1
|
Zhang S, Liu G, Wang C, Guo A, Chen Y. Enhanced immunogenicity of a BoHV-1 gG-/tk- vaccine. Vaccine 2025; 47:126704. [PMID: 39778477 DOI: 10.1016/j.vaccine.2025.126704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Bovine herpesvirus type 1 (BoHV-1) is a widespread respiratory infection that significantly impacts cattle health worldwide. To address this issue in China, we previously developed a novel double gene-deleted vaccine targeting gG and tk. In this study, we further evaluated the efficacy of this vaccine by challenging vaccinated cattle with a prevalent wild-type BoHV-1 strain and comparing its effectiveness against a commercially available inactivated BoHV-1 vaccine. Post-immunization, all cattle maintained normal rectal temperatures and exhibited no respiratory symptoms. Cattle receiving the gene-deleted vaccine showed a significant increase in the expression of immune markers IFN-γ and TNF-α. Following exposure to wild-type BoHV-1, all immunized groups produced high levels of neutralizing antibodies and specific gB antibodies. Notably, virus shedding was significantly lower in the vaccinated groups compared to the non-immune challenge group. Histological analysis of lung tissues revealed that vaccinated calves had more intact lung structure than their unimmunized counterparts after the challenge. Additionally, the gG-/tk- gene-deleted vaccine demonstrated a higher protective rate based on the average scores of clinical symptoms and lung lesions. Overall, the BoHV-1 gG-/tk- gene-deleted vaccine outperformed the other vaccines tested. This study confirms that the gene-deleted vaccine provides robust protection and superior immunogenicity compared to existing inactivated vaccines, underscoring its potential for future market application.
Collapse
Affiliation(s)
- Sen Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Guoxing Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Chen Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Aizhen Guo
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of development of veterinary diagnostic products, Ministry of Agriculture and Rural Affair, Wuhan 430070, China.
| | - Yingyu Chen
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of development of veterinary diagnostic products, Ministry of Agriculture and Rural Affair, Wuhan 430070, China.
| |
Collapse
|
2
|
El-Mayet F, Jones C. Stress Can Induce Bovine Alpha-Herpesvirus 1 (BoHV-1) Reactivation from Latency. Viruses 2024; 16:1675. [PMID: 39599791 PMCID: PMC11599084 DOI: 10.3390/v16111675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Bovine alpha-herpesvirus 1 (BoHV-1) is a significant problem for the cattle industry, in part because the virus establishes latency, and stressful stimuli increase the incidence of reactivation from latency. Sensory neurons in trigeminal ganglia and unknown cells in pharyngeal tonsils are importantsites for latency. Reactivation from latency can lead to reproductive problems in pregnant cows, virus transmission to young calves, suppression of immune responses, and bacterial pneumonia. BoHV-1 is also a significant cofactor in bovine respiratory disease (BRD). Stress, as mimicked by the synthetic corticosteroid dexamethasone, reproducibly initiates reactivation from latency. Stress-mediated activation of the glucocorticoid receptor (GR) stimulates viral replication and transactivation of viral promoters that drive the expression of infected cell protein 0 (bICP0) and bICP4. Notably, GR and Krüppel-like factor 15 (KLF15) form a feed-forward transcription loop that cooperatively transactivates immediate early transcription unit 1 (IEtu1 promoter). Two pioneer transcription factors, GR and KLF4, cooperatively transactivate the bICP0 early promoter. Pioneer transcription factors bind silent viral heterochromatin, remodel chromatin, and activate gene expression. Thus, wepredict that these novel transcription factors mediate early stages of BoHV-1 reactivation from latency.
Collapse
Affiliation(s)
- Fouad El-Mayet
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA;
- Department of Virology, Faculty of Veterinary Medicine, Benha University, Benha 74078, Egypt
| | - Clinton Jones
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA;
| |
Collapse
|
3
|
Pavulraj S, Stout RW, Paulsen DB, Chowdhury SI. A Quadruple Gene-Deleted Live BoHV-1 Subunit RVFV Vaccine Vector Reactivates from Latency and Replicates in the TG Neurons of Calves but Is Not Transported to and Shed from Nasal Mucosa. Viruses 2024; 16:1497. [PMID: 39339973 PMCID: PMC11437494 DOI: 10.3390/v16091497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Bovine herpesvirus type 1 (BoHV-1) establishes lifelong latency in trigeminal ganglionic (TG) neurons following intranasal and ocular infection in cattle. Periodically, the latent virus reactivates in the TG due to stress and is transported anterogradely to nerve endings in the nasal epithelium, where the virus replicates and sheds. Consequently, BoHV-1 is transmitted to susceptible animals and maintained in the cattle population. Modified live BoHV-1 vaccine strains (BoHV-1 MLV) also have a similar latency reactivation. Therefore, they circulate and are maintained in cattle herds. Additionally, they can regain virulence and cause vaccine outbreaks because they mutate and recombine with other circulating field wild-type (wt) strains. Recently, we constructed a BoHV-1 quadruple mutant virus (BoHV-1qmv) that lacks immune evasive properties due to UL49.5 and glycoprotein G (gG) deletions. In addition, it also lacks the gE cytoplasmic tail (gE CT) and Us9 gene sequences designed to make it safe, increase its vaccine efficacy against BoHV-1, and restrict its anterograde neuronal transport noted above. Further, we engineered the BoHV-1qmv-vector to serve as a subunit vaccine against the Rift Valley fever virus (BoHV-1qmv Sub-RVFV) (doi: 10.3390/v15112183). In this study, we determined the latency reactivation and nasal virus shedding properties of BoHV-1qmv (vector) and BoHV-1qmv-vectored subunit RVFV (BoHV-1qmv sub-RVFV) vaccine virus in calves in comparison to the BoHV-1 wild-type (wt) following intranasal inoculation. The real-time PCR results showed that BoHV-1 wt- but not the BoHV-1qmv vector- and BoHV-1qmv Sub-RVFV-inoculated calves shed virus in the nose following dexamethasone-induced latency reactivation; however, like the BoHV-1 wt, both the BoHV-1qmv vector and BoHV-1qmv Sub-RVFV viruses established latency, were reactivated, and replicated in the TG neurons. These results are consistent with the anterograde neurotransport function of the gE CT and Us9 sequences, which are deleted in the BoHV-1qmv and BoHV-1qmv Sub-RVFV.
Collapse
Affiliation(s)
| | | | | | - Shafiqul I. Chowdhury
- Louisiana Animal Disease Diagnostic Laboratory, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (S.P.); (R.W.S.); (D.B.P.)
| |
Collapse
|
4
|
Krishnagopal A, van Drunen Littel-van den Hurk S. The biology and development of vaccines for bovine alphaherpesvirus 1. Vet J 2024; 306:106152. [PMID: 38821207 DOI: 10.1016/j.tvjl.2024.106152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Bovine alphaherpesvirus type 1 (BoAHV-1) infections lead to compromised herd health and significantly reduced productivity of affected cattle. While BoAHV-1 may cause rhinotracheitis, conjunctivitis, genital infections, and abortions, respiratory tract infections constitute the predominant clinical disease. Immune suppression induced by BoAHV-1 may contribute to co-infections initiating the bovine respiratory disease complex. In this review, the emphasis is to recapitulate the biology and the vaccine technologies currently in use and in development for BoAHV-1, and to discuss the major limitations. Studies on the life cycle and host interactions of BoAHV-1 have resulted in the identification of virulence factors. While several vaccine types, such as vectored vaccines and subunit vaccines, are under investigation, modified live and inactivated BoAHV-1 vaccines are still most frequently used in most areas of the world, whereas attenuated and inactivated marker vaccines are in use in Europe. The knowledge gained from studies on the biology of BoAHV-1 can form a basis for the rational design of future vaccines.
Collapse
Affiliation(s)
- Akshaya Krishnagopal
- Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Sylvia van Drunen Littel-van den Hurk
- Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada.
| |
Collapse
|
5
|
Quintero Barbosa JS, Alméciga-Díaz CJ, Pérez SE, Gutierrez MF. Humoral Immune Response of Mice against a Vaccine Candidate Composed of a Chimera of gB of Bovine Alphaherpesviruses 1 and 5. Vaccines (Basel) 2023; 11:1173. [PMID: 37514988 PMCID: PMC10386439 DOI: 10.3390/vaccines11071173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 07/30/2023] Open
Abstract
Infectious bovine rhinotracheitis (IBR) and bovine meningoencephalitis are caused by Bovine alphaherpesvirus (BoHV) types 1 and 5, which seriously threaten the global cattle industry. Vaccination to improve immunity is the most direct and effective means to prevent these conditions. Glycoprotein B (gB) is essential for the attachment of both viruses to permissive cells, and is a major target of the host immune system, inducing a strong humoral response. The aim of this study was to evaluate, in a murine model, the immune response of a candidate vaccine formulation composed of a chimeric BoHV-1 and BoHV-5 gB (DgB), expressed in Komagataella phaffii. The chimeric DgB vaccine adjuvanted with Montanide 50 ISA V2 or aluminum hydroxide was administered intramuscularly or subcutaneously. A control group and a group that received a commercial vaccine were inoculated subcutaneously. Higher titers of neutralizing antibodies against BoHV-1, BoHV-5, and a natural BoHV-1/5 recombinant strain were obtained with the oil-based candidate vaccine formulation administered intramuscularly. The results demonstrated that the chimeric DgB conserved important epitopes that were able to stimulate a humoral immune response capable of neutralizing BoHV-1, BoHV-5, and the recombinant strain, suggesting that the vaccine antigen is a promising candidate to be further evaluated in cattle.
Collapse
Affiliation(s)
- Juan Sebastian Quintero Barbosa
- Virology Laboratory, Infectious Diseases Group, Microbiology Department, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C. 110231, Colombia
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C. 110231, Colombia
| | - Sandra E Pérez
- Tandil Veterinary Research Center (CIVETAN)-CONICET, Faculty of Veterinary Sciences, National University of the Center of the Province of Buenos Aires, Tandil B7000GHG, Argentina
| | - María Fernanda Gutierrez
- Virology Laboratory, Infectious Diseases Group, Microbiology Department, Faculty of Science, Pontificia Universidad Javeriana, Bogotá D.C. 110231, Colombia
| |
Collapse
|
6
|
Losinno A, Vissani MA, Sanchez D, Damiani AM. Equid herpesvirus type 3 infection produces membrane-associated and secreted forms of glycoprotein G that are not required for efficient cell-to-cell spread of the virus in vitro. Arch Virol 2023; 168:122. [PMID: 36977931 DOI: 10.1007/s00705-023-05727-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/28/2023] [Indexed: 03/30/2023]
Abstract
The ORF 70 gene of equid alphaherpesvirus type 3 (EHV-3) encodes glycoprotein G (gG), which is conserved in the majority of alphaherpesviruses. This glycoprotein is located in the viral envelope and has the characteristic of being secreted into the culture medium after proteolytic processing. It modulates the antiviral immune response of the host by interacting with chemokines. The aim of this study was to identify and characterize EHV-3 gG. By constructing viruses with HA-tagged gG, it was possible to detect gG in lysates of infected cells, their supernatants, and purified virions. A 100-, 60-, and 17-kDa form of the protein were detected in viral particles, while a 60-kDa form was identified in supernatants of infected cells. The role of EHV-3 gG in the viral infection cycle was assessed by the construction of a gG-minus EHV-3 mutant and its gG-positive revertant. When growth characteristics in an equine dermal fibroblast cell line were compared, the plaque size and the growth kinetics of the gG-minus mutant were similar to those of the revertant virus, suggesting that EHV-3 gG does not play a role in direct cell-to-cell transmission or virus proliferation of EHV-3 in tissue culture. The identification and characterization of EHV-3 gG described here provide a solid background for further studies to assess whether this glycoprotein has a function in modulating the host immune response.
