1
|
Cao M, Wang R, Xu X, Hou X, Wang W, Zhang X, Ma C, Zhang Y, Shi D, Yang J, Ma H. Engineering of peptide assemblies for adaptable protein delivery to achieve efficient intracellular biocatalysis. J Colloid Interface Sci 2024; 683:457-467. [PMID: 39693883 DOI: 10.1016/j.jcis.2024.12.097] [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: 09/23/2024] [Revised: 12/11/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024]
Abstract
Efficient intracellular delivery of native proteins remains a big challenge, which greatly hinders the development of protein therapy. Here, we report a generalizable peptide vector that can encapsulate and deliver various proteins to achieve efficient intracellular biocatalysis. The peptide was rationally designed to be cationic amphiphilic peptide that consist of four functional fragments, that is, a hydrophobic domain to promote molecular assembly, an enzyme-cleavable fragment to introduce stimuli-responsibility, several cationic arginine (Arg) residues to enhance cell interaction and transmembrane efficiency, and the cystine (Cys) residues with redox sensitivity to adjust the stability of the peptide/protein complexes as needed. The peptide can co-assemble with proteins to form stable complexes in aqueous solution under mild condition. The complexes enter cell mainly through caveolae- and lipid raft-mediated endocytosis, giving a delivery efficiency of up to ∼97.2 %. They can then achieve efficient lysosomal escape and disassociation to release native proteins inside cells in response to intracellular stimuli. More strikingly, the delivered protein's bioactivity can be well maintained and the two model proteins of β-galactosidase (β-Gal) and horseradish peroxidase (HRP) both showed excellent intracellular biocatalytic activity. The study develops a versatile and adjustable peptide carrier platform for protein delivery and highlights impactful structure-function relationships, providing a new chemical guide for the design and optimization of functional protein nanocarriers.
Collapse
Affiliation(s)
- Meiwen Cao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China.
| | - Rui Wang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Xiaomin Xu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Xinyue Hou
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Wentao Wang
- Department of Radiochemistry, China Institute of Atomic Energy, Beijing 102413, China.
| | - Xiaoming Zhang
- School of Science, Optoelectronics Research Center, Minzu University of China, Beijing 100081, China
| | - Chen Ma
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Yuxuan Zhang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Daikui Shi
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Jianing Yang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Hongchao Ma
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| |
Collapse
|
2
|
Kaya B, Smith H, Chen Y, Azad MG, M Russell T, Richardson V, Bernhardt PV, Dharmasivam M, Richardson DR. Targeting lysosomes by design: novel N-acridine thiosemicarbazones that enable direct detection of intracellular drug localization and overcome P-glycoprotein (Pgp)-mediated resistance. Chem Sci 2024:d4sc04339a. [PMID: 39165729 PMCID: PMC11331336 DOI: 10.1039/d4sc04339a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 08/22/2024] Open
Abstract
Innovative N-acridine thiosemicarbazones (NATs) were designed along with their iron(iii), copper(ii), and zinc(ii) complexes. Lysosomal targeting was promoted by specifically incorporating the lysosomotropic Pgp substrate, acridine, into the thiosemicarbazone scaffold to maintain the tridentate N, N, S-donor system. The acridine moiety enables a significant advance in thiosemicarbazone design, since: (1) it enables tracking of the drugs by confocal microscopy using its inherent fluorescence; (2) it is lysosomotropic enabling lysosomal targeting; and (3) as acridine is a P-glycoprotein (Pgp) substrate, it facilitates lysosomal targeting, resulting in the drug overcoming Pgp-mediated resistance. These new N-acridine analogues are novel, and this is the first time that acridine has been specifically added to the thiosemicarbazone framework to achieve the three important properties above. These new agents displayed markedly greater anti-proliferative activity against resistant Pgp-expressing cells than very low Pgp-expressing cells. The anti-proliferative activity of NATs against multiple Pgp-positive cancer cell-types (colon, lung, and cervical carcinoma) was abrogated by the third generation Pgp inhibitor, Elacridar, and also Pgp siRNA that down-regulated Pgp. Confocal microscopy demonstrated that low Pgp in KB31 (-Pgp) cells resulted in acridine's proclivity for DNA intercalation promoting NAT nuclear-targeting. In contrast, high Pgp in KBV1 (+Pgp) cells led to NAT lysosomal sequestration, preventing its nuclear localisation. High Pgp expression in KBV1 (+Pgp) cells resulted in co-localization of NATs with the lysosomal marker, LysoTracker™, that was significantly (p < 0.001) greater than the positive control, the di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) Zn(ii) complex, [Zn(DpC)2]. Incorporation of acridine into the thiosemicarbazone scaffold led to Pgp-mediated transport into lysosomes to overcome Pgp-resistance.
Collapse
Affiliation(s)
- Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Henry Smith
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Yanbing Chen
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland Brisbane 4072 Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine Nagoya 466-8550 Japan
| |
Collapse
|
3
|
Qadeer A, Wajid A, Rafey HA, Nawaz S, Khan S, Rahman SU, Alzahrani KJ, Khan MZ, Alsabi MNS, Ullah H, Safi SZ, Xia Z, Zahoor M. Exploring extracellular vesicles in zoonotic helminth biology: implications for diagnosis, therapeutic and delivery. Front Cell Infect Microbiol 2024; 14:1424838. [PMID: 39165921 PMCID: PMC11333462 DOI: 10.3389/fcimb.2024.1424838] [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: 04/28/2024] [Accepted: 07/03/2024] [Indexed: 08/22/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as key intercellular communication and pathogenesis mediators. Parasitic organisms' helminths, cause widespread infections with significant health impacts worldwide. Recent research has shed light on the role of EVs in the lifecycle, immune evasion, and disease progression of these parasitic organisms. These tiny membrane-bound organelles including microvesicles and exosomes, facilitate the transfer of proteins, lipids, mRNAs, and microRNAs between cells. EVs have been isolated from various bodily fluids, offering a potential diagnostic and therapeutic avenue for combating infectious agents. According to recent research, EVs from helminths hold great promise in the diagnosis of parasitic infections due to their specificity, early detection capabilities, accessibility, and the potential for staging and monitoring infections, promote intercellular communication, and are a viable therapeutic tool for the treatment of infectious agents. Exploring host-parasite interactions has identified promising new targets for diagnostic, therapy, and vaccine development against helminths. This literature review delves into EVS's origin, nature, biogenesis, and composition in these parasitic organisms. It also highlights the proteins and miRNAs involved in EV release, providing a comprehensive summary of the latest findings on the significance of EVs in the biology of helminths, promising targets for therapeutic and diagnostic biomarkers.
Collapse
Affiliation(s)
- Abdul Qadeer
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Abdul Wajid
- Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | - Hafiz Abdul Rafey
- Shifa College of Pharmaceutical Sciences, Faculty of Pharmaceutical and Allied Health Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Saqib Nawaz
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Sawar Khan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Sajid Ur Rahman
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Khalid J. Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Muhammad Zahoor Khan
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng, Shandong, China
| | - Mohammad Nafi Solaiman Alsabi
- Department of Basic Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Hanif Ullah
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu, China
| | - Sher Zaman Safi
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, Selangor, Malaysia
| | - Zanxian Xia
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Muhammad Zahoor
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
Tanabe MB, Caravedo MA, Clinton White A, Cabada MM. An Update on the Pathogenesis of Fascioliasis: What Do We Know? Res Rep Trop Med 2024; 15:13-24. [PMID: 38371362 PMCID: PMC10874186 DOI: 10.2147/rrtm.s397138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/27/2024] [Indexed: 02/20/2024] Open
Abstract
Fasciola hepatica is a trematode parasite distributed worldwide. It is known to cause disease in mammals, producing significant economic loses to livestock industry and burden to human health. After ingestion, the parasites migrate through the liver and mature in the bile ducts. A better understanding of the parasite's immunopathogenesis would help to develop efficacious therapeutics and vaccines. Currently, much of our knowledge comes from in vitro and in vivo studies in animal models. Relatively little is known about the host-parasite interactions in humans. Here, we provide a narrative review of what is currently know about the pathogenesis and host immune responses to F. hepatica summarizing the evidence available from the multiple hosts that this parasite infects.
Collapse
Affiliation(s)
- Melinda B Tanabe
- Division of Infectious Disease, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Maria A Caravedo
- Division of Infectious Disease, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - A Clinton White
- Division of Infectious Disease, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
- Cusco Branch – Alexander von Humboldt Tropical Medicine Institute, Universidad Peruana Cayetano Heredia, Cusco, Peru
| | - Miguel M Cabada
- Division of Infectious Disease, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
- Cusco Branch – Alexander von Humboldt Tropical Medicine Institute, Universidad Peruana Cayetano Heredia, Cusco, Peru
| |
Collapse
|
5
|
Chakraborty P, Aravindhan V, Mukherjee S. Helminth-derived biomacromolecules as therapeutic agents for treating inflammatory and infectious diseases: What lessons do we get from recent findings? Int J Biol Macromol 2023; 241:124649. [PMID: 37119907 DOI: 10.1016/j.ijbiomac.2023.124649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Despite the tremendous progress in healthcare sectors, a number of life-threatening infectious, inflammatory, and autoimmune diseases are continuously challenging mankind throughout the globe. In this context, recent successes in utilizing helminth parasite-derived bioactive macromolecules viz. glycoproteins, enzymes, polysaccharides, lipids/lipoproteins, nucleic acids/nucleotides, and small organic molecules for treating various disorders primarily resulted from inflammation. Among the several parasites that infect humans, helminths (cestodes, nematodes, and trematodes) are known as efficient immune manipulators owing to their explicit ability to modulate and modify the innate and adaptive immune responses of humans. These molecules selectively bind to immune receptors on innate and adaptive immune cells and trigger multiple signaling pathways to elicit anti-inflammatory cytokines, expansion of alternatively activated macrophages, T-helper 2, and immunoregulatory T regulatory cell types to induce an anti-inflammatory milieu. Reduction of pro-inflammatory responses and repair of tissue damage by these anti-inflammatory mediators have been exploited for treating a number of autoimmune, allergic, and metabolic diseases. Herein, the potential and promises of different helminths/helminth-derived products as therapeutic agents in ameliorating immunopathology of different human diseases and their mechanistic insights of function at cell and molecular level alongside the molecular signaling cross-talks have been reviewed by incorporating up-to-date findings achieved in the field.
Collapse
Affiliation(s)
- Pritha Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India
| | | | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India.
| |
Collapse
|
6
|
Quinteros SL, von Krusenstiern E, Snyder NW, Tanaka A, O’Brien B, Donnelly S. The helminth derived peptide FhHDM-1 redirects macrophage metabolism towards glutaminolysis to regulate the pro-inflammatory response. Front Immunol 2023; 14:1018076. [PMID: 36761766 PMCID: PMC9905698 DOI: 10.3389/fimmu.2023.1018076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
We have previously identified an immune modulating peptide, termed FhHDM-1, within the secretions of the liver fluke, Fasciola hepatica, which is sufficiently potent to prevent the progression of type 1 diabetes and multiple sclerosis in murine models of disease. Here, we have determined that the FhHDM-1 peptide regulates inflammation by reprogramming macrophage metabolism. Specifically, FhHDM-1 switched macrophage metabolism to a dependence on oxidative phosphorylation fuelled by fatty acids and supported by the induction of glutaminolysis. The catabolism of glutamine also resulted in an accumulation of alpha ketoglutarate (α-KG). These changes in metabolic activity were associated with a concomitant reduction in glycolytic flux, and the subsequent decrease in TNF and IL-6 production at the protein level. Interestingly, FhHDM-1 treated macrophages did not express the characteristic genes of an M2 phenotype, thereby indicating the specific regulation of inflammation, as opposed to the induction of an anti-inflammatory phenotype per se. Use of an inactive derivative of FhHDM-1, which did not modulate macrophage responses, revealed that the regulation of immune responses was dependent on the ability of FhHDM-1 to modulate lysosomal pH. These results identify a novel functional association between the lysosome and mitochondrial metabolism in macrophages, and further highlight the significant therapeutic potential of FhHDM-1 to prevent inflammation.
Collapse
Affiliation(s)
- Susel Loli Quinteros
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | | | - Nathaniel W. Snyder
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Akane Tanaka
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Bronwyn O’Brien
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia,*Correspondence: Sheila Donnelly,
| |
Collapse
|
7
|
Wound healing approach based on excretory-secretory product and lysate of liver flukes. Sci Rep 2022; 12:21639. [PMID: 36517588 PMCID: PMC9751068 DOI: 10.1038/s41598-022-26275-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Exogenous bioactive peptides are considered promising for the wound healing therapy in humans. In this regard, parasitic trematodes proteins may potentially become a new perspective agents. Foodborne trematode Opisthorchis felineus is widespread in Europe and has the ability to stimulate proliferation of bile duct epithelium. In this study, we investigated skin wound healing potential of O. felineus proteins in mouse model. C57Bl/6 mice were inflicted with superficial wounds with 8 mm diameter. Experimental groups included several non-specific controls and specific treatment groups (excretory-secretory product and lysate). After 10 days of the experiment, the percentage of wound healing in the specific treatment groups significantly exceeded the control values. We also found that wound treatment with excretory-secretory product and worm lysate resulted in: (i) inflammation reducing, (ii) vascular response modulating, (iii) type 1 collagen deposition promoting dermal ECM remodeling. An additional proteomic analysis of excretory-secretory product and worm lysate samples was revealed 111 common proteins. The obtained data indicate a high wound-healing potential of liver fluke proteins and open prospects for further research as new therapeutic approaches.
Collapse
|
8
|
Dalmolin SP, Pedó RT, da Rosa TH, de Souza Silva JM, Farinon M, Gasparini ML, Chiela ECF, Paz AH, Sehabiague MPC, Ferreira HB, do Espírito Santo RC, da Costa Gonçalves F, Xavier RM. Fasciola hepatica extract suppresses fibroblast-like synoviocytes in vitro and alleviates experimental arthritis. Adv Rheumatol 2022; 62:43. [PMID: 36371346 DOI: 10.1186/s42358-022-00275-y] [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: 05/16/2022] [Accepted: 10/29/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovial inflammation, fibroblast-like synoviocytes (FLS) activation and joint destruction. Fasciola hepatica is a platyhelminth that releases excretory-secretory immunomodulatory products capable of suppressing the Th1 immune response. Despite the effectiveness of available treatments for inducing disease remission, current options are not successful in all patients and may cause side effects. Thus, we evaluated the therapeutic potential of F. hepatica extract on FLS from RA patients and arthritis models. METHODS FLS were isolated from synovial fluid of RA patients, cultured, and exposed to F. hepatica extract (60, 80, and 100 µg/ml) for different time points to assess cell viability, adherence, migration and invasion. For in vivo experiments, mice with antigen (AIA) and collagen (CIA) induced arthritis received a 200 µg/dose of F. hepatica extract daily. Statistical analysis was performed by ANOVA and Student's t-test using GraphPad Prism 6.0. RESULTS In vitro assays showed that extract decreased FLS cell viability at concentration of 100 µg/ml (83.8% ± 5.0 extract vs. 100.0% ± 0.0 control; p < 0.05), adherence in 20% (92.0 cells ± 5.8 extract vs. 116.3 cells ± 7.9 control; p < 0.05), migratory potential (69.5% ± 17.6 extract vs. 100.0% control; p < 0.05), and cell invasiveness potential through the matrigel (76.0% ± 8.4 extract vs. 100.0% control; p < 0.01). The extract reduced leukocyte migration by 56% (40 × 104 leukocytes/knee ± 19.00) compared to control (90.90 × 104 leukocytes/knee ± 12.90) (p < 0.01) and nociception (6.37 g ± 0.99 extract vs. 3.81 g ± 1.44 control; p < 0.001) in AIA and delayed clinical onset of CIA (11.75 ± 2.96 extract vs. 14.00 ± 2.56 control; p = 0.126). CONCLUSION Our results point out a potential immunomodulatory effect of F. hepatica extract in RA models. Therefore, the characterization of promising new immunomodulatory molecules should be pursued, as they can promote the development of new therapies. Trial registration Collection of synovial liquid and in vitro procedures were approved by the Ethics Committee with Certificate of Presentation of Ethical Appreciation in Plataforma Brasil (CAAE: 89044918.8.0000.5327; date of registration: 26/07/2018).
Collapse
Affiliation(s)
- Suelen Pizzolatto Dalmolin
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Renata Ternus Pedó
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Thales Hein da Rosa
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jordana Miranda de Souza Silva
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mirian Farinon
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria Luísa Gasparini
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo Cremonese Filippi Chiela
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Serviço de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Centro de Pesquisas Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ana Helena Paz
- Programa de Pós-Graduação Ciências em Gastroenterologia e Hepatologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Serviço de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Centro de Pesquisas Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Martín Pablo Cancela Sehabiague
- Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Henrique Bunselmeyer Ferreira
- Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafaela Cavalheiro do Espírito Santo
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fabiany da Costa Gonçalves
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, GD 3015, Rotterdam, The Netherlands.
| | - Ricardo Machado Xavier
- Laboratório de Doenças Autoimunes, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil. .,Programa de Pós-Graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350; 6º Andar, Porto Alegre, RS, 90035-903, Brazil.
| |
Collapse
|
9
|
Carson JP, Robinson MW, Ramm GA, Gobert GN. Synthetic peptides derived from the Schistosoma mansoni secretory protein Sm16 induce contrasting responses in hepatic stellate cells. Exp Parasitol 2022; 236-237:108255. [PMID: 35385714 DOI: 10.1016/j.exppara.2022.108255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/04/2022]
|
10
|
Choezom D, Gross JC. Neutral Sphingomyelinase 2 controls exosomes secretion via counteracting V-ATPase-mediated endosome acidification. J Cell Sci 2022; 135:274565. [PMID: 35050379 PMCID: PMC8919340 DOI: 10.1242/jcs.259324] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/29/2021] [Indexed: 11/25/2022] Open
Abstract
During endosome maturation, neutral sphingomyelinase 2 (nSMase2, encoded by SMPD3) is involved in budding of intraluminal vesicles (ILVs) into late endosomes or multivesicular bodies (MVBs). Fusion of these with the plasma membrane results in secretion of exosomes or small extracellular vesicles (sEVs). Here, we report that nSMase2 activity controls sEV secretion through modulation of vacuolar H+-ATPase (V-ATPase) activity. Specifically, we show that nSMase2 inhibition induces V-ATPase complex assembly that drives MVB lumen acidification and consequently reduces sEV secretion. Conversely, we further demonstrate that stimulating nSMase2 activity with the inflammatory cytokine TNFα (also known as TNF) decreases acidification and increases sEV secretion. Thus, we find that nSMase2 activity affects MVB membrane lipid composition to counteract V-ATPase-mediated endosome acidification, thereby shifting MVB fate towards sEV secretion. This article has an associated First Person interview with the first author of the paper. Summary: Changing neutral sphingomyelinase 2 activity regulates small extracellular vesicle secretion through modulation of V-ATPase activity.
Collapse
Affiliation(s)
- Dolma Choezom
- Developmental Biochemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Julia Christina Gross
- Developmental Biochemistry, University Medical Center Goettingen, Goettingen, Germany
- Hematology and Oncology, University Medical Center Goettingen, Goettingen, Germany
- Health and Medical University Potsdam, Potsdam, Germany
| |
Collapse
|
11
|
Shi W, Xu N, Wang X, Vallée I, Liu M, Liu X. Helminth Therapy for Immune-Mediated Inflammatory Diseases: Current and Future Perspectives. J Inflamm Res 2022; 15:475-491. [PMID: 35087284 PMCID: PMC8789313 DOI: 10.2147/jir.s348079] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Affiliation(s)
- Wenjie Shi
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Ning Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Xuelin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Isabelle Vallée
- UMR BIPAR, Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
- Correspondence: Xiaolei Liu; Mingyuan Liu, Tel +86-15943092280; +86-13019125996, Email ;
| |
Collapse
|
12
|
Camaya I, Mok TY, Lund M, To J, Braidy N, Robinson MW, Santos J, O'Brien B, Donnelly S. The parasite-derived peptide FhHDM-1 activates the PI3K/Akt pathway to prevent cytokine-induced apoptosis of β-cells. J Mol Med (Berl) 2021; 99:1605-1621. [PMID: 34374810 DOI: 10.1007/s00109-021-02122-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/13/2021] [Accepted: 07/27/2021] [Indexed: 12/31/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterised by the destruction of the insulin-producing beta (β)-cells within the pancreatic islets. We have previously identified a novel parasite-derived molecule, termed Fasciola hepatica helminth defence molecule 1 (FhHDM-1), that prevents T1D development in non-obese diabetic (NOD) mice. In this study, proteomic analyses of pancreas tissue from NOD mice suggested that FhHDM-1 activated the PI3K/Akt signalling pathway, which is associated with β-cell metabolism, survival and proliferation. Consistent with this finding, FhHDM-1 preserved β-cell mass in NOD mice. Examination of the biodistribution of FhHDM-1 after intraperitoneal administration in NOD mice revealed that the parasite peptide localised to the pancreas, suggesting that it exerted a direct effect on the survival/function of β-cells. This was confirmed in vitro, as the interaction of FhHDM-1 with the NOD-derived β-cell line, NIT-1, resulted in increased levels of phosphorylated Akt, increased NADH and NADPH and reduced activity of the NAD-dependent DNA nick sensor, poly(ADP-ribose) polymerase (PARP-1). As a consequence, β-cell survival was enhanced and apoptosis was prevented in the presence of the pro-inflammatory cytokines that destroy β-cells during T1D pathogenesis. Similarly, FhHDM-1 protected primary human islets from cytokine-induced apoptosis. Importantly, while FhHDM-1 promoted β-cell survival, it did not induce proliferation. Collectively, these data indicate that FhHDM-1 has significant therapeutic applications to promote β-cell survival, which is required for T1D and T2D prevention and islet transplantation. KEY MESSAGES: FhHDM-1 preserves β-cell mass in NOD mice and prevents the development of T1D. FhHDM-1 enhances phosphorylation of Akt in mouse β-cell lines. FhHDM-1 increases levels of NADH/NADPH in mouse β-cell lines in vitro. FhHDM-1 prevents cytokine-induced cell death of mouse β-cell lines and primary human β-cells in vitro via activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Inah Camaya
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Tsz Y Mok
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Maria Lund
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Joyce To
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, University of New South Wales, Sydney, Randwick, Australia
| | - Mark W Robinson
- School of Biological Sciences, Queen's University, Belfast, Northern Ireland, UK
| | - Jerran Santos
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Bronwyn O'Brien
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia.
| |
Collapse
|
13
|
Lalor R, Cwiklinski K, Calvani NED, Dorey A, Hamon S, Corrales JL, Dalton JP, De Marco Verissimo C. Pathogenicity and virulence of the liver flukes Fasciola hepatica and Fasciola Gigantica that cause the zoonosis Fasciolosis. Virulence 2021; 12:2839-2867. [PMID: 34696693 PMCID: PMC8632118 DOI: 10.1080/21505594.2021.1996520] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fasciolosis caused by the liver flukes Fasciola hepatica and Fasciola gigantica is one of the most important neglected parasitic diseases of humans and animals. The ability of the parasites to infect and multiply in their intermediate snail hosts, and their adaptation to a wide variety of mammalian definitive hosts contribute to their high transmissibility and distribution. Within the mammalian host, the trauma caused by the immature flukes burrowing through the liver parenchyma is associated with most of the pathogenesis. Similarly, the feeding activity and the physical presence of large flukes in the bile ducts can lead to anemia, inflammation, obstruction and cholangitis. The high frequency of non-synonymous polymorphisms found in Fasciola spp. genes allows for adaptation and invasion of a broad range of hosts. This is also facilitated by parasite’s excretory-secretory (ES) molecules that mediate physiological changes that allows their establishment within the host. ES contains cathepsin peptidases that aid parasite invasion by degrading collagen and fibronectin. In the bile ducts, cathepsin-L is critical to hemoglobin digestion during feeding activities. Other molecules (peroxiredoxin, cathepsin-L and Kunitz-type inhibitor) stimulate a strong immune response polarized toward a Treg/Th2 phenotype that favors fluke’s survival. Helminth defense molecule, fatty acid binding proteins, Fasciola-specific glycans and miRNAs modulate host pro-inflammatory responses, while antioxidant scavenger enzymes work in an orchestrated way to deter host oxidant-mediated damage. Combining these strategies Fasciola spp. survive for decades within their mammalian host, where they reproduce and spread to become one of the most widespread zoonotic worm parasites in the world.
Collapse
Affiliation(s)
- Richard Lalor
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - Krystyna Cwiklinski
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - Nichola Eliza Davies Calvani
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - Amber Dorey
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - Siobhán Hamon
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - Jesús López Corrales
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - John Pius Dalton
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - Carolina De Marco Verissimo
- Molecular Parasitology Laboratory, Centre for One Health and Ryan Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
14
|
Dorey A, Cwiklinski K, Rooney J, De Marco Verissimo C, López Corrales J, Jewhurst H, Fazekas B, Calvani NED, Hamon S, Gaughan S, Dalton JP, Lalor R. Autonomous Non Antioxidant Roles for Fasciola hepatica Secreted Thioredoxin-1 and Peroxiredoxin-1. Front Cell Infect Microbiol 2021; 11:667272. [PMID: 34026663 PMCID: PMC8131638 DOI: 10.3389/fcimb.2021.667272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/22/2021] [Indexed: 01/07/2023] Open
Abstract
Trematode parasites of the genus Fasciola are the cause of liver fluke disease (fasciolosis) in humans and their livestock. Infection of the host involves invasion through the intestinal wall followed by migration in the liver that results in extensive damage, before the parasite settles as a mature egg-laying adult in the bile ducts. Genomic and transcriptomic studies revealed that increased metabolic stress during the rapid growth and development of F. hepatica is balanced with the up-regulation of the thiol-independent antioxidant system. In this cascade system thioredoxin/glutathione reductase (TGR) reduces thioredoxin (Trx), which then reduces and activates peroxiredoxin (Prx), whose major function is to protect cells against the damaging hydrogen peroxide free radicals. F. hepatica expresses a single TGR, three Trx and three Prx genes; however, the transcriptional expression of Trx1 and Prx1 far out-weighs (>50-fold) other members of their family, and both are major components of the parasite secretome. While Prx1 possesses a leader signal peptide that directs its secretion through the classical pathway and explains why this enzyme is found freely soluble in the secretome, Trx1 lacks a leader peptide and is secreted via an alternative pathway that packages the majority of this enzyme into extracellular vesicles (EVs). Here we propose that F. hepatica Prx1 and Trx1 do not function as part of the parasite’s stress-inducible thiol-dependant cascade, but play autonomous roles in defence against the general anti-pathogen oxidative burst by innate immune cells, in the modulation of host immune responses and regulation of inflammation.
Collapse
Affiliation(s)
- Amber Dorey
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Krystyna Cwiklinski
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - James Rooney
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Carolina De Marco Verissimo
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Jesús López Corrales
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Heather Jewhurst
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Barbara Fazekas
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Nichola Eliza Davies Calvani
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Siobhán Hamon
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Siobhán Gaughan
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - John P Dalton
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| | - Richard Lalor
- Molecular Parasitology Laboratory, Centre of One Health (COH), Ryan Institute, National University of Ireland, Galway, Ireland
| |
Collapse
|
15
|
Fasciola hepatica hijacks host macrophage miRNA machinery to modulate early innate immune responses. Sci Rep 2021; 11:6712. [PMID: 33762636 PMCID: PMC7990952 DOI: 10.1038/s41598-021-86125-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 03/08/2021] [Indexed: 01/31/2023] Open
Abstract
Fasciola hepatica, a global worm parasite of humans and their livestock, regulates host innate immune responses within hours of infection. Host macrophages, essential to the first-line defence mechanisms, are quickly restricted in their ability to initiate a classic protective pro-inflammatory immune response. We found that macrophages from infected animals are enriched with parasite-derived micro(mi)RNAs. The most abundant of these miRNAs, fhe-miR-125b, is released by the parasite via exosomes and is homologous to a mammalian miRNA, hsa-miR-125b, that is known to regulate the activation of pro-inflammatory M1 macrophages. We show that the parasite fhe-miR-125b loads onto the mammalian Argonaut protein (Ago-2) within macrophages during infection and, therefore, propose that it mimics host miR-125b to negatively regulate the production of inflammatory cytokines. The hijacking of the miRNA machinery controlling innate cell function could be a fundamental mechanism by which worm parasites disarm the early immune responses of their host to ensure successful infection.
Collapse
|
16
|
Bennett APS, Robinson MW. Trematode Proteomics: Recent Advances and Future Directions. Pathogens 2021; 10:348. [PMID: 33809501 PMCID: PMC7998542 DOI: 10.3390/pathogens10030348] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/09/2021] [Accepted: 03/14/2021] [Indexed: 12/14/2022] Open
Abstract
Trematodes cause disease in millions of people worldwide, but the absence of commercial vaccines has led to an over-reliance on a handful of monotherapies to control infections. Since drug-resistant fluke populations are emerging, a deeper understanding of parasite biology and host interactions is required to identify new drug targets and immunogenic vaccine candidates. Mass spectrometry-based proteomics represents a key tool to that end. Recent studies have capitalised on the wider availability of annotated helminth genomes to achieve greater coverage of trematode proteomes and discover new aspects of the host-parasite relationship. This review focusses on these latest advances. These include how the protein components of fluke extracellular vesicles have given insight into their biogenesis and cellular interactions. In addition, how the integration of transcriptome/proteome datasets has revealed that the expression and secretion of selected families of liver fluke virulence factors and immunomodulators are regulated in accordance with parasite development and migration within the mammalian host. Furthermore, we discuss the use of immunoproteomics as a tool to identify vaccine candidates associated with protective antibody responses. Finally, we highlight how established and emerging technologies, such as laser microdissection and single-cell proteomics, could be exploited to resolve the protein profiles of discrete trematode tissues or cell types which, in combination with functional tools, could pinpoint optimal targets for fluke control.
Collapse
Affiliation(s)
| | - Mark W. Robinson
- School of Biological Sciences, Queen’s University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, Northern Ireland, UK;
| |
Collapse
|
17
|
Huson KM, Atcheson E, Oliver NAM, Best P, Barley JP, Hanna REB, McNeilly TN, Fang Y, Haldenby S, Paterson S, Robinson MW. Transcriptome and Secretome Analysis of Intra-Mammalian Life-Stages of Calicophoron daubneyi Reveals Adaptation to a Unique Host Environment. Mol Cell Proteomics 2021; 20:100055. [PMID: 33581320 PMCID: PMC7973311 DOI: 10.1074/mcp.ra120.002175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Paramphistomosis, caused by the rumen fluke, Calicophoron daubneyi, is a parasitic infection of ruminant livestock, which has seen a rapid rise in prevalence throughout Western Europe in recent years. After ingestion of metacercariae (parasite cysts) by the mammalian host, newly excysted juveniles (NEJs) emerge and invade the duodenal submucosa, which causes significant pathology in heavy infections. The immature flukes then migrate upward, along the gastrointestinal tract, and enter the rumen where they mature and begin to produce eggs. Despite their emergence, and sporadic outbreaks of acute disease, we know little about the molecular mechanisms used by C. daubneyi to establish infection, acquire nutrients, and avoid the host immune response. Here, transcriptome analysis of four intramammalian life-cycle stages, integrated with secretome analysis of the NEJ and adult parasites (responsible for acute and chronic diseases, respectively), revealed how the expression and secretion of selected families of virulence factors and immunomodulators are regulated in accordance with fluke development and migration. Our data show that while a family of cathepsins B with varying S2 subsite residues (indicating distinct substrate specificities) is differentially secreted by NEJs and adult flukes, cathepsins L and F are secreted in low abundance by NEJs only. We found that C. daubneyi has an expanded family of aspartic peptidases, which is upregulated in adult worms, although they are under-represented in the secretome. The most abundant proteins in adult fluke secretions were helminth defense molecules that likely establish an immune environment permissive to fluke survival and/or neutralize pathogen-associated molecular patterns such as bacterial lipopolysaccharide in the microbiome-rich rumen. The distinct collection of molecules secreted by C. daubneyi allowed the development of the first coproantigen-based ELISA for paramphistomosis which, importantly, did not recognize antigens from other helminths commonly found as coinfections with rumen fluke.
Collapse
Affiliation(s)
- Kathryn M Huson
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland
| | - Erwan Atcheson
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland
| | - Nicola A M Oliver
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland
| | - Philip Best
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland
| | - Jason P Barley
- Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, Northern Ireland
| | - Robert E B Hanna
- Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, Northern Ireland
| | - Tom N McNeilly
- Disease Control Department, Moredun Research Institute, Edinburgh, Scotland
| | - Yongxiang Fang
- Centre for Genomic Research, University of Liverpool, Liverpool, England
| | - Sam Haldenby
- Centre for Genomic Research, University of Liverpool, Liverpool, England
| | - Steve Paterson
- Centre for Genomic Research, University of Liverpool, Liverpool, England
| | - Mark W Robinson
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland.
| |
Collapse
|
18
|
Zafra R, Buffoni L, Pérez-Caballero R, Molina-Hernández V, Ruiz-Campillo MT, Pérez J, Martínez-Moreno Á, Martínez Moreno FJ. Efficacy of a multivalent vaccine against Fasciola hepatica infection in sheep. Vet Res 2021; 52:13. [PMID: 33509286 PMCID: PMC7841919 DOI: 10.1186/s13567-021-00895-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
In this work we report the protection found in a vaccination trial performed in sheep with two different vaccines composed each one by a cocktail of antigens (rCL1, rPrx, rHDM and rLAP) formulated in two different adjuvants (Montanide ISA 61 VG (G1) and Alhydrogel®(G2)). The parameters of protection tested were fluke burden, faecal egg count and evaluation of hepatic lesions. In vaccinated group 1 we found a significant decrease in fluke burden in comparison to both unimmunised and infected control group (37.2%; p = 0.002) and to vaccinated group 2 (Alhydrogel®) (27.08%; p = 0.016). The lower fluke burden found in G1 was accompanied by a decrease in egg output of 28.71% in comparison with the infected control group. Additionally, gross hepatic lesions found in vaccine 1 group showed a significant decrease (p = 0.03) in comparison with unimmunised-infected group. The serological study showed the highest level for both IgG1 and IgG2 in animals from group 1. All these data support the hypothesis of protection found in vaccine 1 group.
Collapse
Affiliation(s)
- Rafael Zafra
- Animal Health Department (Parasitology and Parasitic Diseases), Faculty of Veterinary Medicine, University of Córdoba, Sanidad Animal Building, Rabanales Campus, Córdoba, Spain
| | - Leandro Buffoni
- Animal Health Department (Parasitology and Parasitic Diseases), Faculty of Veterinary Medicine, University of Córdoba, Sanidad Animal Building, Rabanales Campus, Córdoba, Spain.
| | - Raúl Pérez-Caballero
- Animal Health Department (Parasitology and Parasitic Diseases), Faculty of Veterinary Medicine, University of Córdoba, Sanidad Animal Building, Rabanales Campus, Córdoba, Spain
| | - Verónica Molina-Hernández
- Department of Anatomy, Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, Sanidad Animal Building, Rabanales Campus, Córdoba, Spain
| | - María T Ruiz-Campillo
- Department of Anatomy, Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, Sanidad Animal Building, Rabanales Campus, Córdoba, Spain
| | - José Pérez
- Department of Anatomy, Comparative Pathology and Toxicology, Faculty of Veterinary Medicine, University of Córdoba, Sanidad Animal Building, Rabanales Campus, Córdoba, Spain
| | - Álvaro Martínez-Moreno
- Animal Health Department (Parasitology and Parasitic Diseases), Faculty of Veterinary Medicine, University of Córdoba, Sanidad Animal Building, Rabanales Campus, Córdoba, Spain
| | - Francisco J Martínez Moreno
- Animal Health Department (Parasitology and Parasitic Diseases), Faculty of Veterinary Medicine, University of Córdoba, Sanidad Animal Building, Rabanales Campus, Córdoba, Spain
| |
Collapse
|
19
|
Cwiklinski K, Robinson MW, Donnelly S, Dalton JP. Complementary transcriptomic and proteomic analyses reveal the cellular and molecular processes that drive growth and development of Fasciola hepatica in the host liver. BMC Genomics 2021; 22:46. [PMID: 33430759 PMCID: PMC7797711 DOI: 10.1186/s12864-020-07326-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 12/17/2022] Open
Abstract
Background The major pathogenesis associated with Fasciola hepatica infection results from the extensive tissue damage caused by the tunnelling and feeding activity of immature flukes during their migration, growth and development in the liver. This is compounded by the pathology caused by host innate and adaptive immune responses that struggle to simultaneously counter infection and repair tissue damage. Results Complementary transcriptomic and proteomic approaches defined the F. hepatica factors associated with their migration in the liver, and the resulting immune-pathogenesis. Immature liver-stage flukes express ~ 8000 transcripts that are enriched for transcription and translation processes reflective of intensive protein production and signal transduction pathways. Key pathways that regulate neoblast/pluripotent cells, including the PI3K-Akt signalling pathway, are particularly dominant and emphasise the importance of neoblast-like cells for the parasite’s rapid development. The liver-stage parasites display different secretome profiles, reflecting their distinct niche within the host, and supports the view that cathepsin peptidases, cathepsin peptidase inhibitors, saposins and leucine aminopeptidases play a central role in the parasite’s destructive migration, and digestion of host tissue and blood. Immature flukes are also primed for countering immune attack by secreting immunomodulating fatty acid binding proteins (FABP) and helminth defence molecules (FhHDM). Combined with published host microarray data, our results suggest that considerable immune cell infiltration and subsequent fibrosis of the liver tissue exacerbates oxidative stress within parenchyma that compels the expression of a range of antioxidant molecules within both host and parasite. Conclusions The migration of immature F. hepatica parasites within the liver is associated with an increase in protein production, expression of signalling pathways and neoblast proliferation that drive their rapid growth and development. The secretion of a defined set of molecules, particularly cathepsin L peptidases, peptidase-inhibitors, saponins, immune-regulators and antioxidants allow the parasite to negotiate the liver micro-environment, immune attack and increasing levels of oxidative stress. This data contributes to the growing F. hepatica -omics information that can be exploited to understand parasite development more fully and for the design of novel control strategies to prevent host liver tissue destruction and pathology.
Collapse
Affiliation(s)
- Krystyna Cwiklinski
- Zoology Department, School of Natural Sciences, Centre for One Health, Ryan Institute, National University of Ireland Galway, Galway, Ireland.
| | - Mark W Robinson
- School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Sheila Donnelly
- Zoology Department, School of Natural Sciences, Centre for One Health, Ryan Institute, National University of Ireland Galway, Galway, Ireland.,The School of Life Sciences, University of Technology, Sydney, Australia
| | - John P Dalton
- Zoology Department, School of Natural Sciences, Centre for One Health, Ryan Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
20
|
Polyphosphate is an extracellular signal that can facilitate bacterial survival in eukaryotic cells. Proc Natl Acad Sci U S A 2020; 117:31923-31934. [PMID: 33268492 DOI: 10.1073/pnas.2012009117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Polyphosphate is a linear chain of phosphate residues and is present in organisms ranging from bacteria to humans. Pathogens such as Mycobacterium tuberculosis accumulate polyphosphate, and reduced expression of the polyphosphate kinase that synthesizes polyphosphate decreases their survival. How polyphosphate potentiates pathogenicity is poorly understood. Escherichia coli K-12 do not accumulate detectable levels of extracellular polyphosphate and have poor survival after phagocytosis by Dictyostelium discoideum or human macrophages. In contrast, Mycobacterium smegmatis and Mycobacterium tuberculosis accumulate detectable levels of extracellular polyphosphate, and have relatively better survival after phagocytosis by D. discoideum or macrophages. Adding extracellular polyphosphate increased E. coli survival after phagocytosis by D. discoideum and macrophages. Reducing expression of polyphosphate kinase 1 in M. smegmatis reduced extracellular polyphosphate and reduced survival in D. discoideum and macrophages, and this was reversed by the addition of extracellular polyphosphate. Conversely, treatment of D. discoideum and macrophages with recombinant yeast exopolyphosphatase reduced the survival of phagocytosed M. smegmatis or M. tuberculosis D. discoideum cells lacking the putative polyphosphate receptor GrlD had reduced sensitivity to polyphosphate and, compared to wild-type cells, showed increased killing of phagocytosed E. coli and M. smegmatis Polyphosphate inhibited phagosome acidification and lysosome activity in D. discoideum and macrophages and reduced early endosomal markers in macrophages. Together, these results suggest that bacterial polyphosphate potentiates pathogenicity by acting as an extracellular signal that inhibits phagosome maturation.
Collapse
|
21
|
Dolasia K, Nazar F, Mukhopadhyay S. Mycobacterium tuberculosis PPE18 protein inhibits MHC class II antigen presentation and B cell response in mice. Eur J Immunol 2020; 51:603-619. [PMID: 33084017 DOI: 10.1002/eji.201848071] [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: 01/09/2019] [Revised: 09/22/2020] [Accepted: 10/19/2020] [Indexed: 01/18/2023]
Abstract
PPE18 protein belongs to PE/PPE family of Mycobacterium tuberculosis. We reported earlier that PPE18 protein provides survival advantage to M. tuberculosis during infection. In the current study, we found that PPE18 inhibits MHC class II-mediated antigen presentation by macrophages in a dose-dependent manner without affecting the surface level of MHC class II or co-stimulatory molecules. PPE18 does not affect antigen uptake or presentation of preprocessed peptide by macrophages. Antigen degradation was found to be inhibited by PPE18 protein due to perturbation in phagolysosomal acidification. PPE18-mediated inhibition of MHC class II antigen presentation caused poorer activation of CD4 T cells. Mice infected with M. smegmatis expressing PPE18 exhibited reduced maturation and activation of B cells and had decreased Mycobacteria-specific antibody titers. Thus M. tuberculosis probably utilizes PPE18 to inhibit MHC class II antigen presentation causing poorer activation of adaptive immune responses. This study may be useful in understanding host-pathogen interaction and open up directions of designing novel therapeutics targeting PPE18 to tackle this nefarious pathogen.
Collapse
Affiliation(s)
- Komal Dolasia
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, India
| | - Faiza Nazar
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| |
Collapse
|
22
|
Wang J, Tan W, Li G, Wu D, He H, Xu J, Yi M, Zhang Y, Aghvami SA, Fraden S, Xu B. Enzymatic Insertion of Lipids Increases Membrane Tension for Inhibiting Drug Resistant Cancer Cells. Chemistry 2020; 26:15116-15120. [PMID: 32579262 DOI: 10.1002/chem.202002974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Indexed: 12/19/2022]
Abstract
Although lipids contribute to cancer drug resistance, it is challenging to target diverse range of lipids. Here, we show enzymatically inserting exceedingly simple synthetic lipids into membranes for increasing membrane tension and selectively inhibiting drug resistant cancer cells. The lipid, formed by conjugating dodecylamine to d-phosphotyrosine, self-assembles to form micelles. Enzymatic dephosphorylation of the micelles inserts the lipids into membranes and increases membrane tension. The micelles effectively inhibit a drug resistant glioblastoma cell (T98G) or a triple-negative breast cancer cell (HCC1937), without inducing acquired drug resistance. Moreover, the enzymatic reaction of the micelles promotes the accumulation of the lipids in the membranes of subcellular organelles (e.g., endoplasmic reticulum (ER), Golgi, and mitochondria), thus activating multiple regulated cell death pathways. This work, in which for the first time membrane tension is increased to inhibit cancer cells, illustrates a new and powerful supramolecular approach for antagonizing difficult drug targets.
Collapse
Affiliation(s)
- Jiaqing Wang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Guanying Li
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan
| | - Difei Wu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Jiashu Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Meihui Yi
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Ye Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan
| | - S Ali Aghvami
- Department of Physic, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Seth Fraden
- Department of Physic, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| |
Collapse
|
23
|
Corral-Ruiz GM, Sánchez-Torres LE. Fasciola hepatica-derived molecules as potential immunomodulators. Acta Trop 2020; 210:105548. [PMID: 32505597 DOI: 10.1016/j.actatropica.2020.105548] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/21/2020] [Accepted: 05/18/2020] [Indexed: 01/15/2023]
Abstract
Through the years, helminths have co-existed with many species. This process has allowed parasites to live within them for long periods and, in some cases, to generate offspring. In particular, this ability has allowed Fasciola hepatica to survive the diverse immunological responses faced within its wide range of hosts. The vast repertoire of molecules that are constantly secreted in large quantities by the parasite, acts directly on several cells of the immune system affecting their antiparasitic capacities. Interestingly, these molecules can direct the host immune response to an anti-inflammatory and regulatory phenotype that assures the survival of the parasite with less harm to the host. Based on these observations, some of the products of F. hepatica, as well as those of other helminths, have been studied, either as a total extract, extracellular vesicles or as purified molecules, to establish and characterize their anti-inflammatory mechanisms. Until now, the results obtained encourage further research directed to discover new helminth-derived alternatives to replace current therapies, which can be useful for people suffering from inflammatory diseases like autoimmunity or allergy processes that affect their life quality. In this review, some of the most studied molecules derived from F. hepatica and their modulating capacities are discussed.
Collapse
Affiliation(s)
- Gerardo Manuel Corral-Ruiz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala, s/n, 11340 Ciudad de México, México
| | - Luvia Enid Sánchez-Torres
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala, s/n, 11340 Ciudad de México, México.
| |
Collapse
|
24
|
Ryan S, Shiels J, Taggart CC, Dalton JP, Weldon S. Fasciola hepatica-Derived Molecules as Regulators of the Host Immune Response. Front Immunol 2020; 11:2182. [PMID: 32983184 PMCID: PMC7492538 DOI: 10.3389/fimmu.2020.02182] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022] Open
Abstract
Helminths (worms) are one of the most successful organisms in nature given their ability to infect millions of humans and animals worldwide. Their success can be attributed to their ability to modulate the host immune response for their own benefit by releasing excretory-secretory (ES) products. Accordingly, ES products have been lauded as a potential source of immunomodulators/biotherapeutics for an array of inflammatory diseases. However, there is a significant lack of knowledge regarding the specific interactions between these products and cells of the immune response. Many different compounds have been identified within the helminth "secretome," including antioxidants, proteases, mucin-like peptides, as well as helminth defense molecules (HDMs), each with unique influences on the host inflammatory response. HDMs are a conserved group of proteins initially discovered in the secretome of the liver fluke, Fasciola hepatica. HDMs interact with cell membranes without cytotoxic effects and do not exert antimicrobial activity, suggesting that these peptides evolved specifically for immunomodulatory purposes. A peptide generated from the HDM sequence, termed FhHDM-1, has shown extensive anti-inflammatory abilities in clinically relevant models of diseases such as diabetes, multiple sclerosis, asthma, and acute lung injury, offering hope for the development of a new class of therapeutics. In this review, the current knowledge of host immunomodulation by a range of F. hepatica ES products, particularly FhHDM-1, will be discussed. Immune regulators, including HDMs, have been identified from other helminths and will also be outlined to broaden our understanding of the variety of effects these potent molecules exert on immune cells.
Collapse
Affiliation(s)
- Sinéad Ryan
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Jenna Shiels
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Clifford C Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - John P Dalton
- Centre of One Health (COH), Ryan Institute, School of Natural Sciences, National University of Ireland Galway, Galway, United Kingdom
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
25
|
Shiels J, Cwiklinski K, Alvarado R, Thivierge K, Cotton S, Gonzales Santana B, To J, Donnelly S, Taggart CC, Weldon S, Dalton JP. Schistosoma mansoni immunomodulatory molecule Sm16/SPO-1/SmSLP is a member of the trematode-specific helminth defence molecules (HDMs). PLoS Negl Trop Dis 2020; 14:e0008470. [PMID: 32644998 PMCID: PMC7373315 DOI: 10.1371/journal.pntd.0008470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/21/2020] [Accepted: 06/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Sm16, also known as SPO-1 and SmSLP, is a low molecular weight protein (~16kDa) secreted by the digenean trematode parasite Schistosoma mansoni, one of the main causative agents of human schistosomiasis. The molecule is secreted from the acetabular gland of the cercariae during skin invasion and is believed to perform an immune-suppressive function to protect the invading parasite from innate immune cell attack. METHODOLOGY/PRINCIPAL FINDINGS We show that Sm16 homologues of the Schistosomatoidea family are phylogenetically related to the helminth defence molecule (HDM) family of immunomodulatory peptides first described in Fasciola hepatica. Interrogation of 69 helminths genomes demonstrates that HDMs are exclusive to trematode species. Structural analyses of Sm16 shows that it consists predominantly of an amphipathic alpha-helix, much like other HDMs. In S. mansoni, Sm16 is highly expressed in the cercariae and eggs but not in adult worms, suggesting that the molecule is of importance not only during skin invasion but also in the pro-inflammatory response to eggs in the liver tissues. Recombinant Sm16 and a synthetic form, Sm16 (34-117), bind to macrophages and are internalised into the endosomal/lysosomal system. Sm16 (34-117) elicited a weak pro-inflammatory response in macrophages in vitro but also suppressed the production of bacterial lipopolysaccharide (LPS)-induced inflammatory cytokines. Evaluation of the transcriptome of human macrophages treated with a synthetic Sm16 (34-117) demonstrates that the peptide exerts significant immunomodulatory effects alone, as well as in the presence of LPS. Pathways most significantly influenced by Sm16 (34-117) were those involving transcription factors peroxisome proliferator-activated receptor (PPAR) and liver X receptors/retinoid X receptor (LXR/RXR) which are intricately involved in regulating the cellular metabolism of macrophages (fatty acid, cholesterol and glucose homeostasis) and are central to inflammatory responses. CONCLUSIONS/SIGNIFICANCE These results offer new insights into the structure and function of a well-known immunomodulatory molecule, Sm16, and places it within a wider family of trematode-specific small molecule HDM immune-modulators with immuno-biotherapeutic possibilities.
Collapse
Affiliation(s)
- Jenna Shiels
- School of Biological Sciences, Queen’s University Belfast, Northern Ireland
- Airway Innate Immunity Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine (WWIEM), School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Northern Ireland
| | - Krystyna Cwiklinski
- School of Biological Sciences, Queen’s University Belfast, Northern Ireland
- Center of One Health (COH) and Ryan Institute, School of Natural Science, National University of Ireland Galway, Galway, Ireland
| | - Raquel Alvarado
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW, Australia
| | - Karine Thivierge
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada
| | - Sophie Cotton
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada
| | | | - Joyce To
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW, Australia
| | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW, Australia
| | - Clifford C. Taggart
- Airway Innate Immunity Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine (WWIEM), School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Northern Ireland
| | - Sinead Weldon
- Airway Innate Immunity Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine (WWIEM), School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Northern Ireland
| | - John P. Dalton
- School of Biological Sciences, Queen’s University Belfast, Northern Ireland
- Center of One Health (COH) and Ryan Institute, School of Natural Science, National University of Ireland Galway, Galway, Ireland
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
26
|
Maizels RM. Regulation of immunity and allergy by helminth parasites. Allergy 2020; 75:524-534. [PMID: 31187881 DOI: 10.1111/all.13944] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/26/2022]
Abstract
There is increasing interest in helminth parasite modulation of the immune system, both from the fundamental perspective of the "arms race" between host and parasite, and equally importantly, to understand if parasites offer new pathways to abate and control untoward immune responses in humans. This article reviews the epidemiological and experimental evidence for parasite down-regulation of host immunity and immunopathology, in allergy and other immune disorders, and recent progress towards defining the mechanisms and molecular mediators which parasites exploit in order to modulate their host. Among these are novel products that interfere with epithelial cell alarmins, dendritic cell activation, macrophage function and T-cell responsiveness through the promotion of an immunoregulatory environment. These modulatory effects assist parasites to establish and survive, while dampening immune reactivity to allergens, autoantigens and microbiome determinants.
Collapse
Affiliation(s)
- Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunology and Inflammation University of Glasgow Glasgow UK
| |
Collapse
|
27
|
Felizatti AP, Zeraik AE, Basso LG, Kumagai PS, Lopes JL, Wallace B, Araujo AP, DeMarco R. Interactions of amphipathic α-helical MEG proteins from Schistosoma mansoni with membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183173. [DOI: 10.1016/j.bbamem.2019.183173] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 02/04/2023]
|
28
|
Kang JM, Yoo WG, Lê HG, Lee J, Sohn WM, Na BK. Clonorchis sinensis MF6p/HDM (CsMF6p/HDM) induces pro-inflammatory immune response in RAW 264.7 macrophage cells via NF-κB-dependent MAPK pathways. Parasit Vectors 2020; 13:20. [PMID: 31931867 PMCID: PMC6958574 DOI: 10.1186/s13071-020-3882-0] [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: 10/02/2019] [Accepted: 01/01/2020] [Indexed: 12/12/2022] Open
Abstract
Background MF6p/host defense molecules (HDMs) are a broad family of small proteins secreted by helminth parasites. Although the physiological role of MF6p/HDMs in trematode parasites is not fully understood, their potential biological function in maintaining heme homeostasis and modulating host immune response has been proposed. Methods A gene encoding the MF6p/HDM of Clonorchis sinensis (CsMF6p/HDM) was cloned. Recombinant CsMF6p/HDM (rCsMF6p/HDM) was expressed in Escherichia coli. The biochemical and immunological properties of rCsMF6/HDM were analyzed. CsMF6p/HDM induced pro-inflammatory response in RAW 264.7 cells was analyzed by cytokine array assay, reverse transcription polymerase chain reaction, and enzyme-linked immunosorbent assay. The structural feature of CsMF6p/HDM was analyzed by three-dimensional modeling and molecular docking simulations. Results The CsMF6p/HDM shares a high level of amino acid sequence similarity with orthologs from other trematodes and is expressed in diverse developmental stages of the parasite. The rCsMF6p/HDM bound to bacteria-derived lipopolysaccharide (LPS), without effectively neutralizing LPS-induced inflammatory response in RAW 264.7 macrophage cells. Rather, the rCsMF6p/HDM induced pro-inflammatory immune response, which is characterized by the expression of TNF-α and IL-6, in RAW 264.7 cells. The rCsMF6p/HDM-induced pro-inflammatory immune response was regulated by JNK and p38 MAPKs, and was effectively down-regulated via inhibition of NF-κB. The structural analysis of CsMF6p/HDM and the docking simulation with LPS suggested insufficient capture of LPS by CsMF6p/HDM, which suggested that rCsMF6p/HDM could not effectively neutralize LPS-induced inflammatory response in RAW 264.7 cells. Conclusions Although rCsMF6p/HDM binds to LPS, the binding affinity may not be sufficient to maintain a stable complex of rCsMF6p/HDM and LPS. Moreover, the rCsMF6p/HDM-induced pro-inflammatory response is characterized by the release of IL-6 and TNF-α in RAW 264.7 macrophage cells. The pro-inflammatory response induced by rCsMF6p/HDM is mediated via NF-κB-dependent MAPK signaling pathway. These results collectively suggest that CsMF6p/HDM mediates C. sinensis-induced inflammation cascades that eventually lead to hepatobiliary diseases.![]()
Collapse
Affiliation(s)
- Jung-Mi Kang
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Won Gi Yoo
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, 06974, Republic of Korea
| | - Hương Giang Lê
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Jinyoung Lee
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,Department of Tropical Medicine, and Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Woon-Mok Sohn
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea
| | - Byoung-Kuk Na
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea. .,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea.
| |
Collapse
|
29
|
Abstract
Fasciola hepatica, the common liver fluke, causes infection of livestock throughout temperate regions of the globe. This helminth parasite has an indirect lifecycle, relying on the presence of the mud snail to complete its transition from egg to definitive host (Beesley et al., Transbound Emerg Dis 65:199-216, 2017). Within the definitive host, the parasite excysts in the intestine forming a newly excysted juvenile (NEJ) and migrates via the peritoneal cavity to the liver. Disease resulting from infection can be acute or chronic depending on the host and the number of parasites present. Sheep may succumb to a fatal acute infection if the challenge of metacercariae is great enough. However, in cattle chronic disease is the most likely outcome with parasites surviving for long periods of time. Annual losses are estimated to be in the region of US$ 2000 million to the agricultural industry (Beesley et al., Transbound Emerg Dis 65:199-216, 2017). Management of the disease depends heavily on chemotherapy with triclabendazole being the drug of choice, consistent use for over 20 years has resulted in drug-resistant strains emerging worldwide (Beesley et al., Int J Parasitol 47:11-20, 2017). A more sustainable approach to control would be through vaccination and indeed a lead candidate has been identified, cathepsin L1. Despite these promising results the parasite continues to confound our own and host efforts to generate long-lasting and effective immunity. In this brief review we focus our attention on those mechanisms that the parasite utilises to circumvent the innate based defense mechanisms within the host.
Collapse
Affiliation(s)
- Robin J Flynn
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK.
| | - Mayowa Musah-Eroje
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| |
Collapse
|
30
|
Khan YA, Maurya SK, Kulkarni C, Tiwari MC, Nagar GK, Chattopadhyay N. Fasciola
helminth defense molecule‐1 protects against experimental arthritis by inhibiting osteoclast formation and function without modulating the systemic immune response. FASEB J 2019; 34:1091-1106. [DOI: 10.1096/fj.201901480rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/05/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Yasir Akhtar Khan
- Division of Endocrinology CSIR‐Central Drug Research Institute Lucknow India
- Section of Parasitology Department of Zoology Aligarh Muslim University Aligarh India
| | | | - Chirag Kulkarni
- Division of Endocrinology CSIR‐Central Drug Research Institute Lucknow India
- Academy of Scientific and Innovative Research CSIR‐Central Drug Research Institute Lucknow India
| | | | - Geet Kumar Nagar
- Division of Endocrinology CSIR‐Central Drug Research Institute Lucknow India
| | - Naibedya Chattopadhyay
- Division of Endocrinology CSIR‐Central Drug Research Institute Lucknow India
- Academy of Scientific and Innovative Research CSIR‐Central Drug Research Institute Lucknow India
| |
Collapse
|
31
|
Engineering universal cells that evade immune detection. Nat Rev Immunol 2019; 19:723-733. [DOI: 10.1038/s41577-019-0200-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2019] [Indexed: 12/15/2022]
|
32
|
Hammond K, Lewis H, Faruqui N, Russell C, Hoogenboom BW, Ryadnov MG. Helminth Defense Molecules as Design Templates for Membrane Active Antibiotics. ACS Infect Dis 2019; 5:1471-1479. [PMID: 31117348 DOI: 10.1021/acsinfecdis.9b00157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A design template for membrane active antibiotics against microbial and tumor cells is described. The template is an amino acid sequence that combines the properties of helminth defense molecules, which are not cytolytic, with the properties of host-defense peptides, which disrupt microbial membranes. Like helminth defense molecules, the template folds into an amphipathic helix in both mammalian host and microbial phospholipid membranes. Unlike these molecules, the template exhibits antimicrobial and anticancer properties that are comparable to those of antimicrobial and anticancer antibiotics. The selective antibiotic activity of the template builds upon a functional synergy between three distinctive faces of the helix, which is in contrast to two faces of membrane-disrupting amphipathic structures. This synergy enables the template to adapt pore formation mechanisms according to the nature of the target membrane, inducing the lysis of microbial and tumor cells.
Collapse
Affiliation(s)
- Katharine Hammond
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
- Department of Physics & Astronomy, University College London, Gower Street, London WC1E 6BT, United Kingdom
- London Centre for Nanotechnology, University College London, Gordon Street, London WC1H 0AH, United Kingdom
| | - Helen Lewis
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
| | - Nilofar Faruqui
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
| | - Craig Russell
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
| | - Bart W. Hoogenboom
- Department of Physics & Astronomy, University College London, Gower Street, London WC1E 6BT, United Kingdom
- London Centre for Nanotechnology, University College London, Gordon Street, London WC1H 0AH, United Kingdom
| | - Maxim G. Ryadnov
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, United Kingdom
- Department of Physics, King’s College London, Strand Lane, London WC2R, United Kingdom
| |
Collapse
|
33
|
de la Torre-Escudero E, Gerlach JQ, Bennett APS, Cwiklinski K, Jewhurst HL, Huson KM, Joshi L, Kilcoyne M, O’Neill S, Dalton JP, Robinson MW. Surface molecules of extracellular vesicles secreted by the helminth pathogen Fasciola hepatica direct their internalisation by host cells. PLoS Negl Trop Dis 2019; 13:e0007087. [PMID: 30657764 PMCID: PMC6355031 DOI: 10.1371/journal.pntd.0007087] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 01/31/2019] [Accepted: 12/18/2018] [Indexed: 01/22/2023] Open
Abstract
Helminth parasites secrete extracellular vesicles (EVs) that can be internalised by host immune cells resulting in modulation of host immunity. While the molecular cargo of EVs have been characterised in many parasites, little is known about the surface-exposed molecules that participate in ligand-receptor interactions with the host cell surface to initiate vesicle docking and subsequent internalisation. Using a membrane-impermeable biotin reagent to capture proteins displayed on the outer membrane surface of two EV sub-populations (termed 15k and 120k EVs) released by adult F. hepatica, we describe 380 surface proteins including an array of virulence factors, membrane transport proteins and molecules involved in EV biogenesis/trafficking. Proteomics and immunohistochemical analysis show that the 120k EVs have an endosomal origin and may be released from the parasite via the protonephridial (excretory) system whilst the larger 15k EVs are released from the gastrodermal epithelial cells that line the fluke gut. A parallel lectin microarray strategy was used to profile the topology of major surface oligosaccharides of intact fluorogenically-labelled EVs as they would be displayed to the host. Lectin profiles corresponding to glycoconjugates exposed on the surface of the 15 K and 120K EV sub-populations are practically identical but are distinct from those of the parasite surface tegument, although all are predominated by high mannose sugars. We found that while the F. hepatica EVs were resistant to exo- and endo-glycosidases, the glyco-amidase PNGase F drastically remodelled the surface oligosaccharides and blocked the uptake of EVs by host macrophages. In contrast, pre-treatment with antibodies obtained from infected hosts, or purified antibodies raised against the extracellular domains of specific EV surface proteins (DM9-containing protein, CD63 receptor and myoferlin), significantly enhanced their cellular internalisation. This work highlights the diversity of EV biogenesis and trafficking pathways used by F. hepatica and sheds light on the molecular interaction between parasite EVs and host cells. Over the last decade, it has become recognised that extracellular vesicles (EVs) are important mediators of communication by transferring molecular signals (including proteins, lipids, complex carbohydrates, mRNA, microRNA and other non-coding RNA species), between cells. Variously described as exosomes or microvesicles depending on their cellular origin and mode of biogenesis, EVs perform a variety of roles in the maintenance of normal physiology but also participate in pathological settings. EVs also play an important role in host-pathogen interactions, with recent work suggesting that they contribute to helminth immunomodulatory strategies. Here we have identified the proteins and sugars displayed on the outer surface of two sub-types of EVs released by the helminth pathogen Fasciola hepatica. We show that the proteins are antigenic and direct EV internalisation by host macrophages. Our study provides a better understanding of how parasite-derived EVs interact with host cells which is important for future development of therapeutics/vaccines that target this interface.
Collapse
Affiliation(s)
- Eduardo de la Torre-Escudero
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Jared Q. Gerlach
- Glycoscience Group, Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Ireland
| | - Adam P. S. Bennett
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Krystyna Cwiklinski
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Heather L. Jewhurst
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Kathryn M. Huson
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Lokesh Joshi
- Glycoscience Group, Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Ireland
| | - Michelle Kilcoyne
- Carbohydrate Signalling Group, Discipline of Microbiology, School of Natural Sciences, National University of Ireland Galway, Ireland
| | - Sandra O’Neill
- School of Biotechnology, Dublin City University, Dublin 9, Republic of Ireland
| | - John P. Dalton
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Mark W. Robinson
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
- * E-mail:
| |
Collapse
|
34
|
Maizels RM, Smits HH, McSorley HJ. Modulation of Host Immunity by Helminths: The Expanding Repertoire of Parasite Effector Molecules. Immunity 2018; 49:801-818. [PMID: 30462997 PMCID: PMC6269126 DOI: 10.1016/j.immuni.2018.10.016] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/13/2018] [Accepted: 10/30/2018] [Indexed: 02/09/2023]
Abstract
Helminths are extraordinarily successful parasites due to their ability to modulate the host immune response. They have evolved a spectrum of immunomodulatory molecules that are now beginning to be defined, heralding a molecular revolution in parasite immunology. These discoveries have the potential both to transform our understanding of parasite adaptation to the host and to develop possible therapies for immune-mediated disease. In this review we will summarize the current state of the art in parasite immunomodulation and discuss perspectives on future areas for research and discovery.
Collapse
Affiliation(s)
- Rick M Maizels
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | | | - Henry J McSorley
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
35
|
Musah-Eroje M, Flynn RJ. Fasciola hepatica, TGF-β and host mimicry: the enemy within. Curr Opin Microbiol 2018; 46:80-85. [PMID: 30317150 DOI: 10.1016/j.mib.2018.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/29/2018] [Accepted: 09/24/2018] [Indexed: 11/16/2022]
Abstract
Helminths parasites undergo developmental changes and migration within their definitive host, in addition to establishing chronic infection. Essential to this is the evasion of host immune responses; the canonical Th2 response is effective at removing parasites resident in the intestine. Conversely, helminths also promote the development of antigen-specific anergy and regulation. This often limits pathology but allows parasite survival, parasite effectors mediating this are the subject of intense study. They may be useful as future vaccine targets or xenogenic therapeutics. Fasciola hepatica possesses a family of TGF-like molecules of which one member, FhTLM, is capable of promoting intrinsic and extrinsic effects. Here we review the extrinsic effects of FhTLM on the host macrophage and its consequences for protective immunity. This review also discusses the specificities of FhTLM in light a very recent description of a nematode TGF-β mimic and the effects of endogenous TGF-β.
Collapse
Affiliation(s)
- Mayowa Musah-Eroje
- School of Veterinary Medicine and Science, University of Nottingham, LE12 5RD, United Kingdom
| | - Robin J Flynn
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, L3 5RF, United Kingdom.
| |
Collapse
|
36
|
Ewanchuk BW, Yates RM. The phagosome and redox control of antigen processing. Free Radic Biol Med 2018; 125:53-61. [PMID: 29578071 DOI: 10.1016/j.freeradbiomed.2018.03.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/09/2018] [Accepted: 03/20/2018] [Indexed: 11/22/2022]
Abstract
In addition to debris clearance and antimicrobial function, versatile organelles known as phagosomes play an essential role in the processing of exogenous antigen in antigen presenting cells. While there has been much attention on human leukocyte antigen haplotypes in the determination of antigenic peptide repertoires, the lumenal biochemistries within phagosomes and endosomes are emerging as equally-important determinants of peptide epitope composition and immunodominance. Recently, the lumenal redox microenvironment within these degradative compartments has been shown to impact two key antigenic processing chemistries: proteolysis by lysosomal cysteine proteases and disulfide reduction of protein antigens. Through manipulation of the balance between oxidative and reductive capacities in the phagosome-principally by modulating NADPH oxidase (NOX2) and γ-interferon-inducible lysosomal thiol reductase (GILT) activities-studies have demonstrated changes to antigen processing patterns leading to modified repertoires of antigenic peptides available for presentation, and subsequently, altered disease progression in T cell-driven autoimmunity. This review focuses on the mechanisms and consequences of redox-mediated phagosomal antigen processing, and the potential downstream implications to tolerance and autoimmunity.
Collapse
Affiliation(s)
- Benjamin W Ewanchuk
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|
37
|
Helminth Antigen-Conditioned Dendritic Cells Generate Anti-Inflammatory Cd4 T Cells Independent of Antigen Presentation via Major Histocompatibility Complex Class II. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2589-2604. [PMID: 30121255 DOI: 10.1016/j.ajpath.2018.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022]
Abstract
A recently identified feature of the host response to infection with helminth parasites is suppression of concomitant disease. Dendritic cells (DCs) exposed to antigens from the tapeworm Hymenolepis diminuta significantly reduce the severity of dinitrobenzene sulfonic acid-induced colitis in mice. Here we elucidate mechanisms underlying this cellular immunotherapy. We show a requirement for Ccr7 expression on transferred H. diminuta antigen-treated (HD)-DCs, suggesting that homing to secondary lymphoid tissues is important for suppression of colitis. Furthermore, sodium metaperiodate-sensitive helminth-derived glycans are required to drive the anti-colitic response in recipient mice. Induction of Th2-type cytokines and Gata-3+Cd4+ cells in secondary lymphoid tissues is dependent on major histocompatibility complex class II (MHC II) protein expression on transferred DCs, although remarkably, transfer of MHC II-/- HD-DCs still attenuated dinitrobenzene sulfonic acid-induced colitis in recipient mice. Moreover, transfer of Cd4+ splenic T cells retrieved from mice administered MHC II-/- HD-DCs suppressed dinitrobenzene sulfonic acid-induced colitis in recipient mice. Our studies reveal that HD-DCs can suppress colitis via an alternative MHC II-independent pathway that involves, in part, mobilization of T-cell responses. These data support the utility of HD-DCs in blocking colitis, revealing a requirement for Ccr7 and providing for HD-DC autologous immunotherapy for disease in which MHC II expression and/or function is compromised.
Collapse
|
38
|
da Silva ED, Cancela M, Monteiro KM, Ferreira HB, Zaha A. Antigen B from Echinococcus granulosus enters mammalian cells by endocytic pathways. PLoS Negl Trop Dis 2018; 12:e0006473. [PMID: 29727452 PMCID: PMC5955594 DOI: 10.1371/journal.pntd.0006473] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 05/16/2018] [Accepted: 04/23/2018] [Indexed: 01/18/2023] Open
Abstract
Background Cystic hydatid disease is a zoonosis caused by the larval stage (hydatid) of Echinococcus granulosus (Cestoda, Taeniidae). The hydatid develops in the viscera of intermediate host as a unilocular structure filled by the hydatid fluid, which contains parasitic excretory/secretory products. The lipoprotein Antigen B (AgB) is the major component of E. granulosus metacestode hydatid fluid. Functionally, AgB has been implicated in immunomodulation and lipid transport. However, the mechanisms underlying AgB functions are not completely known. Methodology/Principal findings In this study, we investigated AgB interactions with different mammalian cell types and the pathways involved in its internalization. AgB uptake was observed in four different cell lines, NIH-3T3, A549, J774 and RH. Inhibition of caveolae/raft-mediated endocytosis causes about 50 and 69% decrease in AgB internalization by RH and A549 cells, respectively. Interestingly, AgB colocalized with the raft endocytic marker, but also showed a partial colocalization with the clathrin endocytic marker. Finally, AgB colocalized with an endolysosomal tracker, providing evidence for a possible AgB destination after endocytosis. Conclusions/Significance The results indicate that caveolae/raft-mediated endocytosis is the main route to AgB internalization, and that a clathrin-mediated entry may also occur at a lower frequency. A possible fate for AgB after endocytosis seems to be the endolysosomal system. Cellular internalization and further access to subcellular compartments could be a requirement for AgB functions as a lipid carrier and/or immunomodulatory molecule, contributing to create a more permissive microenvironment to metacestode development and survival. Antigen B (AgB) is an oligomeric lipoprotein highly abundant in Echinococcus granulosus hydatid fluid. AgB has already been characterized as an immunomodulatory protein, capable of inducing a permissive immune response to parasite development. Also, an important role in lipid acquisition is attributed to AgB, because it has been found associated to different classes of host lipids. However, the mechanisms of interaction employed by AgB to perform its functions remain undetermined. In this study, we demonstrate that mammalian cells are able to internalize E. granulosus AgB in culture and found that specific mechanisms of endocytosis are involved. Our results extend the understanding of AgB biological role indicating cellular internalization as a mechanism of interaction, which in turn, may represent a target to intervention.
Collapse
Affiliation(s)
- Edileuza Danieli da Silva
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Martin Cancela
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Karina Mariante Monteiro
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Henrique Bunselmeyer Ferreira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Arnaldo Zaha
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- * E-mail:
| |
Collapse
|
39
|
Orbegozo-Medina RA, Martínez-Sernández V, González-Warleta M, Castro-Hermida JA, Mezo M, Ubeira FM. Vaccination of sheep with Quil-A® adjuvant expands the antibody repertoire to the Fasciola MF6p/FhHDM-1 antigen and administered together impair the growth and antigen release of flukes. Vaccine 2018. [DOI: 10.1016/j.vaccine.2018.02.115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
40
|
Schwartz C, Hams E, Fallon PG. Helminth Modulation of Lung Inflammation. Trends Parasitol 2018; 34:388-403. [PMID: 29339033 DOI: 10.1016/j.pt.2017.12.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/11/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022]
Abstract
Parasitic helminths must establish chronic infections to complete their life cycle and therefore are potent modulators of multiple facets of host physiology. Parasitic helminths have coevolved with humans to become arguably master selectors of our immune system, whereby they have impacted on the selection of genes with beneficial mutations for both host and parasite. While helminth infections of humans are a significant health burden, studies have shown that helminths or helminth products can alter susceptibility to unrelated infectious or inflammatory diseases. This has generated interest in the use of helminth infections or molecules as therapeutics. In this review, we focus on the impact of helminth infections on pulmonary immunity, especially with regard to homeostatic lung function, pulmonary viral and bacterial (co)infections, and asthma.
Collapse
Affiliation(s)
- Christian Schwartz
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| | - Emily Hams
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Padraic G Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
41
|
Cwiklinski K, Jewhurst H, McVeigh P, Barbour T, Maule AG, Tort J, O'Neill SM, Robinson MW, Donnelly S, Dalton JP. Infection by the Helminth Parasite Fasciola hepatica Requires Rapid Regulation of Metabolic, Virulence, and Invasive Factors to Adjust to Its Mammalian Host. Mol Cell Proteomics 2018; 17:792-809. [PMID: 29321187 PMCID: PMC5880117 DOI: 10.1074/mcp.ra117.000445] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
The parasite Fasciola hepatica infects a broad range of mammals with
impunity. Following ingestion of parasites (metacercariae) by the host, newly
excysted juveniles (NEJ) emerge from their cysts, rapidly penetrate the duodenal wall
and migrate to the liver. Successful infection takes just a few hours and involves
negotiating hurdles presented by host macromolecules, tissues and micro-environments,
as well as the immune system. Here, transcriptome and proteome analysis of ex
vivo F. hepatica metacercariae and NEJ reveal the rapidity and multitude
of metabolic and developmental alterations that take place in order for the parasite
to establish infection. We found that metacercariae despite being encased in a cyst
are metabolically active, and primed for infection. Following excystment, NEJ expend
vital energy stores and rapidly adjust their metabolic pathways to cope with their
new and increasingly anaerobic environment. Temperature increases induce neoblast
proliferation and the remarkable up-regulation of genes associated with growth and
development. Cysteine proteases synthesized by gastrodermal cells are secreted to
facilitate invasion and tissue degradation, and tegumental transporters, such as
aquaporins, are varied to deal with osmotic/salinity changes. Major proteins of the
total NEJ secretome include proteases, protease inhibitors and anti-oxidants, and an
array of immunomodulators that likely disarm host innate immune effector cells. Thus,
the challenges of infection by F. hepatica parasites are met by
rapid metabolic and physiological adjustments that expedite tissue invasion and
immune evasion; these changes facilitate parasite growth, development and maturation.
Our molecular analysis of the critical processes involved in host invasion has
identified key targets for future drug and vaccine strategies directed at preventing
parasite infection.
Collapse
Affiliation(s)
- Krystyna Cwiklinski
- From the ‡School of Biological Sciences, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK;
| | - Heather Jewhurst
- From the ‡School of Biological Sciences, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Paul McVeigh
- From the ‡School of Biological Sciences, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK.,§Institute for Global Food Security (IGFS), Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Tara Barbour
- From the ‡School of Biological Sciences, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Aaron G Maule
- From the ‡School of Biological Sciences, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK.,§Institute for Global Food Security (IGFS), Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Jose Tort
- ¶Departamento de Genética, Facultad de Medicina, Universidad de la República, Uruguay
| | | | - Mark W Robinson
- From the ‡School of Biological Sciences, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK.,§Institute for Global Food Security (IGFS), Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Sheila Donnelly
- **The i3 Institute and School of Medical and Molecular Biosciences, University of Technology, Sydney, Australia
| | - John P Dalton
- From the ‡School of Biological Sciences, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK.,§Institute for Global Food Security (IGFS), Queen's University Belfast, Belfast, Northern Ireland, UK
| |
Collapse
|
42
|
Navarro S, Pickering DA, Ferreira IB, Jones L, Ryan S, Troy S, Leech A, Hotez PJ, Zhan B, Laha T, Prentice R, Sparwasser T, Croese J, Engwerda CR, Upham JW, Julia V, Giacomin PR, Loukas A. Hookworm recombinant protein promotes regulatory T cell responses that suppress experimental asthma. Sci Transl Med 2017; 8:362ra143. [PMID: 27797959 DOI: 10.1126/scitranslmed.aaf8807] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/01/2016] [Indexed: 12/12/2022]
Abstract
In the developed world, declining prevalence of some parasitic infections correlates with increased incidence of allergic and autoimmune disorders. Moreover, experimental human infection with some parasitic worms confers protection against inflammatory diseases in phase 2 clinical trials. Parasitic worms manipulate the immune system by secreting immunoregulatory molecules that offer promise as a novel therapeutic modality for inflammatory diseases. We identify a protein secreted by hookworms, anti-inflammatory protein-2 (AIP-2), that suppressed airway inflammation in a mouse model of asthma, reduced expression of costimulatory markers on human dendritic cells (DCs), and suppressed proliferation ex vivo of T cells from human subjects with house dust mite allergy. In mice, AIP-2 was primarily captured by mesenteric CD103+ DCs and suppression of airway inflammation was dependent on both DCs and Foxp3+ regulatory T cells (Tregs) that originated in the mesenteric lymph nodes (MLNs) and accumulated in distant mucosal sites. Transplantation of MLNs from AIP-2-treated mice into naïve hosts revealed a lymphoid tissue conditioning that promoted Treg induction and long-term maintenance. Our findings indicate that recombinant AIP-2 could serve as a novel curative therapeutic for allergic asthma and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Severine Navarro
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.
| | - Darren A Pickering
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Ivana B Ferreira
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Linda Jones
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Stephanie Ryan
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Sally Troy
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Andrew Leech
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | | | - Bin Zhan
- Baylor College of Medicine, Houston, TX 77030, USA
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Roger Prentice
- Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - John Croese
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.,Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | | | - John W Upham
- University of Queensland, Brisbane, Queensland, Australia.,Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Valerie Julia
- CNRS UMR7275, INSERM U1080, Université de Nice Sophia Antipolis, Nice, France
| | - Paul R Giacomin
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.
| |
Collapse
|
43
|
Novel Therapeutics for Multiple Sclerosis Designed by Parasitic Worms. Int J Mol Sci 2017; 18:ijms18102141. [PMID: 29027962 PMCID: PMC5666823 DOI: 10.3390/ijms18102141] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 12/17/2022] Open
Abstract
The evolutionary response to endemic infections with parasitic worms (helminth) was the development of a distinct regulatory immune profile arising from the need to encapsulate the helminths while simultaneously repairing tissue damage. According to the old friend's hypothesis, the diminished exposure to these parasites in the developed world has resulted in a dysregulated immune response that contributes to the increased incidence of immune mediated diseases such as Multiple Sclerosis (MS). Indeed, the global distribution of MS shows an inverse correlation to the prevalence of helminth infection. On this basis, the possibility of treating MS with helminth infection has been explored in animal models and phase 1 and 2 human clinical trials. However, the possibility also exists that the individual immune modulatory molecules secreted by helminth parasites may offer a more defined therapeutic strategy.
Collapse
|
44
|
Martínez-Sernández V, Mezo M, González-Warleta M, Perteguer MJ, Gárate T, Romarís F, Ubeira FM. Delineating distinct heme-scavenging and -binding functions of domains in MF6p/helminth defense molecule (HDM) proteins from parasitic flatworms. J Biol Chem 2017; 292:8667-8682. [PMID: 28348084 DOI: 10.1074/jbc.m116.771675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/17/2017] [Indexed: 12/27/2022] Open
Abstract
MF6p/FhHDM-1 is a small protein secreted by the parasitic flatworm (trematode) Fasciola hepatica that belongs to a broad family of heme-binding proteins (MF6p/helminth defense molecules (HDMs)). MF6p/HDMs are of interest for understanding heme homeostasis in trematodes and as potential targets for the development of new flukicides. Moreover, interest in these molecules has also increased because of their immunomodulatory properties. Here we have extended our previous findings on the mechanism of MF6p/HDM-heme interactions and mapped the protein regions required for heme binding and for other biological functions. Our data revealed that MF6p/FhHDM-1 forms high-molecular-weight complexes when associated with heme and that these complexes are reorganized by a stacking procedure to form fibril-like and granular nanostructures. Furthermore, we showed that MF6p/FhHDM-1 is a transitory heme-binding protein as protein·heme complexes can be disrupted by contact with an apoprotein (e.g. apomyoglobin) with higher affinity for heme. We also demonstrated that (i) the heme-binding region is located in the MF6p/FhHDM-1 C-terminal moiety, which also inhibits the peroxidase-like activity of heme, and (ii) MF6p/HDMs from other trematodes, such as Opisthorchis viverrini and Paragonimus westermani, also bind heme. Finally, we observed that the N-terminal, but not the C-terminal, moiety of MF6p/HDMs has a predicted structural analogy with cell-penetrating peptides and that both the entire protein and the peptide corresponding to the N-terminal moiety of MF6p/FhHDM-1 interact in vitro with cell membranes in hemin-preconditioned erythrocytes. Our findings suggest that MF6p/HDMs can transport heme in trematodes and thereby shield the parasite from the harmful effects of heme.
Collapse
Affiliation(s)
- Victoria Martínez-Sernández
- From the Laboratorio de Parasitología, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Mercedes Mezo
- the Laboratorio de Parasitología, Centro de Investigaciones Agrarias de Mabegondo, Instituto Galego da Calidade Alimentaria (INGACAL), 15318 Abegondo, A Coruña, Spain, and
| | - Marta González-Warleta
- the Laboratorio de Parasitología, Centro de Investigaciones Agrarias de Mabegondo, Instituto Galego da Calidade Alimentaria (INGACAL), 15318 Abegondo, A Coruña, Spain, and
| | - María J Perteguer
- the Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Teresa Gárate
- the Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Fernanda Romarís
- From the Laboratorio de Parasitología, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Florencio M Ubeira
- From the Laboratorio de Parasitología, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain,
| |
Collapse
|
45
|
Lund ME, Greer J, Dixit A, Alvarado R, McCauley-Winter P, To J, Tanaka A, Hutchinson AT, Robinson MW, Simpson AM, O'Brien BA, Dalton JP, Donnelly S. A parasite-derived 68-mer peptide ameliorates autoimmune disease in murine models of Type 1 diabetes and multiple sclerosis. Sci Rep 2016; 6:37789. [PMID: 27883079 PMCID: PMC5121616 DOI: 10.1038/srep37789] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 11/02/2016] [Indexed: 12/16/2022] Open
Abstract
Helminth parasites secrete molecules that potently modulate the immune responses of their hosts and, therefore, have potential for the treatment of immune-mediated human diseases. FhHDM-1, a 68-mer peptide secreted by the helminth parasite Fasciola hepatica, ameliorated disease in two different murine models of autoimmunity, type 1 diabetes and relapsing-remitting immune-mediated demyelination. Unexpectedly, FhHDM-1 treatment did not affect the proliferation of auto-antigen specific T cells or their production of cytokines. However, in both conditions, the reduction in clinical symptoms was associated with the absence of immune cell infiltrates in the target organ (islets and the brain tissue). Furthermore, after parenteral administration, the FhHDM-1 peptide interacted with macrophages and reduced their capacity to secrete pro-inflammatory cytokines, such as TNF and IL-6. We propose this inhibition of innate pro-inflammatory immune responses, which are central to the initiation of autoimmunity in both diseases, prevented the trafficking of autoreactive lymphocytes from the periphery to the site of autoimmunity (as opposed to directly modulating their function per se), and thus prevented tissue destruction. The ability of FhHDM-1 to modulate macrophage function, combined with its efficacy in disease prevention in multiple models, suggests that FhHDM-1 has considerable potential as a treatment for autoimmune diseases.
Collapse
Affiliation(s)
- Maria E Lund
- The School of Life Sciences, University of Technology Sydney, New South Wales, Australia
| | - Judith Greer
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Aakanksha Dixit
- The University of Queensland, UQ Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Raquel Alvarado
- The School of Life Sciences, University of Technology Sydney, New South Wales, Australia
| | | | - Joyce To
- The School of Life Sciences, University of Technology Sydney, New South Wales, Australia
| | - Akane Tanaka
- The School of Life Sciences, University of Technology Sydney, New South Wales, Australia
| | - Andrew T Hutchinson
- The School of Life Sciences, University of Technology Sydney, New South Wales, Australia.,The Centre for Health Technology, University of Technology Sydney, New South Wales, Australia
| | - Mark W Robinson
- Medical Biology Center, School of Biological Sciences, Queen's University, Belfast, Northern Ireland, United Kingdom
| | - Ann M Simpson
- The School of Life Sciences, University of Technology Sydney, New South Wales, Australia.,The Centre for Health Technology, University of Technology Sydney, New South Wales, Australia
| | - Bronwyn A O'Brien
- The School of Life Sciences, University of Technology Sydney, New South Wales, Australia.,The Centre for Health Technology, University of Technology Sydney, New South Wales, Australia
| | - John P Dalton
- Medical Biology Center, School of Biological Sciences, Queen's University, Belfast, Northern Ireland, United Kingdom
| | - Sheila Donnelly
- The School of Life Sciences, University of Technology Sydney, New South Wales, Australia
| |
Collapse
|
46
|
Liu Q, Huang SY, Yue DM, Wang JL, Wang Y, Li X, Zhu XQ. Proteomic analysis of Fasciola hepatica excretory and secretory products (FhESPs) involved in interacting with host PBMCs and cytokines by shotgun LC-MS/MS. Parasitol Res 2016; 116:627-635. [PMID: 27866265 DOI: 10.1007/s00436-016-5327-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/08/2016] [Indexed: 11/25/2022]
Abstract
Fasciola hepatica is a helminth parasite with a worldwide distribution, which can cause chronic liver disease, fasciolosis, leading to economic losses in the livestock and public health in many countries. Control is mostly reliant on the use of drugs, and as a result, drug resistance has now emerged. The identification of F. hepatica genes involved in interaction between the parasite and host immune system is utmost important to elucidate the evasion mechanisms of the parasite and develop more effective strategies against fasciolosis. In this study, we aimed to identify molecules in F. hepatica excretory and secretory products (FhESPs) interacting with the host peripheral blood mononuclear cells (PBMCs), Th1-like cytokines (IL2 and IFN-γ), and Th17-like cytokines (IL17) by Co-IP combined with tandem mass spectrometry. The results showed that 14, 16, and 9 proteins in FhESPs could bind with IL2, IL17, and IFN-γ, respectively, which indicated that adult F. hepatica may evade the host immune responses through directly interplaying with cytokines. In addition, nine proteins in FhESPs could adhere to PBMCs. Our findings provided potential targets as immuno-regulators, and will be helpful to elucidate the molecular basis of host-parasite interactions and search for new potential proteins as vaccine and drug target candidates.
Collapse
Affiliation(s)
- Qing Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, People's Republic of China
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, 410128, People's Republic of China
| | - Si-Yang Huang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, People's Republic of China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China.
| | - Dong-Mei Yue
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, People's Republic of China
| | - Jin-Lei Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, People's Republic of China
| | - Yujian Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Xiangrui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China
| |
Collapse
|
47
|
Alvarado R, To J, Lund ME, Pinar A, Mansell A, Robinson MW, O'Brien BA, Dalton JP, Donnelly S. The immune modulatory peptide FhHDM-1 secreted by the helminth Fasciola hepatica prevents NLRP3 inflammasome activation by inhibiting endolysosomal acidification in macrophages. FASEB J 2016; 31:85-95. [PMID: 27682204 DOI: 10.1096/fj.201500093r] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 09/16/2016] [Indexed: 12/31/2022]
Abstract
The NLRP3 inflammasome is a multimeric protein complex that controls the production of IL-1β, a cytokine that influences the development of both innate and adaptive immune responses. Helminth parasites secrete molecules that interact with innate immune cells, modulating their activity to ultimately determine the phenotype of differentiated T cells, thus creating an immune environment that is conducive to sustaining chronic infection. We show that one of these molecules, FhHDM-1, a cathelicidin-like peptide secreted by the helminth parasite, Fasciola hepatica, inhibits the activation of the NLRP3 inflammasome resulting in reduced secretion of IL-1β by macrophages. FhHDM-1 had no effect on the synthesis of pro-IL-1β. Rather, the inhibitory effect was associated with the capacity of the peptide to prevent acidification of the endolysosome. The activation of cathepsin B protease by lysosomal destabilization was prevented in FhHDM-1-treated macrophages. By contrast, peptide derivatives of FhHDM-1 that did not alter the lysosomal pH did not inhibit secretion of IL-1β. We propose a novel immune modulatory strategy used by F. hepatica, whereby secretion of the FhHDM-1 peptide impairs the activation of NLRP3 by lysosomal cathepsin B protease, which prevents the downstream production of IL-1β and the development of protective T helper 1 type immune responses that are detrimental to parasite survival.-Alvarado, R., To, J., Lund, M. E., Pinar, A., Mansell, A., Robinson, M. W., O'Brien, B. A., Dalton, J. P., Donnelly, S. The immune modulatory peptide FhHDM-1 secreted by the helminth Fasciola hepatica prevents NLRP3 inflammasome activation by inhibiting endolysosomal acidification in macrophages.
Collapse
Affiliation(s)
- Raquel Alvarado
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Joyce To
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Maria E Lund
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Anita Pinar
- Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia; and
| | - Ashley Mansell
- Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia; and
| | - Mark W Robinson
- School of Biological Sciences, Queen's University, Belfast, Northern Ireland
| | - Bronwyn A O'Brien
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - John P Dalton
- School of Biological Sciences, Queen's University, Belfast, Northern Ireland
| | - Sheila Donnelly
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia;
| |
Collapse
|
48
|
Maizels RM, McSorley HJ. Regulation of the host immune system by helminth parasites. J Allergy Clin Immunol 2016; 138:666-675. [PMID: 27476889 PMCID: PMC5010150 DOI: 10.1016/j.jaci.2016.07.007] [Citation(s) in RCA: 363] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 01/01/2023]
Abstract
Helminth parasite infections are associated with a battery of immunomodulatory mechanisms that affect all facets of the host immune response to ensure their persistence within the host. This broad-spectrum modulation of host immunity has intended and unintended consequences, both advantageous and disadvantageous. Thus the host can benefit from suppression of collateral damage during parasite infection and from reduced allergic, autoimmune, and inflammatory reactions. However, helminth infection can also be detrimental in reducing vaccine responses, increasing susceptibility to coinfection and potentially reducing tumor immunosurveillance. In this review we will summarize the panoply of immunomodulatory mechanisms used by helminths, their potential utility in human disease, and prospective areas of future research.
Collapse
Affiliation(s)
- Rick M Maizels
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.
| | - Henry J McSorley
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
| |
Collapse
|
49
|
Smith D, Tikhonova IG, Jewhurst HL, Drysdale OC, Dvořák J, Robinson MW, Cwiklinski K, Dalton JP. Unexpected Activity of a Novel Kunitz-type Inhibitor: INHIBITION OF CYSTEINE PROTEASES BUT NOT SERINE PROTEASES. J Biol Chem 2016; 291:19220-34. [PMID: 27422822 PMCID: PMC5016662 DOI: 10.1074/jbc.m116.724344] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Indexed: 12/15/2022] Open
Abstract
Kunitz-type (KT) protease inhibitors are low molecular weight proteins classically defined as serine protease inhibitors. We identified a novel secreted KT inhibitor associated with the gut and parenchymal tissues of the infective juvenile stage of Fasciola hepatica, a helminth parasite of medical and veterinary importance. Unexpectedly, recombinant KT inhibitor (rFhKT1) exhibited no inhibitory activity toward serine proteases but was a potent inhibitor of the major secreted cathepsin L cysteine proteases of F. hepatica, FhCL1 and FhCL2, and of human cathepsins L and K (Ki = 0.4-27 nm). FhKT1 prevented the auto-catalytic activation of FhCL1 and FhCL2 and formed stable complexes with the mature enzymes. Pulldown experiments from adult parasite culture medium showed that rFhKT1 interacts specifically with native secreted FhCL1, FhCL2, and FhCL5. Substitution of the unusual P1 Leu15 within the exposed reactive loop of FhKT1 for the more commonly found Arg (FhKT1Leu15/Arg15) had modest adverse effects on the cysteine protease inhibition but conferred potent activity against the serine protease trypsin (Ki = 1.5 nm). Computational docking and sequence analysis provided hypotheses for the exclusive binding of FhKT1 to cysteine proteases, the importance of the Leu15 in anchoring the inhibitor into the S2 active site pocket, and the inhibitor's selectivity toward FhCL1, FhCL2, and human cathepsins L and K. FhKT1 represents a novel evolutionary adaptation of KT protease inhibitors by F. hepatica, with its prime purpose likely in the regulation of the major parasite-secreted proteases and/or cathepsin L-like proteases of its host.
Collapse
Affiliation(s)
| | - Irina G Tikhonova
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, United Kingdom
| | | | | | - Jan Dvořák
- From the School of Biological Sciences and
| | | | | | | |
Collapse
|
50
|
Korhonen PK, Pozio E, La Rosa G, Chang BCH, Koehler AV, Hoberg EP, Boag PR, Tan P, Jex AR, Hofmann A, Sternberg PW, Young ND, Gasser RB. Phylogenomic and biogeographic reconstruction of the Trichinella complex. Nat Commun 2016; 7:10513. [PMID: 26830005 PMCID: PMC4740406 DOI: 10.1038/ncomms10513] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 12/18/2015] [Indexed: 01/21/2023] Open
Abstract
Trichinellosis is a globally important food-borne parasitic disease of humans caused by roundworms of the Trichinella complex. Extensive biological diversity is reflected in substantial ecological and genetic variability within and among Trichinella taxa, and major controversy surrounds the systematics of this complex. Here we report the sequencing and assembly of 16 draft genomes representing all 12 recognized Trichinella species and genotypes, define protein-coding gene sets and assess genetic differences among these taxa. Using thousands of shared single-copy orthologous gene sequences, we fully reconstruct, for the first time, a phylogeny and biogeography for the Trichinella complex, and show that encapsulated and non-encapsulated Trichinella taxa diverged from their most recent common ancestor ∼21 million years ago (mya), with taxon diversifications commencing ∼10-7 mya.
Collapse
Affiliation(s)
- Pasi K Korhonen
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Edoardo Pozio
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Giuseppe La Rosa
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Bill C H Chang
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia.,Yourgene Bioscience, Shu-Lin District, New Taipei City 23863, Taiwan
| | - Anson V Koehler
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Eric P Hoberg
- United States National Parasite Collection, US Department of Agriculture, Agricultural Research Service, Beltsville, Maryland 20705, USA
| | - Peter R Boag
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Patrick Tan
- Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Republic of Singapore.,Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 138672, Republic of Singapore
| | - Aaron R Jex
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Andreas Hofmann
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia.,Structural Chemistry Program, Eskitis Institute, Griffith University, Brisbane, Queensland 4111, Australia
| | - Paul W Sternberg
- Division of Biology, Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California 91125, USA
| | - Neil D Young
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Robin B Gasser
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|