Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Int J Mol Sci ; 24(19)2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37834238

ABSTRACT

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.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Animals , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/metabolism , Ebolavirus/physiology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Mammals
2.
J Virol ; 95(3)2021 01 13.
Article in English | MEDLINE | ID: mdl-33208442

ABSTRACT

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.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , GTP-Binding Proteins/metabolism , Golgi Apparatus/physiology , Hepacivirus/isolation & purification , Hepatitis C/virology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Apoptosis , CARD Signaling Adaptor Proteins/genetics , GTP-Binding Proteins/genetics , Golgi Apparatus/virology , Hepatitis C/metabolism , Hepatitis C/pathology , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
3.
J Virol ; 94(22)2020 10 27.
Article in English | MEDLINE | ID: mdl-32878892

ABSTRACT

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.


Subject(s)
DEAD Box Protein 58/metabolism , Yellow fever virus/metabolism , eIF-2 Kinase/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Hepatocellular , Cell Line , Cell Line, Tumor , Cytokines/metabolism , DEAD Box Protein 58/deficiency , DEAD Box Protein 58/genetics , DNA Helicases/genetics , Gene Knockdown Techniques , Haplorhini , Hepatocytes/virology , Humans , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , RNA, Small Interfering , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Receptors, Immunologic , T-Cell Intracellular Antigen-1/genetics , Transcriptome , eIF-2 Kinase/genetics
4.
Aging Cell ; 18(3): e12887, 2019 06.
Article in English | MEDLINE | ID: mdl-30821420

ABSTRACT

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.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Disease Models, Animal , Spatial Memory , eIF-2 Kinase/metabolism , Alzheimer Disease/pathology , Animals , Behavior, Animal/drug effects , Brain/pathology , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , eIF-2 Kinase/deficiency
5.
Sci Rep ; 8(1): 11185, 2018 07 25.
Article in English | MEDLINE | ID: mdl-30046100

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular/genetics , Hepacivirus/genetics , Liver Neoplasms/genetics , beta Catenin/genetics , Active Transport, Cell Nucleus/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Genotype , HEK293 Cells , Hepacivirus/pathogenicity , Hepatitis C/genetics , Hepatitis C/pathology , Hepatitis C/virology , Humans , Liver Neoplasms/pathology , Liver Neoplasms/virology , T Cell Transcription Factor 1/genetics , Wnt Signaling Pathway/genetics
6.
Dev Comp Immunol ; 85: 95-107, 2018 08.
Article in English | MEDLINE | ID: mdl-29635006

ABSTRACT

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.


Subject(s)
2',5'-Oligoadenylate Synthetase/genetics , Adenosine Deaminase/genetics , Antiviral Agents/pharmacology , Chiroptera/genetics , Interferons/pharmacology , eIF-2 Kinase/genetics , Animals , Cell Line , Poly I-C/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Virus Diseases/genetics
7.
Liver Int ; 38(3): 432-442, 2018 03.
Article in English | MEDLINE | ID: mdl-28752959

ABSTRACT

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.


Subject(s)
Disease Progression , Hepatitis C/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/genetics , Aged , Alleles , Case-Control Studies , Female , Gene Frequency , Genetic Variation , Genotype , Humans , Liver/physiopathology , Logistic Models , Male , Middle Aged , Sex Factors
8.
Dev Comp Immunol ; 81: 1-7, 2018 04.
Article in English | MEDLINE | ID: mdl-29122634

ABSTRACT

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.


Subject(s)
Chiroptera/immunology , DEAD Box Protein 58/genetics , Lung/cytology , Rabies virus/physiology , Toll-Like Receptors/genetics , Animals , Cell Line, Transformed , Chiroptera/genetics , Cloning, Molecular , Disease Reservoirs , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon-alpha/metabolism , Interferon-beta/metabolism , Lung/immunology , Poly I-C/immunology , RNA, Double-Stranded/immunology , Signal Transduction
9.
Sci Rep ; 7(1): 16129, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170442

ABSTRACT

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.


Subject(s)
Inflammation/metabolism , RNA-Binding Proteins/metabolism , eIF-2 Kinase/metabolism , HEK293 Cells , Humans , Inflammation/immunology , Phosphorylation/genetics , Phosphorylation/physiology , Protein Binding/genetics , Protein Binding/physiology , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , eIF-2 Kinase/genetics
10.
Proc Natl Acad Sci U S A ; 114(17): E3462-E3471, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28389568

ABSTRACT

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.


Subject(s)
Autophagy , GTP-Binding Proteins/metabolism , Golgi Apparatus/metabolism , Hepacivirus/physiology , Intracellular Membranes/metabolism , Virus Replication/physiology , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line, Tumor , GTP-Binding Proteins/genetics , Golgi Apparatus/genetics , Golgi Apparatus/virology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Intracellular Membranes/virology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation/genetics
11.
J Alzheimers Dis ; 54(4): 1659-1670, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27636848

ABSTRACT

In Alzheimer's disease (AD), the amyloid cascade hypothesis proposes that amyloid-beta (Aß) neurotoxicity leads to neuroinflammation, synaptic loss, and neuronal degeneration. In AD patients, anti-amyloid immunotherapies did not succeed because they were possibly administered late in AD progression. Modulating new targets associated with Aß toxicity, such as PKR (double-stranded RNA dependent kinase), and JNK (c-Jun N-terminal kinase) is a major goal for neuroprotection. These two pro-apoptotic kinases are activated in AD brains and involved in Aß production, tau phosphorylation, neuroinflammation, and neuronal death. In HEK cells transfected with siRNA directed against PKR, and in PKR knockout (PKR-/-) mice neurons, we showed that PKR triggers JNK activation. Aß-induced neuronal apoptosis, measured by cleaved PARP (Poly ADP-ribose polymerase) and cleaved caspase 3 levels, was reduced in PKR-/- neurons. Two selective JNK inhibitory peptides also produced a striking reduction of Aß toxicity. Finally, the dual inhibition of PKR and JNK nearly abolished Aß toxicity in primary cultured neurons. These results reveal that dual kinase inhibition can afford neuroprotection and this approach is worth being tested in in vivo AD and oxidative stress models.


Subject(s)
Amyloid beta-Peptides/toxicity , Cerebral Cortex/drug effects , Neurons/drug effects , Neuroprotection/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/enzymology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/enzymology , Neuroprotection/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology
12.
J Psychiatry Neurosci ; 40(3): 151-61, 2015 May.
Article in English | MEDLINE | ID: mdl-25455349

ABSTRACT

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.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Brain/enzymology , Brain/pathology , Mitogen-Activated Protein Kinase 10/metabolism , Aged , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Biomarkers/cerebrospinal fluid , Cohort Studies , Disease Progression , Female , Humans , Male , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Peptide Fragments/metabolism , Plaque, Amyloid/enzymology , Plaque, Amyloid/pathology , tau Proteins/cerebrospinal fluid
13.
Virus Res ; 193: 65-77, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25064266

ABSTRACT

The synthesis of proteins from viral mRNA is the first step towards viral assembly. Viruses are dependent upon the cellular translation machinery to synthesize their own proteins. The synthesis of proteins from the human immunodeficiency virus (HIV) type 1 and 2 RNAs utilize several alternative mechanisms. The regulation of viral protein production requires a constant interplay between viral requirements and the cell response to viral infection. Among the antiviral cell responses, the interferon-induced RNA activated protein kinase, PKR, regulates the cellular and viral translation. During HIV-1 infection, PKR activation is highly regulated by viral and cellular factors. The cellular TAR RNA Binding Protein, TRBP, the Adenosine Deaminase acting on RNA, ADAR1, and the PKR Activator, PACT, play important roles. Recent data show that PACT changes its function from activator to inhibitor in HIV-1 infected cells. Therefore, HIV-1 has evolved to replicate in cells in which TRBP, ADAR1 and PACT prevent PKR activation to allow efficient viral protein synthesis. This proper translation will initiate the assembly of viral particles.


Subject(s)
HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , RNA-Binding Proteins/metabolism , Virus Replication , eIF-2 Kinase/metabolism , HIV-2/physiology , Humans , Protein Biosynthesis , RNA, Viral , Signal Transduction
14.
J Virol ; 87(14): 8241-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23658442

ABSTRACT

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.


Subject(s)
Bluetongue virus/immunology , Immunity, Innate/immunology , Interferon Type I/antagonists & inhibitors , Signal Transduction/immunology , Viral Nonstructural Proteins/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunity, Innate/drug effects , Interferon Type I/biosynthesis , Luciferases , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Viral Nonstructural Proteins/pharmacology
15.
Viruses ; 4(11): 2598-635, 2012 Oct 29.
Article in English | MEDLINE | ID: mdl-23202496

ABSTRACT

The double-stranded RNA-dependent protein kinase PKR plays multiple roles in cells, in response to different stress situations. As a member of the interferon (IFN)­Stimulated Genes, PKR was initially recognized as an actor in the antiviral action of IFN, due to its ability to control translation, through phosphorylation, of the alpha subunit of eukaryotic initiation factor 2 (eIF2a). As such, PKR participates in the generation of stress granules, or autophagy and a number of viruses have designed strategies to inhibit its action. However, PKR deficient mice resist most viral infections, indicating that PKR may play other roles in the cell other than just acting as an antiviral agent. Indeed, PKR regulates several signaling pathways, either as an adapter protein and/or using its kinase activity. Here we review the role of PKR as an eIF2a kinase, its participation in the regulation of the NF-kB, p38MAPK and insulin pathways, and we focus on its role during infection with the hepatitis C virus (HCV). PKR binds the HCV IRES RNA, cooperates with some functions of the HCV core protein and may represent a target for NS5A or E2. Novel data points out for a role of PKR as a pro-HCV agent, both as an adapter protein and as an eIF2a-kinase, and in cooperation with the di-ubiquitin-like protein ISG15. Developing pharmaceutical inhibitors of PKR may help in resolving some viral infections as well as stress-related damages.


Subject(s)
Hepatitis C/metabolism , RNA, Double-Stranded/metabolism , Signal Transduction , Stress, Physiological , eIF-2 Kinase/metabolism , Animals , Enzyme Activation , Hepatitis C/genetics , Hepatitis C/immunology , Humans , Protein Transport , eIF-2 Kinase/genetics
16.
J Virol ; 86(10): 5817-28, 2012 May.
Article in English | MEDLINE | ID: mdl-22438548

ABSTRACT

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.


Subject(s)
Bluetongue virus/immunology , Bluetongue/immunology , Dendritic Cells/immunology , Interferon Type I/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology , Animals , Bluetongue/genetics , Bluetongue/virology , Bluetongue virus/genetics , Bluetongue virus/physiology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/virology , Female , Immunity, Innate , Interferon Type I/genetics , Membrane Glycoproteins , Myeloid Differentiation Factor 88/genetics , Receptors, Interleukin-1 , Sheep/immunology , Sheep/virology , Signal Transduction , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/genetics
17.
Brain Pathol ; 22(2): 219-29, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21790829

ABSTRACT

The neuropathological hallmarks of Alzheimer's disease (AD) include senile plaques made of Aß peptide, neurofibrillary tangles containing hyperphosphorylated tau protein and neuronal loss. The pro-apoptotic kinase PKR can be activated by Aß and can phosphorylate tau protein via GSK3ß kinase activation. The activated form of PKR (pPKR) accumulates in affected neurons and could participate in neuronal degeneration in AD. The mechanism of abnormal PKR activation in AD is not elucidated but could be linked to the PKR activator PACT. PACT stainings, and levels were assessed in the brains of AD patients and in APP/PS1 knock-in transgenic mice and in cell cultures exposed to stresses. We showed that PACT and pPKR colocalizations are enhanced in AD brains. Their levels are increased and correlated in AD and APP/PS1 knock-in mice brains. In human neuroblastoma cells exposed to Aß, tunicamycin or H2O2, PACT and pPKR concentrations are increased. PACT then PKR inhibitions indicate that PACT is upstream of PKR activation. Our findings demonstrate that PACT levels are enhanced in AD brains and could partly be caused by the action of Aß. In addition, PACT participates in PKR activation. The PACT-PKR pathway represents a potential link between Aß accumulation, PKR activation and tau phosphorylation.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , RNA-Binding Proteins/biosynthesis , eIF-2 Kinase/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Cell Line, Tumor , Enzyme Induction/physiology , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Phosphorylation , RNA-Binding Proteins/genetics , tau Proteins/metabolism
18.
PLoS Pathog ; 7(10): e1002289, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22022264

ABSTRACT

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.


Subject(s)
Cytokines/metabolism , Hepacivirus/immunology , Hepacivirus/metabolism , Interferons/biosynthesis , Receptors, Retinoic Acid/metabolism , Ubiquitins/metabolism , eIF-2 Kinase/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Profiling , Hepacivirus/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Humans , Interferon Regulatory Factor-3/biosynthesis , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferons/genetics , RNA Interference , RNA, Small Interfering , RNA, Viral/metabolism , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Signal Transduction , TNF Receptor-Associated Factor 3/biosynthesis , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Ubiquitination , Ubiquitins/biosynthesis , Ubiquitins/genetics , eIF-2 Kinase/biosynthesis , eIF-2 Kinase/genetics
19.
Cell Res ; 21(6): 895-910, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21200404

ABSTRACT

Recognition of viral RNA structures by the cytosolic sensor retinoic acid-inducible gene-I (RIG-I) results in the activation of signaling cascades that culminate with the generation of the type I interferon (IFN) antiviral response. Onset of antiviral and inflammatory responses to viral pathogens necessitates the regulated spatiotemporal recruitment of signaling adapters, kinases and transcriptional proteins to the mitochondrial antiviral signaling protein (MAVS). We previously demonstrated that the serine/threonine kinase IKKε is recruited to the C-terminal region of MAVS following Sendai or vesicular stomatitis virus (VSV) infection, mediated by Lys63-linked polyubiquitination of MAVS at Lys500, resulting in inhibition of downstream IFN signaling (Paz et al, Mol Cell Biol, 2009). In this study, we demonstrate that C-terminus of MAVS harbors a novel TRAF3-binding site in the aa450-468 region of MAVS. A consensus TRAF-interacting motif (TIM), 455-PEENEY-460, within this site is required for TRAF3 binding and activation of IFN antiviral response genes, whereas mutation of the TIM eliminates TRAF3 binding and the downstream IFN response. Reconstitution of MAVS(-/-) mouse embryo fibroblasts with a construct expressing a TIM-mutated version of MAVS failed to restore the antiviral response or block VSV replication, whereas wild-type MAVS reconstituted antiviral inhibition of VSV replication. Furthermore, recruitment of IKKε to an adjacent C-terminal site (aa 468-540) in MAVS via Lys500 ubiquitination decreased TRAF3 binding and protein stability, thus contributing to IKKε-mediated shutdown of the IFN response. This study demonstrates that MAVS harbors a functional C-terminal TRAF3-binding site that participates in positive and negative regulation of the IFN antiviral response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Feedback, Physiological , Immunity, Innate , Interferon Type I/metabolism , TNF Receptor-Associated Factor 3/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Gene Knockout Techniques , Humans , I-kappa B Kinase/metabolism , Interferon Type I/immunology , Mice , Mutagenesis, Site-Directed , Mutation, Missense , Peptide Fragments/metabolism , Protein Binding , Protein Stability , Protein Structure, Tertiary , Respirovirus Infections/immunology , Sendai virus/immunology , TNF Receptor-Associated Factor 3/immunology , Vesicular Stomatitis/immunology , Vesiculovirus/immunology
20.
PLoS One ; 5(5): e10575, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20485506

ABSTRACT

Hepatitis C virus is a poor inducer of interferon (IFN), although its structured viral RNA can bind the RNA helicase RIG-I, and activate the IFN-induction pathway. Low IFN induction has been attributed to HCV NS3/4A protease-mediated cleavage of the mitochondria-adapter MAVS. Here, we have investigated the early events of IFN induction upon HCV infection, using the cell-cultured HCV JFH1 strain and the new HCV-permissive hepatoma-derived Huh7.25.CD81 cell subclone. These cells depend on ectopic expression of the RIG-I ubiquitinating enzyme TRIM25 to induce IFN through the RIG-I/MAVS pathway. We observed induction of IFN during the first 12 hrs of HCV infection, after which a decline occurred which was more abrupt at the protein than at the RNA level, revealing a novel HCV-mediated control of IFN induction at the level of translation. The cellular protein kinase PKR is an important regulator of translation, through the phosphorylation of its substrate the eIF2alpha initiation factor. A comparison of the expression of luciferase placed under the control of an eIF2alpha-dependent (IRES(EMCV)) or independent (IRES(HCV)) RNA showed a specific HCV-mediated inhibition of eIF2alpha-dependent translation. We demonstrated that HCV infection triggers the phosphorylation of both PKR and eIF2alpha at 12 and 15 hrs post-infection. PKR silencing, as well as treatment with PKR pharmacological inhibitors, restored IFN induction in JFH1-infected cells, at least until 18 hrs post-infection, at which time a decrease in IFN expression could be attributed to NS3/4A-mediated MAVS cleavage. Importantly, both PKR silencing and PKR inhibitors led to inhibition of HCV yields in cells that express functional RIG-I/MAVS. In conclusion, here we provide the first evidence that HCV uses PKR to restrain its ability to induce IFN through the RIG-I/MAVS pathway. This opens up new possibilities to assay PKR chemical inhibitors for their potential to boost innate immunity in HCV infection.


Subject(s)
Hepacivirus/immunology , Interferons/biosynthesis , eIF-2 Kinase/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Eukaryotic Initiation Factor-2/metabolism , Hepacivirus/drug effects , Hepatitis C/immunology , Hepatitis C/virology , Humans , Kinetics , Models, Immunological , Phosphorylation/drug effects , Protein Biosynthesis , Protein Kinase Inhibitors/pharmacology , Substrate Specificity/drug effects , Time Factors , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism , eIF-2 Kinase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...