1
|
Pinazo MJ, Malchiodi E, Ioset JR, Bivona A, Gollob KJ, Dutra WO. Challenges and advancements in the development of vaccines and therapies against Chagas disease. THE LANCET. MICROBE 2024; 5:100972. [PMID: 39303738 DOI: 10.1016/j.lanmic.2024.100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, presents a substantial global health burden, affecting millions of individuals worldwide and posing a continual risk of infection. Despite the high mortality and morbidity rates, effective vaccines to prevent infection by the parasite remain elusive, and the drugs currently available are suboptimal. Understanding the intricate dynamics of parasite-host interactions and the resulting immune responses, which contribute to both protection and pathology, is crucial for the development of effective vaccines and therapies against Chagas disease. In this Series paper, we discuss the challenges associated with discovering and translating prophylactic and therapeutic strategies from the laboratory bench to clinical application. We highlight ongoing efforts in vaccine and new drug development, with a focus on more advanced candidates for vaccines and drugs. We also discuss potential solutions, emphasising the importance of collaborative research efforts, sustained funding, and a comprehensive understanding of host-parasite interactions and immunopathology to advance the development of new vaccines and therapies against Chagas disease.
Collapse
Affiliation(s)
| | - Emilio Malchiodi
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Instituto de Estudios de la Inmunidad Humoral (IDEHU) and Instituto de Microbiologia y Parasitologia Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | | | - Augusto Bivona
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Instituto de Estudios de la Inmunidad Humoral (IDEHU) and Instituto de Microbiologia y Parasitologia Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Kenneth J Gollob
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Belo Horizonte, Brazil
| | - Walderez O Dutra
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Belo Horizonte, Brazil; Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
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
|
Ramos-Vega A, Monreal-Escalante E, Rosales-Mendoza S, Bañuelos-Hernández B, Dumonteil E, Angulo C. Trypanosoma cruzi Tc24 Antigen Expressed and Orally Delivered by Schizochytrium sp. Microalga is Immunogenic in Mice. Mol Biotechnol 2024; 66:1376-1388. [PMID: 37344711 DOI: 10.1007/s12033-023-00763-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/22/2023] [Indexed: 06/23/2023]
Abstract
Chagas disease-caused by the parasite Trypanosoma cruzi-is a neglected tropical disease for which available drugs are not fully effective in the chronic stage and a vaccine is not available yet. Microalgae represent a promising platform for the production and oral delivery of low-cost vaccines. Herein, we report a vaccine prototype against T. cruzi produced in a microalgae platform, based on the candidate antigen Tc24 with a C terminus fusion with the Co1 peptide (Tc24:Co1 vaccine prototype). After modeling the tertiary structure, in silico studies suggested that the chimeric protein is antigenic, not allergenic, and molecular docking indicated binding with Toll-like receptors 2 and 4. Thus, Tc24:Co1 was expressed in the marine microalga Schizochytrium sp., and Western blot confirmed the expression at 48 h after induction, with a yield of 632 µg/L of algal culture (300 μg/g of lyophilized algal cells) as measured by the enzyme-linked immunosorbent assay (ELISA). Upon oral administration of whole-cell Schizochytrium sp. expressing Tc24:Co1 (7.5 µg or 15 µg of Tc24:Co1 doses) in mice, specific serum IgG and intestinal mucosa IgA responses were detected in addition to an increase in serum Th1/Th2 cytokines. In conclusion, Schizochytrium sp.-expressing Tc24:Co1 is a promising oral vaccine prototype to be evaluated in an animal model of Trypanosoma cruzi infection.
Collapse
Affiliation(s)
- Abel Ramos-Vega
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., La Paz, BCS, Mexico
| | - Elizabeth Monreal-Escalante
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., La Paz, BCS, Mexico.
- CONACYT-Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Col. Playa Palo de Santa Rita Sur, Av. Instituto Politécnico Nacional 195, CP. 23096, La Paz, BCS, Mexico.
| | - Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, UASLP, San Luis Potosí, Mexico
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, UASLP, San Luis Potosí, Mexico
| | | | - Eric Dumonteil
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Carlos Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., La Paz, BCS, Mexico.
| |
Collapse
|
4
|
Soprano LL, Ferrero MR, Jacobs T, Couto AS, Duschak VG. Hallmarks of the relationship between host and Trypanosoma cruzi sulfated glycoconjugates along the course of Chagas disease. Front Cell Infect Microbiol 2023; 13:1028496. [PMID: 37256110 PMCID: PMC10225527 DOI: 10.3389/fcimb.2023.1028496] [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: 08/26/2022] [Accepted: 04/17/2023] [Indexed: 06/01/2023] Open
Abstract
American Trypanosomiasis or Chagas disease (ChD), a major problem that is still endemic in large areas of Latin America, is caused by Trypanosoma cruzi. This agent holds a major antigen, cruzipain (Cz). Its C-terminal domain (C-T) is retained in the glycoprotein mature form and bears several post-translational modifications. Glycoproteins containing sulfated N-linked oligosaccharides have been mostly implicated in numerous specific procedures of molecular recognition. The presence of sulfated oligosaccharides was demonstrated in Cz, also in a minor abundant antigen with serine-carboxypeptidase (SCP) activity, as well as in parasite sulfatides. Sulfate-bearing glycoproteins in Trypanosomatids are targets of specific immune responses. T. cruzi chronically infected subjects mount specific humoral immune responses to sulfated Cz. Unexpectedly, in the absence of infection, mice immunized with C-T, but not with sulfate-depleted C-T, showed ultrastructural heart anomalous pathological effects. Moreover, the synthetic anionic sugar conjugate GlcNAc6SO3-BSA showed to mimic the N-glycan-linked sulfated epitope (sulfotope) humoral responses that natural Cz elicits. Furthermore, it has been reported that sulfotopes participate via the binding of sialic acid Ig-like-specific lectins (Siglecs) to sulfosialylated glycoproteins in the immunomodulation by host-parasite interaction as well as in the parasite infection process. Strikingly, recent evidence involved Cz-sulfotope-specific antibodies in the immunopathogenesis and infection processes during the experimental ChD. Remarkably, sera from chronically T. cruzi-infected individuals with mild disease displayed higher levels of IgG2 antibodies specific for sulfated glycoproteins and sulfatides than those with more severe forms of the disease, evidencing that T. cruzi sulfotopes are antigenic independently of the sulfated glycoconjugate type. Ongoing assays indicate that antibodies specific for sulfotopes might be considered biomarkers of human cardiac ChD progression, playing a role as predictors of stability from the early mild stages of chronic ChD.
Collapse
Affiliation(s)
- Luciana L. Soprano
- Area of Protein Biochemistry and Parasite Glycobiology, Research Department National Institute of Parasitology (INP)”Dr. Mario Fatala Chaben”, National Administration of Health Institutes (ANLIS)-Malbrán, National Health Department, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Maximiliano R. Ferrero
- Max-Planck Heart and Lung Laboratory, Research Institute in Biomedicine in Buenos Aires (IBioBA), Argentine-Department of Internal Medicine II, University Medical Center Giessen and Marburg, Giessen, Germany
| | - Thomas Jacobs
- Immunology Department, Bernhard Notch Institute of Tropical Medicine, Hamburg, Germany
| | - Alicia S. Couto
- Faculty in Exact and Natural Sciences (FCEN), Chemical Organic Department-National Council of Scientific and Technical Research (CONICET), Center of CarboHydrates (CHIHIDECAR), University of Buenos Aires, Buenos Aires, Argentina
| | - Vilma G. Duschak
- Area of Protein Biochemistry and Parasite Glycobiology, Research Department National Institute of Parasitology (INP)”Dr. Mario Fatala Chaben”, National Administration of Health Institutes (ANLIS)-Malbrán, National Health Department, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| |
Collapse
|
5
|
S. I. C, Julio AP, De Souza W, A. M. P. Editorial: Understanding anti-trypanosomatid immune responses: The key to developing protective strategies against them. Front Immunol 2022; 13:993315. [PMID: 36211393 PMCID: PMC9535140 DOI: 10.3389/fimmu.2022.993315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Cazorla S. I.
- Centro de Referencia para Lactobacilos – CERELA (CONICET), Tucumán, Argentina
- Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
- *Correspondence: Cazorla S. I.,
| | - Alonso-Padilla Julio
- Instituto Salud Global de Barcelona (ISGlobal), Barcelona, Spain
- CIBERINFEC, ISCIII—CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - W. De Souza
- Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- CMABio, Universidade do Estado do Amazonas-UEA, Manaus, Amazonas, Brazil
| | - Padilla A. M.
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
6
|
Soprano LL, Ferrero MR, Landoni M, García GA, Esteva MI, Couto AS, Duschak VG. Cruzipain Sulfotopes-Specific Antibodies Generate Cardiac Tissue Abnormalities and Favor Trypanosoma cruzi Infection in the BALB/c Mice Model of Experimental Chagas Disease. Front Cell Infect Microbiol 2022; 11:814276. [PMID: 35059328 PMCID: PMC8763857 DOI: 10.3389/fcimb.2021.814276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Trypanosoma cruzi cruzipain (Cz) bears a C-terminal domain (C-T) that contains sulfated epitopes “sulfotopes” (GlcNAc6S) on its unique N-glycosylation site. The effects of in vivo exposure to GlcNAc6S on heart tissue ultrastructure, immune responses, and along the outcome of infection by T. cruzi, were evaluated in a murine experimental model, BALB/c, using three independent strategies. First, mice were pre-exposed to C-T by immunization. C-T-immunized mice (C-TIM) showed IgG2a/IgG1 <1, induced the production of cytokines from Th2, Th17, and Th1 profiles with respect to those of dC-TIM, which only induced IL-10 respect to the control mice. Surprisingly, after sublethal challenge, both C-TIM and dC-TIM showed significantly higher parasitemia and mortality than the control group. Second, mice exposed to BSA-GlcNAc6S as immunogen (BSA-GlcNAc6SIM) showed: severe ultrastructural cardiac alterations while BSA-GlcNAcIM conserved the regular tissue architecture with slight myofibril changes; a strong highly specific humoral-immune-response reproducing the IgG-isotype-profile obtained with C-TIM; and a significant memory-T-cell-response demonstrating sulfotope-immunodominance with respect to BSA-GlcNAcIM. After sublethal challenge, BSA-GlcNAc6SIM showed exacerbated parasitemias, despite elevated IFN-γ levels were registered. In both cases, the abrogation of ultrastructural alterations when using desulfated immunogens supported the direct involvement of sulfotopes and/or indirect effect through their specific antibodies, in the induction of tissue damage. Finally, a third strategy using a passive transference of sulfotope-specific antibodies (IgG-GlcNAc6S) showed the detrimental activity of IgG-GlcNAc6S on mice cardiac tissue, and mice treated with IgG-GlcNAc6S after a sublethal dose of T. cruzi, surprisingly reached higher parasitemias than control groups. These findings confirmed the indirect role of the sulfotopes, via their IgG-GlcNAc6S, both in the immunopathogenicity as well as favoring T. cruzi infection.
Collapse
Affiliation(s)
- Luciana L Soprano
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina
| | - Maximiliano R Ferrero
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina
| | - Malena Landoni
- Organic Chemistry Department, Natural and Exact Sciences Faculty; Research Center in Carbohydrates (CIHIDECAR), University of Buenos Aires, Buenos Aires, Argentina.,Ministry of Science, Technology and Innovation, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Gabriela A García
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina.,Ministry of Science, Technology and Innovation, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Mónica I Esteva
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina
| | - Alicia S Couto
- Organic Chemistry Department, Natural and Exact Sciences Faculty; Research Center in Carbohydrates (CIHIDECAR), University of Buenos Aires, Buenos Aires, Argentina.,Ministry of Science, Technology and Innovation, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Vilma G Duschak
- Area of Biochemistry of Proteins and Glycobiology of Parasites, Research Department, National Institute of Parasitology "Dr. Mario Fatala Chaben", ANLIS-Malbrán, Health Department, Ciudad Autónoma de Buenos Aires (CABA, 1063), Buenos Aires, Argentina.,Ministry of Science, Technology and Innovation, National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| |
Collapse
|
7
|
Rawal K, Sinha R, Abbasi BA, Chaudhary A, Nath SK, Kumari P, Preeti P, Saraf D, Singh S, Mishra K, Gupta P, Mishra A, Sharma T, Gupta S, Singh P, Sood S, Subramani P, Dubey AK, Strych U, Hotez PJ, Bottazzi ME. Identification of vaccine targets in pathogens and design of a vaccine using computational approaches. Sci Rep 2021; 11:17626. [PMID: 34475453 PMCID: PMC8413327 DOI: 10.1038/s41598-021-96863-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Antigen identification is an important step in the vaccine development process. Computational approaches including deep learning systems can play an important role in the identification of vaccine targets using genomic and proteomic information. Here, we present a new computational system to discover and analyse novel vaccine targets leading to the design of a multi-epitope subunit vaccine candidate. The system incorporates reverse vaccinology and immuno-informatics tools to screen genomic and proteomic datasets of several pathogens such as Trypanosoma cruzi, Plasmodium falciparum, and Vibrio cholerae to identify potential vaccine candidates (PVC). Further, as a case study, we performed a detailed analysis of the genomic and proteomic dataset of T. cruzi (CL Brenner and Y strain) to shortlist eight proteins as possible vaccine antigen candidates using properties such as secretory/surface-exposed nature, low transmembrane helix (< 2), essentiality, virulence, antigenic, and non-homology with host/gut flora proteins. Subsequently, highly antigenic and immunogenic MHC class I, MHC class II and B cell epitopes were extracted from top-ranking vaccine targets. The designed vaccine construct containing 24 epitopes, 3 adjuvants, and 4 linkers was analysed for its physicochemical properties using different tools, including docking analysis. Immunological simulation studies suggested significant levels of T-helper, T-cytotoxic cells, and IgG1 will be elicited upon administration of such a putative multi-epitope vaccine construct. The vaccine construct is predicted to be soluble, stable, non-allergenic, non-toxic, and to offer cross-protection against related Trypanosoma species and strains. Further, studies are required to validate safety and immunogenicity of the vaccine.
Collapse
Affiliation(s)
- Kamal Rawal
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India.
| | - Robin Sinha
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Bilal Ahmed Abbasi
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Amit Chaudhary
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Swarsat Kaushik Nath
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Priya Kumari
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - P Preeti
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Devansh Saraf
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Shachee Singh
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Kartik Mishra
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Pranjay Gupta
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Astha Mishra
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Trapti Sharma
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Srijanee Gupta
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Prashant Singh
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Shriya Sood
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Preeti Subramani
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Aman Kumar Dubey
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Biology, Baylor University, Waco, TX, USA
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Biology, Baylor University, Waco, TX, USA
| |
Collapse
|
8
|
Dumonteil E, Herrera C. The Case for the Development of a Chagas Disease Vaccine: Why? How? When? Trop Med Infect Dis 2021; 6:tropicalmed6010016. [PMID: 33530605 PMCID: PMC7851737 DOI: 10.3390/tropicalmed6010016] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/06/2021] [Accepted: 01/19/2021] [Indexed: 11/16/2022] Open
Abstract
Chagas disease is a major neglected tropical disease, transmitted predominantly by triatomine insect vectors, but also through congenital and oral routes. While endemic in the Americas, it has turned into a global disease. Because of the current drug treatment limitations, a vaccine would represent a major advancement for better control of the disease. Here, we review some of the rationale, advances, and challenges for the ongoing development of a vaccine against Chagas disease. Recent pre-clinical studies in murine models have further expanded (i) the range of vaccine platforms and formulations tested; (ii) our understanding of the immune correlates for protection; and (iii) the extent of vaccine effects on cardiac function, beyond survival and parasite burden. We further discuss outstanding issues and opportunities to move Chagas disease development forward in the near future.
Collapse
|
9
|
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: 0.8] [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
|
10
|
Antonoglou MB, Sánchez Alberti A, Redolfi DM, Bivona AE, Fernández Lynch MJ, Noli Truant S, Sarratea MB, Iannantuono López LV, Malchiodi EL, Fernández MM. Heterologous Chimeric Construct Comprising a Modified Bacterial Superantigen and a Cruzipain Domain Confers Protection Against Trypanosoma cruzi Infection. Front Immunol 2020; 11:1279. [PMID: 32695105 PMCID: PMC7338481 DOI: 10.3389/fimmu.2020.01279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/20/2020] [Indexed: 01/18/2023] Open
Abstract
Chagas disease is an endemic chronic parasitosis in Latin America affecting more than 7 million people. Around 100 million people are currently at risk of acquiring the infection; however, no effective vaccine has been developed yet. Trypanosoma cruzi is the etiological agent of this parasitosis and as an intracellular protozoan it can reside within different tissues, mainly muscle cells, evading host immunity and allowing progression towards the chronic stage of the disease. Considering this intracellular parasitism triggers strong cellular immunity that, besides being necessary to limit infection, is not sufficient to eradicate the parasite from tissues, a differential immune response is required and new strategies for vaccines against Chagas disease need to be explored. In this work, we designed, cloned and expressed a chimeric molecule, named NCz-SEGN24A, comprising a parasite antigen, the N-terminal domain of the major cysteine protease of T. cruzi, cruzipain (Nt-Cz), and a non-toxic form of the staphylococcal superantigen (SAg) G, SEG, with the residue Asn24 mutated to Ala (N24A). The mutant SAg SEGN24A, retains its ability to trigger classical activation of macrophages without inducing T cell apoptosis. To evaluate, as a proof of concept, the immunogenicity and efficacy of the chimeric immunogen vs. its individual antigens, C3H mice were immunized intramuscularly with NCz-SEGN24A co-adjuvanted with CpG-ODN, or the recombinant proteins Nt-Cz plus SEGN24A with the same adjuvant. Vaccinated mice significantly produced Nt-Cz-specific IgG titers after immunization and developed higher IgG2a than IgG1 titers. Specific cell-mediated immunity was assessed by in-vivo DTH and significant responses were obtained. To assess protection, mice were challenged with trypomastigotes of T. cruzi. Both schemes reduced the parasite load throughout the acute phase, but only mice immunized with NCz-SEGN24A showed significant differences against control; moreover, these mice maintained 100% survival. These results encourage testing mutated superantigens fused to specific antigens as immune modulators against pathogens.
Collapse
Affiliation(s)
- María Belén Antonoglou
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andrés Sánchez Alberti
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela María Redolfi
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Augusto Ernesto Bivona
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Julieta Fernández Lynch
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sofía Noli Truant
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Belén Sarratea
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Laura Valeria Iannantuono López
- Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Emilio Luis Malchiodi
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marisa Mariel Fernández
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
11
|
Evaluation of the Immunoprotective Potential of Recombinant Paraflagellar Rod Proteins of Trypanosoma evansi in Mice. Vaccines (Basel) 2020; 8:vaccines8010084. [PMID: 32059486 PMCID: PMC7157580 DOI: 10.3390/vaccines8010084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 12/15/2022] Open
Abstract
Trypanosomosis, caused by Trypanosoma evansi, is an economically significant disease of livestock. Systematic antigenic variation by the parasite has undermined prospects for the development of a protective vaccine that targets the immunodominant surface antigens, encouraging exploration of alternatives. The paraflagellar rod (PFR), constituent proteins of the flagellum, are prominent non-variable vaccine candidates for T. evansi owing to their strategic location. Two major PFR constituent proteins, PFR1 (1770bp) and PFR2 (1800bp), were expressed using Escherichia coli. Swiss albino mice were immunized with the purified recombinant TePFR1 (89KDa) and TePFR2 (88KDa) proteins, as well as with the mix of the combined proteins at equimolar concentrations, and subsequently challenged with virulent T. evansi. The PFR-specific humoral response was assessed by ELISA. Cytometric bead-based assay was used to measure the cytokine response and flow cytometry for quantification of the cytokines. The recombinant TePFR proteins induced specific humoral responses in mice, including IgG1 followed by IgG2a and IgG2b. A balanced cytokine response induced by rTePFR 1 and 2 protein vaccination associated with extended survival and improved control of parasitemia following lethal challenge. The observation confirms the immunoprophylactic potential of the covert antigens of T. evansi.
