1
|
Telleria J, Costales JA. An Overview of Trypanosoma cruzi Biology Through the Lens of Proteomics: A Review. Pathogens 2025; 14:337. [PMID: 40333120 PMCID: PMC12030004 DOI: 10.3390/pathogens14040337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 05/09/2025] Open
Abstract
The protozoan parasite Trypanosoma cruzi, causative agent of Chagas disease, affects millions of people in endemic Latin American countries and beyond. In Latin America, Chagas disease is an important cause of death and disability, for which vaccines are lacking and improved treatment options are required. Additionally, the factors governing the development of a variety of clinical manifestations during Chagas disease, ranging from complete lack of symptoms to severe irreversible chronic organ damage (mainly cardiac or digestive), remain largely unknown. Much remains to be learned regarding the biology of T. cruzi in order to enhance our understanding of these lines of inquiry. In this context, proteomic methods have been leveraged to investigate different parasite strains, life-cycle forms, subcellular compartments, macromolecular complexes, signaling events and secreted molecules. The factors driving morphological transformation during the life cycle, the composition and functions of the parasite's organelles and secreted molecules as well as the determinants of pathogenicity have been explored via proteomic methods, yielding insights into the fundamental processes behind the parasite biology and informing drug design and vaccine development. Importantly, the correlation between the wide genetic and phenotypic variability displayed by T. cruzi has been examined through proteomic methods as well. Here, we review the literature on T. cruzi proteomics and discuss it in the light of its limitations and in the context of the parasite's genetic diversity.
Collapse
Affiliation(s)
- Jenny Telleria
- Institut de Recherche Pour le Développement (IRD), UMR Intertryp IRD-CIRAD, 34398 Montpellier, Cedex 5, France;
| | - Jaime A. Costales
- Centro de Investigación Para la Salud en América Latina, Pontificia Universidad Católica del Ecuador, Quito 170525, Ecuador
| |
Collapse
|
2
|
Henao MA, Cortes I, Isaza JP. In Silico Discovery of Antigenic-Secreted Proteins to Diagnostic Human Toxocariasis. Acta Parasitol 2025; 70:54. [PMID: 39918631 PMCID: PMC11805878 DOI: 10.1007/s11686-024-00966-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/11/2024] [Indexed: 02/09/2025]
Abstract
BACKGROUND Human toxocariasis is a helminthic zoonosis caused by infection of Toxocara canis or T. cati. Humans can be infected by through ingestion of embryonated eggs from contaminated water, food or soil. Diagnosis is challenging, immunodiagnosis tests are commonly implemented with major pitfalls in the cross-reactivity with other pathogens, particularly in endemic areas. METHODS With the aim of identify species-specific genes encoding for highly expressed antigenic proteins, a list of parasites that may infect humans and that might present similar clinical symptoms to T. canis infections was built. Only organisms whose genomes were completely sequenced and the proteome predicted were included. First, orthologous proteins were detected and the subcellular localization of T. canis proteins was predicted. In order to identify differentially expressed genes encoding proteins in larvae L3, pair-wise comparisons among transcriptomes from body parts and genders were performed. Finally, all secreted proteins classified as species-specific of T. canis, whose genes were upregulated in larvae L3 were included in an antigenic prediction. RESULTS Twenty-eight parasites were included in the analyses, proteins of T. canis were clustered in 11,399 groups, however, 279 were species-specific groups which represent 816 proteins. Three hundred and twenty-two proteins were predicted to be secreted and upregulated in larvae L3, however, after filtering these proteins by their orthology inference, only three proteins met all the features included in this study (species-specific, upregulated, secreted, and antigenic potential). To conclude, our strategy in the study is a rational approach for discovering antigenic proteins to be used in diagnosis.
Collapse
Affiliation(s)
- María A Henao
- Facultad de Medicina, Grupo Biología de Sistema, Universidad Pontificia Bolivariana, Circular 1a 70-01, Build 11C - 417, Medellín, Colombia
| | - Isabella Cortes
- Facultad de Medicina, Grupo Biología de Sistema, Universidad Pontificia Bolivariana, Circular 1a 70-01, Build 11C - 417, Medellín, Colombia
| | - Juan P Isaza
- Facultad de Medicina, Grupo Biología de Sistema, Universidad Pontificia Bolivariana, Circular 1a 70-01, Build 11C - 417, Medellín, Colombia.
| |
Collapse
|
3
|
Serrano-Rodríguez M, Araya JE, Cortez M, Orrego PR. Cytotoxic Effect of Trypanosoma cruzi Calcineurin B Against Melanoma and Adenocarcinoma Cells In Vitro. Adv Pharmacol Pharm Sci 2024; 2024:5394494. [PMID: 39640496 PMCID: PMC11620811 DOI: 10.1155/adpp/5394494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/30/2024] [Accepted: 10/26/2024] [Indexed: 12/07/2024] Open
Abstract
Chagas disease caused by the obligate intracellular flagellate protozoan Trypanozoma cruzi infects about 6 million people. From the 1930s to the present, the antitumor capacity of T. cruzi has been studied; however, the identification of the responsible molecules for this effect remains undiscovered. Calcineurin, a calcium/calmodulin-dependent serine/threonine phosphatase, is a heterodimer consisting of a catalytic subunit (CaNA) and a regulatory subunit (CaNB). It has been described that T. cruzi CaN is involved in the cell invasion and proliferation of the parasite. Recently, extracellular human CaNB has been demonstrated to be capable of inhibiting tumor growth cells, conferring an antitumor effect; however, the extracellular role of T. cruzi CaNB (TcCaNB) is still unknown. The objective of this work was to investigate the antitumor potential of TcCaNB by interacting with membrane proteins and evaluating its effects on the viability, proliferation, and morphology of tumor cells in vitro. Additionally, the possible mechanism of action of TcCaNB was explored. Murine melanoma (B16-F10), human cervical adenocarcinoma (HeLa), and African green monkey kidney epithelial (Vero) cell lines were employed for in vitro assays. Far Western blot and immunofluorescence were performed to assess the interaction of TcCaNB with membrane proteins, and the effect of TcCaNB on cell viability and proliferation was evaluated using the MTS assay and the CyQUANT NF assay, respectively. The effect of the caspase inhibitor Z-VAD-FMK on TcCaNB-stimulated tumor cells was investigated to determine if TcCaNB-induced cell death was associated with apoptosis. To assess cell cycle progression, TcCaNB-treated cells were analyzed by flow cytometry. In this study, the results showed an interaction of TcCaNB with cell membrane proteins in B16-F10 and HeLa tumor lines, indicating that TcCaNB is capable of decreasing viability and proliferation of B16-F10 and HeLa cells, with no significant effect observed in Vero cells. Furthermore, morphological changes were observed in tumor cells treated with TcCaNB. DNA fragmentations and inhibition of caspases with Z-VAD-FMK partially counteracted the cytotoxic effects of TcCaNB on tumor cells, suggesting that TcCaNB-induced cell death might be associated with apoptosis. Additionally, TcCaNB caused S phase cell cycle arrest in HeLa cells, with an increase in the sub-G1 population indicative of apoptosis, while no significant effects were observed in Vero cells.
Collapse
Affiliation(s)
- Mayela Serrano-Rodríguez
- Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta 1240000, Chile
| | - Jorge E. Araya
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta 1240000, Chile
| | - Mauro Cortez
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Patricio R. Orrego
- Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta 1240000, Chile
| |
Collapse
|
4
|
Silva TA, Thomas D, Siqueira-Neto JL, Calvet CM. Pirfenidone Prevents Heart Fibrosis during Chronic Chagas Disease Cardiomyopathy. Int J Mol Sci 2024; 25:7302. [PMID: 39000409 PMCID: PMC11242150 DOI: 10.3390/ijms25137302] [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: 11/18/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 07/16/2024] Open
Abstract
Cardiac fibrosis is a severe outcome of Chagas disease (CD), caused by the protozoan Trypanosoma cruzi. Clinical evidence revealed a correlation between fibrosis levels with impaired cardiac performance in CD patients. Therefore, we sought to analyze the effect of inhibitors of TGF-β (pirfenidone), p38-MAPK (losmapimod) and c-Jun (SP600125) on the modulation of collagen deposition in cardiac fibroblasts (CF) and in vivo models of T. cruzi chronic infection. Sirius Red/Fast Green dye was used to quantify both collagen expression and total protein amount, assessing cytotoxicity. The compounds were also used to treat C57/Bl6 mice chronically infected with T. cruzi, Brazil strain. We identified an anti-fibrotic effect in vitro for pirfenidone (TGF-β inhibitor, IC50 114.3 μM), losmapimod (p38 inhibitor, IC50 17.6 μM) and SP600125 (c-Jun inhibitor, IC50 3.9 μM). This effect was independent of CF proliferation since these compounds do not affect T. cruzi-induced host cell multiplication as measured by BrdU incorporation. Assays of chronic infection of mice with T. cruzi have shown a reduction in heart collagen by pirfenidone. These results propose a novel approach to fibrosis therapy in CD, with the prospect of repurposing pirfenidone to prevent the onset of ECM accumulation in the hearts of the patients.
Collapse
Affiliation(s)
- Tatiana Araújo Silva
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute (IOC), FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil;
| | - Diane Thomas
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.T.); (J.L.S.-N.)
| | - Jair L. Siqueira-Neto
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.T.); (J.L.S.-N.)
| | - Claudia Magalhaes Calvet
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute (IOC), FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil;
| |
Collapse
|
5
|
Ossowski MS, Gallardo JP, Niborski LL, Rodríguez-Durán J, Lapadula WJ, Juri Ayub M, Chadi R, Hernandez Y, Fernandez ML, Potenza M, Gómez KA. Characterization of Novel Trypanosoma cruzi-Specific Antigen with Potential Use in the Diagnosis of Chagas Disease. Int J Mol Sci 2024; 25:1202. [PMID: 38256275 PMCID: PMC10816184 DOI: 10.3390/ijms25021202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Chagas disease is caused by the parasite Trypanosoma cruzi. In humans, it evolves into a chronic disease, eventually resulting in cardiac, digestive, and/or neurological disorders. In the present study, we characterized a novel T. cruzi antigen named Tc323 (TcCLB.504087.20), recognized by a single-chain monoclonal antibody (scFv 6B6) isolated from the B cells of patients with cardiomyopathy related to chronic Chagas disease. Tc323, a ~323 kDa protein, is an uncharacterized protein showing putative quinoprotein alcohol dehydrogenase-like domains. A computational molecular docking study revealed that the scFv 6B6 binds to an internal domain of Tc323. Immunofluorescence microscopy and Western Blot showed that Tc323 is expressed in the main developmental forms of T. cruzi, localized intracellularly and exhibiting a membrane-associated pattern. According to phylogenetic analysis, Tc323 is highly conserved throughout evolution in all the lineages of T. cruzi so far identified, but it is absent in Leishmania spp. and Trypanosoma brucei. Most interestingly, only plasma samples from patients infected with T. cruzi and those with mixed infection with Leishmania spp. reacted against Tc323. Collectively, our findings demonstrate that Tc323 is a promising candidate for the differential serodiagnosis of chronic Chagas disease in areas where T. cruzi and Leishmania spp. infections coexist.
Collapse
Affiliation(s)
- Micaela S. Ossowski
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Juan Pablo Gallardo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Leticia L. Niborski
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Jessica Rodríguez-Durán
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Walter J. Lapadula
- Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), Facultad de Química Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis 5700, Argentina; (W.J.L.); (M.J.A.)
| | - Maximiliano Juri Ayub
- Instituto Multidisciplinario de Investigaciones Biológicas de San Luis (IMIBIO-SL-CONICET), Facultad de Química Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis 5700, Argentina; (W.J.L.); (M.J.A.)
| | - Raúl Chadi
- Hospital General de Agudos “Dr. Ignacio Pirovano”, Buenos Aires 1430, Argentina;
| | - Yolanda Hernandez
- Instituto Nacional de Parasitología “Dr. Mario Fatala Chaben”, Buenos Aires 1063, Argentina; (Y.H.); (M.L.F.)
| | - Marisa L. Fernandez
- Instituto Nacional de Parasitología “Dr. Mario Fatala Chaben”, Buenos Aires 1063, Argentina; (Y.H.); (M.L.F.)
| | - Mariana Potenza
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| | - Karina A. Gómez
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires 1428, Argentina; (M.S.O.); (J.P.G.); (L.L.N.); (J.R.-D.)
| |
Collapse
|
6
|
Chiribao ML, Díaz-Viraqué F, Libisch MG, Batthyány C, Cunha N, De Souza W, Parodi-Talice A, Robello C. Paracrine Signaling Mediated by the Cytosolic Tryparedoxin Peroxidase of Trypanosoma cruzi. Pathogens 2024; 13:67. [PMID: 38251374 PMCID: PMC10818299 DOI: 10.3390/pathogens13010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Peroxiredoxins are abundant and ubiquitous proteins that participate in different cellular functions, such as oxidant detoxification, protein folding, and intracellular signaling. Under different cellular conditions, peroxiredoxins can be secreted by different parasites, promoting the induction of immune responses in hosts. In this work, we demonstrated that the cytosolic tryparedoxin peroxidase of Trypanosoma cruzi (cTXNPx) is secreted by epimastigotes and trypomastigotes associated with extracellular vesicles and also as a vesicle-free protein. By confocal microscopy, we show that cTXNPx can enter host cells by an active mechanism both through vesicles and as a recombinant protein. Transcriptomic analysis revealed that cTXNPx induces endoplasmic reticulum stress and interleukin-8 expression in epithelial cells. This analysis also suggested alterations in cholesterol metabolism in cTXNPx-treated cells, which was confirmed by immunofluorescence showing the accumulation of LDL and the induction of LDL receptors in both epithelial cells and macrophages. BrdU incorporation assays and qPCR showed that cTXNPx has a mitogenic, proliferative, and proinflammatory effect on these cells in a dose-dependent manner. Importantly, we also demonstrated that cTXNPx acts as a paracrine virulence factor, increasing the susceptibility to infection in cTXNPx-pretreated epithelial cells by approximately 40%. Although the results presented in this work are from in vitro studies and likely underestimate the complexity of parasite-host interactions, our work suggests a relevant role for this protein in establishing infection.
Collapse
Affiliation(s)
- María Laura Chiribao
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11000, Uruguay;
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - Florencia Díaz-Viraqué
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - María Gabriela Libisch
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Montevideo 11000, Uruguay;
| | - Narcisa Cunha
- Instituto de Biofísica Carlos Chagas Filho, Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (N.C.); (W.D.S.)
| | - Wanderley De Souza
- Instituto de Biofísica Carlos Chagas Filho, Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (N.C.); (W.D.S.)
| | - Adriana Parodi-Talice
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
- Sección Genética Evolutiva, Instituto de Biología, Facultad de Ciencias, Universidad de la República, Montevideo 11000, Uruguay
| | - Carlos Robello
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11000, Uruguay;
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| |
Collapse
|
7
|
Morrison LJ, Steketee PC, Tettey MD, Matthews KR. Pathogenicity and virulence of African trypanosomes: From laboratory models to clinically relevant hosts. Virulence 2023; 14:2150445. [PMID: 36419235 DOI: 10.1080/21505594.2022.2150445] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
African trypanosomes are vector-borne protozoa, which cause significant human and animal disease across sub-Saharan Africa, and animal disease across Asia and South America. In humans, infection is caused by variants of Trypanosoma brucei, and is characterized by varying rate of progression to neurological disease, caused by parasites exiting the vasculature and entering the brain. Animal disease is caused by multiple species of trypanosome, primarily T. congolense, T. vivax, and T. brucei. These trypanosomes also infect multiple species of mammalian host, and this complexity of trypanosome and host diversity is reflected in the spectrum of severity of disease in animal trypanosomiasis, ranging from hyperacute infections associated with mortality to long-term chronic infections, and is also a main reason why designing interventions for animal trypanosomiasis is so challenging. In this review, we will provide an overview of the current understanding of trypanosome determinants of infection progression and severity, covering laboratory models of disease, as well as human and livestock disease. We will also highlight gaps in knowledge and capabilities, which represent opportunities to both further our fundamental understanding of how trypanosomes cause disease, as well as facilitating the development of the novel interventions that are so badly needed to reduce the burden of disease caused by these important pathogens.
Collapse
Affiliation(s)
- Liam J Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Pieter C Steketee
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Mabel D Tettey
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Keith R Matthews
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
8
|
Morales-Velásquez M, Barón-Vera JP, Osorio-Pulgarín MI, Sánchez-Jiménez MM, Ospina-Villa JD. Biomarkers for the diagnosis, treatment follow-up, and prediction of cardiac complications in Chagas disease in chronic phase: Recent advances. Parasite Immunol 2023; 45:e13013. [PMID: 37795913 DOI: 10.1111/pim.13013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/10/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023]
Abstract
Chagas disease is caused by the Trypanosoma cruzi parasite and is transmitted by infected triatomine bugs. This infection affects approximately 8 million people in the Americas, and due to globalisation and displacement, it is becoming increasingly common to find infected patients worldwide. Diagnosis of the disease in its acute form is relatively simple, as the parasite can be detected in peripheral blood smears, and symptoms are visible. However, in its chronic condition, the parasite is almost undetectable, and indirect tests are necessary to determine the presence of antibodies in infected patients. It is important to note that a single test is not enough to confirm the disease in this phase, as a second serological test should confirm the diagnosis. If the results are contradictory, a third test should be performed to confirm or discard the disease. Unfortunately, laboratories may not have access to all necessary tests in many rural areas where the disease is more frequent. Rapid tests to diagnose this disease present problems, such as significant variations in sensitivity and specificity in different countries. Therefore, searching for new biomarkers that allow for optimal correlation is essential. In this work, we have searched scientific literature from the last 10 years for mentions of novel biomarkers for diagnosis, treatment follow-up, and prediction of cardiac complications in Chagas disease in its chronic phase.
Collapse
Affiliation(s)
| | - Juan Pablo Barón-Vera
- Instituto Colombiano de Medicina Tropical, Universidad CES, Sabaneta, Antioquia, Colombia
| | | | | | | |
Collapse
|
9
|
Morales-Velásquez M, Barón-Vera JP, Pulgarín-Osorio MI, Sánchez-Jiménez MM, Ospina-Villa JD. Identification of the ATPase alpha subunit of Trypanosoma cruzi as a potential biomarker for the diagnosis of Chagas disease. Biomarkers 2023; 28:599-607. [PMID: 37667642 DOI: 10.1080/1354750x.2023.2255756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Chagas disease (CD) is considered by the World Health Organisation (WHO) a neglected disease endemic to the Americas, but it has spread throughout the world due to migrations. The disease is almost 100% curable if detected in time. Still, the lack of rapid diagnostic tests with sufficient sensitivity and specificity leads to a chronic phase with a mortality of about 50,000 people worldwide per year. METHODS Using the total proteins extracted from serum samples of patients confirmed with chronic phase CD; we performed the Bio-SELEX strategy. The best aptamers were selected using next-generation sequencing (NGS) based on their most abundant sequences (reads and rpm). Then, selected aptamers were used to isolate potential biomarkers directly from serum samples of patients with chronic phase CD using pull-down and mass spectrometry experiments. RESULTS CH1 aptamer was the aptamer selected after the NGS results analysis. The pull-down and mass spectrometry experiments identified the presence of the ATPase alpha subunit of T. cruzi circulating in serum samples of patients with chronic phase CD. CONCLUSIONS We report the ATPase alpha subunit of T. cruzi as a potential biomarker for chronic phase CD and CH1 aptamer as a potential tool for diagnosing CD.
Collapse
Affiliation(s)
- M Morales-Velásquez
- Tropical Medicine, Instituto Colombiano de Medicina Tropical - ICMT, Universidad CES, Medellin, Colombia
| | - J P Barón-Vera
- Tropical Medicine, Instituto Colombiano de Medicina Tropical - ICMT, Universidad CES, Medellin, Colombia
| | - M I Pulgarín-Osorio
- Tropical Medicine, Instituto Colombiano de Medicina Tropical - ICMT, Universidad CES, Medellin, Colombia
| | - M M Sánchez-Jiménez
- Tropical Medicine, Instituto Colombiano de Medicina Tropical - ICMT, Universidad CES, Medellin, Colombia
| | - J D Ospina-Villa
- Tropical Medicine, Instituto Colombiano de Medicina Tropical - ICMT, Universidad CES, Medellin, Colombia
| |
Collapse
|
10
|
Bunkofske ME, Perumal N, White B, Strauch EM, Tarleton R. Epitopes in the Glycosylphosphatidylinositol Attachment Signal Peptide of Trypanosoma cruzi Mucin Proteins Generate Robust but Delayed and Nonprotective CD8+ T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:420-430. [PMID: 36603035 PMCID: PMC9898211 DOI: 10.4049/jimmunol.2200723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023]
Abstract
Infection with the protozoan parasite Trypanosoma cruzi elicits substantial CD8+ T cell responses that disproportionately target epitopes encoded in the large trans-sialidase (TS) gene family. Within the C57BL/6 infection model, a significant proportion (30-40%) of the T. cruzi-specific CD8+ T cell response targets two immunodominant TS epitopes, TSKb18 and TSKb20. However, both TS-specific CD8+ T cell responses are dispensable for immune control, and TS-based vaccines have no demonstrable impact on parasite persistence, a determinant of disease. Besides TS, the specificity and protective capacity of CD8+ T cells that mediate immune control of T. cruzi infection are unknown. With the goal of identifying alternative CD8+ T cell targets, we designed and screened a representative set of genome-wide, in silico-predicted epitopes. Our screen identified a previously uncharacterized, to our knowledge, T cell epitope MUCKb25, found within mucin family proteins, the third most expanded large gene family in T. cruzi. The MUCKb25-specific response was characterized by delayed kinetics, relative to TS-specific responses, and extensive cross-reactivity with a large number of endogenous epitope variants. Similar to TS-specific responses, the MUCKb25 response was dispensable for control of the infection, and vaccination to generate MUCK-specific CD8+ T cells failed to confer protection. The lack of protection by MUCK vaccination was partly attributed to the fact that MUCKb25-specific T cells exhibit limited recognition of T. cruzi-infected host cells. Overall, these results indicate that the CD8+ T cell compartment in many T. cruzi-infected mice is occupied by cells with minimal apparent effector potential.
Collapse
Affiliation(s)
- Molly E. Bunkofske
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Natasha Perumal
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Brooke White
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Eva-Maria Strauch
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Rick Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
11
|
Ramírez JL. The Elusive Trypanosoma cruzi Disperse Gene Protein Family (DGF-1). Pathogens 2023; 12:pathogens12020292. [PMID: 36839564 PMCID: PMC9967923 DOI: 10.3390/pathogens12020292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/12/2023] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi infections, is included in the group of neglected diseases, and efforts to develop new therapeutic or immunoprevention approaches have not been successful. After the publication of the T. cruzi genome, the number of molecular and biochemical studies on this parasite has increased considerably, many of which are focused on families of variant surface proteins, especially trans-sialidases, mucins, and mucin-associated proteins. The disperse gene protein 1 family (DGF-1) is one of the most abundant families in the T. cruzi genome; however, the large gene size, high copy numbers, and low antibody titers detected in infected humans make it an unattractive study target. However, here we argue that given the ubiquitous presence in all T. cruzi species, and physicochemical characteristics, the DGF-1 gene family may play and important role in host-parasite interactions.
Collapse
Affiliation(s)
- José Luis Ramírez
- Instituto de Estudios Avanzados, Caracas, Venezuela and Universidad Central de Venezuela, Caracas 1080, Venezuela
| |
Collapse
|
12
|
Moreira RS, Calomeno NA, das Neves GB, do Nascimento LFN, Filho VB, Wagner G, Miletti LC. Trypanosoma evansi secretome carries potential biomarkers for Surra diagnosis. J Proteomics 2023; 272:104789. [PMID: 36464092 DOI: 10.1016/j.jprot.2022.104789] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
Trypanosoma evansi is a parasite that is phylogenetically close to Trypanosoma brucei and is the causative agent of a disease known as surra. Surra is responsible for a high mortality rate in livestock and large economic losses in the Americas, Africa, and Asia. This work aimed to analyze in vitro secreted proteins from T. evansi and identify potential treatment and diagnostic biomarkers for surra diagnosis. Two groups were used. In one group the parasites were purified using a DEAE-Cellulose column and maintained in a secretion medium while in the other group the parasites were not purified. Each group was further divided to be maintained at either 37 °C or 27 °C. We identified 246 proteins through mass spectrometry and found that the temperature appears to modulate protein secretion. We found minimal variations in the protein pools from pure and non-purified sets. We observed an emphasis on proteins associated to vesicles, glycolysis, and cellular homeostasis through the enrichment of GO. Also, we found that most secretome proteins share homologous proteins with T. b. brucei, T. b. gambiense, T. vivax, T. equiperdum, and T. b. rhodesiense secretome but unique T. evansi epitopes with potential biomarkers for surra diagnosis were detected. SIGNIFICANCE: Trypanosoma evansi is a parasite of African origin that is phylogenetically close to Trypanosoma brucei. As with other trypanosomatids and blood parasites, its infection causes non-pathognomonic symptoms, which makes its diagnosis difficult. One great problem is the fact that no diagnostic test differentiates between Trypanosoma equiperdum and T. evansi, which is a problem in South America and Asia, and Africa. Thus, it is urgent to study the biochemistry of the parasite to discover proteins that can be used for differential diagnosis or be possible therapeutic targets. In addition, the study of the secretome can point out proteins that are used by the parasite in its interactions with the host, helping to understand the progression of the disease.
Collapse
Affiliation(s)
- Renato Simões Moreira
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil; Instituto Federal de Santa Catarina (IFSC), Campus Gaspar, R. Adriano Kormann, 510 - Bela Vista, Gaspar, SC 89111-009, Brazil
| | - Nathália Anderson Calomeno
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil
| | - Gabriella Bassi das Neves
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil
| | - Luiz Flávio Nepomuceno do Nascimento
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil
| | - Vilmar Benetti Filho
- Laboratório de Bioinformática, Universidade Federal de Santa Catarina, Campus João David Ferreira Lima, Setor F, Bloco A, Sala 318, Caixa postal 476, Trindade, Florianópolis, SC 88040-970, Brazil
| | - Glauber Wagner
- Laboratório de Bioinformática, Universidade Federal de Santa Catarina, Campus João David Ferreira Lima, Setor F, Bloco A, Sala 318, Caixa postal 476, Trindade, Florianópolis, SC 88040-970, Brazil
| | - Luiz Claudio Miletti
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil.
| |
Collapse
|
13
|
Sato S, Dacher M, Kurumizaka H. Nucleosome Structures Built from Highly Divergent Histones: Parasites and Giant DNA Viruses. EPIGENOMES 2022; 6:22. [PMID: 35997368 PMCID: PMC9396995 DOI: 10.3390/epigenomes6030022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
In eukaryotes, genomic DNA is bound with histone proteins and packaged into chromatin. The nucleosome, a fundamental unit of chromatin, regulates the accessibility of DNA to enzymes involved in gene regulation. During the past few years, structural analyses of chromatin architectures have been limited to evolutionarily related organisms. The amino acid sequences of histone proteins are highly conserved from humans to yeasts, but are divergent in the deeply branching protozoan groups, including human parasites that are directly related to human health. Certain large DNA viruses, as well as archaeal organisms, contain distant homologs of eukaryotic histone proteins. The divergent sequences give rise to unique and distinct nucleosome architectures, although the fundamental principles of histone folding and DNA contact are highly conserved. In this article, we review the structures and biophysical properties of nucleosomes containing histones from the human parasites Giardia lamblia and Leishmania major, and histone-like proteins from the Marseilleviridae amoeba virus family. The presented data confirm the sharing of the overall DNA compaction system among evolutionally distant species and clarify the deviations from the species-specific nature of the nucleosome.
Collapse
Affiliation(s)
| | | | - Hitoshi Kurumizaka
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan; (S.S.); (M.D.)
| |
Collapse
|
14
|
Gulin JEN, Bisio MMC, Rocco D, Altcheh J, Solana ME, García-Bournissen F. Miltefosine and Benznidazole Combination Improve Anti-Trypanosoma cruzi In Vitro and In Vivo Efficacy. Front Cell Infect Microbiol 2022; 12:855119. [PMID: 35865815 PMCID: PMC9294734 DOI: 10.3389/fcimb.2022.855119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
Drug repurposing and combination therapy have been proposed as cost-effective strategies to improve Chagas disease treatment. Miltefosine (MLT), a synthetic alkylphospholipid initially developed for breast cancer and repositioned for leishmaniasis, is a promising candidate against Trypanosoma cruzi infection. This study evaluates the efficacy of MLT as a monodrug and combined with benznidazole (BZ) in both in vitro and in vivo models of infection with T. cruzi (VD strain, DTU TcVI). MLT exhibited in vitro activity on amastigotes and trypomastigotes with values of IC50 = 0.51 µM (0.48 µM; 0,55 µM) and LC50 = 31.17 µM (29.56 µM; 32.87 µM), respectively. Drug interaction was studied with the fixed-ration method. The sum of the fractional inhibitory concentrations (ΣFICs) resulted in ∑FIC= 0.45 for trypomastigotes and ∑FIC= 0.71 for amastigotes, suggesting in vitro synergistic and additive effects, respectively. No cytotoxic effects on host cells were observed. MLT efficacy was also evaluated in a murine model of acute infection alone or combined with BZ. Treatment was well tolerated with few adverse effects, and all treated animals displayed significantly lower mean peak parasitemia and mortality than infected non-treated controls (p<0.05). The in vivo studies showed that MLT led to a dose-dependent parasitostatic effect as monotherapy which could be improved by combining with BZ, preventing parasitemia rebound after a stringent immunosuppression protocol. These results support MLT activity in clinically relevant stages from T. cruzi, and it is the first report of positive interaction with BZ, providing further support for evaluating combined schemes using MLT and exploring synthetic alkylphospholipids as drug candidates.
Collapse
Affiliation(s)
- Julián Ernesto Nicolás Gulin
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños “Dr. Ricardo Gutiérrez, Ministerio de Salud, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina Universidad de Buenos Aires (UBA) – CONICET, Buenos Aires, Argentina
| | - Margarita María Catalina Bisio
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños “Dr. Ricardo Gutiérrez, Ministerio de Salud, Buenos Aires, Argentina
- Instituto Nacional de Parasitología (INP) ‘Dr. Mario Fatala Chaben’-Administración Nacional de Laboratorios e Institutos de Salud (ANLIS) ‘Dr. Carlos G. Malbrán’, CONICET, Buenos Aires, Argentina
| | - Daniela Rocco
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños “Dr. Ricardo Gutiérrez, Ministerio de Salud, Buenos Aires, Argentina
| | - Jaime Altcheh
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños “Dr. Ricardo Gutiérrez, Ministerio de Salud, Buenos Aires, Argentina
| | - María Elisa Solana
- Instituto de Microbiología y Parasitología Médica (IMPaM), Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Ciencias Básicas, Universidad Nacional de Luján, Buenos Aires, Argentina
| | - Facundo García-Bournissen
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños “Dr. Ricardo Gutiérrez, Ministerio de Salud, Buenos Aires, Argentina
- Division of Pediatric Clinical Pharmacology, Department of Pediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
- *Correspondence: Facundo García-Bournissen,
| |
Collapse
|
15
|
Molecular Recognition of Surface Trans-Sialidases in Extracellular Vesicles of the Parasite Trypanosoma cruzi Using Atomic Force Microscopy (AFM). Int J Mol Sci 2022; 23:ijms23137193. [PMID: 35806197 PMCID: PMC9266976 DOI: 10.3390/ijms23137193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 02/06/2023] Open
Abstract
Trans-sialidases (TS) are important constitutive macromolecules of the secretome present on the surface of Trypanosoma cruzi (T. cruzi) that play a central role as a virulence factor in Chagas disease. These enzymes have been related to infectivity, escape from immune surveillance and pathogenesis exhibited by this protozoan parasite. In this work, atomic force microscopy (AFM)-based single molecule-force spectroscopy is implemented as a suitable technique for the detection and location of functional TS on the surface of extracellular vesicles (EVs) released by tissue-culture cell-derived trypomastigotes (Ex-TcT). For that purpose, AFM cantilevers with functionalized tips bearing the anti-TS monoclonal antibody mAb 39 as a sense biomolecule are engineered using a covalent chemical ligation based on vinyl sulfonate click chemistry; a reliable, simple and efficient methodology for the molecular recognition of TS using the antibody-antigen interaction. Measurements of the breakdown forces between anti-TS mAb 39 antibodies and EVs performed to elucidate adhesion and forces involved in the recognition events demonstrate that EVs isolated from tissue-culture cell-derived trypomastigotes of T. cruzi are enriched in TS. Additionally, a mapping of the TS binding sites with submicrometer-scale resolution is provided. This work represents the first AFM-based molecular recognition study of Ex-TcT using an antibody-tethered AFM probe.
Collapse
|
16
|
Silvestre A, Shintre SS, Rachidi N. Released Parasite-Derived Kinases as Novel Targets for Antiparasitic Therapies. Front Cell Infect Microbiol 2022; 12:825458. [PMID: 35252034 PMCID: PMC8893276 DOI: 10.3389/fcimb.2022.825458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
The efficient manipulation of their host cell is an essential feature of intracellular parasites. Most molecular mechanisms governing the subversion of host cell by protozoan parasites involve the release of parasite-derived molecules into the host cell cytoplasm and direct interaction with host proteins. Among these released proteins, kinases are particularly important as they govern the subversion of important host pathways, such as signalling or metabolic pathways. These enzymes, which catalyse the transfer of a phosphate group from ATP onto serine, threonine, tyrosine or histidine residues to covalently modify proteins, are involved in numerous essential biological processes such as cell cycle or transport. Although little is known about the role of most of the released parasite-derived kinases in the host cell, they are examples of kinases hijacking host cellular pathways such as signal transduction or apoptosis, which are essential for immune response evasion as well as parasite survival and development. Here we present the current knowledge on released protozoan kinases and their involvement in host-pathogen interactions. We also highlight the knowledge gaps remaining before considering those kinases - involved in host signalling subversion - as antiparasitic drug targets.
Collapse
Affiliation(s)
- Anne Silvestre
- INRAE, Université de Tours, ISP, Nouzilly, France
- *Correspondence: Anne Silvestre, ; Najma Rachidi,
| | - Sharvani Shrinivas Shintre
- INRAE, Université de Tours, ISP, Nouzilly, France
- Institut Pasteur, Université de Paris and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Najma Rachidi
- Institut Pasteur, Université de Paris and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
- *Correspondence: Anne Silvestre, ; Najma Rachidi,
| |
Collapse
|
17
|
Baraibar AM, de Pascual R, Rodriguez Angulo HO, Mijares A, Hernández-Guijo JM. Pro-arrhythmogenic effects of Trypanosoma cruzi conditioned medium proteins in a model of bovine chromaffin cells. Parasitology 2021; 148:1612-1623. [PMID: 34384512 PMCID: PMC11010060 DOI: 10.1017/s003118202100130x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/05/2022]
Abstract
Asymptomatic sudden death is the principal cause of mortality in Chagas disease. There is little information about molecular mechanisms involved in the pathophysiology of malignant arrhythmias in Chagasic patients. Previous studies have involved Trypanosoma cruzi secretion proteins in the genesis of arrhythmias ex vivo, but the molecular mechanisms involved are still unresolved. Thus, the aim was to determine the effect of these secreted proteins on the cellular excitability throughout to test its effects on catecholamine secretion, sodium-, calcium-, and potassium-conductance and action potential (AP) firing. Conditioned medium was obtained from the co-culture of T. cruzi and Vero cells (African green monkey kidney cells) and ultra-filtered for concentrating immunogenic high molecular weight parasite proteins. Chromaffin cells were assessed with the parasite and Vero cells control medium. Parasite-secreted proteins induce catecholamine secretion in a dose-dependent manner. Additionally, T. cruzi conditioned medium induced depression of both calcium conductance and calcium and voltage-dependent potassium current. Interestingly, this fact was related to the abolishment of the hyperpolarization phase of the AP produced by the parasite medium. Taken together, these results suggest that T. cruzi proteins may be involved in the genesis of pro-arrhythmic conditions that could influence the appearance of malignant arrhythmias in Chagasic patients.
Collapse
Affiliation(s)
- A. M. Baraibar
- Department of Neuroscience, University of Minnesota, 4-158 Jackson Hall, 321 Church St s.e., Minneapolis, MN55455, USA
| | - R. de Pascual
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029Madrid, Spain
- Institute ‘Teófilo Hernando’, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029Madrid, Spain
| | - H. O. Rodriguez Angulo
- Laboratorio de Fisiología de Parásitos, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - A. Mijares
- Laboratorio de Fisiología de Parásitos, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - J. M. Hernández-Guijo
- Department of Pharmacology and Therapeutic, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029Madrid, Spain
- Institute ‘Teófilo Hernando’, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029Madrid, Spain
- IRYCIS, School of Medicine, Universidad Autónoma de Madrid, Av. Arzobispo Morcillo 4, 28029Madrid, Spain
| |
Collapse
|
18
|
Valenzuela-Muñoz V, Benavente BP, Casuso A, Leal Y, Valenzuela-Miranda D, Núñez-Acuña G, Sáez-Vera C, Gallardo-Escárate C. Transcriptome and morphological analysis in Caligus rogercresseyi uncover the effects of Atlantic salmon vaccination with IPath®. FISH & SHELLFISH IMMUNOLOGY 2021; 117:169-178. [PMID: 34389379 DOI: 10.1016/j.fsi.2021.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
It is known that iron transporter proteins and their regulation can modulate the fish's immune system, suggesting these proteins as a potential candidate for fish vaccines. Previous studies have evidenced the effects of Atlantic salmon immunized with the chimeric iron-related protein named IPath® against bacterial and ectoparasitic infections. The present study aimed to explore the transcriptome modulation and the morphology of the sea louse Caligus rogercresseyi in response to Atlantic salmon injected with IPath®. Herein, Atlantic salmon were injected with IPath® and challenged to sea lice in controlled laboratory conditions. Then, female adults were collected after 25 days post-infection for molecular and morphological evaluation. Transcriptome analysis conducted in lice collected from immunized fish revealed high modulation of transcripts compared with the control groups. Notably, the low number of up/downregulated transcripts was mainly found in lice exposed to the IPath® fish group. Among the top-25 differentially expressed genes, Vitellogenin, Cytochrome oxidases, and proteases genes were strongly downregulated, suggesting that IPath® can alter lipid transport, hydrogen ion transmembrane transport, and proteolysis. The morphological analysis in lice collected from IPath® fish revealed abnormal embryogenesis and inflammatory processes of the genital segment. Furthermore, head kidney, spleen, and skin were also analyzed in immunized fish to evaluate the transcription expression of immune and iron homeostasis-related genes. The results showed downregulation of TLR22, MCHII, IL-1β, ALAs, HO, BLVr, GSHPx, and Ferritin genes in head kidney and skin tissues; meanwhile, those genes did not show significant differences in spleen tissue. Overall, our findings suggest that IPath® can be used to enhance the fish immune response, showing a promissory commercial application against lice infections.
Collapse
Affiliation(s)
- Valentina Valenzuela-Muñoz
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile; Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepción, Chile
| | - Bárbara P Benavente
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile; Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepción, Chile
| | - Antonio Casuso
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile; Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepción, Chile
| | - Yeny Leal
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile; Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepción, Chile
| | - Diego Valenzuela-Miranda
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile; Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepción, Chile
| | - Gustavo Núñez-Acuña
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile; Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepción, Chile
| | - Constanza Sáez-Vera
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile; Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepción, Chile
| | - Cristian Gallardo-Escárate
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepción, Chile; Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepción, Chile.
| |
Collapse
|
19
|
Bisio MMC, Rivero R, Gonzalez N, Ballering G, D'Amico I, Kessler C, Moroni S, Moscatelli G, Ruiz AM, Altcheh J. Diagnostic Accuracy of Two Molecular Tools for Diagnosis of Congenital Chagas Disease. Mol Diagn Ther 2021; 25:791-801. [PMID: 34426953 PMCID: PMC8382099 DOI: 10.1007/s40291-021-00553-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE The real prevalence of congenital Chagas disease is undefined because of difficulties in the detection of Trypanosoma cruzi by microscopic examination. The aim of this study was to determine the diagnostic accuracy of two molecular diagnostic tools, qPCR and LAMP, in the diagnosis of congenital Chagas disease in a clinical setting. METHODS To this end, we conducted a prospective cohort study in a tertiary care center, of infants under 9 months of age, born in Buenos Aires to women with Chagas disease. Blood samples were collected for microscopic examination and molecular diagnosis at baseline. If negative, infants were followed up until 9 months of age to determine a final diagnosis by serology. In-house qPCR and LAMP previously validated were challenged as index tests. RESULTS A total of 154 participants were potentially eligible, 120 of whom were enrolled. Finally, 102 (66.2%) of them fulfilled the follow-up. The diagnosis of congenital Chagas disease was confirmed in 13 infants and excluded in 89. Both the sensitivity and specificity of the qPCR were 100.0% (95% confidence interval 75.3-100.0 and 95% confidence interval 95.9-100.0, respectively), whereas the sensitivity and specificity of LAMP were 69.2% (95% confidence interval 38.6-90.9) and 100% (95% confidence interval 95.9-100.0), respectively. CONCLUSIONS The qPCR agreed with the current diagnostic algorithm, and was a reliable and sensitive tool to detect congenital Chagas disease earlier, providing an appropriate and timely identification of infected infants requiring treatment. LAMP was able to detect congenital Chagas disease in infected infants by naked-eye visualization in accordance with a microscopic examination. The advantages of molecular diagnostic tools should be taken into account by the health system to improve congenital Chagas disease diagnosis.
Collapse
Affiliation(s)
- Margarita María Catalina Bisio
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina. .,Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET-GCBA), Buenos Aires, Argentina.
| | - Rocío Rivero
- Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Buenos Aires, Argentina
| | - Nicolás Gonzalez
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina
| | - Griselda Ballering
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina
| | - Indira D'Amico
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina
| | - Camila Kessler
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET-GCBA), Buenos Aires, Argentina
| | - Samanta Moroni
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina
| | - Guillermo Moscatelli
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina.,Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET-GCBA), Buenos Aires, Argentina
| | - Andrés Mariano Ruiz
- Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Jaime Altcheh
- Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", CP 1425 Gallo 1330, Buenos Aires, Argentina. .,Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET-GCBA), Buenos Aires, Argentina.
| |
Collapse
|
20
|
Gulin JEN, Rocco DM, Alonso V, Cribb P, Altcheh J, García-Bournissen F. Optimization and biological validation of an in vitro assay using the transfected Dm28c/pLacZ Trypanosoma cruzi strain. Biol Methods Protoc 2021; 6:bpab004. [PMID: 34386588 PMCID: PMC8355463 DOI: 10.1093/biomethods/bpab004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/15/2021] [Accepted: 07/01/2021] [Indexed: 12/03/2022] Open
Abstract
There is an urgent need to develop safer and more effective drugs for Chagas disease, as the current treatment relies on benznidazole (BZ) and nifurtimox (NFX). Using the Trypanosoma cruzi Dm28c strain genetically engineered to express the Escherichia coli β-galactosidase gene, lacZ, we have adapted and validated an easy, quick and reliable in vitro assay suitable for high-throughput screening for candidate compounds with anti-T. cruzi activity. In vitro studies were conducted to determine trypomastigotes sensitivity to BZ and NFX from Dm28c/pLacZ strain by comparing the conventional labour-intensive microscopy counting method with the colourimetric assay. Drug concentrations producing the lysis of 50% of trypomastigotes (lytic concentration 50%) were 41.36 and 17.99 µM for BZ and NFX, respectively, when measured by microscopy and 44.74 and 38.94 µM, for the colourimetric method, respectively. The optimal conditions for the amastigote development inhibitory assay were established considering the parasite–host relationship (i.e. multiplicity of infection) and interaction time, the time for colourimetric readout and the incubation time with the β-galactosidase substrate. The drug concentrations resulting in 50% amastigote development inhibition obtained with the colourimetric assay were 2.31 µM for BZ and 0.97 µM for NFX, similar to the reported values for the Dm28c wild strain (2.80 and 1.5 µM, respectively). In summary, a colourimetric assay using the Dm28c/pLacZ strain of T. cruzi has been set up, obtaining biologically meaningful sensibility values with the reference compounds on both trypomastigotes and amastigotes forms. This development could be applied to high-throughput screening programmes aiming to identify compounds with anti-T. cruzi in vitro activity.
Collapse
Affiliation(s)
- Julián Ernesto Nicolás Gulin
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Gallo 1330 (C1425EFD), Buenos Aires, Argentina.,Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", Gallo 1330 (C1425EFD), Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires (UBA) Facultad de Medicina-CONICET, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
| | - Daniela Marisa Rocco
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Gallo 1330 (C1425EFD), Buenos Aires, Argentina.,Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", Gallo 1330 (C1425EFD), Buenos Aires, Argentina
| | - Victoria Alonso
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (IBR-CONICET-UNR), Suipacha 531 (2000), Rosario, Argentina
| | - Pamela Cribb
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (IBR-CONICET-UNR), Suipacha 531 (2000), Rosario, Argentina
| | - Jaime Altcheh
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Gallo 1330 (C1425EFD), Buenos Aires, Argentina.,Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", Gallo 1330 (C1425EFD), Buenos Aires, Argentina
| | - Facundo García-Bournissen
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Gobierno de la Ciudad de Buenos Aires (GCBA), Gallo 1330 (C1425EFD), Buenos Aires, Argentina.,Servicio de Parasitología y Enfermedad de Chagas, Hospital de Niños "Dr. Ricardo Gutiérrez", Gallo 1330 (C1425EFD), Buenos Aires, Argentina.,Division of Paediatric Clinical Pharmacology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, 800 Commissioners Rd. E., Rm. B1-437, London, Canada
| |
Collapse
|
21
|
López L, Chiribao ML, Girard MC, Gómez KA, Carasi P, Fernandez M, Hernandez Y, Robello C, Freire T, Piñeyro MD. The cytosolic tryparedoxin peroxidase from Trypanosoma cruzi induces a pro-inflammatory Th1 immune response in a peroxidatic cysteine-dependent manner. Immunology 2021; 163:46-59. [PMID: 33410127 PMCID: PMC8044337 DOI: 10.1111/imm.13302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/25/2020] [Accepted: 12/19/2020] [Indexed: 12/25/2022] Open
Abstract
Trypanosoma cruzi cytosolic tryparedoxin peroxidase (c-TXNPx) is a 2-Cys peroxiredoxin (Prx) with an important role in detoxifying host cell oxidative molecules during parasite infection. c-TXNPx is a virulence factor, as its overexpression enhances parasite infectivity and resistance to exogenous oxidation. As Prxs from other organisms possess immunomodulatory properties, we studied the effects of c-TXNPx in the immune response and analysed whether the presence of the peroxidatic cysteine is necessary to mediate these properties. To this end, we used a recombinant c-TXNPx and a mutant version (c-TXNPxC52S) lacking the peroxidatic cysteine. We first analysed the oligomerization profile, oxidation state and peroxidase activity of both proteins by gel filtration, Western blot and enzymatic assay, respectively. To investigate their immunological properties, we analysed the phenotype and functional activity of macrophage and dendritic cells and the T-cell response by flow cytometry after injection into mice. Our results show that c-TXNPx, but not c-TXNPxC52S, induces the recruitment of IL-12/23p40-producing innate antigen-presenting cells and promotes a strong specific Th1 immune response. Finally, we studied the cellular and humoral immune response developed in the context of parasite natural infection and found that only wild-type c-TXNPx induces proliferation and high levels of IFN-γ secretion in PBMC from chronic patients without demonstrable cardiac manifestations. In conclusion, we demonstrate that c-TXNPx possesses pro-inflammatory properties that depend on the presence of peroxidatic cysteine that is essential for peroxidase activity and quaternary structure of the protein and could contribute to rational design of immune-based strategies against Chagas disease.
Collapse
Affiliation(s)
- Lucía López
- Laboratorio de Inmunomodulación y Desarrollo de VacunasDepartamento de InmunobiologíaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
- Unidad de Biología MolecularInstitut Pasteur MontevideoMontevideoUruguay
| | - María Laura Chiribao
- Unidad de Biología MolecularInstitut Pasteur MontevideoMontevideoUruguay
- Departamento de BioquímicaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| | - Magalí C. Girard
- Laboratorio de Inmunología de las Infecciones por TripanosomátidosInstituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI‐CONICET)Buenos AiresArgentina
| | - Karina A. Gómez
- Laboratorio de Inmunología de las Infecciones por TripanosomátidosInstituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI‐CONICET)Buenos AiresArgentina
| | - Paula Carasi
- Laboratorio de Inmunomodulación y Desarrollo de VacunasDepartamento de InmunobiologíaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| | - Marisa Fernandez
- Instituto Nacional de Parasitología ‘Doctor Mario Fatala Chabén’Buenos AiresArgentina
| | - Yolanda Hernandez
- Instituto Nacional de Parasitología ‘Doctor Mario Fatala Chabén’Buenos AiresArgentina
| | - Carlos Robello
- Unidad de Biología MolecularInstitut Pasteur MontevideoMontevideoUruguay
- Departamento de BioquímicaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| | - Teresa Freire
- Laboratorio de Inmunomodulación y Desarrollo de VacunasDepartamento de InmunobiologíaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| | - María Dolores Piñeyro
- Unidad de Biología MolecularInstitut Pasteur MontevideoMontevideoUruguay
- Departamento de BioquímicaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| |
Collapse
|
22
|
Ferri G, Edreira MM. All Roads Lead to Cytosol: Trypanosoma cruzi Multi-Strategic Approach to Invasion. Front Cell Infect Microbiol 2021; 11:634793. [PMID: 33747982 PMCID: PMC7973469 DOI: 10.3389/fcimb.2021.634793] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022] Open
Abstract
T. cruzi has a complex life cycle involving four developmental stages namely, epimastigotes, metacyclic trypomastigotes, amastigotes and bloodstream trypomastigotes. Although trypomastigotes are the infective forms, extracellular amastigotes have also shown the ability to invade host cells. Both stages can invade a broad spectrum of host tissues, in fact, almost any nucleated cell can be the target of infection. To add complexity, the parasite presents high genetic variability with differential characteristics such as infectivity. In this review, we address the several strategies T. cruzi has developed to subvert the host cell signaling machinery in order to gain access to the host cell cytoplasm. Special attention is made to the numerous parasite/host protein interactions and to the set of signaling cascades activated during the formation of a parasite-containing vesicle, the parasitophorous vacuole, from which the parasite escapes to the cytosol, where differentiation and replication take place.
Collapse
Affiliation(s)
- Gabriel Ferri
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
| | - Martin M Edreira
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina.,Laboratorio de Biología Molecular de Trypanosoma, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina.,Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
23
|
A novel Trypanosoma cruzi secreted antigen as a potential biomarker of Chagas disease. Sci Rep 2020; 10:19591. [PMID: 33177582 PMCID: PMC7658208 DOI: 10.1038/s41598-020-76508-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 10/26/2020] [Indexed: 01/30/2023] Open
Abstract
Chagas drug discovery has been hampered by a lack of validated assays to establish treatment efficacy in pre-clinical animal models and in patients infected with T. cruzi. Reduced levels of parasite secreted antigens in the blood of infected hosts could be used to demonstrate treatment efficacy. A published proteomic study of parasite secreted antigens identified the hypothetical protein Tc_5171 as a secreted antigen. In this report, we developed Tc_5171 specific antibodies and showed that the native protein was expressed by the three life cycle stages of the parasite. Anti-peptide antibodies were able to detect the parasite antigen in blood of infected mice during the acute and the chronic phase of infection. Benznidazole treatment of infected mice significantly reduced their blood antigen levels. Of clinical significance, patients diagnosed with Chagas disease, either asymptomatic or with cardiac clinical symptoms had significantly higher Tc_5171 antigen levels compared to endemic controls. Pair-wise analysis, before and after Benznidazole treatment, of patients with asymptomatic Chagas disease showed a significant reduction in antigen levels post treatment. Taken together, our results indicate that Tc_5171 could be used as a novel biomarker of Chagas disease for diagnosis and to assess treatment efficacy.
Collapse
|
24
|
Genomic Organization and Generation of Genetic Variability in the RHS (Retrotransposon Hot Spot) Protein Multigene Family in Trypanosoma cruzi. Genes (Basel) 2020; 11:genes11091085. [PMID: 32957642 PMCID: PMC7563717 DOI: 10.3390/genes11091085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
Retrotransposon Hot Spot (RHS) is the most abundant gene family in Trypanosoma cruzi, with unknown function in this parasite. The aim of this work was to shed light on the organization and expression of RHS in T. cruzi. The diversity of the RHS protein family in T. cruzi was demonstrated by phylogenetic and recombination analyses. Transcribed sequences carrying the RHS domain were classified into ten distinct groups of monophyletic origin. We identified numerous recombination events among the RHS and traced the origins of the donors and target sequences. The transcribed RHS genes have a mosaic structure that may contain fragments of different RHS inserted in the target sequence. About 30% of RHS sequences are located in the subtelomere, a region very susceptible to recombination. The evolution of the RHS family has been marked by many events, including gene duplication by unequal mitotic crossing-over, homologous, as well as ectopic recombination, and gene conversion. The expression of RHS was analyzed by immunofluorescence and immunoblotting using anti-RHS antibodies. RHS proteins are evenly distributed in the nuclear region of T. cruzi replicative forms (amastigote and epimastigote), suggesting that they could be involved in the control of the chromatin structure and gene expression, as has been proposed for T. brucei.
Collapse
|
25
|
Watanabe Costa R, Batista MF, Meneghelli I, Vidal RO, Nájera CA, Mendes AC, Andrade-Lima IA, da Silveira JF, Lopes LR, Ferreira LRP, Antoneli F, Bahia D. Comparative Analysis of the Secretome and Interactome of Trypanosoma cruzi and Trypanosoma rangeli Reveals Species Specific Immune Response Modulating Proteins. Front Immunol 2020; 11:1774. [PMID: 32973747 PMCID: PMC7481403 DOI: 10.3389/fimmu.2020.01774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/02/2020] [Indexed: 12/04/2022] Open
Abstract
Chagas disease, a zoonosis caused by the flagellate protozoan Trypanosoma cruzi, is a chronic and systemic parasitic infection that affects ~5–7 million people worldwide, mainly in Latin America. Chagas disease is an emerging public health problem due to the lack of vaccines and effective treatments. According to recent studies, several T. cruzi secreted proteins interact with the human host during cell invasion. Moreover, some comparative studies with T. rangeli, which is non-pathogenic in humans, have been performed to identify proteins directly involved in the pathogenesis of the disease. In this study, we present an integrated analysis of canonical putative secreted proteins (PSPs) from both species. Additionally, we propose an interactome with human host and gene family clusters, and a phylogenetic inference of a selected protein. In total, we identified 322 exclusively PSPs in T. cruzi and 202 in T. rangeli. Among the PSPs identified in T. cruzi, we found several trans-sialidases, mucins, MASPs, proteins with phospholipase 2 domains (PLA2-like), and proteins with Hsp70 domains (Hsp70-like) which have been previously characterized and demonstrated to be related to T. cruzi virulence. PSPs found in T. rangeli were related to protozoan metabolism, specifically carboxylases and phosphatases. Furthermore, we also identified PSPs that may interact with the human immune system, including heat shock and MASP proteins, but in a lower number compared to T. cruzi. Interestingly, we describe a hypothetical hybrid interactome of PSPs which reveals that T. cruzi secreted molecules may be down-regulating IL-17 whilst T. rangeli may enhance the production of IL-15. These results will pave the way for a better understanding of the pathophysiology of Chagas disease and may ultimately lead to the identification of molecular targets, such as key PSPs, that could be used to minimize the health outcomes of Chagas disease by modulating the immune response triggered by T. cruzi infection.
Collapse
Affiliation(s)
- Renata Watanabe Costa
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marina Ferreira Batista
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabela Meneghelli
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ramon Oliveira Vidal
- The Berlin Institute for Medical Systems Biology-Max Delbrück Center for Molecular Medicine in the Helmholtz Association in Berlin, Berlin, Germany.,Laboratorio Nacional de Biociências (LNBio), Campinas, São Paulo, Brazil
| | - Carlos Alcides Nájera
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Clara Mendes
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela Augusta Andrade-Lima
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luciano Rodrigo Lopes
- Departamento de Informática em Saúde, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ludmila Rodrigues Pinto Ferreira
- RNA Systems Biology Lab (RSBL), Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Antoneli
- Departamento de Informática em Saúde, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
26
|
Prevalence and Epitope Recognition of Anti- Trypanosoma cruzi Antibodies in Two Procyonid Species: Implications for Host Resistance. Pathogens 2020; 9:pathogens9060464. [PMID: 32545481 PMCID: PMC7350377 DOI: 10.3390/pathogens9060464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/01/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022] Open
Abstract
More than 180 mammalian species have been found naturally infected with Trypanosoma cruzi. Many of them play an important role in the maintenance of this parasite. In particular, new studies have appeared which indicate that some species of Procyonidae family may play a role as T. cruzi hosts, however, more data are needed to evaluate their long-term physiological response to parasite infection, especially for specific antibodies. In this study, antibodies to T. cruzi were detected and prevalence and epitope recognition were assessed by ELISA (using discrete typing unit (DTU) I as antigen) and WB (using DTU I and DTU II as antigens) and sera from two procyonid species obtained through five-year follow-up of two semicaptive populations living in the same habitat. Marked heterogeneity in antigens recognition between species and differences in seroprevalence (p = 0.0002) between white-nosed coatis (Nasua narica), 51.8% (115/222), and common raccoons (Procyon lotor), 28.3% (23/81), were found. Antigens with high molecular weight when DTU-I was used were the most recognized, while a greater antigen diversity recognition was observed with DTU-II; for white-nosed coatis, low-molecular-weight antigens were mainly recognized, while for common raccoons proteins with molecular weights greater than 80 kDa were recognized most. These divergent humoral immune responses could be related to an alleged pattern of recognition receptors and major histocompatibility complex molecules difference in the procyonids species.
Collapse
|
27
|
Parthasarathy A, Kalesh K. Defeating the trypanosomatid trio: proteomics of the protozoan parasites causing neglected tropical diseases. RSC Med Chem 2020; 11:625-645. [PMID: 33479664 PMCID: PMC7549140 DOI: 10.1039/d0md00122h] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022] Open
Abstract
Mass spectrometry-based proteomics enables accurate measurement of the modulations of proteins on a large scale upon perturbation and facilitates the understanding of the functional roles of proteins in biological systems. It is a particularly relevant methodology for studying Leishmania spp., Trypanosoma cruzi and Trypanosoma brucei, as the gene expression in these parasites is primarily regulated by posttranscriptional mechanisms. Large-scale proteomics studies have revealed a plethora of information regarding modulated proteins and their molecular interactions during various life processes of the protozoans, including stress adaptation, life cycle changes and interactions with the host. Important molecular processes within the parasite that regulate the activity and subcellular localisation of its proteins, including several co- and post-translational modifications, are also accurately captured by modern proteomics mass spectrometry techniques. Finally, in combination with synthetic chemistry, proteomic techniques facilitate unbiased profiling of targets and off-targets of pharmacologically active compounds in the parasites. This provides important data sets for their mechanism of action studies, thereby aiding drug development programmes.
Collapse
Affiliation(s)
- Anutthaman Parthasarathy
- Rochester Institute of Technology , Thomas H. Gosnell School of Life Sciences , 85 Lomb Memorial Dr , Rochester , NY 14623 , USA
| | - Karunakaran Kalesh
- Department of Chemistry , Durham University , Lower Mount Joy, South Road , Durham DH1 3LE , UK .
| |
Collapse
|
28
|
Lampaki D, Diepold A, Glatter T. A Serial Sample Processing Strategy with Improved Performance for in-Depth Quantitative Analysis of Type III Secretion Events in Pseudomonas aeruginosa. J Proteome Res 2020; 19:543-553. [PMID: 31814412 DOI: 10.1021/acs.jproteome.9b00628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The efficient analysis of secretomes is important to study the mechanisms of bacterial secretion. However, secretome analysis of bacteria that rely on rich media for optimal secretion via modern quantitative shotgun proteomics workflows is often hampered by the higher degree of sample impurities. This may be a reason for the low number of quantitative secretome investigations in such cases. We assessed the efficiency and amenability for rich media secretome analysis of different workflows including precipitation, SP3, and a combined, serial workflow. Using the model organism Pseudomonas aeruginosa, we found that the combined TCA-SP3 strategy outperformed the other tested methods on all monitored qualitative and quantitative levels. This method proved to be most efficient in the recovery of proteins secreted by the type III secretion system (T3SS), including all known effector proteins and secretion machinery components. We monitored the compositional changes of secretome samples over time, and observed a strong increase in the secreted protein fraction by the T3SS 2 to 3 h after T3SS induction. Our study conceptually illustrates how the combination of TCA precipitation and SP3 results in orthogonality in depleting sample impurities accompanied by improved chromatographic peptide separation, and more efficient MS detection with improved quantification parameters.
Collapse
Affiliation(s)
- Dimitrios Lampaki
- Core Facility for Mass Spectrometry and Proteomics , Max Planck Institute for Terrestrial Microbiology , Karl-von-Frisch-Str. 10 , D-35043 Marburg , Germany.,Department of Ecophysiology , Max Planck Institute for Terrestrial Microbiology , Karl-von-Frisch-Str. 10 , D-35043 Marburg , Germany
| | - Andreas Diepold
- Department of Ecophysiology , Max Planck Institute for Terrestrial Microbiology , Karl-von-Frisch-Str. 10 , D-35043 Marburg , Germany
| | - Timo Glatter
- Core Facility for Mass Spectrometry and Proteomics , Max Planck Institute for Terrestrial Microbiology , Karl-von-Frisch-Str. 10 , D-35043 Marburg , Germany
| |
Collapse
|
29
|
Dacher M, Tachiwana H, Horikoshi N, Kujirai T, Taguchi H, Kimura H, Kurumizaka H. Incorporation and influence of Leishmania histone H3 in chromatin. Nucleic Acids Res 2019; 47:11637-11648. [PMID: 31722422 PMCID: PMC7145708 DOI: 10.1093/nar/gkz1040] [Citation(s) in RCA: 8] [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: 09/13/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022] Open
Abstract
Immunopathologies caused by Leishmania cause severe human morbidity and mortality. This protozoan parasite invades and persists inside host cells, resulting in disease development. Leishmania modifies the epigenomic status of the host cells, thus probably averting the host cell defense mechanism. To accomplish this, Leishmania may change the host cell chromatin structure. However, the mechanism by which the parasite changes the host cell chromatin has not been characterized. In the present study, we found that ectopically produced Leishmania histone H3, LmaH3, which mimics the secreted LmaH3 in infected cells, is incorporated into chromatin in human cells. A crystallographic analysis revealed that LmaH3 forms nucleosomes with human histones H2A, H2B and H4. We found that LmaH3 was less stably incorporated into the nucleosome, as compared to human H3.1. Consistently, we observed that LmaH3-H4 association was remarkably weakened. Mutational analyses revealed that the specific LmaH3 Trp35, Gln57 and Met98 residues, which correspond to the H3.1 Tyr41, Arg63 and Phe104 residues, might be responsible for the instability of the LmaH3 nucleosome. Nucleosomes containing LmaH3 resisted the Mg2+-mediated compaction of the chromatin fiber. These distinct physical characteristics of LmaH3 support the possibility that histones secreted by parasites during infection may modulate the host chromatin structure.
Collapse
Affiliation(s)
- Mariko Dacher
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hiroaki Tachiwana
- Department of Cancer Biology, The Cancer Institute of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Naoki Horikoshi
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Tomoya Kujirai
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hiroyuki Taguchi
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Hiroshi Kimura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
| | - Hitoshi Kurumizaka
- Laboratory of Chromatin Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| |
Collapse
|
30
|
Systematic Review of the Epidemiology of Chagas Disease in the Americas: a Call for Standardized Reporting of Chagas Disease Prevalence. CURRENT TROPICAL MEDICINE REPORTS 2019. [DOI: 10.1007/s40475-019-00177-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Retana Moreira L, Rodríguez Serrano F, Osuna A. Extracellular vesicles of Trypanosoma cruzi tissue-culture cell-derived trypomastigotes: Induction of physiological changes in non-parasitized culture cells. PLoS Negl Trop Dis 2019; 13:e0007163. [PMID: 30789912 PMCID: PMC6383987 DOI: 10.1371/journal.pntd.0007163] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/16/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Trypanosoma cruzi is the obligate intracellular parasite that causes Chagas disease. The pathogenesis of this disease is a multifactorial complex process that involves a large number of molecules and particles, including the extracellular vesicles. The presence of EVs of T. cruzi was first described in 1979 and, since then, research regarding these particles has been increasing. Some of the functions described for these EVs include the increase in heart parasitism and the immunomodulation and evasion of the host immune response. Also, EVs may be involved in parasite adhesion to host cells and host cell invasion. METHODOLOGY/PRINCIPAL FINDINGS EVs (exosomes) of the Pan4 strain of T. cruzi were isolated by differential centrifugation, and measured and quantified by TEM, NTA and DLS. The effect of EVs in increasing the parasitization of Vero cells was evaluated and the ED50 was calculated. Changes in cell permeability induced by EVs were evaluated in Vero and HL-1 cardiomyocyte cells using cell viability techniques such as trypan blue and MTT assays, and by confocal microscopy. The intracellular mobilization of Ca2+ and the disruption of the actin cytoskeleton induced by EVs over Vero cells were followed-up in time using confocal microscopy. To evaluate the effect of EVs over the cell cycle, cell cycle analyses using flow cytometry and Western blotting of the phosphorylated and non-phosphorylated protein of Retinoblastoma were performed. CONCLUSION/SIGNIFICANCE The incubation of cells with EVs of trypomastigotes of the Pan4 strain of T. cruzi induce a number of changes in the host cells that include a change in cell permeability and higher intracellular levels of Ca2+ that can alter the dynamics of the actin cytoskeleton and arrest the cell cycle at G0/G1 prior to the DNA synthesis necessary to complete mitosis. These changes aid the invasion of host cells and augment the percentage of cell parasitization.
Collapse
Affiliation(s)
- Lissette Retana Moreira
- Instituto de Biotecnología, Grupo de Bioquímica y Parasitología Molecular, Departamento de Parasitología, Universidad de Granada, Granada, Spain
| | | | - Antonio Osuna
- Instituto de Biotecnología, Grupo de Bioquímica y Parasitología Molecular, Departamento de Parasitología, Universidad de Granada, Granada, Spain
- * E-mail:
| |
Collapse
|
32
|
Garg G, Singh K, Ali V. Proteomic approaches unravel the intricacy of secreted proteins of Leishmania: An updated review. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:913-923. [DOI: 10.1016/j.bbapap.2018.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/16/2018] [Accepted: 05/22/2018] [Indexed: 02/03/2023]
|
33
|
Caeiro LD, Alba-Soto CD, Rizzi M, Solana ME, Rodriguez G, Chidichimo AM, Rodriguez ME, Sánchez DO, Levy GV, Tekiel V. The protein family TcTASV-C is a novel Trypanosoma cruzi virulence factor secreted in extracellular vesicles by trypomastigotes and highly expressed in bloodstream forms. PLoS Negl Trop Dis 2018; 12:e0006475. [PMID: 29727453 PMCID: PMC5955593 DOI: 10.1371/journal.pntd.0006475] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 05/16/2018] [Accepted: 04/24/2018] [Indexed: 12/19/2022] Open
Abstract
TcTASV-C is a protein family of about 15 members that is expressed only in the trypomastigote stage of Trypanosoma cruzi. We have previously shown that TcTASV-C is located at the parasite surface and secreted to the medium. Here we report that the expression of different TcTASV-C genes occurs simultaneously at the trypomastigote stage and while some secreted and parasite-associated products are found in both fractions, others are different. Secreted TcTASV-C are mainly shedded through trypomastigote extracellular vesicles, of which they are an abundant constituent, despite its scarce expression on culture-derived trypomastigotes. In contrast, TcTASV-C is highly expressed in bloodstream trypomastigotes; its upregulation in bloodstream parasites was observed in different T. cruzi strains and was specific for TcTASV-C, suggesting that some host-molecules trigger TcTASV-C expression. TcTASV-C is also strongly secreted by bloodstream parasites. A DNA prime—protein boost immunization scheme with TcTASV-C was only partially effective to control the infection in mice challenged with a highly virulent T. cruzi strain. Vaccination triggered a strong humoral response that delayed the appearance of bloodstream trypomastigotes at the early phase of the infection. Linear epitopes recognized by vaccinated mice were mapped within the TcTASV-C family motif, suggesting that blockade of secreted TcTASV-C impacts on the settlement of infection. Furthermore, although experimental and naturally T. cruzi-infected hosts did not react with antigens from extracellular vesicles, vaccinated and challenged mice recognized not only TcTASV-C but also other vesicle-antigens. We hypothesize that TcTASV-C is involved in the establishment of the initial T. cruzi infection in the mammalian host. Altogether, these results point towards TcTASV-C as a novel secreted virulence factor of T. cruzi trypomastigotes. Trypanosoma cruzi is the kinetoplastid parasite that causes Chagas’ disease, a neglected infection endemic in Latin America and emerging worldwide. Being vaccines currently unavailable and treatments not completely effective, identification and characterization of parasite molecules that can be target for these interventions are urgently needed. Of particular interest are surface anchored and secreted proteins involved in parasite—host interplay. Recently, extracellular vesicles released from protozoan pathogens have been shown to alter host cell function favoring the establishment of infection. Trypomastigotes are the disseminating stage of T. cruzi, being their presence in peripheral blood a hallmark of early acute infection in mammals. While the most abundant proteins of the trypomastigote surface are fairly well characterized, little is known about other, less abundant and more recently discovered multigenic families, which could have critical functions in the parasite—host interaction. The T. cruziTrypomastigote Alanine, Valine and Serine rich proteins (TcTASV) belong to a medium-size multigene family of ~40 members that remained unobserved until a few years ago when it was identified through a trypomastigote-enriched cDNA library. Almost simultaneously, an expression library immunization approach designed to discover novel vaccine antigens in T. cruzi, spotlighted the TcTASV-C subfamily, as a fragment of a TcTASV-C gene was identified in a pool of protective clones. A distinctive feature that characterizes TcTASV proteins–and particularly the TcTASV-C subfamily- is their predominant expression in trypomastigotes. Recent transcriptomic and proteomic studies uphold our previous observations that the TcTASV family is over-represented in the trypomastigote stage, and therefore could represent an interesting target for rational intervention against T. cruzi infection. Here show that TcTASV-C is mainly secreted through extracellular vesicles (EVs) of trypomastigotes, and is a major cargo of its content. We have also shown that TcTASV-C is much more expressed in trypomastigotes purified from blood from infected mice than in trypomastigotes harvested from in vitro cultures, suggesting that host molecules should trigger TcTASV-C expression in vivo during the infection. The immunization of mice with TcTASV-C interfered with the early acute phase of T. cruzi infection through a strong humoral immune response. TcTASV-C should be considered as a novel secreted virulence factor of T. cruzi trypomastigotes and -although its biological function is still unknown- we hypothesize its participation in the early steps of T cruzi infection in the mammalian host.
Collapse
Affiliation(s)
- Lucas D Caeiro
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Catalina D Alba-Soto
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médicas (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Mariana Rizzi
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - María Elisa Solana
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médicas (IMPaM), UBA-CONICET, Buenos Aires, Argentina.,Departamento de Cs. Básicas, Universidad Nacional de Luján, Luján, Buenos Aires, Argentina
| | - Giselle Rodriguez
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Agustina M Chidichimo
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Matías E Rodriguez
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Daniel O Sánchez
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Gabriela V Levy
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Valeria Tekiel
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| |
Collapse
|
34
|
Holzmuller P, Geiger A, Nzoumbou-Boko R, Pissarra J, Hamrouni S, Rodrigues V, Dauchy FA, Lemesre JL, Vincendeau P, Bras-Gonçalves R. Trypanosomatid Infections: How Do Parasites and Their Excreted-Secreted Factors Modulate the Inducible Metabolism of l-Arginine in Macrophages? Front Immunol 2018; 9:778. [PMID: 29731753 PMCID: PMC5921530 DOI: 10.3389/fimmu.2018.00778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 03/28/2018] [Indexed: 12/20/2022] Open
Abstract
Mononuclear phagocytes (monocytes, dendritic cells, and macrophages) are among the first host cells to face intra- and extracellular protozoan parasites such as trypanosomatids, and significant expansion of macrophages has been observed in infected hosts. They play essential roles in the outcome of infections caused by trypanosomatids, as they can not only exert a powerful antimicrobial activity but also promote parasite proliferation. These varied functions, linked to their phenotypic and metabolic plasticity, are exerted via distinct activation states, in which l-arginine metabolism plays a pivotal role. Depending on the environmental factors and immune response elements, l-arginine metabolites contribute to parasite elimination, mainly through nitric oxide (NO) synthesis, or to parasite proliferation, through l-ornithine and polyamine production. To survive and adapt to their hosts, parasites such as trypanosomatids developed mechanisms of interaction to modulate macrophage activation in their favor, by manipulating several cellular metabolic pathways. Recent reports emphasize that some excreted-secreted (ES) molecules from parasites and sugar-binding host receptors play a major role in this dialog, particularly in the modulation of the macrophage's inducible l-arginine metabolism. Preventing l-arginine dysregulation by drugs or by immunization against trypanosomatid ES molecules or by blocking partner host molecules may control early infection and is a promising way to tackle neglected diseases including Chagas disease, leishmaniases, and African trypanosomiases. The present review summarizes recent knowledge on trypanosomatids and their ES factors with regard to their influence on macrophage activation pathways, mainly the NO synthase/arginase balance. The review ends with prospects for the use of biological knowledge to develop new strategies of interference in the infectious processes used by trypanosomatids, in particular for the development of vaccines or immunotherapeutic approaches.
Collapse
Affiliation(s)
- Philippe Holzmuller
- CIRAD, Montpellier, France.,UMR 117 ASTRE "Animal, Santé, Territoire, Risques et Ecosystèmes", Univ. Montpellier (I-MUSE), CIRAD, INRA, Montpellier, France
| | - Anne Geiger
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| | - Romaric Nzoumbou-Boko
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France.,Univ. Bordeaux, UMR 177 INTERTRYP, Bordeaux, France.,CHU Bordeaux, Laboratoire de Parasitologie-Mycologie, Bordeaux, France
| | - Joana Pissarra
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| | - Sarra Hamrouni
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| | - Valérie Rodrigues
- CIRAD, Montpellier, France.,UMR 117 ASTRE "Animal, Santé, Territoire, Risques et Ecosystèmes", Univ. Montpellier (I-MUSE), CIRAD, INRA, Montpellier, France
| | - Frédéric-Antoine Dauchy
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France.,Univ. Bordeaux, UMR 177 INTERTRYP, Bordeaux, France.,CHU Bordeaux, Département des Maladies Infectieuses et Tropicales, Bordeaux, France
| | - Jean-Loup Lemesre
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| | - Philippe Vincendeau
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France.,Univ. Bordeaux, UMR 177 INTERTRYP, Bordeaux, France.,CHU Bordeaux, Laboratoire de Parasitologie-Mycologie, Bordeaux, France
| | - Rachel Bras-Gonçalves
- UMR 177 INTERTRYP "Interactions Hôte-Vecteur-Parasite-Environnement dans les maladies tropicales négligées dues aux Trypanosomatidae", Univ. Montpellier (I-MUSE), CIRAD, IRD, Univ. Bordeaux 2, Univ. Lyon 1, Montpellier, France
| |
Collapse
|
35
|
Ribeiro KS, Vasconcellos CI, Soares RP, Mendes MT, Ellis CC, Aguilera-Flores M, de Almeida IC, Schenkman S, Iwai LK, Torrecilhas AC. Proteomic analysis reveals different composition of extracellular vesicles released by two Trypanosoma cruzi strains associated with their distinct interaction with host cells. J Extracell Vesicles 2018; 7:1463779. [PMID: 29696081 PMCID: PMC5912195 DOI: 10.1080/20013078.2018.1463779] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/07/2018] [Indexed: 12/12/2022] Open
Abstract
Trypanosoma cruzi, the aetiologic agent of Chagas disease, releases vesicles containing a wide range of surface molecules known to affect the host immunological responses and the cellular infectivity. Here, we compared the secretome of two distinct strains (Y and YuYu) of T. cruzi, which were previously shown to differentially modulate host innate and acquired immune responses. Tissue culture-derived trypomastigotes of both strains secreted extracellular vesicles (EVs), as demonstrated by electron scanning microscopy. EVs were purified by exclusion chromatography or ultracentrifugation and quantitated using nanoparticle tracking analysis. Trypomastigotes from YuYu strain released higher number of EVs than those from Y strain, enriched with virulence factors trans-sialidase (TS) and cruzipain. Proteomic analysis confirmed the increased abundance of proteins coded by the TS gene family, mucin-like glycoproteins, and some typical exosomal proteins in the YuYu strain, which also showed considerable differences between purified EVs and vesicle-free fraction as compared to the Y strain. To evaluate whether such differences were related to parasite infectivity, J774 macrophages and LLC-MK2 kidney cells were preincubated with purified EVs from both strains and then infected with Y strain trypomastigotes. EVs released by YuYu strain caused a lower infection but higher intracellular proliferation in J774 macrophages than EVs from Y strain. In contrast, YuYu strain-derived EVs caused higher infection of LLC-MK2 cells than Y strain-derived EVs. In conclusion, quantitative and qualitative differences in EVs and secreted proteins from different T. cruzi strains may correlate with infectivity/virulence during the host-parasite interaction.
Collapse
Affiliation(s)
| | | | | | - Maria Tays Mendes
- Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, TX, USA
| | - Cameron C Ellis
- Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, TX, USA
| | - Marcela Aguilera-Flores
- Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, TX, USA
| | - Igor Correia de Almeida
- Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, TX, USA
| | - Sergio Schenkman
- Departamento de Microbiologia, Imunologia e Parasitologia, UNIFESP, São Paulo, Brazil
| | - Leo Kei Iwai
- Laboratório Especial de Toxicologia Aplicada (LETA), Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | | |
Collapse
|