Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Nat Commun ; 14(1): 4503, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37495576

ABSTRACT

The reduced prevalence of insulin resistance and type 2 diabetes in countries with endemic parasitic worm infections suggests a protective role for worms against metabolic disorders, however clinical evidence has been non-existent. This 2-year randomised, double-blinded clinical trial in Australia of hookworm infection in 40 male and female adults at risk of type 2 diabetes assessed the safety and potential metabolic benefits of treatment with either 20 (n = 14) or 40 (n = 13) Necator americanus larvae (L3) or Placebo (n = 13) (Registration ACTRN12617000818336). Primary outcome was safety defined by adverse events and completion rate. Homoeostatic model assessment of insulin resistance, fasting blood glucose and body mass were key secondary outcomes. Adverse events were more frequent in hookworm-treated participants, where 44% experienced expected gastrointestinal symptoms, but completion rates were comparable to Placebo. Fasting glucose and insulin resistance were lowered in both hookworm-treated groups at 1 year, and body mass was reduced after L3-20 treatment at 2 years. This study suggests hookworm infection is safe in people at risk of type 2 diabetes and associated with improved insulin resistance, warranting further exploration of the benefits of hookworms on metabolic health.


Subject(s)
Diabetes Mellitus, Type 2 , Hookworm Infections , Insulin Resistance , Animals , Male , Female , Hookworm Infections/complications , Hookworm Infections/drug therapy , Hookworm Infections/epidemiology , Necator americanus , Fasting
2.
Proc Natl Acad Sci U S A ; 119(36): e2202795119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037362

ABSTRACT

Parasitic helminth infections, while a major cause of neglected tropical disease burden, negatively correlate with the incidence of immune-mediated inflammatory diseases such as inflammatory bowel diseases (IBD). To evade expulsion, helminths have developed sophisticated mechanisms to regulate their host's immune responses. Controlled experimental human helminth infections have been assessed clinically for treating inflammatory conditions; however, such a radical therapeutic modality has challenges. An alternative approach is to harness the immunomodulatory properties within the worm's excretory-secretory (ES) complement, its secretome. Here, we report a biologics discovery and validation pipeline to generate and screen in vivo a recombinant cell-free secretome library of helminth-derived immunomodulatory proteins. We successfully expressed 78 recombinant ES proteins from gastrointestinal hookworms and screened the crude in vitro translation reactions for anti-IBD properties in a mouse model of acute colitis. After statistical filtering and ranking, 20 proteins conferred significant protection against various parameters of colitis. Lead candidates from distinct protein families, including annexins, transthyretins, nematode-specific retinol-binding proteins, and SCP/TAPS were identified. Representative proteins were produced in mammalian cells and further validated, including ex vivo suppression of inflammatory cytokine secretion by T cells from IBD patient colon biopsies. Proteins identified herein offer promise as novel, safe, and mechanistically differentiated biologics for treating the globally increasing burden of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents , Biological Products , Colitis , Helminth Proteins , Inflammatory Bowel Diseases , Animals , Anti-Inflammatory Agents/pharmacology , Biological Products/pharmacology , Colitis/drug therapy , Helminth Proteins/genetics , Helminth Proteins/pharmacology , Helminths , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/parasitology , Mice
3.
8.
Clin Transl Gastroenterol ; 11(12): e00274, 2020 12.
Article in English | MEDLINE | ID: mdl-33512796

ABSTRACT

INTRODUCTION: Celiac disease is an autoimmune disorder where intestinal immunopathology arises after gluten consumption. Previous studies suggested that hookworm infection restores gluten tolerance; however, these studies were small (n = 12) and not placebo controlled. METHODS: We undertook a randomized, placebo-controlled trial of hookworm infection in 54 people with celiac disease. The 94-week study involved treatment with either 20 or 40 Necator americanus third-stage larvae (L3-20 or L3-40) or placebo, followed by escalating gluten consumption (50 mg/d for 12 weeks, 1 g intermittent twice weekly for 12 weeks, 2 g/d sustained for 6 weeks, liberal diet for 1 year). RESULTS: Successful study completion rates at week 42 (primary outcome) were similar in each group (placebo: 57%, L3-20: 37%, and L3-40: 44%; P = 0.61), however gluten-related adverse events were significantly reduced in hookworm-treated participants: Median (range) adverse events/participant were as follows: placebo, 4 (1-9); L3-20, 1 (0-9); and L3-40, 0 (0-3) (P = 0.019). Duodenal villous height:crypt depth deteriorated similarly compared with their enrolment values in each group (mean change [95% confidence interval]: placebo, -0.6 [-1.3 to 0.2]; L3-20, -0.5 [-0.8 to 0.2]; and L3-40, -1.1 [-1.8 to 0.4]; P = 0.12). A retrospective analysis revealed that 9 of the 40 L3-treated participants failed to establish hookworm infections. Although week 42 completion rates were similar in hookworm-positive vs hookworm-negative participants (48% vs 44%, P = 0.43), quality of life symptom scores were lower in hookworm-positive participants after intermittent gluten challenge (mean [95% confidence interval]: 38.9 [33.9-44] vs 45.9 [39.2-52.6]). DISCUSSION: Hookworm infection does not restore tolerance to sustained moderate consumption of gluten (2 g/d) but was associated with improved symptom scores after intermittent consumption of lower, intermittent gluten doses.


Subject(s)
Celiac Disease/therapy , Glutens/immunology , Larva/metabolism , Necator americanus/metabolism , Therapy with Helminths/methods , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Celiac Disease/immunology , Double-Blind Method , Female , Glutens/administration & dosage , Glutens/metabolism , Humans , Male , Middle Aged , Quality of Life , Therapy with Helminths/adverse effects , Treatment Outcome , Young Adult
9.
BMC Endocr Disord ; 19(1): 136, 2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31829172

ABSTRACT

BACKGROUND: Abdominal obesity and presence of the metabolic syndrome (MetS) significantly increase the risk of developing diseases such as Type 2 diabetes mellitus (T2DM) with escalating emergence of MetS and T2DM constituting a significant public health crisis worldwide. Lower prevalence of inflammatory and metabolic diseases such as T2DM in countries with higher incidences of helminth infections suggested a potential role for these parasites in the prevention and management of certain diseases. Recent studies confirmed the potential protective nature of helminth infection against MetS and T2DM via immunomodulation or, potentially, alteration of the intestinal microbiota. This Phase 1b safety and tolerability trial aims to assess the effect of inoculation with helminths on physical and metabolic parameters, immune responses, and the microbiome in otherwise healthy women and men. METHODS: Participants eligible for inclusion are adults aged 18-50 with central obesity and a minimum of one additional feature of MetS recruited from the local community with a recruitment target of 54. In a randomised, double-blind, placebo-controlled design, three groups will receive either 20 or 40 stage three larvae of the human hookworm Necator americanus or a placebo. Eligible participants will provide blood and faecal samples at their baseline and 6-monthly assessment visits for a total of 24 months with an optional extension to 36 months. During each scheduled visit, participants will also undergo a full physical examination and complete diet (PREDIMED), physical activity, and patient health (PHQ-9) questionnaires. Outcome measurements include tolerability and safety of infection with Necator americanus, changes in metabolic and immunological parameters, and changes in the composition of the faecal microbiome. DISCUSSION: Rising cost of healthcare associated with obesity-induced metabolic diseases urgently calls for new approaches in disease prevention. Findings from this trial will provide valuable information regarding the potential mechanisms by which hookworms, potentially via alterations in the microbiota, may positively influence metabolic health. TRIAL REGISTRATION: The protocol was registered on ANZCTR.org.au on 05 June 2017 with identifier ACTRN12617000818336. Alternatively, a Google search using the above trial registration number will yield a direct link to the trial protocol within the ANZCTR website.


Subject(s)
Metabolic Syndrome/therapy , Necatoriasis , Obesity/complications , Therapy with Helminths/methods , Adolescent , Adult , Animals , Diabetes Mellitus, Type 2/prevention & control , Double-Blind Method , Feces/microbiology , Female , Gastrointestinal Microbiome/physiology , Humans , Immunomodulation , Larva , Male , Metabolic Syndrome/immunology , Metabolic Syndrome/microbiology , Middle Aged , Necator americanus , Necatoriasis/immunology , Necatoriasis/microbiology , Placebos , Queensland , Therapy with Helminths/adverse effects , Treatment Outcome
10.
J Clin Gastroenterol ; 53(3): e113-e116, 2019 03.
Article in English | MEDLINE | ID: mdl-29570172

ABSTRACT

GOALS: To provide preliminary evidence that sessile serrated adenomas (SSA) are low-risk polyps in young patients. BACKGROUND: SSAs are the dominant polyp of the serrated neoplasia pathway and as such are the precursor of up to 20% of colorectal carcinomas (CRC). Up to 90% of these cancers are expected to harbor a BRAF mutation. SSAs are being diagnosed with increasing frequency in young patients, placing a significant burden on colonoscopic services. Evidence to direct the surveillance intervals for these young patients is not available. STUDY: We utilized 2 patient cohorts comprising (1) a consecutive series of patients who underwent outpatient colonoscopy through a tertiary hospital and (2) a consecutive series of resection specimens for CRC processed through a gastrointestinal pathology service. The prevalence of SSAs by age was determined in the patients undergoing colonoscopy and compared with the ages of patients with BRAF mutated CRC in the pathology series. RESULTS: The prevalence of SSAs was similar irrespective of age. By comparison, BRAF mutated CRCs were very rare (3.8% of cases) in patients younger than 50 years of age and uncommon (9.3% of cases) in patients younger than 60 years of age, but increased to 39.8% in patients older than 80 years of age. CONCLUSIONS: These results suggest that SSAs develop at a young age, but have a prolonged dwell time and are unlikely to develop into cancer in patients younger than 60 years of age. These findings highlight the need for further targeted research to determine the most appropriate surveillance intervals for young patients with sporadic SSAs.


Subject(s)
Adenoma/pathology , Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Proto-Oncogene Proteins B-raf/genetics , Adenoma/epidemiology , Adenoma/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Cohort Studies , Colonic Polyps/epidemiology , Colonic Polyps/genetics , Colonoscopy , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Disease Progression , Female , Humans , Male , Middle Aged , Mutation , Prevalence , Retrospective Studies , Time Factors
11.
Cell Rep ; 23(4): 1085-1098, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29694887

ABSTRACT

Inflammasomes promote immunity to microbial pathogens by regulating the function of IL-1-family cytokines such as IL-18 and IL-1ß. However, the roles for inflammasomes during parasitic helminth infections remain unclear. We demonstrate that mice and humans infected with gastrointestinal nematodes display increased IL-18 secretion, which in Trichuris-infected or worm antigen-treated mice and in macrophages co-cultured with Trichuris antigens or exosome-like vesicles was dependent on the NLRP3 inflammasome. NLRP3-deficient mice displayed reduced pro-inflammatory type 1 cytokine responses and augmented protective type 2 immunity, which was reversed by IL-18 administration. NLRP3-dependent suppression of immunity partially required CD4+ cells but was apparent even in Rag1-/- mice that lack adaptive immune cells, suggesting that NLRP3 influences both innate and adaptive immunity. These data highlight a role for NLRP3 in limiting protective immunity to helminths, suggesting that targeting the NLRP3 inflammasome may be an approach for limiting the disease burden associated with helminth infections.


Subject(s)
Adaptive Immunity , Antigens, Helminth/immunology , Immunity, Innate , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Trichuriasis/immunology , Trichuris/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Female , Humans , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Trichuriasis/genetics , Trichuriasis/pathology
12.
Clin Transl Immunology ; 6(10): e157, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29114386

ABSTRACT

Gastrointestinal parasites, hookworms in particular, have evolved to cause minimal harm to their hosts when present in small numbers, allowing them to establish chronic infections for decades. They do so by creating an immunoregulatory environment that promotes their own survival, but paradoxically also benefits the host by protecting against the onset of many inflammatory diseases. To harness the therapeutic value of hookworms without using live parasites, we have examined the protective properties of the recombinant protein anti-inflammatory protein (AIP)-1, secreted in abundance by hookworms within the intestinal mucosa, in experimental colitis. Colitic inflammation assessed by weight loss, colon atrophy, oedema, ulceration and necrosis, as well as abdominal adhesion was significantly suppressed in mice treated with a single intraperitoneal dose of AIP-1 at 1 mg kg-1. Local infiltration of inflammatory cells was also significantly reduced, with minimal goblet cell loss and preserved mucosal architecture. Treatment with AIP-1 promoted the production of colon interleukin (IL)-10, transforming growth factor (TGF)-ß and thymic stromal lymphopoietin (TSLP), resulting in the suppression of tumour necrosis factor (TNF)-α, IL-13 and IL-17 A cytokines and granulocyte macrophage colony-stimulating factor (GM-CSF), CX motif chemokine (CXCL)-11 and cyclooxygenase synthase (COX)-2 mRNA transcripts. AIP-1 promoted the accumulation of regulatory T cells in the colon likely allowing rapid healing of the colon mucosa. Hookworm recombinant AIP-1 is a novel therapeutic candidate for the treatment of inflammatory bowel diseases that can be explored for the prevention of acute inflammatory relapses, an important cause of colorectal cancer.

13.
J Dig Dis ; 18(3): 160-168, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28188978

ABSTRACT

OBJECTIVE: Aspiration risk, especially with propofol sedation, remains a concern after split-dose bowel preparation of up to 1 L polyethylene glycol for the procedure. We aimed to identify the ideal timing of bowel preparation to achieve optimal colon cleansing with no increased risk of aspiration. METHODS: A total of 892 consecutive patients undergoing simultaneous esophagogastroduodenoscopy (EGD) and colonoscopy were prospectively recruited. Residual gastric volume (RGV) and pH of gastric contents were measured at EGD, and patients' characteristics, runway time (duration between completion of the final liter of bowel preparation and colonoscopy commencement), and cleansing quality were recorded. RESULTS: A shorter runway time resulted in better colon cleansing (r = -0.124, P < 0.001). No correlation between runway time and RGV or pH was found (r = -0.017, P = 0.62 and r = -0.030, P = 0.47, respectively). RGV and pH did not differ significantly with runway time of 4 or 5 h. RGV with runway time ≤3 h was 35.9 ± 11.8 mL and 17.4 ± 0.6 mL after runway time >3 h (P < 0.001). No aspiration pneumonia occurred. The only factors independently related to higher RGV were younger age and male sex. CONCLUSIONS: The consumption of bowel preparation agent within 3-4 h before propofol sedation resulted in a similar RGV and pH as those achieved by more prolonged fasting, with no increased risk of aspiration even in patients perceived to be at high risk.


Subject(s)
Cathartics/administration & dosage , Colonoscopy/methods , Hypnotics and Sedatives , Polyethylene Glycols/administration & dosage , Propofol , Adolescent , Adult , Aged , Aged, 80 and over , Cathartics/adverse effects , Conscious Sedation/methods , Drug Administration Schedule , Endoscopy, Digestive System/methods , Female , Gastric Acidity Determination , Gastrointestinal Contents , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Pneumonia, Aspiration/etiology , Pneumonia, Aspiration/prevention & control , Polyethylene Glycols/adverse effects , Prospective Studies , Young Adult
14.
Intern Med J ; 47(3): 318-323, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27860102

ABSTRACT

BACKGROUND: Sessile serrated adenomas (SSA) are the polyp precursor of 15-20% of colorectal carcinomas. There is debate about their prevalence and increasing discussion about the need for a serrated polyp detection rate as a quality indicator for colonoscopy. AIMS: To assess the prevalence of SSA at an outpatient gastroenterology service. METHODS: This is a retrospective study of an unselected consecutive series of patients who had an outpatient colonoscopy between April 2013 and May 2014. The colonoscopy reports were reviewed to identify age, gender, indication for procedure, completion, withdrawal time, adequacy of bowel preparation, number, size and location of polyps. The pathology of all polyps was centrally reviewed by a gastrointestinal pathologist. RESULTS: A total of 707 patients underwent colonoscopy within the study period. The mean age of the cohort was 58 years, and 50.6% were female. Polyp(s) were identified in 66.5% of patients. The SSA detection rate was 20.1%, and the adenoma detection rate was 48.0%. SSA detection was associated with longer withdrawal times. Conventional adenoma detection was associated with older age, male gender, longer withdrawal time and a positive faecal occult blood test result. CONCLUSION: SSA are highly prevalent in an unselected series of patients attending a gastroenterology outpatient department. Identifying and removing these polyps may help prevent interval colorectal carcinoma. This result may serve as a benchmark for a high-quality colonoscopy service.


Subject(s)
Adenoma/epidemiology , Colonic Polyps/epidemiology , Colonoscopy , Colorectal Neoplasms/epidemiology , Early Detection of Cancer , Adenoma/diagnosis , Adenoma/pathology , Aged , Australia/epidemiology , Benchmarking , Colonic Polyps/diagnosis , Colonic Polyps/pathology , Colonoscopy/economics , Colonoscopy/statistics & numerical data , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/prevention & control , Cost-Benefit Analysis , Early Detection of Cancer/economics , Female , Humans , Male , Middle Aged , Outpatients , Prevalence , Retrospective Studies
15.
Nat Rev Dis Primers ; 2: 16088, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27929101

ABSTRACT

Hookworms are soil-transmitted nematode parasites that can reside for many years in the small intestine of their human hosts; Necator americanus is the predominant infecting species. Adult worms feed on the blood of a host and can cause iron deficiency anaemia, especially in high-risk populations (children and women of childbearing age). Almost 500 million people in developing tropical countries are infected, and simulation models estimate that hookworm infection is responsible for >4 million disability-adjusted life years lost annually. Humans mount an immune response to hookworms, but it is mostly unsuccessful at removing adult worms from the bowel. Accordingly, the host switches to an immune-tolerant state that enables hookworms to reside in the gut for many years. Although anthelmintic drugs are available and widely used, their efficacy varies and the drugs do not prevent reinfection. Thus, other control strategies aimed at improving water quality, sanitation and hygiene are needed. In addition, efforts are underway to develop a human hookworm vaccine through public-private partnerships. However, hookworms could also be a resource; as hookworms have the capability to regulate the host's inflammation, researchers are experimentally infecting patients to treat some inflammatory diseases as an approach to discover new anti-inflammatory molecules. This area of endeavour might well yield new biotherapeutics for autoimmune and allergic diseases.


Subject(s)
Hookworm Infections/complications , Hookworm Infections/physiopathology , Albendazole/pharmacology , Albendazole/therapeutic use , Ancylostomatoidea/immunology , Ancylostomatoidea/pathogenicity , Anemia/complications , Anemia/etiology , Animals , Anthelmintics/pharmacology , Anthelmintics/therapeutic use , Factor VIIa/adverse effects , Factor XIa/adverse effects , Factor Xa/adverse effects , Feces/parasitology , Hemorrhage/etiology , Hemorrhage/parasitology , Hookworm Infections/epidemiology , Humans , Intestine, Small/parasitology , Intestine, Small/physiopathology , Larva Migrans/etiology , Mebendazole/pharmacology , Mebendazole/therapeutic use , Necator americanus/immunology , Necator americanus/pathogenicity , Prevalence , Risk Factors , Soil/parasitology
16.
Sci Rep ; 6: 36797, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27827438

ABSTRACT

A reduced diversity of the gastrointestinal commensal microbiota is associated with the development of several inflammatory diseases. Recent reports in humans and animal models have demonstrated the beneficial therapeutic effects of infections by parasitic worms (helminths) in some inflammatory disorders, such as inflammatory bowel disease (IBD) and coeliac disease (CeD). Interestingly, these studies have described how helminths may alter the intestinal microbiota, potentially representing a mechanism by which they regulate inflammation. However, for practical reasons, these reports have primarily analysed the faecal microbiota. In the present investigation, we have assessed, for the first time, the changes in the microbiota at the site of infection by a parasitic helminth (hookworm) and gluten-dependent inflammation in humans with CeD using biopsy tissue from the duodenum. Hookworm infection and gluten exposure were associated with an increased abundance of species within the Bacteroides phylum, as well as increases in the richness and diversity of the tissue-resident microbiota within the intestine, results that are consistent with previous reports using other helminth species in humans and animal models. Hence, this may represent a mechanism by which parasitic helminths may restore intestinal immune homeostasis and exert a therapeutic benefit in CeD, and potentially other inflammatory disorders.


Subject(s)
Ancylostomatoidea/physiology , Bacteria/classification , Celiac Disease/microbiology , Duodenum/microbiology , Ancylostomatoidea/immunology , Animals , Bacteria/genetics , Bacteria/isolation & purification , Celiac Disease/immunology , Celiac Disease/parasitology , Duodenum/immunology , Duodenum/parasitology , Feces/microbiology , Humans , Microbiota , Sequence Analysis, DNA
17.
Sci Transl Med ; 8(362): 362ra143, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27797959

ABSTRACT

In the developed world, declining prevalence of some parasitic infections correlates with increased incidence of allergic and autoimmune disorders. Moreover, experimental human infection with some parasitic worms confers protection against inflammatory diseases in phase 2 clinical trials. Parasitic worms manipulate the immune system by secreting immunoregulatory molecules that offer promise as a novel therapeutic modality for inflammatory diseases. We identify a protein secreted by hookworms, anti-inflammatory protein-2 (AIP-2), that suppressed airway inflammation in a mouse model of asthma, reduced expression of costimulatory markers on human dendritic cells (DCs), and suppressed proliferation ex vivo of T cells from human subjects with house dust mite allergy. In mice, AIP-2 was primarily captured by mesenteric CD103+ DCs and suppression of airway inflammation was dependent on both DCs and Foxp3+ regulatory T cells (Tregs) that originated in the mesenteric lymph nodes (MLNs) and accumulated in distant mucosal sites. Transplantation of MLNs from AIP-2-treated mice into naïve hosts revealed a lymphoid tissue conditioning that promoted Treg induction and long-term maintenance. Our findings indicate that recombinant AIP-2 could serve as a novel curative therapeutic for allergic asthma and potentially other inflammatory diseases.


Subject(s)
Asthma/blood , Helminth Proteins/pharmacology , Hypersensitivity/therapy , Recombinant Proteins/pharmacology , T-Lymphocytes, Regulatory/immunology , Adult , Ancylostomatoidea , Animals , Antigens, CD/metabolism , Asthma/immunology , Cell Proliferation , Cytokines/immunology , Dendritic Cells/immunology , Disease Models, Animal , Female , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Inflammation , Integrin alpha Chains/metabolism , Male , Mice , Mucous Membrane/metabolism , Prevalence , Pyroglyphidae
18.
Korean J Parasitol ; 54(4): 533-6, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27658607

ABSTRACT

A 26-year-old male member of the Australian Defense Force presented with a history of central abdominal pain of 4 weeks duration and peripheral eosinophilia consistent with eosinophilic enteritis. Acute hookworm disease was diagnosed as the cause. Adult worms recovered from feces after therapy with albendazole were morphologically consistent with Ancylostoma ceylanicum. As the patient had been deployed with the Regional Assistance Mission to Solomon Islands for 6 months prior to this presentation, it is very likely that the A. ceylanicum was acquired in Solomon Islands. Until now, it has been assumed that any Ancylostoma spp. recovered from humans in Solomon Islands is A. duodenale. However, this case demonstrates that human hookworm infection acquired in the Solomon Islands could be caused by A. ceylanicum.


Subject(s)
Ancylostoma/isolation & purification , Ancylostomiasis/diagnosis , Ancylostomiasis/pathology , Enteritis/etiology , Enteritis/pathology , Eosinophilia/etiology , Eosinophilia/pathology , Gastritis/etiology , Gastritis/pathology , Adult , Albendazole/therapeutic use , Ancylostomiasis/drug therapy , Ancylostomiasis/parasitology , Animals , Anthelmintics/therapeutic use , Australia , Enteritis/drug therapy , Enteritis/parasitology , Eosinophilia/drug therapy , Eosinophilia/parasitology , Feces/parasitology , Gastritis/drug therapy , Gastritis/parasitology , Humans , Male , Melanesia , Military Personnel
19.
Immunity ; 43(5): 998-1010, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26522986

ABSTRACT

Intestinal helminths are potent regulators of their host's immune system and can ameliorate inflammatory diseases such as allergic asthma. In the present study we have assessed whether this anti-inflammatory activity was purely intrinsic to helminths, or whether it also involved crosstalk with the local microbiota. We report that chronic infection with the murine helminth Heligmosomoides polygyrus bakeri (Hpb) altered the intestinal habitat, allowing increased short chain fatty acid (SCFA) production. Transfer of the Hpb-modified microbiota alone was sufficient to mediate protection against allergic asthma. The helminth-induced anti-inflammatory cytokine secretion and regulatory T cell suppressor activity that mediated the protection required the G protein-coupled receptor (GPR)-41. A similar alteration in the metabolic potential of intestinal bacterial communities was observed with diverse parasitic and host species, suggesting that this represents an evolutionary conserved mechanism of host-microbe-helminth interactions.


Subject(s)
Gastrointestinal Microbiome/immunology , Helminths/immunology , Hypersensitivity/immunology , Inflammation/immunology , Inflammation/parasitology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Adult , Aged , Animals , Asthma/immunology , Asthma/microbiology , Asthma/parasitology , Cytokines/immunology , Fatty Acids/immunology , Female , Humans , Hypersensitivity/microbiology , Hypersensitivity/parasitology , Inflammation/microbiology , Intestinal Mucosa/parasitology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nematospiroides dubius/immunology , Receptors, G-Protein-Coupled/immunology , Strongylida Infections/immunology , Strongylida Infections/microbiology , Strongylida Infections/parasitology
20.
Sci Rep ; 5: 13797, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26381211

ABSTRACT

The intestinal microbiota plays a critical role in the development of the immune system. Recent investigations have highlighted the potential of helminth therapy for treating a range of inflammatory disorders, including celiac disease (CeD); however, the mechanisms by which helminths modulate the immune response of the human host and ameliorate CeD pathology are unknown. In this study, we investigated the potential role of alterations in the human gut microbiota in helminth-mediated suppression of an inflammatory disease. We assessed the qualitative and quantitative changes in the microbiota of human volunteers with CeD prior to and following infection with human hookworms, and following challenge with escalating doses of dietary gluten. Experimental hookworm infection of the trial subjects resulted in maintenance of the composition of the intestinal flora, even after a moderate gluten challenge. Notably, we observed a significant increase in microbial species richness over the course of the trial, which could represent a potential mechanism by which hookworms can regulate gluten-induced inflammation and maintain intestinal immune homeostasis.


Subject(s)
Ancylostomatoidea , Biodiversity , Celiac Disease/etiology , Gastrointestinal Microbiome , Glutens/metabolism , Hookworm Infections , Ancylostomatoidea/immunology , Animals , Case-Control Studies , Celiac Disease/therapy , Feces/microbiology , Gastrointestinal Microbiome/immunology , High-Throughput Nucleotide Sequencing , Hookworm Infections/immunology , Humans , Immunomodulation , Metagenome , Metagenomics/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...