1
|
Dias N, Dias M, Ribeiro A, Gomes N, Moraes A, Wesley M, Gonzaga C, Ramos DDAR, Braz S, Dallago B, de Carvalho JL, Hagström L, Nitz N, Hecht M. Network Analysis of Pathogenesis Markers in Murine Chagas Disease Under Antimicrobial Treatment. Microorganisms 2024; 12:2332. [PMID: 39597721 PMCID: PMC11596328 DOI: 10.3390/microorganisms12112332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Chagas disease (CD), a disease affecting millions globally, remains shrouded in scientific uncertainty, particularly regarding the role of the intestinal microbiota in disease progression. This study investigates the effects of antibiotic-induced microbiota depletion on parasite burden, immune responses, and clinical outcomes in BALB/c mice infected with either the Trypanosoma cruzi Colombiana or CL Brener strains. Mice were treated with a broad-spectrum antibiotic cocktail before infection, and parasite burden was quantified via qPCR at 30 and 100 days post-infection (dpi). Immune responses were analyzed using flow cytometry and ELISA, while histopathology was conducted on cardiac and intestinal tissues. Antibiotic treatment uncovered strain-specific correlations, with Colombiana infections affecting Bifidobacterium populations and CL Brener infections linked to Lactobacillus. Microbiota depletion initially reduced parasite burden in the heart and intestine, but an increase was observed in the chronic phase, except in the CL Brener-infected gut, where an early burden spike was followed by a decline. Antibiotic-induced bacterial shifts, such as reductions in Bacteroides and Bifidobacterium, promoted a more pro-inflammatory immune profile. These findings highlight the importance of microbiota and strain-specific factors in CD and suggest further research into microbiota manipulation as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Nayra Dias
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Marina Dias
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Andressa Ribeiro
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Nélio Gomes
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Aline Moraes
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Moisés Wesley
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Carlito Gonzaga
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Doralina do Amaral Rabello Ramos
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Shélida Braz
- Institute of Exact and Technological Sciences, Federal University of Amazonas, Manaus 69000-000, Brazil;
| | - Bruno Dallago
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Juliana Lott de Carvalho
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Luciana Hagström
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Nadjar Nitz
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| | - Mariana Hecht
- Interdisciplinary Laboratory of Biosciences, Department of Pathology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (N.D.); (M.D.); (A.R.); (N.G.); (A.M.); (M.W.); (C.G.); (D.d.A.R.R.); (B.D.); (J.L.d.C.); (L.H.); (N.N.)
| |
Collapse
|
2
|
Salassa BN, Cueto JA, Vanrell MC, López MB, Descoteaux A, Labriola CA, Romano PS. The host Rab9a/Rab32 axis is actively recruited to the Trypanosoma cruzi parasitophorous vacuole and benefits the infection cycle. Mol Microbiol 2024; 122:643-659. [PMID: 38193389 DOI: 10.1111/mmi.15217] [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: 05/16/2023] [Revised: 10/26/2023] [Accepted: 12/15/2023] [Indexed: 01/10/2024]
Abstract
Trypanosoma cruzi, the etiological agent of Chagas disease is a protozoan parasite that infects phagocytic and non-phagocytic mammalian cells. At early stages of infection, trypomastigotes, the infective forms of this parasite, localize in a vesicular compartment called the T. cruzi parasitophorous vacuole until the exit of parasites to the host cell cytoplasm where continue their infective cycle. Rab proteins participate in the membrane traffic's molecular machinery, functioning as central regulators of vesicle recognition and transport. In previous work, we demonstrated that endocytic Rabs are key factors of the T. cruzi infection process in non-phagocytic cells, regulating the formation and the maturation of the vacuole. In this work, we identified and characterized other molecular components of the vesicular transport pathways and their participation in the T. cruzi infection. We found that Rab9a and Rab32, two regulators of the endocytic and autophagic pathways, were actively recruited to the T. cruzi vacuoles and favored the late stages of the infective process. The recruitment was specific and dependent on T. cruzi protein synthesis. Interestingly, Rab32 association depends on the presence of Rab9a in the vacuolar membrane, while the inhibition of the cysteine-protease cruzipain, a T. cruzi virulence factor, significantly decreases both Rab9a and Rab32 association with the vacuole. In summary, this work showed for the first time that specific molecules produced and secreted by the parasite can subvert intracellular components of host cells to benefit the infection. These new data shed light on the complex map of interactions between T. cruzi and the host cell and introduce concepts that can be useful in finding new forms of intervention against this parasite in the future.
Collapse
Affiliation(s)
- Betiana Nebaí Salassa
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora-Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Odontología, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Juan Agustín Cueto
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora-Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Cristina Vanrell
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora-Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Área Biología celular y molecular, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Belén López
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora-Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Área Biología celular y molecular, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Albert Descoteaux
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, Quebec, Canada
- Infectiopole INRS, Laval, Quebec, Canada
| | - Carlos Alberto Labriola
- Laboratorio de Biología estructural y celular, Fundación Instituto Leloir (FIL-CONICET), Buenos Aires, Argentina
| | - Patricia Silvia Romano
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora-Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Área Biología celular y molecular, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
3
|
Macedo-da-Silva J, Mule SN, Rosa-Fernandes L, Palmisano G. A computational pipeline elucidating functions of conserved hypothetical Trypanosoma cruzi proteins based on public proteomic data. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 138:401-428. [PMID: 38220431 DOI: 10.1016/bs.apcsb.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
The proteome is complex, dynamic, and functionally diverse. Functional proteomics aims to characterize the functions of proteins in biological systems. However, there is a delay in annotating the function of proteins, even in model organisms. This gap is even greater in other organisms, including Trypanosoma cruzi, the causative agent of the parasitic, systemic, and sometimes fatal disease called Chagas disease. About 99.8% of Trypanosoma cruzi proteome is not manually annotated (unreviewed), among which>25% are conserved hypothetical proteins (CHPs), calling attention to the knowledge gap on the protein content of this organism. CHPs are conserved proteins among different species of various evolutionary lineages; however, they lack functional validation. This study describes a bioinformatics pipeline applied to public proteomic data to infer possible biological functions of conserved hypothetical Trypanosoma cruzi proteins. Here, the adopted strategy consisted of collecting differentially expressed proteins between the epimastigote and metacyclic trypomastigotes stages of Trypanosoma cruzi; followed by the functional characterization of these CHPs applying a manifold learning technique for dimension reduction and 3D structure homology analysis (Spalog). We found a panel of 25 and 26 upregulated proteins in the epimastigote and metacyclic trypomastigote stages, respectively; among these, 18 CHPs (8 in the epimastigote stage and 10 in the metacyclic stage) were characterized. The data generated corroborate the literature and complement the functional analyses of differentially regulated proteins at each stage, as they attribute potential functions to CHPs, which are frequently identified in Trypanosoma cruzi proteomics studies. However, it is important to point out that experimental validation is required to deepen our understanding of the CHPs.
