Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Immunity ; 46(2): 301-314, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228284

ABSTRACT

Respiratory syncytial virus (RSV) is the major cause of lower respiratory tract infections in infants and is characterized by pulmonary infiltration of B cells in fatal cases. We analyzed the B cell compartment in human newborns and identified a population of neonatal regulatory B lymphocytes (nBreg cells) that produced interleukin 10 (IL-10) in response to RSV infection. The polyreactive B cell receptor of nBreg cells interacted with RSV protein F and induced upregulation of chemokine receptor CX3CR1. CX3CR1 interacted with RSV glycoprotein G, leading to nBreg cell infection and IL-10 production that dampened T helper 1 (Th1) cytokine production. In the respiratory tract of neonates with severe RSV-induced acute bronchiolitis, RSV-infected nBreg cell frequencies correlated with increased viral load and decreased blood memory Th1 cell frequencies. Thus, the frequency of nBreg cells is predictive of the severity of acute bronchiolitis disease and nBreg cell activity may constitute an early-life host response that favors microbial pathogenesis.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Bronchiolitis, Viral/immunology , Receptors, Chemokine/immunology , Respiratory Syncytial Virus Infections/immunology , B-Lymphocytes, Regulatory/virology , Bronchiolitis, Viral/pathology , CD4-Positive T-Lymphocytes/immunology , CX3C Chemokine Receptor 1 , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Gene Expression Profiling , Humans , Infant, Newborn , Lymphocyte Activation/immunology , Oligonucleotide Array Sequence Analysis , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses , Transcriptome
2.
Eur J Immunol ; : e2350954, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38837415

ABSTRACT

Hexokinases (HKs) control the first step of glucose catabolism. A switch of expression from liver HK (glucokinase, GCK) to the tumor isoenzyme HK2 is observed in hepatocellular carcinoma progression. Our prior work revealed that HK isoenzyme switch in hepatocytes not only regulates hepatic metabolic functions but also modulates innate immunity and sensitivity to Natural Killer (NK) cell cytotoxicity. This study investigates the impact of HK2 expression and its mitochondrial binding on the resistance of human liver cancer cells to NK-cell-induced cytolysis. We have shown that HK2 expression induces resistance to NK cell cytotoxicity in a process requiring mitochondrial binding of HK2. Neither HK2 nor GCK expression affects target cells' ability to activate NK cells. In contrast, mitochondrial binding of HK2 reduces effector caspase 3/7 activity both at baseline and upon NK-cell activation. Furthermore, HK2 tethering to mitochondria enhances their resistance to cytochrome c release triggered by tBID. These findings indicate that HK2 mitochondrial binding in liver cancer cells is an intrinsic resistance factor to cytolysis and an escape mechanism from immune surveillance.

3.
Nature ; 574(7777): 259-263, 2019 10.
Article in English | MEDLINE | ID: mdl-31554973

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging alphavirus that is transmitted to humans by mosquito bites and causes musculoskeletal and joint pain1,2. Despite intensive investigations, the human cellular factors that are critical for CHIKV infection remain unknown, hampering the understanding of viral pathogenesis and the development of anti-CHIKV therapies. Here we identified the four-and-a-half LIM domain protein 1 (FHL1)3 as a host factor that is required for CHIKV permissiveness and pathogenesis in humans and mice. Ablation of FHL1 expression results in the inhibition of infection by several CHIKV strains and o'nyong-nyong virus, but not by other alphaviruses and flaviviruses. Conversely, expression of FHL1 promotes CHIKV infection in cells that do not normally express it. FHL1 interacts directly with the hypervariable domain of the nsP3 protein of CHIKV and is essential for the replication of viral RNA. FHL1 is highly expressed in CHIKV-target cells and is particularly abundant in muscles3,4. Dermal fibroblasts and muscle cells derived from patients with Emery-Dreifuss muscular dystrophy that lack functional FHL15 are resistant to CHIKV infection. Furthermore,  CHIKV infection  is undetectable in Fhl1-knockout mice. Overall, this study shows that FHL1 is a key factor expressed by the host that enables CHIKV infection and identifies the interaction between nsP3 and FHL1 as a promising target for the development of anti-CHIKV therapies.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Host-Derived Cellular Factors/metabolism , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Animals , Cells, Cultured , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Chikungunya virus/genetics , Chikungunya virus/growth & development , Female , Fibroblasts/virology , HEK293 Cells , Host-Derived Cellular Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/deficiency , LIM Domain Proteins/genetics , Male , Mice , Muscle Proteins/deficiency , Muscle Proteins/genetics , Myoblasts/virology , O'nyong-nyong Virus/growth & development , O'nyong-nyong Virus/pathogenicity , Protein Binding , RNA, Viral/biosynthesis , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
4.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33402530

ABSTRACT

The recent emergence and reemergence of viruses in the human population has highlighted the need to develop broader panels of therapeutic molecules. High-throughput screening assays opening access to untargeted steps of the viral replication cycle will provide powerful leverage to identify innovative antiviral molecules. We report here the development of an innovative protein complementation assay, termed αCentauri, to measure viral translocation between subcellular compartments. As a proof of concept, the Centauri fragment was either tethered to the nuclear pore complex or sequestered in the nucleus, while the complementary α fragment (<16 amino acids) was attached to the integrase proteins of infectious HIV-1. The translocation of viral ribonucleoproteins from the cytoplasm to the nuclear envelope or to the nucleoplasm efficiently reconstituted superfolder green fluorescent protein or NanoLuc αCentauri reporters. These fluorescence- or bioluminescence-based assays offer a robust readout of specific steps of viral infection in a multiwell format that is compatible for high-throughput screening and is validated by a short hairpin RNA-based prototype screen.


Subject(s)
High-Throughput Screening Assays/methods , Virus Diseases/metabolism , Virus Replication/physiology , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Green Fluorescent Proteins/metabolism , HIV Infections/metabolism , HeLa Cells , Humans , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Ribonucleoproteins/metabolism , Virus Replication/drug effects
5.
J Virol ; 96(7): e0196221, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35266803

ABSTRACT

Dengue virus (DENV) is a mosquito-borne flavivirus responsible for dengue disease, a major human health concern for which no effective treatment is available. DENV relies heavily on the host cellular machinery for productive infection. Here, we show that the scaffold protein RACK1, which is part of the DENV replication complex, mediates infection by binding to the 40S ribosomal subunit. Mass spectrometry analysis of RACK1 partners coupled to an RNA interference screen-identified Vigilin and SERBP1 as DENV host-dependency factors. Both are RNA-binding proteins that interact with the DENV genome. Genetic ablation of Vigilin or SERBP1 rendered cells poorly susceptible to DENV, as well as related flaviviruses, by hampering the translation and replication steps. Finally, we established that a Vigilin or SERBP1 mutant lacking RACK1 binding but still interacting with the viral RNA is unable to mediate DENV infection. We propose that RACK1 recruits Vigilin and SERBP1, linking the DENV genome to the translation machinery for efficient infection. IMPORTANCE We recently identified the scaffolding RACK1 protein as an important host-dependency factor for dengue virus (DENV), a positive-stranded RNA virus responsible for the most prevalent mosquito-borne viral disease worldwide. Here, we have performed the first RACK1 interactome in human cells and identified Vigilin and SERBP1 as DENV host-dependency factors. Both are RNA-binding proteins that interact with the DENV RNA to regulate viral replication. Importantly, Vigilin and SERBP1 interact with RACK1 and the DENV viral RNA (vRNA) to mediate viral replication. Overall, our results suggest that RACK1 acts as a binding platform at the surface of the 40S ribosomal subunit to recruit Vigilin and SERBP1, which may therefore function as linkers between the viral RNA and the translation machinery to facilitate infection.


Subject(s)
Dengue Virus , Dengue , RNA-Binding Proteins , Animals , Dengue/physiopathology , Dengue Virus/physiology , Host Microbial Interactions/physiology , Humans , Neoplasm Proteins/metabolism , RNA, Viral/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors for Activated C Kinase/metabolism , Virus Replication
6.
Vet Res ; 54(1): 18, 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36864517

ABSTRACT

Vaccination is one of the most widely used strategies to protect horses against pathogens. However, available equine vaccines often have limitations, as they do not always provide effective, long-term protection and booster injections are often required. In addition, research efforts are needed to develop effective vaccines against emerging equine pathogens. In this review, we provide an inventory of approved adjuvants for equine vaccines worldwide, and discuss their composition and mode of action when available. A wide range of adjuvants are used in marketed vaccines for horses, the main families being aluminium salts, emulsions, polymers, saponins and ISCOMs. We also present veterinary adjuvants that are already used for vaccination in other species and are currently evaluated in horses to improve equine vaccination and to meet the expected level of protection against pathogens in the equine industry. Finally, we discuss new adjuvants such as liposomes, polylactic acid polymers, inulin, poly-ε-caprolactone nanoparticles and co-polymers that are in development. Our objective is to help professionals in the horse industry understand the composition of marketed equine vaccines in a context of mistrust towards vaccines. Besides, this review provides researchers with a list of adjuvants, either approved or at least evaluated in horses, that could be used either alone or in combination to develop new vaccines.


Subject(s)
Adjuvants, Immunologic , Nanoparticles , Horses , Animals , Adjuvants, Immunologic/pharmacology , Vaccination/veterinary , Nanoparticles/therapeutic use , Polymers
7.
J Virol ; 95(22): e0091221, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34431698

ABSTRACT

Respiratory syncytial virus (RSV) is the main cause of acute respiratory infections in young children and also has a major impact on the elderly and immunocompromised people. In the absence of a vaccine or efficient treatment, a better understanding of RSV interactions with the host antiviral response during infection is needed. Previous studies revealed that cytoplasmic inclusion bodies (IBs), where viral replication and transcription occur, could play a major role in the control of innate immunity during infection by recruiting cellular proteins involved in the host antiviral response. We recently showed that the morphogenesis of IBs relies on a liquid-liquid-phase separation mechanism depending on the interaction between viral nucleoprotein (N) and phosphoprotein (P). These scaffold proteins are expected to play a central role in the recruitment of cellular proteins to IBs. Here, we performed a yeast two-hybrid screen using RSV N protein as bait and identified the cellular protein TAX1BP1 as a potential partner of this viral protein. This interaction was validated by pulldown and immunoprecipitation assays. We showed that TAX1BP1 suppression has only a limited impact on RSV infection in cell cultures. However, RSV replication is decreased in TAX1BP1-deficient (TAX1BP1 knockout [TAX1BP1KO]) mice, whereas the production of inflammatory and antiviral cytokines is enhanced. In vitro infection of wild-type or TAX1BP1KO alveolar macrophages confirmed that the innate immune response to RSV infection is enhanced in the absence of TAX1BP1. Altogether, our results suggest that RSV could hijack TAX1BP1 to restrain the host immune response during infection. IMPORTANCE Respiratory syncytial virus (RSV), which is the leading cause of lower respiratory tract illness in infants, remains a medical problem in the absence of a vaccine or efficient treatment. This virus is also recognized as a main pathogen in the elderly and immunocompromised people, and the occurrence of coinfections (with other respiratory viruses and bacteria) amplifies the risks of developing respiratory distress. In this context, a better understanding of the pathogenesis associated with viral respiratory infections, which depends on both viral replication and the host immune response, is needed. The present study reveals that the cellular protein TAX1BP1, which interacts with the RSV nucleoprotein N, participates in the control of the innate immune response during RSV infection, suggesting that the N-TAX1BP1 interaction represents a new target for the development of antivirals.


Subject(s)
Intracellular Signaling Peptides and Proteins/immunology , Neoplasm Proteins/immunology , Nucleocapsid Proteins/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Cell Line , Cricetinae , Humans , Immunity, Innate , Mice , Mice, Knockout , Virus Replication
8.
Proc Natl Acad Sci U S A ; 116(22): 10968-10977, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31076555

ABSTRACT

New therapeutic strategies targeting influenza are actively sought due to limitations in current drugs available. Host-directed therapy is an emerging concept to target host functions involved in pathogen life cycles and/or pathogenesis, rather than pathogen components themselves. From this perspective, we focused on an essential host partner of influenza viruses, the RED-SMU1 splicing complex. Here, we identified two synthetic molecules targeting an α-helix/groove interface essential for RED-SMU1 complex assembly. We solved the structure of the SMU1 N-terminal domain in complex with RED or bound to one of the molecules identified to disrupt this complex. We show that these compounds inhibiting RED-SMU1 interaction also decrease endogenous RED-SMU1 levels and inhibit viral mRNA splicing and viral multiplication, while preserving cell viability. Overall, our data demonstrate the potential of RED-SMU1 destabilizing molecules as an antiviral therapy that could be active against a wide range of influenza viruses and be less prone to drug resistance.


Subject(s)
Antiviral Agents/pharmacology , Chromosomal Proteins, Non-Histone/metabolism , Cytokines/metabolism , Orthomyxoviridae/drug effects , RNA Splicing Factors/metabolism , A549 Cells , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cytokines/chemistry , Cytokines/genetics , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Molecular Docking Simulation , Orthomyxoviridae/pathogenicity , Protein Binding/drug effects , Protein Stability/drug effects , RNA Splicing , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , Spliceosomes/drug effects
9.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055105

ABSTRACT

Hepatitis C virus (HCV) relies on cellular lipid metabolism for its replication, and actively modulates lipogenesis and lipid trafficking in infected hepatocytes. This translates into an intracellular accumulation of triglycerides leading to liver steatosis, cirrhosis and hepatocellular carcinoma, which are hallmarks of HCV pathogenesis. While the interaction of HCV with hepatocyte metabolic pathways is patent, how viral proteins are able to redirect central carbon metabolism towards lipogenesis is unclear. Here, we report that the HCV protein NS5A activates the glucokinase (GCK) isoenzyme of hexokinases through its D2 domain (NS5A-D2). GCK is the first rate-limiting enzyme of glycolysis in normal hepatocytes whose expression is replaced by the hexokinase 2 (HK2) isoenzyme in hepatocellular carcinoma cell lines. We took advantage of a unique cellular model specifically engineered to re-express GCK instead of HK2 in the Huh7 cell line to evaluate the consequences of NS5A-D2 expression on central carbon and lipid metabolism. NS5A-D2 increased glucose consumption but decreased glycogen storage. This was accompanied by an altered mitochondrial respiration, an accumulation of intracellular triglycerides and an increased production of very-low density lipoproteins. Altogether, our results show that NS5A-D2 can reprogram central carbon metabolism towards a more energetic and glycolytic phenotype compatible with HCV needs for replication.


Subject(s)
Glucokinase/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatocytes/metabolism , Hepatocytes/virology , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , Glycogen/metabolism , Glycolysis , Host-Pathogen Interactions , Humans , Lipid Metabolism , Lipogenesis , Mitochondria/metabolism , Protein Binding , Protein Interaction Domains and Motifs , RNA-Dependent RNA Polymerase/chemistry , Viral Nonstructural Proteins/chemistry
10.
RNA Biol ; 18(5): 732-744, 2021 05.
Article in English | MEDLINE | ID: mdl-33459164

ABSTRACT

Flaviviruses such as the dengue (DENV) and the Zika virus (ZIKV) are important human pathogens causing around 100 million symptomatic infections each year. During infection, small subgenomic flavivirus RNAs (sfRNAs) are formed inside the infected host cell as a result of incomplete degradation of the viral RNA genome by cellular exoribonuclease XRN1. Although the full extent of sfRNA functions is to be revealed, these non-coding RNAs are key virulence factors and their detrimental effects on multiple cellular processes seem to consistently involve molecular interactions with RNA-binding proteins (RBPs). Discovery of such sfRNA-binding host-factors has followed established biochemical pull-down approaches skewed towards highly abundant proteins hampering proteome-wide coverage. Yeast three-hybrid (Y3H) systems represent an attractive alternative approach. To facilitate proteome-wide screens for RBP, we revisited and improved existing RNA-Y3H methodology by (1) implementing full-length ORF libraries in combination with (2) efficient yeast mating to increase screening depth and sensitivity, and (3) stringent negative controls to eliminate over-representation of non-specific RNA-binders. These improvements were validated employing the well-characterized interaction between DDX6 (DEAD-box helicase 6) and sfRNA of DENV as paradigm. Our advanced Y3H system was used to screen for human proteins binding to DENV and ZIKV sfRNA, resulting in a list of 69 putative sfRNA-binders, including several previously reported as well as numerous novel RBP host factors. Our methodology requiring no sophisticated infrastructure or analytic pipeline may be employed for the discovery of meaningful RNA-protein interactions at large scale in other fields.


Subject(s)
Host-Pathogen Interactions , Protein Interaction Maps , RNA, Viral/metabolism , RNA-Binding Proteins/isolation & purification , Cells, Cultured , Dengue/genetics , Dengue/metabolism , Dengue Virus/genetics , Genome, Human , Humans , Open Reading Frames/genetics , Organisms, Genetically Modified , Protein Binding , RNA Stability , RNA, Viral/genetics , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae , Two-Hybrid System Techniques , Zika Virus/genetics , Zika Virus Infection/genetics , Zika Virus Infection/metabolism
11.
Int J Mol Sci ; 22(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669407

ABSTRACT

La Reunion island in the South West Indian Ocean is now endemic for dengue following the introduction of dengue virus serotype 2 (DENV-2) cosmopolitan-I genotype in 2017. DENV-2 infection causes a wide spectrum of clinical manifestations ranging from flu-like disease to severe dengue. The nonstructural glycoprotein 1 (NS1) has been identified as playing a key role in dengue disease severity. The intracellular NS1 exists as a homodimer, whereas a fraction is driven towards the plasma membrane or released as a soluble hexameric protein. Here, we characterized the NS1 glycoproteins from clinical isolates DES-14 and RUN-18 that were collected during the DENV-2 epidemics in Tanzania in 2014 and La Reunion island in 2018, respectively. In relation to hepatotropism of the DENV, expression of recombinant DES-14 NS1 and RUN-18 NS1 glycoproteins was compared in human hepatoma Huh7 cells. We observed that RUN-18 NS1 was poorly stable in Huh7 cells compared to DES-14 NS1. The instability of RUN-18 NS1 leading to a low level of NS1 secretion mostly relates to lysine residues on positions 272 and 324. Our data raise the issue of the consequences of a defect in NS1 stability in human hepatocytes in relation to the major role of NS1 in the pathogenesis of the DENV-2 infection.


Subject(s)
Dengue Virus/metabolism , Dengue/epidemiology , Dengue/metabolism , Epidemics , Genotype , Lysine/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Substitution , Antigens, Viral/chemistry , Antigens, Viral/genetics , Cell Line, Tumor , Dengue/virology , HEK293 Cells , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Protein Multimerization , Protein Stability , Recombinant Proteins/chemistry , Reunion/epidemiology , Serogroup , Tanzania/epidemiology , Transfection , Viral Nonstructural Proteins/genetics
12.
J Transl Med ; 18(1): 319, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32811513

ABSTRACT

In less than 20 years, three deadly coronaviruses, SARS-CoV, MERS-CoV and SARS-CoV-2, have emerged in human population causing hundreds to hundreds of thousands of deaths. Other coronaviruses are causing epizootic representing a significant threat for both domestic and wild animals. Members of this viral family have the longest genome of all RNA viruses, and express up to 29 proteins establishing complex interactions with the host proteome. Deciphering these interactions is essential to identify cellular pathways hijacked by these viruses to replicate and escape innate immunity. Virus-host interactions also provide key information to select targets for antiviral drug development. Here, we have manually curated the literature to assemble a unique dataset of 1311 coronavirus-host protein-protein interactions. Functional enrichment and network-based analyses showed coronavirus connections to RNA processing and translation, DNA damage and pathogen sensing, interferon production, and metabolic pathways. In particular, this global analysis pinpointed overlooked interactions with translation modulators (GIGYF2-EIF4E2), components of the nuclear pore, proteins involved in mitochondria homeostasis (PHB, PHB2, STOML2), and methylation pathways (MAT2A/B). Finally, interactome data provided a rational for the antiviral activity of some drugs inhibiting coronaviruses replication. Altogether, this work describing the current landscape of coronavirus-host interactions provides valuable hints for understanding the pathophysiology of coronavirus infections and developing effective antiviral therapies.


Subject(s)
Coronavirus Infections/metabolism , Coronavirus/metabolism , Host-Pathogen Interactions/physiology , Protein Interaction Maps , Viral Proteins/metabolism , Animals , Betacoronavirus/physiology , COVID-19 , Coronavirus/chemistry , Coronavirus Infections/virology , Databases, Protein , Humans , Mitochondrial Proteins/metabolism , Pandemics , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Prohibitins , SARS-CoV-2 , Transcription Factors/metabolism , Virus Replication/genetics
13.
Virologie (Montrouge) ; 22(1): 1-13, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-33111665

ABSTRACT

Environmental pollution is of concern to civil society and as the problem intensifies, there is increasing pressure on politicians and polluters to assess and mitigate this risk. In addition, the emergence (or re-emergence) of viral pathologies such as dengue or chikungunya has also become a major concern requiring appropriate measures. Unfortunately, these two issues may well collide with unpredictable consequences in the next decades. Indeed, a growing number of studies suggests that organic pollutants could alter the innate antiviral response, including the type I interferon system (IFN-I). Such interactions could have significant consequences on the susceptibility of populations to viral infections, but also modify responses and protection induced by vaccines or favor the development of autoimmune diseases. The purpose of this review is to take stock of the known interactions between organic pollutants and the IFN-I response, and to present questions that should be addressed in the future in order to better assess this risk.

14.
Virologie (Montrouge) ; 22(1): 27-40, 2018 02 01.
Article in French | MEDLINE | ID: mdl-33111666

ABSTRACT

Environmental pollution is of concern to civil society and as the problem intensifies, there is increasing pressure on politicians and polluters to assess and mitigate this risk. In addition, the emergence (or re-emergence) of viral pathologies such as dengue or chikungunya has also become a major concern requiring appropriate measures. Unfortunately, these two issues may well collide with unpredictable consequences in the next decades. Indeed, a growing number of studies suggests that organic pollutants could alter the innate antiviral response, including the type I interferon system (IFN-I). Such interactions could have significant consequences on the susceptibility of populations to viral infections, but also modify responses and protection induced by vaccines or favor the development of autoimmune diseases. The purpose of this review is to take stock of the known interactions between organic pollutants and the IFN-I response, and to present questions that should be addressed in the future in order to better assess this risk.

15.
Article in English | MEDLINE | ID: mdl-28807907

ABSTRACT

De novo pyrimidine biosynthesis is a key metabolic pathway involved in multiple biosynthetic processes. Here, we identified an original series of 3-(1H-indol-3-yl)-2,3-dihydro-4H-furo[3,2-c]chromen-4-one derivatives as a new class of pyrimidine biosynthesis inhibitors formed by two edge-fused polycyclic moieties. We show that identified compounds exhibit broad-spectrum antiviral activity and immunostimulatory properties, in line with recent reports linking de novo pyrimidine biosynthesis with innate defense mechanisms against viruses. Most importantly, we establish that pyrimidine deprivation can amplify the production of both type I and type III interferons by cells stimulated with retinoic acid-inducible gene 1 (RIG-I) ligands. Altogether, our results further expand the current panel of pyrimidine biosynthesis inhibitors and illustrate how the production of antiviral interferons is tightly coupled to this metabolic pathway. Functional and structural similarities between this new chemical series and dicoumarol, which was reported before to inhibit pyrimidine biosynthesis at the dihydroorotate dehydrogenase (DHODH) step, are discussed.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya virus/immunology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Interferon Type I/biosynthesis , Interferons/biosynthesis , Measles virus/immunology , Pyrimidines/biosynthesis , Antiviral Agents/chemistry , Cell Line , Chromones/chemistry , Dicumarol/pharmacology , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Immunity, Innate/immunology , Indoles/chemistry , Interferon Type I/immunology , Interferons/immunology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Structure-Activity Relationship , Interferon Lambda
16.
J Biol Chem ; 290(8): 4631-4646, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25505242

ABSTRACT

After cell entry, HIV undergoes rapid transport toward the nucleus using microtubules and microfilaments. Neither the cellular cytoplasmic components nor the viral proteins that interact to mediate transport have yet been identified. Using a yeast two-hybrid screen, we identified four cytoskeletal components as putative interaction partners for HIV-1 p24 capsid protein: MAP1A, MAP1S, CKAP1, and WIRE. Depletion of MAP1A/MAP1S in indicator cell lines and primary human macrophages led to a profound reduction in HIV-1 infectivity as a result of impaired retrograde trafficking, demonstrated by a characteristic accumulation of capsids away from the nuclear membrane, and an overall defect in nuclear import. MAP1A/MAP1S did not impact microtubule network integrity or cell morphology but contributed to microtubule stabilization, which was shown previously to facilitate infection. In addition, we found that MAP1 proteins interact with HIV-1 cores both in vitro and in infected cells and that interaction involves MAP1 light chain LC2. Depletion of MAP1 proteins reduced the association of HIV-1 capsids with both dynamic and stable microtubules, suggesting that MAP1 proteins help tether incoming viral capsids to the microtubular network, thus promoting cytoplasmic trafficking. This work shows for the first time that following entry into target cells, HIV-1 interacts with the cytoskeleton via its p24 capsid protein. Moreover, our results support a role for MAP1 proteins in promoting efficient retrograde trafficking of HIV-1 by stimulating the formation of stable microtubules and mediating the association of HIV-1 cores with microtubules.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , HIV-1/metabolism , Macrophages/metabolism , Microtubule-Associated Proteins/metabolism , Active Transport, Cell Nucleus/genetics , Carrier Proteins/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/virology , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , HIV-1/genetics , Humans , Macrophages/pathology , Macrophages/virology , Microfilament Proteins , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Microtubules/pathology
17.
J Gen Virol ; 97(9): 2194-2200, 2016 09.
Article in English | MEDLINE | ID: mdl-27405867

ABSTRACT

Poliovirus (PV)-induced apoptosis seems to play a major role in central nervous system (CNS) tissue injury, a crucial feature of the pathogenesis of poliomyelitis. We have previously shown that calcium (Ca2+) flux from the endoplasmic reticulum (ER) to the cytosol during PV infection is involved in apoptosis induction in human neuroblastoma cells. We show here that PV infection is associated with a transient upregulation of Herp (homocysteine-induced ER protein), a protein known to promote the degradation of ER-resident Ca2+ channels. Herp gene transcription is controlled by the transcription factor CREB3 (cAMP response element-binding protein 3). We found that the CREB3/Herp pathway limited the increase in cytosolic Ca2+ concentration and apoptosis early in PV infection. This may reduce the extent of PV-induced damage to the CNS during poliomyelitis.


Subject(s)
Apoptosis , Calcium/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Host-Pathogen Interactions , Membrane Proteins/metabolism , Poliovirus/immunology , Poliovirus/pathogenicity , Cell Line , Humans , Neurons/immunology , Neurons/metabolism , Neurons/virology , Signal Transduction
18.
J Virol ; 89(22): 11233-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26311883

ABSTRACT

UNLABELLED: To date, the majority of work on RNA virus replication fidelity has focused on the viral RNA polymerase, while the potential role of other viral replicase proteins in this process is poorly understood. Previous studies used resistance to broad-spectrum RNA mutagens, such as ribavirin, to identify polymerases with increased fidelity that avoid misincorporation of such base analogues. We identified a novel variant in the alphavirus viral helicase/protease, nonstructural protein 2 (nsP2) that operates in concert with the viral polymerase nsP4 to further alter replication complex fidelity, a functional linkage that was conserved among the alphavirus genus. Purified chikungunya virus nsP2 presented delayed helicase activity of the high-fidelity enzyme, and yet purified replication complexes manifested stronger RNA polymerization kinetics. Because mutagenic nucleoside analogs such as ribavirin also affect intracellular nucleotide pools, we addressed the link between nucleotide depletion and replication fidelity by using purine and pyrimidine biosynthesis inhibitors. High-fidelity viruses were more resistant to these conditions, and viral growth could be rescued by the addition of exogenous nucleosides, suggesting that mutagenesis by base analogues requires nucleotide pool depletion. This study describes a novel function for nsP2, highlighting the role of other components of the replication complex in regulating viral replication fidelity, and suggests that viruses can alter their replication complex fidelity to overcome intracellular nucleotide-depleting conditions. IMPORTANCE: Previous studies using the RNA mutagen ribavirin to select for drug-resistant variants have highlighted the essential role of the viral RNA-dependent RNA polymerase in regulating replication fidelity. However, the role of other viral replicase components in replication fidelity has not been studied in detail. We identified here an RNA mutagen-resistant variant of the nsP2 helicase/protease that conferred increased fidelity and yet could not operate in the same manner as high-fidelity polymerases. We show that the alphavirus helicase is a key component of the fidelity-regulating machinery. Our data show that the RNA mutagenic activity of compounds such as ribavirin is coupled to and potentiated by nucleotide depletion and that RNA viruses can fine-tune their replication fidelity when faced with an intracellular environment depleted of nucleotides.


Subject(s)
Chikungunya virus/physiology , Cysteine Endopeptidases/metabolism , RNA Helicases/metabolism , RNA-Dependent RNA Polymerase/metabolism , Virus Replication/physiology , Animals , Antiviral Agents/pharmacology , Base Sequence , Cell Line , Chikungunya virus/genetics , Chlorocebus aethiops , Cricetinae , Cysteine Endopeptidases/genetics , DNA Replication/drug effects , HeLa Cells , Humans , Mutation/genetics , Nucleotides/deficiency , Purines/biosynthesis , Pyrimidines/biosynthesis , RNA Helicases/genetics , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/genetics , Ribavirin/pharmacology , Sequence Analysis, RNA , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Vero Cells , Virus Replication/genetics
19.
PLoS Pathog ; 10(6): e1004164, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24945353

ABSTRACT

Influenza A viruses are major pathogens in humans and in animals, whose genome consists of eight single-stranded RNA segments of negative polarity. Viral mRNAs are synthesized by the viral RNA-dependent RNA polymerase in the nucleus of infected cells, in close association with the cellular transcriptional machinery. Two proteins essential for viral multiplication, the exportin NS2/NEP and the ion channel protein M2, are produced by splicing of the NS1 and M1 mRNAs, respectively. Here we identify two human spliceosomal factors, RED and SMU1, that control the expression of NS2/NEP and are required for efficient viral multiplication. We provide several lines of evidence that in infected cells, the hetero-trimeric viral polymerase recruits a complex formed by RED and SMU1 through interaction with its PB2 and PB1 subunits. We demonstrate that the splicing of the NS1 viral mRNA is specifically affected in cells depleted of RED or SMU1, leading to a decreased production of the spliced mRNA species NS2, and to a reduced NS2/NS1 protein ratio. In agreement with the exportin function of NS2, these defects impair the transport of newly synthesized viral ribonucleoproteins from the nucleus to the cytoplasm, and strongly reduce the production of infectious influenza virions. Overall, our results unravel a new mechanism of viral subversion of the cellular splicing machinery, by establishing that the human splicing factors RED and SMU1 act jointly as key regulators of influenza virus gene expression. In addition, our data point to a central role of the viral RNA polymerase in coupling transcription and alternative splicing of the viral mRNAs.


Subject(s)
Alternative Splicing , Chromosomal Proteins, Non-Histone/metabolism , Cytokines/metabolism , Influenza A virus/physiology , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Active Transport, Cell Nucleus , Cell Line , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cytokines/antagonists & inhibitors , Cytokines/chemistry , Cytokines/genetics , Gene Silencing , Host-Pathogen Interactions , Humans , Influenza A virus/enzymology , Karyopherins/genetics , Karyopherins/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , RNA-Dependent RNA Polymerase/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Spliceosomes/enzymology , Spliceosomes/metabolism , Two-Hybrid System Techniques , Viral Proteins/genetics , Virus Replication
20.
J Gen Virol ; 96(Pt 4): 782-792, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25537374

ABSTRACT

There is no large-scale therapy available against human respiratory syncytial virus (hRSV), a major pathogen responsible for acute respiratory diseases. Macaques represent an interesting animal model to evaluate potential treatments because of their genetic, anatomical and immunological proximity with humans. However, the parameters that influence hRSV growth and control in this model are still poorly understood. We have documented in the following study the influence of age as well as repeated infections on the virological, clinical and immunological parameters of this animal model. Following intranasal inoculation, hRSV replicated in the upper respiratory tract for less than 15 days with no clinical signs regardless of age. Interestingly, we observed the induction of a local immune response at the nasal mucosa as assessed by expression profiles of inflammatory and IFN-stimulated genes. Animals also developed specific antibodies and were immune to reinfection. Thus, we showed that even in infant macaques, intranasal hRSV infection induced both local and systemic immune responses to efficiently control the virus.


Subject(s)
Macaca fascicularis/immunology , Nasal Mucosa/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Respiratory Tract Infections/immunology , Administration, Intranasal/methods , Age Factors , Animals , Antibodies, Viral/immunology , Cell Line , Disease Models, Animal , Female , Humans , Interferons/immunology , Macaca fascicularis/virology , Male , Nasal Mucosa/virology , Pregnancy , Respiratory Syncytial Virus Infections/virology , Respiratory Tract Infections/virology , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL