Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jun 1.
Published in final edited form as: Parasite Immunol. 2015 Jun;37(6):314–323. doi: 10.1111/pim.12193

Helminths and the microbiota: parts of the hygiene hypothesis

P’ng Loke 1,*, Yvonne AL Lim 2,*
PMCID: PMC4428757  NIHMSID: NIHMS684423  PMID: 25869420

Abstract

In modern societies, diseases that are driven by dysregulated immune responses are increasing at an alarming pace, such as inflammatory bowel diseases and diabetes. There is an urgent need to understand these epidemiological trends, which are likely to be driven by the changing environment of the last few decades. There are complex interactions between human genetic factors and this changing environment that is leading to the increasing prevalence of metabolic and inflammatory diseases. Alterations to human gut bacterial communities (the microbiota) and lowered prevalence of helminth infections are potential environmental factors contributing to immune dysregulation. Helminths have co-evolved with the gut microbiota and their mammalian hosts. This three-way interaction is beginning to be characterized and the knowledge gained may enable the design of new therapeutic strategies to treat metabolic and inflammatory diseases. However, these complex interactions need to be carefully investigated in the context of host genetic backgrounds in order to identify optimal treatment strategies. The complex nature of these interactions raises the possibility that only with highly personalized treatment, with knowledge of individual genetic and microbiota communities, will therapeutic interventions be successful for a majority of the individuals suffering from these complex diseases of immune dysregulation.

Introduction

Whilst our previous generations were faced with constant threats of diseases caused by infectious agents (accounting for substantial proportions of human mortality (1)), the current generation is facing an epidemic of diseases associated with dysregulated inflammation (2). These include many metabolic diseases (3). Through defining the mechanisms underlying obesity, diabetes and atherosclerosis (4), we now know that dysregulated or unresolved inflammation influences many of these diseases. There is no doubt from the epidemiological perspective that there is an increase in inflammatory mediated diseases over the last 50 years. Since this is a timeframe that is unfeasible for genetic changes to have occurred in the human population, environmental factors are clearly responsible for these alterations in disease landscapes. However, genetic factors are as important in determining the consequence of these changes in response to environmental changes, as they are to response to infections with infectious pathogens (5). Hence, it is necessary to understand the interaction between genetic and environmental factors (6) to unravel causes behind the increasing rates of inflammatory diseases.

In order to avoid expulsion, helminths have evolved mechanisms to regulate the immune system of their hosts(7). They do not benefit from replicating within the host or utilizing intra-host population genetic approaches towards avoiding the immune response, mainly through antigenic variation (a common strategy for viruses, bacteria and protozoan parasites). Therefore, helminths have evolved production of various immune regulatory molecules (8) during the process of co-evolution, which are encoded into their genomes (9). These genomes are therefore a source of immune regulatory molecules (10). The ability of helminths to regulate the mammalian immune system is a fascinating topic, which has drawn increasing interest as the potential to exploit this knowledge for the treatment of autoimmune diseases is increasingly being considered (11, 12).

Concurrently, there has been an explosion of knowledge and interest regarding the many physiological effects of commensal bacteria (often termed the microbiota) that live with their mammalian hosts(13). While they clearly affect the ability of the host to metabolize and absorb nutrients (as was previously recognized), there has been a surge of interests in how these commensal communities regulate the immune system of their hosts(1416). At the same time, there has been an increasing appreciation of how shifts in these bacterial communities may alter the immune response of their hosts and lead to increased inflammatory responses(17). Since alterations to the microbiota clearly has immune consequences and there are dramatic differences in the microbial communities of residents in developing and developed countries (18) with different rates of inflammatory diseases, there is also much interest to relate microbial changes to the hygiene hypothesis(19).

The most widespread helminth infections in man, as well as in most other mammals, are the intestinal helminths (2022). This is also the locale of the majority of commensal bacteria in mammals. Undoubtedly, there must be some important interactions between these organisms that reside in the same niche (23, 24). In this review, we will discuss some of the recent developments in characterizing the interactions between helminths and the microbiota and the consequences that this may have on the immune response of the mammalian hosts.

Helminth immune responses and the hygiene hypothesis

Helminth infections are generally associated with a Type 2 response, as well as with activation of an immune regulatory network(25). It is widely regarded that the Type 2 response has likely evolved to minimize the virulence of helminth infection to their mammalian hosts (26). This response enables parasite resistance (or expulsion) by limiting the number helminths that can live in our intestinal tract (27, 28) and also promoting the repair of tissue damage that is caused by the helminths that have colonized our tissues (26, 29). This response is characterized by the production of cytokines such as interleukin-4 (IL-4), IL-5, IL-9 and IL-13, which in turn activates a broad range of downstream effector mechanisms (30, 31) that are necessary for host expulsion and tolerance of helminth infections.

IL-4 and IL-13 are key components of this response and they signal through the IL-4Ra and STAT6 (32). Acting on intestinal epithelial cells (IECs), these cytokines promote goblet cell differentiation, increased mucus production, proliferation and turnover of the IECs (27, 28). In the context of the gut microbiota, these alterations to the intestinal environment may be important for maintaining the mucosal barrier and limiting aberrant inflammatory responses triggered by the luminal or adherent bacteria in the mucosa (33). When this Type 2 response is knocked out during helminth infections in mice it can lead to lethal sepsis from compromised gut integrity and leakage of gut bacteria (26). Contraction of intestinal muscles and increased fluid flow into the lumen may help flush the worms out of the gut (27, 28), but are also likely to alter the environmental niche for gut microbial communities (23).

It was first noted from field studies that the peripheral T cells from chronically helminth-infected patients are unresponsive to stimulation with parasite antigens and responses to other antigens are also reduced (7, 34). Subsequently, investigators have worked to define the regulatory mechanisms that may be reducing responses during helminth infection. In addition to the TH2 response described above, regulatory T cells (35), regulatory B cells (36, 37) and alternatively activated (or M2) macrophages (3840) have all been identified as important components of the immune regulatory network activated by helminth infections (31). These same mechanisms may be responsible for preventing immune dysregulation as part of the hygiene hypothesis(12).

Some of the early field evidence to relate allergies and helminths came from Gabon, where there is a high prevalence of schistosomiasis (41). Infected children had lower responses to allergen challenge (skin test to house dust mite antigen) than uninfected children (42). Importantly, deworming the children with anti-helminthics increased reactivity to allergens (43), providing evidence for a causal effect of the worms on downregulating reactivity to the skin allergens. Since this study, there have been other studies in Brazil (44) and Equador (45) that have provided supporting evidence that helminth infections could suppress allergic inflammation (46). Since allergic inflammation and helminth infections result in strong Type 2 responses, why would helminths suppress allergen reactivity? This would not be consistent with the paradigm of a TH2 response inhibiting TH1 or TH17 responses, in which case we would expect helminths to suppress TH1/TH17 driven diseases such as type 1 diabetes(47), multiple sclerosis (48) and Crohn’s disease(12). The answer may lie in the increased levels of IL-10, a highly potent immunosuppressive cytokine. IL-10 may attenuate the ability of basophils to respond to antibody-mediated activation. While the early studies were on schistosomiasis, there is also evidence that gastrointestinal nematodes can also reduce allergic inflammation in the skin (46). However, this effect did not appear to extend to a suppressive effect on developing clinical asthma, indicating perhaps that once chronic pathogenic mechanisms have been established they cannot be overturned by helminths.

The effects of helminth infection may be especially important in early life or even in pregnant mothers. A recent study in Africa enrolled 2507 pregnant women in a randomized, double blind, placebo controlled trial with different anti-helminthic drug treatment(s) to examine immune responses against infant vaccines (49) and the health outcome of offspring. There was neither a beneficial or detrimental effect on infectious disease outcomes or on responses to Bacillus Calmette–Guérin vaccine (BCG) immunization (49, 50). Surprisingly, the incidence of eczema increased with anti-helminthic treatment (50, 51). These results suggest that deworming during pregnancy might promote allergic disease in offspring. This large randomized study provides clinical evidence that helminth infection during pregnancy may protect children from immune dysregulation, as well as indicating that helminth infection during pregnancy does not have a negative effect on the offspring.

