Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 591: 124-129, 2022 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-34815077

RESUMO

Mendelian susceptibility to mycobacterial disease (MSMD) is a rare monogenetic disease, which is characterized by susceptibility to some weakly virulent mycobacteria. Here, we explored the pathogenic genes and molecular mechanisms of MSMD patients. We recruited three patients diagnosed with MSMD from two families. Two novel mutations (c.1228A > G, p.K410E and c.2071A > G, p.M691V) in STAT1 gene were identified from two families. The translocation of K410E mutant STAT1 protein into nucleus was not affected. The binding ability between gamma-activating sequence (GAS) and K410E mutant STAT1 protein was significantly reduced, which will reduce the interaction between STAT1 protein with the promoters of target genes. The M691V mutant STAT1 protein cannot translocate into the nucleus after IFN-γ stimulation, which will affect the STAT1 protein form gamma-activating factors (GAF) and bind the GAS in the promoter region of downstream target genes. Taken together, our results showed that the mutation of K410E led to impaired binding of STAT1 to target DNA, and the mutation of M691V prevented the transport of STAT1 into the nucleus, which led to MSMD. Together, we identified two novel mutations (c.1228A > G, p.K410E and c.2071A > G, p.M691V) in STAT1 gene in MSMD patients, and deciphered the molecular mechanism of MSMD caused by STAT1 mutations.


Assuntos
Predisposição Genética para Doença , Mutação/genética , Infecções por Mycobacterium/genética , Infecções por Mycobacterium/microbiologia , Fator de Transcrição STAT1/genética , Sequência de Aminoácidos , Sequência de Bases , Núcleo Celular/metabolismo , DNA/metabolismo , Feminino , Células HEK293 , Células HeLa , Humanos , Masculino , Proteínas Mutantes/química , Proteínas Mutantes/genética , Linhagem , Ligação Proteica , Domínios Proteicos , Transporte Proteico , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/metabolismo , Frações Subcelulares/metabolismo
2.
Carbohydr Polym ; 268: 118214, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34127216

RESUMO

Phellinus baumii is used to treat inflammatory bowel disease (IBD) and gastroenteritis. In this study, a 46 kDa heteropolysaccharide SHPS-1 was isolated from fruiting bodies of P. baumii. SHPS-1 consisted of arabinose, mannose, glucose, and galactose at a molar ratio of 2.2:15.7:49.3:32.8. SHPS-1 had a backbone containing 1,3-linked ß-D-Glcp and 1,6-linked α-D-Galp residues, and Araf, Manp and Galp units were attached as oligosaccharidic side chains to the backbone at C-6 of some glucopyranoses. SHPS-1 decreased phosphorylation level of STAT-1 and expression levels of STAT-1 targeted genes such as iNOS and TNF-α in lipopolysaccharide-stimulated macrophage RAW 264.7 cells. Furthermore, SHPS-1 promoted the expression of IL-10 and macrophage mannose receptor CD 206, markers of tissue repairing macrophages. SHPS-1 alleviated ulcerative colitis in mice by decreasing pro-inflammatory genes and increasing anti-inflammatory and tissue repairing genes. Collectively, SHPS-1 polysaccharide from P. baumii had anti-inflammatory activity and can potentially treat IBD.


Assuntos
Anti-Inflamatórios/uso terapêutico , Basidiomycota/química , Colite Ulcerativa/tratamento farmacológico , Polissacarídeos Fúngicos/uso terapêutico , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/isolamento & purificação , Sequência de Carboidratos , Colite Ulcerativa/induzido quimicamente , Citocinas/metabolismo , Sulfato de Dextrana , Carpóforos/química , Polissacarídeos Fúngicos/química , Polissacarídeos Fúngicos/isolamento & purificação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Células RAW 264.7 , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
Nat Commun ; 12(1): 3931, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34168143

RESUMO

STAT1α is a key transcription factor driving pro-inflammatory responses in macrophages. We found that the interferon gamma (IFNγ)-regulated transcriptional program in macrophages is controlled by ADP-ribosylation (ADPRylation) of STAT1α, a post-translational modification resulting in the site-specific covalent attachment of ADP-ribose moieties. PARP-1, the major nuclear poly(ADP-ribose) polymerase (PARP), supports IFNγ-stimulated enhancer formation by regulating the genome-wide binding and IFNγ-dependent transcriptional activation of STAT1α. It does so by ADPRylating STAT1α on specific residues in its DNA-binding domain (DBD) and transcription activation (TA) domain. ADPRylation of the DBD controls STAT1α binding to its cognate DNA elements, whereas ADPRylation of the TA domain regulates enhancer activation by modulating STAT1α phosphorylation and p300 acetyltransferase activity. Loss of ADPRylation at either site leads to diminished IFNγ-dependent transcription and downstream pro-inflammatory responses. We conclude that PARP-1-mediated ADPRylation of STAT1α drives distinct enhancer activation mechanisms and is a critical regulator of inflammatory responses in macrophages.


Assuntos
ADP-Ribosilação , Interferon gama/metabolismo , Macrófagos/fisiologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Animais , Sítios de Ligação , DNA/metabolismo , Elementos Facilitadores Genéticos , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Fator de Transcrição STAT1/química , Ativação Transcricional
4.
Cell Res ; 31(2): 206-218, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32759968

RESUMO

Type I interferons (IFN-I) protect us from viral infections. Signal transducer and activator of transcription 2 (STAT2) is a key component of interferon-stimulated gene factor 3 (ISGF3), which drives gene expression in response to IFN-I. Using electron microscopy, we found that, in naive cells, U-STAT2, lacking the activating tyrosine phosphorylation, forms a heterodimer with U-STAT1 in an inactive, anti-parallel conformation. A novel phosphorylation of STAT2 on T404 promotes IFN-I signaling by disrupting the U-STAT1-U-STAT2 dimer, facilitating the tyrosine phosphorylation of STATs 1 and 2 and enhancing the DNA-binding ability of ISGF3. IKK-ε, activated by virus infection, phosphorylates T404 directly. Mice with a T-A mutation at the corresponding residue (T403) are highly susceptible to virus infections. We conclude that T404 phosphorylation drives a critical conformational switch that, by boosting the response to IFN-I in infected cells, enables a swift and efficient antiviral defense.


Assuntos
Herpes Simples/metabolismo , Multimerização Proteica/genética , Infecções por Rhabdoviridae/metabolismo , Fator de Transcrição STAT1/química , Fator de Transcrição STAT2/química , Transdução de Sinais/genética , Simplexvirus/metabolismo , Vírus da Estomatite Vesicular Indiana/metabolismo , Animais , Chlorocebus aethiops , Fibroblastos/metabolismo , Fibroblastos/virologia , Células HEK293 , Células HeLa , Herpes Simples/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação/genética , Conformação Proteica , Interferência de RNA , Infecções por Rhabdoviridae/virologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Células Vero
5.
PLoS One ; 15(7): e0228302, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32628668

RESUMO

Programmed death ligand 1 (PD-L1) has been recently shown to be a major obstacle to antiviral immunity by binding to its receptor programmed death 1 (PD-1) on specific IFN-γ producing T cells in chronic hepatitis B. Currently, IFN-α is widely used to treat hepatitis B virus (HBV) infection, but its antiviral effect vary greatly and the mechanism is not totally clear. We found that IFN-α/γ induced a marked increase of PD-L1 expression in hepatocytes. Signal and activators of transcription (Stat1) was then identified as a major transcription factor involved in IFN-α/γ-mediated PD-L1 elevation both in vitro and in mice. Blockage of the PD-L1/PD-1 interaction by a specific mAb greatly enhanced HBV-specific T cell activity by the gp96 adjuvanted therapeutic vaccine, and promoted HBV clearance in HBV transgenic mice. Our results demonstrate the IFN-α/γ-Stat1-PD-L1 axis plays an important role in mediating T cell hyporesponsiveness and inactivating liver-infiltrating T cells in the hepatic microenvironment. These data raise further potential interest in enhancing the anti-HBV efficacy of IFN-α and therapeutic vaccines.


Assuntos
Antígeno B7-H1/metabolismo , Vírus da Hepatite B/imunologia , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Fator de Transcrição STAT1/metabolismo , Linfócitos T/imunologia , Regulação para Cima/efeitos dos fármacos , Animais , Anticorpos Monoclonais/uso terapêutico , Antígeno B7-H1/química , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Sítios de Ligação , Linhagem Celular , Hepatite B/tratamento farmacológico , Hepatite B/veterinária , Antígenos de Superfície da Hepatite B/sangue , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Receptor de Morte Celular Programada 1/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição STAT1/química , Linfócitos T/metabolismo
6.
J Virol ; 94(17)2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32581091

RESUMO

Measles virus (MeV) is a highly immunotropic and contagious pathogen that can even diminish preexisting antibodies and remains a major cause of childhood morbidity and mortality worldwide despite the availability of effective vaccines. MeV is one of the most extensively studied viruses with respect to the mechanisms of JAK-STAT antagonism. Of the three proteins translated from the MeV P gene, P and V are essential for inactivation of this pathway. However, the lack of data from direct analyses of the underlying interactions means that the detailed molecular mechanism of antagonism remains unresolved. Here, we prepared recombinant MeV V protein, which is responsible for human JAK-STAT antagonism, and a panel of variants, enabling the biophysical characterization of V protein, including direct V/STAT1 and V/STAT2 interaction assays. Unambiguous direct interactions between the host and viral factors, in the absence of other factors such as Jak1 or Tyk2, were observed, and the dissociation constants were quantified for the first time. Our data indicate that interactions between the C-terminal region of V and STAT2 is 1 order of magnitude stronger than that of the N-terminal region of V and STAT1. We also clarified that these interactions are completely independent of each other. Moreover, results of size exclusion chromatography demonstrated that addition of MeV-V displaces STAT2-core, a rigid region of STAT2 lacking the N- and C-terminal domains, from preformed complexes of STAT2-core/IRF-associated domain (IRF9). These results provide a novel model whereby MeV-V can not only inhibit the STAT2/IRF9 interaction but also disrupt preassembled interferon-stimulated gene factor 3.IMPORTANCE To evade host immunity, many pathogenic viruses inactivate host Janus kinase signal transducer and activator of transcription (STAT) signaling pathways using diverse strategies. Measles virus utilizes P and V proteins to counteract this signaling pathway. Data derived largely from cell-based assays have indicated several amino acid residues of P and V proteins as important. However, biophysical properties of V protein or its direct interaction with STAT molecules using purified proteins have not been studied. We have developed novel molecular tools enabling us to identify a novel molecular mechanism for immune evasion whereby V protein disrupts critical immune complexes, providing a clear strategy by which measles virus can suppress interferon-mediated antiviral gene expression.


Assuntos
Fator Gênico 3 Estimulado por Interferon, Subunidade gama/química , Vírus do Sarampo/metabolismo , Fosfoproteínas/química , Fator de Transcrição STAT2/química , Proteínas Virais/química , Sítios de Ligação , Expressão Gênica , Humanos , Evasão da Resposta Imune , Imunidade Inata , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Janus Quinases/metabolismo , Vírus do Sarampo/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Domínios Proteicos , Domínios e Motivos de Interação entre Proteínas , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/metabolismo , Dedos de Zinco
7.
J Photochem Photobiol B ; 202: 111704, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31743829

RESUMO

Ultraviolet B (UVB) induces inflammation and causes skin aging. The signs of skin aging, such as wrinkles, discolored spots, loss of skin moisture, and disruption of the skin barrier, are mostly caused by inflammatory signaling among various skin layers. The cells on the outermost surface of the skin are keratinocytes; these cells protect the skin against environmental stress and play an important role in immunomodulation by secreting cytokines in response to environmental stress. In the present study, we found that UVB activates STAT1 to mediate inflammatory signaling, yet STAT1 (S272) and STAT (Y702) shows different responses against UVB exposure. Anhua drak tea is a post-fermented dark tea produced in Anhua and Xinhua country in Hunan province of China. Treatment with 2S,3R-6-methoxycarbonylgallocatechin (MCGE), an epigallocatechin gallate derivative isolated from black tea (Anhua dark tea), effectively suppresses STAT1 activation and inflammatory cytokines, and activates Nrf2 pathway to protect cells from reactive oxygen species production in UVB exposed keratinocyte cells (HaCaT). Interestingly, the effects of MCGE were independent on MAPK signaling pathway. Moreover, MCGE regulates inflammatory cytokines in monocyte-keratinocyte (THP-1, HaCaT) co-culture and macrophage differentiation models. These results suggest that MCGE potentially can be used as a photoprotective agent against UVB-induced inflammatory responses.


Assuntos
Catequina/análogos & derivados , Catequina/farmacologia , Protetores contra Radiação/farmacologia , Transdução de Sinais/efeitos dos fármacos , Chá/química , Raios Ultravioleta , Sítios de Ligação , Catequina/química , Catequina/isolamento & purificação , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos da radiação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Técnicas de Cocultura , Citocinas/metabolismo , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/efeitos da radiação , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Estrutura Terciária de Proteína , Protetores contra Radiação/química , Protetores contra Radiação/isolamento & purificação , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos da radiação , Chá/metabolismo
8.
Cell Rep ; 29(7): 1934-1945.e8, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31722208

RESUMO

To evade immunity, many viruses express interferon antagonists that target STAT transcription factors as a major component of pathogenesis. Because of a lack of direct structural data, these interfaces are poorly understood. We report the structural analysis of full-length STAT1 binding to an interferon antagonist of a human pathogenic virus. The interface revealed by transferred cross-saturation NMR is complex, involving multiple regions in both the viral and cellular proteins. Molecular mapping analysis, combined with biophysical characterization and in vitro/in vivo functional assays, indicates that the interface is significant in disease caused by a pathogenic field-strain lyssavirus, with critical roles for contacts between the STAT1 coiled-coil/DNA-binding domains and specific regions within the viral protein. These data elucidate the potentially complex nature of IFN antagonist/STAT interactions, and the spatial relationship of protein interfaces that mediate immune evasion and replication, providing insight into how viruses can regulate these essential functions via single multifunctional proteins.


