Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
EMBO Rep ; 17(11): 1609-1623, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27596623

RESUMO

The CHD1 gene, encoding the chromo-domain helicase DNA-binding protein-1, is one of the most frequently deleted genes in prostate cancer. Here, we examined the role of CHD1 in DNA double-strand break (DSB) repair in prostate cancer cells. We show that CHD1 is required for the recruitment of CtIP to chromatin and subsequent end resection during DNA DSB repair. Our data support a role for CHD1 in opening the chromatin around the DSB to facilitate the recruitment of homologous recombination (HR) proteins. Consequently, depletion of CHD1 specifically affects HR-mediated DNA repair but not non-homologous end joining. Together, we provide evidence for a previously unknown role of CHD1 in DNA DSB repair via HR and show that CHD1 depletion sensitizes cells to PARP inhibitors, which has potential therapeutic relevance. Our findings suggest that CHD1 deletion, like BRCA1/2 mutation in ovarian cancer, may serve as a marker for prostate cancer patient stratification and the utilization of targeted therapies such as PARP inhibitors, which specifically target tumors with HR defects.


Assuntos
DNA Helicases/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Biomarcadores , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Cromatina , Quebras de DNA de Cadeia Dupla , DNA Helicases/deficiência , DNA Helicases/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Endodesoxirribonucleases , Humanos , Masculino , Proteínas Nucleares/genética , Inibidores de Poli(ADP-Ribose) Polimerases/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Neoplasias da Próstata/genética , Reparo de DNA por Recombinação
3.
Biochem Biophys Res Commun ; 473(1): 200-205, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-27012198

RESUMO

Recent studies involving several viral systems have highlighted the importance of cellular intrinsic defense mechanisms through nuclear antiviral proteins that restrict viral propagation. These factors include among others components of PML nuclear bodies, the nuclear DNA sensor IFI16, and a potential restriction factor PHF13/SPOC1. For several nuclear replicating DNA viruses, it was shown that these factors sense and target viral genomes immediately upon nuclear import. In contrast to the anticipated view, we recently found that incoming adenoviral genomes are not targeted by PML nuclear bodies. Here we further explored cellular responses against adenoviral infection by focusing on specific conditions as well as additional nuclear antiviral factors. In line with our previous findings, we show that neither interferon treatment nor the use of specific isoforms of PML nuclear body components results in co-localization between incoming adenoviral genomes and the subnuclear domains. Furthermore, our imaging analyses indicated that neither IFI16 nor PHF13/SPOC1 are likely to target incoming adenoviral genomes. Thus our findings suggest that incoming adenoviral genomes may be able to escape from a large repertoire of nuclear antiviral mechanisms, providing a rationale for the efficient initiation of lytic replication cycle.


Assuntos
Adenoviridae/genética , Proteínas de Ligação a DNA/imunologia , Genoma Viral , Proteínas Nucleares/imunologia , Fosfoproteínas/imunologia , Fatores de Transcrição/imunologia , Adenoviridae/fisiologia , Infecções por Adenoviridae/imunologia , Linhagem Celular Tumoral , Técnica Indireta de Fluorescência para Anticorpo , Interações Hospedeiro-Patógeno , Humanos , Interferons/farmacologia , Microscopia de Fluorescência , Neutrófilos/citologia , Neutrófilos/virologia , Proteínas Nucleares/genética , Replicação Viral
4.
PLoS Pathog ; 9(11): e1003775, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24278021

RESUMO

Little is known about immediate phases after viral infection and how an incoming viral genome complex counteracts host cell defenses, before the start of viral gene expression. Adenovirus (Ad) serves as an ideal model, since entry and onset of gene expression are rapid and highly efficient, and mechanisms used 24-48 hours post infection to counteract host antiviral and DNA repair factors (e.g. p53, Mre11, Daxx) are well studied. Here, we identify an even earlier host cell target for Ad, the chromatin-associated factor and epigenetic reader, SPOC1, recently found recruited to double strand breaks, and playing a role in DNA damage response. SPOC1 co-localized with viral replication centers in the host cell nucleus, interacted with Ad DNA, and repressed viral gene expression at the transcriptional level. We discovered that this SPOC1-mediated restriction imposed upon Ad growth is relieved by its functional association with the Ad major core protein pVII that enters with the viral genome, followed by E1B-55K/E4orf6-dependent proteasomal degradation of SPOC1. Mimicking removal of SPOC1 in the cell, knock down of this cellular restriction factor using RNAi techniques resulted in significantly increased Ad replication, including enhanced viral gene expression. However, depletion of SPOC1 also reduced the efficiency of E1B-55K transcriptional repression of cellular promoters, with possible implications for viral transformation. Intriguingly, not exclusive to Ad infection, other human pathogenic viruses (HSV-1, HSV-2, HIV-1, and HCV) also depleted SPOC1 in infected cells. Our findings provide a general model for how pathogenic human viruses antagonize intrinsic SPOC1-mediated antiviral responses in their host cells. A better understanding of viral entry and early restrictive functions in host cells should provide new perspectives for developing antiviral agents and therapies. Conversely, for Ad vectors used in gene therapy, counteracting mechanisms eradicating incoming viral DNA would increase Ad vector efficacy and safety for the patient.


Assuntos
Adenoviridae/metabolismo , Infecções por Adenovirus Humanos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Imunidade Inata , Proteólise , Fatores de Transcrição/metabolismo , Adenoviridae/genética , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/genética , Proteínas E4 de Adenovirus/metabolismo , Infecções por Adenovirus Humanos/genética , Proteínas de Ligação a DNA/genética , Células HEK293 , Humanos , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Fatores de Transcrição/genética
6.
Nucleic Acids Res ; 40(22): 11363-79, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23034801

RESUMO

Survival time-associated plant homeodomain (PHD) finger protein in Ovarian Cancer 1 (SPOC1, also known as PHF13) is known to modulate chromatin structure and is essential for testicular stem-cell differentiation. Here we show that SPOC1 is recruited to DNA double-strand breaks (DSBs) in an ATM-dependent manner. Moreover, SPOC1 localizes at endogenous repair foci, including OPT domains and accumulates at large DSB repair foci characteristic for delayed repair at heterochromatic sites. SPOC1 depletion enhances the kinetics of ionizing radiation-induced foci (IRIF) formation after γ-irradiation (γ-IR), non-homologous end-joining (NHEJ) repair activity, and cellular radioresistance, but impairs homologous recombination (HR) repair. Conversely, SPOC1 overexpression delays IRIF formation and γH2AX expansion, reduces NHEJ repair activity and enhances cellular radiosensitivity. SPOC1 mediates dose-dependent changes in chromatin association of DNA compaction factors KAP-1, HP1-α and H3K9 methyltransferases (KMT) GLP, G9A and SETDB1. In addition, SPOC1 interacts with KAP-1 and H3K9 KMTs, inhibits KAP-1 phosphorylation and enhances H3K9 trimethylation. These findings provide the first evidence for a function of SPOC1 in DNA damage response (DDR) and repair. SPOC1 acts as a modulator of repair kinetics and choice of pathways. This involves its dose-dependent effects on DNA damage sensors, repair mediators and key regulators of chromatin structure.


Assuntos
Cromatina/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Raios gama , Heterocromatina , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Tolerância a Radiação , Reparo de DNA por Recombinação , Proteínas Repressoras/metabolismo , Proteína 28 com Motivo Tripartido
7.
J Cell Sci ; 124(Pt 18): 3137-48, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21852425

RESUMO

SPOC1 (PHF13) is a recently identified protein that has been shown to dynamically associate with somatic chromatin, to modulate chromatin compaction and to be important for proper cell division. Here, we report on the expression of SPOC1 in promyelocytic leukaemia zinc finger (PLZF)-positive undifferentiated spermatogonial stem cells (SSCs) of the mouse testis. To investigate further the biological function of SPOC1 in germ cells we generated Spoc1 mutant mice from a gene-trap embryonic stem cell clone. Postpubertal homozygous Spoc1(-/-) animals displayed a pronounced progressive loss of germ cells from an initially normal germ epithelium of the testis tubules leading to testis hypoplasia. This loss first affected non-SSC stages of germ cells and then, at a later time point, the undifferentiated spermatogonia. Remarkably, successive loss of all germ cells (at >20 weeks of age) was preceded by a transient increase in the number of undifferentiated A(aligned) (A(al)) spermatogonia in younger mice (at >10 weeks of age). The number of primary Spoc1(-/-) gonocytes, the proliferation of germ cells, and the initiation and progression of meiosis was normal, but we noted a significantly elevated level of apoptosis in the Spoc1(-/-) testis. Taken together, the data argue that SPOC1 is indispensable for stem cell differentiation in the testis and for sustained spermatogenesis.


Assuntos
Células-Tronco Adultas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Testículo/metabolismo , Fatores de Transcrição/metabolismo , Células-Tronco Adultas/patologia , Animais , Apoptose/genética , Diferenciação Celular/genética , Sobrevivência Celular/genética , Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Mutação/genética , Espermatogênese/genética , Espermatogônias/patologia , Testículo/patologia , Fatores de Transcrição/genética
9.
Histochem Cell Biol ; 138(5): 759-72, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22777346

RESUMO

Human spermatogonia (Spg) and their fetal precursors express fibroblast growth factor receptor 3 (FGFR3). To further elucidate the role of FGFR3 in the control of Spg self-renewal, proliferation, and/or differentiation, and to narrow down the FGFR3-positive cell type(s) in the normal adult human testis, tissue sections and whole mount preparations of seminiferous tubules were analyzed combining immunofluorescence and confocal fluorescence microscopy. FGFR3 protein was chiefly observed in cellular membranes and cytoplasmic vesicles of a subpopulation of type A Spg, which comprised the chromatin rarefaction zone-containing type A(dark). Cytoplasmic expression of FGFR3 and nuclear expression of proliferation-associated antigen KI-67 were mutually exclusive. Similarly, FGFR3-positive Spg were negative for Doublesex and Mab-3 related transcription factor 1 (DMRT1). By contrast, undifferentiated embryonic cell transcription factor 1 (UTF1) and survival time-associated PHD finger in ovarian cancer 1 protein (SPOC1) were co-expressed in the nuclei of FGFR3-positive Spg. Whole mounted seminiferous tubules illustrated the clonogenic arrangement of the FGFR3/UTF1 double-positive Spg, which mainly occurred as pairs or quadruplets and, different from the KIT-positive Spg, showed no overlap with KI-67 labeled clusters. Taken together, in the adult human testis, FGFR3 expression is a feature of small clones of rarely dividing type A Spg which resemble "undifferentiated" Spg, including the spermatogonial stem cells.


Assuntos
Divisão Celular , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/biossíntese , Espermatogônias/metabolismo , Membrana Celular/metabolismo , Vesículas Citoplasmáticas/metabolismo , Proteínas de Ligação a DNA/biossíntese , Imunofluorescência , Humanos , Antígeno Ki-67/biossíntese , Masculino , Microscopia Confocal , Proteínas Nucleares/biossíntese , Proteínas Proto-Oncogênicas c-kit/análise , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/análise , Túbulos Seminíferos/citologia , Túbulos Seminíferos/metabolismo , Espermatogênese/fisiologia , Testículo/citologia , Testículo/metabolismo , Transativadores/biossíntese , Fatores de Transcrição/análise , Fatores de Transcrição/biossíntese
10.
Reproduction ; 143(1): 45-57, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22046055

RESUMO

It is unclear whether the distinct nuclear morphologies of human A(dark) (Ad) and A(pale) (Ap) spermatogonia are manifestations of different stages of germ cell development or phases of the mitotic cycle, or whether they may reflect still unknown molecular differences. According to the classical description by Clermont, human dark type A spermatogonium (Ad) may contain one, sometimes two or three nuclear 'vacuolar spaces' representing chromatin rarefaction zones. These structures were readily discerned in paraffin sections of human testis tissue during immunohistochemical and immunofluorescence analyses and thus represented robust morphological markers for our study. While a majority of the marker proteins tested did not discriminate between spermatogonia with and without chromatin rarefaction zones, doublesex- and mab-3-related transcription factor (DMRT1), tyrosine kinase receptor c-Kit/CD117 (KIT) and proliferation-associated antigen Ki-67 (KI-67) appeared to be restricted to subtypes which lacked the rarefaction zones. Conversely, exosome component 10 (EXOSC10) was found to accumulate within the rarefaction zones, which points to a possible role of this nuclear domain in RNA processing.


Assuntos
Espermatogônias/citologia , Espermatogônias/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Cromatina/metabolismo , Cromatina/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Exorribonucleases/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Microscopia Confocal , Microscopia de Fluorescência , Proteínas Nucleares/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Espermatogênese , Espermatogônias/classificação , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
11.
Curr Opin Cell Biol ; 15(3): 351-7, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12787779

RESUMO

The p53 tumour suppressor is crucial for the ability of the cell to either arrest cell cycle progression or activate apoptosis in response to stimuli that may impinge on genomic stability. p53 activation is controlled by mechanisms involving post-translational modifications, protein interactions and modulation of subcellular localisation. Recently, p53 was identified within nuclear bodies, particular subnuclear structures that can provide a 'platform' where interaction of p53 with specific cofactors is favoured. Modulation of recruitment/release of some of these components and modifications might be required for directing p53 toward one or another of its downstream response pathways.


Assuntos
Estruturas do Núcleo Celular/fisiologia , Reparo do DNA/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Humanos
12.
Nat Cell Biol ; 4(1): 1-10, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11740489

RESUMO

Transcriptional activity of p53, a central regulatory switch in a network controlling cell proliferation and apoptosis, is modulated by protein stability and post-translational modifications including phosphorylation and acetylation. Here we demonstrate that the human serine/threonine kinase homeodomain-interacting protein kinase-2 (HIPK2) colocalizes and interacts with p53 and CREB-binding protein (CBP) within promyelocytic leukaemia (PML) nuclear bodies. HIPK2 is activated by ultraviolet (UV) radiation and selectively phosphorylates p53 at Ser 46, thus facilitating the CBP-mediated acetylation of p53 at Lys 382, and promoting p53-dependent gene expression. Accordingly, the kinase function of HIPK2 mediates the increased expression of p53 target genes, which results in growth arrest and the enhancement of UV-induced apoptosis. Interference with HIPK2 expression by antisense oligonucleotides impairs UV-induced apoptosis. Our results imply that HIPK2 is a novel regulator of p53 effector functions involved in cell growth, proliferation and apoptosis.


Assuntos
Apoptose/genética , Proteínas de Transporte/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , Transativadores/genética , Fatores de Transcrição/genética , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Animais , Apoptose/efeitos da radiação , Proteína de Ligação a CREB , Proteínas de Transporte/metabolismo , Proteínas de Transporte/efeitos da radiação , Divisão Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Ativação Enzimática/efeitos da radiação , Genes Supressores de Tumor , Humanos , Camundongos , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Oligonucleotídeos Antissenso , Proteína da Leucemia Promielocítica , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/efeitos da radiação , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor , Raios Ultravioleta
13.
N Engl J Med ; 354(17): 1807-12, 2006 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-16641397

RESUMO

The reverse-transcriptase inhibitor lamivudine (Zeffix, GlaxoSmithKline) is often used to treat chronic infection with hepatitis B virus (HBV) until resistance develops. Treatment may then be switched to the reverse-transcriptase inhibitor adefovir (Hepsera, Gilead), which has a lower frequency of resistance. Here, we describe three cases of primary adefovir resistance that were sensitive to tenofovir (Viread, Gilead). All three cases involved a rare HBV variant with a valine at position 233 of the reverse-transcriptase domain instead of isoleucine (rtI233V), as in the wild-type virus. This HBV variant also displayed resistance to adefovir and sensitivity to tenofovir in vitro.


Assuntos
Adenina/análogos & derivados , Farmacorresistência Viral , Vírus da Hepatite B/genética , Hepatite B Crônica/tratamento farmacológico , Organofosfonatos/uso terapêutico , DNA Polimerase Dirigida por RNA/genética , Inibidores da Transcriptase Reversa/uso terapêutico , Adenina/farmacologia , Adenina/uso terapêutico , Sequência de Aminoácidos , Antivirais/farmacologia , Antivirais/uso terapêutico , DNA Viral/biossíntese , Farmacorresistência Viral/genética , Feminino , Hepatite B Crônica/virologia , Humanos , Lamivudina/farmacologia , Lamivudina/uso terapêutico , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação , Organofosfonatos/farmacologia , Inibidores da Transcriptase Reversa/farmacologia , Tenofovir , Viremia
14.
Nucleic Acids Res ; 34(1): 353-63, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16410615

RESUMO

The posttranscriptional regulatory element (PRE) is considered to enhance hepatitis B virus (HBV) gene expression by facilitating the nuclear export of intronless viral subgenomic RNAs. Its role in the RNA metabolism of the viral pregenomic RNA (pgRNA) is currently unknown. We identified a positively cis-acting splicing regulatory element (SRE-1) and present two lines of evidence for its functionality. Firstly, in a heterologous context SRE-1 functionally substitutes for a retroviral bidirectional exonic splicing enhancer (ESE). As expected, SRE-1 is a splicing enhancer also in its natural viral sequence context, since deletion of SRE-1 reduces splicing of pgRNA in cell culture experiments. Secondly, we show that stimulation of HBV RNA splicing by the splicing factor PSF was repressed by the PRE. Analysis of a variety of PSF mutants indicated that RNA-binding and protein-protein interaction were required to enhance splicing. In addition, we show that the PRE contributed to pgRNA stability, but has little influence on its nuclear export. Herein, we report for the first time that the PRE harbors splicing stimulating and inhibiting regulatory elements controlling processing of the viral pregenome. We discuss a model in which the regulation of pgRNA splicing depends on cellular factors interacting with the PRE.


Assuntos
Regulação Viral da Expressão Gênica , Vírus da Hepatite B/genética , Splicing de RNA , RNA Viral/química , RNA Viral/metabolismo , Sequências Reguladoras de Ácido Ribonucleico , Linhagem Celular Tumoral , Vírus da Hepatite B/metabolismo , Humanos , Fator de Processamento Associado a PTB , Proteínas de Ligação a RNA/metabolismo
15.
World J Gastroenterol ; 13(1): 91-103, 2007 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-17206758

RESUMO

The human hepatitis B virus (HBV) and the duck hepatitis B virus (DHBV) share several fundamental features. Both viruses have a partially double-stranded DNA genome that is replicated via a RNA intermediate and the coding open reading frames (ORFs) overlap extensively. In addition, the genomic and structural organization, as well as replication and biological characteristics, are very similar in both viruses. Most of the key features of hepadnaviral infection were first discovered in the DHBV model system and subsequently confirmed for HBV. There are, however, several differences between human HBV and DHBV. This review will focus on the molecular and cellular biology, evolution, and host adaptation of the avian hepatitis B viruses with particular emphasis on DHBV as a model system.


Assuntos
Avihepadnavirus/genética , Avihepadnavirus/fisiologia , Infecções por Hepadnaviridae/patologia , Sequência de Aminoácidos , Animais , Avihepadnavirus/crescimento & desenvolvimento , Avihepadnavirus/patogenicidade , DNA Viral/genética , Modelos Animais de Doenças , Patos , Infecções por Hepadnaviridae/tratamento farmacológico , Infecções por Hepadnaviridae/fisiopatologia , Vírus da Hepatite B do Pato/genética , Vírus da Hepatite B do Pato/crescimento & desenvolvimento , Vírus da Hepatite B do Pato/patogenicidade , Vírus da Hepatite B do Pato/fisiologia , Dados de Sequência Molecular , Morfogênese/fisiologia , Tropismo/fisiologia , Proteínas Virais/análise , Proteínas Virais/fisiologia , Vacinas Virais/genética , Vacinas Virais/uso terapêutico , Internalização do Vírus , Replicação Viral/fisiologia
16.
Cancer Res ; 63(23): 8271-7, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14678985

RESUMO

Homeodomain-interacting protein kinase 2 (HIPK2) is a serine/threonine kinase involved in transcriptional regulation and apoptosis. Here we demonstrate that HIPK2 regulates transforming growth factor (TGF) beta-induced c-Jun NH(2)-terminal kinase (JNK) activation and apoptosis. HIPK2 colocalizes with Daxx, a protein acting in TGF-beta-induced JNK activation and apoptosis, in promyelocytic leukemia (PML) nuclear bodies, and triggers PML-nuclear body disruption and release of Daxx. HIPK2 interacts in vitro and in vivo via its kinase domain with Daxx, and a fraction of Daxx coprecipitates with HIPK2 under physiological conditions. Moreover, overexpression of HIPK2 leads to Daxx phosphorylation, and ectopic expression of HIPK2 activates the JNK signaling pathway, which is enhanced by coexpression of Daxx. HIPK2 signals to JNK via a pathway using Daxx and the mitogen-activated protein kinase kinases MKK4/SEK1 and MKK7. Ectopic expression of HIPK2 and Daxx potentiates TGF-beta-induced apoptosis in human p53-deficient hepatocellular carcinoma cells. Finally, we demonstrate that knockdown of endogenous HIPK2 using RNA interference inhibits TGF-beta-induced JNK activation and apoptosis. Taken together, our findings indicate that HIPK2 participates in the TGF-beta signaling pathway leading to JNK activation and apoptosis.


Assuntos
Apoptose/fisiologia , Carcinoma Hepatocelular/enzimologia , Proteínas de Transporte/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias Hepáticas/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Proteínas Correpressoras , Ativação Enzimática , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Leucemia Promielocítica Aguda/metabolismo , Neoplasias Hepáticas/patologia , MAP Quinase Quinase 6 , MAP Quinase Quinase 7 , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
17.
Cancer Res ; 63(15): 4310-4, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12907596

RESUMO

Here we demonstrate that endogenous human homeodomain-interacting protein kinase (HIPK) 2 and the highly homologous kinase HIPK3 are found in a novel subnuclear domain, the HIPK domains. These are distinct from other subnuclear structures such as Cajal bodies and nucleoli and show only a partial colocalization with promyelocytic leukemia (PML) nuclear bodies (PML-NBs). A kinase inactive HIPK2 point mutant is localized in the nucleoplasm. The occurrence of HIPK domains in PML-/- fibroblasts reveals their independence from the PML protein. HIPK2 can be almost completely recruited to PML-NBs by the PML isoform PML IV, but not by PML-III. PML IV-mediated recruitment of HIPK2 does not rely on its kinase function and also occurs in PML-/- fibroblasts, showing that this PML isoform is sufficient for recruitment of HIPK2. Whereas the architecture of HIPK domains is PML independent, HIPK2-mediated enhancement of p53-dependent transcription, p53 serine 46 phosphorylation and the antiproliferative function of HIPK2 strictly rely on the presence of PML.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Transporte/genética , Ciclo Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Humanos , Osteossarcoma/genética , Osteossarcoma/metabolismo , Fosforilação , Proteína da Leucemia Promielocítica , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Frações Subcelulares/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese , Proteínas Supressoras de Tumor
18.
BMC Res Notes ; 9: 128, 2016 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-26921094

RESUMO

Recessive mutations in the MPV17 gene cause mitochondrial DNA depletion syndrome, a fatal infantile genetic liver disease in humans. Loss of function in mice leads to glomerulosclerosis and sensineural deafness accompanied with mitochondrial DNA depletion. Mutations in the yeast homolog Sym1, and in the zebra fish homolog tra cause interesting, but not obviously related phenotypes, although the human gene can complement the yeast Sym1 mutation. The MPV17 protein is a hydrophobic membrane protein of 176 amino acids and unknown function. Initially localised in murine peroxisomes, it was later reported to be a mitochondrial inner membrane protein in humans and in yeast. To resolve this contradiction we tested two new mouse monoclonal antibodies directed against the human MPV17 protein in Western blots and immunohistochemistry on human U2OS cells. One of these monoclonal antibodies showed specific reactivity to a protein of 20 kD absent in MPV17 negative mouse cells. Immunofluorescence studies revealed colocalisation with peroxisomal, endosomal and lysosomal markers, but not with mitochondria. This data reveal a novel connection between a possible peroxisomal/endosomal/lysosomal function and mitochondrial DNA depletion.


Assuntos
Anticorpos Monoclonais/química , Endossomos/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/genética , Proteínas Mitocondriais/genética , Peroxissomos/metabolismo , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/isolamento & purificação , Linhagem Celular Tumoral , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Endossomos/ultraestrutura , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Imunofluorescência , Expressão Gênica , Humanos , Lisossomos/ultraestrutura , Proteínas de Membrana/metabolismo , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/metabolismo , Mutação , Osteoblastos/metabolismo , Osteoblastos/ultraestrutura , Peroxissomos/ultraestrutura , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Elife ; 52016 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-27223324

RESUMO

PHF13 is a chromatin affiliated protein with a functional role in differentiation, cell division, DNA damage response and higher chromatin order. To gain insight into PHF13's ability to modulate these processes, we elucidate the mechanisms targeting PHF13 to chromatin, its genome wide localization and its molecular chromatin context. Size exclusion chromatography, mass spectrometry, X-ray crystallography and ChIP sequencing demonstrate that PHF13 binds chromatin in a multivalent fashion via direct interactions with H3K4me2/3 and DNA, and indirectly via interactions with PRC2 and RNA PolII. Furthermore, PHF13 depletion disrupted the interactions between PRC2, RNA PolII S5P, H3K4me3 and H3K27me3 and resulted in the up and down regulation of genes functionally enriched in transcriptional regulation, DNA binding, cell cycle, differentiation and chromatin organization. Together our findings argue that PHF13 is an H3K4me2/3 molecular reader and transcriptional co-regulator, affording it the ability to impact different chromatin processes.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Histonas/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Cromatografia em Gel , Cristalografia por Raios X , Regulação da Expressão Gênica , Humanos , Espectrometria de Massas , Camundongos , Ligação Proteica
20.
Oncogene ; 21(43): 6606-13, 2002 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-12242658

RESUMO

Two established activities of the multifunctional human hepatitis B virus X-protein are its transactivating and pro-apoptotic potential. We analysed whether X-proteins from other orthohepadnaviruses and the newly discovered avihepadnaviral X-proteins have similar functions as HBx. Previously, we have shown that HBx suppresses oncogenic transformation of primary rat embryo fibroblasts (REF) by induction of apoptosis. Using this system, we found that the wildtype X-proteins of woodchuck, ground squirrel, arctic squirrel and woolly monkey hepatitis B virus exhibit similar levels of pro-apoptotic activity as HBx, whereas mutants with carboxyterminal deletions were severely impaired in this activity. A strong correlation between the pro-apoptotic and transactivating abilities of the mammalian X-proteins was found. The newly discovered avihepadnaviral X-like proteins showed similar and Raf-MAPK pathway-dependent transactivating abilities and induced apoptosis in the REF-assay. Our data indicate that the transactivating and pro-apoptotic activities reside in the carboxyterminal half of orthohepadnaviral X and are conserved in avihepadnaviral X-proteins.


Assuntos
Apoptose , Hepadnaviridae/química , Transativadores/fisiologia , Ativação Transcricional , Proteínas Virais/fisiologia , Animais , Hepadnaviridae/genética , Ratos , Ratos Endogâmicos F344 , Proteínas Virais Reguladoras e Acessórias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA