1
|
Ascanio LC, Carroll S, Paniz-Mondolfi A, Ramírez JD. In vitro diagnostic methods of Chagas disease in the clinical laboratory: a scoping review. Front Microbiol 2024; 15:1393992. [PMID: 38746745 PMCID: PMC11091413 DOI: 10.3389/fmicb.2024.1393992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/02/2024] [Indexed: 01/31/2025] Open
Abstract
Background Chagas disease (CD), caused by Trypanosoma cruzi, is a global health concern with expanding geographical reach. Despite improved and accessible test methods, diagnosing CD in its various phases remains complex. The existence of clinical scenarios, including immunosuppressed patients, transplant-related CD reactivation, transfusion-associated cases, and orally transmitted acute infections, adds to the diagnostic challenge. No singular gold standard test exists for all phases, and recommendations from PAHO and the CDC advocate for the use of two serological methods for chronic CD diagnosis, while molecular methods or direct parasite detection are suggested for the acute phase. Given the complexity in the diagnostic landscape of CD, the goal of this scoping review is to characterize available diagnostic tests for CD in the clinical laboratory. Methods A literature search in PubMed was conducted on studies related to In vitro diagnosis (IVD) in humans published in English, Spanish, or Portuguese language as of 28 August 2023, and extended backward with no predefined time frame. Studies underwent title and abstract screening, followed by full-text review. Studies included were classified based on the diagnostic method used. Test methods were grouped as serological, molecular, and other methods. Performance, availability, and regulatory status were also characterized. Results Out of 85 studies included in the final review, 115 different tests were identified. These tests comprised 89 serological test types, 21 molecular test types, and 5 other test methods. Predominant serological tests included ELISA (38 studies, 44.70%), Rapid tests (19 studies, 22.35%), and chemiluminescence (10 studies, 11.76%). Among molecular tests, Polymerase Chain Reaction (PCR) assays were notable. Twenty-eight tests were approved globally for IVD or donor testing, all being serological methods. Molecular assays lacked approval for IVD in the United States, with only European and Colombian regulatory acceptance. Discussion and conclusion Serological tests, specifically ELISAs, remain the most used and commercially available diagnostic methods. This makes sense considering that most Chagas disease diagnoses occur in the chronic phase and that the WHO gold standard relies on 2 serological tests to establish the diagnosis of chronic Chagas. ELISAs are feasible and relatively low-cost, with good performance with sensitivities ranging between 77.4% and 100%, and with specificities ranging between 84.2% and 100%. Molecular methods allow the detection of specific variants but rely on the parasite's presence, which limits their utility to parasitemia levels. Depending on the PCR method and the phase of the disease, the sensitivity ranged from 58.88 to 100% while the mean specificity ranged from 68.8% to 100%. Despite their performance, molecular testing remains mostly unavailable for IVD use. Only 3 molecular tests are approved for IVD, which are available only in Europe. Six commercial serological assays approved by the FDA are available for blood and organ donor screening. Currently, there are no guidelines for testing CD oral outbreaks. Although more evidence is needed on how testing methods should be used in special clinical scenarios, a comprehensive approach of clinical assessment and diagnostics tests, including not IVD methods, is required for an accurate CD diagnosis.
Collapse
Affiliation(s)
- Luis C. Ascanio
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Savannah Carroll
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alberto Paniz-Mondolfi
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Juan David Ramírez
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
2
|
Benevides L, Sacramento LA, Pioto F, Barretto GD, Carregaro V, Silva JS. Blimp-1 signaling pathways in T lymphocytes is essential to control the Trypanosoma cruzi infection-induced inflammation. Front Immunol 2023; 14:1268196. [PMID: 37908369 PMCID: PMC10614018 DOI: 10.3389/fimmu.2023.1268196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/25/2023] [Indexed: 11/02/2023] Open
Abstract
In many infectious diseases, the pathogen-induced inflammatory response could result in protective immunity that should be regulated to prevent tissue damage and death. In fact, in Trypanosoma cruzi infection, the innate immune and the inflammatory response should be perfectly controlled to avoid significant lesions and death. Here, we investigate the role of Blimp-1 expression in T cells in resistance to T. cruzi infection. Therefore, using mice with Blimp-1 deficiency in T cells (CKO) we determined its role in the controlling parasites growth and lesions during the acute phase of infection. Infection of mice with Blimp-1 ablation in T cells resulted failure the cytotoxic CD8+ T cells and in marked Th1-mediated inflammation, high IFN-γ and TNF production, and activation of inflammatory monocyte. Interestingly, despite high nitric-oxide synthase activation (NOS-2), parasitemia and mortality in CKO mice were increased compared with infected WT mice. Furthermore, infected-CKO mice exhibited hepatic lesions characteristic of steatosis, with significant AST and ALT activity. Mechanistically, Blimp-1 signaling in T cells induces cytotoxic CD8+ T cell activation and restricts parasite replication. In contrast, Blimp-1 represses the Th1 response, leading to a decreased monocyte activation, less NOS-2 activation, and, consequently preventing hepatic damage and dysfunction. These data demonstrate that T. cruzi-induced disease is multifactorial and that the increased IFN-γ, NO production, and dysfunction of CD8+ T cells contribute to host death. These findings have important implications for the design of potential vaccines against Chagas disease.
Collapse
Affiliation(s)
- Luciana Benevides
- Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, SP, Brazil
- Department of Biochemistry and Immunology Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Lais A. Sacramento
- Department of Biochemistry and Immunology Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Franciele Pioto
- Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, SP, Brazil
| | | | - Vanessa Carregaro
- Department of Biochemistry and Immunology Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, SP, Brazil
| | - João S. Silva
- Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, SP, Brazil
- Department of Biochemistry and Immunology Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
3
|
Francisco AF, Saade U, Jayawardhana S, Pottel H, Scandale I, Chatelain E, Liehl P, Kelly JM, Zrein M. Comparing in vivo bioluminescence imaging and the Multi-Cruzi immunoassay platform to develop improved Chagas disease diagnostic procedures and biomarkers for monitoring parasitological cure. PLoS Negl Trop Dis 2022; 16:e0010827. [PMID: 36190992 PMCID: PMC9560623 DOI: 10.1371/journal.pntd.0010827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/13/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chagas disease is caused by the protozoan parasite Trypanosoma cruzi and is a serious public health problem throughout Latin America. With 6 million people infected, there is a major international effort to develop new drugs. In the chronic phase of the disease, the parasite burden is extremely low, infections are highly focal at a tissue/organ level, and bloodstream parasites are only intermittently detectable. As a result, clinical trials are constrained by difficulties associated with determining parasitological cure. Even highly sensitive PCR methodologies can be unreliable, with a tendency to produce "false-cure" readouts. Improved diagnostic techniques and biomarkers for cure are therefore an important medical need. METHODOLOGY/PRINCIPAL FINDINGS Using an experimental mouse model, we have combined a multiplex assay system and highly sensitive bioluminescence imaging to evaluate serological procedures for diagnosis of T. cruzi infections and confirmation of parasitological cure. We identified a set of three antigens that in the context of the multiplex serology system, provide a rapid, reactive and highly accurate read-out of both acute and chronic T. cruzi infection. In addition, we describe specific antibody responses where down-regulation can be correlated with benznidazole-mediated parasite reduction and others where upregulation is associated with persistent infection. One specific antibody (IBAG39) highly correlated with the bioluminescence flux and represents a promising therapy monitoring biomarker in mice. CONCLUSIONS/SIGNIFICANCE Robust, high-throughput methodologies for monitoring the efficacy of anti-T. cruzi drug treatment are urgently required. Using our experimental systems, we have identified markers of infection or parasite reduction that merit assessing in a clinical setting for the longitudinal monitoring of drug-treated patients.
Collapse
Affiliation(s)
- Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | | | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Ivan Scandale
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | | | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
- * E-mail:
| | | |
Collapse
|
4
|
Montoya AL, Gil ER, Heydemann EL, Estevao IL, Luna BE, Ellis CC, Jankuru SR, Alarcón de Noya B, Noya O, Zago MP, Almeida IC, Michael K. Specific Recognition of β-Galactofuranose-Containing Glycans of Synthetic Neoglycoproteins by Sera of Chronic Chagas Disease Patients. Molecules 2022; 27:411. [PMID: 35056727 PMCID: PMC8781757 DOI: 10.3390/molecules27020411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 11/20/2022] Open
Abstract
Chagas disease (CD) can be accurately diagnosed by detecting Trypanosoma cruzi in patients' blood using polymerase chain reaction (PCR). However, parasite-derived biomarkers are of great interest for the serological diagnosis and early evaluation of chemotherapeutic efficacy when PCR may fail, owing to a blood parasite load below the method's limit of detection. Previously, we focused on the detection of specific anti-α-galactopyranosyl (α-Gal) antibodies in chronic CD (CCD) patients elicited by α-Gal glycotopes copiously expressed on insect-derived and mammal-dwelling infective parasite stages. Nevertheless, these stages also abundantly express cell surface glycosylphosphatidylinositol (GPI)-anchored glycoproteins and glycoinositolphospholipids (GIPLs) bearing nonreducing terminal β-galactofuranosyl (β-Galf) residues, which are equally foreign to humans and, therefore, highly immunogenic. Here we report that CCD patients' sera react specifically with synthetic β-Galf-containing glycans. We took a reversed immunoglycomics approach that entailed: (a) Synthesis of T. cruzi GIPL-derived Galfβ1,3Manpα-(CH2)3SH (glycan G29SH) and Galfβ1,3Manpα1,2-[Galfβ1,3]Manpα-(CH2)3SH (glycan G32SH); and (b) preparation of neoglycoproteins NGP29b and NGP32b, and their evaluation in a chemiluminescent immunoassay. Receiver-operating characteristic analysis revealed that NGP32b can distinguish CCD sera from sera of healthy individuals with 85.3% sensitivity and 100% specificity. This suggests that Galfβ1,3Manpα1,2-[Galfβ1,3]Manpα is an immunodominant glycotope and that NGP32b could potentially be used as a novel CCD biomarker.
Collapse
Affiliation(s)
- Alba L. Montoya
- Department of Chemistry and Biochemistry, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (A.L.M.); (E.R.G.); (E.L.H.); (S.R.J.)
| | - Eileni R. Gil
- Department of Chemistry and Biochemistry, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (A.L.M.); (E.R.G.); (E.L.H.); (S.R.J.)
| | - Emily L. Heydemann
- Department of Chemistry and Biochemistry, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (A.L.M.); (E.R.G.); (E.L.H.); (S.R.J.)
| | - Igor L. Estevao
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (I.L.E.); (B.E.L.); (C.C.E.)
| | - Bianca E. Luna
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (I.L.E.); (B.E.L.); (C.C.E.)
| | - Cameron C. Ellis
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (I.L.E.); (B.E.L.); (C.C.E.)
| | - Sohan R. Jankuru
- Department of Chemistry and Biochemistry, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (A.L.M.); (E.R.G.); (E.L.H.); (S.R.J.)
| | - Belkisyolé Alarcón de Noya
- Sección de Inmunología, Instituto de Medicina Tropical, Facultad de Medicina, Universidad Central de Venezuela, Caracas 1041-A, Venezuela; (B.A.d.N.); (O.N.)
| | - Oscar Noya
- Sección de Inmunología, Instituto de Medicina Tropical, Facultad de Medicina, Universidad Central de Venezuela, Caracas 1041-A, Venezuela; (B.A.d.N.); (O.N.)
- Centro para Estudios Sobre Malaria, Instituto de Altos Estudios “Dr. Arnoldo Gabaldón”, Instituto Nacional de Higiene Rafael Rangel, Ministerio del Poder Popular para la Salud, Caracas 1041-A, Venezuela
| | - Maria Paola Zago
- Instituto de Patología Experimental, Facultad de Ciencias de la Salud, Universidad Nacional de Salta (UNSa)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Salta 4400, Argentina;
| | - Igor C. Almeida
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (I.L.E.); (B.E.L.); (C.C.E.)
| | - Katja Michael
- Department of Chemistry and Biochemistry, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (A.L.M.); (E.R.G.); (E.L.H.); (S.R.J.)
| |
Collapse
|
5
|
The Oxidative Stress and Chronic Inflammatory Process in Chagas Disease: Role of Exosomes and Contributing Genetic Factors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:4993452. [PMID: 34976301 PMCID: PMC8718323 DOI: 10.1155/2021/4993452] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/27/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022]
Abstract
Chagas disease is a neglected tropical disease caused by the flagellated protozoa Trypanosoma cruzi that affects several million people mainly in Latin American countries. Chagas disease has two phases, which are acute and chronic, both separated by an indeterminate time period in which the infected individual is relatively asymptomatic. The acute phase extends for 40-60 days with atypical and mild symptoms; however, about 30% of the infected patients will develop a symptomatic chronic phase, which is characterized by either cardiac, digestive, neurological, or endocrine problems. Cardiomyopathy is the most important and severe result of Chagas disease, which leads to left ventricular systolic dysfunction, heart failure, and sudden cardiac death. Most deaths are due to heart failure (70%) and sudden death (30%) resulting from cardiomyopathy. During the chronic phase, T. cruzi-infected macrophages respond with the production of proinflammatory cytokines and production of superoxide and nitric oxide by the NADPH oxidase 2 (NOX2) and inducible nitric oxide synthase (iNOS) enzymes, respectively. During the chronic phase, myocardial changes are produced as a result of chronic inflammation, oxidative stress, fibrosis, and cell death. The cellular inflammatory response is mainly the result of activation of the NF-κB-dependent pathway, which activates gene expression of inflammatory cytokines, leading to progressive tissue damage. The persisting production of reactive oxygen species (ROS) is the result of mitochondrial dysfunction in the cardiomyocytes. In this review, we will discuss inflammation and oxidative damage which is produced in the heart during the chronic phase of Chagas disease and recent evidence on the role of macrophages and the production of proinflammatory cytokines during the acute phase and the origin of macrophages/monocytes during the chronic phase of Chagas disease. We will also discuss the contributing factors and mechanisms leading to the chronic inflammation of the cardiac tissue during the chronic phase of the disease as well as the innate and adaptive host immune response. The contribution of genetic factors to the progression of the chronic inflammatory cardiomyopathy of chronic Chagas disease is also discussed. The secreted extracellular vesicles (exosomes) produced for both T. cruzi and infected host cells can play key roles in the host immune response, and those roles are described. Lastly, we describe potential treatments to attenuate the chronic inflammation of the cardiac tissue, designed to improve heart function in chagasic patients.
Collapse
|
6
|
Beasley EA, Pessôa-Pereira D, Scorza BM, Petersen CA. Epidemiologic, Clinical and Immunological Consequences of Co-Infections during Canine Leishmaniosis. Animals (Basel) 2021; 11:3206. [PMID: 34827938 PMCID: PMC8614518 DOI: 10.3390/ani11113206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 11/24/2022] Open
Abstract
Canine leishmaniosis (CanL) is a vector-borne, parasitic disease. CanL is endemic in the Mediterranean basin and South America but also found in Northern Africa, Asia, and the U.S. Regions with both competent sand fly vectors and L. infantum parasites are also endemic for additional infectious diseases that could cause co-infections in dogs. Growing evidence indicates that co-infections can impact immunologic responses and thus the clinical course of both CanL and the comorbid disease(s). The aim for this review is to summarize epidemiologic, clinical, and immunologic factors contributing to eight primary co-infections reported with CanL: Ehrlichia spp., Anaplasma spp., Borrelia spp., Babesia spp., Trypanosoma cruzi, Toxoplasma gondii, Dirofilaria immitis, Paracoccidioides braziliensis. Co-infection causes mechanistic differences in immunity which can alter diagnostics, therapeutic management, and prognosis of dogs with CanL. More research is needed to further explore immunomodulation during CanL co-infection(s) and their clinical impact.
Collapse
Affiliation(s)
- Erin A. Beasley
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (E.A.B.); (D.P.-P.); (B.M.S.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA
| | - Danielle Pessôa-Pereira
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (E.A.B.); (D.P.-P.); (B.M.S.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA
| | - Breanna M. Scorza
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (E.A.B.); (D.P.-P.); (B.M.S.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA
| | - Christine A. Petersen
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (E.A.B.); (D.P.-P.); (B.M.S.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
7
|
Carlier Y, Truyens C, Muraille E. Is Antibody-Dependent Enhancement of Trypanosoma cruzi Infection Contributing to Congenital/Neonatal Chagas Disease? Front Immunol 2021; 12:723516. [PMID: 34566981 PMCID: PMC8461104 DOI: 10.3389/fimmu.2021.723516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/26/2021] [Indexed: 11/13/2022] Open
Abstract
The newborns of women infected with the parasite Trypanosoma cruzi (the agent of Chagas disease) can be infected either before birth (congenitally), or after birth (as e.g., by vector route). Congenital Chagas disease can induce high levels of neonatal morbidity and mortality. Parasite-infected pregnant women transmit antibodies to their fetus. Antibodies, by opsonizing parasites, can promote phagocytosis and killing of T. cruzi by cells expressing FcγR, on the mandatory condition that such cells are sufficiently activated in an inflammatory context. Antibody-dependent enhancement (ADE) is a mechanism well described in viral infections, by which antibodies enhance entry of infectious agents into host cells by exploiting the phagocytic FcγR pathway. Previously reported Chagas disease studies highlighted a severe reduction of the maternal-fetal/neonatal inflammatory context in parasite-transmitting pregnant women and their congenitally infected newborns. Otherwise, experimental observations brought to light ADE of T. cruzi infection (involving FcγR) in mouse pups displaying maternally transferred antibodies, out of an inflammatory context. Herein, based on such data, we discuss the previously unconsidered possibility of a role of ADE in the trans-placental parasite transmission, and/or the development of severe and mortal clinical forms of congenital/neonatal Chagas disease in newborns of T. cruzi-infected mothers.
Collapse
Affiliation(s)
- Yves Carlier
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Bruxelles, Belgium.,Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Carine Truyens
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Eric Muraille
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| |
Collapse
|
8
|
Versteeg L, Adhikari R, Poveda C, Villar-Mondragon MJ, Jones KM, Hotez PJ, Bottazzi ME, Tijhaar E, Pollet J. Location and expression kinetics of Tc24 in different life stages of Trypanosoma cruzi. PLoS Negl Trop Dis 2021; 15:e0009689. [PMID: 34478444 PMCID: PMC8415617 DOI: 10.1371/journal.pntd.0009689] [Citation(s) in RCA: 10] [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: 03/17/2021] [Accepted: 07/27/2021] [Indexed: 12/03/2022] Open
Abstract
Tc24-C4, a modified recombinant flagellar calcium-binding protein of Trypanosoma cruzi, is under development as a therapeutic subunit vaccine candidate to prevent or delay progression of chronic Chagasic cardiomyopathy. When combined with Toll-like receptor agonists, Tc24-C4 immunization reduces parasitemia, parasites in cardiac tissue, and cardiac fibrosis and inflammation in animal models. To support further research on the vaccine candidate and its mechanism of action, murine monoclonal antibodies (mAbs) against Tc24-C4 were generated. Here, we report new findings made with mAb Tc24-C4/884 that detects Tc24-WT and Tc24-C4, as well as native Tc24 in T. cruzi on ELISA, western blots, and different imaging techniques. Surprisingly, detection of Tc24 by Tc24-C/884 in fixed T. cruzi trypomastigotes required permeabilization of the parasite, revealing that Tc24 is not exposed on the surface of T. cruzi, making a direct role of antibodies in the induced protection after Tc24-C4 immunization less likely. We further observed that after immunostaining T. cruzi-infected cells with mAb Tc24-C4/884, the expression of Tc24 decreases significantly when T. cruzi trypomastigotes enter host cells and transform into amastigotes. However, Tc24 is then upregulated in association with parasite flagellar growth linked to re-transformation into the trypomastigote form, prior to host cellular escape. These observations are discussed in the context of potential mechanisms of vaccine immunity.
Collapse
Affiliation(s)
- Leroy Versteeg
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
- Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands
| | - Rakesh Adhikari
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
| | - Cristina Poveda
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
| | - Maria Jose Villar-Mondragon
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kathryn M. Jones
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
| | - Peter J. Hotez
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Maria Elena Bottazzi
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Edwin Tijhaar
- Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands
| | - Jeroen Pollet
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States of America
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
9
|
Choudhuri S, Rios L, Vázquez-Chagoyán JC, Garg NJ. Oxidative stress implications for therapeutic vaccine development against Chagas disease. Expert Rev Vaccines 2021; 20:1395-1406. [PMID: 34406892 DOI: 10.1080/14760584.2021.1969230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Pathogenesis of Chagas disease (CD) caused by the protozoan parasite Trypanosoma cruzi (T. cruzi) involves chronic oxidative and inflammatory stress. In this review, we discuss the research efforts in therapeutic vaccine development to date and the potential challenges imposed by oxidative stress in achieving an efficient therapeutic vaccine against CD. AREAS COVERED This review covers the immune and nonimmune mechanisms of reactive oxygen species production and immune response patterns during T. cruzi infection in CD. A discussion on immunotherapy development efforts, the efficacy of antigen-based immune therapies against T. cruzi, and the role of antioxidants as adjuvants is discussed to provide promising insights to developing future treatment strategies against CD. EXPERT OPINION Administration of therapeutic vaccines can be a good option to confront persistent parasitemia in CD by achieving a rapid, short-lived stimulation of type 1 cell-mediated immunity. At the same time, adjunct therapies could play a critical role in the preservation of mitochondrial metabolism and cardiac muscle contractility in CD. We propose combined therapy with antigen-based vaccine and small molecules to control the pathological oxidative insult would be effective in the conservation of cardiac structure and function in CD.
Collapse
Affiliation(s)
- Subhadip Choudhuri
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lizette Rios
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Juan Carlos Vázquez-Chagoyán
- Centro de Investigación y Estudios Avanzados En Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca, México
| | - Nisha Jain Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Tx, USA
| |
Collapse
|
10
|
Choudhury SD. Nano-Medicines a Hope for Chagas Disease! Front Mol Biosci 2021; 8:655435. [PMID: 34141721 PMCID: PMC8204082 DOI: 10.3389/fmolb.2021.655435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Chagas disease, is a vector-mediated tropical disease whose causative agent is a parasitic protozoan named Trypanosoma cruzi. It is a very severe health issue in South America and Mexico infecting millions of people every year. Protozoan T. cruzi gets transmitted to human through Triatominae, a subfamily of the Reduviidae, and do not have any effective treatment or preventative available. The lack of economic gains from this tropical parasitic infection, has always been the reason behind its negligence by researchers and drug manufacturers for many decades. Hence there is an enormous requirement for more efficient and novel strategies to reduce the fatality associated with these diseases. Even, available diagnosis protocols are outdated and inefficient and there is an urgent need for rapid high throughput diagnostics as well as management protocol. The current advancement of nanotechnology in the field of healthcare has generated hope for better management of many tropical diseases including Chagas disease. Nanoparticulate systems for drug delivery like poloxamer coated nanosuspension of benzimidazole have shown promising results in reducing toxicity, elevating efficacy and bioavailability of the active compound against the pathogen, by prolonging release, thereby increasing the therapeutic index. Moreover, nanoparticle-based drug delivery has shown promising results in inducing the host’s immune response against the pathogen with very few side effects. Besides, advances in diagnostic assays, such as nanosensors, aided in the accurate detection of the parasite. In this review, we provide an insight into the life cycle stages of the pathogen in both vertebrate host and the insect vector, along with an overview of the current therapy for Chagas disease and its limitations; nano carrier-based delivery systems for antichagasic agents, we also address the advancement of nano vaccines and nano-diagnostic techniques, for treatment of Chagas disease, majorly focusing on the novel perspectives in combating the disease.
Collapse
|
11
|
Niborski LL, Potenza M, Chirivi RGS, Simonetti L, Ossowski MS, Grippo V, May M, Staquicini DI, Parodi-Talice A, Robello C, Comini MA, Alonso GD, Raats JMH, Gómez KA. Recombinant antibody against Trypanosoma cruzi from patients with chronic Chagas heart disease recognizes mammalian nervous system. EBioMedicine 2021; 63:103206. [PMID: 33429173 PMCID: PMC7809174 DOI: 10.1016/j.ebiom.2020.103206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/15/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND To deeply understand the role of antibodies in the context of Trypanosoma cruzi infection, we decided to characterize A2R1, a parasite antibody selected from single-chain variable fragment (scFv) phage display libraries constructed from B cells of chronic Chagas heart disease patients. METHODS Immunoblot, ELISA, cytometry, immunofluorescence and immunohistochemical assays were used to characterize A2R1 reactivity. To identify the antibody target, we performed an immunoprecipitation and two-dimensional electrophoresis coupled to mass spectrometry and confirmed A2R1 specific interaction by producing the antigen in different expression systems. Based on these data, we carried out a comparative in silico analysis of the protein target´s orthologues, focusing mainly on post-translational modifications. FINDINGS A2R1 recognizes a parasite protein of ~50 kDa present in all life cycle stages of T. cruzi, as well as in other members of the kinetoplastid family, showing a defined immunofluorescence labeling pattern consistent with the cytoskeleton. A2R1 binds to tubulin, but this interaction relies on its post-translational modifications. Interestingly, this antibody also targets mammalian tubulin only present in brain, staining in and around cell bodies of the human peripheral and central nervous system. INTERPRETATION Our findings demonstrate for the first time the existence of a human antibody against T. cruzi tubulin capable of cross-reacting with a human neural protein. This work re-emphasizes the role of molecular mimicry between host and parasitic antigens in the development of pathological manifestations of T. cruzi infection.
Collapse
Affiliation(s)
- Leticia L Niborski
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires, Argentina
| | - Mariana Potenza
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires, Argentina
| | | | | | - Micaela S Ossowski
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires, Argentina
| | - Vanina Grippo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires, Argentina
| | - Maria May
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Daniela I Staquicini
- Departamento de Microbiología, Inmunología e Parasitología, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Adriana Parodi-Talice
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay; Sección Genética, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Carlos Robello
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay; Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Marcelo A Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Guillermo D Alonso
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires, Argentina
| | | | - Karina A Gómez
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
12
|
Rios LE, Vázquez-Chagoyán JC, Pacheco AO, Zago MP, Garg NJ. Immunity and vaccine development efforts against Trypanosoma cruzi. Acta Trop 2019; 200:105168. [PMID: 31513763 PMCID: PMC7409534 DOI: 10.1016/j.actatropica.2019.105168] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 08/27/2019] [Accepted: 09/07/2019] [Indexed: 12/28/2022]
Abstract
Trypanosoma cruzi (T. cruzi) is the causative agent for Chagas disease (CD). There is a critical lack of methods for prevention of infection or treatment of acute infection and chronic disease. Studies in experimental models have suggested that the protective immunity against T. cruzi infection requires the elicitation of Th1 cytokines, lytic antibodies and the concerted activities of macrophages, T helper cells, and cytotoxic T lymphocytes (CTLs). In this review, we summarize the research efforts in vaccine development to date and the challenges faced in achieving an efficient prophylactic or therapeutic vaccine against human CD.
Collapse
Affiliation(s)
- Lizette E Rios
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | - Juan Carlos Vázquez-Chagoyán
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca, México
| | - Antonio Ortega Pacheco
- Departamento de Salud Animal y Medicina Preventiva, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - M Paola Zago
- Instituto de Patología Experimental, Universidad Nacional de Salta - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Salta, Argentina
| | - Nisha J Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX.
| |
Collapse
|
13
|
Quijia Quezada C, Azevedo CS, Charneau S, Santana JM, Chorilli M, Carneiro MB, Bastos IMD. Advances in nanocarriers as drug delivery systems in Chagas disease. Int J Nanomedicine 2019; 14:6407-6424. [PMID: 31496694 PMCID: PMC6691952 DOI: 10.2147/ijn.s206109] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Chagas disease is one of the most important public health problems in Latin America due to its high mortality and morbidity levels. There is no effective treatment for this disease since drugs are usually toxic with low bioavailability. Serious efforts to achieve disease control and eventual eradication have been unsuccessful to date, emphasizing the need for rapid diagnosis, drug development, and a reliable vaccine. Novel systems for drug and vaccine administration based on nanocarriers represent a promising avenue for Chagas disease treatment. Nanoparticulate systems can reduce toxicity, and increase the efficacy and bioavailability of active compounds by prolonging release, and therefore improve the therapeutic index. Moreover, nanoparticles are able to interact with the host's immune system, modulating the immune response to favour the elimination of pathogenic microorganisms. In addition, new advances in diagnostic assays, such as nanobiosensors, are beneficial in that they enable precise identification of the pathogen. In this review, we provide an overview of the strategies and nanocarrier-based delivery systems for antichagasic agents, such as liposomes, micelles, nanoemulsions, polymeric and non-polymeric nanoparticles. We address recent progress, with a particular focus on the advances of nanovaccines and nanodiagnostics, exploring new perspectives on Chagas disease treatment.
Collapse
Affiliation(s)
- Christian Quijia Quezada
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
- Department of Drugs and Medicines, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Clênia S Azevedo
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Sébastien Charneau
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Jaime M Santana
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Marcella B Carneiro
- Electron Microscopy Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Izabela Marques Dourado Bastos
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| |
Collapse
|
14
|
Trypanosoma cruzi immunoproteome: Calpain-like CAP5.5 differentially detected throughout distinct stages of human Chagas disease cardiomyopathy. J Proteomics 2019; 194:179-190. [DOI: 10.1016/j.jprot.2018.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/22/2018] [Accepted: 11/28/2018] [Indexed: 12/26/2022]
|
15
|
Acevedo GR, Girard MC, Gómez KA. The Unsolved Jigsaw Puzzle of the Immune Response in Chagas Disease. Front Immunol 2018; 9:1929. [PMID: 30197647 PMCID: PMC6117404 DOI: 10.3389/fimmu.2018.01929] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
Trypanosoma cruzi interacts with the different arms of the innate and adaptive host's immune response in a very complex and flowery manner. The history of host-parasite co-evolution has provided this protozoan with means of resisting, escaping or subverting the mechanisms of immunity and establishing a chronic infection. Despite many decades of research on the subject, the infection remains incurable, and the factors that steer chronic Chagas disease from an asymptomatic state to clinical onset are still unclear. As the relationship between T. cruzi and the host immune system is intricate, so is the amount and diversity of scientific knowledge on the matter. Many of the mechanisms of immunity are fairly well understood, but unveiling the factors that lead each of these to success or failure, within the coordinated response as a whole, requires further research. The intention behind this Review is to compile the available information on the different aspects of the immune response, with an emphasis on those phenomena that have been studied and confirmed in the human host. For ease of comprehension, it has been subdivided in sections that cover the main humoral and cell-mediated components involved therein. However, we also intend to underline that these elements are not independent, but function intimately and concertedly. Here, we summarize years of investigation carried out to unravel the puzzling interplay between the host and the parasite.
Collapse
Affiliation(s)
| | | | - Karina A. Gómez
- Laboratorio de Inmunología de las Infecciones por Tripanosomátidos, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
16
|
Díaz-Viraqué F, Chiribao ML, Trochine A, González-Herrera F, Castillo C, Liempi A, Kemmerling U, Maya JD, Robello C. Old Yellow Enzyme from Trypanosoma cruzi Exhibits In Vivo Prostaglandin F 2α Synthase Activity and Has a Key Role in Parasite Infection and Drug Susceptibility. Front Immunol 2018; 9:456. [PMID: 29563916 PMCID: PMC5845897 DOI: 10.3389/fimmu.2018.00456] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/20/2018] [Indexed: 01/26/2023] Open
Abstract
The discovery that trypanosomatids, unicellular organisms of the order Kinetoplastida, are capable of synthesizing prostaglandins raised questions about the role of these molecules during parasitic infections. Multiple studies indicate that prostaglandins could be related to the infection processes and pathogenesis in trypanosomatids. This work aimed to unveil the role of the prostaglandin F2α synthase TcOYE in the establishment of Trypanosoma cruzi infection, the causative agent of Chagas disease. This chronic disease affects several million people in Latin America causing high morbidity and mortality. Here, we propose a prokaryotic evolutionary origin for TcOYE, and then we used in vitro and in vivo experiments to show that T. cruzi prostaglandin F2α synthase plays an important role in modulating the infection process. TcOYE overexpressing parasites were less able to complete the infective cycle in cell culture infections and increased cardiac tissue parasitic load in infected mice. Additionally, parasites overexpressing the enzyme increased PGF2α synthesis from arachidonic acid. Finally, an increase in benznidazole and nifurtimox susceptibility in TcOYE overexpressing parasites showed its participation in activating the currently anti-chagasic drugs, which added to its observed ability to confer resistance to hydrogen peroxide, highlights the relevance of this enzyme in multiple events including host-parasite interaction.
Collapse
Affiliation(s)
| | - María Laura Chiribao
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departamento de Bioquímica, Facultad de Medicina Universidad de la República, Montevideo, Uruguay
| | - Andrea Trochine
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Fabiola González-Herrera
- Programa de Farmacología Molecular y Clínica - ICBM, Facultad de Medicina Universidad de Chile, Santiago de Chile, Chile
| | - Christian Castillo
- Programa de Anatomía y Biología del Desarrollo - ICBM, Facultad de Medicina Universidad De Chile, Santiago de Chile, Chile
| | - Ana Liempi
- Programa de Anatomía y Biología del Desarrollo - ICBM, Facultad de Medicina Universidad De Chile, Santiago de Chile, Chile
| | - Ulrike Kemmerling
- Programa de Anatomía y Biología del Desarrollo - ICBM, Facultad de Medicina Universidad De Chile, Santiago de Chile, Chile
| | - Juan Diego Maya
- Programa de Farmacología Molecular y Clínica - ICBM, Facultad de Medicina Universidad de Chile, Santiago de Chile, Chile
| | - Carlos Robello
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departamento de Bioquímica, Facultad de Medicina Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
17
|
Lidani KCF, Bavia L, Ambrosio AR, de Messias-Reason IJ. The Complement System: A Prey of Trypanosoma cruzi. Front Microbiol 2017; 8:607. [PMID: 28473804 PMCID: PMC5397499 DOI: 10.3389/fmicb.2017.00607] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/24/2017] [Indexed: 12/27/2022] Open
Abstract
Trypanosoma cruzi is a protozoan parasite known to cause Chagas disease (CD), a neglected sickness that affects around 6-8 million people worldwide. Originally, CD was mainly found in Latin America but more recently, it has been spread to countries in North America, Asia, and Europe due the international migration from endemic areas. Thus, at present CD represents an important concern of global public health. Most of individuals that are infected by T. cruzi may remain in asymptomatic form all lifelong, but up to 40% of them will develop cardiomyopathy, digestive mega syndromes, or both. The interaction between the T. cruzi infective forms and host-related immune factors represents a key point for a better understanding of the physiopathology of CD. In this context, the complement, as one of the first line of host defense against infection was shown to play an important role in recognizing T. cruzi metacyclic trypomastigotes and in controlling parasite invasion. The complement consists of at least 35 or more plasma proteins and cell surface receptors/regulators, which can be activated by three pathways: classical (CP), lectin (LP), and alternative (AP). The CP and LP are mainly initiated by immune complexes or pathogen-associated molecular patterns (PAMPs), respectively, whereas AP is spontaneously activated by hydrolysis of C3. Once activated, several relevant complement functions are generated which include opsonization and phagocytosis of particles or microorganisms and cell lysis. An important step during T. cruzi infection is when intracellular trypomastigotes are release to bloodstream where they may be target by complement. Nevertheless, the parasite uses a sequence of events in order to escape from complement-mediated lysis. In fact, several T. cruzi molecules are known to interfere in the initiation of all three pathways and in the assembly of C3 convertase, a key step in the activation of complement. Moreover, T. cruzi promotes secretion of plasma membrane-derived vesicles from host cells, which prevent the activity of C3 convertase C4b2a and thereby may hinder complement. In this review, we aim to present an overview on the strategies used by T. cruzi in order to circumvent the activation of complement and, consequently, its biological effects.
Collapse
Affiliation(s)
| | | | | | - Iara J. de Messias-Reason
- Laboratory of Molecular Immunopathology, Clinical Hospital, Federal University of ParanáCuritiba, Brazil
| |
Collapse
|
18
|
Barry MA, Wang Q, Jones KM, Heffernan MJ, Buhaya MH, Beaumier CM, Keegan BP, Zhan B, Dumonteil E, Bottazzi ME, Hotez PJ. A therapeutic nanoparticle vaccine against Trypanosoma cruzi in a BALB/c mouse model of Chagas disease. Hum Vaccin Immunother 2016; 12:976-87. [PMID: 26890466 DOI: 10.1080/21645515.2015.1119346] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, results in an acute febrile illness that progresses to chronic chagasic cardiomyopathy in 30% of patients. Current treatments have significant side effects and poor efficacy during the chronic phase; therefore, there is an urgent need for new treatment modalities. A robust TH1-mediated immune response correlates with favorable clinical outcomes. A therapeutic vaccine administered to infected individuals could bolster the immune response, thereby slowing or stopping the progression of chagasic cardiomyopathy. Prior work in mice has identified an efficacious T. cruzi DNA vaccine encoding Tc24. To elicit a similar protective cell-mediated immune response to a Tc24 recombinant protein, we utilized a poly(lactic-co-glycolic acid) nanoparticle delivery system in conjunction with CpG motif-containing oligodeoxynucleotides as an immunomodulatory adjuvant. In a BALB/c mouse model, the vaccine produced a TH1-biased immune response, as demonstrated by a significant increase in antigen-specific IFNγ-producing splenocytes, IgG2a titers, and proliferative capacity of CD8(+) T cells. When tested for therapeutic efficacy, significantly reduced systemic parasitemia was seen during peak parasitemia. Additionally, there was a significant reduction in cardiac parasite burden and inflammatory cell infiltrate. This is the first study demonstrating immunogenicity and efficacy of a therapeutic Chagas vaccine using a nanoparticle delivery system.
Collapse
Affiliation(s)
- Meagan A Barry
- a Interdepartmental Program in Translational Biology and Molecular Medicine , Baylor College of Medicine , Houston , TX , USA.,b Medical Scientist Training Program, Baylor College of Medicine , Houston , TX , USA.,c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA
| | - Qian Wang
- c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA
| | - Kathryn M Jones
- c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA.,d National School of Tropical Medicine, Baylor College of Medicine , Houston , TX , USA
| | - Michael J Heffernan
- a Interdepartmental Program in Translational Biology and Molecular Medicine , Baylor College of Medicine , Houston , TX , USA.,c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA.,d National School of Tropical Medicine, Baylor College of Medicine , Houston , TX , USA.,h Department of Molecular Virology and Microbiology , Baylor College of Medicine , Houston , TX , USA
| | - Munir H Buhaya
- e Summer Medical and Research Training Program, Baylor College of Medicine , Houston , TX , USA
| | - Coreen M Beaumier
- c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA
| | - Brian P Keegan
- c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA
| | - Bin Zhan
- c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA
| | - Eric Dumonteil
- f Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr Hideyo Noguchi, Universidad Autónoma de Yucatán , Mérida , Mexico.,g Department of Tropical Medicine , School of Public Health and Tropical Medicine, Tulane University , New Orleans , LA , USA
| | - Maria Elena Bottazzi
- a Interdepartmental Program in Translational Biology and Molecular Medicine , Baylor College of Medicine , Houston , TX , USA.,c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA.,d National School of Tropical Medicine, Baylor College of Medicine , Houston , TX , USA.,h Department of Molecular Virology and Microbiology , Baylor College of Medicine , Houston , TX , USA
| | - Peter J Hotez
- a Interdepartmental Program in Translational Biology and Molecular Medicine , Baylor College of Medicine , Houston , TX , USA.,c Department of Pediatrics , Section of Pediatric Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine , Houston , TX , USA.,d National School of Tropical Medicine, Baylor College of Medicine , Houston , TX , USA.,h Department of Molecular Virology and Microbiology , Baylor College of Medicine , Houston , TX , USA
| |
Collapse
|
19
|
de Morais CGV, Castro Lima AK, Terra R, dos Santos RF, Da-Silva SAG, Dutra PML. The Dialogue of the Host-Parasite Relationship: Leishmania spp. and Trypanosoma cruzi Infection. BIOMED RESEARCH INTERNATIONAL 2015; 2015:324915. [PMID: 26090399 PMCID: PMC4450238 DOI: 10.1155/2015/324915] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 01/11/2023]
Abstract
The intracellular protozoa Leishmania spp. and Trypanosoma cruzi and the causative agents of Leishmaniasis and Chagas disease, respectively, belong to the Trypanosomatidae family. Together, these two neglected tropical diseases affect approximately 25 million people worldwide. Whether the host can control the infection or develops disease depends on the complex interaction between parasite and host. Parasite surface and secreted molecules are involved in triggering specific signaling pathways essential for parasite entry and intracellular survival. The recognition of the parasite antigens by host immune cells generates a specific immune response. Leishmania spp. and T. cruzi have a multifaceted repertoire of strategies to evade or subvert the immune system by interfering with a range of signal transduction pathways in host cells, which causes the inhibition of the protective response and contributes to their persistence in the host. The current therapeutic strategies in leishmaniasis and trypanosomiasis are very limited. Efficacy is variable, toxicity is high, and the emergence of resistance is increasingly common. In this review, we discuss the molecular basis of the host-parasite interaction of Leishmania and Trypanosoma cruzi infection and their mechanisms of subverting the immune response and how this knowledge can be used as a tool for the development of new drugs.
Collapse
Affiliation(s)
- Carlos Gustavo Vieira de Morais
- Laboratório de Bioquímica de Protozoários e Imunofisiologia do Exercício, Disciplina de Parasitologia, DMIP, FCM, Universidade do Estado do Rio de Janeiro, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 5° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
- Programa de Pós Graduação em Microbiologia/FCM/UERJ, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 3° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Ana Karina Castro Lima
- Laboratório de Bioquímica de Protozoários e Imunofisiologia do Exercício, Disciplina de Parasitologia, DMIP, FCM, Universidade do Estado do Rio de Janeiro, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 5° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Rodrigo Terra
- Laboratório de Bioquímica de Protozoários e Imunofisiologia do Exercício, Disciplina de Parasitologia, DMIP, FCM, Universidade do Estado do Rio de Janeiro, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 5° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
- Programa de Pós Graduação em Fisiopatologia Clínica e Experimental/FCM/UERJ, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 5° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Rosiane Freire dos Santos
- Programa de Pós Graduação em Microbiologia/FCM/UERJ, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 3° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
- Laboratório de Imunofarmacologia Parasitária, Disciplina de Parasitologia, DMIP, FCM, Universidade do Estado do Rio de Janeiro, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 5° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Silvia Amaral Gonçalves Da-Silva
- Laboratório de Imunofarmacologia Parasitária, Disciplina de Parasitologia, DMIP, FCM, Universidade do Estado do Rio de Janeiro, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 5° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Patrícia Maria Lourenço Dutra
- Laboratório de Bioquímica de Protozoários e Imunofisiologia do Exercício, Disciplina de Parasitologia, DMIP, FCM, Universidade do Estado do Rio de Janeiro, Avenida Professor Manuel de Abreu 444, Pavilhão Américo Piquet Carneiro, 5° andar, Vila Isabel, 20550-170 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
20
|
Austen JM, Ryan U, Ditcham WGF, Friend JA, Reid SA. The innate resistance of Trypanosoma copemani to human serum. Exp Parasitol 2015; 153:105-10. [PMID: 25816975 DOI: 10.1016/j.exppara.2015.03.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/12/2015] [Accepted: 03/20/2015] [Indexed: 11/26/2022]
Abstract
Trypanosoma copemani is known to be infective to a variety of Australian marsupials. Characterisation of this parasite revealed the presence of stercorarian-like life-cycle stages in culture, which are similar to T. rangeli and T. cruzi. The blood incubation infectivity test (BIIT) was adapted and used to determine if T. copemani, like T. cruzi and T. rangeli, has the potential to grow in the presence of human serum. To eliminate any effects of anticoagulants on the complement system and on human high density lipoprotein (HDL), only fresh whole human blood was used. Trypanosoma copemani was observed by microscopy in all human blood cultures from day 5 to day 19 post inoculation (PI). The mechanism for normal human serum (NHS) resistance in T. copemani is not known. The results of this study show that at least one native Australian trypanosome species may have the potential to be infective for humans.
Collapse
Affiliation(s)
- J M Austen
- School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch, Western Australia 6150, Australia
| | - U Ryan
- School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch, Western Australia 6150, Australia.
| | - W G F Ditcham
- School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch, Western Australia 6150, Australia
| | - J A Friend
- Department of Parks and Wildlife, 120 Albany Highway, Albany, Western Australia 6330, Australia
| | - S A Reid
- School of Population Health, Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston Road, Herston, QLD 4006, Australia
| |
Collapse
|
21
|
Schmitz V, Almeida LN, Svensjö E, Monteiro AC, Köhl J, Scharfstein J. C5a and Bradykinin Receptor Cross-Talk Regulates Innate and Adaptive Immunity inTrypanosoma cruziInfection. THE JOURNAL OF IMMUNOLOGY 2014; 193:3613-23. [DOI: 10.4049/jimmunol.1302417] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
22
|
Santamaria C, Chatelain E, Jackson Y, Miao Q, Ward BJ, Chappuis F, Ndao M. Serum biomarkers predictive of cure in Chagas disease patients after nifurtimox treatment. BMC Infect Dis 2014; 14:302. [PMID: 24894358 PMCID: PMC4059459 DOI: 10.1186/1471-2334-14-302] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/28/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Chagas disease (CD), caused by the protozoan Trypanosoma cruzi, remains an important public health issue in many Central and South American countries, as well as non-endemic areas with high rates of immigration from these countries. Existing treatment options for CD are limited and often unsatisfactory. Moreover the lack of post-treatment tests of cure limits the development of new drugs. To address this issue, we sought to identify serum biomarkers following nifurtimox (Nfx) treatment that could be used as an early test of cure and/or markers of a therapeutic response. METHODS Human sera from Chagas patients pre- and post-treatment with Nfx (n = 37) were compared to samples from healthy subjects (n = 37) using a range of proteomic and immunologic techniques. Biomarker peaks with the best discriminatory power were further characterized. RESULTS Using serum samples (n = 111), we validated the presence of five key biomarkers identified in our previous study, namely human apolipoprotein A-I (APOA1) and specific fragments thereof and one fragment of human fibronectin (FN1). In chagasic serum samples all biomarkers except full-length APOA1 were upregulated. These five biomarkers returned to normal in 43% (16/37) of the patients treated with Nfx at three years after treatment. CONCLUSIONS The normalization of biomarker patterns strongly associated with CD suggests that these markers can be used to identify patients in whom Nfx treatment is successful. We believe that these are the first biomarkers predictive of cure in CD patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Momar Ndao
- National Reference Center for Parasitology, Research Institute of the McGill University Health Centre, Department of Medicine, Division of Infectious Diseases, Montreal General Hospital, 1650 Cedar Ave,, Room R3-137, Montreal, Quebec H3G 1A4, Canada.
| |
Collapse
|
23
|
Interspecific variation in redox status regulation and immune defence in five bat species: the role of ectoparasites. Oecologia 2014; 175:811-23. [DOI: 10.1007/s00442-014-2959-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 04/18/2014] [Indexed: 12/31/2022]
|
24
|
Salgado-Jiménez B, Arce-Fonseca M, Baylón-Pacheco L, Talamás-Rohana P, Rosales-Encina JL. Differential immune response in mice immunized with the A, R or C domain from TcSP protein of Trypanosoma cruzi or with the coding DNAs. Parasite Immunol 2013; 35:32-41. [PMID: 23106492 DOI: 10.1111/pim.12017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 10/17/2012] [Indexed: 11/26/2022]
Abstract
In a murine model of experimental Trypanosoma cruzi (H8 strain) infection, we investigated the induction of protective immunity against the domains [amino (A), repeats (R) and carboxyl (C)] of the surface protein (SP), a member of the trans-sialidase (TS) superfamily. Recombinant proteins and plasmid DNA coding for the respective proteins were used to immunize BALB/c mice, and the humoral response and cytokine levels were analysed. Immunization with the recombinant proteins induced higher levels of anti-TcSP antibodies than immunization with the corresponding DNAs, and analysis of serum cytokines showed that immunization with both recombinant proteins and naked DNA resulted in a Th1-Th2 mixed T-cell response. Mice immunized with either recombinant proteins or plasmid DNA were infected with blood trypomastigotes. The recombinant protein-immunized mice showed a variable reduction in peak parasitemia, and most died by day 60. Only the pBKTcSPR-immunized mice exhibited a significant reduction in peak parasitemia and survived the lethal challenge. DNA-based immunization with DNA coding for the repeats domain of TcSP is a good candidate for the development of a vaccine against experimental T. cruzi infection.
Collapse
Affiliation(s)
- B Salgado-Jiménez
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, México, D.F, México
| | | | | | | | | |
Collapse
|
25
|
Arce-Fonseca M, Ballinas-Verdugo MA, Zenteno ERA, Suárez-Flores D, Carrillo-Sánchez SC, Alejandre-Aguilar R, Rosales-Encina JL, Reyes PA, Rodríguez-Morales O. Specific humoral and cellular immunity induced by Trypanosoma cruzi DNA immunization in a canine model. Vet Res 2013; 44:15. [PMID: 23497041 PMCID: PMC3601012 DOI: 10.1186/1297-9716-44-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 02/07/2013] [Indexed: 11/18/2022] Open
Abstract
Chagas disease has a high incidence in Mexico and other Latin American countries. Because one of the most important known methods of prevention is vector control, which has been effective only in certain areas of South America, the development of a vaccine to protect people at risk has been proposed. In this study, we assessed the cellular and humoral immune response generated following immunization with pBCSP and pBCSSP4 plasmids containing the genes encoding a trans-sialidase protein (present in all three forms of T. cruzi) and an amastigote specific glycoprotein, respectively, in a canine model. Thirty-five beagle dogs were divided randomly into 5 groups (n = 7) and were immunized twice intramuscularly with 500 μg of pBCSSP4, pBCSP, pBk-CMV (empty plasmid) or saline solution. Fifteen days after the last immunization the 4 groups were infected intraperitoneally with 500 000 metacyclic trypomastigotes. The fifth group was unimmunized/infected. The parasitaemia in the immunized/infected dogs was for a shorter period (14 vs. 29 days) and the parasite load was lower. The concentration of IgG1 (0.612 ± 0.019 O.D.) and IgG2 (1.167 ± 0.097 O.D.) subclasses was measured (absorbance) 15 days after the last immunization with both recombinant plasmids, the majority of which were IgG2. The treatment of parasites using the serum from dogs immunized with pBCSP and pBCSSP4 plasmids produced 54% (± 11.8) and 68% (± 21.4) complement-mediated lysis, respectively. At 12 h post immunization, an increase in cytokines was not observed; however, vaccination with pBCSSP4 significantly increased the levels of IFN-γ and IL-10 at 9 months post-infection. The recombinant plasmid immunization stimulated the spleen cell proliferation showing a positive stimulatory index above 2.0. In conclusion, immunization using both genes effectively induces a humoral and cellular immune response.
Collapse
Affiliation(s)
- Minerva Arce-Fonseca
- Department of Molecular Biology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano No, 1, Col, Sección XVI, Tlalpan, Mexico City, CP 14080, Mexico.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gupta S, Wan X, Zago MP, Sellers VCM, Silva TS, Assiah D, Dhiman M, Nuñez S, Petersen JR, Vázquez-Chagoyán JC, Estrada-Franco JG, Garg NJ. Antigenicity and diagnostic potential of vaccine candidates in human Chagas disease. PLoS Negl Trop Dis 2013; 7:e2018. [PMID: 23350012 PMCID: PMC3547861 DOI: 10.1371/journal.pntd.0002018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 12/04/2012] [Indexed: 12/15/2022] Open
Abstract
Background Chagas disease, caused by Trypanosoma cruzi, is endemic in Latin America and an emerging infectious disease in the US and Europe. We have shown TcG1, TcG2, and TcG4 antigens elicit protective immunity to T. cruzi in mice and dogs. Herein, we investigated antigenicity of the recombinant proteins in humans to determine their potential utility for the development of next generation diagnostics for screening of T. cruzi infection and Chagas disease. Methods and Results Sera samples from inhabitants of the endemic areas of Argentina-Bolivia and Mexico-Guatemala were analyzed in 1st-phase for anti-T. cruzi antibody response by traditional serology tests; and in 2nd-phase for antibody response to the recombinant antigens (individually or mixed) by an ELISA. We noted similar antibody response to candidate antigens in sera samples from inhabitants of Argentina and Mexico (n = 175). The IgG antibodies to TcG1, TcG2, and TcG4 (individually) and TcGmix were present in 62–71%, 65–78% and 72–82%, and 89–93% of the subjects, respectively, identified to be seropositive by traditional serology. Recombinant TcG1- (93.6%), TcG2- (96%), TcG4- (94.6%) and TcGmix- (98%) based ELISA exhibited significantly higher specificity compared to that noted for T. cruzi trypomastigote-based ELISA (77.8%) in diagnosing T. cruzi-infection and avoiding cross-reactivity to Leishmania spp. No significant correlation was noted in the sera levels of antibody response and clinical severity of Chagas disease in seropositive subjects. Conclusions Three candidate antigens were recognized by antibody response in chagasic patients from two distinct study sites and expressed in diverse strains of the circulating parasites. A multiplex ELISA detecting antibody response to three antigens was highly sensitive and specific in diagnosing T. cruzi infection in humans, suggesting that a diagnostic kit based on TcG1, TcG2 and TcG4 recombinant proteins will be useful in diverse situations. Chagas disease is the most common cause of congestive heart failure related deaths among young adults in the endemic areas of South and Central America and Mexico. Diagnosis and treatment of T. cruzi infection has remained difficult and challenging after 100 years of its identification. In >95% of human cases, T. cruzi infection remains undiagnosed until several years later when chronic evolution of progressive disease results in clinical symptoms associated with cardiac damage. Diagnosis generally depends on the measurement of T. cruzi–specific antibodies that can result in false positives. A conclusive diagnosis of T. cruzi infection thus often requires multiple serological tests, in combination with epidemiological data and clinical symptoms. In this study, we investigated the antibody response to TcG1, TcG2, and TcG4 in clinically characterized chagasic patients. These antigens were identified as vaccine candidates and shown to elicit protective immunity to T. cruzi and Chagas disease in experimental animals. Our data show the serology test developed using the TcGmix (multiplex ELISA) is a significantly better alternative to epimastigote extracts currently used in T. cruzi serodiagnosis or the trypomastigote lysate used in this study for comparison purposes.
Collapse
Affiliation(s)
- Shivali Gupta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Machado FS, Dutra WO, Esper L, Gollob K, Teixeira MM, Factor SM, Weiss LM, Nagajyothi F, Tanowitz HB, Garg NJ. Current understanding of immunity to Trypanosoma cruzi infection and pathogenesis of Chagas disease. Semin Immunopathol 2012; 34:753-70. [PMID: 23076807 PMCID: PMC3498515 DOI: 10.1007/s00281-012-0351-7] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 09/21/2012] [Indexed: 02/06/2023]
Abstract
Chagas disease caused by Trypanosoma cruzi remains an important neglected tropical disease and a cause of significant morbidity and mortality. No longer confined to endemic areas of Latin America, it is now found in non-endemic areas due to immigration. The parasite may persist in any tissue, but in recent years, there has been increased recognition of adipose tissue both as an early target of infection and a reservoir of chronic infection. The major complications of this disease are cardiomyopathy and megasyndromes involving the gastrointestinal tract. The pathogenesis of Chagas disease is complex and multifactorial involving many interactive pathways. The significance of innate immunity, including the contributions of cytokines, chemokines, reactive oxygen species, and oxidative stress, has been emphasized. The role of the components of the eicosanoid pathway such as thromboxane A(2) and the lipoxins has been demonstrated to have profound effects as both pro- and anti-inflammatory factors. Additionally, we discuss the vasoconstrictive actions of thromboxane A(2) and endothelin-1 in Chagas disease. Human immunity to T. cruzi infection and its role in pathogen control and disease progression have not been fully investigated. However, recently, it was demonstrated that a reduction in the anti-inflammatory cytokine IL-10 was associated with clinically significant chronic chagasic cardiomyopathy.
Collapse
Affiliation(s)
- Fabiana S. Machado
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Program in Health Sciences: Infectious Diseases and Tropical Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walderez O. Dutra
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Departments of Microbiology and Immunology and Pathology, Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX
| | - Lisia Esper
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Program in Health Sciences: Infectious Diseases and Tropical Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Kenneth Gollob
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Santa Casa Hospital, Belo Horizonte, Brazil
- SRI International, Biosciences Division, Menlo Park, CA
- National Institute of Science and Technology in Tropical Diseases, Belo Horizonte, MG, Brazil
| | - Mauro M. Teixeira
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Program in Health Sciences: Infectious Diseases and Tropical Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Stephen M. Factor
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Fnu Nagajyothi
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
| | - Herbert B. Tanowitz
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Nisha J. Garg
- Departments of Microbiology and Immunology and Pathology, Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
28
|
Sánchez Valdéz FJ, Pérez Brandán C, Zago MP, Labriola C, Ferreira A, Basombrío MÁ. Trypanosoma cruzi carrying a monoallelic deletion of the calreticulin (TcCRT) gene are susceptible to complement mediated killing and defective in their metacyclogenesis. Mol Immunol 2012; 53:198-205. [PMID: 22954747 DOI: 10.1016/j.molimm.2012.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 06/25/2012] [Accepted: 08/07/2012] [Indexed: 11/17/2022]
Abstract
Trypanosoma cruzi calreticulin (TcCRT) can hijack complement C1, mannan-binding lectin and ficolins from serum thus inhibiting the classical and lectin complement pathway activation respectively. To understand the in vivo biological functions of TcCRT in T. cruzi we generated a clonal cell line lacking one TcCRT allele (TcCRT+/-) and another clone overexpressing it (TcCRT+). Both clones were derived from the TCC T. cruzi strain. As expected, TcCRT+/- epimastigotes showed impairment on TcCRT synthesis, whereas TcCRT+ ones showed increased protein levels. In correlation to this, monoallelic mutant parasites were significantly susceptible to killing by the complement machinery. On the contrary, TcCRT+ parasites showed higher levels of resistance to killing mediate by the classical and lectin but not the alternative pathway. The involvement of surface TcCRT in depleting C1 was demonstrated through restoration of serum killing activity by addition of exogenous C1. In axenic cultures, a reduced propagation rate of TcCRT+/- parasites was observed. Moreover, TcCRT+/- parasites presented a reduced rate of differentiation in in vitro assays. As shown by down- or upregulation of TcCRT expression this gene seems to play a major role in providing T. cruzi with the ability to resist complement system.
Collapse
|
29
|
Cestari I, Ansa-Addo E, Deolindo P, Inal JM, Ramirez MI. Trypanosoma cruzi immune evasion mediated by host cell-derived microvesicles. THE JOURNAL OF IMMUNOLOGY 2012; 188:1942-52. [PMID: 22262654 DOI: 10.4049/jimmunol.1102053] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The innate immune system is the first mechanism of vertebrate defense against pathogen infection. In this study, we present evidence for a novel immune evasion mechanism of Trypanosoma cruzi, mediated by host cell plasma membrane-derived vesicles. We found that T. cruzi metacyclic trypomastigotes induced microvesicle release from blood cells early in infection. Upon their release, microvesicles formed a complex on the T. cruzi surface with the complement C3 convertase, leading to its stabilization and inhibition, and ultimately resulting in increased parasite survival. Furthermore, we found that TGF-β-bearing microvesicles released from monocytes and lymphocytes promoted rapid cell invasion by T. cruzi, which also contributed to parasites escaping the complement attack. In addition, in vivo infection with T. cruzi showed a rapid increase of microvesicle levels in mouse plasma, and infection with exogenous microvesicles resulted in increased T. cruzi parasitemia. Altogether, these data support a role for microvesicles contributing to T. cruzi evasion of innate immunity.
Collapse
Affiliation(s)
- Igor Cestari
- Laboratório de Biologia Molecular de Parasitas e Vetores, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | | | | | | | | |
Collapse
|
30
|
Vázquez-Chagoyán JC, Gupta S, Garg NJ. Vaccine development against Trypanosoma cruzi and Chagas disease. ADVANCES IN PARASITOLOGY 2011; 75:121-46. [PMID: 21820554 DOI: 10.1016/b978-0-12-385863-4.00006-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathology of Chagas disease presents a complicated and diverse picture in humans. The major complications and destructive evolutionary outcomes of chronic infection by Trypanosoma cruzi in humans include ventricular fibrillation, thromboembolism and congestive heart failure. Studies in animal models and human patients have revealed the pathogenic mechanisms during disease progression, pathology of disease and features of protective immunity. Accordingly, several antigens, antigen-delivery vehicles and adjuvants have been tested to elicit immune protection to T. cruzi in experimental animals. This review summarizes the research efforts in vaccine development against Chagas disease during the past decade.
Collapse
Affiliation(s)
- Juan C Vázquez-Chagoyán
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Estado de México, Toluca, Mexico
| | | | | |
Collapse
|
31
|
Oladiran A, Belosevic M. Immune evasion strategies of trypanosomes: a review. J Parasitol 2011; 98:284-92. [PMID: 22007969 DOI: 10.1645/ge-2925.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Trypanosomes are digenetic protozoans that infect domestic and wild animals, as well as humans. They cause important medical and veterinary diseases, making them a major public health concern. There are many species of trypanosomes that infect virtually all vertebrate taxa. They typically cycle between insect or leech vectors and vertebrate hosts, and they undergo biochemical and morphological changes in the process. Trypanosomes have received much attention in the last 4 decades because of the diseases they cause and their remarkable armamentarium of immune evasion mechanisms. The completed genome sequences of trypanosomes have revealed an extensive array of molecules that contribute to various immune evasion mechanisms. The different species interact uniquely with their vertebrate hosts with a wide range of evasion strategies and some of the most fascinating immune evasion mechanisms, including antigenic variation that was first described in the trypanosomes. This review focuses on the variety of strategies that these parasites have evolved to evade or modulate immunity of endothermic and ectothermic vertebrates.
Collapse
Affiliation(s)
- Ayoola Oladiran
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
32
|
Pena DA, Eger I, Nogueira L, Heck N, Menin Á, Báfica A, Steindel M. Selection of TcII Trypanosoma cruzi population following macrophage infection. J Infect Dis 2011; 204:478-86. [PMID: 21742848 DOI: 10.1093/infdis/jir292] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Chagas disease is caused by the protozoan parasite Trypanosoma cruzi, which exhibits a high genetic variability. TcI, TcII, or mixed TcI/TcII strains may be found during acute human infection while mainly TcII parasites are present at the chronic stage of disease. In a previously studied Chagas disease outbreak, we identified mixed TcI/TcII strains in the vector Triatoma tibiamaculata and only TcII strains in infected humans, indicating that T. cruzi populations may be selected within the human host. METHODS Utilizing molecular typing and cell biology techniques, we investigated the interaction of TcI, TcII, and mixed TcI/TcII strains with macrophages, an important cell population implicated in controlling protozoan infection. RESULTS TcII but not TcI strains were selected by both human and murine macrophages in vitro and by peritoneal cavity cells in vivo. Biological analysis revealed that, compared with TcI, TcII strains display higher infective and multiplicative ability as well as lower doubling time inside macrophages. However, TcI and TcII strains present similar susceptibility to interferon-γ-activated macrophages in vitro. CONCLUSIONS Taken together, our results reveal the existence of an intracellular selection process in macrophages that favors TcII, but not TcI, when infection occurs with vector-derived mixed TcI/TcII strains.
Collapse
Affiliation(s)
- Darlene A Pena
- Laboratory of Protozoology, Department of Microbiology, Immunology, and Parasitology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | | | | | | | | | | | | |
Collapse
|
33
|
Gupta S, Garg NJ. Prophylactic efficacy of TcVac2 against Trypanosoma cruzi in mice. PLoS Negl Trop Dis 2010; 4:e797. [PMID: 20706586 PMCID: PMC2919396 DOI: 10.1371/journal.pntd.0000797] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 07/15/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chagas disease is a major health problem in Latin America, and an emerging infectious disease in the US. Previously, we have screened the Trypanosoma cruzi sequence database by a computational/bioinformatics approach, and identified antigens that exhibited the characteristics of vaccine candidates. METHODOLOGY We investigated the protective efficacy of a multi-component DNA-prime/protein-boost vaccine (TcVac2) constituted of the selected candidates and cytokine (IL-12 and GM-CSF) expression plasmids in a murine model. C57BL/6 mice were immunized with antigen-encoding plasmids plus cytokine adjuvants, followed by recombinant proteins; and two-weeks later, challenged with T. cruzi trypomastigotes. ELISA and flow cytometry were employed to measure humoral (antibody isotypes) and cellular (lymphocyte proliferation, CD4(+) and CD8(+) T cell phenotype and cytokines) responses. Myocardial pathology was evaluated by H&E and Masson's trichrome staining. PRINCIPAL FINDINGS TcVac2 induced a strong antigen-specific antibody response (IgG2b>IgG1) and a moderate level of lymphocyte proliferation in mice. Upon challenge infection, TcVac2-vaccinated mice expanded the IgG2b/IgG1 antibodies and elicited a substantial CD8(+) T cell response associated with type 1 cytokines (IFN-gamma and TNF-alpha) that resulted in control of acute parasite burden. During chronic phase, antibody response persisted, splenic activation of CD8(+) T cells and IFN-gamma/TNF-alpha cytokines subsided, and IL-4/IL-10 cytokines became dominant in vaccinated mice. The tissue parasitism, inflammation, and fibrosis in heart and skeletal muscle of TcVac2-vaccinated chronic mice were undetectable by histological techniques. In comparison, mice injected with vector or cytokines only responded to T. cruzi by elicitation of a mixed (type 1/type 2) antibody, T cell and cytokine response, and exhibited persistent parasite burden and immunopathology in the myocardium. CONCLUSION TcVac2-induced activation of type 1 antibody and lymphocyte responses provided resistance to acute T. cruzi infection, and consequently, prevented the evolution of chronic immunopathology associated with parasite persistence in chagasic hearts.
Collapse
Affiliation(s)
- Shivali Gupta
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nisha Jain Garg
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Member of the Institute for Infections and Immunity, Center for Biodefense and Emerging Infectious Diseases, and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
34
|
Antiangiogenic and antitumor effects of Trypanosoma cruzi Calreticulin. PLoS Negl Trop Dis 2010; 4:e730. [PMID: 20625551 PMCID: PMC2897838 DOI: 10.1371/journal.pntd.0000730] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 05/11/2010] [Indexed: 01/26/2023] Open
Abstract
Background In Latin America, 18 million people are infected with Trypanosoma cruzi, the agent of Chagas' disease, with the greatest economic burden. Vertebrate calreticulins (CRT) are multifunctional, intra- and extracellular proteins. In the endoplasmic reticulum (ER) they bind calcium and act as chaperones. Since human CRT (HuCRT) is antiangiogenic and suppresses tumor growth, the presence of these functions in the parasite orthologue may have consequences in the host/parasite interaction. Previously, we have cloned and expressed T. cruzi calreticulin (TcCRT) and shown that TcCRT, translocated from the ER to the area of trypomastigote flagellum emergence, promotes infectivity, inactivates the complement system and inhibits angiogenesis in the chorioallantoid chicken egg membrane. Most likely, derived from these properties, TcCRT displays in vivo inhibitory effects against an experimental mammary tumor. Methodology and Principal Findings TcCRT (or its N-terminal vasostatin-like domain, N-TcCRT) a) Abrogates capillary growth in the ex vivo rat aortic ring assay, b) Inhibits capillary morphogenesis in a human umbilical vein endothelial cell (HUVEC) assay, c) Inhibits migration and proliferation of HUVECs and the human endothelial cell line Eahy926. In these assays TcCRT was more effective, in molar terms, than HuCRT: d) In confocal microscopy, live HUVECs and EAhy926 cells, are recognized by FITC-TcCRT, followed by its internalization and accumulation around the host cell nuclei, a phenomenon that is abrogated by Fucoidin, a specific scavenger receptor ligand and, e) Inhibits in vivo the growth of the murine mammary TA3 MTXR tumor cell line. Conclusions/Significance We describe herein antiangiogenic and antitumor properties of a parasite chaperone molecule, specifically TcCRT. Perhaps, by virtue of its capacity to inhibit angiogenesis (and the complement system), TcCRT is anti-inflammatory, thus impairing the antiparasite immune response. The TcCRT antiangiogenic effect could also explain, at least partially, the in vivo antitumor effects reported herein and the reports proposing antitumor properties for T. cruzi infection. In Latin America, 18 million people are infected with Trypanosoma cruzi, a protozoan that causes Chagas' disease. Vertebrate calreticulins (CRTs) are multifunctional, intra- and extracellular calcium binding, chaperone proteins. Since human CRT (HuCRT) inhibits capillary growth (angiogenesis) and suppresses tumor growth, the presence of these functions in T. cruzi CRT (TcCRT) may have interesting consequences in the host/parasite interactions. Previously, we have cloned and expressed TcCRT and shown that, when translocated from the endoplasmic reticulum to the area of trypomastigote flagellum emergence, it promotes infectivity, inactivates the complement system, an innate defense arm and inhibits angiogenesis in the chorioallantoid chicken egg membrane. TcCRT inhibits angiogenesis, since it interferes with endothelial cell multiplication, migration and capillary morphogenesis in vitro, as well as angiogenesis in rat aortic rings. The parasite molecule also displays important antitumor effects. In these activities, TcCRT is more effective than the human counterpart. Perhaps, by virtue of its capacity to inhibit angiogenesis, TcCRT is anti-inflammatory, thus impairing the antiparasite immune response. The TcCRT antiangiogenic effect could also explain, at least partially, the in vivo antitumor effects reported herein and the reports proposing antitumor properties for T. cruzi infection.
Collapse
|
35
|
Oladiran A, Belosevic M. Trypanosoma carassii calreticulin binds host complement component C1q and inhibits classical complement pathway-mediated lysis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2010; 34:396-405. [PMID: 19913050 DOI: 10.1016/j.dci.2009.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 11/04/2009] [Accepted: 11/05/2009] [Indexed: 05/28/2023]
Abstract
Trypanosoma carassii is an extracellular parasite of economically important fish species that has evolved several strategies to circumvent host immune responses. Proteomic analysis of the excreted/secreted (ES) and surface molecules of the parasite has revealed a number of proteins that may be involved in host-parasite interactions. Among the parasite molecules identified in the ES of T. carassii was calreticulin. We cloned and produced T. carassii calreticulin (rTcaCRT), and generated a rabbit polyclonal antibody to the recombinant protein. The incubation of parasites with rabbit anti-rTcaCRT affinity-purified IgG antibody indicated substantial CRT levels on the surface of trypanosomes, as well as internal structures of permeabilized organisms. Recombinant parasite calreticulin bound several molecules in host serum including the first complement component, C1q. The host C1q specifically interacted with parasite CRT since the C1q-dependent lysis of sensitized sheep erythrocytes was inhibited by rTcaCRT. Our findings suggest that CRT may be used by the parasite to inhibit hosts' classical complement pathway.
Collapse
Affiliation(s)
- Ayoola Oladiran
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
36
|
Cestari IDS, Krarup A, Sim RB, Inal JM, Ramirez MI. Role of early lectin pathway activation in the complement-mediated killing of Trypanosoma cruzi. Mol Immunol 2009; 47:426-37. [PMID: 19783051 DOI: 10.1016/j.molimm.2009.08.030] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 08/21/2009] [Accepted: 08/28/2009] [Indexed: 11/15/2022]
Abstract
The complement system is the first line of defence against pathogen infection and can be activated by the classic, alternative and lectin pathways. Trypanosoma cruzi, the causative agent of Chagas disease, has to evade complement system killing and invade the host cells to progress in infection. T. cruzi infectious stages resist complement-mediated killing by expressing surface receptors, which dissociate or prevent C3 convertase formation. Here, we present the first evidence that T. cruzi activates the complement lectin pathway. We detected rapid binding of mannan-binding lectin, H-ficolin, and L-ficolin to the surface of T. cruzi, and found that serum depleted of these molecules failed to kill parasites. Furthermore, lectin pathway activation by T. cruzi required the MBL-associated serine protease 2 (MASP2) activity resulting in C2 factor cleavage. In addition, we demonstrate that the infectious stage of T. cruzi inhibits the lectin pathway activation and complement killing expressing the complement C2 receptor inhibitor trispanning (CRIT) protein. Transgenic parasites overexpressing CRIT were highly resistant to complement-mediated killing. CRIT-derived peptides inhibited both C2 binding to the surface of T. cruzi and parasite killing. Biochemical studies revealed that the CRIT extracellular domain 1 inhibits MASP2 cleavage of C2 factor and thereby impairs C3 convertase formation. Our findings establish that the complement lectin pathway recognizes T. cruzi and provide molecular insights into how the infectious stage inhibits this activation to resist complement system killing.
Collapse
Affiliation(s)
- Igor dos S Cestari
- Instituto Oswaldo Cruz-Fiocruz, Laboratório de Biologia Molecular de Parasitas e Vetores, Rio de Janeiro, 21040-900, Brazil
| | | | | | | | | |
Collapse
|
37
|
Krettli AU. The utility of anti-trypomastigote lytic antibodies for determining cure of Trypanosoma cruzi infections in treated patients: an overview and perspectives. Mem Inst Oswaldo Cruz 2009; 104 Suppl 1:142-51. [DOI: 10.1590/s0074-02762009000900020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Accepted: 06/09/2009] [Indexed: 11/22/2022] Open
|
38
|
Reduction of parasite levels in blood improves pregnancy outcome during experimental Trypanosoma cruzi infection. Parasitology 2009; 136:627-39. [PMID: 19366478 DOI: 10.1017/s0031182009005770] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Infection with a myotropic Trypanosoma cruzi clone induces maternal fertility failure. In the current work, we evaluated whether reduction of maternal parasitaemia before mating has beneficial effects on pregnancy outcome. Female mice were subjected to benznidazole (Bz) treatment after infection. On day 30 of therapy, mating was assessed and pregnancy outcome was determined on day 14 of gestation. Fetal resorptions diminished in T. cruzi-infected Bz-treated mice compared with T. cruzi-infected untreated mice. This was in agreement with the reduction in the blood/solid tissue parasite load and with the percentage of necrotic foci in placental samples from T. cruzi-infected Bz-treated females. To study eventual changes in the immune homeostasis of T. cruzi-infected Bz-treated mice, activation of the immune system was evaluated at the end of Bz therapy and before mating. We found specific IgG1 reduction resulting in a predominance of specific IgG2a, reduced numbers of CD69+ CD4+ cells and diminished frequency and numbers of CD44+ T cells. Concanavalin A-stimulated splenocytes from T. cruzi-infected Bz-treated mice produced higher amounts of IFN-gamma than T. cruzi-infected untreated mice. These results indicate that reduction of maternal parasite load improves pregnancy outcome. These findings correlate with a favourable modulation of the immune response.
Collapse
|
39
|
Cestari IDS, Evans-Osses I, Freitas JC, Inal JM, Ramirez MI. Complement C2 receptor inhibitor trispanning confers an increased ability to resist complement-mediated lysis in Trypanosoma cruzi. J Infect Dis 2008; 198:1276-83. [PMID: 18781865 DOI: 10.1086/592167] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The ability to resist complement differs between the Y and Colombiana Trypanosoma cruzi strains. We found that the Y strain of T. cruzi was more able to resist the classical and lectin pathways of complement activation than the Colombiana strain. The complement C2 receptor inhibitor trispanning gene (CRIT) is highly conserved in both strains. At the protein level, CRIT is expressed only in stationary-phase epimastigotes of the Y but not the Colombiana strain and is expressed in infectious metacyclic trypomastigotes of both strains. Y strain epimastigotes with an overexpressed CRIT gene (pTEX-CRIT) had higher survival in normal human serum (NHS). Overexpression of the Y strain CRIT gene in Colombiana epimastigote forms increased the parasite's resistance to lysis mediated by the classical and lectin pathways but not to lysis mediated by alternative pathways. CRIT involvement on the parasite surface was confirmed by showing that the lytic activity of NHS against epimastigotes could be restored by adding excess C2.
Collapse
Affiliation(s)
- Igor Dos S Cestari
- Departamento de Bioquímica e Biologia Molecular, Instituto Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
40
|
León-Pérez F, Gómez-Garcia L, Alejandre-Aguilar R, López R, Monteón VM. Mexican Trypanosoma cruzi isolates: in vitro susceptibility of epimastigotes to anti-trypanosoma cruzi drugs and metacyclic forms to complement-mediated lysis. Vector Borne Zoonotic Dis 2008; 7:330-6. [PMID: 17760512 DOI: 10.1089/vbz.2006.0604] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Trypanosoma cruzi has a clonal organization with an ample array of genetic and phenotypic features and probably anaploid constitution. Consequently, the biological behavior, biochemistry, and molecular attributes may be distinctive for each parasite strain in different geographical regions. As far as we know, there is no published information on the susceptibility of Mexican T. cruzi stocks to anti-T. cruzi drugs such as benznidazole and gentian violet, or on its resistance to complement-mediated lysis. We studied 10 Mexican T. cruzi isolates from different geographical areas, such as the pacific coast (Oaxaca, Guerrero, and Nayarit States), central part of Mexico (Guanajuato State), Gulf of Mexico (Veracruz State), and the Yucatan Peninsula (Campeche State). We searched for the natural resistance to drugs in in vitro assay against the 10 Mexican isolates using epimastigote forms and the complement-mediated lysis using metacyclic trypomastigotes insect-derived in three of them (one for each geographic region). In general, we observed high resistance to benznidazole in all the Mexican isolates tested, but in the complement-mediated lysis test, they showed moderate to high susceptibility. Although it is necessary to expand this study by using trypomastigotes and the intracellular form to verify its biological role, we suggest that Mexican T. cruzi parasites may have a variable susceptibility to antibody-mediated lysis and high resistance to benznidazole.
Collapse
Affiliation(s)
- Floribeth León-Pérez
- Centro Investigación Enfermedades Tropicales, Universidad Autónoma de Campeche, Campeche, Mexico
| | | | | | | | | |
Collapse
|
41
|
Oral vaccination with Salmonella enterica as a cruzipain-DNA delivery system confers protective immunity against Trypanosoma cruzi. Infect Immun 2007; 76:324-33. [PMID: 17967857 DOI: 10.1128/iai.01163-07] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
To stimulate both local and systemic immune responses against Trypanosoma cruzi, Salmonella enterica serovar Typhimurium aroA was exploited as a DNA delivery system for cruzipain (SCz). In a murine model we compared SCz alone (GI) or coadministered with Salmonella carrying a plasmid encoding granulocyte-macrophage colony-stimulating factor (GII), as well as protocols in which SCz priming was followed by boosting with recombinant cruzipain (rCz) admixed with either CpG-ODN (GIII) or MALP-2, a synthetic derivative of a macrophage-activating lipopeptide of 2 kDa from Mycoplasma fermentans (GIV). The results showed that protocols that included four oral doses of SCz (GI) elicited mainly a mucosal response characterized by immunoglobulin A (IgA) secretion and proliferation of gut-associated lymphoid tissue cells, with weak systemic responses. In contrast, the protocol that included a boost with rCz plus CpG (GIII) triggered stronger systemic responses in terms of Cz-specific serum IgG titers, splenocyte proliferation, gamma interferon (IFN-gamma) secretion, and delayed-type hypersensitivity response. Trypomastigote challenge of vaccinated mice resulted in significantly lower levels of parasitemia compared to controls. Protection was abolished by depletion of either CD4+ or CD8+ T cells. Parasite control was also evident from the reduction of tissue damage, as revealed by histopathologic studies and serum levels of enzymes that are markers of muscle injury in chronic Chagas' disease (i.e., creatine kinase, aspartate aminotransferase, and lactate dehydrogenase). Enhanced release of IFN-gamma and interleukin-2 was observed in GI and GII upon restimulation of splenocytes in the nonparasitic phase of infection. Our results indicate that Salmonella-mediated delivery of Cz-DNA by itself promotes the elicitation of an immune response that controls T. cruzi infection, thereby reducing parasite loads and subsequent damage to muscle tissues.
Collapse
|
42
|
Garg N, Bhatia V. Current status and future prospects for a vaccine against American trypanosomiasis. Expert Rev Vaccines 2007; 4:867-80. [PMID: 16372882 DOI: 10.1586/14760584.4.6.867] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The clinically relevant pathognomonic consequences of human infection by Trypanosoma cruzi are dilation and hypertrophy of the left ventricle walls and thinning of the apex. The major complications and debilitating evolutionary outcomes of chronic infection include ventricular fibrillation, thromboembolism and congestive heart failure. American trypanosomiasis (Chagas disease) poses serious public healthcare and budgetary concerns. The currently available drugs, although effective against acute infection, are highly toxic and ineffective in arresting or attenuating clinical disease symptoms in chronic patients. The development of an efficacious prophylactic vaccine faces many challenges, and progress is slow, despite several years of effort. Studies in animal models and human patients have revealed the pathogenic mechanisms during disease progression, pathology of disease and features of protective immunity. Accordingly, several antigens, antigen-delivery vehicles and adjuvants have been tested in animal models, and some efforts have been successful in controlling infection and disease. This review will summarize the accumulated knowledge about the parasite and disease, as well as pathogenesis and protective immunity. The authors will discuss the efforts to date, and the challenges faced in achieving an efficient prophylactic vaccine against human American trypanosomiasis, and present the future perspectives.
Collapse
Affiliation(s)
- Nisha Garg
- Sealy Center for Vaccine Development, Department of Microbiology, Immunology and Pathology, University of Texas Medical Branch, Galveston TX 77555, USA.
| | | |
Collapse
|
43
|
Chamekh M, Vercruysse V, Habib M, Lorent M, Goldman M, Allaoui A, Vray B. Transfection of Trypanosoma cruzi with host CD40 ligand results in improved control of parasite infection. Infect Immun 2005; 73:6552-61. [PMID: 16177330 PMCID: PMC1230987 DOI: 10.1128/iai.73.10.6552-6561.2005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2005] [Revised: 03/09/2005] [Accepted: 07/04/2005] [Indexed: 11/20/2022] Open
Abstract
We have previously shown that infection by Trypanosoma cruzi, a parasitic protozoan, is reduced by injection of CD40 ligand (CD40L)-transfected 3T3 fibroblasts (D. Chaussabel, F. Jacobs, J. de Jonge, M. de Veerman, Y. Carlier, K. Thielemans, M. Goldman, and B. Vray, Infect. Immun. 67:1929-1934, 1999). This prompted us to transfect T. cruzi with the murine CD40L gene and to study the consequences of this transfection on the course of infection. For this, epimastigotes (Y strain) were electroporated with the pTEX vector alone or the pTEX-CD40L construct, and transfected cells were selected for their resistance to Geneticin G418. Then strain Y-, pTEX-, and pTEX-CD40L-transfected epimastigotes were transformed by metacyclogenesis into mammalian infective forms called Y, YpTEX, and YpTEX-CD40L trypomastigotes. Transfection of the CD40L gene and expression of the CD40L protein were assessed by reverse transcription-PCR and Western blot analysis. The three strains of parasites were infective in vitro for mouse peritoneal macrophages. When organisms were inoculated into mice, a very low level of parasitemia and no mortality were seen with the YpTEX-CD40L strain compared to the Y and YpTEX strains. Furthermore, the proliferative capacity and the secretion of gamma interferon were both preserved in spleen cells (SCs) from YpTEX-CD40L-infected mice but not with SCs from Y- and YpTEX-infected mice. These results suggest that the CD40L produced by transfected T. cruzi is involved in the modulation of an antiparasite immune response. Moreover, mice surviving YpTEX-CD40L infection resisted a challenge infection with the wild-type strain. Taken together, our data demonstrate the feasibility of generating a T. cruzi strain expressing a bioactive host costimulatory molecule that counteracts the immunodeficiency induced by the parasite during infection and enhances protective immunity against a challenge infection.
Collapse
Affiliation(s)
- Mustapha Chamekh
- Laboratoire de Bactériologie Moléculaire, Faculté de Médecine, Université Libre de Bruxelles, Belgium
| | | | | | | | | | | | | |
Collapse
|
44
|
Ferreira V, Molina MC, Schwaeble W, Lemus D, Ferreira A. Does Trypanosoma cruzi calreticulin modulate the complement system and angiogenesis? Trends Parasitol 2005; 21:169-74. [PMID: 15780838 DOI: 10.1016/j.pt.2005.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Calreticulin, a calcium-binding protein that is highly conserved in its multiple functions, is present in a wide spectrum of subcellular compartments in virtually every cell of higher organisms. In this article, we propose a dual role for parasite calreticulin, with emphasis on the Trypanosoma cruzi model. By modulating the vertebrate complement system, calreticulin might provide the parasite with an effective immune-escape mechanism. Alternatively, by inhibiting angiogenesis, the parasite molecule might protect the host from ongoing neoplasic aggressions. Many questions are still unanswered, particularly those regarding the consequences that these interactions could have in vivo for both the parasite and the host.
Collapse
Affiliation(s)
- Viviana Ferreira
- Programa de Inmunología, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile.
| | | | | | | | | |
Collapse
|
45
|
Garzon E, Genna F, Bosseno MF, Simony-La Fontaine J, Radal M, Sereno D, Mathieu-Daude F, Ouaissi A, Brenière SF. Differential infectivity and immunopathology in murine experimental infections by two natural clones belonging to theTrypanosoma cruziI lineage. Parasitology 2005; 131:109-19. [PMID: 16038402 DOI: 10.1017/s003118200400722x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immunopathology of Chagas' disease in Balb/c mice infected with 2Trypanosoma cruziclones, belonging to theT. cruziI lineage and presenting differentin vitrovirulence (P/209 cl1>SO34 cl4) was compared. In the acute phase, evading mechanisms such as parasite-induced lymphocyte polyclonal activation and T cell immunosuppression were higher in mice infected with the clone giving a higher parasitaemia (P/209 cl1). A similar increase of non-specific isotypes was observed in both infections with IgG2a prevalence. Interestingly, CD8+ cell hypercellularity and lymphocyte immunosuppression were observed during the chronic phase (245 days post-infection) in mice infected by the most virulent clone. In the same way, the parasite-specific antibody response was more intense in P/209 cl1-infected mice over the acute phase. During the chronic phase this response remarkably dropped down in SO34 cl4-infected mice exclusively. Finally, P/209 cl1-infected mice presented a more severe inflammation and tissue damage in heart and quadriceps than SO34 cl4-infected mice. This comparative study showed differences between the two clones: a higher virulencein vivobeing clearly associated with a greater ability to induce evasion mechanisms and severe tissue damage.
Collapse
Affiliation(s)
- E Garzon
- Institut de Recherche pour le Développement, IRD, UR 008 Pathogénie et Epidémiologie des Trypanosomatidés, 911 Av. Agropolis, BP 64501, 34394 Montpellier Cedex 5, France
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ferreira V, Valck C, Sánchez G, Gingras A, Tzima S, Molina MC, Sim R, Schwaeble W, Ferreira A. The classical activation pathway of the human complement system is specifically inhibited by calreticulin from Trypanosoma cruzi. THE JOURNAL OF IMMUNOLOGY 2004; 172:3042-50. [PMID: 14978109 DOI: 10.4049/jimmunol.172.5.3042] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The high resistance of Trypanosoma cruzi trypomastigotes, the causal agent of Chagas' disease, to complement involves several parasite strategies. In these in vitro studies, we show that T. cruzi calreticulin (TcCRT) and two subfragments thereof (TcCRT S and TcCRT R domains) bind specifically to recognition subcomponents of the classical and lectin activation pathways (i.e., to collagenous tails of C1q and to mannan-binding lectin) of the human complement system. As a consequence of this binding, specific functional inhibition of the classical pathway and impaired mannan-binding lectin to mannose were observed. By flow cytometry, TcCRT was detected on the surface of viable trypomastigotes and, by confocal microscopy, colocalization of human C1q with surface TcCRT of infective trypomastigotes was visualized. Taken together, these findings imply that TcCRT may be a critical factor contributing to the ability of trypomastigotes to interfere at the earliest stages of complement activation.
Collapse
Affiliation(s)
- Viviana Ferreira
- Immunology and Molecular Biology Disciplinary Programs, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ferreira V, Molina MC, Valck C, Rojas A, Aguilar L, Ramírez G, Schwaeble W, Ferreira A. Role of calreticulin from parasites in its interaction with vertebrate hosts. Mol Immunol 2004; 40:1279-91. [PMID: 15128045 DOI: 10.1016/j.molimm.2003.11.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although parasites range from protozoan to complex, evolutionary advanced arthropods, in general, a hallmark of parasite life cycles is their ability to adapt to changes in temperature, pH and host defense strategies. Calreticulin, a calcium-binding protein, highly conserved and multifunctional, is present in every cell of higher organisms, except erythrocytes. The surprising array of calreticulin-associated functions include lectin-like chaperoning, calcium storage and signaling, modulation of gene expression, cell adhesion, enhancement of phagocytosis of C1q or collectin opsonized apoptotic cells, inhibition of angiogenesis and tumoral growth, inhibition of perforin pore formation in T and NK cells, and inhibition of C1q-dependent complement activation. Likewise, calreticulin is present in a wide spectrum of sub cellular compartments. Parasite calreticulin shows a surprisingly high degree of conservation within the framework of its functional domains. Its role within the parasite/host relationship needs to be assessed further, in particular with regard to its impact on parasite infectivity, by helping to evade from its hosts' immune response. With special emphasis on calreticulin from Trypanosoma cruzi, the intracellular protozoan agent of American trypanosomiasis (Chagas' disease), we wish to exemplify and highlight the various implications of parasite calreticulin, within the pathophysiology of parasite-mediated human and animal disease.
Collapse
Affiliation(s)
- Viviana Ferreira
- Programa de Immunología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Shichinohe K, Shimizu-Suganuma M, Ghazizadeh M, Ishizaki M. Supplementation of heterologous complement induces anti-Thy-1.1 nephritis in the Mongolian gerbil (Meriones unguiculatus). J Vet Med Sci 2002; 64:463-7. [PMID: 12130828 DOI: 10.1292/jvms.64.463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anti-Thy-1.1 nephritis in the rat is a popular experimental model for mesangial proliferative glomerulonephritis (GN). This model is characterized by direct binding of anti-Thy-1.1 antibody with Thy-1.1 antigen expressed on mesangial cells (MCs) of glomeruli in the rat. A single injection of anti-rat thymocyte serum (ARTS) results in GN with proteinuria and extensive mesangiolysis. Development of mesangiolysis and proteinuria are complement-dependent. We previously demonstrated Thy-1.1 antigen, similar to the rat, in thymocytes, brain cells and MCs of the kidney in the Mongolian gerbil (MG). In this study, we attempted to develop a MG nephritis model, but an injection of ARTS did not induce GN. An additional injection of guinea pig serum as a complement after ARTS injection resulted in anti-Thy-1.1 nephritis in MG. Degeneration of MCs and neutrophil infiltration were observed 1 hr after GP serum injection. Mesangiolysis and fibrin exudation occurred 12 hr after the injection and MC proliferation was apparent 7 days after the injection. In the complement-dependent hemolytic test, MG serum could not hemolyze sheep erythrocytes. These results suggested low activity, or depletion of some factors, in complements of MG serum.
Collapse
Affiliation(s)
- Kazuhiro Shichinohe
- Department of Laboratory Animal Science, Nippon Medical School, Tokyo, Japan
| | | | | | | |
Collapse
|
49
|
Matsumoto TK, Cotrim PC, da Silveira JF, Stolf AMS, Umezawa ES. Trypanosoma cruzi: isolation of an immunodominant peptide of TESA (Trypomastigote Excreted-Secreted Antigens) by gene cloning. Diagn Microbiol Infect Dis 2002; 42:187-92. [PMID: 11929690 DOI: 10.1016/s0732-8893(01)00348-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Trypomastigote forms of Trypanosoma cruzi excrete-secrete several molecules, which are immunodominant during the human infection. This complex antigenic mixture termed TESA (Trypomastigote Excreted-Secreted Antigens) presents a 150-160 kDa band that shows excellent specificity and sensitivity in Chagas' disease diagnosis by immunoblotting. Here we describe the isolation and the antigenic characterization of a recombinant peptide (TESA-1) containing a 10 kDa T. cruzi peptide that belongs to the 150-160 kDa TESA fraction. The clone was isolated by screening a T. cruzi genomic expression library with chagasic antibodies reactive to the 150-160 kDa band of TESA immunoblots. After expression, the recombinant peptide TESA-1 was purified and used to immunize rabbits. Anti-TESA-1 immunesera specifically recognized the 150-160 kDa fraction of TESA-blots from eight different T. cruzi strains. The TESA-1 peptide reacted with 82.2% of chagasic patient sera by immunoblotting, showing that it harbors most of the antigenic epitopes that account for the high reactivity of the 150-160 kDa band of TESA.
Collapse
|