1
|
Wang P, Fredj Z, Zhang H, Rong G, Bian S, Sawan M. Blocking Superantigen-Mediated Diseases: Challenges and Future Trends. J Immunol Res 2024; 2024:2313062. [PMID: 38268531 PMCID: PMC10807946 DOI: 10.1155/2024/2313062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/15/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024] Open
Abstract
Superantigens are virulence factors secreted by microorganisms that can cause various immune diseases, such as overactivating the immune system, resulting in cytokine storms, rheumatoid arthritis, and multiple sclerosis. Some studies have demonstrated that superantigens do not require intracellular processing and instated bind as intact proteins to the antigen-binding groove of major histocompatibility complex II on antigen-presenting cells, resulting in the activation of T cells with different T-cell receptor Vβ and subsequent overstimulation. To combat superantigen-mediated diseases, researchers have employed different approaches, such as antibodies and simulated peptides. However, due to the complex nature of superantigens, these approaches have not been entirely successful in achieving optimal therapeutic outcomes. CD28 interacts with members of the B7 molecule family to activate T cells. Its mimicking peptide has been suggested as a potential candidate to block superantigens, but it can lead to reduced T-cell activity while increasing the host's infection risk. Thus, this review focuses on the use of drug delivery methods to accurately target and block superantigens, while reducing the adverse effects associated with CD28 mimic peptides. We believe that this method has the potential to provide an effective and safe therapeutic strategy for superantigen-mediated diseases.
Collapse
Affiliation(s)
- Pengbo Wang
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Zina Fredj
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Hongyong Zhang
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Guoguang Rong
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Sumin Bian
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Mohamad Sawan
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| |
Collapse
|
2
|
Jensen KJ, Tolstrup LK, Knobel DL, Aaby P, Jungersen G, Larsen LE, Kristensen CS, Benn CS. Non-specific effects of maternal and offspring rabies vaccination on mortality and antibiotic use in a Danish pig herd: A randomized trial. Vaccine 2022; 40:1665-1673. [PMID: 33840563 DOI: 10.1016/j.vaccine.2021.03.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Human non-live vaccines have been associated with detrimental non-specific effects (NSE), particularly in females. A large trial found 2-fold increased overall mortality in girls receiving a new malaria vaccine compared to the rabies vaccine used as a coontrol; a beneficial NSE of the rabies vaccine was proposed. Conversely, in dogs increased mortality was seen in females but not males following rabies vaccination of puppies born to immunized mothers. We investigated NSE of non-live rabies vaccine in piglets and the potential modifying effect of maternal priming with rabies vaccine. METHODS In a Danish herd of commercial rabies virus-free pigs, 575 pregnant sows (2-3 weeks before scheduled farrowing) and 5747 of their offspring (median 6-day-old) were allocated (1:1) to non-live rabies vaccine (Versiguard rabies vet) or no rabies vaccine. Outcomes were overall mortality and antibiotic treatment until departure from the nursery (approximately age 12 weeks/30 kgs). RESULTS Until weaning, overall offspring mortality was 2.2% (127 piglets died, rabies vaccine: n = 69; control: n = 58), the proportion ratio (PR) being 1.19 (95% confidence interval: 0.84-1.68). Until end of follow-up, mortality was 4.1% (233, rabies vaccine: n = 115; control = 118, PR: 0.97 (0.76-1.25)). Prior sow rabies vaccination did not affect piglet mortality. For mortality as well as risk of antibiotic treatment before weaning, there was indication of a beneficial effect of rabies vaccine in female piglets, but a negative effect in (castrated) male piglets from rabies-naïve sows. Prior sow vaccination significantly modified the vaccine effect estimate in female piglets toward a detrimental effect of rabies vaccine on treatment risk. These effects had waned by 12 weeks of age. CONCLUSION The study did not support the hypothesized beneficial NSE of rabies vaccine. Although under-powered for subgroup analyses, the study indicated effect modification by sex and maternal vaccination. Results could be different in a herd with higher mortality and infectious burden.
Collapse
Affiliation(s)
- Kristoffer Jarlov Jensen
- Bandim Health Project, Department of Clinical Research, University of Southern Denmark, Copenhagen, Denmark; Copenhagen Phase IV Unit, Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark; Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark.
| | | | - Darryn L Knobel
- Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis; Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
| | - Peter Aaby
- Bandim Health Project, INDEPTH Network, Bissau, Guinea-Bissau
| | - Gregers Jungersen
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Lars Erik Larsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Christine Stabell Benn
- Bandim Health Project, Department of Clinical Research, University of Southern Denmark, Copenhagen, Denmark; Danish Institute for Advanced Study, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
3
|
Martinón-Torres F, Bosch X, Rappuoli R, Ladhani S, Redondo E, Vesikari T, García-Sastre A, Rivero-Calle I, Gómez-Rial J, Salas A, Martín C, Finn A, Butler R. TIPICO IX: report of the 9 th interactive infectious disease workshop on infectious diseases and vaccines. Hum Vaccin Immunother 2019; 15:2405-2415. [PMID: 31158041 PMCID: PMC6816368 DOI: 10.1080/21645515.2019.1609823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Ninth Interactive Infectious Disease workshop TIPICO was held on November 22–23, 2018, in Santiago de Compostela, Spain. This 2-day academic experience addressed current and topical issues in the field of infectious diseases and vaccination. Summary findings of the meeting include: cervical cancer elimination will be possible in the future, thanks to the implementation of global vaccination action plans in combination with appropriate screening interventions. The introduction of appropriate immunization programs is key to maintain the success of current effective vaccines such as those against meningococcal disease or rotavirus infection. Additionally, reduced dose schedules might improve the efficiency of some vaccines (i.e., PCV13). New vaccines to improve current preventive alternatives are under development (e.g., against tuberculosis or influenza virus), while others to protect against infectious diseases with no current available vaccines (e.g., enterovirus, parechovirus and flaviviruses) need to be developed. Vaccinomics will be fundamental in this process, while infectomics will allow the application of precision medicine. Further research is also required to understand the impact of heterologous vaccine effects. Finally, vaccination requires education at all levels (individuals, community, healthcare professionals) to ensure its success by helping to overcome major barriers such as vaccine hesitancy and false contraindications.
