1
|
Chakraborty A, Bayry J, Mukherjee S. Helminth-derived biomolecules as potential therapeutics against ulcerative colitis. Immunotherapy 2024; 16:635-640. [PMID: 38888436 PMCID: PMC11404699 DOI: 10.1080/1750743x.2024.2360382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Affiliation(s)
- Ankita Chakraborty
- Integrative Biochemistry & Immunology Laboratory (IBIL), Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, 713340, India
| | - Jagadeesh Bayry
- Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678623, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory (IBIL), Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, 713340, India
| |
Collapse
|
2
|
Truscott J, Guan X, Fury H, Atagozli T, Metwali A, Liu W, Li Y, Li RW, Elliott DE, Blazar BR, Ince MN. After Bone Marrow Transplantation, the Cell-Intrinsic Th2 Pathway Promotes Recipient T Lymphocyte Survival and Regulates Graft-versus-Host Disease. Immunohorizons 2023; 7:442-455. [PMID: 37294277 PMCID: PMC10580113 DOI: 10.4049/immunohorizons.2300021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/10/2023] Open
Abstract
Recipient T cells can aggravate or regulate lethal and devastating graft-versus-host disease (GVHD) after bone marrow transplantation (BMT). In this context, we have shown before that intestinal immune conditioning with helminths is associated with survival of recipient T cells and Th2 pathway-dependent regulation of GVHD. We investigated the mechanism of survival of recipient T cells and their contribution to GVHD pathogenesis in this helminth infection and BMT model after myeloablative preparation with total body irradiation in mice. Our results indicate that the helminth-induced Th2 pathway directly promotes the survival of recipient T cells after total body irradiation. Th2 cells also directly stimulate recipient T cells to produce TGF-β, which is required to regulate donor T cell-mediated immune attack of GVHD and can thereby contribute to recipient T cell survival after BMT. Moreover, we show that recipient T cells, conditioned to produce Th2 cytokines and TGF-β after helminth infection, are fundamentally necessary for GVHD regulation. Taken together, reprogrammed or immune-conditioned recipient T cells after helminth infection are crucial elements of Th2- and TGF-β-dependent regulation of GVHD after BMT, and their survival is dependent on cell-intrinsic Th2 signaling.
Collapse
Affiliation(s)
- Jamie Truscott
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Xiaoqun Guan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Veterans Administration Medical Center, Iowa City, IA
| | - Hope Fury
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Veterans Administration Medical Center, Iowa City, IA
| | - Tyler Atagozli
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Veterans Administration Medical Center, Iowa City, IA
| | - Ahmed Metwali
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Veterans Administration Medical Center, Iowa City, IA
| | - Weiren Liu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Veterans Administration Medical Center, Iowa City, IA
| | - Yue Li
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Veterans Administration Medical Center, Iowa City, IA
| | - Robert W. Li
- Animal Parasitic Diseases Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD
| | - David E. Elliott
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Veterans Administration Medical Center, Iowa City, IA
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - M. Nedim Ince
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Veterans Administration Medical Center, Iowa City, IA
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
3
|
Atagozli T, Elliott DE, Ince MN. Helminth Lessons in Inflammatory Bowel Diseases (IBD). Biomedicines 2023; 11:1200. [PMID: 37189818 PMCID: PMC10135676 DOI: 10.3390/biomedicines11041200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Helminths are multicellular invertebrates that colonize the gut of many vertebrate animals including humans. This colonization can result in pathology, which requires treatment. It can also lead to a commensal and possibly even a symbiotic relationship where the helminth and the host benefit from each other's presence. Epidemiological data have linked helminth exposure to protection from immune disorders that include a wide range of diseases, such as allergies, autoimmune illnesses, and idiopathic inflammatory disorders of the gut, which are grouped as inflammatory bowel diseases (IBD). Treatment of moderate to severe IBD involves the use of immune modulators and biologics, which can cause life-threatening complications. In this setting, their safety profile makes helminths or helminth products attractive as novel therapeutic approaches to treat IBD or other immune disorders. Helminths stimulate T helper-2 (Th2) and immune regulatory pathways, which are targeted in IBD treatment. Epidemiological explorations, basic science studies, and clinical research on helminths can lead to the development of safe, potent, and novel therapeutic approaches to prevent or treat IBD in addition to other immune disorders.
Collapse
Affiliation(s)
- Tyler Atagozli
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
| | - David E. Elliott
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - Mirac Nedim Ince
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| |
Collapse
|
4
|
Abstract
Transforming Growth Factor-β is a potent regulator of the immune system, acting at every stage from thymic differentiation, population of the periphery, control of responsiveness, tissue repair and generation of memory. It is therefore a central player in the immune response to infectious pathogens, but its contribution is often clouded by multiple roles acting on different cells in time and space. Hence, context is all-important in understanding when TGF-β is beneficial or detrimental to the outcome of infection. In this review, a full range of infectious agents from viruses to helminth parasites are explored within this framework, drawing contrasts and general conclusions about the importance of TGF-β in these diseases.
Collapse
Affiliation(s)
- Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
5
|
Hang L, Kumar S, Blum AM, Urban JF, Fantini MC, Weinstock JV. Heligmosomoides polygyrus bakeri Infection Decreases Smad7 Expression in Intestinal CD4 + T Cells, Which Allows TGF-β to Induce IL-10-Producing Regulatory T Cells That Block Colitis. THE JOURNAL OF IMMUNOLOGY 2019; 202:2473-2481. [PMID: 30850474 DOI: 10.4049/jimmunol.1801392] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/04/2019] [Indexed: 12/25/2022]
Abstract
Helminthic infections modulate host immunity and may protect their hosts from developing immunological diseases like inflammatory bowel disease. Induction of regulatory T cells (Tregs) may be an important part of this protective process. Heligmosomoides polygyrus bakeri infection also promotes the production of the regulatory cytokines TGF-β and IL-10 in the gut. In the intestines, TGF-β helps induce regulatory T cells. This study used Foxp3/IL-10 double reporter mice to investigate the effect of TGF-β on the differentiation of colon and mesenteric lymph node-derived murine Foxp3- IL-10- CD4+ T cells into their regulatory phenotypes. Foxp3- IL-10- CD4+ T cells from H. polygyrus bakeri-infected mice, as opposed to T cells from uninfected animals, cultured in vitro with TGF-β and anti-CD3/CD28 mAb differentiated into Foxp3+ and/or IL-10+ T cells. The IL-10-producing T cells nearly all displayed CD25. Smad7 is a natural inhibitor of TGF-β signaling. In contrast to gut T cells from uninfected mice, Foxp3- IL10- CD4+ T cells from H. polygyrus bakeri-infected mice displayed reduced Smad7 expression and responded to TGF-β with Smad2/3 phosphorylation. The TGF-β-induced Tregs that express IL-10 blocked colitis when transferred into the Rag/CD25- CD4+ T cell transfer model of inflammatory bowel disease. TGF-β had a greatly diminished capacity to induce Tregs in H. polygyrus bakeri-infected transgenic mice with constitutively high T cell-specific Smad7 expression. Thus, infection with H. polygyrus bakeri causes down-modulation in Smad7 expression in intestinal CD4+ T cells, which allows the TGF-β produced in response to the infection to induce the Tregs that prevent colitis.
Collapse
Affiliation(s)
- Long Hang
- Division of Gastroenterology/Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111
| | - Sangeeta Kumar
- Division of Gastroenterology/Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111
| | - Arthur M Blum
- Division of Gastroenterology/Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111
| | - Joseph F Urban
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705; and
| | - Massimo C Fantini
- Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Joel V Weinstock
- Division of Gastroenterology/Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111;
| |
Collapse
|
6
|
Li Y, Guan X, Liu W, Chen HL, Truscott J, Beyatli S, Metwali A, Weiner GJ, Zavazava N, Blumberg RS, Urban JF, Blazar BR, Elliott DE, Ince MN. Helminth-Induced Production of TGF-β and Suppression of Graft-versus-Host Disease Is Dependent on IL-4 Production by Host Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:2910-2922. [PMID: 30291167 DOI: 10.4049/jimmunol.1700638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/07/2018] [Indexed: 02/07/2023]
Abstract
Helminths stimulate the secretion of Th2 cytokines, like IL-4, and suppress lethal graft-versus-host disease (GVHD) after bone marrow transplantation. This suppression depends on the production of immune-modulatory TGF-β and is associated with TGF-β-dependent in vivo expansion of Foxp3+ regulatory T cells (Treg). In vivo expansion of Tregs is under investigation for its potential as a therapy for GVHD. Nonetheless, the mechanism of induced and TGF-β-dependent in vivo expansion of Tregs, in a Th2 polarized environment after helminth infection, is unknown. In this study, we show that helminth-induced IL-4 production by host cells is critical to the induction and maintenance of TGF-β secretion, TGF-β-dependent expansion of Foxp3+ Tregs, and the suppression of GVHD. In mice with GVHD, the expanding donor Tregs express the Th2-driving transcription factor, GATA3, which is required for helminth-induced production of IL-4 and TGF-β. In contrast, TGF-β is not necessary for GATA3 expression by Foxp3+ Tregs or by Foxp3- CD4 T cells. Various cell types of innate or adaptive immune compartments produce high quantities of IL-4 after helminth infection. As a result, IL-4-mediated suppression of GVHD does not require invariant NKT cells of the host, a cell type known to produce IL-4 and suppress GVHD in other models. Thus, TGF-β generation, in a manner dependent on IL-4 secretion by host cells and GATA3 expression, constitutes a critical effector arm of helminthic immune modulation that promotes the in vivo expansion of Tregs and suppresses GVHD.
Collapse
Affiliation(s)
- Yue Li
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Xiaoqun Guan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Weiren Liu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Hung-Lin Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Jamie Truscott
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Sonay Beyatli
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Ahmed Metwali
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - George J Weiner
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242.,Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Nicholas Zavazava
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242.,Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Richard S Blumberg
- Department of Internal Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Joseph F Urban
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705; and
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | - David E Elliott
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242.,Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - M Nedim Ince
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; .,Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
7
|
Li Y, Liu W, Guan X, Truscott J, Creemers JW, Chen HL, Pesu M, El Abiad RG, Karacay B, Urban JF, Elliott DE, Kaplan MH, Blazar BR, Ince MN. STAT6 and Furin Are Successive Triggers for the Production of TGF-β by T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:2612-2623. [PMID: 30266770 DOI: 10.4049/jimmunol.1700808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/03/2018] [Indexed: 01/11/2023]
Abstract
Production of TGF-β by T cells is key to various aspects of immune homeostasis, with defects in this process causing or aggravating immune-mediated disorders. The molecular mechanisms that lead to TGF-β generation by T cells remain largely unknown. To address this issue, we take advantage of the fact that intestinal helminths stimulate Th2 cells besides triggering TGF-β generation by T lymphocytes and regulate immune-mediated disorders. We show that the Th2 cell-inducing transcription factor STAT6 is necessary and sufficient for the expression of TGF-β propeptide in T cells. STAT6 is also necessary for several helminth-triggered events in mice, such as TGF-β-dependent suppression of alloreactive inflammation in graft-versus-host disease. Besides STAT6, helminth-induced secretion of active TGF-β requires cleavage of propeptide by the endopeptidase furin. Thus, for the immune regulatory pathway necessary for TGF-β production by T cells, our results support a two-step model, composed of STAT6 and furin.
Collapse
Affiliation(s)
- Yue Li
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Weiren Liu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Xiaqun Guan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Jamie Truscott
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - John W Creemers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven, B-3000 Belgium
| | - Hung-Lin Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Marko Pesu
- Immunoregulation, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, FI-33520 Tampere, Finland.,Department of Dermatology, Tampere University Hospital, FI-33520 Tampere, Finland
| | - Rami G El Abiad
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Bahri Karacay
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Joseph F Urban
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705
| | - David E Elliott
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Mark H Kaplan
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research and Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455; and
| | - M Nedim Ince
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; .,Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
8
|
Zhou S, Qi Q, Wang X, Zhang L, Xu L, Dong L, Zhu J, Li Y, Wang X, Xu Z, Liu F, Hu W, Zhou L, Chen X, Su C. SjHSP60 induces CD4 + CD25 + Foxp3 + Tregs via TLR4-Mal-drived production of TGF-β in macrophages. Immunol Cell Biol 2018; 96:958-968. [PMID: 29697865 DOI: 10.1111/imcb.12160] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 03/25/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
CD4+ CD25+ Foxp3+ regulatory T cells (Tregs) play a pivotal role in limiting immunopathological damage to host organs after schistosome infection. Transforming growth factor-β (TGF-β) is an essential factor for the periphery conversion of CD4+ CD25- T cells into CD4+ CD25+ Foxp3+ Tregs by inducing the key transcription factor Foxp3. Antigen presenting cells (APCs), which highly express TGF-β, are involved in parasite antigen-induced Treg conversion in peripheral. However, the mechanisms underlying high TGF-β induction in APCs by parasite antigens remain to be clarified during schistosome infection. Here, we demonstrated that Schistosoma japonicum stress protein, heat shock protein 60 (SjHSP60), promoted TGF-β production in macrophages (Mφ). Furthermore, we showed that activation of TLR4-Mal (MyD88 adaptor-like protein) signaling by SjHSP60 is necessary for induction of TGF-β expression in Mφ, which subsequently promoted Treg induction. Our results not only demonstrate a novel mechanism of TGF-β production in Mφ for inducing Tregs in mice with schistosomiasis, but also allude to the possibility of targeting parasite stress protein for potential therapeutics.
Collapse
Affiliation(s)
- Sha Zhou
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianqian Qi
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaofan Wang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lina Zhang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Xu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liyang Dong
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuefeng Wang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhipeng Xu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Liu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Hu
- Department of Microbiology and Microbial Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, USA
| | - Xiaojun Chen
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Su
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Leung JM, Graham AL, Knowles SCL. Parasite-Microbiota Interactions With the Vertebrate Gut: Synthesis Through an Ecological Lens. Front Microbiol 2018; 9:843. [PMID: 29867790 PMCID: PMC5960673 DOI: 10.3389/fmicb.2018.00843] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/12/2018] [Indexed: 12/14/2022] Open
Abstract
The vertebrate gut teems with a large, diverse, and dynamic bacterial community that has pervasive effects on gut physiology, metabolism, and immunity. Under natural conditions, these microbes share their habitat with a similarly dynamic community of eukaryotes (helminths, protozoa, and fungi), many of which are well-known parasites. Both parasites and the prokaryotic microbiota can dramatically alter the physical and immune landscape of the gut, creating ample opportunities for them to interact. Such interactions may critically alter infection outcomes and affect overall host health and disease. For instance, parasite infection can change how a host interacts with its bacterial flora, either driving or protecting against dysbiosis and inflammatory disease. Conversely, the microbiota can alter a parasite's colonization success, replication, and virulence, shifting it along the parasitism-mutualism spectrum. The mechanisms and consequences of these interactions are just starting to be elucidated in an emergent transdisciplinary area at the boundary of microbiology and parasitology. However, heterogeneity in experimental designs, host and parasite species, and a largely phenomenological and taxonomic approach to synthesizing the literature have meant that common themes across studies remain elusive. Here, we use an ecological perspective to review the literature on interactions between the prokaryotic microbiota and eukaryotic parasites in the vertebrate gut. Using knowledge about parasite biology and ecology, we discuss mechanisms by which they may interact with gut microbes, the consequences of such interactions for host health, and how understanding parasite-microbiota interactions may lead to novel approaches in disease control.
Collapse
Affiliation(s)
- Jacqueline M Leung
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, United States
| | - Andrea L Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, United States
| | | |
Collapse
|
10
|
Zakeri A, Borji H, Haghparast A. Interaction Between Helminths and Toll-Like Receptors: Possibilities and Potentials for Asthma Therapy. Int Rev Immunol 2016; 35:219-48. [PMID: 27120222 DOI: 10.3109/08830185.2015.1096936] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Toll-like receptors (TLRs) are essential components of the innate immune system. They play an important role in the pathogenesis of allergic diseases, especially asthma. Since TLRs significantly orchestrate innate and adaptive immune response, their manipulation has widely been considered as a potential approach to control asthma symptoms. It is well established that helminths have immunoregulatory effects on host immune responses, especially innate immunity. They release bioactive molecules such as excretory-secretory (ES) products manipulating TLRs expression and signaling. Thus, given the promising results derived from preclinical studies, harnessing helminth-derived molecules affecting TLRs can be considered as a potential biological therapy for allergic diseases. Prospectively, the data that are available at present suggest that, in the near future, it is possible that helminth antigens will offer new therapeutic strategies and druggable targets for fighting allergic diseases. This review describes the interactions between helminths and TLRs and discusses the potential possibilities for asthma therapy. In this opinion paper, the authors aimed to review the updated literatures on the interplay between helminths, TLRs, and asthma with a view to proposing helminth-based asthma therapy.
Collapse
Affiliation(s)
- Amin Zakeri
- a Parasitology Section, Department of Pathobiology , Faculty of Veterinary Medicine, Ferdowsi University of Mashhad , Mashhad , Iran.,b Immunology Sections, Department of Pathobiology, Faculty of Veterinary Medicine , Faculty of Veterinary Medicine, Ferdowsi University of Mashhad , Mashhad , Iran
| | - Hassan Borji
- a Parasitology Section, Department of Pathobiology , Faculty of Veterinary Medicine, Ferdowsi University of Mashhad , Mashhad , Iran
| | - Alireza Haghparast
- b Immunology Sections, Department of Pathobiology, Faculty of Veterinary Medicine , Faculty of Veterinary Medicine, Ferdowsi University of Mashhad , Mashhad , Iran.,c Biotechnology Section, Department of Pathobiology , Faculty of Veterinary Medicine, Ferdowsi University of Mashhad , Mashhad , Iran
| |
Collapse
|
11
|
Reynolds LA, Finlay BB, Maizels RM. Cohabitation in the Intestine: Interactions among Helminth Parasites, Bacterial Microbiota, and Host Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 195:4059-66. [PMID: 26477048 DOI: 10.4049/jimmunol.1501432] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Both intestinal helminth parasites and certain bacterial microbiota species have been credited with strong immunomodulatory effects. Recent studies reported that the presence of helminth infection alters the composition of the bacterial intestinal microbiota and, conversely, that the presence and composition of the bacterial microbiota affect helminth colonization and persistence within mammalian hosts. This article reviews recent findings on these reciprocal relationships, in both human populations and mouse models, at the level of potential mechanistic pathways and the implications these bear for immunomodulatory effects on allergic and autoimmune disorders. Understanding the multidirectional complex interactions among intestinal microbes, helminth parasites, and the host immune system allows for a more holistic approach when using probiotics, prebiotics, synbiotics, antibiotics, and anthelmintics, as well as when designing treatments for autoimmune and allergic conditions.
Collapse
Affiliation(s)
- Lisa A Reynolds
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; and
| | - Rick M Maizels
- Centre for Immunity, Infection, and Evolution, Institute of Immunology and Infection Research, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| |
Collapse
|
12
|
|
13
|
Neither primary nor memory immunity to Mycobacterium tuberculosis infection is compromised in mice with chronic enteric helminth infection. Infect Immun 2015; 83:1217-23. [PMID: 25605766 DOI: 10.1128/iai.03004-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Previously we had reported that Nippostrongylus brasiliensis, a helminth with a lung migratory phase, affected host resistance against Mycobacterium tuberculosis infection through the induction of alternatively activated (M2) macrophages. Several helminth species do not have an obligatory lung migratory phase but establish chronic infections in the host that include potent immune downregulatory effects, in part mediated through induction of a FoxP3(+) T regulatory cell (Treg) response. Treg cells exhibit duality in their functions in host defense against M. tuberculosis infection since their depletion leads to enhanced priming of T cells in the lymph nodes and attendant improved control of M. tuberculosis infection, while their presence in the lung granuloma protects against excessive inflammation. Heligmosomoides polygyrus is a strictly murine enteric nematode that induces a strong FoxP3 Treg response in the host. Therefore, in this study we investigated whether host immunity to M. tuberculosis infection would be modulated in mice with chronic H. polygyrus infection. We report that neither primary nor memory immunity conferred by Mycobacterium bovis BCG vaccination was affected in mice with chronic enteric helminth infection, despite a systemic increase in FoxP3(+) T regulatory cells. The findings indicate that anti-M. tuberculosis immunity is not similarly affected by all helminth species and highlight the need to consider this inequality in human coinfection studies.
Collapse
|
14
|
Li Y, Chen HL, Bannick N, Henry M, Holm AN, Metwali A, Urban JF, Rothman PB, Weiner GJ, Blazar BR, Elliott DE, Ince MN. Intestinal helminths regulate lethal acute graft-versus-host disease and preserve the graft-versus-tumor effect in mice. THE JOURNAL OF IMMUNOLOGY 2014; 194:1011-20. [PMID: 25527786 DOI: 10.4049/jimmunol.1303099] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Donor T lymphocyte transfer with hematopoietic stem cells suppresses residual tumor growth (graft-versus-tumor [GVT]) in cancer patients undergoing bone marrow transplantation (BMT). However, donor T cell reactivity to host organs causes severe and potentially lethal inflammation called graft-versus-host disease (GVHD). High-dose steroids or other immunosuppressive drugs are used to treat GVHD that have limited ability to control the inflammation while incurring long-term toxicity. Novel strategies are needed to modulate GVHD, preserve GVT, and improve the outcome of BMT. Regulatory T cells (Tregs) control alloantigen-sensitized inflammation of GVHD, sustain GVT, and prevent mortality in BMT. Helminths colonizing the alimentary tract dramatically increase the Treg activity, thereby modulating intestinal or systemic inflammatory responses. These observations led us to hypothesize that helminths can regulate GVHD and maintain GVT in mice. Acute GVHD was induced in helminth (Heligmosomoides polygyrus)-infected or uninfected BALB/c recipients of C57BL/6 donor grafts. Helminth infection suppressed donor T cell inflammatory cytokine generation and reduced GVHD-related mortality, but maintained GVT. H. polygyrus colonization promoted the survival of TGF-β-generating recipient Tregs after a conditioning regimen with total body irradiation and led to a TGF-β-dependent in vivo expansion/maturation of donor Tregs after BMT. Helminths did not control GVHD when T cells unresponsive to TGF-β-mediated immune regulation were used as donor T lymphocytes. These results suggest that helminths suppress acute GVHD using Tregs and TGF-β-dependent pathways in mice. Helminthic regulation of GVHD and GVT through intestinal immune conditioning may improve the outcome of BMT.
Collapse
Affiliation(s)
- Yue Li
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Hung-Lin Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Nadine Bannick
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Michael Henry
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Adrian N Holm
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Ahmed Metwali
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Joseph F Urban
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705; and
| | - Paul B Rothman
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - George J Weiner
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | - David E Elliott
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - M Nedim Ince
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242;
| |
Collapse
|
15
|
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: 5.8] [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
|
16
|
Zaph C, Cooper PJ, Harris NL. Mucosal immune responses following intestinal nematode infection. Parasite Immunol 2014; 36:439-52. [PMID: 25201407 PMCID: PMC4312905 DOI: 10.1111/pim.12090] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/05/2013] [Indexed: 12/12/2022]
Abstract
In most natural environments, the large majority of mammals harbour parasitic helminths that often live as adults within the intestine for prolonged periods (1-2 years). Although these organisms have been eradicated to a large extent within westernized human populations, those living within rural areas of developing countries continue to suffer from high infection rates. Indeed, recent estimates indicate that approximately 2.5 billion people worldwide, mainly children, currently suffer from infection with intestinal helminths (also known as geohelminths and soil-transmitted helminths) . Paradoxically, the eradication of helminths is thought to contribute to the increased incidence of autoimmune diseases and allergy observed in developed countries. In this review, we will summarize our current understanding of host-helminth interactions at the mucosal surface that result in parasite expulsion or permit the establishment of chronic infections with luminal dwelling adult worms. We will also provide insight into the adaptive immune mechanisms that provide immune protection against re-infection with helminth larvae, a process that is likely to be key to the future development of successful vaccination strategies. Lastly, the contribution of helminths to immune modulation and particularly to the treatment of allergy and inflammatory bowel disease will be discussed.
Collapse
Affiliation(s)
- C Zaph
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
17
|
Incidence of active pulmonary tuberculosis in patients with coincident filarial and/or intestinal helminth infections followed longitudinally in South India. PLoS One 2014; 9:e94603. [PMID: 24728010 PMCID: PMC3984271 DOI: 10.1371/journal.pone.0094603] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/18/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Filarial (and other helminth) infections are known to modulate mycobacteria-specific pro-inflammatory cytokine responses necessary for maintaining latency in tuberculosis (TB). We sought to address whether helminth co-infection alters progression to active pulmonary TB in a co-endemic area of South India. METHODS/PRINCIPAL FINDINGS Incidence of active pulmonary TB was assessed in 5096 subjects from five villages among helminth-infected (hel⁺) and -uninfected (hel⁻) groups. Baseline stool examinations, circulating filarial antigen, and tuberculin skin testing (PPD) were performed along with chest radiographs, sputum microscopy, and culture. During three follow-up visits each 2.5 years, patients were assessed using PPD tests and questionnaires and--for those with potential symptoms of TB--sputum microscopy and culture. Of the 5096 subjects, 1923 were found to be hel⁺ and 3173 were hel⁻. Follow up interval stool examination could not be performed. In each group, 21 developed active TB over the course of the study. After adjusting for sex, age, BCG vaccination status, and PPD positivity, no difference was seen in active TB incidence between hel⁺ and hel- groups either at baseline (relative risk (RR) 1.60; 95% confidence interval (CI): 0.69, 3.71, P = 0·27), or when followed prospectively (RR 1.24; 95% CI: 0.48, 3.18, P = 0·66). CONCLUSIONS/SIGNIFICANCE Our findings suggest that, despite the immunomodulatory effects of helminth infection, baseline co-morbid infection with these parasites had little effect on the clinical progression from latent to active pulmonary TB.
Collapse
|
18
|
Rausch S, Held J, Fischer A, Heimesaat MM, Kühl AA, Bereswill S, Hartmann S. Small intestinal nematode infection of mice is associated with increased enterobacterial loads alongside the intestinal tract. PLoS One 2013; 8:e74026. [PMID: 24040152 PMCID: PMC3769368 DOI: 10.1371/journal.pone.0074026] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/25/2013] [Indexed: 01/04/2023] Open
Abstract
Parasitic nematodes are potent modulators of immune reactivity in mice and men. Intestinal nematodes live in close contact with commensal gut bacteria, provoke biased Th2 immune responses upon infection, and subsequently lead to changes in gut physiology. We hypothesized that murine nematode infection is associated with distinct changes of the intestinal bacterial microbiota composition. We here studied intestinal inflammatory and immune responses in mice following infection with the hookworm Heligmosomoidespolygyrusbakeri and applied cultural and molecular techniques to quantitatively assess intestinal microbiota changes in the ileum, cecum and colon. At day 14 post nematode infection, mice harbored significantly higher numbers of γ-Proteobacteria/Enterobacteriaceae and members of the Bacteroides/Prevotella group in their cecum as compared to uninfected controls. Abundance of Gram-positive species such as Lactobacilli, Clostridia as well as the total bacterial load was not affected by worm infection. The altered microbiota composition was independent of the IL-4/-13 – STAT6 signaling axis, as infected IL-4Rα-/- mice showed a similar increase in enterobacterial loads. In conclusion, infection with an enteric nematode is accompanied by distinct intestinal microbiota changes towards higher abundance of gram-negative commensal species at the small intestinal site of infection (and inflammation), but also in the parasite-free large intestinal tract. Further studies should unravel the impact of nematode-induced microbiota changes in inflammatory bowel disease to allow for a better understanding of how theses parasites interfere with intestinal inflammation and bacterial communities in men.
Collapse
MESH Headings
- Animals
- Bacterial Load
- Cytokines/biosynthesis
- Enterobacteriaceae/classification
- Enterobacteriaceae/genetics
- Enterobacteriaceae/growth & development
- Female
- Interleukin-4 Receptor alpha Subunit/genetics
- Interleukin-4 Receptor alpha Subunit/metabolism
- Intestinal Diseases, Parasitic/immunology
- Intestinal Diseases, Parasitic/microbiology
- Intestinal Mucosa/microbiology
- Intestinal Mucosa/parasitology
- Intestinal Mucosa/pathology
- Intestine, Small/immunology
- Intestine, Small/microbiology
- Intestine, Small/parasitology
- Intestine, Small/pathology
- Mice
- Mice, Knockout
- Microbiota
- Nematode Infections/immunology
- Nematode Infections/microbiology
- Nematode Infections/parasitology
- RNA, Bacterial
- RNA, Ribosomal, 16S
- Signal Transduction
Collapse
Affiliation(s)
- Sebastian Rausch
- Institute of Immunology, Department of Veterinary Medicine, Freie Universität, Berlin, Germany
- * E-mail:
| | - Josephin Held
- Department of Neuropathology, Charité - University Medicine Berlin, Berlin, Germany
| | - André Fischer
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Markus M. Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Anja A. Kühl
- Department of Internal Medicine, Rheumatology and Clinical Immunology/Research Center ImmunoSciences (RCIS), Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Susanne Hartmann
- Institute of Immunology, Department of Veterinary Medicine, Freie Universität, Berlin, Germany
| |
Collapse
|
19
|
Friberg IM, Little S, Ralli C, Lowe A, Hall A, Jackson JA, Bradley JE. Macroparasites at peripheral sites of infection are major and dynamic modifiers of systemic antimicrobial pattern recognition responses. Mol Ecol 2013; 22:2810-26. [PMID: 23379442 DOI: 10.1111/mec.12212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/06/2012] [Accepted: 12/11/2012] [Indexed: 12/20/2022]
Abstract
Immune defences and the maintenance of immunological homeostasis in the face of pathogenic and commensal microbial exposures are channelled by innate antimicrobial pattern recognition receptors (PRRs) such as toll-like receptors (TLRs). Whilst PRR-mediated response programmes are the result of long-term host-pathogen or host-commensal co-evolutionary dynamics involving microbes, an additional possibility is that macroparasitic co-infections may be a significant modifier of such interactions. We demonstrate experimentally that macroparasites (the model gastrointestinal nematode, Heligmosomoides) at peripheral sites of infection cause substantial alteration of the expression and function of TLRs at a systemic level (in cultured splenocytes), predominantly up-regulating TLR2, TLR4 and TLR9-mediated cytokine responses at times of high standing worm burdens. We consistently observed such effects in BALB/c and C57BL/6 mice under single-pulse and trickle exposures to Heligmosomoides larvae and in SWR and CBA mice under single-pulse exposures. A complementary long-term survey of TLR2-mediated tumour necrosis factor-alpha responses in wild wood mice (Apodemus sylvaticus) was consistent with substantial effects of macroparasites under some environmental conditions. A general pattern, though, was for the associations of macroparasites with TLR function to be temporally dynamic and context-dependent: varying with different conditions of infection exposure in the field and laboratory and with host genetic strain in the laboratory. These results are compelling evidence that macroparasites are a major and dynamic modifier of systemic innate antimicrobial responsiveness in naturally occurring mammals and thus likely to be an important influence on the interaction between microbial exposures and the immune system.
Collapse
Affiliation(s)
- I M Friberg
- School of Biology, University of Nottingham, Nottingham, UK
| | | | | | | | | | | | | |
Collapse
|
20
|
Whelan RAK, Hartmann S, Rausch S. Nematode modulation of inflammatory bowel disease. PROTOPLASMA 2012; 249:871-886. [PMID: 22086188 PMCID: PMC3459088 DOI: 10.1007/s00709-011-0342-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 10/11/2011] [Indexed: 05/31/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic disease arising due to a culmination of genetic, environmental, and lifestyle-associated factors and resulting in an excessive pro-inflammatory response to bacterial populations in the gastrointestinal tract. The prevalence of IBD in developing nations is relatively low, and it has been proposed that this is directly correlated with a high incidence of helminth infections in these areas. Gastrointestinal nematodes are the most prevalent parasitic worms, and they efficiently modulate the immune system of their hosts in order to establish chronic infections. Thus, they may be capable of suppressing unrelated inflammation in disorders such as IBD. This review describes how nematodes, or their products, suppress innate and adaptive pro-inflammatory immune responses and how the mechanisms involved in the induction of anti-nematode responses regulate colitis in experimental models and clinical trials with IBD patients. We also discuss how refinement of nematode-derived therapies should ultimately result in the development of potent new therapeutics of clinical inflammatory disorders.
Collapse
Affiliation(s)
- Rose A. K. Whelan
- Department of Molecular Parasitology, Humboldt University of Berlin, 10115 Berlin, Germany
| | - Susanne Hartmann
- Department of Molecular Parasitology, Humboldt University of Berlin, 10115 Berlin, Germany
| | - Sebastian Rausch
- Department of Molecular Parasitology, Humboldt University of Berlin, 10115 Berlin, Germany
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Tuberculosis (TB) continues to be a significant problem and a major cause of morbidity and mortality in the developing world despite decades of intensive efforts to combat the disease. The poverty in these endemic areas is associated with an increased incidence of tropical helminthic infections. The purpose of this review is to bring to the fore, the urgent need to unravel the potential consequences of helminth coinfection to tuberculosis disease pathogenesis and transmission. RECENT FINDINGS There is now strong experimental evidence that helminth-induced T helper (Th)2 and T regulatory (Treg) responses impinge on host resistance against Mycobacterium tuberculosis (Mtb) infection. Several studies show that Th1 response is reduced in helminth coinfected hosts. Emerging studies also indicate that helminth-induced alternatively activated macrophages contribute to enhanced susceptibility to TB. Despite studies showing an association between helminthes and diminished Th1 immunity, maternal antihelminthic treatment had no effect on an infant's response to bacille Calmette-Guerin vaccination. SUMMARY Whether helminthes affect tuberculosis (TB) disease is still an open question and clinical trials are warranted to determine at the population level whether helminthes enhance TB incidence and transmission and diminish the protective immune response to vaccines. Consequently, mass deworming of infected individuals could contribute toward overall improvement of global public health.
Collapse
|
22
|
Korpe PS, Petri WA. Environmental enteropathy: critical implications of a poorly understood condition. Trends Mol Med 2012; 18:328-36. [PMID: 22633998 DOI: 10.1016/j.molmed.2012.04.007] [Citation(s) in RCA: 332] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 04/12/2012] [Accepted: 04/20/2012] [Indexed: 12/13/2022]
Abstract
Environmental enteropathy (also called tropical enteropathy) is a subclinical condition caused by constant fecal-oral contamination and resulting in blunting of intestinal villi and intestinal inflammation. Although these histological changes were discovered decades ago, the clinical impact of environmental enteropathy is just starting to be recognized. The failure of nutritional interventions and oral vaccines in the developing world may be attributed to environmental enteropathy, as the intestinal absorptive and immunologic functions are significantly deranged. Here we review the existing literature and examine potential mechanisms of pathogenesis for this poorly understood condition.
Collapse
Affiliation(s)
- Poonum S Korpe
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908-1337, USA
| | | |
Collapse
|
23
|
Elliott DE, Weinstock JV. Helminth-host immunological interactions: prevention and control of immune-mediated diseases. Ann N Y Acad Sci 2012; 1247:83-96. [PMID: 22239614 DOI: 10.1111/j.1749-6632.2011.06292.x] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Exposure to commensal and pathogenic organisms strongly influences our immune system. Exposure to helminths was frequent before humans constructed their current highly hygienic environment. Today, in highly industrialized countries, contact between humans and helminths is rare. Congruent with the decline in helminth infections is an increase in the prevalence of autoimmune and inflammatory disease. It is possible that exclusion of helminths from the environment has permitted the emergence of immune-mediated disease. We review the protective effects of helminths on expression of inflammatory bowel disease, multiple sclerosis, and animal models of these and other inflammatory diseases. We also review the immune pathways altered by helminths that may afford protection from these illnesses. Helminth exposure tends to inhibit IFN-γ and IL-17 production, promote IL-4, IL-10, and TGF-β release, induce CD4(+) T cell Foxp3 expression, and generate regulatory macrophages, dendritic cells, and B cells. Helminths enable protective pathways that may vary by specific species and disease model. Helminths or their products likely have therapeutic potential to control or prevent immune-mediated illness.
Collapse
Affiliation(s)
- David E Elliott
- Division of Gastroenterology, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
24
|
Sun S, Wang X, Wu X, Zhao Y, Wang F, Liu X, Song Y, Wu Z, Liu M. Toll-like receptor activation by helminths or helminth products to alleviate inflammatory bowel disease. Parasit Vectors 2011; 4:186. [PMID: 21943110 PMCID: PMC3199248 DOI: 10.1186/1756-3305-4-186] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Accepted: 09/27/2011] [Indexed: 12/11/2022] Open
Abstract
Helminth infection may modulate the expression of Toll like receptors (TLR) in dendritic cells (DCs) and modify the responsiveness of DCs to TLR ligands. This may regulate aberrant intestinal inflammation in humans with helminthes and may thus help alleviate inflammation associated with human inflammatory bowel disease (IBD). Epidemiological and experimental data provide further evidence that reducing helminth infections increases the incidence rate of such autoimmune diseases. Fine control of inflammation in the TLR pathway is highly desirable for effective host defense. Thus, the use of antagonists of TLR-signaling and agonists of their negative regulators from helminths or helminth products should be considered for the treatment of IBD.
Collapse
Affiliation(s)
- ShuMin Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Zoonosis Research Centre of State Key Laboratory for Molecular Virology and Genetic Engineering, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Changchun 130062, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Walk ST, Blum AM, Ewing SAS, Weinstock JV, Young VB. Alteration of the murine gut microbiota during infection with the parasitic helminth Heligmosomoides polygyrus. Inflamm Bowel Dis 2010; 16:1841-9. [PMID: 20848461 PMCID: PMC2959136 DOI: 10.1002/ibd.21299] [Citation(s) in RCA: 246] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND In a murine model of inflammatory bowel disease (IBD), treatment of colitis in IL-10 gene-deficient mice with the parasitic helminth Heligmosomoides polygyrus ameliorates colonic inflammation. The cellular and molecular mechanisms driving this therapeutic host response are being studied vigorously. One proposed mechanism is that H. polygyrus infection favors the outgrowth or suppression of certain bacteria, which in turn help modulate host immunity. METHODS To quantify the effect of H. polygyrus infection on the composition of the gastrointestinal (GI) tract microbiota, we conducted two independent microbial ecology analyses of C57BL/6 mice. We obtained and analyzed 3,353 bacterial 16S rRNA encoding gene sequences from the ileum and cecum of infected and uninfected mice as well as incective H. polygyrus larvae at the outset of the second experiment and adult worms taken directly from the mouse duodenum at the end of the second experiment. RESULTS We found that a significant shift in the abundance and relative distribution of bacterial species in the ileum of mice is associated with H. polygyrus infection. Members of the bacterial family Lactobacillaceae significantly increased in abundance in the ileum of infected mice reproducibly in two independent experiments despite having different microbiotas present at the outset of each experiment. CONCLUSIONS These data support the concept that helminth infection shifts the composition of intestinal bacteria. The clinical consequences of these shifts in intestinal flora are yet to be explored.
Collapse
Affiliation(s)
- Seth T. Walk
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Arthur M. Blum
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tufts New England Medical Center, Boston MA 02111, USA
| | - Sarah Ang-Sheng Ewing
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Joel V. Weinstock
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tufts New England Medical Center, Boston MA 02111, USA
| | - Vincent B. Young
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Health System, Ann Arbor, MI 48109, USA, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
26
|
Macroparasites, innate immunity and immunoregulation: developing natural models. Trends Parasitol 2010; 26:540-9. [PMID: 20634138 DOI: 10.1016/j.pt.2010.06.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2009] [Revised: 06/03/2010] [Accepted: 06/07/2010] [Indexed: 12/25/2022]
Abstract
Innate immune receptors carry out surveillance for infection threats and are a proximal controller of the threshold and intensity at which inflammatory responses occur. As such, they are a natural focus for understanding how inflammatory immune reactivity is regulated. This review highlights how little data there are relating to the effect of macroparasites on systemic innate receptor responses. The idea is developed that studies on innate immune function in wild animals exposed to a natural profile of infections, including macroparasites, might be a valuable model in which to test hypotheses about the ultimate cause of aberrant inflammation in modern human populations.
Collapse
|
27
|
Helminthic therapy: using worms to treat immune-mediated disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 666:157-66. [PMID: 20054982 DOI: 10.1007/978-1-4419-1601-3_12] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
There is an epidemic of immune-mediated disease in highly-developed industrialized countries. Such diseases, like inflammatory bowel disease, multiple sclerosis and asthma increase in prevalence as populations adopt modern hygienic practices. These practices prevent exposure to parasitic worms (helminths). Epidemiologic studies suggest that people who carry helminths have less immune-mediated disease. Mice colonized with helminths are protected from disease in models of colitis, encephalitis, Type 1 diabetes and asthma. Clinical trials show that exposure to helminths reduce disease activity in patients with ulcerative colitis or Crohn's disease. This chapter reviews some of the work showing that colonization with helminths alters immune responses, against dysregulated inflammation. These helminth-host immune interactions have potentially important implications for the treatment of immune-mediated diseases.
Collapse
|
28
|
Signal-transducing mechanisms of ketamine-caused inhibition of interleukin-1β gene expression in lipopolysaccharide-stimulated murine macrophage-like Raw 264.7 cells. Toxicol Appl Pharmacol 2009; 240:15-25. [DOI: 10.1016/j.taap.2009.06.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 06/07/2009] [Accepted: 06/15/2009] [Indexed: 01/08/2023]
|
29
|
Ince MN, Elliott DE, Setiawan T, Metwali A, Blum A, Chen HL, Urban JF, Flavell RA, Weinstock JV. Role of T cell TGF-beta signaling in intestinal cytokine responses and helminthic immune modulation. Eur J Immunol 2009; 39:1870-8. [PMID: 19544487 PMCID: PMC2882993 DOI: 10.1002/eji.200838956] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Colonization with helminthic parasites induces mucosal regulatory cytokines, like IL-10 or TGF-beta, that are important in suppressing colitis. Helminths induce mucosal T cell IL-10 secretion and regulate lamina propria mononuclear cell (LPMC) Th1 cytokine generation in an IL-10-dependent manner in WT mice. Helminths also stimulate mucosal TGF-beta release. As TGF-beta exerts major regulatory effects on T lymphocytes, we investigated the role of T lymphocyte TGF-beta signaling in helminthic modulation of intestinal immunity. T cell TGF-beta signaling is interrupted in TGF-beta receptor II dominant negative (TGF-betaRII DN) mice by T-cell-specific over-expression of a TGF-betaRII DN. We studied LPMC responses in WT and TGF-betaRII DN mice that were uninfected or colonized with the nematode, Heligmosomoides polygyrus. Our results indicate an essential role of T cell TGF-beta signaling in limiting mucosal Th1 and Th2 responses. Furthermore, we demonstrate that helminthic induction of intestinal T cell IL-10 secretion requires intact T cell TGF-beta-signaling pathway. Helminths fail to curtail robust, dysregulated intestinal Th1 cytokine production and chronic colitis in TGF-betaRII DN mice. Thus, T cell TGF-beta signaling is essential for helminthic stimulation of mucosal IL-10 production, helminthic modulation of intestinal IFN-gamma generation and H. polygyrus-mediated suppression of chronic colitis.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Colitis/immunology
- Colitis/metabolism
- Colitis/parasitology
- Cytokines/metabolism
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Host-Parasite Interactions
- Interferon-gamma/metabolism
- Interleukin-10/metabolism
- Intestinal Diseases, Parasitic/immunology
- Intestinal Diseases, Parasitic/metabolism
- Intestinal Diseases, Parasitic/parasitology
- Intestine, Small/cytology
- Intestine, Small/metabolism
- Intestine, Small/parasitology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutant Proteins/genetics
- Mutant Proteins/metabolism
- Nematospiroides dubius/physiology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Signal Transduction/genetics
- Signal Transduction/physiology
- Strongylida Infections/immunology
- Strongylida Infections/metabolism
- Strongylida Infections/parasitology
- T-Lymphocytes/cytology
- T-Lymphocytes/metabolism
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- M Nedim Ince
- Department of Internal Medicine, University of Iowa, Iowa City, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
There is limited data on the human mucosal immune response to geohelminths, but extensive data from experimental animals. Geohelminth infections may modulate mucosal immunity with effects on parasite expulsion or persistence and mucosal inflammation. Geohelminths are considered to have important effects on immunity to mucosal vaccines, infectious disease susceptibility, and anti-inflammatory effects in inflammatory bowel disease and asthma. This review will discuss the findings of studies of human immunity to geohelminths and their potential effects on non-parasite mucosal immune responses. Such effects are likely to be of public health importance in middle- and low-income countries where these parasites are endemic. There is a need for human studies on the effects of geohelminth infections on mucosal immunity and the potential for anthelmintic treatment to modify these effects. Such studies are likely to provide important insights into the regulation of mucosal immunity and inflammation, and the development of more effective mucosal vaccines.
Collapse
|
31
|
Helmby H. Gastrointestinal nematode infection exacerbates malaria-induced liver pathology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:5663-71. [PMID: 19380813 PMCID: PMC2796717 DOI: 10.4049/jimmunol.0803790] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mixed parasite infections are common in many parts of the world, but little is known of the effects of concomitant parasite infections on the immune response or severity of clinical disease. We have used the nonlethal malaria infection model of Plasmodium chabaudi AS in combination with the gastrointestinal nematode Heligmosomoides bakeri polygyrus to investigate the impact of nematode infections on malarial morbidity and antimalarial immunity. The data demonstrate that wild-type C57BL/6 mice coinfected with both parasites simultaneously exhibit a striking increase in mortality, while mice deficient in IFN-gamma or IL-23 survive coinfection. The increase in mortality in wild-type mice was associated with severe liver pathology characterized by extensive coagulative necrosis and an increase in hepatic IFN-gamma, IL-17, and IL-22 mRNA expression. This is the first demonstration of increased malaria-associated pathology associated with a switch toward a proinflammatory environment, involving not only IFN-gamma but also the IL-17/IL-23 axis, as a result of coinfection with a gastrointestinal helminth.
Collapse
Affiliation(s)
- Helena Helmby
- Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| |
Collapse
|
32
|
Jackson JA, Friberg IM, Little S, Bradley JE. Review series on helminths, immune modulation and the hygiene hypothesis: immunity against helminths and immunological phenomena in modern human populations: coevolutionary legacies? Immunology 2009; 126:18-27. [PMID: 19120495 DOI: 10.1111/j.1365-2567.2008.03010.x] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Although the molecules and cells involved in triggering immune responses against parasitic worms (helminths) remain enigmatic, research has continued to implicate expansions of T-helper type 2 (Th2) cells and regulatory T-helper (T(reg)) cells as a characteristic response to these organisms. An intimate association has also emerged between Th2 responses and wound-healing functions. As helminth infections in humans are associated with a strong Th2/T(reg) immunoregulatory footprint (often termed a 'modified Th2' response), plausible links have been made to increased susceptibility to microbial pathogens in helminth-infected populations in the tropics and to the breakdowns in immunological control (allergy and autoimmunity) that are increasing in frequency in helminth-free developed countries. Removal of helminths and their anti-inflammatory influence may also have hazards for populations exposed to infectious agents, such as malaria and influenza, whose worst effects are mediated by excessive inflammatory reactions. The patterns seen in the control of helminth immunity are discussed from an evolutionary perspective. Whilst an inability to correctly regulate the immune system in the absence of helminth infection might seem highly counter-adaptive, the very ancient and pervasive relationship between vertebrates and helminths supports a view that immunological control networks have been selected to function within the context of a modified Th2 environment. The absence of immunoregulatory stimuli from helminths may therefore uncover maladaptations that were not previously exposed to selection.
Collapse
Affiliation(s)
- Joseph A Jackson
- School of Biology, The University of Nottingham, University Park, Nottingham, UK
| | | | | | | |
Collapse
|
33
|
Abstract
Helminths are parasitic animals that have evolved over 100,000,000 years to live in the intestinal track or other locations of their hosts. Colonization of humans with these organisms was nearly universal until the early 20th century. More than 1,000,000,000 people in less developed countries carry helminths even today. Helminths must quell their host's immune system to successfully colonize. It is likely that helminths sense hostile changes in the local host environment and take action to control such responses. Inflammatory bowel disease (IBD) probably results from an inappropriately vigorous immune response to contents of the intestinal lumen. Environmental factors strongly affect the risk for IBD. People living in less developed countries are protected from IBD. The "IBD hygiene hypothesis" states that raising children in extremely hygienic environments negatively affects immune development, which predisposes them to immunological diseases like IBD later in life. Modern day absence of exposure to intestinal helminths appears to be an important environmental factor contributing to development of these illnesses. Helminths interact with both host innate and adoptive immunity to stimulate immune regulatory circuitry and to dampen effector pathways that drive aberrant inflammation. The first prototype worm therapies directed against immunological diseases are now under study in the United States and various countries around the world. Additional studies are in the advanced planning stage.
Collapse
Affiliation(s)
- Joel V Weinstock
- Tufts New England Medical Center, Boston, Massachusetts 02111, USA.
| | | |
Collapse
|
34
|
Herbert DR, Orekov T, Perkins C, Finkelman FD. IL-10 and TGF-beta redundantly protect against severe liver injury and mortality during acute schistosomiasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7214-20. [PMID: 18981143 PMCID: PMC2921214 DOI: 10.4049/jimmunol.181.10.7214] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The cytokines IL-10 and TGF-beta regulate immunity and inflammation. IL-10 is known to suppress the extent of hepatic damage caused by parasite ova during natural infection with Schistosoma mansoni, but the role of TGF-beta is less clear. Cytokine blockade studies in mice revealed that anti-IL-10R mAb treatment during acute infection modestly increased cytokine production and liver damage, whereas selective anti-TGF-beta mAb treatment had marginal effects. In contrast, mice administered both mAbs developed severe hepatic inflammation, with enlarged, necrotic liver granulomas, cachexia, and >80% mortality by 8 wk postinfection, despite increased numbers of CD4(+)CD25(+)Foxp3(+) T regulatory cells. Blocking both IL-10 and TGF-beta at the onset of egg production also significantly increased IL-4, IL-6, TNF, IFN-gamma, and IL-17 production and markedly increased hepatic, peritoneal, and splenic neutrophilia. In contrast, coadministration of anti-IL-10R and TGF-beta mAbs had little effect upon parasite ova-induced intestinal pathology or development of alternatively activated macrophages, which are required to suppress intestinal pathology. This suggests that inflammation is controlled during acute S. mansoni infection by two distinct, organ-specific mechanisms: TGF-beta and IL-10 redundantly suppress hepatic inflammation while intestinal inflammation is regulated by alternatively activated macrophages.
Collapse
Affiliation(s)
- De'Broski R Herbert
- Research Service (151), Cincinnati Veterans Administration Medical Center, Cincinnati, OH 45220, USA.
| | | | | | | |
Collapse
|
35
|
Motomura Y, Wang H, Deng Y, El-Sharkawy RT, Verdu EF, Khan WI. Helminth antigen-based strategy to ameliorate inflammation in an experimental model of colitis. Clin Exp Immunol 2008; 155:88-95. [PMID: 19016806 DOI: 10.1111/j.1365-2249.2008.03805.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is the most common and serious chronic inflammatory condition of the gut. Among the distinct T helper (Th) cell subsets, a Th1 type response is associated predominantly with Crohn's disease (CD) while helminth infections generate a strong Th2 type response. IBD is most prevalent in developed countries but rare in countries where infections with helminths are common. Thus, it has been hypothesized that infection with helminth infection influence the development of CD and recent clinical and experimental studies suggest strongly a beneficial role of helminth infection in IBD. In the present study we examined the effects of rectal submucosal administration of helminth antigens on subsequent experimental colitis. Mice were treated with Trichinella spiralis antigens prior to the induction of dinitrobenzenesulphonic acid (DNBS)-induced colitis and were killed 3 days post-DNBS to assess colonic damage macroscopically, histologically and by myeloperoxidase (MPO) activity, inducible nitric oxide synthase (iNOS) and cytokine levels. Previous treatment with T. spiralis antigens reduced the severity of colitis significantly, as assessed macroscopically and histologically, and reduced the mortality rate. This benefit was correlated with a down-regulation of MPO activity, interleukin (IL)-1beta production and iNOS expression and an up-regulation of IL-13 and transforming growth factor-beta production in colon. These results clearly show a beneficial role of local treatment with helminth antigens for experimental colitis and prompt consideration of helminth antigen-based therapy for IBD instead of infection with live parasites.
Collapse
Affiliation(s)
- Y Motomura
- Intestinal Diseases Research Program, Department of Pathology, McMaster University, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
36
|
Wang LJ, Cao Y, Shi HN. Helminth infections and intestinal inflammation. World J Gastroenterol 2008; 14:5125-32. [PMID: 18777588 PMCID: PMC2744001 DOI: 10.3748/wjg.14.5125] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 08/14/2008] [Accepted: 08/21/2008] [Indexed: 02/06/2023] Open
Abstract
Evidence from epidemiological studies indicates an inverse correlation between the incidence of certain immune-mediated diseases, including inflammatory bowel diseases (IBD), and exposure to helminths. Helminth parasites are the classic inducers of Th2 responses. The Th2-polarized T cell response driven by helminth infection has been linked to the attenuation of some damaging Th1 driven inflammatory responses, preventing some Th1-mediated autoimmune diseases in the host, including experimentally induced colitis. Helminth parasites (the porcine whipworm, Trichuris suis) have been tested for treating IBD patients, resulting in clinical amelioration of the disease. As a result, there is a great deal of interest in the research community in exploring the therapeutic use of helminth parasites for the control of immune-mediated diseases, including IBD. However, recent studies have provided evidence indicating the exacerbating effects of helminths on bacterial as well as non-infectious colitis in animal models. Therefore, a better understanding of mechanisms by which helminths modulate host immune responses in the gut may reveal novel, more effective and safer approaches to helminth-based therapy of IBD.
Collapse
|
37
|
Levin A, Shibolet O. Toll-like receptors in inflammatory bowel disease-stepping into uncharted territory. World J Gastroenterol 2008; 14:5149-53. [PMID: 18777591 PMCID: PMC2744004 DOI: 10.3748/wjg.14.5149] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ulcerative colitis and Crohn’s disease are chronic relapsing-remitting inflammatory processes of the intestinal tract. The etiology of these diseases is currently unknown. However, inflammation is hypothesized to result from inappropriate activation of mucosal immunity by luminal antigens in genetically susceptible individuals. Toll-like receptors (TLRs) are a family of transmembrane proteins that act as microbial pattern recognition receptors. They are crucial initiators of innate immune responses. The role of TLRs in the pathogenesis of inflammatory bowel disease (IBD) has not been fully elucidated. In this review, we aim to analyze the available data connecting individual TLRs to intestinal inflammation and IBD.
Collapse
|
38
|
Abstract
There has been an alarming increase in the incidence of autoimmune and allergic diseases in Western countries in the past few decades. However, in countries endemic for parasitic helminth infections, such diseases remain relatively rare. Hence, it has been hypothesised that helminths may protect against the development of autoimmunity and allergy. This article reviews the evidence supporting this idea with respect to helminths of the phylum Nematoda (nematodes), considering data from human studies and animal models of inflammatory disease. The nature and mode of action of nematode-derived molecules with immunomodulatory properties are considered, and their therapeutic efficacy in models of autoimmunity and allergy described. The recent and future use of nematodes and their products in treating human disease are also discussed.
Collapse
Affiliation(s)
- William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | | |
Collapse
|
39
|
Abstract
Carefully chosen immunological measurements, informed by recent advances in our understanding of the diversity and control of immune mechanisms, can add great interpretative value to ecological studies of infection. This is especially so for co-infection studies, where interactions between species are often mediated via the host's immune response. Here we consider how immunological measurements can strengthen inference in different types of co-infection analysis. In particular, we identify how measuring immune response variables in field studies can help reveal inter-species interactions otherwise obscured by confounding processes operating on count or prevalence data. Furthermore, we suggest that, due to the difficulty of quantifying microbial pathogen communities in field studies, innate responses against broad pathogen types (mediated by pattern response receptors) may be useful quantitative markers of exposure to bacteria and viruses. An ultimate goal of ecological co-infection studies may also be to understand how dynamics within host-parasite assemblages emerge from trade-offs involving different arms of the immune system. We reflect on the phenotypic measures that might best represent levels of responsiveness and bias in immune function. These include mediators associated with different T-helper cell subsets and innate responses controlled by pattern response receptors, such as the Toll-like receptors (TLRs).
Collapse
|
40
|
Cario E. Therapeutic impact of toll-like receptors on inflammatory bowel diseases: a multiple-edged sword. Inflamm Bowel Dis 2008; 14:411-21. [PMID: 17941072 DOI: 10.1002/ibd.20294] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Recent studies have begun to define the mechanisms through which Toll-like receptors (TLRs) regulate intestinal homeostasis in health and disease. Current therapies for inflammatory bowel diseases (IBDs) mostly aim at interrupting the inflammatory cascade through agents that regulate TH1 or TH2 cytokine responses. As recognition grows for TLR dysfunction to play a role in IBD pathogenesis, TLRs could provide another valid interventional target for novel therapy development. However, seemingly contradictory results from studying different murine models of colitis have so far confounded whether therapeutically useful modulation of TLRs is best accomplished by activating, inhibiting, or rather a combination of both at different stages of mucosal disease. This review evaluates potential strategies as well as their rationale and future prospects.
Collapse
Affiliation(s)
- Elke Cario
- Division of Gastroenterology & Hepatology, University Hospital of Essen, University of Duisburg-Essen, Institutsgruppe I, Virchowstrasse 171, Essen, Germany.
| |
Collapse
|
41
|
Zaccone P, Burton OT, Cooke A. Interplay of parasite-driven immune responses and autoimmunity. Trends Parasitol 2008; 24:35-42. [PMID: 18055264 DOI: 10.1016/j.pt.2007.10.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 09/29/2007] [Accepted: 10/01/2007] [Indexed: 12/24/2022]
Abstract
As more facts emerge regarding the ways in which parasite-derived molecules modulate the host immune response, it is possible to envisage how a lack of infection by agents that once infected humans commonly might contribute to the rise in autoimmune disease. Through effects on cells of both the innate and adaptive arms of the immune response, parasites can orchestrate a range of outcomes that are beneficial not only to parasites, in terms of facilitating their life cycles, but also to their host, in limiting pathology.
Collapse
Affiliation(s)
- Paola Zaccone
- Department of Pathology, Tennis Court Road, Cambridge, CB2 1QP, UK
| | | | | |
Collapse
|
42
|
Urban JF, Steenhard NR, Solano-Aguilar GI, Dawson HD, Iweala OI, Nagler CR, Noland GS, Kumar N, Anthony RM, Shea-Donohue T, Weinstock J, Gause WC. Infection with parasitic nematodes confounds vaccination efficacy. Vet Parasitol 2007; 148:14-20. [PMID: 17587500 PMCID: PMC1959410 DOI: 10.1016/j.vetpar.2007.05.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
T helper (Th) cells produce signature cytokine patterns, induced largely by intracellular versus extracellular pathogens that provide the cellular and molecular basis for counter regulatory expression of protective immunity during concurrent infections. The production of IL-12 and IFN-gamma, for example, resulting from exposure to many bacterial, viral, and protozoan pathogens is responsible for Th1-derived protective responses that also can inhibit development of Th2-cells expressing IL-4-dependent immunity to extracellular helminth parasites and vice versa. In a similar manner, concurrent helminth infection alters optimal vaccine-induced responses in humans and livestock; however, the consequences of this condition have not been adequately studied especially in the context of a challenge infection following vaccination. Demands for new and effective vaccines to control chronic and emerging diseases, and the need for rapid deployment of vaccines for bio security concerns requires a systematic evaluation of confounding factors that limit vaccine efficacy. One common albeit overlooked confounder is the presence of gastrointestinal nematode parasites in populations of humans and livestock targeted for vaccination. This is particularly important in areas of the world were helminth infections are prevalent, but the interplay between parasites and emerging diseases that can be transmitted worldwide make this a global issue. In addition, it is not clear if the epidemic in allergic disease in industrialized countries substitutes for geohelminth infection to interfere with effective vaccination regimens. This presentation will focus on recent vaccination studies in mice experimentally infected with Heligmosomoides polygyrus to model the condition of gastrointestinal parasite infestation in mammalian populations targeted for vaccination. In addition, a large animal vaccination and challenge model against Mycoplasma hyopneumonia in swine exposed to Ascaris suum will provide a specific example of the need for further work in this area, and for controlled field studies to assess the impact of other similar scenarios.
Collapse
Affiliation(s)
- Joseph F Urban
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705-2350, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Molecular and immunologic mechanisms underlying inflammation in inflammatory bowel disease (IBD) are largely unknown. Recent studies have helped better characterize genetic and environmental factors associated with colitis. Discoveries of genetic variants have confirmed that IBD is a bacteria and cytokine-driven pathologic immune response. Data have demonstrated that certain T cell subsets are important in executing the inflammatory cascade. Insufficient regulatory cell activity or modulatory cytokine production results in unrestrained inflammation. Biologic agents that block inflammatory cytokines (anti-TNFalpha antibodies) have been used successfully to treat IBD. Recent advances have also identified mucosal regulatory pathways.
Collapse
Affiliation(s)
- M Nedim Ince
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, 4611 JCP, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | | |
Collapse
|
44
|
Setiawan T, Metwali A, Blum AM, Ince MN, Urban JF, Elliott DE, Weinstock JV. Heligmosomoides polygyrus promotes regulatory T-cell cytokine production in the murine normal distal intestine. Infect Immun 2007; 75:4655-63. [PMID: 17606601 PMCID: PMC1951154 DOI: 10.1128/iai.00358-07] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Helminths down-regulate inflammation and may prevent development of several autoimmune illnesses, such as inflammatory bowel disease. We determined if exposure to the duodenal helminth Heligmosomoides polygyrus establishes cytokine pathways in the distal intestine that may protect from intestinal inflammation. Mice received 200 H. polygyrus larvae and were studied 2 weeks later. Lamina propria mononuclear cells (LPMC) were isolated from the terminal ileum for analysis and in vitro experiments. Mice with H. polygyrus were resistant to trinitrobenzenesulfonic acid (TNBS)-induced colitis, a Th1 cytokine-dependent inflammation. Heligmosomoides polygyrus did not change the normal microscopic appearance of the terminal ileum and colon and minimally affected LPMC composition. However, colonization altered LPMC cytokine profiles, blocking gamma interferon (IFN-gamma) and interleukin 12 (IL-12) p40 release but promoting IL-4, IL-5, IL-13, and IL-10 secretion. IL-10 blockade in vitro with anti-IL-10 receptor (IL-10R) monoclonal antibody restored LPMC IFN-gamma and IL-12 p40 secretion. IL-10 blockade in vivo worsened TNBS colitis in H. polygyrus-colonized mice. Lamina propria CD4(+) T cells isolated from colonized mice inhibited IFN-gamma production by splenic T cells from worm-free mice. This inhibition did not require cell contact and was dependent on IL-10. Heligmosomoides polygyrus colonization inhibits Th1 and promotes Th2 and regulatory cytokine production in distant intestinal regions without changing histology or LPMC composition. IL-10 is particularly important for limiting the Th1 response. The T-cell origin of these cytokines demonstrates mucosal regulatory T-cell induction.
Collapse
Affiliation(s)
- Tommy Setiawan
- Division of Gastroenterology (4611 JCP), University of Iowa Hospital and Clinics, 200 Hawkins Drive, Iowa City, IA 52242-1009, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Nematode parasites cause significant infections in both humans and animals. They are complex, multicellular organisms that present unique challenges for the host, in particular with respect to the recognition of their unusual surface structures by the innate defence system. The innate immune system is now recognized to be a critical component in the development of an adaptive effector response as well as a driver of vaccine-induced immunity. This paper will give an overview of current research on the innate barriers and immune mechanisms, cells, and receptors involved in the innate host response to nematode parasites. It will also review the 'nematode-associated molecular patterns' that may be specifically recognized by the host, in addition to other signals, such as nervous stimulation and tissue damage, that may alert the innate system to parasite invasion.
Collapse
Affiliation(s)
- M J de Veer
- Animal Biotechnology Research Laboratories, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | | | | |
Collapse
|
46
|
Coombes JL, Maloy KJ. Control of intestinal homeostasis by regulatory T cells and dendritic cells. Semin Immunol 2007; 19:116-26. [PMID: 17320411 DOI: 10.1016/j.smim.2007.01.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 01/08/2007] [Indexed: 02/07/2023]
Abstract
Many different pathways contribute to the maintenance of tolerance to harmless antigens in the intestine. When these important pathways are compromised, chronic intestinal inflammation can develop. In particular, naturally occurring CD4+CD25+ regulatory T cells have been shown to play an important role in the prevention and cure of colitis in animal models of intestinal inflammation. These regulatory T cell responses may be influenced by the local environment in the intestine. For example, functionally specialised populations of dendritic cells exist in the intestine which may favour regulatory type responses. Understanding how these pathways intersect may lead to the development of more specific therapies for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Janine L Coombes
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | |
Collapse
|
47
|
Reddy A, Fried B. The use of Trichuris suis and other helminth therapies to treat Crohn’s disease. Parasitol Res 2007; 100:921-7. [PMID: 17206504 DOI: 10.1007/s00436-006-0416-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Accepted: 11/25/2006] [Indexed: 12/31/2022]
Abstract
Infections with gastrointestinal (GI) nematodes are prevalent worldwide, despite the fact that anti-helminthic medications are regarded as safe, efficient, and widely available globally. In this review, we highlight the potential therapeutic benefits that may be realized through the clinical use of Trichuris suis and other helminths for Crohn's disease (CD). Long-lived helminthic parasites are remarkable in their ability to down-regulate host immunity, protecting themselves from elimination, and also minimize severe pathological host changes. This review summarizes what is known about the underlying mechanisms that may account for the observed patterns in humans treated with helminths for CD. The Th2 arm of the immune system is emphasized as a component of primary importance in the association between the host immune system and GI nematode infections. Although GI nematode infections in humans cause significant morbidity and mortality, the existence and nature of protective mechanisms these helminths may confer remain largely unclear.
Collapse
Affiliation(s)
- Aditya Reddy
- Department of Biology, Lafayette College, Easton, PA 18042, USA.
| | | |
Collapse
|
48
|
Elliott DE, Summers RW, Weinstock JV. Helminths as governors of immune-mediated inflammation. Int J Parasitol 2006; 37:457-64. [PMID: 17313951 DOI: 10.1016/j.ijpara.2006.12.009] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 11/28/2006] [Accepted: 12/06/2006] [Indexed: 12/11/2022]
Abstract
Immune-mediated diseases (e.g. inflammatory bowel disease, asthma, multiple sclerosis and autoimmune diabetes) are increasing in prevalence and emerge as populations adopt meticulously hygienic lifestyles. This change in lifestyles precludes exposure to helminths (parasitic worms). Loss of natural helminth exposure removes a previously universal Th2 and regulatory immune biasing imparted by these organisms. Helminths protect animals from developing immune-mediated diseases (colitis, reactive airway disease, encephalitis and diabetes). Clinical trials show that exposure to helminths can reduce disease activity in patients with ulcerative colitis or Crohn's disease. This paper summarises work by multiple groups demonstrating that colonization with helminths alters immune reactivity and protects against disease from dysregulated inflammation.
Collapse
Affiliation(s)
- David E Elliott
- Department of Internal Medicine, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242-1009, USA.
| | | | | |
Collapse
|
49
|
Iweala OI, Nagler CR. Immune privilege in the gut: the establishment and maintenance of non-responsiveness to dietary antigens and commensal flora. Immunol Rev 2006; 213:82-100. [PMID: 16972898 DOI: 10.1111/j.1600-065x.2006.00431.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Immune privilege in the gut is the result of a complex interplay between the gut microbiome, gut luminal antigens, and the intestinal epithelial barrier. Composed of both physical and immunochemical components, the intestinal barrier secretes immunoregulatory mediators that promote the generation of tolerogenic antigen-presenting cells, phagocytic innate immune cells characterized by 'inflammatory anergy', and regulatory cells of the adaptive immune system. Innate immune cells mediate controlled transepithelial transport of luminal antigens as far as the mesenteric lymph nodes, where the intestinal and peripheral immune systems intersect. This promotes the generation of adaptive regulatory lymphocytes that actively suppress effector cell responses against gut luminal antigens and flora. The net result is the generation of tolerance to dietary antigens and the maintenance of gut homeostasis. Dysregulation of this complex immunoregulatory network leads to diseases such as food allergy and inflammatory bowel disease. Future therapies for these diseases will likely involve the functional restoration of the barrier and regulatory cell functions at the epithelial/luminal interface.
Collapse
Affiliation(s)
- Onyinye I Iweala
- Mucosal Immunology Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-4404, USA
| | | |
Collapse
|
50
|
Affiliation(s)
- Robert W. Summers
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City, IA 52242
| | - David E. Elliott
- Department of Internal Medicine University of Iowa Carver College of Medicine Iowa City, IA 52242
| | - Joel V. Weinstock
- Department of Gastroenterology/ Hepatology Tufts-New England Medical Center Boston, MA 02111
| |
Collapse
|