Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(19)2023 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-37834238

RESUMO

Infection with Ebola virus (EBOV) is responsible for hemorrhagic fever in humans with a high mortality rate. Combined efforts of prevention and therapeutic intervention are required to tackle highly variable RNA viruses, whose infections often lead to outbreaks. Here, we have screened the 2P2I3D chemical library using a nanoluciferase-based protein complementation assay (NPCA) and isolated two compounds that disrupt the interaction of the EBOV protein fragment VP35IID with the N-terminus of the dsRNA-binding proteins PKR and PACT, involved in IFN response and/or intrinsic immunity, respectively. The two compounds inhibited EBOV infection in cell culture as well as infection by measles virus (MV) independently of IFN induction. Consequently, we propose that the compounds are antiviral by restoring intrinsic immunity driven by PACT. Given that PACT is highly conserved across mammals, our data support further testing of the compounds in other species, as well as against other negative-sense RNA viruses.


Assuntos
Ebolavirus , Doença pelo Vírus Ebola , Humanos , Animais , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/metabolismo , Ebolavirus/fisiologia , Antivirais/farmacologia , Antivirais/uso terapêutico , Mamíferos
2.
J Virol ; 95(3)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33208442

RESUMO

Hepatitis C virus (HCV) infection triggers Golgi fragmentation through the Golgi-resident protein immunity-related GTPase M (IRGM). Here, we report the roles of NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) and ASC (apoptosis-associated speck-like protein containing a caspase activation and recruitment domain [CARD]), two inflammasome components, in the initial events leading to this fragmentation. We show that ASC resides at the Golgi with IRGM at homeostasis. Upon infection, ASC dissociates from both IRGM and the Golgi and associates with HCV-induced NLRP3. NLRP3 silencing inhibits Golgi fragmentation. ASC silencing disrupts the Golgi structure in both control and infected cells and reduces the localization of IRGM at the Golgi. IRGM depletion in the ASC-silenced cells cannot totally restore the Golgi structure. These data highlight a role for ASC, upstream of the formation of the inflammasome, in regulating IRGM through its control on the Golgi. A similar mechanism occurs in response to nigericin treatment, but not in cells infected with another member of the Flaviviridae family, Zika virus (ZIKV). We propose a model for a newly ascribed function of the inflammasome components in Golgi structural remodeling during certain stimuli.IMPORTANCE Numerous pathogens can affect cellular homeostasis and organelle dynamics. Hepatitis C virus (HCV) triggers Golgi fragmentation through the immunity-related GTPase M (IRGM), a resident Golgi protein, to enhance its lipid supply for replication. Here, we reveal the role of the inflammasome components NLRP3 and ASC in this process, thus uncovering a new interplay between effectors of inflammation and viral infection or stress. We show that the inflammasome component ASC resides at the Golgi under homeostasis and associates with IRGM. Upon HCV infection, ASC is recruited to NLRP3 and dissociates from IRGM, causing Golgi fragmentation. Our results uncover that aside from their known function in the inflammation response, these host defense regulators also ensure the maintenance of intact intracellular structure in homeostasis, while their activation relieves factors leading to Golgi remodeling.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Complexo de Golgi/fisiologia , Hepacivirus/isolamento & purificação , Hepatite C/virologia , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Apoptose , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas de Ligação ao GTP/genética , Complexo de Golgi/virologia , Hepatite C/metabolismo , Hepatite C/patologia , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
3.
J Virol ; 94(22)2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-32878892

RESUMO

Yellow fever virus (YFV) is an RNA virus primarily targeting the liver. Severe YF cases are responsible for hemorrhagic fever, plausibly precipitated by excessive proinflammatory cytokine response. Pathogen recognition receptors (PRRs), such as the cytoplasmic retinoic acid inducible gene I (RIG-I)-like receptors (RLRs), and the viral RNA sensor protein kinase R (PKR), are known to initiate a proinflammatory response upon recognition of viral genomes. Here, we sought to reveal the main determinants responsible for the acute cytokine expression occurring in human hepatocytes following YFV infection. Using a RIG-I-defective human hepatoma cell line, we found that RIG-I largely contributes to cytokine secretion upon YFV infection. In infected RIG-I-proficient hepatoma cells, RIG-I was localized in stress granules. These granules are large aggregates of stalled translation preinitiation complexes known to concentrate RLRs and PKR and are so far recognized as hubs orchestrating RNA virus sensing. Stable knockdown of PKR in hepatoma cells revealed that PKR contributes to both stress granule formation and cytokine induction upon YFV infection. However, stress granule disruption did not affect the cytokine response to YFV infection, as assessed by small interfering RNA (siRNA)-knockdown-mediated inhibition of stress granule assembly. Finally, no viral RNA was detected in stress granules using a fluorescence in situ hybridization approach coupled with immunofluorescence. Our findings suggest that both RIG-I and PKR mediate proinflammatory cytokine induction in YFV-infected hepatocytes, in a stress granule-independent manner. Therefore, by showing the uncoupling of the cytokine response from the stress granule formation, our model challenges the current view in which stress granules are required for the mounting of the acute antiviral response.IMPORTANCE Yellow fever is a mosquito-borne acute hemorrhagic disease caused by yellow fever virus (YFV). The mechanisms responsible for its pathogenesis remain largely unknown, although increased inflammation has been linked to worsened outcome. YFV targets the liver, where it primarily infects hepatocytes. We found that two RNA-sensing proteins, RIG-I and PKR, participate in the induction of proinflammatory mediators in human hepatocytes infected with YFV. We show that YFV infection promotes the formation of cytoplasmic structures, termed stress granules, in a PKR- but not RIG-I-dependent manner. While stress granules were previously postulated to be essential platforms for immune activation, we found that they are not required for the production of proinflammatory mediators upon YFV infection. Collectively, our work uncovered molecular events triggered by the replication of YFV, which could prove instrumental in clarifying the pathogenesis of the disease, with possible repercussions for disease management.


Assuntos
Proteína DEAD-box 58/metabolismo , Vírus da Febre Amarela/metabolismo , eIF-2 Quinase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Carcinoma Hepatocelular , Linhagem Celular , Linhagem Celular Tumoral , Citocinas/metabolismo , Proteína DEAD-box 58/deficiência , Proteína DEAD-box 58/genética , DNA Helicases/genética , Técnicas de Silenciamento de Genes , Haplorrinos , Hepatócitos/virologia , Humanos , Proteínas de Ligação a Poli-ADP-Ribose/genética , RNA Helicases/genética , Proteínas com Motivo de Reconhecimento de RNA/genética , RNA Interferente Pequeno , RNA Viral/genética , Proteínas de Ligação a RNA/genética , Receptores Imunológicos , Antígeno-1 Intracelular de Células T/genética , Transcriptoma , eIF-2 Quinase/genética
4.
Proc Natl Acad Sci U S A ; 114(17): E3462-E3471, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28389568

RESUMO

Positive-stranded RNA viruses, such as hepatitis C virus (HCV), assemble their viral replication complexes by remodeling host intracellular membranes to a membranous web. The precise composition of these replication complexes and the detailed mechanisms by which they are formed are incompletely understood. Here we show that the human immunity-related GTPase M (IRGM), known to contribute to autophagy, plays a previously unrecognized role in this process. We show that IRGM is localized at the Golgi apparatus and regulates the fragmentation of Golgi membranes in response to HCV infection, leading to colocalization of Golgi vesicles with replicating HCV. Our results show that IRGM controls phosphorylation of GBF1, a guanine nucleotide exchange factor for Arf-GTPases, which normally operates in Golgi membrane dynamics and vesicle coating in resting cells. We also find that HCV triggers IRGM-mediated phosphorylation of the early autophagy initiator ULK1, thereby providing mechanistic insight into the role of IRGM in HCV-mediated autophagy. Collectively, our results identify IRGM as a key Golgi-situated regulator that links intracellular membrane remodeling by autophagy and Golgi fragmentation with viral replication.


Assuntos
Autofagia , Proteínas de Ligação ao GTP/metabolismo , Complexo de Golgi/metabolismo , Hepacivirus/fisiologia , Membranas Intracelulares/metabolismo , Replicação Viral/fisiologia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação ao GTP/genética , Complexo de Golgi/genética , Complexo de Golgi/virologia , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Membranas Intracelulares/virologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosforilação/genética
5.
Liver Int ; 38(3): 432-442, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28752959

RESUMO

BACKGROUND & AIMS: The natural outcomes of hepatitis C virus (HCV) as well as the progression of the liver disease are highly variable and depend primarily on an efficient immune response. As toll-like receptors seven (TLR7) and eight (TLR8) are important effectors of the innate immunity, this study aims to evaluate the association between TLR7 and TLR8 polymorphisms and the HCV infection outcomes in Moroccan subjects. METHODS: In this case-control study, 643 subjects including 293 mild chronic hepatitis patients, 119 with advanced liver disease (AdLD), 93 with HCV spontaneous clearance and 138 healthy controls were genotyped using TaqMan SNPs assays. RESULTS: Patients carrying TLR7 rs179008-A allele were more likely to clear the virus spontaneously (P = .0001 for women, and P < .001 for men). Besides, carriage of TLR7 rs179009-A allele was associated with a twofold increase in spontaneous viral clearance in female patients (P = .0002), but not in men. In addition, we observed that TLR7 rs179008-T and rs179009-G alleles increased the risk of disease progression in both sexes (P < .05). TLR8 rs3764880-G allele was associated with spontaneous HCV clearance in both sexes (P < .0001) albeit with an apparently stronger association in males (OR = 6.02 for men vs 2.2 for women). In males, TLR8 rs3764879-C and TLR8 rs3764880-A alleles were significantly associated with AdLD status (P < .05). CONCLUSIONS: Our results suggest that variations in TLR7 and TLR8 genes modulate the clearance and progression of HCV infection with different magnitudes between sexes. Our results refine, therefore, our understanding of the sex-specific differences observed regarding the susceptibility to chronic hepatitis.


Assuntos
Progressão da Doença , Hepatite C/genética , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética , Idoso , Alelos , Estudos de Casos e Controles , Feminino , Frequência do Gene , Variação Genética , Genótipo , Humanos , Fígado/fisiopatologia , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Fatores Sexuais
6.
J Psychiatry Neurosci ; 40(3): 151-61, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25455349

RESUMO

BACKGROUND: Alzheimer disease is characterized by cognitive decline, senile plaques of ß-amyloid (Aß) peptides, neurofibrillary tangles composed of hyperphosphorylated τ proteins and neuronal loss. Aß and τ are useful markers in the cerebrospinal fluid (CSF). C-Jun N-terminal kinases (JNKs) are serine-threonine protein kinases activated by phosphorylation and involved in neuronal death. METHODS: In this study, Western blots, enzyme-linked immunosorbent assay and histological approaches were used to assess the concentrations of Aß, τ and JNK isoforms in postmortem brain tissue samples (10 Alzheimer disease and 10 control) and in CSF samples from 30 living patients with Alzheimer disease and 27 controls with neurologic disease excluding Alzheimer disease. Patients with Alzheimer disease were followed for 1-3 years and assessed using Mini-Mental State Examination scores. RESULTS: The biochemical and morphological results showed a significant increase of JNK3 and phosphorylated JNK levels in patients with Alzheimer disease, and JNK3 levels correlated with Aß42 levels. Confocal microscopy revealed that JNK3 was associated with Aß in senile plaques. The JNK3 levels in the CSF were significantly elevated in patients with Alzheimer disease and correlated statistically with the rate of cognitive decline in a mixed linear model. LIMITATIONS: The study involved different samples grouped into 3 small cohorts. Evaluation of JNK3 in CSF was possible only with immunoblot analysis. CONCLUSION: We found that JNK3 levels are increased in brain tissue and CSF from patients with Alzheimer disease. The finding that increased JNK3 levels in CSF could reflect the rate of cognitive decline is new and merits further investigation.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Encéfalo/enzimologia , Encéfalo/patologia , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Idoso , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/metabolismo , Biomarcadores/líquido cefalorraquidiano , Estudos de Coortes , Progressão da Doença , Feminino , Humanos , Masculino , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/enzimologia , Placa Amiloide/patologia , Proteínas tau/líquido cefalorraquidiano
7.
J Virol ; 87(14): 8241-6, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23658442

RESUMO

Upon infection with Bluetongue virus (BTV), an arthropod-borne virus, type I interferon (IFN-I) is produced in vivo and in vitro. IFN-I is essential for the establishment of an antiviral cellular response, and most if not all viruses have elaborated strategies to counteract its action. In this study, we assessed the ability of BTV to interfere with IFN-I synthesis and identified the nonstructural viral protein NS3 as an antagonist of the IFN-I system.


Assuntos
Vírus Bluetongue/imunologia , Imunidade Inata/imunologia , Interferon Tipo I/antagonistas & inibidores , Transdução de Sinais/imunologia , Proteínas não Estruturais Virais/metabolismo , Western Blotting , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon Tipo I/biossíntese , Luciferases , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Proteínas não Estruturais Virais/farmacologia
8.
J Virol ; 86(10): 5817-28, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438548

RESUMO

Dendritic cells (DCs), especially plasmacytoid DCs (pDCs), produce large amounts of alpha/beta interferon (IFN-α/ß) upon infection with DNA or RNA viruses, which has impacts on the physiopathology of the viral infections and on the quality of the adaptive immunity. However, little is known about the IFN-α/ß production by DCs during infections by double-stranded RNA (dsRNA) viruses. We present here novel information about the production of IFN-α/ß induced by bluetongue virus (BTV), a vector-borne dsRNA Orbivirus of ruminants, in sheep primary DCs. We found that BTV induced IFN-α/ß in skin lymph and in blood in vivo. Although BTV replicated in a substantial fraction of the conventional DCs (cDCs) and pDCs in vitro, only pDCs responded to BTV by producing a significant amount of IFN-α/ß. BTV replication in pDCs was not mandatory for IFN-α/ß production since it was still induced by UV-inactivated BTV (UV-BTV). Other inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and IL-12p40, were also induced by UV-BTV in primary pDCs. The induction of IFN-α/ß required endo-/lysosomal acidification and maturation. However, despite being an RNA virus, UV-BTV did not signal through Toll-like receptor 7 (TLR7) for IFN-α/ß induction. In contrast, pathways involving the MyD88 adaptor and kinases dsRNA-activated protein kinase (PKR) and stress-activated protein kinase (SAPK)/Jun N-terminal protein kinase (JNK) were implicated. This work highlights the importance of pDCs for the production of innate immunity cytokines induced by a dsRNA virus, and it shows that a dsRNA virus can induce IFN-α/ß in pDCs via a novel TLR-independent and Myd88-dependent pathway. These findings have implications for the design of efficient vaccines against dsRNA viruses.


Assuntos
Vírus Bluetongue/imunologia , Bluetongue/imunologia , Células Dendríticas/imunologia , Interferon Tipo I/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , Animais , Bluetongue/genética , Bluetongue/virologia , Vírus Bluetongue/genética , Vírus Bluetongue/fisiologia , Células Cultivadas , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/virologia , Feminino , Imunidade Inata , Interferon Tipo I/genética , Glicoproteínas de Membrana , Fator 88 de Diferenciação Mieloide/genética , Receptores de Interleucina-1 , Ovinos/imunologia , Ovinos/virologia , Transdução de Sinais , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética
9.
PLoS Pathog ; 7(10): e1002289, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22022264

RESUMO

Recognition of viral RNA structures by the intracytosolic RNA helicase RIG-I triggers induction of innate immunity. Efficient induction requires RIG-I ubiquitination by the E3 ligase TRIM25, its interaction with the mitochondria-bound MAVS protein, recruitment of TRAF3, IRF3- and NF-κB-kinases and transcription of Interferon (IFN). In addition, IRF3 alone induces some of the Interferon-Stimulated Genes (ISGs), referred to as early ISGs. Infection of hepatocytes with Hepatitis C virus (HCV) results in poor production of IFN despite recognition of the viral RNA by RIG-I but can lead to induction of early ISGs. HCV was shown to inhibit IFN production by cleaving MAVS through its NS3/4A protease and by controlling cellular translation through activation of PKR, an eIF2α-kinase containing dsRNA-binding domains (DRBD). Here, we have identified a third mode of control of IFN induction by HCV. Using HCVcc and the Huh7.25.CD81 cells, we found that HCV controls RIG-I ubiquitination through the di-ubiquitine-like protein ISG15, one of the early ISGs. A transcriptome analysis performed on Huh7.25.CD81 cells silenced or not for PKR and infected with JFH1 revealed that HCV infection leads to induction of 49 PKR-dependent genes, including ISG15 and several early ISGs. Silencing experiments revealed that this novel PKR-dependent pathway involves MAVS, TRAF3 and IRF3 but not RIG-I, and that it does not induce IFN. Use of PKR inhibitors showed that this pathway requires the DRBD but not the kinase activity of PKR. We then demonstrated that PKR interacts with HCV RNA and MAVS prior to RIG-I. In conclusion, HCV recruits PKR early in infection as a sensor to trigger induction of several IRF3-dependent genes. Among those, ISG15 acts to negatively control the RIG-I/MAVS pathway, at the level of RIG-I ubiquitination.These data give novel insights in the machinery involved in the early events of innate immune response.


Assuntos
Citocinas/metabolismo , Hepacivirus/imunologia , Hepacivirus/metabolismo , Interferons/biossíntese , Receptores do Ácido Retinoico/metabolismo , Ubiquitinas/metabolismo , eIF-2 Quinase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Citocinas/biossíntese , Citocinas/genética , Perfilação da Expressão Gênica , Hepacivirus/genética , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Fator Regulador 3 de Interferon/biossíntese , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interferons/genética , Interferência de RNA , RNA Interferente Pequeno , RNA Viral/metabolismo , Receptores do Ácido Retinoico/biossíntese , Receptores do Ácido Retinoico/genética , Transdução de Sinais , Fator 3 Associado a Receptor de TNF/biossíntese , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/metabolismo , Ubiquitinação , Ubiquitinas/biossíntese , Ubiquitinas/genética , eIF-2 Quinase/biossíntese , eIF-2 Quinase/genética
10.
Mol Membr Biol ; 27(1): 45-62, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20001747

RESUMO

The Hepatitis C virus (HCV) NS4B protein, a multispanning endoplasmic reticulum (ER) membrane protein, generates intracellular rearrangements of ER-derived membranes, essential for HCV replication. In this study, we characterized NS4B elements involved in the process of targeting, association and retention in the ER membrane. We investigated the localization and membrane association of a number of C- or N-terminal NS4B deletions expressed as GFP chimeras by biochemical and fluorescence microscopy techniques. A second set of GFP-NS4B chimeras containing the plasma membrane ecto-ATPase CD39 at the C-terminus of each NS4B deletion mutant was used to further examine the role of N-terminal NS4B sequences in ER retention. Several structural elements, besides the first two transmembrane domains (TMs), within the NS4B N-terminal half (residues 1-130) were found to mediate association of the NS4B-GFP chimeras with ER membranes. Both TM1 and TM2 are required for ER anchoring and retention but are not sufficient for ER retention. Sequences upstream of TM1 are also required. These include two putative amphipathic alpha-helices and a Leucine Rich Repeat-like motif, a sequence highly conserved in all HCV genotypes. The N-terminal 55peptidic sequence, containing the 1st amphipathic helix, mediates association of the 55N-GFP chimera with cellular membranes including the ER, but is dispensable for ER targeting of the entire NS4B molecule. Importantly, the C-terminal 70peptidic sequence can associate with membranes positive for ER markers in the absence of any predicted TMs. In conclusion, HCV NS4B targeting and retention in the ER results from the concerted action of several NS4B structural elements.


Assuntos
Retículo Endoplasmático/metabolismo , Hepacivirus/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Motivos de Aminoácidos/fisiologia , Antígenos CD/genética , Antígenos CD/metabolismo , Apirase/genética , Apirase/metabolismo , Linhagem Celular Tumoral , Retículo Endoplasmático/genética , Retículo Endoplasmático/virologia , Humanos , Mapeamento de Peptídeos/métodos , Estrutura Terciária de Proteína/fisiologia , Proteínas não Estruturais Virais/genética
11.
J Virol ; 83(19): 10119-28, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19605474

RESUMO

The interferon-induced protein kinase RNA activated (PKR) is activated after virus infection. This activation is transient during the human immunodeficiency virus type 1 (HIV-1) infection of lymphocytes, and the protein is not activated at the peak of infection. We observed that interferon-induced adenosine deaminase acting on RNA 1-p150 (ADAR1-p150) and ADAR1-p110 expression increases while the virus replicates actively. Furthermore, both forms of ADAR1 show enhanced interactions with PKR at the peak of HIV infection, suggesting a role for this protein in the regulation of PKR activation. We observed that ADAR1-p150, as previously shown for the TAR RNA binding protein (TRBP), reverses the PKR inhibition of HIV expression and production in HEK 293T cells. This activity requires the Z-DNA binding motif and the three double-stranded RNA binding domains but not the catalytic domain. In astrocytic cells, ADAR1-p150 increased HIV expression and production to an extent similar to that of TRBP. Small interfering RNAs against ADAR1-p150 moderately decreased HIV production. These results indicate that two interferon-induced proteins, ADAR1 and PKR, have antagonistic functions on HIV production. They suggest that ADAR1 and TRBP belong to a multiprotein complex that inhibits PKR during the HIV infection of lymphocytes.


Assuntos
Adenosina Desaminase/genética , Adenosina Desaminase/fisiologia , Regulação Viral da Expressão Gênica , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Linfócitos/virologia , Replicação Viral , eIF-2 Quinase/metabolismo , Motivos de Aminoácidos , Linhagem Celular , Fator de Iniciação 2 em Eucariotos/metabolismo , HIV-1/metabolismo , Humanos , Células Jurkat , Modelos Biológicos , Fosforilação , Proteínas de Ligação a RNA , Transfecção
12.
J Virol ; 82(1): 569-74, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17942559

RESUMO

Hepatitis C virus-positive serum (HCVser, genotypes 1a to 3a) or HCV cell culture (JFH1/HCVcc) infection of primary normal human hepatocytes was assessed by measuring intracellular HCV RNA strands. Anti-CD81 antibodies and siRNA-CD81 silencing markedly inhibited (>90%) HCVser infection irrespective of HCV genotype, viral load, or liver donor, while hCD81-large intracellular loop (LEL) had no effect. However, JFH1/HCVcc infection of hepatocytes was modestly inhibited (40 to 60%) by both hCD81-LEL and anti-CD81 antibodies. In conclusion, CD81 is involved in HCVser infection of human hepatocytes, and comparative studies of HCVser versus JFH1/HCVcc infection of human hepatocytes and Huh-7.5 cells revealed that the cell-virion combination is determinant of the entry process.


Assuntos
Antígenos CD/fisiologia , Hepacivirus/fisiologia , Hepatócitos/virologia , Receptores Virais/fisiologia , Internalização do Vírus , Adolescente , Adulto , Idoso , Antígenos CD/genética , Antígenos CD/imunologia , Células Cultivadas , Feminino , Inativação Gênica , Hepatócitos/química , Humanos , Masculino , Pessoa de Meia-Idade , RNA Viral/análise , Receptores Virais/antagonistas & inibidores , Receptores Virais/genética , Receptores Virais/imunologia , Tetraspanina 28
13.
Aging Cell ; 18(3): e12887, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30821420

RESUMO

Brain lesions in Alzheimer's disease (AD) include amyloid plaques made of Aß peptides and neurofibrillary tangles composed of hyperphosphorylated tau protein with synaptic and neuronal loss and neuroinflammation. Aß oligomers can trigger tau phosphorylation and neuronal alterations through activation of neuronal kinases leading to progressive cognitive decline. PKR is a ubiquitous pro-apoptotic serine/threonine kinase, and levels of activated PKR are increased in AD brains and AD CSF. In addition, PKR regulates negatively memory formation in mice. To assess the role of PKR in an AD in vivo model, we crossed 5xFAD transgenic mice with PKR knockout (PKRKO) mice and we explored the contribution of PKR on cognition and brain lesions in the 5xFAD mouse model of AD as well as in neuron-microglia co-cultures exposed to the innate immunity activator lipopolysaccharide (LPS). Nine-month-old double-mutant mice revealed significantly improved memory consolidation with the new object location test, starmaze test, and elevated plus maze test as compared to 5xFAD mice. Brain amyloid accumulation and BACE1 levels were statistically decreased in double-mutant mice. Apoptosis, neurodegeneration markers, and synaptic alterations were significantly reduced in double-mutant mice as well as neuroinflammation markers such as microglial load and brain cytokine levels. Using cocultures, we found that PKR in neurons was essential for LPS microglia-induced neuronal death. Our results demonstrate the clear involvement of PKR in abnormal spatial memory and brain lesions in the 5xFAD model and underline its interest as a target for neuroprotection in AD.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Memória Espacial , eIF-2 Quinase/metabolismo , Doença de Alzheimer/patologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/patologia , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , eIF-2 Quinase/deficiência
14.
Sci STKE ; 2007(384): pe20, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17473309

RESUMO

The cytoplasmic CARD-containing DExD/H box RNA helicases RIG-I and MDA5 act as sensors of viral infections through recognition of viral double-stranded (ds) RNAs. They both associate with the mitochondrial adaptor IPS-1 (also referred to as MAVS, VISA, and CARDIF) through homotypic CARD-CARD interactions. IPS-1, in turn, triggers signaling pathways, including activation of the protein kinases TBK1 and IKKepsilon, responsible for the phosphorylation of IRF3, a key transcription factor involved in interferon (IFN) synthesis, one essential element of the innate immune response. RIG-I remains in an autoinhibited state in the absence of dsRNA, through an internal repressor domain (RD) that binds within both its CARD and its RNA helicase domains and therefore acts in cis to control its multimerization and interaction with IPS-1. Ectopic expression of the RD prevents signaling and increases cell permissiveness to viruses, including hepatitis C virus. LGP2, which is another DExD/H RNA helicase of the RIG-I and MDA5 family and which is devoid of CARD domain, negatively controls IFN induction at different levels: by sequestering dsRNA, by blocking RIG-I's multimerization in trans through a domain analogous to the RIG-I RD, and by competing with the protein kinase IKKepsilon for a common interaction site on IPS-1. The ability of RIG-I and LGP2 to exert such a feedback control at the earliest steps of IFN synthesis allows the cells to exert a tight regulation of the induction of the innate immune response.


Assuntos
RNA Helicases DEAD-box/fisiologia , Interferons/biossíntese , RNA Helicases/fisiologia , Linhagem Celular , Proteína DEAD-box 58 , Humanos , Imunidade Inata , Fosforilação , Receptores Imunológicos , Transdução de Sinais
15.
Sci Rep ; 8(1): 11185, 2018 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-30046100

RESUMO

Clinical studies have suggested association of some hepatitis C virus (HCV) subtypes or isolates with progression toward hepatocellular carcinoma (HCC). HCV core protein has been reported to interfere with host Wnt/ß-catenin pathway, a cell fate-determining pathway, which plays a major role in HCC. Here, we investigated the impact of HCV core genetic variability in the dysregulation of Wnt/ß-catenin pathway. We used both transient expression of core proteins from clinical isolates of HCV subtypes 1a (Cambodia), 4a (Romania) and 4f (Cameroon) and infection systems based on a set of engineered intergenotypic recombinant viruses encoding core from these various clinical strains. We found that TCF transcription factor-dependent reporter activity was upregulated by core in a strain-specific manner. We documented core sequence-specific transcriptional upregulation of several ß-catenin downstream target genes associated with cell proliferation and malignant transformation, fibrogenesis or fat accumulation. The extent of ß-catenin nuclear translocation varied in accordance with ß-catenin downstream gene upregulation in infected cells. Pairwise comparisons of subgenotypic core recombinants and mutated core variants unveiled the critical role of core residues 64 and 71 in these dysregulations. In conclusion, this work identified natural core polymorphisms involved in HCV strain-specific activation of Wnt/ß-catenin pathway in relevant infection systems.


Assuntos
Carcinoma Hepatocelular/genética , Hepacivirus/genética , Neoplasias Hepáticas/genética , beta Catenina/genética , Transporte Ativo do Núcleo Celular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , Genótipo , Células HEK293 , Hepacivirus/patogenicidade , Hepatite C/genética , Hepatite C/patologia , Hepatite C/virologia , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Fator 1 de Transcrição de Linfócitos T/genética , Via de Sinalização Wnt/genética
16.
Dev Comp Immunol ; 85: 95-107, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29635006

RESUMO

Bats are known to harbor many zoonotic viruses, some of which are pathogenic to other mammals while they seem to be harmless in bats. As the interferon (IFN) response represents the first line of defense against viral infections in mammals, it is hypothesized that activation of the IFN system is one of the mechanisms enabling bats to co-exist with viruses. We have previously reported induction of type I IFN in a cell line from the common vampire bat, Desmodus rotundus, upon polyinosinic:polycytidylic acid (poly(I:C)) stimulation. To deepen our knowledge on D. rotundus' IFN-I antiviral response, we molecularly characterized three interferon-stimulated genes (ISGs), OAS1, PKR and ADAR1, closely implicated in the IFN-I antiviral response, and tested their functionality in our cellular model. We first found that D. rotundus encoded two OAS1 paralogs, OAS1a and OAS1b, and that the functional domains of the four ISGs characterized were highly conserved with those of other mammals. Despite their significant transcription level in the absence of stimulation, the transcription of the four ISGs characterized was enhanced by poly(I:C). In addition, the transcription of OAS1a and OAS1b appears to be differentially regulated. These findings demonstrate an active ISG antiviral response in D. rotundus in which OAS1b may play an important role.


Assuntos
2',5'-Oligoadenilato Sintetase/genética , Adenosina Desaminase/genética , Antivirais/farmacologia , Quirópteros/genética , Interferons/farmacologia , eIF-2 Quinase/genética , Animais , Linhagem Celular , Poli I-C/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Viroses/genética
17.
Dev Comp Immunol ; 81: 1-7, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29122634

RESUMO

Though the common vampire bat, Desmodus rotundus, is known as the main rabies virus reservoir in Latin America, no tools are available to investigate its antiviral innate immune system. To characterize the IFN-I pathway, we established an immortalized cell line from a D. rotundus fetal lung named FLuDero. Then we molecularly characterized some of the Toll-like receptors (TLR3, 7, 8 and 9), the three RIG-I-like receptor members, as well as IFNα1 and IFNß. Challenging the FLuDero cell line with poly (I:C) resulted in an up-regulation of both IFNα1 and IFNß and the induction of expression of the different pattern recognition receptors characterized. These findings provide evidence of the intact dsRNA recognition machinery and the IFN-I signaling pathway in our cellular model. Herein, we generated a sum of insightful specific molecular and cellular tools that will serve as a useful model to study virus-host interactions of the common vampire bat.


Assuntos
Quirópteros/imunologia , Proteína DEAD-box 58/genética , Pulmão/citologia , Vírus da Raiva/fisiologia , Receptores Toll-Like/genética , Animais , Linhagem Celular Transformada , Quirópteros/genética , Clonagem Molecular , Reservatórios de Doenças , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Pulmão/imunologia , Poli I-C/imunologia , RNA de Cadeia Dupla/imunologia , Transdução de Sinais
18.
Cell Signal ; 18(11): 1865-75, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16600570

RESUMO

The interferon-induced double-stranded RNA (dsRNA)-activated protein kinase (PKR) has been shown to activate NF-kappaB independently of its kinase function after interaction with the IKK complex. In order to investigate the mechanism of NF-kappaB activation by PKR, we identified the domain of PKR responsible for stimulating the NF-kappaB pathway in PKR-deficient fibroblasts using an NF-kappaB dependent reporter assay. The N-terminal 1-265 AA of PKR activates NF-kappaB, whereas the 1-180 AA N-terminus restricted to the two dsRNA Binding Domains (DRBD), the third basic domain alone (AA 181-265), or the C-terminus of PKR (AA 266-550) were unable to stimulate the expression of the NF-kappaB dependent reporter gene. Using confocal microscopy, we confirmed that PKR full length as well as PKR N-terminus colocalized with IKKbeta. By GST-pulldown analysis, using different PKR domains, we then revealed the specific ability of the PKR N-terminus 1-265 to bind to and activate IKK and showed that this activation requires the integrity of the IKK complex. This activation is not only due to DRBDs since the DRBD fragment 1-180 failed to inhibit PKR 1-265 induced NF-kappaB activation. Our results therefore demonstrate that the ability of PKR to mediate NF-kappaB activation resides in its full N-terminus, and requires both DRBDs and the third basic domain.


Assuntos
Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , eIF-2 Quinase/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Técnicas de Transferência de Genes , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Estrutura Terciária de Proteína/fisiologia , Regulação para Cima , eIF-2 Quinase/genética
19.
World J Gastroenterol ; 13(17): 2446-54, 2007 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-17552028

RESUMO

The innate immune response is triggered by a variety of pathogens, including viruses, and requires rapid induction of type I interferons (IFN), such as IFNbeta and IFNalpha. IFN induction occurs when specific pathogen motifs bind to specific cellular receptors. In non-professional immune, virally-infected cells, IFN induction is essentially initiated after the binding of dsRNA structures to TLR3 receptors or to intracytosolic RNA helicases, such as RIG-I/MDA5. This leads to the recruitment of specific adaptors, such as TRIF for TLR3 and the mitochondrial-associated IPS-1/VISA/MAVS/CARDIF adapter protein for the RNA helicases, and the ultimate recruitment of kinases, such as MAPKs, the canonical IKK complex and the TBK1/IKKepsilon kinases, which activate the transcription factors ATF-2/c-jun, NF-kappaB and IRF3, respectively. The coordinated action of these transcription factors leads to induction of IFN and of pro-inflammatory cytokines and to the establishment of the innate immune response. HCV can cleave both the adapters TRIF and IPS-1/VISA/MAVS/CARDIF through the action of its NS3/4A protease. This provokes abrogation of the induction of the IFN and cytokine pathways and favours viral propagation and presumably HCV chronic infection.


Assuntos
Hepacivirus/fisiologia , Interferons/fisiologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Hepatite C/enzimologia , Hepatite C/imunologia , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , RNA Helicases/metabolismo , Receptores Toll-Like/fisiologia
20.
Sci Rep ; 7(1): 16129, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-29170442

RESUMO

PKR is a cellular kinase involved in the regulation of the integrative stress response (ISR) and pro-inflammatory pathways. Two N-terminal dsRNA Binding Domains (DRBD) are required for activation of PKR, by interaction with either dsRNA or PACT, another cellular DRBD-containing protein. A role for PKR and PACT in inflammatory processes linked to neurodegenerative diseases has been proposed and raised interest for pharmacological PKR inhibitors. However, the role of PKR in inflammation is subject to controversy. We identified the flavonoid luteolin as an inhibitor of the PKR/PACT interaction at the level of their DRBDs using high-throughput screening of chemical libraries by homogeneous time-resolved fluorescence. This was further validated using NanoLuc-Based Protein Complementation Assay. Luteolin inhibits PKR phosphorylation, the ISR and the induction of pro-inflammatory cytokines in human THP1 macrophages submitted to oxidative stress and toll-like receptor (TLR) agonist. Similarly, luteolin inhibits induction of pro-inflammatory cytokines in murine microglial macrophages. In contrast, luteolin increased activation of the inflammasome, in a PKR-independent manner. Collectively, these data delineate the importance of PKR in the inflammation process to the ISR and induction of pro-inflammatory cytokines. Pharmacological inhibitors of PKR should be used in combination with drugs targeting directly the inflammasome.


Assuntos
Inflamação/metabolismo , Proteínas de Ligação a RNA/metabolismo , eIF-2 Quinase/metabolismo , Células HEK293 , Humanos , Inflamação/imunologia , Fosforilação/genética , Fosforilação/fisiologia , Ligação Proteica/genética , Ligação Proteica/fisiologia , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/genética , eIF-2 Quinase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA