Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-37773612

RESUMO

Neutrophils are essential for host defense against Staphylococcus aureus (S. aureus). The neuro-repellent, SLIT2, potently inhibits neutrophil chemotaxis, and might, therefore, be expected to impair antibacterial responses. We report here that, unexpectedly, neutrophils exposed to the N-terminal SLIT2 (N-SLIT2) fragment kill extracellular S. aureus more efficiently. N-SLIT2 amplifies reactive oxygen species production in response to the bacteria by activating p38 mitogen-activated protein kinase that in turn phosphorylates NCF1, an essential subunit of the NADPH oxidase complex. N-SLIT2 also enhances the exocytosis of neutrophil secondary granules. In a murine model of S. aureus skin and soft tissue infection (SSTI), local SLIT2 levels fall initially but increase subsequently, peaking at 3 days after infection. Of note, the neutralization of endogenous SLIT2 worsens SSTI. Temporal fluctuations in local SLIT2 levels may promote neutrophil recruitment and retention at the infection site and hasten bacterial clearance by augmenting neutrophil oxidative burst and degranulation. Collectively, these actions of SLIT2 coordinate innate immune responses to limit susceptibility to S. aureus.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Animais , Humanos , Camundongos , Quimiotaxia de Leucócito , Imunidade Inata , Neutrófilos , Infecções Estafilocócicas/microbiologia
2.
Life Sci Alliance ; 6(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37311584

RESUMO

SLIT/ROBO signaling impacts many aspects of tissue development and homeostasis, in part, through the regulation of cell growth and proliferation. Recent studies have also linked SLIT/ROBO signaling to the regulation of diverse phagocyte functions. However, the mechanisms by which SLIT/ROBO signaling acts at the nexus of cellular growth control and innate immunity remain enigmatic. Here, we show that SLIT2-mediated activation of ROBO1 leads to inhibition of mTORC1 kinase activity in macrophages, leading to dephosphorylation of its downstream targets, including transcription factor EB and ULK1. Consequently, SLIT2 augments lysosome biogenesis, potently induces autophagy, and robustly promotes the killing of bacteria within phagosomes. Concordant with these results, we demonstrate decreased lysosomal content and accumulated peroxisomes in the spinal cords of embryos from Robo1 -/- , Robo2 -/- double knockout mice. We also show that impediment of auto/paracrine SLIT-ROBO signaling axis in cancer cells leads to hyperactivation of mTORC1 and inhibition of autophagy. Together, these findings elucidate a central role of chemorepellent SLIT2 in the regulation of mTORC1 activity with important implications for innate immunity and cancer cell survival.


Assuntos
Proteínas do Tecido Nervoso , Receptores Imunológicos , Animais , Camundongos , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Lisossomos , Bactérias , Alvo Mecanístico do Complexo 1 de Rapamicina
3.
Nat Commun ; 9(1): 3199, 2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-30097581

RESUMO

The nucleus represents a cellular compartment where the discrimination of self from non-self nucleic acids is vital. While emerging evidence establishes a nuclear non-self DNA sensing paradigm, the nuclear sensing of non-self RNA, such as that from nuclear-replicating RNA viruses, remains unexplored. Here, we report the identification of nuclear-resident RIG-I actively involved in nuclear viral RNA sensing. The nuclear RIG-I, along with its cytoplasmic counterpart, senses influenza A virus (IAV) nuclear replication leading to a cooperative induction of type I interferon response. Its activation signals through the canonical signaling axis and establishes an effective antiviral state restricting IAV replication. The exclusive signaling specificity conferred by nuclear RIG-I is reinforced by its inability to sense cytoplasmic-replicating Sendai virus and appreciable sensing of hepatitis B virus pregenomic RNA in the nucleus. These results refine the RNA sensing paradigm for nuclear-replicating viruses and reveal a previously unrecognized subcellular milieu for RIG-I-like receptor sensing.


