Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Invest Ophthalmol Vis Sci ; 60(13): 4479-4488, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31661548

RESUMO

Purpose: Glutamate excitotoxicity seems to contribute to retinal ganglion cell (RGC) death in various eye diseases, but the underlying molecular mechanisms are not fully understood. We studied the roles of cyclin-dependent kinase inhibitors Cdkn2a and Cdkn2b, known as cellular stress-related senescence markers, in N-methyl-d-aspartate (NMDA)-induced RGC death. Methods: Gene expression was analyzed using quantitative reverse transcription (qRT)-PCR, in situ hybridization, and immunochemistry. Cdkn2a and Cdkn2b gain- and loss-of-function experiments were performed using the adeno-associated virus type 2 (AAV2)-mediated gene delivery system. AAV2-CRISPR-Cas9-mediated knockout of Cdkn2a or Cdkn2b was validated using cultured cells by T7 endonuclease I assay and Western blot analysis. The effects of altered expression of Cdkn2a and Cdkn2b on NMDA-induced RGC death were evaluated by quantification of RNA binding protein with multiple splicing (Rbpms)-immunoreactive RGCs. Results: Intravitreal NMDA injection resulted in upregulation of Cdkn2a and Cdkn2b expression in RGCs of the mouse retina. AAV2-mediated overexpression of Cdkn2b led to increased expression of Cdkn2a in RGCs, but not vice versa. Overexpression of Cdkn2b, but not Cdkn2a, resulted in a further reduction in RGC viability in NMDA-injected retinas. However, excessive levels of Cdkn2a or Cdkn2b had no effect on RGC viability in healthy mice. AAV2-CRISPR-Cas9-mediated knockout of either Cdkn2a or Cdkn2b attenuated NMDA-induced RGC death. Conclusions: Cdkn2a and Cdkn2b have pivotal roles in the regulation of excitotoxic RGC degeneration under NMDA-induced pathologic conditions. Our findings imply that Cdkn2a and Cdkn2b are novel therapeutic targets for ocular diseases displaying excitotoxicity-induced neuronal degeneration.


Assuntos
Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Agonistas de Aminoácidos Excitatórios/toxicidade , N-Metilaspartato/toxicidade , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Western Blotting , Sistemas CRISPR-Cas , Morte Celular , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Dependovirus , Imunoquímica , Hibridização In Situ , Injeções Intravítreas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parvovirinae/genética , Células Ganglionares da Retina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
2.
Exp Hematol ; 67: 32-40.e3, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30172749

RESUMO

Acute myeloid leukemia (AML) is a heterogeneous hematopoietic disorder initiated from a small subset of leukemia stem cell (LSC), which presents unrestricted self-renewal and proliferation. Long non-coding RNA HOTAIR is abundantly expressed and plays oncogenic roles in solid cancer and AML. However, whether HOTAIR regulates the self-renewal of LSC is largely unknown. Here, we reported that the expression of HOTAIR was increased in LSC than in normal hematological stem and progenitor cells (HSPCs). HOTAIR inhibition by short hairpin RNAs (shRNAs) decreased colony formation in leukemia cell lines and primary AML blasts. We then investigated the role of HOTAIR in leukemia in vivo. HOTAIR knockdown extends the survival time in U937-transplanted NSG mice. Furthermore, HOTAIR knockdown reduced infiltration of leukemic blasts, decreased frequency of LSC, and prolonged overall survival in MLL-AF9-induced murine leukemia, suggesting that HOTAIR is required for the maintenance of AML. Mechanistically, HOTAIR inhibited p15 expression through zeste homolog 2 (EZH2)-enrolled tri-methylation of Lys 27 of histone H3 (H3K27me3) in p15 promoter. In addition, p15 partially reversed the decrease of colony and proliferation induced by HOTAIR knockdown, suggesting that p15 plays an important role in the leukemogenesis by HOTAIR. In conclusion, our study suggests that HOTAIR facilitates leukemogenesis by enhancing self-renewal of LSC. HOTAIR might be a potential target for anti-LSC therapy.


Assuntos
Autorrenovação Celular/fisiologia , Transformação Celular Neoplásica/genética , Inibidor de Quinase Dependente de Ciclina p15/antagonistas & inibidores , Inativação Gênica , Código das Histonas/genética , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/citologia , RNA Longo não Codificante/fisiologia , RNA Neoplásico/fisiologia , Animais , Transplante de Medula Óssea , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Regulação Leucêmica da Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Terapia de Alvo Molecular , Proteínas de Fusão Oncogênica/genética , Regiões Promotoras Genéticas , Ensaio Tumoral de Célula-Tronco , Células U937
3.
J Gastroenterol Hepatol ; 33(4): 887-893, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29063720

RESUMO

BACKGROUND AND AIM: CDKN2A/CDKN2B locus on 9p21 is reported to be associated with various diseases, including cancer and cardiovascular and inflammatory diseases. Significant downregulation of CDKN2B-AS1 in inflamed colon tissue of inflammatory bowel disease (IBD) cases was reported in Europeans. This study aimed to confirm the suggestive association of CDKN2A/CDKN2B with IBD identified in our recent genome-wide association study (GWAS). METHODS: We examined the association of CDKN2A/CDKN2B locus with IBD in an additional sample of 574 IBD cases and 542 controls, totaling 4068 cases and 8074 controls. In silico study was performed at various levels for functional annotation of the causal variant. Co-localization of the GWAS association signals and the corresponding expression quantitative trait loci in IBD-related tissues was evaluated using eCAVIAR. RESULTS: An expanded GWAS showed genome-wide significant association of rs3731257 at 9p21 with IBD (odds ratio = 1.17, 95% confidence interval = 1.12-1.22, Pcombined  = 5.68 × 10-9 ) and Crohn's disease (odds ratio = 1.22, 95% confidence interval = 1.15-1.28, Pcombined  = 8.85 × 10-9 ) in the Korean population. Co-localization study suggested that both CDKN2B-AS1 and CDKN2A might be functionally associated with the locus in the small intestine. CONCLUSIONS: rs3731257 in CDKN2A/CDKN2B is an IBD-susceptible locus in Koreans, with a suggestive role for small intestine-specific gene regulation. Our findings suggested that alterations of the CDKN2A/CDKN2B locus could affect the pathophysiology of IBD.


Assuntos
Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor de Quinase Dependente de Ciclina p18/genética , Estudo de Associação Genômica Ampla , Doenças Inflamatórias Intestinais/genética , Povo Asiático/genética , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p18/fisiologia , Regulação da Expressão Gênica/genética , Loci Gênicos/genética , Predisposição Genética para Doença/genética , Humanos , Locos de Características Quantitativas/genética
5.
Aging Clin Exp Res ; 28(5): 813-21, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26526028

RESUMO

BACKGROUND AND AIMS: Cyclin-dependent kinase inhibitor p15(INK4b) is thought to be an important player in regulating astrocytic cell cycle. However, little is known with regard to the expression of p15(INK4b) and its function in hippocampal astrocytes. This study evaluated the expression of p15(INK4b) and its function during different development stages in hippocampal astrocytes. METHODS: In this study, we cultured hippocampal astrocytes from neonatal adult and aged rats. The expression of p15(INK4b) in neonatal, adult and aged astrocytes was examined. Short interfering RNA (siRNA) was then used to study the functional effects of p15(INK4b) down-regulation during cell cycle regulation. RESULTS: We found the expression of p15(INK4b) in hippocampal astrocytes was detectable on postnatal day 7, was expressed at moderate levels in adult mice (9 months old) astrocytes and peaked in aged rat (24 months old) astrocytes. Incubation with siRNA significantly suppressed p15(INK4b) expression at the mRNA and protein levels in astrocytes. Down-regulation of p15(INK4b) increased [(3)H]-thymidine incorporation into DNA and allowed cells to pass the G0/G1-S checkpoint in aged but not in neonatal or adult astrocytes. CONCLUSIONS: These observations suggest p15(INK4b) is expressed at a steady level in neonatal and adult rat hippocampal astrocytes with no effect on cell cycle regulation. Importantly, aged astrocyte cell cycle regulation was significantly affected by high expression levels of p15(INK4b) suggesting a role for p15(INK4b) in cell cycle regulation when it is expressed at high but not moderate or low levels in hippocampal astrocytes.