Collapse
Affiliation(s)
- Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Sao Paulo, Brazil
| | - Simon Ngao Mule
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Sao Paulo, Brazil
| | - Livia Rosa-Fernandes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Sao Paulo, Brazil; Centre for Motor Neuron Disease Research, Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Sydney, NSW, Australia
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Sao Paulo, Brazil; School of Natural Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Dutra Barroso Gomes N, Paula Magalhães E, Rodrigues Ribeiro L, Cavalcante JW, Morais Gomes Maia M, Cunha da Silva FR, Ali A, Machado Marinho M, Silva Marinho E, Silva Dos Santos H, Costa Martins AM, Róseo Paula Pessoa Bezerra de Menezes R. Trypanocidal potential of synthetic p-aminochalcones: In silico and in vitro evaluation. Bioorg Chem 2023; 141:106931. [PMID: 37879182 DOI: 10.1016/j.bioorg.2023.106931] [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: 07/19/2023] [Revised: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023]
Abstract
Chagas disease (CD) is a neglected tropical disease of worldwide health concern, caused by the flagellate protozoan Trypanosoma cruzi (T. cruzi), endemic in Latin America and present in North America and Europe. The WHO recommended drug for CD, benznidazole has low safety profile and several limitations. Therefore, an entity with better therapeutic potential to treat CD is required. Chalcones are an important class of compounds, which have shown antichagasic potential. Thus, the objective of this study was to evaluate the activity of synthetic p-aminochalcones against T. cruzi. Chalcones 1 and 2 were synthesized by Claisen-Schmidt condensation and characterized by both spectroscopic and theoretical methods. Initially, they were submitted to molecular docking simulations using cruzain and trypanothione reductase (TR) enzymes. It was expected to observe the possible interactions of chalcones with the catalytic site and other important regions of these main pharmacological targets of T. cruzi. Their cytotoxicity within host cells were assessed by MTT reduction assay using LLC-MK2 cells, with CC50 = 85.6 ± 9.2 µM and 1115 ± 381.7 µM for chalcones 1 and 2, respectively. These molecules were also tested against epimastigote and trypomastigote life forms of T. cruzi, causing reduction in the number of viable parasites. For the evaluation of the effect on intracellular amastigotes, infected LLC-MK2 cells were incubated with the chalcones for 24 h, causing reduction in the percentage of infected cells and the number of amastigotes/100 cells. Finally, flow cytometry assays were performed for analyzing cell death mechanisms (7-AAD/AxPE labelling), cytoplasmic ROS accumulation (DCFH-DA assay) and mitochondrial transmembrane potential disruption (Rho123 assay). Both chalcones (1 and 2) caused membrane damage, ROS accumulation and mitochondrial depolarization. In conclusion, the synthetic p-aminochalcones presented trypanocidal effect, causing membrane damage and oxidative stress. Their mechanism of action may be related to cruzain and TR inhibition.
Collapse
Affiliation(s)
| | - Emanuel Paula Magalhães
- Post-Graduate Program in Pharmaceutical Sciences, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lyanna Rodrigues Ribeiro
- Post-Graduate Program in Pharmaceutical Sciences, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | | | | | - Arif Ali
- Post-Graduate Program in Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Márcia Machado Marinho
- Theoretical and Eletrochemical Chemistry Research Group, State University of Ceará, Limoeiro do Norte, CE, Brazil; State University of Vale do Acaraú, Center for Exact Sciences and Technology, Sobral, CE, Brazil
| | - Emmanuel Silva Marinho
- Theoretical and Eletrochemical Chemistry Research Group, State University of Ceará, Limoeiro do Norte, CE, Brazil
| | - Hélcio Silva Dos Santos
- State University of Vale do Acaraú, Center for Exact Sciences and Technology, Sobral, CE, Brazil
| | - Alice Maria Costa Martins
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | | |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Manchola Varón NC, Dos Santos GRRM, Colli W, Alves MJM. Interaction With the Extracellular Matrix Triggers Calcium Signaling in Trypanosoma cruzi Prior to Cell Invasion. Front Cell Infect Microbiol 2021; 11:731372. [PMID: 34671568 PMCID: PMC8521164 DOI: 10.3389/fcimb.2021.731372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/23/2021] [Indexed: 11/26/2022] Open
Abstract
Trypanosoma cruzi, the etiological agent of Chagas disease in humans, infects a wide variety of vertebrates. Trypomastigotes, the parasite infective forms, invade mammalian cells by a still poorly understood mechanism. Adhesion of tissue culture- derived trypomastigotes to the extracellular matrix (ECM) prior to cell invasion has been shown to be a relevant part of the process. Changes in phosphorylation, S-nitrosylation, and nitration levels of proteins, in the late phase of the interaction (2 h), leading to the reprogramming of both trypomastigotes metabolism and the DNA binding profile of modified histones, were described by our group. Here, the involvement of calcium signaling at a very early phase of parasite interaction with ECM is described. Increments in the intracellular calcium concentrations during trypomastigotes-ECM interaction depends on the Ca2+ uptake from the extracellular medium, since it is inhibited by EGTA or Nifedipine, an inhibitor of the L-type voltage gated Ca2+ channels and sphingosine-dependent plasma membrane Ca2+ channel, but not by Vanadate, an inhibitor of the plasma membrane Ca2+-ATPase. Furthermore, Nifedipine inhibits the invasion of host cells by tissue culture- derived trypomastigotes in a dose-dependent manner, reaching 95% inhibition at 100 µM Nifedipine. These data indicate the importance of both Ca2+ uptake from the medium and parasite-ECM interaction for host-cell invasion. Previous treatment of ECM with protease abolishes the Ca2+ uptake, further reinforcing the possibility that these events may be connected. The mitochondrion plays a relevant role in Ca2+ homeostasis in trypomastigotes during their interaction with ECM, as shown by the increment of the intracellular Ca2+ concentration in the presence of Antimycin A, in contrast to other calcium homeostasis disruptors, such as Cyclopiazonic acid for endoplasmic reticulum and Bafilomycin A for acidocalcisome. Total phosphatase activity in the parasite decreases in the presence of Nifedipine, EGTA, and Okadaic acid, implying a role of calcium in the phosphorylation level of proteins that are interacting with the ECM in tissue culture- derived trypomastigotes. In summary, we describe here the increment of Ca2+ at an early phase of the trypomastigotes interaction with ECM, implicating both nifedipine-sensitive Ca2+ channels in the influx of Ca2+ and the mitochondrion as the relevant organelle in Ca2+ homeostasis. The data unravel a complex sequence of events prior to host cell invasion itself.
Collapse
Affiliation(s)
- Nubia Carolina Manchola Varón
- Laboratory of Biochemistry of Parasites, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Walter Colli
- Laboratory of Biochemistry of Parasites, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Maria Julia M Alves
- Laboratory of Biochemistry of Parasites, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Chadha A, Chadee K. The NF-κB Pathway: Modulation by Entamoeba histolytica and Other Protozoan Parasites. Front Cell Infect Microbiol 2021; 11:748404. [PMID: 34595137 PMCID: PMC8476871 DOI: 10.3389/fcimb.2021.748404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Protozoan parasites have led to worldwide devastation because of their ability to cause infectious diseases. They have evolved as successful pathogens in part because of their remarkable and sophisticated ways to evade innate host defenses. This holds true for both intracellular and extracellular parasites that deploy multiple strategies to circumvent innate host defenses for their survival. The different strategies protozoan parasites use include hijacking the host cellular signaling pathways and transcription factors. In particular, the nuclear factor-κB (NF-κB) pathway seems to be an attractive target for different pathogens owing to their central role in regulating prompt innate immune responses in host defense. NF-κB is a ubiquitous transcription factor that plays an indispensable role not only in regulating immediate immune responses against invading pathogens but is also a critical regulator of cell proliferation and survival. The major immunomodulatory components include parasite surface and secreted proteins/enzymes and stimulation of host cells intracellular pathways and inflammatory caspases that directly or indirectly interfere with the NF-κB pathway to thwart immune responses that are directed for containment and/or elimination of the pathogen. To showcase how protozoan parasites exploits the NF-κB signaling pathway, this review highlights recent advances from Entamoeba histolytica and other protozoan parasites in contact with host cells that induce outside-in and inside-out signaling to modulate NF-κB in disease pathogenesis and survival in the host.
