Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21.015
Filter
1.
Nat Commun ; 15(1): 6548, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095394

ABSTRACT

Eastern equine encephalitis virus (EEEV) is the most virulent alphavirus that infects humans, and many survivors develop neurological sequelae, including paralysis and intellectual disability. Alphavirus spike proteins comprise trimers of heterodimers of glycoproteins E2 and E1 that mediate binding to cellular receptors and fusion of virus and host cell membranes during entry. We recently identified very-low density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2) as cellular receptors for EEEV and a distantly related alphavirus, Semliki Forest virus (SFV). Here, we use single-particle cryo-electron microscopy (cryo-EM) to determine structures of the EEEV and SFV spike glycoproteins bound to the VLDLR ligand-binding domain and found that EEEV and SFV interact with the same cellular receptor through divergent binding modes. Our studies suggest that the ability of LDLR-related proteins to interact with viral spike proteins through very small footprints with flexible binding modes results in a low evolutionary barrier to the acquisition of LDLR-related proteins as cellular receptors for diverse sets of viruses.


Subject(s)
Cryoelectron Microscopy , Encephalitis Virus, Eastern Equine , Receptors, LDL , Receptors, LDL/metabolism , Receptors, LDL/chemistry , Encephalitis Virus, Eastern Equine/metabolism , Encephalitis Virus, Eastern Equine/ultrastructure , Humans , Animals , Semliki forest virus/metabolism , Protein Binding , Receptors, Virus/metabolism , Receptors, Virus/chemistry , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/ultrastructure , Models, Molecular
2.
Arch Virol ; 169(8): 172, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39096433

ABSTRACT

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.


Subject(s)
Baculoviridae , Capripoxvirus , Enzyme-Linked Immunosorbent Assay , Goat Diseases , Goats , Viral Envelope Proteins , Capripoxvirus/genetics , Capripoxvirus/isolation & purification , Baculoviridae/genetics , Animals , Goat Diseases/virology , Goat Diseases/diagnosis , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Goats/virology , Enzyme-Linked Immunosorbent Assay/methods , Poxviridae Infections/diagnosis , Poxviridae Infections/veterinary , Poxviridae Infections/virology , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/immunology , Virion/genetics , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Sf9 Cells , Antigens, Viral/genetics , Antigens, Viral/immunology , Cell Line , Gene Expression
3.
Arch Virol ; 169(8): 167, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020055

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Viral Envelope Proteins , Humans , Epstein-Barr Virus Infections/prevention & control , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Antibodies, Neutralizing/immunology , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Antibodies, Viral/immunology , Virus Internalization , Animals , Viral Vaccines/immunology , Viral Proteins/immunology , Viral Proteins/genetics , Membrane Glycoproteins , Molecular Chaperones
4.
PLoS One ; 19(7): e0301987, 2024.
Article in English | MEDLINE | ID: mdl-38995916

ABSTRACT

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.


Subject(s)
Antibodies, Viral , Herpesvirus 1, Equid , Horse Diseases , Viral Envelope Proteins , Animals , Horses/immunology , Herpesvirus 1, Equid/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Envelope Proteins/immunology , Horse Diseases/virology , Horse Diseases/immunology , Horse Diseases/prevention & control , Herpesvirus 4, Equid/immunology , Herpesviridae Infections/veterinary , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Cross Reactions/immunology , Enzyme-Linked Immunosorbent Assay , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Protein Domains/immunology
5.
Methods Mol Biol ; 2824: 121-133, 2024.
Article in English | MEDLINE | ID: mdl-39039410

ABSTRACT

The Rift Valley fever virus (RVFV), transmitted through mosquito bites, leads to severe illness in humans and livestock throughout Africa and the Arabian Peninsula, causing significant morbidity and mortality. As of now, there are no verified and efficacious drugs or licensed vaccines accessible for the prevention or treatment of RVFV infections in both humans and livestock. The mature RVFV virion has two envelope proteins on its surface: glycoprotein N (GN) and glycoprotein C (GC). These proteins play a significant role in facilitating the virus's entry into the host cell, making them prominent targets for entry mechanism research as well as targets for drugs and vaccine development. The initial stage in obtaining atomic-resolution structural and mechanistic information on viral entry as well as developing biochemical and biophysical research tools involves recombinant protein production. In this chapter, we describe a simplified and scalable protocol facilitating the generation of high-quality, high-titer baculovirus virus for expression and purification of RVFV GC, utilizing the baculovirus-mediated expression system in insect cells.


Subject(s)
Baculoviridae , Recombinant Proteins , Rift Valley fever virus , Viral Envelope Proteins , Baculoviridae/genetics , Animals , Viral Envelope Proteins/genetics , Viral Envelope Proteins/isolation & purification , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Rift Valley fever virus/genetics , Sf9 Cells , Gene Expression , Humans , Genetic Vectors/genetics , Cloning, Molecular/methods
6.
Arch Virol ; 169(7): 155, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951272

ABSTRACT

Given the high prevalence of avian leukosis virus subgroup K (ALV-K) in chickens in China, the positive rate of ALV-K in local chickens in Henan province was investigated, and the genetic region encoding the glycoprotein gp85 of isolates from positive chickens was analyzed. The positive rate of ALV-K in local chickens in Henan was found to be 87.2% (41/47). Phylogenetic analysis of gp85 sequences revealed six clusters that differed in their host range regions (hr1 and hr2) and variable regions (vr1, vr2, and vr3). Evidence of recombination of hr1, hr2, vr1, vr2, and vr3 was observed between the different clusters. The isolate HN23LS02 appears to have obtained its hr1 and hr2 regions from separate lineages via recombination but without having a significant affect on the replication capacity of the virus.


Subject(s)
Avian Leukosis Virus , Avian Leukosis , Chickens , Host Specificity , Phylogeny , Poultry Diseases , Recombination, Genetic , Viral Envelope Proteins , Animals , Avian Leukosis Virus/genetics , Avian Leukosis Virus/classification , Avian Leukosis Virus/isolation & purification , Chickens/virology , Avian Leukosis/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Poultry Diseases/virology , China
7.
Virology ; 597: 110152, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38968676

ABSTRACT

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.


Subject(s)
Adjuvants, Immunologic , Alphavirus , Antibodies, Viral , Antigens, Viral , Liposomes , Viral Envelope Proteins , Viral Vaccines , Animals , Antibodies, Viral/immunology , Mice , Liposomes/immunology , Alphavirus/immunology , Antigens, Viral/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Encephalitis Virus, Venezuelan Equine/immunology , Female , Antibodies, Neutralizing/immunology , Chikungunya virus/immunology , Mice, Inbred BALB C , Immunoglobulin G/immunology , Immunoglobulin G/blood
8.
Front Immunol ; 15: 1408510, 2024.
Article in English | MEDLINE | ID: mdl-39021566

ABSTRACT

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.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Equid , Vaccines, Attenuated , Animals , Vaccines, Attenuated/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/immunology , Herpesvirus 1, Equid/genetics , Horses , Mesocricetus , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Cricetinae , Horse Diseases/prevention & control , Horse Diseases/immunology , Horse Diseases/virology , Viral Vaccines/immunology , Viral Vaccines/genetics , Cell Line , Mutation
9.
Antiviral Res ; 228: 105955, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964614

ABSTRACT

High levels of hepatitis B virus (HBV) surface antigen (HBsAg) in the blood of chronic HBV carriers are considered to drive the exhaustion of antigen-specific T and B lymphocytes and thus responsible for the persistence of infection. Accordingly, therapeutic elimination of HBsAg may facilitate the activation of adaptive antiviral immune responses against HBV and achieve a functional cure of chronic hepatitis B. We discovered recently that an amphipathic alpha helix spanning W156 to R169 of HBV small envelope (S) protein plays an essential role in the morphogenesis of subviral particles (SVPs) and metabolism of S protein. We thus hypothesized that pharmacological disruption of SVP morphogenesis may induce intracellular degradation of S protein and reduce HBsAg secretion. To identify inhibitors of SVP biogenesis, we screened 4417 bioactive compounds with a HepG2-derived cell line expressing HBV S protein and efficiently secreting small spherical SVPs. The screen identified 24 compounds that reduced intracellular SVPs and secreted HBsAg in a concentration-dependent manner. However, 18 of those compounds inhibited the secretion of HBsAg and HBeAg in HBV replicon transfected HepG2 cells at similar efficiency, suggesting each of those compounds may disrupt a common cellular function required for the synthesis and/or secretion of these viral proteins. Interestingly, lycorine more efficiently inhibited the secretion of HBsAg in HepG2 cells transfected with HBV replicons, HepG2.2.15 cells and HBV infected - HepG2 cells expressing sodium taurocholate cotransporting polypeptide (NTCP). The structure activity relationship and antiviral mechanism of lycorine against HBV have been determined.


Subject(s)
Antiviral Agents , Hepatitis B Surface Antigens , Hepatitis B virus , Humans , Hepatitis B virus/drug effects , Antiviral Agents/pharmacology , Hepatitis B Surface Antigens/metabolism , Hep G2 Cells , Virus Assembly/drug effects , Virion/drug effects , Drug Discovery , Virus Replication/drug effects , Small Molecule Libraries/pharmacology , Viral Envelope Proteins/metabolism , Hepatitis B e Antigens/metabolism
10.
Egypt J Immunol ; 31(3): 95-112, 2024 07.
Article in English | MEDLINE | ID: mdl-38995715

ABSTRACT

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.


Subject(s)
Hepacivirus , Hepatitis C , Mice, Inbred BALB C , Vaccines, DNA , Viral Hepatitis Vaccines , Animals , Hepacivirus/immunology , Hepacivirus/genetics , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Mice , Viral Hepatitis Vaccines/immunology , Hepatitis C/prevention & control , Hepatitis C/immunology , Humans , Immunogenicity, Vaccine/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Viral Core Proteins/immunology , Viral Core Proteins/genetics , Female , Hepatitis C Antibodies/immunology , Hepatitis C Antibodies/blood
11.
Protein Expr Purif ; 222: 106543, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38971211

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing , Bombyx , Dengue Virus , Mice, Inbred BALB C , Viral Envelope Proteins , Animals , Bombyx/genetics , Bombyx/virology , Bombyx/metabolism , Dengue Virus/genetics , Dengue Virus/immunology , Mice , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/biosynthesis , Antibodies, Neutralizing/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Capsid Proteins/chemistry , Capsid Proteins/biosynthesis , Dengue Vaccines/immunology , Dengue Vaccines/genetics , Antibodies, Viral/immunology , Dengue/immunology , Dengue/virology , Serogroup , Protein Domains , Female
12.
Sci Adv ; 10(27): eadl1888, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959313

ABSTRACT

We present structures of three immature tick-borne encephalitis virus (TBEV) isolates. Our atomic models of the major viral components, the E and prM proteins, indicate that the pr domains of prM have a critical role in holding the heterohexameric prM3E3 spikes in a metastable conformation. Destabilization of the prM furin-sensitive loop at acidic pH facilitates its processing. The prM topology and domain assignment in TBEV is similar to the mosquito-borne Binjari virus, but is in contrast to other immature flavivirus models. These results support that prM cleavage, the collapse of E protein ectodomains onto the virion surface, the large movement of the membrane domains of both E and M, and the release of the pr fragment from the particle render the virus mature and infectious. Our work favors the collapse model of flavivirus maturation warranting further studies of immature flaviviruses to determine the sequence of events and mechanistic details driving flavivirus maturation.


Subject(s)
Encephalitis Viruses, Tick-Borne , Viral Envelope Proteins , Encephalitis Viruses, Tick-Borne/physiology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Models, Molecular , Flavivirus/physiology , Animals , Virion , Encephalitis, Tick-Borne/virology , Humans
13.
J Virol ; 98(7): e0049924, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38953631

ABSTRACT

Tibroviruses are novel rhabdoviruses detected in humans, cattle, and arthropods. Four tibroviruses are known to infect humans: Bas-Congo virus (BASV), Ekpoma virus 1 (EKV-1), Ekpoma virus 2, and Mundri virus. However, since none of them has been isolated, their biological properties are largely unknown. We aimed to characterize the human tibrovirus glycoprotein (G), which likely plays a pivotal role in viral tropism and pathogenicity. Human tibrovirus Gs were found to share some primary structures and display 14 conserved cysteine residues, although their overall amino acid homology was low (29%-48%). Multiple potential glycosylation sites were found on the G molecules, and endoglycosidase H- and peptide-N-glycosidase F-sensitive glycosylation was confirmed. AlphaFold-predicted three-dimensional (3D) structures of human tibrovirus Gs were overall similar. Membrane fusion mediated by these tibrovirus Gs was induced by acidic pH. The low pH-induced conformational change that triggers fusion was reversible. Virus-like particles (VLPs) were produced by transient expression of Gs in cultured cells and used to produce mouse antisera. Using vesicular stomatitis Indiana virus pseudotyped with Gs, we found that the antisera to the respective tibrovirus VLPs showed limited cross-neutralizing activity. It was also found that human C-type lectins and T-cell immunoglobulin mucin 1 acted as attachment factors for G-mediated entry into cells. Interestingly, BASV-G showed the highest ability to utilize these molecules. The viruses infected a wide range of cell lines with preferential tropism for human-derived cells whereas the preference of EKV-1 was unique compared with the other human tibroviruses. These findings provide fundamental information to understand the biological properties of the human tibroviruses. IMPORTANCE: Human tibroviruses are poorly characterized emerging rhabdoviruses associated with either asymptomatic infection or severe disease with a case fatality rate as high as 60% in humans. However, the extent and burden of human infection as well as factors behind differences in infection outcomes are largely unknown. In this study, we characterized human tibrovirus glycoproteins, which play a key role in virus-host interactions, mainly focusing on their structural and antigenic differences and cellular tropism. Our results provide critical information for understanding the biological properties of these novel viruses and for developing appropriate preparedness interventions such as diagnostic tools, vaccines, and effective therapies.


Subject(s)
Viral Envelope Proteins , Humans , Animals , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Mice , Glycosylation , Virus Internalization , Viral Tropism , Cell Line , Mucin-1/metabolism , HEK293 Cells , Antibodies, Viral/immunology , Amino Acid Sequence
14.
Viruses ; 16(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39066177

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing , Evolution, Molecular , HIV-1 , Macaca mulatta , Phylogeny , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Simian Acquired Immunodeficiency Syndrome/virology , HIV-1/genetics , HIV-1/immunology , HIV-1/classification , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Genetic Variation , Animals, Newborn , HIV Antibodies/immunology , HIV Antibodies/blood , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , HIV Infections/virology
15.
Front Immunol ; 15: 1438371, 2024.
Article in English | MEDLINE | ID: mdl-39081314

ABSTRACT

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.


Subject(s)
Circoviridae Infections , Circovirus , Herpesvirus 1, Suid , Animals , Circovirus/immunology , Circovirus/genetics , Mice , Herpesvirus 1, Suid/immunology , Herpesvirus 1, Suid/genetics , Circoviridae Infections/immunology , Circoviridae Infections/prevention & control , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Swine , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Capsid Proteins/immunology , Capsid Proteins/genetics , Vaccines, Synthetic/immunology , Pseudorabies/immunology , Pseudorabies/prevention & control , Female , Mice, Inbred BALB C
16.
Front Immunol ; 15: 1419634, 2024.
Article in English | MEDLINE | ID: mdl-39081325

ABSTRACT

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.


Subject(s)
Herpes Zoster Vaccine , Herpesvirus 3, Human , Immunity, Cellular , Nanoparticles , Viral Envelope Proteins , Animals , Mice , Herpesvirus 3, Human/immunology , Viral Envelope Proteins/immunology , Herpes Zoster Vaccine/immunology , Herpes Zoster Vaccine/administration & dosage , Macaca mulatta , Herpes Zoster/prevention & control , Herpes Zoster/immunology , Female , Antibodies, Viral/immunology , Antibodies, Viral/blood , Adjuvants, Immunologic/administration & dosage , Humans , Antigens, Viral/immunology , Immunogenicity, Vaccine , Mice, Inbred BALB C , Nanovaccines
17.
Front Cell Infect Microbiol ; 14: 1404431, 2024.
Article in English | MEDLINE | ID: mdl-39081866

ABSTRACT

Introduction: Endogenous retroviruses (ERVs), which originated from exogenous retroviral infections of germline cells millions of years ago and were inherited by subsequent generations as per Mendelian inheritance patterns, predominantly comprise non-protein-coding sequences due to the accumulation of mutations, insertions, deletions, and truncations. Nevertheless, recent studies have revealed that ERVs play a crucial role in diverse biological processes by encoding various proteins. Methods: In this study, we successfully identified an ERV envelope (env) gene in a mink species. A phylogenetic tree of mink ERV-V env and reference sequences was constructed using Bayesian methods and maximum-likelihood inference. Results: Phylogenetic analyses indicated a significant degree of sequence conservation and positive selection within the env-surface open reading frame. Additionally, qRT-PCR revealed diverse patterns of mink ERV-V env expression in various tissues. The expression of mink ERV-V env gene in testicular tissue strongly correlated with the seasonal reproductive cycles of minks. Discussion: Our study suggests that the ERV-V env gene in mink may have been repurposed for host functions.


Subject(s)
Endogenous Retroviruses , Mink , Phylogeny , Endogenous Retroviruses/genetics , Animals , Mink/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Seasons , Reproduction/genetics , Male , Testis/virology , Bayes Theorem
18.
Commun Biol ; 7(1): 871, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020082

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Ebolavirus , Hemorrhagic Fever, Ebola , Polysaccharides , Viral Envelope Proteins , Animals , Ebolavirus/immunology , Antibodies, Monoclonal/immunology , Mice , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/prevention & control , Polysaccharides/immunology , Antibodies, Viral/immunology , Humans , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Complement Activation , Mice, Inbred BALB C , Female , Complement System Proteins/immunology , Complement System Proteins/metabolism , Glycoproteins/immunology
19.
J Mol Model ; 30(8): 295, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39083139

ABSTRACT

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.


Subject(s)
Flavivirus , Flavivirus/immunology , Humans , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Computational Biology , Amino Acid Sequence , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Sequence Homology, Amino Acid , Epitopes/immunology , Epitopes/chemistry , Models, Molecular , Encephalitis Viruses, Tick-Borne/immunology
20.
Virus Res ; 347: 199430, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38964470

ABSTRACT

A multistep priming process involving furin and endosomal cathepsin B and L (CatB/L) has been described for the Orthoebolavirus zairense (EBOV) glycoprotein GP. Inhibition or knockdown of either furin or endosomal cathepsins, however, did not prevent virus multiplication in cell cultures. Moreover, an EBOV mutant lacking the furin cleavage motif (RRTRR→AGTAA) was able to replicate and cause fatal disease in nonhuman primates, indicating that furin cleavage may be dispensable for virus infectivity. Here, by using protease inhibitors and EBOV GP-carrying recombinant vesicular stomatitis virus (VSV) and transcription and replication-competent virus-like particles (trVLPs) we found that processing of EBOV GP is mediated by different proteases in different cell lines depending on the protease repertoire available. Endosomal cathepsins were essential for EBOV GP entry in Huh-7 but not in Vero cells, in which trypsin-like proteases and stably expressed trypsin-like transmembrane serine protease 2 (TMPRSS2) supported wild-type EBOV GP and EBOV GP_AGTAA mutant entry. Furthermore, we show that the EBOV GP_AGTAA mutant is cleaved into fusion-competent GP2 by TMPRSS2 and by CatL at a so far unknown site. Fluorescence microscopy co-localization studies indicate that EBOV GP cleavage by TMPRSS2 may occur in the TGN prior to virus release or in the late endosome at the stage of virus entry into a new cell. Our data show that EBOV GP must be proteolytically activated to support virus entry but has even greater flexibility in terms of proteases and the precise cleavage site than previously assumed.


Subject(s)
Cathepsin L , Ebolavirus , Furin , Serine Endopeptidases , Viral Envelope Proteins , Virus Internalization , Cathepsin L/metabolism , Cathepsin L/genetics , Furin/metabolism , Furin/genetics , Ebolavirus/genetics , Ebolavirus/physiology , Ebolavirus/metabolism , Animals , Humans , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Chlorocebus aethiops , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Proteolysis , Vero Cells , Cell Line , Endosomes/metabolism , Endosomes/virology
SELECTION OF CITATIONS
SEARCH DETAIL