1
|
Ortiz R, Barajas A, Pons-Grífols A, Trinité B, Tarrés-Freixas F, Rovirosa C, Urrea V, Barreiro A, Gonzalez-Tendero A, Rovira-Rigau M, Cardona M, Ferrer L, Clotet B, Carrillo J, Aguilar-Gurrieri C, Blanco J. Production and Immunogenicity of FeLV Gag-Based VLPs Exposing a Stabilized FeLV Envelope Glycoprotein. Viruses 2024; 16:987. [PMID: 38932278 PMCID: PMC11209239 DOI: 10.3390/v16060987] [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: 04/30/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
The envelope glycoprotein (Env) of retroviruses, such as the Feline leukemia virus (FeLV), is the main target of neutralizing humoral response, and therefore, a promising vaccine candidate, despite its reported poor immunogenicity. The incorporation of mutations that stabilize analogous proteins from other viruses in their prefusion conformation (e.g., HIV Env, SARS-CoV-2 S, or RSV F glycoproteins) has improved their capability to induce neutralizing protective immune responses. Therefore, we have stabilized the FeLV Env protein following a strategy based on the incorporation of a disulfide bond and an Ile/Pro mutation (SOSIP) previously used to generate soluble HIV Env trimers. We have characterized this SOSIP-FeLV Env in its soluble form and as a transmembrane protein present at high density on the surface of FeLV Gag-based VLPs. Furthermore, we have tested its immunogenicity in DNA-immunization assays in C57BL/6 mice. Low anti-FeLV Env responses were detected in SOSIP-FeLV soluble protein-immunized animals; however, unexpectedly no responses were detected in the animals immunized with SOSIP-FeLV Gag-based VLPs. In contrast, high humoral response against FeLV Gag was observed in the animals immunized with control Gag VLPs lacking SOSIP-FeLV Env, while this response was significantly impaired when the VLPs incorporated SOSIP-FeLV Env. Our data suggest that FeLV Env can be stabilized as a soluble protein and can be expressed in high-density VLPs. However, when formulated as a DNA vaccine, SOSIP-FeLV Env remains poorly immunogenic, a limitation that must be overcome to develop an effective FeLV vaccine.
Collapse
MESH Headings
- Animals
- Mice
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice, Inbred C57BL
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/genetics
- Leukemia Virus, Feline/immunology
- Leukemia Virus, Feline/genetics
- Gene Products, gag/immunology
- Gene Products, gag/genetics
- Female
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/administration & dosage
- Humans
- Cats
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Immunogenicity, Vaccine
Collapse
Affiliation(s)
- Raquel Ortiz
- IrsiCaixa, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Ana Barajas
- IrsiCaixa, 08916 Badalona, Spain
- Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
| | - Anna Pons-Grífols
- IrsiCaixa, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | | | | | | | | | | | | | | | | | | | - Bonaventura Clotet
- IrsiCaixa, 08916 Badalona, Spain
- Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Infectious Diseases Department, Germans Trias I Pujol Hospital, 08916 Badalona, Spain
| | | | | | - Julià Blanco
- IrsiCaixa, 08916 Badalona, Spain
- Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Germans Trias I Pujol Research Institute (IGTP), 08916 Badalona, Spain
- CIBERINFEC, ISCIII, 28029 Madrid, Spain
| |
Collapse
|
2
|
Wang W, Wang S, Meng X, Zhao Y, Li N, Wang T, Feng N, Yan F, Xia X. A virus-like particle candidate vaccine based on CRISPR/Cas9 gene editing technology elicits broad-spectrum protection against SARS-CoV-2. Antiviral Res 2024; 225:105854. [PMID: 38447647 DOI: 10.1016/j.antiviral.2024.105854] [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: 10/29/2023] [Revised: 02/08/2024] [Accepted: 03/02/2024] [Indexed: 03/08/2024]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with frequent mutations has seriously damaged the effectiveness of the 2019 coronavirus disease (COVID-19) vaccine. There is an urgent need to develop a broad-spectrum vaccine while elucidating the underlying immune mechanisms. Here, we developed a SARS-CoV-2 virus-like particles (VLPs) vaccine based on the Canarypox-virus vector (ALVAC-VLPs) using CRISPR/Cas9. Immunization with ALVAC-VLPs showed the effectively induce SARS-CoV-2 specific T and B cell responses to resist the lethal challenge of mouse adaptive strains. Notably, ALVAC-VLPs conferred protection in golden hamsters against SARS-CoV-2 Wuhan-Hu-1 (wild-type, WT) and variants (Beta, Delta, Omicron BA.1, and BA.2), as evidenced by the prevention of weight loss, reduction in lung and turbinate tissue damage, and decreased viral load. Further investigation into the mechanism of immune response induced by ALVAC-VLPs revealed that toll-like receptor 4 (TLR4) mediates the recruitment of dendritic cells (DCs) to secondary lymphoid organs, thereby initiating follicle assisted T (Tfh) cell differentiation, the proliferation of germinal center (GC) B cells and plasma cell production. These findings demonstrate the immunogenicity and efficacy of the safe ALVAC-VLPs vaccine against SARS-CoV-2 and provide valuable insight into the development of COVID-19 vaccine strategies.
Collapse
Affiliation(s)
- Weiqi Wang
- College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, China; Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Xianyong Meng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China; College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, Jilin, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Nan Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China.
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China.
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, China; Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China.
| |
Collapse
|
3
|
Weidinger AK, Bergmann M, König M, Zablotski Y, Hartmann K. Anti-rabies humoral immune response in cats after concurrent vs separate vaccination against rabies and feline leukaemia virus using canarypox-vectored vaccines. J Feline Med Surg 2024; 26:1098612X231218643. [PMID: 38358295 PMCID: PMC10911302 DOI: 10.1177/1098612x231218643] [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] [Indexed: 02/16/2024]
Abstract
OBJECTIVES Some expert groups recommend that cats should be vaccinated with non-adjuvanted feline leukaemia virus (FeLV) and rabies vector vaccines, which, in the European Union, are currently not licensed for concurrent use and have to be administered at least 14 days apart (different from the USA) and thus at separate visits, which is associated with more stress for cats and owners. The aim of this study was to assess the anti-rabies antibody response in cats after vaccination against rabies and FeLV at concurrent vs separate (4 weeks apart) visits using two canarypox-vectored vaccines (Purevax Rabies and Purevax FeLV; Boehringer Ingelheim) and to evaluate the occurrence of vaccine-associated adverse events (VAAEs). METHODS Healthy FeLV antigen-negative client-owned kittens (n = 106) were prospectively included in this randomised study. All kittens received primary vaccinations against rabies (week 0) and FeLV (weeks 4 and 8). After 1 year, the study group (n = 52) received booster vaccinations against rabies and FeLV concurrently at the same visit (weeks 50-52). The control group (n = 54) received booster vaccinations against rabies (weeks 50-52) and FeLV (weeks 54-56) separately. Anti-rabies virus antibodies (anti-RAV Ab) were determined by fluorescent antibody virus neutralisation assay at weeks 4, 50-52 and 54-56, and compared between both groups using a Mann-Whitney U-test. RESULTS Four weeks after the first rabies vaccination, 87/106 (82.1%) kittens had a titre ⩾0.5 IU/ml and 19/106 (17.9%) had a titre <0.5 IU/ml. Four weeks after the 1-year rabies booster, all cats had adequate anti-RAV Ab according to the World Organisation for Animal Health (⩾0.5 IU/ml), and the titres of the study group (median = 14.30 IU/ml) and the control group (median = 21.39 IU/ml) did not differ significantly (P = 0.141). VAAEs were observed in 7/106 (6.6%) cats. CONCLUSIONS AND RELEVANCE Concurrent administration of Purevax FeLV and Purevax Rabies vector vaccines at the 1-year booster does not interfere with the development of anti-RAV Ab or cause more adverse effects and thus represents a better option than separate vaccination visits for cats and owners.
Collapse
Affiliation(s)
- Anna-Karina Weidinger
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Michèle Bergmann
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Matthias König
- Institute of Virology, Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Hessen, Germany
| | - Yury Zablotski
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Katrin Hartmann
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
4
|
Piewbang C, Wardhani SW, Siripoonsub J, Sirivisoot S, Rungsipipat A, Techangamsuwan S. Domestic cat hepadnavirus detection in blood and tissue samples of cats with lymphoma. Vet Q 2023; 43:1-10. [PMID: 37768269 PMCID: PMC10563604 DOI: 10.1080/01652176.2023.2265172] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/26/2023] [Indexed: 09/29/2023] Open
Abstract
Domestic cat hepadnavirus (DCH), a relative hepatitis B virus (HBV) in human, has been recently identified in cats; however, association of DCH infection with lymphoma in cats is not investigated. To determine the association between DCH infection and feline lymphoma, seven hundred and seventeen cats included 131 cats with lymphoma (68 blood and 63 tumor samples) and 586 (526 blood and 60 lymph node samples) cats without lymphoma. DCH DNA was investigated in blood and formalin-fixed paraffin-embedded (FFPE) tissues by quantitative polymerase chain reaction (qPCR). The FFPE lymphoma tissues were immunohistochemically subtyped, and the localization of DCH in lymphoma sections was investigated using in situ hybridization (ISH). Feline retroviral infection was investigated in the DCH-positive cases. DCH DNA was detected in 16.18% (11/68) (p = 0.002; odds ratio [OR], 5.15; 95% confidence interval [CI], 2.33-11.36) of blood and 9.52% (6/63) (p = 0.028; OR, 13.68; 95% CI, 0.75-248.36) of neoplastic samples obtained from lymphoma cats, whereas only 3.61% (19/526) of blood obtained from non-lymphoma cats was positive for DCH detection. Within the DCH-positive lymphoma, in 3/6 cats, feline leukemia virus was co-detected, and in 6/6 were B-cell lymphoma (p > 0.9; OR, 1.93; 95% CI, 0.09-37.89) and were multicentric form (p = 0.008; OR, 1.327; 95% CI, 0.06-31.18). DCH was found in the CD79-positive pleomorphic cells. Cats with lymphoma were more likely to be positive for DCH than cats without lymphoma, and infection associated with lymphoma development needs further investigations.
Collapse
Affiliation(s)
- Chutchai Piewbang
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Animal Virome and Diagnostic Development Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sabrina Wahyu Wardhani
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Animal Virome and Diagnostic Development Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Jedsada Siripoonsub
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Companion Animal Cancer, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sirintra Sirivisoot
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Companion Animal Cancer, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Anudep Rungsipipat
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Companion Animal Cancer, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Somporn Techangamsuwan
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Animal Virome and Diagnostic Development Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
5
|
Skandorff I, Ragonnaud E, Gille J, Andersson AM, Schrödel S, Duvnjak L, Turner L, Thirion C, Wagner R, Holst PJ. Human Ad19a/64 HERV-W Vaccines Uncover Immunosuppression Domain-Dependent T-Cell Response Differences in Inbred Mice. Int J Mol Sci 2023; 24:9972. [PMID: 37373123 DOI: 10.3390/ijms24129972] [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: 05/17/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Expression of human endogenous retrovirus type W (HERV-W) has been linked to cancer, making HERV-W antigens potential targets for therapeutic cancer vaccines. In a previous study, we effectively treated established tumours in mice by using adenoviral-vectored vaccines targeting the murine endogenous retrovirus envelope and group-specific antigen (Gag) of melanoma-associated retrovirus (MelARV) in combination with anti-PD-1. To break the immunological tolerance to MelARV, we mutated the immunosuppressive domain (ISD) of the MelARV envelope. However, reports on the immunogenicity of the HERV-W envelope, Syncytin-1, and its ISD are conflicting. To identify the most effective HERV-W cancer vaccine candidate, we evaluated the immunogenicity of vaccines encoding either the wild-type or mutated HERV-W envelope ISD in vitro and in vivo. Here, we show that the wild-type HERV-W vaccine generated higher activation of murine antigen-presenting cells and higher specific T-cell responses than the ISD-mutated counterpart. We also found that the wild-type HERV-W vaccine was sufficient to increase the probability of survival in mice subjected to HERV-W envelope-expressing tumours compared to a control vaccine. These findings provide the foundation for developing a therapeutic cancer vaccine targeting HERV-W-positive cancers in humans.
Collapse
Affiliation(s)
- Isabella Skandorff
- Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- InProTher, COBIS, Ole Maaloesvej 3, 2200 Copenhagen, Denmark
| | - Emeline Ragonnaud
- InProTher, COBIS, Ole Maaloesvej 3, 2200 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Jasmin Gille
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology, University of Regensburg, 93053 Regensburg, Germany
| | | | - Silke Schrödel
- Sirion Biotech GmbH, Am Haag 6, 82166 Graefelfing, Germany
| | - Lara Duvnjak
- Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- InProTher, COBIS, Ole Maaloesvej 3, 2200 Copenhagen, Denmark
| | - Louise Turner
- Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | | | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology, University of Regensburg, 93053 Regensburg, Germany
| | - Peter Johannes Holst
- InProTher, COBIS, Ole Maaloesvej 3, 2200 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
6
|
Ortiz R, Barajas A, Pons-Grífols A, Trinité B, Tarrés-Freixas F, Rovirosa C, Urrea V, Barreiro A, Gonzalez-Tendero A, Cardona M, Ferrer L, Clotet B, Carrillo J, Aguilar-Gurrieri C, Blanco J. Exploring FeLV-Gag-Based VLPs as a New Vaccine Platform-Analysis of Production and Immunogenicity. Int J Mol Sci 2023; 24:ijms24109025. [PMID: 37240371 DOI: 10.3390/ijms24109025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/11/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Feline leukemia virus (FeLV) is one of the most prevalent infectious diseases in domestic cats. Although different commercial vaccines are available, none of them provides full protection. Thus, efforts to design a more efficient vaccine are needed. Our group has successfully engineered HIV-1 Gag-based VLPs that induce a potent and functional immune response against the HIV-1 transmembrane protein gp41. Here, we propose to use this concept to generate FeLV-Gag-based VLPs as a novel vaccine strategy against this retrovirus. By analogy to our HIV-1 platform, a fragment of the FeLV transmembrane p15E protein was exposed on FeLV-Gag-based VLPs. After optimization of Gag sequences, the immunogenicity of the selected candidates was evaluated in C57BL/6 and BALB/c mice, showing strong cellular and humoral responses to Gag but failing to generate anti-p15E antibodies. Altogether, this study not only tests the versatility of the enveloped VLP-based vaccine platform but also sheds light on FeLV vaccine research.
Collapse
Affiliation(s)
- Raquel Ortiz
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Ana Barajas
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Medicine Department, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
| | - Anna Pons-Grífols
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Benjamin Trinité
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
| | | | - Carla Rovirosa
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
| | - Victor Urrea
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
| | | | | | | | | | - Bonaventura Clotet
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Medicine Department, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Infectious Diseases Department, Germans Trias I Pujol Hospital, 08916 Badalona, Spain
| | - Jorge Carrillo
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- CIBERINFEC, ISCIII, 28029 Madrid, Spain
| | | | - Julià Blanco
- AIDS Research Institute, IrsiCaixa, Campus Can Ruti, 08916 Badalona, Spain
- Doctorate School, Microbiology Department, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Doctorate School, Medicine Department, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- CIBERINFEC, ISCIII, 28029 Madrid, Spain
- Germans Trias I Pujol Research Institute (IGTP), Campus Can Ruti, 08916 Badalona, Spain
| |
Collapse
|
7
|
A recombinant measles virus vaccine strongly reduces SHIV viremia and virus reservoir establishment in macaques. NPJ Vaccines 2021; 6:123. [PMID: 34686669 PMCID: PMC8536681 DOI: 10.1038/s41541-021-00385-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/20/2021] [Indexed: 11/22/2022] Open
Abstract
Replicative vectors derived from live-attenuated measles virus (MV) carrying additional non-measles vaccine antigens have long demonstrated safety and immunogenicity in humans despite pre-existing immunity to measles. Here, we report the vaccination of cynomolgus macaques with MV replicative vectors expressing simian-human immunodeficiency virus Gag, Env, and Nef antigens (MV-SHIV Wt) either wild type or mutated in the immunosuppressive (IS) domains of Nef and Env antigens (MV-SHIV Mt). We found that the inactivation of Nef and Env IS domains by targeted mutations led to the induction of significantly enhanced post-prime cellular immune responses. After repeated challenges with low doses of SHIV-SF162p3, vaccinees were protected against high viremia, resulting in a 2-Log reduction in peak viremia, accelerated viral clearance, and a decrease -even complete protection for nearly half of the monkeys- in reservoir cell infection. This study demonstrates the potential of a replicative viral vector derived from the safe and widely used measles vaccine in the development of a future human vaccine against HIV-1.
Collapse
|
8
|
Lokossou AG, Toudic C, Nguyen PT, Elisseeff X, Vargas A, Rassart É, Lafond J, Leduc L, Bourgault S, Gilbert C, Scorza T, Tolosa J, Barbeau B. Endogenous retrovirus-encoded Syncytin-2 contributes to exosome-mediated immunosuppression of T cells†. Biol Reprod 2021; 102:185-198. [PMID: 31318021 DOI: 10.1093/biolre/ioz124] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/21/2018] [Accepted: 07/11/2019] [Indexed: 12/24/2022] Open
Abstract
Modulation of the activation status of immune cell populations during pregnancy depends on placental villous cytotrophoblast (VCT) cells and the syncytiotrophoblast (STB). Failure in the establishment of this immunoregulatory function leads to pregnancy complications. Our laboratory has been studying Syncytin-2 (Syn-2), an endogenous retroviral protein expressed in placenta and on the surface of placental exosomes. This protein plays an important role not only in STB formation through its fusogenic properties, but also through its immunosuppressive domain (ISD). Considering that Syn-2 expression is importantly reduced in preeclamptic placentas, we were interested in addressing its possible immunoregulatory effects on T cells. Activated Jurkat T cells and peripheral blood mononuclear cells (PBMCs) were treated with monomeric or dimerized version of a control or a Syn-2 ISD peptide. Change in phosphorylation levels of ERK1/2 MAP kinases was selectively noted in Jurkat cells treated with the dimerized ISD peptide. Upon incubation with the dimerized Syn-2 ISD peptide, significant reduction in Th1 cytokine production was further demonstrated by ELISA and Human Th1/Th2 Panel Multi-Analyte Flow Assay. To determine if exosome-associated Syn-2 could also be immunosuppressive placental exosomes were incubated with activated Jurkat and PBMCs. Quantification of Th1 cytokines in the supernatants revealed severe reduction in T cell activation. Interestingly, exosomes from Syn-2-silenced VCT incubated with PBMCs were less suppressive when compared with exosome derived from VCT transfected with control small interfering RNA (siRNA). Our results suggest that Syn-2 is an important immune regulator both locally and systemically, via its association with placental exosomes.
Collapse
Affiliation(s)
- Adjimon G Lokossou
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, Quebec, Canada.,Centre de recherche BioMed, Montreal, Quebec, Canada
| | - Caroline Toudic
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, Quebec, Canada.,Centre de recherche BioMed, Montreal, Quebec, Canada
| | - Phuong Trang Nguyen
- Centre de recherche BioMed, Montreal, Quebec, Canada.,Université du Québec à Montréal, Department of Chemistry, Montreal, Quebec, Canada
| | - Xavier Elisseeff
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, Quebec, Canada.,Centre de recherche BioMed, Montreal, Quebec, Canada
| | - Amandine Vargas
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Éric Rassart
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, Quebec, Canada.,Centre de recherche BioMed, Montreal, Quebec, Canada
| | - Julie Lafond
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, Quebec, Canada.,Centre de recherche BioMed, Montreal, Quebec, Canada
| | - Line Leduc
- CHU Ste-Justine, Montreal, Quebec, Canada
| | - Steve Bourgault
- Centre de recherche BioMed, Montreal, Quebec, Canada.,Université du Québec à Montréal, Department of Chemistry, Montreal, Quebec, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Montreal, Quebec, Canada
| | | | - Tatiana Scorza
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, Quebec, Canada.,Centre de recherche BioMed, Montreal, Quebec, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Montreal, Quebec, Canada
| | - Jorge Tolosa
- Mothers and Babies Research Centre and Hunter Medical Research Institute, The University of Newcastle, Newcastle, Australia
| | - Benoit Barbeau
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, Quebec, Canada.,Centre de recherche BioMed, Montreal, Quebec, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Aida V, Pliasas VC, Neasham PJ, North JF, McWhorter KL, Glover SR, Kyriakis CS. Novel Vaccine Technologies in Veterinary Medicine: A Herald to Human Medicine Vaccines. Front Vet Sci 2021; 8:654289. [PMID: 33937377 PMCID: PMC8083957 DOI: 10.3389/fvets.2021.654289] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/17/2021] [Indexed: 01/10/2023] Open
Abstract
The success of inactivated and live-attenuated vaccines has enhanced livestock productivity, promoted food security, and attenuated the morbidity and mortality of several human, animal, and zoonotic diseases. However, these traditional vaccine technologies are not without fault. The efficacy of inactivated vaccines can be suboptimal with particular pathogens and safety concerns arise with live-attenuated vaccines. Additionally, the rate of emerging infectious diseases continues to increase and with that the need to quickly deploy new vaccines. Unfortunately, first generation vaccines are not conducive to such urgencies. Within the last three decades, veterinary medicine has spearheaded the advancement in novel vaccine development to circumvent several of the flaws associated with classical vaccines. These third generation vaccines, including DNA, RNA and recombinant viral-vector vaccines, induce both humoral and cellular immune response, are economically manufactured, safe to use, and can be utilized to differentiate infected from vaccinated animals. The present article offers a review of commercially available novel vaccine technologies currently utilized in companion animal, food animal, and wildlife disease control.
Collapse
Affiliation(s)
- Virginia Aida
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - Vasilis C. Pliasas
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - Peter J. Neasham
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - J. Fletcher North
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - Kirklin L. McWhorter
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Department of Chemistry, Emory University, Atlanta, GA, United States
| | - Sheniqua R. Glover
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - Constantinos S. Kyriakis
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| |
Collapse
|
10
|
Skirmuntt EC, Katzourakis A. The evolution of endogenous retroviral envelope genes in bats and their potential contribution to host biology. Virus Res 2019; 270:197645. [PMID: 31271763 DOI: 10.1016/j.virusres.2019.197645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022]
Abstract
Bats are the primary reservoirs and carriers of a wide range of viruses of unknown infectivity and pathogenic potential. Some of those if transmitted to other species can cause enormous economic losses in agriculture, and mortality in humans. Bats can be persistently infected with viruses while not showing any symptoms of disease, despite having high virus titre levels in their tissues and shedding virions for months or years after primary infection. It has been suggested that the lack of symptoms of viral infections and low mortality rate in bats might be due to immune adaptations that result from their long-term co-evolution with viruses. In this study, we screened all publicly available bat genomes from six bat families within which we have identified several envelope sequences of retroviral origin (gammaretroviruses). We analysed the identified sequences with Bayesian methods and maximum-likelihood inference to generate a phylogenetic tree with additional reference sequences of known endogenous and exogenous viral envelope genes. We also identified groups of orthologous viral envelopes and analysed them to determine if any of them might be an EVE (endogenous virus element) with an EDI (EVE- derived immunity) function or a candidate for a bat syncytin gene, which is an endogenized viral envelope, mostly known from its function in placentation in animals. Our study shows that bat genomes contain a substantial number of large, intact envelopes with open reading frames, which were found clustering closely on a phylogenetic tree reconstruction with syncytin sequences of other species. That might indicate that such sequences are good candidates for further bat-syncytin/EDI search.
Collapse
Affiliation(s)
- Emilia Cecylia Skirmuntt
- Department of Zoology, University of Oxford, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK
| | - Aris Katzourakis
- Department of Zoology, University of Oxford, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK.
| |
Collapse
|
11
|
Ivanusic D, Pietsch H, König J, Denner J. Absence of IL-10 production by human PBMCs co-cultivated with human cells expressing or secreting retroviral immunosuppressive domains. PLoS One 2018; 13:e0200570. [PMID: 30001404 PMCID: PMC6042780 DOI: 10.1371/journal.pone.0200570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/28/2018] [Indexed: 11/29/2022] Open
Abstract
Immunosuppression by retroviruses including the human immunodeficiency virus—1 (HIV-1) is well known, however the mechanisms how retroviruses induce this immunosuppression is not fully investigated. It was shown that non-infectious retroviral particles as well as retroviral or recombinant retroviral transmembrane envelope (TM) proteins demonstrated immunosuppressive properties. The same was shown for peptides corresponding to a highly conserved domain in the TM protein. This domain is called immunosuppressive (ISU) domain and it induces modulation of the cytokine release of peripheral blood mononuclear cells (PBMCs) from healthy donors. In addition, it changes the gene expression of these cells. Common indications for the immunosuppressive activity were tumour growth in vivo and interleukin—10 (IL-10) release from human PBMCs in vitro. Single mutations in the ISU domain abrogated the immunosuppressive activity. In order to develop a new model system for the expression of the ISU domain and presentation to PBMCs which is not prone to possible endotoxin contaminations, two expression systems were developed. In the first system, designated pOUT, retroviral proteins containing the ISU domain were expressed and released into the cell culture medium, and in the second system, tANCHOR, the ISU domain was presented by a tetraspanin-anchored sequence on the cell surface of human cells. Both systems were exploited to express the wild-type (wt) ISU domains of HIV-1, of the porcine endogenous retrovirus (PERV) and of the murine leukaemia virus (MuLV) as well as to express mutants (mut) of these ISU domains. PERV is of special interest in the context of virus safety of xenotransplantation using pig organs. Expression of the TM proteins was demonstrated by confocal laser scanning microscopy, ELISA and Western blot analyses using specific antibodies. However, when cells expressing and releasing the ISU were co-incubated with human PBMCs, no increased production of IL-10 was observed when compared with the mutants. Similar results were obtained when the released TM proteins were concentrated by immunoprecipitation and added to PBMCs. We suggest that the absence of IL-10 induction can be explained by a low amount of protein, by the lack of a biologically active conformation or the absence of additional factors.
Collapse
|
12
|
Kawasaki J, Nishigaki K. Tracking the Continuous Evolutionary Processes of an Endogenous Retrovirus of the Domestic Cat: ERV-DC. Viruses 2018; 10:v10040179. [PMID: 29642384 PMCID: PMC5923473 DOI: 10.3390/v10040179] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/02/2018] [Accepted: 04/05/2018] [Indexed: 12/23/2022] Open
Abstract
An endogenous retrovirus (ERV) is a remnant of an ancient retroviral infection in the host genome. Although most ERVs have lost their viral productivity, a few ERVs retain their replication capacity. In addition, partially inactivated ERVs can present a potential risk to the host via their encoded virulence factors or the generation of novel viruses by viral recombination. ERVs can also eventually acquire a biological function, and this ability has been a driving force of host evolution. Therefore, the presence of an ERV can be harmful or beneficial to the host. Various reports about paleovirology have revealed each event in ERV evolution, but the continuous processes of ERV evolution over millions of years are mainly unknown. A unique ERV family, ERV-DC, is present in the domestic cat (Felis silvestriscatus) genome. ERV-DC proviruses are phylogenetically classified into three genotypes, and the specific characteristics of each genotype have been clarified: their capacity to produce infectious viruses; their recombination with other retroviruses, such as feline leukemia virus or RD-114; and their biological functions as host antiviral factors. In this review, we describe ERV-DC-related phenomena and discuss the continuous changes in the evolution of this ERV in the domestic cat.
Collapse
Affiliation(s)
- Junna Kawasaki
- Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan.
| | - Kazuo Nishigaki
- Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan.
| |
Collapse
|
13
|
Wetzel D, Rolf T, Suckow M, Kranz A, Barbian A, Chan JA, Leitsch J, Weniger M, Jenzelewski V, Kouskousis B, Palmer C, Beeson JG, Schembecker G, Merz J, Piontek M. Establishment of a yeast-based VLP platform for antigen presentation. Microb Cell Fact 2018; 17:17. [PMID: 29402276 PMCID: PMC5798182 DOI: 10.1186/s12934-018-0868-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 01/27/2018] [Indexed: 12/26/2022] Open
Abstract
Background Chimeric virus-like particles (VLP) allow the display of foreign antigens on their surface and have proved valuable in the development of safe subunit vaccines or drug delivery. However, finding an inexpensive production system and a VLP scaffold that allows stable incorporation of diverse, large foreign antigens are major challenges in this field. Results In this study, a versatile and cost-effective platform for chimeric VLP development was established. The membrane integral small surface protein (dS) of the duck hepatitis B virus was chosen as VLP scaffold and the industrially applied and safe yeast Hansenula polymorpha (syn. Pichia angusta, Ogataea polymorpha) as the heterologous expression host. Eight different, large molecular weight antigens of up to 412 amino acids derived from four animal-infecting viruses were genetically fused to the dS and recombinant production strains were isolated. In all cases, the fusion protein was well expressed and upon co-production with dS, chimeric VLP containing both proteins could be generated. Purification was accomplished by a downstream process adapted from the production of a recombinant hepatitis B VLP vaccine. Chimeric VLP were up to 95% pure on protein level and contained up to 33% fusion protein. Immunological data supported surface exposure of the foreign antigens on the native VLP. Approximately 40 mg of chimeric VLP per 100 g dry cell weight could be isolated. This is highly comparable to values reported for the optimized production of human hepatitis B VLP. Purified chimeric VLP were shown to be essentially stable for 6 months at 4 °C. Conclusions The dS-based VLP scaffold tolerates the incorporation of a variety of large molecular weight foreign protein sequences. It is applicable for the display of highly immunogenic antigens originating from a variety of pathogens. The yeast-based production system allows cost-effective production that is not limited to small-scale fundamental research. Thus, the dS-based VLP platform is highly efficient for antigen presentation and should be considered in the development of future vaccines.
Collapse
Affiliation(s)
- David Wetzel
- ARTES Biotechnology GmbH, Elisabeth-Selbert-Straße 9, 40764, Langenfeld, Germany. .,Laboratory of Plant and Process Design, Technical University of Dortmund, Emil-Figge-Straße 70, 44227, Dortmund, Germany.
| | - Theresa Rolf
- ARTES Biotechnology GmbH, Elisabeth-Selbert-Straße 9, 40764, Langenfeld, Germany
| | - Manfred Suckow
- ARTES Biotechnology GmbH, Elisabeth-Selbert-Straße 9, 40764, Langenfeld, Germany
| | - Andreas Kranz
- ARTES Biotechnology GmbH, Elisabeth-Selbert-Straße 9, 40764, Langenfeld, Germany
| | - Andreas Barbian
- Institute for Anatomy I, Düsseldorf University Hospital, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Jo-Anne Chan
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Joachim Leitsch
- ARTES Biotechnology GmbH, Elisabeth-Selbert-Straße 9, 40764, Langenfeld, Germany
| | - Michael Weniger
- ARTES Biotechnology GmbH, Elisabeth-Selbert-Straße 9, 40764, Langenfeld, Germany
| | - Volker Jenzelewski
- ARTES Biotechnology GmbH, Elisabeth-Selbert-Straße 9, 40764, Langenfeld, Germany
| | - Betty Kouskousis
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Catherine Palmer
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - James G Beeson
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Gerhard Schembecker
- Laboratory of Plant and Process Design, Technical University of Dortmund, Emil-Figge-Straße 70, 44227, Dortmund, Germany
| | - Juliane Merz
- Laboratory of Plant and Process Design, Technical University of Dortmund, Emil-Figge-Straße 70, 44227, Dortmund, Germany
| | - Michael Piontek
- ARTES Biotechnology GmbH, Elisabeth-Selbert-Straße 9, 40764, Langenfeld, Germany
| |
Collapse
|
14
|
Joglekar AV, Sandoval S. Pseudotyped Lentiviral Vectors: One Vector, Many Guises. Hum Gene Ther Methods 2017; 28:291-301. [DOI: 10.1089/hgtb.2017.084] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Alok V. Joglekar
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| | - Salemiz Sandoval
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| |
Collapse
|
15
|
Grosenbaugh DA, Frances-Duvert V, Abedi S, Feilmeier B, Ru H, Poulet H. Efficacy of a nonadjuvanted recombinant FeLV vaccine and two inactivated FeLV vaccines when subject to consistent virulent FeLV challenge conditions. Biologicals 2017; 49:76-80. [PMID: 28734742 DOI: 10.1016/j.biologicals.2016.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/19/2016] [Accepted: 10/17/2016] [Indexed: 11/25/2022] Open
Abstract
The purpose of this study was to compare the efficacy of three FeLV vaccines, under identical conditions in a laboratory challenge model that closely mimics natural infection. Four groups of cats (n = 20 per group) were administered two doses of vaccine, 21 days apart, starting at 9-10 weeks of age (Purevax® FeLV, Versifel® FeLV, Nobivac® feline 2-FeLV, and a placebo). Cats were challenged 3 weeks later with a virulent, heterologous FeLV isolate. FeLV antigenemia was determined at weekly intervals from 3 to 15 weeks postchallenge. Circulating proviral DNA was determined on terminal PBMC samples. Following challenge, persistent antigenemia developed in 15 (75%) placebo-vaccinated cats, 3 (15%) cats in the Versifel FeLV vaccinated group, and 1 cat (5%) each in the Purevax FeLV and the Nobivac FeLV vaccinated groups. The prevented fractions for three vaccine groups were 93%, 93%, and 80% respectively. The adjusted p-values for all vaccine group comparisons fail to approach statistical significance. There was excellent agreement between proviral FeLV DNA in circulating PBMCs and persistent antigenemia. It is shown that when cats are managed under the same conditions during a virulent challenge, via the normal route of infection, the tested vaccines all show a comparable degree of protection.
Collapse
Affiliation(s)
| | - Valérie Frances-Duvert
- Merial, Research and Development, Lyon Gerland Laboratories, 254 rue Marcel Mérieux, 69007, Lyon, France
| | - Shabnam Abedi
- Merial, Research and Development, 1730 Olympic Dr., Athens, GA 30601, USA
| | - Bradley Feilmeier
- Merial, Research and Development, 1730 Olympic Dr., Athens, GA 30601, USA
| | - Hongyu Ru
- Merial, Research and Development, 1730 Olympic Dr., Athens, GA 30601, USA
| | - Hervé Poulet
- Merial, Research and Development, Lyon Gerland Laboratories, 254 rue Marcel Mérieux, 69007, Lyon, France
| |
Collapse
|
16
|
Lemaître C, Tsang J, Bireau C, Heidmann T, Dewannieux M. A human endogenous retrovirus-derived gene that can contribute to oncogenesis by activating the ERK pathway and inducing migration and invasion. PLoS Pathog 2017. [PMID: 28651004 PMCID: PMC5501692 DOI: 10.1371/journal.ppat.1006451] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Endogenous retroviruses are cellular genes of retroviral origin captured by their host during the course of evolution and represent around 8% of the human genome. Although most are defective and transcriptionally silenced, some are still able to generate retroviral-like particles and proteins. Among these, the HERV-K(HML2) family is remarkable since its members have amplified relatively recently and many of them still have full length coding genes. Furthermore, they are induced in cancers, especially in melanoma, breast cancer and germ cell tumours, where viral particles, as well as the envelope protein (Env), can be detected. Here we show that HERV-K(HML2) Env per se has oncogenic properties. Its expression in a non-tumourigenic human breast epithelial cell line induces epithelial to mesenchymal transition (EMT), often associated with tumour aggressiveness and metastasis. In our model, this is typified by key modifications in a set of molecular markers, changes in cell morphology and enhanced cell motility. Remarkably, microarrays performed in 293T cells reveal that HERV-K(HML2) Env is a strong inducer of several transcription factors, namely ETV4, ETV5 and EGR1, which are downstream effectors of the MAPK ERK1/2 and are associated with cellular transformation. We demonstrate that HERV-K(HML2) Env effectively activates the ERK1/2 pathway in our experimental setting and that this activation depends on the Env cytoplasmic tail. In addition, this phenomenon is very specific, being absent with every other retroviral Env tested, except for Jaagsiekte Sheep Retrovirus (JSRV) Env, which is already known to have transforming properties in vivo. Though HERV-K Env is not directly transforming by itself, the newly discovered properties of this protein may contribute to oncogenesis. Nearly half the DNA of mammals consists of reitarated, selfish elements that can move and amplify within the genome. With time, some of these elements are recruited by the host and the proteins they encode are used to fulfill physiological functions, whereas other elements have conserved some of their pathological properties and contribute to the development of diseases. The human HERV-K(HML2) elements originated from an ancestral infection of the primate germline by an infectious retrovirus that has been maintained and amplified in the human lineage. It is associated with several pathologies in modern humans, in particular cancer of the breast, germline and skin. We show that the HERV-K(HML2) envelope protein is able to activate a major cellular signalling pathway often involved in human cancers, and that its expression promotes a series of cellular changes that are characteristic of cancer development. Altogether, this study indicates that the expression of HERV-K(HML2) elements is not only a marker of cancer, but can also directly participate to tumourigenesis via the newly discovered oncogenic properties carried by the envelope protein.
Collapse
Affiliation(s)
- Cécile Lemaître
- CNRS, UMR 9196, Institut Gustave Roussy, Villejuif, France
- Université Paris-Sud, Orsay, France
- Université Paris Denis Diderot, Sorbonne Paris-Cité, Paris, France
| | - Jhen Tsang
- CNRS, UMR 9196, Institut Gustave Roussy, Villejuif, France
- Université Paris-Sud, Orsay, France
| | - Caroline Bireau
- CNRS, UMR 9196, Institut Gustave Roussy, Villejuif, France
- Université Paris-Sud, Orsay, France
| | - Thierry Heidmann
- CNRS, UMR 9196, Institut Gustave Roussy, Villejuif, France
- Université Paris-Sud, Orsay, France
- * E-mail: (MD); (TH)
| | - Marie Dewannieux
- CNRS, UMR 9196, Institut Gustave Roussy, Villejuif, France
- Université Paris-Sud, Orsay, France
- * E-mail: (MD); (TH)
| |
Collapse
|
17
|
Interference of retroviral envelope with vaccine-induced CD8 + T cell responses is relieved by co-administration of cytokine-encoding vectors. Retrovirology 2017; 14:28. [PMID: 28449719 PMCID: PMC5408827 DOI: 10.1186/s12977-017-0352-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/20/2017] [Indexed: 11/13/2022] Open
Abstract
Background Retroviral envelope (Env) proteins are known to exhibit immunosuppressive properties, which become apparent not only in retroviral infections, but also in gene-based immunizations using retroviral immunogens, where envelope interferes with the induction of CD8+ T cell responses towards another, simultaneously or subsequently delivered, immunogen. Results In the Friend retrovirus mouse model, immunization with a plasmid encoding the Friend murine leukemia virus (F-MuLV) Leader-Gag protein resulted in induction of a strong GagL85–93-specific CD8+ T cell response, while the response was completely abrogated by co-immunization with an F-MuLV Env-encoding plasmid. In order to overcome this interference of retroviral envelope, we employed plasmids encoding the cytokines interleukin (IL) 1β, IL2, IL12, IL15, IL21, IL28A or granulocyte–macrophage colony-stimulating factor (GM-CSF) as genetic adjuvants. Co-application of plasmids encoding IL2, IL12, IL21, IL28A and especially GM-CSF rescued the induction of GagL85–93-specific CD8+ T cells in mice vaccinated with FV Leader-Gag and Env. Mice that were immunized with plasmids encoding Leader-Gag and Env and the cytokines IL1β, IL12, IL15, IL28A or GM-CSF, but not Leader-Gag and Env without any cytokine, showed significantly reduced viral loads upon a high-dose Friend virus challenge infection. Conclusions Our data demonstrate the potency of cytokine-encoding vectors as adjuvants and immune modulators in composite vaccines for anti-retroviral immunization. Electronic supplementary material The online version of this article (doi:10.1186/s12977-017-0352-7) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Waugh CA, Hanger J, Loader J, King A, Hobbs M, Johnson R, Timms P. Infection with koala retrovirus subgroup B (KoRV-B), but not KoRV-A, is associated with chlamydial disease in free-ranging koalas (Phascolarctos cinereus). Sci Rep 2017; 7:134. [PMID: 28273935 PMCID: PMC5427818 DOI: 10.1038/s41598-017-00137-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/09/2017] [Indexed: 02/02/2023] Open
Abstract
The virulence of chlamydial infection in wild koalas is highly variable between individuals. Some koalas can be infected (PCR positive) with Chlamydia for long periods but remain asymptomatic, whereas others develop clinical disease. Chlamydia in the koala has traditionally been studied without regard to coinfection with other pathogens, although koalas are usually subject to infection with koala retrovirus (KoRV). Retroviruses can be immunosuppressive, and there is evidence of an immunosuppressive effect of KoRV in vitro. Originally thought to be a single endogenous strain, a new, potentially more virulent exogenous variant (KoRV-B) was recently reported. We hypothesized that KoRV-B might significantly alter chlamydial disease outcomes in koalas, presumably via immunosuppression. By studying sub-groups of Chlamydia and KoRV infected koalas in the wild, we found that neither total KoRV load (either viraemia or proviral copies per genome), nor chlamydial infection level or strain type, was significantly associated with chlamydial disease risk. However, PCR positivity with KoRV-B was significantly associated with chlamydial disease in koalas (p = 0.02961). This represents an example of a recently evolved virus variant that may be predisposing its host (the koala) to overt clinical disease when co-infected with an otherwise asymptomatic bacterial pathogen (Chlamydia).
Collapse
Affiliation(s)
- Courtney A Waugh
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, 4558, Queensland, Australia.,Department of Biology, Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Jonathan Hanger
- Endeavour Veterinary Ecology, 1695 Pumicestone Rd, Toorbul, 4510, Queensland, Australia
| | - Joanne Loader
- Endeavour Veterinary Ecology, 1695 Pumicestone Rd, Toorbul, 4510, Queensland, Australia
| | - Andrew King
- Australian Museum Research Institute, Australian Museum, 1 William Street, Sydney, NSW, 2010, Australia
| | - Matthew Hobbs
- Australian Museum Research Institute, Australian Museum, 1 William Street, Sydney, NSW, 2010, Australia
| | - Rebecca Johnson
- Australian Museum Research Institute, Australian Museum, 1 William Street, Sydney, NSW, 2010, Australia
| | - Peter Timms
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, 4558, Queensland, Australia.
| |
Collapse
|
19
|
Jenkins B, Eksmond U, Young G, Kassiotis G. Antigenicity of peptides comprising the immunosuppressive domain of the retroviral envelope glycoprotein. Wellcome Open Res 2016. [DOI: 10.12688/wellcomeopenres.10269.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To achieve persistent infection of the host, viruses often subvert or suppress host immunity through mechanisms that are not entirely understood. The envelope glycoprotein of several retroviruses is thought to possess potent immunosuppressive activity, mapped to a 17-amino acid residue conserved domain. Synthetic peptides corresponding to this immunosuppressive domain can inhibit lymphocyte activation, whereas mutation of key domain residues can increase the lymphocyte response to linked antigenic epitopes. Using three T cell receptors (TCRs) of defined specificity, we examine the effect of the immunosuppressive domain on the T cell response to their respective antigenic peptides. We find that fusion of a T cell epitope to the immunosuppressive domain can greatly modulate its potency. However, the effects heavily depend on the particular combination of TCR and peptide-major histocompatibility complex class II (pMHC II), and are mimicked by sequence-scrambled peptides of similar length, suggesting they operate at the level of TCR-pMHC interaction. These results offer an alternative explanation for the immunogenicity of T cell epitopes comprising the putative immunosuppressive domain, which is more consistent with an effect on peptide antigenicity than true immunosuppressive activity.
Collapse
|
20
|
Jenkins B, Eksmond U, Young G, Kassiotis G. Antigenicity of peptides comprising the immunosuppressive domain of the retroviral envelope glycoprotein. Wellcome Open Res 2016; 1:22. [PMID: 28111636 PMCID: PMC5242373 DOI: 10.12688/wellcomeopenres.10269.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To achieve persistent infection of the host, viruses often subvert or suppress host immunity through mechanisms that are not entirely understood. The envelope glycoprotein of several retroviruses is thought to possess potent immunosuppressive activity, mapped to a 17-amino acid residue conserved domain. Synthetic peptides corresponding to this immunosuppressive domain can inhibit lymphocyte activation, whereas mutation of key domain residues can increase the lymphocyte response to linked antigenic epitopes. Using three T cell receptors (TCRs) of defined specificity, we examine the effect of the immunosuppressive domain on the T cell response to their respective antigenic peptides. We find that fusion of a T cell epitope to the immunosuppressive domain can greatly modulate its potency. However, the effects heavily depend on the particular combination of TCR and peptide-major histocompatibility complex class II (pMHC II), and are mimicked by sequence-scrambled peptides of similar length, suggesting they operate at the level of TCR-pMHC interaction. These results offer an alternative explanation for the immunogenicity of T cell epitopes comprising the putative immunosuppressive domain, which is more consistent with an effect on peptide antigenicity than true immunosuppressive activity.
Collapse
Affiliation(s)
| | | | | | - George Kassiotis
- The Francis Crick Institute, London, UK.,Department of Medicine, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
21
|
Guillaume-Vasselin V, Lemaitre L, Dhondt KP, Tedeschi L, Poulard A, Charreyre C, Horvat B. Protection from Hendra virus infection with Canarypox recombinant vaccine. NPJ Vaccines 2016; 1:16003. [PMID: 29263849 PMCID: PMC5707888 DOI: 10.1038/npjvaccines.2016.3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 04/14/2016] [Accepted: 05/25/2016] [Indexed: 11/18/2022] Open
Abstract
Hendra virus (HeV) is an emerging zoonotic pathogen, which causes severe respiratory illness and encephalitis in humans and horses. Since its first appearance in 1994, spillovers of HeV from its natural reservoir fruit bats occur on almost an annual basis. The high mortality rate in both humans and horses and the wide-ranging reservoir distribution are making HeV a serious public health problem, especially for people exposed to sick horses. This study has aimed to develop an efficient low-cost HeV vaccine for horses based on Canarypox recombinant vector expressing HeV glycoproteins, attachment glycoprotein (G) and fusion protein (F). This vaccine was used to immunise hamsters and then challenged intraperitoneally with HeV 3 weeks later. The higher tested dose of the vaccine efficiently prevented oropharyngeal virus shedding and protected animals from clinical disease and virus-induced mortality. Vaccine induced generation of seroneutralising antibodies and prevented virus-induced histopathological changes and a production of viral RNA and antigens in animal tissues. Interestingly, some vaccinated animals, including those immunised at a lower dose, were protected in the absence of detectable specific antibodies, suggesting the induction of an efficient virus-specific cellular immunity. Finally, ponies immunised using the same vaccination protocol as hamsters developed strong seroneutralising titres against both HeV and closely related Nipah virus, indicating that this vaccine may have the ability to induce cross-protection against Henipavirus infection. These data suggest that Canarypox-based vectors encoding for HeV glycoproteins present very promising new vaccine candidate to prevent infection and shedding of the highly lethal HeV.
Collapse
Affiliation(s)
- Vanessa Guillaume-Vasselin
- CIRI, International Center for Infectiology Research, Lyon, France.,Inserm, U1111, Lyon, France.,CNRS, UMR5308, Lyon, France.,Université Lyon 1, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Kévin P Dhondt
- CIRI, International Center for Infectiology Research, Lyon, France.,Inserm, U1111, Lyon, France.,CNRS, UMR5308, Lyon, France.,Université Lyon 1, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | - Branka Horvat
- CIRI, International Center for Infectiology Research, Lyon, France.,Inserm, U1111, Lyon, France.,CNRS, UMR5308, Lyon, France.,Université Lyon 1, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
22
|
Reply to "Immunosuppression in a Comparative Study of Feline Leukemia Virus Vaccines". CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 22:1296-7. [PMID: 26604265 DOI: 10.1128/cvi.00504-15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Immunosuppression in a Comparative Study of Feline Leukemia Virus Vaccines. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 22:1294-5. [PMID: 26604264 DOI: 10.1128/cvi.00497-15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
In response to Letter to the Editor (Regina Hofmann-Lehmann, Laura S. Levy and Brian Willett)—Comparing the efficacy of FeLV vaccines. Vaccine 2015; 33:2739-40. [DOI: 10.1016/j.vaccine.2015.01.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 01/16/2015] [Accepted: 01/26/2015] [Indexed: 11/18/2022]
|
25
|
Denner J. Treatment of Ebola virus infections with inhibitors of TLR4. Med Hypotheses 2015; 85:253-7. [PMID: 26003830 DOI: 10.1016/j.mehy.2015.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 05/04/2015] [Accepted: 05/10/2015] [Indexed: 01/25/2023]
Abstract
Ebola virus (EBOV) infection is associated with modulation of cytokine expression in infected patients. EBOV has been shown to interact directly with immune cells (at minimum with macrophages and dendritic cells) and modulation of cytokine expression has also been observed in vitro, which is similar to that in vivo. The modulation of cytokine expression observed in vitro was independent of virus infection and the glycoprotein GP1,2 was shown to be necessary and sufficient for cytokine modulation. Interestingly, similar changes in gene expression were observed in cells treated with lipopolysaccharide (LPS). As evidence suggests that GP1,2 and LPS use the same receptor, it is tempting to evaluate whether compounds that can inhibit signal transduction by LPS, e.g., TAK-242, can also reduce EBOV-induced pathogenesis.
Collapse
|
26
|
Lokossou AG, Toudic C, Barbeau B. Implication of human endogenous retrovirus envelope proteins in placental functions. Viruses 2014; 6:4609-27. [PMID: 25421890 PMCID: PMC4246240 DOI: 10.3390/v6114609] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 11/02/2014] [Accepted: 11/07/2014] [Indexed: 12/21/2022] Open
Abstract
Human endogenous retroviruses (ERVs) represent 8% of the total human genome. Although the majority of these ancient proviral sequences have only retained non-coding long terminal repeats (LTRs), a number of “endogenized” retroviral genes encode functional proteins. Previous studies have underlined the implication of these ERV-derived proteins in the development and the function of the placenta. In this review, we summarize recent findings showing that two ERV genes, termed Syncytin-1 and Syncytin-2, which encode former envelope (Env) proteins, trigger fusion events between villous cytotrophoblasts and the peripheral multinucleated syncytiotrophoblast layer. Such fusion events maintain the stability of this latter cell structure, which plays an important role in fetal development by the active secretion of various soluble factors, gas exchange and regulation of fetomaternal immunotolerance. We also highlight new studies showing that these ERV proteins, in addition to their localization at the cell surface of cytotrophoblasts, are also incorporated on the surface of various extracellular microvesicles, including exosomes. Such exosome-associated proteins could be involved in the various functions attributed to these vesicles and could provide a form of tropism. Additionally, through their immunosuppressive domains, these ERV proteins could also contribute to fetomaternal immunotolerance in a local and more distal manner. These various aspects of the implication of Syncytin-1 and -2 in placental function are also addressed in the context of the placenta-related disorder, preeclampsia.
Collapse
Affiliation(s)
- Adjimon Gatien Lokossou
- Département des Sciences Biologiques and Centre de recherche BioMed, Université du Québec à Montréal, 2080 Saint-Urbain, Montréal, PQ H2X 3X8, Canada.
| | - Caroline Toudic
- Département des Sciences Biologiques and Centre de recherche BioMed, Université du Québec à Montréal, 2080 Saint-Urbain, Montréal, PQ H2X 3X8, Canada.
| | - Benoit Barbeau
- Département des Sciences Biologiques and Centre de recherche BioMed, Université du Québec à Montréal, 2080 Saint-Urbain, Montréal, PQ H2X 3X8, Canada.
| |
Collapse
|
27
|
The transmembrane proteins contribute to immunodeficiencies induced by HIV-1 and other retroviruses. AIDS 2014; 28:1081-90. [PMID: 24445366 DOI: 10.1097/qad.0000000000000195] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Many microorganisms including retroviruses suppress the immune system of the infected host in order to maintain infection. Unfortunately, it is still unclear how retroviruses induce immunosuppression. There is increasing evidence of a common mechanism based on their transmembrane envelope proteins. This review therefore summarizes evidence of the involvement of the transmembrane envelope proteins in the immunopathogenesis of different retroviruses including HIV-1. Mutations in the immunosuppressive (isu) domain of the transmembrane envelope protein of several retroviruses abrogate the immunosuppressive activities in vitro and in vivo. Most importantly, virus sequences with such abrogating mutations were never found in HIV-1-infected individuals despite the fact that the mutated viruses are replication-competent. However, there is also evidence for additional, perhaps even divergent, strategies for each retrovirus. For example, in contrast to many other retroviruses, the HIV directly interacts with immune cells and infects them. In addition, HIV uses several accessory proteins to evade the immune response. Furthermore, the possible contribution of the transmembrane envelope proteins of endogenous retroviruses to immunosuppression when expressed on tumor cells or in the placenta is analyzed.
Collapse
|