Collapse
Affiliation(s)
- Federico Martinón-Torres
- Translational Paediatrics and Infectious Diseases, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela , Santiago de Compostela , Spain.,Genetics, Vaccines and Infections Research group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidad de Santiago de Compostela , Santiago de Compostela , Spain
| | - Xavier Bosch
- Cancer Epidemiology Research Programme (e-oncología), Catalan Institute of Oncology, L'Hospitalet de Llobregat , Barcelona , Spain.,Cancer Prevention and Palliative Care Program, IDIBELL, L'Hospitalet de Llobregat , Barcelona , Spain
| | - Rino Rappuoli
- R&D Centre, GlaxoSmithKline , Siena , Italy.,Department of Medicine, Imperial College London , London , UK
| | - Shamez Ladhani
- Immunisation Department, Public Health England , London , UK
| | - Esther Redondo
- International Vaccination Center of Madrid , Madrid , Spain.,Grupo de Actividades Preventivas y Salud Pública SEMERGEN , Madrid , Spain
| | - Timo Vesikari
- Faculty of Medicine and Life Sciences, Vaccine Research Center, University of Tampere , Tampere , Finland
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York , NY , USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai , New York , NY , USA.,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Irene Rivero-Calle
- Translational Paediatrics and Infectious Diseases, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela , Santiago de Compostela , Spain.,Genetics, Vaccines and Infections Research group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidad de Santiago de Compostela , Santiago de Compostela , Spain
| | - José Gómez-Rial
- Genetics, Vaccines and Infections Research group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidad de Santiago de Compostela , Santiago de Compostela , Spain
| | - Antonio Salas
- Genetics, Vaccines and Infections Research group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Universidad de Santiago de Compostela , Santiago de Compostela , Spain.,Unidade de Xenética, Instituto de Ciencias Forenses (INCIFOR), Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, of the Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago (SERGAS) , Galicia , Spain
| | - Carlos Martín
- Faculty of Medicine, Microbiology Department, University of Zaragoza , Zaragoza , Spain.,CIBER of Respiratory Diseases, Instituto de Salud Carlos III , Madrid , Spain
| | - Adam Finn
- Bristol Children's Vaccine Centre, Schools of Cellular and Molecular Medicine and Population Health Sciences, University of Bristol , Bristol , UK
| | | |
Collapse
|
4
|
Gessner BD, Knobel DL, Conan A, Finn A. Could the RTS,S/AS01 meningitis safety signal really be a protective effect of rabies vaccine? Vaccine 2017; 35:716-721. [PMID: 28065475 DOI: 10.1016/j.vaccine.2016.12.067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/18/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
The RTS,S/AS01 malaria vaccine has been associated with meningitis and cerebral malaria safety signals. Key characteristics of the meningitis signal include presence, in the 5-17month but not the 6-12week age group, of delayed and variable meningitis onset after vaccination, and multiple etiologies. For both meningitis and cerebral malaria, the 5-17month old age group control arm had abnormally low incidences while other arms in both age groups had meningitis and cerebral malaria incidences similar to background rates. No single hypothesis postulating an adverse effect from RTS,S/AS01 unites these observations. Unlike the 6-12week group, the control population in the 5-17month old age group received rabies vaccine. This raises the possibility that non-specific rabies vaccine effects had a protective effect against central nervous system infection, a hypothesis consistent with the epidemiologic data. The lack of a confirmed biologic mechanism for such an effect emphasizes the need for additional studies.
Collapse
Affiliation(s)
| | - Darryn L Knobel
- Ross University School of Veterinary Medicine, Island Main Road, West Farm, Saint Kitts and Nevis
| | - Anne Conan
- Ross University School of Veterinary Medicine, Island Main Road, West Farm, Saint Kitts and Nevis
| | - Adam Finn
- Bristol Children's Vaccine Centre, University of Bristol, United Kingdom
| |
Collapse
|
5
|
Zhang J, Ruan X, Zan J, Zheng X, Yan Y, Liao M, Zhou J. Efficient generation of monoclonal antibodies against major structural proteins of rabies virus with suckling mouse brain antigen. Monoclon Antib Immunodiagn Immunother 2014; 33:94-100. [PMID: 24746150 DOI: 10.1089/mab.2013.0087] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The rabies virus is a neurotropic virus that causes fatal disease in humans and animals. However, not all commercial antibodies against rabies virus (RABV) structural proteins are generally available, and production of high-quality monoclonal antibodies (MAbs) requires high purification of virus particles and special facilities and is time-consuming. By using RABV-infected suckling mouse brain as antigens in this study, 11 hybridoma cells secreting MAbs against RABV were obtained, which showed strong reactivity with RABV-infected Vero cells in immunofluorescence assay. Among the 11 MAbs, three MAbs (1B11, 1C8, and 8H12) showed a neutralizing effect to RABV, while MAb 4B7 recognized the recombinant nucleoprotein (N) of RABV expressed in Vero cells; seven MAbs (1H3, 3H7, 4E7, 4G3, 5A10, 6C9, and 7B3) reacted specifically with phosphoprotein (P) of RABV. The MAbs developed in this study will be useful in establishing a diagnostic test and study on the interactions between RABV and its host.
Collapse
Affiliation(s)
- Jinyang Zhang
- 1 Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang University , Hangzhou, P.R. China
| | | | | | | | | | | | | |
Collapse
|
6
|
Orange JS, Ballow M, Stiehm ER, Ballas ZK, Chinen J, De La Morena M, Kumararatne D, Harville TO, Hesterberg P, Koleilat M, McGhee S, Perez EE, Raasch J, Scherzer R, Schroeder H, Seroogy C, Huissoon A, Sorensen RU, Katial R. Use and interpretation of diagnostic vaccination in primary immunodeficiency: a working group report of the Basic and Clinical Immunology Interest Section of the American Academy of Allergy, Asthma & Immunology. J Allergy Clin Immunol 2012; 130:S1-24. [PMID: 22935624 DOI: 10.1016/j.jaci.2012.07.002] [Citation(s) in RCA: 325] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 07/02/2012] [Accepted: 07/03/2012] [Indexed: 12/24/2022]
Abstract
A major diagnostic intervention in the consideration of many patients suspected to have primary immunodeficiency diseases (PIDDs) is the application and interpretation of vaccination. Specifically, the antibody response to antigenic challenge with vaccines can provide substantive insight into the status of human immune function. There are numerous vaccines that are commonly used in healthy individuals, as well as others that are available for specialized applications. Both can potentially be used to facilitate consideration of PIDD. However, the application of vaccines and interpretation of antibody responses in this context are complex. These rely on consideration of numerous existing specific studies, interpolation of data from healthy populations, current diagnostic guidelines, and expert subspecialist practice. This document represents an attempt of a working group of the American Academy of Allergy, Asthma & Immunology to provide further guidance and synthesis in this use of vaccination for diagnostic purposes in consideration of PIDD, as well as to identify key areas for further research.
Collapse
Affiliation(s)
- Jordan S Orange
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Development of combined vaccines for rabies and immunocontraception. Vaccine 2010; 27:7202-9. [PMID: 19925954 DOI: 10.1016/j.vaccine.2009.09.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 09/03/2009] [Indexed: 11/24/2022]
Abstract
Rabies prevention and appropriate population management of free-ranging animals have an important role to play in the eventual elimination of rabies in dogs. An effective sterilant based on rabies vaccines has the potential to create a supportive measure of public acceptability and to reduce associated clinic visit costs. We inserted the coding sequence of gonadotropin-releasing hormone (GnRH) into different locations within the rabies virus ERA glycoprotein (G) gene, and demonstrated that the amino terminus (N), antigenic site IIa, and the junction between the ecto- and cytoplasmic domains (C) of the G were suitable sites for GnRH insertion. The rescued recombinant rabies viruses ERA-N-GnRH and ERA-C-GnRH grew as well as the parental ERA virus, reaching 1x10(9)ffu/ml in cell culture. Insertion and expression of the GnRH were stable in the viruses after multiple passages in vitro. To increase immunogenicity of the GnRH peptide, two copies of GnRH, aligned in tandem, were fused to the N terminus of the G. The recombinant rabies virus ERA-N-2GnRH was recovered and grown to high titers in cell culture. All GnRH-carrying rabies viruses induced antibodies against GnRH in immunized mice and protected 100% of the animals after rabies virus challenge. The recombinant viruses reacted strongly with the serum from a GonaCon-immunized animal. The GnRH-carrying rabies viruses have significant potential in rabies and animal population control.
Collapse
|
8
|
Loza-Rubio E, Molina-Güarneros J, Montaño-Hirose JA. Nucleocapsid of rabies virus improve immune response of an inactivated avian influenza vaccine. Vet Res Commun 2009; 33:589-95. [DOI: 10.1007/s11259-009-9206-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Accepted: 01/12/2009] [Indexed: 12/01/2022]
|
9
|
Rojas-Anaya E, Loza-Rubio E, Olivera-Flores MT, Gomez-Lim M. Expression of rabies virus G protein in carrots (Daucus carota). Transgenic Res 2009; 18:911-9. [PMID: 19479338 DOI: 10.1007/s11248-009-9278-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Accepted: 04/30/2009] [Indexed: 11/26/2022]
Abstract
Antigens derived from various pathogens can readily be synthesized at high levels in plants in their authentic forms. Such antigens administered orally can induce an immune response and, in some cases, result in protection against a subsequent challenge. We here report the expression of rabies virus G protein into carrots. The G gene was subcloned into the pUCpSSrabG vector and then used to transform carrot embryogenic cells by particle bombardment. The carrot cells were selected in liquid medium, a method previously unreported. The presence of the transgene was verified by PCR, and by RT-PCR. By western blot, G protein transgene was identified in 93.3% of adult carrot roots. The G protein was quantified by densitometric analysis (range 0.4-1.2%). The expressed protein was antigenic in mice. This confirms that the carrot is an adequate system for antigen expression.
