1
|
Rosa RSL, Leal da Silva M, Bernardi RC. Atomistic Insights into gp82 Binding: A Microsecond, Million-Atom Exploration of Trypanosoma cruzi Host-Cell Invasion. Biochemistry 2025. [PMID: 40152296 DOI: 10.1021/acs.biochem.4c00710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, affects millions globally, leading to severe cardiac and gastrointestinal complications in its chronic phase. The invasion of host cells by T. cruzi is mediated by the interaction between the parasite's glycoprotein gp82 and the human receptor lysosome-associated membrane protein 2 (LAMP2). While experimental studies have identified a few residues involved in this interaction, a comprehensive molecular-level understanding has been lacking. In this study, we present a 1.44-million-atom computational model of the gp82 complex, including over 3300 lipids, glycosylation sites, and full molecular representations of gp82 and LAMP2, making it the most complete model of a parasite-host interaction to date. Using microsecond-long molecular dynamics simulations and dynamic network analysis, we identified critical residue interactions, including novel regions of contact that were previously uncharacterized. Our findings also highlight the significance of the transmembrane domain of LAMP2 in stabilizing the complex. These insights extend beyond traditional hydrogen bond interactions, revealing a complex network of cooperative motions that facilitate T. cruzi invasion. This study not only confirms key experimental observations but also uncovers new molecular targets for therapeutic intervention, offering a potential pathway to disrupt T. cruzi infection and combat Chagas disease.
Collapse
Affiliation(s)
- Raissa S L Rosa
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, United States
- Programa de Pós-Graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ 21040-360, Brazil
| | - Manuela Leal da Silva
- Programa de Pós-Graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ 21040-360, Brazil
- Instituto de Biodiversidade e Sustentabilidade (NUPEM), Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil
| | - Rafael C Bernardi
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
2
|
Hirosawa KM, Sato Y, Kasai RS, Yamaguchi E, Komura N, Ando H, Hoshino A, Yokota Y, Suzuki KGN. Uptake of small extracellular vesicles by recipient cells is facilitated by paracrine adhesion signaling. Nat Commun 2025; 16:2419. [PMID: 40075063 PMCID: PMC11903687 DOI: 10.1038/s41467-025-57617-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Small extracellular vesicles (sEVs) play crucial roles in intercellular communication. However, the internalization of individual sEVs by recipient cells has not been directly observed. Here, we examined these mechanisms using state-of-the-art imaging techniques. Single-molecule imaging shows that tumor-derived sEVs can be classified into several subtypes. Simultaneous single-sEV particle tracking and observation of super-resolution movies of membrane invaginations in living cells reveal that all sEV subtypes are internalized via clathrin-independent endocytosis mediated by galectin-3 and lysosome-associated membrane protein-2C, while some subtypes that recruited raft markers are internalized through caveolae. Integrin β1 and talin-1 accumulate in recipient cell plasma membranes beneath all sEV subtypes. Paracrine, but not autocrine, sEV binding triggers Ca2+ mobilization induced by the activation of Src family kinases and phospholipase Cγ. Subsequent Ca2+-induced activation of calcineurin-dynamin promotes sEV internalization, leading to the recycling pathway. Thus, we clarified the detailed mechanisms of sEV internalization driven by paracrine adhesion signaling.
Collapse
Affiliation(s)
- Koichiro M Hirosawa
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Yusuke Sato
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, 980-8578, Japan
| | - Rinshi S Kasai
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, 104-0045, Japan
| | - Eriko Yamaguchi
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Naoko Komura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Hiromune Ando
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 606-8501, Japan
- Innovation Research Center for Quantum Medicine. Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan
| | - Ayuko Hoshino
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, 153-8904, Japan
- Inamori Research Institute for Science, Inamori Foundation, Kyoto, 600-8411, Japan
| | - Yasunari Yokota
- Department of Electrical, Electronics and Computer Engineering, Faculty of Engineering, Gifu University, Gifu, 501-1193, Japan
| | - Kenichi G N Suzuki
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan.
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, 104-0045, Japan.
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 606-8501, Japan.
- Innovation Research Center for Quantum Medicine. Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan.
| |
Collapse
|
3
|
Ansa‐Addo EA, Pathak P, McCrossan MV, Volpato Rossi I, Abdullahi M, Stratton D, Lange S, Ramirez MI, Inal JM. Monocyte-derived extracellular vesicles, stimulated by Trypanosoma cruzi, enhance cellular invasion in vitro via activated TGF-β1. J Extracell Vesicles 2024; 13:e70014. [PMID: 39611395 PMCID: PMC11605483 DOI: 10.1002/jev2.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/06/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024] Open
Abstract
During cell invasion, large Extracellular Vesicle (lEV) release from host cells was dose-dependently triggered by Trypanosoma cruzi metacyclic trypomastigotes (Mtr). This lEV release was inhibited when IP3-mediated Ca2+ exit from the ER and further Ca2+ entry from plasma membrane channels was blocked, but whilst any store-independent Ca2+ entry (SICE) could continue unabated. That lEV release was equally inhibited if all entry from external sources was blocked by chelation of external Ca2+ points to the major contributor to Mtr-triggered host cell lEV release being IP3/store-mediated Ca2+ release, SICE playing a minor role. Host cell lEVs were released through Mtr interaction with host cell lipid raft domains, integrins, and mechanosensitive ion channels, whereupon [Ca2+]cyt increased (50 to 750 nM) within 15 s. lEV release and cell entry of T. cruzi, which increased up to 30 and 60 mpi, respectively, as well as raised actin depolymerization at 60 mpi, were all reduced by TRPC inhibitor, GsMTx-4. Vesicle release and infection was also reduced with RGD peptide, methyl-β-cyclodextrin, knockdown of calpain and with the calpain inhibitor, calpeptin. Restoration of lEV levels, whether with lEVs from infected or uninfected epithelial cells, did not restore invasion, but supplementation with lEVs from infected monocytes, did. We provide evidence of THP-1 monocyte-derived lEV interaction with Mtr (lipid mixing by R18-dequenching; flow cytometry showing transfer to Mtr of R18 from R18-lEVs and of LAP(TGF-β1). Active, mature TGF-β1 (at 175 pg/×105 in THP-1 lEVs) was detected in concentrated lEV-/cell-free supernatant by western blotting, only after THP-1 lEVs had interacted with Mtr. The TGF-β1 receptor (TβRI) inhibitor, SB-431542, reduced the enhanced cellular invasion due to monocyte-lEVs.
