Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 230
Filtrer
1.
BMC Vet Res ; 20(1): 301, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38971791

RÉSUMÉ

BACKGROUND: Foot-and-mouth disease (FMD) is a devastating disease affecting cloven-hoofed animals, that leads to significant economic losses in affected countries and regions. Currently, there is an evident inclination towards the utilization of nanoparticles as powerful platforms for innovative vaccine development. Therefore, this study developed a ferritin-based nanoparticle (FNP) vaccine that displays a neutralizing epitope of foot-and-mouth disease virus (FMDV) VP1 (aa 140-158) on the surface of FNP, and evaluated the immunogenicity and protective efficacy of these FNPs in mouse and guinea pig models to provide a strategy for developing potential FMD vaccines. RESULTS: This study expressed the recombinant proteins Hpf, HPF-NE and HPF-T34E via an E. coli expression system. The results showed that the recombinant proteins Hpf, Hpf-NE and Hpf-T34E could be effectively assembled into nanoparticles. Subsequently, we evaluated the immunogenicity of the Hpf, Hpf-NE and Hpf-T34E proteins in mice, as well as the immunogenicity and protectiveness of the Hpf-T34E protein in guinea pigs. The results of the mouse experiment showed that the immune efficacy in the Hpf-T34E group was greater than the Hpf-NE group. The results from guinea pigs immunized with Hpf-T34E showed that the immune efficacy was largely consistent with the immunogenicity of the FMD inactivated vaccine (IV) and could confer partial protection against FMDV challenge in guinea pigs. CONCLUSIONS: The Hpf-T34E nanoparticles stand out as a superior choice for a subunit vaccine candidate against FMD, offering effective protection in FMDV-infected model animals. FNP-based vaccines exhibit excellent safety and immunogenicity, thus representing a promising strategy for the continued development of highly efficient and safe FMD vaccines.


Sujet(s)
Épitopes , Ferritines , Virus de la fièvre aphteuse , Fièvre aphteuse , Nanoparticules , Vaccins antiviraux , Animaux , Cochons d'Inde , Fièvre aphteuse/prévention et contrôle , Fièvre aphteuse/immunologie , Virus de la fièvre aphteuse/immunologie , Ferritines/immunologie , Vaccins antiviraux/immunologie , Épitopes/immunologie , Souris , Femelle , Souris de lignée BALB C , Protéines recombinantes/immunologie , Protéines de capside
2.
Front Microbiol ; 15: 1419615, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952452

RÉSUMÉ

African swine fever (ASF) is an infectious disease characterized by hemorrhagic fever, which is highly pathogenic and causes severe mortality in domestic pigs. It is caused by the African swine fever virus (ASFV). ASFV is a large DNA virus and primarily infects porcine monocyte macrophages. The interaction between ASFV and host macrophages is the major reason for gross pathological lesions caused by ASFV. Necroptosis is an inflammatory programmed cell death and plays an important immune role during virus infection. However, whether and how ASFV induces macrophage necroptosis and the effect of necroptosis signaling on host immunity and ASFV infection remains unknown. This study uncovered that ASFV infection activates the necroptosis signaling in vivo and macrophage necroptosis in vitro. Further evidence showed that ASFV infection upregulates the expression of ZBP1 and RIPK3 to consist of the ZBP1-RIPK3-MLKL necrosome and further activates macrophage necroptosis. Subsequently, multiple Z-DNA sequences were predicted to be present in the ASFV genome. The Z-DNA signals were further confirmed to be present and colocalized with ZBP1 in the cytoplasm and nucleus of ASFV-infected cells. Moreover, ZBP1-mediated macrophage necroptosis provoked the extracellular release of proinflammatory cytokines, including TNF-α and IL-1ß induced by ASFV infection. Finally, we demonstrated that ZBP1-mediated necroptosis signaling inhibits ASFV replication in host macrophages. Our findings uncovered a novel mechanism by which ASFV induces macrophage necroptosis by facilitating Z-DNA accumulation and ZBP1 necrosome assembly, providing significant insights into the pathogenesis of ASFV infection.

3.
mBio ; 15(8): e0154924, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-38953350

RÉSUMÉ

Metabolism in host cells can be modulated after viral infection, favoring viral survival or clearance. Here, we report that lipid droplet (LD) synthesis in host cells can be modulated by yin yang 1 (YY1) after porcine reproductive and respiratory syndrome virus (PRRSV) infection, resulting in active antiviral activity. As a ubiquitously distributed transcription factor, there was increased expression of YY1 upon PRRSV infection both in vitro and in vivo. YY1 silencing promoted the replication of PRRSV, whereas YY1 overexpression inhibited PRRSV replication. PRRSV infection led to a marked increase in LDs, while YY1 knockout inhibited LD synthesis, and YY1 overexpression enhanced LD accumulation, indicating that YY1 reprograms PRRSV infection-induced intracellular LD synthesis. We also showed that the viral components do not colocalize with LDs during PRRSV infection, and the effect of exogenously induced LD synthesis on PRRSV replication is nearly lethal. Moreover, we demonstrated that YY1 affects the synthesis of LDs by regulating the expression of lipid metabolism genes. YY1 negatively regulates the expression of fatty acid synthase (FASN) to weaken the fatty acid synthesis pathway and positively regulates the expression of peroxisome proliferator-activated receptor gamma (PPARγ) to promote the synthesis of LDs, thus inhibiting PRRSV replication. These novel findings indicate that YY1 plays a crucial role in regulating PRRSV replication by reprogramming LD synthesis. Therefore, our study provides a novel mechanism of host resistance to PRRSV and suggests potential new antiviral strategies against PRRSV infection.IMPORTANCEPorcine reproductive and respiratory virus (PRRSV) has caused incalculable economic damage to the global pig industry since it was first discovered in the 1980s. However, conventional vaccines do not provide satisfactory protection. It is well known that viruses are parasitic pathogens, and the completion of their replication life cycle is highly dependent on host cells. A better understanding of host resistance to PRRSV infection is essential for developing safe and effective strategies to control PRRSV. Here, we report a crucial host antiviral molecule, yin yang 1 (YY1), which is induced to be expressed upon PRRSV infection and subsequently inhibits virus replication by reprogramming lipid droplet (LD) synthesis through transcriptional regulation. Our work provides a novel antiviral mechanism against PRRSV infection and suggests that targeting YY1 could be a new strategy for controlling PRRSV.


Sujet(s)
Gouttelettes lipidiques , Virus du syndrome respiratoire et reproducteur porcin , Réplication virale , Facteur de transcription YY1 , Facteur de transcription YY1/métabolisme , Facteur de transcription YY1/génétique , Animaux , Virus du syndrome respiratoire et reproducteur porcin/physiologie , Virus du syndrome respiratoire et reproducteur porcin/génétique , Suidae , Gouttelettes lipidiques/métabolisme , Syndrome dysgénésique et respiratoire porcin/virologie , Syndrome dysgénésique et respiratoire porcin/métabolisme , Syndrome dysgénésique et respiratoire porcin/génétique , Lignée cellulaire , Métabolisme lipidique , Interactions hôte-pathogène
4.
J Med Chem ; 67(15): 12932-12944, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-38996365

RÉSUMÉ

Toll-like receptor 2 (TLR2) plays a crucial role in detecting microbial pathogen-associated molecular patterns, offering potential applications as an adjuvant for vaccines and antitumor therapies. Here, we present the gram-scale synthesis of CaLGL-1 and its derivatives, natural products known for activating mouse TLR2 (EC50 = 3.2 µM). This synthesis involves a streamlined six-step reaction sequence utilizing oxidant-promoted acetalization, effectively preserving the acid-sensitive glycosidic bond for maintaining the compounds' functional integrity. Our structure-activity relationship studies identified R-7d as a potent human TLR2 activator. It demonstrated subnanomolar activity (EC50 = 116 pM) in human THP-1 cells, comparable to that of diprovocim (EC50 = 110 pM). Experiments revealed that R-7d enhances NF-kB promoter activation through TLR2/TLR1 heterodimers rather than TLR2/TLR6. The discovery of R-7d as a robust human TLR2 agonist opens up new possibilities for combination therapies.


Sujet(s)
Récepteur de type Toll-2 , Humains , Récepteur de type Toll-2/agonistes , Récepteur de type Toll-2/métabolisme , Relation structure-activité , Cellules THP-1 , Facteur de transcription NF-kappa B/métabolisme , Récepteur de type Toll-1/agonistes , Récepteur de type Toll-1/métabolisme
5.
Anal Chem ; 96(29): 12120-12128, 2024 07 23.
Article de Anglais | MEDLINE | ID: mdl-38990044

RÉSUMÉ

Dihydro-nicotinamide adenine dinucleotide (NADH) detection is crucial since it is a vital coenzyme in organism metabolism. Compared to the traditional method based on natural NADH oxidase (NOX), nanozymes with multienzyme-like activity can catalyze multistage reactions in a singular setup, simplifying detection processes and enhancing sensitivity. In this study, an innovative NADH detection method was developed using iron-doped carbon (Fe@C) nanozyme synthesized from metal-organic frameworks with in situ reduced Pt clusters. This nanozyme composite (Pt/Fe@C) demonstrated dual NOX and peroxidase-like characteristics, significantly enhancing the catalytic efficiency and enabling NADH conversion to NAD+ and H2O2 with subsequent detection. The collaborative research involving both experimental and theoretical simulations has uncovered the catalytic process and the cooperative effect of Fe and Pt atoms, leading to enhanced oxygen adsorption and activation, as well as a decrease in the energy barrier of the key step in the H2O2 decomposition process. These findings indicate that the catalytic performance of Pt/Fe@C in NOX-like and POD-like reactions can be significantly improved. The colorimetric sensor detects NADH with a limit of detection as low as 0.4 nM, signifying a breakthrough in enzyme-mimicking nanozyme technology for precise NADH measurement.


Sujet(s)
Carbone , Réseaux organométalliques , NAD , Platine , NAD/composition chimique , Réseaux organométalliques/composition chimique , Platine/composition chimique , Carbone/composition chimique , Fer/composition chimique , Peroxyde d'hydrogène/composition chimique , Colorimétrie/méthodes , Humains , Catalyse , Complexes multienzymatiques/composition chimique , Complexes multienzymatiques/métabolisme , Matériaux biomimétiques/composition chimique , Limite de détection , NADH, NADPH oxidoreductases
6.
J Virol ; 98(7): e0058524, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38869319

RÉSUMÉ

Senecavirus A (SVA), a picornavirus, causes vesicular diseases and epidemic transient neonatal losses in swine, resulting in a multifaceted economic impact on the swine industry. SVA counteracts host antiviral response through multiple strategies facilitatng viral infection and transmission. However, the mechanism of how SVA modulates interferon (IFN) response remains elusive. Here, we demonstrate that SVA 3C protease (3Cpro) blocks the transduction of Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway to antagonize type I IFN response. Mechanistically, 3Cpro selectively cleaves and degrades STAT1 and STAT2 while does not target JAK1, JAK2, and IRF9, through its protease activity. Notably, SVA 3Cpro cleaves human and porcine STAT1 on a Leucine (L)-Aspartic acid (D) motif, specifically L693/D694. In the case of STAT2, two cleavage sites were identified: glutamine (Q) 707 was identified in both human and porcine, while the second cleavage pattern differed, with residues 754-757 (Valine-Leucine-Glutamine-Serine motifs) in human STAT2 and Q758 in porcine STAT2. These cleavage patterns by SVA 3Cpro partially differ from previously reported classical motifs recognized by other picornaviral 3Cpro, highlighting the distinct characteristics of SVA 3Cpro. Together, these results reveal a mechanism by which SVA 3Cpro antagonizes IFN-induced antiviral response but also expands our knowledge about the substrate recognition patterns for picornaviral 3Cpro.IMPORTANCESenecavirus A (SVA), the only member in the Senecavirus genus within the Picornaviridae family, causes vesicular diseases in pigs that are clinically indistinguishable from foot-and-mouth disease (FMD), a highly contagious viral disease listed by the World Organization for Animal Health (WOAH). Interferon (IFN)-mediated antiviral response plays a pivotal role in restricting and controlling viral infection. Picornaviruses evolved numerous strategies to antagonize host antiviral response. However, how SVA modulates the JAK-STAT signaling pathway, influencing the type I IFN response, remains elusive. Here, we identify that 3Cpro, a protease of SVA, functions as an antagonist for the IFN response. 3Cpro utilizes its protease activity to cleave STAT1 and STAT2, thereby diminishing the host IFN response to promote SVA infection. Our findings underscore the significance of 3Cpro as a key virulence factor in the antagonism of the type I signaling pathway during SVA infection.


Sujet(s)
Cysteine endopeptidases , Infections à Picornaviridae , Picornaviridae , Facteur de transcription STAT-1 , Facteur de transcription STAT-2 , Transduction du signal , Protéines virales , Animaux , Suidae , Facteur de transcription STAT-2/métabolisme , Humains , Infections à Picornaviridae/virologie , Infections à Picornaviridae/immunologie , Infections à Picornaviridae/métabolisme , Facteur de transcription STAT-1/métabolisme , Cysteine endopeptidases/métabolisme , Protéines virales/métabolisme , Protéases virales 3C , Maladies des porcs/virologie , Maladies des porcs/immunologie , Cellules HEK293 , Interféron de type I/antagonistes et inhibiteurs , Interféron de type I/métabolisme , Lignée cellulaire , Janus kinases/métabolisme , Janus kinases/antagonistes et inhibiteurs
7.
Autophagy ; : 1-20, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38752369

RÉSUMÉ

Macroautophagy/autophagy and apoptosis are pivotal interconnected host cell responses to viral infection, including picornaviruses. Here, the VP3 proteins of picornaviruses were determined to trigger autophagy, with the autophagic flux being triggered by the TP53-BAD-BAX axis. Using foot-and-mouth disease virus (FMDV) as a model system, we unraveled a novel mechanism of how picornavirus hijacks autophagy to bolster viral replication and enhance pathogenesis. FMDV infection induced both autophagy and apoptosis in vivo and in vitro. FMDV VP3 protein facilitated the phosphorylation and translocation of TP53 from the nucleus into the mitochondria, resulting in BAD-mediated apoptosis and BECN1-mediated autophagy. The amino acid Gly129 in VP3 is essential for its interaction with TP53, and crucial for induction of autophagy and apoptosis. VP3-induced autophagy and apoptosis are both essential for FMDV replication, while, autophagy plays a more important role in VP3-mediated pathogenesis. Mutation of Gly129 to Ala129 in VP3 abrogated the autophagic regulatory function of VP3, which significantly decreased the viral replication and pathogenesis of FMDV. This suggested that VP3-induced autophagy benefits viral replication and pathogenesis. Importantly, this Gly is conserved and showed a common function in various picornaviruses. This study provides insight for developing broad-spectrum antivirals and genetic engineering attenuated vaccines against picornaviruses.Abbreviations: 3-MA, 3-methyladenine; ATG, autophagy related; BAD, BCL2 associated agonist of cell death; BAK1, BCL2 antagonist/killer 1; BAX, BCL2 associated X, apoptosis regulator; BBC3/PUMA, BCL2 binding component 3; BCL2, BCL2 apoptosis regulator; BID, BH3 interacting domain death agonist; BIP-V5, BAX inhibitor peptide V5; CFLAR/FLIP, CASP8 and FADD like apoptosis regulator; CPE, cytopathic effects; CQ, chloroquine; CV, coxsackievirus; DAPK, death associated protein kinase; DRAM, DNA damage regulated autophagy modulator; EV71, enterovirus 71; FMDV, foot-and-mouth disease virus; HAV, hepatitis A virus; KD, knockdown; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; MOI, multiplicity of infection; MTOR, mechanistic target of rapamycin kinase; PML, promyelocytic leukemia; PV, poliovirus; SVA, Seneca Valley virus; TCID50, 50% tissue culture infectious doses; TOR, target of rapamycin. TP53/p53, tumor protein p53; WCL, whole-cell lysate.

9.
Appl Microbiol Biotechnol ; 108(1): 350, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38809284

RÉSUMÉ

The African swine fever virus (ASFV) has the ability to infect pigs and cause a highly contagious acute fever that can result in a mortality rate as high as 100%. Due to the viral epidemic, the pig industry worldwide has suffered significant financial setbacks. The absence of a proven vaccine for ASFV necessitates the development of a sensitive and reliable serological diagnostic method, enabling laboratories to effectively and expeditiously detect ASFV infection. In this study, four strains of monoclonal antibodies (mAbs) against p72, namely, 5A1, 4C4, 8A9, and 5E10, were generated through recombinant expression of p72, the main capsid protein of ASFV, and immunized mice with it. Epitope localization was performed by truncated overlapping polypeptides. The results indicate that 5A1 and 4C4 recognized the amino acid 20-39 aa, 8A9 and 5E10 are recognized at 263-282 aa, which is consistent with the reported 265-280 aa epitopes. Conserved analysis revealed 20-39 aa is a high conservation of the epitopes in the ASFV genotypes. Moreover, a blocking ELISA assay for detection ASFV antibody based on 4C4 monoclonal antibody was developed and assessed. The receiver-operating characteristic (ROC) was performed to identify the best threshold value using 87 negative and 67 positive samples. The established test exhibited an area under the curve (AUC) of 0.9997, with a 95% confidence interval ranging from 99.87 to 100%. Furthermore, the test achieved a diagnostic sensitivity of 100% (with a 95% confidence interval of 95.72 to 100%) and a specificity of 98.51% (with a 95% confidence interval of 92.02 to 99.92%) when the threshold was set at 41.97%. The inter- and intra-batch coefficient of variation were below 10%, demonstrating the exceptional repeatability of the method. This method can detect the positive standard serum at a dilution as high as 1:512. Subsequently, an exceptional blocking ELISA assay was established with high diagnostic sensitivity and specificity, providing a novel tool for detecting ASFV antibodies. KEY POINTS: • Four strains of ASFV monoclonal antibodies against p72 were prepared and their epitopes were identified. • Blocking ELISA method was established based on monoclonal antibody 4C4 with an identified conservative epitope. • The established blocking ELISA method has a good effect on the detection of ASFV antibody.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Anticorps monoclonaux , Anticorps antiviraux , Protéines de capside , Test ELISA , Cartographie épitopique , Animaux , Anticorps monoclonaux/immunologie , Virus de la peste porcine africaine/immunologie , Virus de la peste porcine africaine/génétique , Test ELISA/méthodes , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Suidae , Peste porcine africaine/diagnostic , Peste porcine africaine/immunologie , Peste porcine africaine/virologie , Souris , Protéines de capside/immunologie , Protéines de capside/génétique , Souris de lignée BALB C , Sensibilité et spécificité , Épitopes/immunologie
10.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1536-1547, 2024 May 25.
Article de Chinois | MEDLINE | ID: mdl-38783814

RÉSUMÉ

The aim of this study was to prepare a mouse monoclonal antibody against the nonstructural protein 1 (NS1) of respiratory syncytial virus (RSV) to analyze its expression and distribution during transfection and infection. Additionally, we aimed to evaluate the antibody's application in immunoprecipitation assay. Firstly, the NS1 gene fragment was cloned into a prokaryotic plasmid and expressed in Escherichia coli. The resulting NS1 protein was then purified by affinity chromatography, and used to immunize the BALB/c mice. Subsequently, hybridoma cells capable of stably secreting the NS1 monoclonal antibody were selected using indirect enzyme linked immunosorbent assay (ELISA). This monoclonal antibody was employed in both indirect immunofluorescence assay (IFA) and Western blotting to analyze the expression and distribution of RSV NS1 in overexpressed and infected cells. Finally, the reliability of this monoclonal antibody was evaluated through the immunoprecipitation assay. The results showed that the RSV NS1 protein was successfully expressed and purified. Following immunization of mice with this protein, we obtained a highly specific RSV NS1 monoclonal antibody, which belonged to the IgG1 subtype with an antibody titer of 1:15 360 000. Using this monoclonal antibody, the RSV NS1 protein was identified in both transfected and infected cells. The IFA results revealed predominant distribution of NS1 in the cytoplasm and nucleus. Moreover, we confirmed that this monoclonal antibody could effectively bind specifically to NS1 protein in cell lysates, making it suitable as a capture antibody in immunoprecipitation assay. In conclusion, our study successfully achieved production of the RSV NS1 protein through a prokaryotic expression system and prepared a specific monoclonal antibody against NS1. This antibody demonstrates the ability to specifically identify the NS1 protein and can be used in the immunoprecipitation assay, thereby laying a foundation for the functional studies of the NS1 protein.


Sujet(s)
Anticorps monoclonaux , Protéines virales non structurales , Animaux , Femelle , Souris , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/génétique , Anticorps antiviraux/immunologie , Escherichia coli/génétique , Escherichia coli/métabolisme , Hybridomes/immunologie , Souris de lignée BALB C , Virus respiratoires syncytiaux/immunologie , Virus respiratoires syncytiaux/génétique , Protéines virales non structurales/immunologie , Protéines virales non structurales/génétique
11.
Appl Microbiol Biotechnol ; 108(1): 283, 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38573435

RÉSUMÉ

Porcine reproductive and respiratory syndrome (PRRS) is an immunosuppressive disease caused by the porcine reproductive and respiratory syndrome virus (PRRSV). Current vaccine prevention and treatment approaches for PRRS are not adequate, and commercial vaccines do not provide sufficient cross-immune protection. Therefore, establishing a precise, sensitive, simple, and rapid serological diagnostic approach for detecting PRRSV antibodies is crucial. The present study used quantum dot fluorescent microspheres (QDFM) as tracers, covalently linked to the PRRSV N protein, to develop an immunochromatography strip (ICS) for detecting PRRSV antibodies. Monoclonal antibodies against PRRSV nucleocapsid (N) and membrane (M) proteins were both coated on nitrocellulose membranes as control (C) and test (T) lines, respectively. QDFM ICS identified PRRSV antibodies under 10 min with high sensitivity and specificity. The specificity assay revealed no cross-reactivity with the other tested viruses. The sensitivity assay revealed that the minimum detection limit was 1.2 ng/mL when the maximum dilution was 1:2,048, comparable to the sensitivity of enzyme-linked immunosorbent assay (ELISA) kits. Moreover, compared to PRRSV ELISA antibody detection kits, the sensitivity, specificity, and accuracy of QDFM ICS after analyzing 189 clinical samples were 96.7%, 97.9%, and 97.4%, respectively. Notably, the test strips can be stored for up to 6 months at 4 °C and up to 4 months at room temperature (18-25 °C). In conclusion, QDFM ICS offers the advantages of rapid detection time, high specificity and sensitivity, and affordability, indicating its potential for on-site PRRS screening. KEY POINTS: • QDFM ICS is a novel method for on-site and in-lab detection of PRRSV antibodies • Its sensitivity, specificity, and accuracy are on par with commercial ELISA kits • QDFM ICS rapidly identifies PRRSV, aiding the swine industry address the evolving virus.


Sujet(s)
Syndrome dysgénésique et respiratoire porcin , Virus du syndrome respiratoire et reproducteur porcin , Boîtes quantiques , Animaux , Suidae , Microsphères , Syndrome dysgénésique et respiratoire porcin/diagnostic , Agents colorants , Anticorps antiviraux , Chromatographie d'affinité
12.
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
13.
Vet Microbiol ; 293: 110074, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38603982

RÉSUMÉ

African swine fever (ASF) is a highly impactful infectious disease in the swine industry, leading to substantial economic losses globally. The causative agent, African swine fever virus (ASFV), possesses intricate pathogenesis, warranting further exploration. In this study, we investigated the impact of ASFV infection on host gene transcription and organelle changes through macrophage transcriptome sequencing and ultrastructural transmission electron microscopy observation. According to the results of the transcriptome sequencing, ASFV infection led to significant alterations in the gene expression pattern of porcine bone marrow derived macrophages (BMDMs), with 2404 genes showing upregulation and 1579 genes downregulation. Cytokines, and chemokines were significant changes in the expression of BMDMs; there was significant activation of pattern recognition receptors such as Toll-like receptors and Nod-like receptors. According to the observation of the ultrastructure, mitochondrial damage and mitochondrial autophagy were widely present in ASFV-infected cells. The reduced number of macrophage pseudopodia suggested that virus-induced structural changes may compromise pathogen recognition, phagocytosis, and signal communication in macrophages. Additionally, the decreased size and inhibited acidification of secondary lysosomes in macrophages implied suppressed phagocytosis. Overall, ASFV infection resulted in significant changes in the expression of cytokines and chemokines, accompanied by the activation of NLR and TLR signaling pathways. We reported for the first time that ASFV infection led to a reduction in pseudopodia numbers and a decrease in the size and acidification of secondary lysosomes.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Cytokines , Macrophages , Animaux , Virus de la peste porcine africaine/génétique , Virus de la peste porcine africaine/ultrastructure , Virus de la peste porcine africaine/immunologie , Peste porcine africaine/virologie , Peste porcine africaine/immunologie , Suidae , Macrophages/virologie , Cytokines/génétique , Cytokines/métabolisme , Transcriptome , Phagocytose , Transduction du signal , Microscopie électronique à transmission , Mitochondries/ultrastructure
14.
Virulence ; 15(1): 2333562, 2024 12.
Article de Anglais | MEDLINE | ID: mdl-38622757

RÉSUMÉ

The Picornaviridae are a large group of positive-sense, single-stranded RNA viruses, and most research has focused on the Enterovirus genus, given they present a severe health risk to humans. Other picornaviruses, such as foot-and-mouth disease virus (FMDV) and senecavirus A (SVA), affect agricultural production with high animal mortality to cause huge economic losses. The 3Dpol protein of picornaviruses is widely known to be used for genome replication; however, a growing number of studies have demonstrated its non-polymerase roles, including modulation of host cell biological processes, viral replication complex assembly and localization, autophagy, and innate immune responses. Currently, there is no effective vaccine to control picornavirus diseases widely, and clinical therapeutic strategies have limited efficiency in combating infections. Many efforts have been made to develop different types of drugs to prohibit virus survival; the most important target for drug development is the virus polymerase, a necessary element for virus replication. For picornaviruses, there are also active efforts in targeted 3Dpol drug development. This paper reviews the interaction of 3Dpol proteins with the host and the progress of drug development targeting 3Dpol.


Sujet(s)
Enterovirus , Virus de la fièvre aphteuse , Infections à Picornaviridae , Animaux , Humains , Produits du gène pol/métabolisme , Virus de la fièvre aphteuse/génétique , Virus de la fièvre aphteuse/métabolisme , Réplication virale , ARN viral/génétique
15.
PLoS Pathog ; 20(3): e1012104, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38512977

RÉSUMÉ

The interaction between foot-and-mouth disease virus (FMDV) and the host is extremely important for virus infection, but there are few researches on it, which is not conducive to vaccine development and FMD control. In this study, we designed a porcine genome-scale CRISPR/Cas9 knockout library containing 93,859 single guide RNAs targeting 16,886 protein-coding genes, 25 long ncRNAs, and 463 microRNAs. Using this library, several previously unreported genes required for FMDV infection are highly enriched post-FMDV selection in IBRS-2 cells. Follow-up studies confirmed the dependency of FMDV on these genes, and we identified a functional role for one of the FMDV-related host genes: TOB1 (Transducer of ERBB2.1). TOB1-knockout significantly inhibits FMDV infection by positively regulating the expression of RIG-I and MDA5. We further found that TOB1-knockout led to more accumulation of mRNA transcripts of transcription factor CEBPA, and thus its protein, which further enhanced transcription of RIG-I and MDA5 genes. In addition, TOB1-knockout was shown to inhibit FMDV adsorption and internalization mediated by EGFR/ERBB2 pathway. Finally, the FMDV lethal challenge on TOB1-knockout mice confirmed that the deletion of TOB1 inhibited FMDV infection in vivo. These results identify TOB1 as a key host factor involved in FMDV infection in pigs.


Sujet(s)
Virus de la fièvre aphteuse , Fièvre aphteuse , Animaux , Souris , Récepteurs ErbB/métabolisme , Fièvre aphteuse/génétique , Virus de la fièvre aphteuse/génétique , Régulation de l'expression des gènes , , Suidae
16.
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
17.
J Virol ; 98(4): e0014624, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38440983

RÉSUMÉ

Peste des petits ruminants is an acute and highly contagious disease caused by the Peste des petits ruminants virus (PPRV). Host proteins play a crucial role in viral replication. However, the effect of fusion (F) protein-interacting partners on PPRV infection is poorly understood. In this study, we found that the expression of goat plasminogen activator urokinase (PLAU) gradually decreased in a time- and dose-dependent manner in PPRV-infected goat alveolar macrophages (GAMs). Goat PLAU was subsequently identified using co-immunoprecipitation and confocal microscopy as an F protein binding partner. The overexpression of goat PLAU inhibited PPRV growth and replication, whereas silencing goat PLAU promoted viral growth and replication. Additionally, we confirmed that goat PLAU interacted with a virus-induced signaling adapter (VISA) to antagonize F-mediated VISA degradation, increasing the production of type I interferon. We also found that goat PLAU reduced the inhibition of PPRV replication in VISA-knockdown GAMs. Our results show that the host protein PLAU inhibits the growth and replication of PPRV by VISA-triggering RIG-I-like receptors and provides insight into the host protein that antagonizes PPRV immunosuppression.IMPORTANCEThe role of host proteins that interact with Peste des petits ruminants virus (PPRV) fusion (F) protein in PPRV replication is poorly understood. This study confirmed that goat plasminogen activator urokinase (PLAU) interacts with the PPRV F protein. We further discovered that goat PLAU inhibited PPRV replication by enhancing virus-induced signaling adapter (VISA) expression and reducing the ability of the F protein to degrade VISA. These findings offer insights into host resistance to viral invasion and suggest new strategies and directions for developing PPR vaccines.


Sujet(s)
Maladies des chèvres , Capra , Interactions hôte-pathogène , Peste des petits ruminants , Virus de la peste des petits ruminants , Activateur du plasminogène de type urokinase , Protéines de fusion virale , Animaux , Protéines adaptatrices de la transduction du signal/métabolisme , Protéine-58 à domaine DEAD/métabolisme , Maladies des chèvres/immunologie , Maladies des chèvres/métabolisme , Maladies des chèvres/virologie , Capra/immunologie , Capra/virologie , Macrophages alvéolaires , Peste des petits ruminants/immunologie , Peste des petits ruminants/métabolisme , Peste des petits ruminants/virologie , Virus de la peste des petits ruminants/croissance et développement , Virus de la peste des petits ruminants/immunologie , Virus de la peste des petits ruminants/métabolisme , Liaison aux protéines , Activateur du plasminogène de type urokinase/génétique , Activateur du plasminogène de type urokinase/métabolisme , Protéines de fusion virale/métabolisme
18.
Clin Immunol ; 262: 110169, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38479440

RÉSUMÉ

The process of autophagy, a conservative evolutionary mechanism, is responsible for the removal of surplus and undesirable cytoplasmic components, thereby ensuring cellular homeostasis. Autophagy exhibits a remarkable level of selectivity by employing a multitude of cargo receptors that possess the ability to bind both ubiquitinated cargoes and autophagosomes. In the context of viral infections, selective autophagy plays a crucial role in regulating the innate immune system. Notably, numerous viruses have developed strategies to counteract, evade, or exploit the antiviral effects of selective autophagy. This review encompasses the latest research progress of selective autophagy in regulating innate immunity and virus infectious.


Sujet(s)
Maladies virales , Virus , Humains , Immunité innée , Autophagie/physiologie , Homéostasie
19.
Virology ; 595: 110056, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38552409

RÉSUMÉ

The Peste des petits ruminant virus (PPRV) is a member of the Paramyxoviridae family and is classified into the genus Measles virus. PPRV predominantly infects small ruminants, leading to mortality rates of nearly 100%, which have caused significant economic losses in developing countries. Host proteins are important in virus replication, but the PPRV nucleocapsid (N) protein-host interacting partners for regulating PPRV replication remain unclear. The present study confirmed the interaction between PPRV-N and the host protein vimentin by co-immunoprecipitation and co-localization experiments. Overexpression of vimentin suppressed PPRV replication, whereas vimentin knockdown had the opposite effect. Mechanistically, N was subjected to degradation via the ubiquitin/proteasome pathway, where vimentin recruits the E3 ubiquitin ligase NEDD4L to fulfill N-ubiquitination, resulting in the degradation of the N protein. These findings suggest that the host protein vimentin and E3 ubiquitin ligase NEDD4L have an anti-PPRV effect.


Sujet(s)
Protéines nucléocapside , Virus de la peste des petits ruminants , Vimentine , Réplication virale , Protéines nucléocapside/métabolisme , Protéines nucléocapside/génétique , Vimentine/métabolisme , Vimentine/génétique , Animaux , Virus de la peste des petits ruminants/physiologie , Virus de la peste des petits ruminants/génétique , Virus de la peste des petits ruminants/métabolisme , Humains , Ubiquitination , Interactions hôte-pathogène , Cellules HEK293 , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Lignée cellulaire , Peste des petits ruminants/virologie , Peste des petits ruminants/métabolisme , Liaison aux protéines
20.
Virol Sin ; 39(3): 378-389, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38499154

RÉSUMÉ

Foot-and-mouth disease (FMD) is a highly contagious and economically important disease, which is caused by the FMD virus (FMDV). Although the cell receptor for FMDV has been identified, the specific mechanism of FMDV internalization after infection remains unknown. In this study, we found that kinesin family member 5B (KIF5B) plays a vital role during FMDV internalization. Moreover, we confirmed the interaction between KIF5B and FMDV structural protein VP1 by co-immunoprecipitation (Co-IP) and co-localization in FMDV-infected cells. In particular, the stalk [amino acids (aa) 413-678] domain of KIF5B was indispensable for KIF5B-VP1 interaction. Moreover, overexpression of KIF5B dramatically enhanced FMDV replication; consistently, knockdown or knockout of KIF5B suppressed FMDV replication. Furthermore, we also demonstrated that KIF5B promotes the internalization of FMDV via regulating clathrin uncoating. KIF5B also promotes the transmission of viral particles to early and late endosomes during the early stages of infection. In conclusion, our results demonstrate that KIF5B promotes the internalization of FMDV via regulating clathrin uncoating and intracellular transport. This study may provide a new therapeutic target for developing FMDV antiviral drugs.


Sujet(s)
Virus de la fièvre aphteuse , Kinésine , Pénétration virale , Réplication virale , Kinésine/métabolisme , Kinésine/génétique , Virus de la fièvre aphteuse/physiologie , Virus de la fièvre aphteuse/génétique , Virus de la fièvre aphteuse/métabolisme , Animaux , Fièvre aphteuse/virologie , Fièvre aphteuse/métabolisme , Protéines de capside/métabolisme , Protéines de capside/génétique , Lignée cellulaire , Humains , Endosomes/métabolisme , Endosomes/virologie , Cellules HEK293
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE