benzyloxycarbonylleucyl-leucyl-leucine-aldehyde and epoxomicin

benzyloxycarbonylleucyl-leucyl-leucine-aldehyde has been researched along with epoxomicin* in 62 studies

Reviews

2 review(s) available for benzyloxycarbonylleucyl-leucyl-leucine-aldehyde and epoxomicin

ArticleYear
Viral entry and the ubiquitin-proteasome system.
    Cellular microbiology, 2021, Volume: 23, Issue:2

    Viruses confiscate cellular components of the ubiquitin-proteasome system (UPS) to facilitate many aspects of the infectious cycle. The 26S proteasome is an ATP-dependent, multisubunit proteolytic machine present in all eukaryotic cells. The proteasome executes the controlled degradation of functional proteins, as well as the hydrolysis of aberrantly folded polypeptides. There is growing evidence for the role of the UPS in viral entry. The UPS assists in several steps of the initiation of infection, including endosomal escape of the entering virion, intracellular transport of incoming nucleocapsids and uncoating of the viral genome. Inhibitors of proteasome activity, including MG132, epoxomicin, lactacystin and bortezomib have been integral to developments in this area. Here, we review the mechanistic details of UPS involvement in the entry process of viruses from a multitude of families. The possibility of proteasome inhibitors as therapeutic antiviral agents is highlighted.

    Topics: Acetylcysteine; Animals; Antiviral Agents; Bortezomib; Host Microbial Interactions; Humans; Leupeptins; Nucleocapsid; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Proteolysis; Ubiquitin; Virion; Virus Internalization; Virus Physiological Phenomena; Viruses

2021
Turnover of StAR protein: roles for the proteasome and mitochondrial proteases.
    Molecular and cellular endocrinology, 2007, Volume: 265-266

    Steroidogenic acute regulatory protein (StAR) is a mitochondrial protein essential for massive synthesis of steroid hormones in the adrenal and the gonads. Our studies suggest that once synthesized on free polyribosomes, StAR preprotein either associates with the outer mitochondrial membrane to mediate transfer of cholesterol substrate required for steroidgenesis, or it is degraded by the proteasome. Proteasome inhibitors can prevent the turnover of StAR preprotein and other matrix-targeted preproteins. Once imported, excessive accumulation of inactive StAR in the matrix is avoided by a rapid turnover. Unexpectedly, mitochondrial StAR turnover can be inhibited by two proteasome inhibitors, i.e., MG132 and clasto-lactacystin beta-lactone, but not epoxomicin. Use of those inhibitors and immuno-electron microscopy data enabled a clear distinction between two pools of intra-mitochondrial StAR, one degraded by matrix protease(s) shortly after import, while the rest of the protein undergoes a slower and inhibitor resistant degradation following translocation onto to the matrix face of the inner membranes.

    Topics: Animals; Humans; Lactones; Leupeptins; Mitochondria; Mitochondrial Membranes; Oligopeptides; Peptide Hydrolases; Phosphoproteins; Protease Inhibitors; Proteasome Endopeptidase Complex

2007

Other Studies

60 other study(ies) available for benzyloxycarbonylleucyl-leucyl-leucine-aldehyde and epoxomicin

ArticleYear
Effects of proteasome inhibitors on cytokines, metalloproteinases and their inhibitors and collagen type-I expression in periprosthetic tissues and fibroblasts from loose arthroplasty endoprostheses.
    Connective tissue research, 2019, Volume: 60, Issue:6

    Topics: Arthroplasty, Replacement, Knee; Bone-Implant Interface; Collagen Type I; Collagenases; Female; Fibroblasts; Gene Expression Regulation; Humans; Interleukin-6; Knee Prosthesis; Leupeptins; Male; Oligopeptides; Tissue Inhibitor of Metalloproteinase-1

2019
The Effects of Proteasome Inhibitors on Telomerase Activity and Regulation in Multiple Myeloma Cells.
    International journal of molecular sciences, 2019, May-21, Volume: 20, Issue:10

    The importance of telomerase, the enzyme that maintains telomere length, has been reported in many malignancies in general and in multiple myeloma (MM) in particular. Proteasome inhibitors are clinically used to combat effectively MM. Since the mechanism of action of proteasome inhibitors has not been fully described we sought to clarify its potential effect on telomerase activity (TA) in MM cells. Previously we showed that the first generation proteasome inhibitor bortezomib (Brt) inhibits TA in MM cells by both transcriptional and post-translational mechanisms and has a potential clinical significance. In the current study we focused around the anti- telomerase activity of the new generation of proteasome inhibitors, epoxomicin (EP) and MG-132 in order to clarify whether telomerase inhibition represents a class effect. We have exposed MM cell lines, ARP-1, CAG, RPMI 8226 and U266 to EP or MG and the following parameters were assessed: viability; TA, hTERT expression, the binding of hTERT (human telomerase reverse transcriptase) transcription factors and post-translational modifications. Epoxomicin and MG-132 differentially downregulated the proliferation and TA in all MM cell lines. The downregulation of TA and the expression of hTERT were faster in CAG than in ARP-1 cells. Epoxomicin was more potent than MG-132 and therefore further mechanistic studies were performed using this compound. The inhibition of TA was mainly transcriptionally regulated. The binding of three positive regulator transcription factors: SP1, c-Myc and NF-κB to the hTERT promoter was decreased by EP in CAG cells as well as their total cellular expression. In ARP-1 cells the SP1 and c-MYC binding and protein levels were similarly affected by EP while NF-κB was not affected. Interestingly, the transcription factor WT-1 (Wilms' tumor-1) exhibited an increased binding to the hTERT promoter while its total cellular amount remained unchanged. Our results combined with our previous study of bortezomib define telomerase as a general target for proteasome inhibitors. The inhibitory effect of TA is exerted by several regulatory levels, transcriptional and post translational. SP1, C-Myc and NF-κB were involved in mediating these effects. A novel finding of this study is the role of WT-1 in the regulation of telomerase which appears as a negative regulator of hTERT expression. The results of this study may contribute to future development of telomerase inhibition as a therapeutic modality in

    Topics: Cell Line, Tumor; Gene Expression Regulation, Neoplastic; Humans; Leupeptins; Multiple Myeloma; NF-kappa B; Oligopeptides; Promoter Regions, Genetic; Proteasome Inhibitors; Sp1 Transcription Factor; Telomerase; Transcription, Genetic

2019
Functional proteasome complex is required for turnover of islet amyloid polypeptide in pancreatic β-cells.
    The Journal of biological chemistry, 2018, 09-14, Volume: 293, Issue:37

    Human islet amyloid polypeptide (hIAPP) is the principal constituent of amyloid deposits and toxic oligomers in the pancreatic islets. Together with hyperglycemia, hIAPP-derived oligomers and aggregates are important culprits in type 2 diabetes mellitus (T2DM). Here, we explored the role of the cell's main proteolytic complex, the proteasome, in hIAPP turnover in normal and stressed β-cells evoked by chronic hyperglycemia. Moderate inhibition (10-35%) of proteasome activity/function in cultured human islets by the proteasome inhibitor lactacystin enhanced intracellular accumulation of hIAPP. Unexpectedly, prolonged (>1 h) and marked (>50%) impairment of proteasome activity/function had a strong inhibitory effect on hIAPP transcription and secretion from normal and stressed β-cells. This negative compensatory feedback mechanism for controlling IAPP turnover was also observed in the lactacystin-treated rat insulinoma β-cell line (INS 832/13), demonstrating the presence of an evolutionarily conserved mechanism for IAPP production. In line with these

    Topics: Acetylcysteine; Animals; Cell Line, Tumor; Diabetes Mellitus, Type 2; Down-Regulation; Hepatocyte Nuclear Factor 3-beta; Humans; Insulin-Secreting Cells; Insulinoma; Islet Amyloid Polypeptide; Leupeptins; Mice; Oligopeptides; Promoter Regions, Genetic; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Rats

2018
Intramitochondrial adenylyl cyclase controls the turnover of nuclear-encoded subunits and activity of mammalian complex I of the respiratory chain.
    Biochimica et biophysica acta, 2015, Volume: 1853, Issue:1

    In mammalian cells the nuclear-encoded subunits of complex I are imported into mitochondria, where they are assembled with mt-DNA encoded subunits in the complex, or exchanged with pre-existing copies in the complex. The present work shows that in fibroblast cultures inhibition by KH7 of cAMP production in the mitochondrial matrix by soluble adenylyl cyclase (sAC) results in decreased amounts of free non-incorporated nuclear-encoded NDUFS4, NDUFV2 and NDUFA9 subunits of the catalytic moiety and inhibition of the activity of complex I. Addition of permeant 8-Br-cAMP prevents this effect of KH7. KH7 inhibits accumulation in isolated rat-liver mitochondria and incorporation in complex I of "in vitro" produced, radiolabeled NDUFS4 and NDUFV2 subunits. 8-Br-cAMP prevents also this effect of KH7. Use of protease inhibitors shows that intramitochondrial cAMP exerts this positive effect on complex I by preventing digestion of nuclear-encoded subunits by mitochondrial protease(s), whose activity is promoted by KH7 and H89, an inhibitor of PKA.

    Topics: 8-Bromo Cyclic Adenosine Monophosphate; Adenylyl Cyclases; Cells, Cultured; Cyclic AMP; Electron Transport Complex I; Fibroblasts; Humans; Leupeptins; Mitochondria; Oligopeptides; Protease Inhibitors; Protein Subunits

2015
Tumor necrosis factor-α-induced apoptosis of gastric cancer MKN28 cells: accelerated degradation of the inhibitor of apoptosis family members.
    Archives of biochemistry and biophysics, 2015, Jan-15, Volume: 566

    The role of the inhibitor of apoptosis (IAP) family members in tumor necrosis factor-α (TNF-α)-induced apoptosis of human gastric cancer MKN28 cells was explored. TNF-α induced up-regulation of cIAP2, whereas cycloheximide (CHX) induced down-regulation of XIAP and survivin. Degradation of cIAP1 and XIAP, but not survivin, was accelerated by co-treatment of cells with TNF-α and CHX, and TNF-α-induced up-regulation of cIAP2 was inhibited by BMS-345541 (NF-κB inhibitor). Treatment of MKN28 cells with TNF-α plus CHX induced degradation of survivin and activation of caspase-8 and -3, followed by degradation of cIAP1 and XIAP and apoptosis. Proteasome inhibitors (MG132 and epoxomicin) suppressed TNF-α plus CHX-induced degradation of survivin, cIAP1, and XIAP as well as apoptosis. A caspase inhibitor (z-VAD-fmk) suppressed TNF-α plus CHX-induced apoptosis, but allowed degradation of survivin, cIAP1 and XIAP. TNF-α receptor 1 and 2 were expressed on MKN28 cells. The magnitude of apoptosis induced by TNF-α plus BMS-345541 was much less than that induced by TNF-α plus CHX. These findings suggest that TNF-α plus CHX-induced apoptosis of gastric cancer MKN28 cells may be caused by accelerated degradation of the IAP family members (survivin, cIAP1, and XIAP), in addition to inhibition of NF-κB-dependent synthesis of anti-apoptotic molecules.

    Topics: Amino Acid Chloromethyl Ketones; Apoptosis; Baculoviral IAP Repeat-Containing 3 Protein; Caspase 3; Caspase 8; Cell Line, Tumor; Cycloheximide; Gene Expression Regulation, Neoplastic; Humans; Imidazoles; Inhibitor of Apoptosis Proteins; Leupeptins; NF-kappa B; Oligopeptides; Proteolysis; Quinoxalines; Signal Transduction; Survivin; Tumor Necrosis Factor-alpha; Ubiquitin-Protein Ligases; X-Linked Inhibitor of Apoptosis Protein

2015
Superior antimitogenic and chemosensitization activities of the combination treatment of the histone deacetylase inhibitor apicidin and proteasome inhibitors on human colorectal cancer cells.
    International journal of oncology, 2014, Volume: 44, Issue:1

    Despite the effectiveness of histone deacetylase inhibitors, proteasome inhibitors and cytotoxic drugs on human cancers, none of these types of treatments by themselves has been sufficient to eradicate the disease. The combination of different modalities may hold enormous potential for eliciting therapeutic results. In the current study, we examined the effects of treatment with the histone deacetylase inhibitor (HDACI) apicidin (APC) in combination with proteasome inhibitors on human colorectal cancer cells. The molecular mechanisms of the combined treatments and their potential to sensitize colorectal cancer cells to chemotherapies were also investigated. Cancer cells were exposed to the agents alone and in combination, and cell growth inhibition was determined by MTT and colony formation assays. HDAC, proteasome and NF-κB activities as well as reactive oxygen species (ROS) were monitored. Cell cycle perturbation and induction of apoptosis were assessed by flow cytometry. The expression of cell cycle/apoptosis- and cytoprotective/stress-related genes was determined by quantitative PCR and EIA, respectively. The potentiation of cancer cell sensitivity to chemotherapies upon APC/PI combination treatment was also studied. The combination of APC and MG132, PI-1 or epoxomicin potently inhibited cancer cell growth, disrupted the cell cycle, induced apoptosis, decreased NF-κB activity and increased ROS production. These events were accompanied by the altered expression of genes associated with the cell cycle, apoptosis and cytoprotection/stress regulation. The combination treatment markedly enhanced the chemosensitivity of colorectal cancer cells (50-3.7 x 10(4)-fold) in a drug-, APC/PI combination- and colorectal cancer subtype-dependent manner. The results of this study have implications for the development of com-binatorial treatments that include HDACIs, PIs and conventional chemotherapeutic drugs, suggesting a potential therapeutic synergy with general applicability to various types of cancers.

    Topics: Antineoplastic Combined Chemotherapy Protocols; Apoptosis; Cell Line, Tumor; Cell Proliferation; Colorectal Neoplasms; Drug Synergism; Humans; Leupeptins; NF-kappa B; Oligopeptides; Peptides, Cyclic; Proteasome Endopeptidase Complex; Reactive Oxygen Species

2014
MCPIP1 contributes to the toxicity of proteasome inhibitor MG-132 in HeLa cells by the inhibition of NF-κB.
    Molecular and cellular biochemistry, 2014, Volume: 395, Issue:1-2

    Recently, we have shown that the treatment of cells with proteasome inhibitor MG-132 results in the induction of expression of monocyte chemotactic protein-1 induced protein 1 (MCPIP1). MCPIP1 is a ribonuclease, responsible for the degradation of transcripts encoding certain pro-inflammatory cytokines. The protein is also known as an inhibitor of NF-κB transcription factor. Thanks to its molecular properties, MCPIP1 is considered as a regulator of inflammation, differentiation, and survival. Using siRNA technology, we show here that MCPIP1 expression contributes to the toxic properties of MG-132 in HeLa cells. The inhibition of proteasome by MG-132 and epoxomicin markedly increased MCPIP1 expression. While MG-132 induces HeLa cell death, down-regulation of MCPIP1 expression by siRNA partially protects HeLa cells from MG-132 toxicity and restores Nuclear factor-κB (NF-κB) activity, inhibited by MG-132 treatment. Inversely, overexpression of MCPIP1 decreased constitutive activity of NF-κB and limited the survival of HeLa cells, as we have shown in the previous study. Interestingly, although MG-132 decreased the expression of IκBα and increased p65 phosphorylation, the inhibition of constitutive NF-κB activity was observed in MG-132-treated cells. Since the elevated constitutive activity of NF-κB is one of the mechanisms providing increased survival of cancer cells, including HeLa cells, we propose that death-promoting properties of MCPIP1 in MG-132-treated HeLa cells may, at least partially, derive from the negative effect on the constitutive NF-κB activity.

    Topics: Apoptosis; Cell Proliferation; Cysteine Proteinase Inhibitors; HeLa Cells; Hep G2 Cells; Humans; Leupeptins; NF-kappa B; Oligopeptides; Ribonucleases; Transcription Factors; Up-Regulation

2014
Proteasome-mediated processing of Nrf1 is essential for coordinate induction of all proteasome subunits and p97.
    Current biology : CB, 2014, Jul-21, Volume: 24, Issue:14

    Proteasome inhibitors are widely used in the treatment of multiple myeloma and as research tools. Additionally, diminished proteasome function may contribute to neuronal dysfunction. In response to these inhibitors, cells enhance the expression of proteasome subunits by the transcription factor Nrf1. Here, we investigate the mechanisms by which decreased proteasome function triggers production of new proteasomes via Nrf1.. Exposure of myeloma or neuronal cells to proteasome inhibitors (bortezomib, epoxomicin, and MG132), but not to proteotoxic or ER stress, caused a 2- to 4-fold increase within 4 hr in mRNAs for all 26S subunits. In addition, p97 and its cofactors (Npl4, Ufd1, and p47), PA200, and USP14 were induced, but expression of immunoproteasome-specific subunits was suppressed. Nrf1 mediates this induction of proteasomes and p97, but only upon exposure to low concentrations of inhibitors that partially inhibit proteolysis. Surprisingly, high concentrations of these inhibitors prevent this compensatory response. Nrf1 is normally ER-bound, and its release requires its deglycosylation and ubiquitination. Normally ubiquitinated Nrf1 is rapidly degraded, but when partially inhibited, proteasomes carry out limited proteolysis and release the processed Nrf1 (lacking its N-terminal region) from the ER, which allows it to enter the nucleus and promote gene expression.. When fully active, proteasomes degrade Nrf1, but when partially inhibited, they perform limited proteolysis that generates the active form of Nrf1. This elegant mechanism allows cells to compensate for reduced proteasome function by enhancing production of 26S subunits and p97.

    Topics: Boronic Acids; Bortezomib; Cell Line; Endoplasmic Reticulum; HEK293 Cells; Humans; Leupeptins; Nuclear Proteins; Nuclear Respiratory Factor 1; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Proteolysis; Pyrazines; RNA Interference; RNA, Small Interfering; Ubiquitin Thiolesterase; Ubiquitination; Unfolded Protein Response

2014
Nrf2- and ATF4-dependent upregulation of xCT modulates the sensitivity of T24 bladder carcinoma cells to proteasome inhibition.
    Molecular and cellular biology, 2014, Sep-15, Volume: 34, Issue:18

    The ubiquitin-proteasome pathway degrades ubiquitinated proteins to remove damaged or misfolded protein and thus plays an important role in the maintenance of many important cellular processes. Because the pathway is also crucial for tumor cell growth and survival, proteasome inhibition by specific inhibitors exhibits potent antitumor effects in many cancer cells. xCT, a subunit of the cystine antiporter system xc (-), plays an important role in cellular cysteine and glutathione homeostasis. Several recent reports have revealed that xCT is involved in cancer cell survival; however, it was unknown whether xCT affects the cytotoxic effects of proteasome inhibitors. In this study, we found that two stress-inducible transcription factors, Nrf2 and ATF4, were upregulated by proteasome inhibition and cooperatively enhance human xCT gene expression upon proteasome inhibition. In addition, we demonstrated that the knockdown of xCT by small interfering RNA (siRNA) or pharmacological inhibition of xCT by sulfasalazine (SASP) or (S)-4-carboxyphenylglycine (CPG) significantly increased the sensitivity of T24 cells to proteasome inhibition. These results suggest that the simultaneous inhibition of both the proteasome and xCT could have therapeutic benefits in the treatment of bladder tumors.

    Topics: Activating Transcription Factor 4; Amino Acid Transport System y+; Antineoplastic Agents; Boronic Acids; Bortezomib; Cell Line, Tumor; Cysteine; Gene Expression Regulation, Neoplastic; Gene Knockdown Techniques; Glutathione; Glycine; HEK293 Cells; HeLa Cells; Humans; Leupeptins; NF-E2-Related Factor 2; Oligopeptides; Proteasome Endopeptidase Complex; Pyrazines; RNA, Small Interfering; Sulfasalazine; Urinary Bladder Neoplasms

2014
Broad-spectrum antimalarial activity of peptido sulfonyl fluorides, a new class of proteasome inhibitors.
    Antimicrobial agents and chemotherapy, 2013, Volume: 57, Issue:8

    Despite declining numbers of cases and deaths, malaria remains a major public health problem in many parts of the world. Today, case management relies heavily on a single class of antimalarial compounds: artemisinins. Hence, development of resistance against artemisinins may destroy current malaria control strategies. Beyond malaria control are elimination and eradication programs that will require drugs with good activity against acute infection but also with preventive and transmission-blocking properties. Consequently, new antimalarials are needed not only to ensure malaria control but also for elimination and eradication efforts. In this study, we introduce peptido sulfonyl fluorides (PSF) as a new class of compounds with antiplasmodial activity. We show that PSF target the plasmodial proteasome and act on all asexual stages of the intraerythrocytic cycle and on gametocytes. PSF showed activities at concentrations as low as 20 nM against multidrug-resistant and chloroquine-sensitive Plasmodium falciparum laboratory strains and clinical isolates from Gabon. Structural requirements for activity were identified, and cytotoxicity in human HeLa or HEK 293 cells was low. The lead PSF PW28 suppressed growth of Plasmodium berghei in vivo but showed signs of toxicity in mice. Considering their modular structure and broad spectrum of activity against different stages of the plasmodial life cycle, proteasome inhibitors based on PSF have a great potential for further development as preclinical candidate compounds with improved species-specific activity and less toxicity.

    Topics: Animals; Antimalarials; Chloroquine; Drug Evaluation, Preclinical; Drug Resistance, Multiple; Female; HEK293 Cells; HeLa Cells; Humans; Leupeptins; Mice; Oligopeptides; Parasitic Sensitivity Tests; Plasmodium berghei; Plasmodium falciparum; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Schizonts; Sulfinic Acids

2013
The ubiquitin-proteasome system regulates the stability and activity of the glucose sensor glucokinase in pancreatic β-cells.
    The Biochemical journal, 2013, Dec-01, Volume: 456, Issue:2

    The ubiquitin-proteasome system is important to maintain pancreatic β-cell function. Inhibition of the proteasome significantly reduced glucose-induced insulin secretion. Key regulators of the stimulus/secretion cascade seem to be affected by protein misfolding if the proteasome is down-regulated as recently reported in humans with Type 2 diabetes. It remains unknown, however, whether the glucose sensor enzyme glucokinase is involved in this process. A direct interaction between glucokinase and ubiquitin could be shown in vivo by FRET, suggesting regulation of glucokinase by the proteasome. After proteasome inhibition glucokinase activity was significantly reduced in MIN6 cells, whereas the protein content was increased, indicating protein misfolding. Enhancing the availability of chaperones by cyclohexamide could induce refolding and restored glucokinase activity. Glucokinase aggregation due to proteasome blocking with MG132, bortezomib, epoxomicin or lactacystin could be detected in MIN6 cells, primary β-cells and hepatocytes using fluorescence-based assays. Glucokinase aggresome formation proceeded microtubule-assisted and was avoided by cyclohexamide. Thus the results of the present study provide support for glucokinase misfolding and aggregation in case of a diminished capacity of the ubiquitin-proteasome system in pancreatic β-cells. In the Type 2 diabetic situation this could contribute to reduced glucose-induced insulin secretion.

    Topics: Acetylcysteine; Animals; Chlorocebus aethiops; COS Cells; Cycloheximide; Glucokinase; Glucose; Hepatocytes; Humans; Insulin; Insulin Secretion; Insulin-Secreting Cells; Leupeptins; Mice; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Stability; Protein Synthesis Inhibitors; Proteolysis; Single-Cell Analysis; Ubiquitin; Ubiquitination

2013
Proteolytic pathways induced by herbicides that inhibit amino acid biosynthesis.
    PloS one, 2013, Volume: 8, Issue:9

    The herbicides glyphosate (Gly) and imazamox (Imx) inhibit the biosynthesis of aromatic and branched-chain amino acids, respectively. Although these herbicides inhibit different pathways, they have been reported to show several common physiological effects in their modes of action, such as increasing free amino acid contents and decreasing soluble protein contents. To investigate proteolytic activities upon treatment with Gly and Imx, pea plants grown in hydroponic culture were treated with Imx or Gly, and the proteolytic profile of the roots was evaluated through fluorogenic kinetic assays and activity-based protein profiling.. Several common changes in proteolytic activity were detected following Gly and Imx treatment. Both herbicides induced the ubiquitin-26 S proteasome system and papain-like cysteine proteases. In contrast, the activities of vacuolar processing enzymes, cysteine proteases and metacaspase 9 were reduced following treatment with both herbicides. Moreover, the activities of several putative serine protease were similarly increased or decreased following treatment with both herbicides. In contrast, an increase in YVADase activity was observed under Imx treatment versus a decrease under Gly treatment.. These results suggest that several proteolytic pathways are responsible for protein degradation upon herbicide treatment, although the specific role of each proteolytic activity remains to be determined.

    Topics: Amino Acids; Blotting, Western; Cysteine Proteinase Inhibitors; Glycine; Glyphosate; Herbicides; Hydroponics; Imidazoles; Leucine; Leupeptins; Oligopeptides; Pisum sativum; Plant Proteins; Plant Roots; Proteasome Endopeptidase Complex; Proteolysis; Signal Transduction; Time Factors

2013
The origin of proteasome-inhibitor resistant HLA class I peptidomes: a study with HLA-A*68:01.
    Molecular & cellular proteomics : MCP, 2012, Volume: 11, Issue:1

    Some HLA class I molecules bind a significant fraction of their constitutive peptidomes in the presence of proteasome inhibitors. In this study, A*68:01-bound peptides, and their parental proteins, were characterized through massive mass spectrometry sequencing to refine its binding motif, including the nearly exclusive preference for C-terminal basic residues. Stable isotope tagging was used to distinguish proteasome-inhibitor sensitive and resistant ligands. The latter accounted for less than 20% of the peptidome and, like in HLA-B27, arose predominantly from small and basic proteins. Under the conditions used for proteasome inhibition in vivo, epoxomicin and MG-132 incompletely inhibited the hydrolysis of fluorogenic substrates specific for the tryptic or for both the tryptic and chymotryptic subspecificities, respectively. This incomplete inhibition was also reflected in the cleavage of synthetic peptide precursors of A*68:01 ligands. For these substrates, the inhibition of the proteasome resulted in altered cleavage patterns. However these alterations did not upset the balance between cleavage at peptide bonds resulting in epitope destruction and those leading to their generation. The results indicate that inhibitor-resistant HLA class I ligands are not necessarily produced by non-proteasomal pathways. However, their generation is not simply explained by decreased epitope destruction upon incomplete proteasomal inhibition and may require additional proteolytic steps acting on incompletely processed proteasomal products.

    Topics: Amino Acid Sequence; Cell Line; Cysteine Proteinase Inhibitors; HLA-A Antigens; Humans; Leupeptins; Ligands; Oligopeptides; Peptides; Proteasome Endopeptidase Complex

2012
High sensitivity of embryonic stem cells to proteasome inhibitors correlates with low expression of heat shock protein and decrease of pluripotent cell marker expression.
    BMB reports, 2012, Volume: 45, Issue:5

    The ubiquitin-proteasome system is a major proteolytic system for nonlysosomal degradation of cellular proteins. Here, we investigated the response of mouse embryonic stem (ES) cells under proteotoxic stress. Proteasome inhibitors induced expression of heat shock protein 70 (HSP70) in a concentration- and time-dependent manner, and also induced apoptosis of ES cells. Importantly, more apoptotic cells were observed in ES cells compared with other somatic cells. To understand this phenomenon, we further investigated the expression of HSP70 and pluripotent cell markers. HSP70 expression was more significantly increased in somatic cells than in ES cells, and expression levels of pluripotent cell markers such as Oct4 and Nanog were decreased in ES cells. These results suggest that higher sensitivity of ES cells to proteotoxic stress may be related with lower capacity of HSP70 expression and decreased pluripotent cell marker expression, which is essential for the survival of ES cells.

    Topics: Acetylcysteine; Animals; Biomarkers; Cell Differentiation; Cells, Cultured; Cysteine Proteinase Inhibitors; Down-Regulation; Drug Resistance; Embryonic Stem Cells; Gene Expression Regulation, Developmental; HEK293 Cells; HSP70 Heat-Shock Proteins; Humans; Leupeptins; Mice; Oligopeptides; Pluripotent Stem Cells; Proteasome Inhibitors

2012
Recruitment and dynamics of proteasome association with rhTRIM5α cytoplasmic complexes during HIV-1 infection.
    Traffic (Copenhagen, Denmark), 2012, Volume: 13, Issue:9

    A variety of proteins have been identified that restrict infection by different viruses. One such restriction factor is the rhesus macaque variant of TRIM5α (rhTRIM5α), which potently blocks infection by HIV-1. The block to infection mediated by rhTRIM5α occurs early after entry into the host cell, generally prior to reverse transcription. However, proteasome inhibitors reveal an intermediate step of restriction in which virus can complete reverse transcription, but still fails to infect the cell. While proteasome inhibitors have been a useful tool in understanding how restriction takes place, the role of the proteasome itself during restriction has not yet been examined. Here, we characterize the interaction of rhTRIM5α and incoming virions with the proteasome. We show that proteasomes localize to rhTRIM5α cytoplasmic bodies, and this localization is more evident when the activity of the proteasome is inhibited pharmacologically. We also show that restricted virus associates with complexes of proteasomes and rhTRIM5α, suggesting that rhTRIM5α utilizes the proteasome during restriction. Finally, live cell imaging experiments reveal that virus associates with proteasomes, and proteasome inhibition affects the duration of association. Taken together, these studies implicate the proteasome as playing a functional role during rhTRIM5α restriction of incoming virions.

    Topics: Animals; Cells, Cultured; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cytoplasm; HeLa Cells; HIV-1; Humans; Leupeptins; Macaca mulatta; Oligopeptides; Proteasome Endopeptidase Complex; Proteins; Ubiquitin-Protein Ligases; Virion

2012
Proteasome inhibition induces developmentally deregulated programs of apoptotic and autophagic cell death during Drosophila melanogaster oogenesis.
    Cell biology international, 2011, Volume: 35, Issue:1

    Ubiquitin/proteasome-mediated degradation of eukaryotic proteins is critically implicated in a number of signalling pathways and cellular processes. To specifically impair proteasome activities, in vitro developing Drosophila melanogaster egg chambers were exposed to the MG132 or epoxomicin proteasome inhibitors, while a GAL4/UAS binary genetic system was employed to generate double transgenic flies overexpressing β2 and β6 conditional mutant proteasome subunits in a cell type-specific manner. MG132 and epoxomicin administration resulted in severe deregulation of in vitro developing egg chambers, which was tightly associated with precocious induction of nurse cell-specific apoptotic and autophagic death programmes, featured by actin cytoskeleton disorganization, nuclear chromatin condensation, DRICE caspase activation and autophagosome accumulation. In vivo targeted overexpression of β2 and β6 conditional mutants, specifically in the nurse cell compartment, led to a notable up-regulation of sporadic apoptosis potency during early and mid-oogenesis 'checkpoints', thus reasonably justifying the observed reduction in eclosion efficiency. Furthermore, in response to the intracellular abundance of β2 and β6 conditional mutant forms, specifically in numerous tissues of third instar larval stage, the developmental course was arrested, and lethal phenotypes were obtained at this particular embryonic period, with the double transgenic heterozygote embryos being unable to further proceed to complete maturation to adult flies. Our data demonstrate that physiological proteasome function is required to ensure normal oogenesis and embryogenesis in D. melanogaster, since targeted and cell type-dependent proteasome inactivation initiates developmentally deregulated apoptotic and autophagic mechanisms.

    Topics: Animals; Animals, Genetically Modified; Apoptosis; Autophagy; Cysteine Proteinase Inhibitors; Drosophila melanogaster; Larva; Leupeptins; Mutation; Oligopeptides; Oogenesis; Proteasome Endopeptidase Complex; Proteasome Inhibitors

2011
Semi-high throughput method of measuring proteasome inhibition in vitro and in cultured cells.
    Cell biology and toxicology, 2011, Volume: 27, Issue:2

    The ubiquitin proteasome-proteolytic pathway has emerged as one of the most significant pathways in modulating protein homeostasis under both normal and disease states. The use of proteasome inhibitors (PI) has played a pivotal role in understanding protein turn over. The main objective of this work was to develop a comprehensive, fast, and reliable, yet simple in vitro assay that would allow for the identification and characterization of a wide range of PIs. The assays consist of a 96-well plate high throughput (HTP) method to assess proteasome activity in Hs578T breast cancer cell extracts, purified 20S proteasome, using a fluorogenic substrate, Suc-leu-leu-val-tyr-7-AMC, specific to the chymotrypsin-like enzymatic activity of the proteasome. We showed that the chymotrypsin-like activity of the proteasome was inhibited in the two in vitro systems, albeit to different degrees. The assay system also includes two cell-based assays consisting of a vector expressing a fusion protein of green fluorescent protein (gfp) and Mouse Ornithine Decarboxylase (MODC) in Zs578T (parental Hs578T carrying the vector that expresses the fusion protein). In the cell-based assay analyses (qualitatively by microscopy and quantitatively by flow cytometry), treatment of Zs578T with PIs prevented the degradation of MODC, accumulated gfp, indicative of increased proteasome inhibition. Because no single assay represents a definitive proof of proteasome inhibitory activity, combined, these assays should serve as a comprehensive benchmark for the identification and partial characterization of novel inhibitors. In summary, the four-step assay protocol can easily be adapted into a high throughput format to rapidly screen unknown inhibitors.

    Topics: Acetylcysteine; Animals; Biological Assay; Boronic Acids; Bortezomib; Cell Extracts; Cells, Cultured; Chymotrypsin; High-Throughput Screening Assays; Inhibitory Concentration 50; Leupeptins; Mice; Models, Biological; Oligopeptides; Ornithine Decarboxylase; Protease Inhibitors; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Pyrazines; Recombinant Fusion Proteins; Time Factors

2011
Proteasome inhibitors induce the presentation of an Epstein-Barr virus nuclear antigen 1-derived cytotoxic T lymphocyte epitope in Burkitt's lymphoma cells.
    Immunology, 2011, Volume: 133, Issue:1

    The Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1) is generally expressed in all EBV-associated tumours and is therefore an interesting target for immunotherapy. However, evidence for the recognition and elimination of EBV-transformed and Burkitt's lymphoma (BL) cells by cytotoxic T lymphocytes (CTLs) specific for endogenously presented EBNA1-derived epitopes remains elusive. We confirm here that CTLs specific for the HLA-B35/B53-presented EBNA1-derived HPVGEADYFEY (HPV) epitope are detectable in the majority of HLA-B35 individuals, and recognize EBV-transformed B lymphocytes, thereby demonstrating that the GAr domain does not fully inhibit the class I presentation of the HPV epitope. In contrast, BL cells are not recognized by HPV-specific CTLs, suggesting that other mechanisms contribute to providing a full protection from EBNA1-specific CTL-mediated lysis. One of the major differences between BL cells and lymphoplastoid cell lines (LCLs) is the proteasome; indeed, proteasomes from BL cells demonstrate far lower chymotryptic and tryptic-like activities compared with proteasomes from LCLs. Hence, inefficient proteasomal processing is likely to be the main reason for the poor presentation of this epitope in BL cells. Interestingly, we show that treatments with proteasome inhibitors partially restore the capacity of BL cells to present the HPV epitope. This indicates that proteasomes from BL cells, although less efficient in degrading reference substrates than proteasomes from LCLs, are able to destroy the HPV epitope, which can, however, be generated and presented after partial inhibition of the proteasome. These findings suggest the use of proteasome inhibitors, alone or in combination with other drugs, as a strategy for the treatment of EBNA1-carrying tumours.

    Topics: Antigen Presentation; Blotting, Western; Boronic Acids; Bortezomib; Burkitt Lymphoma; Cell Line; Epitopes, T-Lymphocyte; Epstein-Barr Virus Nuclear Antigens; Fluorescent Antibody Technique; Humans; Leupeptins; Oligopeptides; Protease Inhibitors; Proteasome Endopeptidase Complex; Pyrazines; T-Lymphocytes, Cytotoxic

2011
Mutations in PINK1 and Parkin impair ubiquitination of Mitofusins in human fibroblasts.
    PloS one, 2011, Mar-08, Volume: 6, Issue:3

    PINK1 and Parkin mutations cause recessive Parkinson's disease (PD). In Drosophila and SH-SY5Y cells, Parkin is recruited by PINK1 to damaged mitochondria, where it ubiquitinates Mitofusins and consequently promotes mitochondrial fission and mitophagy.Here, we investigated the impact of mutations in endogenous PINK1 and Parkin on the ubiquitination of mitochondrial fusion and fission factors and the mitochondrial network structure. Treating control fibroblasts with mitochondrial membrane potential (Δψ) inhibitors or H(2)O(2) resulted in ubiquitination of Mfn1/2 but not of OPA1 or Fis1. Ubiquitination of Mitofusins through the PINK1/Parkin pathway was observed within 1 h of treatment. Upon combined inhibition of Δψ and the ubiquitin proteasome system (UPS), no ubiquitination of Mitofusins was detected. Regarding morphological changes, we observed a trend towards increased mitochondrial branching in PD patient cells upon mitochondrial stress.For the first time in PD patient-derived cells, we demonstrate that mutations in PINK1 and Parkin impair ubiquitination of Mitofusins. In the presence of UPS inhibitors, ubiquitinated Mitofusin is deubiquitinated by the UPS but not degraded, suggesting that the UPS is involved in Mitofusin degradation.

    Topics: Carbonyl Cyanide m-Chlorophenyl Hydrazone; Fibroblasts; GTP Phosphohydrolases; Humans; Hydrogen Peroxide; Leupeptins; Macrolides; Membrane Proteins; Membrane Transport Proteins; Mitochondria; Mitochondrial Membrane Transport Proteins; Mitochondrial Proteins; Models, Biological; Mutation; Oligopeptides; Proteasome Endopeptidase Complex; Protein Kinases; Protein Transport; Ubiquitin; Ubiquitin-Protein Ligases; Ubiquitination; Valinomycin

2011
Leishmania donovani: proteasome-mediated down-regulation of methionine adenosyltransferase.
    Parasitology, 2011, Volume: 138, Issue:9

    Methionine adenosyltransferase (MAT) is an important enzyme for metabolic processes, to the extent that its product, S-adenosylmethionine (AdoMet), plays a key role in trans-methylation, trans-sulphuration and polyamine synthesis. Previous studies have shown that a MAT-overexpressing strain of Leishmania donovani controls AdoMet production, keeping the intracellular AdoMet concentration at levels that are compatible with cell survival. This unexpected result, together with the fact that MAT activity and abundance changed with time in culture, suggests that different regulatory mechanisms acting beyond the post-transcriptional level are controlling this protein. In order to gain an insight into these mechanisms, several experiments were carried out to explain the MAT abundance during promastigote cell growth. Determination of MAT turnover in cycloheximide (CHX)-treated cultures resulted in a surprising 5-fold increase in MAT turnover compared to CHX-untreated cultures. This increase agrees with a stabilization of the MAT protein, whose integrity was maintained during culture. The presence of proteasome inhibitors, namely MG-132, MG-115, epoxomycin and lactacystin in the culture medium prevented MAT degradation in both MAT-overexpressing and 'mock-transfected' leishmanial strains. The role of the ubiquitin (Ub) pathway in MAT down-regulation was supported using immunoprecipitation experiments. Immunoprecipitated MAT cross-reacted with anti-Ub antibodies, which provides evidence of a proteasome-mediated down-regulation of the leishmanial MAT abundance.

    Topics: Acetylcysteine; Cell Culture Techniques; Cloning, Molecular; Cycloheximide; Down-Regulation; Electrophoresis, Polyacrylamide Gel; Gene Expression; Immunoprecipitation; Kinetics; Leishmania donovani; Leishmaniasis, Visceral; Leupeptins; Methionine Adenosyltransferase; Oligopeptides; Plasmids; Protease Inhibitors; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Processing, Post-Translational; Recombinant Proteins; S-Adenosylmethionine; Transfection; Ubiquitin

2011
Relative contribution of different l-arginine sources to the substrate supply of endothelial nitric oxide synthase.
    Journal of molecular and cellular cardiology, 2011, Volume: 51, Issue:5

    In certain cases of endothelial dysfunction l-arginine becomes rate-limiting for NO synthesis in spite of sufficiently high plasma concentrations of the amino acid. To better understand this phenomenon, we investigated routes of substrate supply to endothelial nitric oxide synthase (eNOS). Our previous data with human umbilical vein (HUVEC) and EA.hy.926 endothelial cells demonstrated that eNOS can obtain its substrate from the conversion of l-citrulline to l-arginine and from protein breakdown. In the present study, we determined the quantitative contribution of proteasomal and lysosomal protein degradation and investigated to what extent extracellular peptides and l-citrulline can provide substrate to eNOS. The RFL-6 reporter cell assay was used to measure eNOS activity in human EA.hy926 endothelial cells. Individual proteasome and lysosome inhibition reduced eNOS activity in EA.hy926 cells only slightly. However, the combined inhibition had a pronounced reducing effect. eNOS activity was fully restored by supplementing either l-citrulline or l-arginine-containing dipeptides. Histidine prevented the restoration of eNOS activity by the dipeptide, suggesting that a transporter accepting both, peptides and histidine, mediates the uptake of the extracellular peptide. In fact, the peptide and histidine transporter PHT1 was expressed in EA.hy926 cells and HUVECs (qRT/PCR). Our study thus demonstrates that l-citrulline and l-arginine-containing peptides derived from either intracellular protein breakdown or from the extracellular space seem to be good substrate sources for eNOS.

    Topics: Arginine; Atherosclerosis; Biological Transport; Cell Line; Chloroquine; Chromatography, High Pressure Liquid; Citrulline; Dipeptides; Endothelium, Vascular; Genes, Reporter; Histidine; Human Umbilical Vein Endothelial Cells; Humans; Leupeptins; Lysosomes; Membrane Transport Proteins; Nitric Oxide; Nitric Oxide Synthase Type III; Oligopeptides; Protease Inhibitors; Proteasome Endopeptidase Complex; Proteolysis

2011
The cell senescence inducing gene product MORF4 is regulated by degradation via the ubiquitin/proteasome pathway.
    Experimental cell research, 2010, Jan-01, Volume: 316, Issue:1

    After undergoing several rounds of divisions normal human fibroblasts enter a terminally non-dividing state referred to as cellular or replicative senescence. We cloned MORF4 (mortality factor on human chromosome 4), as a cellular senescence inducing gene that caused immortal cells assigned to complementation group B for indefinite division to stop dividing. To facilitate analyses of this gene, which is toxic to cells at low levels, we obtained stable clones of HeLa cells expressing a tetracycline-induced MORF4 construct that could be induced by doxycycline in a dose-dependent manner. MORF4 induction resulted in reduced colony formation after 14 days of culture, as previously observed. We determined that MORF4 protein was unstable and that addition of the proteasome inhibitor MG132 resulted in the accumulation of the protein. Following removal of MG132 the protein was rapidly degraded. Subcellular fractionation following MG132 treatment demonstrated that the protein accumulates primarily in the cytoplasm with some amounts present in the nucleus. It is therefore possible that MORF4 protein, which escapes degradation in the cytoplasm, is transported to the nucleus where it is functional. The results suggest that levels of MORF4 in cells must be tightly controlled and one mechanism involves stability of the protein.

    Topics: Benzoates; Cell Nucleus; Cell Proliferation; Cycloheximide; Cysteine Proteinase Inhibitors; Cytoplasm; Doxycycline; Enzyme Inhibitors; Furans; Gene Expression Regulation; HeLa Cells; Humans; Leupeptins; Oligopeptides; Peptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Processing, Post-Translational; Pyrazoles; Recombinant Fusion Proteins; Transcription Factors; Transfection; Ubiquitin; Ubiquitin-Activating Enzymes

2010
Zinc mesoporphyrin induces rapid proteasomal degradation of hepatitis C nonstructural 5A protein in human hepatoma cells.
    Gastroenterology, 2010, Volume: 138, Issue:5

    The nonstructural 5A (NS5A) protein of hepatitis C virus (HCV) plays a critical role in HCV replication and is an attractive target for the therapy of HCV infection. So far, little is known about the posttranslational regulation of NS5A protein and its precise role in HCV RNA replication. Our objectives were to elucidate the down-regulation of NS5A protein and HCV RNA replication by zinc mesoporphyrin (ZnMP) and the mechanism by which this process occurs.. Human hepatoma cells expressing HCV proteins were used to investigate the posttranslational regulation of ZnMP on NS5A protein by Western blots and immunoprecipitation. Real-time quantitative reverse transcription polymerase chain reaction was used to determine the effects of ZnMP on HCV RNA replication.. ZnMP selectively and markedly down-regulated NS5A protein levels by increasing degradation of NS5A protein (half-life fell from 18.7 hours to 2.7 hours). The proteasome inhibitors epoxomicin and MG132 significantly abrogated degradation of NS5A protein by ZnMP without affecting levels of NS5A in the absence of ZnMP. Analysis of immunoprecipitates with an antiubiquitin antibody revealed polyubiquitination of NS5A, suggesting that ZnMP induces ubiquitination of NS5A protein. In addition, 10 micromol/L of ZnMP reduced HCV replication by approximately 63% in the Con1 replicon cells, approximately 70% in J6/Japanese fulminant hepatitis 1 HCV-transfected cells, and approximately 90% in J6/Japanese fulminant hepatitis 1 HCV-infected cells without affecting cell viability.. ZnMP produces a rapid and profound down-regulation of the NS5A protein by enhancing its polyubiquitination and proteasome-dependent catabolism. ZnMP may hold promise as a novel agent to treat HCV infection.

    Topics: Antiviral Agents; Carcinoma, Hepatocellular; Cell Line, Tumor; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Down-Regulation; Half-Life; Hepacivirus; Humans; Leupeptins; Liver Neoplasms; Metalloporphyrins; Oligopeptides; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; Protein Stability; RNA, Viral; Time Factors; Transfection; Ubiquitination; Viral Nonstructural Proteins; Virus Replication

2010
Induced, selective proteolysis of MLK3 negatively regulates MLK3/JNK signalling.
    The Biochemical journal, 2010, Apr-14, Volume: 427, Issue:3

    MLK3 (mixed lineage kinase 3) is a MAP3K [MAPK (mitogen-activated protein kinase) kinase kinase] that activates multiple MAPK pathways, including the JNK (c-Jun N-terminal kinase) pathway. Immunoblotting of lysates from cells ectopically expressing active MLK3 revealed an additional immunoreactive band corresponding to a CTF (C-terminal fragment) of MLK3. In the present paper we provide evidence that MLK3 undergoes proteolysis to generate a stable CTF in response to different stimuli, including PMA and TNFalpha (tumour necrosis factor alpha). The cleavage site was deduced by Edman sequencing as between Gln251 and Pro252, which is within the kinase domain of MLK3. Based on our homology model of the kinase domain of MLK3, the region containing the cleavage site is predicted to reside on a flexible solvent-accessible loop. Site-directed mutagenesis studies revealed that Leu250 and Gln251 are required for recognition by the 'MLK3 protease', reminiscent of the substrate specificity of the coronavirus 3C and 3CL proteases. Whereas numerous mammalian protease inhibitors have no effect on MLK3 proteolysis, blockade of the proteasome through epoxomicin or MG132 abolishes PMA-induced production of the CTF of MLK3. This CTF is able to heterodimerize with full-length MLK3, and interact with the active form of the small GTPase Cdc42, resulting in diminished activation loop phosphorylation of MLK3 and reduced signalling to JNK. Thus this novel proteolytic processing of MLK3 may negatively control MLK3 signalling to JNK.

    Topics: Blotting, Western; Cell Line; Cysteine Proteinase Inhibitors; Electrophoresis, Polyacrylamide Gel; Humans; Immunoprecipitation; JNK Mitogen-Activated Protein Kinases; Leupeptins; MAP Kinase Kinase Kinases; Mitogen-Activated Protein Kinase Kinase Kinase 11; Mutagenesis, Site-Directed; Oligopeptides; Phorbol Esters; Proteasome Endopeptidase Complex; Signal Transduction

2010
The ubiquitin-proteasome system plays an important role during various stages of the coronavirus infection cycle.
    Journal of virology, 2010, Volume: 84, Issue:15

    The ubiquitin-proteasome system (UPS) is a key player in regulating the intracellular sorting and degradation of proteins. In this study we investigated the role of the UPS in different steps of the coronavirus (CoV) infection cycle. Inhibition of the proteasome by different chemical compounds (i.e., MG132, epoxomicin, and Velcade) appeared to not only impair entry but also RNA synthesis and subsequent protein expression of different CoVs (i.e., mouse hepatitis virus [MHV], feline infectious peritonitis virus, and severe acute respiratory syndrome CoV). MHV assembly and release were, however, not appreciably affected by these compounds. The inhibitory effect on CoV protein expression did not appear to result from a general inhibition of translation due to induction of a cellular stress response by the inhibitors. Stress-induced phosphorylation of eukaryotic translation initiation factor 2alpha (eIF2alpha) generally results in impaired initiation of protein synthesis, but the sensitivity of MHV infection to proteasome inhibitors was unchanged in cells lacking a phosphorylatable eIF2alpha. MHV infection was affected not only by inhibition of the proteasome but also by interfering with protein ubiquitination. Viral protein expression was reduced in cells expressing a temperature-sensitive ubiquitin-activating enzyme E1 at the restrictive temperature, as well as in cells in which ubiquitin was depleted by using small interfering RNAs. Under these conditions, the susceptibility of the cells to virus infection was, however, not affected, excluding an important role of ubiquitination in virus entry. Our observations reveal an important role of the UPS in multiple steps of the CoV infection cycle and identify the UPS as a potential drug target to modulate the impact of CoV infection.

    Topics: Animals; Boronic Acids; Bortezomib; Cats; Cell Line; Chlorocebus aethiops; Coronavirus Infections; Coronavirus, Feline; Leupeptins; Mice; Murine hepatitis virus; Oligopeptides; Protease Inhibitors; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Pyrazines; Severe acute respiratory syndrome-related coronavirus; Ubiquitin; Virus Internalization; Virus Release; Virus Replication

2010
Effect of endogenous mutant and wild-type PINK1 on Parkin in fibroblasts from Parkinson disease patients.
    Human molecular genetics, 2010, Aug-15, Volume: 19, Issue:16

    Mutations in the PTEN-induced putative kinase 1 (PINK1), a mitochondrial serine-threonine kinase, and Parkin, an E3 ubiquitin ligase, are associated with autosomal-recessive forms of Parkinson disease (PD). Both are involved in the maintenance of mitochondrial integrity and protection from multiple stressors. Recently, Parkin was demonstrated to be recruited to impaired mitochondria in a PINK1-dependent manner, where it triggers mitophagy. Using primary human dermal fibroblasts originating from PD patients with various PINK1 mutations, we showed at the endogenous level that (i) PINK1 regulates the stress-induced decrease of endogenous Parkin; (ii) mitochondrially localized PINK1 mediates the stress-induced mitochondrial translocation of Parkin; (iii) endogenous PINK1 is stabilized on depolarized mitochondria; and (iv) mitochondrial accumulation of full-length PINK1 is sufficient but not necessary for the stress-induced loss of Parkin signal and its mitochondrial translocation. Furthermore, we showed that different stressors, depolarizing or non-depolarizing, led to the same effect on detectable Parkin levels and its mitochondrial targeting. Although this effect on Parkin was independent of the mitochondrial membrane potential, we demonstrate a differential effect of depolarizing versus non-depolarizing stressors on endogenous levels of PINK1. Our study shows the necessity to introduce an environmental factor, i.e. stress, to visualize the differences in the interaction of PINK1 and Parkin in mutants versus controls. Establishing human fibroblasts as a suitable model for studying this interaction, we extend data from animal and other cellular models and provide experimental evidence for the generally held notion of PD as a condition with a combined genetic and environmental etiology.

    Topics: Blotting, Western; Cell Line; Cells, Cultured; Cysteine Proteinase Inhibitors; Cytosol; Fibroblasts; Humans; Hydrogen Peroxide; Ionophores; Leupeptins; Mitochondria; Mutation; Oligopeptides; Oxidants; Parkinson Disease; Protein Kinases; Protein Transport; RNA Interference; Transfection; Ubiquitin-Protein Ligases; Valinomycin

2010
Proteasomal inhibition reduces parkin mRNA in PC12 and SH-SY5Y cells.
    Parkinsonism & related disorders, 2009, Volume: 15, Issue:3

    Mutations in the gene encoding the E3 ubiquitin-protein ligase parkin have been shown to be a common genetic cause of familial early-onset Parkinson's disease (PD). In addition to its function in the ubiquitin-proteasome system (UPS), parkin has been ascribed general neuroprotective properties. Stress and mutation induced decreases in parkin solubility leading to compromised cytoprotection have recently been reported. We systematically investigated whether PD-related stresses including MG132 and epoxomicin (proteasomal impairment), tunicamycin (unfolded protein stress), and rotenone (mitochondrial dysfunction) resulted in expressional changes of parkin and other E3 ubiquitin ligases (dorfin, SIAH-1). Rotenone and tunicamycin did not change parkin mRNA levels, whereas proteasomal inhibition resulted in a reduction of parkin mRNA in PC12 cells as well as in SH-SY5Y cells. Therefore, surprisingly, cells did not react with a compensatory parkin upregulation under proteasomal inhibition, although, in parallel, parkin protein shifted to the insoluble fraction, reducing soluble parkin levels in the cytosol. Since the mRNA of the parkin-coregulated gene PACRG paralleled the parkin mRNA at least partly, we suspect a promoter-driven mechanism. Our study, therefore, shows a link between proteasomal impairment and parkin expression levels in cell culture, which is intriguing in the context of the described and debated proteasomal dysfunction in the substantia nigra of PD patients.

    Topics: Animals; Cell Line, Tumor; Cysteine Proteinase Inhibitors; Enzyme Inhibitors; Gene Expression Regulation; Humans; Leupeptins; Neuroblastoma; Nuclear Proteins; Oligopeptides; PC12 Cells; Proteasome Inhibitors; Rats; RNA, Messenger; Rotenone; Time Factors; Tunicamycin; Ubiquitin-Protein Ligases

2009
Inhibition of the ubiquitin-proteasome system prevents vaccinia virus DNA replication and expression of intermediate and late genes.
    Journal of virology, 2009, Volume: 83, Issue:6

    The ubiquitin-proteasome system has a central role in the degradation of intracellular proteins and regulates a variety of functions. Viruses belonging to several different families utilize or modulate the system for their advantage. Here we showed that the proteasome inhibitors MG132 and epoxomicin blocked a postentry step in vaccinia virus (VACV) replication. When proteasome inhibitors were added after virus attachment, early gene expression was prolonged and the expression of intermediate and late genes was almost undetectable. By varying the time of the removal and addition of MG132, the adverse effect of the proteasome inhibitors was narrowly focused on events occurring 2 to 4 h after infection, the time of the onset of viral DNA synthesis. Further analyses confirmed that genome replication was inhibited by both MG132 and epoxomicin, which would account for the effect on intermediate and late gene expression. The virus-induced replication of a transfected plasmid was also inhibited, indicating that the block was not at the step of viral DNA uncoating. UBEI-41, an inhibitor of the ubiquitin-activating enzyme E1, also prevented late gene expression, supporting the role of the ubiquitin-proteasome system in VACV replication. Neither the overexpression of ubiquitin nor the addition of an autophagy inhibitor was able to counter the inhibitory effects of MG132. Further studies of the role of the ubiquitin-proteasome system for VACV replication may provide new insights into virus-host interactions and suggest potential antipoxviral drugs.

    Topics: Cell Line; DNA Replication; Enzyme Inhibitors; Host-Pathogen Interactions; Humans; Leupeptins; Oligopeptides; Proteasome Inhibitors; Ubiquitin; Vaccinia virus; Virus Replication

2009
Participation of the human sperm proteasome in the capacitation process and its regulation by protein kinase A and tyrosine kinase.
    Biology of reproduction, 2009, Volume: 80, Issue:5

    The proteasome is a multicatalytic cellular complex present in human sperm that plays a significant role during several steps of mammalian fertilization. Here, we present evidence that the proteasome is involved in human sperm capacitation. Aliquots of highly motile sperm were incubated with proteasome inhibitors MG132 or epoxomicin. The percentage of capacitated sperm, the chymotrypsin-like activity of the proteasome, cAMP content, and the pattern of protein phosphorylation were assayed by using the chlortetracycline hydrochloride assay, a fluorogenic substrate, the cAMP enzyme immunoassay kit, and Western blot analysis, respectively. Our results indicate that treatment of sperm with proteasome inhibitors blocks the capacitation process, does not alter cAMP concentration, and changes the pattern of protein phosphorylation. To elucidate how proteasome activity is regulated during capacitation, sperm were incubated with: 1) tyrosine kinase (TK) inhibitors (genistein or herbimycin A); 2) protein kinase (PK) A inhibitors or activators (H89 and Rp-cAMPS, and 8-Br-cAMP, respectively); or 3) PKC inhibitors (tamoxifen or staurosporin) at different capacitation times. The chymotrypsin-like activity and degree of phosphorylation of the proteasome were then assayed. The results indicate that sperm treatment with TK and PKA inhibitors significantly decreases the chymotrypsin-like activity of the proteasome during capacitation. Immunoprecipitation and Western blot results suggest that the proteasome is phosphorylated during capacitation in a TK- and PKA-dependent pathway. In conclusion, we suggest that the sperm proteasome participates in the capacitation process, and that its activity is modulated by PKs.

