1
|
Thammasonthijarern N, Boonnak K, Reamtong O, Krasae T, Thankansakul J, Phongphaew W, Ampawong S, Adisakwattana P. Amelioration of ovalbumin-induced lung inflammation in a mouse model by Trichinella spiralis novel cystatin. Vet World 2023; 16:2366-2373. [PMID: 38152266 PMCID: PMC10750734 DOI: 10.14202/vetworld.2023.2366-2373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/23/2023] [Indexed: 12/29/2023] Open
Abstract
Background and Aims Asthma, a chronic disease affecting humans and animals, has recently become increasingly prevalent and steadily widespread. The alternative treatment of asthma using helminth infections or helminth-derived immunomodulatory molecules (IMs) has been evaluated and demonstrated significant amelioration of disease severity index in vitro and in vivo. Trichinella spiralis, a parasitic nematode and its IMs, elicits a potential to relieve asthma and other immune-related disorders. In this study, we investigated the immunomodulatory function of recombinant T. spiralis novel cystatin (rTsCstN) in ameliorating acute inflammatory asthma disorders in a murine model. Materials and Methods Female BALB/c mice were sensitized using intraperitoneal injection of ovalbumin (OVA)/alum and subsequently challenged with intranasal administration of OVA alone or OVA + rTsCstN for 3 consecutive days, producing OVA-induced allergic asthma models. To evaluate the therapeutic efficacy of rTsCstN, the inflammatory cells and cytokines in bronchoalveolar lavage fluid (BALF) and OVA-specific immunoglobulin E levels in serum were assessed. Histological alterations in the lung tissues were determined by hematoxylin and eosin (H&E) staining and eventually scored for the extent of inflammatory cell infiltration. Results The asthmatic mouse models challenged with OVA + rTsCstN demonstrated a significant reduction of eosinophils (p < 0.01), macrophages (p < 0.05), and cytokines tumor necrosis factor-α (p < 0.05) and interferon (IFN)-γ (p < 0.05) in BALF when compared with the mice challenged with OVA alone. However, the levels of interleukin (IL)-4 and IL-10 remained unchanged. Histological examination revealed that mice administered OVA + rTsCstN were less likely to have inflammatory cell infiltration in their perivascular and peribronchial lung tissues than those administered OVA alone. Conclusion Recombinant T. spiralis novel cystatin demonstrated immunomodulatory effects to reduce severe pathogenic alterations in asthma mouse models, encouraging a viable alternative treatment for asthma and other immunoregulatory disorders in humans and animals in the future.
Collapse
Affiliation(s)
- Nipa Thammasonthijarern
- Department of Parasitology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Kobporn Boonnak
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Thanyaluk Krasae
- Laboratory Animal Science Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Janyaporn Thankansakul
- Kasetsart University Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Wallaya Phongphaew
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Li Z, Wang X, Zhang W, Yang W, Xu B, Hu W. Excretory/Secretory Products from Schistosoma japonicum Eggs Alleviate Ovalbumin-Induced Allergic Airway Inflammation. PLoS Negl Trop Dis 2023; 17:e0011625. [PMID: 37788409 PMCID: PMC10547495 DOI: 10.1371/journal.pntd.0011625] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
INTRODUCTION Excretory/secretory products (ESPs) derived from helminths have been reported to effectively control allergic inflammation, which have better therapeutic prospects than live parasite infections. However, it remains unknown whether ESPs from schistosome eggs can protect against allergies, despite reports alleging that schistosome infection could alleviate disordered allergic inflammation. METHOD In the present study, we investigated the protective effects of ESPs from Schistosoma japonicum eggs (ESP-SJE) on asthmatic inflammation. Firstly, we successfully established an allergic airway inflammation model in mice by alum-adjuvanted ovalbumin (OVA) sensitization and challenge. ESP-SJE were administered intraperitoneally on days -1 and 13 (before sensitization), on day 20 (before challenge), and on days 21-24 (challenge phase). RESULTS The results showed that ESP-SJE treatment significantly reduced the infiltration of inflammatory cells, especially eosinophils into the lung tissue, inhibited the production of the total and OVA-specific IgE during OVA-sensitized and -challenged phases, respectively, and suppressed the secretion of Th2-type inflammatory cytokines (IL-4). Additionally, ESP-SJE treatment significantly upregulated the regulatory T cells (Tregs) in the lung tissue during OVA challenge. Furthermore, using liquid chromatography-mass spectrometry analysis and Treg induction experiments in vitro, we might identify nine potential therapeutic proteins against allergic inflammation in ESP-SJE. The targets of these candidate proteins included glutathione S-transferase, egg protein CP422 precursor, tubulin alpha-2/alpha-4 chain, actin-2, T-complex protein 1 subunit beta, histone H₄, whey acidic protein core region, and molecular chaperone HtpG. CONCLUSION Taken together, the results discussed herein demonstrated that ESP-SJE could significantly alleviate OVA-induced asthmatic inflammation in a murine model, which might be mediated by the upregulation of Treg in lung tissues that may be induced by the potential modulatory proteins. Therefore, potential proteins in ESP-SJE might be the best candidates to be tested for therapeutic application of asthma, thus pointing out to a possible new therapy for allergic airway inflammation.
Collapse
Affiliation(s)
- Zhidan Li
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, P. R. China
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
| | - Xiaoling Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Wenbin Yang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Bin Xu
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
| | - Wei Hu
- National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Centre for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, China
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- College of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
3
|
Immunomodulating Hydrogels as Stealth Platform for Drug Delivery Applications. Pharmaceutics 2022; 14:pharmaceutics14102244. [PMID: 36297679 PMCID: PMC9610165 DOI: 10.3390/pharmaceutics14102244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/23/2022] [Accepted: 10/05/2022] [Indexed: 11/23/2022] Open
Abstract
Non-targeted persistent immune activation or suppression by different drug delivery platforms can cause adverse and chronic physiological effects including cancer and arthritis. Therefore, non-toxic materials that do not trigger an immunogenic response during delivery are crucial for safe and effective in vivo treatment. Hydrogels are excellent candidates that can be engineered to control immune responses by modulating biomolecule release/adsorption, improving regeneration of lymphoid tissues, and enhancing function during antigen presentation. This review discusses the aspects of hydrogel-based systems used as drug delivery platforms for various diseases. A detailed investigation on different immunomodulation strategies for various delivery options and deliberate upon the outlook of such drug delivery platforms are conducted.
Collapse
|
4
|
Kondo Y, Ito D, Tademoto S, Itami N, Nishikata S, Takashima E, Tsuboi T, Fukumoto S, Otsuki H. Molecular cloning and characterization of plerocercoid-immunosuppressive factor from Spirometra erinaceieuropaei. Parasitol Int 2020; 76:102062. [PMID: 31978597 DOI: 10.1016/j.parint.2020.102062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/14/2020] [Accepted: 01/17/2020] [Indexed: 01/15/2023]
Abstract
A platyhelminth, Spirometra erinaceieuropaei, belonging to the class Cestoda, causes human sparganosis, and infection with its larva results in subtle inflammation in the body of its host. We previously reported the purification of a glycoprotein, plerocercoid-immunosuppressive factor (P-ISF) from the excretory/secretory products of S. erinaceieuropaei plerocercoids that may be involved in immuno-modification. We determined the sequence of P-ISF from the N-terminal and the internal 10 amino acids of P-ISF using degenerate PCR and 5'- and 3'-RACE methods. The putative gene encoding P-ISF was 1443 bp long and the gene contained 10 exons and 9 introns in a genomic DNA of size 5205 bp. P-ISF consists of 480 amino acids including the N-terminal signal peptide sequence, and has two unknown domains,-cestoda cysteine-rich domains (CCDs) and a fibronectin type III domain between the two CCDs. All cysteine residues were conserved in the two CCDs, which shared 62% amino acid identities. Homologous analysis revealed that the CCDs were homologous with an unknown protein of Diphyllobothrium latum. To produce specific antibodies, we expressed recombinant P-ISF (rP-ISF) using wheat germ protein synthetic system. P-ISF was localized in the sub-cutaneous tissues and the parenchymal tissues of plerocercoids. Transcription of P-ISF was detected only in plerocercoid stage, but not in adult stage. Western blotting also showed a band in plerocercoide stage but not in adult. The rP-ISF did not suppress nitrite production in RAW 264.7 cells stimulated with LPS, and this might be due to lack of carbohydrate chains in the recombinant protein.
Collapse
Affiliation(s)
- Yoko Kondo
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Daisuke Ito
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Sayuri Tademoto
- Technical Department, Tottori University, Yonago 683-8503, Japan
| | - Nanase Itami
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Shuma Nishikata
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan
| | - Soji Fukumoto
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; Tottori Medical Career Support Center, Tottori University Hospital, Yonago 683-8504, Japan
| | - Hitoshi Otsuki
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan.
| |
Collapse
|
5
|
Alexandre-Silva GM, Brito-Souza PA, Oliveira AC, Cerni FA, Zottich U, Pucca MB. The hygiene hypothesis at a glance: Early exposures, immune mechanism and novel therapies. Acta Trop 2018; 188:16-26. [PMID: 30165069 DOI: 10.1016/j.actatropica.2018.08.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/20/2018] [Accepted: 08/25/2018] [Indexed: 02/07/2023]
Abstract
The hygiene hypothesis was proposed almost three decades ago. Nevertheless, its mechanism still remains with relevant controversies. Some studies defend that early exposures during childhood to microbes and parasites are key determinants to prevent allergies and autoimmune diseases; however, other studies demonstrated that these early exposures can even potentiate the clinical scenario of the diseases. Based on several studies covering the influences of microbiome, parasites, related theories and others, this review focuses on recent advances in the hygiene hypothesis field. In addition, the main immunological mechanisms underlying the hygiene hypothesis are also discussed. We also strongly encourage that researchers do not consider the hygiene hypothesis as a theory based strictly on hygiene habits, but a theory combining diverse influences, as illustrated in this review as the hygiene hypothesis net.
Collapse
|
6
|
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: 14.9] [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
|
7
|
Floudas A, Cluxton CD, Fahel J, Khan AR, Saunders SP, Amu S, Alcami A, Fallon PG. Composition of the Schistosoma mansoni worm secretome: Identification of immune modulatory Cyclophilin A. PLoS Negl Trop Dis 2017; 11:e0006012. [PMID: 29073139 PMCID: PMC5681295 DOI: 10.1371/journal.pntd.0006012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/10/2017] [Accepted: 10/04/2017] [Indexed: 12/16/2022] Open
Abstract
The helminth Schistosoma mansoni modulates the infected host's immune system to facilitate its own survival, by producing excretory/secretory molecules that interact with a variety of the host's cell types including those of the immune system. Herein, we characterise the S. mansoni adult male worm secretome and identify 111 proteins, including 7 vaccine candidates and several molecules with potential immunomodulatory activity. Amongst the molecules present in the secretome, a 17-19kDa protein analogous to human cyclophilin A was identified. Given the ability of cyclophilin A to modulate the immune system by regulating antigen presenting cell activity, we sought to determine whether recombinant S. mansoni Cyclophilin A (rSmCypA) is capable of modulating bone-marrow derived dendritic cell (BMDC) and T cell responses under in vitro conditions. rSmCypA was enzymatically active and able to alter the pro-inflammatory cytokine profile of LPS-activated dendritic cells. rSmCypA also modulated DC function in the induction of CD4+ T cell proliferation with a preferential expansion of Treg cells. This work demonstrates the unique protein composition of the S. mansoni male worm secretome and immunomodulatory activity of S. mansoni Cyclophilin A.
Collapse
Affiliation(s)
- Achilleas Floudas
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Christopher D. Cluxton
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Julia Fahel
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Adnan R. Khan
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sean P. Saunders
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sylvie Amu
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Antonio Alcami
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Padraic G. Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- * E-mail:
| |
Collapse
|
8
|
Abstract
MS incidence has significantly increased during the second half of the 20th century, generating considerable interest in analyzing the basis for this rise in the developed world. Particular emphasis is being placed on the role infections might play in exacerbating or preventing disease onset. Epidemiological data suggest that improvement in sanitation conditions and reduced exposure to infection might explain, at least in part, these changes. The hygiene hypothesis is not new and is currently used to explain the increasing incidence of allergies and other autoimmune diseases. Because helminths are powerful modulators of host immunity, some authors hypothesize that reduced parasite exposure due to improved hygiene conditions may favor MS development. We discuss epidemiological, experimental, clinical and molecular data supporting the protective role of helminthes against MS. Better understanding of host–parasite interactions caused by specific parasite molecules with immunomodulatory effects will help combat allergies and autoimmune disease without the price of untoward infection as a side-effect.
Collapse
|
9
|
Wang X, Wang J, Liang Y, Ni H, Shi L, Xu C, Zhou Y, Su Y, Mou X, Chen D, Mao C. Schistosoma japonicum HSP60-derived peptide SJMHE1 suppresses delayed-type hypersensitivity in a murine model. Parasit Vectors 2016; 9:147. [PMID: 26971312 PMCID: PMC4789290 DOI: 10.1186/s13071-016-1434-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/05/2016] [Indexed: 12/20/2022] Open
Abstract
Background Parasite-derived molecules with immunomodulatory properties, which have been optimised during host-parasite co-evolution, exhibit potential applications as novel immunotherapeutics. We have previously demonstrated that Schistosoma japonicum HSP60-derived peptide SJMHE1 induces CD4+CD25+ regulatory T-cells (Tregs) and that adoptively transferred SJMHE1-induced CD4+CD25+ Tregs inhibit delayed-type hypersensitivity (DTH) in mice. However, multiple concerns regarding this method render this treatment unsuitable. To gain further insights into the potential effects of SJMHE1, we used ovalbumin (OVA)-induced DTH and evaluated the effect of SJMHE1 on DTH mice. Methods BALB/c mice were sensitised with OVA alone or combined with SJMHE1 and then challenged with OVA to induce DTH. We first analysed the potential effects of SJMHE1 by measuring DTH responses, T-cell responses, cytokine secretion, and Treg proportions. We then evaluated the expression levels of IL-10 and TGF-β1 in CD4+CD25+ T-cells during DTH and Treg generation to identify the mechanism by which SJMHE1 suppresses DTH. Results SJMHE1 modulated the effector response against OVA-induced DTH and stimulated the production of the anti-inflammatory cytokines IL-10 and TGF-β1 in immunised mice through a mechanism involving CD4+CD25+ Tregs. SJMHE1-induced CD4+CD25+ Tregs expressed high levels of CTLA-4, IL-10, and TGF-β1, which substantially contributed to the suppressive activity during DTH. The administration of SJMHE1 to DTH in mice led to the expansion of CD4+CD25+ Tregs from CD4+CD25− T-cells in the periphery, which inhibited DTH responses. Conclusions Our study proves that the parasite-driven peptide suppresses DTH in mice, which may confer a new option for inflammation treatment.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China. .,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Jun Wang
- Department of Nuclear Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Yong Liang
- Clinical Laboratory, Huai'an Hospital Affiliated of Xuzhou Medical College, Huaian, Jiangsu, 223300, China
| | - Hongchang Ni
- Department of Nuclear Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Liang Shi
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chengcheng Xu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yuepeng Zhou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yuting Su
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiao Mou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Deyu Chen
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chaoming Mao
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| |
Collapse
|
10
|
Shepherd C, Navarro S, Wangchuk P, Wilson D, Daly NL, Loukas A. Identifying the immunomodulatory components of helminths. Parasite Immunol 2015; 37:293-303. [PMID: 25854639 DOI: 10.1111/pim.12192] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 03/18/2015] [Indexed: 12/13/2022]
Abstract
Immunomodulatory components of helminths offer great promise as an entirely new class of biologics for the treatment of inflammatory diseases. Here, we discuss the emerging themes in helminth-driven immunomodulation in the context of therapeutic drug discovery. We broadly define the approaches that are currently applied by researchers to identify these helminth molecules, highlighting key areas of potential exploitation that have been mostly neglected thus far, notably small molecules. Finally, we propose that the investigation of immunomodulatory compounds will enable the translation of current and future research efforts into potential treatments for autoimmune and allergic diseases, while at the same time yielding new insights into the molecular interface of host-parasite biology.
Collapse
Affiliation(s)
- C Shepherd
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Qld, Australia
| | | | | | | | | | | |
Collapse
|
11
|
Shen SW, Lu Y, Li F, Shen ZH, Xu M, Yao WF, Feng YB, Yun JT, Wang YP, Ling W, Qi HJ, Tong DX. The potential long-term effect of previous schistosome infection reduces the risk of metabolic syndrome among Chinese men. Parasite Immunol 2015; 37:333-9. [PMID: 25809087 DOI: 10.1111/pim.12187] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 03/13/2015] [Indexed: 12/19/2022]
Affiliation(s)
- S.-W. Shen
- Wuxi No. 2 People's Hospital Affiliated to Nanjing Medical University; Wuxi City Jiangsu Province China
| | - Y. Lu
- Jiangsu Provincial Taihu Cadre's Sanatorium of Jiangsu Provincial People's Hospital Group; Wuxi City Jiangsu Province China
| | - F. Li
- Jiangsu Provincial Taihu Cadre's Sanatorium of Jiangsu Provincial People's Hospital Group; Wuxi City Jiangsu Province China
| | - Z.-H. Shen
- Jiangsu Provincial Taihu Cadre's Sanatorium of Jiangsu Provincial People's Hospital Group; Wuxi City Jiangsu Province China
| | - M. Xu
- Jiangsu Institute of Parasitic Diseases; Wuxi City Jiangsu Province China
| | - W.-F. Yao
- Wuxi No. 2 People's Hospital Affiliated to Nanjing Medical University; Wuxi City Jiangsu Province China
| | - Y.-B. Feng
- Jiangsu Provincial Taihu Cadre's Sanatorium of Jiangsu Provincial People's Hospital Group; Wuxi City Jiangsu Province China
| | - J.-T. Yun
- Jiangsu Provincial Taihu Cadre's Sanatorium of Jiangsu Provincial People's Hospital Group; Wuxi City Jiangsu Province China
| | - Y.-P. Wang
- Wuxi No. 2 People's Hospital Affiliated to Nanjing Medical University; Wuxi City Jiangsu Province China
| | - W. Ling
- Jiangsu Provincial Taihu Cadre's Sanatorium of Jiangsu Provincial People's Hospital Group; Wuxi City Jiangsu Province China
| | - H.-J. Qi
- Jiangsu Provincial Taihu Cadre's Sanatorium of Jiangsu Provincial People's Hospital Group; Wuxi City Jiangsu Province China
| | - D.-X. Tong
- Jiangsu Provincial Taihu Cadre's Sanatorium of Jiangsu Provincial People's Hospital Group; Wuxi City Jiangsu Province China
| |
Collapse
|
12
|
Potential long-term effects of previous schistosome infection may reduce the atherogenic index of plasma in Chinese men. Int J Parasitol 2015; 45:289-94. [DOI: 10.1016/j.ijpara.2015.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/25/2014] [Accepted: 01/01/2015] [Indexed: 12/24/2022]
|
13
|
McSorley HJ, Blair NF, Robertson E, Maizels RM. Suppression of OVA-alum induced allergy by Heligmosomoides polygyrus products is MyD88-, TRIF-, regulatory T- and B cell-independent, but is associated with reduced innate lymphoid cell activation. Exp Parasitol 2015; 158:8-17. [PMID: 25728231 DOI: 10.1016/j.exppara.2015.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/09/2015] [Accepted: 02/22/2015] [Indexed: 02/06/2023]
Abstract
The murine intestinal nematode Heligmosomoides polygyrus exerts multiple immunomodulatory effects in the host, including the suppression of allergic inflammation in mice sensitized to allergen presented with alum adjuvant. Similar suppression is attained by co-administration of H. polygyrus excretory/secretory products (HES) with the sensitizing dose of ovalbumin (OVA) in alum. We investigated the mechanism of suppression by HES in this model, and found it was maintained in MyD88xTRIF-deficient mice, implying no role for helminth- or host-derived TLR ligands, or IL-1 family cytokines that signal in a MyD88- or TRIF-dependent manner. We also found suppression was unchanged in µMT mice, which lack B2 B cells, and that suppression was not abrogated when regulatory T cells were depleted in Foxp3.LuciDTR-4 mice. However, reduced IL-5 production was seen in the first 12 h after injection of OVA-alum when HES was co-administered, associated with reduced activation of IL-5(+) and IL-13(+) group 2 innate lymphoid cells. Thus, the suppressive effects of HES on alum-mediated OVA sensitization are reflected in the very earliest innate response to allergen exposure in vivo.
Collapse
Affiliation(s)
- Henry J McSorley
- Institute of Immunology and Infection Research, and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK; Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Natalie F Blair
- Institute of Immunology and Infection Research, and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK
| | - Elaine Robertson
- Institute of Immunology and Infection Research, and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK
| | - Rick M Maizels
- Institute of Immunology and Infection Research, and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK
| |
Collapse
|
14
|
Weinstock JV, Elliott DE. Helminth infections decrease host susceptibility to immune-mediated diseases. THE JOURNAL OF IMMUNOLOGY 2014; 193:3239-47. [PMID: 25240019 DOI: 10.4049/jimmunol.1400927] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Helminthic infection has become rare in highly industrialized nations. Concurrent with the decline in helminthic infection has been an increase in the prevalence of inflammatory disease. Removal of helminths from our environment and their powerful effects on host immunity may have contributed to this increase. Several helminth species can abrogate disease in murine models of inflammatory bowel disease, type 1 diabetes, multiple sclerosis, and other conditions. Helminths evoke immune regulatory pathways often involving dendritic cells, regulatory T cells, and macrophages that help to control disease. Cytokines, such as IL-4, IL-10, and TGF-β, have a role. Notable is the helminthic modulatory effect on innate immunity, which impedes development of aberrant adaptive immunity. Investigators are identifying key helminth-derived immune modulatory molecules that may have therapeutic usefulness in the control of inflammatory disease.
Collapse
Affiliation(s)
- Joel V Weinstock
- Division of Gastroenterology, Tufts Medical Center, Boston, MA 02111; and
| | - David E Elliott
- Division of Gastroenterology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
15
|
Oikonomopoulou K, Brinc D, Hadjisavvas A, Christofi G, Kyriacou K, Diamandis EP. The bifacial role of helminths in cancer: Involvement of immune and non-immune mechanisms. Crit Rev Clin Lab Sci 2014; 51:138-48. [DOI: 10.3109/10408363.2014.886180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
16
|
Khan AR, Amu S, Saunders SP, Fallon PG. The generation of regulatory B cells by helminth parasites. Methods Mol Biol 2014; 1190:143-162. [PMID: 25015279 DOI: 10.1007/978-1-4939-1161-5_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The activation of B cells by pathogens to elicit antibody production is a central facet of immunity. Pathogens also evoke the expansion of B cells that can function in the regulation of immunity. Different pathogens have been described with the capacity to drive such regulatory B (Breg) cells. One group of pathogens, parasitic helminths, has been used experimentally to identify and explore immunological mechanisms of Breg cells. Several species of helminths have demonstrated the capacity to expand Breg cell populations in mice, while the presence of Breg cells in humans infected with certain helminths has also been identified. Herein, we outline how the helminth Schistosoma mansoni can expand Breg cellular responses in vitro. We describe the establishment of a laboratory-based S. mansoni life cycle and methodology for detecting Breg cells via flow cytometry.
Collapse
Affiliation(s)
- Adnan R Khan
- School of Medicine, Trinity Biomedical Sciences Institute, , Trinity College Dublin, College Green, Dublin 2, Ireland
| | | | | | | |
Collapse
|
17
|
Possible use of Trichuris suis ova in autism spectrum disorders therapy. Med Hypotheses 2013; 81:1-4. [PMID: 23597946 DOI: 10.1016/j.mehy.2013.03.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 03/01/2013] [Accepted: 03/17/2013] [Indexed: 12/24/2022]
Abstract
Autism and autism spectrum disorders (ASDs) are heterogeneous, severe neurodevelopmental pathologies. The main core symptoms are: dysfunctions in social interactions and communication skills, restricted interests, repetitive and stereotypic verbal and non-verbal behaviors. Several biochemical processes are associated with ASDs: oxidative stress; endoplasmic reticulum stress; decreased methylation capacity; limited production of glutathione; mitochondrial dysfunction; intestinal dysbiosis; increased toxic metal burden; immune dysregulation. Current available treatments for ASDs can be divided into behavioral, nutritional and medical approaches, although no defined standard approach exists. Current drugs fail to benefit the ASD core symptoms and can have marked adverse effects, are mainly palliative and only sometimes efficacy in attenuating specific autistic behaviors. Helminthic therapy shows potential for application as anti-inflammatory agent. Several human diseases can be treated by helminths (i.e. inflammatory bowel disease, asthma, multiple sclerosis and autoimmune diabetes). Trichuris suis ova (TSO) show strong immunomodulatory properties. Authors hypothesize that TSO could be useful in addressing ASD immune dysregulations. TSO could be a novel therapeutic option for ASD management.
Collapse
|
18
|
Khan AR, Fallon PG. Helminth therapies: translating the unknown unknowns to known knowns. Int J Parasitol 2013; 43:293-9. [PMID: 23291459 DOI: 10.1016/j.ijpara.2012.12.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/01/2012] [Accepted: 12/03/2012] [Indexed: 12/26/2022]
Abstract
The use of live helminth infections is currently in clinical trials as a novel approach for the treatment of a range of allergic and autoimmune diseases. This rapid progression from observational studies some 20 years ago to helminth clinical trials can be attributed to huge advances in not just pre-clinical and clinical evidence, pertaining to the efficacy of these parasites in unrelated diseases, but also a greater understanding of the complex immunological mechanisms that underpin these effects. Helminths have exerted significant evolutionary selective pressures on the host immune genome or "immunome". Studies on helminths were pivotal in a paradigm shift in immunology with recent discoveries of a number of novel immune cell populations. Critically, these new discoveries highlight the need to further understand the underlying mechanism behind the desirable therapeutic effects that helminths offer. With these unknown unknowns there is the distinct possibility that a true, fundamental modus operandi for helminth therapy will arrive long after it has been established in the clinic.
Collapse
Affiliation(s)
- Adnan R Khan
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, and National Children's Research Centre, Our Lady's Children's Hospital, Ireland
| | | |
Collapse
|
19
|
Kim SE, Kim JH, Min BH, Bae YM, Hong ST, Choi MH. Crude extracts of Caenorhabditis elegans suppress airway inflammation in a murine model of allergic asthma. PLoS One 2012; 7:e35447. [PMID: 22558152 PMCID: PMC3338843 DOI: 10.1371/journal.pone.0035447] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 03/16/2012] [Indexed: 12/26/2022] Open
Abstract
Epidemiological studies suggest an inverse relationship between helminth infections and allergic disease, and several helminth-derived products have been shown to suppress allergic responses in animals. This study was undertaken to evaluate the effect of a crude extract of Caenorhabditis elegans on allergic airway inflammation in a murine model of asthma. Allergic airway inflammation was induced in BALB/c mice by sensitization with ovalbumin. The effect of the C. elegans crude extract on the development of asthma and on established asthma was evaluated by analyzing airway hyperresponsiveness, serum antibody titers, lung histology and cell counts and cytokine levels in the bronchoalveolar lavage fluid. The role of IFN-γ in the suppression of asthma by the C. elegans crude extract was investigated in IFN-γ knockout and wild-type mice. When mice were sensitized with ovalbumin together with the crude extract of C. elegans, cellular infiltration into the lung was dramatically reduced in comparison with the ovalbumin-treated group. Treatment of mice with the C. elegans crude extract significantly decreased methacholine-induced airway hyperresponsiveness and the total cell counts and levels of IL-4, IL-5 and IL-13 in the bronchoalveolar lavage fluid but increased the levels of IFN-γ and IL-12. Sensitization with the C. elegans crude extract significantly diminished the IgE and IgG1 responses but provoked elevated IgG2a levels. However, the suppressive effect of the C. elegans crude extract was abolished in IFN-γ knockout mice, and the Th2 responses in these mice were as strong as those in wild-type mice sensitized with ovalbumin. The crude extract of C. elegans also suppressed the airway inflammation associated with established asthma. This study provides new insights into immune modulation by the C. elegans crude extract, which suppressed airway inflammation in mice not only during the development of asthma but also after its establishment by skewing allergen-induced Th2 responses to Th1 responses.
Collapse
Affiliation(s)
- Sung Eun Kim
- Department of Parasitology and Tropical Medicine, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Korea
| | - Jae-Hwan Kim
- Department of Parasitology and Tropical Medicine, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Korea
| | - Byung-Hoon Min
- Department of Parasitology and Tropical Medicine, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Korea
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Mee Bae
- Department of Parasitology and Tropical Medicine, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Korea
| | - Sung-Tae Hong
- Department of Parasitology and Tropical Medicine, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Korea
| | - Min-Ho Choi
- Department of Parasitology and Tropical Medicine, Seoul National University College of Medicine, and Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Korea
- * E-mail:
| |
Collapse
|
20
|
Song X, Shen J, Wen H, Zhong Z, Luo Q, Chu D, Qi Y, Xu Y, Wei W. Impact of Schistosoma japonicum infection on collagen-induced arthritis in DBA/1 mice: a murine model of human rheumatoid arthritis. PLoS One 2011; 6:e23453. [PMID: 21858123 PMCID: PMC3152573 DOI: 10.1371/journal.pone.0023453] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Accepted: 07/18/2011] [Indexed: 12/26/2022] Open
Abstract
Background The hygiene hypothesis suggests that helminth infections prevent a range of autoimmune diseases. Methodology/Principal Findings To investigate the effects of S. japonicum infection on collagen-induced arthritis (CIA), male DBA/1 mice were challenged with unisexual or bisexual S. japonicum cercariae two weeks prior to bovine type II collagen (CII) immunization or at the onset of CIA. S. japonicum infection prior to CII immunization significantly reduced the severity of CIA. ELISA (enzyme linked immunosorbent assay) showed that the levels of anti-CII IgG and IgG2a were reduced in prior schistosome-infected mice, while anti-CII IgG1 was elevated. Splenocyte proliferation against both polyclonal and antigen-specific stimuli was reduced by prior schistosome infection as measured by tritiated thymidine incorporation (3H-TdR). Cytokine profiles and CD4+ T cells subpopulation analysis by ELISA and flow cytometry (FCM) demonstrated that prior schistosome infection resulted in a significant down-regulation of pro-inflammatory cytokines (IFN-γ, TNF-α, IL-1β and IL-6) and Th1 cells, together with up-regulation of the anti-inflammatory cytokine IL-10 and Th2 cells. Interestingly, the expansion of Treg cells and the reduction of Th17 cells were only observed in bisexually infected mice. In addition, prior schistosome infection notably reduced the expression of pro-inflammatory cytokines and receptor activator of NF-κB ligand (RANKL) in the inflamed joint. However, the disease was exacerbated at one week after infection when established CIA mice were challenged with bisexual cercariae. Conclusion/Significance Our data provide direct evidence that the Th2 response evoked by prior S. japonicum infection can suppress the Th1 response and pro-inflammatory mediator and that bisexual infection with egg-laying up-regulates the Treg response and down-regulates the Th17 response, resulting in an amelioration of autoimmune arthritis. The beneficial effects might depend on the establishment of a Th2-dominant response rather than the presence of the eggs. Our results suggest that anti-inflammatory molecules from the parasite could treat autoimmune diseases.
Collapse
MESH Headings
- Animals
- Antibodies, Anti-Idiotypic/immunology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/immunology
- Cell Proliferation
- Cells, Cultured
- Collagen Type II/immunology
- Disease Models, Animal
- Female
- Humans
- Hygiene Hypothesis
- Immunization
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-4/immunology
- Interleukin-4/metabolism
- Mice
- Mice, Inbred DBA
- Schistosoma japonicum/immunology
- Schistosoma japonicum/parasitology
- Schistosomiasis japonica/immunology
- Severity of Illness Index
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Therapy with Helminths
Collapse
Affiliation(s)
- Xiaorong Song
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui, China
- The Key Laboratory of Microbiology and Parasitology, and The Key Laboratory of Zoonoses, Anhui Medical University, Hefei, Anhui, China
- Institute of Anhui Cardiovascular Research, Anhui Provincial Hospital, Hefei, Anhui, China
| | - Jilong Shen
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui, China
- The Key Laboratory of Microbiology and Parasitology, and The Key Laboratory of Zoonoses, Anhui Medical University, Hefei, Anhui, China
- * E-mail:
| | - Huiqin Wen
- The Key Laboratory of Microbiology and Parasitology, and The Key Laboratory of Zoonoses, Anhui Medical University, Hefei, Anhui, China
| | - Zhengrong Zhong
- The Key Laboratory of Microbiology and Parasitology, and The Key Laboratory of Zoonoses, Anhui Medical University, Hefei, Anhui, China
| | - Qinli Luo
- The Key Laboratory of Microbiology and Parasitology, and The Key Laboratory of Zoonoses, Anhui Medical University, Hefei, Anhui, China
| | - Deyong Chu
- The Key Laboratory of Microbiology and Parasitology, and The Key Laboratory of Zoonoses, Anhui Medical University, Hefei, Anhui, China
| | - Yao Qi
- The Key Laboratory of Microbiology and Parasitology, and The Key Laboratory of Zoonoses, Anhui Medical University, Hefei, Anhui, China
| | - Yuanhong Xu
- The Key Laboratory of Microbiology and Parasitology, and The Key Laboratory of Zoonoses, Anhui Medical University, Hefei, Anhui, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
21
|
Hewitson JP, Harcus Y, Murray J, van Agtmaal M, Filbey KJ, Grainger JR, Bridgett S, Blaxter ML, Ashton PD, Ashford DA, Curwen RS, Wilson RA, Dowle AA, Maizels RM. Proteomic analysis of secretory products from the model gastrointestinal nematode Heligmosomoides polygyrus reveals dominance of venom allergen-like (VAL) proteins. J Proteomics 2011; 74:1573-94. [PMID: 21722761 DOI: 10.1016/j.jprot.2011.06.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2011] [Revised: 05/20/2011] [Accepted: 06/05/2011] [Indexed: 01/25/2023]
Abstract
The intestinal helminth parasite, Heligmosomoides polygyrus bakeri offers a tractable experimental model for human hookworm infections such as Ancylostoma duodenale and veterinary parasites such as Haemonchus contortus. Parasite excretory-secretory (ES) products represent the major focus for immunological and biochemical analyses, and contain immunomodulatory molecules responsible for nematode immune evasion. In a proteomic analysis of adult H. polygyrus secretions (termed HES) matched to an extensive transcriptomic dataset, we identified 374 HES proteins by LC-MS/MS, which were distinct from those in somatic extract HEx, comprising 446 identified proteins, confirming selective export of ES proteins. The predominant secreted protein families were proteases (astacins and other metalloproteases, aspartic, cysteine and serine-type proteases), lysozymes, apyrases and acetylcholinesterases. The most abundant products were members of the highly divergent venom allergen-like (VAL) family, related to Ancylostoma secreted protein (ASP); 25 homologues were identified, with VAL-1 and -2 also shown to be associated with the parasite surface. The dominance of VAL proteins is similar to profiles reported for Ancylostoma and Haemonchus ES products. Overall, this study shows that the secretions of H. polygyrus closely parallel those of clinically important GI nematodes, confirming the value of this parasite as a model of helminth infection.
Collapse
Affiliation(s)
- James P Hewitson
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3JT, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|