Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.677
Filtrer
1.
Arch Virol ; 169(8): 172, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39096433

RÉSUMÉ

Goatpox and sheeppox are highly contagious and economically important viral diseases of small ruminants. Due to the risk they pose to animal health, livestock production, and international trade, capripoxviruses are a considerable threat to the livestock economy. In this study, we expressed two core proteins (A4L and A12L) and one extracellular enveloped virion protein (A33R) of goatpox virus in a baculovirus expression vector system and evaluated their use as diagnostic antigens in ELISA. Full-length A4L, A12L, and A33R genes of the GTPV Uttarkashi strain were amplified, cloned into the pFastBac HT A donor vector, and introduced into DH10Bac cells containing a baculovirus shuttle vector plasmid to generate recombinant bacmids. The recombinant baculoviruses were produced in Sf-21 cells by transfection, and proteins were expressed in TN5 insect cells. The recombinant proteins were analysed by SDS-PAGE and confirmed by western blot, with expected sizes of ~30 kDa, ~31 kDa, and ~32 kDa for A4L, A12L, and A33R, respectively. The recombinant proteins were purified, and the immunoreactivity of the purified proteins was confirmed by western blot using anti-GTPV serum. The antigenic specificity of the expressed proteins as diagnostic antigens was evaluated by testing their reactivity with infected, vaccinated, and negative GTPV/SPPV serum in indirect ELISA, and the A33R-based indirect ELISA was optimized. The diagnostic sensitivity and specificity of the A33R-based indirect ELISA were found to be of 89% and 94% for goats and 98% and 91%, for sheep, respectively. No cross-reactivity was observed with other related viruses. The recombinant-A33R-based indirect ELISA developed in the present study shows that it has potential for the detection of antibodies in GTPV and SPPV infected/vaccinated animals.


Sujet(s)
Baculoviridae , Capripoxvirus , Test ELISA , Maladies des chèvres , Capra , Protéines de l'enveloppe virale , Capripoxvirus/génétique , Capripoxvirus/isolement et purification , Baculoviridae/génétique , Animaux , Maladies des chèvres/virologie , Maladies des chèvres/diagnostic , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/immunologie , Capra/virologie , Test ELISA/méthodes , Infections à Poxviridae/diagnostic , Infections à Poxviridae/médecine vétérinaire , Infections à Poxviridae/virologie , Protéines recombinantes/génétique , Protéines recombinantes/isolement et purification , Protéines recombinantes/immunologie , Virion/génétique , Protéines du core viral/génétique , Protéines du core viral/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Cellules Sf9 , Antigènes viraux/génétique , Antigènes viraux/immunologie , Lignée cellulaire , Expression des gènes
2.
J Mol Model ; 30(8): 295, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39083139

RÉSUMÉ

CONTEXT: Flaviviruses cause severe encephalitic or hemorrhagic diseases in humans. Its members, Kyasanur forest disease virus (KFDV) and Alkhumra hemorrhagic fever virus (ALKV), cause hemorrhagic fever and are prevalent in India and Saudi Arabia, respectively, while the tick-borne encephalitis virus (TBEV) causes a dangerous encephalitic infection in Europe and Asia. However, little information is available about the targets of immune responses for these deadly viruses. Here, we predict potential antigenic peptide epitopes of viral envelope protein for inducing a cell-mediated and humoral immune response. METHODS: Using the Immune Epitope Database and Analysis Resource (IEDB-AR), we identified 13 MHC-I and two MHC-II dominant conserved epitopes in KFDV and ALKV and six MHC-I and three MHC-II epitopes in TBEV envelope proteins. Parallelly, we also predicted B-cell linear and discontinuous envelope protein epitopes for these viruses. Interestingly, the epitopes are conserved in all three viral envelope proteins. Further, the discontinuous epitopes are structurally compared with the available DENV, ZIKV, WNV, TBEV, and LIV envelope protein antibody structures. Overall structural comparison analyses highlight (i) lateral ridge epitope in the ED-III domain of E protein, and (ii) envelope dimer epitope (EDE) could be targeted for developing potent vaccine candidates as well as therapeutic antibody production. Moreover, existing structural and biochemical functions of the same epitopes in homologous viruses are predicted to have a reduced antibody-dependent enhancement (ADE) effect on flaviviral infection.


Sujet(s)
Flavivirus , Flavivirus/immunologie , Humains , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/composition chimique , Biologie informatique , Séquence d'acides aminés , Déterminants antigéniques des lymphocytes B/immunologie , Déterminants antigéniques des lymphocytes B/composition chimique , Similitude de séquences d'acides aminés , Épitopes/immunologie , Épitopes/composition chimique , Modèles moléculaires , Virus de l'encéphalite à tiques (sous-groupe)/immunologie
3.
Egypt J Immunol ; 31(3): 95-112, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38995715

RÉSUMÉ

In this study, we aimed to evaluate the immunogenic profile of a chimeric DNA-based hepatitis C virus (HCV) vaccine candidate encoding the full-length viral core-E1-E2 (HCV-CE) fragment. The vaccine candidate was designed to uniformly express the HCV genotype 4 core-E1-E2 protein. The recombinant HCV-CE protein was bacterially expressed in C41 (DE3) cells, and then BALB/c mice were immunized with different combinations of DNA/DNA or DNA/protein prime/boost immunizations. The proper construction of our vaccine candidate was confirmed by specific amplification of the encoded fragments and basic local alignment search tool (BLAST) results of the nucleotide sequence, which revealed a high degree of similarity with several HCV serotypes/genotypes. The platform for bacterial expression was optimized to maximize the yield of the purified recombinant HCV-CE protein. The recombinant protein showed high specific antigenicity against the sera of HCV-infected patients according to the ELISA and western blot results. The predicted B- and T-cell epitopes showed high antigenic and interferon-γ (IFN-γ) induction potential, in addition to cross-genotype conservation and population coverage. The mice antisera further demonstrated a remarkable ability to capture 100% of the native viral antigens circulating in the sera of HCV patients, with no cross-reactivity detected in control sera. In conclusion, the proposed HCV vaccination strategy demonstrated promising potential regarding its safety, immunogenicity, and population coverage.


Sujet(s)
Hepacivirus , Hépatite C , Souris de lignée BALB C , Vaccins à ADN , Vaccins contre les hépatites virales , Animaux , Hepacivirus/immunologie , Hepacivirus/génétique , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Souris , Vaccins contre les hépatites virales/immunologie , Hépatite C/prévention et contrôle , Hépatite C/immunologie , Humains , Immunogénicité des vaccins/immunologie , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Protéines du core viral/immunologie , Protéines du core viral/génétique , Femelle , Anticorps de l'hépatite C/immunologie , Anticorps de l'hépatite C/sang
4.
Virology ; 597: 110152, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38968676

RÉSUMÉ

Cobalt-porphyrin phospholipid displays recombinant protein antigens on liposome surfaces via antigen polyhistidine-tag (His-tag), and when combined with monophosphorylated lipid A and QS-21 yields the "CPQ" vaccine adjuvant system. In this proof of principle study, CPQ was used to generate vaccine prototypes that elicited antibodies for two different alphaviruses (AV). Mice were immunized with computationally designed, His-tagged, physicochemical property consensus (PCPcon) protein antigens representing the variable B-domain of the envelope protein 2 (E2) from the serotype specific Venezuelan Equine Encephalitis Virus (VEEVcon) or a broad-spectrum AV-antigen termed EVCcon. The CPQ adjuvant enhanced the antigenicity of both proteins without eliciting detectable anti-His-tag antibodies. Antibodies elicited from mice immunized with antigens admixed with CPQ showed orders-of-magnitude higher levels of antigen-specific IgG compared to alternative control adjuvants. The ELISA results correlated with antiviral activity against VEEV strain TC83 and more weakly to Chikungunya virus 118/25. Thus, display of E.coli-produced His-tagged E2 protein segments on the surface of immunogenic liposomes elicits high levels of antigen-specific and AV neutralizing antibodies in mice with vaccination, while facilitating vaccine preparation and providing dose-sparing potential.


Sujet(s)
Adjuvants immunologiques , Alphavirus , Anticorps antiviraux , Antigènes viraux , Liposomes , Protéines de l'enveloppe virale , Vaccins antiviraux , Animaux , Anticorps antiviraux/immunologie , Souris , Liposomes/immunologie , Alphavirus/immunologie , Antigènes viraux/immunologie , Protéines de l'enveloppe virale/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Adjuvants immunologiques/administration et posologie , Virus de l'encéphalite équine du Venezuela/immunologie , Femelle , Anticorps neutralisants/immunologie , Virus du chikungunya/immunologie , Souris de lignée BALB C , Immunoglobuline G/immunologie , Immunoglobuline G/sang
5.
Front Immunol ; 15: 1408510, 2024.
Article de Anglais | MEDLINE | ID: mdl-39021566

RÉSUMÉ

Equid alphaherpesvirus 1 (EqAHV1) is a viral pathogen known to cause respiratory disease, neurologic syndromes, and abortion storms in horses. Currently, there are no vaccines that provide complete protection against EqAHV1. Marker vaccines and the differentiation of infected and vaccinated animals (DIVA) strategy are effective for preventing and controlling outbreaks but have not been used for the prevention of EqAHV1 infection. Glycoprotein 2 (gp2), located on the envelope of viruses (EqAHV1), exhibits high antigenicity and functions as a molecular marker for DIVA. In this study, a series of EqAHV1 mutants with deletion of gp2 along with other virulence genes (TK, UL24/TK, gI/gE) were engineered. The mutant viruses were studied in vitro and then in an in vivo experiment using Golden Syrian hamsters to assess the extent of viral attenuation and the immune response elicited by the mutant viruses in comparison to the wild-type (WT) virus. Compared with the WT strain, the YM2019 Δgp2, ΔTK/gp2, and ΔUL24/TK/gp2 strains exhibited reduced growth in RK-13 cells, while the ΔgI/gE/gp2 strain exhibited significantly impaired proliferation. The YM2019 Δgp2 strain induced clinical signs and mortality in hamsters. In contrast, the YM2019 ΔTK/gp2 and ΔUL24/TK/gp2 variants displayed diminished pathogenicity, causing no observable clinical signs or fatalities. Immunization with nasal vaccines containing YM2019 ΔTK/gp2 and ΔUL24/TK/gp2 elicited a robust immune response in hamsters. In particular, compared with the vaccine containing the ΔTK/gp2 strain, the vaccine containing the ΔUL24/TK/gp2 strain demonstrated enhanced immune protection upon challenge with the WT virus. Furthermore, an ELISA for gp2 was established and refined to accurately differentiate between infected and vaccinated animals. These results confirm that the ΔUL24/TK/gp2 strain is a safe and effective live attenuated vaccine candidate for controlling EqAHV1 infection.


Sujet(s)
Infections à Herpesviridae , Herpèsvirus équin de type 1 , Vaccins atténués , Animaux , Vaccins atténués/immunologie , Infections à Herpesviridae/prévention et contrôle , Infections à Herpesviridae/immunologie , Infections à Herpesviridae/virologie , Infections à Herpesviridae/médecine vétérinaire , Herpèsvirus équin de type 1/immunologie , Herpèsvirus équin de type 1/génétique , Equus caballus , Mesocricetus , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Cricetinae , Maladies des chevaux/prévention et contrôle , Maladies des chevaux/immunologie , Maladies des chevaux/virologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Lignée cellulaire , Mutation
6.
Arch Virol ; 169(8): 167, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39020055

RÉSUMÉ

Epstein‒Barr virus (EBV) is a double-stranded DNA virus belonging to the family Orthoherpesviridae that is associated with the development of various tumors, such as lymphoma, nasopharyngeal carcinoma, and gastric cancer. There are no uniformly effective treatments for human EBV infection, and vaccines and immunotherapies are currently the main research directions. The glycoproteins gB and gH/gL are surface glycoproteins that are common to all herpesviruses, with subtle differences in structure and function between different viruses. The core membrane fusion machinery constituted by EBV gB and gH/gL is an important target of neutralizing antibodies in epithelial EBV infection due to its essential role in the fusion of viral and target cell membranes. In this article, we review the main modes of EBV infection, the structure and function of the core fusion machinery gB and gH/gL, and the development of neutralizing antibodies and prophylactic vaccines based on this target.


Sujet(s)
Anticorps neutralisants , Infections à virus Epstein-Barr , Herpèsvirus humain de type 4 , Protéines de l'enveloppe virale , Humains , Infections à virus Epstein-Barr/prévention et contrôle , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/virologie , Anticorps neutralisants/immunologie , Herpèsvirus humain de type 4/immunologie , Herpèsvirus humain de type 4/génétique , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Anticorps antiviraux/immunologie , Pénétration virale , Animaux , Vaccins antiviraux/immunologie , Protéines virales/immunologie , Protéines virales/génétique , Glycoprotéines membranaires , Chaperons moléculaires
7.
Sci Rep ; 14(1): 17645, 2024 07 31.
Article de Anglais | MEDLINE | ID: mdl-39085250

RÉSUMÉ

The Middle East has witnessed a greater spread of infectious Dengue viruses, with serotype 2 (DENV-2) being the most prevalent form. Through this work, multi-epitope peptide vaccines against DENV-2 that target E and nonstructural (NS1) proteins were generated through an immunoinformatic approach. MHC class I and II and LBL epitopes among NS1 and envelope E proteins sequences were predicted and their antigenicity, toxicity, and allergenicity were investigated. Studies of the population coverage denoted the high prevalence of NS1 and envelope-E epitopes among different countries where DENV-2 endemic. Further, both the CTL and HTL epitopes retrieved from NS1 epitopes exhibited high conservancies' percentages with other DENV serotypes (1, 3, and 4). Three vaccine constructs were created and the expected immune responses for the constructs were estimated using C-IMMSIM and HADDOCK (against TLR 2,3,4,5, and 7). Molecular dynamics simulation for vaccine construct 2 with TLR4 denoted high binding affinity and stability of the construct with the receptor which might foretell favorable in vivo interaction and immune responses.


Sujet(s)
Vaccins contre la dengue , Virus de la dengue , Dengue , Sérogroupe , Vaccins sous-unitaires , Protéines virales non structurales , Virus de la dengue/immunologie , Vaccins sous-unitaires/immunologie , Vaccins contre la dengue/immunologie , Humains , Dengue/prévention et contrôle , Dengue/immunologie , Dengue/virologie , Protéines virales non structurales/immunologie , Biologie informatique/méthodes , Déterminants antigéniques des lymphocytes T/immunologie , Protéines de l'enveloppe virale/immunologie , Simulation de dynamique moléculaire , Épitopes/immunologie , Épitopes/composition chimique ,
8.
PLoS One ; 19(7): e0301987, 2024.
Article de Anglais | MEDLINE | ID: mdl-38995916

RÉSUMÉ

Equid alphaherpesviruses 1 (EHV-1) and 4 (EHV-4) are closely related and both endemic in horses worldwide. Both viruses replicate in the upper respiratory tract, but EHV-1 may additionally lead to abortion and equine herpesvirus myeloencephalopathy (EHM). We focused on antibody responses in horses against the receptor-binding glycoprotein D of EHV-1 (gD1), which shares a 77% amino acid identity with its counterpart in EHV-4 (gD4). Both antigens give rise to cross-reacting antibodies, including neutralizing antibodies. However, immunity against EHV-4 is not considered protective against EHM. While a diagnostic ELISA to discriminate between EHV-1 and EHV-4 infections is available based on type-specific fragments of glycoprotein G (gG1 and gG4, respectively), the type-specific antibody reaction against gD1 has not yet been sufficiently addressed. Starting from the N-terminus of gD1, we developed luciferase immunoprecipitation system (LIPS) assays, using gD1-fragments of increasing size as antigens, i.e. gD1_83 (comprising the first 83 amino acids), gD1_160, gD1_180, and gD1_402 (the full-length molecule). These assays were then used to analyse panels of horse sera from Switzerland (n = 60) and Iceland (n = 50), the latter of which is considered EHV-1 free. We detected only one true negative horse serum from Iceland, whereas all other sera in both panels were seropositive for both gG4 (ELISA) and gD1 (LIPS against gD1_402). In contrast, seropositivity against gG1 was rather rare (35% Swiss sera; 14% Icelandic sera). Therefore, a high percentage of antibodies against gD1 could be attributed to cross-reaction and due to EHV-4 infections. In contrast, the gD1_83 fragment was able to identify sera with type-specific antibodies against gD1. Interestingly, those sera stemmed almost exclusively from vaccinated horses. Although it is uncertain that the N-terminal epitopes of gD1 addressed in this communication are linked to better protection, we suggest that in future vaccine developments, type-common antigens should be avoided, while a broad range of type-specific antigens should be favored.


Sujet(s)
Anticorps antiviraux , Herpèsvirus équin de type 1 , Maladies des chevaux , Protéines de l'enveloppe virale , Animaux , Equus caballus/immunologie , Herpèsvirus équin de type 1/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Protéines de l'enveloppe virale/immunologie , Maladies des chevaux/virologie , Maladies des chevaux/immunologie , Maladies des chevaux/prévention et contrôle , Herpèsvirus équin de type 4/immunologie , Infections à Herpesviridae/médecine vétérinaire , Infections à Herpesviridae/immunologie , Infections à Herpesviridae/virologie , Réactions croisées/immunologie , Test ELISA , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Domaines protéiques/immunologie
9.
Protein Expr Purif ; 222: 106543, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38971211

RÉSUMÉ

Dengue virus (DENV) is a considerable public health threat affecting millions of people globally. Vaccines for dengue are an important strategy to reduce the disease burden. We expressed capsid (C2) and envelope domain III of dengue virus serotype 2 (2EDIII) separately in the silkworm expression system. We conjugated them employing the monomeric streptavidin (mSA2) and biotin affinity to display the antigenic 2EDIII on the C2-forming capsid-like particle (CLP). Purified 2EDIII-displaying C2 (CLP/2EDIII) was immunogenic in BALB/c mice, eliciting neutralizing antibodies confirmed by a single-round infectious particle (SRIP) neutralization assay. Th1 cytokine levels were upregulated for the CLP/2EDIII group, and the anti-inflammatory IL-10 and pro-inflammatory IL-6 cytokine levels were also raised compared to the 2EDIII and the control groups. Elevated cytokine levels for CLP/2EDIII indicate the importance of displaying the 2EDIII as CLP/2EDIII rather than as an individual subunit. This study is the first to express the C2 protein as self-assembling CLP in vivo and 2EDIII separately in the silkworm expression system and conjugate them to form a monovalent CLP. Thus, this CLP/2EDIII display method may pave the way for an efficient tetravalent dengue vaccine candidate.


Sujet(s)
Anticorps neutralisants , Bombyx , Virus de la dengue , Souris de lignée BALB C , Protéines de l'enveloppe virale , Animaux , Bombyx/génétique , Bombyx/virologie , Bombyx/métabolisme , Virus de la dengue/génétique , Virus de la dengue/immunologie , Souris , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/composition chimique , Protéines de l'enveloppe virale/biosynthèse , Anticorps neutralisants/immunologie , Protéines de capside/génétique , Protéines de capside/immunologie , Protéines de capside/composition chimique , Protéines de capside/biosynthèse , Vaccins contre la dengue/immunologie , Vaccins contre la dengue/génétique , Anticorps antiviraux/immunologie , Dengue/immunologie , Dengue/virologie , Sérogroupe , Domaines protéiques , Femelle
10.
Commun Biol ; 7(1): 871, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39020082

RÉSUMÉ

Antibodies to Ebola virus glycoprotein (EBOV GP) represent an important correlate of the vaccine efficiency and infection survival. Both neutralization and some of the Fc-mediated effects are known to contribute the protection conferred by antibodies of various epitope specificities. At the same time, the role of the complement system remains unclear. Here, we compare complement activation by two groups of representative monoclonal antibodies (mAbs) interacting with the glycan cap (GC) or the membrane-proximal external region (MPER) of GP. Binding of GC-specific mAbs to GP induces complement-dependent cytotoxicity (CDC) in the GP-expressing cell line via C3 deposition on GP in contrast to MPER-specific mAbs. In the mouse model of EBOV infection, depletion of the complement system leads to an impairment of protection exerted by one of the GC-specific, but not MPER-specific mAbs. Our data suggest that activation of the complement system represents an important mechanism of antiviral protection by GC antibodies.


Sujet(s)
Anticorps monoclonaux , Anticorps antiviraux , Ebolavirus , Fièvre hémorragique à virus Ebola , Polyosides , Protéines de l'enveloppe virale , Animaux , Ebolavirus/immunologie , Anticorps monoclonaux/immunologie , Souris , Fièvre hémorragique à virus Ebola/immunologie , Fièvre hémorragique à virus Ebola/virologie , Fièvre hémorragique à virus Ebola/prévention et contrôle , Polyosides/immunologie , Anticorps antiviraux/immunologie , Humains , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/métabolisme , Activation du complément , Souris de lignée BALB C , Femelle , Protéines du système du complément/immunologie , Protéines du système du complément/métabolisme , Glycoprotéines/immunologie
11.
Viruses ; 16(7)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39066177

RÉSUMÉ

We recently demonstrated that Simian-HIV (SHIV)-infected neonate rhesus macaques (RMs) generated heterologous HIV-1 neutralizing antibodies (NAbs) with broadly-NAb (bNAb) characteristics at a higher frequency compared with their corresponding dam. Here, we characterized genetic diversity in Env sequences from four neonate or adult/dam RM pairs: in two pairs, neonate and dam RMs made heterologous HIV-1 NAbs; in one pair, neither the neonate nor the dam made heterologous HIV-1 NAbs; and in another pair, only the neonate made heterologous HIV-1 NAbs. Phylogenetic and sequence diversity analyses of longitudinal Envs revealed that a higher genetic diversity, within the host and away from the infecting SHIV strain, was correlated with heterologous HIV-1 NAb development. We identified 22 Env variable sites, of which 9 were associated with heterologous HIV-1 NAb development; 3/9 sites had mutations previously linked to HIV-1 Env bNAb development. These data suggested that viral diversity drives heterologous HIV-1 NAb development, and the faster accumulation of viral diversity in neonate RMs may be a potential mechanism underlying bNAb induction in pediatric populations. Moreover, these data may inform candidate Env immunogens to guide precursor B cells to bNAb status via vaccination by the Env-based selection of bNAb lineage members with the appropriate mutations associated with neutralization breadth.


Sujet(s)
Anticorps neutralisants , Évolution moléculaire , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Macaca mulatta , Phylogenèse , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Virus de l'immunodéficience simienne/génétique , Virus de l'immunodéficience simienne/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/classification , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Variation génétique , Animaux nouveau-nés , Anticorps anti-VIH/immunologie , Anticorps anti-VIH/sang , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/immunologie , Infections à VIH/virologie
12.
Front Immunol ; 15: 1438371, 2024.
Article de Anglais | MEDLINE | ID: mdl-39081314

RÉSUMÉ

Introduction: Variant pseudorabies virus (PRV) is a newly emerged zoonotic pathogen that can cause human blindness. PRV can take advantage of its large genome and multiple non-essential genes to construct recombinant attenuated vaccines carrying foreign genes. However, a major problem is that the foreign genes in recombinant PRV are only integrated into the genome for independent expression, rather than assembled on the surface of virion. Methods: We reported a recombinant PRV with deleted gE/TK genes and an inserted porcine circovirus virus 2 (PCV2) Cap gene into the extracellular domain of the PRV gE gene using the Cre-loxP recombinant system combined with the CRISPR-Cas9 gene editing system. This recombinant PRV (PRV-Cap), with the envelope-embedded Cap protein, exhibits a similar replication ability to its parental virus. Results: An immunogenicity assay revealed that PRV-Cap immunized mice have 100% resistance to lethal PRV and PCV2 attacks. Neutralization antibody and ELISPOT detections indicated that PRV-Cap can enhance neutralizing antibodies to PRV and produce IFN-γ secreting T cells specific for both PRV and PCV2. Immunological mechanistic investigation revealed that initial immunization with PRV-Cap stimulates significantly early activation and expansion of CD69+ T cells, promoting the activation of CD4 Tfh cell dependent germinal B cells and producing effectively specific effector memory T and B cells. Booster immunization with PRV-Cap recalled the activation of PRV-specific IFN-γ+IL-2+CD4+ T cells and IFN-γ+TNF-α+CD8+ T cells, as well as PCV2-specific IFN-γ+TNF-α+CD8+ T cells. Conclusion: Collectively, our data suggested an immunological mechanism in that the recombinant PRV with envelope-assembled PCV2 Cap protein can serve as an excellent vaccine candidate for combined immunity against PRV and PCV2, and provided a cost-effective method for the production of PRV- PCV2 vaccine.


Sujet(s)
Infections à Circoviridae , Circovirus , Herpèsvirus porcin de type 1 , Animaux , Circovirus/immunologie , Circovirus/génétique , Souris , Herpèsvirus porcin de type 1/immunologie , Herpèsvirus porcin de type 1/génétique , Infections à Circoviridae/immunologie , Infections à Circoviridae/prévention et contrôle , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Vaccins antiviraux/immunologie , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Suidae , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Protéines de capside/immunologie , Protéines de capside/génétique , Vaccins synthétiques/immunologie , Maladie d'Aujeszky/immunologie , Maladie d'Aujeszky/prévention et contrôle , Femelle , Souris de lignée BALB C
13.
Front Immunol ; 15: 1419634, 2024.
Article de Anglais | MEDLINE | ID: mdl-39081325

RÉSUMÉ

Herpes zoster (HZ), also known as shingles, remains a significant global health issue and most commonly seen in elderly individuals with an early exposure history to varicella-zoster virus (VZV). Currently, the licensed vaccine Shingrix, which comprises a recombinant VZV glycoprotein E (gE) formulated with a potent adjuvant AS01B, is the most effective shingles vaccine on the market. However, undesired reactogenicity and increasing global demand causing vaccine shortage, prompting the development of novel shingles vaccines. Here, we developed novel vaccine candidates utilising multiple nanoparticle (NP) platforms to display the recombinant gE antigen, formulated in an MF59-biosimilar adjuvant. In naïve mice, all tested NP vaccines induced higher humoral and cellular immune responses than Shingrix, among which, the gEM candidate induced the highest cellular response. In live attenuated VZV (VZV LAV)-primed mouse and rhesus macaque models, the gEM candidate elicited superior cell-mediated immunity (CMI) over Shingrix. Collectively, we demonstrated that NP technology remains a suitable tool for developing shingles vaccine, and the reported gEM construct is a highly promising candidate in the next-generation shingles vaccine development.


Sujet(s)
Vaccin contre le zona , Herpèsvirus humain de type 3 , Immunité cellulaire , Nanoparticules , Protéines de l'enveloppe virale , Animaux , Souris , Herpèsvirus humain de type 3/immunologie , Protéines de l'enveloppe virale/immunologie , Vaccin contre le zona/immunologie , Vaccin contre le zona/administration et posologie , Macaca mulatta , Zona/prévention et contrôle , Zona/immunologie , Femelle , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Adjuvants immunologiques/administration et posologie , Humains , Antigènes viraux/immunologie , Immunogénicité des vaccins , Souris de lignée BALB C ,
14.
J Virol ; 98(7): e0021324, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38832789

RÉSUMÉ

Human cytomegalovirus (HCMV) is a ß-herpesvirus that poses severe disease risk for immunocompromised patients who experience primary infection or reactivation. Development and optimization of safe and effective anti-HCMV therapeutics is of urgent necessity for the prevention and treatment of HCMV-associated diseases in diverse populations. The use of neutralizing monoclonal antibodies (mAbs) to limit HCMV infection poses a promising therapeutic strategy, as anti-HCMV mAbs largely inhibit infection by targeting virion glycoprotein complexes. In contrast, the small-molecule compounds currently approved for patients (e.g., ganciclovir, letermovir, and maribavir) target later stages of the HCMV life cycle. Here, we present a broadly neutralizing human mAb, designated 1C10, elicited from a VelocImmune mouse immunized with infectious HCMV particles. Clone 1C10 neutralizes infection after virion binding to cells by targeting gH/gL envelope complexes and potently reduces infection of diverse HCMV strains in fibroblast, trophoblast, and epithelial cells. Antibody competition assays found that 1C10 recognizes a region of gH associated with broad neutralization and binds to soluble pentamer in the low nanomolar range. Importantly, 1C10 treatment significantly reduced virus proliferation in both fibroblast and epithelial cells. Further, the combination treatment of mAb 1C10 with ganciclovir reduced HCMV infection and proliferation in a synergistic manner. This work characterizes a neutralizing human mAb for potential use as a HCMV treatment, as well as a possible therapeutic strategy utilizing combination-based treatments targeting disparate steps of the viral life cycle. Collectively, the findings support an antibody-based therapy to effectively treat patients at risk for HCMV-associated diseases. IMPORTANCE: Human cytomegalovirus is a herpesvirus that infects a large proportion of the population and can cause significant disease in diverse patient populations whose immune systems are suppressed or compromised. The development and optimization of safe anti-HCMV therapeutics, especially those that have viral targets and inhibition mechanisms different from current HCMV treatments, are of urgent necessity to better public health. Human monoclonal antibodies (mAbs) that prevent HCMV entry of cells were identified by immunizing transgenic mice and screened for broad and effective neutralization capability. Here, we describe one such mAb, which was found to target gH/gL envelope complexes and effectively limit HCMV infection and dissemination. Further, administration of the antibody in combination with the antiviral drug ganciclovir inhibited HCMV in a synergistic manner, highlighting this approach and the use of anti-HCMV mAbs more broadly, as a potential therapeutic strategy for the treatment of diverse patient populations.


Sujet(s)
Anticorps monoclonaux , Anticorps antiviraux , Infections à cytomégalovirus , Cytomegalovirus , Souris transgéniques , Protéines de l'enveloppe virale , Animaux , Humains , Cytomegalovirus/immunologie , Cytomegalovirus/effets des médicaments et des substances chimiques , Souris , Anticorps antiviraux/immunologie , Infections à cytomégalovirus/immunologie , Infections à cytomégalovirus/prévention et contrôle , Infections à cytomégalovirus/virologie , Protéines de l'enveloppe virale/immunologie , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/pharmacologie , Virion/immunologie , Fibroblastes/virologie , Réplication virale/effets des médicaments et des substances chimiques , Anticorps neutralisants à large spectre/immunologie , Antiviraux/pharmacologie , Immunisation
15.
J Virol ; 98(7): e0015524, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38832790

RÉSUMÉ

Marburg virus infection in humans is associated with case fatality rates that can reach up to 90%, but to date, there are no approved vaccines or monoclonal antibody (mAb) countermeasures. Here, we immunized Rhesus macaques with multivalent combinations of filovirus glycoprotein (GP) antigens belonging to Marburg, Sudan, and Ebola viruses to generate monospecific and cross-reactive antibody responses against them. From the animal that developed the highest titers of Marburg virus GP-specific neutralizing antibodies, we sorted single memory B cells using a heterologous Ravn virus GP probe and cloned and characterized a panel of 34 mAbs belonging to 28 unique lineages. Antibody specificities were assessed by overlapping pepscan and binding competition analyses, revealing that roughly a third of the lineages mapped to the conserved receptor binding region, including potent neutralizing lineages that were confirmed by negative stain electron microscopy to target this region. Additional lineages targeted a protective region on GP2, while others were found to possess cross-filovirus reactivity. Our study advances the understanding of orthomarburgvirus glycoprotein antigenicity and furthers efforts to develop candidate antibody countermeasures against these lethal viruses. IMPORTANCE: Marburg viruses were the first filoviruses characterized to emerge in humans in 1967 and cause severe hemorrhagic fever with average case fatality rates of ~50%. Although mAb countermeasures have been approved for clinical use against the related Ebola viruses, there are currently no approved countermeasures against Marburg viruses. We successfully isolated a panel of orthomarburgvirus GP-specific mAbs from a macaque immunized with a multivalent combination of filovirus antigens. Our analyses revealed that roughly half of the antibodies in the panel mapped to regions on the glycoprotein shown to protect from infection, including the host cell receptor binding domain and a protective region on the membrane-anchoring subunit. Other antibodies in the panel exhibited broad filovirus GP recognition. Our study describes the discovery of a diverse panel of cross-reactive macaque antibodies targeting orthomarburgvirus and other filovirus GPs and provides candidate immunotherapeutics for further study and development.


Sujet(s)
Anticorps monoclonaux , Anticorps neutralisants , Anticorps antiviraux , Réactions croisées , Macaca mulatta , Maladie de Marbourg , Marburgvirus , Animaux , Marburgvirus/immunologie , Anticorps antiviraux/immunologie , Anticorps neutralisants/immunologie , Anticorps monoclonaux/immunologie , Maladie de Marbourg/immunologie , Maladie de Marbourg/prévention et contrôle , Réactions croisées/immunologie , Glycoprotéines/immunologie , Protéines de l'enveloppe virale/immunologie , Immunisation , Humains , Ebolavirus/immunologie , Antigènes viraux/immunologie
16.
mBio ; 15(7): e0109224, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38847539

RÉSUMÉ

Herpes B virus (BV) is a zoonotic virus and belongs to the genus Simplexvius, the same genus as human herpes simplex virus (HSV). BV typically establishes asymptomatic infection in its natural hosts, macaque monkeys. However, in humans, BV infection causes serious neurological diseases and death. As such, BV research can only be conducted in a high containment level facility (i.e., biosafety level [BSL] 4), and the mechanisms of BV entry have not been fully elucidated. In this study, we generated a pseudotyped vesicular stomatitis virus (VSV) expressing BV glycoproteins using G-complemented VSV∆G system, which we named VSV/BVpv. We found that four BV glycoproteins (i.e., gB, gD, gH, and gL) were required for the production of a high-titer VSV/BVpv. Moreover, VSV/BVpv cell entry was dependent on the binding of gD to its cellular receptor nectin-1. Pretreatment of Vero cells with endosomal acidification inhibitors did not affect the VSV/BVpv infection. The result indicated that VSV/BVpv entry occurred by direct fusion with the plasma membrane of Vero cells and suggested that the entry pathway was similar to that of native HSV. Furthermore, we developed a VSV/BVpv-based chemiluminescence reduction neutralization test (CRNT), which detected the neutralization antibodies against BV in macaque plasma samples with high sensitivity and specificity. Crucially, the VSV/BVpv generated in this study can be used under BSL-2 condition to study the initial entry process through gD-nectin-1 interaction and the direct fusion of BV with the plasma membrane of Vero cells.IMPORTANCEHerpes B virus (BV) is a highly pathogenic zoonotic virus against humans. BV belongs to the genus Simplexvius, the same genus as human herpes simplex virus (HSV). By contrast to HSV, cell entry mechanisms of BV are not fully understood. The research procedures to manipulate infectious BV should be conducted in biosafety level (BSL)-4 facilities. As pseudotyped viruses provide a safe viral entry model because of their inability to produce infectious progeny virus, we tried to generate a pseudotyped vesicular stomatitis virus bearing BV glycoproteins (VSV/BVpv) by modification of expression constructs of BV glycoproteins, and successfully obtained VSV/BVpv with a high titer. This study has provided novel information for constructing VSV/BVpv and its usefulness to study BV infection.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Pénétration virale , Animaux , Anticorps neutralisants/immunologie , Chlorocebus aethiops , Cellules Vero , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Humains , Tests de neutralisation , Vesiculovirus/génétique , Vesiculovirus/immunologie , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/métabolisme , Glycoprotéines/génétique , Glycoprotéines/immunologie , Glycoprotéines/métabolisme , Virus de la stomatite vésiculeuse de type Indiana/génétique , Virus de la stomatite vésiculeuse de type Indiana/immunologie , Protéines virales/génétique , Protéines virales/immunologie , Protéines virales/métabolisme
17.
Arch Microbiol ; 206(7): 312, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38900285

RÉSUMÉ

Dengue virus (DENV) is the leading cause of numerous deaths every year due to its high infectivity. In this study we have tried to target the DENV envelope protein receptor binding domain, the region crucial for binding to host receptors which leads to membrane fusion and entry of the viral genome into the human host cell. We have taken 13 known FDA approved antiviral therapeutic antibodies from therapeutic antibody database and tried to repurpose them against the DENV envelope protein. Based on the humanness analysis, 10 antibodies were selected against the DENV envelope protein. Computational affinity maturation of the 10 selected antibodies was performed to increase their binding affinity and specificity against the DENV envelope protein which ultimately led to 8 mutant antibodies having better binding affinity than the native ones. Molecular Dynamics (MD) simulation shows that, the stability of the complexes involving both the native and mutant antibodies were found to be the same although the binding energy between the protein and the respective antibodies was seen to improve upon computational affinity maturation. Contact analyses show similar robustness of the interaction for both the mutant and native antibodies during complex formation with the DENV envelope protein. This has led to the selection of total 18 antibodies including 10 natural and 8 affinity matured mutants which have a high probability of interacting with the DENV envelope protein. Finally, based on all these analyses along with heated MD simulation, Bamlanivimab, Etesivimab and Tixagevimab with a mutation of residue 100 of the heavy chain from serine to tyrosine were selected as prospective therapeutic antibodies to combat DENV infection. This study may open a new avenue in designing therapeutics to combat Dengue viral infection.


Sujet(s)
Anticorps antiviraux , Virus de la dengue , Dengue , Simulation de dynamique moléculaire , Protéines de l'enveloppe virale , Virus de la dengue/immunologie , Virus de la dengue/génétique , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/métabolisme , Protéines de l'enveloppe virale/composition chimique , Humains , Anticorps antiviraux/immunologie , Dengue/immunologie , Dengue/traitement médicamenteux , Dengue/virologie , Antiviraux/pharmacologie , Repositionnement des médicaments , Liaison aux protéines
18.
Int J Pharm ; 660: 124320, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-38866086

RÉSUMÉ

Zika virus (ZIKV) is a mosquito-borne flavivirus that highly susceptibly causes Guillain-Barré syndrome and microcephaly in newborns. Vaccination is one of the most effective measures for preventing infectious diseases. However, there is currently no approved vaccine to prevent ZIKV infection. Here, we developed nanoparticle (NP) vaccines by covalently conjugating self-assembled 24-subunit ferritin to the envelope structural protein subunit of ZIKV to achieve antigen polyaggregation. The immunogenicityof the NP vaccine was evaluated in mice. Compared to monomer vaccines, the NP vaccine achieved effective antigen presentation, promoted the differentiation of follicular T helper cells in lymph nodes, and induced significantly greater antigen-specific humoral and cellular immune responses. Moreover, the NP vaccine enhanced high-affinity antigen-specific IgG antibody levels, increased secretion of the cytokines IL-4 and IFN-γ by splenocytes, significantly activated T/B lymphocytes, and improved the generation of memory T/B cells. In addition, no significant adverse reactions occurred when NP vaccine was combined with adjuvants. Overall, ferritin-based NP vaccines are safe and effective ZIKV vaccine candidates.


Sujet(s)
Ferritines , Nanoparticules , Vaccins antiviraux , Infection par le virus Zika , Virus Zika , Animaux , Virus Zika/immunologie , Infection par le virus Zika/prévention et contrôle , Infection par le virus Zika/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Femelle , Souris , Ferritines/immunologie , Immunogénicité des vaccins , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Immunité cellulaire/effets des médicaments et des substances chimiques , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Immunité humorale/effets des médicaments et des substances chimiques , Lymphocytes B/immunologie , Souris de lignée BALB C , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/administration et posologie ,
19.
Proc Natl Acad Sci U S A ; 121(24): e2400145121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38833465

RÉSUMÉ

Microalgae are promising production platforms for the cost-effective production of recombinant proteins. We have recently established that the red alga Porphyridium purpureum provides superior transgene expression properties, due to the episomal maintenance of transformation vectors as multicopy plasmids in the nucleus. Here, we have explored the potential of Porphyridium to synthesize complex pharmaceutical proteins to high levels. Testing expression constructs for a candidate subunit vaccine against the hepatitis C virus (HCV), we show that the soluble HCV E2 glycoprotein can be produced in transgenic algal cultures to high levels. The antigen undergoes faithful posttranslational modification by N-glycosylation and is recognized by conformationally selective antibodies, suggesting that it adopts a proper antigenic conformation in the endoplasmic reticulum of red algal cells. We also report the experimental determination of the structure of the N-glycan moiety that is attached to glycosylated proteins in Porphyridium. Finally, we demonstrate the immunogenicity of the HCV antigen produced in red algae when administered by injection as pure protein or by feeding of algal biomass.


Sujet(s)
Hepacivirus , Porphyridium , Porphyridium/métabolisme , Porphyridium/immunologie , Porphyridium/génétique , Hepacivirus/immunologie , Hepacivirus/génétique , Glycosylation , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/métabolisme , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , Protéines recombinantes/métabolisme , Animaux
20.
Front Immunol ; 15: 1352404, 2024.
Article de Anglais | MEDLINE | ID: mdl-38846950

RÉSUMÉ

Background: CD2v, a critical outer envelope glycoprotein of the African swine fever virus (ASFV), plays a central role in the hemadsorption phenomenon during ASFV infection and is recognized as an essential immunoprotective protein. Monoclonal antibodies (mAbs) targeting CD2v have demonstrated promise in both diagnosing and combating African swine fever (ASF). The objective of this study was to develop specific monoclonal antibodies against CD2v. Methods: In this investigation, Recombinant CD2v was expressed in eukaryotic cells, and murine mAbs were generated through meticulous screening and hybridoma cloning. Various techniques, including indirect enzyme-linked immunosorbent assay (ELISA), western blotting, immunofluorescence assay (IFA), and bio-layer interferometry (BLI), were employed to characterize the mAbs. Epitope mapping was conducted using truncation mutants and epitope peptide mapping. Results: An optimal antibody pair for a highly sensitive sandwich ELISA was identified, and the antigenic structures recognized by the mAbs were elucidated. Two linear epitopes highly conserved in ASFV genotype II strains, particularly in Chinese endemic strains, were identified, along with a unique glycosylated epitope. Three mAbs, 2B25, 3G25, and 8G1, effectively blocked CD2v-induced NF-κB activation. Conclusions: This study provides valuable insights into the antigenic structure of ASFV CD2v. The mAbs obtained in this study hold great potential for use in the development of ASF diagnostic strategies, and the identified epitopes may contribute to vaccine development against ASFV.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Anticorps monoclonaux , Cartographie épitopique , Facteur de transcription NF-kappa B , Animaux , Virus de la peste porcine africaine/immunologie , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/immunologie , Suidae , Souris , Peste porcine africaine/immunologie , Peste porcine africaine/virologie , Anticorps monoclonaux/immunologie , Protéines de l'enveloppe virale/immunologie , Épitopes/immunologie , Anticorps antiviraux/immunologie , Souris de lignée BALB C
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE