Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 15(1): 76, 2024 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-38245531

RESUMO

The Hippo pathway's main effector, Yes-associated protein (YAP), plays a crucial role in tumorigenesis as a transcriptional coactivator. YAP's phosphorylation by core upstream components of the Hippo pathway, such as mammalian Ste20 kinase 1/2 (MST1/2), mitogen-activated protein kinase kinase kinase kinases (MAP4Ks), and their substrate, large tumor suppressor 1/2 (LATS1/2), influences YAP's subcellular localization, stability, and transcriptional activity. However, recent research suggests the existence of alternative pathways that phosphorylate YAP, independent of these core upstream Hippo pathway components, raising questions about additional means to inactivate YAP. In this study, we present evidence demonstrating that TSSK1B, a calcium/calmodulin-dependent protein kinase (CAMK) superfamily member, is a negative regulator of YAP, suppressing cellular proliferation and oncogenic transformation. Mechanistically, TSSK1B inhibits YAP through two distinct pathways. Firstly, the LKB1-TSSK1B axis directly phosphorylates YAP at Ser94, inhibiting the YAP-TEAD complex's formation and suppressing its target genes' expression. Secondly, the TSSK1B-LATS1/2 axis inhibits YAP via phosphorylation at Ser127. Our findings reveal the involvement of TSSK1B-mediated molecular mechanisms in the Hippo-YAP pathway, emphasizing the importance of multilevel regulation in critical cellular decision-making processes.


Assuntos
Via de Sinalização Hippo , Transdução de Sinais , Animais , Humanos , Fosforilação , Proteínas de Sinalização YAP , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transformação Celular Neoplásica/metabolismo , Proliferação de Células/fisiologia , Fosfoproteínas/metabolismo , Mamíferos
2.
Nucleic Acids Res ; 48(7): 3678-3691, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32123907

RESUMO

Genomic instability resulting from defective DNA damage responses or repair causes several abnormalities, including progressive cerebellar ataxia, for which the molecular mechanisms are not well understood. Here, we report a new murine model of cerebellar ataxia resulting from concomitant inactivation of POLB and ATM. POLB is one of key enzymes for the repair of damaged or chemically modified bases, including methylated cytosine, but selective inactivation of Polb during neurogenesis affects only a subpopulation of cortical interneurons despite the accumulation of DNA damage throughout the brain. However, dual inactivation of Polb and Atm resulted in ataxia without significant neuropathological defects in the cerebellum. ATM is a protein kinase that responds to DNA strand breaks, and mutations in ATM are responsible for Ataxia Telangiectasia, which is characterized by progressive cerebellar ataxia. In the cerebella of mice deficient for both Polb and Atm, the most downregulated gene was Itpr1, likely because of misregulated DNA methylation cycle. ITPR1 is known to mediate calcium homeostasis, and ITPR1 mutations result in genetic diseases with cerebellar ataxia. Our data suggest that dysregulation of ITPR1 in the cerebellum could be one of contributing factors to progressive ataxia observed in human genomic instability syndromes.


Assuntos
Ataxia Cerebelar/genética , Cerebelo/metabolismo , Metilação de DNA , DNA Polimerase beta/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Encéfalo/embriologia , Encéfalo/patologia , Cerebelo/anormalidades , Cerebelo/patologia , Citosina/metabolismo , Dano ao DNA , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Camundongos , Camundongos Knockout , Neurogênese/genética
3.
Cell Death Dis ; 9(11): 1079, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30348983

RESUMO

Remodeling and spacing factor 1 (RSF1), which is one of chromatin-remodeling factors, has been linked to the DNA damage response (DDR) and DNA repair. However, the biological consequence of RSF1 deficiency in DDR in vivo and its molecular mechanisms remain unknown. Because defective DDR is related to neuropathological phenotypes, we developed neural-specific Rsf1 knockout mice. Rsf1 deficiency did not result in any neuropathological abnormalities, but prevented neural apoptosis triggered by excessive DNA strand breaks during neurogenesis. Likewise, cell death was significantly reduced in RSF1 deficient human cell lines after DNA damage, and the global transcriptome of these cells revealed that the expressions of p53 downstream genes were significantly reduced upon DNA strand breaks. Inactivation of these genes resulted from decreased binding of p53/p300 complex and subsequent reduction of H3 acetylation at their promoters. Our data show that RSF1 is necessary for p53-dependent gene expression in response to DNA strand breaks via controlling the accessibility of p53/p300 complex to its target genes and contributes to the maintenance of cellular integrity.


Assuntos
Apoptose/genética , Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Dano ao DNA/genética , Proteínas Nucleares/genética , Transativadores/genética , Transcrição Gênica/genética , Proteína Supressora de Tumor p53/genética , Animais , Linhagem Celular Tumoral , DNA/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Feminino , Células HCT116 , Humanos , Masculino , Camundongos , Camundongos Knockout
4.
Viruses ; 7(9): 5133-44, 2015 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-26402693

RESUMO

The antiviral effect of a catalytic RNA-hydrolyzing antibody, 3D8 scFv, for intranasal administration against avian influenza virus (H1N1) was described. The recombinant 3D8 scFv protein prevented BALB/c mice against H1N1 influenza virus infection by degradation of the viral RNA genome through its intrinsic RNA-hydrolyzing activity. Intranasal administration of 3D8 scFv (50 µg/day) for five days prior to infection demonstrated an antiviral activity (70% survival) against H1N1 infection. The antiviral ability of 3D8 scFv to penetrate into epithelial cells from bronchial cavity via the respiratory mucosal layer was confirmed by immunohistochemistry, qRT-PCR, and histopathological examination. The antiviral activity of 3D8 scFv against H1N1 virus infection was not due to host immune cytokines or chemokines, but rather to direct antiviral RNA-hydrolyzing activity of 3D8 scFv against the viral RNA genome. Taken together, our results suggest that the RNase activity of 3D8 scFv, coupled with its ability to penetrate epithelial cells through the respiratory mucosal layer, directly prevents H1N1 virus infection in a mouse model system.


Assuntos
Anticorpos Catalíticos/administração & dosagem , Antivirais/administração & dosagem , Células Epiteliais/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Ribonucleases/administração & dosagem , Anticorpos de Cadeia Única/administração & dosagem , Administração Intranasal , Animais , Antivirais/farmacocinética , Hidrólise , Camundongos Endogâmicos BALB C , RNA Viral/metabolismo , Anticorpos de Cadeia Única/farmacocinética , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...