Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
EMBO J ; 38(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30643018

RESUMO

Adult neurogenesis is involved in cognitive performance but studies that manipulated this process to improve brain function are scarce. Here, we characterized a genetic mouse model in which neural stem cells (NSC) of the subventricular zone (SVZ) were temporarily expanded by conditional expression of the cell cycle regulators Cdk4/cyclinD1, thus increasing neurogenesis. We found that supernumerary neurons matured and integrated in the olfactory bulb similarly to physiologically generated newborn neurons displaying a correct expression of molecular markers, morphology and electrophysiological activity. Olfactory performance upon increased neurogenesis was unchanged when mice were tested on relatively easy tasks using distinct odor stimuli. In contrast, intriguingly, increasing neurogenesis improved the discrimination ability of mice when challenged with a difficult task using mixtures of highly similar odorants. Together, our study provides a mammalian model to control the expansion of somatic stem cells that can in principle be applied to any tissue for basic research and models of therapy. By applying this to NSC of the SVZ, we highlighted the importance of adult neurogenesis to specifically improve performance in a challenging olfactory task.


Assuntos
Aprendizagem por Discriminação , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Odorantes/análise , Bulbo Olfatório/fisiologia , Animais , Ciclina D1/fisiologia , Quinase 4 Dependente de Ciclina/fisiologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Bulbo Olfatório/citologia , Bulbo Olfatório/efeitos dos fármacos
2.
Mol Cell ; 53(2): 193-208, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24389100

RESUMO

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.


Assuntos
Cromatina/metabolismo , Quinase 6 Dependente de Ciclina/fisiologia , NF-kappa B/genética , Ciclo Celular/genética , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/fisiologia , Quinase 6 Dependente de Ciclina/análise , Quinase 6 Dependente de Ciclina/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Interleucina-1/metabolismo , Interleucina-1/fisiologia , Interleucina-8/genética , Interleucina-8/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , MAP Quinase Quinase Quinases/fisiologia , Regiões Promotoras Genéticas , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelA/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
3.
Biochem Biophys Res Commun ; 532(3): 400-405, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-32878707

RESUMO

Hypoxia could cause vascular smooth muscle hypertrophy, leading to high pulmonary circulation resistance, pulmonary artery (PA) pressure, even pulmonary arterial hypertension (PAH). Recent studies have demonstrated the ability of mesenchymal stem cell (MSC) to ameliorate PAH but the mechanism was controversial. In this study, we revealed that the growth rate of pulmonary artery smooth muscle cells (PASMCs) treated with hypoxia was significantly increased than normal and showed lower expression of potassium channels. However, cells co-cultured with MSC showed decreased proliferation capability and down-regulated expression of ion channel of PAMSCs. The protein array data showed that the changes of PAMSCs was substantially associated with a high level of tumor necrosis factor alpha (TNFα) secretion from MSC. We further demonstrated that TNFα rescued the cell behavior of PAMSCs through activating the expression of P53 and NF-kB and inducing cell cycle arrest by P21/CDK2/CDK4 downregulation. These findings suggested that MSCs could attenuate abnormal function of PAMSCs by TNFα secretion, which was more or less associated with the beneficial effects of MSC on improving PAH.


Assuntos
Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Hipóxia/complicações , Hipóxia/fisiopatologia , Células-Tronco Mesenquimais/fisiologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Pontos de Checagem do Ciclo Celular , Técnicas de Cocultura , Quinase 2 Dependente de Ciclina/fisiologia , Quinase 4 Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Humanos , Hipertensão Pulmonar/patologia , Hipóxia/patologia , Células-Tronco Mesenquimais/patologia , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/fisiologia , Proteômica , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Transdução de Sinais
4.
Hepatology ; 70(5): 1614-1630, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31077409

RESUMO

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.


Assuntos
Neoplasias dos Ductos Biliares/etiologia , Colangiocarcinoma/etiologia , Quinase 4 Dependente de Ciclina/fisiologia , Quinase 6 Dependente de Ciclina/fisiologia , Animais , Humanos , Camundongos , Células Tumorais Cultivadas
5.
Int J Cancer ; 140(5): 1147-1158, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-27668411

RESUMO

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.


Assuntos
Linfócitos B/efeitos dos fármacos , Interleucina-10/fisiologia , Receptor Toll-Like 9/fisiologia , Linfócitos B/citologia , Divisão Celular , Linhagem Celular Transformada , Transformação Celular Neoplásica , Células Cultivadas , Ilhas de CpG , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/fisiologia , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/fisiologia , Sinergismo Farmacológico , Regulação da Expressão Gênica , Humanos , Interleucina-10/farmacologia , Linfoma/etiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Fase S/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Receptor Toll-Like 9/agonistas , Fator de Transcrição RelA/metabolismo
6.
Acta Biochim Biophys Sin (Shanghai) ; 49(7): 635-642, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28510621

RESUMO

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.


Assuntos
Ciclina D1/análise , MicroRNAs/fisiologia , Células-Tronco Embrionárias Murinas/química , Animais , Proliferação de Células , Células Cultivadas , Ciclina D1/genética , Quinase 4 Dependente de Ciclina/fisiologia , Quinase 6 Dependente de Ciclina/fisiologia , Fase G1 , Camundongos , Células-Tronco Embrionárias Murinas/citologia
7.
EMBO J ; 30(8): 1563-76, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21399612

RESUMO

RB is a key substrate of Cdks and an important regulator of the mammalian cell cycle. RB either represses E2Fs that promote cell proliferation or enhances the activity of cell-specific factors that promote differentiation, although the mechanism that facilitates this dual interaction is unclear. Here, we demonstrate that RB associates with and stabilizes pancreatic duodenal homeobox-1 (Pdx-1) that is essential for embryonic pancreas development and adult ß-cell function. Interestingly, Pdx-1 utilizes a conserved RB-interaction motif (RIM) that is also present in E2Fs. Point mutations within the RIM reduce RB-Pdx-1 complex formation, destabilize Pdx-1 and promote its proteasomal degradation. Glucose regulates RB and Pdx-1 levels, RB/Pdx-1 complex formation and Pdx-1 degradation. RB occupies the promoters of ß-cell-specific genes, and knockdown of RB results in reduced expression of Pdx-1 and its target genes. Further, RB-deficiency in vivo results in reduced pancreas size due to decreased proliferation of Pdx-1(+) pancreatic progenitors, increased apoptosis and aberrant expression of regulators of pancreatic development. These results demonstrate an unanticipated regulatory mechanism for pancreatic development and ß-cell function, which involves RB-mediated stabilization of the pancreas-specific transcription factor Pdx-1.


Assuntos
Fatores de Transcrição E2F/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Pâncreas/citologia , Pâncreas/metabolismo , Proteína do Retinoblastoma/metabolismo , Transativadores/química , Transativadores/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Western Blotting , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Imunoprecipitação da Cromatina , Quinase 4 Dependente de Ciclina/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Regulação da Expressão Gênica , Glucose/farmacologia , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Fosforilação , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Proteína do Retinoblastoma/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Nutr Cancer ; 67(3): 514-22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25751508

RESUMO

2-Deoxyglucose (2DG) is an anticancer drug with excellent safety profile. Because of its higher dose requirements, its potential is yet to translate into a monotherapy. However, recently, 2DG has been tested as an adjunct in established chemotherapeutic regimens. 2DG enhanced the potency of several chemotherapeutic agents but not all. The rationale selection of known chemotherapeutic agents to use with 2DG is hampered because of the lack of complete understanding of mechanism behind 2DG anticancer effects. Although, 2DG is a well-known glycolytic inhibitor, which inhibits the key glycolytic enzyme hexokinase, its anticancer effects cannot be fully explained by this simplistic mechanism alone. In this article, we have shown for the first time that 2DG induced a transient expression of p21 and a continuous expression of p53 in colorectal cancer cells (SW620). The treatment also caused cell cycle arrest at G0/G1 phase and induced apoptosis through the mitochondrial pathway. The effects of 2DG on p21 and p53 protein levels were totally independent of its inhibitory effect on either hexokinase or ATP levels. Results from this study provides key insights into novel molecular mechanisms of 2DG and directs rational selection of other anticancer drugs to combine with 2DG in colorectal cancer treatment.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Desoxiglucose/farmacologia , Glicólise/fisiologia , Trifosfato de Adenosina/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Quinase 4 Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/análise , Humanos , Proteína Supressora de Tumor p53/análise
9.
Hepatology ; 53(5): 1676-84, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21520178

