leupeptins has been researched along with acetylleucyl-leucyl-norleucinal* in 299 studies
1 review(s) available for leupeptins and acetylleucyl-leucyl-norleucinal
Article | Year |
---|---|
How close are we to implementing a genetic risk score for coronary heart disease?
Genome-wide association meta-analysis have now identified more than 150 loci where common variants (SNPs) are significantly associated with coronary heart disease (CHD) and CHD end points. Areas covered: The authors review publications from their own laboratory and published recently where identified CHD risk SNPs are used in combination, and 'scaled' by their effect size, to create a 'weighted' Genetic risk Score (GRS), which, in combination with an individual's classical CHD risk factors, can be used to identify those at overall low, intermediate and high future risk. Those at highest risk can be offered life-style and therapeutic options to reduce their risk and those at intermediate levels can be monitored. Expert commentary: The authors discuss the selection of the best variants to be included in the GRS, and the potential utility of such scores in different clinical settings. The limitations of the current data sets and the way forward in the next 5 years is discussed. Topics: Coronary Disease; Genetic Association Studies; Genetic Predisposition to Disease; Genetic Testing; Genome-Wide Association Study; Humans; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Leupeptins; Medication Adherence; Polymorphism, Single Nucleotide; Practice Guidelines as Topic; Primary Prevention; Risk Assessment; Risk Factors; Sex Factors; United Kingdom | 2017 |
298 other study(ies) available for leupeptins and acetylleucyl-leucyl-norleucinal
Article | Year |
---|---|
Calpain inhibitor prevents atherosclerosis in apolipoprotein E knockout mice by regulating mRNA expression of genes related to cholesterol uptake and efflux.
We previously reported that a calpain inhibitor (CAI) prevents the development of atherosclerosis in rats. This study aimed to investigate the effects of CAI (1 mg/kg) on atherosclerosis in apolipoprotein E knockout (ApoE KO) mice that were fed a high-fat diet (HFD) and explore the underlying mechanism by analyzing the expression of genes related to the uptake and efflux of cholesterol.. Atherosclerotic plaques were evaluated. The activity of calpain in the aorta and that of superoxide dismutase (SOD) in the serum were assessed. Lipid profiles in the serum and liver were examined. Serum oxidized low-density lipoprotein (oxLDL), malondialdehyde (MDA), tumor necrosis factor (TNF-α), and interleukin-6 (IL-6) levels were measured. The mRNA expressions of CD68, TNF-α, IL-6, CD36, scavenger receptor (SR-A), peroxisome proliferator-activated receptor gamma (PPAR-γ), liver-x-receptor alpha (LXR-α), and ATP-binding cassette transporter class A1 (ABCA1) in the aorta and peritoneal macrophages were also evaluated.. CAI reduced calpain activity in the aorta. CAI also impeded atherosclerotic lesion formation and mRNA expression of CD68 in the aorta and peritoneal macrophages of ApoE KO mice compared with those of mice receiving HFD. However, CAI had no effect on body weight and lipid levels in both the serum and liver. CAI significantly decreased MDA, oxLDL, TNF-α, and IL-6 levels and increased SOD activity in the serum. Moreover, CAI significantly inhibited the mRNA expression of TNF-α and IL-6 genes in the aorta and peritoneal macrophages. In addition, CAI significantly downregulated the mRNA expression of scavenger receptors CD36 and SR-A and upregulated the expression of genes involved in the cholesterol efflux pathway, i.e., PPAR-γ, LXR-α, and ABCA1 in the aorta and peritoneal macrophages.. CAI inhibited the development of atherosclerotic lesions in ApoE KO mice, and this effect might be related to the reduction of oxidative stress and inflammation and the improvement of cholesterol intake and efflux pathways. Topics: Animals; Antigens, CD; Antigens, Differentiation, Myelomonocytic; Aorta; Aortic Diseases; Atherosclerosis; ATP Binding Cassette Transporter 1; Calpain; Cholesterol; Cysteine Proteinase Inhibitors; Disease Models, Animal; Gene Expression Regulation; Leupeptins; Lipid Metabolism; Liver X Receptors; Macrophages, Peritoneal; Male; Mice, Inbred C57BL; Mice, Knockout, ApoE; Plaque, Atherosclerotic; PPAR gamma; RNA, Messenger; Scavenger Receptors, Class A | 2022 |
Identification of Phosphorylated Calpain 3 in Rat Brain Mitochondria under mPTP Opening.
The protein phosphorylation of the membrane-bound mitochondrial proteins has become of interest from the point of view of its regulatory role of the function of the respiratory chain, opening of the mitochondrial permeability transition pore (mPTP), and initiation of apoptosis. Earlier, we noticed that upon phosphorylation of proteins in some proteins, the degree of their phosphorylation increases with the opening of mPTP. Two isoforms of myelin basic protein and cyclic nucleotide phosphodiesterase were identified in rat brain non-synaptic mitochondria and it was concluded that they are involved in mPTP regulation. In the present study, using the mass spectrometry method, the phosphorylated protein was identified as Calpain 3 in rat brain non-synaptic mitochondria. In the present study, the phosphoprotein Calpain-3 (p94) (CAPN3) was identified in the rat brain mitochondria as a phosphorylated truncated form of p60-62 kDa by two-dimensional electrophoresis and mass spectrometry. We showed that the calpain inhibitor, calpeptin, was able to suppress the Ca Topics: Animals; Apoptosis; Brain; Calcium; Calcium Signaling; Calpain; Cysteine Proteinase Inhibitors; Dipeptides; Isoenzymes; Leupeptins; Male; Mitochondria; Mitochondrial Membranes; Mitochondrial Permeability Transition Pore; Molecular Weight; Muscle Proteins; Phosphorylation; Protein Transport; Rats | 2021 |
Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of Nsp5 main protease.
The coronavirus 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), spread around the world with unprecedented health and socio-economic effects for the global population. While different vaccines are now being made available, very few antiviral drugs have been approved. The main viral protease (nsp5) of SARS-CoV-2 provides an excellent target for antivirals, due to its essential and conserved function in the viral replication cycle. We have expressed, purified and developed assays for nsp5 protease activity. We screened the nsp5 protease against a custom chemical library of over 5000 characterised pharmaceuticals. We identified calpain inhibitor I and three different peptidyl fluoromethylketones (FMK) as inhibitors of nsp5 activity in vitro, with IC50 values in the low micromolar range. By altering the sequence of our peptidomimetic FMK inhibitors to better mimic the substrate sequence of nsp5, we generated an inhibitor with a subnanomolar IC50. Calpain inhibitor I inhibited viral infection in monkey-derived Vero E6 cells, with an EC50 in the low micromolar range. The most potent and commercially available peptidyl-FMK compound inhibited viral growth in Vero E6 cells to some extent, while our custom peptidyl FMK inhibitor offered a marked antiviral improvement. Topics: Amino Acid Chloromethyl Ketones; Animals; Antiviral Agents; Azoles; Chlorocebus aethiops; Coronavirus 3C Proteases; Drug Evaluation, Preclinical; Enzyme Assays; Fluorescence Resonance Energy Transfer; High-Throughput Screening Assays; Isoindoles; Leupeptins; Organoselenium Compounds; Peptidomimetics; Reproducibility of Results; RNA-Binding Proteins; SARS-CoV-2; Small Molecule Libraries; Vero Cells; Viral Nonstructural Proteins | 2021 |
Proteasome Inhibitors Bortezomib and Carfilzomib Stimulate the Transport Activity of Human Organic Anion Transporter 1.
Organic anion transporter 1 (OAT1), expressed at the basolateral membrane of renal proximal tubule epithelial cells, mediates the renal excretion of many clinically important drugs. Previous study in our laboratory demonstrated that ubiquitin conjugation to OAT1 leads to OAT1 internalization from the cell surface and subsequent degradation. The current study showed that the ubiquitinated OAT1 accumulated in the presence of the proteasomal inhibitors MG132 and ALLN rather than the lysosomal inhibitors leupeptin and pepstatin A, suggesting that ubiquitinated OAT1 degrades through proteasomes. Anticancer drugs bortezomib and carfilzomib target the ubiquitin-proteasome pathway. We therefore investigate the roles of bortezomib and carfilzomib in reversing the ubiquitination-induced downregulation of OAT1 expression and transport activity. We showed that bortezomib and carfilzomib extremely increased the ubiquitinated OAT1, which correlated well with an enhanced OAT1-mediated transport of p-aminohippuric acid and an enhanced OAT1 surface expression. The augmented OAT1 expression and transport activity after the treatment with bortezomib and carfilzomib resulted from a reduced rate of OAT1 degradation. Consistent with this, we found decreased 20S proteasomal activity in cells that were exposed to bortezomib and carfilzomib. In conclusion, this study identified the pathway in which ubiquitinated OAT1 degrades and unveiled a novel role of anticancer drugs bortezomib and carfilzomib in their regulation of OAT1 expression and transport activity. SIGNIFICANCE STATEMENT: Bortezomib and carfilzomib are two Food and Drug Administration-approved anticancer drugs, and proteasome is the drug target. In this study, we unveiled a new role of bortezomib and carfilzomib in enhancing OAT1 expression and transport activity by preventing the degradation of ubiquitinated OAT1 in proteasomes. This finding provides a new strategy in regulating OAT1 function that can be used to accelerate the clearance of drugs, metabolites, or toxins and reverse the decreased expression under disease conditions. Topics: Antineoplastic Agents; Biological Transport, Active; Bortezomib; HEK293 Cells; Humans; Leupeptins; Oligopeptides; Organic Anion Transport Protein 1; p-Aminohippuric Acid; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Proteolysis; Ubiquitination | 2020 |
Calpain-dependent Beclin1 cleavage stimulates senescence-associated cell death in HT22 hippocampal cells under the oxidative stress conditions.
Oxidative damage in neurons including glutamate excitotoxicity has been linked to increasing numbers of neuropathological conditions. Under these conditions, cells trigger several different cellular responses such as autophagy, apoptosis, necrosis and senescence. However, the connection between these responses is not well understood. In this study, we found that the 60-kDa BECN1 was specifically degraded to a 40-kDa fragment in hippocampal HT22 cells treated with 5 mM glutamate. Increased BECN1 cleavage was specifically associated with a decrease in cell viability under oxidative stress. Interestingly, this BECN1 cleavage was specifically inhibited by a calpain inhibitor ALLN but was not affected by other protease inhibitors. Also, the BECN1 cleavage was not detected in calpain-4-deficient cell lines. Furthermore, calpain cleaved BECN1 at a specific site between the coiled-coil domain and Bcl2 homology 3 domain, which is associated with the anti-apoptotic protein Bcl-2. Moreover, some cellular senescence markers, including β-galactosidase, p21, p27 Topics: Animals; Apoptosis; Beclin-1; Calpain; Caspases; Cell Line; Cell Survival; Cellular Senescence; Glutamic Acid; HeLa Cells; Hippocampus; Humans; Leupeptins; Mice; Neurons; NIH 3T3 Cells; Oxidative Stress; Reactive Oxygen Species | 2019 |
The association between HLA and non-Hodgkin lymphoma subtypes, among a transplant-indicated population.
Several studies have implicated HLA in non-Hodgkin lymphoma (NHL) subtype etiology. However, NHL patients indicated for stem cell transplants are underrepresented in these reports. We therefore evaluated the association between HLA and NHL subtypes among a transplant-indicated population. One thousand three hundred and sixty-six NHL patients HLA-typed and indicated for transplant at the City of Hope National Medical Center (Duarte, CA) were compared to 10,271 prospective donors. Odds ratios and 95% confidence intervals were calculated for HLA haplotype and alleles, adjusted for sex and age. The Topics: Adult; Clinical Decision-Making; Diagnosis, Differential; Disease Management; Female; Genetic Association Studies; Genetic Predisposition to Disease; Haplotypes; Hematopoietic Stem Cell Transplantation; HLA Antigens; Humans; Leupeptins; Lymphoma, Non-Hodgkin; Male; Middle Aged | 2019 |
Sepia Ink Oligopeptide Induces Apoptosis of Lung Cancer Cells via Mitochondrial Pathway.
Our previous study suggested the anti-tumor activity of sepia ink oligopeptide (SIO). Here we sought to investigate the underlying molecular mechanism.. Cell proliferation was evaluated by cell counting kit-8 (CCK-8) assay. Cell apoptosis was determined by Annexin V/Propidium Iodide (PI) staining. The mitochondria pathway was characterized by quantification of Bcl-2, Bax, Caspase-9 and Cyto-C. The death receptor pathway was analyzed by determinement of Fas, Caspase-8 and NIK. The endoplasmic reticulum (ER)-dependent pathway was determined by measurement the expression of CHOP, Caspase-12, GRP78 and Calpain. The associated gene expression was quantified by RT-PCR and protein level was determined by immunoblotting.. We demonstrated treatment with structurally modified SIO (CSIO, 5 µM) significantly inhibited cell proliferation and induced apoptosis in lung cancer cell line A549. The mitochondrial pathway, death receptor pathway and ER stress induced apoptosis were stimulated upon CSIO treatment. The administration with respective inhibitors including midiv-1 (50 µM for 2 h), PDTC (20 µM PDTC for 30 min) and ALLN (20 mM ALLN for 5 h) readily reversed the apoptosis inducing effect of CSIO.. Our data demonstrates that CSIO is capable of induction apoptosis in lung cancer cell line, which is mediated by all three classical apoptotic pathways. Our results warrant further in vivo investigations of the anti-tumor potential of CSIO. Topics: A549 Cells; Animals; Apoptosis; bcl-2-Associated X Protein; Calpain; Caspase 12; Caspase 8; Caspase 9; Cell Proliferation; Endoplasmic Reticulum; Endoplasmic Reticulum Chaperone BiP; Endoplasmic Reticulum Stress; fas Receptor; Humans; Ink; Leupeptins; Lung Neoplasms; Mitochondria; Oligopeptides; Proline; Proto-Oncogene Proteins c-bcl-2; Sepia; Thiocarbamates; Transcription Factor CHOP | 2018 |
The role and mechanism of chaperones Calnexin/Calreticulin in which ALLN selectively rescues the trafficking defective of HERG-A561V mutation.
Topics: Calnexin; Calreticulin; ERG1 Potassium Channel; HEK293 Cells; Humans; Leupeptins; Long QT Syndrome; Molecular Chaperones; Mutation; Patch-Clamp Techniques; Propranolol; Protein Transport | 2018 |
Ischemia-reperfusion induces death receptor-independent necroptosis via calpain-STAT3 activation in a lung transplant setting.
Ischemia-reperfusion (I/R)-induced lung injury undermines lung transplantation (LTx) outcomes by predisposing lung grafts to primary graft dysfunction (PGD). Necrosis is a feature of I/R lung injury. However, regulated necrosis (RN) with specific signaling pathways has not been explored in an LTx setting. In this study, we investigated the role of RN in I/R-induced lung injury. To study I/R-induced cell death, we simulated an LTx procedure using our cell culture model with human lung epithelial (BEAS-2B) cells. After 18 h of cold ischemic time (CIT) followed by reperfusion, caspase-independent cell death, mitochondrial reactive oxygen species production, and mitochondrial membrane permeability were significantly increased. N-acetyl-Leu-Leu-norleucinal (ALLN) (calpain inhibitor) or necrostatin-1 (Nec-1) [receptor interacting serine/threonine kinase 1 (RIPK1) inhibitor] reduced these changes. ALLN altered RIPK1/RIPK3 expression and mixed lineage kinase domain-like (MLKL) phosphorylation, whereas Nec-1 did not change calpain/calpastatin expression. Furthermore, signal transducer and activator of transcription 3 (STAT3) was demonstrated to be downstream of calpain and regulate RIPK3 expression and MLKL phosphorylation during I/R. This calpain-STAT3-RIPK axis induces endoplasmic reticulum stress and mitochondrial calcium dysregulation. LTx patients' samples demonstrate that RIPK1, MLKL, and STAT3 mRNA expression increased from CIT to reperfusion. Moreover, the expressions of the key proteins are higher in PGD samples than in non-PGD samples. Cell death associated with prolonged lung preservation is mediated by the calpain-STAT3-RIPK axis. Inhibition of RIPK and/or calpain pathways could be an effective therapy in LTx. Topics: Apoptosis; Calpain; Cells, Cultured; Humans; Imidazoles; Indoles; Leupeptins; Lung Transplantation; Necrosis; Phosphorylation; Primary Graft Dysfunction; Receptor-Interacting Protein Serine-Threonine Kinases; Receptors, Death Domain; Reperfusion Injury; Signal Transduction; STAT3 Transcription Factor | 2018 |
A susceptibility locus for classical Hodgkin lymphoma at 8q24 near MYC/PVT1 predicts patient outcome in two independent cohorts.
Topics: Chromosomes, Human, Pair 8; Genes, myc; Genetic Loci; Genetic Predisposition to Disease; Hodgkin Disease; Humans; Leupeptins; Polymorphism, Single Nucleotide; Prognosis; RNA, Long Noncoding; Survival Analysis | 2018 |
Schizophrenia-Associated hERG channel Kv11.1-3.1 Exhibits a Unique Trafficking Deficit that is Rescued Through Proteasome Inhibition for High Throughput Screening.
The primate-specific brain voltage-gated potassium channel isoform Kv11.1-3.1 has been identified as a novel therapeutic target for the treatment of schizophrenia. While this ether-a-go-go related K(+)channel has shown clinical relevance, drug discovery efforts have been hampered due to low and inconsistent activity in cell-based assays. This poor activity is hypothesized to result from poor trafficking via the lack of an intact channel-stabilizing Per-Ant-Sim (PAS) domain. Here we characterize Kv11.1-3.1 cellular localization and show decreased channel expression and cell surface trafficking relative to the PAS-domain containing major isoform, Kv11.1-1A. Using small molecule inhibition of proteasome degradation, cellular expression and plasma membrane trafficking are rescued. These findings implicate the importance of the unfolded-protein response and endoplasmic reticulum associated degradation pathways in the expression and regulation of this schizophrenia risk factor. Utilizing this identified phenomenon, an electrophysiological and high throughput in-vitro fluorescent assay platform has been developed for drug discovery in order to explore a potentially new class of cognitive therapeutics. Topics: Bortezomib; Cell Membrane; Drug Discovery; Ether-A-Go-Go Potassium Channels; Gene Expression; HEK293 Cells; High-Throughput Screening Assays; Humans; Intracellular Space; Leupeptins; Mutation; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Interaction Domains and Motifs; Protein Isoforms; Protein Transport; Schizophrenia | 2016 |
The Toxic Effect of ALLN on Primary Rat Retinal Neurons.
N-acetyl-leucyl-leucyl-norleucinal (ALLN), an inhibitor of proteasomes and calpain, is widely used to reduce proteasomes or calpain-mediated cell death in rodents. However, ALLN is toxic to retinal neurons to some extent. At the concentration of 10 μM, ALLN is non-toxic to cortical neurons, but induces cell death of retinal neurons in vitro. The tolerance concentration of ALLN for retinal neurons is unclear, and the precise mechanism of cell death induced by ALLN remains elusive. In this study, we investigated the toxic effect of ALLN on primary retinal neurons. The 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed no significant changes of cell viability at 1 μM but decreased cell viability after treatment of ALLN at 2.5, 5, and 7.5 μM. Lactate dehydrogenase (LDH) release was highly elevated and propidium iodide (PI)-positive cells were significantly increased at 2.5, 5, and 7.5 μM after all treatment times. Moreover, the protein levels of caspase-3 were up-regulated at 5 and 7.5 μM after 12 and 24 h of ALLN treatment. The ratio of Bax/Bcl-2 was raised and Annexin V-positive cells were increased at 5 and 7.5 μM after 12 and 24 h of ALLN treatment. However, there were no significant changes in either the ratio of microtubule-associated protein 1 light chain 3 (LC3) II/LC3 I or monodansylcadaverine (MDC) staining. Our data clearly show that at the concentrations equal to and higher than 2.5 μM, ALLN may induce cell death of primary retinal neurons by necrosis and apoptosis, but not autophagy. These suggest that primary retinal neurons are more susceptible to ALLN treatment and provide a possible mechanism for the cell death of ALLN-sensitive cells in ALLN injury. Topics: Animals; Apoptosis; Autophagy; bcl-2-Associated X Protein; Blotting, Western; Caspase 3; Cell Survival; Cells, Cultured; Dose-Response Relationship, Drug; Drug Evaluation, Preclinical; Fluorescent Antibody Technique; L-Lactate Dehydrogenase; Leupeptins; Microtubule-Associated Proteins; Mitochondria; Necrosis; Proto-Oncogene Proteins c-bcl-2; Rats, Sprague-Dawley; Retinal Neurons; Time Factors | 2016 |
Calpain-dependent truncated form of TrkB-FL increases in neurodegenerative processes.
Recent findings indicate that the mechanisms that drive reshaping of the nervous system are aberrantly activated in epilepsy and several neurodegenerative diseases. The recurrent seizures in epilepsy, particularly in the condition called status epilepticus, can cause permanent neurological damage, resulting in cognitive dysfunction and other serious neurological conditions. In this study, we used an in vitro model of status epilepticus to examine the role of calpain in the degeneration of hippocampal neurons. We grew neurons on a culture system that allowed studying the dendritic and axonal domains separately from the cell bodies. We found that a recently characterized calpain substrate, the neurotrophin receptor TrkB, is cleaved in the dendritic and axonal domain of neurons committed to die, and this constitutes an early step in the neuronal degeneration process. While the full-length TrkB (TrkB-FL) levels decreased, the truncated form of TrkB (Tc TrkB-FL) concurrently increased, leading to a TrkB-FL/Tc TrkB-FL imbalance, which is thought to be causally linked to neurodegeneration. We further show that the treatment with N-acetyl-Leu-Leu-norleucinal, a specific calpain activity blocker, fully protects the neuronal processes from degeneration, prevents the TrkB-FL/Tc TrkB-FL imbalance, and provides full neuroprotection. Moreover, the use of the TrkB antagonist ANA 12 at the time when the levels of TrkB-FL were significantly decreased, totally blocked neuronal death, suggesting that Tc TrkB-FL may have a role in neuronal death. These results indicate that the imbalance of these neurotrophins receptors plays a key role in neurite degeneration induced by seizures. Topics: Animals; Calcium; Calpain; Cell Death; Cells, Cultured; Hippocampus; Leupeptins; Neurons; Protease Inhibitors; Proteolysis; Rats; Rats, Wistar; Receptor, trkB; Status Epilepticus | 2016 |
Iron overload-induced calcium signals modulate mitochondrial fragmentation in HT-22 hippocampal neuron cells.
Iron is necessary for neuronal functions; however, excessive iron accumulation caused by impairment of iron balance could damage neurons. Neuronal iron accumulation has been observed in several neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. Nevertheless, the precise mechanisms underlying iron toxicity in neuron cells are not fully understood. In this study, we investigated the mechanism underlying iron overload-induced mitochondrial fragmentation in HT-22 hippocampal neuron cells that were incubated with ferric ammonium citrate (FAC). Mitochondrial fragmentation via dephosphorylation of Drp1 (Ser637) and increased apoptotic neuronal death were observed in FAC-stimulated HT-22 cells. Furthermore, the levels of intracellular calcium (Ca(2+)) were increased by iron overload. Notably, chelation of intracellular Ca(2+) rescued mitochondrial fragmentation and neuronal cell death. In addition, iron overload activated calcineurin through the Ca(2+)/calmodulin and Ca(2+)/calpain pathways. Pretreatment with the calmodulin inhibitor W13 and the calpain inhibitor ALLN attenuated iron overload-induced mitochondrial fragmentation and neuronal cell death. Therefore, these findings suggest that Ca(2+)-mediated calcineurin signals are a key player in iron-induced neurotoxicity by regulating mitochondrial dynamics. We believe that our results may contribute to the development of novel therapies for iron toxicity related neurodegenerative disorders. Topics: Animals; Calcineurin; Calcium; Calcium Signaling; Calmodulin; Calpain; Cell Death; Cell Line; Cell Survival; Dynamins; Ferric Compounds; Hippocampus; Iron; Iron Overload; Leupeptins; Mice; Mitochondria; Neurons; Quaternary Ammonium Compounds; Sulfonamides | 2016 |
Inhibition of calpain on oxygen glucose deprivation-induced RGC-5 necroptosis.
The purpose of this study was to investigate the effect of inhibition of calpain on retinal ganglion cell-5 (RGC-5) necroptosis following oxygen glucose deprivation (OGD). RGC-5 cells were cultured in Dulbecco's-modified essential medium and necroptosis was induced by 8-h OGD. PI staining and flow cytometry were performed to detect RGC-5 necrosis. The calpain expression was detected by Western blotting and immunofluorescence staining. The calpain activity was tested by activity detection kit. Flow cytometry was used to detect the effect of calpain on RGC-5 necroptosis following OGD with or without N-acetyl-leucyl-leucyl-norleucinal (ALLN) pre-treatment. Western blot was used to detect the protein level of truncated apoptosis inducing factor (tAIF) in RGC-5 cells following OGD. The results showed that there was an up-regulation of the calpain expression and activity following OGD. Upon adding ALLN, the calpain activity was inhibited and tAIF was reduced following OGD along with the decreased number of RGC-5 necroptosis. In conclusion, calpain was involved in OGD-induced RGC-5 necroptosis with the increased expression of its downstream molecule tAIF. Topics: Animals; Apoptosis Inducing Factor; Calpain; Gene Expression Regulation; Glucose; Humans; Leupeptins; Mice; Oxygen; Retinal Ganglion Cells; Retinal Necrosis Syndrome, Acute | 2016 |
Activation of Both the Calpain and Ubiquitin-Proteasome Systems Contributes to Septic Cardiomyopathy through Dystrophin Loss/Disruption and mTOR Inhibition.
Cardiac dysfunction caused by the impairment of myocardial contractility has been recognized as an important factor contributing to the high mortality in sepsis. Calpain activation in the heart takes place in response to increased intracellular calcium influx resulting in proteolysis of structural and contractile proteins with subsequent myocardial dysfunction. The purpose of the present study was to test the hypothesis that increased levels of calpain in the septic heart leads to disruption of structural and contractile proteins and that administration of calpain inhibitor-1 (N-acetyl-leucinyl-leucinyl-norleucinal (ALLN)) after sepsis induced by cecal ligation and puncture prevents cardiac protein degradation. We also tested the hypothesis that calpain plays a role in the modulation of protein synthesis/degradation through the activation of proteasome-dependent proteolysis and inhibition of the mTOR pathway. Severe sepsis significantly increased heart calpain-1 levels and promoted ubiquitin and Pa28β over-expression with a reduction in the mTOR levels. In addition, sepsis reduced the expression of structural proteins dystrophin and β-dystroglycan as well as the contractile proteins actin and myosin. ALLN administration prevented sepsis-induced increases in calpain and ubiquitin levels in the heart, which resulted in decreased of structural and contractile proteins degradation and basal mTOR expression levels were re-established. Our results support the concept that increased calpain concentrations may be part of an important mechanism of sepsis-induced cardiac muscle proteolysis. Topics: Actins; Animals; Calpain; Cardiomyopathies; Disease Models, Animal; Dystrophin; Gene Expression; Leupeptins; Male; Mice; Mice, Inbred C57BL; Microscopy, Fluorescence; Myocardium; Myosins; Proteasome Endopeptidase Complex; Proteolysis; Sepsis; TOR Serine-Threonine Kinases; Ubiquitin | 2016 |
Specific calpain activity evaluation in Plasmodium parasites.
In the intraerythrocytic trophozoite stages of Plasmodium falciparum, the calcium-dependent cysteine protease calpain (Pf-calpain) has an important role in the parasite calcium modulation and cell development. We established specific conditions to follow by confocal microscopy and spectrofluorimetry measurements the intracellular activity of Pf-calpain in live cells. The catalytic activity was measured using the fluorogenic Z-Phe-Arg-MCA (where Z is carbobenzoxy and MCA is 4-methylcoumaryl-7-amide). The calmodulin inhibitor calmidazolium and the sarcoplasmic reticulum calcium ATPase inhibitor thapsigargin were used for modifications in the cytosolic calcium concentrations that persisted in the absence of extracellular calcium. The observed calcium-dependent peptidase activity was greatly inhibited by specific cysteine protease inhibitor E-64 and by the selective calpain inhibitor ALLN (N-acetyl-l-leucyl-l-leucyl-l-norleucinal). Taken together, we observed that intracellular Pf-calpain can be selectively detected and is the main calcium-dependent protease in the intraerythrocytic stages of the parasite. The method described here can be helpful in cell metabolism studies and antimalarial drug screening. Topics: Animals; Calcium; Calpain; Cysteine Proteinase Inhibitors; Leupeptins; Male; Mice; Mice, Inbred BALB C; Microscopy, Confocal; Plasmodium chabaudi; Plasmodium falciparum; Protozoan Proteins; Spectrometry, Fluorescence | 2015 |
Cathepsin L Mediates the Degradation of Novel APP C-Terminal Fragments.
Alzheimer's disease (AD) is characterized by the deposition of amyloid β (Aβ), a peptide generated from proteolytic processing of its precursor, amyloid precursor protein (APP). Canonical APP proteolysis occurs via α-, β-, and γ-secretases. APP is also actively degraded by protein degradation systems. By pharmacologically inhibiting protein degradation with ALLN, we observed an accumulation of several novel APP C-terminal fragments (CTFs). The two major novel CTFs migrated around 15 and 25 kDa and can be observed across multiple cell types. The process was independent of cytotoxicity or protein synthesis. We further determine that the accumulation of the novel CTFs is not mediated by proteasome or calpain inhibition, but by cathepsin L inhibition. Moreover, these novel CTFs are not generated by an increased amount of BACE. Here, we name the CTF of 25 kDa as η-CTF (eta-CTF). Our data suggest that under physiological conditions, a subset of APP undergoes alternative processing and the intermediate products, the 15 kDa CTFs, and the η-CTFs aret rapidly degraded and/or processed via the protein degradation machinery, specifically, cathepsin L. Topics: Amyloid beta-Protein Precursor; Amyloid Precursor Protein Secretases; Animals; Calpain; Cathepsin L; Cattle; Cell Line, Tumor; Cells, Cultured; Humans; L-Lactate Dehydrogenase; Leupeptins; Peptide Fragments; Proteolysis; Rats; Ubiquitination | 2015 |
Activated microglia/macrophage whey acidic protein (AMWAP) inhibits NFκB signaling and induces a neuroprotective phenotype in microglia.
Microglia reactivity is a hallmark of neurodegenerative diseases. We have previously identified activated microglia/macrophage whey acidic protein (AMWAP) as a counter-regulator of pro-inflammatory response. Here, we studied its mechanisms of action with a focus on toll-like receptor (TLR) and nuclear factor κB (NFκB) signaling.. Recombinant AMWAP was produced in Escherichia coli and HEK293 EBNA cells and purified by affinity chromatography. AMWAP uptake was identified by fluorescent labeling, and pro-inflammatory microglia markers were measured by qRT-PCR after stimulation with TLR ligands. NFκB pathway proteins were assessed by immunocytochemistry, Western blot, and immunoprecipitation. A 20S proteasome activity assay was used to investigate the anti-peptidase activity of AMWAP. Microglial neurotoxicity was estimated by nitrite measurement and quantification of caspase 3/7 levels in 661W photoreceptors cultured in the presence of microglia-conditioned medium. Microglial proliferation was investigated using flow cytometry, and their phagocytosis was monitored by the uptake of 661W photoreceptor debris.. AMWAP was secreted from lipopolysaccharide (LPS)-activated microglia and recombinant AMWAP reduced gene transcription of IL6, iNOS, CCL2, CASP11, and TNFα in BV-2 microglia treated with LPS as TLR4 ligand. This effect was replicated with murine embryonic stem cell-derived microglia (ESdM) and primary brain microglia. AMWAP also diminished pro-inflammatory markers in microglia activated with the TLR2 ligand zymosan but had no effects on IL6, iNOS, and CCL2 transcription in cells treated with CpG oligodeoxynucleotides as TLR9 ligand. Microglial uptake of AMWAP effectively inhibited TLR4-dependent NFκB activation by preventing IRAK-1 and IκBα proteolysis. No inhibition of IκBα phosphorylation or ubiquitination and no influence on overall 20S proteasome activity were observed. Functionally, both microglial nitric oxide (NO) secretion and 661W photoreceptor apoptosis were significantly reduced after AMWAP treatment. AMWAP promoted the filopodia formation of microglia and increased the phagocytic uptake of apoptotic 661W photoreceptor cells.. AMWAP is secreted from reactive microglia and acts in a paracrine fashion to counter-balance TLR2/TLR4-induced reactivity through NFκB inhibition. AMWAP also induces a neuroprotective microglial phenotype with reduced neurotoxicity and increased phagocytosis. We therefore hypothesize that anti-inflammatory whey acidic proteins could have a therapeutic potential in neurodegenerative diseases of the brain and the retina. Topics: Animals; Antibodies; Apoptosis; Cell Line, Transformed; Cell Proliferation; Enzyme Inhibitors; Humans; Leupeptins; Lipopolysaccharides; Mice; Microglia; Nerve Tissue Proteins; Neuroprotective Agents; NF-kappa B; Nitrites; Phagocytosis; Photoreceptor Cells; Proteasome Endopeptidase Complex; RNA, Messenger; Signal Transduction | 2015 |
Zinc pyrithione inhibits caspase-3 activity, promotes ErbB1-ErbB2 heterodimerization and suppresses ErbB2 downregulation in cardiomyocytes subjected to ischemia/reperfusion.
Heart tissue becomes zinc-depleted and the capacity to mobilize labile zinc is diminished, indicating zinc dyshomeostasis during ischemia/reperfusion (I/R). Apparently, zinc pyrithione restores the basal zinc levels during I/R and prevents apoptosis by activating phosphatidyl inositol-3-kinase/Akt and targeting mitochondrial permeability transition. Receptor tyrosine kinases of the ErbB family (ErbB1 to ErbB4) are cell surface proteins that can regulate cell growth, proliferation and survival. Previous studies have shown that zinc pyrithione-induced activation of PI3kinase/Akt requires ErbB2 expression. On the other hand, while I/R decreases ErbB2 levels causing cardiomyocyte dysfunction and cell death, zinc pyrithione restores ErbB2 levels and maintains cardiomyocyte function. H9c2 cells expressed all the four ErbBs, although the expression of ErbB1 and ErbB2 were higher compared to ErbB3 and ErbB4. Hypoxia/Reoxygenation (H/R) had opposing effects on the mRNA expression of ErbB1 and ErbB2. ErbB2 mRNA levels were enhanced, but corresponding ErbB2 protein levels decreased after reoxygenation. H/R induced the degradation of ErbB2 in caspase-3 dependent manner, with the formation of a 25kDa fragment. This fragment could be detected after H/R only upon treatment of the cells with a proteasomal inhibitor, ALLN, suggesting that caspase-mediated cleavage of 185kDa ErbB2 results in C-terminal cleavage and formation of 25kDa fragment, which is further degraded by proteasome. Heterodimerization and phosphorylation of ErbB2/ErbB1 which decreased upon reoxygenation, was promoted by zinc pyrithione. Zinc pyrithione effectively suppressed the caspase activation, decreased the proteolytic cleavage of ErbB2, enhanced the phosphorylation and activation of ErbB1-ErbB2 complexes and improved the cell survival after hypoxia/reoxygenation. Topics: Amino Acid Chloromethyl Ketones; Animals; Benzothiazoles; Cardiotonic Agents; Caspase 3; Caspase Inhibitors; Cell Hypoxia; Chlorides; Down-Regulation; ErbB Receptors; Lapatinib; Leupeptins; Myocytes, Cardiac; Organometallic Compounds; Phosphorylation; Proteasome Inhibitors; Protein Kinase Inhibitors; Protein Multimerization; Pyridines; Quinazolines; Rats; Receptor, ErbB-2; RNA, Messenger; Thiones; Tyrphostins; Zinc Compounds | 2015 |
Sorafenib enhances proteasome inhibitor-induced cell death via inactivation of Akt and stress-activated protein kinases.
Advanced hepatocellular carcinoma (HCC) responds poorly to conventional systemic therapies. Therefore, new effective therapy strategies are urgently needed. Molecular targeted therapies have entered the field of anti-neoplastic treatment and are being used on their own and in combination with other drugs. Sorafenib inhibits proliferation and angiogenesis of HCC by suppressing the Raf serine/threonine kinases and the receptor tyrosine kinases. The proteasome inhibitor bortezomib has shown activity in a variety of solid tumors, including HCC. However, the precise anti-proliferative mechanisms of these agents remain unclear.. We treated human hepatoma cell lines (Huh7 and Hep3B) and immortalized human hepatocyte (OUMS29) with sorafenib and/or proteasome inhibitors, including epoxomicin and acetyl-leucyl-leucyl-norleucinal. Cytotoxic effects were examined by morphometric analyses of apoptosis and necrosis. Apoptosis was also evaluated by Western blotting of keratin18, PARP and caspase3. The activity of Akt and stress-activated protein kinases was examined by Western blotting.. Both sorafenib and proteasome inhibitors induced apoptosis in Huh7 and OUMS29. However, sorafenib attenuated proteasome inhibitor-induced apoptosis. Sorafenib induced necrosis, especially in combination with proteasome inhibitors. Sorafenib induced down-regulation of Akt synergistically in combination with proteasome inhibitors in Huh7. Sorafenib inhibited both the JNK and p38 pathways in a time- and dose-dependent manner. In addition, sorafenib also inhibited proteasome inhibitor-mediated JNK and p38 activation in both Huh7 and OUMS29.. Sorafenib enhances the anti-proliferative effect of proteasome inhibitors in part by inactivating the Akt signaling pathway and modulating stress-activated protein kinases. The combination of these agents could be an ideal molecular targeted therapy for HCC. Topics: Antineoplastic Agents; Apoptosis; Carcinoma, Hepatocellular; Cell Death; Cell Line, Tumor; Down-Regulation; Drug Synergism; Humans; Leupeptins; Liver Neoplasms; Molecular Targeted Therapy; Niacinamide; Oligopeptides; Phenylurea Compounds; Proteasome Inhibitors; Protein Kinase Inhibitors; Protein Kinases; Proto-Oncogene Proteins c-akt; Signal Transduction; Sorafenib | 2014 |
Protective effect of S-allyl-L-cysteine against endoplasmic reticulum stress-induced neuronal death is mediated by inhibition of calpain.
Endoplasmic reticulum (ER) stress, implicated in various neurodegenerative processes, increases the level of intracellular Ca(2+) and leads to activation of calpain, a Ca(2+)-dependent cysteine protease. We have shown previously that S-allyl-L-cysteine (SAC) in aged garlic extracts significantly protects cultured rat hippocampal neurons (HPNs) against ER stress-induced neurotoxicity. The neuroprotective effect of SAC was compared with those of the related antioxidant compounds, L-cysteine (CYS) and N-acetylcysteine (NAC), on calpain activity in HPNs and also in vitro. SAC, but not CYS or NAC, reversibly restored the survival of HPNs and increased the degradation of α-spectrin, a substrate for calpain, induced by tunicamycin, a typical ER stress inducer. Activities of μ- and m-calpains in vitro were also concentration dependently suppressed by SAC, but not by CYS or NAC. At submaximal concentration, although ALLN (5 pM), which blocks the active site of calpain, and calpastatin (100 pM), an endogenous calpain-inhibitor protein, additively inhibited μ-calpain activity in vitro in combination with SAC, the effect of PD150606 (25 μM), which prevents interaction of Ca(2+) with the Ca(2+)-binding site of calpain, was unaffected by SAC. In contrast, SAC (1 mM) significantly reversed the effect of PD150606 at a concentration that elicited supramaximal inhibition (100 μM), but did not affect ALLN (1 nM)- and calpastatin (100 nM)-induced inhibition of μ-calpain activity. These results suggest that the protective effects of SAC against ER stress-induced neuronal cell death are not attributable to antioxidant activity, but to suppression of calpain through interaction with its Ca(2+)-binding site. Topics: Animals; Apoptosis; Calcium-Binding Proteins; Calpain; Cell Survival; Cells, Cultured; Cysteine; Dipeptides; Endoplasmic Reticulum Stress; Hippocampus; Leupeptins; Neurons; Neuroprotective Agents; Oxidative Stress; Rats; Rats, Wistar; Spectrin | 2014 |
Candida albicans and Candida parapsilosis rapidly up-regulate galectin-3 secretion by human gingival epithelial cells.
Galectin-3 is a β-galactoside-binding C-type lectin that plays an important role in innate immunity. The purpose of this study was to determine whether Candida albicans and Candida parapsilosis up-regulate galectin-3 secretion by human gingival epithelial cells and gingival fibroblasts. Ca9-22, a human gingival epithelial cell line, and human gingival fibroblasts were incubated in the presence or absence of C. albicans or C. parapsilosis without serum. Levels of secreted human galectin-3 in culture supernatants were measured by enzyme-linked immunosorbent assay. We also pretreated Ca9-22 cells with cytochalasin D (an actin polymerization inhibitor), ALLN (a calpain inhibitor) and LY294002 [a phosphatidylinositol-3 kinase (PI3K) inhibitor] to determine whether the up-regulation of galectin-3 secretion was mediated by cytoskeletal changes, protease activity, or PI3K signaling. Galectin-3 secretion was significantly and rapidly up-regulated by live C. albicans and C. parapsilosis, as well as heat-killed C. albicans. In addition, cytochalasin D, LY294002 and ALLN did not inhibit the up-regulation in galectin-3 secretion. These results suggest that both live and heat-killed C. albicans and C. parapsilosis may increase the activity of the innate immune system and invasion by other microorganisms via up-regulation of galectin-3 secretion. Topics: Actins; Blood Proteins; Calpain; Candida albicans; Cell Line; Chromones; Cytochalasin D; Enzyme-Linked Immunosorbent Assay; Epithelial Cells; Fibroblasts; Galectin 3; Galectins; Gingiva; Humans; Leupeptins; Morpholines; Phosphoinositide-3 Kinase Inhibitors | 2014 |
Protein synthesis dependence of growth cone collapse induced by different Nogo-A-domains.
The protein Nogo-A regulates axon growth in the developing and mature nervous system, and this is carried out by two distinct domains in the protein, Nogo-A-Δ20 and Nogo-66. The differences in the signalling pathways engaged in axon growth cones by these domains are not well characterized, and have been investigated in this study.. We analyzed growth cone collapse induced by the Nogo-A domains Nogo-A-Δ20 and Nogo-66 using explanted chick dorsal root ganglion neurons growing on laminin/poly-lysine substratum. Collapse induced by purified Nogo-A-Δ20 peptide is dependent on protein synthesis whereas that induced by Nogo-66 peptide is not. Nogo-A-Δ20-induced collapse is accompanied by a protein synthesis-dependent rise in RhoA expression in the growth cone, but is unaffected by proteasomal catalytic site inhibition. Conversely Nogo-66-induced collapse is inhibited ∼ 50% by proteasomal catalytic site inhibition.. Growth cone collapse induced by the Nogo-A domains Nogo-A-Δ20 and Nogo-66 is mediated by signalling pathways with distinguishable characteristics concerning their dependence on protein synthesis and proteasomal function. Topics: Animals; Anisomycin; Chick Embryo; Ganglia, Spinal; Gene Expression Regulation, Developmental; Growth Cones; Laminin; Leupeptins; Myelin Proteins; Nogo Proteins; Polylysine; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Biosynthesis; Protein Structure, Tertiary; Protein Synthesis Inhibitors; Recombinant Proteins; rhoA GTP-Binding Protein; Signal Transduction; Tissue Culture Techniques | 2014 |
Re-trafficking of hERG reverses long QT syndrome 2 phenotype in human iPS-derived cardiomyocytes.
Long QT syndrome 2 (LQTS2) caused by missense mutations in hERG channel is clinically associated with abnormally prolonged ventricular repolarization and sudden cardiac deaths. Modelling monogenic arrhythmogenic diseases using human-induced pluripotent stem cells (hiPSCs) offers unprecedented mechanistic insights into disease pathogenesis. We utilized LQTS2-hiPSC-derived cardiomyocytes (CMs) to elucidate pathological changes and to demonstrate reversal of LQTS2 phenotype in a therapeutic intervention using a pharmacological agent, (N-[N-(N-acetyl-l-leucyl)-l-leucyl]-l-norleucine) (ALLN).. We generated LQTS2-specific CMs (A561V missense mutation in KCNH2) from iPSCs using the virus-free reprogramming method. These CMs recapitulate dysfunction of hERG potassium channel with diminished IKr currents, prolonged repolarization durations, and elevated arrhythmogenesis due to reduced membrane localization of glycosylated/mature hERG. Dysregulated expression of folding chaperones and processing proteasomes coupled with sequestered hERG in the endoplasmic reticulum confirmed trafficking-induced disease manifestation. Treatment with ALLN, not only increased membrane localization of mature hERG but also reduced repolarization, increased IKr currents and reduced arrhythmogenic events. Diverged from biophysical interference of hERG channel, our results show that modulation of chaperone proteins could be therapeutic in LQTS2 treatment.. Our in vitro study shows an alternative approach to rescue diseased LQTS2 phenotype via corrective re-trafficking therapy using a small chemical molecule, such as ALLN. This potentially novel approach may have ramifications in other clinically relevant trafficking disorders. Topics: Animals; ERG1 Potassium Channel; Ether-A-Go-Go Potassium Channels; Humans; Induced Pluripotent Stem Cells; Leupeptins; Long QT Syndrome; Male; Mice; Mutation; Myocytes, Cardiac; Phenotype; Protein Transport | 2014 |
Hepatic lipase release is inhibited by a purinergic induction of autophagy.
We have shown that extracellular adenosine diphosphate (ADP) affects lipoprotein secretion from liver cells by stimulating cellular autophagic degradation. In this study, we investigated the effect of ADP and cellular autophagy on hepatic lipase (HL) release from human liver cells.. Depletion of media serum stimulates an autophagic response in liver cells, which parallels an 8-fold increase in the release of ADP into the media and a complete inhibition of HL release. Treatment of cells with exogenous ADP stimulates cellular autophagy and also blocks HL release. Treatment with the autophagic stimulant and proteasomal inhibitor, ALLN (25 µM), reduces cellular HL levels and blocks HL release at 4h. In contrast, treatment with the autophagy inhibitor, 3-methyladenine (3-MA) (5 mM), increases cellular HL levels and stimulates HL release. ADP acts through the G-protein coupled receptor, P2Y13, to stimulate autophagy. siRNA-targeted reduction in P2Y13 protein expression stimulates the release of HL by 5 to 8-fold, while overexpression of P2Y13 blocks HL release. HL release from liver cells is therefore inhibited by a purinergic induction of autophagy. To evaluate the effect of extracellular ADP on the processing of HL, we expressed a V5-epitope tag-labeled HL (HL-V5) and then measured secretion, uptake and degradation. Two isoforms of HL-V5, at 62 and 68 kDa, are released from HepG2 cells, but only the 62 kDa protein undergoes reuptake / internalization. The 62 kDa HL-V5 isoform progressively accumulates in the cell over 24h, with no detectible modification or degradation. Treatment of liver cells with ADP has no effect on HL-V5 internalization or degradation at 30 min and 4h.. These studies show that extracellular nucleotides act to prevent HL accumulation in the media by stimulating cellular autophagic degradation and blocking HL release. Topics: Adenine; Adenosine Diphosphate; Autophagy; Endocytosis; Hep G2 Cells; Hepatocytes; Humans; Leupeptins; Lipase; Microtubule-Associated Proteins; Protein Isoforms; Receptors, Purinergic P2; RNA Interference; RNA, Small Interfering; Time Factors | 2014 |
Protective effect of calpain inhibitor N-acetyl-L-leucyl-L-leucyl-L-norleucinal on acute alcohol consumption related cardiomyopathy.
Excessive alcohol consumption and alcoholism cause medical problems with high mortality and morbidity rates. In this study we aimed to decrease the alcohol related tissue damage by inhibiting calpain activation which plays an important role in apoptosis and necrosis, in rats with cardiomyopathy induced by acute alcohol consumption. Male Sprague-Dawley rats were separated into four groups (control, vehicle, alcohol and alcohol + inhibitor) with 10 rats in each. Control group received isocaloric maltose while vehicle group received isocaloric maltose with DMSO, and alcohol group received 8 g/kg absolute ethanol by gavage. Inhibitor group received 20 mg/kg calpain inhibitor 1 intraperitonally prior to alcohol administration. Calpain activities, cathepsin L levels and cytochrome c release rates were significantly increased in alcohol group compared to control group (p < 0.05). Serum CK MB and BNP levels of alcohol group were excessively increased compared to control group (respectively p < 0.001 and p < 0.01). Serum BNP levels of alcohol + inhibitor group were significantly (p < 0.05) decreased compared to alcohol group. In addition to these, histological evaluation of light microscope images and the results of DNA fragmentation and immunohistochemical caspase-3 activity results showed significant improvement of these parameters in alcohol + inhibitor group compared to alcohol group. Results of our biochemical and histological evaluation results revealed that the calpain inhibitor N-acetyl-leu-leu-norleucinal may have an ameliorating effect on acute alcohol consumption related cardiac tissue damage due to its effects on cell death pathways. Topics: Animals; Binge Drinking; Calpain; Cardiomyopathies; Caspase 3; Cathepsin B; Cathepsin L; Creatine Kinase, MB Form; Cysteine Proteinase Inhibitors; Cytochromes c; Disease Models, Animal; Humans; Immunohistochemistry; Leupeptins; Male; Myocardium; Nerve Tissue Proteins | 2014 |
UbcH10 overexpression increases carcinogenesis and blocks ALLN susceptibility in colorectal cancer.
Cyclins are essential for cell proliferation, the cell cycle and tumorigenesis in all eukaryotes. UbcH10 regulates the degradation of cyclins in a ubiquitin-dependent manner. Here, we report that UbcH10 is likely involved in tumorigenesis. We found that cancer cells exposed to n-acetyl-leu-leu-norleucinal (ALLN) treatment and UbcH10 depletion exhibit a synergistic therapeutic effect. Abundant expression of UbcH10 drives resistance to ALLN-induced cell death, while cells deficient in UbcH10 were susceptible to ALLN-induced cell death. The depletion of UbcH10 hindered tumorigenesis both in vitro and in vivo, as assessed by colony formation, growth curve, soft agar and xenograft assays. These phenotypes were efficiently rescued through the introduction of recombinant UbcH10. In the UbcH10-deficient cells, alterations in the expression of cyclins led to cell cycle changes and subsequently decreases in tumorigenesis. The tumorigenesis of xenograft tumors from UbcH10-deficient cells treated with ALLN was decreased relative to wild-type cells treated with ALLN in nude mice. On the molecular level, we observed that UbcH10 deficiency enhances the activation of caspase 8 and caspase 3 but not caspase 9 to impair cell viability upon ALLN treatment. Collectively, our results suggest that, as an oncogene, UbcH10 is a potential drug target for the treatment of colorectal cancer. Topics: Adenocarcinoma; Animals; Carcinogenesis; Caspases; Cell Cycle; Cell Death; Cell Line, Tumor; Cell Proliferation; Colorectal Neoplasms; Cyclins; Dependovirus; Female; Gene Expression Regulation, Neoplastic; Genetic Vectors; Humans; Leupeptins; Mice; Mice, Knockout; Mice, Nude; Signal Transduction; Ubiquitin; Ubiquitin-Conjugating Enzymes; Xenograft Model Antitumor Assays | 2014 |
N-terminal cleavage and release of the ectodomain of Flt1 is mediated via ADAM10 and ADAM 17 and regulated by VEGFR2 and the Flt1 intracellular domain.
Flt is one of the cell surface VEGF receptors which can be cleaved to release an N-terminal extracellular fragment which, like alternately transcribed soluble Flt1 (sFlt1), can antagonize the effects of VEGF. In HUVEC and in HEK293 cells where Flt1 was expressed, metalloprotease inhibitors reduced Flt1 N-terminal cleavage. Overexpression of ADAM10 and ADAM17 increased cleavage while knockdown of ADAM10 and ADAM17 reduced N-terminal cleavage suggesting that these metalloproteases were responsible for Flt1 cleavage. Protein kinase C (PKC) activation increased the abundance and the cleavage of Flt1 but this did not require any residues within the intracellular portion of Flt1. ALLN, a proteasomal inhibitor, increased the abundance of Flt1 which was additive to the effect of PKC. Removal of the entire cytosolic region of Flt1 appeared to stimulate cleavage of Flt1 and Flt1 was no longer sensitive to ALLN suggesting that the cytosolic region contained a degradation domain. Knock down of c-CBL, a ring finger ubiquitin ligase, in HEK293 cells increased the expression of Flt1 although it did not appear to require a previously published tyrosine residue (1333Y) in the C-terminus of Flt1. Increasing VEGFR2 expression increased VEGF-stimulated sFlt1 expression and progressively reduced the cleavage of Flt1 with Flt1 staying bound to VEGFR2 as a heterodimer. Our results imply that secreted sFlt1 and cleaved Flt1 will tend to have local effects as a VEGF antagonist when released from cells expressing VEGFR2 and more distant effects when released from cells lacking VEGFR2. Topics: ADAM Proteins; ADAM10 Protein; ADAM17 Protein; Amyloid Precursor Protein Secretases; Dipeptides; HEK293 Cells; Human Umbilical Vein Endothelial Cells; Humans; Hydroxamic Acids; Leupeptins; Matrix Metalloproteinase Inhibitors; Membrane Proteins; Mutation; Protein Structure, Tertiary; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factor Receptor-1; Vascular Endothelial Growth Factor Receptor-2 | 2014 |
ALLN hinders HCT116 tumor growth through Bax-dependent apoptosis.
Continual high expression of cysteine proteases calpain I and II have been implicated in tumorigenicity; conversely, N-acetyl-leu-leunorleucinal (ALLN), which inhibits calpain I and II, should also influence tumor growth and carcinogenesis. To explore the role of ALLN against colon cancer and in promoting apoptosis, we used colon cancer HCT116 cell lines, p53 or Bax-deficient HCT116 cell lines. Cell viability and tumor growth decreased in a concentration-dependent manner when treated with 0-26μM ALLN. Treatment with ALLN induced apoptosis in HCT116 cell; however, flow cytometry showed that apoptosis significantly decreased in Bax-deficient HCT116 cell lines, but not in p53-deficient HCT116 cell lines. In addition, the ALLN-induced apoptosis response was through Bax translocation from cytosol to mitochondria. In this study we showed intraperitoneally injected ALLN to inhibit colon tumor formation in nude mice, and found ALLN to inhibit tumor growth in colon cancer cells, mainly through apoptosis that depends on translocation of Bax to a mitochondrial endogenous pathway; this implies a molecular mechanism for ALLN against human colon cancer. These results suggest that ALLN could become a novel agent for prevention of colon cancer. Topics: Apoptosis; bcl-2-Associated X Protein; Cell Division; Cell Line, Tumor; Humans; Leupeptins | 2013 |
A role for the protein tyrosine phosphatase CD45 in macrophage adhesion through the regulation of paxillin degradation.
CD45 is a protein tyrosine phosphatase expressed on all cells of hematopoietic origin that is known to regulate Src family kinases. In macrophages, the absence of CD45 has been linked to defects in adhesion, however the molecular mechanisms involved remain poorly defined. In this study, we show that bone marrow derived macrophages from CD45-deficient mice exhibit abnormal cell morphology and defective motility. These defects are accompanied by substantially decreased levels of the cytoskeletal-associated protein paxillin, without affecting the levels of other proteins. Degradation of paxillin in CD45-deficient macrophages is calpain-mediated, as treatment with a calpain inhibitor restores paxillin levels in these cells and enhances cell spreading. Inhibition of the tyrosine kinases proline-rich tyrosine kinase (Pyk2) and focal adhesion kinase (FAK), kinases that are capable of mediating tyrosine phosphorylation of paxillin, also restored paxillin levels, indicating a role for these kinases in the CD45-dependent regulation of paxillin. These data demonstrate that CD45 functions to regulate Pyk2/FAK activity, likely through the activity of Src family kinases, which in turn regulates the levels of paxillin to modulate macrophage adhesion and migration. Topics: Animals; Blotting, Western; Calpain; Cell Adhesion; Cell Movement; Cell Shape; Cells, Cultured; Cysteine Proteinase Inhibitors; Cytoskeleton; Focal Adhesion Kinase 1; Focal Adhesion Kinase 2; Leukocyte Common Antigens; Leupeptins; Macrophages; Mice; Mice, Inbred C57BL; Mice, Knockout; Microscopy, Confocal; Paxillin; Phosphorylation; Proteolysis; src-Family Kinases; Time-Lapse Imaging | 2013 |
Proteasome inhibition in cancer is associated with enhanced tumor targeting by the adeno-associated virus/phage.
Bacteriophage (phage), which are viruses that infect bacteria only, have shown promise as vehicles for targeted cancer gene therapy, albeit with poor efficiency. Recently, we generated an improved version of phage vectors by incorporating cis genetic elements of adeno-associated virus (AAV). This novel AAV/phage hybrid (AAVP) efficiently delivered systemically administered therapeutic genes to various tumor targets by displaying an integrin tumor-targeting ligand on the phage capsid. However, inherent limitations in bacteriophage mean that these AAVP vectors still need to be improved. One of the limitations of AAVP in mammalian cells may be its susceptibility to proteasomal degradation. The proteasome is upregulated in cancer and it is known that it constitutes a barrier to gene delivery by certain eukaryotic viruses. We report here that inhibition of proteasome improved targeted reporter gene delivery by AAVP in cancer cells in vitro and in tumors in vivo after intravenous vector administration to tumor-bearing mice. We also show enhanced targeted tumor cell killing by AAVP upon proteasome inhibition. The AAVP particles persisted significantly in cancer cells in vitro and in tumors in vivo after systemic administration, and accumulated polyubiquitinated coat proteins. Our results suggest that the proteasome is indeed a barrier to tumor targeting by AAVP and indicate that a combination of proteasome-inhibiting drugs and AAVP should be considered for clinical anticancer therapy. Topics: Animals; Bacteriophages; Cell Line; Cell Line, Tumor; Dependovirus; Fluorescent Antibody Technique; Humans; Leupeptins; Mice; Proteasome Endopeptidase Complex | 2013 |
ALLN rescues an in vitro excitatory synaptic transmission deficit in Lis1 mutant mice.
LIS1 gene mutations lead to a rare neurological disorder, classical lissencephaly, characterized by brain malformations, mental retardation, seizures, and premature death. Mice heterozygous for Lis1 (Lis1(+/-)) exhibit cortical malformations, defects in neuronal migration, increased glutamate-mediated synaptic transmission, and spontaneous electrographic seizures. Recent work demonstrated that in utero treatment of Lis1(+/-) mutant dams with ALLN, a calpain inhibitor, partially rescues neuronal migration defects in the offspring. Given the challenges of in utero drug administration, we examined the therapeutic potential of ALLN on postnatal lissencephalic cells. Voltage- and current-clamp studies were performed with acute hippocampal slices obtained from Lis1 mutant mice and age-matched littermate control mice. Specifically, we determined whether postnatal ALLN treatment can reverse excitatory synaptic transmission deficits, namely, an increase in spontaneous and miniature excitatory postsynaptic current (EPSC) frequency, on CA1 pyramidal neurons observed in tissue slices from Lis1(+/-) mice. We found that acute application of ALLN restored spontaneous and miniature EPSC frequencies to wild-type levels without affecting inhibitory postsynaptic synaptic current. Furthermore, Western blot analysis of protein expression, including proteins involved in excitatory synaptic transmission, demonstrated that ALLN blocks the cleavage of the calpain substrate αII-spectrin but does not rescue Lis1 protein levels in Lis1(+/-) mutants. Topics: 1-Alkyl-2-acetylglycerophosphocholine Esterase; Animals; Calpain; Cysteine Proteinase Inhibitors; Excitatory Postsynaptic Potentials; Gene Expression; Heterozygote; Leupeptins; Lissencephaly; Mice; Mice, Mutant Strains; Microtubule-Associated Proteins; Miniature Postsynaptic Potentials; Mutation; Proteolysis; Pyramidal Cells; Spectrin | 2013 |
Endoplasmic spreading requires coalescence of vimentin intermediate filaments at force-bearing adhesions.
For cells to develop long-range forces and carry materials to the periphery, the microtubule and organelle-rich region at the center of the cell-the endoplasm-needs to extend to near the cell edge. Depletion of the actin cross-linking protein filamin A (FlnA) causes a collapse of the endoplasm into a sphere around the nucleus of fibroblasts and disruption of matrix adhesions, indicating that FlnA is involved in endoplasmic spreading and adhesion growth. Here, we report that treatment with the calpain inhibitor N-[N-(N-acetyl-l-leucyl)-l-leucyl]-l-norleucine (ALLN) restores endoplasmic spreading as well as focal adhesion (FA) growth on fibronectin-coated surfaces in a Fln-depleted background. Addback of calpain-uncleavable talin, not full-length talin, achieves a similar effect in Fln-depleted cells and indicates a crucial role for talin in endoplasmic spreading. Because FA maturation involves the vimentin intermediate filament (vIF) network, we also examined the role of vIFs in endoplasmic spreading. Wild-type cells expressing a vimentin variant incapable of polymerization exhibit deficient endoplasmic spreading as well as defects in FA growth. ALLN treatment restores FA growth despite the lack of vIFs but does not restore endoplasmic spreading, implying that vIFs are essential for endoplasm spreading. Consistent with that hypothesis, vIFs are always displaced from adhesions when the endoplasm does not spread. In Fln-depleted cells, vIFs extend beyond adhesions, nearly to the cell edge. Finally, inhibiting myosin II-mediated contraction blocks endoplasmic spreading and adhesion growth. Thus we propose a model in which myosin II-mediated forces and coalescence of vIFs at mature FAs are required for endoplasmic spreading. Topics: Animals; Calpain; Cell Movement; Cells, Cultured; Contractile Proteins; Cysteine Proteinase Inhibitors; Cytoskeleton; Embryo, Mammalian; Fibroblasts; Filamins; Focal Adhesions; Immunohistochemistry; Leupeptins; Luminescent Proteins; Mice; Mice, Knockout; Microfilament Proteins; Microscopy, Fluorescence; Microtubules; Models, Biological; Myosin Type II; RNA Interference; Talin; Vimentin | 2013 |
Calpains participate in nerve terminal degeneration induced by spider and snake presynaptic neurotoxins.
α-latrotoxin and snake presynaptic phospholipases A2 neurotoxins target the presynaptic membrane of axon terminals of the neuromuscular junction causing paralysis. These neurotoxins display different biochemical activities, but similarly alter the presynaptic membrane permeability causing Ca(2+) overload within the nerve terminals, which in turn induces nerve degeneration. Using different methods, here we show that the calcium-activated proteases calpains are involved in the cytoskeletal rearrangements that we have previously documented in neurons exposed to α-latrotoxin or to snake presynaptic phospholipases A2 neurotoxins. These results indicate that calpains, activated by the massive calcium influx from the extracellular medium, target fundamental components of neuronal cytoskeleton such as spectrin and neurofilaments, whose cleavage is functional to the ensuing nerve terminal fragmentation. Topics: Acrylates; Animals; Animals, Newborn; Calcium Signaling; Calpain; Cell Membrane Permeability; Cells, Cultured; Cytoskeleton; Dipeptides; Leupeptins; Nerve Degeneration; Neurofilament Proteins; Neurons; Neurotoxins; Phospholipases A2; Presynaptic Terminals; Rats; Rats, Wistar; Snake Venoms; Spectrin; Spider Venoms | 2013 |
Anti-malarial activity of new N-acetyl-L-leucyl-L-leucyl-L-norleucinal (ALLN) derivatives against Plasmodium falciparum.
Malaria is the most common of the parasitic diseases in tropical and subtropical regions. Adverse side effects of anti-malarial drugs have precluded them as a potential clinical drug. In this study, novel derivatives of N-acetyl-L-leucyl-L-leucyl-L-norleucinal (ALLN) based on a variety of dipeptidyl α,β-unsaturated amides containing lysine as a part were synthesized and evaluated. Lower toxicity was achieved by reducing or eliminating the tendency of forming chemically reactive and toxic intermediates and metabolites. The synthesized compounds were evaluated for anti-malarial efficacy against Plasmodium falciparum and cytotoxicity in human epitheloid carcinoma cervix (HeLa cells) by estimating the therapeutic index (TI). N-Methyl amide with N'-Boc protection among them exhibited strong anti-malarial activity and N-methyl amide with N'-m-methylbenzyl amide showed excellent anti-malarial activity with much lower toxicity than the ALLN. Therefore, the two chemicals, as well as the underlying design rationale, could be useful in the discovery and development of new anti-malarial drugs. Topics: Amides; Antimalarials; Dipeptides; HeLa Cells; Humans; Leupeptins; Plasmodium falciparum | 2013 |
Presenilin-2 regulates the degradation of RBP-Jk protein through p38 mitogen-activated protein kinase.
Transcriptional regulation performs a central role in Notch1 signaling by recombining binding protein Suppressor of Hairless (RBP-Jk)--a signaling pathway that is widely involved in determination of cell fate. Our earlier work demonstrated the possible regulation of the Notch1-RBP-Jk pathway through protein degradation of RBP-Jk; however, the potential regulator for the degradation of RBP-Jk remains to be determined. Here, we report that the expression of endogenous and exogenous RBP-Jk was increased significantly in cells treated with proteasome- and lysosome-specific inhibitors. The effects of these inhibitors on RBP-Jk occurred in a dose- and time-dependent manner. The level of RBP-Jk protein was higher in presenilin-2 (PS2)-knockout cells than in presenilin-1 (PS1)-knockout cells. Furthermore, the level of RBP-Jk was decreased by expression of PS2 in PS1 and PS2 double-knockout cells. We also found that PS1-knockout cells treated with a specific inhibitor of p38 mitogen-activated protein kinase ∂ (MAPK) had significantly increased levels of RBP-Jk. p38 MAPK phosphorylates RBP-Jk at Thr339 by physical binding, which subsequently induces the degradation and ubiquitylation of the RBP-Jk protein. Collectively, our results indicate that PS2 modulates the degradation of RBP-Jk through phosphorylation by p38 MAPK. Topics: Acetylcysteine; Ammonium Chloride; Cell Line; Chloroquine; Cysteine Proteinase Inhibitors; Gene Knockout Techniques; HEK293 Cells; Humans; Immunoglobulin J Recombination Signal Sequence-Binding Protein; Leupeptins; Lysosomes; MAP Kinase Signaling System; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Presenilin-1; Presenilin-2; Proteasome Inhibitors; Proteolysis; Receptor, Notch1; Transcription, Genetic; Transcriptional Activation; Ubiquitination | 2012 |
Degradation of mouse NTE-related esterase by macroautophagy and the proteasome.
NTE-related esterase (NRE) is a novel endoplasmic reticulum-anchored lysophospholipase with high homology to neuropathy target esterase (NTE). However, little is known about the regulation of NRE protein. In the current study, we investigated the degradation pathways of mouse NRE (mNRE) in mammalian cells. Based on experiments with inhibitors and inducer of protein degradation pathways, we provide here the first evidence that mNRE is degraded by macroautophagy as well as by the proteasome. Moreover, the contribution of protein domains to the degradation of mNRE was investigated, which showed that the transmembrane and regulatory domain played a role in the degradation of mNRE by macroautophagy and the proteasome respectively. In contrast the C-terminal catalytic domain was not involved in both degradation pathways of mNRE. These findings showed for the first time that the degradation pathways in controlling mNRE quantity and may provide further insight into structure and regulation of mNRE. Topics: Adenine; Animals; Autophagy; Carboxylic Ester Hydrolases; Cycloheximide; Green Fluorescent Proteins; HEK293 Cells; Humans; Leupeptins; Mice; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Biosynthesis; Protein Structure, Tertiary; Proteolysis; Recombinant Fusion Proteins | 2012 |
Synthesis and evaluation of peptidyl α,β-unsaturated carbonyl derivatives as anti-malarial calpain inhibitors.
Malarial calpain is a cysteine protease believed to be a central mediator essential for parasitic activities. N-Acetyl-L-leucyl-L-leucyl-L-norleucinal (ALLN), a calpain inhibitor, showed an excellent inhibitory effect on the erythrocytic stages of Plasmodium falciparum. However the aldehyde group of ALLN makes it susceptible to metabolism. Therefore, we designed α,β-unsaturated carbonyl peptides that could serve as electrophiles for cysteine residues in calpain. Among the synthetic analogs based on the structure of ALLN, peptidyl esters 7, 8 and 9 showed the most potent anti-malarial effects, with the same IC50 values of 5.0 μM. Also they showed the high selective toxicity for the malaria versus Hela cell with 40.6, 69.2 and 24.3 fold for 7, 8 and 9, respectively. Dipeptidyl α,β-unsaturated carbonyl derivatives consisting of two amino acids gave better anti-malarial effects than those consisting with one amino acid. The fluctuation in anti-malarial activity with small changes in chemical structure indicates the possibilities of improving synthetic analogs. Topics: Antimalarials; Calpain; Cell Proliferation; Chromatography; Cysteine Proteinase Inhibitors; Flow Cytometry; HeLa Cells; Humans; Leupeptins; Magnetic Resonance Spectroscopy; Molecular Structure; Parasitic Sensitivity Tests; Plasmodium falciparum; Protozoan Proteins; Spectroscopy, Fourier Transform Infrared; Structure-Activity Relationship | 2012 |
Calpain inhibitors exhibit matrix metalloproteinase-2 inhibitory activity.
Matrix metalloproteinase (MMP)-2 is a zinc-dependent endopeptidase which, alongside its known extracellular actions, plays fundamental roles in oxidative stress-induced injury to the heart. Intracellular cleavage targets of MMP-2 selectively mediating this injury include the sarcomeric proteins troponin I, myosin light chain-1 and titin; some of these are also targeted by calpains. In myocardial ischemia and reperfusion injury, inhibitors of MMP-2 and some calpain inhibitors were shown to improve the recovery of contractile function. We hypothesized that the protective effects of calpain inhibitors may be due in part to their ability to inhibit MMP-2. Four calpain inhibitors (calpain inhibitor III, ALLM, ALLN, and PD-150606) were tested for their ability to inhibit MMP-2 in comparison to the selective MMP inhibitor ONO-4817. At 100 μM, all calpain inhibitors, except ALLM, showed significant inhibition of MMP-2 gelatinolytic activity. When assessed by the troponin I proteolysis assay, both ALLN and PD-150606, but neither ALLM nor calpain inhibitor III (at 20 μM), significantly inhibited MMP-2 activity. Using a fluorogenic MMP substrate peptide OmniMMP in a kinetic assay the rank order of IC(50) values against MMP-2 were: PD-150606 Topics: Acrylates; Calpain; Catalysis; Cell Line; Cysteine Proteinase Inhibitors; Dipeptides; Gelatin; Humans; Inhibitory Concentration 50; Leupeptins; Matrix Metalloproteinase 2; Matrix Metalloproteinase Inhibitors; Oligopeptides; Phenyl Ethers; Troponin I | 2012 |
Regulated specific proteolysis of the Cajal body marker protein coilin.
Cajal bodies (CB) are subnuclear domains that contain various proteins with diverse functions including the CB marker protein coilin. In this study, we investigate the proteolytic activity of calpain on coilin. Here, we report a 28-kDa cleaved coilin fragment detected by two coilin antibodies that is cell cycle regulated, with levels that are consistently reduced during mitosis. We further show that an in vitro calpain assay with full-length or C-terminal coilin recombinant protein releases the same size cleaved fragment. Furthermore, addition of exogenous RNA to purified coilin induces proteolysis by calpain. We also report that the relative levels of this cleaved coilin fragment are susceptible to changes induced by various cell stressors, and that coilin localization is affected by inhibition or knockdown of calpain both under normal and stressed conditions. Collectively, our data suggest that coilin is subjected to regulated specific proteolysis by calpain, and this processing may play a role in the regulation of coilin activity and CB formation. Topics: Calpain; Cell Cycle; Cell Nucleolus; Coiled Bodies; HeLa Cells; Humans; Leupeptins; Nuclear Proteins; Proteolysis | 2012 |
Proteasome inhibitors induce nucleolar aggregation of proteasome target proteins and polyadenylated RNA by altering ubiquitin availability.
The ubiquitin-proteasome pathway is essential for most cellular processes, including protein quality control, cell cycle, transcription, signaling, protein transport, DNA repair and stress responses. Hampered proteasome activity leads to the accumulation of polyubiquitylated proteins, endoplastic reticulum (ER) stress and even cell death. The ability of chemical proteasome inhibitors (PIs) to induce apoptosis is utilized in cancer therapy. During PI treatment, misfolded proteins accrue to cytoplasmic aggresomes. The formation of aggresome-like structures in the nucleus has remained obscure. We identify here a nucleolus-associated RNA-protein aggregate (NoA) formed by the inhibition of proteasome activity in mammalian cells. The aggregate forms within the nucleolus and is dependent on nucleolar integrity, yet is a separate structure, lacking nucleolar marker proteins, ribosomal RNA (rRNA) and rRNA synthesis activity. The NoAs contain polyadenylated RNA, conjugated ubiquitin and numerous nucleoplasmic proteasome target proteins. Several of these are key factors in oncogenesis, including transcription factors p53 and retinoblastoma protein (Rb), several cell cycle-regulating cyclins and cyclin-dependent kinases (CDKs), and stress response kinases ataxia-telangiectasia mutated (ATM) and Chk1. The aggregate formation depends on ubiquitin availability, as shown by modulating the levels of ubiquitin and deubiquitinases. Furthermore, inhibition of chromosome region maintenance 1 protein homolog (CRM1) export pathway aggravates the formation of NoAs. Taken together, we identify here a novel nuclear stress body, which forms upon proteasome inactivity within the nucleolus and is detectable in mammalian cell lines and in human tissue. These findings show that the nucleolus controls protein and RNA surveillance and export by the ubiquitin pathway in a previously unidentified manner, and provide mechanistic insight into the cellular effects of PIs. Topics: Acetylcysteine; Ataxia Telangiectasia Mutated Proteins; Cell Cycle Proteins; Cell Line; Cell Nucleolus; Checkpoint Kinase 1; Cyclin-Dependent Kinases; Cyclins; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Humans; Leucine; Leupeptins; Nuclear Proteins; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Kinases; Protein Serine-Threonine Kinases; RNA, Messenger; Transcription Factors; Tumor Suppressor Proteins; Ubiquitin | 2011 |
Sur8/Shoc2 involves both inhibition of differentiation and maintenance of self-renewal of neural progenitor cells via modulation of extracellular signal-regulated kinase signaling.
Sur8/Shoc2 is a scaffold protein that regulates the Ras-extracellular signal-regulated kinase (ERK) pathway. However, the roles of Sur8 in cellular physiologies are poorly understood. In this study, Sur8 was severely repressed in the course of neural progenitor cell (NPC) differentiation in the cerebral cortex of developing rat embryos. Similarly, Sur8 was also critically reduced in cultured NPCs, which were induced differentiation by removal of basic fibroblast growth factor (bFGF). Sur8 regulation occurs at the protein level rather than at the mRNA level as revealed by both in situ hybridization and reverse transcriptase polymerase chain reaction analyses. The role of Sur8 in NPC differentiation was confirmed by lentivirus-mediated Sur8 knockdown, which resulted in increased differentiation, whereas exogenous expression of Sur8 inhibited differentiation. Contrastingly, NPC proliferation was promoted by overexpression, but was suppressed by Sur8 knockdown. The role of Sur8 as an antidifferentiation factor in the developing rat brain was confirmed by an ex vivo embryo culture system combined with the lentivirus-mediated Sur8 knockdown. The numbers and sizes of neurospheres were reduced, but neuronal outgrowth was enhanced by the Sur8 knockdown. The Ras-ERK pathway is involved in Sur8-mediated regulations of differentiation, as the treatment of ERK kinase (MEK) inhibitors blocks the effects of Sur8. The regulations of NPCs' differentiation and proliferation by the Ras-ERK pathway were also shown by the rescues of the effects of bFGF depletion, neuronal differentiation, and antiproliferation by epidermal growth factor. In summary, Sur8 is an antidifferentiation factor that stimulates proliferation for maintenance of self-renewal in NPCs via modulation of the Ras-ERK pathway. Topics: Animals; Brain; Cell Differentiation; Cell Proliferation; Extracellular Signal-Regulated MAP Kinases; Fibroblast Growth Factor 2; Intracellular Signaling Peptides and Proteins; Leupeptins; MAP Kinase Signaling System; Neural Stem Cells; Rats; Rats, Sprague-Dawley; RNA Interference; RNA, Messenger; RNA, Small Interfering | 2011 |
A lipid-droplet-targeted O-GlcNAcase isoform is a key regulator of the proteasome.
Protein-O-linked N-Acetyl-β-D-glucosaminidase (O-GlcNAcase, OGA; also known as hexosaminidase C) participates in a nutrient-sensing, hexosamine signaling pathway by removing O-linked N-acetylglucosamine (O-GlcNAc) from key target proteins. Perturbations in O-GlcNAc signaling have been linked to Alzheimer's disease, diabetes and cancer. Mammalian O-GlcNAcase exists as two major spliced isoforms differing only by the presence (OGA-L) or absence (OGA-S) of a histone-acetyltransferase domain. Here we demonstrate that OGA-S accumulates on the surface of nascent lipid droplets with perilipin-2; both of these proteins are stabilized by proteasome inhibition. We show that selective downregulation of OGA-S results in global proteasome inhibition and the striking accumulation of ubiquitinylated proteins. OGA-S knockdown increased levels of perilipin-2 and perilipin-3 suggesting that O-GlcNAc-dependent regulation of proteasomes might occur on the surface of lipid droplets. By locally activating proteasomes during maturation of the nascent lipid droplet, OGA-S could participate in an O-GlcNAc-dependent feedback loop regulating lipid droplet surface remodeling. Our findings therefore suggest a mechanistic link between hexosamine signaling and lipid droplet assembly and mobilization. Topics: Animals; beta-N-Acetylhexosaminidases; Cysteine Proteinase Inhibitors; Cytoplasmic Vesicles; HeLa Cells; Humans; Leupeptins; Lipid Metabolism; Membrane Proteins; Mice; Perilipin-2; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Isoforms; Protein Transport; Proteolysis; RNA, Small Interfering | 2011 |
Stimulation of human formyl peptide receptors by calpain inhibitors: homology modeling of receptors and ligand docking simulation.
Calpain inhibitors, including peptide aldehydes (N-acetyl-Leu-Leu-Nle-CHO and N-acetyl-Leu-Leu-Met-CHO) and α-mercapto-acrylic acid derivatives (PD150606 and PD151746), have been shown to stimulate phagocyte functions via activation of human formyl peptide receptor (hFPR) and/or hFPR-like 1 (hFPRL1). Using the homology modeling of the receptors and the ligand docking simulation, here we show that these calpain inhibitors could bind to the putative N-formyl-Met-Leu-Phe (fMLF) binding site on hFPR and/or hFPRL1. The studies with HEK-293 cells stably expressing hFPR or hFPRL1 showed that the concentrations of calpain inhibitors required to induce an increase in cytoplasmic free Ca(2+) ([Ca(2+)](i)) was much higher (>100 folds) than those of fMLF and Trp-Lys-Tyr-Met-Val-D-Met (WKYMVm). HEK-293 cells expressing hFPR or hFPRL1 with the mutated fMLF binding site never exhibited the [Ca(2+)](i) response to calpain inhibitors. When the optimal concentrations of each stimulus were used, pretreatment of cells with fMLF or WKYMVm abolished an increase in [Ca(2+)](i) induced by calpain inhibitors as well as the same stimulus, whereas pretreatment of cells with calpain inhibitors significantly suppressed, but never abolished, the [Ca(2+)](i) response induced by fMLF or WKYMVm, suggesting that the binding affinity of the inhibitors to the putative fMLF binding site may be lower than that of fMLF or WKYMVm. Topics: Acrylates; Base Sequence; Binding Sites; Calcium Signaling; Computer Simulation; Cysteine Proteinase Inhibitors; DNA Primers; Glycoproteins; HEK293 Cells; Humans; Leupeptins; Ligands; Models, Molecular; N-Formylmethionine Leucyl-Phenylalanine; Oligopeptides; Receptors, Formyl Peptide; Receptors, Lipoxin; Recombinant Proteins; Structural Homology, Protein | 2011 |
Targeted cancer stem cell therapies start with proper identification of the target.
Topics: Antineoplastic Agents; Brain Neoplasms; Cell Culture Techniques; Cysteine Proteinase Inhibitors; Drug Resistance, Neoplasm; Glioma; Humans; Leupeptins; Neoplastic Stem Cells; Phenotype; Proteasome Endopeptidase Complex; Reproducibility of Results | 2010 |
Activation of mitochondrial calpain and release of apoptosis-inducing factor from mitochondria in RCS rat retinal degeneration.
The present study was performed to investigate changes of cytosolic and mitochondrial calpain activities, and effects of intravitreously injected calpain inhibitor on photoreceptor apoptosis in Royal College of Surgeon's (RCS) rats. Time courses of activities for both cytosolic and mitochondrial calpains and amount of calpastatin in RCS rat retina were analyzed by subcellular fractionation, calpain assay and western blotting. Calpain assay was colorimetrically performed using Suc-LLVY-Glo as substrate. Effects of intravitreously injected calpain inhibitor (ALLN and PD150606) on RCS rat retinal degeneration were analyzed by TUNEL staining. Effects of mitochondrial calpain activity on activation and translocation of apoptosis-inducing factor (AIF) were analyzed by western blotting. Mitochondrial calpain started to be significantly activated at postnatal (p) 28 days in RCS rat retina, whereas cytosolic micro-calpain was activated at p 35 days, although specific activity of mitochondrial calpain was 13% compared to cytosolic micro-calpain. Intravitreously injected ALLN and PD150606 effectively inhibited photoreceptor apoptosis only when injected at p 25 days, but did not inhibit photoreceptor apoptosis when injected at p 32 days. Parts of AIF were truncated/activated by mitochondrial calpains and translocated to the nucleus. These results suggest that 1), calpain presents not only in the cytosolic fraction but also in the mitochondrial fraction in RCS rat retina; 2), mitochondrial calpain is activated earlier than cytosolic calpain during retinal degeneration in RCS rats; 3), photoreceptor apoptosis may be regulated by not only calpain systems but also other mechanisms; 4), mitochondrial calpain may activate AIF to induce apoptosis; and 5), calpain inhibitors may be partially effective to inhibit photoreceptor apoptosis in RCS rats. The present study provides new insights into the molecular basis for photoreceptor apoptosis in RCS rats and the future possibility of new pharmaceutical treatments for retinitis pigmentosa. Topics: Acrylates; Animals; Apoptosis; Apoptosis Inducing Factor; Blotting, Western; Calcium-Binding Proteins; Calpain; Cysteine Proteinase Inhibitors; Cytosol; Electrophoresis, Polyacrylamide Gel; Immunoenzyme Techniques; In Situ Nick-End Labeling; Leupeptins; Mitochondria; Photoreceptor Cells, Vertebrate; Rats; Rats, Mutant Strains; Retinal Degeneration | 2010 |
Extensive autolytic fragmentation of membranous versus cytosolic calpain following myocardial ischemia-reperfusion.
We investigated calpain activation in the heart during ischemia-reperfusion (I-R) by immunologically mapping the fragmentation patterns of calpain and selected calpain substrates. Western blots showed the intact 78 kDa large subunit of membrane-associated calpain was autolytically fragmented to 56 and 43 kDa signature immunopeptides following I-R. Under these conditions, the 78 kDa calpain large subunit from crude cytosolic fractions was markedly less fragmented, with only weakly stained autolytic peptides detected at higher molecular weights (70 and 64 kDa). Western blots also showed corresponding calpain-like degradation products (150 and 145 kDa) of membrane-associated alpha-fodrin (240 kDa) following I-R, but in crude myofibrils alpha-fodrin degradation occurred in a manner uncharacteristic of calpain. For control hearts perfused in the absence of ischemia, autolytic fragmentation of calpain and calpain-like alpha-fodrin degradation were completely absent from most subcellular fractions. The exception was sarcolemma-enriched membranes, where significant calpain autolysis and calpain-like alpha-fodrin degradation were detected. In purified sarcoplasmic reticulum membranes, RyR2 and SERCA2 proteins were also highly degraded, but for RyR2 this did not occur in a manner characteristic of calpain. When I-R-treated hearts were perfused with peptidyl calpain inhibitors (ALLN or ALLM; 25 micromol/L), calpain autolysis and calpain-like degradation of alpha-fodrin were equally attenuated by each inhibitor. However, only ALLN protected against early loss of developed pressure in hearts following I-R, with no functionally protective effect of ALLM observed. Our studies suggest calpain is preferentially activated at membranes following I-R, possibly contributing to impaired ion channel function implicated by others in I-R injury. Topics: Animals; Autolysis; Blotting, Western; Calpain; Cell Fractionation; Cytosol; Disease Models, Animal; Dogs; Electrophoresis, Polyacrylamide Gel; Intracellular Membranes; Leupeptins; Male; Muscle, Skeletal; Myocardial Reperfusion Injury; Myocardium; Oligopeptides; Rabbits; Rats; Rats, Sprague-Dawley | 2010 |
The production of 85 kDa N-terminal fragment of apolipoprotein B in mutant HepG2 cells generated by targeted modification of apoB gene occurs by ALLN-inhibitable protease cleavage during translocation.
To study the mechanism of low levels of full length and truncated apoB in individuals heterozygous for apoB truncation, a non-sense mutation was introduced in one of the three alleles of apob gene of HepG2 cells by homologous recombination. Despite very low levels of apoB-82 (1-2%) in the media, a prominent N-terminal apoB protein of 85 kDa (apoB-15) was secreted that fractionated at d>1.065 in density gradient ultracentrifugation. The mechanism of production of this short protein was studied by (35)S-methionine pulse-chase experiment. Oleate prevented presecretory degradation of apoB-100 in the cell and resulted in increased secretion of newly synthesized apoB-100 with decreases in the apoB-15, suggesting that rescue of pre-secretary intracellular degradation of apoB restricted the production and secretion of apoB-15. Further investigation on the degradation of transmembrane forms of apoB, in the presence and absence of a cysteine protease inhibitor, N-acetyl-leucyl-leucyl-norleucinal (ALLN), showed appearance of detectable levels of newly synthesized apoB-82 in the cell and the media together with increased apoB-100 secretion, and reduction in the secretion of apoB-15. Compared to ER membrane, the levels of apoB were higher in the luminal content, and presence of both oleate and ALLN had additive effect on apoB secretion. These results suggest that the presence of improper folding of apoB during translocation led to the cleavage of both apoB-100 and apoB-82 by ALLN-sensitive protease and generation of 85 kDa N-terminal fragment of apoB. Topics: Apolipoproteins B; Codon, Nonsense; Cysteine Proteases; Cysteine Proteinase Inhibitors; Hep G2 Cells; Humans; Hypobetalipoproteinemia, Familial, Apolipoprotein B; Leupeptins; Protein Folding; Protein Transport | 2010 |
Calpain modulates capacitation and acrosome reaction through cleavage of the spectrin cytoskeleton.
Research on fertilization in mammalian species has revealed that Ca(2+) is an important player in biochemical and physiological events enabling the sperm to penetrate the oocyte. Ca(2+) is a signal transducer that particularly mediates capacitation and acrosome reaction (AR). Before becoming fertilization competent, sperm must experience several molecular, biochemical, and physiological changes where Ca(2+) plays a pivotal role. Calpain-1 and calpain-2 are Ca(2+)-dependent proteases widely studied in mammalian sperm; they have been involved in capacitation and AR but little is known about their mechanism. In this work, we establish the association of calpastatin with calpain-1 and the changes undergone by this complex during capacitation in guinea pig sperm. We found that calpain-1 is relocated and translocated from cytoplasm to plasma membrane (PM) during capacitation, where it could cleave spectrin, one of the proteins of the PM-associated cytoskeleton, and facilitates AR. The aforementioned results were dependent on the calpastatin phosphorylation and the presence of extracellular Ca(2+). Our findings underline the contribution of the sperm cytoskeleton in the regulation of both capacitation and AR. In addition, our findings also reveal one of the mechanisms by which calpain and calcium exert its function in sperm. Topics: Acrosome Reaction; Animals; Blotting, Western; Calcium-Binding Proteins; Calpain; Cytoskeleton; Dipeptides; Guinea Pigs; Leupeptins; Male; Microscopy, Electron; Spectrin; Sperm Capacitation; Spermatozoa | 2010 |
HIV-1 Tat C-terminus is cleaved by calpain 1: implication for Tat-mediated neurotoxicity.
HIV-Encephalopathy (HIVE) is a common neurological disorder associated with HIV-1 infection and AIDS. The activity of the HIV trans-activating protein Tat is thought to contribute to neuronal pathogenesis. While Tat proteins from primary virus isolates consist of 101 or more amino acids, 72 and 86 amino acids forms of Tat are commonly used for in vitro studies. Although Tat72 contains the minimal domain required for viral replication, other activities of Tat appear to vary according to its length, sub-cellular localization, cell type and the stage of cellular differentiation. In this study, we investigated the stability of intracellular Tat101 during proliferation and differentiation of neuronal cells in culture. We have utilized rat neuronal progenitors as a model of neuronal cell proliferation and differentiation, as well as rat primary cortical neurons as a model of fully differentiated cells. Our results indicate that, upon internalization, Tat101 was degraded more rapidly in proliferating cells than in cells which either underwent neuronal differentiation or were fully differentiated. Intracellular degradation of Tat was prevented by the calpain 1 inhibitor, ALLN, in both proliferating and differentiated cells. Inhibition of calpain 1 by calpastatin peptide also prevented Tat cleavage. In vitro calpain digestion and mass spectrometry analysis further demonstrated that the sequence of Tat sensitive to calpain cleavage was located in the C-terminus of this viral protein, between amino acids 68 and 69. Moreover, cleavage of Tat101 by calpain 1 increased neurotoxic effect of this viral protein and presence of the calpain inhibitor protected neuronal cells from Tat-mediated toxicity. Topics: Amino Acid Sequence; Animals; Calpain; Cell Differentiation; Cell Nucleus; Cell Proliferation; Cells, Cultured; Cerebral Cortex; Enzyme Inhibitors; Intracellular Space; Leupeptins; Molecular Sequence Data; Neurons; Protein Processing, Post-Translational; Protein Stability; Protein Transport; Rats; Stem Cells; Subcellular Fractions; tat Gene Products, Human Immunodeficiency Virus | 2009 |
An ezrin/calpain/PI3K/AMPK/eNOSs1179 signaling cascade mediating VEGF-dependent endothelial nitric oxide production.
Calpain was recently reported to mediate vascular endothelial growth factor (VEGF)-induced angiogenesis. In the present study, we investigated detailed molecular mechanisms. VEGF (100 ng/mL) induced a marked increase in endothelial cell production of NO(*), specifically detected by electron spin resonance. This response was abolished by inhibition of calpain with N-acetyl-leucyl-leucyl-norleucinal (ALLN) or Calpeptin. Both also diminished membrane-specific calpain activation by VEGF, which was intriguingly attenuated by silencing ezrin with RNA interference. A rapid membrane colocalization of calpain and ezrin occurred as short as 10 minutes after VEGF stimulation. AKT, AMP-dependent kinase (AMPK), and endothelial nitric oxide synthase (eNOS)(s1179) phosphorylations in VEGF-stimulated endothelial cells were markedly enhanced, which were however significantly attenuated by either ALLN, Calpeptin, or ezrin small interfering RNA, as well as by Wortmannin or compound C (respectively for phosphatidylinositol 3-kinase [PI3K] or AMPK). The latter 3 also abolished VEGF induction of NO(*). These data indicate that AMPK and AKT are both downstream of PI3K and that AKT activation is partially dependent on AMPK. The interrelationship between AMPK and AKT, although known to be individually important in mediating VEGF activation of eNOS, is clearly characterized. Furthermore, AMPK/AKT/eNOS(s1179) was found downstream of a calpain/ezrin membrane interaction. These data no doubt provide new insights into the long mystified signaling gap between VEGF receptors and PI3K/AKT or AMPK-dependent eNOS activation. In view of the well-established significance of VEGF-dependent angiogenesis, these findings might have broad and important implications in cardiovascular pathophysiology. Topics: AMP-Activated Protein Kinases; Animals; Aorta; Calpain; Cattle; Cells, Cultured; Cytoskeletal Proteins; Dipeptides; Endothelial Cells; Endothelium, Vascular; Enzyme Activation; Hydrogen Peroxide; Leupeptins; Membrane Proteins; Nitric Oxide; Nitric Oxide Synthase Type III; Phosphatidylinositol 3-Kinases; Phosphorylation; Protein Interaction Mapping; Protein Processing, Post-Translational; Proto-Oncogene Proteins c-akt; RNA, Small Interfering; Signal Transduction; Vascular Endothelial Growth Factor A | 2009 |
Calpain-mediated degradation of G-substrate plays a critical role in retinal excitotoxicity for amacrine cells.
The role of neuronal N-methyl-D-aspartate (NMDA) receptor-mediated intracellular signaling has been elucidated in both physiological and pathological conditions. However, the details of relative vulnerability for excitotoxicity remain unknown. Retinal excitotoxicity is involved in various diseases leading to irreversible blindness. Here, we used the visual system and explored the mechanistic details of the NMDA-elicited intracellular events, especially in the amacrine cells, which are the most vulnerable type of neuron in the retina. G-substrate, a specific substrate of cyclic guanosine 3',5'-monophosphate (cGMP)-dependent protein kinase, is colocalized with amacrine cells and acts as an endogenous inhibitor of protein phosphatase. To elucidate how G-substrate was involved in NMDA-induced amacrine cell death, the immunohistochemical analysis with G-substrate antibody was performed following NMDA injury. In vivo, NMDA immediately decreased G-substrate immunoreactivity, and the suppression of calpain activation using ALLN or calpain III, an inhibitor of calpain, blocked this decrease. In vitro, degraded fragments of G-substrate were detected within 10 min after coincubation of G-substrate and calpain. Moreover, G-substrate knockout (G-substrate(-/-)) mice were more susceptible to NMDA injury than wild-type mice. ALLN did not have a neuroprotective effect in G-substrate(-/-) mice. These data strongly suggest that calpain-mediated loss of G-substrate represents an important mechanism contributing to NMDA-induced amacrine cell death. Topics: Amacrine Cells; Analysis of Variance; Animals; Blotting, Western; Calpain; Cell Death; Cysteine Proteinase Inhibitors; Dipeptides; Immunohistochemistry; In Situ Nick-End Labeling; Leupeptins; Mice; Mice, Inbred C57BL; Mice, Knockout; N-Methylaspartate; Nerve Tissue Proteins; Proto-Oncogene Proteins c-akt; Rats; Rats, Sprague-Dawley; Retina | 2009 |
Reduced amyloidogenic processing of the amyloid beta-protein precursor by the small-molecule Differentiation Inducing Factor-1.
The detection of cell cycle proteins in Alzheimer's disease (AD) brains may represent an early event leading to neurodegeneration. To identify cell cycle modifiers with anti-Abeta properties, we assessed the effect of Differentiation-Inducing Factor-1 (DIF-1), a unique, small-molecule from Dictyostelium discoideum, on the proteolysis of the amyloid beta-protein precursor (APP) in a variety of different cell types. We show that DIF-1 slows cell cycle progression through G0/G1 that correlates with a reduction in cyclin D1 protein levels. Western blot analysis of DIF-treated cells and conditioned medium revealed decreases in the levels of secreted APP, mature APP, and C-terminal fragments. Assessment of conditioned media by sandwich ELISA showed reduced levels of Abeta40 and Abeta42, also demonstrating that treatment with DIF-1 effectively decreases the ratio of Abeta42 to Abeta40. In addition, DIF-1 significantly diminished APP phosphorylation at residue T668. Interestingly, site-directed mutagenesis of APP residue Thr668 to alanine or glutamic acid abolished the effect of DIF-1 on APP proteolysis and restored secreted levels of Abeta. Finally, DIF-1 prevented the accumulation of APP C-terminal fragments induced by the proteasome inhibitor lactacystin, and calpain inhibitor N-acetyl-leucyl-leucyl-norleucinal (ALLN). Our findings suggest that DIF-1 affects G0/G1-associated amyloidogenic processing of APP by a gamma-secretase-, proteasome- and calpain-insensitive pathway, and that this effect requires the presence of residue Thr668. Topics: Acetylcysteine; Amyloid beta-Peptides; Amyloid beta-Protein Precursor; Animals; Benzazepines; Cell Line; Cell Line, Tumor; CHO Cells; Cricetinae; Cricetulus; Cyclin D1; Fibroblasts; Glioma; Hexanones; Humans; Hydrocarbons, Chlorinated; Indoles; Leupeptins; Mice; Peptide Fragments; Proteasome Inhibitors; Purines; Recombinant Fusion Proteins; Roscovitine; Threonine | 2009 |
Inflammatory NF-kappaB activation promotes hepatic apolipoprotein B100 secretion: evidence for a link between hepatic inflammation and lipoprotein production.
Insulin-resistant states are commonly associated with chronic inflammation and hepatic overproduction of apolipoprotein B100 (apoB100), leading to hypertriglyceridemia and a metabolic dyslipidemic profile. Molecular mechanisms linking hepatic inflammatory cascades and the pathways of apoB100-lipoprotein production are, however, unknown. In the present study, we employed a diet-induced, insulin-resistant hamster model, as well as cell culture studies, to investigate the potential link between activation of hepatic inflammatory nuclear factor-kappaB (NF-kappaB) signaling cascade and the synthesis and secretion of apoB100-containing lipoproteins. Using an established insulin-resistant animal model, the fructose-fed hamster, we found that feeding fructose (previously shown to induce hepatic inflammation) for as little as 4 days reduced hepatic IkappaB (inhibitor of NF-kappaB) level, indicating activation of the inflammatory NF-kappaB cascade. Importantly, IKK (IkappaB kinase) inhibition was found to suppress apoB100 overproduction in fructose-fed hamster hepatocytes. As IKK, the upstream activator of NF-kappaB has been shown to inhibit insulin signaling, and insulin is a major regulator of apoB100, we modulated IKK activity in primary hamster hepatocytes and HepG2 cells and assessed the effects on hepatic apoB100 biosynthesis. Inhibition of the IKK-NF-kappaB pathway by BMS345541 and activation of the pathway by adenoviral-mediated IKK overexpression decreased and increased newly synthesized apoB100 levels, respectively. Pulse-chase and metabolic labeling experiments revealed that IKK activation regulates apoB100 levels at the levels of apoB100 biosynthesis and protein stability. Inhibition of the IKK-NF-kappaB pathway significantly enhanced proteasomal degradation of hepatic apoB100, while direct IKK activation led to reduced degradation and increased apoB100 mRNA translation. Together, our results reveal important links between modulation of the inflammatory IKK-NF-kappaB signaling cascade and hepatic synthesis and secretion of apoB100-containing lipoproteins. Hepatic inflammation may be an important underlying factor in hepatic apoB100 overproduction observed in insulin resistance. Topics: Administration, Oral; Animals; Apolipoprotein B-100; Cell Line, Tumor; Cell-Free System; Cricetinae; Cysteine Proteinase Inhibitors; Extracellular Signal-Regulated MAP Kinases; Fructose; Gene Expression; Hepatocytes; Humans; I-kappa B Kinase; I-kappa B Proteins; Imidazoles; Inflammation; Leupeptins; Lipid Metabolism; Lipoproteins; Lipoproteins, VLDL; Liver; Male; Mesocricetus; NF-kappa B; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Biosynthesis; Protein Kinase Inhibitors; Quinoxalines; Transfection; Tumor Necrosis Factor-alpha | 2009 |
Glucosamine-induced endoplasmic reticulum stress attenuates apolipoprotein B100 synthesis via PERK signaling.
Glucosamine impairs hepatic apolipoprotein B100 (apoB100) production by inducing endoplasmic reticulum (ER) stress and enhancing cotranslational and posttranslational apoB100 degradation (Qiu, W., R. K. Avramoglu, A. C. Rutledge, J. Tsai, and K. Adeli. Mechanisms of glucosamine-induced suppression of the hepatic assembly and secretion of apolipoprotein B-100-containing lipoproteins. J. Lipid Res. 2006. 47: 1749-1761). Here, we report that glucosamine also regulates apoB100 protein synthesis via ER-stress-induced PERK activation. Short-term (4 h) glucosamine treatment of HepG2 cells reduced both cellular (by 62%) and secreted apoB100 (by 43%) without altering apoB100 mRNA. Treatment with proteasomal inhibitors only partially prevented the suppressive effects of glucosamine, suggesting that mechanisms other than proteasomal degradation may also be involved. Glucosamine-induced ER stress was associated with a significantly reduced apoB100 synthesis with no significant change in posttranslational decay rates, suggesting that glucosamine exerted its effect early during apoB biosynthesis. The role of PERK and its substrate, alpha-subunit of eukaryotic initiation factor 2 (eIF2alpha), in the suppressive effects of glucosamine on apoB synthesis was then investigated. Coexpression of apoB15 (normally resistant to intracellular degradation) with wild-type double stranded (ds) RNA activated protein kinase (PKR)-like endoplasmic reticulum kinase (PERK) in COS-7 cells resulted in a dramatic reduction in the levels of newly synthesized apoB15. Interestingly, cotransfection with apoB15 and a kinase inactive PERK mutant (K618A) increased apoB15 expression. In addition, short-term glucosamine treatment stimulated an increase in phosphorylation of PERK and eIF2alpha. Taken together, these data suggest that in addition to the induction of ER-associated degradation and other degradative pathways, ER stress is associated with suppression of apoB synthesis via a PERK-dependent mechanism. Topics: Acetylcysteine; Activating Transcription Factor 6; Animals; Apolipoprotein B-100; Cell Line; eIF-2 Kinase; Endoplasmic Reticulum; Endoplasmic Reticulum Chaperone BiP; Endoribonucleases; Eukaryotic Initiation Factor-2; Gene Expression Regulation; Glucosamine; Heat-Shock Proteins; Humans; Leupeptins; Membrane Proteins; Phosphorylation; Protein Serine-Threonine Kinases; RNA; Signal Transduction; Time Factors | 2009 |
Effects of dopamine on LC3-II activation as a marker of autophagy in a neuroblastoma cell model.
Dopamine at 100-500 microM has toxic effects on human SH-SY5Y neuroblastoma cells, manifested as apoptotic cell loss and strong autophagy. The molecular mechanisms and types of dopamine-induced cell death are not yet well known. Their identification is important in the study of neurodegenerative diseases that specifically involve dopaminergic neurons. We looked for changes in expression and content of proteins involved in apoptosis and autophagy after dopamine treatment. All the changes found were prevented by avoiding dopamine oxidation with N-acetylcysteine, indicating a key role for the products of dopamine oxidation in dopamine toxicity. As early as 1-2h after treatment we found an increase in hypoxia-inducible factor-1alpha (HIF-1alpha) and an accumulation of ubiquitinated proteins. Proteins regulated by HIF-1alpha and involved in apoptosis and/or autophagy, such as p53, Puma and Bnip3, were subsequently increased. However, apoptotic parameters (caspase-3, caspase-7, PARP) were only activated after 12h of 500muM dopamine treatment. Autophagy, monitored by the LC3-II increase after LC3-I linkage to autophagic vacuoles, was evident after 6h of treatment with both 100 and 500 microM dopamine. The mTOR pathway was inhibited by dopamine, probably due to the intracellular redox changes and energy depletion leading to AMPK activation. However, this mechanism is not sufficient to explain the high LC3-II activation caused by dopamine: the LC3-II increase was not reversed by IGF-1, which prevented this effect when caused by the mTOR inhibitor rapamycin. Our results suggest that the aggregation of ubiquitinated non-degraded proteins may be the main cause of LC3-II activation and autophagy. As we have reported previously, cytosolic dopamine may cause damage by autophagy in neuroblastoma cells (and presumably in dopaminergic neurons), which develops to apoptosis and leads to cell degeneration. Topics: Analysis of Variance; Apoptosis Regulatory Proteins; Autophagy; Caspase 3; Cell Line, Tumor; Cell Survival; Cysteine Proteinase Inhibitors; Dopamine; Dopamine Agents; Dose-Response Relationship, Drug; Fluoresceins; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Leupeptins; Microscopy, Electron, Transmission; Microtubule-Associated Proteins; Nerve Tissue Proteins; Neuroblastoma; Oncogene Protein v-akt; Oxidative Stress; Phosphatidylinositol 3-Kinases; Protein Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-bcl-2; Signal Transduction; TOR Serine-Threonine Kinases | 2009 |
Decreased protein synthesis of Hsp27 associated with cellular toxicity in a cell model of Machado-Joseph disease.
Machado-Joseph disease is an autosomal dominant spinocerebellar degeneration caused by the expansion of a polyglutamine tract within the gene product, ataxin-3. We have previously shown that increased oxidative stress and decreased expression of Hsp27 may be contributory factors to the disease progression. In this study, we utilized neuroblastoma SK-N-SH cells stably transfected with full-length expanded ataxin-3 to further investigate the mechanism(s) resulting in the decreased expression of Hsp27. Results from 35S-methionine pulse-chase labeling and protein degradation assays revealed that decreased Hsp27 in mutant MJD cells is due to defects in protein synthesis. Our results further demonstrated that Hsp27 degradation is independent of the proteasome degradation pathway. In addition, we showed that overexpression of Hsp27 desensitizes mutant MJD cells to apoptotic stress. Taken together, these findings provide the first evidence that expanded ataxin-3 interferes with Hsp27 synthesis, which may contribute to the impairment of the cells' ability to respond to stresses and trigger the progression of this late-onset disease. Topics: Animals; Ataxin-3; Blotting, Western; Cell Line, Tumor; Chlorocebus aethiops; COS Cells; Cysteine Proteinase Inhibitors; Gene Expression; Heat-Shock Proteins; HSP27 Heat-Shock Proteins; Humans; Leupeptins; Machado-Joseph Disease; Molecular Chaperones; Nerve Tissue Proteins; Neurons; Nuclear Proteins; Peptides; Repressor Proteins; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Stress, Physiological; Transfection | 2009 |
Cytosolic Aryl sulfotransferase 4A1 interacts with the peptidyl prolyl cis-trans isomerase Pin1.
Sulfonation by cytosolic sulfotransferases plays an important role in the metabolism of both endogenous and exogenous compounds. Sulfotransferase 4A1 (SULT4A1) is a novel sulfotransferase found primarily in neurons in the brain. It is highly conserved between species, but no substantial enzyme activity has been identified for the protein. Consequently, little is known about the role of this enzyme in the brain. We performed a yeast two-hybrid screen of a human brain library to isolate potential SULT4A1-interacting proteins that might identify the role or regulation of the sulfotransferase in humans. The screen isolated the peptidyl-prolyl cis-trans isomerase Pin1. Its interaction with SULT4A1 was confirmed by coimmunoprecipitation studies in HeLa cells and by in vitro pull-down of expressed proteins. Moreover, Pin1 binding was dependent on phosphorylation of the SULT4A1 protein. Pin1 destabilized SULT4A1, decreasing its half-life from more than 8 h to approximately 4.5 h. This effect was dependent on the isomerase activity of Pin1 and was inhibited by okadaic acid, suggesting a role for the phosphatase PP2A. Pin1-mediated SULT4A1 degradation did not involve the proteosomes or macroautophagy, but it was inhibited by the calpain antagonists N-acetyl-Leu-Leu-Nle-CHO and Z-Val-Phe-CHO. Finally, Pin1 binding was mapped to two threonine-proline motifs (Thr(8) and Thr(11)) that are not present in any of the other human cytosolic sulfotransferases. Our findings suggest that SULT4A1 is subject to post-translational modification that alters its stability in the cell. These modifications may also be important for enzyme activity, which explains why specific substrates for SULT4A1 have not yet been identified. Topics: Amino Acid Motifs; Amino Acid Sequence; Binding Sites; Calpain; Cysteine Proteinase Inhibitors; Cytosol; Escherichia coli; Escherichia coli Proteins; Glutathione Transferase; Half-Life; HeLa Cells; Humans; Leupeptins; Molecular Sequence Data; NIMA-Interacting Peptidylprolyl Isomerase; Okadaic Acid; Peptidylprolyl Isomerase; Phosphorylation; Proline; Protein Binding; Recombinant Proteins; Serine; Sulfotransferases; Threonine; Transfection; Two-Hybrid System Techniques | 2009 |
Antimalarial effect of N-acetyl-L-Leucyl-L-leucyl-L-norleucinal by the inhibition of Plasmodium falciparum Calpain.
The biological understanding of malaria parasites has increased considerably over the past two decades with the discovery of many potential targets for the development of new antimalarial drugs. Calpain, a cysteine protease of Plasmodium falciparum, is believed to be a central mediator essential for parasitic activity. However, the utility of calpain as a potential anti-malarial target in P. falciparum has not been fully determined. In the present study, we determined the effect of N-acetyl-L-Leucyl-L-leucyl-L-norleucinal (ALLN)-treatment on the expression of calpain in erythrocytic stages of P. falciparum and its usefulness as an antimalarial chemotherapeutic agent. ALLN was shown to have low toxicity to HeLa cells but high toxicity to malaria. ALLN inhibited the expression of calpain in ring, trophozoite and schizont stages when treated for 48 h. Also, after 48 h, samples were characterized by 6.15% and 0% parasitemia without ALLN treatment and with ALLN treatment, respectively. Brightfield and confocal microscopy revealed that ALLN treatment affects merozoite maturation. As ALLN concentration increased from 1 muM to 100 microM, ring stage parasites did not mature into the schizont stage. When ALLN treatment was continued for 48 h, it also significantly inhibited the maturation of ring-stage parasites into trophozoite or schizont stages and survival of malarial parasites. Taken together, these findings suggest that ALLN inhibit the maturation and survival of P. falciparum and calpain expression, and thus has potential utility as an antimalarial chemotherapeutic agent. Topics: Animals; Antimalarials; Calpain; Drug Evaluation, Preclinical; Erythrocytes; HeLa Cells; Humans; Leupeptins; Parasitemia; Plasmodium falciparum; Protozoan Proteins; Schizonts; Trophozoites | 2009 |
Inhibition of calpain increases LIS1 expression and partially rescues in vivo phenotypes in a mouse model of lissencephaly.
Lissencephaly is a devastating neurological disorder caused by defective neuronal migration. LIS1 (official symbol PAFAH1B1, for platelet-activating factor acetylhydrolase, isoform 1b, subunit 1) was identified as the gene mutated in individuals with lissencephaly, and it was found to regulate cytoplasmic dynein function and localization. Here we show that inhibition or knockdown of calpains protects LIS1 from proteolysis, resulting in the augmentation of LIS1 amounts in Lis1(+/-) mouse embryonic fibroblast cells and rescue of the aberrant distribution of cytoplasmic dynein, mitochondria and beta-COP-positive vesicles. We also show that calpain inhibitors improve neuronal migration of Lis1(+/-) cerebellar granular neurons. Intraperitoneal injection of the calpain inhibitor ALLN to pregnant Lis1(+/-) dams rescued apoptotic neuronal cell death and neuronal migration defects in Lis1(+/-) offspring. Furthermore, in utero knockdown of calpain by short hairpin RNA rescued defective cortical layering in Lis1(+/-) mice. Thus, calpain inhibition is a potential therapeutic intervention for lissencephaly. Topics: 1-Alkyl-2-acetylglycerophosphocholine Esterase; Animals; Calpain; Cell Movement; Cells, Cultured; Cerebral Cortex; Cysteine Proteinase Inhibitors; Disease Models, Animal; Dyneins; Embryo, Mammalian; Female; Fibroblasts; Gene Expression Regulation, Developmental; Leucine; Leupeptins; Lissencephaly; Mice; Mice, Knockout; Microtubule-Associated Proteins; Models, Neurological; Neurons; Phenotype; Pregnancy | 2009 |
Role of Plasmodium falciparum digestive vacuole plasmepsins in the specificity and antimalarial mode of action of cysteine and aspartic protease inhibitors.
Hemoglobin (Hb) degradation is essential for the growth of the intraerythrocytic stages of malarial parasites. This process, which occurs inside an acidic digestive vacuole (DV), is thought to involve the action of four aspartic proteases, termed plasmepsins (PMs). These enzymes have received considerable attention as potential antimalarial drug targets. Leveraging the availability of a set of PM-knockout lines generated in Plasmodium falciparum, we report here that a wide range of previously characterized or novel aspartic protease inhibitors exert their antimalarial activities independently of their effect on the DV PMs. We also assayed compounds previously shown to inhibit cysteine proteases residing in the DV. The most striking observation was a ninefold increase in the potency of the calpain inhibitor N-acetyl-leucinyl-leucinyl-norleucinal (ALLN) against parasites lacking all four DV PMs. Genetic ablation of PM III or PM IV also decreased the level of parasite resistance to the beta-hematin binding antimalarial chloroquine. On the basis of the findings of drug susceptibility and isobologram assays, as well as the findings of studies of the inhibition of Hb degradation, morphological analyses, and stage specificity, we conclude that the DV PMs and falcipain cysteine proteases act cooperatively in Hb hydrolysis. We also identify several aspartic protease inhibitors, designed to target DV PMs, which appear to act on alternative targets early in the intraerythrocytic life cycle. These include the potent diphenylurea compound GB-III-32, which was found to be fourfold less potent against a P. falciparum line overexpressing plasmepsin X than against the parental nontransformed parasite line. The identification of the mode of action of these inhibitors will be important for future antimalarial drug discovery efforts focusing on aspartic proteases. Topics: Animals; Antimalarials; Aspartic Acid Endopeptidases; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Hemoglobins; Hydrolysis; Leupeptins; Malaria, Falciparum; Parasitic Sensitivity Tests; Plasmodium falciparum; Protease Inhibitors | 2009 |
Hypoxic resistance to articular chondrocyte apoptosis--a possible mechanism of maintaining homeostasis of normal articular cartilage.
Hypoxia and hypoxia-related genes are important factors in articular chondrocytes during cartilage homeostasis and osteoarthritis. We have investigated the various apoptotic factors that show significance in synovial fluid obtained from normal and experimental osteoarthritic animal models and have evaluated the effect of hypoxia on articular chondrocyte apoptosis induced by these apoptotic factors. Mature beagle dogs underwent surgical transections of ligaments and medial meniscectomies to explore the underlying mechanisms of osteoarthritis. Cartilage and synovial fluid obtained from normal animals and those with osteoarthritis were evaluated via proteasome inhibition, tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) protein expression, mitochondrial transmembrane potential and levels of reactive oxygen species. Canine chondrocytes were exposed to the proteasome inhibitor N-acetyl-Leu-Leu-Norleu-al and treated with recombinant TRAIL protein under normoxic and hypoxic conditions, measuring chondrocyte cell viability, proteasome activity and levels of apoptotic factors. TRAIL protein expression and ubiquitinated proteins were increased significantly, but the proteasome activity in the synovial fluid of osteoarthritic joints relative to that in normal joints was not. Primary cultured articular chondrocytes cotreated with the proteasome inhibitor and TRAIL progressed to severe apoptosis under normoxic conditions, but the sensitization caused by the combined treatment was suppressed by exposure to hypoxia. Caspase-8 activation, c-Jun N-terminal kinase phosphorylation, the mitochondrial transmembrane potential and the generation of reactive oxygen species involved in cell death regulation were significantly inhibited under hypoxic conditions. These findings suggest that proteasome inhibition and TRAIL may be possible mechanisms in cartilage degradation and joint-related diseases. Furthermore, the maintenance of hypoxic conditions or therapy with hypoxia-related genes in the joint may be successful for the treatment of joint-related diseases, including osteoarthritis. Topics: Animals; Apoptosis; Cartilage, Articular; Caspase 8; Cells, Cultured; Chondrocytes; Dogs; Female; Homeostasis; Hypoxia; JNK Mitogen-Activated Protein Kinases; Leupeptins; Membrane Potential, Mitochondrial; Osteoarthritis; Proteasome Inhibitors; Reactive Oxygen Species; Synovial Fluid; TNF-Related Apoptosis-Inducing Ligand; Ubiquitinated Proteins | 2009 |
The C-terminal region of serotonin transporter is important for its trafficking and glycosylation.
We investigated the effects of brefeldin A and ilimaquinone, inhibitors of membrane trafficking, using serotonin transporter (SERT)-expressing COS-7 cells. Both drugs significantly inhibited the serotonin uptake activity of SERT and caused SERT to be retained in the endoplasmic reticulum (ER), indicating that membrane trafficking is an important factor for SERT functional regulation. In agreement with previous reports, a C-terminal-deletion mutant of SERT (SERTDeltaCT) mostly localized to the ER and completely lacked serotonin uptake activity. To further elucidate the role of the C-terminus of SERT, we investigated whether overexpression of FLAG-tagged SERT C-terminus (FLAG-SERT-CT) affected the serotonin uptake activity and glycosylation of SERT. Interestingly, when concomitantly expressed with full-length FLAG-SERT in COS-7 cells, FLAG-SERT-CT increased the serotonin uptake activity and mature glycosylation of FLAG-SERT. These results indicate that the C-terminal region of SERT plays a crucial role in the functional regulation of SERT via membrane trafficking and glycosylation. In addition, proteasome inhibitors induced apparent ER stress, significantly decreased the serotonin uptake activity and mature glycosylation of SERT and caused SERT to be localized to the ER, suggesting that SERT function would be attenuated via membrane trafficking in pathological states that trigger ER stress. Topics: Acetylcysteine; Animals; Binding Sites; Brefeldin A; Chlorocebus aethiops; COS Cells; Cysteine Proteinase Inhibitors; Endoplasmic Reticulum; Glycosylation; Leupeptins; Membrane Transport Modulators; Protein Structure, Tertiary; Protein Transport; Quinones; Sequence Deletion; Serotonin; Serotonin Plasma Membrane Transport Proteins; Sesquiterpenes; Structure-Activity Relationship; Transfection | 2009 |
Mycobacterium bovis bacillus Calmette-Guerin induces CCL5 secretion via the Toll-like receptor 2-NF-kappaB and -Jun N-terminal kinase signaling pathways.
In response to Mycobacterium bovis bacillus Calmette-Guérin (BCG), CC chemokines are secreted from host cells to attract components of the innate and adaptive immune systems to the site of infection. Toll-like receptor 2 (TLR2) has been shown to recognize M. bovis BCG and to initiate signaling pathways that result in enhanced secretion of CC chemokines. Despite the essential requirement of TLR2 in M. bovis BCG infection, the mechanisms by which it induces secretion of CC chemokines are not well defined. In this study, we report that stimulation of HEK293 cells expressing human TLR2 with M. bovis BCG resulted in increased CCL2 and CCL5 secretion, as determined by an enzyme-linked immunosorbent assay. M. bovis BCG infection resulted in the activation of c-Jun N-terminal kinase (JNK), and the inhibition of JNK activity had a significant effect on M. bovis BCG-dependent CCL5 secretion in TLR2-expressing cells but no effect on M. bovis BCG-dependent CCL2 secretion from infected HEK293 cells expressing human TLR2. The M. bovis BCG-induced CCL5 release was attenuated by sulfasalazine (a well-described inhibitor of NF-kappaB activity), BAY 11-7082 (an IkappaB phosphorylation inhibitor), and ALLN (a well-described inhibitor of NF-kappaB activation that prevents degradation of IkappaB and eventually results in a lack of translocated NF-kappaB in the nucleus). In addition, stimulation of TLR2-expressing cells with M. bovis BCG resulted in translocation of NF-kappaB subunits from the cytoplasmic to the nuclear fraction, and stimulation of cells with M. bovis BCG activated IkappaB kinase alphabeta. These findings indicate that M. bovis BCG induces CCL5 production through mechanisms that include a TLR2-dependent component that requires JNK and NF-kappaB activities. Topics: Cell Line; Cell Nucleus; Chemokine CCL2; Chemokine CCL5; Cytoplasm; Enzyme Inhibitors; Enzyme-Linked Immunosorbent Assay; Humans; JNK Mitogen-Activated Protein Kinases; Leupeptins; Mycobacterium bovis; NF-kappa B; Nitriles; Protein Transport; Sulfasalazine; Sulfones; Toll-Like Receptor 2 | 2008 |
Antioxidants block proteasome inhibitor function in endometrial carcinoma cells.
We have recently demonstrated that proteasome inhibitors can be effective in inducing apoptotic cell death in endometrial carcinoma cell lines and primary culture explants. Increasing evidence suggests that reactive oxygen species are responsible for proteasome inhibitor-induced cell killing. Antioxidants can thus block apoptosis (cell death) triggered by proteasome inhibition. Here, we have evaluated the effects of different antioxidants (edaravone and tiron) on endometrial carcinoma cells treated with aldehyde proteasome inhibitors (MG-132 or ALLN), the boronic acid-based proteasome inhibitor (bortezomib) and the epoxyketone, epoxomicin. We show that tiron specifically inhibited the cytotoxic effects of bortezomib, whereas edaravone inhibited cell death caused by aldehyde-based proteasome inhibitors. We have, however, found that edaravone completely inhibited accumulation of ubiquitin and proteasome activity decrease caused by MG-132 or ALLN, but not by bortezomib. Conversely, tiron inhibited the ubiquitin accumulation and proteasome activity decrease caused by bortezomib. These results suggest that edaravone and tiron rescue cells of proteasome inhibitors from cell death, by inhibiting blockade of proteasome caused by MG-132 and ALLN or bortezomib, respectively. We also tested other antioxidants, and we found that vitamin C inhibited bortezomib-induced cell death. Similar to tiron, vitamin C inhibited cell death by blocking the ability of bortezomib to inhibit the proteasome. Until now, all the antioxidants that blocked proteasome inhibitor-induced cell death also blocked the proteasome inhibitor mechanism of action. Topics: Antioxidants; Antipyrine; Apoptosis; Ascorbic Acid; Blotting, Western; Boronic Acids; Bortezomib; Butylated Hydroxyanisole; Caspase 3; Caspase 9; Caspase Inhibitors; Cell Line, Tumor; Cell Survival; Coumarins; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Edaravone; Endometrial Neoplasms; Ergothioneine; Female; Humans; Leupeptins; Oligopeptides; Proteasome Inhibitors; Pyrazines; Ubiquitin; Vitamin E; Vitamins | 2008 |
Down-regulation of brain-pancreas relative protein in diabetic rats and by high glucose in PC12 cells: prevention by calpain inhibitors.
Brain-pancreas relative protein (BPRP) is a novel protein that we found in our laboratory. Previously we demonstrated that it is involved in ischemia and depression. In light of the putative association between diabetes and clinical depression, and the selective expression of BPRP in brain and pancreas, the present study examined whether BPRP levels are affected by induction of diabetes by alloxan injection in rats and exposure to high glucose levels in PC12 cells. Western blot and immunohistochemical analyses revealed that BPRP levels were decreased in the hippocampal CA1 neurons of diabetic rats 4 and 8 weeks post-alloxan injection and in PC12 cells 48 h after exposure to high concentrations of glucose. BPRP protein levels were not affected by osmolarity control treatments with mannitol. Follow-up pharmacological experiments in PC12 cells revealed that glucose-induced BPRP down-regulation was markedly attenuated by the calpain inhibitors N-acetyl-Leu-Leu-norleucinal (ALLN) or calpeptin, but not the proteasome-specific inhibitor carbobenzoxy-Leu-Leu-leucinal (MG132). The ability of calpain inhibitors to specifically counter the effects of high glucose exposure on BPRP levels further suggests that BPRP and calpain activity may contribute to diabetes complications in the central nervous system. Topics: Animals; Calpain; Cysteine Proteinase Inhibitors; Diabetes Mellitus, Experimental; Dipeptides; Down-Regulation; Glucose; Half-Life; Hippocampus; Insulin; Leupeptins; Male; Nerve Tissue Proteins; Neurons; PC12 Cells; Rats; Rats, Sprague-Dawley; Time Factors | 2008 |
Autophagy modulates keratin-containing inclusion formation and apoptosis in cell culture in a context-dependent fashion.
The major pathways for protein degradation are the proteasomal and lysosomal systems. Derangement of protein degradation causes the formation of intracellular inclusions, and apoptosis and is associated with several diseases. We utilized hepatocyte-derived cell lines to examine the consequences of the cytoplasmic hepatocyte Mallory-Denk body-like inclusions on organelle organization, autophagy and apoptosis, and tested the hypothesis that autophagy affects inclusion turnover. Proteasome inhibitors (PIs) generate keratin-containing Mallory-Denk body-like inclusions in cultured cells and cause reorganization of mitochondria and other organelles, autophagy and apoptosis. In cultured hepatoma cells, caspase inhibition blocks PI-induced apoptosis but not inclusion formation or autophagy activation. Autophagy induction by rapamycin decreases the extent of PI-induced inclusions and apoptosis in Huh7 and OUMS29 cells. Surprisingly, blocking of autophagy sequestration by 3 methyl adenine or beclin 1 siRNA, but not bafilomycin A1 inhibition of autophagic degradation, also inhibits inclusion formation in the tested cells. Therefore, autophagy can be upstream of apoptosis and may promote or alleviate inclusion formation in cell culture in a context-dependent manner via putative autophagy-associated molecular triggers. Manipulation of autophagy may offer a strategy to address the importance of inclusion formation and its significance in inclusion-associated diseases. Topics: Apoptosis; Autophagy; Cell Line; Cycloheximide; Cysteine Proteinase Inhibitors; Humans; Inclusion Bodies; Keratins; Leupeptins; Organelles; Proteasome Inhibitors; Protein Synthesis Inhibitors | 2008 |
UBE1L represses PML/RAR{alpha} by targeting the PML domain for ISG15ylation.
Acute promyelocytic leukemia (APL) is characterized by expression of promyelocytic leukemia (PML)/retinoic acid (RA) receptor alpha (RARalpha) protein and all-trans-RA-mediated clinical remissions. RA treatment can confer PML/RARalpha degradation, overcoming dominant-negative effects of this oncogenic protein. The present study uncovered independent retinoid degradation mechanisms, targeting different domains of PML/RARalpha. RA treatment is known to repress PML/RARalpha and augment ubiquitin-activating enzyme-E1-like (UBE1L) protein expression in NB4-S1 APL cells. We previously reported RA-induced UBE1L and the IFN-stimulated gene, 15-kDa protein ISG15ylation in APL cells. Whether the ubiquitin-like protein ISG15 directly conjugates with PML/RARalpha was not explored previously and is examined in this study. Transient transfection experiments with different PML/RARalpha domains revealed that RA treatment preferentially down-regulated the RARalpha domain, whereas UBE1L targeted the PML domain for repression. As expected, ubiquitin-specific protease 18 (UBP43/USP18), the ISG15 deconjugase, opposed UBE1L but not RA-dependent PML/RARalpha degradation. In contrast, the proteasomal inhibitor, N-acetyl-leucinyl-leucinyl-norleucinal, inhibited both UBE1L- and RA-mediated PML/RARalpha degradation. Notably, UBE1L induced ISG15ylation of the PML domain of PML/RARalpha, causing its repression. These findings confirmed that RA triggers PML/RARalpha degradation through different domains and distinct mechanisms. Taken together, these findings advance prior work by establishing two pathways converge on the same oncogenic protein to cause its degradation and thereby promote antineoplastic effects. The molecular pharmacologic implications of these findings are discussed. Topics: Animals; Antineoplastic Agents; Bronchi; Cells, Cultured; Chlorocebus aethiops; COS Cells; Cytokines; Endopeptidases; Gene Expression Regulation, Leukemic; Humans; Immunoblotting; Immunoprecipitation; Leukemia, Promyelocytic, Acute; Leupeptins; Oncogene Proteins, Fusion; Protein Processing, Post-Translational; Protein Structure, Tertiary; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Signal Transduction; Transfection; Tretinoin; Ubiquitin Thiolesterase; Ubiquitin-Activating Enzymes; Ubiquitins | 2008 |
Oxidative stress and calpain inhibition induce alpha B-crystallin phosphorylation via p38-MAPK and calcium signalling pathways in H9c2 cells.
We investigated the response of alphaB-crystallin to oxidative stress and calpain inhibition in an attempt to elucidate the signalling pathways mediating its phosphorylation. Given the high expression levels of alphaB-crystallin in cardiac muscle one can evaluate the significance of its participation in preservation of homeostasis under adverse conditions. H9c2 cardiac myoblasts were used as our experimental model since their response reflects the signal transduction pathways activated by stress conditions in the myocardium. Thus, in H9c2 cells treated with H2O2 the mechanism regulating alphaB-crystallin phosphorylation was found to involve p38-MAPK/MSK1 as well as intracellular free calcium levels. Our immunocytochemical experiments demonstrated phosphorylated alphaB-crystallin to be co-localized with tubulin, potentially preserving cytoskeletal architecture under these interventions. In H9c2 cells treated with calpain inhibitors (ALLN, ALLM) alphaB-crystallin exhibited a p38-MAPK- and [Ca 2+](i)-dependent phosphorylation pattern since the latter was ablated in the presence of the selective p38-MAPK inhibitor SB203580 and calcium chelator BAPTA-AM. Calpain activity repression ultimately led to apoptosis confirmed by PARP fragmentation and chromatin condensation. However, the apoptotic pathway activated by ALLM and ALLN differed, underlying the diverse transduction mechanisms stimulated. In addition to this, an anti-apoptotic role for phospho-alphaB-crystallin was verified by confirmation of its interaction with pro-caspase 3, hindering its cleavage and subsequent activation. Collectively, our findings underline alphaB-crystallin crucial role as a participant of cardiac cells early response to stressful stimuli compromising their survival. Topics: alpha-Crystallin B Chain; Animals; Apoptosis; Calcium Signaling; Calpain; Caspase 3; Cell Line; Cell Nucleus; Cytochromes c; Egtazic Acid; Hydrogen Peroxide; Leupeptins; Myocytes, Cardiac; Oligopeptides; Oxidative Stress; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Phosphoserine; Poly(ADP-ribose) Polymerases; Protein Transport; Rats; Ribosomal Protein S6 Kinases, 90-kDa; Time Factors; Tubulin | 2008 |
Complex effects of IL1A polymorphism and calpain inhibitors on interleukin 1 alpha (IL-1 alpha) mRNA levels and secretion of IL-1 alpha protein.
Alleles of IL1A-889(C>T) and IL1A+4845(G>T) are in linkage disequilibrium. Interleukin 1alpha (IL-1alpha) is produced as a precursor protein and cleaved at positions 117-118 by calpain, generating a mature protein for export. IL1A+4845 affects amino acids expressed at position 114 and hence may modulate calpain-mediated cleavage. We sought evidence for this mechanism in intact cells. Blood leukocytes from heterozygous donors released more IL-1alpha protein than cells from IL1A(1,1) donors, while release from IL1A(2,2) cells was variable. Genotype did not affect levels of IL-1alpha mRNA, so differential cleavage of the precursor is a feasible mechanism. However, genotype also had no effect on inhibition of IL-1alpha release by pretreatment with calpain inhibitors, and calpain inhibitors reduced IL-1alpha and tumor necrosis factor alpha mRNA levels. Hence, calpain inhibitors probably affect inhibition of signal transduction pathway rather than cleavage of IL-1alpha protein. As ratios of mu-calpain/calpastatin were lowest in heterozygous donors, genetically determined IL-1alpha levels may modulate transcription of calpain and calpastatin. This could reduce the impact of IL1A genotype on IL-1alpha secretion and amplify individual variation in levels generated in culture. Topics: Acrylates; Calpain; Cells, Cultured; Dipeptides; Glycoproteins; Humans; Interleukin-1alpha; Leupeptins; Polymorphism, Genetic; RNA, Messenger | 2008 |
Lithium ions increase hepatic glycogen synthase stability through a proteasome-related mechanism.
Incubation of rat hepatocytes with LiCl resulted in an overall increase in the activity ratio of glycogen synthase (GS), concomitantly with a decrease in active GS kinase-3 levels. GS total activity was also increased in a dose- and time-dependent manner. This latter effect correlated with the amount of immunoreactive enzyme determined by immunoblotting. Cycloheximide and actinomycin-D did not modify LiCl action on GS activity. Lithium ions did not induce any changes in GS mRNA levels. Furthermore, the increase in the total amount of GS induced by LiCl was further augmented after addition of a specific, calpain and proteasome inhibitor. Our results indicate that LiCl increases hepatocyte GS activity through increasing both the activation state of the enzyme and its cellular content. This latter increase is mediated through a modification of the proteasome-regulated proteolytic pathway of the enzyme. Topics: Animals; Calpain; Cations, Monovalent; Cells, Cultured; Cycloheximide; Cysteine Proteinase Inhibitors; Dactinomycin; Enzyme Activation; Glycogen Synthase; Glycogen Synthase Kinase 3; Hepatocytes; Leupeptins; Lithium Chloride; Liver Glycogen; Male; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Synthesis Inhibitors; Rats; Rats, Sprague-Dawley; RNA, Messenger | 2007 |
Involvement of NO/cGMP signaling in the apoptotic and anti-angiogenic effects of beta-lapachone on endothelial cells in vitro.
Neovascularization is an essential process in tumor development, it is conceivable that anti-angiogenic treatment may block tumor growth. In angiogenesis, nitric oxide (NO) is an important factor which mediates vascular endothelial cell growth and migration. beta-Lapachone (3,4-dihydro-2,2-dimethyl-2H-naphtho-[1,2-b]pyran-5,6-dione), a natural product extracted from the lapacho tree (Tabebuia avellanedae), has been demonstrated to possess anti-cancer and anti-viral effects. Whether beta-lapachone can induce endothelial cell death or has an anti-angiogenic effect is still an enigma. We investigated the in vitro effect of beta-lapachone on endothelial cells, including human vascular endothelial cell line, EAhy926, and human umbilical vascular endothelial cells (HUVEC). Our results revealed that (1) the intracellular cGMP levels and the mitochondria membrane potential (MMP) decreased, and calpain and caspases were activated, during beta-lapachone-induced endothelial cell death; (2) co-treatment with calpain inhibitors (ALLM or ALLN) or the intracellular calcium chelator, BAPTA, but not the general caspase inhibitor, zVAD-fmk, provided significant protection against apoptosis by preventing the beta-lapachone-induced MMP decrease and cytoplasmic calcium increase; (3) addition of NO downregulated the beta-lapachone-induced cGMP depletion and protected the cells from apoptosis by blocking the MMP decrease and the calcium increase; and (4) exogenous NO protects endothelial cells against the cell death induced by beta-lapachone, but not the anti-angiogenic effect. From all the data above, we demonstrated that NO can attenuate the apoptotic effect of beta-lapachone on human endothelial cells and suggest that beta-lapachone may have potential as an anti-angiogenic drug. Topics: Angiogenesis Inhibitors; Apoptosis; Arginine; Calcium; Calpain; Caspases; Cell Line; Cell Survival; Chelating Agents; Cyclic GMP; Dose-Response Relationship, Drug; Egtazic Acid; Endothelial Cells; Enzyme Activation; Enzyme Inhibitors; Humans; Leupeptins; Membrane Potential, Mitochondrial; Naphthoquinones; Neovascularization, Physiologic; NG-Nitroarginine Methyl Ester; Nitric Oxide; Nitric Oxide Donors; Nitric Oxide Synthase Type III; Oligopeptides; Signal Transduction; Time Factors | 2007 |
Ischemia promotes calpain-mediated degradation of p120-catenin in SH-SY5Y cells.
p120-catenin contributes to the cadherin-mediated adhesion and aggregation of cells. mu-Calpain was activated and p120-catenin was degraded after 36 h of ischemia in differentiated SH-SY5Y cells. Calpain inhibitors Cbz-Val-Phe-H (MDL28170, 20 microM) and N-acetyl-leucyl-leucyl-norleucinal (ALLN, 20 microM) increased the levels of dephosphorylated p120-catenin, aggregation, and cell survival as detected by reduced LDH release in ischemic cells. However, a proteasome inhibitor lactacystin had no such effects. This is the first report of the calpain-mediated degradation of p120-catenin and an association between the level of dephosphorylated p120-catenin and cell aggregation in ischemic neuronal cells. Topics: Acetylcysteine; Calpain; Catenins; Cell Adhesion Molecules; Cell Aggregation; Cell Death; Cell Line, Tumor; Delta Catenin; Dipeptides; Humans; Ischemia; Leupeptins; Neuroblastoma; Phosphoproteins | 2007 |
Sterol regulatory element binding proteins downregulate LDL receptor-related protein (LRP1) expression and LRP1-mediated aggregated LDL uptake by human macrophages.
In the extracellular intima, extracellular matrix proteoglycans favor LDL retention and aggregation (agLDL). In contrast to native LDL (nLDL), agLDL induces high intracellular cholesteryl ester (CE) accumulation in macrophages. It has been suggested that LDL receptor-related protein (LRP1) is involved in agLDL binding and internalization by macrophages. The aim of this work was to analyze whether sterol regulatory element binding proteins (SREBPs) modulate LRP1 expression and LRP1-mediated agLDL uptake by human monocyte-derived macrophages (HMDM).. The treatment of HMDM with small anti-LRP1 interfering RNA (siRNA-LRP1) led to the specific inhibition of LRP1 mRNA expression and also to the inhibition of LRP1 protein expression in these cells. In siRNA-LRP1-treated HMDM, CE accumulation from agLDL uptake (84.66+/-5 microg CE/mg protein) was reduced by 95.76+/-5.22%. This suggests that LRP1 plays a pivotal role in agLDL uptake by HMDM. N-acetyl-leucyl-leucyl-norleucinal (ALLN), an inhibitor of SREBP catabolism, maintained high levels of active SREBP-2 and SREBP-1 even in the presence of nLDL and agLDL. Therefore, ALLN induced LDL receptor (LDLR) upregulation. Concomitantly, a strong downregulation of LRP1 mRNA and LRP1 protein was observed in ALLN-treated macrophages. By decreasing LRP1 expression levels, ALLN reduced CE accumulation from agLDL at all tested concentrations.. These results suggest that high levels of active SREBPs downregulate LRP1 expression and intracellular CE accumulation in HMDM. Topics: Analysis of Variance; Atherosclerosis; Blotting, Western; Cells, Cultured; Cholesterol Esters; Coronary Vessels; Down-Regulation; Gene Expression Regulation; Humans; Leupeptins; Lipoproteins, LDL; Low Density Lipoprotein Receptor-Related Protein-1; Macrophages; Reverse Transcriptase Polymerase Chain Reaction; RNA Interference; RNA, Messenger; RNA, Small Interfering; Statistics, Nonparametric; Sterol Regulatory Element Binding Proteins | 2007 |
Oxidative stress induces the endoplasmic reticulum stress and facilitates inclusion formation in cultured cells.
The precise mechanism of formation and significance of Mallory bodies (MBs) are poorly understood. The endoplasmic reticulum (ER) is the organelle responsible for proper folding and elimination of unfolded proteins. Therefore, failure of this function increases defective proteins in the cell.. We examined the effects of oxidative stress on induction of ER stress and keratin 8 and 18 (K8/18)-containing inclusion formation in cultured human hepatoma cells and hepatocytes by immunofluorescence and immunoblot analyses.. Generation of H(2)O(2) was detected in glucose oxidase (GO)-treated cells by 2',7'-dichlorodihydrofluorescein diacetate and co-treatment with GO and acetyl-leucyl-leucyl-norleucinal (ALLN), a proteasome inhibitor, induced formation of extensive keratin inclusions that were inhibited by pre-treatment with N-acetyl-cysteine. These inclusions shared similar features with MBs by immunofluorescence analysis. Electron microscopy showed that these structures appeared near the nuclei, surrounded by filamentous structures. GO and ALLN upregulated the expression of ER stress markers, however, 4-phenylbutyrate, a chemical chaperone, reduced formation of inclusions and expression of the ER stress markers.. The oxidative stress coupled with limited inhibition of the proteasome induces dysfunction of the ER and results in inclusion formation in cultured cells. This suggests that ER stress plays a role in MB formation in liver disease. Topics: Cysteine Proteinase Inhibitors; Endoplasmic Reticulum; Fluoresceins; Glucose Oxidase; Humans; Hydrogen Peroxide; Inclusion Bodies; Keratin-18; Keratin-8; Leupeptins; Liver Diseases; Oxidative Stress; Phenylbutyrates; Proteasome Inhibitors; Tumor Cells, Cultured | 2007 |
Clostridium difficile toxin A-induced apoptosis is p53-independent but depends on glucosylation of Rho GTPases.
Clostridium difficile toxin A (TcdA) is one of two homologous glucosyltransferases that mono-glucosylate Rho GTPases. HT29 cells were challenged with wild-type and mutant TcdA to investigate the mechanism by which apoptosis is induced. The TcdA-induced re-organization of the actin cytoskeleton led to an increased number of cells within the G2/M phase. Depolymerization of the actin filaments with subsequent G2/M arrest, however, was not causative for apoptosis, as shown in a comparative study using latrunculin B. The activation of caspase-3, -8, and -9 strictly depended on the glucosylation of Rho GTPases. Apoptosis measured by flow cytometry was completely abolished by a pan-caspase inhibitor (z-VAD-fmk). Interestingly, cleavage of procaspase-3 and Bid was not inhibited by z-VAD-fmk, but was inhibited by the calpain/cathepsin inhibitor ALLM. Cleavage of procaspase-8 was susceptible to inhibition by z-VAD-fmk and to the caspase-3 inhibitor Ac-DMQD-CHO, indicating a contribution to the activation of caspase-3 in an amplifying manner. Although TcdA induced mitochondrial damage and cytochrome c release, p53 was not activated or up-regulated. A p53-independent apoptotic effect was also checked by treatment of HCT 116 p53(-/-) cells. In summary, TcdA-induced apoptosis in HT29 cells depends on glucosylation of Rho GTPases leading to activation of cathepsins and caspase-3. Topics: Actins; Apoptosis; Bacterial Toxins; Caspases; Cathepsins; Cell Cycle; Enterotoxins; Enzyme Activation; Glucosides; Glycosyltransferases; HT29 Cells; Humans; Leupeptins; Models, Biological; Protein Processing, Post-Translational; rho GTP-Binding Proteins; Tumor Suppressor Protein p53 | 2007 |
Cleavage of focal adhesion kinase (FAK) is essential in adipocyte differentiation.
During adipocyte differentiation, the cells experience dramatic alterations in morphology, motility and cell-ECM contact. Focal adhesion kinase (pp125FAK), a widely expressed non-receptor tyrosine kinase in integrin signaling, has been reported to participate in these events in various cells. Utilizing 3T3-L1 cells and primary rat preadipocytes, we explored the role of FAK in adipocyte differentiation. Gradual cleavage of FAK was demonstrated during adipcoyte differentiation, both in vitro and in vivo. This cleavage of FAK was mediated by calpain. Inhibition of calpain activity resulted in the rescue of FAK degradation, accompanied with the disturbance of final maturation of adipocyte. Our study revealed that FAK participated in adipocyte differentiation, and its cleavage by calpain was required to fulfill the final maturation of adipocytes. Topics: 3T3-L1 Cells; Adipocytes; Adipose Tissue; Animals; Blotting, Western; Calcimycin; Calpain; Cell Differentiation; Cell Nucleus; Cells, Cultured; Chromones; Cysteine Proteinase Inhibitors; Enzyme Inhibitors; Focal Adhesion Protein-Tyrosine Kinases; Ionophores; Leupeptins; Mice; Microscopy, Confocal; Morpholines; Peptide Fragments; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Rats; Signal Transduction; Time Factors | 2007 |
Regulation of plasma membrane Ca2+-ATPase in human platelets by calpain.
The plasma membrane Ca(2+)-ATPase (PMCA) plays an essential role in maintaining low cytosolic Ca(2+) in resting human platelets by extruding Ca(2+) from the cytoplasm across the plasma membrane. Since PMCA is the main agent of Ca(2+) efflux in platelets, it is a key point for regulation of platelet Ca(2+) metabolism. PMCA has been shown to be an excellent substrate for the Ca(2+)-activated cysteine protease calpain, a major platelet protein that is turned on during platelet activation. The objectives of the present work were to determine if PMCA is degraded during thrombin- and collagen-mediated platelet activation, and if calpain is responsible. The kinetics of PMCA degradation during platelet activation were analysed using SDS polyacrylamide gel electrophoresis and immunoblotting. The role of calpain was tested using the calpain inhibitors calpeptin and ALLN. Platelet activation mediated by both collagen and thrombin resulted in degradation of 60% of platelet PMCA within 18 minutes. Calpeptin and ALLN significantly inhibited the rate and extent of PMCA degradation. We conclude that calpain-mediated degradation of PMCA during platelet activation likely contributes significantly to Ca(2+) regulation and, therefore, to platelet function. Topics: Blood Platelets; Calcium; Calcium-Transporting ATPases; Calpain; Cell Membrane; Collagen; Dipeptides; Enzyme Activation; Humans; Leupeptins; Platelet Activation; Thrombin | 2007 |
Babesia bovis: effects of cysteine protease inhibitors on in vitro growth.
In the present study, we examined the effects of four kinds of cysteine protease inhibitors (E64, E64d, leupeptin, and ALLN) on the in vitro asexual growth of Babesia bovis. Of these, only the lipophilic inhibitors, E64d and ALLN, were found to effectively inhibit the growth of B. bovis. In further experiments, E64d, but not ALLN, significantly suppressed the parasite's invasion of host erythrocytes, while both chemicals, especially ALLN, inhibited the parasite's replication within the infected erythrocytes. These data suggested the presence of cysteine protease(s) derived from B. bovis, in which the protease(s) would play important roles in the erythrocyte invasion and/or replication processes of the parasite. Topics: Animals; Babesia bovis; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Erythrocytes; Leucine; Leupeptins | 2007 |
Endothelial gap junctions are down-regulated by arsenic trioxide.
We investigated the effect of As(2)O(3), an anti-cancer drug, on endothelial gap junctions. Human aortic endothelial cells (HAEC) were treated with As(2)O(3) at 1, 10, 100, and 1000 ng/ml and the cells were examined to evaluate the expression of connexin43 (Cx43) and to assess gap-junction communication. Endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) were measured to assess for endothelial dysfunction. Male Sprague-Dawley rats were given intravenous As(2)O(3) (200 mug/kg/day) or saline for 4 weeks, after which aortic endothelial gap junctions, eNOS, and circulating NO levels were evaluated. We found that HAEC Cx43 transcripts and gap junctions were reduced and gap-junction communication was attenuated by As(2)O(3). This decrease of Cx43 gap junctions was prevented by the addition of protease inhibitors. At a dose of 100 ng/ml of As(2)O(3), eNOS was reduced at 48 h, but NO was markedly reduced by 1 h. In animals treated with As(2)O(3), endothelial gap junctions comprising Cx37, Cx40, or Cx43 were all reduced in amount, while eNOS and circulating NO levels remained unchanged. In both in vitro and in vivo rat experiments, endothelial gap junctions were consistently reduced by As(2)O(3), unlike the response of eNOS and NO, which were decreased in cells but not in the rat aortic endothelium. The reduction in Cx43 involved both down-regulation at the transcriptional level and increased degradation. These findings indicate that gap-junction communication in the vascular endothelium is inhibited by treatment with As(2)O(3). Topics: Animals; Antineoplastic Agents; Arsenic Trioxide; Arsenicals; Blotting, Western; Cell Line; Connexin 43; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Down-Regulation; Endothelium, Vascular; Gap Junctions; Humans; Immunohistochemistry; Leupeptins; Male; Nitric Oxide; Nitric Oxide Synthase Type III; Oxides; Rats; Rats, Sprague-Dawley; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Time Factors; von Willebrand Factor | 2007 |
p21 (CDKN1A) is a negative regulator of p53 stability.
Cell cycle arrest in response to DNA damage involves protein stabilization and consequent upregulation of p53, which induces transcription of cyclin-dependent kinase inhibitor p21 (CDKN1A). We now show that p21 acts as a negative regulator of the cellular levels of p53. p21 knockdown by short hairpin RNA strongly increased p53 upregulation by a DNA-damaging drug doxorubicin in HT1080 fibrosarcoma cells. A protease inhibitor N-Ac-Leu-Leu-norleucinal (ALLN) drastically increased the amount of p53 in HCT116 colon carcinoma cells, but it had no effect on the already high p53 level in a p21(-/-) derivative of this cell line. Inhibition of transcription, which increases p53 levels in different cell lines due to the degradation of p53-destabilizing proteins such as Mdm2, failed to increase but instead decreased the amount of p53 in p21(-/-) cells, despite a drastic decrease in the level of Mdm2. These results indicate that p21 acts as a negative regulator of p53 stability in different cell types. p53 regulation by p21 may provide a negative regulatory loop that limits p53 induction. Topics: Cell Cycle; Cell Line, Tumor; Cyclin-Dependent Kinase Inhibitor p21; DNA Damage; Doxorubicin; Gene Expression Regulation; Humans; Immunoblotting; Leupeptins; Proto-Oncogene Proteins c-mdm2; Tumor Suppressor Protein p53 | 2007 |
Sp3 and sp4 transcription factor levels are increased in brains of patients with Alzheimer's disease.
Alzheimer's disease (AD) is characterized by extracellular Abeta peptide deposition originating from amyloid precursor protein cleavage and intracellular neurofibrillary tangles resulting from pathological tau protein aggregation. These processes are accompanied by dramatic neuronal losses, further leading to different cognitive impairments. Neuronal death signalings involve gene expression modifications that rely on transcription factor alterations. Herein, we investigated the fate of the Sp family of transcription factors in postmortem brains from patients with AD disease and in different contexts of neuronal death.. By immunohistochemistry we found that the Sp3 and Sp4 levels were dramatically increased and associated with neurofibrillary tangles and pathological tau presence in neurons from the CA1 region of the hippocampus, as well as the entorhinal cortex of AD patient brains. The Sp transcription factor expression levels were further analyzed in cortical neurons in which death is induced by amyloid precursor protein signaling targeting. While the Sp1 levels remained constant, the Sp4 levels were slightly upregulated in response to the death signal. The Sp3 isoforms were rather degraded. Interestingly, when overexpressed by transfection experiments, the three Sp family members induced neuronal apoptosis, Sp3 and Sp4 being the most potent proapoptotic factors over Sp1.. Our data evidence Sp3 and Sp4 as new hallmarks of AD in postmortem human brains and further point out that Sp proteins are potential triggers of neuronal death signaling cascades. Topics: Alzheimer Disease; Amyloid beta-Protein Precursor; Animals; Animals, Newborn; Antibodies; Apoptosis; Brain; Case-Control Studies; Cells, Cultured; Cerebellum; Drug Interactions; Gene Expression Regulation; Humans; Leupeptins; Mice; Neurons; Oligopeptides; Postmortem Changes; Sp3 Transcription Factor; Sp4 Transcription Factor; tau Proteins; Time Factors; Transfection | 2007 |
Estrogen attenuates cardiac ischemia-reperfusion injury via inhibition of calpain-mediated bid cleavage.
Although several studies have shown that the administration of 17beta-estradiol (estrogen) is cardioprotective to ischemia-reperfusion (I/R), the molecular mechanisms are largely unknown. Therefore, we investigated the effects of estrogen on myocardial I/R injury in rat that were sham operated (Sham), ovariectomized (OVX), or ovariectomized and then given estrogen supplementation (OE). Langendorff-perfused rat hearts were subjected to I/R stimuli and the effects of estrogen were examined on cardiac performance. Additionally, we examined the mechanism of estrogen-mediated inhibition of apoptosis. Depression in cardiac contractile function and an increment of calpain activity were observed during I/R in the OVX rats. Estrogen replacement recovered cardiac contractile function and attenuated calpain activity, Bid cleavage, and caspases activities. Through in vitro assay using cardiomyocytes, we demonstrated that addition of H2O2 (100 microM) significantly increased calpain activity, which was attenuated by estrogen. Moreover, calpain activity was inhibited by calpain inhibitors such as ALLN or leupeptin, but not by caspase-8 inhibitor peptide. These results suggest that estrogen protects the heart against I/R injury through the decrease of calpain activity, Bid cleavage and caspase-8 activity. These apoptotic mechanisms may play a critical role on I/R-associated cardiac damage. Topics: Animals; Apoptosis; bcl-2-Associated X Protein; BH3 Interacting Domain Death Agonist Protein; Calpain; Caspase Inhibitors; Caspases; Cells, Cultured; Cysteine Proteinase Inhibitors; Disease Models, Animal; Enzyme Activation; Estradiol; Female; Hydrogen Peroxide; Leupeptins; Myocardial Contraction; Myocardial Reperfusion Injury; Myocardium; Myocytes, Cardiac; Ovariectomy; Proto-Oncogene Proteins c-bcl-2; Rats; Rats, Sprague-Dawley; Severity of Illness Index | 2007 |
Modulation of proteasome activity by vitamin E in THP-1 monocytes.
In THP-1 monocytes, cellular proteasome inhibition by ritonavir or ALLN is associated with increased production of oxidative stress. Both compounds produced comparable amounts of oxidative stress; however, normalization by alpha-tocopherol occurred solely after inhibition by ritonavir, and not by ALLN. Similar to that, alpha-tocopherol could normalize the reduced formation of 3-nitrotyrosine-modified proteins only after ritonavir treatment. In the absence of any proteasome inhibitor, intrinsic cellular proteasome activity was not modulated by alpha-, beta-, and gamma-tocopherols; however, delta-tocopherol, alpha-tocotrienol, and alpha-tocopheryl phosphate could significantly inhibit cellular proteasome activity and increased the level of p27(Kip1) and p53. Since oxidative stress was reduced by alpha-tocopherol only after proteasome inhibition by ritonavir and not by ALLN, it is concluded that, in this experimental system, alpha-tocopherol does not act as an antioxidant but interferes with the inhibitory effect of ritonavir. Topics: Cell Line; Cyclin-Dependent Kinase Inhibitor p27; Humans; Intracellular Signaling Peptides and Proteins; Leupeptins; Monocytes; Oxidative Stress; Protease Inhibitors; Proteasome Endopeptidase Complex; Ritonavir; Tocopherols; Tocotrienols; Tumor Suppressor Protein p53; Tyrosine; Vitamin E | 2007 |
The selective inhibition of nitric oxide production in the avian macrophage cell line HD11.
The production of reactive nitrogen, nitric oxide (NO), has previously been demonstrated to be a major mechanism by which the innate immune system defends against microbial invasion. The induction of many antimicrobial mechanisms is regulated by numerous components during the transduction of the signal from the cell surface to the cell nucleus where response genes are upregulated. Toll-like cell surface receptor activation often leads to sequential modulation of protein tyrosine kinases (PTK), mitogen activated protein kinases (MAPK), degradation of I kappa B (IkappaB) regulatory molecules which, in turn, release the nuclear factor-kappa B (NF-kappaB) family proteins for translocation into the nucleus and subsequent gene transcription. The purpose of this study was to investigate components of the upstream signal transduction pathway induced by bacterial and viral-like stimulation of NO for antimicrobial defense by the transformed chicken macrophage cell line, HD11. We quantified the production of nitrite by chicken macrophages after exposure to selective pharmacological inhibitors of specific signal transduction components prior to stimulation by polyinosinic polycytidylic acid (poly I:C), formalin-fixed Enterococcus gallinarum (EG) or formalin-fixed Klebsiella pneumoniae (KP). We found that NO production induced by dsRNA or bacteria was reduced in a dose dependent manner by specific inhibitors of PTK, p38 MAPK, IkappaB, and NF-kappaB. Inhibition efficacy varied dependent on stimulation by bacterial or viral-like ligands. In general, NO production induced by bacterial stimulation was most effectively reduced by inhibition of p38 MAPK and least effectively reduced by inhibition of IkappaB. NF-kappaB and IkappaB inhibition affected NO production induced by dsRNA more than that induced by bacterial stimulation. Topics: Animals; Caffeic Acids; Cell Line; Chickens; Enterococcus; Enzyme Inhibitors; Genistein; I-kappa B Proteins; Klebsiella pneumoniae; Leupeptins; Macrophage Activation; Macrophages; NF-kappa B; Nitric Oxide; p38 Mitogen-Activated Protein Kinases; Phenylethyl Alcohol; Poly I-C; Poultry Diseases; Protein-Tyrosine Kinases; RNA, Double-Stranded; Signal Transduction | 2006 |
Degradation of perilipin is mediated through ubiquitination-proteasome pathway.
Perilipin protein coats the surface of intracellular lipid droplets and plays fundamental roles in lipid droplet formation and triacylglycerol hydrolysis. Perilipin is transcriptionally regulated through peroxisome proliferator-activated receptor and post-translationally stabilized by stored intracellular neutral lipids. In this study, we show that perilipin protein accumulates in transfected Chinese hamster ovary cells cultured in the presence of fatty acids but in turn is destabilized when lipid precursors for triacylglycerol synthesis are removed from culture serum. Adding fatty acids in the culture medium prevents the degradation of perilipin. Moreover, specific proteasome inhibitors, MG132, lactacystin, and ALLN, block the degradation, whereas inhibitors of other proteases are ineffective. Pulse-chase experiments confirm that perilipin is degraded through proteasome, a process that is inhibited by MG132 or ALLN and blunted by the addition of oleic acid. We have detected the co-immunoprecipitation of perilipin and ubiquitin, thus confirming that perilipin is conjugated to poly-ubiquitin and targeted for proteasomal degradation. Treatment with MG132 increases the expression of perilipin associated with lipid droplets as well as modestly throughout the cytosol. We conclude that the degradation of perilipin is mediated through an ubiquitination-proteasome pathway, which suggests another mode for the post-translational regulation of perilipin. Topics: Animals; Carrier Proteins; CHO Cells; Cricetinae; Culture Media; Fatty Acids; Leupeptins; Oleic Acid; Perilipin-1; Phosphoproteins; Proteasome Endopeptidase Complex; Recombinant Proteins; Transfection; Triglycerides; Ubiquitin | 2006 |
Prodomain processing of recombinant plasmepsin II and IV, the aspartic proteases of Plasmodium falciparum, is auto- and trans-catalytic.
Prodomain processing of the four food vacuole plasmepsins (PMs), the malarial aspartic proteases, is prerequisite for their activity on hemoglobin degradation of the parasite Plasmodium falciparum. Although previous studies have suggested the involvement of a calpain-like PM convertase in the processing of PMs, the underlying mechanism of their processing remains to be clarified. Here, to investigate the mechanism by which food vacuole PM II and IV are processed, we used their wild-type and mutant proteins in which the catalytic Asp residue in two active-site motifs was mutated, as well as protease inhibitors. Autocatalytic processing of wild-type PM II and IV was inhibited only by an aspartic protease inhibitor pepstatin A. Unexpectedly, their proteolytic activities were inhibited not only by pepstatin A but also by calpain inhibitor ALLN. The active-site mutants of both PM II and IV showed neither autocatalytic processing nor proteolytic activities. However, the mutants of both PMs were efficiently processed upon incubation with their respective wild type proteins. Furthermore, the mutants of both PMs were processed upon incubation with each other's wild-type PM in both pepstatin A- and ALLN-sensitive manners. These results suggest that the processing of PM II and IV occurs via an intra- and inter-molecular autocatalytic event as well as via a transcatalytic event between them. Topics: Animals; Aspartic Acid Endopeptidases; Biotransformation; Catalysis; Leupeptins; Mutation; Pepstatins; Plasmodium falciparum; Protein Structure, Tertiary; Protozoan Proteins; Recombinant Fusion Proteins | 2006 |
Multiple signaling pathways mediate compaction of collagen matrices by EGF-stimulated fibroblasts.
Fibroblasts stimulated by EGF within collagen matrices generate contraction forces that are likely of importance to cell migration and matrix compaction during wound healing. We have employed an in vitro fibroblast-embedded collagen matrix compaction assay to ascertain signaling pathway components downstream of EGFR activation leading to generation and transmission of contractile force. EGF compacts this floating collagen matrix to a similar extent as PDGF. We demonstrate that compaction requires EGFR kinase activity, yet is maximal in magnitude at intermediate EGF concentrations. This suggests that transmission of EGFR-induced contractile force to the matrix can be mitigated by consequent anti-adhesive effects of EGFR signaling in a dose-dependent manner. Treatment with pharmacological inhibitors demonstrated involvement of the signaling components extracellular signal-regulated kinase (ERK), Rho kinase, and myosin light chain kinase (MLCK) in the force generation and/or transmission process. Moreover, treatment with the pan-calpain inhibitor ALLN and isoform-specific downregulation of m-calpain (CAPN2) using RNA interference determined m-calpain to be a key component of the EGF-induced force response. ALLN treatment modulated the compaction response in a biphasic manner, enhancing matrix deformation to the greatest extent at intermediate concentrations. Our findings have thus identified key signals downstream of EGFR, which integrate in a complex manner to generate and transmit contractile forces to yield matrix deformation. Topics: Animals; Cell Line, Tumor; Cells, Cultured; Collagen; Dose-Response Relationship, Drug; Down-Regulation; Epidermal Growth Factor; Extracellular Matrix; Fibroblasts; Glycoproteins; Humans; Integrin beta1; Intracellular Signaling Peptides and Proteins; Leupeptins; Mice; Mitogen-Activated Protein Kinase 3; Peptides; Protein Serine-Threonine Kinases; rho-Associated Kinases; RNA, Small Interfering; Signal Transduction | 2006 |
Cytoplasmic lipid droplets are sites of convergence of proteasomal and autophagic degradation of apolipoprotein B.
Lipid esters stored in cytoplasmic lipid droplets (CLDs) of hepatocytes are used to synthesize very low-density lipoproteins (VLDLs), into which apolipoprotein B (ApoB) is integrated cotranslationally. In the present study, by using Huh7 cells, derived from human hepatoma and competent for VLDL secretion, we found that ApoB is highly concentrated around CLDs to make "ApoB-crescents." ApoB-crescents were seen in <10% of Huh7 cells under normal conditions, but the ratio increased to nearly 50% after 12 h of proteasomal inhibition by N-acetyl-L-leucinyl-L-leucinyl-L-norleucinal. Electron microscopy showed ApoB to be localized to a cluster of electron-lucent particles 50-100 nm in diameter adhering to CLDs. ApoB, proteasome subunits, and ubiquitinated proteins were detected in the CLD fraction, and this ApoB was ubiquitinated. Interestingly, proteasome inhibition also caused increases in autophagic vacuoles and ApoB in lysosomes. ApoB-crescents began to decrease after 12-24 h of proteasomal inhibition, but the decrease was blocked by an autophagy inhibitor, 3-methyladenine. Inhibition of autophagy alone caused an increase in ApoB-crescents. These observations indicate that both proteasomal and autophagy/lysosomal degradation of ApoB occur around CLDs and that the CLD surface functions as a unique platform for convergence of the two pathways. Topics: Apolipoproteins B; Autophagy; Carcinoma, Hepatocellular; Cell Line, Tumor; Cysteine Proteinase Inhibitors; Cytoplasm; Humans; Leupeptins; Lipids; Lipoproteins, VLDL; Liver Neoplasms; Proteasome Endopeptidase Complex | 2006 |
Remodeling of connexin 43 in the diabetic rat heart.
In the Streptozotocin-induced diabetic rat heart, a decrease in the conductivity and suppression of electrical cell-to-cell coupling were observed. To clarify this mechanism, the present study was performed to investigate alterations of the gap junction connexin 43 (Cx43) using immunoblotting, immunohistochemistry, electron-microscopic analyses. An enhanced activation of PKCepsilon, an augmentation of PKCepsilon-mediated phosphorylation of Cx43, a decrease in the total amount of Cx43, a reduction in the area of immunoreactive particles for Cx43 at the intercalated disk, distribution of Cx43 to cell periphery or cytoplasm and the internalization approximately annular profiles of the gap junction were all characteristically recognized in the diabetic heart. Such abnormalities in the expression of Cx43 were alleviated by treatment with either lysosomal (NH(4)Cl, Leupeptin) or proteasomal inhibitor (ALLN). These results suggest that the PKCepsilon-mediated hyperphosphorylation of Cx43 makes Cx43 vulnerable to proteolytic degradation and that a decrease in the conductivity in the diabetic heart is also caused by a decrease in the number of gap junction channels due to an acceleration of the proteolytic degradation of Cx43. The remodeling of Cx43 induced by the activation of PKCepsilon may therefore contribute to the formation of the arrhythmogenic substrate in the diabetic heart. The cardioprotective effect of the remodeling of Cx43 by PKCepsilon is discussed. Topics: Animals; Cell Communication; Connexin 43; Diabetes Mellitus, Experimental; Electrophysiology; Gap Junctions; Heart; Immunoblotting; Immunohistochemistry; Leupeptins; Male; Microscopy, Electron; Myocardium; Phosphorylation; Protein Isoforms; Protein Kinase C; Rats; Rats, Wistar; Streptozocin | 2006 |
Differential regulation of eotaxin-1/CCL11 and eotaxin-3/CCL26 production by the TNF-alpha and IL-4 stimulated human lung fibroblast.
Allergic asthma and allergic dermatitis are chronic inflammatory diseases and are characterized by an accumulation of eosinophils at sites of inflammation. Eotaxin-1/CCL11 and eotaxin-3/CCL26 are members of the CC chemokine family, which are known to be potent chemoattractants for eosinophils. We observed that a human lung fibroblast, HFL-1 produces eotaxin-1 and -3 in response to TNF-alpha plus IL-4 stimulation, accompanied with NF-kappaB and STAT6 activation. We explored which signaling pathways are operative in the production of eotaxin-1 and -3 using several inhibitors. Eotaxin-1/CCL11 production was inhibited by a p38 mitogen-activated protein kinase (MAPK) inhibitor, SB203580, but not by the MEK (MAPK/ERK kinase) inhibitors, PD98059 and U0126. In contrast, eotaxin-3/CCL26 production was inhibited similarly by PD98059 as well as U0126 and SB203580. In addition, two proteasome inhibitors, N-acetyl-leucyl-leucyl-norleucinal (ALLN) and bortezomib with significant inhibitory activity on NF-kappaB activation, inhibited eotaxin-1/CCL11 production with IC50 8 microM for ALLN and IC50 16 nM for bortezomib. In contrast, eotaxin-3/CCL26 production was not inhibited significantly up to 10 microM of ALLN (IC50 16 microM) and up to 10 nM of bortezomib (IC50 11 nM), giving inhibition of eotaxin-3/CCL26 less sensitive than eotaxin-1/CCL11 production by the proteasome inhibitors. Synergistic inhibition was observed among lower doses of SB203580 and proteasome inhibitors, particularly in the eotaxin-1/CCL11 production. No such prominent synergism was found on the eotaxin-3/CCL26 production. The suppression of eotaxin family production by these inhibitors may be efficacious against allergic diseases. Topics: Asthma; Boronic Acids; Bortezomib; Cell Line; Chemokine CCL11; Chemokine CCL26; Chemokines, CC; Drug Synergism; Enzyme Inhibitors; Fibroblasts; Humans; Hypersensitivity; Immunoblotting; Interleukin-4; Leupeptins; Lung; Mitogen-Activated Protein Kinases; NF-kappa B; Pyrazines; Recombinant Proteins; STAT6 Transcription Factor; Tumor Necrosis Factor-alpha | 2006 |
In vivo heteromer formation. Expression of soluble betaA4-crystallin requires coexpression of a heteromeric partner.
The beta-crystallins are a family of long-lived, abundant structural proteins that are coexpressed in the vertebrate lens. As beta-crystallins form heteromers, a process that involves transient exposure of hydrophobic interfaces, we have examined whether in vivobeta-crystallin assembly is enhanced by protein chaperones, either small heat shock proteins, Hsp27 or alphaB-crystallin, or Hsp70. We show here that betaA4-crystallin is abundantly expressed in HeLa cells, but rapidly degraded, irrespective of the presence of Hsp27, alphaB-crystallin or Hsp70. Degradation is even enhanced by Hsp70. Coexpression of betaA4-crystallin with betaB2-crystallin yielded abundant soluble betaA4-betaB2-crystallin heteromers; betaB1-crystallin was much less effective in solubilizing betaA4-crystallin. As betaB2-crystallin competed for betaA4-crystallin with Hsp70 and the proteasomal degradation pathway, betaB2-crystallin probably captures an unstable betaA4-crystallin intermediate. We suggest that the proper folding of betaA4-crystallin is not mediated by general chaperones but requires a heteromeric partner, which then also acts as a dedicated chaperone towards betaA4-crystallin. Topics: Animals; beta-Crystallin A Chain; beta-Crystallin B Chain; beta-Crystallins; Cricetinae; Enzyme Inhibitors; Gene Expression Regulation; Genes, Reporter; Heat-Shock Proteins; HeLa Cells; HSP70 Heat-Shock Proteins; Humans; Isoelectric Focusing; Leupeptins; Luciferases; Molecular Chaperones; Mutation; Protein Folding; Solubility | 2006 |
Secreted proteins from Mycobacterium tuberculosis gain access to the cytosolic MHC class-I antigen-processing pathway.
CD8+ T cells play an important role in the host response to infection with Mycobacterium tuberculosis (Mtb). Mtb resides in an arrested phagosome that is phenotypically similar to an early endosome. The mechanisms by which Mtb-derived Ags gain access to the HLA-I-processing pathway are incompletely characterized. Studies with CD8+ T cell lines have suggested that Mtb Ags gain access to the HLA-I pathway in an alternate vacuolar pathway that is both brefeldin A (BFA) and TAP independent. To define the requirements of entry of Ag into the HLA-I pathway, we have used human CD8+ T cell clones specific for the secreted Mtb Ag CFP10. Human monocyte-derived dendritic cells were pulsed with CFP10 expressed in a recombinant adenovirus, surface adsorbed to microspheres, or in its native form by Mtb. When delivered by adenovirus, processing and presentation of CFP10 were blocked by both BFA and the proteasomal blocker lactacystin. In contrast, processing of CFP10 adsorbed to the surface of microspheres was not affected by either of these Ag-processing inhibitors. BFA, lactacystin, and TAP inhibition blocked the recognition of Mtb-infected dendritic cells, suggesting that processing was via a cytosolic pathway for this secreted protein Ag. We conclude that secreted proteins from Mtb can be processed in a BFA- and proteasome-dependent manner, consistent with egress of Ag into the cytosol and subsequent loading of proteasomally derived peptides. Topics: Adenoviruses, Human; Amino Acid Sequence; Antigen Presentation; ATP-Binding Cassette Transporters; Bacterial Proteins; Brefeldin A; Cell Line; Cell Line, Transformed; Clone Cells; Cytosol; Histocompatibility Antigens Class I; Humans; Immediate-Early Proteins; Lactic Acid; Leupeptins; Microspheres; Molecular Sequence Data; Mycobacterium tuberculosis; Polyglycolic Acid; Polylactic Acid-Polyglycolic Acid Copolymer; Polymers; Proteasome Inhibitors; Signal Transduction; Viral Proteins | 2006 |
Albumin induces endoplasmic reticulum stress and apoptosis in renal proximal tubular cells.
Chronic proteinuria appears to be a key factor in tubulointerstitial damage. Recent studies have emphasized a pathogenic role of endoplasmic reticulum (ER) stress which is induced by the accumulation of misfolded proteins in ER, extracellular stress, etc. In the present study, we investigated ER stress and ER stress-induced apoptosis in proximal tubular cells (PTCs). Immortalized rat PTCs (IRPTCs) were cultured with bovine serum albumin (BSA). The viability of IRPTCs decreased proportionately with BSA overload in a time-dependent manner. Quantitative real-time polymerase chain reaction analysis revealed that 40 mg/ml BSA increases mRNA of ER stress markers by 7.7- and 4.6-fold (glucose-regulated protein 78 (GRP78) and oxygen-regulated protein 150 (ORP150), respectively) as compared to control. The increased expression of ORP150 and GRP78 in IRPTCs with albumin overload was detected by Western blot and immunofluorescence study. These in vitro observations were supported by in vivo studies, which demonstrated that ER stress proteins were upregulated at PTCs in experimental proteinuric rats. Furthermore, increased ER stress-induced apoptosis and activation of caspase-12 were observed in IRPTCs with albumin overload and kidneys of experimental proteinuric rats. We confirmed that apoptotic cell death was attenuated by co-incubation with caspase-3 inhibitor or calpain inhibitors. These results indicate that the ER stress-induced apoptosis pathway contributed to the insult of tubular cells by proteinuria. In conclusion, renal tubular cells exposed to high protein load suffer from ER stress. ER stress may subsequently lead to tubular damage by activation of caspase-12. Topics: Acrylates; Animals; Apoptosis; Calpain; Caspase 12; Caspase 3; Caspase Inhibitors; Caspases; Cell Death; Cell Line; Cell Survival; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Endoplasmic Reticulum; Gene Expression Regulation; Heat-Shock Proteins; HSP70 Heat-Shock Proteins; Kidney Tubules, Proximal; Leupeptins; Molecular Chaperones; Oligopeptides; Proteins; Proteinuria; Rats; Serum Albumin, Bovine; Stress, Physiological | 2006 |
[Effect of ubiquitin-proteasome pathway on inflammatory reaction in intestine and its barrier function in rats with postburn sepsis].
To study the effect of ubiquitin-proteasome pathway inhibition on intestinal nuclear factor-KappaB (NF-KappaB) activity and tumor necrosis factor-alpha (TNF-alpha) release as well as plasma diamine oxidase (DAO) activity in rats with postburn sepsis.. Rats were subjected to 30% total body surface area (TBSA) full-thickness scald injury, followed by intraperitoneal injection of lipopolysaccharide (LPS) to mimic postburn sepsis. Sixty Wistar rats were randomly divided into normal control group, sepsis group, sepsis with proteasome inhibitor N-Acetyl leucinyl leucinyl norleucinal (ALLN) treatment group and sepsis with NF-KappaB inhibitor pyrrolidine dithiocarbamate (PDTC) treatment group. NF-KappaB activity, TNF-alpha protein content, and plasma DAO activity were determined by electrophoretic mobility shift assay (EMSA), enzyme-linked immunosorbent assay (ELISA), and spectrophotometric method, respectively.. The results showed that NF-KappaB activity was markedly activated and reached its peak 1 hour after scalding and injection of LPS in each group (all P<0.01), then reduced gradually. Both ALLN and PDTC could decrease intestinal NF-KappaB activity at 1 hour and 2 hours after injury. TNF-alpha release was reduced by ALLN at 1 hour after injury (P<0.01). Plasma DAO activity was significantly elevated after scalding and injection of LPS (P<0.01). Pretreatment with PDTC or ALLN could not lower the activity of DAO.. The results suggest that early treatment with inhibitor of ubiquitin-proteasome pathway might decrease the intestinal inflammatory reaction, but exert no effect on intestinal barrier function in rats with postburn sepsis. Topics: Amine Oxidase (Copper-Containing); Animals; Burns; Cysteine Proteinase Inhibitors; Disease Models, Animal; Intestinal Mucosa; Intestines; Leupeptins; Male; NF-kappa B; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Pyrrolidines; Random Allocation; Rats; Rats, Wistar; Sepsis; Thiocarbamates; Tumor Necrosis Factor-alpha; Ubiquitin | 2006 |
Perturbed interactions of mutant proteolipid protein/DM20 with cholesterol and lipid rafts in oligodendroglia: implications for dysmyelination in spastic paraplegia.
Missense mutations in the human PLP1 gene lead to dysmyelinating diseases with a broad range of clinical severity, ranging from severe Pelizaeus-Merzbacher disease (PMD) to milder spastic paraplegia type 2 (SPG-2). The molecular pathology has been generally attributed to endoplasmic reticulum (ER) retention of misfolded proteolipid protein (PLP) (and its splice isoform DM20) and induction of the unfolded protein response. As opposed to previous studies of heterologous expression systems, we have analyzed PLP/DM20 trafficking in oligodendroglial cells, thereby revealing differences between PMD and SPG-2-associated PLP/DM20 isoforms. PLP(A242V) and DM20(A242V) (jimpy-msd in mice), associated with severe PMD-like phenotype in vivo, were not only retained in the ER but also interfered with oligodendroglial process formation. In contrast, glial cells expressing SPG-2-associated PLP(I186T) or DM20(I186T) (rumpshaker in mice) developed processes, and mutant PLP/DM20 reached a late endosomal/lysosomal compartment. Unexpectedly, PLP/DM20 with either substitution exhibited impaired cholesterol binding, and the association with lipid raft microdomains was strongly reduced. Turnover analysis demonstrated that mutant PLP was rapidly degraded in oligodendroglial cells, with half-lives for PLP > PLP(I186T) > PLP(A242V). Protein degradation was specifically sensitive to proteasome inhibition, although PLP/DM20(I186T) degradation was also affected by inhibition of lysosomal enzymes. We conclude that, in addition to ER retention and unfolded protein response (UPR) induction, impaired cholesterol binding and lipid raft association are characteristic cellular defects of PLP1-missense mutations. Mutant protein is rapidly cleared and does not accumulate in oligodendroglial cells. Whereas UPR-induced cell death governs the PMD phenotype of the msd mutation, we propose that impaired cholesterol and lipid raft interaction of the rsh protein may contribute to the dysmyelination observed in SPG-2. Topics: Animals; Blotting, Western; Cells, Cultured; Cholesterol; Cricetinae; Cricetulus; Cysteine Proteinase Inhibitors; Gene Expression; Immunohistochemistry; Immunoprecipitation; Leupeptins; Membrane Microdomains; Mice; Mice, Neurologic Mutants; Mutant Proteins; Myelin Proteolipid Protein; Nerve Tissue Proteins; Oligodendroglia; Protein Transport; Subcellular Fractions; Time Factors; Transfection | 2006 |
Antitumorigenic effect of proteasome inhibitors on insulinoma cells.
Malignant insulinoma is a critical cancer form with a poor prognosis. Because cure by surgery is infrequent, effective chemotherapy is in demand. Induction of cell death in tumor cells by proteasome inhibitors is emerging as a potential strategy in cancer therapy. Here we investigated whether inhibition of the proteasome has an antitumorigenic potential in insulinoma cells. Exposure of mouse betaTC3 insulinoma cells to the proteasome inhibitor N-Acetyl-Leu-Leu-Nle-CHO (ALLN) reduced cell viability, activated caspase-3, induced apoptosis, and suppressed insulin release. Treatment with ALLN also resulted in phosphorylation of c-jun N-terminal kinase (JNK) and an increase in in vitro phosphorylation of c-jun. In insulinoma cells with impaired JNK signaling, ALLN-induced apoptosis was significantly suppressed. Another proteasome inhibitor, lactacystin, also stimulated JNK activation, caused activation of caspase-3, suppressed cell viability, and induced apoptosis in betaTC3 and rat INS-1E cells. Both ALLN and lactacystin caused a marked decrease in the cellular amount of the JNK scaffold protein JNK-interacting protein 1/islet-brain-1. In primary pancreatic rat islet cells, proteasome inhibition reduced insulin secretion but had no impact on cell viability and even partially protected against the toxic effect of proinflammatory cytokines. Our findings demonstrate that proteasome inhibitors possess antitumorigenic and antiinsulinogenic effects on insulinoma cells. Topics: Acetylcysteine; Adaptor Proteins, Signal Transducing; Animals; Antineoplastic Agents; Apoptosis; Binding Sites; Cell Line, Tumor; Cysteine Proteinase Inhibitors; Insulinoma; JNK Mitogen-Activated Protein Kinases; Leupeptins; Mice; Pancreatic Neoplasms; Proteasome Inhibitors; Rats; Signal Transduction; Tumor Suppressor Protein p53 | 2005 |
Corticosterone modulates noradrenaline-induced melatonin synthesis through inhibition of nuclear factor kappa B.
In chronically inflamed animals, adrenal hormones exert a positive control on the secretion of melatonin by the pineal gland. In this paper, the mechanism of corticosterone as a modulator of melatonin and N-acetylserotonin (NAS) was determined. Rat pineal glands in culture, stimulated for 5 hr with noradrenaline (10 nm), were previously incubated with corticosterone (1.0 nm-1.0 microm) for 48 hr in the presence or absence of the glucocorticoid receptor (GR) antagonist, mifepristone (1.0 microm), the proteasome inhibitor, N-acetyl-leucinyl-leucinyl-norleucinal-H (ALLN, 12.5 microm) or the antagonist of the nuclear factor kappa B (NFkappaB), pyrrolidinedithiocarbamate (PDTC, 12.5 microm). Corticosterone potentiated noradrenaline-induced melatonin and NAS production in a bell-shaped manner. The increase in NAS (12.9 +/- 2.7, n=6 versus 34.3 +/- 8.3 ng per pineal) and melatonin (16.3 +/- 2.0, n=6 versus 44.3 +/- 12.9 ng per pineal) content induced by 1 microm corticosterone was blocked by mifepristone, and mimicked by ALLN and PDTC. The presence of GRs was shown by [3H]-dexamethasone binding (0.30 +/- 0.09 pmol/mg protein) and corticosterone inhibition of NFkappaB nuclear translocation was demonstrated by electromobility shift assay. Therefore, corticosterone potentiates noradrenaline-induced melatonin and NAS production through GR inhibition of NFkappaB nuclear translocation. To the best of our knowledge, this is the first time that this relevant pathway for passive and acquired immune response is shown to modulate melatonin production in pineal gland. Topics: Animals; Base Sequence; Corticosterone; Dexamethasone; DNA; Female; Hormone Antagonists; Leupeptins; Male; Melatonin; Mifepristone; NF-kappa B; Norepinephrine; Organ Culture Techniques; Pineal Gland; Pyrrolidines; Rats; Rats, Wistar; Receptors, Glucocorticoid; Serotonin; Thiocarbamates | 2005 |
Role of glycogen synthase kinase 3beta in rapamycin-mediated cell cycle regulation and chemosensitivity.
The mammalian target of rapamycin is a serine-threonine kinase that regulates cell cycle progression. Rapamycin and its analogues inhibit the mammalian target of rapamycin and are being actively investigated in clinical trials as novel targeted anticancer agents. Although cyclin D1 is down-regulated by rapamycin, the role of this down-regulation in rapamycin-mediated growth inhibition and the mechanism of cyclin D1 down-regulation are not well understood. Here, we show that overexpression of cyclin D1 partially overcomes rapamycin-induced cell cycle arrest and inhibition of anchorage-dependent growth in breast cancer cells. Rapamycin not only decreases endogenous cyclin D1 levels but also decreases the expression of transfected cyclin D1, suggesting that this is at least in part caused by accelerated proteolysis. Indeed, rapamycin decreases the half-life of cyclin D1 protein, and the rapamycin-induced decrease in cyclin D1 levels is partially abrogated by proteasome inhibitor N-acetyl-leucyl-leucyl-norleucinal. Rapamycin treatment leads to an increase in the kinase activity of glycogen synthase kinase 3beta (GSK3beta), a known regulator of cyclin D1 proteolysis. Rapamycin-induced down-regulation of cyclin D1 is inhibited by the GSK3beta inhibitors lithium chloride, SB216763, and SB415286. Rapamycin-induced G1 arrest is abrogated by nonspecific GSK3beta inhibitor lithium chloride but not by selective inhibitor SB216763, suggesting that GSK3beta is not essential for rapamycin-mediated G1 arrest. However, rapamycin inhibits cell growth significantly more in GSK3beta wild-type cells than in GSK3beta-null cells, suggesting that GSK3beta enhances rapamycin-mediated growth inhibition. In addition, rapamycin enhances paclitaxel-induced apoptosis through the mitochondrial death pathway; this is inhibited by selective GSK3beta inhibitors SB216763 and SB415286. Furthermore, rapamycin significantly enhances paclitaxel-induced cytotoxicity in GSK3beta wild-type but not in GSK3beta-null cells, suggesting a critical role for GSK3beta in rapamycin-mediated paclitaxel-sensitization. Taken together, these results show that GSK3beta plays an important role in rapamycin-mediated cell cycle regulation and chemosensitivity and thus significantly potentiates the antitumor effects of rapamycin. Topics: Aminophenols; Antibiotics, Antineoplastic; Antimanic Agents; Apoptosis; Breast Neoplasms; Cell Cycle; Cyclin D1; Cysteine Proteinase Inhibitors; Down-Regulation; Drug Resistance, Neoplasm; Female; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Half-Life; Humans; Indoles; Leupeptins; Lithium Chloride; Maleimides; Mitochondria; NF-kappa B; Paclitaxel; Proteasome Inhibitors; Sirolimus | 2005 |
Degradation of tyrosinase induced by phenylthiourea occurs following Golgi maturation.
Tyrosinase, the rate-limiting enzyme of melanin synthesis, is a di-copper metalloprotein that catalyzes the conversion of L-tyrosine to L-DOPAquinone. Phenylthiourea (PTU) is a well-known inhibitor of tyrosinase and melanin synthesis and is known to interact with sweet potato catechol oxidase, an enzyme possessing copper binding domain homology to tyrosinase. While PTU is frequently used to induce hypopigmentation in biological systems, little is known about its effects on tyrosinase and other melanogenic proteins. We have found that PTU induces degradation of tyrosinase but not of other melanogenic proteins including the tyrosinase-related metalloproteins tyrosinase-related protein (Tyrp)1 and Tyrp2. Using pulse-chase analysis coupled with glycosidase digestion, we observed that tyrosinase degradation occurs following complete maturation of the protein and that degradation was reversed by cysteine protease inhibitor E64 but not proteasome inhibitor N-acetyl-L-leucinyl-L-leucinyl-L-norleucinal. We conclude that PTU specifically induces tyrosinse degradation following Golgi maturation. Our data suggest that in addition to well-known ER-directed quality control, tyrosinase is also subject to post-Golgi quality control. Topics: Animals; Cell Line; Cysteine Proteinase Inhibitors; Golgi Apparatus; Leucine; Leupeptins; Melanins; Melanocytes; Mice; Monophenol Monooxygenase; Oxidoreductases; Phenylthiourea; Proteasome Inhibitors | 2005 |
Degradation of trafficking-defective long QT syndrome type II mutant channels by the ubiquitin-proteasome pathway.
Mutations in the human ether-a-go-go-related gene (hERG) cause chromosome 7-linked long QT syndrome type II (LQT2). We have shown previously that LQT2 mutations lead to endoplasmic reticulum (ER) retention and rapid degradation of mutant hERG proteins. In this study we examined the role of the ubiquitin-proteasome pathway in the degradation of the LQT2 mutation Y611H. We showed that proteasome inhibitors N-acetyl-L-leucyl-L-leucyl-L-norleucinal and lactacystin but not lysosome inhibitor leupeptin inhibited the degradation of Y611H mutant channels. In addition, ER mannosidase I inhibitor kifunensine and down-regulation of EDEM (ER degradation-enhancing alpha-mannosidase-like protein) also suppressed the degradation of Y611H mutant channels. Proteasome inhibition but not mannosidase inhibition led to the accumulation of full-length hERG protein in the cytosol. The hERG protein accumulated in the cytosol was deglycosylated. Proteasome inhibition also resulted in the accumulation of polyubiquitinated hERG channels. These results suggest that the degradation of LQT2 mutant channels is mediated by the cytosolic proteasome in a process that involves mannose trimming, polyubiquitination, and deglycosylation of mutant channels. Topics: Acetylcysteine; Alkaloids; Blotting, Western; Cell Line; Cell Membrane; Cysteine Proteinase Inhibitors; Cytosol; Down-Regulation; Electrophoresis, Polyacrylamide Gel; Endoplasmic Reticulum; Enzyme Inhibitors; Glycosylation; Green Fluorescent Proteins; Humans; Immunoprecipitation; Leupeptins; Long QT Syndrome; Mutation; Potassium Channels; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Ribonucleases; Subcellular Fractions; Time Factors; Transfection; Ubiquitin | 2005 |
Adeno-associated virus (AAV)-7 and -8 poorly transduce vascular endothelial cells and are sensitive to proteasomal degradation.
Transduction of the vascular endothelium by adeno-associated virus (AAV) vectors would have broad appeal for gene therapy. However, levels of transduction by AAV serotype-2 are low, an observation linked to deficiencies in endothelial cell binding, sequestration of virions in the extracellular matrix and/or virion degradation by the proteasome. Strategies to improve transduction of endothelial cells include AAV-2 capsid targeting using small peptides isolated by phage display or the use of alternate serotypes. Previously, we have shown that AAV serotypes-3 through -6 transduce endothelial cells with poor efficiency. Recently, AAV serotypes-7 and -8 have been shown to mediate efficient transduction of the skeletal muscle and liver, respectively, although their infectivity profile for vascular cells has not been addressed. Here, we show that AAV-7 and -8 also transduce endothelial cells with poor efficiency and the levels of transgene expression are markedly enhanced by inhibition of the proteasome. In both cases proteasome blockade enhances the nuclear translocation of virions. We further show that this is vascular cell-type selective since transduction of smooth muscle cells is not sensitive to proteasome inhibition. Analysis in intact blood vessels corroborated these findings and suggests that proteasome degradation is a common limiting factor for endothelial cell transduction by AAV vectors. Topics: Animals; Aorta, Thoracic; Cell Line; Cells, Cultured; Dependovirus; DNA, Viral; Endothelial Cells; Genetic Therapy; Genetic Vectors; Humans; Leupeptins; Mice; Muscle, Smooth, Vascular; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Rats; Rats, Inbred WKY; Saphenous Vein; Serotyping; Transduction, Genetic | 2005 |
Participation of the ubiquitin-proteasome pathway in rat oocyte activation.
The role of the ubiquitin-proteasome pathway (UPP) in mitosis is well known. However, its role in meiotic division is still poorly documented, especially in the activation of mammalian oocytes. In this study, the role of proteasome in the spontaneous and parthenogenetic activation of rat oocytes was investigated. We found that ALLN, an inhibitor of proteasome, when applied to metaphase II oocytes, inhibited spontaneous activation, blocked extrusion of the second polar body (PB) and caused the withdrawal of the partially extruded second PB. ALLN also inhibited the parthenogenetic activation induced by cycloheximide, but had no effect on the formation of pronuclei in activated eggs. In metaphase and anaphase, ubiquitin and proteasome localized to the meiotic spindle, concentrating on both sides of the oocyte-second PB boundary during PB extrusion. This pattern of cellular distribution suggests that UPP may have a role in regulating nuclear division and cytokinesis. Ubiquitin was seen to form a ring around the pronucleus, whereas proteasome was evenly distributed in the pronuclear region. Taken together, our results indicate that (1) UPP is required for the transitions of oocytes from metaphase II to anaphase II and from anaphase II to the end of meiosis; and (2) the UPP plays a role in cytokinesis of the second meiotic division. Topics: Animals; Cell Nucleus Structures; Cells, Cultured; Colchicine; Cycloheximide; Cysteine Proteinase Inhibitors; Cytochalasin B; Female; Leupeptins; Meiosis; Oocytes; Parthenogenesis; Proteasome Endopeptidase Complex; Rats; Rats, Sprague-Dawley; Subcellular Fractions; Ubiquitin | 2005 |
Regulation of polymeric immunoglobulin receptor expression by reovirus.
Polymeric immunoglobulin receptor (pIgR) transcytoses dimeric IgA and IgA-coated immune complexes from the lamina propria across epithelia and into secretions. The effect of reovirus infection on regulation of pIgR expression in the human intestinal epithelial cell line HT-29 was characterized in this report. Both replication-competent and UV-inactivated reovirus at m.o.i. equivalents of 1-100 p.f.u. per cell upregulated pIgR mRNA by 24 h post-infection and intracellular pIgR protein was increased at 48 h following exposure to UV-inactivated virus. Binding of virus to HT-29 cells was required, as pre-incubating virus with specific antiserum, but not non-immune serum, inhibited reovirus-mediated pIgR upregulation. Endosomal acidification leading to uncoating of virus is a required step for pIgR upregulation, as ammonium chloride or bafilomycin A1 pre-treatment inhibited virus-induced pIgR upregulation. Inhibition experiments using the calpain inhibitor N-acetyl-leucyl-leucyl-norleucinal suggested that calpains are involved in reovirus-mediated pIgR upregulation. Upregulation of pIgR following virus infection appears to be an innate immune response against invading pathogens that could help the host clear infection effectively. Signalling induced by microbes and their products may serve to augment pIgR-mediated transcytosis of IgA, linking the innate and acquired immune responses to viruses. Topics: Calpain; Cell Line, Tumor; Dose-Response Relationship, Drug; Humans; Intestinal Mucosa; Leupeptins; Mammalian orthoreovirus 3; Receptors, Polymeric Immunoglobulin; Ultraviolet Rays; Up-Regulation | 2005 |
Activation of multiple pathways during photoreceptor apoptosis in the rd mouse.
The primary purpose of this study was to characterize photoreceptor apoptosis in the rd mouse. Given that apoptosis is the final common pathway in many cases of retinal degeneration, the ability to retard or even arrest this process may ameliorate retinal disorders such as retinitis pigmentosa (RP). The absence of any recognized therapy emphasizes the fact that a detailed knowledge of the molecular events involved is necessary to identify rational targets for therapeutic intervention.. Flow cytometry was used to measure physical and chemical characteristics in the photoreceptor population. Individual cells flow in suspension past one or more lasers, scattering light and emitting fluorescence. Western blot techniques demonstrated cleavage of calpain-specific substrates. Retinal explant cultures were used for inhibitor studies. Postnatal day 10 (P(10)) rd retinas were cultured without retinal pigment epithelium (RPE) attached up to P(17).. This study demonstrated calcium overload in the cytosol and subsequently in mitochondria. Mitochondrial membrane depolarization and reactive oxygen species (ROS) were detected later, during the peak of cell death. Analysis of downstream events indicated early activation of calcium-activated calpains. Treatment of rd retinal explants with the calpain inhibitor N-acetyl-Leu-Leu-Nle-CHO (ALLN) successfully inhibited calpain-induced alpha-fodrin cleavage, yet it did not protect against photoreceptor degeneration. Finally, the results demonstrate an increase in the levels of both precursor and processed forms of the aspartate protease cathepsin D.. Excessive calcium influx is an early event that initiates the activation of calcium-activated proteases. However, these proteases are not singularly the cause of death, because their inhibition does not prevent apoptosis. Indeed, the results presented herein suggest that multiple pathways are involved and that each of these components may have to be addressed for cell death to be successfully inhibited. Topics: Animals; Apoptosis; Blotting, Western; Calcium; Calpain; Cathepsin D; Cell-Free System; Flow Cytometry; Fluorescent Antibody Technique, Indirect; Immunoenzyme Techniques; In Situ Nick-End Labeling; Leupeptins; Mice; Mice, Inbred C3H; Mice, Inbred C57BL; Organ Culture Techniques; Photoreceptor Cells, Vertebrate; Reactive Oxygen Species; Retinal Degeneration; Signal Transduction | 2005 |
Hepatitis C virus nonstructural proteins inhibit apolipoprotein B100 secretion.
Host genes involved in lipid metabolism are differentially regulated during the early stages of hepatitis C virus (HCV) infection. The majority of lipids synthesized in the liver are exported to other tissues in the form of lipoproteins. The formation of these lipoproteins is dependent upon the association of triglycerides with apolipoprotein B100. Using the HCV subgenomic replicon expression system, we show that secretion of apoB100 is significantly reduced. Inhibition of apoB100 degradation by ALLN did not improve secretion. Triglyceride levels as well as microsomal triglyceride transfer protein mRNA and activity levels were reduced in replicon-expressing cells, indicating potential reasons for the observed decrease. Further evidence is presented for the interaction between the HCV nonstructural protein 5A and apoB100. These results provide further insight into the alteration of lipid metabolism by HCV. Topics: Apolipoprotein B-100; Apolipoproteins B; Carcinoma, Hepatocellular; Carrier Proteins; Cell Line, Tumor; Cysteine Proteinase Inhibitors; Genes, Reporter; Genome, Viral; Hepacivirus; Humans; Immunoprecipitation; Leupeptins; Lipids; Liver; Liver Neoplasms; Oleic Acid; Plasmids; Promoter Regions, Genetic; Protein Binding; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Time Factors; Transcription, Genetic; Transfection; Triglycerides; Viral Nonstructural Proteins | 2005 |
Octanoate reduces very low-density lipoprotein secretion by decreasing the synthesis of apolipoprotein B in primary cultures of chicken hepatocytes.
Fatty acids of varying lengths and saturation differentially affect plasma apolipoprotein B (apoB) levels. To identify the mechanisms underlying the effect of octanoate on very low-density lipoprotein (VLDL) secretion, chicken primary hepatocytes were incubated with either fatty acid-bovine serum albumin (BSA) complexes or BSA alone. Addition of octanoate to culture medium significantly reduced VLDL-triacylglycerol (TG), VLDL-cholesterol and apoB secretion from hepatocytes compared to both control cultures with BSA only and palmitate treatments, but did not modulate intracellular TG accumulation. However, no differences in cellular microsomal triglyceride transfer protein levels were observed in the cultures with saturated fatty acid. In pulse-chase studies, octanoate treatment resulted in reduced apoB-100 synthesis, in agreement with its promotion of secretion. This characteristic effect of octanoate was confirmed by addition of a protease inhibitor, N-acetyl-leucyl-leucyl-norleucinal (ALLN), to hepatocyte cultures. Analysis showed that the level of apoB mRNA was lower in cultures supplemented with octanoate than in the control cultures, but no significant changes were observed in the levels of apolipoprotein A-I, fatty acid synthase and 3-hydroxy-3-methylglutaryl-CoA reductase mRNA as a result of octanoate treatment. Time-course studies indicate that a 50% reduction in apoB mRNA levels requires 12 h of incubation with octanoate. We conclude that octanoate reduced VLDL secretion by the specific down-regulation of apoB gene expression and impairment of subsequent synthesis of apoB, not by the modulation of intracellular apoB degradation, which is known to be a major regulatory target of VLDL secretion of other fatty acids. Topics: Animals; Apolipoprotein A-I; Apolipoproteins B; Caprylates; Carrier Proteins; Cells, Cultured; Chickens; Cholesterol, VLDL; Cysteine Proteinase Inhibitors; Down-Regulation; Fatty Acid Synthases; Hepatocytes; Hydroxymethylglutaryl CoA Reductases; Leupeptins; Lipoproteins, VLDL; Male; Palmitic Acid; Triglycerides | 2005 |
Involvement of calpain in melanogenesis of mouse B16 melanoma cells.
In the current study, the involvement of calpain, a cysteine proteinase in the regulation of melanogenesis was examined using mouse B16 melanoma cells. In response to alpha-melanocyte-stimulating hormone (a-MSH), B16 melanoma cells underwent differentiation characterized by increased melanin biosynthesis. The total calapain activity was decreased within 2 h following alpha-MSH-treatment, and restored to the initial level in 6-12 h. To further investigate the involvement of calpain in the regulation of melanogenesis, the effect of calpain inhibitors on alpha-MSH-induced melanogenesis was examined. Inhibition of calpain by either N-acetyl-Leu-Leu-norleucinal (ALLN) or calpastatin (CS) peptide blocked alpha-MSH-induced melanogenesis. The magnitude of inhibition of melanin biosynthesis was well correlated with a decrease in the activity of tyrosinase, a key regulatory enzyme in melanogenesis. Treatment of B16 cells with ALLN caused marked decrease in both tyrosinase protein and mRNA levels. These results indicate that calpain would be involved in the melanogenic signaling by modulating the expression of tyrosinase in mouse B16 melanoma cells. Topics: alpha-MSH; Animals; Blotting, Western; Calcium-Binding Proteins; Calpain; Cell Differentiation; Cysteine Proteinase Inhibitors; Electrophoresis, Polyacrylamide Gel; Hormones; Indolequinones; Leupeptins; Melanins; Melanocytes; Melanoma, Experimental; Mice; Monophenol Monooxygenase; RNA, Messenger; Tumor Cells, Cultured | 2005 |
Human ABCA7 supports apolipoprotein-mediated release of cellular cholesterol and phospholipid to generate high density lipoprotein.
Apolipoprotein-mediated release of cellular cholesterol and phospholipids was induced in HEK293 cells by expressing human ATP-binding cassette transporter A7 (ABCA7) and ABC transporter A1 (ABCA1) proteins, whether transient or stable, to generate cholesterol-rich high density lipoprotein (HDL). Green fluorescent protein (GFP) attached at their C termini did not influence the lipid release reactions. Transfected ABCA7-GFP induced apolipoprotein-mediated assembly of cholesterol-containing HDL also in L929 cells, which otherwise generate only cholesterol-deficient HDL with their endogenous ABCA1. Time-dependent release of cholesterol and phospholipid by apolipoprotein A (apoA)-I was parallel both with ABCA1 and with ABCA7 when highly expressed in HEK293 cells, but dose-dependent profiles of lipid release on apoA-I and apoA-II were somewhat different between ABCA1 and ABCA7. Analyses of the stable clones with ABCA1-GFP (293/2c) and ABCA7-GFP (293/6c) by using the same vector indicated some differences in regulation of their activities by protein kinase modulators. Dibutyryl cyclic AMP increased ABCA1-GFP and the release of cholesterol and phospholipid in 293/2c but increased neither ABCA7-GFP nor the lipid release in 293/6c. Expression of ABCA1-GFP- and apoA-I-mediated lipid release were enhanced in parallel by phorbol 12-myristate 13-acetate (PMA) in 293/2c cells. In contrast, the same treatment of 293/6c increased ABCA7-GFP, but apoA-I-mediated lipid release was significantly suppressed. Despite these different responses to PMA, all of the effects of PMA were reversed by a specific protein kinase C inhibitor Gö6976, suggesting that the changes were in fact due to protein kinase C activation. A thiol protease inhibitor, N-acetyl-Leu-Leu-norleucinal, increased the protein levels of ABCA1-GFP in 293/2c and ABCA7-GFP in 293/6c, indicating their common degradation pathway. The data indicated that human ABCA7 would compensate the function of ABCA1 for release of cell cholesterol in a certain condition(s), but post-transcriptional regulation of their activity is different. Topics: Apolipoprotein A-I; Apolipoproteins; ATP Binding Cassette Transporter 1; ATP-Binding Cassette Transporters; Bucladesine; Carbazoles; Cell Line; Cholesterol; Cloning, Molecular; Cysteine Proteinase Inhibitors; DNA, Complementary; Enzyme Inhibitors; Humans; Indoles; Kinetics; Leupeptins; Lipoproteins, HDL; Phospholipids; Recombinant Proteins; Tetradecanoylphorbol Acetate; Transfection | 2004 |
Amino acid sequences within the beta1 domain of human apolipoprotein B can mediate rapid intracellular degradation.
Apolipoprotein B (apoB)-48 contains a region termed the beta1 domain that is predicted to be composed of extensive amphipathic beta-strands. Analysis of truncated apoB variants revealed that sequences between the carboxyl termini of apoB-37 and apoB-42 governed the secretion efficiency and intracellular stability of apoB. Although apoB-37, apoB-34, and apoB-29 were stable and secreted efficiently, apoB-42 and apoB-100 were secreted poorly and were degraded by an acetyl-leucyl-leucyl-norleucinal (ALLN)-sensitive pathway. Amino acid sequence analysis suggested that a segment between the carboxyl termini of apoB-38 and apoB-42 was 63% homologous to fatty acid binding proteins (FABPs), which contain orthogonal beta-sheets. To test the hypothesis that sequences from the beta1 domain are involved in apoB degradation, fusion proteins were created that contained apoB-29 linked to fragments derived from the beta1 domain of apoB or to liver FABP. Fusion proteins containing the beta1 domain segments apoB-34-42 or apoB-37-42 were degraded rapidly, whereas other fusion proteins were stable and secreted efficiently. Degradation was ALLN-sensitive, and the apoB-34-42 segment increased the association of the apoB protein with the cytosolic surface of the microsomal membrane. Our data suggest that the presence of specific sequences in the beta1 domain of human apoB increases degradation by promoting the cytosolic exposure of the protein, although not all regions of the beta1 domain are functionally equivalent. Topics: Amino Acid Sequence; Animals; Apolipoprotein B-100; Apolipoprotein B-48; Apolipoproteins B; Cell Line, Tumor; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Fatty Acids; Genetic Vectors; Humans; Leupeptins; Multienzyme Complexes; Proteasome Endopeptidase Complex; Protein Binding; Protein Structure, Secondary; Protein Structure, Tertiary; Rats; Recombinant Proteins; Sequence Alignment; Sequence Deletion | 2004 |
Both insulin resistance and diabetes in Psammomys obesus upregulate the hepatic machinery involved in intracellular VLDL assembly.
In the current study, we examined the mechanisms that regulate hepatic apolipoprotein B (apoB)-containing lipoprotein secretion in Psammomys obesus, a good animal model for the investigation of insulin resistance and diabetes.. When fed chow ad libitum, 22% maintained normoglycemia and normoinsulinemia (group A), 33% exhibited normoglycemia and appreciable hyperinsulinemia (group B), and 45% developed overt diabetes (group C). Body weight gain, plasma free fatty acid elevation, hypertriglyceridemia, and hypercholesterolemia characterized groups B and C. Triton WR-1339 injection, at fasting, resulted in higher plasma VLDL-triglyceride and VLDL-apoB accumulation in groups B and C, suggesting increased VLDL production by the liver. Pulse-chase labeling experiments in cultured hepatocytes with [35S]methionine revealed reduced intracellular degradation and enhanced secretion of newly synthesized apoB in groups B and C. Concomitant with the raised triglyceride and cholesterol contents in the livers of groups B and C, there was an increase in lipogenesis and in the activity of microsomal triglyceride transfer protein, monoacylglycerol acyltransferase, and diacylglycerol transferase. Pretreatment of hepatocytes with proteasomal inhibitors eliminated the differences in apoB secretion among groups A, B, and C.. Our data indicate that both insulin resistance and diabetes triggered the intracellular machinery involved in VLDL assembly and secretion. Topics: Acetylcysteine; Animals; Apolipoproteins B; Cells, Cultured; Cysteine Endopeptidases; Diabetes Mellitus; Disease Models, Animal; Gerbillinae; Hepatocytes; Hypercholesterolemia; Hyperinsulinism; Hypertriglyceridemia; Insulin; Insulin Resistance; Leupeptins; Lipoproteins, VLDL; Liver; Multienzyme Complexes; Protease Inhibitors; Proteasome Endopeptidase Complex | 2004 |
Induction of apoptosis by the proteasome inhibitor MG132 in human HCC cells: Possible correlation with specific caspase-dependent cleavage of beta-catenin and inhibition of beta-catenin-mediated transactivation.
Proteasome inhibitors, like MG132, can exert cell growth inhibitory and apoptotic effects in different tumor types. The apoptotic mechanism of these compounds involves the activation of the effector caspases. beta-catenin, also an oncogene, represents one of the substrates of these proteases, but the consequences of its cleavage are poorly understood. We investigated its function during apoptosis induced by MG132 in three hepatocellular carcinoma (HCC) cell lines, endowed (HepG2 and HuH-6) or not (HA22T/VGH) with activating mutations of beta-catenin. Induction of apoptosis was associated with cell growth inhibition, accumulation of the cells at the G(2)/M phases of the cell cycle, as well as with fragmentation of beta-catenin (but not of alpha- or gamma-catenin) in all the cell lines. The cleavage of beta-catenin was inhibited by the caspase inhibitors Z-VAD-fmk and Z-DEVD-fmk. Fragmented beta-catenin was found in the nuclei of the treated cells. Analyses through the reporter plasmid pTOPflash showed that MG132 significantly reduces Tcf transcriptional activity in the cells. This was associated with a decrease in the mRNA expression of survivin and c-myc, which are target genes of the APC/beta-catenin/Tcf signaling. Nevertheless, Z-VAD-fmk or Z-DEVD-fmk did not reverse the MG132 effects on Tcf transcriptional activity, suggesting that the compound may affect this activity also by other mechanisms. Overall, the present study supports the therapeutic potential of the proteasome inhibitors in HCC. Topics: alpha Catenin; Amino Acid Chloromethyl Ketones; Apoptosis; beta Catenin; Carcinoma, Hepatocellular; Caspase Inhibitors; Caspases; Cell Division; Cell Line, Tumor; Cell Nucleus; Cysteine Proteinase Inhibitors; Cytoskeletal Proteins; Cytosol; Desmoplakins; DNA-Binding Proteins; G2 Phase; gamma Catenin; Humans; Inhibitor of Apoptosis Proteins; Leupeptins; Lymphoid Enhancer-Binding Factor 1; Microtubule-Associated Proteins; Neoplasm Proteins; Oligopeptides; Peptide Fragments; Proto-Oncogene Proteins c-myc; RNA, Messenger; Survivin; Trans-Activators; Transcription Factors; Transcriptional Activation | 2004 |
Specific chemopreventive agents trigger proteasomal degradation of G1 cyclins: implications for combination therapy.
There is a need to identify cancer chemoprevention mechanisms. We reported previously that all-trans-retinoic acid (RA) prevented carcinogenic transformation of BEAS-2B immortalized human bronchial epithelial cells by causing G(1) arrest, permitting repair of genomic DNA damage. G(1) arrest was triggered by cyclin D1 proteolysis via ubiquitin-dependent degradation. This study investigated which chemopreventive agents activated this degradation program and whether cyclin E was also degraded.. This study examined whether: (a) cyclin E protein was affected by RA treatment; (b) cyclin degradation occurred in derived BEAS-2B-R1 cells that were partially resistant to RA; and (c) other candidate chemopreventive agents caused cyclin degradation.. RA treatment triggered degradation of cyclin E protein, and ALLN, a proteasomal inhibitor, inhibited this degradation. Induction of the retinoic acid receptor beta, growth suppression, and cyclin degradation were each inhibited in BEAS-2B-R1 cells. Transfection experiments in BEAS-2B cells indicated that RA treatment repressed expression of wild-type cyclin D1 and cyclin E, but ALLN inhibited this degradation. Mutation of threonine 286 stabilized transfected cyclin D1, and mutations of threonines 62 and 380 stabilized transfected cyclin E, despite RA treatment. Specific chemopreventive agents triggered cyclin degradation. Nonclassical retinoids (fenretinide and retinoid X receptor agonists) and a synthetic triterpenoid (2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid) each suppressed BEAS-2B growth and activated this degradation program. However, a vitamin D3 analog (RO-24-5531), a cyclooxygenase inhibitor (indomethacin), and a peroxisome proliferator-activated receptor gamma agonist (rosiglitazone) each suppressed BEAS-2B growth, but did not cause cyclin degradation. BEAS-2B-R1 cells remained responsive to nonclassical retinoids and to 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid.. Specific chemopreventive agents activate cyclin proteolysis. Yet, broad resistance did not occur after acquired resistance to a single agent. This provides a therapeutic rationale for combination chemoprevention with agents activating non-cross-resistant pathways. Topics: Anticarcinogenic Agents; Antineoplastic Combined Chemotherapy Protocols; Bronchi; Cell Culture Techniques; Cell Division; Cell Line; Cyclin D1; Cyclin E; Cyclin G; Cyclin G1; Cyclins; DNA Damage; Dose-Response Relationship, Drug; Epithelial Cells; G1 Phase; Humans; Immunoblotting; Leupeptins; Mutation; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Retinoids; Reverse Transcriptase Polymerase Chain Reaction; Threonine; Transcription, Genetic; Transfection; Tretinoin | 2004 |
Transcriptional repression of E2F gene by proteasome inhibitors in human osteosarcoma cells.
E2F family of transcription factors regulates the transcription of genes required for DNA synthesis. E2F is itself controlled by a series of transcriptional and post-transcriptional pathways. Here we provide evidence that proteasome inhibitor-mediated E2F1 gene down-regulation is regulated by transcriptional events. Using the proteasome-specific inhibitors, MG132 and lactacystin, we show that the p53, the cdk inhibitors p21 and p27, and cyclin A are degraded by the ubiquitin-proteasome pathway in human osteosarcoma cells. Interestingly, the expression levels of E2F1 and E2F2 are down-regulated by proteasome inhibitors. E2F promoter and RT-PCR assay clearly demonstrated that proteasome inhibitors could reduce E2F transcriptional activation. However, MG132-induced repression of E2F1 and E2F2 is not associated with ROS generation. Topics: Acetylcysteine; Antioxidants; Cell Cycle Proteins; Cell Line, Tumor; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Down-Regulation; E2F Transcription Factors; E2F1 Transcription Factor; E2F2 Transcription Factor; Glutathione; Humans; Leupeptins; Multienzyme Complexes; Osteosarcoma; Transcription Factors; Transcription, Genetic; Transfection | 2004 |
Inhibitors of cysteine cathepsin and calpain do not prevent ultraviolet-B-induced apoptosis in human keratinocytes and HeLa cells.
Caspases, members of the cysteine protease family, execute UVB-induced apoptosis in several cell lines and keratinocytes. Several researchers investigating UVB-induced apoptosis have demonstrated a dose-dependent protective effect of the synthetic peptide caspase inhibitor zVAD-fmk. However, zVAD-fmk displays a dose-dependent protective effect against UVB-induced apoptosis, even at doses higher than those required to block all known proapoptotic caspases. In addition, it is known that zVAD-fmk also inhibits other cysteine proteases including cathepsins and calpains, and these proteases have recently been demonstrated to play a role in the execution of programmed cell death induced by other stimuli, e.g. TNF-alpha. The purpose of the present study was therefore to investigate whether inhibitors of cysteine cathepsins and calpains could prevent UVB-induced apoptosis in HeLa cells and keratinocytes. This was done by investigating the effect of the irreversible cysteine protease inhibitor zFA-fmk, the cathepsin B inhibitor CA-074-Me and the calpain inhibitor ALLN on the viability of UVB-irradiated human keratinocytes and HeLa cells. At concentrations of 10 microM and above zVAD-fmk conferred partial dose-dependent protection against UVB-induced apoptosis in HeLa cells and keratinocytes. Moreover, caspase-3 activity was completely blocked at zVAD-fmk concentrations of 1 microM in HeLa cells. This indicates that caspase-independent mechanisms could be involved in UVB-induced apoptosis. However, the protease inhibitors zFA-fmk, CA-074-Me and ALLN all failed to prevent UVB-induced apoptosis in HeLa cells and keratinocytes. In conclusion, the protective effect of zVAD-fmk at high concentrations indicates that other proteases than caspases are active in the execution of UVB-induced apoptosis but further studies are needed to identify these proteases. Topics: Amino Acid Chloromethyl Ketones; Apoptosis; Calpain; Caspase Inhibitors; Cathepsins; Cells, Cultured; Cysteine Proteinase Inhibitors; Dipeptides; HeLa Cells; Humans; Keratinocytes; Ketones; Leupeptins; Phospholipases A; Ultraviolet Rays | 2004 |
Phosphorylation of the histone deacetylase 7 modulates its stability and association with 14-3-3 proteins.
Class II histone deacetylases (HDACs) play a role in myogenesis and inhibit transcriptional activation by myocyte enhancer factors 2. A distinct feature of class II HDACs is their ability to shuttle between the nucleus and the cytoplasm in a cell type- and signal-dependent manner. We demonstrate here that treatment with the 26 S proteosome inhibitors, MG132 and ALLN, leads to detection of ubiquitinated HDAC7 and causes accumulation of cytoplasmic HDAC7. We also show that treatment with calyculin A, a protein phosphatase inhibitor, leads to a marked increase of HDAC7 but not HDAC5. The increase in HDAC7 is accompanied by enhanced interaction between 14-3-3 proteins and HDAC7. HDAC7 mutations that prevent the interaction with 14-3-3 proteins also block calyculin A-mediated stabilization. Expression of constitutively active calcium/calmodulin-dependent kinase I stabilizes HDAC7 and causes an increased association between HDAC7 and 14-3-3. Together, our results suggest that calcium/calmodulin-dependent kinase I-mediated phosphorylation of HDAC7 acts, in part, to promote association of HDAC7 with 14-3-3 and stabilizes HDAC7. Topics: 14-3-3 Proteins; Cell Line; Cell Nucleus; Cysteine Proteinase Inhibitors; Cytoplasm; Enzyme Inhibitors; Histone Deacetylases; Humans; Leupeptins; Marine Toxins; Models, Biological; Mutation; Oxazoles; Peptide Hydrolases; Phosphorylation; Plasmids; Proteasome Endopeptidase Complex; Protein Binding; Protein Biosynthesis; Time Factors; Transcription, Genetic; Transcriptional Activation; Transfection; Tyrosine 3-Monooxygenase; Up-Regulation | 2004 |
Calpain inhibitors improve myocardial dysfunction and inflammation induced by endotoxin in rats.
Excessive activation of calpains has been implicated in the pathophysiology of inflammation, trauma, and ischemia reperfusion injury. Here, we investigated the effects of calpain inhibition on myocardial dysfunction and inflammation induced by endotoxin in rats. Rats were treated i.v. with endotoxin (10 mg/kg) or endotoxin plus calpain inhibitors and were then prepared after 4 h for myocardial contractility assessment, detection of endothelium leukocyte interactions, and plasma TNF-alpha, nitrite/nitrate, and endocan levels. Compared with vehicle-treated rats, hearts from endotoxin-treated rats had reduced systolic performance that was partially prevented by calpain inhibitors, i.e., acetyl-leucyl-leucyl-arginal (leupeptin), carbobenzoxy-valyl-phenylalanial (calpain inhibitor III), and N-acetyl-leucinyl-leucinyl-norleucinal (ALLN). Leupeptin and calpain inhibitor III reduced plasma TNF-alpha levels in endotoxin-treated rats. ALLN reduced plasma TNF-alpha and nitrite/nitrate levels in endotoxin-treated rats. Endotoxin treatment increased mesenteric venule leukocyte rolling (10 +/- 3 leukocytes/min vs. 44 +/- 10 leukocytes/min; P < 0.01) and adhesion (2 +/- 2 leukocytes/min vs. 15 +/- 3 leukocytes/min; P < 0.01), which was reduced by calpain inhibitors. Attenuation of leukocyte endothelium interactions observed in calpain inhibitor-treated rats with sepsis was associated with increases in plasma anti-adhesion molecule endocan. In conclusion, calpain inhibitors improved endotoxin-induced cardiac dysfunction, which may be attributed to the modulation of endothelium leukocyte interactions in the inflamed vasculature. Topics: Animals; Cardiomyopathies; Endotoxins; Glycoproteins; Heart; In Vitro Techniques; Inflammation; Injections, Intravenous; Leukocyte Rolling; Leupeptins; Male; Myocardial Contraction; Myocardium; Nitrates; Nitrites; Rats; Rats, Sprague-Dawley; Shock, Septic; Tumor Necrosis Factor-alpha | 2004 |
Endoplasmic reticulum stress triggers an acute proteasome-dependent degradation of ATF6.
ATF6, a 670 amino acid endoplasmic reticulum (ER) transmembrane glycoprotein with the electrophoretic mobility of a 90 kDa protein, is a key transcriptional activator of the unfolded protein response (UPR) that allows mammalian cells to maintain cellular homeostasis when the cells are subjected to a variety of environmental and physiological stress. Previous studies have established that ATF6 is a short-lived protein, the activation of which involves relocation from the ER to the Golgi where it is cleaved by the S1P/S2P protease system to generate a nuclear form that acts as a transcriptional activator for ER-stress inducible target genes such as Grp78/BiP. We report here that in addition to this process, ER-stress mediated by thapsigargin triggers an acute proteasomal degradation of the pre-existing pool of p90ATF6 independent of S1P/S2P cleavage. We showed that ATF6 is a direct target of proteasome-ubiquitin pathway, and this process can be suppressed by proteasome inhibitors, ALLN and MG115. We further observed that in non-stressed cells, p90ATF6 can be stabilized by MG115 but not ALLN and that treatment of cells with MG115 results in Grp78 induction in the absence of ER stress. These studies suggest that ER-stress induced acute, transit degradation of p90ATF6 could represent a novel cellular defense mechanism to prevent premature cell death resulting from p90ATF6 activation. Further, inhibition of proteasome activity can result in chaperone protein gene induction through stabilization of p90ATF6 as well as accumulation of malfolded proteins. Topics: Activating Transcription Factor 6; Animals; CHO Cells; Cricetinae; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Endoplasmic Reticulum; Endoplasmic Reticulum Chaperone BiP; Enzyme Inhibitors; Heat-Shock Proteins; Humans; Kidney; Leupeptins; Mice; Molecular Chaperones; NF-kappa B; NIH 3T3 Cells; Protease Inhibitors; Proteasome Endopeptidase Complex; Thapsigargin; Transcription Factors | 2004 |
Ubiquitin-proteasome pathway modulates mouse oocyte meiotic maturation and fertilization via regulation of MAPK cascade and cyclin B1 degradation.
Degradation of proteins mediated by ubiquitin-proteasome pathway (UPP) plays important roles in the regulation of eukaryotic cell cycle. In this study, the functional roles and regulatory mechanisms of UPP in mouse oocyte meiotic maturation, fertilization, and early embryonic cleavage were studied by drug-treatment, Western blot, antibody microinjection, and confocal microscopy. The meiotic resumption of both cumulus-enclosed oocytes and denuded oocytes was stimulated by two potent, reversible, and cell-permeable proteasome inhibitors, ALLN and MG-132. The metaphase I spindle assembly was prevented, and the distribution of ubiquitin, cyclin B1, and polo-like kinase 1 (Plk1) was also distorted. When UPP was inhibited, mitogen-activated protein kinase (MAPK)/p90rsk phosphorylation was not affected, but the cyclin B1 degradation that occurs during normal metaphase-anaphase transition was not observed. During oocyte activation, the emission of second polar body (PB2) and the pronuclear formation were inhibited by ALLN or MG-132. In oocytes microinjected with ubiquitin antibodies, PB2 emission and pronuclear formation were also inhibited after in vitro fertilization. The expression of cyclin B1 and the phosphorylation of MAPK/p90rsk could still be detected in ALLN or MG-132-treated oocytes even at 8 h after parthenogenetic activation or insemination, which may account for the inhibition of PB2 emission and pronuclear formation. We also for the first time investigated the subcellular localization of ubiquitin protein at different stages of oocyte and early embryo development. Ubiquitin protein was accumulated in the germinal vesicle (GV), the region between the separating homologous chromosomes, the midbody, the pronuclei, and the region between the separating sister chromatids. In conclusion, our results suggest that the UPP plays important roles in oocyte meiosis resumption, spindle assembly, polar body emission, and pronuclear formation, probably by regulating cyclin B1 degradation and MAPK/p90rsk phosphorylation. Topics: Animals; Antibodies; Cell Cycle Proteins; Cyclin B; Cyclin B1; Cysteine Proteinase Inhibitors; Female; Fertilization in Vitro; Leupeptins; MAP Kinase Signaling System; Meiosis; Mice; Oocytes; Parthenogenesis; Polo-Like Kinase 1; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Kinases; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Subcellular Fractions; Ubiquitin | 2004 |
Inhibition of calpain is a component of nitric oxide-induced down-regulation of human mast cell adhesion.
Nitric oxide is an important messenger that regulates mast cell activity by modifications to gene expression and intracellular pathways associated with exocytosis and adhesion. Integrin interactions with extracellular matrix components modulate an array of cell activities, including mediator production and secretion. To investigate the molecular mechanisms underlying NO regulation of mast cell function, we studied its effects on adhesion of a human mast cell line (HMC-1) to fibronectin (FN). The NO donors S-nitrosoglutathione and S-nitroso-N-acetylpenicillamine strongly down-regulated the adhesion of HMC-1 to FN. Inhibitors of soluble guanylate cyclase and protein kinase G did not alter the response of cells to NO. A peroxynitrite scavenger did not affect modulation of adhesion by NO, nor could the effect of NO be mimicked by the peroxynitrite-producing compound 3-morpholinosydnonimine. NO donors inhibited the cysteine protease, calpain, while calpain inhibitors mimicked the effect of NO and led to a decrease in the ability of HMC-1 cells to adhere to FN. Thus, NO is an effective down-regulator of human mast cell adhesion. The mechanism for this action does not involve peroxynitrite or activation of soluble guanylate cyclase. Instead, a portion of NO-induced down-regulation of adhesion may be attributed to inhibition of the cysteine protease, calpain, an enzyme that has been associated with control of integrin activation in other cell types. The inhibition of calpain is most likely mediated via nitrosylation of its active site thiol group. Calpain may represent a novel therapeutic target for the regulation of mast cell activity in inflammatory disorders. Topics: Calpain; Cell Adhesion; Cyclic GMP; Dipeptides; Down-Regulation; Enzyme Activation; Enzyme Inhibitors; Fibronectins; Humans; Leupeptins; Mast Cells; Nitric Oxide; Nitric Oxide Donors; Peroxynitrous Acid; Protein Binding; S-Nitroso-N-Acetylpenicillamine; S-Nitrosoglutathione; Tumor Cells, Cultured | 2003 |
TNF-related apoptosis-inducing ligand (TRAIL) frequently induces apoptosis in Philadelphia chromosome-positive leukemia cells.
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL) have been implicated in antitumor immunity and therapy. In the present study, we investigated the sensitivity of Philadelphia chromosome (Ph1)-positive leukemia cell lines to TRAIL- or FasL-induced cell death to explore the possible contribution of these molecules to immunotherapy against Ph1-positive leukemias. TRAIL, but not FasL, effectively induced apoptotic cell death in most of 5 chronic myelogenous leukemia-derived and 7 acute leukemia-derived Ph1-positive cell lines. The sensitivity to TRAIL was correlated with cell-surface expression of death-inducing receptors DR4 and/or DR5. The TRAIL-induced cell death was caspase-dependent and enhanced by nuclear factor kappa B inhibitors. Moreover, primary leukemia cells from Ph1-positive acute lymphoblastic leukemia patients were also sensitive to TRAIL, but not to FasL, depending on DR4/DR5 expression. Fas-associated death domain protein (FADD) and caspase-8, components of death-inducing signaling complex (DISC), as well as FLIP (FLICE [Fas-associating protein with death domain-like interleukin-1-converting enzyme]/caspase-8 inhibitory protein), a negative regulator of caspase-8, were expressed ubiquitously in Ph1-positive leukemia cell lines irrespective of their differential sensitivities to TRAIL and FasL. Notably, TRAIL could induce cell death in the Ph1-positive leukemia cell lines that were refractory to a BCR-ABL-specific tyrosine kinase inhibitor imatinib mesylate (STI571; Novartis Pharma, Basel, Switzerland). These results suggested the potential utility of recombinant TRAIL as a novel therapeutic agent and the possible contribution of endogenously expressed TRAIL to immunotherapy against Ph1-positive leukemias. Topics: Amino Acid Chloromethyl Ketones; Apoptosis; Apoptosis Regulatory Proteins; Arabidopsis Proteins; Benzamides; Carrier Proteins; CASP8 and FADD-Like Apoptosis Regulating Protein; Caspase 1; Death Domain Receptor Signaling Adaptor Proteins; Drug Resistance, Neoplasm; Drug Screening Assays, Antitumor; Enzyme Inhibitors; Fas Ligand Protein; Fatty Acid Desaturases; Fusion Proteins, bcr-abl; Humans; Imatinib Mesylate; Intracellular Signaling Peptides and Proteins; Leukemia, Myelogenous, Chronic, BCR-ABL Positive; Leupeptins; Membrane Glycoproteins; Neoplasm Proteins; Neoplastic Stem Cells; NF-kappa B; Peptides; Piperazines; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Pyrimidines; Receptors, TNF-Related Apoptosis-Inducing Ligand; Receptors, Tumor Necrosis Factor; Recombinant Proteins; TNF-Related Apoptosis-Inducing Ligand; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha | 2003 |
Extracellular matrix enhances heregulin-dependent BRCA1 phosphorylation and suppresses BRCA1 expression through its C terminus.
Germ line mutations in the breast cancer susceptibility gene BRCA1 account for the increased risk of early onset of familial breast cancer, whereas overexpression of the ErbB family of receptor tyrosine kinases has been linked to the development of nonfamilial or sporadic breast cancer. To analyze whether there is a link between these two regulatory molecules, we studied the effects of ErbB-2 activation by heregulin (HRG) on BRCA1 function. It was previously demonstrated that HRG induced the phosphorylation of BRCA1, which was mediated by the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Since altered interaction between cells and the surrounding extracellular matrix (ECM) is a common feature in a variety of tumors and since ECM modulates intracellular signaling, we hypothesized that ECM may affect the expression and HRG-dependent phosphorylation of BRCA1. Following stimulation by HRG, a strong increase in [(3)H]thymidine incorporation was observed in human T47D breast cancer cells seeded on plastic (PL). When T47D cells were seeded on laminin (LAM) or Matrigel, HRG induced a significantly higher proliferation than it did in cells seeded on PL. T47D cells seeded on poly-L-lysine had an abrogated mitogenic response, indicating the involvement of integrins in this process. HRG treatment induced a transient phosphorylation of BRCA1 that was enhanced in T47D cells grown on LAM. LAM-enhanced BRCA1 phosphorylation was mediated through alpha(6) integrin upon HRG stimulation. Accordingly, T47D cells grown on LAM had the greatest increase in ErbB-2 activation, PI3K activity, and phosphorylation of Akt. A similar pattern of BRCA1 mRNA expression was observed when T47D cells were seeded on PL, LAM, or COL4. There was a significant decrease in the steady state of the BRCA1 mRNA level on both the LAM and COL4 matrices compared to that for cells seeded on PL. In addition, HRG stimulation caused a significant decrease in BRCA1 mRNA expression that was dependent on protein synthesis. Pretreatment with both the calpain inhibitor ALLN (N-acetyl-Leu-Leu-norleucinal) and the proteosome inhibitor lactacystin inhibited the HRG-induced down-regulation of BRCA1 mRNA expression. Likewise, there was a strong decrease in the protein level of BRCA1 in T47D cells 4 h after treatment with HRG compared to its level in control nontreated T47D cells. Pretreatment with the proteosome inhibitors ALLN, lactacystin, and PSI [N-benzyloxycarbonyl-Ile-Glu-(O-t-butyl)-Ala-leucinal] inhibited Topics: Acetylcysteine; Blotting, Northern; Blotting, Western; BRCA1 Protein; Cell Cycle; Cell Division; Cell Survival; Collagen; Cyclin-Dependent Kinase 4; Cyclin-Dependent Kinases; Cysteine Proteinase Inhibitors; Down-Regulation; Drug Combinations; Enzyme Inhibitors; Extracellular Matrix; Genetic Vectors; Germ-Line Mutation; Humans; Immunoblotting; Laminin; Leupeptins; MAP Kinase Signaling System; Neuregulin-1; Phenotype; Phosphatidylinositol 3-Kinases; Phosphorylation; Polylysine; Precipitin Tests; Protein Binding; Protein Structure, Tertiary; Proteoglycans; Proto-Oncogene Proteins; Receptor, ErbB-2; RNA; RNA, Messenger; Signal Transduction; Time Factors; Transfection; Tumor Cells, Cultured | 2003 |
Dictyostelium differentiation-inducing factor-3 activates glycogen synthase kinase-3beta and degrades cyclin D1 in mammalian cells.
In search of chemical substances applicable for the treatment of cancer and other proliferative disorders, we studied the signal transduction of Dictyostelium differentiation-inducing factors (DIFs) in mammalian cells mainly using HeLa cells. Although DIF-1 and DIF-3 both strongly inhibited cell proliferation by inducing G(0)/G(1) arrest, DIF-3 was more effective than DIF-1. DIF-3 suppressed cyclin D1 expression at both mRNA and protein levels, whereas the overexpression of cyclin D1 overrode DIF-3-induced cell cycle arrest. The DIF-3-induced decrease in the amount of cyclin D1 protein preceded the reduction in the level of cyclin D1 mRNA. The decrease in cyclin D1 protein seemed to be caused by accelerated proteolysis, since it was abrogated by N-acetyl-Leu-Leu-norleucinal, a proteasome inhibitor. DIF-3-induced degradation of cyclin D1 was also prevented by treatment with lithium chloride, an inhibitor of glycogen synthase kinase-3beta (GSK-3beta), suggesting that DIF-3 induced cyclin D1 proteolysis through the activation of GSK-3beta. Indeed, DIF-3 dephosphorylated Ser(9) and phosphorylated tyrosine on GSK-3beta, and it stimulated GSK-3beta activity in an in vitro kinase assay. Moreover, DIF-3 was revealed to induce the nuclear translocation of GSK-3beta by immunofluorescent microscopy and immunoblotting of subcellular protein fractions. These results suggested that DIF-3 activates GSK-3beta to accelerate the proteolysis of cyclin D1 and that this mechanism is involved in the DIF-3-induced G(0)/G(1) arrest in mammalian cells. Topics: Animals; Cell Cycle; Cell Division; Cell Line; Cells, Cultured; Cyclin D1; Cyclin D2; Cyclin D3; Cyclins; Cysteine Endopeptidases; Dictyostelium; Electrophoresis, Polyacrylamide Gel; Endothelium, Vascular; G1 Phase; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; HeLa Cells; Hexanones; Humans; Leupeptins; Lithium Chloride; Microscopy, Fluorescence; Multienzyme Complexes; Phosphorylation; Proteasome Endopeptidase Complex; Resting Phase, Cell Cycle; Reverse Transcriptase Polymerase Chain Reaction; RNA; RNA, Messenger; Signal Transduction; Time Factors; Transfection; Umbilical Veins | 2003 |
I kappa B overexpression in cardiomyocytes prevents NF-kappa B translocation and provides cardioprotection in trauma.
This study examined the effects of either IkappaBalpha overexpression (transgenic mice) or N-acetyl-leucinyl-leucinyl-norleucinal (ALLN) administration (proteosome inhibitor in wild-type mice) on cardiomyocyte secretion of tumor necrosis factor-alpha (TNF-alpha) and on cardiac performance after burn trauma. Transgenic mice were divided into four experimental groups. IkappaBalpha overexpressing mice were given a third-degree scald burn over 40% of the total body surface area or wild-type littermates were given either a scald or sham burn to provide appropriate controls. Pharmacological studies included ALLN (20 mg/kg) administration in either burned wild-type mice or wild-type shams. Burn trauma in wild-type mice promoted nuclear factor-kappaB (NF-kappaB) nuclear translocation, cardiomyocyte secretion of TNF-alpha, and impaired cardiac performance. IkappaBalpha overexpression or ALLN treatment of burn trauma prevented NF-kappaB activation in cardiac tissue, prevented cardiomyocyte secretion of TNF-alpha, and ablated burn-mediated cardiac contractile dysfunction. These data suggest that NF-kappaB activation and inflammatory cytokine secretion play a significant role in postburn myocardial abnormalities. Topics: Animals; Burns; Cysteine Proteinase Inhibitors; Cytokines; Female; I-kappa B Proteins; Leupeptins; Male; Mice; Mice, Inbred C57BL; Mice, Transgenic; Myocardial Contraction; Myocytes, Cardiac; NF-kappa B; NF-KappaB Inhibitor alpha; Protein Transport; Tumor Necrosis Factor-alpha; Wounds and Injuries | 2003 |
Transmembrane lipid transfer is crucial for providing neutral lipids during very low density lipoprotein assembly in endoplasmic reticulum.
Very low density lipoprotein (VLDL), a large particle containing apolipoprotein B (apoB) and large amounts of neutral lipids, is formed in the luminal space within the endoplasmic reticulum (ER) of hepatic cells. The assembly mechanism of VLDL particles is a tightly regulated process where apoB, associated with an insufficient amount of lipids, is selectively degraded intracellularly. In this study we found that treatment of HuH-7 human hepatoma cells with verapamil inhibited secretion of apoB-containing lipoprotein particles through increasing degradation of apoB. Addition of N-acetylleucyl-leucyl-norleucinal, an inhibitor of proteasome and other cysteinyl proteases that are responsible for apoB degradation, restored apoB recovery from verapamil-treated cells. De novo synthesis of lipids from [14C]acetate was increased in the presence of verapamil, suggesting that verapamil decreases lipid availability for apoB thus leading to the secretion of apoB-containing lipoprotein. We prepared cytosolic fractions from cells preincubated with [14C]acetate and used as a donor of radioactive lipids. When this cytosolic fraction was incubated with microsomes isolated separately, radioactive triglyceride (TG) accumulated in the luminal space of the microsomes. The transfer of radioactive TG from the cytosolic fraction to the microsomal lumen was inhibited in the presence of verapamil, suggesting that there is a verapamil-sensitive mechanism for TG transfer across ER membranes that is involved in formation of apoB-containing lipoprotein particles in ER. Verapamil showed no inhibitory effect on microsomal TG transfer protein, a well known lipid transfer protein in ER. We propose from these results that there is novel machinery for transmembrane movement of neutral lipids, which is involved in providing TG for apoB during VLDL assembly in ER. Topics: Acetates; Albumins; Apolipoproteins B; ATP Binding Cassette Transporter, Subfamily B, Member 1; Calcium Channel Blockers; Calcium Channels; Cell Membrane; Cholesterol; Culture Media, Conditioned; Cysteine Proteinase Inhibitors; Cytosol; Dose-Response Relationship, Drug; Endoplasmic Reticulum; Humans; Leupeptins; Lipid Metabolism; Lipoproteins, VLDL; Microsomes; Time Factors; Triglycerides; Tumor Cells, Cultured; Verapamil | 2003 |
Synergistic induction of apoptosis of rheumatoid arthritis synovial cells by H(2)O(2) and N-acetyl-leucyl-leucyl-norleucinal.
The effects of proteolysis inhibitors on hydrogen peroxide (H(2)O(2))-induced apoptosis were examined in cultured human synovial cells of rheumatoid arthritis (RA) patients. RA synovial cells were resistant to apoptosis induced by H(2)O(2). In the presence of 100 microM N-acetyl-leucyl-leucyl-norleucinal (ALLN, known as calpain inhibitor 1 and also a proteasome inhibitor), but not N-acetyl-leucyl-leucyl-methioninal (ALLM), apoptotic cell death was elicited by 400 microM H(2)O(2) at a concentration that alone never induced cell death. ALLN induced the expression of tumor suppressor p53 protein and p21(WAF-1) protein, probably through inhibition of proteasome. H(2)O(2) further potentiated ALLN-induced p53 expression. H(2)O(2) appeared to activate c-Jun N-terminal kinase (JNK) as well as extracellular signal-regulated kinase (ERK) and AKT. After administration of H(2)O(2) and p53 induction by ALLN, we found that either one alone is insufficient to induce apoptosis of RA synovial cells but their combination synergistically does so. These results suggest that induction of p53 by ALLN may be potentially important for triggering H(2)O(2)-induced apoptosis processes in RA synovial cells. Topics: Apoptosis; Arthritis, Rheumatoid; Calpain; Cells, Cultured; Cysteine Proteinase Inhibitors; Drug Synergism; Humans; Hydrogen Peroxide; Leupeptins; Synovial Membrane | 2003 |
Food vacuole plasmepsins are processed at a conserved site by an acidic convertase activity in Plasmodium falciparum.
Intraerythrocytic Plasmodium falciparum digests vast amounts of hemoglobin within an acidic food vacuole (FV). Four homologous aspartic proteases participate in hemoglobin degradation within the FV. Plasmepsin (PM) I and II are thought to initiate degradation of the native hemoglobin molecule. PM IV and histo-aspartic protease (HAP) act on denatured globin further downstream in the pathway. PM I and II have been shown to be synthesized as zymogens and activated by proteolytic removal of a propiece. In this study, we have determined that the proteolytic processing of FV plasmepsins occurs immediately after a conserved Leu-Gly dipeptidyl motif with uniform kinetics and pH and inhibitor sensitivities. We have developed a cell-free in vitro processing assay that generates correctly processed plasmepsins. Our data suggest that proplasmepsin processing is not autocatalytic, but rather is mediated by a separate processing enzyme. This convertase requires acidic conditions and is blocked only by the calpain inhibitors, suggesting that it may be an atypical calpain-like protease. Topics: Amino Acid Motifs; Amino Acid Sequence; Animals; Aspartic Acid Endopeptidases; Calpain; Cell-Free System; Endopeptidases; Hydrogen-Ion Concentration; Kinetics; Leupeptins; Molecular Sequence Data; Plasmodium falciparum; Protease Inhibitors; Protein Processing, Post-Translational; Protozoan Proteins; Vacuoles | 2003 |
Radiosensitization by inhibition of IkappaB-alpha phosphorylation in human glioma cells.
To assess the role of nuclear factor kappaB (NFKB) in cellular radiosensitivity, three different IkappaB-alpha (also known as NFKBIA) expression plasmids, i.e., S-IkappaB (mutations at (32, 36)Ser), Y-IkappaB (a mutation at (42)Tyr), and SY-IkappaB, were constructed and introduced into human brain tumor M054 cells. The clones were named as M054-S8, M054-Y2 and M054-SY4, respectively. Compared to the parental cell line, M054-S8 and M054-Y2 cells were more sensitive to X rays while M054-SY4 cells exhibited the greatest sensitivity. After treatment with N-acetyl-Leu-Leu-norleucinal, a proteasome inhibitor, the X-ray sensitivity of M054-S8 and M054-SY4 cells did not change, while that of M054-Y2 cells and the parental cells was enhanced. An increase in X-ray sensitivity accompanied by a decrease in translocation of NFKB to the nucleus in parental cells was observed after treatment with pervanadate, an inhibitor of tyrosine phosphatase, as well as in M054-S8 and M054-SY4 cells. Repair of potentially lethal damage (PLD) was observed in the parental cells but not in the clones. Four hours after irradiation (8 Gy), the expression of TP53 and phospho-p53 ((15)Ser) was induced in the parental cells but not in M054-S8, M054-Y2 or M054-SY4 cells. Our data suggest that inhibition of IkappaB-alpha phosphorylation at serine or tyrosine acts independently in sensitizing cells to X rays. NFKB may play a role in determining radiosensitivity and PLD repair in malignant glioma cells; TP53 may also be involved. Topics: Cell Survival; Cloning, Molecular; DNA Repair; Gene Expression Regulation, Neoplastic; Glioma; Humans; Leupeptins; Mutagenesis, Site-Directed; NF-kappa B; Phosphorylation; Radiation Tolerance; Transfection; Tumor Cells, Cultured; Vanadates | 2003 |
Interleukin-10 activation of the interleukin-10E1 pathway and tissue inhibitor of metalloproteinase-1 expression is enhanced by proteasome inhibitors in primary prostate tumor lines.
The interleukin-10 (IL-10) activation of Janus kinase (JAK) family members (JAK1/TYK2) and IL-10E1 is subsequently inactivated by approximately 3-4 h in primary prostate tumor lines. We examined the effect of proteasome inhibition on IL-10 activation of the IL-10E1 pathway following stimulation of HPCA-10a cells. Treatment of HPCA-10a cells with the proteasome inhibitor, N-acetyl-L-leucinyl-L-leucinyl-norleucinal (LLnL), led to stable tyrosine phosphorylation of the IL-10 receptor and IL-10E1 following stimulation. Further investigation showed that these stable phosphorylation events were the result of prolonged activation of JAK1 and TYK2 plus IL-10E1. IL-10E1 signaling normally induced the expression of tissue inhibitor of metalloproteinase-1 (TIMP-1) and LLnL treatment of the HPCA-10a and HPCA-10c cells significantly enhanced IL-10 induction of TIMP-1 levels to block tumor cell invasion in modified Boyden chamber invasion assays. These observations were confirmed using pharmacologic inhibitors by Western blot and ELISAs. In the presence of LLnL, stable phosphorylation of IL-10E1 and induction of TIMP-1 was abrogated if the tyrosine kinase inhibitor, staurosporine, was added. The effect of staurosporine on IL-10E1 phosphorylation and TIMP-1 could be overcome if the phosphatase inhibitor, vanadate, was also added, suggesting that phosphorylated IL-10E1 could be stabilized by phosphatase, but not by proteasome inhibition. These observations are consistent with the hypothesis that proteasome-mediated protein degradation can modulate the activity of the IL-10E1 pathway and TIMP-1 induction by regulating the deactivation of JAK1/TYK2. Topics: Antibodies; Cell Line, Tumor; Electrophoretic Mobility Shift Assay; Enzyme-Linked Immunosorbent Assay; Gene Expression Regulation; Humans; Interleukin-10; Leupeptins; Male; Phosphorylation; Phosphotyrosine; Prostatic Neoplasms; Protease Inhibitors; Proto-Oncogene Proteins c-rel; Signal Transduction; Staurosporine; Tissue Inhibitor of Metalloproteinase-1 | 2003 |
Ubiquitin-dependent and -independent proteasomal degradation of apoB associated with endoplasmic reticulum and Golgi apparatus, respectively, in HepG2 cells.
Studies in hepatocyte cultures indicate that apolipoprotein (apo) B-100 production is regulated largely by intracellular degradation and the proteasome pathway is a major mechanism for the degradation. In the present study, we have examined the detailed itinerary of apoB degradation through its secretory pathway in HepG2 cells. We found that ubiquitin-dependent proteasomal degradation of apoB largely occurred on the cytosolic surface of rough and smooth endoplasmic reticulum (ER) and that a small proportion of apoB was dislodged from the secretory organelles into the cytosolic compartment where it underwent ubiquitination for proteasomal degradation. The transmembrane conformation of apoB persisted as the protein was transported through the Golgi apparatus. We further demonstrated that proteasomal degradation of apoB was associated the Golgi apparatus but Golgi-associated apoB was not ubiquitinated, indicating an ubiquitin-independent proteasomal degradation of apoB is associated with this organelle. We conclude that apoB undergoes proteasomal degradation while going through different compartments of the secretory pathway; further, ER-associated proteasomal degradation of apoB in the ER is ubiquitin-dependent whereas that occurring in the Golgi is ubiquitin-independent. Topics: Apolipoprotein B-100; Apolipoproteins B; Biological Transport; Calpain; Carcinoma, Hepatocellular; Cell Compartmentation; Cysteine Endopeptidases; Cytosol; Endoplasmic Reticulum; Enzyme Inhibitors; Fluorescent Antibody Technique; Golgi Apparatus; Humans; Leupeptins; Microsomes, Liver; Multienzyme Complexes; Proteasome Endopeptidase Complex; Tumor Cells, Cultured; Ubiquitin | 2003 |
Involvement of the proteasome in IL-1beta induced suppression of islets of Langerhans in the rat.
The cytokine IL-1beta suppresses rodent islets of Langerhans in vitro. Presently we used inhibitors of the proteasome to investigate if these compounds could counteract the suppressive effects of the cytokine. Thus, isolated rat islets were cultured and pre-treated with proteasome inhibitors and subsequently exposed for 48 h to 25 U/ml human IL-1beta. After this period functional tests were carried out. The rate of glucose oxidation (pmol/10 islets x 90 min) was suppressed by IL-1beta (115 +/- 17 vs. control 380 +/- 57). Pre-treatment with 10 microM of the proteasome inhibitor MG115 (N-carbobenzoxyl-leu-leu-norvalinal) and 100 microM of the calpain inhibitor norLEU (N-acetyl-leu-leu-norleucinal; known to affect proteasome activity) counteracted the suppressive effects (253 +/- 17 and 262 +/- 10 respectively). The calpain inhibitor alIMET (N-acetyl-leu-leu-methional) had no effect. MG115 (10 microM) and norLEU (100 microM) blocked nitric oxide formation induced by IL-1beta, while alIMET was without effect. We also investigated if IL-1beta could influence the expression of two inducible proteasome subunits, namely LMP2 and LMP7, and found that the cytokine increased the mRNA expression of the proteasome subunit LMP2 in islets, and that the proteasome inhibitor MG115 prevented this increase. In conclusion our study shows that IL-1beta increases the transcription of the proteasome subunit LMP2, and that the proteasome is involved in IL-1beta induced suppression of islet function. Moreover, the observation that inhibitors of the proteasome protect islets against IL-1beta induced inhibition of glucose metabolism, suggests that these compounds might be worthwile to explore in future therapies against the development of type 1 diabetes. Topics: Animals; Base Sequence; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA; DNA Primers; Electrophoretic Mobility Shift Assay; Glucose; Insulin; Insulin Secretion; Interleukin-1; Islets of Langerhans; Leupeptins; Male; Multienzyme Complexes; Nitrites; Proteasome Endopeptidase Complex; Rats; Rats, Sprague-Dawley; Reverse Transcriptase Polymerase Chain Reaction | 2003 |
Regulation of ubiquitous 6-phosphofructo-2-kinase by the ubiquitin-proteasome proteolytic pathway during myogenic C2C12 cell differentiation.
6-Phosphofructo-2-kinase catalyzes the synthesis and degradation of fructose 2,6-bisphosphate, activator of phosphofructokinase-1 and inhibitor of fructose 1,6-bisphosphatase. These properties confer to this bifunctional enzyme a key role in the control of glycolysis and gluconeogenesis. Several mammalian isozymes generated by alternative splicing from four genes, designated pfkfb1-4, have been identified. The results presented in this study demonstrate the expression of the pfkfb3 gene in C2C12 cells and its downregulation during myogenic cell differentiation. We also show that the decrease of ubiquitous 6-phosphofructo-2-kinase isozyme levels, product of pfkfb3 gene, is due to its enhanced degradation through the ubiquitin-proteasome proteolytic pathway. Topics: Animals; Cell Differentiation; Cells, Cultured; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Gene Expression Regulation, Enzymologic; Humans; Isoenzymes; Leupeptins; Multienzyme Complexes; Myoblasts; Phosphofructokinase-2; Proteasome Endopeptidase Complex; Proteins; Ubiquitin | 2003 |
Calpain facilitates GLUT4 vesicle translocation during insulin-stimulated glucose uptake in adipocytes.
Calpains are a family of non-lysosomal cysteine proteases. Recent studies have identified a member of the calpain family of proteases, calpain 10, as a putative diabetes-susceptibility gene that may be involved in the development of type 2 diabetes. Inhibition of calpain activity has been shown to reduce insulin-stimulated glucose uptake in isolated rat-muscle strips and adipocytes. In this report, we examine the mechanism by which calpain affects insulin-stimulated glucose uptake in 3T3-L1 adipocytes. Inhibition of calpain activity resulted in approx. a 60% decrease in insulin-stimulated glucose uptake. Furthermore, inhibition of calpain activity prevented the translocation of insulin-responsive glucose transporter 4 (GLUT4) vesicles to the plasma membrane, as demonstrated by fluorescent microscopy of whole cells and isolated plasma membranes; it did not, however, alter the total GLUT4 protein content. While inhibition of calpain did not affect the insulin-mediated proximal steps of the phosphoinositide 3-kinase pathway, it did prevent the insulin-stimulated cortical actin reorganization required for GLUT4 translocation. Specific inhibition of calpain 10 by antisense expression reduced insulin-stimulated GLUT4 translocation and actin reorganization. Based on these findings, we propose a role for calpain in the actin reorganization required for insulin-stimulated GLUT4 translocation to the plasma membrane in 3T3-L1 adipocytes. These studies identify calpain as a novel factor involved in GLUT4 vesicle trafficking and suggest a link between calpain activity and the development of type 2 diabetes. Topics: Actin Cytoskeleton; Adipocytes; Animals; Biological Transport; Calpain; Cell Line; Cell Membrane; Cysteine Proteinase Inhibitors; Glucose; Glucose Transporter Type 4; Insulin; Leupeptins; Monosaccharide Transport Proteins; Muscle Proteins; Phosphorylation; Protein Transport; Signal Transduction; Transport Vesicles | 2003 |
The mechanism of alphaB-crystallin gene expression by proteasome inhibition.
The mechanism of small heat shock protein/alphaB-crystallin gene expression by proteasome inhibition was investigated. Expression of alphaB-crystallin was induced efficiently only by proteasome inhibition and not by heat shock while expression of HSP27 was induced efficiently by both proteasome inhibition and heat shock. The promoter of the alphaB-crystallin gene contains two conserved heat shock elements, one located between -397 and -374 and the other between -57 and -37, relative to the transcription start site. Electrophoretic mobility shift assay (EMSA) revealed that proteasome inhibition induces binding of heat shock factors to both heat shock elements in the alphaB-crystallin gene promoter. However, a transient transfection assay using deletion constructs of the alphaB-crystallin gene promoter showed that the region between -373 and -58 plays an important role in promoter activity. These results indicate the presence of differential response mechanisms of alphaB-crystallin gene expression to proteasome inhibition and heat shock, and that the activation of heat shock elements is not sufficient for the efficient induction of the alphaB-crystallin gene by proteasome inhibition. Topics: Acetylcysteine; alpha-Crystallin B Chain; Animals; Cell Line; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Gene Expression; Heat-Shock Response; Leupeptins; Multienzyme Complexes; Myocytes, Cardiac; Promoter Regions, Genetic; Proteasome Endopeptidase Complex; Rats | 2003 |
Proteasome-mediated degradation of RNase L in response to phorbol-12-myristate-13-acetate (PMA) treatment of mouse L929 cells.
2'-5' Oligoadenylate (2-5A)-dependent RNase L is one of the key enzymes involved in the molecular mechanisms of interferon (IFN) function. Although the regulation of RNase L by 2-5A has been studied extensively, relatively little is known about how RNase L is controlled by posttranslational processes. Here, we report that phorbol-12-myristate-13-acetate (PMA) treatment of mouse L929 fibroblasts caused rapid degradation of RNase L in a dose-dependent and time-dependent manner. RNase L levels were decreased to 40% of control levels after only 5 min exposure of cells to PMA, suggesting the involvement of protein kinase C (PKC). After PMA treatment for 1 h, RNase L levels decreased to 18% of the pretreatment levels. Decay of RNase L was measured by 2-5A binding assay, ribonuclease activity, and protein levels in Western blots probed with antibody to murine RNase L. PMA treatment caused decreases in the levels of RNase L in both cytoplasm and nucleus. To explore the mechanism of RNase L degradation, we treated cells with the selective proteasome inhibitors, ALLN, MG132, and PSI, prior to PMA treatment. These inhibitors completely blocked the degradation of RNase L caused by PMA. Our results show a novel regulatory pathway for RNase L that could have an impact on its antitumor and antiviral functions. Topics: Animals; Apoptosis; Cell Line; Cell Nucleus; Cysteine Endopeptidases; Cytoplasm; Down-Regulation; Endoribonucleases; Interferon-alpha; Leupeptins; Mice; Multienzyme Complexes; Oligopeptides; Proteasome Endopeptidase Complex; Protein Transport; Tetradecanoylphorbol Acetate | 2003 |
Inhibition of apolipoprotein B secretion by taurocholate is controlled by the N-terminal end of the protein in rat hepatoma McArdle-RH7777 cells.
Bile salts (BS) inhibit the secretion of apolipoprotein B (apoB) and triacylglycerol (TG) in primary rat, mouse and human hepatocytes and in mice in vivo. We investigated whether lipidation of apoB into a lipoprotein particle is required for this inhibitory action of BS. The sodium/taurocholate co-transporting polypeptide (Ntcp) was co-expressed in McArdle-RH7777 (McA-RH7777) cells stably expressing the full-length human apoB100 (h-apoB100, secreted as TG-rich lipoprotein particles) or carboxyl-truncated human apoB18 (h-apoB18, secreted in lipid-free form). The doubly transfected cell lines (h-apoB/r-Ntcp) effectively accumulated taurocholic acid (TC). TC incubation decreased the secretion of endogenous rat apoB100 (-50%) and h-apoB18 (-35%), but did not affect secretion of rat apoA-I. Pulse-chase experiments (35S-methionine) indicated that the impaired secretion of radiolabeled h-apoB18 and h-apoB100 was associated with accelerated intracellular degradation. The calpain protease inhibitor N-acetyl-leucyl-leucyl-norleucinal (ALLN) partially inhibited intracellular apoB degradation but did not affect the amount of either h-apoB18 or h-apoB100 secreted into the medium, indicating that inhibition of apoB secretion by TC is not due to calpain-dependent proteasomal degradation. We conclude that TC does not inhibit apoB secretion by interference with its lipidation, but rather involves a mechanism dependent on the N-terminal end of apoB. Topics: Animals; Apolipoprotein A-I; Apolipoprotein B-100; Apolipoproteins B; Carrier Proteins; Cell Line, Tumor; Cysteine Proteinase Inhibitors; Hepatocytes; Leupeptins; Lipoproteins, VLDL; Liver Neoplasms, Experimental; Membrane Transport Proteins; Organic Anion Transporters, Sodium-Dependent; Rats; Symporters; Taurocholic Acid; Time Factors; Transfection; Triglycerides | 2003 |
Activation of monocytic cells through Fc gamma receptors induces the expression of macrophage-inflammatory protein (MIP)-1 alpha, MIP-1 beta, and RANTES.
Monocytic cells were stimulated with IgG-OVA equivalence immune complexes, mAb reacting with FcgammaRI, FcgammaRIIA, and FcgammaRIII, LPS, TNF-alpha, and the combination of ionomycin and phorbol ester, to address their effects on the expression of the mRNAs encoding for chemokines. Stimulation of monocytes with immune complexes induced a rapid expression of macrophage-inflammatory protein (MIP)-1alpha, MIP-1beta, and IL-8 mRNAs. In contrast, RANTES mRNA was already detectable in resting cells and only increased after 16 h of stimulation. A similar pattern was observed following homotypic stimulation of FcgammaR with mAb reacting with FcgammaRI and FcgammaRIIA, but not with a mAb reacting with FcgammaRIII, a subtype of receptor not expressed in THP-1 cells, thus indicating that both FcgammaRI and FcgammaRIIA are involved in the response. The pattern of chemokine induction elicited by LPS and the combination of ionomycin and PMA showed some similarities to those produced by FcgammaR cross-linking, although expression of IFN-gamma-inducible protein 10 mRNA was also observed in response to those agonists. The production of MIP-1alpha, MIP-1beta, and RANTES proteins encompassing the induction of their mRNAs was confirmed by specific ELISA. Experiments to address the transcription factors involved in the regulation of MIP-1alpha using pharmacological agents and EMSA showed the possible involvement of CCAAT/enhancer-binding protein beta sites and ruled out the functional significance of both NF-AT and AP-1 sites. Topics: Antigen-Antibody Complex; Base Sequence; Chemokine CCL3; Chemokine CCL4; Chemokine CCL5; Cross-Linking Reagents; Gene Expression Regulation; Humans; Interleukin-8; Leupeptins; Macrophage Activation; Macrophage Inflammatory Proteins; Molecular Sequence Data; Monocytes; NF-kappa B; Receptors, IgG; RNA, Messenger; Salicylates; Tumor Cells, Cultured | 2002 |
Low-density lipoprotein upregulates low-density lipoprotein receptor-related protein expression in vascular smooth muscle cells: possible involvement of sterol regulatory element binding protein-2-dependent mechanism.
Low-density lipoprotein (LDL) receptor-related protein (LRP) is highly expressed in vascular smooth muscle cells (VSMCs) of both normal and atherosclerotic lesions. However, little is known about LRP regulation in the vascular wall.. We analyzed the regulation of LRP expression in vitro in human VSMCs cultured with native LDL (nLDL) or aggregated LDL (agLDL) by semiquantitative reverse transcriptase-polymerase chain reaction, real-time polymerase chain reaction, and Western blot and in vivo during diet-induced hypercholesterolemia by in situ hybridization. LRP expression in human VSMCs is increased by nLDL and agLDL in a time- and dose-dependent manner. Maximal induction of LRP mRNA expression was observed after 24 hours of exposure to LDL. However, agLDL induced higher LRP mRNA expression (3.0-fold) than nLDL (1.76-fold). LRP mRNA upregulation was associated with an increase on LRP protein expression with the greatest induction by agLDL. VSMC-LRP upregulation induced by nLDL or agLDL was reduced by an inhibitor of sterol regulatory element binding protein (SREBP) catabolism (N-acetyl-leucyl-leucyl-norleucinal). In situ hybridization analysis indicates that there is a higher VSMC-LRP expression in hypercholesterolemic than in normocholesterolemic pig aortas.. These results indicate that LRP expression in VSMCs is upregulated by intravascular and systemic LDL. Topics: Animals; Aorta, Abdominal; Blotting, Western; Cells, Cultured; Cholesterol, Dietary; Chromatography, Thin Layer; Disease Models, Animal; DNA-Binding Proteins; Dose-Response Relationship, Drug; Female; Humans; Hypercholesterolemia; In Situ Hybridization; Leupeptins; Lipoproteins, LDL; Low Density Lipoprotein Receptor-Related Protein-1; Macromolecular Substances; Muscle, Smooth, Vascular; Polymers; Receptors, LDL; RNA, Messenger; Signal Transduction; Sterol Regulatory Element Binding Protein 2; Swine; Transcription Factors; Up-Regulation | 2002 |
Signaling pathways for TNF production induced by human aminoacyl-tRNA synthetase-associating factor, p43.
The p43 protein is associated with human macromolecular aminoacyl tRNA synthetase complex and secreted to up-regulate diverse proinflammatory genes including TNF. Here we focused on the p43-induced TNF production and determined its responsible signal pathway. The p43-induced TNF production was mediated by the activation of MAPK family members, ERK and p38 MAPK, and by IkappaB degradation leading to the activation of NFkappaB. We also studied the upstream molecules for ERK and p38 MAPK by using a variety of inhibitors. The inhibitors for protein kinase C (PKC) and phospholipase C (PLC) prevented the p43-induced TNF production. Interestingly, all of the effective drugs inhibited the ERK activity, while the drugs had no effects on p38 MAPK activity and IkappaB degradation. Together, the p43-induced TNF production was controlled by NFkB, p38 MAPK, and ERK that is dependent on the activities of PLC and PKC. Topics: Cytokines; Enzyme Inhibitors; Estrenes; Gene Expression Regulation; Humans; I-kappa B Proteins; Leupeptins; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Monocytes; Neoplasm Proteins; NF-kappa B; p38 Mitogen-Activated Protein Kinases; Phosphatidylinositol 3-Kinases; Phospholipase C gamma; Protein Kinase C; Pyrrolidinones; RNA-Binding Proteins; Signal Transduction; Staurosporine; Tumor Necrosis Factor-alpha; Type C Phospholipases | 2002 |
Wnt signals are transmitted through N-terminally dephosphorylated beta-catenin.
beta-catenin mediates Wnt signaling by acting as the essential co-activator for TCF transcription factors. Wnt signaling increases the half-life and therefore the absolute level of beta-catenin in responding cells. The current model states that these changes in beta-catenin stability set the threshold for Wnt signaling. However, we find that pharmacological inhibition of proteasome activity by ALLN leads to accumulation of cytosolic beta-catenin but not to increased TCF-mediated transcription. In addition, in temperature-sensitive ubiquitylation mutant CHO cells inhibition of ubiquitylation increases beta-catenin levels, but does not induce transcriptional activation of TCF reporter genes. Using an antibody specific for beta-catenin dephosphorylated at residues Ser37 and Thr41, we show that Wnt signals specifically increase the levels of dephosphorylated beta-catenin, whereas ALLN does not. We conclude that changes in the phosphorylation status of the N-terminus of beta-catenin that occur upon Wnt signaling independently affect the signaling properties and half-life of beta-catenin. Hence, Wnt signals are transduced via N-terminally dephosphorylated beta-catenin. Topics: Animals; beta Catenin; Calpain; CHO Cells; Cricetinae; Cytoskeletal Proteins; DNA-Binding Proteins; Leupeptins; Lymphoid Enhancer-Binding Factor 1; Phosphorylation; Proto-Oncogene Proteins; Signal Transduction; Trans-Activators; Transcription Factors; Transcription, Genetic; Wnt Proteins; Zebrafish Proteins | 2002 |
Regulation of transformed state by calpastatin via PKCepsilon in NIH3T3 mouse fibroblasts.
Ca(2+)-activated neutral protease calpain is ubiquitously expressed and may have pleiotropic biological functions. We have previously reported that repeated treatment of NIH3T3 mouse fibroblasts with the calpain inhibitor N-acetyl-Leu-Leu-norleucinal (ALLN) resulted in the induction of transformed foci [T. Hiwasa, T. Sawada, and S. Sakiyama (1990) Carcinogenesis 11, 75-80]. To elucidate further the effects of calpain in malignant transformation of NIH3T3 cells, calpastatin, an endogenous specific inhibitor of calpain, was expressed in NIH3T3 cells by transfection with cDNA. G418-selected calpastatin-expressing clones showed a significant increase in the anchorage-independent growth ability. A similar increase in cloning efficiency in soft agar medium was also observed in calpain small-subunit-transfected clones. On the other hand, reduced expression of calpastatin achieved by transfection with calpastatin antisense cDNA in Ha-ras-transformed NIH3T3 (ras-NIH) cells caused morphological reversion as well as a decrease in anchorage-independent growth. When NIH3T3 cells were treated with ALLN for 3 days, cell growth was stimulated by approximately 10%. This growth stimulation by ALLN was not observed in ras-NIH cells, but recovered by expression of a dominant negative form of protein kinase C (PKC)epsilon but not by that of PKCalpha. Western blotting analysis showed that an increase in PKCepsilon was much more prominent than that of PKCalpha in NIH3T3 cells after treatment with ALLN. These results are concordant with the notion that calpain suppresses malignant transformation by predominant degradation of PKCepsilon. Topics: 3T3 Cells; Animals; Blotting, Western; Calcium; Calcium-Binding Proteins; Cell Division; Cell Line; Cysteine Proteinase Inhibitors; DNA, Complementary; Gene Expression Regulation; Genes, Dominant; Humans; Isoenzymes; Leupeptins; Mice; Phenotype; Protein Kinase C; Protein Kinase C-alpha; Protein Kinase C-epsilon; ras Proteins; Time Factors; Transfection | 2002 |
Proteasomal degradation of retinoblastoma-related p130 during adipocyte differentiation.
Within 24 h of hormonally stimulated 3T3-L1 adipocyte differentiation, there are dramatic changes in the protein levels of p130 and p107, two members of the retinoblastoma tumor suppressor gene family. Designated the "p103:p107" switch, this alteration is characterized by a rapid and transient drop in p130 protein levels accompanied by a transient increase in both p107 mRNA and protein levels. Using protease inhibitors, the specific proteolytic pathway involved in degradation of p130 was examined. Treatment of cells with N-acetyl-leu-leu-norleucinal, an inhibitor that blocks proteolytic activity of type I calpain and the 26S proteasome, resulted in a complete block in the degradation of p130 protein, as well as adipocyte differentiation, suggesting that one of these pathways is involved in regulating p130 protein levels. Similar analysis with lactacystin, a specific inhibitor of the 26S proteasome, also resulted in a complete block in both differentiation and p130 degradation. Furthermore, both inhibitors blocked the increase in p107 protein levels normally observed on Day 1, suggesting that the p130:p107 switch is required for adipocyte differentiation and one of the early molecular events involved in activating the p130:p107 switch is the specific degradation of p130 by the 26S proteasome. Topics: Acetylcysteine; Adipocytes; Animals; Calpain; Cell Differentiation; Cell Line; Cysteine Proteinase Inhibitors; Kinetics; Leupeptins; Nuclear Proteins; Peptide Hydrolases; Phosphoproteins; Proteasome Endopeptidase Complex; Proteins; Retinoblastoma-Like Protein p130 | 2002 |
Ubiquitin/proteasome-dependent degradation of D-type cyclins is linked to tumor necrosis factor-induced cell cycle arrest.
Tumor necrosis factor-alpha (TNF) is well known for its cytotoxic effect on malignant cells. Its role in cell cycle control is relatively less known. In this study, we found that TNF induced G(1) arrest of TF-1 and MV4-11 cells while simultaneously causing apoptosis. Treatment of the cells with TNF for 48 h caused cell cycle arrest, accompanied by dephosphorylation of pRb and reduction in D-type cyclin expression. The down-regulation of the D-type cyclins resulted in approximately 50-80% decrease of the cyclin-dependent kinase activities. Cells treated with calpain-dependent inhibitor ALLN and apoptosis inhibitor zVAD-FMK suppressed degradation of IkappaBalpha and activation of caspase 3, respectively. However, treatment of cells with these two inhibitors was not able to prevent TNF-induced down-regulation of the D-type cyclins. In contrast, proteasome inhibitor MG-132 and lactacystin blocked both TNF-induced degradation of IkappaBalpha and down-regulation of D-type cyclins. These data suggest that down-regulation of D-type cyclins by TNF may be proteasome-proteolysis dependent. Additional support for this conclusion was obtained from experiments showing an increase of proteasome activity in TNF-treated cells and in vitro degradation of cyclin D3 by 26 S proteasome. Topics: Amino Acid Chloromethyl Ketones; Apoptosis; Blotting, Western; Calpain; Caspase 3; Caspases; Cell Cycle; Cell Line; Cyclin D; Cyclin D3; Cyclins; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Dose-Response Relationship, Drug; Down-Regulation; Enzyme Activation; G1 Phase; Humans; I-kappa B Proteins; Leupeptins; Multienzyme Complexes; NF-KappaB Inhibitor alpha; Peptide Hydrolases; Plasmids; Precipitin Tests; Proteasome Endopeptidase Complex; Protein Binding; Protein Biosynthesis; Ribonucleases; RNA; RNA, Messenger; Spectrometry, Fluorescence; Time Factors; Transcription, Genetic; Tumor Necrosis Factor-alpha; Ubiquitin | 2002 |
Helical apolipoproteins stabilize ATP-binding cassette transporter A1 by protecting it from thiol protease-mediated degradation.
ATP-binding cassette transporter (ABC) A1 was increased by apolipoprotein A-I without an increase of its message in THP-1 cells. The pulse label study demonstrated that apoA-I retarded degradation of ABCA1. Similar changes were demonstrated by apoA-II, but the effect of high density lipoprotein was almost negligible on the basis of equivalent protein concentration. Thiol protease inhibitors (leupeptin and N-acetyl-Leu-Leu-norleucinal (ALLN)) increased ABCA1 and slowed its decay in the cells, whereas none of the proteosome-specific inhibitor lactacystin, other protease inhibitors, or the lysosomal inhibitor NH(4)Cl showed such effects. The effects of apoA-I and ALLN were additive for the increase of ABCA1, and the apoA-I-mediated cellular lipid release was enhanced by ALLN. The data suggest that ABCA1 is rapidly degraded by a thiol protease(s) in the cells unless helical apolipoproteins in their lipid-free form stabilize ABCA1 by protecting it from protease-mediated degradation. Topics: Adenosine Triphosphate; Apolipoproteins; Apolipoproteins E; ATP Binding Cassette Transporter 1; ATP-Binding Cassette Transporters; Cell Differentiation; Cell Line; Cell Membrane; Cholesterol; Cysteine Proteinase Inhibitors; DNA, Complementary; Dose-Response Relationship, Drug; Humans; Immunoblotting; Leupeptins; Lipid Metabolism; Precipitin Tests; Protease Inhibitors; Protein Binding; Reverse Transcriptase Polymerase Chain Reaction; Tetradecanoylphorbol Acetate; Time Factors | 2002 |
The time course of cardiac NF-kappaB activation and TNF-alpha secretion by cardiac myocytes after burn injury: contribution to burn-related cardiac contractile dysfunction.
Previous studies have suggested that cardiac synthesis of TNF-alpha contributes to myocardial dysfunction in several models of trauma, sepsis and ischemia. Therefore, it is likely that myocyte secretion of TNF-alpha occurs early after major burn trauma, contributing to progressive cardiac contractile dysfunction that is characteristic of thermal injury. This study examined the time course of nuclear translocation of the transcription factor NF-kappaB, the time course of TNF-alpha secretion by cardiomyocytes after burn trauma, and the development of cardiac contractile defects. Rats were given burn injury over 40% TBSA (sham burns included for controls), and fluid resuscitation included lactated Ringer's solution, 4 mL/kg/%burn. Subsets of rats were sacrificed at several times postburn (1, 2, 4, 8, 12, 18 and 24 h), hearts were harvested to prepare cardiomyocytes (N = 4 rats/group/time period), to prepare nuclear fractions to measure burn-induced NF-kappaB activation (N = 3-4 rats/group/time period), or to examine the time course of postburn cardiac contractile dysfunction (N = 6-7 rats/group/time period). Despite aggressive fluid resuscitation, burn trauma activated NF-kappaB 4 h postburn, and this activation persisted over the 24 h study period. In addition, burn trauma produced a time-related increase in TNF-alpha secretion by cardiac myocytes with cytokine secretion evident 1 h postburn. Cardiac dysfunction occurred 8 h postburn and persisted over the 24 h study period. Administration of a strategy designed to inhibit NF-kappaB activation (N-acetyl-leucinyl-leucinyl-norleucinal, ALLN, 50 mg/kg, in additional groups of burn rats) inhibited TNF-alpha secretion by cardiac myocytes and improved myocardial function. This study confirms that burn trauma activates myocardial NF-kappaB and promotes cardiomyocyte secretion of TNF-alpha. This inflammatory cascade preceded the appearance of cardiac dysfunction, suggesting that cardiac myocyte derived TNF-alpha contributes, in part, to postburn cardiac contractile deficits. Topics: Animals; Burns; Heart; Heart Diseases; Hemodynamics; In Vitro Techniques; Inflammation Mediators; Kinetics; Leupeptins; Myocardial Contraction; Myocardium; NF-kappa B; Rats; Rats, Sprague-Dawley; RNA, Messenger; Tumor Necrosis Factor-alpha | 2002 |
Copper uptake is required for pyrrolidine dithiocarbamate-mediated oxidation and protein level increase of p53 in cells.
The p53 tumour-suppressor protein is a transcription factor that activates the expression of genes involved in cell cycle arrest, apoptosis and DNA repair. The p53 protein is vulnerable to oxidation at cysteine thiol groups. The metal-chelating dithiocarbamates, pyrrolidine dithiocarbamate (PDTC), diethyldithiocarbamate, ethylene(bis)dithiocarbamate and H(2)O(2) were tested for their oxidative effects on p53 in cultured human breast cancer cells. Only PDTC oxidized p53, although all oxidants tested increased the p53 level. Inductively coupled plasma MS analysis indicated that the addition of 60 microM PDTC increased the cellular copper concentration by 4-fold, which was the highest level of copper accumulated amongst all the oxidants tested. Bathocuproinedisulphonic acid, a membrane-impermeable Cu(I) chelator inhibited the PDTC-mediated copper accumulation. Bathocuproinedisulphonic acid as well as the hydroxyl radical scavenger d-mannitol inhibited the PDTC-dependent increase in p53 protein and oxidation. Our results show that a low level of copper accumulation in the range of 25-40 microg/g of cellular protein increases the steady-state levels of p53. At copper accumulation levels higher than 60 microg/g of cellular protein, p53 is oxidized. These results suggest that p53 is vulnerable to free radical-mediated oxidation at cysteine residues. Topics: Animals; Antioxidants; Breast Neoplasms; Cells, Cultured; Chelating Agents; Copper; Cysteine; Cysteine Proteinase Inhibitors; Ditiocarb; Female; Fibroblasts; Free Radicals; Humans; Hydrogen Peroxide; Leupeptins; Mannitol; Molecular Structure; Oxidants; Oxidation-Reduction; Pyrrolidines; Rats; Recombinant Proteins; Thiobarbituric Acid Reactive Substances; Thiocarbamates; Tumor Suppressor Protein p53 | 2002 |
Dislocation and degradation from the ER are regulated by cytosolic stress.
Akey step in ER-associated degradation (ERAD) is dislocation of the substrate protein from the ER into the cytosol to gain access to the proteasome. Very little is known about how this process is regulated, especially in the case of polytopic proteins. Using pulse-chase analysis combined with subcellular fractionation, we show that connexins, the four transmembrane structural components of gap junctions, can be chased in an intact form from the ER membrane into the cytosol of proteasome inhibitor-treated cells. Dislocation of endogenously expressed connexin from the ER was reduced 50-80% when the cytosolic heat shock response was induced by mild oxidative or thermal stress, but not by treatments that instead upregulate the ER unfolded protein response. Cytosolic but not ER stresses slowed the normally rapid degradation of connexins, and led to a striking increase in gap junction formation and function in otherwise assembly-inefficient cell types. These treatments also inhibited the dislocation and turnover of a connexin-unrelated ERAD substrate, unassembled major histocompatibility complex class I heavy chain. Our findings demonstrate that dislocation is negatively regulated by physiologically relevant, nonlethal stress. They also reveal a previously unrecognized relationship between cytosolic stress and intercellular communication. Topics: Animals; Cell Line; CHO Cells; Connexins; Cricetinae; Cysteine Proteinase Inhibitors; Cytosol; Endoplasmic Reticulum; Gap Junctions; Heat-Shock Response; Leupeptins; Oxidation-Reduction; Peptide Hydrolases; Proteasome Endopeptidase Complex | 2002 |
Hydroxy-methylglutaryl-coenzyme A reductase inhibition promotes endothelial nitric oxide synthase activation through a decrease in caveolin abundance.
Hypercholesterolemia is causally associated with defects of endothelial nitric oxide (NO)-dependent vasodilation. Increased uptake of cholesterol by endothelial cells (ECs) upregulates the abundance of the structural protein caveolin-1 and impairs NO release through the stabilization of the inhibitory heterocomplex between caveolin-1 and endothelial NO synthase (eNOS). Therefore, we examined whether the hydroxy-methylglutaryl-coenzyme A reductase inhibitor atorvastatin modulates caveolin abundance, eNOS activity, and NO release through a reduction in endogenous cholesterol levels.. ECs were incubated with increasing doses of atorvastatin in the absence or in the presence of human LDL cholesterol (LDL-Chol) fractions in the presence of antioxidants. Our results show that atorvastatin (10 nmol/L to 1 micromol/L) reduced caveolin-1 abundance in the absence (-75%) and in the presence (-20% to 70%) of LDL-Chol. This was paralleled by a decreased inhibitory interaction between caveolin-1 and eNOS and a restoration and/or potentiation of the basal (+45%) and agonist-stimulated (+107%) eNOS activity. These effects were observed in the absence of changes in eNOS abundance and were reversed with mevalonate. In the presence of LDL-Chol, atorvastatin also promoted the agonist-induced association of eNOS and the chaperone Hsp90, resulting in the potentiation of eNOS activation.. We provide biochemical and functional evidence that atorvastatin promotes NO production by decreasing caveolin-1 expression in ECs, regardless of the level of extracellular LDL-Chol. These findings highlight the therapeutic potential of inhibiting cholesterol synthesis in peripheral cells to correct NO-dependent endothelial dysfunction associated with hypercholesterolemia and possibly other diseases. Topics: Animals; Atorvastatin; Cattle; Caveolin 1; Caveolins; CCAAT-Enhancer-Binding Proteins; Cells, Cultured; Cholesterol; Cholesterol, LDL; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Dose-Response Relationship, Drug; Endothelium, Vascular; Heptanoic Acids; HSP90 Heat-Shock Proteins; Humans; Hydroxymethylglutaryl CoA Reductases; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Leupeptins; Lipoproteins, LDL; Nitric Oxide; Nitric Oxide Synthase; Nitric Oxide Synthase Type III; Pyrroles; Sterol Regulatory Element Binding Protein 1; Transcription Factors; Up-Regulation | 2001 |
A mutation of the Wilson disease protein, ATP7B, is degraded in the proteasomes and forms protein aggregates.
Wilson disease is a genetic disorder characterized by the accumulation of copper in the body as a result of a defect of copper excretion from hepatocytes. The intracellular localization of the Wilson disease gene product, ATP7B, was recently identified as the late endosomes. Various mutations have been documented in patients with Wilson disease. The clinical manifestations vary greatly among the patients; however, there is little information on the genotype-phenotype correlation.. We investigated the distribution of a common ATP7B mutant His1069Gln and a mutant Asp1270Ser by expressing the mutants tagged with green fluorescent protein in Huh7 and HEK293 cells. Intracellular organelles were visualized by fluorescence microscopy.. Although the wild-type ATP7B and Asp1270Ser mutant localized in the late endosomes, His1069Gln mutant did not locate in the late endosomes and was degraded by the proteasomes in the cytoplasm. Furthermore, His1069Gln formed aggresomes composed of the degradates and intermediate filaments at the microtubule-organizing center. These aggresomes were similar to Mallory bodies on electron microscopy.. The different protein properties of ATP7B mutants may explain the variety of clinical spectrums in patients with Wilson disease. Topics: Acetylcysteine; Adenosine Triphosphatases; Carrier Proteins; Cation Transport Proteins; Cell Line; Copper-Transporting ATPases; Cysteine Endopeptidases; Cytoskeleton; Fluorescent Antibody Technique; Humans; Leupeptins; Microscopy, Confocal; Microscopy, Electron; Multienzyme Complexes; Mutation; Proteasome Endopeptidase Complex; Tissue Distribution | 2001 |
Role of proteasomal degradation in the cell cycle-dependent regulation of DNA topoisomerase IIalpha expression.
1DNA topoisomerase II (topo II) is a nuclear enzyme that modifies DNA topology and also serves as a target to mediate the cytotoxicity of several antineoplastic agents. Several reports have demonstrated that a reduction of topo II is associated with reduced sensitivity to these agents. Topo II exists as two isoforms in mammalian cells: topo IIalpha and topo IIbeta. In MCF-7 cells, the half-life (mean +/- SEM) values of topo IIalpha and topo IIbeta in situ were 6.6 +/- 0.3 and 17.6 +/- 2.3 hr, respectively, as determined by [(35)S]methionine/cysteine pulse-chase analysis. Degradation of topo IIalpha in situ was abrogated by the presence of proteasome inhibitors, and the relative activities were carbobenzoxy-leucyl-leucyl-leucinal (MG132) > carbobenzoxy-leucyl-leucyl-norvalinal (MG115) > ALLN congruent with lactacystin. ATP-dependent degradation of topo IIalpha, but not topo IIbeta, was observed in extracts of asynchronously dividing HeLa and MCF-7 cells. Furthermore, degradation of topo IIalpha was abrogated by the proteasome inhibitors MG132 and MG115, but not by lactacystin, in extracts of asynchronously dividing MCF-7 cells. Finally, degradation of topo IIalpha, but not topo IIbeta, was observed to occur in a cell cycle-dependent fashion, in extracts of synchronized HeLa cells, with maximal loss of the alpha isoform occurring 2 hr after release from mitotic arrest. This degradation of topo IIalpha appeared to be facilitated by an ATP-dependent activity. Furthermore, high molecular weight bands (>200 kDa), which may represent polyubiquitinated-topo IIalpha conjugates, were also detected in extracts of synchronized HeLa cells. This study provides evidence for a role of the ubiquitin-proteasome pathway in the cell cycle-dependent regulation of topo IIalpha expression. Topics: Acetylcysteine; Adenosine Triphosphate; Antigens, Neoplasm; Cell Cycle; Cell Extracts; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA Topoisomerases, Type II; DNA-Binding Proteins; HeLa Cells; Humans; Isoenzymes; Leupeptins; Multienzyme Complexes; Proteasome Endopeptidase Complex; Tumor Cells, Cultured | 2001 |
Peptide transport by the multidrug resistance protein MRP1.
Small hydrophobic peptides were studied as possible substrates of the multidrug resistance protein (MRP)-1 (ABCC1) transmembrane transporter molecule. As observed earlier for P-glycoprotein- (Pgp; ABCB1) overexpressing cells, MRP1-overexpressing cells, including cells stably transfected with the MRP1 cDNA, showed distinct resistance to the cytotoxic peptide N-acetyl-Leu-Leu-norleucinal (ALLN). Resistance to this peptide and another toxic peptide derivative, which is based on a Thr-His-Thr-Nle-Glu-Gly backbone conjugated to butyl and benzyl groups (4A6), could be reversed by MRP1 inhibitors. The reduced toxicity of 4A6 in MRP1-overexpressing cells was found to be associated with lower accumulation of a fluorescein-labeled derivative of this peptide. Glutathione (GSH) depletion had a clear effect on resistance to ALLN but hardly affected 4A6 resistance. In a limited structure-activity study using peptides that are analogous to 4A6, MRP1-overexpressing cells were found to be resistant to these peptides as well. Remarkably, when selecting A2780 ovarian cancer cells for resistance to ALLN, even in the absence of Pgp blockers, resulting cell lines had up-regulated MRP1, rather than any of the other currently known multidrug resistance transporter molecules including Pgp, MRP2 (ABCC2), MRP3 (ABCC3), MRP5 (ABCCS), and the breast cancer resistance protein ABCG2. ALLN-resistant, MRP1-overexpressing cells were found to be cross-resistant to 4A6 and the classical multidrug resistance drugs doxorubicin, vincristine, and etoposide. This establishes MRP1 as a transporter for small hydrophobic peptides. More extensive structure-activity relationship studies should allow the identification of clinically useful peptide antagonists of MRP1. Topics: Anti-Bacterial Agents; Antimetabolites, Antineoplastic; ATP-Binding Cassette Transporters; Biological Transport; Buthionine Sulfoximine; Drug Resistance, Multiple; Drug Resistance, Neoplasm; Drug Synergism; HL-60 Cells; Humans; Leupeptins; Multidrug Resistance-Associated Protein 2; Multidrug Resistance-Associated Proteins; Oligopeptides; Tumor Cells, Cultured; Valinomycin | 2001 |
Impairment of the ubiquitin-proteasome system by protein aggregation.
Intracellular deposition of aggregated and ubiquitylated proteins is a prominent cytopathological feature of most neurodegenerative disorders. Whether protein aggregates themselves are pathogenic or are the consequence of an underlying molecular lesion is unclear. Here, we report that protein aggregation directly impaired the function of the ubiquitin-proteasome system. Transient expression of two unrelated aggregation-prone proteins, a huntingtin fragment containing a pathogenic polyglutamine repeat and a folding mutant of cystic fibrosis transmembrane conductance regulator, caused nearly complete inhibition of the ubiquitin-proteasome system. Because of the central role of ubiquitin-dependent proteolysis in regulating fundamental cellular events such as cell division and apoptosis, our data suggest a potential mechanism linking protein aggregation to cellular disregulation and cell death. Topics: Acetylcysteine; Amino Acid Sequence; Cell Death; Cell Line; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cystic Fibrosis Transmembrane Conductance Regulator; Endoplasmic Reticulum; G2 Phase; Green Fluorescent Proteins; Humans; Huntingtin Protein; Inclusion Bodies; Leupeptins; Luminescent Proteins; Molecular Sequence Data; Multienzyme Complexes; Nerve Tissue Proteins; Nuclear Proteins; Proteasome Endopeptidase Complex; Recombinant Fusion Proteins; Transfection; Ubiquitins | 2001 |
Dendritic growth induced by BMP-7 requires Smad1 and proteasome activity.
Bone morphogenetic proteins (BMPs) induce dendritic growth in cultured sympathetic neurons; however, the signaling pathways that mediate this dendrite-promoting activity have not been previously characterized. Here we report studies of the signaling events that regulate the growth of these afferent processes. We find that Smad1 is expressed in sympathetic neurons and that BMPs rapidly induce its phosphorylation and translocation from the cytoplasm to the nucleus. Furthermore, a dominant negative form of Smad1 inhibits BMP-7-induced dendritic growth, suggesting a requirement for Smad1 activation in this biological activity of BMP-7. A physical interaction between Smad1 and components involved in the proteasome-mediated degradation system was detected with a yeast two-hybrid screen, thereby prompting an examination of the effects of proteasome inhibitors on dendritic growth. Lactacystin and ALLN (N-acetyl-Leu-Leu-norleucinal) selectively blocked BMP-7-induced dendritic growth without adversely affecting either cell viability or axonal growth. Moreover, studies of transfected P19 cells suggest that the proteasome inhibitors directly block the effects of Smad1 on the transcriptional activity of the Tlx-2 promoter. These data indicate that BMP-induced dendritic growth requires Smad1 activation and involves proteasome-mediated degradation events. Topics: Acetylcysteine; Animals; Bone Morphogenetic Protein 7; Bone Morphogenetic Proteins; Cell Nucleus; Cells, Cultured; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Dendrites; DNA-Binding Proteins; Gene Expression; Leupeptins; Multienzyme Complexes; Neurons; Neuroprotective Agents; Oligopeptides; Proteasome Endopeptidase Complex; Rats; Rats, Sprague-Dawley; Smad Proteins; Smad1 Protein; Superior Cervical Ganglion; Trans-Activators; Transcriptional Activation; Transfection; Transforming Growth Factor beta | 2001 |
Alpha-synuclein-enhanced green fluorescent protein fusion proteins form proteasome sensitive inclusions in primary neurons.
Alpha-synuclein accumulates in the brains of sporadic Parkinson's disease patients as a major component of Lewy bodies, and mutations in alpha-synuclein are associated with familial forms of Parkinson's disease. The pathogenic mechanisms that precede and promote the aggregation of alpha-synuclein into Lewy bodies in neurons remain to be determined. Here, we constructed a series of alpha-synuclein-enhanced green fluorescent protein (alpha-synucleinEGFP, SynEGFP) fusion proteins to address whether the Parkinson's disease-associated mutations alter the subcellular distribution of alpha-synuclein, and to use as a tool for experimental manipulations to induce aggregate formation. When transfected into mouse cultured primary neurons, the 49-kDa alpha-synucleinEGFP fusion proteins are partially truncated to a approximately 27-kDa form. This non-fluorescent carboxy-terminally modified fusion protein spontaneously forms inclusions in the neuronal cytoplasm. A marked increase in the accumulation of inclusions is detected following treatment with each of three proteasome inhibitors, n-acetyl-leu-leu-norleucinal, lactacystin and MG132. Interestingly, Ala30Pro alpha-synucleinEGFP does not form the cytoplasmic inclusions that are characteristic of wild-type and Ala53Thr alpha-synucleinEGFP, supporting the idea that the Ala30Pro alpha-synuclein protein conformation differs from wild-type alpha-synuclein. Similar inclusions are formed if alpha-synuclein carboxy-terminus is modified by the addition of a V5/6xHistidine epitope tag. By contrast, overexpression of unmodified alpha-synuclein does not lead to aggregate formation. Furthermore, synphilin-1, an alpha-synuclein interacting protein also found in Lewy bodies, colocalizes with the carboxy-terminally truncated alpha-synuclein fusion protein in discrete cytoplasmic inclusions.Our finding that manipulations of the carboxy-terminus of alpha-synuclein lead to inclusion formation may provide a model for studies of the pathogenic mechanisms of alpha-synuclein aggregation in Lewy bodies. Topics: alpha-Synuclein; Animals; Brain; Carrier Proteins; Cerebral Cortex; Cryoprotective Agents; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Dimethyl Sulfoxide; Fetus; Genes, Reporter; Green Fluorescent Proteins; Humans; Immunohistochemistry; Inclusion Bodies; Intracellular Signaling Peptides and Proteins; Leupeptins; Luminescent Proteins; Mice; Multienzyme Complexes; Nerve Tissue Proteins; Neurons; Parkinson Disease; Proteasome Endopeptidase Complex; Protein Structure, Tertiary; Recombinant Fusion Proteins; Synucleins; Tumor Cells, Cultured | 2001 |
Proteasome inhibitors induce Fas-mediated apoptosis by c-Myc accumulation and subsequent induction of FasL message in human glioma cells.
Proteasome inhibitors were shown previously to induce mitochondria-independent and caspase-3-dependent apoptosis in human glioma cell lines by unknown mechanisms. Here, we showed that treatment with proteasome inhibitors, lactacystin or acetyl-leucinyl-leucinyl-norleucinal, led to elevation of the steady-state c-Myc protein but not c-myc mRNA, suggesting the accumulation of c-Myc protein by proteasome inhibitors. In addition, the marked association of c-Myc protein with ubiquitin by treatment with proteasome inhibitors indicated the involvement of proteasome in c-Myc proteolysis and the stabilization of c-Myc protein by proteasome inhibitors in vivo. The expression of Fas (also termed CD95 or APO-1) mRNA, if analyzed by reverse transcriptase polymerase chain reaction assay, was found to occur constitutively, and increased slightly by the treatment with proteasome inhibitors. In contrast, the expression of Fas ligand (FasL) mRNA was markedly induced temporarily before the activation of caspase-3 by the treatment. Agonistic anti-Fas antibody (CH11) induced apoptotic cell death, suggesting the presence of a functional Fas receptor. In addition, proteasome inhibitor-induced apoptosis was prevented by the addition of antagonistic anti-FasL antibody (4A5) or z-IETD.fmk, a potent inhibitor of caspase-8, indicating the involvement of the Fas receptor-ligand apoptotic signaling system in proteasome inhibitor-mediated apoptosis. Thus, it is suggested that proteasome inhibitors cause the accumulation of c-Myc protein which induces transiently FasL message to stimulate the Fas receptor-ligand apoptotic signaling pathway. Topics: Acetylcysteine; Apoptosis; Base Sequence; Brain Neoplasms; Cysteine Proteinase Inhibitors; DNA Primers; Fas Ligand Protein; fas Receptor; Glioma; Humans; Leupeptins; Membrane Glycoproteins; Proto-Oncogene Proteins c-myc; Tumor Cells, Cultured | 2001 |
Evidence for the Involvement of annexin 6 in the trafficking between the endocytic compartment and lysosomes.
Annexins are a family of calcium-dependent phospholipid-binding proteins, which have been implicated in a variety of biological processes including membrane trafficking. The annexin 6/lgp120 prelysosomal compartment of NRK cells was loaded with low-density lipoprotein (LDL) and then its transport from this endocytic compartment and its degradation in lysosomes were studied. NRK cells were microinjected with the mutated annexin 6 (anx6(1-175)), to assess the possible involvement of annexin 6 in the transport of LDL from the prelysosomal compartment. The results indicated that microinjection of mutated annexin 6, in NRK cells, showed the accumulation of LDL in larger endocytic structures, denoting retention of LDL in the prelysosomal compartment. To confirm the involvement of annexin 6 in the trafficking and the degradation of LDL we used CHO cells transfected with mutated annexin 6(1-175). Thus, in agreement with NRK cells the results obtained in CHO cells demonstrated a significant inhibition of LDL degradation in CHO cells expressing the mutated form of annexin 6 compared to controls overexpressing wild-type annexin 6. Therefore, we conclude that annexin 6 is involved in the trafficking events leading to LDL degradation. Topics: Animals; Annexin A6; Calpain; Carbocyanines; Cell Compartmentation; CHO Cells; Cricetinae; Endocytosis; Fluorescent Antibody Technique; Fluorescent Dyes; Genetic Vectors; Iodine Radioisotopes; Leupeptins; Lipoproteins, LDL; Lysosomes; Protein Transport; Spectrin; Subcellular Fractions; Transport Vesicles | 2001 |
Proteasome-mediated degradation of BRCA1 protein in MCF-7 human breast cancer cells.
The breast and ovarian cancer susceptibility gene BRCA1 encodes a nuclear phosphoprotein, which functions as a tumor suppressor gene. We present several lines of evidence for the mechanism of BRCA1 degradation through the ubiquitin-proteasome pathway by using specific inhibitors of the proteasome in human MCF-7 breast carcinoma cells. The levels of BRCA1 protein were up-regulated by proteasome inhibitors, such as MG-132 and ALLnL, suggesting rapid degradation via the ubiquitin-proteasome pathway. The enhanced loss of BRCA1 protein by taxol, okadaic acid or nocodazole treatment was prevented by the proteasome inhibitors, while inhibition of other proteases was ineffective. Accumulation and proteasomal degradation of BRCA1 protein appear to be restricted entirely to the nucleus. We also detected that high molecular weight BRCA1 protein species appeared after proteasome inhibitor treatments, which indicated that ubiquitinated species were present. Moreover the half-life of BRCA1 protein was significantly increased in response to inhibition of proteasome activity. Our present data demonstrate that BRCA1 protein may be degraded through the ubiquitin-proteasome mediated pathway. Topics: Apoptosis; Blotting, Western; BRCA1 Protein; CDC2-CDC28 Kinases; Cell Cycle; Cyclin G; Cyclin G1; Cyclin-Dependent Kinase 2; Cyclin-Dependent Kinase 4; Cyclin-Dependent Kinases; Cyclins; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Fluorescent Antibody Technique; Humans; Leupeptins; Multienzyme Complexes; Paclitaxel; Poly(ADP-ribose) Polymerases; Proteasome Endopeptidase Complex; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Tumor Cells, Cultured; Tumor Suppressor Protein p53; Up-Regulation | 2001 |
Proteasomes and ubiquitin are involved in the turnover of the wild-type prion protein.
Prion diseases propagate by converting a normal glycoprotein of the host, PrP(C), into a pathogenic "prion" conformation. Several misfolding mutants of PrP(C) are degraded through the ER-associated degradation (ERAD)-proteasome pathway. In their infectious form, prion diseases such as bovine spongiform encephalopathy involve PrP(C) of wild-type sequence. In contrast to mutant PrP, wild-type PrP(C) was hitherto thought to be stable in the ER and thus immune to ERAD. Using proteasome inhibitors, we now show that approximately 10% of nascent PrP(C) molecules are diverted into the ERAD pathway. Cells incubated with N-acetyl-leucinal-leucinal-norleucinal (ALLN), lactacystin or MG132 accumulated both detergent-soluble and insoluble PrP species. The insoluble fraction included an unglycosylated 26 kDa PrP species with a protease-resistant core, and a M(r) "ladder" that contained ubiquitylated PrP. Our results show for the first time that wild-type PrP(C) molecules are subjected to ERAD, in the course of which they are dislocated into the cytosol and ubiquitylated. The presence of wild-type PrP molecules in the cytosol may have potential pathogenic implications. Topics: Animals; Brefeldin A; CHO Cells; Cricetinae; Cysteine Endopeptidases; Cytoplasm; Endoplasmic Reticulum; Leupeptins; Membrane Proteins; Mice; Multienzyme Complexes; Protease Inhibitors; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; PrPC Proteins; Solubility; Tumor Cells, Cultured; Ubiquitin | 2001 |
Chemotropic responses of retinal growth cones mediated by rapid local protein synthesis and degradation.
Growth cones contain mRNAs, translation machinery, and, as we report here, protein degradation machinery. We show that isolated retinal growth cones immediately lose their ability to turn in a chemotropic gradient of netrin-1 or Sema3A when translation is inhibited. Translation inhibition also prevents Sema3A-induced collapse, while LPA-induced collapse is not affected. Inhibition of proteasome function blocks responses to netrin-1 and LPA but does not affect Sema3A responses. We further demonstrate in isolated growth cones that netrin-1 and Sema3A activate translation initiation factors and stimulate a marked rise in protein synthesis within minutes, while netrin-1 and LPA elicit similar rises in ubiquitin-protein conjugates. These results suggest that guidance molecules steer axon growth by triggering rapid local changes in protein levels in growth cones. Topics: Amanitins; Animals; Carrier Proteins; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Drosophila Proteins; Eukaryotic Initiation Factor-4E; Eye Proteins; Glycoproteins; Growth Cones; Leupeptins; Lipopolysaccharides; Multienzyme Complexes; Nerve Growth Factors; Netrin-1; Nucleic Acid Synthesis Inhibitors; Peptide Initiation Factors; Phosphatidylinositol 3-Kinases; Phosphoproteins; Proteasome Endopeptidase Complex; Protein Biosynthesis; Receptor Protein-Tyrosine Kinases; Retina; Semaphorin-3A; Tumor Suppressor Proteins; Ubiquitin; Xenopus | 2001 |
Characterization of peroxisomal 3-hydroxy-3-methylglutaryl coenzyme A reductase in UT2 cells: sterol biosynthesis, phosphorylation, degradation, and statin inhibition.
We have previously identified a CHO cell line (UT2 cells) that expresses only one 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase protein which is localized exclusively in peroxisomes [Engfelt, H.W., Shackelford, J.E., Aboushadi, N., Jessani, N., Masuda, K., Paton, V.G., Keller, G.A., and Krisans, S.K. (1997) J. Biol. Chem. 272, 24579-24587]. In this study, we utilized the UT2 cells to determine the properties of the peroxisomal reductase independent of the endoplasmic reticulum (ER) HMG-CoA reductase. We demonstrated major differences between the two proteins. The peroxisomal reductase is not the rate-limiting enzyme for cholesterol biosynthesis in UT2 cells. The peroxisomal reductase protein is not phosphorylated, and its activity is not altered in the presence of inhibitors of cellular phosphatases. Its rate of degradation is not accelerated in response to mevalonate. Finally, the degradation process is not blocked by N-acetyl-Leu-Leu-norleucinal (ALLN). Furthermore, the peroxisomal HMG-CoA reductase is significantly more resistant to inhibition by statins. Taken together, the data support the conclusion that the peroxisomal reductase is functionally and structurally different from the ER HMG-CoA reductase. Topics: Acetic Acid; Acetyl-CoA C-Acetyltransferase; Adenosine Triphosphate; Animals; Carbon Radioisotopes; Cell Cycle; Cell Survival; CHO Cells; Cholesterol; Clone Cells; Cricetinae; Deuterium Oxide; Enzyme Activation; Fatty Acids, Unsaturated; Hydroxymethylglutaryl CoA Reductases; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Hydroxymethylglutaryl-CoA Synthase; Lactones; Leupeptins; Lovastatin; Mevalonic Acid; Peroxisomes; Phosphorylation; Simvastatin; Tritium | 2000 |
Pervanadate-induced nuclear factor-kappaB activation requires tyrosine phosphorylation and degradation of IkappaBalpha. Comparison with tumor necrosis factor-alpha.
Tumor necrosis factor activates nuclear transcription factor kappaB (NF-kappaB) by inducing serine phosphorylation of the inhibitory subunit of NF-kappaB (IkappaBalpha), which leads to its ubiquitination and degradation. In contrast, pervanadate (PV) activates NF-kappaB and induces tyrosine phosphorylation of IkappaBalpha (Singh, S., Darney, B. G., and Aggarwal, B. B. (1996) J. Biol. Chem. 271, 31049-31054; Imbert, V., Rupec, R. A., Antonia, L., Pahl, H. L., Traenckner, E. B.-M., Mueller-Dieckmann, C., Farahifar, D., Rossi, B., Auderger, P., Baeuerle, P. A., and Peyron, J.-F. (1996) Cell 86, 787-798). Whether PV also induces IkappaBalpha degradation and whether degradation is required for NF-kappaB activation are not understood. We investigated the effect of PV-induced tyrosine phosphorylation on IkappaBalpha degradation and NF-kappaB activation. PV activated NF-kappaB, as determined by DNA binding, NF-kappaB-dependent reporter gene expression, and phosphorylation and degradation of IkappaBalpha. Maximum degradation of IkappaBalpha occurred at 180 min, followed by NF-kappaB-dependent IkappaBalpha resynthesis. N-Acetylleucylleucylnorlucinal, a proteasome inhibitor, blocked both IkappaBalpha degradation and NF-kappaB activation, suggesting that the IkappaBalpha degradation is required for NF-kappaB activation. PV did not induce serine phosphorylation of IkappaBalpha but induced phosphorylation at tyrosine residue 42. Unlike tumor necrosis factor (TNF), PV did not induce ubiquitination of IkappaBalpha. Like TNF, however, PV induced phosphorylation and degradation of IkappaBalpha, and subsequent NF-kappaB activation, which could be blocked by N-tosyl-L-phenylalanine chloromethyl ketone, calpeptin, and pyrrolidine dithiocarbomate, suggesting a close link between PV-induced NF-kappaB activation and IkappaBalpha degradation. Overall, our studies demonstrate that PV activates NF-kappaB, which, unlike TNF, requires tyrosine phosphorylation of IkappaBalpha and its degradation. Topics: Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; HeLa Cells; Humans; I-kappa B Proteins; Leupeptins; Multienzyme Complexes; NF-kappa B; NF-KappaB Inhibitor alpha; Phosphorylation; Proteasome Endopeptidase Complex; Protein Tyrosine Phosphatases; Transcription, Genetic; Tumor Necrosis Factor-alpha; Tyrosine; U937 Cells; Vanadates | 2000 |
Accumulation and degradation in the endoplasmic reticulum of a truncated ER-60 devoid of C-terminal amino acid residues.
The accumulation and degradation in the endoplasmic reticulum (ER) of a truncated ER-60 protease, from which the C-terminal 89 amino acid residues have been deleted (K 417 ochre), was examined. K 417 ochre overexpressed in COS-1 cells is not secreted into the medium, but accumulates as insoluble aggregates in non-ionic detergent without degradation in unusual clump membrane structures. K 417 ochre, stably expressed, forms soluble aggregates in non-ionic detergent and is distributed in the reticular structures of ER. Under these conditions, K 417 ochre is not secreted into the medium but is degraded with a half-life time of more than 8 h. Since K 417 ochre/C all S, in which all the Cys residues of K 417 ochre are replaced by Ser, also forms aggregates, an inter-disulfide bond appears unnecessary for aggregation. In both types of aggregates, Ig heavy chain binding protein, calnexin, glucose regulated protein 94, calreticulin, ERp72, and protein disulfide isomerase are scarcely found. Since degradation of the stably expressed K 417 ochre was not inhibited by lactacystin, leupeptin, NH(4)Cl, or cytocharasin B, but was inhibited by N-acetyl-leucyl-leucyl-norleucinal, the self-aggregated abnormal protein in the lumen of ER is assumed to be degraded by an unknown protease system other than proteasome, lysosome or autophagy. Topics: Acetylcysteine; Animals; Calcium-Binding Proteins; Calnexin; Calreticulin; Carrier Proteins; Cell Membrane; COS Cells; Culture Media; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytochalasin B; Endoplasmic Reticulum; Endoplasmic Reticulum Chaperone BiP; Half-Life; Heat-Shock Proteins; HSP70 Heat-Shock Proteins; Leupeptins; Membrane Proteins; Molecular Chaperones; Protein Disulfide-Isomerases; Recombinant Proteins; Ribonucleoproteins | 2000 |
Caspases cleave the amino-terminal calpain inhibitory unit of calpastatin during apoptosis in human Jurkat T cells.
We have previously reported the activation of procalpain mu (precursor for low-calcium-requiring calpain) in apoptotic cells using a cleavage-site-directed antibody specific to active calpain [Kikuchi, H. and Imajoh-Ohmi, S. (1995) Cell Death Differ. 2, 195-199]. In this study, calpastatin, the endogenous inhibitor protein for calpain, was cleaved to a 90-kDa polypeptide during apoptosis in human Jurkat T cells. The limited proteolysis of calpastatin preceded the autolytic activation of procalpain. Inhibitors for caspases rescued the cells from apoptosis and simultaneously inhibited the cleavage of calpastatin. The full-length recombinant calpastatin was also cleaved by caspase-3 or caspase-7 at Asp-233 into the same size fragment. Cys-241 was also targeted by these caspases in vitro but not in apoptotic cells. Caspase-digested calpastatin lost its amino-terminal inhibitory unit, and inhibited three moles of calpain per mole. Our findings suggest that caspases trigger the decontrol of calpain activity suppression by degrading calpastatin. Topics: Amino Acid Chloromethyl Ketones; Antibodies, Monoclonal; Apoptosis; Aspartic Acid; Calcium-Binding Proteins; Calpain; Caspase Inhibitors; Caspases; Cysteine Proteinase Inhibitors; fas Receptor; Humans; Jurkat Cells; Leupeptins; Oligopeptides; Poly(ADP-ribose) Polymerases; Recombinant Proteins; Tumor Necrosis Factor-alpha | 2000 |
Mitotic clonal expansion during preadipocyte differentiation: calpain-mediated turnover of p27.
Evidence is presented that calpain, a calcium-activated protease, degrades the cyclin-dependent kinase inhibitor, p27, during the mitotic clonal expansion phase of 3T3-L1 preadipocyte differentiation. Calpain activity is required during an early stage of the adipocyte differentiation program. Thus, inhibition of calpain with N-acetyl-Leu-Leu-norleucinal (ALLN) blocks clonal expansion and acquisition of the adipocyte phenotype only when added between 12 and 24 h after the induction of differentiation. Likewise, inhibition of calpain by overexpression of calpastatin, the specific endogenous inhibitor of calpain, prevents 2-day post-confluent preadipocytes from reentering the cell cycle triggered by the differentiation inducers. Inhibition of calpain with ALLN causes preadipocytes to arrest just prior to S phase and prevents phosphorylation of the retinoblastoma gene product, DNA replication, clonal expansion, and subsequent adipocyte differentiation but does not affect the expression of immediate early genes (i.e. fos, jun, C/EBPbeta, and C/EBPdelta). Inhibition of calpain by either ALLN or by overexpression of calpastatin blocks the degradation of p27. p27 is degraded in vitro by cell-free extracts from clonally expanding preadipocytes that contain "active" calpain but not by extracts from pre-mitotic preadipocytes that do not. This action is inhibited by calpastatin or ALLN. Likewise, p27 in preadipocyte extracts is a substrate for purified calpain; this proteolytic action was inhibited by heat inactivation, EGTA, or ALLN. Thus, extracellular signals from the differentiation inducers appear to activate calpain, which degrades p27 allowing density-dependent inhibited preadipocytes to reenter the cell cycle and undergo mitotic clonal expansion. Topics: 3T3 Cells; Adipocytes; Animals; Calpain; Cell Cycle Proteins; Cell Differentiation; Clone Cells; Cyclin-Dependent Kinase Inhibitor p27; Cyclin-Dependent Kinases; Cysteine Proteinase Inhibitors; Cytoplasm; Gene Expression Regulation; Genes, Immediate-Early; Kinetics; Leupeptins; Mice; Microtubule-Associated Proteins; Mitosis; Recombinant Proteins; Time Factors; Transfection; Triglycerides; Tumor Suppressor Proteins | 2000 |
Differential localization and regulation of death and decoy receptors for TNF-related apoptosis-inducing ligand (TRAIL) in human melanoma cells.
Induction of apoptosis in cells by TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF family, is believed to be regulated by expression of two death-inducing and two inhibitory (decoy) receptors on the cell surface. In previous studies we found no correlation between expression of decoy receptors and susceptibility of human melanoma cells to TRAIL-induced apoptosis. In view of this, we studied the localization of the receptors in melanoma cells by confocal microscopy to better understand their function. We show that the death receptors TRAIL-R1 and R2 are located in the trans-Golgi network, whereas the inhibitory receptors TRAIL-R3 and -R4 are located in the nucleus. After exposure to TRAIL, TRAIL-R1 and -R2 are internalized into endosomes, whereas TRAIL-R3 and -R4 undergo relocation from the nucleus to the cytoplasm and cell membranes. This movement of decoy receptors was dependent on signals from TRAIL-R1 and -R2, as shown by blocking experiments with Abs to TRAIL-R1 and -R2. The location of TRAIL-R1, -R3, and -R4 in melanoma cells transfected with cDNA for these receptors was similar to that in nontransfected cells. Transfection of TRAIL-R3 and -R4 increased resistance of the melanoma lines to TRAIL-induced apoptosis even in melanoma lines that naturally expressed these receptors. These results indicate that abnormalities in "decoy" receptor location or function may contribute to sensitivity of melanoma to TRAIL-induced apoptosis and suggest that further studies are needed on the functional significance of their nuclear location and TRAIL-induced movement within cells. Topics: Amino Acid Chloromethyl Ketones; Apoptosis; Apoptosis Regulatory Proteins; Brefeldin A; Cell Nucleus; Cysteine Proteinase Inhibitors; DNA, Complementary; GPI-Linked Proteins; Humans; Leupeptins; Ligands; Melanoma; Membrane Glycoproteins; Protein Synthesis Inhibitors; Receptors, TNF-Related Apoptosis-Inducing Ligand; Receptors, Tumor Necrosis Factor; Receptors, Tumor Necrosis Factor, Member 10c; Signal Transduction; Subcellular Fractions; TNF-Related Apoptosis-Inducing Ligand; Transfection; Tumor Cells, Cultured; Tumor Necrosis Factor Decoy Receptors; Tumor Necrosis Factor-alpha | 2000 |
Keratins turn over by ubiquitination in a phosphorylation-modulated fashion.
Keratin polypeptides 8 and 18 (K8/18) are intermediate filament (IF) proteins that are expressed in glandular epithelia. Although the mechanism of keratin turnover is poorly understood, caspase-mediated degradation of type I keratins occurs during apoptosis and the proteasome pathway has been indirectly implicated in keratin turnover based on colocalization of keratin-ubiquitin antibody staining. Here we show that K8 and K18 are ubiquitinated based on cotransfection of His-tagged ubiquitin and human K8 and/or K18 cDNAs, followed by purification of ubiquitinated proteins and immunoblotting with keratin antibodies. Transfection of K8 or K18 alone yields higher levels of keratin ubiquitination as compared with cotransfection of K8/18, likely due to stabilization of the keratin heteropolymer. Most of the ubiquitinated species partition with the noncytosolic keratin fraction. Proteasome inhibition stabilizes K8 and K18 turnover, and is associated with accumulation of phosphorylated keratins, which indicates that although keratins are stable they still turnover. Analysis of K8 and K18 ubiquitination and degradation showed that K8 phosphorylation contributes to its stabilization. Our results provide direct evidence for K8 and K18 ubiquitination, in a phosphorylation modulated fashion, as a mechanism for regulating their turnover and suggest that other IF proteins could undergo similar regulation. These and other data offer a model that links keratin ubiquitination and hyperphosphorylation that, in turn, are associated with Mallory body deposits in a variety of liver diseases. Topics: Cell Line; Cysteine Endopeptidases; Enzyme Inhibitors; Humans; Intermediate Filament Proteins; Keratins; Leupeptins; Multienzyme Complexes; Mutation; Phosphorylation; Proteasome Endopeptidase Complex; Transfection; Ubiquitins | 2000 |
Inhibiting amyloid precursor protein C-terminal cleavage promotes an interaction with presenilin 1.
Presenilin 1 (PS1) plays a pivotal role in the production of the amyloid-beta protein, which is central to the pathogenesis of Alzheimer's disease. It has been demonstrated that PS1 regulates the gamma-secretase proteolysis of the amyloid precursor protein (APP) C-terminal fragment (APP-C100), which is the final step in amyloid-beta protein production. The mechanism and detailed pathway of this PS1 activity has yet to be fully resolved, but it may be due to a presenilin-controlled trafficking of the APP fragment or possibly an inherent PS1 proteolytic activity. We have investigated the possibility of a direct interaction of PS1 and the APP-C100 within the high molecular mass presenilin complex. However, the APP-C100 is rapidly degraded, and if it forms, then any PS1.APP complex is likely to be very transitory. To circumvent this problem, we have utilized the protease inhibitor N-acetyl-leucyl-norleucinal (LLnL) and the lysosomotropic agent NH(4)Cl, which inhibits the turnover of the APP-C100. Under these conditions, levels of the fragment increased appreciably, and as shown by glycerol gradient analysis, the APP-C100 shifted to a higher molecular mass complex that overlapped with PS1. Immunoprecipitation studies demonstrated that a significant population of the APP-C100 co-precipitated with PS1. These findings suggest that PS1 may mediate the shuttling of APP fragments and/or facilitate their presentation for gamma-secretase cleavage through a direct interaction. Topics: Alzheimer Disease; Ammonium Chloride; Amyloid beta-Protein Precursor; Amyloid Precursor Protein Secretases; Animals; Aspartic Acid Endopeptidases; Cell Line; Centrifugation, Density Gradient; Cysteine Proteinase Inhibitors; Dogs; Endopeptidases; Humans; Leupeptins; Membrane Proteins; Peptide Fragments; Precipitin Tests; Presenilin-1; Transfection | 2000 |
Proteasome-mediated degradation of the papillomavirus E2-TA protein is regulated by phosphorylation and can modulate viral genome copy number.
The bovine papillomavirus E2 proteins regulate viral transcription, replication, and episomal genome maintenance. We have previously mapped the major phosphorylation sites of the E2 proteins to serine residues 298 and 301 and shown that mutation of serine residue 301 to alanine leads to a dramatic (10- to 20-fold) increase in viral DNA copy number. In this study we analyzed how phosphorylation regulates E2 protein function. S301 is located in a PEST sequence; these sequences are often found in proteins with a short half-life and can be regulated by phosphorylation. We show here that the E2 protein is ubiquitinated and degraded by the proteasome. Mutation of serine 301 to alanine increases the half-life of E2 from approximately 50 min to 160 min. Furthermore, the A301 E2 protein shows greatly reduced ubiquitination and degradation by the proteasome. These results suggest that the E2 protein level is regulated by phosphorylation, which in turn determines viral episomal copy number. Topics: Alanine; Animals; Bovine papillomavirus 1; Cattle; Cell Line; Chlorocebus aethiops; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; DNA, Viral; Gene Dosage; Genome, Viral; Lactones; Leupeptins; Multienzyme Complexes; Mutagenesis, Site-Directed; Papilloma; Papillomavirus Infections; Phosphorylation; Proteasome Endopeptidase Complex; Serine; Viral Proteins; Warts | 2000 |
Inhibition of translocation of nascent apolipoprotein B across the endoplasmic reticulum membrane is associated with selective inhibition of the synthesis of apolipoprotein B.
In HepG2 cells, inhibition of apolipoprotein B100 (apoB) translocation across the endoplasmic reticulum by an microsomal triglyceride transfer protein (MTP) inhibitor (CP-10447) in the presence of N-acetyl-leucinyl-norleucinal, a proteasomal inhibitor, results in accumulation of newly synthesized apoB in the translocation channel. Here we demonstrated that such accumulation led to a specific reduction of apoB synthesis. ApoB mRNA levels remained unchanged, but we observed reduced rates of elongation of nascent apoB in puromycin-synchronized cells pretreated with MTP inhibitor. This observation was consistent with a longer half-ribosome transit time for the synthesis of apoB in MTP-inhibited cells. Initiation of translation of apoB mRNA was not impaired by MTP inhibition. Overall, these findings suggest that translocation arrest of apoB in the endoplasmic reticulum channel can exert a selective and negative effect on the synthesis of apoB at the stage of elongation. Topics: Apolipoproteins B; Biological Transport; Carrier Proteins; Cell Line; Endoplasmic Reticulum; Humans; Leupeptins; Methaqualone; Ribosomes; RNA, Messenger | 2000 |
The docking protein HEF1 is an apoptotic mediator at focal adhesion sites.
HEF1 (human enhancer of filamentation 1) is a member of a docking protein family that includes p130(Cas) and Efs. Through assembly of multiple protein interactions at focal adhesion sites, these proteins activate signaling cascades in response to integrin receptor binding of the extracellular matrix. The HEF1 protein is cell cycle regulated, with full-length forms cleaved in mitosis at a caspase consensus site to generate an amino-terminal 55-kDa form that localizes to the mitotic spindle. The identification of a caspase cleavage site in HEF1 led us to investigate whether HEF1 belongs to a select group of caspase substrates cleaved in apoptosis to promote the morphological changes characteristic of programmed cell death. Significantly, inducing expression of HEF1 in MCF-7 or HeLa cells causes extensive apoptosis, as assessed by multiple criteria. Endogenous HEF1 is cleaved into 65- and 55-kDa fragments and a newly detected 28-kDa form in response to the induction of apoptosis, paralleling cleavage of poly(ADP-ribose) polymerase and focal adhesion kinase (FAK); the death-promoting activity of over-expressed HEF1 is associated with production of the 28-kDa form. While the generation of the cleaved HEF1 forms is caspase dependent, the accumulation of HEF1 forms is further regulated by the proteasome, as the proteasome inhibitors N-acetyl-L-leucinyl-L-leucinyl-L-norleucinyl and lactacystin enhance their stability. Finally, the induction of HEF1 expression also increases Jun N-terminal protein kinase (JNK) activation, and activated JNK colocalizes with HEF1, implicating this pathway in HEF1 action. Based on these results, we propose that dysregulation of HEF1 and its family members along with FAK may signal the destruction of focal adhesion sites and regulate the onset of apoptosis. Topics: Acetylcysteine; Adaptor Proteins, Signal Transducing; Apoptosis; Caspase 3; Caspase 7; Caspase Inhibitors; Caspases; Cell Death; Crk-Associated Substrate Protein; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Focal Adhesion Kinase 1; Focal Adhesion Protein-Tyrosine Kinases; HeLa Cells; Humans; JNK Mitogen-Activated Protein Kinases; Leupeptins; Mitogen-Activated Protein Kinases; Mitosis; Multienzyme Complexes; Mutation; Oligopeptides; Peptide Fragments; Phosphoproteins; Poly(ADP-ribose) Polymerases; Proteasome Endopeptidase Complex; Protein Isoforms; Protein-Tyrosine Kinases; Proteins; Retinoblastoma-Like Protein p130; Signal Transduction; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha | 2000 |
Aldehyde-mannan antigen complexes target the MHC class I antigen-presentation pathway.
Antigens such as MUC1 coupled to oxidized mannan lead to rapid and efficient MHC class I presentation to CD8+ cells and a preferential T1 response; after reduction there is class II presentation and a T2 immune response. We now show that the selective advantage of the oxidized mannan-MUC1 is due to the presence of aldehydes and not Schiff bases, and that oxidized mannan-MUC1 binds to the mannose and not scavenger receptors and is internalized and presented by MHC class I molecules 1,000 times more efficiently than when reduced. After internalization there is rapid access to the class I pathway via endosomes but not lysosomes, proteasomal processing and transport to the endoplasmic reticulum, Golgi apparatus and cell surface. Aldehydes cause rapid entry into the class I pathway, and can therefore direct the subsequent immune response. Topics: Aldehydes; Animals; Antigen Presentation; Calpain; Cathepsin B; Cells, Cultured; CHO Cells; Cricetinae; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Endosomes; Histocompatibility Antigens Class I; Humans; Lectins, C-Type; Leupeptins; Lysosomes; Macrophages, Peritoneal; Mannans; Mannose Receptor; Mannose-Binding Lectins; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Mice, Inbred DBA; Mucin-1; Multienzyme Complexes; Oxidation-Reduction; Peptide Fragments; Proteasome Endopeptidase Complex; Receptors, Cell Surface; Schiff Bases | 2000 |
Protease inhibitor-induced apoptosis: accumulation of wt p53, p21WAF1/CIP1, and induction of apoptosis are independent markers of proteasome inhibition.
Inhibitors of proteases are currently emerging as a potential anti-cancer modality. Nonselective protease inhibitors are cytotoxic to leukemia and cancer cell lines and we found that this cytotoxicity is correlated with their potency as inhibitors of the proteasome but not as inhibitors of calpain and cathepsin. Highly selective inhibitors of the proteasome were more cytotoxic and fast-acting than less selective inhibitors (PS341>>ALLN>>ALLM). Induction of wt p53 correlated with inhibition of the proteasome and antiproliferative effect in MCF7, a breast cancer cell line, which was resistant to apoptosis caused by proteasome inhibitors. In contrast, inhibitors of the proteasome induced apoptosis in four leukemia cell lines lacking wt p53. The order of sensitivity of leukemia cells was: Jurkat>HL60> or =U937>>K562. The highly selective proteasome inhibitor PS-341 induced cell death with an IC50 as low as 5 nM in apoptosis-prone leukemia cells. Cell death was preceded by p21WAF1/CIP1 accumulation, an alternative marker of proteasome inhibition, and by cleavage of PARP and Rb proteins and nuclear fragmentation. Inhibition of caspases abrogated PARP cleavage and nuclear fragmentation and delayed, but did not completely prevent cell death caused by PS-341. Reintroduction of wt p53 into p53-null PC3 prostate carcinoma cells did not increase their sensitivity to proteasome inhibitors. Likewise, comparison of parental and p21-deficient cells demonstrated that p21WAF1/CIP1 was dispensable for proteasome inhibitor-induced cytotoxicity. We conclude that accumulation of wt p53 and induction of apoptosis are independent markers of proteasome inhibition. Topics: Acetylcysteine; Acrylates; Amino Acid Chloromethyl Ketones; Apoptosis; Boronic Acids; Bortezomib; Calpain; Cathepsins; Cell Division; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Cysteine Endopeptidases; Drug Synergism; Genes, p53; Humans; Jurkat Cells; Leupeptins; Multienzyme Complexes; Neoplasm Proteins; Neoplasms; Oligopeptides; Protease Inhibitors; Proteasome Endopeptidase Complex; Pyrazines; Tumor Cells, Cultured; Tumor Suppressor Protein p53; U937 Cells | 2000 |
Prevention of etoposide-induced apoptosis by proteasome inhibitors in a human leukemic cell line but not in fresh acute leukemia blasts. A differential role of NF-kappab activation.
Recent research indicates that the proteasome is one of the non-caspase proteases involved in apoptotic signaling pathways. Nuclear factor-kappaB (NF-kappaB) activation, one of the key factors in apoptosis, can be prevented through abrogation of IkappaBalpha degradation by proteasome inhibition. We have investigated the effects of the proteasome inhibitors carbobenzoxyl-L-leucyl-L-leucyl-L-leucinal (MG132) and N-acetyl-L-leucinyl-L-leucinyl-L-norleucinal (LLnL) on apoptosis and NF-kappaB activation induced by etoposide, using a human leukemia cell line (U937) and leukemia blasts freshly isolated from patients with acute leukemia. Pretreatment of U937 cells with MG132 or LLnL inhibited etoposide-induced morphological apoptosis and caspase-3 activation. Furthermore, MG132 or LLnL prevented NF-kappaB activation and IkappaBalpha degradation, but not IkappaBalpha phosphorylation at Ser32. Other inhibitors of NF-kappaB activation, including pyrrrolidine dithiocarbamate (an antioxidant) and the peptide SN50 (an inhibitor of translocation of activated NF-kappaB into the nucleus), also attenuated etoposide-induced apoptosis. In leukemia blasts, although proteasome inhibitors suppressed NF-kappaB activation induced by etoposide, they were unable to prevent morphological apoptosis. Moreover, proteasome inhibitors by themselves caused apoptosis in leukemia blasts at the concentrations employed in this study. These results suggest that the role that NF-kappaB plays in apoptosis induced by etoposide in a human leukemia cell line may be different from the role it plays in freshly isolated leukemia blasts. Topics: Antineoplastic Agents, Phytogenic; Apoptosis; Caspase 3; Caspases; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Etoposide; Humans; I-kappa B Proteins; Leupeptins; Multienzyme Complexes; NF-kappa B; NF-KappaB Inhibitor alpha; Phosphorylation; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Proteasome Endopeptidase Complex; Tumor Cells, Cultured; U937 Cells | 2000 |
The ubiquitin-proteasome pathway mediates the regulated degradation of mammalian 3-hydroxy-3-methylglutaryl-coenzyme A reductase.
3-Hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR), the key regulatory enzyme in the mevalonate (MVA) pathway, is rapidly degraded in mammalian cells supplemented with sterols or MVA. This accelerated turnover was blocked by N-acetyl-leucyl-leucyl-norleucinal (ALLN), MG-132, and lactacystin, and to a lesser extent by N-acetyl-leucyl-leucyl-methional (ALLM), indicating the involvement of the 26 S proteasome. Proteasome inhibition led to enhanced accumulation of high molecular weight polyubiquitin conjugates of HMGR and of HMGal, a chimera between the membrane domain of HMGR and beta-galactosidase. Importantly, increased amounts of polyubiquitinated HMGR and HMGal were observed upon treating cells with sterols or MVA. Cycloheximide inhibited the sterol-stimulated degradation of HMGR concomitantly with a marked reduction in polyubiquitination of the enzyme. Inhibition of squalene synthase with zaragozic acid blocked the MVA- but not sterol-stimulated ubiquitination and degradation of HMGR. Thus, similar to yeast, the ubiquitin-proteasome pathway is involved in the metabolically regulated turnover of mammalian HMGR. Yet, the data indicate divergence between yeast and mammals and suggest distinct roles for sterol and nonsterol metabolic signals in the regulated ubiquitination and degradation of mammalian HMGR. Topics: Animals; beta-Galactosidase; Bridged Bicyclo Compounds, Heterocyclic; Cell Line; Cholesterol; Cricetinae; Cycloheximide; Cysteine Proteinase Inhibitors; Farnesyl-Diphosphate Farnesyltransferase; Humans; Hydroxycholesterols; Hydroxymethylglutaryl CoA Reductases; Leupeptins; Lovastatin; Oligopeptides; Peptide Hydrolases; Precipitin Tests; Proteasome Endopeptidase Complex; Recombinant Fusion Proteins; Tricarboxylic Acids; Ubiquitins | 2000 |
Activation-dependent degradation of protein kinase C eta.
Prolonged activation of protein kinase Cs (PKCs) by long-term treatment of cells with phorbol ester tumor promoters down-regulates the expression of many PKCs. To investigate the molecular mechanisms involved in the down-regulation of PKC eta, we expressed the novel PKCs eta and θ and various mutant forms in baby hamster kidney cells. Upon overexpression, constitutively active PKC eta, but not wild type or kinase-dead PKC eta, underwent rapid degradation to generate several lower molecular weight polypeptides. When co-expressed with active kinases, kinase-dead PKC eta with a pseudosubstrate site mutation designed to give an active conformation was down-regulated while the wild type PKC eta was not. These results suggest requirements for kinase activity and an active conformation for down-regulation of PKC eta. Treatment with the proteasome inhibitors N-Ac-Leu-Leu-norleucinal and lactacystin led to accumulation of PKC eta proteolytic products and potentially ubiquitinated forms. While wild type PKC eta localizes mostly to the detergent-soluble fraction of the cell, a significant portion of full-length constitutively active PKC eta and of kinase-dead, active conformation PKC eta were found in the detergent-insoluble fraction. Several proteolytic fragments of constitutively active PKC eta also were found in the detergent insoluble fraction. These full-length and proteolytic fragments of PKC eta in the detergent-insoluble fraction accumulated further in the presence of proteasome inhibitors. These data suggest that active conformation PKC eta accumulates in the detergent-insoluble compartment, is degraded by proteolysis in the presence of kinase activity, and that the cleavage products undergo further degradation via ubiquitin-mediated degradation in the proteasome. Oncogene (2000) 19, 4263 - 4272 Topics: Acetylcysteine; Animals; Cell Fractionation; Cell Line; Chick Embryo; Cricetinae; Cysteine Endopeptidases; Detergents; Enzyme Activation; Enzyme Inhibitors; Genetic Vectors; Humans; Isoenzymes; Kidney; Leupeptins; Membrane Proteins; Mesocricetus; Mice; Multienzyme Complexes; Mutagenesis, Site-Directed; Proteasome Endopeptidase Complex; Protein Conformation; Protein Kinase C; Protein Kinase C-theta; Rats; Recombinant Fusion Proteins; Sindbis Virus; Tumor Cells, Cultured; Ubiquitins | 2000 |
Ganglioside GD3 and its mimetics induce cytochrome c release from mitochondria.
Ganglioside GD3 induced the release of cytochrome c from isolated rat liver mitochondria. This process was completely prevented by cyclosporin A and partially prevented by a cysteine protease inhibitor, n-acetyl-leu-leu-norleucinal. Cyclosporin A is a potent inhibitor of the permeability transition pore, whereas n-acetyl-leu-leu-norleucinal has no effect on this pore. These results indicate that the release of cytochrome c from mitochondria requires both the opening of the permeability transition pore and a cysteine protease inhibitor-sensitive mechanism. Gangliosides GD1a, GD1b, GT1b, and GQ1b along with the synthetic GD3 mimetics TMS-42 and CI-22, which are glycerophospholipids carrying a disialo residue, also induced cytochrome c release. In contrast, gangliosides GM1, GM2, and GM3 did not induce cytochrome c release. These results indicate that two sialo residues must play an important role in the induction of cytochrome c release by gangliosides. Topics: Animals; Blotting, Western; Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone; Cell-Free System; Cyclosporine; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytochrome c Group; Dose-Response Relationship, Drug; Egtazic Acid; Gangliosides; Ion Channels; Leupeptins; Membrane Proteins; Mitochondria, Liver; Mitochondrial Membrane Transport Proteins; Mitochondrial Permeability Transition Pore; Molecular Mimicry; Oligomycins; Rats | 2000 |
Inhibition of NF-kappaB by IkappaB prevents cytokine-induced NO production and promotes enterocyte apoptosis in vitro.
Nuclear factor-kappaB (N-kappaB) plays a key role in gut inflammation. NF-kappaB up-regulates proinflammatory genes encoding cytokines, adhesion molecules, and inducible nitric oxide synthase (iNOS). However, NF-kappaB has also been shown to up-regulate protective or anti-apoptotic factors. We utilized an adenoviral vector carrying a super-repressor form of the inhibitor of NF-kappaB, IkappaB, to examine the effects of NF-kappaB inhibition on cytokine-induced nitric oxide production and apoptosis in rat small intestinal epithelial cells (IEC-6). Chemical inhibitors of NF-kappaB, including pyrrolidine dithiocarbamate (PDTC), tosyl-lysine-chloromethylketone (TLCK), genistein, and N-acetyl-leu-leu-norleucinal (n-LLnL) were also utilized. Treatment of AdIkappaB-transfected cells with cytomix [1000 U/mL IFN-gamma, 1 nM IL-1beta, and 10 ng/mL tumor necrosis factor alpha (TNFalpha)] or TNFalpha-containing cytokine combinations resulted in inhibition of cytokine-induced nitrite production and a marked increase in apoptosis compared to control cells. Apoptosis occurred independently of nitric oxide (NO) production since exogenous sources of NO did not inhibit apoptosis. Inducible NOS and clAP were down-regulated in AdIkappaB-transfected cells treated with cytomix. TLCK and LLnL treatment also induced apoptosis in cytomix-treated cells, while PDTC and genistein did not. Thus, although NF-kappaB up-regulates various pro-inflammatory genes, it may also have protective or anti-apoptotic effects in enterocytes. NF-kappaB appears necessary for upregulating cIAP in IEC-6 cells upon cytokine exposure. Topics: Animals; Apoptosis; Cells, Cultured; Cysteine Endopeptidases; Cytokines; Enterocytes; Enzyme Inhibitors; Genistein; I-kappa B Proteins; Leupeptins; Multienzyme Complexes; Neoplasm Proteins; NF-kappa B; Nitric Oxide; Nitric Oxide Donors; Nitric Oxide Synthase; Nitric Oxide Synthase Type II; Nitrites; Penicillamine; Proteasome Endopeptidase Complex; Protein Transport; Pyrrolidines; Rats; Recombinant Proteins; Thiocarbamates; Tosyllysine Chloromethyl Ketone; Up-Regulation | 2000 |
HLA-B polymorphism affects interactions with multiple endoplasmic reticulum proteins.
To explore the nature of amino acid substitutions that influence association with TAP, we compared a site-directed mutant of HLA-B*0702 (Y116D) to unmutated HLA-B7 in regard to TAP interaction. We found that the mutant had stronger association with TAP, and, in addition, with tapasin and calreticulin. These data confirm the importance of position 116 for TAP association, and indicate that (1) an aspartic acid at the 116 position can facilitate the interaction, and (2) association with tapasin and calreticulin is affected along with TAP. Furthermore, we tested three natural subtypes of HLA-B15, and found that a B15 subtype with a tyrosine at position 116 (B*1510) was strongly associated not only with TAP, but also with tapasin and calreticulin. In contrast, two B15 subtypes with a serine at position 116 (B*1518 and B*1501) exhibited very little or no association with any of these proteins. Thus, very closely related HLA-B subtypes can differ in regard to interaction with the entire assembly complex. Interestingly, when their surface expression was tested by flow cytometry, the HLA-B15 subtypes with little to no detectable intracellular assembly complex association had a slightly, yet consistently, higher level of the open heavy chain form than did the B15 subtype with intracellular assembly complex association. These data suggest that the relatively low strength or short length of interaction between endoplasmic reticulum proteins and natural HLA class I molecules can decrease their surface stability. Topics: Amino Acid Substitution; Animals; Antibodies, Monoclonal; Antiporters; Aspartic Acid; ATP Binding Cassette Transporter, Subfamily B, Member 2; ATP-Binding Cassette Transporters; Calcium-Binding Proteins; Calreticulin; Cysteine Endopeptidases; Endoplasmic Reticulum; HLA-B7 Antigen; Humans; Immunoglobulin G; Immunoglobulins; Leupeptins; Membrane Transport Proteins; Multienzyme Complexes; Mutagenesis, Site-Directed; Polymorphism, Genetic; Protease Inhibitors; Proteasome Endopeptidase Complex; Protein Binding; Rabbits; Ribonucleoproteins; Serine; Tyrosine | 2000 |
Modulation of hepatic lipoprotein synthesis and secretion by taxifolin, a plant flavonoid.
In the present study, the effects of taxifolin, a plant flavonoid, on lipid, apolipoprotein B (apoB), and apolipoprotein A-I (apoA-I) synthesis and secretion were determined in HepG2 cells. Pretreatment of cells with (+/-)-taxifolin led to an inhibition of cholesterol synthesis in a dose- and time-dependent manner, with an 86 +/- 3% inhibition at 200 microM observed within 24 h. As to the mechanism underlying this inhibitory effect, taxifolin was shown to inhibit the activity of HMG-CoA reductase by 47 +/- 7%. In addition, cellular cholesterol esterification, and triacylglycerol and phospholipid syntheses, were also significantly suppressed in the presence of taxifolin. ApoA-I and apoB synthesis and secretion were then studied by pulse-chase experiments. ApoA-I secretion was found to increase by 36 +/- 10%. In contrast, an average reduction of 61 +/- 8% in labeled apoB in the medium was apparent with taxifolin. This effect on secretion appeared not to be exerted at the transcriptional level. Rather, the effect on apoB secretion was found to be exerted in the early stages of apoB degradation and to be sensitive to dithiothreitol (DTT) and insensitive to N-acetyl-leucyl-leucyl-norleucinal, suggesting a proteolytic pathway involving a DTT-sensitive protease. Fractionation of secreted apoB revealed a slight shift in the distribution of secreted apoB-containing lipoproteins. Cholesteryl ester, rather than triacylglycerol, was shown to be the lipid that primarily regulated apoB secretion. In summary, our data suggest that taxifolin decreases hepatic lipid synthesis with a concomitant decrease and increase in apoB and apoA-I secretion, respectively. Topics: Anticholesteremic Agents; Apolipoprotein A-I; Apolipoproteins B; Cholesterol; Dithiothreitol; Flavonols; Humans; Hydrolysis; Hydroxymethylglutaryl CoA Reductases; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Leupeptins; Liver; Quercetin; RNA, Messenger; Tumor Cells, Cultured | 2000 |
Identification of an apoptotic cleavage product of BARD1 as an autoantigen: a potential factor in the antitumoral response mediated by apoptotic bodies.
We have shown previously that rats can be cured from induced peritoneal colon carcinomatosis by injections of apoptotic bodies derived from tumor cells and interleukin 2. This curative treatment generated a tumor-specific cytotoxic T-cell response associated with a humoral response. Autoantibodies from sera of cured rats strongly recognized a Mr 67,000 protein from apoptotic bodies and weakly reacted with a protein of Mr approximately 97,000 in PROb parental cells. We now show that these autoantibodies are directed against BARD1, originally identified as a protein interacting with the product of the breast cancer gene 1, BRCA1. We demonstrate that the Mr 67,000 antigen is a cleaved form of BARD1 present in apoptotic bodies derived from rat and human colon and mammary carcinoma cell lines. Moreover, we show that the cleavage site of BARD1 is located NH2 terminally but downstream of the RING domain essential for BARD1 and BRCA1 protein interaction. In vitro studies using [35S]methionine-labeled human BARD1 and apoptotic cellular extracts derived from SW48 carcinoma cells indicate that BARD1 proteolysis occurs at an early stage of apoptosis and in a cell cycle-dependent manner. This hydrolysis is inhibited by EGTA, and the calpain inhibitor I, N-acetyl-leu-leu-norleucinal, but not by several caspases inhibitors, suggesting that BARD1 is hydrolyzed by the calcium-dependent cysteine proteases, calpains. Thus, the highly immunogenic form of cleaved BARD1 could contribute to the antitumoral response mediated by apoptotic bodies. Topics: Amino Acid Sequence; Animals; Antibodies, Monoclonal; Apoptosis; Autoantigens; Blotting, Western; BRCA1 Protein; Breast Neoplasms; Calpain; Carrier Proteins; Cell Cycle; Cell Fractionation; Cloning, Molecular; Colonic Neoplasms; Cysteine Proteinase Inhibitors; DNA, Complementary; Egtazic Acid; Enzyme Inhibitors; Gene Library; Humans; Leupeptins; Mammary Neoplasms, Animal; Mice; Molecular Sequence Data; Precipitin Tests; Protein Binding; Protein Structure, Tertiary; Rats; Sequence Homology, Amino Acid; Tumor Cells, Cultured; Tumor Suppressor Proteins; Ubiquitin-Protein Ligases | 2000 |
Translocation-arrested apolipoprotein B evades proteasome degradation via a sterol-sensitive block in ubiquitin conjugation.
In this study, we explored how sterol metabolism altered by the expression of cholesterol-7alpha-hydroxylase NADPH:oxygen oxidoreductase (7alpha-hydroxylase) affects the ubiquitin-dependent proteasome degradation of translocation-arrested apoB53 in Chinese hamster ovary cells. Stable expression of two different plasmids that encode either rat or human 7alpha-hydroxylase inhibited the ubiquitin conjugation of apoB and its subsequent degradation by the proteasome. Oxysterols (25-hydroxycholesterol and 7-ketocholesterol) reversed the inhibition of apoB degradation caused by 7alpha-hydroxylase. The combined results suggest that the normally rapid proteasome degradation of translocation-arrested apoB can be regulated by a sterol-sensitive polyubiquitin conjugation step in the endoplasmic reticulum. Blocked ubiquitin-dependent proteasome degradation caused translocation-arrested apoB to become sequestered in segregated membrane domains. Our results described for the first time a novel mechanism through which the "quality control" proteasome endoplasmic reticulum degradative pathway of translocation-arrested apoB is linked to sterol metabolism. Sterol-sensitive blocked ubiquitin conjugation appears to selectively inhibit the proteasome degradation of apoB, but not 7alpha-hydroxylase protein, with no impairment of cell vitality or function. Our findings may help to explain why the hepatic production of lipoproteins is increased when familial hypertriglyceridemic patients are treated with drugs that activate 7alpha-hydroxylase (e.g. bile acid-binding resins). Topics: Animals; Apolipoproteins B; Biological Transport; Carrier Proteins; Cells, Cultured; CHO Cells; Cholesterol 7-alpha-Hydroxylase; Cricetinae; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Endoplasmic Reticulum; Humans; Hydroxycholesterols; Leupeptins; Microsomes; Multienzyme Complexes; Proteasome Endopeptidase Complex; Rats; RNA, Messenger; Transfection; Ubiquitins | 1999 |
Photo-oxidative stress down-modulates the activity of nuclear factor-kappaB via involvement of caspase-1, leading to apoptosis of photoreceptor cells.
The mechanisms of photoreceptor cell death via apoptosis, in retinal dystrophies, are largely not understood. In the present report we show that visible light exposure of mouse cultured 661W photoreceptor cells at 4.5 milliwatt/cm2 caused a significant increase in oxidative damage of 661W cells, leading to apoptosis of these cells. These cells show constitutive expression of nuclear factor-kappaB (NF-kappaB), and light exposure of photoreceptor cells results in lowering of NF-kappaB levels in both the nuclear and cytosolic fractions in a time-dependent manner. Immunoblot analysis of IkappaBalpha and p50, and p65 (RelA) subunits of NF-kappaB, suggested that photo-oxidative stress results in their depletion. Immunocytochemical studies using antibody to RelA subunit of NF-kappaB further revealed the presence of this subunit constitutively both in the nucleus and cytoplasm of the 661W cells. Upon exposure to photo-oxidative stress, a depletion of the cytoplasmic and nuclear RelA subunit was observed. The depletion of NF-kappaB appears to be mediated through involvement of caspase-1. Furthermore, transfection of these cells with a dominant negative mutant IkappaBalpha greatly enhanced the kinetics of down modulation of NF-kappaB, resulting in a faster photo-oxidative stress-induced apoptosis. Taken together, these studies show that the presence of NF-kappaB RelA subunit in the nucleus is essential for protection of photoreceptor cells against apoptosis mediated by an oxidative pathway. Topics: Animals; Apoptosis; Base Sequence; Blotting, Western; Caspase 1; Cattle; Cell Line; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA Primers; Down-Regulation; Glutathione; Leupeptins; Light; Lipid Peroxidation; Mice; Mice, Transgenic; Multienzyme Complexes; NF-kappa B; Oxidative Stress; Photoreceptor Cells; Proteasome Endopeptidase Complex | 1999 |
Presecretory degradation of apolipoprotein [a] is mediated by the proteasome pathway.
Plasma levels of atherogenic lipoprotein [a] (Lp[a]) vary over a 1000-fold range and are largely determined by the gene for its unique glycoprotein, apolipoprotein [a] (apo[a]). The apo[a] locus comprises more than 100 alleles, encoding proteins from <300 to >800 kDa. Using primary baboon hepatocyte cultures, we previously demonstrated that differences in the secretion efficiency of apo[a] allelic variants contribute to the variation in plasma Lp[a] levels. In the current study, we investigated the mechanism of apo[a] presecretory degradation. The proteasome inhibitors, acetyl-leucyl-leucyl-norleucinal and lactacystin, prevented apo[a] degradation and increased apo[a] secretion. Transfection with an HA-tagged ubiquitin construct demonstrated the accumulation of ubiquitinated apo[a] in the presence of lactacystin. These results suggest a role for the cytoplasmic proteasome in apo[a] proteolysis. Apo[a] that accumulated intracellularly in the presence of lactacystin remained sensitive to endo-B-N-glucosaminidase H, and apo[a] degradation was reversibly inhibited by brefeldin A, suggesting that transport to a post-endoplasmic reticulum (ER) pre-medial Golgi compartment is required for apo[a] degradation. Newly synthesized apo[a] bound to the ER chaperone calnexin and conditions that enhanced this interaction prevented apo[a] degradation, suggesting that calnexin can protect apo[a] from proteolysis. These studies provide further support for the role of the proteasome in endoplasmic reticulum quality control, and expand this role to one that influences plasma levels of the atherogenic lipoprotein Lp[a].-White, A. L., B. Guerra, J. Wang, and R. E. Lanford. Presecretory degradation of apolipoprotein[a] is mediated by the proteasome pathway. Topics: Acetylcysteine; Animals; Apolipoproteins A; Calnexin; Cells, Cultured; Cysteine Endopeptidases; Electrophoresis, Polyacrylamide Gel; Endoplasmic Reticulum; Fabaceae; Hepatocytes; Hexosaminidases; Leupeptins; Mannosidases; Molecular Chaperones; Multienzyme Complexes; Papio; Precipitin Tests; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; Transfection; Ubiquitin | 1999 |
Role of calpain in adipocyte differentiation.
Evidence is presented that the calcium-activated protease, calpain, is required for differentiation of 3T3-L1 preadipocytes into adipocytes induced by methylisobutylxanthine (a cAMP phosphodiesterase inhibitor), dexamethasone, and insulin. Calpain is expressed by preadipocytes and its level falls during differentiation. Exposure of preadipocytes to the calpain inhibitor N-acetyl-Leu-Leu-norleucinal or overexpression of calpastatin, a specific endogenous inhibitor of calpain, blocks expression of adipocyte-specific genes, notably the CCAAT/enhancer-binding protein (C/EBP)alpha gene, and acquisition of the adipocyte phenotype. The inhibitor disrupts the differentiation-inducing effect of methylisobutylxanthine (by means of the cAMP-signaling pathway), but is without effect on differentiation induced by dexamethasone or insulin. N-acetyl-Leu-Leu-norleucinal, or overexpression of calpastatin, inhibits reporter gene expression mediated by the C/EBPalpha gene promoter by preventing C/EBPbeta, a transcriptional activator of the C/EBPalpha gene, from binding to the promoter. These findings implicate calpain in the transcriptional activation of the C/EBPalpha gene, a process required for terminal adipocyte differentiation. Topics: 1-Methyl-3-isobutylxanthine; 3T3 Cells; Adipocytes; Animals; Calcium-Binding Proteins; Calpain; CCAAT-Enhancer-Binding Proteins; Cell Differentiation; Dexamethasone; DNA-Binding Proteins; Insulin; Leupeptins; Mice; Nuclear Proteins; Recombinant Proteins; Transcription Factors; Transfection | 1999 |
Coupling assembly of the E-cadherin/beta-catenin complex to efficient endoplasmic reticulum exit and basal-lateral membrane targeting of E-cadherin in polarized MDCK cells.
The E-cadherin/catenin complex regulates Ca++-dependent cell-cell adhesion and is localized to the basal-lateral membrane of polarized epithelial cells. Little is known about mechanisms of complex assembly or intracellular trafficking, or how these processes might ultimately regulate adhesion functions of the complex at the cell surface. The cytoplasmic domain of E-cadherin contains two putative basal-lateral sorting motifs, which are homologous to sorting signals in the low density lipoprotein receptor, but an alanine scan across tyrosine residues in these motifs did not affect the fidelity of newly synthesized E-cadherin delivery to the basal-lateral membrane of MDCK cells. Nevertheless, sorting signals are located in the cytoplasmic domain since a chimeric protein (GP2CAD1), comprising the extracellular domain of GP2 (an apical membrane protein) and the transmembrane and cytoplasmic domains of E-cadherin, was efficiently and specifically delivered to the basal-lateral membrane. Systematic deletion and recombination of specific regions of the cytoplasmic domain of GP2CAD1 resulted in delivery of <10% of these newly synthesized proteins to both apical and basal-lateral membrane domains. Significantly, >90% of each mutant protein was retained in the ER. None of these mutants formed a strong interaction with beta-catenin, which normally occurs shortly after E-cadherin synthesis. In addition, a simple deletion mutation of E-cadherin that lacks beta-catenin binding is also localized intracellularly. Thus, beta-catenin binding to the whole cytoplasmic domain of E-cadherin correlates with efficient and targeted delivery of E-cadherin to the lateral plasma membrane. In this capacity, we suggest that beta-catenin acts as a chauffeur, to facilitate transport of E-cadherin out of the ER and the plasma membrane. Topics: Amino Acid Sequence; Animals; Base Sequence; beta Catenin; Binding Sites; Cadherins; Cell Adhesion; Cell Line; Cell Membrane; Cell Polarity; Chloroquine; Cytoplasm; Cytoskeletal Proteins; Dogs; Endoplasmic Reticulum; Leupeptins; Macromolecular Substances; Molecular Sequence Data; Plasmids; Rats; Recombinant Fusion Proteins; Trans-Activators | 1999 |
Dietary fish oils inhibit early events in the assembly of very low density lipoproteins and target apoB for degradation within the rough endoplasmic reticulum of hamster hepatocytes.
Dietary fish oils inhibited secretion and stimulated intracellular degradation of apolipoprotein (apo)B in hamster hepatocytes, while dietary sunflower oils stimulated secretion and had no effect on degradation of apoB. To investigate the intracellular site at which fish oils act, we have made use of our previous observations that inhibition of degradation by N-acetyl-leucyl-leucyl-norleucinal (ALLN) results in accumulation of apoB in the trans -Golgi membrane and does not stimulate secretion, while inhibition of degradation by o-phenanthroline results in accumulation of apoB in the rough endoplasmic reticulum membrane and stimulates secretion. Thus, ALLN protects apoB which has been diverted from secretion and o -phenanthroline protects apoB which is targetted for secretion. Addition of o -phenantholine to the incubation medium of hepatocytes from fish oil-fed hamsters inhibited degradation of apoB and stimulated its secretion in particles of the density of VLDL, while addition of ALLN had no effect. These observations suggest that dietary fish oils reversibly inhibit early steps in the assembly of very low density lipoprotein precursors and target apoB for degradation in the rough endoplasmic reticulum. Topics: Animals; Apolipoprotein B-100; Apolipoproteins B; Biomarkers; Cells, Cultured; Cricetinae; Dietary Fats, Unsaturated; Endoplasmic Reticulum, Rough; Fish Oils; Leupeptins; Lipoproteins, VLDL; Liver; Male; Oleic Acid; Phenanthrolines; Plant Oils; Protease Inhibitors | 1999 |
Complement component C3 production in IL-1beta-stimulated human intestinal epithelial cells is blocked by NF-kappaB inhibitors and by transfection with ser 32/36 mutant IkappaBalpha.
Recent studies suggest that interleukin-1beta (IL-1beta) stimulates the production of the acute phase protein complement component C3 in human intestinal epithelial cells. The transcription factor NF-kappaB activates different genes involved in the response to cytokines. It is not known if IL-1beta-induced C3 production in the enterocyte is regulated by NF-kappaB.. Cultured Caco-2 cells, a human intestinal epithelial cell line, were treated with one of the NF-kappaB inhibitors, tosyl-lys-chloromethylketone (TLCK), genistein, or pyrrolidine dithiocarbamate (PDTC), or with N-acetyl-leu-leu-norleucinal (LLnL), a proteasome inhibitor known to block the degradation of Ikappabeta, the cytosolic inhibitor of NF-kappaB. Following this treatment, the Caco-2 cells were stimulated with IL-1beta, and C3 levels in the culture medium were measured after 24 h by ELISA. C3 mRNA levels were determined after 4 h by Northern blot analysis. In other experiments, Caco-2 cells were transfected with a mutant IkappaBalpha in which serines 32 and 36 were substituted by alanine. This mutation prevents IkBalpha phosphorylation and subsequent NF-kappaB nuclear translocation. After transfection, the cells were stimulated with IL-1beta, and C3 levels in the culture medium were measured after 24 h. Cytosolic IkappaBalpha was determined by Western blot analysis.. TLCK, genistein, and LLnL each inhibited IL-1beta-induced C3 production in a dose-dependent fashion. These responses were associated with decreased C3 mRNA levels. In contrast, PDTC did not influence C3 production or C3 mRNA in the Caco-2 cells. Transfection of the Caco-2 cells with the Ser 32/36 mutant IkBalpha resulted in maintained IkappaBalpha levels and decreased IL-beta-induced C3 production.. IL-1beta-stimulated C3 production in the enterocyte may be regulated by NF-kappaB. Topics: Base Sequence; Caco-2 Cells; Complement C3; DNA Primers; DNA-Binding Proteins; Genistein; Humans; I-kappa B Proteins; Interleukin-1; Intestinal Mucosa; Leupeptins; Mutation; NF-kappa B; NF-KappaB Inhibitor alpha; Pyrrolidines; RNA, Messenger; Thiocarbamates; Tosyllysine Chloromethyl Ketone; Transfection | 1999 |
Calpain inhibitor entrapped in liposome rescues ischemic neuronal damage.
Transient forebrain ischemia induces activation of calpain and proteolysis of a neuronal cytoskeleton, fodrin, in gerbil hippocampus. This phenomenon precedes delayed neuronal death in hippocampal CA1 neurons. We examined effects of a calpain inhibitor on delayed neuronal death after transient forebrain ischemia. In gerbils, a selective calpain inhibitor entrapped in liposome was given transvenously and 30 min later, 5-min forebrain ischemia was produced by occlusion of both common carotid arteries. On day 7, CA1 neuronal damage was examined in the hippocampal slices stained with cresyl violet. Calpain-induced proteolysis of fodrin was also examined by immunohistochemistry and immunoblot. Additionally, to assure entrapment of the inhibitor by CA1 neurons, the inhibitor-liposome complex was labeled with FITC and given to gerbils. Fluorescence in the hippocampal slices was examined by confocal laser scanning microscope. Selective CA1 neuronal damage induced by forebrain ischemia was prevented by administration of the inhibitor in a dose-dependent manner. Calpain-induced proteolysis of fodrin was also extinguished by the calpain inhibitor in a dose-dependent manner. Bright fluorescence of the FITC-labeled inhibitor was observed in the CA1 neurons. The data show an important role of calpain in the development of the ischemic delayed neuronal death. Calpain seems to produce neuronal damage by degrading neuronal cytoskeleton. Our data also show a palliative effect of the calpain inhibitor on the neurotoxic damage, which offers a new and potent treatment of transient forebrain cerebral ischemia. Topics: Animals; Blood-Brain Barrier; Blotting, Western; Calpain; Carrier Proteins; Cell Death; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Drug Carriers; Fluorescein-5-isothiocyanate; Fluorescent Dyes; Gerbillinae; Hippocampus; Immunohistochemistry; Injections, Intravenous; Ischemic Attack, Transient; Leupeptins; Liposomes; Microfilament Proteins; Microscopy, Confocal; Neurons; Prosencephalon | 1999 |
Calpain inhibitor I increases beta-amyloid peptide production by inhibiting the degradation of the substrate of gamma-secretase. Evidence that substrate availability limits beta-amyloid peptide production.
The calpain inhibitor N-acetyl-leucyl-leucyl-norleucinal (ALLN) has been reported to have complex effects on the production of the beta-amyloid peptide (Abeta). In this study, the effects of ALLN on the processing of the amyloid precursor protein (APP) to Abeta were examined in 293 cells expressing APP or the C-terminal 100 amino acids of APP (C100). In cells expressing APP or low levels of C100, ALLN increased Abeta40 and Abeta42 secretion at low concentrations, decreased Abeta40 and Abeta42 secretion at high concentrations, and increased cellular levels of C100 in a concentration-dependent manner by inhibiting C100 degradation. Low concentrations of ALLN increased Abeta42 secretion more dramatically than Abeta40 secretion. ALLN treatment of cells expressing high levels of C100 did not alter cellular C100 levels and inhibited Abeta40 and Abeta42 secretion with similar IC50 values. These results suggest that C100 can be processed both by gamma-secretase and by a degradation pathway that is inhibited by low concentrations of ALLN. The data are consistent with inhibition of gamma-secretase by high concentrations of ALLN but do not support previous assertions that ALLN is a selective inhibitor of the gamma-secretase producing Abeta40. Rather, Abeta42 secretion may be more dependent on C100 substrate concentration than Abeta40 secretion. Topics: Alzheimer Disease; Amino Acid Sequence; Amyloid beta-Peptides; Amyloid beta-Protein Precursor; Amyloid Precursor Protein Secretases; Aspartic Acid Endopeptidases; Calpain; Cell Line; Endopeptidases; Gene Expression Regulation; Humans; Leupeptins; Membrane Proteins; Molecular Sequence Data; Mutation; Peptide Fragments; Presenilin-1; Protease Inhibitors; Transfection | 1999 |
Posttranslational regulation of the retinoblastoma gene family member p107 by calpain protease.
The retinoblastoma protein plays a critical role in regulating the G1/S transition. Less is known about the function and regulation of the homologous pocket protein p107. Here we present evidence for the posttranslational regulation of p107 by the Ca2+-activated protease calpain. Three negative growth regulators, the HMG-CoA reductase inhibitor lovastatin, the antimetabolite 5-fluorouracil, and the cyclic nucleotide dibutyryl cAMP were found to induce cell type-specific loss of p107 protein which was reversible by the calpain inhibitor leucyl-leucyl-norleucinal but not by the serine protease inhibitor phenylmethylsulfonylfluoride, caspase inhibitors, or lactacystin, a specific inhibitor of the 26S proteasome. Purified calpain induced Ca2+-dependent p107 degradation in cell lysates. Transient expression of the specific calpain inhibitor calpastatin blocked the loss of p107 protein in lovastatin-treated cells, and the half-life of p107 was markedly lengthened in lovastatian-treated cells stably transfected with a calpastatin expression vector versus cells transfected with vector alone. The data presented here demonstrate down-regulation of p107 protein in response to various antiproliferative signals, and implicate calpain in p107 posttranslational regulation. Topics: Acetylcysteine; Amino Acid Chloromethyl Ketones; Bucladesine; Calpain; Cyclin B; Cyclin B1; Cysteine Proteinase Inhibitors; Dipeptides; Fluorouracil; Humans; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Ketones; Leupeptins; Lovastatin; Nuclear Proteins; Protein Processing, Post-Translational; Retinoblastoma Protein; Retinoblastoma-Like Protein p107; Tumor Cells, Cultured | 1999 |
Regulation of beta-catenin signaling by the B56 subunit of protein phosphatase 2A.
Dysregulation of Wnt-beta-catenin signaling disrupts axis formation in vertebrate embryos and underlies multiple human malignancies. The adenomatous polyposis coli (APC) protein, axin, and glycogen synthase kinase 3beta form a Wnt-regulated signaling complex that mediates the phosphorylation-dependent degradation of beta-catenin. A protein phosphatase 2A (PP2A) regulatory subunit, B56, interacted with APC in the yeast two-hybrid system. Expression of B56 reduced the abundance of beta-catenin and inhibited transcription of beta-catenin target genes in mammalian cells and Xenopus embryo explants. The B56-dependent decrease in beta-catenin was blocked by oncogenic mutations in beta-catenin or APC, and by proteasome inhibitors. B56 may direct PP2A to dephosphorylate specific components of the APC-dependent signaling complex and thereby inhibit Wnt signaling. Topics: Adenomatous Polyposis Coli Protein; Animals; beta Catenin; Calcium-Calmodulin-Dependent Protein Kinases; Cell Line; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytoskeletal Proteins; Down-Regulation; Genes, Reporter; Glycogen Synthase Kinase 3; Glycogen Synthase Kinases; Humans; Leupeptins; Multienzyme Complexes; Mutation; Phosphoprotein Phosphatases; Phosphorylation; Proteasome Endopeptidase Complex; Protein Phosphatase 2; Proto-Oncogene Proteins; Signal Transduction; Trans-Activators; Transcriptional Activation; Transfection; Tumor Cells, Cultured; Wnt Proteins; Xenopus; Xenopus Proteins; Zebrafish Proteins | 1999 |
Chinese hamster ovary cells require the coexpression of microsomal triglyceride transfer protein and cholesterol 7alpha-hydroxylase for the assembly and secretion of apolipoprotein B-containing lipoproteins.
Due to the absence of microsomal triglyceride transfer protein (MTP), Chinese hamster ovary (CHO) cells lack the ability to translocate apoB into the lumen of the endoplasmic reticulum, causing apoB to be rapidly degraded by an N-acetyl-leucyl-leucyl-norleucinal-inhibitable process. The goal of this study was to examine if expression of MTP, whose genetic deletion is responsible for the human recessive disorder abetalipoproteinemia, would recapitulate the lipoprotein assembly pathway in CHO cells. Unexpectedly, expression of MTP mRNA and protein in CHO cells did not allow apoB-containing lipoproteins to be assembled and secreted by CHO cells expressing apoB53. Although expression of MTP in cells allowed apoB to completely enter the endoplasmic reticulum, it was degraded by a proteolytic process that was inhibited by dithiothreitol (1 mM) and chloroquine (100 microM), but resistant to N-acetyl-leucyl-leucyl-norleucinal. In marked contrast, coexpression of the liver-specific gene product cholesterol 7alpha-hydroxylase with MTP resulted in levels of MTP lipid transfer activity that were similar to those in mouse liver and allowed intact apoB53 to be secreted as a lipoprotein particle. These data suggest that, although MTP-facilitated lipid transport is not required for apoB translocation, it is required for the secretion of apoB-containing lipoproteins. We propose that, in CHO cells, MTP plays two roles in the assembly and secretion of apoB-containing lipoproteins: 1) it acts as a chaperone that facilitates apoB53 translocation, and 2) its lipid transfer activity allows apoB-containing lipoproteins to be assembled and secreted. Our results suggest that the phenotype of the cell (e.g. expression of cholesterol 7alpha-hydroxylase by the liver) may profoundly influence the metabolic relationships determining how apoB is processed into lipoproteins and/or degraded. Topics: Animals; Apolipoproteins B; Biological Transport; Carrier Proteins; Chloroquine; CHO Cells; Cholesterol 7-alpha-Hydroxylase; Cricetinae; Dithiothreitol; Endoplasmic Reticulum; HeLa Cells; Humans; Leupeptins; Lipoproteins; Liver; Mice; Transfection | 1999 |
Distinct secretases, a cysteine protease and a serine protease, generate the C termini of amyloid beta-proteins Abeta1-40 and Abeta1-42, respectively.
The carboxy-terminal ends of the 40- and 42-amino acids amyloid beta-protein (Abeta) may be generated by the action of at least two different proteases termed gamma(40)- and gamma(42)-secretase, respectively. To examine the cleavage specificity of the two proteases, we treated amyloid precursor protein (APP)-transfected cell cultures with several dipeptidyl aldehydes including N-benzyloxycarbonyl-Leu-leucinal (Z-LL-CHO) and the newly synthesized N-benzyloxycarbonyl-Val-leucinal (Z-VL-CHO). All dipeptidyl aldehydes tested inhibited production of both Abeta1-40 and Abeta1-42. Changes in the P1 and P2 residues of these aldehydes, however, indicated that the amino acids occupying these positions are important for the efficient inhibition of gamma-secretases. Peptidyl aldehydes inhibit both cysteine and serine proteases, suggesting that the two gamma-secretases belong to one of these mechanistic classes. To differentiate between the two classes of proteases, we treated our cultures with the specific cysteine protease inhibitor E-64d. This agent inhibited production of secreted Abeta1-40, with a concomitant accumulation of its cellular precursor indicating that gamma(40)-secretase is a cysteine protease. In contrast, this treatment increased production of secreted Abeta1-42. No inhibition of Abeta production was observed with the potent calpain inhibitor I (acetyl-Leu-Leu-norleucinal), suggesting that calpain is not involved. Together, these results indicate that gamma(40)-secretase is a cysteine protease distinct from calpain, whereas gamma(42)-secretase may be a serine protease. In addition, the two secretases may compete for the same substrate. Dipeptidyl aldehyde treatment of cultures transfected with APP carrying the Swedish mutation resulted in the accumulation of the beta-secretase C-terminal APP fragment and a decrease of the alpha-secretase C-terminal APP fragment, indicating that this mutation shifts APP cleavage from the alpha-secretase site to the beta-secretase site. Topics: Aldehydes; Alzheimer Disease; Amyloid beta-Peptides; Amyloid Precursor Protein Secretases; Animals; Calpain; CHO Cells; Cricetinae; Cysteine Endopeptidases; Dipeptides; Endopeptidases; Enzyme Inhibitors; Leupeptins; Peptide Fragments; Serine Endopeptidases | 1999 |
Proteasome blockers inhibit TNF-alpha release by lipopolysaccharide stimulated macrophages and microglia: implications for HIV-1 dementia.
HIV-1 infection of the central nervous system can cause severe neurologic disease although only microglial cells and brain macrophages are susceptible to productive viral infection. Substances secreted by infected cells are thought to cause disease indirectly. Tumor necrosis factor alpha (TNF-alpha) is one candidate neurotoxin and is upregulated during HIV-1 infection of the brain, likely via activation of the transcription factor NF-kappaB. We used the proteasome inhibitors, MG132 and ALLN (N-acetyl-Leu-Leu-Norleucinal), to inhibit NF-kappaB activation in primary human fetal microglia (PHFM) and primary monocyte derived-macrophages, and showed that they could block TNF-alpha release stimulated by lipopolysaccharide (LPS) or TNF-alpha. In addition, we performed electrophoretic mobility shift analysis and determined that in microglia, the p50/p65 heterodimer of NF-kappaB is activated by LPS stimulation, and is inhibited by MG132. Thus, blockade of NF-kappaB activation in microglia in vitro can decrease production of TNF-alpha and may prove to be a novel strategy for treating HIV-1 dementia. Topics: AIDS Dementia Complex; Cells, Cultured; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA Probes; Enzyme Activation; Fetus; Gene Expression; HIV-1; Humans; Leupeptins; Lipopolysaccharides; Macrophages; Microglia; Multienzyme Complexes; NF-kappa B; Phagocytosis; Proteasome Endopeptidase Complex; Tumor Necrosis Factor-alpha | 1999 |
The ATM protein is required for sustained activation of NF-kappaB following DNA damage.
Cells lacking an intact ATM gene are hypersensitive to ionizing radiation and show multiple defects in the cell cycle-coupled checkpoints. DNA damage usually triggers cell cycle arrest through, among other things, the activation of p53. Another DNA-damage responsive factor is NF-kappaB. It is activated by various stress situations, including oxidative stress, and by DNA-damaging compounds such as topoisomerase poisons. We found that cells from Ataxia Telangiectasia patients exhibit a defect in NF-kappaB activation in response to treatment with camptothecin, a topoisomerase I poison. In AT cells, this activation is shortened or suppressed, compared to that observed in normal cells. Ectopic expression of the ATM protein in AT cells increases the activation of NF-kappaB in response to camptothecin. MO59J glioblastoma cells that do not express the DNA-PK catalytic subunit respond normally to camptothecin. These results support the hypothesis that NF-kappaB is a DNA damage-responsive transcription factor and that its activation pathway by DNA damage shares some components with the one leading to p53 activation. Topics: Adolescent; Age Factors; Antineoplastic Agents, Phytogenic; Ataxia Telangiectasia; Ataxia Telangiectasia Mutated Proteins; Brain Neoplasms; Camptothecin; Cell Cycle Proteins; Cells, Cultured; Child; Child, Preschool; DNA Damage; DNA-Activated Protein Kinase; DNA-Binding Proteins; Enzyme Inhibitors; Fibroblasts; Gene Expression Regulation; Glioblastoma; Humans; I-kappa B Proteins; Leupeptins; NF-kappa B; NF-KappaB Inhibitor alpha; Nuclear Proteins; Phosphoric Monoester Hydrolases; Protease Inhibitors; Protein Serine-Threonine Kinases; Proteins; Radiation Tolerance; Recombinant Fusion Proteins; Topoisomerase I Inhibitors; Transcription, Genetic; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha; Tumor Suppressor Protein p53; Tumor Suppressor Proteins | 1999 |
Inhibition of cytoplasmic antigen, glucose- 6-phosphate dehydrogenase, by VH-CH1, an intracellular Fd fragment antibody derived from a semisynthetic Fd fragment phage display library.
A library of Fd fragment antibody binding proteins was created by random mutation of 15 nucleotides within the CDRIII region of the immunoglobulin heavy chain gene and displayed as Fd coat protein fusion constructs of M13 phage. The library was screened for those VHbinding sites that bound glucose-6-phosphate dehydrogenase (G6PD). One isolate (DH27bp) inhibited G6PD activity by 85 %. The DH27bpgene was re-engineered, placed in a eukaryotic expression vector having an isopropyl-beta-delta-thiogalactopyranoside (IPTG) inducible promoter, and transfected and then expressed in Chinese hamster V79 cells. G6PD activity was completely inhibited. Removal of IPTG reverted the cell to full G6PD activity. The intracellular dynamics of the G6PD/DH27bpcomplex showed that when the proteasomes of cells expressing DH27bpwere inhibited (N -acetyl-Leu-Leu-norleucinal or lactacystin) G6PD activity increased. Metabolic labelling of newly synthesized IPTG-induced proteins during/absence of proteasomal inhibitors showed that both G6PD and DH27bpare signaled for degradation when the intracellular complex is formed. Furthermore, semi-quantitative RT/PCR demonstrated that G6PD mRNA is upregulated over the time course of G6PD inactivation by DH27bpFd binding protein. These effects were not observed in those cells expressing a non-mutated Fd (UMHC) or in IPTG-treated non-transduced V79 cells. Our results demonstrate that an Fd-based intracellular binding protein can find and disable the function of a specific intracellular target and once the Fd expression is repressed the activity of intracellular targeted protein can revert to normal. Topics: Acetylcysteine; Animals; Cell Line; Cricetinae; Cricetulus; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytoplasm; Gene Expression Regulation, Enzymologic; Glucosephosphate Dehydrogenase; Immunoglobulin Constant Regions; Immunoglobulin Heavy Chains; Immunoglobulin Variable Region; Inovirus; Isopropyl Thiogalactoside; Kinetics; Leupeptins; Multienzyme Complexes; Peptide Library; Proteasome Endopeptidase Complex; Recombinant Proteins; Reverse Transcriptase Polymerase Chain Reaction; Transfection | 1999 |
Protective effects of calpain inhibitor for prolonged hypothermic cardiac preservation.
For successful organ transplantation, it is important to properly preserve the donor organ. This study was carried out to investigate tissue damage generated by the activation of calpain during prolonged hypothermic cardiac preservation using specific antibodies for mu- and m-calpain proenzymes, and to ensure the protective effect of calpain inhibitor 1 (N-acetyl-leucyl-leucyl-norleucinal).. Excised rat hearts were divided into two groups: in Group I, the heart was arrested and immersed in University of Wisconsin solution with 20 microM of calpain inhibitor 1 (n = 28) and in Group N, the heart was arrested and immersed in University of Wisconsin solution without calpain inhibitor (n = 27). After a 12-hour preservation period at 4 degrees C, the hearts were reperfused on an isolated perfusion apparatus. Separation of the myocardial calpain isozymes was carried out by DEAE cellulose chromatography and both calpain proenzymes were detected by immunoblotting.. The cardiac function was more satisfactorily maintained in Group I in comparison with Group N. Remarkable leakage of creatine kinase, glutamic-oxaloacetic transaminase and lactate dehydrogenase was detected in Group N, while it was efficiently suppressed in Group I. During ischemia, mu-calpain proenzyme decreased in Group N (p < 0.01), but there was no significant change in m-calpain. However, during reperfusion, both mu- and m-calpains decreased more in Group N (p < 0.01).. Activation of calpain proenzymes and a decrease in cardiac function during preservation and reperfusion were demonstrated. The use of calpain inhibitor to protect against tissue damage was suggested as being useful for the prolonged preservation of the heart. Topics: Adenosine; Allopurinol; Animals; Antibodies; Calpain; Enzyme Precursors; Glutathione; Heart; Heart Transplantation; Hypothermia, Induced; Insulin; Leupeptins; Male; Organ Preservation; Organ Preservation Solutions; Raffinose; Rats; Rats, Wistar; Time Factors | 1999 |
Inhibition of E6 induced degradation of p53 is not sufficient for stabilization of p53 protein in cervical tumour derived cell lines.
The E6 proteins derived from tumour associated papillomavirus types target the cellular tumour suppressor protein p53 for ubiquitin mediated degradation. In cell lines derived from cervical tumours the p53 protein is present in very low amounts, but it can be activated by appropriate DNA damaging agents, indicating that functional p53 is present within these lines. Recent studies have also shown that different polymorphic forms of the p53 protein are differentially susceptible to E6 mediated degradation. Therefore we have been interested in analysing the effects of different HPV E6 proteins upon p53 levels in a variety of cervical tumour derived cell lines. We show that inhibition of E6 mediated degradation of p53 frequently results in increased levels of p53 expression. However, there are notable exceptions to this where increased p53 levels are only obtained following DNA damage and proteasome inhibition. We also show in E6 expressing cells, that as well as p53 being targeted for degradation, the localization of p53 to the nucleus is also inhibited, consistent with previous observations which indicate that degradation of p53 is not essential for E6 mediated inhibition of p53 function. These results have important implications for any potential therapies which might aim to block E6 mediated degradation of p53. Topics: Acetylcysteine; Adenocarcinoma; Animals; Antibiotics, Antineoplastic; Breast Neoplasms; Carcinoma; Cell Nucleus; Cysteine Proteinase Inhibitors; DNA Damage; DNA-Binding Proteins; Female; Fibrosarcoma; Humans; Leupeptins; Mitomycin; Oncogene Proteins, Viral; Papillomaviridae; Polymorphism, Genetic; Repressor Proteins; Tumor Cells, Cultured; Tumor Suppressor Protein p53; Uterine Cervical Neoplasms | 1999 |
Ubiquitin-proteasome system is involved in induction of LFA-1/ICAM-1-dependent adhesion of HL-60 cells.
Membrane-permeable proteasome inhibitors, lactacystin (LC) and N-acetyl-Leu-Leu-norleucinal (ALLN), but not calpain inhibitor Z-Leu-leucinal (ZLL), prevented LFA-1/ICAM-1-dependent cellular adhesion of TPA-stimulated HL-60 cells. These proteasome inhibitors affected neither the induction of monocytic differentiation nor the accompanying protein-tyrosine phosphorylation. They suppressed the increase in the avidity of LFA-1 to ICAM-1 without changing the expression of these molecules. Immunoblotting using monoclonal antibody FK-1, which reacts specifically with polyubiquitinated proteins, demonstrated that the proteasome inhibitors caused the drastic accumulation of the polyubiquitinated proteins in the membrane fraction of TPA-treated HL-60 cells. This indicates that accompanying activation of LFA-1, TPA induces the polyubiquitination of the membrane proteins, which are rapidly degraded by proteasomes. These data taken together show that proteolysis mediated by the ubiquitin-proteasome system is a prerequisite for the induction of LFA-1-dependent adhesion of HL-60 cells. Topics: Acetylcysteine; Adenosine Triphosphatases; Antibody Affinity; Cell Adhesion; Cell Differentiation; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; HL-60 Cells; Humans; Intercellular Adhesion Molecule-1; Leupeptins; Lymphocyte Function-Associated Antigen-1; Monocytes; Multienzyme Complexes; Proteasome Endopeptidase Complex; Tetradecanoylphorbol Acetate; Tretinoin; Ubiquitins | 1999 |
Growth inhibition by CDK-cyclin and PCNA binding domains of p21 occurs by distinct mechanisms and is regulated by ubiquitin-proteasome pathway.
The CDK inhibitor, p21WAF1/Cip1 blocks cell cycle progression. In vitro, the N-terminus of p21 binds and inhibits CDK-cyclin kinase activity, whereas the C-terminus binds and inhibits PCNA (proliferating cell nuclear antigen) function. PCNA is essential for processivity of both DNA polymerase delta and epsilon. We have performed a detailed analysis of growth inhibition by the N- and C-terminal regions of p21, and determined whether the N- and C-terminal regions mediate this effect by different mechanisms. Expression of either the N- or the C-terminal region of p21 inhibits DNA synthesis and cell growth, but not as efficiently as full length p21. The effectiveness of the two p21 domains is dependent on their stability which is determined by the ubiquitin-proteasome pathway. The stabilization of the N- and C-terminal region of p21 increases their effectiveness as inhibitors of DNA synthesis to levels comparable to full length p21. Inhibition of DNA synthesis by the N-terminal region of p21 involves suppression of E2F activity. In contrast, inhibition by the C-terminal region of p21 is not accompanied by suppression of E2F activity, but is mediated via PCNA binding. The C-terminal region of p21 therefore inhibits cell growth by a mechanism distinct from that of the N-terminal region containing the CDK-cyclin inhibitory domain. Topics: 3T3 Cells; Acetylcysteine; Animals; CDC2-CDC28 Kinases; Cyclin-Dependent Kinase 2; Cyclin-Dependent Kinase Inhibitor p21; Cyclin-Dependent Kinases; Cyclins; Cycloheximide; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Gene Deletion; Gene Expression Regulation, Neoplastic; Growth Inhibitors; Hemagglutinins; Humans; Leupeptins; Mice; Models, Genetic; Multienzyme Complexes; Mutagenesis; Osteosarcoma; Proliferating Cell Nuclear Antigen; Proteasome Endopeptidase Complex; Protein Serine-Threonine Kinases; Protein Synthesis Inhibitors; Time Factors; Transfection; Tumor Cells, Cultured; Ubiquitins | 1999 |
Caspase-induced proteolysis of the cyclin-dependent kinase inhibitor p27Kip1 mediates its anti-apoptotic activity.
The caspase-mediated cleavage of a limited number of cellular proteins is a common feature of apoptotic cell death. This cleavage usually inhibits the function of the target protein or generates peptides that actively contribute to the death process. In the present study, we demonstrate that the cyclin-dependent kinase inhibitor p27Kip1 is cleaved by caspases in human leukemic cells exposed to apoptotic stimuli. We have shown recently that p27Kip1 overexpression delayed leukemic cell death in response to cytotoxic drugs. In transient transfection experiments, the p23 and the p15 N-terminal peptides generated by p27Kip1 proteolysis demonstrate an anti-apoptotic effect similar to that induced by the wild-type protein, whereas cleavage-resistant mutants have lost their protective effect. Moreover, stable transfection of a cleavage-resistant mutant of p27Kip1 sensitizes leukemic cells to drug-induced cell death. Altogether, these results indicate that proteolysis of p27Kip1 triggered by caspases mediates the anti-apoptotic activity of the protein. Topics: Amino Acid Chloromethyl Ketones; Apoptosis; Base Sequence; Calpain; Caspase 3; Caspase 6; Caspase 8; Caspase 9; Caspase Inhibitors; Caspases; CDC2-CDC28 Kinases; Cell Cycle Proteins; Cyclin-Dependent Kinase 2; Cyclin-Dependent Kinase Inhibitor p27; Cyclin-Dependent Kinases; Cysteine Proteinase Inhibitors; Etoposide; Humans; Leukemia; Leupeptins; Microtubule-Associated Proteins; Molecular Sequence Data; Mutation; Nucleic Acid Synthesis Inhibitors; Oligopeptides; Protein Serine-Threonine Kinases; Thimerosal; Tumor Cells, Cultured; Tumor Suppressor Protein p53; Tumor Suppressor Proteins | 1999 |
Calpain inhibitor causes accumulation of ubiquitinated P-glycoprotein at the cell surface: possible role of calpain in P-glycoprotein turnover.
P-glycoprotein (Pgp) is a plasma-membrane glycoprotein that confers multi-drug resistance (MDR) on cells and displays ATP-driven drug pumping. The possible contribution of calpain-mediated proteolytic pathways to the functional regulation of the Pgp molecule was evaluated using K562/DXR, MDR cells. N-Acetyl-L-leucyl-L-leucyl-norleucinal was effluxed by Pgp, but N-benzyloxycarbonyl-L-leucyl-L-leucinal (zLLal), an inhibitor of calpain, retarded the degradation of Pgp leading to accumulation of the molecule largely at the cell surface membrane. Treatment with brefeldin A did not obstruct the zLLal-induced Pgp accumulation. NH4Cl increased the cytoplasmic Pgp level, with a slight to significant decrease at the cell surface membrane. Ubiquitin-ELISA and western blot analysis confirmed that the Pgp molecule, which accumulated mainly at the cell surface, was ubiquitinated. However, lactacystin did not show any accumulation of Pgp in either the cytoplasm or the cell surface membrane, suggesting that the proteasome did not participate in the phenomenon. Additionally, the Pgp was limitedly proteolyzed by calpain into two 98 kDa and 69 kDa, fragments within one minute. Despite the increased accumulation of Pgp at the cell surface after treatment with calpain inhibitor, the cytoplasmic doxorubicin level of the cells treated with a calpain inhibitor was higher than that of non-treated cells and approached that of parental cells. These results indicated that calpain involved Pgp turnover and that calpain inhibition induced ubiquitinated Pgp-accumulation mainly at the cell surface membrane with a reduction in its own functions suggesting that the modulation of Pgp-turnover involves MDR-reversal by another approach. Topics: Acetylcysteine; Ammonium Chloride; ATP Binding Cassette Transporter, Subfamily B; Blotting, Western; Brefeldin A; Calpain; Cell Membrane; Cysteine Proteinase Inhibitors; Dipeptides; Doxorubicin; Enzyme-Linked Immunosorbent Assay; Glycoproteins; Humans; K562 Cells; Leucine; Leupeptins; Membrane Proteins; Protein Synthesis Inhibitors; Time Factors; Ubiquitins | 1999 |
Regulation of BRCA1 by protein degradation.
BRCA1, a tumor suppressor protein implicated in hereditary forms of breast and ovarian cancer, is transcriptionally regulated in a proliferation-dependent manner. In this study, we demonstrate a substantial role for proteolysis in regulating the BRCA1 steady-state protein level in several cell lines. N-acetyl-leu-leu-norleucinal (ALLN), an inhibitor of the proteasome, calpain, and cathepsins, caused BRCA1 protein to accumulate in the nucleus of several human breast, prostate, and melanoma cell lines which express low or undetectable basal levels of BRCA1 protein, but not in cells with high basal expression of BRCA1. Protease inhibition did not increase BRCA1 synthesis, nor change its mRNA level, but it dramatically prolonged the protein's half-life. In contrast to ALLN, lactacystin and PS341, two specific proteasome inhibitors, as well as calpastatin peptide and PD150606, two selective calpain inhibitors, had no effect on BRCA1 stability, whereas ALLM, an effective calpain and cathepsin inhibitor but weak proteasome inhibitor, did stimulate accumulation of BRCA1. Moreover, three inhibitors of acidic cysteine proteases, chloroquine, ammonium chloride and bafilomycin, were as effective as ALLN. These results demonstrate that degradation by a cathepsin-like protease in fine balance with BRCA1 transcription is responsible for maintaining the low steady-state level of BRCA1 protein seen in many cancer cells. Topics: BRCA1 Protein; Calpain; Carrier Proteins; Cell Nucleus; Cysteine Endopeptidases; Dactinomycin; Down-Regulation; Endopeptidases; Exportin 1 Protein; Gene Expression Regulation; Half-Life; Humans; Hydrolysis; Karyopherins; Leupeptins; Multienzyme Complexes; Proteasome Endopeptidase Complex; Receptors, Cytoplasmic and Nuclear; RNA, Messenger; Tumor Cells, Cultured | 1999 |
Role of c-Jun N-terminal kinase 1 (JNK1) in cell cycle checkpoint activated by the protease inhibitor N-acetyl-leucinyl-leucinyl-norleucinal.
The cysteine protease inhibitor N-acetyl-leucinyl-leucinyl-norleucinal (LLnL) inhibited the growth of the Calu-1 lung carcinoma cells and induced a prolonged cell cycle arrest in the S phase. c-Jun N-terminal kinases (JNKs) participate in cellular responses to mitogenic stimuli, environmental stresses, and apoptotic signals but its role in cell cycle checkpoint control has not been elucidated. In this report, we examined the role of JNK in LLnL-induced S phase checkpoint by overexpression of a dominant-negative mutant of JNK1 (JNK1-APF) in Calu-1 cells. Expression of high levels of JNK1-APF blocked the growth-inhibitory effects of LLnL and abrogated S phase arrest induced by LLnL. These results support the role of JNK in the activation of cell cycle checkpoint induced by LLnL. Topics: Anisomycin; Cell Cycle; Cell Line; Cysteine Proteinase Inhibitors; Enzyme Activation; JNK Mitogen-Activated Protein Kinases; Leupeptins; Mitogen-Activated Protein Kinases | 1999 |
Reduced MAP kinase phosphatase-1 degradation after p42/p44MAPK-dependent phosphorylation.
The mitogen-activated protein (MAP) kinase cascade is inactivated at the level of MAP kinase by members of the MAP kinase phosphatase (MKP) family, including MKP-1. MKP-1 was a labile protein in CCL39 hamster fibroblasts; its degradation was attenuated by inhibitors of the ubiquitin-directed proteasome complex. MKP-1 was a target in vivo and in vitro for p42(MAPK) or p44(MAPK), which phosphorylates MKP-1 on two carboxyl-terminal serine residues, Serine 359 and Serine 364. This phosphorylation did not modify MKP-1's intrinsic ability to dephosphorylate p44(MAPK) but led to stabilization of the protein. These results illustrate the importance of regulated protein degradation in the control of mitogenic signaling. Topics: Animals; Blood; Cell Cycle Proteins; Cell Division; Cell Line; Cricetinae; Culture Media; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Dual Specificity Phosphatase 1; Estradiol; Humans; Immediate-Early Proteins; Leucine; Leupeptins; MAP Kinase Signaling System; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Multienzyme Complexes; Mutation; Nitrophenols; Organophosphorus Compounds; Phosphoprotein Phosphatases; Phosphorylation; Proteasome Endopeptidase Complex; Protein Phosphatase 1; Protein Tyrosine Phosphatases; Ubiquitins | 1999 |
The deubiquitinating enzyme Fam interacts with and stabilizes beta-catenin.
In the ubiquitin-proteasome pathway, the ubiquitinated substrates either undergo degradation by the proteasome or stabilization through the action of the deubiquitinating enzyme. We have previously found that the deubiquitinating enzyme Fam is colocalized with AF-6, one of the effectors of the Ras small GTPase, at cell-cell contact sites in epithelial cells and interacts with AF-6 in vivo and in vitro. Fam has deubiquitinating activity in vitro and prevents the ubiquitination of AF-6 in intact cells. The degradation of beta-catenin, which accumulates at the cell-cell contact sites as a cadherin/catenin complex, is thought to be regulated by the ubiquitin-proteasome pathway. These observations prompted us to examine the possible Fam regulation of the stabilization of beta-catenin.. We found that Fam interacted with beta-catenin both in vivo and in vitro. The Fam-binding site of beta-catenin mapped to the region close to the APC or Axin-binding site of beta-catenin. Over-expression of Fam in mouse L cells resulted in an elevation of beta-catenin levels and in an elongation of the half-life of beta-catenin. In these L cells, Fam was colocalized with beta-catenin at the dot-like structures in the cytoplasm.. These results indicate that Fam interacts with and stabilizes beta-catenin in vivo, presumably through the deubiquitination of beta-catenin. Topics: Animals; beta Catenin; Binding Sites; Cadherins; Cell Line; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytoskeletal Proteins; Dogs; Endopeptidases; Kinesins; Kinetics; L Cells; Leupeptins; Mice; Multienzyme Complexes; Myosins; Oligopeptides; Proteasome Endopeptidase Complex; ras Proteins; Recombinant Fusion Proteins; Signal Transduction; Trans-Activators; Transfection; Ubiquitin Thiolesterase; Ubiquitins | 1999 |
Stability of the Listeria monocytogenes ActA protein in mammalian cells is regulated by the N-end rule pathway.
Upon infection of mammalian cells, Listeria monocytogenes lyses the phagosome and enters the cytosol, where it secretes proteins necessary for its intracellular growth cycle. Consequently, bacterial proteins exposed to the cytosol are potential targets for degradation by host cytosolic proteases. One pathway for degradation of host cytosolic proteins, the N-end rule pathway, involves recognition of the N-terminal amino acid and is mediated by the proteasome. However, very few natural N-end rule substrates have been identified. We have examined the L. monocytogenes ActA protein as a potential target for this pathway. ActA is an essential determinant of L. monocytogenes pathogenesis that is required to induce actin-based motility and cell-to-cell spread. We show that the half-life of a secreted form of ActA can be altered in the mammalian cytosol by changing the N-terminal amino acid. Moreover, the introduction of a destabilizing N-terminus into the functional, surface-bound form of ActA results in a small-plaque phenotype in L2 cells, which is partially reversible by an inhibitor of the proteasome. These results indicate that the L. monocytogenes ActA protein is a natural N-end rule substrate, and that optimal function of ActA in mediating cell-to-cell spread is dependent upon its intracellular turnover rate. Topics: Animals; Arginine; Bacterial Proteins; Cell Line; Colony Count, Microbial; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Half-Life; Leupeptins; Listeria monocytogenes; Membrane Proteins; Multienzyme Complexes; Mutation; Proteasome Endopeptidase Complex | 1999 |
Identification of tropoelastin as a ligand for the 65-kD FK506-binding protein, FKBP65, in the secretory pathway.
The folding and trafficking of tropoelastin is thought to be mediated by intracellular chaperones, although the identity and role of any tropoelastin chaperone remain to be determined. To identify proteins that are associated with tropoelastin intracellularly, bifunctional chemical cross-linkers were used to covalently stabilize interactions between tropoelastin and associated proteins in the secretory pathway in intact fetal bovine auricular chondrocytes. Immunoprecipitation of tropoelastin from cell lysates after cross-linking and analysis by SDS-PAGE showed the presence of two proteins of approximately 74 kD (p74) and 78 kD (p78) that coimmunoprecipitated with tropoelastin. Microsequencing of peptide fragments from a cyanogen bromide digest of p78 identified this protein as BiP and sequence analysis identified p74 as the peptidyl-prolyl cis-trans isomerase, FKPB65. The appearance of BiP and FKBP65 in the immunoprecipitations could be enhanced by the addition of brefeldin A (BFA) and N-acetyl-leu-leu-norleucinal (ALLN) to the culture medium for the final 4 h of labeling. Tropoelastin accumulates in the fused ER/Golgi compartment in the presence of BFA if its degradation is inhibited by ALLN (Davis, E.C., and R.P. Mecham. 1996. J. Biol. Chem. 271:3787-3794). The use of BFA and other secretion-disrupting agents suggests that the association of tropoelastin with FKBP65 occurs in the ER. Results from this study provide the first identification of a ligand for an FKBP in the secretory pathway and suggest that the prolyl cis-trans isomerase activity of FKBP65 may be important for the proper folding of the proline-rich tropoelastin molecule before secretion. Topics: Amino Acid Isomerases; Animals; Brefeldin A; Carrier Proteins; Cattle; Cell Compartmentation; Centrifugation, Density Gradient; Cyclopentanes; Cysteine; DNA-Binding Proteins; Heat-Shock Proteins; Leupeptins; Ligands; Methionine; Molecular Weight; Protein Folding; Protein Synthesis Inhibitors; Tacrolimus; Tacrolimus Binding Proteins; Tropoelastin | 1998 |
A promoter recruitment mechanism for tumor necrosis factor-alpha-induced interleukin-8 transcription in type II pulmonary epithelial cells. Dependence on nuclear abundance of Rel A, NF-kappaB1, and c-Rel transcription factors.
The alveolar macrophage-derived peptide tumor necrosis factor-alpha (TNFalpha) initiates pulmonary inflammation through its ability to stimulate interleukin-8 (IL-8) synthesis in alveolar epithelial cells through an incompletely described transcriptional mechanism. In this study, we use the technique of ligation-mediated polymerase chain reaction (LMPCR) to record changes in transcription factor occupancy of the IL-8 promoter after TNFalpha stimulation of A549 human alveolar cells. Using dimethylsulfate/LMPCR, no detectable proteins bind the TATA box in unstimulated cells. By contrast, TNFalpha rapidly induces protection of G residues at -79 and -80 coincident with endogenous IL-8 gene transcription. Using DNase I/LMPCR, we observe inducible protection of nucleotides -60 to -99 (the TNF response element) and nucleotides -3 to -32 (containing the TATA box). Surprisingly, extensive TATA box protection is only seen after TNFalpha stimulation. Using a two-step microaffinity isolation/Western immunoblot DNA binding assay, we observe that the NF-kappaB subunits Rel A, NF-kappaB1, and c-Rel inducibly bind the TNF response element; these proteins undergo rapid TNFalpha-inducible increases in nuclear abundance as a consequence of IkappaBalpha proteolysis. Furthermore, the peptide aldehyde N-acetyl-Leu-Leu-norleucinal, an agent that blocks both IkappaBalpha proteolysis and NF-kappaB subunit translocation, abrogates recombinant human TNFalpha-inducible IL-8 gene transcription. These studies demonstrate that IL-8 is activated by a promoter recruitment mechanism in alveolar epithelial cells, where NF-kappaB subunit translocation is required for (and coincident with) binding of the constitutively active TATA box-binding proteins. Topics: Binding Sites; Blotting, Western; Cell Line; Epithelial Cells; Humans; Hydrolysis; Interleukin-8; Kinetics; Leupeptins; Lung; Promoter Regions, Genetic; Transcription Factors; Transcription, Genetic; Tumor Necrosis Factor-alpha | 1998 |
Sepsis-induced increase in muscle proteolysis is blocked by specific proteasome inhibitors.
Recent studies suggest that sepsis stimulates ubiquitin-dependent protein breakdown in skeletal muscle. The 20S proteasome is the catalytic core of the ubiquitin-dependent proteolytic pathway. We tested the effects in vitro of the proteasome inhibitors N-acetyl-L-leucinyl-L-leucinal-L-norleucinal (LLnL) and lactacystin on protein breakdown in incubated muscles from septic rats. LLnL resulted in a dose- and time-dependent inhibition of protein breakdown in muscles from septic rats. Lactacystin blocked both total and myofibrillar muscle protein breakdown. In addition to inhibiting protein breakdown, LLnL reduced muscle protein synthesis and increased ubiquitin mRNA levels, probably reflecting inhibited proteasome-associated ribonuclease activity. Inhibited muscle protein breakdown caused by LLnL or lactacystin supports the concept that the ubiquitin-proteasome pathway plays a central role in sepsis-induced muscle proteolysis. The results suggest that muscle catabolism during sepsis may be inhibited by targeting specific molecular mechanisms of muscle proteolysis. Topics: Acetylcysteine; Animals; Cecum; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Dactinomycin; Kinetics; Leupeptins; Male; Multienzyme Complexes; Muscle Proteins; Muscle, Skeletal; Proteasome Endopeptidase Complex; Rats; Rats, Sprague-Dawley; RNA, Messenger; Sepsis; Transcription, Genetic; Ubiquitins | 1998 |
A proteasome inhibitor, an antioxidant, or a salicylate, but not a glucocorticoid, blocks constitutive and cytokine-inducible expression of P-selectin in human endothelial cells.
Proteasome inhibitors, antioxidants, salicylates, or glucocorticoids block the cytokine-induced expression of the endothelial cell adhesion molecules E-selectin, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1. These pharmacological agents have been assumed to inhibit the expression of adhesion molecules primarily by blocking activation of the transcription factor NF-kappaB. We found that the proteasome inhibitor ALLN, the antioxidant PDTC, or sodium salicylate, but not the glucocorticoid dexamethasone, inhibited both the constitutive and the interleukin-4- or oncostatin M-induced expression of the adhesion molecule P-selectin in human endothelial cells. ALLN, PDTC, or sodium salicylate decreased P-selectin expression without a detectable requirement for inhibition of NF-kappaB activation or for an intact kappaB element in the P-selectin gene. These results extend the potential anti-inflammatory utility of such drugs to inhibition of P-selectin expression and suggest that they have important actions that do not involve the NF-kappaB system. Topics: Animals; Antioxidants; Aorta; Cattle; Cells, Cultured; Cysteine Proteinase Inhibitors; Cytokines; Dexamethasone; Endothelium, Vascular; Gene Expression; Humans; Interleukin-4; Leupeptins; NF-kappa B; Oncostatin M; P-Selectin; Peptides; Pyrrolidines; Sodium Salicylate; Thiocarbamates; Umbilical Veins | 1998 |
Low environmental pH is responsible for the induction of nitric-oxide synthase in macrophages. Evidence for involvement of nuclear factor-kappaB activation.
Stimulation of macrophages with endotoxin and/or cytokines is responsible for the expression of the inducible isoform of nitric oxide synthase (iNOS). Because macrophages are exposed to low pH within the microenvironment of inflammatory lesions, the potential role of acidic pH as an additional regulator of iNOS was investigated. Substitution of the culture medium of rat peritoneal macrophages at pH 7.4 with medium at pH 7.0 up-regulated iNOS activity, as reflected by a 2.5-fold increase in nitrite accumulation. The increase in iNOS activity was associated with a similar increase in iNOS mRNA expression that reflected an increase in iNOS mRNA synthesis rather than stability. Low environmental pH-induced iNOS gene transcription involved the activation of nuclear factor-kappaB (NF-kappaB) transcription factor since exposure of macrophages to low environmental pH both increased NF-kappaB binding activity in the nucleus and enhanced NF-kappaB-driven reporter gene expression. In addition, treatment of macrophages with pyrrolidine dithiocarbamate or n-acetyl-leucinyl-leucinyl-norleucinal, two drugs preventing NF-kappaB translocation to the nucleus, canceled low pH-induced nitrite accumulation. The overall mechanism required the synthesis of tumor necrosis factor alpha (TNFalpha). Indeed, 1) elevated TNFalpha bioactivity was observed in the medium of macrophages exposed to pH 7.0, and 2) incubation of macrophages with a neutralizing anti-TNFalpha antibody impaired both NF-kappaB activation and nitrite accumulation in response to acid challenge. In summary, exposure of macrophages to acidic microenvironment in inflammatory lesions leads to the up-regulation of iNOS activity through the activation of NF-kappaB. Topics: Acids; Amiloride; Animals; Autocrine Communication; Cell Nucleus; Culture Media; Cysteine Proteinase Inhibitors; Enzyme Induction; Genes, Reporter; Hydrogen-Ion Concentration; Leupeptins; Macrophages, Peritoneal; Male; NF-kappa B; Nitric Oxide; Nitric Oxide Synthase; Nitric Oxide Synthase Type II; Nitrites; Pyrrolidines; Rats; Rats, Sprague-Dawley; Thiocarbamates; Transcription, Genetic; Tumor Necrosis Factor-alpha | 1998 |
Calcium ionophore-induced degradation of neurofilament and cell death in MSN neuroblastoma cells.
Extensive necrotic death of MSN neuroblastoma cells could be induced after incubation with the calcium ionophore, A23187. The reaction was concentration-dependent and time course-dependent. Levels of the 66 kd/alpha-internexin neurofilament protein (NF-66) and the cognate heat shock protein 70 (Hsc 70) decreased during the Ca2+-activated cell death. Addition of the calcium chelator, ethylene glycol-bis(beta-aminoethyl ether) N,N,N',N'-tetraacetic acid (EGTA) restored the normal level of NF-66 and partially that of the Hsc 70. Use of either calpain I or calpain II inhibitor could alleviate the reduction of 66 kd protein during the ionophore treatment whereas only calpain I inhibitor treatment was effective in restoring the normal level of the Hsc 70. Neither of these calpain inhibitors could block the ionophore triggered cell death. EGTA was toxic to cells in a wide range of concentration suggesting a calcium-independent activation of cell death mechanism. Topics: Calcimycin; Calpain; Carrier Proteins; Cell Death; HSC70 Heat-Shock Proteins; HSP70 Heat-Shock Proteins; Humans; Intermediate Filament Proteins; Leupeptins; Nerve Tissue Proteins; Neuroblastoma; Neurofibrils; Oligopeptides; Tumor Cells, Cultured | 1998 |
Defect in an intrahepatic degradation of apolipoprotein B in suncus: an animal model of hypobetalipoproteinemia.
We have previously shown that fatty liver is easily induced in suncus by starvation and that the plasma level of apolipoprotein B (apo B) is very low. We also found that hepatic acyl coenzyme A cholesterol acyltransferase (ACAT) activity is almost absent in the animals, resulting in decreased cholesteryl ester contents in the liver. A deficiency of cholesteryl ester in suncus liver may be one of the reasons for the defect in the assembly process of apo B-containing lipoproteins, leading to a low level of plasma apo B. Another possible explanation for the induction of fatty liver in suncus is a defect in apo B-processing in the liver. In this study, we investigated the hepatic synthetic rate and intrahepatic degradation of apo B using primary cultured hepatocytes derived from suncus and rats. In order to estimate intrahepatic degradation of apo B, we added N-acetylleucyl-leucynorleucinal to the culture medium as an inhibitor of apo B degradation. The basal synthesis of apo B in suncus hepatocytes was 50% of that in rat. Intracellular degradation of apo B was not observed in suncus hepatocytes, while it was obvious in rat hepatocytes. This evidence suggests that the lower secretion rate of apo B lipoprotein is not due to the intrahepatic degradation of apo B, but may be due to the low synthetic rate of apo B. Topics: Animals; Apolipoproteins B; Cells, Cultured; Disease Models, Animal; Hypobetalipoproteinemias; Leupeptins; Liver; Male; Rats; Shrews | 1998 |
Interleukin-3-induced activation of the JAK/STAT pathway is prolonged by proteasome inhibitors.
One facet of cytokine receptor signaling involves the activation of signal transducers and activators of transcription (STATs). STATs are rapidly activated via tyrosine phosphorylation by Janus kinase (JAK) family members and subsequently inactivated within a short period. We investigated the effect of proteasome inhibition on interleukin-3 (IL-3) activation of the JAK/STAT pathway following stimulation of Ba/F3 cells. Treatment of Ba/F3 cells with the proteasome inhibitor, N-acetyl-L-leucinyl-L-leucinyl-norleucinal (LLnL), led to stable tyrosine phosphorylation of the IL-3 receptor, beta common (betac), and STAT5 following stimulation. The effects of LLnL were not restricted to the JAK/STAT pathway, as Shc and mitogen-activated protein kinase (MAPK) phosphorylation were also prolonged in LLnL-treated cells. Further investigation showed these stable phosphorylation events were the result of prolonged activation of JAK2 and JAK1. These observations were confirmed using pharmacologic inhibitors. In the presence of LLnL, stable phosphorylation of STAT5 and betac was abrogated if the tyrosine kinase inhibitor, staurosporine, was added. The effect of staurosporine on STAT5 phosphorylation could be overcome if the phosphatase inhibitor, vanadate, was also added, suggesting phosphorylated STAT5 could be stabilized by phosphatase, but not by proteasome inhibition per se. These observations are consistent with the hypothesis that proteasome-mediated protein degradation can modulate the activity of the JAK/STAT pathway by regulating the deactivation of JAK. Topics: Animals; Cell Line; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Enzyme Activation; Enzyme Inhibitors; Interleukin-3; Janus Kinase 2; Leupeptins; Mice; Milk Proteins; Multienzyme Complexes; Phosphoprotein Phosphatases; Proteasome Endopeptidase Complex; Protein-Tyrosine Kinases; Proto-Oncogene Proteins; Receptors, Interleukin-3; Signal Transduction; STAT5 Transcription Factor; Staurosporine; Trans-Activators; Ubiquitins; Vanadates | 1998 |
Inhibitory function of p21Cip1/WAF1 in differentiation of primary mouse keratinocytes independent of cell cycle control.
The cyclin-dependent kinase inhibitor p21(Cip1/WAF1) has been implicated as an inducer of differentiation. However, although expression of p21 is increased in postmitotic cells immediately adjacent to the proliferative compartment, its expression is decreased in cells further along the differentiation program. Expression of the p21 protein was decreased in terminally differentiated primary keratinocytes of mice, and this occurred by a proteasome-dependent pathway. Forced expression of p21 in these cells inhibited the expression of markers of terminal differentiation at both the protein and messenger RNA levels. These inhibitory effects on differentiation were not observed with a carboxyl-terminal truncation mutant or with the unrelated cyclin-dependent kinase inhibitor p16(INK4a), although all these molecules exerted similar inhibition of cell growth. These findings reveal an inhibitory role of p21 in the late stages of differentiation that does not result from the effects of p21 on the cell cycle. Topics: Acetylcysteine; Adenoviridae; Animals; Animals, Newborn; Cell Cycle; Cell Differentiation; Cells, Cultured; Cyclin-Dependent Kinase Inhibitor p21; Cyclin-Dependent Kinases; Cyclins; Enzyme Inhibitors; Gene Expression Regulation; Keratinocytes; Leupeptins; Membrane Proteins; Mice; Mutation; Promoter Regions, Genetic; Protein Precursors; RNA, Messenger; Succinates; Transfection | 1998 |
Rapamycin inhibition of the G1 to S transition is mediated by effects on cyclin D1 mRNA and protein stability.
The immunosuppressant rapamycin has been shown previously to inhibit the G1/S transition in several cell types by prolonging the G1 phase of the cell cycle. This process appears to be controlled, in part, by the rapamycin-sensitive FK506-binding protein-rapamycin-associated protein-p70 S6 kinase (p70(S6k)) pathway and the cyclin-dependent kinases (Cdk). We now show that in serum-stimulated NIH 3T3 cells, rapamycin treatment delays the accumulation of cyclin D1 mRNA during progression through G1. Rapamycin also appears to affect stability of the transcript. The combined transcriptional and post-transcriptional effects of the drug ultimately result in decreased levels of cyclin D1 protein. Moreover, degradation of newly synthesized cyclin D1 protein is accelerated by rapamycin, a process prevented by inclusion of the proteasome inhibitor, N-acetyl-Leu-Leu-norleucinal. The overall effect of rapamycin on cyclin D1 leads, in turn, to impaired formation of active complexes with Cdk4, a process which triggers retargeting of the p27(Kip1) inhibitor to cyclin E/Cdk2. In view of this novel experimental evidence, we discuss a possible mechanism for the rapamycin-induced cell cycle arrest at the G1/S transition. Topics: 3T3 Cells; Animals; Apoptosis; Carrier Proteins; Cell Cycle; Cell Cycle Proteins; Cyclin D1; Cyclin-Dependent Kinase Inhibitor p27; Cyclin-Dependent Kinases; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; G1 Phase; Heat-Shock Proteins; Immunosuppressive Agents; Leupeptins; Mice; Microtubule-Associated Proteins; Phosphorylation; Polyenes; Retinoblastoma Protein; Ribosomal Protein S6 Kinases; RNA, Messenger; Sirolimus; Tacrolimus Binding Proteins; Transcription, Genetic; Tumor Suppressor Proteins | 1998 |
Effects of tocotrienol on the intracellular translocation and degradation of apolipoprotein B: possible involvement of a proteasome independent pathway.
gamma-Tocotrienol (gamma-T3), a HMG CoA reductase inhibitor, was previously shown to stimulate the intracellular degradation of apolipoprotein B (apoB) in HepG2 cells. The aim of this study was to explore the effects of gamma-T3 on the proteasome dependent co-translational degradation and the proteasome independent post-translational degradation of apoB. Previous studies have shown that apoB translocation across the endoplasmic reticulum (ER) membrane governs the co-translational degradative pathway of apoB. Therefore, we first examined the effects of gamma-T3 on this pathway using a specific translocation assay derived from HepG2 cells. Our results indicated that gamma-T3 reduced the efficiency of apoB translocation across the ER membrane, suggesting that co-translational degradation may be partially involved. Evidence of an ER associated post-translational degradation was also provided upon pre-treating digitonin-permeabilized HepG2 cells with a proteasome inhibitor, lactacystin. When chased for 2h, ER degradation of apoB was observed and was further enhanced in the presence of gamma-T3 versus untreated control, in spite of proteasome inhibition. Combined with the ability of ALLN, a proteasome and cysteine protease inhibitor, to block the post-translational degradation of apoB, the data suggest that gamma-T3 diverted more apoB to a cytosolic proteasomal dependent and possibly an ER-associated proteasomal independent degradation pathways. Topics: Acetylcysteine; Apolipoproteins B; Biological Transport; Cell Membrane Permeability; Chromans; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Endoplasmic Reticulum; Humans; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Leupeptins; Liver; Multienzyme Complexes; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; Tumor Cells, Cultured; Vitamin E | 1998 |
IL-1beta-induced monocyte chemoattractant protein-1 gene expression in endothelial cells is blocked by proteasome inhibitors.
Human monocyte chemoattractant protein-1 (MCP-1) is expressed by a variety of cell types in response to various stimuli. MCP-1 expressed by the endothelium plays an important role in cell migration and activation. MCP-1 is a major chemoattractant for monocytes, T lymphocytes, and basophils. In the present study, we present evidence that the proteasome complex is involved in mediating the interleukin (IL)-1beta induction of MCP-1 in endothelial cells. We present evidence that a proteasome inhibitor, N-acetyl-leucinyl-leucinyl-norleucinal (norLeu), and the protease inhibitor tosyl-Phe-chloromethylketone (TPCK) block IL-1beta induction of MCP-1 protein expression. norLeu and TPCK also blocked IL-1beta-induced MCP-1 promoter-driven reporter gene expression as well as nuclear factor (NF)-kappaB-mediated reporter gene expression. The effects of norLeu were due to its inhibition of the proteasome rather than calpain, because other calpain inhibitors had no effect on MCP-1 expression. In contrast to TPCK, which blocked NF-kappaB translocation to the nucleus, norLeu had no effect on NF-kappaB nuclear translocation or IL-1beta-induced phosphorylation of p65. This study demonstrates that the proteasome pathway is involved in IL-1beta-induced MCP-1 gene expression in human endothelial cells. Topics: Cells, Cultured; Chemokine CCL2; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Endothelium, Vascular; Gene Expression Regulation; Humans; Interleukin-1; Leupeptins; Multienzyme Complexes; NF-kappa B; Proteasome Endopeptidase Complex; Serine Proteinase Inhibitors; Tosylphenylalanyl Chloromethyl Ketone | 1998 |
Proteasomes can either generate or destroy MHC class I epitopes: evidence for nonproteasomal epitope generation in the cytosol.
Proteasomes have been implicated in the production of the majority of peptides that associate with MHC class I molecules. We used two different proteasome inhibitors, the peptide aldehyde N-acetyl-L-leucyl-L-leucyl-L-norleucinal (LLnL) and the highly specific inhibitor lactacystin, to examine the role of proteasomes in generating peptide epitopes associated with HLA-A*0201. Neither LLnL nor lactacystin was able to completely block the expression of the HLA-A*0201. Furthermore, the effects of LLnL and lactacystin on the expression of different categories of specific epitopes, TAP independent vs TAP dependent and derived from either cytosolic or membrane proteins, were assessed. As predicted, presentation of two TAP-dependent epitopes was blocked by LLnL and lactacystin, while a TAP-independent epitope that is processed in the endoplasmic reticulum was unaffected by either inhibitor. Surprisingly, both LLnL and lactacystin increased rather than inhibited the expression of a cytosolically transcribed and TAP-dependent peptide from the influenza A virus M1 protein. Mass spectrometric analyses of in vitro proteasome digests of a synthetic 24 mer containing this epitope revealed no digestion products of any length that included the intact epitope. Instead, the major species resulted from cleavage sites within the epitope. Although cleavage at these sites was inhibitable by LLnL and lactacystin, epitope-containing species were still not produced. We conclude that proteasomes may in some cases actually destroy epitopes that would otherwise be destined for presentation by class I molecules. These results suggest that some epitopes are generated by nonproteasomal proteases in the cytosol. Topics: Acetylcysteine; Amino Acid Sequence; Animals; ATP Binding Cassette Transporter, Subfamily B, Member 2; ATP-Binding Cassette Transporters; Cell Line; Cell-Free System; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytosol; Cytotoxicity, Immunologic; Epitopes, T-Lymphocyte; Glycine; Histocompatibility Antigens Class I; HLA-A Antigens; Humans; Leupeptins; Membrane Proteins; Mice; Molecular Sequence Data; Multienzyme Complexes; Proteasome Endopeptidase Complex; T-Lymphocytes, Cytotoxic; Time Factors; Viral Matrix Proteins | 1998 |
Cell cycle analysis and synchronization of the Xenopus cell line XL2.
We have determined the length of the cell cycle and its different phases in a permanent Xenopus tadpole cell line, XL2. Following BrdU labeling, the total length of the cell cycle was estimated as 28 h. The different phases of the cell cycle, G1, S, G2, and M were, respectively, 14 h, 10 h 45 min, 2 h 30 min, and 54 min. Knowing these parameters, we were able to develop methods that selectively enrich cells in different phases of the cycle. Treatment with aphidicolin resulted in a S phase block in which more than 85% of the cells showed S phase chromosomes. Almost 60% of the cells were arrested in mitosis after a double block with aphidicolin/nocodazole or aphidicolin/ALLN (acetyl-leucyl-leucyl-norleucinal) treatment. This synchronization protocol will greatly facilitate studies of biochemical events associated with specific gene regulation through the cell cycle. Our synchronization protocol does not disturb cell metabolism as the expression of cyclin B2 during the cell cycle is in agreement with the results obtained with mammalian cells. Topics: Animals; Aphidicolin; Cell Cycle; Cell Line; Cyclin B; Leupeptins; Mitosis; Nocodazole; S Phase; Tubulin; Xenopus laevis | 1998 |
Involvement of proteasomes in migration and matrix metalloproteinase-9 production of oral squamous cell carcinoma.
We investigated whether proteasomes were involved in the invasiveness of oral squamous cell carcinoma (SCC) cells. The migration of SCC cells through a gelatin-coated membrane was enhanced with tumor necrosis factor alpha (TNF alpha), which was strongly inhibited by a peptide aldehyde, N-acetyl-Leu-Leu-norleucinal (ALLN), but not by its structurally related compound, N-acetyl-Leu-Leu-methioninal (ALLM). Since ALLN is a more potent inhibitor against proteasomal proteolysis than ALLM, cell migration inhibited by ALLN may thus likely depend on proteasomes. The TNF alpha-induced migration through gelatin appeared to be associated with the gelatinolytic activity from the cells, since TNF alpha strongly enhanced the production of matrix metalloproteinase (MMP)-9/gelatinase B in the SCC cells, as detected by gelatin zymography. The production of MMP-9 was also inhibited by pretreatment with ALLN, but not ALLM, in a dose-dependent manner. Moreover, ALLN could block the activation and nuclear translocation of a transcription-activating factor, NF-kappaB, which is known to regulate MMP-9 expression in TNF alpha-stimulated SCC cells. The TNF alpha-induced degradation of IkappaB alpha was also suppressed by ALLN treatment, thus implying that the molecule linking proteasome to MMP-9 production should be IkappaB alpha. We finally reconfirmed the involvement of proteasomes in the invasive behavior of oral SCC using lactacystin, a specific proteasome inhibitor, which could prevent TNF alpha from enhancing MMP-9 production, NF-kappaB activation, induction of MMP-9 mRNA and cell migration. Topics: Carcinoma, Squamous Cell; Collagenases; Cysteine Proteinase Inhibitors; Gelatinases; Humans; Leupeptins; Matrix Metalloproteinase 2; Matrix Metalloproteinase 9; Matrix Metalloproteinase Inhibitors; Metalloendopeptidases; Mouth Neoplasms; Neoplasm Invasiveness; Neoplasm Proteins; NF-kappa B; Oligopeptides; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha | 1998 |
Proteasome blockers inhibit protein breakdown in skeletal muscle after burn injury in rats.
1. Burn injury stimulates ubiquitin-dependent protein breakdown in skeletal muscle. The 20S proteasome is the proteolytic core of the 26S proteasome that degrades ubiquitin conjugates. We examined the effects of the proteasome inhibitors N-acetyl-L-leucinyl-L-leucinal-L-norleucinal (LLnL), lactacystin and beta-lactone on protein breakdown in muscles from burned rats. 2. A full-thickness burn of 30% total body surface area was inflicted on the back of rats. Control rats underwent a sham procedure. After 24 h, extensor digitorum longus muscles were incubated in the absence or presence of 20S proteasome blocker and protein turnover rates and ubiquitin mRNA levels were determined. 3. LLnL resulted in a dose- and time-dependent inhibition of total protein breakdown in incubated muscles from burned rats. Lactacystin and beta-lactone blocked both total and myofibrillar muscle protein breakdown. In addition to inhibiting protein breakdown, LLnL increased ubiquitin mRNA levels, possibly reflecting inhibited proteasome-associated RNase activity. 4. Inhibited muscle protein breakdown caused by LLnL, lactacystin and beta-lactone supports the concept that the ubiquitin-proteasome pathway plays a central role in burn-induced muscle proteolysis. Because the proteasome has multiple important functions in the cell, in addition to regulating general protein breakdown, further studies are needed to test the role of proteasome blockers in the treatment or prevention of muscle catabolism. Topics: Acetylcysteine; Animals; Burns; Culture Techniques; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Lactones; Leupeptins; Male; Muscle Proteins; Muscle, Skeletal; Rats; Rats, Sprague-Dawley; RNA, Messenger; Time Factors; Tyrosine; Ubiquitins | 1998 |
Annexin VI-mediated loss of spectrin during coated pit budding is coupled to delivery of LDL to lysosomes.
Previously we reported that annexin VI is required for the budding of clathrin-coated pits from human fibroblast plasma membranes in vitro. Here we show that annexin VI bound to the NH2-terminal 28-kD portion of membrane spectrin is as effective as cytosolic annexin VI in supporting coated pit budding. Annexin VI-dependent budding is accompanied by the loss of approximately 50% of the spectrin from the membrane and is blocked by the cysteine protease inhibitor N-acetyl-leucyl-leucyl-norleucinal (ALLN). Incubation of fibroblasts in the presence of ALLN initially blocks the uptake of low density lipoprotein (LDL), but the cells recover after 1 h and internalize LDL with normal kinetics. The LDL internalized under these conditions, however, fails to migrate to the center of the cell and is not degraded. ALLN-treated cells have twice as many coated pits and twofold more membrane clathrin, suggesting that new coated pits have assembled. Annexin VI is not required for the budding of these new coated pits and ALLN does not inhibit. Finally, microinjection of a truncated annexin VI that inhibits budding in vitro has the same effect on LDL internalization as ALLN. These findings suggest that fibroblasts are able to make at least two types of coated pits, one of which requires the annexin VI-dependent activation of a cysteine protease to disconnect the clathrin lattice from the spectrin membrane cytoskeleton during the final stages of budding. Topics: Annexin A6; Clathrin; Coated Pits, Cell-Membrane; Cysteine Proteinase Inhibitors; Cytoskeleton; Endocytosis; Fibroblasts; Humans; Leupeptins; Lipoproteins, LDL; Lysosomes; Membrane Proteins; Microscopy, Fluorescence; Peptide Fragments; Protein Binding; Recombinant Proteins; Spectrin | 1998 |
The Ras target AF-6 is a substrate of the fam deubiquitinating enzyme.
The Ras target AF-6 has been shown to serve as one of the peripheral components of cell-cell adhesions, and is thought to participate in cell-cell adhesion regulation downstream of Ras. We here purified an AF-6-interacting protein with a molecular mass of approximately 220 kD (p220) to investigate the function of AF-6 at cell-cell adhesions. The peptide sequences of p220 were identical to the amino acid sequences of mouse Fam. Fam is homologous to a deubiquitinating enzyme in Drosophila, the product of the fat facets gene. Recent genetic analyses indicate that the deubiquitinating activity of the fat facets product plays a critical role in controlling the cell fate. We found that Fam accumulated at the cell-cell contact sites of MDCKII cells, but not at free ends of plasma membranes. Fam was partially colocalized with AF-6 and interacted with AF-6 in vivo and in vitro. We also showed that AF-6 was ubiquitinated in intact cells, and that Fam prevented the ubiquitination of AF-6. Topics: Animals; Brain; Cattle; Cell Adhesion; Cell Line; Cysteine Endopeptidases; Drosophila Proteins; Endopeptidases; Fluorescent Antibody Technique; Genes, ras; Immunohistochemistry; Kinesins; Leupeptins; Multienzyme Complexes; Myosins; Proteasome Endopeptidase Complex; Recombinant Fusion Proteins; Transfection; Ubiquitin Thiolesterase; Ubiquitins | 1998 |
Enhanced in vitro cytotoxicity and cytostasis of the combination of onconase with a proteasome inhibitor.
In proliferating cells the turnover rate of proteins responsible for regulation of the cell cycle progression, namely cyclins and inhibitors of the cyclin-dependent kinases (CDKs) and phosphatases, is rapid and their cellular level is modulated at the transcriptional, translational and/or degradation (via proteasome pathway) stages. Inhibition of proteasome function results in accumulation of rapidly turning over proteins and, thus, causes an imbalance of the cell cycle regulatory components, and loss of their regulatory function. Indeed, it has been shown that proteasome inhibitors perturb the cell cycle progression. Onconase, a novel RNase which has anti-tumor activity and is in clinical trials, has previously been shown to suppress protein synthesis, presumably by degradation of intracellular RNA, preferentially tRNA. By interfering with regulation of expression of cyclins and/or CDK-inhibitors, onconase also may induce the imbalance of these proteins and potentiate the effect of proteasome inhibitors. In the present study, we observed that the combinations of onconase with peptide-aldehyde inhibitors of calpain and proteasome such as the N-acetyl-leucinyl-leucinyl-norleucinal (LLnL) and the N-acetyl-leucinyl-valinyl-phenylalaninal (LVP), but not N-acetyl-leucinyl-leucinyl-methioninal (LLM), were synergistic in suppressing cell proliferation and inducing apoptosis in three human tumor cell lines: A-549 lung adenocarcinoma, DU-145 prostatic carcinoma, and MDA-MB-231 breast carcinoma. The observed cytotoxicity may also be a result of prevention of the induction of the 'survival' genes by the nuclear factor kappaB (NFkappaB) by onconase and proteasome inhibitors. The data indicate that such combinations should be further tested as potential anti-cancer regimens. Topics: Antineoplastic Agents; Clone Cells; Colony-Forming Units Assay; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Doxorubicin; Drug Synergism; Egg Proteins; Flow Cytometry; Formazans; Humans; Leupeptins; Multienzyme Complexes; Oligopeptides; Proteasome Endopeptidase Complex; Ribonucleases; Tetrazolium Salts; Toxicity Tests; Tumor Cells, Cultured | 1998 |
Proteolysis of connexin43-containing gap junctions in normal and heat-stressed cardiac myocytes.
The present studies were performed to examine the degradation of connexin43-containing gap junctions by the lysosome or the proteasome in normal and heat-stressed cultures of neonatal rat ventricular myocytes.. Primary cultures were prepared from neonatal rat ventricular myocytes. Connexin43 was detected by immunoblotting, immunofluorescence, or immunoprecipitation. Gap junction profiles were detected by transmission electron microscopy.. Immunoblots of whole cell lysates demonstrated increased levels of connexin43 in cultures treated with lysosomal inhibitors (chloroquine, leupeptin, E-64, or ammonium chloride) or proteasomal inhibitors (lactacystin or ALLN). Pulse-chase experiments showed that the half-life of connexin43 was 1.4 h in control cultures, but was prolonged to 2.0 or 2.8 h in cultures treated with chloroquine or lactacystin, respectively. Immunofluorescence and electron microscopy showed a significant increase in the number of gap junction profiles in myocytes treated with either chloroquine or lactacystin. Heat treatment of cultures (43.5 degrees C for 30 min) produced a rapid loss of connexin43 as detected by immunoblotting or immunofluorescence. Heat-induced connexin43 degradation was prevented by simultaneous treatment with lactacystin, ALLN, or chloroquine. Connexin43 levels and distribution returned to normal by 3 h following a heat shock and were resistant to a subsequent repeat heat stress. The heat shock also led to production of HSP70 as detected by immunoblotting.. These data suggest that Cx43 gap junctions in myocytes are degraded by the proteasome and the lysosome, that this proteolysis can be augmented by heat stress, and that inducible factors such as HSP70 may protect against Cx43 degradation. Topics: Acetylcysteine; Ammonium Chloride; Animals; Cells, Cultured; Chloroquine; Connexin 43; Cysteine Proteinase Inhibitors; Fluorescent Antibody Technique; Gap Junctions; Hot Temperature; HSP70 Heat-Shock Proteins; Immunoblotting; Leucine; Leupeptins; Lysosomes; Microscopy, Electron; Myocardium; Oligopeptides; Precipitin Tests; Protease Inhibitors; Rats | 1998 |
Proteasomes regulate erythropoietin receptor and signal transducer and activator of transcription 5 (STAT5) activation. Possible involvement of the ubiquitinated Cis protein.
Cis is an Src homology 2 domain-containing protein, which binds to the erythropoietin receptor and decreases erythropoietin-stimulated cell proliferation. We show that Cis associates with the second tyrosine residue of the intracellular domain of the erythropoietin receptor (Tyr401). Two forms of Cis with molecular masses of 32 and 37 kDa were detected, and we demonstrate that the 37-kDa protein resulted from post-translational modifications of the 32-kDa form. Anti-ubiquitin antibodies recognized the 37-kDa form of Cis and the proteasome inhibitors N-acetyl-leucyl-leucyl-norleucinal and lactacystin inhibited its degradation, showing that the 37-kDa form of Cis is a ubiquitinated protein, which seems to be rapidly degraded by the proteasome. In erythropoietin-stimulated UT-7 cells, the activation of the erythropoietin receptor and signal transducer and activator of transcription 5 (STAT5) was transient and returned to basal levels after 30-60 min of erythropoietin stimulation. In contrast, these proteins remained strongly phosphorylated, and STAT5 remained activated for at least 120 min in the presence of proteasome inhibitors. These experiments demonstrate that the proteasomes are involved in the down-regulation of the erythropoietin receptor activation signals. Because the proteasome inhibitors induced the accumulation of both the ubiquitinated form of Cis and the Cis-erythropoietin receptor complexes, our results suggest that the ubiquitinated form of Cis could be involved in the proteasome-mediated inactivation of the erythropoietin receptor. Topics: Acetylcysteine; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Humans; Immediate-Early Proteins; Leupeptins; Milk Proteins; Multienzyme Complexes; Proteasome Endopeptidase Complex; Protein Binding; Protein Processing, Post-Translational; Receptors, Erythropoietin; Signal Transduction; src Homology Domains; STAT5 Transcription Factor; Suppressor of Cytokine Signaling Proteins; Trans-Activators; Tyrosine; Ubiquitins | 1998 |
Constitutive apoptosis in human neutrophils requires synergy between calpains and the proteasome downstream of caspases.
Programmed cell death invariably requires the activation of proteolytic cascades that are not yet well defined but are initiated after apical caspase activation. We provide evidence that calpains and the proteasome function synergistically downstream of caspases to assist the constitutive apoptotic program of aging neutrophils, which plays an important role in resolution of inflammatory responses. Inhibitor studies indicated that "tethering" of preapoptotic senescent neutrophils to human macrophages required caspase activity. However, the development of morphological features characteristic of apoptosis, including nuclear morphology, PS exposure, surface protein shedding, and the capacity to be ingested by macrophages, required the downstream action of either calpains or the proteasome. Calpain activities were constitutively active in freshly isolated neutrophils and responsible for rearrangements in the protein composition and structure of the plasmalemmal cytoskeleton as they aged in culture and underwent apoptosis. This included a dissociation of protein(s) from F-actin, a candidate mechanism for increased susceptibility to cleavage, and a loss in immunodetectable alpha-actinin and ezrin, two actin-binding, membrane-anchoring proteins. These results clarify roles for different classes of proteases in a physiologically important form of constitutive apoptosis. Topics: Actinin; Actins; Apoptosis; Calpain; Caspases; Cell Communication; Cells, Cultured; Cellular Senescence; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytoskeletal Proteins; Dipeptides; Drug Synergism; Humans; Leupeptins; Multienzyme Complexes; Neutrophils; Phosphoproteins; Proteasome Endopeptidase Complex | 1998 |
Calpain involvement in calphostin C-induced apoptosis.
A major problem in assessing the role of calpains in apoptosis induction concerns the fact that calpain inhibitors can also impair the activity of the proteasome, also reported to be involved in apoptosis. Herein we showed that apoptosis induced by calphostin C in U937 human promonocytic leukemia cells was associated, at its onset, with enhanced protein (poly)ubiquitination. This observation prompted us to study whether protein degradation through the ubiquitin/proteasome pathway was involved in apoptosis induction. We found that N-acetyl-Leu-Leu-norleucinal (50 microM), a proteasome as well as a calpain inhibitor, was able to reduce calphostin C-induced apoptosis by approximately 60%, whereas lactacystin (10 microM), a specific proteasome inhibitor, was ineffective. These results suggest that calphostin C-induced apoptosis is partly calpain-mediated, but does not require protein degradation through the ubiquitin/proteasome pathway. Topics: Acetylcysteine; Antibiotics, Antineoplastic; Apoptosis; Calpain; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Humans; Leupeptins; Multienzyme Complexes; Naphthalenes; Neoplasm Proteins; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; U937 Cells; Ubiquitins | 1998 |
Coupling of mitosis to the completion of S phase through Cdc34-mediated degradation of Wee1.
The dependence of mitosis on the completion of the period of DNA replication in the cell cycle [synthesis (S) phase] ensures that chromosome segregation occurs only after the genome has been fully duplicated. A key negative regulator of mitosis, the protein kinase Wee1, was degraded in a Cdc34-dependent fashion in Xenopus egg extracts. This proteolysis event was required for a timely entrance into mitosis and was inhibited when DNA replication was blocked. Therefore, the DNA replication checkpoint can prevent mitosis by suppressing the proteolysis of Wee1 during S phase. Topics: Anaphase-Promoting Complex-Cyclosome; Animals; Aphidicolin; cdc25 Phosphatases; Cell Cycle Proteins; Cell Nucleus; Cyclin-Dependent Kinase Inhibitor p27; DNA Replication; Female; G2 Phase; Leupeptins; Ligases; Male; Maturation-Promoting Factor; Microtubule-Associated Proteins; Mitosis; Nuclear Proteins; Okadaic Acid; Ovum; Phosphoprotein Phosphatases; Protein Serine-Threonine Kinases; Protein-Tyrosine Kinases; Proteins; Recombinant Proteins; S Phase; Spermatozoa; Tumor Suppressor Proteins; Ubiquitin-Protein Ligase Complexes; Ubiquitin-Protein Ligases; Ubiquitins; Xenopus; Xenopus Proteins | 1998 |
Characterization of two Chinese hamster ovary cell lines expressing the COOH-terminal domains of sterol regulatory element-binding protein (SREBP)-1.
Sterol regulatory element-binding proteins (SREBPs) regulate transcription of genes encoding enzymes in the cholesterol biosynthetic pathway and the LDL receptor. These proteins are synthesized as membrane-bound precursors and processed to generate the NH2-terminal domains, mature transcription factors. We established two Chinese hamster ovary (CHO) cell lines, CHO-421 expressing the truncated hamster SREBP-1 (amino acids 421-1133) with two transmenbrane domains and CHO-557 expressing another truncated SREBP-1 (amino acids 557-1133) without any transmembrane domains, to investigate the fate of the COOH terminus after cleavage of the NH2-terminal mature SREBP. The cell fractionation experiments revealed that the two proteins, regardless of the absence of transmembrane domains in the SREBP (557-1133), similarly localized in the nuclear envelope and the microsomal membrane fractions, suggesting that these proteins appear to be tightly bound to a membrane protein(s) localizing on the nuclear and endoplasmic reticulum (ER) membranes. Although we predicted that overexpression of the COOH-terminal domains, which were thought to be involved in the regulation of SREBP processing, would result in disruption of the SREBP-dependent transcriptional regulation of several genes, the mRNA levels for 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) synthase in these two cell lines were regulated in a sterol-dependent manner. Pulse-chase experiments revealed that the SREBP (421-1133) was relatively rapidly degraded (t1/2 = 4-6 hr) and that the intracellular cholesterol level did not affect the half-life time. The degradation of the SREBP (421-1133) was not suppressed by the treatment of a calpain inhibitor, N-acetyl-leucyl-leucyl-norleucynal (ALLN), which blocks the proteolysis of some proteins within or near the ER. In CHO-557 cells the SREBP (557-1133) was much more rapidly degraded (t1/2 = 1-2 hr), suggesting that the cytosolic COOH-terminal domain is accessible to the enzymatic attacks from the cytoplasm. Taken together, overexpression of the COOH-terminal domains does not affect the regulation of SREBP processing and the domains are rapidly turned over by the cytosolic proteolytic process distinct from the ALLN-sensitive ER degradative pathway. Topics: Animals; Calpain; CCAAT-Enhancer-Binding Proteins; Cell Nucleus; CHO Cells; Cricetinae; DNA-Binding Proteins; Electrophoresis, Polyacrylamide Gel; Endoplasmic Reticulum; Gene Deletion; Hydroxymethylglutaryl-CoA Synthase; Leupeptins; Microsomes; Molecular Weight; Nuclear Proteins; Peptide Fragments; RNA, Messenger; Sterol Regulatory Element Binding Protein 1; Transcription Factors; Transfection | 1998 |
The proteasome-specific inhibitor lactacystin blocks presentation of cytotoxic T lymphocyte epitopes in human and murine cells.
We describe the effect of the proteasome specific inhibitor lactacystin on the metabolic stability of influenza nucleoprotein (NP) and on the generation of antigens presented by human and murine class I molecules of the major histocompatibility complex to cytotoxic T lymphocytes (CTL). We show that cells treated with lactacystin fail to present influenza antigens to influenza-specific CTL, but retain the capacity to present defined epitopes expressed as peptides intracellularly by recombinant vaccinia viruses. This block in antigen presentation can be overcome by expressing the viral protein within the lumen of the endoplasmic reticulum, confirming the specificity of lactacystin for cytosolic proteases. We also show that the effect of lactacystin on antigen presentation correlates with the block of breakdown of a rapidly degraded form of the influenza NP linked to ubiquitin. These results demonstrate that proteasome-dependent degradation plays an important role in the cytosolic generation of CTL epitopes. Topics: Acetylcysteine; Animals; Antigen-Presenting Cells; Antigens, Viral; Cells, Cultured; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytosol; Epitopes; H-2 Antigens; Humans; Leupeptins; Mice; Multienzyme Complexes; Nucleocapsid Proteins; Nucleoproteins; Orthomyxoviridae; Peptides; Proteasome Endopeptidase Complex; Signal Transduction; T-Lymphocytes, Cytotoxic; Vaccinia virus; Viral Core Proteins | 1997 |
N-acetyl-leucinyl-leucinyl-norleucinal inhibits lipopolysaccharide-induced NF-kappaB activation and prevents TNF and IL-6 synthesis in vivo.
The effects of N-acetyl-leucinyl-leucinyl-norleucinal (ALLN), a potent inhibitor of proteolysis catalyzed by proteasomes, on the activation of NF-kappaB in vitro and in vivo have been examined. Confirming earlier observations, ALLN inhibits the activation of NF-kappaB in macrophage cultures stimulated with LPS, resulting in the intracellular accumulation of IkappaB and p105. The synthesis of TNF, a reaction dependent upon NF-kappaB activation, is blocked by ALLN. Treatment of mice with LPS results in the induction of TNF and IL-6 within 90 min followed by lethal shock at 24 hr. In mice pretreated with ALLN, serum TNF and IL-6 levels were significantly lower than those in untreated animals. These studies suggest that the proteasome is a novel target for the identification of agents that may be useful in the treatment of those diseases whose etiology is dependent on the activation of NF-kappaB. Topics: Animals; Cells, Cultured; Feasibility Studies; Female; Gene Expression Regulation; Interleukin-6; Leupeptins; Lipopolysaccharides; Macrophages; Mice; NF-kappa B; Sepsis; Tumor Necrosis Factor-alpha | 1997 |
Misfolded major histocompatibility complex class I heavy chains are translocated into the cytoplasm and degraded by the proteasome.
N-acetyl-L-leucyl-L-leucyl-L-norleucinal (LLnL), which reversibly inhibits the proteasome in addition to other proteases, and a more specific irreversible inhibitor of the proteasome, lactacystin, were found to cause the accumulation of major histocompatibility complex (MHC) class I heavy chains in the cytosol of the beta2-microglobulin-deficient cell line Daudi and the TAP-deficient cell line .174. These cell lines, which are severely impaired in their ability to fold MHC class I heavy chain, showed an accumulation of soluble class I heavy chains at different rates over a period of hours in the presence of LLnL. The accumulation of soluble class I heavy chains in the presence of either LLnL or lactacystin was easily revealed in Daudi and .174 but almost undetectable in a Daudi transfectant expressing beta2-microglobulin and in 45.1, the wild-type parent of .174. The soluble class I heavy chain was also found to be devoid of its N-linked glycan and to be located in the cytosol. When the gene for ICP47, a herpes simplex virus protein that blocks the translocation of peptides into the endoplasmic reticulum, was transfected into 45.1, a similar accumulation of soluble MHC class I heavy chain was detectable. These data suggest that in cells where the MHC class I molecule is unable to assemble properly, the misfolded heavy chain is removed from the endoplasmic reticulum to the cytosol, deglycosylated, and degraded by the proteasome. Topics: Acetylcysteine; beta 2-Microglobulin; Blotting, Western; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytoplasm; Endoplasmic Reticulum; Enzyme Inhibitors; Glycosylation; Histocompatibility Antigens Class I; Humans; Kinetics; Leupeptins; Multienzyme Complexes; Proteasome Endopeptidase Complex; Protein Folding; Solubility; Transfection; Tumor Cells, Cultured | 1997 |
Proteasome inhibition leads to a heat-shock response, induction of endoplasmic reticulum chaperones, and thermotolerance.
The accumulation of misfolded proteins in the cytosol leads to increased expression of heat-shock proteins, while accumulation of such proteins in the endoplasmic reticulum (ER) stimulates the expression of many ER resident proteins, most of which function as molecular chaperones. Recently, inhibitors of the proteasome have been identified that can block the rapid degradation of abnormal cytosolic and ER-associated proteins. We therefore tested whether these agents, by causing the accumulation of abnormal proteins, might stimulate the expression of cytosolic heat-shock proteins and/or ER molecular chaperones and thereby induce thermotolerance. Exposure of Madin-Darby canine kidney cells to various proteasome inhibitors, including the peptide aldehydes (MG132, MG115, N-acetyl-leucyl-leucyl-norleucinal) and lactacystin, inhibited the degradation of short-lived proteins and increased markedly the levels of mRNAs encoding cytosolic heat-shock proteins (Hsp70, polyubiquitin) and ER chaperones (BiP, Grp94, ERp72), as shown by Northern blot analysis. However, inhibitors of cysteine proteases (E64), serine proteases (leupeptin), or metalloproteases (1, 10-phenanthroline) had no effect on the levels of these mRNAs. The relative efficacies of the peptide aldehyde inhibitors in inducing these mRNAs correlated with their potencies against the proteasome. Furthermore, reduction of the aldehyde group of MG132 decreased its inhibitory effect on proteolysis and largely prevented the induction of these mRNAs. Although treatment with the proteasome inhibitors caused rapid increases in mRNA levels (as early as 2 h after treatment with MG132), the inhibitors did not detectably affect total protein synthesis, total protein secretion, ER morphology, or the retention of ER-lumenal proteins, even after 18 h of treatment. Together, the findings suggest that inhibition of proteasome function induces heat-shock proteins and ER chaperones due to the accumulation of sufficient amounts of abnormal proteins and/or the inhibition of degradation of a key regulatory factor (e.g. heat-shock factor). Since expression of heat-shock proteins can protect cells from thermal injury, we tested whether the proteasome inhibitors might also confer thermotolerance. Treatment of cells with MG132 for as little as 2 h, markedly increased the survival of cells subjected to high temperatures (up to 46 degrees C). Thus, these agents may have applications in protecting against cell injury. Topics: Acetylcysteine; Animals; Calpain; Cell Line; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Dogs; Endoplasmic Reticulum; Fungal Proteins; Heat-Shock Proteins; HSP70 Heat-Shock Proteins; Leupeptins; Membrane Glycoproteins; Membrane Proteins; Molecular Chaperones; Multienzyme Complexes; Proteasome Endopeptidase Complex; Protein Conformation; RNA, Messenger | 1997 |
Evidence of proteasome-mediated cytochrome P-450 degradation.
The cytochrome P-450 family of enzymes performs an incredibly diverse range of detoxification and oxidation reactions within the cell and constitutes between 5 and 10% of protein in hepatic endoplasmic reticulum. In this report it is demonstrated that constitutively expressed membranous P-450s are targeted for destruction by the proteasome, in a process which is ubiquitin-independent and is demonstrated in vitro to require prior labilization of the enzyme. This process was specific for P-450s CYP1A2, CYP2E1, CYP3A, and CYP4A and was not demonstrated to be involved in the turnover of CYP1A1, CYP2B1/2, or NADPH reductase. In reconstitution experiments using purified proteasomes and microsomal fractions, labilized P-450 conformations are protected from 20 S proteasome degradation by substrate addition, with proteolysis occurring while P-450s are still attached to the endoplasmic reticulum. Topics: Acetylcysteine; Animals; Aryl Hydrocarbon Hydroxylases; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytochrome P-450 CYP1A2; Cytochrome P-450 CYP2E1; Cytochrome P-450 CYP3A; Cytochrome P-450 CYP4A; Cytochrome P-450 Enzyme System; Dose-Response Relationship, Drug; Leupeptins; Microsomes, Liver; Mixed Function Oxygenases; Multienzyme Complexes; Oxidoreductases, N-Demethylating; Proteasome Endopeptidase Complex; Rats; Rats, Sprague-Dawley | 1997 |
Mechanism for biphasic rel A. NF-kappaB1 nuclear translocation in tumor necrosis factor alpha-stimulated hepatocytes.
The proinflammatory cytokine, tumor necrosis factor alpha (TNFalpha), is a potent activator of angiotensinogen gene transcription in hepatocytes by activation of latent nuclear factor-kappaB (NF-kappaB) DNA binding activity. In this study, we examine the kinetics of TNFalpha-activated translocation of the 65-kDa (Rel A) and 50-kDa (NF-kappaB1) NF-kappaB subunits mediated by inhibitor (IkappaB) proteolysis in HepG2 hepatoblastoma cells. HepG2 cells express the IkappaB members IkappaBalpha, IkappaBbeta, and IkappaBgamma. In response to TNFalpha, Rel A.NF-kappaB1 translocation and DNA binding activity follows a biphasic profile, with an "early" induction (15-30 min), followed by a nadir to control levels at 60 min, and a "late" induction (>120 min). The early phase of Rel A.NF-kappaB1 translocation depends on simultaneous proteolysis of both IkappaBalpha and IkappaBbeta isoforms; IkappaBgamma is inert to TNFalpha treatment. The 60-min nadir is due to a rapid IkappaBalpha resynthesis that reassociates with Rel A and completely inhibits its DNA binding activity; the 60-min nadir is not observed when IkappaBalpha resynthesis is prevented by cycloheximide treatment. By contrast, selective inhibition of IkappaBbeta proteolysis by pretreatment of HepG2 cells with the peptide aldehyde N-acetyl-Leu-Leu-norleucinal completely blocks the late phase of Rel A.NF-kappaB1 translocation. These studies indicate the presence of inducible and constitutive cytoplasmic NF-kappaB pools in hepatocytes. TNFalpha induces a coordinated proteolysis and resynthesis of IkappaB isoforms to produce dynamic changes in NF-kappaB nuclear abundance. Topics: Cell Nucleus; Cysteine Proteinase Inhibitors; DNA; DNA-Binding Proteins; Humans; I-kappa B Proteins; Leupeptins; Liver; NF-kappa B; NF-kappa B p50 Subunit; NF-KappaB Inhibitor alpha; Protein Precursors; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-rel; Recombinant Proteins; Transcription Factor RelA; Transcription Factor RelB; Transcription Factors; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha | 1997 |
A two-site model for ApoB degradation in HepG2 cells.
Newly synthesized apolipoprotein B (apoB) undergoes rapid degradation in a pre-Golgi compartment in HepG2 cells. A major site of this early degradation seems to be on the cytosolic side of the endoplasmic reticulum (ER) membrane and is sensitive to N-acetyl-leucinyl-leucinyl-norleucinal (ALLN), which can inhibit neutral cysteine proteases and/or proteasome activity. Oleate (OA) treatment, which facilitates translocation of nascent apoB across the ER membrane, also reduces early degradation. In the present studies, we have used brefeldin A (BFA), which inhibits vesicular transport from the ER to the Golgi, to demonstrate that apoB can also be degraded by an ER luminal proteolytic activity that is distinct from the ALLN-sensitive proteases. Thus, when BFA-treated HepG2 cells were co-treated with ALLN, which protects apoB but does not facilitate its translocation into the ER lumen, degradation of newly synthesized apoB was significantly reduced compared with cells incubated with BFA alone. However, apoB degradation was rapid and complete when OA was added to media containing either BFA or ALLN/BFA. These results suggested that OA, by increasing translocation of nascent apoB into the ER lumen, exposed apoB to an ALLN-resistant proteolytic pathway. When we incubated HepG2 cells with dithiothreitol (DTT)/OA/BFA or DTT/OA/ALLN/BFA, degradation of apoB was inhibited. Furthermore, addition of DTT resulted in the accumulation of a 70-kDa amino-terminal fragment of apoB. Both full-length and amino-terminal apoB were degraded if DTT was removed from the incubation media; both were secreted if only BFA was removed. Thus, even after apoB is translocated into the ER lumen (thereby avoiding the initial proteolytic pathway), it can potentially be degraded by a lumenal proteolytic process that is ALLN-resistant but DTT-sensitive. The present results, together with previous studies, suggest that at least two distinct steps may be involved in the posttranslational degradation of apoB: 1) the first occurs while apoB is partially translocated and is ALLN-sensitive; and 2) the second occurs in the ER lumen and is DTT-sensitive. Finally, our results support the hypothesis that degradation of partially translocated apoB generates a 70-kDa amino-terminal fragment that is mainly degraded in the ER lumen by a DTT-sensitive pathway. Topics: Apolipoproteins B; Biological Transport; Brefeldin A; Cyclopentanes; Cysteine Proteinase Inhibitors; Dithiothreitol; Endoplasmic Reticulum; Humans; Leupeptins; Liver; Models, Biological; Oleic Acid; Peptide Fragments; Protein Synthesis Inhibitors | 1997 |
Effects of an inhibitor of cathepsin L on bone resorption in thyroparathyroidectomized and ovariectomized rats.
The process of bone resorption by osteoclasts involves the dissolution of mineral salts and enzymatic degradation of the mainly collagenous extracellular matrix. Cysteine proteinases, which can efficiently degrade collagen at acidic pH, have been suggested to play an important role in the bone resorptive process. The cysteine proteinase cathepsin L is secreted by osteoclasts, and inhibitors of this enzyme can prevent bone resorption in vitro. The activity of acetyl-leu-leu-norleucinol (ALLN), a selective inhibitor of cathepsin L, was investigated in two models of bone resorption in vivo. In the first study, the ability of ALLN to inhibit bone resorption was investigated in Ro-13-6298 (arotinoid)-treated thyroparathyroidectomized (TPTX) rats. ALLN [100 mg/kg, intraperitoneally (i.p.)] inhibited hypercalcemia by 62.8% acutely (p < 0.001), compared to 94.9% (p < 0.001) inhibition by salmon calcitonin (sCT) (10 IU/kg, subcutaneously). In rats treated for 3 days with ALLN, arotinoid-induced reduction in cortical bone mineral density measured by peripheral quantitative computed tomography (pQCT) was inhibited by 86.4% (p < 0.05) in rats treated with ALLN 100 mg/kg, i.p., and by 82% in rats treated with 50 mg/kg, i.p. (p < 0.05). In a second study, the efficacy of ALLN was tested in a longitudinal study in ovariectomized (ovx) rats. Bone loss, measured by pQCT, was unaffected by treatment with ALLN. The bisphosphonate alendronate, however, inhibited bone loss in this model. These data demonstrate the ability of a cathepsin L inhibitor to inhibit bone resorption in arotinoid-treated TPTX rats, a process which may be dependent on the activity of cathepsin L-like cysteine proteinases. In contrast to its effects in TPTX rats, ALLN had no inhibitory activity on bone resorption in ovx rats. It is possible that in chronic bone resorption in ovx rats, the activity of other enzymes such as cathepsins OC-2 or K allows the process of resorption to continue even when cathepsin L is inhibited by ALLN. Further studies are required to determine why the activity of ALLN varies between different animal models. These data indicate that there may be variations in the effects of drugs in different animal models of bone resorption which should be considered when investigating novel antiresorptive therapies. Topics: Animals; Benzoates; Bone Resorption; Cathepsin L; Cathepsins; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Disease Models, Animal; Endopeptidases; Female; Humans; Hypercalcemia; Leupeptins; Male; Osteoporosis; Ovariectomy; Ovary; Parathyroid Glands; Parathyroidectomy; Rats; Rats, Wistar; Retinoids; Thyroid Gland; Thyroidectomy | 1997 |
Insulin inhibition of proteasome activity in intact cells.
Cellular homeostasis requires regulation of protein turnover. Protein degradation is an essential component of this process and is inhibited by insulin. The importance of cytosolic proteolysis in overall cellular protein degradation is increasingly apparent and an insulin effect on this system has been suggested but not proven. The present study shows that a membrane permeable substrate of the proteasome is degraded in HepG2 cells and that insulin inhibits its degradation both by isolated proteasomes and by intact cells. Inhibitors of the proteasome suppress degradation, and in the presence of these inhibitors insulin has no further effect. This is the first demonstration that insulin inhibition of cellular protein degradation is due to an effect on proteasomes. Topics: Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Humans; Hydrolysis; Insulin; Leupeptins; Multienzyme Complexes; Oligopeptides; Proteasome Endopeptidase Complex; Tumor Cells, Cultured | 1997 |
Geldanamycin-stimulated destabilization of mutated p53 is mediated by the proteasome in vivo.
Mutation of the tumor suppressor gene p53 is the most common genetic abnormality detected in human cancers. Wild type p53 is a short-lived protein with very low basal intracellular levels. Most mutated forms of the protein, however, display markedly increased intracellular levels as an essential feature of their positive transforming activity. In this report, we have used selective inhibitors of the 20S proteasome to demonstrate that processing of p53 by ubiquitination and proteasome-mediated degradation is impaired by commonly occuring mutations of the protein. We found that this impairment of p53 turnover can be reversed by treatment of tumor cells with the benzoquinone ansamycin, geldanamycin, leading to a marked reduction in intracellular p53 levels. Finally, using cells which over-express a mutant p53 protein, we were able to demonstrate that restoration of proteasome-mediated degradation by geldanamycin is accompanied by p53 polyubiquitination. Although much remains to be learned about the mechanisms involved, our data demonstrate that selective de-stabilization of mutant transforming proteins such as p53 can be achieved pharmacologically with agents such as geldanamycin which modify the function of molecular chaperone proteins within tumor cells. Topics: Acetylcysteine; Animals; Benzoquinones; Cycloheximide; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Detergents; Enzyme Inhibitors; Half-Life; Humans; Lactams, Macrocyclic; Leupeptins; Mice; Multienzyme Complexes; Mutation; Octoxynol; Polyethylene Glycols; Proteasome Endopeptidase Complex; Protein Synthesis Inhibitors; Quinones; Rats; Tumor Cells, Cultured; Tumor Suppressor Protein p53; Ubiquitins | 1997 |
Inhibitors of the proteasome reduce the accelerated proteolysis in atrophying rat skeletal muscles.
Several observations have suggested that the enhanced proteolysis and atrophy of skeletal muscle in various pathological states is due primarily to activation of the ubiquitin-proteasome pathway. To test this idea, we investigated whether peptide aldehyde inhibitors of the proteasome, N-acetyl-leucyl-leucyl-norleucinal (LLN), or the more potent CBZ-leucyl-leucyl-leucinal (MG132) suppressed proteolysis in incubated rat skeletal muscles. These agents (e.g., MG132 at 10 microM) inhibited nonlysosomal protein breakdown by up to 50% (P < 0.01), and this effect was rapidly reversed upon removal of the inhibitor. The peptide aldehydes did not alter protein synthesis or amino acid pools, but improved overall protein balance in the muscle. Upon treatment with MG132, ubiquitin-conjugated proteins accumulated in the muscle. The inhibition of muscle proteolysis correlated with efficacy against the proteasome, although these agents could also inhibit calpain-dependent proteolysis induced with Ca2+. These inhibitors had much larger effects on proteolysis in atrophying muscles than in controls. In the denervated soleus undergoing atrophy, the increase in ATP-dependent proteolysis was reduced 70% by MG132 (P < 0.001). Similarly, the rise in muscle proteolysis induced by administering thyroid hormones was reduced 40-70% by the inhibitors. Finally, in rats made septic by cecal puncture, the increase in muscle proteolysis was completely blocked by MG132. Thus, the enhanced proteolysis in many catabolic states (including denervation, hyperthyroidism, and sepsis) is due to a proteasome-dependent pathway, and inhibition of proteasome function may be a useful approach to reduce muscle wasting. Topics: Animals; Atrophy; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Diaphragm; Kinetics; Leupeptins; Male; Multienzyme Complexes; Muscle, Skeletal; Proteasome Endopeptidase Complex; Proteins; Rats; Rats, Inbred Strains | 1997 |
Transcriptional squelching by ectopic expression of E2F-1 and p53 is alleviated by proteasome inhibitors MG-132 and lactacystin.
The transcription factors p53 and E2F-1 play important roles in the control of cell cycle progression. In transient transfection experiments, expression of E2F-1, other E2F family members, or p53 squelched transcription from cotransfected plasmids in a dose-dependent manner. Although the proteasome inhibitors MG-132 and lactacystin markedly increased the level of expression of E2F-1 and p53, these inhibitors completely alleviated squelching by both proteins. Several observations indicate MG-132 alleviates squelching by influencing the conformation of newly synthesized p53 and E2F-1:MG-132 increased the fraction of wild type p53 bound by a monoclonal antibody which preferentially recognizes mutant conformers of p53, increased binding of hsp70 to p53 and inhibited nuclear accumulation of both p53 and E2F-1, but not the pocket protein p107. The protease inhibitors ALLN and ALLM did not influence expression of E2F-1 or p53, nor did they alleviate squelching by either transcription factor. Because MG-132 and lactacycstin are highly specific inhibitors of the proteasome protease, our results suggest that the proteasome influences post-translational processes involved in proper folding and cytoplasmic clearing of E2F-1 and p53. Topics: Acetylcysteine; Animals; beta-Galactosidase; Blotting, Northern; Carrier Proteins; Cell Cycle Proteins; CHO Cells; Cricetinae; Cycloheximide; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; E2F Transcription Factors; E2F1 Transcription Factor; Leupeptins; Oligopeptides; Protein Synthesis Inhibitors; Retinoblastoma-Binding Protein 1; RNA, Messenger; Transcription Factors; Transcription, Genetic; Transcriptional Activation; Transfection; Tumor Suppressor Protein p53; Ubiquitins | 1997 |
The effect of the proteasome inhibitor lactacystin on the presentation of transporter associated with antigen processing (TAP)-dependent and TAP-independent peptide epitopes by class I molecules.
Cells were treated with two proteolytic inhibitors, N-acetyl-leucyl-leucyl-norleucinal and lactacystin, the latter reported to be a specific inhibitor for the proteasome. Both inhibitors retarded the maturation of endo-H-resistant forms of murine and human class I molecules from their endo-H-sensitive precursors in cell lines with functional TAP proteins. HLA-A2 maturation readily occurs in TAP-deficient T2 cells, and it has been shown that the peptides associated with A2 are derived from the leader segment of proteins in the secretory pathway. This maturation is inhibited by N-acetyl-leucyl-leucyl-norleucinal but not lactacystin, indicating that the proteasome is not required for the generation of HLA-A2 binding peptides in these cells. The murine class Ib molecule Qa-1b presents a leader peptide derived from D-end class I molecules to alloreactive CTL. Since this presentation is dependent on the expression of TAP proteins, we determined if this requirement reflects a need for the proteasome to process this peptide. We found that lactacystin did not inhibit the maturation of endo-H-resistant forms of Qa-1b that are dependent on this leader peptide for its maturation, nor did it inhibit the expression of this peptide-Qa-1b complex in a functional assay. Thus, unlike conventional cytosolic peptides, leader peptides (regardless of whether they are dependent on TAP for their presentation) do not require the proteasome for processing. Topics: Acetylcysteine; Animals; Antigen Presentation; ATP Binding Cassette Transporter, Subfamily B, Member 2; ATP-Binding Cassette Transporters; Biological Transport; Cysteine Endopeptidases; Endoplasmic Reticulum; Epitopes; Hexosaminidases; Histocompatibility Antigens Class I; HLA-A2 Antigen; Humans; Leupeptins; Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase; Mice; Multienzyme Complexes; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; Protein Sorting Signals; Self Tolerance; T-Lymphocytes, Cytotoxic | 1997 |
Cyclosporine A is an uncompetitive inhibitor of proteasome activity and prevents NF-kappaB activation.
Cyclosporine A is an immunosuppressive agent that is used clinically in the prevention of transplant rejection and development of graft-versus-host disease. Recently, cyclosporine A has been shown to possess anti-inflammatory properties and is capable of inhibiting lipopolysaccharide-induced NF-kappaB activation. Ubiquitin-mediated proteasomal proteolysis plays a critical role in signal-induced NF-kappaB activation since it regulates both IkappaB degradation and p105 processing, it is also involved in the production of peptides for the assembly of MHC class I molecules. We report here that cylcosporine A acts as an uncompetitive inhibitor of the chymotrypsin-like activity of the 20S proteasome in vitro and that it suppresses lipopolysaccharide-induced IkappaB degradation and p105 processing in vivo demonstrating that inhibition of proteasome proteolysis is the mechanism by which cyclosporine A prevents NF-kappaB activation. A structurally unrelated immunosuppressant, rapamycin, did not inhibit the 20S proteasome in vitro. Topics: Animals; Brain; Cattle; Cyclosporine; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Leupeptins; Lipopolysaccharides; Multienzyme Complexes; NF-kappa B; NF-kappa B p50 Subunit; Polyenes; Proteasome Endopeptidase Complex; Protein Precursors; Proto-Oncogene Proteins; Sirolimus; Transcription Factor RelB; Transcription Factors; Tumor Necrosis Factor-alpha | 1997 |
Identification and functional requirement of Cu(I) and its ligands within coagulation factor VIII.
Coagulation factor VIII (FVIII) is a heterodimer consisting of a light chain of 80 kDa (domains A3-C1-C2) in a metal ion-dependent association with a 220-kDa heavy chain (domains A1-A2-B). The nature of the metal ion-dependent association between the heavy and light chains was investigated using atomic absorption spectroscopy, electron paramagnetic resonance spectroscopy (EPR), and site-directed mutagenesis and expression of the FVIII cDNA. Whereas copper ion was not detected in intact recombinant FVIII, EDTA dissociation of the chains yielded an EPR signal consistent with 1 mol of Cu(I)/mol of active protein, supporting the hypothesis that a single molecule of reduced copper ion is buried within intact FVIII and is released and oxidized upon treatment with EDTA. Cu(I), and not Cu(II), was able to reconstitute FVIII activity from dissociated chains, demonstrating a requirement for Cu(I) in FVIII function. Three potential copper ion binding sites exist within FVIII: one type-2 site and two type-1 sites. The importance of these potential copper ion ligands was tested by studying the effect of site-directed mutants. Of the two histidines that compose the type-2 binding site, the His-1957 --> Ala mutant displayed secretion, light and heavy chain assembly, and activity similar to wild-type FVIII, while mutant His-99 --> Ala was partially defective for secretion and had low levels of heavy and light chain association and activity. In contrast, FVIII having the mutation Cys-310 --> Ser within the type-1 copper binding site in the A1 domain was inactive and partially defective for secretion from the cell, and the heavy and light chains of the secreted protein were not associated. Mutant Cys-2000 --> Ser within the A3 domain displayed secretion, assembly, and activity similar to that for wild-type FVIII. These results support the hypothesis that Cu(I) is buried within the type-1 copper binding site within the A1 domain and is required for FVIII chain association and activity. Topics: Amino Acid Substitution; Animals; Binding Sites; CHO Cells; Copper; COS Cells; Cricetinae; Culture Media, Conditioned; Cysteine Proteinase Inhibitors; Electron Spin Resonance Spectroscopy; Factor VIII; Humans; Kinetics; Leupeptins; Ligands; Macromolecular Substances; Mutagenesis, Site-Directed; Recombinant Proteins; Spectrophotometry, Atomic | 1997 |
Nitric oxide attenuates reoxygenation-induced ICAM-1 expression in coronary microvascular endothelium: role of NFkappaB.
Enhanced leukocyte adhesion has been shown to occur in post-ischemic reperfused hearts due to the upregulation of specific cell-surface adhesion molecules. Therefore, we investigated the influence of 4 h of reoxygenation after 20 h of hypoxia on ICAM-1 induction in primary cultures of rat coronary microvascular endothelial cells (CMEC). ICAM-1 surface expression as well as oxygen free radical formation were measured by flow cytometry. Changes in ICAM-1 mRNA levels were assessed by Northern blot and activation of NFkappaB and AP-1 signalling were analysed by electrophoretic mobility shift assays (EMSA) in CMEC lysates. Although hypoxia alone did not affect cell-surface ICAM-1 expression, 4 h of reoxygenation induced a significant upregulation of ICAM-1. ICAM-1 mRNA could not be found after hypoxia alone, but could be detected as early as 1 h following reoxygenation. Unlike AP-1, the activation of which could be detected in CMEC lysates following hypoxia alone, NFkappaB binding activity was induced only following reoxygenation, concurrent with an increase in the formation of reactive oxygen species (ROS). A proteasome inhibitor, nor-Leu (25 microM) inhibited NFkappaB activation by reoxygenation and ICAM-1 expression. Blockade of endogenous nitric oxide (NO) synthesis in CMEC with L-nitroarginine (10 microM) accentuated post-reoxygenation ICAM-1 expression. Finally, an exogenous NO donor, S-nitrosoacetyl-penicillamine (SNAP, 100 microM), suppressed the generation of ROS upon reoxygenation, and blocked the activation of NFkappaB and the upregulation of ICAM-1. Thus, ICAM-1 upregulation in CMEC primary cultures is not induced by hypoxia alone, but appears shortly after reoxygenation in the absence of exogenous cytokines or inflammatory cells. Because upregulation of AP-1 through hypoxia alone did not affect ICAM-1 expression, we conclude that redox-sensitive NFkappaB activation triggers ICAM-1 upregulation. NO inhibits reoxygenation-specific ICAM-1 upregulation, most likely by diminishing oxidative stress that leads to NFkappaB activation. Topics: Animals; Cells, Cultured; Coronary Vessels; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Endothelium, Vascular; Intercellular Adhesion Molecule-1; Leupeptins; Multienzyme Complexes; NF-kappa B; Nitric Oxide; Oxygen; Proteasome Endopeptidase Complex; Rats | 1997 |
Proteolysis of erythrocyte-type and brain-type ankyrins in rat heart after postischemic reperfusion.
Ankyrin links cytoskeleton and integral membrane proteins and is proteolyzed in vitro by calpain, a Ca2+-dependent protease. In the present study, we examined the localization of two ankyrin isoforms, erythrocyte (red blood cell)-type (ankyrin(R)) and brain-type (ankyrin(B)), and their proteolysis after ischemia-reperfusion in the subcellular fractions of perfused rat heart by immunoblotting and by immunohistochemistry using specific antibodies. Both isoforms were observed to be distributed chiefly in the myofibril-nucleus (1,OOOx g pellet: P1) fraction, while ankyrin(R) was located substantially in the membrane (100,000x g pellet: P2) fraction. Reperfusion after 10 min or more of global ischemia induced preferential proteolysis of ankyrin(R) in the P2 fraction and ankyrin(B) in the P1 fraction. The proteolysis of ankyrin(R), but not ankyrin(B), was effectively inhibited by the synthetic calpain inhibitor acethyl-leucyl-leucyl-norleucinal. The immunohistochemical examination showed that anti-ankyrin(R) delineated striations, sarcolemma and nuclei, and the staining was decreased after ischemia-reperfusion, while anti-ankyrin(B) showed diffuse staining. The proteolysis of ankyrin(R) may interfere with force conduction through disruption of the linkage between integral membrane proteins and the myofibril-cytoskeleton. Topics: Animals; Ankyrins; Brain; Calpain; Cell Fractionation; Cysteine Proteinase Inhibitors; Erythrocytes; In Vitro Techniques; Leupeptins; Male; Molecular Weight; Myocardial Ischemia; Myocardial Reperfusion Injury; Myocardium; Peptide Fragments; Rats; Rats, Wistar | 1997 |
Insulin stimulates haptotactic migration of human epidermal keratinocytes through activation of NF-kappa B transcription factor.
Insulin-mediated cell motility as well as the role of transcription factors in insulin-activated intracellular signal events have not been extensively studied. In this report we have examined whether insulin could mediate haptotactic migration of cultured human epidermal keratinocytes through activation of transcription factor NF-kappa B. Insulin caused a dose-dependent stimulation of keratinocyte migration that maximally reached 2-fold at 2 x 10(-7) M hormone. This phenomenon was independent of the nature of the extracellular matrix component (collagen I or laminin5/nicein) on which the cells migrated, indicating that a specific integrin-ligand complex is not required. A 10(-7) M insulin treatment of keratinocytes resulted in activation of a major kappa B DNA binding complex within 15 to 30 minutes, which was identified as the p65/p50 NF-kappa B heterodimer by electrophoretic mobility shift assays. The activation induced nuclear translocation of cytosolic pools of NF-kappa B factor. Pyrrolidine dithiocarbamate and N-acetyl-leucinyl-leucinyl-norleucinal H (two compounds that differentially inhibit I kappa B alpha degradation and, thus, NF-kappa B activation) reversed the insulin-stimulated keratinocyte haptotactic migration without affecting insulin receptor activation. These compounds inhibited the insulin-induced nuclear translocation of NF-kappa B as detected by confocal laser scanning microscopy. Taken together our experiments demonstrate that insulin stimulates haptotactic migration of human epidermal keratinocytes through activation of NF-kappa B transcription factor. They emphasize the ability of insulin to stimulate keratinocyte movement and provide a first clue to the mechanism of insulin-induced haptotactic signaling. Topics: Antioxidants; Cell Movement; Cells, Cultured; Cysteine Proteinase Inhibitors; Epidermal Cells; Extracellular Matrix Proteins; Humans; Hypoglycemic Agents; Insulin; Keratinocytes; Leupeptins; NF-kappa B; Pyrrolidines; Thiocarbamates | 1997 |
Rapid deubiquitination of nucleosomal histones in human tumor cells caused by proteasome inhibitors and stress response inducers: effects on replication, transcription, translation, and the cellular stress response.
The proteasome inhibitors, lactacystin and N-acetyl-leucyl-leucyl-norlucinal, caused a rapid and near-complete loss of approximately 22-23-kDa ubiquitinated nucleoproteins, which we have identified as monoubiquitinated nucleosomal histones H2A and H2B by immunological and two-dimensional electrophoretic techniques. In human SKBr3 breast tumor cells, depletion of monoubiquitinated histones by the proteasome inhibitors coincided with the accumulation of high molecular weight ubiquitinated proteins in both nucleoprotein and cytosolic fractions and decreased unconjugated ubiquitin in the cytosol, without changes in the nonubiquitinated core histones. Unconjugated ubiquitin was not detected in isolated tumor cell nuclei. A similar loss in monoubiquitinated histones occurred in cells harboring a defective, temperature-sensitive mutation of the ubiquitin-activating E1 enzyme, after these cells were elevated from 33 degrees C to the non-permissive temperature of 39 degrees C. DNA replication and RNA transcription were decreased by the proteasome inhibitors most strongly after 90% of the ubiquitin had been removed from ubiquitinated histones H2A and H2B, suggesting a relationship between the nucleosomal histone ubiquitin status and the processing of genetic information. Interestingly, although both proteasome inhibitors caused a generalized decrease in methionine incorporation into proteins, they strongly induced the synthesis of the hsp72 and hsp90 stress proteins. Finally, treating cells with heat-shock at 43 degrees C, with stress response-provoking chemicals or with several other proteasome inhibitors caused ubiquitinated proteins to accumulate, depleted free ubiquitin, and concomitantly decreased nucleosomal monoubiquitinated histones. These results suggest that deubiquitination of nucleosomal histones H2A and H2B may play a previously unrecognized role in the cellular stress response, as well as in the processing of chromatin, and emphasize the important role of the proteasome in cellular homeostasis. Topics: Acetylcysteine; Breast Neoplasms; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytosol; DNA Replication; Female; Histones; Hot Temperature; Humans; Leupeptins; Multienzyme Complexes; Nucleoproteins; Nucleosomes; Proteasome Endopeptidase Complex; Protein Biosynthesis; Transcription, Genetic; Tumor Cells, Cultured; Ubiquitins | 1997 |
Phosphorylation of human CDC25B phosphatase by CDK1-cyclin A triggers its proteasome-dependent degradation.
In eukaryotes the activity of CDK1 (CDC2), a cyclin-dependent kinase that initiates the structural changes that culminate in the segregation of chromosomes at mitosis, is regulated by the synergistic and opposing activities of a cascade of kinases and phosphatases. Dephosphorylation of threonine 14 and tyrosine 15 of CDK1 by the CDC25 phosphatases is a key step in the activation of the CDK1-cyclin B protein kinase. Little is currently known about the role and the regulation of CDC25B. Here we report in vitro and in vivo data that indicate that CDC25B is degraded by the proteasome. This degradation is dependent upon phosphorylation by the CDK1-cyclin A complex but not by CDK1-cyclin B. These results indicate that CDK1-cyclin A phosphorylation targets CDC25B for degradation and that this might be an important component of cell cycle regulation at the G2/M transition. Topics: Calpain; CDC2 Protein Kinase; cdc25 Phosphatases; Cell Cycle; Cell Cycle Proteins; Cyclin A; Cysteine Endopeptidases; HeLa Cells; Humans; Leupeptins; Multienzyme Complexes; Phosphoprotein Phosphatases; Phosphorylation; Proteasome Endopeptidase Complex; Threonine; Tyrosine | 1997 |
Conformational changes in apolipoprotein B modulate intracellular assembly and degradation of ApoB-containing lipoprotein particles in HepG2 cells.
The linkage between the conformation of apolipoprotein B100 (apoB) and the intracellular assembly and degradation of apoB-containing lipoproteins was investigated in the present study. Disruption of disulfide bond formation in newly synthesized apoB molecules through the use of the reducing agent DTT resulted in a decrease in the secretion of apoB-containing lipoproteins from HepG2 cells compared with control cells. The synthesis of total apoB (apoB100 plus nascent chains), as well as a number of control proteins, such as albumin and alpha 1-antitrypsin, was decreased significantly in DTT-treated cells. However, the intracellular accumulation of full-length apoB100 molecules was not inhibited in the presence of DTT. Subcellular fractionation indicated that apoB molecules isolated from the microsomes of DTT-treated cells had an increased association with the microsomal membrane compared with apoB isolated from untreated cells. Analysis of the distribution of apoB-containing lipoproteins from the lumen of isolated microsomes demonstrated that in the presence of DTT, there was a shift in the distribution, such that there was a decrease in the formation of HDL-sized (lipid-poor) apoB-containing lipoproteins and a decrease in the formation of LDL/VLDL apoB particles. Alterations in apoB conformation and their impact on degradation were also investigated by using DTT and by inhibiting N-linked glycosylation with tunicamycin. DTT appeared to change the rate and pattern of apoB degradation. Degradation was accelerated in both intact and permeabilized HepG2 cells. ApoB degradation occurred in DTT-treated permeabilized cells without the usual generation of the 70-kD and 335-kD fragments and was largely N-acetyl-leucyl-leucyl-norleucinal (ALLN) insensitive. In tunicamycin-treated cells, DTT further accelerated the degradation of unglycosylated apoB. Overall, the data suggest that the misfolding of apoB may prevent the proper association of apoB with lipids, resulting in impairment of the assembly of mature apoB-containing lipoproteins. Alteration in the conformation of apoB also appears to alter the degradation pathway of apoB, such that the protein is degraded through a pathway that is at least in part ALLN insensitive. Topics: Animals; Apolipoprotein B-100; Apolipoproteins B; Carcinoma, Hepatocellular; Cystine; Dithiothreitol; Glycosylation; Humans; Leupeptins; Lipoproteins; Lipoproteins, HDL; Lipoproteins, LDL; Lipoproteins, VLDL; Liver; Liver Neoplasms; Microsomes, Liver; Molecular Weight; Neoplasm Proteins; Protease Inhibitors; Protein Conformation; Protein Denaturation; Protein Folding; Protein Processing, Post-Translational; Sulfhydryl Reagents; Tumor Cells, Cultured; Tunicamycin | 1997 |
Blocking intracellular degradation of the erythropoietin and asialoglycoprotein receptors by calpain inhibitors does not result in the same increase in the levels of their membrane and secreted forms.
The erythropoietin receptor (EPO-R), a type 1 membrane glycoprotein, is degraded mainly in the lysosomes or endosomes, whereas the asialoglycoprotein receptor (ASGP-R) H2a subunit, a type 2 membrane glycoprotein, is degraded exclusively in the endoplasmic reticulum. The present study describes compounds that inhibit the intracellular degradation of these receptors in an efficient manner. However, the levels of cell-surface expression and secretion of their soluble exoplasmic domains were not enhanced to the same extent. The calpain inhibitors N-acetyl-leucyl-leucyl-norleucinal (ALLN) and N-acetyl-leucyl-leucyl-methional (ALLM) inhibited EPO-R degradation profoundly. After 3 h of chase using Ba/F3 cells and NIH 3T3 fibroblasts expressing the EPO-R, virtually all of the receptor molecules were degraded, whereas 80% of the pulse-labelled receptor remained intact in the presence of the inhibitor. EPO-R cell-surface expression was elevated 1.5-fold after 1 h of incubation with ALLN. In the absence of protein synthesis, ALLN caused the accumulation of non-degraded EPO-R molecules in endosomes and lysosomes, as determined by double immunofluorescence labelling of NIH 3T3 cells expressing EPO-Rs. In Ba/F3 cells expressing a soluble EPO-R, ALLN treatment increased secretion of the soluble exoplasmic domain of the EPO-R 2-5-fold. Similarly, in NIH 3T3 cells singly transfected with the ASGP-R H2a subunit cDNA, ALLN inhibited degradation of the ASGP-R H2a subunit precursor, as well as the degradation of the 35 kDa proteolytic fragment corresponding to the receptor ectodomain, by 3-6-fold. However, accumulation of secreted proteolytic fragment in the medium was augmented in the presence of ALLN by only 1.75-fold. In cells expressing the G78R mutant of the ASGP-R H2a subunit, which is not cleaved to the 35 kDa fragment [Yuk and Lodish (1993) J. Cell Biol. 123, 1735-1749], degradation of the precursor was inhibited. Overall, our data suggest the involvement of cysteine proteinases located in the endoplasmic reticulum, as well as in post-Golgi compartments, in degradation of the EPO-R and the ASGP-R H2a subunit. The much lower effect of the inhibitory compounds on cell-surface and secreted forms of the EPO-R and ASGP-R H2a subunit illustrates the complexity and the tight regulation of the cellular localization and stability of membrane proteins. Topics: 3T3 Cells; Animals; Asialoglycoprotein Receptor; Asialoglycoproteins; Calpain; Cell Line; Cell Membrane; Cysteine Proteinase Inhibitors; Hydrolysis; Leupeptins; Mice; Receptors, Cell Surface; Receptors, Erythropoietin | 1996 |
Proteasome inhibitors block VCAM-1 and ICAM-1 gene expression in endothelial cells without affecting nuclear translocation of nuclear factor-kappa B.
Endothelial cells play a major role in recruiting leukocytes to sites of inflammation. This is accomplished, at least in part, by up-regulation of cell surface adhesion molecules, including VCAM-1 and ICAM-1, in response to cytokines. In this report, we investigated the role of the proteasome complex in mediating the interleukin (IL)- 1 beta induction of VCAM-1 and ICAM-1 gene expression in human endothelial cells. We present evidence that a proteasome inhibitor, n-acetyl-leucinyl-leucinyl-norleucinal (norLEU), as well as specific protease inhibitors, n-tosyl-Lys-chloromethylketone and N-tosyl-Phe-chloromethylketone, blocked IL-1 beta induction of VCAM-1 and ICAM-1 promoter-driven reporter gene expression in stably transfected endothelial cells. These inhibitors also blocked cytokine induced cell surface expression of VCAM-1 and ICAM-1 by human umbilical vein endothelial cells. As expected, the protease inhibitors blocked the activation of nuclear factor (NF)-kappa B in response to IL-1 beta stimulation. In contrast, norLEU did not prevent IL-1 beta-induced nuclear translocation of NF-kappa B. The effects of norLEU were specific because it did not inhibit the IL-1 beta induction of plasminogen activator inhibitor type 1 gene expression. This study demonstrates that inhibition of the proteolytic activity of the proteasome blocks IL-1 beta induction of VCAM-1 and ICAM-1 gene expression in human endothelial cells. Topics: Base Sequence; Biological Transport; Calpain; Cell Nucleus; Cells, Cultured; Cysteine Endopeptidases; Endothelium, Vascular; Gene Expression Regulation; Humans; Intercellular Adhesion Molecule-1; Interleukin-1; Leupeptins; Molecular Sequence Data; Multienzyme Complexes; NF-kappa B; Plasminogen Activator Inhibitor 1; Protease Inhibitors; Proteasome Endopeptidase Complex; Recombinant Fusion Proteins; RNA, Messenger; Tosyllysine Chloromethyl Ketone; Tosylphenylalanyl Chloromethyl Ketone; Transfection; Vascular Cell Adhesion Molecule-1 | 1996 |
Selective degradation of accumulated secretory proteins in the endoplasmic reticulum. A possible clearance pathway for abnormal tropoelastin.
The specific pathway of tropoelastin secretion was investigated in fetal calf ligamentum nuchae (FCL) cells using brefeldin A (BFA) to disrupt the secretory pathway. Electron microscopic studies of BFA-treated FCL cells showed ultrastructural changes consistent with the reported effects of BFA on intracellular organelles. When FCL cells were labeled with [3H]leucine in the presence of BFA, radiolabeled tropoelastin was not secreted, nor was there an intracellular accumulation of the protein. In contrast, fibronectin accumulated within the cells in the presence of BFA. Northern analysis of mRNA levels in FCL cells showed that the message for tropoelastin was unaffected by BFA treatment. Pulse chase experiments conducted in the presence of BFA demonstrated that the tropoelastin retained within the cells was rapidly degraded. Ammonium chloride, nocodazole, and cycloheximide had no effect on the degradation of tropoelastin, indicating that the degradation did not involve the endosome/lysosome pathway, movement via microtubules, or a short-lived protein, respectively. Incubation of FCL cells with BFA in the presence of N-acetyl-Leu-Leu-norleucinal, however, allowed tropoelastin to steadily accumulate in the cells. Cells pulsed in the presence of BFA alone showed that tropoelastin initially accumulates within the cells for approximately 1 h prior to being degraded, thus indicating that a critical threshold of tropoelastin must be reached before degradation can occur. Results from this study provide evidence for selective degradation of a soluble secreted protein by a cysteine protease following retention of the protein in the endoplasmic reticulum. Topics: Ammonium Chloride; Animals; Blotting, Northern; Brefeldin A; Cattle; Cells, Cultured; Cycloheximide; Cyclopentanes; Endoplasmic Reticulum; Leucine; Leupeptins; Ligaments; Microscopy, Electron; Nocodazole; Organelles; Protein Synthesis Inhibitors; RNA, Messenger; Serine Proteinase Inhibitors; Transcription, Genetic; Tropoelastin | 1996 |
The protease inhibitor, N-acetyl-L-leucyl-L-leucyl-leucyl-L-norleucinal, decreases the pool of major histocompatibility complex class I-binding peptides and inhibits peptide trimming in the endoplasmic reticulum.
N-acetyl-L-leucyl-L-leucyl-L-norleucinal, (LLnL), which inhibits proteasomes in addition to other proteases, was found to prolong the association of major histocompatibility complex class I molecules with the transporters associated with antigen processing (TAP), and to slow their transport out of the endoplasmic reticulum (ER). LLnL induced a reversible accumulation of ubiquitinated proteins and changed the spectrum of peptides bound by class I molecules. These effects can probably be attributed to proteasome inhibition. Unexpectedly, in the TAP-deficient cell line .174, the rate of intracellular transport of human histocompatibility leukocyte antigen (HLA) A2 was also reduced by LLnL, and the generation of most HLA-A2-associated signal sequence peptides was inhibited. The inhibition of HLA-A2 transport in .174 cells was found to be less sensitive to LLnL than in wild-type cells, and a similar difference was found for a second protease inhibitor, benzyloxycarbonyl-L-leucyl-L-leucyl-L-phenylalanilal. These data suggest that under some conditions such inhibitors can block trimming of peptides by an ER peptidase in addition to inhibiting cytosolic peptide generation. Topics: Amino Acid Sequence; Biological Transport; Carrier Proteins; Cell Compartmentation; Cysteine Proteinase Inhibitors; Cytosol; Endoplasmic Reticulum; Histocompatibility Antigens Class I; Leupeptins; Molecular Sequence Data; Oligopeptides; Peptides; Protein Binding | 1996 |
Proteasomes play an essential role in thymocyte apoptosis.
Cell death in many different organisms requires the activation of proteolytic cascades involving cytosolic proteases. Here we describe a novel requirement in thymocyte cell death for the 20S proteasome, a highly conserved multicatalytic protease found in all eukaryotes. Specific inhibitors of proteasome function blocked cell death induced by ionizing radiation, glucocorticoids or phorbol ester. In addition to inhibiting apoptosis, these signals prevented the cleavage of poly(ADP-ribose) polymerase that accompanies many cell deaths. Since overall rates of protein degradation were not altered significantly during cell death in thymocytes, these results suggest that the proteasome may either degrade regulatory protein(s) that normally inhibit the apoptotic pathway or may proteolytically activate protein(s) than promote cell death. Topics: Acetylcysteine; Animals; Apoptosis; Blotting, Western; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Dexamethasone; DNA Ligases; Leupeptins; Mice; Mice, Inbred BALB C; Multienzyme Complexes; Proteasome Endopeptidase Complex; Proteins; T-Lymphocytes | 1996 |
Involvement of the proteasome in the programmed cell death of NGF-deprived sympathetic neurons.
Sympathetic neurons undergo programmed cell death (PCD) upon deprivation of nerve growth factor (NGF). PCD of neurons is blocked by inhibitors of the interleukin-1beta converting enzyme (ICE)/Ced-3-like cysteine protease, indicating involvement of this class of proteases in the cell death programme. Here we demonstrate that the proteolytic activities of the proteasome are also essential in PCD of neurons. Nanomolar concentrations of several proteasome inhibitors, including the highly selective inhibitor lactacystin, not only prolonged survival of NGF-deprived neurons but also prevented processing of poly(ADP-ribose) polymerase which is known to be cleaved by an ICE/Ced-3 family member during PCD. These results demonstrate that the proteasome is a key regulator of neuronal PCD and that, within this process, it is involved upstream of proteases of the ICE/Ced-3 family. This order of events was confirmed in macrophages where lactacystin inhibited the proteolytic activation of precursor ICE and the subsequent generation of active interleukin-1beta. Topics: Acetylcysteine; Animals; Apoptosis; Calcium-Binding Proteins; Caspase 1; Cell Survival; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA Ligases; Electrophoresis, Polyacrylamide Gel; Leupeptins; Multienzyme Complexes; Nerve Growth Factors; Neurons; Poly(ADP-ribose) Polymerases; Proteasome Endopeptidase Complex; Rats; Sympathetic Nervous System | 1996 |
Calpain inhibitors protect against depolarization-induced neurofilament protein loss of septo-hippocampal neurons in culture.
We examined the effect of a 6 min depolarization with 60 mM KCl and 1.8, 2.8 or 5.8 mM extracellular CaCl2 on neurofilament proteins of high (NF-H), medium (NF-M) and low (NF-L) molecular weight in primary septohippocampal cultures. One day after depolarization, Western blot analyses revealed losses of all three neurofilament proteins. Increasing the extracellular calcium concentration from 1.8 to 5.8 mM CaCl2 in the presence of 60 mM KCl produced increased losses of all three neurofilament proteins to approximately 80% of control values in the absence of cell death. Calcium-dependent losses of the neurofilament proteins correlated with calcium-dependent increases in calpain 1-mediated breakdown products of alpha-spectrin. Calpain inhibitors 1 and 2, applied immediately after depolarization and made available to cultures for 24 h, reduced losses of all three neurofilament proteins to approximately 14% of control values. The protective effects of calpain inhibitors 1 and 2 were influenced by different levels of extracellular calcium. Qualitative immunohistochemical evaluations confirmed semiquantitative Western blot data on neurofilament loss and protection by calpain inhibitors 1 and 2. We propose that brief depolarization causes loss of neurofilament proteins, possibly due to calpain activation. Thus, calpain inhibitors could represent a viable strategy for preserving the cytoskeletal structure of injured neurons. Topics: Animals; Calcium Chloride; Calpain; Cells, Cultured; Cysteine Proteinase Inhibitors; Hippocampus; Leupeptins; Membrane Potentials; Nerve Tissue Proteins; Neurofilament Proteins; Neurons; Potassium Chloride; Rats; Septum Pellucidum; Spectrin | 1996 |
Cyclodextrins as templates for the presentation of protease inhibitors.
Mono(6-succinylamido-6-deoxy)-beta-cyclodextrin was synthesized by classical carbohydrate chemistry and used as a template mono-functionalized with the linear, fully flexible 4C-spacer carboxylate for covalent linkage of the calpain inhibitor leucyl-leucyl-norleucinal. Spectroscopic analyses of the conjugate do not support a self-inclusion of part of the hydrophobic peptide tail, but confirm its intra- or intermolecular interaction with the template moiety that leads to full water solubility. The inhibitory potency of the beta-cyclodextrin/peptide aldehyde construct was compared with that of the parent Ac-Leu-Leu-Nle-H against cathepsin B and calpain. Despite the large size of the template the inhibition of cathepsin B was only slightly reduced in full agreement with the X-ray structure of this enzyme which shows full accessibility of the S-subsites. For this enzyme the 4C-spacer is apparently sufficient to guarantee optimal interaction of the peptide tail with the binding cleft. Conversely, for mu-calpain a significantly decreased inhibitory potency was obtained with the conjugate suggesting steric interference of the template in the binding process. These results show that the beneficial properties of the cyclodextrin template can be retained in conjugates with bioactive peptides if attention is paid to optimize in each case the size and nature of the spacer for optimal recognition of the grafted biomolecule. Topics: beta-Cyclodextrins; Calpain; Carbohydrate Sequence; Circular Dichroism; Cyclodextrins; Cysteine Proteinase Inhibitors; Kinetics; Leupeptins; Molecular Sequence Data | 1996 |
Evidence that a rapidly turning over protein, normally degraded by proteasomes, regulates hsp72 gene transcription in HepG2 cells.
Heat shock protein 72/73 (Hsp70) is a cytosolic molecular chaperone that carries out fundamental roles under both normal and stress situations. There is great interest in delineating the mechanisms whereby Hsp70 levels are regulated. We observed that N-acetyl-leucyl-leucyl-norleucinal (ALLN), a synthetic aldehydic tripeptide that inhibits proteasomes, markedly induced Hsp70 levels (up to 30-fold above base line in HepG2 cells and human endothelial cells). Induction of Hsp70 by ALLN was dose-dependent and not related to cell toxicity. ALLN selectively increased Hsp70 levels without affecting Hsp25, Hsp27, Hsp60, Hsp86, Hsp90, Hsp104, or Bip (immunoglobulin heavy chain binding protein) in HepG2 cells. ALLN induced Hsp70 not only by stabilizing the protein but also by dramatically increasing its synthesis. The modulation of Hsp70 synthesis by ALLN resulted from a rapid and marked increase in transcription of the hsp72 gene, since the induction of hsp72 mRNA was blocked in cells co-treated with actinomycin D. hsp72 mRNA levels were affected in a time-dependent manner by exposure to ALLN; significant elevations occurred within 60 min of treatment, and a decline to background levels was observed by 7 h of recovery. The ALLN-induced increase in hsp72 gene expression was associated with trimerization of the heat shock transcriptional factor (HSF1). ALLN did not affect the steady-state level of HSF1 protein. The effects of ALLN appeared to require de novo protein synthesis, since the induction of both HSF1 trimerization and hsp72 transcription was blocked by co-treatment with cycloheximide. When we tested a series of protease inhibitors, only the related aldehydic tripeptides, N-acetyl-leucyl-leucyl-methioninal and the proteasome inhibitor, Cbz-leucyl-leucyl-leucinal, induced Hsp70 levels. The specific proteasome inhibitor, lactacystin, which has a different structure, also induced Hsp70 levels. Overall, our results suggest that a rapidly turning over protein that is normally degraded by proteasomes may be involved in the regulation of Hsp70 synthesis via effects on the hsp70 transcriptional factor, HSF1. Topics: Biopolymers; Cell Line; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Heat Shock Transcription Factors; Heat-Shock Proteins; HSP72 Heat-Shock Proteins; Humans; Hydrolysis; Leupeptins; Transcription Factors; Transcription, Genetic; Up-Regulation | 1996 |
Calpain inhibitors and serine protease inhibitors can produce apoptosis in HL-60 cells.
Recent investigations indicate that proteolysis is an important event in generation of the apoptosis phenotype. Although various proteases have been suggested to be candidates for this proteolysis, the results from different laboratories are inconsistent. In the present studies, HL-60 cells were treated with cycloheximide to investigate proteases involved in apoptosis. The calpain inhibitors benzyloxycarbonyl-Leu-Leu-Tyr diazomethylketone and acetyl-Leu-Leu-Nle aldehyde were not capable of preventing apoptosis induced by cycloheximide. In the absence of cycloheximide, these two inhibitors could initiate apoptosis in HL-60 cells. The thiol protease inhibitor benzyloxycarbonyl-Leu-Val-Gly diazomethylketone neither prevented nor produced apoptosis. The serine protease inhibitors 3,4-dichloroisocoumarin (DCI) and tosyl-Phe chloromethylketone (TPCK) also induced apoptosis in the absence of cycloheximide. On the other hand, the latter two inhibitors decreased cycloheximide-induced apoptosis, assessed either by cell morphologic changes or DNA ladder generation. Benzyloxycarbonyl-Val-Ala-Asp fluoromethyl ketone and iodoacetamide, inactivators of interleukin 1beta-converting enzyme (ICE)-like proteases, did not produce apoptosis and inhibited the induction of apoptosis by cycloheximide, calpain inhibitors, or serine protease inhibitors. These results are consistent with the ICE-like proteases having a central role in proteolysis during apoptosis, while calpain-like proteases and the serine proteases sensitive to DCI or TPCK are not required for generation of the apoptosis phenotype in HL-60 cells. Topics: Apoptosis; Calpain; Coumarins; Cycloheximide; Cysteine Proteinase Inhibitors; Diazomethane; HL-60 Cells; Humans; Isocoumarins; Leupeptins; Oligopeptides; Protein Synthesis Inhibitors; Serine Proteinase Inhibitors | 1996 |
p53-dependent cell cycle arrest induced by N-acetyl-L-leucinyl-L-leucinyl-L-norleucinal in platelet-derived growth factor-stimulated human fibroblasts.
Proteases are known to play important roles in cell growth control, although the underlying mechanisms are still poorly understood. Here we show that the protease inhibitor N-acetyl-L-leucinyl-L-leucinyl-L-norleucinal induced cell cycle arrest in platelet-derived growth factor-stimulated human fibroblasts at the G1/S boundary of the cell cycle by inhibiting the proteasome. Inhibition of the proteasome resulted in accumulation of the tumor suppressor p53, which was followed by an increase in the amount of the cyclin-dependent kinase-inhibitor p21. As a consequence, both phosphorylation and activity of the cyclin-dependent kinase 2/cyclin E complex were inhibited. We further observed that the retinoblastoma gene product, pRb, remained in the hypophosphorylated state, thus preventing cells from progression into the S-phase. These studies strongly support the hypothesis that the proteasome is a key regulator in the G1-phase of cell cycle progression. Topics: Calpain; Cathepsins; Cell Cycle; Cells, Cultured; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Fibroblasts; Humans; Leupeptins; Lung; Multienzyme Complexes; Platelet-Derived Growth Factor; Proteasome Endopeptidase Complex; Tumor Suppressor Protein p53 | 1996 |
Microsomal triacylglycerol transfer protein prevents presecretory degradation of apolipoprotein B-100. A dithiothreitol-sensitive protease is involved.
The role of microsomal triacylglycerol transfer protein (MTP) in the secretion of apolipoprotein B-100 (apoB-100) has been studied using an inhibitor of MTP: 4'-bromo-3'-methylmetaqualone. In vitro, this compound inhibits trioleoylglycerol transfer between lipid vesicles mediated by MTP with an IC50 of 0.9 microM whereas it does not inhibit the lipid transfer mediated by the cholesteryl ester transfer protein. In HepG2 cells, 4'-bromo-3'-methylmetaqualone inhibits the secretion of apoB-100 with an IC50 of 0.3 microM, without affecting the secretion of several other proteins like apoA-I or albumin. Moreover, there is no accumulation of apoB-100 in treated cells. Oleic acid, which increases apoB-100 secretion, only slightly modifies the IC50 of 4'-bromo-3'-methylmetaqualone (0.5 microM). The latter has no effect on the synthesis of major lipids within the cell, but decreases the secretion of triacylglycerol into apoB-100-containing lipoproteins. Pulse/chase experiments reveal that 4'-bromo-3'-methylmetaqualone acts on apoB-100 production either at the co-translational or post-translational level. The cysteine protease inhibitor N-acetyl-leucyl-leucyl-norleucinal does not protect apoB-100 from the 4'-bromo-3'-methylmetaqualone effect but seems to be involved in a later step of apoB-100 intracellular degradation. By contrast, dithiothreitol can totally reverse the effect of the MTP inhibitor on apoB-100 production. The mechanism of MTP-mediated lipid assembly with apoB-100 is discussed. Topics: Apolipoprotein B-100; Apolipoproteins B; Carrier Proteins; Cell Line; Cholesterol Ester Transfer Proteins; Cysteine Proteinase Inhibitors; Dithiothreitol; Endopeptidases; Glycoproteins; Humans; In Vitro Techniques; Leupeptins; Lipid Metabolism; Lipids; Methaqualone; Microsomes; Triglycerides | 1996 |
Ubiquitin-proteasome pathway mediates intracellular degradation of apolipoprotein B.
Newly synthesized apolipoprotein B (apoB) is degraded by a proteolytic process in the pre-Golgi compartment that can be inhibited by N-acetyl-L-leucinyl-L-leucinyl-L-norleucinal (ALLN) but not by several other protease inhibitors. We have tested the hypothesis that the ubiquitin-proteasome pathway is involved in the intracellular degradation of apoB in liver cells. We found that inhibitors of proteasomes blocked the degradation of apoB in cultured human hepatoma (HepG2) cells. Protein degradation by proteasomes is ATP-dependent, and ATP depletion by dinitrophenol and 2-deoxyglucose also inhibited apoB degradation in these cells. Furthermore, the intracellular human apoB isolated by immunoprecipitation was shown to react specifically with anti-ubiquitin antibody by immunoblotting. This result was corroborated by sequential immunoprecipitation of [35S]methionine-labeled proteins by anti-human apoB and anti-ubiquitin antisera. In contrast, secreted apoB was not ubiquitinated. The amount of intracellular ubiquitinated apoB was increased by the proteasome inhibitors, ALLN and carbobenzoxyl-leucinyl-leucinyl-norvalinal-H (MG115). Our findings suggest that the ubiquitin-proteasome pathway is one mechanism for the intracellular degradation of apoB. Topics: Adenosine Triphosphate; Antibodies; Apolipoproteins B; Blotting, Western; Cysteine Endopeptidases; Electrophoresis, Polyacrylamide Gel; Endoplasmic Reticulum; Leupeptins; Liver; Models, Biological; Multienzyme Complexes; Precipitin Tests; Protease Inhibitors; Proteasome Endopeptidase Complex; Tumor Cells, Cultured; Ubiquitins | 1996 |
Apolipoprotein B-100 destined for lipoprotein assembly and intracellular degradation undergoes efficient translocation across the endoplasmic reticulum membrane.
It has been proposed that inefficient translocation across the endoplasmic reticulum (ER) membrane gives rise to transmembrane forms of apolipoprotein B-100 (apoB). However, we previously demonstrated that the amino-terminal 50% of apoB (apoB-50) was efficiently translocated across the ER membrane in the nonhepatic cell line COS-1. To determine whether liver-specific factors modulate apoB membrane translocation or topology, hybrid proteins containing 300 amino acid overlapping segments of apoB-48 were transiently expressed in HepG2 cells and their protease sensitivities were examined in membrane vesicles. The hybrid proteins demonstrated the same range of protection from exogenously added protease (75-100%) as a transfected secretory control protein. When endogenous apoB was examined, its protection from trypsin in intact membranes was -80%, a value similar to that of two endogenous secretory control proteins, transferrin and alpha 2-macroglobulin. No discretely sized fragments of apoB were generated by trypsin digestion of membranes unless they were first permeabilized with detergent. In contrast to the behavior of apoB and other control proteins, albumin predominantly resisted degradation by trypsin in both intact and detergent permeabilized membranes. HepG2 cells were treated with ALLN, a protease inhibitor that has been proposed to inhibit the turnover of partially translocated forms of apoB. Although an -6-fold increase in intracellular apoB was observed in ALLN-treated cells, no corresponding increase in protease sensitivity was observed. These results indicate that the efficient translocation of apoB across the ER membrane occurs independently of its ability to undergo assembly into a secretion competent lipoprotein. Topics: Apolipoprotein B-100; Apolipoprotein B-48; Apolipoproteins B; Biological Transport; Carcinoma, Hepatocellular; Endoplasmic Reticulum; Humans; Immunoblotting; Intracellular Membranes; Leupeptins; Lipoproteins; Liver; Liver Neoplasms; Protease Inhibitors; Trypsin; Tumor Cells, Cultured | 1996 |
Aberrant stress-induced phosphorylation of perikaryal neurofilaments.
The aberrant phosphorylation of the neurofilament high molecular weight subunit (NFH) in the neuronal perikaryon is a common feature of several neurological diseases. We demonstrated a strong correlation between hyperphosphorylation of the NFH carboxyl-terminal domain and activation of stress-activated protein kinase (SAPK) -gamma in PC12 cells. Agents that activated SAPKgamma in PC12 cells also caused the hyperphosphorylation of perikaryal NFH in cultured dorsal root ganglion neurons. The NFH carboxyl-terminal domain was phosphorylated by SAPKgamma in vitro, and the use of peptide substrates indicated that this event occurred preferentially at KSPXE motifs. We propose that SAPKgamma, perhaps in concert with other SAPKs, is involved in the abnormal phosphorylation of perikaryal NFH. This finding could lead to new insights into the etiology of several neurological diseases. Topics: Amino Acid Sequence; Animals; Calcium-Calmodulin-Dependent Protein Kinases; Enzyme Activation; Fluorescent Antibody Technique, Indirect; Ganglia, Spinal; Leupeptins; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 12; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinases; Molecular Sequence Data; Nerve Growth Factors; Neurofilament Proteins; Neurons; Oligopeptides; PC12 Cells; Phosphorylation; Protease Inhibitors; Protein Kinases; Rats; Stress, Physiological; Structure-Activity Relationship | 1996 |
In HepG2 cells, translocation, not degradation, determines the fate of the de novo synthesized apolipoprotein B.
Previous studies show that translocation and degradation of apolipoprotein B (apoB), two processes occurring on or within the endoplasmic reticulum, determine how much de novo synthesized apoB is secreted. We determined which of these processes regulates the intracellular fate of apoB by examining whether degradation determines how much apoB is translocated or if translocation determines how much apoB is degraded. HepG2 cells, treated with the cysteine active site protease inhibitor ALLN, previously shown to block the degradation of translocation-arrested apoB in Chinese hamster ovary cells (Du, E., Kurth, J., Wang, S.-L., Humiston, P., and Davis, R.A. (1994) J. Biol. Chem. 269, 24169-24176), showed a 10-fold increase in the accumulation of de novo synthesized [35S]methionine-labeled apoB. The majority (80%) of the apoB accumulated in response to ALLN was in the microsomal fraction. In contrast, ALLN did not effect apoB secretion. Since ALLN did not effect the intracellular accumulation of [35S]methionine-labeled albumin and other proteins (trichloroacetic acid-precipitable [35S]methionine-labeled proteins), its effect on apoB was specific. Pulse-chase studies showed that ALLN dramatically reduced the first-order rate of removal of [35S]methionine-labeled apoB from the cell but did not effect its rate of secretion. The finding that ALLN caused the intracellular accumulation of incompletely translated chains of apoB suggests that at least some of the degradation occurs at the ribosomal level. Moreover, 85% of the apoB that accumulated in isolated microsomes in response to ALLN was accessible to exogenous trypsin, indicating this pool of apoB was incompletely translocated. The combined data suggest that translocation, not degradation, determines the intracellular fate of de novo synthesized apoB. Topics: Apolipoprotein B-100; Apolipoproteins B; Biological Transport; Cell Line; Cysteine Proteinase Inhibitors; Hydrolysis; Leupeptins; Liver; Microsomes, Liver | 1995 |
The proteasome pathway is required for cytokine-induced endothelial-leukocyte adhesion molecule expression.
Multiple cell adhesion proteins are up-regulated in vascular endothelial cells in response to TNF alpha and other inflammatory cytokines. This increase in cell adhesion gene expression is thought to require the transcription factor NF-kappa B. Here, we show that peptide aldehyde inhibitors of the proteasome, a multicatalytic protease recently shown to be required for the activation of NF-kappa B, block TNF alpha induction of the leukocyte adhesion molecules E-selectin, VCAM-1, and ICAM-1. Striking functional consequences of this inhibition were observed in analyses of leukocyte-endothelial interactions under defined flow conditions. Lymphocyte attachment to TNF alpha-treated endothelial monolayers was totally blocked, while neutrophil attachment was partially reduced but transmigration was essentially prevented. Topics: Base Sequence; Calpain; Cell Adhesion; Cell Adhesion Molecules; Cells, Cultured; Cysteine Endopeptidases; E-Selectin; Endothelium, Vascular; Gene Expression; Humans; In Vitro Techniques; Intercellular Adhesion Molecule-1; Interleukin-8; Leukocytes; Leupeptins; Molecular Sequence Data; Multienzyme Complexes; NF-kappa B; Oligodeoxyribonucleotides; Proteasome Endopeptidase Complex; Proto-Oncogene Proteins; RNA, Messenger; Time Factors; Transcription Factor RelB; Transcription Factors; Tumor Necrosis Factor-alpha; Vascular Cell Adhesion Molecule-1 | 1995 |
Multiple proteolytic systems, including the proteasome, contribute to CFTR processing.
The molecular components of the quality control system that rapidly degrades abnormal membrane and secretory proteins have not been identified. The cystic fibrosis transmembrane conductance regulator (CFTR) is an integral membrane protein to which this quality control is stringently applied; approximately 75% of the wild-type precursor and 100% of the delta F508 CFTR variant found in most CF patients are rapidly degraded before exiting from the ER. We now show that this ER degradation is sensitive to inhibitors of the cytosolic proteasome, including lactacystin and certain peptide aldehydes. One of the latter compounds, MG-132, also completely blocks the ATP-dependent conversion of the wild-type precursor to the native folded form that enables escape from degradation. Hence, CFTR and presumably other intrinsic membrane proteins are substrates for proteasomal degradation during their maturation within the ER. Topics: Acetylcysteine; Animals; ATP Binding Cassette Transporter, Subfamily B, Member 1; CHO Cells; Cricetinae; Cysteine Endopeptidases; Cystic Fibrosis; Cystic Fibrosis Transmembrane Conductance Regulator; Endopeptidases; Endoplasmic Reticulum; Humans; Leupeptins; Multienzyme Complexes; Oligopeptides; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; Recombinant Fusion Proteins; Ubiquitins | 1995 |
N- and C-terminal sequences control degradation of MAD3/I kappa B alpha in response to inducers of NF-kappa B activity.
The proteolytic degradation of the inhibitory protein MAD3/I kappa B alpha in response to extracellular stimulation is a prerequisite step in the activation of the transcription factor NF-kappa B. Analysis of the expression of human I kappa B alpha protein in stable transfectants of mouse 70Z/3 cells shows that, as for the endogenous murine protein, exogenous I kappa B alpha is degraded in response to inducers of NF-kappa B activity, such as phorbol myristate acetate or lipopolysaccharide. In addition, pretreatment of the cells with the proteasome inhibitor N-Ac-Leu-Leu-norleucinal inhibits this ligand-induced degradation and, in agreement with previous studies, stabilizes a hyperphosphorylated form of the human I kappa B alpha protein. By expressing mutant forms of the human protein in this cell line, we have been able to delineate the sequences responsible for both the ligand-induced phosphorylation and the degradation of I kappa B alpha. Our results show that deletion of the C terminus of the I kappa B alpha molecule up to amino acid 279 abolishes constitutive but not ligand-inducible phosphorylation and inhibits ligand-inducible degradation. Further analysis reveals that the inducible phosphorylation of I kappa B alpha maps to two serines in the N terminus of the protein (residues 32 and 36) and that the mutation of either residue is sufficient to abolish ligand-induced degradation, whereas both residues must be mutated to abolish inducible phosphorylation of the protein. We propose that treatment of 70Z/3 cells with either phorbol myristate acetate or lipopolysaccharide induces a kinase activity which phosphorylates serines 32 and that these phosphorylations target the protein for rapid proteolytic degradation, possibly by the ubiquitin-26S proteasome pathway, thus allowing NF-kappa B to translocate to the nucleus and to activate gene expression. Topics: Amino Acid Sequence; Animals; B-Lymphocytes; Cell Line; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Humans; I-kappa B Proteins; Leupeptins; Lipopolysaccharides; Mice; Molecular Sequence Data; Mutation; NF-kappa B; NF-KappaB Inhibitor alpha; Phosphorylation; Protein Serine-Threonine Kinases; Sequence Deletion; Serine; Tetradecanoylphorbol Acetate; Transfection | 1995 |
Inhibition of secretion from isolated rat alveolar epithelial type II cells by the cell permeant calpain inhibitor II (N-acetyl-leucyl-leucyl-methioninal).
Although several signal transduction pathways, including activation of specific protein kinases have been proposed and studied for the secretory processes of lung surfactant from alveolar epithelial type II cells, the role of proteolytic processing by calpains (calcium-activated neutral proteases) in secretion has not been investigated. Therefore, we examined the effect of cell permeable calpain inhibitor I (N-acetyl-leucyl-leucyl-norleucinal) and II (N-acetyl-leucyl-leucyl-methioninal) on secretion to test the hypothesis that calpains participate in the secretory processes of alveolar epithelial type II cells. Calpain inhibitor I preferentially inhibits micro (mu)-calpain while inhibitor II inhibits milli (m)-calpain. Isolated type II cells were prelabelled with [3H]-choline for 18-24 h. To measure secretion, [3H]-labelled disaturated phosphatidylcholine (DSPC) released in the medium was monitored. Basal secretion of DSPC was maximally (87%) depressed by the presence of 10 microM inhibitor II. Secretagogue-stimulated secretion was also modulated by inhibitor II treatment. Stimulation with calcium ionophore A23187 enhanced secretion 3-fold. However, cells pre-exposed to inhibitor II displayed a 90% reduction of calcium-stimulated secretion. Terbutaline (10 microM) and ATP (1 mM) each increased secretion 2- and 4-fold, respectively. However, the inhibitor-treated cells, exposed to the same stimuli, attained only 53 or 62% of these increases. Calpain inhibitor I, on the other hand, inhibited neither basal nor stimulated secretion. The results suggest that m-calpain, the major isozyme of lung calpain requiring mM calcium for activity in vitro, is involved in the secretory pathways of alveolar epithelial type II cells. Topics: Amino Acid Sequence; Animals; Calcium; Calpain; Cell Membrane Permeability; Epithelial Cells; Epithelium; In Vitro Techniques; Leupeptins; Molecular Sequence Data; Oligopeptides; Phosphatidylcholines; Protease Inhibitors; Pulmonary Alveoli; Rats | 1995 |
The gap junction protein connexin43 is degraded via the ubiquitin proteasome pathway.
We investigated the degradation of the gap junction protein connexin43 in E36 Chinese hamster ovary cells and rat cardiomyocyte-derived BWEM cells. Treatment of E36 cells with the lysosomotropic amine, primaquine, for 16 h doubled the amount of connexin43 detected by immunoblotting and modestly increased the half-life of connexin43 in pulse-chase studies, suggesting that the lysosome played a minor role in connexin43 proteolysis. In contrast, treatment with the proteasomal inhibitor N-acetyl-L-leucyl-L-leucinyl-norleucinal led to a 6-fold accumulation of connexin43 and increased the half-life of connexin43 to approximately 9 h. The role of ubiquitin in connexin43 degradation was examined in an E36-derived mutant, ts20, which contains a thermolabile ubiquitin-activating enzyme, E1. E36 and ts20 cells grown at the permissive temperature contained similar amounts of connexin43 detectable by immunoblotting. Heat treatment dramatically reduced the amount of connexin43 detected in E36 cells, while connexin43 levels in heat-treated ts20 cells did not change. E36 cells that were heat-treated in the presence of N-acetyl-L-leucyl-L-leucinyl-norleucinal did not lose their connexin43. Pulse-chase experiments showed the reversibility of the block to connexin43 degradation in ts20 cells that were returned to the permissive temperature. Finally, sequential immunoprecipitation using anti-connexin43 and anti-ubiquitin antibodies demonstrated polyubiquitination of connexin43. These results indicate that ubiquitin-mediated proteasomal proteolysis may be the major mechanism of degradation of connexin43. Topics: Animals; Cells, Cultured; CHO Cells; Connexin 43; Cricetinae; Cysteine Endopeptidases; Electrophoresis, Polyacrylamide Gel; Half-Life; Immunoblotting; Kinetics; Leupeptins; Multienzyme Complexes; Myocardium; Protease Inhibitors; Proteasome Endopeptidase Complex; Rats; Time Factors; Ubiquitins | 1995 |
Role of the ubiquitin-proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27.
The p27 mammalian cell cycle protein is an inhibitor of cyclin-dependent kinases. Both in vivo and in vitro, p27 was found to be degraded by the ubiquitin-proteasome pathway. The human ubiquitin-conjugating enzymes Ubc2 and Ubc3 were specifically involved in the ubiquitination of p27. Compared with proliferating cells, quiescent cells exhibited a smaller amount of p27 ubiquitinating activity, which accounted for the marked increase of p27 half-life measured in these cells. Thus, the abundance of p27 in cells is regulated by degradation. The specific proteolysis of p27 may represent a mechanism for regulating the activity of cyclin-dependent kinases. Topics: Adenosine Triphosphate; Anaphase-Promoting Complex-Cyclosome; Animals; Cell Cycle Proteins; Cell Line; Cyclin-Dependent Kinase Inhibitor p27; Cyclin-Dependent Kinases; Cysteine Endopeptidases; Electroporation; Enzyme Inhibitors; Humans; Kinetics; Leupeptins; Ligases; Mice; Microtubule-Associated Proteins; Multienzyme Complexes; Proteasome Endopeptidase Complex; Rabbits; Recombinant Proteins; Succinates; Tumor Cells, Cultured; Tumor Suppressor Proteins; Ubiquitin-Conjugating Enzymes; Ubiquitin-Protein Ligase Complexes; Ubiquitin-Protein Ligases; Ubiquitins | 1995 |
Effect and cellular site of action of cysteine protease inhibitors on the cholesterol esterification pathway in macrophages and Chinese hamster ovary cells.
Stimulation of intracellular cholesterol esterification, which is catalyzed by the enzyme acyl-CoA:cholesterol O-acyltransferase (ACAT), by atherogenic lipoproteins in macrophages is a key step in the development of atheroma foam cells. Since other aspects of intracellular cholesterol metabolism involve proteolytic reactions, we looked for evidence of intracellular proteolysis in the stimulation of the cholesterol esterification pathway. When macrophages and CHO cells were incubated with the cysteine protease inhibitor N-acetylleucylleucylnorleucinal (ALLN), the ability of beta-very-low-density lipoprotein (beta-VLDL) and free cholesterol-rich liposomes to stimulate cholesterol esterification was inhibited by 60-90%. Epoxysuccinylleucylamido-3-methylbutane ethyl ester (EST), a cysteine protease inhibitor structurally different from ALLN, also inhibited beta-VLDL-induced cholesterol esterification in CHO cells. The inhibitory effect of the protease inhibitors could not be explained by decreased net expansion of cellular cholesterol pools, inhibition of lipoprotein cholesteryl ester hydrolysis, or blockage of cholesterol trafficking through the lysosomal pathway. Furthermore, stimulation of cholesterol esterification by 25-hydroxycholesterol and sphingomyelinase was not inhibited by ALLN, indicating that ALLN is not acting as a direct ACAT inhibitor in the cells, and suggesting that the ALLN effect is specific for methods of stimulating cholesterol esterification that expand cellular cholesterol pools. Previous studies have shown that inhibition of protein synthesis (e.g., by cycloheximide) stimulates cholesterol esterification in macrophages and CHO cells, suggesting the presence of a short-lived protein inhibitor of cholesterol esterification. Herein, we show that, when added after cycloheximide, ALLN does not inhibit cycloheximide-induced cholesterol esterification in either cell type. The data in this report are consistent with a novel model in which a proteolytic reaction mediates the stimulation of cholesterol esterification specifically by expanded cellular cholesterol pools. The apparent protease-dependent step is not dependent upon lysosomal trafficking of cholesterol and is proximal to the ACAT enzyme itself; it may function by cleaving an endogenous inhibitor of the interaction of expanded cellular cholesterol pools with ACAT. Topics: Animals; CHO Cells; Cholesterol Esters; Cricetinae; Cycloheximide; Cysteine Proteinase Inhibitors; Esterification; Female; Hydroxycholesterols; Leucine; Leupeptins; Lipoproteins, VLDL; Macrophages, Peritoneal; Mice; Mice, Inbred ICR; Microscopy, Fluorescence; Sphingomyelin Phosphodiesterase | 1995 |
Crystal structure of the 20S proteasome from the archaeon T. acidophilum at 3.4 A resolution.
The three-dimensional structure of the proteasome from the archaebacterium Thermoplasma acidophilum has been elucidated by x-ray crystallographic analysis by means of isomorphous replacement and cyclic averaging. The atomic model was built and refined to a crystallographic R factor of 22.1 percent. The 673-kilodalton protease complex consists of 14 copies of two different subunits, alpha and beta, forming a barrel-shaped structure of four stacked rings. The two inner rings consist of seven beta subunits each, and the two outer rings consist of seven alpha subunits each. A narrow channel controls access to the three inner compartments. The alpha 7 beta 7 beta 7 alpha 7 subunit assembly has 72-point group symmetry. The structures of the alpha and beta subunits are similar, consisting of a core of two antiparallel beta sheets that is flanked by alpha helices on both sides. The binding of a peptide aldehyde inhibitor marks the active site in the central cavity at the amino termini of the beta subunits and suggests a novel proteolytic mechanism. Topics: Amino Acid Sequence; Archaeal Proteins; Binding Sites; Chaperonin 60; Computer Graphics; Crystallography, X-Ray; Cysteine Endopeptidases; Endopeptidases; Fourier Analysis; Hydrogen Bonding; Leupeptins; Models, Molecular; Molecular Sequence Data; Multienzyme Complexes; Protease Inhibitors; Proteasome Endopeptidase Complex; Protein Conformation; Protein Folding; Protein Structure, Secondary; Proteins; Thermoplasma | 1995 |
Parathyroid hormone-induced retraction of MC3T3-E1 osteoblastic cells is attenuated by the calpain inhibitor N-Ac-Leu-Leu-norleucinal.
Parathyroid hormone (PTH) binding to its osteoblastic receptors stimulates cytoplasmic retraction within minutes. We hypothesized that the calpains (calcium-activated papain-like enzymes) contribute to PTH-induced osteoblastic retraction by catalyzing regulatory hydrolysis of cytoskeletal structural proteins or enzymes important in cytokinesis. N-Ac-Leu-Leu-norleucinal (ALLN), a reversible calpain inhibitor, was tested for its ability to inhibit PTH-induced retraction in murine MC3T3-E1 osteoblastic cells. ALLN inhibited PTH-induced retraction for 30 minutes in cells cultured on polystyrene cultureware or gelatin-coated glass cover slips, supporting the hypothesis that PTH-induced activation of the calpains contributes to short-term changes in MC3T3-E1 cell shape. Inhibition of PTH-induced retraction occurred on two substrata, suggesting that interactions between the extracellular matrix and cell surface proteins are not the sole determinants of morphology. Intracellular events, such as hydrolysis of focal adherens junction proteins on the cytoplasmic face of the plasma membrane, may contribute to PTH-induced retraction. Topics: Animals; Calpain; Cell Adhesion; Cell Size; Cells, Cultured; Glycoproteins; Leupeptins; Mice; Osteoblasts; Parathyroid Hormone | 1995 |
The molecular mechanism of the induction of the low density lipoprotein receptor by chenodeoxycholic acid in cultured human cells.
In a cultured human hepatoblastoma cell line, Hep G2, chenodeoxycholic acid (CDCA) induced LDL receptor mRNA levels approximately 4 fold and mRNA levels for HMG-CoA reductase and HMG-CoA synthase two fold. In contrast, the mRNA levels for mevalonate kinase, farnesyl pyrophosphate synthase and squalene synthase were not changed significantly. The pattern of the induction of the sterol-sensitive genes was similar to the induction by N-acetyl-leucyl-leucyl-norleucinal (ALLN), an SREBP degradation inhibitor, suggesting that CDCA may increase mature SREBPs. CDCA could inhibit the 25-hydroxycholesterol mediated inactivation of SREBP without affecting mRNA levels of SREBPs. These results suggest that CDCA can affect sterol metabolism by a novel mechanism involving the inhibition of the oxysterol-mediated inactivation of SREBP. Topics: Alkyl and Aryl Transferases; Base Sequence; Cell Line; Chenodeoxycholic Acid; Codon; DNA-Binding Proteins; Farnesyl-Diphosphate Farnesyltransferase; Gene Expression; Geranyltranstransferase; Hepatoblastoma; Humans; Hydroxycholesterols; Hydroxymethylglutaryl CoA Reductases; Hydroxymethylglutaryl-CoA Synthase; Leupeptins; Liver Neoplasms; Phosphotransferases (Alcohol Group Acceptor); Protease Inhibitors; Receptors, LDL; Regulatory Sequences, Nucleic Acid; RNA, Messenger; Transferases; Tumor Cells, Cultured | 1995 |
Increase of cyclin B by overexpression of cystatin alpha.
Degradation of cyclin B was effectively suppressed when cells were treated with ALLN (N-acetylleucylleucylnorleucinal) which inhibits proteasome, calpain and cysteine proteinase cathepsins. In order to examine which protease degrades cyclin B, the effect of a cathepsin inhibitor, cystatin alpha, was investigated. The cystatin alpha gene was inserted into an inducible expression vector, pMSG, and transfected into NIH3T3 mouse fibroblasts. The expression of cystatin alpha was induced effectively in the transfected cells after treatment with dexamethasone. Overexpression of cystatin alpha resulted in an increase of the amount of cyclin B, suggesting that cysteine proteinase cathepsins might be involved in the degradation of cyclin B. Topics: 3T3 Cells; Animals; Anti-Inflammatory Agents; Base Sequence; Cyclins; Cystatins; Cysteine Proteinase Inhibitors; Dexamethasone; Gene Expression; Leupeptins; Mice; Molecular Sequence Data; Rats; Transfection | 1995 |
Increased phosphorylation of neurofilament subunits in PC12 cells and rat dorsal root ganglion neurons treated with N-Acetyl-Leu-Leu-norleucinal.
Treatment of PC12 cells or dorsal root ganglion neurons with the protease inhibitor, N-Acetyl-Leu-Leu-norleucinal, stimulated phosphorylation of the mid-sized and heavy neurofilament subunits and caused the heavy subunit in the perikarya of dorsal root ganglion neurons to become hyperphosphorylated. The closely related inhibitor, N-Acetyl-Leu-Leu-methioninal, did not produce a similar effect. Okadaic acid increased the phosphorylation state of the heavy neurofilament subunit in PC12 cells in a fashion similar to N-Acetyl-Leu-Leu-norleucinal and the effect of both compounds together was greater than for either one alone. There was no increase in cyclin-dependent kinase 5-immunoprecipitable histone H1 kinase activity in PC12 cells treated with N-Acetyl-Leu-Leu-norleucinal despite the presence of enzyme protein. The present study demonstrates that a protease inhibitor can induce the hyperphosphorylation of neurofilament subunits to a level normally seen only in axons. This suggests that perturbations in intracellular proteolysis may lead to the accumulation of phosphorylated neurofilament epitopes in neuronal perikarya in certain pathological states. The results also show that the carboxy-terminal tail domains of the two largest neurofilament subunits are phosphorylated even when cyclin dependent kinase 5 is inactive, indicating that other neuronal kinases are involved in the phosphorylation of Lys-Ser-Pro repeats. Topics: Amino Acid Sequence; Animals; Blotting, Western; Electrophoresis, Polyacrylamide Gel; Ganglia, Spinal; Humans; Hydrogen-Ion Concentration; Leupeptins; Molecular Sequence Data; Neurofilament Proteins; Neurons; PC12 Cells; Phosphorylation; Precipitin Tests; Protein Kinases; Rats; Stimulation, Chemical | 1995 |
The regulated degradation of a 3-hydroxy-3-methylglutaryl-coenzyme A reductase reporter construct occurs in the endoplasmic reticulum.
The rate-limiting enzyme in cholesterol biosynthesis, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG CoA) reductase, is regulated at a number of levels. One important mechanism is regulation of the half-life of the protein by a controlled proteolytic system. This comes about in response to downstream products of the sterol biosynthetic pathway. Little is known about this system, including where in the cell this regulated degradation occurs. HMG CoA reductase resides in the endoplasmic reticulum. To localize the site of regulated degradation of HMG CoA reductase, we used a construct that fuses the N-terminal membrane-anchoring domain of HMG CoA reductase in-frame with beta-galactosidase as a reporter domain (HM-Gal). HM-Gal has previously been shown to reproduce faithfully the degradative properties of native HMG CoA reductase (Chun et al. (1990) J. Biol. Chem. 265, 22004-22010). CHO cells transfected with DNA encoding HM-Gal were exposed to mevalonic acid, which enhances the rate of HMG CoA reductase degradation several fold, and leads to the reduction of the steady state levels of HM-Gal by 80-90%. To accumulate HMG CoA reductase at the site of degradation, cells were simultaneously treated with N-acetyl-leucyl-leucyl-norleucinal (ALLN), which inhibits the protease responsible for reductase degradation. HM-Gal was localized morphologically by immunofluorescence and biochemically by measuring beta-galactosidase activity in Percoll gradients of cellular homogenates. Using either technique HM-Gal localization was indistinguishable from that of ER markers in both control cells and in cells treated to accumulate HMG CoA reductase at the site of degradation.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; beta-Galactosidase; Biological Transport; CHO Cells; Cricetinae; Cricetulus; Endopeptidases; Endoplasmic Reticulum; Gene Expression Regulation, Enzymologic; Genes, Reporter; Half-Life; Hydroxymethylglutaryl CoA Reductases; Leupeptins; Membrane Glycoproteins; Protease Inhibitors; Protein Processing, Post-Translational; Recombinant Fusion Proteins; Viral Envelope Proteins | 1994 |
Sterol mediated regulation of SREBP-1a,1b,1c and SREBP-2 in cultured human cells.
Under conditions of cholesterol depletion and SREBP-1 accumulation, changes in the levels of sterol regulatory element binding protein(s) (SREBPs) and sterol regulated gene mRNA were studied in Hep G2 cells by RNase protection assay. Cholesterol depletion increased the expression of mRNAs for cholesterol biosynthetic enzymes and low density lipoprotein (LDL) receptor. mRNAs levels for SREBP-1c and SREBP-2 were also increased by the cholesterol depletion. In contrast, levels for SREBP-1a and 1b (1a/b) mRNA increased transiently and then decreased. To examine the effect of SREBP-1 accumulation, Hep G2 cells were incubated with a SREBP-1 degradation inhibitor, N-acetyl-leucyl-leucyl-norleucinal (ALLN). The ALLN treatment increased the LDL receptor mRNA significantly, and also increased mRNA levels for HMG-CoA reductase, SREBP-1a/b and SREBP-2. The mRNA level for squalene synthase was not changed, and for SREBP-1c was decreased by the treatment. In conclusion, the regulation of differential expression of SREBP mRNA may be involved in sterol mediated regulation of gene expression. Moreover, the regulation of the SREBP-1 level may be a critical step in the regulation of sterol mediated LDL receptor expression. Topics: Amino Acid Sequence; CCAAT-Enhancer-Binding Proteins; Cholesterol; Cysteine Proteinase Inhibitors; DNA-Binding Proteins; Gene Expression Regulation; Humans; Leupeptins; Molecular Sequence Data; Nuclear Proteins; RNA, Messenger; Sterol Regulatory Element Binding Protein 1; Sterol Regulatory Element Binding Protein 2; Sterols; Transcription Factors; Tumor Cells, Cultured | 1994 |
Inhibitors of the proteasome block the degradation of most cell proteins and the generation of peptides presented on MHC class I molecules.
Reagents that inhibit the ubiquitin-proteasome proteolytic pathway in cells have not been available. Peptide aldehydes that inhibit major peptidase activities of the 20S and 26S proteasomes are shown to reduce the degradation of protein and ubiquitinated protein substrates by 26S particles. Unlike inhibitors of lysosomal proteolysis, these compounds inhibit the degradation of not only abnormal and short-lived polypeptides but also long-lived proteins in intact cells. We used these agents to test the importance of the proteasome in antigen presentation. When ovalbumin is introduced into the cytosol of lymphoblasts, these inhibitors block the presentation on MHC class I molecules of an ovalbumin-derived peptide by preventing its proteolytic generation. By preventing peptide production from cell proteins, these inhibitors block the assembly of class I molecules. Therefore, the proteasome catalyzes the degradation of the vast majority of cell proteins and generates most peptides presented on MHC class I molecules. Topics: Amino Acid Sequence; Animals; Antigen Presentation; B-Lymphocytes; Cysteine Endopeptidases; Dose-Response Relationship, Drug; Hematopoietic Stem Cells; Histocompatibility Antigens Class I; Leupeptins; Lymphocytes; Mice; Molecular Sequence Data; Multienzyme Complexes; Ovalbumin; Protease Inhibitors; Proteasome Endopeptidase Complex; T-Lymphocytes; Ubiquitins | 1994 |
The ubiquitin-proteasome pathway is required for processing the NF-kappa B1 precursor protein and the activation of NF-kappa B.
We demonstrate an essential role for the proteasome complex in two proteolytic processes required for activation of the transcription factor NF-kappa B. The p105 precursor of the p50 subunit of NF-kappa B is processed in vitro by an ATP-dependent process that requires proteasomes and ubiquitin conjugation. The C-terminal region of p105 is rapidly degraded, leaving the N-terminal p50 domain. p105 processing can be blocked in intact cells with inhibitors of the proteasome or in yeast with proteasome mutants. These inhibitors also block the activation of NF-kappa B and the rapid degradation of I kappa B alpha induced by tumor necrosis factor alpha. Thus, the ubiquitin-proteasome pathway functions not only in the complete degradation of polypeptides, but also in the regulated processing of precursors into active proteins. Topics: Adenosine Triphosphate; Amino Acid Sequence; Animals; Cells, Cultured; Cysteine Endopeptidases; DNA-Binding Proteins; Humans; I-kappa B Proteins; Leupeptins; Models, Biological; Molecular Sequence Data; Multienzyme Complexes; NF-kappa B; NF-kappa B p50 Subunit; NF-KappaB Inhibitor alpha; Protease Inhibitors; Proteasome Endopeptidase Complex; Protein Precursors; Protein Processing, Post-Translational; Ubiquitins | 1994 |
Comparison of the effect of calpain inhibitors on two extralysosomal proteinases: the multicatalytic proteinase complex and m-calpain.
The potencies of three peptide aldehyde inhibitors of calpain (calpain inhibitors 1 and 2 and calpeptin) as inhibitors of four catalytic activities of the multicatalytic proteinase complex (MPC) were compared with their potencies as inhibitors of m-calpain. The chymotrypsinlike activity (cleavage after hydrophobic amino acids) and the caseinolytic activity (degradation of beta-casein) of MPC were strongly inhibited by calpain inhibitors 1 and 2 (IC50 values in the low micromolar range). Cleavage by MPC after acidic amino acids (peptidylglutamyl-peptide bond hydrolyzing activity) and basic amino acids (trypsinlike activity) was inhibited less effectively, declining moderately with increasing concentrations of calpain inhibitors 1 and 2. Calpeptin only weakly inhibited the four MPC activities, yet was the most potent inhibitor of m-calpain. These results indicate that caution must be exercised when calpain inhibitors 1 and 2 are used to infer calpain function. Calpeptin may be a better choice for such studies, although its effect on other cysteine or serine proteinases remains to be determined. Topics: Amino Acid Sequence; Animals; Calpain; Cattle; Cysteine Endopeptidases; Dipeptides; Kinetics; Leupeptins; Molecular Sequence Data; Multienzyme Complexes; Oligopeptides; Pituitary Gland; Protease Inhibitors; Proteasome Endopeptidase Complex; Substrate Specificity | 1994 |
Proteolysis and lipid-facilitated translocation are distinct but competitive processes that regulate secretion of apolipoprotein B in Hep G2 cells.
Under lipid-poor conditions, most newly synthesized apolipoprotein B100 (apoB) undergoes rapid degradation in Hep G2 cells such that only a small fraction of newly synthesized apoB is actually secreted. Addition of oleate to Hep G2 culture medium stimulates apoB secretion by a post-translational mechanism. In the current studies we have explored oleate-stimulation of apoB secretion by using calpain inhibitor I, N-acetyl-leucyl-leucyl-norleucinal (ALLN), a compound that inhibits the intracellular degradation of 3-hydroxy-3-methylglutaryl-coenzyme A reductase and the T cell receptor alpha subunit. Preincubation of Hep G2 cells with ALLN (40 micrograms/ml) for 1 h markedly inhibited degradation of newly synthesized apoB. Whereas only 32% of newly labeled apoB remained intact (cells+medium) in control cells after a 10-min pulse with [3H]leucine followed by a 60-min chase, 84% of labeled apoB was intact in ALLN-treated cells. However, most of the ALLN-protected apoB remained intracellular, as ALLN did not stimulate the rate of apoB secretion over the control rate (12 versus 9.2%). Although secretion of apoB was not accelerated, the protection afforded by ALLN continued for several hours, and labeled apoB continued to be secreted over 3 h of chase after which secretion ceased. The protection afforded by ALLN resulted in 37% of labeled apoB secreted by 3 h compared to 15% in control cells. In contrast, simultaneous treatment of cells with ALLN and oleate both accelerated and increased total apoB secretion, such that 36% of initially labeled apoB was recovered in the medium by 60 min and 71% of labeled apoB was secreted by 180 min of chase. These data show that ALLN and oleate affect apoB metabolism by different mechanisms. Although ALLN can protect nascent apoB from rapid early intracellular degradation, it does not accelerate apoB secretion. In contrast, although our results can not rule out the possibility that oleate may directly inhibit proteolysis of apoB, oleate appears to protect apoB mainly by facilitating transport of apoB out of a protease-containing compartment associated with the endoplasmic reticulum. Topics: Apolipoproteins B; Biological Transport; Calpain; Humans; In Vitro Techniques; Leupeptins; Liver; Oleic Acid; Oleic Acids; Protein Processing, Post-Translational; Secretory Rate; Tumor Cells, Cultured | 1993 |
Endosomal proteolysis precedes ricin A-chain toxicity in macrophages.
Ricin A-chain is delivered into macrophages via receptor-mediated endocytosis. We have found that following uptake via the mannose receptor, ricin A-chain is rapidly cleaved by endosomal proteases. Inhibition of endosomal proteases such as cathepsin D and B leads to the accumulation of toxin inside the cell. Inhibition of cathepsin D reduces ricin A-chain cytotoxicity, while blocking cathepsin B enhances cytotoxicity. Similar results were obtained using fibroblasts transfected with the mannose receptor. Our data strongly suggest that the activation or membrane translocation of ricin A-chain is dependent upon the action of specific proteases. Topics: Animals; Bone Marrow; Bone Marrow Cells; Cathepsin B; Cathepsin D; Cells, Cultured; Intracellular Membranes; Leupeptins; Macrophages; Male; Mice; Protease Inhibitors; Ricin | 1993 |
In vivo inhibition of cyclin B degradation and induction of cell-cycle arrest in mammalian cells by the neutral cysteine protease inhibitor N-acetylleucylleucylnorleucinal.
The cytotoxic neutral cysteine protease inhibitor N-acetylleucylleucylnorleucinal (ALLN) inhibits cell-cycle progression in CHO cells, affecting the G1/S and metaphase-anaphase transition points, as well as S phase. Mitotic arrest induced by ALLN is associated with the inhibition of cyclin B degradation. At mitosis-inhibiting concentrations of ALLN, cells undergo nuclear-envelope breakdown, spindle formation, chromosome condensation, and congression to the metaphase plate. However, normal anaphase events do not occur, and cells arrest in a metaphase configuration for a prolonged period. Steady-state levels of cyclin B increase to greater than normal mitotic levels, and cyclin B is not degraded for an extended period. Histone H1 kinase activity remains elevated during mitotic arrest. Duration of mitotic arrest depends on ALLN concentration; high concentrations (> 50 micrograms/ml) produce a prolonged mitotic arrest, whereas at lower concentrations, cells are transiently delayed through mitosis (up to 4-12 hr), after which they undergo aberrant cell division resulting in randomly sized daughter cells containing variable amounts of DNA. Cyclin B degradation fails to occur, and histone H1 kinase remains activated for the duration of mitotic arrest at all ALLN concentrations. Topics: Animals; Cell Cycle; CHO Cells; Cricetinae; Cyclins; Cysteine Proteinase Inhibitors; Flow Cytometry; Fluorescent Antibody Technique; Kinetics; Leupeptins; Mitosis; Protamine Kinase; Time Factors | 1993 |
Cellular detoxification of tripeptidyl aldehydes by an aldo-keto reductase.
Calpain inhibitor I, N-acetyl-leucyl-leucyl-norleucinal (ALLN), a cell-permeable synthetic tripeptide with an aldehyde at its C terminus specifically inhibits the activity of cysteine proteases. Since the regulated degradation of 3-hydroxy-3-methylglutaryl-CoA reductase in Chinese hamster ovary (CHO) cells is blocked by ALLN and ALLN has a cytotoxic effect on cells, we attempted to isolate ALLN-resistant cells that overproduce an ALLN-sensitive protease(s). However, we obtained an ALLN-resistant cell line that overproduced P-glycoprotein (Sharma, R. C., Inoue, S., Roitelman, J., Schimke, R. T., and Simoni, R. D. (1992) J. Biol. Chem. 267, 5731-5734). To circumvent the multidrug resistance (MDR) phenotype during selection, we have stepwise selected an ALLN-resistant cell line of CHO cells in the presence of verapamil, a competitive inhibitor of P-glycoprotein. These non-MDR ALLN-resistant cells overexpress a 35-kDa protein and have increased aldo-keto reductase activity. Partial amino acid sequences of the 35-kDa protein are highly homologous to members of the aldo-keto reductase superfamily. The aldo-keto reductases are NADPH-dependent oxidoreductases and catalyze reduction of a wide range of carbonyl compounds such as aldehydes, sugars, and ketones. Our findings support the concept that a physiological function for aldo-keto reductases may be detoxification. Topics: Alcohol Oxidoreductases; Amino Acid Sequence; Animals; Cells, Cultured; CHO Cells; Cricetinae; Cytokinins; Drug Resistance; Humans; Leupeptins; Molecular Sequence Data; Oligopeptides; Protease Inhibitors | 1993 |
Inhibition of apolipoprotein E degradation in a post-Golgi compartment by a cysteine protease inhibitor.
In our prior studies on lipoprotein stimulation of apolipoprotein E (apoE) secretion in HepG2 cells, it became clear that a proportion of the newly synthesized apoE was degraded intracellularly (Ye, S. Q., Olson, L. M., Reardon, C. A., and Getz, G. S. (1992) J. Biol. Chem. 267, 21961-21966). The present study was designed to determine the nature of the proteases and the intracellular sites involved in newly synthesized apoE degradation. The effect of seven protease inhibitors on total apoE levels was examined. Only N-acetyl-leucyl-leucyl-norleucinal (ALLN), a cysteine protease inhibitor, significantly blocked apoE degradation in HepG2 cells. The amount of total apoE from cells chased with ALLN for 4 h was increased by 1.58 +/- 0.05-fold relative to the controls (n = 11, p < 0.01). ALLN extended the half-life of apoE from 2.61 h to 4.38 h (p < 0.01). This effect occurs in a post-Golgi compartment since in the presence of brefeldin A, ALLN had no effect on intracellular apoE levels. Chloroquine and NH4Cl significantly reduced apoE degradation; however, ALLN plus either of these reagents appear to have an additive effect. The amount of apoE in cells chased in Ca(2+)-free medium was significantly higher than that in cells chased in Ca(2+)-containing medium (1.70 +/- 0.07-fold, n = 6, p < 0.01). ALLN plus Ca(2+)-free medium had no additive effect. ALLN had no significant influence on the degradation of albumin but had a similar effect on transfected apoE in Chinese hamster ovary cells. Overall, these data suggest that apoE may be degraded in a post-Golgi compartment of HepG2 and Chinese hamster ovary cells by lysosomal enzymes and cytosolic Ca(2+)-dependent cysteine proteases. ALLN inhibits the latter. Topics: Albumins; Ammonium Chloride; Animals; Apolipoproteins E; Brefeldin A; Calcium; Cathepsin B; Cathepsin L; Cathepsins; Chloroquine; CHO Cells; Cricetinae; Cyclopentanes; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Endopeptidases; Golgi Apparatus; Humans; Kinetics; Leupeptins; Molecular Sequence Data; Transfection; Tumor Cells, Cultured | 1993 |
Peptide transport by the multidrug resistance pump.
The membrane P-glycoprotein (P170) is an ATP-hydrolyzing transmembrane pump, and elevated levels of P170, due to higher expression with or without amplification of the multidrug resistance gene (mdr1), result in resistance to a variety of chemotherapeutic agents in mammalian cells. The function of the P170 pump has been proposed as a protection against toxic substances present in animal diets. Here we describe a Chinese hamster ovary cell line that was selected for resistance to a synthetic tripeptide, N-acetyl-leucyl-leucyl-norleucinal (ALLN). This ALLN-resistant variant shows the classical multidrug resistance (MDR) phenotype, including overexpression and amplification of the mdr1 gene. Additionally, a mouse embryo cell line overexpressing the transfected mdr1 gene is likewise resistant to ALLN. Our results demonstrate that P170 is capable of transporting peptides and raise the possibility that the mdr1 gene product or other MDR-like genes, present in the genome of mammalian cells, may be involved in secretion of peptides or cellular proteins as is the case with the structurally similar hylB and ste6 gene products of Escherichia coli and yeast, respectively. Topics: 3T3 Cells; Animals; ATP Binding Cassette Transporter, Subfamily B, Member 1; Cell Survival; CHO Cells; Cricetinae; Drug Resistance; Escherichia coli; Kinetics; Leupeptins; Membrane Glycoproteins; Mice; Phenotype; Protease Inhibitors | 1992 |
Neuronal fodrin proteolysis occurs independently of excitatory amino acid-induced neurotoxicity.
In cultured cerebellar granule cells, the total amount of fodrin alpha subunit increased 3-fold between 0 and 10 days in vitro and fodrin mRNA increased 5-fold. The exposure of cerebellar neurons to NMDA induced the accumulation of a 150 kd proteolytic fragment of fodrin. The NMDA-induced breakdown of fodrin was time-, concentration-, and Ca2(+)-dependent and was inhibited by APV, Mg2+, or the calpain I inhibitor N-acetyl-Leu-Leu-norleucinal. Kainate caused fodrin proteolysis through indirect activation of NMDA receptors. Quisqualate was ineffective. The NMDA-induced degradation of fodrin occurred under conditions that did not cause degeneration of cultured cerebellar neurons. These results show that Ca2+/calpain I-dependent proteolysis of fodrin is selectively associated with NMDA receptor activation; however, fodrin proteolysis per se does not play a causal role in NMDA-induced toxicity in cerebellar granule cells. Topics: Animals; Blotting, Northern; Calcium; Calcium Channels; Calpain; Carrier Proteins; Cattle; Cells, Cultured; Cerebellum; Dose-Response Relationship, Drug; Fluorescent Antibody Technique; Kainic Acid; Leupeptins; Microfilament Proteins; N-Methylaspartate; Neurons; Receptors, Glutamate; Receptors, N-Methyl-D-Aspartate; Receptors, Neurotransmitter; RNA, Messenger; Time Factors | 1991 |
Inhibition of degradation of 3-hydroxy-3-methylglutaryl-coenzyme A reductase in vivo by cysteine protease inhibitors.
3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase is a key regulatory enzyme of cholesterol biosynthesis and is located in the endoplasmic reticulum (ER). A fusion protein, HMGal, consisting of the membrane domain of HMG-CoA reductase fused to Escherichia coli beta-galactosidase and expressed in Chinese hamster ovary (CHO) cells from the SV40 promoter, was previously constructed and was found to respond to regulatory signals for degradation in a similar fashion to the intact HMG-CoA reductase. Degradation of both HMG-CoA reductase and HMGal in CHO cells was enhanced by addition of mevalonate or low density lipoprotein (LDL). In this report we show that 2 cysteine protease inhibitors, N-acetyl-leucyl-leucyl-norleucinal (ALLN) and N-acetyl-leucyl-leucyl-methioninal (ALLM), completely inhibit the mevalonate- or LDL-accelerated degradation of HMG-CoA reductase and HMGal and also block the basal degradation of these enzymes. It has been shown that in vitro these protease inhibitors inhibit the activities of Ca(2+)-dependent neutral proteases as well as lysosomal proteases, including cathepsin L, cathepsin b, and cathepsin D. However, the mevalonate-accelerated degradation of HMG-CoA reductase and HMGal is not affected by lysosomotropic agents, suggesting that the site of action of these inhibitor peptides in preventing the degradation is not the cathepsins. In brefeldin A-treated cells, where protein export from the ER is blocked, ALLN is still effective in inhibiting the degradation of HMG-CoA reductase and HMGal. These results indicate the involvement of non-lysosomal Ca(2+)-dependent proteases in the basal and the accelerated degradation of HMG-CoA reductase and HMGal. Enzymatic assays in vitro and immunoblot analyses have revealed calpain- and calpastatin-like proteins in CHO cells. The activities and the amount of these proteins do not change under conditions of enhanced degradation, indicating that the levels of these proteins are not subject to mevalonate regulation. Topics: Animals; beta-Galactosidase; Calcium; Cell Line; Chloroquine; Cysteine Proteinase Inhibitors; Egtazic Acid; Hydroxymethylglutaryl CoA Reductases; Kinetics; Leupeptins; Oligopeptides; Recombinant Fusion Proteins; Transfection | 1991 |
Multiple proteases regulate neurite outgrowth in NB2a/dl neuroblastoma cells.
Mouse NB2a/dl neuroblastoma cells elaborate axonal neurites in response to various chemical treatments including dibutyryl cyclic AMP and serum deprivation. Hirudin, a specific inhibitor of thrombin, initiated neurite outgrowth in NB2a/dl cells cultured in the presence of serum; however, these neurites typically retracted within 24 h. The cysteine protease inhibitors leupeptin and N-acetyl-leucyl-leucyl-norleucinal (CI; preferential inhibitor of micromolar calpain but also inhibits millimolar calpain) at 10(-6) M considerably enhanced neurite outgrowth induced by serum deprivation, but could not induce neuritogenesis in the presence of serum. A third cysteine protease inhibitor, N-acetyl-leucyl-leucyl-methional (CII; preferential inhibitor of millimolar calpain but also inhibits micromolar calpain), had no detectable effects by itself. Cells treated simultaneously with hirudin and either leupeptin, CI, or CII elaborated stable neurites in the presence of serum. Cell-free enzyme assays demonstrated that hirudin inhibited thrombin but not calpain, CI and CII inhibited calpain but not thrombin, and leupeptin inhibited both proteases. These results imply that distinct proteolytic events, possibly involving more than one protease, regulate the initiation and subsequent elongation and stabilization of axonal neurites. Since the addition of exogenous thrombin or calpain to serum-free medium did not modify neurite outgrowth, the proteolytic events affected by these inhibitors may be intracellular or involve proteases distinct from thrombin or calpain. Topics: Animals; Axons; Blood; Calpain; Endopeptidases; Hirudins; Leupeptins; Neuroblastoma; Osmolar Concentration; Protease Inhibitors; Thrombin; Tumor Cells, Cultured | 1991 |
Possible involvement of a novel protease in neurite outgrowth of PC12 cells.
A tripeptide aldehyde protease inhibitor (Ac-Leu-Leu-Nle-al, ALLNal), among many other protease inhibitors, initiates neurite outgrowth in PC12h cells. The neuritogenesis induced by this inhibitor is different in some aspects from that induced by NGF; one or two long neurites are initiated from the cell body and differentiation is transient. The results provide evidence that the protease inhibitor and nerve growth factor elicit neurite initiation by different mechanisms and suggest the existence of a novel protease which modulates neurite initiation in PC12 cells. To identify the target protease, Leu-Leu-Leu-al was immobilized and used as a ligand for affinity chromatography of the protease. A protein with an apparent molecular weight of 33 kDa was isolated specifically from the cytoplasmic fraction of PC12 cells using the affinity column. The same protein was identified in the brains of 1-day postnatal rats, but the amount was much less in the brains of adult rats. Thus, we suggest that the 33-kDa protein regulates neurite initiation in nervous systems, possibly as a protease which degrades membrane proteins or the cytoskeletal framework. Topics: Adrenal Gland Neoplasms; Animals; Axons; Chromatography, Affinity; Dose-Response Relationship, Drug; Endopeptidases; Leupeptins; Molecular Weight; Osmolar Concentration; Pheochromocytoma; Protease Inhibitors; Tumor Cells, Cultured | 1990 |
The neurite-initiating effect of a tripeptide aldehyde protease inhibitor on PC12h cells.
We report here the possible involvement of a new protease in neurite initiation by PC12h cells. Addition of a leupeptin analogue (Ac-Leu-Leu-Nle-al, ALLNal) to PC12h cells on culture plates coated with collagen type I caused de novo neurite outgrowth. Other protease inhibitors (Ac-Leu-Leu-Met-al, leupeptin, E64c, E64d, soybean trypsin inhibitor, hirudin, aprotinin, diisofluorophosphate, 6-aminocapric acid, and pepstatin A) could not mimic this neurite-initiating action. ALLNal induced the initiation of one or two long neurites from the cell body, and increased the cellular level of acetylcholinesterase to an extent similar to nerve growth factor (NGF). However, ALLNal-induced neuritogenesis is different from that induced by NGF, in which many neurites are induced from a single cell body. In addition, in contrast to neurons induced by NGF, which survive for a long time, ALLNal-induced differentiation was transient, and after 48 h percentage of cells bearing neurites started to decrease. After about 120 h exposure to ALLNal, neurites had mostly disappeared and the acetylcholinesterase activity level was not as great as that produced by NGF. These results provide evidence that ALLNal and NGF elicit neurite initiation by different mechanisms, and suggest the existence of a regulatory system of neuronal differentiation through specific protease-protease inhibitor interaction. Topics: Acetylcholinesterase; Amino Acid Sequence; Animals; Axons; Bucladesine; Cell Adhesion; Chemical Phenomena; Chemistry, Physical; Collagen; Growth; Leupeptins; Molecular Sequence Data; Nerve Fibers; Nerve Growth Factors; Oligopeptides; Pheochromocytoma; Protease Inhibitors; Rats; Time Factors; Tumor Cells, Cultured | 1989 |