Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
Add more filters

Publication year range
1.
PLoS Genet ; 14(12): e1007790, 2018 12.
Article in English | MEDLINE | ID: mdl-30557355

ABSTRACT

Accumulating evidence indicates that miRNAs can be promising diagnostic and/or prognostic markers for various cancers. In this study, we identified a novel miRNA, miR-3607-3p, and its targets in non-small cell lung cancer (NSCLC). The expression of miR-3607-3p was measured and its correlation with patient prognosis was determined. Ectopic expression in NSCLC cells, xenografts, and metastasis models was used to evaluate the effects of miR-3607-3p on proliferation and migration of NSCLC. Luciferase assay and western blotting were performed to validate the potential targets of miR-3607-3p after preliminary screening by microarray analysis and computer-aided algorithms. We demonstrated that miR-3607-3p was downregulated in NSCLC tissues and that miR-3607-3p might act as an independent predictor for overall survival in NSCLC. Moreover, serum miR-3607-3p may be a novel and stable marker for NSCLC. We found that overexpression of miR-3607-3p inhibited cell proliferation, colony formation, migration and invasion, and hampered the cell cycle of NSCLC cell lines in vitro. Our results suggested that miR-3607-3p directly targets TGFBR1 and CCNE2. In accordance with in vitro studies, we confirmed that miR-3607-3p functions as a potent suppressor miRNA of NSCLC. We showed that miR-3607-3p agomir could reduce tumor growth and inhibit TGFBR1 and CCNE2 protein expression. Taken together, our findings indicate that miR-3607-3p can inhibit NSCLC cell growth and metastasis by targeting TGFBR1 and CCNE2 protein expression, and provide new evidence of miR-3607-3p as a potential non-invasive biomarker and therapeutic target for NSCLC.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Cyclins/antagonists & inhibitors , Lung Neoplasms/genetics , Lung Neoplasms/therapy , RNA, Small Nucleolar/genetics , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Aged , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclins/genetics , Down-Regulation , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis , Prognosis , RNA, Small Nucleolar/antagonists & inhibitors , RNA, Small Nucleolar/blood , Receptor, Transforming Growth Factor-beta Type I/genetics
2.
J Biol Chem ; 293(32): 12542-12562, 2018 08 10.
Article in English | MEDLINE | ID: mdl-29907572

ABSTRACT

The avian reovirus p17 protein is a nucleocytoplasmic shuttling protein. Although we have demonstrated that p17 causes cell growth retardation via activation of p53, the precise mechanisms remain unclear. This is the first report that avian reovirus p17 possesses broad inhibitory effects on cell cycle CDKs, cyclins, CDK-cyclin complexes, and CDK-activating kinase activity in various mammalian, avian, and cancer cell lines. Suppression of CDK activity by p17 occurs by direct binding to CDKs, cyclins, and CDK-cyclin complexes; transcriptional down-regulation of CDKs; cytoplasmic retention of CDKs and cyclins; and inhibition of CDK-activating kinase activity by promoting p53-cyclin H interaction. p17 binds to CDK-cyclin except for CDK1-cyclin B1 and CDK7-cyclin H complexes. We have determined that the negatively charged 151LAVXDVDA(E/D)DGADPN165 motif in cyclin B1 interacts with a positively charged region of CDK1. p17 mimics the cyclin B1 sequence to compete for CDK1 binding. The PSTAIRE motif is not required for interaction of CDK1-cyclin B1, but it is required for other CDK-cyclin complexes. p17 interacts with cyclins by its cyclin-binding motif, 125RXL127 Sequence and mutagenic analyses of p17 indicated that a 140WXFD143 motif and residues Asp-113 and Lys-122 in p17 are critical for CDK2 and CDK6 binding, leading to their sequestration in the cytoplasm. Exogenous expression of p17 significantly enhanced virus replication, whereas p17 mutants with low binding ability to cell cycle CDKs had no effect on virus yield, suggesting that p17 inhibits cell growth and the cell cycle, benefiting virus replication. An in vivo tumorigenesis assay also showed a significant reduction in tumor size.


Subject(s)
Cyclin H/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Orthoreovirus, Avian/physiology , Tumor Suppressor Protein p53/metabolism , Viral Proteins/metabolism , Animals , Cell Cycle , Chick Embryo , Chlorocebus aethiops , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin H/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/antagonists & inhibitors , Humans , Reoviridae Infections/virology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Vero Cells , Viral Proteins/genetics
3.
Cell Physiol Biochem ; 49(2): 432-446, 2018.
Article in English | MEDLINE | ID: mdl-30157476

ABSTRACT

BACKGROUND/AIMS: Next-generation sequencing (NGS) has revealed abundant long noncoding RNAs (lncRNAs) that have been characterized as critical components of cancer biology in humans. The present study aims to investigate the role of the lncRNA KCNQ1OT1 in breast cancer (BRCA) as well as the underlying molecular mechanisms and functions of KCNQ1OT1 involved in the progression of BRCA. METHODS: The Cancer Genome Atlas (TCGA) and StarBase v2.0 were used to obtain the required gene data. Dual luciferase reporter gene assays were conducted to verify the relevant intermolecular target relationships. QRT-PCR and Western blot were performed to measure the expression levels of different molecules. Cell proliferation was detected by using the MTT and colony formation assays, while cell migration and invasion were examined by transwell assay. Variations in cell apoptosis and cell cycle were determined through flow cytometry. A tumor xenograft model was applied to assess tumor growth in vivo. RESULTS: KCNQ1OT1 was found to be remarkably highly expressed in BRCA tissues and cells. KCNQ1OT1 modulated CCNE2 through sponging miR-145 in BRCA. KCNQ1OT1 promoted tumor growth in vivo by regulating miR-145/CCNE2. CONCLUSION: The KCNQ1OT1/miR-145/CCNE2 axis plays a critical regulatory role in BRCA, potentially giving rise to BRCA tumorigenesis and progression. These findings provide valuable evidence for improving the diagnosis and treatment of BRCA in the future.


Subject(s)
Cyclins/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cyclins/antagonists & inhibitors , Cyclins/genetics , Female , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , PPAR gamma/genetics , PPAR gamma/metabolism , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , RNA Interference , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use
4.
J Cell Mol Med ; 21(8): 1619-1635, 2017 08.
Article in English | MEDLINE | ID: mdl-28244683

ABSTRACT

Although translational research into autosomal dominant polycystic kidney disease (ADPKD) and its pathogenesis has made considerable progress, there is presently lack of standardized animal model for preclinical trials. In this study, we developed an orthologous mouse model of human ADPKD by cross-mating Pkd2 conditional-knockout mice (Pkd2f3 ) to Cre transgenic mice in which Cre is driven by a spectrum of kidney-related promoters. By systematically characterizing the mouse model, we found that Pkd2f3/f3 mice with a Cre transgene driven by the mouse villin-1 promoter (Vil-Cre;Pkd2f3/f3 ) develop overt cysts in the kidney, liver and pancreas and die of end-stage renal disease (ESRD) at 4-6 months of age. To determine whether these Vil-Cre;Pkd2f3/f3 mice were suitable for preclinical trials, we treated the mice with the high-dose mammalian target of rapamycin (mTOR) inhibitor rapamycin. High-dose rapamycin significantly increased the lifespan, lowered the cystic index and kidney/body weight ratio and improved renal function in Vil-Cre;Pkd2f3/f3 mice in a time- and dose-dependent manner. In addition, we further found that rapamycin arrested aberrant epithelial-cell proliferation in the ADPKD kidney by down-regulating the cell-cycle-associated cyclin-dependent kinase 1 (CDK1) and cyclins, namely cyclin A, cyclin B, cyclin D1 and cyclin E, demonstrating a direct link between mTOR signalling changes and the polycystin-2 dysfunction in cystogenesis. Our newly developed ADPKD model provides a practical platform for translating in vivo preclinical results into ADPKD therapies. The newly defined molecular mechanism by which rapamycin suppresses proliferation via inhibiting abnormally elevated CDK1 and cyclins offers clues to new molecular targets for ADPKD treatment.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , CDC2 Protein Kinase/antagonists & inhibitors , Cell Cycle/drug effects , Cyclins/antagonists & inhibitors , Polycystic Kidney, Autosomal Dominant/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cyclins/genetics , Cyclins/metabolism , Dose-Response Relationship, Drug , Female , Founder Effect , Gene Expression Regulation , Humans , Integrases/genetics , Integrases/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Promoter Regions, Genetic , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , TRPP Cation Channels/antagonists & inhibitors , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
5.
J Biol Chem ; 291(20): 10716-25, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26969161