RESUMO

UNLABELLED: Transforming growth factor beta (TGF-ß) is an important regulator of cell growth, and loss of TGF-ß signaling is a hallmark of carcinogenesis. The Smad3/4 adaptor protein ß2-spectrin (ß2SP) is emerging as a potent regulator of tumorigenesis through its ability to modulate the tumor suppressor function of TGF-ß. However, to date the role of the TGF-ß signaling pathway at specific stages of the development of hepatocellular carcinoma (HCC), particularly in relation to the activation of other oncogenic pathways, remains poorly delineated. Here we identify a mechanism by which ß2SP, a crucial Smad3 adaptor, modulates cyclin dependent kinase 4 (CDK4), cell cycle progression, and suppression of HCC. Increased expression of ß2SP inhibits phosphorylation of the retinoblastoma gene product (Rb) and markedly reduces CDK4 expression to a far greater extent than other CDKs and cyclins. Furthermore, suppression of CDK4 by ß2SP efficiently restores Rb hypophosphorylation and cell cycle arrest in G(1) . We further demonstrate that ß2SP interacts with CDK4 and Smad3 in a competitive and TGF-ß-dependent manner. In addition, haploinsufficiency of cdk4 in ß2sp(+/-) mice results in a dramatic decline in HCC formation compared to that observed in ß2sp(+/-) mice. CONCLUSION: ß2SP deficiency leads to CDK4 activation and contributes to dysregulation of the cell cycle, cellular proliferation, oncogene overexpression, and the formation of HCCs. Our data highlight CDK4 as an attractive target for the pharmacologic inhibition of HCC and demonstrate the importance of ß2sp(+/-) mice as a model of preclinical efficacy in the treatment of HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Proliferação de Células , Quinase 4 Dependente de Ciclina/fisiologia , Neoplasias Hepáticas/patologia , Espectrina/fisiologia , Animais , Quinase 4 Dependente de Ciclina/genética , Haploinsuficiência , Camundongos , Proteína Smad3/fisiologia , Fator de Crescimento Transformador beta
10.
Blood ; 113(12): 2706-14, 2009 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-19060242

RESUMO

Stem cell factor (SCF) promotes synergistic cellular proliferation in combination with several growth factors, and appears important for normal natural killer (NK)-cell development. CD34(+) hematopoietic precursor cells (HPCs) require interleukin-15 (IL-15) for differentiation into human NK cells, and this effect can be mimicked by IL-2. Culture of CD34(+) HPCs or some primary human NK cells in IL-2/15 and SCF results in enhanced growth compared with either cytokine alone. The molecular mechanisms responsible for this are unknown and were investigated in the present work. Activation of NK cells by IL-2/15 increases expression of c-kit whose kinase activity is required for synergy with IL-2/15 signaling. Mitogen-activated protein kinase (MAPK) signaling intermediaries that are activated both by SCF and IL-2/15 are enhanced in combination to facilitate earlier cell-cycle entry. The effect results at least in part via enhanced MAPK-mediated modulation of p27 and CDK4. Collectively the data reveal a novel mechanism by which SCF enhances cellular proliferation in combination with IL-2/15 in primary human NK cells.


Assuntos
Interleucina-2/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Fator de Células-Tronco/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Meios de Cultura Livres de Soro , Quinase 4 Dependente de Ciclina/fisiologia , Sinergismo Farmacológico , Humanos , Interleucina-15/farmacologia , Células Matadoras Naturais/citologia , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-kit/biossíntese , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/fisiologia , Piridinas/farmacologia , Proteínas Recombinantes/farmacologia , Tirfostinas/farmacologia , Regulação para Cima/efeitos dos fármacos
11.
Mol Pharmacol ; 77(2): 195-201, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19917880

RESUMO

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor with constitutive activities and those induced by xenobiotic ligands, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). One unexplained cellular role for the AHR is its ability to promote cell cycle progression in the absence of exogenous ligands, whereas treatment with exogenous ligands induces cell cycle arrest. Within the cell cycle, progression from G(1) to S phase is controlled by sequential phosphorylation of the retinoblastoma protein (RB1) by cyclin D-cyclin-dependent kinase (CDK) 4/6 complexes. In this study, the functional interactions between the AHR, CDK4, and cyclin D1 (CCND1) were investigated as a potential mechanism for the cell cycle regulation by the AHR. Time course cell cycle and molecular experiments were performed in human breast cancer cells. The results demonstrated that the AHR and CDK4 interact within the cell cycle, and the interaction was disrupted upon TCDD treatment. The disruption was temporally correlated with G(1) cell cycle arrest and decreased phosphorylation of RB1. Biochemical reconstitution assays using in vitro-translated protein recapitulated the AHR and CDK4 interaction and showed that CCND1 was also part of the complex. In vitro assays for CDK4 kinase activity demonstrated that RB1 phosphorylation by the AHR/CDK4/CCND1 complex was reduced in the presence of TCDD. The results suggest that the AHR interacts in a complex with CDK4 and CCND1 in the absence of exogenous ligands to facilitate cell cycle progression. This interaction is disrupted by exogenous ligands, such as TCDD, to induce G(1) cell cycle arrest.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/fisiologia , Quinase 4 Dependente de Ciclina/fisiologia , Receptores de Hidrocarboneto Arílico/fisiologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Dibenzodioxinas Policloradas/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia
12.
Endocrinology ; 161(10)2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32877917

RESUMO

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.


Assuntos
Ciclina D1/genética , Quinase 4 Dependente de Ciclina/fisiologia , Quinase 6 Dependente de Ciclina/fisiologia , Neoplasias das Paratireoides/genética , Neoplasias das Paratireoides/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Ciclina D1/metabolismo , Hiperparatireoidismo/genética , Hiperparatireoidismo/metabolismo , Hiperparatireoidismo/patologia , Camundongos , Camundongos Transgênicos , Mutação , Neoplasias das Paratireoides/patologia , Fosforilação/genética , Transdução de Sinais/genética
13.
Cell Rep ; 31(2): 107504, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32294430

RESUMO

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.


Assuntos
Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Aminopiridinas/farmacologia , Animais , Benzimidazóis/farmacologia , Neoplasias da Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Ciclina D/metabolismo , Ciclina D/fisiologia , Quinase 4 Dependente de Ciclina/fisiologia , Quinase 6 Dependente de Ciclina/fisiologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Camundongos , Fosforilação , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
14.
Dermatol Online J ; 15(3): 1, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19379645

RESUMO

The influences exerted by the epidermal growth factor receptor (EGFR) on the skin act at multiple levels, which involve compartments that normally express EGFR. These include the basal and suprabasal layers of the epidermis, sebaceous glands, and the outer root sheath of the hair follicles. The physiological roles of EGFR ensure epidermal renewal and integrity, along with a gatekeeping and function and hair growth stimulation functions. Important cellular functions that are altered during EGF receptor blocking therapy consist of epidermal differentiation, proliferation, apoptosis, and migration, with an overall dominating effect of inducing growth arrest and terminal differentiation of the keratinocytes in the basal layers. The effects of EGFR blockage on the hair cycle include terminal differentiation of the hair follicle, which in certain cases may be associated with trichomegaly. Trichomegaly of the eyelashes may occur as an isolated occurrence or, frequently, as part of a generalized phenomenon that may be associated with the use of the EGFR inhibitors. Molecular changes associated with EGFR blockage are discussed, relevant to their association with hair growth. Modulation of Akt, AP2alpha, CDK4, Notch-1, p27KIP1, and Hedgehog expression are involved in the initiation of the hair cycle and inducement of the anagen phase, followed by proliferation and differentiation of the hair follicles. Epidermal growth factor receptor inhibitors have been developed as therapeutic molecules directed against cancer; in these regimens the knowledge of EGF receptor signaling functions has been translated into significant clinical results. However, among their various collateral effects on the skin, hair growth is observed to occur in certain patients. A particular "wavy" hair phenotype is observed during the pharmacological EGFR receptor blockade, just as in murine transgenic models that carry loss of function of TGF-alpha or EGFR genes. A better characterization of the individual roles pertaining to the EGF family ligands and receptors, has the potential provide new strategies for the management of hair loss.


Assuntos
Alopecia/terapia , Receptores ErbB/fisiologia , Animais , Antralina/farmacologia , Antralina/uso terapêutico , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Pesquisa Biomédica , Medicina Clínica , Quinase 4 Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p27 , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/fisiologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/deficiência , Oftalmopatias/complicações , Oftalmopatias/fisiopatologia , Cabelo/anormalidades , Cabelo/crescimento & desenvolvimento , Folículo Piloso/fisiologia , Proteínas Hedgehog/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/análise , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Camundongos , Família Multigênica , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Notch/fisiologia , Transdução de Sinais , Fator de Transcrição AP-2/fisiologia , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética
15.
Oncogene ; 38(25): 4962-4976, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30833638

RESUMO

The retinoblastoma protein (RB) restricts cell cycle gene expression and entry into the cell cycle. The RB-related protein p130 forms the DREAM (DP, RB-like, E2F, and MuvB) complex and contributes to repression of cell cycle-dependent genes during quiescence. Although both RB and DREAM bind and repress an overlapping set of E2F-dependent gene promoters, it remains unclear whether they cooperate to restrict cell cycle entry. To test the specific contributions of RB and DREAM, we generated RB and p130 knockout cells in primary human fibroblasts. Knockout of both p130 and RB yielded higher levels of cell cycle gene expression in G0 and G1 cells compared to cells with knockout of RB alone, indicating a role for DREAM and RB in repression of cell cycle genes. We observed that RB had a dominant role in E2F-dependent gene repression during mid to late G1 while DREAM activity was more prominent during G0 and early G1. Cyclin D-Cyclin-Dependent Kinase 4 (CDK4)-dependent phosphorylation of p130 occurred during early G1, and led to the release of p130 and MuvB from E2F4 and decreased p130 and MuvB binding to cell cycle promoters. Specific inhibition of CDK4 activity by palbociclib blocked DREAM complex disassembly during cell cycle entry. In addition, sensitivity to CDK4 inhibition was dependent on RB and an intact DREAM complex in both normal cells as well as in palbociclib-sensitive cancer cell lines. Although RB knockout cells were partially resistant to CDK4 inhibition, RB and p130 double knockout cells were significantly more resistant to palbociclib treatment. These results indicate that DREAM cooperates with RB in repressing E2F-dependent gene expression and cell cycle entry and supports a role for DREAM as a therapeutic target in cancer.


Assuntos
Ciclo Celular/genética , Proliferação de Células/genética , Ciclina D/fisiologia , Quinase 4 Dependente de Ciclina/fisiologia , Proteínas Interatuantes com Canais de Kv/fisiologia , Proteínas Repressoras/fisiologia , Proteína do Retinoblastoma/fisiologia , Células A549 , Pontos de Checagem do Ciclo Celular/genética , Células Cultivadas , Regulação para Baixo/genética , Regulação da Expressão Gênica , Humanos , Recém-Nascido , Masculino , Transdução de Sinais/genética
16.
Cancer Res ; 79(20): 5245-5259, 2019 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-31395606

RESUMO

Cyclin-dependent kinase 4 (CDK4) is well-known for its role in regulating the cell cycle, however, its role in cancer metabolism, especially mTOR signaling, is undefined. In this study, we established a connection between CDK4 and lysosomes, an emerging metabolic organelle crucial for mTORC1 activation. On the one hand, CDK4 phosphorylated the tumor suppressor folliculin (FLCN), regulating mTORC1 recruitment to the lysosomal surface in response to amino acids. On the other hand, CDK4 directly regulated lysosomal function and was essential for lysosomal degradation, ultimately regulating mTORC1 activity. Pharmacologic inhibition or genetic inactivation of CDK4, other than retaining FLCN at the lysosomal surface, led to the accumulation of undigested material inside lysosomes, which impaired the autophagic flux and induced cancer cell senescence in vitro and in xenograft models. Importantly, the use of CDK4 inhibitors in therapy is known to cause senescence but not cell death. To overcome this phenomenon and based on our findings, we increased the autophagic flux in cancer cells by using an AMPK activator in combination with a CDK4 inhibitor. The cotreatment induced autophagy (AMPK activation) and impaired lysosomal function (CDK4 inhibition), resulting in cell death and tumor regression. Altogether, we uncovered a previously unknown role for CDK4 in lysosomal biology and propose a novel therapeutic strategy to target cancer cells. SIGNIFICANCE: These findings uncover a novel function of CDK4 in lysosomal biology, which promotes cancer progression by activating mTORC1; targeting this function offers a new therapeutic strategy for cancer treatment.


Assuntos
Quinase 4 Dependente de Ciclina/fisiologia , Lisossomos/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Neoplasias/fisiologia , Adenilato Quinase/metabolismo , Aminopiridinas/farmacologia , Aminopiridinas/uso terapêutico , Animais , Autofagossomos/fisiologia , Autofagia/fisiologia , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Compostos de Bifenilo , Linhagem Celular Tumoral , Senescência Celular/fisiologia , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/genética , Sinergismo Farmacológico , Feminino , Técnicas de Inativação de Genes , Humanos , Insulina/fisiologia , Lisossomos/ultraestrutura , Camundongos , Camundongos Endogâmicos NOD , Terapia de Alvo Molecular , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteínas Proto-Oncogênicas/metabolismo , Pironas/farmacologia , Pironas/uso terapêutico , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Tiofenos/farmacologia , Tiofenos/uso terapêutico , Proteínas Supressoras de Tumor/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Clin Invest ; 115(11): 3228-38, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16239970

RESUMO

Vascular SMC proliferation is a crucial event in occlusive cardiovascular diseases. PPARalpha is a nuclear receptor controlling lipid metabolism and inflammation, but its role in the regulation of SMC growth remains to be established. Here, we show that PPARalpha controls SMC cell-cycle progression at the G1/S transition by targeting the cyclin-dependent kinase inhibitor and tumor suppressor p16(INK4a) (p16), resulting in an inhibition of retinoblastoma protein phosphorylation. PPARalpha activates p16 gene transcription by both binding to a canonical PPAR-response element and interacting with the transcription factor Sp1 at specific proximal Sp1-binding sites of the p16 promoter. In a carotid arterial-injury mouse model, p16 deficiency results in an enhanced SMC proliferation underlying intimal hyperplasia. Moreover, PPARalpha activation inhibits SMC growth in vivo, and this effect requires p16 expression. These results identify an unexpected role for p16 in SMC cell-cycle control and demonstrate that PPARalpha inhibits SMC proliferation through p16. Thus, the PPARalpha/p16 pathway may be a potential pharmacological target for the prevention of cardiovascular occlusive complications of atherosclerosis.


Assuntos
Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidores do Crescimento/fisiologia , Músculo Liso Vascular/patologia , PPAR alfa/fisiologia , Túnica Íntima/patologia , Regulação para Cima , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Células Cultivadas , Quinase 4 Dependente de Ciclina/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Regulação para Baixo/genética , Fase G1/genética , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Células HeLa , Humanos , Hiperplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , PPAR alfa/deficiência , PPAR alfa/genética , Fosforilação , Proteína do Retinoblastoma/metabolismo , Fase S/genética , Transcrição Gênica/fisiologia , Túnica Íntima/citologia , Regulação para Cima/genética
18.
Naunyn Schmiedebergs Arch Pharmacol ; 377(2): 167-77, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18228003

RESUMO

The initiation of endoplasmic reticulum (ER) stress has been suggested to play potential roles in hepatocarcinogenesis. However, many obstacles remain as to whether ER stress plays a role in carcinogenesis or tumoricide. This study sought to identify the signals that can serve as anticancer effectors in cells in response to ER stress. Tunicamycin (an N-glycosylation inhibitor) inhibited cell proliferation with IC(50) values of 0.19 and 0.62 microg/ml in hepatoma (Hep) 3B and HepG2 cells, respectively. It induced G1 arrest of the cell cycle in both cell lines. The anticancer mechanism of tunicamycin was investigated in Hep3B cells. Tunicamycin induced a rapid decline of cyclin D1 and cyclin A expression and an early increase of glucose-related protein (GRP) 78 and growth arrest and DNA damage-inducible transcription factor (GADD) 153 levels. Cyclin A was the most sensitive regulator to tunicamycin-triggered degradation mechanism. The association of p27(Kip1) with cyclin D1/cyclin-dependent kinase (Cdk) 4 was also increased by tunicamycin. The inhibition of GADD153 expression by transfection of GADD153 antisense did not modify tunicamycin-induced G1 arrest and cyclin/Cdk expressions. The knockdown of GRP78 expression by the siRNA transfection technique moderately increased tunicamycin-induced apoptosis but not the antiproliferative effect by sulforhodamine B assay. We suggest that tunicamycin induces G1 arrest through down-regulation of cyclins and Cdks, in which cyclin A is more susceptible to ER stress-triggered degradation mechanism in Hep3B cells. The increased association of p27(Kip1) with cyclin D1/Cdk4 may also contribute to tunicamycin-induced cell-cycle arrest. GADD153 and GRP78 play a minor role in tunicamycin-mediated antiproliferative effect, although GRP78 moderately inhibits apoptosis in Hep3B cells. These data provide evidence that cell-cycle regulators are susceptible factors in hepatocellular carcinoma (HCC) responsive to ER stress.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Retículo Endoplasmático/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Tunicamicina/farmacologia , Carcinoma Hepatocelular/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina D1/fisiologia , Quinase 4 Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p27 , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias Hepáticas/patologia , Chaperonas Moleculares/fisiologia , Fator de Transcrição CHOP/fisiologia
19.
Breast Cancer ; 25(6): 742-752, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29951881

RESUMO

BACKGROUND: MiRNAs regulate a variety of biological processes, such as cell proliferation and apoptosis and play critical roles in cancer progression. Accumulating studies have demonstrated that miR-1301-3p could regulate the development and progression of multiple cancers, but its biological behaviors in breast cancer (BC) are still elusive. METHODS: The expression of miR-1301-3p was determined in BC tissues and cell lines using quantitative real-time PCR analysis. The effects of miR-1301-3p on BC cell growth, proliferation, cell cycle distribution, and apoptosis were also explored in vitro using MTT, colony formation and Flow cytometry assays. The potential target gene of miR-1301-3p was determined by dual-luciferase reporter assay and verified by quantitative real-time PCR and western blot analysis. RESULTS: We found the expression of miR-1301-3p was observably significantly down-regulated in BC tissues and cell lines. MiR-1301-3p expression in BC tissues was significantly associated with tumor size and clinical stage. Gain-of-function assays demonstrated that miR-1301-3p inhibited the cell growth and proliferation in breast cancer cell lines, MCF-7 and T-47D. Moreover, up-regulation of miR-1301-3p induced cell cycle G0/G1 phase arrest and apoptosis. Mechanistically, up-regulation of miR-1301-3p reduced the expression of CDK4, Cyclin D1, Bcl-2, but elevated the expression of p21, Bad and Bax. ICT1 was confirmed as a direct target of miR-1301-3p. Furthermore, ICT1 overexpression could partially reverse the effects of miR-1301-3p on BC cell proliferation, cell cycle progression and apoptosis. CONCLUSION: Our observations suggested that miR-1301-3p inhibits cell proliferation via inducing cell cycle arrest and apoptosis through targeting ICT1, and might be a therapeutic target for BC.


Assuntos
Apoptose , Neoplasias da Mama/patologia , Pontos de Checagem da Fase G1 do Ciclo Celular , MicroRNAs/fisiologia , Proteínas/genética , Adulto , Idoso , Linhagem Celular Tumoral , Proliferação de Células , Quinase 4 Dependente de Ciclina/análise , Quinase 4 Dependente de Ciclina/fisiologia , Feminino , Humanos , MicroRNAs/análise , Pessoa de Meia-Idade , Proteínas Ribossômicas
20.
J Orthop Res ; 36(6): 1581-1589, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29194728

RESUMO

Chordomas are rare bone tumors and treatment is commonly based on a combination of surgery and radiotherapy. There is no standard chemotherapy treatment for chordoma. The aim of this study was to determine the expression of cyclin-dependent kinase 4 (CDK4) in chordoma and its therapeutic implications. We evaluated CDK4 expression both in chordoma cell lines and in chordoma tissues. Also, we investigated the functional roles of CDK4 in chordoma cell growth and proliferation. Furthermore, the therapeutic implications of targeting CDK4 in chordoma were evaluated. We found CDK4 highly expressed in chordoma cell lines and in a majority (97.7%) of chordoma tissues. Higher CDK4 expression correlated with metastasis and recurrence of chordoma. Treatment of chordoma cells using CDK4 inhibitor palbociclib could efficiently inhibit chordoma cells growth and proliferation. These data demonstrate that targeting CDK4 may be useful as a novel strategy in the treatment of chordoma. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1581-1589, 2018.


Assuntos
Neoplasias Ósseas/patologia , Cordoma/patologia , Quinase 4 Dependente de Ciclina/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Ósseas/química , Neoplasias Ósseas/tratamento farmacológico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cordoma/química , Cordoma/tratamento farmacológico , Quinase 4 Dependente de Ciclina/análise , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosforilação , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA