1
|
Hernández-Flores A, Elías-Díaz D, Cubillo-Cervantes B, Ibarra-Cerdeña CN, Morán D, Arnal A, Chaves A. Fighting Strategies Against Chagas' Disease: A Review. Pathogens 2025; 14:183. [PMID: 40005558 PMCID: PMC11858460 DOI: 10.3390/pathogens14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/08/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, remains a significant public health challenge, particularly in Latin America, where it is one of the most neglected diseases and is primarily transmitted by triatomine insects. The disease exhibits complexity due to its diverse transmission routes, including vectorial and non-vectorial mechanisms such as blood transfusions and congenital transmission. Effective monitoring and control strategies are critical to mitigating its impact. This review focuses on current monitoring and control efforts, emphasizing the importance of enhanced surveillance systems, improved risk assessments, and integrated vector control programs. Surveillance plays a pivotal role in early detection and timely intervention, particularly in endemic regions, while vector control remains central to reducing transmission. Moreover, the development of novel diagnostic tools, treatments, and vaccines is a crucial step in advancing control efforts. This review also highlights the involvement of local governments, international organizations, and civil society in executing these strategies, stressing the need for sustained political commitment to ensure the success of public health programs. By addressing key challenges in monitoring, control, and prevention, this review aims to provide insights and recommendations to further global efforts in reducing the burden of Chagas disease.
Collapse
Affiliation(s)
- Andrea Hernández-Flores
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, Av. Universidad #3000, Mexico City 04510, Mexico; (A.H.-F.); (A.A.)
| | - Debora Elías-Díaz
- Sistema de Estudios de Posgrado Posgrado en Biología, Universidad de Costa Rica, San José 11501-206, Costa Rica; (D.E.-D.); (B.C.-C.)
| | - Bernadeth Cubillo-Cervantes
- Sistema de Estudios de Posgrado Posgrado en Biología, Universidad de Costa Rica, San José 11501-206, Costa Rica; (D.E.-D.); (B.C.-C.)
| | - Carlos N. Ibarra-Cerdeña
- Departamento de Ecología Humana, Centro de Investigación y de Estudios Avanzados (Cinvestav), Unidad Mérida, Merida 97205, Mexico;
| | - David Morán
- Unidad de Ecología y Epidemiología, Programa Arbovirus y Zoonoses, Centro para Estudios de Salud, Universidad del Valle de Guatemala, Guatemala City 01015, Guatemala;
| | - Audrey Arnal
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, Av. Universidad #3000, Mexico City 04510, Mexico; (A.H.-F.); (A.A.)
- MIVEGEC, IRD, CNRS, Université de Montpellier, 34394 Montpellier, France
- International Joint Laboratory IRD/UNAM ELDORADO, Merida 97205, Mexico
| | - Andrea Chaves
- Centro Nacional de Innovaciones Biotecnológicas (CENIBiot), CeNAT, Conare, San José 1174-1200, Costa Rica
- Escuela de Biología, Universidad de Costa Rica, San José 11501-206, Costa Rica
| |
Collapse
|
2
|
Farani PSG, Jones KM, Poveda C. Treatments and the Perspectives of Developing a Vaccine for Chagas Disease. Vaccines (Basel) 2024; 12:870. [PMID: 39203996 PMCID: PMC11359273 DOI: 10.3390/vaccines12080870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Chagas disease (CD) treatment and vaccine development are critical due to the significant health burden caused by the disease, especially in Latin America. Current treatments include benznidazole and nifurtimox, which are most effective in the acute phase of the disease but less so in the chronic phase, often with significant side effects. Here, using the available literature, we summarize the progress in vaccine development and new treatments that promise to reduce CD incidence and improve the quality of life for those at risk, particularly in endemic regions. New treatment options, such as posaconazole and fexinidazole, are being explored to improve efficacy and reduce adverse effects. Vaccine development for CD remains a high priority. The complex life stages and genetic diversity of Trypanosoma cruzi present challenges, but several promising vaccine candidates are under investigation. These efforts focus on stimulating a protective immune response through various innovative approaches.
Collapse
Affiliation(s)
- Priscila Silva Grijó Farani
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Kathryn Marie Jones
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristina Poveda
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
3
|
Vázquez ME, Zabala BA, Mesías AC, Biscari L, Kaufman CD, Alloatti A, Siano F, Picariello G, Corbalán NS, Lenis BA, Toscano MA, Parodi CM, Brandán CMP, Acuña L. Protective Efficacy of the Epitope-Conjugated Antigen N-Tc52/TSkb20 in Mitigating Trypanosoma cruzi Infection through CD8+ T-Cells and IFNγ Responses. Vaccines (Basel) 2024; 12:621. [PMID: 38932350 PMCID: PMC11209121 DOI: 10.3390/vaccines12060621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 06/28/2024] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, remains a major public health challenge affecting millions in Latin America and worldwide. Although significant progress has been made in vector control, no vaccine exists to prevent infection or mitigate disease pathogenesis. We developed a rationally designed chimeric protein vaccine, N-Tc52/TSkb20, incorporating immunodominant epitopes from two T. cruzi antigens, the amino-terminal portion of Tc52 and the TSkb20 epitope derived from trans-sialidase. The objectives of this study were to construct and characterize the antigen and evaluate its protective potential in an immunoprophylactic murine model of T. cruzi infection. The N-Tc52/TSkb20 protein was recombinantly expressed in E. coli and its identity was confirmed using mass spectrometry and Western blotting. Immunization with the chimeric protein significantly controlled parasitemia and reduced the heart, colon, and skeletal muscle parasite burdens compared to non-vaccinated mice. Protection was superior to vaccination with the individual parental antigen components. Mechanistically, the vaccine induced potent CD8+ T-cell and IFNγ responses against the incorporated epitopes and a protective IgG antibody profile. A relatively low IL-10 response favored early parasite control. These results validate the promising multi-epitope approach and support the continued development of this type of rational vaccine design strategy against Chagas disease.
Collapse
Affiliation(s)
- María Elisa Vázquez
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Brenda A. Zabala
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Andrea C. Mesías
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Lucia Biscari
- Instituto de Inmunología Clínica y Experimental de Rosario, IDICER—CONICET—UNR, Rosario 2000, Argentina; (L.B.); (C.D.K.); (A.A.)
| | - Cintia D. Kaufman
- Instituto de Inmunología Clínica y Experimental de Rosario, IDICER—CONICET—UNR, Rosario 2000, Argentina; (L.B.); (C.D.K.); (A.A.)
| | - Andrés Alloatti
- Instituto de Inmunología Clínica y Experimental de Rosario, IDICER—CONICET—UNR, Rosario 2000, Argentina; (L.B.); (C.D.K.); (A.A.)
| | - Francesco Siano
- Istituto di Scienze dell’ Alimentazione—Consiglio Nazionale delle Ricerche (CNR), 83100 Avellino, Italy; (F.S.); (G.P.)
| | - Gianluca Picariello
- Istituto di Scienze dell’ Alimentazione—Consiglio Nazionale delle Ricerche (CNR), 83100 Avellino, Italy; (F.S.); (G.P.)
| | - Natalia S. Corbalán
- Facultad de Ciencias Naturales, Universidad Nacional de Salta, Salta A4400, Argentina;
| | - Bladimiro A. Lenis
- Unidad de Conocimiento Traslacional, Hospital Arturo Oñativia, Salta A4400, Argentina; (B.A.L.); (M.A.T.)
| | - Marta A. Toscano
- Unidad de Conocimiento Traslacional, Hospital Arturo Oñativia, Salta A4400, Argentina; (B.A.L.); (M.A.T.)
| | - Cecilia M. Parodi
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Cecilia M. Pérez Brandán
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| | - Leonardo Acuña
- Unidad de Biotecnología y Protozoarios, Instituto de Patología Experimental “Dr. Miguel Ángel Basombrío”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Salta, Salta A4400, Argentina; (M.E.V.); (B.A.Z.); (A.C.M.); (C.M.P.)
| |
Collapse
|
4
|
Bunkofske ME, Perumal N, White B, Strauch EM, Tarleton R. Epitopes in the Glycosylphosphatidylinositol Attachment Signal Peptide of Trypanosoma cruzi Mucin Proteins Generate Robust but Delayed and Nonprotective CD8+ T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:420-430. [PMID: 36603035 PMCID: PMC9898211 DOI: 10.4049/jimmunol.2200723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023]
Abstract
Infection with the protozoan parasite Trypanosoma cruzi elicits substantial CD8+ T cell responses that disproportionately target epitopes encoded in the large trans-sialidase (TS) gene family. Within the C57BL/6 infection model, a significant proportion (30-40%) of the T. cruzi-specific CD8+ T cell response targets two immunodominant TS epitopes, TSKb18 and TSKb20. However, both TS-specific CD8+ T cell responses are dispensable for immune control, and TS-based vaccines have no demonstrable impact on parasite persistence, a determinant of disease. Besides TS, the specificity and protective capacity of CD8+ T cells that mediate immune control of T. cruzi infection are unknown. With the goal of identifying alternative CD8+ T cell targets, we designed and screened a representative set of genome-wide, in silico-predicted epitopes. Our screen identified a previously uncharacterized, to our knowledge, T cell epitope MUCKb25, found within mucin family proteins, the third most expanded large gene family in T. cruzi. The MUCKb25-specific response was characterized by delayed kinetics, relative to TS-specific responses, and extensive cross-reactivity with a large number of endogenous epitope variants. Similar to TS-specific responses, the MUCKb25 response was dispensable for control of the infection, and vaccination to generate MUCK-specific CD8+ T cells failed to confer protection. The lack of protection by MUCK vaccination was partly attributed to the fact that MUCKb25-specific T cells exhibit limited recognition of T. cruzi-infected host cells. Overall, these results indicate that the CD8+ T cell compartment in many T. cruzi-infected mice is occupied by cells with minimal apparent effector potential.
Collapse
Affiliation(s)
- Molly E. Bunkofske
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Natasha Perumal
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Brooke White
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Eva-Maria Strauch
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Rick Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
5
|
Biscari L, Maza MC, Farré C, Kaufman CD, Amigorena S, Fresno M, Gironès N, Alloatti A. Sec22b-dependent antigen cross-presentation is a significant contributor of T cell priming during infection with the parasite Trypanosoma cruzi. Front Cell Dev Biol 2023; 11:1138571. [PMID: 36936692 PMCID: PMC10014565 DOI: 10.3389/fcell.2023.1138571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/17/2023] [Indexed: 03/05/2023] Open
Abstract
Antigen cross-presentation is a vital mechanism of dendritic cells and other antigen presenting cells to orchestrate the priming of cytotoxic responses towards killing of infected or cancer cells. In this process, exogenous antigens are internalized by dendritic cells, processed, loaded onto MHC class I molecules and presented to CD8+ T cells to activate them. Sec22b is an ER-Golgi Intermediate Compartment resident SNARE protein that, in partnership with sintaxin4, coordinates the recruitment of the transporter associated with antigen processing protein and the peptide loading complex to phagosomes, where antigenic peptides that have been proteolyzed in the cytosol are loaded in MHC class I molecules and transported to the cell membrane. The silencing of Sec22b in dendritic cells primary cultures and conditionally in dendritic cells of C57BL/6 mice, critically impairs antigen cross-presentation, but neither affects other antigen presentation routes nor cytokine production and secretion. Mice with Sec22b conditionally silenced in dendritic cells (Sec22b-/-) show deficient priming of CD8+ T lymphocytes, fail to control tumor growth, and are resistant to anti-checkpoint immunotherapy. In this work, we show that Sec22b-/- mice elicit a deficient specific CD8+ T cell response when challenged with sublethal doses of Trypanosoma cruzi trypomastigotes that is associated with increased blood parasitemia and diminished survival.
Collapse
Affiliation(s)
- Lucía Biscari
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), CONICET, Universidad Nacional de Rosario, Rosario, Argentina
| | - Ma Carmen Maza
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Cecilia Farré
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), CONICET, Universidad Nacional de Rosario, Rosario, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Cintia Daniela Kaufman
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), CONICET, Universidad Nacional de Rosario, Rosario, Argentina
| | - Sebastian Amigorena
- Institut Curie, INSERM U932, Immunity and Cancer, PSL University, Paris, France
| | - Manuel Fresno
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, Madrid, Spain
| | - Núria Gironès
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, Madrid, Spain
| | - Andrés Alloatti
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), CONICET, Universidad Nacional de Rosario, Rosario, Argentina
- *Correspondence: Andrés Alloatti,
| |
Collapse
|
6
|
Montalvo-Ocotoxtle IG, Rojas-Velasco G, Rodríguez-Morales O, Arce-Fonseca M, Baeza-Herrera LA, Arzate-Ramírez A, Meléndez-Ramírez G, Manzur-Sandoval D, Lara-Romero ML, Reyes-Ortega A, Espinosa-González P, Palacios-Rosas E. Chagas Heart Disease: Beyond a Single Complication, from Asymptomatic Disease to Heart Failure. J Clin Med 2022; 11:7262. [PMID: 36555880 PMCID: PMC9784121 DOI: 10.3390/jcm11247262] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Chagas cardiomyopathy (CC), caused by the protozoan Trypanosoma cruzi, is an important cause of cardiovascular morbidity and mortality in developing countries. It is estimated that 6 to 7 million people worldwide are infected, and it is predicted that it will be responsible for 200,000 deaths by 2025. The World Health Organization (WHO) considers Chagas disease (CD) as a Neglected Tropical Disease (NTD), which must be acknowledged and detected in time, as it remains a clinical and diagnostic challenge in both endemic and non-endemic regions and at different levels of care. The literature on CC was analyzed by searching different databases (Medline, Cochrane Central, EMBASE, PubMed, Google Scholar, EBSCO) from 1968 until October 2022. Multicenter and bioinformatics trials, systematic and bibliographic reviews, international guidelines, and clinical cases were included. The reference lists of the included papers were checked. No linguistic restrictions or study designs were applied. This review is intended to address the current incidence and prevalence of CD and to identify the main pathogenic mechanisms, clinical presentation, and diagnosis of CC.
Collapse
Affiliation(s)
- Isis G. Montalvo-Ocotoxtle
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Gustavo Rojas-Velasco
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Olivia Rodríguez-Morales
- Department of Molecular Biology, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Minerva Arce-Fonseca
- Department of Molecular Biology, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Luis A. Baeza-Herrera
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Arturo Arzate-Ramírez
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Gabriela Meléndez-Ramírez
- Magnetic Resonance Imaging Department, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Daniel Manzur-Sandoval
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Mayra L. Lara-Romero
- Academic Department of Health Sciences, School of Sciences, Universidad de las Américas Puebla, Ex Hacienda Sta. Catarina Mártir S/N. San Andrés Cholula, Puebla 72810, Mexico
| | - Antonio Reyes-Ortega
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Patricia Espinosa-González
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Erika Palacios-Rosas
- Academic Department of Health Sciences, School of Sciences, Universidad de las Américas Puebla, Ex Hacienda Sta. Catarina Mártir S/N. San Andrés Cholula, Puebla 72810, Mexico
| |
Collapse
|
7
|
Caeiro LD, Masip YE, Rizzi M, Rodríguez ME, Pueblas Castro C, Sánchez DO, Coria ML, Cassataro J, Tekiel V. The Trypanosoma cruzi TcTASV-C protein subfamily administrated with U-Omp19 promotes a protective response against a lethal challenge in mice. Vaccine 2020; 38:7645-7653. [PMID: 33071003 DOI: 10.1016/j.vaccine.2020.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 01/08/2023]
Abstract
The development of a Chagaś disease vaccine has yet the need for the identification of novel combinations of antigens and adjuvants. Here, the performance of TcTASV-C proteins that are virulence factors of trypomastigotes and belong to a novel surface protein family specific for T. cruzi, have been evaluated as antigens for a prophylactic vaccine. Several immunization schemes in which TcTASV-C was combined with aluminum hydroxide, saponin and/or U-Omp19 were assayed. Aluminum hydroxide and saponin were assayed together to trigger different pathways of the immune response simultaneously. U-Omp19 is a promising novel adjuvant able to promote a Th1 immune response with IFNg production, thus an interesting molecule to be tested as adjuvant for the control of T. cruzi infection. Therefore, U-Omp19 was added to the aluminum hydroxide-saponin formulation as well as assayed individually with TcTASV-C. The immunization with TcTASV-C and U-Omp19 had the best performance as a prophylactic vaccine. Mice presented the lowest parasitemias and improved survival by 40% after being challenged with a highly virulent T. cruzi strain, which promoted 100% mortality in all other immunized groups. Immunization with TcTASV-C and U-Omp19 triggered cellular responses with IFN-γ and IL-17 production and with lytic antibodies that could explain the protection achieved by this vaccination scheme. To our knowledge, this is the first time that U-Omp19 is tested with a defined T. cruzi antigen in a vaccine formulation.
Collapse
Affiliation(s)
- Lucas D Caeiro
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| | - Yamil E Masip
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| | - Mariana Rizzi
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| | - Matías E Rodríguez
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| | - Celeste Pueblas Castro
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| | - Daniel O Sánchez
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| | - M Lorena Coria
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| | - Valeria Tekiel
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín (UNSAM) - CONICET, Av. 25 de Mayo y Francia, Campus UNSAM, San Martín (1650), Provincia de Buenos Aires, Argentina.
| |
Collapse
|
8
|
DNA Vaccine Treatment in Dogs Experimentally Infected with Trypanosoma cruzi. J Immunol Res 2020; 2020:9794575. [PMID: 32455143 PMCID: PMC7222601 DOI: 10.1155/2020/9794575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/08/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023] Open
Abstract
Chagas disease is a chronic and potentially lethal disorder caused by the parasite Trypanosoma cruzi, and an effective treatment has not been developed for chronic Chagas disease. The objective of this study was to determine the effectiveness of a therapeutic DNA vaccine containing T. cruzi genes in dogs with experimentally induced Chagas disease through clinical, pathological, and immunological analyses. Infection of Beagle dogs with the H8 T. cruzi strain was performed intraperitoneally with 3500 metacyclic trypomastigotes/kg body weight. Two weeks after infection, plasmid DNA immunotherapy was administered thrice at 15-day intervals. The clinical (physical and cabinet studies), immunological (antibody and cytokine profiles and lymphoproliferation), and macro- and microscopic pathological findings were described. A significant increase in IgG and cell proliferation was recorded after immunotherapy, and the highest stimulation index (3.02) was observed in dogs treated with the pBCSSP4 plasmid. The second treatment with both plasmids induced an increase in IL-1, and the third treatment with the pBCSSP4 plasmid induced an increase in IL-6. The pBCSP plasmid had a good Th1 response regulated by high levels of IFN-gamma and TNF-alpha, whereas the combination of the two plasmids did not have a synergistic effect. Electrocardiographic studies registered lower abnormalities and the lowest number of individuals with abnormalities in each group treated with the therapeutic vaccine. Echocardiograms showed that the pBCSSP4 plasmid immunotherapy preserved cardiac structure and function to a greater extent and prevented cardiomegaly. The two plasmids alone controlled the infection moderately by a reduction in the inflammatory infiltrates in heart tissue. The immunotherapy was able to reduce the magnitude of cardiac lesions and modulate the cellular immune response; the pBCSP treatment showed a clear Th1 response; and pBCSSP4 induced a balanced Th1/Th2 immune response that prevented severe cardiac involvement. The pBCSSP4 plasmid had a better effect on most of the parameters evaluated in this study; therefore, this plasmid can be considered an optional treatment against Chagas disease in naturally infected dogs.
Collapse
|
9
|
Hegazy-Hassan W, Zepeda-Escobar JA, Ochoa-García L, Contreras-Ortíz JME, Tenorio-Borroto E, Barbabosa-Pliego A, Aparicio-Burgos JE, Oros-Pantoja R, Rivas-Santiago B, Díaz-Albiter H, Garg NJ, Vázquez-Chagoyán JC. TcVac1 vaccine delivery by intradermal electroporation enhances vaccine induced immune protection against Trypanosoma cruzi infection in mice. Vaccine 2018; 37:248-257. [PMID: 30497833 DOI: 10.1016/j.vaccine.2018.11.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
Abstract
The efforts for the development and testing of vaccines against Trypanosoma cruzi infection have increased during the past years. We have designed a TcVac series of vaccines composed of T. cruzi derived, GPI-anchored membrane antigens. The TcVac vaccines have been shown to elicit humoral and cellular mediated immune responses and provide significant (but not complete) control of experimental infection in mice and dogs. Herein, we aimed to test two immunization protocols for the delivery of DNA-prime/DNA-boost vaccine (TcVac1) composed of TcG2 and TcG4 antigens in a BALB/c mouse model. Mice were immunized with TcVac1 through intradermal/electroporation (IDE) or intramuscular (IM) routes, challenged with T. cruzi, and evaluated during acute phase of infection. The humoral immune response was evaluated through the assessment of anti-TcG2 and anti-TcG4 IgG subtypes by using an ELISA. Cellular immune response was assessed through a lymphocyte proliferation assay. Finally, clinical and morphopathological aspects were evaluated for all experimental animals. Our results demonstrated that when comparing TcVac1 IDE delivery vs IM delivery, the former induced significantly higher level of antigen-specific antibody response (IgG2a + IgG2b > IgG1) and lymphocyte proliferation, which expanded in response to challenge infection. Histological evaluation after challenge infection showed infiltration of inflammatory cells (macrophages and lymphocytes) in the heart and skeletal tissue of all infected mice. However, the largest increase in inflammatory infiltrate was observed in TcVac1_IDE/Tc mice when compared with TcVac1_IM/Tc or non-vaccinated/infected mice. The extent of tissue inflammatory infiltrate was directly associated with the control of tissue amastigote nests in vaccinated/infected (vs. non-vaccinated/infected) mice. Our results suggest that IDE delivery improves the protective efficacy of TcVac1 vaccine against T. cruzi infection in mice when compared with IM delivery of the vaccine.
Collapse
Affiliation(s)
- Wael Hegazy-Hassan
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - José Antonio Zepeda-Escobar
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - Laucel Ochoa-García
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico; Laboratorio Estatal de Salud Pública del Instituto de Salud del Estado de México, Independencia Oriente #1310 Colonia: Reforma y FFCC, CP. 50070 Toluca, Estado de México, Mexico
| | - J M Eloy Contreras-Ortíz
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - Esvieta Tenorio-Borroto
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - Alberto Barbabosa-Pliego
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - José Esteban Aparicio-Burgos
- Universidad Autónoma del Estado de Hidalgo, Escuela Superior de Apan, Carretera Apan-Calpulalpan, Km. 8, Chimalpa Tlalayote S/N, Colonia Chimalpa, Apan, C.P. 43920 Hidalgo, Mexico
| | - Rigoberto Oros-Pantoja
- Facultad de Medicina, Universidad Autónoma del Estado de México, Departamento de Neurociencias, Tollocan esq. Jesus Carranza S/N, Colonia Moderna de la Cruz, C.P. 50180 Estado de México, Toluca, Mexico
| | - Bruno Rivas-Santiago
- Unidad de Investigación Médica Zacatecas-IMSS, Interior de la Alameda, 45, Centro, C.P. 98000 Zacatecas, Mexico
| | - Héctor Díaz-Albiter
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, University Place, Glasgow G12 8TA, United Kingdom; El Colegio de la Frontera Sur, Carretera Villahermosa-Reforma Km 15.5, Ranchería Guineo, sección II, CP 86280 Villahermosa, Tabasco, Mexico
| | - Nisha Jain Garg
- Departments of Microbiology & Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1070, United States; Departments of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1070, United States
| | - Juan Carlos Vázquez-Chagoyán
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico.
| |
Collapse
|
10
|
Differential binding of the HIV-1 envelope to phosphatidylserine receptors. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017. [PMID: 28622976 PMCID: PMC5593811 DOI: 10.1016/j.bbamem.2017.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Prior work has shown that the HIV-1 envelope of the human immunodeficiency virus (HIV) interacts directly with T-cell immunoglobulin mucin (TIM) family proteins. Herein, we demonstrate that HIV-1 envelope glycoproteins from varying HIV-1 clades bind differentially to TIM proteins and functionally similar proteins acting as phosphatidylserine (PtdSer) receptors. Using enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR) technology, we show that lysate containing HIV-1 envelope and recombinant HIV-1 envelope glycoproteins bind TIM-4 and advanced glycosylation end product-specific receptor (AGER). The complex binding of HIV-1 UG21 gp140 to TIM-4 or AGER suggests a biphasic interaction with these proteins. HIV-1 glycoproteins bind PS-binding proteins as confirmed by ELISA and SPR. HIV-1 glycoproteins from multiple clades bind to bind phosphatidylserine binding proteins. Surface plasmon resonance is used to characterize the binding kinetics of HIV-1 glycoprotein and phosphatidylserine. HIV-1 UG21 gp140 clade D binds TIM-4 or AGER in a biphasic manner.
