Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.059
Filter
1.
Methods Mol Biol ; 2854: 9-18, 2025.
Article in English | MEDLINE | ID: mdl-39192113

ABSTRACT

Antiviral innate immunity is the first line of defence against viruses. The interferon (IFN) signaling pathway, the DNA damage response (DDR), apoptosis, endoplasmic reticulum (ER) stress, and autophagy are involved in antiviral innate immunity. Viruses abrogate the antiviral immune response of cells to replication in various ways. Viral genes/proteins play a key role in evading antiviral innate immunity. Here, we will discuss the interference of viruses with antiviral innate immunity and the strategy for identifying viral gene/protein immune evasion.


Subject(s)
Immunity, Innate , Humans , Viral Proteins/immunology , Viral Proteins/genetics , Viruses/immunology , Viruses/genetics , Immune Evasion , Virus Diseases/immunology , Virus Diseases/virology , Animals , Genes, Viral , Autophagy/immunology , Host-Pathogen Interactions/immunology , Signal Transduction/immunology
2.
Methods Mol Biol ; 2854: 213-220, 2025.
Article in English | MEDLINE | ID: mdl-39192132

ABSTRACT

Yeast two-hybrid (YTH) technology is a powerful tool for studying protein interactions and has been widely used in various fields of molecular biology, including the study of antiviral innate immunity. This chapter presents detailed information and experimental procedures for identifying virus-host protein interactions involved in immune regulation using yeast two-hybrid technology.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate , Two-Hybrid System Techniques , Humans , Host-Pathogen Interactions/immunology , Viral Proteins/immunology , Viral Proteins/metabolism , Saccharomyces cerevisiae/immunology , Saccharomyces cerevisiae/genetics , Protein Binding , Protein Interaction Mapping/methods
3.
Nat Commun ; 15(1): 8712, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379405

ABSTRACT

Influenza viruses cause substantial morbidity and mortality every year despite seasonal vaccination. mRNA-based vaccines have the potential to elicit more protective immune responses, but for maximal breadth and durability, it is desirable to deliver both the viral hemagglutinin and neuraminidase glycoproteins. Delivering multiple antigens individually, however, complicates manufacturing and increases cost, thus it would be beneficial to express both proteins from a single mRNA. Here, we develop an mRNA genetic configuration that allows the simultaneous expression of unmodified, full-length NA and HA proteins from a single open reading frame. We apply this approach to glycoproteins from contemporary influenza A and B viruses and, after vaccination, observe high levels of functional antibodies and protection from disease in female mouse and male ferret challenge models. This approach may further efforts to utilize mRNA technology to improve seasonal vaccine efficacy by efficiently delivering multiple viral antigens simultaneously and in their native state.


Subject(s)
Antibodies, Viral , Ferrets , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , RNA, Messenger , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Female , Mice , Male , Neuraminidase/immunology , Neuraminidase/genetics , Antibodies, Viral/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Mice, Inbred BALB C , Influenza B virus/immunology , Influenza B virus/genetics , Influenza A virus/immunology , Influenza A virus/genetics , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Glycoproteins/immunology , Glycoproteins/genetics , Viral Proteins/immunology , Viral Proteins/genetics , Antigens, Viral/immunology , Antigens, Viral/genetics , Vaccination/methods
4.
J Gen Virol ; 105(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39392059

ABSTRACT

The emergence of Singapore grouper iridovirus (SGIV) has caused huge losses to grouper farming. SGIV is a DNA virus and belongs to the genus Ranavirus. Groupers infected with SGIV showed haemorrhaging and swelling of the spleen, with a mortality rate of more than 90% within a week. Therefore, it is of great significance to study the escape mechanism of SGIV from host innate immunity for the prevention and treatment of viral diseases in grouper. In this study, the viral proteins that interact with EccGAS were identified by mass spectrometry, and the SGIV VP12 protein that inhibits cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-mediated antiviral innate immunity was screened by the dual-luciferase reporter gene assay. VP12 belongs to the late gene of the virus. The immunofluorescence analysis demonstrated that VP12 was aggregated and distributed in the cytoplasm during the early stage of virus infection and translocated into the nucleus at the late stage of virus infection. VP12 inhibited the activation of IFN3, ISRE and NF-κB promoter activities mediated by cGAS-STING, EcTBK1 and EcIRF3. Quantitative real-time PCR analysis showed that VP12 inhibited the expression of interferon-related genes, including those mediated by cGAS-STING. VP12 enhanced the inhibition of IFN3, ISRE and NF-κB promoter activity by EccGAS, EccGAS-mab-21 and EccGAS-delete-mab21. The interaction between VP12 and EccGAS was found to be domain independent. The immunoprecipitation results demonstrated that VP12 interacted and co-localized with EccGAS, EcTBK1 and EcIRF3. VP12 degraded the protein levels of EcTBK1 and EcIRF3 and degraded EcIRF3 through the protease pathway. These results suggest that SGIV VP12 protein escapes the cGAS-STING signalling pathway and degrades EcIRF3 protein expression through the protease pathway.


Subject(s)
DNA Virus Infections , Immunity, Innate , Membrane Proteins , Nucleotidyltransferases , Ranavirus , Signal Transduction , Animals , Ranavirus/immunology , Ranavirus/physiology , DNA Virus Infections/immunology , DNA Virus Infections/virology , DNA Virus Infections/veterinary , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Fish Diseases/immunology , Fish Diseases/virology , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Immune Evasion , Host-Pathogen Interactions/immunology
5.
Virus Res ; 349: 199459, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39237037

ABSTRACT

Avian hepatitis E virus (HEV) has resulted in significant economic losses in the poultry industry. There is currently no commercial vaccination available to prevent avian HEV infection. Previously, a novel epitope (601TFPS604) was discovered in the ORF2 protein of avian HEV. In this study, peptides were synthesized and assessed for their ability to provide immunoprotecting against avian HEV infection in poultry. Twenty-five Hy-Line Variety Brown laying hens were randomly divided into five groups; groups 1 to 3 respectively immunized with RLLDRLSRTFPS, PETRRLLDRLSR (irrelevant peptide control), or truncated avian HEV ORF2 protein (aa 339-606), while group 4 (negative control) was mock-immunized with PBS and group 5 (normal control) was not immunized or challenged. After the challenge, all hens in groups 2 and 4 showed seroconversion, fecal virus shedding, viremia, alanine aminotransferase (ALT) level increasing, liver lesions and HEV antigen in the liver. There were no pathogenic effects in other groups. Collectively, all of these findings showed that hens were completely protected against avian HEV infection when they were immunized with the peptide containing TFPS of the avian HEV ORF2 protein.


Subject(s)
Chickens , Hepatitis, Viral, Animal , Hepevirus , Poultry Diseases , Viral Proteins , Animals , Poultry Diseases/prevention & control , Poultry Diseases/virology , Poultry Diseases/immunology , Hepevirus/immunology , Hepevirus/genetics , Hepatitis, Viral, Animal/prevention & control , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/virology , Viral Proteins/immunology , Viral Proteins/genetics , Viral Hepatitis Vaccines/immunology , Female , Peptides/immunology , Peptides/chemical synthesis , Peptides/genetics , Virus Shedding , RNA Virus Infections/prevention & control , RNA Virus Infections/veterinary , RNA Virus Infections/immunology , Viral Vaccines/immunology , Liver/virology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Feces/virology
6.
J Med Virol ; 96(9): e29923, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39291820

ABSTRACT

Arthropod-borne viruses, such as dengue virus (DENV), pose significant global health threats, with DENV alone infecting around 400 million people annually and causing outbreaks beyond endemic regions. This study aimed to enhance serological diagnosis and discover new drugs by identifying immunogenic protein regions of DENV. Utilizing a comprehensive approach, the study focused on peptides capable of distinguishing DENV from other flavivirus infections through serological analyses. Over 200 patients with confirmed arbovirus infection were profiled using high-density pan flavivirus peptide arrays comprising 6253 peptides and the computational method matrix of local coupling energy (MLCE). Twenty-four peptides from nonstructural and structural viral proteins were identified as specifically recognized by individuals with DENV infection. Six peptides were confirmed to distinguish DENV from Zika virus (ZIKV), West Nile virus (WNV), Yellow Fever virus (YFV), Usutu virus (USUV), and Chikungunya virus (CHIKV) infections, as well as healthy controls. Moreover, the combination of two immunogenic peptides emerged as a potential serum biomarker for DENV infection. These peptides, mapping to highly accessible regions on protein structures, show promise for diagnostic and prophylactic strategies against flavivirus infections. The described methodology holds broader applicability in the serodiagnosis of infectious diseases.


