1
|
Pandey H, Tang DWT, Wong SH, Lal D. Helminths in alternative therapeutics of inflammatory bowel disease. Intest Res 2025; 23:8-22. [PMID: 39916482 PMCID: PMC11834367 DOI: 10.5217/ir.2023.00059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/24/2023] [Accepted: 11/01/2023] [Indexed: 02/20/2025] Open
Abstract
Inflammatory bowel disease (IBD), which includes Crohn's disease and ulcerative colitis, is a nonspecific chronic inflammation of the gastrointestinal tract. Despite recent advances in therapeutics and newer management strategies, IBD largely remains untreatable. Helminth therapy is a promising alternative therapeutic for IBD that has gained some attention in the last two decades. Helminths have immunomodulatory effects and can alter the gut microbiota. The immunomodulatory effects include a strong Th2 immune response, T-regulatory cell response, and the production of regulatory cytokines. Although concrete evidence regarding the efficacy of helminth therapy in IBD is lacking, clinical studies and studies done in animal models have shown some promise. Most clinical studies have shown that helminth therapy is safe and easily tolerable. Extensive work has been done on the whipworm Trichuris, but other helminths, including Schistosoma, Trichinella, Heligmosomoides, and Ancylostoma, have also been explored for pre-clinical and animal studies. This review article summarizes the potential of helminth therapy as an alternative therapeutic or an adjuvant to the existing therapeutic procedures for IBD treatment.
Collapse
Affiliation(s)
| | - Daryl W. T. Tang
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Sunny H. Wong
- Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Devi Lal
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| |
Collapse
|
2
|
Miao S, Li J, Chen Y, Zhao W, Xu M, Liu F, Zou X, Dong X. Targeting gut microbiota and metabolism profiles with coated sodium butyrate to ameliorate high-energy and low-protein diet-induced intestinal barrier dysfunction in laying hens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:104-116. [PMID: 39635416 PMCID: PMC11615920 DOI: 10.1016/j.aninu.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 12/07/2024]
Abstract
High energy diets are a risk factor for intestinal barrier damage. Butyrate, a major energy source for intestinal epithelial cells, has been shown to improve barrier dysfunction and modulate the gut microbiota. In this trial, we examined the preventative effect of coated sodium butyrate (CSB) on high-energy and low-protein (HELP)-induced intestinal barrier injury in laying hens, and also worked to determine the underlying mechanisms by an integrative analysis of gut microbiota and the metabolome. A total of 216 healthy 28-week-old Huafeng laying hens were randomly assigned to 3 groups with 6 replicates each: the CON group (normal diet), HELP group (HELP diet) and CH500 group (500 mg/kg CSB added to HELP diet). The duration of the trial encompassed a period of 10 weeks. The results revealed that CSB treatment improved the laying rate and mitigated the detrimental effects on intestinal barrier function and the inflammatory response induced by the HELP diet in laying hens (P < 0.05). Microbial profiling analysis revealed that the CSB treatment reshaped the HELP-perturbed gut microbiota and promoted the growth of beneficial bacteria (P < 0.05). Untargeted metabolomics analysis revealed that CSB reduced the metabolites associated with intestinal inflammation (P < 0.05). In conclusion, CSB did not merely modulate alterations in the gut microbiota composition and microbial metabolites but also yielded increased egg production, while mitigating intestinal barrier dysfunction and inflammatory responses induced by HELP in laying hens.
Collapse
Affiliation(s)
- Sasa Miao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiankui Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Chen
- Hangzhou Zhejiang University Animal Hospital Co., Ltd., Hangzhou 310058, China
| | - Wenyan Zhao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mengru Xu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fang Liu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoting Zou
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyang Dong
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
3
|
Blackwell KH, Walk HMG, Peters LR, Gunlikson EM, Bright JC, Kominsky DJ, Walk ST. A simplified protocol for deriving sterile, infectious murine Heligmosomoides polygyrus bakeri larvae. STAR Protoc 2024; 5:103144. [PMID: 39167493 PMCID: PMC11381856 DOI: 10.1016/j.xpro.2024.103144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 08/23/2024] Open
Abstract
Gastrointestinal helminth infection occurs within a diverse microbiome, complicating the interpretation of whether effects are caused by the parasite versus the microbial community. Here, we present a protocol for deriving sterile larvae of the murine helminth, Heligmosomoides polygyrus bakeri (H. polygyrus), providing experimental control of the microbiome. We describe steps for sterilizing with a bleach solution and developing into infectious larvae using E. coli. We then detail procedures for removing bacterial contaminants before harvesting to ensure the generation of germ-free larvae.
Collapse
Affiliation(s)
- Karlin H Blackwell
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA.
| | - Heather M G Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - L Robert Peters
- Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D., 1000 Oakland Dr., Kalamazoo, MI 49008, USA
| | - Emily M Gunlikson
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Jack C Bright
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Douglas J Kominsky
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA.
| | - Seth T Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA.
| |
Collapse
|
4
|
Zhang Y, Hardy LC, Kapita CM, Hall JA, Arbeeva L, Campbell E, Urban JF, Belkaid Y, Nagler CR, Iweala OI. Intestinal Helminth Infection Impairs Oral and Parenteral Vaccine Efficacy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:389-402. [PMID: 37272847 PMCID: PMC10524302 DOI: 10.4049/jimmunol.2300084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023]
Abstract
The impact of endemic parasitic infection on vaccine efficacy is an important consideration for vaccine development and deployment. We have examined whether intestinal infection with the natural murine helminth Heligmosomoides polygyrus bakeri alters Ag-specific Ab and cellular immune responses to oral and parenteral vaccination in mice. Oral vaccination of mice with a clinically relevant, live, attenuated, recombinant Salmonella vaccine expressing chicken egg OVA (Salmonella-OVA) induced the accumulation of activated, OVA-specific T effector cells rather than OVA-specific regulatory T cells in the GALT. Intestinal helminth infection significantly reduced Th1-skewed Ab responses to oral vaccination with Salmonella-OVA. Activated, adoptively transferred, OVA-specific CD4+ T cells accumulated in draining mesenteric lymph nodes of vaccinated mice, regardless of their helminth infection status. However, helminth infection increased the frequencies of adoptively transferred OVA-specific CD4+ T cells producing IL-4 and IL-10 in the mesenteric lymph node. Ab responses to the oral Salmonella-OVA vaccine were reduced in helminth-free mice adoptively transferred with OVA-specific CD4+ T cells harvested from mice with intestinal helminth infection. Intestinal helminth infection also significantly reduced Th2-skewed Ab responses to parenteral vaccination with OVA adsorbed to alum. These findings suggest that vaccine-specific CD4+ T cells induced in the context of helminth infection retain durable immunomodulatory properties and may promote blunted Ab responses to vaccination. They also underscore the potential need to treat parasitic infection before mass vaccination campaigns in helminth-endemic areas.