Collapse
|
11
|
A Novel Vaccine Approach for Chagas Disease Using Rare Adenovirus Serotype 48 Vectors. Viruses 2016; 8:78. [PMID: 26978385 PMCID: PMC4810268 DOI: 10.3390/v8030078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/26/2016] [Accepted: 03/03/2016] [Indexed: 12/28/2022] Open
Abstract
Due to the increasing amount of people afflicted worldwide with Chagas disease and an increasing prevalence in the United States, there is a greater need to develop a safe and effective vaccine for this neglected disease. Adenovirus serotype 5 (Ad5) is the most common adenovirus vector used for gene therapy and vaccine approaches, but its efficacy is limited by preexisting vector immunity in humans resulting from natural infections. Therefore, we have employed rare serotype adenovirus 48 (Ad48) as an alternative choice for adenovirus/Chagas vaccine therapy. In this study, we modified Ad5 and Ad48 vectors to contain T. cruzi’s amastigote surface protein 2 (ASP-2) in the adenoviral early gene. We also modified Ad5 and Ad48 vectors to utilize the “Antigen Capsid-Incorporation” strategy by adding T. cruzi epitopes to protein IX (pIX). Mice that were immunized with the modified vectors were able to elicit T. cruzi-specific humoral and cellular responses. This study indicates that Ad48-modified vectors function comparable to or even premium to Ad5-modified vectors. This study provides novel data demonstrating that Ad48 can be used as a potential adenovirus vaccine vector against Chagas disease.
Collapse
|
12
|
Adenoviral Vector Vaccines Antigen Transgene. ADENOVIRAL VECTORS FOR GENE THERAPY 2016. [PMCID: PMC7150117 DOI: 10.1016/b978-0-12-800276-6.00021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the past decade adenovirus-based vaccines have progressed from preclinical studies, which universally showed the vectors’ high immunogenicity, to testing in humans. Clinical trials showed that adenovirus vectors are well tolerated by humans. They induce robust immune responses that can be expanded by booster immunization. The effect of preexisting neutralizing antibodies on vectors’ immunogenicity appears to be less severe than was observed in experimental animals and can readily be circumvented by using vectors to which most humans lack neutralizing antibodies. Additional clinical studies are needed to firmly establish the efficacy of adenoviral vector vaccines.
Collapse
|
13
|
Tanowitz HB, Machado FS, Spray DC, Friedman JM, Weiss OS, Lora JN, Nagajyothi J, Moraes DN, Garg NJ, Nunes MCP, Ribeiro ALP. Developments in the management of Chagas cardiomyopathy. Expert Rev Cardiovasc Ther 2015; 13:1393-409. [PMID: 26496376 DOI: 10.1586/14779072.2015.1103648] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Over 100 years have elapsed since the discovery of Chagas disease and there is still much to learn regarding pathogenesis and treatment. Although there are antiparasitic drugs available, such as benznidazole and nifurtimox, they are not totally reliable and often toxic. A recently released negative clinical trial with benznidazole in patients with chronic Chagas cardiomyopathy further reinforces the concerns regarding its effectiveness. New drugs and new delivery systems, including those based on nanotechnology, are being sought. Although vaccine development is still in its infancy, the reality of a therapeutic vaccine remains a challenge. New ECG methods may help to recognize patients prone to developing malignant ventricular arrhythmias. The management of heart failure, stroke and arrhythmias also remains a challenge. Although animal experiments have suggested that stem cell based therapy may be therapeutic in the management of heart failure in Chagas cardiomyopathy, clinical trials have not been promising.
Collapse
Affiliation(s)
- Herbert B Tanowitz
- a Department of Pathology , Albert Einstein College of Medicine , Bronx , NY , USA.,b Department of Medicine , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Fabiana S Machado
- c Department of Biochemistry and Immunology, Institute of Biological Science , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil.,d Program in Health Sciences: Infectious Diseases and Tropical Medicine, Medical School , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - David C Spray
- b Department of Medicine , Albert Einstein College of Medicine , Bronx , NY , USA.,e Dominick P. Purpura Department of Neuroscience , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Joel M Friedman
- f Department of Physiology & Biophysics , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Oren S Weiss
- a Department of Pathology , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Jose N Lora
- a Department of Pathology , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Jyothi Nagajyothi
- g Public Health Research Institute, New Jersey Medical School , Rutgers University , Newark , NJ , USA
| | - Diego N Moraes
- d Program in Health Sciences: Infectious Diseases and Tropical Medicine, Medical School , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil.,h Department of Internal Medicine and University Hospital , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - Nisha Jain Garg
- i Department of Microbiology & Immunology and Institute for Human Infections and Immunity , University of Texas Medical Branch , Galveston , TX , USA
| | - Maria Carmo P Nunes
- d Program in Health Sciences: Infectious Diseases and Tropical Medicine, Medical School , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil.,h Department of Internal Medicine and University Hospital , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - Antonio Luiz P Ribeiro
- d Program in Health Sciences: Infectious Diseases and Tropical Medicine, Medical School , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil.,h Department of Internal Medicine and University Hospital , Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| |
Collapse
|
14
|
Gupta S, Garg NJ. A Two-Component DNA-Prime/Protein-Boost Vaccination Strategy for Eliciting Long-Term, Protective T Cell Immunity against Trypanosoma cruzi. PLoS Pathog 2015; 11:e1004828. [PMID: 25951312 PMCID: PMC4423834 DOI: 10.1371/journal.ppat.1004828] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/23/2015] [Indexed: 12/15/2022] Open
Abstract
In this study, we evaluated the long-term efficacy of a two-component subunit vaccine against Trypanosoma cruzi infection. C57BL/6 mice were immunized with TcG2/TcG4 vaccine delivered by a DNA-prime/Protein-boost (D/P) approach and challenged with T. cruzi at 120 or 180 days post-vaccination (dpv). We examined whether vaccine-primed T cell immunity was capable of rapid expansion and intercepting the infecting T. cruzi. Our data showed that D/P vaccine elicited CD4+ (30-38%) and CD8+ (22-42%) T cells maintained an effector phenotype up to 180 dpv, and were capable of responding to antigenic stimulus or challenge infection by a rapid expansion (CD8>CD4) with type 1 cytokine (IFNγ+ and TFNα+) production and cytolytic T lymphocyte (CTL) activity. Subsequently, challenge infection at 120 or 180 dpv, resulted in 2-3-fold lower parasite burden in vaccinated mice than was noted in unvaccinated/infected mice. Co-delivery of IL-12- and GMCSF-encoding expression plasmids provided no significant benefits in enhancing the anti-parasite efficacy of the vaccine-induced T cell immunity. Booster immunization (bi) with recombinant TcG2/TcG4 proteins 3-months after primary vaccine enhanced the protective efficacy, evidenced by an enhanced expansion (1.2-2.8-fold increase) of parasite-specific, type 1 CD4+ and CD8+ T cells and a potent CTL response capable of providing significantly improved (3-4.5-fold) control of infecting T. cruzi. Further, CD8+T cells in vaccinated/bi mice were predominantly of central memory phenotype, and capable of responding to challenge infection 4-6-months post bi by a rapid expansion to a poly-functional effector phenotype, and providing a 1.5-2.3-fold reduction in tissue parasite replication. We conclude that the TcG2/TcG4 D/P vaccine provided long-term anti-T. cruzi T cell immunity, and bi would be an effective strategy to maintain or enhance the vaccine-induced protective immunity against T. cruzi infection and Chagas disease. Chagas disease, caused by Trypanosoma cruzi infection, represents the third greatest tropical disease burden in the world. No vaccine or suitable treatment is available for control of this infection. Based upon several studies we have conducted, we believe that TcG2 and TcG4 candidate antigens that are highly conserved in T. cruzi, expressed in clinically relevant forms of the parasite, and recognized by both B and T cell responses in multiple hosts, are an excellent choice for subunit vaccine development. In this study, we demonstrate that the delivery of TcG2 and TcG4 as a DNA-prime/protein-boost vaccine provided long-term protection from challenge infection, and this protection was associated with elicitation of long-lived CD8+ effector T cells. The longevity and efficacy of vaccine could be enhanced by booster immunization. We believe that this is the first report demonstrating a) a subunit vaccine can be useful in achieving long-term protection against T. cruzi infection and Chagas disease, and b) the effector T cells can be long-lived and play a role in vaccine elicited protection from parasitic infection.
Collapse
Affiliation(s)
- Shivali Gupta
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch (UTMB), Galveston, Texas, United States of America
- * E-mail: (SG); (NJG)
| | - Nisha J. Garg
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch (UTMB), Galveston, Texas, United States of America
- Department of Pathology, School of Medicine, University of Texas Medical Branch (UTMB), Galveston, Texas, United States of America
- Institute for Human Infections and Immunity and the Sealy Center for Vaccine Development, University of Texas Medical Branch (UTMB), Galveston, Texas, United States of America
- * E-mail: (SG); (NJG)
| |
Collapse
|
15
|
Efficacy of a trans-sialidase-ISCOMATRIX subunit vaccine candidate to protect against experimental Chagas disease. Vaccine 2015; 33:1274-83. [DOI: 10.1016/j.vaccine.2015.01.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 12/19/2014] [Accepted: 01/14/2015] [Indexed: 12/12/2022]
|
16
|
Arce-Fonseca M, Rios-Castro M, Carrillo-Sánchez SDC, Martínez-Cruz M, Rodríguez-Morales O. Prophylactic and therapeutic DNA vaccines against Chagas disease. Parasit Vectors 2015; 8:121. [PMID: 25885641 PMCID: PMC4343048 DOI: 10.1186/s13071-015-0738-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 02/13/2015] [Indexed: 12/26/2022] Open
Abstract
Chagas disease is a zoonosis caused by Trypanosoma cruzi in which the most affected organ is the heart. Conventional chemotherapy has a very low effectiveness; despite recent efforts, there is currently no better or more effective treatment available. DNA vaccines provide a new alternative for both prevention and treatment of a variety of infectious disorders, including Chagas disease. Recombinant DNA technology has allowed some vaccines to be developed using recombinant proteins or virus-like particles capable of inducing both a humoral and cellular specific immune response. This type of immunization has been successfully used in preclinical studies and there are diverse models for viral, bacterial and/or parasitic diseases, allergies, tumors and other diseases. Therefore, several research groups have been given the task of designing a DNA vaccine against experimental infection with T. cruzi. In this review we explain what DNA vaccines are and the most recent studies that have been done to develop them with prophylactic or therapeutic purposes against Chagas disease.
