Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 619(7969): 363-370, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37407814

RESUMO

In mammalian cells, the decision to proliferate is thought to be irreversibly made at the restriction point of the cell cycle1,2, when mitogen signalling engages a positive feedback loop between cyclin A2/cyclin-dependent kinase 2 (CDK2) and the retinoblastoma protein3-5. Contrary to this textbook model, here we show that the decision to proliferate is actually fully reversible. Instead, we find that all cycling cells will exit the cell cycle in the absence of mitogens unless they make it to mitosis and divide first. This temporal competition between two fates, mitosis and cell cycle exit, arises because cyclin A2/CDK2 activity depends upon CDK4/6 activity throughout the cell cycle, not just in G1 phase. Without mitogens, mitosis is only observed when the half-life of cyclin A2 protein is long enough to sustain CDK2 activity throughout G2/M. Thus, cells are dependent on mitogens and CDK4/6 activity to maintain CDK2 activity and retinoblastoma protein phosphorylation throughout interphase. Consequently, even a 2-h delay in a cell's progression towards mitosis can induce cell cycle exit if mitogen signalling is lost. Our results uncover the molecular mechanism underlying the restriction point phenomenon, reveal an unexpected role for CDK4/6 activity in S and G2 phases and explain the behaviour of all cells following loss of mitogen signalling.


Assuntos
Ciclo Celular , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Fase G2 , Fase S , Animais , Ciclina A2/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/metabolismo , Mitógenos/deficiência , Mitógenos/metabolismo , Mitose , Fosforilação , Proteína do Retinoblastoma/química , Proteína do Retinoblastoma/metabolismo , Quinase 6 Dependente de Ciclina/deficiência , Quinase 6 Dependente de Ciclina/metabolismo , Fase G1
2.
Protein Sci ; 29(4): 978-990, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31930537

RESUMO

Most genes in evolutionarily complex genomes are expressed to multiple protein isoforms, but there is not yet any simple high-throughput approach to identify these isoforms. Using an oversimplified top-down LC-MS/MS strategy, we detected, around the 26-kD position of SDS-PAGE, proteins produced from 782 genes in a Cdk4-/- mouse embryonic fibroblast cell line. Interestingly, only 213 (27.24%, about one-fourth) of these 782 genes have their proteins with a theoretical molecular mass (TMM) 10% smaller or larger than 26 kD, that is, between 23 and 29 kD, the range set as allowed variation in SDS-PAGE. These 213 proteins are considered as the wild type (WT). The remaining three-fourths includes proteins from 66 (9.44%) genes with a TMM smaller than 23 kD and proteins from 503 (64.32%, nearly two-thirds) genes with a TMM larger than 29 kD; these proteins are categorized into a larger-group or a smaller-group, respectively, for their appearance at a higher or lower position of SDS-PAGE. For instance, at this 26-kD position we detected proteins from the Rps27a, Snrpf, Hist1h4a, and Rps25 genes whose proteins' TMM is 8.6, 9.7, 11.4, and 13.7 kD, respectively, and detected proteins from the Plelc1 and Prkdc genes, whose largest isoform is 533.9 and 471.1 kD, respectively. We extrapolate that many of those proteins migrating unexpectedly in SDS-PAGE may be isoforms besides the WT protein. Moreover, we also detected a Cdk4 protein in this Cdk4-/- cell line, thus wondering whether some of other gene-knockout cells or organisms show similar incompleteness of the knockout.


