51
|
Helmby H. Human helminth therapy to treat inflammatory disorders - where do we stand? BMC Immunol 2015; 16:12. [PMID: 25884706 PMCID: PMC4374592 DOI: 10.1186/s12865-015-0074-3] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/05/2015] [Indexed: 12/22/2022] Open
Abstract
Parasitic helminths have evolved together with the mammalian immune system over many millennia and as such they have become remarkably efficient modulators in order to promote their own survival. Their ability to alter and/or suppress immune responses could be beneficial to the host by helping control excessive inflammatory responses and animal models and pre-clinical trials have all suggested a beneficial effect of helminth infections on inflammatory bowel conditions, MS, asthma and atopy. Thus, helminth therapy has been suggested as a possible treatment method for autoimmune and other inflammatory disorders in humans.
Collapse
Affiliation(s)
- Helena Helmby
- Department of Immunology and Infection, Faculty of infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel street, London, WC1E 7HT, UK.
| |
Collapse
|
52
|
Mohammadi R, Hosseini-Safa A, Ehsani Ardakani MJ, Rostami-Nejad M. The relationship between intestinal parasites and some immune-mediated intestinal conditions. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2015; 8:123-31. [PMID: 25926937 PMCID: PMC4403024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/03/2015] [Indexed: 11/16/2022]
Abstract
Over the last decades, the incidence of infestation by minor parasites has decreased in developed countries. Infectious agents can also suppress autoimmune and allergic disorders. Some investigations show that various protozoa and helminthes are connected with the main immune-mediated intestinal conditions including celiac disease (CD), inflammatory bowel diseases (IBD) and irritable bowel syndrome (IBS). Celiac disease is a digestive and autoimmune disorder that can damage the small intestine and characterized by a multitude gastrointestinal (GI) and extra GI symptoms. IBD (including ulcerative colitis and Crohn's disease) is a group of inflammatory conditions of the small intestine and colon. The etiology of IBD is unknown, but it may be related to instability in the intestinal microflora that leading to an immoderate inflammatory response to commensal microbiota. Irritable bowel syndrome (IBS) is a common, long-term condition of the digestive system. Bloating, diarrhoea and/or constipation are nonspecific symptoms of IBS. Various studies have shown that some intestinal parasites can effect on immune system of infected hosts and in some cases, they are able to modify and change the host's immune responses, particularly in autoimmune disorders like celiac disease and IBD. The main objective of this review is to investigate the relationship between intestinal parasites and different inflammatory bowel disorders.
Collapse
Affiliation(s)
- Rasoul Mohammadi
- Department of Medical Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran,
| | - Ahmad Hosseini-Safa
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
53
|
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
|
54
|
Finlay CM, Walsh KP, Mills KHG. Induction of regulatory cells by helminth parasites: exploitation for the treatment of inflammatory diseases. Immunol Rev 2014; 259:206-30. [PMID: 24712468 DOI: 10.1111/imr.12164] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Helminth parasites are highly successful pathogens, chronically infecting a quarter of the world's population, causing significant morbidity but rarely causing death. Protective immunity and expulsion of helminths is mediated by T-helper 2 (Th2) cells, type 2 (M2) macrophages, type 2 innate lymphoid cells, and eosinophils. Failure to mount these type 2 immune responses can result in immunopathology mediated by Th1 or Th17 cells. Helminths have evolved a wide variety of approaches for immune suppression, especially the generation of regulatory T cells and anti-inflammatory cytokines interleukin-10 and transforming growth factor-β. This is a very effective strategy for subverting protective immune responses to prolong their survival in the host but has the bystander effect of modulating immune responses to unrelated antigens. Epidemiological studies in humans have shown that infection with helminth parasites is associated with a low incidence of allergy/asthma and autoimmunity in developing countries. Experimental studies in mice have demonstrated that regulatory immune responses induced by helminth can suppress Th2 and Th1/Th17 responses that mediate allergy and autoimmunity, respectively. This has provided a rational explanation of the 'hygiene hypothesis' and has also led to the exploitation of helminths or their immunomodulatory products in the development of new immunosuppressive therapies for inflammatory diseases in humans.
Collapse
Affiliation(s)
- Conor M Finlay
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
55
|
Gaze S, Bethony JM, Periago MV. Immunology of experimental and natural human hookworm infection. Parasite Immunol 2014; 36:358-66. [PMID: 25337625 DOI: 10.1111/pim.12088] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human hookworm infection is one amongst the most prevalent of the neglected tropical diseases. An informative experimental animal model, that is, one that parallels a human infection, is not available for the study of human hookworm infection. Much of our current understanding of the human immune response during hookworm infection relies on the studies from experimental infection of hookworm-naïve individuals or the natural infections from individuals residing in hookworm-endemic areas. The experimental human infections tend to be acute, dose-controlled infections, often with a low larval inoculum so that they are well tolerated by human volunteers. Natural hookworm infections usually occur in areas where hookworm transmission is constant and infection is chronic. In cases where there has been drug administration in an endemic area, re-infection often occurs quickly even amongst those who were treated. Hence, although many of the characteristics of experimental and natural hookworm infection differ, both models have elements in common: mainly an intense Th2 response with the production of total and specific IgE as well as elevated levels of eosinophilia, IL-5, IL-10 and TNF. While hookworm infection affects millions of individuals worldwide, much of the human immunology of this infection still needs to be studied and understood.
Collapse
|
56
|
Bouchery T, Kyle R, Ronchese F, Le Gros G. The Differentiation of CD4(+) T-Helper Cell Subsets in the Context of Helminth Parasite Infection. Front Immunol 2014; 5:487. [PMID: 25360134 PMCID: PMC4197778 DOI: 10.3389/fimmu.2014.00487] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/22/2014] [Indexed: 12/13/2022] Open
Abstract
Helminths are credited with being the major selective force driving the evolution of the so-called “type 2” immune responses in vertebrate animals, with their size and infection strategies presenting unique challenges to the immune system. Originally, type 2 immune responses were defined by the presence and activities of the CD4+ T-helper 2 subset producing the canonical cytokines IL-4, IL-5, and IL-13. This picture is now being challenged by the discovery of a more complex pattern of CD4+ T-helper cell subsets that appear during infection, including Tregs, Th17, Tfh, and more recently, Th22, Th9, and ThGM. In addition, a clearer view of the mechanisms by which helminths and their products selectively prime the CD4+ T-cell subsets is emerging. In this review, we have focused on recent data concerning the selective priming, differentiation, and functional role of CD4+ T-helper cell subsets in the context of helminth infection. We argue for a re-evaluation of the original Th2 paradigm and discuss how the observed plasticity of the T-helper subsets may enable the parasitized host to achieve an appropriate compromise between elimination, tissue repair, containment, and pathology.
Collapse
Affiliation(s)
- Tiffany Bouchery
- Malaghan Institute of Medical Research , Wellington , New Zealand
| | - Ryan Kyle
- Malaghan Institute of Medical Research , Wellington , New Zealand
| | - Franca Ronchese
- Malaghan Institute of Medical Research , Wellington , New Zealand
| | - Graham Le Gros
- Malaghan Institute of Medical Research , Wellington , New Zealand ; Victoria University of Wellington , Wellington , New Zealand
| |
Collapse
|
57
|
Schabussova I, Wiedermann U. Allergy and worms: let's bring back old friends? Wien Med Wochenschr 2014; 164:382-91. [PMID: 25281198 DOI: 10.1007/s10354-014-0308-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 08/20/2014] [Indexed: 01/15/2023]
Abstract
PURPOSE OF REVIEW In order to survive in their host, parasitic worms (helminths) have evolved cunning strategies to manipulate the host immune system, some of which may lead to protection from immune dysregulatory diseases such as allergy. Thus, loss of exposure to helminths due to a highly hygienic life style might have contributed to the fact that living in an industrialized country is being associated with an increased prevalence of allergic diseases. However, it must be pointed out that certain helminth infections can in fact induce an allergic phenotype. Factors such as different parasite species, timing of infection in relation to allergic sensitization, or duration and intensity of infection may influence the association between helminth infections and the development or clinical course of allergic disease. In the present article, we review studies that have explored the interaction between helminth infections and allergy in epidemiological and experimental studies. Furthermore, the possibility of using helminths or helminth-derived molecules for the treatment of allergic diseases is discussed with a focus on evidence from clinical trials. RECENT FINDINGS During the past 10 years, many exciting and important studies have found that certain helminth infections protect against the development of allergic diseases. Not surprisingly, several clinical trials investigated the effects of deliberate exposure to parasites like porcine whipworm (Trichuris suis) or hookworm (Necator americanus) to develop "helminth therapies". Although they proved to be a safe option to control aberrant inflammation, the final goal is to identify the parasite-derived immunnomodulatory molecules responsible for protective effects.
Collapse
Affiliation(s)
- Irma Schabussova
- Institut für Spezifische Prophylaxe und Tropenmedizin, Medizinische Universität Wien, Kinderspitalgasse 15, 1090, Vienna, Austria,
| | | |
Collapse
|
58
|
Experimental hookworm infection and gluten microchallenge promote tolerance in celiac disease. J Allergy Clin Immunol 2014; 135:508-16. [PMID: 25248819 DOI: 10.1016/j.jaci.2014.07.022] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND Celiac disease (CeD) is a common gluten-sensitive autoimmune enteropathy. A gluten-free diet is an effective treatment, but compliance is demanding; hence, new treatment strategies for CeD are required. OBJECTIVE Parasitic helminths hold promise for treating inflammatory disorders, so we examined the influence of experimental hookworm infection on the predicted outcomes of escalating gluten challenges in CeD subjects. METHODS A 52-week study was conducted involving 12 adults with diet-managed CeD. Subjects were inoculated with 20 Necator americanus larvae, and escalating gluten challenges consumed as pasta were subsequently administered: (1) 10 to 50 mg for 12 weeks (microchallenge); (2) 25 mg daily + 1 g twice weekly for 12 weeks (GC-1g); and (3) 3 g daily (60-75 straws of spaghetti) for 2 weeks (GC-3g). Symptomatic, serologic, and histological outcomes evaluated gluten toxicity. Regulatory and inflammatory T cell populations in blood and mucosa were examined. RESULTS Two gluten-intolerant subjects were withdrawn after microchallenge. Ten completed GC-1g, 8 of whom enrolled in and completed GC-3g. PRIMARY OUTCOMES median villous height-to-crypt depth ratios (2.60-2.63; P = .98) did not decrease as predicted after GC-1g, and the mean IgA-tissue transglutaminase titers declined, contrary to the predicted rise after GC-3g. SECONDARY OUTCOMES quality of life scores improved (46.3-40.6; P = .05); celiac symptom indices (24.3-24.3; P = .53), intra-epithelial lymphocyte percentages (32.5-35.0; P = .47), and Marsh scores were unchanged by gluten challenge. Intestinal T cells expressing IFNγ were reduced following hookworm infection (23.9%-11.5%; P = .04), with corresponding increases in CD4(+) Foxp3(+) regulatory T cells (0.19%-1.12%; P = .001). CONCLUSIONS Necator americanus and gluten microchallenge promoted tolerance and stabilized or improved all tested indices of gluten toxicity in CeD subjects.
Collapse
|
59
|
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
|
60
|
Ludvigsson JF, Bai JC, Biagi F, Card TR, Ciacci C, Ciclitira PJ, Green PHR, Hadjivassiliou M, Holdoway A, van Heel DA, Kaukinen K, Leffler DA, Leonard JN, Lundin KEA, McGough N, Davidson M, Murray JA, Swift GL, Walker MM, Zingone F, Sanders DS. Diagnosis and management of adult coeliac disease: guidelines from the British Society of Gastroenterology. Gut 2014; 63:1210-28. [PMID: 24917550 PMCID: PMC4112432 DOI: 10.1136/gutjnl-2013-306578] [Citation(s) in RCA: 761] [Impact Index Per Article: 69.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A multidisciplinary panel of 18 physicians and 3 non-physicians from eight countries (Sweden, UK, Argentina, Australia, Italy, Finland, Norway and the USA) reviewed the literature on diagnosis and management of adult coeliac disease (CD). This paper presents the recommendations of the British Society of Gastroenterology. Areas of controversies were explored through phone meetings and web surveys. Nine working groups examined the following areas of CD diagnosis and management: classification of CD; genetics and immunology; diagnostics; serology and endoscopy; follow-up; gluten-free diet; refractory CD and malignancies; quality of life; novel treatments; patient support; and screening for CD.
Collapse
Affiliation(s)
- Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden Department of Paediatrics, Örebro University Hospital, Örebro, Sweden
| | - Julio C Bai
- Department of Medicine, Dr C. Bonorino Udaondo Gastroenterology Hospital, Del Salvador University, Buenos Aires, Argentina
| | - Federico Biagi
- Coeliac Centre/1st Department of Internal Medicine, University of Pavia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Timothy R Card
- University of Nottingham, Department of Epidemiology and Public Health, Nottingham City Hospital, Nottingham, UK
| | - Carolina Ciacci
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Paul J Ciclitira
- Gastroenterology, Division of Nutritional Sciences, King's College London, The Rayne Institute, St Thomas Hospital, London, UK
| | - Peter H R Green
- Coeliac Disease Center at Columbia University, New York, New York, USA
| | | | - Anne Holdoway
- Registered dietitian and representative of the British Dietetic Association, Bath, Somerset, UK
| | - David A van Heel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Katri Kaukinen
- School of Medicine, University of Tampere, Tampere, Finland Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland Department of Medicine, Seinäjoki Central Hospital, Finland
| | - Daniel A Leffler
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan N Leonard
- Department of Dermatology, Imperial College NHS Healthcare Trust, St Mary's Hospital, London, UK
| | - Knut E A Lundin
- Department of Gastroenterology, Centre for Immune Regulation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Mike Davidson
- Patient Representative & Regional Chairman for Coeliac UK, Sheffield, UK
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Department of Immunology Mayo Clinic, Rochester, Minnesota, USA
| | - Gillian L Swift
- Department of Gastroenterology, University Hospital Llandough, Wales, UK
| | - Marjorie M Walker
- Anatomical Pathology, University of Newcastle, Faculty of Health and Medicine, School of Medicine & Public Health, Callaghan, Australia
| | - Fabiana Zingone
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - David S Sanders
- Gastroenterology and Liver Unit, Royal Hallamshire Hospital & University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
61
|
Maizels RM, McSorley HJ, Smyth DJ. Helminths in the hygiene hypothesis: sooner or later? Clin Exp Immunol 2014; 177:38-46. [PMID: 24749722 PMCID: PMC4089153 DOI: 10.1111/cei.12353] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2014] [Indexed: 02/07/2023] Open
Abstract
There is increasing recognition that exposures to infectious agents evoke fundamental effects on the development and behaviour of the immune system. Moreover, where infections (especially parasitic infections) have declined, immune responses appear to be increasingly prone to hyperactivity. For example, epidemiological studies of parasite-endemic areas indicate that prenatal or early-life experience of infections can imprint an individual's immunological reactivity. However, the ability of helminths to dampen pathology in established inflammatory diseases implies that they can have therapeutic effects even if the immune system has developed in a low-infection setting. With recent investigations of how parasites are able to modulate host immune pathology at the level of individual parasite molecules and host cell populations, we are now able to dissect the nature of the host-parasite interaction at both the initiation and recall phases of the immune response. Thus the question remains - is the influence of parasites on immunity one that acts primarily in early life, and at initiation of the immune response, or in adulthood and when recall responses occur? In short, parasite immunosuppression - sooner or later?
Collapse
Affiliation(s)
- R M Maizels
- Institute for Immunology and Infection Research, Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
62
|
Wammes LJ, Mpairwe H, Elliott AM, Yazdanbakhsh M. Helminth therapy or elimination: epidemiological, immunological, and clinical considerations. THE LANCET. INFECTIOUS DISEASES 2014; 14:1150-1162. [PMID: 24981042 DOI: 10.1016/s1473-3099(14)70771-6] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Deworming is rightly advocated to prevent helminth-induced morbidity. Nevertheless, in affluent countries, the deliberate infection of patients with worms is being explored as a possible treatment for inflammatory diseases. Several clinical trials are currently registered, for example, to assess the safety or efficacy of Trichuris suis ova in allergies, inflammatory bowel diseases, multiple sclerosis, rheumatoid arthritis, psoriasis, and autism, and the Necator americanus larvae for allergic rhinitis, asthma, coeliac disease, and multiple sclerosis. Studies in animals provide strong evidence that helminths can not only downregulate parasite-specific immune responses, but also modulate autoimmune and allergic inflammatory responses and improve metabolic homoeostasis. This finding suggests that deworming could lead to the emergence of inflammatory and metabolic conditions in countries that are not prepared for these new epidemics. Further studies in endemic countries are needed to assess this risk and to enhance understanding of how helminths modulate inflammatory and metabolic pathways. Studies are similarly needed in non-endemic countries to move helminth-related interventions that show promise in animals, and in phase 1 and 2 studies in human beings, into the therapeutic development pipeline.
Collapse
Affiliation(s)
- Linda J Wammes
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Harriet Mpairwe
- MRC/Uganda Virus Research Institute, Uganda Research Unit on AIDS, Entebbe, Uganda
| | - Alison M Elliott
- MRC/Uganda Virus Research Institute, Uganda Research Unit on AIDS, Entebbe, Uganda; London School of Hygiene and Tropical Medicine, London, UK
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
63
|
Abstract
Human gastrointestinal bacteria often share their environment with parasitic worms, allowing physical and physiological interaction between the two groups. Such associations have the potential to affect host health as well as the bacterial and helminth populations. Although still in its early stages, research on the interaction between the microbiome and parasitic helminths in humans offers the potential to improve health by manipulating the microbiome. Previously, supplementation with various nutritional compounds has been found to increase the abundance of potentially beneficial gut commensal bacteria. Thus, nutritional microbiome manipulation to produce an environment which may decrease malnutrition associated with helminth infection and/or aid host recovery from disease is conceivable. This review discusses the influence of the gut microbiota and helminths on host nutrition and immunity and the subsequent effects on the human host's overall health. It also discusses changes occurring in the microbiota upon helminth infections and the underlying mechanisms leading to these changes. There are still significant knowledge gaps which need to be filled before meaningful progress can be made in translating knowledge from studying the human gut microbiome into therapeutic strategies. Ultimately this review aims to discuss our current knowledge as well as highlight areas requiring further investigation.
Collapse
|
64
|
Hotez PJ, Diemert D, Bacon KM, Beaumier C, Bethony JM, Bottazzi ME, Brooker S, Couto AR, Freire MDS, Homma A, Lee BY, Loukas A, Loblack M, Morel CM, Oliveira RC, Russell PK. The Human Hookworm Vaccine. Vaccine 2014; 31 Suppl 2:B227-32. [PMID: 23598487 PMCID: PMC3988917 DOI: 10.1016/j.vaccine.2012.11.034] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 10/17/2012] [Accepted: 11/09/2012] [Indexed: 01/08/2023]
Abstract
Hookworm infection is one of the world's most common neglected tropical diseases and a leading cause of iron deficiency anemia in low- and middle-income countries. A Human Hookworm Vaccine is currently being developed by the Sabin Vaccine Institute and is in phase 1 clinical testing. The candidate vaccine is comprised of two recombinant antigens known as Na-GST-1 and Na-APR-1, each of which is an important parasite enzyme required for hookworms to successfully utilize host blood as a source of energy. The recombinant proteins are formulated on Alhydrogel® and are being tested in combination with a synthetic Toll-like receptor 4 agonist. The aim of the vaccine is to induce anti-enzyme antibodies that will reduce both host blood loss and the number of hookworms attached to the gut. Transfer of the manufacturing technology to the Oswaldo Cruz Foundation (FIOCRUZ)/Bio-Manguinhos (a Brazilian public sector developing country vaccine manufacturer) is planned, with a clinical development plan that could lead to registration of the vaccine in Brazil. The vaccine would also need to be introduced in the poorest regions of Africa and Asia, where hookworm infection is highly endemic. Ultimately, the vaccine could become an essential tool for achieving hookworm control and elimination, a key target in the 2012 London Declaration on Neglected Tropical Diseases.
Collapse
Affiliation(s)
- Peter J Hotez
- Sabin Vaccine Institute Product Development Partnership, Houston, TX, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Nunn CL, Brezine C, Jolles AE, Ezenwa VO. Interactions between Micro- and Macroparasites Predict Microparasite Species Richness across Primates. Am Nat 2014; 183:494-505. [DOI: 10.1086/675362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
66
|
Heylen M, Ruyssers NE, Gielis EM, Vanhomwegen E, Pelckmans PA, Moreels TG, De Man JG, De Winter BY. Of worms, mice and man: an overview of experimental and clinical helminth-based therapy for inflammatory bowel disease. Pharmacol Ther 2014; 143:153-67. [PMID: 24603369 DOI: 10.1016/j.pharmthera.2014.02.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 12/17/2022]
Abstract
The incidence of inflammatory and autoimmune disorders is highest in well-developed countries which is directly related to their higher hygienic standards: it is suggested that the lack of exposure to helminths contributes to the susceptibility for immune-related diseases. Epidemiological, experimental and clinical data support the idea that helminths provide protection against immune-mediated diseases such as inflammatory bowel disease (IBD). The most likely mechanism for the suppression of immune responses by helminths is the release of helminth-derived immunomodulatory molecules. This article reviews the experimental and clinical studies investigating the therapeutic potential of helminth-based therapy in IBD and also focuses on the current knowledge of its immunomodulatory mechanisms of action highlighting innate as well as adaptive immune mechanisms. Identifying the mechanisms by which these helminths and helminth-derived molecules modulate the immune system will help in creating novel drugs for the treatment of IBD and other disorders that result from an overactive immune response.
Collapse
Affiliation(s)
- Marthe Heylen
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Nathalie E Ruyssers
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Els M Gielis
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Els Vanhomwegen
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Paul A Pelckmans
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Division of Gastroenterology & Hepatology, Antwerp, Belgium
| | - Tom G Moreels
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Division of Gastroenterology & Hepatology, Antwerp, Belgium
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
67
|
Hassan K, A-Kader H. Celiac disease: the search for adjunctive or alternative therapies. Expert Rev Gastroenterol Hepatol 2014; 8:313-21. [PMID: 24490653 DOI: 10.1586/17474124.2014.882769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Celiac disease is a widespread disorder caused by intolerance to gluten, a common protein in food. Currently, a life-long gluten-free diet is the only available treatment for patients with celiac disease. However, adherence to gluten-free diet is difficult due to the widespread use of wheat-derived gluten in the food industry. Therefore, there is a pressing need for the development of novel non-dietary therapies. In this article, we will review several promising strategies focusing on reducing gluten immunogenicity or sequestering to gluten prevent its uptake by the intestinal epithelium. Other possible treatment strategies that will be reviewed include the suppression of the adaptive immune response, permeability modulation and the use of systemic T-cell or cytokine blockers.
Collapse
Affiliation(s)
- Kareem Hassan
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, The University of Arizona, Tucson AZ, USA
| | | |
Collapse
|
68
|
Samaşca G, Sur G, Lupan I. Current trends and investigative developments in celiac disease. Immunol Invest 2014; 42:273-84. [PMID: 23883196 DOI: 10.3109/08820139.2013.777074] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Celiac disease has become extensively studied. What could be the cause? Increasing the accuracy of diagnostic tests for celiac disease has led to more discovered cases. Serological diagnosis of celiac disease has undergone important changes in recent years. Application of serological tests has reflected the diagnostic performance of tissue transglutaminase antibody and endomysial antibody as screening tests for celiac disease but also the progress of new serological tests as the antibodies against synthetic deamidated gliadin peptides. Serological tests are largely responsible for the recognition that celiac disease is not a rare disease. The Consensus in celiac disease from 2008 conducted under the aegis of the European Society of Pediatric Gastroenterology, Hepatology and Nutrition jointly with North American Society of Pediatric Gastroenterology, Hepatology and Nutrition which agreed that "Celiac disease is an immune-mediated enteropathy that can affect any system or organ and that can present itself with a wide range of clinical manifestations of variable severity" was confirmed. But increasing prevalence of this disease has led to the need for new methods of treatment among patients with celiac disease. Studies on quality of life of patients with celiac disease have questioned the gluten-free diet. As such new therapies, like TG2 inhibitors, the copolymer P (HEMA-co-SS) and other new experimental therapies, have emerged in celiac disease. The new therapies in celiac disease are based on new investigations in gluten toxicity screening, like K562(S)-cell agglutination, A1 and G12 monoclonal antibodies and proteomics. In this paper we want to present the investigative developments in celiac disease. We also want to find whether a new treatment in celiac disease is necessary.
Collapse
Affiliation(s)
- Gabriel Samaşca
- Department of Immunology, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | | | | |
Collapse
|
69
|
Abstract
In the last 50 years, environmental factors such as helminth infections have been proposed to explain why autoimmunity is less prevalent in the developing world; this proposal has been termed the hygiene or old friends hypothesis. The epidemiology of MS shows an inverse correlation with helminth infections. Positive effects of helminths in animal models of MS and observational studies in people with MS naturally infected with helminths suggest that those organisms can act as immune regulators and led to clinical trials of helminth therapy. The goal of helminth therapy is to introduce parasitic organisms into people with MS in a controlled and predictable fashion, and to prevent immune-mediated disease without increasing the risk of pathology with high parasite load. This chapter focuses on intestinal worms as they are the current choice as a therapeutic strategy in a number of autoimmune diseases, including MS. Here we review current data regarding the rationale and the current state of research in the field of helminth therapies in MS.
Collapse
|
70
|
Abstract
Coeliac disease is a common and fairly well-characterized systemic disorder that mainly affects the small intestine, but also has extraintestinal manifestations. The environmental trigger (gluten derived from wheat, rye and barley), the genetic predisposition conferred by the HLA-DQ2 and HLA-DQ8 haplotypes and many steps in the disease pathogenesis are known. This knowledge has enabled researchers to suggest novel alternative treatments or adjunctive therapies to the gluten-free diet, which is currently the only available and effective treatment for the condition. This Review focuses on emerging and potential treatment strategies that are based on the current concept of the disease pathophysiology. The search for novel future treatment modes, including nonpharmacological and pharmacological approaches, is also outlined. The potential pitfalls associated with the various research avenues are also discussed.
Collapse
|
71
|
Tennyson CA, Simpson S, Lebwohl B, Lewis S, Green PHR. Interest in medical therapy for celiac disease. Therap Adv Gastroenterol 2013; 6:358-64. [PMID: 24003336 PMCID: PMC3756636 DOI: 10.1177/1756283x13492580] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES A gluten-free diet is the treatment for celiac disease, but pharmaceutical agents are being developed. The level of interest amongst patients in using a medication to treat celiac disease is unknown. This study examined the level of interest amongst patients in medication to treat celiac disease. METHODS A questionnaire was distributed to celiac disease patients and data were collected on demographics, presentation, and interest in medication. Three validated celiac disease-specific instruments were incorporated: Celiac Disease Associated Quality of Life, the Celiac Symptom Index, and the Celiac Dietary Adherence Test. RESULTS Responses were received from 365 individuals with biopsy-proven celiac disease. Respondents were 78% (n = 276) female, 48% (n = 170) over 50 years of age, and experienced a classical (diarrhea predominant) presentation in 44% (n = 154). Of the 339 individuals answering the question regarding use of a medication to treat celiac disease, 66% were interested. Interest was greatest in older individuals (71% >50 years of age versus 60% <50 years of age, p = 0.0415), men (78% men versus 62% women, p = 0.0083), frequent restaurant customers (76% versus 58%, p = 0.0006), those dissatisfied with their weight (73% versus 51%, p = 0.0003) and those concerned with the cost of a gluten-free diet (77% versus 64%, p = 0.0176). Length of time since diagnosis, education, presentation, and symptoms with gluten exposure did not demonstrate any effect. Interest in medication was associated with a worse quality of life (CD-QOL 69.4 versus 80.1, p < 0.0001). CONCLUSIONS Most individuals with celiac disease are interested in using a medication. Interest was highest among men, older individuals, frequent restaurant customers, individuals dissatisfied with their weight or concerned with the cost of a gluten-free diet, and those with a worse quality of life.
Collapse
Affiliation(s)
| | - Suzanne Simpson
- Columbia University College of Physicians and Surgeons, Division of Digestive and Liver Diseases, New York, NY, USA
| | - Benjamin Lebwohl
- Columbia University College of Physicians and Surgeons, Division of Digestive and Liver Diseases, New York, NY, USA
| | - Suzanne Lewis
- Columbia University College of Physicians and Surgeons, Division of Digestive and Liver Diseases, New York, NY, USA
| | - Peter H. R. Green
- The Celiac Disease Center at Columbia University, 180 Fort Washington Avenue, #936, New York, NY 10032, USA
| |
Collapse
|
72
|
Prasanphanich NS, Mickum ML, Heimburg-Molinaro J, Cummings RD. Glycoconjugates in host-helminth interactions. Front Immunol 2013; 4:240. [PMID: 24009607 PMCID: PMC3755266 DOI: 10.3389/fimmu.2013.00240] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/03/2013] [Indexed: 12/20/2022] Open
Abstract
Helminths are multicellular parasitic worms that comprise a major class of human pathogens and cause an immense amount of suffering worldwide. Helminths possess an abundance of complex and unique glycoconjugates that interact with both the innate and adaptive arms of immunity in definitive and intermediate hosts. These glycoconjugates represent a major untapped reservoir of immunomodulatory compounds, which have the potential to treat autoimmune and inflammatory disorders, and antigenic glycans, which could be exploited as vaccines and diagnostics. This review will survey current knowledge of the interactions between helminth glycans and host immunity and highlight the gaps in our understanding which are relevant to advancing therapeutics, vaccine development, and diagnostics.
Collapse
Affiliation(s)
- Nina Salinger Prasanphanich
- Department of Biochemistry, Glycomics Center of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Megan L. Mickum
- Department of Biochemistry, Glycomics Center of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Jamie Heimburg-Molinaro
- Department of Biochemistry, Glycomics Center of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard D. Cummings
- Department of Biochemistry, Glycomics Center of Emory University, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
73
|
Schabussova I, Ul-Haq O, Hoflehner E, Akgün J, Wagner A, Loupal G, Joachim A, Ruttkowski B, Maizels RM, Wiedermann U. Oesophagostomum dentatum extract modulates T cell-dependent immune responses to bystander antigens and prevents the development of allergy in mice. PLoS One 2013; 8:e67544. [PMID: 23844022 PMCID: PMC3699627 DOI: 10.1371/journal.pone.0067544] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 05/19/2013] [Indexed: 12/26/2022] Open
Abstract
One third of the human population is currently infected by one or more species of parasitic helminths. Certain helminths establish long-term chronic infections resulting in a modulation of the host’s immune system with attenuated responsiveness to “bystander” antigens such as allergens or vaccines. In this study we investigated whether parasite-derived products suppress the development of allergic inflammation in a mouse model. We show that extract derived from adult male Oesophagostomum dentatum (eMOD) induced Th2 and regulatory responses in BALB/c mice. Stimulation of bone marrow-derived dendritic cells induced production of regulatory cytokines IL-10 and TGF-beta. In a mouse model of birch pollen allergy, co-administration of eMOD with sensitizing allergen Bet v 1 markedly reduced the production of allergen-specific antibodies in serum as well as IgE-dependent basophil degranulation. Furthermore, eMOD prevented the development of airway inflammation, as demonstrated by attenuation of bronchoalveolar lavages eosinophil influx, peribronchial inflammatory infiltrate, and mucus secretion in lungs and IL-4 and IL-5 levels in lung cell cultures. Reduced secretion of Th2-related cytokines by birch pollen-re-stimulated splenocytes and mesenteric lymph node cells was observed in eMOD-treated/sensitized and challenged mice in comparison to sensitized and challenged controls. The suppressive effects of eMOD were heat-stable. Immunization with model antigens in the presence of eMOD reduced production of antibodies to thymus-dependent but not to thymus-independent antigen, suggesting that suppression of the immune responses by eMOD was mediated by interference with antigen presenting cell or T helper cell function but did not directly suppress B cell function. In conclusion, we have shown that eMOD possesses immunomodulatory properties and that heat-stable factors in eMOD are responsible for the dramatic suppression of allergic responses in a mouse model of type I allergy. The identification and characterization of parasite-derived immune-modulating molecules might have potential for designing novel prophylactic/therapeutic strategies for immune-mediated diseases.
Collapse
Affiliation(s)
- Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Hookworm excretory/secretory products induce interleukin-4 (IL-4)+ IL-10+ CD4+ T cell responses and suppress pathology in a mouse model of colitis. Infect Immun 2013; 81:2104-11. [PMID: 23545299 DOI: 10.1128/iai.00563-12] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Evidence from human studies and mouse models shows that infection with parasitic helminths has a suppressive effect on the pathogenesis of some inflammatory diseases. Recently, we and others have shown that some of the suppressive effects of hookworms reside in their excretory/secretory (ES) products. Here, we demonstrate that ES products of the hookworm Ancylostoma caninum (AcES) suppress intestinal pathology in a model of chemically induced colitis. This suppression was associated with potent induction of a type 2 cytokine response characterized by coexpression of interleukin-4 (IL-4) and IL-10 by CD4(+) T cells, downregulation of proinflammatory cytokine expression in the draining lymph nodes and the colon, and recruitment of alternatively activated (M2) macrophages and eosinophils to the site of ES administration. Protease digestion and heat denaturation of AcES resulted in impaired induction of CD4(+) IL-4(+) IL-10(+) cell responses and diminished ability to suppress colitis, indicating that protein component(s) are responsible for some of the immunosuppressive effects of AcES. Identification of the specific parasite-derived molecules responsible for reducing pathology during chemically induced colitis could lead to the development of novel therapeutics for the treatment of human inflammatory bowel disease.
Collapse
|
75
|
Zaccone P, Cooke A. Vaccine against autoimmune disease: can helminths or their products provide a therapy? Curr Opin Immunol 2013; 25:418-23. [PMID: 23465465 DOI: 10.1016/j.coi.2013.02.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 02/12/2013] [Indexed: 12/25/2022]
Abstract
There is an increasing interest in exploiting the immunomodulatory effects of helminths and their products in treatment of diseases such as allergy, autoimmunity and inflammatory bowel disease. Detailed examination of the ways in which helminth derived products interact with the host immune system and with host physiology has revealed that these may be multifaceted and have almost certainly arisen following co-evolution of helminths and their hosts. Clinical trials have been initiated with encouraging results in the treatment of inflammatory bowel disease and also Multiple Sclerosis. Identification of key pathways that are manipulated by helminths to ameliorate ongoing inflammatory conditions increases the prospect of developing novel therapies for the treatment and possible prevention of a range of debilitating and life threatening conditions.
Collapse
Affiliation(s)
- Paola Zaccone
- Department of Pathology, University of Cambridge, Tennis Court Rd., Cambridge, UK
| | | |
Collapse
|
76
|
Leung JM, Loke P. A role for IL-22 in the relationship between intestinal helminths, gut microbiota and mucosal immunity. Int J Parasitol 2013; 43:253-7. [PMID: 23178750 PMCID: PMC3955947 DOI: 10.1016/j.ijpara.2012.10.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 10/23/2012] [Accepted: 10/24/2012] [Indexed: 12/22/2022]
Abstract
The intestinal tract is home to nematodes as well as commensal bacteria (microbiota), which have coevolved with the mammalian host. The mucosal immune system must balance between an appropriate response to dangerous pathogens and an inappropriate response to commensal microbiota that may breach the epithelial barrier, in order to maintain intestinal homeostasis. IL-22 has been shown to play a critical role in maintaining barrier homeostasis against intestinal pathogens and commensal bacteria. Here we review the advances in our understanding of the role of IL-22 in helminth infections, as well as in response to commensal and pathogenic bacteria of the intestinal tract. We then consider the relationship between intestinal helminths and gut microbiota and hypothesize that this relationship may explain how helminths may improve symptoms of inflammatory bowel diseases. We propose that by inducing an immune response that includes IL-22, intestinal helminths may enhance the mucosal barrier function of the intestinal epithelium. This may restore the mucosal microbiota populations from dysbiosis associated with colitis and improve intestinal homeostasis.
Collapse
Affiliation(s)
| | - P’ng Loke
- Department of Microbiology, New York University, New York, NY, USA
| |
Collapse
|
77
|
Stoven S, Murray JA, Marietta EV. Latest in vitro and in vivo models of celiac disease. Expert Opin Drug Discov 2013; 8:445-57. [PMID: 23293929 DOI: 10.1517/17460441.2013.761203] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Currently, the only treatment for celiac disease is a gluten-free diet, and there is an increased desire for alternative therapies. In vitro and in vivo models of celiac disease have been generated in order to better understand the pathogenesis of celiac disease, and this review will discuss these models as well as the testing of alternative therapies using these models. AREAS COVERED The research discussed describes the different in vitro and in vivo models of celiac disease that currently exist and how they have contributed to our understanding of how gluten can stimulate both innate and adaptive immune responses in celiac patients. We also provide a summary on the alternative therapies that have been tested with these models and discuss whether subsequent clinical trials were done based on these tests done with these models of celiac disease. EXPERT OPINION Only a few of the alternative therapies that have been tested with animal models have gone on to clinical trials; however, those that did go on to clinical trial have provided promising results from a safety standpoint. Further trials are required to determine if some of these therapies may serve as an effective adjunct to a gluten-free diet to alleviate the adverse affects associated with accidental gluten exposure. A "magic-bullet" approach may not be the answer to celiac disease, but possibly a future cocktail of these different therapeutics may allow celiac patients to consume an unrestricted diet.
Collapse
Affiliation(s)
- Samantha Stoven
- Mayo Clinic, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN 55905, USA
| | | | | |
Collapse
|
78
|
Croese J, Gaze ST, Loukas A. Changed gluten immunity in celiac disease by Necator americanus provides new insights into autoimmunity. Int J Parasitol 2013; 43:275-82. [PMID: 23291460 DOI: 10.1016/j.ijpara.2012.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/09/2012] [Accepted: 12/12/2012] [Indexed: 12/26/2022]
Abstract
We recently completed clinical trials in people with diet-treated celiac disease who were purposefully infected with the ubiquitous human hookworm, Necator americanus. Hookworm infection elicited not only parasite-specific immunity but also modified the host's immune response to gluten. After infection, mucosal IL-1β and IL-22 responses were enhanced, but IFNγ and IL-17A levels and circulating regulatory T cells following gluten challenge were suppressed, and the adaptive response to gluten acquired a helper T cell type-2 profile. In this review, we briefly, (i) highlight the utility celiac disease offers autoimmune research, (ii) discuss safety and personal experience with N. americanus, (iii) summarise the direct and bystander impact that hookworm infection has on mucosal immunity to the parasite and gluten, respectively, and (iv) speculate why this hookworm's success depends on healing its host and how this might impact on a propensity to autoimmunity.
Collapse
Affiliation(s)
- John Croese
- The Department of Gastroenterology, The Townsville Hospital, Townsville, Australia.
| | | | | |
Collapse
|
79
|
McSorley HJ, Hewitson JP, Maizels RM. Immunomodulation by helminth parasites: defining mechanisms and mediators. Int J Parasitol 2013; 43:301-10. [PMID: 23291463 DOI: 10.1016/j.ijpara.2012.11.011] [Citation(s) in RCA: 225] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 11/28/2012] [Accepted: 11/29/2012] [Indexed: 12/26/2022]
Abstract
Epidemiological and interventional human studies, as well as experiments in animal models, strongly indicate that helminth parasitic infections can confer protection from immune dysregulatory diseases such as allergy, autoimmunity and colitis. Here, we review the immunological pathways that helminths exploit to downregulate immune responses, both against bystander specificities such as allergens and against antigens from the parasites themselves. In particular, we focus on a highly informative laboratory system, the mouse intestinal nematode, Heligmosomoides polygyrus, as a tractable model of host-parasite interaction at the cellular and molecular levels. Analysis of the molecules released in vitro (as excretory-secretory products) and their cellular targets is identifying individual parasite molecules and gene families implicated in immunomodulation, and which hold potential for future human therapy of immunopathological conditions.
Collapse
Affiliation(s)
- Henry J McSorley
- Institute of Immunology and Infection Research, University of Edinburgh, UK.
| | | | | |
Collapse
|
80
|
Matoori S, Fuhrmann G, Leroux JC. Celiac disease: a challenging disease for pharmaceutical scientists. Pharm Res 2012; 30:619-26. [PMID: 23229860 DOI: 10.1007/s11095-012-0951-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Accepted: 11/30/2012] [Indexed: 12/19/2022]
Abstract
Celiac disease (CD) is an immune-mediated enteropathy triggered by the ingestion of gluten-containing grains that affects ~1% of the white ethnic population. In the last decades, a rise in prevalence of CD has been observed that cannot be fully explained by improved diagnostics. Genetic predisposition greatly influences the susceptibility of individuals towards CD, though environmental factors also play a role. With no pharmacological treatments available, the only option to keep CD in remission is a strict and permanent exclusion of dietary gluten. Such a gluten-free diet is difficult to maintain because of gluten's omnipresence in food (e.g., additive in processed food). The development of adjuvant therapies which would permit the intake of small amounts of gluten would be desirable to improve the quality of life of patients on a gluten-free diet. Such therapies include gluten-degrading enzymes, polymeric binders, desensitizing vaccines, anti-inflammatory drugs, transglutaminase 2 inhibitors, and HLA-DQ2 blockers. However, many of these approaches pose pharmaceutical challenges with respect to drug formulation and stability, or application route and dosing interval. This perspective article discusses how pharmaceutical scientists may deal with these challenges and contribute to the implementation of novel therapeutic options for patients with CD.
Collapse
Affiliation(s)
- Simon Matoori
- Department of Chemistry and Applied Biosciences, ETH Zurich Institute of Pharmaceutical Sciences, Wolfgang-Pauli-Str. 10, HCI, 8093, Zurich, Switzerland
| | | | | |
Collapse
|
81
|
The hookworm pharmacopoeia for inflammatory diseases. Int J Parasitol 2012; 43:225-31. [PMID: 23220091 DOI: 10.1016/j.ijpara.2012.11.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/12/2012] [Accepted: 11/14/2012] [Indexed: 01/18/2023]
Abstract
In the developed world, declining prevalence of parasitic infections correlates with increased incidence of allergic and autoimmune disorders. Current treatments for these chronic inflammatory conditions have little to no effect on their prevalence and are referred to as "controllers" rather than cures. There has been limited success in therapeutically targeting allergic and autoimmune pathways, leaving an unmet need for development of effective anti-inflammatories. We discuss the benefit of hookworm infections and the parasite's ability to condition the immune system to prevent allergic asthma and inflammatory bowel diseases. We then examine the immunomodulatory properties of selected hookworm-derived proteins in these two models of inflammation. While hookworm protein therapy has yet to be fully exploited, the identification of these proteins and the mechanisms by which they skew the immune system will provide new avenues for controlling and optimally reversing key pathological processes important in allergic and inflammatory bowel diseases.
Collapse
|
82
|
Broadhurst MJ, Ardeshir A, Kanwar B, Mirpuri J, Gundra UM, Leung JM, Wiens KE, Vujkovic-Cvijin I, Kim CC, Yarovinsky F, Lerche NW, McCune JM, Loke P. Therapeutic helminth infection of macaques with idiopathic chronic diarrhea alters the inflammatory signature and mucosal microbiota of the colon. PLoS Pathog 2012; 8:e1003000. [PMID: 23166490 PMCID: PMC3499566 DOI: 10.1371/journal.ppat.1003000] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 09/13/2012] [Indexed: 12/19/2022] Open
Abstract
Idiopathic chronic diarrhea (ICD) is a leading cause of morbidity amongst rhesus monkeys kept in captivity. Here, we show that exposure of affected animals to the whipworm Trichuris trichiura led to clinical improvement in fecal consistency, accompanied by weight gain, in four out of the five treated monkeys. By flow cytometry analysis of pinch biopsies collected during colonoscopies before and after treatment, we found an induction of a mucosal TH2 response following helminth treatment that was associated with a decrease in activated CD4+ Ki67+ cells. In parallel, expression profiling with oligonucleotide microarrays and real-time PCR analysis revealed reductions in TH1-type inflammatory gene expression and increased expression of genes associated with IgE signaling, mast cell activation, eosinophil recruitment, alternative activation of macrophages, and worm expulsion. By quantifying bacterial 16S rRNA in pinch biopsies using real-time PCR analysis, we found reduced bacterial attachment to the intestinal mucosa post-treatment. Finally, deep sequencing of bacterial 16S rRNA revealed changes to the composition of microbial communities attached to the intestinal mucosa following helminth treatment. Thus, the genus Streptophyta of the phylum Cyanobacteria was vastly increased in abundance in three out of five ICD monkeys relative to healthy controls, but was reduced to control levels post-treatment; by contrast, the phylum Tenericutes was expanded post-treatment. These findings suggest that helminth treatment in primates can ameliorate colitis by restoring mucosal barrier functions and reducing overall bacterial attachment, and also by altering the communities of attached bacteria. These results also define ICD in monkeys as a tractable preclinical model for ulcerative colitis in which these effects can be further investigated. Young macaques kept in captivity at Primate Research Centers often develop chronic diarrhea, which is difficult to treat because it is poorly understood. This disease shares many features with ulcerative colitis, which is an autoimmune disease affecting the intestinal tract of humans. Recently, parasitic worms have been used in clinical trials to treat inflammatory bowel diseases in humans with positive results, but very little is known about how worms can improve symptoms. We performed a trial where we treated macaques suffering from chronic diarrhea with human whipworms, collecting gut biopsies before and after treatment. We found that 4 out of the 5 treated macaques improved their symptoms and studied the changes in their gut immune responses, as they got better. We found that after treatment with worms, the monkeys had less bacteria attached to their intestinal wall and a reduced inflammatory response to the gut bacteria. Additionally, the composition of gut bacteria was altered in the sick macaques and was restored close to normal after treatment with whipworms. These results provide a potential mechanism by which parasitic worms may improve the symptoms of intestinal inflammation, by reducing the immune response against intestinal bacteria.
Collapse
Affiliation(s)
- Mara Jana Broadhurst
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MJB); (PL)
| | - Amir Ardeshir
- California National Primate Research Center, Davis, California, United States of America
| | - Bittoo Kanwar
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Julie Mirpuri
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Uma Mahesh Gundra
- Department of Microbiology, New York University, New York, New York, United States of America
| | - Jacqueline M. Leung
- Department of Microbiology, New York University, New York, New York, United States of America
| | - Kirsten E. Wiens
- Department of Microbiology, New York University, New York, New York, United States of America
| | - Ivan Vujkovic-Cvijin
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Charlie C. Kim
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Felix Yarovinsky
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Nicholas W. Lerche
- California National Primate Research Center, Davis, California, United States of America
| | - Joseph M. McCune
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - P'ng Loke
- Department of Microbiology, New York University, New York, New York, United States of America
- * E-mail: (MJB); (PL)
| |
Collapse
|
83
|
Abstract
PURPOSE OF REVIEW There is something about living in an industrialized country that dramatically increases the risk of acquiring inflammatory bowel disease (IBD). Loss of routine exposure to parasitic worms (helminths), due to modern highly hygienic life styles, likely contributes to this risk. This article reviews current understanding on how helminths influence intestinal inflammation and mucosal immune responses. RECENT FINDINGS IBD emerges in populations as regions develop socioeconomically and lose exposure to previously ubiquitous helminthic infections. Helminthic infections provided strong selective pressure for the dissemination of gene variants, many of which predispose to development of IBD. In animal models of IBD, helminth colonization suppresses intestinal inflammation through multiple mechanisms including induction of innate and adaptive regulatory circuits. Trials using helminths like hookworm (Necator americanus) or porcine whipworm (Trichuris suis) show that they are safe and may be effective therapies for the control of the aberrant intestinal inflammation seen in Crohn's disease and ulcerative colitis. SUMMARY Evidence is accumulating that highly hygienic living conditions create risk for developing immune-mediated disease such as IBD. To live in their host, helminths have developed the ability to activate cells of innate and adaptive immunity that suppress inflammation. Therapeutic trials using helminths are in progress.
Collapse
|
84
|
McSorley HJ, O'Gorman MT, Blair N, Sutherland TE, Filbey KJ, Maizels RM. Suppression of type 2 immunity and allergic airway inflammation by secreted products of the helminth Heligmosomoides polygyrus. Eur J Immunol 2012; 42:2667-82. [PMID: 22706967 PMCID: PMC4916998 DOI: 10.1002/eji.201142161] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 05/11/2012] [Accepted: 06/06/2012] [Indexed: 01/23/2023]
Abstract
Allergic asthma is less prevalent in countries with parasitic helminth infections, and mice infected with parasites such as Heligmosomoides polygyrus are protected from allergic airway inflammation. To establish whether suppression of allergy could be mediated by soluble products of this helminth, we tested H. polygyrus excretory-secretory (HES) material for its ability to impair allergic inflammation. When HES was added to sensitising doses of ovalbumin, the subsequent allergic airway response was suppressed, with ablated cell infiltration, a lower ratio of effector (CD4(+) CD25(+) Foxp3(-) ) to regulatory (CD4(+) Foxp3(+) ) T (Treg) cells, and reduced Th1, Th2 and Th17 cytokine production. HES exposure reduced IL-5 responses and eosinophilia, abolished IgE production and inhibited the type 2 innate molecules arginase-1 and RELM-α (resistin-like molecule-α). Although HES contains a TGF-β-like activity, similar effects in modulating allergy were not observed when administering mammalian TGF-β alone. HES also protected previously sensitised mice, suppressing recruitment of eosinophils to the airways when given at challenge, but no change in Th or Treg cell populations was apparent. Because heat-treatment of HES did not impair suppression at sensitisation, but compromised its ability to suppress at challenge, we propose that HES contains distinct heat-stable and heat-labile immunomodulatory molecules, which modulate pro-allergic adaptive and innate cell populations.
Collapse
Affiliation(s)
| | | | | | | | | | - Rick M Maizels
- Institute of Immunology and Infection Research University of Edinburgh, Edinburgh, EH9 3JT, UK
| |
Collapse
|
85
|
Mukherjee R, Kelly CP, Schuppan D. Nondietary therapies for celiac disease. Gastrointest Endosc Clin N Am 2012; 22:811-31. [PMID: 23083995 DOI: 10.1016/j.giec.2012.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Currently, the only available therapy for celiac disease is strict lifelong adherence to a gluten-free diet (GFD). Although safe and effective, the GFD is not ideal. It is frequently expensive, of limited nutritional value, and not readily available in many countries. Consequently, a need exists for novel, nondietary therapies for celiac disease. Based on the current understanding of celiac disease pathogenesis, several potential targets of therapeutic intervention exist. These novel strategies provide promise of alternative, adjunctive treatment options but also raise important questions regarding safety, efficacy, and monitoring of long-term treatment effect.
Collapse
Affiliation(s)
- Rupa Mukherjee
- Department of Medicine, Division of Gastroenterology, The Celiac Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | | | | |
Collapse
|
86
|
Abstract
Helminth parasites infect almost one-third of the world's population, primarily in tropical regions. However, regions where helminth parasites are endemic record much lower prevalences of allergies and autoimmune diseases, suggesting that parasites may protect against immunopathological syndromes. Most helminth diseases are spectral in nature, with a large proportion of relatively asymptomatic cases and a subset of patients who develop severe pathologies. The maintenance of the asymptomatic state is now recognized as reflecting an immunoregulatory environment, which may be promoted by parasites, and involves multiple levels of host regulatory cells and cytokines; a breakdown of this regulation is observed in pathological disease. Currently, there is much interest in whether helminth-associated immune regulation may ameliorate allergy and autoimmunity, with investigations in both laboratory models and human trials. Understanding and exploiting the interactions between these parasites and the host regulatory network are therefore likely to highlight new strategies to control both infectious and immunological diseases.
Collapse
|
87
|
Pearson MS, Tribolet L, Cantacessi C, Periago MV, Valero MA, Valerio MA, Jariwala AR, Hotez P, Diemert D, Loukas A, Bethony J. Molecular mechanisms of hookworm disease: stealth, virulence, and vaccines. J Allergy Clin Immunol 2012; 130:13-21. [PMID: 22742835 DOI: 10.1016/j.jaci.2012.05.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 05/24/2012] [Accepted: 05/25/2012] [Indexed: 12/31/2022]
Abstract
Hookworms produce a vast repertoire of structurally and functionally diverse molecules that mediate their long-term survival and pathogenesis within a human host. Many of these molecules are secreted by the parasite, after which they interact with critical components of host biology, including processes that are key to host survival. The most important of these interactions is the hookworm's interruption of nutrient acquisition by the host through its ingestion and digestion of host blood. This results in iron deficiency and eventually the microcytic hypochromic anemia or iron deficiency anemia that is the clinical hallmark of hookworm infection. Other molecular mechanisms of hookworm infection cause a systematic suppression of the host immune response to both the parasite and to bystander antigens (eg, vaccines or allergens). This is achieved by a series of molecules that assist the parasite in the stealthy evasion of the host immune response. This review will summarize the current knowledge of the molecular mechanisms used by hookworms to survive for extended periods in the human host (up to 7 years or longer) and examine the pivotal contributions of these molecular mechanisms to chronic hookworm parasitism and host clinical outcomes.
Collapse
Affiliation(s)
- Mark S Pearson
- Center for Biodiscovery and Molecular Development of Therapeutics, James Cook University, Cairns, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
|
89
|
Lindfors K, Lähdeaho ML, Kalliokoski S, Kurppa K, Collin P, Mäki M, Kaukinen K. Future treatment strategies for celiac disease. Expert Opin Ther Targets 2012; 16:665-75. [PMID: 22620264 DOI: 10.1517/14728222.2012.688808] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Ingestion of dietary gluten in wheat, rye and barley by celiac patients leads to small-bowel mucosal villous atrophy, crypt hyperplasia and massive inflammation, often coupled with clinical symptoms and signs. Currently, the only effective treatment is a strict life-long gluten-free diet excluding all gluten-containing food products. In view of the extremely restricted nature of the diet, new treatment options would clearly be desirable. AREAS COVERED The improved understanding of celiac disease pathogenesis has enabled researchers to suggest alternative strategies to treat the disorder. This review covers current approaches toward developing an appropriate drug and discusses the possible problems associated with these approaches. EXPERT OPINION Phase II clinical trials are already ongoing to test the efficacy of novel alternative treatments for celiac disease. Before any of the candidates can enter Phase III trials, however, researchers must develop novel reliable non-invasive surrogate markers for intestinal injury and disease activity which also accurately reflect patient-related outcomes. Any novel medication for celiac disease should be as effective and safe as the gluten-free diet, and this constitutes a challenge for drug development. It is thus likely that the first medication entering the market will be supplementary to rather than substitute the gluten-free diet.
Collapse
Affiliation(s)
- Katri Lindfors
- University of Tampere and Tampere University Hospital, School of Medicine, Pediatric Research Center, Finn-Medi 3, FIN-33014, Tampere, Finland.
| | | | | | | | | | | | | |
Collapse
|
90
|
Wolff MJ, Broadhurst MJ, Loke P. Helminthic therapy: improving mucosal barrier function. Trends Parasitol 2012; 28:187-94. [PMID: 22464690 DOI: 10.1016/j.pt.2012.02.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 02/29/2012] [Accepted: 02/29/2012] [Indexed: 12/21/2022]
Abstract
The epidemiology of autoimmune diseases and helminth infections led to suggestions that helminths could improve inflammatory conditions, which was then tested using animal models. This has translated to clinical investigations aimed at the safe and controlled reintroduction of helminthic exposure to patients suffering from autoimmune diseases (so-called 'helminthic therapy') in an effort to mitigate the inflammatory response. In this review, we summarize the results of recent clinical trials of helminthic therapy, with particular attention to mechanisms of action. Whereas previous reviews have emphasized immune regulatory mechanisms activated by helminths, we propose that enhancement of mucosal barrier function may have an equally important role in improving conditions of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Martin J Wolff
- Division of Gastroenterology, Department of Medicine, New York University School of Medicine, New York, NY 10010, USA
| | | | | |
Collapse
|
91
|
Gaze S, McSorley HJ, Daveson J, Jones D, Bethony JM, Oliveira LM, Speare R, McCarthy JS, Engwerda CR, Croese J, Loukas A. Characterising the mucosal and systemic immune responses to experimental human hookworm infection. PLoS Pathog 2012; 8:e1002520. [PMID: 22346753 PMCID: PMC3276555 DOI: 10.1371/journal.ppat.1002520] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 12/21/2011] [Indexed: 11/18/2022] Open
Abstract
The mucosal cytokine response of healthy humans to parasitic helminths has never been reported. We investigated the systemic and mucosal cytokine responses to hookworm infection in experimentally infected, previously hookworm naive individuals from non-endemic areas. We collected both peripheral blood and duodenal biopsies to assess the systemic immune response, as well as the response at the site of adult worm establishment. Our results show that experimental hookworm infection leads to a strong systemic and mucosal Th2 (IL-4, IL-5, IL-9 and IL-13) and regulatory (IL-10 and TGF-β) response, with some evidence of a Th1 (IFN-γ and IL-2) response. Despite upregulation after patency of both IL-15 and ALDH1A2, a known Th17-inducing combination in inflammatory diseases, we saw no evidence of a Th17 (IL-17) response. Moreover, we observed strong suppression of mucosal IL-23 and upregulation of IL-22 during established hookworm infection, suggesting a potential mechanism by which Th17 responses are suppressed, and highlighting the potential that hookworms and their secreted proteins offer as therapeutics for human inflammatory diseases. Parasitic worms reside in the gastrointestinal tracts of billions of humans in developing countries. Despite the enormous disease burdens associated with these infections, very little is known about the immune response in the gut tissue of humans to these parasites. We conducted a clinical trial where we obtained gut biopsies from people experimentally infected with hookworms and present here the first report of the immune response by healthy human gut tissue to a parasitic worm. We show that hookworms suppress the production of pro-inflammatory molecules and promote the expression of anti-inflammatory and wound healing molecules in the gut, providing a potential mechanism by which parasitic worms reside for long periods in their human hosts and suppress inflammation associated with auto-immune diseases.
Collapse
Affiliation(s)
- Soraya Gaze
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
| | - Henry J. McSorley
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
| | - James Daveson
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Di Jones
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | | | | - Richard Speare
- Anton Breinl Centre, James Cook University, Townsville, Queensland, Australia
| | - James S. McCarthy
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | | - John Croese
- The Townsville Hospital and James Cook University, Townsville, Queensland, Australia
| | - Alex Loukas
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
- * E-mail:
| |
Collapse
|
92
|
Bancroft AJ, Hayes KS, Grencis RK. Life on the edge: the balance between macrofauna, microflora and host immunity. Trends Parasitol 2012; 28:93-8. [PMID: 22257556 DOI: 10.1016/j.pt.2011.12.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 12/16/2011] [Accepted: 12/19/2011] [Indexed: 12/26/2022]
Abstract
Mammals, microflora and gut-dwelling macrofauna have co-evolved over many millions of years until relatively recently when the geographical prevalence of macrofauna in humans has become restricted to the developing world. Immune homeostasis relies on a balance in the composition of intestinal microflora; long-lived macrofauna have also been shown to regulate immune function, and their absence in Western lifestyles is suggested to be a factor for the increasing frequency of allergy and autoimmunity. The intestinal nematode Trichuris muris was recently demonstrated to utilise microflora to initiate its life cycle. The interdependence on one another of all three factors is such that when the balance is perturbed it must be realigned or the consequences may be detrimental to the mammalian host.
Collapse
Affiliation(s)
- Allison J Bancroft
- Manchester Immunology Group, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | | | | |
Collapse
|