1
|
Latifi T, Kachooei A, Jalilvand S, Zafarian S, Roohvand F, Shoja Z. Correlates of immune protection against human rotaviruses: natural infection and vaccination. Arch Virol 2024; 169:72. [PMID: 38459213 DOI: 10.1007/s00705-024-05975-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/12/2023] [Indexed: 03/10/2024]
Abstract
Species A rotaviruses are the leading viral cause of acute gastroenteritis in children under 5 years of age worldwide. Despite progress in the characterization of the pathogenesis and immunology of rotavirus-induced gastroenteritis, correlates of protection (CoPs) in the course of either natural infection or vaccine-induced immunity are not fully understood. There are numerous factors such as serological responses (IgA and IgG), the presence of maternal antibodies (Abs) in breast milk, changes in the intestinal microbiome, and rotavirus structural and non-structural proteins that contribute to the outcome of the CoP. Indeed, while an intestinal IgA response and its surrogate, the serum IgA level, are suggested as the principal CoPs for oral rotavirus vaccines, the IgG level is more likely to be a CoP for parenteral non-replicating rotavirus vaccines. Integrating clinical and immunological data will be instrumental in improving rotavirus vaccine efficacy, especially in low- and middle-income countries, where vaccine efficacy is significantly lower than in high-income countries. Further knowledge on CoPs against rotavirus disease will be helpful for next-generation vaccine development. Herein, available data and literature on interacting components and proposed CoPs against human rotavirus disease are reviewed, and limitations and gaps in our knowledge in this area are discussed.
Collapse
Affiliation(s)
- Tayebeh Latifi
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Atefeh Kachooei
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Saman Zafarian
- Department of Microbial Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
- Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
Kumar D, Shepherd FK, Springer NL, Mwangi W, Marthaler DG. Rotavirus Infection in Swine: Genotypic Diversity, Immune Responses, and Role of Gut Microbiome in Rotavirus Immunity. Pathogens 2022; 11:pathogens11101078. [PMID: 36297136 PMCID: PMC9607047 DOI: 10.3390/pathogens11101078] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/13/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Rotaviruses (RVs) are endemic in swine populations, and all swine herds certainly have a history of RV infection and circulation. Rotavirus A (RVA) and C (RVC) are the most common among all RV species reported in swine. RVA was considered most prevalent and pathogenic in swine; however, RVC has been emerging as a significant cause of enteritis in newborn piglets. RV eradication from swine herds is not practically achievable, hence producers’ mainly focus on minimizing the production impact of RV infections by reducing mortality and diarrhea. Since no intra-uterine passage of immunoglobulins occur in swine during gestation, newborn piglets are highly susceptible to RV infection at birth. Boosting lactogenic immunity in gilts by using vaccines and natural planned exposure (NPE) is currently the only way to prevent RV infections in piglets. RVs are highly diverse and multiple RV species have been reported from swine, which also contributes to the difficulties in preventing RV diarrhea in swine herds. Human RV-gut microbiome studies support a link between microbiome composition and oral RV immunogenicity. Such information is completely lacking for RVs in swine. It is not known how RV infection affects the functionality or structure of gut microbiome in swine. In this review, we provide a detailed overview of genotypic diversity of swine RVs, host-ranges, innate and adaptive immune responses to RVs, homotypic and heterotypic immunity to RVs, current methods used for RV management in swine herds, role of maternal immunity in piglet protection, and prospects of investigating swine gut microbiota in providing immunity against rotaviruses.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
- Correspondence: (D.K.); (W.M.); (D.G.M.); Tel.: +1-804-503-1241 (D.K.)
| | - Frances K Shepherd
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55108, USA
| | - Nora L. Springer
- Clinical Pathology, Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| | - Waithaka Mwangi
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
- Correspondence: (D.K.); (W.M.); (D.G.M.); Tel.: +1-804-503-1241 (D.K.)
| | - Douglas G. Marthaler
- Indical Inc., 1317 Edgewater Dr #3722, Orlando, FL 32804, USA
- Correspondence: (D.K.); (W.M.); (D.G.M.); Tel.: +1-804-503-1241 (D.K.)
| |
Collapse
|
3
|
Endt K, Wollmann Y, Haug J, Bernig C, Feigl M, Heiseke A, Kalla M, Hochrein H, Suter M, Chaplin P, Volkmann A. A Recombinant MVA-Based RSV Vaccine Induces T-Cell and Antibody Responses That Cooperate in the Protection Against RSV Infection. Front Immunol 2022; 13:841471. [PMID: 35774800 PMCID: PMC9238321 DOI: 10.3389/fimmu.2022.841471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/13/2022] [Indexed: 11/25/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes a respiratory disease with a potentially fatal outcome especially in infants and elderly individuals. Several vaccines failed in pivotal clinical trials, and to date, no vaccine against RSV has been licensed. We have developed an RSV vaccine based on the recombinant Modified Vaccinia Virus Ankara-BN® (MVA-RSV), containing five RSV-specific antigens that induced antibody and T-cell responses, which is currently tested in clinical trials. Here, the immunological mechanisms of protection were evaluated to determine viral loads in lungs upon vaccination of mice with MVA-RSV followed by intranasal RSV challenge. Depletion of CD4 or CD8 T cells, serum transfer, and the use of genetically engineered mice lacking the ability to generate either RSV-specific antibodies (T11µMT), the IgA isotype (IgA knockout), or CD8 T cells (β2M knockout) revealed that complete protection from RSV challenge is dependent on CD4 and CD8 T cells as well as antibodies, including IgA. Thus, MVA-RSV vaccination optimally protects against RSV infection by employing multiple arms of the adaptive immune system.
Collapse
Affiliation(s)
- Kathrin Endt
- Bavarian Nordic GmbH, Martinsried, Germany
- *Correspondence: Kathrin Endt,
| | | | - Jana Haug
- Bavarian Nordic GmbH, Martinsried, Germany
| | | | | | | | | | | | - Mark Suter
- University of Zürich, Dekanat Vetsuisse-Fakultät Immunology, Zurich, Switzerland
| | | | | |
Collapse
|
4
|
Shoja Z, Jalilvand S, Latifi T, Roohvand F. Rotavirus VP6: involvement in immunogenicity, adjuvant activity, and use as a vector for heterologous peptides, drug delivery, and production of nano-biomaterials. Arch Virol 2022; 167:1013-1023. [PMID: 35292854 PMCID: PMC8923333 DOI: 10.1007/s00705-022-05407-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/26/2022] [Indexed: 12/15/2022]
Abstract
The first-generation, live attenuated rotavirus (RV) vaccines, such as RotaTeq and Rotarix, were successful in reducing the number of RV-induced acute gastroenteritis (AGE) and child deaths globally. However, the low efficacy of these first-generation oral vaccines, coupled with safety concerns, required development of improved RV vaccines. The highly conserved structural protein VP6 is highly immunogenic, and it can generate self-assembled nano-sized structures, including tubes and spheres (virus-like particles; VLPs). Amongst the RV proteins, only VP6 shows these features. Interestingly, VP6-assembled structures, in addition to being highly immunogenic, have several other useful characteristics that could allow them to be used as adjuvants, immunological carriers, and drug-delivery vehicles as well as acting a scaffold for production of valuable nano-biomaterials. This review provides an overview of the self-assembled nano-sized structures of VP6-tubes/VLPs and their various functions.
Collapse
Affiliation(s)
- Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
5
|
T-Cell Responses after Rotavirus Infection or Vaccination in Children: A Systematic Review. Viruses 2022; 14:v14030459. [PMID: 35336866 PMCID: PMC8951614 DOI: 10.3390/v14030459] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
Cellular immunity against rotavirus in children is incompletely understood. This review describes the current understanding of T-cell immunity to rotavirus in children. A systematic literature search was conducted in Embase, MEDLINE, Web of Science, and Global Health databases using a combination of “t-cell”, “rotavirus” and “child” keywords to extract data from relevant articles published from January 1973 to March 2020. Only seventeen articles were identified. Rotavirus-specific T-cell immunity in children develops and broadens reactivity with increasing age. Whilst occurring in close association with antibody responses, T-cell responses are more transient but can occur in absence of detectable antibody responses. Rotavirus-induced T-cell immunity is largely of the gut homing phenotype and predominantly involves Th1 and cytotoxic subsets that may be influenced by IL-10 Tregs. However, rotavirus-specific T-cell responses in children are generally of low frequencies in peripheral blood and are limited in comparison to other infecting pathogens and in adults. The available research reviewed here characterizes the T-cell immune response in children. There is a need for further research investigating the protective associations of rotavirus-specific T-cell responses against infection or vaccination and the standardization of rotavirus-specific T-cells assays in children.
Collapse
|
6
|
Devi YD, Devi A, Gogoi H, Dehingia B, Doley R, Buragohain AK, Singh CS, Borah PP, Rao CD, Ray P, Varghese GM, Kumar S, Namsa ND. Exploring rotavirus proteome to identify potential B- and T-cell epitope using computational immunoinformatics. Heliyon 2020; 6:e05760. [PMID: 33426322 PMCID: PMC7779714 DOI: 10.1016/j.heliyon.2020.e05760] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/02/2020] [Accepted: 12/14/2020] [Indexed: 11/28/2022] Open
Abstract
Rotavirus is the most common cause of acute gastroenteritis in infants and children worldwide. The functional correlation of B- and T-cells to long-lasting immunity against rotavirus infection in the literature is limited. In this work, a series of computational immuno-informatics approaches were applied and identified 28 linear B-cells, 26 conformational B-cell, 44 TC cell and 40 TH cell binding epitopes for structural and non-structural proteins of rotavirus. Further selection of putative B and T cell epitopes in the multi-epitope vaccine construct was carried out based on immunogenicity, conservancy, allergenicity and the helical content of predicted epitopes. An in-silico vaccine constructs was developed using an N-terminal adjuvant (RGD motif) followed by TC and TH cell epitopes and B-cell epitope with an appropriate linker. Multi-threading models of multi-epitope vaccine construct with B- and T-cell epitopes were generated and molecular dynamics simulation was performed to determine the stability of designed vaccine. Codon optimized multi-epitope vaccine antigens was expressed and affinity purified using the E. coli expression system. Further the T cell epitope presentation assay using the recombinant multi-epitope constructs and the T cell epitope predicted and identified in this study have not been investigated. Multi-epitope vaccine construct encompassing predicted B- and T-cell epitopes may help to generate long-term immune responses against rotavirus. The computational findings reported in this study may provide information in developing epitope-based vaccine and diagnostic assay for rotavirus-led diarrhea in children's.
Collapse
Affiliation(s)
- Yengkhom Damayanti Devi
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam 784 028, Assam, India
| | - Arpita Devi
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam 784 028, Assam, India
| | - Hemanga Gogoi
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam 784 028, Assam, India
| | - Bondita Dehingia
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam 784 028, Assam, India
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam 784 028, Assam, India
| | | | - Ch Shyamsunder Singh
- Department of Paediatrics, Regional Institute of Medical Sciences, Imphal, India
| | - Partha Pratim Borah
- Department of Paediatrics and Neonatology, Pratiksha Hospital, Guwahati, India
| | - C Durga Rao
- School of Liberal Arts and Basic Sciences, SRM University AP, Amaravati, India
| | - Pratima Ray
- Department of Biotechnology, Jamia Hamdard, Delhi, India
| | - George M Varghese
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, India
| | - Nima D Namsa
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam 784 028, Assam, India
| |
Collapse
|
7
|
Rotavirus VP6 Adjuvant Effect on Norovirus GII.4 Virus-Like Particle Uptake and Presentation by Bone Marrow-Derived Dendritic Cells In Vitro and In Vivo. J Immunol Res 2020; 2020:3194704. [PMID: 32411793 PMCID: PMC7204108 DOI: 10.1155/2020/3194704] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/20/2019] [Indexed: 12/31/2022] Open
Abstract
We have previously shown that rotavirus (RV) inner capsid protein VP6 has an adjuvant effect on norovirus (NoV) virus-like particle- (VLP-) induced immune responses and studied the adjuvant mechanism in immortalized cell lines used as antigen-presenting cells (APCs). Here, we investigated the uptake and presentation of RV VP6 and NoV GII.4 VLPs by primary bone marrow-derived dendritic cells (BMDCs). The adjuvant effect of VP6 on GII.4 VLP presentation and NoV-specific immune response induction by BMDC in vivo was also studied. Intracellular staining demonstrated that BMDCs internalized both antigens, but VP6 more efficiently than NoV VLPs. Both antigens were processed and presented to antigen-primed T cells, which responded by robust interferon γ secretion. When GII.4 VLPs and VP6 were mixed in the same pulsing reaction, a subpopulation of the cells had uptaken both antigens. Furthermore, VP6 copulsing increased GII.4 VLP uptake by 37% and activated BMDCs to secrete 2-5-fold increased levels of interleukin 6 and tumor necrosis factor α compared to VLP pulsing alone. When in vitro-pulsed BMDCs were transferred to syngeneic BALB/c mice, VP6 improved NoV-specific antibody responses. The results of this study support the earlier findings of VP6 adjuvant effect in vitro and in vivo.
Collapse
|
8
|
Heinimäki S, Hankaniemi MM, Sioofy-Khojine AB, Laitinen OH, Hyöty H, Hytönen VP, Vesikari T, Blazevic V. Combination of three virus-derived nanoparticles as a vaccine against enteric pathogens; enterovirus, norovirus and rotavirus. Vaccine 2019; 37:7509-7518. [DOI: 10.1016/j.vaccine.2019.09.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/23/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022]
|
9
|
T lymphocytes in the intestinal mucosa: defense and tolerance. Cell Mol Immunol 2019; 16:216-224. [PMID: 30787416 DOI: 10.1038/s41423-019-0208-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 02/01/2019] [Indexed: 02/06/2023] Open
Abstract
Although lymphocytes are known to circulate throughout lymphoid tissues and blood, they also establish residency in nonlymphoid organs, most prominently in barrier tissues, such as the intestines. The adaptation of T lymphocytes to intestinal environments requires constant discrimination between natural stimulation from commensal flora and food and pathogens that need to be cleared. Genetic variations that cause a defective defense or a break in tolerance along with environmental cues, such as infection or imbalances in the gut microbiota known as dysbiosis, can trigger several immune disorders via the activation of T lymphocytes in the intestines. Elucidation of the immune mechanisms that distinguish between commensal flora and pathogenic organisms may reveal therapeutic targets for the prevention or modulation of inflammatory diseases and boost the efficacy of cancer immunotherapy. In this review, we discuss the development and adaptation of T lymphocytes in the intestine, how these cells protect the host against pathogenic infections while tolerating food antigens and commensal microbiota, and the potential implications of targeting these cells for disease management and therapeutics.
Collapse
|
10
|
Afchangi A, Jalilvand S, Mohajel N, Marashi SM, Shoja Z. Rotavirus VP6 as a potential vaccine candidate. Rev Med Virol 2019; 29:e2027. [DOI: 10.1002/rmv.2027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Atefeh Afchangi
- Virology Department, School of Public Health; Tehran University of Medical Sciences; Tehran Iran
| | - Somayeh Jalilvand
- Virology Department, School of Public Health; Tehran University of Medical Sciences; Tehran Iran
| | - Nasir Mohajel
- Virology Department; Pasteur Institute of Iran; Tehran Iran
| | - Sayed Mahdi Marashi
- Virology Department, School of Public Health; Tehran University of Medical Sciences; Tehran Iran
| | | |
Collapse
|
11
|
Antonio-Herrera L, Badillo-Godinez O, Medina-Contreras O, Tepale-Segura A, García-Lozano A, Gutierrez-Xicotencatl L, Soldevila G, Esquivel-Guadarrama FR, Idoyaga J, Bonifaz LC. The Nontoxic Cholera B Subunit Is a Potent Adjuvant for Intradermal DC-Targeted Vaccination. Front Immunol 2018; 9:2212. [PMID: 30319653 PMCID: PMC6171476 DOI: 10.3389/fimmu.2018.02212] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022] Open
Abstract
CD4+ T cells are major players in the immune response against several diseases; including AIDS, leishmaniasis, tuberculosis, influenza and cancer. Their activation has been successfully achieved by administering antigen coupled with antibodies, against DC-specific receptors in combination with adjuvants. Unfortunately, most of the adjuvants used so far in experimental models are unsuitable for human use. Therefore, human DC-targeted vaccination awaits the description of potent, yet nontoxic adjuvants. The nontoxic cholera B subunit (CTB) can be safely used in humans and it has the potential to activate CD4+ T cell responses. However, it remains unclear whether CTB can promote DC activation and can act as an adjuvant for DC-targeted antigens. Here, we evaluated the CTB's capacity to activate DCs and CD4+ T cell responses, and to generate long-lasting protective immunity. Intradermal (i.d.) administration of CTB promoted late and prolonged activation and accumulation of skin and lymphoid-resident DCs. When CTB was co-administered with anti-DEC205-OVA, it promoted CD4+ T cell expansion, differentiation, and infiltration to peripheral nonlymphoid tissues, i.e., the skin, lungs and intestine. Indeed, CTB promoted a polyfunctional CD4+ T cell response, including the priming of Th1 and Th17 cells, as well as resident memory T (RM) cell differentiation in peripheral nonlymphoid tissues. It is worth noting that CTB together with a DC-targeted antigen promoted local and systemic protection against experimental melanoma and murine rotavirus. We conclude that CTB administered i.d. can be used as an adjuvant to DC-targeted antigens for the induction of broad CD4+ T cell responses as well as for promoting long-lasting protective immunity.
Collapse
Affiliation(s)
- Laura Antonio-Herrera
- Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Inmunoquímica, Mexico City, Mexico.,Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Oscar Badillo-Godinez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SS, Cuernavaca, Mexico
| | - Oscar Medina-Contreras
- Immunology and Proteomics Laboratory, Mexico Children's Hospital "Federico Gómez", Mexico City, Mexico
| | - Araceli Tepale-Segura
- Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Inmunoquímica, Mexico City, Mexico
| | - Alberto García-Lozano
- Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Inmunoquímica, Mexico City, Mexico
| | | | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States
| | - Laura C Bonifaz
- Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Inmunoquímica, Mexico City, Mexico
| |
Collapse
|
12
|
Temprana CF, Argüelles MH, Gutierrez NM, Barril PA, Esteban LE, Silvestre D, Mandile MG, Glikmann G, Castello AA. Rotavirus VP6 protein mucosally delivered by cell wall-derived particles from Lactococcus lactis induces protection against infection in a murine model. PLoS One 2018; 13:e0203700. [PMID: 30192869 PMCID: PMC6128627 DOI: 10.1371/journal.pone.0203700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/24/2018] [Indexed: 01/21/2023] Open
Abstract
Rotaviruses are the primary cause of acute gastroenteritis in children worldwide. Although the implementation of live attenuated vaccines has reduced the number of rotavirus-associated deaths, variance in their effectiveness has been reported in different countries. This fact, among other concerns, leads to continuous efforts for the development of new generation of vaccines against rotavirus.In this work, we describe the obtention of cell wall-derived particles from a recombinant Lactococcus lactis expressing a cell wall-anchored version of the rotavirus VP6 protein. After confirming by SDS-PAGE, Western blot, flow cytometry and electronic immunomicroscopy that these particles were carrying the VP6 protein, their immunogenic potential was evaluated in adult BALB/c mice. For that, mucosal immunizations (oral or intranasal), with or without the dmLT [(double mutant Escherichia coli heat labile toxin LT(R192G/L211A)] adjuvant were performed. The results showed that these cell wall-derived particles were able to generate anti-rotavirus IgG and IgA antibodies only when administered intranasally, whether the adjuvant was present or not. However, the presence of dmLT was necessary to confer protection against rotavirus infection, which was evidenced by a 79.5 percent viral shedding reduction.In summary, this work describes the production of cell wall-derived particles which were able to induce a protective immune response after intranasal immunization. Further studies are needed to characterize the immune response elicited by these particles as well as to determine their potential as an alternative to the use of live L. lactis for mucosal antigen delivery.
Collapse
Affiliation(s)
- C. Facundo Temprana
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
- * E-mail: (AAC); (CFT)
| | - Marcelo H. Argüelles
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
| | - Nicolás M. Gutierrez
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
| | - Patricia A. Barril
- Laboratorio de Microbiología de los Alimentos, Centro de Investigación y Asistencia Técnica a la Industria (CIATI A.C.)–CONICET, Centenario, Neuquén, Argentina
| | - Laura E. Esteban
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
| | - Dalila Silvestre
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
| | - Marcelo G. Mandile
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
| | - Graciela Glikmann
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
| | - Alejandro A. Castello
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- Instituto de Ciencias de la Salud, Universidad Nacional Arturo Jauretche, Florencio Varela, Buenos Aires, Argentina
- * E-mail: (AAC); (CFT)
| |
Collapse
|
13
|
Heinimäki S, Malm M, Vesikari T, Blazevic V. Intradermal and intranasal immunizations with oligomeric middle layer rotavirus VP6 induce Th1, Th2 and Th17 T cell subsets and CD4 + T lymphocytes with cytotoxic potential. Antiviral Res 2018; 157:1-8. [DOI: 10.1016/j.antiviral.2018.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 12/26/2022]
|
14
|
Labastida-Conde RG, Ramírez-Pliego O, Peleteiro-Olmedo M, Lopez-Guerrero DV, Badillo-Godinez OD, Gutiérrez-Xicoténcatl MDL, Rosas-Salgado G, González-Fernández Á, Esquivel-Guadarrama FR, Santana MA. Flagellin is a Th1 polarizing factor for human CD4 + T cells and induces protection in a murine neonatal vaccination model of rotavirus infection. Vaccine 2018; 36:4188-4197. [PMID: 29891347 DOI: 10.1016/j.vaccine.2018.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/29/2018] [Accepted: 06/03/2018] [Indexed: 02/07/2023]
Abstract
Neonates have an increased susceptibility to infections, particularly those caused by intracellular pathogens, leading to high morbidity and mortality rates. This is partly because of a poor response of neonatal CD4+ T cells, leading to deficient antibody production and a low production of IFN-γ, resulting in deficient elimination of intracellular pathogens. The poor memory response of human neonates has underpinned the need for improving vaccine formulations. Molecular adjuvants that improve the response of neonatal lymphocytes, such as the ligands of toll-like receptors (TLRs), are attractive candidates. Among them, flagellin, the TLR5 ligand, is effective at very low doses; prior immunity to flagellin does not impair its adjuvant activity. Human CD4+ and CD8+ T cells express TLR5. We found that flagellin induces the expression of IFN-γ, IL-1β and IL-12 in mononuclear cells from human neonate and adult donors. When human naïve CD4+ T cells were activated in the presence of flagellin, there was high level of expression of IFN-γ in both neonates and adults. Furthermore, flagellin induced IFN-γ production in Th1 cells obtained from adult donors; in the Th2 population, it inhibited IL-4 cytokine production. Flagellin also promoted expression of the IFN-γ receptor in naive CD4+ T cells from neonates and adults. To test the adjuvant capacity of flagellin in vivo, we used a murine neonate vaccination model for infection with rotavirus, a pathogen responsible for severe diarrhea in young infants. Using the conserved VP6 antigen, we observed an 80% protection against rotavirus infection in the presence of flagellin, but only in those mice previously primed in the neonatal period. Our data suggest that flagellin could be an attractive adjuvant for achieving a Th1 response.
Collapse
Affiliation(s)
| | - Oscar Ramírez-Pliego
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Mercedes Peleteiro-Olmedo
- Inmunología, Centro de Investigaciones Biomédicas (CINBIO), Centro Singular de Investigación de Galicia, Instituto de Investigación Sanitaria Galicia Sur, Universidad de Vigo, Campus Universitario de Vigo, 36310 Vigo, Spain
| | | | | | | | - Gabriela Rosas-Salgado
- Facultad de Medicina, Universidad Autonoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - África González-Fernández
- Inmunología, Centro de Investigaciones Biomédicas (CINBIO), Centro Singular de Investigación de Galicia, Instituto de Investigación Sanitaria Galicia Sur, Universidad de Vigo, Campus Universitario de Vigo, 36310 Vigo, Spain
| | | | - M Angélica Santana
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
15
|
Arnold MM. Rotavirus vaccines: why continued investment in research is necessary. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018; 5:73-81. [PMID: 29805958 PMCID: PMC5967271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
PURPOSE OF REVIEW Rotavirus vaccines were first introduced more than a decade ago and have had a tremendous impact on reducing the number of hospitalizations and deaths due to rotavirus-associated diarrhea. This review will discuss current rotavirus vaccines, post-licensure surveillance, progress in non-replicating vaccine development, and why continued research is important for understanding a virus that remains a globally leading cause of death due to diarrhea. RECENT FINDINGS Research advances have enhanced our understanding of how vaccines induce protection against subsequent severe disease, how the virus replicates and spreads in the face of the host immune system, and basic mechanisms governing the viral life cycle. SUMMARY Much remains to be learned about how to improve vaccine success, what are the molecular determinants of host range and virulence, and what are the interactions of the virus with the host that drive its replicative success, among many other important questions.
Collapse
Affiliation(s)
- Michelle M. Arnold
- Corresponding author: Michelle M. Arnold, , Telephone: 318-675-4731, ORCID: 0000-0001-9219-3097
| |
Collapse
|
16
|
Frederick DR, Goggins JA, Sabbagh LM, Freytag LC, Clements JD, McLachlan JB. Adjuvant selection regulates gut migration and phenotypic diversity of antigen-specific CD4 + T cells following parenteral immunization. Mucosal Immunol 2018; 11:549-561. [PMID: 28792004 PMCID: PMC6252260 DOI: 10.1038/mi.2017.70] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 06/28/2017] [Indexed: 02/04/2023]
Abstract
Infectious diarrheal diseases are the second leading cause of death in children under 5 years, making vaccines against these diseases a high priority. It is known that certain vaccine adjuvants, chiefly bacterial ADP-ribosylating enterotoxins, can induce mucosal antibodies when delivered parenterally. Based on this, we reasoned vaccine-specific mucosal cellular immunity could be induced via parenteral immunization with these adjuvants. Here, we show that, in contrast to the Toll-like receptor-9 agonist CpG, intradermal immunization with non-toxic double-mutant heat-labile toxin (dmLT) from enterotoxigenic Escherichia coli drove endogenous, antigen-specific CD4+ T cells to expand and upregulate the gut-homing integrin α4β7. This was followed by T-cell migration into gut-draining lymph nodes and both small and large intestines. We also found that dmLT produces a balanced T-helper 1 and 17 (Th1 and Th17) response, whereas T cells from CpG immunized mice were predominantly Th1. Immunization with dmLT preferentially engaged CD103+ dendritic cells (DCs) compared with CpG, and mice deficient in CD103+ DCs were unable to fully license antigen-specific T-cell migration to the intestinal mucosae following parenteral immunization. This work has the potential to redirect the design of existing and next generation vaccines to elicit pathogen-specific immunity in the intestinal tract with non-mucosal immunization.
Collapse
Affiliation(s)
- Daniel R. Frederick
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA 70112
| | - J. Alan Goggins
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA 70112
| | - Leila M. Sabbagh
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA 70112
| | - Lucy C. Freytag
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA 70112
| | - John D. Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA 70112
| | - James B. McLachlan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA 70112
| |
Collapse
|
17
|
Arnold MM. Rotavirus Vaccines: Why Continued Investment in Research Is Necessary. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018. [DOI: 10.1007/s40588-018-0079-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
18
|
Kondakova OA, Nikitin NA, Trifonova EA, Atabekov JG, Karpova OV. Rotavirus Vaccines: New Strategies and Approaches. ACTA ACUST UNITED AC 2018. [DOI: 10.3103/s0096392517040071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
19
|
Afchangi A, Arashkia A, Shahosseini Z, Jalilvand S, Marashi SM, Roohvand F, Mohajel N, Shoja Z. Immunization of Mice by Rotavirus NSP4-VP6 Fusion Protein Elicited Stronger Responses Compared to VP6 Alone. Viral Immunol 2017; 31:233-241. [PMID: 29185875 DOI: 10.1089/vim.2017.0075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Due to the limitations and safety issues of the two currently approved live attenuated rotavirus (RV) vaccines "RotaTeq and Rotarix," studies on nonreplicating sources of RV vaccines and search for proper RV antigens are actively carried out. The adjuvant activity of NSP4 and highly immunogenic properties of RV VP6 protein prompted us to consider the construction of a NSP4112-175-VP6 fusion protein and to assess the anti-VP6 IgG, IgA, and IgG subclass responses induced by Escherichia coli-derived NSP4-VP6 fusion protein compared to that of VP6 protein with/without formulation in Montanide ISA 50V2 (M50) in BALB/c mice. Results indicated to the proper expression of the fused NSP4-VP6 and VP6 proteins in E. coli. Intraperitoneal immunization by M50 formulated NSP4-VP6 fusion protein (M5+NSP4-VP6) induced the highest titration of VP6-specific IgG and IgA responses compared to the other groups. Indeed, the presence of NSP4 resulted to the induction of stronger humoral immune responses against the fused protein compared to that elicited by administration of VP6 protein alone (with/without M50 formulation), implying the adjuvant properties of NSP4 for the fused protein. Moreover, the "M50+NSP4-VP6" formulation induced higher serum IgG2a titers than IgG1 and increased Interferon-γ levels, despite unchanged interleukin-4 amounts compared to other groups, indicating Th1-oriented responses with a possible role of NSP4. In conclusion, this study further highlights the potentiality of NSP4-VP6 fusion protein as an efficient and cost-effective immunogen in the field of RV vaccine development.
Collapse
Affiliation(s)
- Atefeh Afchangi
- 1 Virology Department, School of Public Health (SPH), Tehran University of Medical Sciences (TUMS) , Tehran, Iran .,2 Virology Department, Pasteur Institute of Iran , Tehran, Iran
| | - Arash Arashkia
- 2 Virology Department, Pasteur Institute of Iran , Tehran, Iran
| | | | - Somayeh Jalilvand
- 1 Virology Department, School of Public Health (SPH), Tehran University of Medical Sciences (TUMS) , Tehran, Iran
| | - Sayed Mahdi Marashi
- 1 Virology Department, School of Public Health (SPH), Tehran University of Medical Sciences (TUMS) , Tehran, Iran
| | - Farzin Roohvand
- 2 Virology Department, Pasteur Institute of Iran , Tehran, Iran
| | - Nasir Mohajel
- 2 Virology Department, Pasteur Institute of Iran , Tehran, Iran
| | | |
Collapse
|
20
|
Palaschak B, Marsic D, Herzog RW, Zolotukhin S, Markusic DM. An Immune-Competent Murine Model to Study Elimination of AAV-Transduced Hepatocytes by Capsid-Specific CD8 + T Cells. Mol Ther Methods Clin Dev 2017; 5:142-152. [PMID: 28480313 PMCID: PMC5415329 DOI: 10.1016/j.omtm.2017.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 04/13/2017] [Indexed: 01/13/2023]
Abstract
Multiple independent adeno-associated virus (AAV) gene therapy clinical trials for hemophilia B, utilizing different AAV serotypes, have reported a vector dose-dependent loss of circulating factor IX (FIX) protein associated with capsid-specific CD8+ T cell (Cap-CD8) elimination of transduced hepatocytes. Hemophilia B patients who develop transient transaminitis and loss of FIX protein may be stabilized with the immune-suppressive (IS) drug prednisolone, but do not all recover lost FIX expression, whereas some patients fail to respond to IS. We developed the first animal model demonstrating Cap-CD8 infiltration and elimination of AAV-transduced hepatocytes of immune-deficient mice. Here, we extend this model to an immune-competent host where Cap-CD8 transfer to AAV2-F9-treated mice significantly reduced circulating and hepatocyte FIX expression. Further, we studied two high-expressing liver tropic AAV2 variants, AAV2-LiA and AAV2-LiC, obtained from a rationally designed capsid library. Unlike AAV2, Cap-CD8 did not initially reduce circulating FIX levels for either variant. However, FIX levels were significantly reduced in AAV2-LiC-F9-treated, but not AAV2-LiA-F9-treated, mice at the study endpoint. Going forward, the immune-competent model may provide an opportunity to induce immunological memory directed against a surrogate AAV capsid antigen and study recall responses following AAV gene transfer.
Collapse
Affiliation(s)
- Brett Palaschak
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Damien Marsic
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Roland W. Herzog
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Sergei Zolotukhin
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - David M. Markusic
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
21
|
Wen K, Bui T, Weiss M, Li G, Kocher J, Yang X, Jobst PM, Vaught T, Ramsoondar J, Ball S, Clark-Deener S, Ayares D, Yuan L. B-Cell-Deficient and CD8 T-Cell-Depleted Gnotobiotic Pigs for the Study of Human Rotavirus Vaccine-Induced Protective Immune Responses. Viral Immunol 2016; 29:112-27. [PMID: 26824402 PMCID: PMC4782039 DOI: 10.1089/vim.2015.0105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genetically modified pigs have become available recently. In this study, we established the gnotobiotic pig model of human rotavirus (HRV) infection using cloned pigs with homozygous disruption in the gene encoding immunoglobulin heavy chain (HCKO), which totally impairs B-cell development. To clarify importance of B cells and cytotoxic T cells in rotavirus immunity, CD8 cells in a subset of the pigs were depleted by injecting antipig CD8 antibodies and the immune phenotypes of all pigs were examined. HCKO pigs, CD8 cell-depleted HCKO pigs, and wild-type (WT) pigs were vaccinated with an attenuated HRV vaccine and challenged with virulent HRV. Protection against HRV infection and diarrhea was assessed postchallenge and detailed T-cell subset responses were determined pre- and postchallenge. Significantly longer duration of virus shedding was seen in vaccinated HCKO pigs than in WT pigs, indicating the importance of B cells in vaccine-induced protective immunity. Vaccinated HCKO/CD8(-) pigs shed significantly higher number of infectious virus than WT pigs and non-CD8-depleted HCKO pigs, indicating the importance of CD8 T cells in controlling virus replication. Therefore, both B cells and CD8 T cells play an important role in the protection against rotavirus infection. HCKO and HCKO/CD8(-) pigs did not differ significantly in diarrhea and virus shedding postchallenge; increased CD4 and CD8(-) γδ T-cell responses probably compensated partially for the lack of CD8 T cells. This study demonstrated that HCKO pigs can serve as a valuable model for dissection of protective immune responses against viral infections and diseases.
Collapse
Affiliation(s)
- Ke Wen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Tammy Bui
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Mariah Weiss
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Guohua Li
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Jacob Kocher
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Xingdong Yang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Peter M. Jobst
- Teaching & Research Animal Care Support Service, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | | | | | | | - Sherrie Clark-Deener
- Department of Large Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | | | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| |
Collapse
|
22
|
Jalilvand S, Marashi SM, Shoja Z. Rotavirus VP6 preparations as a non-replicating vaccine candidates. Vaccine 2015; 33:3281-7. [PMID: 26021725 DOI: 10.1016/j.vaccine.2015.05.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/05/2015] [Accepted: 05/13/2015] [Indexed: 01/03/2023]
Abstract
Rotavirus (RV) structural proteins VP4 and VP7, located on the surface of viral particles, elicit neutralizing antibodies (Abs) and are therefore considered to be important components of RV vaccines. However, despite inducing neutralizing Abs, limits of cross-neutralizing activity and lack of full correlation with protection limit the usefulness of these proteins as protective agents against RV disease. VP6 protein, which forms the middle layer of RV particles, is discussed as an alternative vaccine candidate since it can induce cross-protective immune responses against different RV strains although the Ab raised is not neutralizing. This report reviews different functions of VP6 that can lead to considering it as an alternative vaccine against RV disease.
Collapse
Affiliation(s)
- Somayeh Jalilvand
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Mahdi Marashi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
23
|
Badillo-Godinez O, Gutierrez-Xicotencatl L, Plett-Torres T, Pedroza-Saavedra A, Gonzalez-Jaimes A, Chihu-Amparan L, Maldonado-Gama M, Espino-Solis G, Bonifaz LC, Esquivel-Guadarrama F. Targeting of rotavirus VP6 to DEC-205 induces protection against the infection in mice. Vaccine 2015; 33:4228-37. [PMID: 25850020 DOI: 10.1016/j.vaccine.2015.03.080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/17/2015] [Accepted: 03/24/2015] [Indexed: 01/15/2023]
Abstract
Rotavirus (RV) is the primary etiologic agent of severe gastroenteritis in human infants. Although two attenuated RV-based vaccines have been licensed to be applied worldwide, they are not so effective in low-income countries, and the induced protection mechanisms have not been clearly established. Thus, it is important to develop new generation vaccines that induce long lasting heterotypic immunity. VP6 constitutes the middle layer protein of the RV virion. It is the most conserved protein and it is the target of protective T-cells; therefore, it is a potential candidate antigen for a new generation vaccine against the RV infection. We determined whether targeting the DEC-205 present in dendritic cells (DCs) with RV VP6 could induce protection at the intestinal level. VP6 was cross-linked to a monoclonal antibody (mAb) against murine DEC-205 (αDEC-205:VP6), and BALB/c mice were inoculated subcutaneously (s.c.) twice with the conjugated containing 1.5 μg of VP6 in the presence of polyinosinic-polycytidylic acid (Poly I:C) as adjuvant. As controls and following the same protocol, mice were immunized with ovalbumin (OVA) cross-linked to the mAb anti-DEC-205 (αDEC-205:OVA), VP6 cross-linked to a control isotype mAb (Isotype:VP6), 3 μg of VP6 alone, Poly I:C or PBS. Two weeks after the last inoculation, mice were orally challenged with a murine RV. Mice immunized with α-DEC-205:VP6 and VP6 alone presented similar levels of serum Abs to VP6 previous to the virus challenge. However, after the virus challenge, only α-DEC-205:VP6 induced up to a 45% IgA-independent protection. Memory T-helper (Th) cells from the spleen and the mesenteric lymph node (MLN) showed a Th1-type response upon antigen stimulation in vitro. These results show that when VP6 is administered parenterally targeting DEC-205, it can induce protection at the intestinal level at a very low dose, and this protection may be Th1-type cell dependent.
Collapse
Affiliation(s)
- O Badillo-Godinez
- Laboratorio de Inmunidad Viral, Facultad de Medicina, UAEM, Cuernavaca, MOR, Mexico; Facultad de Ciencias, UAEM, Cuernavaca, MOR, Mexico
| | | | - T Plett-Torres
- CISEI-Instituto Nacional de Salud Publica, Cuernavaca, MOR, Mexico
| | | | | | - L Chihu-Amparan
- CISEI-Instituto Nacional de Salud Publica, Cuernavaca, MOR, Mexico
| | - M Maldonado-Gama
- CISEI-Instituto Nacional de Salud Publica, Cuernavaca, MOR, Mexico
| | - G Espino-Solis
- Instituto de Biotecnologia, UNAM, Cuernavaca, MOR, Mexico
| | - L C Bonifaz
- Unidad de Inmunohistoquimica, CMN, Hospital Siglo XXI, IMSS, Mexico, D.F., Mexico
| | - F Esquivel-Guadarrama
- Laboratorio de Inmunidad Viral, Facultad de Medicina, UAEM, Cuernavaca, MOR, Mexico.
| |
Collapse
|
24
|
Hodgins DC, Chattha K, Vlasova A, Parreño V, Corbeil LB, Renukaradhya GJ, Saif LJ. Mucosal Veterinary Vaccines. Mucosal Immunol 2015. [PMCID: PMC7149859 DOI: 10.1016/b978-0-12-415847-4.00068-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
Rodríguez M, Wood C, Sanchez-López R, Castro-Acosta RM, Ramírez OT, Palomares LA. Understanding internalization of rotavirus VP6 nanotubes by cells: towards a recombinant vaccine. Arch Virol 2014; 159:1005-15. [PMID: 24232915 DOI: 10.1007/s00705-013-1916-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 10/30/2013] [Indexed: 11/24/2022]
Abstract
Rotavirus VP6 nanotubes are an attractive option for a recombinant vaccine against rotavirus disease. Protection against rotavirus infection and an adjuvant effect have been observed upon immunization with VP6 nanotubes. However, little information exists on how VP6 nanotubes interact with cells and trigger an immune response. In this work, the interaction between VP6 nanotubes and different cell lines was characterized. VP6 nanotubes were not cytotoxic to any of the animal or human cell lines tested. Uptake of nanotubes into cells was cell-line-dependent, as only THP1 and J774 macrophage cells internalized them. Moreover, the size and spatial arrangement of VP6 assembled into nanotubes allowed their uptake by macrophages, as double-layered rotavirus-like particles also displaying VP6 in their surface were not taken up. The internalization of VP6 nanotubes was inhibited by methyl-β-cyclodextrin, but not by genistein, indicating that nanotube entry is specific, depends on the presence of cholesterol in the plasma membrane, and does not require the activity of tyrosine kinases. The information generated here expands our understanding of the interaction of protein nanotubes with cells, which is useful for the application of VP6 nanotubes as a vaccine.
Collapse
|
26
|
Comprehensive analysis of contributions from protein conformational stability and major histocompatibility complex class II-peptide binding affinity to CD4+ epitope immunogenicity in HIV-1 envelope glycoprotein. J Virol 2014; 88:9605-15. [PMID: 24920818 DOI: 10.1128/jvi.00789-14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED Helper T-cell epitope dominance in human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein gp120 is not adequately explained by peptide binding to major histocompatibility complex (MHC) proteins. Antigen processing potentially influences epitope dominance, but few, if any, studies have attempted to reconcile the influences of antigen processing and MHC protein binding for all helper T-cell epitopes of an antigen. Epitopes of gp120 identified in both humans and mice occur on the C-terminal flanks of flexible segments that are likely to be proteolytic cleavage sites. In this study, the influence of gp120 conformation on the dominance pattern in gp120 from HIV strain 89.6 was examined in CBA mice, whose MHC class II protein has one of the most well defined peptide-binding preferences. Only one of six dominant epitopes contained the most conserved element of the I-Ak binding motif, an aspartic acid. Destabilization of the gp120 conformation by deletion of single disulfide bonds preferentially enhanced responses to the cryptic I-Ak motif-containing sequences, as reported by T-cell proliferation or cytokine secretion. Conversely, inclusion of CpG in the adjuvant with gp120 enhanced responses to the dominant CD4+ T-cell epitopes. The gp120 destabilization affected secretion of some cytokines more than others, suggesting that antigen conformation could modulate T-cell functions through mechanisms of antigen processing. IMPORTANCE CD4+ helper T cells play an essential role in protection against HIV and other pathogens. Thus, the sites of helper T-cell recognition, the dominant epitopes, are targets for vaccine design; and the corresponding T cells may provide markers for monitoring infection and immunity. However, T-cell epitopes are difficult to identify and predict. It is also unclear whether CD4+ T cells specific for one epitope are more protective than T cells specific for other epitopes. This work shows that the three-dimensional (3D) structure of an HIV protein partially determines which epitopes are dominant, most likely by controlling the breakdown of HIV into peptides. Moreover, some types of signals from CD4+ T cells are affected by the HIV protein 3D structure; and thus the protectiveness of a particular peptide vaccine could be related to its location in the 3D structure.
Collapse
|
27
|
Vaccination with a single CD4 T cell peptide epitope from a Salmonella type III-secreted effector protein provides protection against lethal infection. Infect Immun 2014; 82:2424-33. [PMID: 24686055 DOI: 10.1128/iai.00052-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Salmonella infections affect millions worldwide and remain a significant cause of morbidity and mortality. It is known from mouse studies that CD4 T cells are essential mediators of immunity against Salmonella infection, yet it is not clear whether targeting CD4 T cell responses directly with peptide vaccines against Salmonella can be effective in combating infection. Additionally, it is not known whether T cell responses elicited against Salmonella secreted effector proteins can provide protective immunity against infection. In this study, we investigated both of these possibilities using prime-boost immunization of susceptible mice with a single CD4 T cell peptide epitope from Salmonella secreted effector protein I (SseI), a component of the Salmonella type III secretion system. This immunization conferred significant protection against lethal oral infection, equivalent to that conferred by whole heat-killed Salmonella bacteria. Surprisingly, a well-characterized T cell epitope from the flagellar protein FliC afforded no protection compared to immunization with an irrelevant control peptide. The protective response appeared to be most associated with polyfunctional CD4 T cells raised against the SseI peptide, since no antibodies were produced against any of the peptides and very little CD8 T cell response was observed. Overall, this study demonstrates that eliciting CD4 T cell responses against components of the Salmonella type III secretion system can contribute to protection against infection and should be considered in the design of future Salmonella subunit vaccines.
Collapse
|
28
|
Azevedo MP, Vlasova AN, Saif LJ. Human rotavirus virus-like particle vaccines evaluated in a neonatal gnotobiotic pig model of human rotavirus disease. Expert Rev Vaccines 2014; 12:169-81. [DOI: 10.1586/erv.13.3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
29
|
Gonzalez D, Rimondi A, Perez Aguirreburualde M, Mozgovoj M, Bellido D, Wigdorovitz A, Dus Santos M. Quantitation of cytokine gene expression by real time PCR in bovine milk and colostrum cells from cows immunized with a bovine rotavirus VP6 experimental vaccine. Res Vet Sci 2013; 95:703-8. [DOI: 10.1016/j.rvsc.2013.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Revised: 03/04/2013] [Accepted: 03/24/2013] [Indexed: 10/26/2022]
|
30
|
Tamminen K, Lappalainen S, Huhti L, Vesikari T, Blazevic V. Trivalent combination vaccine induces broad heterologous immune responses to norovirus and rotavirus in mice. PLoS One 2013; 8:e70409. [PMID: 23922988 PMCID: PMC3724941 DOI: 10.1371/journal.pone.0070409] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/18/2013] [Indexed: 01/14/2023] Open
Abstract
Rotavirus (RV) and norovirus (NoV) are the two major causes of viral gastroenteritis (GE) in children worldwide. We have developed an injectable vaccine design to prevent infection or GE induced with these enteric viruses. The trivalent combination vaccine consists of NoV capsid (VP1) derived virus-like particles (VLPs) of GI-3 and GII-4 representing the two major NoV genogroups and tubular RV recombinant VP6 (rVP6), the most conserved and abundant RV protein. Each component was produced in insect cells by a recombinant baculovirus expression system and combined in vitro. The vaccine components were administered intramuscularly to BALB/c mice either separately or in the trivalent combination. High levels of NoV and RV type specific serum IgGs with high avidity (>50%) as well as intestinal IgGs were detected in the immunized mice. Cross-reactive IgG antibodies were also elicited against heterologous NoV VLPs not used for immunization (GII-4 NO, GII-12 and GI-1 VLPs) and to different RVs from cell cultures. NoV-specific serum antibodies blocked binding of homologous and heterologous VLPs to the putative receptors, histo-blood group antigens, suggesting broad NoV neutralizing activity of the sera. Mucosal antibodies of mice immunized with the trivalent combination vaccine inhibited RV infection in vitro. In addition, cross-reactive T cell immune responses to NoV and RV-specific antigens were detected. All the responses were sustained for up to six months. No mutual inhibition of the components in the trivalent vaccine combination was observed. In conclusion, the NoV GI and GII VLPs combination induced broader cross-reactive and potentially neutralizing immune responses than either of the VLPs alone. Therefore, trivalent vaccine might induce protective immune responses to the vast majority of circulating NoV and RV genotypes.
Collapse
Affiliation(s)
- Kirsi Tamminen
- Vaccine Research Center, University of Tampere School of Medicine, Tampere, Finland
| | - Suvi Lappalainen
- Vaccine Research Center, University of Tampere School of Medicine, Tampere, Finland
| | - Leena Huhti
- Vaccine Research Center, University of Tampere School of Medicine, Tampere, Finland
| | - Timo Vesikari
- Vaccine Research Center, University of Tampere School of Medicine, Tampere, Finland
| | - Vesna Blazevic
- Vaccine Research Center, University of Tampere School of Medicine, Tampere, Finland
- * E-mail:
| |
Collapse
|
31
|
Uzri D, Greenberg HB. Characterization of rotavirus RNAs that activate innate immune signaling through the RIG-I-like receptors. PLoS One 2013; 8:e69825. [PMID: 23894547 PMCID: PMC3720929 DOI: 10.1371/journal.pone.0069825] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/17/2013] [Indexed: 12/24/2022] Open
Abstract
In mammalian cells, the first line of defense against viral pathogens is the innate immune response, which is characterized by induction of type I interferons (IFN) and other pro-inflammatory cytokines that establish an antiviral milieu both in infected cells and in neighboring uninfected cells. Rotavirus, a double-stranded RNA virus of the Reoviridae family, is the primary etiological agent of severe diarrhea in young children worldwide. Previous studies demonstrated that rotavirus replication induces a MAVS-dependent type I IFN response that involves both RIG-I and MDA5, two cytoplasmic viral RNA sensors. This study reports the isolation and characterization of rotavirus RNAs that activate IFN signaling. Using an in vitro approach with purified rotavirus double-layer particles, nascent single-stranded RNA (ssRNA) transcripts (termed in vitro ssRNA) were found to be potent IFN inducers. In addition, large RNAs isolated from rotavirus-infected cells six hours post-infection (termed in vivo 6 hr large RNAs), also activated IFN signaling, whereas a comparable large RNA fraction isolated from cells infected for only one hour lacked this stimulatory activity. Experiments using knockout murine embryonic fibroblasts showed that RIG-I is required for and MDA5 partly contributes to innate immune signaling by both in vitro ssRNA and in vivo 6 hr large RNAs. Enzymatic studies demonstrated that in vitro ssRNA and in vivo 6 hr large RNA samples contain uncapped RNAs with exposed 5’ phosphate groups. RNAs lacking 2’-O-methylated 5’ cap structures were also detected in the in vivo 6 hr large RNA sample. Taken together, our data provide strong evidence that the rotavirus VP3 enzyme, which encodes both guanylyltransferase and methyltransferase activities, is not completely efficient at either 5’ capping or 2’-O-methylation of the 5’ cap structures of viral transcripts, and in this way produces RNA patterns that activate innate immune signaling through the RIG-I-like receptors.
Collapse
Affiliation(s)
- Dina Uzri
- Departments of Medicine and Microbiology & Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Harry B. Greenberg
- Departments of Medicine and Microbiology & Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Lappalainen S, Tamminen K, Vesikari T, Blazevic V. Comparative immunogenicity in mice of rotavirus VP6 tubular structures and virus-like particles. Hum Vaccin Immunother 2013; 9:1991-2001. [PMID: 23777748 DOI: 10.4161/hv.25249] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Rotavirus (RV) is the most important cause of severe gastroenteritis in children worldwide. Current live RV vaccines are efficacious but show lower efficacy in developing countries, as well as a low risk of intussusception. This has led to the development of parenteral non-live candidate vaccines against RV. RV capsid VP6 protein is highly conserved and the most abundant RV protein forming highly immunogenic oligomeric structures with multivalent antigen expression. Both recombinant VP6 (rVP6) or double-layered (dl) 2/6-virus-like particles (VLPs), might be considered as the simplest RV subunit vaccine candidates. Human rVP6 protein and dl2/6-VLPs were produced in Sf9 insect cells by baculovirus expression system. Formation of rVP6 tubules and VLPs were confirmed by electron microscopy. BALB/c mice were immunized intramuscularly, and immune responses were analyzed. Both rVP6 and dl2/6-VLPs induced a balanced Th1-type and Th2-type response and high levels of serum IgG antibodies with cross-reactivity against different RV strains (Wa, SC2, BrB, 69M, L26, WC3, and RRV). In addition, mucosal VP6-specific IgG and IgA antibodies were detected in feces and vaginal washes (VW) of immunized animals. Importantly, VWs of immunized mice inhibited RV Wa and RRV infection in vitro. Immunization with either protein preparation induced a similar level of VP6-specific, interferon-γ secreting CD4(+) T cells in response to different RVs or the 18-mer peptide (AA 242-259), a VP6-specific CD4(+) T cell epitope. RV rVP6 and dl2/6-VLPs induced equally strong humoral and cellular responses against RV in mice and therefore, may be considered as non-live vaccine candidates against RV.
Collapse
Affiliation(s)
- Suvi Lappalainen
- Vaccine Research Center; University of Tampere Medical School; Tampere, Finland
| | | | | | | |
Collapse
|
33
|
Alteration of the thymic T cell repertoire by rotavirus infection is associated with delayed type 1 diabetes development in non-obese diabetic mice. PLoS One 2013; 8:e59182. [PMID: 23554993 PMCID: PMC3598695 DOI: 10.1371/journal.pone.0059182] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 02/12/2013] [Indexed: 12/12/2022] Open
Abstract
Rotaviruses are implicated as a viral trigger for the acceleration of type 1 diabetes in children. Infection of adult non-obese diabetic (NOD) mice with rotavirus strain RRV accelerates diabetes development, whereas RRV infection in infant NOD mice delays diabetes onset. In this study of infant mice, RRV titers and lymphocyte populations in the intestine, mesenteric lymph nodes (MLN) and thymus of NOD mice were compared with those in diabetes-resistant BALB/c and C57BL/6 mice. Enhanced intestinal RRV infection occurred in NOD mice compared with the other mouse strains. This was associated with increases in the frequency of CD8αβ TCRαβ intraepithelial lymphocytes, and their PD-L1 expression. Virus spread to the MLN and T cell numbers there also were greatest in NOD mice. Thymic RRV infection is shown here in all mouse strains, often in combination with alterations in T cell ontogeny. Infection lowered thymocyte numbers in infant NOD and C57BL/6 mice, whereas thymocyte production was unaltered overall in infant BALB/c mice. In the NOD mouse thymus, effector CD4+ T cell numbers were reduced by infection, whereas regulatory T cell numbers were maintained. It is proposed that maintenance of thymic regulatory T cell numbers may contribute to the increased suppression of inflammatory T cells in response to a strong stimulus observed in pancreatic lymph nodes of adult mice infected as infants. These findings show that rotavirus replication is enhanced in diabetes-prone mice, and provide evidence that thymic T cell alterations may contribute to the delayed diabetes onset following RRV infection.
Collapse
|
34
|
|
35
|
HSV-1 amplicon vectors launch the production of heterologous rotavirus-like particles and induce rotavirus-specific immune responses in mice. Mol Ther 2012; 20:1810-20. [PMID: 22713696 DOI: 10.1038/mt.2012.108] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Virus-like particles (VLPs) are promising vaccine candidates because they represent viral antigens in the authentic conformation of the virion and are therefore readily recognized by the immune system. As VLPs do not contain genetic material they are safer than attenuated virus vaccines. In this study, herpes simplex virus type 1 (HSV-1) amplicon vectors were constructed to coexpress the rotavirus (RV) structural genes VP2, VP6, and VP7 and were used as platforms to launch the production of RV-like particles (RVLPs) in vector-infected mammalian cells. Despite the observed splicing of VP6 RNA, full-length VP6 protein and RVLPs were efficiently produced. Intramuscular injection of mice with the amplicon vectors as a two-dose regimen without adjuvants resulted in RV-specific humoral immune responses and, most importantly, immunized mice were partially protected at the mucosal level from challenge with live wild-type (wt) RV. This work provides proof of principle for the application of HSV-1 amplicon vectors that mediate the efficient production of heterologous VLPs as genetic vaccines.
Collapse
|
36
|
Blazevic V, Lappalainen S, Nurminen K, Huhti L, Vesikari T. Norovirus VLPs and rotavirus VP6 protein as combined vaccine for childhood gastroenteritis. Vaccine 2011; 29:8126-33. [DOI: 10.1016/j.vaccine.2011.08.026] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/16/2011] [Accepted: 08/05/2011] [Indexed: 11/26/2022]
|
37
|
Abstract
A “Meeting on Upstream Rotavirus Vaccines and Emerging Vaccine Producers” was held at the World Health Organization in Geneva, Switzerland on March 28–30, 2006. The purpose was to discuss, evaluate, and weigh the importance of additional rotavirus vaccine candidates following the successful international licensure of rotavirus vaccines by two major pharmaceutical companies (GlaxoSmithKline and Merck) that had been in development for many years. Both licensed vaccines are composed of live rotaviruses that are delivered orally as have been all candidate rotavirus vaccines evaluated in humans. Each is built on the experience gained with previous candidates whose development had either been discontinued or, in the case of the previously licensed rhesus rotavirus reassortant vaccine (Rotashield), was withdrawn by its manufacturer after the discovery of a rare association with intussusception. Although which alternative candidate vaccines should be supported for development and where this should be done are controversial topics, there was general agreement expressed at the Geneva meeting that further development of alternative candidates is a high priority. This development will help insure that the most safe, effective and economic vaccines are available to children in Third World nations where the vast majority of the >600,000 deaths due to rotavirus occur each year. This review is intended to provide the history and present status of rotavirus vaccines as well as a perspective on the future development of candidate vaccines as a means of promulgating plans suggested at the Geneva meeting.
Collapse
Affiliation(s)
- Richard L Ward
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | | | | |
Collapse
|
38
|
Abstract
The immunological mediators that clear rotavirus antigenemia or viremia remain undefined. Immunodeficient mice and antibody transfer were used to test whether lymphocytes or rotavirus-specific serum antibodies are essential for resolving antigenemia. Clearance of antigenemia required lymphocytes, but neither T nor B lymphocytes were absolutely required. Transfer of convalescent-phase or nonneutralizing rotavirus-specific serum antibodies to the systemic compartment of severe-combined-immunodeficient (SCID) mice temporarily suppressed the onset or level of chronic rotavirus antigenemia. Our findings provide the first report demonstrating that clearance of rotavirus antigenemia and possibly viremia are mediated by multiple effector lymphocyte subsets and serum antibodies.
Collapse
|
39
|
Zhou H, Guo L, Wang M, Qu J, Zhao Z, Wang J, Hung T. Prime immunization with rotavirus VLP 2/6 followed by boosting with an adenovirus expressing VP6 induces protective immunization against rotavirus in mice. Virol J 2011; 8:3. [PMID: 21205330 PMCID: PMC3024956 DOI: 10.1186/1743-422x-8-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Accepted: 01/05/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Rotavirus (RV) is the main cause of severe gastroenteritis in children. An effective vaccination regime against RV can substantially reduce morbidity and mortality. Previous studies have demonstrated the efficacy of virus-like particles formed by RV VP2 and VP6 (VLP2/6), as well as that of recombinant adenovirus expressing RV VP6 (rAd), in eliciting protective immunities against RV. However, the efficacy of such prime-boost strategy, which incorporates VLP and rAd in inducing protective immunities against RV, has not been addressed. We assessed the immune effects of different regimens in mice, including rAd prime-VLP2/6 boost (rAd+VLP), VLP2/6 prime-rAd boost (VLP+rAd), rAd alone, and VLP alone. RESULTS Mice immunized with the VLP+rAd regimen elicit stronger humoral, mucosal, and cellular immune responses than those immunized with other regimens. RV challenging experiments showed that the highest reduction (92.9%) in viral shedding was achieved in the VLP+rAd group when compared with rAd+VLP (25%), VLP alone (75%), or rAd alone (40%) treatment groups. The reduction in RV shedding in mice correlated with fecal IgG (r = 0.95773, P = 0.04227) and IgA (r = 0.96137, P = 0.038663). CONCLUSIONS A VLP2/6 prime-rAd boost regimen is effective in conferring immunoprotection against RV challenge in mice. This finding may lay the groundwork for an alternative strategy in novel RV vaccine development.
Collapse
Affiliation(s)
- Hongli Zhou
- State Key Laboratory for Molecular Virology and Genetic Engineering, Institute of Pathogen Biology, Chinese Academy Medical Sciences & Peking Union Medical College, Dong Dan San Tiao, Beijing 100730, PR China
| | | | | | | | | | | | | |
Collapse
|
40
|
Mendicino M, Ramsoondar J, Phelps C, Vaught T, Ball S, LeRoith T, Monahan J, Chen S, Dandro A, Boone J, Jobst P, Vance A, Wertz N, Bergman Z, Sun XZ, Polejaeva I, Butler J, Dai Y, Ayares D, Wells K. Generation of antibody- and B cell-deficient pigs by targeted disruption of the J-region gene segment of the heavy chain locus. Transgenic Res 2010; 20:625-41. [PMID: 20872248 PMCID: PMC7089184 DOI: 10.1007/s11248-010-9444-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 09/13/2010] [Indexed: 01/22/2023]
Abstract
A poly(A)-trap gene targeting strategy was used to disrupt the single functional heavy chain (HC) joining region (JH) of swine in primary fibroblasts. Genetically modified piglets were then generated via somatic cell nuclear transfer (SCNT) and bred to yield litters comprising JH wild-type littermate (+/+), JH heterozygous knockout (±) and JH homozygous knockout (−/−) piglets in the expected Mendelian ratio of 1:2:1. There are only two other targeted loci previously published in swine, and this is the first successful poly(A)-trap strategy ever published in a livestock species. In either blood or secondary lymphoid tissues, flow cytometry, RT-PCR and ELISA detected no circulating IgM+ B cells, and no transcription or secretion of immunoglobulin (Ig) isotypes, respectively in JH −/− pigs. Histochemical and immunohistochemical (IHC) studies failed to detect lymph node (LN) follicles or CD79α+ B cells, respectively in JH −/− pigs. T cell receptor (TCR)β transcription and T cells were detected in JH −/− pigs. When reared conventionally, JH −/− pigs succumbed to bacterial infections after weaning. These antibody (Ab)- and B cell-deficient pigs have significant value as models for both veterinary and human research to discriminate cellular and humoral protective immunity to infectious agents. Thus, these pigs may aid in vaccine development for infectious agents such as the pandemic porcine reproductive and respiratory syndrome virus (PRRSV) and H1N1 swine flu. These pigs are also a first significant step towards generating a pig that expresses fully human, antigen-specific polyclonal Ab to target numerous incurable infectious diseases with high unmet clinical need.
Collapse
Affiliation(s)
- M Mendicino
- Revivicor, Inc., 1700 Kraft Drive, Blacksburg, VA 24060, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Development of a Bacillus subtilis-based rotavirus vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1647-55. [PMID: 20810679 DOI: 10.1128/cvi.00135-10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Bacillus subtilis vaccine strains engineered to express either group A bovine or murine rotavirus VP6 were tested in adult mice for their ability to induce immune responses and provide protection against rotavirus challenge. Mice were inoculated intranasally with spores or vegetative cells of the recombinant strains of B. subtilis. To enhance mucosal immunity, whole cholera toxin (CT) or a mutant form (R192G) of Escherichia coli heat-labile toxin (mLT) were included as adjuvants. To evaluate vaccine efficacy, the immunized mice were challenged orally with EDIM EW murine rotavirus and monitored daily for 7 days for virus shedding in feces. Mice immunized with either VP6 spore or VP6 vegetative cell vaccines raised serum anti-VP6 IgG enzyme-linked immunosorbent assay (ELISA) titers, whereas only the VP6 spore vaccines generated fecal anti-VP6 IgA ELISA titers. Mice in groups that were immunized with VP6 spore vaccines plus CT or mLT showed significant reductions in virus shedding, whereas the groups of mice immunized with VP6 vegetative cell vaccines showed no difference in virus shedding compared with mice immunized with control spores or cells. These results demonstrate that intranasal inoculation with B. subtilis spore-based rotavirus vaccines is effective in generating protective immunity against rotavirus challenge in mice.
Collapse
|
42
|
Wu CR, Jiang X, He ST, Yang B, Hu JZ, Cai R. Effects of QWBZP on T-cell subsets and their cytokines in intestinal mucosa of HRV infection suckling mice. JOURNAL OF ETHNOPHARMACOLOGY 2010; 131:130-4. [PMID: 20600774 DOI: 10.1016/j.jep.2010.06.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 06/05/2010] [Accepted: 06/10/2010] [Indexed: 05/04/2023]
Abstract
AIM OF THIS STUDY Qiwei Baizhu Powder (QWBZP) is a traditional herbal prescription that has been used traditionally for the treatment of infantile diarrhea, including the infantile diarrhea caused by Human Rotavirus (HRV). In this study, we investigated the pharmacological activity of QWBZP extract. MATERIALS AND METHODS NIH suckling mice with HRV induced diarrhea were used. Density of CD3(+), CD4(+) and CD8(+) T cells, mRNA expression of IL-2, IFN-gamma, IL-4 and IL-10 in intestinal mucosa epithelial cells were assayed. RESULTS QWBZP extract promoted the expressions of mRNA of IL-2, IL-4, IL-10 and IFN-gamma in intestinal mucosa epithelial cells. Also, we found that the density of CD8(+) cells in intestinal mucosa epithelial cells was significantly lower in QWBZP group than in Model group, while the density of CD8(+) cells was significantly higher in QWBZP group than in Model group. CONCLUSION These data suggest that QWBZP extract may exhibit antiviral effects through modulating the densities of T-cell subsets and the expressions of their cytokines in small intestinal mucosa epithelial cells.
Collapse
Affiliation(s)
- Can-rong Wu
- Department of Etiology and Immunology, Hunan University of TCM, Shaoshan Road, Changsha, Hunan 410007, PR China.
| | | | | | | | | | | |
Collapse
|
43
|
Gonzalez DD, Mozgovoj MV, Bellido D, Rodriguez DV, Fernandez FM, Wigdorovitz A, Parreño VG, Dus Santos MJ. Evaluation of a bovine rotavirus VP6 vaccine efficacy in the calf model of infection and disease. Vet Immunol Immunopathol 2010; 137:155-60. [PMID: 20546933 DOI: 10.1016/j.vetimm.2010.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 04/15/2010] [Accepted: 04/23/2010] [Indexed: 12/31/2022]
Abstract
Group A bovine rotavirus (BRV) is the major cause of acute viral gastroenteritis in neonatal calves worldwide. Due to the early susceptibility to the infection prevention strategies are based on the improvement of passive immunity levels through cow vaccination in the last third of gestation. The major capsid antigen (VP6) of BRV is the most immunogenic viral protein and it is highly conserved among group A BRV. In this work, VP6 protein from BRV C-486 strain (P[1]G6) was expressed in insect cells using the baculovirus expression vector system. Recombinant VP6 was used to immunize cows and vaccine's efficacy was assessed in a colostrum-deprived calf model of BRV infection and disease. Immune colostrum pool was generated using first and second milking of the immunized cows. Calves receiving one dose of immune colostrum within the first 6h of life, or colostrum-deprived calves were orally inoculated with virulent BRV at 2 days of age. The animals were monitored for diarrhea, virus shedding and isotype-specific antibodies responses to BRV in both feces and serum. Calves receiving VP6-immune colostrum showed a reduction of both diarrhea and virus shedding (in terms of viral titer and excretion period) in comparison with the colostrum-deprived calves.
Collapse
Affiliation(s)
- Diego Daniel Gonzalez
- Instituto de Virología, CICVyA, INTA, Las Cabañas y de los Reseros s/n, Castelar, 1686 Hurlingham, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Variation in antagonism of the interferon response to rotavirus NSP1 results in differential infectivity in mouse embryonic fibroblasts. J Virol 2009; 83:6987-94. [PMID: 19420080 DOI: 10.1128/jvi.00585-09] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Rotavirus NSP1 has been shown to function as an E3 ubiquitin ligase that mediates proteasome-dependent degradation of interferon (IFN) regulatory factors (IRF), including IRF3, -5, and -7, and suppresses the cellular type I IFN response. However, the effect of rotavirus NSP1 on viral replication is not well defined. Prior studies used genetic analysis of selected reassortants to link NSP1 with host range restriction in the mouse, suggesting that homologous and heterologous rotaviruses might use their different abilities to antagonize the IFN response as the basis of their host tropisms. Using a mouse embryonic fibroblast (MEF) model, we demonstrate that heterologous bovine (UK and NCDV) and porcine (OSU) rotaviruses fail to effectively degrade cellular IRF3, resulting in IRF3 activation and beta IFN (IFN-beta) secretion. As a consequence of this failure, replication of these viruses is severely restricted in IFN-competent wild-type, but not in IFN-deficient (IFN-alpha/beta/gamma receptor- or STAT1-deficient) MEFs. On the other hand, homologous murine rotaviruses (ETD or EHP) or the heterologous simian rotavirus (rhesus rotavirus [RRV]) efficiently degrade cellular IRF3, diminish IRF3 activation and IFN-beta secretion and are not replication restricted in wild-type MEFs. Genetic reassortant analysis between UK and RRV maps the distinctive phenotypes of IFN antagonism and growth restriction in wild-type MEFs to NSP1. Therefore, there is a direct relationship between the replication efficiencies of different rotavirus strains in MEFs and strain-related variations in NSP1-mediated antagonism of the type I IFN response.
Collapse
|
45
|
Verhoeven D, Teijaro JR, Farber DL. Heterogeneous memory T cells in antiviral immunity and immunopathology. Viral Immunol 2008; 21:99-113. [PMID: 18476772 DOI: 10.1089/vim.2008.0002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Memory T cells are generated following an initial viral infection, and have the potential for mediating robust protective immunity to viral re-challenge due to their rapid and enhanced functional responses. In recent years, it has become clear that the memory T cell response to most viruses is remarkably diverse in phenotype, function, and tissue distribution, and can undergo dynamic changes during its long-term maintenance in vivo. However, the role of this variegation and compartmentalizationof memory T cells in protective immunity to viruses remains unclear. In this review,we discuss the diverse features of memory T cells that can delineate different subsets, the characteristics of memory T cells thus far identified to promote protective immune responses, and how the heterogeneous nature of memory T cells may also promote immunopathology during antiviral responses. We propose that given the profound heterogeneity of memory T cells, regulation of memory T cells during secondary responses could focus the response to participation of specific subsets,and/or inhibit memory T-cell subsets and functions that can lead to immunopathology.
Collapse
Affiliation(s)
- David Verhoeven
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
46
|
Feng N, Kim B, Fenaux M, Nguyen H, Vo P, Omary MB, Greenberg HB. Role of interferon in homologous and heterologous rotavirus infection in the intestines and extraintestinal organs of suckling mice. J Virol 2008; 82:7578-90. [PMID: 18495762 PMCID: PMC2493311 DOI: 10.1128/jvi.00391-08] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Accepted: 05/12/2008] [Indexed: 02/05/2023] Open
Abstract
Recent studies demonstrated that viremia and extraintestinal rotavirus infection are common in acutely infected humans and animals, while systemic diseases appear to be rare. Intraperitoneal infection of newborn mice with rhesus rotavirus (RRV) results in biliary atresia (BA), and this condition is influenced by the host interferon response. We studied orally inoculated 5-day-old suckling mice that were deficient in interferon (IFN) signaling to evaluate the role of interferon on the outcome of local and systemic infection after enteric inoculation. We found that systemic replication of RRV, but not murine rotavirus strain EC, was greatly enhanced in IFN-alpha/beta and IFN-gamma receptor double-knockout (KO) or STAT1 KO mice but not in mice deficient in B- or T-cell immunity. The enhanced replication of RRV was associated with a lethal hepatitis, pancreatitis, and BA, while no systemic disease was observed in strain EC-infected interferon-deficient mice. In IFN-alpha/beta receptor KO mice the extraintestinal infection and systemic disease were only moderately increased, while RRV infection was not augmented and systemic disease was not present in IFN-gamma receptor KO mice. The increase of systemic infection in IFN-deficient mice was also observed during simian strain SA11 infection but not following bovine NCDV, porcine OSU, or murine strain EW infection. Our data indicate that the requirements for the interferon system to inhibit intestinal and extraintestinal viral replication in suckling mice vary among different heterologous and homologous rotavirus strains, and this variation is associated with lethal systemic disease.
Collapse
Affiliation(s)
- N Feng
- Stanford University, Stanford, California, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Istrate C, Hinkula J, Charpilienne A, Poncet D, Cohen J, Svensson L, Johansen K. Parenteral administration of RF 8-2/6/7 rotavirus-like particles in a one-dose regimen induce protective immunity in mice. Vaccine 2008; 26:4594-601. [PMID: 18588935 DOI: 10.1016/j.vaccine.2008.05.089] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Revised: 05/21/2008] [Accepted: 05/25/2008] [Indexed: 10/21/2022]
Abstract
Rotavirus virus-like particles (RV-VLPs) represent a novel strategy for development of a rotavirus subunit vaccine. In this study, RF 8-2/6/7-VLPs with rotavirus VP8 protein (amino acid 1-241 of VP4) fused to the amino terminal end of a truncated VP2, were evaluated for their immunogenic and protective properties. A single intramuscular dose of, either 2 or 20 microg, RF 8-2/6/7-VLPs alone or combined with a potent adjuvant poly[di(carboxylatophenoxy)]phosphazene] (PCPP) induced rotavirus-specific serum IgG and IgA, fecal IgG titers that were enhanced 5-90-fold by adjuvant. Passive protective immunity was achieved in offspring to dams vaccinated with 2 and 20 microg RV-VLPs in presence of adjuvant and 20 microg RV-VLP without adjuvant.
Collapse
Affiliation(s)
- Claudia Istrate
- Instituto de Biologia Experimental e Tecnologica and Instituto de Tecnologia Quimica e Biologica, SE-171 76 Oeiras, Portugal
| | | | | | | | | | | | | |
Collapse
|
48
|
Yuan L, Wen K, Azevedo MSP, Gonzalez AM, Zhang W, Saif LJ. Virus-specific intestinal IFN-gamma producing T cell responses induced by human rotavirus infection and vaccines are correlated with protection against rotavirus diarrhea in gnotobiotic pigs. Vaccine 2008; 26:3322-31. [PMID: 18456375 DOI: 10.1016/j.vaccine.2008.03.085] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 03/26/2008] [Accepted: 03/31/2008] [Indexed: 11/17/2022]
Abstract
We examined rotavirus-specific IFN-gamma producing CD4+, CD8+ and CD4+CD8+ T cell responses in gnotobiotic pigs infected with a virulent human rotavirus (VirHRV) or vaccinated with an attenuated (Att) HRV vaccine (AttHRV3x or AttHRV2x) or an AttHRV oral priming and 2/6-virus-like particle (VLP) intranasal boosting (AttHRV-2/6VLP) regimen. In VirHRV infected pigs, HRV-specific IFN-gamma producing T cells reside primarily in ileum. AttHRV-2/6VLP induced similar frequencies of intestinal IFN-gamma producing T cells as the VirHRV, whereas AttHRV3x or 2x vaccines were less effective. Protection rates against rotavirus diarrhea upon VirHRV challenge significantly correlated (r=0.97-1.0, p<0.005) with frequencies of intestinal IFN-gamma producing T cells, suggesting their role in protective immunity.
Collapse
Affiliation(s)
- Lijuan Yuan
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH 44691, USA.
| | | | | | | | | | | |
Collapse
|
49
|
McNeal MM, Stone SC, Basu M, Clements JD, Choi AHC, Ward RL. IFN-gamma is the only anti-rotavirus cytokine found after in vitro stimulation of memory CD4+ T cells from mice immunized with a chimeric VP6 protein. Viral Immunol 2008; 20:571-84. [PMID: 18158731 DOI: 10.1089/vim.2007.0055] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
CD4+ T cells are the only lymphocytes required for protection of mice against rotavirus shedding after mucosal immunization with chimeric VP6 (MBP::VP6) and the adjuvant LT(R192G). One possible effector of protection is CD4+ T-cell cytokines. To determine if memory CD4+ T cells of immunized mice produce cytokines with direct anti-rotavirus activity, an in vitro infection model was developed using mouse CMT-93 cells and rhesus rotavirus (RRV). Spleen and lamina propria (LP) cells, as well as purified splenic CD4T cells obtained after intranasal immunization of BALB/c mice with MBP::VP6/LT(R192G) released large quantities of two cytokines (IL-17 and IFN-gamma) into cell supernatants when stimulated with MBP::VP6. Production of these same cytokines is rapidly upregulated in intestinal lymphocytes after rotavirus inoculation of immunized mice. IL-17 pretreatment of CMT-93 cells had no effect on subsequent RRV replication, but IFN-gamma was the most potent inhibitor within a panel of nine cytokines tested. Supernatants obtained after in vitro stimulation of splenic CD4+ T cells of immunized mice had high levels of anti-RRV activity and their pretreatment with mAb against IFN-gamma caused essentially complete loss of activity. Thus, IFN-gamma was the only cytokine identified in stimulated CD4+ T cells from immunized mice that directly inhibited rotavirus replication.
Collapse
Affiliation(s)
- Monica M McNeal
- Division of Infectious Diseases, Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Zhao W, Pahar B, Sestak K. Identification of Rotavirus VP6-Specific CD4+ T Cell Epitopes in a G1P[8] Human Rotavirus-Infected Rhesus Macaque. Virology (Auckl) 2008; 1:9-15. [PMID: 20401320 DOI: 10.4137/vrt.s563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A non-human primate model was used to evaluate its potential for identification of rotavirus viral protein 6 (VP6) CD4+ T cell epitopes. Four juvenile rhesus macaques were inoculated with a mixed inoculum (G1P[8] and G9P[8]) of human rotaviruses. Infection accompanied by G1P[8] shedding was achieved in the two macaques that had no rotavirus immunoglobulin A (IgA) in plasma. To measure the interferon gamma (IFN-γ) and tumor necrosis factor (TNF) anti-viral cytokines produced by peripheral CD4+ cells that recognize VP6 epitopes, whole blood cells from one infected macaque were stimulated in vitro with VP6 peptides. Stimulation with peptide pools derived from the simian rotavirus VP6(161-395) region revealed reactivity of CD4+ T cells with the VP6(281-331) domain. A VP6(301-315) region was identified as the epitope responsible for IFN-γ production while a broader VP6(293-327) domain was linked to TNF production. These results suggest that human rotavirus-infected macaques can be used for identification of additional epitopes and domains to address specific questions related to the development of pediatric vaccines.
Collapse
Affiliation(s)
- Wei Zhao
- Tulane National Primate Research Center, Covington, LA, U.S.A
| | | | | |
Collapse
|