1
|
Horn M, Bieliková L, Vostoupalová A, Švéda J, Mareš M. An update on proteases and protease inhibitors from trematodes. ADVANCES IN PARASITOLOGY 2024; 126:97-176. [PMID: 39448195 DOI: 10.1016/bs.apar.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Trematodes, a class of parasitic flatworms, are responsible for a variety of devastating diseases in humans and animals, with schistosomiasis and fascioliasis being prominent examples. Trematode proteolytic systems involved in the host-parasite interaction have emerged as key contributors to the success of trematodes in establishing and maintaining infections. This review concentrates on diverse proteases and protease inhibitors employed by trematodes and provides an update on recent advances in their molecular-level characterization, with a focus on function, structure, and therapeutic target potential.
Collapse
Affiliation(s)
- Martin Horn
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lucia Bieliková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Vostoupalová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jakub Švéda
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michael Mareš
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
2
|
Stark KA, Rinaldi G, Costain A, Clare S, Tolley C, Almeida A, McCarthy C, Harcourt K, Brandt C, Lawley TD, Berriman M, MacDonald AS, Forde-Thomas JE, Hulme BJ, Hoffmann KF, Cantacessi C, Cortés A. Gut microbiota and immune profiling of microbiota-humanised versus wildtype mouse models of hepatointestinal schistosomiasis. Anim Microbiome 2024; 6:36. [PMID: 38918824 PMCID: PMC11201864 DOI: 10.1186/s42523-024-00318-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Mounting evidence of the occurrence of direct and indirect interactions between the human blood fluke, Schistosoma mansoni, and the gut microbiota of rodent models raises questions on the potential role(s) of the latter in the pathophysiology of hepatointestinal schistosomiasis. However, substantial differences in both the composition and function between the gut microbiota of laboratory rodents and that of humans hinders an in-depth understanding of the significance of such interactions for human schistosomiasis. Taking advantage of the availability of a human microbiota-associated mouse model (HMA), we have previously highlighted differences in infection-associated changes in gut microbiota composition between HMA and wildtype (WT) mice. To further explore the dynamics of schistosome-microbiota relationships in HMA mice, in this study we (i) characterize qualitative and quantitative changes in gut microbiota composition of a distinct line of HMA mice (D2 HMA) infected with S. mansoni prior to and following the onset of parasite egg production; (ii) profile local and systemic immune responses against the parasite in HMA as well as WT mice and (iii) assess levels of faecal inflammatory markers and occult blood as indirect measures of gut tissue damage. We show that patent S. mansoni infection is associated with reduced bacterial alpha diversity in the gut of D2 HMA mice, alongside expansion of hydrogen sulphide-producing bacteria. Similar systemic humoral responses against S. mansoni in WT and D2 HMA mice, as well as levels of faecal lipocalin and markers of alternatively activated macrophages, suggest that these are independent of baseline gut microbiota composition. Qualitative comparative analyses between faecal microbial profiles of S. mansoni-infected WT and distinct lines of HMA mice reveal that, while infection-induced alterations of the gut microbiota composition are highly dependent on the baseline flora, bile acid composition and metabolism may represent key elements of schistosome-microbiota interactions through the gut-liver axis.
Collapse
Affiliation(s)
- K A Stark
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - G Rinaldi
- Department of Life Sciences, Aberystwyth University, Aberystwyth, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - A Costain
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - S Clare
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - C Tolley
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - A Almeida
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - C McCarthy
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - K Harcourt
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - C Brandt
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - T D Lawley
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - M Berriman
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - A S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - J E Forde-Thomas
- Department of Life Sciences, Aberystwyth University, Aberystwyth, UK
| | - B J Hulme
- Department of Life Sciences, Aberystwyth University, Aberystwyth, UK
| | - K F Hoffmann
- Department of Life Sciences, Aberystwyth University, Aberystwyth, UK
| | - C Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| | - A Cortés
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Valencia, Spain
| |
Collapse
|
3
|
Perera DJ, Koger-Pease C, Paulini K, Daoudi M, Ndao M. Beyond schistosomiasis: unraveling co-infections and altered immunity. Clin Microbiol Rev 2024; 37:e0009823. [PMID: 38319102 PMCID: PMC10938899 DOI: 10.1128/cmr.00098-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by the helminth Schistosoma spp. and has the second highest global impact of all parasites. Schistosoma are transmitted through contact with contaminated fresh water predominantly in Africa, Asia, the Middle East, and South America. Due to the widespread prevalence of Schistosoma, co-infection with other infectious agents is common but often poorly described. Herein, we review recent literature describing the impact of Schistosoma co-infection between species and Schistosoma co-infection with blood-borne protozoa, soil-transmitted helminths, various intestinal protozoa, Mycobacterium, Salmonella, various urinary tract infection-causing agents, and viral pathogens. In each case, disease severity and, of particular interest, the immune landscape, are altered as a consequence of co-infection. Understanding the impact of schistosomiasis co-infections will be important when considering treatment strategies and vaccine development moving forward.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Cal Koger-Pease
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kayla Paulini
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Mohamed Daoudi
