Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Int J Mol Sci ; 22(11)2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34199510

ABSTRACT

During aggressive cancer progression, cancer cells adapt to unique microenvironments by withstanding various cellular stresses, including endoplasmic reticulum (ER) stress. However, the mechanism whereby cancer cells overcome the ER stress to survive remains to be elucidated. Herein, we demonstrated that microtubule acetylation in cancer cells grown on a stiff matrix promotes cancer progression by preventing excessive ER stress. Downregulation of microtubule acetylation using shRNA or CRSIPR/Cas9 techniques targeting ATAT1, which encodes α-tubulin N-acetyltransferase (αTAT1), resulted in the upregulation of ER stress markers, changes in ER morphology, and enhanced tunicamycin-induced UPR signaling in cancer cells. A set of genes involved in cancer progression, especially focal adhesion genes, were downregulated in both ATAT1-knockout and tunicamycin-treated cells, whereas ATAT1 overexpression restored the gene expression inhibited by tunicamycin. Finally, the expression of ATAT1 and ER stress marker genes were negatively correlated in various breast cancer types. Taken together, our results suggest that disruption of microtubule acetylation is a potent therapeutic tool for preventing breast cancer progression through the upregulation of ER stress. Moreover, ATAT1 and ER stress marker genes may be useful diagnostic markers in various breast cancer types.


Subject(s)
Acetyltransferases/genetics , Breast Neoplasms/genetics , Endoplasmic Reticulum Stress/genetics , Microtubule Proteins/genetics , Tunicamycin/pharmacology , Acetylation/drug effects , Acetyltransferases/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Microtubule Proteins/antagonists & inhibitors , Microtubules/drug effects , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Tumor Microenvironment/drug effects
2.
Comput Biol Chem ; 93: 107513, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34052673

ABSTRACT

Post-translation modification of microtubules is associated with many diseases like cancer. Alpha Tubulin Acetyltransferase 1 (ATAT1) is a major enzyme that acetylates 'Lys-40' in alpha-tubulin on the luminal side of microtubules and is a drug target that lacks inhibitors. Here, we developed pharmacophore anchor models of ATAT1 which were constructed statistically using thousands of docked compounds, for drug design and investigating binding mechanisms. Our models infer the compound moiety preferences with the physico-chemical properties for the ATAT1 binding site. The results from the pharmacophore anchor models show the three main sub-pockets, including S1 acetyl site, S2 adenine site, and S3 diphosphate site with anchors, where conserved moieties interact with respective sub-pocket residues in each site and help in guiding inhibitor discovery. We validated these key anchors by analyzing 162 homologous protein sequences (>99 species) and over 10 structures with various bound ligands and mutations. Our results were consistent with previous works also providing new interesting insights. Our models applied in virtual screening predicted several ATAT1 potential inhibitors. We believe that our model is useful for future inhibitor discovery and for guiding lead optimization.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Microtubule Proteins/antagonists & inhibitors , Molecular Docking Simulation , Acetyltransferases/genetics , Acetyltransferases/metabolism , Enzyme Inhibitors/chemistry , Humans , Ligands , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Mutation , Protein Processing, Post-Translational
3.
BMB Rep ; 52(5): 342-347, 2019 May.
Article in English | MEDLINE | ID: mdl-31068247

ABSTRACT

Methylation is a primary epigenetic mechanism regulating gene expression. 5-aza-2'-deoxycytidine is an FDA-approved drug prescribed for treatment of cancer by inhibiting DNA-Methyl-Transferase 1 (DNMT1). Results of this study suggest that prolonged treatment with 5-aza-2'-deoxycytidine could induce centrosome abnormalities in cancer cells and that CEP131, a centrosome protein, is regulated by DNMT1. Interestingly, cancer cell growth was attenuated in vitro and in vivo by inhibiting the expression of Cep131. Finally, Cep131-deficient cells were more sensitive to treatment with DNMT1 inhibitors. These findings suggest that Cep131 is a potential novel anti-cancer target. Agents that can inhibit this protein may be useful alone or in combination with DNMT1 inhibitors to treat cancer. [BMB Reports 2019; 52(5): 342-347].


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , Decitabine/pharmacology , Microtubule Proteins/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cytoskeletal Proteins , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/drug effects , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Epigenesis, Genetic , Female , HEK293 Cells , HeLa Cells , Humans , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
4.
DNA Cell Biol ; 38(5): 476-484, 2019 May.
Article in English | MEDLINE | ID: mdl-30835546

ABSTRACT

Recently, sperm-associated antigen 6 (SPAG6), a member of the cancer-testis antigen family, has been shown to be involved in tumorigenesis. An increasing number of studies have shown that SPAG6 expression is associated with the pathogenesis of myelodysplastic syndrome (MDS). However, the mechanism has not been clearly elucidated. Our previous results indicated that SPAG6 affected cell apoptosis in MDS. In this study, we used reverse transcription-quantitative polymerase chain reaction (RT-qPCR) to demonstrate that the mRNA expression of SPAG6 in bone marrow cells of patients with MDS or MDS-derived acute myeloid leukemia (MDS-AML) was higher than that of cancer-free patients. Kaplan-Meier survival curve analysis of published AML found that patients with high expression of SPAG6 had poor survival. The results of the cell counting kit-8, FACS, RT-qPCR, and Western blotting assays indicated that SPAG6 knockdown in the SKM-1 cell line inhibited cell proliferation, and affected cell cycle and differentiation. Furthermore, we found that SPAG6 knockdown affected the proliferation of SKM-1 cells by mediating the G1-to-S transition of the cell cycle. Moreover, we demonstrated that the antiproliferative effect of SPAG6 knockdown was associated with the upregulation of the cyclin-dependent kinase inhibitor p27Kip1, and regulation of the AKT/FOXO pathway. These findings indicated that SPAG6 might be a potential therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Forkhead Box Protein O1/metabolism , Leukemia, Myeloid, Acute/pathology , Microtubule Proteins/metabolism , Myelodysplastic Syndromes/pathology , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Cycle , Cell Proliferation , Female , Follow-Up Studies , Forkhead Box Protein O1/genetics , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Microtubule Proteins/antagonists & inhibitors , Microtubule Proteins/genetics , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/genetics , Tumor Cells, Cultured
5.
Int J Oncol ; 53(1): 297-306, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29749435

ABSTRACT

Apoptosis is a multi-step mechanism of cell self­destruction for maintaining cellular homeostatic balance. Accumulating evidence indicates that abnormal apoptosis promotes the evolution and progression of myelodysplastic syndromes (MDS). As a novel cancer-testis antigen, sperm­associated antigen 6 (SPAG6) has been reported to regulate apoptosis through the tumor necrosis factor-related apoptosis-inducing ligand signaling pathway in the MDS cell line SKM­1. However, the mechanism of the intrinsic cell death pathway for apoptosis induction by SPAG6 silencing is unclear. In the present study, the in vitro effects of SPAG6 silencing were investigated in SKM­1 cells through extensive biochemical and molecular approaches. Western blotting and reverse transcription-quantitative polymerase chain reaction were used to detect the expression of SPAG6 and activation of PTEN/PI3K/AKT signal pathway. Additionally, SKM­1 cells transduced with SPAG6 short hairpin RNA (shRNA) lentivirus were treated with the phosphatidylionositol 3-kinase (PI3K) inhibitor LY294002 or pan caspase inhibitor z­VAD­fmk and the apoptosis rates were measured by flow cytometry, and the expressions of associated proteins were examined by western blot analysis. A mouse xenograft model was also used to further evaluate the effects of SPAG6 knockdown on inducing tumor apoptosis in vivo. Lentivirus-mediated knockdown of SPAG6 in SKM­1 cells increased phosphatase and tensin homolog (PTEN) expression and reduced protein kinase B (AKT) phosphorylation, which in turn resulted in cell apoptosis as evidenced by induced myeloid leukaemia cell differentiation protein Mcl­1 downregulation, cytochrome c release and increased caspase­9 expression. Consistently, the PI3K inhibitor LY294002 synergistically enhanced apoptosis of SKM­1 cells when co-administered with SPAG6 shRNA lentivirus. Furthermore, treatment with the pan caspase inhibitor z­VAD­fmk failed to prevent PTEN activation upon SPAG6 knockdown, suggesting that SPAG6-regulated PTEN expression was caspase activation-independent. In addition, SPAG6 knockdown was associated with DNMT1 downregulation, implying that SPAG6 may indirectly control PTEN expression via DNA methylation. Furthermore, tumor tissues from nonobese diabetic/severe combined immunodeficient mice inoculated with SPAG6-shRNA lentivirus pre-infected SKM­1 cells exhibited significantly elevated apoptosis in the extrinsic and intrinsic pathways. These results demonstrate that SPAG6 silencing induces PTEN expression to regulate apoptosis though the PI3K/AKT pathway, indicating that SPAG6 may be a potential therapeutic target for MDS.


Subject(s)
Microtubule Proteins/genetics , Myelodysplastic Syndromes/genetics , Neoplasm Proteins/genetics , PTEN Phosphohydrolase/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mice , Microtubule Proteins/antagonists & inhibitors , Myelodysplastic Syndromes/pathology , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/genetics , Xenograft Model Antitumor Assays
6.
J Pharmacol Exp Ther ; 364(1): 46-54, 2018 01.
Article in English | MEDLINE | ID: mdl-29061656

ABSTRACT

Although cancer cell genetic instability contributes to characteristics that mediate tumorigenicity, it also contributes to the tumor-selective toxicity of some chemotherapy drugs. This synthetic lethality can be enhanced by inhibitors of DNA repair. To exploit this potential Achilles heel, we tested the ability of a RAD51 inhibitor to potentiate the cytotoxicity of chemotherapy drugs. 2-(Benzylsulfonyl)-1-(1H-indol-3-yl)-1,2-dihydroisoquinoline (IBR2) inhibits RAD51-mediated DNA double-strand break repair but also enhances cytotoxicity of the Bcr-Abl inhibitor imatinib. The potential for synergy between IBR2 and more drugs was examined in vitro across a spectrum of cancer cell lines from various tissues. Cells were exposed to IBR2 simultaneously with inhibitors of receptor tyrosine kinases, DNA-damaging agents, or microtubule disruptors. IBR2, at concentrations that inhibited proliferation between 0% and 75%, enhanced toxicity by up to 80% of imatinib and regorafenib (targets RAF and kit); epidermal growth factor receptor inhibitors erlotinib, gefitinib, afatinib, and osimertinib; and vincristine, an inhibitor of microtubule function. However, IBR2 antagonized the action of olaparib, cisplatin, melphalan, and irinotecan. A vincristine-resistant squamous cell line was not cross resistant to imatinib, but IBR2 and another RAD51 inhibitor (B02) enhanced imatinib toxicity in this cell line, its HN-5a parent, and the colon cancer line HT-29 by up to 60% and much better than verapamil, a P-glycoprotein inhibitor (P < 0.05). Given the disparate agents the functions of which are enhanced by IBR2, the mechanisms of enhancement may be multimodal. Whether RAD51 is common to these mechanisms remains to be elucidated, but it provides the potential for selectivity to tumor cells.


Subject(s)
Cell Proliferation/drug effects , Indoles/administration & dosage , Microtubule Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Rad51 Recombinase/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Tetrahydroisoquinolines/administration & dosage , A549 Cells , Antineoplastic Agents/administration & dosage , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Drug Synergism , HEK293 Cells , HT29 Cells , Humans , K562 Cells , MCF-7 Cells , Microtubule Proteins/metabolism , Rad51 Recombinase/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
7.
Sci Rep ; 7(1): 13753, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29062069

ABSTRACT

The formation of paired helical filaments (PHF), which are composed of hyperphosphorylated Tau protein dissociating from microtubules, is one of the pathological hallmarks of Alzheimer's disease (AD) and other tauopathies. The most important phosphatase that is capable of dephosphorylating Tau at AD specific phospho-sites is protein phosphatase 2 A (PP2A). Here we show that resveratrol, a polyphenol, significantly induces PP2A activity and reduces Tau phosphorylation at PP2A-dependent epitopes. The increase in PP2A activity is caused by decreased expression of the MID1 ubiquitin ligase that mediates ubiquitin-specific modification and degradation of the catalytic subunit of PP2A when bound to microtubules. Interestingly, we further show that MID1 expression is elevated in AD tissue. Our data suggest a key role of MID1 in the pathology of AD and related tauopathies. Together with previous studies showing that resveratrol reduces ß-amyloid toxicity they also give evidence of a promising role for resveratrol in the prophylaxis and therapy of AD.


Subject(s)
Alzheimer Disease/drug therapy , Microtubule Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Protein Interaction Maps/drug effects , Protein Phosphatase 2/antagonists & inhibitors , Proteins/antagonists & inhibitors , Resveratrol/pharmacology , Transcription Factors/antagonists & inhibitors , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cells, Cultured , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , HEK293 Cells , Humans , Mice , Microtubule Proteins/metabolism , Microtubules/drug effects , Microtubules/metabolism , Neurofibrillary Tangles , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/metabolism , Phosphorylation , Protein Phosphatase 2/metabolism , Proteins/metabolism , Proteolysis , Transcription Factors/metabolism , Ubiquitin-Protein Ligases
8.
J Pharm Biomed Anal ; 128: 226-235, 2016 Sep 05.
Article in English | MEDLINE | ID: mdl-27281578

ABSTRACT

Antibody-drug conjugates (ADCs) play an increasingly important role for targeted cancer treatment. One class of ADCs has attracted particular interest in drug development. These ADCs employ a cleavable chemistry linkage for drugs and utilize the reduced interchain disulfide cysteine residues for conjugation. In this work, a novel bioanalytical method for the quantification of a cleavable antibody-conjugated drug in plasma was developed, qualified, and implemented. This novel method significantly improves throughput by combining a microwave-assisted, enzymatic cleavage of conjugated drugs from ADCs with a 96-well based sample preparation procedure to immunocapture ADCs in plasma. The released drug is subsequently quantified using a LC/MS/MS method. Our results represent a high-throughput, generic, and sensitive quantification method for antibody-conjugated microtubule inhibitors (such as MMAE) for preclinical PK/PD studies. The linear range of the standard curve for antibody conjugated drug (MMAE) was from 2.01 to 2010ng/mL with an excellent linearity (r(2)>0.997). The intra-run precision was below 8.14% and accuracy was from -7.71% to -1.08%. No matrix effect or carryover was observed for this method. This method was successfully used to measure the level of conjugated drug in a preclinical PK/PD study in mice.


Subject(s)
Immunoconjugates/analysis , Immunoconjugates/pharmacokinetics , Animals , Azides/chemistry , Biotin/chemistry , Cathepsin B/chemistry , Chromatography, High Pressure Liquid , Enzymes/chemistry , Female , Hydrolysis , Mice , Mice, SCID , Microtubule Proteins/antagonists & inhibitors , Microwaves , Oligopeptides/chemistry , Propanolamines/chemistry , Quality Control , Reproducibility of Results , Tandem Mass Spectrometry
9.
PLoS Biol ; 13(3): e1002087, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25764135

ABSTRACT

Epithelial morphogenesis involves a dramatic reorganisation of the microtubule cytoskeleton. How this complex process is controlled at the molecular level is still largely unknown. Here, we report that the centrosomal microtubule (MT)-binding protein CAP350 localises at adherens junctions in epithelial cells. By two-hybrid screening, we identified a direct interaction of CAP350 with the adhesion protein α-catenin that was further confirmed by co-immunoprecipitation experiments. Block of epithelial cadherin (E-cadherin)-mediated cell-cell adhesion or α-catenin depletion prevented CAP350 localisation at cell-cell junctions. Knocking down junction-located CAP350 inhibited the establishment of an apico-basal array of microtubules and impaired the acquisition of columnar shape in Madin-Darby canine kidney II (MDCKII) cells grown as polarised epithelia. Furthermore, MDCKII cystogenesis was also defective in junctional CAP350-depleted cells. CAP350-depleted MDCKII cysts were smaller and contained either multiple lumens or no lumen. Membrane polarity was not affected, but cortical microtubule bundles did not properly form. Our results indicate that CAP350 may act as an adaptor between adherens junctions and microtubules, thus regulating epithelial differentiation and contributing to the definition of cell architecture. We also uncover a central role of α-catenin in global cytoskeleton remodelling, in which it acts not only on actin but also on MT reorganisation during epithelial morphogenesis.


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Microtubule Proteins/genetics , Morphogenesis/genetics , Nuclear Proteins/genetics , alpha Catenin/genetics , Adherens Junctions/metabolism , Adherens Junctions/ultrastructure , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion , Cell Line , Cell Polarity , Cell Shape , Dogs , Embryo, Nonmammalian , Epithelial Cells/cytology , Genetic Vectors , Humans , Lentivirus/genetics , Madin Darby Canine Kidney Cells , Microtubule Proteins/antagonists & inhibitors , Microtubule Proteins/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Oryzias , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Two-Hybrid System Techniques , alpha Catenin/metabolism
10.
Int J Oncol ; 46(2): 649-56, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25405588

ABSTRACT

SPAG6, which is a novel cancer-testis antigen, is overexpressed in myeloid malignancies. Previously, SPAG6 was found in UPD (uniparental disomy) region of myeloid cell DNA from MDS patients and reported that SPAG6 may be a predictive marker of minimal residual disease in pediatric acute myeloid, but the biological role of SPAG6 in myeloid malignancies remains unclear. The present study was undertaken to determine the expression and functional significance of SPAG6 in malignant myeloid hematologic cell lines. A short hairpin RNA (shRNA) targeting SPAG6 was designed that could specifically inhibit SPAG6 expression at the mRNA and protein levels when introduced into the malignant myeloid hematologic cell lines SKM-1 and K562. The results from flow cytometry and CCK-8 assays showed that SPAG6 silencing inhibited the proliferation of SKM-1/K562 by inducing apoptosis. Furthermore, SPAG6 silencing resulted in activation of caspase-3, -9 and -8 and upregulated the mRNA and protein expression of p53 and PTEN. Then, we subcutaneously inoculated the monoclonal cells into NOD/SCID mice to establish xenograft models, and we found that the SPAG6-shRNA lentivirus dramatically inhibited tumor growth and increased apoptosis in vivo. These findings demonstrate that SPAG6 might have a role in malignant myeloid hematologic cell proliferation and apoptosis by regulating caspase proteins and p53, suggesting that SPAG6 may be a potential therapeutic target.


Subject(s)
Cell Proliferation/genetics , Microtubule Proteins/genetics , Myelodysplastic Syndromes/genetics , Tumor Suppressor Protein p53/biosynthesis , Animals , Apoptosis/genetics , Caspases/biosynthesis , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , K562 Cells , Mice , Microtubule Proteins/antagonists & inhibitors , Myelodysplastic Syndromes/pathology , Myeloid Cells/metabolism , Transcriptional Activation/genetics , Xenograft Model Antitumor Assays
11.
BMC Cancer ; 14: 52, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24484909

ABSTRACT

BACKGROUND: Metformin is an approved drug prescribed for diabetes. Its role as an anti-cancer agent has drawn significant attention because of its minimal side effects and low cost. However, its mechanism of anti-tumour action has not yet been fully clarified. METHODS: The effect on cell growth was assessed by cell counting. Western blot was used for analysis of protein levels, Boyden chamber assays for analyses of cell migration and co-immunoprecipitation (CoIP) followed by western blot, PCR or qPCR for analysis of protein-protein and protein-mRNA interactions. RESULTS: Metformin showed an anti-proliferative effect on a wide range of prostate cancer cells. It disrupted the AR translational MID1 regulator complex leading to release of the associated AR mRNA and subsequently to downregulation of AR protein in AR positive cell lines. Inhibition of AR positive and negative prostate cancer cells by metformin suggests involvement of additional targets. The inhibitory effect of metformin was mimicked by disruption of the MID1-α4/PP2A protein complex by siRNA knockdown of MID1 or α4 whereas AMPK activation was not required. CONCLUSIONS: Findings reported herein uncover a mechanism for the anti-tumor activity of metformin in prostate cancer, which is independent of its anti-diabetic effects. These data provide a rationale for the use of metformin in the treatment of hormone naïve and castration-resistant prostate cancer and suggest AR is an important indirect target of metformin.


Subject(s)
Antineoplastic Agents/pharmacology , Down-Regulation/physiology , Metformin/pharmacology , Microtubule Proteins/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Transcription Factors/metabolism , Antineoplastic Agents/therapeutic use , Cell Movement/drug effects , Cell Movement/physiology , Down-Regulation/drug effects , Humans , Male , Metformin/therapeutic use , Microtubule Proteins/antagonists & inhibitors , Microtubule Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Treatment Outcome , Ubiquitin-Protein Ligases
13.
Plant J ; 55(3): 443-54, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18410482

ABSTRACT

Previous studies have demonstrated that the major storage protein RNAs found in the rice endosperm are transported as particles via actomyosin to specific subdomains of the cortical endoplasmic reticulum. In this study, we examined the potential role of OsTudor-SN, a major cytoskeletal-associated RNA binding protein, in RNA transport and localization. OsTudor-SN molecules occur as high-molecular-weight forms, the integrity of which are sensitive to RNase. Immunoprecipitation followed by RT-PCR showed that OsTudor-SN binds prolamine and glutelin RNAs. Immunofluorescence studies using affinity-purified antibodies show that OsTudor-SNs exists as particles in the cytoplasm, and are distributed to both the protein body endoplasmic reticulum (ER) and cisternal ER. Examination of OsTudor-SN particles in transgenic rice plants expressing GFP-tagged prolamine RNA transport particles showed co-localization of OsTudor-SN and GFP, suggesting a role in RNA transport. Consistent with this view, GFP-tagged OsTudor-SN is observed in living endosperm sections as moving particles, a property inhibited by microfilament inhibitors. Downregulation of OsTudor-SN by antisense and RNAi resulted in a decrease in steady state prolamine RNA and protein levels, and a reduction in the number of prolamine protein bodies. Collectively, these results show that OsTudor-SN is a component of the RNA transport particle, and may control storage protein biosynthesis by regulating one or more processes leading to the transport, localization and anchoring of their RNAs to the cortical ER.


Subject(s)
Cytoplasm/metabolism , Microtubule Proteins/physiology , Oryza/metabolism , Plant Proteins/physiology , RNA, Plant/metabolism , RNA-Binding Proteins/physiology , Gene Expression Regulation, Plant , Green Fluorescent Proteins/analysis , Microtubule Proteins/antagonists & inhibitors , Microtubule Proteins/chemistry , Oryza/genetics , Oryza/growth & development , Plant Proteins/antagonists & inhibitors , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Prolamins , RNA Interference , RNA Transport , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , Recombinant Fusion Proteins/analysis , Seeds/genetics , Seeds/growth & development , Seeds/metabolism
14.
Curr Drug Targets ; 7(3): 305-26, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16515529

ABSTRACT

Antimitotic agents have been the most successful pharmacological agents for the treatment of cancer. The term "antimitotic agent" has traditionally been synonymous with tubulin-targeting compounds, but as a consequence of the large number of new compounds and mechanisms that have been identified recently, a much broader definition is currently needed. This review attempts to provide a broad overview of compounds and their cognate protein targets which result in a block in mitosis. Focus has been placed on agents that act directly on the mitotic machinery rather than on targets further upstream such as growth factor receptors.


Subject(s)
Antimitotic Agents/pharmacology , Biological Products/pharmacology , Actins/metabolism , Animals , Antimitotic Agents/classification , Binding Sites/drug effects , Biological Products/classification , Colchicine/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Humans , Kinesins/antagonists & inhibitors , Microtubule Proteins/antagonists & inhibitors , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , Proteasome Inhibitors , Sulfhydryl Compounds/pharmacology , Topoisomerase Inhibitors , Tubulin/drug effects , Vinca Alkaloids/pharmacology
15.
FEBS Lett ; 580(5): 1425-30, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16460735

ABSTRACT

Clathrin light chain (CL) b purified from bovine brain postmicrotubule supernatant and identified by mass spectrometry potently inhibited a catalytic activity of a major protein phosphatase (PP) that was copurified with microtubules and recognized by antiPP1 antibodies. CLb similarly affected the catalytic subunit and holoenzyme of the PP, little inhibiting the activity of PP2A. Although the CLb from clathrin-coated vesicles was several hundredfold weaker than our purified CLb, the CLb in the postmicrotubule supernatant, independent of whether it was sedimentable or soluble, was as active as the purified CLb. Thus CLb may be a potent regulator of the PP.


Subject(s)
Clathrin Light Chains/pharmacology , Microtubule Proteins/antagonists & inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Amino Acid Sequence , Animals , Cattle , Clathrin Light Chains/isolation & purification , Clathrin-Coated Vesicles , Mass Spectrometry , Protein Phosphatase 2 , Solubility
16.
Arch Pharm Res ; 27(4): 436-41, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15180310

ABSTRACT

SJ compounds (SJ8002 and related compounds) are a group of novel anticancer agents (Cho, Chung, Lee, Kwon, Kang, Joo, and Oh. PCT/KR02/00392). To explore the anticancer mechanism of these compounds, we examined the effect of SJ8002 on microtubules of six human cell lines. At a high concentration (2 microg/mL), SJ8002 effectively disrupted microtubules of the six cell lines within 1 h. At lower concentrations (0.05 to approximately 1.0 microg/mL), the antimicrotubule activity of SJ8002 varied defending on cell lines. The inhibition of in vitro polymerization of pure tubulin by SJ8002 suggested that SJ8002 acts on free tubulin, inhibits the polymerization of tubulin dimer into microtubules, and hence induces the depolymerization of microtubules.


Subject(s)
Acridines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Microtubule Proteins/metabolism , Microtubules/drug effects , Microtubules/metabolism , Piperazines/pharmacology , Pyridines/pharmacology , Acridines/chemistry , Cell Line , Cell Line, Tumor , HeLa Cells , Humans , Microtubule Proteins/antagonists & inhibitors , Piperazines/chemistry , Pyridines/chemistry
17.
Cardiovasc Toxicol ; 2(2): 75-89, 2002.
Article in English | MEDLINE | ID: mdl-12271151

ABSTRACT

Microtubules (MTs) are dynamic, cytoskeletal fibers that are found in every eukaryotic cell type. MTs serve a wide range of functions, including cell division, membrane and vesicle transport, and motility. As such, MTs play pivotal roles in cardiac development and function. Agents that disrupt normal MT function, including such therapeutic agents as vincristine and paclitaxel, have also been shown to affect essential cardiac activities such as sarcomere mechanics, beat rate, and the secretion of important molecules (e.g., atrial natriuretic factor). Disease states that lead to either ischemia- or pressure overload- induced cardiac hypertrophy also alter the microtubule cytoskeleton in several ways. A fuller understanding of the contributions of MTs to cardiac development and function will be necessary to minimize the deleterious side effects of the therapeutic application of MT-disrupting drugs. This review summarizes current hypotheses and experimental results that demonstrate the central role of MTs in heart cell function and disease.


Subject(s)
Heart Diseases/physiopathology , Microtubule Proteins/physiology , Microtubules/physiology , Growth/drug effects , Humans , Microtubule Proteins/antagonists & inhibitors , Microtubules/drug effects , Myocardium/cytology , Myofibrils/physiology , United States
18.
J Antibiot (Tokyo) ; 55(3): 256-62, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12014440

ABSTRACT

A new screening method to detect antimitotic substances utilizing purified porcine brain microtubule proteins was developed. This method observes the inhibitory and stimulatory activities on microtubule polymerization and inhibitory activity on depolymerization in sequence. Two glycolipids, 1-O-beta-D-galactopyranosyl-2,3-di-O-acylglycerol and 1-O-tetrahydroxycyclopentyl-2-O-acyl-3-O-alkylglycerol were isolated from Okinawan marine sponge Pseudoceratina sp. by this screening method. These compounds stimulated the microtubule polymerization at 10 degrees C.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Glycolipids/isolation & purification , Glycolipids/pharmacology , Porifera/chemistry , Tubulin/metabolism , Animals , Antineoplastic Agents/chemistry , Biopolymers/antagonists & inhibitors , Biopolymers/metabolism , Brain/metabolism , Drug Screening Assays, Antitumor , Glycolipids/chemistry , Microtubule Proteins/antagonists & inhibitors , Microtubule Proteins/metabolism , Porifera/metabolism , Swine
19.
J Neurosci Res ; 66(4): 601-11, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11746380

ABSTRACT

Superior cervical ganglion (SCG) cells from neonatal rats underwent apoptosis upon treatment with colchicine, a microtubule-disrupting agent. Western blotting and activity measurements showed that caspase-3 was indeed activated, but its peptide inhibitor (Ac-DEVD-CHO) neither suppressed nuclear fragmentation nor rescued the neurons from cell death. z-VAD-fmk, the general inhibitor of caspases, prevented nuclear fragmentation and delayed the cell death. Moreover, N-alpha-tosyl-L-lysine chloromethyl ketone (TLCK), but not N-alpha-tosyl-L-phenylalanine chloromethyl ketone (TPCK), prevented nuclear fragmentation and provided neuronal protection as well. The combination of both z-VAD-fmk and TLCK provided a long-term neuronal protection (>4 days), whereas neither one alone could do so, suggesting that there are both caspase-dependent and -independent pathways. TLCK-sensitive serine protease is also likely to act upstream of caspase-3 in a caspase-dependent pathway. Electron microscopic observations demonstrated that z-VAD-fmk suppressed nuclear fragmentation and improved mitochondrial swelling, but failed to prevent vesicular formation, which resulted in a slowly-occurring necrosis. More importantly, TLCK effectively blocked this abundant vesicular formation along with suppressing chromatin condensation. Thus, the combination of both conferred a nearly normal morphology, which is consistent with the results of cell survival experiments. These findings clearly indicate that TLCK-sensitive serine protease plays multiple roles in caspase-dependent and -independent pathways of colchicine-induced cell death, and suggest a novel mechanism underlying a necrotic pathway involving ER swelling and vesicular formation.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Necrosis , Nervous System/enzymology , Neurons/enzymology , Serine Endopeptidases/metabolism , Signal Transduction/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Bucladesine/pharmacology , Caspase 3 , Caspases/drug effects , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cells, Cultured/ultrastructure , Colchicine/pharmacology , Enzyme Inhibitors , Immunohistochemistry , Microscopy, Electron , Microtubule Proteins/antagonists & inhibitors , Microtubule Proteins/metabolism , Nervous System/drug effects , Nervous System/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Neuroprotective Agents/pharmacology , Potassium/pharmacology , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Serine Endopeptidases/drug effects , Serine Proteinase Inhibitors/pharmacology , Signal Transduction/physiology , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/enzymology , Superior Cervical Ganglion/ultrastructure , Tosyllysine Chloromethyl Ketone/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology
20.
Biochem Pharmacol ; 62(5): 581-91, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11585055

ABSTRACT

Vinblastine and other microtubule inhibitors are important antitumor agents that cause mitotic arrest, and induce apoptosis through poorly understood mechanisms, in a wide variety of cell lines. The activating protein 1 (AP-1) transcription factor is a major target of the c-Jun NH2-terminal kinase (JNK) signaling pathway, which is activated by microtubule inhibitors. Therefore, we examined the effect of vinblastine on AP-1 composition and activity in human KB-3 carcinoma cells. Vinblastine caused highly selective effects on AP-1 proteins in a concentration- and time-dependent manner. Specifically, c-Jun, expressed at a low level in control cells, was greatly increased and phosphorylated, Jun D was phosphorylated, Jun B underwent phosphorylation and subsequently became undetectable, and Fra 1 expression was also greatly increased. In contrast. Fra 2, c-Fos, and Fos B were relatively unchanged by vinblastine. Changes in AP-1 preceded caspase 3 activation and, therefore, occurred prior to the commitment phase of apoptosis. With the exception of c-Jun, which was not affected by paclitaxel, the same alterations in AP-1 proteins occurred after exposure to vincristine, paclitaxel, and colchicine, demonstrating that these are general responses to microtubule inhibition. Supershift assays demonstrated that in control cells, AP-1 binding activity was mediated by Jun D/Fra 2 heterodimers, whereas after vinblastine treatment, AP-1 complexes also containing c-Jun and Fra 1 were present, suggesting that induction of these latter proteins by vinblastine is functionally significant. Consistent with these observations, vinblastine stimulated AP-1-dependent luciferase reporter gene transcription. These findings suggest that alterations in AP-1 composition and activity may be key events in the early response of KB-3 cells to microtubule inhibitors.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Vinblastine/pharmacology , Binding Sites , Caspase 3 , Caspases/metabolism , DNA/drug effects , DNA/metabolism , Gene Expression/drug effects , Humans , Microtubule Proteins/antagonists & inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-jun/drug effects , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/genetics , Transcription, Genetic/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...