1
|
Kim A, Gorman H, Moreau F, McManus M, Dufour A, Chadee K. Human Mucin-2 Mucin-Producing Colonic Goblet-Like Cells Secrete the Chemokine CXCL8 by Activating Multiple Proinflammatory Pathways in Response to Entamoeba histolytica. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00080-X. [PMID: 40122459 DOI: 10.1016/j.ajpath.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
The mucus layer produced by highly stressed goblet cells forms a protective shield in the gut to protect the underlying mucosal epithelial cells from external threats. Hypersecretion and depletion of mucin-2 (MUC2) mucin from goblet cells is characteristic of symptomatic Entamoeba histolytica infections. It was hypothesized that MUC2 depleted goblet cells could mount a second line of innate host defense by producing proinflammatory cytokines. To investigate this, it was determined whether E. histolytica could stimulate proinflammatory responses in wild-type (WT) high MUC2 mucin-producing goblet-like cells and in CRISPR-Cas9 gene-edited MUC2KO cells. In response to live E. histolytica and soluble E. histolytica proteins, WT and, to a lesser extent, MUC2KO cells produced high levels of CXCL8. Entamoeba histolytica temporally induced greater levels of CXCL8 mRNA expression and protein secretion in WT versus MUC2KO cells, which was abrogated with alleviation of endoplasmic reticulum stress with the NADPH-oxidase inhibitor diphenyleneiodonium chloride. WT cells produced elevated reactive oxygen species that induced longer half-lives of CXCL8 transcripts, which was abrogated with diphenyleneiodonium chloride. Western blotting and proteomic analyses revealed that WT cells, but not MUC2KO cells, were basally primed to respond to external stressors and responded to E. histolytica through rapid activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase, mitogen-activated protein kinase/p38, and phosphatidylinositol 3-kinase/Akt pathways, to induce CXCL8. These results suggest that colonic goblet-like cells defend against E. histolytica infections by hypersecreting mucus and to produce the chemokine, CXCL8, to recruit neutrophils.
Collapse
Affiliation(s)
- Ariel Kim
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - Hayley Gorman
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - France Moreau
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - Mackenzie McManus
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - Antoine Dufour
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada.
| |
Collapse
|
2
|
Zhang H, Li Q, Zhou H, Feng M, Zhao Y, Zhou R, Chen L, Tachibana H, Cheng X. Identification and characterization of a carbohydrate recognition domain-like region in Entamoeba histolytica Gal/GalNAc lectin intermediate subunit. Microbiol Spectr 2024; 12:e0053824. [PMID: 39365081 PMCID: PMC11537071 DOI: 10.1128/spectrum.00538-24] [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: 02/29/2024] [Accepted: 09/06/2024] [Indexed: 10/05/2024] Open
Abstract
Entamoeba histolytica is an enteric protozoan parasite that causes human amebic colitis and extraintestinal abscesses. As a prerequisite for parasite colonization and invasion, adherence of E. histolytica is predominantly mediated by galactose (Gal)- and N-acetyl-d-galactosamine (GalNAc)-inhibitable lectins. The intermediate subunit (Igl) of Gal-/GalNAc-inhibitable lectin is a cysteine-rich protein containing multiple CXXC motifs and is considered a key factor affecting trophozoite's pathogenicity. However, details of the function of Igl during parasite adherence remain unclear. Here, using segmentally expressed Igl proteins and a CHO cell model transfected with Igl fragments, we identified a carbohydrate-recognition domain (CRD)-like region between amino acids 989 and 1,088. Through single- and double-point mutations in the Igl segments, two core CXXC motifs responsible for carbohydrate recognition in the CRD-like region, which are highly conserved among several lectins, were confirmed. In addition to adhesion, the roles of CRD-like region and its core CXXC motifs in various pathogenic effects were further explored. To our knowledge, this is the first report showing an adhesion-related region in E. histolytica Igl. The identification and characterization of this CRD-like region provides further insights into molecular mechanisms underlying E. histolytica pathogenicity and also aids in the determination of a potential drug target in this parasite. IMPORTANCE Entamoeba histolytica adhesion mainly depends on galactose (Gal)-/N-acetyl-d-galactosamine (GalNAc)-inhibitable lectins, subsequently triggering a series of amebic reactions. Among the three subunits of Gal-/GalNAc-inhibitable lectin, heavy subunit and intermediate subunit (Igl) have exhibited lectin activity, but that of Igl remains poorly understood. In this study, we confirmed a carbohydrate-recognition domain (CRD)-like limiting region in E. histolytica Igl and further identified its two core CXXC motifs responsible for carbohydrate recognition. Moreover, the role of Igl's CRD-like region and its CXXC motifs in hemolysis and pathogenic effects was explored. This is the first study to determine an adhesion-related region in E. histolytica Igl protein, providing a new reference direction for subsequent research studies. Since the potential homogeneity of galectin-2 in several mammals and Igl CRD-like region, it could be meaningful to relate the corresponding pathogeneses and phenotypes of these two proteins. Except for adhesion, studies on the involvement of Igl CRD-like region in different parasite-host interactions are also promising.
Collapse
Affiliation(s)
- Hongze Zhang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qingshan Li
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hang Zhou
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meng Feng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yanqing Zhao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ruixue Zhou
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lijun Chen
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hiroshi Tachibana
- Department of Infectious Diseases, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Xunjia Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Partida-Rodríguez O, Brown EM, Woodward SE, Cirstea M, Reynolds LA, Petersen C, Vogt SL, Peña-Díaz J, Thorson L, Arrieta MC, Hernández EG, Rojas-Velázquez L, Moran P, González Rivas E, Serrano-Vázquez A, Pérez-Juárez H, Torres J, Ximénez C, Finlay BB. Fecal microbiota transplantation from protozoa-exposed donors downregulates immune response in a germ-free mouse model, its role in immune response and physiology of the intestine. PLoS One 2024; 19:e0312775. [PMID: 39466773 PMCID: PMC11515975 DOI: 10.1371/journal.pone.0312775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
Intestinal parasites are part of the intestinal ecosystem and have been shown to establish close interactions with the intestinal microbiota. However, little is known about the influence of intestinal protozoa on the regulation of the immune response. In this study, we analyzed the regulation of the immune response of germ-free mice transplanted with fecal microbiota (FMT) from individuals with multiple parasitic protozoans (P) and non-parasitized individuals (NP). We determined the production of intestinal cytokines, the lymphocyte populations in both the colon and the spleen, and the genetic expression of markers of intestinal epithelial integrity. We observed a general downregulation of the intestinal immune response in mice receiving FMT-P. We found significantly lower intestinal production of the cytokines IL-6, TNF, IFN-γ, MCP-1, IL-10, and IL-12 in the FMT-P. Furthermore, a significant decrease in the proportion of CD3+, CD4+, and Foxp3+ T regulatory cells (Treg) was observed in both, the colon and spleen with FMT-P in contrast to FMT-NP. We also found that in FMT-P mice there was a significant decrease in tjp1 expression in all three regions of the small intestine; ocln in the ileum; reg3γ in the duodenum and relmβ in both the duodenum and ileum. We also found an increase in colonic mucus layer thickness in mice colonized with FMT-P in contrast with FMT-NP. Finally, our results suggest that gut protozoa, such as Blastocystis hominis, Entamoeba coli, Endolimax nana, Entamoeba histolytica/E. dispar, Iodamoeba bütschlii, and Chilomastix mesnili consortia affect the immunoinflammatory state and induce functional changes in the intestine via the gut microbiota. Likewise, it allows us to establish an FMT model in germ-free mice as a viable alternative to explore the effects that exposure to intestinal parasites could have on the immune response in humans.
Collapse
Affiliation(s)
- Oswaldo Partida-Rodríguez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Eric M. Brown
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Sarah E. Woodward
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Mihai Cirstea
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Lisa A. Reynolds
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Microbiology, Faculty of Science, University of Victoria, Victoria, Canada
| | - Charisse Petersen
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Stefanie L. Vogt
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Jorge Peña-Díaz
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Lisa Thorson
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Marie-Claire Arrieta
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Eric G. Hernández
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Liliana Rojas-Velázquez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Patricia Moran
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Enrique González Rivas
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Angélica Serrano-Vázquez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Horacio Pérez-Juárez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas y Parasitarias, Instituto Mexicano del Seguro Social (IMSS), Mexico, Mexico
| | - Cecilia Ximénez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - B. B. Finlay
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
4
|
Otálora-Otálora BA, Payán-Gómez C, López-Rivera JJ, Pedroza-Aconcha NB, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Global transcriptomic network analysis of the crosstalk between microbiota and cancer-related cells in the oral-gut-lung axis. Front Cell Infect Microbiol 2024; 14:1425388. [PMID: 39228892 PMCID: PMC11368877 DOI: 10.3389/fcimb.2024.1425388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 09/05/2024] Open
Abstract
Background The diagnosis and treatment of lung, colon, and gastric cancer through the histologic characteristics and genomic biomarkers have not had a strong impact on the mortality rates of the top three global causes of death by cancer. Methods Twenty-five transcriptomic analyses (10 lung cancer, 10 gastric cancer, and 5 colon cancer datasets) followed our own bioinformatic pipeline based on the utilization of specialized libraries from the R language and DAVID´s gene enrichment analyses to identify a regulatory metafirm network of transcription factors and target genes common in every type of cancer, with experimental evidence that supports its relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during the tumoral process. The network's regulatory functional and signaling pathways might depend on the constant crosstalk with the microbiome network established in the oral-gut-lung axis. Results The global transcriptomic network analysis highlighted the impact of transcription factors (SOX4, TCF3, TEAD4, ETV4, and FOXM1) that might be related to stem cell programming and cancer progression through the regulation of the expression of genes, such as cancer-cell membrane receptors, that interact with several microorganisms, including human T-cell leukemia virus 1 (HTLV-1), the human papilloma virus (HPV), the Epstein-Barr virus (EBV), and SARS-CoV-2. These interactions can trigger the MAPK, non-canonical WNT, and IFN signaling pathways, which regulate key transcription factor overexpression during the establishment and progression of lung, colon, and gastric cancer, respectively, along with the formation of the microbiome network. Conclusion The global transcriptomic network analysis highlights the important interaction between key transcription factors in lung, colon, and gastric cancer, which regulates the expression of cancer-cell membrane receptors for the interaction with the microbiome network during the tumorigenic process.
Collapse
Affiliation(s)
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz, Colombia
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá, Colombia
| | | | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá, Colombia
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá, Colombia
| | | |
Collapse
|
5
|
Zanatta D, Betanzos A, Azuara-Liceaga E, Montaño S, Orozco E. Entamoeba histolytica: EhADH, an Alix Protein, Participates in Several Virulence Events through Its Different Domains. Int J Mol Sci 2024; 25:7609. [PMID: 39062867 PMCID: PMC11277477 DOI: 10.3390/ijms25147609] [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: 06/03/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Entamoeba histolytica is the protozoan causative of human amoebiasis. The EhADH adhesin (687 aa) is a protein involved in tissue invasion, phagocytosis and host-cell lysis. EhADH adheres to the prey and follows its arrival to the multivesicular bodies. It is an accessory protein of the endosomal sorting complexes required for transport (ESCRT) machinery. Here, to study the role of different parts of EhADH during virulence events, we produced trophozoites overexpressing the three domains of EhADH, Bro1 (1-400 aa), Linker (246-446 aa) and Adh (444-687 aa) to evaluate their role in virulence. The TrophozBro11-400 slightly increased adherence and phagocytosis, but these trophozoites showed a higher ability to destroy cell monolayers, augment the permeability of cultured epithelial cells and mouse colon, and produce more damage to hamster livers. The TrophozLinker226-446 also increased the virulence properties, but with lower effect than the TrophozBro11-400. In addition, this fragment participates in cholesterol transport and GTPase binding. Interestingly, the TrophozAdh444-687 produced the highest effect on adherence and phagocytosis, but it poorly influenced the monolayers destruction; nevertheless, they augmented the colon and liver damage. To identify the protein partners of each domain, we used recombinant peptides. Pull-down assays and mass spectrometry showed that Bro1 domain interplays with EhADH, Gal/GalNAc lectin, EhCPs, ESCRT machinery components and cytoskeleton proteins. While EhADH, ubiquitin, EhRabB, EhNPC1 and EhHSP70 were associated to the Linker domain, and EhADH, EhHSP70, EhPrx and metabolic enzymes interacted to the Adh domain. The diverse protein association confirms that EhADH is a versatile molecule with multiple functions probably given by its capacity to form distinct molecular complexes.
Collapse
Affiliation(s)
- Dxinegueela Zanatta
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of National Polytechnic Institute, Mexico City 07360, Mexico;
| | - Abigail Betanzos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of National Polytechnic Institute, Mexico City 07360, Mexico;
| | - Elisa Azuara-Liceaga
- Postgraduate in Genomic Sciences, Autonomous University of Mexico City, Mexico City 03100, Mexico;
| | - Sarita Montaño
- Laboratory of Bioinformatics and Molecular Simulation, Faculty of Biological Chemistry Sciences, Autonomous University of Sinaloa, Sinaloa 80030, Mexico;
| | - Esther Orozco
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of National Polytechnic Institute, Mexico City 07360, Mexico;
| |
Collapse
|
6
|
Guillén N. Pathogenicity and virulence of Entamoeba histolytica, the agent of amoebiasis. Virulence 2023; 14:2158656. [PMID: 36519347 DOI: 10.1080/21505594.2022.2158656] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
The amoeba parasite Entamoeba histolytica is the causative agent of human amebiasis, an enteropathic disease affecting millions of people worldwide. This ancient protozoan is an elementary example of how parasites evolve with humans, e.g. taking advantage of multiple mechanisms to evade immune responses, interacting with microbiota for nutritional and protective needs, utilizing host resources for growth, division, and encystation. These skills of E. histolytica perpetuate the species and incidence of infection. However, in 10% of infected cases, the parasite turns into a pathogen; the host-parasite equilibrium is then disorganized, and the simple lifecycle based on two cell forms, trophozoites and cysts, becomes unbalanced. Trophozoites acquire a virulent phenotype which, when non-controlled, leads to intestinal invasion with the onset of amoebiasis symptoms. Virulent E. histolytica must cross mucus, epithelium, connective tissue and possibly blood. This highly mobile parasite faces various stresses and a powerful host immune response, with oxidative stress being a challenge for its survival. New emerging research avenues and omics technologies target gene regulation to determine human or parasitic factors activated upon infection, their role in virulence activation, and in pathogenesis; this research bears in mind that E. histolytica is a resident of the complex intestinal ecosystem. The goal is to eradicate amoebiasis from the planet, but the parasitic life of E. histolytica is ancient and complex and will likely continue to evolve with humans. Advances in these topics are summarized here.
Collapse
Affiliation(s)
- Nancy Guillén
- Cell Biology and Infection Department, Institut Pasteur and Centre National de la Recherche Scientifique CNRS-ERM9195, Paris, France
| |
Collapse
|
7
|
Argüello-García R, Carrero JC, Ortega-Pierres MG. Extracellular Cysteine Proteases of Key Intestinal Protozoan Pathogens-Factors Linked to Virulence and Pathogenicity. Int J Mol Sci 2023; 24:12850. [PMID: 37629029 PMCID: PMC10454693 DOI: 10.3390/ijms241612850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Intestinal diseases caused by protistan parasites of the genera Giardia (giardiasis), Entamoeba (amoebiasis), Cryptosporidium (cryptosporidiosis) and Blastocystis (blastocystosis) represent a major burden in human and animal populations worldwide due to the severity of diarrhea and/or inflammation in susceptible hosts. These pathogens interact with epithelial cells, promoting increased paracellular permeability and enterocyte cell death (mainly apoptosis), which precede physiological and immunological disorders. Some cell-surface-anchored and molecules secreted from these parasites function as virulence markers, of which peptide hydrolases, particularly cysteine proteases (CPs), are abundant and have versatile lytic activities. Upon secretion, CPs can affect host tissues and immune responses beyond the site of parasite colonization, thereby increasing the pathogens' virulence. The four intestinal protists considered here are known to secrete predominantly clan A (C1- and C2-type) CPs, some of which have been characterized. CPs of Giardia duodenalis (e.g., Giardipain-1) and Entamoeba histolytica (EhCPs 1-6 and EhCP112) degrade mucin and villin, cause damage to intercellular junction proteins, induce apoptosis in epithelial cells and degrade immunoglobulins, cytokines and defensins. In Cryptosporidium, five Cryptopains are encoded in its genome, but only Cryptopains 4 and 5 are likely secreted. In Blastocystis sp., a legumain-activated CP, called Blastopain-1, and legumain itself have been detected in the extracellular medium, and the former has similar adverse effects on epithelial integrity and enterocyte survival. Due to their different functions, these enzymes could represent novel drug targets. Indeed, some promising results with CP inhibitors, such as vinyl sulfones (K11777 and WRR605), the garlic derivative, allicin, and purified amoebic CPs have been obtained in experimental models, suggesting that these enzymes might be useful drug targets.
Collapse
Affiliation(s)
- Raúl Argüello-García
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City 07360, Mexico;
| | - Julio César Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), México City 04510, Mexico
| | - M. Guadalupe Ortega-Pierres
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City 07360, Mexico;
| |
Collapse
|
8
|
Gorman H, Moreau F, Dufour A, Chadee K. IgGFc-binding protein and MUC2 mucin produced by colonic goblet-like cells spatially interact non-covalently and regulate wound healing. Front Immunol 2023; 14:1211336. [PMID: 37359538 PMCID: PMC10285406 DOI: 10.3389/fimmu.2023.1211336] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
The colonic mucus bilayer is the first line of innate host defense that at the same time houses and nourishes the commensal microbiota. The major components of mucus secreted by goblet cells are MUC2 mucin and the mucus-associated protein, FCGBP (IgGFc-binding protein). In this study, we determine if FCGBP and MUC2 mucin were biosynthesized and interacted together to spatially enhance the structural integrity of secreted mucus and its role in epithelial barrier function. MUC2 and FCGBP were coordinately regulated temporally in goblet-like cells and in response to a mucus secretagogue but not in CRISPR-Cas9 gene-edited MUC2 KO cells. Whereas ~85% of MUC2 was colocalized with FCGBP in mucin granules, ~50% of FCGBP was diffusely distributed in the cytoplasm of goblet-like cells. STRING-db v11 analysis of the mucin granule proteome revealed no protein-protein interaction between MUC2 and FCGBP. However, FCGBP interacted with other mucus-associated proteins. FCGBP and MUC2 interacted via N-linked glycans and were non-covalently bound in secreted mucus with cleaved low molecular weight FCGBP fragments. In MUC2 KO, cytoplasmic FCGBP was significantly increased and diffusely distributed in wounded cells that healed by enhanced proliferation and migration within 2 days, whereas, in WT cells, MUC2 and FCGBP were highly polarized at the wound margin which impeded wound closure by 6 days. In DSS colitis, restitution and healed lesions in Muc2+/+ but not Muc2-/- littermates, were accompanied by a rapid increase in Fcgbp mRNA and delayed protein expression at 12- and 15-days post DSS, implicating a potential novel endogenous protective role for FCGBP in wound healing to maintain epithelial barrier function.
Collapse
Affiliation(s)
- Hayley Gorman
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - France Moreau
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Kris Chadee
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
9
|
Liu Y, Yu Z, Zhu L, Ma S, Luo Y, Liang H, Liu Q, Chen J, Guli S, Chen X. Orchestration of MUC2 - The key regulatory target of gut barrier and homeostasis: A review. Int J Biol Macromol 2023; 236:123862. [PMID: 36870625 DOI: 10.1016/j.ijbiomac.2023.123862] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
The gut mucosa of human is covered by mucus, functioning as a crucial defense line for the intestine against external stimuli and pathogens. Mucin2 (MUC2) is a subtype of secretory mucins generated by goblet cells and is the major macromolecular component of mucus. Currently, there is an increasing interest on the investigations of MUC2, noting that its function is far beyond a maintainer of the mucus barrier. Moreover, numerous gut diseases are associated with dysregulated MUC2 production. Appropriate production level of MUC2 and mucus contributes to gut barrier function and homeostasis. The production of MUC2 is regulated by a series of physiological processes, which are orchestrated by various bioactive molecules, signaling pathways and gut microbiota, etc., forming a complex regulatory network. Incorporating the latest findings, this review provided a comprehensive summary of MUC2, including its structure, significance and secretory process. Furthermore, we also summarized the molecular mechanisms of the regulation of MUC2 production aiming to provide developmental directions for future researches on MUC2, which can act as a potential prognostic indicator and targeted therapeutic manipulation for diseases. Collectively, we elucidated the micro-level mechanisms underlying MUC2-related phenotypes, hoping to offer some constructive guidance for intestinal and overall health of mankind.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Lanping Zhu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Shuang Ma
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Yang Luo
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Huixi Liang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Qinlingfei Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Jihua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Sitan Guli
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China.
| |
Collapse
|
10
|
Jasni N, Saidin S, Kin WW, Arifin N, Othman N. Entamoeba histolytica: Membrane and Non-Membrane Protein Structure, Function, Immune Response Interaction, and Vaccine Development. MEMBRANES 2022; 12:1079. [PMID: 36363634 PMCID: PMC9695907 DOI: 10.3390/membranes12111079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Entamoeba histolytica is a protozoan parasite that is the causative agent of amoebiasis. This parasite has caused widespread infection in India, Africa, Mexico, and Central and South America, and results in 100,000 deaths yearly. An immune response is a body's mechanism for eradicating and fighting against substances it sees as harmful or foreign. E. histolytica biological membranes are considered foreign and immunogenic to the human body, thereby initiating the body's immune responses. Understanding immune response and antigen interaction are essential for vaccine development. Thus, this review aims to identify and understand the protein structure, function, and interaction of the biological membrane with the immune response, which could contribute to vaccine development. Furthermore, the current trend of vaccine development studies to combat amoebiasis is also reviewed.
Collapse
Affiliation(s)
- Nurhana Jasni
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia
| | - Syazwan Saidin
- Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, Tanjung Malim 35900, Malaysia
| | - Wong Weng Kin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Norsyahida Arifin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia
| | - Nurulhasanah Othman
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia
| |
Collapse
|
11
|
Wang S, Moreau F, Chadee K. Gasdermins in Innate Host Defense Against Entamoeba histolytica and Other Protozoan Parasites. Front Immunol 2022; 13:900553. [PMID: 35795683 PMCID: PMC9251357 DOI: 10.3389/fimmu.2022.900553] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Gasdermins (GSDMs) are a group of proteins that are cleaved by inflammatory caspases to induce pore formation in the plasma membrane to cause membrane permeabilization and lytic cell death or pyroptosis. All GSDMs share a conserved structure, containing a cytotoxic N-terminal (NT) pore-forming domain and a C-terminal (CT) repressor domain. Entamoeba histolytica (Eh) in contact with macrophages, triggers outside-in signaling to activate inflammatory caspase-4/1 via the noncanonical and canonical pathway to promote cleavage of gasdermin D (GSDMD). Cleavage of GSDMD removes the auto-inhibition that masks the active pore-forming NT domain in the full-length protein by interactions with GSDM-CT. The cleaved NT-GSDMD monomers then oligomerize to form pores in the plasma membrane to facilitate the release of IL-1β and IL-18 with a measured amount of pyroptosis. Pyroptosis is an effective way to counteract intracellular parasites, which exploit replicative niche to avoid killing. To date, most GSDMs have been verified to perform pore-forming activity and GSDMD-induced pyroptosis is rapidly emerging as a mechanism of anti-microbial host defence. Here, we review our comprehensive and current knowledge on the expression, activation, biological functions, and regulation of GSDMD cleavage with emphases on physiological scenario and related dysfunctions of each GSDM member as executioner of cell death, cytokine secretion and inflammation against Eh and other protozoan parasitic infections.
Collapse
Affiliation(s)
| | | | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
12
|
Quantitative Proteomics Reveals Metabolic Reprogramming in Host Cells Induced by Trophozoites and Intermediate Subunit of Gal/GalNAc Lectins from Entamoeba histolytica. mSystems 2022; 7:e0135321. [PMID: 35343800 PMCID: PMC9040881 DOI: 10.1128/msystems.01353-21] [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] [Indexed: 11/21/2022] Open
Abstract
Entamoeba histolytica is an intestinal protozoan parasite with remarkable ability to kill and phagocytose host cells, causing amoebic colitis and extraintestinal abscesses. The intermediate subunit (Igl) of galactose (Gal)- and N-acetyl-d-galactosamine (GalNAc)-specific lectins is considered an important surface antigen involved in the pathogenesis of E. histolytica. Here, we applied mass spectrometry-based quantitative proteomics technology to analyze the protein expression profile changes occurring in host Caco2 cells incubated with E. histolytica trophozoites or stimulated by purified native Igl protein. The expression levels of 1,490 and 489 proteins were significantly altered in the E. histolytica-treated and Igl-treated groups, respectively, among 6,875 proteins totally identified. Intriguingly, central carbon metabolism of host cells was suppressed in both E. histolytica-treated and Igl-treated groups, with evidence of decreased expression levels of several key enzymes, including pyruvate kinase muscle type 2, presenting a Warburg-like effect in host cells. Besides, Igl had potential physical interactions with central carbon metabolism enzymes and the proteolytic degradation family members proteasome subunit alpha and beta, which may be responsible for the degradation of key enzymes in carbon metabolism. These results provided a novel perspective on the pathogenic mechanism of E. histolytica and compelling evidence supporting the important role of Igl in the virulence of E. histolytica. IMPORTANCE Metabolic reprogramming is considered a hallmark of some infectious diseases. However, in amoebiasis, a neglected tropical disease caused by protozoan parasite E. histolytica, metabolic changes in host cells have yet to be proven. In this study, advanced data-independent acquisition mass spectrometry-based quantitative proteomics was applied to investigate the overall host cellular metabolic changes as high-throughput proteomics could measure molecular changes in a cell or tissue with high efficiency. Enrichment analysis of differentially expressed proteins showed biological processes and cellular pathways related to amoeba infection and Igl cytotoxicity. Specifically, central carbon metabolism of host cells was dramatically suppressed in both E. histolytica-treated and Igl-treated groups, indicating the occurrence of a Warburg-like effect induced by trophozoites or Igl from E. histolytica. Distinct differences in ubiquitin-mediated proteolysis, rapamycin (mTOR) signaling pathway, autophagy, endocytosis, and tight junctions provided novel perspectives on the pathogenic mechanism of E. histolytica.
Collapse
|
13
|
Al-Zubadi WFH, Al-Masoudi HK, Abdul-Lateef LA. Detection and Sequencing of Iron Superoxide Dismutase Gene in Entamoeba histolytica Isolated from Patients with Diarrhea in Iraq. ARCHIVES OF RAZI INSTITUTE 2021; 76:1289-1295. [PMID: 35355778 PMCID: PMC8934092 DOI: 10.22092/ari.2021.356101.1777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/02/2021] [Indexed: 06/14/2023]
Abstract
Parasites are among serious health problems that threaten the public health of humans. Entamoeba histolytica is considered the second or third most common cause of death among parasitic diseases. The present study was conducted to determine the molecular detection and sequencing of iron-containing superoxide dismutase (FeSOD) genes of E. histolytica. This cross-sectional study was carried out within March-August 2020. Genomic DNA from stool samples was extracted using an AccuPrep® stool DNA Extraction Kit. Afterward, four DNA samples were subjected to sequence alignment analysis of the FeSOD gene in local E. histolytica human isolated using a Clustal W alignment tool (Mega 6.0).The iron-containing superoxide dismutase gene was present at 62.8% in 15 out of 24 E. histolytica samples. The results of sequence alignment analysis of the FeSOD gene in local E.histolytica human isolates indicated the number of mutations in each of the 4 isolates. Accordingly, there were 2 (16.66%) silent mutations and 1(6.66%) nonsense mutation. There are a variety of genetic strains of E. histolytica associated with diarrheal illness in Iraqi patients, which are unique to this country.
Collapse
Affiliation(s)
- W F H Al-Zubadi
- Department of Microbiology, College of Medicine, University of Babylon, Babylon, Iraq
| | - H K Al-Masoudi
- Department of Microbiology, College of Medicine, University of Babylon, Babylon, Iraq
| | - L A Abdul-Lateef
- Department of Microbiology, College of Medicine, University of Babylon, Babylon, Iraq
| |
Collapse
|
14
|
Chadha A, Moreau F, Wang S, Dufour A, Chadee K. Entamoeba histolytica activation of caspase-1 degrades cullin that attenuates NF-κB dependent signaling from macrophages. PLoS Pathog 2021; 17:e1009936. [PMID: 34499701 PMCID: PMC8454965 DOI: 10.1371/journal.ppat.1009936] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/21/2021] [Accepted: 09/01/2021] [Indexed: 12/18/2022] Open
Abstract
While Entamoeba histolytica (Eh)-induced pro-inflammatory responses are critical in disease pathogenesis, the downstream signaling pathways that subsequently dampens inflammation and the immune response remains unclear. Eh in contact with macrophages suppresses NF-κB signaling while favoring NLRP3-dependent pro-inflammatory cytokine production by an unknown mechanism. Cullin-1 and cullin-5 (cullin-1/5) assembled into a multi-subunit RING E3 ubiquitin ligase complex are substrates for neddylation that regulates the ubiquitination pathway important in NF-κB activity and pro-inflammatory cytokine production. In this study, we showed that upon live Eh contact with human macrophages, cullin-1/4A/4B/5 but not cullin-2/3, were degraded within 10 minutes. Similar degradation of cullin-1/5 were observed from colonic epithelial cells and proximal colonic loops tissues of mice inoculated with live Eh. Degradation of cullin-1/5 was dependent on Eh-induced activation of caspase-1 via the NLRP3 inflammasome. Unlike cullin-4B, the degradation of cullin-4A was partially dependent on caspase-1 and was inhibited with a pan caspase inhibitor. Cullin-1/5 degradation was dependent on Eh cysteine proteinases EhCP-A1 and EhCP-A4, but not EhCP-A5, based on pharmacological inhibition of the cysteine proteinases and EhCP-A5 deficient parasites. siRNA silencing of cullin-1/5 decreased the phosphorylation of pIκ-Bα in response to Eh and LPS stimulation and downregulated NF-κB-dependent TNF-α mRNA expression and TNF-α and MCP-1 pro-inflammatory cytokine production. These results unravel a unique outside-in strategy employed by Eh to attenuate NF-κB-dependent pro-inflammatory responses via NLRP3 activation of caspase-1 that degraded cullin-1/5 from macrophages. The protozoan parasite Entamoeba histolytica (Eh) is the etiologic agent for the disease amebiasis. It is a potent pathogen that deploys an arsenal of virulence factors to trigger and subvert host immune defenses. One of the hallmark features of the disease is amebic colitis and in extreme cases, it can lead to abscesses of the liver and brain. For unknown reasons, the parasite breaches colonic mucosal barriers and invade underlying tissues. The host immune system plays a decisive role in determining the outcome of the disease. At the molecular level, the interaction of Eh with macrophage is a turning point in shaping pro-inflammatory responses. Understanding host-pathogen intricacies at the molecular level is key in determining the complexity of the disease. In the context of amebiasis, the underlying molecular events that occur at the Eh-macrophage intercellular junction are partly unravelled. Here we sought to interrogate the mechanisms by which NF-κB signaling is aborted following Eh-macrophage contact and found two regulatory scaffold proteins, cullin-1 and -5 (cullin-1/5) of the multiple E3 ligase complex, are degraded leading to dampening of NF-κB signaling. During Eh-macrophage contact, cullin-1/4A/4B/5 were rapidly degraded whereas cullin-2/3 were not. The degradation of cullin-1/5 was highly dependent on Eh-induced caspase-1 activation via the NLRP3 inflammasome. In contrast, the degradation of cullin-4A but not cullin-4B, was partially dependent on caspase-1 and was inhibited with a cell-permeable pan caspase inhibitor. Intriguingly, we found that Eh virulence factor EhCP-A1 and EhCP-A4, but not EhCP-A5, played an important role in mediating the degradation of these proteins. Silencing cullin-1/5 decreased the phosphorylation of Iκ-Bα in response to Eh and LPS stimulation that markedly downregulated NF-κB-dependent TNF-α mRNA expression and TNF-α and MCP-1 pro-inflammatory cytokine production. This study unravelled a novel role for Eh-induced NLRP3 inflammasome activation of caspase-1 that intersected with the NF-κB pathway leading to the degradation of the novel substrates cullin-1/5 that regulates NF-κB-dependent pro-inflammatory cytokine production.