ABSTRACT

The oncogenic phosphatase of regenerating liver 2 (PRL-2) has been shown to regulate intracellular magnesium levels by forming a complex through an extended amino acid loop present in the Bateman module of the CNNM3 magnesium transporter. Here we identified highly conserved residues located on this amino acid loop critical for the binding with PRL-2. A single point mutation (D426A) of one of those critical amino acids was found to completely disrupt PRL-2·human Cyclin M 3 (CNNM3) complex formation. Whole-cell voltage clamping revealed that expression of CNNM3 influenced the surface current, whereas overexpression of the binding mutant had no effect, indicating that the binding of PRL-2 to CNNM3 is important for the activity of the complex. Interestingly, overexpression of the CNNM3 D426A-binding mutant in cancer cells decreased their ability to proliferate under magnesium-deprived situations and under anchorage-independent growth conditions, demonstrating a PRL-2·CNNM3 complex-dependent oncogenic advantage in a more stringent environment. We further confirmed the importance of this complex in vivo using an orthotopic xenograft breast cancer model. Finally, because molecular modeling showed that the Asp-426 side chain in CNNM3 buries into the catalytic cavity of PRL-2, we showed that a PRL inhibitor could abrogate complex formation, resulting in a decrease in proliferation of human breast cancer cells. In summary, we provide evidence that this fundamental regulatory aspect of PRL-2 in cancer cells could potentially lead to broadly applicable and innovative therapeutic avenues.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cyclins/antagonists & inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Conserved Sequence , Cyclins/chemistry , Cyclins/genetics , Female , Humans , Mice , Mice, Nude , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Point Mutation , Protein Interaction Domains and Motifs/drug effects , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Pyridones/pharmacology , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
6.
BMC Cancer ; 17(1): 138, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28202053

ABSTRACT

BACKGROUND: Cisplatin (DDP)-based chemotherapy is the common first-line therapy for lung cancer. However, their efficacy is often limited by primary drug resistance and/or acquired drug resistance. The aim of this study was to investigate the function of miRNA-146a (miR-146a) in DDP-resistant non-small cell lung cancer (NSCLC), as well as the underlying mechanisms. METHODS: The effect of overexpression of miR-146a and/or knockdown of cyclin J (CCNJ) in A549/DDP and SPC-A1/DDP cells were investigated as follows. The cellular sensitivity to DDP, cell apoptosis, cell cycle and cell mobility were detected by CCK-8, flow cytometry, hoechst staining and cell invasion/migration assay, respectively. The effects of miR-146a overexpression in NSCLC resistant cells were further analyzed in a nude mouse xenograft model. RESULTS: Overexpression of miR-146a and/or knockdown of CCNJ significantly increased the sensitivity to DDP in A549/DDP and SPC-A1/DDP cells compared to NC group via arresting cell cycle, enhancing cell apoptosis, inhibiting cell viability and motility in vitro and in vivo. Furthermore, miR-146a could specially degrade the mRNA of CCNJ, as examined by dual luciferase report assay. CONCLUSION: The study indicates a crucial role of miR-146a in the development of acquired drug resistance to DDP in NSCLC cells. Further understanding of miR-146a mediated crosstalk networks may promote the clinical use of miR-146a analogue in NSCLC therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/pharmacology , Cyclins/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , MicroRNAs/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclins/genetics , Cyclins/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 286(1): 147-59, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21044963

ABSTRACT

Here, we report the identification of the RNA binding motif protein RBM15B/OTT3 as a new CDK11(p110) binding partner that alters the effects of CDK11 on splicing. RBM15B was initially identified as a binding partner of the Epstein-Barr virus mRNA export factor and, more recently, as a cofactor of the nuclear export receptor NXF1. In this study, we found that RBM15B co-elutes with CDK11(p110), cyclin L2α, and serine-arginine (SR) proteins, including SF2/ASF, in a large nuclear complex of ∼1-MDa molecular mass following size exclusion chromatography. Using co-immunoprecipitation experiments and in vitro pulldown assays, we mapped two distinct domains of RBM15B that are essential for its direct interaction with the N-terminal extension of CDK11(p110), cyclin L2α, and SR proteins such as 9G8 and SF2/ASF. Finally, we established that RBM15B is a functional competitor of the SR proteins SF2/ASF and 9G8, inhibits formation of the functional spliceosomal E complex, and antagonizes the positive effect of the CDK11(p110)-cyclin L2α complex on splicing both in vitro and in vivo.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/antagonists & inhibitors , Cyclins/metabolism , Nuclear Proteins/antagonists & inhibitors , RNA Splicing , RNA-Binding Proteins/metabolism , Animals , Binding, Competitive , Cell Nucleus/metabolism , HEK293 Cells , Humans , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/genetics , RNA-Binding Proteins/antagonists & inhibitors , Serine-Arginine Splicing Factors , Spliceosomes/metabolism
8.
Microbiology (Reading) ; 158(Pt 9): 2262-2271, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22723289

ABSTRACT

The poultry disease coccidiosis, caused by infection with Eimeria spp. apicomplexan parasites, is responsible for enormous economic losses to the global poultry industry. The rapid increase of resistance to therapeutic agents, as well as the expense of vaccination with live attenuated vaccines, requires the development of new effective treatments for coccidiosis. Because of their key regulatory function in the eukaryotic cell cycle, cyclin-dependent kinases (CDKs) are prominent drug targets. The Eimeria tenella CDC2-related kinase 2 (EtCRK2) is a validated drug target that can be activated in vitro by the CDK activator XlRINGO (Xenopus laevis rapid inducer of G2/M progression in oocytes). Bioinformatics analyses revealed four putative E. tenella cyclins (EtCYCs) that are closely related to cyclins found in the human apicomplexan parasite Plasmodium falciparum. EtCYC3a was cloned, expressed in Escherichia coli and purified in a complex with EtCRK2. Using the non-radioactive time-resolved fluorescence energy transfer (TR-FRET) assay, we demonstrated the ability of EtCYC3a to activate EtCRK2 as shown previously for XlRINGO. The EtCRK2/EtCYC3a complex was used for a combined in vitro and in silico high-throughput screening approach, which resulted in three lead structures, a naphthoquinone, an 8-hydroxyquinoline and a 2-pyrimidinyl-aminopiperidine-propane-2-ol. This constitutes a promising starting point for the subsequent lead optimization phase and the development of novel anticoccidial drugs.


Subject(s)
Antiprotozoal Agents/isolation & purification , CDC2 Protein Kinase/antagonists & inhibitors , Cyclins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Eimeria tenella/enzymology , High-Throughput Screening Assays/methods , Plasmodium falciparum/enzymology , Animals , CDC2 Protein Kinase/metabolism , Computational Biology/methods , Cyclins/metabolism , Enzyme Inhibitors/isolation & purification , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism
9.
PLoS Pathog ; 6(6): e1000967, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585629

ABSTRACT

Global gene expression data combined with bioinformatic analysis provides strong evidence that mammalian miRNAs mediate repression of gene expression primarily through binding sites within the 3' untranslated region (UTR). Using RNA induced silencing complex immunoprecipitation (RISC-IP) techniques we have identified multiple cellular targets for a human cytomegalovirus (HCMV) miRNA, miR-US25-1. Strikingly, this miRNA binds target sites primarily within 5'UTRs, mediating significant reduction in gene expression. Intriguingly, many of the genes targeted by miR-US25-1 are associated with cell cycle control, including cyclin E2, BRCC3, EID1, MAPRE2, and CD147, suggesting that miR-US25-1 is targeting genes within a related pathway. Deletion of miR-US25-1 from HCMV results in over expression of cyclin E2 in the context of viral infection. Our studies demonstrate that a viral miRNA mediates translational repression of multiple cellular genes by targeting mRNA 5'UTRs.


Subject(s)
5' Untranslated Regions/genetics , Cell Cycle Proteins/genetics , Gene Expression Regulation , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , Virus Replication/genetics , Blotting, Western , Cells, Cultured , Cyclins/antagonists & inhibitors , Cyclins/genetics , Cyclins/metabolism , Cytomegalovirus/physiology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Down-Regulation , Humans , Kidney/cytology , Kidney/metabolism , Luciferases/metabolism , RNA-Induced Silencing Complex/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Gynecol Oncol ; 125(2): 433-40, 2012 May.
Article in English | MEDLINE | ID: mdl-22252097

ABSTRACT

OBJECTIVE: These studies were designed to determine whether minocycline inhibits ovarian cancer growth in vitro and in vivo and the molecular mechanisms involved. MATERIALS AND METHODS: The effect of minocycline on ovarian cancer cell proliferation, cell cycle progression and apoptosis was assessed using human ovarian cancer cell lines OVCAR-3, SKOV-3 and A2780. Then, the capacity of minocycline to inhibit growth of OVCAR-3 xenografts in female nude mice was examined. RESULTS: Minocycline inhibited cell proliferation and colony formation, down-regulated cyclins A, B and E leading to arrest of cells in the G(0) phase of the cycle and suppression of DNA synthesis. Furthermore, exposure of these cells to minocycline led to DNA laddering, activation of caspase-3 and cleavage of PARP-1. In nude mice bearing sub-cutaneous tumors, minocycline suppressed tumor proliferation index, angiogenesis and tumor growth. CONCLUSION: These findings provide the initial basis for further evaluation of minocycline in the treatment of ovarian cancer.


Subject(s)
Minocycline/pharmacology , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclins/antagonists & inhibitors , DNA, Neoplasm/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Survival Rate , Xenograft Model Antitumor Assays
11.
BMC Complement Altern Med ; 12: 208, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23122182

ABSTRACT

BACKGROUND: The anticancer and immunomodulatory activity of mung bean sprouts (MBS) and the underlying mechanisms against human cervical and hepatocarcinoma cancer cells were explored. METHODS: MBS cytotoxicity and MBS-induced anticancer cytokines, TNF-α and IFN-ß from cancer cells, and immunological cytokines, IL-4, IFN-γ, and IL-10 from peripheral mononuclear cells (PMNC) were assessed by MTS and ELISA assays. Apoptotic cells were investigated by flow cytometry. The expression level of apoptotic genes (Bax, BCL-2, Capsases 7-9) and cell cycle regulatory genes (cyclin D, E, and A) and tumor suppressor proteins (p27, p21, and p53) was assessed by real-time qPCR in the cancer cells treated with extract IC50. RESULTS: The cytotoxicity on normal human cells was significantly different from HeLa and HepG2 cells, 163.97 ± 5.73, 13.3 ± 0.89, and 14.04 ± 1.5 mg/ml, respectively. The selectivity index (SI) was 12.44 ± 0.83 for HeLa and 11.94 ± 1.2 for HepG2 cells. Increased levels of TNF-α and IFN-ß were observed in the treated HeLa and HepG2 culture supernatants when compared with untreated cells. MBS extract was shown to be an immunopolarizing agent by inducing IFNγ and inhibiting IL-4 production by PBMC; this leads to triggering of CMI and cellular cytotoxicity. The extract induced apoptosis, in a dose and time dependent manner, in treated HeLa and HepG2, but not in untreated, cells (P < 0.05). The treatment significantly induced cell cycle arrest in G0/G1 in HeLa cells. The percentage of cells in G0/G1 phase of the treated HeLa cells increased from 62.87 ± 2.1%, in untreated cells, to 80.48 ± 2.97%. Interestingly, MBS IC50 induced the expression of apoptosis and tumor suppressor related genes in both HeLa and HepG2 cells. MBS extract succeeded in inducing cdk-inhibitors, p21, p53, and p27 in HeLa cells while it induced only p53 in HepG2 cells (P < 0.05). This is a clue for the cell type- specific interaction of the studied extract. These proteins inhibit the cyclin-cdk complexes apart from the presence of some other components that might stimulate some cyclins such as cyclin E, A, and D. CONCLUSION: MBS extract was shown to be a potent anticancer agent granting new prospects of anticancer therapy using natural products.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Fabaceae , Immunologic Factors/therapeutic use , Liver Neoplasms/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/antagonists & inhibitors , Cytokines/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression/drug effects , HeLa Cells , Hep G2 Cells , Humans , Immunity, Cellular/drug effects , Immunologic Factors/pharmacology , Inhibitory Concentration 50 , Leukocytes, Mononuclear , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Plant Extracts/pharmacology , Seeds , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
12.
Tsitol Genet ; 46(2): 3-11, 2012.
Article in Russian | MEDLINE | ID: mdl-22679818