Assuntos
Astrócitos , Ciclo Celular/fisiologia , Senescência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Hipocampo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Células Cultivadas , Regulação para Baixo , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , RNA Interferente Pequeno/fisiologia , Ratos , Transdução de Sinais/fisiologia
6.
PLoS One ; 8(6): e66602, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23825548

RESUMO

The main role of Telomerase Reverse Transcriptase (TERT) is to protect telomere length from shortening during cell division. However, recent works have revealed the existence of a pool of TERT associated to mitochondria, where it plays a role in survival. We here show that in fully differentiated neurons the largest pool of cytoplasmic TERT associates to TIA1 positive RNA granules, where it binds the messenger RNA of the cyclin kinase inhibitor p15INK4B. Upon stress, p15INK4B and TERT dissociate and p15INK4B undergoes efficient translation, allowing its pro-survival function. These results unveil another mechanism implicated in the survival of fully differentiated neurons.


Assuntos
Ciclo Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Neurônios/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , RNA/metabolismo , Telomerase/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Wistar
7.
Blood ; 119(21): 5005-15, 2012 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-22461492

RESUMO

The tumor suppressor p15Ink4b is frequently inactivated by methylation in acute myeloid leukemia and premalignant myeloid disorders. Dendritic cells (DCs) as potent APCs play critical regulatory roles in antileukemic immune responses. In the present study, we investigated whether p15Ink4b can function as modulator of DC development. The expression of p15Ink4b is induced strongly during differentiation and activation of DCs, and its loss resulted in significant quantitative and qualitative impairments of conventional DC (cDC) development. Accordingly, ex vivo-generated BM-derived DCs from p15Ink4b-knockout mice express significantly decreased levels of the antigen-presenting (MHC II) and costimulatory (CD80 and CD86) molecules and have impaired immunostimulatory functions, such as antigen uptake and T-cell stimulation. Reexpression of p15Ink4b in progenitors restored these defects, and confirmed a positive role for p15Ink4b during cDC differentiation and maturation. Furthermore, we have shown herein that p15Ink4b expression increases phosphorylation of Erk1/Erk2 kinases, which leads to an elevated activity of the PU.1 transcription factor. In conclusion, our results establish p15Ink4b as an important modulator of cDC development and implicate a novel function for this tumor suppressor in the regulation of adaptive immune responses.


Assuntos
Diferenciação Celular/genética , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Células Dendríticas/fisiologia , Imunidade Adaptativa/genética , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Metilação de DNA/fisiologia , Células Dendríticas/metabolismo , Deleção de Genes , Regulação da Expressão Gênica/imunologia , Regulação da Expressão Gênica/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Regiões Promotoras Genéticas/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/fisiologia
8.
Carcinogenesis ; 31(10): 1889-96, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20663777

RESUMO

DNA double-strand breaks (DSBs) are the most deleterious lesion inflicted by ionizing radiation. Although DSBs are potentially carcinogenic, it is not clear whether complex DSBs that are refractory to repair are more potently tumorigenic compared with simple breaks that can be rapidly repaired, correctly or incorrectly, by mammalian cells. We previously demonstrated that complex DSBs induced by high-linear energy transfer (LET) Fe ions are repaired slowly and incompletely, whereas those induced by low-LET gamma rays are repaired efficiently by mammalian cells. To determine whether Fe-induced DSBs are more potently tumorigenic than gamma ray-induced breaks, we irradiated 'sensitized' murine astrocytes that were deficient in Ink4a and Arf tumor suppressors and injected the surviving cells subcutaneously into nude mice. Using this model system, we find that Fe ions are potently tumorigenic, generating tumors with significantly higher frequency and shorter latency compared with tumors generated by gamma rays. Tumor formation by Fe-irradiated cells is accompanied by rampant genomic instability and multiple genomic changes, the most interesting of which is loss of the p15/Ink4b tumor suppressor due to deletion of a chromosomal region harboring the CDKN2A and CDKN2B loci. The additional loss of p15/Ink4b in tumors derived from cells that are already deficient in p16/Ink4a bolsters the hypothesis that p15 plays an important role in tumor suppression, especially in the absence of p16. Indeed, we find that reexpression of p15 in tumor-derived cells significantly attenuates the tumorigenic potential of these cells, indicating that p15 loss may be a critical event in tumorigenesis triggered by complex DSBs.


Assuntos
Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Quebras de DNA de Cadeia Dupla , Neoplasias/etiologia , Animais , Células Cultivadas , Aberrações Cromossômicas , Deleção Cromossômica , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Reparo do DNA , Instabilidade Genômica , Humanos , Camundongos
9.
Hepatology ; 52(3): 966-74, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20583212

RESUMO

UNLABELLED: Senescence induction could be used as an effective treatment for hepatocellular carcinoma (HCC). However, major senescence inducers (p53 and p16(Ink4a)) are frequently inactivated in these cancers. We tested whether transforming growth factor-beta (TGF-beta) could serve as a potential senescence inducer in HCC. First, we screened for HCC cell lines with intact TGF-beta signaling that leads to small mothers against decapentaplegic (Smad)-targeted gene activation. Five cell lines met this condition, and all of them displayed a strong senescence response to TGF-beta1 (1-5 ng/mL) treatment. Upon treatment, c-myc was down-regulated, p21(Cip1) and p15(Ink4b) were up-regulated, and cells were arrested at G(1). The expression of p16(Ink4a) was not induced, and the senescence response was independent of p53 status. A short exposure of less than 1 minute was sufficient for a robust senescence response. Forced expression of p21(Cip1) and p15(Ink4b) recapitulated TGF-beta1 effects. Senescence response was associated with reduced nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) induction and intracellular reactive oxygen species (ROS) accumulation. The treatment of cells with the ROS scavenger N-acetyl-L-cysteine, or silencing of the NOX4 gene, rescued p21(Cip1) and p15(Ink4b) accumulation as well as the growth arrest in response to TGF-beta. Human HCC tumors raised in immunodeficient mice also displayed TGF-beta1-induced senescence. More importantly, peritumoral injection of TGF-beta1 (2 ng) at 4-day intervals reduced tumor growth by more than 75%. In contrast, the deletion of TGF-beta receptor 2 abolished in vitro senescence response and greatly accelerated in vivo tumor growth. CONCLUSION: TGF-beta induces p53-independent and p16(Ink4a)-independent, but Nox4-dependent, p21(Cip1)-dependent, p15(Ink4b)-dependent, and ROS-dependent senescence arrest in well-differentiated HCC cells. Moreover, TGF-beta-induced senescence in vivo is associated with a strong antitumor response against HCC.


Assuntos
Carcinoma Hepatocelular/fisiopatologia , Proliferação de Células , Senescência Celular/fisiologia , Neoplasias Hepáticas/fisiopatologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Humanos , Fígado/metabolismo , Cirrose Hepática/metabolismo , Camundongos , Camundongos Nus , NADPH Oxidase 4 , NADPH Oxidases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Transplante Heterólogo
10.
Carcinogenesis ; 30(7): 1252-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19451188

RESUMO

Id2 is a member of the helix-loop-helix (HLH) family of transcription regulators known to antagonize basic HLH transcription factors and proteins of the retinoblastoma tumor suppressor family and is implicated in the regulation of proliferation, differentiation, apoptosis and carcinogenesis. To investigate its proposed role in tumorigenesis, Id2 or deletion mutants were re-expressed in Id2(-/-) dermal fibroblasts. Ectopic expression of Id2 or mutants containing the central HLH domain increased S-phase cells, cell proliferation in low and normal serum and induced tumorigenesis when grafted or subcutaneously injected into athymic mice. Similar to their downregulation in human tumors, the expression of cyclin-dependent kinase inhibitors p27(KIP1) and p15(INK4b) was decreased by Id2; the former by downregulation of its promoter by the Id2 HLH domain-mediated sequestration of E12/E47. Re-expression of p27(KIP1) in Id2-overexpressing cells reverted the hyperproliferative and tumorigenic phenotype, implicating Id2 as an oncogene working through p27(KIP1). These results tie together the previously observed misregulation of Id2 with a novel mechanism for tumorigenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Fibroblastos/metabolismo , Proteína 2 Inibidora de Diferenciação/fisiologia , Fatores de Transcrição TCF/fisiologia , Animais , Proliferação de Células , Transformação Celular Neoplásica/patologia , Células Cultivadas , Fibroblastos/patologia , Sequências Hélice-Alça-Hélice , Proteína 2 Inibidora de Diferenciação/genética , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Fase S , Proteína 1 Semelhante ao Fator 7 de Transcrição
11.
Pigment Cell Melanoma Res ; 22(1): 99-110, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18983539

RESUMO

Expression profile analysis clusters Gpnmb with known pigment genes, Tyrp1, Dct, and Si. During development, Gpnmb is expressed in a pattern similar to Mitf, Dct and Si with expression vastly reduced in Mitf mutant animals. Unlike Dct and Si, Gpnmb remains expressed in a discrete population of caudal melanoblasts in Sox10-deficient embryos. To understand the transcriptional regulation of Gpnmb we performed a whole genome annotation of 2,460,048 consensus MITF binding sites, and cross-referenced this with evolutionarily conserved genomic sequences at the GPNMB locus. One conserved element, GPNMB-MCS3, contained two MITF consensus sites, significantly increased luciferase activity in melanocytes and was sufficient to drive expression in melanoblasts in vivo. Deletion of the 5'-most MITF consensus site dramatically reduced enhancer activity indicating a significant role for this site in Gpnmb transcriptional regulation. Future analysis of the Gpnmb locus will provide insight into the transcriptional regulation of melanocytes, and Gpnmb expression can be used as a marker for analyzing melanocyte development and disease progression.


Assuntos
Proteínas do Olho/genética , Melanócitos/metabolismo , Glicoproteínas de Membrana/genética , Fator de Transcrição Associado à Microftalmia/genética , Animais , Antígenos de Neoplasias/fisiologia , Sequência de Bases , Sítios de Ligação , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Elementos Facilitadores Genéticos , Proteínas do Olho/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Oxirredutases Intramoleculares/fisiologia , Luciferases/metabolismo , Antígeno MART-1 , Melanócitos/citologia , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição Associado à Microftalmia/metabolismo , Dados de Sequência Molecular , Células NIH 3T3 , Proteínas de Neoplasias/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredutases/fisiologia , Pigmentação , Fatores de Transcrição SOXE/fisiologia , Homologia de Sequência do Ácido Nucleico , Ativação Transcricional , Peixe-Zebra , Antígeno gp100 de Melanoma
12.
Leuk Res ; 32(7): 1101-11, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18037485

RESUMO

Inactivation of p15(Ink4b) expression by promoter hypermethylation occurs in up to 80% of acute myeloid leukemia (AML) cases and is particularly common in the FAB-M2 subtype of AML, which is characterized by the presence of the RUNX1-ETO translocation in 40% of cases. To establish whether the loss of p15(Ink4b) contributes to AML progression in association with RUNX1-ETO, we have expressed the RUNX1-ETO fusion protein from a retroviral vector in hematopoietic progenitor cells isolated from wild-type, p15(Ink4b) or p16(Ink4a) knockout bone marrow. Analysis of lethally irradiated recipient mice reconstituted with RUNX1-ETO-expressing cells showed that neither p15(Ink4b) or p16(Ink4a) loss significantly accelerated disease progression over the time period of one year post-transplantation. Loss of p15(Ink4b) alone resulted in increased myeloid progenitor cell frequencies in bone marrow by 10-month post-transplant and a 19-fold increase in the frequency of Lin(-)c-Kit(+)Sca-1(+) (LKS) cells that was not associated with expansion of long-term reconstituting HSC. These results strongly suggest that p15(Ink4b) loss must be accompanied by additional oncogenic changes for RUNX1-ETO-associated AML to develop.


Assuntos
Diferenciação Celular/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Leucemia Mieloide Aguda/fisiopatologia , Animais , Sequência de Bases , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Primers do DNA , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL
13.
Oncogene ; 26(55): 7665-74, 2007 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-17599058

RESUMO

Cell-cycle inhibitors of the Cip/Kip and INK4 families are involved in cellular senescence and tumor suppression. Some of these proteins, p21(Cip1), p16(INK4a) and p15(INK4b), are coexpressed in response to antiproliferative signals such as cellular senescence resulting in cell-cycle arrest. To understand the roles of these inhibitors and their synergistic effect, we have characterized the growth properties and senescent behavior of primary cells deficient in p21(Cip1) and expressing an endogenous Cdk4(R24C) (cyclin-dependent kinase) mutant (Cdk4(R24C) knock-in cells) insensitive to INK4 proteins. Inactivation of both p21(Cip1) and INK4 pathways strongly cooperate in suppressing cellular senescence in vitro. These double mutant cells behavior as immortal cultures and display high sensitivity to cellular transformation by oncogenes. Moreover, mice double mutant in the INK4 and p21(Cip1) pathways (Cdk4(R24C); p21(Cip1)-null mice) display an increased incidence of specific sarcomas, suggesting a significant cooperation between these two families of cell-cycle inhibitors in senescence responses and tumor suppression in vivo.


Assuntos
Senescência Celular/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/fisiologia , Sarcoma/genética , Animais , Quinase 4 Dependente de Ciclina/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Camundongos , Camundongos Mutantes , Sarcoma/metabolismo , Sarcoma/patologia
14.
J Biol Chem ; 282(33): 24471-6, 2007 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-17553787

RESUMO

The extracellular matrix is a crucial component in determining cell fate. Fibrillar collagen in its native form inhibits cell proliferation, whereas in its monomeric form it stimulates proliferation. The observation of elevated levels of p27(KIP1) in cells plated in the presence of fibrillar collagen has led to the assumption that this kinase inhibitor was responsible for cell cycle arrest on fibrillar collagen. Here we provide evidence that p15(INK4b), rather than p27(KIP1), is the cyclin-dependent kinase inhibitor responsible for G0/G1 arrest of human melanoma cells grown on fibrillar collagen. Additionally, we demonstrate that fibrillar collagen can also arrest cells at the G2 phase, which is mediated in part by p21(CIP1). Our data, in addition to identifying cyclin-dependent kinase inhibitors important in cell cycle arrest mediated by fibrillar collagen, demonstrate the complexity of cell cycle regulation and indicate that modulating a single cyclin-dependent kinase inhibitor does not disrupt cell proliferation in the presence of fibrillar collagen.


Assuntos
Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Colágenos Fibrilares/fisiologia , Melanoma/patologia , Inibidor de Quinase Dependente de Ciclina p27 , Fase G2 , Humanos , Fase de Repouso do Ciclo Celular
15.
Exp Hematol ; 35(3): 394-406, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17309820

RESUMO

OBJECTIVE: The tumor suppressor p15Ink4b (Ink4b) is a cell-cycle inhibitor that is inactivated in a high percentage of acute myeloid leukemia and myeloid dysplasia syndrome cases. Despite this, the role of Ink4b in hematopoiesis remains unclear. Here we examined the role of Ink4b in blood cell formation using Ink4b-deficient (Ink4b(-/-)) mice. METHODS: We compared the bone marrow (BM) of Ink4b(-/-) and wild-type mice using flow cytometric, colony-forming unit and competitive repopulating assays (CRA). The proliferation, differentiation, self-renewal, and apoptosis of progenitor cells were further compared by in vitro and in vivo methods. RESULTS: BM from Ink4b(-/-) mice contained increased numbers of granulocyte-monocyte progenitors and Gr-1(+) cells and showed a competitive advantage over wild-type cells in myeloid cell formation by CRA. Ink4b(-/-) progenitors did not demonstrate increased proliferation, self-renewing potential, or reduced apoptosis. Instead, Ink4b(-/-) common myeloid progenitors (CMPs) showed increased myeloid progenitor formation concomitant with reduced erythroid potential. CONCLUSIONS: This work establishes a role for Ink4b in regulating the differentiation of CMPs and indicates that loss of Ink4b enhances the formation of myeloid progenitors.


Assuntos
Inibidor de Quinase Dependente de Ciclina p15/deficiência , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Células-Tronco Hematopoéticas/citologia , Células Mieloides/citologia , Animais , Apoptose/fisiologia , Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias/métodos , Citometria de Fluxo , Granulócitos/citologia , Células-Tronco Hematopoéticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Células Mieloides/fisiologia
16.
Mol Vis ; 12: 983-94, 2006 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-16943770

RESUMO

PURPOSE: To evaluate the role of cyclin-dependent kinase inhibitors p57 and p15 in transforming growth factor (TGF)-beta1 or TGF-beta2 inhibited proliferation of primary cultured human limbal epithelial cells using short interfering RNA (siRNA). METHODS: Primary cultured human limbal epithelial cells were treated with TGF-beta1 or TGF-beta2 for 6 and 24 h, and total RNA extracted for RT-PCR and real-time PCR using primers for p21, p27, and p57 (CipP/Kip family) and p15 and p19 (INK4 family). Proteins were extracted for western blot analysis of p57 and p15. For RNA interference, primary cultured human limbal epithelial cells were transfected with annealed double-stranded siRNA (67 nM) specific for p57, p15, or siRNA-Fluorescein (siRNA-F; as a negative control) followed by treatment with TGF-beta1 or TGF-beta2 at 1 ng/ml. P57 and p15 were quantitatively detected by real-time PCR and western blot; and immunolocalized by immunofluorescent staining. The effects of TGF-beta1 or TGF-beta2 on cell proliferation were evaluated by BrdU incorporation and MTT assay. RESULTS: TGF-beta1 or TGF-beta2 significantly inhibited primary cultured human limbal epithelial cell proliferation measured by BrdU incorporation and MTT assay. TGF-beta1 or TGF-beta2 upregulated the expression of p57 and p15 mRNA and protein, but did not effect the expression of p19, p21, or p27. The siRNA transfection efficiency of these cells was 75% and no cellular toxicity was observed by 24 h. The TGF-beta1 or TGF-beta2 stimulated expression of p57 and p15 mRNA were markedly blocked by siRNA-p57 or siRNA-p15, respectively, but not by siRNA-F. The TGF-beta1 or TGF-beta2 suppression of epithelial proliferation measured by BrdU incorporation and MTT generation was increased to near normal levels by siRNA-p57 or siRNA-p15. Western blot and immunofluorescent staining showed that levels of p57 and p15 proteins were equally reduced in the cytoplasm and nucleus. CONCLUSIONS: These findings demonstrate that TGF-beta1 and/or TGF-beta2 inhibit proliferation of primary cultured human limbal epithelial cells and that p57 and p15 play roles in this process.


Assuntos
Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor de Quinase Dependente de Ciclina p57/fisiologia , Limbo da Córnea/citologia , Fator de Crescimento Transformador beta/farmacologia , Inibidor de Quinase Dependente de Ciclina p15/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p57/genética , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Células Epiteliais/citologia , Humanos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Transfecção , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta2
17.
Cancer J Sci Am ; 2(4): 221-4, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-9166536

RESUMO

PURPOSE: We investigate whether p16INK4 and p15INK4B inhibit cellular proliferation and exert a growth suppressive effect on esophageal cancer cells. MATERIALS AND METHODS: The growth suppressive effects of p16INK4 and p15INK4B were evaluated by transfecting vectors containing the p16INK4 cDNA or the p15INK4B cDNA, or both, constitutively driven by a cytomegalovirus promoter, into two human esophageal cancer cell lines containing or lacking endogenous p16INK4 and/or p15INK4B. RESULTS: These experiments demonstrated that in both cells lines tested, the numbers of cells surviving dramatically decreased in p16INK4-transfected and p15INK4B-transfected cells compared with control vector-transfected cells. There was no significant difference in the degree of growth inhibition between p16INK4-transfected and pI5INK4B-transfected cell lines. CONCLUSIONS: These results suggest that p16INK4 and p15INK4B play important roles in the initiation or promotion of esophageal cancer. The inactivation of p16INK4 and p15INK4B may contribute to uncontrolled growth in human cancer.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Inibidor de Quinase Dependente de Ciclina p15/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Neoplasias Esofágicas/patologia , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Neoplasias Esofágicas/metabolismo , Humanos , Técnicas In Vitro , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA