Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
1.
Cell Death Discov ; 10(1): 298, 2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38909032

RÉSUMÉ

LIMA1 is a LIM domain and Actin binding 1 protein that acts as a skeleton protein to promote cholesterol absorption, which makes it an ideal target for interfering with lipid metabolism. However, the detailed regulation of LIMA1 remains unclear. Here, we identified that ring finger protein 40 (RNF40), an E3 ubiquitin ligase previously known as an epigenetic modifier to increase H2B ubiquitination, mediated the ubiquitination of LIMA1 and thereby promoted its degradation in a proteasome-dependent manner. Fraction studies revealed that the 1-166aa fragment of LIMA1 was indispensable for the interaction with RNF40, and at least two domains of RNF40 might mediate the association of RNF40 with LIMA1. Notably, treatment with simvastatin dramatically decreased the levels of CHO and TG in control cells rather than cells with overexpressed LIMA1. Moreover, RNF40 significantly decreased lipid content, which could be reversed by LIMA1 overexpression. These findings suggest that E3 ubiquitin ligase RNF40 could directly target LIMA1 and promote its protein degradation in cytoplasm, leading to the suppression of lipid accumulation mediated by LIMA1. Collectively, this study unveils that RNF40 is a novel E3 ubiquitin ligase of LIMA1, which underpins its high therapeutic value to combat dysregulation of lipid metabolism.

2.
Viruses ; 16(4)2024 04 17.
Article de Anglais | MEDLINE | ID: mdl-38675963

RÉSUMÉ

Southern Africa Territories 2 (SAT2) foot-and-mouth disease (FMD) has crossed long-standing regional boundaries in recent years and entered the Middle East. However, the existing vaccines offer poor cross-protection against the circulating strains in the field. Therefore, there is an urgent need for an alternative design approach for vaccines in anticipation of a pandemic of SAT2 Foot-and-mouth disease virus (FMDV). The porcine parvovirus (PPV) VP2 protein can embed exogenous epitopes into the four loops on its surface, assemble into virus-like particles (VLPs), and induce antibodies and cytokines to PPV and the exogenous epitope. In this study, chimeric porcine parvovirus VP2 VLPs (chimeric PPV-SAT2-VLPs) expressing the T-and/or B-cell epitopes of the structural protein VP1 of FMDV SAT2 were produced using the recombinant pFastBac™ Dual vector of baculoviruses in Sf9 and HF cells We used the Bac-to-Bac system to construct the recombinant baculoviruses. The VP2-VLP--SAT2 chimeras displayed chimeric T-cell epitope (amino acids 21-40 of VP1) and/or the B-cell epitope (amino acids 135-174) of SAT FMDV VP1 by substitution of the corresponding regions at the N terminus (amino acids 2-23) and/or loop 2 and/or loop 4 of the PPV VP2 protein, respectively. In mice, the chimeric PPV-SAT2-VLPs induced specific antibodies against PPV and the VP1 protein of SAT2 FMDV. The VP2-VLP-SAT2 chimeras induced specific antibodies to PPV and the VP1 protein specific epitopes of FMDV SAT2. In this study, as a proof-of-concept, successfully generated chimeric PPV-VP2 VLPs expressing epitopes of the structural protein VP1 of FMDV SAT2 that has a potential to prevent FMDV SAT2 and PPV infection in pigs.


Sujet(s)
Anticorps antiviraux , Antigènes viraux , Protéines de capside , Virus de la fièvre aphteuse , Fièvre aphteuse , Parvovirus porcin , Vaccins à pseudo-particules virales , Vaccins antiviraux , Animaux , Virus de la fièvre aphteuse/immunologie , Virus de la fièvre aphteuse/génétique , Souris , Fièvre aphteuse/immunologie , Fièvre aphteuse/prévention et contrôle , Fièvre aphteuse/virologie , Protéines de capside/immunologie , Protéines de capside/génétique , Parvovirus porcin/immunologie , Parvovirus porcin/génétique , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Vaccins à pseudo-particules virales/immunologie , Vaccins à pseudo-particules virales/génétique , Suidae , Immunité humorale , Immunité cellulaire , Déterminants antigéniques des lymphocytes T/immunologie , Déterminants antigéniques des lymphocytes T/génétique , Déterminants antigéniques des lymphocytes B/immunologie , Déterminants antigéniques des lymphocytes B/génétique , Sérogroupe , Souris de lignée BALB C , Femelle , Épitopes/immunologie , Épitopes/génétique , Cellules Sf9 , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang
3.
Viruses ; 16(3)2024 03 13.
Article de Anglais | MEDLINE | ID: mdl-38543813

RÉSUMÉ

African swine fever (ASF) is a highly contagious and hemorrhagic disease caused by infection with the African swine fever virus (ASFV), resulting in a mortality rate of up to 100%. Currently, there are no effective treatments and commercially available vaccines for ASF. Therefore, it is crucial to identify biochemicals derived from host cells that can impede ASFV replication, with the aim of preventing and controlling ASF. The ASFV is an acellular organism that promotes self-replication by hijacking the metabolic machinery and biochemical resources of host cells. ASFV specifically alters the utilization of glucose and glutamine, which are the primary metabolic sources in mammalian cells. This study aimed to investigate the impact of glucose and glutamine metabolic dynamics on the rate of ASFV replication. Our findings demonstrate that ASFV infection favors using glutamine as a metabolic fuel to facilitate self-replication. ASFV replication can be substantially inhibited by blocking glutamine metabolism. The metabolomics analysis of the host cell after late-stage ASFV infection revealed a significant disruption of normal glutamine metabolic pathways due to the abundant expression of PLA (phenyllactic acid). Pretreatment with PLA also inhibited ASFV proliferation and glutamine consumption following infection. The metabolomic analysis also showed that PLA pretreatment greatly slowed down the metabolism of amino acids and nucleotides that depend on glutamine. The depletion of these building blocks directly hindered the replication of ASFV by decreasing the biosynthetic precursors produced during the replication of ASFV's progeny virus. These findings provide valuable insight into the possibility of pursuing the development of antiviral drugs against ASFV that selectively target metabolic pathways.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Lactates , Suidae , Animaux , Glutamine , Glucose , Polyesters/pharmacologie , Réplication virale , Mammifères
4.
J Virol ; 97(8): e0058623, 2023 08 31.
Article de Anglais | MEDLINE | ID: mdl-37582206

RÉSUMÉ

African swine fever (ASF) is a devastating disease caused by the African swine fever virus (ASFV) that adversely affects the pig industry. The spleen is the main target organ of ASFV; however, the function of metabolites in the spleen during ASFV infection is yet to be investigated. To define the metabolic changes in the spleen after ASFV infection, untargeted and targeted metabolomics analyses of spleens from ASFV-infected pigs were conducted. Untargeted metabolomics analysis revealed 540 metabolites with significant differential levels. Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that these metabolites were mainly enriched in metabolic pathways, including nucleotide metabolism, purine metabolism, arginine biosynthesis, and neuroactive ligand-receptor interaction. Moreover, 134 of 540 metabolites quantified by targeted metabolomics analysis had differential levels and were enriched in metabolic pathways such as the biosynthesis of cofactors, ABC transporters, and biosynthesis of amino acids. Furthermore, coalition analysis of untargeted and targeted metabolomics data revealed that the levels of acylcarnitines, which are intermediates of fatty acid ß-oxidation, were significantly increased in ASFV-infected spleens compared with those in the uninfected spleens. Moreover, inhibiting fatty acid ß-oxidation significantly reduced ASFV replication, indicating that fatty acid ß-oxidation is essential for this process. To our knowledge, this is the first report presenting the metabolite profiles of ASFV-infected pigs. This study revealed a new mechanism of ASFV-mediated regulation of host metabolism. These findings provide new insights into the pathogenic mechanisms of ASFV, which will benefit the development of target drugs for ASFV replication. IMPORTANCE African swine fever virus, the only member of the Asfarviridae family, relies on hijacking host metabolism to meet the demand for self-replication. However, the change in host metabolism after African swine fever virus (ASFV) infection remains unknown. Here, we analyzed the metabolic changes in the pig spleen after ASFV infection for the first time. ASFV infection increased the levels of acylcarnitines. Inhibition of the production and metabolism of acylcarnitines inhibited ASFV replication. Acylcarnitines are the vital intermediates of fatty acid ß-oxidation. This study highlights the critical role of fatty acid ß-oxidation in ASFV infection, which may help identify target drugs to control African swine fever disease.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Carnitine , Rate , Réplication virale , Animaux , Virus de la peste porcine africaine/physiologie , Acides gras/métabolisme , Métabolomique , Rate/métabolisme , Suidae , Carnitine/analyse
5.
J Immunol ; 210(3): 283-296, 2023 02 01.
Article de Anglais | MEDLINE | ID: mdl-36548461

RÉSUMÉ

Foot-and-mouth disease virus (FMDV) is the causative agent of foot-and-mouth disease, one of the most highly infectious animal viruses throughout the world. The JAK-STAT signaling pathway is a highly conserved pathway for IFN-ß-induced antiviral gene expression. Previous studies have shown that FMDV can strongly suppress the innate immune response. Moreover, although STAT1 and STAT2 (STAT1/2) have been well established in JAK-STAT signaling-induced antiviral gene expression, whether FMDV proteins inhibit IFN-ß-induced JAK-STAT signaling remains poorly understood. In this study, we described the Lb leader protease (Lbpro) of FMDV as a candidate for inhibiting IFN-ß-induced signaling transduction via directly interacting with STAT1/2. We further showed that Lbpro colocalized with STAT1/2 to inhibit their nuclear translocation. Importantly, Lbpro cleaved STAT1/2 to inhibit IFN-ß-induced signal transduction, whereas the catalytically inactive mutant of LC51A (Lbpro with cysteine substituted with alanine at amino acid residue 51) had no effect on the stability of STAT1/2 proteins. The cleavage of the STAT1/2 proteins was also determined during FMDV infection in vitro. Lbpro could cleave the residues between 252 and 502 aa for STAT1 and the site spanning residues 140 - 150 aa (QQHEIESRIL) for STAT2. The in vivo results showed that Lbpro can cleave STAT1/2 in pigs. Overall, our findings suggest that FMDV Lbpro-mediated targeting of STAT1/2 may reveal a novel mechanism for viral immune evasion.


Sujet(s)
Endopeptidases , Virus de la fièvre aphteuse , Interféron bêta , Facteur de transcription STAT-1 , Facteur de transcription STAT-2 , Animaux , Virus de la fièvre aphteuse/enzymologie , Immunité innée , Peptide hydrolases , Transduction du signal , Suidae , Interféron bêta/immunologie
6.
J Virol ; 96(12): e0031722, 2022 06 22.
Article de Anglais | MEDLINE | ID: mdl-35604142

RÉSUMÉ

The RIG-I-like receptor signaling pathway is crucial for producing type I interferon (IFN-I) against RNA viruses. The present study observed that viral infection increased annexin-A1 (ANXA1) expression, and ANXA1 then promoted RNA virus-induced IFN-I production. Compared to ANXA1 wild-type cells, ANXA1-/- knockout cells showed IFN-ß production decreasing after viral stimulation. RNA virus stimulation induced ANXA1 to regulate IFN-ß production through the TBK1-IRF3 axis but not through the NF-κB axis. ANXA1 also interacted with JAK1 and STAT1 to increase signal transduction induced by IFN-ß or IFN-γ. We assessed the effect of ANXA1 on the replication of foot-and-mouth disease virus (FMDV) and found that ANXA1 inhibits FMDV replication dependent on IFN-I production. FMDV 3A plays critical roles in viral replication and host range. The results showed that FMDV 3A interacts with ANXA1 to inhibit its ability to promote IFN-ß production. We also demonstrated that FMDV 3A inhibits the formation of ANXA1-TBK1 complex. These results indicate that ANXA1 positively regulates RNA virus-stimulated IFN-ß production and FMDV 3A antagonizes ANXA1-promoted IFN-ß production to modulate viral replication. IMPORTANCE FMDV is a pathogen that causes one of the world's most destructive and highly contagious animal diseases. The FMDV 3A protein plays a critical role in viral replication and host range. Although 3A is one of the viral proteins that influences FMDV virulence, its underlying mechanisms remain unclear. ANXA1 is involved in immune activation against pathogens. The present study demonstrated that FMDV increases ANXA1 expression, while ANXA1 inhibits FMDV replication. The results also showed that ANXA1 promotes RNA virus-induced IFN-I production through the IRF3 axis at VISA and TBK1 levels. ANXA1 was also found to interact with JAK1 and STAT1 to strengthen signal transduction induced by IFN-ß and IFN-γ. 3A interacted with ANXA1 to inhibit ANXA1-TBK1 complex formation, thereby antagonizing the inhibitory effect of ANXA1 on FMDV replication. This study helps to elucidate the mechanism underlying the effect of the 3A protein on FMDV replication.


Sujet(s)
Annexine A1 , Virus de la fièvre aphteuse , Réplication virale , Animaux , Annexine A1/métabolisme , Virus de la fièvre aphteuse/génétique , Virus de la fièvre aphteuse/métabolisme , Virus de la fièvre aphteuse/physiologie , Interactions hôte-pathogène , Facteur-3 de régulation d'interféron , Interféron bêta/métabolisme , Interféron gamma , Janus kinase 1/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Facteur de transcription STAT-1/métabolisme
7.
PLoS Negl Trop Dis ; 16(5): e0010435, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35639780

RÉSUMÉ

Almost all Echinococcus multilocularis (Em) infections occur in the liver of the intermediate host, causing a lethal zoonotic helminthic disease, alveolar echinococcosis (AE). However, the long non-coding RNAs (lncRNAs) expression profiles of the host and the potential regulatory function of lncRNA during Em infection are poorly understood. In this study, the profiles of lncRNAs and mRNAs in the liver of mice at different time points after Em infection were explored by microarray. Thirty-one differentially expressed mRNAs (DEMs) and 68 differentially expressed lncRNAs (DELs) were found continuously dysregulated. These DEMs were notably enriched in "antigen processing and presentation", "Th1 and Th2 cell differentiation" and "Th17 cell differentiation" pathways. The potential predicted function of DELs revealed that most DELs might influence Th17 cell differentiation and TGF-ß/Smad pathway of host by trans-regulating SMAD3, STAT1, and early growth response (EGR) genes. At 30 days post-infection (dpi), up-regulated DEMs were enriched in Toll-like and RIG-I-like receptor signaling pathways, which were validated by qRT-PCR, Western blotting and downstream cytokines detection. Furthermore, flow cytometric analysis and serum levels of the corresponding cytokines confirmed the changes in cell-mediated immunity in host during Em infection that showed Th1 and Th17-type CD4+ T-cells were predominant at the early infection stage whereas Th2-type CD4+ T-cells were significantly higher at the middle/late stage. Collectively, our study revealed the potential regulatory functions of lncRNAs in modulating host Th cell subsets and provide novel clues in understanding the influence of Em infection on host innate and adaptive immune response.


Sujet(s)
Echinococcus multilocularis , ARN long non codant , Animaux , Cytokines/métabolisme , Échinococcose , Foie/métabolisme , Souris , ARN long non codant/génétique , ARN messager/génétique
8.
Microbiol Spectr ; 10(2): e0209721, 2022 04 27.
Article de Anglais | MEDLINE | ID: mdl-35254168

RÉSUMÉ

Senecavirus A (SVA) infection induces inflammation in animals, such as fever, diarrhea, vesicles and erosions, and even death. The inflammatory cytokine interleukin-1ß (IL-1ß) plays a pivotal role in inflammatory responses to combat microbes. Although SVA infection can produce inflammatory clinical symptoms, the modulation of IL-1ß production by SVA infection remains unknown at present. Here, both in vitro and in vivo, SVA robustly induced IL-1ß production in macrophages and pigs. Infection performed in NOD-, LRR-, and pyrin domain-containing three (NLRP3) knockdown cells indicated that NLRP3 is essential for SVA-induced IL-1ß secretion. Importantly, we identified that the 1 to 154 amino acid (aa) portion of SVA 3D binds to the NLRP3 NACHT domain to activate NLRP3 inflammasome assembly and IL-1ß secretion. In addition, the SVA 3D protein interacts with IKKα and IKKß to induce NF-κB activation, which facilitates pro-IL-1ß transcription. Meanwhile, 3D induces p65 nucleus entry. Moreover, SVA 3D induces calcium influx and potassium efflux, which triggers IL-1ß secretion. Ion channels might be related to 3D binding with NLRP3, resulting in NLRP3-ASC complex assembly. We found that 3D protein expression induced tissue hemorrhage and swelling in the mice model. Consistently, expression of 3D in mice caused IL-1ß maturation and secretion. In the natural host of pigs, we confirmed that 3D also induced IL-1ß production. Our data reveal a novel mechanism underlying the activation of the NLRP3 inflammasome after SVA 3D expression, which provides clues for controlling pig's inflammation during the SVA infection. IMPORTANCE Inflammation refers to the response of the immune system to viral, bacterial, and fungal infections or other foreign particles in the body, which can involve the production of a wide array of soluble inflammatory mediators. The NLRP3 inflammasome is one of the best-characterized inflammasome leading to IL-1ß production and maturation. Senecavirus A (SVA) is an oncolytic virus that can cause fever, vesicles and erosions, severe fatal diarrhea, and even the sudden death of piglets. In this study, we demonstrated that 1 to 154 aa of SVA polymerase protein 3D interacts with the NACHT domain of NLRP3 to induce IL-1ß production via the NF-κB signaling pathway and ion channel signal. Our study unveils the mechanism underlying the regulation of inflammasome assembly and production of IL-1ß in response to SVA infection that will help better understand the modulation of host inflammation in pathogens invasion and development of the vaccine.


Sujet(s)
Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Animaux , Diarrhée , Inflammasomes/métabolisme , Inflammation , Canaux ioniques , Souris , Souris de lignée NOD , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Picornaviridae , Suidae
9.
Biomed Res Int ; 2021: 7377299, 2021.
Article de Anglais | MEDLINE | ID: mdl-34869770

RÉSUMÉ

OBJECTIVE: In this study, we explored the influence of single nucleotide polymorphism (SNP) in the noncoding region of intercellular adhesion molecule 1 (ICAM1) gene on the occurrence and metastasis of primary hepatocellular carcinoma (PHC). METHODS: Sanger sequencing was used to analyze the genotypes of rs3093032, rs923366, and rs281437 locus in the 3'untranslated region (UTR) of the ICAM1 gene. The level of plasma ICAM1 was analyzed by enzyme-linked immunosorbent assay (ELISA). RESULTS: After adjusting for risk factors such as BMI, smoking, drinking, family history of tumors, and hepatitis B virus test results, the CT genotype at rs3093032 of the ICAM1 gene (OR = 0.19, 95% CI: 0.08-0.44, P < 0.01), dominance model (OR = 0.23, 95% CI: 0.11-0.48, P < 0.01), and T allele (OR = 0.27, 95% CI: 0.14-0.53, P < 0.01) were related to the reduced risk of PHC susceptibility. rs923366 locus CT genotype (OR = 0.63, 95% CI: 0.44-0.90, P = 0.01), TT genotype (OR = 0.23, 95% CI: 0.10-0.53, P < 0.01), dominant model (OR = 0.55, 95% CI: 0.39-0.77, P < 0.01), recessive model (OR = 0.28, 95% CI: 0.12-0.62, P < 0.01), and T allele (OR = 0.55, 95% CI: 0.42-0.73, P < 0.01) were related to a reduction in the risk of PHC susceptibility. rs281437 locus CT genotype (OR = 2.08, 95% CI: 1.40-3.09, P < 0.01), TT genotype (OR = 5.20, 95% CI: 2.22-12.17, P < 0.01), dominant model (OR = 2.45, 95% CI: 1.69-3.54, P < 0.01), recessive model (OR = 4.32, 95% CI: 1.86-10.06, P < 0.01), and T allele (OR = 2.46, 95% CI: 1.79-3.38, P < 0.01) were significantly related to the increased risk of PHC susceptibility. SNPs at rs3093032, rs923366, and rs281437 of the ICAM1 gene were significantly correlated with TNM stage and tumor metastasis of PHC patients (P < 0.05). CONCLUSION: SNPs at rs3093032, rs923366, and rs281437 in the 3'UTR region of the ICAM1 gene are related to the occurrence and metastasis of PHC.


Sujet(s)
Régions 3' non traduites/génétique , Carcinome hépatocellulaire/génétique , Prédisposition génétique à une maladie/génétique , Molécule-1 d'adhérence intercellulaire/génétique , Tumeurs du foie/génétique , Métastase tumorale/génétique , Polymorphisme de nucléotide simple/génétique , Allèles , Carcinome hépatocellulaire/anatomopathologie , Femelle , Fréquence d'allèle/génétique , Génotype , Hépatite B/génétique , Virus de l'hépatite B/pathogénicité , Humains , Tumeurs du foie/anatomopathologie , Mâle , Adulte d'âge moyen , Métastase tumorale/anatomopathologie , Processus néoplasiques , Facteurs de risque
10.
Front Vet Sci ; 8: 697194, 2021.
Article de Anglais | MEDLINE | ID: mdl-34805327

RÉSUMÉ

Pathogens of viral origin produce a large variety of infectious diseases in livestock. It is essential to establish the best practices in animal care and an efficient way to stop and prevent infectious diseases that impact animal husbandry. So far, the greatest way to combat the disease is to adopt a vaccine policy. In the fight against infectious diseases, vaccines are very popular. Vaccination's fundamental concept is to utilize particular antigens, either endogenous or exogenous to induce immunity against the antigens or cells. In light of how past emerging and reemerging infectious diseases and pandemics were handled, examining the vaccination methods and technological platforms utilized for the animals may provide some useful insights. New vaccine manufacturing methods have evolved because of developments in technology and medicine and our broad knowledge of immunology, molecular biology, microbiology, and biochemistry, among other basic science disciplines. Genetic engineering, proteomics, and other advanced technologies have aided in implementing novel vaccine theories, resulting in the discovery of new ruminant vaccines and the improvement of existing ones. Subunit vaccines, recombinant vaccines, DNA vaccines, and vectored vaccines are increasingly gaining scientific and public attention as the next generation of vaccines and are being seen as viable replacements to conventional vaccines. The current review looks at the effects and implications of recent ruminant vaccine advances in terms of evolving microbiology, immunology, and molecular biology.

11.
J Inflamm Res ; 14: 1145-1163, 2021.
Article de Anglais | MEDLINE | ID: mdl-33814921

RÉSUMÉ

Inflammation refers to the response of the immune system to viral, bacterial, and fungal infections, or other foreign particles in the body, which can involve the production of a wide array of soluble inflammatory mediators. It is important for the development of many RNA virus-infected diseases. The primary factors through which the infection becomes inflammation involve inflammasome. Inflammasomes are proteins complex that the activation is responsive to specific pathogens, host cell damage, and other environmental stimuli. Inflammasomes bring about the maturation of various pro-inflammatory cytokines such as IL-18 and IL-1ß in order to mediate the innate immune defense mechanisms. Many RNA viruses and their components, such as encephalomyocarditis virus (EMCV) 2B viroporin, the viral RNA of hepatitis C virus, the influenza virus M2 viroporin, the respiratory syncytial virus (RSV) small hydrophobic (SH) viroporin, and the human rhinovirus (HRV) 2B viroporin can activate the Nod-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome to influence the inflammatory response. On the other hand, several viruses use virus-encoded proteins to suppress inflammation activation, such as the influenza virus NS1 protein and the measles virus (MV) V protein. In this review, we summarize how RNA virus infection leads to the activation or inhibition of the NLRP3 inflammasome.

12.
J Immunol ; 206(3): 566-579, 2021 02 01.
Article de Anglais | MEDLINE | ID: mdl-33380495

RÉSUMÉ

Peste des petits ruminants virus (PPRV) is a Morbillivirus that causes highly contagious and severe disease in various ruminants. PPRV infection leads to a severe inhibition of host antiviral immune response. Our previous study demonstrated that PPRV V protein blocks IFN response by targeting STAT proteins. In the current study, we identified the phosphoprotein (P) as a novel antagonistic factor of PPRV to counteract host antiviral innate immune response. PPRV P protein significantly suppressed RIG-I-like receptor pathway signaling and impaired IFN-ß and ISGs expression by targeting IFN regulatory factor (IRF)3 in both human embryonic kidney 293T cells and primary goat fibroblasts. The 1-102 region of P protein was critical for the antagonistic function of P protein. P protein interacted with IRF association domain (IAD) of IRF3 to block the interaction between TBK1 and IRF3. The interaction between TBK1 and the IAD of IRF3 is responsible for triggering the phosphorylation of IRF3. P protein competed with TBK1 to bind to the IAD of IRF3 that contributed to the decreased phosphorylation of IRF3, which, in turn, interfered with the dimerization of IRF3 and blocked IRF3 nuclear transportation. Besides, we also found that P protein interacted with IRF5 and IRF8. However, the involved mechanism remains unknown. Taken together, our results reveal a novel mechanism by which PPRV P protein antagonizes host antiviral innate immune response by interacting with the transcription factor IRF3, thereby inhibiting the type I IFN production and promoting viral replication.


Sujet(s)
Protéine-58 à domaine DEAD/métabolisme , Fibroblastes/physiologie , Facteur-3 de régulation d'interféron/métabolisme , Peste des petits ruminants/immunologie , Virus de la peste des petits ruminants/physiologie , Phosphoprotéines/métabolisme , Protéines virales/métabolisme , Animaux , Cellules cultivées , Capra , Humains , Échappement immunitaire , Immunité innée , Facteur-3 de régulation d'interféron/génétique , Transduction du signal , Réplication virale
13.
Viruses ; 14(1)2021 12 23.
Article de Anglais | MEDLINE | ID: mdl-35062226

RÉSUMÉ

Foot-and-mouth disease virus (FMDV) infection causes inflammatory clinical symptoms, such as high fever and vesicular lesions, even death of animals. Interleukin-1ß (IL-1ß) is an inflammatory cytokine that plays an essential role in inflammatory responses against viral infection. The viruses have developed multiple strategies to induce the inflammatory responses, including regulation of IL-1ß production. However, the molecular mechanism underlying the induction of IL-1ß by FMDV remains not fully understood. Here, we found that FMDV robustly induced IL-1ß production in macrophages and pigs. Infection of Casp-1 inhibitor-treated cells and NOD-, LRR- and pyrin domain-containing 3 (NLRP3)-knockdown cells indicated that NLRP3 is essential for FMDV-induced IL-1ß secretion. More importantly, we found that FMDV Lpro associates with the NACHT and LRR domains of NLRP3 to promote NLRP3 inflammasome assembly and IL-1ß secretion. Moreover, FMDV Lpro induces calcium influx and potassium efflux, which trigger NLRP3 activation. Our data revealed the mechanism underlying the activation of the NLRP3 inflammasome after FMDV Lpro expression, thus providing insights for the control of FMDV infection-induced inflammation.


Sujet(s)
Endopeptidases/métabolisme , Virus de la fièvre aphteuse/métabolisme , Fièvre aphteuse/immunologie , Interleukine-1 bêta/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Animaux , Lignée cellulaire , Fièvre aphteuse/virologie , Humains , Inflammasomes/métabolisme , Canaux ioniques/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/composition chimique , Domaines protéiques , Suidae
14.
J Virol ; 93(16)2019 08 15.
Article de Anglais | MEDLINE | ID: mdl-31167907

RÉSUMÉ

Peste des petits ruminants virus (PPRV) is the etiological agent of peste des petits ruminants, causing acute immunosuppression in its natural hosts. However, the molecular mechanisms by which PPRV antagonizes the host immune responses have not been fully characterized. In particular, how PPRV suppresses the activation of the host RIG-I-like receptor (RLR) pathway has yet to be clarified. In this study, we demonstrated that PPRV infection significantly suppresses RLR pathway activation and type I interferon (IFN) production and identified PPRV N protein as an extremely important antagonistic viral factor that suppresses beta interferon (IFN-ß) and IFN-stimulated gene (ISG) expression. A detailed analysis showed that PPRV N protein inhibited type I IFN production by targeting interferon regulatory factor 3 (IRF3), a key molecule in the RLR pathway required for type I IFN induction. PPRV N protein interacted with IRF3 (but not with other components of the RLR pathway, including MDA5, RIG-I, VISA, TBK1, and MITA) and abrogated the phosphorylation of IRF3. As expected, PPRV N protein also considerably impaired the nuclear translocation of IRF3. The TBK1-IRF3 interaction was involved significantly in IRF3 phosphorylation, and we showed that PPRV N protein inhibits the association between TBK1 and IRF3, which in turn inhibits IRF3 phosphorylation. The amino acid region 106 to 210 of PPRV N protein was determined to be essential for suppressing the nuclear translocation of IRF3 and IFN-ß production, and the 140 to 400 region of IRF3 was identified as the crucial region for the N-IRF3 interaction. Together, our findings demonstrate a new mechanism evolved by PPRV to inhibit type I IFN production and provide structural insights into the immunosuppression caused by PPRV.IMPORTANCE Peste des petits ruminants is a highly contagious animal disease affecting small ruminants, which threatens both small livestock and endangered susceptible wildlife populations in many countries. The causative agent, peste des petits ruminants virus (PPRV), often causes acute immunosuppression in its natural hosts during infection. Here, for the first time, we demonstrate that N protein, the most abundant protein of PPRV, plays an extremely important role in suppression of interferon regulatory factor 3 (IRF3) function and type I interferon (IFN) production by interfering with the formation of the TBK1-IRF3 complex. This study explored a novel antagonistic mechanism of PPRV.


Sujet(s)
Interactions hôte-pathogène , Facteur-3 de régulation d'interféron/métabolisme , Interféron bêta/biosynthèse , Protéines nucléocapside/métabolisme , Peste des petits ruminants/métabolisme , Peste des petits ruminants/virologie , Virus de la peste des petits ruminants/physiologie , Animaux , Immunomodulation , Interféron bêta/génétique , Phosphorylation , Régions promotrices (génétique) , Liaison aux protéines , Transport des protéines , Activation de la transcription
15.
Acta Trop ; 161: 68-72, 2016 Sep.
Article de Anglais | MEDLINE | ID: mdl-27260667

RÉSUMÉ

The disease microsporidiosis is found worldwide and is mainly caused by Enterocytozoon bieneusi. E. bieneusi can infect a wide range of hosts; however, information regarding the prevalence and genotyping of E. bieneusi infection in raccoon dogs (Nyctereutes procyonoides) is limited. Therefore, in 2015, we examined 305 faecel samples from 80 farmed raccoon dogs in Jilin Province, from 54 in Hebei Province, from 72 in Liaoning Province, from 29 in Shandong Province, and from 40 in Heilongjiang Province. The overall prevalence of E. bieneusi infection in farmed raccoon dogs was 22.30%. Logistic regression analysis suggests that age, gender and region of raccoon dogs were highly related to the prevalence of E. bieneusi infection. Moreover, six E. bieneusi internal transcribed spacer (ITS) region sequences, including four known genotypes, namely D, CHN-DC1, NCF2, and CHN-F1, and two novel genotypes (NCR1 and NCR2), were identified in the present study. The present study firstly indicated the existence of E. bieneusi genotypes NCF2, NCR1, NCR2and CHN-F1 in infected raccoon dogs in Northern China. Integrated control strategies should be implemented to limit E. bieneusi infection in farmed raccoon dogs, and to prevent transmission of this disease to other animals and humans.


Sujet(s)
Entérocytozoon/génétique , Fèces/microbiologie , Microsporidiose/épidémiologie , Chiens viverrins/microbiologie , Animaux , Chine/épidémiologie , Chiens , Génotype , Prévalence , Facteurs de risque
16.
Virology ; 483: 54-63, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25965795

RÉSUMÉ

Peste des petits ruminants virus (PPRV) causes a fatal disease in small ruminants. V protein of PPRV plays a pivotal role in interfering with host innate immunity by blocking IFNs signaling through interacting with STAT1 and STAT2. In the present study, the results demonstrated that PPRV V protein blocks IFN actions in a dose dependent manner and restrains the translocation of STAT1/2 proteins. We speculate that the translocation inhibition might be caused by the interfering of the downstream of STAT protein. Mutagenesis defines that Cys cluster and Trp motif of PPRV V protein are essential for STAT-mediated IFN signaling. These findings give a new sight for the further studies to understand the delicate mechanism of PPRV to escape the IFN signaling.


Sujet(s)
Acides aminés/métabolisme , Interférons/antagonistes et inhibiteurs , Virus de la peste des petits ruminants/physiologie , Cartes d'interactions protéiques , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-2/métabolisme , Protéines virales/métabolisme , Acides aminés/génétique , Animaux , Lignée cellulaire , Analyse de mutations d'ADN , Humains , Échappement immunitaire , Mutation faux-sens , Liaison aux protéines , Transduction du signal , Protéines virales/génétique
17.
Korean J Parasitol ; 53(6): 689-97, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26797435

RÉSUMÉ

The tapeworm Taenia solium is an important human zoonotic parasite that causes great economic loss and also endangers public health. At present, an effective vaccine that will prevent infection and chemotherapy without any side effect remains to be developed. In this study, codon usage patterns in the T. solium genome were examined through 8,484 protein-coding genes. Neutrality analysis showed that T. solium had a narrow GC distribution, and a significant correlation was observed between GC12 and GC3. Examination of an NC (ENC vs GC3s)-plot showed a few genes on or close to the expected curve, but the majority of points with low-ENC (the effective number of codons) values were detected below the expected curve, suggesting that mutational bias plays a major role in shaping codon usage. The Parity Rule 2 plot (PR2) analysis showed that GC and AT were not used proportionally. We also identified 26 optimal codons in the T. solium genome, all of which ended with either a G or C residue. These optimal codons in the T. solium genome are likely consistent with tRNAs that are highly expressed in the cell, suggesting that mutational and translational selection forces are probably driving factors of codon usage bias in the T. solium genome.


Sujet(s)
Codon/génétique , Génome d'helminthe , Protéines d'helminthes/génétique , Taenia solium/génétique , Animaux , Séquence nucléotidique , Évolution moléculaire , Données de séquences moléculaires
18.
PLoS One ; 9(8): e104446, 2014.
Article de Anglais | MEDLINE | ID: mdl-25119375

RÉSUMÉ

The adjuvant effects of Lactobacillus acidophilus on DNA vaccination are not fully understood. It has been hypothesized that swine-derived Lactobacillus acidophilus SW1 (LASW1) could function as an immune adjuvant to enhance antigen-specific immune responses after foot-and-mouth disease (FMD) DNA vaccination in mice. To evaluate the effect of oral LASW1 on the immune response to a DNA vaccine (pRC/CMV-vp1) harboring FMD VP1 gene, anti-FMDV antibody and its isotypes, T-cell proliferation, and cytokine detection were investigated. The results showed that LASW1 was able to enhance FMDV-specific antibody levels and FMDV-neutralizing antibodies. After a booster vaccine, the anti-FMDV antibody titers and FMDV-neutralizing antibodies levels induced by pRC/CMV-vp1 were higher in mice treated with LSAW1 than in the group immunized with pRC/CMV-vp1 alone (the control). Using T-cell proliferation, the stimulation index of the LASW1 group was significantly higher in response to ConA and 146S antigen (P<0.05) than in the control group. Importantly, higher concentrations of IFN-γ and IFN-γ-producing cells were also observed in splenocytes isolated from the experimental LASW1 mice, indicating that INF-γ secretion is important to the immune response to LASW1. The results indicate that LASW1 is a promising immune adjuvant in DNA vaccination against FMD when administrated orally.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Antigènes bactériens/pharmacologie , Virus de la fièvre aphteuse/immunologie , Lactobacillus acidophilus/immunologie , Vaccins à ADN/immunologie , Vaccins antiviraux/immunologie , Adjuvants immunologiques/administration et posologie , Administration par voie orale , Analyse de variance , Animaux , Antigènes bactériens/administration et posologie , Antigènes bactériens/immunologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytokines/immunologie , Test ELISpot , Souris , Lymphocytes T/immunologie
19.
Virol J ; 8: 476, 2011 Oct 21.
Article de Anglais | MEDLINE | ID: mdl-22014033

RÉSUMÉ

BACKGROUND: Porcine reproductive and respitatory syndrome virus (PRRSV) is a recently emerged pathogen and severely affects swine populations worldwide. The replication of PRRSV is tightly controlled by viral gene expression and the codon usage of translation initiation region within each gene could potentially regulate the translation rate. Therefore, a better understanding of the codon usage pattern of the initiation translation region would shed light on the regulation of PRRSV gene expression. RESULTS: In this study, the codon usage in the translation initiation region and in the whole coding sequence was compared in PRRSV ORF1a and ORFs2-7. To investigate the potential role of codon usage in affecting the translation initiation rate, we established a codon usage model for PRRSV translation initiation region. We observed that some non-preferential codons are preferentially used in the translation initiation region in particular ORFs. Although some positions vary with codons, they intend to use codons with negative CUB. Furthermore, our model of codon usage showed that the conserved pattern of CUB is not directly consensus with the conserved sequence, but shaped under the translation selection. CONCLUSIONS: The non-variation pattern with negative CUB in the PRRSV translation initiation region scanned by ribosomes is considered the rate-limiting step in the translation process.


Sujet(s)
Codon , Régulation de l'expression des gènes viraux , Génome viral , Initiation de la traduction , Virus du syndrome respiratoire et reproducteur porcin/génétique , Cadres ouverts de lecture , Ribosomes/métabolisme , Protéines virales/biosynthèse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...