Collapse
Affiliation(s)
- Edith Rojas-Anaya
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico, México DF
| | | | | | | |
Collapse
|
10
|
Perea Arango I, Loza Rubio E, Rojas Anaya E, Olivera Flores T, Gonzalez de la Vara L, Gómez Lim MA. Expression of the rabies virus nucleoprotein in plants at high-levels and evaluation of immune responses in mice. PLANT CELL REPORTS 2008; 27:677-85. [PMID: 18270708 DOI: 10.1007/s00299-007-0324-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Revised: 01/26/2007] [Accepted: 02/10/2007] [Indexed: 05/02/2023]
Abstract
Transgenic plants have been employed successfully as a low-cost system for the production of therapeutically valuable proteins including antibodies, antigens and hormones. Here, we report expression of a full-length nucleoprotein gene of rabies virus in transgenic tomato plants. The nucleoprotein was also transiently expressed in Nicotiana benthamiana plants by agroinfiltration. In both cases, the nucleoprotein was expressed at high levels, 1-5% of total soluble protein in tomato and 45% in N. benthamiana. Previously, only epitopes of the nucleoprotein had been expressed in plants. The presence and expression of the transgene was verified by PCR, Southern, northern and western blots. Mice were immunized both intraperitoneally (i.p.) and orally with tomato protein extracts containing the N protein induced the production of antibodies. The antibody titer of mice immunized i.p., was at least four times higher than that of mice immunized orally. These results were reflected in the challenge experiments where i.p.-immunized mice were partially protected against a peripheral virus challenge whereas orally immunized mice were not. This protection was comparable to that obtained in previous experiments employing different expression systems. Work is in progress to express both G and N proteins in transgenic plants and evaluate protection in mice.
Collapse
Affiliation(s)
- Irene Perea Arango
- Departamento de Ingeniería Genética de Plantas, Libramiento Norte Carretera Irapuato-León, Irapuato, 36500, Guanajuato, Mexico
| | | | | | | | | | | |
Collapse
|
11
|
Morales ME, Rico G, Gómez JL, Alonso R, Cortés R, Silva R, Giménez JA, Kretschmer R, Aguilar-Setién A. Could the homologous sequence of anti-inflammatory pentapeptide (MLIF) produced by Entamoeba histolytica in the N protein of rabies virus affect the inflammatory process? Parasitol Res 2005; 98:232-6. [PMID: 16341883 DOI: 10.1007/s00436-005-0027-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2005] [Accepted: 09/20/2005] [Indexed: 10/25/2022]
Abstract
Amebiasis and rabies are public health problems, and they have in common a poor inflammatory effect in the target organs that they affect. In the GenBank, it was found that the anti-inflammatory peptide monocyte locomotion inhibitory factor (MLIF) produced by Entamoeba histolytica homologates 80%, with a fragment of the N protein of the rabies virus. We speculated if the N protein could contribute to the scant inflammatory reaction produced by rabies virus in central nervous system. The N protein was obtained and studied in vitro and in vivo. The N protein, as MLIF, inhibited the respiratory burst in human mononuclear phagocytes (43%, p<0.05), but in contrast to MLIF, it increased chemotaxis and it did not significantly inhibit delayed hypersensitivity skin reaction to 1-chloro-2-4-dinitrobenzene in guinea pigs. Therefore, the full peptide sequence has to be present or it has to be cleaved-free from the large recombinant N protein molecule (55 kDa) to become active.
Collapse
Affiliation(s)
- M E Morales
- Unidad de Investigación Médica en Inmunología, Unidad Médica de Alta Especialidad, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Mexico City, DF, Mexico.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Finke S, Conzelmann KK. Recombinant rhabdoviruses: vectors for vaccine development and gene therapy. Curr Top Microbiol Immunol 2005; 292:165-200. [PMID: 15981472 DOI: 10.1007/3-540-27485-5_8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The establishment of methods to recover rhabdoviruses from cDNA, so-called reverse genetics systems, has made it possible to genetically engineer rhabdoviruses and to study all aspects of the virus life cycle by introducing defined mutations into the viral genomes. It has also opened the way to make use of the viruses in biomedical applications such as vaccination, gene therapy, or oncolytic virotherapy. The typical gene expression mode of rhabdoviruses, a high genetic stability, and the propensity to tolerate changes in the virus envelope have made rhabdoviruses attractive, targetable gene expression vectors. This chapter provides an overview on the possibilities to manipulate biological properties of the rhabdoviruses that may be important for further development of vaccine vectors and examples of recombinant rhabdoviruses expressing foreign genes and antigens.
Collapse
Affiliation(s)
- S Finke
- Max von Pettenkofer-Institut & Genzentrum, Ludwig-Maximilians-Universität, Feodor-Lynen-Str. 25, 81377 Munich, Germany.
| | | |
Collapse
|
13
|
Abstract
In the absence of treatment, infection with a variety of rabies virus strains most often results in a lethal outcome. This can be averted by prompt immunization following exposure demonstrating that the development of anti-rabies viral immunity prior to extensive infection of neurons is protective. Otherwise it might be expected that immune clearance of the virus would result in neurological sequelae. Thus, the capacity of a rabies virus to induce a protective immune response is a major, negative determinant of its pathogenicity and highly pathogenic rabies viruses have characteristics that avoid triggering protective immune responses. On the other hand, there is evidence that certain aspects of immunity may contribute to the pathogenesis of rabies under certain circumstances. The relationship between rabies virus and the immune system of the host is the focus of this review.
Collapse
Affiliation(s)
- D Craig Hooper
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
14
|
Nakamichi K, Inoue S, Takasaki T, Morimoto K, Kurane I. Rabies virus stimulates nitric oxide production and CXC chemokine ligand 10 expression in macrophages through activation of extracellular signal-regulated kinases 1 and 2. J Virol 2004; 78:9376-88. [PMID: 15308732 PMCID: PMC506932 DOI: 10.1128/jvi.78.17.9376-9388.2004] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Macrophages represent an essential part of innate immunity, and the viral infection of macrophages results in the release of multiple proinflammatory mediators, such as nitric oxide (NO), cytokines, and chemokines. This study was undertaken to define the molecular mechanism of macrophage activation in response to rabies virus (RV) infection. In RAW264 murine macrophage cells, a well-characterized macrophage model, RV replication was strictly restricted, whereas cell proliferation was significantly enhanced upon RV inoculation. Transcriptional analyses for the expression of inducible forms of NO synthase (iNOS), cytokines, and chemokines revealed that RV virions potentiate the gene expression of iNOS and CXC chemokine ligand 10 (CXCL10), a major chemoattractant of T helper cell type 1. However, RV stimulation had little or no effect on the expression profiles of proinflammatory cytokines and other types of chemokines. In macrophages stimulated with UV-inactivated RV virions, as well as infectious viruses, the phosphorylation of extracellular signal-regulated kinase (ERK) 1 and 2, members of the mitogen-activated protein kinase family, was significantly induced. Specific inhibitors of MAPK/ERK kinase reduced the RV-induced production of NO and CXCL10. Furthermore, the RV-induced activation of the ERK1/2 pathway was severely impaired by the neutralization of the endosomal and lysosomal pH environment with lysosomotropic agents, indicating that endocytosis is a key step leading to the activation of ERK1/2 signaling. Taken together, these results suggest that the ERK1/2-mediated signaling pathway plays a cardinal role in the selective activation of macrophages in response to RV virions, thereby regulating cellular functions during virus infection.
Collapse
Affiliation(s)
- Kazuo Nakamichi
- Department of Virology I, National Institute of Infectious Diseases, Toyama, Shinjuku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
15
|
Torres BA, Perrin GQ, Mujtaba MG, Subramaniam PS, Anderson AK, Johnson HM. Superantigen enhancement of specific immunity: antibody production and signaling pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2907-14. [PMID: 12218104 DOI: 10.4049/jimmunol.169.6.2907] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Superantigens are microbial proteins that induce massive activation, proliferation, and cytokine production by CD4+ T cells via specific Vbeta elements on the TCR. In this study we examine superantigen enhancement of Ag-specific CD4+ T cell activity for humoral B cell responses to T-dependent Ags BSA and HIV gp120 envelope, type I T-independent Ag LPS, and type II T-independent Ag pneumococcal polysaccharides. Injection of BSA followed by a combination of superantigens staphylococcal enterotoxin A and staphylococcal enterotoxin B (SEB) 7 days later enhanced the anti-BSA Ab response in mice approximately 4-fold as compared with mice given BSA alone. The anti-gp120 response was enhanced approximately 3-fold by superantigens. The type II T-independent Ag pneumococcal polysaccharide response was enhanced approximately 2.3-fold by superantigens, whereas no effect was observed on the response to the type I T-independent Ag LPS. The superantigen effect was completely blocked by the CD4+ T cell inhibitory cytokine IL-10. SEB-stimulated human CD4+ T cells were examined to determine the role of the mitogen-activated protein (MAP) kinase signal transduction pathway in superantigen activation of T cells. Inhibitors of the mitogen pathway of MAP kinase blocked SEB-induced proliferation and IFN-gamma production, while an inhibitor of the p38 stress pathway had no effect. Consistent with this, SEB activated extracellular signal-regulated kinase/MAP kinase as well as MAP kinase-interacting kinase, a kinase that phosphorylates eIF4E, which is an important component of the eukaryotic protein synthesis initiation complex. Both kinases were inhibited by IL-10. Thus, superantigens enhance humoral immunity via Ag-specific CD4+ T cells involving the stress-independent pathway of MAP kinase.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Animals
- Antibody Specificity/immunology
- CD4-Positive T-Lymphocytes/enzymology
- CD4-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Enterotoxins/administration & dosage
- Enterotoxins/immunology
- Enterotoxins/pharmacology
- Epitopes, T-Lymphocyte/immunology
- Female
- HIV Antibodies/biosynthesis
- HIV Antibodies/blood
- HIV Envelope Protein gp120/administration & dosage
- HIV Envelope Protein gp120/immunology
- Humans
- Immunity, Cellular/immunology
- Immunoglobulin G/biosynthesis
- Immunoglobulin G/blood
- Injections, Intraperitoneal
- Lymphocyte Activation/immunology
- MAP Kinase Signaling System/immunology
- Mice
- Mice, Inbred C57BL
- Serum Albumin, Bovine/administration & dosage
- Serum Albumin, Bovine/immunology
- Serum Albumin, Bovine/pharmacology
- Superantigens/administration & dosage
- Superantigens/immunology
Collapse
Affiliation(s)
- Barbara A Torres
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | | | | | | | | | | |
Collapse
|
16
|
Wirth S, Bille F, Koenig S, Wehrli N, Miconnet I, Lévy F, Diggelmann H, Romero P, Acha-Orbea H. Testing mouse mammary tumor virus superantigen as adjuvant in cytotoxic T-lymphocyte responses against a melanoma tumor antigen. Int J Cancer 2002; 99:201-6. [PMID: 11979434 DOI: 10.1002/ijc.10237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cytotoxic T cells represent a powerful strategy for antitumor treatment. Depending on the route of injection, an important role for CD4 T cell-mediated help was observed in the induction of this response. For this reason, we investigated whether induction of a CTL response to the HLA-A2-restricted immunodominant peptide melanoma antigen Melan-A was improved by using rVVs expressing the CTL-defined epitope alone or in combination with an SAg. In the latter case, the few infected dendritic cells simultaneously presented an SAg and an antigen, i.e., peptide. Here, we show that the anti-Melan-A response was efficiently induced but not significantly improved by coexpression of the SAg.
Collapse
Affiliation(s)
- Susanne Wirth
- Institute of Microbiology, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Piza AT, Pieri KMS, Lusa GM, Caporale GMM, Terreran MT, Machado LA, Zanetti CR. Effect of the contents and form of rabies glycoprotein on the potency of rabies vaccination in cattle. Mem Inst Oswaldo Cruz 2002; 97:265-8. [PMID: 12016455 DOI: 10.1590/s0074-02762002000200022] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
One of the methods used for controlling cattle rabies in Brazil consists of vaccination. Sometimes, however, rabies occurs in cattle supposedly protected. Since rabies vaccine batches are officially controlled by tests performed on laboratory animals, it is questionable whether the minimal mandatory requirements really correspond to immunogenicity in the target species. We have analyzed the association among potencies of rabies vaccines tested by the NIH test, the contents and form (free-soluble or virus-attached) of rabies glycoprotein (G) in the vaccine batches, and the virus-neutralizing antibodies (VNA) titers elicited in cattle. No correlation was found between G contents in the vaccine batches and the NIH values, whatever the presentation of G. There was no correlation either between NIH values and VNA titers elicited in cattle. There was, however, a positive correlation (r = 0.8681; p = 0.0001) between the amounts of virion-attached G present in the vaccine batches and VNA elicited in cattle. This was not observed when the same analysis was performed with total-glycoprotein or free-soluble glycoprotein. The study demonstrated that NIH values can not predict the effect of the immunogen in cattle. On the other hand, the quantification of virus-attached rabies glycoprotein has a strong correlation with VNA elicited in cattle.
Collapse
Affiliation(s)
- A T Piza
- Vallée S.A., São Paulo, SP, Brasil
| | | | | | | | | | | | | |
Collapse
|
18
|
Ghaffari G, Passalacqua DJ, Bender BS, Briggs DJ, Goodenow MM, Sleasman JW. Human lymphocyte proliferation responses following primary immunization with rabies vaccine as neoantigen. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2001; 8:880-3. [PMID: 11527796 PMCID: PMC96164 DOI: 10.1128/cdli.8.5.880-883.2001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Evaluation of the T-cell immune response following primary antigenic challenge with a neoantigen is a critical aspect of assessment of the cellular immune response. While many antigens can be used to accurately assess in vitro T-cell proliferation to a recall antigen, only a few neoantigens have been tested for their capacities to measure T-cell responses in vitro to a primary immunization. Rabies vaccination is an excellent candidate for the testing of T-cell proliferation responses to a primary immunization because few individuals have been exposed to rabies virus antigens. In the present study 14 rabies vaccine-naïve, healthy adult volunteers were immunized against rabies virus, and T-cell proliferation and antibody responses were measured before and after vaccination. Optimal lymphocyte proliferation to soluble rabies virus antigen occurred after 8 days in culture. The average level of uptake of tritiated thymidine postimmunization was 29,620 +/- 4,448 cpm, whereas preimmunization levels were 12,660 +/- 3,448 cpm (P = 0.002). All individuals showed increases in rabies virus antibody titers from <0.05 to 5.59 +/- 1.64 IU/ml. The degree of proliferation to tetanus toxoid as a recall antigen was similar to the response to rabies virus antigen among the cohort. Due to high levels of preimmunization proliferation, four subjects failed to demonstrate a twofold increase in response to rabies virus antigen. The high levels of T-cell responses may be due to a viral superantigen effect in some individuals. Rabies vaccination offers a safe and effective means for measurement of both T- and B-cell immune responses to a neoantigen in healthy and immune suppressed individuals.
Collapse
Affiliation(s)
- G Ghaffari
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, 32610, USA
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
As shown in a number of species, susceptibility to infectious diseases can be efficiently reduced following application of inactivated parapox ovis viruses (iPPOV). However, the basic mechanism for this stimulating capacity of iPPOV remains unclear. When analyzed, the interaction of iPPOV with porcine peripheral blood mononuclear cells was seen to involve T helper cells as the main target cell population responding to iPPOV. These cells displayed a strong proliferation, and were the major source for the observed increased levels of IL-2. Activation of the T helper cells was MHC class II dependent, but not MHC class II restricted: cellular processing of iPPOV was not required for presentation by autologous, allogeneic or xenogeneic MHC class II molecules. Furthermore, CD3 and CD4 molecules were involved in the stimulation, indicating a receptor-mediated activation of T helper cells. The results demonstrated typical characteristics of a superantigen-induced response providing evidence for a viral component within PPOV functioning as superantigen(s) in swine.
Collapse
Affiliation(s)
- V Fachinger
- Institute of Immunology, Federal Research Centre for Virus Diseases of Animals, Tübingen, Germany
| | | | | |
Collapse
|
20
|
Huang CC, Coppola MA, Nguyen P, Carragher D, Rohl C, Flynn KJ, Altman JD, Blackman MA. Effect of Staphylococcus enterotoxin B on the concurrent CD8(+) T cell response to influenza virus infection. Cell Immunol 2000; 204:1-10. [PMID: 11006012 DOI: 10.1006/cimm.2000.1692] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bacterial superantigens have potent in vivo effects. Respiratory viral infections are often associated with secondary bacterial infections, raising the likelihood of exposure to bacterial superantigens after the initiation of the anti-viral immune response. In this study, the general and V beta-specific effects of exposure to Staphylococcal enterotoxin B (SEB) during influenza virus infection on both the ongoing acute and the subsequent recall CD8(+) T cell responses were analyzed, using the well-characterized murine influenza model system and tetrameric MHC/peptide reagents to directly identify virus-specific T cells. The results show that although superantigen exposure during the primary viral infection caused delayed viral clearance, there was remarkably little effect of SEB on the magnitude or TCR repertoire of the ongoing cytolytic T cell response or on the recall response elicited by secondary viral infection. Thus, despite the well-characterized immunomodulatory effects of SEB, there was surprisingly little interference with concurrent anti-viral immunity.
Collapse
Affiliation(s)
- C C Huang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Macphail S. Superantigens: mechanisms by which they may induce, exacerbate and control autoimmune diseases. Int Rev Immunol 1999; 18:141-80. [PMID: 10614742 DOI: 10.3109/08830189909043022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Superantigens are polypeptide molecules produced by a broad range of infectious microorganisms which elicit excessive and toxic T-cell responses in mammalian hosts. In light of this property and the fact that autoimmune diseases are frequently the sequelae of microbial infections, it has been suggested that superantigens may be etiologic agents of autoreactive immunological responses resulting in initiation, exacerbation or relapse of autoimmune diseases. This article relates the biology of superantigens to possible mechanisms by which they may exert these activities and reviews the evidence for their roles in various human and animal models of autoimmune disease. Finally, a mechanism of active suppression by superantigen-activated CD4+ T-cells that could be exploited for therapy as well as prophylaxis of human autoimmune diseases is proposed.
Collapse
Affiliation(s)
- S Macphail
- Department of Surgery, North Shore University Hospital, New York University School of Medicine and Cornell University Medical College, Manhasset, USA.
| |
Collapse
|
22
|
Abstract
Superantigens (SAGs) are a class of immunostimulatory and disease-causing proteins of bacterial or viral origin with the ability to activate large fractions (5-20%) of the T cell population. Activation requires simultaneous interaction of the SAG with the V beta domain of the T cell receptor (TCR) and with major histocompatibility complex (MHC) class II molecules on the surface of an antigen-presenting cell. Recent advances in knowledge of the three-dimensional structure of bacterial SAGs, and of their complexes with MHC class II molecules and the TCR beta chain, provide a framework for understanding the molecular basis of T cell activation by these potent mitogens. These structures along with those of TCR-peptide/MHC complexes reveal how SAGs circumvent the normal mechanism for T cell activation by peptide/MHC and how they stimulate T cells expressing TCR beta chains from a number of different families, resulting in polyclonal T cell activation. The crystal structures also provide insights into the basis for the specificity of different SAGs for particular TCR beta chains, and for the observed influence of the TCR alpha chain on SAG reactivity. These studies open the way to the design of SAG variants with altered binding properties for TCR and MHC for use as tools in dissecting structure-activity relationships in this system.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Autoimmune Diseases/immunology
- Foodborne Diseases/immunology
- Histocompatibility Antigens Class II/chemistry
- Histocompatibility Antigens Class II/metabolism
- Humans
- Immunotherapy
- Kinetics
- Lymphocyte Activation/physiology
- Macromolecular Substances
- Models, Molecular
- Molecular Sequence Data
- Protein Conformation
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Shock, Septic/immunology
- Superantigens/administration & dosage
- Superantigens/chemistry
- Superantigens/metabolism
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- H Li
- Center for Advanced Research in Biotechnology, University of Maryland Biotechnology Institute, Rockville 20850, USA
| | | | | | | |
Collapse
|
23
|
Leutenegger CM, Hofmann-Lehmann R, Holznagel E, Cuisinier AM, Wolfensberger C, Duquesne V, Cronier J, Allenspach K, Aubert A, Ossent P, Lutz H. Partial protection by vaccination with recombinant feline immunodeficiency virus surface glycoproteins. AIDS Res Hum Retroviruses 1998; 14:275-83. [PMID: 9491919 DOI: 10.1089/aid.1998.14.275] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In an effort to induce a strong immune response that might protect against feline immunodeficiency virus (FIV) challenge infection, three groups of five specified pathogen-free (spf) cats each were immunized subcutaneously with different FIV antigen preparations. Immunizations were done at weeks 0, 2, and 4 with 100 microg of recombinant SU from an FIV Zurich 2 (FIV Z2) strain expressed by E. coli (group 1) or the baculovirus expression system (groups 2 and 3) adsorbed on aluminum hydroxyde and administered with QS-21 (groups 1 and 2) or Freund's adjuvant together with the recombinant nucleocapsid protein (protein NC) of rabies virus (group 3). Protein NC was described to act as an exogenous superantigen. Group 3 cats demonstrated the highest detectable antibody response to the vaccine antigen as determined by ELISA and Western blot analysis. All immunized cats together with seven control animals were challenged with 20 CID50 of cat lymphocyte-grown FIV Z2 3 weeks following the last immunization. Whereas virus was readily recovered from peripheral blood lymphocytes of seven of seven nonvaccinated control cats following this challenge dose, virus was not recovered from two cats of groups 1 and 2. All cats in groups 2 and 3 showed a provirus load significantly decreased to 3% of that of controls up to week 8 after challenge infection. Eleven of 15 vaccinated cats and 5 of 7 control cats developed virus-neutralizing antibodies by week 8 after challenge infection. The two cats negative on virus isolation remained seronegative, developed no detectable virus-neutralizing activities, but were repeatedly positive in provirus PCR. Moreover, starting at week 1 after challenge, both cats showed the lowest provirus load in their respective groups. These results indicate that immunization with recombinant FIV SU in conjunction with appropriate adjuvants may lead to partial protection against FIV challenge infection.
Collapse
Affiliation(s)
- C M Leutenegger
- Clinical Laboratory, Department of Internal Veterinary Medicine, University of Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Thoulouze MI, Lafage M, Montano-Hirose JA, Lafon M. Rabies virus infects mouse and human lymphocytes and induces apoptosis. J Virol 1997; 71:7372-80. [PMID: 9311815 PMCID: PMC192082 DOI: 10.1128/jvi.71.10.7372-7380.1997] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Attenuated and highly neurovirulent rabies virus strains have distinct cellular tropisms. Highly neurovirulent strains such as the challenge virus standard (CVS) are highly neurotropic, whereas the attenuated strain ERA also infects nonneuronal cells. We report that both rabies virus strains infect activated murine lymphocytes and the human lymphoblastoid Jurkat T-cell line in vitro. The lymphocytes are more permissive to the attenuated ERA rabies virus strain than to the CVS strain in both cases. We also report that in contrast to that of the CVS strain, ERA viral replication induces apoptosis of infected Jurkat T cells, and cell death is concomitant with viral glycoprotein expression, suggesting that this protein has a role in the induction of apoptosis. Our data indicate that (i) rabies virus infects lymphocytes, (ii) lymphocyte infection with the attenuated rabies virus strain causes apoptosis, and (iii) apoptosis does not hinder rabies virus production. In contrast to CVS, ERA rabies virus and other attenuated rabies virus vaccines stimulate a strong immune response and are efficient live vaccines. The paradoxical finding that a rabies virus triggers a strong immune response despite the fact that it infects lymphocytes and induces apoptosis is discussed in terms of the function of apoptosis in the immune response.
Collapse
Affiliation(s)
- M I Thoulouze
- Département de Virologie, Unité de Neurovirologie et Régénération du Système Nerveux, Institut Pasteur, Paris, France
| | | | | | | |
Collapse
|
25
|
Delcourt M, Thibodeau J, Denis F, Sekaly RP. Paracrine transfer of mouse mammary tumor virus superantigen. J Exp Med 1997; 185:471-80. [PMID: 9053447 PMCID: PMC2196028 DOI: 10.1084/jem.185.3.471] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Transfer of vSAG7, the endogenous superantigen encoded in the Mtv7 locus, from MHC class II to MHC class II+ cells has been suggested to occur both in vivo and in vitro. This transfer usually leads to the activation and deletion of T cells expressing responsive V beta s. However, there is no direct molecular evidence for such a transfer. We have developed an in vitro system which confirms this property of vSAGs. vSAG7 was transfected into a class II murine fibroblastic line. Coculture of these cells with class II+ cells and murine T cell hybridomas expressing the specific V beta s led to high levels of IL-2 production which was specifically inhibited by vSAG7- and MHC class II-specific mAbs. Moreover, injection of vSAG7+ class II+ cells in mice led to expansion of V beta 6+ CD4+ cells. We show that this transfer activity is paracrine but does not require cell-to-cell contact. Indeed, vSAG7 was transferred across semi-permeable membranes. Transfer can occur both from class II+ and class II+ cells, indicating that MHC class II does not sequester vSAG7. Finally, competition experiments using bacterial toxins with well defined binding sites showed that the transferred vSAG7 fragment binds to the alpha 1 domain of HLA-DR.
Collapse
Affiliation(s)
- M Delcourt
- Laboratoire d'Immunologie Institut de Recherches Cliniques de Montréal, Canada
| | | | | | | |
Collapse
|
26
|
Mouse Mammary Tumor Virus: Immunological Interplays between Virus and Host **This article was accepted for publication on 1 October 1996. Adv Immunol 1997. [DOI: 10.1016/s0065-2776(08)60743-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|