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
4
|
Tang CL, Lian Z, Ding FR, Liang J, Li XY. Schistosoma-related molecules as a new strategy to combat type 1 diabetes through immune regulation. Parasitol Int 2024; 98:102818. [PMID: 37848126 DOI: 10.1016/j.parint.2023.102818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/08/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023]
Abstract
The study of immune regulation mechanisms induced by parasites may help develop new treatment methods for inflammatory diseases including type 1 diabetes, which is related to type 1 immune responses. The negative correlation between schistosomiasis infection and type 1 diabetes has been confirmed, and the mechanism of Schistosoma-mediated prevention of type 1 diabetes may be related to the adaptive and innate immune systems. Schistosoma-related molecules affect immune cell composition and macrophage polarization and stimulate an increase in natural killer T cells. Furthermore, Schistosoma-related molecules can regulate the adaptive immune responses related to the prevention of type 1 diabetes and change the Th1/Th2 and Th17/Treg axis. Our previous review showed the role of regulatory T cells in the protective of type 1 diabetes mediated by Schistosoma. Here, we aim to review the other mechanisms of schistosomiasis infection and Schistosoma-related products in regulating the immune response associated with the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital, Wuhan University of Science and Technology, Wuhan 430063, China
| | - Zhan Lian
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan 430030, China
| | - Fan-Rong Ding
- Wuchang Hospital, Wuhan University of Science and Technology, Wuhan 430063, China
| | - Jun Liang
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan 430030, China.
| | - Xiang-You Li
- Wuchang Hospital, Wuhan University of Science and Technology, Wuhan 430063, China.
| |
Collapse
|
5
|
Perera DJ, Domenech P, Babuadze GG, Naghibosadat M, Alvarez F, Koger-Pease C, Labrie L, Stuible M, Durocher Y, Piccirillo CA, Lametti A, Fiset PO, Elahi SM, Kobinger GP, Gilbert R, Olivier M, Kozak R, Reed MB, Ndao M. BCG administration promotes the long-term protection afforded by a single-dose intranasal adenovirus-based SARS-CoV-2 vaccine. iScience 2023; 26:107612. [PMID: 37670783 PMCID: PMC10475483 DOI: 10.1016/j.isci.2023.107612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/19/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
Recent publications have explored intranasal (i.n.) adenovirus-based (Ad) vaccines as an effective strategy for SARS-CoV-2 in pre-clinical models. However, the effects of prior immunizations and infections have yet to be considered. Here, we investigate the immunomodulatory effects of Mycobacterium bovis BCG pre-immunization followed by vaccination with an S-protein-expressing i.n. Ad, termed Ad(Spike). While i.n. Ad(Spike) retains some protective effect after 6 months, a single administration of BCG-Danish prior to Ad(Spike) potentiates its ability to control viral replication of the B.1.351 SARS-CoV-2 variant within the respiratory tract. Though BCG-Danish did not affect Ad(Spike)-generated humoral immunity, it promoted the generation of cytotoxic/Th1 responses over suppressive FoxP3+ TREG cells in the lungs of infected mice. Thus, this vaccination strategy may prove useful in limiting future pandemics by potentiating the long-term efficacy of mucosal vaccines within the context of the widely distributed BCG vaccine.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Pilar Domenech
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- McGill International TB Centre, McGill University, Montréal, QC, Canada
| | - George Giorgi Babuadze
- Department of Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Maedeh Naghibosadat
- Department of Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Fernando Alvarez
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Cal Koger-Pease
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Lydia Labrie
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Matthew Stuible
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Yves Durocher
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - André Lametti
- Department of Pathology, McGill University, Montréal, QC, Canada
| | | | - Seyyed Mehdy Elahi
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Gary P. Kobinger
- Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Rénald Gilbert
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, QC, Canada
| | - Martin Olivier
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Robert Kozak
- Department of Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Molecular Diagnostics, Division of Microbiology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Michael B. Reed
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- McGill International TB Centre, McGill University, Montréal, QC, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- National Reference Centre for Parasitology, McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
6
|
Stark KA, Rinaldi G, Cortés A, Costain A, MacDonald AS, Cantacessi C. The role of the host gut microbiome in the pathophysiology of schistosomiasis. Parasite Immunol 2023; 45:e12970. [PMID: 36655799 DOI: 10.1111/pim.12970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/06/2023] [Accepted: 01/15/2023] [Indexed: 01/20/2023]
Abstract
The pathophysiology of schistosomiasis is linked to the formation of fibrous granulomas around eggs that become trapped in host tissues, particularly the intestines and liver, during their migration to reach the lumen of the vertebrate gut. While the development of Schistosoma egg-induced granulomas is the result of finely regulated crosstalk between egg-secreted antigens and host immunity, evidence has started to emerge of the likely contribution of an additional player-the host gut microbiota-to pathological processes that culminate with the formation of these tissue lesions. Uncovering the role(s) of schistosome-mediated changes in gut microbiome composition and function in granuloma formation and, more broadly, in the pathophysiology of schistosomiasis, will shed light on the mechanisms underlying this three-way parasite-host-microbiome interplay. Such knowledge may, in turn, pave the way towards the discovery of novel therapeutic targets and control strategies.
Collapse
Affiliation(s)
- Klara A Stark
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Gabriel Rinaldi
- Department of Life Sciences, Edward Llwyd Building, Aberystwyth University, Aberystwyth, UK
| | - Alba Cortés
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, València, Spain
| | - Alice Costain
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Salmonella Typhimurium expressing chromosomally integrated Schistosoma mansoni Cathepsin B protects against schistosomiasis in mice. NPJ Vaccines 2023; 8:27. [PMID: 36849453 PMCID: PMC9969381 DOI: 10.1038/s41541-023-00599-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/13/2023] [Indexed: 03/01/2023] Open
Abstract
Schistosomiasis threatens hundreds of millions of people worldwide. The larval stage of Schistosoma mansoni migrates through the lung and adult worms reside adjacent to the colonic mucosa. Several candidate vaccines are in preclinical development, but none is designed to elicit both systemic and mucosal responses. We have repurposed an attenuated Salmonella enterica Typhimurium strain (YS1646) to express Cathepsin B (CatB), a digestive enzyme important for the juvenile and adult stages of the S. mansoni life cycle. Previous studies have demonstrated the prophylactic and therapeutic efficacy of our plasmid-based vaccine. Here, we have generated chromosomally integrated (CI) YS1646 strains that express CatB to produce a viable candidate vaccine for eventual human use (stability, no antibiotic resistance). 6-8-week-old C57BL/6 mice were vaccinated in a multimodal oral (PO) and intramuscular (IM) regimen, and then sacrificed 3 weeks later. The PO + IM group had significantly higher anti-CatB IgG titers with greater avidity and mounted significant intestinal anti-CatB IgA responses compared to PBS control mice (all P < 0.0001). Multimodal vaccination generated balanced TH1/TH2 humoral and cellular immune responses. Production of IFNγ by both CD4+ and CD8+ T cells was confirmed by flow cytometry (P < 0.0001 & P < 0.01). Multimodal vaccination reduced worm burden by 80.4%, hepatic egg counts by 75.2%, and intestinal egg burden by 78.4% (all P < 0.0001). A stable and safe vaccine that has both prophylactic and therapeutic activity would be ideal for use in conjunction with praziquantel mass treatment campaigns.
Collapse
|
8
|
Recent Advances in the Development of Adenovirus-Vectored Vaccines for Parasitic Infections. Pharmaceuticals (Basel) 2023; 16:ph16030334. [PMID: 36986434 PMCID: PMC10058461 DOI: 10.3390/ph16030334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Vaccines against parasites have lagged centuries behind those against viral and bacterial infections, despite the devastating morbidity and widespread effects of parasitic diseases across the globe. One of the greatest hurdles to parasite vaccine development has been the lack of vaccine strategies able to elicit the complex and multifaceted immune responses needed to abrogate parasitic persistence. Viral vectors, especially adenovirus (AdV) vectors, have emerged as a potential solution for complex disease targets, including HIV, tuberculosis, and parasitic diseases, to name a few. AdVs are highly immunogenic and are uniquely able to drive CD8+ T cell responses, which are known to be correlates of immunity in infections with most protozoan and some helminthic parasites. This review presents recent developments in AdV-vectored vaccines targeting five major human parasitic diseases: malaria, Chagas disease, schistosomiasis, leishmaniasis, and toxoplasmosis. Many AdV-vectored vaccines have been developed for these diseases, utilizing a wide variety of vectors, antigens, and modes of delivery. AdV-vectored vaccines are a promising approach for the historically challenging target of human parasitic diseases.
Collapse
|
9
|
Ahmad G. Schistosomiasis: Basic requirements for the development of a subunit vaccine, using genetic vectors. EBioMedicine 2022; 82:104162. [PMID: 35841870 PMCID: PMC9297073 DOI: 10.1016/j.ebiom.2022.104162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 10/31/2022] Open
|