Assuntos
Imunidade Inata , Lyssavirus , Fator de Transcrição STAT1 , Animais , Células COS , Chlorocebus aethiops , Feminino , Células HEK293 , Humanos , Lyssavirus/química , Lyssavirus/imunologia , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , Ressonância Magnética Nuclear Biomolecular , Domínios Proteicos , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia
9.
Proc Natl Acad Sci U S A ; 116(35): 17480-17491, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31413201

RESUMO

In contrast to the importance of type II interferon-γ (IFN-γ) in control of toxoplasmosis, the role of type I IFN is less clear. We demonstrate here that TgIST, a secreted effector previously implicated in blocking type II IFN-γ signaling, also blocked IFN-ß responses by inhibiting STAT1/STAT2-mediated transcription in infected cells. Consistent with a role for type I IFN in cell intrinsic control, ∆Tgist mutants were more susceptible to growth inhibition by murine and human macrophages activated with IFN-ß. Additionally, type I IFN was important for production of IFN-γ by natural killer (NK) cells and recruitment of inflammatory monocytes at the site of infection. Mice lacking type I IFN receptors (Ifnar1-/-) showed increased mortality following infection with wild-type parasites and decreased virulence of ∆Tgist parasites was restored in Ifnar1-/- mice. The findings highlight the importance of type I IFN in control of toxoplasmosis and illuminate a parasite mechanism to counteract the effects of both type I and II IFN-mediated host defenses.


Assuntos
Interferon Tipo I/metabolismo , Proteínas de Protozoários/metabolismo , Transdução de Sinais , Toxoplasma/fisiologia , Toxoplasmose/metabolismo , Toxoplasmose/parasitologia , Perfilação da Expressão Gênica , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Ligação Proteica , Multimerização Proteica , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/química , Fator de Transcrição STAT2/metabolismo , Toxoplasmose/imunologia
10.
Int J Cancer ; 145(6): 1635-1647, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31228268

RESUMO

Uterine serous carcinoma (USC) is a highly aggressive histological subtype of endometrial cancers harboring highly metastatic and chemoresistant features. Our previous study showed that STAT1 is highly expressed in USC and acts as a key molecule that is positively correlated with tumor progression, but it remains unclear whether STAT1 is relevant to the malicious chemorefractory nature of USC. In the present study, we investigated the regulatory role of STAT1 toward platinum-cytotoxicity in USC. STAT1 suppression sensitized USC cells to increase cisplatin-mediated apoptosis (p < 0.001). Furthermore, phosphorylation of STAT1 was prominently observed on serine-727 (pSTAT1-Ser727), but not on tyrosine-701, in the nucleus of USC cells treated with cisplatin. Mechanistically, the inhibition of pSTAT1-Ser727 by dominant-negative plasmid elevated cisplatin-mediated apoptosis by increasing intracellular accumulation of cisplatin through upregulation of CTR1 expression. TBB has an inhibitory effect on casein kinase 2 (CK2), which phosphorylate STAT1 at serine residues. Sequential treatment with TBB and cisplatin on USC cells greatly reduced nuclear pSTAT1-Ser727, enhanced intracellular accumulation of cisplatin, and subsequently increased apoptosis. Tumor load was significantly reduced by combination therapy of TBB and cisplatin in in vivo xenograft models (p < 0.001). Our results collectively suggest that pSTAT1-Ser727 may play a key role in platinum resistance as well as tumor progression in USC. Thus, targeting the STAT1 pathway via CK2 inhibitor can be a novel method for attenuating the chemorefractory nature of USC.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Cistadenocarcinoma Seroso/tratamento farmacológico , Fator de Transcrição STAT1/metabolismo , Serina/metabolismo , Neoplasias Uterinas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimioterapia Adjuvante , Cisplatino/farmacologia , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Xenoenxertos , Humanos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Fosforilação , Fator de Transcrição STAT1/química , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
11.
Fish Shellfish Immunol ; 87: 386-394, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30703549

RESUMO

Signal transducer and activator of transcription 1 (STAT1) plays an important role in the Janus kinase (JAK)-STAT signaling of human and mammals; however, the mechanism of STAT1 in innate immune activation of teleost fishes remains largely unknown. In this study, two STAT1 homologues (bcSTAT1a and bcSTAT1b) of black carp (Mylopharyngodon piceus) have been cloned and characterized. Both bcSTAT1a and bcSTAT1b transcription in host cells was obviously increased in response to the stimulation of poly (I:C), lipopolysaccharide (LPS), grass carp reovirus (GCRV) and interferon (IFN); however, the increase rate of bcSTAT1b transcription post stimulation was obviously higher than that of bcSTAT1a. bcSTAT1a and bcSTAT1b were distributed in both cytoplasm and nucleus in the immunofluorescence staining assay. Self-association of bcSTAT1a and bcSTAT1b, and the interaction between bcSTAT1a and bcSTAT1b have been detected through co-immunoprecipitation (co-IP) assay; and the data of native polyacrylamide gel electrophoresis (PAGE) implied that bcSTAT1a and bcSTAT1b might form homodimer and heterodimer in vivo like their mammalian counterparts. Both bcSTAT1a and bcSTAT1b presented IFN-inducing ability in report assay, and both bcSTAT1a and bcSTAT1b showed antiviral activities against GCRV in EPC cells. Our data support the conclusion that both bcSTAT1a and bcSTAT1b play important roles in host antiviral innate immune activation initiated by GCRV.


Assuntos
Carpas/genética , Carpas/imunologia , Doenças dos Peixes/imunologia , Expressão Gênica/imunologia , Imunidade Inata/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Sequência de Aminoácidos , Animais , Proteínas de Peixes/química , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Perfilação da Expressão Gênica/veterinária , Filogenia , Fator de Transcrição STAT1/química , Alinhamento de Sequência/veterinária
12.
Fish Shellfish Immunol ; 86: 956-964, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30590158

RESUMO

Signal transducer and activator of transcription 1 (STAT1) is one of critical signal transduction proteins of interferon (IFN) pathway and the structure and function of this protein have been well identified in mammals, but the information about the STAT1 is still limited in teleost fishes. In the present study, the full-length cDNA sequence of STAT1 (AjSTAT1) in Japanese eel (Anguilla japonica) was identified and characterized. Multiple alignment of the amino acid sequence showed that the AjSTAT1 protein has the typical conserved domains including the amino-terminal, coiled-coil, DNA-binding, linker, Src homology 2 (SH2), transcriptional activation domains (TAD). Quantitative real-time polymerase chain reaction (qRT-PCR) analysis revealed a broad expression for AjSTAT1 in a wide range of tissues, with the predominant expression in liver, followed by the spleen, intestine, gills, skin, kidney, and the very low expression in heart and muscle. The AjSTAT1 expressions in liver, spleen and kidney were significantly induced following injection with LPS, the viral mimic poly I:C, and Aeromonas hydrophila infection. In vitro, the AjSTAT1 transcripts of Japanese eel liver cells were significantly enhanced by the treatment of poly I:C or the stimulation of the high concentration of Aeromonas hydrophila (1 × 107 cfu/mL and 1 × 108 cfu/mL). Subcellular localization showed that in the natural state AjSTAT1was uniformly distributed in the cytoplasm, but AjSTAT1 was found to aggregated in the cytoplasm as well as partly in the nucleus after the stimulation of LPS and poly I:C. These results collectively suggested AjSTAT1 is an important transcription factor possibly involved in Japanese eel defense against viral and bacterial infection.


Assuntos
Anguilla/metabolismo , Proteínas de Peixes/genética , Fator de Transcrição STAT1/metabolismo , Aeromonas hydrophila , Sequência de Aminoácidos , Animais , DNA Complementar/genética , DNA Complementar/metabolismo , Doenças dos Peixes/microbiologia , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Negativas/veterinária , Lipopolissacarídeos/farmacologia , Poli I-C/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/genética , Análise de Sequência de DNA
13.
Nature ; 562(7728): 538-544, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30323286

RESUMO

The transcriptional co-activator p300 is a histone acetyltransferase (HAT) that is typically recruited to transcriptional enhancers and regulates gene expression by acetylating chromatin. Here we show that the activation of p300 directly depends on the activation and oligomerization status of transcription factor ligands. Using two model transcription factors, IRF3 and STAT1, we demonstrate that transcription factor dimerization enables the trans-autoacetylation of p300 in a highly conserved and intrinsically disordered autoinhibitory lysine-rich loop, resulting in p300 activation. We describe a crystal structure of p300 in which the autoinhibitory loop invades the active site of a neighbouring HAT domain, revealing a snapshot of a trans-autoacetylation reaction intermediate. Substrate access to the active site involves the rearrangement of an autoinhibitory RING domain. Our data explain how cellular signalling and the activation and dimerization of transcription factors control the activation of p300, and therefore explain why gene transcription is associated with chromatin acetylation.


Assuntos
Multimerização Proteica , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Fatores de Transcrição de p300-CBP/química , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Domínio Catalítico , Cromatina/química , Cromatina/metabolismo , Cristalografia por Raios X , Ativação Enzimática , Humanos , Fator Regulador 3 de Interferon/química , Fator Regulador 3 de Interferon/metabolismo , Ligantes , Lisina/química , Lisina/metabolismo , Modelos Moleculares , Domínios Proteicos , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/metabolismo , Transcrição Gênica
14.
Front Immunol ; 9: 1135, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29892288

RESUMO

Interferon (IFN)-I and IFN-II both induce IFN-stimulated gene (ISG) expression through Janus kinase (JAK)-dependent phosphorylation of signal transducer and activator of transcription (STAT) 1 and STAT2. STAT1 homodimers, known as γ-activated factor (GAF), activate transcription in response to all types of IFNs by direct binding to IFN-II activation site (γ-activated sequence)-containing genes. Association of interferon regulatory factor (IRF) 9 with STAT1-STAT2 heterodimers [known as interferon-stimulated gene factor 3 (ISGF3)] or with STAT2 homodimers (STAT2/IRF9) in response to IFN-I, redirects these complexes to a distinct group of target genes harboring the interferon-stimulated response element (ISRE). Similarly, IRF1 regulates expression of ISGs in response to IFN-I and IFN-II by directly binding the ISRE or IRF-responsive element. In addition, evidence is accumulating for an IFN-independent and -dependent role of unphosphorylated STAT1 and STAT2, with or without IRF9, and IRF1 in basal as well as long-term ISG expression. This review provides insight into the existence of an intracellular amplifier circuit regulating ISG expression and controlling long-term cellular responsiveness to IFN-I and IFN-II. The exact timely steps that take place during IFN-activated feedback regulation and the control of ISG transcription and long-term cellular responsiveness to IFN-I and IFN-II is currently not clear. Based on existing literature and our novel data, we predict the existence of a multifaceted intracellular amplifier circuit that depends on unphosphorylated and phosphorylated ISGF3 and GAF complexes and IRF1. In a combinatorial and timely fashion, these complexes mediate prolonged ISG expression and control cellular responsiveness to IFN-I and IFN-II. This proposed intracellular amplifier circuit also provides a molecular explanation for the existing overlap between IFN-I and IFN-II activated ISG expression.


Assuntos
Retroalimentação Fisiológica , Fatores Reguladores de Interferon/genética , Fator Gênico 3 Estimulado por Interferon/genética , Interferons/metabolismo , Animais , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Mutação em Linhagem Germinativa , Humanos , Fatores Reguladores de Interferon/metabolismo , Fator Gênico 3 Estimulado por Interferon/metabolismo , Mutação , Ligação Proteica , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/química , Fator de Transcrição STAT2/metabolismo
15.
J Biomol Struct Dyn ; 36(3): 767-778, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28166455

RESUMO

Oct4 is a master regulator of the induction and maintenance of cellular pluripotency, and has crucial roles in early stages of differentiation. It is the only factor that cannot be substituted by other members of the same protein family to induce pluripotency. However, although Oct4 nuclear transport and delivery to target DNA are critical events for reprogramming to pluripotency, little is known about the molecular mechanism. Oct4 is imported to the nucleus by the classical nuclear transport mechanism, which requires importin α as an adaptor to bind the nuclear localization signal (NLS). Although there are structures of complexes of the NLS of transcription factors (TFs) in complex with importin α, there are no structures available for complexes involving intact TFs. We have therefore modeled the structure of the complex of the whole Oct4 POU domain and importin α2 using protein-protein docking and molecular dynamics. The model explains how the Ebola virus VP24 protein has a negative effect on the nuclear import of STAT1 by importin α but not on Oct4, and how Nup 50 facilitates cargo release from importin α. The model demonstrates the structural differences between the Oct4 importin α bound and DNA bound crystal states. We propose that the 'expanded linker' between the two DNA-binding domains of Oct4 is an intrinsically disordered region and that its conformational changes have a key role in the recognition/binding to both DNA and importin α. Moreover, we propose that this structural change enables efficient delivery to DNA after release from importin α.


Assuntos
Doença pelo Vírus Ebola/genética , Fator 3 de Transcrição de Octâmero/química , Proteínas Virais/química , alfa Carioferinas/química , Transporte Ativo do Núcleo Celular/genética , Sítios de Ligação , Núcleo Celular/química , Núcleo Celular/genética , Reprogramação Celular/genética , Ebolavirus/química , Ebolavirus/genética , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/virologia , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Fator 3 de Transcrição de Octâmero/genética , Ligação Proteica , Mapas de Interação de Proteínas , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/genética , Proteínas Virais/genética , alfa Carioferinas/genética
16.
J Biol Chem ; 292(48): 19752-19766, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28978648

RESUMO

Sendai virus (SeV), which causes respiratory diseases in rodents, possesses the C protein that blocks the signal transduction of interferon (IFN), thereby escaping from host innate immunity. We previously demonstrated by using protein crystallography that two molecules of Y3 (the C-terminal half of the C protein) can bind to the homodimer of the N-terminal domain of STAT1 (STAT1ND), elucidating the mechanism of inhibition of IFN-γ signal transduction. SeV C protein also blocks the signal transduction of IFN-α/ß by inhibiting the phosphorylation of STAT1 and STAT2, although the mechanism for the inhibition is unclear. Therefore, we sought to elucidate the mechanism of inhibition of the IFN signal transduction via STAT1 and STAT2. Small angle X-ray scattering analysis indicated that STAT1ND associates with the N-terminal domain of STAT2 (STAT2ND) with the help of a Gly-rich linker. We generated a linker-less recombinant protein possessing a STAT1ND:STAT2ND heterodimeric structure via an artificial disulfide bond. Analytical size-exclusion chromatography and surface plasmon resonance revealed that one molecule of Y3 can associate with a linker-less recombinant protein. We propose that one molecule of C protein associates with the STAT1:STAT2 heterodimer, inducing a conformational change to an antiparallel form, which is easily dephosphorylated. This suggests that association of C protein with the STAT1ND:STAT2ND heterodimer is an important factor to block the IFN-α/ß signal transduction.


Assuntos
Interferon Tipo I/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Vírus Sendai/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo , Linhagem Celular , Cristalografia por Raios X , Dimerização , Humanos , Fosforilação , Conformação Proteica , Fator de Transcrição STAT1/química , Fator de Transcrição STAT2/química
17.
Fish Shellfish Immunol ; 69: 128-141, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28818616

RESUMO

Signal transducers and activators of transcription 1 (STAT1) is critically involved in mediating cytokine-driven signaling, and triggers the transcription of target genes to activate cellular functions. Although the structural and functional aspects of STAT members have been well described in mammals, only limited information is available for the STAT genes in teleost fishes. In the present study, two variants of STAT1 genes (RbSTAT1 and RbSTAT1L) were identified from rock bream and characterized at the cDNA and genomic sequence levels. RbSTAT1 and RbSTAT1L were found to share a common domain architecture with mammalian STAT1. Phylogenetic analysis revealed that RbSTAT1 shows a common evolutionary trajectory with other STAT1 counterparts, whereas RbSTAT1L showed a separate path, implying that it could be a novel member of the STAT family. The genomic organizations of RbSTAT1 and RbSTAT1L illustrated a similar exon-intron pattern with 23 exons in the coding sequence. Transcription factor-binding sites, which are mostly involved in the regulation of immune responses, were predicted at the putative promoter regions of the RbSTAT1 and RbSTAT1L genes. SYBR Green qPCR analysis revealed the ubiquitous expression of RbSTAT1 and RbSTAT1L transcripts in different fish tissues with the highest level observed in peripheral blood cells. Significantly modulated transcripts were noted upon viral (rock bream iridovirus [RBIV]), bacterial (Edwardsiella tarda and Streptococcus iniae), and pathogen-associated molecular pattern (lipopolysaccharide and poly I:C) stimulations. The WST-1 cell viability assay affirmed the potential antiviral capacity of RbSTAT1 and RbSTAT1L against RBIV. A possible role of RbSTAT1 and RbSTAT1L in the wound healing process was revealed according to their modulated expression in injured fish. In addition, the transcriptional regulation of RbSTAT1 and RbSTAT1L was analyzed by qPCR following stimulation with rock bream interleukin-10. Taken together, these findings suggest that the STAT1-mediated Janus kinase/STAT pathway might at least in part be involved in the regulatory mechanisms underlying the immune defensive roles against microbial pathogens and the wound healing process.


Assuntos
Doenças dos Peixes/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Sequência de Aminoácidos , Animais , Infecções por Vírus de DNA/imunologia , Edwardsiella tarda/fisiologia , Infecções por Enterobacteriaceae/imunologia , Proteínas de Peixes/química , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Perfilação da Expressão Gênica , Iridoviridae/fisiologia , Lipopolissacarídeos/farmacologia , Perciformes , Filogenia , Poli I-C/farmacologia , Distribuição Aleatória , Fator de Transcrição STAT1/química , Alinhamento de Sequência/veterinária , Infecções Estreptocócicas/imunologia , Streptococcus iniae/fisiologia
18.
J Virol ; 91(12)2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28404846

RESUMO

The high mutation rates of the influenza virus genome facilitate the generation of viral escape mutants, rendering vaccines and drugs against influenza virus-encoded targets potentially ineffective. Therefore, we identified host cell determinants dispensable for the host but crucial for virus replication, with the goal of preventing viral escape and finding effective antivirals. To identify these host factors, we screened 2,732 human genes using RNA interference and focused on one of the identified host factors, the double plant homeodomain fingers 2 (DPF2/REQ) gene, for this study. We found that knockdown of DPF2 in cells infected with influenza virus resulted in decreased expression of viral proteins and RNA. Furthermore, production of progeny virus was reduced by two logs in the multiple-cycle growth kinetics assay. We also found that DPF2 was involved in the replication of seasonal influenza A and B viruses. Because DPF2 plays a crucial role in the noncanonical NF-κB pathway, which negatively regulates type I interferon (IFN) induction, we examined the relationship between DPF2 and IFN responses during viral infection. The results showed that knockdown of DPF2 resulted in increased expression of IFN-ß and induced phosphorylation of STAT1 in infected cells. In addition, high levels of several cytokines/chemokines (interleukin-8 [IL-8], IP-10, and IL-6) and antiviral proteins (MxA and ISG56) were produced by DPF2 knockdown cells. In conclusion, we identified a novel host factor, DPF2, that is required for influenza virus to evade the host immune response and that may serve as a potential antiviral target.IMPORTANCE Influenza virus is responsible for seasonal epidemics and occasional pandemics and is an ongoing threat to public health worldwide. Influenza virus relies heavily on cellular factors to complete its life cycle. Here we identified a novel host factor, DPF2, which is involved in influenza virus infection. Our results showed that DPF2 plays a crucial role in the replication and propagation of influenza virus. DPF2 functions in the noncanonical NF-κB pathway, which negatively regulates type I IFN induction. Thus, we investigated the relationship between the IFN response and DPF2 in influenza virus infection. Upon influenza virus infection, DPF2 dysregulated IFN-ß induction and expression of cytokines/chemokines and antiviral proteins. This study provides evidence that influenza virus utilizes DPF2 to escape host innate immunity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Vírus da Influenza A/fisiologia , Interferon beta/biossíntese , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Linhagem Celular , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/imunologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Humanos , Imunidade Inata , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/imunologia , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Interferon beta/genética , Interleucina-6/biossíntese , Interleucina-6/imunologia , Interleucina-8/biossíntese , Interleucina-8/imunologia , Cinética , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/imunologia , NF-kappa B/imunologia , NF-kappa B/metabolismo , Interferência de RNA , Proteínas de Ligação a RNA , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Replicação Viral
19.
Oncotarget ; 8(20): 33779-33795, 2017 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-28422713

RESUMO

Inhibition of oncogenic transcriptional programs is a promising therapeutic strategy. A substituted tricyclic benzimidazole, SEL120-34A, is a novel inhibitor of Cyclin-dependent kinase 8 (CDK8), which regulates transcription by associating with the Mediator complex. X-ray crystallography has shown SEL120-34A to be a type I inhibitor forming halogen bonds with the protein's hinge region and hydrophobic complementarities within its front pocket. SEL120-34A inhibits phosphorylation of STAT1 S727 and STAT5 S726 in cancer cells in vitro. Consistently, regulation of STATs- and NUP98-HOXA9- dependent transcription has been observed as a dominant mechanism of action in vivo. Treatment with the compound resulted in a differential efficacy on AML cells with elevated STAT5 S726 levels and stem cell characteristics. In contrast, resistant cells were negative for activated STAT5 and revealed lineage commitment. In vivo efficacy in xenotransplanted AML models correlated with significant repression of STAT5 S726. Favorable pharmacokinetics, confirmed safety and in vivo efficacy provide a rationale for the further clinical development of SEL120-34A as a personalized therapeutic approach in AML.


Assuntos
Antineoplásicos/farmacologia , Quinase 8 Dependente de Ciclina/antagonistas & inibidores , Leucemia Mieloide Aguda/metabolismo , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Quinase 8 Dependente de Ciclina/química , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Modelos Moleculares , Conformação Molecular , Fosforilação/efeitos dos fármacos , Ligação Proteica , Inibidores de Proteínas Quinases/química , Fator de Transcrição STAT1/química , Fator de Transcrição STAT5/química , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Virology ; 506: 1-6, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28282567

RESUMO

The genetic basis for a dramatically increased virus susceptibility phenotype of MHC-II knockout mice acquired during routine maintenance of the mouse strain was determined. Segregation of the susceptibility allele from the defective MHC-II locus combined with sequence capture and sequencing showed that a Y37L substitution in STAT1 accounted for high flavivirus susceptibility of a newly derived mouse strain, designated Tuara. Interestingly, the mutation in STAT1 gene gave only partial inactivation of the type I interferon antiviral pathway. Accordingly, merely a relatively small impairment of interferon α/ß signalling is sufficient to overcome the ability of the host to control the infection.


Assuntos
Infecções por Flaviviridae/virologia , Flaviviridae/fisiologia , Fator de Transcrição STAT1/imunologia , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Flaviviridae/genética , Infecções por Flaviviridae/genética , Infecções por Flaviviridae/imunologia , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...