Assuntos
Antivirais/farmacologia , Núcleo Celular/metabolismo , Proteína DEAD-box 58/metabolismo , Imunidade Inata , Replicação Viral/fisiologia , Células A549 , Animais , Compartimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Cães , Células HEK293 , Humanos , Vírus da Influenza A/efeitos dos fármacos , Células Madin Darby de Rim Canino , Ligação Proteica/efeitos dos fármacos , RNA Viral/metabolismo , Receptores Imunológicos , Ribonucleoproteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
FEBS Lett ; 592(13): 2323-2333, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29885262

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates lipid metabolism. A mutual interplay of lipid homeostasis and innate immune system has been increasingly recognized. We, therefore, studied the effect of PCSK9 on interferon (IFN) ß expression. We show that PCSK9 decreases IFNß promoter/enhancer activity, mRNA and protein levels, and its downstream 2',5'-oligoadenylate synthetase-1 mRNA level. ProPCSK9, but not the cleaved PCSK9, down-regulates IFNß promoter/enhancer activity. Moreover, PCSK9 decreases IFNß promoter/enhancer activity through the positive regulatory domain IV region where the activating transcription factor-2 (ATF-2)/c-Jun heterodimer binds. Mechanistically, we demonstrate an interaction between PCSK9 and ATF-2, which reduces ATF-2/c-Jun dimerization and ATF-2/c-Jun binding to the IFNß enhancer. This novel function of PCSK9 should have important implications in optimizing the clinical use of PCSK9 inhibitors.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Interferon beta/genética , Pró-Proteína Convertase 9/fisiologia , Células Cultivadas , Regulação para Baixo/genética , Células HEK293 , Humanos , Interferon beta/metabolismo , Metabolismo dos Lipídeos/genética , Regiões Promotoras Genéticas , Pró-Proteína Convertase 9/metabolismo , Ligação Proteica
5.
Virology ; 520: 94-102, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29803738

RESUMO

HBV HCV co-infection leads to more severe liver diseases including liver cancer than mono-infections. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumor suppressor, inhibits sterol regulatory element binding protein-1 (SREBP-1). In this study, we characterized the effect of the PTEN - SREBP-1 pathway on HBV HCV co-replication in a cellular model. We found that HBV and HCV can co-replicate in Huh-7 cells with no interference. Overexpression of PTEN inhibits, whereas PTEN knockdown enhances, HBV replication as well as HBV and HCV co-replication. Knocking down SREBP-1 decreases HBV replication in an HBx-dependent manner. SREBP-1 knockdown also decreases HCV replication. PTEN knockdown is concomitant with increased nuclear SREBP-1 levels. PTEN and SREBP-1 double knockdown results in intermediate levels of HBV and HCV replication in mono- and co-replication scenarios. Taken together, we demonstrated, for the first time, that the PTEN - SREBP-1 pathway can regulate HBV HCV co-replication.


Assuntos
Hepacivirus/fisiologia , Vírus da Hepatite B/fisiologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Replicação Viral , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Replicação do DNA , Técnicas de Silenciamento de Genes , Hepatócitos/virologia , Humanos , Neoplasias Hepáticas/virologia , PTEN Fosfo-Hidrolase/deficiência , Proteína de Ligação a Elemento Regulador de Esterol 1/deficiência , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
6.
Biochem Biophys Res Commun ; 496(4): 1229-1235, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29397939

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secretory serine protease mainly expressed in liver. Although PCSK9 has been shown to inhibit hepatitis C virus (HCV) entry and replication, whether HCV regulates PCSK9 transcription has not been well studied. PCSK9 promoter activity is modulated by numerous transcription factors including sterol-regulatory element binding protein (SREBP)-1a, -1c, -2, hepatocyte nuclear factor-1 (HNF-1), and forkhead box O3 (FoxO3). Since they are differently regulated by HCV, we studied the effects of these transcription factors on PCSK9 promoter activity in the context of HCV infection and replication. We demonstrated that PCSK9 promoter activity was up-regulated after HCV infection and in HCV genomic replicon cells. We also studied the effects of HCV proteins on the PCSK9 promoter activity. While HCV structural proteins core, E1, and E2 had no effect, NS2, NS3, NS3-4A, NS5A and NS5B enhanced, and p7 and NS4B decreased PCSK9 promoter activity. Furthermore, we showed that transcription factors SREBP-1c, HNF-1α and specificity protein 1 increased PCSK9 promoter activity in HCV replicon cells, whereas SREBP-1a, HNF-1ß and FoxO3 had an inhibitory effect. These results demonstrated the molecular mechanisms of how HCV modulates PCSK9 promoter activity and advanced our understanding on the mutual interactions between HCV and PCSK9.


Assuntos
Regulação Enzimológica da Expressão Gênica/genética , Regulação Viral da Expressão Gênica/genética , Hepacivirus/genética , Hepatócitos/enzimologia , Hepatócitos/virologia , Regiões Promotoras Genéticas/genética , Pró-Proteína Convertase 9/genética , Linhagem Celular , Ativação Enzimática , Humanos , Ligação Proteica
7.
J Gen Virol ; 99(1): 44-61, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29235977

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease actively involved in regulating lipid homeostasis. Although PCSK9 has been shown to inhibit hepatitis C virus (HCV) entry and replication, the underlying mechanisms have not been thoroughly characterized. Moreover, whether PCSK9 regulates HCV translation and assembly/secretion has not been determined. We therefore further studied the effects of PCSK9 on the HCV life cycle. We showed that PCSK9 did not affect HCV translation or assembly/secretion. Overexpression of PCSK9 inhibited HCV replication in HCV genomic replicon cells in a dose-dependent manner and after cell culture-derived HCV (HCVcc) infection. Knocking down PCSK9 increased HCV replication. The gain-of-function (D374Y) or loss-of-function (Δaa. 31-52) PCSK9 mutants for low-density lipoprotein receptor (LDLR) degradation had no effect on HCV replication, suggesting that HCV replication inhibition by PCSK9 was not due to LDLR degradation. The uncleaved ProPCSK9, but not cleaved PCSK9, down-regulated HCV replication, suggesting that the auto-cleavage of PCSK9 affected HCV replication. We also found that PCSK9 interacted with NS5A through NS5A aa. 95-215, and this region played an important role in NS5A dimerization, NS5A-RNA binding and was essential for HCV replication. More importantly, NS5A dimerization and NS5A-RNA binding were suppressed by PCSK9 upon interaction. These results suggested that PCSK9 inhibited HCV replication through interaction with NS5A. Our study should help optimize anti-HCV treatment regimen in patients with abnormal lipid profiles.


Assuntos
Hepacivirus/metabolismo , Hepatócitos/metabolismo , Interações Hospedeiro-Patógeno , Pró-Proteína Convertase 9/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Sítios de Ligação , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HEK293 , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Hepatócitos/virologia , Humanos , Mutação , Inibidores de PCSK9 , Plasmídeos/química , Plasmídeos/metabolismo , Pró-Proteína Convertase 9/genética , Ligação Proteica , Proteólise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transdução de Sinais , Transfecção , Proteínas não Estruturais Virais/genética
8.
Virology ; 511: 1-8, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28783500

RESUMO

Hepatitis C virus (HCV) infection is a confirmed risk factor for hepatocellular carcinoma (HCC). Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) possesses tumor suppression function that is frequently defective in HCC tumors. PTEN-Long, a translation isoform of PTEN, functions in a cell non-autonomous manner. In this study, we demonstrated that intracellular overexpression of PTEN-Long inhibits HCV replication. More importantly, we showed that treatment with extracellular PTEN-Long protein inhibits HCV replication in a dose-dependent manner. Furthermore, we showed that PTEN-Long interacts with HCV core protein and this interaction is required for HCV replication inhibition by PTEN-Long. In summary, we demonstrated, for the first time, that PTEN-Long protein, an isoform of the canonical PTEN and in the form of extracellular protein treatment, inhibits HCV replication. Our study offers an opportunity for developing additional anti-HCV agents.


Assuntos
Expressão Gênica , Hepacivirus/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Replicação Viral , Antivirais/farmacologia , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/virologia , PTEN Fosfo-Hidrolase/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas do Core Viral/metabolismo
9.
Sci Rep ; 7(1): 3695, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28623358

RESUMO

Hepatitis C virus (HCV) infection leads to severe liver diseases including hepatocellular carcinoma (HCC). Phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumour suppressor, is frequently mutated or deleted in HCC tumors. PTEN has previously been demonstrated to inhibit HCV secretion. In this study, we determined the effects of PTEN on the other steps in HCV life cycle, including entry, translation, and replication. We showed that PTEN inhibits HCV entry through its lipid phosphatase activity. PTEN has no effect on HCV RNA translation. PTEN decreases HCV replication and the protein phosphatase activity of PTEN is essential for this function. PTEN interacts with the HCV core protein and requires R50 in domain I of HCV core and PTEN residues 1-185 for this interaction. This interaction is required for PTEN-mediated inhibition of HCV replication. This gives rise to a reduction in PTEN levels and intracellular lipid abundance, which may in turn regulate HCV replication. HCV core domain I protein increases the lipid phosphatase activity of PTEN in an in vitro assay, suggesting that HCV infection can also regulate PTEN. Taken together, our results demonstrated an important regulatory role of PTEN in the HCV life cycle.


Assuntos
Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepatite C/virologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral , Linhagem Celular , Ativação Enzimática , Regulação Viral da Expressão Gênica , Humanos , Metabolismo dos Lipídeos , PTEN Fosfo-Hidrolase/química , Ligação Proteica , Biossíntese de Proteínas , Domínios e Motivos de Interação entre Proteínas , RNA Viral , Proteínas do Core Viral/química , Internalização do Vírus
10.
Mediators Inflamm ; 2015: 495370, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26074679

RESUMO

Hepatitis E virus (HEV) is a small nonenveloped single-stranded positive-sense RNA virus and is one of the major causes for acute hepatitis worldwide. CXCL-8 is a small multifunctional proinflammatory chemokine. It was reported recently that HEV infection significantly upregulates CXCL-8 gene expression. In this study, we investigated the mechanism of HEV-induced CXCL-8 transcriptional activation. Using CXCL-8 promoter reporters of different lengths ranging from -1400 to -173, we showed that -173 promoter has the highest promoter activity in the presence of HEV genomic RNA, indicating that the -173 promoter contains sequences responsible for CXCL-8 activation by HEV. Ectopic expression of the ORF-1 protein can upregulate the -173 CXCL-8 promoter activity. In contrast, expression of the ORF-2 protein suppresses the CXCL-8 promoter activity and expression of the ORF-3 protein has no effect on the CXCL-8 promoter activity. We further showed that AP-1 is required for CXCL-8 activation because neither HEV genomic RNA nor the ORF-1 protein can upregulate the -173 CXCL-8 promoter in the absence of the AP-1 binding sequence. Taken together, our results showed that HEV and HEV ORF-1 protein activate the CXCL-8 promoter via AP-1. This novel function of HEV ORF-1 protein should contribute to our understanding of HEV-host interactions and HEV-associated pathogenesis.


Assuntos
Vírus da Hepatite E/genética , Interleucina-8/genética , Regiões Promotoras Genéticas/genética , Fator de Transcrição AP-1/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Vírus da Hepatite E/fisiologia , Humanos , Fator de Transcrição AP-1/genética , Transcrição Gênica/genética
11.
J Gen Virol ; 96(8): 2114-2121, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25862017

RESUMO

Hepatitis C virus (HCV) non-structural protein 5A (NS5A) is essential for viral replication; however, its effect on HCV RNA translation remains controversial partially due to the use of reporters lacking the 3' UTR, where NS5A binds to the poly(U/UC) sequence. We investigated the role of NS5A in HCV translation using a monocistronic RNA containing a Renilla luciferase gene flanked by the HCV UTRs. We found that NS5A downregulated viral RNA translation in a dose-dependent manner. This downregulation required both the 5' and 3' UTRs of HCV because substitution of either sequence with the 5' and 3' UTRs of enterovirus 71 or a cap structure at the 5' end eliminated the effects of NS5A on translation. Translation of the HCV genomic RNA was also downregulated by NS5A. The inhibition of HCV translation by NS5A required the poly(U/UC) sequence in the 3' UTR as NS5A did not affect translation when it was deleted. In addition, we showed that, whilst the amphipathic α-helix of NS5A has no effect on viral translation, the three domains of NS5A can inhibit translation independently, also dependent on the presence of the poly(U/UC) sequence in the 3' UTR. These results suggested that NS5A downregulated HCV RNA translation through a mechanism involving the poly(U/UC) sequence in the 3' UTR.


Assuntos
Regiões 3' não Traduzidas , Regulação Viral da Expressão Gênica , Hepacivirus/genética , Hepatite C/virologia , Biossíntese de Proteínas , RNA Viral/genética , Proteínas não Estruturais Virais/metabolismo , Regiões 5' não Traduzidas , Regulação para Baixo , Hepacivirus/metabolismo , Humanos , RNA Viral/química , RNA Viral/metabolismo , Proteínas não Estruturais Virais/genética
12.
PLoS One ; 10(2): e0117514, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25658101

RESUMO

Hepatitis C virus (HCV) is a small positive-sense single-stranded RNA virus that causes severe liver diseases. Current anti-HCV therapies involving direct-acting antivirals have significantly enhanced efficacy in comparison to traditional interferon and ribavirin combination. However, further improvement is needed to eradicate HCV. Anacardic acid (AnA) is a phytochemical compound that can inhibit the activity of various cellular enzymes including histone acetyltransferases (HATs). In this study, we investigated the effects of AnA on different phases of HCV life cycle. Our data showed that AnA can inhibit HCV entry, replication, translation, and virion secretion in a dose-dependent manner with no measurable effects on cell viability. In addition, we showed that two HAT inhibitors and knocking down HAT (PCAF) by RNAi can reduce HCV replication, suggesting a mechanism of AnA's inhibitory effects on HCV. Elucidation of the AnA-mediated inhibitory mechanism should facilitate the development of new drug candidates for HCV infection.


Assuntos
Ácidos Anacárdicos/farmacologia , Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Inibidores Enzimáticos/metabolismo , Hepacivirus/fisiologia , Histona Acetiltransferases/metabolismo , Humanos
13.
World J Gastroenterol ; 19(47): 8929-39, 2013 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-24379618

RESUMO

Replication of hepatitis C virus (HCV) depends on the interaction of viral proteins with various host cellular proteins and signalling pathways. Similar to cellular proteins, post-translational modifications (PTMs) of HCV proteins are essential for proper protein function and regulation, thus, directly affecting viral life cycle and the generation of infectious virus particles. Cleavage of the HCV polyprotein by cellular and viral proteases into more than 10 proteins represents an early protein modification step after translation of the HCV positive-stranded RNA genome. The key modifications include the regulated intramembranous proteolytic cleavage of core protein, disulfide bond formation of core, glycosylation of HCV envelope proteins E1 and E2, methylation of nonstructural protein 3 (NS3), biotinylation of NS4A, ubiquitination of NS5B and phosphorylation of core and NS5B. Other modifications like ubiquitination of core and palmitoylation of core and NS4B proteins have been reported as well. For some modifications such as phosphorylation of NS3 and NS5A and acetylation of NS3, we have limited understanding of their effects on HCV replication and pathogenesis while the impact of other modifications is far from clear. In this review, we summarize the available information on PTMs of HCV proteins and discuss their relevance to HCV replication and pathogenesis.


Assuntos
Hepacivirus/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Virais/metabolismo , Animais , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/patogenicidade , Humanos , Proteínas Virais/genética , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...