Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pathol ; 246(2): 134-140, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29952003

RESUMO

Geminin, a DNA replication licensing inhibitor, ensures faithful DNA replication in vertebrates. Several studies have shown that geminin depletion in vitro results in rereplication and DNA damage, whereas increased expression of geminin has been observed in human cancers. However, conditional inactivation of geminin during embryogenesis has not revealed any detectable DNA replication defects. In order to examine its role in vivo, we conditionally inactivated geminin in the murine colon and lung, and assessed chemically induced carcinogenesis. We show here that mice lacking geminin develop a significantly higher number of tumors and bear a larger tumor burden than sham-treated controls in urethane-induced lung and azoxymethane/dextran sodium sulfate-induced colon carcinogenesis. Survival is also significantly reduced in mice lacking geminin during lung carcinogenesis. A significant increase in the total number and grade of lesions (hyperplasias, adenomas, and carcinomas) was also confirmed by hematoxylin and eosin staining. Moreover, increased genomic aberrations, identified by increased ATR and γH2AX expression, was detected with immunohistochemistry analysis. In addition, we analyzed geminin expression in human colon cancer, and found increased expression, as well as a positive correlation with ATM/ATR levels and a non-monotonic association with γH2AX. Taken together, our data demonstrate that geminin acts as a tumor suppressor by safeguarding genome stability, whereas its overexpression is also associated with genomic instability. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Adenoma/genética , Carcinoma/genética , Neoplasias do Colo/genética , Geminina/genética , Genes Supressores de Tumor , Instabilidade Genômica , Neoplasias Pulmonares/genética , Adenoma/induzido quimicamente , Adenoma/metabolismo , Adenoma/patologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Azoximetano , Carcinoma/induzido quimicamente , Carcinoma/metabolismo , Carcinoma/patologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Geminina/deficiência , Geminina/metabolismo , Predisposição Genética para Doença , Histonas/metabolismo , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fosforilação , Uretana
2.
Dev Biol ; 409(2): 392-405, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26658318

RESUMO

Neural crest cells comprise a multipotent, migratory cell population that generates a diverse array of cell and tissue types, during vertebrate development. Enteric Nervous System controls the function of the gastrointestinal tract and is mainly derived from the vagal and sacral neural crest cells. Deregulation on self-renewal and differentiation of the enteric neural crest cells is evident in enteric nervous system disorders, such as Hirschsprung disease, characterized by the absence of ganglia in a variable length of the distal bowel. Here we show that Geminin is essential for Enteric Nervous System generation as mice that lacked Geminin expression specifically in neural crest cells revealed decreased generation of vagal neural crest cells, and enteric neural crest cells (ENCCs). Geminin-deficient ENCCs showed increased apoptosis and decreased cell proliferation during the early stages of gut colonization. Furthermore, decreased number of committed ENCCs in vivo and the decreased self-renewal capacity of enteric progenitor cells in vitro, resulted in almost total aganglionosis resembling a severe case of Hirschsprung disease. Our results suggest that Geminin is an important regulator of self-renewal and survival of enteric nervous system progenitor cells.


Assuntos
Sistema Nervoso Entérico/patologia , Geminina/metabolismo , Doença de Hirschsprung/metabolismo , Doença de Hirschsprung/patologia , Crista Neural/metabolismo , Células-Tronco/metabolismo , Animais , Contagem de Células , Morte Celular , Diferenciação Celular , Proliferação de Células , Autorrenovação Celular , Geminina/deficiência , Deleção de Genes , Intestinos/patologia , Camundongos , Crista Neural/citologia , Neuroglia/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo
3.
Stem Cells ; 33(11): 3239-53, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26140583

RESUMO

Geminin is a dual-function protein unique to multicellular animals with roles in modulating gene expression and preventing DNA re-replication. Here, we show that geminin is essential at the beginning of mammalian development to prevent DNA re-replication in pluripotent cells, exemplified by embryonic stem cells, as they undergo self-renewal and differentiation. Embryonic stem cells, embryonic fibroblasts, and immortalized fibroblasts were characterized before and after geminin was depleted either by gene ablation or siRNA. Depletion of geminin under conditions that promote either self-renewal or differentiation rapidly induced DNA re-replication, followed by DNA damage, then a DNA damage response, and finally apoptosis. Once differentiation had occurred, geminin was no longer essential for viability, although it continued to contribute to preventing DNA re-replication induced DNA damage. No relationship was detected between expression of geminin and genes associated with either pluripotency or differentiation. Thus, the primary role of geminin at the beginning of mammalian development is to prevent DNA re-replication-dependent apoptosis, a role previously believed essential only in cancer cells. These results suggest that regulation of gene expression by geminin occurs only after pluripotent cells differentiate into cells in which geminin is not essential for viability.


Assuntos
Apoptose/fisiologia , Diferenciação Celular/fisiologia , Replicação do DNA/fisiologia , Células-Tronco Embrionárias/fisiologia , Geminina/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Geminina/deficiência , Camundongos , Camundongos Transgênicos
4.
Mol Cell Biol ; 35(4): 699-715, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25487575

RESUMO

DNA rereplication is a major form of aberrant replication that causes genomic instabilities, such as gene amplification. However, little is known about which DNA polymerases are involved in the process. Here, we report that low-fidelity Y-family polymerases (Y-Pols), Pol η, Pol ι, Pol κ, and REV1, significantly contribute to DNA synthesis during rereplication, while the replicative polymerases, Pol δ and Pol ε, play an important role in rereplication, as expected. When rereplication was induced by depletion of geminin, these polymerases were recruited to rereplication sites in human cell lines. This finding was supported by RNA interference (RNAi)-mediated knockdown of the polymerases, which suppressed rereplication induced by geminin depletion. Interestingly, epistatic analysis indicated that Y-Pols collaborate in a common pathway, independently of replicative polymerases. We also provide evidence for a catalytic role for Pol η and the involvement of Pol η and Pol κ in cyclin E-induced rereplication. Collectively, our findings indicate that, unlike normal S-phase replication, rereplication induced by geminin depletion and oncogene activation requires significant contributions of both Y-Pols and replicative polymerases. These findings offer important mechanistic insights into cancer genomic instability.


Assuntos
Reparo do DNA , Replicação do DNA , DNA de Neoplasias/genética , DNA Polimerase Dirigida por DNA/genética , Regulação Neoplásica da Expressão Gênica , Genoma , Linhagem Celular Tumoral , Ciclina E/genética , Ciclina E/metabolismo , Dano ao DNA , DNA de Neoplasias/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Geminina/deficiência , Geminina/genética , Vetores Genéticos , Instabilidade Genômica , Células HCT116 , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Lentivirus/genética , Transgenes
5.
Development ; 142(1): 70-81, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25516969

RESUMO

Balancing stem cell self-renewal and initiation of lineage specification programs is essential for the development and homeostasis of the hematopoietic system. We have specifically ablated geminin in the developing murine hematopoietic system and observed profound defects in the generation of mature blood cells, leading to embryonic lethality. Hematopoietic stem cells (HSCs) accumulated in the fetal liver following geminin ablation, while committed progenitors were reduced. Genome-wide transcriptome analysis identified key HSC transcription factors as being upregulated upon geminin deletion, revealing a gene network linked with geminin that controls fetal hematopoiesis. In order to obtain mechanistic insight into the ability of geminin to regulate transcription, we examined Hoxa9 as an example of a key gene in definitive hematopoiesis. We demonstrate that in human K562 cells geminin is associated with HOXA9 regulatory elements and its absence increases HOXA9 transcription similarly to that observed in vivo. Moreover, silencing geminin reduced recruitment of the PRC2 component SUZ12 to the HOXA9 locus and resulted in an increase in RNA polymerase II recruitment and H3K4 trimethylation (H3K4me3), whereas the repressive marks H3K9me3 and H3K27me3 were reduced. The chromatin landscape was also modified at the regulatory regions of HOXA10 and GATA1. K562 cells showed a reduced ability to differentiate to erythrocytes and megakaryocytes upon geminin silencing. Our data suggest that geminin is indispensable for fetal hematopoiesis and regulates the generation of a physiological pool of stem and progenitor cells in the fetal hematopoietic system.


Assuntos
Feto/citologia , Geminina/deficiência , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/citologia , Fatores de Transcrição/genética , Animais , Contagem de Células , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Epigênese Genética , Geminina/metabolismo , Ontologia Genética , Loci Gênicos , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células K562 , Fígado/citologia , Fígado/embriologia , Camundongos , Proteínas de Neoplasias , Complexo Repressor Polycomb 2/metabolismo , Processamento de Proteína Pós-Traducional , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição/metabolismo , Transcriptoma/genética
6.
Cardiovasc Drugs Ther ; 28(5): 407-14, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25189787

RESUMO

PURPOSE: Geminin has been correlated with vascular smooth muscle cell (VSMC) proliferation, but its mechanism is unclear. We selectively silenced the geminin gene of rat VSMCs by using RNAi technology and examined how geminin regulated VSMC proliferation. METHODS: By using RNA interference in A10 cells and flow cytometry, (3)H-thymidine and 5-ethynyl-2'-deoxyuridine (EdU) measurements were used to detect VSMC proliferation. We performed a Western blot, polymerase chain reaction, and immunohistochemistry to detect the expression and location of geminin and cyclin-dependent kinase-1 (CDK1) in VSMCs. RESULTS: Silencing geminin significantly increased (3)H-thymidine and EdU incorporation in VSMCs. We observed a significant increase in (3)H-thymidine incorporation 24 h after a serum challenge in the geminin-RNAi-lentiviral vector group (4401.38 ± 438.39 cpm/mg), versus the non-targeting geminin-lentiviral vector (2836.88 ± 476.18 cpm/mg) and control groups (3069.50 ± 508.18 cpm/mg; P < 0.05). In the geminin-RNAi-lentiviral vector group, the EdU-positive cell rate was significantly increased (0.75 ± 0.03; P < 0.05), versus the non-targeting geminin-lentiviral vector (0.41 ± 0.0) or control group (0.40 ± 0.03). Geminin promoted VSMC proliferation, accelerating G0/G1-S cell-cycle progression (G0/G1 cells, 10 % decrease; S-phase cells, approximate 6 % increase) 12 h after serum withdrawal. Both CDK1 protein and mRNA expression were significantly increased in the positive group versus the controls. The immunofluorescence and co-immunoprecipitation results revealed a close interaction existed between CDK1 and the geminin gene in VSMC proliferation. CONCLUSIONS: Geminin gene inhibition could augment VSMC proliferation by increasing CDK1 expression; thus, geminin may be a potential target for treating vascular diseases, specifically VSMCs.


Assuntos
Proliferação de Células , Quinases Ciclina-Dependentes/biossíntese , Geminina/deficiência , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Regulação para Cima , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Geminina/efeitos dos fármacos , Geminina/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ratos , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...