1
|
Aranda-Chan V, Cárdenas-Guerra RE, Otero-Pedraza A, Pacindo-Cabrales EE, Flores-Pucheta CI, Montes-Flores O, Arroyo R, Ortega-López J. Insights into Peptidyl-Prolyl cis- trans Isomerases from Clinically Important Protozoans: From Structure to Potential Biotechnological Applications. Pathogens 2024; 13:644. [PMID: 39204244 PMCID: PMC11357558 DOI: 10.3390/pathogens13080644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
Peptidyl-prolyl cis/trans isomerases (PPIases) are present in a wide variety of microorganisms, including protozoan parasites such as Trypanosoma cruzi, Trypanosoma brucei, Trichomonas vaginalis, Leishmania major, Leishmania donovani, Plasmodium falciparum, Plasmodium vivax, Entamoeba histolytica, Giardia intestinalis, Cryptosporidium parvum, and Cryptosporidium hominis, all of which cause important neglected diseases. PPIases are classified as cyclophilins, FKBPs, or parvulins and play crucial roles in catalyzing the cis-trans isomerization of the peptide bond preceding a proline residue. This activity assists in correct protein folding. However, experimentally, the biological structure-function characterization of PPIases from these protozoan parasites has been poorly addressed. The recombinant production of these enzymes is highly relevant for this ongoing research. Thus, this review explores the structural diversity, functions, recombinant production, activity, and inhibition of protozoan PPIases. We also highlight their potential as biotechnological tools for the in vitro refolding of other recombinant proteins from these parasites. These applications are invaluable for the development of diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Verónica Aranda-Chan
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Rosa Elena Cárdenas-Guerra
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Alejandro Otero-Pedraza
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Esdras Enoc Pacindo-Cabrales
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Claudia Ivonne Flores-Pucheta
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Octavio Montes-Flores
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Rossana Arroyo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico;
| | - Jaime Ortega-López
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| |
Collapse
|
2
|
Cárdenas-Guerra RE, Montes-Flores O, Nava-Pintor EE, Reséndiz-Cardiel G, Flores-Pucheta CI, Rodríguez-Gavaldón YI, Arroyo R, Bottazzi ME, Hotez PJ, Ortega-López J. Chagasin from Trypanosoma cruzi as a molecular scaffold to express epitopes of TSA-1 as soluble recombinant chimeras. Protein Expr Purif 2024; 218:106458. [PMID: 38423156 DOI: 10.1016/j.pep.2024.106458] [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: 09/05/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Trypanosoma cruzi is the causative agent of Chagas disease, a global public health problem. New therapeutic drugs and biologics are needed. The TSA-1 recombinant protein of T. cruzi is one such promising antigen for developing a therapeutic vaccine. However, it is overexpressed in E. coli as inclusion bodies, requiring an additional refolding step. As an alternative, in this study, we propose the endogenous cysteine protease inhibitor chagasin as a molecular scaffold to generate chimeric proteins. These proteins will contain combinations of two of the five conserved epitopes (E1 to E5) of TSA-1 in the L4 and L6 chagasin loops. Twenty chimeras (Q1-Q20) were designed, and their solubility was predicted using bioinformatics tools. Nine chimeras with different degrees of solubility were selected and expressed in E. coli BL21 (DE3). Western blot assays with anti-6x-His and anti-chagasin antibodies confirmed the expression of soluble recombinant chimeras. Both theoretically and experimentally, the Q12 (E5-E3) chimera was the most soluble, and the Q20 (E4-E5) the most insoluble protein. Q4 (E5-E1) and Q8 (E5-E2) chimeras were classified as proteins with medium solubility that exhibited the highest yield in the soluble fraction. Notably, Q4 has a yield of 239 mg/L, well above the yield of recombinant chagasin (16.5 mg/L) expressed in a soluble form. The expression of the Q4 chimera was scaled up to a 7 L fermenter obtaining a yield of 490 mg/L. These data show that chagasin can serve as a molecular scaffold for the expression of TSA-1 epitopes in the form of soluble chimeras.
Collapse
Affiliation(s)
- Rosa Elena Cárdenas-Guerra
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Octavio Montes-Flores
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Edgar Ezequiel Nava-Pintor
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Gerardo Reséndiz-Cardiel
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Claudia Ivonne Flores-Pucheta
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Yasmín Irene Rodríguez-Gavaldón
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Rossana Arroyo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department 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, Department of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jaime Ortega-López
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, CP 07360, Mexico City, Mexico.
| |
Collapse
|
3
|
Dumonteil E, Tu W, Desale H, Goff K, Marx P, Ortega-Lopez J, Herrera C. Immunoglobulin and T cell receptor repertoire changes induced by a prototype vaccine against Chagas disease in naïve rhesus macaques. J Biomed Sci 2024; 31:58. [PMID: 38824576 PMCID: PMC11143712 DOI: 10.1186/s12929-024-01050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 05/27/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND A vaccine against Trypanosoma cruzi, the agent of Chagas disease, would be an excellent additional tool for disease control. A recombinant vaccine based on Tc24 and TSA1 parasite antigens was found to be safe and immunogenic in naïve macaques. METHODS We used RNA-sequencing and performed a transcriptomic analysis of PBMC responses to vaccination of naïve macaques after each vaccine dose, to shed light on the immunogenicity of this vaccine and guide the optimization of doses and formulation. We identified differentially expressed genes and pathways and characterized immunoglobulin and T cell receptor repertoires. RESULTS RNA-sequencing analysis indicated a clear transcriptomic response of PBMCs after three vaccine doses, with the up-regulation of several immune cell activation pathways and a broad non-polarized immune profile. Analysis of the IgG repertoire showed that it had a rapid turnover with novel IgGs produced following each vaccine dose, while the TCR repertoire presented several persisting clones that were expanded after each vaccine dose. CONCLUSIONS These data suggest that three vaccine doses may be needed for optimum immunogenicity and support the further evaluation of the protective efficacy of this vaccine.
Collapse
Affiliation(s)
- Eric Dumonteil
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, 1440 Canal St, New Orleans, Louisiana, 70112, USA.
| | - Weihong Tu
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, 1440 Canal St, New Orleans, Louisiana, 70112, USA
| | - Hans Desale
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, 1440 Canal St, New Orleans, Louisiana, 70112, USA
| | - Kelly Goff
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Preston Marx
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, 1440 Canal St, New Orleans, Louisiana, 70112, USA
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Jaime Ortega-Lopez
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico, México
| | - Claudia Herrera
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, 1440 Canal St, New Orleans, Louisiana, 70112, USA
| |
Collapse
|
4
|
Ramos-Vega A, Dzul-Huchim VM, Villanueva-Lizama LE, Dumonteil E, Rosales-Mendoza S, Bañuelos-Hernández B, Angulo M, Cruz-Chan JV, Monreal-Escalante E, Angulo C. Protective efficacy of the oral vaccine Tc24:Co1 produced in Schizochytrium sp. against Trypanosoma cruzi infection in a mouse model. Microb Pathog 2024; 186:106488. [PMID: 38061668 DOI: 10.1016/j.micpath.2023.106488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 01/02/2024]
Abstract
Trypanosoma cruzi parasite - causal Chagas disease agent - affects about 7 million people; no vaccine is available, and current medications have not been entirely effective. Multidisciplinary efforts are necessary for developing clinical vaccine prototypes. Thus, this research study aims to assess the expressed and whole-cell administration protection of the oral vaccine prototype Tc24:Co1 using Schizochytrium sp. microalga. High recombinant protein expression yields (675 μg/L) of algal culture were obtained. Additionally, Schizochytrium sp.-Tc24:Co1 resulted stable at 4 °C for up to six months and at 25 °C for three months. After receiving four oral doses of the vaccine, the mice showed a significant humoral immune response and a parasitemia reduction associated with a lack of heart inflammatory damage compared with the unvaccinated controls. The Schizochytrium sp.-Tc24:Co1 vaccine demonstrates to be promising as a prototype for further development showing protective effects against a T. cruzi challenge in a mouse model.
Collapse
Affiliation(s)
- Abel Ramos-Vega
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, La Paz, B.C.S., Mexico
| | - Victor Manuel Dzul-Huchim
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Liliana Estefania Villanueva-Lizama
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Eric Dumonteil
- Deparment of Tropical Medicine and Infectious Diseases, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - 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
| | | | - Miriam Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, La Paz, B.C.S., Mexico
| | - Julio Vladimir Cruz-Chan
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Elizabeth Monreal-Escalante
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, La Paz, B.C.S., Mexico; CONAHCYT-Centro de Investigaciones Biológicas del Noroeste, La Paz, B.C.S., Mexico.
| | - Carlos Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, La Paz, B.C.S., Mexico.
| |
Collapse
|
5
|
Dumonteil E, Herrera C, Marx PA. Safety and preservation of cardiac function following therapeutic vaccination against Trypanosoma cruzi in rhesus macaques. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023; 56:400-407. [PMID: 36210315 PMCID: PMC10131272 DOI: 10.1016/j.jmii.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/10/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Chronic Chagasic cardiomyopathy is responsible for a large disease burden in the Americas, and a therapeutic vaccine would be highly desirable. We tested the safety and efficacy of a therapeutic DNA vaccine encoding antigens TSA-1 and Tc24 for preventing cardiac alterations in experimentally infected macaques. A secondary objective was to evaluate the feasibility of detecting changes in cardiac alterations in these animals. METHODS Naïve rhesus macaques were infected with Trypanosoma cruzi and treated with three doses of DNA vaccines. RESULTS Blood cell counts and chemistry indicated that therapeutic vaccination was safe, as hepatic and renal function appeared unaffected by the vaccination and/or infection with T. cruzi. Electrocardiographic (ECG) recordings indicated that no marked arrhythmias developed up to 7 months post-infection. Univariate analysis of ECG parameters found no significant differences in any of these parameters between vaccinated and control macaques. However, linear discriminant analysis revealed that control macaques presented clear alterations in their ECG patterns at 7 months post-infection, indicative of the onset of conduction defects and cardiac alterations, and these changes were prevented in vaccine treated macaques. CONCLUSIONS This is the first evidence that therapeutic vaccination against T. cruzi can prevent cardiac alterations in non-human primates, strengthening the rationale for developing a human vaccine against Chagas disease.
Collapse
Affiliation(s)
- Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA.
| | - Claudia Herrera
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Preston A Marx
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA; Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| |
Collapse
|
6
|
Haghighi M, Khorasani A, Karimi P, Keshavarz R, Mahdavi M. Different Formulations of Inactivated SARS-CoV-2 Vaccine Candidates in Human Compatible Adjuvants: Potency Studies in Mice Showed Different Platforms of Immune Responses. Viral Immunol 2022; 35:663-672. [PMID: 36534465 DOI: 10.1089/vim.2022.0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Several inactivated SARS-CoV-2 vaccines have been approved for human use, but are not highly potent. In this study, different formulations of the inactivated SARS-CoV-2 virus were developed in Alum, Montanide 51VG, and Montanide ISA720VG adjuvants, followed by assessment of immune responses. The SARS-CoV-2 virus was inactivated with formalin and formulated in the adjuvants. BALB/c mice were immunized subcutaneously with 4 μg of vaccines on days 0 and 14; (IL-4) and (IFN-g), cytotoxic T lymphocyte (CTL) activity, and specific immunoglobulin G (IgG) titer and IgG1, IgG2a, and IgG2a/IgG1 ratio, and anti-receptor-binding domain (RBD) IgG response were assessed 2 weeks after the final immunization. Immunization with SARS-CoV-2-Montanide ISA51VG showed a significant increase in the IFN-γ cytokine versus SARS-CoV-2-Alum, SARS-CoV-2-Montanide ISA720VG, and control groups (p < 0.0033). Cytokine IL-4 response in SARS-CoV-2-Alum group showed a significant increase compared with SARS-CoV-2-Montanide ISA51VG, SARS-CoV-2-Montanide ISA720VG, and control groups (p < 0.0206). In addition, SARS-CoV-2-Montanide ISA51VG vaccine induced the highest IFN-γ/IL-4 cytokine ratio versus other groups (p < 0.0004). CTL activity in SARS-CoV-2-Montanide ISA51VG and SARS-CoV-2-Montanide ISA720VG groups showed a significant increase compared with SARS-CoV-2-Alum and control groups (p < 0.0075). Specific IgG titer in SARS-CoV-2-Montanide ISA51 VG and SARS-CoV-2-Montanide ISA720VG showed a significant increase compared with SARS-CoV-2-Alum and control groups (p < 0.0143). Results from specific IgG1and IgG2a in SARS-CoV-2-Alum, SARS-CoV-2-Montanide ISA51VG, and SARS-CoV-2-Montanide ISA720VG vaccine showed a significant increase compared with phosphate buffer saline (PBS) group (p < 0.0001), but SARS-CoV-2-Montanide ISA51VG and SARS-CoV-2-Montanide ISA 720VG groups showed the highest IgG2a/IgG1 ratio and a significant increase compared with SARS-CoV-2-Alum group (p < 0.0379). Moreover, inactivated SARS-CoV-2+Alum and SARS-CoV-2-Montanide ISA 720VG groups demonstrated a significant increase in anti-RBD IgG response versus the SARS-CoV-2-Montanide ISA51VG group. It seems that the type of vaccine formulation is a critical parameter, influencing the immunologic pattern and vaccine potency and human-compatible oil-based adjuvants were more potent than Alum adjuvant in the vaccine formulation.
Collapse
Affiliation(s)
- Melika Haghighi
- Department of FMD Vaccine Production, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Akbar Khorasani
- Department of FMD Vaccine Production, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Pegah Karimi
- Department of Biochemistry, Faculty of Basic Sciences, Islamic Azad University, Tehran, Iran
| | - Rouhollah Keshavarz
- PPD Tuberculin Department, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran.,Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Immunotherapy Group, The Institute of Pharmaceutical Science (TIPS), Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
7
|
González-López C, Chen WH, Alfaro-Chacón A, Villanueva-Lizama LE, Rosado-Vallado M, Ramirez-Sierra MJ, Teh-Poot CF, Pollet J, Asojo O, Jones KM, Hotez PJ, Elena Bottazzi M, Cruz-Chan JV. A novel multi-epitope recombinant protein elicits an antigen-specific CD8+ T cells response in Trypanosoma cruzi-infected mice. Vaccine 2022; 40:6445-6449. [PMID: 36184402 DOI: 10.1016/j.vaccine.2022.09.068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 02/05/2023]
Abstract
About 6.5 million people worldwide are afflicted by Chagas disease, which is caused by the protozoan parasite Trypanosoma cruzi. The development of a therapeutic vaccine to prevent the progression of Chagasic cardiomyopathy has been proposed as an alternative for antiparasitic chemotherapy. Bioinformatics tools can predict MHC class I CD8 + epitopes for inclusion in a single recombinant protein with the goal to develop a multivalent vaccine. We expressed a novel recombinant protein Tc24-C4.10E harboring ten nonameric CD8 + epitopes and using Tc24-C4 protein as scaffold to evaluate the therapeutic effect in acute T. cruzi infection. T. cruzi-infected mice were immunized with Tc24-C4.10E or Tc24-C4 in a 50-day model of acute infection. Tc24-C4.10E-treated mice showed a decreased parasitemia compared to the Tc24-C4 (non-adjuvant) immunized mice or control group. Moreover, Tc24-C4.10E induced a higher stimulation index of CD8 + T cells producing IFNγ and IL-4 cytokines. These results suggest that the addition of the MHC Class I epitopes to Tc24-C4 can synergize the antigen-specific cellular immune responses, providing proof-of-concept that this approach could lead to the development of a promising vaccine candidate for Chagas disease.
Collapse
Affiliation(s)
- Cristina González-López
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México; Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Wen-Hsiang Chen
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Departments of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Andrea Alfaro-Chacón
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Liliana E Villanueva-Lizama
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México; Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Miguel Rosado-Vallado
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Maria Jesús Ramirez-Sierra
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Christian F Teh-Poot
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Jeroen Pollet
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Departments of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Oluwatoyin Asojo
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Chemistry and Biochemistry, Hampton University, VA, USA
| | - Kathryn M Jones
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Departments of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Departments of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Departments of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Julio Vladimir Cruz-Chan
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México; Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
8
|
Dzul-Huchim VM, Ramirez-Sierra MJ, Martinez-Vega PP, Rosado-Vallado ME, Arana-Argaez VE, Ortega-Lopez J, Gusovsky F, Dumonteil E, Cruz-Chan JV, Hotez P, Bottazzi ME, Villanueva-Lizama LE. Vaccine-linked chemotherapy with a low dose of benznidazole plus a bivalent recombinant protein vaccine prevents the development of cardiac fibrosis caused by Trypanosoma cruzi in chronically-infected BALB/c mice. PLoS Negl Trop Dis 2022; 16:e0010258. [PMID: 36095001 PMCID: PMC9499242 DOI: 10.1371/journal.pntd.0010258] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/22/2022] [Accepted: 08/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background Chagas disease (CD) is caused by Trypanosoma cruzi and affects 6–7 million people worldwide. Approximately 30% of chronic patients develop chronic chagasic cardiomyopathy (CCC) after decades. Benznidazole (BNZ), one of the first-line chemotherapy used for CD, induces toxicity and fails to halt the progression of CCC in chronic patients. The recombinant parasite-derived antigens, including Tc24, Tc24-C4, TSA-1, and TSA-1-C4 with Toll-like receptor 4 (TLR-4) agonist-adjuvants reduce cardiac parasite burdens, heart inflammation, and fibrosis, leading us to envision their use as immunotherapy together with BNZ. Given genetic immunization (DNA vaccines) encoding Tc24 and TSA-1 induce protective immunity in mice and dogs, we propose that immunization with the corresponding recombinant proteins offers an alternative and feasible strategy to develop these antigens as a bivalent human vaccine. We hypothesized that a low dose of BNZ in combination with a therapeutic vaccine (TSA-1-C4 and Tc24-C4 antigens formulated with a synthetic TLR-4 agonist-adjuvant, E6020-SE) given during early chronic infection, could prevent cardiac disease progression and provide antigen-specific T cell immunity. Methodology/ Principal findings We evaluated the therapeutic vaccine candidate plus BNZ (25 mg/kg/day/7 days) given on days 72 and 79 post-infection (p.i) (early chronic phase). Fibrosis, inflammation, and parasite burden were quantified in heart tissue at day 200 p.i. (late chronic phase). Further, spleen cells were collected to evaluate antigen-specific CD4+ and CD8+ T cell immune response, using flow cytometry. We found that vaccine-linked BNZ treated mice had lower cardiac fibrosis compared to the infected untreated control group. Moreover, cells from mice that received the immunotherapy had higher stimulation index of antigen-specific CD8+Perforin+ T cells as well as antigen-specific central memory T cells compared to the infected untreated control. Conclusions Our results suggest that the bivalent immunotherapy together with BNZ treatment given during early chronic infection protects BALB/c mice against cardiac fibrosis progression and activates a strong CD8+ T cell response by in vitro restimulation, evidencing the induction of a long-lasting T. cruzi-immunity. Chagas disease (CD) is a neglected tropical disease caused by the parasite Trypanosoma cruzi, transmitted through contact with infected feces of vectors bugs. CD can induce cardiac abnormalities including the development of fibrosis and eventually death. Benznidazole (BNZ) is the first-line drug approved against CD, however, its toxicity and lack of efficacy in the chronic phase have limited its use. Previous studies have demonstrated the feasibility of reducing doses of BNZ given in combination with therapeutic vaccines during the acute phase of CD, which increases its tolerability and reduces adverse side effects. Considering that patients are often diagnosed until more advanced stages of the disease, its necessary to evaluate therapies given in the chronic phase of CD. In this study, we evaluated a vaccine formulated with the recombinant T. cruzi-antigens TSA-1-C4 and Tc24-C4 and the adjuvant E6020-SE in combination with a low dose of BNZ given during the chronic phase of T. cruzi-infection using a murine model. The authors found that the combination therapy protects mice against cardiac fibrosis progression, allows the activation of a CD8+ T cell response, and induces a prolonged memory response against T. cruzi. This study supports the development of the vaccine-linked chemotherapy approach to prevent T. cruzi chronic infection.
Collapse
Affiliation(s)
- Victor Manuel Dzul-Huchim
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Maria Jesus Ramirez-Sierra
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Pedro Pablo Martinez-Vega
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Miguel Enrique Rosado-Vallado
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Victor Ermilo Arana-Argaez
- Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Jaime Ortega-Lopez
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Fabian Gusovsky
- Eisai, Inc., Eisai Inc, Andover, Massachusetts, United States of America
| | - Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Los Angeles, United States of America
| | - Julio Vladimir Cruz-Chan
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Peter Hotez
- Texas Children’s Center for Vaccine Development, Departments of Pediatrics and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - María Elena Bottazzi
- Texas Children’s Center for Vaccine Development, Departments of Pediatrics and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Liliana Estefania Villanueva-Lizama
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
- * E-mail:
| |
Collapse
|
9
|
Roa-Velázquez D, Xoconostle-Cázares B, Benítez-Cardoza CG, Ortega-López J, Shoshani L, Morales-Ríos E, Gallardo-Hernández S. Expression, purification, and refolding of the recombinant extracellular domain β 1-subunit of the dog Na +/K +-ATPase of the epithelial cells. Protein Expr Purif 2022; 200:106167. [PMID: 36057422 DOI: 10.1016/j.pep.2022.106167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022]
Abstract
The β1-subunit of the Na+/K+-ATPase is a cell membrane protein, beyond its classic functions, it is also a cell adhesion molecule. β1-subunits on the lateral membrane of dog kidney epithelial cells trans-interact with β1-subunits from another neighboring cells. The β-β interaction is essential for the formation and stabilization of intercellular junctions. Previous studies on site-directed mutagenesis and in silico revealed that the interaction interface involves residues 198-207 and 221-229. However, it is necessary to report the interaction interface at the structural level experimentally. Here, we describe the successful cloning, overexpression in E. coli, and purification of the extracellular domain of the β1-subunit from inclusion bodies. Experimental characterization by size exclusion chromatography and DLS indicated similar hydrodynamic properties of the protein refolded. Structural analysis by circular dichroism and Raman spectroscopy revealed the secondary structures in the folded protein of type β-sheet, α-helix, random coil, and turn. We also performed β1-β1 interaction assays with the recombinant protein, showing dimers' formation (6xHisβ1-β1). Given our results, the recombinant extracellular domain of the β1-subunit is highly similar to the native protein, therefore the current work in our laboratory aims to characterize at the atomic level the interaction interface between EDβ1.