Collapse
Affiliation(s)
- Attinder Chadha
- Departments of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - France Moreau
- Departments of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Shanshan Wang
- Departments of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Antoine Dufour
- Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Biochemistry and Molecular Biology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Kris Chadee
- Departments of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
15
|
Uribe-Querol E, Rosales C. Immune Response to the Enteric Parasite Entamoeba histolytica. Physiology (Bethesda) 2021; 35:244-260. [PMID: 32490746 DOI: 10.1152/physiol.00038.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Entamoeba histolytica is a protozoan parasite responsible for amoebiasis, a disease with a high prevalence in developing countries. Establishing an amoebic infection involves interplay between pathogenic factors for invasion and tissue damage, and immune responses for protecting the host. Here, we review the pathogenicity of E. histolytica and summarize the latest knowledge on immune response and immune evasion mechanisms during amoebiasis.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
16
|
Rosales C. Neutrophils vs. amoebas: Immunity against the protozoan parasite Entamoeba histolytica. J Leukoc Biol 2021; 110:1241-1252. [PMID: 34085314 DOI: 10.1002/jlb.4mr0521-849rr] [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: 02/05/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 12/29/2022] Open
Abstract
Entamoeba histolytica is a protozoan parasite with high prevalence in developing countries, and causes amoebiasis. This disease affects the intestine and the liver, and is the third leading cause of human deaths among parasite infections. E. histolytica infection of the intestine or liver is associated with a strong inflammation characterized by a large number of infiltrating neutrophils. Consequently, several reports suggest that neutrophils play a protective role in amoebiasis. However, other reports indicate that amoebas making direct contact with neutrophils provoke lysis of these leukocytes, resulting in the release of their lytic enzymes, which in turn provoke tissue damage. Therefore, the role of neutrophils in this parasitic infection remains controversial. Neutrophils migrate from the circulation to sites of infection, where they display several antimicrobial functions, including phagocytosis, degranulation, and formation of neutrophil extracellular traps (NET). Recently, it was found that E. histolytica trophozoites are capable of inducing NET formation. Neutrophils in touch with amoebas launched NET in an explosive manner around the amoebas and completely covered them in nebulous DNA and cell aggregates where parasites got immobilized and killed. In addition, the phenotype of neutrophils can be modified by the microbiome resulting in protection against amoebas. This review describes the mechanisms of E. histolytica infection and discusses the novel view of how neutrophils are involved in innate immunity defense against amoebiasis. Also, the mechanisms on how the microbiome modulates neutrophil function are described.
Collapse
Affiliation(s)
- Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
17
|
Uddin MJ, Leslie JL, Petri WA. Host Protective Mechanisms to Intestinal Amebiasis. Trends Parasitol 2021; 37:165-175. [PMID: 33502317 PMCID: PMC7840892 DOI: 10.1016/j.pt.2020.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
The protozoan parasite Entamoeba histolytica is the causative agent of amebiasis, an infection that manifests as colitis and, in some cases, liver abscess. A better understanding of host protective factors is key to developing an effective remedy. Recently, significant advances have been made in understanding the mechanisms of MUC2 production by goblet cells upon amebic infection, regulation of antimicrobial peptide production by Paneth cells, the interaction of commensal microbiota with immune stimulation, and host genetics in conferring protection from amebiasis. In addition to host pathways that may serve as potential therapeutic targets, significant progress has also been made with respect to development of a vaccine against amebiasis. Here, we aim to highlight the current understanding and knowledge gaps critically.
Collapse
Affiliation(s)
- Md Jashim Uddin
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jhansi L Leslie
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - William A Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
18
|
Flores-Sanchez F, Chavez-Dueñas L, Sanchez-Villamil J, Navarro-Garcia F. Pic Protein From Enteroaggregative E. coli Induces Different Mechanisms for Its Dual Activity as a Mucus Secretagogue and a Mucinase. Front Immunol 2020; 11:564953. [PMID: 33281812 PMCID: PMC7705071 DOI: 10.3389/fimmu.2020.564953] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
A hallmark of enteroaggregative Escherichia coli (EAEC) infection is the formation of an intestinal biofilm, which comprises a mucus layer with immersed bacteria. Pic is an autotransporter secreted by EAEC, and other E. coli pathotypes, and has been involved in two apparently contradictory phenotypes, as a mucus secretagogue and as a mucinase. Here, we investigated this Pic dual activity, mucus secretagogue capability and mucinolytic activity, in human goblet cells that secrete MUC2 and MUC5AC. Pic induced mucus hypersecretion directly in the goblet cells, without other intestinal cell types involved. At the same time, Pic exhibited strong proteolytic activity on the secreted mucins. These activities were independent since a mutation in the serine protease motif (PicS258I) abolished mucin degradation while maintaining the mucus secretagogue activity intact. Furthermore, deoxycholic acid (DCA)-induced mucins were proteolytically degraded when goblet cells were co-incubated with DCA/Pic, while co-incubation with DCA/PicS258I induced a synergistic effect on mucus hypersecretion. Pic was more efficient degrading MUC5AC than MUC2, but no degradation was detected with Pic inactivated at the active site by mutation or pharmacological inhibition. Remarkably, Pic cleaved MUC2 and MUC5AC in the C-terminal domain, leaving N-terminal subproducts, impacting the feature of gel-forming mucins and allowing mucus layer penetration by EAEC. Astonishingly, Pic stimulated rapid mucin secretion in goblet-like cells by activating the intracellular calcium pathway resulting from the PLC signal transduction pathway, leading to the production of DAG and releasing IP3, a second messenger of calcium signaling. Therefore, the dual activity of Pic, as a mucus secretagogue and a mucinase, is relevant in the context of carbon source generation and mucus layer penetration, allowing EAEC to live within the layer of mucus but also access epithelial cells.
Collapse
Affiliation(s)
- Fernando Flores-Sanchez
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Lucia Chavez-Dueñas
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Javier Sanchez-Villamil
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| |
Collapse
|
19
|
Grondin JA, Kwon YH, Far PM, Haq S, Khan WI. Mucins in Intestinal Mucosal Defense and Inflammation: Learning From Clinical and Experimental Studies. Front Immunol 2020; 11:2054. [PMID: 33013869 PMCID: PMC7500085 DOI: 10.3389/fimmu.2020.02054] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/28/2020] [Indexed: 12/24/2022] Open
Abstract
Throughout the gastrointestinal (GI) tract, a distinct mucus layer composed of highly glycosylated proteins called mucins plays an essential role in providing lubrication for the passage of food, participating in cell signaling pathways and protecting the host epithelium from commensal microorganisms and invading pathogens, as well as toxins and other environmental irritants. These mucins can be broadly classified into either secreted gel-forming mucins, those that provide the structural backbone for the mucus barrier, or transmembrane mucins, those that form the glycocalyx layer covering the underlying epithelial cells. Goblet cells dispersed among the intestinal epithelial cells are chiefly responsible for the synthesis and secretion of mucins within the gut and are heavily influenced by interactions with the immune system. Evidence from both clinical and animal studies have indicated that several GI conditions, including inflammatory bowel disease (IBD), colorectal cancer, and numerous enteric infections are accompanied by considerable changes in mucin quality and quantity. These changes include, but are not limited to, impaired goblet cell function, synthesis dysregulation, and altered post-translational modifications. The current review aims to highlight the structural and functional features as well as the production and immunological regulation of mucins and the impact these key elements have within the context of barrier function and host defense in intestinal inflammation.
Collapse
Affiliation(s)
- Jensine A Grondin
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Yun Han Kwon
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Parsa Mehraban Far
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Sabah Haq
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
20
|
Begum S, Gorman H, Chadha A, Chadee K. Role of inflammasomes in innate host defense against Entamoeba histolytica. J Leukoc Biol 2020; 108:801-812. [PMID: 32498132 DOI: 10.1002/jlb.3mr0420-465r] [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: 12/10/2019] [Revised: 04/27/2020] [Accepted: 05/12/2020] [Indexed: 12/22/2022] Open
Abstract
Intestinal amebiasis is the disease caused by the extracellular protozoan parasite Entamoeba histolytica (Eh) that induces a dynamic and heterogeneous interaction profile with the host immune system during disease pathogenesis. In 90% of asymptomatic infection, Eh resides with indigenous microbiota in the outer mucus layer of the colon without prompting an immune response. However, for reasons that remain unclear, in a minority of the Eh-infected individuals, this fine tolerated relationship is switched to a pathogenic phenotype and advanced to an increasingly complex host-parasite interaction. Eh disease susceptibility depends on parasite virulence factors and their interactions with indigenous bacteria, disruption of the mucus bilayers, and adherence to the epithelium provoking host immune cells to evoke a robust pro-inflammatory response mediated by inflammatory caspases and inflammasome activation. To understand Eh pathogenicity and innate host immune responses, this review highlights recent advances in our understanding of how Eh induces outside-in signaling via Mϕs to activate inflammatory caspases and inflammasome to regulate pro-inflammatory responses.
Collapse
Affiliation(s)
- Sharmin Begum
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Hayley Gorman
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Attinder Chadha
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| |
Collapse
|
21
|
Entamoeba histolytica stimulates the alarmin molecule HMGB1 from macrophages to amplify innate host defenses. Mucosal Immunol 2020; 13:344-356. [PMID: 31772322 DOI: 10.1038/s41385-019-0233-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/22/2019] [Accepted: 11/07/2019] [Indexed: 02/04/2023]
Abstract
Even though Entamoeba histolytica (Eh)-induced host pro-inflammatory responses play a critical role in disease, we know very little about the host factors that regulate this response. Direct contact between host cell and Eh signify the highest level of danger, and to eliminate this threat, the host immune system elicits an augmented immune response. To understand the mechanisms of this response, we investigated the induction and release of the endogenous alarmin molecule high-mobility group box 1 (HMGB1) that act as a pro-inflammatory cytokine and chemoattractant during Eh infection. Eh in contact with macrophage induced a dose- and time-dependent secretion of HMGB1 in the absence of cell death. Secretion of HMGB1 was facilitated by Eh surface Gal-lectin-activated phosphoinositide 3-kinase and nuclear factor-κB signaling and up-regulation of histone acetyltransferase activity to trigger acetylated HMGB1 translocation from the nucleus. Unlike lipopolysaccharide, Eh-induced HMGB1 release was independent of caspase-1-mediated inflammasome and gasdermin D pores. In vivo, Eh inoculation in specific pathogen-free but not germ-free mice was associated with high levels of pro-inflammatory cytokines such as tumor necrosis factor-α, interleukin-1β, and keratinocyte-derived chemokine, which was suppressed with HMGB1 neutralization. This study reveals that Eh-induced active secretion of the HMGB1 plays a key role in shaping the pro-inflammatory landscape critical in innate host defense against amebiasis.
Collapse
|
22
|
Bao X, Wiehe R, Dommisch H, Schaefer A. Entamoeba gingivalis Causes Oral Inflammation and Tissue Destruction. J Dent Res 2020; 99:561-567. [DOI: 10.1177/0022034520901738] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A metagenomics analysis showed a strongly increased frequency of the protozoan Entamoeba gingivalis in inflamed periodontal pockets, where it contributed the second-most abundant rRNA after human rRNA. This observation and the close biological relationship to Entamoeba histolytica, which causes inflammation and tissue destruction in the colon of predisposed individuals, raised our concern about its putative role in the pathogenesis of periodontitis. Histochemical staining of gingival epithelium inflamed from generalized severe chronic periodontitis visualized the presence of E. gingivalis in conjunction with abundant neutrophils. We showed that on disruption of the epithelial barrier, E. gingivalis invaded gingival tissue, where it moved and fed on host cells. We validated the frequency of E. gingivalis in 158 patients with periodontitis and healthy controls by polymerase chain reaction and microscopy. In the cases, we detected the parasite in 77% of inflamed periodontal sites and 22% of healthy sites; 15% of healthy oral cavities were colonized by E. gingivalis. In primary gingival epithelial cells, we demonstrated by quantitative real-time polymerase chain reaction that infection with E. gingivalis but not with the oral bacterial pathogen Porphyromonas gingivalis strongly upregulated the inflammatory cytokine IL8 (1,900 fold, P = 2 × 10–4) and the epithelial barrier gene MUC21 (8-fold, P = 7 × 10–4). In gingival fibroblasts, we showed upregulation of the collagenase MMP13 (11-fold, P = 3 × 10–4). Direct contact of E. gingivalis to gingival epithelial cells inhibited cell proliferation. We indicated the strong virulence potential of E. gingivalis and showed that the mechanisms of tissue invasion and destruction are similar to the colonic protozoan parasite E. histolytica. In conjunction with abundant colonization of inflamed periodontal sites and the known resistance of Entamoeba species to neutrophils, antimicrobial peptides, and various antibiotics, our results raise the awareness of this protozoan as a potential and, to date, underrated microbial driver of destructive forms of periodontitis.
Collapse
Affiliation(s)
- X. Bao
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Dental and Craniofacial Sciences, Dept. of Periodontology and Synoptic Dentistry, Berlin, Germany
| | - R. Wiehe
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Dental and Craniofacial Sciences, Dept. of Periodontology and Synoptic Dentistry, Berlin, Germany
| | - H. Dommisch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Dental and Craniofacial Sciences, Dept. of Periodontology and Synoptic Dentistry, Berlin, Germany
| | - A.S. Schaefer
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Dental and Craniofacial Sciences, Dept. of Periodontology and Synoptic Dentistry, Berlin, Germany
| |
Collapse
|
23
|
Al-Hilfi AAU, Al-Malak MK, Al-Tomah MAU. Histopathological study of invasive and non-invasive Entamoeba spp. in experimental rats. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2019; 43:166. [DOI: 10.1186/s42269-019-0209-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 10/15/2019] [Indexed: 09/01/2023]
Abstract
Abstract
Background
The genus Entamoeba has many species that are invasive or non-invasive (E. histolytica, E. dispar, and E. moshkovskii).The invasive E. histolytica is the main pathogenic amoeba in human. Amoebiasis involves several stages starting with the adherence of the parasite to the intestinal epithelium, followed by degradation, tissue invasion, and distribution to other organs.
Results
The current study investigates the pathological changes of Entamoeba spp. infection in both rectum and cecum of experimental rats. The results showed the histological changes at the 7th, 14th, and 28th day post-infection for the three species. E. histolytica and E. moshkovskii infection showed less pathological changes compared to E. histolytica. These changes include the attachment of the trophozoites to the mucosal layer, significant surface epithelial changes such as dissociation and degeneration in the mucosal layer, and ulceration of the apical surface. Inflammatory cells infiltrate the varied regions, extending into the deep mucosa causing mild architectural alterations. These are features of amoebiasis.
Conclusion
The pathological changes reported in E. dispar and E. moshkovskii were less severe than E. histolytica.
Collapse
|
24
|
Sheats MK, Yin Q, Fang S, Park J, Crews AL, Parikh I, Dickson B, Adler KB. MARCKS and Lung Disease. Am J Respir Cell Mol Biol 2019; 60:16-27. [PMID: 30339463 DOI: 10.1165/rcmb.2018-0285tr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MARCKS (myristoylated alanine-rich C kinase substrate) is a prominent PKC substrate expressed in all eukaryotic cells. It is known to bind to and cross-link actin filaments, to serve as a bridge between Ca2+/calmodulin and PKC signaling, and to sequester the signaling molecule phosphatidylinositol 4,5-bisphosphate in the plasma membrane. Since the mid-1980s, this evolutionarily conserved and ubiquitously expressed protein has been associated with regulating cellular events that require dynamic actin reorganization, including cellular adhesion, migration, and exocytosis. More recently, translational studies have implicated MARCKS in the pathophysiology of a number of airway diseases, including chronic obstructive pulmonary disease, asthma, lung cancer, and acute lung injury/acute respiratory distress syndrome. This article summarizes the structure and cellular function of MARCKS (also including MARCKS family proteins and MARCKSL1 [MARCKS-like protein 1]). Evidence for MARCKS's role in several lung diseases is discussed, as are the technological innovations that took MARCKS-targeting strategies from theoretical to therapeutic. Descriptions and updates derived from ongoing clinical trials that are investigating inhalation of a MARCKS-targeting peptide as therapy for patients with chronic bronchitis, lung cancer, and ARDS are provided.
Collapse
Affiliation(s)
| | - Qi Yin
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| | - Shijing Fang
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| | - Joungjoa Park
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| | - Anne L Crews
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| | - Indu Parikh
- 3 BioMarck Pharmaceuticals, Durham, North Carolina
| | | | - Kenneth B Adler
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| |
Collapse
|
25
|
Cornick S, Kumar M, Moreau F, Gaisano H, Chadee K. VAMP8-mediated MUC2 mucin exocytosis from colonic goblet cells maintains innate intestinal homeostasis. Nat Commun 2019; 10:4306. [PMID: 31541089 PMCID: PMC6754373 DOI: 10.1038/s41467-019-11811-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
The mucus layer is the first line of innate host defense in the gut that protects the epithelium by spatially separating commensal bacteria. MUC2 mucin is produced and stored by goblet cells that is constitutively exocytosed or hyper secreted upon sensing a threat. How coordinated mucus exocytosis maintains homeostasis in the intestinal epithelium and modulates the immunological landscape remains elusive. Here we describe how the vesicle SNARE protein VAMP8 coordinates mucin exocytosis from goblet cells. Vamp8-/- exhibit a mild pro-inflammatory state basally due to an altered mucus layer and increased encounters with microbial antigens. Microbial diversity shifts to a detrimental microbiota with an increase abundance of pathogenic and mucolytic bacteria. To alleviate the heavy microbial burden and inflammatory state basally, Vamp8-/- skews towards tolerance. Despite this, Vamp8-/- is highly susceptible to both chemical and infectious colitis demonstrating the fragility of the intestinal mucosa without proper mucus exocytosis mechanisms.
Collapse
Affiliation(s)
- Steve Cornick
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Manish Kumar
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - France Moreau
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Herbert Gaisano
- Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
26
|
Rangel-Castañeda IA, Carranza-Rosales P, Guzmán-Delgado NE, Hernández-Hernández JM, González-Pozos S, Pérez-Rangel A, Castillo-Romero A. Curcumin Attenuates the Pathogenicity of Entamoeba histolytica by Regulating the Expression of Virulence Factors in an Ex-Vivo Model Infection. Pathogens 2019; 8:pathogens8030127. [PMID: 31443160 PMCID: PMC6789811 DOI: 10.3390/pathogens8030127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 12/12/2022] Open
Abstract
Infection with the enteric protozoan Entamoeba histolytica is still a serious public health problem, especially in developing countries. Amoebic liver abscess (ALA) is the most common extraintestinal manifestation of the amoebiasis, and it can lead to serious and potentially life-threatening complications in some people. ALA can be cured by metronidazole (MTZ); however, because it has poor activity against luminal trophozoites, 40–60% of treated patients get repeated episodes of invasive disease and require repeated treatments that can induce resistance to MTZ, this may emerge as an important public health problem. Anti-virulence strategies that impair the virulence of pathogens are one of the novel approaches to solving the problem. In this study, we found that low doses of curcumin (10 and 50 μM) attenuate the virulence of E. histolytica without affecting trophozoites growth or triggering liver injury. Curcumin (CUR) decreases the expression of genes associated with E. histolytica virulence (gal/galnac lectin, ehcp1, ehcp5, and amoebapore), and is correlated with significantly lower amoebic invasion. In addition, oxidative stress is critically involved in the etiopathology of amoebic liver abscess; our results show no changes in mRNA expression levels of superoxide dismutase (SOD) and catalase (CAT) after E. histolytica infection, with or without CUR. This study provides clear evidence that curcumin could be an anti-virulence agent against E. histolytica, and makes it an attractive potential starting point for effective treatments that reduce downstream amoebic liver abscess.
Collapse
Affiliation(s)
- Itzia Azucena Rangel-Castañeda
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Pilar Carranza-Rosales
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, Mexico
| | | | - José Manuel Hernández-Hernández
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Sirenia González-Pozos
- Unidad de Microscopía Electrónica LaNSE, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Armando Pérez-Rangel
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Araceli Castillo-Romero
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico.
| |
Collapse
|
27
|
Betanzos A, Bañuelos C, Orozco E. Host Invasion by Pathogenic Amoebae: Epithelial Disruption by Parasite Proteins. Genes (Basel) 2019; 10:E618. [PMID: 31416298 PMCID: PMC6723116 DOI: 10.3390/genes10080618] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
The epithelium represents the first and most extensive line of defence against pathogens, toxins and pollutant agents in humans. In general, pathogens have developed strategies to overcome this barrier and use it as an entrance to the organism. Entamoeba histolytica, Naegleriafowleri and Acanthamoeba spp. are amoebae mainly responsible for intestinal dysentery, meningoencephalitis and keratitis, respectively. These amoebae cause significant morbidity and mortality rates. Thus, the identification, characterization and validation of molecules participating in host-parasite interactions can provide attractive targets to timely intervene disease progress. In this work, we present a compendium of the parasite adhesins, lectins, proteases, hydrolases, kinases, and others, that participate in key pathogenic events. Special focus is made for the analysis of assorted molecules and mechanisms involved in the interaction of the parasites with epithelial surface receptors, changes in epithelial junctional markers, implications on the barrier function, among others. This review allows the assessment of initial host-pathogen interaction, to correlate it to the potential of parasite invasion.
Collapse
Affiliation(s)
- Abigail Betanzos
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Mexico City 03940, Mexico
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Cecilia Bañuelos
- Coordinación General de Programas de Posgrado Multidisciplinarios, Programa de Doctorado Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, CINVESTAV-IPN, Mexico City 07360, Mexico
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV-IPN, Mexico City 07360, Mexico.
| |
Collapse
|
28
|
Allain T, Fekete E, Buret AG. Giardia Cysteine Proteases: The Teeth behind the Smile. Trends Parasitol 2019; 35:636-648. [DOI: 10.1016/j.pt.2019.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 01/15/2023]
|
29
|
Abstract
Entamoeba histolytica (Eh) is a protozoan parasite of humans that colonizes the outer colonic mucus layer. Under conditions not fully understood, Eh breaches innate host defenses and invades the intestinal mucosa-causing amebic colitis and liver abscess. In asymptomatic infection, Eh interacts with and feeds on resident microbiota that forms biofilms on the outer colonic mucus layer. Despite the close association between Eh and commensal microbiota, we still lack basic knowledge on whether microbiota and/or their metabolites influence Eh virulence traits critical in disease pathogenesis. In the pathogenesis of intestinal amebiasis, Eh overcomes the protective mucus layer using a combination of mucinase/glycosidase and potent mucus secretagogue activity. In this addendum, we discuss the interconnected role of a healthy mucus barrier and the role commensal microbiota play in shaping innate host defense against Eh-induced pro-inflammatory and secretory responses critical in disease pathogenesis.
Collapse
Affiliation(s)
- Aralia Leon-Coria
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Manish Kumar
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada,CONTACT Kris Chadee Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Shahi P, Moreau F, Chadee K. Entamoeba histolytica Cyclooxygenase-Like Protein Regulates Cysteine Protease Expression and Virulence. Front Cell Infect Microbiol 2019; 8:447. [PMID: 30687644 PMCID: PMC6333869 DOI: 10.3389/fcimb.2018.00447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022] Open
Abstract
The intestinal protozoan parasite Entamoeba histolytica (Eh) causes amebiasis associated with severe diarrhea and/or liver abscess. Eh pathogenesis is multifactorial requiring both parasite virulent molecules and host-induced innate immune responses. Eh-induced host pro-inflammatory responses plays a critical role in disease pathogenesis by causing damage to tissues allowing parasites access to systemic sites. Eh cyclooxygenase (EhCox) derived prostaglandin E2 stimulates the chemokine IL-8 from mucosal epithelial cells that recruits neutrophils to the site of infection to exacerbate disease. At present, it is not known how EhCox is regulated or whether it affects the expression of other proteins in Eh. In this study, we found that gene silencing of EhCox (EhCoxgs) markedly increased endogenous cysteine protease (CP) protein expression and virulence without altering CP gene transcripts. Live virulent Eh pretreated with arachidonic acid substrate to enhance PGE2 production or aspirin to inhibit EhCox enzyme activity or addition of exogenous PGE2 to Eh had no effect on EhCP activity. Increased CP enzyme activity in EhCoxgs was stable and significantly enhanced erythrophagocytosis, cytopathic effects on colonic epithelial cells and elicited pro-inflammatory cytokines in mice colonic loops. Acute infection with EhCoxgs in colonic loops increased inflammation associated with high levels of myeloperoxidase activity. This study has identified EhCox protein as one of the important endogenous regulators of cysteine protease activity. Alterations of CP activity in response to Cox gene silencing may be a negative feedback mechanism in Eh to limit proteolytic activity during colonization that can inadvertently trigger inflammation in the gut.
Collapse
Affiliation(s)
| | | | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
31
|
Betanzos A, Zanatta D, Bañuelos C, Hernández-Nava E, Cuellar P, Orozco E. Epithelial Cells Expressing EhADH, An Entamoeba histolytica Adhesin, Exhibit Increased Tight Junction Proteins. Front Cell Infect Microbiol 2018; 8:340. [PMID: 30324093 PMCID: PMC6172307 DOI: 10.3389/fcimb.2018.00340] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 09/07/2018] [Indexed: 12/15/2022] Open
Abstract
In Entamoeba histolytica, the EhADH adhesin together with the EhCP112 cysteine protease, form a 124 kDa complex named EhCPADH. This complex participates in trophozoite adherence, phagocytosis and cytolysis of target cells. EhCPADH and EhCP112 are both involved on epithelium damage, by opening tight junctions (TJ) and reaching other intercellular junctions. EhADH is a scaffold protein belonging to the ALIX family that contains a Bro1 domain, expresses at plasma membrane, endosomes and cytoplasm of trophozoites, and is also secreted to the medium. Contribution of EhADH to TJ opening still remains unknown. In this paper, to elucidate the role of EhADH on epithelium injury, we followed two strategies: producing a recombinant protein (rEhADH) and transfecting the ehadh gene in MDCK cells. Results from the first strategy revealed that rEhADH reached the intercellular space of epithelial cells and co-localized with claudin-1 and occludin at TJ region; later, rEhADH was mainly internalized by clathrin-coated vesicles. In the second strategy, MDCK cells expressing EhADH (MDCK-EhADH) showed the adhesin at plasma membrane. In addition, MDCK-EHADH cells exhibited adhesive features, producing epithelial aggregation and adherence to erythrocytes, as described in trophozoites. Surprisingly, the adhesin expression produced an increase of claudin-1, occludin, ZO-1 and ZO-2 at TJ, and also the transepithelial electric resistance (TEER), which is a measure of TJ gate function. Moreover, MDCK-EhADH cells resulted more susceptible to trophozoites attack, as showed by TEER and cytopathic experiments. Overall, our results indicated that EhADH disturbed TJ from the extracellular space and also intracellularly, suggesting that EhADH affects by itself TJ proteins, and possibly synergizes the action of other parasite molecules during epithelial invasion.
Collapse
Affiliation(s)
- Abigail Betanzos
- Consejo Nacional de Ciencia y Tecnología, Mexico City, Mexico.,Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Dxinegueela Zanatta
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Cecilia Bañuelos
- Coordinación General de Programas de Posgrado Multidisciplinarios, Programa de Doctorado Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Patricia Cuellar
- Centro Regional de Educación Superior, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
32
|
Gastelum-Martínez A, León-Sicairos C, Plata-Guzmán L, Soto-Castro L, León-Sicairos N, de la Garza M. Iron-modulated virulence factors of Entamoeba histolytica. Future Microbiol 2018; 13:1329-1341. [PMID: 30238768 DOI: 10.2217/fmb-2018-0066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Entamoeba histolytica is a human parasite that causes amoebiasis, a disease that affects the colon and liver and is prevalent worldwide. This protozoan requires a high concentration of iron to survive and reproduce. Iron modulates the expression of parasite virulence factors, including hemoglobinases, hemoglobin-binding proteins and cysteine proteases, as well as proteins related to the amoebic cytoskeleton. This review summarizes the virulence factors that are affected by iron, resulting in upregulation or downregulation of E. histolytica genes. This review also discusses the functionality of iron in the mechanisms of pathogenesis.
Collapse
Affiliation(s)
- Aurora Gastelum-Martínez
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas. Universidad Autónoma de Sinaloa. Av. de las Américas y Josefa Ortiz (Cd. Universitaria) Culiacán 80030, Sinaloa, Mexico
| | - Claudia León-Sicairos
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas. Universidad Autónoma de Sinaloa. Av. de las Américas y Josefa Ortiz (Cd. Universitaria) Culiacán 80030, Sinaloa, Mexico
| | - Laura Plata-Guzmán
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas. Universidad Autónoma de Sinaloa. Av. de las Américas y Josefa Ortiz (Cd. Universitaria) Culiacán 80030, Sinaloa, Mexico
| | - Liliana Soto-Castro
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas. Universidad Autónoma de Sinaloa. Av. de las Américas y Josefa Ortiz (Cd. Universitaria) Culiacán 80030, Sinaloa, Mexico
| | - Nidia León-Sicairos
- CIASaP Facultad de Medicina. Universidad Autónoma de Sinaloa. Cedros y Sauces Frac. Fresnos. Culiacán 80246, Sinaloa, México; Departamento de Investigación, Hospital Pediátrico de Sinaloa. Boulevard Constitución S/N, Col. Jorge Almada, Culiacán 80200, Sinaloa, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Colonia San Pedro Zacatenco 07360, CdMx, Mexico
| |
Collapse
|
33
|
Entamoeba histolytica Alters Ileal Paneth Cell Functions in Intact and Muc2 Mucin Deficiency. Infect Immun 2018; 86:IAI.00208-18. [PMID: 29685982 DOI: 10.1128/iai.00208-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/14/2018] [Indexed: 12/19/2022] Open
Abstract
Enteric α-defensins, termed cryptdins (Crps) in mice, and lysozymes secreted by Paneth cells contribute to innate host defense in the ileum. Antimicrobial factors, including lysozymes and β-defensins, are often embedded in luminal glycosylated colonic Muc2 mucin secreted by goblet cells that form the protective mucus layer critical for gut homeostasis and pathogen invasion. In this study, we investigated ileal innate immunity against Entamoeba histolytica, the causative agent of intestinal amebiasis, by inoculating parasites in closed ileal loops in Muc2+/+ and Muc2-/- littermates and quantifying Paneth cell localization (lysozyme expression) and function (Crp secretion). Relative to Muc2+/+ littermates, Muc2-/- littermates showed a disorganized mislocalization of Paneth cells that was diffusely distributed, with elevated lysozyme secretion in the crypts and on villi in response to E. histolytica Inhibition of E. histolytica Gal/GalNAc lectin (Gal-lectin) binding with exogenous galactose and Entamoeba histolytica cysteine proteinase 5 (EhCP5)-negative E. histolytica had no effect on parasite-induced erratic Paneth cell lysozyme synthesis. Although the basal ileal expression of Crp genes was unaffected in Muc2-/- mice in response to E. histolytica, there was a robust release of proinflammatory cytokines and Crp peptide secretions in luminal exudates that was also present in the colon. Interestingly, E. histolytica-secreted cysteine proteinases cleaved the proregion of Crp4 but not the active form. These findings define Muc2 mucin as an essential component of ileal barrier function that regulates the localization and function of Paneth cells critical for host defense against microbes.
Collapse
|
34
|
Li G, Yang S, Shen P, Wu B, Sun T, Sun H, Ji F, Zhou D. SCF/c-KIT signaling promotes mucus secretion of colonic goblet cells and development of mucinous colorectal adenocarcinoma. Am J Cancer Res 2018; 8:1064-1073. [PMID: 30034943 PMCID: PMC6048403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/16/2018] [Indexed: 06/08/2023] Open
Abstract
Mucinous colorectal adenocarcinoma (MCA) is characterized by a great mount of extracellular mucus fundamentally composed of Mucin2 (MUC2) which is significantly correlated with the high malignancy and strong invasive ability of MCA. However, rare is known about the underlying mechanism of the mucus accumulation in MCA. Our latest study demonstrated that SCF/c-KIT signaling was highly activated in MCA patients and mouse model, which up-regulated MUC2 transcription. In the present study, we paid a special interest in whether and how SCF/c-KIT signaling promoted mucus secretion by using wild-type (WT) C57BL mice and their littermates who harbor mutational c-kit gene (Wadsm/m), clinical colorectal cancer (CRC) samples, as well as human CRC cell lines. Our results clearly showed that the inner mucus layer of colon was thinner and the intracellular mucin residual was more in Wadsm/m mice than those in WT mice by Alcian blue and PAS staining, suggesting that the mucus secretion process was crippled when SCF/c-KIT signaling was hypo-activated. Inhibiting SCF/c-KIT signaling by Imatinib also resulted in weakened mucus secretion in WT mice. Intraperitoneal administration of MANS which competitively inhibits the activity of the vesicular transport protein MARCKS efficiently reduced mucus secretion in colonic goblet cells of WT mice. Significantly, phosphorylated MARCKS (p-MARCKS) was overtly decreased in colonic mucosa of Wadsm/m mice compared with WT mice, indicating that SCF/c-KIT signaling-regulated mucus secretion was probably mediated by MARCKS activation. Similar results were obtained in MCA patients and mouse model. Moreover, SCF/c-KIT signaling was activated or inhibited in HT-29 and LS174T CRC cells, which potently increased or decreased MARCKS activity, respectively. Finally, we found that PKCδ, a known kinase for MARCKS, was activated in WT and MCA mice along with MARCKS. Inhibition or activation of SCF/c-KIT signaling resulted in decreased or increased PKCδ activity respectively in vitro. In conclusion, we demonstrated that SCF/c-KIT signaling can promote the mucus secretion by activating PKCδ-MARCKS, which provided a new insight into understanding the mechanism of mucus secretion of goblet cells and MCA development.
Collapse
Affiliation(s)
- Guilan Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical UniversityBeijing 100069, P. R. China
| | - Shu Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical UniversityBeijing 100069, P. R. China
- Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchBeijing 100069, P. R. China
- Cancer Institute of Capital Medical UniversityBeijing 100069, P. R. China
| | - Ping Shen
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical UniversityBeijing 100069, P. R. China
| | - Bo Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical UniversityBeijing 100069, P. R. China
- Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchBeijing 100069, P. R. China
- Cancer Institute of Capital Medical UniversityBeijing 100069, P. R. China
| | - Tingyi Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical UniversityBeijing 100069, P. R. China
- Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchBeijing 100069, P. R. China
- Cancer Institute of Capital Medical UniversityBeijing 100069, P. R. China
| | - Haimei Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical UniversityBeijing 100069, P. R. China
- Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchBeijing 100069, P. R. China
- Cancer Institute of Capital Medical UniversityBeijing 100069, P. R. China
| | - Fengqing Ji
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical UniversityBeijing 100069, P. R. China
| | - Deshan Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical UniversityBeijing 100069, P. R. China
- Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchBeijing 100069, P. R. China
- Cancer Institute of Capital Medical UniversityBeijing 100069, P. R. China
| |
Collapse
|
35
|
Cornick S, Chadee K. Entamoeba histolytica: Host parasite interactions at the colonic epithelium. Tissue Barriers 2018; 5:e1283386. [PMID: 28452682 DOI: 10.1080/21688370.2017.1283386] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Entamoeba histolytica (Eh) is the protozoan parasite responsible for intestinal amebiasis and interacts dynamically with the host intestinal epithelium during disease pathogenesis. A multifaceted pathogenesis profile accounts for why 90% of individuals infected with Eh are largely asymptomatic. For 100 millions individuals that are infected each year, key interactions within the intestinal mucosa dictate disease susceptibility. The ability for Eh to induce amebic colitis and disseminate into extraintestinal organs depends on the parasite competing with indigenous bacteria and overcoming the mucus barrier, binding to host cells inducing their cell death, invasion through the mucosa and outsmarting the immune system. In this review we summarize how Eh interacts with the intestinal epithelium and subverts host defense mechanisms in disease pathogenesis.
Collapse
Affiliation(s)
- Steve Cornick
- a Department of Microbiology, Immunology and Infectious Diseases , Snyder Institute for Chronic Diseases, University of Calgary , Calgary , Alberta , Canada
| | - Kris Chadee
- a Department of Microbiology, Immunology and Infectious Diseases , Snyder Institute for Chronic Diseases, University of Calgary , Calgary , Alberta , Canada
| |
Collapse
|
36
|
Beyond the Matrix: The Many Non-ECM Ligands for Integrins. Int J Mol Sci 2018; 19:ijms19020449. [PMID: 29393909 PMCID: PMC5855671 DOI: 10.3390/ijms19020449] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/21/2018] [Accepted: 01/30/2018] [Indexed: 12/17/2022] Open
Abstract
The traditional view of integrins portrays these highly conserved cell surface receptors as mediators of cellular attachment to the extracellular matrix (ECM), and to a lesser degree, as coordinators of leukocyte adhesion to the endothelium. These canonical activities are indispensable; however, there is also a wide variety of integrin functions mediated by non-ECM ligands that transcend the traditional roles of integrins. Some of these unorthodox roles involve cell-cell interactions and are engaged to support immune functions such as leukocyte transmigration, recognition of opsonization factors, and stimulation of neutrophil extracellular traps. Other cell-cell interactions mediated by integrins include hematopoietic stem cell and tumor cell homing to target tissues. Integrins also serve as cell-surface receptors for various growth factors, hormones, and small molecules. Interestingly, integrins have also been exploited by a wide variety of organisms including viruses and bacteria to support infectious activities such as cellular adhesion and/or cellular internalization. Additionally, the disruption of integrin function through the use of soluble integrin ligands is a common strategy adopted by several parasites in order to inhibit blood clotting during hematophagy, or by venomous snakes to kill prey. In this review, we strive to go beyond the matrix and summarize non-ECM ligands that interact with integrins in order to highlight these non-traditional functions of integrins.
Collapse
|
37
|
Abstract
The protozoan parasite Entamoeba histolytica is the microbial agent of amoebiasis - an infection that is endemic worldwide and is associated with high morbidity and mortality rates. As the disease develops, virulent E. histolytica deplete the mucus layer, interact with the intestinal epithelium, and then degrade the colonic mucosa and disrupt the extracellular matrix (ECM). Our research demonstrated that virulent parasites with an invasive phenotype display rapid, highly specific changes in their transcriptome (notably for essential factors involved in carbohydrate metabolism and the processing of glycosylated residues). Moreover, combined activation of parasite and host lytic enzymes leads to the destruction of the intestinal parenchyma. Together, these enzymes degrade the mucus layer and the ECM, and trigger the inflammatory response essential to the development of amoebiasis.
Collapse
|
38
|
Entamoeba histolytica-Induced Mucin Exocytosis Is Mediated by VAMP8 and Is Critical in Mucosal Innate Host Defense. mBio 2017; 8:mBio.01323-17. [PMID: 28974617 PMCID: PMC5626970 DOI: 10.1128/mbio.01323-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Intestinal mucus secretion is critical in maintaining mucosal host defense against a myriad of pathogens by preventing direct association with the epithelium. Entamoeba histolytica specifically binds colonic MUC2 mucin and also induces potent hypersecretion from goblet cells; however, characterization of the nature of the mechanisms controlling mucus release remains elusive. In this report, we identify vesicle SNARE vesicle-associated membrane protein 8 (VAMP8) present on mucin granules as orchestrating regulated exocytosis in human goblet cells in response to the presence of E. histolytica. VAMP8 was specifically activated during E. histolytica infection, and ablation of VAMP8 led to impaired mucin secretion. As a consequence, loss of VAMP8 increased E. histolytica adherence to epithelial cells associated with enhanced cell death through apoptosis characterized by caspase 3 and 9 cleavages and DNA fragmentation. With the mucosal barrier compromised in Vamp8−/− animals, E. histolytica induced an aggressive proinflammatory response with elevated levels of interleukin-1 alpha (IL-1α), IL-1β, and tumor necrosis factor alpha (TNF-α) secretion. This report is the first to characterize regulated mucin exocytosis in intestinal goblet cells in response to a pathogen and the downstream consequences of improper mucin secretion in mucosal barrier defense. The intestinal tract is exposed to countless substances and pathogens, and yet homeostasis is maintained, in part by the mucus layer that houses the microbiota and spatially separates potential threats from the underlying single layer of epithelium. Despite the critical role of mucus in innate host defense, characterization of the mechanisms by which mucus is secreted from specialized goblet cells in the gut remains elusive. Here, we describe the machinery that regulates mucus secretion as well as the consequence during infection with the colonic pathogen Entamoeba histolytica. Abolishment of the key machinery protein VAMP8 abrogated mucus release in cultured human colonic goblet cells and during E. histolytica infection in Vamp8−/− mice, which showed enhanced amoeba contact and killing of epithelial cells, triggering a potent proinflammatory response. This report highlights the importance of the VAMP8 secretory machinery in facilitating mucus release from intestinal goblet cells and the dire consequences that occur during disease pathogenesis if these pathways are not functional.
Collapse
|
39
|
St-Pierre J, Moreau F, Cornick S, Quach J, Begum S, Aracely Fernandez L, Gorman H, Chadee K. The macrophage cytoskeleton acts as a contact sensor upon interaction with Entamoeba histolytica to trigger IL-1β secretion. PLoS Pathog 2017; 13:e1006592. [PMID: 28837696 PMCID: PMC5587335 DOI: 10.1371/journal.ppat.1006592] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/06/2017] [Accepted: 08/18/2017] [Indexed: 01/09/2023] Open
Abstract
Entamoeba histolytica (Eh) is the causative agent of amebiasis, one of the major causes of dysentery-related morbidity worldwide. Recent studies have underlined the importance of the intercellular junction between Eh and host cells as a determinant in the pathogenesis of amebiasis. Despite the fact that direct contact and ligation between Eh surface Gal-lectin and EhCP-A5 with macrophage α5β1 integrin are absolute requirements for NLRP3 inflammasome activation and IL-1β release, many other undefined molecular events and downstream signaling occur at the interface of Eh and macrophage. In this study, we investigated the molecular events at the intercellular junction that lead to recognition of Eh through modulation of the macrophage cytoskeleton. Upon Eh contact with macrophages key cytoskeletal-associated proteins were rapidly post-translationally modified only with live Eh but not with soluble Eh proteins or fragments. Eh ligation with macrophages rapidly activated caspase-6 dependent cleavage of the cytoskeletal proteins talin, Pyk2 and paxillin and caused robust release of the pro-inflammatory cytokine, IL-1β. Macrophage cytoskeletal cleavages were dependent on Eh cysteine proteinases EhCP-A1 and EhCP-A4 but not EhCP-A5 based on pharmacological blockade of Eh enzyme inhibitors and EhCP-A5 deficient parasites. These results unravel a model where the intercellular junction between macrophages and Eh form an area of highly interacting proteins that implicate the macrophage cytoskeleton as a sensor for Eh contact that leads downstream to subsequent inflammatory immune responses. The protozoan parasite Entamoeba histolytica can establish an enteric infection in human hosts that leads to symptoms ranging from diarrhea to abscesses in the liver and the brain. Host susceptibility to amebic infection is in part determined by the quality and potency of the host immune response that occurs once the parasite overcomes the mucus bilayers and colonic epithelial barriers, and invades underlying tissues. At the cellular level, one of the key events that shape the inflammatory response occurs during direct parasite interaction with host macrophages via surface proteins. The ensuing cascades of intracellular signaling events have only partly been uncovered. Interestingly, only direct interaction between live parasites and macrophages, as opposed to soluble factors or dead parasites, is a prerequisite to the generation of a prompt raging pro-inflammatory response. We have sought to further elucidate the mechanisms by which macrophages distinguish live parasites and found that the macrophage cell skeleton undergoes rapid significant alteration upon Eh contact. Furthermore, we uncovered a previously unknown role for two Eh enzymes in triggering macrophage pro-inflammatory responses. Through this work, we gain a better understanding of the molecular interactions that occur at the macrophage-ameba interface that regulate host inflammatory responses.
Collapse
Affiliation(s)
- Joëlle St-Pierre
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - France Moreau
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Steve Cornick
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jeanie Quach
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Sharmin Begum
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Luz Aracely Fernandez
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Hayley Gorman
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
40
|
Amat CB, Motta JP, Fekete E, Moreau F, Chadee K, Buret AG. Cysteine Protease-Dependent Mucous Disruptions and Differential Mucin Gene Expression in Giardia duodenalis Infection. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2486-2498. [PMID: 28823873 DOI: 10.1016/j.ajpath.2017.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 02/08/2023]
Abstract
The intestinal mucous layer provides a critical host defense against pathogen exposure and epithelial injury, yet little is known about how enteropathogens may circumvent this physiologic barrier. Giardia duodenalis is a small intestinal parasite responsible for diarrheal disease and chronic postinfectious illness. This study reveals a complex interaction at the surface of epithelial cells, between G. duodenalis and the intestinal mucous layer. Here, we reveal mechanisms whereby G. duodenalis evades and disrupts the first line of host defense by degrading human mucin-2 (MUC2), depleting mucin stores and inducing differential gene expression in the mouse small and large intestines. Human colonic biopsy specimens exposed to G. duodenalis were depleted of mucus, and in vivo mice infected with G. duodenalis had a thinner mucous layer and demonstrated differential Muc2 and Muc5ac mucin gene expression. Infection in Muc2-/- mice elevated trophozoite colonization in the small intestine and impaired weight gain. In vitro, human LS174T goblet-like cells were depleted of mucus and had elevated levels of MUC2 mRNA expression after G. duodenalis exposure. Importantly, the cysteine protease inhibitor E64 prevented mucous degradation, mucin depletion, and the increase in MUC2 expression. This article describes a novel role for Giardia's cysteine proteases in pathogenesis and how Giardia's disruptions of the mucous barrier facilitate bacterial translocation that may contribute to the onset and propagation of disease.
Collapse
Affiliation(s)
- Christina B Amat
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Jean-Paul Motta
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Elena Fekete
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - France Moreau
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, Calgary, Alberta, Canada.
| | - Andre G Buret
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
41
|
Cuellar P, Hernández-Nava E, García-Rivera G, Chávez-Munguía B, Schnoor M, Betanzos A, Orozco E. Entamoeba histolytica EhCP112 Dislocates and Degrades Claudin-1 and Claudin-2 at Tight Junctions of the Intestinal Epithelium. Front Cell Infect Microbiol 2017; 7:372. [PMID: 28861400 PMCID: PMC5561765 DOI: 10.3389/fcimb.2017.00372] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/02/2017] [Indexed: 12/11/2022] Open
Abstract
During intestinal invasion, Entamoeba histolytica opens tight junctions (TJs) reflected by transepithelial electrical resistance (TEER) dropping. To explore the molecular mechanisms underlying this, we studied in vitro and in vivo the damage produced by the recombinant E. histolytica cysteine protease (rEhCP112) on TJ functions and proteins. rEhCP112 reduced TEER in Caco-2 cells in a dose- and time-dependent manner; and EhCP112-overexpressing trophozoites provoked major epithelial injury compared to control trophozoites. rEhCP112 penetrated through the intercellular space, and consequently the ion flux increased and the TJs fence function was disturbed. However, macromolecular flux was not altered. Functional in vitro assays revealed specific association of rEhCP112 with claudin-1 and claudin-2, that are both involved in regulating ion flux and fence function. Of note, rEhCP112 did not interact with occludin that is responsible for regulating macromolecular flux. Moreover, rEhCP112 degraded and delocalized claudin-1, thus affecting interepithelial adhesion. Concomitantly, expression of the leaky claudin-2 at TJ, first increased and then it was degraded. In vivo, rEhCP112 increased intestinal epithelial permeability in the mouse colon, likely due to apical erosion and claudin-1 and claudin-2 degradation. In conclusion, we provide evidence that EhCP112 causes epithelial dysfunction by specifically altering claudins at TJ. Thus, EhCP112 could be a potential target for therapeutic approaches against amoebiasis.
Collapse
Affiliation(s)
- Patricia Cuellar
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico NacionalMexico, Mexico
| | - Elizabeth Hernández-Nava
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico NacionalMexico, Mexico
| | - Guillermina García-Rivera
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico NacionalMexico, Mexico
| | - Bibiana Chávez-Munguía
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico NacionalMexico, Mexico
| | - Michael Schnoor
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico NacionalMexico, Mexico
| | - Abigail Betanzos
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico NacionalMexico, Mexico.,Consejo Nacional de Ciencia y TecnologíaMexico, Mexico
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico NacionalMexico, Mexico
| |
Collapse
|
42
|
Ngobeni R, Abhyankar MM, Jiang NM, Farr LA, Samie A, Haque R, Moonah SN. Entamoeba histolytica-Encoded Homolog of Macrophage Migration Inhibitory Factor Contributes to Mucosal Inflammation during Amebic Colitis. J Infect Dis 2017; 215:1294-1302. [PMID: 28186296 DOI: 10.1093/infdis/jix076] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022] Open
Abstract
Understanding the mechanisms by which Entamoeba histolytica drives gut inflammation is critical for the development of improved preventive and therapeutic strategies. E. histolytica encodes a homolog of the human cytokine macrophage migration inhibitory factor (MIF). Here, we investigated the role of E. histolytica MIF (EhMIF) during infection. We found that the concentration of fecal EhMIF correlated with the level of intestinal inflammation in persons with intestinal amebiasis. Mice treated with antibodies that specifically block EhMIF had reduced chemokine expression and neutrophil infiltration in the mucosa. In addition to antibody-mediated neutralization, we used a genetic approach to test the effect of EhMIF on mucosal inflammation. Mice infected with parasites overexpressing EhMIF had increased chemokine expression, neutrophil influx, and mucosal damage. Together, these results uncover a specific parasite protein that increases mucosal inflammation, expands our knowledge of host-parasite interaction during amebic colitis, and highlights a potential immunomodulatory target.
Collapse
Affiliation(s)
- Renay Ngobeni
- Department of Medicine, University of Virginia Health System, Charlottesville
| | | | - Nona M Jiang
- Department of Medicine, University of Virginia Health System, Charlottesville
| | - Laura A Farr
- Department of Medicine, University of Virginia Health System, Charlottesville
| | - Amidou Samie
- Department of Microbiology, University of Venda, Limpopo Province, South Africa; and
| | - Rashidul Haque
- International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Shannon N Moonah
- Department of Medicine, University of Virginia Health System, Charlottesville
| |
Collapse
|
43
|
MUC2 Mucin and Butyrate Contribute to the Synthesis of the Antimicrobial Peptide Cathelicidin in Response to Entamoeba histolytica- and Dextran Sodium Sulfate-Induced Colitis. Infect Immun 2017; 85:IAI.00905-16. [PMID: 28069814 DOI: 10.1128/iai.00905-16] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/21/2016] [Indexed: 12/14/2022] Open
Abstract
Embedded in the colonic mucus are cathelicidins, small cationic peptides secreted by colonic epithelial cells. Humans and mice have one cathelicidin-related antimicrobial peptide (CRAMP) each, LL-37/hCAP-18 and Cramp, respectively, with related structure and functions. Altered production of MUC2 mucin and antimicrobial peptides is characteristic of intestinal amebiasis. The interactions between MUC2 mucin and cathelicidins in conferring innate immunity against Entamoeba histolytica are not well characterized. In this study, we quantified whether MUC2 expression and release could regulate the expression and secretion of cathelicidin LL-37 in colonic epithelial cells and in the colon. The synthesis of LL-37 was enhanced with butyrate (a product of bacterial fermentation) and interleukin-1β (IL-1β) (a proinflammatory cytokine in colitis) in the presence of exogenously added purified MUC2. The LL-37 responses to butyrate and IL-1β were higher in high-MUC2-producing cells than in lentivirus short hairpin RNA (shRNA) MUC2-silenced cells. Activation of cyclic adenylyl cyclase (AMP) and mitogen-activated protein kinase (MAPK) signaling pathways was necessary for the simultaneous expression of MUC2 and cathelicidins. In Muc2 mucin-deficient (Muc2-/-) mice, murine cathelicidin (Cramp) was significantly reduced compared to that in Muc2+/- and Muc2+/+ littermates. E. histolytica-induced acute inflammation in colonic loops stimulated high levels of cathelicidin in Muc2+/+ but not in Muc2-/- littermates. In dextran sodium sulfate (DSS)-induced colitis in Muc2+/+ mice, which depletes the mucus barrier and goblet cell mucin, Cramp expression was significantly enhanced during restitution. These studies demonstrate regulatory mechanisms between MUC2 and cathelicidins in the colonic mucosa where an intact mucus barrier is essential for expression and secretion of cathelicidins in response to E. histolytica- and DSS-induced colitis.
Collapse
|
44
|
Meyer M, Fehling H, Matthiesen J, Lorenzen S, Schuldt K, Bernin H, Zaruba M, Lender C, Ernst T, Ittrich H, Roeder T, Tannich E, Lotter H, Bruchhaus I. Overexpression of Differentially Expressed Genes Identified in Non-pathogenic and Pathogenic Entamoeba histolytica Clones Allow Identification of New Pathogenicity Factors Involved in Amoebic Liver Abscess Formation. PLoS Pathog 2016; 12:e1005853. [PMID: 27575775 PMCID: PMC5004846 DOI: 10.1371/journal.ppat.1005853] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/09/2016] [Indexed: 02/07/2023] Open
Abstract
We here compared pathogenic (p) and non-pathogenic (np) isolates of Entamoeba histolytica to identify molecules involved in the ability of this parasite to induce amoebic liver abscess (ALA)-like lesions in two rodent models for the disease. We performed a comprehensive analysis of 12 clones (A1–A12) derived from a non-pathogenic isolate HM-1:IMSS-A and 12 clones (B1–B12) derived from a pathogenic isolate HM-1:IMSS-B. “Non-pathogenicity” included the induction of small and quickly resolved lesions while “pathogenicity” comprised larger abscess development that overstayed day 7 post infection. All A-clones were designated as non-pathogenic, whereas 4 out of 12 B-clones lost their ability to induce ALAs in gerbils. No correlation between ALA formation and cysteine peptidase (CP) activity, haemolytic activity, erythrophagocytosis, motility or cytopathic activity was found. To identify the molecular framework underlying different pathogenic phenotypes, three clones were selected for in-depth transcriptome analyses. Comparison of a non-pathogenic clone A1np with pathogenic clone B2p revealed 76 differentially expressed genes, whereas comparison of a non-pathogenic clone B8np with B2p revealed only 19 differentially expressed genes. Only six genes were found to be similarly regulated in the two non-pathogenic clones A1np and B8np in comparison with the pathogenic clone B2p. Based on these analyses, we chose 20 candidate genes and evaluated their roles in ALA formation using the respective gene-overexpressing transfectants. We conclude that different mechanisms lead to loss of pathogenicity. In total, we identified eight proteins, comprising a metallopeptidase, C2 domain proteins, alcohol dehydrogenases and hypothetical proteins, that affect the pathogenicity of E. histolytica. The pathogen Entamoeba histolytica can live asymptomatically in the human gut, or it can disrupt the intestinal barrier and induce life-threatening abscesses in different organs, most often in the liver. The molecular framework that enables this invasive, highly pathogenic phenotype is still not well understood. In order to identify factors that are positively or negatively correlated for invasion and destruction of the liver, we used a unique tool, E. histolytica clones that differ dramatically in their pathogenicity, while sharing almost identical genetic background. Based on comprehensive transcriptome studies of these clones, we identified a set of candidate genes that are potentially involved in pathogenicity. Using ectopic overexpression of the most promising candidates, either in pathogenic or in non-pathogenic Entamoeba clones, we identified genes where high expression reduced pathogenicity and only one gene that increased pathogenicity to a certain extend. Taken together, the current study identifies novel pathogenicity factors of E. histolytica and highlights the observation that various different genes contribute to pathogenicity.
Collapse
Affiliation(s)
- Martin Meyer
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Helena Fehling
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jenny Matthiesen
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stephan Lorenzen
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Kathrin Schuldt
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Hannah Bernin
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Mareen Zaruba
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Corinna Lender
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Thomas Ernst
- Diagnostic and Interventional Radiology Department and Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Harald Ittrich
- Diagnostic and Interventional Radiology Department and Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Roeder
- Zoological Institute, Molecular Physiology, Christian-Albrechts University Kiel, Kiel, Germany
| | - Egbert Tannich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Hannelore Lotter
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Iris Bruchhaus
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|