While we have thus far described helminths in a positive light with regard to immune regulation, it is important to note though that parasitic helminths are not commensals. In fact they are perhaps the greatest of all neglected diseases (21). There is a heterogeneous immune response to helminth infections in any endemic population (31). Individuals that mount particularly weak or inappropriate immune responses to the parasites may result in carrying heavy worm burdens. These heavy worm burdens are then likely to cause pathology associated with heavy infection (20). In contrast, there will also be some individuals that mount exceedingly strong immune responses to the helminths, these individuals are likely to carry very low worm burdens, but may suffer collateral damage from immune driven pathology (29). For the majority of individuals there is a balance between tolerance (52) and resistance to the presence of the helminths, which enables a small number of parasites to survive within the host without avert pathology (31). Under these circumstances, there is an optimal interaction whereby immune reactivity and parasite-induced suppression of immune responses are balanced to minimize pathology from either heavy worm burden or immune pathology (53). Studies on Soay sheep on an isolated island in Scotland, demonstrate that there can be a balance between reproductive fitness and survival that is driven by the magnitude of an immune response (54), indicating that there is selection for tolerating the presence of helminth infections (55) with a moderate immune response, without causing collateral damage that reduces reproductive ability.

Perhaps helminths should thus be considered to be “pathobionts”, potentially disease causing organisms, which under normal circumstances live as a symbiont with their hosts (56). This also depends on how we define “normal” circumstances. Depending on the type of helminth infection, some would be more likely to be “normally” symbiotic (e.g. gastrointestinal helminths like Trichuris and Ascaris) while others would be weighed more towards the disease-causing spectrum (e.g. schistosomiasis). In general, the gastrointestinal tract is a body site more tolerant to accommodating helminth colonization without pathology.

Microbiota and the hygiene hypothesis

The populations of microbes that are found primarily in the gut of mammals are considered to be their “microbiota”. There is ever increasing evidence that the microbiota plays an important role in regulating homeostasis of their host and consequently has effects on many different disease processes (17). Many different physiological functions, including metabolism, cognitive development (57) and immune responses are affected by the composition of the microbiota. Importantly, alterations to bacterial communities in early life appear to be most important and can have long-term consequences to physiology in adult life (58). There have been many recent reviews on the relationship between the microbiota and human diseases (59,60) and just a few examples are highlighted here.

Inflammation is an important component of many metabolic diseases (61). The relationship between obesity and the gut microbiota is among the best studied of host-microbiota interactions (62). The increase in obesity in the last few decades is striking and clearly a result of environmental alterations. While diet and lifestyle clearly are important contributors, the composition of gut microbial communities can also contribute to progression of obesity. Obesity is now widely considered to be an inflammatory disorder and disruption of specific immune pathways; especially bacterial sensors (e.g. TLR5 (63) and Inflammasome components(64)) result in phenotypes that are associated with increased weight, adiposity or other metabolic effects. Recent studies perturbing the microbiota with low sub-therapeutic doses of antibiotics during early life, has dramatic effects on adiposity in mice, especially when combined with high fat diets. These metabolic changes were also associated with alterations to immune cell function, specifically a reduction in TH17 responses(58).

Inflammatory bowel diseases (IBD), are complex disorders that include Crohn’s disease (CD) and ulcerative colitis. These autoimmune diseases are characterized by recurring inflammation in the gastrointestinal tract (65). While the causes of IBD are still to be determined, there is general consensus that abnormal responses to the gut microbiota are a key component of the disease manifestation (65, 66). Hence, more is known about the role of the microbiota in this disease, which may serve as a model for studies to better understand host-microbiota interactions (59). Imbalances in commensal bacterial populations, referred to as dysbiosis, is a common feature among IBD patients (67). Genetic risk factors are also involved in host-microbe interactions, suggesting a disease model in which certain commensal microbes drive an exaggerated inflammatory response in genetically susceptible individuals. Hence, IBDs are complex diseases and different combinations of genetic and environmental factors (e.g. the microbiota) may require different treatment options. However, animal models that recreate this multi-hit pathogenesis mechanism have been lacking, making it difficult to understand the relationship between genetic, microbial, and other environmental risk factors. We recently developed a mouse model to understand dysbiosis, in which the combination of genetic susceptibility and microbial imbalances generates intestinal inflammation (68). These types of model may enable the gene-microbe-helminth interactions in the future.

Whilst the gut microbiota is restricted to the intestinal tract, metabolites produced by the bacterial communities can influence peripheral tissues (69, 70). In a remarkable study, supplementation of dietary fiber to mice led to increased concentrations of circulating short chain fatty acids (SCFA), as a result of metabolizing the fibers by the gut microbiota(71). This increase in SCFAs in turn protected the mice from allergic inflammation(71). SCFAs have also been shown to regulate intestinal inflammation and promote differentiation of Foxp3+ Tregs(72) (69). These finding provide a potential mechanistic link between alterations to our modern diet (low fiber high fat) and increased susceptibility to inflammatory diseases. Further studies, especially translational studies in human subjects with dietary alterations (73, 74) should validate some of these hypotheses.

As a result of the Human Microbiome Project, we now have a detailed catalog of the healthy human microbiota from individuals of the developed world (75, 76). The microbiome refers to the genomes of the microbiota. However, it is clear from the few studies conducted so far that the gut microbiota of developing countries are extremely different to residents from the developed world (18, 77, 78). Notably, helminth infection status of individuals analyzed from developing countries has not been well described in most of these studies. Diet certainly would contribute significantly to differences between the developing and developed countries. But there is growing evidence that helminth infections may also have a substantial impact on the human microbiota. Recent studies have also emerged indicating that specific taxa within the microbiota of individuals living in developing countries have profound effects on the health outcome of these people; for example, promoting the recovery from cholera (79), and affecting malnutrition (80, 81). The effects of helminth infections in these interactions remain to be determined.

Helminths and microbial communities

Just as the immune system has co-evolved with the gut microbiota, it has co-evolved to tolerate the presence of helminth infections. Hence, it is undoubtedly the case that the helminths and the gut microbiota must have co-evolved also. While this is still a new area of investigation, there is growing evidence that intestinal helminths can alter the composition of the gut microbiota.

Some of the earlier experiments were conducted on pigs. Trichuris suis is associated with exacerbation of campylobacteriosis (82). More recent data with next generation sequencing showed that Mucispirillum bacteria, which colonize mucus, were substantially increased in infected animals (83). Shotgun sequencing indicated that T. suis infection reduces carbohydrate metabolism, coinciding with reductions in Ruminococcus bacterium that are cellulolytic (83). A study to compare relationship between the mucosal immune response and worm burden was possible because at a later time point (53 days) some pigs were cleared of adult worms, whereas others were still colonized, enabling the comparison of infected pigs that retained worms with pigs that had cleared worms (84). Interestingly, Campylobacter was more common in worm bearing pigs. Greater worm burden was also associated with increased expression of inflammatory genes, arg1, cxcr2, c3ar1, il6, muc5ac and ptgs2 (84). However, the functional relationship between the inflammatory response, worm burden and bacterial communities is difficult to determine in pigs and easier to investigate in mice.

Heligmosomoides polygyrus can alter gut microbiota of healthy mice (8587) in the large intestine, despite the parasite occupying the small intestine. Members of the Lactobacillaceae family were increased in abundance in several independent studies (8587), as were the Enterobacteria (86, 87). Very early experiments with germ-free mice had found that fewer adult worms were recovered from these mice, which was associated with increased eosinophilia, granulomas and thickening of the small intestinal wall (88), however this finding still awaits validation in the modern germ-free setting.

Intriguingly, the abundance of Lactobacillus was found to correlate positively with infection intensity of H. polygyrus, in addition to H. polygyrus increasing the abundance of Lactobacillus (87). Indeed, the introduction of exogenous Lactobacillus to mice promoted establishment of the worm infection (87). This synergistic relationship between worm and bacteria was reminiscent of another nematode, Trichuris muris, which utilizes the cecal microbiota to provide environmental cues for hatching of the larvae (89). It is still unclear if T. muris can also alter the intestinal microbiota to promote expansion of bacterial taxa that can enable hatching. Since these worms need to find mates in order to produce eggs, it would be sensible to adopt such strategies. The relationship between H. polygyrus and Lactobacillus could be driven by an expansion of regulatory T cells, which is correlated with exogenous delivery of Lactobacillus (87) and has been demonstrated to promote H. polygyrus colonization. Increase in Lactobacillaceae is not restricted to the gastrointestinal nematodes and interestingly the liver fluke Opisthorchis viverrini also increases this group of organisms (90), along with the Lachnospiraceae and Ruminococcaceae. One possibility is that these alterations are driven by the Type 2 response, a common feature of these different helminth infections.

There are now a few reported studies from humans in endemic regions infected with helminths, with many others in the pipeline (unpublished). We performed a cross-sectional study on 51 individuals from two Orang Asli villages in Malaysia, of which 36 (70.6%) were infected by helminths (91). The Orang Asli are an indigenous population who live in the rural and semi-urban areas of Peninsular Malaysia where helminth infection remains prevalent. We found that the helminth-colonized individuals among these villagers had greater species richness and number of observed OTUs, compared to the uninfected individuals. Recently, we have followed up these studies with a longitudinal deworming study and confirmed that deworming decreases microbial diversity, indicating a causal relationship between helminth infections and microbial diversity (unpublished). However, a different study conducted in Ecuador did not find significant effects of helminth infection on microbial diversity(92). In this study, the investigators compared uninfected with infected children, but also compared children before and after treatment with deworming medication. There were also no significant differences in bacterial composition. Hence, there may be geographic and cultural differences that could influence these studies, or technical differences in sample storage, extraction and sequencing platforms may also affect the results.

In summary, experimental models suggest that helminths and the microbiota may interact in positive and negative feedback loops, whereby a particular worm (e.g. H. polygyrus) is better suited to an environment with certain bacterial communities (e.g. abundant Lactobacilli) and therefore produces factors that promotes the growth of bacterial communities that favors it’s presence. This may also be a strategy to exclude other worms, or enteric organisms from occupying this niche. The heterogeneity of worm burdens that is typically observed in an endemically infected human population, with 15% of the population often carrying 80% of the worm burden (21, 93), may well be a reflection of this interaction. This is a testable hypothesis for future field studies. The results from endemic human studies are still variable at the moment and definitive conclusions will await further reports.

Helminths as treatment for inflammatory diseases

Gastroenterologists at the University of Iowa led the way in taking the emerging concepts of immune regulation by helminths into translational research on inflammatory bowel disease patients (9496). Trichuris suis was utilized as a therapeutic agent because it produces a self-limited colonization in humans and remains isolated to the gastrointestinal tract (97). An exploratory open-label study of seven patients with IBD was first reported in 2003. In this study and subsequent clinical trials of Trichuris suis ova (TSO) in Iowa, there were significant improvements in patient outcomes for both UC and Crohn’s disease with essentially no adverse effects (9496). In a randomized, placebo-controlled, double-blind study of 54 subjects with moderate to severe UC, after 12 weeks of treatment, 43.3% of the individuals treated with TSO had improved symptoms compared to 16.7% in the placebo group, which was statistically significant.

Subsequently, small investigator initiated trials were established to evaluate efficacy for multiple sclerosis (98, 99), based on observations in Argentina indicating positive effects of helminth infections on MS patients (100). There was also a larger randomized double blind, placebo-controlled trial of 100 subjects with allergic rhinitis (101). The allergic rhinitis trial did not show any significant effect on symptom scores or allergic reactivity. While there was no effect on symptoms, this trial provided an indication of potential side effects for relatively healthy individuals, not suffering from chronic gastrointestinal disorders. A few side effects were documented (102), including diarrhea, excessive flatulence, and upper abdominal pain. These events peaked 30 to 50 days after the first treatment with TSO, but were generally transient (median duration of two days) and subsided. Perhaps this is an indication of alterations in gut microbiota as a result of TSO.

The other helminth studied in clinical trials is the hookworm N. americanus, which has been tested on asthma and celiac disease patients. TSO is being used in clinical trials with dosages as high as 7500 eggs. For N. americanus there may be a narrower therapeutic window between achieving effective immune modulation and unacceptable adverse events. Doses higher than 10 larvae already correlate with increased adverse events (103). In a randomized, double-blinded trial with 32 asthma subjects, the differences between the treated and placebo groups were non-significant (104). N. americanus has also been tested in a randomized double blind clinical trial on 20 patients with well-controlled clinically inactive celiac disease (105). In this initial study, subjects underwent a five-day gluten challenge 12 weeks post inoculation and no significant difference was observed in pathologic grade or systemic inflammatory immune response after gluten challenge between the hookworm infected and placebo treated groups. However, a biological response was elicited to N. americanus in these subjects with a strong mucosal TH2 responses as well as increased IL-22 expression accompanied by declines in IFNg and IL-17A secretion from cultured duodenal pinch biopsies (106, 107). Remarkably, with a different study design (108), with escalating gluten challenges, N. americanus combined with gluten microchallenge showed promise in promoting tolerance to larger challenges of subsequent gluten challenge. These results clearly illustrate the importance of devising an appropriate intervention strategy when utilizing helminths as a therapeutic agent.

Whereas in the previous section, we discussed early studies on the effects of helminth infections on the microbiota of humans living in endemic regions, there is also an emerging body of work on the microbiota from experimental treatments of individuals with helminth infections. We performed a longitudinal treatment study of juvenile macaques suffering from idiopathic chronic colitis, treated with Trichuris trichiura eggs we received from a self-infected individual (109). This colitis condition is a major cause of required veterinary attention in primate research centers. Four of the 5 treated macaques responded positively to infection and gained weight progressively. When we examined the composition of the mucosal microbiota from the biopsy specimens, we found that microbial diversity was significantly reduced in colitic macaques compared to healthy macaques (consistent with previous reports (110)) and diversity was partially restored post treatment. The bacterial communities post treatment was also more similar to healthy macaques. These results indicated that Trichuris infection might reverse dysbiosis in these macaques with colitis. In a study of healthy celiac disease patients infected with N. americanus, fecal samples were analyzed from 8 healthy celiac disease patients on a long-term gluten-free diet before and after infection with N. americanus (105107) and analyzed by 16S sequencing. The results showed a small increase in microbial species richness, but there was not a dramatic effect on community structure, diversity or relative abundance of individual bacterial species. One possibility why the effects on the microbiota was minor could be because N. americanus colonizes the small intestine, rather than the colon.

The only large placebo controlled trials thus far conducted with the GMP grade helminth product, TSO, (for allergic rhinitis and Crohn’s disease) have failed to demonstrate significant improvement of symptoms compared to placebo alone. While these results have been discouraging, it is important to note that our understanding of how TSO may affect a wide range of patients is poor. Currently, there are no good ways to measure correlates of biological activity. With conventional drugs (e.g. antibodies and small molecules), determination of accurate pharmacokinetics is important for dosing. For a live therapeutic organism, this information is unavailable. This scenario is more similar to a therapeutic vaccine, where investigators can still measure antibody responses or T cell response to antigens as a correlate of biological activity. In the case of TSO, hatching rates and hence parasite loads could vary significantly between individuals, and is not well understood. The composition of gut microbiota from different individuals may result in different hatching rates, since hatching of the eggs has been shown to be dependent on gut bacteria (23, 89). Hence, many challenges remain for this form of new disease treatment, as is often the case for “first in class” therapeutics.

Challenges

Immunologists tend to get confused about the diverse biological properties of different helminths. Differences in life cycles, location of larvae relative to adult forms and the migration process through different body cavities, blood vessels and lymphatic systems tend to be under appreciated. “Worms are a way to induce Type 2 responses”, is a common thought process for some immunologists. While it is indeed true that Type 2 responses are a general feature of most helminth infections, and indeed features of immune regulation are also shared between most helminth infections, it is also important to note that the relative magnitude of the Type 2 response and immune regulatory response varies greatly depending on which helminth infection is being investigated (111). Indeed, the type of response is also greatly affected by the dosage of parasites used and of course the strain of mice under investigation (28).

In the same vein as above, there is often a failure to appreciate the complexities of the many different types of autoimmune or inflammatory diseases. Indeed within the same type of disease, there are huge variations in manifestations of pathology and disease course, indicating perhaps that there are actually a number of different diseases that are being categorized together, but are in effect, many separate diseases. Whereas with cancer, there is an increasing appreciation of sub-categorizing cancer types based on molecular profiles (112116), autoimmune and inflammatory diseases are still often being treated as a single entity, because we lack a molecular and cellular understanding of the different disease manifestations.

Thus, while there are clear examples in mouse models (11), and in individual cases (117), where a particular helminth can have dramatic effects at improving disease pathology of an inflammatory disease, there is also unlimited possibilities in pairing different helminth infections with different autoimmune and inflammatory diseases, and we are poorly placed at the moment to do so, because we do not understand the complexities of the different infections and inflammatory diseases. It is therefore dangerous to predict that any helminth infection can suppress any particular inflammatory disease, because it is just as likely to have unintended consequences of exacerbating disease (118, 119). What is likely required instead is the careful study and pairing of specific helminths (or helminth products), with specific immunological features with certain specific (perhaps even subsets?) diseases. It is unlikely that any single helminth is going to have a beneficial effect on several different inflammatory diseases.

Future directions

It is clear that gut microbial communities can exert a strong influence on the host immune response (14, 15, 120). In mouse experimental models, we can determine that these effects are definitely happening in the absence of helminth infections. While we are equally certain that helminth infections can exert strong influences on the immune response of the host (121), it has not been clearly demonstrated if these immune effects can occur in the absence of gut bacteria. Hence, it is possible that certain elements of immune regulation by helminths may be caused by alterations to the bacterial communities of the host. Germ-free mice experiments with helminth infections may clarify this in the future. In a recent study on virus-helminth co-infection (122), helminths impaired antiviral immunity, but the immune regulation was still present in germ-free mice, indicating that regulation can occur independently of the microbiota. Whether this observation is a general rule will require further investigation. Clearly, we have a lot to learn about the many different types of transkingdom interactions that can occur between the mammalian hosts, viruses, bacteria, fungi and helminths. How these interactions may impact the regulation of the immune system will be the subject of future studies.

Bacterial communities have a tendency to stabilize into several distinct clusters (sometimes referred to as enterotypes(123)). These community structures appear to be relatively resilient to perturbations (13). It has been suggested that once community structures have stabilized, considerably more “activation energy” may be required to alter these structures (59). One possibility is that helminths (or the immune response to helminths) are a driving force favoring specific bacterial communities that are more immune regulatory (24). An exciting area of further research will be to characterize the dynamic interactions between helminth infections and bacterial communities to determine whether they may promote stability and resilience of certain communities, as well as decoupling the immunological effects of the microbiota from the helminths.

Acknowledgements

PL and YALL are funded by a UM/MoHE High Impact Research (H-20001-00-E000061) grant from University of Malaya (YALL and PL), the Kevin and Masha Keating Family Foundation (PL), National Institutes of Health (AI093811 and AI094166) and the Broad Medical Research Program. The funders had no role in the decision to publish, or preparation of the manuscript.

Footnotes

Disclosures: The authors declare that they do not have potential conflicts of interest.

References

  • 1.Cohen ML. Changing patterns of infectious disease. Nature. 2000 Aug 17;406(6797):762–767. doi: 10.1038/35021206. PubMed PMID: 10963605. [DOI] [PubMed] [Google Scholar]
  • 2.Murray CJ, Lopez AD. Alternative projections of mortality and disability by cause 1990–2020: Global Burden of Disease Study. Lancet. 1997 May 24;349(9064):1498–1504. doi: 10.1016/S0140-6736(96)07492-2. PubMed PMID: 9167458. [DOI] [PubMed] [Google Scholar]
  • 3.Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA. 2002 Jan 16;287(3):356–359. doi: 10.1001/jama.287.3.356. PubMed PMID: 11790215. [DOI] [PubMed] [Google Scholar]
  • 4.Ross R. Atherosclerosis--an inflammatory disease. N Engl J Med. 1999 Jan 14;340(2):115–126. doi: 10.1056/NEJM199901143400207. PubMed PMID: 9887164. [DOI] [PubMed] [Google Scholar]
  • 5.Casanova JL, Abel L. Human genetics of infectious diseases: a unified theory. EMBO J. 2007 Feb 21;26(4):915–922. doi: 10.1038/sj.emboj.7601558. PubMed PMID: 17255931. Pubmed Central PMCID: 1852849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Spor A, Koren O, Ley R. Unravelling the effects of the environment and host genotype on the gut microbiome. Nat Rev Microbiol. 2011 Apr;9(4):279–290. doi: 10.1038/nrmicro2540. PubMed PMID: 21407244. [DOI] [PubMed] [Google Scholar]
  • 7.McSorley HJ, Maizels RM. Helminth infections and host immune regulation. Clin Microbiol Rev. 2012 Oct;25(4):585–608. doi: 10.1128/CMR.05040-11. PubMed PMID: 23034321. Pubmed Central PMCID: 3485755. Epub 2012/10/05. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Harnett W, Harnett MM. Helminth-derived immunomodulators: can understanding the worm produce the pill? Nature reviews. 2010 Apr;10(4):278–284. doi: 10.1038/nri2730. PubMed PMID: 20224568. Epub 2010/03/13. eng. [DOI] [PubMed] [Google Scholar]
  • 9.Brindley PJ, Mitreva M, Ghedin E, Lustigman S. Helminth genomics: The implications for human health. PLoS Negl Trop Dis. 2009;3(10):e538. doi: 10.1371/journal.pntd.0000538. PubMed PMID: 19855829. Pubmed Central PMCID: 2757907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Maizels RM, Yazdanbakhsh M. Immune regulation by helminth parasites: cellular and molecular mechanisms. Nature reviews. 2003 Sep;3(9):733–744. doi: 10.1038/nri1183. PubMed PMID: 12949497. eng. [DOI] [PubMed] [Google Scholar]
  • 11.Elliott DE, Summers RW, Weinstock JV. Helminths as governors of immune-mediated inflammation. International journal for parasitology. 2007 Apr;37(5):457–464. doi: 10.1016/j.ijpara.2006.12.009. PubMed PMID: 17313951. eng. [DOI] [PubMed] [Google Scholar]
  • 12.Elliott DE, Weinstock JV. Helminth-host immunological interactions: prevention and control of immune-mediated diseases. Annals of the New York Academy of Sciences. 2011 Jan;1247:83–96. doi: 10.1111/j.1749-6632.2011.06292.x. PubMed PMID: 22239614. Epub 2012/01/14. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012 Sep 13;489(7415):220–230. doi: 10.1038/nature11550. PubMed PMID: 22972295. Pubmed Central PMCID: 3577372. Epub 2012/09/14. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Honda K, Littman DR. The Microbiome in Infectious Disease and Inflammation. Annu Rev Immunol. 2012 Mar 24; doi: 10.1146/annurev-immunol-020711-074937. PubMed PMID: 22224764. Epub 2012/01/10. Eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012 Jun 8;336(6086):1268–1273. doi: 10.1126/science.1223490. PubMed PMID: 22674334. Epub 2012/06/08. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ivanov II, Littman DR. Modulation of immune homeostasis by commensal bacteria. Curr Opin Microbiol. Feb;14(1):106–114. doi: 10.1016/j.mib.2010.12.003. PubMed PMID: 21215684. Pubmed Central PMCID: 3123735. Epub 2011/01/11. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Cho I, Blaser MJ. The human microbiome: at the interface of health and disease. Nat Rev Genet. 2012 Apr;13(4):260–270. doi: 10.1038/nrg3182. PubMed PMID: 22411464. Epub 2012/03/14. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012 Jun 14;486(7402):222–227. doi: 10.1038/nature11053. PubMed PMID: 22699611. Pubmed Central PMCID: 3376388. Epub 2012/06/16. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Blaser MJ, Falkow S. What are the consequences of the disappearing human microbiota? Nat Rev Microbiol. 2009 Dec;7(12):887–894. doi: 10.1038/nrmicro2245. PubMed PMID: 19898491. Epub 2009/11/10. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bethony J, Brooker S, Albonico M, Geiger SM, Loukas A, Diemert D, et al. Soil-transmitted helminth infections: ascariasis, trichuriasis, and hookworm. Lancet. 2006 May 6;367(9521):1521–1532. doi: 10.1016/S0140-6736(06)68653-4. PubMed PMID: 16679166. [DOI] [PubMed] [Google Scholar]
  • 21.Hotez PJ, Brindley PJ, Bethony JM, King CH, Pearce EJ, Jacobson J. Helminth infections: the great neglected tropical diseases. The Journal of clinical investigation. 2008 Apr;118(4):1311–1321. doi: 10.1172/JCI34261. PubMed PMID: 18382743. Pubmed Central PMCID: 2276811. Epub 2008/04/03. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hotez PJ, Mistry N, Rubinstein J, Sachs JD. Integrating neglected tropical diseases into AIDS, tuberculosis, and malaria control. The New England journal of medicine. 2011 Jun 2;364(22):2086–2089. doi: 10.1056/NEJMp1014637. PubMed PMID: 21631320. [DOI] [PubMed] [Google Scholar]
  • 23.Bancroft AJ, Hayes KS, Grencis RK. Life on the edge: the balance between macrofauna, microflora and host immunity. Trends in parasitology. Mar;28(3):93–98. doi: 10.1016/j.pt.2011.12.001. PubMed PMID: 22257556. Epub 2012/01/20. eng. [DOI] [PubMed] [Google Scholar]
  • 24.Leung JM, Loke P. A role for IL-22 in the relationship between intestinal helminths, gut microbiota and mucosal immunity. International journal for parasitology. 2012 Nov 20; doi: 10.1016/j.ijpara.2012.10.015. PubMed PMID: 23178750. Epub 2012/11/28. Eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Maizels RM, Balic A, Gomez-Escobar N, Nair M, Taylor MD, Allen JE. Helminth parasites--masters of regulation. Immunol Rev. 2004 Oct;201:89–116. doi: 10.1111/j.0105-2896.2004.00191.x. PubMed PMID: 15361235. Epub 2004/09/14. eng. [DOI] [PubMed] [Google Scholar]
  • 26.Allen JE, Wynn TA. Evolution of Th2 immunity: a rapid repair response to tissue destructive pathogens. PLoS Pathog. 2011 May;7(5):e1002003. doi: 10.1371/journal.ppat.1002003. PubMed PMID: 21589896. Pubmed Central PMCID: 3093361. Epub 2011/05/19. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Artis D, Grencis RK. The intestinal epithelium: sensors to effectors in nematode infection. Mucosal Immunol. 2008 Jul;1(4):252–264. doi: 10.1038/mi.2008.21. PubMed PMID: 19079187. Epub 2008/12/17. eng. [DOI] [PubMed] [Google Scholar]
  • 28.Grencis RK, Humphreys NE, Bancroft AJ. Immunity to gastrointestinal nematodes: mechanisms and myths. Immunol Rev. 2014 Jul;260(1):183–205. doi: 10.1111/imr.12188. PubMed PMID: 24942690. Pubmed Central PMCID: 4141702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gause WC, Wynn TA, Allen JE. Type 2 immunity and wound healing: evolutionary refinement of adaptive immunity by helminths. Nature reviews. 2013 Jul 5; doi: 10.1038/nri3476. PubMed PMID: 23827958. Epub 2013/07/06. Eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pulendran B, Artis D. New paradigms in type 2 immunity. Science. 2012 Jul 27;337(6093):431–435. doi: 10.1126/science.1221064. PubMed PMID: 22837519. Epub 2012/07/28. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Allen JE, Maizels RM. Diversity and dialogue in immunity to helminths. Nature reviews. 2011 Jun;11(6):375–388. doi: 10.1038/nri2992. PubMed PMID: 21610741. Epub 2011/05/26. eng. [DOI] [PubMed] [Google Scholar]
  • 32.O'Shea JJ, Plenge R. JAK and STAT signaling molecules in immunoregulation and immune-mediated disease. Immunity. 2012 Apr 20;36(4):542–550. doi: 10.1016/j.immuni.2012.03.014. PubMed PMID: 22520847. Pubmed Central PMCID: 3499974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wolff MJ, Broadhurst MJ, Loke P. Helminthic therapy: improving mucosal barrier function. Trends in parasitology. 2012 May;28(5):187–194. doi: 10.1016/j.pt.2012.02.008. PubMed PMID: 22464690. Epub 2012/04/03. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yazdanbakhsh M, Kremsner PG, van Ree R. Allergy, parasites, and the hygiene hypothesis. Science. 2002 Apr 19;296(5567):490–494. doi: 10.1126/science.296.5567.490. PubMed PMID: 11964470. Epub 2002/04/20. eng. [DOI] [PubMed] [Google Scholar]
  • 35.Taylor MD, van der Werf N, Maizels RM. T cells in helminth infection: the regulators and the regulated. Trends in immunology. 2012 Apr;33(4):181–189. doi: 10.1016/j.it.2012.01.001. PubMed PMID: 22398370. Epub 2012/03/09. eng. [DOI] [PubMed] [Google Scholar]
  • 36.Hussaarts L, van der Vlugt LE, Yazdanbakhsh M, Smits HH. Regulatory B-cell induction by helminths: implications for allergic disease. J Allergy Clin Immunol. 2011 Oct;128(4):733–739. doi: 10.1016/j.jaci.2011.05.012. PubMed PMID: 21684587. Epub 2011/06/21. eng. [DOI] [PubMed] [Google Scholar]
  • 37.Mangan NE, Fallon RE, Smith P, van Rooijen N, McKenzie AN, Fallon PG. Helminth infection protects mice from anaphylaxis via IL-10-producing B cells. J Immunol. 2004 Nov 15;173(10):6346–6356. doi: 10.4049/jimmunol.173.10.6346. PubMed PMID: 15528374. eng. [DOI] [PubMed] [Google Scholar]
  • 38.Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010 May 28;32(5):593–604. doi: 10.1016/j.immuni.2010.05.007. PubMed PMID: 20510870. Epub 2010/06/01. eng. [DOI] [PubMed] [Google Scholar]
  • 39.Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013 Apr 25;496(7446):445–455. doi: 10.1038/nature12034. PubMed PMID: 23619691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Van Dyken SJ, Locksley RM. Interleukin-4- and Interleukin-13-Mediated Alternatively Activated Macrophages: Roles in Homeostasis and Disease. Annu Rev Immunol. 2013 Jan 3; doi: 10.1146/annurev-immunol-032712-095906. PubMed PMID: 23298208. Epub 2013/01/10. Eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Smits HH, Everts B, Hartgers FC, Yazdanbakhsh M. Chronic helminth infections protect against allergic diseases by active regulatory processes. Current allergy and asthma reports. 2010 Jan;10(1):3–12. doi: 10.1007/s11882-009-0085-3. PubMed PMID: 20425508. Pubmed Central PMCID: 2816799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.van den Biggelaar AHJ, van Ree R, Rodrigues LC, Lell B, Deelder AM, Kremsner PG, et al. Decreased atopy in children infected with Schistosoma haematobium: a role for parasite-induced interleukin-10. The Lancet. 2000;356(9243):1723–1727. doi: 10.1016/S0140-6736(00)03206-2. [DOI] [PubMed] [Google Scholar]
  • 43.van den Biggelaar AH, Rodrigues LC, van Ree R, van der Zee JS, Hoeksma-Kruize YC, Souverijn JH, et al. Long-term treatment of intestinal helminths increases mite skin-test reactivity in Gabonese schoolchildren. J Infect Dis. 2004 Mar 1;189(5):892–900. doi: 10.1086/381767. PubMed PMID: 14976607. [DOI] [PubMed] [Google Scholar]
  • 44.Araujo MI, Lopes AA, Medeiros M, Cruz AA, Sousa-Atta L, Sole D, et al. Inverse association between skin response to aeroallergens and Schistosoma mansoni infection. International archives of allergy and immunology. 2000 Oct;123(2):145–148. doi: 10.1159/000024433. PubMed PMID: 11060486. [DOI] [PubMed] [Google Scholar]
  • 45.Cooper PJ, Chico ME, Rodrigues LC, Ordonez M, Strachan D, Griffin GE, et al. Reduced risk of atopy among school-age children infected with geohelminth parasites in a rural area of the tropics. J Allergy Clin Immunol. 2003 May;111(5):995–1000. doi: 10.1067/mai.2003.1348. PubMed PMID: 12743563. [DOI] [PubMed] [Google Scholar]
  • 46.Cooper PJ. Interactions between helminth parasites and allergy. Current opinion in allergy and clinical immunology. 2009 Feb;9(1):29–37. doi: 10.1097/ACI.0b013e32831f44a6. PubMed PMID: 19106698. Pubmed Central PMCID: 2680069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Cooke A. Review series on helminths, immune modulation and the hygiene hypothesis: how might infection modulate the onset of type 1 diabetes? Immunology. 2009 Jan;126(1):12–17. doi: 10.1111/j.1365-2567.2008.03009.x. PubMed PMID: 19120494. Pubmed Central PMCID: 2632708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fleming JO, Isaak A, Lee JE, Luzzio CC, Carrithers MD, Cook TD, et al. Probiotic helminth administration in relapsing-remitting multiple sclerosis: a phase 1 study. Mult Scler. 2011 Jun;17(6):743–754. doi: 10.1177/1352458511398054. PubMed PMID: 21372112. Epub 2011/03/05. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Webb EL, Mawa PA, Ndibazza J, Kizito D, Namatovu A, Kyosiimire-Lugemwa J, et al. Effect of single-dose anthelmintic treatment during pregnancy on an infant's response to immunisation and on susceptibility to infectious diseases in infancy: a randomised, double-blind, placebo-controlled trial. Lancet. 2011 Jan 1;377(9759):52–62. doi: 10.1016/S0140-6736(10)61457-2. PubMed PMID: 21176950. Pubmed Central PMCID: 3018567. Epub 2010/12/24. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ndibazza J, Mpairwe H, Webb EL, Mawa PA, Nampijja M, Muhangi L, et al. Impact of anthelminthic treatment in pregnancy and childhood on immunisations, infections and eczema in childhood: a randomised controlled trial. PLoS One. 2012;7(12):e50325. doi: 10.1371/journal.pone.0050325. PubMed PMID: 23236367. Pubmed Central PMCID: 3517620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Mpairwe H, Webb EL, Muhangi L, Ndibazza J, Akishule D, Nampijja M, et al. Anthelminthic treatment during pregnancy is associated with increased risk of infantile eczema: randomised-controlled trial results. Pediatric allergy and immunology : official publication of the European Society of Pediatric Allergy and Immunology. 2011 May;22(3):305–312. doi: 10.1111/j.1399-3038.2010.01122.x. PubMed PMID: 21255083. Pubmed Central PMCID: 3130136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Medzhitov R, Schneider DS, Soares MP. Disease tolerance as a defense strategy. Science. 2012 Feb 24;335(6071):936–941. doi: 10.1126/science.1214935. PubMed PMID: 22363001. Epub 2012/03/01. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Little TJ, Shuker DM, Colegrave N, Day T, Graham AL. The coevolution of virulence: tolerance in perspective. PLoS Pathog. 2010;6(9):e1001006. doi: 10.1371/journal.ppat.1001006. PubMed PMID: 20838464. Pubmed Central PMCID: 2936544. Epub 2010/09/15. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Graham AL, Hayward AD, Watt KA, Pilkington JG, Pemberton JM, Nussey DH. Fitness correlates of heritable variation in antibody responsiveness in a wild mammal. Science. 2010 Oct 29;330(6004):662–665. doi: 10.1126/science.1194878. PubMed PMID: 21030656. Epub 2010/10/30. eng. [DOI] [PubMed] [Google Scholar]
  • 55.Hayward AD, Nussey DH, Wilson AJ, Berenos C, Pilkington JG, Watt KA, et al. Natural selection on individual variation in tolerance of gastrointestinal nematode infection. PLoS biology. 2014 Jul;12(7):e1001917. doi: 10.1371/journal.pbio.1001917. PubMed PMID: 25072883. Pubmed Central PMCID: 4114752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Chow J, Tang H, Mazmanian SK. Pathobionts of the gastrointestinal microbiota and inflammatory disease. Curr Opin Immunol. 2011 Aug;23(4):473–480. doi: 10.1016/j.coi.2011.07.010. PubMed PMID: 21856139. Pubmed Central PMCID: 3426444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nature reviews Neuroscience. 2012 Oct;13(10):701–712. doi: 10.1038/nrn3346. PubMed PMID: 22968153. [DOI] [PubMed] [Google Scholar]
  • 58.Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, et al. Altering the Intestinal Microbiota during a Critical Developmental Window Has Lasting Metabolic Consequences. Cell. 2014 Aug 14;158(4):705–721. doi: 10.1016/j.cell.2014.05.052. PubMed PMID: 25126780. Pubmed Central PMCID: 4134513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Huttenhower C, Kostic AD, Xavier RJ. Inflammatory Bowel Disease as a Model for Translating the Microbiome. Immunity. 2014 Jun 19;40(6):843–854. doi: 10.1016/j.immuni.2014.05.013. PubMed PMID: 24950204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012 Mar 16;148(6):1258–1270. doi: 10.1016/j.cell.2012.01.035. PubMed PMID: 22424233. Epub 2012/03/20. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Odegaard JI, Chawla A. Alternative macrophage activation and metabolism. Annual review of pathology. 2011;6:275–297. doi: 10.1146/annurev-pathol-011110-130138. PubMed PMID: 21034223. Pubmed Central PMCID: 3381938. Epub 2010/11/03. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tilg H, Kaser A. Gut microbiome, obesity, and metabolic dysfunction. The Journal of clinical investigation. 2011 Jun;121(6):2126–2132. doi: 10.1172/JCI58109. PubMed PMID: 21633181. Pubmed Central PMCID: 3104783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, Mwangi S, Srinivasan S, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science. Apr 9;328(5975):228–231. doi: 10.1126/science.1179721. PubMed PMID: 20203013. Epub 2010/03/06. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Strowig T, Henao-Mejia J, Elinav E, Flavell R. Inflammasomes in health and disease. Nature. 2012 Jan 19;481(7381):278–286. doi: 10.1038/nature10759. PubMed PMID: 22258606. [DOI] [PubMed] [Google Scholar]
  • 65.Abraham C, Cho JH. Inflammatory bowel disease. The New England journal of medicine. 2009 Nov 19;361(21):2066–2078. doi: 10.1056/NEJMra0804647. PubMed PMID: 19923578. Epub 2009/11/20. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annu Rev Immunol. 2010;28:573–621. doi: 10.1146/annurev-immunol-030409-101225. PubMed PMID: 20192811. Epub 2010/03/03. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tamboli CP, Neut C, Desreumaux P, Colombel JF. Dysbiosis in inflammatory bowel disease. Gut. 2004 Jan;53(1):1–4. doi: 10.1136/gut.53.1.1. PubMed PMID: 14684564. Pubmed Central PMCID: 1773911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ramanan D, Tang MS, Bowcutt R, Loke P, Cadwell K. Bacterial Sensor Nod2 Prevents Inflammation of the Small Intestine by Restricting the Expansion of the Commensal Bacteroides vulgatus. Immunity. 2014 Aug 21;41(2):311–324. doi: 10.1016/j.immuni.2014.06.015. PubMed PMID: 25088769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013 Dec 19;504(7480):451–455. doi: 10.1038/nature12726. PubMed PMID: 24226773. Pubmed Central PMCID: 3869884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Thorburn AN, Macia L, Mackay CR. Diet, metabolites, and "western-lifestyle" inflammatory diseases. Immunity. 2014 Jun 19;40(6):833–842. doi: 10.1016/j.immuni.2014.05.014. PubMed PMID: 24950203. [DOI] [PubMed] [Google Scholar]
  • 71.Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014 Feb;20(2):159–166. doi: 10.1038/nm.3444. PubMed PMID: 24390308. [DOI] [PubMed] [Google Scholar]
  • 72.Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013 Aug 2;341(6145):569–573. doi: 10.1126/science.1241165. PubMed PMID: 23828891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014 Jan 23;505(7484):559–563. doi: 10.1038/nature12820. PubMed PMID: 24336217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011 Oct 7;334(6052):105–108. doi: 10.1126/science.1208344. PubMed PMID: 21885731. Pubmed Central PMCID: 3368382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Structure, function and diversity of the healthy human microbiome. Nature. Jun 14;486(7402):207–214. doi: 10.1038/nature11234. PubMed PMID: 22699609. Epub 2012/06/16. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.A framework for human microbiome research. Nature. Jun 14;486(7402):215–221. doi: 10.1038/nature11209. PubMed PMID: 22699610. Pubmed Central PMCID: 3377744. Epub 2012/06/16. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Blaser MJ, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Estrada I, et al. Distinct cutaneous bacterial assemblages in a sampling of South American Amerindians and US residents. ISME J. 2013;7(1):85–95. doi: 10.1038/ismej.2012.81. PubMed PMID: 22895161. Pubmed Central PMCID: 3526177. Epub 2012/08/17. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010 Aug 17;107(33):14691–14696. doi: 10.1073/pnas.1005963107. PubMed PMID: 20679230. Pubmed Central PMCID: 2930426. Epub 2010/08/04. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hsiao A, Ahmed AM, Subramanian S, Griffin NW, Drewry LL, Petri WA, Jr, et al. Members of the human gut microbiota involved in recovery from Vibrio cholerae infection. Nature. 2014 Nov 20;515(7527):423–426. doi: 10.1038/nature13738. PubMed PMID: 25231861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Subramanian S, Huq S, Yatsunenko T, Haque R, Mahfuz M, Alam MA, et al. Persistent gut microbiota immaturity in malnourished Bangladeshi children. Nature. 2014 Jun 19;510(7505):417–421. doi: 10.1038/nature13421. PubMed PMID: 24896187. Pubmed Central PMCID: 4189846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, Cheng J, et al. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science. 2013 Feb 1;339(6119):548–554. doi: 10.1126/science.1229000. PubMed PMID: 23363771. Pubmed Central PMCID: 3667500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mansfield LS, Gauthier DT, Abner SR, Jones KM, Wilder SR, Urban JF. Enhancement of disease and pathology by synergy of Trichuris suis and Campylobacter jejuni in the colon of immunologically naive swine. The American journal of tropical medicine and hygiene. 2003 Mar;68(3):70–80. PubMed PMID: 12685626. Epub 2003/04/11. eng. [PubMed] [Google Scholar]
  • 83.Li RW, Wu S, Li W, Navarro K, Couch RD, Hill D, et al. Alterations in the porcine colon microbiota induced by the gastrointestinal nematode Trichuris suis. Infection and immunity. 2012 Jun;80(6):2150–2157. doi: 10.1128/IAI.00141-12. PubMed PMID: 22493085. Pubmed Central PMCID: 3370577. Epub 2012/04/12. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wu S, Li RW, Li W, Beshah E, Dawson HD, Urban JF., Jr Worm burden-dependent disruption of the porcine colon microbiota by Trichuris suis infection. PLoS One. 2012;7(4):e35470. doi: 10.1371/journal.pone.0035470. PubMed PMID: 22532855. Pubmed Central PMCID: 3332011. Epub 2012/04/26. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Walk ST, Blum AM, Ewing SA, Weinstock JV, Young VB. Alteration of the murine gut microbiota during infection with the parasitic helminth Heligmosomoides polygyrus. Inflamm Bowel Dis. 2010 Nov;16(11):1841–1849. doi: 10.1002/ibd.21299. PubMed PMID: 20848461. Pubmed Central PMCID: 2959136. Epub 2010/09/18. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Rausch S, Held J, Fischer A, Heimesaat MM, Kuhl AA, Bereswill S, et al. Small intestinal nematode infection of mice is associated with increased enterobacterial loads alongside the intestinal tract. PLoS One. 2013;8(9):e74026. doi: 10.1371/journal.pone.0074026. PubMed PMID: 24040152. Pubmed Central PMCID: 3769368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Reynolds LA, Smith KA, Filbey KJ, Harcus Y, Hewitson JP, Redpath SA, et al. Commensal-pathogen interactions in the intestinal tract: lactobacilli promote infection with, and are promoted by, helminth parasites. Gut Microbes. 2014 Jul 1;5(4):522–532. doi: 10.4161/gmic.32155. PubMed PMID: 25144609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wescott RB. Experimental Nematospiroides dubius infection in germfree and conventional mice. Experimental parasitology. 1968 Apr;22(2):245–249. doi: 10.1016/0014-4894(68)90099-4. PubMed PMID: 5652501. Epub 1968/04/01. eng. [DOI] [PubMed] [Google Scholar]
  • 89.Hayes KS, Bancroft AJ, Goldrick M, Portsmouth C, Roberts IS, Grencis RK. Exploitation of the intestinal microflora by the parasitic nematode Trichuris muris. Science. 2010 Jun 11;328(5984):1391–1394. doi: 10.1126/science.1187703. PubMed PMID: 20538949. Epub 2010/06/12. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Plieskatt JL, Deenonpoe R, Mulvenna JP, Krause L, Sripa B, Bethony JM, et al. Infection with the carcinogenic liver fluke Opisthorchis viverrini modifies intestinal and biliary microbiome. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2013 Nov;27(11):4572–4584. doi: 10.1096/fj.13-232751. PubMed PMID: 23925654. Pubmed Central PMCID: 3804743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Lee SC, Tang MS, Lim YA, Choy SH, Kurtz ZD, Cox LM, et al. Helminth colonization is associated with increased diversity of the gut microbiota. PLoS Negl Trop Dis. 2014 May;8(5):e2880. doi: 10.1371/journal.pntd.0002880. PubMed PMID: 24851867. Pubmed Central PMCID: 4031128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cooper P, Walker AW, Reyes J, Chico M, Salter SJ, Vaca M, et al. Patent human infections with the whipworm, Trichuris trichiura, are not associated with alterations in the faecal microbiota. PLoS One. 2013;8(10):e76573. doi: 10.1371/journal.pone.0076573. PubMed PMID: 24124574. Pubmed Central PMCID: 3790696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Maizels RM, Bundy DA, Selkirk ME, Smith DF, Anderson RM. Immunological modulation and evasion by helminth parasites in human populations. Nature. 1993 Oct 28;365(6449):797–805. doi: 10.1038/365797a0. PubMed PMID: 8413664. Epub 1993/10/28. eng. [DOI] [PubMed] [Google Scholar]
  • 94.Summers RW, Elliott DE, Qadir K, Urban JF, Thompson R, Weinstock JV. Trichuris suis seems to be safe and possibly effective in the treatment of inflammatory bowel disease. Am J Gastroenterol. 2003;98(9):2034–2041. doi: 10.1111/j.1572-0241.2003.07660.x. [DOI] [PubMed] [Google Scholar]
  • 95.Summers RW, Elliott DE, Urban JF, Jr, Thompson R, Weinstock JV. Trichuris suis therapy in Crohn's disease. Gut. 2005 Jan;54(1):87–90. doi: 10.1136/gut.2004.041749. PubMed PMID: 15591509. Pubmed Central PMCID: 1774382. Epub 2004/12/14. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Summers RW, Elliott DE, Urban JF, Jr, Thompson RA, Weinstock JV. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology. 2005 Apr;128(4):825–832. doi: 10.1053/j.gastro.2005.01.005. PubMed PMID: 15825065. eng. [DOI] [PubMed] [Google Scholar]
  • 97.Jouvin MH, Kinet JP. Trichuris suis ova: Testing a helminth-based therapy as an extension of the hygiene hypothesis. J Allergy Clin Immunol. 2012 Jul;130(1):3–10. doi: 10.1016/j.jaci.2012.05.028. PubMed PMID: 22742834. Epub 2012/06/30. eng. [DOI] [PubMed] [Google Scholar]
  • 98.Fleming J, Isaak A, Lee J, Luzzio C, Carrithers M, Cook T, et al. Probiotic helminth administration in relapsing–remitting multiple sclerosis: a phase 1 study. Multiple Sclerosis Journal. 2011 Jun 1;17(6):743–754. doi: 10.1177/1352458511398054. 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Fleming JO. Helminth therapy and multiple sclerosis. International journal for parasitology. 2013 Mar;43(3–4):259–274. doi: 10.1016/j.ijpara.2012.10.025. PubMed PMID: 23298637. [DOI] [PubMed] [Google Scholar]
  • 100.Correale J, Farez MF. The impact of environmental infections (parasites) on MS activity. Mult Scler. 2011 Oct;17(10):1162–1169. doi: 10.1177/1352458511418027. PubMed PMID: 21980148. Epub 2011/10/08. eng. [DOI] [PubMed] [Google Scholar]
  • 101.Bager P, Arnved J, Ronborg S, Wohlfahrt J, Poulsen LK, Westergaard T, et al. Trichuris suis ova therapy for allergic rhinitis: a randomized, double-blind, placebo-controlled clinical trial. J Allergy Clin Immunol. 2010 Jan;125(1):123–130. e1–e3. doi: 10.1016/j.jaci.2009.08.006. PubMed PMID: 19800680. Epub 2009/10/06. eng. [DOI] [PubMed] [Google Scholar]
  • 102.Bager P, Kapel C, Roepstorff A, Thamsborg S, Arnved J, Ronborg S, et al. Symptoms after ingestion of pig whipworm Trichuris suis eggs in a randomized placebo-controlled double-blind clinical trial. PLoS One. 2011;6(8):e22346. doi: 10.1371/journal.pone.0022346. PubMed PMID: 21829616. Pubmed Central PMCID: 3149054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Mortimer K, Brown A, Feary J, Jagger C, Lewis S, Antoniak M, et al. Dose-ranging study for trials of therapeutic infection with Necator americanus in humans. The American journal of tropical medicine and hygiene. 2006 Nov;75(5):914–920. PubMed PMID: 17123987. eng. [PubMed] [Google Scholar]
  • 104.Feary JR, Venn AJ, Mortimer K, Brown AP, Hooi D, Falcone FH, et al. Experimental hookworm infection: a randomized placebo-controlled trial in asthma. Clin Exp Allergy. 2010 Feb;40(2):299–306. doi: 10.1111/j.1365-2222.2009.03433.x. PubMed PMID: 20030661. Pubmed Central PMCID: 2814083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Daveson AJ, Jones DM, Gaze S, McSorley H, Clouston A, Pascoe A, et al. Effect of hookworm infection on wheat challenge in celiac disease--a randomised double-blinded placebo controlled trial. PLoS One. 2011;6(3):e17366. doi: 10.1371/journal.pone.0017366. PubMed PMID: 21408161. Pubmed Central PMCID: 3050888. Epub 2011/03/17. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gaze S, McSorley HJ, Daveson J, Jones D, Bethony JM, Oliveira LM, et al. Characterising the mucosal and systemic immune responses to experimental human hookworm infection. PLoS Pathog. 2012 Feb;8(2):e1002520. doi: 10.1371/journal.ppat.1002520. PubMed PMID: 22346753. Pubmed Central PMCID: 3276555. Epub 2012/02/22. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.McSorley HJ, Gaze S, Daveson J, Jones D, Anderson RP, Clouston A, et al. Suppression of inflammatory immune responses in celiac disease by experimental hookworm infection. PLoS One. 2011;6(9):e24092. doi: 10.1371/journal.pone.0024092. PubMed PMID: 21949691. Pubmed Central PMCID: 3174943. Epub 2011/09/29. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Croese J, Giacomin P, Navarro S, Clouston A, McCann L, Dougall A, et al. Experimental hookworm infection and gluten microchallenge promote tolerance in celiac disease. J Allergy Clin Immunol. 2014 Aug 29; doi: 10.1016/j.jaci.2014.07.022. PubMed PMID: 25248819. [DOI] [PubMed] [Google Scholar]
  • 109.Broadhurst MJ, Ardeshir A, Kanwar B, Mirpuri J, Gundra UM, Leung JM, et al. Therapeutic helminth infection of macaques with idiopathic chronic diarrhea alters the inflammatory signature and mucosal microbiota of the colon. PLoS Pathog. 2012 Nov;8(11):e1003000. doi: 10.1371/journal.ppat.1003000. PubMed PMID: 23166490. Epub 2012/11/21. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.McKenna P, Hoffmann C, Minkah N, Aye PP, Lackner A, Liu Z, et al. The macaque gut microbiome in health, lentiviral infection, and chronic enterocolitis. PLoS Pathog. 2008 Feb 8;4(2):e20. doi: 10.1371/journal.ppat.0040020. PubMed PMID: 18248093. Pubmed Central PMCID: 2222957. Epub 2008/02/06. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Fallon PG, Mangan NE. Suppression of TH2-type allergic reactions by helminth infection. Nature reviews. 2007 Mar;7(3):220–230. doi: 10.1038/nri2039. PubMed PMID: 17318233. [DOI] [PubMed] [Google Scholar]
  • 112.Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet. 2011 Nov 19;378(9805):1812–1823. doi: 10.1016/S0140-6736(11)61539-0. PubMed PMID: 22098854. [DOI] [PubMed] [Google Scholar]
  • 113.Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000 Feb 3;403(6769):503–511. doi: 10.1038/35000501. PubMed PMID: 10676951. Epub 2000/02/17. eng. [DOI] [PubMed] [Google Scholar]
  • 114.Baehner FL, Lee M, Demeure MJ, Bussey KJ, Kiefer JA, Barrett MT. Genomic signatures of cancer: basis for individualized risk assessment, selective staging and therapy. J Surg Oncol. May 1;103(6):563–573. doi: 10.1002/jso.21838. PubMed PMID: 21480251. Epub 2011/04/12. eng. [DOI] [PubMed] [Google Scholar]
  • 115.Bharti A, Ma PC, Salgia R. Biomarker discovery in lung cancer--promises and challenges of clinical proteomics. Mass spectrometry reviews. 2007 May-Jun;26(3):451–466. doi: 10.1002/mas.20125. PubMed PMID: 17407130. eng. [DOI] [PubMed] [Google Scholar]
  • 116.Liu JJ, Cutler G, Li W, Pan Z, Peng S, Hoey T, et al. Multiclass cancer classification and biomarker discovery using GA-based algorithms. Bioinformatics (Oxford, England) 2005 Jun 1;21(11):2691–2697. doi: 10.1093/bioinformatics/bti419. PubMed PMID: 15814557. eng. [DOI] [PubMed] [Google Scholar]
  • 117.Broadhurst MJ, Leung JM, Kashyap V, McCune JM, Mahadevan U, McKerrow JH, et al. IL-22+ CD4+ T cells are associated with therapeutic trichuris trichiura infection in an ulcerative colitis patient. Sci Transl Med. 2010 Dec 1;2(60):60ra88. doi: 10.1126/scitranslmed.3001500. PubMed PMID: 21123809. Epub 2010/12/03. eng. [DOI] [PubMed] [Google Scholar]
  • 118.Hunter MM, Wang A, McKay DM. Helminth infection enhances disease in a murine TH2 model of colitis. Gastroenterology. 2007 Apr;132(4):1320–1330. doi: 10.1053/j.gastro.2007.01.038. PubMed PMID: 17408663. Epub 2007/04/06. eng. [DOI] [PubMed] [Google Scholar]
  • 119.Wang A, Fernando M, Leung G, Phan V, Smyth D, McKay DM. Exacerbation of oxazolone colitis by infection with the helminth Hymenolepis diminuta: involvement of IL-5 and eosinophils. Am J Pathol. 2010 Dec;177(6):2850–2859. doi: 10.2353/ajpath.2010.100537. PubMed PMID: 21037078. Pubmed Central PMCID: 2993289. Epub 2010/11/03. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Garrett WS, Gordon JI, Glimcher LH. Homeostasis and inflammation in the intestine. Cell. 2010 Mar 19;140(6):859–870. doi: 10.1016/j.cell.2010.01.023. PubMed PMID: 20303876. Pubmed Central PMCID: 2845719. Epub 2010/03/23. eng. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Girgis NM, Gundra UM, Loke P. Immune regulation during helminth infections. PLoS Pathog. 2013 Apr;9(4):e1003250. doi: 10.1371/journal.ppat.1003250. PubMed PMID: 23637593. Pubmed Central PMCID: 3630086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Osborne LC, Monticelli LA, Nice TJ, Sutherland TE, Siracusa MC, Hepworth MR, et al. Coinfection. Virus-helminth coinfection reveals a microbiota-independent mechanism of immunomodulation. Science. 2014 Aug 1;345(6196):578–582. doi: 10.1126/science.1256942. PubMed PMID: 25082704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, et al. Enterotypes of the human gut microbiome. Nature. 2011 May 12;473(7346):174–180. doi: 10.1038/nature09944. PubMed PMID: 21508958. Pubmed Central PMCID: 3728647. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES