Skip to main content
Gut Microbes logoLink to Gut Microbes
. 2017 Feb 10;8(3):238–252. doi: 10.1080/19490976.2017.1290757

Intestinal microbiota, fecal microbiota transplantation, and inflammatory bowel disease

Alexa R Weingarden 1, Byron P Vaughn 1,
PMCID: PMC5479396  PMID: 28609251

ABSTRACT

Inflammatory bowel disease (IBD) is a complex set of diseases that lead to chronic inflammation in the gastrointestinal tract. Although the etiology of IBD is not fully understood, it is well-known that the intestinal microbiota is associated with the development and maintenance of IBD. Manipulation of the gut microbiota, therefore, may represent a target for IBD therapy. Fecal microbiota transplantation (FMT), where fecal microbiota from a healthy donor is transplanted into a patient's GI tract, is already a successful therapy for Clostridium difficile infection. FMT is currently being explored as a potential therapy for IBD as well. In this review, the associations between the gut microbiota and IBD and the emerging data on FMT for IBD will be discussed.

KEYWORDS: Inflammatory Bowel Disease, Crohn's disease, ulcerative colitis, fecal microbiota transplant

Introduction

Inflammatory bowel disease (IBD) affects over 1 million individuals in the United States alone, and the incidence of these diseases in both the US and in developed countries worldwide continues to grow.1,2 The current paradigm of the pathophysiology of IBD is an inappropriate immune response to the microbiota in a genetically susceptible individual.3,4 IBD is grouped into predominantly phenotypic patterns based on the location of inflammation: in Crohn's disease (CD), the inflammation can be in any part of the intestine, while in ulcerative colitis (UC) the inflammation is limited to the colon. In IBD, an abnormal intestinal microbiota (dysbiosis) is clearly associated with certain disease phenotypes, and may be a causal or synergistic factor in perpetuating chronic inflammation. Thus, manipulating the intestinal microbiota represents a potential treatment of IBD.2 One form of manipulating the microbiota is through fecal microbiota transplantation (FMT), where fecal microbiota from a healthy donor are transplanted into the distal GI tract of a patient. FMT has already emerged as a successful therapy for Clostridium difficile infection,5-7 and is currently being explored as a potential treatment of IBD.8-10 This review will outline the associations of IBD and the gut microbiota, and then discuss the current data on fecal microbiota transplantation in IBD.

Associations between the intestinal microbiota and IBD

Microbiota and early intestinal immune system development

Evidence in both human and mouse studies strongly suggests that early intestinal immune system development is highly dependent on the intestinal microbiota,11 which in turn may impact susceptibility to IBD. It is well-established that germ-free animals have an under-developed intestinal immune system compared with conventionally raised animals, characterized by smaller and fewer Peyer's patches, mesenteric lymph nodes, and isolated lymphoid follicles.12,13 Germ-free mice also lack certain helper T cell subtypes in their intestinal tracts.14 Although many of these changes can be ameliorated by the later introduction of specific pathogen-free microbiota,15 transcriptional profiles in the jejunum and colon of these mice remain altered compared to conventionally-raised mice,16 suggesting that there is a critical period during which exposure to microbes informs appropriate immune and mucosal development.

Appropriate exposure to a healthy microbiota early in life appears to be important in the resistance of chemical induced colitis later in life. In germ-free mice, there is an accumulation of invariant natural killer T (iNKT) cells in the lamina propria of the colon, with subsequent worsening of colitis upon exposure to oxazolone compared with conventionally-raised animals.17 Although introduction of a conventional microbiota to neonatal germ-free mice protected animals against both the accumulation of iNKT cells and worsened colitis, these effects were not seen following conventionalization of adult animals. At least in this model, early exposure to microbiota is critical to normal immune development and protection against colitis.

Recent work has demonstrated that maternal microbial exposure may also play a role in immune system development and protection against inflammation.18 Transient colonization (gestational only) of otherwise germ-free pregnant mice induced innate immune development in subsequent germ-free offspring, which in turn decreased the inflammatory response of pups to microbial molecules. These findings suggest that microbial manipulation in utero may already set the stage for inflammation later in life.

Studies in humans have also suggested that early-life manipulation of the gut microbiota, primarily through the use of antibiotics, is associated with altered susceptibility to IBD. Case-control analyses demonstrate that early exposure to antibiotics increases the risk of IBD later in life.19-21 Interestingly this risk appears to be the highest for the first year in life. Similarly, another study noted that early-childhood infectious diseases, including gastroenteritis and respiratory infections, were protective against the development of IBD.22 While the associations are compelling, it remains unclear if these microbial changes are causal for IBD, or rather markers for an underlying immune dysfunction that eventually manifests as IBD. However, overall, it appears that early-life microbial exposures in mice and humans influence the development of the intestinal immune system in such a way to promote or protect against the development of IBD.

NOD2 – bridging the immune system and commensal bacteria

One of the clearest human genetic associations with IBD is the nucleotide-binding oligomerization domain-containing protein 2 (NOD2), an intracellular immune receptor for components of the bacterial cell wall.23,24 NOD2 polymorphisms confer an increased risk for the development of Crohn's disease.23,25,26 Mice lacking Nod2, as well as humans with NOD2 mutations, have an altered microbiota.27-30 In both humans and animal models, a lack of functional NOD2 results in an increased abundance of the Bacteroidetes phylum, one of the major bacterial phyla found in the gut.28 Interestingly, Nod2-deficient mice do not spontaneously develop colitis..31 However, compared with wild type mice, Nod2-deficient mice have increased susceptibility to dextran sodium sulfate (DSS)-induced colitis, which is both transmissible to wild-type animals via cohousing and ameliorated by fecal transplantation from wild-type donors.32 This suggests that the risk of colitis from NOD2 mutations is from the resulting dysbiosis and can be ameliorated or worsened by altering the microbiota.

Alterations in other key immune pathways may also explain why individuals with NOD2 mutations are predisposed to, but do not always develop, IBD. NOD2 is important in the clearance of intracellular pathogens through autophagy via interaction with ATG16L1.33,34 Notably in Crohn's disease with NOD2 variants, this effect is absent, resulting in failure to induce autophagy for intracellular Salmonella enterica ser. Typhimurium, adherent-invasive Escherichia coli, and Shigella flexneri.25 Again, it appears that the underlying mutation alone does not cause inflammation, but rather the resulting changes to the microbiota; in this case through failure to clear potentially pathogenic microbes

The intestinal microbiota is required for mouse models of colitis

The majority of mouse models of colitis require a microbiota to develop intestinal inflammation.35 Although no mouse model can fully replicate the complex pathophysiology of human IBD, several models that spontaneously develop inflammation when housed under typical conditions show no evidence of inflammation when raised under germ-free conditions.36-38 These findings support the role of the intestinal microbiota in the development of inflammation and IBD.

The importance of the intestinal microbiota in inflammation in animal models is demonstrated via transfer of inflammation-associated microbial populations into germ-free mice. In some mouse models of intestinal inflammation, the transfer of fecal pellets from conventionally raised or specific pathogen free animals with an IBD-like phenotype into germ-free animals recapitulates both the phenotype and the dysbiotic microbiota.39,40 In certain murine models (including Nod2-deficient mice as above,32 T-bet and Rag-2 deficient (TRUC) mice,41 and Nlrp-6 deficient mice)42 colitis can be reproduced in conventionally-raised wild-type animals by cohousing, an effect presumably mediated by the microbiota. Similarly, Casp3/11-deficient mice, which are protected against DSS-induced colitis, lose this protection when cohoused with wild-type mice,43 while cohousing Il10-deficient mice with Apoe-deficient mice worsens colitis in IL-10 knockout animals.44 This provides evidence not only of the significance of the microbiota in intestinal inflammation, but also suggests that this inflammation is transmissible.

Specific bacteria are associated with IBD

Certain bacteria are implicated in the development of intestinal inflammation in animal models. For example, Proteus mirabilis and Klebsiella pneumoniae correlate with colitis in T-bet−/− x Rag2−/− mice, a mouse model of ulcerative colitis.45 Additionally, these bacteria may be associated with maternal transmission of disease, potentially indicating a causal link. Recent work found that Bilophila wadsworthia, a typically low-abundance commensal organism, is associated with colitis in Il10−/− mice.46 In this study, an increase in dietary milk fat increased the proportion of taurine-conjugated bile acids in the colon, subsequently increasing organic sulfur compounds available to sulfite-reducing microbes including B. wadsworthia, which resulted in an expansion of this organism. B. wadsworthia can activate dendritic cells to promote a Th1-mediated colitis, likely explaining why its expansion induced colitis in a susceptible host, and also linking diet to colonic inflammation. Another recent study identified Atopobium parvulum as a driver of colitis via altered metabolism of H2S, and subsequent amelioration of colitis with the H2S scavenger bismuth.47 Finally, supernatant from Fusobacterium varium cultures, isolated from the colonic mucosa of UC patients, can induce colonic ulcer formation in mice.48 These findings indicate that individual commensal organisms have the capability of inducing colitis in certain mouse models, although the translation to human IBD is less apparent.