Subject(s)
Flavivirus Infections , Flavivirus , Protein Array Analysis , Humans , Flavivirus Infections/diagnosis , Flavivirus Infections/immunology , Flavivirus/immunology , Protein Array Analysis/methods , Peptides/immunology , Vaccine Development , Computational Biology/methods , Dengue/diagnosis , Dengue/immunology , Dengue/blood , Dengue Virus/immunology , Dengue Virus/genetics , High-Throughput Screening Assays/methods , Serologic Tests/methods , Biomarkers/blood , Viral Proteins/immunology , Adult , Antibodies, Viral/blood , Middle Aged , Male , Female , Zika Virus/immunology
7.
Viruses ; 16(9)2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39339941

ABSTRACT

African swine fever (ASF) is an acute infectious disease with a high mortality rate in both domestic and wild boars. Commercial vaccines or antiviral drugs for ASF were not available due to the complex diversity of the structure and genome of its pathogen African swine fever virus (ASFV). In recent years, there have been many reports on candidate strains of attenuated vaccines for ASFV. In this study, we obtained a recombinant virus named SY18ΔL60LΔCD2v by simultaneously deleting the L60L gene and CD2v gene from highly virulent strain SY18. In vitro, SY18ΔL60LΔCD2v displayed a decreased growth kinetic compared to that of parental SY18. In vivo, high doses (105 TCID50) of SY18ΔL60LΔCD2v can protect pigs (5/5) from attacks by the parental SY18 strain (102 TCID50). Low doses (102 TCID50) of SY18ΔL60LΔCD2v only protected 20% of pigs (1/5) from attacks by the parental SY18 strain (102 TCID50). The results indicated that the absence of these two genes in SY18 could induce protection against the homologous parental strain, and there were no obvious clinical symptoms or viremia. These results indicate that the SY18ΔL60LΔCD2v strain can serve as a new live attenuated vaccine candidate for the prevention and control of ASFV infection.


Subject(s)
African Swine Fever Virus , African Swine Fever , Gene Deletion , Vaccines, Attenuated , Viral Vaccines , African Swine Fever Virus/genetics , African Swine Fever Virus/immunology , African Swine Fever Virus/pathogenicity , Animals , Swine , African Swine Fever/prevention & control , African Swine Fever/virology , African Swine Fever/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/administration & dosage , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Proteins/genetics , Viral Proteins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viremia/prevention & control
8.
Viruses ; 16(9)2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39339948

ABSTRACT

Equid alphaherpesvirus 1 (EHV-1) has been linked to the emergence of neurological disorders, with the horse racing industry experiencing significant impacts from outbreaks of equine herpesvirus myeloencephalopathy (EHM). Building robust immune memory before pathogen exposure enables rapid recognition and elimination, preventing infection. This is crucial for effectively managing EHV-1. Removing neuropathogenic factors and immune evasion genes to develop live attenuated vaccines appears to be a successful strategy for EHV-1 vaccines. We created mutant viruses without ORF38 and ORF37/38 and validated their neuropathogenicity and immunogenicity in hamsters. The ∆ORF38 strain caused brain tissue damage at high doses, whereas the ∆ORF37/38 strain did not. Dexamethasone was used to confirm latent herpesvirus infection and reactivation. Dexamethasone injection increased viral DNA load in the brains of hamsters infected with the parental and ∆ORF38 strains, but not in those infected with the ∆ORF37/38 strain. Immunizing hamsters intranasally with the ∆ORF37/38 strain as a live vaccine produced a stronger immune response compared to the ∆ORF38 strain at the same dose. The hamsters demonstrated effective protection against a lethal challenge with the parental strain. This suggests that the deletion of ORF37/38 may effectively inhibit latent viral infection, reduce the neuropathogenicity of EHV-1, and induce a protective immune response.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Equid , Vaccines, Attenuated , Animals , Cricetinae , Female , Brain/virology , Brain/pathology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesviridae Infections/immunology , Herpesvirus 1, Equid/genetics , Herpesvirus 1, Equid/immunology , Herpesvirus 1, Equid/pathogenicity , Horse Diseases/virology , Horse Diseases/prevention & control , Horse Diseases/immunology , Horses , Latent Infection/immunology , Latent Infection/virology , Mesocricetus , Open Reading Frames , Sequence Deletion , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/administration & dosage , Viral Load , Viral Proteins/genetics , Viral Proteins/immunology , Virus Latency , Rabbits
9.
Hum Vaccin Immunother ; 20(1): 2403175, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39291424

ABSTRACT

Influenza neuraminidase (NA) is a promising target for a broadly protective vaccine. In this study, the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology was used to develop N2 NA vaccine candidates. The unique wild type (WT) N2 sequences of human and swine influenza strains isolated between 1957 and 2019 were used to design the COBRA N2-A NA vaccine, while the unique WT N2 sequences of human influenza strains isolated between 2000 and 2019 were used to design the COBRA N2-B NA vaccine. Sera collected from COBRA N2 NA vaccinated mice showed more broadly reactive antibody responses against a broad panel of H×N2 influenza virus strains than sera collected from mice vaccinated with WT N2 NA vaccines. Antibodies elicited by COBRA or WT N2 NA antigens cross react with recent human H3N2 influenza viruses from different clades, while the antibodies elicited by A/Switzerland/9715293/2013 hemagglutinin (HA) reacted with viruses from the same clade. Furthermore, mice vaccinated with COBRA N2-B NA vaccine had lower viral lung titers compared to mock vaccinated mice when challenged with human H3N2 influenza viruses. Thus, the COBRA N2 NA vaccines elicit broadly protective murine anti-NA antibodies against multiple strains across subtypes and the viral loads were significantly decreased in the lungs of the mice in the COBRA N2 NA vaccine groups, compared to the mice in the mock vaccinated group, indicating that the COBRA-based N2 subtype NA vaccines have a potential to be a component in a universal influenza vaccine.


Subject(s)
Antibodies, Viral , Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Animals , Female , Humans , Mice , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cross Reactions/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Influenza, Human/immunology , Lung/virology , Lung/immunology , Mice, Inbred BALB C , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Viral Load , Viral Proteins/immunology
10.
Front Cell Infect Microbiol ; 14: 1427588, 2024.
Article in English | MEDLINE | ID: mdl-39318476

ABSTRACT

P32 protein serves as a crucial structural component of Goat pox virus (GTPV), which causes a highly virulent infectious disease in sheep and goats. Despite the fact that P32 has been widely expressed in the previous studies, it is difficult to obtain recombinant P32 efficiently. This study aimed to achieve soluble expression of P32 recombinant protein and to develop its specific monoclonal antibody. The gene fragment of P32Δ (GP32Δ) was synthesized by optimizing the coding sequence of amino acids 1-246 of the known goatpox P32 protein. Subsequently, GP32Δ was cloned into a prokaryotic expression vector for expression and purification, resulting in the successful production of soluble recombinant protein rP32Δ. Utilizing rP32Δ, an indirect ELISA method was established by immunizing 6-week-old BALB/c mice with inactivated GTPV as the antigen. Through hybridoma technology, three monoclonal antibody hybridoma cell lines secreting anti-goat pox virus rP32Δ were screened, designated as 2F3, 3E8, and 4H5, respectively. These monoclonal antibodies, classified as IgG1, IgG2a, and IgG2b, respectively, with κappa light chains, were characterized following ascites preparation and purification. Indirect ELISA results demonstrated that the ELISA potency of the three monoclonal antibodies exceeded 1:12800. Furthermore, Western blot analysis revealed specific reactivity of both 3E8 and 4H5 with rP32Δ, while immunofluorescence assays confirmed 3E8's ability to specifically recognize GTPV in cells. The preceding findings demonstrate the successful acquisition of the soluble expressed recombinant P32 protein and its specific monoclonal antibody 3E8 in this study, thereby laying a foundational material basis for the establishment of a GTPV detection method.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Capripoxvirus , Enzyme-Linked Immunosorbent Assay , Goats , Mice, Inbred BALB C , Recombinant Proteins , Animals , Antibodies, Monoclonal/immunology , Capripoxvirus/genetics , Capripoxvirus/immunology , Antibodies, Viral/immunology , Mice , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Hybridomas , Immunoglobulin G , Gene Expression , Viral Proteins/genetics , Viral Proteins/immunology , Poxviridae Infections/immunology , Female , Goat Diseases/virology , Cloning, Molecular
11.
J Virol ; 98(9): e0120624, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39207137

ABSTRACT

The signal sequences of the human cytomegalovirus (CMV) UL40 protein and its rhesus CMV (RhCMV) counterpart, Rh67, contain a peptide (VMAPRT[L/V][F/I/L/V]L, VL9) that is presented by major histocompatibility complex (MHC) antigen E (MHC-E). The CMV VL9 peptides replace VL9 peptides derived from classical MHC (Ia) signal sequences, which are lost when CMV disrupts antigen processing and presentation and MHC Ia expression. This allows infected cells to maintain MHC-E surface expression and escape killing by Natural Killer cells. We demonstrate that processing of the Rh67 VL9 peptide mirrors that of UL40, despite the lack of sequence conservation between the two proteins. Processing of both VL9 peptides is dependent on cleavage of their signal sequences by the host protease signal peptide peptidase. As previously shown for UL40, up-regulation of MHC-E expression by Rh67 requires only its signal sequence, with sequences upstream of VL9 critical for conferring independence from TAP, the transporter associated with antigen processing. Our results also suggest that the mature UL40 and Rh67 proteins contribute to CMV immune evasion by decreasing surface expression of MHC Ia. Unexpectedly, while the Rh67 VL9 peptide is resistant to the effects of Rh67, UL40 can partially counteract the up-regulation of MHC-E expression mediated by its own VL9 peptide. This suggests differences in the mechanisms by which the two VL9 peptides up-regulate MHC-E, and further work will be required to determine if any such differences have implications for translating a RhCMV-vectored simian immunodeficiency virus (SIV) vaccine to HIV-1 using human CMV as a vector. IMPORTANCE: The protective immune response induced by a rhesus cytomegalovirus (RhCMV)-vectored simian immunodeficiency virus (SIV) vaccine in rhesus macaques depends on the presence of the viral Rh67 gene in the vaccine. The Rh67 protein contains a peptide that allows the RhCMV-infected cells to maintain expression of major histocompatibility complex (MHC) antigen E at the cell surface. We show that production of this peptide, referred to as "VL9," mirrors that of the equivalent peptide present in the human cytomegalovirus (CMV) protein UL40, despite the little sequence similarity between the two CMV proteins. We also show that the mature UL40 and Rh67 proteins, which have no previously described function, also contribute to CMV immune evasion by reducing cell surface expression of MHC proteins important for the immune system to detect infected cells. Despite these similarities, our work also reveals possible differences between Rh67 and UL40, and these may have implications for the use of human CMV as the vector for a potential HIV-1 vaccine.


Subject(s)
Cytomegalovirus , Histocompatibility Antigens Class I , Macaca mulatta , Viral Proteins , Cytomegalovirus/immunology , Humans , Animals , Viral Proteins/metabolism , Viral Proteins/immunology , Viral Proteins/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Antigen Presentation , Protein Sorting Signals , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Immune Evasion
12.
Antiviral Res ; 230: 105978, 2024 10.
Article in English | MEDLINE | ID: mdl-39117282

ABSTRACT

Seasonal influenza is an annually severe crisis for global public health, and an ideal influenza vaccine is expected to provide broad protection against constantly drifted strains. Compared to highly flexible hemagglutinin (HA), increasing data have demonstrated that neuraminidase (NA) might be a potential target against influenza variants. In the present study, a series of genetic algorithm-based mosaic NA were designed, and then cloned into recombinant DNA and replication-defective Vesicular Stomatitis Virus (VSV) vector as a novel influenza vaccine candidate. Our Results showed that DNA prime/VSV boost strategy elicited a robust NA-specific Th1-dominated immune response, but the traditional inactivated influenza vaccine elicited a Th2-dominated immune response. More importantly, the superior NA-specific immunity induced by our strategy could confer both a full protection against lethal homologous influenza challenge and a partial protection against heterologous influenza infection. These findings will provide insights on designing NA-based universal vaccine strategy against influenza variants.


Subject(s)
Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Neuraminidase/immunology , Neuraminidase/genetics , Influenza Vaccines/immunology , Influenza Vaccines/genetics , Animals , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Mice , T-Lymphocytes/immunology , Mice, Inbred BALB C , Female , Humans , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Th1 Cells/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood
13.
Viruses ; 16(8)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39205158

ABSTRACT

Mpox (formerly known as monkeypox) is a zoonotic disease caused by monkeypox virus (MPXV), a DNA virus belonging to the Orthopoxvirus genus, in the Poxviridae family. The disease constitutes a moderate risk to public health at the global level. The MPXV A29L protein plays a crucial role in coordinating virion assembly and facilitating important virus-host interactions. This study focused on the expression, purification, and recombinant protein synthesis of the A29L protein of MPXV using prokaryotic systems. Using hybridoma technology, we successfully generated the monoclonal antibodies (mAbs) 1E12 and 4B2, which specifically recognize the A29L protein. These mAbs were found to be suitable for use in indirect immunofluorescence assays (IFA), Western blotting, and immunoprecipitation (IP). Our investigation also revealed that mAbs 1E12 and 4B2 could detect the A27L protein, a homologous protein found in the vaccinia virus Western Reserve (VACV WR) strain, using IFA, Western blotting, and immunoprecipitation (IP). Using mAbs 1E12 and 4B2 as primary immunological probes, A27L protein expression was detected as early as 6 h postinfection with VACV WR, with increasing protein levels being observed throughout the infection. This study enhances our understanding of the protein structure and function of MPXV and contributes to the development of specific MPXV detection methods.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Monkeypox virus , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Animals , Mice , Antibodies, Viral/immunology , Monkeypox virus/immunology , Monkeypox virus/genetics , Mice, Inbred BALB C , Viral Proteins/immunology , Viral Proteins/genetics , Humans , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Female , Vaccinia virus/immunology , Vaccinia virus/genetics , Hybridomas
14.
Viruses ; 16(8)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39205259

ABSTRACT

The E3 ubiquitin ligase TRIM7 is known to have dual roles during viral infections. Like other TRIM proteins, TRIM7 can regulate the IFN pathway via the regulation of the cytosolic receptors RIG-I or MDA-5, which promote the production of type I interferons (IFN-I) and antiviral immune responses. Alternatively, under certain infectious conditions, TRIM7 can negatively regulate IFN-I signaling, resulting in increased virus replication. A growing body of evidence has also shown that TRIM7 can, in some cases, ubiquitinate viral proteins to promote viral replication and pathogenesis, while in other cases it can promote degradation of viral proteins through the proteasome, reducing virus infection. TRIM7 can also regulate the host inflammatory response and modulate the production of inflammatory cytokines, which can lead to detrimental inflammation. TRIM7 can also protect the host during infection by reducing cellular apoptosis. Here, we discuss the multiple functions of TRIM7 during viral infections and its potential as a therapeutic target.


Subject(s)
Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Virus Diseases , Virus Replication , Humans , Virus Diseases/immunology , Virus Diseases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Animals , Ubiquitination , Signal Transduction , Interferon Type I/metabolism , Interferon Type I/immunology , Immunity, Innate , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Host-Pathogen Interactions/immunology
15.
Int J Mol Sci ; 25(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39201592

ABSTRACT

The African swine fever virus (ASFV) is a large enveloped DNA virus that causes a highly pathogenic hemorrhagic disease in both domestic pigs and wild boars. The ASFV genome contains a double-stranded DNA encoding more than 150 proteins. The ASFV possesses only one protease, pS273R, which is important for virion assembly and host immune evasion. Therefore, the specific monoclonal antibody (mAb) against pS273R is useful for ASFV research. Here, we generated two specific anti-pS273R mAbs named 2F3 and 3C2, both of which were successfully applied for ELISA, Western blotting, and immunofluorescence assays. Further, we showed that both 2F3 and 3C2 mAbs recognize a new epitope of N terminal 1-25 amino acids of pS273R protein, which is highly conserved across different ASFV strains including all genotype I and II strains. Based on the recognized epitope, an indirect ELISA was established and was effective in detecting antibodies during ASFV infection. To conclude, the specific pS273R mAbs and corresponding epitope identified will strongly promote ASFV serological diagnosis and vaccine research.


Subject(s)
African Swine Fever Virus , African Swine Fever , Antibodies, Monoclonal , Epitopes , African Swine Fever Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Epitopes/immunology , Swine , Mice , African Swine Fever/immunology , African Swine Fever/virology , Antibodies, Viral/immunology , Mice, Inbred BALB C , Viral Proteins/immunology , Peptide Hydrolases/immunology , Peptide Hydrolases/metabolism , Antigens, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping
16.
Eur J Immunol ; 54(10): e2451135, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39086059

ABSTRACT

Inflammasomes are essential for host defense, recognizing foreign or stress signals to trigger immune responses, including maturation of IL-1 family cytokines and pyroptosis. Here, NLRP1 is emerging as an important sensor of viral infection in barrier tissues. NLRP1 is activated by various stimuli, including viral double-stranded (ds) RNA, ribotoxic stress, and inhibition of dipeptidyl peptidases 8 and 9 (DPP8/9). However, certain viruses, most notably the vaccinia virus, have evolved strategies to subvert inflammasome activation or effector functions. Using the modified vaccinia virus Ankara (MVA) as a model, we investigated how the vaccinia virus inhibits inflammasome activation. We confirmed that the early gene F1L plays a critical role in inhibiting NLRP1 inflammasome activation. Interestingly, it blocks dsRNA and ribotoxic stress-dependent NLRP1 activation without affecting its DPP9-inhibition-mediated activation. Complementation and loss-of-function experiments demonstrated the sufficiency and necessity of F1L in blocking NLRP1 activation. Furthermore, we found that F1L-deficient, but not wild-type MVA, induced ZAKα activation. Indeed, an F1L-deficient virus was found to disrupt protein translation more prominently than an unmodified virus, suggesting that F1L acts in part upstream of ZAKα. These findings underscore the inhibitory role of F1L on NLRP1 inflammasome activation and provide insight into viral evasion of host defenses and the intricate mechanisms of inflammasome activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Inflammasomes , NLR Proteins , Vaccinia virus , Vaccinia virus/immunology , Inflammasomes/immunology , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , NLR Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/immunology , HEK293 Cells , Viral Proteins/immunology , Viral Proteins/metabolism , Viral Proteins/genetics , Vaccinia/immunology , Animals , Mice , Immune Evasion
17.
Nature ; 633(8030): 710-717, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39187718

ABSTRACT

The rapid evolution of viruses generates proteins that are essential for infectivity and replication but with unknown functions, due to extreme sequence divergence1. Here, using a database of 67,715 newly predicted protein structures from 4,463 eukaryotic viral species, we found that 62% of viral proteins are structurally distinct and lack homologues in the AlphaFold database2,3. Among the remaining 38% of viral proteins, many have non-viral structural analogues that revealed surprising similarities between human pathogens and their eukaryotic hosts. Structural comparisons suggested putative functions for up to 25% of unannotated viral proteins, including those with roles in the evasion of innate immunity. In particular, RNA ligase T-like phosphodiesterases were found to resemble phage-encoded proteins that hydrolyse the host immune-activating cyclic dinucleotides 3',3'- and 2',3'-cyclic GMP-AMP (cGAMP). Experimental analysis showed that RNA ligase T homologues encoded by avian poxviruses similarly hydrolyse cGAMP, showing that RNA ligase T-mediated targeting of cGAMP is an evolutionarily conserved mechanism of immune evasion that is present in both bacteriophage and eukaryotic viruses. Together, the viral protein structural database and analyses presented here afford new opportunities to identify mechanisms of virus-host interactions that are common across the virome.


Subject(s)
Protein Folding , Viral Proteins , Virome , Animals , Humans , Bacteriophages/enzymology , Bacteriophages/immunology , Hydrolysis , Immune Evasion/immunology , Immunity, Innate/immunology , Models, Molecular , Nucleotides, Cyclic/chemistry , Nucleotides, Cyclic/immunology , Nucleotides, Cyclic/metabolism , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/chemistry , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Proteins/metabolism , Virome/immunology , Virome/physiology , Databases, Protein , Host Microbial Interactions
18.
Nature ; 634(8033): 424-431, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111359

ABSTRACT

Viruses compete with each other for limited cellular resources, and some deliver defence mechanisms that protect the host from competing genetic parasites1. The phage antirestriction induced system (PARIS) is a defence system, often encoded in viral genomes, that is composed of a 55 kDa ABC ATPase (AriA) and a 35 kDa TOPRIM nuclease (AriB)2. However, the mechanism by which AriA and AriB function in phage defence is unknown. Here we show that AriA and AriB assemble into a 425 kDa supramolecular immune complex. We use cryo-electron microscopy to determine the structure of this complex, thereby explaining how six molecules of AriA assemble into a propeller-shaped scaffold that coordinates three subunits of AriB. ATP-dependent detection of foreign proteins triggers the release of AriB, which assembles into a homodimeric nuclease that blocks infection by cleaving host lysine transfer RNA. Phage T5 subverts PARIS immunity through expression of a lysine transfer RNA variant that is not cleaved by PARIS, thereby restoring viral infection. Collectively, these data explain how AriA functions as an ATP-dependent sensor that detects viral proteins and activates the AriB toxin. PARIS is one of an emerging set of immune systems that form macromolecular complexes for the recognition of foreign proteins, rather than foreign nucleic acids3.


Subject(s)
Adenosine Triphosphate , Cryoelectron Microscopy , Models, Molecular , Adenosine Triphosphate/metabolism , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/immunology , RNA, Transfer/metabolism , RNA, Transfer/chemistry , RNA, Transfer/genetics , Genome, Viral/genetics , Bacteriophages/genetics
19.
Int J Biol Macromol ; 279(Pt 4): 134852, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39159796

ABSTRACT

The African swine fever virus (ASFV), a highly contagious pathogen responsible for African swine fever (ASF), causes significant economic losses in the global pork industry. Due to its large and complex structure, ASFV remains refractory to commercial vaccine development, necessitating the creation of rapid, sensitive, and specific diagnostic tools for disease control. In this study, quantum dots were conjugated to ASFV p72 protein to establish a fluorescent immunochromatographic assay for detecting ASFV-specific antibodies. The assay test strips contained four adjacent pads arranged sequentially: a sample-application pad, a pad containing mobile antigen-probe conjugate, a nitrocellulose readout pad featuring a test line containing immobilised staphylococcal protein A and a control line containing immobilised monoclonal antibodies against the ASFV p72 protein, and an absorbent pad driving the directional flow of liquid via capillary action. The resulting fluorescence immunochromatographic assay demonstrated highly sensitive and specific ASFV antibody detection in under 15 min. Specificity testing showed no cross-reactivity with serum antibodies against other viruses and sensitivity surpassing that of commercial ASFV antibody colloidal gold immunochromatographic test strips. This novel approach offers rapid detection, excellent specificity, and high sensitivity, and supports the future development of fluorescent immunochromatographic test strips for ASFV antibody detection.


Subject(s)
African Swine Fever Virus , Antibodies, Viral , Chromatography, Affinity , African Swine Fever Virus/immunology , Animals , Chromatography, Affinity/methods , Antibodies, Viral/immunology , Swine , African Swine Fever/diagnosis , African Swine Fever/immunology , African Swine Fever/virology , Quantum Dots/chemistry , Fluorescence , Viral Proteins/immunology , Immunoassay/methods
20.
J Gen Virol ; 105(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-39167082

ABSTRACT

Molluscum contagiosum virus (MCV) is a human-specific poxvirus that causes a highly common but mild infection characterized by distinctive and persistent papular skin lesions. These lesions can persist for long periods without an effective clearance response from the host. MCV, like all poxviruses, encodes multiple known immunosuppressive proteins which target innate immune signalling pathways involved in viral nucleic acid sensing, interferon production and inflammation which should trigger antiviral immunity leading to clearance. Two major families of transcription factors responsible for driving the immune response to viruses are the NF-κB and the interferon regulatory factor (IRF) families. While NF-κB broadly drives pro-inflammatory gene expression and IRFs chiefly drive interferon induction, both collaborate in transactivating many of the same genes in a concerted immune response to viral infection. Here, we report that the MCV protein MC089 specifically inhibits IRF activation from both DNA- and RNA-sensing pathways, making it the first characterized MCV inhibitor to selectively target IRF activation to date. MC089 interacts with proteins required for IRF activation, namely IKKε, TBKBP1 and NAP1. Additionally, MC089 targets RNA sensing by associating with the RNA-sensing adaptor protein mitochondrial antiviral-signalling protein on mitochondria. MC089 displays specificity in its inhibition of IRF3 activation by suppressing immunostimulatory nucleic acid-induced serine 396 phosphorylation without affecting the phosphorylation of serine 386. The selective interaction of MC089 with IRF-regulatory proteins and site-specific inhibition of IRF3 phosphorylation may offer a tool to provide novel insights into the biology of IRF3 regulation.


Subject(s)
Interferon Regulatory Factor-3 , Molluscum contagiosum virus , Viral Proteins , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Molluscum contagiosum virus/immunology , Molluscum contagiosum virus/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Signal Transduction , Immunity, Innate , HEK293 Cells , Host-Pathogen Interactions/immunology
SELECTION OF CITATIONS
SEARCH DETAIL