Collapse
Affiliation(s)
- Daniela Roa-Velázquez
- Programa de Doctorado en Nanociencias y Nanotecnología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Beatriz Xoconostle-Cázares
- Departamento de Bioingeniería y Biotecnología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Claudia G Benítez-Cardoza
- Laboratorio de Investigación Bioquímica, Escuela Nacional de Medicina y Homeopatía-Instituto Politécnico Nacional, Guillermo Massieu Helguera 239, Ciudad de México, 07320, Mexico.
| | - Jaime Ortega-López
- Departamento de Bioingeniería y Biotecnología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Liora Shoshani
- Departamento de Fisiología Biofísica y Neurociencias, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Edgar Morales-Ríos
- Departamento de Bioquímica, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| | - Salvador Gallardo-Hernández
- Departamento de Física, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Ciudad de México, 07360, Mexico.
| |
Collapse
|
10
|
Paula AT, Ribeiro KVG, Cardoso KF, Bastos DSS, Santos EC, Novaes RD, Cardoso SA, Oliveira LL. Protective immunity triggered by ectonucleoside triphosphate diphosphohydrolase-based biopharmaceuticals attenuates cardiac parasitism and prevents mortality in Trypanosoma cruzi infection. Bioorg Med Chem 2022; 72:116966. [PMID: 35998390 DOI: 10.1016/j.bmc.2022.116966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/04/2022] [Accepted: 08/12/2022] [Indexed: 11/26/2022]
Abstract
Chagas disease is a potentially fatal infection in 21 endemic Latin America countries for which the effectiveness of reference antiparasitic chemotherapy is limited. Thus, we developed three biopharmaceuticals and evaluated the effectiveness of different immunization strategies (recombinant protein NTPDase-1 [rNTPDase-1], DNA plasmid encoding Trypanosoma cruzi NTPDase-1 [TcNTPDase-1] and DNA-NTPDase-1 prime/rNTPDase-1 boost [Prime-boost]) based on the surface ecto-nucleoside triphosphate diphosphohydrolase (ecto-NTPDase) enzyme of T. cruzi in animals challenged with a virulent strain (Y) of this parasite. BALB/c mice were immunized three times at 30 days intervals, challenged with T. cruzi 15 days after the last immunization, and euthanized 30 days after T. cruzi challenge. Our results showed limited polarization of specific anti-ecto-NTPDase immunoglobulins in mice receiving both immunization protocols. Conversely, the Prime-boost strategy stimulated the Th1 protective phenotype, upregulating TNF-α and downregulating IL-10 production while increasing the activation/distribution of CD3+/CD8+, CD4+/CD44hi and CD8+/CD44hi/CD62L cells in immunized and infected mice. Furthermore, IL-6 and IL10 levels were reduced, while the distribution of CD4+/CD44hi and CD3+/CD8+ cells was increased from rNTPDase-1 and DNA-NTPDase1-based immunization strategies. Animals receiving DNA-NTPDase1 and Prime-boost protocols before T. cruzi challenged exhibited an enhanced immunological response associated with IL-17 upregulation and remarkable downregulation of heart parasitism (T. cruzi DNA) and mortality. These findings indicated that NTPDase-1 with Prime-boost strategy induced a protective and sustained Th17 response, enhancing host resistance against T. cruzi. Thus, ecto-NTPDase is a potentially relevant and applicable in the development of biopharmaceuticals with greater immunoprophylactic potential for Chagas disease.
Collapse
Affiliation(s)
| | | | | | | | - Eliziária Cardoso Santos
- School of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, MG, Brazil
| | - Rômulo Dias Novaes
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil
| | - Silvia Almeida Cardoso
- Department of Medicine and Nursing, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil
| | | |
Collapse
|
11
|
Machine Learning-Based Feature Selection and Classification for the Experimental Diagnosis of Trypanosoma cruzi. ELECTRONICS 2022. [DOI: 10.3390/electronics11050785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chagas disease, caused by the Trypanosoma cruzi (T. cruzi) parasite, is the third most common parasitosis worldwide. Most of the infected subjects can remain asymptomatic without an opportune and early detection or an objective diagnostic is not conducted. Frequently, the disease manifests itself after a long time, accompanied by severe heart disease or by sudden death. Thus, the diagnosis is a complex and challenging process where several factors must be considered. In this paper, a novel pipeline is presented integrating temporal data from four modalities (electrocardiography signals, echocardiography images, Doppler spectrum, and ELISA antibody titers), multiple features selection analyses by a univariate analysis and a machine learning-based selection. The method includes an automatic dichotomous classification of animal status (control vs. infected) based on Random Forest, Extremely Randomized Trees, Decision Trees, and Support Vector Machine. The most relevant multimodal attributes found were ELISA (IgGT, IgG1, IgG2a), electrocardiography (SR mean, QT and ST intervals), ascending aorta Doppler signals, and echocardiography (left ventricle diameter during diastole). Concerning automatic classification from selected features, the best accuracy of control vs. acute infection groups was 93.3 ± 13.3% for cross-validation and 100% in the final test; for control vs. chronic infection groups, it was 100% and 100%, respectively. We conclude that the proposed machine learning-based approach can be of help to obtain a robust and objective diagnosis in early T. cruzi infection stages.
Collapse
|
12
|
Pathogen diversity, immunity, and the fate of infections: lessons learned from Trypanosoma cruzi human–host interactions. THE LANCET MICROBE 2022; 3:e711-e722. [DOI: 10.1016/s2666-5247(21)00265-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/02/2023] Open
|
13
|
Ward AI, Lewis MD, Taylor MC, Kelly JM. Incomplete Recruitment of Protective T Cells Is Associated with Trypanosoma cruzi Persistence in the Mouse Colon. Infect Immun 2022; 90:e0038221. [PMID: 34780279 PMCID: PMC8853677 DOI: 10.1128/iai.00382-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Trypanosoma cruzi is the etiological agent of Chagas disease. Following T cell-mediated suppression of acute-phase infection, this intracellular eukaryotic pathogen persists long-term in a limited subset of tissues at extremely low levels. The reasons for this tissue-specific chronicity are not understood. Using a dual bioluminescent-fluorescent reporter strain and highly sensitive tissue imaging that allows experimental infections to be monitored at single-cell resolution, we undertook a systematic analysis of the immunological microenvironments of rare parasitized cells in the mouse colon, a key site of persistence. We demonstrate that incomplete recruitment of T cells to a subset of colonic infection foci permits the occurrence of repeated cycles of intracellular parasite replication and differentiation to motile trypomastigotes at a frequency sufficient to perpetuate chronic infections. The lifelong persistence of parasites in this tissue site continues despite the presence, at a systemic level, of a highly effective T cell response. Overcoming this low-level dynamic host-parasite equilibrium represents a major challenge for vaccine development.
Collapse
Affiliation(s)
- Alexander I. Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
14
|
Lartey NL, Valle-Reyes S, Vargas-Robles H, Jiménez-Camacho KE, Guerrero-Fonseca IM, Castellanos-Martínez R, Montoya-García A, García-Cordero J, Cedillo-Barrón L, Nava P, Filisola-Villaseñor JG, Roa-Velázquez D, Zavala-Vargas DI, Morales-Ríos E, Salinas-Lara C, Vadillo E, Schnoor M. ADAM17/MMP inhibition prevents neutrophilia and lung injury in a mouse model of COVID-19. J Leukoc Biol 2021; 111:1147-1158. [PMID: 34826347 PMCID: PMC9015574 DOI: 10.1002/jlb.3cova0421-195rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/06/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Severe coronavirus disease 2019 (COVID‐19) is characterized by lung injury, cytokine storm, and increased neutrophil‐to‐lymphocyte ratio (NLR). Current therapies focus on reducing viral replication and inflammatory responses, but no specific treatment exists to prevent the development of severe COVID‐19 in infected individuals. Angiotensin‐converting enzyme‐2 (ACE2) is the receptor for SARS‐CoV‐2, the virus causing COVID‐19, but it is also critical for maintaining the correct functionality of lung epithelium and endothelium. Coronaviruses induce activation of a disintegrin and metalloprotease 17 (ADAM17) and shedding of ACE2 from the cell surface resulting in exacerbated inflammatory responses. Thus, we hypothesized that ADAM17 inhibition ameliorates COVID‐19‐related lung inflammation. We employed a preclinical mouse model using intratracheal instillation of a combination of polyinosinic:polycytidylic acid (poly(I:C)) and the receptor‐binding domain of the SARS‐CoV‐2 spike protein (RBD‐S) to mimic lung damage associated with COVID‐19. Histologic analysis of inflamed mice confirmed the expected signs of lung injury including edema, fibrosis, vascular congestion, and leukocyte infiltration. Moreover, inflamed mice also showed an increased NLR as observed in critically ill COVID‐19 patients. Administration of the ADAM17/MMP inhibitors apratastat and TMI‐1 significantly improved lung histology and prevented leukocyte infiltration. Reduced leukocyte recruitment could be explained by reduced production of proinflammatory cytokines and lower levels of the endothelial adhesion molecules ICAM‐1 and VCAM‐1. Additionally, the NLR was significantly reduced by ADAM17/MMP inhibition. Thus, we propose inhibition of ADAM17/MMP as a novel promising treatment strategy in SARS‐CoV‐2‐infected individuals to prevent the progression toward severe COVID‐19.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Porfirio Nava
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | | | | | | | | | | | - Eduardo Vadillo
- Oncology Research Unit, Hospital de Oncología, Centro Médico Nacional Siglo XXI, Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine, CINVESTAV-IPN, Mexico City, Mexico
| |
Collapse
|
15
|
Galindo A, Javier-Reyna R, García-Rivera G, Bañuelos C, Montaño S, Ortega-Lopez J, Chávez-Munguía B, Salazar-Villatoro L, Orozco E. EhVps23: A Component of ESCRT-I That Participates in Vesicular Trafficking and Phagocytosis of Entamoeba histolytica. Front Cell Infect Microbiol 2021; 11:770759. [PMID: 34778112 PMCID: PMC8588831 DOI: 10.3389/fcimb.2021.770759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/12/2021] [Indexed: 11/26/2022] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) is formed by ESCRT-0, ESCRT-I, ESCRT-II, ESCRT-III complexes, and accessory proteins. It conducts vesicular trafficking in eukaryotes through the formation of vesicles and membrane fission and fusion events. The trophozoites of Entamoeba histolytica, the protozoan responsible for human amoebiasis, presents an active membrane movement in basal state that increases during phagocytosis and tissue invasion. ESCRT-III complex has a pivotal role during these events, but ESCRT-0, ESCRT-I and ESCRT-II have been poorly studied. Here, we unveiled the E. histolytica ESCRT-I complex and its implication in vesicular trafficking and phagocytosis, as well as the molecular relationships with other phagocytosis-involved molecules. We found a gene encoding for a putative EhVps23 protein with the ubiquitin-binding and Vps23 core domains. In basal state, it was in the plasma membrane, cytoplasmic vesicles and multivesicular bodies, whereas during phagocytosis it was extensively ubiquitinated and detected in phagosomes and connected vesicles. Docking analysis, immunoprecipitation assays and microscopy studies evidenced its interaction with EhUbiquitin, EhADH, EhVps32 proteins, and the lysobisphosphatidic acid phospholipid. The knocking down of the Ehvps23 gene resulted in lower rates of phagocytosis. Our results disclosed the concert of finely regulated molecules and vesicular structures participating in vesicular trafficking-related events with a pivotal role of EhVps23.
Collapse
Affiliation(s)
- Ausencio Galindo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Rosario Javier-Reyna
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Guillermina García-Rivera
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Cecilia Bañuelos
- Programa Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Sarita Montaño
- Laboratorio de Bioinformática y Simulación Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa, Sinaloa, Mexico
| | - Jaime Ortega-Lopez
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Bibiana Chávez-Munguía
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Lizbeth Salazar-Villatoro
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| |
Collapse
|
16
|
da Costa KM, Marques da Fonseca L, dos Reis JS, Santos MARDC, Previato JO, Mendonça-Previato L, Freire-de-Lima L. Trypanosoma cruzi trans-Sialidase as a Potential Vaccine Target Against Chagas Disease. Front Cell Infect Microbiol 2021; 11:768450. [PMID: 34765570 PMCID: PMC8576188 DOI: 10.3389/fcimb.2021.768450] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/12/2021] [Indexed: 01/21/2023] Open
Abstract
Chagas' disease is caused by the protozoan Trypanosoma cruzi, described in the early 20th century by the Brazilian physician Dr. Carlos Chagas. There was a great amount of research devoted to diagnosis, treatment and prevention of the disease. One of the most important discoveries made since then, impacting the understanding of how the parasite interacts with the host's immune system, was the description of trans-sialidase. It is an unique enzyme, capable of masking the parasite's presence from the host, while at the same time dampening the activation of CD8+ T cells, the most important components of the immune response. Since the description of Chagas' disease in 1909, extensive research has identified important events in the disease in order to understand the biochemical mechanism that modulates T. cruzi-host cell interactions and the ability of the parasite to ensure its survival. The importance of the trans-sialidase enzyme brought life to many studies for the design of diagnostic tests, drugs and vaccines. While many groups have been prolific, such efforts have encountered problems, among them: the fact that while T. cruzi have many genes that are unique to the parasite, it relies on multiple copies of them and the difficulty in providing epitopes that result in effective and robust immune responses. In this review, we aim to convey the importance of trans-sialidase as well as to provide a history, including the initial failures and the most promising successes in the chasing of a working vaccine for a disease that is endemic in many tropical countries, including Brazil.
Collapse
Affiliation(s)
- Kelli Monteiro da Costa
- Laboratório de Glicobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | - Lucia Mendonça-Previato
- Laboratório de Glicobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Freire-de-Lima
- Laboratório de Glicobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Núñez-Muñoz L, Marcelino-Pérez G, Calderón-Pérez B, Pérez-Saldívar M, Acosta-Virgen K, González-Conchillos H, Vargas-Hernández B, Olivares-Martínez A, Ruiz-Medrano R, Roa-Velázquez D, Morales-Ríos E, Ramos-Flores J, Torres-Franco G, Peláez-González D, Fernández-Hernández J, Espinosa-Cantellano M, Tapia-Sidas D, Ramírez-Pool JA, Padilla-Viveros A, Xoconostle-Cázares B. Recombinant Antigens Based on Non-Glycosylated Regions from RBD SARS-CoV-2 as Potential Vaccine Candidates against COVID-19. Vaccines (Basel) 2021; 9:928. [PMID: 34452053 PMCID: PMC8402574 DOI: 10.3390/vaccines9080928] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/22/2023] Open
Abstract
The Receptor-Binding Domain (RBD) of the Spike (S) protein from Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has glycosylation sites which can limit the production of reliable antigens expressed in prokaryotic platforms, due to glycan-mediated evasion of the host immune response. However, protein regions without glycosylated residues capable of inducing neutralizing antibodies could be useful for antigen production in systems that do not carry the glycosylation machinery. To test this hypothesis, the potential antigens NG06 and NG19, located within the non-glycosylated S-RBD region, were selected and expressed in Escherichia coli, purified by FPLC and employed to determine their immunogenic potential through detection of antibodies in serum from immunized rabbits, mice, and COVID-19 patients. IgG antibodies from sera of COVID-19-recovered patients detected the recombinant antigens NG06 and NG19 (A450 nm = 0.80 ± 0.33; 1.13 ± 0.33; and 0.11 ± 0.08 for and negatives controls, respectively). Also, the purified antigens were able to raise polyclonal antibodies in animal models evoking a strong immune response with neutralizing activity in mice model. This research highlights the usefulness of antigens based on the non-N-glycosylated region of RBD from SARS-CoV-2 for candidate vaccine development.
Collapse
Affiliation(s)
- Leandro Núñez-Muñoz
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Gabriel Marcelino-Pérez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
- Doctoral Program in Nanosciences and Nanotechnology, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Berenice Calderón-Pérez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Miriam Pérez-Saldívar
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Karla Acosta-Virgen
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Hugo González-Conchillos
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Brenda Vargas-Hernández
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Ana Olivares-Martínez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Roberto Ruiz-Medrano
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Daniela Roa-Velázquez
- Doctoral Program in Nanosciences and Nanotechnology, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Edgar Morales-Ríos
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Jorge Ramos-Flores
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Gustavo Torres-Franco
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Diana Peláez-González
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Jorge Fernández-Hernández
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Martha Espinosa-Cantellano
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Diana Tapia-Sidas
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - José Abrahan Ramírez-Pool
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - América Padilla-Viveros
- Transdisciplinary Doctoral Program in Scientific and Technological Development for Society, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Beatriz Xoconostle-Cázares
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| |
Collapse
|
18
|
Hernández-Cuevas NA, Marín-Cervera A, Garcia-Polanco S, Martínez-Vega P, Rosado-Vallado M, Dumonteil E. Fibronectin degradation as biomarker for Trypanosoma cruzi infection and treatment monitoring in mice. Parasitology 2021; 148:1067-1073. [PMID: 34024298 PMCID: PMC11010125 DOI: 10.1017/s0031182021000809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 11/06/2022]
Abstract
Biomarkers (coming from host or parasite) to monitor Chagas disease (CD) progression as well as the therapeutic response in chronic CD are critically needed, since seronegativization, which may be considered the best indicator of therapeutic cure, takes several years to be observed in adults. Several molecules have been suggested as biomarkers for CD, however, they have to be validated. Taking advantage of mouse models of Trypanosoma cruzi infection, we investigated changes in the degradation profile of fibronectin in plasma. The degradation profile of fibronectin was different in the acute phase compared to the chronic phase of the infection. Fibronectin fragments of approximately 150, 100, 40 and 30 kDa were identified. Furthermore, those degradation profiles correlated with acute parasitaemia as well as with cardiac parasite burden and tissue damage during the infection. The usefulness of fibronectin degradation as a biomarker for therapeutic response following drug treatment and immunotherapeutic vaccination also was evaluated and a decreased fibronectin degradation profile was observed upon benznidazole or a vaccine candidate treatment.
Collapse
Affiliation(s)
- Nora Adriana Hernández-Cuevas
- Laboratorio de Parasitología, Centro de Investigaciones Regionales ‘Dr. Hideyo Noguchi’, Universidad Autónoma de Yucatán, Mérida, México
| | - Andrea Marín-Cervera
- Laboratorio de Parasitología, Centro de Investigaciones Regionales ‘Dr. Hideyo Noguchi’, Universidad Autónoma de Yucatán, Mérida, México
| | - Shineily Garcia-Polanco
- Laboratorio de Parasitología, Centro de Investigaciones Regionales ‘Dr. Hideyo Noguchi’, Universidad Autónoma de Yucatán, Mérida, México
| | - Pedro Martínez-Vega
- Laboratorio de Parasitología, Centro de Investigaciones Regionales ‘Dr. Hideyo Noguchi’, Universidad Autónoma de Yucatán, Mérida, México
| | - Miguel Rosado-Vallado
- Laboratorio de Parasitología, Centro de Investigaciones Regionales ‘Dr. Hideyo Noguchi’, Universidad Autónoma de Yucatán, Mérida, México
| | - Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| |
Collapse
|
19
|
Ramos-Vega A, Monreal-Escalante E, Dumonteil E, Bañuelos-Hernández B, Angulo C. Plant-made vaccines against parasites: bioinspired perspectives to fight against Chagas disease. Expert Rev Vaccines 2021; 20:1373-1388. [PMID: 33612044 DOI: 10.1080/14760584.2021.1893170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Three decades of evidence have demonstrated that plants are an affordable platform for biopharmaceutical production and delivery. For instance, several plant-made recombinant proteins have been approved for commercialization under good manufacturing practice (GMP). Thus far, plant-based vaccine prototypes have been evaluated at pre- and clinical levels. Particularly, plant-made vaccines against parasitic diseases, such as malaria, cysticercosis, and toxoplasmosis have been successfully produced and orally delivered with promising outcomes in terms of immunogenicity and protection. The experience on several approaches and technical strategies over 30 years accounts for their potential low-cost, high scalability, and easy administration.Areas covered: This platform is an open technology to fight against Chagas disease, one of the most important neglected tropical diseases worldwide.Expert opinion: This review provides a perspective for the potential use of plants as a production platform and delivery system of Trypanosoma cruzi recombinant antigens, analyzing the advantages and limitations with respect to plant-made vaccines produced for other parasitic diseases. Plant-made vaccines are envisioned to fight against Chagas disease and other neglected tropical diseases in those countries suffering endemic prevalence.
Collapse
Affiliation(s)
- Abel Ramos-Vega
- Grupo de Inmunología & Vacunología. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.c.s. C.p., México
| | - Elizabeth Monreal-Escalante
- Grupo de Inmunología & Vacunología. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.c.s. C.p., México.,CONACYT- Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.c.s. C.p, México
| | - Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Bernardo Bañuelos-Hernández
- Facultad de Agronomía Y Veterinaria, Universidad de La Salle Bajio, Avenida Universidad 602, Lomas del Campestre, León Guanajuato, México
| | - Carlos Angulo
- Grupo de Inmunología & Vacunología. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.c.s. C.p., México
| |
Collapse
|
20
|
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
|
21
|
Arnal A, Villanueva‐Lizama L, Teh‐Poot C, Herrera C, Dumonteil E. Extent of polymorphism and selection pressure on the Trypanosoma cruzi vaccine candidate antigen Tc24. Evol Appl 2020; 13:2663-2672. [PMID: 33294015 PMCID: PMC7691455 DOI: 10.1111/eva.13068] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/30/2020] [Accepted: 07/12/2020] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, is a major public health problem in the Americas, and existing drugs have severe limitations. In this context, a vaccine would be an attractive alternative for disease control. One of the difficulties in developing an effective vaccine lies in the high genetic diversity of T. cruzi. In this study, we evaluated the level of sequence diversity of the leading vaccine candidate Tc24 in multiple parasite strains. METHODS AND RESULTS We quantified its level of polymorphism within and between T. cruzi discrete typing units (DTUs) and how this potential polymorphism is structured by different selective pressures. We observed a low level of polymorphism of Tc24 protein, weakly associated with parasite DTUs, but not with the geographic origin of the strains. In particular, Tc24 was under strong purifying selection pressure and predicted CD8+ T-cell epitopes were mostly conserved. Tc24 strong conservation may be associated with structural/functional constrains to preserve EF hand domains and their calcium-binding loops, and Tc24 is likely important for the parasite fitness. DISCUSSION Together, these results show that a vaccine based on Tc24 is likely to be effective against a wide diversity of parasite strains across the American continent, and further development of this vaccine candidate should be a high priority.
Collapse
Affiliation(s)
- Audrey Arnal
- Laboratorio de ParasitologíaCentro de Investigaciones Regionales “DrHideyo Noguchi”Universidad Autónoma de YucatánMéridaMexico
- Departamento de Ecología de la BiodiversidadInstituto de EcologíaUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMéxico
| | - Liliana Villanueva‐Lizama
- Laboratorio de ParasitologíaCentro de Investigaciones Regionales “DrHideyo Noguchi”Universidad Autónoma de YucatánMéridaMexico
| | - Christian Teh‐Poot
- Laboratorio de ParasitologíaCentro de Investigaciones Regionales “DrHideyo Noguchi”Universidad Autónoma de YucatánMéridaMexico
| | - Claudia Herrera
- Department of Tropical MedicineSchool of Public Health and Tropical MedicineTulane UniversityNew OrleansLAUSA
- Vector‐Borne and Infectious Disease Research CenterTulane UniversityNew OrleansLAUSA
| | - Eric Dumonteil
- Department of Tropical MedicineSchool of Public Health and Tropical MedicineTulane UniversityNew OrleansLAUSA
- Vector‐Borne and Infectious Disease Research CenterTulane UniversityNew OrleansLAUSA
| |
Collapse
|
22
|
Cerny N, Bivona AE, Sanchez Alberti A, Trinitario SN, Morales C, Cardoso Landaburu A, Cazorla SI, Malchiodi EL. Cruzipain and Its Physiological Inhibitor, Chagasin, as a DNA-Based Therapeutic Vaccine Against Trypanosoma cruzi. Front Immunol 2020; 11:565142. [PMID: 33162979 PMCID: PMC7583359 DOI: 10.3389/fimmu.2020.565142] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
Chagas disease caused by the protozoan parasite Trypanosoma cruzi is endemic in 21 Latin American countries and the southern United States and now is spreading into several other countries due to migration. Despite the efforts to control the vector throughout the Americas, currently, there are almost seven million infected people worldwide, causing ~10,000 deaths per year, and 70 million people at risk to acquire the infection. Chagas disease treatment is restricted only to two parasiticidal drugs, benznidazole and nifurtimox, which are effective during the acute and early infections but have not been found to be as effective in chronic infection. No prophylactic or therapeutic vaccine for human use has been communicated at this moment. Here, we evaluate in a mouse model a therapeutic DNA vaccine combining Cruzipain (Cz), a T. cruzi cysteine protease that proved to be protective in several settings, and Chagasin (Chg), which is the natural Cz inhibitor. The DNAs of both antigens, as well as a plasmid encoding GM-CSF as adjuvant, were orally administrated and delivered by an attenuated Salmonella strain to treat mice during the acute phase of T. cruzi infection. The bicomponent vaccine based on Salmonella carrying Cz and Chg (SChg+SCz) was able to improve the protection obtained by each antigen as monocomponent therapeutic vaccine and significantly increased the titers of antigen- and parasite-specific antibodies. More importantly, the bicomponent vaccine triggered a robust cellular response with interferon gamma (IFN-γ) secretion that rapidly reduced the parasitemia during the acute phase and decreased the tissue damage in the chronic stage of the infection, suggesting it could be an effective tool to ameliorate the pathology associated to Chagas disease.
Collapse
Affiliation(s)
- Natacha Cerny
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Ricardo A. Margni (IDEHU, UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Departamento de Microbiología Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Augusto Ernesto Bivona
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Ricardo A. Margni (IDEHU, UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Departamento de Microbiología Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andrés Sanchez Alberti
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Ricardo A. Margni (IDEHU, UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Departamento de Microbiología Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sebastián Nicolás Trinitario
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Ricardo A. Margni (IDEHU, UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Departamento de Microbiología Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Celina Morales
- Instituto de Fisiopatología Cardiovascular, Departamento de Patología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro Cardoso Landaburu
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Ricardo A. Margni (IDEHU, UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Departamento de Microbiología Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvia Inés Cazorla
- Instituto de Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Departamento de Microbiología Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Laboratorio de Inmunología, Centro de Referencia Para Lactobacilos (CERELA-CONICET), Tucumán, Argentina
| | - Emilio Luis Malchiodi
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Ricardo A. Margni (IDEHU, UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Departamento de Microbiología Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
23
|
Villanueva-Lizama LE, Cruz-Chan JV, Versteeg L, Teh-Poot CF, Hoffman K, Kendricks A, Keegan B, Pollet J, Gusovsky F, Hotez PJ, Bottazzi ME, Jones KM. TLR4 agonist protects against Trypanosoma cruzi acute lethal infection by decreasing cardiac parasite burdens. Parasite Immunol 2020; 42:e12769. [PMID: 32592180 DOI: 10.1111/pim.12769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 01/11/2023]
Abstract
E6020 is a synthetic agonist of Toll-like receptor-4 (TLR4). The purpose of this study was to evaluate the effect of different doses of E6020-SE on Trypanosoma cruzi-specific immune responses and its ability to confer protection against acute lethal infection in mice. Forty female BALB/c were infected with 500 trypomastigotes of T cruzi H1 strain, divided into four groups (n = 10) and treated at 7- and 14-day post-infection (dpi) with different doses of E6020-SE or PBS (control). Survival was followed for 51 days, mice were euthanized and hearts were collected to evaluate parasite burden, inflammation and fibrosis. We found significantly higher survival and lower parasite burdens in mice injected with E6020-SE at all doses compared to the control group. However, E6020-SE treatment did not significantly reduce cardiac inflammation or fibrosis. On the other hand, E6020-SE modulated Th1 and Th2 cytokines, decreasing IFN-γ and IL-4 in a dose-dependent manner after stimulation with parasite antigens. We conclude that E6020-SE alone increased survival by decreasing cardiac parasite burdens in BALB/c mice acutely infected with T cruzi but failed to prevent cardiac damage. Our results suggest that for optimal protection, a vaccine antigen is necessary to balance and orient a protective immune response.
Collapse
Affiliation(s)
- Liliana E Villanueva-Lizama
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Julio V Cruz-Chan
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Leroy Versteeg
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Christian F Teh-Poot
- Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, México
| | - Kristyn Hoffman
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - April Kendricks
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Brian Keegan
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Jeroen Pollet
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | | | - Peter J Hotez
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Department of Biology, Baylor University, Waco, TX, USA.,James A. Baker III Institute for Public Policy, Rice University, Houston, TX, USA.,Hagler Institute for Advanced Study at Texas A&M University, College Station, TX, USA
| | - Maria Elena Bottazzi
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Department of Biology, Baylor University, Waco, TX, USA
| | - Kathryn M Jones
- Department of Pediatrics and National School of Tropical Medicine, Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
24
|
Dumonteil E, Herrera C, Tu W, Goff K, Fahlberg M, Haupt E, Kaur A, Marx PA, Ortega-Lopez J, Hotez PJ, Bottazzi ME. Safety and immunogenicity of a recombinant vaccine against Trypanosoma cruzi in Rhesus macaques. Vaccine 2020; 38:4584-4591. [PMID: 32417142 DOI: 10.1016/j.vaccine.2020.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/21/2020] [Accepted: 05/05/2020] [Indexed: 11/30/2022]
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi is one of the most important neglected parasitic diseases in the Americas. Vaccines represent an attractive complementary strategy for the control of T. cruzi infection and pre-clinical studies in mice demonstrated that trypomastigote surface antigen (TSA-1) and the flagellar calcium-binding (Tc24) parasite antigens are promising candidates for vaccine development. We performed here the first evaluation of the safety and immunogenicity of two recombinant vaccine antigens (named TSA1-C4 and Tc24-C4) in naïve non-human primates. Three rhesus macaques received 3 doses of each recombinant protein, formulated with E6020 (Eisai Co., Ltd.), a novel Toll-like receptor-4 agonist, in a stable emulsion. All parameters from blood chemistry and blood cell counts were stable over the course of the study and unaffected by the vaccine. A specific IgG response against both antigens was detectable after the first vaccine dose, and increased with the second dose. After three vaccine doses, stimulation of PBMCs with a peptide pool derived from TSA1-C4 resulted in the induction of TSA1-C4-specific TNFα-, IL-2- and IFNγ-producing CD4+ in one or two animals while stimulation with a peptide pool derived from Tc24-C4 only activated IFNγ-producing CD4+T cells in one animal. In two animals there was also activation of TSA1-C4-specific IL2-producing CD8+ T cells. This is the first report of the immunogenicity of T. cruzi-derived recombinant antigens formulated as an emulsion with a TLR4 agonist in a non-human primate model. Our results strongly support the need for further evaluation of the preventive efficacy of this type of vaccine in non-human primates and explore the effect of the vaccine in a therapeutic model of naturally-infected Chagasic non-human primates, which would strengthen the rationale for the clinical development as a human vaccine against Chagas disease.
Collapse
Affiliation(s)
- Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA.
| | - Claudia Herrera
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Weihong Tu
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Kelly Goff
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Marissa Fahlberg
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Erin Haupt
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Preston A Marx
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA; Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Jaime Ortega-Lopez
- Departmento de Biotecnología y Bioingeniería, CINVESTAV, Mexico, D.F., Mexico
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics and National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
25
|
Cerbán FM, Stempin CC, Volpini X, Carrera Silva EA, Gea S, Motran CC. Signaling pathways that regulate Trypanosoma cruzi infection and immune response. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165707. [DOI: 10.1016/j.bbadis.2020.165707] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023]
|
26
|
Drug-cured experimental Trypanosoma cruzi infections confer long-lasting and cross-strain protection. PLoS Negl Trop Dis 2020; 14:e0007717. [PMID: 32302312 PMCID: PMC7190179 DOI: 10.1371/journal.pntd.0007717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 04/29/2020] [Accepted: 02/11/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The long term and complex nature of Chagas disease in humans has restricted studies on vaccine feasibility. Animal models also have limitations due to technical difficulties in monitoring the extremely low parasite burden that is characteristic of chronic stage infections. Advances in imaging technology offer alternative approaches that circumvent these problems. Here, we describe the use of highly sensitive whole body in vivo imaging to assess the efficacy of recombinant viral vector vaccines and benznidazole-cured infections to protect mice from challenge with Trypanosoma cruzi. METHODOLOGY/PRINCIPAL FINDINGS Mice were infected with T. cruzi strains modified to express a red-shifted luciferase reporter. Using bioluminescence imaging, we assessed the degree of immunity to re-infection conferred after benznidazole-cure. Those infected for 14 days or more, prior to the onset of benznidazole treatment, were highly protected from challenge with both homologous and heterologous strains. There was a >99% reduction in parasite burden, with parasites frequently undetectable after homologous challenge. This level of protection was considerably greater than that achieved with recombinant vaccines. It was also independent of the route of infection or size of the challenge inoculum, and was long-lasting, with no significant diminution in immunity after almost a year. When the primary infection was benznidazole-treated after 4 days (before completion of the first cycle of intracellular infection), the degree of protection was much reduced, an outcome associated with a minimal T. cruzi-specific IFN-γ+ T cell response. CONCLUSIONS/SIGNIFICANCE Our findings suggest that a protective Chagas disease vaccine must have the ability to eliminate parasites before they reach organs/tissues, such as the GI tract, where once established, they become largely refractory to the induced immune response.
Collapse
|
27
|
Bivona AE, Alberti AS, Cerny N, Trinitario SN, Malchiodi EL. Chagas disease vaccine design: the search for an efficient Trypanosoma cruzi immune-mediated control. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165658. [PMID: 31904415 DOI: 10.1016/j.bbadis.2019.165658] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/20/2019] [Indexed: 12/21/2022]
Abstract
Chagas disease is currently endemic to 21 Latin-American countries and has also become a global concern because of globalization and mass migration of chronically infected individuals. Prophylactic and therapeutic vaccination might contribute to control the infection and the pathology, as complement of other strategies such as vector control and chemotherapy. Ideal prophylactic vaccine would produce sterilizing immunity; however, a reduction of the parasite burden would prevent progression from Trypanosoma cruzi infection to Chagas disease. A therapeutic vaccine for Chagas disease may improve or even replace the treatment with current drugs which have several side effects and require long term treatment that frequently leads to therapeutic withdrawal. Here, we will review some aspects about sub-unit vaccines, the rationale behind the selection of the immunogen, the role of adjuvants, the advantages and limitations of DNA-based vaccines and the idea of therapeutic vaccines. One of the main limitations to advance vaccine development against Chagas disease is the high number of variables that must be considered and the lack of uniform criteria among research laboratories. To make possible comparisons, much of this review will be focused on experiments that kept many variables constant including antigen mass/doses, type of eukaryotic plasmid, DNA-delivery system, mice strain and sex, lethal and sublethal model of infection, and similar immunogenicity and efficacy assessments.
Collapse
Affiliation(s)
- Augusto E Bivona
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Dr. Ricardo A. Margni (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
| | - Andrés Sánchez Alberti
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Dr. Ricardo A. Margni (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
| | - Natacha Cerny
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Dr. Ricardo A. Margni (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
| | - Sebastián N Trinitario
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Dr. Ricardo A. Margni (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
| | - Emilio L Malchiodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Prof. Dr. Ricardo A. Margni (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.
| |
Collapse
|
28
|
Dumonteil E, Herrera C, Buekens P. A therapeutic preconceptional vaccine against Chagas disease: A novel indication that could reduce congenital transmission and accelerate vaccine development. PLoS Negl Trop Dis 2019; 13:e0006985. [PMID: 30703092 PMCID: PMC6354953 DOI: 10.1371/journal.pntd.0006985] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, United States of America
- * E-mail:
| | - Claudia Herrera
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, United States of America
| | - Pierre Buekens
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| |
Collapse
|