Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Am J Physiol Endocrinol Metab ; 316(4): E590-E604, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30668150

ABSTRACT

Parathyroid hormone (PTH) and its related peptide (PTH-related peptide 1-34) are two of the Food and Drug Administration-approved bone-promoting drugs for age-related osteoporosis. Treatment with PTH stimulates bone formation. However, the molecular mechanisms of PTH-mediated osteoblast differentiation and cell proliferation are still not completely understood. In this study, we showed that PTH induced endoplasmic reticulum (ER) stress in osteoblasts through the PKR-like endoplasmic reticulum kinase (PERK)-eukaryotic initiation factor 2α (EIF2α)-activating transcription factor 4 (ATF4)-signaling pathway. After separately blocking PERK-EIF2α-ATF4 signaling with two different inhibitors [AMG'44 and integrated stress response inhibitor (ISRIB)] or specific small interfering RNA for PERK and ATF4, the following targets were all downregulated: expression of osteoblast differentiation markers [runt-related transcription factor 2 (Runx2), alkaline phosphatase (Alp), type I collagen (Col1a1), and osteocalcin (Ocn)], cell proliferation markers (CyclinE, CyclinD, and CDC2), amino acid import (Glyt1), and metabolism-related genes (Asns). Additionally, Alp-positive staining cells, Alp activity, matrix mineralization, Ocn secretion, and cell proliferation indexes were inhibited. Interestingly, we found that salubrinal enhanced PTH-induced osteoblast differentiation and proliferation by maintenance of phosphorylation of EIF2α. Furthermore, we observed that PTH increased the association between heat shock protein 90 (HSP90) and PERK and maintained PERK protein stabilization in the early stages of PTH-induced ER stress. Treatment of MC3T3-E1 cells with geldanamycin, an HSP90 inhibitor, decreased PERK protein expression and inhibited osteoblast differentiation and cell proliferation upon PTH treatment. Taken together, our data demonstrate that PTH regulates osteoblast differentiation and cell proliferation, partly by activating the HSP90-dependent PERK-EIF2α-ATF4 signaling pathway.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Activating Transcription Factor 4/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Benzoquinones/pharmacology , CDC2 Protein Kinase/drug effects , CDC2 Protein Kinase/metabolism , Cell Line , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Cyclin D/drug effects , Cyclin D/metabolism , Cyclin E/drug effects , Cyclin E/metabolism , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Glycine Plasma Membrane Transport Proteins/drug effects , Glycine Plasma Membrane Transport Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/drug effects , HSP90 Heat-Shock Proteins/metabolism , Lactams, Macrocyclic/pharmacology , Mice , Osteoblasts/metabolism , Osteocalcin/drug effects , Osteocalcin/metabolism , Signal Transduction , eIF-2 Kinase/metabolism
2.
Asian J Androl ; 17(6): 996-1005, 2015.
Article in English | MEDLINE | ID: mdl-26289399

ABSTRACT

This study was designed to explore the regulatory effects of male germ cell secreting factor NODAL on Sertoli cell fate decisions from obstructive azoospermia (OA) and nonobstructive azoospermia (NOA) patients. Human Sertoli cells and male germ cells were isolated using two-step enzymatic digestion and SATPUT from testes of azoospermia patients. Expression of NODAL and its multiple receptors in human Sertoli cells and male germ cells were characterized by reverse transcription-polymerase chain reaction (RT-PCR) and immunochemistry. Human recombinant NODAL and its receptor inhibitor SB431542 were employed to probe their effect on the proliferation of Sertoli cells using the CCK-8 assay. Quantitative PCR and Western blots were utilized to assess the expression of Sertoli cell functional genes and proteins. NODAL was found to be expressed in male germ cells but not in Sertoli cells, whereas its receptors ALK4, ALK7, and ACTR-IIB were detected in Sertoli cells and germ cells, suggesting that NODAL plays a regulatory role in Sertoli cells and germ cells via a paracrine and autocrine pathway, respectively. Human recombinant NODAL could promote the proliferation of human Sertoli cells. The expression of cell cycle regulators, including CYCLIN A, CYCLIN D1 and CYCLIN E, was not remarkably affected by NODAL signaling. NODAL enhanced the expression of essential growth factors, including GDNF, SCF, and BMP4, whereas SB431542 decreased their levels. There was not homogeneity of genes changes by NODAL treatment in Sertoli cells from OA and Sertoli cell-only syndrome (SCO) patients. Collectively, this study demonstrates that NODAL produced by human male germ cells regulates proliferation and numerous gene expression of Sertoli cells.


Subject(s)
Azoospermia/metabolism , Cell Proliferation/drug effects , Nodal Protein/pharmacology , Recombinant Proteins/pharmacology , Sertoli Cells/drug effects , Spermatozoa/metabolism , Activin Receptors, Type I/metabolism , Activin Receptors, Type II/metabolism , Adult , Autocrine Communication , Benzamides/pharmacology , Blotting, Western , Bone Morphogenetic Protein 4/drug effects , Bone Morphogenetic Protein 4/metabolism , Cyclin A/drug effects , Cyclin A/metabolism , Cyclin D1/drug effects , Cyclin D1/metabolism , Cyclin E/drug effects , Cyclin E/metabolism , Dioxoles/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Immunohistochemistry , Male , Nodal Protein/metabolism , Paracrine Communication , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Factor/drug effects , Stem Cell Factor/metabolism
3.
Womens Health (Lond) ; 10(1): 45-57, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24328598

ABSTRACT

Uterine serous carcinoma (USC) is a highly aggressive variant of endometrial cancer. Although it only represents less than 10% of all cases, it accounts for a disproportionate number of deaths from endometrial cancer. Comprehensive surgical staging followed by carboplatin and paclitaxel chemotherapy represents the mainstay of USC therapy. Vaginal cuff brachytherapy is also of potential benefit in USC. Recent whole-exome sequencing studies have demonstrated gain of function of the HER2/NEU gene, as well as driver mutations in the PIK3CA/AKT/mTOR and cyclin E/FBXW7 oncogenic pathways in a large number of USCs. These results emphasize the relevance of these novel therapeutic targets for biologic therapy of chemotherapy-resistant recurrent USC.


Subject(s)
Antineoplastic Agents/therapeutic use , Cystadenocarcinoma, Serous/drug therapy , Molecular Targeted Therapy/methods , Neoplasm Recurrence, Local/drug therapy , Uterine Neoplasms/drug therapy , Class I Phosphatidylinositol 3-Kinases , Cyclin E/drug effects , Cyclin E/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Drug Resistance, Neoplasm/drug effects , Female , Genes, erbB-2/drug effects , Genes, erbB-2/genetics , Humans , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/genetics , Randomized Controlled Trials as Topic , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
4.
Cell Cycle ; 11(7): 1374-82, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22421144

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is the sixth leading cancer in the world; the main risk factors are alcohol and tobacco use. Advancements in therapies have yet to improve the prognosis of HNSCC. The connection between diabetes and cancer is being recognized, and metformin has been shown to decrease cancer incidence in diabetic patients. Accordingly, here, for the first time, we investigated metformin's efficacy on the growth and viability of human HNSCC FaDU and Detroit cells. Our results show that metformin treatment (5-20 mM) dose-dependently inhibits the growth of both cell lines. In FaDU cells, metformin caused 18-57% and 35-81% growth inhibition after 48 and 72 h treatments, respectively. Similarly, in Detroit 562 cells, 48 and 72 h metformin treatment resulted in 20-57% and 33-82% inhibition, respectively. Mechanistically, metformin caused G 1 arrest, which coincided with a decrease in the protein levels of CDKs (2, 4 and 6), cyclins (D1 and E) and CDK inhibitors (p15, p16, p18 and p27), but no change in p19 and p21. Metformin also decreased the levels of oncogenic proteins Skp2 and ß-Trcp. In other studies, metformin decreased the phosphorylation of 4E-BP1 at Ser65, Thr37/46 and Thr70 sites, but drastically increased the phosphorylation of EF2 at Thr56 and AMPK at Thr172, which results in global translational inhibition. In summary, the observed wide spectrum of mechanistic effects of metformin on HNSCC cells provides support for the anticancer capability of the drug and its potential use in future therapies.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Metformin/pharmacology , Protein Biosynthesis/drug effects , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cyclin D1/metabolism , Cyclin E/analysis , Cyclin E/drug effects , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Head and Neck Neoplasms/pathology , Humans , Neoplasm Proteins/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck , beta-Transducin Repeat-Containing Proteins/metabolism , rho GTP-Binding Proteins/metabolism
5.
Anticancer Drugs ; 23(4): 370-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22185819

ABSTRACT

Artemisinin, a sesquiterpene phytolactone derived from Artemisia annua, is a potent antimalarial compound with promising anticancer properties, although the mechanism of its anticancer signaling is not well understood. Artemisinin inhibited proliferation and induced a strong G1 cell cycle arrest of cultured MCF7 cells, an estrogen-responsive human breast cancer cell line that represents an early-stage cancer phenotype, and effectively inhibited the in-vivo growth of MCF7 cell-derived tumors from xenografts in athymic nude mice. Artemisinin also induced a growth arrest of tumorigenic human breast cancer cell lines with preneoplastic and late stage cancer phenotypes, but failed to arrest the growth of a nontumorigenic human mammary cell line. Concurrent with the cell cycle arrest of MCF7 cells, artemisinin selectively downregulated the transcript and protein levels of the CDK2 and CDK4 cyclin-dependent kinases, cyclin E, cyclin D1, and the E2F1 transcription factor. Analysis of CDK2 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK2 gene expression was accounted for by the loss of CDK2 promoter activity. Chromatin immunoprecipitation revealed that artemisinin inhibited E2F1 interactions with the endogenous MCF7 cell CDK2 and cyclin E promoters. Moreover, constitutive expression of exogenous E2F1 prevented the artemisinin-induced cell cycle arrest and downregulation of CDK2 and cyclin E gene expression. Taken together, our results demonstrate that the artemisinin disruption of E2F1 transcription factor expression mediates the cell cycle arrest of human breast cancer cells and represents a critical transcriptional pathway by which artemisinin controls human reproductive cancer cell growth.


Subject(s)
Antineoplastic Agents/pharmacology , Artemisinins/pharmacology , Breast Neoplasms/drug therapy , E2F1 Transcription Factor/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclin D1/drug effects , Cyclin D1/metabolism , Cyclin E/drug effects , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/drug effects , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/drug effects , Cyclin-Dependent Kinase 4/metabolism , E2F1 Transcription Factor/metabolism , Female , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
6.
J Exp Ther Oncol ; 9(3): 175-81, 2011.
Article in English | MEDLINE | ID: mdl-22070048

ABSTRACT

Ovarian cancer is the leading cause of death from gynecological cancer. The high mortality rate reflets the lack of early diagnosis and limited treatment alternatives. We have observed a number of properties of zinc cytotoxicity that make it attractive from a therapeutic standpoint. Using SKOV3 and ES2 cells, ovarian cancer cell lines that demonstrate varied degrees of resistance to known therapeutics, we show that zinc killing is time and concentration dependent. Death is preceded by distinct changes in cell shape and size. The effects of zinc are additive with cisplatin or doxorubicin, whose morphological effects are distinct from those of zinc. Cytotoxicity of paclitaxel is minimal, making it difficult to determine additivity with zinc. Paclitaxel results in changes in cell shape and size similar to those of zinc but has different effects on cell cycle progression and cyclin expression. The data indicate that the means by which zinc kills ovarian cancer cells is distinct from currently used chemotherapeutics. Based on the properties reported here, zinc has the potential to be developed as either a primary treatment or as a second line of defense against cancers that have developed resistance to currently used chemotherapeutics.


Subject(s)
Cell Cycle Checkpoints/drug effects , Ovarian Neoplasms/drug therapy , Trace Elements/pharmacology , Zinc/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Cyclin A/drug effects , Cyclin A/metabolism , Cyclin D/drug effects , Cyclin D/metabolism , Cyclin E/drug effects , Cyclin E/metabolism , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Synergism , Female , Humans , Paclitaxel/pharmacology , Pyridines/pharmacology , Thiones/pharmacology
7.
Arch Oral Biol ; 55(12): 938-45, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20832772

ABSTRACT

Voltage gated chloride channels (ClCs) play an important role in the regulation of intracellular pH and cell volume homeostasis. Mutations of these genes result in genetic diseases with abnormal bone deformation and body size, indicating that ClCs may have a role in chondrogenesis. In the present study, we isolated chicken mandibular mesenchymal cells (CMMC) from Hamburg-Hamilton (HH) stage 26 chick embryos and induced chondrocyte maturation by using ascorbic acid and ß-glycerophosphate (AA-BGP). We also determined the effect of the chloride channel inhibitor NPPB [5-nitro-2-(3-phenylpropylamino) benzoic acid] on regulation of growth, differentiation, and gene expression in these cells using MTT and real-time PCR assays. We found that CLCN1 and CLCN3-7 mRNA were expressed in CMMC and NPPB reduced expression of CLCN3, CLCN5, and CLCN7 mRNA in these cells. At the same time, NPPB inhibited the growth of the CMMC, but had no effect on the mRNA level of cyclin D1 and cyclin E (P>0.05) with/without AA-BGP treatment. AA-BGP increased markers for early chondrocyte differentiation including type II collagen, aggrecan (P<0.01) and Sox9 (P<0.05), whilst had no effect on the late chondrocyte differentiation marker type X collagen. NPPB antagonized AA-BGP-induced expression of type II collagen and aggrecan (P<0.05). Furthermore, NPPB downregulated type X collagen (P<0.05) with/without AA-BGP treatment. We conclude that abundant chloride channel genes in CMMC play important roles in regulating chondrocyte proliferation and differentiation. Type X collagen might function as a target of chloride channel inhibitors during the differentiation process.


Subject(s)
Chloride Channels/physiology , Chondrogenesis/physiology , Mandible/embryology , Mesoderm/embryology , Aggrecans/drug effects , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chick Embryo , Chloride Channels/analysis , Chloride Channels/antagonists & inhibitors , Chondrocytes/drug effects , Chondrogenesis/drug effects , Collagen Type II/drug effects , Collagen Type X/drug effects , Cyclin D1/drug effects , Cyclin E/drug effects , Gene Expression Regulation, Developmental/drug effects , Glycerophosphates/pharmacology , Mandible/drug effects , Mesoderm/cytology , Mesoderm/drug effects , Muscle Proteins/analysis , Muscle Proteins/antagonists & inhibitors , Nitrobenzoates/pharmacology , SOX9 Transcription Factor/drug effects
8.
Brain Res ; 1359: 272-80, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20813099

ABSTRACT

Methylmercury (MeHg) is a potent environmental neurotoxin that shows toxicity to developing central nervous system (CNS), causing brain damage in children even at low exposure levels. However, the mechanisms for its effect on CNS are not well understood. In current study, primary cultures of progenitor cells from embryonic cerebral cortex were used as a model system to study the potential effect and the underlying mechanism of MeHg on neural progenitor cells. Results showed that, in cultured cortical progenitor cells, 48-h exposure to low-level of MeHg (at 2.5 nM, 5 nM and 50 nM, respectively) caused G1/S cell cycle arrest in a dose-dependent manner without inducing cell death. Interestingly, the expression of cyclin E, which promotes G1/S transition, but not cyclin D1 and CDK2, was selectively downregulated by exposure of MeHg. In addition, low-level of MeHg inhibited the maintenance of ERK1/2 phosphorylation, possibly by abolishing the late phase ERK1/2 activation induced by bFGF. Thus, MeHg may induce proliferation inhibition and cell cycle arrest of neural progenitor cells via regulating cyclin E expression and perturbing a pathway that involves ERK1/2.


Subject(s)
Cell Proliferation/drug effects , Methylmercury Compounds/toxicity , Neural Stem Cells/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cyclin E/drug effects , Cyclin E/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique , Neural Stem Cells/metabolism , Rats , Rats, Sprague-Dawley
9.
Breast Cancer Res Treat ; 121(2): 355-64, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19641990

ABSTRACT

In contrast to deoxyribose or arabinose containing nucleoside analogs that are currently established for cancer therapeutics, 8-chloro-adenosine (8-Cl-Ado) possesses a ribose sugar. This unique nucleoside analog is RNA-directed and is in a phase I clinical trial for hematological malignancies. RNA-directed therapies are effective for the treatment of many malignancies as their activities are primarily aimed at short-lived transcripts, which are typically encoded by genes that promote the growth and survival of tumor cells such as cyclin E in breast cancer. Based on this, we hypothesized that 8-Cl-Ado, a transcription inhibitor, will be effective for the treatment of breast cancer cells. The metabolism of 8-Cl-Ado and the effect on ATP in the breast cancer cell lines MCF-7 and BT-474 were measured using HPLC analysis. In these cells, 8-Cl-Ado was effectively taken up, converted to its cytotoxic metabolite, 8-Cl-ATP, and depleted the endogenous ATP levels. This in turn led to an inhibition of RNA synthesis. The RNA synthesis inhibition was associated with a depletion of cyclin E expression, which is indicative of a diminished tumorigenic phenotype. The final outcome of 8-Cl-Ado treatment of the breast cancer cells was growth inhibition due to an induction of apoptosis and a loss of clonogenic survival. These results indicate that 8-Cl-Ado, which is currently in clinic for hematological malignancies, may be an effective agent for the treatment of breast cancer.


Subject(s)
2-Chloroadenosine/analogs & derivatives , Antineoplastic Agents/metabolism , Breast Neoplasms/metabolism , Cyclin E/metabolism , RNA/drug effects , 2-Chloroadenosine/metabolism , 2-Chloroadenosine/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Separation , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cyclin E/drug effects , Female , Flow Cytometry , Humans , Reverse Transcriptase Polymerase Chain Reaction
10.
Anticancer Drugs ; 20(4): 230-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19288605

ABSTRACT

A number of polyamine analogs are currently used in various clinical trials as cancer treatment and it is important to investigate their effects not only on cancer cells but also on normal cells. Treatment with polyamine analogs depletes cells of polyamines and inhibits cell proliferation, but the analogs cannot take over the normal function of the natural polyamines in the cell. In this study, the normal-like breast epithelial cell line MCF-10A was treated with the polyamine analog N',N"-diethylnorspermine (DENSPM). The cells were then studied using a bromodeoxyuridine- DNA flow cytometry method as well as western blot. The ability of both normal-like and breast cancer cells to recover from DENSPM treatment was also studied. DENSPM treatment of MCF-10A cells resulted in a prolongation of the S and G2 +M phases, followed by a G1/S block. The p53/p21/RB1 pathway was involved in the G1/S block as shown by increased levels of p53 and p21 detected by western blot. Decreased levels of cyclin E1, cyclin A2, and cyclin B1 in DENSPM-treated cells can explain the prolongation of cell cycle phases that occurred before the G1/S block. We also show that MCF-10A cells rapidly recover from DENSPM-induced growth inhibition in contrast to four human breast cancer cell lines. The goal of cancer treatment is to cause minimal and reversible damage to normal cells, while cancer cells should be eliminated. Altogether, the data show that treatment with polyamine analogs spares normal cells, while negatively affecting the cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Spermine/analogs & derivatives , Blotting, Western , Breast Neoplasms/pathology , Bromodeoxyuridine , Cell Line , Cell Line, Tumor , Cyclin A/drug effects , Cyclin A/metabolism , Cyclin A2 , Cyclin B/drug effects , Cyclin B/metabolism , Cyclin B1 , Cyclin E/drug effects , Cyclin E/metabolism , DNA , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Flow Cytometry , Humans , Oncogene Proteins/drug effects , Oncogene Proteins/metabolism , Spermine/pharmacology
11.
Biochem Pharmacol ; 77(2): 151-8, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18977205

ABSTRACT

Cyclin E is the Cdk2-regulatory subunit required for the initiation of DNA replication at the G1/S transition. It accumulates in late G1 phase and gets rapidly degraded by the ubiquitin/proteasome pathway during S phase. The degradation of cyclin E is a consequence of its phosphorylation and subsequent isomerization by the peptidyl-prolyl isomerase Pin1. We show that in the colon cancer cells HT-29 the inhibition of the chaperone function of Hsp90 by geldanamycin (GA) enhances the ubiquitinylation of cyclin E and triggers active degradation via the proteasome pathway. As Hsp90 forms multiprotein complexes with and regulates the function and cell contents of numerous signaling proteins, this observation suggests a direct interaction between Hsp90 and cyclin E. However, experiments using cell lysate fractionation did not reveal the presence of complexes containing both Hsp90 and cyclin E. Coupled transcription/translation experiments also failed to detect the formation of complexes between newly synthesized cyclin E and Hsp90. We conclude that Hsp90 can regulate the degradation of cellular proteins without binding to them, by an indirect mechanism. This conclusion postulates a new category of proteins that are affected by the inactivation of Hsp90. Our observations do not support the possible involvement of a PPIase in this indirect mechanism. Besides, we did not observe active geldanamycin-dependent degradation of cyclin E in the prostate cancer-derived cell line DU-145, indicating that the Hsp90-dependent stabilization of cyclin E requires specific regulatory mechanism which may be lost in certain types of cancer cells.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Benzoquinones/pharmacology , Cyclin E/metabolism , HSP90 Heat-Shock Proteins/physiology , Lactams, Macrocyclic/pharmacology , Aphidicolin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Colonic Neoplasms , Cyclin E/drug effects , Cyclin E/genetics , DNA Replication/physiology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Homeostasis , Humans , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/drug effects , Prostatic Neoplasms , Protein Biosynthesis , Transcription, Genetic , Ubiquitin/metabolism
12.
Environ Toxicol ; 24(3): 243-58, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18683188

ABSTRACT

Humic acid (HA) in well water used by the inhabitants for drinking is one of the possible etiological factors for Blackfoot disease (BFD). In this study, the ability of HA to inhibit cell cycle progression and induce apoptosis in cultured smooth muscle cells (SMCs; A7r5) was investigated. Treatment of the SMCs at various HA concentrations (25-200 microg/mL) resulted in sequences of events marked by apoptosis, as shown by loss of cell viability, morphology change, and internucleosomal DNA fragmentation. HA-induced apoptotic cell death that is associated with loss of mitochondrial membrane potential (Delta Psi m), cytochrome c translocation, caspase-3, -8, and -9 activation, poly ADP-ribose polymerase (PARP) degradation, dysregulation of Bcl-2 and Bax, and upregulation of p53 and phospholyrated p53 (p-p53) in SMCs. Flow cytometry analysis demonstrated that HA blocked cell cycle progress in the G1 phase in SMCs. This blockade of cell cycle was associated with reduced amounts of cyclin D1, CDK4, cyclin E, CDK2, and hyperphosphorylated retinoblastoma protein (pRb) in a time-dependent manner. Apparent DNA strand breaks (DNA damage) were also detected in a dose-dependent manner using Single-cell gel electrophoresis assay (comet assay). Furthermore, HA induced dose-dependent elevation of reactive oxygen species (ROS) level in SMCs, and antioxidant vitamin C and Trolox effectively suppressed HA-induced DNA damage and dysregulation of Bcl-2/Bax. Our findings suggest that HA-induced DNA damage, cell cycle arrest, and apoptosis in SMCs may be an underlying mechanisms for the atherosclerosis and thrombosis observed in the BFD endemic region.


Subject(s)
Apoptosis , G1 Phase/drug effects , Humic Substances/toxicity , Muscle, Smooth, Vascular/drug effects , Animals , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Collagen Type XI/metabolism , Cyclin D1/drug effects , Cyclin D1/genetics , Cyclin E/drug effects , Cyclin E/genetics , Cyclin-Dependent Kinase 2/drug effects , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 4/drug effects , Cyclin-Dependent Kinase 4/genetics , Cytochromes c/metabolism , DNA Fragmentation , Membrane Potential, Mitochondrial/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Retinoblastoma Protein/metabolism , bcl-2-Associated X Protein/metabolism
13.
Virology ; 367(1): 1-9, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17568647

ABSTRACT

Epidemiological studies have demonstrated that 15 different mucosal human papillomavirus (HPV) types of the genus alpha of the HPV phylogetic tree are classified as high risk for cervical cancer development. Three additional HPV types of the same genus, HPV26, 53 and 66, are classified as probable high-risk types. In this study, we have characterized the biological properties of the E7 oncoproteins from these three HPV types. All of the corresponding E7 proteins were able to associate with retinoblastoma protein (pRb) and up-regulated the expression of several positive cell cycle regulators, i.e. CDK2, cyclin A and cylin E. However, HPV26 E7 appears to be more efficient than HPV53 and 66 E7 in up-regulating the transcription of cyclin A. Unlike E7 from the high-risk type HPV16 protein, HPV26, 53 and 66 did not efficiently promote pRb degradation. In addition, E7 from these viruses was able to promote proliferation of primary human keratinocytes and circumvent G1 arrest imposed by overexpression of p16(INK4a), but with less efficiency than the high-risk HPV16 E7. Together, our data show that in vitro properties of these E7 proteins correlate with the epidemiological classification of HPV26, 53 and 66 as HPV types with an intermediate risk for cervical cancer development.


Subject(s)
Alphapapillomavirus/classification , Alphapapillomavirus/pathogenicity , Cell Transformation, Neoplastic , Keratinocytes/virology , Papillomavirus E7 Proteins/metabolism , Alphapapillomavirus/metabolism , Cell Line , Cell Line, Transformed , Cells, Cultured , Cyclin A/drug effects , Cyclin A/metabolism , Cyclin E/drug effects , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/drug effects , Cyclin-Dependent Kinase 2/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , G1 Phase/drug effects , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/pharmacology , Papillomavirus Infections/virology , Retinoblastoma Protein/metabolism , Risk Factors , Up-Regulation , Uterine Cervical Neoplasms/virology
14.
Br J Cancer ; 95(11): 1514-24, 2006 Dec 04.
Article in English | MEDLINE | ID: mdl-17088910

ABSTRACT

The cisplatin analogue 1R,2R-diaminocyclohexane(trans-diacetato)(dichloro)platinum(IV) (DAP) is a DNA-damaging agent that will be entering clinical trials for its potent cytotoxic effects against cisplatin-resistant tumour cells. This cytotoxicity may reside in its ability to selectively activate G1-phase checkpoint response by inhibiting CDKs via the p53/p21 pathway. We have now evaluated the role of another CDK inhibitor p27 as a contributor to DAP-mediated inhibition of G1-phase CDK2 activity. Our studies in ovarian A2780 tumour cells demonstrate that p27 levels induced by DAP are comparable to or greater than those seen for p21. The induction of p27 is not through a transcriptional mechanism, but rather is due to a four-fold increase in protein stabilisation through a mechanism dependent on p21. Moreover, DAP-induced p21 promoted the selective increase of p27 in the CDK2 complex, but not in CDK4 complex, and this selective increase contributed to inhibition of the CDK2 kinase activity. The inhibited complex contained either p27 or p21, but not both, with the relative levels of cyclin E associated with p27 and p21 indicating that about 25% of the inhibition of CDK2 activity was due to p27 and 75% due to p21. This study provides the first evidence that p27 upregulation is directly attributable to activation of the p53/p21 pathway by a DNA-damaging agent, and promulgates p53/p21/p27 axis as a significant component of checkpoint response.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/analogs & derivatives , Cyclin E/drug effects , Cyclin-Dependent Kinase 2/drug effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , DNA Damage , Proliferating Cell Nuclear Antigen/drug effects , Blotting, Northern , Cell Line, Tumor , Cisplatin/pharmacology , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinases/drug effects , Cyclin-Dependent Kinases/metabolism , Female , Humans , Immunoblotting , Immunoprecipitation , Organoplatinum Compounds , Plasmids , Proliferating Cell Nuclear Antigen/metabolism , RNA, Small Interfering , Transfection , Up-Regulation
15.
Cancer Lett ; 234(2): 199-208, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-15885897

ABSTRACT

Caffeic acid phenethyl ester (CAPE), a component of honeybee propolis, has been reported to hold various biochemical responses. In the preliminary study, we found that CAPE inhibited the growth of C6 glioma cells in a dose dependent and time dependent manner as shown by the results of trypan blue dye exclusion assay and cell proliferation assay. In addition, the cell number percentage of the G0/G1 phase increased to 85% after the treatment with 50 microM of CAPE for 24h. After treatment with CAPE (50 microM) for 6h, it demonstrated that the protein level of hyperphosphorylated pRb decreased, and cyclin dependent kinase inhibitors p21, p27, and p16 were marked up-regulated. The association of CDK2 and cyclin E that affects the CDK2 activity decreased. When C6 cells were grown as xenografts in nude mice, treatment with CAPE (1-10mg/kg; ip) induced a significant dose dependent decrease in tumor growth by evaluating tumor volume and tumor weight. Histochemical and immunohistochemical analysis revealed that CAPE treatment significantly reduced the number of mitotic cells and proliferating cell nuclear antigen (PCNA)-positive cells in C6 glioma. These results suggest that CAPE presents an antitumor potential for glioma by inhibiting the growth of tumor cells.


Subject(s)
Caffeic Acids/pharmacology , Cell Proliferation/drug effects , Glioma/drug therapy , Neoplasms, Experimental/drug therapy , Phenylethyl Alcohol/analogs & derivatives , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin E/drug effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Dose-Response Relationship, Drug , Immunoblotting , Immunohistochemistry , In Vitro Techniques , Mice , Phenylethyl Alcohol/pharmacology , Rats , Retinoblastoma Protein/drug effects
16.
Int J Artif Organs ; 28(1): 44-50, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15742309

ABSTRACT

In mammalian cells, cellular differentiation into specific cell types is usually preceded by growth arrest. On the other hand, the induced differentiation may also be preceded by an enhanced G1-S transition of the cell cycle prior to the growth arrest. This suggests that an early increase in proliferation is in some way a prerequisite for subsequent differentiation. We therefore attempted to assess whether we could produce human hepatocytes with further differentiated functions by promoting G1-S transition in a butyrate-treated human hepatocyte cell line. A cyclin E-over-expressing cell line was established by transfecting human cyclin E cDNA. Upon butyrate treatment, the cyclin E-over-expressing cells exhibited a significantly increased albumin-secreting and ammonia-detoxifying capacity when compared to the control cells. In particular, the ornithine transcarbamylase activity was increased in these cells. Collectively, these results implicate that the cyclin E over-expression may augment the hepatocyte-specific functions during the butyrate-induced differentiation process of human hepatocytes by enhancing G1-S cell cycle transition.


Subject(s)
Butyrates/pharmacology , Cyclin E/drug effects , Hepatocytes/drug effects , Albumins/metabolism , Ammonia/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/analysis , Cell Differentiation/drug effects , Cell Line , Cell Proliferation , Cyclin E/genetics , DNA-Binding Proteins/analysis , G1 Phase/drug effects , Gene Expression Regulation/genetics , Hepatocyte Nuclear Factor 4 , Humans , Ornithine Carbamoyltransferase/drug effects , Phosphoproteins/analysis , S Phase/drug effects , Transcription Factor CHOP , Transcription Factors/analysis , Transfection , Urea/analysis
17.
Anticancer Drugs ; 15(6): 625-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15205608

ABSTRACT

3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors have been developed as lipid-lowering drugs, and are well recognized to reduce morbidity and mortality from coronary artery disease. Several recent experimental studies have focused on the inhibitory effects of HMG-CoA reductase inhibitor on tumor cell growth in vitro and in vivo, dependent on a direct effect on cancer cells. In the present study, we aimed to investigate the potential anti-angiogenic effect of pravastatin and its mechanism of action. Using human umbilical vein endothelial cells (HUVECs) as a model of angiogenesis, we investigated the effect of pravastatin on the various steps of angiogenesis, including endothelial cell proliferation and adhesion to extracellular matrix proteins. Pravastatin induced a dose-dependent decrease in the proliferative activity of endothelial cells, which was dependent on the cell cycle arrest to the G1 phase and not on cell apoptosis. G1 arrest was due to the decrease of cyclin D, cyclin E and cyclin-dependent kinase 2 levels. In addition, pravastatin inhibited tube formation on Matrigel and adhesion to extracellular matrix, but did not affect matrix metalloproteinase production. The present results demonstrate the anti-angiogenic activity of pravastatin and its potential use as an anticancer drug is suggested.


Subject(s)
Endothelium, Vascular/cytology , G1 Phase/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neovascularization, Pathologic/prevention & control , Pravastatin/pharmacology , Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , CDC2-CDC28 Kinases/drug effects , CDC2-CDC28 Kinases/genetics , CDC2-CDC28 Kinases/metabolism , Cell Division/drug effects , Cells, Cultured , Cyclin D , Cyclin E/drug effects , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclins/drug effects , Cyclins/genetics , Cyclins/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression/drug effects , Gene Expression/physiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 9/chemistry , Neovascularization, Pathologic/drug therapy , Polyisoprenyl Phosphates/pharmacology , Pravastatin/antagonists & inhibitors , Pravastatin/therapeutic use , Randomized Controlled Trials as Topic , Sesquiterpenes , Umbilical Veins/cytology , Umbilical Veins/drug effects
18.
J Oral Sci ; 46(1): 37-44, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15141722

ABSTRACT

It has previously been demonstrated that gingival fibroblasts derived from nifedipine-reactive patients (nifedipine responders) show a greater cell proliferation rate than those from nifedipine non-reactive patients (nifedipine non-responders) in the presence of 1 microM nifedipine. The aim of the present study was to characterize cell cycle differences between nifedipine responder and non-responder fibroblast cells and determine the effect of basic fibroblast growth factor (bFGF) on cell cycle progression. Further, the effect of bFGF on cyclins A, B1, D1, E, and CDKs 1, 2, 4, 6 mRNA expression in responder and non-responder cells was investigated. A population of nifedipine responder cells underwent progression to S and G2/M phases from G0/G1 phase in the presence of 10% fetal calf serum or 10 ng/ml bFGF was greater than nifedipine non-responder cells. mRNA expression of cyclins A, B1, D1, E and CDKs 1, 2, 4, 6 in the presence of 10 ng/ml bFGF was generally greater in nifedipine responder cells than non-responder cells. These results indicate that nifedipine responder cells may be more susceptible to growth factors such as bFGF with a resultant increase in expression of cyclins and CDKs in responder compared with non-responder cells.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Fibroblasts/drug effects , Gingiva/drug effects , Gingival Overgrowth/pathology , Nifedipine/adverse effects , Proto-Oncogene Proteins , CDC2 Protein Kinase/drug effects , CDC2-CDC28 Kinases/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cells, Cultured , Cyclin A/drug effects , Cyclin B/drug effects , Cyclin B1 , Cyclin D1/drug effects , Cyclin E/drug effects , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinases/drug effects , G1 Phase/drug effects , G2 Phase/drug effects , Gingiva/cytology , Gingival Overgrowth/physiopathology , Humans , Mitosis/drug effects , Resting Phase, Cell Cycle/drug effects , S Phase/drug effects
19.
Int J Cancer ; 109(5): 643-52, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14999769

ABSTRACT

The effects of GA, an ansamycin antibiotic in development as a lead anticancer drug, were studied in mouse BP-A31 fibroblasts and in human cancer-derived cell lines. GA and related molecules act by inhibiting the chaperone function of the Hsp90 protein through competition for ATP binding. The antiproliferative effects of GA have been attributed to destabilization of the Raf-1 protein, one of the targets of Hsp90, and to the resulting inhibition of MAPK. Addition of GA to BP-A31 cells, synchronously progressing through the G(1) phase, inhibited Rb hyperphosphorylation and G(1)/S transition irrespective of the time of addition. The G(1) arrest was accompanied by a progressive decrease in Raf-1 content, especially of the phosphorylated form; however, GA caused only partial inhibition of MAPK phosphorylation. We show that GA triggers a rapid and marked decrease in the kinase activity of the cyclin E/cdk2 complex coupled with a decline in both total and cdk2-associated cyclin E. In transient transfection experiments, inhibition of cyclin E expression by GA was correlated with inhibition of the transcriptional activity of the cyclin E gene promoter. Inhibition of cdk4 activity by GA was observed 3 hr after addition of the drug to late G(1) cells but not after a short (1 hr) exposure, as revealed by the phosphorylation of Rb on the Ser(780) residue. In human cancer-derived cell lines expressing or not a functional Rb protein, GA blocked proliferation and inhibited the transcriptional activity of the cyclin E gene promoter. In these cell lines, the antiproliferative effect of GA was not limited to the G(1) phase, suggesting the existence of multiple cellular targets of the drug.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , CDC2-CDC28 Kinases/drug effects , Cyclin E/drug effects , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Interphase/drug effects , Quinones/pharmacology , Benzoquinones , Blotting, Western , CDC2-CDC28 Kinases/metabolism , Cell Cycle/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lactams, Macrocyclic , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Chaperones/antagonists & inhibitors , Phosphorylation/drug effects , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Retinoblastoma Protein/drug effects , Retinoblastoma Protein/metabolism , Transfection , Tumor Cells, Cultured
20.
Am J Pathol ; 164(3): 937-46, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14982847

ABSTRACT

Lipoxins (LX) are endogenously produced eicosanoids with a spectrum of bioactions that suggest anti-inflammatory, pro-resolution roles for these agents. Mesangial cell (MC) proliferation plays a pivotal role in the pathophysiology of glomerular inflammation and is coupled to sclerosis and tubulointerstitial fibrosis. We have previously reported that LXA4 acts through a specific G-protein-coupled-receptor (GPCR) to modulate MC proliferation in response to the proinflammatory mediators LTD4 and platelet-derived growth factor (PDGF). Further investigations revealed that these effects were mediated by modulation of receptor tyrosine kinase activity. Here we have explored the underlying mechanisms and report inhibition of growth factor (PDGF; epithelial growth factor) activation of Akt/PKB by LXA4. LXA4 (10 nmol/L) modulates PDGF-induced (10 ng/ml, 24 hours) decrements in the levels of cyclin kinase inhibitors p21Cip1 and p27Kip1. PDGF-induced increases in CDK2-cyclin E complex formation are also inhibited by LXA4. The potential of LXA4 as an anti-inflammatory therapeutic is compromised by its degradation; this has been circumvented by synthesis of stable analogs. We report that 15-(R/S)-methyl-LXA4 and 16-phenoxy-LXA4 mimic the native compound with respect to modulation of cell proliferation and PDGF-induced changes in cell cycle proteins. In vivo, MC proliferation in response to PDGF is associated with TGFbeta1 production and the subsequent development of renal fibrosis. Here we demonstrate that prolonged (24 to 48 hours) exposure to PDGF is associated with autocrine TGFbeta1 production, which is significantly reduced by LXA4. In aggregate these data demonstrate that LX inhibit PDGF stimulated proliferation via modulation of the PI-3-kinase pathway preventing mitogen-elicited G1-S phase progression and suggest the therapeutic potential of LX as anti-fibrotic agents.


Subject(s)
Cell Cycle/drug effects , Glomerular Mesangium/drug effects , Growth Substances/metabolism , Lipoxins/pharmacology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/drug effects , Blotting, Western , CDC2-CDC28 Kinases/drug effects , CDC2-CDC28 Kinases/metabolism , Cell Cycle Proteins/drug effects , Cells, Cultured , Cyclin E/drug effects , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/drug effects , Enzyme Activation/drug effects , Glomerular Mesangium/cytology , Humans , Immunohistochemistry , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-akt , Tumor Suppressor Proteins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...