Collapse
Affiliation(s)
- Ephraim A. Ansa‐Addo
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- Pelotonia Institute for Immuno‐Oncology, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Paras Pathak
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- Medical Research Council HarwellHarwell Science and Innovation Campus, Genotyping CoreOxfordshireUK
| | | | - Izadora Volpato Rossi
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- School of Life and Medical Sciences, Biosciences Research GroupUniversity of HertfordshireHatfieldUK
- Carlos Chagas InstituteFundacao Oswaldo Cruz, (FIOCRUZ‐PR)CuritibaBrazil
- Postgraduate Program in Cellular and Molecular BiologyFederal University of ParanáCuritibaBrazil
| | - Mahamed Abdullahi
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- National Mycobacterium Reference Service‐South (NMRS‐South) ColindaleLondonUK
| | - Dan Stratton
- School of Life, Health & Chemical SciencesThe Open UniversityMilton KeynesUK
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life SciencesUniversity of WestminsterLondonUK
- University College London, Institute of Women's HealthLondonUK
| | - Marcel I. Ramirez
- Carlos Chagas InstituteFundacao Oswaldo Cruz, (FIOCRUZ‐PR)CuritibaBrazil
| | - Jameel M. Inal
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- School of Life and Medical Sciences, Biosciences Research GroupUniversity of HertfordshireHatfieldUK
| |
Collapse
|
4
|
Rosa RSL, Leal da Silva M, Bernardi RC. Atomistic Insights into gp82 Binding: A Microsecond, Million-Atom Exploration of Trypanosoma cruzi Host-Cell Invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619626. [PMID: 39484421 PMCID: PMC11526924 DOI: 10.1101/2024.10.22.619626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi , affects millions globally, leading to severe cardiac and gastrointestinal complications in its chronic phase. The invasion of host cells by T. cruzi is mediated by the interaction between the parasite's glycoprotein gp82 and the human receptor lysosome-associated membrane protein 2 (LAMP2). While experimental studies have identified a few residues involved in this interaction, a comprehensive molecular-level understanding has been lacking. In this study, we present a 1.44-million-atom computational model of the gp82 complex, including over 3,300 lipids, glycosylation sites, and full molecular representations of gp82 and LAMP2, making it the most complete model of a parasite-host interaction to date. Using microsecond-long molecular dynamics simulations and dynamic network analysis, we identified critical residue interactions, including novel regions of contact that were previously uncharacterized. Our findings also highlight the significance of the transmembrane domain of LAMP2 in stabilizing the complex. These insights extend beyond traditional hydrogen bond interactions, revealing a complex network of cooperative motions that facilitate T. cruzi invasion. This study not only confirms key experimental observations but also uncovers new molecular targets for therapeutic intervention, offering a potential pathway to disrupt T. cruzi infection and combat Chagas disease.
Collapse
|
5
|
Rossi IV, de Souza DAS, Ramirez MI. The End Justifies the Means: Chagas Disease from a Perspective of the Host- Trypanosoma cruzi Interaction. Life (Basel) 2024; 14:488. [PMID: 38672758 PMCID: PMC11050810 DOI: 10.3390/life14040488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
The neglected Chagas disease (CD) is caused by the protozoan parasite Trypanosoma cruzi. Despite CD dispersion throughout the world, it prevails in tropical areas affecting mainly poor communities, causing devastating health, social and economic consequences. Clinically, CD is marked by a mildly symptomatic acute phase, and a chronic phase characterized by cardiac and/or digestive complications. Current treatment for CD relies on medications with strong side effects and reduced effectiveness. The complex interaction between the parasite and the host outlines the etiology and progression of CD. The unique characteristics and high adaptability of T. cruzi, its mechanisms of persistence, and evasion of the immune system seem to influence the course of the disease. Despite the efforts to uncover the pathology of CD, there are many gaps in understanding how it is established and reaches chronicity. Also, the lack of effective treatments and protective vaccines constitute challenges for public health. Here, we explain the background in which CD is established, from the peculiarities of T. cruzi molecular biology to the development of the host's immune response leading to the pathophysiology of CD. We also discuss the state of the art of treatments for CD and current challenges in basic and applied science.
Collapse
Affiliation(s)
- Izadora Volpato Rossi
- Graduate Program in Microbiology, Parasitology and Pathology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil;
- Laboratory of Cell Biology, Carlos Chagas Institute/Oswaldo Cruz Foundation (FIOCRUZ-PR), Curitiba 81310-020, PR, Brazil;
| | - Denise Andréa Silva de Souza
- Laboratory of Cell Biology, Carlos Chagas Institute/Oswaldo Cruz Foundation (FIOCRUZ-PR), Curitiba 81310-020, PR, Brazil;
| | - Marcel Ivan Ramirez
- Graduate Program in Microbiology, Parasitology and Pathology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil;
- Laboratory of Cell Biology, Carlos Chagas Institute/Oswaldo Cruz Foundation (FIOCRUZ-PR), Curitiba 81310-020, PR, Brazil;
| |
Collapse
|
6
|
Alvim JM, Venturini G, Oliveira TGM, Seidman JG, Seidman CE, Krieger JE, Pereira AC. mTOR signaling inhibition decreases lysosome migration and impairs the success of Trypanosoma cruzi infection and replication in cardiomyocytes. Acta Trop 2023; 240:106845. [PMID: 36709791 DOI: 10.1016/j.actatropica.2023.106845] [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: 11/01/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
Chagas disease is caused by the parasite Trypanosoma cruzi (T. cruzi) and, among all the chronic manifestations of the disease, Chronic Chagas Cardiomyopathy (CCC) is the most severe outcome. Despite high burden and public health importance in Latin America, there is a gap in understanding the molecular mechanisms that results in CCC development. Previous studies showed that T. cruzi uses the host machinery for infection and replication, including the repurposing of the responses to intracellular infection such as mitochondrial activity, vacuolar membrane, and lysosomal activation in benefit of parasite infection and replication. One common signaling upstream to many responses to parasite infection is mTOR pathway, previous associated to several downstream cellular mechanisms including autophagy, mitophagy and lysosomal activation. Here, using human iPSC derived cardiomyocytes (hiPSCCM), we show the mTOR pathway is activated in hiPSCCM after T. cruzi infection, and the inhibition of mTOR with rapamycin reduced number of T. cruzi 48 h post infection (hpi). Rapamycin treatment also reduced lysosome migration from nuclei region to cell periphery resulting in less T. cruzi inside the parasitophorous vacuole (PV) in the first hour of infection. In addition, the number of parasites leaving the PV to the cytoplasm to replicate in later times of infection was also lower after rapamycin treatment. Altogether, our data suggest that host's mTOR activation concomitant with parasite infection modulates lysosome migration and that T. cruzi uses this mechanism to achieve infection and replication. Modulating this mechanism with rapamycin impaired the success of T. cruzi life cycle independent of mitophagy.
Collapse
Affiliation(s)
- Juliana M Alvim
- Heart Institute, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil; Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil
| | - Gabriela Venturini
- Heart Institute, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil; Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil; Department of Genetics, Harvard Medical School, United States.
| | - Theo G M Oliveira
- Heart Institute, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil; Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil; Fundação Pró-Sangue Hemocentro de São Paulo, Brazil
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, United States; Brigham and Women's Hospital, Harvard Medical School, United States; Howard Hughes Medical Institute (HHMI), United States
| | - José E Krieger
- Heart Institute, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil; Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil
| | - Alexandre C Pereira
- Heart Institute, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil; Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Brazil; Department of Genetics, Harvard Medical School, United States
| |
Collapse
|
7
|
Garcez EM, Gomes N, Moraes AS, Pogue R, Uenishi RH, Hecht M, Carvalho JL. Extracellular vesicles in the context of Chagas Disease - A systematic review. Acta Trop 2023; 242:106899. [PMID: 36935050 DOI: 10.1016/j.actatropica.2023.106899] [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: 01/25/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
Extracellular vesicle (EVs) traffic is considered an important cellular communication process between cells that can be part of a single organism or belong to different living beings. The relevance of EV-mediated cellular communication is increasingly studied and appreciated, especially in relation to pathological conditions, including parasitic disorders, in which the EV release and uptake processes have been documented. In the context of Chagas Disease (CD), EVs have been explored, however, current data have not been systematically revised in order to provide an overview of the published literature and the main results obtained thus far. In this systematic review, 25 studies involving the investigation of EVs in CD were identified. The studies involved Trypanosoma cruzi (Tc)-derived EVs (Tc-EVs), as well as EVs derived from T. cruzi-infected mammalian cells-derived EVs, mainly isolated by ultracentrifugation and poorly characterized. The objectives of the identified studies included the characterization of the protein and RNA cargo of Tc-EVs, as well as investigation of EVs in parasitic infections and immune-related processes. Overall, our systematic review reveals that EVs play critical roles in several mechanisms related to the interaction between T. cruzi and mammalian hosts, their contribution to immune system evasion by the parasite, and to chronic inflammation in the host. Future studies will benefit from the consolidation of isolation and characterization methods, as well as the elucidation of the role of EVs in CD.
Collapse
Affiliation(s)
- Emãnuella Melgaço Garcez
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Nélio Gomes
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Aline Silva Moraes
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Robert Pogue
- Genomic Sciences and Biotechnology Program. Catholic University of Brasília, 71966-700, Brasília, DF, Brazil
| | - Rosa Harumi Uenishi
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Mariana Hecht
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil
| | - Juliana Lott Carvalho
- Multidisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, 70910-900, Brasília, DF, Brazil; Genomic Sciences and Biotechnology Program. Catholic University of Brasília, 71966-700, Brasília, DF, Brazil.
| |
Collapse
|
8
|
Selvapandiyan A, Puri N, Kumar P, Alam A, Ehtesham NZ, Griffin G, Hasnain SE. Zooming in on common immune evasion mechanisms of pathogens in phagolysosomes: potential broad-spectrum therapeutic targets against infectious diseases. FEMS Microbiol Rev 2023; 47:6780197. [PMID: 36309472 DOI: 10.1093/femsre/fuac041] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 01/19/2023] Open
Abstract
The intracellular viral, bacterial, or parasitic pathogens evade the host immune challenges to propagate and cause fatal diseases. The microbes overpower host immunity at various levels including during entry into host cells, phagosome formation, phagosome maturation, phagosome-lysosome fusion forming phagolysosomes, acidification of phagolysosomes, and at times after escape into the cytosol. Phagolysosome is the final organelle in the phagocyte with sophisticated mechanisms to degrade the pathogens. The immune evasion strategies by the pathogens include the arrest of host cell apoptosis, decrease in reactive oxygen species, the elevation of Th2 anti-inflammatory response, avoidance of autophagy and antigen cross-presentation pathways, and escape from phagolysosomal killing. Since the phagolysosome organelle in relation to infection/cure is seldom discussed in the literature, we summarize here the common host as well as pathogen targets manipulated or utilized by the pathogens established in phagosomes and phagolysosomes, to hijack the host immune system for their benefit. These common molecules or pathways can be broad-spectrum therapeutic targets for drug development for intervention against infectious diseases caused by different intracellular pathogens.
Collapse
Affiliation(s)
| | - Niti Puri
- Cellular and Molecular Immunology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pankaj Kumar
- Department of Biochemistry, Jamia Hamdard, New Delhi, 110062, India.,Centre for Tuberculosis Research, Department of Medicine, Johns Hopkins University, Baltimore, MD, 21218, United States
| | - Anwar Alam
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India.,Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India
| | - Nasreen Zafar Ehtesham
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
| | - George Griffin
- Department of Cellular and Molecular Medicine, St. George's University of London, London, SW17 0RE, United Kingdom
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India.,Department of Life Science, School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, 201310, India
| |
Collapse
|
9
|
Depletion of Na+/H+ Exchanger Isoform 1 Increases the Host Cell Resistance to Trypanosoma cruzi Invasion. Pathogens 2022; 11:pathogens11111294. [DOI: 10.3390/pathogens11111294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Na+/H+ exchanger isoform 1 (NHE1), a member of a large family of integral membrane proteins, plays a role in regulating the cortical actin cytoskeleton. Trypanosoma cruzi, the agent of Chagas disease, depends on F-actin rearrangement and lysosome mobilization to invade host cells. To determine the involvement of NHE1 in T. cruzi metacyclic trypomastigote (MT) internalization, the effect of treatment in cells with NHE1 inhibitor amiloride or of NHE1 depletion was examined in human epithelial cells. MT invasion decreased in amiloride-treated and NHE1-depleted cells. The phosphorylation profile of diverse protein kinases, whose activation is associated with remodeling of actin fibers, was analyzed in amiloride-treated and NHE1-depleted cells. In amiloride-treated cells, the phosphorylation levels of protein kinase C (PKC), focal adhesion kinase (FAK) and Akt were similar to those of untreated cells, whereas those of extracellular signal-regulated protein kinases (ERK1/2) increased. In NHE1-deficient cells, with marked alteration in the actin cytoskeleton architecture and in lysosome distribution, the levels of phospho-PKC and phospho-FAK decreased, whereas those of phospho-Akt and phospho-ERK1/2 increased. These data indicate that NHE1 plays a role in MT invasion, by maintaining the activation status of diverse protein kinases in check and preventing the inappropriate F-actin arrangement that affects lysosome distribution.
Collapse
|
10
|
Onofre TS, Loch L, Ferreira Rodrigues JP, Macedo S, Yoshida N. Gp35/50 mucin molecules of Trypanosoma cruzi metacyclic forms that mediate host cell invasion interact with annexin A2. PLoS Negl Trop Dis 2022; 16:e0010788. [PMID: 36190932 PMCID: PMC9529151 DOI: 10.1371/journal.pntd.0010788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/05/2022] [Indexed: 11/07/2022] Open
Abstract
Host cell invasion is a critical step for infection by Trypanosoma cruzi, the agent of Chagas disease. In natural infection, T. cruzi metacyclic trypomastigote (MT) forms establish the first interaction with host cells. The gp35/50 mucin molecules expressed in MT have been implicated in cell invasion process, but the mechanisms involved are not well understood. We performed a series of experiments to elucidate the mode of gp35/50-mediated MT internalization. Comparing two parasite strains from genetically divergent groups, G strain (TcI) and CL strain (TcVI), expressing variant forms of mucins, we demonstrated that G strain mucins participate in MT invasion. Only G strain-derived mucins bound to HeLa cells in a receptor-dependent manner and significantly inhibited G strain MT invasion. CL strain MT internalization was not affected by mucins from either strain. HeLa cell invasion by G strain MT was associated with actin recruitment and did not rely on lysosome mobilization. To examine the involvement of annexin A2, which plays a role in actin dynamic, annexin A2-depleted HeLa cells were generated. Annexin A2-deficient cell lines were significantly more resistant than wild type controls to G strain MT invasion. In a co-immunoprecipitation assay, to check whether annexin A2 might be the receptor for mucins, protein A/G magnetic beads crosslinked with monoclonal antibody to G strain mucins were incubated with detergent extracts of MT and HeLa cells. Binding of gp35/50 mucins to annexin A2 was detected. Both G strain MT and purified mucins induced focal adhesion kinase activation in HeLa cells. By confocal immunofluorescence microscopy, colocalization of invading G strain MT with clathrin was visualized. Inhibition of clathrin-coated vesicle formation reduced parasite internalization. Taken together, our data indicate that gp35/50-mediated MT invasion is accomplished through interaction with host cell annexin A2 and clathrin-dependent endocytosis. Host cell invasion by Trypanosoma cruzi, the agent of Chagas disease, is critical for the establishment of infection. Metacyclic trypomastigote (MT) forms are responsible for the initial T. cruzi-host cell interaction. Mucin molecules expressed on MT surface have been implicated in target cell invasion process, but the underlying mechanism are not fully understood. In this study, we aimed at elucidating the mode of mucin-mediated MT internalization. We found that requirement of mucins for MT invasion is T. cruzi strain-dependent. Experiments with G strain MTs, which rely on mucins and on target cell actin for internalization, revealed that mucin molecules bind to annexin A2, a protein that plays a role in actin dynamic. Annexin A2-deficient cell lines were generated and found to be significantly more resistant than wild type controls to MT invasion. Both MT and purified mucins induced focal adhesion kinase activation in host cells. By confocal immunofluorescence microscopy, invading MT was found to colocalize with clathrin, a protein that plays a role in endocytosis. Inhibition of clathrin-coated vesicle formation reduced parasite internalization. From these data we infer that mucin-mediated MT invasion is accomplished through interaction with host cell annexin A2 and clathrin-dependent endocytosis.
Collapse
Affiliation(s)
- Thiago Souza Onofre
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Leonardo Loch
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - João Paulo Ferreira Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Silene Macedo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil,* E-mail:
| |
Collapse
|
11
|
Spirandelli da Costa M, Borges BC, Marques IT, de Oliveira RC, Teixeira TL, de Gouveia Santos J, Silva CVD. Pentachloropseudilin treatment impairs host cell invasion by Trypanosoma cruzi. Chembiochem 2022; 23:e202200349. [PMID: 35839379 DOI: 10.1002/cbic.202200349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/14/2022] [Indexed: 11/08/2022]
Abstract
Pentachloropseudilin (PClP) is a reversible and allosteric inhibitor of type 1 myosin. Here, we addressed the impact of PClP treatment of Trypanosoma cruzi and mammalian host cell on the parasite migration, cell adhesion and invasion. We observed that PClP was not toxic to either T. cruzi or host cell. Moreover, treatment of T. cruzi with PClP inhbited parasite motility, host cell adhesion and invasion. Treatment of host cell with PClP also impaired parasite invasion probably by decreasing lysosome migration to the entry site of the parasite. Therefore, PClP treatment impaired fundamental processes necessary for a successful T. cruzi infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Claudio Vieira da Silva
- Universidade Federal de Uberlândia, Imunologia, Rua Piauí, Bloco 2B sala 200, 38400096, Uberlândia, BRAZIL
| |
Collapse
|
12
|
Knüsel S, Jenni A, Benninger M, Bütikofer P, Roditi I. Persistence of Trypanosoma brucei as early procyclic forms and social motility are dependent on glycosylphosphatidylinositol transamidase. Mol Microbiol 2021; 117:802-817. [PMID: 34954848 PMCID: PMC9303471 DOI: 10.1111/mmi.14873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 12/04/2022]
Abstract
Glycosylphosphatidylinositol (GPI)‐linked molecules are surface‐exposed membrane components that influence the infectivity, virulence and transmission of many eukaryotic pathogens. Procyclic (insect midgut) forms of Trypanosoma brucei do not require GPI‐anchored proteins for growth in suspension culture. Deletion of TbGPI8, and inactivation of the GPI:protein transamidase complex, is tolerated by cultured procyclic forms. Using a conditional knockout, we show TbGPI8 is required for social motility (SoMo). This collective migration by cultured early procyclic forms has been linked to colonization of the tsetse fly digestive tract. The SoMo‐negative phenotype was observed after a lag phase with respect to loss of TbGPI8 and correlated with an unexpectedly slow loss of procyclins, the major GPI‐anchored proteins. Procyclins are not essential for SoMo, however, suggesting a requirement for at least one other GPI‐anchored protein. Loss of TbGPI8 initiates the transition from early to late procyclic forms; this effect was observed in a subpopulation in suspension culture, and was more pronounced when cells were cultured on SoMo plates. Our results indicate two, potentially interlinked, scenarios that may explain the previously reported failure of TbGPI8 deletion mutants to establish a midgut infection in the tsetse fly: interference with stage‐specific gene expression and absence of SoMo.
Collapse
Affiliation(s)
- Sebastian Knüsel
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland
| | - Aurelio Jenni
- Institute of Biochemistry and Molecular Medicine, University of Bern, 3012, Bern, Switzerland.,Graduate School for Chemical and Biomedical Sciences, University of Bern, 3012, Bern, Switzerland
| | - Mattias Benninger
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland
| | - Peter Bütikofer
- Institute of Biochemistry and Molecular Medicine, University of Bern, 3012, Bern, Switzerland
| | - Isabel Roditi
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland
| |
Collapse
|
13
|
Loch L, Onofre TS, Rodrigues JPF, Yoshida N. Shedding of Trypanosoma cruzi Surface Molecules That Regulate Host Cell Invasion Involves Phospholipase C and Increases Upon Sterol Depletion. Front Cell Infect Microbiol 2021; 11:769722. [PMID: 34737979 PMCID: PMC8560688 DOI: 10.3389/fcimb.2021.769722] [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: 09/02/2021] [Accepted: 09/27/2021] [Indexed: 12/05/2022] Open
Abstract
Metacyclic trypomastigote (MT) forms of Trypanosoma cruzi have been shown to release into medium gp82 and gp90, the stage-specific surface molecules that regulate host cell invasion, either in vesicles or in soluble form. Here, we found that during interaction of poorly invasive G strain with the host cell, gp82 and gp90 were released in vesicle-like forms, whereas no such release by highly invasive CL strain was observed. Shedding of vesicles of varying sizes by CL and G strains was visualized by scanning electron microscopy, and the protein profile of conditioned medium (CM) of the two strains was similar, but the content of gp82 and gp90 differed, with both molecules being detected in G strain as bands of high intensity in Western blotting, whereas in CL strain, they were barely detectable. Confocal images revealed a distinct distribution of gp82 and gp90 on MT surface of CL and G strains. In cell invasion assays, addition of G strain CM resulted in decreased CL strain internalization. Depletion of gp82 in G strain CM, by treatment with specific mAb-coupled magnetic beads, increased its inhibitory effect on CL strain invasion, in contrast to CM depleted in gp90. The effect of cholesterol-depleting drug methyl-β-cyclodextrin (MβCD) on gp82 and gp90 release by MTs was also examined. G strain MTs, untreated or treated with MβCD, were incubated in serum-containing medium or in nutrient-depleted PBS++, and the CM generated under these conditions was analyzed by Western blotting. In PBS++, gp82 and gp90 were released at lower levels by untreated MTs, as compared with MβCD-treated parasites. CM from untreated and MβCD-treated G strain, generated in PBS++, inhibited CL strain internalization. Treatment of CL strain MTs with MβCD resulted in increased gp82 and gp90 shedding and in decreased host cell invasion. The involvement of phospholipase C (PLC) on gp82 and gp90 shedding was also investigated. The CM from G strain MTs pretreated with specific PLC inhibitor contained lower levels of gp82 and gp90, as compared with untreated parasites. Our results contribute to shed light on the mechanism by which T. cruzi releases surface molecules implicated in host cell invasion.
Collapse
Affiliation(s)
- Leonardo Loch
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Thiago Souza Onofre
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - João Paulo Ferreira Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Rodríguez-Bejarano OH, Avendaño C, Patarroyo MA. Mechanisms Associated with Trypanosoma cruzi Host Target Cell Adhesion, Recognition and Internalization. Life (Basel) 2021; 11:534. [PMID: 34207491 PMCID: PMC8227291 DOI: 10.3390/life11060534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Chagas disease is caused by the kinetoplastid parasite Trypanosoma cruzi, which is mainly transmitted by hematophagous insect bites. The parasite's lifecycle has an obligate intracellular phase (amastigotes), while metacyclic and bloodstream-trypomastigotes are its infective forms. Mammalian host cell recognition of the parasite involves the interaction of numerous parasite and host cell plasma membrane molecules and domains (known as lipid rafts), thereby ensuring internalization by activating endocytosis mechanisms triggered by various signaling cascades in both host cells and the parasite. This increases cytoplasmatic Ca2+ and cAMP levels; cytoskeleton remodeling and endosome and lysosome intracellular system association are triggered, leading to parasitophorous vacuole formation. Its membrane becomes modified by containing the parasite's infectious form within it. Once it has become internalized, the parasite seeks parasitophorous vacuole lysis for continuing its intracellular lifecycle, fragmenting such a vacuole's membrane. This review covers the cellular and molecular mechanisms involved in T. cruzi adhesion to, recognition of and internalization in host target cells.
Collapse
Affiliation(s)
- Oscar Hernán Rodríguez-Bejarano
- Health Sciences Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222#55-37, Bogotá 111166, Colombia;
| | - Catalina Avendaño
- Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222#55-37, Bogotá 111166, Colombia;
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia
- Health Sciences Division, Main Campus, Universidad Santo Tomás, Carrera 9#51-11, Bogotá 110231, Colombia
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia
| |
Collapse
|
15
|
Jin T, Guan N, Du Y, Zhang X, Li J, Xia X. Cronobacter sakazakii ATCC 29544 Translocated Human Brain Microvascular Endothelial Cells via Endocytosis, Apoptosis Induction, and Disruption of Tight Junction. Front Microbiol 2021; 12:675020. [PMID: 34163451 PMCID: PMC8215149 DOI: 10.3389/fmicb.2021.675020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/23/2021] [Indexed: 01/19/2023] Open
Abstract
Cronobacter sakazakii (C. sakazakii) is an emerging opportunistic foodborne pathogen that can cause neonatal necrotizing enterocolitis, meningitis, sepsis in neonates and infants with a relatively high mortality rate. Bacterial transcytosis across the human brain microvascular endothelial cells (HBMEC) is vital for C. sakazakii to induce neonatal meningitis. However, few studies focus on the mechanisms by which C. sakazakii translocates HBMEC. In this study, the translocation processes of C. sakazakii on HBMEC were explored. C. sakazakii strains could effectively adhere to, invade and intracellularly survive in HBMEC. The strain ATCC 29544 exhibited the highest translocation efficiency across HBMEC monolayer among four tested strains. Bacteria-contained intracellular endosomes were detected in C. sakazakii-infected HBMEC by a transmission electron microscope. Endocytosis-related proteins CD44, Rab5, Rab7, and LAMP2 were increased after infection, while the level of Cathepsin L did not change. C. sakazakii induced TLR4/NF-κB inflammatory signal pathway activation in HBMEC, with increased NO production and elevated mRNA levels of IL-8, IL-6, TNF-α, IL-1β, iNOS, and COX-2. C. sakazakii infection also caused LDH release, caspase-3 activation, and HBMEC apoptosis. Meanwhile, increased Dextran-FITC permeability and decreased trans epithelial electric resistance indicated that C. sakazakii disrupted tight junction of HBMEC monolayers, which was confirmed by the decreased levels of tight junction-related proteins ZO-1 and Occludin. These findings suggest that C. sakazakii induced intracellular bacterial endocytosis, stimulated inflammation and apoptosis, disrupted monolayer tight junction in HBMEC, which all together contribute to bacterial translocation.
Collapse
Affiliation(s)
- Tong Jin
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
| | - Ning Guan
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
| | - Yuhang Du
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
| | - Xinpeng Zhang
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
| | - Jiahui Li
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
| | - Xiaodong Xia
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
- National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
16
|
LAMP2 deficiency attenuates the neurodegeneration markers induced by HSV-1 infection. Neurochem Int 2021; 146:105032. [PMID: 33781848 DOI: 10.1016/j.neuint.2021.105032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 11/22/2022]
Abstract
Mounting evidence suggests a major role of infectious agents in the pathogenesis of sporadic Alzheimer's disease (AD). Among them, herpes simplex virus type 1 (HSV-1) infection has emerged as a major factor in the etiology of AD. HSV-1 is able to induce some of the main alterations of the disease such as hyperphosphorylation of tau protein and accumulation of amyloid-β peptide. Functional genomic analysis of a cell model of HSV-1 infection and oxidative stress developed in our laboratory revealed lysosomal system to be the main pathway altered, and the lysosome-associated membrane protein 2 (LAMP2) gene one of the most strongly modulated genes. The aim of this work is to study LAMP2 as an AD candidate gene and to investigate its role in the neurodegeneration induced by HSV-1 using a LAMP2 knockdown cell model. LAMP2 deficiency led to a significant reduction of viral DNA replication and formation of infectious particles. In addition, tau hyperphosphorylation and inhibition of Aβ secretion induced by the virus were attenuated by the absence of LAMP2. Finally, genetic association studies revealed LAMP2 genetic variants to be associated with AD risk. In summary, our data indicate that LAMP2 could be a suitable candidate to mediate the AD-like phenotype caused by HSV-1.
Collapse
|
17
|
Onofre TS, Rodrigues JPF, Shio MT, Macedo S, Juliano MA, Yoshida N. Interaction of Trypanosoma cruzi Gp82 With Host Cell LAMP2 Induces Protein Kinase C Activation and Promotes Invasion. Front Cell Infect Microbiol 2021; 11:627888. [PMID: 33777840 PMCID: PMC7996063 DOI: 10.3389/fcimb.2021.627888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/23/2021] [Indexed: 11/13/2022] Open
Abstract
The surface molecule gp82 of metacyclic trypomastigote (MT) forms of Trypanosoma cruzi, the protozoan parasite that causes Chagas disease, mediates the host cell invasion, a process critical for the establishment of infection. Gp82 is known to bind to the target cell in a receptor-dependent manner, triggering Ca2+ signal, actin cytoskeleton rearrangement and lysosome spreading. The host cell receptor for gp82 was recently identified as LAMP2, the major lysosome membrane-associated protein. To further clarify the mechanisms of MT invasion, we aimed in this study at identifying the LAMP2 domain that interacts with gp82 and investigated whether target cell PKC and ERK1/2, previously suggested to be implicated in MT invasion, are activated by gp82. Interaction of MT, or the recombinant gp82 (r-gp82), with human epithelial HeLa cells induced the activation of Ca2+-dependent PKC and ERK1/2. The LAMP2 sequence predicted to bind gp82 was mapped and the synthetic peptide based on that sequence inhibited MT invasion, impaired the binding of r-gp82 to HeLa cells, and blocked the PKC and ERK1/2 activation induced by r-gp82. Treatment of HeLa cells with specific inhibitor of focal adhesion kinase resulted in inhibition of r-gp82-induced PKC and ERK1/2 activation, as well as in alteration of the actin cytoskeleton architecture. PKC activation by r-gp82 was also impaired by treatment of HeLa cells with inhibitor of phospholipase C, which mediates the production of diacylglycerol, which activates PKC, and inositol 1,4,5-triphosphate that releases Ca2+ from intracellular stores. Taken together, our results indicate that recognition of MT gp82 by LAMP2 induces in the host cell the activation of phosholipase C, with generation of products that contribute for PKC activation and the downstream ERK1/2. This chain of events leads to the actin cytoskeleton disruption and lysosome spreading, promoting MT internalization.
Collapse
Affiliation(s)
- Thiago Souza Onofre
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - João Paulo Ferreira Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marina Tiemi Shio
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Silene Macedo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Aparecida Juliano
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Ferri G, Edreira MM. All Roads Lead to Cytosol: Trypanosoma cruzi Multi-Strategic Approach to Invasion. Front Cell Infect Microbiol 2021; 11:634793. [PMID: 33747982 PMCID: PMC7973469 DOI: 10.3389/fcimb.2021.634793] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022] Open
Abstract
T. cruzi has a complex life cycle involving four developmental stages namely, epimastigotes, metacyclic trypomastigotes, amastigotes and bloodstream trypomastigotes. Although trypomastigotes are the infective forms, extracellular amastigotes have also shown the ability to invade host cells. Both stages can invade a broad spectrum of host tissues, in fact, almost any nucleated cell can be the target of infection. To add complexity, the parasite presents high genetic variability with differential characteristics such as infectivity. In this review, we address the several strategies T. cruzi has developed to subvert the host cell signaling machinery in order to gain access to the host cell cytoplasm. Special attention is made to the numerous parasite/host protein interactions and to the set of signaling cascades activated during the formation of a parasite-containing vesicle, the parasitophorous vacuole, from which the parasite escapes to the cytosol, where differentiation and replication take place.
Collapse
Affiliation(s)
- Gabriel Ferri
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina
| | - Martin M Edreira
- CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina.,Laboratorio de Biología Molecular de Trypanosoma, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina.,Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
19
|
Velásquez-Ortiz N, Ramírez JD. Understanding the oral transmission of Trypanosoma cruzi as a veterinary and medical foodborne zoonosis. Res Vet Sci 2020; 132:448-461. [PMID: 32781335 DOI: 10.1016/j.rvsc.2020.07.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
Chagas disease is a neglected tropical disease transmitted by the protozoan Trypanosoma cruzi that lately has been highlighted because several outbreaks attributed to oral transmission of the parasite have occurred. These outbreaks are characterized by high mortality rates and massive infections that cannot be related to other types of transmission such as the vectorial route. Oral transmission of Chagas disease has been reported in Brazil, Colombia, Venezuela, Bolivia, Ecuador, Argentina and French Guiana, most of them are massive oral outbreaks caused by the ingestion of beverages and food contaminated with triatomine feces or parasites' reservoirs secretions and considered since 2012 as a foodborne disease. In this review, we present the current status and all available data regarding oral transmission of Chagas disease, highlighting its relevance as a veterinary and medical foodborne zoonosis.
Collapse
Affiliation(s)
- Natalia Velásquez-Ortiz
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Departamento de Biología, Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
20
|
Rodríguez ME, Rizzi M, Caeiro LD, Masip YE, Perrone A, Sánchez DO, Búa J, Tekiel V. Transmigration of Trypanosoma cruzi trypomastigotes through 3D cultures resembling a physiological environment. Cell Microbiol 2020; 22:e13207. [PMID: 32270902 DOI: 10.1111/cmi.13207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/05/2020] [Accepted: 03/29/2020] [Indexed: 12/24/2022]
Abstract
To disseminate and colonise tissues in the mammalian host, Trypanosoma cruzi trypomastogotes should cross several biological barriers. How this process occurs or its impact in the outcome of the disease is largely speculative. We examined the in vitro transmigration of trypomastigotes through three-dimensional cultures (spheroids) to understand the tissular dissemination of different T. cruzi strains. Virulent strains were highly invasive: trypomastigotes deeply transmigrate up to 50 μm inside spheroids and were evenly distributed at the spheroid surface. Parasites inside spheroids were systematically observed in the space between cells suggesting a paracellular route of transmigration. On the contrary, poorly virulent strains presented a weak migratory capacity and remained in the external layers of spheroids with a patch-like distribution pattern. The invasiveness-understood as the ability to transmigrate deep into spheroids-was not a transferable feature between strains, neither by soluble or secreted factors nor by co-cultivation of trypomastigotes from invasive and non-invasive strains. Besides, we demonstrated that T. cruzi isolates from children that were born congenitally infected presented a highly migrant phenotype while an isolate from an infected mother (that never transmitted the infection to any of her children) presented significantly less migration. In brief, we demonstrated that in a 3D microenvironment each strain presents a characteristic migration pattern that can be associated to their in vivo behaviour. Altogether, data presented here repositionate spheroids as a valuable tool to study host-pathogen interactions.
Collapse
Affiliation(s)
- Matías Exequiel Rodríguez
- Instituto de Investigaciones Biotecnológicas "Dr. R. Ugalde" (IIBIO) Universidad Nacional de San Martín (UNSAM)-CONICET, Buenos Aires, Argentina
| | - Mariana Rizzi
- Instituto de Investigaciones Biotecnológicas "Dr. R. Ugalde" (IIBIO) Universidad Nacional de San Martín (UNSAM)-CONICET, Buenos Aires, Argentina
| | - Lucas D Caeiro
- Instituto de Investigaciones Biotecnológicas "Dr. R. Ugalde" (IIBIO) Universidad Nacional de San Martín (UNSAM)-CONICET, Buenos Aires, Argentina
| | - Yamil E Masip
- Instituto de Investigaciones Biotecnológicas "Dr. R. Ugalde" (IIBIO) Universidad Nacional de San Martín (UNSAM)-CONICET, Buenos Aires, Argentina
| | - Alina Perrone
- Instituto Nacional de Parasitología "Dr Mario Fatala Chaben", ANLIS-Carlos G. Malbrán, Buenos Aires, Argentina
| | - Daniel O Sánchez
- Instituto de Investigaciones Biotecnológicas "Dr. R. Ugalde" (IIBIO) Universidad Nacional de San Martín (UNSAM)-CONICET, Buenos Aires, Argentina
| | - Jacqueline Búa
- Instituto Nacional de Parasitología "Dr Mario Fatala Chaben", ANLIS-Carlos G. Malbrán, Buenos Aires, Argentina
| | - Valeria Tekiel
- Instituto de Investigaciones Biotecnológicas "Dr. R. Ugalde" (IIBIO) Universidad Nacional de San Martín (UNSAM)-CONICET, Buenos Aires, Argentina
| |
Collapse
|
21
|
Horta MF, Andrade LO, Martins-Duarte ÉS, Castro-Gomes T. Cell invasion by intracellular parasites - the many roads to infection. J Cell Sci 2020; 133:133/4/jcs232488. [PMID: 32079731 DOI: 10.1242/jcs.232488] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracellular parasites from the genera Toxoplasma, Plasmodium, Trypanosoma, Leishmania and from the phylum Microsporidia are, respectively, the causative agents of toxoplasmosis, malaria, Chagas disease, leishmaniasis and microsporidiosis, illnesses that kill millions of people around the globe. Crossing the host cell plasma membrane (PM) is an obstacle these parasites must overcome to establish themselves intracellularly and so cause diseases. The mechanisms of cell invasion are quite diverse and include (1) formation of moving junctions that drive parasites into host cells, as for the protozoans Toxoplasma gondii and Plasmodium spp., (2) subversion of endocytic pathways used by the host cell to repair PM, as for Trypanosoma cruzi and Leishmania, (3) induction of phagocytosis as for Leishmania or (4) endocytosis of parasites induced by specialized structures, such as the polar tubes present in microsporidian species. Understanding the early steps of cell entry is essential for the development of vaccines and drugs for the prevention or treatment of these diseases, and thus enormous research efforts have been made to unveil their underlying biological mechanisms. This Review will focus on these mechanisms and the factors involved, with an emphasis on the recent insights into the cell biology of invasion by these pathogens.
Collapse
Affiliation(s)
- Maria Fátima Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Luciana Oliveira Andrade
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Érica Santos Martins-Duarte
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Thiago Castro-Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| |
Collapse
|
22
|
Campetella O, Buscaglia CA, Mucci J, Leguizamón MS. Parasite-host glycan interactions during Trypanosoma cruzi infection: trans-Sialidase rides the show. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165692. [PMID: 31972227 DOI: 10.1016/j.bbadis.2020.165692] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/07/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
Many important pathogen-host interactions rely on highly specific carbohydrate binding events. In the case of the protozoan Trypanosoma cruzi, the causative agent of Chagas disease, glycointeractions involving sialic acid (SA) residues are pivotal for parasite infectivity, escape from immune surveillance and pathogenesis. Though unable to synthesize SA de novo, T. cruzi displays a unique trans-Sialidase (TS) enzyme, which is able to cleave terminal SA residues from host donor glycoconjugates and transfer them onto parasite surface mucins, thus generating protective/adhesive structures. In addition, this parasite sheds TS into the bloodstream, as a way of modifying the surface SA signature, and thereby the signaling/functional properties of mammalian host target cells on its own advantage. Here, we discuss the pathogenic aspects of T. cruzi TS: its molecular adaptations, the multiplicity of interactions in which it is involved during infections, and the array of novel and appealing targets for intervention in Chagas disease provided by TS-remodeled sialoglycophenotypes.
Collapse
Affiliation(s)
- Oscar Campetella
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Carlos A Buscaglia
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Juan Mucci
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Susana Leguizamón
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
23
|
Onofre TS, Rodrigues JPF, Yoshida N. Depletion of Host Cell Focal Adhesion Kinase Increases the Susceptibility to Invasion by Trypanosoma cruzi Metacyclic Forms. Front Cell Infect Microbiol 2019; 9:231. [PMID: 31297342 PMCID: PMC6607697 DOI: 10.3389/fcimb.2019.00231] [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] [Received: 04/11/2019] [Accepted: 06/11/2019] [Indexed: 11/13/2022] Open
Abstract
Focal adhesion kinase (FAK), a cytoplasmic protein tyrosine kinase (PTK), is implicated in diverse cellular processes, including the regulation of F-actin dynamics. Host cell F-actin rearrangement is critical for invasion of Trypanosoma cruzi, the protozoan parasite that causes Chagas disease. It is unknown whether FAK is involved in the internalization process of metacyclic trypomastigote (MT), the parasite form that is important for vectorial transmission. MT can enter the mammalian host through the ocular mucosa, lesion in the skin, or by the oral route. Oral infection by MT is currently a mode of transmission responsible for outbreaks of acute Chagas disease. Here we addressed the question by generating HeLa cell lines deficient in FAK. Host cell invasion assays showed that, as compared to control wild type (WT) cells, FAK-deficient cells were significantly more susceptible to parasite invasion. Lysosome spreading and a disarranged actin cytoskeleton, two features associated with susceptibility to MT invasion, were detected in FAK-deficient cells, as opposed to WT cells that exhibited a more organized F-actin arrangement, and lysosomes concentrated in the perinuclear area. As compared to WT cells, the capacity of FAK-deficient cells to bind a recombinant protein based on gp82, the MT surface molecule that mediates invasion, was higher. On the other hand, when treated with FAK-specific inhibitor PF573228, WT cells exhibited a dense meshwork of actin filaments, lysosome accumulation around the nucleus, and had increased resistance to MT invasion. In cells treated with PF573228, the phosphorylation levels of FAK were reduced and, as a consequence of FAK inactivation, diminished phosphorylation of extracellular signal-regulated protein kinases (ERK1/2) was observed. Fibronectin, known to impair MT invasion, induced the formation of thick bundles of F-actin and ERK1/2 dephosphorylation.
Collapse
Affiliation(s)
- Thiago Souza Onofre
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - João Paulo Ferreira Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
24
|
Rodrigues JPF, Souza Onofre T, Barbosa BC, Ferreira ÉR, Bonfim‐Melo A, Yoshida N. Host cell protein LAMP-2 is the receptor for Trypanosoma cruzi surface molecule gp82 that mediates invasion. Cell Microbiol 2019; 21:e13003. [PMID: 30609224 PMCID: PMC6590364 DOI: 10.1111/cmi.13003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/22/2018] [Accepted: 12/28/2018] [Indexed: 02/04/2023]
Abstract
Host cell invasion by Trypanosoma cruzi metacyclic trypomastigote (MT) is mediated by MT-specific surface molecule gp82, which binds to a still unidentified receptor, inducing lysosome spreading and exocytosis required for the parasitophorous vacuole formation. We examined the involvement of the major lysosome membrane-associated LAMP proteins in MT invasion. First, human epithelial HeLa cells were incubated with MT in the presence of antibody to LAMP-1 or LAMP-2. Antibody to LAMP-2, but not to LAMP-1, significantly reduced MT invasion. Next, HeLa cells depleted in LAMP-1 or LAMP-2 were generated. Cells deficient in LAMP-2, but not in LAMP-1, were significantly more resistant to MT invasion than wild-type controls. The possibility that LAMP-2 might be the receptor for gp82 was examined by co-immunoprecipitation assays. Protein A/G magnetic beads cross-linked with antibody directed to LAMP-1 or LAMP-2 were incubated with HeLa cell and MT detergent extracts. Gp82 bound to LAMP-2 but not to LAMP-1. Binding of the recombinant gp82 protein to wild-type and LAMP-1-deficient cells, which was dose dependent and saturable, had a similar profile and was much higher as compared with LAMP-2-depleted cells. These data indicate that MT invasion is accomplished through recognition of gp82 by its receptor LAMP-2.
Collapse
Affiliation(s)
- João Paulo Ferreira Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloBrazil
| | - Thiago Souza Onofre
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloBrazil
| | - Bruno Couto Barbosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloBrazil
| | - Éden Ramalho Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloBrazil
| | - Alexis Bonfim‐Melo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloBrazil
| | - Nobuko Yoshida
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de MedicinaUniversidade Federal de São PauloSão PauloBrazil
| |
Collapse
|