Collapse
|
12
|
Cabrera G, Marcipar I. Vaccines and the regulatory arm of the immune system. An overview from the Trypanosoma cruzi infection model. Vaccine 2019; 37:3628-3637. [PMID: 31155420 DOI: 10.1016/j.vaccine.2019.05.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 04/13/2019] [Accepted: 05/03/2019] [Indexed: 01/06/2023]
Abstract
The knowledge that the immune system is composed of a regulatory/suppressor arm added a new point of view to better understand the nature of several pathologies including cancer, transplants, infections and autoimmune diseases. The striking discoveries concerning molecules and cells involved in this kind of regulation were followed by the elucidation of equally notable mechanisms used by several pathogens to manipulate the host immune system. Vaccines against pathogens are an invaluable tool developed to help the immune system cope with a potential infection or prevent disease pathology. Nowadays, there is accumulated evidence indicating that the powerful stimulation capacity of vaccines influences not only the effector arm of the immune system but also cells with regulatory/suppressor capacity, such as myeloid derived suppressor cells (MDSCs) and Foxp3+ regulatory T cells (Tregs). Trypanosoma cruzi (T. cruzi) is a protozoan parasite with a complex life cycle that has evolved several strategies to influence the regulatory immune response. Although diverse vaccine formulations have been able to stimulate the effector response, achieving non-sterilizing protection against T. cruzi, the influence of the vaccine candidates on the regulatory machinery has scarcely been assessed. This fact may not only reveal important information concerning how vaccines may influence cells with regulatory/suppressor capacity but also open the possibility to analyze whether vaccines are able to disrupt the mechanisms used by some pathogens to manipulate the host regulatory circuits. The aim of this review is to summarize and discuss available data related to the role of cellular components, like MDSCs and Foxp3+ Tregs, during T. cruzi infection, and the potential utility of those populations as additional targets for the rational design of vaccines.
Collapse
Affiliation(s)
- Gabriel Cabrera
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| | - Iván Marcipar
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| |
Collapse
|
13
|
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: 2.0] [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
|
14
|
Laurella LC, Cerny N, Bivona AE, Sánchez Alberti A, Giberti G, Malchiodi EL, Martino VS, Catalan CA, Alonso MR, Cazorla SI, Sülsen VP. Assessment of sesquiterpene lactones isolated from Mikania plants species for their potential efficacy against Trypanosoma cruzi and Leishmania sp. PLoS Negl Trop Dis 2017; 11:e0005929. [PMID: 28945741 PMCID: PMC5629014 DOI: 10.1371/journal.pntd.0005929] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/05/2017] [Accepted: 09/04/2017] [Indexed: 12/16/2022] Open
Abstract
Four sesquiterpene lactones, mikanolide, deoxymikanolide, dihydromikanolide and scandenolide, were isolated by a bioassay-guided fractionation of Mikania variifolia and Mikania micrantha dichloromethane extracts. Mikanolide and deoxymikanolide were the major compounds in both extracts (2.2% and 0.4% for Mikania variifolia and 21.0% and 6.4% for Mikania micrantha respectively, calculated on extract dry weight). Mikanolide, deoxymikanolide and dihydromikanolide were active against Trypanosoma cruzi epimastigotes (50% inhibitory concentrations of 0.7, 0.08 and 2.5 μg/mL, for each compound respectively). These sesquiterpene lactones were also active against the bloodstream trypomastigotes (50% inhibitory concentrations for each compound were 2.1, 1.5 and 0.3 μg/mL, respectively) and against amastigotes (50% inhibitory concentrations for each compound were 4.5, 6.3 and 8.5 μg/mL, respectively). By contrast, scandenolide was not active on Trypanosoma cruzi. Besides, mikanolide and deoxymikanolide were also active on Leishmania braziliensis promastigotes (50% inhibitory concentrations of 5.1 and 11.5 μg/mL, respectively). The four sesquiterpene lactones were tested for their cytotoxicity on THP 1 cells. Deoxymikanolide presented the highest selectivity index for trypomastigotes (SI = 54) and amastigotes (SI = 12.5). In an in vivo model of Trypanosoma cruzi infection, deoxymikanolide was able to decrease the parasitemia and the weight loss associated to the acute phase of the parasite infection. More importantly, while 100% of control mice died by day 22 after receiving a lethal T. cruzi infection, 70% of deoxymikanolide-treated mice survived. We also observed that this compound increased TNF-α and IL-12 production by macrophages, which could contribute to control T. cruzi infection.
Collapse
Affiliation(s)
- Laura C. Laurella
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacognosia, Buenos Aires, Argentina
| | - Natacha Cerny
- CONICET—Universidad Nacional de Luján, Instituto de Ecología y Desarrollo Sustentable (INEDES), Luján, Argentina
| | - Augusto E. Bivona
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, Argentina, Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- CONICET- Universidad de Buenos Aires, Instituto de Microbiología y Parasitología Médica—CONICET (IMPaM), Facultad de Medicina, Piso 13, Buenos Aires, Argentina
| | - Andrés Sánchez Alberti
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, Argentina, Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- CONICET- Universidad de Buenos Aires, Instituto de Microbiología y Parasitología Médica—CONICET (IMPaM), Facultad de Medicina, Piso 13, Buenos Aires, Argentina
| | - Gustavo Giberti
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacognosia, Buenos Aires, Argentina
- CONICET–Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco—CONICET (IQUIMEFA), Buenos Aires, Argentina
| | - Emilio L. Malchiodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, Argentina, Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- CONICET- Universidad de Buenos Aires, Instituto de Microbiología y Parasitología Médica—CONICET (IMPaM), Facultad de Medicina, Piso 13, Buenos Aires, Argentina
| | - Virginia S. Martino
- CONICET–Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco—CONICET (IQUIMEFA), Buenos Aires, Argentina
| | - Cesar A. Catalan
- CONICET–Universidad Nacional de Tucumán, Instituto de Química del Noroeste—CONICET (INQUINOA), Ayacucho 471 (T4000INI), San Miguel de Tucumán, Argentina
| | - María Rosario Alonso
- CONICET–Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco—CONICET (IQUIMEFA), Buenos Aires, Argentina
| | - Silvia I. Cazorla
- CONICET- Universidad de Buenos Aires, Instituto de Microbiología y Parasitología Médica—CONICET (IMPaM), Facultad de Medicina, Piso 13, Buenos Aires, Argentina
- CONICET–Centro de Referencia para Lactobacilos (CERELA), Batalla de Chacabuco 145, San Miguel de Tucumán, Argentina
- * E-mail: (VPS); (SIC)
| | - Valeria P. Sülsen
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacognosia, Buenos Aires, Argentina
- CONICET–Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco—CONICET (IQUIMEFA), Buenos Aires, Argentina
- * E-mail: (VPS); (SIC)
| |
Collapse
|
15
|
Prochetto E, Roldán C, Bontempi IA, Bertona D, Peverengo L, Vicco MH, Rodeles LM, Pérez AR, Marcipar IS, Cabrera G. Trans-sialidase-based vaccine candidate protects against Trypanosoma cruzi infection, not only inducing an effector immune response but also affecting cells with regulatory/suppressor phenotype. Oncotarget 2017; 8:58003-58020. [PMID: 28938533 PMCID: PMC5601629 DOI: 10.18632/oncotarget.18217] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/08/2017] [Indexed: 01/07/2023] Open
Abstract
Prophylactic and/or therapeutic vaccines have an important potential to control Trypanosoma cruzi (T. cruzi)infection. The involvement of regulatory/suppressor immune cells after an immunization treatment and T. cruzi infection has never been addressed. Here we show that a new trans-sialidase-based immunogen (TSf) was able to confer protection, correlating not only with beneficial changes in effector immune parameters, but also influencing populations of cells related to immune control. Regarding the effector response, mice immunized with TSf showed a TS-specific antibody response, significant delayed-type hypersensitivity (DTH) reactivity and increased production of IFN-γ by CD8+ splenocytes. After a challenge with T. cruzi, TSf-immunized mice showed 90% survival and low parasitemia as compared with 40% survival and high parasitemia in PBS-immunized mice. In relation to the regulatory/suppressor arm of the immune system, after T. cruzi infection TSf-immunized mice showed an increase in spleen CD4+ Foxp3+ regulatory T cells (Treg) as compared to PBS-inoculated and infected mice. Moreover, although T. cruzi infection elicited a notable increase in myeloid derived suppressor cells (MDSC) in the spleen of PBS-inoculated mice, TSf-immunized mice showed a significantly lower increase of MDSC. Results presented herein highlight the need of studying the immune response as a whole when a vaccine candidate is rationally tested.
Collapse
Affiliation(s)
- Estefanía Prochetto
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Carolina Roldán
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Iván A Bontempi
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Daiana Bertona
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Luz Peverengo
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Miguel H Vicco
- Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Luz M Rodeles
- Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ana R Pérez
- IDICER-CONICET and Instituto de Inmunología, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Santa Fe, Argentina
| | - Iván S Marcipar
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gabriel Cabrera
- Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| |
Collapse
|
16
|
Matos MN, Cazorla SI, Schulze K, Ebensen T, Guzmán CA, Malchiodi EL. Immunization with Tc52 or its amino terminal domain adjuvanted with c-di-AMP induces Th17+Th1 specific immune responses and confers protection against Trypanosoma cruzi. PLoS Negl Trop Dis 2017; 11:e0005300. [PMID: 28234897 PMCID: PMC5342303 DOI: 10.1371/journal.pntd.0005300] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/08/2017] [Accepted: 01/03/2017] [Indexed: 12/21/2022] Open
Abstract
The development of new adjuvants enables fine modulation of the elicited immune responses. Ideally, the use of one or more adjuvants should result in the induction of a protective immune response against the specific pathogen. We have evaluated the immune response and protection against Trypanosoma cruzi infection in mice vaccinated with recombinant Tc52 or its N- and C-terminal domains (NTc52 and CTc52) adjuvanted either with the STING (Stimulator of Interferon Genes) agonist cyclic di-AMP (c-di-AMP), a pegylated derivative of α-galactosylceramide (αGC-PEG), or oligodeoxynucleotides containing unmethylated CpG motifs (ODN-CpG). All groups immunized with the recombinant proteins plus adjuvant: Tc52+c-di-AMP, NTc52+c-di-AMP, CTc52+c-di-AMP, NTc52+c-di-AMP+αGC-PEG, NTc52+CpG, developed significantly higher anti-Tc52 IgG titers than controls. Groups immunized with c-di-AMP and Tc52, NTc52 or CTc52 showed the highest Tc52-specific IgA titers in nasal lavages. All groups immunized with the recombinant proteins plus adjuvant developed a strong specific cellular immune response in splenocytes and lymph node cells with significant differences for groups immunized with c-di-AMP and Tc52, NTc52 or CTc52. These groups also showed high levels of Tc52-specific IL-17 and IFN-γ producing cells, while NTc52+CpG group only showed significant difference with control in IFN-γ producing cells. Groups immunized with c-di-AMP and Tc52, NTc52 or CTc52 developed predominantly a Th17 and Th1immune response, whereas for NTc52+CpG it was a dominant Th1 response. It was previously described that αGC-PEG inhibits Th17 differentiation by activating NKT cells. Thus, in this work we have also included a group immunized with both adjuvants (NTc52+c-di-AMP+αGC-PEG) with the aim to modulate the Th17 response induced by c-di-AMP. This group showed a significant reduction in the number of Tc52-specific IL-17 producing splenocytes, as compared to the group NTc52+c-di-AMP, which has in turn correlated with a reduction in protection against infection. These results suggest that the Th17 immune response developed after immunizing with NTc52+c-di-AMP could have a protective role against T. cruzi infection. Groups NTc52+c-di-AMP, Tc52+c-di-AMP and NTc52PB, were the ones that showed better protection against infection with lower parasitemia and weight loss, and higher survival. Chagas disease is a parasitic disease caused by a protozoan parasite (Trypanosoma cruzi) which has a complex life cycle including insect vector and mammalians. In Latin America, 7–10 million people are infected, 100 million people are at risk of infection, and about 56,000 new infection cases and 12,000 deaths are registered annually. Migration spread the geographic distribution of the disease to North America and Europe. The infection in humans has an initial acute stage followed by a chronic stage where up to 30% of patients develop cardiac alterations and 10% develop digestive, neurological or mixed alterations. The acute infection is hardly detected and there is not drug to treat the chronic infection. Thus, there is an urgent need for prophylactic and therapeutic vaccines development. Several attempts to find a vaccine antigen has been made and the protein Tc52 is a good candidate. In a vaccine composition, as important as the antigen is the adjuvants, which are substances able to increase, improve or modified the immune response. This research provides information about the immune response and protection against Trypanosoma cruzi infection elicited by Tc52 or portions of this molecule using different adjuvants.
Collapse
Affiliation(s)
- Marina N. Matos
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Silvia I. Cazorla
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Emilio L. Malchiodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
17
|
Matthews QL, Farrow AL, Rachakonda G, Gu L, Nde P, Krendelchtchikov A, Pratap S, Sakhare SS, Sabbaj S, Lima MF, Villalta F. Epitope Capsid-Incorporation: New Effective Approach for Vaccine Development for Chagas Disease. Pathog Immun 2016; 1:214-233. [PMID: 27709126 PMCID: PMC5046838 DOI: 10.20411/pai.v1i2.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Previously we reported that a hexon-modified adenovirus (Ad) vector containing the invasive neutralizing epitope of Trypanosoma cruzi (T. cruzi) trypomastigote gp83 (Ad5-gp83) provided immunoprotection against T. cruzi infection. The purpose of this work was to design an improved vaccine for T. cruzi using a novel epitope capsid incorporation strategy. Thus, we evaluated the immunoprotection raised by co-immunization with Ad5-gp83 and an Ad vector containing an epitope (ASP-M) of the T. cruzi amastigote surface protein 2. Methods: Protein IX (pIX)-modified Ad vector (Ad5-pIX-ASP-M) was generated, characterized, and validated. C3H/He mice were immunized with Ad5-pIX-ASP-M and Ad5-gp83 and the cell-mediated responses were evaluated by enzyme-linked immunospot (ELISPOT) assay and intracellular staining. Immunized mice were challenged with T. cruzi to evaluate the vaccine efficacy. Results: Our findings indicate that Ad5-pIX-ASP-M was viable. Specific CD8+ T-cell mediated responses prior to the challenge show an increase in IFNγ and TNFα production. A single immunization with Ad5-pIX-ASP-M provided protection from T. cruzi infection, but co-immunizations with Ad5-pIX-ASP-M and Ad5-gp83 provided a higher immunoprotection and increased survival rate of mice. Conclusions: Overall, these results suggest that the combination of gp83 and ASP-M specific epitopes onto the capsid-incorporated adenoviruses would provide superior protection against Chagas disease as compared with Ad5-gp83 alone.
Collapse
Affiliation(s)
- Qiana L Matthews
- Department of Biological Sciences, Alabama State University, Montgomery, AL; Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, AL
| | - Anitra L Farrow
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, AL
| | - Girish Rachakonda
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | - Linlin Gu
- Division of Pulmonary, Allergy and Critical Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Pius Nde
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | | | - Siddharth Pratap
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | - Shruti S Sakhare
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | - Steffanie Sabbaj
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, AL
| | - Maria F Lima
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| | - Fernando Villalta
- Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, TN
| |
Collapse
|
18
|
Abstract
ABSTRACT
Parasites are an important cause of human disease worldwide. The clinical severity and outcome of parasitic disease is often dependent on the immune status of the host. Specific parasitic diseases discussed in this chapter are amebiasis, giardiasis, cryptosporidiosis, cyclosporiasis, cystoisosporiasis, microsporidosis, granulomatous amebic encephalitis, toxoplasmosis, leishmaniasis, Chagas disease, malaria, babesiosis, strongyloidiasis, and scabies.
Collapse
|
19
|
Basso B, Marini V, Gauna D, Frias M. Vaccination of dogs with Trypanosoma rangeli induces antibodies against Trypanosoma cruzi in a rural area of Córdoba, Argentina. Mem Inst Oswaldo Cruz 2016; 111:271-4. [PMID: 27074257 PMCID: PMC4830117 DOI: 10.1590/0074-02760160019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/02/2016] [Indexed: 12/11/2022] Open
Abstract
Dogs play a major role in the domestic cycle of Trypanosoma cruzi, acting as reservoirs. In a previous work we have developed a model of vaccination of dogs in captivity with nonpathogenic Trypanosoma rangeli epimastigotes, resulting in the production of protective antibodies against T. cruzi, with dramatic decrease of parasitaemia upon challenge with 100,000 virulent forms of this parasite. The aim of this work was to evaluate the immunogenicity of this vaccine in dogs living in a rural area. Domestic dogs, free from T. cruzi infection, received three immunisations with fixed T. rangeli epimastigotes. Dogs were not challenged with T. cruzi, but they were left in their environment. This immunisation induced antibodies against T. cruzi for more than three years in dogs in their natural habitat, while control dogs remained serologically negative.
Collapse
Affiliation(s)
- Beatriz Basso
- School of Medicine, National University of Córdoba, Córdoba, Argentina
| | | | | | - Maria Frias
- School of Medicine, National University of Córdoba, Córdoba, Argentina
| |
Collapse
|
20
|
Nascentes GAN, Hernández CG, Rabelo RADS, Coelho RF, Morais FRD, Marques T, Batista LR, Meira WSF, Oliveira CJFD, Lages Silva E, Ramírez LE. The Driving of Immune Response by Th1 Adjuvants in Immunization of Mice with Trypanosoma cruzi marinkellei Elicits a Controversial Infection Control. Vector Borne Zoonotic Dis 2016; 16:317-25. [PMID: 26959861 DOI: 10.1089/vbz.2015.1874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In previous studies, we have demonstrated that inoculation with a Trypanosoma cruzi marinkellei (avirulent RM1 strain) was able to reduce parasitemia in mice challenged with T. cruzi, although it was not able to prevent histopathological lesions. Th1 response stimulation by immunization is necessary for T. cruzi infection control, but the resistance is also dependent on immunoregulatory mechanisms, which can be induced by adjuvants. Thus, we evaluated whether inoculation of T. cruzi marinkellei associated with administration of different adjuvants would be capable of inducing different patterns of immune response to maximize the immune response against T. cruzi (virulent Romildo strain) infection. Two hundred eighty nonisogenic mice were divided into 14 groups according to the immunization scheme and the subsequent challenge with virulent Romildo T. cruzi strain. Nonimmunized groups and animals inoculated without adjuvants were also included. Immune protection was not observed with Th2 adjuvants (incomplete Freund's adjuvant [IFA] and Alum) due to high parasitemia. Th1/Th2-polarizing adjuvants also did not induce immune protection because inulin was unable to maintain survival, and immune-stimulating complexes induced intense inflammatory processes. Animals sensitized with RM1 strain without adjuvants were able to reduce parasitemia, increase survival, and protect against severe histological lesions, followed by adequate cytokine stimulation. Finally, our results demonstrate that the early and balanced IFN-γ production becomes critical to promote protection and that Th1 adjuvant elicited a controversial infection control due to increased histopathological damage. Therefore, the host's immunomodulation remains one of the most important challenges in the research for effective protection against T. cruzi infection. Similarly, the identification of protective antigens in the RM1 strain of T. cruzi marinkellei may contribute to further studies on vaccine development against human Chagas disease.
Collapse
Affiliation(s)
- Gabriel Antonio Nogueira Nascentes
- 1 Microbiology and Immunology Discipline, Federal Institute of Education , Science and Technology at Triângulo Mineiro (IFTM), Uberaba, Brazil
| | - César Gómez Hernández
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Rosiley Aparecida de Souza Rabelo
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Raquel Fernandes Coelho
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Fabiana Rossetto de Morais
- 3 School of Pharmaceutical Sciences at Ribeirão Preto, University of São Paulo (USP) , Ribeirão Preto, Brazil
| | - Tatiane Marques
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Lara Rocha Batista
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Wendell Sérgio Ferreira Meira
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Carlo José Freire de Oliveira
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Eliane Lages Silva
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Luis Eduardo Ramírez
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| |
Collapse
|
21
|
Involvement of sulfates from cruzipain, a major antigen of Trypanosoma cruzi, in the interaction with immunomodulatory molecule Siglec-E. Med Microbiol Immunol 2015; 205:21-35. [PMID: 26047932 DOI: 10.1007/s00430-015-0421-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/15/2015] [Indexed: 10/23/2022]
Abstract
In order to investigate the involvement of sulfated groups in the Trypanosoma cruzi host-parasite relationship, we studied the interaction between the major cysteine proteinase of T. cruzi, cruzipain (Cz), a sulfate-containing sialylated molecule and the sialic acid-binding immunoglobulin like lectin-E (Siglec-E). To this aim, ELISA, indirect immunofluorescence assays and flow cytometry, using mouse Siglec-E-Fc fusion molecules and glycoproteins of parasites, were performed. Competition assays verified that the lectins, Maackia amurensis II (Mal II) and Siglec-E-Fc, compete for the same binding sites. Taking into account that Mal II binding remains unaltered by sulfation, we established this lectin as sialylation degree control. Proteins of an enriched microsomal fraction showed the highest binding to Siglec-E as compared with those from the other parasite subcellular fractions. ELISA assays and the affinity purification of Cz by a Siglec-E column confirmed the interaction between both molecules. The significant decrease in binding of Siglec-E-Fc to Cz and to its C-terminal domain (C-T) after desulfation of these molecules suggests that sulfates contribute to the interaction between Siglec-E-Fc and these glycoproteins. Competitive ELISA assays confirmed the involvement of sulfated epitopes in the affinity between Siglec-E and Cz, probably modified by natural protein environment. Interestingly, data from flow cytometry of untreated and chlorate-treated parasites suggested that sulfates are not primary receptors, but enhance the binding of Siglec-E to trypomastigotic forms. Altogether, our findings support the notion that sulfate-containing sialylated glycoproteins interact with Siglec-E, an ortholog protein of human Siglec-9, and might modulate the immune response of the host, favoring parasitemia and persistence of the parasite.
Collapse
|
22
|
Experimental Vaccines against Chagas Disease: A Journey through History. J Immunol Res 2015; 2015:489758. [PMID: 26090490 PMCID: PMC4452192 DOI: 10.1155/2015/489758] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 12/13/2022] Open
Abstract
Chagas disease, or American trypanosomiasis, which is caused by the protozoan parasite Trypanosoma cruzi, is primarily a vector disease endemic in 21 Latin American countries, including Mexico. Although many vector control programs have been implemented, T. cruzi has not been eradicated. The development of an anti-T. cruzi vaccine for prophylactic and therapeutic purposes may significantly contribute to the transmission control of Chagas disease. Immune protection against experimental infection with T. cruzi has been studied since the second decade of the last century, and many types of immunogens have been used subsequently, such as killed or attenuated parasites and new DNA vaccines. This primary prevention strategy appears feasible, effective, safe, and inexpensive, although problems remain. The objective of this review is to summarize the research efforts about the development of vaccines against Chagas disease worldwide. A thorough literature review was conducted by searching PubMed with the terms “Chagas disease” and “American trypanosomiasis” together with “vaccines” or “immunization”. In addition, reports and journals not cited in PubMed were identified. Publications in English, Spanish, and Portuguese were reviewed.
Collapse
|
23
|
Pereira IR, Vilar-Pereira G, Marques V, da Silva AA, Caetano B, Moreira OC, Machado AV, Bruna-Romero O, Rodrigues MM, Gazzinelli RT, Lannes-Vieira J. A human type 5 adenovirus-based Trypanosoma cruzi therapeutic vaccine re-programs immune response and reverses chronic cardiomyopathy. PLoS Pathog 2015; 11:e1004594. [PMID: 25617628 PMCID: PMC4305326 DOI: 10.1371/journal.ppat.1004594] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/02/2014] [Indexed: 12/21/2022] Open
Abstract
Chagas disease (CD), caused by the protozoan Trypanosoma cruzi, is a prototypical neglected tropical disease. Specific immunity promotes acute phase survival. Nevertheless, one-third of CD patients develop chronic chagasic cardiomyopathy (CCC) associated with parasite persistence and immunological unbalance. Currently, the therapeutic management of patients only mitigates CCC symptoms. Therefore, a vaccine arises as an alternative to stimulate protective immunity and thereby prevent, delay progression and even reverse CCC. We examined this hypothesis by vaccinating mice with replication-defective human Type 5 recombinant adenoviruses (rAd) carrying sequences of amastigote surface protein-2 (rAdASP2) and trans-sialidase (rAdTS) T. cruzi antigens. For prophylactic vaccination, naïve C57BL/6 mice were immunized with rAdASP2+rAdTS (rAdVax) using a homologous prime/boost protocol before challenge with the Colombian strain. For therapeutic vaccination, rAdVax administration was initiated at 120 days post-infection (dpi), when mice were afflicted by CCC. Mice were analyzed for electrical abnormalities, immune response and cardiac parasitism and tissue damage. Prophylactic immunization with rAdVax induced antibodies and H-2Kb-restricted cytotoxic and interferon (IFN)γ-producing CD8+ T-cells, reduced acute heart parasitism and electrical abnormalities in the chronic phase. Therapeutic vaccination increased survival and reduced electrical abnormalities after the prime (analysis at 160 dpi) and the boost (analysis at 180 and 230 dpi). Post-therapy mice exhibited less heart injury and electrical abnormalities compared with pre-therapy mice. rAdVax therapeutic vaccination preserved specific IFNγ-mediated immunity but reduced the response to polyclonal stimuli (anti-CD3 plus anti-CD28), CD107a+ CD8+ T-cell frequency and plasma nitric oxide (NO) levels. Moreover, therapeutic rAdVax reshaped immunity in the heart tissue as reduced the number of perforin+ cells, preserved the number of IFNγ+ cells, increased the expression of IFNγ mRNA but reduced inducible NO synthase mRNA. Vaccine-based immunostimulation with rAd might offer a rational alternative for re-programming the immune response to preserve and, moreover, recover tissue injury in Chagas’ heart disease. The idea that Chagas disease (CD) has an important autoimmune involvement contributed to delay the development of therapies and vaccines. CD is a parasitic neglected disease which afflicts millions of people mostly in Latin America. The cardiac form is the main clinical manifestation of CD. Currently, patients with access to therapy receive medicaments that only mitigate symptoms. Because of the limited prospect of treatment, vaccine reemerged as a strategy to prevent infection, interfere with CD progression and, moreover, reverse heart abnormalities. Here we tested a recombinant adenovirus carrying sequences of ASP2 and TS T. cruzi antigens (rAdVax) as prophylactic and therapeutic tool using a model of chronic Chagas’ heart disease. We showed that prophylactic vaccination reduced heart parasite load, inflammation and electrical abnormalities. The rAdVax therapeutic vaccination also reduced heart injury and improved electrical function, preserved specific IFNγ-mediated immunity but reduced response to polyclonal stimuli, CD107a+ CD8+ T-cell frequency and plasma nitric oxide levels. Moreover, therapeutic rAdVax preserved the number IFNγ+ cells, but decreased perforin+ cells in the heart tissue. Therefore, our results support the hypothesis that vaccination can modify the immunological unbalance that concurs to Chagas’ heart disease to improve prognosis.
Collapse
Affiliation(s)
- Isabela Resende Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Glaucia Vilar-Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Virgínia Marques
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrea Alice da Silva
- Departamento de Patologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Bráulia Caetano
- Centro de Pesquisas René Rachou, Fiocruz, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Otacilio Cruz Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre Vieira Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Oscar Bruna-Romero
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Maurício Martins Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, São Paulo, Brazil
| | - Ricardo Tostes Gazzinelli
- Centro de Pesquisas René Rachou, Fiocruz, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
24
|
Sánchez-Valdéz FJ, Pérez Brandán C, Ferreira A, Basombrío MÁ. Gene-deleted live-attenuated Trypanosoma cruzi parasites as vaccines to protect against Chagas disease. Expert Rev Vaccines 2014; 14:681-97. [PMID: 25496192 DOI: 10.1586/14760584.2015.989989] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan parasite Trypanosoma cruzi. This illness is now becoming global, mainly due to congenital transmission, and so far, there are no prophylactic or therapeutic vaccines available to either prevent or treat Chagas disease. Therefore, different approaches aimed at identifying new protective immunogens are urgently needed. Live vaccines are likely to be more efficient in inducing protection, but safety issues linked with their use have been raised. The development of improved protozoan genetic manipulation tools and genomic and biological information has helped to increase the safety of live vaccines. These advances have generated a renewed interest in the use of genetically attenuated parasites as vaccines against Chagas disease. This review discusses the protective capacity of genetically attenuated parasite vaccines and the challenges and perspectives for the development of an effective whole-parasite Chagas disease vaccine.
Collapse
|
25
|
Effects of chlorate on the sulfation process of Trypanosoma cruzi glycoconjugates. Implication of parasite sulfates in cellular invasion. Acta Trop 2014; 137:161-73. [PMID: 24879929 DOI: 10.1016/j.actatropica.2014.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 05/17/2014] [Accepted: 05/20/2014] [Indexed: 02/06/2023]
Abstract
Sulfation, a post-translational modification which plays a key role in various biological processes, is inhibited by competition with chlorate. In Trypanosoma cruzi, the agent of Chagas' disease, sulfated structures have been described as part of glycolipids and we have reported sulfated high-mannose type oligosaccharides in the C-T domain of the cruzipain (Cz) glycoprotein. However, sulfation pathways have not been described yet in this parasite. Herein, we studied the effect of chlorate treatment on T. cruzi with the aim to gain some knowledge about sulfation metabolism and the role of sulfated molecules in this parasite. In chlorate-treated epimastigotes, immunoblotting with anti-sulfates enriched Cz IgGs (AS-enriched IgGs) showed Cz undersulfation. Accordingly, a Cz mobility shift toward higher isoelectric points was observed in 2D-PAGE probed with anti-Cz antibodies. Ultrastructural membrane abnormalities and a significant decrease of dark lipid reservosomes were shown by electron microscopy and a significant decrease in sulfatide levels was confirmed by TLC/UV-MALDI-TOF-MS analysis. Altogether, these results suggest T. cruzi sulfation occurs via PAPS. Sulfated epitopes in trypomastigote and amastigote forms were evidenced using AS-enriched IgGs by immunoblotting. Their presence on trypomastigotes surface was demonstrated by flow cytometry and IF with Cz/dCz specific antibodies. Interestingly, the percentage of infected cardiac HL-1 cells decreased 40% when using chlorate-treated trypomastigotes, suggesting sulfates are involved in the invasion process. The same effect was observed when cells were pre-incubated with dCz, dC-T or an anti-high mannose receptor (HMR) antibody, suggesting Cz sulfates and HMR are also involved in the infection process by T. cruzi.
Collapse
|
26
|
Cazorla SI, Matos MN, Cerny N, Ramirez C, Alberti AS, Bivona AE, Morales C, Guzmán CA, Malchiodi EL. Oral multicomponent DNA vaccine delivered by attenuated Salmonella elicited immunoprotection against American trypanosomiasis. J Infect Dis 2014; 211:698-707. [PMID: 25160983 DOI: 10.1093/infdis/jiu480] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We have reported that attenuated Salmonella (S) carrying plasmids encoding the cysteine protease cruzipain (Cz) protects against Trypanosoma cruzi infection. Here, we determined whether immunoprotection could be improved by the oral coadministration of 3 Salmonella carrying the plasmids that encode the antigens Cz, Tc52, and Tc24. SCz+STc52+STc24-immunized mice presented an increased antibody response against each antigen compared with those in the single antigen-immunized groups, as well as higher trypomastigotes antibody-mediated lyses and cell invasion inhibition compared with controls. SCz+STc52+STc24-immunized and -challenged mice rendered lower parasitemia. Weight loss after infection was detected in all mice except those in the SCz+STc52+STc24 group. Moreover, cardiomyopathy-associated enzyme activity was significantly lower in SCz+STc24+STc52-immunized mice compared with controls. Few or no abnormalities were found in muscle tissues of SCz+STc24+STc52-immunized mice, whereas controls presented with inflammatory foci, necrosis, and amastigote nests. We conclude that a multicomponent approach that targets several invasion and metabolic mechanisms improves protection compared with single-component vaccines.