    Topics: 8-Bromo Cyclic Adenosine Monophosphate; Benzoquinones; Cyclic AMP; Cyclic AMP-Dependent Protein Kinases; Enzyme Activation; Enzyme Inhibitors; Genistein; Humans; In Vitro Techniques; Isoquinolines; Lactams, Macrocyclic; Leupeptins; Male; Oligopeptides; Phosphorylation; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein-Tyrosine Kinases; Rifabutin; Sperm Capacitation; Spermatozoa; Sulfonamides; Thionucleotides

2009
Proteasome inhibitor differentially regulates expression of the major immediate early genes of human cytomegalovirus in human central nervous system-derived cell lines.
    Virus research, 2009, Volume: 142, Issue:1-2

    Proteasome inhibitor, which inhibits NF-kappaB activation, has been reported to activate c-Jun N-terminal kinase (JNK)-c-Jun pathway. In this study, we investigated the effects of proteasome inhibitor on the human cytomegalovirus (HCMV) major immediate early (MIE) gene expression in human central nervous system (CNS)-derived cell lines. Treatment of HCMV-infected 118MGC glioma and U373-MG astrocytoma cells with three proteasome inhibitors, MG132, clasto-lactacystin beta-lactone, and epoxomicin, suppressed MIE protein expression. In contrast, in HCMV-infected IMR-32 neuroblastoma cells, the proteasome inhibitors increased MIE protein expression, even in the presence of NF-kappaB inhibitor SN-50. A luciferase reporter assay demonstrated that MG132 markedly elevated the MIE promoter/enhancer (MIEP) activity in IMR-32 cells, but down-regulated it in 118MGC and U373-MG cells. Mutation in five cAMP response elements (CREs) within the MIEP resulted in a loss of the ability to respond to MG132 in IMR-32 cells. Moreover, Western blotting analysis revealed that MG132 induced c-Jun phosphorylation in all three CNS-derived cell lines, whereas a high level of activating transcription factor-2 (ATF-2) phosphorylation was observed only in IMR-32 cells. Finally, MG132-induced MIE protein expression was suppressed by JNK inhibitor that reduced the phosphorylation levels of both c-Jun and ATF-2. Taken together, these results suggest that the proteasome inhibitors activate CRE binding proteins consisting of c-Jun and ATF-2 through activating the JNK-c-Jun pathway, thereby inducing MIE protein synthesis in IMR-32 cells under the condition where NF-kappaB activity is inhibited.

    Topics: Activating Transcription Factor 2; Cell Line; Central Nervous System; Cytomegalovirus; Cytomegalovirus Infections; Gene Expression Regulation, Viral; Humans; Immediate-Early Proteins; Lactones; Leupeptins; Oligopeptides; Protease Inhibitors; Proteasome Inhibitors; Proto-Oncogene Proteins c-jun

2009
Perturbation of DNA repair pathways by proteasome inhibitors corresponds to enhanced chemosensitivity of cells to DNA damage-inducing agents.
    Cancer chemotherapy and pharmacology, 2009, Volume: 64, Issue:5

    Breast cancer treatment often employs DNA double-strand breaks (DSBs), such as that induced by irradiation or anticancer agents. Ubiquitination is required at the site of DNA damage and plays a crucial role in the DSB repair pathway. We investigated the effect of proteasome inhibitors on the pathway after exposure to chemotherapeutic agents and examined its correlation with cytotoxicity.. Cells were exposed for 1 h to DNA damage-inducing chemotherapeutic agents. After DNA damage, nuclear foci formation of conjugated ubiquitin (Ub-foci) and cell viability were examined in the absence or presence of proteasome inhibitors MG132 and epoxomicin.. Proteasome inhibitors trapped conjugated ubiquitin in the cytosol and blocked irinotecan (CPT-11)- and epirubicin-induced Ub-foci formation in MCF10A cells and HeLa cells, but not in MCF7 cells. MG132 sensitized MCF10A cells to CPT-11 and epirubicin treatment, demonstrating a synergistic effect. This synergistic effect is likely due to the failure to repair DNA, because a significant rise in unrepaired DNA damage was observed in the cells treated with MG132. On the other hand, no synergy was observed in MCF7 cells or when MG132 was combined with docetaxel.. The synergistic effect of proteasome inhibitors in combination with DNA damage-inducing agents warrants further investigating into its effectiveness in the treatment of breast cancer.

    Topics: Antibiotics, Antineoplastic; Antineoplastic Agents; Camptothecin; Cell Line, Tumor; Cell Nucleus; Cell Survival; Comet Assay; DNA Damage; DNA Repair; Epirubicin; Female; HeLa Cells; Humans; Immunohistochemistry; Irinotecan; Leupeptins; Oligopeptides; Proteasome Inhibitors; Ubiquitin

2009
Transcriptional and posttranslational regulation of clusterin by the two main cellular proteolytic pathways.
    Free radical biology & medicine, 2009, May-01, Volume: 46, Issue:9

    Clusterin/apolipoprotein J (CLU) is a secreted glycoprotein associated with many severe physiological disturbances that represent states of increased oxidative stress, such as aging, cancer, atherosclerosis, diabetes, and renal and neurodegenerative diseases. The aim of our work was to examine the effect of proteasome and lysosome inhibition on CLU expression and to determine whether those proteolytic pathways are implicated in CLU gene regulation and protein degradation. To this end we used two different model systems, namely the U-2 OS osteosarcoma cell line and the WI38 primary human embryonic lung fibroblasts. We report that proteasome inhibition promotes both heat-shock factor 1 (HSF-1)-dependent CLU gene expression induction and protein accumulation due to reduced degradation. In contrast, lysosome inhibition results in elevated levels of CLU protein but does not affect the CLU mRNA levels. We also provide direct evidence that both the intracellular precursor, psCLU, and the mature secreted, sCLU, isoforms constitute proteolytic substrates of the proteasome and the lysosome. Overall our findings indicate that CLU overexpression after proteasome inhibition relates to both positive gene transcriptional regulation by HSF-1 and posttranslational protein accumulation due to reduced proteasomal and lysosomal degradation.

    Topics: Antibodies, Blocking; Cell Line, Tumor; Clusterin; DNA-Binding Proteins; Fibroblasts; Gene Expression Regulation; Heat Shock Transcription Factors; Humans; Hydrolysis; Leupeptins; Lysosomes; Male; Oligopeptides; Osteosarcoma; Proteasome Inhibitors; Protein Processing, Post-Translational; Transcription Factors; Transcriptional Activation; Transfection; Ubiquitin

2009
Proteasome inhibitors impair RANKL-induced NF-kappaB activity in osteoclast-like cells via disruption of p62, TRAF6, CYLD, and IkappaBalpha signaling cascades.
    Journal of cellular physiology, 2009, Volume: 220, Issue:2

    Proteasome inhibitors represent a promising therapy for the treatment of relapsed and/or refractory multiple myeloma, a disease that is concomitant with osteolysis and enhanced osteoclast formation. While blockade of the proteosome pathway has been recently shown to influence osteoclast formation and function, the precise molecular cascade underlying these effects is presently unclear. Here, we provide evidence that proteasome inhibitors directly impair osteoclast formation and function via the disruption of key RANK-mediated signaling cascades. Disruption of the proteosome pathway using selective inhibitors (MG-132, MG-115, and epoxomicin) resulted in the accumulation of p62 and CYLD, and altered the subcellular targeting and distribution of p62 and TRAF6 in osteoclast-like cells. Proteosome inhibition also blocked RANKL-induced NF-kappaB activation, IkappaBalpha degradation and nuclear translocation of p65. The disruption in RANK-signaling correlated dose-dependently with an impairment in osteoclastogenesis, with relative potency epoxomicin > MG-132 > MG-115 based on equimolar concentrations. In addition, these inhibitors were found to impact osteoclastic microtubule organization and attenuate bone resorption. Based on these data we propose that deregulation of key RANK-mediated signaling cascades (p62, TRAF6, CYLD, and IkappaBalpha) underscores proteasome-mediated inhibition of osteolytic bone conditions.

    Topics: Actins; Animals; Bone Resorption; Cell Line; Cysteine; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Deubiquitinating Enzyme CYLD; Erythropoietin; Humans; I-kappa B Proteins; Leupeptins; Mice; Mice, Inbred C57BL; Microtubules; NF-kappa B; NF-KappaB Inhibitor alpha; Oligopeptides; Osteoclasts; Proteasome Endopeptidase Complex; Proteasome Inhibitors; RANK Ligand; Signal Transduction; Synaptotagmin I; TNF Receptor-Associated Factor 6; Transcription Factor TFIIH; Transcription Factors

2009
Effect of proteasome inhibitors on endotoxin-induced diaphragm dysfunction.
    American journal of physiology. Lung cellular and molecular physiology, 2009, Volume: 296, Issue:6

    Infections produce severe respiratory muscle dysfunction. It is known that the proteasome proteolytic system is activated in skeletal muscle in sepsis, and it has been postulated that this degradative pathway is responsible for inducing skeletal muscle weakness and wasting. The objective of this study was to determine if administration of proteasomal inhibitors (MG132, epoxomicin, bortezomib) can prevent sepsis-induced diaphragm weakness. Rats were given either 1) saline (0.5 ml ip), 2) endotoxin (12 mg/kg ip), 3) endotoxin plus MG132 (2.5 mg/kg), 4) endotoxin plus epoxomicin (1 micromol/kg), or 5) endotoxin plus bortezomib (0.05 mg/kg). Animals were killed either 48 or 96 h after injections, and assessments were made of diaphragm proteolysis, force-frequency relationships, mass, protein content, and caspase activation. Endotoxin increased proteolysis (P <0.001). MG132, epoxomicin, and bortezomib each prevented the endotoxin-induced increase in proteolysis (P <0.01). Endotoxin induced severe reductions in diaphragm force generation by 48 h (P <0.01); none of the proteasomal inhibitors prevented loss of force. Endotoxin induced significant reductions in diaphragm mass and protein content by 96 h (P <0.01); neither MG132 nor epoxomicin prevented loss of mass or protein, but bortezomib attenuated the reduction in protein content (P <0.05). Endotoxin increased diaphragm caspase-3 activity (P <0.01); caspase-3 activity remained high when either MG132, epoxomicin, or bortezomib were given. These data suggest proteasomal inhibitors are not an adequate treatment to prevent endotoxin-induced diaphragmatic dysfunction.

    Topics: Animals; Atrophy; Boronic Acids; Bortezomib; Caspase 3; Cysteine Proteinase Inhibitors; Diaphragm; Endotoxemia; Enzyme Activation; Leupeptins; Male; Muscle Contraction; Muscle Weakness; Oligopeptides; Organ Size; Protease Inhibitors; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Pyrazines; Rats; Rats, Sprague-Dawley; Sepsis; Tyrosine

2009
Regulation of stathmin phosphorylation in mouse liver progenitor-29 cells during proteasome inhibition.
    Proteomics, 2009, Volume: 9, Issue:19

    Proteasome inhibitors are potential therapeutic agents in the treatment of hepatocarcinoma and other liver diseases. The analysis of alternative protein phosphorylation states might contribute to elucidate the underlying mechanisms of proteasome inhibitor-induced apoptosis. We have investigated the response of mouse liver progenitor-29 (MLP-29) cells to MG132 using a combination of phosphoprotein affinity chromatography, DIGE, and nano LC-MS/MS. Thirteen unique deregulated phosphoproteins involved in chaperone activity, stress response, mRNA processing and cell cycle control were unambiguously identified. Alterations in NDRG1 and stathmin suggest new mechanisms associated to proteasome inhibitor-induced apoptosis in MLP-29 cells. Particularly, a transient modification of the phosphorylation state of Ser(16), Ser(25) and Ser(38), which are involved in the regulation of stathmin activity, was detected in three distinct isoforms upon proteasome inhibition. The parallel deregulation of calcium/calmodulin-activated protein kinase II, extracellular regulated kinase-1/2 and cyclin-dependent kinase-2, might explain the modified phosphorylation pattern of stathmin. Interestingly, stathmin phosphorylation profile was also modified in response to epoxomicin treatment, a more specific proteasome inhibitor. In summary, we report here data supporting that regulation of NDRG1 and stathmin by phosphorylation at specific Ser/Thr residues may participate in the cellular response induced by proteasome inhibitors.

    Topics: Animals; Cell Cycle Proteins; Cell Line; Cysteine Proteinase Inhibitors; Humans; Intracellular Signaling Peptides and Proteins; Leupeptins; Liver; Mice; Molecular Sequence Data; Oligopeptides; Phosphorylation; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Stathmin; Stem Cells

2009
Overexpression of endoplasmic reticulum-resident chaperone attenuates cardiomyocyte death induced by proteasome inhibition.
    Cardiovascular research, 2008, Sep-01, Volume: 79, Issue:4

    Proteasome inhibitors are a novel class of anticancer agents that induce tumour cell death via endoplasmic reticulum (ER) stress. Since ER stress is involved in the development of heart failure, we investigated the role of ER-initiated cardiomyocyte death by proteasome inhibition.. Rat neonatal cardiomyocytes were used in this study. Proteasome activity was assayed using proteasome peptidase substrates. Cell viability and apoptosis were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenol tetrazolium bromide and flow cytometry, respectively. Western blot analysis, real-time polymerase chain reaction (PCR) and reverse transcriptional PCR were used to detect the expression of protein and messenger ribonucleic acid (RNA). The location of overexpressed glucose-regulated protein (GRP) 78 was observed by confocal fluorescence microscopy. Proteasome inhibition induced cardiomyocyte death and activated ER stress-induced transcriptional factor ATF6, but not XBP1 (X-box binding protein 1), without up-regulating ER chaperones. ER-initiated apoptosis signalling, including cytosine-cytosine-adenine-adenine-thymine enhancer-binding protein (C/EBP) homologous protein (CHOP), c-Jun-N-terminal kinase (JNK), and caspase-12, was activated by proteasome inhibition. Short interference RNA targeting CHOP, but not the blockage of caspase-12 or JNK pathway, attenuated cardiomyocyte death. Overexpression of GRP78 suppressed both CHOP expression and cardiomyocyte death by proteasome inhibition.. These findings demonstrate that proteasome inhibition induces ER-initiated cardiomyocyte death via CHOP-dependent pathways without compensatory up-regulation of ER chaperones. Supplement and/or pharmacological induction of GRP78 can attenuate cardiac damage by proteasome inhibition.

    Topics: Activating Transcription Factor 6; Animals; Animals, Newborn; Antineoplastic Agents; Apoptosis; Caspase 12; Cell Survival; Cells, Cultured; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Endoplasmic Reticulum; Heat-Shock Proteins; JNK Mitogen-Activated Protein Kinases; Leupeptins; Molecular Chaperones; Myocytes, Cardiac; Oligopeptides; Oxidative Stress; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Rats; RNA Interference; RNA, Small Interfering; Signal Transduction; Transcription Factor CHOP; Tunicamycin

2008
Proteasome inhibition and stress compromise the heart in chemotherapy.
    Cardiovascular research, 2008, Sep-01, Volume: 79, Issue:4

    Topics: Animals; Antineoplastic Agents; Apoptosis; Cell Survival; Cysteine Proteinase Inhibitors; Endoplasmic Reticulum; Humans; Leupeptins; Molecular Chaperones; Myocytes, Cardiac; Oligopeptides; Oxidative Stress; Proteasome Endopeptidase Complex; Proteasome Inhibitors

2008
Antioxidants block proteasome inhibitor function in endometrial carcinoma cells.
    Anti-cancer drugs, 2008, Volume: 19, Issue:2

    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
Zinc mesoporphyrin induces rapid and marked degradation of the transcription factor Bach1 and up-regulates HO-1.
    Biochimica et biophysica acta, 2008, Volume: 1779, Issue:3

    Heme oxygenase 1 (HO-1) is the first and rate-controlling enzyme in heme degradation. Bach1 is a mammalian transcriptional repressor of HO-1. To understand how zinc mesoporphyrin (ZnMP) induces the expression of HO-1, we investigated the effects of ZnMP on Bach1 mRNA and protein levels in human hepatoma Huh-7 cells by quantitative RT-PCR and Western blots. We found that ZnMP markedly up-regulated HO-1 mRNA and protein levels, and rapidly and significantly decreased Bach1 protein levels by increasing degradation of Bach1 protein [half life (t(1/2)) from 19 h to 45 min], whereas ZnMP did not influence Bach1 mRNA levels. The proteasome inhibitors, epoxomicin and MG132, significantly inhibited degradation of Bach1 by ZnMP in a dose-dependent fashion, indicating that the degradation of Bach1 by ZnMP is proteasome-dependent. Purified Bach1 C-terminal fragment bound heme, but there was no evidence for binding of ZnMP to the heme-binding region of Bach1. In conclusion, ZnMP produces profound post-transcriptional down-regulation of Bach1 protein levels and transcriptional up-regulation of HO-1. Our results indicate that ZnMP up-regulates HO-1 gene expression by markedly increasing Bach1 protein degradation in a proteasome-dependent manner.

    Topics: Basic-Leucine Zipper Transcription Factors; Cell Line, Tumor; Fanconi Anemia Complementation Group Proteins; Gene Expression Regulation, Enzymologic; Heme; Heme Oxygenase-1; Humans; Leupeptins; Metalloporphyrins; Oligopeptides; Protein Binding; Recombinant Proteins; RNA, Messenger; Up-Regulation

2008
Dose-dependent inhibition of proteasome activity by a mutant ubiquitin associated with neurodegenerative disease.
    Journal of cell science, 2007, May-01, Volume: 120, Issue:Pt 9

    The ubiquitin-proteasome system is the main regulated intracellular proteolytic pathway. Increasing evidence implicates impairment of this system in the pathogenesis of diseases with ubiquitin-positive pathology. A mutant ubiquitin, UBB(+1), accumulates in the pathological hallmarks of tauopathies, including Alzheimer's disease, polyglutamine diseases, liver disease and muscle disease and serves as an endogenous reporter for proteasomal dysfunction in these diseases. UBB(+1) is a substrate for proteasomal degradation, however it can also inhibit the proteasome. Here, we show that UBB(+1) properties shift from substrate to inhibitor in a dose-dependent manner in cell culture using an inducible UBB(+1) expression system. At low expression levels, UBB(+1) was efficiently degraded by the proteasome. At high levels, the proteasome failed to degrade UBB(+1), causing its accumulation, which subsequently induced a reversible functional impairment of the ubiquitin-proteasome system. Also in brain slice cultures, UBB(+1) accumulation and concomitant proteasome inhibition was only induced at high expression levels. Our findings show that by varying UBB(+1) expression levels, the dual proteasome substrate and inhibitory properties can be optimally used to serve as a research tool to study the ubiquitin-proteasome system and to further elucidate the role of aberrations of this pathway in disease.

    Topics: Animals; Blotting, Western; Cerebral Cortex; Cysteine Proteinase Inhibitors; Cytosol; Dose-Response Relationship, Drug; Doxorubicin; Flow Cytometry; Frameshift Mutation; Gene Expression; Green Fluorescent Proteins; HeLa Cells; Humans; Leupeptins; Mice; Mice, Inbred C57BL; Mice, Transgenic; Models, Biological; Neurodegenerative Diseases; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Tissue Culture Techniques; Transfection; Ubiquitin

2007
The E3 Ligase MuRF1 degrades myosin heavy chain protein in dexamethasone-treated skeletal muscle.
    Cell metabolism, 2007, Volume: 6, Issue:5

    Skeletal muscle atrophy occurs as a side effect of treatment with synthetic glucocorticoids such as dexamethasone (DEX) and is a hallmark of cachectic syndromes associated with increased cortisol levels. The E3 ubiquitin ligase MuRF1 (muscle RING finger protein 1) is transcriptionally upregulated by DEX treatment. Differentiated myotubes treated with DEX undergo depletion of myosin heavy chain protein (MYH), which physically associates with MuRF1. This loss of MYH can be blocked by inhibition of MuRF1 expression. When wild-type and MuRF1(-/-) mice are treated with DEX, the MuRF1(-/-) animals exhibit a relative sparing of MYH. In vitro, MuRF1 is shown to function as an E3 ubiquitin ligase for MYH. These data identify the mechanism by which MYH is depleted under atrophy conditions and demonstrate that inhibition of a single E3 ligase, MuRF1, is sufficient to maintain this important sarcomeric protein.

    Topics: Animals; Blotting, Western; Cell Line; Dexamethasone; Gene Expression; Glucocorticoids; Leupeptins; Mice; Mice, Knockout; Muscle Proteins; Muscle, Skeletal; Myosin Heavy Chains; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Binding; Protein Isoforms; RNA Interference; Tripartite Motif Proteins; Ubiquitin-Protein Ligases; Ubiquitination

2007
Ubiquitin-proteasome system stress sensitizes ovarian cancer to proteasome inhibitor-induced apoptosis.
    Cancer research, 2006, Apr-01, Volume: 66, Issue:7

    The ubiquitin-proteasome system (UPS) mediates targeted protein degradation. Notably, the UPS determines levels of key checkpoint proteins controlling apoptosis and proliferation by controlling protein half-life. Herein, we show that ovarian carcinoma manifests an overstressed UPS by comparison with normal tissues by accumulation of ubiquitinated proteins despite elevated proteasome levels. Elevated levels of total ubiquitinated proteins and 19S and 20S proteasome subunits are evident in both low-grade and high-grade ovarian carcinoma tissues relative to benign ovarian tumors and in ovarian carcinoma cell lines relative to immortalized surface epithelium. We find that ovarian carcinoma cell lines exhibit greater sensitivity to apoptosis in response to proteasome inhibitors than immortalized ovarian surface epithelial cells. This sensitivity correlates with increased cellular proliferation rate and UPS stress rather than absolute proteasome levels. Proteasomal inhibition in vitro induces cell cycle arrest and the accumulation of p21 and p27 and triggers apoptosis via activation of caspase-3. Furthermore, treatment with the licensed proteasome inhibitor PS-341 slows the growth of ES-2 ovarian carcinoma xenograft in immunodeficient mice. In sum, elevated proliferation and metabolic rate resulting from malignant transformation of the epithelium stresses the UPS and renders ovarian carcinoma more sensitive to apoptosis in response to proteasomal inhibition.

    Topics: Animals; Apoptosis; Boronic Acids; Bortezomib; Caspases; Cell Division; Cell Line, Tumor; Female; G2 Phase; Humans; Leupeptins; Mice; Mice, Nude; Oligopeptides; Ovarian Neoplasms; Protease Inhibitors; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Pyrazines; Ubiquitin; Xenograft Model Antitumor Assays

2006
Roles of the ubiquitin/proteasome pathway in pollen tube growth with emphasis on MG132-induced alterations in ultrastructure, cytoskeleton, and cell wall components.
    Plant physiology, 2006, Volume: 141, Issue:4

    The ubiquitin/proteasome pathway represents one of the most important proteolytic systems in eukaryotes and has been proposed as being involved in pollen tube growth, but the mechanism of this involvement is still unclear. Here, we report that proteasome inhibitors MG132 and epoxomicin significantly prevented Picea wilsonii pollen tube development and markedly altered tube morphology in a dose- and time-dependent manner, while hardly similar effects were detected when cysteine-protease inhibitor E-64 was used. Fluorogenic kinetic assays using fluorogenic substrate sLLVY-AMC confirmed MG132-induced inhibition of proteasome activity. The inhibitor-induced accumulation of ubiquitinated proteins (UbPs) was also observed using immunoblotting. Transmission electron microscopy revealed that MG132 induces endoplasmic reticulum (ER)-derived cytoplasmic vacuolization. Immunogold-labeling analysis demonstrated a significant accumulation of UbPs in degraded cytosol and dilated ER in MG132-treated pollen tubes. Fluorescence labeling with fluorescein isothiocyanate-phalloidin and beta-tubulin antibody revealed that MG132 disrupts the organization of F-actin and microtubules and consequently affects cytoplasmic streaming in pollen tubes. However, tip-focused Ca2+ gradient, albeit reduced, seemingly persists after MG132 treatment. Finally, fluorescence labeling with antipectin antibodies and calcofluor indicated that MG132 treatment induces a sharp decline in pectins and cellulose. This result was confirmed by Fourier transform infrared analysis, thus demonstrating for the first time the inhibitor-induced weakening of tube walls. Taken together, these findings suggest that MG132 treatment promotes the accumulation of UbPs in pollen tubes, which induces ER-derived cytoplasmic vacuolization and depolymerization of cytoskeleton and consequently strongly affects the deposition of cell wall components, providing a mechanistic framework for the functions of proteasome in the tip growth of pollen tubes.

    Topics: Actin Cytoskeleton; Cell Wall; Cellulose; Cytoplasmic Streaming; Cytoskeleton; Endoplasmic Reticulum; Immunohistochemistry; Leupeptins; Microscopy, Electron, Transmission; Microtubules; Models, Biological; Oligopeptides; Pectins; Picea; Pollen; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Ubiquitin; Vacuoles

2006
Proteasome inhibitors sensitize colon carcinoma cells to TRAIL-induced apoptosis via enhanced release of Smac/DIABLO from the mitochondria.
    Pathology oncology research : POR, 2006, Volume: 12, Issue:3

    The synergistic interaction between proteasome inhibitors and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a promising approach to induce cell death in tumor cells. However, the molecular and biochemical mechanisms of this synergism have been proven to be cell type specific. We therefore focused our investigation on TRAIL-resistant colon carcinoma cells in this study. DNA fragmentation, mitochondrial membrane depolarization and increased caspase-3-like enzyme activity was exclusively induced only by combined treatment with proteasome inhibitors (epoxomicin, MG132, bortezomib/PS-341) and TRAIL. The expression level of anti-apoptotic proteins (XIAP, survivin, Bcl-2, Bcl-XL), regulated by NF-kappaB transcription factor, was not effected by any of these treatments. TRAIL alone induced only partial activation of caspase-3 (p20), while the combination of TRAIL and proteasome inhibition led to the full proteolytic activation of caspase-3 (p17). Only the combination treatment induced marked membrane depolarization and the release of cytochrome c, HtrA2/Omi and Smac/DIABLO. Apoptosis-inducing factor (AIF) was not released in any of these conditions. These results are consistent with a model where the full activation of caspase-3 by caspase-8 is dependent on the release of Smac/DIABLO in response to the combined treatment. This molecular mechanism, independent of the inhibition NF-kappaB activity, may provide rationale for the combination treatment of colon carcinomas with proteasome inhibitors and recombinant TRAIL or agonistic antibody of TRAIL receptors.

    Topics: Antineoplastic Combined Chemotherapy Protocols; Apoptosis; Apoptosis Regulatory Proteins; Boronic Acids; Bortezomib; Caspase 3; Caspase 8; Colonic Neoplasms; Drug Synergism; Enzyme Activation; Humans; Intracellular Signaling Peptides and Proteins; Leupeptins; Mitochondria; Mitochondrial Proteins; Oligopeptides; Protease Inhibitors; Proteasome Inhibitors; Pyrazines; TNF-Related Apoptosis-Inducing Ligand

2006
Antiviral activity of proteasome inhibitors in herpes simplex virus-1 infection: role of nuclear factor-kappaB.
    Antiviral therapy, 2006, Volume: 11, Issue:8

    Herpes simplex virus type 1 (HSV-1) is a potent inducer of nuclear factor-KB (NF-kappaB), a cellular transcription factor with a crucial role in promoting inflammation and controlling cell proliferation and survival.. On the basis of the recent demonstration that HSV-1-induced NF-kappaB is actively recruited to KB-binding sites in the HSV-1 infected-cell protein 0 (ICPO) promoter enhancing viral transcription and replication, we investigated the effect of proteasome inhibitors MG132, MG115 and epoxomicin, which block NF-kappaB function by preventing the degradation of the inhibitory proteins IkappaBalpha, on HSV-1-induced NF-kappaB activation and viral replication.. Antiviral activity of proteasome inhibitors was analysed in HSV-1-infected HEp2 cells by determining infective virus titres by CPE50%, viral RNA synthesis by RT-PCR, and viral protein synthesis by immunoblot analysis or immunofluorescence. ICPO transcription was studied in transient transfection experiments using the ICPO promoter-luciferase IE1-Luc construct. IkappaBalpha degradation and NF-kappaB activity were determined by immunoblot analysis and EMSA, respectively.. Proteasome inhibitors were found to prevent HSV-1-induced NF-kappaB activation in the early phase of infection. Block of virus-induced NF-kappaB activation resulted in inhibiting HSV-1 ICPO gene expression, in decreasing the level of immediate-early and late viral proteins, and ultimately in greatly suppressing viral replication. The antiviral effect was lost if treatment was started after NF-kappaB activation, and appeared to be independent of the HSV-1-induced activation of the JNK pathway.. Proteasome inhibitors possess NF-kappaB-dependent antiherpetic activity. The results described further identify the IKK/NF-kappaB pathway as a suitable target for novel antiherpetic drugs.

    Topics: Animals; Antiviral Agents; Cell Line, Tumor; Chlorocebus aethiops; Down-Regulation; Herpesvirus 1, Human; Humans; Immediate-Early Proteins; Leupeptins; NF-kappa B; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Transcription Factor AP-1; Vero Cells

2006
Evidence for a protective role of Mcl-1 in proteasome inhibitor-induced apoptosis.
    Blood, 2005, Apr-15, Volume: 105, Issue:8

    Proteasome inhibitors exhibit antitumor activity against malignancies of different histology. Yet, the mechanisms underlying this effect are poorly understood. Recent evidence indicates that antiapoptotic factors may also accumulate as a consequence of exposure to these drugs, possibly reducing their cytotoxicity. These include the Bcl-2 family member Mcl-1, whose down-regulation has been proposed to initiate apoptosis in response to genotoxic stimuli. In this study, we found that proteasome inhibitors release cyotochrome c and second mitochondria-derived activator of caspase (SMAC)/Diablo and trigger the subsequent apoptotic cascade in spite of concomitant Mcl-1 increase. However, our data indicate that subtraction of Mcl-1 during apoptosis, although not required for early release of proapoptotic factors, is probably relevant in speeding up cell demise, since RNA interference-mediated Mcl-1 silencing is lethal in lymphoma cells. Consistent with this, the cytotoxic effects of proteasome inhibitors are enhanced when Mcl-1 increase is impeded. Thus, this study identifies Mcl-1 accumulation as an unwanted molecular consequence of exposure to proteasome inhibitors, which slows down their proapoptotic effects. Pharmacologic or genetic approaches targeting Mcl-1, including therapeutic RNAi, may increase the effectiveness of these compounds.

    Topics: Acetylcysteine; Apoptosis; Apoptosis Regulatory Proteins; Carrier Proteins; Caspases; Cell Survival; Complement Membrane Attack Complex; Complement System Proteins; Cysteine Proteinase Inhibitors; Cytochromes c; Gene Expression; Glycoproteins; Humans; Intracellular Signaling Peptides and Proteins; Jurkat Cells; Leupeptins; Lymphocytes; Mitochondria; Mitochondrial Proteins; Myeloid Cell Leukemia Sequence 1 Protein; Neoplasm Proteins; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Proto-Oncogene Proteins c-bcl-2; RNA Interference

2005
Cooperative cytotoxicity of proteasome inhibitors and tumor necrosis factor-related apoptosis-inducing ligand in chemoresistant Bcl-2-overexpressing cells.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2005, Jun-01, Volume: 11, Issue:11

    Bcl-2 overexpression is frequently detected in lymphoid malignancies, being associated with poor prognosis and reduced response to therapy. Here, we evaluated whether Bcl-2 overexpression affects the cytotoxic activity of proteasome inhibitors taken alone or in association with conventional anticancer drugs or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL).. Jurkat cells engineered to overexpress Bcl-2 were treated with proteasome inhibitors (MG132, epoxomicin, and bortezomib), anticancer drugs (etoposide and doxorubicin), TRAIL, or combinations of these compounds. Cell death and loss of mitochondrial transmembrane potential were detected by flow cytometry. Cytosolic relocalization of cytochrome c and SMAC/Diablo, caspase cleavage, and Bcl-2 and Mcl-1 levels were determined by immunoblotting. Nuclear factor-kappaB inhibition was done by retroviral transduction with a dominant-negative mutant of IkappaBalpha.. Bcl-2 overexpression results in significant inhibition of apoptosis in response to proteasome inhibitors, antiblastics, and TRAIL. Addition of TRAIL to proteasome inhibitors results in a synergistic cytotoxic effect in Bcl-2-overexpressing cells, whereas this result is not reproduced by the combination of proteasome inhibitors with antiblastic drugs. Importantly, proteasome inhibitors plus TRAIL induce mitochondrial dysfunction irrespective of up-regulated Bcl-2. Bcl-2 cleavage to a fragment with putative proapoptotic activity and elimination of antiapoptotic Mcl-1 may both play a role in proteasome inhibitors-TRAIL cooperation. Conversely, nuclear factor-kappaB inhibition by proteasome inhibitors is per se insufficient to explain the observed synergy.. Combined proteasome inhibitors and TRAIL overcome the apoptotic threshold raised by Bcl-2 and may prove useful in the treatment of chemoresistant malignancies with up-regulated Bcl-2.

    Topics: Antineoplastic Agents; Apoptosis; Apoptosis Regulatory Proteins; Boronic Acids; Bortezomib; Cell Line; Cell Survival; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Doxorubicin; Drug Resistance, Neoplasm; Drug Synergism; Etoposide; Flow Cytometry; Humans; Immunoblotting; Jurkat Cells; Leupeptins; Membrane Glycoproteins; Mitochondria; Myeloid Cell Leukemia Sequence 1 Protein; Neoplasm Proteins; NF-kappa B; Oligopeptides; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Proto-Oncogene Proteins c-bcl-2; Pyrazines; TNF-Related Apoptosis-Inducing Ligand; Tumor Necrosis Factor-alpha

2005
Deubiquitination by proteasome is coordinated with substrate translocation for proteolysis in vivo.
    Experimental cell research, 2005, Jul-15, Volume: 307, Issue:2

    The 26S proteasome mediates degradation of protein substrates labeled with polyUb chains. After recognition by the 19S proteasome regulatory complex, polyUb chains are disassembled and substrates are processed in the 20S core of proteasome. However, the exact relationship of degradation-associated deubiquitination to substrate processing remains unclear. Here, using Ub-based tagging strategies, we provided evidence that removable polyUb chains serve as the signal for proteolytic processing of ubiquitinated substrates. We showed that inhibition of the proteasome by proteasome inhibitor MG132 results in trapping of the substrate in the proteasome. Such a trapping allows proteasomal cleavage of attached non-removable Ub mutant (UbV75,76), which is otherwise a "difficult" deubiquitination substrate. Characterization of deubiquitination and degradation intermediates, generated due to incomplete proteolytic inhibition, revealed changes in proteolytic cleavage sites, within the Gal4-VP16 model substrate, suggesting that the copy number of attached UbV75,76 affects substrate processing. Conversion of lysine48 to arginine48 in UbV75,76 did not have significant effect on in vivo polyubiquitination of multiple Ub-fused substrates, but considerably reduced proteolytic intermediates. Taken together, the results support a model in which deubiquitination process is a crucial event for proteolysis of ubiquitinated substrates and such an event is coordinated with substrate translocation.

    Topics: Amino Acid Substitution; Arginine; Blotting, Western; Cell Line, Tumor; Cysteine Proteinase Inhibitors; Gene Expression; HeLa Cells; Humans; Leupeptins; Lysine; Mutation; Oligopeptides; Peptide Fragments; Protease Inhibitors; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Protein Subunits; Proteins; Recombinant Fusion Proteins; Trans-Activators; Transfection; Ubiquitin

2005
The proteasome metabolizes peptide-mediated nonviral gene delivery systems.
    Gene therapy, 2005, Volume: 12, Issue:21

    The proteasome is a multisubunit cytosolic protein complex responsible for degrading cytosolic proteins. Several studies have implicated its involvement in the processing of viral particles used for gene delivery, thereby limiting the efficiency of gene transfer. Peptide-based nonviral gene delivery systems are sufficiently similar to viral particles in their size and surface properties and thereby could also be recognized and metabolized by the proteasome. The present study utilized proteasome inhibitors (MG 115 and MG 132) to establish that peptide DNA condensates are metabolized by the proteasome, thereby limiting their gene transfer efficiency. Transfection of HepG2 or cystic fibrosis/T1 (CF/T1) cells with CWK18 DNA condensates in the presence of MG 115 or MG 132 resulted in significantly enhanced gene expression. MG 115 and MG 132 increased luciferase expression 30-fold in a dose-dependent manner in HepG2 and CF/T1. The enhanced gene expression correlated directly with proteasome inhibition, and was not the result of lysosomal enzyme inhibition. The enhanced transfection was specific for peptide DNA condensates, whereas Lipofectamine- and polyethylenimine-mediated gene transfer were significantly blocked. A series of novel gene transfer peptides containing intrinsic GA proteasome inhibitors (CWK18(GA)n, where n=4, 6, 8 and 10) were synthesized and found to inhibit the proteasome. The gene transfer efficiency mediated by these peptides in four different cell lines established that a GA repeat of four is sufficient to block the proteasome and significantly enhance the gene transfer. Together, these results implicate the proteasome as a previously undiscovered route of metabolism of peptide-based nonviral gene delivery systems and provide a rationale for the use of proteasome inhibition to increase gene transfer efficiency.

    Topics: Cell Line; Cell Line, Tumor; Cystic Fibrosis; DNA; Gene Expression; Genetic Therapy; Humans; Leupeptins; Lipids; Luciferases; Lung; Oligopeptides; Peptides; Polyethyleneimine; Protease Inhibitors; Proteasome Endopeptidase Complex; Transfection

2005
Proteasome inhibition induces a senescence-like phenotype in primary human fibroblasts cultures.
    Biogerontology, 2004, Volume: 5, Issue:1

    Senescent human fibroblasts exhibit several genetic and biochemical differences as compared to their young counterparts including abnormalities of the main proteolytic mechanism, namely the proteasome. Specifically, we and others have shown that there is an impaired function of the proteasome, as senescent cells have reduced proteolytic activities and less proteasome content. In a complementary work we have recently shown that inhibition of the proteasome by a specific inhibitor induces a senescence-like phenotype in young WI38 fibroblasts [Chondrogianni et al. (2003) J Biol Chem 278: 28026-28037]. In this study we tested whether the induction of a senescence-like phenotype following treatment with proteasome inhibitors is a common feature of primary human fibroblasts. A comparative biochemical analysis, after employing three different human fibroblasts cell lines (IMR90, MRC5 and WI38 cells), as well as two proteasome inhibitors (epoxomicin and MG132), has shown that proteasome inhibition results in the appearance of a senescence-like phenotype in all cell lines used. Proteasome inhibitors treated cells were irreversibly stopped dividing, exhibited positive staining to beta-galactosidase as well as reduced CT-L and PGPH activities. In summary, these data reveal the fundamental role of the proteasome in the progression of replicative senescence and open new dimensions towards a better understanding of protein degradation.

    Topics: beta-Galactosidase; Cell Division; Cell Line; Cellular Senescence; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Endopeptidases; Fibroblasts; Humans; Leupeptins; Multienzyme Complexes; Oligopeptides; Peptide Hydrolases; Phenotype; Proteasome Endopeptidase Complex

2004
Ubiquitin-dependent lysosomal degradation of the HNE-modified proteins in lens epithelial cells.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2004, Volume: 18, Issue:12

    4-hydroxynonenal (HNE), a highly reactive lipid peroxidation product, may adversely modify proteins. Accumulation of HNE-modified proteins may be responsible for pathological lesions associated with oxidative stress. The objective of this work was to determine how HNE-modified proteins are removed from cells. The data showed that alphaB-crystallin modified by HNE was ubiquitinated at a faster rate than that of native alphaB-crystallin in a cell-free system. However, its susceptibility to proteasome-dependent degradation in the cell-free system did not increase. When delivered into cultured lens epithelial cells, HNE-modified alphaB-crystallin was degraded at a faster rate than that of unmodified alphaB-crystallin. Inhibition of the lysosomal activity stabilized HNE-modified alphaB-crystallin, but inhibition of the proteasome activity alone had little effect. To determine if other HNE-modified proteins are also degraded in a ubiquitin-dependent lysosomal pathway, lens epithelial cells were treated with HNE and the removal of HNE-modified proteins in the cells was monitored. The levels of HNE-modified proteins in the cell decreased rapidly upon removal of HNE from the medium. Depletion of ATP or the presence of MG132, a proteasome/lysosome inhibitor, resulted in stabilization of HNE-modified proteins. However, proteasome-specific inhibitors, lactacystin-beta-lactone and epoxomicin, could not stabilize HNE-modified proteins in the cells. In contrast, chloroquine, a lysosome inhibitor, stabilized HNE-modified proteins. The enrichment of HNE-modified proteins in the fraction of ubiquitin conjugates suggests that HNE-modified proteins are preferentially ubiquitinated. Taken together, these findings show that HNE-modified proteins are degraded via a novel ubiquitin and lysosomal-dependent but proteasome-independent pathway.

    Topics: Acetylcysteine; Adenosine Triphosphate; Aldehydes; alpha-Crystallin B Chain; Cell-Free System; Cells, Cultured; Epithelial Cells; Humans; Lens, Crystalline; Leupeptins; Lysosomes; Oligopeptides; Oxidative Stress; Proteasome Endopeptidase Complex; Proteasome Inhibitors; Ubiquitin

2004
Proteasome inhibitors: their effects on arachidonic acid release from cells in culture and arachidonic acid metabolism in rat liver cells.
    BMC pharmacology, 2004, Aug-05, Volume: 4

    I have postulated that arachidonic acid release from rat liver cells is associated with cancer chemoprevention. Since it has been reported that inhibition of proteasome activities may prevent cancer, the effects of proteasome inhibitors on arachidonic acid release from cells and on prostaglandin I2 production in rat liver cells were studied.. The proteasome inhibitors, epoxomicin, lactacystin and carbobenzoxy-leucyl-leucyl-leucinal, stimulate the release of arachidonic acid from rat glial, human colon carcinoma, human breast carcinoma and the rat liver cells. They also stimulate basal and induced prostacycin production in the rat liver cells. The stimulated arachidonic acid release and basal prostaglandin I2 production in rat liver cells is inhibited by actinomycin D.. Stimulation of arachidonic acid release and arachidonic acid metabolism may be associated with some of the biologic effects observed after proteasome inhibition, e.g. prevention of tumor growth, induction of apoptosis, stimulation of bone formation.

    Topics: Acetylcysteine; Animals; Arachidonic Acid; Breast Neoplasms; Cell Line; Cell Line, Tumor; HT29 Cells; Humans; Leupeptins; Liver; Neuroglia; Oligopeptides; Proteasome Inhibitors; Rats

2004
Assessment of proteasome activity in cell lysates and tissue homogenates using peptide substrates.
    The international journal of biochemistry & cell biology, 2003, Volume: 35, Issue:5

    The ubiquitin-proteasome pathway is a major route of degradation of cell proteins. It also plays an essential role in maintaining cell homeostasis by degrading many rate-limiting enzymes and critical regulatory proteins. Alterations in proteasome activity have been implicated in a number of pathologies including Parkinson's disease, Alzheimer's disease and diabetes. The eukaryotic proteasome is a multicatalytic protease characterized by three activities with distinct specificities against peptide substrates. Although substrates were identified which could selectively measure the individual activities in the purified proteasome little data is available on how specific those substrates are for proteasomal activity when used with biological samples which may contain many other active peptidases. Here we examine the three major peptidase activities in lysates of two cell types and in a liver cytosol fraction in the presence of specific proteasome inhibitors and after fractionation by gel permeation chromatography. We demonstrate that other proteinases present in these preparations can degrade the commonly used proteasome substrates under the standard assay conditions. We develop a simple method for separating the proteasome from the lower molecular weight proteases using a 500kDa molecular weight cut-off membrane. This allows proteasome activity to be accurately measured in crude biological samples and may have quite broad applicability. We also identify low molecular weight tryptic activity in both the cell and tissue preparations which could not be inhibited by the proteasome inhibitor epoxomycin but was inhibitable by two cysteine proteinase inhibitors and by lactacystin suggesting that lactacystin may not be completely proteasome specific.

    Topics: Acetylcysteine; Animals; Cell Line; Chromatography, Gel; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Humans; Leupeptins; Mice; Multienzyme Complexes; Oligopeptides; Peptide Hydrolases; Proteasome Endopeptidase Complex; Rats; Substrate Specificity; Ubiquitin

2003
Inducible p27(Kip1) expression inhibits proliferation of K562 cells and protects against apoptosis induction by proteasome inhibitors.
    Cell death and differentiation, 2003, Volume: 10, Issue:3

    Overexpression of the cyclin-dependent kinase inhibitor p27(Kip1) has been demonstrated to induce cell cycle arrest and apoptosis in various cancer cell lines, but has also been associated with the opposite effect of enhanced survival of tumor cells and increased resistance towards chemotherapeutic treatment. To address the question of how p27(Kip1) expression is related to apoptosis induction, we studied doxycycline-regulated p27(Kip1) expression in K562 erythroleukemia cells. p27(Kip1) expression effectively retards proliferation, but it is not sufficient to induce apoptosis in K562 cells. p27(Kip1)-expressing K562 cells, however, become resistant to apoptosis induction by the proteasome inhibitors PSI, MG132 and epoxomicin, in contrast to wild-type K562 cells that are efficiently killed. Cell cycle arrest in the S phase by aphidicolin, which is not associated with an accumulation of p27(Kip1) protein, did not protect K562 cells against the cytotoxic effect of the proteasome inhibitor PSI. The expression levels of p27(Kip1) thus constitute an important parameter, which decides on the overall sensitivity of cells against the cytotoxic effect of proteasome inhibitors.

    Topics: Antibiotics, Antineoplastic; Aphidicolin; Apoptosis; Blotting, Western; Caspases; Cell Cycle; Cell Cycle Proteins; Cell Division; Cell Nucleus; Cell Separation; Cyclin-Dependent Kinase Inhibitor p27; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Dose-Response Relationship, Drug; Doxorubicin; Flow Cytometry; Genetic Vectors; Hemoglobins; Humans; K562 Cells; Leupeptins; Microscopy, Fluorescence; Models, Genetic; Multienzyme Complexes; Oligopeptides; Proteasome Endopeptidase Complex; Retroviridae; S Phase; Time Factors; Transfection; Tumor Suppressor Proteins

2003
Central role of the proteasome in senescence and survival of human fibroblasts: induction of a senescence-like phenotype upon its inhibition and resistance to stress upon its activation.
    The Journal of biological chemistry, 2003, Jul-25, Volume: 278, Issue:30

    Normal human fibroblasts undergo a limited number of divisions in culture and progressively they reach a state of irreversible growth arrest, a process termed as replicative senescence. The proteasome is the major cellular proteolytic machinery, the function of which is impaired during replicative senescence. However, the exact causes of its malfunction in these conditions are unknown. Using WI38 fibroblasts as a model for cellular senescence we have observed reduced levels of proteasomal peptidase activities coupled with increased levels of both oxidized and ubiquitinated proteins in senescent cells. We have found the catalytic subunits of the 20 S complex and subunits of the 19 S regulatory complex to be down-regulated in senescent cells. This is accompanied by a decrease in the level of both 20 S and 26 S complexes. Partial inhibition of proteasomes in young cells caused by treatment with specific inhibitors induced a senescence-like phenotype, thus demonstrating the fundamental importance of the proteasome for retaining cellular maintenance and homeostasis. Stable overexpression of beta1 and beta5 subunits in WI38 established cell lines was shown to induce elevated expression levels of beta1 subunit in beta5 transfectants and vice versa. Transfectants possess increased proteasome activities and most importantly, increased capacity to cope better with various stresses. In summary these data demonstrate the central role of the proteasome during cellular senescence and survival as well as provide insights toward a better understanding of proteasome regulation.

    Topics: beta-Galactosidase; Cell Division; Cell Line; Cell Survival; Cellular Senescence; Chromatography, Gel; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; DNA; Down-Regulation; Electrophoresis, Gel, Two-Dimensional; Fibroblasts; Humans; Leupeptins; Microscopy, Confocal; Microscopy, Fluorescence; Multienzyme Complexes; Oligopeptides; Oxidative Stress; Peptide Hydrolases; Phenotype; Precipitin Tests; Proteasome Endopeptidase Complex; Time Factors; Transfection

2003
A transgenic mouse model of the ubiquitin/proteasome system.
    Nature biotechnology, 2003, Volume: 21, Issue:8

    Impairment of the ubiquitin/proteasome system has been proposed to play a role in neurodegenerative disorders such as Alzheimer and Parkinson diseases. Although recent studies confirmed that some disease-related proteins block proteasomal degradation, and despite the existence of excellent animal models of both diseases, in vivo data about the system are lacking. We have developed a model for in vivo analysis of the ubiquitin/proteasome system by generating mouse strains transgenic for a green fluorescent protein (GFP) reporter carrying a constitutively active degradation signal. Administration of proteasome inhibitors to the transgenic animals resulted in a substantial accumulation of GFP in multiple tissues, confirming the in vivo functionality of the reporter. Moreover, accumulation of the reporter was induced in primary neurons by UBB+1, an aberrant ubiquitin found in Alzheimer disease. These transgenic animals provide a tool for monitoring the status of the ubiquitin/proteasome system in physiologic or pathologic conditions.

    Topics: Alzheimer Disease; Animals; Boronic Acids; Cells, Cultured; Cysteine Endopeptidases; Fibroblasts; Leupeptins; Mice; Mice, Inbred C57BL; Mice, Transgenic; Microscopy, Fluorescence; Models, Animal; Multienzyme Complexes; Myocytes, Cardiac; Neurodegenerative Diseases; Neurons; Oligopeptides; Organ Specificity; Parkinson Disease; Proteasome Endopeptidase Complex; Recombinant Fusion Proteins; Tissue Distribution; Ubiquitin

2003
Conversion of PrP to a self-perpetuating PrPSc-like conformation in the cytosol.
    Science (New York, N.Y.), 2002, Nov-29, Volume: 298, Issue:5599

    A rare conformation of the prion protein, PrPSc, is found only in mammals with transmissible prion diseases and represents either the infectious agent itself or a major component of it. The mechanism for initiating PrPSc formation is unknown. We report that PrP retrogradely transported out of the endoplasmic reticulum produced both amorphous aggregates and a PrPSc-like conformation in the cytosol. The distribution between these forms correlated with the rate of appearance in the cytosol. Once conversion to the PrPSc-like conformation occurred, it was sustained. Thus, PrP has an inherent capacity to promote its own conformational conversion in mammalian cells. These observations might explain the origin of PrPSc.

    Topics: Acetylcysteine; Animals; COS Cells; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Cystic Fibrosis Transmembrane Conductance Regulator; Cytosol; Endopeptidase K; Endoplasmic Reticulum; Leupeptins; Mice; Models, Biological; Multienzyme Complexes; Neurons; Oligopeptides; Prions; Proteasome Endopeptidase Complex; Protein Conformation; Protein Folding; Protein Transport; PrPSc Proteins; Solubility; Transfection; Tumor Cells, Cultured

2002
Cells adapted to the proteasome inhibitor 4-hydroxy- 5-iodo-3-nitrophenylacetyl-Leu-Leu-leucinal-vinyl sulfone require enzymatically active proteasomes for continued survival.
    Proceedings of the National Academy of Sciences of the United States of America, 2001, Jan-16, Volume: 98, Issue:2

    The proteasome is the primary protease used by cells for degrading proteins and generating peptide ligands for class I molecules of the major histocompatibility complex. Based on the properties of cells adapted to grow in the presence of the proteasome inhibitor 4-hydroxy-5-iodo-3-nitrophenylacetyl-Leu-Leu-leucinal-vinyl sulfone (NLVS), it was proposed that proteasomes can be replaced by alternative proteolytic systems, particularly a large proteolytic complex with a tripeptidyl peptidase II activity. Here we show that NLVS-adapted cells retain sensitivity to a number of highly specific proteasome inhibitors with regard to antigenic peptide generation, accumulation of polyubiquitinated proteins, degradation of p53, and cell viability. In addition, we show that in the same assays (with a single minor exception), NLVS-adapted cells are about as sensitive as nonselected cells to Ala-Ala-Phe-chloromethylketone, a specific inhibitor of tripeptidyl peptidase II activity. Based on these findings, we conclude that proteasomes still have essential proteolytic functions in adapted cells that are not replaced by Ala-Ala-Phe-chloromethylketone-sensitive proteases.

    Topics: Amino Acid Chloromethyl Ketones; Aminopeptidases; Animals; Antigen Presentation; Antigens; Boronic Acids; Bortezomib; CD8-Positive T-Lymphocytes; Cell Survival; Cysteine Endopeptidases; Dipeptidyl-Peptidases and Tripeptidyl-Peptidases; Drug Resistance; Endopeptidases; Enzyme Activation; H-2 Antigens; Leupeptins; Lymphoma, T-Cell; Mice; Multienzyme Complexes; Neoplasm Proteins; Oligopeptides; Peptide Fragments; Phenols; Protease Inhibitors; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; Pyrazines; Selection, Genetic; Serine Endopeptidases; Sulfones; Thymus Neoplasms; Tumor Cells, Cultured; Tumor Suppressor Protein p53; Tyramine; Ubiquitins

2001
The proteasome participates in degradation of mutant alpha 1-antitrypsin Z in the endoplasmic reticulum of hepatoma-derived hepatocytes.
    The Journal of biological chemistry, 2001, Nov-30, Volume: 276, Issue:48

    Because retention of mutant alpha(1)-antitrypsin (alpha(1)-AT) Z in the endoplasmic reticulum (ER) is associated with liver disease in alpha(1)-AT-deficient individuals, the mechanism by which this aggregated glycoprotein is degraded has received considerable attention. In previous studies using stable transfected human fibroblast cell lines and a cell-free microsomal translocation system, we found evidence for involvement of the proteasome in degradation of alpha(1)-ATZ (Qu, D., Teckman, J. H., Omura, S., and Perlmutter, D. H. (1996) J. Biol. Chem. 271, 22791-22795). In more recent studies, Cabral et al. (Cabral, C. M., Choudhury, P., Liu, Y., and Sifers, R. N. (2000) J. Biol. Chem. 275, 25015-25022) found that degradation of alpha(1)-ATZ in a stable transfected murine hepatoma cell line was inhibited by tyrosine phosphatase inhibitors, but not by the proteasomal inhibitor lactacystin and concluded that the proteasome was only involved in ER degradation of alpha(1)-ATZ in nonhepatocytic cell types or in cell types with levels of alpha(1)-AT expression that are substantial lower than that which occurs in hepatocytes. To examine this important issue in further detail, in this study we established rat and murine hepatoma cell lines with constitutive and inducible expression of alpha(1)-ATZ. In each of these cell lines degradation of alpha(1)-ATZ was inhibited by lactacystin, MG132, epoxomicin, and clasto-lactacystin beta-lactone. Using the inducible expression system to regulate the relative level of alpha(1)-ATZ expression, we found that lactacystin had a similar inhibitory effect on degradation of alpha(1)-ATZ at high and low levels of alpha(1)-AT expression. Although there is substantial evidence that other mechanisms contribute to ER degradation of alpha(1)-ATZ, the data reported here indicate that the proteasome plays an important role in many cell types including hepatocytes.

    Topics: Acetylcysteine; alpha 1-Antitrypsin; Animals; Antibiotics, Antineoplastic; Carcinoma, Hepatocellular; Cell Line; Cells, Cultured; Cysteine Endopeptidases; Electrophoresis, Polyacrylamide Gel; Endoplasmic Reticulum; Fibroblasts; HeLa Cells; Hepatocytes; Humans; Lactones; Leupeptins; Liver; Mice; Multienzyme Complexes; Mutation; Oligopeptides; Precipitin Tests; Proteasome Endopeptidase Complex; Protein Binding; Rats; Time Factors; Transfection; Tumor Cells, Cultured

2001
Ubiquitin is part of the retrovirus budding machinery.
    Proceedings of the National Academy of Sciences of the United States of America, 2000, Nov-21, Volume: 97, Issue:24

    Retroviruses contain relatively large amounts of ubiquitin, but the significance of this finding has been unknown. Here, we show that drugs that are known to reduce the level of free ubiquitin in the cell dramatically reduced the release of Rous sarcoma virus, an avian retrovirus. This effect was suppressed by overexpressing ubiquitin and also by directly fusing ubiquitin to the C terminus of Gag, the viral protein that directs budding and particle release. The block to budding was found to be at the plasma membrane, and electron microscopy revealed that the reduced level of ubiquitin results in a failure of mature virus particles to separate from each other and from the plasma membrane during budding. These data indicate that ubiquitin is actually part of the budding machinery.

    Topics: Acetylcysteine; Animals; Avian Sarcoma Viruses; Cell Line; Cell Membrane; Cysteine Endopeptidases; Endocytosis; Gene Products, gag; Leupeptins; Multienzyme Complexes; Oligopeptides; Protease Inhibitors; Proteasome Endopeptidase Complex; Quail; Recombinant Fusion Proteins; Ubiquitins; Virus Replication

2000