Collapse
Affiliation(s)
- Antonella Losinno
- Instituto de Medicina y Biología Experimental de Cuyo IMBECU, CCT Mendoza, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Aldana Vissani
- Instituto de Virología, CICVyA, INTA, Las Cabañas y Los Reseros s/n, 1712, Castelar, Argentina
- Escuela de Veterinaria, Universidad del Salvador, Pilar, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Diego Sanchez
- Instituto de Medicina y Biología Experimental de Cuyo IMBECU, CCT Mendoza, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Armando Mario Damiani
- Instituto de Medicina y Biología Experimental de Cuyo IMBECU, CCT Mendoza, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
7
|
Quintero Barbosa JS, Rojas HYT, Gonzalez J, Espejo-Mojica AJ, Díaz CJA, Gutierrez MF. Characterization and expression of domains of Alphaherpesvirus bovine 1/5 envelope glycoproteins B in Komagataella phaffi. BMC Vet Res 2023; 19:28. [PMID: 36721143 PMCID: PMC9887784 DOI: 10.1186/s12917-023-03590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Bovine herpes virus (BoHV 1 and BoHV-5) are the causative agents of infectious bovine rhinotracheitis (IBR). IBR is responsible for important economic losses in the cattle industry. The envelope glycoprotein B (gB) is essential for BoHV infection of cattle's upper respiratory and genital tract. gB is one of the main candidate antigens for a potential recombinant vaccine since it induces a strong and persistent immune response. RESULTS In this study, gB of BoHV-1 and BoHV-5 was characterized in terms of function, structure, and antigenicity through bioinformatics tools. gB showed conserved sequence and structure, so, both domains named PH Like 1 and 2 domains of each virus were selected for the design of a bivalent vaccine candidate. The immunoinformatic study showed that these two domains have epitopes recognizable by B and T lymphocytes, followed by this, the cDNA domains from BoHV-1/5 gB (Domains-gB) were transformed into the yeast Komagataella phaffii GS115 (previously known as Pichia pastoris). A recombinant protein with molecular weight of about 110 kDa was obtained from the culture media. The vaccine candidate protein (Domains-gB) was recognized by a monoclonal antibody from a commercial ELISA kit used for IBR diagnostic, which may suggest that the epitopes are conserved of the entire infectious virus. CONCLUSION Overall, it was shown that the recombinant domains of BoHV-1/5 gB have antigenic and immunogenic properties similar to the native gB. This vaccine candidate is promising to be used in future studies to assess its immunogenicity in an animal model.
Collapse
Affiliation(s)
- Juan Sebastián Quintero Barbosa
- grid.41312.350000 0001 1033 6040Virology Laboratory, Infectious Diseases Group, Microbiology Department, Faculty of Science Pontificia, Universidad Javeriana, Bogotá, D.C Colombia
| | - Heidy Yohana Triana Rojas
- grid.41312.350000 0001 1033 6040Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C Colombia
| | - Janneth Gonzalez
- grid.41312.350000 0001 1033 6040Nutrition and Biochemistry Department, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C Colombia
| | - Angela Johana Espejo-Mojica
- grid.41312.350000 0001 1033 6040Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C Colombia
| | - Carlos Javier Alméciga Díaz
- grid.41312.350000 0001 1033 6040Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C Colombia
| | - María Fernanda Gutierrez
- grid.41312.350000 0001 1033 6040Virology Laboratory, Infectious Diseases Group, Microbiology Department, Faculty of Science Pontificia, Universidad Javeriana, Bogotá, D.C Colombia
| |
Collapse
|
8
|
Liu CY, Guo H, Zhao HZ, Hou LN, Wen YJ, Wang FX. Recombinant Bovine Herpesvirus Type I Expressing the Bovine Viral Diarrhea Virus E2 Protein Could Effectively Prevent Infection by Two Viruses. Viruses 2022; 14:v14081618. [PMID: 35893683 PMCID: PMC9331970 DOI: 10.3390/v14081618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 12/03/2022] Open
Abstract
Bovine respiratory disease complex (BRDC) is a comprehensive disease in cattle caused by various viral and bacterial infections. Among them, bovine herpesvirus type I (BoHV−1) and bovine viral diarrhea virus (BVDV) play important roles and have caused huge financial losses for the cattle industry worldwide. At present, vaccines against BRDC include trivalent attenuated BoHV−1, BVDV−1, and BVDV−2 live vaccines, BoHV−1 live attenuated vaccines, and BoHV−1/BVDV bivalent live attenuated vaccines, which have limitations in terms of their safety and efficacy. To solve these problems, we optimized the codon of the BVDV−1 E2 gene, added the signal peptide sequence of the BoHV−1 gD gene, expressed double BVDV−1 E2 glycoproteins in tandem at the BoHV−1 gE gene site, and constructed a BoHV−1 genetics-engineered vectored vaccine with gE gene deletion, named BoHV−1 gE/E2−Linker−E2+ and BoHV−1 ΔgE. This study compared the protective effects in BoHV−1, BoHV−1 ΔgE, BoHV−1 gE/E2−Linker−E2+, and BVDV−1 inactivated antigen immunized guinea pigs and calves. The results showed that BoHV−1 gE/E2−Linker−E2+ could successfully induce guinea pigs and calves to produce specific neutralizing antibodies against BVDV−1. In addition, after BoHV−1 and BVDV−1 challenges, BoHV−1 gE/E2−Linker−E2+ can produce a specific neutralizing antibody response against BoHV−1 and BVDV−1 infections. Calves immunized with this type of virus can be distinguished as either vaccinated animals (gE-) or naturally infected animals (gE+). In summary, our data suggest that BoHV−1 gE/E2−Linker−E2+ and BoHV−1 ΔgE have great potential to prevent BVDV−1 or BoHV−1 infection.
Collapse
|
9
|
Zarski LM, Vaala WE, Barnett DC, Bain FT, Soboll Hussey G. A Live-Attenuated Equine Influenza Vaccine Stimulates Innate Immunity in Equine Respiratory Epithelial Cell Cultures That Could Provide Protection From Equine Herpesvirus 1. Front Vet Sci 2021; 8:674850. [PMID: 34179166 PMCID: PMC8224402 DOI: 10.3389/fvets.2021.674850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/23/2021] [Indexed: 01/04/2023] Open
Abstract
Equine herpesvirus 1 (EHV-1) ubiquitously infects horses worldwide and causes respiratory disease, abortion, and equine herpesvirus myeloencephalopathy. Protection against EHV-1 disease is elusive due to establishment of latency and immune-modulatory features of the virus. These include the modulation of interferons, cytokines, chemokines, antigen presentation, and cellular immunity. Because the modulation of immunity likely occurs at the site of first infection—the respiratory epithelium, we hypothesized that the mucosal influenza vaccine Flu Avert® I.N. (Flu Avert), which is known to stimulate strong antiviral responses, will enhance antiviral innate immunity, and that these responses would also provide protection from EHV-1 infection. To test our hypothesis, primary equine respiratory epithelial cells (ERECs) were treated with Flu Avert, and innate immunity was evaluated for 10 days following treatment. The timing of Flu Avert treatment was also evaluated for optimal effectiveness to reduce EHV-1 replication by modulating early immune responses to EHV-1. The induction of interferons, cytokine and chemokine mRNA expression, and protein secretion was evaluated by high-throughput qPCR and multiplex protein analysis. Intracellular and extracellular EHV-1 titers were determined by qPCR. Flu Avert treatment resulted in the modulation of IL-8, CCL2, and CXCL9 starting at days 5 and 6 post-treatment. Coinciding with the timing of optimal chemokine induction, our data also suggested the same timing for reduction of EHV-1 replication. In combination, our results suggest that Flu Avert may be effective at counteracting some of the immune-modulatory properties of EHV-1 at the airway epithelium and the peak for this response occurs 5–8 days post-Flu Avert treatment. Future in vivo studies are needed to investigate Flu Avert as a prophylactic in situations where EHV-1 exposure may occur.
Collapse
Affiliation(s)
- Lila M Zarski
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Veterinary Medical Center, East Lansing, MI, United States
| | | | | | | | - Gisela Soboll Hussey
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Veterinary Medical Center, East Lansing, MI, United States
| |
Collapse
|
10
|
The Bacterial and Viral Agents of BRDC: Immune Evasion and Vaccine Developments. Vaccines (Basel) 2021; 9:vaccines9040337. [PMID: 33916119 PMCID: PMC8066859 DOI: 10.3390/vaccines9040337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/19/2022] Open
Abstract
Bovine respiratory disease complex (BRDC) is a multifactorial disease of cattle which presents as bacterial and viral pneumonia. The causative agents of BRDC work in synergy to suppress the host immune response and increase the colonisation of the lower respiratory tracts by pathogenic bacteria. Environmental stress and/or viral infection predispose cattle to secondary bacterial infections via suppression of key innate and adaptive immune mechanisms. This allows bacteria to descend the respiratory tract unchallenged. BRDC is the costliest disease among feedlot cattle, and whilst vaccines exist for individual pathogens, there is still a lack of evidence for the efficacy of these vaccines and uncertainty surrounding the optimum timing of delivery. This review outlines the immunosuppressive actions of the individual pathogens involved in BRDC and highlights the key issues in the development of vaccinations against them.
Collapse
|
11
|
Gowthaman V, Kumar S, Koul M, Dave U, Murthy TRGK, Munuswamy P, Tiwari R, Karthik K, Dhama K, Michalak I, Joshi SK. Infectious laryngotracheitis: Etiology, epidemiology, pathobiology, and advances in diagnosis and control - a comprehensive review. Vet Q 2021; 40:140-161. [PMID: 32315579 PMCID: PMC7241549 DOI: 10.1080/01652176.2020.1759845] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Infectious laryngotracheitis (ILT) is a highly contagious upper respiratory tract disease of chicken caused by a Gallid herpesvirus 1 (GaHV-1) belonging to the genus Iltovirus, and subfamily Alphaherpesvirinae within Herpesviridae family. The disease is characterized by conjunctivitis, sinusitis, oculo-nasal discharge, respiratory distress, bloody mucus, swollen orbital sinuses, high morbidity, considerable mortality and decreased egg production. It is well established in highly dense poultry producing areas of the world due to characteristic latency and carrier status of the virus. Co-infections with other respiratory pathogens and environmental factors adversely affect the respiratory system and prolong the course of the disease. Latently infected chickens are the primary source of ILT virus (ILTV) outbreaks irrespective of vaccination. Apart from conventional diagnostic methods including isolation and identification of ILTV, serological detection, advanced biotechnological tools such as PCR, quantitative real-time PCR, next generation sequencing, and others are being used in accurate diagnosis and epidemiological studies of ILTV. Vaccination is followed with the use of conventional vaccines including modified live attenuated ILTV vaccines, and advanced recombinant vector vaccines expressing different ILTV glycoproteins, but still these candidates frequently fail to reduce challenge virus shedding. Some herbal components have proved to be beneficial in reducing the severity of the clinical disease. The present review discusses ILT with respect to its current status, virus characteristics, epidemiology, transmission, pathobiology, and advances in diagnosis, vaccination and control strategies to counter this important disease of poultry.
Collapse
Affiliation(s)
- Vasudevan Gowthaman
- Poultry Disease Diagnosis and Surveillance Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Namakkal, Tamil Nadu, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Monika Koul
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Urmil Dave
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - T R Gopala Krishna Murthy
- Poultry Disease Diagnosis and Surveillance Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Namakkal, Tamil Nadu, India
| | - Palanivelu Munuswamy
- Division of Pathology, ICAR - Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU), Mathura, Uttar Pradesh, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, Tamil Nadu, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR - Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Izabela Michalak
- Faculty of Chemistry, Department of Advanced Material Technologies, Wrocław University of Science and Technology, Wrocław, Poland
| | - Sunil K Joshi
- Department of Microbiology & Immunology, Department of Pediatrics, Division of Hematology, Oncology and Bone Marrow Transplantation, University of Miami School of Medicine, Miami, Florida, USA
| |
Collapse
|
12
|
Laval K, Poelaert KCK, Van Cleemput J, Zhao J, Vandekerckhove AP, Gryspeerdt AC, Garré B, van der Meulen K, Baghi HB, Dubale HN, Zarak I, Van Crombrugge E, Nauwynck HJ. The Pathogenesis and Immune Evasive Mechanisms of Equine Herpesvirus Type 1. Front Microbiol 2021; 12:662686. [PMID: 33746936 PMCID: PMC7970122 DOI: 10.3389/fmicb.2021.662686] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Equine herpesvirus type 1 (EHV-1) is an alphaherpesvirus related to pseudorabies virus (PRV) and varicella-zoster virus (VZV). This virus is one of the major pathogens affecting horses worldwide. EHV-1 is responsible for respiratory disorders, abortion, neonatal foal death and equine herpes myeloencephalopathy (EHM). Over the last decade, EHV-1 has received growing attention due to the frequent outbreaks of abortions and/or EHM causing serious economical losses to the horse industry worldwide. To date, there are no effective antiviral drugs and current vaccines do not provide full protection against EHV-1-associated diseases. Therefore, there is an urgent need to gain a better understanding of the pathogenesis of EHV-1 in order to develop effective therapies. The main objective of this review is to provide state-of-the-art information on the pathogenesis of EHV-1. We also highlight recent findings on EHV-1 immune evasive strategies at the level of the upper respiratory tract, blood circulation and endothelium of target organs allowing the virus to disseminate undetected in the host. Finally, we discuss novel approaches for drug development based on our current knowledge of the pathogenesis of EHV-1.
Collapse
Affiliation(s)
- Kathlyn Laval
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Katrien C K Poelaert
- Division of Virology, Department Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jolien Van Cleemput
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Jing Zhao
- Shenzhen International Institute for Biomedical Research, Shenzhen, China
| | | | | | | | | | - Hossein B Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haileleul N Dubale
- College of Veterinary Medicine and Agriculture, Addis Ababa University, Bishoftu, Ethiopia
| | - Ines Zarak
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eline Van Crombrugge
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hans J Nauwynck
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
13
|
BoHV-1-Vectored BVDV-2 Subunit Vaccine Induces BVDV Cross-Reactive Cellular Immune Responses and Protects against BVDV-2 Challenge. Vaccines (Basel) 2021; 9:vaccines9010046. [PMID: 33451136 PMCID: PMC7828602 DOI: 10.3390/vaccines9010046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/04/2021] [Accepted: 01/08/2021] [Indexed: 02/07/2023] Open
Abstract
The bovine respiratory disease complex (BRDC) remains a major problem for both beef and dairy cattle industries worldwide. BRDC frequently involves an initial viral respiratory infection resulting in immunosuppression, which creates a favorable condition for fatal secondary bacterial infection. Current polyvalent modified live vaccines against bovine herpesvirus type 1(BoHV-1) and bovine viral diarrhea virus (BVDV) have limitations concerning their safety and efficacy. To address these shortcomings and safety issues, we have constructed a quadruple gene mutated BoHV-1 vaccine vector (BoHV-1 QMV), which expresses BVDV type 2, chimeric E2 and Flag-tagged Erns-fused with bovine granulocyte monocyte colony-stimulating factor (GM-CSF) designated here as QMV-BVD2*. Here we compared the safety, immunogenicity, and protective efficacy of QMV-BVD2* vaccination in calves against BVDV-2 with Zoetis Bovi-shield Gold 3 trivalent (BoHV-1, BVDV types 1 and 2) vaccine. The QMV-BVD2* prototype subunit vaccine induced the BoHV-1 and BVDV-2 neutralizing antibody responses along with BVDV-1 and -2 cross-reactive cellular immune responses. Moreover, after a virulent BVDV-2 challenge, the QMV-BVD2* prototype subunit vaccine conferred a more rapid recall BVDV-2-specific neutralizing antibody response and considerably better recall BVDV types 1 and 2-cross protective cellular immune responses than that of the Zoetis Bovi-shield Gold 3.
Collapse
|
14
|
Poelaert KCK, Van Cleemput J, Laval K, Xie J, Favoreel HW, Nauwynck HJ. Equine herpesvirus 1 infection orchestrates the expression of chemokines in equine respiratory epithelial cells. J Gen Virol 2020; 100:1567-1579. [PMID: 31490114 DOI: 10.1099/jgv.0.001317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ancestral equine herpesvirus 1 (EHV1), closely related to human herpes viruses, exploits leukocytes to reach its target organs, accordingly evading the immune surveillance system. Circulating EHV1 strains can be divided into abortigenic/neurovirulent, causing reproductive/neurological disorders. Neurovirulent EHV1 more efficiently recruits monocytic CD172a+ cells to the upper respiratory tract (URT), while abortigenic EHV1 tempers monocyte migration. Whether similar results could be expected for T lymphocytes is not known. Therefore, we questioned whether differences in T cell recruitment could be associated with variations in cell tropism between both EHV1 phenotypes, and which viral proteins might be involved. The expression of CXCL9 and CXCL10 was evaluated in abortigenic/neurovirulent EHV1-inoculated primary respiratory epithelial cells (ERECs). The bioactivity of chemokines was tested with a functional migration assay. Replication of neurovirulent EHV1 in the URT resulted in an enhanced expression/bioactivity of CXCL9 and CXCL10, compared to abortigenic EHV1. Interestingly, deletion of glycoprotein 2 resulted in an increased recruitment of both monocytic CD172a+ cells and T lymphocytes to the corresponding EREC supernatants. Our data reveal a novel function of EHV1-gp2, tempering leukocyte migration to the URT, further indicating a sophisticated virus-mediated orchestration of leukocyte recruitment to the URT.
Collapse
Affiliation(s)
- Katrien C K Poelaert
- Department of Virology, Immunology and Parasitology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Jolien Van Cleemput
- Department of Molecular Biology 301 Schultz Laboratory, Princeton University Washington Rd, Princeton, NJ 08544, USA.,Department of Virology, Immunology and Parasitology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Kathlyn Laval
- Department of Molecular Biology 301 Schultz Laboratory, Princeton University Washington Rd, Princeton, NJ 08544, USA
| | - Jiexiong Xie
- Department of Virology, Immunology and Parasitology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Herman W Favoreel
- Department of Virology, Immunology and Parasitology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Hans J Nauwynck
- Department of Virology, Immunology and Parasitology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
15
|
Semisynthesis of an evasin from tick saliva reveals a critical role of tyrosine sulfation for chemokine binding and inhibition. Proc Natl Acad Sci U S A 2020; 117:12657-12664. [PMID: 32461364 DOI: 10.1073/pnas.2000605117] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Blood-feeding arthropods produce antiinflammatory salivary proteins called evasins that function through inhibition of chemokine-receptor signaling in the host. Herein, we show that the evasin ACA-01 from the Amblyomma cajennense tick can be posttranslationally sulfated at two tyrosine residues, albeit as a mixture of sulfated variants. Homogenously sulfated variants of the proteins were efficiently assembled via a semisynthetic native chemical ligation strategy. Sulfation significantly improved the binding affinity of ACA-01 for a range of proinflammatory chemokines and enhanced the ability of ACA-01 to inhibit chemokine signaling through cognate receptors. Comparisons of evasin sequences and structural data suggest that tyrosine sulfation serves as a receptor mimetic strategy for recognizing and suppressing the proinflammatory activity of a wide variety of mammalian chemokines. As such, the incorporation of this posttranslational modification (PTM) or mimics thereof into evasins may provide a strategy to optimize tick salivary proteins for antiinflammatory applications.
Collapse
|
16
|
Crijns H, Vanheule V, Proost P. Targeting Chemokine-Glycosaminoglycan Interactions to Inhibit Inflammation. Front Immunol 2020; 11:483. [PMID: 32296423 PMCID: PMC7138053 DOI: 10.3389/fimmu.2020.00483] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Leukocyte migration into tissues depends on the activity of chemokines that form concentration gradients to guide leukocytes to a specific site. Interaction of chemokines with their specific G protein-coupled receptors (GPCRs) on leukocytes induces leukocyte adhesion to the endothelial cells, followed by extravasation of the leukocytes and subsequent directed migration along the chemotactic gradient. Interaction of chemokines with glycosaminoglycans (GAGs) is crucial for extravasation in vivo. Chemokines need to interact with GAGs on endothelial cells and in the extracellular matrix in tissues in order to be presented on the endothelium of blood vessels and to create a concentration gradient. Local chemokine retention establishes a chemokine gradient and prevents diffusion and degradation. During the last two decades, research aiming at reducing chemokine activity mainly focused on the identification of inhibitors of the interaction between chemokines and their cognate GPCRs. This approach only resulted in limited success. However, an alternative strategy, targeting chemokine-GAG interactions, may be a promising approach to inhibit chemokine activity and inflammation. On this line, proteins derived from viruses and parasites that bind chemokines or GAGs may have the potential to interfere with chemokine-GAG interactions. Alternatively, chemokine mimetics, including truncated chemokines and mutant chemokines, can compete with chemokines for binding to GAGs. Such truncated or mutated chemokines are characterized by a strong binding affinity for GAGs and abrogated binding to their chemokine receptors. Finally, Spiegelmers that mask the GAG-binding site on chemokines, thereby preventing chemokine-GAG interactions, were developed. In this review, the importance of GAGs for chemokine activity in vivo and strategies that could be employed to target chemokine-GAG interactions will be discussed in the context of inflammation.
Collapse
Affiliation(s)
- Helena Crijns
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vincent Vanheule
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
17
|
Brito AF, Pinney JW. The evolution of protein domain repertoires: Shedding light on the origins of the Herpesviridae family. Virus Evol 2020; 6:veaa001. [PMID: 32042448 PMCID: PMC7000910 DOI: 10.1093/ve/veaa001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Herpesviruses (HVs, Family: Herpesviridae) have large genomes that encode hundreds of proteins. Apart from amino acid mutations, protein domain acquisitions, duplications and losses are also common modes of evolution. HV domain repertoires differ across species, and only a core set is shared among all species, aspect that raises a question: How have HV domain repertoires diverged while keeping some similarities? To answer such question, we used profile Hidden Markov Models (HMMs) to search for domains in all possible translated open reading frames (ORFs) of fully sequenced HV genomes. With at least 274 domains being identified, we built a matrix of domain counts per species, and applied a parsimony method to reconstruct the ancestral states of these domains along the HV phylogeny. It revealed events of domain gain, duplication, and loss over more than 400 millions of years, where Alpha-, Beta-, and GammaHVs expanded and condensed their domain repertoires at distinct rates. Most of the acquired domains perform ‘Modulation and Control’, ‘Envelope’, or ‘Auxiliary’ functions, categories that showed high flexibility (number of domains) and redundancy (number of copies). Conversely, few gains and duplications were observed for domains involved in ‘Capsid assembly and structure’, and ‘DNA Replication, recombination and metabolism’. Among the forty-one primordial domains encoded by Herpesviridae ancestors, twenty-eight are still found in all present-day HVs. Because of their distinct evolutionary strategies, HV domain repertoires are very specific at the subfamily, genus and species levels. Differences in domain composition may not only explain HV host range and tissue tropism, but also provide hints to the origins of HVs.
Collapse
Affiliation(s)
- Anderson F Brito
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - John W Pinney
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
18
|
Atasoy MO, Rohaim MA, Munir M. Simultaneous Deletion of Virulence Factors and Insertion of Antigens into the Infectious Laryngotracheitis Virus Using NHEJ-CRISPR/Cas9 and Cre-Lox System for Construction of a Stable Vaccine Vector. Vaccines (Basel) 2019; 7:vaccines7040207. [PMID: 31817447 PMCID: PMC6963826 DOI: 10.3390/vaccines7040207] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Infectious laryngotracheitis virus (ILTV) is a promising vaccine vector due to its heterologous gene accommodation capabilities, low pathogenicity, and potential to induce cellular and humoral arms of immunity. Owing to these characteristics, different gene-deletion versions of ILTVs have been successfully deployed as a vector platform for the development of recombinant vaccines against multiple avian viruses using conventional recombination methods, which are tedious, time-demanding, and error-prone. Here, we applied a versatile, and customisable clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 accompanied with Cre–Lox system to simultaneously delete virulence factors and to insert foreign genes in the ILTV genome. Using this pipeline, we successfully deleted thymidine kinase (TK) and unique short 4 (US4) genes and inserted fusion (F) gene of the Newcastle disease virus without adversely affecting ILTV replication and expression of the F protein. Taken together, the proposed approach offers novel tools to attenuate (by deletion of virulence factor) and to generate multivalent (by insertion of heterologous genes) vaccine vectors to protect chickens against pathogens of poultry and public health importance.
Collapse
|
19
|
Jones C. Bovine Herpesvirus 1 Counteracts Immune Responses and Immune-Surveillance to Enhance Pathogenesis and Virus Transmission. Front Immunol 2019; 10:1008. [PMID: 31134079 PMCID: PMC6514135 DOI: 10.3389/fimmu.2019.01008] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/18/2019] [Indexed: 12/21/2022] Open
Abstract
Infection of cattle by bovine herpesvirus 1 (BoHV-1) can culminate in upper respiratory tract disorders, conjunctivitis, or genital disorders. Infection also consistently leads to transient immune-suppression. BoHV-1 is the number one infectious agent in cattle that is associated with abortions in cattle. BoHV-1, as other α-herpesvirinae subfamily members, establishes latency in sensory neurons. Stressful stimuli, mimicked by the synthetic corticosteroid dexamethasone, consistently induce reactivation from latency in latently infected calves and rabbits. Increased corticosteroid levels due to stress have a two-pronged effect on reactivation from latency by: (1) directly stimulating viral gene expression and replication, and (2) impairing antiviral immune responses, thus enhancing virus spread and transmission. BoHV-1 encodes several proteins, bICP0, bICP27, gG, UL49.5, and VP8, which interfere with key antiviral innate immune responses in the absence of other viral genes. Furthermore, the ability of BoHV-1 to infect lymphocytes and induce apoptosis, in particular CD4+ T cells, has negative impacts on immune responses during acute infection. BoHV-1 induced immune-suppression can initiate the poly-microbial disorder known as bovine respiratory disease complex, which costs the US cattle industry more than one billion dollars annually. Furthermore, interfering with antiviral responses may promote viral spread to ovaries and the developing fetus, thus enhancing reproductive issues associated with BoHV-1 infection of cows or pregnant cows. The focus of this review is to describe the known mechanisms, direct and indirect, by which BoHV-1 interferes with antiviral immune responses during the course of infection.
Collapse
Affiliation(s)
- Clinton Jones
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
20
|
Heidarieh H, Alcamí A. Mechanism of action of the viral chemokine-binding protein E163 from ectromelia virus. J Biol Chem 2018; 293:17418-17429. [PMID: 30257868 DOI: 10.1074/jbc.ra118.004432] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/13/2018] [Indexed: 12/16/2022] Open
Abstract
Chemokines interact with glycosaminoglycans (GAGs) at the cellular surface and to specific cell-surface receptors to activate signaling pathways. The GAG interaction allows the formation of a chemotactic gradient of chemokine required for cell haptotaxis and chemokine oligomerization. Poxviruses encode secreted chemokine-binding proteins with no sequence similarity to their cellular counterparts to modulate the host immune system. The E163 protein from ectromelia virus, the causative agent of mousepox, binds chemokines through their GAG-binding domain. In addition, E163 interacts with GAGs to be anchored at the cell surface, but its ability to interfere with chemokine-GAG interactions has not been demonstrated. We report the identification of the GAG-binding regions in E163 and the generation of mutant forms deficient of GAG binding. Chemokine binding assays show that some of the E163 GAG-binding sites are also involved in the interaction with chemokines. By using recombinant GAG-binding mutant forms we demonstrate that E163 prevents the interaction of chemokines with cell-surface GAGs, providing mechanisms for the immunomodulatory activity of the viral chemokine-binding protein E163.
Collapse
Affiliation(s)
- Haleh Heidarieh
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Antonio Alcamí
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
21
|
Infectious Laryngotracheitis Virus Viral Chemokine-Binding Protein Glycoprotein G Alters Transcription of Key Inflammatory Mediators In Vitro and In Vivo. J Virol 2017; 92:JVI.01534-17. [PMID: 29070686 DOI: 10.1128/jvi.01534-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/11/2017] [Indexed: 01/05/2023] Open
Abstract
Infectious laryngotracheitis virus (ILTV) is an alphaherpesvirus that infects chickens, causing upper respiratory tract disease and significant losses to poultry industries worldwide. Glycoprotein G (gG) is a broad-range viral chemokine-binding protein conserved among most alphaherpesviruses, including ILTV. A number of studies comparing the immunological parameters between infection with gG-expressing and gG-deficient ILTV strains have demonstrated that expression of gG is associated with increased virulence, modification of the amount and the composition of the inflammatory response, and modulation of the immune responses toward antibody production and away from cell-mediated immune responses. The aims of the current study were to examine the establishment of infection and inflammation by ILTV and determine how gG influences that response to infection. In vitro infection studies using tracheal organ tissue specimen cultures and blood-derived monocytes and in vivo infection studies in specific-pathogen-free chickens showed that leukocyte recruitment to the site of infection is an important component of the induced pathology and that this is influenced by the expression of ILTV gG and changes in the transcription of the chicken orthologues of mammalian CXC chemokine ligand 8 (CXCL8), chicken CXCLi1 and chicken CXCLi2, among other cytokines and chemokines. The results from this study demonstrate that ILTV gG interferes with chemokine and cytokine transcription at different steps of the inflammatory cascade, thus altering inflammation, virulence, and the balance of the immune response to infection.IMPORTANCE Infectious laryngotracheitis virus is an alphaherpesvirus that expresses gG, a conserved broad-range viral chemokine-binding protein known to interfere with host immune responses. However, little is known about how gG modifies virulence and influences the inflammatory signaling cascade associated with infection. Here, data from in vitro and in vivo infection studies are presented. These data show that gG has a direct impact on the transcription of cytokines and chemokine ligands in vitro (such as chicken CXCL8 orthologues, among others), which explains the altered balance of the inflammatory response that is associated with gG during ILTV infection of the upper respiratory tract of chickens. This is the first report to associate gG with the dysregulation of cytokine transcription at different stages of the inflammatory cascade triggered by ILTV infection of the natural host.
Collapse
|
22
|
Graybill C, Claypool DJ, Brinton JT, Levin MJ, Lee KS. Cytokines Produced in Response to Varicella-Zoster Virus Infection of ARPE-19 Cells Stimulate Lymphocyte Chemotaxis. J Infect Dis 2017; 216:1038-1047. [PMID: 28968855 DOI: 10.1093/infdis/jix426] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/15/2017] [Indexed: 12/14/2022] Open
Abstract
Posterior uveitis is an ocular complication that can occur with reactivation of varicella-zoster virus (VZV). It may lead to loss of vision due to retinal detachment and chronic inflammation, which often causes more severe disease than the virus infection itself. To increase our understanding of the immune response, we infected the retinal pigment epithelial (RPE) cell line, ARPE-19, with cell-associated VZV and compared its response to that of the MeWo cell line using multiplex assays. We observed (1) a difference in the magnitude and kinetics of cytokine responses between the 2 cell types and (2) differential migration of CD4+ and CD8+ T cells towards these cytokines. Thus, our data provide information about the cytokine and lymphocytic responses to VZV infection of RPE cells, thereby providing a useful platform for future studies to address mechanisms underlying the immunopathology of VZV-associated posterior uveitis.
Collapse
Affiliation(s)
| | | | - John T Brinton
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora
| | - Myron J Levin
- Department of Pediatrics, Section of Infectious Diseases
| | | |
Collapse
|
23
|
Brito AF, Pinney JW. Protein-Protein Interactions in Virus-Host Systems. Front Microbiol 2017; 8:1557. [PMID: 28861068 PMCID: PMC5562681 DOI: 10.3389/fmicb.2017.01557] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/02/2017] [Indexed: 01/10/2023] Open
Abstract
To study virus–host protein interactions, knowledge about viral and host protein architectures and repertoires, their particular evolutionary mechanisms, and information on relevant sources of biological data is essential. The purpose of this review article is to provide a thorough overview about these aspects. Protein domains are basic units defining protein interactions, and the uniqueness of viral domain repertoires, their mode of evolution, and their roles during viral infection make viruses interesting models of study. Mutations at protein interfaces can reduce or increase their binding affinities by changing protein electrostatics and structural properties. During the course of a viral infection, both pathogen and cellular proteins are constantly competing for binding partners. Endogenous interfaces mediating intraspecific interactions—viral–viral or host–host interactions—are constantly targeted and inhibited by exogenous interfaces mediating viral–host interactions. From a biomedical perspective, blocking such interactions is the main mechanism underlying antiviral therapies. Some proteins are able to bind multiple partners, and their modes of interaction define how fast these “hub proteins” evolve. “Party hubs” have multiple interfaces; they establish simultaneous/stable (domain–domain) interactions, and tend to evolve slowly. On the other hand, “date hubs” have few interfaces; they establish transient/weak (domain–motif) interactions by means of short linear peptides (15 or fewer residues), and can evolve faster. Viral infections are mediated by several protein–protein interactions (PPIs), which can be represented as networks (protein interaction networks, PINs), with proteins being depicted as nodes, and their interactions as edges. It has been suggested that viral proteins tend to establish interactions with more central and highly connected host proteins. In an evolutionary arms race, viral and host proteins are constantly changing their interface residues, either to evade or to optimize their binding capabilities. Apart from gaining and losing interactions via rewiring mechanisms, virus–host PINs also evolve via gene duplication (paralogy); conservation (orthology); horizontal gene transfer (HGT) (xenology); and molecular mimicry (convergence). The last sections of this review focus on PPI experimental approaches and their limitations, and provide an overview of sources of biomolecular data for studying virus–host protein interactions.
Collapse
Affiliation(s)
- Anderson F Brito
- Department of Life Sciences, Centre for Integrative Systems Biology and Bioinformatics, Imperial College LondonLondon, United Kingdom
| | - John W Pinney
- Department of Life Sciences, Centre for Integrative Systems Biology and Bioinformatics, Imperial College LondonLondon, United Kingdom
| |
Collapse
|
24
|
Hart J, MacHugh ND, Sheldrake T, Nielsen M, Morrison WI. Identification of immediate early gene products of bovine herpes virus 1 (BHV-1) as dominant antigens recognized by CD8 T cells in immune cattle. J Gen Virol 2017; 98:1843-1854. [PMID: 28671533 DOI: 10.1099/jgv.0.000823] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In common with other herpes viruses, bovine herpes virus 1 (BHV-1) induces strong virus-specific CD8 T-cell responses. However, there is a paucity of information on the antigenic specificity of the responding T-cells. The development of a system to generate virus-specific CD8 T-cell lines from BHV-1-immune cattle, employing Theileria-transformed cell lines for antigen presentation, has enabled us to address this issue. Use of this system allowed the study to screen for CD8 T-cell antigens that are efficiently presented on the surface of virus-infected cells. Screening of a panel of 16 candidate viral gene products with CD8 T-cell lines from 3 BHV-1-immune cattle of defined MHC genotypes identified 4 antigens, including 3 immediate early (IE) gene products (ICP4, ICP22 and Circ) and a tegument protein (UL49). Identification of the MHC restriction specificities revealed that the antigens were presented by two or three class I MHC alleles in each animal. Six CD8 T-cell epitopes were identified in the three IE proteins by screening of synthetic peptides. Use of an algorithm (NetMHCpan) that predicts the peptide-binding characteristics of restricting MHC alleles confirmed and, in some cases refined, the identity of the epitopes. Analyses of the epitope specificity of the CD8 T-cell lines showed that a large component of the response is directed against these IE epitopes. The results indicate that these IE gene products are dominant targets of the CD8 T-cell response in BHV-I-immune cattle and hence are prime-candidate antigens for the generation of a subunit vaccine.
Collapse
Affiliation(s)
- Jane Hart
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Niall D MacHugh
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Tara Sheldrake
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Morten Nielsen
- Department of Bio and Health Informatics, Centre for Biological Science Sequence Analysis, The Technical University, Lyngby, Denmark
| | - W Ivan Morrison
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK.,Biotechnological Research Institute, National University of San Martin, San Martin, Buenos Aires, Argentina
| |
Collapse
|
25
|
Varicella zoster virus glycoprotein C increases chemokine-mediated leukocyte migration. PLoS Pathog 2017; 13:e1006346. [PMID: 28542541 PMCID: PMC5444840 DOI: 10.1371/journal.ppat.1006346] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/11/2017] [Indexed: 02/07/2023] Open
Abstract
Varicella zoster virus (VZV) is a highly prevalent human pathogen that establishes latency in neurons of the peripheral nervous system. Primary infection causes varicella whereas reactivation results in zoster, which is often followed by chronic pain in adults. Following infection of epithelial cells in the respiratory tract, VZV spreads within the host by hijacking leukocytes, including T cells, in the tonsils and other regional lymph nodes, and modifying their activity. In spite of its importance in pathogenesis, the mechanism of dissemination remains poorly understood. Here we addressed the influence of VZV on leukocyte migration and found that the purified recombinant soluble ectodomain of VZV glycoprotein C (rSgC) binds chemokines with high affinity. Functional experiments show that VZV rSgC potentiates chemokine activity, enhancing the migration of monocyte and T cell lines and, most importantly, human tonsillar leukocytes at low chemokine concentrations. Binding and potentiation of chemokine activity occurs through the C-terminal part of gC ectodomain, containing predicted immunoglobulin-like domains. The mechanism of action of VZV rSgC requires interaction with the chemokine and signalling through the chemokine receptor. Finally, we show that VZV viral particles enhance chemokine-dependent T cell migration and that gC is partially required for this activity. We propose that VZV gC activity facilitates the recruitment and subsequent infection of leukocytes and thereby enhances VZV systemic dissemination in humans. Varicella zoster virus (VZV) causes two main pathologies in humans, chickenpox during primary infection, and shingles following reactivation. The latter is a painful condition that is often followed by chronic pain in a large numbers of shingles patients. Despite the existence of a vaccine, shingles-related complications cause expenses of more than $1 billion per year in the USA alone. Following primary infection, the virus infects leukocytes including T cells, spreading to the skin causing chickenpox. Direct infection of neurons from leukocytes has also been postulated. Given the relevance of leukocytes in VZV biology and the importance of chemokines in directing their migration, we investigated whether VZV modulates the function of chemokines. Our results show that VZV glycoprotein C potentiates the activity of chemokines, inducing higher migration of human leukocytes at low chemokine concentration. This may attract additional susceptible leukocytes to the site of infection enhancing virus spread and pathogenesis.
Collapse
|
26
|
Mechanisms of immunomodulation by mammalian and viral decoy receptors: insights from structures. Nat Rev Immunol 2016; 17:112-129. [PMID: 28028310 DOI: 10.1038/nri.2016.134] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Immune responses are regulated by effector cytokines and chemokines that signal through cell surface receptors. Mammalian decoy receptors - which are typically soluble or inactive versions of cell surface receptors or soluble protein modules termed binding proteins - modulate and antagonize signalling by canonical effector-receptor complexes. Viruses have developed a diverse array of molecular decoys to evade host immune responses; these include viral homologues of host cytokines, chemokines and chemokine receptors; variants of host receptors with new functions; and novel decoy receptors that do not have host counterparts. Over the past decade, the number of known mammalian and viral decoy receptors has increased considerably, yet a comprehensive curation of the corresponding structure-mechanism relationships has not been carried out. In this Review, we provide a comprehensive resource on this topic with a view to better understanding the roles and evolutionary relationships of mammalian and viral decoy receptors, and the opportunities for leveraging their therapeutic potential.
Collapse
|
27
|
Sharif S, Nakatani Y, Wise L, Corbett M, Real NC, Stuart GS, Lateef Z, Krause K, Mercer AA, Fleming SB. A Broad-Spectrum Chemokine-Binding Protein of Bovine Papular Stomatitis Virus Inhibits Neutrophil and Monocyte Infiltration in Inflammatory and Wound Models of Mouse Skin. PLoS One 2016; 11:e0168007. [PMID: 27936239 PMCID: PMC5148066 DOI: 10.1371/journal.pone.0168007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 11/23/2016] [Indexed: 12/21/2022] Open
Abstract
Bovine papular stomatitis virus (BPSV) is a Parapoxvirus that induces acute pustular skin lesions in cattle and is transmissible to humans. Previous studies have shown that BPSV encodes a distinctive chemokine-binding protein (CBP). Chemokines are critically involved in the trafficking of immune cells to sites of inflammation and infected tissue, suggesting that the CBP plays a role in immune evasion by preventing immune cells reaching sites of infection. We hypothesised that the BPSV-CBP binds a wide range of inflammatory chemokines particularly those involved in BPSV skin infection, and inhibits the recruitment of immune cells from the blood into inflamed skin. Molecular analysis of the purified protein revealed that the BPSV-CBP is a homodimeric polypeptide with a MW of 82.4 kDa whilst a comprehensive screen of inflammatory chemokines by surface plasmon resonance showed high-affinity binding to a range of chemokines within the CXC, CC and XC subfamilies. Structural analysis of BPSV-CBP, based on the crystal structure of orf virus CBP, provided a probable explanation for these chemokine specificities at a molecular level. Functional analysis of the BPSV-CBP using transwell migration assays demonstrated that it potently inhibited chemotaxis of murine neutrophils and monocytes in response to CXCL1, CXCL2 as well as CCL2, CCL3 and CCL5 chemokines. In order to examine the effects of CBP in vivo, we used murine skin models to determine its impact on inflammatory cell recruitment such as that observed during BPSV infection. Intradermal injection of BPSV-CBP blocked the influx of neutrophils and monocytes in murine skin in which inflammation was induced with lipopolysaccharide. Furthermore, intradermal injection of BPSV-CBP into injured skin, which more closely mimics BPSV lesions, delayed the influx of neutrophils and reduced the recruitment of MHC-II+ immune cells to the wound bed. Our findings suggest that the CBP could be important in pathogenesis of BPSV infections.
Collapse
Affiliation(s)
- Saeed Sharif
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Yoshio Nakatani
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Lyn Wise
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Michael Corbett
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Nicola C. Real
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Gabriella S. Stuart
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Zabeen Lateef
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Kurt Krause
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Andrew A. Mercer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B. Fleming
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- * E-mail:
| |
Collapse
|
28
|
González-Motos V, Kropp KA, Viejo-Borbolla A. Chemokine binding proteins: An immunomodulatory strategy going viral. Cytokine Growth Factor Rev 2016; 30:71-80. [DOI: 10.1016/j.cytogfr.2016.02.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/27/2016] [Indexed: 01/13/2023]
|
29
|
Bonvin P, Power CA, Proudfoot AEI. Evasins: Therapeutic Potential of a New Family of Chemokine-Binding Proteins from Ticks. Front Immunol 2016; 7:208. [PMID: 27375615 PMCID: PMC4894869 DOI: 10.3389/fimmu.2016.00208] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/15/2016] [Indexed: 02/03/2023] Open
Abstract
Blood-sucking parasites, such as ticks, remain attached to their hosts for relatively long periods of time in order to obtain their blood meal without eliciting an immune response. One mechanism used to avoid rejection is the inhibition of the recruitment of immune cells, which can be achieved by a class of chemokine-binding proteins (CKBPs) known as Evasins. We have identified three distinct Evasins produced by the salivary glands of the common brown dog tick, Rhipicephalus sanguineus. They display different selectivities for chemokines, the first two identified show a narrow selectivity profile, while the third has a broader binding spectrum. The Evasins showed efficacy in animal models of inflammatory disease. Here, we will discuss the potential of their development for therapeutic use, addressing both the advantages and disadvantages that this entails.
Collapse
Affiliation(s)
- Pauline Bonvin
- Geneva Research Centre, Merck Serono S.A., Geneva, Switzerland; Research Department, Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | - Amanda E I Proudfoot
- Geneva Research Centre, Merck Serono S.A., Geneva, Switzerland; Research Department, Novimmune S.A., Plan-les-Ouates, Switzerland
| |
Collapse
|
30
|
Heidarieh H, Hernáez B, Alcamí A. Immune modulation by virus-encoded secreted chemokine binding proteins. Virus Res 2015; 209:67-75. [PMID: 25791735 DOI: 10.1016/j.virusres.2015.02.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/23/2015] [Indexed: 10/23/2022]
Abstract
Chemokines are chemoattractant cytokines that mediate the migration of immune cells to sites of infection which play an important role in innate and adaptive immunity. As an immune evasion strategy, large DNA viruses (herpesviruses and poxviruses) encode soluble chemokine binding proteins that bind chemokines with high affinity, even though they do not show sequence similarity to cellular chemokine receptors. This review summarizes the different secreted viral chemokine binding proteins described to date, with special emphasis on the diverse mechanisms of action they exhibit to interfere with chemokine function and their specific contribution to virus pathogenesis.
Collapse
Affiliation(s)
- Haleh Heidarieh
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid), Cantoblanco, Madrid, Spain
| | - Bruno Hernáez
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid), Cantoblanco, Madrid, Spain
| | - Antonio Alcamí
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid), Cantoblanco, Madrid, Spain.
| |
Collapse
|
31
|
Baker L, Chitas AML, Hartley CA, Coppo MJC, Vaz PK, Stent A, Gilkerson JR, Devlin JM, Every AL. Recombinant herpesvirus glycoprotein G improves the protective immune response to Helicobacter pylori vaccination in a mouse model of disease. PLoS One 2014; 9:e96563. [PMID: 24794215 PMCID: PMC4008605 DOI: 10.1371/journal.pone.0096563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/08/2014] [Indexed: 12/12/2022] Open
Abstract
Alphaherpesviruses, which have co-evolved with their hosts for more than 200 million years, evade and subvert host immune responses, in part, by expression of immuno-modulatory molecules. Alphaherpesviruses express a single, broadly conserved chemokine decoy receptor, glycoprotein G (gG), which can bind multiple chemokine classes from multiple species, including human and mouse. Previously, we demonstrated that infection of chickens with an infectious laryngotracheitis virus (ILTV) mutant deficient in gG resulted in altered host immune responses compared to infection with wild-type virus. The ability of gG to disrupt the chemokine network has the potential to be used therapeutically. Here we investigated whether gG from ILTV or equine herpesvirus 1 (EHV-1) could modulate the protective immune response induced by the Helicobacter pylori vaccine antigen, catalase (KatA). Subcutaneous immunisation of mice with KatA together with EHV-1 gG, but not ILTV gG, induced significantly higher anti-KatA IgG than KatA alone. Importantly, subcutaneous or intranasal immunisation with KatA and EHV-1 gG both resulted in significantly lower colonization levels of H. pylori colonization following challenge, compared to mice vaccinated with KatA alone. Indeed, the lowest colonization levels were observed in mice vaccinated with KatA and EHV-1 gG, subcutaneously. In contrast, formulations containing ILTV gG did not affect H. pylori colonisation levels. The difference in efficacy between EHV-1 gG and ILTV gG may reflect the different spectrum of chemokines bound by the two proteins. Together, these data indicate that the immuno-modulatory properties of viral gGs could be harnessed for improving immune responses to vaccine antigens. Future studies should focus on the mechanism of action and whether gG may have other therapeutic applications.
Collapse
Affiliation(s)
- Louise Baker
- Centre for Animal Biotechnology, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Andre M. L. Chitas
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Carol A. Hartley
- Centre for Equine Infectious Disease, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Mauricio J. C. Coppo
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Paola K. Vaz
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew Stent
- Centre for Animal Biotechnology, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - James R. Gilkerson
- Centre for Equine Infectious Disease, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Joanne M. Devlin
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Alison L. Every
- Centre for Animal Biotechnology, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
- * E-mail:
| |
Collapse
|
32
|
van Drunen Littel-van den Hurk S. Cell-mediated immune responses induced by BHV-1: rational vaccine design. Expert Rev Vaccines 2014; 6:369-80. [PMID: 17542752 DOI: 10.1586/14760584.6.3.369] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Bovine herpesvirus-1 (BHV-1) is one of the major respiratory pathogens in cattle worldwide. Although antibodies have been correlated with protection and recovery from BHV-1 infection, the cell-mediated immune response is also a critical defense mechanism because cell-to-cell spread occurs before hematogenous spread. Furthermore, induction of robust T-cell memory is critical for the long-term duration of immunity. Among current commercial vaccines, the attenuated conventional vaccines induce a balanced immune response and long-term memory but may result in viral shedding. By contrast, inactivated vaccines primarily elicit a humoral immune response and relative short-term memory. These vaccines do not allow differentiation of vaccinated from infected cattle. Recent efforts are focusing on the development of vaccines that induce a balanced immune response and long-term memory, as well as having differentiation markers. This includes well-defined genetically engineered gene-deleted, subunit and vectored vaccines.
Collapse
|
33
|
Vidick S, Leroy B, Palmeira L, Machiels B, Mast J, François S, Wattiez R, Vanderplasschen A, Gillet L. Proteomic characterization of murid herpesvirus 4 extracellular virions. PLoS One 2013; 8:e83842. [PMID: 24386290 PMCID: PMC3875534 DOI: 10.1371/journal.pone.0083842] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 11/18/2013] [Indexed: 12/18/2022] Open
Abstract
Gammaherpesvirinae, such as the human Epstein-Barr virus (EBV) and the Kaposi’s sarcoma associated herpesvirus (KSHV) are highly prevalent pathogens that have been associated with several neoplastic diseases. As EBV and KSHV are host-range specific and replicate poorly in vitro, animal counterparts such as Murid herpesvirus-4 (MuHV-4) have been widely used as models. In this study, we used MuHV-4 in order to improve the knowledge about proteins that compose gammaherpesviruses virions. To this end, MuHV-4 extracellular virions were isolated and structural proteins were identified using liquid chromatography tandem mass spectrometry-based proteomic approaches. These analyses allowed the identification of 31 structural proteins encoded by the MuHV-4 genome which were classified as capsid (8), envelope (9), tegument (13) and unclassified (1) structural proteins. In addition, we estimated the relative abundance of the identified proteins in MuHV-4 virions by using exponentially modified protein abundance index analyses. In parallel, several host proteins were found in purified MuHV-4 virions including Annexin A2. Although Annexin A2 has previously been detected in different virions from various families, its role in the virion remains controversial. Interestingly, despite its relatively high abundance in virions, Annexin A2 was not essential for the growth of MuHV-4 in vitro. Altogether, these results extend previous work aimed at determining the composition of gammaherpesvirus virions and provide novel insights for understanding MuHV-4 biology.
Collapse
Affiliation(s)
- Sarah Vidick
- Department of Infectious Diseases, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Baptiste Leroy
- Department of Proteomics and Microbiology, Research Institute for Biosciences Interdisciplinary Mass Spectrometry Center (CISMa), University of Mons, Mons, Belgium
| | - Leonor Palmeira
- Department of Infectious Diseases, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Bénédicte Machiels
- Department of Infectious Diseases, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Jan Mast
- Electron Microscopy Unit, Veterinary and Agrochemical Research Centre, Brussels, Belgium
| | - Sylvie François
- Department of Infectious Diseases, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Ruddy Wattiez
- Department of Proteomics and Microbiology, Research Institute for Biosciences Interdisciplinary Mass Spectrometry Center (CISMa), University of Mons, Mons, Belgium
| | - Alain Vanderplasschen
- Department of Infectious Diseases, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Laurent Gillet
- Department of Infectious Diseases, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
34
|
Coppo MJC, Hartley CA, Devlin JM. Immune responses to infectious laryngotracheitis virus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:454-462. [PMID: 23567343 DOI: 10.1016/j.dci.2013.03.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 03/28/2013] [Accepted: 03/28/2013] [Indexed: 06/02/2023]
Abstract
Infectious laryngotracheitis (ILT) is an upper respiratory tract disease in chickens caused by infectious laryngotracheitis virus (ILTV), an alphaherpesvirus. Despite the extensive use of attenuated, and more recently recombinant, vaccines for the control of this disease, ILT continues to affect the intensive poultry industries worldwide. Innate and cell-mediated, rather than humoral immune responses, have been identified as responsible for protection against disease. This review examines the current understandings in innate and adaptive immune responses towards ILTV, as well as the role of ILTV glycoprotein G in modulating the host immune response towards infection. Protective immunity induced by ILT vaccines is also examined. The increasing availability of tools and reagents for the characterisation of avian innate and cell-mediated immune responses are expected to further our understanding of immunity against ILTV and drive the development of new generation vaccines towards enhanced control of this disease.
Collapse
Affiliation(s)
- Mauricio J C Coppo
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Victoria, Australia
| | | | | |
Collapse
|
35
|
Ma G, Azab W, Osterrieder N. Equine herpesviruses type 1 (EHV-1) and 4 (EHV-4)—Masters of co-evolution and a constant threat to equids and beyond. Vet Microbiol 2013; 167:123-34. [DOI: 10.1016/j.vetmic.2013.06.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 05/08/2013] [Accepted: 06/28/2013] [Indexed: 12/28/2022]
|
36
|
Liu Z, Bethunaickan R, Sahu R, Brenner M, Laragione T, Gulko PS, Davidson A. The Multiple Chemokine-Binding Bovine Herpesvirus 1 Glycoprotein G (BHV1gG) Inhibits Polymorphonuclear Cell but Not Monocyte Migration into Inflammatory Sites. Mol Med 2013. [DOI: 10.2119/molmed.2012.00339] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
37
|
Biswas S, Bandyopadhyay S, Dimri U, H. Patra P. Bovine herpesvirus-1 (BHV-1) – a re-emerging concern in livestock: a revisit to its biology, epidemiology, diagnosis, and prophylaxis. Vet Q 2013; 33:68-81. [DOI: 10.1080/01652176.2013.799301] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Suman Biswas
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, Bareilly, India
| | | | - Umesh Dimri
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, Bareilly, India
| | - Pabitra H. Patra
- Department of Pharmacology & Toxicology, C.V.Sc. & AH, Tripura, India
| |
Collapse
|
38
|
Abstract
AbstractBovine herpesvirus 1 (BHV-1) causes a variety of diseases and is globally distributed. It infects via mucosal epithelium, leading to rapid lytic replication and latent infection, primarily in sensory ganglia. Large amounts of virus can be excreted by the host on primary infection or upon recrudescence of latent infection, resulting in disease spread. The bovine immune response to BHV-1 is rapid, robust, balanced, and long-lasting. The innate immune system is the first to respond to the infection, with type I interferons (IFNs), inflammatory cytokines, killing of infected host cells, and priming of a balanced adaptive immune response. The virus possesses a variety of immune evasion strategies, including inhibition of type I IFN production, chemokine and complement binding, infection of macrophages and neutrophils, and latency. BHV-1 immune suppression contributes to the severity of its disease manifestations and to the bovine respiratory disease complex, the leading cause of cattle death loss in the USA.
Collapse
|
39
|
Mahmoudian A, Markham PF, Noormohammadi AH, Devlin JM, Browning GF. Differential transcription patterns in wild-type and glycoprotein G-deleted infectious laryngotracheitis viruses. Avian Pathol 2013; 42:253-9. [PMID: 23611157 DOI: 10.1080/03079457.2013.780649] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Infectious laryngotracheitis virus (ILTV) causes severe respiratory disease in poultry throughout the world. Recently the role of glycoprotein G (gG) in ILTV pathogenesis has been investigated and it has been shown to have chemokine-binding activity. An ILTV vaccine candidate deficient in gG has been developed and the deletion has been shown to alter the host's immune response to the virus. To understand the effect of the gG gene on transcription of other viral genes, the global expression profile of 72 ILTV genes in gG-deleted and wild-type ILTVs were investigated both in vivo and in vitro using quantitative reverse transcription-polymerase chain reaction. Several genes were differentially expressed in the different viruses in LMH cell cultures or in the tracheas of infected birds, and the expression of a number of genes, including ICP27, gC, gJ, Ul7 and UL40, differed significantly both in vivo and in vitro, suggesting that they had direct or indirect roles in virulence. This study has provided insights into the interactions between gG and other ILTV genes that may have a role in virulence.
Collapse
Affiliation(s)
- Alireza Mahmoudian
- Asia-Pacific Centre for Animal Health, The University of Melbourne, Melbourne, Victoria, Australia.
| | | | | | | | | |
Collapse
|
40
|
Abstract
Mice are commonly used as an experimental model to investigate the Equid herpesvirus 1 (EHV-1) infection. This model easily reproduces the disease, and the clinical signs are more or less similar to those observed in the horse, the natural host. During natural infection, the acute course of respiratory infection is mandatory for the development of adaptive immune response. Since interactions between EHV-1 and anesthetics are possible, the study investigated whether the early events of murine pulmonary immune response could be affected by different anesthetics. Therefore, mice were experimentally infected with a unique EHV-1 strain under the effects of ether, ketamine/xylazine, or isoflurane. Clinical signs and histopathological lesions in the lungs were described, and the cell death and proliferation rates of sham-inoculated or infected animals were quantified using immunohistochemistry. Clinical signs were more severe in animals anesthetized with ether. Qualitative differences in the recruited inflammatory cells were observed following application of anesthesia. The level of infection between the infected groups was not statistically significant. However, lungs from ketamine/xylazine-anesthetized animals showed the highest cell death rates, whereas those from isoflurane-anesthetized animals showed the highest proliferation rates. It has been emphasized that anesthetics alone or their interactions with EHV-1 modify the response against the infection. An appropriate selection of the anesthetic during experimental studies is relevant to minimize wrong conclusions.
Collapse
|
41
|
Epperson ML, Lee CA, Fremont DH. Subversion of cytokine networks by virally encoded decoy receptors. Immunol Rev 2012; 250:199-215. [PMID: 23046131 PMCID: PMC3693748 DOI: 10.1111/imr.12009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
During the course of evolution, viruses have captured or created a diverse array of open reading frames, which encode for proteins that serve to evade and sabotage the host innate and adaptive immune responses that would otherwise lead to their elimination. These viral genomes are some of the best textbooks of immunology ever written. The established arsenal of immunomodulatory proteins encoded by viruses is large and growing, and includes specificities for virtually all known inflammatory pathways and targets. The focus of this review is on herpes and poxvirus-encoded cytokine and chemokine-binding proteins that serve to undermine the coordination of host immune surveillance. Structural and mechanistic studies of these decoy receptors have provided a wealth of information, not only about viral pathogenesis but also about the inner workings of cytokine signaling networks.
Collapse
Affiliation(s)
- Megan L Epperson
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
42
|
Legione AR, Coppo MJC, Lee SW, Noormohammadi AH, Hartley CA, Browning GF, Gilkerson JR, O'Rourke D, Devlin JM. Safety and vaccine efficacy of a glycoprotein G deficient strain of infectious laryngotracheitis virus delivered in ovo. Vaccine 2012; 30:7193-8. [PMID: 23084851 DOI: 10.1016/j.vaccine.2012.10.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 08/21/2012] [Accepted: 10/05/2012] [Indexed: 10/27/2022]
Abstract
Infectious laryngotracheitis virus (ILTV), an alphaherpesvirus, causes respiratory disease in chickens and is commonly controlled by vaccination with conventionally attenuated vaccines. Glycoprotein G (gG) is a virulence factor in ILTV and a gG deficient strain of ILTV (ΔgG-ILTV) has shown potential for use as a vaccine. In the poultry industry vaccination via drinking water is common, but technology is now available to allow quicker and more accurate in ovo vaccination of embryos at 18 days of incubation. In this study ΔgG-ILTV was delivered to chicken embryos at three different doses (10(2), 10(3) and 10(4) plaque forming units per egg) using manual in ovo vaccination. At 20 days after hatching, birds were challenged intra-tracheally with wild type ILTV and protection was measured. In ovo vaccination was shown to be safe, as there were no developmental differences between birds from hatching up to 20 days of age, as measured by weight gain. The highest dose of vaccine was the most efficacious, resulting in a weight gain not significantly different from unvaccinated/unchallenged birds seven days after challenge. In contrast, birds vaccinated with the lowest dose showed weight gains not significantly different from unvaccinated/challenged birds. Gross pathology and histopathology of the trachea reflected these observations, with birds vaccinated with the highest dose having less severe lesions. However, qPCR results suggested the vaccine did not prevent the challenge virus replicating in the trachea. This study is the first to assess in ovo delivery of a live attenuated ILTV vaccine and shows that in ovo vaccination with ΔgG-ILTV can be both safe and efficacious.
Collapse
Affiliation(s)
- Alistair R Legione
- Asia-Pacific Centre for Animal Health, The University of Melbourne, Victoria, 3010, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
The role of secreted glycoprotein G of equine herpesvirus type 1 and type 4 (EHV-1 and EHV-4) in immune modulation and virulence. Virus Res 2012; 169:203-11. [PMID: 22902480 DOI: 10.1016/j.virusres.2012.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 07/23/2012] [Accepted: 07/25/2012] [Indexed: 11/24/2022]
Abstract
Equine herpesvirus type 1 and 4 (EHV-1 and EHV-4) are important pathogens of horses worldwide. Infection with EHV-4 usually remains restricted to the upper respiratory tract, whereas infection with EHV-1 can generalize after leukocyte-associated viremia. Here we examined whether differences in the immunomodulatory glycoprotein G (gG) between the two viruses determine EHV-1's ability to cause systemic infection. To this end, mutant viruses were constructed based on the neurovirulent EHV-1 strain OH-03, in which the entire gG gene or parts thereof were exchanged with EHV-4 gG sequences. In vitro chemotaxis assays showed that supernatants of cells infected with the various gG mutant viruses interfered to variable degrees with neutrophil migration. More specifically, supernatants of cells infected with the gG deletion virus (vOH-ΔgG1) or OH-03 expressing EHV-4 gG (vOH-gG4) were unable to interfere with chemotaxis. Re-insertion of the predicted chemokine-binding region of EHV-1 gG in the vOH-gG4 mutant (vOH-gG4hyp1) did not completely restore the ability to inhibit neutrophil migration, whereas insertion of the hypervariable region of EHV-4 gG into vOH-03 (vOH-gG1hyp4) did not lead to a complete loss of chemokine-binding function. Very similar results were obtained in an in vivo study where the amount of neutrophils present in bronchioalveolar lavages (BALs) of mice infected with the different mutants was analyzed by flow cytometry. Taken together, our results show that, in a virus background, the hypervariable region is not solely responsible for the immunomodulatory potential of EHV-1 gG.
Collapse
|
44
|
Van de Walle GR, Osterrieder N. Profiling chemokine–glycoprotein G interactions: implications for alphaherpesviral immune evasion. Future Virol 2012. [DOI: 10.2217/fvl.12.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Evaluation of: Viejo-Borbolla A, Martinez-Martín N, Nel HJ et al. Enhancement of chemokine function as an immunomodulatory strategy employed by human herpesviruses. PLoS Pathog. 8(2), e1002497 (2012). The study of immunomodulation by alphaherpesviral proteins targeting the chemokine network remains an area of active research. The article by Viejo-Borbolla et al. evaluates the modulation of chemokines by human HSV-1 and HSV-2. The authors report that secreted recombinant glycoprotein G (gG) of both viruses was able to bind with high affinity to a wide range of CC and CXC chemokines. Interestingly, and in contrast to other viral chemokine binding proteins produced by animal herpesviruses, the investigators found that human herpesvirus-encoded secreted gG1 and secreted gG2 do enhance and not inhibit chemotaxis. This article provides additional insights into the role in immune evasion of alphaherpesviral gGs, but at the same time raises intriguing questions. Among those questions are why and when animal and human alphaherpesviruses diverged in their strategies to manipulate the actions of chemokines and how these apparent differences influence pathogenesis and the final outcome of infection.
Collapse
Affiliation(s)
- Gerlinde R Van de Walle
- Department of Comparative Physiology & Biometrics, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium
| | - Nikolaus Osterrieder
- Institute für Virologie, Freie Universität Berlin, Philippstraße 13, 10115 Berlin, Germany
| |
Collapse
|
45
|
Harishankar A, Jambulingam M, Gowrishankar R, Venkatachalam A, Vetrivel U, Ravichandran S, Yesupadam SM, Madhavan HNR. Phylogenetic comparison of exonic US4, US7 and UL44 regions of clinical herpes simplex virus type 1 isolates showed lack of association between their anatomic sites of infection and genotypic/sub genotypic classification. Virol J 2012; 9:65. [PMID: 22416856 PMCID: PMC3359161 DOI: 10.1186/1743-422x-9-65] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 03/14/2012] [Indexed: 11/21/2022] Open
Abstract
Background HSV-1 genome is a mosaic of recombinants. Clinical Herpes simplex virus -1 (HSV1) isolates were already genotyped as A, B and C types based on nucleotide variations at Unique Short (US) 4 (gG) and US 7 (gI) regions through phylogeny. Analysis of Glycoprotein C (gC) exon present on the Unique Long (UL) region had also revealed the existence of different genotypes. Glycoprotein C is mainly involved in initial viral attachment to heparan sulphate on host cell surface facilitating the virus's binding and penetration into cell. As the amount of heparan sulphate on the host cell surface varies according to the cell type, it is plausible that different genotypes bind differentially to cell types. Hence, this study was framed to determine the existence of novel genotypes/sub genotypes in the US or UL regions which could associate with clinical entities. Results All the twenty five isolates analyzed in this study were of genotype A as per their gG gene sequences. In case of gI gene, 16 out of 25 were found to be type A and the remaining nine were type B putative intergenic recombinants. Intragenic recombinations were also encountered in both the US genes, with gG possessing novel subgenotypes, arbitrarily designated A1 and A2. The 9 type B isolates of gI genes also branched out into 2 clades due to genetic variations. Glycoprotein C of UL region had two distinct genotypic clades α and β, whose topological distribution was significantly different from that of the US region. Neither the US nor UL regions, however, showed any preference among the genotypes to a specific anatomic site of infection. Even the non synonymous variations identified in the functional domain of gC, were not confined to a particular genotype/clinical entity. Conclusion The analyses of the US and UL regions of the HSV-1 genome showed the existence of variegated genotypes in these two regions. In contrary to the documented literature, in which Asian strains were concluded as more conserved than European ones, our study showed the existence of a higher degree of variability among Indian strains. However, the identified novel genotypes and subgenotypes were not found associated with clinical entities.
Collapse
Affiliation(s)
- Anusha Harishankar
- Larsen & Toubro Microbiology Research Centre, Sankara Nethralaya, No,18, College Road, Chennai 600006, India
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Enhancement of chemokine function as an immunomodulatory strategy employed by human herpesviruses. PLoS Pathog 2012; 8:e1002497. [PMID: 22319442 PMCID: PMC3271085 DOI: 10.1371/journal.ppat.1002497] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 12/06/2011] [Indexed: 01/10/2023] Open
Abstract
Herpes simplex virus (HSV) types 1 and 2 are highly prevalent human neurotropic pathogens that cause a variety of diseases, including lethal encephalitis. The relationship between HSV and the host immune system is one of the main determinants of the infection outcome. Chemokines play relevant roles in antiviral response and immunopathology, but the modulation of chemokine function by HSV is not well understood. We have addressed the modulation of chemokine function mediated by HSV. By using surface plasmon resonance and crosslinking assays we show that secreted glycoprotein G (SgG) from both HSV-1 and HSV-2 binds chemokines with high affinity. Chemokine binding activity was also observed in the supernatant of HSV-2 infected cells and in the plasma membrane of cells infected with HSV-1 wild type but not with a gG deficient HSV-1 mutant. Cell-binding and competition experiments indicate that the interaction takes place through the glycosaminoglycan-binding domain of the chemokine. The functional relevance of the interaction was determined both in vitro, by performing transwell assays, time-lapse microscopy, and signal transduction experiments; and in vivo, using the air pouch model of inflammation. Interestingly, and in contrast to what has been observed for previously described viral chemokine binding proteins, HSV SgGs do not inhibit chemokine function. On the contrary, HSV SgGs enhance chemotaxis both in vitro and in vivo through increasing directionality, potency and receptor signaling. This is the first report, to our knowledge, of a viral chemokine binding protein from a human pathogen that increases chemokine function and points towards a previously undescribed strategy of immune modulation mediated by viruses. Chemokines are chemotactic cytokines that direct the flux of leukocytes to the site of injury and infection, playing a relevant role in the antiviral response. An uncontrolled, unorganized chemokine response is beneath the onset and maintenance of several immunopathologies. During millions of years of evolution, viruses have developed strategies to modulate the host immune system. One of such strategies consists on the secretion of viral proteins that bind to and inhibit the function of chemokines. However, the modulation of the chemokine network mediated by the highly prevalent human pathogen herpes simplex virus (HSV) is unknown. We have addressed this issue and show that HSV-1, causing cold sores and encephalitis and HSV-2, causing urogenital tract infections, interact with chemokines. We determined that the viral protein responsible for such activity is glycoprotein G (gG). gG binds chemokines with high affinity and, in contrast to all viral chemokine binding proteins described to date that inhibit chemokine function, we found that HSV gG potentiates chemokine function in vitro and in vivo. The implications of such potentiation in HSV viral cycle, pathogenesis and chemokine function are discussed.
Collapse
|
47
|
Azab W, El-Sheikh A, Abdel-Gawad A. In vitro characterization of EHV-4 gG-deleted mutant. Virus Genes 2011; 44:109-11. [PMID: 21960433 DOI: 10.1007/s11262-011-0677-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2011] [Accepted: 09/16/2011] [Indexed: 11/25/2022]
Abstract
Equine herpesvirus 4 (EHV-4) is an important pathogen that causes respiratory tract disease in horse populations worldwide. Glycoprotein G (gG) homologs have been identified in several alphaherpesviruses as minor non-essential membrane-anchored glycoproteins. In this study, EHV-4 gG deletion mutant has been generated by using bacterial artificial chromosome technology to investigate the role of gG in viral pathogenesis. Our findings reported here revealed no significant difference between parental EHV-4 and gG-negative strain in their replication cycle in cell culture. Furthermore, virus titers and plaque formation were comparable in both viruses. It is noteworthy that these findings disagree with the previously published study describing gG deletion in another EHV-4 strain.
Collapse
Affiliation(s)
- Walid Azab
- Department of Virology, Zagazig University, Zagazig, Egypt.
| | | | | |
Collapse
|
48
|
Zhang M, Fu S, Deng M, Xie Q, Xu H, Liu Z, Hu C, Chen H, Guo A. Attenuation of bovine herpesvirus type 1 by deletion of its glycoprotein G and tk genes and protection against virulent viral challenge. Vaccine 2011; 29:8943-50. [PMID: 21959327 DOI: 10.1016/j.vaccine.2011.09.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 09/09/2011] [Accepted: 09/13/2011] [Indexed: 10/17/2022]
Abstract
To develop a novel vaccine against infectious bovine rhinotracheitis (IBR), a bovine herpesvirus-1 (BoHV-1) mutant was constructed by deleting the genes for glycoprotein G (gG) and thymidine kinase (tk) through homologous recombination. The resulting sequences for both genes were shown to be correct and a gG expression defect was also confirmed. A parallel study of the BoHV-1 gG(-)/tk(-), gE(-)/tk(-) mutants and wild type (wt) in 31 calves was performed at three different doses, 4×10(5)PFU, 4×10(6)PFU and 4×10(7)PFU. Compared to wt BoHV-1, inoculation of BoHV-1 gG(-)/tk(-) and gE(-)/tk(-) produced no clinical signs and the virus was not reactivated by dexamethasone (dex). Inoculation of BoHV-1 gG(-)/tk(-) at the doses of 4×10(6) and 4×10(7)PFU provided full clinical protection for the cattle against wt BoHV-1 challenge at 4×10(7)PFU/calf. Although the mutants were associated with significantly lower levels of serum neutralizing antibody, interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) than wt BoHV-1 on days 3, 5 and 7 after immunization, stimulation of IFN-β by BoHV-1 gG(-)/tk(-) was significantly higher than that of wt BoHV-1 and gE(-)/tk(-) on days 3 and 5. We conclude that BoHV-1 gG(-)/tk(-) was attenuated adequately and that it maintains the ability to stimulate immune protection. Therefore, it may be a promising candidate for a marker vaccine against IBR.
Collapse
Affiliation(s)
- Minmin Zhang
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Xue X, Lu Q, Wei H, Wang D, Chen D, He G, Huang L, Wang H, Wang X. Structural basis of chemokine sequestration by CrmD, a poxvirus-encoded tumor necrosis factor receptor. PLoS Pathog 2011; 7:e1002162. [PMID: 21829356 PMCID: PMC3145792 DOI: 10.1371/journal.ppat.1002162] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 05/28/2011] [Indexed: 11/25/2022] Open
Abstract
Pathogens have evolved sophisticated mechanisms to evade detection and destruction by the host immune system. Large DNA viruses encode homologues of chemokines and their receptors, as well as chemokine-binding proteins (CKBPs) to modulate the chemokine network in host response. The SECRET domain (smallpox virus-encoded chemokine receptor) represents a new family of viral CKBPs that binds a subset of chemokines from different classes to inhibit their activities, either independently or fused with viral tumor necrosis factor receptors (vTNFRs). Here we present the crystal structures of the SECRET domain of vTNFR CrmD encoded by ectromelia virus and its complex with chemokine CX3CL1. The SECRET domain adopts a β-sandwich fold and utilizes its β-sheet I surface to interact with CX3CL1, representing a new chemokine-binding manner of viral CKBPs. Structure-based mutagenesis and biochemical analysis identified important basic residues in the 40s loop of CX3CL1 for the interaction. Mutation of corresponding acidic residues in the SECRET domain also affected the binding for other chemokines, indicating that the SECRET domain binds different chemokines in a similar manner. We further showed that heparin inhibited the binding of CX3CL1 by the SECRET domain and the SECRET domain inhibited RAW264.7 cell migration induced by CX3CL1. These results together shed light on the structural basis for the SECRET domain to inhibit chemokine activities by interfering with both chemokine-GAG and chemokine-receptor interactions. Chemokines are a family of small proteins that help the immune system fight against invading pathogens by inducing the white blood cells to the areas of infection and inflammation. Due to the important roles of chemokines in immune response, the pathogens evolve diverse mechanisms to neutralize their activities. One example is that large DNA viruses, such as poxviruses and herpesviruses can produce chemokine binding proteins (CKBPs) to sequester chemokines during the infection. The SECRET domain represents a new family of viral CKBPs that was originally identified as a C-terminal extension of the viral tumor necrosis factor receptors (vTNFRs). We determined the three-dimensional structures of the SECRET domain and its complex with chemokine CX3CL1, revealing a new chemokine-binding manner of viral CKBPs. We also showed that other chemokines from different classes may be bound by the SECRET domain in a way similar to that observed in the SECRET/CX3CL1 complex structure. Our biochemical and chemotaxis assays also suggest that the SECRET domain is able to interfere with both chemokine-GAG and chemokine-receptor interactions, both of which are essential for chemokine activities in vivo.
Collapse
Affiliation(s)
- Xiaoguang Xue
- Center for Structural Biology, School of Life Sciences, Ministry of Education Key Laboratory of Protein Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Qingyu Lu
- Center for Structural Biology, School of Life Sciences, Ministry of Education Key Laboratory of Protein Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Hui Wei
- Center for Structural Biology, School of Life Sciences, Ministry of Education Key Laboratory of Protein Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Dongli Wang
- Center for Structural Biology, School of Life Sciences, Ministry of Education Key Laboratory of Protein Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Dongwei Chen
- School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Guangjun He
- Center for Structural Biology, School of Life Sciences, Ministry of Education Key Laboratory of Protein Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Li Huang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Hanzhong Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Xinquan Wang
- Center for Structural Biology, School of Life Sciences, Ministry of Education Key Laboratory of Protein Sciences, Tsinghua University, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
50
|
Azab W, Kato K, Abdel-Gawad A, Tohya Y, Akashi H. Equine herpesvirus 4: recent advances using BAC technology. Vet Microbiol 2011; 150:1-14. [PMID: 21292410 DOI: 10.1016/j.vetmic.2011.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 12/17/2010] [Accepted: 01/03/2011] [Indexed: 10/18/2022]
Abstract
The equine herpesviruses are major infectious pathogens that threaten equine health. Equine herpesvirus 4 (EHV-4) is an important equine pathogen that causes respiratory tract disease, known as rhinopneumonitis, among horses worldwide. EHV-4 genome manipulation with subsequent understanding of the viral gene functions has always been difficult due to the limited number of susceptible cell lines and the absence of small-animal models of the infection. Efficient generation of mutants of EHV-4 would significantly contribute to the rapid and accurate characterization of the viral genes. This problem has been solved recently by the cloning of the genome of EHV-4 as a stable and infectious bacterial artificial chromosome (BAC) without any deletions of the viral genes. Very low copy BAC vectors are the mainstay of present genomic research because of the high stability of inserted clones and the possibility of mutating any gene target in a relatively short time. Manipulation of EHV-4 genome is now feasible using the power of BAC technology, and should aid greatly in assessing the role of viral genes in the virus-host interaction.
Collapse
Affiliation(s)
- Walid Azab
- Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| | | | | | | | | |
Collapse
|