Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
J Clin Gastroenterol ; 58(2): 120-130, 2024 02 01.
Article in English | MEDLINE | ID: mdl-37682003

ABSTRACT

BACKGROUND: Hyperbaric oxygen therapy (HBOT) delivers 100% oxygen in a pressurized chamber, increasing tissue oxygen levels and regulating inflammatory pathways. Mounting evidence suggests that HBOT may be effective for inflammatory bowel disease. Our systematic review and meta-analysis aimed to quantify the efficacy and safety of HBOT in fistulizing Crohn's disease (CD). METHODS: A systematic review was conducted using the EMBASE, Web of Science, Pubmed, and Cochrane Library databases according to the "Preferred Reporting Items for Systematic Reviews and Meta-analyses" criteria. Study bias was assessed using the Cochrane Handbook guidelines. RESULTS: Sixteen studies with 164 patients were included in the analysis. For all fistula subtypes, the pooled overall clinical response was 87% (95% CI: 0.70-0.95, I2 = 0) and the pooled clinical remission was 59% (95% CI: 0.35-0.80, I2 = 0). The overall clinical response was 89%, 84%, and 29% for perianal, enterocutaneous, and rectovaginal fistulas, respectively. On meta-regression, hours in the chamber and the number of HBOT sessions were not found to correlate with clinical response. The pooled number of adverse events was low at 51.7 per 10,000 HBOT sessions for all fistula types (95% CI: 16.8-159.3, I2 = 0). The risk of bias was observed across all studies. CONCLUSION: HBOT is a safe and potentially effective treatment option for fistulizing CD. Randomized control trials are needed to substantiate the benefit of HBOT in fistulizing CD.


Subject(s)
Crohn Disease , Hyperbaric Oxygenation , Female , Humans , Crohn Disease/therapy , Fistula/therapy , Hyperbaric Oxygenation/adverse effects , Oxygen/therapeutic use , Treatment Outcome
2.
Sci Rep ; 9(1): 19127, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836772

ABSTRACT

The ability of helminth parasite infections to manipulate the immune system of their host towards T regulatory responses has been proposed to suppress the inflammatory response. The aim of this study was to investigate the protective and therapeutic effect of Syphacia obvelata in the treatment of experimental DSS -induced colitis. 50 male C57BL/6 mice were divided into 5 groups: healthy uninfected controls, DSS colitis, receiving only S. obv, preventive (S. obv + DSS) and therapeutic group (DSS + S.obv). Colitis intensity was investigated by measuring body weight changes, stool consistency/bleeding and colon length. To evaluate the immune responses induced by this nematode, TNF-α, IL-10, IL-17, IFN-γ and expressing of FoxP3+ T cells were measured in mesenteric lymph nodes and Peyer's patches cells. Mice in preventive and therapeutic groups treated with S. obv egg significantly ameliorated the severity of the DSS colitis, indicated by the reduced disease manifestations, improved histopathological scores correlated with the up regulation of Treg responses and down regulation of proinflammatory cytokines. S. obv can prevention and reverse on-going murine DSS colitis. The data suggest that induction of Tregs and change in cytokine profiles during helminthic therapies were responsible for reversed inflammatory events in IBD.


Subject(s)
Colitis/immunology , Colitis/parasitology , Oxyuroidea/physiology , Animals , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Immune System , Inflammation , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Peyer's Patches/immunology , T-Lymphocytes, Regulatory/parasitology , Tumor Necrosis Factor-alpha/metabolism
3.
J Immunol ; 202(8): 2473-2481, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30850474

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-ß and IL-10 in the gut. In the intestines, TGF-ß helps induce regulatory T cells. This study used Foxp3/IL-10 double reporter mice to investigate the effect of TGF-ß 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-ß 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-ß 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-ß with Smad2/3 phosphorylation. The TGF-ß-induced Tregs that express IL-10 blocked colitis when transferred into the Rag/CD25- CD4+ T cell transfer model of inflammatory bowel disease. TGF-ß 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-ß produced in response to the infection to induce the Tregs that prevent colitis.


Subject(s)
Colitis/immunology , Interleukin-10/immunology , Nematospiroides dubius/immunology , Smad7 Protein/immunology , Strongylida Infections/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Colitis/pathology , Colitis/prevention & control , Interleukin-10/genetics , Mice , Mice, Transgenic , Smad7 Protein/genetics , Strongylida Infections/genetics , Strongylida Infections/pathology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics
4.
Sci Rep ; 8(1): 15921, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30374177

ABSTRACT

Parasitic worms have a remarkable ability to modulate host immune responses through several mechanisms including excreted/secreted proteins (ESP), yet the exact nature of these proteins and their targets often remains elusive. Here, we performed mass spectrometry analyses of ESP (TsESP) from larval and adult stages of the pig whipworm Trichuris suis (Ts) and identified ~350 proteins. Transcriptomic analyses revealed large subsets of differentially expressed genes in the various life cycle stages of the parasite. Exposure of bone marrow-derived macrophages and dendritic cells to TsESP markedly diminished secretion of the pro-inflammatory cytokines TNFα and IL-12p70. Conversely, TsESP exposure strongly induced release of the anti-inflammatory cytokine IL-10, and also induced high levels of nitric oxide (NO) and upregulated arginase activity in macrophages. Interestingly, TsESP failed to directly induce CD4+ CD25+ FoxP3+ regulatory T cells (Treg cells), while OVA-pulsed TsESP-treated dendritic cells suppressed antigen-specific OT-II CD4+ T cell proliferation. Fractionation of TsESP identified a subset of proteins that promoted anti-inflammatory functions, an activity that was recapitulated using recombinant T. suis triosephosphate isomerase (TPI) and nucleoside diphosphate kinase (NDK). Our study helps illuminate the intricate balance that is characteristic of parasite-host interactions at the immunological interface, and further establishes the principle that specific parasite-derived proteins can modulate immune cell functions.


Subject(s)
Helminth Proteins/metabolism , Trichuris/metabolism , Animals , Arginase/metabolism , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Life Cycle Stages , Macrophages/cytology , Macrophages/metabolism , Nitric Oxide/metabolism , Swine/parasitology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Trichuris/growth & development
5.
J Immunol ; 197(7): 2948-57, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27559049

ABSTRACT

Helminthic infections modulate host immunity and may protect people in less-developed countries from developing immunological diseases. In a murine colitis model, the helminth Heligmosomoides polygyrus bakeri prevents colitis via induction of regulatory dendritic cells (DCs). The mechanism driving the development of these regulatory DCs is unexplored. There is decreased expression of the intracellular signaling pathway spleen tyrosine kinase (Syk) in intestinal DCs from H. polygyrus bakeri-infected mice. To explore the importance of this observation, it was shown that intestinal DCs from DC-specific Syk(-/-) mice were powerful inhibitors of murine colitis, suggesting that loss of Syk was sufficient to convert these cells into their regulatory phenotype. DCs sense gut flora and damaged epithelium via expression of C-type lectin receptors, many of which signal through the Syk signaling pathway. It was observed that gut DCs express mRNA encoding for C-type lectin (CLEC) 7A, CLEC9A, CLEC12A, and CLEC4N. H. polygyrus bakeri infection downmodulated CLEC mRNA expression in these cells. Focusing on CLEC7A, which encodes for the dectin-1 receptor, flow analysis showed that H. polygyrus bakeri decreases dectin-1 expression on the intestinal DC subsets that drive Th1/Th17 development. DCs become unresponsive to the dectin-1 agonist curdlan and fail to phosphorylate Syk after agonist stimulation. Soluble worm products can block CLEC7A and Syk mRNA expression in gut DCs from uninfected mice after a brief in vitro exposure. Thus, downmodulation of Syk expression and phosphorylation in intestinal DCs could be important mechanisms through which helminths induce regulatory DCs that limit colitis.


Subject(s)
Colitis/prevention & control , Dendritic Cells/immunology , Dendritic Cells/metabolism , Intestines/cytology , Signal Transduction , Syk Kinase/metabolism , Animals , Colitis/immunology , Colitis/parasitology , Disease Models, Animal , Intestines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Syk Kinase/deficiency , Syk Kinase/genetics
6.
Science ; 351(6279): 1329-33, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26847546

ABSTRACT

The intestinal epithelium forms an essential barrier between a host and its microbiota. Protozoa and helminths are members of the gut microbiota of mammals, including humans, yet the many ways that gut epithelial cells orchestrate responses to these eukaryotes remain unclear. Here we show that tuft cells, which are taste-chemosensory epithelial cells, accumulate during parasite colonization and infection. Disruption of chemosensory signaling through the loss of TRMP5 abrogates the expansion of tuft cells, goblet cells, eosinophils, and type 2 innate lymphoid cells during parasite colonization. Tuft cells are the primary source of the parasite-induced cytokine interleukin-25, which indirectly induces tuft cell expansion by promoting interleukin-13 production by innate lymphoid cells. Our results identify intestinal tuft cells as critical sentinels in the gut epithelium that promote type 2 immunity in response to intestinal parasites.


Subject(s)
Chemoreceptor Cells/immunology , Intestinal Diseases, Parasitic/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Microbiota/immunology , TRPM Cation Channels/immunology , Animals , Doublecortin-Like Kinases , Eosinophils/immunology , Goblet Cells/immunology , Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Immunity, Mucosal , Interleukin-13/immunology , Interleukin-17/immunology , Intestinal Diseases, Parasitic/parasitology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Serine-Threonine Kinases/immunology , Protozoan Infections/immunology , Protozoan Infections/parasitology , Signal Transduction , Taste , Transducin/genetics , Transducin/immunology , Tritrichomonas/immunology
7.
Clin Rev Allergy Immunol ; 49(2): 227-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25326880

ABSTRACT

Many immune-mediated diseases like inflammatory bowel disease, multiple sclerosis, type 1 diabetes, asthma, and food allergy appeared to have increased in frequency in developed countries in the latter part of the twentieth century. Reports from less developed countries suggest that the "epidemic" of immune-mediated diseases now is spreading into these regions as well. The "hygiene hypothesis" was developed to partly explain this phenomenon. It has been proposed that modern-day sanitary living has altered our exposure to organisms that provided protection from these diseases in the past. Alternations in the composition of our intestinal flora and fauna could play a role. Helminths are a group of worm-like parasitic organisms that have adapted to live in various regions of their hosts. Epidemiological and some clinical data suggest that these organisms can protect people from developing immune-mediated diseases. Animal experimentation has shown that helminths stimulate the production of regulatory cytokines, activate regulatory T cells, and induce regulatory dendritic cells and macrophages. This could be the mechanism by which they protect the host from these diseases. Early clinical studies also suggest that helminths may prove useful for treating immunological diseases. More sophisticated clinical studies are underway, testing live helminth agents as therapeutic agents. Also, a strong effort is ongoing to discover the agents produced by helminths that modulate host immune responses with an eye on developing new, highly effective immune modulatory therapeutic agent.


Subject(s)
Helminths/immunology , Immune System Diseases/therapy , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Therapy with Helminths , Animals , Antigens, Helminth/immunology , Clinical Trials as Topic , Cytokines/metabolism , Humans , Hygiene Hypothesis , Immune System Diseases/immunology , Immune System Diseases/parasitology , Immunomodulation , Macrophages/parasitology , T-Lymphocytes, Regulatory/parasitology
8.
J Immunol ; 193(7): 3239-47, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25240019

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-ß, 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.


Subject(s)
Autoimmune Diseases/immunology , Dendritic Cells/immunology , Helminthiasis/immunology , Helminths/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/immunology , Humans , Immunity, Innate , Mice
9.
Clin Gastroenterol Hepatol ; 12(5): 749-55, 2014 May.
Article in English | MEDLINE | ID: mdl-24632348

ABSTRACT

Cells can innately recognize generic products of viruses, bacteria, fungi, or injured tissue by engagement of pattern recognition receptors. Innate immune cells rapidly respond to this engagement to control commensals, thwart pathogens, and/or prompt repair. Insufficient or excessive activation of the innate immune response results in disease. This review focuses on pattern recognition receptors and cells of the innate immune system that are important for intestinal function. Our improving knowledge pertaining to this important aspect of our immune response is opening potential important new therapeutic opportunities for the treatment of disease.


Subject(s)
Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/pathology , Immunity, Innate , Cytokines/metabolism , Humans , Leukocytes/immunology , Receptors, Pattern Recognition/metabolism
10.
J Immunol ; 191(4): 1927-34, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851695

ABSTRACT

Helminthic infections protect mice from colitis in murine models of inflammatory bowel disease and also may protect people. Helminths like Heligmosomoides polygyrus bakeri can induce regulatory T cells (Treg). Experiments explored whether H. polygyrus bakeri infection could protect mice from colitis through activation of colonic Treg and examined mechanisms of action. We showed that H. polygyrus bakeri infection increased the number of T cells expressing Foxp3 in the colon. More importantly, Foxp3(+)/IL-10(-) and Foxp3(+)/IL-10(+) T cell subsets isolated from the colon of H. polygyrus bakeri-infected mice prevented colitis when adoptively transferred into a murine model of inflammatory bowel disease, whereas Treg from uninfected mice could not provide protection. Only the transferred colonic Foxp3(+)/IL-10(-) T cells from H. polygyrus bakeri-infected mice readily accumulated in the colon and mesenteric lymph nodes of recipient mice, and they reconstituted the Foxp3(+)/IL-10(-) and Foxp3(+)/IL-10(+) T cell subsets. However, transferred Foxp3(+)/IL-10(+) T cells disappeared. IL-10 expression by Foxp3(+) T cells was necessary for colitis prevention. Thus, H. polygyrus bakeri infection activates colonic Foxp3(+) T cells, making them highly regulatory. The Foxp3(+) T cells that fail to express IL-10 may be critical for populating the colon with the Foxp3(+)/IL-10(+) T cells, which are required to control colitis.


Subject(s)
Colitis/prevention & control , Colon/immunology , Intestinal Diseases, Parasitic/immunology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Therapy with Helminths , Animals , Colitis/immunology , Colitis/parasitology , Colon/parasitology , Cytokines/biosynthesis , Cytokines/metabolism , DNA-Binding Proteins/deficiency , Disease Models, Animal , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/deficiency , Genes, Reporter , Graft Survival , Helminthiasis, Animal/immunology , Immunotherapy, Adoptive , Inflammatory Bowel Diseases/therapy , Interleukin-10/analysis , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mesentery , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/chemistry , T-Lymphocytes, Regulatory/transplantation
11.
Inflamm Bowel Dis ; 19(3): 672-3, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23429450
12.
Int J Parasitol ; 43(3-4): 245-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23178819

ABSTRACT

Modern hygienic lifestyles are associated with the emergence of inflammatory bowel disease (IBD) which now afflicts millions of people in highly-developed countries. Meticulous hygiene interrupts conduits of transmission required for ubiquitous exposure to parasitic worms (helminths). We proposed that loss of exposure to helminths permits development of IBD. Early clinical trials suggested that exposure to helminths such as Trichuris suis or Necator americanus can improve IBD. Over the last several years, processes to "medicinalize"T. suis have been developed and use of this helminth is now being studied in large multi-center clinical trials. Concurrently, we and others have identified some of the immune regulatory mechanisms elicited by helminth exposure that suppress inappropriate intestinal inflammation. These efforts could soon result in new therapies for patients with IBD.


Subject(s)
Helminths/immunology , Inflammatory Bowel Diseases/therapy , Therapy with Helminths , Animals , Helminths/physiology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/parasitology
15.
Curr Opin Gastroenterol ; 28(6): 551-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23079675

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.


Subject(s)
Helminthiasis/immunology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Animals , Disease Models, Animal , Helminthiasis/epidemiology , Helminths/immunology , Humans , Hygiene Hypothesis , Immunity, Mucosal , Inflammation/parasitology , Inflammatory Bowel Diseases/epidemiology , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/parasitology , Socioeconomic Factors , Therapy with Helminths
16.
J Immunol ; 189(5): 2512-20, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22844110

ABSTRACT

Immunological diseases such as inflammatory bowel disease (IBD) are infrequent in less developed countries, possibly because helminths provide protection by modulating host immunity. In IBD murine models, the helminth Heligmosomoides polygyrus bakeri prevents colitis. It was determined whether H. polygyrus bakeri mediated IBD protection by altering dendritic cell (DC) function. We used a Rag IBD model where animals were reconstituted with IL10⁻/⁻ T cells, making them susceptible to IBD and with OVA Ag-responsive OT2 T cells, allowing study of a gut antigenic response. Intestinal DC from H. polygyrus bakeri-infected Rag mice added to lamina propria mononuclear cells (LPMC) isolated from colitic animals blocked OVA IFN-γ/IL-17 responses in vitro through direct contact with the inflammatory LPMC. DC from uninfected Rag mice displayed no regulatory activity. Transfer of DC from H. polygyrus bakeri-infected mice into Rag mice reconstituted with IL10⁻/⁻ T cells protected animals from IBD, and LPMC from these mice lost OVA responsiveness. After DC transfer, OT2 T cells populated the intestines normally. However, the OT2 T cells were rendered Ag nonresponsive through regulatory action of LPMC non-T cells. The process of regulation appeared to be regulatory T cell independent. Thus, H. polygyrus bakeri modulates intestinal DC function, rendering them tolerogenic. This appears to be an important mechanism through which H. polygyrus bakeri suppresses colitis. IFN-γ and IL-17 are colitogenic. The capacity of these DC to block a gut Ag-specific IFN-γ/IL-17 T cell response also is significant.


Subject(s)
Colitis/immunology , Dendritic Cells/immunology , Immune Tolerance , Inflammatory Bowel Diseases/immunology , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , Cells, Cultured , Colitis/parasitology , Colitis/prevention & control , Dendritic Cells/parasitology , Dendritic Cells/pathology , Disease Models, Animal , Enterocolitis/immunology , Enterocolitis/parasitology , Enterocolitis/prevention & control , Epitopes, T-Lymphocyte/immunology , Inflammatory Bowel Diseases/parasitology , Inflammatory Bowel Diseases/prevention & control , Interleukin-10/administration & dosage , Interleukin-10/deficiency , Interleukin-10/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nematospiroides dubius/immunology , Strongylida Infections/pathology , Strongylida Infections/prevention & control , T-Lymphocyte Subsets/parasitology , T-Lymphocyte Subsets/pathology
17.
Ann N Y Acad Sci ; 1247: 83-96, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22239614

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-ß 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.


Subject(s)
Helminths/pathogenicity , Host-Parasite Interactions/immunology , Immune System Diseases/prevention & control , Animals , Helminths/immunology , Humans , Immune System Diseases/immunology , Immune System Diseases/parasitology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/parasitology , Inflammatory Bowel Diseases/prevention & control , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-7/immunology , Interleukin-7/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/parasitology , Multiple Sclerosis/prevention & control
18.
Allergy Asthma Proc ; 33(6): 519-24, 2012.
Article in English | MEDLINE | ID: mdl-23394511

ABSTRACT

Diagnosis of eosinophilic esophagitis (EoE) and determination of response to therapy is based on histological assessment of the esophagus, which requires upper endoscopy. In children, in whom a dietary approach is commonly used, multiple endoscopies are needed, because foods are eliminated and then gradually reintroduced. Ideally, noninvasive methods could supplement or replace upper endoscopy to facilitate management. Fractionated exhaled nitric oxide (FeNO) has been proposed as a useful measure for monitoring disease activity in studies of patients with eosinophil-predominant asthma and in other atopic disorders. Thus, we evaluated whether FeNO levels could be a useful biomarker to assess the response to therapy in EoE patients. This study was designed to determine whether there is a change in FeNO levels during treatment with topical corticosteroids and whether changes correlated with clinical response. This was a prospective, multicenter study that enrolled nonasthmatic patients with established EoE. FeNO levels and symptom scores were measured at baseline, biweekly during 6-week swallowed fluticasone treatment, and 4 weeks posttreatment. Twelve patients completed the trial. We found a statistically significant difference between median pre- and posttreatment FeNO levels [20.3 ppb (16.0 -29.0 ppb) vs 17.6 ppb (11.7 -27.3 ppb), [corrected] p=0.009]. However, neither the pretreatment FeNO level, a change of FeNO level after 2 weeks of treatment, nor the FeNO level at the end of treatment confidently predicted a clinical or histological response. Although our findings suggest nitric oxide possibly has a physiological role in EoE, our observations do not support a role of FeNo determination for management of EoE.


Subject(s)
Eosinophilic Esophagitis/drug therapy , Eosinophilic Esophagitis/metabolism , Nitric Oxide/metabolism , Adolescent , Adult , Aged , Biomarkers/chemistry , Biomarkers/metabolism , Child , Eosinophilic Esophagitis/diagnosis , Exhalation , Female , Humans , Male , Middle Aged , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/chemistry , Pilot Projects , Prospective Studies , Young Adult
19.
Inflamm Bowel Dis ; 16(11): 1841-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20848461

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.


Subject(s)
Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Nematospiroides dubius/immunology , Animals , Disease Models, Animal , Genes, rRNA , Ileum/immunology , Interleukin-10/genetics , Lactobacillaceae/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
20.
J Immunol ; 185(6): 3184-9, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20702728

ABSTRACT

Less developed countries have a low incidence of immunological diseases like inflammatory bowel disease (IBD), perhaps prevented by the high prevalence of helminth infections in their populations. In the Rag IL-10(-/-) T cell transfer model of colitis, Heligmosomoides polygyrus, an intestinal helminth, prevents and reverses intestinal inflammation. This model of colitis was used to explore the importance of innate immunity in H. polygyrus protection from IBD. Rag mice briefly exposed to H. polygyrus before reconstitution with IL-10(-/-) colitogenic T cells are protected from colitis. Exposure to H. polygyrus before introduction of IL-10(-/-) and OT2 T cells reduced the capacity of the intestinal mucosa to make IFN-gamma and IL-17 after either anti-CD3 mAb or OVA stimulation. This depressed cytokine response was evident even in the absence of colitis, suggesting that the downmodulation in proinflammatory cytokine secretion was not just secondary to improvement in intestinal inflammation. Following H. polygyrus infection, dendritic cells (DCs) from the lamina propria of Rag mice displayed decreased expression of CD80 and CD86, and heightened expression of plasmacytoid dendritic cell Ag-1 and CD40. They were also less responsive to lamina proprias, producing less IL-12p40 and IL-10. Also diminished was their capacity to present OVA to OT2 T cells. These experiments infer that H. polygyrus does not require direct interactions with T or B cells to render animals resistant to colitis. DCs have an important role in driving both murine and human IBD. Data suggest that phenotypic alternations in mucosal DC function are part of the regulatory process.


Subject(s)
Colitis/immunology , Colitis/prevention & control , Immunity, Innate , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Animals , Cells, Cultured , Colitis/parasitology , Dendritic Cells/immunology , Dendritic Cells/parasitology , Disease Models, Animal , Immunity, Innate/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/parasitology , Inflammatory Bowel Diseases/prevention & control , Interleukin-10/deficiency , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucous Membrane/immunology , Mucous Membrane/parasitology , Mucous Membrane/pathology , Ovalbumin/immunology , Strongylida Infections/genetics , Strongylida Infections/parasitology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/parasitology , T-Lymphocyte Subsets/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...