1
|
Ngambia Freitas FS, De Vooght L, Njiokou F, Abeele JVD, Bossard G, Tchicaya B, Corrales RM, Ravel S, Geiger A, Berthier-Teyssedre D. Evaluation of two candidate molecules-TCTP and cecropin-on the establishment of Trypanosoma brucei gambiense into the gut of Glossina palpalis gambiensis. INSECT SCIENCE 2025. [PMID: 40090966 DOI: 10.1111/1744-7917.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 03/19/2025]
Abstract
Trypanosomiasis, transmitted by tsetse flies (Glossina spp.), poses a significant health threat in 36 sub-Saharan African countries. Current control methods targeting tsetse flies, while effective, allow reinfestation. This study investigates paratransgenesis, a novel strategy to engineer symbiotic bacteria in tsetse flies, Sodalis glossinidius, to deliver anti-trypanosome compounds. Disrupting the trypanosome life cycle within the fly and reducing parasite transmission could offer a sustainable solution for trypanosomiasis control. In this context, we tested the effect of cecropin, reported to be lethal for Trypanosoma cruzi (Chagas disease) and TbgTCTP (Translationally Controlled Tumor Protein from Trypanosoma brucei gambiense), previously reported to modulate the growth of bacteria isolated from the fly microbiome, to delay the first peak of parasitemia and the death of trypanosome-infected mice. We have successfully cloned and transfected the genes encoding the two proteins into Sodalis strains. These Sodalis recombinant strains (recSodalisTbgTCTP and recSodaliscecropin) have been then microinjected into the L3 larval stage of Glossina palpalis gambiensis flies. The stability of the cloned genes was checked up to the 20th day after microinjection of recSodalis. The rate of fly emergence from untreated pupae was 95%; it was reduced by nearly 50% due to the mechanical injury caused by microinjection. It decreased to nearly 7% when larvae were injected with recSodalisTbgTCTP, which suggests TCTP could have a lethal impact to larvae development. When challenged with T. brucei gambiense, a slightly lower, but statistically non-significant, infection rate was recorded in flies harboring recSodaliscecropin compared to control flies. The effect of recSodalisTbgTCTP could not be measured due to the very low rate of fly emergence after corresponding treatment of the larvae. The results do not allow to conclude on the effect of cecropin or TCTP, delivered by para-transgenesis into the fly's gut, on the fly infection by the trypanosome. Nevertheless, the results are encouraging insofar as the technical approach works on the couple G. p. gambiensis/T. brucei gambiense. The next step will be to optimize the system and test other targets chosen among the ESPs (Excreted-Secreted Proteins) of the trypanosome secretum, or the differentially expressed genes associated with the sensitivity/resistance of the fly to trypanosome infection.
Collapse
Affiliation(s)
- François Sougal Ngambia Freitas
- Laboratory of Parasitology and Ecology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
- IRD-CIRAD, UMR INTERTRYP, Univ Montpellier, Cirad, IRD, Montpellier, France
| | - Linda De Vooght
- Department of Biomedical Sciences, Unit of Veterinary Protozoology, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Flobert Njiokou
- Laboratory of Parasitology and Ecology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Jan Van Den Abeele
- Department of Biomedical Sciences, Unit of Veterinary Protozoology, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Géraldine Bossard
- IRD-CIRAD, UMR INTERTRYP, Univ Montpellier, Cirad, IRD, Montpellier, France
| | | | | | - Sophie Ravel
- IRD-CIRAD, UMR INTERTRYP, Univ Montpellier, Cirad, IRD, Montpellier, France
| | - Anne Geiger
- IRD-CIRAD, UMR INTERTRYP, Univ Montpellier, Cirad, IRD, Montpellier, France
| | | |
Collapse
|
2
|
Aranda-Chan V, Cárdenas-Guerra RE, Otero-Pedraza A, Pacindo-Cabrales EE, Flores-Pucheta CI, Montes-Flores O, Arroyo R, Ortega-López J. Insights into Peptidyl-Prolyl cis- trans Isomerases from Clinically Important Protozoans: From Structure to Potential Biotechnological Applications. Pathogens 2024; 13:644. [PMID: 39204244 PMCID: PMC11357558 DOI: 10.3390/pathogens13080644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
Peptidyl-prolyl cis/trans isomerases (PPIases) are present in a wide variety of microorganisms, including protozoan parasites such as Trypanosoma cruzi, Trypanosoma brucei, Trichomonas vaginalis, Leishmania major, Leishmania donovani, Plasmodium falciparum, Plasmodium vivax, Entamoeba histolytica, Giardia intestinalis, Cryptosporidium parvum, and Cryptosporidium hominis, all of which cause important neglected diseases. PPIases are classified as cyclophilins, FKBPs, or parvulins and play crucial roles in catalyzing the cis-trans isomerization of the peptide bond preceding a proline residue. This activity assists in correct protein folding. However, experimentally, the biological structure-function characterization of PPIases from these protozoan parasites has been poorly addressed. The recombinant production of these enzymes is highly relevant for this ongoing research. Thus, this review explores the structural diversity, functions, recombinant production, activity, and inhibition of protozoan PPIases. We also highlight their potential as biotechnological tools for the in vitro refolding of other recombinant proteins from these parasites. These applications are invaluable for the development of diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Verónica Aranda-Chan
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Rosa Elena Cárdenas-Guerra
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Alejandro Otero-Pedraza
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Esdras Enoc Pacindo-Cabrales
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Claudia Ivonne Flores-Pucheta
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Octavio Montes-Flores
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| | - Rossana Arroyo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico;
| | - Jaime Ortega-López
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN # 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, Mexico City 07360, Mexico; (V.A.-C.); (R.E.C.-G.); (A.O.-P.); (E.E.P.-C.); (C.I.F.-P.); (O.M.-F.)
| |
Collapse
|
3
|
Medeiros EG, Valente MR, Honorato L, Ferreira MDS, Mendoza SR, Gonçalves DDS, Martins Alcântara L, Gomes KX, Pinto MR, Nakayasu ES, Clair G, da Rocha IFM, dos Reis FCG, Rodrigues ML, Alves LR, Nimrichter L, Casadevall A, Guimarães AJ. Comprehensive characterization of extracellular vesicles produced by environmental (Neff) and clinical (T4) strains of Acanthamoeba castellanii. mSystems 2024; 9:e0122623. [PMID: 38717186 PMCID: PMC11237502 DOI: 10.1128/msystems.01226-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/01/2024] [Indexed: 06/19/2024] Open
Abstract
We conducted a comprehensive comparative analysis of extracellular vesicles (EVs) from two Acanthamoeba castellanii strains, Neff (environmental) and T4 (clinical). Morphological analysis via transmission electron microscopy revealed slightly larger Neff EVs (average = 194.5 nm) compared to more polydisperse T4 EVs (average = 168.4 nm). Nanoparticle tracking analysis (NTA) and dynamic light scattering validated these differences. Proteomic analysis of the EVs identified 1,352 proteins, with 1,107 common, 161 exclusive in Neff, and 84 exclusively in T4 EVs. Gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) mapping revealed distinct molecular functions and biological processes and notably, the T4 EVs enrichment in serine proteases, aligned with its pathogenicity. Lipidomic analysis revealed a prevalence of unsaturated lipid species in Neff EVs, particularly triacylglycerols, phosphatidylethanolamines (PEs), and phosphatidylserine, while T4 EVs were enriched in diacylglycerols and diacylglyceryl trimethylhomoserine, phosphatidylcholine and less unsaturated PEs, suggesting differences in lipid metabolism and membrane permeability. Metabolomic analysis indicated Neff EVs enrichment in glycerolipid metabolism, glycolysis, and nucleotide synthesis, while T4 EVs, methionine metabolism. Furthermore, RNA-seq of EVs revealed differential transcript between the strains, with Neff EVs enriched in transcripts related to gluconeogenesis and translation, suggesting gene regulation and metabolic shift, while in the T4 EVs transcripts were associated with signal transduction and protein kinase activity, indicating rapid responses to environmental changes. In this novel study, data integration highlighted the differences in enzyme profiles, metabolic processes, and potential origins of EVs in the two strains shedding light on the diversity and complexity of A. castellanii EVs and having implications for understanding host-pathogen interactions and developing targeted interventions for Acanthamoeba-related diseases.IMPORTANCEA comprehensive and fully comparative analysis of extracellular vesicles (EVs) from two Acanthamoeba castellanii strains of distinct virulence, a Neff (environmental) and T4 (clinical), revealed striking differences in their morphology and protein, lipid, metabolites, and transcripts levels. Data integration highlighted the differences in enzyme profiles, metabolic processes, and potential distinct origin of EVs from both strains, shedding light on the diversity and complexity of A. castellanii EVs, with direct implications for understanding host-pathogen interactions, disease mechanisms, and developing new therapies for the clinical intervention of Acanthamoeba-related diseases.
Collapse
Affiliation(s)
- Elisa Gonçalves Medeiros
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Michele Ramos Valente
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Leandro Honorato
- Departamento de Microbiologia Geral, Laboratório de Glicobiologia de Eucariotos, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Marina da Silva Ferreira
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Susana Ruiz Mendoza
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Diego de Souza Gonçalves
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lucas Martins Alcântara
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Kamilla Xavier Gomes
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Departamento de Microbiologia Geral, Laboratório de Glicobiologia de Eucariotos, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Marcia Ribeiro Pinto
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Ernesto S. Nakayasu
- Biological Science Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | | | - Flavia C. G. dos Reis
- Instituto Carlos Chagas, Fundação Oswaldo Cruz, Fiocruz, Curitiba, Paraná, Brazil
- Centro de Desenvolvimento Tecnológico em Saúde (CDTS), Fiocruz, Rio de Janeiro, Brazil
| | - Marcio L. Rodrigues
- Instituto Carlos Chagas, Fundação Oswaldo Cruz, Fiocruz, Curitiba, Paraná, Brazil
- Instituto de Microbiologia Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
| | - Lysangela R. Alves
- Instituto Carlos Chagas, Fundação Oswaldo Cruz, Fiocruz, Curitiba, Paraná, Brazil
| | - Leonardo Nimrichter
- Departamento de Microbiologia Geral, Laboratório de Glicobiologia de Eucariotos, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Rede Micologia RJ–Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Allan Jefferson Guimarães
- Departamento de Microbiologia e Parasitologia, Laboratório de Bioquímica e Imunologia das Micoses, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Rede Micologia RJ–Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Chiribao ML, Díaz-Viraqué F, Libisch MG, Batthyány C, Cunha N, De Souza W, Parodi-Talice A, Robello C. Paracrine Signaling Mediated by the Cytosolic Tryparedoxin Peroxidase of Trypanosoma cruzi. Pathogens 2024; 13:67. [PMID: 38251374 PMCID: PMC10818299 DOI: 10.3390/pathogens13010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Peroxiredoxins are abundant and ubiquitous proteins that participate in different cellular functions, such as oxidant detoxification, protein folding, and intracellular signaling. Under different cellular conditions, peroxiredoxins can be secreted by different parasites, promoting the induction of immune responses in hosts. In this work, we demonstrated that the cytosolic tryparedoxin peroxidase of Trypanosoma cruzi (cTXNPx) is secreted by epimastigotes and trypomastigotes associated with extracellular vesicles and also as a vesicle-free protein. By confocal microscopy, we show that cTXNPx can enter host cells by an active mechanism both through vesicles and as a recombinant protein. Transcriptomic analysis revealed that cTXNPx induces endoplasmic reticulum stress and interleukin-8 expression in epithelial cells. This analysis also suggested alterations in cholesterol metabolism in cTXNPx-treated cells, which was confirmed by immunofluorescence showing the accumulation of LDL and the induction of LDL receptors in both epithelial cells and macrophages. BrdU incorporation assays and qPCR showed that cTXNPx has a mitogenic, proliferative, and proinflammatory effect on these cells in a dose-dependent manner. Importantly, we also demonstrated that cTXNPx acts as a paracrine virulence factor, increasing the susceptibility to infection in cTXNPx-pretreated epithelial cells by approximately 40%. Although the results presented in this work are from in vitro studies and likely underestimate the complexity of parasite-host interactions, our work suggests a relevant role for this protein in establishing infection.
Collapse
Affiliation(s)
- María Laura Chiribao
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11000, Uruguay;
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - Florencia Díaz-Viraqué
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - María Gabriela Libisch
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Montevideo 11000, Uruguay;
| | - Narcisa Cunha
- Instituto de Biofísica Carlos Chagas Filho, Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (N.C.); (W.D.S.)
| | - Wanderley De Souza
- Instituto de Biofísica Carlos Chagas Filho, Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (N.C.); (W.D.S.)
| | - Adriana Parodi-Talice
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
- Sección Genética Evolutiva, Instituto de Biología, Facultad de Ciencias, Universidad de la República, Montevideo 11000, Uruguay
| | - Carlos Robello
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11000, Uruguay;
- Laboratorio de Interacciones Hospedero–Patógeno—UBM, Institut Pasteur Montevideo, Montevideo 11000, Uruguay; (F.D.-V.); (M.G.L.)
| |
Collapse
|
5
|
Morrison LJ, Steketee PC, Tettey MD, Matthews KR. Pathogenicity and virulence of African trypanosomes: From laboratory models to clinically relevant hosts. Virulence 2023; 14:2150445. [PMID: 36419235 DOI: 10.1080/21505594.2022.2150445] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
African trypanosomes are vector-borne protozoa, which cause significant human and animal disease across sub-Saharan Africa, and animal disease across Asia and South America. In humans, infection is caused by variants of Trypanosoma brucei, and is characterized by varying rate of progression to neurological disease, caused by parasites exiting the vasculature and entering the brain. Animal disease is caused by multiple species of trypanosome, primarily T. congolense, T. vivax, and T. brucei. These trypanosomes also infect multiple species of mammalian host, and this complexity of trypanosome and host diversity is reflected in the spectrum of severity of disease in animal trypanosomiasis, ranging from hyperacute infections associated with mortality to long-term chronic infections, and is also a main reason why designing interventions for animal trypanosomiasis is so challenging. In this review, we will provide an overview of the current understanding of trypanosome determinants of infection progression and severity, covering laboratory models of disease, as well as human and livestock disease. We will also highlight gaps in knowledge and capabilities, which represent opportunities to both further our fundamental understanding of how trypanosomes cause disease, as well as facilitating the development of the novel interventions that are so badly needed to reduce the burden of disease caused by these important pathogens.
Collapse
Affiliation(s)
- Liam J Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Pieter C Steketee
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Mabel D Tettey
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Keith R Matthews
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
6
|
Jamabo M, Mahlalela M, Edkins AL, Boshoff A. Tackling Sleeping Sickness: Current and Promising Therapeutics and Treatment Strategies. Int J Mol Sci 2023; 24:12529. [PMID: 37569903 PMCID: PMC10420020 DOI: 10.3390/ijms241512529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Human African trypanosomiasis is a neglected tropical disease caused by the extracellular protozoan parasite Trypanosoma brucei, and targeted for eradication by 2030. The COVID-19 pandemic contributed to the lengthening of the proposed time frame for eliminating human African trypanosomiasis as control programs were interrupted. Armed with extensive antigenic variation and the depletion of the B cell population during an infectious cycle, attempts to develop a vaccine have remained unachievable. With the absence of a vaccine, control of the disease has relied heavily on intensive screening measures and the use of drugs. The chemotherapeutics previously available for disease management were plagued by issues such as toxicity, resistance, and difficulty in administration. The approval of the latest and first oral drug, fexinidazole, is a major chemotherapeutic achievement for the treatment of human African trypanosomiasis in the past few decades. Timely and accurate diagnosis is essential for effective treatment, while poor compliance and resistance remain outstanding challenges. Drug discovery is on-going, and herein we review the recent advances in anti-trypanosomal drug discovery, including novel potential drug targets. The numerous challenges associated with disease eradication will also be addressed.
Collapse
Affiliation(s)
- Miebaka Jamabo
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Maduma Mahlalela
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Adrienne L. Edkins
- Department of Biochemistry and Microbiology, Biomedical Biotechnology Research Centre (BioBRU), Rhodes University, Makhanda 6139, South Africa;
| | - Aileen Boshoff
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| |
Collapse
|
7
|
Gómez-Chávez F, Murrieta-Coxca JM, Caballero-Ortega H, Morales-Prieto DM, Markert UR. Host-pathogen interactions mediated by extracellular vesicles in Toxoplasma gondii infection during pregnancy. J Reprod Immunol 2023; 158:103957. [PMID: 37253287 DOI: 10.1016/j.jri.2023.103957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/01/2023]
Abstract
Molecular communication between a pathogen and its host is crucial for a successful interplay. Extracellular vesicles (EVs) act as mediators for the delivery of molecular signals among pathogens or between pathogens and the host. Toxoplasma gondii (T. gondii), an intracellular parasite with a worldwide presence, produces EVs itself, or induces the secretion of EVs from infected host cells potentially having capacities to modulate the host immune response. T. gondii infection is particularly important during pregnancy. Depending on the gestational age at the time of infection, the parasite can be transmitted through the placenta to the fetus, causing clinical complications such as jaundice, hepatosplenomegaly, chorioretinitis, cranioencephalic abnormalities, or even death. T. gondii infection is related to a pro-inflammatory immune response in both mother and fetus, which may enhance parasite transmission, but the implication of EV signaling in this process remains unclear. In this review, we summarize the current knowledge on EV release from T. gondii and its human host cells in regard to the immunological consequences and the passage through the placenta.
Collapse
Affiliation(s)
- Fernando Gómez-Chávez
- Sección de Estudios de Posgrado e Investigación, Escuela Nacional de Medicina y Homeopatía-Instituto Politécnico Nacional, Mexico City, Mexico; Programa de Posgrado en Ciencia y Tecnología de Vacunas y Bioterapéuticos, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Heriberto Caballero-Ortega
- Secretaría de Salud, Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, Mexico City, Mexico
| | | | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany.
| |
Collapse
|
8
|
McWilliam KR. Cell-cell communication in African trypanosomes. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001388. [PMID: 37643128 PMCID: PMC10482365 DOI: 10.1099/mic.0.001388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Years of research have shown us that unicellular organisms do not exist entirely in isolation, but rather that they are capable of an altogether far more sociable way of living. Single cells produce, receive and interpret signals, coordinating and changing their behaviour according to the information received. Although this cell-cell communication has long been considered the norm in the bacterial world, an increasing body of knowledge is demonstrating that single-celled eukaryotic parasites also maintain active social lives. This communication can drive parasite development, facilitate the invasion of new niches and, ultimately, influence infection outcome. In this review, I present the evidence for cell-cell communication during the life cycle of the African trypanosomes, from their mammalian hosts to their insect vectors, and reflect on the many remaining unanswered questions in this fascinating field.
Collapse
Affiliation(s)
- K. R. McWilliam
- Institute for Immunology and Infection Research, School of Biological Sciences, King’s Buildings, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| |
Collapse
|
9
|
Moreira RS, Calomeno NA, das Neves GB, do Nascimento LFN, Filho VB, Wagner G, Miletti LC. Trypanosoma evansi secretome carries potential biomarkers for Surra diagnosis. J Proteomics 2023; 272:104789. [PMID: 36464092 DOI: 10.1016/j.jprot.2022.104789] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
Trypanosoma evansi is a parasite that is phylogenetically close to Trypanosoma brucei and is the causative agent of a disease known as surra. Surra is responsible for a high mortality rate in livestock and large economic losses in the Americas, Africa, and Asia. This work aimed to analyze in vitro secreted proteins from T. evansi and identify potential treatment and diagnostic biomarkers for surra diagnosis. Two groups were used. In one group the parasites were purified using a DEAE-Cellulose column and maintained in a secretion medium while in the other group the parasites were not purified. Each group was further divided to be maintained at either 37 °C or 27 °C. We identified 246 proteins through mass spectrometry and found that the temperature appears to modulate protein secretion. We found minimal variations in the protein pools from pure and non-purified sets. We observed an emphasis on proteins associated to vesicles, glycolysis, and cellular homeostasis through the enrichment of GO. Also, we found that most secretome proteins share homologous proteins with T. b. brucei, T. b. gambiense, T. vivax, T. equiperdum, and T. b. rhodesiense secretome but unique T. evansi epitopes with potential biomarkers for surra diagnosis were detected. SIGNIFICANCE: Trypanosoma evansi is a parasite of African origin that is phylogenetically close to Trypanosoma brucei. As with other trypanosomatids and blood parasites, its infection causes non-pathognomonic symptoms, which makes its diagnosis difficult. One great problem is the fact that no diagnostic test differentiates between Trypanosoma equiperdum and T. evansi, which is a problem in South America and Asia, and Africa. Thus, it is urgent to study the biochemistry of the parasite to discover proteins that can be used for differential diagnosis or be possible therapeutic targets. In addition, the study of the secretome can point out proteins that are used by the parasite in its interactions with the host, helping to understand the progression of the disease.
Collapse
Affiliation(s)
- Renato Simões Moreira
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil; Instituto Federal de Santa Catarina (IFSC), Campus Gaspar, R. Adriano Kormann, 510 - Bela Vista, Gaspar, SC 89111-009, Brazil
| | - Nathália Anderson Calomeno
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil
| | - Gabriella Bassi das Neves
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil
| | - Luiz Flávio Nepomuceno do Nascimento
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil
| | - Vilmar Benetti Filho
- Laboratório de Bioinformática, Universidade Federal de Santa Catarina, Campus João David Ferreira Lima, Setor F, Bloco A, Sala 318, Caixa postal 476, Trindade, Florianópolis, SC 88040-970, Brazil
| | - Glauber Wagner
- Laboratório de Bioinformática, Universidade Federal de Santa Catarina, Campus João David Ferreira Lima, Setor F, Bloco A, Sala 318, Caixa postal 476, Trindade, Florianópolis, SC 88040-970, Brazil
| | - Luiz Claudio Miletti
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil.
| |
Collapse
|
10
|
Rangel-Ramírez VV, González-Sánchez HM, Lucio-García C. Exosomes: from biology to immunotherapy in infectious diseases. Infect Dis (Lond) 2023; 55:79-107. [PMID: 36562253 DOI: 10.1080/23744235.2022.2149852] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Exosomes are extracellular vesicles derived from the endosomal compartment, which are released by all kinds of eukaryotic and prokaryotic organisms. These vesicles contain a variety of biomolecules that differ both in quantity and type depending on the origin and cellular state. Exosomes are internalized by recipient cells, delivering their content and thus contributing to cell-cell communication in health and disease. During infections exosomes may exert a dual role, on one hand, they can transmit pathogen-related molecules mediating further infection and damage, and on the other hand, they can protect the host by activating the immune response and reducing pathogen spread. Selective packaging of pathogenic components may mediate these effects. Recently, quantitative analysis of samples by omics technologies has allowed a deep characterization of the proteins, lipids, RNA, and metabolite cargoes of exosomes. Knowledge about the content of these vesicles may facilitate their therapeutic application. Furthermore, as exosomes have been detected in almost all biological fluids, pathogenic or host-derived components can be identified in liquid biopsies, making them suitable for diagnosis and prognosis. This review attempts to organize the recent findings on exosome composition and function during viral, bacterial, fungal, and protozoan infections, and their contribution to host defense or to pathogen spread. Moreover, we summarize the current perspectives and future directions regarding the potential application of exosomes for prophylactic and therapeutic purposes.
Collapse
Affiliation(s)
| | | | - César Lucio-García
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, México
| |
Collapse
|
11
|
Cruz Camacho A, Alfandari D, Kozela E, Regev-Rudzki N. Biogenesis of extracellular vesicles in protozoan parasites: The ESCRT complex in the trafficking fast lane? PLoS Pathog 2023; 19:e1011140. [PMID: 36821560 PMCID: PMC9949670 DOI: 10.1371/journal.ppat.1011140] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Extracellular vesicles (EVs) provide a central mechanism of cell-cell communication. While EVs are found in most organisms, their pathogenesis-promoting roles in parasites are of particular interest given the potential for medical insight and consequential therapeutic intervention. Yet, a key feature of EVs in human parasitic protozoa remains elusive: their mechanisms of biogenesis. Here, we survey the current knowledge on the biogenesis pathways of EVs secreted by the four main clades of human parasitic protozoa: apicomplexans, trypanosomatids, flagellates, and amoebae. In particular, we shine a light on findings pertaining to the Endosomal Sorting Complex Required for Transport (ESCRT) machinery, as in mammals it plays important roles in EV biogenesis. This review highlights the diversity in EV biogenesis in protozoa, as well as the related involvement of the ESCRT system in these unique organisms.
Collapse
Affiliation(s)
- Abel Cruz Camacho
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel Alfandari
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Ewa Kozela
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
12
|
Mabille D, Dirkx L, Thys S, Vermeersch M, Montenye D, Govaerts M, Hendrickx S, Takac P, Van Weyenbergh J, Pintelon I, Delputte P, Maes L, Pérez-Morga D, Timmermans JP, Caljon G. Impact of pulmonary African trypanosomes on the immunology and function of the lung. Nat Commun 2022; 13:7083. [PMID: 36400767 PMCID: PMC9674601 DOI: 10.1038/s41467-022-34757-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022] Open
Abstract
Approximately 20% of sleeping sickness patients exhibit respiratory complications, however, with a largely unknown role of the parasite. Here we show that tsetse fly-transmitted Trypanosoma brucei parasites rapidly and permanently colonize the lungs and occupy the extravascular spaces surrounding the blood vessels of the alveoli and bronchi. They are present as nests of multiplying parasites exhibiting close interactions with collagen and active secretion of extracellular vesicles. The local immune response shows a substantial increase of monocytes, macrophages, dendritic cells and γδ and activated αβ T cells and a later influx of neutrophils. Interestingly, parasite presence results in a significant reduction of B cells, eosinophils and natural killer cells. T. brucei infected mice show no infection-associated pulmonary dysfunction, mirroring the limited pulmonary clinical complications during sleeping sickness. However, the substantial reduction of the various immune cells may render individuals more susceptible to opportunistic infections, as evident by a co-infection experiment with respiratory syncytial virus. Collectively, these observations provide insights into a largely overlooked target organ, and may trigger new diagnostic and supportive therapeutic approaches for sleeping sickness.
Collapse
Affiliation(s)
- Dorien Mabille
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Sofie Thys
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Daniel Montenye
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Matthias Govaerts
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Peter Takac
- Institute of Zoology, Slovak Academy of Sciences, 84506, Bratislava, Slovakia
- Scientica, Ltd., 83106, Bratislava, Slovakia
| | - Johan Van Weyenbergh
- Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute of Medical Research, KU Leuven, Leuven, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - David Pérez-Morga
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
13
|
Shokouhy M, Sarvnaz H, Taslimi Y, Lajevardi MS, Habibzadeh S, Mizbani A, Shekari F, Behbahani M, Torrecilhas AC, Rafati S. Isolation, characterization, and functional study of extracellular vesicles derived from Leishmania tarentolae. Front Cell Infect Microbiol 2022; 12:921410. [PMID: 35992172 PMCID: PMC9381964 DOI: 10.3389/fcimb.2022.921410] [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: 04/15/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Leishmania (L.) species are protozoan parasites with a complex life cycle consisting of a number of developmental forms that alternate between the sand fly vector and their host. The non-pathogenic species L. tarentolae is not able to induce an active infection in a human host. It has been observed that, in pathogenic species, extracellular vesicles (EVs) could exacerbate the infection. However, so far, there is no report on the identification, isolation, and characterization of L. tarentolae EVs. In this study, we have isolated and characterized EVs from L. tarentolaeGFP+ (tEVs) along with L. majorGFP+ as a reference and positive control. The EVs secreted by these two species demonstrated similar particle size distribution (approximately 200 nm) in scanning electron microscopy and nanoparticle tracking analysis. Moreover, the said EVs showed similar protein content, and GFP and GP63 proteins were detected in both using dot blot analysis. Furthermore, we could detect Leishmania-derived GP63 protein in THP-1 cells treated with tEVs. Interestingly, we observed a significant increase in the production of IFN-γ, TNF-α, and IL-1β, while there were no significant differences in IL-6 levels in THP-1 cells treated with tEVs following an infection with L. major compared with another group of macrophages that were treated with L. major EVs prior to the infection. Another exciting observation of this study was a significant decrease in parasite load in tEV-treated Leishmania-infected macrophages. In addition, in comparison with another group of Leishmania-infected macrophages which was not exposed to any EVs, tEV managed to increase IFN-γ and decrease IL-6 and the parasite burden. In conclusion, we report for the first time that L. tarentolae can release EVs and provide evidence that tEVs are able to control the infection in human macrophages, making them a great potential platform for drug delivery, at least for parasitic infections.
Collapse
Affiliation(s)
- Mehrdad Shokouhy
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Hamzeh Sarvnaz
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mahya Sadat Lajevardi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Habibzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Amir Mizbani
- Department of Health Science and Technology, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology Cell Science, Research Center, Royan Institute for Stem Cell Biology and Technology, Academic center tor Education, Culture and Research (ACECR), Tehran, Iran
| | - Mandana Behbahani
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), Diadema, Brazil
- *Correspondence: Ana Claudia Torrecilhas, ; Sima Rafati, ;
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Ana Claudia Torrecilhas, ; Sima Rafati, ;
| |
Collapse
|
14
|
Tettey MD, Rojas F, Matthews KR. Extracellular release of two peptidases dominates generation of the trypanosome quorum-sensing signal. Nat Commun 2022; 13:3322. [PMID: 35680928 PMCID: PMC9184580 DOI: 10.1038/s41467-022-31057-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/31/2022] [Indexed: 01/22/2023] Open
Abstract
Trypanosomes causing African sleeping sickness use quorum-sensing (QS) to generate transmission-competent stumpy forms in mammalian hosts. This density-dependent process is signalled by oligopeptides that stimulate the signal transduction pathway leading to stumpy formation. Here, using mass spectrometry analysis, we identify peptidases released by trypanosomes and, for 12 peptidases, confirm their extracellular delivery. Thereafter, we determine the contribution of each peptidase to QS signal production using systematic inducible overexpression in vivo, and confirm this activity operates through the physiological QS signalling pathway. Gene knockout of the QS-active peptidases identifies two enzymes, oligopeptidase B and metallocarboxypeptidase 1, that significantly reduce QS when ablated individually. Further, combinatorial gene knockout of both peptidases confirms their dominance in the generation of the QS signal, with peptidase release of oligopeptidase B mediated via an unconventional protein secretion pathway. This work identifies how the QS signal driving trypanosome virulence and transmission is generated in mammalian hosts.
Collapse
Affiliation(s)
- Mabel Deladem Tettey
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH93FL, UK
| | - Federico Rojas
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH93FL, UK.
- Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4AT, UK.
| | - Keith R Matthews
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH93FL, UK.
| |
Collapse
|
15
|
Monic S, Lamy A, Thonnus M, Bizarra-Rebelo T, Bringaud F, Smith TK, Figueiredo LM, Rivière L. A novel lipase with dual localisation in Trypanosoma brucei. Sci Rep 2022; 12:4766. [PMID: 35306507 PMCID: PMC8934347 DOI: 10.1038/s41598-022-08546-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 03/03/2022] [Indexed: 12/05/2022] Open
Abstract
Phospholipases are esterases involved in lipid catabolism. In pathogenic micro-organisms (bacteria, fungi, parasites) they often play a critical role in virulence and pathogenicity. A few phospholipases (PL) have been characterised so far at the gene and protein level in unicellular parasites including African trypanosomes (AT). They could play a role in different processes such as host–pathogen interaction, antigenic variation, intermediary metabolism. By mining the genome database of AT we found putative new phospholipase candidate genes and here we provided biochemical evidence that one of these has lipolytic activity. This protein has a unique non-canonical glycosome targeting signal responsible for its dual localisation in the cytosol and the peroxisomes-related organelles named glycosomes. We also show that this new phospholipase is excreted by these pathogens and that antibodies directed against this protein are generated during an experimental infection with T. brucei gambiense, a subspecies responsible for infection in humans. This feature makes this protein a possible tool for diagnosis.
Collapse
|
16
|
Miranda MR, Sayé M, Reigada C, Galceran F, Rengifo M, Maciel BJ, Digirolamo FA, Pereira CA. Revisiting trypanosomatid nucleoside diphosphate kinases. Mem Inst Oswaldo Cruz 2022; 116:e210339. [PMID: 35170678 PMCID: PMC8833001 DOI: 10.1590/0074-02760210339] [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: 10/18/2021] [Accepted: 11/26/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND An increasing amount of research has led to the positioning of nucleoside diphosphate kinases (NDPK/NDK) as key metabolic enzymes among all organisms. They contribute to the maintenance the intracellular di- and tri- phosphate nucleoside homeostasis, but they also are involved in widely diverse processes such as gene regulation, apoptosis, signal transduction and many other regulatory roles. OBJETIVE Examine in depth the NDPKs of trypanosomatid parasites responsible for devastating human diseases (e.g., Trypanosoma cruzi, Trypanosoma brucei and Leishmania spp.) which deserve special attention. METHODS The earliest and latest advances in the topic were explored, focusing on trypanosomatid NDPK features, multifunctionality and suitability as molecular drug targets. FINDINGS Trypanosomatid NDPKs appear to play functions different from their host counterparts. Evidences indicate that they would perform key roles in the parasite metabolism such as nucleotide homeostasis, drug resistance, DNA damage responses and gene regulation, as well as host-parasite interactions, infection, virulence and immune evasion, placing them as attractive pharmacological targets. MAIN CONCLUSIONS NDPKs are very interesting multifunctional enzymes. In the present review, the potential of trypanosomatid NDPKs was highlighted, raising awareness of their value not only with respect to parasite biology but also as molecular targets.
Collapse
Affiliation(s)
- Mariana R Miranda
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A Lanari, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina,+ Corresponding author: /
| | - Melisa Sayé
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A Lanari, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Chantal Reigada
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A Lanari, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Facundo Galceran
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A Lanari, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Marcos Rengifo
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A Lanari, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Belen J Maciel
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A Lanari, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Fabio A Digirolamo
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A Lanari, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Claudio A Pereira
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A Lanari, Buenos Aires, Argentina,Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| |
Collapse
|
17
|
Temporão A, Sanches-Vaz M, Luís R, Nunes-Cabaço H, Smith TK, Prudêncio M, Figueiredo LM. Excreted Trypanosoma brucei proteins inhibit Plasmodium hepatic infection. PLoS Negl Trop Dis 2021; 15:e0009912. [PMID: 34714824 PMCID: PMC8580256 DOI: 10.1371/journal.pntd.0009912] [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: 05/31/2021] [Revised: 11/10/2021] [Accepted: 10/15/2021] [Indexed: 11/25/2022] Open
Abstract
Malaria, a disease caused by Plasmodium parasites, remains a major threat to public health globally. It is the most common disease in patients with sleeping sickness, another parasitic illness, caused by Trypanosoma brucei. We have previously shown that a T. brucei infection impairs a secondary P. berghei liver infection and decreases malaria severity in mice. However, whether this effect requires an active trypanosome infection remained unknown. Here, we show that Plasmodium liver infection can also be inhibited by the serum of a mouse previously infected by T. brucei and by total protein lysates of this kinetoplastid. Biochemical characterisation showed that the anti-Plasmodium activity of the total T. brucei lysates depends on its protein fraction, but is independent of the abundant variant surface glycoprotein. Finally, we found that the protein(s) responsible for the inhibition of Plasmodium infection is/are present within a fraction of ~350 proteins that are excreted to the bloodstream of the host. We conclude that the defence mechanism developed by trypanosomes against Plasmodium relies on protein excretion. This study opens the door to the identification of novel antiplasmodial intervention strategies.
Collapse
Affiliation(s)
- Adriana Temporão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Margarida Sanches-Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rafael Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Helena Nunes-Cabaço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Terry K. Smith
- Schools of Biology and Chemistry Biomedical Sciences Research Complex, The North Haugh, The University, St. Andrews, Scotland, United Kingdom
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Luisa M. Figueiredo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
18
|
Bardet-Biedl syndrome proteins modulate the release of bioactive extracellular vesicles. Nat Commun 2021; 12:5671. [PMID: 34580290 PMCID: PMC8476602 DOI: 10.1038/s41467-021-25929-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Primary cilia are microtubule based sensory organelles important for receiving and processing cellular signals. Recent studies have shown that cilia also release extracellular vesicles (EVs). Because EVs have been shown to exert various physiological functions, these findings have the potential to alter our understanding of how primary cilia regulate specific signalling pathways. So far the focus has been on lgEVs budding directly from the ciliary membrane. An association between cilia and MVB-derived smEVs has not yet been described. We show that ciliary mutant mammalian cells demonstrate increased secretion of small EVs (smEVs) and a change in EV composition. Characterisation of smEV cargo identified signalling molecules that are differentially loaded upon ciliary dysfunction. Furthermore, we show that these smEVs are biologically active and modulate the WNT response in recipient cells. These results provide us with insights into smEV-dependent ciliary signalling mechanisms which might underly ciliopathy disease pathogenesis. Extracellular vesicles (EV) are known to be released from the primary cilium, but the role ciliary proteins play in EV biogenesis remains unexplored. Here, the authors demonstrate increased secretion of small EVs with altered cargo composition from cells with known ciliarelated mutations. Wnt related molecules made up a majority of altered cargo
Collapse
|
19
|
Magez S, Li Z, Nguyen HTT, Pinto Torres JE, Van Wielendaele P, Radwanska M, Began J, Zoll S, Sterckx YGJ. The History of Anti-Trypanosome Vaccine Development Shows That Highly Immunogenic and Exposed Pathogen-Derived Antigens Are Not Necessarily Good Target Candidates: Enolase and ISG75 as Examples. Pathogens 2021; 10:pathogens10081050. [PMID: 34451514 PMCID: PMC8400590 DOI: 10.3390/pathogens10081050] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 12/02/2022] Open
Abstract
Salivarian trypanosomes comprise a group of extracellular anthroponotic and zoonotic parasites. The only sustainable method for global control of these infection is through vaccination of livestock animals. Despite multiple reports describing promising laboratory results, no single field-applicable solution has been successful so far. Conventionally, vaccine research focusses mostly on exposed immunogenic antigens, or the structural molecular knowledge of surface exposed invariant immunogens. Unfortunately, extracellular parasites (or parasites with extracellular life stages) have devised efficient defense systems against host antibody attacks, so they can deal with the mammalian humoral immune response. In the case of trypanosomes, it appears that these mechanisms have been perfected, leading to vaccine failure in natural hosts. Here, we provide two examples of potential vaccine candidates that, despite being immunogenic and accessible to the immune system, failed to induce a functionally protective memory response. First, trypanosomal enolase was tested as a vaccine candidate, as it was recently characterized as a highly conserved enzyme that is readily recognized during infection by the host antibody response. Secondly, we re-addressed a vaccine approach towards the Invariant Surface Glycoprotein ISG75, and showed that despite being highly immunogenic, trypanosomes can avoid anti-ISG75 mediated parasitemia control.
Collapse
Affiliation(s)
- Stefan Magez
- Laboratory of Cellular and Molecular Immunology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (Z.L.); (H.T.T.N.); (J.E.P.T.)
- Department of Biochemistry and Microbiology, Ghent University, Ledeganckstraat 35, 9000 Ghent, Belgium
- Laboratory for Biomedical Research, Department of Molecular Biotechnology, Environment Technology and Food Technology, Ghent University Global Campus, Songdomunhwa-Ro 119-5, Yeonsu-Gu, Incheon 406-840, Korea;
- Correspondence:
| | - Zeng Li
- Laboratory of Cellular and Molecular Immunology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (Z.L.); (H.T.T.N.); (J.E.P.T.)
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium; (P.V.W.); (Y.G.-J.S.)
| | - Hang Thi Thu Nguyen
- Laboratory of Cellular and Molecular Immunology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (Z.L.); (H.T.T.N.); (J.E.P.T.)
- Department of Biochemistry and Microbiology, Ghent University, Ledeganckstraat 35, 9000 Ghent, Belgium
- Laboratory for Biomedical Research, Department of Molecular Biotechnology, Environment Technology and Food Technology, Ghent University Global Campus, Songdomunhwa-Ro 119-5, Yeonsu-Gu, Incheon 406-840, Korea;
| | - Joar Esteban Pinto Torres
- Laboratory of Cellular and Molecular Immunology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; (Z.L.); (H.T.T.N.); (J.E.P.T.)
| | - Pieter Van Wielendaele
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium; (P.V.W.); (Y.G.-J.S.)
| | - Magdalena Radwanska
- Laboratory for Biomedical Research, Department of Molecular Biotechnology, Environment Technology and Food Technology, Ghent University Global Campus, Songdomunhwa-Ro 119-5, Yeonsu-Gu, Incheon 406-840, Korea;
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark Zwijnaarde 71, 9000 Ghent, Belgium
| | - Jakub Began
- Laboratory of Structural Parasitology, Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo Namesti 2, 16610 Prague 6, Czech Republic; (J.B.); (S.Z.)
| | - Sebastian Zoll
- Laboratory of Structural Parasitology, Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo Namesti 2, 16610 Prague 6, Czech Republic; (J.B.); (S.Z.)
| | - Yann G.-J. Sterckx
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Wilrijk, Belgium; (P.V.W.); (Y.G.-J.S.)
| |
Collapse
|
20
|
De Sousa KP, Potriquet J, Mulvenna J, Sotillo J, Groves PL, Loukas A, Apte SH, Doolan DL. Proteomic identification of the contents of small extracellular vesicles from in vivo Plasmodium yoelii infection. Int J Parasitol 2021; 52:35-45. [PMID: 34339723 DOI: 10.1016/j.ijpara.2021.06.001] [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] [Received: 03/15/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022]
Abstract
Small extracellular vesicles, including exosomes, are formed by the endocytic pathway and contain genetic and protein material which reflect the contents of their cells of origin. These contents have a role in vesicle-mediated information transfer, as well as physiological and pathological functions. Thus, these vesicles are of great interest as therapeutic targets, or as vehicles for immunomodulatory control. In Plasmodium spp. infections, vesicles derived from the parasite or parasite-infected cells have been shown to induce the expression of pro-inflammatory elements, which have been correlated with manifestations of clinical disease. Herein, we characterised the protein cargo of naturally occurring sEVs in the plasma of P. yoelii-infected mice. After in vivo infections, extracellular vesicles in the size range of exosomes were collected by sequential centrifugation/ultracentrifugation followed by isopycnic gradient separation. Analysis of the vesicles was performed by transmission electron microscopy, dynamic light scattering, SDS-PAGE and flow cytometry. LC-MS analysis followed by bioinformatics analysis predicted parasite protein cargo associated with exosomes. Within these small extracellular vesicles, we identified proteins of interest as vaccine candidates, uncharacterized proteins which may be targets of T cell immunoreactivity, and proteins involved in metabolic processes, regulation, homeostasis and immunity. Importantly, the small extracellular vesicles studied in our work were obtained from in vivo infection rather than from the supernatant of in vitro cultures. These findings add to the growing interest in parasite small extracellular vesicles, further our understanding of the interactions between host and parasite, and identify novel proteins which may represent potential targets for vaccination against malaria.
Collapse
Affiliation(s)
- Karina P De Sousa
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Jeremy Potriquet
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns QLD 4878 Australia
| | - Jason Mulvenna
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Javier Sotillo
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns QLD 4878 Australia; Parasitology Reference and Research Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Penny L Groves
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Alex Loukas
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Simon H Apte
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns QLD 4878 Australia
| | - Denise L Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns QLD 4878 Australia.
| |
Collapse
|
21
|
Steketee PC, Dickie EA, Iremonger J, Crouch K, Paxton E, Jayaraman S, Alfituri OA, Awuah-Mensah G, Ritchie R, Schnaufer A, Rowan T, de Koning HP, Gadelha C, Wickstead B, Barrett MP, Morrison LJ. Divergent metabolism between Trypanosoma congolense and Trypanosoma brucei results in differential sensitivity to metabolic inhibition. PLoS Pathog 2021; 17:e1009734. [PMID: 34310651 PMCID: PMC8384185 DOI: 10.1371/journal.ppat.1009734] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/24/2021] [Accepted: 06/21/2021] [Indexed: 11/18/2022] Open
Abstract
Animal African Trypanosomiasis (AAT) is a debilitating livestock disease prevalent across sub-Saharan Africa, a main cause of which is the protozoan parasite Trypanosoma congolense. In comparison to the well-studied T. brucei, there is a major paucity of knowledge regarding the biology of T. congolense. Here, we use a combination of omics technologies and novel genetic tools to characterise core metabolism in T. congolense mammalian-infective bloodstream-form parasites, and test whether metabolic differences compared to T. brucei impact upon sensitivity to metabolic inhibition. Like the bloodstream stage of T. brucei, glycolysis plays a major part in T. congolense energy metabolism. However, the rate of glucose uptake is significantly lower in bloodstream stage T. congolense, with cells remaining viable when cultured in concentrations as low as 2 mM. Instead of pyruvate, the primary glycolytic endpoints are succinate, malate and acetate. Transcriptomics analysis showed higher levels of transcripts associated with the mitochondrial pyruvate dehydrogenase complex, acetate generation, and the glycosomal succinate shunt in T. congolense, compared to T. brucei. Stable-isotope labelling of glucose enabled the comparison of carbon usage between T. brucei and T. congolense, highlighting differences in nucleotide and saturated fatty acid metabolism. To validate the metabolic similarities and differences, both species were treated with metabolic inhibitors, confirming that electron transport chain activity is not essential in T. congolense. However, the parasite exhibits increased sensitivity to inhibition of mitochondrial pyruvate import, compared to T. brucei. Strikingly, T. congolense exhibited significant resistance to inhibitors of fatty acid synthesis, including a 780-fold higher EC50 for the lipase and fatty acid synthase inhibitor Orlistat, compared to T. brucei. These data highlight that bloodstream form T. congolense diverges from T. brucei in key areas of metabolism, with several features that are intermediate between bloodstream- and insect-stage T. brucei. These results have implications for drug development, mechanisms of drug resistance and host-pathogen interactions.
Collapse
Affiliation(s)
- Pieter C Steketee
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Emily A Dickie
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - James Iremonger
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Kathryn Crouch
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Edith Paxton
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Siddharth Jayaraman
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Omar A Alfituri
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Ryan Ritchie
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Achim Schnaufer
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Tim Rowan
- Global Alliance for Livestock Veterinary Medicines, Edinburgh, United Kingdom
| | - Harry P de Koning
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Catarina Gadelha
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Bill Wickstead
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Michael P Barrett
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.,Glasgow Polyomics, University of Glasgow, Glasgow, United Kingdom
| | - Liam J Morrison
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
22
|
Zuma AA, Dos Santos Barrias E, de Souza W. Basic Biology of Trypanosoma cruzi. Curr Pharm Des 2021; 27:1671-1732. [PMID: 33272165 DOI: 10.2174/1381612826999201203213527] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/01/2020] [Accepted: 10/08/2020] [Indexed: 11/22/2022]
Abstract
The present review addresses basic aspects of the biology of the pathogenic protozoa Trypanosoma cruzi and some comparative information of Trypanosoma brucei. Like eukaryotic cells, their cellular organization is similar to that of mammalian hosts. However, these parasites present structural particularities. That is why the following topics are emphasized in this paper: developmental stages of the life cycle in the vertebrate and invertebrate hosts; the cytoskeleton of the protozoa, especially the sub-pellicular microtubules; the flagellum and its attachment to the protozoan body through specialized junctions; the kinetoplast-mitochondrion complex, including its structural organization and DNA replication; glycosome and its role in the metabolism of the cell; acidocalcisome, describing its morphology, biochemistry, and functional role; cytostome and the endocytic pathway; the organization of the endoplasmic reticulum and Golgi complex; the nucleus, describing its structural organization during interphase and division; and the process of interaction of the parasite with host cells. The unique characteristics of these structures also make them interesting chemotherapeutic targets. Therefore, further understanding of cell biology aspects contributes to the development of drugs for chemotherapy.
Collapse
Affiliation(s)
- Aline A Zuma
- Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emile Dos Santos Barrias
- Laboratorio de Metrologia Aplicada a Ciencias da Vida, Diretoria de Metrologia Aplicada a Ciencias da Vida - Instituto Nacional de Metrologia, Qualidade e Tecnologia (Inmetro), Rio de Janeiro, Brazil
| | - Wanderley de Souza
- Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Luo M, Xu L, Qian Z, Sun X. Infection-Associated Thymic Atrophy. Front Immunol 2021; 12:652538. [PMID: 34113341 PMCID: PMC8186317 DOI: 10.3389/fimmu.2021.652538] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
The thymus is a vital organ of the immune system that plays an essential role in thymocyte development and maturation. Thymic atrophy occurs with age (physiological thymic atrophy) or as a result of viral, bacterial, parasitic or fungal infection (pathological thymic atrophy). Thymic atrophy directly results in loss of thymocytes and/or destruction of the thymic architecture, and indirectly leads to a decrease in naïve T cells and limited T cell receptor diversity. Thus, it is important to recognize the causes and mechanisms that induce thymic atrophy. In this review, we highlight current progress in infection-associated pathogenic thymic atrophy and discuss its possible mechanisms. In addition, we discuss whether extracellular vesicles/exosomes could be potential carriers of pathogenic substances to the thymus, and potential drugs for the treatment of thymic atrophy. Having acknowledged that most current research is limited to serological aspects, we look forward to the possibility of extending future work regarding the impact of neural modulation on thymic atrophy.
Collapse
Affiliation(s)
- Mingli Luo
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Lingxin Xu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Zhengyu Qian
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| |
Collapse
|
24
|
Balmer EA, Faso C. The Road Less Traveled? Unconventional Protein Secretion at Parasite-Host Interfaces. Front Cell Dev Biol 2021; 9:662711. [PMID: 34109175 PMCID: PMC8182054 DOI: 10.3389/fcell.2021.662711] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/07/2021] [Indexed: 01/01/2023] Open
Abstract
Protein secretion in eukaryotic cells is a well-studied process, which has been known for decades and is dealt with by any standard cell biology textbook. However, over the past 20 years, several studies led to the realization that protein secretion as a process might not be as uniform among different cargos as once thought. While in classic canonical secretion proteins carry a signal sequence, the secretory or surface proteome of several organisms demonstrated a lack of such signals in several secreted proteins. Other proteins were found to indeed carry a leader sequence, but simply circumvent the Golgi apparatus, which in canonical secretion is generally responsible for the modification and sorting of secretory proteins after their passage through the endoplasmic reticulum (ER). These alternative mechanisms of protein translocation to, or across, the plasma membrane were collectively termed “unconventional protein secretion” (UPS). To date, many research groups have studied UPS in their respective model organism of choice, with surprising reports on the proportion of unconventionally secreted proteins and their crucial roles for the cell and survival of the organism. Involved in processes such as immune responses and cell proliferation, and including far more different cargo proteins in different organisms than anyone had expected, unconventional secretion does not seem so unconventional after all. Alongside mammalian cells, much work on this topic has been done on protist parasites, including genera Leishmania, Trypanosoma, Plasmodium, Trichomonas, Giardia, and Entamoeba. Studies on protein secretion have mainly focused on parasite-derived virulence factors as a main source of pathogenicity for hosts. Given their need to secrete a variety of substrates, which may not be compatible with canonical secretion pathways, the study of mechanisms for alternative secretion pathways is particularly interesting in protist parasites. In this review, we provide an overview on the current status of knowledge on UPS in parasitic protists preceded by a brief overview of UPS in the mammalian cell model with a focus on IL-1β and FGF-2 as paradigmatic UPS substrates.
Collapse
Affiliation(s)
- Erina A Balmer
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Carmen Faso
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
López L, Chiribao ML, Girard MC, Gómez KA, Carasi P, Fernandez M, Hernandez Y, Robello C, Freire T, Piñeyro MD. The cytosolic tryparedoxin peroxidase from Trypanosoma cruzi induces a pro-inflammatory Th1 immune response in a peroxidatic cysteine-dependent manner. Immunology 2021; 163:46-59. [PMID: 33410127 PMCID: PMC8044337 DOI: 10.1111/imm.13302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/25/2020] [Accepted: 12/19/2020] [Indexed: 12/25/2022] Open
Abstract
Trypanosoma cruzi cytosolic tryparedoxin peroxidase (c-TXNPx) is a 2-Cys peroxiredoxin (Prx) with an important role in detoxifying host cell oxidative molecules during parasite infection. c-TXNPx is a virulence factor, as its overexpression enhances parasite infectivity and resistance to exogenous oxidation. As Prxs from other organisms possess immunomodulatory properties, we studied the effects of c-TXNPx in the immune response and analysed whether the presence of the peroxidatic cysteine is necessary to mediate these properties. To this end, we used a recombinant c-TXNPx and a mutant version (c-TXNPxC52S) lacking the peroxidatic cysteine. We first analysed the oligomerization profile, oxidation state and peroxidase activity of both proteins by gel filtration, Western blot and enzymatic assay, respectively. To investigate their immunological properties, we analysed the phenotype and functional activity of macrophage and dendritic cells and the T-cell response by flow cytometry after injection into mice. Our results show that c-TXNPx, but not c-TXNPxC52S, induces the recruitment of IL-12/23p40-producing innate antigen-presenting cells and promotes a strong specific Th1 immune response. Finally, we studied the cellular and humoral immune response developed in the context of parasite natural infection and found that only wild-type c-TXNPx induces proliferation and high levels of IFN-γ secretion in PBMC from chronic patients without demonstrable cardiac manifestations. In conclusion, we demonstrate that c-TXNPx possesses pro-inflammatory properties that depend on the presence of peroxidatic cysteine that is essential for peroxidase activity and quaternary structure of the protein and could contribute to rational design of immune-based strategies against Chagas disease.
Collapse
Affiliation(s)
- Lucía López
- Laboratorio de Inmunomodulación y Desarrollo de VacunasDepartamento de InmunobiologíaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
- Unidad de Biología MolecularInstitut Pasteur MontevideoMontevideoUruguay
| | - María Laura Chiribao
- Unidad de Biología MolecularInstitut Pasteur MontevideoMontevideoUruguay
- Departamento de BioquímicaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| | - Magalí C. Girard
- Laboratorio de Inmunología de las Infecciones por TripanosomátidosInstituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI‐CONICET)Buenos AiresArgentina
| | - Karina A. Gómez
- Laboratorio de Inmunología de las Infecciones por TripanosomátidosInstituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI‐CONICET)Buenos AiresArgentina
| | - Paula Carasi
- Laboratorio de Inmunomodulación y Desarrollo de VacunasDepartamento de InmunobiologíaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| | - Marisa Fernandez
- Instituto Nacional de Parasitología ‘Doctor Mario Fatala Chabén’Buenos AiresArgentina
| | - Yolanda Hernandez
- Instituto Nacional de Parasitología ‘Doctor Mario Fatala Chabén’Buenos AiresArgentina
| | - Carlos Robello
- Unidad de Biología MolecularInstitut Pasteur MontevideoMontevideoUruguay
- Departamento de BioquímicaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| | - Teresa Freire
- Laboratorio de Inmunomodulación y Desarrollo de VacunasDepartamento de InmunobiologíaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| | - María Dolores Piñeyro
- Unidad de Biología MolecularInstitut Pasteur MontevideoMontevideoUruguay
- Departamento de BioquímicaFacultad de MedicinaUniversidad de La RepúblicaMontevideoUruguay
| |
Collapse
|
26
|
Bamra T, Shafi T, Das S, Kumar M, Dikhit MR, Kumar A, Kumar A, Abhishek K, Pandey K, Sen A, Das P. Leishmania donovani Secretory Mevalonate Kinase Regulates Host Immune Response and Facilitates Phagocytosis. Front Cell Infect Microbiol 2021; 11:641985. [PMID: 33981628 PMCID: PMC8110032 DOI: 10.3389/fcimb.2021.641985] [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: 12/15/2020] [Accepted: 03/31/2021] [Indexed: 11/13/2022] Open
Abstract
Leishmania secretes over 151 proteins during in vitro cultivation. Cellular functions of one such novel protein: mevalonate kinase is discussed here; signifying its importance in Leishmania infection. Visceral Leishmaniasis is a persistent infection, caused by Leishmania donovani in Indian subcontinent. This persistence is partly due to phagocytosis and evasion of host immune response. The underlying mechanism involves secretory proteins of Leishmania parasite; however, related studies are meagre. We have identified a novel secretory Leishmania donovani glycoprotein, Mevalonate kinase (MVK), and shown its importance in parasite internalization and immuno-modulation. In our studies, MVK was found to be secreted maximum after 1 h temperature stress at 37°C. Its secretion was increased by 6.5-fold in phagolysosome-like condition (pH ~5.5, 37°C) than at pH ~7.4 and 25°C. Treatment with MVK modulated host immune system by inducing interleukin-10 and interleukin-4 secretion, suppressing host’s ability to kill the parasite. Peripheral blood mononuclear cell (PBMC)-derived macrophages infected with mevalonate kinase-overexpressing parasites showed an increase in intracellular parasite burden in comparison to infection with vector control parasites. Mechanism behind the increase in phagocytosis and immunosuppression was found to be phosphorylation of mitogen-activated protein (MAP) kinase pathway protein, Extracellular signal-regulated kinases-1/2, and actin scaffold protein, cortactin. Thus, we conclude that Leishmania donovani Mevalonate kinase aids in parasite engulfment and subvert the immune system by interfering with signal transduction pathways in host cells, which causes suppression of the protective response and facilitates their persistence in the host. Our work elucidates the involvement of Leishmania in the process of phagocytosis which is thought to be dependent largely on macrophages and contributes towards better understanding of host pathogen interactions.
Collapse
Affiliation(s)
- Tanvir Bamra
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Taj Shafi
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | | | - Manjay Kumar
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Manas Ranjan Dikhit
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Ajay Kumar
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Ashish Kumar
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Kumar Abhishek
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Krishna Pandey
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Abhik Sen
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Pradeep Das
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| |
Collapse
|
27
|
Dong G, Wagner V, Minguez-Menendez A, Fernandez-Prada C, Olivier M. Extracellular vesicles and leishmaniasis: Current knowledge and promising avenues for future development. Mol Immunol 2021; 135:73-83. [PMID: 33873096 DOI: 10.1016/j.molimm.2021.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 02/09/2023]
Abstract
Extracellular vesicles (EVs) are small, membrane-bound "delivery trucks" that are present in the extracellular environment, including biological fluids. EVs are capable of inducing changes in the physiological status of neighboring cells through the transfer of key macromolecules, and are thought to play a role in a number of pathological processes. Leishmaniasis, caused by the protozoan parasite Leishmania, is an important example. The biology of Leishmania EVs has been studied in detail, and findings point to their role in exacerbation of disease and potential involvement in the perpetuation of drug resistance. Furthermore, the use of EVs for development of vaccines has been explored, as well as their potential use in a number of fields as biomarkers of disease and drug resistance. Here we discuss the latest findings on EVs, with a particular focus on Leishmania, as well as potential avenues for their future development and clinical applications.
Collapse
Affiliation(s)
- George Dong
- Infectious Diseases and Immunology in Global Health Program (IDIGH), The Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Victoria Wagner
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, QC, Canada; The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, Université de Montréal, QC, Canada
| | | | - Christopher Fernandez-Prada
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, QC, Canada; The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, Université de Montréal, QC, Canada.
| | - Martin Olivier
- Infectious Diseases and Immunology in Global Health Program (IDIGH), The Research Institute of the McGill University Health Centre, Montréal, QC, Canada; Departments of Medicine, Microbiology and Immunology, Faculty of Medicine, McGill University, Montréal, QC, Canada.
| |
Collapse
|
28
|
Halliday C, de Castro-Neto A, Alcantara CL, Cunha-E-Silva NL, Vaughan S, Sunter JD. Trypanosomatid Flagellar Pocket from Structure to Function. Trends Parasitol 2021; 37:317-329. [PMID: 33308952 DOI: 10.1016/j.pt.2020.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
The trypanosomatids Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp. are flagellate eukaryotic parasites that cause serious diseases in humans and animals. These parasites have cell shapes defined by a subpellicular microtubule array and all share a number of important cellular features. One of these is the flagellar pocket, an invagination of the cell membrane around the proximal end of the flagellum, which is an important organelle for endo/exocytosis. The flagellar pocket plays a crucial role in parasite pathogenicity and persistence in the host and has a great influence on cell morphogenesis and cell division. Here, we compare the morphology and function of the flagellar pockets between different trypanosomatids, with their life cycles and ecological niches likely influencing these differences.
Collapse
Affiliation(s)
- Clare Halliday
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | - Artur de Castro-Neto
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | - Carolina L Alcantara
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Narcisa L Cunha-E-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Sue Vaughan
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | - Jack D Sunter
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK.
| |
Collapse
|
29
|
Ngambia Freitas FS, Njiokou F, Tsagmo Ngoune JM, Sempere G, Berthier D, Geiger A. Modulation of trypanosome establishment in Glossina palpalis palpalis by its microbiome in the Campo sleeping sickness focus, Cameroon. INFECTION GENETICS AND EVOLUTION 2021; 90:104763. [PMID: 33571685 DOI: 10.1016/j.meegid.2021.104763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 11/30/2022]
Abstract
The purpose of this study was to investigate factors involved in vector competence by analyzing whether the diversity and relative abundance of the different bacterial genera inhabiting the fly's gut could be associated with its trypanosome infection status. This was investigated on 160 randomly selected G. p. palpalis flies - 80 trypanosome-infected, 80 uninfected - collected in 5 villages of the Campo trypanosomiasis focus in South Cameroon. Trypanosome species were identified using specific primers, and the V4 region of the 16S rRNA gene of bacteria was targeted for metabarcoding analysis in order to identify the bacteria and determine microbiome composition. A total of 261 bacterial genera were identified of which only 114 crossed two barriers: a threshold of 0.01% relative abundance and the presence at least in 5 flies. The secondary symbiont Sodalis glossinidius was identified in 50% of the flies but it was not considered since its relative abundance was much lower than the 0.01% relative abundance threshold. The primary symbiont Wigglesworthia displayed 87% relative abundance, the remaining 13% were prominently constituted by the genera Spiroplasma, Tediphilus, Acinetobacter and Pseudomonas. Despite a large diversity in bacterial genera and in their abundance observed in micobiome composition, the statistical analyzes of the 160 tsetse flies showed an association with flies' infection status and the sampling sites. Furthermore, tsetse flies harboring Trypanosoma congolense Savanah type displayed a different composition of bacterial flora compared to uninfected flies. In addition, our study revealed that 36 bacterial genera were present only in uninfected flies, which could therefore suggest a possible involvement in flies' refractoriness; with the exception of Cupriavidus, they were however of low relative abundance. Some genera, including Acinetobacter, Cutibacterium, Pseudomonas and Tepidiphilus, although present both in infected and uninfected flies, were found to be associated with uninfected status of tsetse flies. Hence their effective role deserves to be further evaluated in order to determine whether some of them could become targets for tsetse control of fly vector competence and consequently for the control of the disease. Finally, when comparing the bacterial genera identified in tsetse flies collected during 4 epidemiological surveys, 39 genera were found to be common to flies from at least 2 sampling campaigns.
Collapse
Affiliation(s)
- François Sougal Ngambia Freitas
- INTERTRYP, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France; Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Flobert Njiokou
- Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | | | - Guilhem Sempere
- CIRAD, UMR INTERTRYP, F-34398 Montpellier, France; South Green Bioinformatics Platform, Biodiversity, CIRAD, INRAE, IRD, Montpellier, France; INTERTRYP, Univ Montpellier, CIRAD, IRD, Montpellier, France
| | - David Berthier
- CIRAD, UMR INTERTRYP, F-34398 Montpellier, France; INTERTRYP, Univ Montpellier, CIRAD, IRD, Montpellier, France
| | - Anne Geiger
- INTERTRYP, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France; Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon; Center for Research on Filariasis and other Tropical Diseases (CRFilMT), P.O. Box 5797, Yaoundé, Cameroon.
| |
Collapse
|
30
|
In Silico Identification of New Targets for Diagnosis, Vaccine, and Drug Candidates against Trypanosoma cruzi. DISEASE MARKERS 2021; 2020:9130719. [PMID: 33488847 PMCID: PMC7787821 DOI: 10.1155/2020/9130719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/23/2020] [Accepted: 11/24/2020] [Indexed: 11/17/2022]
Abstract
Chagas disease is a neglected tropical disease caused by the parasite Trypanosoma cruzi. Despite the efforts and distinct methodologies, the search of antigens for diagnosis, vaccine, and drug targets for the disease is still needed. The present study is aimed at identifying possible antigens that could be used for diagnosis, vaccine, and drugs targets against T. cruzi using reverse vaccinology and molecular docking. The genomes of 28 T. cruzi strains available in GenBank (NCBI) were used to obtain the genomic core. Then, subtractive genomics was carried out to identify nonhomologous genes to the host in the core. A total of 2630 conserved proteins in 28 strains of T. cruzi were predicted using OrthoFinder and Diamond software, in which 515 showed no homology to the human host. These proteins were evaluated for their subcellular localization, from which 214 are cytoplasmic and 117 are secreted or present in the plasma membrane. To identify the antigens for diagnosis and vaccine targets, we used the VaxiJen software, and 14 nonhomologous proteins were selected showing high binding efficiency with MHC I and MHC II with potential for in vitro and in vivo tests. When these 14 nonhomologous molecules were compared against other trypanosomatids, it was found that the retrotransposon hot spot (RHS) protein is specific only for T. cruzi parasite suggesting that it could be used for Chagas diagnosis. Such 14 proteins were analyzed using the IEDB software to predict their epitopes in both B and T lymphocytes. Furthermore, molecular docking analysis was performed using the software MHOLline. As a result, we identified 6 possible T. cruzi drug targets that could interact with 4 compounds already known as antiparasitic activities. These 14 protein targets, along with 6 potential drug candidates, can be further validated in future studies, in vivo, regarding Chagas disease.
Collapse
|
31
|
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: 55] [Impact Index Per Article: 11.0] [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
|
32
|
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
|
33
|
Mice immunization with Trypanosoma brucei gambiense translationally controlled tumor protein modulates immunoglobulin and cytokine production, as well as parasitaemia and mice survival after challenge with the parasite. INFECTION GENETICS AND EVOLUTION 2020; 87:104636. [PMID: 33217546 DOI: 10.1016/j.meegid.2020.104636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/06/2020] [Accepted: 11/15/2020] [Indexed: 01/01/2023]
Abstract
Fighting trypanosomiasis with an anti-trypanosome vaccine is ineffective, the parasite being protected by a Variable Surface Glycoprotein (VSG) whose structure is modified at each peak of parasitaemia, which allows it to escape the host's immune defenses. However, the host immunization against an essential factor for the survival of the parasite or the expression of its pathogenicity could achieve the same objective. Here we present the results of mouse immunization against the Translationally Controlled Tumor Protein (TCTP), a protein present in the Trypanosoma brucei gambiense (Tbg) secretome, the parasite responsible for human trypanosomiasis. Mice immunization was followed by infection with Tbg parasites. The production of IgG, IgG1 and IgG2a begun after the second TCTP injection and was dose-dependant, the maximum level of anti-TCTP antibodies remained stable up to 4 days post-infection and then decreased. Regarding cytokines (IL-2, 4, 6, 10, INFγ, TNFα), the most striking result was their total suppression after immunization with the highest TCTP dose. Compared to the control group, the immunized mice displayed a reduced first peak of parasitaemia, a 100% increase in the time to onset of the second peak, and an increased time of mice survival. The effect of immunization was only transient but demonstrated the likely important role that TCTP plays in host-parasite interactions and that some key parasite proteins could reduce infection impact.
Collapse
|
34
|
Varikuti S, Jha BK, Holcomb EA, McDaniel JC, Karpurapu M, Srivastava N, McGwire BS, Satoskar AR, Parinandi NL. The role of vascular endothelium and exosomes in human protozoan parasitic diseases. ACTA ACUST UNITED AC 2020; 4. [PMID: 33089078 PMCID: PMC7575144 DOI: 10.20517/2574-1209.2020.27] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The vascular endothelium is a vital component in maintaining the structure and function of blood vessels. The endothelial cells (ECs) mediate vital regulatory functions such as the proliferation of cells, permeability of various tissue membranes, and exchange of gases, thrombolysis, blood flow, and homeostasis. The vascular endothelium also regulates inflammation and immune cell trafficking, and ECs serve as a replicative niche for many bacterial, viral, and protozoan infectious diseases. Endothelial dysfunction can lead to vasodilation and pro-inflammation, which are the hallmarks of many severe diseases. Exosomes are nanoscale membrane-bound vesicles that emerge from cells and serve as important extracellular components, which facilitate communication between cells and maintain homeostasis during normal and pathophysiological states. Exosomes are also involved in gene transfer, inflammation and antigen presentation, and mediation of the immune response during pathogenic states. Protozoa are a diverse group of unicellular organisms that cause many infectious diseases in humans. In this regard, it is becoming increasingly evident that many protozoan parasites (such as Plasmodium, Trypanosoma, Leishmania, and Toxoplasma) utilize exosomes for the transfer of their virulence factors and effector molecules into the host cells, which manipulate the host gene expression, immune responses, and other biological activities to establish and modulate infection. In this review, we discuss the role of the vascular endothelium and exosomes in and their contribution to pathogenesis in malaria, African sleeping sickness, Chagas disease, and leishmaniasis and toxoplasmosis with an emphasis on their actions on the innate and adaptive immune mechanisms of resistance.
Collapse
Affiliation(s)
- Sanjay Varikuti
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA.,Department of Bioscience & Biotechnology, Banasthali University, Banasthali 304022, India
| | - Bijay Kumar Jha
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Erin A Holcomb
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Jodi C McDaniel
- College of Nursing, The Ohio State University, Columbus, OH 43201, USA
| | - Manjula Karpurapu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Nidhi Srivastava
- Department of Bioscience & Biotechnology, Banasthali University, Banasthali 304022, India
| | - Bradford S McGwire
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Narasimham L Parinandi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| |
Collapse
|
35
|
Olajide JS, Cai J. Perils and Promises of Pathogenic Protozoan Extracellular Vesicles. Front Cell Infect Microbiol 2020; 10:371. [PMID: 32923407 PMCID: PMC7456935 DOI: 10.3389/fcimb.2020.00371] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are membranous structures formed during biological processes in living organisms. For protozoan parasites, secretion of EVs can occur directly from the parasite organellar compartments and through parasite-infected or antigen-stimulated host cells in response to in vitro and in vivo physiological stressors. These secreted EVs characteristically reflect the biochemical features of their parasitic origin and activating stimuli. Here, we review the species-specific morphology and integrity of parasitic protozoan EVs in concurrence with the origin, functions, and internalization process by recipient cells. The activating stimuli for the secretion of EVs in pathogenic protozoa are discoursed alongside their biomolecules and specific immune cell responses to protozoan parasite-derived EVs. We also present some insights on the intricate functions of EVs in the context of protozoan parasitism.
Collapse
Affiliation(s)
- Joshua Seun Olajide
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, China.,Centre for Distance Learning, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Jianping Cai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, China
| |
Collapse
|
36
|
An Unbiased Immunization Strategy Results in the Identification of Enolase as a Potential Marker for Nanobody-Based Detection of Trypanosoma evansi. Vaccines (Basel) 2020; 8:vaccines8030415. [PMID: 32722150 PMCID: PMC7565430 DOI: 10.3390/vaccines8030415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Trypanosoma evansi is a widely spread parasite that causes the debilitating disease “surra” in several types of ungulates. This severely challenges livestock rearing and heavily weighs on the socio-economic development in the affected areas, which include countries on five continents. Active case finding requires a sensitive and specific diagnostic test. In this paper, we describe the application of an unbiased immunization strategy to identify potential biomarkers for Nanobody (Nb)-based detection of T. evansi infections. Alpaca immunization with soluble lysates from different T. evansi strains followed by panning against T. evansi secretome resulted in the selection of a single Nb (Nb11). By combining Nb11-mediated immuno-capturing with mass spectrometry, the T. evansi target antigen was identified as the glycolytic enzyme enolase. Four additional anti-enolase binders were subsequently generated by immunizing another alpaca with the recombinant target enzyme. Together with Nb11, these binders were evaluated for their potential use in a heterologous sandwich detection format. Three Nb pairs were identified as candidates for the further development of an antigen-based assay for Nb-mediated diagnosis of T. evansi infection.
Collapse
|
37
|
de Souza W, Barrias ES. Membrane-bound extracellular vesicles secreted by parasitic protozoa: cellular structures involved in the communication between cells. Parasitol Res 2020; 119:2005-2023. [PMID: 32394001 DOI: 10.1007/s00436-020-06691-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
The focus of this review is a group of structures/organelles collectively known as extracellular vesicles (EVs) that are secreted by most, if not all, cells, varying from mammalian cells to protozoa and even bacteria. They vary in size: some are small (100-200 nm) and others are larger (> 200 nm). In protozoa, however, most of them are small or medium in size (200-400 nm). These include vesicles from different origins. We briefly review the biogenesis of this distinct group that includes (a) exosome, which originates from the multivesicular bodies, an important component of the endocytic pathway; (b) ectosome, formed from a budding process that takes place in the plasma membrane of the cells; (c) vesicles released from the cell surface following a process of patching and capping of ligand/receptor complexes; (d) other processes where tubules secreted by the parasite subsequently originate exosome-like structures. Here, special emphasis is given to EVs secreted by parasitic protozoa such as Leishmania, Trypanosoma, Plasmodium, Toxoplasma, Cryptosporidium, Trichomonas, and Giardia. Most of them have been characterized as exosomes that were isolated using several approaches and characterized by electron microscopy, proteomic analysis, and RNA sequencing. The results obtained show clearly that they present several proteins and different types of RNAs. From the functional point of view, it is now clear that the secreted exosomes can be incorporated by the parasite itself as well as by mammalian cells with which they interact. As a consequence, there is interference both with the parasite (induction of differentiation, changes in infectivity, etc.) and with the host cell. Therefore, the EVs constitute a new system of transference of signals among cells. On the other hand, there are suggestions that exosomes may constitute potential biotechnology tools and are important players of what has been designated as nanobiotechnology. They may constitute an important delivery system for gene therapy and molecular-displaying cell regulation capabilities when incorporated into other cells and even by interfering with the exosomal membrane during its biogenesis, targeting the vesicles via specific ligands to different cell types. These vesicles may reach the bloodstream, overflow through intercellular junctions, and even pass through the central nervous system blood barrier. There is evidence that it is possible to interfere with the composition of the exosomes by interfering with multivesicular body biogenesis.
Collapse
Affiliation(s)
- Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, 21941-900, Brazil. .,Instituto Nacional de Ciência e Tecnologia and Núcleo de Biologia Estrutural e Bioimagens, CENABIO, Rio de Janeiro, Brazil.
| | - Emile S Barrias
- Instituto Nacional de Ciência e Tecnologia and Núcleo de Biologia Estrutural e Bioimagens, CENABIO, Rio de Janeiro, Brazil.,Laboratorio de Metrologia Aplicada à Ciências da Vida, Diretoria de Metrologia Aplicada à Ciências da Vida - Instituto Nacional de Metrologia, Qualidade e Tecnologia (Inmetro), Rio de Janeiro, Brazil
| |
Collapse
|
38
|
Meningher T, Barsheshet Y, Ofir‐Birin Y, Gold D, Brant B, Dekel E, Sidi Y, Schwartz E, Regev‐Rudzki N, Avni O, Avni D. Schistosomal extracellular vesicle-enclosed miRNAs modulate host T helper cell differentiation. EMBO Rep 2020; 21:e47882. [PMID: 31825165 PMCID: PMC6944914 DOI: 10.15252/embr.201947882] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022] Open
Abstract
During the chronic stage of Schistosoma infection, the female lays fertile eggs, triggering a strong anti-parasitic type 2 helper T-cell (Th2) immune response. It is unclear how this Th2 response gradually declines even though the worms live for years and continue to produce eggs. Here, we show that Schistosoma mansoni downregulates Th2 differentiation in an antigen-presenting cell-independent manner, by modulating the Th2-specific transcriptional program. Adult schistosomes secrete miRNA-harboring extracellular vesicles that are internalized by Th cells in vitro. Schistosomal miRNAs are found also in T helper cells isolated from Peyer's patches and mesenteric lymph nodes of infected mice. In T helper cells, the schistosomal miR-10 targets MAP3K7 and consequently downmodulates NF-κB activity, a critical transcription factor for Th2 differentiation and function. Our results explain, at least partially, how schistosomes tune down the Th2 response, and provide further insight into the reciprocal geographic distribution between high prevalence of parasitic infections and immune disorders such as allergy. Furthermore, this worm-host crosstalk mechanism can be harnessed to develop diagnostic and therapeutic approaches for human schistosomiasis and Th2-associated diseases.
Collapse
Affiliation(s)
- Tal Meningher
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
| | | | - Yifat Ofir‐Birin
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Daniel Gold
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Boris Brant
- Azrieli Faculty of MedicineBar Ilan UniversitySafedIsrael
| | - Elya Dekel
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Yechezkel Sidi
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Faculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Eli Schwartz
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
- Faculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
- The Center for Geographic MedicineSheba Medical CenterTel HashomerIsrael
| | - Neta Regev‐Rudzki
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Orly Avni
- Azrieli Faculty of MedicineBar Ilan UniversitySafedIsrael
| | - Dror Avni
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
| |
Collapse
|
39
|
Motta FN, Azevedo CDS, Neves BP, Araújo CND, Grellier P, Santana JMD, Bastos IMD. Oligopeptidase B, a missing enzyme in mammals and a potential drug target for trypanosomatid diseases. Biochimie 2019; 167:207-216. [DOI: 10.1016/j.biochi.2019.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/15/2019] [Indexed: 12/21/2022]
|
40
|
Mandacaru SC, Queiroz RML, Alborghetti MR, de Oliveira LS, de Lima CMR, Bastos IMD, Santana JM, Roepstorff P, Ricart CAO, Charneau S. Exoproteome profiling of Trypanosoma cruzi during amastigogenesis early stages. PLoS One 2019; 14:e0225386. [PMID: 31756194 PMCID: PMC6874342 DOI: 10.1371/journal.pone.0225386] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/04/2019] [Indexed: 11/20/2022] Open
Abstract
Chagas disease is caused by the protozoan Trypanosoma cruzi, affecting around 8 million people worldwide. After host cell invasion, the infective trypomastigote form remains 2–4 hours inside acidic phagolysosomes to differentiate into replicative amastigote form. In vitro acidic-pH-induced axenic amastigogenesis was used here to study this step of the parasite life cycle. After three hours of trypomastigote incubation in amastigogenesis promoting acidic medium (pH 5.0) or control physiological pH (7.4) medium samples were subjected to three rounds of centrifugation followed by ultrafiltration of the supernatants. The resulting exoproteome samples were trypsin digested and analysed by nano flow liquid chromatography coupled to tandem mass spectrometry. Computational protein identification searches yielded 271 and 483 protein groups in the exoproteome at pH 7.4 and pH 5.0, respectively, with 180 common proteins between both conditions. The total amount and diversity of proteins released by parasites almost doubled upon acidic incubation compared to control. Overall, 76.5% of proteins were predicted to be secreted by classical or non-classical pathways and 35.1% of these proteins have predicted transmembrane domains. Classical secretory pathway analysis showed an increased number of mucins and mucin-associated surface proteins after acidic incubation. However, the number of released trans-sialidases and surface GP63 peptidases was higher at pH 7.4. Trans-sialidases and mucins are anchored to the membrane and exhibit an enzyme-substrate relationship. In general, mucins are glycoproteins with immunomodulatory functions in Chagas disease, present mainly in the epimastigote and trypomastigote surfaces and could be enzymatically cleaved and released in the phagolysosome during amastigogenesis. Moreover, evidence for flagella discard during amastigogenesis are addressed. This study provides the first comparative analysis of the exoproteome during amastigogenesis, and the presented data evidence the dynamism of its profile in response to acidic pH-induced differentiation.
Collapse
Affiliation(s)
- Samuel C. Mandacaru
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Rayner M. L. Queiroz
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Marcos R. Alborghetti
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Lucas S. de Oliveira
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Consuelo M. R. de Lima
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Izabela M. D. Bastos
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Jaime M. Santana
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Peter Roepstorff
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Carlos André O. Ricart
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Sébastien Charneau
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- * E-mail:
| |
Collapse
|
41
|
Dozio V, Lejon V, Mumba Ngoyi D, Büscher P, Sanchez JC, Tiberti N. Cerebrospinal Fluid-Derived Microvesicles From Sleeping Sickness Patients Alter Protein Expression in Human Astrocytes. Front Cell Infect Microbiol 2019; 9:391. [PMID: 31824868 PMCID: PMC6879452 DOI: 10.3389/fcimb.2019.00391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022] Open
Abstract
Human African trypanosomiasis (HAT) caused by the extracellular protozoon Trypanosoma brucei, is a neglected tropical disease affecting the poorest communities in sub-Saharan Africa. HAT progresses from a hemolymphatic first stage (S1) to a meningo-encephalitic late stage (S2) when parasites reach the central nervous system (CNS), although the existence of an intermediate stage (Int.) has also been proposed. The pathophysiological mechanisms associated with the development of S2 encephalopathy are yet to be fully elucidated. Here we hypothesized that HAT progression toward S2 might be accompanied by an increased release of microvesicles (MVs), sub-micron elements (0.1–1 μm) involved in inflammatory processes and in the determination of the outcome of infections. We studied the morphology of MVs isolated from HAT cerebrospinal fluid (CSF) by transmission electron microscopy (TEM) and used flow cytometry to show that total-MVs and leukocyte derived-CD45+ MVs are significantly increased in concentration in S2 patients' CSF compared to S1 and Int. samples (n = 12 per group). To assess potential biological properties of these MVs, immortalized human astrocytes were exposed, in vitro, to MVs enriched from S1, Int. or S2 CSF. Data-independent acquisition mass spectrometry analyses showed that S2 MVs induced, compared to Int. or S1 MVs, a strong proteome modulation in astrocytes that resembled the one produced by IFN-γ, a key molecule in HAT pathogenesis. Our results indicate that HAT S2 CSF harbors MVs potentially involved in the mechanisms of pathology associated with HAT late stage. Such vesicles might thus represent a new player to consider in future functional studies.
Collapse
Affiliation(s)
- Vito Dozio
- Translational Biomarker Group, University of Geneva, Geneva, Switzerland
| | - Veerle Lejon
- Intertryp, Institut de Recherche pour le Développement, CIRAD, University of Montpellier, Montpellier, France
| | - Dieudonné Mumba Ngoyi
- Department of Parasitology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of Congo
| | - Philippe Büscher
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Natalia Tiberti
- Translational Biomarker Group, University of Geneva, Geneva, Switzerland.,Department of Infectious - Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Verona, Italy
| |
Collapse
|
42
|
Overview of the role of kinetoplastid surface carbohydrates in infection and host cell invasion: prospects for therapeutic intervention. Parasitology 2019; 146:1743-1754. [PMID: 31603063 PMCID: PMC6939169 DOI: 10.1017/s0031182019001355] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Kinetoplastid parasites are responsible for serious diseases in humans and livestock such as Chagas disease and sleeping sickness (caused by Trypanosoma cruzi and Trypanosoma brucei, respectively), and the different forms of cutaneous, mucocutaneous and visceral leishmaniasis (produced by Leishmania spp). The limited number of antiparasitic drugs available together with the emergence of resistance underscores the need for new therapeutic agents with novel mechanisms of action. The use of agents binding to surface glycans has been recently suggested as a new approach to antitrypanosomal design and a series of peptidic and non-peptidic carbohydrate-binding agents have been identified as antiparasitics showing efficacy in animal models of sleeping sickness. Here we provide an overview of the nature of surface glycans in three kinetoplastid parasites, T. cruzi, T. brucei and Leishmania. Their role in virulence and host cell invasion is highlighted with the aim of identifying specific glycan-lectin interactions and carbohydrate functions that may be the target of novel carbohydrate-binding agents with therapeutic applications.
Collapse
|
43
|
Salas-Sarduy E, Landaburu LU, Carmona AK, Cazzulo JJ, Agüero F, Alvarez VE, Niemirowicz GT. Potent and selective inhibitors for M32 metallocarboxypeptidases identified from high-throughput screening of anti-kinetoplastid chemical boxes. PLoS Negl Trop Dis 2019; 13:e0007560. [PMID: 31329594 PMCID: PMC6675120 DOI: 10.1371/journal.pntd.0007560] [Citation(s) in RCA: 3] [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: 03/06/2019] [Revised: 08/01/2019] [Accepted: 06/18/2019] [Indexed: 11/18/2022] Open
Abstract
Enzymes of the M32 family are Zn-dependent metallocarboxypeptidases (MCPs) widely distributed among prokaryotic organisms and just a few eukaryotes including Trypanosoma brucei and Trypanosoma cruzi, the causative agents of sleeping sickness and Chagas disease, respectively. These enzymes are absent in humans and several functions have been proposed for trypanosomatid M32 MCPs. However, no synthetic inhibitors have been reported so far for these enzymes. Here, we present the identification of a set of inhibitors for TcMCP-1 and TbMCP-1 (two trypanosomatid M32 enzymes sharing 71% protein sequence identity) from the GlaxoSmithKline HAT and CHAGAS chemical boxes; two collections grouping 404 compounds with high antiparasitic potency, drug-likeness, structural diversity and scientific novelty. For this purpose, we adapted continuous fluorescent enzymatic assays to a medium-throughput format and carried out the screening of both collections, followed by the construction of dose-response curves for the most promising hits. As a result, 30 micromolar-range inhibitors were discovered for one or both enzymes. The best hit, TCMDC-143620, showed sub-micromolar affinity for TcMCP-1, inhibited TbMCP-1 in the low micromolar range and was inactive against angiotensin I-converting enzyme (ACE), a potential mammalian off-target structurally related to M32 MCPs. This is the first inhibitor reported for this family of MCPs and considering its potency and specificity, TCMDC-143620 seems to be a promissory starting point to develop more specific and potent chemical tools targeting M32 MCPs from trypanosomatid parasites.
Collapse
Affiliation(s)
- Emir Salas-Sarduy
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–Universidad Nacional de San Martín–CONICET, San Martín, B1650HMP, Buenos Aires, Argentina
| | - Lionel Urán Landaburu
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–Universidad Nacional de San Martín–CONICET, San Martín, B1650HMP, Buenos Aires, Argentina
| | - Adriana K. Carmona
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juan José Cazzulo
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–Universidad Nacional de San Martín–CONICET, San Martín, B1650HMP, Buenos Aires, Argentina
| | - Fernán Agüero
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–Universidad Nacional de San Martín–CONICET, San Martín, B1650HMP, Buenos Aires, Argentina
| | - Vanina E. Alvarez
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–Universidad Nacional de San Martín–CONICET, San Martín, B1650HMP, Buenos Aires, Argentina
| | - Gabriela T. Niemirowicz
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–Universidad Nacional de San Martín–CONICET, San Martín, B1650HMP, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
44
|
Dong G, Filho AL, Olivier M. Modulation of Host-Pathogen Communication by Extracellular Vesicles (EVs) of the Protozoan Parasite Leishmania. Front Cell Infect Microbiol 2019; 9:100. [PMID: 31032233 PMCID: PMC6470181 DOI: 10.3389/fcimb.2019.00100] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/25/2019] [Indexed: 12/21/2022] Open
Abstract
Leishmania genus protozoan parasites have developed various strategies to overcome host cell protective mechanisms favoring their survival and propagation. Recent findings in the field propose a new player in this infectious strategy, the Leishmania exosomes. Exosomes are eukaryotic extracellular vesicles essential to cell communication in various biological contexts. In fact, there have been an increasing number of reports over the last 10 years regarding the role of protozoan parasite exosomes, Leishmania exosomes included, in their capacity to favor infection and propagation within their hosts. In this review, we will discuss the latest findings regarding Leishmania exosome function during infectious conditions with a strong focus on Leishmania-host interaction from a mammalian perspective. We also compare the immunomodulatory properties of Leishmania exosomes to other parasite exosomes, demonstrating the conserved, important role that exosomes play during parasite infection.
Collapse
Affiliation(s)
- George Dong
- Infectious Diseases and Immunity in Global Heath Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Alonso Lira Filho
- Infectious Diseases and Immunity in Global Heath Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Martin Olivier
- Infectious Diseases and Immunity in Global Heath Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
45
|
Gonçalves DDS, Ferreira MDS, Guimarães AJ. Extracellular Vesicles from the Protozoa Acanthamoeba castellanii: Their Role in Pathogenesis, Environmental Adaptation and Potential Applications. Bioengineering (Basel) 2019; 6:bioengineering6010013. [PMID: 30717103 PMCID: PMC6466093 DOI: 10.3390/bioengineering6010013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/22/2019] [Accepted: 01/26/2019] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are membranous compartments of distinct cellular origin and biogenesis, displaying different sizes and include exosomes, microvesicles, and apoptotic bodies. The EVs have been described in almost every living organism, from simple unicellular to higher evolutionary scale multicellular organisms, such as mammals. Several functions have been attributed to these structures, including roles in energy acquisition, cell-to-cell communication, gene expression modulation and pathogenesis. In this review, we described several aspects of the recently characterized EVs of the protozoa Acanthamoeba castellanii, a free-living amoeba (FLA) of emerging epidemiological importance, and compare their features to other parasites' EVs. These A. castellanii EVs are comprised of small microvesicles and exosomes and carry a wide range of molecules involved in many biological processes like cell signaling, carbohydrate metabolism and proteolytic activity, such as kinases, glucanases, and proteases, respectively. Several biomedical applications of these EVs have been proposed lately, including their use in vaccination, biofuel production, and the pharmaceutical industry, such as platforms for drug delivery.
Collapse
Affiliation(s)
- Diego de Souza Gonçalves
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro 24210-130, Brazil.
| | - Marina da Silva Ferreira
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-970, Brazil.
| | - Allan J Guimarães
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro 24210-130, Brazil.
| |
Collapse
|
46
|
Wu Z, Wang L, Li J, Wang L, Wu Z, Sun X. Extracellular Vesicle-Mediated Communication Within Host-Parasite Interactions. Front Immunol 2019; 9:3066. [PMID: 30697211 PMCID: PMC6340962 DOI: 10.3389/fimmu.2018.03066] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are small membrane-surrounded structures released by different kinds of cells (normal, diseased, and transformed cells) in vivo and in vitro that contain large amounts of important substances (such as lipids, proteins, metabolites, DNA, RNA, and non-coding RNA (ncRNA), including miRNA, lncRNA, tRNA, rRNA, snoRNA, and scaRNA) in an evolutionarily conserved manner. EVs, including exosomes, play a role in the transmission of information, and substances between cells that is increasingly being recognized as important. In some infectious diseases such as parasitic diseases, EVs have emerged as a ubiquitous mechanism for mediating communication during host-parasite interactions. EVs can enable multiple modes to transfer virulence factors and effector molecules from parasites to hosts, thereby regulating host gene expression, and immune responses and, consequently, mediating the pathogenic process, which has made us rethink our understanding of the host-parasite interface. Thus, here, we review the present findings regarding EVs (especially exosomes) and recognize the role of EVs in host-parasite interactions. We hope that a better understanding of the mechanisms of parasite-derived EVs may provide new insights for further diagnostic biomarker, vaccine, and therapeutic development.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Lingling Wang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Jiaying Li
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Lifu Wang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Zhongdao Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| |
Collapse
|
47
|
A Major Step towards Defining the Elusive Stumpy Inducing Factor in Trypanosoma brucei. Trends Parasitol 2019; 35:6-8. [PMID: 30554967 DOI: 10.1016/j.pt.2018.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/16/2022]
Abstract
Trypanosoma brucei stumpy forms are the only stage that can transmit from human to tsetse fly. Stumpy formation is regulated by a quorum sensing mechanism that depends on parasite density and an unknown stumpy induction factor (SIF). Recently, an elegant study by Matthews and colleagues (Cell 176, 1-12) has identified several crucial components of this pathway, including the putative SIF and its receptor.
Collapse
|
48
|
Gonçalves DDS, Ferreira MDS, Liedke SC, Gomes KX, de Oliveira GA, Leão PEL, Cesar GV, Seabra SH, Cortines JR, Casadevall A, Nimrichter L, Domont GB, Junqueira MR, Peralta JM, Guimaraes AJ. Extracellular vesicles and vesicle-free secretome of the protozoa Acanthamoeba castellanii under homeostasis and nutritional stress and their damaging potential to host cells. Virulence 2018; 9:818-836. [PMID: 29560793 PMCID: PMC5955443 DOI: 10.1080/21505594.2018.1451184] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022] Open
Abstract
Acanthamoeba castellanii (Ac) are ubiquitously distributed in nature, and by contaminating medical devices such as heart valves and contact lenses, they cause a broad range of clinical presentations to humans. Although several molecules have been described to play a role in Ac pathogenesis, including parasite host-tissue invasion and escaping of host-defense, little information is available on their mechanisms of secretion. Herein, we describe the molecular components secreted by Ac, under different protein availability conditions to simulate host niches. Ac extracellular vesicles (EVs) were morphologically and biochemically characterized. Dynamic light scattering analysis of Ac EVs identified polydisperse populations, which correlated to electron microscopy measurements. High-performance thin liquid chromatography of Ac EVs identified phospholipids, steryl-esters, sterol and free-fatty acid, the last two also characterized by GC-MS. Secretome composition (EVs and EVs-free supernatants) was also determined and proteins biological functions classified. In peptone-yeast-glucose (PYG) medium, a total of 179 proteins were identified (21 common proteins, 89 exclusive of EVs and 69 in EVs-free supernatant). In glucose alone, 205 proteins were identified (134 in EVs, 14 common and 57 proteins in EVs-free supernatant). From those, stress response, oxidative and protein and amino acid metabolism proteins prevailed. Qualitative differences were observed on carbohydrate metabolism enzymes from Krebs cycle and pentose phosphate shunt. Serine proteases and metalloproteinases predominated. Analysis of the cytotoxicity of Ac EVs (upon uptake) and EVs-free supernatant to epithelial and glioblastoma cells revealed a dose-dependent effect. Therefore, the Ac secretome differs depending on nutrient conditions, and is also likely to vary during infection.
Collapse
Affiliation(s)
- Diego de Souza Gonçalves
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
| | - Marina da Silva Ferreira
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susie Coutinho Liedke
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kamilla Xavier Gomes
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
| | - Gabriel Afonso de Oliveira
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
| | - Pedro Ernesto Lopes Leão
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriele Vargas Cesar
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio H. Seabra
- Laboratório de Tecnologia em Cultura de Células, Centro Universitário Estadual da Zona Oeste (UEZO), Rio de Janeiro, Brazil
| | - Juliana Reis Cortines
- Departamento de Virologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Leonardo Nimrichter
- Laboratório de Glicobiologia de Eucariotos, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto Barbosa Domont
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Magno Rodrigues Junqueira
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose Mauro Peralta
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Allan J. Guimaraes
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
49
|
Rojas F, Silvester E, Young J, Milne R, Tettey M, Houston DR, Walkinshaw MD, Pérez-Pi I, Auer M, Denton H, Smith TK, Thompson J, Matthews KR. Oligopeptide Signaling through TbGPR89 Drives Trypanosome Quorum Sensing. Cell 2018; 176:306-317.e16. [PMID: 30503212 PMCID: PMC6333907 DOI: 10.1016/j.cell.2018.10.041] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/13/2018] [Accepted: 10/17/2018] [Indexed: 02/03/2023]
Abstract
Trypanosome parasites control their virulence and spread by using quorum sensing (QS) to generate transmissible “stumpy forms” in their host bloodstream. However, the QS signal “stumpy induction factor” (SIF) and its reception mechanism are unknown. Although trypanosomes lack G protein-coupled receptor signaling, we have identified a surface GPR89-family protein that regulates stumpy formation. TbGPR89 is expressed on bloodstream “slender form” trypanosomes, which receive the SIF signal, and when ectopically expressed, TbGPR89 drives stumpy formation in a SIF-pathway-dependent process. Structural modeling of TbGPR89 predicts unexpected similarity to oligopeptide transporters (POT), and when expressed in bacteria, TbGPR89 transports oligopeptides. Conversely, expression of an E. coli POT in trypanosomes drives parasite differentiation, and oligopeptides promote stumpy formation in vitro. Furthermore, the expression of secreted trypanosome oligopeptidases generates a paracrine signal that accelerates stumpy formation in vivo. Peptidase-generated oligopeptide QS signals being received through TbGPR89 provides a mechanism for both trypanosome SIF production and reception. Trypanosomes use quorum sensing to differentiate to transmissible stumpy forms A GPR89 protein with oligopeptide transport activity drives parasite differentiation Oligopeptide mixtures and synthetic di- and tripeptides promote stumpy formation Released parasite oligopeptidases generate the paracrine quorum sensing signal in vivo
Collapse
Affiliation(s)
- Federico Rojas
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Eleanor Silvester
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Julie Young
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Rachel Milne
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Mabel Tettey
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Douglas R Houston
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Malcolm D Walkinshaw
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Irene Pérez-Pi
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Manfred Auer
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Helen Denton
- School of Biology, BSRC, University of St. Andrews, North Haugh, St. Andrews, Fife KY16 9ST, UK
| | - Terry K Smith
- School of Biology, BSRC, University of St. Andrews, North Haugh, St. Andrews, Fife KY16 9ST, UK
| | - Joanne Thompson
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| | - Keith R Matthews
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| |
Collapse
|
50
|
Geiger A, Malele I, Abd-Alla AM, Njiokou F. Blood feeding tsetse flies as hosts and vectors of mammals-pre-adapted African Trypanosoma: current and expected research directions. BMC Microbiol 2018; 18:162. [PMID: 30470183 PMCID: PMC6251083 DOI: 10.1186/s12866-018-1281-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Research on the zoo-anthropophilic blood feeding tsetse flies' biology conducted, by different teams, in laboratory settings and at the level of the ecosystems- where also co-perpetuate African Trypanosoma- has allowed to unveil and characterize key features of tsetse flies' bacterial symbionts on which rely both (a) the perpetuation of the tsetse fly populations and (b) the completion of the developmental program of the African Trypanosoma. Transcriptomic analyses have already provided much information on tsetse fly genes as well as on genes of the fly symbiotic partners Sodalis glossinidius and Wigglesworthia, which account for the successful onset or not of the African Trypanosoma developmental program. In parallel, identification of the non- symbiotic bacterial communities hosted in the tsetse fly gut has recently been initiated: are briefly introduced those bacteria genera and species common to tsetse flies collected from distinct ecosystems, that could be further studied as potential biologicals preventing the onset of the African Trypanosoma developmental program. Finally, future work will need to concentrate on how to render tsetse flies refractory, and the best means to disseminate them in the field in order to establish an overall refractory fly population.
Collapse
Affiliation(s)
- Anne Geiger
- INTERTRYP, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
| | - Imna Malele
- Vector and Vector Borne Diseases Institute, Majani Mapana, Off Korogwe Road, Box, 1026 Tanga, Tanzania
| | - Adly M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, Vienna, Austria
| | - Flobert Njiokou
- Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| |
Collapse
|