Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 304
Filter
1.
Methods Mol Biol ; 2854: 237-251, 2025.
Article in English | MEDLINE | ID: mdl-39192134

ABSTRACT

The innate immune system is the first line of host defense against infection by pathogenic microorganisms, among which macrophages are important innate immune cells. Macrophages are widely distributed throughout the body and recognize and eliminate viruses through pattern recognition receptors (PRRs) to sense pathogen-associated molecular patterns (PAMPs). In the present chapter, we provide detailed protocols for vesicular stomatitis virus (VSV) amplification, VSV titer detection, isolation of mouse primary peritoneal macrophages, in vitro and in vivo VSV infection, detection of interferon-beta (IFN-ß) expression, and lung injury. These protocols provide efficient and typical methods to evaluate virus-induced innate immunity in vitro and in vivo.


Subject(s)
Immunity, Innate , Interferon-beta , Macrophages, Peritoneal , Vesiculovirus , Animals , Mice , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/virology , Macrophages, Peritoneal/metabolism , Interferon-beta/immunology , Interferon-beta/metabolism , Interferon-beta/genetics , Vesiculovirus/immunology , Vesiculovirus/genetics , Vesicular Stomatitis/immunology , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/immunology , Receptors, Pattern Recognition/metabolism , Receptors, Pattern Recognition/immunology
2.
Proc Natl Acad Sci U S A ; 121(35): e2406421121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39159381

ABSTRACT

Viral infection is frequently assayed by ongoing expression of viral genes. These assays fail to identify cells that have been exposed to the virus but limit or inhibit viral replication. To address this limitation, we used a dual-labeling vesicular stomatitis virus (DL-VSV), which has a deletion of the viral glycoprotein gene, to allow evaluation of primary infection outcomes. This virus encodes Cre, which can stably mark any cell with even a minimal level of viral gene expression. Additionally, the virus encodes GFP, which distinguishes cells with higher levels of viral gene expression, typically due to genome replication. Stereotactic injections of DL-VSV into the murine brain showed that different cell types had very different responses to the virus. Almost all neurons hosted high levels of viral gene expression, while glial cells varied in their responses. Astrocytes (Sox9+) were predominantly productively infected, while oligodendrocytes (Sox10+) were largely abortively infected. Microglial cells (Iba1+) were primarily uninfected. Furthermore, we monitored the early innate immune response to viral infection and identified unique patterns of interferon (IFN) induction. Shortly after infection, microglia were the main producers of IFNb, whereas later, oligodendrocytes were the main producers. IFNb+ cells were primarily abortively infected regardless of cell type. Last, we investigated whether IFN signaling had any impact on the outcome of primary infection and did not observe significant changes, suggesting that intrinsic factors are likely responsible for determining the outcome of primary infection.


Subject(s)
Astrocytes , Animals , Mice , Astrocytes/virology , Astrocytes/metabolism , Virus Replication , Microglia/virology , Microglia/metabolism , Microglia/immunology , Neurons/virology , Neurons/metabolism , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Vesiculovirus/physiology , Vesiculovirus/immunology , Vesiculovirus/genetics , Oligodendroglia/virology , Oligodendroglia/metabolism , Vesicular Stomatitis/virology , Vesicular Stomatitis/immunology , Immunity, Innate , Mice, Inbred C57BL , Brain/virology , Brain/metabolism , Brain/immunology , Neuroglia/virology , Neuroglia/metabolism
3.
Nat Commun ; 15(1): 7267, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39179564

ABSTRACT

Targeted antineoplastic immunotherapies have achieved remarkable clinical outcomes. However, resistance to these therapies due to target absence or antigen shedding limits their efficacy and excludes tumours from candidacy. To address this limitation, here we engineer an oncolytic rhabdovirus, vesicular stomatitis virus (VSVΔ51), to express a truncated targeted antigen, which allows for HER2-targeting with trastuzumab. The truncated HER2 (HER2T) lacks signaling capabilities and is efficiently expressed on infected cell surfaces. VSVΔ51-mediated HER2T expression simulates HER2-positive status in tumours, enabling effective treatment with the antibody-drug conjugate trastuzumab emtansine in vitro, ex vivo, and in vivo. Additionally, we combine VSVΔ51-HER2T with an oncolytic vaccinia virus expressing a HER2-targeted T-cell engager. This dual-virus therapeutic strategy demonstrates potent curative efficacy in vivo in female mice using CD3+ infiltrate for anti-tumour immunity. Our findings showcase the ability to tailor the tumour microenvironment using oncolytic viruses, thereby enhancing compatibility with "off-the-shelf" targeted therapies.


