Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Adv Protein Chem Struct Biol ; 135: 125-177, 2023.
Article En | MEDLINE | ID: mdl-37061330

Serine/threonine kinases called cyclin-dependent kinases (CDKs) interact with cyclins and CDK inhibitors (CKIs) to control the catalytic activity. CDKs are essential controllers of RNA transcription and cell cycle advancement. The ubiquitous overactivity of the cell cycle CDKs is caused by a number of genetic and epigenetic processes in human cancer, and their suppression can result in both cell cycle arrest and apoptosis. This review focused on CDKs, describing their kinase activity, their role in phosphorylation inhibition, and CDK inhibitory proteins (CIP/KIP, INK 4, RPIC). We next compared the role of different CDKs, mainly p21, p27, p57, p16, p15, p18, and p19, in the cell cycle and apoptosis in cancer cells with respect to normal cells. The current work also draws attention to the use of CDKIs as therapeutics, overcoming the pharmacokinetic barriers of pan-CDK inhibitors, analyze new chemical classes that are effective at attacking the CDKs that control the cell cycle (cdk4/6 or cdk2). It also discusses CDKI's drawbacks and its combination therapy against cancer patients. These findings collectively demonstrate the complexity of cancer cell cycles and the need for targeted therapeutic intervention. In order to slow the progression of the disease or enhance clinical outcomes, new medicines may be discovered by researching the relationship between cell death and cell proliferation.


Cell Cycle Proteins , Cyclin-Dependent Kinases , Humans , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cell Cycle , Apoptosis
2.
Contrast Media Mol Imaging ; 2022: 6384334, 2022.
Article En | MEDLINE | ID: mdl-36134118

Most studies have focused on the protective effects of ginkgolide A against ischemia/reperfusion-induced cardiomyopathy and injury of the brain, liver, and other organs, but there are few reports about the protection of lung tissues. This study was designed to clarify the protection of ginkgolide A against lipopolysaccharide (LPS)-induced pulmonary microvascular endothelial cell (PMVEC) injury. PMVECs were extracted and fell into control, LPS, and ginkgolide A groups. Next, we delved into the growth activity and apoptosis rate of cells via the CCK-8 assay and Hoechst staining, independently. Beyond that, western blotting (WB) was implemented for measurement of the expressions of cyclin D1, cyclin-dependent kinase 4 (CDK4), and CDK inhibitor (p21) that pertained to the cell cycle. The target sites of ginkgolide A were confirmed by miRNA array and real-time quantitative PCR. The relationship between miR-224 and p21 was analyzed using dual-luciferase reporter gene assay. Compared with the control group, the LPS group and ginkgolide A group had significantly decreased cell growth activity and relative expressions of cyclin D1 and CDK4 and elevated apoptosis rate and p21 expression. Pronounced elevations were observable in the cell growth activity and expressions of cyclin D1, CDK4, and p21, while the ginkgolide A group presented with a reduced apoptosis rate in comparison with the LPS group (P < 0.05). MiR-224 was the target of ginkgolide A, which had targeted regulatory effects on p21. Ginkgolide A can modulate miR-224 expression and regulate p21 expression in a targeted manner to enhance the resistance of PMVECs to LPS-induced cell apoptosis.


Cyclin D1 , MicroRNAs , Apoptosis , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 4/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Endothelial Cells/metabolism , Ginkgolides , Lactones , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , MicroRNAs/pharmacology
3.
J Neurosci ; 42(14): 2905-2916, 2022 04 06.
Article En | MEDLINE | ID: mdl-35232758

Paternal environmental perturbations can influence the physiology and behavior of offspring. For example, our previous work showed reduced cocaine reinforcement in male, but not female, progeny of rat sires that self-administered cocaine. The information transfer from sire to progeny may occur through epigenetic marks in sperm, encompassing alterations in small noncoding RNAs, including microRNAs (miRNAs) and/or DNA methylation. Here, no reliable changes in miRNAs in the sperm of cocaine- relative to saline-experienced sires were identified. In contrast, 272 differentially methylated regions were observed in sperm between these groups. Two hypomethylated promoter regions in the sperm of cocaine-experienced rats were upstream of cyclin-dependent kinase inhibitor 1a (Cdkn1a). Cdkn1a mRNA also was selectively increased in the NAc of cocaine-sired male (but not female) offspring. Cocaine self-administration also enhanced Cdkn1a expression in the accumbens of cocaine-sired rats. These results suggest that changes in Cdkn1a may play a role in the reduced cocaine reinforcing efficacy observed in cocaine-sired male rats. Introducing a 90 d delay between sire self-administration and breeding reversed both cocaine resistance and the increase in accumbens Cdkn1a mRNA in male offspring, indicating that cocaine-induced epigenetic modifications are eliminated with sperm turnover. Collectively, our results indicate that cocaine self-administration produces hypomethylation of Cdkn1a in sperm and a selective increase in the expression of this gene in the NAc of male offspring, which is associated with blunted cocaine reinforcement.SIGNIFICANCE STATEMENT The relatively new field of transgenerational epigenetics explores the effects of environmental perturbations on offspring behavior and physiology. Our prior work in rats indicated that male, but not female, progeny of sires that self-administered cocaine displayed reduced cocaine reinforcement. The information transfer from sire to progeny may occur through heritable epigenetic marks in sperm, including DNA methylation. The present findings revealed two hypomethylated promoter regions upstream of the Cdkn1a gene in sire sperm. Remarkably, Cdkn1a expression was selectively decreased in offspring NAc, a brain region that regulates cocaine reinforcement.


Cocaine , Cyclin-Dependent Kinase Inhibitor p21 , DNA Methylation , Epigenesis, Genetic , Spermatozoa , Animals , Cocaine/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , DNA Methylation/drug effects , Male , MicroRNAs/metabolism , Nucleus Accumbens , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Spermatozoa/metabolism
4.
Bioorg Chem ; 120: 105634, 2022 03.
Article En | MEDLINE | ID: mdl-35114524

Novel (Z)-3-((4,6-diphenylpyrimidin-2-ylamino)methylene)-2,3-dihydrochromen-4-one derivatives were designed and synthesized to find chemotherapeutic agents. Derivative 9 was selected based on its clonogenicity against cancer cells and synthetic yield for further biological experiments. It showed decreases in aurora kinase A, B, and C phosphorylation from western blot analysis. Derivative 9 upregulated the expression of G1 cell cycle inhibitory proteins including p21 and p27, and G1 progressive cyclin D1, and downregulated G1-to-S progressive cyclins, resulting in cell cycle arrest at the G1/S boundary. It stimulated the cleavage of caspase-9, -3, -7, and poly (ADP-ribose) polymerase, resulting in triggering apoptosis through a caspase-dependent pathway. In addition, derivative 9 inhibited in vivo tumor growth in a syngeneic tumor implantation mouse model. The findings of this study suggest that derivative 9 can be considered as a lead compound for chemotherapeutic agents.


Antineoplastic Agents , Caspases , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Caspases/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Mice , Poly(ADP-ribose) Polymerases/metabolism
5.
Comput Biol Med ; 135: 104640, 2021 08.
Article En | MEDLINE | ID: mdl-34261004

Cisplatin is a DNA-damaging chemotherapeutic agent used for treating cancer. Based on cDNA dataset analysis, we investigated how cisplatin modified gene expression and observed cisplatin-induced dysregulation and system-level variations relating to insulin resistance and type 2 diabetes mellitus (T2DM). T2DM is a multifactorial disease affecting 462 million people in the world, and drug-induced T2DM is a serious issue. To understand this etiology, we designed an integrative, system-level study to identify associations between cisplatin-induced differentially expressed genes (DEGs) and T2DM. From a list of differential expressed genes, cisplatin downregulated the cyclin-dependent kinase inhibitor 1 (CDKN1A), tumor necrosis factor (FAS), and sestrin-1 (SESN1) genes responsible for modifying signaling pathways, including the p53, JAK-STAT, FOXO, MAPK, mTOR, P13-AKT, Toll-like receptor (TLR), adipocytokine, and insulin signaling pathways. These enriched pathways were expressively associated with the disease. We observed significant gene signatures, including SMAD3, IRS, PDK1, PRKAA1, AKT, SOS, RAS, GRB2, MEK1/2, and ERK, interacting with source genes. This study revealed the value of system genetics for identifying the cisplatin-induced genetic variants responsible for the progression of T2DM. Also, by cross-validating gene expression data for T2DM islets, we found that downregulating IRS and PRK families is critical in insulin and T2DM signaling pathways. Cisplatin, by inhibiting CDKN1A, FAS, and SESN1, promotes IRS and PRK activity in a similar way to rosiglitazone (a popular drug used for T2DM treatment). Our integrative, network-based approach can help in understanding the drug-induced pathophysiological mechanisms of diabetes.


Antineoplastic Agents , Diabetes Mellitus, Type 2 , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Heat-Shock Proteins/pharmacology , Heat-Shock Proteins/therapeutic use , Humans , Insulin/pharmacology , Signal Transduction
6.
Crit Rev Eukaryot Gene Expr ; 26(1): 49-62, 2016.
Article En | MEDLINE | ID: mdl-27278885

Cancer develops due to an imbalance between cell proliferation and cell death. Various mechanisms of carcinogenesis as well as of novel anticancer agents that could be targeted for the treatment of cancer have been proposed by different studies. Among these, p21 is recognized as a potent cyclin-dependent kinase inhibitor that facilitates cell-cycle arrest by interacting with different stimuli such as p53, DNA repair process, CDK, E2F1, MYC, PCNA, STAT3 AP4, proteasomes, K1F, CDX2, and ER-α. p21 acts both as a tumor-suppressor gene and an inhibitor of apoptosis by interacting with various molecules and transition factors. In this review, we discuss the complex role of p21 in the development of cancer and as a target in its treatment. We conclude that, in the future, the tumor-suppressor activity of p21 should be the focus of a novel treatment strategies, which may lead to the devolvement of new and selective anti-cancer agents for the targeted therapy of cancers.


Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinase Inhibitor p21/therapeutic use , Disease Progression , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/metabolism
7.
J Microbiol Biotechnol ; 25(3): 413-7, 2015 Mar.
Article En | MEDLINE | ID: mdl-25674801

Recently, we isolated HY253, a novel decahydrofluorene analog with a molecular structure of 7,8a-divinyl-2,4a,4b,5,6,7,8,8a,9,9a-decahydro-1H-fluorene-2,4a,4b,9a-tetraol from the roots of Aralia continentalis, which is known as Dokwhal, a traditional medicinal herb. Moreover, we previously reported its cytotoxic activity on cancer cell proliferation in human lung cancer A549 and cervical cancer HeLa cells. The current study aimed to evaluate its detailed molecular mechanisms in cell cycle arrest and apoptotic induction in human hepatocellular carcinoma HepG2 cells. Flow cytometric analysis of HepG2 cells treated with 60 micrometer HY253 revealed appreciable cell cycle arrest at the G1 phase via inhibition of Rb phosphorylation and down-regulation of cyclin D1. Furthermore, using western blots, we found that up-regulation of cyclin-dependent kinase inhibitors, such as p21(CIP1) and p27(KIP1), was associated with this G1 phase arrest. Moreover, TUNEL assay and immunoblottings revealed apoptotic induction in HepG2 cells treated with 100 micrometer HY253 for 24 h, which is associated with cytochrome c release from mitochondria, via down-regulation of anti-apoptotic Bcl-2 protein, which in turn resulted in activation of caspase-9 and -3, and proteolytic cleavage of poly(ADP-ribose) polymerase (PARP). Accordingly, we suggest that HY253 may be a potent chemotherapeutic hit compound for treating human liver cancer cells via up-regulation and activation of the p53 gene.


Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Fluorenes/pharmacology , Liver Neoplasms/drug therapy , Aralia/chemistry , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cytochromes c/metabolism , Flow Cytometry , G1 Phase/drug effects , Hep G2 Cells , Humans , Mitochondria/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Suppressor Protein p53/genetics
8.
Cancer Lett ; 348(1-2): 177-84, 2014 Jun 28.
Article En | MEDLINE | ID: mdl-24680816

This work describes the effects of elastin-like polypeptide (ELP) with the p21(Waf1/Cip1)-derived cell cycle inhibitory peptide (p21) on pancreatic tumor cells with gemcitabine. The thermo-responsive property of ELP permits use of a mild, local hyperthermia to target tumors for the transport of chemotherapeutics. In this study, a p21-ELP construct with Bac cell penetrating peptide was designed, and its anticancer activities in pancreatic cancer cell lines was examined. In combination with gemcitabine, the peptide demonstrated enhanced in vitro cytotoxicity as well as tumor growth inhibition in an animal model. Our data suggest that this ELP construct, with gemcitabine, may improve pancreatic cancer therapy.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hypothermia, Induced , Pancreatic Neoplasms/drug therapy , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Elastin/metabolism , Elastin/pharmacology , Feasibility Studies , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Retinoblastoma Protein/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Time Factors , Xenograft Model Antitumor Assays , Gemcitabine
9.
Cell Death Dis ; 4: e859, 2013 Oct 17.
Article En | MEDLINE | ID: mdl-24136223

The p90 ribosomal S6 kinase (RSK) family is a group of highly conserved Ser/Thr kinases that promote cell proliferation, growth, motility and survival. As they are almost exclusively activated downstream of extracellular signal-regulated kinases 1 and 2 (ERK1/2), therapeutic intervention by RSK inhibition is less likely to produce such severe side effects as those observed following inhibition of the upstream master regulators Raf, MEK and ERK1/2. Here, we report that BI-D1870, a potent small molecule inhibitor of RSKs, induces apoptosis, although preferentially, in a p21-deficient background. On the other hand, BI-D1870 also induces a strong transcription- and p53-independent accumulation of p21 protein and protects cells from gamma irradiation (γIR)-induced apoptosis, driving them into senescence even in the absence of γIR. Although we identified p21 in in vitro kinase assays as a novel RSK substrate that specifically becomes phosphorylated by RSK1-3 at Ser116 and Ser146, RNA-interference, overexpression and co-immunoprecipitation studies as well as the use of SL0101, another specific RSK inhibitor, revealed that BI-D1870 mediates p21 accumulation via a yet unknown pathway that, besides its off-site targets polo-like kinase-1 and AuroraB, also does also not involve RSKs. Thus, this novel off-target effect of BI-D1870 should be taken into serious consideration in future studies investigating the role of RSKs in cellular signaling and tumorigenesis.


Apoptosis/radiation effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gamma Rays , Pteridines/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Aurora Kinases/metabolism , Benzopyrans/pharmacology , Cell Cycle Proteins/metabolism , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Gene Knockdown Techniques , HCT116 Cells , Humans , Isoenzymes/metabolism , Monosaccharides/pharmacology , Phosphorylation/drug effects , Phosphorylation/radiation effects , Phosphoserine/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Stress, Physiological/drug effects , Stress, Physiological/radiation effects , Substrate Specificity/drug effects , Substrate Specificity/radiation effects , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects , Polo-Like Kinase 1
10.
Exp Biol Med (Maywood) ; 238(6): 589-99, 2013 Jun.
Article En | MEDLINE | ID: mdl-23918871

Triple negative breast cancer (TNBC) represents approximately 15% of the newly diagnosed cancers worldwide and is characterized by tissue lacking in estrogen, progesterone and human epidermal growth factor receptors. TNBC disproportionately affects younger women and women of colour, and new treatments are needed. The opioid growth factor (OGF) - opioid growth factor receptor (OGFr) axis is a determinant of cell proliferation in neoplasia, and OGF is an endogenously produced pentapeptide that inhibits cell replication by interacting with OGFr and upregulating cyclin-dependent inhibitory kinase pathways thus reducing DNA synthesis. In these studies we investigated the presence and function of the OGF-OGFr axis in two human TNBC cell lines, as well as in breast cancer cell lines containing hormonal receptors. TNBC cell lines MDA-MD-231 and BT-20, as well as human breast cancer cells SK-BR-3 and MCF-7, were examined for the presence of pentapeptide and receptors, as well as their response to OGF. Specificity of peptide and receptor was confirmed by antibody neutralization and molecular studies to knockdown classical receptor protein. The requirement for protein transcription and translation and RNA transcription were investigated. Growth of TNBC cells in the presence of OGF and standard of care chemotherapeutic agent paclitaxel was evaluated to determine both efficacy and protective effects against toxicity. OGF treatment inhibited TNBC cells in a dosage related, receptor mediated, and reversible manner. OGF was the specific endogenous opioid to inhibit cell proliferation, and this was mediated by p21 cyclin dependent inhibitory kinase pathways, and required protein and RNA synthesis. OGFr was the specific receptor involved; both peptide and receptor were detected in all four cell lines. OGF treatment inhibited growth of all cancer cell lines evaluated, and reduced cell death in cultures exposed to paclitaxel. The OGF-OGFr axis is present and functioning in TNBC cell lines, and provides a novel biological pathway as potential therapy.


Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Narcotic Antagonists , Opioid Peptides/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Naltrexone/pharmacology , Receptors, Opioid
11.
Int J Radiat Oncol Biol Phys ; 85(4): 1119-26, 2013 Mar 15.
Article En | MEDLINE | ID: mdl-23200172

PURPOSE: To elucidate the mechanism of the therapeutic efficacy of targeted α-particle radiation therapy using (212)Pb-TCMC-trastuzumab together with gemcitabine for treatment of disseminated peritoneal cancers. METHODS AND MATERIALS: Mice bearing human colon cancer LS-174T intraperitoneal xenografts were pretreated with gemcitabine, followed by (212)Pb-TCMC-trastuzumab and compared with controls. RESULTS: Treatment with (212)Pb-TCMC-trastuzumab increased the apoptotic rate in the S-phase-arrested tumors induced by gemcitabine at earlier time points (6 to 24 hours). (212)Pb-TCMC-trastuzumab after gemcitabine pretreatment abrogated G2/M arrest at the same time points, which may be associated with the inhibition of Chk1 phosphorylation and, in turn, cell cycle perturbation, resulting in apoptosis. (212)Pb-TCMC-trastuzumab treatment after gemcitabine pretreatment caused depression of DNA synthesis, DNA double-strand breaks, accumulation of unrepaired DNA, and down-regulation of Rad51 protein, indicating that DNA damage repair was blocked. In addition, modification in the chromatin structure of p21 may be associated with transcriptionally repressed chromatin states, indicating that the open structure was delayed at earlier time points. CONCLUSION: These findings suggest that the cell-killing efficacy of (212)Pb-TCMC-trastuzumab after gemcitabine pretreatment may be associated with abrogation of the G2/M checkpoint, inhibition of DNA damage repair, and chromatin remodeling.


Antibodies, Monoclonal, Humanized/pharmacology , Cell Death/physiology , DNA Repair/radiation effects , Deoxycytidine/analogs & derivatives , Heterocyclic Compounds/pharmacology , Isothiocyanates/pharmacology , Lead Radioisotopes/pharmacology , Rad51 Recombinase/antagonists & inhibitors , Radioimmunotherapy/methods , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis/radiation effects , Cell Line, Tumor , Checkpoint Kinase 1 , Chromatin/drug effects , Chromatin/radiation effects , Combined Modality Therapy/methods , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , DNA Repair/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Female , G2 Phase/physiology , G2 Phase/radiation effects , Humans , In Situ Nick-End Labeling/methods , Linear Energy Transfer/physiology , Mice , Mice, Nude , Mitosis/radiation effects , Peritoneal Neoplasms/radiotherapy , Phosphorylation/radiation effects , Protein Kinases/immunology , Protein Kinases/metabolism , Radiation-Sensitizing Agents/pharmacology , Trastuzumab , Xenograft Model Antitumor Assays/methods , Gemcitabine
12.
Anal Biochem ; 427(1): 69-78, 2012 Aug 01.
Article En | MEDLINE | ID: mdl-22522186

Proliferating cell nuclear antigen (PCNA) is a critical player in cell proliferation. It interacts with a myriad of cellular proteins in genomic DNA replication and cell cycle control. This makes PCNA an attractive target for developing antiproliferative therapeutics. Indeed, the binding of a human tumor suppressor protein, p21, to PCNA contributes to its antiproliferative effect in cells. In this work, we report a fluorescence polarization-based binding assay for determining the affinity between the p21 peptide and human PCNA. To improve the potency of the p21-based PCNA antagonist, we exploited the homotrimeric structure of PCNA and developed multivalent peptide-based PCNA antagonists. The di- and trivalent p21-based antagonists bind to PCNA with low nanomolar dissociation constant. Moreover, we show that the multivalent PCNA antagonists inhibited PCNA-dependent DNA synthesis in a human cell extract with improved avidity when compared with the monovalent p21 peptide. The fluorescence polarization assay holds promise for the discovery of potent small-molecule PCNA inhibitors given its ready adaptability to a high-throughput screening format.


Cyclin-Dependent Kinase Inhibitor p21 , Peptides , Proliferating Cell Nuclear Antigen/analysis , Amino Acid Sequence , Binding Sites , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/chemical synthesis , Cyclin-Dependent Kinase Inhibitor p21/chemistry , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , DNA Replication/drug effects , Fluorescence , HeLa Cells , Humans , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Proliferating Cell Nuclear Antigen/chemistry , Protein Binding , Protein Conformation
13.
Tumori ; 98(6): 804-11, 2012 Nov.
Article En | MEDLINE | ID: mdl-23389370

AIMS AND BACKGROUND: To study the inhibitory effect of p21WAF1/CIP1 activation by saRNA on the growth of human pancreatic cancer cells PANC-1 in vitro and in vivo. METHODS AND STUDY DESIGN: A dsRNA (dsP21) targeting the p21WAF1/CIP1 gene promoter at position-322 relative to the transcription start site was transfected into PANC-1 cells. Expression of mRNA and protein was evaluated by semiquantitative RT-PCR and Western blotting. Proliferation of PANC-1 cells was measured by the MTT method, and the apoptosis rate was detected by flow cytometry. PANC-1 cells were transplanted subcutaneously in nude mice, and the inhibitory effect of dsP21 on tumor growth was observed. RESULTS: The introduction of dsP21 was shown to efficiently up-regulate expression of the p21WAF1/CIP1 gene in PANC-1 cells according to the results of RT-PCR and Western blotting (P <0.01, compared with controls). The inhibitory effect on cell proliferation was confirmed by the MTT test (P <0.05, compared with controls). The apoptosis rate of PANC-1 cells treated with dsP21 was significantly higher than that of the control cells (P <0.01). Our experimental data showed that dsP21-mediated up-regulation of p21 expression exerted an apparent growth inhibitory effect on PANC-1 cells in vivo. CONCLUSIONS: dsP21 targeting the p21WAF1/CIP1 gene promoter can specifically up-regulate expression of the p21WAF1/CIP1 gene in PANC-1 cells. It therefore has a substantially inhibitory effect on cell proliferation in vitro and in vivo and can be used as a new method and material for the gene therapy of pancreatic cancer.


Cyclin-Dependent Kinase Inhibitor p21/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Small Interfering/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Up-Regulation , Xenograft Model Antitumor Assays
14.
J Virol ; 85(13): 6774-83, 2011 Jul.
Article En | MEDLINE | ID: mdl-21525341

Human herpesvirus 6 (HHV-6) is an important immunosuppressive and immunomodulatory virus that primarily infects immune cells and strongly suppresses the proliferation of infected cells. However, the mechanisms responsible for the regulation and suppression mediated by HHV-6 are still unknown. In this study, we examined the ability of HHV-6A to manipulate cell cycle progression in infected cells and explored the potential molecular mechanisms. We demonstrated that infection with HHV-6A imposed a growth-inhibitory effect on HSB-2 cells by inducing cell cycle arrest at the G(2)/M phase. We then showed that the activity of the Cdc2-cyclin B1 complex was significantly decreased in HHV-6A-infected HSB-2 cells. Furthermore, we found that inactivation of Cdc2-cyclin B1 in HHV-6A-infected cells occurred through the inhibitory Tyr15 phosphorylation resulting from elevated Wee1 expression and inactivated Cdc25C. The reduction of Cdc2-cyclin B1 activity in HHV-6-infected cells was also partly due to the increased expression of the cell cycle-regulatory molecule p21 in a p53-dependent manner. In addition, HHV-6A infection activated the DNA damage checkpoint kinases Chk2 and Chk1. Our data suggest that HHV-6A infection induces G(2)/M arrest in infected T cells via various molecular regulatory mechanisms. These results further demonstrate the potential mechanisms involved in immune suppression and modulation mediated by HHV-6 infection, and they provide new insights relevant to the development of novel vaccines and immunotherapeutic approaches.


Cell Division/drug effects , Cell Proliferation/drug effects , G2 Phase/drug effects , Herpesvirus 6, Human/pathogenicity , T-Lymphocytes/virology , CDC2 Protein Kinase , Cell Cycle/drug effects , Cyclin B/genetics , Cyclin B/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cyclin-Dependent Kinases , Down-Regulation , Flow Cytometry , Humans , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/pharmacology , Up-Regulation
15.
Cell Signal ; 22(12): 1864-73, 2010 Dec.
Article En | MEDLINE | ID: mdl-20667470

The p75 neurotrophin receptor (p75(NTR)) plays a critical role in various neuronal and non-neuronal cell types by regulating cell survival, differentiation and proliferation. To evaluate the influence of p75(NTR) in breast cancer development, we have established and characterized breast cancer cells which stably overexpress p75(NTR). We showed that p75(NTR) overexpression per se promoted cell survival to apoptogens with a concomitant slowdown of cell growth. The pro-survival effect is associated with an increased expression of the inhibitor of apoptosis protein-1 (c-IAP1), a decrease of TRAIL-induced cleavage of PARP, procaspase 9 and procaspase 3, and a decrease of cytochrome C release from the mitochondria. The anti-proliferative effect is due to a cell accumulation in G0/G1, associated with a decrease of Rb phosphorylation and an increase of p21(waf1). Interestingly, inhibition of p21(waf1) with siRNA not only restores proliferation but also abolishes the pro-survival effect of p75(NTR), indicating the key role of p21(waf1) in the biological functions of p75(NTR). Finally, using a SCID mice xenograft model, we showed that p75(NTR) overexpression favors tumor growth and strongly increases tumor resistance to anti-tumoral treatment. Together, our findings suggest that p75(NTR) overexpression in breast tumor cells could favor tumor survival and contribute to tumor resistance to drugs. This provides a rationale to consider p75(NTR) as a potential target for the future design of innovative therapeutic strategies.


Breast Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Receptor, Nerve Growth Factor/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Cytochrome c Group/metabolism , Female , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, SCID , Mitochondria/genetics , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Xenograft Model Antitumor Assays/methods
16.
Oncogene ; 29(13): 1952-62, 2010 Apr 01.
Article En | MEDLINE | ID: mdl-20062073

Knockout of heat shock protein Hsp72 was shown to promote chromosomal instability and increase radiation sensitivity of mouse fibroblasts. Here, we report that downregulation of Hsp72 in human tumor cells leads to suppression of a specific branch of the DNA damage response (DDR) that facilitates DNA repair following genotoxic insults, that is, reduced accumulation of the phosphorylated form of histone H2AX (gammaH2AX). This inhibition was due to decreased expression of H2AX as well as higher rate of gammaH2AX dephosphorylation. Formation of gammaH2AX and MDC1 radiation-induced foci was impaired in Hsp72-depleted cells, which in turn enhanced DNA damage, resulting in sensitization of cells to gamma-radiation and doxorubicin. These effects of Hsp72 knockdown were dependent on activation of the p53/p21-signaling pathway. Overall, permanent activation of the p53/p21 signaling in Hsp72-depleted cells specifically impaired the gammaH2AX pathway of the DDR, enhanced DNA damage following genotoxic insults, and led to further stimulation of the p53/p21 pathway, thus creating a positive feedback loop. The resulting strong induction of p21 precipitated senescence following exposure to DNA-damaging agents, thus accounting for higher sensitivity of cells to genotoxic stresses.


Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA Damage , HSP72 Heat-Shock Proteins/deficiency , Histones/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , DNA Repair/genetics , Down-Regulation , Gene Knockout Techniques , HSP72 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/metabolism , Histones/genetics , Humans , Mice , Protein Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
17.
J Med Chem ; 53(1): 492-8, 2010 Jan 14.
Article En | MEDLINE | ID: mdl-19911822

It is important to develop G-quadruplex binding agents that can discriminate between different quadruplexes. Recently we reported the first example that a chiral supramolecular complex can selectively stabilize human telomeric G-quadruplex among different G-quadruplex and duplex DNA, and the two enantiomers show different inhibition effect on telomerase activity. Here, we report that DNA loop sequence can be determinant for this chiral complex G-quadruplex selectivity. Adenine in the diagonal loop plays an important role in G-quadruplex hybrid structural transition, thus, it strongly influences the chiral complex induced DNA structural transition. The complex's preference for human telomeric DNA and its chiral selectivity prompted us to investigate whether the two enantiomers, M and P, can show different effects on cancer cells. The P enantiomer's chiral selectivity has been demonstrated in cancer cells by telomere shortening, beta-galactosidase activity, and up-regulation of cyclin-dependent kinase inhibitors p16 and p21.


Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , DNA/drug effects , DNA/genetics , G-Quadruplexes/drug effects , Macromolecular Substances/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p16/chemistry , Cyclin-Dependent Kinase Inhibitor p21/chemistry , DNA/chemistry , Drug Screening Assays, Antitumor , Humans , Macromolecular Substances/chemistry , Molecular Structure , Stereoisomerism , Structure-Activity Relationship , Substrate Specificity , Telomerase/antagonists & inhibitors
18.
Cell Cycle ; 8(22): 3777-81, 2009 Nov 15.
Article En | MEDLINE | ID: mdl-19855165

Cellular senescence is characterized by irreversible loss of proliferative potential and a large, flat cell morphology. Ectopic p21 and doxorubicin induced cellular senescence in HT1080 and WI-38-tert cell lines. In the same cell lines, the Mdm2 inhibitor nutlin-3a induced p53 but, unexpectedly, caused quiescence (reversible arrest) with a small cell morphology. We discuss that Mdm antagonists could be used in combination with chemotherapy to reversibly arrest normal cells, thus protecting them during chemotherapy of cancer (cyclotherapy).


Cell Cycle/physiology , Cell Proliferation , Drug Therapy/methods , Imidazoles/metabolism , Piperazines/metabolism , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/physiology , Colony-Forming Units Assay , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Doxorubicin/pharmacology , Flow Cytometry , Humans , Immunoblotting , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase
19.
Plast Reconstr Surg ; 123(2 Suppl): 149S-157S, 2009 Feb.
Article En | MEDLINE | ID: mdl-19182674

BACKGROUND: Tissue engineering is often limited by the time required for culture expansion of cells necessary for scaffold seeding. Cell cycle regulators control entry and exit into the cell cycle and as such regulate cellular proliferation rates. The authors hypothesized that transient alteration in cell cycle regulators can be utilized as a means to accelerate stem cell proliferation. METHODS: Mesenchymal stem cells were harvested from wild-type mice and mice deficient in the cell cycle regulator p21. Wild-type cells were treated with small interfering RNA against p21 in two- or three-dimensional cultures in vitro. Cellular proliferation and the potential for cellular differentiation into the bone or fat lineage were assessed. RESULTS: Mesenchymal stem cells treated with small interfering RNA targeting p21 demonstrated a significant decrease in p21 protein and mRNA expression 96 hours after treatment. They also proliferated significantly faster than control cells (2.5 to three times) in both two- and three-dimensional culture. Similarly, cells harvested from p21-deficient mice demonstrated a significant acceleration in cellular proliferation. Inhibition of p21 expression was not associated with significant changes in spontaneous cellular differentiation. However, transient p21 inhibition promoted both osteoblastic and adipogenic differentiation when cells were exposed to differentiation medium. CONCLUSIONS: Transient inhibition of the cell cycle regulator p21 results in significant acceleration of mesenchymal stem cell proliferation without promoting spontaneous cellular differentiation. Exposure to differentiation medium results in increased cellular differentiation toward the osteoblast and fat lineage. Manipulation of cell cycle regulators may represent a novel means by which stem cell proliferation can be accelerated, thereby decreasing the time required for scaffold synthesis in tissue engineering.


Cyclin-Dependent Kinase Inhibitor p21/pharmacology , Mesenchymal Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation , Mice
...