Despite an extensive search, no single specific pathogen appears to cause IBD.49 Certainly some infections cause a phenotype similar to IBD (e.g. intestinal tuberculosis, Campylobacter jejuni, Yersinia enterocolitica and others),50,51 raising the possibility that some subsets of IBD are actually unrecognized intestinal infections. Some potential examples include, Mycobacterium avium subsp. paratuberculosis (MAP) and adherent-invasive E. coli (AIEC), both of which are associated with Crohn's disease. In one study, peripheral blood from patients with active Crohn's disease had MAP DNA prevalence of 68%, while those with Crohn's in any stage had E. coli 80% of the time.52 In subjects whose sera tested positive for antigens against MAP, early uncontrolled trials suggested antibiotics against MAP could induce symptomatic improvement.53-55 A large randomized controlled in trial from Australia found an early clinical benefit of antibiotics for MAP in addition to steroids compared to placebo (week 16, 66% v. 50%, p = 0.02).56 Unfortunately there were no changes in inflammatory parameters, endoscopic endpoints, or maintenance of remission As of this writing, a Phase III trial is underway to further explore the use of antibiotics against MAP in Crohn's disease (NCT01951326).

Helminthic infections and IBD

Non-bacterial microbes, notably parasites, may also have a role to play in the development of IBD. Early epidemiological data linked the decreased incidence of parasitic infections with an increase in Crohn's disease, positing that the mechanism behind this observation may be a shift toward T helper type 1 (Th1) cytokine production.57 Several studies in mice have demonstrated amelioration or prevention of colitis following exposure to a variety of helminths, including Heligmosomoides polygyrus, Trichinella spiralis, and the parasite Schistosoma mansoni.58-61 These organisms may either directly alter the immune response of the host, or may act via modulation of the gut microbiota.62 For example, infection with H. polygyrus in mice appears to alter the intestinal microbiota.63

While some limited data in humans suggests that treatment with Trichuris suis may be a potential therapy for IBD,64,65 phase 2 clinical trials in Crohn's disease did not meet early end points and were stopped (NCT01576471 and NCT01279577).66 A study of Trichuris suis ova in ulcerative colitis failed to recruit sufficient subjects to draw meaningful conclusions (NCT01433471) and further trials in ulcerative colitis were halted (NCT01953354). Necator americanus, a hookworm, has also been studied in IBD.67 Of 9 patients who received hookwork treatment, 7 improved. Hookwork colonization seems to increase the diversity of the gut microbiota,68 although this effect has not always been reproducible.69 Certainly the idea of using a single agent to orchestrate a more diverse microbiota is appealing, as in general diversity is associated with health, however much more research is needed before this is trialed in human diseases.

Global shifts in the gut microbiota and IBD

In addition to associations with specific organisms, substantial work has found that the overall composition of the gut microbiota is highly altered in IBD. Typically, microbial diversity is substantially diminished in patients with IBD compared with healthy individuals.70-72 Furthermore, in contrast to healthy individuals, the fecal microbiota of patients with both Crohn's disease and ulcerative colitis contains a significantly lower proportion of the Bacteroidetes and Firmicutes phyla (particularly Clostridium), which are normally dominant in the human fecal microbiota,12 and a significantly higher proportion of the Proteobacteria phylum.70,71,73,74 Notably, these differences are present even in patients with Crohn's disease who are treatment-naïve, suggesting that intestinal dysbiosis in these patients is not a consequence of therapy, but rather a potential early change in IBD.72

Evidence from post-surgical patients who have been treated for Crohn's disease also suggests a causal relationship between gut microbiota composition and IBD. For example, diversion of the fecal stream via diverting loop ileostomy can prevent Crohn's disease recurrence, an effect that is abrogated upon restoration of bowel continuity.75 One study noted that Crohn's patients with mucosal-associated microbiota that were more similar to healthy individuals were more likely to remain in remission following surgery.76 Specific functions of the microbiota may also be associated with improved outcomes following resection for CD. Notably, the presence of mucosal bacteria associated with saccharolytic metabolism, including Bacteroides, Prevotella, and Parabacteroides species, has been correlated with increased remission compared with the presence of bacteria associated with fermentation and lactic acid production, such as Enterococcus and Veillonella.77 These studies demonstrate the importance of the global gut microbiota composition and function in the development and severity of IBD.

Clinical use of FMT for IBD

Therapeutic manipulation of the gut microbiota

Given these well-described changes in the gut microbiota in patients with IBD, it is unsurprising that several therapeutic strategies have focused on manipulation of the gut microbiota. Indeed, there is a large body of literature on the use of antibiotics and probiotics in IBD. While a full review of antibiotics and probiotics is beyond the scope of this article (especially given other excellent recent reviews),78,79 several points are highlighted to demonstrate the importance of microbiota manipulation in IBD. The use of antibiotics has variable success for the treating IBD, although in one pooled analysis there was a trend toward benefit, particularly in fistulizing or post-surgical Crohn's disease.80 In these 2 settings, it may be that secondary bacterial overgrowth (in an area of a sinus tract or anastomosis) may perpetuate chronic inflammation, in which case antibiotic therapy may ameliorate symptoms. Some studies suggest that specific antibiotics improve disease severity,81-83 although these studies are not always reproducible and often use symptomatic endpoints without measurement of inflammatory parameters. It is possible that decreasing the overall bacterial burden via antibiotics decreases symptoms such as diarrhea or bloating, which subsequently reduces disease activity scores, without necessarily improving the mucosal inflammation. Additionally, as IBD is typically characterized by reduced microbial diversity, it is counterintuitive to think that further reduction in bacterial diversity through antibiotics will reverse the underlying inflammatory process, especially given the associations between early-life antibiotic use and IBD as reviewed above.

An additional method of microbiota manipulation in IBD is the introduction of specific bacteria, or probiotics, in an attempt to control the growth of pathological organisms or shift the global composition of toward a healthier state. E. coli Nissle 1917 is a well-studied probiotic which has been shown to be as effective as mesalazine at maintaining remission in ulcerative colitis.84,85 Other individual probiotics with demonstrated efficacy in IBD include Lactobacillus GG, bifidobacteria strains, and the yeast Saccharomyces boulardii.86-88 One of the most promising probiotic supplements, VSL#3, is a set of 8 bacterial strains that significantly reduces disease severity and induces remission in patients with UC compared to the placebo.89,90 Additionally, VSL#3 can prevent pouchitis following total proctocolectomy and J-pouch formation.91 However, engraftment of probiotics is often poor as demonstrated by lack of detectable probiotic strains 2 weeks following cessation of intake.92

In contrast to both antibiotics and probiotics, fecal microbiota transplantation (FMT) may represent a more robust method of manipulating the gut microbiota as a therapy for patients with IBD. This procedure involves the transfer of processed feces from a donor into the GI tract of a recipient, and has been successfully used to treat infection with C. difficile for nearly 60 y.93 Unlike antibiotics, FMT increases the diversity of fecal bacterial populations in recipients,94,95 likely contributing to its success in C. difficile infection. Furthermore, unlike probiotics, evidence suggests that FMT results in long-term engraftment in recipients with C. difficile infection,96 The scale and content of FMT also varies considerably from probiotic therapy, as donor fecal material contains approximately 1011 bacterial cells per gram of stool, in addition to viruses, fungi, and archaea.97 All together, these factors suggest that FMT may be a more promising therapy for IBD than either antibiotics or probiotics. Below we discuss current evidence available on the use of FMT for IBD.

Ulcerative colitis

Early reports of FMT for UC suggested a reversal of disease in selected patients. The first report of FMT use in UC, in 1989, was of a single patient with no endoscopic or histopathology follow-up.98 The authors later noted in a review that the initial patient treated remained endoscopically and histologically disease free for over 20 y.99 Subsequently, a series of 6 patients who received daily enema administration of donor fecal material for 5 d resulted in cessation of all UC-related medications, and over 1–13 y of follow-up had no clinical, endoscopic, or histologic evidence of UC.100 Notably all patients had at least left sided ulcerative colitis and at least 5 y duration of disease. Later follow-up from this same center reported substantial success, with over 90% of a cohort of 62 patients achieving complete or partial remission.101 Further case series in children with UC demonstrated that FMT was safe and potentially effective in improving disease status.102,103

Several recent case series in adults have shown mixed results for FMT as a treatment of UC. Generally, a few subjects improved, although none reached remission, and the benefit appeared to be short-term.104-108 Interestingly, one study of 12 patients found that the clinical benefit of FMT was associated with a higher proportion of butyrate-producing bacteria in their feces following transplant, suggesting a possible mechanism behind those procedures that are successful.109 Two essential issues with these small case series are single FMT infusions and selection bias in recruitment; patients can be very motivated for an FMT trial to the point of compromising research protocols, as a failed trial of FMT in UC noted.110

Given the mixed success of these small studies, 2 recent randomized trials evaluated the clinical efficacy of FMT in UC. A group from McMaster University used weekly retention enemas with donor fecal material or placebo for 6 weeks.111 Unfortunately, the Data and Safety Monitoring Board discontinued the trial based on futility to reach the primary end point at a planned interim analysis. At that time, 4 of 27 subjects (14.8%) in the FMT arm and 2 of 26 (7.7%) in the placebo arm were in clinical remission. Patients already enrolled in the trial were allowed to complete the study and ultimately 9 of 38 (24%) in the FMT arm were in remission versus 2 of 37 (5%) in the placebo (risk difference: 17%, 95% CI: 2 – 33%). Other end points such as symptomatic improvement or changes in quality of life were similar between the 2 groups. Interestingly in this trial, nearly 40% of the patients in remission following FMT received material from a single donor. Additionally, those with UC for less than one year were more likely to achieve remission. Following FMT all subjects demonstrated increased diversity and similarity to the donors. Overall, the results suggest improved remission rates for patients treated with FMT, possibly dependent on donor fecal composition, the use of multiple FMTs, and early treatment of UC.

A second recent randomized study also investigated the use of FMT in UC. Investigators from the Academic Medical Center in Amsterdam performed FMT with nasoduodenal tube placement following full bowel lavage twice over a 3 week period.112 At the second interim, analysis by the DSMB recommended cessation of the trial due to futility. In the intention to treat analysis, at week 12, 30% of those who received FMT from a donor were in remission vs. 20% of those who received placebo FMT (autologous stool transplant). Regardless of treatment group, responders demonstrated an increase in diversity of fecal microbiota at week 12, whereas non-responders in either group did not have any change in diversity.

These 2 trials differed in dose, frequency, and administration of FMT, as well as type of placebo used, making direct comparisons difficult. However, in light of the results available from case series, these findings suggest that donor selection, disease duration, and successful engraftment of microbiota may all be significant factors affecting clinical outcome following FMT. A recently published abstract describing a third blinded study of 81 patients treated with an intense, multiple FMT regimen (fecal enemas 5 d per week for 8 weeks) also noted significantly improved remission in patients treated with donor fecal material (27% vs. 8% of placebo-treated controls, p = 0.02), suggesting that the number of transplants may also play a role in the success of this therapy.113 Clearly, more work is needed to understand how the microbiota influences mucosal inflammation in ulcerative colitis.

Crohn's disease

Data on FMT for Crohn's disease is somewhat more limited than UC. Case reports have demonstrated mixed results with some suggesting clinical and endoscopic remission while others demonstrated no effect.107,114-116 The earliest report of FMT for Crohn's disease noted symptomatic improvement in one patient over 4 months.117 A more recent case report of a patient with severe, complicated CD also noted successful treatment with FMT.118 A cohort of 30 patients with refractory mid-gut CD (defined as Harvey Bradshaw Index [HBI] ≥ 7) demonstrated 77% clinical remission at one month following a single FMT via nasoduodenal route.119 A small but significant benefit was noted in hemoglobin and albumin at 3 months post-FMT in this study. FMT may also be a potential treatment of pediatric CD, as one recent case series noted remission in 5 of 9 patients (56%) after FMT, with 7 of 9 patients (78%) demonstrating engraftment of donor microbiota.120 Interestingly, although one study did not find a significant decrease in Crohn's disease severity following FMT, patients reported significantly increased quality of life scores after the procedure.108

Prior work from one of us for FMT in active Crohn's disease (defined as HBI ≥ 5) had mixed results.121 Of the 20 subjects enrolled for FMT, 19 had complete follow-up data. While most subjects improved post-FMT, the clinical course was variable with one subject with severe disease before FMT proceeding to colectomy following FMT. Similar to the randomized trials in UC, microbiota diversity increased for clinical responders. Additionally, clinical responders assumed more of their donor profile as measured by week 4 Bray Curtis similarity index. However, the small number and lack of a control group limit the ability to draw conclusions about efficacy in this study. Overall, these findings suggest that FMT may also be a potential therapy for CD. However, much more work is needed to assess both clinical efficacy as well as changes to the gut microbiota of these patients.

Pouchitis

Patients with UC who undergo colectomy may develop inflammation of the surgically-created ileal pouch, a condition known as pouchitis. Although created from small intestinal tissue, the microbiota of the pouch frequently resembles that of the colon.122 Like UC itself, however, patients with pouchitis may have a pouch microbiota that is distinct from that of patients without active inflammation.122,123 Although this suggests that FMT may also be a treatment of pouchitis, small studies have failed to demonstrate remission following FMT.124 One key limitation in FMT for pouchitis is the size of the pouch, which may limit engraftment of a donor microbiota. Although current data are not promising, more data are necessary to understand the effects of FMT on pouchitis.

Systematic reviews of FMT in IBD

Given the overall limited data, some investigators have pooled available data to attempt to discern a signal from the noise. One review found promising results for FMT in IBD, with the majority of patients achieving reduction of symptoms, cessation of IBD medications, and/or clinical remission.125 Another review found similar results, with 78% of patients, both pediatric and adult, achieving remission.126 However, these studies only included data from case reports or series, which are not controlled and subject to publication bias. A more recent systematic review which included a randomized trial for FMT in IBD noted less promising results, with 45% of patients achieving remission (22% of patients with UC, 60.5% of patients with CD).127 The variability in these findings likely reflects the underlying heterogeneity in the primary studies of FMT for IBD, which in turn may be related to several factors such as donor selection, FMT preparation and delivery, and length of diagnosis or severity of disease.

Safety of FMT

In addition to efficacy, safety of the procedure is an important factor in assessing the usefulness of FMT for IBD. Although minimal side effects are typically reported for FMT for C. difficile infection, one case report did note a UC flare following FMT in a patient who was previously in remission for over 20 y before the procedure.128 Similarly, a recent case series noted that 2.9% of patients treated for C. difficile who had underlying IBD required hospitalization for IBD flare after FMT.129 This suggests that the potential for adverse effects in FMT may be greater in IBD than for recurrent C. difficile infection. In studies examining FMT for IBD without C. difficile infection, side effects are typically mild and self-limiting. In one systematic review, no serious adverse events were reported in any included study; all mild to moderate adverse events (such as fever, abdominal tenderness, and CRP elevation) were self-limiting outside a single patient with fever, who was successfully treated with acetaminophen.127 Another study in pediatric patients reported only mild adverse events with one case of moderate abdominal pain, all of which were self-limiting.120 These findings suggest that FMT for IBD is a safe procedure although the numbers in each trial are small. Given the decreased efficacy of FMT for C. difficile infection in patients with underlying IBD,130 it is possible that the increased side-effects noted in this population are confounded by the presence of both diseases.

Future directions

With increasing numbers of studies, including randomized trials, of FMT for IBD (39 registered studies on clinicaltrials.gov as of this writing), we will be faced with more clinical data in an area where a mechanistic understanding is lacking. At the moment, FMT cannot be considered a consistent therapy across trials. In fact, trials to date vary on the method of FMT delivery, bacterial dose, method of stool filtration, frequency of administration, and do not control for other donor factors such as diet. This diversity may ultimately be beneficial for deriving a mechanistic underpinning for FMT in IBD, though currently it limits our interpretation of FMT as a clinical success or failure.

One essential issue to clarify and define is that of engraftment of a microbiota. A common theme among published studies is that those who achieve normalization of the gut microbiota, or are more similar to their donor, typically experience improved clinical outcomes.107,109,112,120,121 A key issue moving forward, therefore, will be to understand how donor microbiota engraftment can be improved, as this alone may lead to increased success with FMT. Perhaps one reason why some studies have had more encouraging results is due to multiple FMTs, which suggests those studies may have achieved a higher rate of engraftment.111

Related to the issue of engraftment is the question of whether or not a so-called “Western” microbiota may even be useful in the treatment of IBD. The prevalence of IBD in developed countries is typically much higher, and increasing at a much steeper rate, compared to developing nations,131 although IBD appears to be increasing in these countries as well.132 Additionally the gut microbiota of individuals in developed countries is substantially different compared to individuals in primitive hunter-gatherer societies.133-135 These changes are usually characterized as a loss of bacterial diversity, suggesting a possible link between the gut microbiota and the rising prevalence of IBD and other autoimmune and inflammatory conditions in developed countries. Recent data from a mouse model suggests that a shift toward a more “Western” diet results in extinction of certain bacteria within the gut,136 further suggesting that a “Western” microbiota may lack key organisms which might be critical to preventing or treating IBD. All of these findings imply that “Western” fecal donors may overall be sub-optimal for treating IBD patients.

Beyond the issues of engraftment and donor choice, a critical issue will be to understand the functions of the microbiota following FMT in those patients who achieve remission. Several studies have suggested that patients with IBD have decreased populations of bacteria capable of producing butyrate, a known anti-inflammatory molecule also used as an energy source by the colonic epithelium.70,137-139 One study has already found that IBD patients who responded to FMT replenished these butyrate-producing bacteria, suggesting that this may a key mechanism to target in the future.109 Ideally, future work will lead to targeted microbial therapies aimed at restoring intestinal microbiota functions currently diminished in patients with IBD.

Lastly, the issue of cause and effect is still an unanswered question. Despite the fact that treatment-naïve patients with IBD have dysbiosis,72 it is still not fully understood if the effects of the early inflammation cause dysbiosis or if the dysbiosis precedes inflammation. Perhaps as IBD appears to be an abnormal immune response to the microbiota, the microbial composition is irrelevant as any generic microbial stimulus will perpetuate inflammation. The truth is likely in between a cure and no effect at all; and while overall the use of FMT for IBD is promising, clearly the course will not be the same as FMT for recurrent C. difficile infection. Determining microbial biomarkers (specific bacterial taxa, pathways, or metabolites) associated with IBD will be essential to establish key outcomes for clinical trials. Continued clinical trials of FMT in IBD without teasing out these underlying mechanisms will continue to result in variable and difficult-to-interpret results. Beyond the potential therapeutic application, microbial patterns may allow us to identify those at risk for IBD, predict phenotypes or disease courses, and perhaps even predict complications of IBD, such as PSC or dysplasia. In conclusion, IBD is clearly associated with dysbiosis, and the available data suggest there may be a role for manipulating the intestinal microbiota in the treatment of these devastating diseases.

Disclosure of potential conflicts of interest

ARW has no conflicts of interest to report.

BPV has received research support for Genetech-Roche and Takeda and speaking/consulting fees from Abbvie and Janssen.

Funding

Research reported in this publication was supported by the National Center for Advancing Translational Sciences of the National Institutes of Health Award Number UL1TR000114. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

References

  • [1].Ventham NT, Kennedy NA, Nimmo ER, Satsangi J. Beyond gene discovery in inflammatory bowel disease: the emerging role of epigenetics. Gastroenterology 2013; 145:293-308; PMID:23751777; http://dx.doi.org/ 10.1053/j.gastro.2013.05.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Miyoshi J, Chang EB. The gut microbiota and inflammatory bowel diseases. Transl Res 2016; 179:38-48; PMID:27371886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Loddo I, Romano C. Inflammatory Bowel Disease: Genetics, Epigenetics, and Pathogenesis. Front Immunol 2015; 6:551; PMID:26579126; http://dx.doi.org/ 10.3389/fimmu.2015.00551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med 2009; 361:2066-78; PMID:19923578; http://dx.doi.org/ 10.1056/NEJMra0804647 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Hamilton MJ, Weingarden AR, Sadowsky MJ, Khoruts A. Standardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infection. Am J Gastroenterol 2012; 107:761-7; PMID:22290405; http://dx.doi.org/ 10.1038/ajg.2011.482 [DOI] [PubMed] [Google Scholar]
  • [6].van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, Visser CE, Kuijper EJ, Bartelsman JF, Tijssen JG, et al.. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med 2013; 368:407-15; PMID:23323867; http://dx.doi.org/ 10.1056/NEJMoa1205037 [DOI] [PubMed] [Google Scholar]
  • [7].Khoruts A, Weingarden AR. Emergence of fecal microbiota transplantation as an approach to repair disrupted microbial gut ecology. Immunol Lett 2014; 162:77-81; PMID:25106113;http://dx.doi.org/ 10.1016/j.imlet.2014.07.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Damman CJ, Miller SI, Surawicz CM, Zisman TL. The microbiome and inflammatory bowel disease: is there a therapeutic role for fecal microbiota transplantation? Am J Gastroenterol 2012; 107:1452-9; PMID:23034604; http://dx.doi.org/ 10.1038/ajg.2012.93 [DOI] [PubMed] [Google Scholar]
  • [9].Dasgupta S, Kasper DL. Relevance of commensal microbiota in the treatment and prevention of inflammatory bowel disease. Inflamm Bowel Dis 2013; 19:2478-89; PMID:23846489;http://dx.doi.org/ 10.1097/MIB.0b013e318297d884 [DOI] [PubMed] [Google Scholar]
  • [10].Kahn SA, Gorawara-Bhat R, Rubin DT. Fecal bacteriotherapy for ulcerative colitis: patients are ready, are we? Inflamm Bowel Dis 2012; 18:676-84; PMID:21618362; http://dx.doi.org/ 10.1002/ibd.21775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Abrams GD, Bauer H, Sprinz H. Influence of the normal flora on mucosal morphology and cellular renewal in the ileum. A comparison of germ-free and conventional mice. Lab Invest 1963; 12:355-64 [PubMed] [Google Scholar]
  • [12].Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature 2007; 449:804-10; PMID:17943116; http://dx.doi.org/ 10.1038/nature06244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 2009; 9:313-23; PMID:19343057; http://dx.doi.org/ 10.1038/nri2515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al.. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009; 139:485-98; PMID:19836068;http://dx.doi.org/ 10.1016/j.cell.2009.09.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Crabbe PA, Nash DR, Bazin H, Eyssen H, Heremans JF. Immunohistochemical observations on lymphoid tissues from conventional and germ-free mice. Lab Invest 1970; 22:448-57; PMID:4911977 [PubMed] [Google Scholar]
  • [16].El Aidy S, Hooiveld G, Tremaroli V, Bäckhed F, Kleerebezem M. The gut microbiota and mucosal homeostasis: colonized at birth or at adulthood, does it matter? Gut Microbes 2013; 4:118-24; PMID:23333858; http://dx.doi.org/ 10.4161/gmic.23362 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Olszak T, An D, Zeissig S, Vera MP, Richter J, Franke A, Glickman JN, Siebert R, Baron RM, Kasper DL, et al.. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 2012; 336:489-93; PMID:22442383; http://dx.doi.org/ 10.1126/science.1219328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Gomez de Aguero M, Ganal-Vonarburg SC, Fuhrer T, Rupp S, Uchimura Y, Li H, Steinert A, Heikenwalder M, Hapfelmeier S, Sauer U, et al.. The maternal microbiota drives early postnatal innate immune development. Science 2016; 351:1296-302; PMID:26989247; http://dx.doi.org/ 10.1126/science.aad2571 [DOI] [PubMed] [Google Scholar]
  • [19].Shaw SY, Blanchard JF, Bernstein CN. Association between the use of antibiotics in the first year of life and pediatric inflammatory bowel disease. Am J Gastroenterol 2010; 105:2687-92; PMID:20940708; http://dx.doi.org/ 10.1038/ajg.2010.398 [DOI] [PubMed] [Google Scholar]
  • [20].Kronman MP, Zaoutis TE, Haynes K, Feng R, Coffin SE. Antibiotic exposure and IBD development among children: a population-based cohort study. Pediatrics 2012; 130:e794-803; PMID:23008454; http://dx.doi.org/ 10.1542/peds.2011-3886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Hviid A, Svanstrom H, Frisch M. Antibiotic use and inflammatory bowel diseases in childhood. Gut 2011; 60:49-54; PMID:20966024; http://dx.doi.org/ 10.1136/gut.2010.219683 [DOI] [PubMed] [Google Scholar]
  • [22].Lopez-Serrano P, Perez-Calle JL, Perez-Fernandez MT, Fernández-Font JM, Boixeda de Miguel D, Fernández-Rodríguez CM. Environmental risk factors in inflammatory bowel diseases. Investigating the hygiene hypothesis: a Spanish case-control study. Scand J Gastroenterol 2010; 45:1464-71 [DOI] [PubMed] [Google Scholar]
  • [23].Hugot JP, Chamaillard M, Zouali H, Lesage S, Cézard JP, Belaiche J, Almer S, Tysk C, O'Morain CA, Gassull M, et al.. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature 2001; 411:599-603; PMID:11385576; http://dx.doi.org/ 10.1038/35079107 [DOI] [PubMed] [Google Scholar]
  • [24].Strober W, Watanabe T. NOD2, an intracellular innate immune sensor involved in host defense and Crohn's disease. Mucosal Immunol 2011; 4:484-95; PMID:21750585; http://dx.doi.org/ 10.1038/mi.2011.29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Fritz T, Niederreiter L, Adolph T, Blumberg RS, Kaser A. Crohn's disease: NOD2, autophagy and ER stress converge. Gut 2011; 60:1580-8; PMID:21252204; http://dx.doi.org/ 10.1136/gut.2009.206466 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, Britton H, Moran T, Karaliuskas R, Duerr RH, et al.. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature 2001; 411:603-6; PMID:11385577; http://dx.doi.org/ 10.1038/35079114 [DOI] [PubMed] [Google Scholar]
  • [27].Petnicki-Ocwieja T, Hrncir T, Liu YJ, Biswas A, Hudcovic T, Tlaskalova-Hogenova H, Kobayashi KS. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci U S A 2009; 106:15813-8; PMID:19805227;http://dx.doi.org/ 10.1073/pnas.0907722106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Rehman A, Sina C, Gavrilova O, Häsler R, Ott S, Baines JF, Schreiber S, Rosenstiel P. Nod2 is essential for temporal development of intestinal microbial communities. Gut 2011; 60:1354-62; PMID:21421666; http://dx.doi.org/ 10.1136/gut.2010.216259 [DOI] [PubMed] [Google Scholar]
  • [29].Mondot S, Barreau F, Al Nabhani Z, Dussaillant M, Le Roux K, Doré J, Leclerc M, Hugot JP, Lepage P. Altered gut microbiota composition in immune-impaired Nod2(−/−) mice. Gut 2012; 61:634-5; PMID:21868489; http://dx.doi.org/ 10.1136/gutjnl-2011-300478 [DOI] [PubMed] [Google Scholar]
  • [30].Smith P, Siddharth J, Pearson R, Holway N, Shaxted M, Butler M, Clark N, Jamontt J, Watson RP, Sanmugalingam D, et al.. Host genetics and environmental factors regulate ecological succession of the mouse colon tissue-associated microbiota. PLoS One 2012; 7:e30273; PMID:22272321;http://dx.doi.org/ 10.1371/journal.pone.0030273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara N, Nuñez G, Flavell RA. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science 2005; 307:731-4; PMID:15692051; http://dx.doi.org/ 10.1126/science.1104911 [DOI] [PubMed] [Google Scholar]
  • [32].Couturier-Maillard A, Secher T, Rehman A, Normand S, De Arcangelis A, Haesler R, Huot L, Grandjean T, Bressenot A, Delanoye-Crespin A, et al.. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest 2013; 123:700-11; PMID:23281400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Travassos LH, Carneiro LA, Ramjeet M, Hussey S, Kim YG, Magalhães JG, Yuan L, Soares F, Chea E, Le Bourhis L, et al.. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat Immunol 2010; 11:55-62; PMID:19898471; http://dx.doi.org/ 10.1038/ni.1823 [DOI] [PubMed] [Google Scholar]
  • [34].Cooney R, Baker J, Brain O, Danis B, Pichulik T, Allan P, Ferguson DJ, Campbell BJ, Jewell D, Simmons A. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med 2010; 16:90-7; PMID:19966812; http://dx.doi.org/ 10.1038/nm.2069 [DOI] [PubMed] [Google Scholar]
  • [35].Elson CO, Cong Y, McCracken VJ, Dimmitt RA, Lorenz RG, Weaver CT. Experimental models of inflammatory bowel disease reveal innate, adaptive, and regulatory mechanisms of host dialogue with the microbiota. Immunol Rev 2005; 206:260-76; PMID:16048554; http://dx.doi.org/ 10.1111/j.0105-2896.2005.00291.x [DOI] [PubMed] [Google Scholar]
  • [36].Balish E, Warner T. Enterococcus faecalis induces inflammatory bowel disease in interleukin-10 knockout mice. Am J Pathol 2002; 160:2253-7; PMID:12057927; http://dx.doi.org/ 10.1016/S0002-9440(10)61172-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Waidmann M, Bechtold O, Frick JS, Lehr HA, Schubert S, Dobrindt U, Loeffler J, Bohn E, Autenrieth IB. Bacteroides vulgatus protects against Escherichia coli-induced colitis in gnotobiotic interleukin-2-deficient mice. Gastroenterology 2003; 125:162-77; PMID:12851881; http://dx.doi.org/ 10.1016/S0016-5085(03)00672-3 [DOI] [PubMed] [Google Scholar]
  • [38].Stepankova R, Powrie F, Kofronova O, Kozakova H, Hudcovic T, Hrncir T, Uhlig H, Read S, Rehakova Z, Benada O, et al.. Segmented filamentous bacteria in a defined bacterial cocktail induce intestinal inflammation in SCID mice reconstituted with CD45RBhigh CD4+ T cells. Inflamm Bowel Dis 2007; 13:1202-11; PMID:17607724; http://dx.doi.org/ 10.1002/ibd.20221 [DOI] [PubMed] [Google Scholar]
  • [39].Eun CS, Mishima Y, Wohlgemuth S, Liu B, Bower M, Carroll IM, Sartor RB. Induction of bacterial antigen-specific colitis by a simplified human microbiota consortium in gnotobiotic interleukin-10−/− mice. Infect Immun 2014; 82:2239-46; PMID:24643531; http://dx.doi.org/ 10.1128/IAI.01513-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Schaubeck M, Clavel T, Calasan J, Lagkouvardos I, Haange SB, Jehmlich N, Basic M, Dupont A, Hornef M, von Bergen M, et al.. Dysbiotic gut microbiota causes transmissible Crohn's disease-like ileitis independent of failure in antimicrobial defence. Gut 2016; 65:225-37; PMID:25887379; http://dx.doi.org/ 10.1136/gutjnl-2015-309333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Garrett WS, Lord GM, Punit S, Lugo-Villarino G, Mazmanian SK, Ito S, Glickman JN, Glimcher LH. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 2007; 131:33-45; PMID:17923086; http://dx.doi.org/ 10.1016/j.cell.2007.08.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, et al.. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011; 145:745-57; PMID:21565393; http://dx.doi.org/ 10.1016/j.cell.2011.04.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Brinkman BM, Becker A, Ayiseh RB, Hildebrand F, Raes J, Huys G, Vandenabeele P. Gut microbiota affects sensitivity to acute DSS-induced colitis independently of host genotype. Inflamm Bowel Dis 2013; 19:2560-7; PMID:24105395; http://dx.doi.org/ 10.1097/MIB.0b013e3182a8759a [DOI] [PubMed] [Google Scholar]
  • [44].Singh V, Kumar M, San Yeoh B, Xiao X, Saha P, Kennett MJ, Vijay-Kumar M. Inhibition of Interleukin-10 Signaling Induces Microbiota-dependent Chronic Colitis in Apolipoprotein E Deficient Mice. Inflamm Bowel Dis 2016; 22:841-52; PMID:26891260; http://dx.doi.org/ 10.1097/MIB.0000000000000699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Garrett WS, Gallini CA, Yatsunenko T, Michaud M, DuBois A, Delaney ML, Punit S, Karlsson M, Bry L, Glickman JN, et al.. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell Host Microbe 2010; 8:292-300; PMID:20833380; http://dx.doi.org/ 10.1016/j.chom.2010.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Devkota S, Wang Y, Musch MW, Leone V, Fehlner-Peach H, Nadimpalli A, Antonopoulos DA, Jabri B, Chang EB. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature 2012; 487:104-8; PMID:22722865 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Mottawea W, Chiang CK, Muhlbauer M, Starr AE, Butcher J, Abujamel T, Deeke SA, Brandel A, Zhou H, Shokralla S, et al.. Altered intestinal microbiota-host mitochondria crosstalk in new onset Crohn's disease. Nat Commun 2016; 7:13419; PMID:27876802; http://dx.doi.org/ 10.1038/ncomms13419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Ohkusa T, Okayasu I, Ogihara T, Morita K, Ogawa M, Sato N. Induction of experimental ulcerative colitis by Fusobacterium varium isolated from colonic mucosa of patients with ulcerative colitis. Gut 2003; 52:79-83; PMID:12477765; http://dx.doi.org/ 10.1136/gut.52.1.79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Eckburg PB, Relman DA. The role of microbes in Crohn's disease. Clin Infect Dis 2007; 44:256-62; PMID:17173227; http://dx.doi.org/ 10.1086/510385 [DOI] [PubMed] [Google Scholar]
  • [50].Sibartie V, Kirwan WO, O'Mahony S, Stack W, Shanahan F. Intestinal tuberculosis mimicking Crohn's disease: lessons relearned in a new era. Eur J Gastroenterol Hepatol 2007; 19:347-9; PMID:17353702; http://dx.doi.org/ 10.1097/MEG.0b013e328012122b [DOI] [PubMed] [Google Scholar]
  • [51].Woodman I, Schofield JB, Haboubi N. The histopathological mimics of inflammatory bowel disease: a critical appraisal. Tech Coloproctol 2015; 19:717-27; PMID:26385573; http://dx.doi.org/ 10.1007/s10151-015-1372-8 [DOI] [PubMed] [Google Scholar]
  • [52].Nazareth N, Magro F, Machado E, Ribeiro TG, Martinho A, Rodrigues P, Alves R, Macedo GN, Gracio D, Coelho R, et al.. Prevalence of Mycobacterium avium subsp. paratuberculosis and Escherichia coli in blood samples from patients with inflammatory bowel disease. Med Microbiol Immunol 2015; 204:681-92; PMID:25994082; http://dx.doi.org/ 10.1007/s00430-015-0420-3 [DOI] [PubMed] [Google Scholar]
  • [53].Gui GP, Thomas PR, Tizard ML, Lake J, Sanderson JD, Hermon-Taylor J. Two-year-outcomes analysis of Crohn's disease treated with rifabutin and macrolide antibiotics. J Antimicrob Chemother 1997; 39:393-400; PMID:9096189; http://dx.doi.org/ 10.1093/jac/39.3.393 [DOI] [PubMed] [Google Scholar]
  • [54].Borody TJ, Leis S, Warren EF, Surace R. Treatment of severe Crohn's disease using antimycobacterial triple therapy–approaching a cure? Dig Liver Dis 2002; 34:29-38; PMID:11926571; http://dx.doi.org/ 10.1016/S1590-8658(02)80056-1 [DOI] [PubMed] [Google Scholar]
  • [55].Shafran I, Kugler L, El-Zaatari FA, Naser SA, Sandoval J. Open clinical trial of rifabutin and clarithromycin therapy in Crohn's disease. Dig Liver Dis 2002; 34:22-8; PMID:11930899; http://dx.doi.org/ 10.1016/S1590-8658(02)80055-X [DOI] [PubMed] [Google Scholar]
  • [56].Selby W, Pavli P, Crotty B, Florin T, Radford-Smith G, Gibson P, Mitchell B, Connell W, Read R, Merrett M, et al.. Two-year combination antibiotic therapy with clarithromycin, rifabutin, and clofazimine for Crohn's disease. Gastroenterology 2007; 132:2313-9; PMID:17570206; http://dx.doi.org/ 10.1053/j.gastro.2007.03.031 [DOI] [PubMed] [Google Scholar]
  • [57].Elliott DE, Urban JJ, Argo CK, Weinstock JV. Does the failure to acquire helminthic parasites predispose to Crohn's disease? Faseb j 2000; 14:1848-55; PMID:10973934; http://dx.doi.org/ 10.1096/fj.99-0885hyp [DOI] [PubMed] [Google Scholar]
  • [58].Khan WI, Blennerhasset PA, Varghese AK, Chowdhury SK, Omsted P, Deng Y, Collins SM. Intestinal nematode infection ameliorates experimental colitis in mice. Infect Immun 2002; 70:5931-7; PMID:12379667; http://dx.doi.org/ 10.1128/IAI.70.11.5931-5937.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Elliott DE, Setiawan T, Metwali A, Blum A, Jr Urban JF, Weinstock JV. Heligmosomoides polygyrus inhibits established colitis in IL-10-deficient mice. Eur J Immunol 2004; 34:2690-8; PMID:15368285; http://dx.doi.org/ 10.1002/eji.200324833 [DOI] [PubMed] [Google Scholar]
  • [60].Ashour DS, Othman AA, Shareef MM, Gaballah HH, Mayah WW. Interactions between Trichinella spiralis infection and induced colitis in mice. J Helminthol 2014; 88:210-8; PMID:23402295; http://dx.doi.org/ 10.1017/S0022149X13000059 [DOI] [PubMed] [Google Scholar]
  • [61].Hasby EA, Hasby Saad MA, Shohieb Z, El Noby K. FoxP3+ T regulatory cells and immunomodulation after Schistosoma mansoni egg antigen immunization in experimental model of inflammatory bowel disease. Cell Immunol 2015; 295:67-76; PMID:25766778; http://dx.doi.org/ 10.1016/j.cellimm.2015.02.013 [DOI] [PubMed] [Google Scholar]
  • [62].Glendinning L, Nausch N, Free A, Taylor DW, Mutapi F. The microbiota and helminths: sharing the same niche in the human host. Parasitology 2014; 141:1255-71; PMID:24901211; http://dx.doi.org/ 10.1017/S0031182014000699 [DOI] [PubMed] [Google Scholar]
  • [63].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; 16:1841-9; PMID:20848461; http://dx.doi.org/ 10.1002/ibd.21299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Summers RW, Elliott DE, Qadir K, Jr 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; 98:2034-41; PMID:14499784; http://dx.doi.org/ 10.1111/j.1572-0241.2003.07660.x [DOI] [PubMed] [Google Scholar]
  • [65].Sandborn WJ, Elliott DE, Weinstock J, Summers RW, Landry-Wheeler A, Silver N, Harnett MD, Hanauer SB. Randomised clinical trial: the safety and tolerability of Trichuris suis ova in patients with Crohn's disease. Aliment Pharmacol Ther 2013; 38:255-63; PMID:23730956; http://dx.doi.org/ 10.1111/apt.12366 [DOI] [PubMed] [Google Scholar]
  • [66].Helmby H. Human helminth therapy to treat inflammatory disorders - where do we stand? BMC Immunol 2015; 16:12; PMID:25884706; http://dx.doi.org/ 10.1186/s12865-015-0074-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Croese J, O'Neil J, Masson J, Cooke S, Melrose W, Pritchard D, Speare R. A proof of concept study establishing Necator americanus in Crohn's patients and reservoir donors. Gut 2006; 55:136-7; PMID:16344586; http://dx.doi.org/ 10.1136/gut.2005.079129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Lee SC, Tang MS, Lim YA, Choy SH, Kurtz ZD, Cox LM, Gundra UM, Cho I, Bonneau R, Blaser MJ, et al.. Helminth colonization is associated with increased diversity of the gut microbiota. PLoS Negl Trop Dis 2014; 8:e2880; PMID:24851867; http://dx.doi.org/ 10.1371/journal.pntd.0002880 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Cantacessi C, Giacomin P, Croese J, Zakrzewski M, Sotillo J, McCann L, Nolan MJ, Mitreva M, Krause L, Loukas A. Impact of experimental hookworm infection on the human gut microbiota. J Infect Dis 2014; 210:1431-4; PMID:24795483; http://dx.doi.org/ 10.1093/infdis/jiu256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A 2007; 104:13780-5; PMID:17699621; http://dx.doi.org/ 10.1073/pnas.0706625104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, Reyes JA, Shah SA, LeLeiko N, Snapper SB, et al.. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 2012; 13:R79; http://dx.doi.org/ 10.1186/gb-2012-13-9-r79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Gevers D, Kugathasan S, Denson LA, Vázquez-Baeza Y, Van Treuren W, Ren B, Schwager E, Knights D, Song SJ, Yassour M, et al.. The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe 2014; 15:382-92; http://dx.doi.org/ 10.1016/j.chom.2014.02.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Swidsinski A, Weber J, Loening-Baucke V, Hale LP, Lochs H. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J Clin Microbiol 2005; 43:3380-9; PMID:16000463; http://dx.doi.org/ 10.1128/JCM.43.7.3380-3389.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Knights D, Silverberg MS, Weersma RK, Gevers D, Dijkstra G, Huang H, Tyler AD, van Sommeren S, Imhann F, Stempak JM, et al.. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med 2014; 6:107; PMID:25587358; http://dx.doi.org/ 10.1186/s13073-014-0107-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Rutgeerts P, Goboes K, Peeters M, Hiele M, Penninckx F, Aerts R, Kerremans R, Vantrappen G. Effect of faecal stream diversion on recurrence of Crohn's disease in the neoterminal ileum. Lancet 1991; 338:771-4; PMID:1681159; http://dx.doi.org/ 10.1016/0140-6736(91)90663-A [DOI] [PubMed] [Google Scholar]
  • [76].Dey N, Soergel DA, Repo S, Brenner SE. Association of gut microbiota with post-operative clinical course in Crohn's disease. BMC Gastroenterol 2013; 13:131; PMID:23964800; http://dx.doi.org/ 10.1186/1471-230X-13-131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].De Cruz P, Kang S, Wagner J, Buckley M, Sim WH, Prideaux L, Lockett T, McSweeney C, Morrison M, Kirkwood CD, et al.. Association between specific mucosa-associated microbiota in Crohn's disease at the time of resection and subsequent disease recurrence: a pilot study. J Gastroenterol Hepatol 2015; 30:268-78; PMID:25087692; http://dx.doi.org/ 10.1111/jgh.12694 [DOI] [PubMed] [Google Scholar]
  • [78].Matijasic M, Mestrovic T, Peric M, Čipčić Paljetak H, Panek M, Vranešić Bender D, Ljubas Kelečić D, Ž Krznarić, Verbanac D. Modulating Composition and Metabolic Activity of the Gut Microbiota in IBD Patients. Int J Mol Sci 2016; 17:pii: E578. PMID:27104515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Sokol H. Probiotics and antibiotics in IBD. Dig Dis 2014; 32(Suppl 1):10-7; PMID:25531348; http://dx.doi.org/ 10.1159/000367820 [DOI] [PubMed] [Google Scholar]
  • [80].Khan KJ, Ullman TA, Ford AC, et al.. Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol 2011; 106:661-73; http://dx.doi.org/ 10.1038/ajg.2011.72 [DOI] [PubMed] [Google Scholar]
  • [81].Sutherland L, Singleton J, Sessions J, Hanauer S, Krawitt E, Rankin G, Summers R, Mekhjian H, Greenberger N, Kelly M, et al.. Double blind, placebo controlled trial of metronidazole in Crohn's disease. Gut 1991; 32:1071-5; PMID:1916494; http://dx.doi.org/ 10.1136/gut.32.9.1071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Prantera C, Lochs H, Campieri M, Scribano ML, Sturniolo GC, Castiglione F, Cottone M. Antibiotic treatment of Crohn's disease: results of a multicentre, double blind, randomized, placebo-controlled trial with rifaximin. Aliment Pharmacol Ther 2006; 23:1117-25; PMID:16611272; http://dx.doi.org/ 10.1111/j.1365-2036.2006.02879.x [DOI] [PubMed] [Google Scholar]
  • [83].Wu XW, Ji HZ, Wang FY. Meta-analysis of ciprofloxacin in treatment of Crohn's disease. Biomed Rep 2015; 3:70-74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Henker J, Muller S, Laass MW, Schreiner A, Schulze J. Probiotic Escherichia coli Nissle 1917 (EcN) for successful remission maintenance of ulcerative colitis in children and adolescents: an open-label pilot study. Z Gastroenterol 2008; 46:874-5; http://dx.doi.org/ 10.1055/s-2008-1027463 [DOI] [PubMed] [Google Scholar]
  • [85].Kruis W, Schutz E, Fric P, Fixa B, Judmaier G, Stolte M. Double-blind comparison of an oral Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther 1997; 11:853-8; PMID:9354192; http://dx.doi.org/ 10.1046/j.1365-2036.1997.00225.x [DOI] [PubMed] [Google Scholar]
  • [86].Kato K, Mizuno S, Umesaki Y, Ishii Y, Sugitani M, Imaoka A, Otsuka M, Hasunuma O, Kurihara R, Iwasaki A, et al.. Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis. Aliment Pharmacol Ther 2004; 20:1133-41; http://dx.doi.org/ 10.1111/j.1365-2036.2004.02268.x [DOI] [PubMed] [Google Scholar]
  • [87].Zocco MA, dal Verme LZ, Cremonini F, Piscaglia AC, Nista EC, Candelli M, Novi M, Rigante D, Cazzato IA, Ojetti V, et al.. Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther 2006; 23:1567-74; PMID:16696804; http://dx.doi.org/ 10.1111/j.1365-2036.2006.02927.x [DOI] [PubMed] [Google Scholar]
  • [88].Guslandi M. Saccharomyces boulardii plus rifaximin in mesalamine-intolerant ulcerative colitis. J Clin Gastroenterol 2010; 44:385. PMID:20104184 [DOI] [PubMed] [Google Scholar]
  • [89].Bibiloni R, Fedorak RN, Tannock GW, Madsen KL, Gionchetti P, Campieri M, De Simone C, Sartor RB. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am J Gastroenterol 2005; 100:1539-46; PMID:15984978; http://dx.doi.org/ 10.1111/j.1572-0241.2005.41794.x [DOI] [PubMed] [Google Scholar]
  • [90].Mardini HE, Grigorian AY. Probiotic mix VSL#3 is effective adjunctive therapy for mild to moderately active ulcerative colitis: a meta-analysis. Inflamm Bowel Dis 2014; 20:1562-7; http://dx.doi.org/ 10.1097/MIB.0000000000000084 [DOI] [PubMed] [Google Scholar]
  • [91].Chapman TM, Plosker GL, Figgitt DP. VSL#3 probiotic mixture: a review of its use in chronic inflammatory bowel diseases. Drugs 2006; 66:1371-87; PMID:16903771; http://dx.doi.org/ 10.2165/00003495-200666100-00006 [DOI] [PubMed] [Google Scholar]
  • [92].Maldonado-Gomez MX, Martinez I, Bottacini F, O'Callaghan A, Ventura M, van Sinderen D, Hillmann B, Vangay P, Knights D, Hutkins RW, et al.. Stable Engraftment of Bifidobacterium longum AH1206 in the Human Gut Depends on Individualized Features of the Resident Microbiome. Cell Host Microbe 2016; 20:515-526; http://dx.doi.org/ 10.1016/j.chom.2016.09.001 [DOI] [PubMed] [Google Scholar]
  • [93].Eiseman B, Silen W, Bascom GS, Kauvar AJ. Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery 1958; 44:854-9; PMID:13592638 [PubMed] [Google Scholar]
  • [94].Khoruts A, Dicksved J, Jansson JK, Sadowsky MJ. Changes in the composition of the human fecal microbiome after bacteriotherapy for recurrent Clostridium difficile-associated diarrhea. J Clin Gastroenterol 2010; 44:354-60; PMID:20048681 [DOI] [PubMed] [Google Scholar]
  • [95].Hamilton MJ, Weingarden AR, Unno T, Khoruts A, Sadowsky MJ. High-throughput DNA sequence analysis reveals stable engraftment of gut microbiota following transplantation of previously frozen fecal bacteria. Gut Microbes 2013; 4:125-35; PMID:23333862; http://dx.doi.org/ 10.4161/gmic.23571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Weingarden A, Gonzalez A, Vazquez-Baeza Y, Weiss S, Humphry G, Berg-Lyons D, Knights D, Unno T, Bobr A, Kang J, et al.. Dynamic changes in short- and long-term bacterial composition following fecal microbiota transplantation for recurrent Clostridium difficile infection. Microbiome 2015; 3:10; PMID:25825673; http://dx.doi.org/ 10.1186/s40168-015-0070-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Bojanova DP, Bordenstein SR. Fecal Transplants: What Is Being Transferred? PLoS Biol 2016; 14:e1002503; PMID:27404502; http://dx.doi.org/ 10.1371/journal.pbio.1002503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Bennet JD, Brinkman M. Treatment of ulcerative colitis by implantation of normal colonic flora. Lancet 1989; 1:164; PMID:2563083; http://dx.doi.org/ 10.1016/S0140-6736(89)91183-5 [DOI] [PubMed] [Google Scholar]
  • [99].Borody TJ, Campbell J. Fecal microbiota transplantation: techniques, applications, and issues. Gastroenterol Clin North Am 2012; 41:781-803; PMID:23101687; http://dx.doi.org/ 10.1016/j.gtc.2012.08.008 [DOI] [PubMed] [Google Scholar]
  • [100].Borody TJ, Warren EF, Leis S, Surace R, Ashman O. Treatment of ulcerative colitis using fecal bacteriotherapy. Journal of Clinical Gastroenterology 2003; 37:42-47; PMID:12811208; http://dx.doi.org/ 10.1097/00004836-200307000-00012 [DOI] [PubMed] [Google Scholar]
  • [101].Borody T, Wettstein A, Campbell J, et al.. Fecal microbiota transplantation in ulcerative colitis: Review of 24 years experience. Am J Gastronerol Conf 77th Annu Sci Meet Am Coll Gastroenterol 2012. Oct 19 - 24, Las Vegas, NV; 107:S665. [Google Scholar]
  • [102].Kunde S, Pham A, Bonczyk S, Crumb T, Duba M, Jr Conrad H, Cloney D, Kugathasan S. Safety, tolerability, and clinical response after fecal transplantation in children and young adults with ulcerative colitis. J Pediatr Gastroenterol Nutr 2013; 56:597-601; PMID:23542823; http://dx.doi.org/ 10.1097/MPG.0b013e318292fa0d [DOI] [PubMed] [Google Scholar]
  • [103].Suskind DL, Singh N, Nielson H, Wahbeh G. Fecal microbial transplant via nasogastric tube for active pediatric ulcerative colitis. J Pediatr Gastroenterol Nutr 2015; 60:27-9; http://dx.doi.org/ 10.1097/MPG.0000000000000544 [DOI] [PubMed] [Google Scholar]
  • [104].Brandt LJ, Aroniadis OC, Mellow M, Kanatzar A, Kelly C, Park T, Stollman N, Rohlke F, Surawicz C. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. Am J Gastroenterol 2012; 107:1079-87; PMID:22450732; http://dx.doi.org/ 10.1038/ajg.2012.60 [DOI] [PubMed] [Google Scholar]
  • [105].Kump PK, Grochenig HP, Lackner S, Trajanoski S, Reicht G, Hoffmann KM, Deutschmann A, Wenzl HH, Petritsch W, Krejs GJ, et al.. Alteration of intestinal dysbiosis by fecal microbiota transplantation does not induce remission in patients with chronic active ulcerative colitis. Inflamm Bowel Dis 2013; 19:2155-65; PMID:23899544;http://dx.doi.org/ 10.1097/MIB.0b013e31829ea325 [DOI] [PubMed] [Google Scholar]
  • [106].Damman CJ, Brittnacher MJ, Hayden HS, et al.. Single colonoscopically administered fecal microbiota transplant for ulcerative colitis-a pilot study to determine therapeutic benefit and graft stability. Paper presented at: Digestive Diseases Week 2014; 2014 May 3-6, Chicago, IL. Gastroenterology 2014:S-460 [Google Scholar]
  • [107].Vermeire S, Joossens M, Verbeke K, Wang J, Machiels K, Sabino J, Ferrante M, Van Assche G, Rutgeerts P, Raes J. Donor Species Richness Determines Faecal Microbiota Transplantation Success in Inflammatory Bowel Disease. J Crohns Colitis 2016; 10:387-94; http://dx.doi.org/ 10.1093/ecco-jcc/jjv203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Wei Y, Zhu W, Gong J, Guo D, Gu L, Li N, Li J. Fecal Microbiota Transplantation Improves the Quality of Life in Patients with Inflammatory Bowel Disease. Gastroenterol Res Pract 2015; 2015:517597; PMID:26146498; http://dx.doi.org/ 10.1155/2015/517597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Libertucci JW, Whelan FJ, Moayyedi P, et al. Investigating the microbiome pre and post fecal microbiota therapy from active ulcerative colitis patients in a randomized placebo controlled trial. Paper presented at: Digestive Diseases Week 2014; 2014 May 3-6, Chicago, IL. Gastroenterology 2014; 146:S-902; http://dx.doi.org/ 10.1016/S0016-5085(14)63280-7 [DOI] [Google Scholar]
  • [110].Kahn SA, Rubin DT. When Subjects Violate the Research Covenant: Lessons Learned from a Failed Clinical Trial of Fecal Microbiota Transplantation. Am J Gastroenterol 2016; 111(11):1508-1510; PMID:27166127 [DOI] [PubMed] [Google Scholar]
  • [111].Moayyedi P, Surette MG, Kim PT, Libertucci J, Wolfe M, Onischi C, Armstrong D, Marshall JK, Kassam Z, Reinisch W, et al.. Fecal Microbiota Transplantation Induces Remission in Patients With Active Ulcerative Colitis in a Randomized Controlled Trial. Gastroenterology 2015; 149(1):102-109; http://dx.doi.org/ 10.1053/j.gastro.2015.04.001 [DOI] [PubMed] [Google Scholar]
  • [112].Rossen NG, Fuentes S, van der Spek MJ, Tijssen JG, Hartman JH, Duflou A, Löwenberg M, van den Brink GR, Mathus-Vliegen EM, de Vos WM, et al.. Findings From a Randomized Controlled Trial of Fecal Transplantation for Patients With Ulcerative Colitis. Gastroenterology 2015; 149(1):110-118; PMID:25836986; http://dx.doi.org/ 10.1053/j.gastro.2015.03.045 [DOI] [PubMed] [Google Scholar]
  • [113].Paramsothy S, Kamm MA, Walsh A, et al.. 600 Multi Donor Intense Faecal Microbiota Transplantation is an Effective Treatment for Resistant Ulcerative Colitis: A Randomised Placebo-Controlled Trial. Paper presented at: Digestive Diseases Week 2016; 2016 May 21-24; San Diego, CA. Gastroenterology 2016; 150:S122-S123; http://dx.doi.org/ 10.1016/S0016-5085(16)30517-0 [DOI] [Google Scholar]
  • [114].Kao D, Hotte N, Gillevet P, Madsen K. Fecal microbiota transplantation inducing remission in Crohn's colitis and the associated changes in fecal microbial profile. J Clin Gastroenterol 2014; 48:625-8; PMID:24667590;http://dx.doi.org/ 10.1097/MCG.0000000000000131 [DOI] [PubMed] [Google Scholar]
  • [115].Vermeire S, Joossens M, Verbeke K, et al.. Pilot Study on the safety and efficacy of faecal microbiota transplantation in refractory Crohn's disease. Paper presented at: Digestive Diseases Week 2012; 2012 May 19-22; San Diego, CA. Gastroenterology 2012; 142:S360; http://dx.doi.org/ 10.1016/S0016-5085(12)61356-0 [DOI] [Google Scholar]
  • [116].Gordon H, Harbord M. A patient with severe Crohn's colitis responds to Faecal Microbiota Transplantation. J Crohns Colitis 2014; 8:256-7; PMID:24239403; http://dx.doi.org/ 10.1016/j.crohns.2013.10.007 [DOI] [PubMed] [Google Scholar]
  • [117].Borody TJ, George L, Andrews P, Brandl S, Noonan S, Cole P, Hyland L, Morgan A, Maysey J, Moore-Jones D. Bowel-flora alteration: a potential cure for inflammatory bowel disease and irritable bowel syndrome? Med J Aust 1989; 150:604. PMID:2783214 [DOI] [PubMed] [Google Scholar]
  • [118].Zhang FM, Wang HG, Wang M, Cui BT, Fan ZN, Ji GZ. Fecal microbiota transplantation for severe enterocolonic fistulizing Crohn's disease. World J Gastroenterol 2013; 19:7213-6; http://dx.doi.org/ 10.3748/wjg.v19.i41.7213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Cui B, Feng Q, Wang H, Wang M, Peng Z, Li P, Huang G, Liu Z, Wu P, Fan Z, et al.. Fecal microbiota transplantation through mid-gut for refractory Crohn's disease: safety, feasibility, and efficacy trial results. J Gastroenterol Hepatol 2015; 30:51-8; http://dx.doi.org/ 10.1111/jgh.12727 [DOI] [PubMed] [Google Scholar]
  • [120].Suskind DL, Brittnacher MJ, Wahbeh G, Shaffer ML, Hayden HS, Qin X, Singh N, Damman CJ, Hager KR, Nielson H, et al.. Fecal microbial transplant effect on clinical outcomes and fecal microbiome in active Crohn's disease. Inflamm Bowel Dis 2015; 21:556-63; http://dx.doi.org/ 10.1097/MIB.0000000000000307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Vaughn BP, Vatanen T, Allegretti JR, Bai A, Xavier RJ, Korzenik J, Gevers D, Ting A, Robson SC, Moss AC. Increased Intestinal Microbial Diversity Following Fecal Microbiota Transplant for Active Crohn's Disease. Inflamm Bowel Dis 2016; 22:2182-90; PMID:27542133; http://dx.doi.org/ 10.1097/MIB.0000000000000893 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Zella GC, Hait EJ, Glavan T, Gevers D, Ward DV, Kitts CL, Korzenik JR. Distinct microbiome in pouchitis compared to healthy pouches in ulcerative colitis and familial adenomatous polyposis. Inflamm Bowel Dis 2011; 17:1092-100; PMID:20845425; http://dx.doi.org/ 10.1002/ibd.21460 [DOI] [PubMed] [Google Scholar]
  • [123].Young VB, Raffals LH, Huse SM, Vital M, Dai D, Schloss PD, Brulc JM, Antonopoulos DA, Arrieta RL, Kwon JH, et al.. Multiphasic analysis of the temporal development of the distal gut microbiota in patients following ileal pouch anal anastomosis. Microbiome 2013; 1:9; http://dx.doi.org/ 10.1186/2049-2618-1-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Landy J, Walker AW, Li JV, Al-Hassi HO, Ronde E, English NR, Mann ER, Bernardo D, McLaughlin SD, Parkhill J, et al.. Variable alterations of the microbiota, without metabolic or immunological change, following faecal microbiota transplantation in patients with chronic pouchitis. Sci Rep 2015; 5:12955; PMID:26264409; http://dx.doi.org/ 10.1038/srep12955 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Anderson JL, Edney RJ, Whelan K. Systematic review: faecal microbiota transplantation in the management of inflammatory bowel disease. Aliment Pharmacol Ther 2012; 36:503-16; PMID:22827693; http://dx.doi.org/ 10.1111/j.1365-2036.2012.05220.x [DOI] [PubMed] [Google Scholar]
  • [126].Sha S, Liang J, Chen M, Xu B, Liang C, Wei N, Wu K. Systematic review: faecal microbiota transplantation therapy for digestive and nondigestive disorders in adults and children. Aliment Pharmacol Ther 2014; 39:1003-32; PMID:24641570; http://dx.doi.org/ 10.1111/apt.12699 [DOI] [PubMed] [Google Scholar]
  • [127].Colman RJ, Rubin DT. Fecal microbiota transplantation as therapy for inflammatory bowel disease: A systematic review and meta-analysis. J Crohns Colitis 2014; 8(12):1569-81; PMID:25223604 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].De Leon LM, Watson JB, Kelly CR. Transient flare of ulcerative colitis after fecal microbiota transplantation for recurrent Clostridium difficile infection. Clin Gastroenterol Hepatol 2013; 11:1036-8; PMID:23669309; http://dx.doi.org/ 10.1016/j.cgh.2013.04.045 [DOI] [PubMed] [Google Scholar]
  • [129].Fischer M, Kao D, Kelly C, Kuchipudi A, Jafri SM, Blumenkehl M, Rex D, Mellow M, Kaur N, Sokol H, et al.. Fecal Microbiota Transplantation is Safe and Efficacious for Recurrent or Refractory Clostridium difficile Infection in Patients with Inflammatory Bowel Disease. Inflamm Bowel Dis 2016; 22:2402-9; PMID:27580384; http://dx.doi.org/ 10.1097/MIB.0000000000000908 [DOI] [PubMed] [Google Scholar]
  • [130].Khoruts A, Rank KM, Newman KM, Viskocil K, Vaughn BP, Hamilton MJ, Sadowsky MJ. Inflammatory Bowel Disease Affects the Outcome of Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection. Clin Gastroenterol Hepatol 2016; 14(10):1433-8; PMID:26905904; http://dx.doi.org/ 10.1016/j.cgh.2016.02.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Ananthakrishnan AN. Epidemiology and risk factors for IBD. Nat Rev Gastroenterol Hepatol 2015; 12:205-17; PMID:25732745;http://dx.doi.org/ 10.1038/nrgastro.2015.34 [DOI] [PubMed] [Google Scholar]
  • [132].Hilmi I, Jaya F, Chua A, Heng WC, Singh H, Goh KL. A first study on the incidence and prevalence of IBD in Malaysia–results from the Kinta Valley IBD Epidemiology Study. J Crohns Colitis 2015; 9:404-9; PMID:25744112; http://dx.doi.org/ 10.1093/ecco-jcc/jjv039 [DOI] [PubMed] [Google Scholar]
  • [133].Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, et al.. Human gut microbiome viewed across age and geography. Nature 2012; 486:222-7; PMID:22699611 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Rampelli S, Schnorr SL, Consolandi C, Turroni S, Severgnini M, Peano C, Brigidi P, Crittenden AN, Henry AG, Candela M. Metagenome Sequencing of the Hadza Hunter-Gatherer Gut Microbiota. Curr Biol 2015; 25:1682-93; PMID:25981789; http://dx.doi.org/ 10.1016/j.cub.2015.04.055 [DOI] [PubMed] [Google Scholar]
  • [135].Obregon-Tito AJ, Tito RY, Metcalf J, Sankaranarayanan K, Clemente JC, Ursell LK, Zech Xu Z, Van Treuren W, Knight R, Gaffney PM, et al.. Subsistence strategies in traditional societies distinguish gut microbiomes. Nat Commun 2015; 6:6505; http://dx.doi.org/ 10.1038/ncomms7505 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS, Sonnenburg JL. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016; 529:212-5; PMID:26762459; http://dx.doi.org/ 10.1038/nature16504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, Ballet V, Claes K, Van Immerseel F, Verbeke K, et al.. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 2014; 63:1275-83; PMID:24021287; http://dx.doi.org/ 10.1136/gutjnl-2013-304833 [DOI] [PubMed] [Google Scholar]
  • [138].Wang W, Chen L, Zhou R, Wang X, Song L, Huang S, Wang G, Xia B. Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J Clin Microbiol 2014; 52:398-406; http://dx.doi.org/ 10.1128/JCM.01500-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Takahashi K, Nishida A, Fujimoto T, Fujii M, Shioya M, Imaeda H, Inatomi O, Bamba S, Sugimoto M, Andoh A. Reduced Abundance of Butyrate-Producing Bacteria Species in the Fecal Microbial Community in Crohn's Disease. Digestion 2016; 93:59-65; http://dx.doi.org/ 10.1159/000441768 [DOI] [PubMed] [Google Scholar]

Articles from Gut Microbes are provided here courtesy of Taylor & Francis

RESOURCES