Collapse
Affiliation(s)
- Yugen Zhang
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599
| | - LaKeya C. Hardy
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599
| | - Camille M. Kapita
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
| | - Jason A. Hall
- National Institute of Allergy and Infectious Diseases Microbiome Program and Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, Center for Human Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Liubov Arbeeva
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
| | - Evelyn Campbell
- Biological Sciences Division, University of Chicago, Chicago, IL, 60637
| | - Joseph F. Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory and Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, 10300 Baltimore Avenue BLDG 307-C BARC-East, Beltsville, MD, 20705
| | - Yasmine Belkaid
- National Institute of Allergy and Infectious Diseases Microbiome Program and Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, Center for Human Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Cathryn R. Nagler
- Biological Sciences Division, University of Chicago, Chicago, IL, 60637
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637
- Center for Immunology and Inflammatory Disease, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Charlestown, MA 02129
| | - Onyinye I. Iweala
- Department of Medicine, Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Chapel Hill, NC, 27599
- Department of Pediatrics, University of North Carolina Food Allergy Initiative, Division of Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599
- Center for Immunology and Inflammatory Disease, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Charlestown, MA 02129
| |
Collapse
|
5
|
Deng L, Wojciech L, Png CW, Koh EY, Aung TT, Kioh DYQ, Chan ECY, Malleret B, Zhang Y, Peng G, Gascoigne NRJ, Tan KSW. Experimental colonization with Blastocystis ST4 is associated with protective immune responses and modulation of gut microbiome in a DSS-induced colitis mouse model. Cell Mol Life Sci 2022; 79:245. [PMID: 35435504 PMCID: PMC9016058 DOI: 10.1007/s00018-022-04271-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/27/2022] [Accepted: 03/22/2022] [Indexed: 02/06/2023]
Abstract
Background Blastocystis is a common gut protistan parasite in humans and animals worldwide, but its interrelationship with the host gut microbiota and mucosal immune responses remains poorly understood. Different murine models of Blastocystis colonization were used to examine the effect of a common Blastocystis subtype (ST4) on host gut microbial community and adaptive immune system. Results Blastocystis ST4-colonized normal healthy mice and Rag1−/− mice asymptomatically and was able to alter the microbial community composition, mainly leading to increases in the proportion of Clostridia vadinBB60 group and Lachnospiraceae NK4A136 group, respectively. Blastocystis ST4 colonization promoted T helper 2 (Th2) response defined by interleukin (IL)-5 and IL-13 cytokine production, and T regulatory (Treg) induction from colonic lamina propria in normal healthy mice. Additionally, we observed that Blastocystis ST4 colonization can maintain the stability of bacterial community composition and induce Th2 and Treg immune responses to promote faster recovery from experimentally induced colitis. Furthermore, fecal microbiota transplantation of Blastocystis ST4-altered gut microbiome to colitis mice reduced the severity of colitis, which was associated with increased production of short-chain fat acids (SCFAs) and anti-inflammatory cytokine IL-10. Conclusions The data confirm our hypothesis that Blastocystis ST4 is a beneficial commensal, and the beneficial effects of Blastocystis ST4 colonization is mediated through modulating of the host gut bacterial composition, SCFAs production, and Th2 and Treg responses in different murine colonization models. Supplementary Information The online version contains supplementary material available at 10.1007/s00018-022-04271-9.
Collapse
Affiliation(s)
- Lei Deng
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore.,The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Lukasz Wojciech
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore
| | - Chin Wen Png
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Eileen Yiling Koh
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Thet Tun Aung
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore
| | - Dorinda Yan Qin Kioh
- Department of Pharmacy, Faculty of Science, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Benoit Malleret
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore.,Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Immunos Building, Singapore, 138648, Singapore
| | - Yongliang Zhang
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Guangneng Peng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| | - Nicholas Robert John Gascoigne
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117597, Singapore
| | - Kevin Shyong Wei Tan
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore.
| |
Collapse
|
6
|
Mitigation of Toxoplasma gondii-induced ileitis by Trichinellaspiralis infection pinpointing immunomodulation. J Parasit Dis 2022; 46:491-501. [DOI: 10.1007/s12639-022-01476-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/09/2022] [Indexed: 10/18/2022] Open
|
7
|
Arai T, Lopes F. Potential of human helminth therapy for resolution of inflammatory bowel disease: The future ahead. Exp Parasitol 2021; 232:108189. [PMID: 34848244 DOI: 10.1016/j.exppara.2021.108189] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/06/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022]
Abstract
Inflammatory bowel disease (IBD) is associated with a dysregulated mucosal immune response in the gastrointestinal tract. The number of patients with IBD has increased worldwide, especially in highly industrialized western societies. The population of patients with IBD in North America is forecasted to reach about four million by 2030; meanwhile, there is no definitive therapy for IBD. Current anti-inflammatory, immunosuppressive, or biological treatment may induce and maintain remission, but not all patients respond to these treatments. Recent studies explored parasitic helminths as a novel modality of therapy due to their potent immunoregulatory properties in humans. Research using IBD animal models infected with a helminth or administered helminth-derived products such as excretory-secretory products has been promising, and helminth-microbiota interactions exert their anti-inflammatory effects by modulating the host immunity. Recent studies also indicate that evidence that helminth-derived metabolites may play a role in anticolitic effects. Thus, the helminth shows a potential benefit for treatment against IBD. Here we review the current feasibility of "helminth therapy" from the laboratory for application in IBD management.
Collapse
Affiliation(s)
- Toshio Arai
- Institution of Parasitology, McGill University, Quebec, Canada; Department of Gastroenterology, Hashimoto Municipal Hospital, Wakayama, Japan
| | - Fernando Lopes
- Institution of Parasitology, McGill University, Quebec, Canada.
| |
Collapse
|
8
|
Smyth DJ, Ren B, White MPJ, McManus C, Webster H, Shek V, Evans C, Pandhal J, Fields F, Maizels RM, Mayfield S. Oral delivery of a functional algal-expressed TGF-β mimic halts colitis in a murine DSS model. J Biotechnol 2021; 340:1-12. [PMID: 34390759 PMCID: PMC8516079 DOI: 10.1016/j.jbiotec.2021.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease (IBD) is a set of immunological disorders which can generate chronic pain and fatigue associated with the inflammatory symptoms. The treatment of IBD remains a significant hurdle with current therapies being only partially effective or having significant side effects, suggesting that new therapies that elicit different modes of action and delivery strategies are required. TGM1 is a TGF-β mimic that was discovered from the intestinal helminth parasite Heligmosomoides polygyrus and is thought to be produced by the parasite to suppress the intestinal inflammation response to help evade host immunity, making it an ideal candidate to be developed as a novel anti-inflammatory bio-therapeutic. Here we utilized the expression system of the edible green algae Chlamydomonas reinhardtii in order to recombinantly produce active TGM1 in a form that could be ingested. C. reinhardtii robustly expressed TGM1, and the resultant recombinant protein is biologically active as measured by regulatory T cell induction. When delivered orally to mice, the algal expressed TGM1 is able to ameliorate weight loss, lymphadenopathy, and disease symptoms in a mouse model of DSS-induced colitis, demonstrating the potential of this biologic as a novel treatment of IBD.