Collapse
Affiliation(s)
- Silvia I Cazorla
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Dr. R. A. Margni, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires (CONICET-UBA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA
| | - Marina N Matos
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Dr. R. A. Margni, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires (CONICET-UBA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA
| | - Natacha Cerny
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Dr. R. A. Margni, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires (CONICET-UBA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA
| | - Carolina Ramirez
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Dr. R. A. Margni, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires (CONICET-UBA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA
| | - Andrés Sanchez Alberti
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Dr. R. A. Margni, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires (CONICET-UBA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA
| | - Augusto E Bivona
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Dr. R. A. Margni, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires (CONICET-UBA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA
| | - Celina Morales
- Departamento de Patología, Facultad de Medicina UBA, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Emilio L Malchiodi
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Dr. R. A. Margni, Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad de Buenos Aires (CONICET-UBA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA
| |
Collapse
|
27
|
Tc52 amino-terminal-domain DNA carried by attenuated Salmonella enterica serovar Typhimurium induces protection against a Trypanosoma cruzi lethal challenge. Infect Immun 2014; 82:4265-75. [PMID: 25069980 DOI: 10.1128/iai.02190-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In this work we immunized mice with DNA encoding full-length Tc52 or its amino- or carboxy-terminal (N- and C-term, respectively) domain carried by attenuated Salmonella as a DNA delivery system. As expected, Salmonella-mediated DNA delivery resulted in low antibody titers and a predominantly Th1 response, as shown by the ratio of IgG2a/IgG1-specific antibodies. Despite modest expression of Tc52 in trypomastigotes, the antibodies elicited by vaccination were able to mediate lysis of the trypomastigotes in the presence of complement and inhibit their invasion of mammal cells in vitro. The strongest functional activity was observed with sera from mice immunized with Salmonella carrying the N-term domain (SN-term), followed by Tc52 (STc52), and the C-term domain (SC-term). All immunized groups developed strong cellular responses, with predominant activation of Th1 cells. However, mice immunized with SN-term showed higher levels of interleukin-10 (IL-10), counterbalancing the inflammatory reaction, and also strong activation of Tc52-specific gamma interferon-positive (IFN-γ(+)) CD8(+) T cells. In agreement with this, although all prototypes conferred protection against infection, immunization with SN-term promoted greater protection than that with SC-term for all parameters tested and slightly better protection than that with STc52, especially in the acute stage of infection. We conclude that the N-terminal domain of Tc52 is the section of the protein that confers maximal protection against infection and propose it as a promising candidate for vaccine development.
Collapse
|
28
|
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: 2.6] [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
|
29
|
Genetic vaccination against experimental infection with myotropic parasite strains of Trypanosoma cruzi. Mediators Inflamm 2014; 2014:605023. [PMID: 25061263 PMCID: PMC4098640 DOI: 10.1155/2014/605023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 05/25/2014] [Indexed: 01/29/2023] Open
Abstract
In earlier studies, we reported that a heterologous prime-boost regimen using recombinant plasmid DNA followed by replication-defective adenovirus vector, both containing Trypanosoma cruzi genes encoding trans-sialidase (TS) and amastigote surface protein (ASP) 2, provided protective immunity against experimental infection with a reticulotropic strain of this human protozoan parasite. Herein, we tested the outcome of genetic vaccination of F1 (CB10XBALB/c) mice challenged with myotropic parasite strains (Brazil and Colombian). Initially, we determined that the coadministration during priming of a DNA plasmid containing the murine IL-12 gene improved the immune response and was essential for protective immunity elicited by the heterologous prime-boost regimen in susceptible male mice against acute lethal infections with these parasites. The prophylactic or therapeutic vaccination of resistant female mice led to a drastic reduction in the number of inflammatory infiltrates in cardiac and skeletal muscles during the chronic phase of infection with either strain. Analysis of the electrocardiographic parameters showed that prophylactic vaccination reduced the frequencies of sinus arrhythmia and atrioventricular block. Our results confirmed that prophylactic vaccination using the TS and ASP-2 genes benefits the host against acute and chronic pathologies caused by T. cruzi and should be further evaluated for the development of a veterinary or human vaccine against Chagas disease.
Collapse
|
30
|
Basso B, Marini V. Experimental Chagas disease. Innate immune response in Balb/c mice previously vaccinated with Trypanosoma rangeli. I. The macrophage shows immunological memory: Reality or fiction? Immunobiology 2013; 219:275-84. [PMID: 24321621 DOI: 10.1016/j.imbio.2013.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/27/2013] [Accepted: 10/31/2013] [Indexed: 12/29/2022]
Abstract
Chagas' disease, caused by Trypanosoma cruzi, is a major vector borne health problem in Latin America and an emerging or re-emerging infectious disease in several countries. Immune response to T. cruzi infection is highly complex and involves many components, both regulators and effectors. Although different parasites have been shown to activate different mechanisms of innate immunity, T. cruzi is often able to survive and replicate in its host because they are well adapted to resisting host defences. An experimental model for vaccinating mice with Trypanosoma rangeli, a parasite closely related to T. cruzi, but nonpathogenic to humans, has been designed in our laboratory, showing protection against challenge with T. cruzi infection. The aim of this work was to analyze some mechanisms of the early innate immune response in T. rangeli vaccinated mice challenged with T. cruzi. For this purpose, some interactions were studied between T. cruzi and peritoneal macrophages of mice vaccinated with T. rangeli, infected or not with T. cruzi and the levels of some molecules or soluble mediators which could modify these interactions. The results in vaccinated animals showed a strong innate immune response, where the adherent cells of the vaccinated mice revealed important phagocytic activity, and some soluble mediator (Respiratory Burst: significantly increase, p ≤ 0.03; NO: the levels of vaccinated animals were lower than those of the control group; Arginasa: significantly increase, p ≤ 0.04). The results showed an important role in the early elimination of the parasites and their close relation with the absence of histological lesions that these animals present with regard to the only infected mice. This behaviour reveals that the macrophages act with some type of memory, recognizing the antigens to which they have previously been exposed, in mice were vaccinated with T. rangeli, which shares epitopes with T. cruzi.
Collapse
Affiliation(s)
- B Basso
- Department of Paediatrics, Neonatology Service, Medicine School, National Cordoba University, Argentina; National Co-ordination of Vector Control, Argentina.
| | - V Marini
- Department of Immunology Medicine School, Catholic University of Cordoba, Argentina
| |
Collapse
|
31
|
Abstract
The trypanosomes cause two neglected tropical diseases, Chagas disease in the Americas and African trypanosomiasis in sub-Saharan Africa. Over recent years a raft of molecular tools have been developed enabling the genetic dissection of many aspects of trypanosome biology, including the mechanisms underlying resistance to some of the current clinical and veterinary drugs. This has led to the identification and characterization of key resistance determinants, including transporters for the anti-Trypanosoma brucei drugs, melarsoprol, pentamidine and eflornithine, and the activator of nifurtimox-benznidazole, the anti-Trypanosoma cruzi drugs. More recently, advances in sequencing technology, combined with the development of RNA interference libraries in the clinically relevant bloodstream form of T. brucei have led to an exponential increase in the number of proteins known to interact either directly or indirectly with the anti-trypanosomal drugs. In this review, we discuss these findings and the technological developments that are set to further revolutionise our understanding of drug-trypanosome interactions. The new knowledge gained should inform the development of novel interventions against the devastating diseases caused by these parasites.
Collapse
|
32
|
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: 19] [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
|
33
|
Kling JC, Körner H. Different regulatory mechanisms in protozoan parasitic infections. Int J Parasitol 2013; 43:417-25. [PMID: 23470812 DOI: 10.1016/j.ijpara.2013.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/01/2013] [Accepted: 02/04/2013] [Indexed: 02/07/2023]
Abstract
The immune response to the protozoan pathogens, Leishmania spp., Trypanosoma spp. and Plasmodium spp., has been studied extensively with particular focus on regulation of the immune response by immunological mechanisms. More specifically, in diseases caused by parasites, immunosuppression frequently prevents immunopathology that can injure the host. However, this allows a small number of parasites to evade the immune response and remain in the host after a clinical cure. The consequences can be chronic infections, which establish a zoonotic or anthroponotic reservoir. This review will highlight some of the identified regulatory mechanisms of the immune system that govern immune responses to parasitic diseases, in particular leishmaniasis, trypanosomiasis and malaria, and discuss implications for the development of efficient vaccines against these diseases.
Collapse
Affiliation(s)
- Jessica C Kling
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | | |
Collapse
|
34
|
Challenge of chronically infected mice with homologous trypanosoma cruzi parasites enhances the immune response but does not modify cardiopathy: implications for the design of a therapeutic vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 20:248-54. [PMID: 23254299 DOI: 10.1128/cvi.00032-12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Chagas disease is a Trypanosoma cruzi-induced zoonosis that has no natural cure. Local damage induced by the parasite and the immune response causes chronic heart and digestive lesions. Efforts to develop a therapeutic vaccine that boosts the immune response to completely clear the parasite are needed because there is no effective treatment for chronically infected patients. In an attempt to modify the host-parasite equilibrium to increase parasite destruction, we analyzed cardiopathy and the immune response in chronically infected mice that were challenged with live homologous parasites. Challenge with a single dose of parasite increased CD4(+) and CD8(+) T cell populations, gamma interferon (IFN-γ) production, and serum-specific IgG levels. However, subpatent parasitemias and cardiac tissue were not affected. Because of the short duration of the immune boost after a single challenge, we next evaluated the impact of four parasite doses, administered 3 weeks apart. At 1 to 2 months after the last dose, the numbers of CD4(+) T cells and IFN-γ-producing CD4(+) memory cells and the CD4(+) T cell proliferative response to T. cruzi antigen were increased in the spleen. The frequency of IFN-γ-producing CD8(+) memory cells in the blood was also increased. However, the sustained challenge did not favor TH1 development; rather, it induced an increase in serum-specific IgG1 levels and mixed TH1/TH2 cytokine production. Moreover, there were no significant changes in cardiac lesions and subpatent parasitemias. In conclusion, we believe that this study may help in elucidating the necessary elements for a successful therapeutic vaccine which may reduce cardiomyopathy in chronically infected human patients.
Collapse
|
35
|
Couto AS, Soprano LL, Landoni M, Pourcelot M, Acosta DM, Bultel L, Parente J, Ferrero MR, Barbier M, Dussouy C, Esteva MI, Kovensky J, Duschak VG. An anionic synthetic sugar containing 6-SO3-NAcGlc mimics the sulfated cruzipain epitope that plays a central role in immune recognition. FEBS J 2012; 279:3665-3679. [DOI: 10.1111/j.1742-4658.2012.08728.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
36
|
Dumonteil E, Bottazzi ME, Zhan B, Heffernan MJ, Jones K, Valenzuela JG, Kamhawi S, Ortega J, de Leon Rosales SP, Lee BY, Bacon KM, Fleischer B, Slingsby BT, Cravioto MB, Tapia-Conyer R, Hotez PJ. Accelerating the development of a therapeutic vaccine for human Chagas disease: rationale and prospects. Expert Rev Vaccines 2012; 11:1043-55. [PMID: 23151163 PMCID: PMC3819810 DOI: 10.1586/erv.12.85] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chagas disease is a leading cause of heart disease affecting approximately 10 million people in Latin America and elsewhere worldwide. The two major drugs available for the treatment of Chagas disease have limited efficacy in Trypanosoma cruzi-infected adults with indeterminate (patients who have seroconverted but do not yet show signs or symptoms) and determinate (patients who have both seroconverted and have clinical disease) status; they require prolonged treatment courses and are poorly tolerated and expensive. As an alternative to chemotherapy, an injectable therapeutic Chagas disease vaccine is under development to prevent or delay Chagasic cardiomyopathy in patients with indeterminate or determinate status. The bivalent vaccine will be comprised of two recombinant T. cruzi antigens, Tc24 and TSA-1, formulated on alum together with the Toll-like receptor 4 agonist, E6020. Proof-of-concept for the efficacy of these antigens was obtained in preclinical testing at the Autonomous University of Yucatan. Here the authors discuss the potential for a therapeutic Chagas vaccine as well as the progress made towards such a vaccine, and the authors articulate a roadmap for the development of the vaccine as planned by the nonprofit Sabin Vaccine Institute Product Development Partnership and Texas Children's Hospital Center for Vaccine Development in collaboration with an international consortium of academic and industrial partners in Mexico, Germany, Japan, and the USA.
Collapse
Affiliation(s)
- Eric Dumonteil
- Laboratorio de Parasitología Centro De Investigaciones Regional, “Dr. Hideo Noguchi” Autonomous University of Yucatan (UADY), Merida, Mexico
| | - Maria Elena Bottazzi
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics (Section of Pediatric Tropical Medicine) and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Bin Zhan
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics (Section of Pediatric Tropical Medicine), National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Michael J Heffernan
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics (Section of Pediatric Tropical Medicine), National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kathryn Jones
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics (Section of Pediatric Tropical Medicine) and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jaime Ortega
- Departamento de Biotecnología y Bioingeniería, Centro de Investigacion y de Estudios Avanzados - Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | | | - Bruce Y Lee
- Public Health Computational and Operations Research (PHICOR), University of Pittsburgh, Pittsburgh PA, USA
| | - Kristina M Bacon
- Public Health Computational and Operations Research (PHICOR), University of Pittsburgh, Pittsburgh PA, USA
| | | | | | | | | | - Peter J Hotez
- Sabin Vaccine Institute and Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics (Section of Pediatric Tropical Medicine) and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
37
|
Lee BY, Bacon KM, Wateska AR, Bottazzi ME, Dumonteil E, Hotez PJ. Modeling the economic value of a Chagas' disease therapeutic vaccine. Hum Vaccin Immunother 2012; 8:1293-301. [PMID: 22894964 DOI: 10.4161/hv.20966] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The health burden of Chagas' disease (resulting from Trypanosoma cruzi infection) in Latin America (estimated to outweigh that of malaria by 5-fold and affect 2-6 million people in Mexico alone) has motivated development of therapeutic vaccines to prevent infection progression to severe disease. Our economic model for a Chagas' therapeutic vaccine in Mexico suggests that a vaccine would be highly cost-effective and in many cases economically dominant (providing both cost savings and health benefits) throughout a range of protection durations, severe adverse event risk, and dosing regimens and would be most likely to provide a positive return on investment if the vaccine prevented (rather than delayed) the onset of cardiomyopathy.
Collapse
Affiliation(s)
- Bruce Y Lee
- Public Health Computational and Operations Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Trypanosoma cruzi paraflagellar rod proteins 2 and 3 contain immunodominant CD8(+) T-cell epitopes that are recognized by cytotoxic T cells from Chagas disease patients. Mol Immunol 2012; 52:289-98. [PMID: 22750229 DOI: 10.1016/j.molimm.2012.05.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 05/24/2012] [Accepted: 05/31/2012] [Indexed: 12/21/2022]
Abstract
The protozoan parasite Trypanosoma cruzi is the etiological agent of Chagas disease. To date, no vaccine is available for protection against T. cruzi infection. The CD8(+) T cells immune response against specific antigens has shown to efficiently control the spread of the parasite in murine experimental infection. However, data concerning CD8(+) response in Chagas patients are still restricted to a few epitopes. We have studied the existence of immunodominant CD8(+) T cell epitopes in the paraflagellar rod proteins 2 and 3 (PFR2 and PFR3) from T. cruzi in a mouse model, and analyzed their recognition by cytotoxic T lymphocytes from Chagas disease patients. Immunization of C57BL/6-A2/K(b) transgenic mice with plasmids coding for the fusion proteins PFR2-HSP70 and PFR3-HSP70 induced a specific CTL response against two PFRs epitopes (PFR2(449-457) and PFR3(481-489)), and showed specific lysis percentages of 24 and 12, respectively. Moreover, the PFR2(19-28), PFR2(156-163), PFR2(449-457), PFR3(428-436), PFR3(475-482) and PFR3(481-489) peptides were observed to have a high binding affinity to the HLA-A*02:01 molecule. Remarkably, these HLA-A*02:01-binding peptides are successfully processed and presented during natural infection by T. cruzi in the context of MHC class I as evidenced by using peptide-pulsed K562-A2 cells as antigen presenting cells. The T cells from Chagas disease chronic patients specific for PFR2/PFR3 selected CD8(+) epitopes showed a pro-inflammatory cytokine secretion profile (IFN-γ, TNF-α and IL-6). A positive Granzime B secretion was observed in three out of 16 patients in response to PFR2(156-163) and PFR2(449-457) peptides, two out of 11 patients in response to PFR2(19-28) peptide and one out of 14 and 11 patients in response to PFR3(428-436) and PFR3(481-489) peptides, respectively. The PFRs-specific cytotoxic activity in purified PBMC was only detected in patients in the indeterminate phase of the disease.
Collapse
|
39
|
Repertoire, genealogy and genomic organization of cruzipain and homologous genes in Trypanosoma cruzi, T. cruzi-like and other trypanosome species. PLoS One 2012; 7:e38385. [PMID: 22685565 PMCID: PMC3369871 DOI: 10.1371/journal.pone.0038385] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 05/04/2012] [Indexed: 12/29/2022] Open
Abstract
Trypanosoma cruzi, the agent of Chagas disease, is a complex of genetically diverse isolates highly phylogenetically related to T. cruzi-like species, Trypanosoma cruzi marinkellei and Trypanosoma dionisii, all sharing morphology of blood and culture forms and development within cells. However, they differ in hosts, vectors and pathogenicity: T. cruzi is a human pathogen infective to virtually all mammals whilst the other two species are non-pathogenic and bat restricted. Previous studies suggest that variations in expression levels and genetic diversity of cruzipain, the major isoform of cathepsin L-like (CATL) enzymes of T. cruzi, correlate with levels of cellular invasion, differentiation, virulence and pathogenicity of distinct strains. In this study, we compared 80 sequences of genes encoding cruzipain from 25 T. cruzi isolates representative of all discrete typing units (DTUs TcI-TcVI) and the new genotype Tcbat and 10 sequences of homologous genes from other species. The catalytic domain repertoires diverged according to DTUs and trypanosome species. Relatively homogeneous sequences are found within and among isolates of the same DTU except TcV and TcVI, which displayed sequences unique or identical to those of TcII and TcIII, supporting their origin from the hybridization between these two DTUs. In network genealogies, sequences from T. cruzi clustered tightly together and closer to T. c. marinkellei than to T. dionisii and largely differed from homologues of T. rangeli and T. b. brucei. Here, analysis of isolates representative of the overall biological and genetic diversity of T. cruzi and closest T. cruzi-like species evidenced DTU- and species-specific polymorphisms corroborating phylogenetic relationships inferred with other genes. Comparison of both phylogenetically close and distant trypanosomes is valuable to understand host-parasite interactions, virulence and pathogenicity. Our findings corroborate cruzipain as valuable target for drugs, vaccine, diagnostic and genotyping approaches.
Collapse
|
40
|
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: 5.3] [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
|
41
|
Nakayasu ES, Sobreira TJP, Torres R, Ganiko L, Oliveira PSL, Marques AF, Almeida IC. Improved proteomic approach for the discovery of potential vaccine targets in Trypanosoma cruzi. J Proteome Res 2012; 11:237-46. [PMID: 22115061 PMCID: PMC3253764 DOI: 10.1021/pr200806s] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chagas disease, caused by Trypanosoma cruzi, is a devastating parasitic infection affecting millions of people. Although many efforts have been made for the development of immunotherapies, there is no available vaccine against this deadly infection. One major hurdle for the rational approach to develop a T. cruzi vaccine is the limited information about the proteins produced by different phylogenetic lineages, strains, and stages of the parasite. Here, we have adapted a 1D nanoHPLC system to perform online 2D LC-MS/MS, using the autosampler to inject the eluting salt solutions in the first dimension separation. The application of this methodology for the proteomic analysis of the infective trypomastigote stage of T. cruzi led to the identification of 1448 nonredundant proteins. Furthermore, about 14% of the identified sequences comprise surface proteins, most of them glycosylphosphatidylinositol (GPI)-anchored and related to parasite pathogenesis. Immunoinformatic analysis revealed thousands of potential peptides with predicted high-binding affinity for major histocompatibility complex (MHC) class I and II molecules. The high diversity of proteins expressed on the trypomastigote surface may have many implications for host-cell invasion and immunoevasion mechanisms triggered by the parasite. Finally, we performed a rational approach to filter potential T-cell epitopes that could be further tested and validated for development of a Chagas disease vaccine.
Collapse
Affiliation(s)
- Ernesto S. Nakayasu
- Department of Biological Sciences, University of Texas at El Paso, 500 W. University Ave., El Paso TX, 79902, USA
| | - Tiago J. P. Sobreira
- National Laboratory for Biosciences (LNBio), National Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
| | - Rafael Torres
- Department of Biological Sciences, University of Texas at El Paso, 500 W. University Ave., El Paso TX, 79902, USA
| | - Luciane Ganiko
- Department of Biological Sciences, University of Texas at El Paso, 500 W. University Ave., El Paso TX, 79902, USA
| | - Paulo S. L. Oliveira
- National Laboratory for Biosciences (LNBio), National Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
| | - Alexandre F. Marques
- Department of Biological Sciences, University of Texas at El Paso, 500 W. University Ave., El Paso TX, 79902, USA
| | - Igor C. Almeida
- Department of Biological Sciences, University of Texas at El Paso, 500 W. University Ave., El Paso TX, 79902, USA
| |
Collapse
|
42
|
Enlarging the "Audacious Goal": elimination of the world's high prevalence neglected tropical diseases. Vaccine 2011; 29 Suppl 4:D104-10. [PMID: 22188933 DOI: 10.1016/j.vaccine.2011.06.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 06/01/2011] [Accepted: 06/09/2011] [Indexed: 12/22/2022]
Abstract
The high prevalence neglected tropical diseases (NTDs) exhibit a global disease burden that exceeds malaria, tuberculosis, and other better known global health conditions; they also represent a potent force in trapping the world's poorest people in poverty. Through extremely low cost national programs of disease mapping and mass drug administration (MDA) for the seven most common NTDs, integrated NTD control and elimination efforts are now in place in more than 14 countries through the support of the United States Agency for International Development (USAID), the British Department for International Development (DFID), and the Global Network for NTDs and its partners. The World Health Organization (WHO) estimates that in 2008 some 670 million people in 75 countries received NTD treatments through these and other sponsored programs. With continued successes the next decade could witness the global elimination of blinding trachoma, human Africa trypanosomiasis (HAT), lymphatic filariasis (LF), onchocerciasis, trachoma, and leprosy as public health problems, in addition to the eradication of dracunculiasis. For other high prevalence NTDs, including hookworm infection, schistosomiasis, Chagas disease and leishmaniasis, new drugs and vaccines may still be required. Increasingly it is recognized that the high prevalence NTDs exhibit extensive geographic overlap and polyparasitism is commonly found throughout the world's low income countries. Therefore, global elimination will also require integrated packages of drugs together with vaccine-linked chemotherapy. Ultimately, the global elimination of the high prevalence NTDs will require continued large-scale support from the U.S. Government and selected European governments, however, the emerging market economies, such as Brazil, China, India, Mexico, and Nigeria, and wealthy countries in the Middle East will also have to substantially contribute.
Collapse
|
43
|
Pellegrini A, Guiñazu N, Giordanengo L, Cano RC, Gea S. The role of Toll-like receptors and adaptive immunity in the development of protective or pathological immune response triggered by the Trypanosoma cruzi protozoan. Future Microbiol 2011; 6:1521-33. [DOI: 10.2217/fmb.11.122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Trypanosoma cruzi, the causal agent of Chagas disease, is an intracellular protozoan parasite that predominantly invades macrophages and cardiomyocytes, leading to persistent infection. Several members of the Toll-like receptor family are crucial for innate immunity to infection and are involved in maintaining tissue homeostasis. This review focuses on recent experimental findings of the innate and adaptive immune response in controlling the parasite and/or in generating heart and liver tissue injury. We also describe the importance of the host’s genetic background in the outcome of the disease and emphasize the importance of studying the response to specific parasite antigens. Understanding the dual participation of the immune response may contribute to the design of new therapies for Chagas disease.
Collapse
Affiliation(s)
- Andrea Pellegrini
- Inmunología, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, CIBICI-CONICET, Universidad Nacional de Córdoba, Ciudad Universitaria, Haya de la Torre y Medina Allende S/N, Córdoba, 5000, Argentina
| | - Natalia Guiñazu
- Inmunología, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, CIBICI-CONICET, Universidad Nacional de Córdoba, Ciudad Universitaria, Haya de la Torre y Medina Allende S/N, Córdoba, 5000, Argentina
| | - Laura Giordanengo
- Inmunología, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, CIBICI-CONICET, Universidad Nacional de Córdoba, Ciudad Universitaria, Haya de la Torre y Medina Allende S/N, Córdoba, 5000, Argentina
| | - Roxana Carolina Cano
- Inmunología, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, CIBICI-CONICET, Universidad Nacional de Córdoba, Ciudad Universitaria, Haya de la Torre y Medina Allende S/N, Córdoba, 5000, Argentina
| | | |
Collapse
|
44
|
Teixeira SM, El-Sayed NM, Araújo PR. The genome and its implications. ADVANCES IN PARASITOLOGY 2011; 75:209-30. [PMID: 21820558 DOI: 10.1016/b978-0-12-385863-4.00010-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Trypanosoma cruzi has a heterogeneous population composed of a pool of strains that circulate in the domestic and sylvatic cycles. Genome sequencing of the clone CL Brener revealed a highly repetitive genome of about 110Mb containing an estimated 22,570 genes. Because of its hybrid nature, sequences representing the two haplotypes have been generated. In addition, a repeat content close to 50% made the assembly of the estimated 41 pairs of chromosomes quite challenging. Similar to other trypanosomatids, the organization of T. cruzi chromosomes was found to be very peculiar, with protein-coding genes organized in long polycistronic transcription units encoding 20 or more proteins in one strand separated by strand switch regions. Another remarkable feature of the T. cruzi genome is the massive expansion of surface protein gene families. Because of the high genetic diversity of the T. cruzi population, sequencing of additional strains and comparative genomic and transcriptome analyses are in progress. Five years after its publication, the genome data have proven to be an essential tool for the study of T. cruzi and increasing efforts to translate this knowledge into the development of new modes of intervention to control Chagas disease are underway.
Collapse
Affiliation(s)
- Santuza M Teixeira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | |
Collapse
|
45
|
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: 66] [Impact Index Per Article: 5.1] [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
|
46
|
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: 1.4] [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
|
47
|
Muscia GC, Cazorla SI, Frank FM, Borosky GL, Buldain GY, Asís SE, Malchiodi EL. Synthesis, trypanocidal activity and molecular modeling studies of 2-alkylaminomethylquinoline derivatives. Eur J Med Chem 2011; 46:3696-703. [PMID: 21664012 DOI: 10.1016/j.ejmech.2011.05.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 04/19/2011] [Accepted: 05/13/2011] [Indexed: 01/15/2023]
Abstract
Research and development of new drugs effective in the treatment of Trypanosoma cruzi infections are a real need for the 16 million people infected in the Americas. In a previous work, a quinoline derivative substituted by a 2-piperidylmethyl moiety showed to be active against Chagas disease and was considered a lead compound for further optimization. A series of ten analogous derivatives were tested against epimastigotes as a first approach. In view of their promising results, six of them were evaluated against the blood and intracellular replicative forms of the parasite in humans. Among them, compound 12 which possesses a 6-acetamidohexylamino substituent showed remarkable improvement in activity against epimastigotes, trypomastigotes and amastigotes compared with the structure lead, as well as a good selectivity index for the two parasite stages present in humans. In addition, treatment of infected mice with compound 12 induced a significant reduction in parasitemia compared with non-treated mice. Molecular modeling studies were performed by computational methods in order to elucidate the factors determining these experimental bioactivities.
Collapse
Affiliation(s)
- Gisela C Muscia
- Departamento de Química Orgánica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113 Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
48
|
Marini V, Moretti E, Bermejo D, Basso B. Vaccination with Trypanosoma rangeli modulates the profiles of immunoglobulins and IL-6 at local and systemic levels in the early phase of Trypanosoma cruzi experimental infection. Mem Inst Oswaldo Cruz 2011; 106:32-7. [PMID: 21340352 DOI: 10.1590/s0074-02762011000100005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 12/08/2010] [Indexed: 12/17/2022] Open
Abstract
In America, there are two species of Trypanosoma that can infect humans: Trypanosoma cruzi, which is responsible for Chagas disease and Trypanosoma rangeli, which is not pathogenic. We have developed a model of vaccination in mice with T. rangeli epimastigotes that protects against T. cruzi infection. The goal of this work was to study the pattern of specific immunoglobulins in the peritoneum (the site of infection) and in the sera of mice immunized with T. rangeli before and after challenge with T. cruzi. Additionally, we studied the effects triggered by antigen-antibodies binding and the levels of key cytokines involved in the humoral response, such as IL-4, IL-5 and IL-6. The immunization triggered the production of antibodies reactive with T. cruzi in peritoneal fluid (PF) and in serum, mainly IgG1 and, to a lesser magnitude, IgG2. Only immunized mice developed specific IgG3 antibodies in their peritoneal cavities. Antibodies were able to bind to the surface of the parasites and agglutinate them. Among the cytokines studied, IL-6 was elevated in PF during early infection, with higher levels in non-immunized-infected mice. The results indicate that T. rangeli vaccination against T. cruzi infection triggers a high production of specific IgG isotypes in PF and sera before infection and modulates the levels of IL-6 in PF in the early periods of infection.
Collapse
Affiliation(s)
- Vanina Marini
- Laboratorio de la Coordinación Nacional de Control de Vectores, Córdoba, Argentina
| | | | | | | |
Collapse
|
49
|
Psilostachyin C: a natural compound with trypanocidal activity. Int J Antimicrob Agents 2011; 37:536-43. [DOI: 10.1016/j.ijantimicag.2011.02.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 01/27/2011] [Accepted: 02/02/2011] [Indexed: 12/27/2022]
|
50
|
Malafaia G, Rodrigues ASDL. [Centenary of the discovery of Chagas disease: challenges and prospects]. Rev Soc Bras Med Trop 2011; 43:483-5. [PMID: 21085853 DOI: 10.1590/s0037-86822010000500001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|