ABSTRACT

In order to investigate the role of various serine/ threonine protein kinases and protein phosphatases in the regulation of mitosis progression in plant cells the influence of cyclin-dependent (olomoucine) and Ca2+ -calmodulin-dependent (W7) protein kinases inhibitors, as well as protein kinase C inhibitors (H7 and staurosporine) and protein phosphatases inhibitor (okadaic acid) on mitosis progression in synchronized tobacco BY-2 cells has been studied. It was found that BY-2 culture treatment with inhibitors of cyclin dependent protein kinases and protein kinase C causes prophase delay, reduces the mitotic index and displaces of mitotic peak as compare with control cells. Inhibition of Ca2+ -calmodulin dependent protein kinases enhances the cell entry into prophase and delays their exit from mitosis. Meanwhile inhibition of serine/threonine protein phosphatases insignificantly enhances of synchronized BY-2 cells entering into all phases of mitosis.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitosis/drug effects , Nicotiana/drug effects , Plant Cells/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/antagonists & inhibitors , Cyclins/metabolism , Kinetin/pharmacology , Mitosis/physiology , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Plant Cells/enzymology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Staurosporine/pharmacology , Sulfonamides/pharmacology , Nicotiana/cytology , Nicotiana/enzymology
13.
Eur J Med Chem ; 228: 114012, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34864331

ABSTRACT

Cyclin-dependent kinase 12 (CDK12) plays a crucial role in DNA-damage response gene transcription and has recently been validated as a promising target in cancer therapy. However, existing CDK12 inhibitors potently inhibit its closest isoform CDK13, which could cause potential toxicity. Therefore, the development of CDK12 inhibitors with isoform-selectivity against CDK13 continues to be a challenge. By taking advantage of the emerging PROteolysis-TArgeting Chimeras (PROTACs) approach, we have synthesized a potent PROTAC degrader PP-C8 based on the noncovalent dual inhibitors of CDK12/13 and demonstrated its specificity for CDK12 over CDK13. Notably, PP-C8 induces profound degradation of cyclin K simultaneously and downregulates the mRNA level of DNA-damage response genes. Global proteomics profiling revealed PP-C8 is highly selective toward CDK12-cyclin K complex. Importantly, PP-C8 demonstrates profound synergistic antiproliferative effects with PARP inhibitor in triple-negative breast cancer (TNBC). The potent and selective CDK12 PROTAC degrader developed in this study could potentially be used to treat CDK12-dependent cancers as combination therapy.


Subject(s)
CDC2 Protein Kinase/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase Inhibitors/pharmacology , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proteolysis/drug effects , Structure-Activity Relationship
14.
Prostate ; 71(1): 32-41, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20607765

ABSTRACT

BACKGROUND: Previous studies demonstrated that ST1959, a triazole derivative endowed with immunomodulatory activities, also exerts inhibitory effects on proliferation and survival of a panel of tumor cells. In this study, we sought to ascertain the effects of ST1959 on the growth of androgen-dependent and androgen-independent prostate cancer (PCa) cells. METHODS: The growth of androgen-dependent (LNCaP) and androgen-independent (PC3, DU-145) cells was analyzed in vitro both in the presence and absence of ST1959. Modulation of cyclin and androgen receptor (AR) expression following treatment with ST1959 was analyzed by Western blot and cytofluorimetric analysis. RESULTS: We observed that ST1959 causes a significant growth inhibition of LNCaP cells without affecting proliferation of androgen-insensitive DU-145 and PC3 cell lines. These effects were associated with G0/G1 cell cycle arrest and down-regulation of cyclin D1, A and B and AR expression. CONCLUSIONS: Our present findings indicate that the anti-proliferative activity of ST1959 on cell growth of androgen-dependent LNCaP PCa cells may be brought about by decreasing expression of functional AR and selected cyclins, ultimately leading to cell growth inhibition.


Subject(s)
Adenocarcinoma/metabolism , Cell Proliferation/drug effects , Cyclins/metabolism , Immunosuppressive Agents/pharmacology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Triazoles/pharmacology , Cell Line, Tumor , Cyclins/antagonists & inhibitors , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male
15.
J Pharmacol Exp Ther ; 339(2): 350-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21799049

ABSTRACT

Doxorubicin (DOX) is a drug commonly used for the treatment of cancer. The development of resistance to DOX is common, and high cumulative doses cause potentially lethal cardiac side effects. HO-3867 (3,5-bis(4-fluorobenzylidene)-1-[(2,2,5,5-tetramethyl-2,5-dihydro-1-hydroxy-pyrrol-3-yl)methyl]piperidin-4-one), a synthetic curcumin analog, has been shown to exhibit both anticancer and cardioprotective effects. However, its cardioprotection in the setting of a conventional cancer therapy has not been established. This work investigated the use of HO-3867 and DOX to achieve a complementary outcome, i.e., increased toxicity toward cancer cells, and reduced cardiac toxicity. Combination treatment was investigated using DOX-resistant MCF-7 breast cancer cells [MCF-7 multidrug-resistant (MDR)] and BALB/c mice. Lower doses of HO-3867 and DOX (5 and 2.5 µM, respectively) reduced viability of MCF-7 MDR cells to an extent significantly greater than that when either drug was used alone, an effect equivalent to that induced by exposure to 50 µM DOX. In normal cardiac cells, the loss of viability from combination treatment was significantly lower than that induced by 50 µM DOX. Increases in apoptotic markers, e.g., cleaved caspase-3, and decreases in fatty acid synthase and pAkt expressions were observed by Western blotting. Mice treated with both HO-3867 and DOX showed significant improvement in cardiac functional parameters compared with mice treated with DOX alone. Reduced expression of Bcl-2 and pAkt was observed in mice treated with DOX alone, whereas mice given combination treatment showed levels similar to control. The study indicates that combination treatment of HO-3867 and DOX is a viable option for treatment of cancer with reduced cardiotoxic side effects.


Subject(s)
Antineoplastic Agents/toxicity , Cardiotonic Agents/pharmacology , Doxorubicin/toxicity , Piperidones/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antioxidants/administration & dosage , Antioxidants/pharmacology , Antioxidants/therapeutic use , Aorta/cytology , Aorta/drug effects , Breast Neoplasms/drug therapy , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/therapeutic use , Caspases/biosynthesis , Cell Line, Tumor , Cyclins/antagonists & inhibitors , Cyclins/biosynthesis , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Echocardiography , Female , Heart/drug effects , Heart Diseases/drug therapy , Humans , Male , Mice , Mice, Inbred BALB C , Piperidones/administration & dosage , Piperidones/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/biosynthesis , fas Receptor/antagonists & inhibitors , fas Receptor/biosynthesis
16.
Nat Med ; 1(4): 309-20, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7585060

ABSTRACT

Conceptual and practical advances in molecular medicine are changing our understanding of cancer pathogenesis. In time this should provide the opportunity to alter the natural history of many cancers.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/antagonists & inhibitors , Neoplasms/etiology , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cyclin-Dependent Kinases/physiology , Cyclins/physiology , Humans , Molecular Biology , Neoplasms/genetics , Neoplasms/physiopathology , Neoplasms/therapy , Tumor Suppressor Protein p53/physiology
17.
Eur J Med Chem ; 214: 113232, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33550184

ABSTRACT

Over 50 tetrahydroindazoles were synthesized after 7-bromo-3,6,6-trimethyl-1-(pyridin-2-yl)-5,6,7,7a-tetrahydro-1H-indazol-4(3aH)-one (3) was identified as a hit compound in a high throughput screen for inhibition of CDK2 in complex with cyclin A. The activity of the most promising analogues was evaluated by inhibition of CDK2 enzyme complexes with various cyclins. Analogues 53 and 59 showed 3-fold better binding affinity for CDK2 and 2- to 10-fold improved inhibitory activity against CDK2/cyclin A1, E, and O compared to screening hit 3. The data from the enzyme and binding assays indicate that the binding of the analogues to a CDK2/cyclin complex is favored over binding to free CDK2. Computational analysis was used to predict a potential binding site at the CDK2/cyclin E1 interface.


Subject(s)
Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Binding Sites/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/metabolism , Cyclins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Indazoles/chemical synthesis , Indazoles/chemistry , MCF-7 Cells , Molecular Structure , Structure-Activity Relationship
18.
Cell Cycle ; 20(8): 808-818, 2021 04.
Article in English | MEDLINE | ID: mdl-33794722

ABSTRACT

The cyclin D-CDK4/6 complex has two distinct functions. Its kinase-dependent role involves its ability to act as serine/threonine kinase, responsible for phosphorylation of substrates required for cell cycle transitions, while its kinase-independent function involves its ability to act as a reservoir for p27Kip1. This association sequesters p27 from cyclin E-CDK2 complexes, allowing them to remain active. The aim of this current study is two-fold: to understand the contribution of the kinase-dependent and kinase-independent functions of CDK4 and CDK6 in epithelial cells and to directly compare CDK4 and CDK6 in a simple model system, TGF-ß treatment, where arrest is initiated by the expression of p15Ink4b. Cells that overexpressed a catalytically inactive, p15-insensitive CDK6 variant (p27 sequestration only mutant) were able to overcome TGF-ß-mediated arrest by maintaining CDK2 activity, while cells expressing the identical mutations in CDK4 were not. This result can be partially explained by the presence of a previously unidentified cyclin D-CDK6 dimer, which serves as a sink for free p27 during TGF-ß treatment, enabling CDK2 to remain inhibitor free. The use of the TGF-ß model system and the characterization of CDK pool dynamics and p27 switching is relevant to the CDK4/6 specific inhibitors, such as palbociclib, whose mechanism of action may resemble that of p15.


Subject(s)
Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 6/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Multimerization/physiology , Transforming Growth Factor beta/toxicity , Cell Cycle Checkpoints/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/antagonists & inhibitors , Cyclins/metabolism , Humans , Protein Multimerization/drug effects
19.
Chem Biol Drug Des ; 98(3): 323-340, 2021 09.
Article in English | MEDLINE | ID: mdl-33905613

ABSTRACT

The melatonin hormone secreted by the pineal gland is involved in physiological functions such as growth and maturation, circadian cycles, and biological activities including antioxidants, anti-tumor, and anti-ischemia. Melatonin not only interacts with proteins but also has functional effects on regulatory RNAs such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs). In this study, we overview various physiological and pathological conditions affecting melatonin through lncRNA and miRNA. The information compiled herein will serve as a solid foundation to formulate ideas for future mechanistic studies on melatonin. It will also provide a chance to more clarify the emerging functions of the non-coding transcriptome.


Subject(s)
Apoptosis/drug effects , Melatonin/pharmacology , RNA, Long Noncoding/metabolism , Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/antagonists & inhibitors , Cyclins/genetics , Cyclins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
20.
Biomed Pharmacother ; 139: 111545, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33873145

ABSTRACT

Tacrolimus (TAC, FK506) is a major calcineurin inhibitor and has been commonly used in treatments of patients with organ transplants and immune diseases. Moreover, tacrolimus is recommended by the treatment guidelines for oral potentially malignant disorders (OPMDs) such as oral lichen planus (OLP). However, whether tacrolimus increases the risk of cancer remains controversial. We observed that in a 4-Nitroquinoline N-oxide (4NQO)-induced oral carcinogenesis model, tacrolimus treatment was associated with a significantly lower ratio of cancer formation (52.94% vs. 90%) and a lower proportion of Ki67 and proliferation cell nuclear antigen (PCNA) -positive cells in lesion areas (P < 0.001). Liver, kidney, and lung functions of rats and the tumor immune microenvironment of the tongue were not affected. These observations suggest that tacrolimus blocked oral carcinogenesis through epithelial cell proliferation inhibition, independent of its immunosuppressive effects. As a processing factor, tacrolimus decreased tumor formation and cell proliferation in different stages of oral squamous cell carcinoma (OSCC) progression in vivo and in vitro. Furthermore, we investigated effects on the cell cycle and expression of related proteins. Tacrolimus induced G1/S phase arrest and significantly downregulated the expression of cyclinD1, cyclinE1, and c-Myc. These results suggest that tacrolimus induces G1/S phase arrest via inhibition of cyclinD1, cyclinE1, and c-Myc expression and retards oral cell carcinogenesis in vitro and in vivo. Thus, application of tacrolimus is a safe therapeutic strategy for treating OPMDs.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Cycle/drug effects , Mouth Neoplasms/prevention & control , Tacrolimus/pharmacology , 4-Nitroquinoline-1-oxide , Animals , Carcinogens , Cellular Microenvironment/drug effects , Cyclins/antagonists & inhibitors , Cyclins/biosynthesis , Genes, myc/drug effects , Ki-67 Antigen , Male , Mice , Mice, Inbred BALB C , Mouth Neoplasms/chemically induced , Mouth Neoplasms/pathology , Proliferating Cell Nuclear Antigen/analysis , Rats , Rats, Sprague-Dawley , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/prevention & control , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL