Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Exp Cell Res ; 317(11): 1534-40, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21324423

RESUMO

Chromatin remodeling is required for transcriptional activation and repression. MRG15 (MORF4L1), a chromatin modulator, is a highly conserved protein and is present in complexes containing histone acetyltransferases (HATs) as well as histone deacetylases (HDACs). Loss of expression of MRG15 in mice and Drosophila results in embryonic lethality and fibroblast and neural stem/progenitor cells cultured from Mrg15 null mouse embryos exhibit marked proliferative defects when compared with wild type cells. To determine the role of MRG15 in cell cycle progression we performed chromatin immunoprecipitation with an antibody to MRG15 on normal human fibroblasts as they entered the cell cycle from a quiescent state, and analyzed various cell cycle gene promoters. The results demonstrated a 3-fold increase in MRG15 occupancy at the cdc2 promoter during S phase of the cell cycle and a concomitant increase in acetylated histone H4. H4 lysine 12 was acetylated at 24 h post-serum stimulation while there was no change in acetylation of lysine 16. HDAC1 and 2 were decreased at this promoter during cell cycle progression. Over-expression of MRG15 in HeLa cells activated a cdc2 promoter-reporter construct in a dose-dependent manner, whereas knockdown of MRG15 resulted in decreased promoter activity. In order to implicate HAT activity, we treated cells with the HAT inhibitor anacardic acid and determined that HAT inhibition results in loss of expression of cdc2 mRNA. Further, chromatin immunoprecipitation with Tip60 localizes the protein to the same 110bp stretch of the cdc2 promoter pulled down by MRG15. Additionally, we determined that cotransfection of MRG15 with the known associated HAT Tip60 had a cooperative effect in activating the cdc2 promoter. These results suggest that MRG15 is acting in a HAT complex involving Tip60 to modify chromatin via acetylation of histone H4 at the cdc2 promoter to activate transcription.


Assuntos
Ciclina B/genética , Genes cdc , Histonas/metabolismo , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo , Acetilação , Ácidos Anacárdicos/farmacologia , Proteína Quinase CDC2 , Células Cultivadas , Imunoprecipitação da Cromatina , Ciclina B/metabolismo , Quinases Ciclina-Dependentes , Fibroblastos/citologia , Fibroblastos/metabolismo , Células HeLa , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 2/antagonistas & inibidores , Humanos , Lisina Acetiltransferase 5 , RNA Mensageiro/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
2.
FASEB J ; 24(10): 3706-19, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20479119

RESUMO

The purpose of this study was to investigate the role of the mutant CUGn RNA in the induction of stress in type 1 myotonic dystrophy (DM1) cells and in the stress-mediated inhibition of protein translation in DM1. To achieve our goals, we performed HPLC-based purification of stress granules (SGs), immunoanalysis of SGs with stress markers TIA-1, CUGBP1, and ph-eIF2, site-specific mutagenesis, and examinations of RNA-protein and protein-protein interactions in myoblasts from control and DM1 patients. The cause-and-effect relationships were addressed in stable cells expressing mutant CUG repeats. We found that the mutant CUGn RNA induces formation of SGs through the increase of the double-stranded RNA-dependent protein kinase (PKR) and following inactivation of eIF2α, one of the substrates of PKR. We show that SGs trap mRNA coding for the DNA repair and remodeling factor MRG15 (MORF4L1), translation of which is regulated by CUGBP1. As the result of the trapping, the levels of MRG15 are reduced in DM1 cells and in CUG-expressing cells. These data show that CUG repeats cause stress in DM1 through the PKR-ph-eIF2α pathway inhibiting translation of certain mRNAs, such as MRG15 mRNA. The repression of protein translation by stress might contribute to the progressive muscle loss in DM1.


Assuntos
Distrofia Miotônica/genética , Biossíntese de Proteínas/genética , Repetições de Trinucleotídeos , Northern Blotting , Western Blotting , Proteínas CELF1 , Linhagem Celular , Cromatografia em Gel , Reparo do DNA , Imunofluorescência , Humanos , Distrofia Miotônica/patologia , Fosforilação , Ligação Proteica , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Exp Cell Res ; 316(1): 92-102, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19769966

RESUMO

After undergoing several rounds of divisions normal human fibroblasts enter a terminally non-dividing state referred to as cellular or replicative senescence. We cloned MORF4 (mortality factor on human chromosome 4), as a cellular senescence inducing gene that caused immortal cells assigned to complementation group B for indefinite division to stop dividing. To facilitate analyses of this gene, which is toxic to cells at low levels, we obtained stable clones of HeLa cells expressing a tetracycline-induced MORF4 construct that could be induced by doxycycline in a dose-dependent manner. MORF4 induction resulted in reduced colony formation after 14 days of culture, as previously observed. We determined that MORF4 protein was unstable and that addition of the proteasome inhibitor MG132 resulted in the accumulation of the protein. Following removal of MG132 the protein was rapidly degraded. Subcellular fractionation following MG132 treatment demonstrated that the protein accumulates primarily in the cytoplasm with some amounts present in the nucleus. It is therefore possible that MORF4 protein, which escapes degradation in the cytoplasm, is transported to the nucleus where it is functional. The results suggest that levels of MORF4 in cells must be tightly controlled and one mechanism involves stability of the protein.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Fatores de Transcrição/metabolismo , Ubiquitina/metabolismo , Benzoatos/farmacologia , Núcleo Celular/metabolismo , Proliferação de Células , Cicloeximida/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Citoplasma/metabolismo , Doxiciclina/farmacologia , Inibidores Enzimáticos/farmacologia , Furanos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Células HeLa , Humanos , Leupeptinas/farmacologia , Oligopeptídeos/farmacologia , Peptídeos/genética , Inibidores de Proteassoma , Pirazóis/farmacologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/genética , Transfecção , Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Enzimas Ativadoras de Ubiquitina/metabolismo
4.
J Neurosci Res ; 87(7): 1522-31, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19115414

RESUMO

Neurogenesis during development depends on the coordinated regulation of self-renewal and differentiation of neural precursor cells (NPCs). Chromatin regulation is a key step in self-renewal activity and fate decision of NPCs. However, the molecular mechanism or mechanisms of this regulation is not fully understood. Here, we demonstrate for the first time that MRG15, a chromatin regulator, is important for proliferation and neural fate decision of NPCs. Neuroepithelia from Mrg15-deficient embryonic brain are much thinner than those from control, and apoptotic cells increase in this region. We isolated NPCs from Mrg15-deficient and wild-type embryonic whole brains and produced neurospheres to measure the self-renewal and differentiation abilities of these cells in vitro. Neurospheres culture from Mrg15-deficient embryo grew less efficiently than those from wild type. Measurement of proliferation by means of BrdU (bromodeoxyuridine) incorporation revealed that Mrg15-deficient NPCs have reduced proliferation ability and apoptotic cells do not increase during in vitro culture. The reduced proliferation of Mrg15-deficient NPCs most likely accounts for the thinner neuroepithelia in Mrg15-deficient embryonic brain. Moreover, we also demonstrate Mrg15-deficient NPCs are defective in differentiation into neurons in vitro. Our results demonstrate that MRG15 has more than one function in neurogenesis and defines a novel role for this chromatin regulator that integrates proliferation and cell-fate determination in neurogenesis during development.


Assuntos
Proliferação de Células , Proteínas Cromossômicas não Histona/metabolismo , Neurogênese/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Transativadores/metabolismo , Adenoviridae , Animais , Apoptose/fisiologia , Encéfalo/embriologia , Encéfalo/fisiologia , Bromodesoxiuridina , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Vetores Genéticos , Proteína Glial Fibrilar Ácida , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Proteínas de Filamentos Intermediários/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Nestina , Células-Tronco/citologia , Transativadores/genética , Tubulina (Proteína)/metabolismo
5.
Mol Cell Biol ; 26(7): 2501-10, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16537896

RESUMO

When treated with DNA-damaging chemotherapy agents, many cancer cells, in vivo and in vitro, undergo a terminal growth arrest and acquire a senescence-like phenotype. We investigated the molecular basis for this in breast cancer cells following a 2-hour treatment with 1 muM doxorubicin. Treated cells arrested in G1 and G2 phases of the cell cycle, with concomitant reductions in S-phase and G2-M regulatory genes. p53 and p21 protein levels increased within hours after treatment and were maintained for 5 to 6 days but were reduced 8 days posttreatment, though the cells remained growth arrested. Levels of p130 rose after drug treatment, and it was the primary RB family member recruited to the S-phase promoters cyclin A and PCNA and G2-M promoters cyclin B and cdc2, remaining present for the entire 8-day time period. In contrast, p107 protein and promoter occupancy levels declined sharply after drug treatment. RB was recruited to only the PCNA promoter. In MCF-7 cells with p130 knockdown, p107 compensated for p130 loss at all cell cycle gene promoters examined, allowing cells to retain the growth arrest phenotype. Cells with p130 and p107 knockdown similarly arrested, while cells with knockdown of all three family members failed to downregulate cyclin A and cyclin B. These results demonstrate a mechanistic role for p130 and compensatory roles for p107 and RB in the long-term senescence-like growth arrest response of breast cancer cells to DNA damage.


Assuntos
Neoplasias da Mama/patologia , Senescência Celular/efeitos dos fármacos , Regulação para Baixo/genética , Doxorrubicina/farmacologia , Genes cdc , Regiões Promotoras Genéticas/genética , Proteína do Retinoblastoma/metabolismo , Acetilação , Fase G1/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Perfilação da Expressão Gênica , Histonas/metabolismo , Humanos , Família Multigênica , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína p107 Retinoblastoma-Like/metabolismo , Proteína p130 Retinoblastoma-Like/deficiência , Proteína p130 Retinoblastoma-Like/metabolismo , Fase S/efeitos dos fármacos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
6.
Mol Cell Biol ; 25(12): 4873-80, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15923606

RESUMO

MRGX is one of the members of MORF4/MRG family of transcriptional regulators, which are involved in cell growth regulation and cellular senescence. We have shown that MRGX and MRG15 associate with Rb in nucleoprotein complexes and regulate B-myb promoter activity. To elucidate the functions of MRGX and to explore its potential role in modulating cell growth in vivo, we have generated MrgX-deficient mice. Characterization of the expression pattern of mouse MrgX demonstrated it was ubiquitously expressed in all tissues of adult mice and also during embryogenesis and overlapped with its homolog Mrg15. MRGX and MRG15 proteins localize predominantly to the chromatin fraction in the nucleus, although a small amount of both proteins localized to the nuclear matrix. Whereas disruption of Mrg15 results in embryonic lethality, absence of MrgX did not impair mouse development and MrgX null mice are healthy and fertile. MrgX-deficient and wild-type mouse embryonic fibroblasts (MEFs) also had similar growth rates and showed no differences in cell cycle-related gene expression in response to serum stimulation. Mrg15 expression in MrgX-deficient tissues and MEFs was not upregulated compared with wild-type tissues and MEFs. MRG15 is highly conserved with orthologs present from humans to yeast and is essential for survival of mice. In contrast, MRGX, which evolved later, is expressed only in vertebrates, suggesting that the lack of phenotype of MrgX-deficient mice is secondary to a compensatory effect by the evolutionarily conserved MRG15 protein but not vice versa.


Assuntos
Proliferação de Células , Proteínas Cromossômicas não Histona/metabolismo , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Fracionamento Celular , Células Cultivadas , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Embrião de Mamíferos/anatomia & histologia , Feminino , Fibroblastos/citologia , Fibroblastos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Genes cdc , Genótipo , Humanos , Substâncias Macromoleculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Distribuição Tecidual , Transativadores/genética , Fatores de Transcrição/genética
7.
Mol Cell Biol ; 25(8): 2924-37, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15798182

RESUMO

MRG15 is a highly conserved protein, and orthologs exist in organisms from yeast to humans. MRG15 associates with at least two nucleoprotein complexes that include histone acetyltransferases and/or histone deacetylases, suggesting it is involved in chromatin remodeling. To study the role of MRG15 in vivo, we generated knockout mice and determined that the phenotype is embryonic lethal, with embryos and the few stillborn pups exhibiting developmental delay. Immunohistochemical analysis indicates that apoptosis in Mrg15-/- embryos is not increased compared with wild-type littermates. However, the number of proliferating cells is significantly reduced in various tissues of the smaller null embryos compared with control littermates. Cell proliferation defects are also observed in Mrg15-/- mouse embryonic fibroblasts. The hearts of the Mrg15-/- embryos exhibit some features of hypertrophic cardiomyopathy. The increase in size of the cardiomyocytes is most likely a response to decreased growth of the cells. Mrg15-/- embryos appeared pale, and microarray analysis revealed that alpha-globin gene expression was decreased in null versus wild-type embryos. We determined by chromatin immunoprecipitation that MRG15 was recruited to the alpha-globin promoter during dimethyl sulfoxide-induced mouse erythroleukemia cell differentiation. These findings demonstrate that MRG15 has an essential role in embryonic development via chromatin remodeling and transcriptional regulation.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Desenvolvimento Embrionário/genética , Genes Letais , Transativadores/fisiologia , Animais , Apoptose/genética , Cardiomegalia/etiologia , Ciclo Celular , Proliferação de Células , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Deleção de Genes , Marcação de Genes , Globinas/genética , Globinas/metabolismo , Coração/embriologia , Pulmão/embriologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Miocárdio/patologia , Regiões Promotoras Genéticas , Distribuição Tecidual , Transativadores/genética
8.
Structure ; 14(1): 151-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16407074

RESUMO

The ubiquitous MRG/MORF family of proteins is involved in cell senescence, or the terminal loss of proliferative potential, a model for aging and tumor suppression at the cellular level. These proteins are defined by the approximately 20 kDa MRG domain that binds a plethora of transcriptional regulators and chromatin-remodeling factors, including the histone deacetylase transcriptional corepressor mSin3A and the novel nuclear protein PAM14, and they are also known components of the Tip60/NuA4 complex via interactions with the MRG binding protein (MRGBP). We present here the crystal structure of a prototypic MRG domain from human MRG15 whose core consists of two orthogonal helix hairpins. Despite the lack of sequence similarity, the core structure has surprisingly striking homology to a DNA-interacting domain of the tyrosine site-specific recombinases XerD, lambda integrase, and Cre. Site-directed mutagenesis studies based on the X-ray structure and bioinformatics identified key residues involved in the binding of PAM14 and MRGBP.


Assuntos
Proliferação de Células , Senescência Celular/fisiologia , Proteínas de Ligação a DNA/química , Homologia Estrutural de Proteína , Fatores de Transcrição/química , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Transporte/metabolismo , Cristalografia por Raios X , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Evolução Molecular , Histona Acetiltransferases , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Nucleares/metabolismo , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
FEBS Lett ; 581(27): 5275-81, 2007 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17961556

RESUMO

MORF4-related gene on chromosome 15 (MRG15) is a core component of the NuA4/Tip60 histone acetyltransferase complex that modifies chromatin structure. We here demonstrate that Mrg15 null and heterozygous mouse embryonic fibroblasts exhibit an impaired DNA-damage response post gamma irradiation, when compared to wild-type cells. Defects in DNA-repair and cell growth, and delayed recruitment of repair proteins to sites of damage were observed. Formation of phosphorylated H2AX and 53BP1 foci was delayed in Mrg15 mutant versus wild-type cells following irradiation. These data implicate a novel role for MRG15 in DNA-damage repair in mammalian cells.


Assuntos
Proteínas Cromossômicas não Histona/deficiência , Proteínas Cromossômicas não Histona/genética , Reparo do DNA , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/efeitos da radiação , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Transativadores/deficiência , Transativadores/genética , Acetilação , Animais , Apoptose/efeitos da radiação , Núcleo Celular/metabolismo , Proliferação de Células/efeitos da radiação , Células Cultivadas , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Raios gama/efeitos adversos , Deleção de Genes , Heterozigoto , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
10.
Ann N Y Acad Sci ; 1100: 299-305, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17460191

RESUMO

The discovery that replicative cellular senescence is a dominant phenotype over immortality led to the discovery that there are at least four unique genetic subgroups of immortal cell lines that use distinct mechanistic pathways to evade cell cycle exit. Study of one of these genetic complementation groups demonstrated that one gene, MORF4, possessed the ability to induce senescence in group B cell lines. The MRG family of genes, of which MORF4 is a member, has since proven important for cellular aging, proliferation, positive and negative transcriptional regulation, and DNA damage repair. MRG15, the evolutionary ancestor of the family, is highly conserved in yeast, C. elegans, drosophila, plants, and mammals and has been implicated in chromatin remodeling in these species. Our proteomics studies have found that MRG15 is unique among mammalian genes in that it associates with both histone deacetylases and histone acetyl transferase complexes, and thus potentially plays a role in both transcriptional silencing and activation. Its knockout in mice is embryonic lethal, resulting in improper organogenesis, as well as cell proliferation and DNA damage repair defects. Future study of these genes will help clarify the role of chromatin remodeling in aging, cellular proliferation, and DNA damage repair.


Assuntos
Envelhecimento , Proteínas de Transporte/fisiologia , Histona Acetiltransferases/fisiologia , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Caenorhabditis elegans , Proteínas de Transporte/metabolismo , Diferenciação Celular , Proliferação de Células , Cromatina/metabolismo , Dano ao DNA , Reparo do DNA , Proteína 7 de Ligação a Ácidos Graxos , Histona Acetiltransferases/metabolismo , Humanos , Camundongos , Camundongos Knockout , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas Supressoras de Tumor/metabolismo
11.
Mol Cell Biol ; 24(19): 8366-73, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15367658

RESUMO

PAM14 has been found to associate in complexes with the MORF4/MRG family of proteins as well as Rb, the tumor suppressor protein. This suggested that it might be involved in cell growth, immortalization, and/or senescence. To elucidate the in vivo function of PAM14, we characterized the expression pattern of mouse Pam14 and generated PAM14-deficient (Pam14(-/-)) mice. Pam14 was widely expressed in all mouse tissues and as early as 7 days during embryonic development. Despite this ubiquitous expression in wild-type mice, Pam14(-/-) mice were healthy and fertile. Response to mitogenic stimulation and production of interleukin-2 were the same in stimulated splenic T cells from Pam14(-/-) mice as in control littermates. Cell growth rates of mouse embryonic fibroblasts (MEFs) from all three genotypes were the same, and immortalized cells were obtained from all cell cultures during continuous culture. There was also no difference in expression of growth-related genes in response to serum stimulation in the null versus control MEFs. These data demonstrate that PAM14 is not essential for normal mouse development and cell cycle control. PAM14 likely acts as an adaptor protein in nucleoprotein complexes and is probably compensated for by another functionally redundant protein(s).


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Nucleares/metabolismo , Proteína do Retinoblastoma/metabolismo , Linfócitos T/fisiologia , Transativadores/metabolismo , Animais , Proteínas de Transporte/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Interleucina-2/biossíntese , Camundongos , Mutagênese Sítio-Dirigida , Proteínas Nucleares/genética , Especificidade de Órgãos
12.
Oncogene ; 23(37): 6209-17, 2004 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-15208661

RESUMO

Defects in interferon (IFN) signaling that result in loss of expression of IFN-inducible proteins are associated with cellular immortalization, an important early event in the development of human cancer. Here we report that loss of IFN-inducible IFI 16 expression in human fibroblasts allows bypass of cellular senescence. We found that levels of IFI 16 mRNA and protein were higher in human old versus young fibroblasts and immortalization of fibroblasts with telomerase resulted in decreased expression of IFI 16. Moreover, overexpression of IFI 16 in immortalized fibroblasts strongly inhibited cell proliferation. Interestingly, knockdown of IFI 16 expression in fibroblasts inhibited p53-mediated transcription, downregulated p21(WAF1) expression, and extended the proliferation potential. Importantly, treatment of immortal cell lines with 5-aza-2'-deoxycytidine, an inhibitor of DNA methyltransferase, resulted in upregulation of IFI 16. Our observations support the idea that increased levels of IFI 16 in older populations of human fibroblasts contribute to cellular senescence.


Assuntos
Senescência Celular/fisiologia , Proteínas Nucleares/fisiologia , Fosfoproteínas/fisiologia , Linhagem Celular Transformada , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Fibroblastos/citologia , Humanos , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Transcrição Gênica/fisiologia , Proteína Supressora de Tumor p53/fisiologia
13.
Neoplasia ; 7(6): 585-93, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16036109

RESUMO

Based on previous studies, a minimal set of genetic alterations that is required to convert normal human fibroblasts into cancer cells has been defined. Essential roles for telomere maintenance and alterations in phosphatase 2A activity were inferred from experiments in which tumorigenicity was tested by injecting cells under the skin of immunodeficient mice. However, in the present experiments, the combination of SV40 large T antigen and activated Ras, without hTERT or SV40 small t antigen, was sufficient to convert nine different primary human fibroblast cell strains to a fully malignant state. The malignant behavior of the cells was demonstrated by growth of the cells into invasive tumors when the cells were injected beneath the kidney capsule of immunodeficient mice. Lung metastases and circulating tumor cells were also detected. These tumors were not immortal; cells entered crisis, from which they could be rescued by expression of hTERT. However, the same cell populations were not tumorigenic when they were injected under the skin. In this site, tumorigenicity required the expression of hTERT and SV40 small t antigen as well as SV40 large T antigen and Ras. The cellular pathways targeted by SV40 large T antigen (p53 and pRb) and those targeted by activated Ras represent a minimal set of genetic alterations required for the conversion of normal human fibroblasts into cancer cells.


Assuntos
Transformação Celular Neoplásica , Fibroblastos/citologia , Animais , Antígenos Transformantes de Poliomavirus/química , Biologia Celular , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Técnicas Genéticas , Humanos , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Metástase Neoplásica/patologia , Proteína do Retinoblastoma/metabolismo , Retroviridae/genética , Telomerase/metabolismo , Telômero/ultraestrutura , Proteína Supressora de Tumor p53/metabolismo
14.
J Gerontol A Biol Sci Med Sci ; 60(5): 543-8, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15972600

RESUMO

Mortality Factor on Chromosome 4 (MORF4) induces senescence in several immortal human cell lines. MORF-related gene on chromosome 15 (MRG15), another expressed family member, is highly conserved and expressed in yeast to humans. To determine the biological functions of human MRG15 (hMRG15) we used RNA-mediated interference (RNAi) to silence mrg-1, the Caenorhabditis elegans ortholog, and its closest homolog Y37D8A.11. Expression of mrg-1 RNAi resulted in sterility, body wall defects, vulval protrusion, and posterior developmental defects in worms. We expressed mrg-1 under its own and the cytomegalovirus promoter in human cells. Both constructs were expressed, indicating that C. elegans promoter elements are functional in mammalian cells. Overexpression from the cytomegalovirus promoter eventually resulted in cell death, possibly due to competition with hMRG15 in endogenous nucleoprotein complexes. Recent data indicate a role for yeast and human MRG15 in transcriptional regulation via chromatin remodeling. Here we demonstrate the importance of mrg-1 in development and reproduction in C. elegans and discuss its potential to impact the aging process.


Assuntos
Envelhecimento/fisiologia , Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Morte Celular/genética , Fatores de Transcrição/genética , Animais , Sequência de Bases , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células HeLa/parasitologia , Humanos , Longevidade/fisiologia , Microscopia Confocal , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
15.
J Gerontol A Biol Sci Med Sci ; 60(7): 820-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16079203

RESUMO

Most normal mammalian cell lines demonstrate limited growth capacity due to the gradual accumulation of senescent cells in the culture. Senescent cells appear initially at a low incidence, but with increasing frequency as the culture accumulates more divisions. Because it has been suggested that senescence is regulated by telomere shortening in human cells, we compared the telomere lengths of the subpopulation of senescent cells, present in presenescent cultures, with those of young cells. Senescent cells were separated from young cycling cells by either bromodeoxyuridine (BrdU) incorporation followed by Hoechst dye and light treatment or DiI staining followed by separation on a high-speed cell sorter. Our results demonstrate that telomeres of early-senescing cells are the same length, and must shorten at the same rate, as cycling sister cells in the culture. Therefore, senescent cells in young mass cultures occur as a result of a stochastic, nontelomere-dependent process that we have described: sudden senescence syndrome.


Assuntos
Senescência Celular/fisiologia , Fibroblastos/metabolismo , Pele/citologia , Telômero/metabolismo , Antimetabólitos/farmacologia , Southern Blotting , Bromodesoxiuridina/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Separação Celular , Células Cultivadas , Criança , DNA/análise , Densitometria , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/ultraestrutura , Humanos , Técnicas In Vitro , Telômero/efeitos dos fármacos , Telômero/genética , Telômero/ultraestrutura
16.
Gene ; 294(1-2): 215-24, 2002 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-12234683

RESUMO

MORF4 (mortality factor on chromosome 4) and the novel related MRG (MORF4-related gene) gene family were identified when MORF4 was shown to induce senescence in a subset of tumor cell lines. The gene on chromosome 15 (MRG15) has high similarity to Drosophila MSL3, which is a component of the dosage compensation complex. MRG15 also has a chromodomain and may therefore function as a chromatin remodeling factor in a complex(es) involving a histone acetyltransferase, similar to MSL3. To complement our studies on human MRG15, we cloned and characterized the mouse MRG15 gene. Mouse MRG15 is expressed ubiquitously in adult tissues and at various embryonic stages, and expression in adult testis is higher than in other tissues. MRG15-b, which is an alternatively spliced form of MRG15-a and has a 39-amino-acid insertion in the chromodomain, is also expressed in all mouse tissues examined and localizes to the nucleus of cells. It is possible that MRG15-b may lack the function of the chromodomain because of the additional amino acids and could potentially be the equivalent of the human MORF4 in the mouse. The mouse MRG15 gene is composed of twelve exons and spans over 24 kb DNA. Using luciferase constructs we have determined that there is a functional promoter sequence 1.8 kb upstream of the ATG start codon. This region contains no TATA box but has GC-rich regions, consistent with the ubiquitous expression we have observed.


Assuntos
Proteínas Cromossômicas não Histona/genética , Perfilação da Expressão Gênica , Regiões Promotoras Genéticas/genética , Transativadores , Região 5'-Flanqueadora/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , DNA/química , DNA/genética , Éxons , Feminino , Genes/genética , Células HeLa , Humanos , Íntrons , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Mapeamento por Restrição , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Transcrição Gênica
17.
Mech Ageing Dev ; 123(8): 927-36, 2002 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-12044941

RESUMO

Cellular senescence or replicative senescence is a state of irreversible growth arrest that somatic cells enter as a result of replicative exhaustion. This can be mimicked by culture manipulations such as Ras oncogene overexpression or treatment with various agents such as sodium butyrate and 5-azacytidine. It is believed that cellular senescence is one of the protective mechanisms against tumor formation. Genetic analyses of cellular senescence have revealed that it is dominant over immortality because whole cell fusion of normal with immortal cells yields hybrids with limited division potential. Only four complementation groups for indefinite division have been identified from extensive studies fusing different immortal human cell lines with each other. The senescence-related genes for three of the complementation groups B-D have been identified on human chromosomes 4, 1, and 7, respectively, by microcell-mediated chromosome transfer, though the existence of senescence-related genes on other chromosomes has been suggested. MORF4 was cloned as the senescence-related gene on human chromosome 4 and is a member of a new gene family, which has multiple transcription factor-like motifs. This gene family may affect cell division by modulating gene expression. Study of this novel gene family should lead to new insights regarding the mechanisms and function of cellular senescence in aging and immortalization.


Assuntos
Senescência Celular/genética , Cromossomos Humanos Par 4 , Fatores de Transcrição/genética , Animais , Fusão Celular , Humanos , Telomerase/metabolismo , Telômero
18.
Exp Gerontol ; 37(10-11): 1157-64, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12470827

RESUMO

Mortalin, also known as mthsp70/PBP74/GRP75, resides in multiple subcellular sites including mitochondria, ER, plasma membrane, cytoplasmic vesicles and cytosol. It is differentially distributed in normal and cancerous cells; the latter, when reverted back to normal phenotype, also show change in mortalin staining pattern similar to normal cells. Depending on its different subcellular niche and binding partner therein, mortalin is expected to perform multiple functions relevant to cell survival, control of proliferation and stress response.


Assuntos
Células/química , Proteínas de Choque Térmico HSP70/análise , Animais , Citoplasma/química , Retículo Endoplasmático/química , Humanos , Mitocôndrias/química , Proteínas Mitocondriais
19.
Curr Drug Targets ; 13(13): 1593-602, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22998188

RESUMO

Cellular senescence is a state of irreversible growth arrest and thought to be a tumor suppressive mechanism. In addition, it has been reported that cellular senescence may play an important role in wound healing, tissue remodeling, organismal aging and age-related diseases. This loss of ability to divide, associated with senescence, is induced by factors that are intrinsic, such as genetically defined pathways and telomere erosion, and extrinsic eg. DNA damage, oxidative stress, over-expression of oncogenes and inadequate growth conditions. The p53/p21 and RB/p16 pathways are key to the cell cycle arrest associated with cellular senescence. Extensive molecular changes occur when cells become senescent, as gene expression profiling of senescent versus young cells has demonstrated, and this is, in part, due to alterations in chromatin structure. Here, we review the molecular basis of the cell cycle arrest in cellular senescence, focusing on chromatin regulation. We also summarize our current knowledge of the role of cellular senescence in vivo.


Assuntos
Senescência Celular/genética , Cromatina/genética , Cromatina/metabolismo , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Humanos
20.
Stem Cell Res ; 7(1): 75-88, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21621175

RESUMO

Chromatin regulation is crucial for many biological processes such as transcriptional regulation, DNA replication, and DNA damage repair. We have found that it is also important for neural stem/progenitor cell (NSC) function and neurogenesis. Here, we demonstrate that expression of the cyclin-dependent kinase inhibitor p21 is specifically up-regulated in Mrg15 deficient NSCs. Knockdown of p21 expression by p21 shRNA results in restoration of cell proliferation. This indicates that p21 is directly involved in the growth defects observed in Mrg15 deficient NSCs. Activated p53 accumulates in Mrg15 deficient NSCs and this most likely accounts for the up-regulation of p21 expression in the cells. We observed decreased p53 and p21 levels and a concomitant increase in the percentage of BrdU positive cells in Mrg15 null cultures following expression of p53 shRNA. DNA damage foci, as indicated by immunostaining for γH2AX and 53BP1, are detectable in a sub-population of Mrg15 deficient NSC cultures under normal growing conditions and the majority of p21-positive cells are also positive for 53BP1 foci. Furthermore, Mrg15 deficient NSCs exhibit severe defects in DNA damage response following ionizing radiation. Our observations highlight the importance of chromatin regulation and DNA damage response in NSC function and maintenance.


Assuntos
Proteínas Cromossômicas não Histona/deficiência , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Células-Tronco Neurais/citologia , Transativadores/deficiência , Animais , Processos de Crescimento Celular/fisiologia , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Reparo do DNA , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Transativadores/genética , Transativadores/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA