naphthoquinones and Neuroblastoma

naphthoquinones has been researched along with Neuroblastoma* in 19 studies

Other Studies

19 other study(ies) available for naphthoquinones and Neuroblastoma

ArticleYear
The DNMT3B Inhibitor Nanaomycin A as a Neuroblastoma Therapeutic Agent.
    Current cancer drug targets, 2023, Volume: 23, Issue:11

    Neuroblastoma is one of the most common childhood solid tumors. Because tumor suppressor genes are often hypermethylated in cancers, DNA methylation has emerged as a target for cancer therapeutics. Nanaomycin A, an inhibitor of DNA methyltransferase 3B, which mediates. To study the antitumor activity of nanaomycin A against neuroblastoma cell lines and its mechanism.. The anti-tumor effect of nanaomycin A on neuroblastoma cell lines was evaluated based on cell viability, DNA methylation levels, apoptosis-related protein expression, and neuronal-associated mRNA expression.. Nanaomycin A decreased genomic DNA methylation levels and induced apoptosis in human neuroblastoma cells. Nanaomycin A also upregulated the expression of mRNAs for several genes related to neuronal maturation.. Nanaomycin A is an effective therapeutic candidate for treating neuroblastoma. Our findings also suggest that the inhibition of DNA methylation is a promising anti-tumor therapy strategy for neuroblastoma.

    Topics: Cell Line, Tumor; Child; DNA (Cytosine-5-)-Methyltransferases; DNA Methylation; DNA Methyltransferase 3B; Humans; Naphthoquinones; Neuroblastoma

2023
Synthesis of thia-Michael-Type Adducts between Naphthoquinones and
    Molecules (Basel, Switzerland), 2022, Sep-01, Volume: 27, Issue:17

    Topics: Acetylcysteine; Cell Line, Tumor; HeLa Cells; Humans; Naphthoquinones; Neuroblastoma; Reactive Oxygen Species

2022
Synthetic 1,4-Naphthoquinones inhibit P2X7 receptors in murine neuroblastoma cells.
    Bioorganic & medicinal chemistry, 2021, 02-01, Volume: 31

    The P2X7 receptor (P2X7R) is an ATP-gated ion channel and potential therapeutic target for new drug development. In this study, we synthesized a series of new 1,4-naphthoquinone (1,4-NQ) derivatives and investigated their antagonistic effects against mouse P2X7R. We explored the ability of the tested substances to block ATP-induced Ca

    Topics: Animals; Antineoplastic Agents; Apoptosis; Cell Cycle Checkpoints; Cell Proliferation; Chromobacterium; Dose-Response Relationship, Drug; Drug Screening Assays, Antitumor; Mice; Models, Molecular; Molecular Structure; Naphthoquinones; Neuroblastoma; Purinergic P2X Receptor Antagonists; Receptors, Purinergic P2X7; Structure-Activity Relationship; Tumor Cells, Cultured

2021
Synthesis, Cytotoxic Activity Evaluation and Quantitative Structure-Activity Analysis of Substituted 5,8-Dihydroxy-1,4-Naphthoquinones and their
    Marine drugs, 2020, Nov-29, Volume: 18, Issue:12

    Based on 6,7-substituted 2,5,8-trihydroxy-1,4-naphtoquinones (1,4-NQs) derived from sea urchins, five new acetyl-

    Topics: Animals; Antineoplastic Agents; Cell Line, Tumor; Cell Survival; Dose-Response Relationship, Drug; Glycosides; Inhibitory Concentration 50; Mice; Molecular Structure; Naphthoquinones; Neuroblastoma; Quantitative Structure-Activity Relationship; Sea Urchins

2020
Anti-GD2 Immunoliposomes for Targeted Delivery of the Survivin Inhibitor Sepantronium Bromide (YM155) to Neuroblastoma Tumor Cells.
    Pharmaceutical research, 2018, Mar-07, Volume: 35, Issue:4

    Sepantronium bromide (YM155) is a hydrophilic quaternary compound that cannot be administered orally due to its low oral bioavailability; it is furthermore rapidly eliminated via the kidneys. The current study aims at improving the pharmacokinetic profile of YM155 by its formulation in immunoliposomes that can achieve its enhanced delivery into tumor tissue and facilitate uptake in neuroblastoma cancer cells.. PEGylated YM155 loaded liposomes composed of DPPC, cholesterol and DSPE-PEG. YM155 loaded immunoliposomes had a size of 170 nm and zeta potential of -10 mV, with an antibody coupling efficiency of 60% andYM155 encapsulation efficiency of14%. Targeted and control liposomal formulations were found to have similar YM155 release rates in a release medium containing 50% serum. An in-vitro toxicity study on KCNR cells showed less toxicity for immunoliposomes as compared to free YM155. In-vivo pharmacokinetic evaluation of YM155 liposomes showed prolonged blood circulation and significantly increased half-lives of liposomal YM155 in tumor tissue, as compared to a bolus injection of free YM155.. YM155 loaded immunoliposomes were successfully formulated and characterized, and initial in-vivo results show their potential for improving the circulation time and tumor accumulation of YM155.

    Topics: Animals; Antibodies; Antineoplastic Agents; Cell Line, Tumor; Drug Compounding; Drug Liberation; Drug Stability; Female; Gangliosides; Half-Life; Humans; Hydrophobic and Hydrophilic Interactions; Imidazoles; Injections, Intravenous; Liposomes; Mice; Mice, Nude; Naphthoquinones; Neuroblastoma; Pilot Projects; Polyethylene Glycols; Survivin; Xenograft Model Antitumor Assays

2018
Lapatinib potentiates cytotoxicity of  YM155 in neuroblastoma via inhibition of the ABCB1 efflux transporter.
    Scientific reports, 2017, 06-08, Volume: 7, Issue:1

    Adverse side effects of cancer agents are of great concern in the context of childhood tumors where they can reduce the quality of life in young patients and cause life-long adverse effects. Synergistic drug combinations can lessen potential toxic side effects through lower dosing and simultaneously help to overcome drug resistance. Neuroblastoma is the most common cancer in infancy and extremely heterogeneous in clinical presentation and features. Applying a systematic pairwise drug combination screen we observed a highly potent synergy in neuroblastoma cells between the EGFR kinase inhibitor lapatinib and the anticancer compound YM155 that is preserved across several neuroblastoma variants. Mechanistically, the synergy was based on a lapatinib induced inhibition of the multidrug-resistance efflux transporter ABCB1, which is frequently expressed in resistant neuroblastoma cells, which allowed prolonged and elevated cytotoxicity of YM155. In addition, the drug combination (i.e. lapatinib plus YM155) decreased neuroblastoma tumor size in an in vivo model.

    Topics: Animals; Antineoplastic Agents; Apoptosis; ATP Binding Cassette Transporter, Subfamily B, Member 1; Cell Line, Tumor; Drug Synergism; Humans; Imidazoles; Lapatinib; N-Myc Proto-Oncogene Protein; Naphthoquinones; Neuroblastoma; Protein Kinase Inhibitors; Receptor, trkA; RNA Interference; Zebrafish

2017
Suppressive effect of topoisomerase inhibitors on JC polyomavirus propagation in human neuroblastoma cells.
    Microbiology and immunology, 2016, Volume: 60, Issue:4

    JC polyomavirus (JCPyV) causes progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease of the central nervous system, in immunocompromised patients. Because no drugs have been approved for treating PML, many antiviral agents are currently being investigated for this purpose. The inhibitory effects of the topoisomerase I inhibitors topotecan and β-lapachone were assessed by investigating viral replication, propagation and viral protein 1 (VP1) production in cultured cells. JCPyV replication was assayed using the human neuroblastoma cell line IMR-32 transfected with the JCPyV plasmid and RT- PCR combined with Dpn I treatment. Dpn I digests the input plasmid DNA containing methylated adenosine, but not newly replicated JCPyV DNA, in IMR-32 cells. It was found that JCPyV replicates less in IMR-32 cells treated with topotecan or β-lapachone than in untreated cells. Moreover, drug treatment of JCI cells, which are IMR-32 cells persistently infected with JCPyV, led to a reduction in the amount of JCPyV DNA and population of VP1-positive cells. These results demonstrate that topotecan and β-lapachone affects JCPyV propagation in human neuroblastoma cell lines, suggesting that topotecan and β-lapachone could potentially be used to treat PML.

    Topics: Antiviral Agents; Cell Line; Cell Proliferation; DNA Replication; DNA, Viral; Humans; JC Virus; Leukoencephalopathy, Progressive Multifocal; Naphthoquinones; Neuroblastoma; Topoisomerase I Inhibitors; Topoisomerase Inhibitors; Topotecan; Virus Replication

2016
Effects of YM155 on survivin levels and viability in neuroblastoma cells with acquired drug resistance.
    Cell death & disease, 2016, 10-13, Volume: 7, Issue:10

    Resistance formation after initial therapy response (acquired resistance) is common in high-risk neuroblastoma patients. YM155 is a drug candidate that was introduced as a survivin suppressant. This mechanism was later challenged, and DNA damage induction and Mcl-1 depletion were suggested instead. Here we investigated the efficacy and mechanism of action of YM155 in neuroblastoma cells with acquired drug resistance. The efficacy of YM155 was determined in neuroblastoma cell lines and their sublines with acquired resistance to clinically relevant drugs. Survivin levels, Mcl-1 levels, and DNA damage formation were determined in response to YM155. RNAi-mediated depletion of survivin, Mcl-1, and p53 was performed to investigate their roles during YM155 treatment. Clinical YM155 concentrations affected the viability of drug-resistant neuroblastoma cells through survivin depletion and p53 activation. MDM2 inhibitor-induced p53 activation further enhanced YM155 activity. Loss of p53 function generally affected anti-neuroblastoma approaches targeting survivin. Upregulation of ABCB1 (causes YM155 efflux) and downregulation of SLC35F2 (causes YM155 uptake) mediated YM155-specific resistance. YM155-adapted cells displayed increased ABCB1 levels, decreased SLC35F2 levels, and a p53 mutation. YM155-adapted neuroblastoma cells were also characterized by decreased sensitivity to RNAi-mediated survivin depletion, further confirming survivin as a critical YM155 target in neuroblastoma. In conclusion, YM155 targets survivin in neuroblastoma. Furthermore, survivin is a promising therapeutic target for p53 wild-type neuroblastomas after resistance acquisition (neuroblastomas are rarely p53-mutated), potentially in combination with p53 activators. In addition, we show that the adaptation of cancer cells to molecular-targeted anticancer drugs is an effective strategy to elucidate a drug's mechanism of action.

    Topics: ATP Binding Cassette Transporter, Subfamily B; Cell Line, Tumor; Cell Survival; DNA Damage; Drug Resistance, Neoplasm; Humans; Imidazoles; Inhibitor of Apoptosis Proteins; Kinetics; Membrane Transport Proteins; Mutation; Naphthoquinones; Neuroblastoma; Piperazines; Proto-Oncogene Proteins c-mdm2; RNA, Small Interfering; Survivin; Tumor Suppressor Protein p53

2016
The BET bromodomain inhibitor exerts the most potent synergistic anticancer effects with quinone-containing compounds and anti-microtubule drugs.
    Oncotarget, 2016, Nov-29, Volume: 7, Issue:48

    BET bromodomain inhibitors are very promising novel anticancer agents, however, single therapy does not cause tumor regression in mice, suggesting the need for combination therapy. After screening a library of 2697 small molecule compounds, we found that two classes of compounds, the quinone-containing compounds such as nanaomycin and anti-microtubule drugs such as vincristine, exerted the best synergistic anticancer effects with the BET bromodomain inhibitor JQ1 in neuroblastoma cells. Mechanistically, the quinone-containing compound nanaomycin induced neuroblastoma cell death but also activated the Nrf2-antioxidant signaling pathway, and the BET bromodomain proteins BRD3 and BRD4 formed a protein complex with Nrf2. Treatment with JQ1 blocked the recruitment of Nrf2 to the antioxidant responsive elements at Nrf2 target gene promoters, and JQ1 exerted synergistic anticancer effects with nanaomycin by blocking the Nrf2-antioxidant signaling pathway. JQ1 and vincristine synergistically induced neuroblastoma cell cycle arrest at the G2/M phase, aberrant mitotic spindle assembly formation and apoptosis, but showed no effect on cell survival in normal non-malignant cells. Importantly, co-treatment with JQ1 and vincristine synergistically suppressed tumor progression in neuroblastoma-bearing mice. These results strongly suggest that patients treated with BET bromodomain inhibitors in clinical trials should be co-treated with vincristine.

    Topics: Animals; Antineoplastic Agents; Azepines; Cell Cycle Proteins; Cell Line, Tumor; Cell Proliferation; Cell Survival; Drug Synergism; Gene Expression Regulation, Neoplastic; Humans; Mice; Naphthoquinones; Neuroblastoma; NF-E2-Related Factor 2; Nuclear Proteins; Protein Domains; RNA-Binding Proteins; Small Molecule Libraries; Transcription Factors; Triazoles; Tubulin Modulators; Vincristine; Xenograft Model Antitumor Assays

2016
A rapid screening system evaluates novel inhibitors of DNA methylation and suggests F-box proteins as potential therapeutic targets for high-risk neuroblastoma.
    Targeted oncology, 2015, Volume: 10, Issue:4

    After extensive research on radiochemotherapy, 5-year survival rates of children with high risk neuroblastoma still do not exceed 50%, owing to adverse side-effects exemplified by doxorubicin-induced cardiomyopathy. A promising new approach is the combination of conventional therapies with specific modulation of cell signaling pathways promoting therapeutic resistance, such as inhibition of aberrant kinase activity or re-expression of silenced tumor suppressor genes by means of chromatin remodeling. In this regard, we established a system that allows to identify potential drug targets as well as to validate respective candidate inhibitors in high-risk neuroblastoma model cell lines. Cell culture, drug exposure, shRNA-mediated knockdown and phenotype analysis are integrated into an efficient and versatile single well-based protocol. By utilizing this system, we assessed RG108, SGI-1027 and nanaomycin A, three novel DNA methyltransferase inhibitors that have not been tested in neuroblastoma cell lines so far, for their potential of synergistic anti-tumor activity in combination with doxorubicin. We found that, similarly to azacytidine, SGI-1027 and nanaomycin A mediate synergistic growth inhibition with doxorubicin independently of N-Myc status. However, they display high cytotoxicity but lack global DNA demethylation activity. Secondly, we conducted a lentiviral shRNA screen of F-box proteins, key regulators of protein stability, and identified Fbxw11/β-TrCP2 as well as Fbxo5/Emi1 as potential therapeutic targets in neuroblastoma. These results complement existing studies and underline the reliability and versatility of our single well-based protocol.

    Topics: Aminoquinolines; Antineoplastic Combined Chemotherapy Protocols; Cell Line, Tumor; Cell Proliferation; DNA Methylation; DNA Modification Methylases; Doxorubicin; Drug Synergism; Enzyme Inhibitors; F-Box Proteins; HEK293 Cells; Humans; Molecular Targeted Therapy; Naphthoquinones; Neuroblastoma; Phthalimides; Pyrimidines; RNA, Small Interfering; Tryptophan

2015
Pharmacological activation of Sirt1 ameliorates polyglutamine-induced toxicity through the regulation of autophagy.
    PloS one, 2013, Volume: 8, Issue:6

    Intracellular accumulation of polyglutamine (polyQ)-expanded Huntingtin (Htt) protein is a hallmark of Huntington's disease (HD). This study evaluated whether activation of Sirt1 by the anti-cancer agent, β-lapachone (β-lap), induces autophagy in human neuroblastoma SH-SY5Y cells, thereby reducing intracellular levels of polyQ aggregates and their concomitant cytotoxicity. Treatment of cells with β-lap markedly diminished the cytotoxicity induced by forced expression of Htt exon 1 containing a pathogenic polyQ stretch fused to green fluorescent protein (HttEx1(97Q)-GFP). β-lap increased autophagy in SH-SY5Y cells, as evidenced by the increased formation of LC3-II and autolysosomes. Furthermore, β-lap reduced HttEx1(97Q)-GFP aggregation, which was significantly prevented by co-incubation with 3-methyladenine, an inhibitor of autophagy. β-lap increased Sirt1 activity, as shown by the increased deacetylation of the Sirt1 substrates, PARP-1 and Atg5, and the nuclear translocation of FOXO1. Both the induction of autophagy and attenuation of HttEx1(97Q)-GFP aggregation by β-lap were significantly prevented by co-incubation with sirtinol, a general sirtuin inhibitor or by co-transfection with shRNA against Sirt1. The pro-autophagic actions of β-lap were further investigated in a transgenic Caenorhabditis elegans (C. elegans) line that expressed Q67 fused to cyanine fluorescent protein (Q67). Notably, β-lap reduced the number of Q67 puncta and restored Q67-induced defects in motility, which were largely prevented by pre-treatment with RNAi against sir-2.1, the C. elegans orthologue of Sirt1. Collectively, these data suggest that β-lap induces autophagy through activation of Sirt1, which in turn leads to a reduction in polyQ aggregation and cellular toxicity. Thus, β-lap provides a novel therapeutic opportunity for the treatment of HD.

    Topics: Adenine; Animals; Animals, Genetically Modified; Apoptosis; Autophagy; Blotting, Western; Caenorhabditis elegans; Cell Movement; Cell Proliferation; Green Fluorescent Proteins; Humans; Huntingtin Protein; Immunoenzyme Techniques; Immunoprecipitation; Microscopy, Fluorescence; Naphthoquinones; Nerve Tissue Proteins; Neuroblastoma; Peptides; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Inhibitors; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; RNA, Small Interfering; Sirtuin 1

2013
Inhibitory effects of LPA1 on cell motile activities stimulated by hydrogen peroxide and 2,3-dimethoxy-1,4-naphthoquinone in fibroblast 3T3 cells.
    Biochemical and biophysical research communications, 2013, Nov-08, Volume: 441, Issue:1

    Reactive oxygen species (ROS) are known to mediate a variety of biological responses, including cell motility. Recently, we indicated that lysophosphatidic acid (LPA) receptor-3 (LPA3) increased cell motile activity stimulated by hydrogen peroxide. In the present study, we assessed the role of LPA1 in the cell motile activity mediated by ROS in mouse fibroblast 3T3 cells. 3T3 cells were treated with hydrogen peroxide and 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) at concentrations of 0.1 and 1 μM for 48 h. In cell motility assays with Cell Culture Inserts, the cell motile activities of 3T3 cells treated with hydrogen peroxide and DMNQ were significantly higher than those of untreated cells. 3T3 cells treated with hydrogen peroxide and DMNQ showed elevated expression levels of the Lpar3 gene, but not the Lpar1 and Lpar2 genes. To investigate the effects of LPA1 on the cell motile activity induced by hydrogen peroxide and DMNQ, Lpar1-overexpressing (3T3-a1) cells were generated from 3T3 cells and treated with hydrogen peroxide and DMNQ. The cell motile activities stimulated by hydrogen peroxide and DMNQ were markedly suppressed in 3T3-a1 cells. These results suggest that LPA signaling via LPA1 inhibits the cell motile activities stimulated by hydrogen peroxide and DMNQ in 3T3 cells.

    Topics: 3T3 Cells; Animals; Cell Line, Tumor; Cell Movement; Cell Proliferation; Diphosphates; Fibroblasts; Gene Expression Regulation; Glycerol; Hydrogen Peroxide; Lysophospholipids; Mice; Naphthoquinones; Neuroblastoma; Rats; Receptors, Lysophosphatidic Acid

2013
Silencing of survivin using YM155 induces apoptosis and chemosensitization in neuroblastomas cells.
    European review for medical and pharmacological sciences, 2013, Volume: 17, Issue:21

    Aggressive cell growth and chemoresistance are notorious obstacles in neuroblastoma therapy. Accumulating evidence suggests that survivin is preferentially expressed in cancer cells and plays a crucial role in cell division and apoptosis dysfunction. Thus, in the present study, we investigated whether silencing of survivin, using a novel small-molecule survivin suppressant, YM155 could suppress the proliferation and induce chemosensitization of neuroblastoma cells.. SH-SY5Y human neuroblastomas cells were treated with YM155 (10 to 500 mM) and/or chemotherapeutic agent cisplatin for 72 hours, and cell viability, apoptosis, mRNA and protein expression level were then evaluated. Furthermore, the efficacy of YM155 combined with cisplatin was further examined in established xenograft models.. YM155 suppressed expression of survivin, inhibited the proliferation and induced apoptosis in SH-SY5Y cells in a concentration-dependent manner. Reduced levels of survivin sensitized SH-SY5Y to the chemotherapeutic agent cisplatin. YM155 showed antiproliferative effects and induced tumor regression and apoptosis in established SH-SY5Y xenograft models. Cisplatin showed antitumor activity against SH-SY5Y cells, it did not induce survivin upregulation. Combination treatment of YM155 and cisplatin induced a greater rate of apoptosis than the sum of the single-treatment rates and promoted tumor regression without enhanced body weight loss in the SH-SY5Y xenograft models.. The concomitant combination of YM155 with cisplatin induced more intense apoptosis compared with each single treatment in vivo and in vitro. YM155 in combination with cisplatin is well tolerated and shows greater efficacy than either agent alone in mouse xenograft models.

    Topics: Animals; Antineoplastic Combined Chemotherapy Protocols; Apoptosis; Cell Line, Tumor; Cell Proliferation; Cell Survival; Cisplatin; Dose-Response Relationship, Drug; Humans; Imidazoles; Inhibitor of Apoptosis Proteins; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Naphthoquinones; Neuroblastoma; RNA, Messenger; Survivin; Xenograft Model Antitumor Assays

2013
Targeted BIRC5 silencing using YM155 causes cell death in neuroblastoma cells with low ABCB1 expression.
    European journal of cancer (Oxford, England : 1990), 2012, Volume: 48, Issue:5

    The BIRC5 (Survivin) gene is located at chromosome 17q in the region that is frequently gained in high risk neuroblastoma. BIRC5 is strongly over expressed in neuroblastoma tumour samples, which correlates to a poor prognosis. We recently validated BIRC5 as a potential therapeutic target by showing that targeted knock down with shRNA's triggers an apoptotic response through mitotic catastrophe. We now tested YM155, a novel small molecule selective BIRC5 suppressant that is currently in phase I/II clinical trials. Drug response curves showed IC50 values in the low nM range (median: 35 nM, range: 0.5-> 10,000 nM) in a panel of 23 neuroblastoma cell lines and four TIC-lines, which resulted from an apoptotic response. Nine out of 23 cell lines were relatively resistant to YM155 with IC50 values > 200 nM, although in the same cells shRNA mediated knock down of BIRC5 caused massive apoptosis. Analysis of differentially expressed genes between five most sensitive and five most resistant cell lines using Affymetrix mRNA expression data revealed ABCB1 (MDR1) as the most predictive gene for resistance to YM155. Inhibition of the multi-drug resistance pump ABCB1 with cyclosporine or knockdown with shRNA prior to treatment with YM155 demonstrated that cell lines with ABCB1 expression became 27-695 times more sensitive to YM155 treatment. We conclude that most neuroblastoma cell lines are sensitive to YM155 in the low nM range and that resistant cells can be sensitised by ABCB1 inhibitors. Therefore YM155 is a promising novel compound for treatment of neuroblastoma with low ABCB1 expression.

    Topics: Apoptosis; ATP Binding Cassette Transporter, Subfamily B; ATP Binding Cassette Transporter, Subfamily B, Member 1; Cell Line, Tumor; Drug Resistance, Neoplasm; Gene Silencing; HEK293 Cells; Humans; Imidazoles; Inhibitor of Apoptosis Proteins; Naphthoquinones; Neuroblastoma; RNA, Small Interfering; Survivin; Xenograft Model Antitumor Assays

2012
γ-Aminobutyric acid type A (GABAA) receptor activation modulates tau phosphorylation.
    The Journal of biological chemistry, 2012, Feb-24, Volume: 287, Issue:9

    Abnormal phosphorylation and aggregation of the microtubule-associated protein Tau are hallmarks of various neurodegenerative diseases, such as Alzheimer disease. Molecular mechanisms that regulate Tau phosphorylation are complex and currently incompletely understood. We have developed a novel live cell reporter system based on protein-fragment complementation assay to study dynamic changes in Tau phosphorylation status. In this assay, fusion proteins of Tau and Pin1 (peptidyl-prolyl cis-trans-isomerase 1) carrying complementary fragments of a luciferase protein serve as a sensor of altered protein-protein interaction between Tau and Pin1, a critical regulator of Tau dephosphorylation at several disease-associated proline-directed phosphorylation sites. Using this system, we identified several structurally distinct GABA(A) receptor modulators as novel regulators of Tau phosphorylation in a chemical library screen. GABA(A) receptor activation promoted specific phosphorylation of Tau at the AT8 epitope (Ser-199/Ser-202/Thr-205) in cultures of mature cortical neurons. Increased Tau phosphorylation by GABA(A) receptor activity was associated with reduced Tau binding to protein phosphatase 2A and was dependent on Cdk5 but not GSK3β kinase activity.

    Topics: Alzheimer Disease; Animals; Cell Line, Tumor; Cyclin-Dependent Kinase 5; Cytoskeleton; Enzyme Inhibitors; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Humans; Marine Toxins; Mice; Naphthoquinones; Nerve Degeneration; Neuroblastoma; NIMA-Interacting Peptidylprolyl Isomerase; Oxazoles; Peptidylprolyl Isomerase; Phosphorylation; Protein Kinase Inhibitors; Protein Phosphatase 2; Purines; Rats; Receptors, GABA-A; Roscovitine; tau Proteins; Tauopathies

2012
Evidence for synergism between cell death mechanisms in a cellular model of neurodegeneration in Parkinson's disease.
    Neurotoxicity research, 2012, Volume: 22, Issue:4

    Delineation of how cell death mechanisms associated with Parkinson's disease (PD) interact and whether they converge would help identify targets for neuroprotective therapies. The purpose of this study was to use a cellular model to address these issues. Catecholaminergic SH-SY5Y neuroblastoma cells were exposed to a range of compounds (dopamine, rotenone, 5,8-dihydroxy-1,4-naphtho-107 quinone [naphthazarin], and Z-Ile-Glu(OBut)-Ala-Leu-al [PSI]) that are neurotoxic when applied to these cells for extended periods of times at specific concentrations. At the concentrations used, these compounds cause cellular stress via mechanisms that mimic those associated with causing neurodegeneration in PD, namely oxidative stress (dopamine), mitochondrial dysfunction (rotenone), lysosomal dysfunction (naphthazarin), and proteasomal dysfunction (PSI). The compounds were applied to the SH-SY5Y cells either alone or in pairs. When applied separately, the compounds produced a significant decrease in cell viability confirming that oxidative stress, mitochondrial, proteosomal, or lysosomal dysfunction can individually result in catecholaminergic cell death. When the compounds were applied in pairs, some of the combinations produced synergistic effects. Analysis of these interactions indicates that proteasomal, lysosomal, and mitochondrial dysfunction is exacerbated by dopamine-induced oxidative stress. Furthermore, inhibition of the proteasome or lysosome or increasing oxidative stress has a synergistic effect on cell viability when combined with mitochondrial dysfunction, suggesting that all cell death mechanisms impair mitochondrial function. Finally, we show that there are reciprocal relationships between oxidative stress, proteasomal dysfunction, and mitochondrial dysfunction, whereas lysosome dysfunction appears to mediate cell death via an independent pathway. Given the highly interactive nature of the various cell death mechanisms linked with PD, we predict that effective neuroprotective strategies should target multiple sites in these pathways, for example oxidative stress and mitochondria.

    Topics: Antineoplastic Agents; Apoptosis; Cell Line, Tumor; Cell Survival; Dopamine; Dose-Response Relationship, Drug; Drug Synergism; Humans; Lysosomes; Mitochondria; Naphthoquinones; Neuroblastoma; Neurotoxins; Proteasome Endopeptidase Complex; Rotenone

2012
Plumbagin, a novel Nrf2/ARE activator, protects against cerebral ischemia.
    Journal of neurochemistry, 2010, Volume: 112, Issue:5

    Many phytochemicals function as noxious agents that protect plants against insects and other damaging organisms. However, at subtoxic doses, the same phytochemicals may activate adaptive cellular stress response pathways that can protect cells against a variety of adverse conditions. We screened a panel of botanical pesticides using cultured human and rodent neuronal cell models, and identified plumbagin as a novel potent activator of the nuclear factor E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway. In vitro, plumbagin increases nuclear localization and transcriptional activity of Nrf2, and induces the expression of the Nrf2/ARE-dependent genes, such as heme oxygenase 1 in human neuroblastoma cells. Plumbagin specifically activates the Nrf2/ARE pathway in primary mixed cultures from ARE-human placental alkaline phosphatase reporter mice. Exposure of neuroblastoma cells and primary cortical neurons to plumbagin provides protection against subsequent oxidative and metabolic insults. The neuroprotective effects of plumbagin are abolished by RNA interference-mediated knockdown of Nrf2 expression. In vivo, administration of plumbagin significantly reduces the amount of brain damage and ameliorates-associated neurological deficits in a mouse model of focal ischemic stroke. Our findings establish precedence for the identification and characterization of neuroprotective phytochemicals based upon their ability to activate adaptive cellular stress response pathways.

    Topics: Animals; Cell Line, Tumor; Cell Survival; Cells, Cultured; Cerebral Cortex; Cerebral Infarction; Disease Models, Animal; Embryo, Mammalian; Gene Expression Regulation; Glucose; Heme Oxygenase-1; Humans; Hypoxia; Infarction, Middle Cerebral Artery; Mice; Mice, Inbred C57BL; Naphthoquinones; Neuroblastoma; Neurologic Examination; Neurons; Neuroprotective Agents; NF-E2-Related Factor 2; Oxidative Stress; Rats; Rats, Sprague-Dawley; Transcription Factor AP-1; Transfection

2010
Effects of naturally occurring compounds on fibril formation and oxidative stress of beta-amyloid.
    Journal of agricultural and food chemistry, 2005, Nov-02, Volume: 53, Issue:22

    Beta-amyloid (betaA)-induced oxidative toxicity on neuronal cells is a principal route in Alzheimer's disease (AD), and its toxicity occurs after fibril formation. Inhibitory or promoting effects of naturally occurring compounds on betaA fibril formation were evaluated. Among 214 tested compounds, curcuminoids, flavone type flavonoids, and naphthoquinones were shown to be potent inhibitors of betaA fibrilization. The addition of the curcuminoids, curcumin, demethoxycurcumin, and bisdemethoxycurcumin strongly inhibited betaA fibril formation. Flavonoids such as quercetin, rhamnetin, and fisetin strongly inhibited betaA fibril formation. Limonoids, cinnamic acids, and catechins enhanced fibril formation in vitro. Anthothecol possessed the most enhancing activity on fibril formation of the compounds tested. On the other hand, it was found that curcuminoids showed cytotoxicity with the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide assay and did not protect HT22 murine neuroblastoma cells from betaA(25-35) insult. Two flavone type flavonoids, morin and quercetin, exhibited no cytotoxicity and strongly protected HT22 murine neuroblastoma cells from betaA(25-35) oxidative attack. Conclusively, morin or quercetin could be a key molecule for the development of therapeutics for AD.

    Topics: Amyloid beta-Peptides; Animals; Cell Line, Tumor; Curcumin; Flavonoids; Mice; Naphthoquinones; Neuroblastoma; Oxidative Stress

2005
Pin1: a therapeutic target in Alzheimer neurodegeneration.
    Journal of molecular neuroscience : MN, 2002, Volume: 19, Issue:3

    In Alzheimer's disease, the peptidyl prolyl cis/trans isomerase Pin1 binds to phospho-Thr231 on Tau proteins and, hence, is found within degenerating neurons, where it is associated to the large amounts of abnormally phosphorylated Tau proteins. Conversely, Pin1 may restore the tubulin polymerization function of these hyperphosphorylated Tau. In the present work, we investigated, both at the cellular and molecular levels, the role of Pin1 in Alzheimer's disease through the study of its interactions with phosphorylated Tau proteins. We also showed that in neuronal cells, Pin1 upregulates the expression of cyclin D1. This, in turn, could facilitate the transition from quiescence to the G1 phase (re-entry in cell cycle) in a neuron and, subsequently, neuronal dedifferentiation and apoptosis. The involvement of Pin1 in the G0/G1 transition in neurons points to its function as a good target for the development of new therapeutic strategies in neurodegenerative disorders.

    Topics: Alzheimer Disease; Cell Line; Cyclin D1; Humans; Magnetic Resonance Spectroscopy; Models, Molecular; Naphthoquinones; Neuroblastoma; Neurons; NIMA-Interacting Peptidylprolyl Isomerase; Peptidylprolyl Isomerase; Phosphopyruvate Hydratase; Phosphorylation; Protein Binding; Protein Structure, Tertiary; Spectrometry, Fluorescence; tau Proteins

2002