Assuntos
Eletroforese em Gel de Poliacrilamida , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Animais , Linhagem Celular , Quinase 4 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/genética , Camundongos
3.
Dev Neurobiol ; 78(7): 660-670, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29570951

RESUMO

Little is known about the molecular players driving proliferation of neural progenitor cells (NPCs) during embryonic mouse development. Here, we demonstrate that proliferation of NPCs in the developing forebrain depends on a particular combination of cell cycle regulators. We have analyzed the requirements for members of the cyclin-dependent kinase (cdk) family using cdk-deficient mice. In the absence of either cdk4 or cdk6, which are both regulators of the G1 phase of the cell cycle, we found no significant effects on the proliferation rate of cortical progenitor cells. However, concomitant loss of cdk4 and cdk6 led to a drastic decrease in the proliferation rate of NPCs, specifically the basal progenitor cells of both the dorsal and ventral forebrain at embryonic day 13.5 (E13.5). Moreover, basal progenitors in the forebrain of Cdk4;Cdk6 double mutant mice exhibited altered cell cycle characteristics. Cdk4;cdk6 deficiency led to an increase in cell cycle length and cell cycle exit of mutant basal progenitor cells in comparison to controls. In contrast, concomitant ablation of cdk2 and cdk6 had no effect on the proliferation of NCPs. Together, our data demonstrate that the expansion of the basal progenitor pool in the developing telencephalon is dependent on the presence of distinct combinations of cdk molecules. Our results provide further evidence for differences in the regulation of proliferation between apical and basal progenitors during cortical development. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 660-670, 2018.


Assuntos
Proliferação de Células/fisiologia , Quinase 4 Dependente de Ciclina/deficiência , Quinase 6 Dependente de Ciclina/deficiência , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Células-Tronco/metabolismo , Animais , Contagem de Células , Ciclo Celular/fisiologia , Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/genética , Camundongos Knockout , Prosencéfalo/patologia , Células-Tronco/patologia
4.
Haematologica ; 100(4): 431-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25616574

RESUMO

Mouse knockouts of Cdk2 and Cdk4 are individually viable whereas the double knockouts are embryonic lethal due to heart defects, and this precludes the investigation of their overlapping roles in definitive hematopoiesis. Here we use a conditional knockout mouse model to investigate the effect of combined loss of Cdk2 and Cdk4 in hematopoietic cells. Cdk2(fl/fl)Cdk4(-/-)vavCre mice are viable but displayed a significant increase in erythrocyte size. Cdk2(fl/fl)Cdk4(-/-)vavCre mouse bone marrow exhibited reduced phosphorylation of the retinoblastoma protein and reduced expression of E2F target genes such as cyclin A2 and Cdk1. Erythroblasts lacking Cdk2 and Cdk4 displayed a lengthened G1 phase due to impaired phosphorylation of the retinoblastoma protein. Deletion of the retinoblastoma protein rescued the increased size displayed by erythrocytes lacking Cdk2 and Cdk4, indicating that the retinoblastoma/Cdk2/Cdk4 pathway regulates erythrocyte size. The recovery of platelet counts following a 5-fluorouracil challenge was delayed in Cdk2(fl/fl)Cdk4(-/-)vavCre mice revealing a critical role for Cdk2 and Cdk4 in stress hematopoiesis. Our data indicate that Cdk2 and Cdk4 play important overlapping roles in homeostatic and stress hematopoiesis, which need to be considered when using broad-spectrum cyclin-dependent kinase inhibitors for cancer therapy.


Assuntos
Plaquetas/metabolismo , Quinase 2 Dependente de Ciclina/genética , Eritrócitos/citologia , Hematopoese/genética , Estresse Fisiológico , Animais , Tamanho Celular , Quinase 2 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/genética , Feminino , Deleção de Genes , Hematócrito , Células-Tronco Hematopoéticas/metabolismo , Imunofenotipagem , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Ploidias , Proteína do Retinoblastoma/genética
5.
Mol Cell Biol ; 34(12): 2121-34, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24687853

RESUMO

Pituitary tumors develop in about one-quarter of the population, and most arise from the anterior lobe (AL). The pituitary gland is particularly sensitive to genetic alteration of genes involved in the cyclin-dependent kinase (CDK) inhibitor (CKI)-CDK-retinoblastoma protein (Rb) pathway. Mice heterozygous for the Rb mutation develop pituitary tumors, with about 20% arising from the AL. Perplexingly, none of the CKI-deficient mice reported thus far develop pituitary AL tumors. In this study, we show that deletion of p19(Ink4d) (p19), a CKI gene, in mice results in spontaneous development of tumors in multiple organs and tissues. Specifically, more than one-half of the mutant mice developed pituitary hyperplasia or tumors predominantly in the AL. Tumor development is associated with increased cell proliferation and enhanced activity of Cdk4 and Cdk6 and phosphorylation of Rb protein. Though Cdk4 is indispensable for postnatal pituitary cell proliferation, it is not required for the hyperproliferative pituitary phenotype caused by p19 loss. Loss of p19 phosphorylates Rb in Cdk4(-/-) pituitary AL cells and mouse embryonic fibroblasts (MEFs) and rescues their proliferation defects, at least partially, through the activation of Cdk6. These results provide the first genetic evidence that p19 is a tumor suppressor and the major CKI gene that controls pituitary AL cell proliferation.


Assuntos
Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Adeno-Hipófise/metabolismo , Adeno-Hipófise/patologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Peso Corporal , Carcinogênese/metabolismo , Carcinogênese/patologia , Proliferação de Células , Quinase 4 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Diabetes Mellitus Experimental/patologia , Embrião de Mamíferos/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Deleção de Genes , Loci Gênicos , Infertilidade Feminina/patologia , Infertilidade Masculina/patologia , Ilhotas Pancreáticas/patologia , Células Lúteas/metabolismo , Células Lúteas/patologia , Masculino , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Fase S
6.
J Clin Invest ; 124(4): 1672-84, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24614102

RESUMO

The G1 kinase CDK4 is amplified or overexpressed in some human tumors and promotes tumorigenesis by inhibiting known tumor suppressors. Here, we report that CDK4 deficiency markedly accelerated lymphoma development in the Eµ-Myc transgenic mouse model of B lymphoma and that silencing or loss of CDK4 augmented the tumorigenic potential of Myc-driven mouse and human B cell lymphoma in transplant models. Accelerated disease in CDK4-deficient Eµ-Myc transgenic mice was associated with rampant genomic instability that was provoked by dysregulation of a FOXO1/RAG1/RAG2 pathway. Specifically, CDK4 phosphorylated and inactivated FOXO1, which prevented FOXO1-dependent induction of Rag1 and Rag2 transcription. CDK4-deficient Eµ-Myc B cells had high levels of the active form of FOXO1 and elevated RAG1 and RAG2. Furthermore, overexpression of RAG1 and RAG2 accelerated lymphoma development in a transplant model, with RAG1/2-expressing tumors exhibiting hallmarks of genomic instability. Evaluation of human tumor samples revealed that CDK4 expression was markedly suppressed, while FOXO1 expression was elevated, in several subtypes of human non-Hodgkin B cell lymphoma. Collectively, these findings establish a context-specific tumor suppressor function for CDK4 that prevents genomic instability, which contributes to B cell lymphoma. Furthermore, our data suggest that targeting CDK4 may increase the risk for the development and/or progression of lymphoma.


Assuntos
Quinase 4 Dependente de Ciclina/deficiência , Genes myc , Instabilidade Genômica , Linfoma de Células B/enzimologia , Linfoma de Células B/genética , Animais , Quinase 4 Dependente de Ciclina/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Xenoenxertos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Linfoma de Células B/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais
7.
Glia ; 61(9): 1443-55, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23839966

RESUMO

Little is known about the molecular mechanisms driving proliferation of glial cells after an insult to the central nervous system (CNS). To test the hypothesis that the G1 regulator cyclin D1 is critical for injury-induced cell division of glial cells, we applied an injury model that causes brain damage within a well-defined region. For this, we injected the neurotoxin ibotenic acid into the prefrontal cortex of adult mice, which leads to a local nerve cell loss but does not affect the survival of glial cells. Here, we show that cyclin D1 immunoreativity increases drastically after neurotoxin injection. We find that the cyclin D1-immunopositive (cyclin D1+) cell population within the lesioned area consists to a large extent of Olig2+ oligodendrocyte progenitor cells. Analysis of cyclin D1-deficient mice demonstrates that the proliferation rate of Olig2+ cells diminishes upon loss of cyclin D1. Further, we show that cyclin-dependent kinase (cdk) 4, but not cdk6 or cdk2, is essential for driving cell division of Olig2-expressing cells in our injury model. These data suggest that distinct cell cycle proteins regulate proliferation of Olig2+ progenitor cells following a CNS insult.


Assuntos
Células-Tronco Adultas/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Lesões Encefálicas/patologia , Proliferação de Células , Córtex Cerebral/patologia , Ciclina D1/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Análise de Variância , Animais , Bromodesoxiuridina , Proliferação de Células/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Ciclina D1/deficiência , Quinase 2 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/deficiência , Quinase 6 Dependente de Ciclina/deficiência , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Ácido Ibotênico/toxicidade , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Neurotoxinas/toxicidade , Fator de Transcrição 2 de Oligodendrócitos , Fatores de Tempo
8.
Exp Cell Res ; 315(17): 2914-20, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19619530

RESUMO

Mip130/LIN-9 is part of a large complex that includes homologs of the Drosophila dREAM (drosophila RB-like, E2F, and Myb) and C. elegans DRM complexes. This complex also includes proteins such as Mip40/LIN-37, Mip120/LIN-54, and LIN-52. In mammalian cells, Mip130/LIN-9 specifically associates with the p107/p130-E2F4 repressor complex in G0/G1 and with B-Myb in S-phase. However, little is known about how the transition occurs and whether Mip130/LIN-9 contributes to the repressor effect of p107/p130. In this report, we demonstrate that Mip130/LIN-9, Mip40/LIN-37, Mip120/LIN-54, and Sin3b form a core complex, the Mip Core Complex or LIN Complex (MCC/LINC), which is detectable in all phases of the cell cycle. This complex specifically recruits transcriptional repressors such as p107, p130, E2F4 and HDAC1 in G0/G1, and B-Myb in S-phase. Importantly, we provide strong evidence that the transition between repressors and activators of transcription is mediated by CDK4, through the phosphorylation of the pocket proteins, p107 and p130. The requirement for CDK4 activity is bypassed by the deletion of the first 84 amino acids (Mip130/LIN-9(Delta84)), since this mutant is unable to interact with p107/p130 in G0/G1, while maintaining its association with B-Myb. Importantly, the Mip130/LIN-9(Delta84) allele rescues the low expression of G1/S genes observed in CDK4(-/-) MEFs demonstrating that Mip130/LIN-9 contributes to the repression of these E2F-regulated genes in G0/G1.


Assuntos
Quimiocina CCL4/genética , Quinase 4 Dependente de Ciclina/metabolismo , Fator de Transcrição E2F4/metabolismo , Proteínas Supressoras de Tumor/genética , Células 3T3 , Animais , Sítios de Ligação , Ciclo Celular , Linhagem Celular , Quimiocina CCL4/química , Quimiocina CCL4/metabolismo , Quinase 4 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/genética , Fator de Transcrição E2F4/deficiência , Fator de Transcrição E2F4/genética , Deleção de Genes , Humanos , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fase S , Deleção de Sequência , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo
9.
Proc Natl Acad Sci U S A ; 106(2): 486-91, 2009 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-19129496

RESUMO

The G(1)/S-phase transition is a well-toned switch in the mammalian cell cycle. Cdk2, Cdk4, and the rate-limiting tumor suppressor retinoblastoma protein (Rb) have been studied in separate animal models, but interactions between the kinases and Rb in vivo have yet to be investigated. To further dissect the regulation of the G(1) to S-phase progression, we generated Cdk2(-/-)Cdk4(-/-)Rb(-/-) (TKO) mutant mice. TKO mice died at midgestation with major defects in the circulatory systems and displayed combined phenotypes of Rb(-/-) and Cdk2(-/-)Cdk4(-/-) mutants. However, TKO mouse embryonic fibroblasts were not only resistant to senescence and became immortal but displayed enhanced S-phase entry and proliferation rates similar to wild type. These effects were more remarkable in hypoxic compared with normoxic conditions. Interestingly, depletion of the pocket proteins by HPV-E7 or p107/p130 shRNA in the absence of Cdk2/Cdk4 elicited a mechanism for the G(1)/S regulation with increased levels of p27(Kip1) binding to Cdk1/cyclin E complexes. Our work indicates that the G(1)/S transition can be controlled in different ways depending on the situation, resembling a regulatory network.


Assuntos
Proteína Quinase CDC2/metabolismo , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/deficiência , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fibroblastos/citologia , Interfase/genética , Proteína do Retinoblastoma/deficiência , Animais , Proliferação de Células , Células Cultivadas , Senescência Celular/genética , Fase G1 , Hipóxia , Camundongos , Camundongos Knockout , Complexos Multiproteicos/fisiologia , Fase S
10.
Mol Cell Neurosci ; 37(3): 519-27, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18191580

RESUMO

Peripheral myelin formation depends on axonal signals that tightly control proliferation and differentiation of the associated Schwann cells. Here we demonstrate that the molecular program controlling proliferation of Schwann cells switches at birth. We have analyzed the requirements for three members of the cyclin-dependent kinase (cdk) family in Schwann cells using cdk-deficient mice. Mice lacking cdk4 showed a drastic decrease in the proliferation rate of Schwann cells at postnatal days 2 and 5, but proliferation was unaffected at embryonic day 18. In contrast, ablation of cdk2 and cdk6 had no significant influence on postnatal Schwann cell proliferation. Taken together, these findings indicate that postnatal Schwann cell proliferation is uniquely controlled by cdk4. Despite the lack of the postnatal wave of Schwann cell proliferation, axons were normally myelinated in adult cdk4-deficient sciatic nerves. Following nerve injury, Schwann cells lacking cdk4 were unable to re-enter the cell cycle, while Schwann cells deficient in cdk2 or cdk6 displayed proliferation rates comparable to controls. We did not observe compensatory effects such as elevated cdk4 levels in uninjured or injured nerves of cdk2 or cdk6-deficient mice. Our data demonstrate that prenatal and postnatal Schwann cell proliferation are driven by distinct molecular cues, and that postnatal proliferation is not a prerequisite for the generation of Schwann cell numbers adequate for correct myelination.


Assuntos
Proliferação de Células , Quinase 4 Dependente de Ciclina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Bainha de Mielina/metabolismo , Células de Schwann/fisiologia , Neuropatia Ciática/enzimologia , Animais , Animais Recém-Nascidos , Bromodesoxiuridina/metabolismo , Ciclo Celular/fisiologia , Células Cultivadas , Quinase 2 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/deficiência , Quinase 6 Dependente de Ciclina/deficiência , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Knockout , Ratos , Degeneração Walleriana/metabolismo
11.
Mol Cell ; 22(4): 432-3, 2006 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-16713571

RESUMO

The mouse embryo is surprisingly resistant to loss of individual cyclins or cyclin-dependent kinases. In a recent issue of Developmental Cell, Berthet et al. (2006) describe collaboration of Cdk2 and Cdk4 in embryogenesis that is revealed only upon their simultaneous loss, resulting in inappropriate activation of the retinoblastoma protein and embryonic lethality.


Assuntos
Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/enzimologia , Animais , Ciclo Celular , Proliferação de Células , Células Cultivadas , Quinase 2 Dependente de Ciclina/deficiência , Quinase 2 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/genética , Desenvolvimento Embrionário , Fibroblastos/citologia , Fibroblastos/enzimologia , Camundongos , Camundongos Knockout
12.
Dev Cell ; 10(5): 563-73, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16678773

RESUMO

Mouse knockouts of Cdk2 and Cdk4 have demonstrated that, individually, these genes are not essential for viability. To investigate whether there is functional redundancy, we have generated double knockout (DKO) mice. Cdk2-/- Cdk4-/- DKOs die during embryogenesis around E15 as a result of heart defects. We observed a gradual decrease of Retinoblastoma protein (Rb) phosphorylation and reduced expression of E2F-target genes, like Cdc2 and cyclin A2, during embryogenesis and in embryonic fibroblasts (MEFs). DKO MEFs are characterized by a decreased proliferation rate, impaired S phase entry, and premature senescence. HPV-E7-mediated inactivation of Rb restored normal expression of E2F-inducible genes, senescence, and proliferation in DKO MEFs. In contrast, loss of p27 did not rescue Cdk2-/- Cdk4-/- phenotypes. Our results demonstrate that Cdk2 and Cdk4 cooperate to phosphorylate Rb in vivo and to couple the G1/S phase transition to mitosis via E2F-dependent regulation of gene expression.


Assuntos
Quinase 2 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/deficiência , Embrião de Mamíferos/anormalidades , Proteína do Retinoblastoma/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Quinase 2 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/genética , Fatores de Transcrição E2F/antagonistas & inibidores , Fibroblastos/citologia , Inativação Gênica , Hematopoese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus , Fenótipo , Fosforilação , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/química
13.
Exp Cell Res ; 312(13): 2465-75, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16730350

RESUMO

It has been proposed that C. elegans LIN-9 functions downstream of CDK4 in a pathway that regulates cell proliferation. Here, we report that mammalian BARA/LIN-9 is a predominantly nuclear protein that inhibits cell proliferation. More importantly, we demonstrate that BARA/LIN-9 also acts downstream of cyclin D/CDK4 in mammalian cells since (i) its antiproliferative effect is partially blocked by coexpression of cyclin D1, and (ii) a mutant form that lacks the first 84 amino acids rescues several phenotypic alterations observed in mice null for cdk4. Interestingly, mutation of BARA/LIN-9 restores the expression of E2F target genes in CDK4 null MEFs, indicating that the wild-type protein plays a role in the expression of genes required for the G1/S transition.


Assuntos
Alelos , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Quinase 4 Dependente de Ciclina/deficiência , Fatores de Transcrição E2F/antagonistas & inibidores , Mutação/genética , Proteínas Repressoras/antagonistas & inibidores , Animais , Ciclo Celular , Quinase 4 Dependente de Ciclina/genética , DNA/biossíntese , Fatores de Transcrição E2F/metabolismo , Embrião de Mamíferos/embriologia , Feminino , Fertilidade/genética , Fibroblastos/citologia , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Células NIH 3T3 , Proteínas Nucleares/metabolismo , Ovário/citologia , Fenótipo , Hipófise/citologia , Proteínas Repressoras/metabolismo , Testículo/citologia , Proteínas Supressoras de Tumor/metabolismo
14.
Int J Biochem Cell Biol ; 38(5-6): 873-93, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16253543

RESUMO

Type 2 diabetes is increasingly viewed as a disease of insulin deficiency due not only to intrinsic pancreatic beta-cell dysfunction but also to reduction of beta-cell mass. It is likely that, in diabetes-prone subjects, the regulated beta-cell turnover that adapts cell mass to body's insulin requirements is impaired, presumably on a genetic basis. We still have a limited knowledge of how and when this derangement occurs and what might be the most effective therapeutic strategy to preserve beta-cell mass. The animal models of type 2 diabetes with reduced beta-cell mass described in this review can be extremely helpful (a) to have insight into the mechanisms underlying the defective growth or accelerated loss of beta-cells leading to the beta-cell mass reduction; (b) to investigate in prospective studies the mechanisms of compensatory adaptation and subsequent failure of a reduced beta-cell mass. Furthermore, these models are of invaluable importance to test the effectiveness of potential therapeutic agents that either stimulate beta-cell growth or inhibit beta-cell death.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Células Secretoras de Insulina/patologia , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Ciclina D2 , Quinase 4 Dependente de Ciclina/deficiência , Ciclinas/deficiência , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 2/patologia , Exenatida , Retardo do Crescimento Fetal/fisiopatologia , Proteínas de Homeodomínio , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Transgênicos , Peptídeos/uso terapêutico , Fosfoproteínas/deficiência , Proteínas Proto-Oncogênicas c-akt/deficiência , Ratos , Receptor de Insulina , Proteínas Quinases S6 Ribossômicas 70-kDa/deficiência , Transativadores/deficiência , Peçonhas/uso terapêutico , eIF-2 Quinase/deficiência
15.
Cancer Res ; 65(22): 10174-8, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16288002

RESUMO

Previous work has shown that cyclin D1 expression is required for neu- and ras-induced, but not wnt- or c-myc-induced, breast tumorigenesis in mice. Although cyclin D1 binds and activates cyclin-dependent kinase 4 (Cdk4), thereby mediating activation of a program of E2F-dependent gene expression, it has been suggested that the oncogenic activities of cyclin D1 are independent of Cdk4. To determine whether Cdk4 expression is required for breast tumorigenesis in mice, we have generated compound mice ectopically expressing the neu or wnt oncogenes in the mammary glands of wild-type and Cdk4-/- mice. Our results show that Cdk4 expression is required for efficient neu-induced tumorigenesis but is dispensable for wnt-induced breast tumorigenesis. In contrast to results previously observed in the mammary glands of cyclin D1-/- virgin females, our results show defects in mammary gland development in Cdk4-/- virgin females, suggesting differences in compensatory mechanisms in the absence of either subunit of the cyclin D1/Cdk4 complex. These results suggest that drugs targeted to inhibit Cdk4 activities could be developed to specifically treat certain breast tumors as Cdk4 is not essential for viability.


Assuntos
Transformação Celular Neoplásica/metabolismo , Quinase 4 Dependente de Ciclina/biossíntese , Genes erbB-2/fisiologia , Neoplasias Mamárias Experimentais/enzimologia , Animais , Transformação Celular Neoplásica/genética , Quinase 2 Dependente de Ciclina/biossíntese , Quinase 2 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/biossíntese , Quinase 6 Dependente de Ciclina/genética , Feminino , Masculino , Glândulas Mamárias Animais/enzimologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Proteína Wnt1/genética
16.
Artigo em Inglês | MEDLINE | ID: mdl-16869759

RESUMO

Most human tumors harbor mutations that misregulate the early phases of the cell cycle. Here, we summarize genetic evidence, mostly obtained in our laboratory using strains of gene-targeted mice, that provides direct experimental support for a role of Cdk4 in tumor development. Moreover, these genetic studies challenge some well-established concepts regarding the role of Cdks during the early phases of the cell cycle. For instance, they have illustrated that Cdk4 and Cdk6 are not essential for cell division during embryonic development except in the hematopoietic system. More surprisingly, mice lacking Cdk2 survive for over 2 years without detectable abnormalities except in their germ cells, indicating that Cdk2 is essential for meiosis but dispensable for the normal mitotic cell cycle. Cdk2 is also dispensable for cell cycle inhibition and tumor suppression by the Cip/Kip inhibitors, p21(Cip1) and p27(Kip1). These observations have important implications not only to understand cell cycle regulation, but also to validate Cdks as potential targets for the development of therapeutic strategies to block proliferation of tumor cells.


Assuntos
Ciclo Celular/genética , Ciclo Celular/fisiologia , Quinases Ciclina-Dependentes/genética , Mutação , Neoplasias/enzimologia , Neoplasias/genética , Animais , Quinase 2 Dependente de Ciclina/deficiência , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/fisiologia , Quinase 4 Dependente de Ciclina/deficiência , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/fisiologia , Quinase 6 Dependente de Ciclina/deficiência , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Quinases Ciclina-Dependentes/deficiência , Quinases Ciclina-Dependentes/fisiologia , Feminino , Morte Fetal/genética , Genes Letais , Humanos , Meiose , Melanoma Experimental/enzimologia , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Mitose , Neoplasias/patologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...