Collapse
Affiliation(s)
- Danielle J Smyth
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Bijie Ren
- California Center for Algae Biotechnology, Division of Biological Sciences, University of California, San Diego, USA
| | - Madeleine P J White
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Caitlin McManus
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Holly Webster
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Vivien Shek
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Caroline Evans
- Bioanalytical Facility, Dept Chemical and Biological Engineering, University of Sheffield, UK
| | - Jagroop Pandhal
- Bioanalytical Facility, Dept Chemical and Biological Engineering, University of Sheffield, UK
| | - Francis Fields
- California Center for Algae Biotechnology, Division of Biological Sciences, University of California, San Diego, USA
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK.
| | - Stephen Mayfield
- California Center for Algae Biotechnology, Division of Biological Sciences, University of California, San Diego, USA.
| |
Collapse
|
9
|
Li J, Liu X, Ding J, Tang B, Bai X, Wang Y, Li S, Liu M, Wang X. Effect of Trichinella spp. or derived antigens on chemically induced inflammatory bowel disease (IBD) in mouse models: A systematic review and meta-analysis. Int Immunopharmacol 2020; 85:106646. [PMID: 32485355 DOI: 10.1016/j.intimp.2020.106646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/14/2020] [Accepted: 05/25/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Trichinella or derived antigens have been suggested to be potential therapeutic agents for inflammatory bowel disease (IBD). We aimed to conduct a systematic review and meta-analysis of the available literature to estimate the effect of Trichinella or derived antigens on chemically induced IBD. METHODS Studies were identified by searching the Cochrane Central Register of Controlled Trials, PubMed, Scopus, Web of Science, and Science Direct from inception to February 2020. We included articles written in English that investigated the effect of Trichinella infection and/or derived products in mouse models of IBD. Studies were pooled, and the combined standard mean difference (SMD) and 95% confidence interval (CI) were calculated using a random-effect or fixed-effect model. RESULTS Thirteen studies were eventually included in the meta-analysis. The results indicated significant differences in the disease activity index (DAI), myeloperoxidase (MPO) activity, macroscopic inflammation score, and microscopic inflammation score between the experimental group and the control group. The anti-inflammatory cytokines interleukin (IL)-4, transforming growth factor-beta (TGF-β), IL-10 and IL-13 were significantly increased in the experimental group compared with the control group, whereas the levels of the proinflammatory cytokines interferon (IFN)-γ, IL-6, TNF-α, and IL-17 were significantly decreased. The percentage of regulatory T (Treg) cells was also significantly increased, while the level of the M1 phenotypic macrophage marker iNOS was significantly decreased and the expression of the M2 phenotypic macrophage marker Arg-1 was significantly increased. CONCLUSION Trichinella infection or derived antigens is effective for the alleviation of IBD in mouse models.
Collapse
Affiliation(s)
- Jian Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Jing Ding
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Bin Tang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Yang Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Shicun Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Xuelin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China.
| |
Collapse
|
10
|
Xu YW, Xing RX, Zhang WH, Li L, Wu Y, Hu J, Wang C, Luo QL, Shen JL, Chen X. Toxoplasma ROP16 I/III ameliorated inflammatory bowel diseases via inducing M2 phenotype of macrophages. World J Gastroenterol 2019; 25:6634-6652. [PMID: 31832003 PMCID: PMC6906210 DOI: 10.3748/wjg.v25.i45.6634] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/03/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized by chronic and non-specific inflammation of the intestinal mucosa and mainly includes ulcerative colitis and Crohn's disease.
AIM To explore the beneficial effect of ToxoROP16I/III-induced M2 phynotype macrophages in homeostasis of IBDs through downregulation of M1 inflammatory cells.
METHODS RAW264.7 macrophages stimulated by lipopolysaccharide (LPS) (M1 cells) were co-cultured with Caco-2 cells as an inflammatory model of IBD in vitro. The expression of ToxoROP16I/III was observed in RAW264.7 macrophages that were transfected with pEGFP-rop16I/III. The phenotypes of M2 and M1 macrophage cells were assessed by quantitative real-time reverse transcriptase polymerase chain reaction and the expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, transforming growth factor (TGF)-β1, IL-10, inducible nitric oxide synthase (iNOS), and arginase-1 (Arg-1) was detected. The expression of iNOS, Arg-1, signal transducer and activator of transcription 3 (Stat3), p-Stat3, Stat6, p-Stat6, programmed death ligand-2 (PD-L2), caspase-3, -8, and -9 was analyzed by Western blotting, and Griess assays were performed to detect nitric oxide (NO). TNF-α, IL-1β, IL-6, TGF-β1, and IL-10 expression in the supernatants was detected by enzyme-linked immunosorbent assay, and Caco-2 cell apoptosis was determined by flow cytometry after mixing M1 cells with M2 cells in a Caco-2 cell co-culture system.
RESULTS M1 cells exhibited significantly increased production of iNOS, NO, TNF-α, IL-1β, and IL-6, while ToxoROP16I/III induced macrophage bias to M2 cells in vitro, showing increased expression of Arg-1, IL-10 and TGF-β1 and elevated production of p-Stat3 and p-Stat6. The mixed M1 and M2 cell culture induced by ToxoROP16I/III exhibited decreased production of NO and iNOS and upregulated expression of Arg-1 and PD-L2. Accordingly, Caco-2 cells became apoptotic, and apoptosis-associated proteins such as caspase-3, -8 and -9 were dampened during co-culture of M1 and M2 cells. Flow cytometry analysis showed that co-culture of M1 cells with Caco-2 cells facilitated the apoptosis of Caco-2 cells, but co-culture of M1 and M2 cells alleviated Caco-2 cell apoptosis.
CONCLUSION ToxoROP16I/III-induced M2 macrophages inhibited apoptosis of Caco-2 cells caused by M1 macrophages. This finding may help gain a better understanding of the underlying mechanism and represent a promising therapeutic strategy for IBDs.
Collapse
Affiliation(s)
- Yong-Wei Xu
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Rui-Xin Xing
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Wen-Hui Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Lu Li
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Yi Wu
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Jing Hu
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Cong Wang
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Zoonoses Anhui, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Qing-Li Luo
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Zoonoses Anhui, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Ji-Long Shen
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Zoonoses Anhui, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Xi Chen
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| |
Collapse
|
11
|
Sun S, Li H, Yuan Y, Wang L, He W, Xie H, Gao S, Cheng R, Qian H, Jiang H, Wang X, Zhan B, Fang Q, Yang X. Preventive and therapeutic effects of Trichinella spiralis adult extracts on allergic inflammation in an experimental asthma mouse model. Parasit Vectors 2019; 12:326. [PMID: 31253164 PMCID: PMC6599242 DOI: 10.1186/s13071-019-3561-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/09/2019] [Indexed: 01/09/2023] Open
Abstract
Background Helminths immunomodulate the host immune system by secreting proteins to create an inhibitory environment as a strategy for survival in the host. As a bystander effect, this balances the host immune system to reduce hypersensitivity to allergens or autoantigens. Based on this, helminth therapy has been used to treat some allergic or autoimmune diseases. As a tissue-dwelling helminth, Trichinella spiralis infection has been identified to have strong immunomodulatory effects; the effective components in the worm have not yet been identified. Methods The soluble extracts of T. spiralis adult worms and muscle larvae were used to treat airway inflammation before and after an ovalbumin (OVA)-sensitization/challenge in an OVA-induced asthma mouse model. The therapeutic effects were observed by measuring the level of inflammation in the lungs. Results The soluble products derived from T. spiralis parasites, especially from adult worms, were able to ameliorate OVA-induced airway inflammatory responses which were associated with reduced eosinophil infiltration, OVA-specific IgE, Th2 cytokine IL-4, and increased IL-10 and TGF-β. The stimulation of the Treg response may contribute to the alleviated allergic inflammation. Conclusions Trichinella spiralis worm extracts stimulate regulatory cytokines that are associated with reduced allergic airway inflammation. The identification of effective components in the adult worm extracts will be a crucial approach for developing a novel therapeutic for allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Siying Sun
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Huihui Li
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Yuan Yuan
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Liyuan Wang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Wenxin He
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Hong Xie
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Shifang Gao
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Ruoxue Cheng
- Pharmacy College of Anhui Medical University, Hefei, 230001, China
| | - Haichun Qian
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Hui Jiang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Xiaoli Wang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Bin Zhan
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qiang Fang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China. .,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China.
| | - Xiaodi Yang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China. .,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China.
| |
Collapse
|
12
|
Hookworm-Derived Metabolites Suppress Pathology in a Mouse Model of Colitis and Inhibit Secretion of Key Inflammatory Cytokines in Primary Human Leukocytes. Infect Immun 2019; 87:IAI.00851-18. [PMID: 30670556 DOI: 10.1128/iai.00851-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/15/2019] [Indexed: 12/13/2022] Open
Abstract
Iatrogenic hookworm therapy shows promise for treating disorders that result from a dysregulated immune system, including inflammatory bowel disease (IBD). Using a murine model of trinitrobenzenesulfonic acid-induced colitis and human peripheral blood mononuclear cells, we demonstrated that low-molecular-weight metabolites derived from both somatic extracts (LMWM-SE) and excretory-secretory products (LMWM-ESP) of the hookworm, Ancylostoma caninum, display anti-inflammatory properties. Administration to mice of LMWM-ESP as well as sequentially extracted fractions of LMWM-SE using both methanol (SE-MeOH) and hexane-dichloromethane-acetonitrile (SE-HDA) resulted in significant protection against T cell-mediated immunopathology, clinical signs of colitis, and impaired histological colon architecture. To assess bioactivity in human cells, we stimulated primary human leukocytes with lipopolysaccharide in the presence of hookworm extracts and showed that SE-HDA suppressed ex vivo production of inflammatory cytokines. Gas chromatography-mass spectrometry (MS) and liquid chromatography-MS analyses revealed the presence of 46 polar metabolites, 22 fatty acids, and five short-chain fatty acids (SCFAs) in the LMWM-SE fraction and 29 polar metabolites, 13 fatty acids, and six SCFAs in the LMWM-ESP fraction. Several of these small metabolites, notably the SCFAs, have been previously reported to have anti-inflammatory properties in various disease settings, including IBD. This is the first report showing that hookworms secrete small molecules with both ex vivo and in vivo anti-inflammatory bioactivity, and this warrants further exploration as a novel approach to the development of anti-inflammatory drugs inspired by coevolution of gut-dwelling hookworms with their vertebrate hosts.
Collapse
|
13
|
Xu J, Liu M, Yu P, Wu L, Lu Y. Effect of recombinant Trichinella spiralis cysteine proteinase inhibitor on TNBS-induced experimental inflammatory bowel disease in mice. Int Immunopharmacol 2018; 66:28-40. [PMID: 30419451 DOI: 10.1016/j.intimp.2018.10.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/26/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic autoimmune disease with a high recurrence rate. Ulcerative colitis (UC) and Crohn's disease (CD) are two types of IBD. At present, parasite-derived cysteine protease inhibitors have received extensive attention from researchers, and experiments have confirmed that these protease have an effect on certain autoimmune diseases. So we conducted experiments to investigate the effect of Trichinella spiralis cysteine protease inhibitors on TNBS-induced mouse CD models. In this experiment, 72 male BALB/c mice aged 6-8 weeks were randomly divided into two groups: prevention group and therapy group. The mice were sacrificed and harvested on the 7th day after the model was established to measure the changes of various indicators of colitis. The comparison of the TsCystatin + TNBS group with the PBS + TNBS group showed that the DAI score, MPO activity, and colonic macroscopic and microscopic damage significantly reduced, IFN-γ significantly decreased, IL-4 expression increased, and NF-κB expression decreased. The percentage of CD4+CD25+Foxp3+ Treg and CD8+CD28- Treg in spleen, and the proportion of CD4+/CD8+ Treg cells decreased. In the therapy group, we found no significant difference between the TNBS+PBS group and TNBS + TsCystatin group. Treatment with TsCystatin exerted a good intervention effect on the TNBS-induced mouse CD model. TsCystatin possibly induced a Th2-type immune response in the body, which balanced the Th1-type immune response induced by TNBS administration, thereby relieving colitis.
Collapse
Affiliation(s)
- Jingyun Xu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Street, Harbin 150030, China
| | - Mingxu Liu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Street, Harbin 150030, China
| | - Pengcheng Yu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Street, Harbin 150030, China
| | - Lijia Wu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Street, Harbin 150030, China
| | - Yixin Lu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Street, Harbin 150030, China.
| |
Collapse
|
14
|
Maruszewska-Cheruiyot M, Donskow-Łysoniewska K, Doligalska M. Helminth Therapy: Advances in the use of Parasitic Worms Against Inflammatory Bowel Diseases and its Challenges. Helminthologia 2018; 55:1-11. [PMID: 31662622 PMCID: PMC6799527 DOI: 10.1515/helm-2017-0048] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022] Open
Abstract
Development of modern medicine and better living conditions in the 20th century helped in reducing a number of cases of infectious diseases. During the same time, expansion of autoimmunological disorders was noticed. Among other are Inflammatory Bowel Diseases (IBD) including ulcerative colitis and Crohn's disease which are chronic and relapsing inflammation of the gastrointestinal tract. Absence of effective treatment in standard therapies effects the search for alternative opportunities. As per hygienic hypothesis increasing number of cases of autoimmune diseases is as a result of reduced exposure to pathogens, especially parasites. Thus, one of the promising remedial acts against IBD and other allergic and autoimmune disorders is "helminth therapy". Cure with helminths seems to be the most effective therapy of IBD currently proposed. Helminth therapy focuses on advantageous results that have been obtained from the clinical trials, but its mechanisms are still unclear. Explanation of this phenomenon would help to develop new drugs against IBD based on helminth immunomodulatory molecules.
Collapse
Affiliation(s)
- M. Maruszewska-Cheruiyot
- Department of Parasitology, Faculty of Biology University of Warsaw, Miecznikowa 1, 02-096Warsaw, Poland
- E-mail:
| | - K. Donskow-Łysoniewska
- Department of Parasitology, Faculty of Biology University of Warsaw, Miecznikowa 1, 02-096Warsaw, Poland
| | - M. Doligalska
- Department of Parasitology, Faculty of Biology University of Warsaw, Miecznikowa 1, 02-096Warsaw, Poland
| |
Collapse
|
15
|
|
16
|
Pastille E, Frede A, McSorley HJ, Gräb J, Adamczyk A, Kollenda S, Hansen W, Epple M, Buer J, Maizels RM, Klopfleisch R, Westendorf AM. Intestinal helminth infection drives carcinogenesis in colitis-associated colon cancer. PLoS Pathog 2017; 13:e1006649. [PMID: 28938014 PMCID: PMC5627963 DOI: 10.1371/journal.ppat.1006649] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 10/04/2017] [Accepted: 09/15/2017] [Indexed: 12/26/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract, strongly associated with an increased risk of colorectal cancer development. Parasitic infections caused by helminths have been shown to modulate the host’s immune response by releasing immunomodulatory molecules and inducing regulatory T cells (Tregs). This immunosuppressive state provoked in the host has been considered as a novel and promising approach to treat IBD patients and alleviate acute intestinal inflammation. On the contrary, specific parasite infections are well known to be directly linked to carcinogenesis. Whether a helminth infection interferes with the development of colitis-associated colon cancer (CAC) is not yet known. In the present study, we demonstrate that the treatment of mice with the intestinal helminth Heligmosomoides polygyrus at the onset of tumor progression in a mouse model of CAC does not alter tumor growth and distribution. In contrast, H. polygyrus infection in the early inflammatory phase of CAC strengthens the inflammatory response and significantly boosts tumor development. Here, H. polygyrus infection was accompanied by long-lasting alterations in the colonic immune cell compartment, with reduced frequencies of colonic CD8+ effector T cells. Moreover, H. polygyrus infection in the course of dextran sulfate sodium (DSS) mediated colitis significantly exacerbates intestinal inflammation by amplifying the release of colonic IL-6 and CXCL1. Thus, our findings indicate that the therapeutic application of helminths during CAC might have tumor-promoting effects and therefore should be well-considered. Evidence from epidemiological studies indicates an inverse correlation between the incidence of certain immune-mediated diseases, including inflammatory bowel diseases, and exposure to helminths. As a consequence, helminth parasites were tested for treating IBD patients, resulting in clinical amelioration of the disease due to the induction of an immunosuppressive microenvironment. However, some infection–related cancers can be attributed to helminth infection, probably due to the generation of a microenvironment that might be conductive to the initiation and development of cancer. In the present study, we aimed to unravel the apparently controversial function of helminths in a mouse model of colitis-associated colon cancer. We show that helminth infection in the onset of colitis and colitis-associated colon cancer does not ameliorate colonic inflammation but activates intestinal immune cells that further facilitate tumor development. Therefore, a better understanding of mechanisms by which helminths modulate host immune responses in the gut should be defined precisely before application of helminths in autoimmune diseases like IBD.
Collapse
Affiliation(s)
- Eva Pastille
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Annika Frede
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Henry J. McSorley
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jessica Gräb
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Alexandra Adamczyk
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sebastian Kollenda
- Institute for Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Duisburg, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Matthias Epple
- Institute for Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Duisburg, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Rick M. Maizels
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universitaet Berlin, Berlin, Germany
| | - Astrid M. Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- * E-mail:
| |
Collapse
|
17
|
Wang M, Wu L, Weng R, Zheng W, Wu Z, Lv Z. Therapeutic potential of helminths in autoimmune diseases: helminth-derived immune-regulators and immune balance. Parasitol Res 2017; 116:2065-2074. [PMID: 28664463 DOI: 10.1007/s00436-017-5544-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 06/21/2017] [Indexed: 12/22/2022]
Abstract
Helminths have accompanied human throughout history by releasing immune-evasion molecules that could counteract an aberrant immune response within the host. In the past decades, helminth infections are becoming less prevalent possibly due to the developed sanitation. Meanwhile, the incidence of autoimmune diseases is increasing, which cannot be exclusively explained by the changes of susceptibility genes. While the hygiene hypothesis casts light on the problem. The infections of helminths are believed to interact with and regulate human immunity with the byproduct of suppressing the autoimmune diseases. Thus, helminths are potential to treat or cure the autoimmune diseases. The therapeutic progresses and possible immune suppression mechanisms are illustrated in the review. The helminths that are studied most intensively include Heligmosomoides polygyrus, Hymenolepis diminuta, Schistosoma mansoni, Trichinella spiralis, and Trichuris suis. Special attentions are paid on the booming animal models and clinical trials that are to detect the efficiency of immune-modulating helminth-derived molecules on autoimmune diseases. These trials provide us with a prosperous clinical perspective, but the precise mechanism of the down-regulatory immune response remains to be clarified. More efforts are needed to be dedicated until these parasite-derived immune modulators could be used in clinic to treat or cure the autoimmune diseases under a standard management.
Collapse
Affiliation(s)
- Meng Wang
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Linxiang Wu
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Rennan Weng
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Weihong Zheng
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Zhongdao Wu
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, China
| | - Zhiyue Lv
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China. .,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China. .,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, China.
| |
Collapse
|
18
|
Varyani F, Fleming JO, Maizels RM. Helminths in the gastrointestinal tract as modulators of immunity and pathology. Am J Physiol Gastrointest Liver Physiol 2017; 312:G537-G549. [PMID: 28302598 PMCID: PMC5495915 DOI: 10.1152/ajpgi.00024.2017] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/06/2017] [Accepted: 03/12/2017] [Indexed: 01/31/2023]
Abstract
Helminth parasites are highly prevalent in many low- and middle-income countries, in which inflammatory bowel disease and other immunopathologies are less frequent than in the developed world. Many of the most common helminths establish themselves in the gastrointestinal tract and can exert counter-inflammatory influences on the host immune system. For these reasons, interest has arisen as to how parasites may ameliorate intestinal inflammation and whether these organisms, or products they release, could offer future therapies for immune disorders. In this review, we discuss interactions between helminth parasites and the mucosal immune system, as well as the progress being made toward identifying mechanisms and molecular mediators through which it may be possible to attenuate pathology in the intestinal tract.
Collapse
Affiliation(s)
- Fumi Varyani
- 1Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom; ,2Edinburgh Clinical Academic Track, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom; and
| | - John O. Fleming
- 3Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Rick M. Maizels
- 1Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom;
| |
Collapse
|
19
|
Khelifi L, Soufli I, Labsi M, Touil-Boukoffa C. Immune-protective effect of echinococcosis on colitis experimental model is dependent of down regulation of TNF-α and NO production. Acta Trop 2017; 166:7-15. [PMID: 27983971 DOI: 10.1016/j.actatropica.2016.10.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/24/2016] [Accepted: 10/26/2016] [Indexed: 12/26/2022]
Abstract
Hydatid disease (echinococcosis) is a chronic, endemic helminthic disease caused by the larval stage of the tapeworm, Echinococcus granulosus. This disease is endemic in many parts of the world, such as the Mediterranean area, and in particular in Algeria. Helminth parasites have developed complex strategies to modulate the immune responses of their hosts through versatile immune-regulatory mechanisms. These mechanisms may regulate immune responses associated with inflammatory diseases such as inflammatory bowel diseases (IBD). the goal of this study was to investigate the effect of Echinococcus granulosus infection on the development of dextran sulfate sodium (DSS)-induced colitis. Our results demonstrated that E. granulosus infection significantly improved the clinical symptoms and histological scores observed during DSS-induced colitis, and also maintained mucus production by goblet cells. Interestingly, this infection reduced Nitric oxide (NO) and tumor necrosis factor α (TNF-α) production and attenuated inducible nitric oxide synthase (iNOS) and nuclear factor-κB (NF-κB) expression in colonic tissues. Collectively, our data support the hygiene hypothesis and indicate that prior infection with E. granulosus can effectively protect mice from DSS-induced colitis by enhancing immune-regulatory mechanisms.
Collapse
Affiliation(s)
- Lila Khelifi
- Laboratory of Cellular and Molecular Biology, Department of Biology, University of Sciences and Technology Houari Boumediene, Algiers, Algeria.
| | - Imene Soufli
- Laboratory of Cellular and Molecular Biology, Department of Biology, University of Sciences and Technology Houari Boumediene, Algiers, Algeria.
| | - Moussa Labsi
- Laboratory of Cellular and Molecular Biology, Department of Biology, University of Sciences and Technology Houari Boumediene, Algiers, Algeria.
| | - Chafia Touil-Boukoffa
- Laboratory of Cellular and Molecular Biology, Department of Biology, University of Sciences and Technology Houari Boumediene, Algiers, Algeria.
| |
Collapse
|
20
|
Hand TW, Vujkovic-Cvijin I, Ridaura VK, Belkaid Y. Linking the Microbiota, Chronic Disease, and the Immune System. Trends Endocrinol Metab 2016; 27:831-843. [PMID: 27623245 PMCID: PMC5116263 DOI: 10.1016/j.tem.2016.08.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
Chronic inflammatory diseases (CIDs) are the most important causes of mortality in the world today and are on the rise. We now know that immune-driven inflammation is critical in the etiology of these diseases, though the environmental triggers and cellular mechanisms that lead to their development are still mysterious. Many CIDs are associated with significant shifts in the microbiota toward inflammatory configurations, which can affect the host both by inducing local and systemic inflammation and by alterations in microbiota-derived metabolites. This review discusses recent findings suggesting that shifts in the microbiota may contribute to chronic disease via effects on the immune system.
Collapse
Affiliation(s)
- Timothy W. Hand
- R.K. Mellon Institute for Pediatric Research, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA, 15224
- Correspondence addressed to: Timothy Hand () or Yasmine Belkaid ()
| | - Ivan Vujkovic-Cvijin
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID/NIH, Bethesda, Maryland 20892, USA
| | - Vanessa K. Ridaura
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID/NIH, Bethesda, Maryland 20892, USA
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID/NIH, Bethesda, Maryland 20892, USA
- National Institute of Allergy and Infectious diseases (NIAID) Microbiome Program, National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| |
Collapse
|
21
|
Xu AT, Lu JT, Ran ZH, Zheng Q. Exosome in intestinal mucosal immunity. J Gastroenterol Hepatol 2016; 31:1694-1699. [PMID: 27061439 DOI: 10.1111/jgh.13413] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/05/2016] [Indexed: 12/17/2022]
Abstract
Intercellular communication of immune cells is critical to elicit efficient inflammatory responses. In intestinal mucosa, imbalance in pro-inflammatory and anti-inflammatory mediators, especially cytokines and chemokines, characterizes the underlying immune mechanisms of inflammatory bowel disease. Exosomes, small membrane vesicles secreted into the extracellular environment, are emerging as another important intercellular messenger in immune responses. A major recent breakthrough in this field unveils the capacity of exosomes to mediate the functional transfer of genetic materials (mRNAs and miRNAs) between immune cells. RAB27A and RAB27B are two small GTPases involved in exosome secretion. With respect to intestinal mucosal immunity, increased number of RAB27A-positive immune cells and RAB27B-positive immune cells are demonstrated in the colonic mucosa of patients with active ulcerative colitis as compared with that of healthy controls. This indicates the important role of exosome-mediated immune responses in the pathogenesis of inflammatory bowel disease. Here, we will discuss the immune properties of exosomes and recent advances in their function with a special focus on intestinal mucosal immunity.
Collapse
Affiliation(s)
- An Tao Xu
- Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai IBD Research Center, Shanghai, China
| | - Jun Tao Lu
- Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai IBD Research Center, Shanghai, China
| | - Zhi Hua Ran
- Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai IBD Research Center, Shanghai, China.
| | - Qing Zheng
- Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai IBD Research Center, Shanghai, China.
| |
Collapse
|
22
|
Morimoto M, Azuma N, Kadowaki H, Abe T, Suto Y. Regulation of type 2 diabetes by helminth-induced Th2 immune response. J Vet Med Sci 2016; 78:1855-1864. [PMID: 27665994 PMCID: PMC5240765 DOI: 10.1292/jvms.16-0183] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Helminth-induced type 2 cytokines increase the number of regulatory T cells and
alternatively activated macrophages, resulting in modulation of the host–immune system.
Studies on these parasite-induced immunoregulatory mechanisms might contribute to the
development of new therapies for inflammatory diseases, including type 2 diabetes (T2D).
Previous studies have suggested that progression of obesity-associated metabolic
abnormalities is under pathophysiological control of CD4+ T cells. Glucose absorption
through the intestinal epithelium reduced after infection in a STAT-6-dependent manner. In
this study, we investigated whether infection with the gastrointestinal nematode parasite
Heligmosomoides polygyrus (Hp) can modulate T2D-associated pathology in
a mouse model (KK-Ay/TaJcl). KK-Ay/TaJcl mice were inoculated with infective third-stage
Hp larvae and studied at Day 8 following infection. Uninfected KK-Ay/TaJcl mice showed
high blood glucose levels even 120 min after administration of glucose by IP injection.
However, it was significantly improved in the infected group. HOMA-IR, fat accumulation
and FAS gene expression in the liver were significantly decreased by Hp infection. GLUT2
gene expression in this group was significantly lower than that in the uninfected diabetic
mice, which might be related to the decrease in glucose absorption in the
parasite-infected intestine. In conclusion, helminth-induced type 2 immune responses might
contribute to T2D disease control.
Collapse
Affiliation(s)
- Motoko Morimoto
- School of Food, Agricultural, and Environmental Sciences, Miyagi University, Sendai City, Miyagi 982-0215, Japan
| | | | | | | | | |
Collapse
|
23
|
Wang S, Xie Y, Yang X, Wang X, Yan K, Zhong Z, Wang X, Xu Y, Zhang Y, Liu F, Shen J. Therapeutic potential of recombinant cystatin from Schistosoma japonicum in TNBS-induced experimental colitis of mice. Parasit Vectors 2016; 9:6. [PMID: 26728323 PMCID: PMC4700642 DOI: 10.1186/s13071-015-1288-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/28/2015] [Indexed: 12/22/2022] Open
Abstract
Background Helminth infections and their components have been shown to have a protective effect on autoimmune diseases. The isolated purified protein from Schisotosoma japonicum and its potential therapeutic effect on trinitrobenzene sulfonic acid (TNBS)-induced colitis could provide an alternative way to treat inflammatory bowel disease (IBDs). Methods Colitis was induced in Balb/c mice by rectal administration of 2.5 % TNBS, followed by intraperitoneal injection of rSjcystatin 50 μg at 6 h and 24 h afterwards. The inflammation was monitored by recording weight change, stool character and bleeding, colon length, macroscopic score (MAO), microscopic score (MIO), myeloperoxidase activity (MPO) and disease activity index (DAI). The potential underlying mechanism was investigated by examining cytokine profiles including Th1 (IFNγ), Th2 (IL-4), Th17 (IL-17A) and Treg subsets from lymphocytes of spleen, mesenteric lymph nodes (MLN) and intestinal lamina propria mononuclear cells (LPMCs) by flow cytometry. The mRNA relative expressions of the cytokines in splenocytes and MLN were analysed by quantitative real time reverse-transcriptase polymerase chain reaction (qRT-PCR). Simultaneously, the concentrations of the cytokines in the colon homogenate supernatants were tested by enzyme-linked immunosorbent assay (ELISA) and key transcription factors were detected by Western blotting. Results Administration of rSjcystatin significantly reduced inflammatory parameters and ameliorated the severity of the TNBS-induced colitis through decreasing IFNγ in three organs and lifting the level of IL-4, IL-13, IL-10, and TGF-β in the colon tissues, with uptrending Tregs in the MLN and LPMC. Conclusion The findings provide evidence that rSjcystatin has a therapeutic potential for diminishing colitis inflammation in Balb/c mice. The immunological mechanism may involve the down-regulation of Th1 response and up-regulation of Th2 and Tregs in the MLN and colon.
Collapse
Affiliation(s)
- Shushu Wang
- Department of Immunology, Anhui Medical University, Hefei, 230022, China. .,Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, Hefei, 230022, China. .,Pediatrics Department of Affiliated Provincial Hospital, Anhui Medical University, Hefei, 230001, China.
| | - Yuanyuan Xie
- Department of Immunology, Anhui Medical University, Hefei, 230022, China. .,Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, Hefei, 230022, China.
| | - Xiaodi Yang
- Department of Microbiology and Parasitology, Bengbu Medical College; Anhui Key Laboratory of Infection and Immunity, Bengbu, 233000, Anhui, China.
| | - Xuesong Wang
- Pediatrics Department of Affiliated Provincial Hospital, Anhui Medical University, Hefei, 230001, China.
| | - Ke Yan
- Department of Laboratory Diagnosis, the Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.
| | - Zhengrong Zhong
- Department of Laboratory Diagnosis, the Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.
| | - Xiaowei Wang
- Department of Laboratory Diagnosis, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yuanhong Xu
- Department of Laboratory Diagnosis, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yi Zhang
- Department of Immunology, Anhui Medical University, Hefei, 230022, China. .,Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, Hefei, 230022, China.
| | - Fang Liu
- Department of Immunology, Anhui Medical University, Hefei, 230022, China. .,Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, Hefei, 230022, China.
| | - Jilong Shen
- Department of Immunology, Anhui Medical University, Hefei, 230022, China. .,Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, Hefei, 230022, China.
| |
Collapse
|
24
|
Khatri V, Amdare N, Tarnekar A, Goswami K, Reddy MVR. Brugia malayi cystatin therapeutically ameliorates dextran sulfate sodium-induced colitis in mice. J Dig Dis 2015; 16:585-94. [PMID: 26358507 DOI: 10.1111/1751-2980.12290] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 08/25/2015] [Accepted: 09/09/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Helminth immunomodulation in the host has been shown to have therapeutic implications in inflammatory bowel diseases. In this study we aimed to evaluate the therapeutic effect of Brugia malayi recombinant cystatin (rBmCys) in a dose-dependent manner on dextran sulfate sodium (DSS)-induced colitis in mice. METHODS The anti-inflammatory activity of rBmCys on mice peritoneal exudate cells was initially analyzed in vitro. BALB/c mice were fed with 5% DSS for 7 days to induce colitis. The colitis mice were treated intraperitoneally with rBmCys (10, 25 or 50 µg for the three different groups of mice) on days 1, 3 and 5 of the DSS administration. Disease severity was assessed by the disease activity index (DAI) and macroscopic and histopathological scores of colon and myeloperoxidase activity in colonic mucosa. Cytokine profiles were measured in sera and cultured splenocytes of treated mice followed by stimulation with rBmCys. RESULTS rBmCys showed anti-inflammatory activity in vitro. Treatment of DSS-induced colitis with rBmCys in mice ameliorated the overall disease severity as reflected by a significant reduction in weight loss, the DAI, mucosal edema, colon damage and myeloperoxidase activity of the colonic mucosa. While the mRNA expressions of IFN-γ, TNF-α, interleukin (IL)-5, IL-6 and IL-17 were downregulated, IL-10 expression was upregulated in the splenocytes of colitis mice treated with rBmCys. The amelioration of DSS-induced colitis occurred in a dose-dependent manner. CONCLUSION The results of this study indicate an anti-inflammatory potential of rBmCys and provide evidence for using this protein as a promising therapeutic agent in ulcerative colitis.
Collapse
Affiliation(s)
- Vishal Khatri
- Department of Biochemistry & JB Tropical Disease Research Center, Mahatma Gandhi Institute of Medical Sciences, Maharashtra, India
| | - Nitin Amdare
- Department of Biochemistry & JB Tropical Disease Research Center, Mahatma Gandhi Institute of Medical Sciences, Maharashtra, India
| | - Aaditya Tarnekar
- Department of Anatomy, Mahatma Gandhi Institute of Medical Sciences, Maharashtra, India
| | - Kalyan Goswami
- Department of Biochemistry & JB Tropical Disease Research Center, Mahatma Gandhi Institute of Medical Sciences, Maharashtra, India
| | - Maryada Venkata Rami Reddy
- Department of Biochemistry & JB Tropical Disease Research Center, Mahatma Gandhi Institute of Medical Sciences, Maharashtra, India
| |
Collapse
|
25
|
Siles-Lucas M, Morchon R, Simon F, Manzano-Roman R. Exosome-transported microRNAs of helminth origin: new tools for allergic and autoimmune diseases therapy? Parasite Immunol 2015; 37:208-14. [PMID: 25712154 DOI: 10.1111/pim.12182] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/17/2015] [Indexed: 12/11/2022]
Abstract
Chronic diseases associated with inflammation show fast annual increase in their incidence. This has been associated with excessive hygiene habits that limit contacts between the immune system and helminth parasites. Helminthic infections induce regulation and expansion of regulatory T cells (Treg) leading to atypical Th2 type immune responses, with downregulation of the inflammatory component usually associated with these type of responses. Many cells, including those of the immune system, produce extracellular vesicles called exosomes which mediate either immune stimulation (DCs) or immune modulation (T cells). The transfer of miRNAs contained in T-cell exosomes has been shown to contribute to downregulate the production of inflammatory mediators. It has been recently described the delivery to the host-parasite interface of exosomes containing miRNAs by helminths and its internalization by host cells. In this sense, helminth microRNAs transported in exosomes and internalized by immune host cells exert an important role in the expansion of Treg cells, resulting in the control of inflammation. We here provide relevant information obtained in the field of exosomes, cell-cell communication and miRNAs, showing the high potential of helminth miRNAs delivered in exosomes to host cells as new therapeutic tools against diseases associated with exacerbated inflammatory responses.
Collapse
Affiliation(s)
- M Siles-Lucas
- Instituto de Recursos Naturales y Agrobiologia de Salamanca (IRNASA-CSIC), Salamanca, Spain
| | | | | | | |
Collapse
|
26
|
Extraintestinal Helminth Infection Limits Pathology and Proinflammatory Cytokine Expression during DSS-Induced Ulcerative Colitis: A Role for Alternatively Activated Macrophages and Prostaglandins. BIOMED RESEARCH INTERNATIONAL 2015; 2015:563425. [PMID: 26090422 PMCID: PMC4450267 DOI: 10.1155/2015/563425] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/14/2014] [Accepted: 10/20/2014] [Indexed: 01/05/2023]
Abstract
Chronic inflammation of the intestinal mucosa is characteristic of inflammatory bowel diseases such as ulcerative colitis and Crohn's disease. Helminth parasites have developed immunomodulatory strategies that may impact the outcome of several inflammatory diseases. Therefore, we investigated whether Taenia crassiceps infection is able to decrease the inflammatory effects of dextran sulfate sodium- (DSS-) induced ulcerative colitis in BALB/c and C57BL/6 mice. Preinfection significantly reduced the manifestations of DSS-induced colitis, as weight loss and shortened colon length, and decreased the disease activity index independently of the genetic background of the mice. Taenia infection decreased systemic levels of proinflammatory cytokines while increasing levels of IL-4 and IL-10, and the inflammatory infiltrate into the colon was also markedly reduced. RT-PCR assays from colon showed that T. crassiceps-infected mice displayed increased expression of Arginase-1 but decreased expression of iNOS compared to DSS-treated uninfected mice. The percentages of T regulatory cells were not increased. The adoptive transfer of alternatively activated macrophages (AAMФs) from infected mice into mice with DSS-induced colitis reduced the severity of colon inflammation. Administration of indomethacin abrogated the anticolitic effect of Taenia. Thus, T. crassiceps infection limits the pathology of ulcerative colitis by suppressing inflammatory responses mechanistically associated with AAMФs and prostaglandins.
Collapse
|
27
|
Heylen M, Ruyssers NE, Gielis EM, Vanhomwegen E, Pelckmans PA, Moreels TG, De Man JG, De Winter BY. Of worms, mice and man: an overview of experimental and clinical helminth-based therapy for inflammatory bowel disease. Pharmacol Ther 2014; 143:153-167. [PMID: 24603369 DOI: 10.1016/j.pharmthera.2014.02.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 12/17/2022]
Abstract
The incidence of inflammatory and autoimmune disorders is highest in well-developed countries which is directly related to their higher hygienic standards: it is suggested that the lack of exposure to helminths contributes to the susceptibility for immune-related diseases. Epidemiological, experimental and clinical data support the idea that helminths provide protection against immune-mediated diseases such as inflammatory bowel disease (IBD). The most likely mechanism for the suppression of immune responses by helminths is the release of helminth-derived immunomodulatory molecules. This article reviews the experimental and clinical studies investigating the therapeutic potential of helminth-based therapy in IBD and also focuses on the current knowledge of its immunomodulatory mechanisms of action highlighting innate as well as adaptive immune mechanisms. Identifying the mechanisms by which these helminths and helminth-derived molecules modulate the immune system will help in creating novel drugs for the treatment of IBD and other disorders that result from an overactive immune response.
Collapse
Affiliation(s)
- Marthe Heylen
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Nathalie E Ruyssers
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Els M Gielis
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Els Vanhomwegen
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Paul A Pelckmans
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Division of Gastroenterology & Hepatology, Antwerp, Belgium
| | - Tom G Moreels
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Division of Gastroenterology & Hepatology, Antwerp, Belgium
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
28
|
Yang X, Yang Y, Wang Y, Zhan B, Gu Y, Cheng Y, Zhu X. Excretory/secretory products from Trichinella spiralis adult worms ameliorate DSS-induced colitis in mice. PLoS One 2014; 9:e96454. [PMID: 24788117 PMCID: PMC4008629 DOI: 10.1371/journal.pone.0096454] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 04/08/2014] [Indexed: 12/19/2022] Open
Abstract
Background Many evidences show the inverse correlation between helminth infection and allergic or autoimmune diseases. Identification and characterization of the active helminth-derived products responsible for the beneficial effects on allergic or inflammatory diseases will provide another feasible approach to treat these diseases. Methods and Findings Colitis was induced in C57BL/6 mice by giving 3% DSS orally for 7 days. During this period, the mice were treated daily with the excretory/secretory products from T. spiralis adult worms (AES) intraperitoneally. The severity of colitis was monitored by measuring body weight, stool consistency or bleeding, colon length and inflammation. To determine the T. spiralis AES product-induced immunological response, Th1, Th2, Th17 and regulatory cytokine profiles were measured in lymphocytes isolated from colon, mesenteric lymph nodes (MLN), and the spleen of treated mice. The CD4+ CD25+ FOXP3+ regulatory T cells (Tregs) were also measured in the spleens and MLN of treated mice. Mice treated with AES significantly ameliorated the severity of the DSS-induced colitis indicated by the reduced disease manifestations, improved macroscopic and microscopic inflammation correlated with the up-regulation of Treg response (increased regulatory cytokines IL-10, TGF-beta and regulatory T cells) and down-regulation of pro-inflammatory cytokines (IFN-gamma, IL-6 and IL-17) in the spleens, MLN and colon of treated mice. Conclusions Our results provide direct evidences that T. spiralis AES have a therapeutic potential for alleviating inflammatory colitis in mice. This effect is possibly mediated by the immunomodulation of regulatory T cells to produce regulatory and anti-inflammatory cytokines and inhibit pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Xiaodi Yang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Microbiology and Parasitology, Bengbu Medical College; Anhui Key Laboratory of Infection and Immunity, Bengbu, Anhui, China
| | - Yaping Yang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yunyun Wang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bin Zhan
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Yuan Gu
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuli Cheng
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinping Zhu
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- * E-mail:
| |
Collapse
|
29
|
Heligmosomoides polygyrus infection reduces severity of type 1 diabetes induced by multiple low-dose streptozotocin in mice via STAT6- and IL-10-independent mechanisms. Exp Parasitol 2013; 135:388-96. [DOI: 10.1016/j.exppara.2013.08.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/01/2013] [Accepted: 08/09/2013] [Indexed: 12/11/2022]
|
30
|
Abstract
Modern hygienic lifestyles are associated with the emergence of inflammatory bowel disease (IBD) which now afflicts millions of people in highly-developed countries. Meticulous hygiene interrupts conduits of transmission required for ubiquitous exposure to parasitic worms (helminths). We proposed that loss of exposure to helminths permits development of IBD. Early clinical trials suggested that exposure to helminths such as Trichuris suis or Necator americanus can improve IBD. Over the last several years, processes to "medicinalize"T. suis have been developed and use of this helminth is now being studied in large multi-center clinical trials. Concurrently, we and others have identified some of the immune regulatory mechanisms elicited by helminth exposure that suppress inappropriate intestinal inflammation. These efforts could soon result in new therapies for patients with IBD.
Collapse
Affiliation(s)
- Joel V Weinstock
- Division of Gastroenterology, Tufts Medical Center, Boston, MA, USA.
| | | |
Collapse
|
31
|
Abstract
Helminth parasites infect almost one-third of the world's population, primarily in tropical regions. However, regions where helminth parasites are endemic record much lower prevalences of allergies and autoimmune diseases, suggesting that parasites may protect against immunopathological syndromes. Most helminth diseases are spectral in nature, with a large proportion of relatively asymptomatic cases and a subset of patients who develop severe pathologies. The maintenance of the asymptomatic state is now recognized as reflecting an immunoregulatory environment, which may be promoted by parasites, and involves multiple levels of host regulatory cells and cytokines; a breakdown of this regulation is observed in pathological disease. Currently, there is much interest in whether helminth-associated immune regulation may ameliorate allergy and autoimmunity, with investigations in both laboratory models and human trials. Understanding and exploiting the interactions between these parasites and the host regulatory network are therefore likely to highlight new strategies to control both infectious and immunological diseases.
Collapse
|