Collapse
Affiliation(s)
- Minerva Arce-Fonseca
- Department of Molecular Biology, Laboratory of Molecular Immunology and Proteomics. Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No. 1, Col. Sección XVI, Tlalpan, C.P. 14080, Mexico City, Mexico.
| | - Martha Rios-Castro
- Department of Molecular Biology, Laboratory of Molecular Immunology and Proteomics. Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No. 1, Col. Sección XVI, Tlalpan, C.P. 14080, Mexico City, Mexico.
| | - Silvia del Carmen Carrillo-Sánchez
- Department of Molecular Biology, Laboratory of Molecular Immunology and Proteomics. Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No. 1, Col. Sección XVI, Tlalpan, C.P. 14080, Mexico City, Mexico.
| | - Mariana Martínez-Cruz
- Department of Molecular Biology, Laboratory of Molecular Immunology and Proteomics. Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No. 1, Col. Sección XVI, Tlalpan, C.P. 14080, Mexico City, Mexico.
| | - Olivia Rodríguez-Morales
- Department of Molecular Biology, Laboratory of Molecular Immunology and Proteomics. Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No. 1, Col. Sección XVI, Tlalpan, C.P. 14080, Mexico City, Mexico.
| |
Collapse
|
17
|
Farrow AL, Rachakonda G, Gu L, Krendelchtchikova V, Nde PN, Pratap S, Lima MF, Villalta F, Matthews QL. Immunization with Hexon modified adenoviral vectors integrated with gp83 epitope provides protection against Trypanosoma cruzi infection. PLoS Negl Trop Dis 2014; 8:e3089. [PMID: 25144771 PMCID: PMC4140675 DOI: 10.1371/journal.pntd.0003089] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/30/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Trypanosoma cruzi is the causative agent of Chagas disease. Chagas disease is an endemic infection that affects over 8 million people throughout Latin America and now has become a global challenge. The current pharmacological treatment of patients is unsuccessful in most cases, highly toxic, and no vaccines are available. The results of inadequate treatment could lead to heart failure resulting in death. Therefore, a vaccine that elicits neutralizing antibodies mediated by cell-mediated immune responses and protection against Chagas disease is necessary. METHODOLOGY/PRINCIPAL FINDINGS The "antigen capsid-incorporation" strategy is based upon the display of the T. cruzi epitope as an integral component of the adenovirus' capsid rather than an encoded transgene. This strategy is predicted to induce a robust humoral immune response to the presented antigen, similar to the response provoked by native Ad capsid proteins. The antigen chosen was T. cruzi gp83, a ligand that is used by T. cruzi to attach to host cells to initiate infection. The gp83 epitope, recognized by the neutralizing MAb 4A4, along with His6 were incorporated into the Ad serotype 5 (Ad5) vector to generate the vector Ad5-HVR1-gp83-18 (Ad5-gp83). This vector was evaluated by molecular and immunological analyses. Vectors were injected to elicit immune responses against gp83 in mouse models. Our findings indicate that mice immunized with the vector Ad5-gp83 and challenged with a lethal dose of T. cruzi trypomastigotes confer strong immunoprotection with significant reduction in parasitemia levels, increased survival rate and induction of neutralizing antibodies. CONCLUSIONS/SIGNIFICANCE This data demonstrates that immunization with adenovirus containing capsid-incorporated T. cruzi antigen elicits a significant anti-gp83-specific response in two different mouse models, and protection against T. cruzi infection by eliciting neutralizing antibodies mediated by cell-mediated immune responses, as evidenced by the production of several Ig isotypes. Taken together, these novel results show that the recombinant Ad5 presenting T. cruzi gp83 antigen is a useful candidate for the development of a vaccine against Chagas disease.
Collapse
Affiliation(s)
- Anitra L. Farrow
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Girish Rachakonda
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Linlin Gu
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Valentina Krendelchtchikova
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Pius N. Nde
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Siddharth Pratap
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Maria F. Lima
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Fernando Villalta
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Qiana L. Matthews
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for AIDS Research, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
18
|
Rodrigues MM, Ersching J. Neglected tropical diseases, bioinformatics, and vaccines. J Infect Dis 2014; 211:175-7. [PMID: 25070940 DOI: 10.1093/infdis/jiu420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mauricio Martins Rodrigues
- Centro de Terapia Celular e Molecular Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, Brazil
| | - Jonatan Ersching
- Centro de Terapia Celular e Molecular Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, Brazil
| |
Collapse
|
19
|
Teh-Poot C, Tzec-Arjona E, Martínez-Vega P, Ramirez-Sierra MJ, Rosado-Vallado M, Dumonteil E. From genome screening to creation of vaccine against Trypanosoma cruzi by use of immunoinformatics. J Infect Dis 2014; 211:258-66. [PMID: 25070943 DOI: 10.1093/infdis/jiu418] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chagas disease is caused by the protozoan parasite Trypanosoma cruzi, and activation of CD8(+) T cells is crucial for a protective immune response. Therefore, the identification of antigens with major histocompatibility complex class I epitopes is a crucial step for vaccine development against T. cruzi. Our aim was to identify novel antigens and epitopes by immunoinformatics analysis of the parasite proteome (12 969 proteins) and to validate their immunotherapeutic potential in infected mice. We identified 172 predicted epitopes, using NetMHC and RANKPEP. The corresponding protein sequences were reanalyzed to generate a consensus prediction, and 26 epitopes were selected for in vivo validation. The interferon γ (IFN-γ) recall response of splenocytes from T. cruzi-infected mice confirmed that 10 of 26 epitopes (38%) induced IFN-γ production. The immunotherapeutic potential of a mixture of all 10 peptides was evaluated in infected mice. The therapeutic vaccine was able to control T. cruzi infection, as evidenced by reduced parasitemia, cardiac tissue inflammation, and parasite burden and increased survival. These findings illustrate the benefits of this approach for the rapid development of a vaccine against pathogens with large genomes. The identified peptides and the proteins from which they are derived are excellent candidates for the development of a vaccine against T. cruzi.
Collapse
Affiliation(s)
- Christian Teh-Poot
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Mexico
| | - Evelyn Tzec-Arjona
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Mexico
| | - Pedro Martínez-Vega
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Mexico
| | - Maria Jesus Ramirez-Sierra
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Mexico
| | - Miguel Rosado-Vallado
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Mexico
| | - Eric Dumonteil
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Mexico Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
20
|
CD8(+) T cell-mediated immunity during Trypanosoma cruzi infection: a path for vaccine development? Mediators Inflamm 2014; 2014:243786. [PMID: 25104879 PMCID: PMC4102079 DOI: 10.1155/2014/243786] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/15/2014] [Indexed: 11/05/2022] Open
Abstract
MHC-restricted CD8+ T cells are important during infection with the intracellular protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease. Experimental studies performed in the past 25 years have elucidated a number of features related to the immune response mediated by these T cells, which are important for establishing the parasite/host equilibrium leading to chronic infection. CD8+ T cells are specific for highly immunodominant antigens expressed by members of the trans-sialidase family. After infection, their activation is delayed, and the cells display a high proliferative activity associated with high apoptotic rates. Although they participate in parasite control and elimination, they are unable to clear the infection due to their low fitness, allowing the parasite to establish the chronic phase when these cells then play an active role in the induction of heart immunopathology. Vaccination with a number of subunit recombinant vaccines aimed at eliciting specific CD8+ T cells can reverse this path, thereby generating a productive immune response that will lead to the control of infection, reduction of symptoms, and reduction of disease transmission. Due to these attributes, activation of CD8+ T lymphocytes may constitute a path for the development of a veterinarian or human vaccine.
Collapse
|
21
|
Cazorla SI, Frank FM, Malchiodi EL. Vaccination approaches againstTrypanosoma cruziinfection. Expert Rev Vaccines 2014; 8:921-35. [DOI: 10.1586/erv.09.45] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
22
|
Mendes ÉA, Fonseca FG, Casério BM, Colina JP, Gazzinelli RT, Caetano BC. Recombinant vaccines against T. gondii: comparison between homologous and heterologous vaccination protocols using two viral vectors expressing SAG1. PLoS One 2013; 8:e63201. [PMID: 23690999 PMCID: PMC3654925 DOI: 10.1371/journal.pone.0063201] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 04/03/2013] [Indexed: 11/19/2022] Open
Abstract
The use of recombinant viral vectors expressing T. gondii antigens is a safe and efficient approach to induce immune response against the parasite and a valuable tool for vaccine development. We have previously protected mice from toxoplasmosis by immunizing the animals with an adenovirus expressing the protein SAG1 (AdSAG1) of T. gondii. We are now looking for ways to improve the vaccination strategy and enhance protection. One limitation of homologous vaccinations (sequential doses of the same vector) is induction of anti-vector immune response that blocks cell transduction, restricts transgene expression and, consequently, compromises the overall outcome of vaccination. One way to avert the effects of anti-vector response is to use different viruses in prime and boost (heterologous vaccination). Bearing this in mind, we generated a modified Vaccinia Virus Ankara encoding SAG1 (MVASAG1), to be tested as boost agent after prime with AdSAG1. Although minor differences were observed in the magnitude of the anti-SAG1 immune response induced by each vaccination protocol, the heterologous immunization with AdSAG1 followed by MVASAG1 resulted in improved capacity to control brain cyst formation in a model of chronic toxoplasmosis in C57BL/6 mice.
Collapse
Affiliation(s)
- Érica Araújo Mendes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flavio G. Fonseca
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Bárbara M. Casério
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Janaína P. Colina
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Tostes Gazzinelli
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
- Division of Infectious Disease and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (RTG); (BCC)
| | - Braulia C. Caetano
- Division of Infectious Disease and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (RTG); (BCC)
| |
Collapse
|
23
|
Gupta S, Garg NJ. TcVac3 induced control of Trypanosoma cruzi infection and chronic myocarditis in mice. PLoS One 2013; 8:e59434. [PMID: 23555672 PMCID: PMC3608676 DOI: 10.1371/journal.pone.0059434] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 02/14/2013] [Indexed: 11/19/2022] Open
Abstract
We characterized the immune responses elicited by a DNA-prime/MVA-boost vaccine (TcVac3) constituted of antigenic candidates (TcG2 and TcG4), shown to be recognized by B and T cell responses in Trypanosoma cruzi (Tc) infected multiple hosts. C57BL/6 mice immunized with TcVac3 elicited a strong antigen-specific, high-avidity, trypanolytic antibody response (IgG2b>IgG1); and a robust antigen- and Tc-specific CD8+T cell response with type-1 cytokine (IFN-γ+TNF-α>IL-4+IL-10) and cytolytic effector (CD8+CD107a+IFN-γ+Perforin+) phenotype. The vaccine-induced effector T cells significantly expanded upon challenge infection and provided >92% control of T. cruzi. Co-delivery of IL-12 and GMCSF cytokine adjuvants didn’t enhance the TcVac3-induced resistance to T. cruzi. In chronic phase, vaccinated/infected mice exhibited a significant decline (up to 70%) in IFN-γ+CD8+T cells, a predominance of immunoregulatory IL-10+/CD4+T and IL10+/CD8+T cells, and presented undetectable tissue parasitism, inflammatory infiltrate, and fibrosis in vaccinated/infected mice. In comparison, control mice responded to challenge infection by a low antibody response, mixed cytokine profile, and consistent activation of pro-inflammatory CD8+T cells associated with parasite persistence and pathologic damage in the heart. We conclude that TcVac3 elicited type-1 effector T cell immunity that effectively controlled T. cruzi infection, and subsequently, predominance of anti-inflammatory responses prevented chronic inflammation and myocarditis in chagasic mice.
Collapse
Affiliation(s)
- Shivali Gupta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Faculty of the Institute for Human Infections and Immunity, Center for Tropical Diseases, and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
24
|
Recombinant yellow fever viruses elicit CD8+ T cell responses and protective immunity against Trypanosoma cruzi. PLoS One 2013; 8:e59347. [PMID: 23527169 PMCID: PMC3601986 DOI: 10.1371/journal.pone.0059347] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 02/13/2013] [Indexed: 12/19/2022] Open
Abstract
Chagas’ disease is a major public health problem affecting nearly 10 million in Latin America. Despite several experimental vaccines have shown to be immunogenic and protective in mouse models, there is not a current vaccine being licensed for humans or in clinical trial against T. cruzi infection. Towards this goal, we used the backbone of Yellow Fever (YF) 17D virus, one of the most effective and well-established human vaccines, to express an immunogenic fragment derived from T. cruzi Amastigote Surface Protein 2 (ASP-2). The cDNA sequence of an ASP-2 fragment was inserted between E and NS1 genes of YF 17D virus through the construction of a recombinant heterologous cassette. The replication ability and genetic stability of recombinant YF virus (YF17D/ENS1/Tc) was confirmed for at least six passages in Vero cells. Immunogenicity studies showed that YF17D/ENS1/Tc virus elicited neutralizing antibodies and gamma interferon (IFN-γ) producing-cells against the YF virus. Also, it was able to prime a CD8+ T cell directed against the transgenic T. cruzi epitope (TEWETGQI) which expanded significantly as measured by T cell-specific production of IFN-γ before and after T. cruzi challenge. However, most important for the purposes of vaccine development was the fact that a more efficient protective response could be seen in mice challenged after vaccination with the YF viral formulation consisting of YF17D/ENS1/Tc and a YF17D recombinant virus expressing the TEWETGQI epitope at the NS2B-3 junction. The superior protective immunity observed might be due to an earlier priming of epitope-specific IFN-γ-producing T CD8+ cells induced by vaccination with this viral formulation. Our results suggest that the use of viral formulations consisting of a mixture of recombinant YF 17D viruses may be a promising strategy to elicit protective immune responses against pathogens, in general.
Collapse
|
25
|
Gupta S, Garg NJ. Delivery of antigenic candidates by a DNA/MVA heterologous approach elicits effector CD8(+)T cell mediated immunity against Trypanosoma cruzi. Vaccine 2012; 30:7179-86. [PMID: 23079191 DOI: 10.1016/j.vaccine.2012.10.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 10/05/2012] [Indexed: 01/22/2023]
Abstract
In this study, we have characterized the immune mechanisms elicited by antigenic candidates, TcG2 and TcG4, delivered by a DNA-prime/MVA-boost approach, and evaluated the host responses to Trypanosoma cruzi infection in C57BL/6 mice. Immunization of mice with antigenic candidates elicited antigen-specific, high-avidity, trypanolytic antibody response (IgG2b>IgG1) and CD8(+)T cells that exhibited type-1 cytolytic effector (CD8(+)CD107a(+)IFN-γ(+)Perforin(+)) phenotype. The extent of TcG2-dependent type 1 B and T cell immunity was higher than that noted in TcG4-immunized mice, and expanded accordingly in response to challenge infection with T. cruzi. The progression of chronic phase in immunized mice was associated with persistence of IgGs, 55-90% reduction in the frequency of proinflammatory (IFN-γ(+) or TNF-α(+)) CD8(+)T cells, and an increase or emergence of immunoregulatory (IL-10(+)) CD4/CD8 T cells. The tissue parasitism, infiltration of inflammatory infiltrate, parasite persistence, and fibrosis were decreased by 82-92% in heart and skeletal muscle of immunized/chronically infected mice. Control mice exhibited a significantly low antibody response, consistent activation of effector CD8(+)T cells dominated by pro-inflammatory phenotype and mixed cytokine profile (IFN-γ+TNF-α>IL-4+IL-10), parasite persistence and pathologic damage in chagasic hearts. We conclude that delivery of TcG2 or TcG4 by DNA-rMVA approach elicits effective antibody and CD8(+)T cell mediated immunity against T. cruzi and Chagas disease. The emergence of type 2 cytokine and T cell response in chronic phase was indicative of prevention of clinical disease.
Collapse
Affiliation(s)
- Shivali Gupta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, United States
| | | |
Collapse
|
26
|
Ono T, Yamaguchi Y, Oguma T, Takayama E, Takashima Y, Tadakuma T, Miyahira Y. Actively induced antigen-specific CD8+ T cells by epitope-bearing parasite pre-infection but not prime/boost virus vector vaccination could ameliorate the course of Plasmodium yoelii blood-stage infection. Vaccine 2012; 30:6270-8. [PMID: 22902783 DOI: 10.1016/j.vaccine.2012.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/31/2012] [Accepted: 08/04/2012] [Indexed: 12/16/2022]
Abstract
The lack of MHC molecules on red blood cells (RBCs) has led to questions regarding the immunological function of CD8(+) T cells against malarial blood-stage (MBS). However, several recent reports contradicting with this concept have suggested that they play an important role in the course of MBS infection. The present study generated genetically engineered murine malaria, Plasmodium yoelii, which expresses a well-defined Trypanosoma cruzi-derived, H-2K(b)-restricted CD8(+) T cell epitope, ANYNFTLV. Prime/boost vaccination by the use of recombinant adenovirus and recombinant modified vaccinia virus Ankara (MVA), which induced an enhanced number of ANYNFTLV-specific CD8(+) T cells, failed to prevent a pathological outcome to occur upon ANYNFTLV-expressing murine MBS infection. This outcome did not change even with the combination of passive transfer of an appreciable number of in vitro-expanded ANYNFTLV-specific CD8(+) T cells. In contrast, the pre-infection of mice with T. cruzi, which intrinsically bears the same CD8(+) T cell epitope significantly improved the survival of ANYNFTLV-expressing malaria-infected mice but not that of control malaria-infected ones. This protective effect was abrogated by the use of a CD8(+) T cell-depleting monoclonal antibody. Although the protective effect was observed only in certain situations, the actively induced antigen-specific CD8(+) T cells could ameliorate the pathologies caused by the MBS. This is the first study to implicate that the active induction of antigen-specific CD8(+) T cells should be included in the development of a vaccine against MBS.
Collapse
Affiliation(s)
- Takeshi Ono
- Department of Global Infectious Diseases and Tropical Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa City, Saitama 359-8513, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Dominguez MR, Ersching J, Lemos R, Machado AV, Bruna-Romero O, Rodrigues MM, de Vasconcelos JRC. Re-circulation of lymphocytes mediated by sphingosine-1-phosphate receptor-1 contributes to resistance against experimental infection with the protozoan parasite Trypanosoma cruzi. Vaccine 2012; 30:2882-91. [PMID: 22381075 DOI: 10.1016/j.vaccine.2012.02.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 01/26/2012] [Accepted: 02/15/2012] [Indexed: 12/23/2022]
Abstract
T-cell mediated immune responses are critical for acquired immunity against infection by the intracellular protozoan parasite Trypanosoma cruzi. Despite its importance, it is currently unknown where protective T cells are primed and whether they need to re-circulate in order to exert their anti-parasitic effector functions. Here, we show that after subcutaneous challenge, CD11c(+)-dependent specific CD8(+) T-cell immune response to immunodominant parasite epitopes arises almost simultaneously in the draining lymph node (LN) and the spleen. However, until day 10 after infection, we observed a clear upregulation of activation markers only on the surface of CD11C(+)PDCA1(+) cells present in the LN and not in the spleen. Therefore, we hypothesized that CD8(+) T cells re-circulated rapidly from the LN to the spleen. We investigated this phenomenon by administering FTY720 to T. cruzi-infected mice to prevent egress of T cells from the LN by interfering specifically with signalling through sphingosine-1-phosphate receptor-1. In T. cruzi-infected mice receiving FTY720, CD8 T-cell immune responses were higher in the draining LN and significantly reduced in their spleen. Most importantly, FTY720 increased susceptibility to infection, as indicated by elevated parasitemia and accelerated mortality. Similarly, administration of FTY720 to mice genetically vaccinated with an immunodominant parasite antigen significantly reduced their protective immunity, as observed by the parasitemia and survival of vaccinated mice. We concluded that re-circulation of lymphocytes mediated by sphingosine-1-phosphate receptor-1 greatly contributes to acquired and vaccine-induced protective immunity against experimental infection with a human protozoan parasite.
Collapse
Affiliation(s)
- Mariana R Dominguez
- Centro de Terapia Celular e Molecular, Universidade Federal de São Paulo-Escola Paulista de Medicina, Brazil
| | | | | | | | | | | | | |
Collapse
|
28
|
Rodrigues MM, Oliveira AC, Bellio M. The Immune Response to Trypanosoma cruzi: Role of Toll-Like Receptors and Perspectives for Vaccine Development. J Parasitol Res 2012; 2012:507874. [PMID: 22496959 PMCID: PMC3306967 DOI: 10.1155/2012/507874] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/31/2011] [Indexed: 02/06/2023] Open
Abstract
In the past ten years, studies have shown the recognition of Trypanosoma cruzi-associated molecular patterns by members of the Toll-like receptor (TLR) family and demonstrated the crucial participation of different TLRs during the experimental infection with this parasite. In the present review, we will focus on the role of TLR-activated pathways in the modulation of both innate and acquired immune responses to T. cruzi infection, as well as discuss the state of the art of vaccine research and development against the causative agent of Chagas disease (or American trypanosomiasis).
Collapse
Affiliation(s)
- Mauricio M. Rodrigues
- Centro de Terapia Celular e Molecular (CTCMol), Universidade Federal de São Paulo (UNIFESP), 04044-010 São Paulo, SP, Brazil
| | - Ana Carolina Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), 21941-902 Rio de Janeiro, RJ, Brazil
| | - Maria Bellio
- Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro (UFRJ), CCS, Avenida Carlos Chagas Filho, 373 Bloco D, sala 35, Cidade Universitária, 21941-902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
29
|
Vázquez-Chagoyán JC, Gupta S, Garg NJ. Vaccine development against Trypanosoma cruzi and Chagas disease. ADVANCES IN PARASITOLOGY 2011; 75:121-46. [PMID: 21820554 DOI: 10.1016/b978-0-12-385863-4.00006-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathology of Chagas disease presents a complicated and diverse picture in humans. The major complications and destructive evolutionary outcomes of chronic infection by Trypanosoma cruzi in humans include ventricular fibrillation, thromboembolism and congestive heart failure. Studies in animal models and human patients have revealed the pathogenic mechanisms during disease progression, pathology of disease and features of protective immunity. Accordingly, several antigens, antigen-delivery vehicles and adjuvants have been tested to elicit immune protection to T. cruzi in experimental animals. This review summarizes the research efforts in vaccine development against Chagas disease during the past decade.
Collapse
Affiliation(s)
- Juan C Vázquez-Chagoyán
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Estado de México, Toluca, Mexico
| | | | | |
Collapse
|
30
|
Quijano-Hernandez I, Dumonteil E. Advances and challenges towards a vaccine against Chagas disease. HUMAN VACCINES 2011; 7:1184-91. [PMID: 22048121 DOI: 10.4161/hv.7.11.17016] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Chagas disease is major public health problem, affecting nearly 10 million people, characterized by cardiac alterations leading to congestive heart failure and death of 20-40% of the patients infected with Trypanosoma cruzi, the protozoan parasite responsible for the disease. A vaccine would be key to improve disease control and we review here the recent advances and challenges of a T. cruzi vaccine. There is a growing consensus that a protective immune response requires the activation of a Th1 immune profile, with the stimulation of CD8 (+) T cells. Several vacines types, including recombinant proteins, DNA and viral vectors, as well as heterologous prime-boost combinations, have been found immunogenic and protective in mouse models, providing proof-of-concept data on the feasibility of a preventive or therapeutic vaccine to control a T. cruzi infection. However, several challenges such as better end-points, safety issues and trial design need to be addressed for further vaccine development to proceed.
Collapse
Affiliation(s)
- Israel Quijano-Hernandez
- Laboratorio de Parasitología, Centro de Investigaciones Regionales 'Dr. Hideyo Noguchi', Universidad Autónoma de Yucatán, Merida, Mexico
| | | |
Collapse
|
31
|
Arce-Fonseca M, Ramos-Ligonio A, López-Monteón A, Salgado-Jiménez B, Talamás-Rohana P, Rosales-Encina JL. A DNA vaccine encoding for TcSSP4 induces protection against acute and chronic infection in experimental Chagas disease. Int J Biol Sci 2011; 7:1230-8. [PMID: 22110377 PMCID: PMC3221361 DOI: 10.7150/ijbs.7.1230] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/01/2011] [Indexed: 12/12/2022] Open
Abstract
Immunization of mice with plasmids containing genes of Trypanosoma cruzi induces protective immunity in the murine model of Chagas disease. A cDNA clone that codes for an amastigote-specific surface protein (TcSSP4) was used as a candidate to develop a DNA vaccine. Mice were immunized with the recombinant protein rTcSSP4 and with cDNA for TcSSP4, and challenged with bloodstream trypomastigotes. Immunization with rTcSSP4 protein makes mice more susceptible to trypomastigote infection, with high mortality rates, whereas mice immunized with a eukaryotic expression plasmid containing the TcSSP4 cDNA were able to control the acute phase of infection. Heart tissue of gene-vaccinated animals did not show myocarditis and tissue damage at 365 days following infection, as compared with control animals. INF-γ was detected in sera of DNA vaccinated mice shortly after immunization, suggesting the development of a Th1 response. The TcSSP4 gene is a promising candidate for the development of an anti-T. cruzi DNA vaccine.
Collapse
Affiliation(s)
- Minerva Arce-Fonseca
- Departamento de Infectómica y Patogenesis Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., México D.F. 07360, México
| | | | | | | | | | | |
Collapse
|
32
|
Sullivan NL, Eickhoff CS, Zhang X, Giddings OK, Lane TE, Hoft DF. Importance of the CCR5-CCL5 axis for mucosal Trypanosoma cruzi protection and B cell activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1358-68. [PMID: 21715689 PMCID: PMC3150841 DOI: 10.4049/jimmunol.1100033] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Trypanosoma cruzi is an intracellular parasite and the causative agent of Chagas disease. Previous work has shown that the chemokine receptor CCR5 plays a role in systemic T. cruzi protection. We evaluated the importance of CCR5 and CCL5 for mucosal protection against natural oral and conjunctival T. cruzi challenges. T. cruzi-immune CCR5(-/-) and wild-type C57BL/6 mice were generated by repeated infectious challenges with T. cruzi. CCR5(-/-) and wild-type mice developed equivalent levels of cellular, humoral, and protective mucosal responses. However, CCR5(-/-)-immune mice produced increased levels of CCL5 in protected gastric tissues, suggesting compensatory signaling through additional receptors. Neutralization of CCL5 in CCR5(-/-)-immune mice resulted in decreased mucosal inflammatory responses, reduced T. cruzi-specific Ab-secreting cells, and significantly less mucosal T. cruzi protection, confirming an important role for CCL5 in optimal immune control of T. cruzi replication at the point of initial mucosal invasion. To investigate further the mechanism responsible for mucosal protection mediated by CCL5-CCR5 signaling, we evaluated the effects of CCL5 on B cells. CCL5 enhanced proliferation and IgM secretion in highly purified B cells triggered by suboptimal doses of LPS. In addition, neutralization of endogenous CCL5 inhibited B cell proliferation and IgM secretion during stimulation of highly purified B cells, indicating that B cell production of CCL5 has important autocrine effects. These findings demonstrate direct effects of CCL5 on B cells, with significant implications for the development of mucosal adjuvants, and further suggest that CCL5 may be important as a general B cell coactivator.
Collapse
Affiliation(s)
- Nicole L Sullivan
- Department of Molecular Microbiology and Immunology, Saint Louis University Medical Center, St. Louis, MO 63104, USA
| | | | | | | | | | | |
Collapse
|
33
|
Oral exposure to Trypanosoma cruzi elicits a systemic CD8⁺ T cell response and protection against heterotopic challenge. Infect Immun 2011; 79:3397-406. [PMID: 21628516 DOI: 10.1128/iai.01080-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Trypanosoma cruzi infects millions of people in Latin America and often leads to the development of Chagas disease. T. cruzi infection can be acquired at or near the bite site of the triatomine vector, but per os infection is also a well-documented mode of transmission, as evidenced by recent microepidemics of acute Chagas disease attributed to the consumption of parasite-contaminated foods and liquids. It would also be convenient to deliver vaccines for T. cruzi by the oral route, particularly live parasite vaccines intended for the immunization of reservoir hosts. For these reasons, we were interested in better understanding immunity to T. cruzi following oral infection or oral vaccination, knowing that the route of infection and site of antigen encounter can have substantial effects on the ensuing immune response. Here, we show that the route of infection does not alter the ability of T. cruzi to establish infection in muscle tissue nor does it impair the generation of a robust CD8(+) T cell response. Importantly, oral vaccination with attenuated parasites provides protection against wild-type (WT) T. cruzi challenge. These results strongly support the development of whole-organism-based vaccines targeting reservoir species as a means to alleviate the burden of Chagas disease in affected regions.
Collapse
|
34
|
Aparicio-Burgos JE, Ochoa-García L, Zepeda-Escobar JA, Gupta S, Dhiman M, Martínez JS, de Oca-Jiménez RM, Arreola MV, Barbabosa-Pliego A, Vázquez-Chagoyán JC, Garg NJ. Testing the efficacy of a multi-component DNA-prime/DNA-boost vaccine against Trypanosoma cruzi infection in dogs. PLoS Negl Trop Dis 2011; 5:e1050. [PMID: 21625470 PMCID: PMC3098890 DOI: 10.1371/journal.pntd.0001050] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 04/15/2011] [Indexed: 11/19/2022] Open
Abstract
Background Trypanosoma cruzi, the etiologic agent of Chagas Disease, is
a major vector borne health problem in Latin America and an emerging
infectious disease in the United States. Methods We tested the efficacy of a multi-component DNA-prime/DNA-boost vaccine
(TcVac1) against experimental T. cruzi infection in a
canine model. Dogs were immunized with antigen-encoding plasmids and
cytokine adjuvants, and two weeks after the last immunization, challenged
with T. cruzi trypomastigotes. We measured antibody
responses by ELISA and haemagglutination assay, parasitemia and infectivity
to triatomines by xenodiagnosis, and performed electrocardiography and
histology to assess myocardial damage and tissue pathology. Results Vaccination with TcVac1 elicited parasite-and antigen-specific IgM and IgG
(IgG2>IgG1) responses. Upon challenge infection, TcVac1-vaccinated dogs,
as compared to non-vaccinated controls dogs, responded to T.
cruzi with a rapid expansion of antibody response, moderately
enhanced CD8+ T cell proliferation and IFN-γ production,
and suppression of phagocytes’ activity evidenced by decreased
myeloperoxidase and nitrite levels. Subsequently, vaccinated dogs controlled
the acute parasitemia by day 37 pi (44 dpi in non-vaccinated dogs), and
exhibited a moderate decline in infectivity to triatomines. TcVac1-immunized
dogs did not control the myocardial parasite burden and electrocardiographic
and histopatholgic cardiac alterations that are the hallmarks of acute
Chagas disease. During the chronic stage, TcVac1-vaccinated dogs exhibited a
moderate decline in cardiac alterations determined by EKG and
anatomo-/histo-pathological analysis while
chronically-infected/non-vaccinated dogs continued to exhibit severe EKG
alterations. Conclusions Overall, these results demonstrated that TcVac1 provided a partial resistance
to T. cruzi infection and Chagas disease, and provide an
impetus to improve the vaccination strategy against Chagas disease. Immunization of dogs with DNA-prime/DNA-boost vaccine (TcVac1) enhanced the
Trypanosoma cruzi-specific type 1 antibody and
CD8+ T cell responses that resulted in an early control of
acute parasitemia and a moderate decline in pathological symptoms during chronic
phase. Further improvement of vaccine-induced immunity would be required to
achieve clinical and epidemiological benefits and prevent transmission of
parasites from vaccinated/infected dogs to triatomines.
Collapse
Affiliation(s)
- José E. Aparicio-Burgos
- Centro de Investigación y Estudios
Avanzados, Universidad Autónoma de Estado de México, Toluca,
México
| | - Laucel Ochoa-García
- Centro de Investigación y Estudios
Avanzados, Universidad Autónoma de Estado de México, Toluca,
México
| | | | - Shivali Gupta
- Department of Microbiology and Immunology,
University of Texas Medical Branch, Galveston, Texas, United States of
America
| | - Monisha Dhiman
- Department of Microbiology and Immunology,
University of Texas Medical Branch, Galveston, Texas, United States of
America
| | - José Simón Martínez
- Centro de Investigación y Estudios
Avanzados, Universidad Autónoma de Estado de México, Toluca,
México
| | | | | | - Alberto Barbabosa-Pliego
- Centro de Investigación y Estudios
Avanzados, Universidad Autónoma de Estado de México, Toluca,
México
| | - Juan C. Vázquez-Chagoyán
- Centro de Investigación y Estudios
Avanzados, Universidad Autónoma de Estado de México, Toluca,
México
- * E-mail: (NJG); (JCV-C)
| | - Nisha Jain Garg
- Department of Microbiology and Immunology,
University of Texas Medical Branch, Galveston, Texas, United States of
America
- Department of Pathology, University of Texas
Medical Branch, Galveston, Texas, United States of America
- Faculty of the Institute for Human Infections
and Immunity, and the Sealy Center for Vaccine Development, University of Texas
Medical Branch, Galveston, Texas, United States of America
- * E-mail: (NJG); (JCV-C)
| |
Collapse
|
35
|
Effect of a combination DNA vaccine for the prevention and therapy of Trypanosoma cruzi infection in mice: role of CD4+ and CD8+ T cells. Vaccine 2010; 28:7414-9. [PMID: 20850536 DOI: 10.1016/j.vaccine.2010.08.104] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2010] [Revised: 08/26/2010] [Accepted: 08/31/2010] [Indexed: 11/22/2022]
Abstract
Chagas disease is a major public health problem, with about 10 million infected people, and DNA vaccines are a promising alternative for the control of Trypanosoma cruzi, the causing agent of the disease. We tested here a new DNA vaccine encoding a combination of two leading parasite antigens, TSA-1 and Tc24, for the prevention and therapy of T. cruzi infection. Immunized Balb/c mice challenged by T. cruzi presented a significantly lower parasitemia and inflammatory cell density in the heart compared to control mice. Similarly, the therapeutic administration of the DNA vaccine was able to significantly reduce the parasitemia and inflammatory reaction in acutely infected Balb/c and C57BL/6 mice, and reduced cardiac tissue inflammation in chronically infected ICR mice. Therapeutic vaccination induced a marked increase in parasite-specific IFNγ producing CD4(+) and CD8(+) T cells in the spleen as well as an increase in CD4(+) and CD8(+) T cells in the infected cardiac tissue. In addition, some effect of the DNA vaccine could still be observed in CD4-knockout C57BL/6 mice, which presented a lower parasitemia and inflammatory cell density, but not in CD8-deficient mice, in which the vaccine had no effect. These results indicate that the activation of CD8(+) T cells plays a major role in the control of the infection by the therapeutic DNA vaccine, and to a somewhat lesser extent CD4(+) T cells. This observation opens interesting perspectives for the potentiation of this DNA vaccine candidate by including additional CD8(+) T cell antigens/epitopes in future vaccine formulations.
Collapse
|
36
|
Gupta S, Garg NJ. Prophylactic efficacy of TcVac2 against Trypanosoma cruzi in mice. PLoS Negl Trop Dis 2010; 4:e797. [PMID: 20706586 PMCID: PMC2919396 DOI: 10.1371/journal.pntd.0000797] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 07/15/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chagas disease is a major health problem in Latin America, and an emerging infectious disease in the US. Previously, we have screened the Trypanosoma cruzi sequence database by a computational/bioinformatics approach, and identified antigens that exhibited the characteristics of vaccine candidates. METHODOLOGY We investigated the protective efficacy of a multi-component DNA-prime/protein-boost vaccine (TcVac2) constituted of the selected candidates and cytokine (IL-12 and GM-CSF) expression plasmids in a murine model. C57BL/6 mice were immunized with antigen-encoding plasmids plus cytokine adjuvants, followed by recombinant proteins; and two-weeks later, challenged with T. cruzi trypomastigotes. ELISA and flow cytometry were employed to measure humoral (antibody isotypes) and cellular (lymphocyte proliferation, CD4(+) and CD8(+) T cell phenotype and cytokines) responses. Myocardial pathology was evaluated by H&E and Masson's trichrome staining. PRINCIPAL FINDINGS TcVac2 induced a strong antigen-specific antibody response (IgG2b>IgG1) and a moderate level of lymphocyte proliferation in mice. Upon challenge infection, TcVac2-vaccinated mice expanded the IgG2b/IgG1 antibodies and elicited a substantial CD8(+) T cell response associated with type 1 cytokines (IFN-gamma and TNF-alpha) that resulted in control of acute parasite burden. During chronic phase, antibody response persisted, splenic activation of CD8(+) T cells and IFN-gamma/TNF-alpha cytokines subsided, and IL-4/IL-10 cytokines became dominant in vaccinated mice. The tissue parasitism, inflammation, and fibrosis in heart and skeletal muscle of TcVac2-vaccinated chronic mice were undetectable by histological techniques. In comparison, mice injected with vector or cytokines only responded to T. cruzi by elicitation of a mixed (type 1/type 2) antibody, T cell and cytokine response, and exhibited persistent parasite burden and immunopathology in the myocardium. CONCLUSION TcVac2-induced activation of type 1 antibody and lymphocyte responses provided resistance to acute T. cruzi infection, and consequently, prevented the evolution of chronic immunopathology associated with parasite persistence in chagasic hearts.
Collapse
Affiliation(s)
- Shivali Gupta
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nisha Jain Garg
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Member of the Institute for Infections and Immunity, Center for Biodefense and Emerging Infectious Diseases, and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
37
|
Takayama E, Ono T, Carnero E, Umemoto S, Yamaguchi Y, Kanayama A, Oguma T, Takashima Y, Tadakuma T, García-Sastre A, Miyahira Y. Quantitative and qualitative features of heterologous virus-vector-induced antigen-specific CD8+ T cells against Trypanosoma cruzi infection. Int J Parasitol 2010; 40:1549-61. [PMID: 20620143 DOI: 10.1016/j.ijpara.2010.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 05/24/2010] [Accepted: 05/26/2010] [Indexed: 12/27/2022]
Abstract
We studied some aspects of the quantitative and qualitative features of heterologous recombinant (re) virus-vector-induced, antigen-specific CD8(+) T cells against Trypanosoma cruzi. We used three different, highly attenuated re-viruses, i.e., influenza virus, adenovirus and vaccinia virus, which all expressed a single, T. cruzi antigen-derived CD8(+) T-cell epitope. The use of two out of three vectors or the triple virus-vector vaccination regimen not only confirmed that the re-vaccinia virus, which was placed last in order for sequential immunisation, was an effective booster for the CD8(+) T-cell immunity in terms of the number of antigen-specific CD8(+) T cells, but also demonstrated that (i) the majority of cells exhibit the effector memory (T(EM)) phenotype, (ii) robustly secrete IFN-γ, (iii) express higher intensity of the CD122 molecule and (iv) present protective activity against T. cruzi infection. In contrast, placing the re-influenza virus last in sequential immunisation had a detrimental effect on the quantitative and qualitative features of CD8(+) T cells. The triple virus-vector vaccination was more effective at inducing a stronger CD8(+) T-cell immunity than using two re-viruses. The different quantitative and qualitative features of CD8(+) T cells induced by different immunisation regimens support the notion that the refinement of the best choice of multiple virus-vector combinations is indispensable for the induction of a maximum number of CD8(+) T cells of high quality.
Collapse
Affiliation(s)
- Eiji Takayama
- Department of Global Infectious Diseases and Tropical Medicine, National Defense Medical College, Tokorozawa City, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Rosenberg CS, Martin DL, Tarleton RL. CD8+ T cells specific for immunodominant trans-sialidase epitopes contribute to control of Trypanosoma cruzi infection but are not required for resistance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:560-8. [PMID: 20530265 PMCID: PMC3784248 DOI: 10.4049/jimmunol.1000432] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD8(+) T cells are essential for controlling Trypanosoma cruzi infection. During Brazil strain infection, C57BL/6 mice expand parasite-specific CD8(+) T cells recognizing the dominant TSKB20 (ANYKFTLV) and subdominant TSKB74 (VNYDFTLV) trans-sialidase gene (TS)-encoded epitopes with up to 40% of all CD8(+) T cells specific for these epitopes. Although this is one of the largest immunodominant T cell responses described for any infection, most mice fail to clear T. cruzi and subsequently develop chronic disease. To determine if immunodominant TS-specific CD8(+) T cells are necessary for resistance to infection, we epitope-tolerized mice by high-dose i.v. injections of TSKB20 or TSKB74 peptides. Tolerance induction led to deletion of TS-specific CD8(+) T cells but did not prevent the expansion of other effector CD8(+) T cell populations. Mice tolerized against either TSKB20 or TSKB74, or both epitopes simultaneously, exhibited transient increases in parasite loads, although ultimately they controlled the acute infection. Furthermore, BALB/c mice tolerized against the TSKD14 peptide effectively controlled acute T. cruzi infection. These data are consistent with the hypothesis that development of high-frequency CD8(+) T cell populations focused on TS-derived epitopes contributes to optimal control of acute infection but is not required for the development of immune resistance.
Collapse
Affiliation(s)
- Charles S. Rosenberg
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA
- Department of Microbiology, University of Georgia, Athens, GA
| | - Diana L. Martin
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA
- Department of Cellular Biology, University of Georgia, Athens, GA
| | - Rick L. Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA
- Department of Cellular Biology, University of Georgia, Athens, GA
| |
Collapse
|
39
|
Dumonteil E. Vaccine development against Trypanosoma cruzi and Leishmania species in the post-genomic era. INFECTION GENETICS AND EVOLUTION 2010; 9:1075-82. [PMID: 19805015 DOI: 10.1016/j.meegid.2009.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 02/17/2009] [Accepted: 02/19/2009] [Indexed: 10/21/2022]
Abstract
Trypanosoma cruzi and the genus Leishmania are protozoan parasites causing diseases of major public health importance, and the recent completion of the sequencing of their genomes has opened new opportunities to further our understanding of the mechanisms required for protection and the development of vaccines. For example, trans-sialidases, one of the largest protein families from T. cruzi, contain dominant CD8+ T cell epitopes, and their use as preventive or therapeutic vaccines in different animal models has provided encouraging results. A much wider range of antigens and vaccine formulations have been tested against Leishmania, and new correlates for protection are being defined, such as the induction of multifunctional Th1 effector cells capable of producing a complex set of cytokines. Also, while a large number of these vaccine candidates have been rather successful in mouse models, their usefulness in more relevant animal models is still poor, in spite of significant immunogenicity. Novel proteomics and genomics approaches are being used for antigen discovery and the identification of new vaccine candidates, some of which have shown promise for the control of infection. These studies cast little doubt that T. cruzi and Leishmania genomes represent major resources for understanding key aspects of the mechanisms of immune protection against these parasites, and the increasing use of these tools will greatly impact vaccine development.
Collapse
Affiliation(s)
- Eric Dumonteil
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr Hideyo Noguchi, Universidad Autónoma de Yucatán, Merida, Yucatan, Mexico
| |
Collapse
|
40
|
Perforin and gamma interferon expression are required for CD4+ and CD8+ T-cell-dependent protective immunity against a human parasite, Trypanosoma cruzi, elicited by heterologous plasmid DNA prime-recombinant adenovirus 5 boost vaccination. Infect Immun 2009; 77:4383-95. [PMID: 19651871 DOI: 10.1128/iai.01459-08] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A heterologous prime-boost strategy using plasmid DNA, followed by replication-defective recombinant adenovirus 5, is being proposed as a powerful way to elicit CD4(+) and CD8(+) T-cell-mediated protective immunity against intracellular pathogens. We confirmed this concept and furthered existing research by providing evidence that the heterologous prime-boost regimen using the gene encoding amastigote surface protein 2 elicited CD4(+) and CD8(+) T-cell-mediated protective immunity (reduction of acute parasitemia and prolonged survival) against experimental infection with Trypanosoma cruzi. Protective immunity correlated with the presence of in vivo antigen-specific cytotoxic activity prior to challenge. Based on this, our second goal was to determine the outcome of infection after heterologous prime-boost immunization of perforin-deficient mice. These mice were highly susceptible to infection. A detailed analysis of the cell-mediated immune responses in immunized perforin-deficient mice showed an impaired gamma interferon (IFN-gamma) secretion by immune spleen cells upon restimulation in vitro with soluble recombinant antigen. In spite of a normal numeric expansion, specific CD8(+) T cells presented several functional defects detected in vivo (cytotoxicity) and in vitro (simultaneous expression of CD107a/IFN-gamma or IFN-gamma/tumor necrosis factor alpha) paralleled by a decreased expression of CD44 and KLRG-1. Our final goal was to determine the importance of IFN-gamma in the presence of highly cytotoxic T cells. Vaccinated IFN-gamma-deficient mice developed highly cytotoxic cells but failed to develop any protective immunity. Our study thus demonstrated a role for perforin and IFN-gamma in a number of T-cell-mediated effector functions and in the antiparasitic immunity generated by a heterologous plasmid DNA prime-adenovirus boost vaccination strategy.
Collapse
|
41
|
Rodrigues MM, Alencar BCD, Claser C, Tzelepis F, Silveira EL, Haolla FA, Dominguez MR, Vasconcelos JR. Swimming against the current: genetic vaccination against Trypanosoma cruzi infection in mice. Mem Inst Oswaldo Cruz 2009; 104 Suppl 1:281-7. [DOI: 10.1590/s0074-02762009000900037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 04/30/2009] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | | | | | | | | | | | - José Ronnie Vasconcelos
- Centro Interdisciplinar de Terapia Gênica; Escola Paulista de Medicina, Brasil; Universidade de São Paulo, Brasil
| |
Collapse
|
42
|
Crager SE, Price M. Prizes and parasites: incentive models for addressing Chagas disease. THE JOURNAL OF LAW, MEDICINE & ETHICS : A JOURNAL OF THE AMERICAN SOCIETY OF LAW, MEDICINE & ETHICS 2009; 37:292-304. [PMID: 19493074 DOI: 10.1111/j.1748-720x.2009.00373.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Recent advances in immunology have provided a foundation of knowledge to understand many of the intricacies involved in manipulating the human response to fight parasitic infections, and a great deal has been learned from malaria vaccine efforts regarding strategies for developing parasite vaccines. There has been some encouraging progress in the development of a Chagas vaccine in animal models. A prize fund for Chagas could be instrumental in ensuring that these efforts are translated into products that benefit patients.
Collapse
|
43
|
Overstreet MG, Cockburn IA, Zavala F. Protective CD8 T cells against Plasmodium liver stages: immunobiology of an 'unnatural' immune response. Immunol Rev 2008; 225:272-83. [PMID: 18837788 PMCID: PMC2597001 DOI: 10.1111/j.1600-065x.2008.00671.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
SUMMARY Immunization with high doses of irradiated sporozoites delivered by the bites of infected mosquitoes has been shown to induce protective responses against malaria, mediated in part by CD8(+) T cells. In contrast, natural transmission involving low exposure to live sporozoite antigen fails to elicit strong immunity. In this review, we examine how irradiated sporozoite immunization breaks the natural host-parasite interaction and induces protective CD8(+) T cells. Upon biting, the malaria-infected mosquitoes deposit parasites in the skin, many of which eventually exit to the bloodstream and infect hepatocytes. However, certain antigens, including the circumsporozoite (CS) protein, remain in the skin and are presented in the draining lymph node. These antigens prime specific CD8(+) T cells, which migrate to the liver where they eliminate parasitized hepatocytes. We discuss the relevance of the different tissue compartments involved in the induction and effector phases of this response, as well as the cellular requirements for priming and memory development of CD8(+) T cells, which include a complete dependence on dendritic cells and a near absolute need for CD4(+) T-cell help. Finally, we discuss the impact of the immunodominant CS protein on this protection and the implications of these findings for vaccine design.
Collapse
Affiliation(s)
- Michael Glen Overstreet
- Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ian Andrew Cockburn
- Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fidel Zavala
- Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
44
|
Fontanella GH, De Vusser K, Laroy W, Daurelio L, Nocito AL, Revelli S, Contreras R. Immunization with an engineered mutant trans-sialidase highly protects mice from experimental Trypanosoma cruzi infection: a vaccine candidate. Vaccine 2008; 26:2322-34. [PMID: 18403070 DOI: 10.1016/j.vaccine.2008.02.060] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 02/21/2008] [Accepted: 02/28/2008] [Indexed: 01/22/2023]
Abstract
Chagas' disease is a major tropical disease for which a cure for chronic phase does not exist yet. Trypanosoma cruzi trans-sialidase (TS) seems to be involved in relevant processes such as infectivity, host survival and, very importantly, disease pathogenesis. In this study, we show that mice vaccinated with an engineered enzymatically deficient mutant TS containing the catalytic domain without the immunodominant SAPA (Shed Acute Phase Antigen) repeats, were highly protected against T. cruzi infection. Adult male BALB/c mice were immunized with mutant protein, purified from Pichia pastoris yeast, using three inoculations in Freund's adjuvant. All immunized mice were protected against challenge with a lethal dose of T. cruzi trypomastigotes. The protected immunized mice developed no clinical or tissue evidence of infection throughout the study. In contrast, 60-90% mortality and 100% occurrence of myocardial lesions were observed in the non-immunized counterparts. Titers of circulating antibody against TS did not correlate with protection, while anti-SAPA antibodies were coincident with disease severity. Further studies indicated that a single inoculation of mutant recombinant protein in Freund's complete adjuvant was not associated with blood or organic alterations, per se. Mutant TS vaccination seems to be a promising tool for immune intervention strategies in Chagas' disease, aimed at preventing T. cruzi-related heart tissue damage.
Collapse
Affiliation(s)
- Germán H Fontanella
- Instituto de Inmunología, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Argentina
| | | | | | | | | | | | | |
Collapse
|
45
|
Cockburn IA, Chakravarty S, Overstreet MG, García-Sastre A, Zavala F. Memory CD8+ T cell responses expand when antigen presentation overcomes T cell self-regulation. THE JOURNAL OF IMMUNOLOGY 2008; 180:64-71. [PMID: 18097005 DOI: 10.4049/jimmunol.180.1.64] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antimicrobial memory CD8+ T cell responses are not readily expanded by either repeated infections or immunizations. This is a major obstacle to the development of T cell vaccines. Prime-boost immunization with heterologous microbes sharing the same CD8+ epitope can induce a large expansion of the CD8+ response; however, different vectors vary greatly in their ability to boost for reasons that are poorly understood. To investigate how efficient memory T cell expansion can occur, we evaluated immune regulatory events and Ag presentation after secondary immunization with strong and weak boosting vectors. We found that dendritic cells were essential for T cell boosting and that Ag presentation by these cells was regulated by cognate memory CD8+ T cells. When weak boosting vectors were used for secondary immunization, pre-established CD8+ T cells were able to effectively curtail Ag presentation, resulting in limited CD8+ T cell expansion. In contrast, a strong boosting vector, vaccinia virus, induced highly efficient Ag presentation that overcame regulation by cognate T cells and induced large numbers of memory CD8+ T cells to expand. Thus, efficient targeting of Ag to dendritic cells in the face of cognate immunity is an important requirement for T cell expansion.
Collapse
Affiliation(s)
- Ian A Cockburn
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
46
|
Millar J, Dissanayake D, Yang TC, Grinshtein N, Evelegh C, Wan Y, Bramson J. The magnitude of the CD8+ T cell response produced by recombinant virus vectors is a function of both the antigen and the vector. Cell Immunol 2008; 250:55-67. [PMID: 18313652 DOI: 10.1016/j.cellimm.2008.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2007] [Revised: 01/16/2008] [Accepted: 01/17/2008] [Indexed: 10/22/2022]
Abstract
Virus-based recombinant vaccines have proven highly effective at generating protective CD8+ T cell responses. Multiple vector platforms are available, however, little is known about the relative influence of the different vectors on the transgene-specific CD8+ T cell population. To address this question, we compared several characteristics of the CD8+ T cell response elicited by recombinant adenovirus (rAd) and vaccinia virus (rVV). We found that following rAd immunization the transgene-specific CD8+ T cell response peaked around day 12 and was larger and more sustained than the response produced by rVV. In addition, the CD8+ T cell response generated by rAd was directed primarily against the transgene, whereas the CD8+ T cell response produced by rVV principally targeted the vector backbone. In addition, we also observed that transgene selection also impacted on the magnitude of the CD8+ T cell response elicited by both vectors. Despite differences in the magnitude of the anti-transgene CD8+ T cell response, both vectors elicited CD8+ T cell populations with similar cytokine production, functional avidity and cytolytic activity. In addition, plasmid priming prior to immunization with either rAd or rVV only impacted the magnitude of the transgene gene specific CD8+ T cell response. Our study demonstrates that both vector and transgene selection can influence the magnitude of the CD8+ T cell response, but they do not influence functionality.
Collapse
Affiliation(s)
- James Millar
- Center for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, Room MDCL-5025, 1200 Main Street West, Hamilton, Ont., Canada L8N 3Z5
| | | | | | | | | | | | | |
Collapse
|
47
|
Gironès N, Carrasco-Marin E, Cuervo H, Guerrero NA, Sanoja C, John S, Flores-Herráez R, Fernández-Prieto L, Chico-Calero I, Salgado H, Carrión J, Fresno M. Role of Trypanosoma cruzi autoreactive T cells in the generation of cardiac pathology. Ann N Y Acad Sci 2007; 1107:434-44. [PMID: 17804572 DOI: 10.1196/annals.1381.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Chagas disease, caused by Trypanosoma cruzi, affects several million people in Central and South America. About 30% of chronic patients develop cardiomyopathy probably caused by parasite persistence and/or autoimmunity. While several cross-reactive antibodies generated during mammal T. cruzi infection have been described, very few cross-reactive T cells have been identified. We performed adoptive transfer experiments of T cells isolated from chronically infected mice. The results showed the generation of cardiac pathology in the absence of parasites. We also transferred cross-reactive SAPA-specific T cells and observed unspecific alterations in heart repolarization, cardiac inflammatory infiltration, and tissue damage.
Collapse
Affiliation(s)
- Núria Gironès
- Centro de Biología Molecular, CSIC-UAM, Universidad Autónoma de Madrid, Cantoblanco, E-28049 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Miyahira Y. Trypanosoma cruzi infection from the view of CD8+ T cell immunity--an infection model for developing T cell vaccine. Parasitol Int 2007; 57:38-48. [PMID: 17728174 DOI: 10.1016/j.parint.2007.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 07/23/2007] [Accepted: 07/24/2007] [Indexed: 11/28/2022]
Abstract
Chagas' disease is caused by Trypanosoma cruzi (T. cruzi) which was once prevalent in Central and South America. Although the recent success in Triatoma vector control has made the disease being possibly "extinct" in the near future, the development of effective preventive and therapeutic vaccines is still necessary to prevent the resurgence of the neglected infection. In addition to the importance for containing the disease, T. cruzi infection presents unique features for elucidating hosts' immune responses against intracellular infectious agents. Due to its biological capacity for invading into principally any types of cells and for causing systemic infection which damages particularly muscle and neural cells, T cell immunity is critical for resolving its infection. Although T cell-mediated immune responses have been, so far, extensively investigated in viral and bacterial infections, parasitic infection such as malaria has presented epoch-making discovery in T cell immunity. Recent advances in the analyses of T cell-mediated immune responses against T. cruzi infection now make this infectious disease potentially more suitable for detecting subtle immunological changes in hosts' immune defense upon modifying immune system. The current review focuses on the usefulness of T. cruzi infection as a model for developing effective CD8(+) T cell-mediated vaccine against intracellular infectious agents.
Collapse
Affiliation(s)
- Yasushi Miyahira
- Department of Global Infectious Diseases and Tropical Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa City, Saitama 359-8513 Japan.
| |
Collapse
|
49
|
Díez H, Guzmán F, Alba MP, Cuéllar A, Thomas MC, López MC, Rosas F, Velasco V, González JM, Patarroyo ME, Puerta CJ. Immunological and structural characterization of an epitope from the Trypanosoma cruzi KMP-11 protein. Peptides 2007; 28:1520-6. [PMID: 17683828 DOI: 10.1016/j.peptides.2007.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 06/15/2007] [Accepted: 06/18/2007] [Indexed: 11/23/2022]
Abstract
The K1 peptide is an HLA-A*0201-restricted cytotoxic epitope derived from the Trypanosoma cruzi KMP-11 protein, this being the etiological agent of Chagas' disease. This work describes the K1 peptide's secondary structure and its recognition by sera from chagasic patients. Circular dichroism and NMR spectroscopy analysis revealed that the K1 peptide adopts an alpha-helical conformation. Fifty-six percent of individuals had anti-K1 and 86% anti-KMP-11 antibodies by ELISA in the chronic Chagas' group and 28 and 68% in the indeterminate Chagas' group, respectively. By contrast, no reactivity was observed in sera from healthy individuals and tuberculosis patients. Antibody response subclass specificity to the K1 peptide was IgG1 and IgG3. Taken together these results support the idea that the K1 peptide acts as a B-cell-inducer epitope during Chagas' disease.
Collapse
Affiliation(s)
- H Díez
- Laboratorio de Parasitología Molecular, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tarleton RL. Immune system recognition of Trypanosoma cruzi. Curr Opin Immunol 2007; 19:430-4. [PMID: 17651955 DOI: 10.1016/j.coi.2007.06.003] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Accepted: 06/08/2007] [Indexed: 10/23/2022]
Abstract
Innate and adaptive cellular immune recognition is crucial for control of the protozoan parasite Trypanosoma cruzi. T. cruzi triggers both MyD88-dependent and TRIF-dependent innate activation pathways in macrophages and dendritic cells. TLR-2 and TLR-9 recognize GPI anchors and parasite DNA, respectively; however other, as yet undefined receptors and ligands, also appear to be involved in innate recognition. CD8(+) T cells distinguish T. cruzi-infected host cells primarily via robust recognition of MHC-associated peptide epitopes from the large and highly diverse trans-sialidase family of surface proteins. To date there has been minimal investigation of linkages between innate immune recognition in vivo and the generation of adaptive cellular immune responses.
Collapse
Affiliation(s)
- Rick L Tarleton
- Center for Tropical & Emerging Global Diseases and Department of Cellular Biology, Coverdell Center for Biomedical Research, 500 D.W. Brooks Drive, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|