Collapse
Affiliation(s)
- Attinder Chadha
- Departments of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Kris Chadee
- Departments of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
9
|
Wiser MF. Unique Endomembrane Systems and Virulence in Pathogenic Protozoa. Life (Basel) 2021; 11:life11080822. [PMID: 34440567 PMCID: PMC8401336 DOI: 10.3390/life11080822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Virulence in pathogenic protozoa is often tied to secretory processes such as the expression of adhesins on parasite surfaces or the secretion of proteases to assisted in tissue invasion and other proteins to avoid the immune system. This review is a broad overview of the endomembrane systems of pathogenic protozoa with a focus on Giardia, Trichomonas, Entamoeba, kinetoplastids, and apicomplexans. The focus is on unique features of these protozoa and how these features relate to virulence. In general, the basic elements of the endocytic and exocytic pathways are present in all protozoa. Some of these elements, especially the endosomal compartments, have been repurposed by the various species and quite often the repurposing is associated with virulence. The Apicomplexa exhibit the most unique endomembrane systems. This includes unique secretory organelles that play a central role in interactions between parasite and host and are involved in the invasion of host cells. Furthermore, as intracellular parasites, the apicomplexans extensively modify their host cells through the secretion of proteins and other material into the host cell. This includes a unique targeting motif for proteins destined for the host cell. Most notable among the apicomplexans is the malaria parasite, which extensively modifies and exports numerous proteins into the host erythrocyte. These modifications of the host erythrocyte include the formation of unique membranes and structures in the host erythrocyte cytoplasm and on the erythrocyte membrane. The transport of parasite proteins to the host erythrocyte involves several unique mechanisms and components, as well as the generation of compartments within the erythrocyte that participate in extraparasite trafficking.
Collapse
Affiliation(s)
- Mark F Wiser
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
10
|
Maldonado E, Rojas DA, Urbina F, Solari A. T. cruzi DNA polymerase beta (Tcpolβ) is phosphorylated in vitro by CK1, CK2 and TcAUK1 leading to the potentiation of its DNA synthesis activity. PLoS Negl Trop Dis 2021; 15:e0009588. [PMID: 34260580 PMCID: PMC8312956 DOI: 10.1371/journal.pntd.0009588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/26/2021] [Accepted: 06/23/2021] [Indexed: 11/19/2022] Open
Abstract
The unicellular protozoan Trypanosoma cruzi is the causing agent of Chagas disease which affects several millions of people around the world. The components of the cell signaling pathways in this parasite have not been well studied yet, although its genome can encode several components able to transduce the signals, such as protein kinases and phosphatases. In a previous work we have found that DNA polymerase β (Tcpolβ) can be phosphorylated in vivo and this modification activates the synthesis activity of the enzyme. Tcpolβ is kinetoplast-located and is a key enzyme in the DNA base excision repair (BER) system. The polypeptide possesses several consensus phosphorylation sites for several protein kinases, however, a direct phosphorylation of those sites by specific kinases has not been reported yet. Tcpolβ has consensus phosphorylation sites for casein kinase 1 (CK1), casein kinase 2 (CK2) and aurora kinase (AUK). Genes encoding orthologues of those kinases exist in T. cruzi and we were able to identify the genes and to express them to investigate whether or no Tcpolβ could be a substrate for in vitro phosphorylation by those kinases. Both CK1 and TcAUK1 have auto-phosphorylation activities and they are able to phosphorylate Tcpolβ. CK2 cannot perform auto-phosphorylation of its subunits, however, it was able to phosphorylate Tcpolβ. Pharmacological inhibitors used to inhibit the homologous mammalian kinases can also inhibit the activity of T. cruzi kinases, although, at higher concentrations. The phosphorylation events carried out by those kinases can potentiate the DNA polymerase activity of Tcpolβ and it is discussed the role of the phosphorylation on the DNA polymerase and lyase activities of Tcpolβ. Taken altogether, indicates that CK1, CK2 and TcAUK1 can play an in vivo role regulating the function of Tcpolβ.
Collapse
Affiliation(s)
- Edio Maldonado
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- * E-mail: (EM); (AS)
| | - Diego A. Rojas
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Fabiola Urbina
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Aldo Solari
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- * E-mail: (EM); (AS)
| |
Collapse
|
11
|
Li SH, Li SD, Wu KL, Li JY, Li HJ, Wang WQ, Yang LJ, Xu JJ, Chang GJ, Zhang YL, Shu QH, Zhuang SS, Ma ZQ, He SM, Zhu M, Wang WL, Huang HL. Transcriptome Analysis Reveals Possible Virulence Factors of Paragonimus proliferus. Curr Bioinform 2021. [DOI: 10.2174/1574893615999200728203648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Objective:
To identify the possible virulence factors (VFs) of P. proliferus.
Methods:
By Illumina HiSeq 4000 RNA-Seq platform, transcriptomes of adult P. proliferus
worms were sequenced to predict VFs via screening the homologues of traditional VFs of parasites
based on the annotations in the functional databases. Homology analysis was also performed to
screen homologous genes between P. proliferus and other four Paragonimus species (i.e., P.
kellicotti, P. skrjabini, P. miyazakii and P. westermani) whose transcriptomes were downloaded
from the National Center for Biotechnology Information (NCBI) database, and then the
differential-expressed homologous genes (DEHGs) were screened via comparisons of P. proliferus
and P. kellicotti, P. skrjabini, P. miyazakii and P. westermani, respectively. Finally, an overlap of
the predicted VFs and DEHGs was performed to identify possible key VFs that do not only belong
to the predicted VFs but also DEHGs.
Results:
A total of 1,509 genes of P. proliferus homologous to traditional VFs, including surface
antigens (SAGs), secreted proteins (SPs), ATP-Binding Cassette (ABC) Transporters, actin-related
proteins (ARPs), aminopeptidases (APases), glycoproteins (GPs), cysteine proteases (CPs), and
heat shock proteins (HSPs), were identified. Meanwhile, homology analysis identified 6279
DEHGs among the five species, of which there were 48 DEHGs being mutually differentialexpressed
among the four pairs of comparisons, such as MRP, Tuba 3, PI3K, WASF2, ADK,
Nop56, DNAH1, PFK-2/FBPase2, Ppp1r7, SSP7. Furthermore, the overlap between the predicted
VFs and DEHGs showed 97 genes of the predicted VFs that simultaneously belonged to DEHGs.
Strikingly, of these 97 genes, only 26, including Chymotrypsin, Leucine APases, Cathepsin L, HSP
70, and so on, were higher expressed in P. proliferus while all the remaining were lower expressed
than in the four other species.
Conclusions:
This work provides a fundamental context for further studies of the pathogenicity of
P. proliferus. Most of the predicted VFs which simultaneously belonged to DEHGs were lower
expressed in P. proliferus.
Collapse
Affiliation(s)
- Sheng-Hao Li
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Shu-De Li
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Kun-Li Wu
- Department of Hepatology, Oncology, Infectious Disease, Health Care or Tuberculosis, the Third People’s Hospital of Kunming, Kunming 650043, China
| | - Jun-Yi Li
- Department of Hepatology, Oncology, Infectious Disease, Health Care or Tuberculosis, the Third People’s Hospital of Kunming, Kunming 650043, China
| | - Hong-Juan Li
- Department of Hepatology, Oncology, Infectious Disease, Health Care or Tuberculosis, the Third People’s Hospital of Kunming, Kunming 650043, China
| | - Wei-Qun Wang
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Li-Jun Yang
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Jing-Jing Xu
- Department of Hepatology, Oncology, Infectious Disease, Health Care or Tuberculosis, the Third People’s Hospital of Kunming, Kunming 650043, China
| | - Guo-Ji Chang
- Department of Hepatology, Oncology, Infectious Disease, Health Care or Tuberculosis, the Third People’s Hospital of Kunming, Kunming 650043, China
| | - Yan-Ling Zhang
- Department of Hepatology, Oncology, Infectious Disease, Health Care or Tuberculosis, the Third People’s Hospital of Kunming, Kunming 650043, China
| | - Qiu-Hong Shu
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Shan-Shan Zhuang
- Department of Clinical Laboratory, Yan’an Hospital of Kunming, Kunming 650000, China
| | - Zhi-Qiang Ma
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Shu-Meiqi He
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Min Zhu
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Wen-Lin Wang
- School of Basic Medicine, Kunming Medical University, Kunming 650504, China
| | - Hong-Li Huang
- Department of Hepatology, Oncology, Infectious Disease, Health Care or Tuberculosis, the Third People’s Hospital of Kunming, Kunming 650043, China
| |
Collapse
|
12
|
Nájera CA, Batista MF, Meneghelli I, Bahia D. Mixed signals - how Trypanosoma cruzi exploits host-cell communication and signaling to establish infection. J Cell Sci 2021; 134:134/5/jcs255687. [PMID: 33692153 DOI: 10.1242/jcs.255687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chagas disease (American trypanosomiasis) is a 'neglected' pathology that affects millions of people worldwide, mainly in Latin America. Trypanosoma cruzi, the causative agent, is an obligate intracellular parasite with a complex and diverse biology that infects several mammalian species, including humans. Because of genetic variability among strains and the presence of four biochemically and morphologically distinct parasite forms, the outcome of T. cruzi infection varies considerably depending on host cell type and parasite strain. During the initial contact, cellular communication is established by host-recognition-mediated responses, followed by parasite adherence and penetration. For this purpose, T. cruzi expresses a variety of proteins that modify the host cell, enabling it to safely reach the cytoplasm. After entry into the host cell, T. cruzi forms a transitory structure termed 'parasitophorous vacuole' (PV), followed by its cytoplasmic replication and differentiation after PV rupture, and subsequent invasion of other cells. The success of infection, maintenance and survival inside host cells is facilitated by the ability of T. cruzi to subvert various host signaling mechanisms. We focus in this Review on the various mechanisms that induce host cytoskeletal rearrangements, activation of autophagy-related proteins and crosstalk among major immune response regulators, as well as recent studies on the JAK-STAT pathway.
Collapse
Affiliation(s)
- Carlos Acides Nájera
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Marina Ferreira Batista
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Isabela Meneghelli
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Diana Bahia
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| |
Collapse
|
13
|
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: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
14
|
Torrecilhas AC, Soares RP, Schenkman S, Fernández-Prada C, Olivier M. Extracellular Vesicles in Trypanosomatids: Host Cell Communication. Front Cell Infect Microbiol 2020; 10:602502. [PMID: 33381465 PMCID: PMC7767885 DOI: 10.3389/fcimb.2020.602502] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022] Open
Abstract
Trypanosoma cruzi, Trypanosoma brucei and Leishmania (Trypanosomatidae: Kinetoplastida) are parasitic protozoan causing Chagas disease, African Trypanosomiasis and Leishmaniases worldwide. They are vector borne diseases transmitted by triatomine bugs, Tsetse fly, and sand flies, respectively. Those diseases cause enormous economic losses and morbidity affecting not only rural and poverty areas but are also spreading to urban areas. During the parasite-host interaction, those organisms release extracellular vesicles (EVs) that are crucial for the immunomodulatory events triggered by the parasites. EVs are involved in cell-cell communication and can act as important pro-inflammatory mediators. Therefore, interface between EVs and host immune responses are crucial for the immunopathological events that those diseases exhibit. Additionally, EVs from these organisms have a role in the invertebrate hosts digestive tracts prior to parasite transmission. This review summarizes the available data on how EVs from those medically important trypanosomatids affect their interaction with vertebrate and invertebrate hosts.
Collapse
Affiliation(s)
- Ana Claudia Torrecilhas
- Departamento de Ciências Farmacêuticas, Federal University of Sao Paulo (UNIFESP), Diadema, Brazil
| | | | - Sergio Schenkman
- Departamento de Microbiologia, Imunologia e Parasitologia, UNIFESP, São Paulo, Brazil
| | | | - Martin Olivier
- The Research Institute of the McGill University Health Centre, McGill University, Montréal, QC, Canada
| |
Collapse
|
15
|
Update on relevant trypanosome peptidases: Validated targets and future challenges. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1869:140577. [PMID: 33271348 DOI: 10.1016/j.bbapap.2020.140577] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/09/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Trypanosoma cruzi, the agent of the American Trypanosomiasis, Chagas disease, and Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense, the agents of Sleeping sickness (Human African Trypanosomiasis, HAT), as well as Trypanosoma brucei brucei, the agent of the cattle disease nagana, contain cysteine, serine, threonine, aspartyl and metallo peptidases. The most abundant among these enzymes are the cysteine proteases from the Clan CA, the Cathepsin L-like cruzipain and rhodesain, and the Cathepsin B-like enzymes, which have essential roles in the parasites and thus are potential targets for chemotherapy. In addition, several other proteases, present in one or both parasites, have been characterized, and some of them are also promising candidates for the developing of new drugs. Recently, new inhibitors, with good selectivity for the parasite proteasomes, have been described and are very promising as lead compounds for the development of new therapies for these neglected diseases. This article is part of a Special Issue entitled: "Play and interplay of proteases in health and disease".
Collapse
|
16
|
The Glycan Structure of T. cruzi mucins Depends on the Host. Insights on the Chameleonic Galactose. Molecules 2020; 25:molecules25173913. [PMID: 32867240 PMCID: PMC7504415 DOI: 10.3390/molecules25173913] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/23/2022] Open
Abstract
Trypanosoma cruzi, the protozoa that causes Chagas disease in humans, is transmitted by insects from the Reduviidae family. The parasite has developed the ability to change the structure of the surface molecules, depending on the host. Among them, the mucins are the most abundant glycoproteins. Structural studies have focused on the epimastigotes and metacyclic trypomastigotes that colonize the insect, and on the mammal trypomastigotes. The carbohydrate in the mucins fulfills crucial functions, the most important of which being the accepting of sialic acid from the host, a process catalyzed by the unique parasite trans-sialidase. The sialylation of the parasite influences the immune response on infection. The O-linked sugars have characteristics that differentiate them from human mucins. One of them is the linkage to the polypeptide chain by the hexosamine, GlcNAc, instead of GalNAc. The main monosaccharide in the mucins oligosaccharides is galactose, and this may be present in three configurations. Whereas β-d-galactopyranose (β-Galp) was found in the insect and the human stages of Trypanosoma cruzi, β-d-galactofuranose (β-Galf) is present only in the mucins of some strains of epimastigotes and α-d-galactopyranose (α-Galp) characterizes the mucins of the bloodstream trypomastigotes. The two last configurations confer high antigenic properties. In this review we discuss the different structures found and we pose the questions that still need investigation on the exchange of the configurations of galactose.
Collapse
|
17
|
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
|
18
|
Rose E, Carvalho JL, Hecht M. Mechanisms of DNA repair in Trypanosoma cruzi: What do we know so far? DNA Repair (Amst) 2020; 91-92:102873. [PMID: 32505694 DOI: 10.1016/j.dnarep.2020.102873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/27/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022]
Abstract
Trypanosoma cruzi is the etiological agent of Chagas Disease, which affects 6-7 million people worldwide. Since the early stages of infection and throughout its life cycle, the parasite is exposed to several genotoxic agents. Furthermore, DNA damage is also part of the mechanism of action of at least a few trypanocidal drugs, including Benznidazole. Thus, it is paramount for the parasite to count on an efficient DNA repair machinery to guarantee genome integrity and survival. The present work provides an up-to-date review of both the conserved and peculiar DNA repair mechanisms described in T. cruzi against oxidative stress, ultraviolet and ionizing radiation, DNA adduct-inducing agents, and Benznidazole. The comprehension of the DNA repair mechanisms of the parasite may shed light on the parasite evolution and possibly pave the way for the development of novel and more effective trypanocidal drugs.
Collapse
Affiliation(s)
- Ester Rose
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil.
| | - Juliana Lott Carvalho
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil; Genomic Sciences and Biotechnology Program, Catholic University of Brasília, Brasília, Brazil
| | - Mariana Hecht
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| |
Collapse
|
19
|
Quebrada Palacio LP, Fernández ER, Hernández-Vásquez Y, Petray PB, Postan M. Circulating T Follicular Helper Cell Abnormalities Associated to Different Clinical Forms of Chronic Chagas Disease. Front Cell Infect Microbiol 2020; 10:126. [PMID: 32296649 PMCID: PMC7136390 DOI: 10.3389/fcimb.2020.00126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/06/2020] [Indexed: 12/31/2022] Open
Abstract
Multiple perturbations of the immune response affecting a range of cells have been reported in Trypanosoma cruzi-infected individuals and associated to clinical manifestations of chronic Chagas disease. There is a paucity of knowledge about the role of T follicular helper (Tfh) cells in this infection. Here, we sought to characterize circulating Tfh (cTfh) cells in chronic Chagas disease patients and to identify potential associations with disease severity in humans. cTfh cells were characterized by flow cytometry in freshly isolated PBMCs from 7 T. cruzi-infected asymptomatic patients (ASYMP), 5 patients with chronic chagasic dilated cardiomyopathy (CCC) and 8 healthy controls, using antibodies against chemokine receptors CXCR5, CXCR3, CCR6, and CCR7. Our results showed significant expansion of CD4+CD45RO+CXCR5+CCR6+ cells in ASYMP and CCC patients, along with a contraction of CD4+CD45RO+CXCR5+CXCR3-CCR6- (cTfh2) cells. ASYMP patients further exhibited decreased CD4+CD45RO+CXCR5+CXCR3+CCR6- (cTfh1) cells and expanded CD4+CD45RO+CXCR5+CXCR3-CCR6+ (cTfh17) cells while CCC patients exhibited significantly increased frequencies of CD4+CD45RO+CXCR5+CCR7+ cells. Linear regression analysis revealed a positive trend of CD4+CD45RO+CXCR5+CXCR3+CCR6+ (cTfh1/17) cells and negative trends of cTfh1 and cTfh2 cells as disease was more severe. There was no correlation between the frequencies of cTfh cells and circulating CD19+IgD-IgG+ cells or serum levels of T. cruzi-specific IgG. These results demonstrate that the cTfh compartment of humans chronically infected with T. cruzi comprises expanded CCR6-expressing cells and reduced cTfh2 cells. The association of discrete phenotypic changes in cTfh subsets with different clinical forms suggests the potential contribution of T follicular helper cells to Chagas heart disease progression.
Collapse
Affiliation(s)
- Luz P Quebrada Palacio
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS/Malbran, Buenos Aires, Argentina
| | - Esteban R Fernández
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS/Malbran, Buenos Aires, Argentina
| | - Yolanda Hernández-Vásquez
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS/Malbran, Buenos Aires, Argentina
| | - Patricia B Petray
- Facultad de Medicina, IMPaM-Instituto de Investigaciones en Microbiología y Parasitología Médica (UBA-CONICET), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Miriam Postan
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS/Malbran, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
20
|
Sangenito LS, Menna-Barreto RFS, d'Avila-Levy CM, Branquinha MH, Santos ALS. Repositioning of HIV Aspartyl Peptidase Inhibitors for Combating the Neglected Human Pathogen Trypanosoma cruzi. Curr Med Chem 2019; 26:6590-6613. [PMID: 31187704 DOI: 10.2174/0929867326666190610152934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/11/2018] [Accepted: 08/23/2018] [Indexed: 12/11/2022]
Abstract
Chagas disease, caused by the flagellate parasite Trypanosoma cruzi, is a wellknown neglected tropical disease. This parasitic illness affects 6-7 million people and can lead to severe myocarditis and/or complications of the digestive tract. The changes in its epidemiology facilitate co-infection with the Human Immunodeficiency Virus (HIV), making even more difficult the diagnosis and prognosis. The parasitic infection is reactivated in T. cruzi/HIV co-infection, with the appearance of unusual manifestations in the chronic phase and the exacerbation of classical clinical signs. The therapeutic arsenal to treat Chagas disease, in all its clinical forms, is restricted basically to two drugs, benznidazole and nifurtimox. Both drugs are extremely toxic and the therapeutic efficacy is still unclear, making the clinical treatment a huge issue to be solved. Therefore, it seems obvious the necessity of new tangible approaches to combat this illness. In this sense, the repositioning of approved drugs appears as an interesting and viable strategy. The discovery of Human Immunodeficiency Virus Aspartyl Peptidase Inhibitors (HIV-PIs) represented a milestone in the treatment of Acquired Immune Deficiency Syndrome (AIDS) and, concomitantly, a marked reduction in both the incidence and prevalence of important bacterial, fungal and parasitic co-infections was clearly observed. Taking all these findings into consideration, the present review summarizes the promising and beneficial data concerning the effects of HIV-PIs on all the evolutionary forms of T. cruzi and in important steps of the parasite's life cycle, which highlight their possible application as alternative drugs to treat Chagas disease.
Collapse
Affiliation(s)
- Leandro S Sangenito
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Rubem F S Menna-Barreto
- Laboratorio de Biologia Celular, Instituto Oswaldo Cruz (IOC), Fundacao Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Cláudia M d'Avila-Levy
- Laboratorio de Estudos Integrados em Protozoologia, Instituto Oswaldo Cruz (IOC), Fundacao Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Marta H Branquinha
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - André L S Santos
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
21
|
da Rocha RFDB, LaRocque-de-Freitas IF, Arcanjo AF, Logullo J, Nunes MP, Freire-de-Lima CG, Decote-Ricardo D. B-1 Cells May Drive Macrophages Susceptibility to Trypanosoma cruzi Infection. Front Microbiol 2019; 10:1598. [PMID: 31338088 PMCID: PMC6629875 DOI: 10.3389/fmicb.2019.01598] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/26/2019] [Indexed: 12/16/2022] Open
Abstract
B-1 cells can directly and indirectly influence the immune response. These cells are known to be excellent producers of natural antibodies and can secrete a variety of immunomodulatory molecules. They are also able to differentiate into B-1 cell-derived phagocytes (B-1CDP). B-1 cells can modulate macrophages to become less effective, and B-1CDP cells are more susceptible in infection models. In this work, we investigated the microbicidal ability of these cells in Trypanosoma cruzi infection in vitro. The results show that macrophages from BALB/c mice are more susceptible to infection than macrophages from XID mice. The resistance observed in macrophages from XID mice was abolished in the presence of B-1 cells, and this event seems to be associated with IL-10 production by B-1 cells, which may have contributed to the decrease of NO production. Additionally, B-1CDP cells were more permissive to intracellular T. cruzi infection than peritoneal macrophages. These findings strongly suggest that B-1 cells and B-1CDP cells have a potential role in the persistence of the parasite in host cells.
Collapse
Affiliation(s)
| | | | - Angelica Fernandes Arcanjo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorgete Logullo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Debora Decote-Ricardo
- Instituto de Veterinária, Universidade Federal Rural do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Espinosa-Pereiro J, Sánchez-Montalvá A, Salvador F, Sao-Avilés A, Sulleiro E, Molina I. A retrospective study on the influence of siblings' relatedness in Bolivian patients with chronic Chagas disease. Parasit Vectors 2019; 12:260. [PMID: 31126327 PMCID: PMC6533702 DOI: 10.1186/s13071-019-3518-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 05/20/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chagas disease is a protozoan infection caused by Trypanosoma cruzi. The disease has a chronic course in which 20-30% of the patients would develop progressive damage to the cardiovascular system and the gastrointestinal tube. We are still unable to predict who will develop end-organ damage but there are some acquired and genetic risk factors already known. RESULTS We reviewed data from 833 patients with serologically confirmed Chagas disease in this retrospective study. Patients were classified as siblings or non-siblings (controls) and the results of pre-treatment blood PCR assay, end-organ damage (cardiac and/or gastrointestinal), and the presence of delayed type hypersensitivity (DTH) skin involvement in patients treated with benznidazole were analyzed. Siblings were grouped by family and we randomly generated groups of 2 or 3 persons with the remaining controls. We classified the results of each variable as concordant or discordant and compared the concordance in these results among the sibling groups with that among control groups. We identified 71 groups of siblings and randomly generated 299 groups of non-related patients. Pre-treatment blood PCR concordance was significantly higher (19%) among siblings compared to controls (P = 0.02), probably due to a higher frequency in pre-treatment positive results. No other statistically significant differences were found. CONCLUSIONS A significant difference was found in the concordance of pre-treatment blood PCR for T. cruzi among siblings compared to non-related controls.
Collapse
Affiliation(s)
- Juan Espinosa-Pereiro
- Infectious Diseases Department, Vall d’Hebron University Hospital, Programa de Salut Internacional de l’Institut Català de la Salut (PROSICS), Barcelona, Spain
| | - Adrián Sánchez-Montalvá
- Infectious Diseases Department, Vall d’Hebron University Hospital, Programa de Salut Internacional de l’Institut Català de la Salut (PROSICS), Barcelona, Spain
- Vall d’Hebron Resarch Institute, University Hospital Vall d’Hebron, Barcelona, Spain
| | - Fernando Salvador
- Infectious Diseases Department, Vall d’Hebron University Hospital, Programa de Salut Internacional de l’Institut Català de la Salut (PROSICS), Barcelona, Spain
- Vall d’Hebron Resarch Institute, University Hospital Vall d’Hebron, Barcelona, Spain
| | - Augusto Sao-Avilés
- Infectious Diseases Department, Vall d’Hebron University Hospital, Programa de Salut Internacional de l’Institut Català de la Salut (PROSICS), Barcelona, Spain
- Cardiac Imaging Unit, University Hospital Vall d’Hebron, Barcelona, Spain
| | - Elena Sulleiro
- Microbiology Department, University Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Israel Molina
- Infectious Diseases Department, Vall d’Hebron University Hospital, Programa de Salut Internacional de l’Institut Català de la Salut (PROSICS), Barcelona, Spain
- Vall d’Hebron Resarch Institute, University Hospital Vall d’Hebron, Barcelona, Spain
| |
Collapse
|
23
|
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
|
24
|
Ferrer MJ, Wehrendt DP, Bonilla M, Comini MA, Tellez-Iñón MT, Potenza M. Production of Recombinant Trypanosoma cruzi Antigens in Leishmania tarentolae. Methods Mol Biol 2019; 1955:105-118. [PMID: 30868522 DOI: 10.1007/978-1-4939-9148-8_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Trypanosomatids are unicellular organisms that colonize a wide diversity of environments and hosts. For instance, Trypanosoma cruzi is a human pathogen responsible for Chagas diseases, while Leishmania tarentolae infects amphibians and became a biotechnological tool suitable for recombinant protein expression. T. cruzi antigens are needed for the development of improved epitope-based methods for diagnosis and treatment of Chagas disease. Molecular cloning for the production of recombinant proteins offers the possibility to obtain T. cruzi antigens at high yield and purity. L. tarentolae appears as the ideal expression host to obtain recombinant T. cruzi antigens with a structure and posttranslational modifications typical of trypanosomatids. In this chapter, we present a protocol for the analytical to mid-scale production of recombinant T. cruzi antigens, using L. tarentolae as expression host (LEXSY® inducible system).
Collapse
Affiliation(s)
- María José Ferrer
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, "Dr. Héctor Torres" (INGEBI-CONICET), Buenos Aires, Argentina
| | - Diana Patricia Wehrendt
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, "Dr. Héctor Torres" (INGEBI-CONICET), Buenos Aires, Argentina
| | - Mariana Bonilla
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Marcelo Alberto Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - María Teresa Tellez-Iñón
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, "Dr. Héctor Torres" (INGEBI-CONICET), Buenos Aires, Argentina
| | - Mariana Potenza
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, "Dr. Héctor Torres" (INGEBI-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
25
|
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]
|
26
|
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
|
27
|
Martins-da-Silva A, Telleria EL, Batista M, Marchini FK, Traub-Csekö YM, Tempone AJ. Identification of Secreted Proteins Involved in Nonspecific dsRNA-Mediated Lutzomyia longipalpis LL5 Cell Antiviral Response. Viruses 2018; 10:v10010043. [PMID: 29346269 PMCID: PMC5795456 DOI: 10.3390/v10010043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 02/07/2023] Open
Abstract
Hematophagous insects transmit infectious diseases. Sand flies are vectors of leishmaniasis, but can also transmit viruses. We have been studying immune responses of Lutzomyia longipalpis, the main vector of visceral leishmaniasis in the Americas. We identified a non-specific antiviral response in L. longipalpis LL5 embryonic cells when treated with non-specific double-stranded RNAs (dsRNAs). This response is reminiscent of interferon response in mammals. We are investigating putative effectors for this antiviral response. Secreted molecules have been implicated in immune responses, including interferon-related responses. We conducted a mass spectrometry analysis of conditioned medium from LL5 cells 24 and 48 h after dsRNA or mock treatment. We identified 304 proteins. At 24 h, 19 proteins had an abundance equal or greater than 2-fold change, while the levels of 17 proteins were reduced when compared to control cells. At the 48 h time point, these numbers were 33 and 71, respectively. The two most abundant secreted peptides at 24 h in the dsRNA-transfected group were phospholipid scramblase, an interferon-inducible protein that mediates antiviral activity, and forskolin-binding protein (FKBP), a member of the immunophilin family, which mediates the effect of immunosuppressive drugs. The transcription profile of most candidates did not follow the pattern of secreted protein abundance.
Collapse
Affiliation(s)
- Andrea Martins-da-Silva
- Laboratório de Biologia Molecular de Parasitas e Vetores, Instituto Oswaldo Cruz-Fiocruz, Av. Brasil 4365, Rio de Janeiro 21040-360, RJ, Brazil.
| | - Erich Loza Telleria
- Laboratório de Biologia Molecular de Parasitas e Vetores, Instituto Oswaldo Cruz-Fiocruz, Av. Brasil 4365, Rio de Janeiro 21040-360, RJ, Brazil.
| | - Michel Batista
- Laboratório de Genômica Funcional, Instituto Carlos Chagas-Fiocruz, Rua Prof. Algacyr Munhoz Mader 3775, Curitiba 81350-010, PR, Brazil.
- Plataforma Espectrometria de Massas-RPT02H, Instituto Carlos Chagas-Fiocruz, Rua Prof. Algacyr Munhoz Mader 3775, Curitiba 81350-010, PR, Brazil.
| | - Fabricio Klerynton Marchini
- Laboratório de Genômica Funcional, Instituto Carlos Chagas-Fiocruz, Rua Prof. Algacyr Munhoz Mader 3775, Curitiba 81350-010, PR, Brazil.
- Plataforma Espectrometria de Massas-RPT02H, Instituto Carlos Chagas-Fiocruz, Rua Prof. Algacyr Munhoz Mader 3775, Curitiba 81350-010, PR, Brazil.
| | - Yara Maria Traub-Csekö
- Laboratório de Biologia Molecular de Parasitas e Vetores, Instituto Oswaldo Cruz-Fiocruz, Av. Brasil 4365, Rio de Janeiro 21040-360, RJ, Brazil.
| | - Antonio Jorge Tempone
- Laboratório de Biologia Molecular de Parasitas e Vetores, Instituto Oswaldo Cruz-Fiocruz, Av. Brasil 4365, Rio de Janeiro 21040-360, RJ, Brazil.
| |
Collapse
|
28
|
Lonien SCH, Malvezi AD, Suzukawa HT, Yamauchi LM, Yamada-Ogatta SF, Rizzo LV, Bordignon J, Pinge-Filho P. Response to Trypanosoma cruzi by Human Blood Cells Enriched with Dentritic Cells Is Controlled by Cyclooxygenase-2 Pathway. Front Microbiol 2017; 8:2020. [PMID: 29118740 PMCID: PMC5660982 DOI: 10.3389/fmicb.2017.02020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 10/03/2017] [Indexed: 12/14/2022] Open
Abstract
Chagas disease (Cd) or American human trypanosomiasis is caused by Trypanosoma cruzi and affects ~7 million people, mostly in Latin America. The infective trypomastigote forms of the parasite can invade several human blood cell populations, including monocytes and dendritic cells (DC). Although these cells display a wide functional diversity, their interactions with T. cruzi via cyclooxygenase (COX) and cyclic adenosine monophosphate (cAMP) dependent pathways have not been analyzed. To exploiting this mechanism, DC-enriched peripheral human blood mononuclear cell populations (DC-PBMC) were used as our model. Our results showed that the treatment of these cell populations with celecoxib (CEL), a cyclooxygenase-2 selective inhibitor or SQ 22,536, an adenilate cyclase inhibitor, significantly caused marked inhibition of T. cruzi infection. In contrast, aspirin (ASA, a non-selective COX-1 and COX-2 inhibitor) treatment did not inhibit the infection of the cells by the parasite and was independent of nitric oxide (NO) production. The expression of co-stimulatory molecules CD80 and CD86 were similar on cells treated or not with both COX-inhibitors. The infection stimulated the release of TNF-α, IL-1β, IL-6, IL-8, and IL-10 production by infected cells. Treatment with ASA or CEL did not affect TNF-α, IL-6, IL-8, IL-10, and NO production by infected cells, but increased IL-1β production by them. Our results suggest a key role of COX-2 and cAMP pathways in T. cruzi invasion process of human blood cells and these pathways may represent targets of new therapeutic options for Cd.
Collapse
Affiliation(s)
- Sandra C H Lonien
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Aparecida D Malvezi
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Helena T Suzukawa
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Lucy M Yamauchi
- Laboratório de Biologia Molecular de Microrganismos, Departamento de Microbiologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Sueli F Yamada-Ogatta
- Laboratório de Biologia Molecular de Microrganismos, Departamento de Microbiologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Luiz V Rizzo
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Juliano Bordignon
- Laboratório de Virologia Molecular, Instituto Carlos Chagas/Fiocruz, Curitiba, Brazil
| | - Phileno Pinge-Filho
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
29
|
Bahia D. A New Trick for a Conserved Enzyme: Mevalonate Kinase, a Glycosomal Enzyme, Can Be Secreted by Trypanosoma cruzi and Modulate Cell Invasion and Signaling. Is It Another Moonlighting Enzyme? Front Cell Infect Microbiol 2017; 7:426. [PMID: 29034216 PMCID: PMC5627032 DOI: 10.3389/fcimb.2017.00426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 09/15/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| |
Collapse
|
30
|
Urquiza JM, Burgos JM, Ojeda DS, Pascuale CA, Leguizamón MS, Quarleri JF. Astrocyte Apoptosis and HIV Replication Are Modulated in Host Cells Coinfected with Trypanosoma cruzi. Front Cell Infect Microbiol 2017; 7:345. [PMID: 28824880 PMCID: PMC5539089 DOI: 10.3389/fcimb.2017.00345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022] Open
Abstract
The protozoan Trypanosoma cruzi is the etiological agent of Chagas disease. In immunosuppressed individuals, as it occurs in the coinfection with human immunodeficiency virus (HIV), the central nervous system may be affected. In this regard, reactivation of Chagas disease is severe and often lethal, and it accounts for meningoencephalitis. Astrocytes play a crucial role in the environment maintenance of healthy neurons; however, they can host HIV and T. cruzi. In this report, human astrocytes were infected in vitro with both genetically modified-pathogens to express alternative fluorophore. As evidenced by fluorescence microscopy and flow cytometry, HIV and T. cruzi coexist in the same astrocyte, likely favoring reciprocal interactions. In this context, lower rates of cell death were observed in both T. cruzi monoinfected-astrocytes and HIV-T. cruzi coinfection in comparison with those infected only with HIV. The level of HIV replication is significantly diminished under T. cruzi coinfection, but without affecting the infectivity of the HIV progeny. This interference with viral replication appears to be related to the T. cruzi multiplication rate or its increased intracellular presence but does not require their intracellular cohabitation or infected cell-to-cell contact. Among several Th1/Th2/Th17 profile-related cytokines, only IL-6 was overexpressed in HIV-T. cruzi coinfection exhibiting its cytoprotective role. This study demonstrates that T. cruzi and HIV are able to coinfect astrocytes thus altering viral replication and apoptosis.
Collapse
Affiliation(s)
- Javier M Urquiza
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Instituto de Investigaciones Biomédicas en Retrovirus y Sida, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Juan M Burgos
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, San Martín, Argentina Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Diego S Ojeda
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Instituto de Investigaciones Biomédicas en Retrovirus y Sida, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Carla A Pascuale
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, San Martín, Argentina Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - M Susana Leguizamón
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, San Martín, Argentina Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Jorge F Quarleri
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Instituto de Investigaciones Biomédicas en Retrovirus y Sida, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| |
Collapse
|
31
|
Freire-de-Lima L, Gentile LB, da Fonseca LM, da Costa KM, Santos Lemos J, Jacques LR, Morrot A, Freire-de-Lima CG, Nunes MP, Takiya CM, Previato JO, Mendonça-Previato L. Role of Inactive and Active Trypanosoma cruzi Trans-sialidases on T Cell Homing and Secretion of Inflammatory Cytokines. Front Microbiol 2017; 8:1307. [PMID: 28744279 PMCID: PMC5504189 DOI: 10.3389/fmicb.2017.01307] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/28/2017] [Indexed: 12/29/2022] Open
Abstract
Trans-sialidase from Trypanosoma cruzi (Tc-TS) belongs to a superfamily of proteins that may have enzymatic activity. While enzymatically active members (Tc-aTS) are able to transfer sialic acid from the host cell sialyl-glycoconjugates onto the parasite or to other molecules on the host cell surface, the inactive members (Tc-iTS) are characterized by their lectinic properties. Over the last 10 years, several papers demonstrated that, individually, Tc-aTS or Tc-iTS is able to modulate several biological events. Since the genes encoding Tc-iTS and Tc-aTS are present in the same copy number, and both proteins portray similar substrate-specificities as well, it would be plausible to speculate that such molecules may compete for the same sialyl-glycan structures and govern numerous immunobiological phenomena. However, their combined effect has never been evaluated in the course of an acute infection. In this study, we investigated the ability of both proteins to modulate the production of inflammatory signals, as well as the homing of T cells to the cardiac tissue of infected mice, events that usually occur during the acute phase of T. cruzi infection. The results showed that the intravenous administration of Tc-iTS, but not Tc-aTS protected the cardiac tissue from injury caused by reduced traffic of inflammatory cells. In addition, the ability of Tc-aTS to modulate the production of inflammatory cytokines was attenuated and/or compromised when Tc-iTS was co-injected in the same proportions. These results suggest that although both proteins present structural similarities and compete for the same sialyl-glycan epitopes, they might present distinct immunomodulatory properties on T cells following T. cruzi infection.
Collapse
Affiliation(s)
- Leonardo Freire-de-Lima
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Luciana B Gentile
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Leonardo M da Fonseca
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Kelli M da Costa
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Jessica Santos Lemos
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Lucas Rodrigues Jacques
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Alexandre Morrot
- Instituto Oswaldo Cruz, Fundação Oswaldo CruzRio de Janeiro, Brazil.,Instituto de Microbiologia, Centro de Ciência da Saúde - Sala D1-035, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Célio G Freire-de-Lima
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Marise P Nunes
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil.,Instituto Oswaldo Cruz, Fundação Oswaldo CruzRio de Janeiro, Brazil
| | - Christina M Takiya
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Jose O Previato
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Lucia Mendonça-Previato
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
32
|
Lidani KCF, Bavia L, Ambrosio AR, de Messias-Reason IJ. The Complement System: A Prey of Trypanosoma cruzi. Front Microbiol 2017; 8:607. [PMID: 28473804 PMCID: PMC5397499 DOI: 10.3389/fmicb.2017.00607] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/24/2017] [Indexed: 12/27/2022] Open
Abstract
Trypanosoma cruzi is a protozoan parasite known to cause Chagas disease (CD), a neglected sickness that affects around 6-8 million people worldwide. Originally, CD was mainly found in Latin America but more recently, it has been spread to countries in North America, Asia, and Europe due the international migration from endemic areas. Thus, at present CD represents an important concern of global public health. Most of individuals that are infected by T. cruzi may remain in asymptomatic form all lifelong, but up to 40% of them will develop cardiomyopathy, digestive mega syndromes, or both. The interaction between the T. cruzi infective forms and host-related immune factors represents a key point for a better understanding of the physiopathology of CD. In this context, the complement, as one of the first line of host defense against infection was shown to play an important role in recognizing T. cruzi metacyclic trypomastigotes and in controlling parasite invasion. The complement consists of at least 35 or more plasma proteins and cell surface receptors/regulators, which can be activated by three pathways: classical (CP), lectin (LP), and alternative (AP). The CP and LP are mainly initiated by immune complexes or pathogen-associated molecular patterns (PAMPs), respectively, whereas AP is spontaneously activated by hydrolysis of C3. Once activated, several relevant complement functions are generated which include opsonization and phagocytosis of particles or microorganisms and cell lysis. An important step during T. cruzi infection is when intracellular trypomastigotes are release to bloodstream where they may be target by complement. Nevertheless, the parasite uses a sequence of events in order to escape from complement-mediated lysis. In fact, several T. cruzi molecules are known to interfere in the initiation of all three pathways and in the assembly of C3 convertase, a key step in the activation of complement. Moreover, T. cruzi promotes secretion of plasma membrane-derived vesicles from host cells, which prevent the activity of C3 convertase C4b2a and thereby may hinder complement. In this review, we aim to present an overview on the strategies used by T. cruzi in order to circumvent the activation of complement and, consequently, its biological effects.
Collapse
Affiliation(s)
| | | | | | - Iara J. de Messias-Reason
- Laboratory of Molecular Immunopathology, Clinical Hospital, Federal University of ParanáCuritiba, Brazil
| |
Collapse
|
33
|
Bayer-Santos E, Marini MM, da Silveira JF. Non-coding RNAs in Host-Pathogen Interactions: Subversion of Mammalian Cell Functions by Protozoan Parasites. Front Microbiol 2017; 8:474. [PMID: 28377760 PMCID: PMC5359270 DOI: 10.3389/fmicb.2017.00474] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 03/08/2017] [Indexed: 01/23/2023] Open
Abstract
Pathogens have evolved mechanisms to modulate host cell functions and avoid recognition and destruction by the host damage response. For many years, researchers have focused on proteins as the main effectors used by pathogens to hijack host cell pathways, but only recently with the development of deep RNA sequencing these molecules were brought to light as key players in infectious diseases. Protozoan parasites such as those from the genera Plasmodium, Toxoplasma, Leishmania, and Trypanosoma cause life-threatening diseases and are responsible for 1000s of deaths worldwide every year. Some of these parasites replicate intracellularly when infecting mammalian hosts, whereas others can survive and replicate extracellularly in the bloodstream. Each of these parasites uses specific evasion mechanisms to avoid being killed by the host defense system. An increasing number of studies have shown that these pathogens can transfer non-coding RNA molecules to the host cells to modulate their functions. This transference usually happens via extracellular vesicles, which are small membrane vesicles secreted by the microorganism. In this mini-review we will combine published work regarding several protozoan parasites that were shown to use non-coding RNAs in inter-kingdom communication and briefly discuss future perspectives in the field.
Collapse
Affiliation(s)
- Ethel Bayer-Santos
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo São Paulo, Brazil
| | - Marjorie M Marini
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| | - José F da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo São Paulo, Brazil
| |
Collapse
|
34
|
Goulart LR, da S. Ribeiro V, Costa-Cruz JM. Anti-parasitic Antibodies from Phage Display. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1053:155-171. [DOI: 10.1007/978-3-319-72077-7_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Macrophages Promote Oxidative Metabolism To Drive Nitric Oxide Generation in Response to Trypanosoma cruzi. Infect Immun 2016; 84:3527-3541. [PMID: 27698021 DOI: 10.1128/iai.00809-16] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 12/18/2022] Open
Abstract
Trypanosoma cruzi is the causative agent of chronic chagasic cardiomyopathy. Why macrophages (mφs), the early responders to infection, fail to achieve parasite clearance is not known. Mouse (RAW 264.7) and human (THP-1 and primary) mφs were infected for 3 h and 18 h with T. cruzi TcI isolates, SylvioX10/4 (SYL, virulent) and TCC (nonpathogenic), which represent mφ stimulation and infection states, respectively. Mφs incubated with lipopolysaccharide and gamma interferon (LPS/IFN-γ) and with interleukin-4 (IL-4) were used as controls. We monitored the cytokine profile (using enzyme-linked immunosorbent assay [ELISA]), reactive oxygen species (ROS; fluorescent probes), nitric oxide (·NO; Griess assay), and metabolic state using a custom-designed mitoxosome array and Seahorse XF24 Analyzer. LPS/IFN-γ treatment of mφs elicited a potent increase in production of tumor necrosis alpha (TNF-α) at 3 h and of ROS and ·NO by 18 h. Upon SYL infection, murine mφs elicited an inflammatory cytokine profile (TNF-α ≫ TGF-β + IL-10) and low levels of ·NO and ROS production. LPS/IFN-γ treatment resulted in the inhibition of oxidative metabolism at the gene expression and functional levels and a switch to the glycolytic pathway in mφs, while IL-4-treated mφs utilized oxidative metabolism to meet energy demands. SYL infection resulted in an intermediate functional metabolic state with increased mitoxosome gene expression and glycolysis, and IFN-γ addition shut down the oxidative metabolism in SYL-infected mφs. Further, TCC- and SYL-stimulated mφs exhibited similar levels of cell proliferation and production of TNF-α and ROS, while TCC-stimulated mφs exhibited up to 2-fold-higher levels of oxidative metabolism and ·NO production than SYL-infected mφs. Inhibiting ATP-coupled O2 consumption suppressed the ·NO generation in SYL-infected mφs. Mitochondrial oxygen consumption constitutes a mechanism for stimulating ·NO production in mφs during T. cruzi infection. Enhancing the oxidative metabolism provides an opportunity for increased ·NO production and pathogen clearance by mφs to limit disease progression.
Collapse
|
36
|
Gil-Jaramillo N, Motta FN, Favali CBF, Bastos IMD, Santana JM. Dendritic Cells: A Double-Edged Sword in Immune Responses during Chagas Disease. Front Microbiol 2016; 7:1076. [PMID: 27471496 PMCID: PMC4943928 DOI: 10.3389/fmicb.2016.01076] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/27/2016] [Indexed: 01/24/2023] Open
Abstract
Dendritic cells (DCs) are the most important member of the antigen presenting cells group due to their ability to recognize antigen at the infection site and their high specialized antigen internalization capacity. These cells have central role in connecting the innate and adaptive immune responses against Trypanosoma cruzi, the causative agent of Chagas disease. These first line defense cells modulate host immune response depending on type, maturation level, cytokine milieu and DC receptor involved in the interactions with T. cruzi, influencing the development of the disease clinic forms. Here, we present a review of DCs-T. cruzi interactions both in human and murine models, pointing out the parasite ability to manipulate DCs activity for the purpose of evading innate immune response and assuring its own survival and persistence.
Collapse
Affiliation(s)
- Natalia Gil-Jaramillo
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
| | - Flávia N. Motta
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
- Faculdade de Ceilândia, Universidade de BrasíliaBrasília, Brazil
| | - Cecília B. F. Favali
- Laboratório de Biologia do Gene, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
| | - Izabela M. D. Bastos
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
| | - Jaime M. Santana
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
| |
Collapse
|