Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Sci Rep ; 10(1): 12879, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32733084

ABSTRACT

The cyclin M (CNNM) family of Mg2+ transporters is reported to promote tumour progression by binding to phosphatase of regenerating liver (PRL) proteins. Here, we established an assay for detection of the binding between the cystathionine-beta-synthase (CBS) domain of human CNNM3 (a region responsible for PRL binding) and human PRL2 using fluorescence resonance energy transfer (FRET) techniques. By fusing YPet to the C-terminus of the CNNM3 CBS domain and CyPet to the N-terminus of PRL2, we performed a FRET-based binding assay with purified proteins in multiwell plates and successfully detected the changes in fluorescence intensity derived from FRET with a reasonable Kd. We then confirmed that the addition of non-YPet-tagged CNNM3 and non-CyPet-tagged PRL proteins inhibited the changes in FRET intensity, whereas non-YPet-tagged CNNM3 with a mutation at the PRL2-binding site did not exhibit such inhibition. Furthermore, newly synthesized peptides derived from the CNNM loop region, with the PRL-binding sequences of the CNNM3 CBS domain, inhibited the interactions between CNNM3 and PRL2. Overall, these results showed that this method can be used for screening to identify inhibitors of CNNM-PRL interactions, potentially for novel anticancer therapy.


Subject(s)
Cyclins , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer , Peptides/chemistry , Protein Tyrosine Phosphatases , Cyclins/antagonists & inhibitors , Cyclins/chemistry , Cyclins/genetics , Humans , Protein Domains , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics
7.
J Exp Clin Cancer Res ; 38(1): 376, 2019 Aug 27.
Article in English | MEDLINE | ID: mdl-31455378

ABSTRACT

BACKGROUND: Metformin has been reported to function as the anti-tumor inhibiting the growth of different types of cancers, including bladder cancer. But there are few reports on the roles of Yap1, the key molecule of Hippo pathway, in the metformin induced inhibition of bladder cancer (BLCA). We are wondering if the inhibitory effect of metformin on bladder cancer is fulfilled via Yap1 and exploring the related mechanism. METHODS: MTS and colony formation assays were used to explore the cellular viabilities and proliferation of BLCA cells challenged by metformin at different concentrations, in vitro. Flow Cytometry (FCM) was used to analyze the cell cycle and the cellular apoptosis of the BLCA cells. Western Blot was performed to detect the expressions of AMPKα, Yap1, CCND1, CCNE1/2 and CDK2/4/6 in the metformin-treated BLCA cell lines. RNAi method was used for the related genetic functional analysis. The relationships among Yap1, TEADs and CCNE1/2 were predicted and evaluated using bioinformatics, dual-luciferase reporter and co-immunoprecipitation (Co-IP) assays. For in vivo experiments, a xenograft model was used to investigate the effects of metformin on the proliferation of BLCA cells. And Immunohistochemistry (IHC) assay was performed to assess the expressions of CCNE1/2 and Yap1 proteins in the tumor tissues from the model. RESULTS: Metformin could inhibit the proliferation of the BLCA cells via inducing the G1 cell cycle arrest without apoptosis. And metformin upregulated the phosphorylated AMPKα and decreased the expressions of Yap1 and CCND1, CCNE1/2 and CDK4/6. AMPK inhibition by compound C (CC) restored the cell proliferation and the G1 cell cycle arrest induced by metformin, in vivo. Knockdown of YAP1 inhibited the proliferation of BLCA cells and caused the cell cycle arrest at G1 phase by decreasing the expressions of CCNE1/2 and other G1 phase related molecules, which has been restored by the Yap 5SA mutant. Bioinformatics analysis showed that trans-factor TEAD4 was highly expressed and positively associated with the expressions of CCNE1 and CCNE2 in BLCA and only TEAD4 was precipitated by Yap1 in the BLCA cells. Further studies demonstrated that Yap1 positively regulated both CCNE1 and CCNE2 expressions via forming complex with TEAD4. Furthermore, we observed that metformin inhibited the cell proliferation by decreasing the expressions of Yap1 and both CCNE1 and CCNE2 in xenograft model. CONCLUSIONS: The results of our study reveal a new potential regulatory pathway in which metformin inhibits cell proliferation via AMPKα/Yap1/TEAD4/CCNE1/2 axis in BLCA cells, providing new insights into novel molecular therapeutic targets for BLCA.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Cyclin E/antagonists & inhibitors , Cyclins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Metformin/pharmacology , Muscle Proteins/metabolism , Oncogene Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Urinary Bladder Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin E/biosynthesis , Cyclin E/genetics , Cyclin E/metabolism , Cyclins/biosynthesis , Cyclins/genetics , Cyclins/metabolism , DNA-Binding Proteins/genetics , Female , G1 Phase/drug effects , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Muscle Proteins/genetics , Oncogene Proteins/biosynthesis , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , TEA Domain Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , YAP-Signaling Proteins
8.
Stem Cell Reports ; 13(2): 274-290, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31327733

ABSTRACT

Bone marrow (BM) niche cells help to keep adult hematopoietic stem cells (HSCs) in a quiescent state via secreted factors and induction of cell-cycle inhibitors. Here, we demonstrate that the adapter protein CABLES1 is a key regulator of long-term hematopoietic homeostasis during stress and aging. Young mice lacking Cables1 displayed hyperproliferation of hematopoietic progenitor cells. This defect was cell intrinsic, since it was reproduced in BM transplantation assays using wild-type animals as recipients. Overexpression and short hairpin RNA-mediated depletion of CABLES1 protein resulted in p21Cip/waf up- and downregulation, respectively. Aged mice lacking Cables1 displayed abnormalities in peripheral blood cell counts accompanied by a significant reduction in HSC compartment, concomitant with an increased mobilization of progenitor cells. In addition, Cables1-/- mice displayed increased sensitivity to the chemotherapeutic agent 5-fluorouracil due to an abnormal microenvironment. Altogether, our findings uncover a key role for CABLES1 in HSC homeostasis and stress hematopoiesis.


Subject(s)
Cyclins/genetics , Hematopoietic Stem Cells/cytology , Aging , Animals , Antigens, CD34/metabolism , Bone Marrow Cells/cytology , Cell Cycle Checkpoints/drug effects , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/antagonists & inhibitors , Cyclins/metabolism , Fluorouracil/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , RNA, Small Interfering/metabolism
9.
Nefrologia (Engl Ed) ; 39(5): 473-481, 2019.
Article in English, Spanish | MEDLINE | ID: mdl-30929891

ABSTRACT

Chronic kidney disease (CKD), cancer and haematological diseases share areas of reciprocal influence. Cancer can affect the kidney either as glomerular lesions or as a result of the toxic effects of medication or radiation with acute (thrombotic microangiopathy, acute kidney injury, interstitial nephropathies among others) or chronic processes (worsening of CKD after nephrectomy due to renal cancer, interstitial fibrosis, hydroelectrolytic disorders). On the other hand, patients who require renal replacement therapy with dialysis and particularly with kidney transplantation are at high risk of onset of cancer due to the immunosuppression situation that they generate. In addition to conventional chemotherapy, innovative treatments have been developed: target agents against growth factors and their receptor; anti-angiogenic drugs; immunoregulatory proteins; cell cycle regulators; and enzyme inhibitors. Other immunotherapeutic approaches have also been developed, such as vaccines, adoptive cell therapy (CAR T cells) or development of antibodies. All these therapeutic advances will improve the outcomes against cancer and haematological diseases, but they are not free from secondary renal problems. Onco-Nephrology is already an important area for the Spanish Society of Nephrology with a large number of inter-consultations. Nephrologists need a better understanding of rapidly evolving areas of cancer biology and its treatment in order to become valued members of the cancer care team and to provide the best nephrology care possible.


Subject(s)
Antineoplastic Agents/adverse effects , Kidney/drug effects , Neoplasms/drug therapy , Renal Insufficiency, Chronic/complications , Acrylonitrile/adverse effects , Acrylonitrile/analogs & derivatives , Acute Kidney Injury/chemically induced , Angiogenesis Inhibitors/adverse effects , Aniline Compounds/adverse effects , Biomarkers/blood , Contrast Media/adverse effects , Creatinine/blood , Cyclins/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Humans , Immunotherapy, Adoptive/methods , Kidney Neoplasms/complications , Kidney Neoplasms/surgery , Molecular Targeted Therapy/adverse effects , Neoplasms/complications , Nephrectomy/adverse effects , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Postoperative Complications/etiology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Referral and Consultation/statistics & numerical data , Renal Dialysis/adverse effects , TOR Serine-Threonine Kinases/antagonists & inhibitors
10.
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
11.
ChemMedChem ; 13(23): 2479-2483, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30328274

ABSTRACT

An in silico screen of 350 000 commercially available compounds was conducted with an unbiased approach to identify potential malaria inhibitors that bind to the Plasmodium falciparum protein kinase 5 (PfPK5) ATP-binding site. PfPK5 is a cyclin-dependent kinase-like protein with high sequence similarity to human cyclin-dependent kinase 2 (HsCDK2), but its precise role in cell-cycle regulation remains unclear. After two-dimensional fingerprinting of the top scoring compounds, 182 candidates were prioritized for biochemical testing based on their structural diversity. Evaluation of these compounds demonstrated that 135 bound to PfPK5 to a similar degree or better than known PfPK5 inhibitors, confirming that the library was enriched with PfPK5-binding compounds. A previously reported triazolodiamine HsCDK2 inhibitor and the screening hit 4-methylumbelliferone were each selected for an analogue study. The results of this study highlight the difficult balance between optimization of PfPK5 affinity and binding selectivity for PfPK5 over its closest human homologue HsCDK2. Our approach enabled the discovery of several new PfPK5-binding compounds from a modest screening campaign and revealed the first scaffold to have improved PfPK5/HsCDK2 selectivity. These steps are critical for the development of PfPK5-targeting probes for functional studies and antimalarials with decreased risks of host toxicity.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Cyclins/antagonists & inhibitors , Plasmodium falciparum/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protozoan Proteins/antagonists & inhibitors , Computer Simulation , Cyclins/metabolism , Drug Discovery , Hep G2 Cells , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Structure-Activity Relationship
12.
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
13.
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
14.
Nat Commun ; 9(1): 1876, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29760377

ABSTRACT

The assembly of prereplicative complex (pre-RC) during G1 phase must be tightly controlled to sustain cell proliferation and maintain genomic stability. Mechanisms to prevent pre-RC formation in G2/M and S phases are well appreciated, whereas how cells ensure efficient pre-RC assembly during G1 is less clear. Here we report that cyclin K regulates pre-RC formation. We find that cyclin K expression positively correlates with cell proliferation, and knockdown of cyclin K or its cognate kinase CDK12 prevents the assembly of pre-RC in G1 phase. Mechanistically we uncover that cyclin K promotes pre-RC assembly by restricting cyclin E1 activity in G1. We identify a cyclin K-dependent, novel phosphorylation site in cyclin E1 that disrupts its interaction with CDK2. Importantly, this antagonistic relationship is largely recapitulated in cyclin E1-overexpressing tumors. We discuss the implications of our findings in light of recent reports linking cyclin K and CDK12 to human tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , G1 Phase Cell Cycle Checkpoints/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/antagonists & inhibitors , Cyclins/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Genomic Instability , HCT116 Cells , Humans , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , S Phase Cell Cycle Checkpoints/genetics
15.
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
16.
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
17.
Sci Rep ; 6: 24661, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27381447

ABSTRACT

Hypoxia-induced pulmonary hypertension (PH), which is characterized by vascular remodeling of blood vessels, is a significant complication of chronic obstructive pulmonary disease (COPD). In this study, we screened 13 candidate miRNAs in pulmonary artery smooth muscle cells (PASMCs) harvested from COPD patients with PH (n = 18) and normal controls (n = 15) and found that the expression of miR-214 was differentially expressed between these two groups. Additionally, cyclin L2 (CCNL2) was validated as a target of miR-214 in PASMCs using a luciferase assay. Based on real-time PCR, immunohistochemistry and western blot, the expression of CCNL2 was substantially downregulated in PASMCs from COPD patients with PH compared with those from normal controls. Moreover, the relationship between miRNA and mRNA expression was confirmed using real-time PCR and western blot in PASMCs transfected with miR-214 mimics. Furthermore, the introduction of miR-214 significantly promoted the proliferation of PASMCs by suppressing cell apoptosis, and this effect was mediated by the downregulation of CCNL2. Exposure of PASMCs to hypoxia significantly increased the expression of miR-214, decreased the expression of CCNL2, and promoted cell proliferation. However, these effects were significantly attenuated by the introduction of miR-214 inhibitors, which significantly downregulated miR-214 expression and upregulated CCNL2 expression.


Subject(s)
Cell Hypoxia , Cyclins/metabolism , Hypertension, Pulmonary/diagnosis , MicroRNAs/metabolism , Pulmonary Disease, Chronic Obstructive/diagnosis , Transcription Factors/metabolism , Vascular Remodeling/physiology , Aged , Animals , Base Sequence , Case-Control Studies , Cells, Cultured , Cyclins/antagonists & inhibitors , Cyclins/genetics , Disease Models, Animal , Female , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Pulmonary Disease, Chronic Obstructive/complications , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Up-Regulation
18.
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
19.
Methods Mol Biol ; 1336: 29-45, 2016.
Article in English | MEDLINE | ID: mdl-26231706

ABSTRACT

The abundance of biochemical and structural knowledge on the Cyclin-Dependent Kinases (CDKs) has provided a comprehensive but not exhaustive insight into the molecular determinants that govern their function mechanisms. The implementation of structural and functional CDK models towards developing novel anticancer strategies that will specifically target individual or multiple CDKs remains a critical need.More than 250 CDKs crystal structures are available to-date, including truncated or whole, modified or not, active or inactive forms, co-crystallized with the cyclins and/or their respective putative inhibitors, though, to our knowledge, there is no NMR solved structure available to date. We hitherto attempt to provide a useful guide from protein production to crystallization for CDK/Inhibitors complexes based on an overview of the already elucidated CDK structures, constructs and the preferable expression vectors in each case, in order to yield the respective crystals.


Subject(s)
Crystallization/methods , Cyclin-Dependent Kinases/chemistry , Cyclins/antagonists & inhibitors , Animals , Baculoviridae , Cell Line , Cloning, Molecular , Crystallography, X-Ray , Cyclins/chemistry , Databases, Protein , Escherichia coli/metabolism , Humans , Insecta , Magnetic Resonance Spectroscopy , Molecular Biology/methods , Phosphorylation , Protein Conformation , Protein Structure, Tertiary
20.
Sci Rep ; 5: 16985, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26592552

ABSTRACT

Cancer cells acquire invasive ability to degrade and adhere to extracellular matrix (ECM) and migrate to adjacent tissues. This ultimately results metastasis. Hence, the present study investigated the in vitro effects of cyclopeptide glycoside, RA-XII on cell adhesion, invasion, proliferation and matrix degradation, and its underlying mechanism in murine breast tumour cells, 4T1. The effect of RA-XII on tumour growth and metastasis in 4T1-bearing mice was also investigated. Our results showed that RA-XII inhibited tumour cell adhesion to collagen, fibronectin and laminin, RA-XII also reduced the expressions of vascular cell adhesion molecule, intracellular adhesion molecule and integrins, and integrin binding. In addition, RA-XII significantly inhibited breast tumour cell migration via interfering cofilin signaling and chemokine receptors. The activities of matrix metalloproteinase-9 and urokinase-type of plasminogen activator, and the expressions of ECM-associated proteinases were attenuated significantly by RA-XII. Furthermore, RA-XII induced G1 phase arrest and inhibited the expressions of cyclins and cyclin-dependent kinases. RA-XII inhibited the expressions of molecules in PI3K/AKT, NF-kappaB, FAK/pSRC, MAPK and EGFR signaling. RA-XII was also shown to have anti-tumour, anti-angiogenic and anti-metastatic activities in metastatic breast tumour-bearing mice. These findings strongly suggested that RA-XII is a potential anti-metastatic agent for breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Extracellular Matrix Proteins/antagonists & inhibitors , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/drug therapy , Peptides, Cyclic/pharmacology , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cofilin 1/antagonists & inhibitors , Cofilin 1/genetics , Cofilin 1/metabolism , Cyclins/antagonists & inhibitors , Cyclins/genetics , Cyclins/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Integrins/antagonists & inhibitors , Integrins/genetics , Integrins/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lymphatic Metastasis , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Protein Binding , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Signal Transduction , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...