Subject(s)
Immunotherapy , Oncolytic Virotherapy , Oncolytic Viruses , Receptor, ErbB-2 , T-Lymphocytes , Trastuzumab , Vaccinia virus , Animals , Female , Humans , Immunotherapy/methods , Mice , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/immunology , Receptor, ErbB-2/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , T-Lymphocytes/immunology , Cell Line, Tumor , Vaccinia virus/genetics , Vaccinia virus/immunology , Trastuzumab/therapeutic use , Trastuzumab/pharmacology , Tumor Microenvironment/immunology , Vesiculovirus/genetics , Vesiculovirus/immunology , Xenograft Model Antitumor Assays , Mice, Inbred BALB C
4.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126087

ABSTRACT

Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 in Angola. From 2021 to 2023, MARV outbreaks occurred in Guinea, Ghana, New Guinea, and Tanzania, emphasizing the expansion of its endemic area into new geographical regions. There are currently no approved vaccines or therapeutics targeting MARV, but several vaccine candidates have shown promise in preclinical studies. We compared three vaccine platforms simultaneously by vaccinating hamsters with either a single dose of an adenovirus-based (ChAdOx-1 MARV) vaccine, an alphavirus replicon-based RNA (LION-MARV) vaccine, or a recombinant vesicular stomatitis virus-based (VSV-MARV) vaccine, all expressing the MARV glycoprotein as the antigen. Lethal challenge with hamster-adapted MARV 4 weeks after vaccination resulted in uniform protection of the VSV-MARV and LION-MARV groups and 83% of the ChAdOx-1 MARV group. Assessment of the antigen-specific humoral response and its functionality revealed vaccine-platform-dependent differences, particularly in the Fc effector functions.


Subject(s)
Marburg Virus Disease , Marburgvirus , Viral Vaccines , Animals , Cricetinae , Viral Vaccines/immunology , Marburgvirus/immunology , Marburg Virus Disease/prevention & control , Marburg Virus Disease/immunology , Disease Models, Animal , Adenoviridae/genetics , Adenoviridae/immunology , Vesiculovirus/immunology , Vesiculovirus/genetics , Antibodies, Viral/immunology , Vaccination/methods
5.
Viruses ; 16(8)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39205155

ABSTRACT

Filoviruses, like the Marburg (MARV) and Ebola (EBOV) viruses, have caused outbreaks associated with significant hemorrhagic morbidity and high fatality rates. Vaccines offer one of the best countermeasures for fatal infection, but to date only the EBOV vaccine has received FDA licensure. Given the limited cross protection between the EBOV vaccine and Marburg hemorrhagic fever (MHF), we analyzed the protective efficacy of a similar vaccine, rVSV-MARV, in the lethal cynomolgus macaque model. NHPs vaccinated with a single dose (as little as 1.6 × 107 pfu) of rVSV-MARV seroconverted to MARV G-protein prior to challenge on day 42. Vaccinemia was measured in all vaccinated primates, self-resolved by day 14 post vaccination. Importantly, all vaccinated NHPs survived lethal MARV challenge, and showed no significant alterations in key markers of morbid disease, including clinical signs, and certain hematological and clinical chemistry parameters. Further, apart from one primate (from which tissues were not collected and no causal link was established), no pathology associated with Marburg disease was observed in vaccinated animals. Taken together, rVSV-MARV is a safe and efficacious vaccine against MHF in cynomolgus macaques.


Subject(s)
Macaca fascicularis , Marburg Virus Disease , Marburgvirus , Vesiculovirus , Viral Vaccines , Animals , Marburg Virus Disease/prevention & control , Marburg Virus Disease/immunology , Marburg Virus Disease/virology , Marburgvirus/immunology , Marburgvirus/genetics , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Vesiculovirus/genetics , Vesiculovirus/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Disease Models, Animal , Vaccination , Male , Female , Vaccine Efficacy , Genetic Vectors , Immunogenicity, Vaccine
6.
Front Cell Infect Microbiol ; 14: 1415695, 2024.
Article in English | MEDLINE | ID: mdl-39035358

ABSTRACT

Histone deacetylates family proteins have been studied for their function in regulating viral replication by deacetylating non-histone proteins. RIG-I (Retinoic acid-inducible gene I) is a critical protein in RNA virus-induced innate antiviral signaling pathways. Our previous research showed that HDAC8 (histone deacetylase 8) involved in innate antiviral immune response, but the underlying mechanism during virus infection is still unclear. In this study, we showed that HDAC8 was involved in the regulation of vesicular stomatitis virus (VSV) replication. Over-expression of HDAC8 inhibited while knockdown promoted VSV replication. Further exploration demonstrated that HDAC8 interacted with and deacetylated RIG-I, which eventually lead to enhance innate antiviral immune response. Collectively, our data clearly demonstrated that HDAC8 inhibited VSV replication by promoting RIG-I mediated interferon production and downstream signaling pathway.


Subject(s)
DEAD Box Protein 58 , Histone Deacetylases , Immunity, Innate , Receptors, Immunologic , Signal Transduction , Vesiculovirus , Virus Replication , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , Humans , Histone Deacetylases/metabolism , Vesiculovirus/immunology , Receptors, Immunologic/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Acetylation , HEK293 Cells , Interferons/metabolism , Interferons/immunology , Cell Line , Host-Pathogen Interactions/immunology , Animals , Vesicular stomatitis Indiana virus/immunology
7.
mBio ; 15(7): e0109224, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38847539

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Virus Internalization , Animals , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , Vero Cells , Antibodies, Viral/immunology , Antibodies, Viral/blood , Humans , Neutralization Tests , Vesiculovirus/genetics , Vesiculovirus/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Glycoproteins/genetics , Glycoproteins/immunology , Glycoproteins/metabolism , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
8.
Nat Commun ; 15(1): 5442, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937436

ABSTRACT

Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.


Subject(s)
Antigens, Neoplasm , B7-H1 Antigen , CD8-Positive T-Lymphocytes , Carcinoma, Hepatocellular , Liver Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Tumor Microenvironment , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Animals , Oncolytic Virotherapy/methods , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/immunology , Tumor Microenvironment/immunology , Mice , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Humans , Liver Neoplasms/therapy , Liver Neoplasms/immunology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Interferon-beta/metabolism , Interferon-beta/immunology , Mice, Inbred C57BL , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , T-Lymphocytes/immunology , Female , Vesiculovirus/immunology , Vesiculovirus/genetics
9.
mBio ; 15(7): e0056824, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38888342

ABSTRACT

The interferon (IFN) system protects mammals from diseases caused by virus infections. IFN synthesis is induced by pattern recognition receptor signaling pathways activated by virus infection. IFN is secreted from the infected cells and acts upon neighboring cells by binding cell surface receptors and triggering induction of hundreds of IFN-stimulated genes and proteins, many of which block different steps of virus replication. The IFN-induced tetratricopeptide repeat proteins (IFIT) are a family of RNA-binding proteins. We and others have previously reported that IFIT2 protects mice from many neurotropic RNA viruses; indeed, Ifit2-/- mice are very susceptible to intranasal or subcutaneous infections with vesicular stomatitis virus (VSV). Here, using a newly generated conditional knockout mouse, we report that ablation of Ifit2 expression only in neuronal cells was sufficient to render mice susceptible to neuropathogenesis caused by intranasal, but not subcutaneous, infection of VSV. Another genetically modified mouse line, expressing a mutant IFIT2 that cannot bind RNA, was as susceptible to VSV infection as Ifit2-/- mice. These results demonstrated that IFIT2 RNA-binding activity is essential for protecting mice against neurological diseases caused by intranasal infection of VSV.IMPORTANCEInterferon's (IFN's) antiviral effects are mediated by the proteins encoded by the interferon-stimulated genes. IFN-stimulated genes (IFIT2) is one such protein, which inhibits replication of many RNA viruses in the mouse brain and the resultant neuropathology. Our study sheds light on how IFIT2 works. By ablating Ifit2 expression only in neuronal cells, using a newly generated conditional knockout mouse line, we showed that Ifit2 induction in the neurons of the infected mouse was necessary for antiviral function of interferon. IFIT2 has no known enzyme activity; instead, it functions by binding to cellular or viral proteins or RNAs. We engineered a new mouse line that expressed a mutant IFIT2 that cannot bind RNA. These mice were very susceptible to infection with vesicular stomatitis virus indicating that the RNA-binding property of IFIT2 was essential for its antiviral function in vivo.


Subject(s)
Mice, Knockout , Neurons , RNA-Binding Proteins , Vesicular Stomatitis , Animals , Mice , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Neurons/virology , Neurons/metabolism , Vesicular Stomatitis/virology , Vesicular Stomatitis/immunology , Vesicular Stomatitis/genetics , Virus Replication , Vesiculovirus/immunology , Vesiculovirus/genetics , Mice, Inbred C57BL , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/genetics , Apoptosis Regulatory Proteins
10.
J Autoimmun ; 146: 103230, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38754237

ABSTRACT

Neonatal Fc receptor (FcRn) recycles immunoglobulin G, and inhibition of FcRn is used clinically for treatment of autoimmune diseases. In this work, using the vesicular stomatitis virus (VSV) mouse infection model system, we determined the role of FcRn during virus infection. While induction of neutralizing antibodies and long-term protection of these antibodies was hardly affected in FcRn deficient mice, FcRn deficiency limited the amount of natural IgG (VSV-specific) antibodies. Lack of natural antibodies (nAbs) limited early control of VSV in macrophages, accelerated propagation of virus in several organs, led to the spread of VSV to the neural tissue resulting in fatal outcomes. Adoptive transfer of natural IgG into FcRn deficient mice limited early propagation of VSV in FcRn deficient mice and enhanced survival of FcRn knockout mice. In line with this, vaccination of FcRn mice with very low dose of VSV prior to infection similarly prevented death after infection. In conclusion we determined the importance of nAbs during VSV infection. Lack of FcRn limited nAbs and thereby enhanced the susceptibility to virus infection.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Histocompatibility Antigens Class I , Immunoglobulin G , Mice, Knockout , Receptors, Fc , Vesicular Stomatitis , Animals , Mice , Immunoglobulin G/immunology , Receptors, Fc/immunology , Receptors, Fc/genetics , Receptors, Fc/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Vesicular Stomatitis/immunology , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , Vesiculovirus/immunology , Vesicular stomatitis Indiana virus/immunology , Disease Models, Animal , Adoptive Transfer , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL
11.
Front Biosci (Landmark Ed) ; 29(5): 195, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38812326

ABSTRACT

BACKGROUND: To investigate the immune responses and protection ability of ultraviolet light (UV)-inactivated recombinant vesicular stomatitis (rVSV)-based vectors that expressed a fusion protein consisting of four copies of the influenza matrix 2 protein ectodomain (tM2e) and the Dendritic Cell (DC)-targeting domain of the Ebola Glycoprotein (EΔM), (rVSV-EΔM-tM2e). METHOD: In our previous study, we demonstrated the effectiveness of rVSV-EΔM-tM2e to induce robust immune responses against influenza M2e and protect against lethal challenges from H1N1 and H3N2 strains. Here, we used UV to inactivate rVSV-EΔM-tM2e and tested its immunogenicity and protection in BALB/c mice from a mouse-adapted H1N1 influenza challenge. Using Enzyme-Linked Immunosorbent Assay (ELISA) and Antibody-Dependent Cellular Cytotoxicity (ADCC), the influenza anti-M2e immune responses specific to human, avian and swine influenza strains induced were characterized. Likewise, the specificity of the anti-M2e immune responses induced in recognizing M2e antigen on the surface of the cell was investigated using Fluorescence-Activated Cell Sorting (FACS) analysis. RESULTS: Like the live attenuated rVSV-EΔM-tM2e, the UV-inactivated rVSV-EΔM-tM2e was highly immunogenic against different influenza M2e from strains of different hosts, including human, swine, and avian, and protected against influenza H1N1 challenge in mice. The FACS analysis demonstrated that the induced immune responses can recognize influenza M2 antigens from human, swine and avian influenza strains. Moreover, the rVSV-EΔM-tM2e also induced ADCC activity against influenza M2e from different host strains. CONCLUSIONS: These findings suggest that UV-inactivated rVSV-EΔM-tM2e could be used as an inactivated vaccine against influenza viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Mice, Inbred BALB C , Orthomyxoviridae Infections , Ultraviolet Rays , Animals , Influenza Vaccines/immunology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Female , Mice , Humans , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Vesiculovirus/immunology , Vesiculovirus/genetics , Vaccines, Inactivated/immunology
12.
Emerg Infect Dis ; 30(5): 1004-1008, 2024 May.
Article in English | MEDLINE | ID: mdl-38666640

ABSTRACT

We evaluated the in vitro effects of lyophilization for 2 vesicular stomatitis virus-based vaccines by using 3 stabilizing formulations and demonstrated protective immunity of lyophilized/reconstituted vaccine in guinea pigs. Lyophilization increased stability of the vaccines, but specific vesicular stomatitis virus-based vaccines will each require extensive analysis to optimize stabilizing formulations.


Subject(s)
Disease Models, Animal , Freeze Drying , Vesicular Stomatitis , Viral Vaccines , Animals , Guinea Pigs , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Vesicular Stomatitis/immunology , Vesicular Stomatitis/prevention & control , Vesicular Stomatitis/virology , Vesiculovirus/immunology , Vesiculovirus/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Vaccine Efficacy , Vesicular stomatitis Indiana virus/immunology
14.
J Virol ; 96(18): e0133722, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36069551

ABSTRACT

COVID-19 and influenza are both highly contagious respiratory diseases that have been serious threats to global public health. It is necessary to develop a bivalent vaccine to control these two infectious diseases simultaneously. In this study, we generated three attenuated replicating recombinant vesicular stomatitis virus (rVSV)-based vaccine candidates against both SARS-CoV-2 and influenza viruses. These rVSV-based vaccines coexpress SARS-CoV-2 Delta spike protein (SP) bearing the C-terminal 17 amino acid (aa) deletion (SPΔC) and I742A point mutation, or the SPΔC with a deletion of S2 domain, or the RBD domain, and a tandem repeat harboring four copies of the highly conserved influenza M2 ectodomain (M2e) that fused with the Ebola glycoprotein DC-targeting/activation domain. Animal immunization studies have shown that these rVSV bivalent vaccines induced efficient humoral and cellular immune responses against both SARS-CoV-2 SP and influenza M2 protein, including high levels of neutralizing antibodies against SARS-CoV-2 Delta and other variant SP-pseudovirus infections. Importantly, immunization of the rVSV bivalent vaccines effectively protected hamsters or mice against the challenges of SARS-CoV-2 Delta variant and lethal H1N1 and H3N2 influenza viruses and significantly reduced respiratory viral loads. Overall, this study provides convincing evidence for the high efficacy of this bivalent vaccine platform to be used and/or easily adapted to produce new vaccines against new or reemerging SARS-CoV-2 variants and influenza A virus infections. IMPORTANCE Given that both COVID-19 and influenza are preferably transmitted through respiratory droplets during the same seasons, it is highly advantageous to develop a bivalent vaccine that could simultaneously protect against both COVID-19 and influenza. In this study, we generated the attenuated replicating recombinant vesicular stomatitis virus (rVSV)-based vaccine candidates that target both spike protein of SARS-Cov-2 Delta variant and the conserved influenza M2 domain. Importantly, these vaccine candidates effectively protected hamsters or mice against the challenges of SARS-CoV-2 Delta variant and lethal H1N1 and H3N2 influenza viruses and significantly reduced respiratory viral loads.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Influenza, Human , Vaccines, Combined , Vesicular Stomatitis , Amino Acids/genetics , Animals , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Cricetinae , Glycoproteins/genetics , Glycoproteins/immunology , Humans , Influenza A Virus, H3N2 Subtype , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Mice , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Combined/immunology , Vaccines, Synthetic/genetics , Vesiculovirus/immunology
15.
Proc Natl Acad Sci U S A ; 119(35): e2110105119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994646

ABSTRACT

The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the main target for neutralizing antibodies (NAbs). The S protein trimer is anchored in the virion membrane in its prefusion (preS) but metastable form. The preS protein has been stabilized by introducing two or six proline substitutions, to generate stabilized, soluble 2P or HexaPro (6P) preS proteins. Currently, it is not known which form is the most immunogenic. Here, we generated recombinant vesicular stomatitis virus (rVSV) expressing preS-2P, preS-HexaPro, and native full-length S, and compared their immunogenicity in mice and hamsters. The rVSV-preS-HexaPro produced and secreted significantly more preS protein compared to rVSV-preS-2P. Importantly, rVSV-preS-HexaPro triggered significantly more preS-specific serum IgG antibody than rVSV-preS-2P in both mice and hamsters. Antibodies induced by preS-HexaPro neutralized the B.1.1.7, B.1.351, P.1, B.1.427, and B.1.617.2 variants approximately two to four times better than those induced by preS-2P. Furthermore, preS-HexaPro induced a more robust Th1-biased cellular immune response than preS-2P. A single dose (104 pfu) immunization with rVSV-preS-HexaPro and rVSV-preS-2P provided complete protection against challenge with mouse-adapted SARS-CoV-2 and B.1.617.2 variant, whereas rVSV-S only conferred partial protection. When the immunization dose was lowered to 103 pfu, rVSV-preS-HexaPro induced two- to sixfold higher antibody responses than rVSV-preS-2P in hamsters. In addition, rVSV-preS-HexaPro conferred 70% protection against lung infection whereas only 30% protection was observed in the rVSV-preS-2P. Collectively, our data demonstrate that both preS-2P and preS-HexaPro are highly efficacious but preS-HexaPro is more immunogenic and protective, highlighting the advantages of using preS-HexaPro in the next generation of SARS-CoV-2 vaccines.


Subject(s)
Proline , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccine Development , Vesicular Stomatitis , Viral Vaccines , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/genetics , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/immunology , Cricetinae , Humans , Mice , Proline/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vesicular Stomatitis/immunology , Vesicular Stomatitis/prevention & control , Vesicular Stomatitis/virology , Vesiculovirus/immunology , Viral Proteins/immunology , Viral Vaccines/immunology
16.
Signal Transduct Target Ther ; 7(1): 22, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075101

ABSTRACT

As a highly pathogenic human coronavirus, SARS-CoV-2 has to counteract an intricate network of antiviral host responses to establish infection and spread. The nucleic acid-induced stress response is an essential component of antiviral defense and is closely related to antiviral innate immunity. However, whether SARS-CoV-2 regulates the stress response pathway to achieve immune evasion remains elusive. In this study, SARS-CoV-2 NSP5 and N protein were found to attenuate antiviral stress granule (avSG) formation. Moreover, NSP5 and N suppressed IFN expression induced by infection of Sendai virus or transfection of a synthetic mimic of dsRNA, poly (I:C), inhibiting TBK1 and IRF3 phosphorylation, and restraining the nuclear translocalization of IRF3. Furthermore, HEK293T cells with ectopic expression of NSP5 or N protein were less resistant to vesicular stomatitis virus infection. Mechanistically, NSP5 suppressed avSG formation and disrupted RIG-I-MAVS complex to attenuate the RIG-I-mediated antiviral immunity. In contrast to the multiple targets of NSP5, the N protein specifically targeted cofactors upstream of RIG-I. The N protein interacted with G3BP1 to prevent avSG formation and to keep the cofactors G3BP1 and PACT from activating RIG-I. Additionally, the N protein also affected the recognition of dsRNA by RIG-I. This study revealed the intimate correlation between SARS-CoV-2, the stress response, and innate antiviral immunity, shedding light on the pathogenic mechanism of COVID-19.


Subject(s)
Coronavirus 3C Proteases/genetics , Coronavirus Nucleocapsid Proteins/genetics , DEAD Box Protein 58/genetics , DNA Helicases/genetics , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , RNA-Binding Proteins/genetics , Receptors, Immunologic/genetics , SARS-CoV-2/genetics , Stress Granules/genetics , Animals , Chlorocebus aethiops , Coronavirus 3C Proteases/immunology , Coronavirus Nucleocapsid Proteins/immunology , DEAD Box Protein 58/immunology , DNA Helicases/immunology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Immune Evasion , Phosphoproteins/genetics , Phosphoproteins/immunology , Poly I-C/pharmacology , Poly-ADP-Ribose Binding Proteins/immunology , Protein Binding , RNA Helicases/immunology , RNA Recognition Motif Proteins/immunology , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA-Binding Proteins/immunology , Receptors, Immunologic/immunology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Sendai virus/genetics , Sendai virus/immunology , Signal Transduction , Stress Granules/drug effects , Stress Granules/immunology , Stress Granules/virology , Vero Cells , Vesiculovirus/genetics , Vesiculovirus/immunology
17.
Viruses ; 14(1)2022 01 05.
Article in English | MEDLINE | ID: mdl-35062293

ABSTRACT

The physiologic function of tripartite motif protein 56 (TRIM56), a ubiquitously expressed E3 ligase classified within the large TRIM protein family, remains elusive. Gene knockdown studies have suggested TRIM56 as a positive regulator of the type I interferon (IFN-I) antiviral response elicited via the Toll-like receptor 3 (TLR3) and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathways, which detect and respond to danger signals-extracellular double-stranded (ds) RNA and cytosolic dsDNA, respectively. However, to what extent these pathways depend on TRIM56 in human cells is unclear. In addition, it is debatable whether TRIM56 plays a part in controlling the expression of IFN-stimulated genes (ISGs) resulting from IFN-I based antiviral treatment. In this study, we created HeLa-derived TRIM56 null cell lines by gene editing and used these cell models to comprehensively examine the impact of endogenous TRIM56 on innate antiviral responses. Our results showed that TRIM56 knockout severely undermined the upregulation of ISGs by extracellular dsRNA and that loss of TRIM56 weakened the response to cytosolic dsDNA. ISG induction and ISGylation following IFN-α stimulation, however, were not compromised by TRIM56 deletion. Using a vesicular stomatitis virus-based antiviral bioactivity assay, we demonstrated that IFN-α could efficiently establish an antiviral state in TRIM56 null cells, providing direct evidence that TRIM56 is not required for the general antiviral action of IFN-I. Altogether, these data ascertain the contributions of TRIM56 to TLR3- and cGAS-STING-dependent antiviral pathways in HeLa cells and add to our understanding of the roles this protein plays in innate immunity.


Subject(s)
DNA/immunology , Interferon-alpha/immunology , RNA, Double-Stranded/immunology , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viruses/immunology , Animals , Chlorocebus aethiops , Cytosol/metabolism , HeLa Cells , Humans , Immunity, Innate , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Toll-Like Receptor 3/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Vero Cells , Vesiculovirus/immunology
18.
J Cell Biochem ; 123(2): 322-346, 2022 02.
Article in English | MEDLINE | ID: mdl-34729821

ABSTRACT

Chandipura vesiculovirus (CHPV) is a rapidly emerging pathogen responsible for causing acute encephalitis. Due to its widespread occurrence in Asian and African countries, this has become a global threat, and there is an urgent need to design an effective and nonallergenic vaccine against this pathogen. The present study aimed to develop a multi-epitope vaccine using an immunoinformatics approach. The conventional method of vaccine design involves large proteins or whole organism which leads to unnecessary antigenic load with increased chances of allergenic reactions. In addition, the process is also very time-consuming and labor-intensive. These limitations can be overcome by peptide-based vaccines comprising short immunogenic peptide fragments that can elicit highly targeted immune responses, avoiding the chances of allergenic reactions, in a relatively shorter time span. The multi-epitope vaccine constructed using CTL, HTL, and IFN-γ epitopes was able to elicit specific immune responses when exposed to the pathogen, in silico. Not only that, molecular docking and molecular dynamics simulation studies confirmed a stable interaction of the vaccine with the immune receptors. Several physicochemical analyses of the designed vaccine candidate confirmed it to be highly immunogenic and nonallergic. The computer-aided analysis performed in this study suggests that the designed multi-epitope vaccine can elicit specific immune responses and can be a potential candidate against CHPV.


Subject(s)
Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Molecular Docking Simulation , Molecular Dynamics Simulation , Vesiculovirus , Viral Vaccines , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Humans , Rhabdoviridae Infections/immunology , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Vesiculovirus/chemistry , Vesiculovirus/immunology , Viral Vaccines/chemistry , Viral Vaccines/immunology
19.
Front Immunol ; 12: 730483, 2021.
Article in English | MEDLINE | ID: mdl-34512666

ABSTRACT

The antiviral innate immunity is the first line of host defense against viral infection. Mitochondrial antiviral signaling protein (MAVS, also named Cardif/IPS-1/VISA) is a critical protein in RNA virus-induced antiviral signaling pathways. Our previous research suggested that E3 ubiquitin-protein ligases RING-finger protein (RNF90) negatively regulate cellular antiviral responses by targeting STING for degradation, though its role in RNA virus infection remains unknown. This study demonstrated that RNF90 negatively regulated RNA virus-triggered antiviral innate immune responses in RNF90-silenced PMA-THP1 cells, RNF90-deficient cells (including HaCaTs, MEFs, and BMDMs), and RNF90-deficient mice. However, RNF90 regulated RNA virus-triggered antiviral innate immune responses independent of STING. RNF90 promoted K48-linked ubiquitination of MAVS and its proteasome-dependent degradation, leading to the inhibition of innate immune responses. Altogether, our findings suggested a novel function and mechanism of RNF90 in antiviral innate immunity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Immunity, Innate , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Vesicular Stomatitis/metabolism , Vesiculovirus/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Chlorocebus aethiops , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , HEK293 Cells , HaCaT Cells , Host-Pathogen Interactions , Humans , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , THP-1 Cells , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology , Ubiquitination , Vero Cells , Vesicular Stomatitis/genetics , Vesicular Stomatitis/immunology , Vesicular Stomatitis/virology , Vesiculovirus/pathogenicity
20.
mBio ; 12(4): e0046321, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34340542

ABSTRACT

Oropouche virus (OROV) infection of humans is associated with a debilitating febrile illness that can progress to meningitis or encephalitis. First isolated from a forest worker in Trinidad and Tobago in 1955, the arbovirus OROV has since been detected throughout the Amazon basin with an estimated 500,000 human infections over 60 years. Like other members of the family Peribunyaviridae, the viral genome exists as 3 single-stranded negative-sense RNA segments. The medium-sized segment encodes a viral glycoprotein complex (GPC) that is proteolytically processed into two viral envelope proteins, Gn and Gc, responsible for attachment and membrane fusion. There are no therapeutics or vaccines to combat OROV infection, and we have little understanding of protective immunity to infection. Here, we generated a replication competent chimeric vesicular stomatitis virus (VSV), in which the endogenous glycoprotein was replaced by the GPC of OROV. Serum from mice immunized by intramuscular injection with VSV-OROV specifically neutralized wild-type OROV, and using peptide arrays we mapped multiple epitopes within an N-terminal variable region of Gc recognized by the immune sera. VSV-OROV lacking this variable region of Gc was also immunogenic in mice producing neutralizing sera that recognize additional regions of Gc. Challenge of both sets of immunized mice with wild-type OROV shows that the VSV-OROV chimeras reduce wild-type viral infection and suggest that antibodies that recognize the variable N terminus of Gc afford less protection than those that target more conserved regions of Gc. IMPORTANCE Oropouche virus (OROV), an orthobunyavirus found in Central and South America, is an emerging public health challenge that causes debilitating febrile illness. OROV is transmitted by arthropods, and increasing mobilization has the potential to significantly increase the spread of OROV globally. Despite this, no therapeutics or vaccines have been developed to combat infection. Using vesicular stomatitis (VSV) as a backbone, we developed a chimeric virus bearing the OROV glycoproteins (VSV-OROV) and tested its ability to elicit a neutralizing antibody response. Our results demonstrate that VSV-OROV produces a strong neutralizing antibody response that is at least partially targeted to the N-terminal region of Gc. Importantly, vaccination with VSV-OROV reduces viral loads in mice challenged with wild-type virus. These data provide novel evidence that targeting the OROV glycoproteins may be an effective vaccination strategy to combat OROV infection.


Subject(s)
Bunyaviridae Infections/prevention & control , Genome, Viral , Orthobunyavirus/genetics , Vesiculovirus/genetics , Vesiculovirus/immunology , Viral Envelope Proteins/genetics , Animals , Antibodies, Neutralizing , Bunyaviridae Infections/immunology , Male , Mice , Mice, Inbred C57BL , Vesicular Stomatitis/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL