Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Endocrinology ; 161(10)2020 10 01.
Article in English | MEDLINE | ID: mdl-32877917

ABSTRACT

The protein product of the cyclin D1 oncogene functions by activating partner cyclin-dependent kinases (cdk)4 or cdk6 to phosphorylate, thereby inactivating, the retinoblastoma protein pRB. Nonclassical, cdk-independent, functions of cyclin D1 have been described but their role in cyclin D1-driven neoplasia, with attendant implications for recently approved cdk4/6 chemotherapeutic inhibitors, requires further examination. We investigated whether cyclin D1's role in parathyroid tumorigenesis in vivo is effected primarily through kinase-dependent or kinase-independent mechanisms. Using a mouse model of cyclin D1-driven parathyroid tumorigenesis (PTH-D1), we generated new transgenic lines harboring a mutant cyclin D1 (KE) that is unable to activate its partner kinases. While this kinase-dead KE mutant effectively drove mammary tumorigenesis in an analogous model, parathyroid-overexpressed cyclin D1 KE mice did not develop the characteristic biochemical hyperparathyroidism or parathyroid hypercellularity of PTH-D1 mice. These results strongly suggest that in parathyroid cells, cyclin D1 drives tumorigenesis predominantly through cdk-dependent mechanisms, in marked contrast with the cdk-independence of cyclin D1-driven mouse mammary cancer. These findings highlight crucial tissue-specific mechanistic differences in cyclin D1-driven tumorigenesis, suggest that parathyroid/endocrine cells may be more tumorigenically vulnerable to acquired genetic perturbations in cdk-mediated proliferative control than other tissues, and carry important considerations for therapeutic intervention.


Subject(s)
Cyclin D1/genetics , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , Parathyroid Neoplasms/genetics , Parathyroid Neoplasms/metabolism , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin D1/metabolism , Hyperparathyroidism/genetics , Hyperparathyroidism/metabolism , Hyperparathyroidism/pathology , Mice , Mice, Transgenic , Mutation , Parathyroid Neoplasms/pathology , Phosphorylation/genetics , Signal Transduction/genetics
2.
Aging (Albany NY) ; 12(8): 6904-6927, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32310823

ABSTRACT

Gastric cancer (GC) is one of the most commonly diagnosed malignancies in digestive tract and its underlying molecular mechanism is still not clear, so we aimed to reveal the relationship between GC and UDP-GlcNAc pyrophosphorylase-1 like 1 (UAP1L1). The detection of UAP1L1 expression in GC tumor and normal tissues was accomplished by immunohistochemistry and demonstrated the upregulation of UAP1L1 in GC, which was statistically associated with tumor grade. GC cell models constructed via transfection of UAP1L1-silencing/overexpressing lentiviruses were employed for evaluating the effects of UAP1L1 knockdown/overexpression on GC in vitro and in vivo. The results indicated that UAP1L1 played important role in development of GC through regulating cell proliferation, colony formation, cell apoptosis and cell migration. Subsequently, CDK6 was identified as a potential target in UAP1L1 induced regulation of GC, downregulation of which exhibited similar inhibition effects on GC with UAP1L1. Moreover, it was demonstrated that the promotion of GC by UAP1L1 overexpression could be significantly attenuated or even reversed by simultaneously silencing CDK6. In conclusion, UAP1L1 was reported to be a tumor promotor in the development and progression of GC which may exert its role through regulating CDK6 and may act as a candidate of therapeutic target in treatment.


Subject(s)
Cyclin-Dependent Kinase 6/physiology , Nucleotidyltransferases/physiology , Stomach Neoplasms/etiology , Adult , Aged , Animals , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Middle Aged , Nucleotidyltransferases/analysis , Nucleotidyltransferases/genetics , Stomach Neoplasms/chemistry , Up-Regulation
3.
Cell Rep ; 31(2): 107504, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294430

ABSTRACT

Cell growth is coupled to cell-cycle progression in mitotically proliferating mammalian cells, but the underlying molecular mechanisms are not well understood. CyclinD-Cdk4/6 is known to phosphorylate RB to promote S-phase entry, but recent work suggests they have additional functions. We show here that CyclinD-Cdk4/6 activates mTORC1 by binding and phosphorylating TSC2 on Ser1217 and Ser1452. Pharmacological inhibition of Cdk4/6 leads to a rapid, TSC2-dependent reduction of mTORC1 activity in multiple human and mouse cell lines, including breast cancer cells. By simultaneously driving mTORC1 and E2F, CyclinD-Cdk4/6 couples cell growth to cell-cycle progression. Consistent with this, we see that mTORC1 activity is cell cycle dependent in proliferating neural stem cells of the adult rodent brain. We find that Cdk4/6 inhibition reduces cell proliferation partly via TSC2 and mTORC1. This is of clinical relevance, because Cdk4/6 inhibitors are used for breast cancer therapy.


Subject(s)
Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Aminopyridines/pharmacology , Animals , Benzimidazoles/pharmacology , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cyclin D/metabolism , Cyclin D/physiology , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , Humans , Mechanistic Target of Rapamycin Complex 1/physiology , Mice , Phosphorylation , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Tuberous Sclerosis Complex 2 Protein/metabolism
4.
Hepatology ; 70(5): 1614-1630, 2019 11.
Article in English | MEDLINE | ID: mdl-31077409

ABSTRACT

Cholangiocarcinoma (CCA) is a bile duct cancer with a very poor prognosis. Currently, there is no effective pharmacological treatment available for it. We showed that CCA ubiquitously relies on cyclin-dependent kinases 4 and 6 (CDK4/6) activity to proliferate. Primary CCA tissues express high levels of cyclin D1 and the specific marker of CDK4/6 activity, phospho-RB Ser780. Treatment of a 15-CCA cell line collection by pharmacological CDK4/6 inhibitors leads to reduced numbers of cells in the S-phase and senescence in most of the CCA cell lines. We found that expression of retinoblastoma protein (pRB) is required for activity of the CDK4/6 inhibitor, and that loss of pRB conferred CDK4/6 inhibitor-drug resistance. We also identified that sensitivity of CCA to CDK4/6 inhibition is associated with the activated KRAS signature. Effectiveness of CDK4/6 inhibition for CCA was confirmed in the three-dimensional spheroid-, xenograft-, and patient-derived xenograft models. Last, we identified a list of genes whose expressions can be used to predict response to the CDK4/6 inhibitor. Conclusion: We investigated a ubiquitous dependency of CCA on CDK4/6 activity and the universal response to CDK4/6 inhibition. We propose that the CDK4/6-pRB pathway is a suitable therapeutic target for CCA treatment.


Subject(s)
Bile Duct Neoplasms/etiology , Cholangiocarcinoma/etiology , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , Animals , Humans , Mice , Tumor Cells, Cultured
5.
Acta Biochim Biophys Sin (Shanghai) ; 49(7): 635-642, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28510621

ABSTRACT

Mouse miR-290 cluster miRNAs are expressed specifically in early embryos and embryonic germ cells. These miRNAs play critical roles in the maintenance of pluripotency and self-renewal. Here, we showed that Cyclin D1 is a direct target gene of miR-290 cluster miRNAs. Negative relationships between the expression of Cyclin D1 protein and miR-290 cluster miRNAs in pluripotent and non-pluripotent cells, as well as in differentiating CGR8 cells were observed. Inhibition of miR-290 cluster miRNAs could arrest cells at the G1 phase and slow down the cell proliferation in CGR8 mouse stem cells. Since miR-290 cluster miRNAs are the most dominant stem-cell-specific miRNAs, our results revealed an important cause for the absence of Cyclin D1 in mouse embryonic stem cells.


Subject(s)
Cyclin D1/analysis , MicroRNAs/physiology , Mouse Embryonic Stem Cells/chemistry , Animals , Cell Proliferation , Cells, Cultured , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , G1 Phase , Mice , Mouse Embryonic Stem Cells/cytology
6.
J Hematol Oncol ; 10(1): 97, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28438180

ABSTRACT

Uncontrolled cell division is the hallmark of cancers. Full understanding of cell cycle regulation would contribute to promising cancer therapies. In particular, cyclin-dependent kinases 4/6 (CDK4/6), which are pivotal drivers of cell proliferation by combination with cyclin D, draw more and more attention. Subsequently, extensive studies were carried out to explore drugs inhibiting CDK4/6 and assess the efficacy and safety of these drugs in cancer, especially breast cancer. Due to the insuperable adverse events and the less activity observed in vivo, the drug development of the initial pan-CDK inhibitor flavopiridol was consequently discontinued, and then highly specific inhibitors were extensively researched and developed, including palbociclib (PD0332991), ribociclib (LEE011), and abemaciclib (LY2835219). Food and Drug Administration has approved palbociclib and ribociclib for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced or metastatic breast cancer, and recent clinical trial data suggest that palbociclib significantly improved clinical outcome when combined with letrozole or fulvestrant. Besides, the favorable effects of abemaciclib on prolonging survival of breast cancer patients have also been observed in clinical trials both for single-agent and combination strategy. In this review, we outline the preclinical and clinical advancement of these three orally bioavailable and highly selective CDK4/6 inhibitors in breast cancer.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Aminopyridines/adverse effects , Aminopyridines/pharmacokinetics , Aminopyridines/therapeutic use , Antineoplastic Agents, Immunological/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Benzimidazoles/therapeutic use , Cell Cycle , Clinical Trials as Topic , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , Drug Screening Assays, Antitumor , Female , Humans , Neoplasm Proteins/adverse effects , Neoplasm Proteins/physiology , Piperazines/adverse effects , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Purines/adverse effects , Purines/pharmacokinetics , Purines/therapeutic use , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Therapies, Investigational
7.
Haematologica ; 102(6): 995-1005, 2017 06.
Article in English | MEDLINE | ID: mdl-28255017

ABSTRACT

Mice lacking Cdk6 kinase activity suffer from mild anemia accompanied by elevated numbers of Ter119+ cells in the bone marrow. The animals show hardly any alterations in erythroid development, indicating that Cdk6 is not required for proliferation and maturation of erythroid cells. There is also no difference in stress erythropoiesis following hemolysis in vivo However, Cdk6-/- erythrocytes have a shortened lifespan and are more sensitive to mechanical stress in vitro, suggesting differences in cytoskeletal architecture. Erythroblasts contain both Cdk4 and Cdk6, while mature erythrocytes apparently lack Cdk4 and their Cdk6 is partly associated with the cytoskeleton. We used mass spectrometry to show that Cdk6 interacts with a number of proteins involved in cytoskeleton organization. Cdk6-/- erythroblasts show impaired F-actin formation and lower levels of gelsolin, which interacts with Cdk6. We also found that Cdk6 regulates the transcription of a panel of genes involved in actin (de-)polymerization. Cdk6-deficient cells are sensitive to drugs that interfere with the cytoskeleton, suggesting that our findings are relevant to the treatment of patients with anemia - and may be relevant to cancer patients treated with the new generation of CDK6 inhibitors.


Subject(s)
Cyclin-Dependent Kinase 6/physiology , Cytoskeleton/ultrastructure , Erythroid Cells/ultrastructure , Actin Cytoskeleton , Actins/metabolism , Anemia , Animals , Gelsolin/metabolism , Gene Expression Regulation , Mass Spectrometry , Mice , Mice, Inbred C57BL
8.
Nat Rev Cancer ; 17(2): 93-115, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28127048

ABSTRACT

Cancer is characterized by uncontrolled tumour cell proliferation resulting from aberrant activity of various cell cycle proteins. Therefore, cell cycle regulators are considered attractive targets in cancer therapy. Intriguingly, animal models demonstrate that some of these proteins are not essential for proliferation of non-transformed cells and development of most tissues. By contrast, many cancers are uniquely dependent on these proteins and hence are selectively sensitive to their inhibition. After decades of research on the physiological functions of cell cycle proteins and their relevance for cancer, this knowledge recently translated into the first approved cancer therapeutic targeting of a direct regulator of the cell cycle. In this Review, we focus on proteins that directly regulate cell cycle progression (such as cyclin-dependent kinases (CDKs)), as well as checkpoint kinases, Aurora kinases and Polo-like kinases (PLKs). We discuss the role of cell cycle proteins in cancer, the rationale for targeting them in cancer treatment and results of clinical trials, as well as the future therapeutic potential of various cell cycle inhibitors.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/physiology , Cell Cycle Proteins/physiology , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/physiology , Clinical Trials as Topic , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/physiology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/physiology , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/physiology , Signal Transduction , Polo-Like Kinase 1
9.
Bull Cancer ; 104(2): 114-122, 2017 Feb.
Article in French | MEDLINE | ID: mdl-28126188

ABSTRACT

Dysregulation of cellular cycle is a key component of carcinogenesis and its targeting represents an interesting approach. Recently, the development of selective inhibitors of the cycle targeting the cyclin-dependent kinases (CDK) 4 and 6 revived interest in this therapeutic class after the failure of pan-inhibitors. Palbociclib, ribociclib, and abemaciclib are the 3 drugs with the most advanced development. They demonstrated preclinical activity in luminal breast cancer models and are under clinical evaluation. The first available studies demonstrate the value of these compounds with an improved prognosis of metastatic patients in combination with endocrine therapy (palbociclib, ribociclib) or in monotherapy (abemaciclib). The results of ongoing studies will clarify the role of these agents in our new strategies and the individualisation of biomarkers will help to define patients who benefit most from this approach.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Neoplasms, Hormone-Dependent/drug therapy , Protein Kinase Inhibitors/therapeutic use , Aminopyridines/adverse effects , Aminopyridines/therapeutic use , Antineoplastic Agents/adverse effects , Benzimidazoles/adverse effects , Benzimidazoles/therapeutic use , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Cycle/physiology , Cell Cycle Checkpoints , Clinical Trials as Topic , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , Female , Humans , Neoplasms, Hormone-Dependent/chemistry , Piperazines/adverse effects , Piperazines/therapeutic use , Protein Kinase Inhibitors/adverse effects , Purines/adverse effects , Purines/therapeutic use , Pyridines/adverse effects , Pyridines/therapeutic use
10.
Int J Cancer ; 140(5): 1147-1158, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27668411

ABSTRACT

A network of autocrine and paracrine signals defines B cell homeostasis and is thought to be involved in transformation processes. Investigating interactions of these microenvironmental factors and their relation to proto-oncogenes as c-Myc (MYC) is fundamental to understand the biology of B cell lymphoma. Therefore, B cells with conditional MYC expression were stimulated with CD40L, insulin-like growth factor 1, α-IgM, Interleukin-10 (IL10) and CpG alone or in combination. The impact of forty different interventions on cell proliferation was investigated in MYC deprived cells and calculated by linear regression. Combination of CpG and IL10 led to a strong synergistic activation of cell proliferation (S-phase/doubling of total cell number) comparable to cells with high MYC expression. A synergistic up-regulation of CDK4, CDK6 and CCND3 expression by IL10 and CpG treatment was causal for this proliferative effect as shown by qRT-PCR analysis and inhibition of the CDK4/6 complex by PD0332991. Furthermore, treatment of stimulated MYC deprived cells with MLN120b, ACHP, Pyridone 6 or Ruxolitinib showed that IL10/CpG induced proliferation and CDK4 expression were JAK/STAT3 and IKK/NF-κB dependent. This was further supported by STAT3 and p65/RELA knockdown experiments, showing strongest effects on cell proliferation and CDK4 expression after double knockdown. Additionally, chromatin immunoprecipitation revealed a dual binding of STAT3 and p65 to the proximal promotor of CDK4 after IL10/CpG treatment. Therefore, the observed synergism of IL10R and TLR9 signalling was able to induce proliferation in a comparable way as aberrant MYC and might play a role in B cell homeostasis or transformation.


Subject(s)
B-Lymphocytes/drug effects , Interleukin-10/physiology , Toll-Like Receptor 9/physiology , B-Lymphocytes/cytology , Cell Division , Cell Line, Transformed , Cell Transformation, Neoplastic , Cells, Cultured , CpG Islands , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/physiology , Drug Synergism , Gene Expression Regulation , Humans , Interleukin-10/pharmacology , Lymphoma/etiology , Proto-Oncogene Proteins c-myc/physiology , S Phase/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 9/agonists , Transcription Factor RelA/metabolism
11.
Blood ; 127(23): 2890-902, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27099147

ABSTRACT

Up to 30% of patients with acute myeloid leukemia have constitutively activating internal tandem duplications (ITDs) of the FLT3 receptor tyrosine kinase. Such mutations are associated with a poor prognosis and a high propensity to relapse after remission. FLT3 inhibitors are being developed as targeted therapy for FLT3-ITD(+) acute myeloid leukemia; however, their use is complicated by rapid development of resistance, which illustrates the need for additional therapeutic targets. We show that the US Food and Drug Administration-approved CDK4/6 kinase inhibitor palbociclib induces apoptosis of FLT3-ITD leukemic cells. The effect is specific for FLT3-mutant cells and is ascribed to the transcriptional activity of CDK6: CDK6 but not its functional homolog CDK4 is found at the promoters of the FLT3 and PIM1 genes, another important leukemogenic driver. There CDK6 regulates transcription in a kinase-dependent manner. Of potential clinical relevance, combined treatment with palbociclib and FLT3 inhibitors results in synergistic cytotoxicity. Simultaneously targeting two critical signaling nodes in leukemogenesis could represent a therapeutic breakthrough, leading to complete remission and overcoming resistance to FLT3 inhibitors.


Subject(s)
Cyclin-Dependent Kinase 6/physiology , Leukemia, Myeloid, Acute/genetics , Piperazines/pharmacology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Pyridines/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Adult , Aged , Animals , Cells, Cultured , Cyclin-Dependent Kinase 6/metabolism , Female , Gene Duplication , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Protein Kinase Inhibitors/therapeutic use , Tandem Repeat Sequences , Transcriptional Activation/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/metabolism
12.
Mol Cell Biochem ; 412(1-2): 289-96, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26699910

ABSTRACT

MicroRNA-1 (miR-1) is approved involved in cardiac hypertrophy, but the underlying molecular mechanisms of miR-1 in cardiac hypertrophy are not well elucidated. The present study aimed to investigate the potential role of miR-1 in modulating CDKs-Rb pathway during cardiomyocyte hypertrophy. A rat model of hypertrophy was established with abdominal aortic constriction, and a cell model of hypertrophy was also achieved based on PE-promoted neonatal rat ventricular cardiomyocytes (NRVCs). We demonstrated that miR-1 expression was markedly decreased in hypertrophic myocardium and hypertrophic cardiomyocytes. Dual luciferase reporter assays revealed that miR-1 interacted with the 3'UTR of CDK6, and miR-1 was verified to inhibit CDK6 expression at the posttranscriptional level. CDK6 protein expression was observed increased in hypertrophic myocardium and hypertrophic cardiomyocytes. Morover, miR-1 mimic, in parallel to CDK6 siRNA, could inhibit PE-induced hypertrophy of NRVCs, with decreases in cell size, newly transcribed RNA, expressions of ANF and ß-MHC, and the phosphorylated pRb. Taken together, our results reveal that derepression of CDK6 and activation of Rb pathway contributes to the effect of attenuation of miR-1 on provoking cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Cyclin-Dependent Kinase 6/physiology , MicroRNAs/metabolism , Myocytes, Cardiac/pathology , Animals , Cyclin-Dependent Kinase 6/genetics , Down-Regulation , Male , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley
13.
Leukemia ; 30(5): 1033-43, 2016 05.
Article in English | MEDLINE | ID: mdl-26707936

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is a high-risk subset of acute leukemia, characterized by frequent activation of Notch1 or AKT signaling, where new therapeutic approaches are needed. We showed previously that cyclin-dependent kinase 6 (CDK6) is required for thymic lymphoblastic lymphoma induced by activated AKT. Here, we show CDK6 is required for initiation and maintenance of Notch-induced T-ALL. In a mouse retroviral model, hematopoietic stem/progenitor cells lacking CDK6 protein or expressing kinase-inactive (K43M) CDK6 are resistant to induction of T-ALL by activated Notch, whereas those expressing INK4-insensitive (R31C) CDK6 are permissive. Pharmacologic inhibition of CDK6 kinase induces CD25 and RUNX1 expression, cell cycle arrest and apoptosis in mouse and human T-ALL. Ablation of Cd25 in a K43M background restores Notch-induced T leukemogenesis, with disease that is resistant to CDK6 inhibitors in vivo. These data support a model whereby CDK6-mediated suppression of CD25 is required for initiation of T-ALL by activated Notch1, and CD25 induction mediates the therapeutic response to CDK6 inhibition in established T-ALL. These results both validate CDK6 as a molecular target for therapy of this subset of T-ALL and suggest that CD25 expression could serve as a biomarker for responsiveness of T-ALL to CDK4/6 inhibitor therapy.


Subject(s)
Cyclin-Dependent Kinase 6/physiology , Interleukin-2 Receptor alpha Subunit/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Receptor, Notch1/physiology , Animals , Apoptosis/drug effects , Carcinogenesis/metabolism , Cell Cycle Checkpoints/drug effects , Core Binding Factor Alpha 2 Subunit/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Receptor, Notch1/metabolism
14.
J BUON ; 20(5): 1267-75, 2015.
Article in English | MEDLINE | ID: mdl-26537074

ABSTRACT

PURPOSE: Clear cell renal cell carcinoma (ccRCC) is characterized with frequent mutations of SETD2 gene and our purpose was to explore targeted therapy for this entity. METHODS: By bioinformatic investigation of two major databases, the Genomics of Drug Sensitivity in Cancer (GDSC) database and The Cancer Genome Atlas (TCGA) database, we identified the selective PI3Kß inhibitors TGX221 and AZD6482 as selective inhibitors for ccRCC with SETD2 mutations, with AZD6482 additionally targeting PIK3CA and CDK6 mutations. RESULTS: Further investigation on AZD6482 profile revealed that mutations in RB1, KRAS, NRAS and APC contributed in drug resistance. Changes in both AZD6482-sensitive and -resistant gene sets showed limited impact on prognosis. Western blotting showed AZD6482 did not induce changes in a panel of major downstream effectors of AKT, but substantially increased PMS2 level. AZD6482 also selectively inhibited migration, invasiveness, and colony formation of ccRCC cells with SETD2 mutations. Integrative network analysis revealed complex interactions between these genes except SETD2. CONCLUSION: AZD6482 is a novel inhibitor with high selectivity for ccRCC SETD2 mutations. Increased activity of PI3K/AKT/PMS2 could play a role in SETD2 mutated ccRCC.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Histone-Lysine N-Methyltransferase/genetics , Kidney Neoplasms/drug therapy , Mutation , Phosphoinositide-3 Kinase Inhibitors , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/physiology , Gene Regulatory Networks , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Phosphatidylinositol 3-Kinases/physiology , Pyrimidinones/pharmacology , ortho-Aminobenzoates/pharmacology
16.
Blood ; 125(1): 90-101, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25342715

ABSTRACT

The cyclin-dependent kinase 6 (CDK6) and CDK4 have redundant functions in regulating cell-cycle progression. We describe a novel role for CDK6 in hematopoietic and leukemic stem cells (hematopoietic stem cells [HSCs] and leukemic stem cells [LSCs]) that exceeds its function as a cell-cycle regulator. Although hematopoiesis appears normal under steady-state conditions, Cdk6(-/-) HSCs do not efficiently repopulate upon competitive transplantation, and Cdk6-deficient mice are significantly more susceptible to 5-fluorouracil treatment. We find that activation of HSCs requires CDK6, which interferes with the transcription of key regulators, including Egr1. Transcriptional profiling of HSCs is consistent with the central role of Egr1. The impaired repopulation capacity extends to BCR-ABL(p210+) LSCs. Transplantation with BCR-ABL(p210+)-infected bone marrow from Cdk6(-/-) mice fails to induce disease, although recipient mice do harbor LSCs. Egr1 knock-down in Cdk6(-/-) BCR-ABL(p210+) LSKs significantly enhances the potential to form colonies, underlining the importance of the CDK6-Egr1 axis. Our findings define CDK6 as an important regulator of stem cell activation and an essential component of a transcriptional complex that suppresses Egr1 in HSCs and LSCs.


Subject(s)
Cyclin-Dependent Kinase 6/physiology , Early Growth Response Protein 1/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Leukemia/metabolism , Animals , Cell Cycle , Cell Transplantation , Cyclin-Dependent Kinase 6/genetics , Disease Progression , Fusion Proteins, bcr-abl/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Poly I-C/metabolism , Stem Cells/cytology , Transcription, Genetic
17.
Oncogene ; 34(4): 496-505, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-24469036

ABSTRACT

Many oncogenic viruses activate nuclear factor-κB (NF-κB) as a part of their replicative cycles. We have shown recently that persistent and potentially oncogenic activation of NF-κB by the human T-lymphotropic virus 1 (HTLV-1) oncoprotein Tax immediately triggers a host senescence response mediated by cyclin-dependent kinase inhibitors: p21(CIP1/WAF1) (p21) and p27(Kip1) (p27) Here we demonstrate that RelA/NF-κB activation by Kaposi sarcoma herpesvirus (KSHV) latency protein vFLIP also leads to p21/p27 upregulation and G1 cell cycle arrest. Remarkably, KSHV vCyclin, another latency protein coexpressed with vFLIP from a bicistronic latency-specific mRNA, was found to prevent the senescence and G1 arrest induced by HTLV-1 Tax and vFLIP, respectively. This is because of the known ability of vCyclin/cyclin-dependent kinase 6 complex to resist p21 and p27 inhibition and cause p27 degradation. In KSHV-transformed BCBL-1 cells, sustained vFLIP expression with small hairpin RNAs-mediated vCyclin depletion resulted in G1 arrest. The functional interdependence of vFLIP and vCyclin explains why they are cotranslated from the same viral mRNA. Importantly, deregulation of the G1 cyclin-dependent kinase can facilitate chronic I-κB kinases/NF-κB activation.


Subject(s)
G1 Phase Cell Cycle Checkpoints , NF-kappa B/metabolism , Viral Proteins/physiology , Cyclin-Dependent Kinase 6/physiology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Gene Products, tax , HeLa Cells , Humans , I-kappa B Kinase/physiology
18.
Mol Cell ; 53(2): 193-208, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24389100

ABSTRACT

Given the intimate link between inflammation and dysregulated cell proliferation in cancer, we investigated cytokine-triggered gene expression in different cell cycle stages. Transcriptome analysis revealed that G1 release through cyclin-dependent kinase 6 (CDK6) and CDK4 primes and cooperates with the cytokine-driven gene response. CDK6 physically and functionally interacts with the NF-κB subunit p65 in the nucleus and is found at promoters of many transcriptionally active NF-κB target genes. CDK6 recruitment to distinct chromatin regions of inflammatory genes was essential for proper loading of p65 to its cognate binding sites and for the function of p65 coactivators, such as TRIP6. Furthermore, cytokine-inducible nuclear translocation and chromatin association of CDK6 depends on the kinase activity of TAK1 and p38. These results have widespread biological implications, as aberrant CDK6 expression or activation that is frequently observed in human tumors modulates NF-κB to shape the cytokine and chemokine repertoires in chronic inflammation and cancer.


Subject(s)
Chromatin/metabolism , Cyclin-Dependent Kinase 6/physiology , NF-kappa B/genetics , Cell Cycle/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/analysis , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Interleukin-1/metabolism , Interleukin-1/physiology , Interleukin-8/genetics , Interleukin-8/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/physiology , Promoter Regions, Genetic , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcription Factor RelA/physiology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/physiology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/physiology
19.
Cell Cycle ; 12(15): 2384-94, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23839043

ABSTRACT

Cyclin D1 and its binding partners CDK4/6 are essential regulators of cell cycle progression and are implicated in cancer progression. Our aim was to investigate a potential regulatory role of these proteins in other essential tumor biological characteristics. Using a panel of breast cancer cell lines and primary human breast cancer samples, we have demonstrated the importance of these cell cycle regulators in both migration and stem-like cell activity. siRNA was used to target cyclin D1 and CDK4/6 expression, having opposing effects on both migration and stem-like cell activity dependent upon estrogen receptor (ER) expression. Inhibition of cyclin D1 or CDK4/6 increases or decreases migration and stem-like cell activity in ER-ve (ER-negative) and ER+ve (ER-positive) breast cancer, respectively. Furthermore, overexpressed cyclin D1 caused decreased migration and stem-like cell activity in ER-ve cells while increasing activity in ER+ve breast cancer cells. Treatment of breast cancer cells with inhibitors of cyclin D1 and CDK4/6 (Flavopiridol/PD0332991), currently in clinical trials, mimicked the effects observed with siRNA treatment. Re-expression of ER in two ER-ve cell lines was sufficient to overcome the effects of either siRNA or clinical inhibitors of cyclin D1 and CDK4/6.   In conclusion, cyclin D1 and CDK4/6 have alternate roles in regulation of migration and stem-like cell activity. Furthermore, these effects are highly dependent upon expression of ER. The significance of these results adds to our general understanding of cancer biology but, most importantly, could be used diagnostically to predict treatment response to cell cycle inhibition in breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Cyclin D1/physiology , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , Neoplastic Stem Cells/enzymology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin D1/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Female , Flavonoids/pharmacology , Gene Knockdown Techniques , Humans , Neoplastic Stem Cells/physiology , Piperidines/pharmacology , RNA, Small Interfering/genetics , Receptors, Estrogen/metabolism , Spheroids, Cellular/enzymology , Spheroids, Cellular/physiology
20.
Cancer Cell ; 22(4): 452-65, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23079656

ABSTRACT

D-type cyclins form complexes with cyclin-dependent kinases (CDK4/6) and promote cell cycle progression. Although cyclin D functions appear largely tissue specific, we demonstrate that cyclin D3 has unique functions in lymphocyte development and cannot be replaced by cyclin D2, which is also expressed during blood differentiation. We show that only combined deletion of p27(Kip1) and retinoblastoma tumor suppressor (Rb) is sufficient to rescue the development of Ccnd3(-/-) thymocytes. Furthermore, we show that a small molecule targeting the kinase function of cyclin D3:CDK4/6 inhibits both cell cycle entry in human T cell acute lymphoblastic leukemia (T-ALL) and disease progression in animal models of T-ALL. These studies identify unique functions for cyclin D3:CDK4/6 complexes and suggest potential therapeutic protocols for this devastating blood tumor.


Subject(s)
Cyclin D3/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Cyclin D2/physiology , Cyclin D3/physiology , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Humans , Lymphocytes/physiology , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Receptor, Notch1/physiology , Retinoblastoma Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...