Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Nov 14, 2014; 20(42): 15657-15663
Published online Nov 14, 2014. doi: 10.3748/wjg.v20.i42.15657
Enteric microbiota leads to new therapeutic strategies for ulcerative colitis
Wei-Xu Chen, Li-Hua Ren, Rui-Hua Shi, Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
Author contributions: Chen WX wrote and reviewed the manuscript; Ren LH contributed to the work; Shi RH reviewed and assisted with editing the manuscript.
Correspondence to: Rui-Hua Shi, MD, PhD, Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, China. ruihuashi@126.com
Telephone: +86-25-83674636 Fax: +86-25-83674636
Received: February 25, 2014
Revised: May 11, 2014
Accepted: June 26, 2014
Published online: November 14, 2014

Abstract

Ulcerative colitis (UC) is a leading form of inflammatory bowel disease that involves chronic relapsing or progressive inflammation. As a significant proportion of UC patients treated with conventional therapies do not achieve remission, there is a pressing need for the development of more effective therapies. The human gut contains a large, diverse, and dynamic population of microorganisms, collectively referred to as the enteric microbiota. There is a symbiotic relationship between the human host and the enteric microbiota, which provides nutrition, protection against pathogenic organisms, and promotes immune homeostasis. An imbalance of the normal enteric microbiota composition (termed dysbiosis) underlies the pathogenesis of UC. A reduction of enteric microbiota diversity has been observed in UC patients, mainly affecting the butyrate-producing bacteria, such as Faecalibacterium prausnitzii, which can repress pro-inflammatory cytokines. Many studies have shown that enteric microbiota plays an important role in anti-inflammatory and immunoregulatory activities, which can benefit UC patients. Therefore, manipulation of the dysbiosis is an attractive approach for UC therapy. Various therapies targeting a restoration of the enteric microbiota have shown efficacy in treating patients with active and chronic forms of UC. Such therapies include fecal microbiota transplantation, probiotics, prebiotics, antibiotics, helminth therapy, and dietary polyphenols, all of which can alter the abundance and composition of the enteric microbiota. Although there have been many large, randomized controlled clinical trials assessing these treatments, the effectiveness and safety of these bacteria-driven therapies need further evaluation. This review focuses on the important role that the enteric microbiota plays in maintaining intestinal homeostasis and discusses new therapeutic strategies targeting the enteric microbiota for UC.

Key Words: Ulcerative colitis, Enteric microbiota, Dysbiosis, Probiotic, Fecal microbiota transplantation, Polyphenol

Core tip: The human gut is comprised of a large, diverse, and dynamic microbiota. The enteric microbiota plays an important role in regulating anti-inflammatory and immunoregulatory activities. An imbalance of the normal enteric microbiota composition, termed dysbiosis, underlies the pathogenesis of ulcerative colitis (UC). Therefore, manipulation of the dysbiosis is an attractive strategy for UC therapy. This review discusses new therapies associated with the regulation of enteric microbiota for UC patients.



INTRODUCTION

Inflammatory bowel disease (IBD) refers to chronic relapsing or progressive inflammatory conditions that may affect the entire gastrointestinal tract. Ulcerative colitis (UC) and Crohn’s disease (CD) are two main clinically defined forms of IBD[1]. In UC, inflammation is limited to the lining of the colon, whereas transmural inflammation occurs along any part of the gastrointestinal tract in CD[2]. The incidence and prevalence of IBD have continued to increase over the past few decades throughout various regions around the world[3]. Although the precise pathogenesis is unknown, it is thought that the etiology of IBD involves dysfunction of the mucosal immune system, which develops from complex interactions between the host immune system and genetic and environmental factors[4].

The gastrointestinal tract is possibly the most complicated immune organ of the entire human body. The intestinal mucosa is continuously exposed to a variety of commensal microbiota and food antigens. The gut must suppress excessive immune responses to antigen stimulation, and thus uses both acquired and innate immune systems to maintain intestinal homeostasis. Evidence suggests that the development and function of the intestinal immune system depend on its specific enteric microbiota, which appears to have co-evolved[5]. However, a disruption in this system can lead to aberrant immune responses to the enteric microbiota and cause chronic inflammation within the gut[5,6]. This review focuses on the important role the enteric microbiota plays in maintaining intestinal homeostasis and discusses new therapeutic strategies for UC that are based on the enteric microbiota.

HUMAN ENTERIC MICROBIOTA

The human gut is a complex anaerobic environment with a large, diverse, and dynamic enteric microbiota composed of more than 100 trillion microorganisms - which is ten times greater than the total estimated number of human cells[7]. Until the 1990s, the characterization of the enteric microbiota was limited to the use of bacteriological culture. However, less than 30% of fecal microorganisms observed under the microscope can be cultured. More recent advances in culture-independent techniques, such as 16S rRNA gene probing-based strategies and metagenomics, have broadened our knowledge of the complexity of this ecosystem[8,9]. The enteric microbiota is comprised of more than 1000 different bacterial species[10]. The majority of these organisms are from seven phyla: Firmicutes, Bacteroidetes, Proteobacteria, Fusobacteria, Verrucomicrobia, Cyanobacteria and Actinobacteria. Over 90% of the human enteric microbiota are from the Firmicutes and Bacteroidetes phyla, though Proteobacteria are also common[11,12].

There is a beneficial, symbiotic relationship between the host and the enteric microbiota. The host provides a nutrient-rich habitat to support the microbiota, which in return confers a huge diversity of genes and metabolic functions. First, the enteric microbiota provides nutrition to the host by fermenting non-digestible substrates and producing short chain fatty acids, and aiding in the absorption of ions, amino acids and vitamins. Second, the microbiota provides a barrier against pathogenic bacteria. Furthermore, the microbiota plays an important role in regulating immune and gut homeostasis[13]. Importantly, an imbalance of this symbiotic state, termed dysbiosis, underlies the pathogenesis of several inflammatory diseases, including allergic skin and respiratory disorders, rheumatoid arthritis, type I diabetes and IBD[14].

Enteric microbiota in IBD

Quantitative and qualitative changes in the composition of the enteric microbiota have been observed in IBD, through a decrease in the diversity and an increase in the concentration of bacterial species[13,15]. Dysbiosis in patients with CD has been well characterized, with a loss of bacteria from the Firmicutes phylum, including Faecalibacterium prausnitzii (F. prausnitzii), the major butyrate-producing bacterium in cluster IV of the Clostridium leptum phylogenetic group in the gut[16]. Butyrate, the key energy source for colonic epithelial cells, represses the production of pro-inflammatory cytokines in the intestinal mucosa. The reduction of F. prausnitzii in mucosal and fecal samples represents the most replicated species-specific finding so far in CD[17-19].

The decrease in enteric microbiota diversity has also been described in UC[20-22]. Researchers found significantly fewer Bacteroides and Clostridium (C. coccoides and C. leptum) in fecal samples of UC patients[23], and a higher amount of Enterococcus and Gammaproteobacteria[21,22,24]. Additionally, Machiels et al[25] found that two well-known butyrate-producing Firmicutes bacteria, Roseburia hominis and F. prausnitzii, were reduced in UC patients. Moreover, the presence of butyrate-producing members of the clostridial cluster was found to vary with disease activity[26]. Varela et al[27] found that counts of F. prausnitzii were associated with relapse and maintenance of clinical remission, with low counts corresponding to short-term remission, and counts increasing during remission. In another study that analyzed both fecal and biopsy UC specimens, F. prausnitzii was sharply decreased in both, whereas Bifidobacterium was significantly increased in the biopsy specimens of active UC[28]. However, the composition of fecal and mucosal-associated microbiota changes throughout the progression of UC[26,29].

It is still not clear whether the observed dysbiosis contributes to the development or is a consequence of UC. Although studies documenting the association between microbial changes and UC do not necessarily predict cause-effect relationships, they at least demonstrate that the enteric microbiota plays an important role in anti-inflammatory and immunoregulatory activities, which can benefit UC patients[30].

THERAPIES ASSOCIATED WITH ENTERIC MICROBIOTA

Current therapeutic strategies for treating UC typically follow a step-up strategy, including mesalazine, steroids, immunosuppressants and biologics[31]. Although a considerable proportion of patients maintain remission when using this approach, a significant number of them experience persistent disease activity and ultimately require colectomy[32]. Therefore, the development of new and effective therapies is needed. Manipulation of the dysbiosis is an attractive therapeutic approach, particularly because the interaction between enteric microbiota and the host immune system perpetuates the inflammation in UC.

Fecal microbiota transplantation

Fecal microbiota transplantation (FMT) involves the administration of fecal material from a healthy donor into the intestinal tract of a recipient. This approach attempts to recover the composition and function of the enteric microbiota in patients with chronic gastrointestinal infections and IBD[33]. FMT was first used in 1985 when standard treatments for Clostridium difficile infection (CDI) failed. The efficacy achieved by this application[34] has since brought greater attention to FMT, which is increasingly being used for treatment of IBD, and UC in particular.

A case report published in 2003 documented six patients with UC who were treated with FMT[35]. Some symptoms of UC improved within one week, and a complete reversal of symptoms was achieved in all patients by four months after FMT. In addition, there was no clinical, colonoscopic, or histologic evidence of UC in any patient after 1 to 13 years, even without any UC medication. Another study observed ten children with mild to moderate UC for four weeks after receiving FMT[36]. Seven of these subjects showed a clinical response within one week, and six maintained a clinical response after one month. Among these, three subjects achieved clinical remission after one week, which was maintained throughout the four-week observation period. Furthermore, no serious adverse events were noted.

In contrast, some studies report only temporary clinical improvement from FMT rather than remission. Kump et al[37] reported short-term clinical improvement with colonoscopic FMT in six patients that were nonresponsive to standard medical therapy. Although there were significant changes in the composition of the intestinal microbiota that resulted in a partial improvement of UC-associated dysbiosis, none of the patients achieved clinical remission. Similar results were obtained in a study of five patients with moderate to severe active UC by Angelberger et al[38]. Only one patient was observed to have some clinical improvement, and none of the five patients achieved remission during the 12-wk study. A transient increase in the similarity to donor microbiota and an increase in phylotype richness were detected in two patients less than four weeks after FMT. Notably, the microbiota of the sole clinical responder was similar to the donor’s, even 12 wk after FMT, which was characterized by successive colonization by the anti-inflammatory and short-chain fatty acid-producing F. prausnitzii, Rosebura faecis and Bacteroides ovatus.

There are several possible explanations for the failure of FMT in some UC patients[39]. First, enteric microbiota in non-responsive patients may be affected by other factors, including dietary intake or exposure to cigarette smoke. Second, the dysbiosis may be a result of UC, rather than the cause. Finally, the type of patients used in these studies may have affected the results, as those with more severe disease and for whom current medical therapies had failed were chosen.

Because of the uncertain effectiveness and safety issues, the study of FMT in UC is complicated. Further studies should identify the best candidates for FMT, such as patients with mild to moderate UC, those who already have a medically induced remission, or those who are newly diagnosed. In addition, more attention should be paid regarding the side effects accompanying FMT. A recent case report of a patient with UC that had been in remission for more than 20 years experienced a UC flare-up after FMT for treatment of CDI[40]. Thus, additional careful studies are needed to determine the effectiveness and safety of FMT, and to facilitate the development of FMT as potential therapy for UC patients.

Probiotics and prebiotics

Probiotics are living non-pathogenic microbes, including Lactobacilli, Bifidobacteria, Enterococci and some yeast species[41], which can benefit the host and affect the structure and function of the enteric microbiota through diverse mechanisms[42]. Prebiotics, which also confer benefits to the host, are defined as non-digestible, selectively fermented, short-chain carbohydrates that allow specific changes in the composition and/or activity of the enteric microbiota[43]. Probiotics and prebiotics have been widely used in the prevention and treatment of important gastroenterological conditions, such as irritable bowel syndrome (IBS) and IBD, and infectious diarrhea.

A probiotic mixture named VSL3, consisting of four strains of Lactobacilli, three strains of Bifidobacteria and one strain of Streptococcus thermophiles, was effectively used as maintenance therapy in patients with recurrent or chronic pouchitis[44-46]. Further evidence supporting the use of probiotics was provided by a one-year, placebo-controlled, double-blind study involving a total of 29 children with active UC who received either VSL3 or a placebo in addition to steroid induction and mesalamine maintenance treatment[47]. A greater number of patients receiving the placebo relapsed within one year, suggesting that VSL3 is effective in maintaining remission. In two randomized, double dummy trials, Nissle 1917, a non-pathogenic strain of Escherichia coli (E. coli), was as effective as standard mesalamine in maintaining remission in UC[48,49]. In both studies, there were no differences in the percentage of patients who relapsed or the safety profile of one-year treatments with E. coli Nissle 1917 or mesalamine. However, an open-label, randomized trial by Zocco et al[50] demonstrated that treatment with Lactobacillus GG was more effective than mesalamine in prolonging the relapse-free time in UC patients.

Probiotics also have the capability to induce remission. In a multicenter, randomized, double-blind, placebo-controlled trial including 147 adult patients with mild to moderate UC, a greater percentage of patients receiving VSL3 for 12 wk achieved remission or had greater activity reduction [reduced UC disease activity index (UCDAI) score] than those receiving a placebo[51]. In a similarly designed clinical trial, 144 patients who received VSL3 for 8 wk in addition to their standard pharmaceutical therapy had greater improvement in UCDAI scores, and a significant reduction in rectal bleeding[52]. A study by Oliva et al[53] demonstrated that local administration of Lactobacillus reuteri ATCC 55730 with standard oral mesalazine reduced the inflammation of rectal mucosa in pediatric UC patients.

The prebiotic germinated barley foodstuff has also been shown to reduce the clinical activity of UC together with the baseline treatment, and to maintain remission[54-56]. The combined use of probiotics and prebiotics, such as Bifidobacterium and galacto-oligosaccharide, has also been shown to improve the clinical status of UC patients[57]. However, other studies using combinations of probiotics and prebiotics have been less successful[58,59]. Future probiotic and prebiotic treatments for UC patients should be more personalized, taking into consideration the age of patient, the phase of disease, and the molecular pattern of dysbiosis[60]. Moreover, therapeutic outcomes may be improved by careful selection of agents, and protective commensal enteric species may be more suitable.

Antibiotics

There have been conflicting results over the past few decades concerning the efficacy of antibacterial therapies for the treatment of UC[31]. However, a combination of broad-spectrum antibiotics completely inhibited spontaneous colitis in animal models, suggesting a possible rationale for antibiotic combination therapy for the treatment of UC[61,62]. A two-week antibiotic combination therapy consisting of amoxicillin, tetracycline and metronidazole (ATM) was shown to be effective against Fusobacterium varium in UC patients[63,64]. F. varium has been detected in the colonic mucosa of a large number of UC patients[65] and can induce UC lesions in experimental animals[66]. ATM treatment was also shown to provide more efficient improvement, remission and steroid withdrawal than a placebo in a double-blind, multicenter trial including 105 patients that was conducted in patients with chronic mild to severe relapsing UC[67]. This treatment has also shown efficacy in the treatment of refractory or steroid-dependent UC[68].

Helminth therapy

Modern hygienic practices prevent exposure to helminth parasites, though epidemiological data suggest that people who carry helminths have fewer immune-mediated diseases[69,70]. Helminth infections may alter the abundance and composition of enteric microbiota, resulting in a greater proportion of anti-inflammatory strains[71]. The biocomplexity of the gut, including the interactions between parasites and the enteric microbiota, can influence inflammatory processes[72]. Studies in rodent models revealed that intestinal parasites and parasite products regulate host immunity and alleviate IBD-like inflammation[73]. In a randomized, double-blind, placebo-controlled trial involving 54 patients with active UC, a greater number of patients who received 12 wk of treatment with Trichuris suis ova showed improvement with no side effects[74]. Similarly, three patients with active UC in an open trial attained remission with 12-wk T. suis ova treatment with no side effects[75].

Dietary polyphenols

Diet can modulate enteric microbiota composition, with a strong effect on human health[76]. Polyphenols are members of a large family of plant-derived compounds that have drawn attention due to their antioxidant and anti-inflammatory properties[77]. Some in vitro and in vivo studies indicate that dietary polyphenols and their metabolites can interact with the enteric microbiota, and enhance the growth of probiotic bacteria[78-84]. Due to the prebiotic potential and anti-inflammatory effects, polyphenol supplementation could potentially serve as a complementary medicinal approach to UC. However, there have been no clinical trials evaluating the use of dietary polyphenols for the treatment of UC.

CONCLUSION

As a form of IBD, UC has its own genetic, pathogenic and therapeutic identity. Despite recent advances in UC therapeutic resources, a considerable proportion of UC patients are still refractory to conventional treatment. Dysbiosis is an important immunologic and pathologic process in UC. Accumulating knowledge of dysbiosis has promoted the use of enteric microbiotal regulation as a novel promising adjuvant in UC therapy. Therapeutic approaches, such as FMT, probiotics, antibiotics, dietary polyphenols, and helminth therapy, can alter the abundance and composition of enteric microbiota, and improve patient outcomes. However, the safety and effectiveness of bacteria-driven therapies need further evaluation.

Footnotes

P- Reviewer: Hiraoka S, Kato J, Yang SK S- Editor: Qi Y L- Editor: Wang TQ E- Editor: Ma S

References
1.  Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annu Rev Immunol. 2010;28:573-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1352]  [Cited by in F6Publishing: 1444]  [Article Influence: 103.1]  [Reference Citation Analysis (0)]
2.  Shale M, Ghosh S. Beyond TNF, Th1 and Th2 in inflammatory bowel disease. Gut. 2008;57:1349-1351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
3.  Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, Benchimol EI, Panaccione R, Ghosh S, Barkema HW. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46-54.e42; quiz e30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3134]  [Cited by in F6Publishing: 3259]  [Article Influence: 271.6]  [Reference Citation Analysis (1)]
4.  Hill DA, Artis D. Intestinal bacteria and the regulation of immune cell homeostasis. Annu Rev Immunol. 2010;28:623-667.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 401]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
5.  Cader MZ, Kaser A. Recent advances in inflammatory bowel disease: mucosal immune cells in intestinal inflammation. Gut. 2013;62:1653-1664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 239]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
6.  Hisamatsu T, Kanai T, Mikami Y, Yoneno K, Matsuoka K, Hibi T. Immune aspects of the pathogenesis of inflammatory bowel disease. Pharmacol Ther. 2013;137:283-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 69]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
7.  Savage DC. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol. 1977;31:107-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1659]  [Cited by in F6Publishing: 1427]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
8.  Fraher MH, O’Toole PW, Quigley EM. Techniques used to characterize the gut microbiota: a guide for the clinician. Nat Rev Gastroenterol Hepatol. 2012;9:312-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 218]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
9.  Reiff C, Kelly D. Inflammatory bowel disease, gut bacteria and probiotic therapy. Int J Med Microbiol. 2010;300:25-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 108]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
10.  Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7052]  [Cited by in F6Publishing: 7119]  [Article Influence: 508.5]  [Reference Citation Analysis (3)]
11.  Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora. Science. 2005;308:1635-1638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5700]  [Cited by in F6Publishing: 5249]  [Article Influence: 276.3]  [Reference Citation Analysis (2)]
12.  Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307:1915-1920.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3394]  [Cited by in F6Publishing: 3308]  [Article Influence: 174.1]  [Reference Citation Analysis (4)]
13.  Guarner F. What is the role of the enteric commensal flora in IBD? Inflamm Bowel Dis. 2008;14 Suppl 2:S83-S84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
14.  Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90:859-904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2501]  [Cited by in F6Publishing: 2604]  [Article Influence: 186.0]  [Reference Citation Analysis (1)]
15.  Chassaing B, Darfeuille-Michaud A. The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel diseases. Gastroenterology. 2011;140:1720-1728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 338]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
16.  Barcenilla A, Pryde SE, Martin JC, Duncan SH, Stewart CS, Henderson C, Flint HJ. Phylogenetic relationships of butyrate-producing bacteria from the human gut. Appl Environ Microbiol. 2000;66:1654-1661.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Joossens M, Huys G, Cnockaert M, De Preter V, Verbeke K, Rutgeerts P, Vandamme P, Vermeire S. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut. 2011;60:631-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 692]  [Cited by in F6Publishing: 703]  [Article Influence: 54.1]  [Reference Citation Analysis (0)]
18.  Walker AW, Sanderson JD, Churcher C, Parkes GC, Hudspith BN, Rayment N, Brostoff J, Parkhill J, Dougan G, Petrovska L. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiol. 2011;11:7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 470]  [Cited by in F6Publishing: 499]  [Article Influence: 38.4]  [Reference Citation Analysis (0)]
19.  Manichanh C, Rigottier-Gois L, Bonnaud E, Gloux K, Pelletier E, Frangeul L, Nalin R, Jarrin C, Chardon P, Marteau P. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut. 2006;55:205-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1633]  [Cited by in F6Publishing: 1589]  [Article Influence: 88.3]  [Reference Citation Analysis (0)]
20.  Rajilić-Stojanović M, Shanahan F, Guarner F, de Vos WM. Phylogenetic analysis of dysbiosis in ulcerative colitis during remission. Inflamm Bowel Dis. 2013;19:481-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 247]  [Cited by in F6Publishing: 254]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
21.  Nemoto H, Kataoka K, Ishikawa H, Ikata K, Arimochi H, Iwasaki T, Ohnishi Y, Kuwahara T, Yasutomo K. Reduced diversity and imbalance of fecal microbiota in patients with ulcerative colitis. Dig Dis Sci. 2012;57:2955-2964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 105]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
22.  Michail S, Durbin M, Turner D, Griffiths AM, Mack DR, Hyams J, Leleiko N, Kenche H, Stolfi A, Wine E. Alterations in the gut microbiome of children with severe ulcerative colitis. Inflamm Bowel Dis. 2012;18:1799-1808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 203]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
23.  Kumari R, Ahuja V, Paul J. Fluctuations in butyrate-producing bacteria in ulcerative colitis patients of North India. World J Gastroenterol. 2013;19:3404-3414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 143]  [Cited by in F6Publishing: 143]  [Article Influence: 13.0]  [Reference Citation Analysis (2)]
24.  Kabeerdoss J, Sankaran V, Pugazhendhi S, Ramakrishna BS. Clostridium leptum group bacteria abundance and diversity in the fecal microbiota of patients with inflammatory bowel disease: a case-control study in India. BMC Gastroenterol. 2013;13:20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 86]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
25.  Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, Ballet V, Claes K, Van Immerseel F, Verbeke K. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014;63:1275-1283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1036]  [Cited by in F6Publishing: 1125]  [Article Influence: 112.5]  [Reference Citation Analysis (2)]
26.  Fite A, Macfarlane S, Furrie E, Bahrami B, Cummings JH, Steinke DT, Macfarlane GT. Longitudinal analyses of gut mucosal microbiotas in ulcerative colitis in relation to patient age and disease severity and duration. J Clin Microbiol. 2013;51:849-856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
27.  Varela E, Manichanh C, Gallart M, Torrejón A, Borruel N, Casellas F, Guarner F, Antolin M. Colonisation by Faecalibacterium prausnitzii and maintenance of clinical remission in patients with ulcerative colitis. Aliment Pharmacol Ther. 2013;38:151-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 158]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
28.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 272]  [Cited by in F6Publishing: 320]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
29.  Martinez C, Antolin M, Santos J, Torrejon A, Casellas F, Borruel N, Guarner F, Malagelada JR. Unstable composition of the fecal microbiota in ulcerative colitis during clinical remission. Am J Gastroenterol. 2008;103:643-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 138]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
30.  Robles Alonso V, Guarner F. Linking the gut microbiota to human health. Br J Nutr. 2013;109 Suppl 2:S21-S26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 187]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
31.  Dignass A, Lindsay JO, Sturm A, Windsor A, Colombel JF, Allez M, D’Haens G, D’Hoore A, Mantzaris G, Novacek G. Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 2: current management. J Crohns Colitis. 2012;6:991-1030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 728]  [Cited by in F6Publishing: 683]  [Article Influence: 56.9]  [Reference Citation Analysis (0)]
32.  Torres J, Danese S, Colombel JF. New therapeutic avenues in ulcerative colitis: thinking out of the box. Gut. 2013;62:1642-1652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
33.  Smits LP, Bouter KE, de Vos WM, Borody TJ, Nieuwdorp M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology. 2013;145:946-953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 420]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
34.  Bakken JS, Borody T, Brandt LJ, Brill JV, Demarco DC, Franzos MA, Kelly C, Khoruts A, Louie T, Martinelli LP. Treating Clostridium difficile infection with fecal microbiota transplantation. Clin Gastroenterol Hepatol. 2011;9:1044-1049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 650]  [Cited by in F6Publishing: 641]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
35.  Borody TJ, Warren EF, Leis S, Surace R, Ashman O. Treatment of ulcerative colitis using fecal bacteriotherapy. J Clin Gastroenterol. 2003;37:42-47.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Kunde S, Pham A, Bonczyk S, Crumb T, Duba M, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 213]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
37.  Kump PK, Gröchenig HP, Lackner S, Trajanoski S, Reicht G, Hoffmann KM, Deutschmann A, Wenzl HH, Petritsch W, Krejs GJ. Alteration of intestinal dysbiosis by fecal microbiota transplantation does not induce remission in patients with chronic active ulcerative colitis. Inflamm Bowel Dis. 2013;19:2155-2165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 169]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
38.  Angelberger S, Reinisch W, Makristathis A, Lichtenberger C, Dejaco C, Papay P, Novacek G, Trauner M, Loy A, Berry D. Temporal bacterial community dynamics vary among ulcerative colitis patients after fecal microbiota transplantation. Am J Gastroenterol. 2013;108:1620-1630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 245]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
39.  Rubin DT. Curbing our enthusiasm for fecal transplantation in ulcerative colitis. Am J Gastroenterol. 2013;108:1631-1633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
40.  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;11:1036-1038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 150]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
41.  Oelschlaeger TA. Mechanisms of probiotic actions - A review. Int J Med Microbiol. 2010;300:57-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 406]  [Cited by in F6Publishing: 378]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
42.  Bron PA, van Baarlen P, Kleerebezem M. Emerging molecular insights into the interaction between probiotics and the host intestinal mucosa. Nat Rev Microbiol. 2012;10:66-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 411]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
43.  Gibson GR, Probert HM, Loo JV, Rastall RA, Roberfroid MB. Dietary modulation of the human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev. 2004;17:259-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1563]  [Cited by in F6Publishing: 1299]  [Article Influence: 108.3]  [Reference Citation Analysis (1)]
44.  Gionchetti P, Rizzello F, Venturi A, Brigidi P, Matteuzzi D, Bazzocchi G, Poggioli G, Miglioli M, Campieri M. Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology. 2000;119:305-309.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Gionchetti P, Rizzello F, Helwig U, Venturi A, Lammers KM, Brigidi P, Vitali B, Poggioli G, Miglioli M, Campieri M. Prophylaxis of pouchitis onset with probiotic therapy: a double-blind, placebo-controlled trial. Gastroenterology. 2003;124:1202-1209.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Mimura T, Rizzello F, Helwig U, Poggioli G, Schreiber S, Talbot IC, Nicholls RJ, Gionchetti P, Campieri M, Kamm MA. Once daily high dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitis. Gut. 2004;53:108-114.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Miele E, Pascarella F, Giannetti E, Quaglietta L, Baldassano RN, Staiano A. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am J Gastroenterol. 2009;104:437-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 350]  [Cited by in F6Publishing: 321]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
48.  Kruis W, Fric P, Pokrotnieks J, Lukás M, Fixa B, Kascák M, Kamm MA, Weismueller J, Beglinger C, Stolte M. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut. 2004;53:1617-1623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 851]  [Cited by in F6Publishing: 761]  [Article Influence: 38.1]  [Reference Citation Analysis (0)]
49.  Rembacken BJ, Snelling AM, Hawkey PM, Chalmers DM, Axon AT. Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial. Lancet. 1999;354:635-639.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Zocco MA, dal Verme LZ, Cremonini F, Piscaglia AC, Nista EC, Candelli M, Novi M, Rigante D, Cazzato IA, Ojetti V. Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther. 2006;23:1567-1574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 276]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
51.  Sood A, Midha V, Makharia GK, Ahuja V, Singal D, Goswami P, Tandon RK. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol. 2009;7:1202-129, 1209.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in F6Publishing: 322]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
52.  Tursi A, Brandimarte G, Papa A, Giglio A, Elisei W, Giorgetti GM, Forti G, Morini S, Hassan C, Pistoia MA. Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a standard pharmaceutical treatment: a double-blind, randomized, placebo-controlled study. Am J Gastroenterol. 2010;105:2218-2227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 313]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
53.  Oliva S, Di Nardo G, Ferrari F, Mallardo S, Rossi P, Patrizi G, Cucchiara S, Stronati L. Randomised clinical trial: the effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis. Aliment Pharmacol Ther. 2012;35:327-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 176]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
54.  Hanai H, Kanauchi O, Mitsuyama K, Andoh A, Takeuchi K, Takayuki I, Araki Y, Fujiyama Y, Toyonaga A, Sata M. Germinated barley foodstuff prolongs remission in patients with ulcerative colitis. Int J Mol Med. 2004;13:643-647.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Kanauchi O, Mitsuyama K, Homma T, Takahama K, Fujiyama Y, Andoh A, Araki Y, Suga T, Hibi T, Naganuma M. Treatment of ulcerative colitis patients by long-term administration of germinated barley foodstuff: multi-center open trial. Int J Mol Med. 2003;12:701-704.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Kanauchi O, Suga T, Tochihara M, Hibi T, Naganuma M, Homma T, Asakura H, Nakano H, Takahama K, Fujiyama Y. Treatment of ulcerative colitis by feeding with germinated barley foodstuff: first report of a multicenter open control trial. J Gastroenterol. 2002;37 Suppl 14:67-72.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Ishikawa H, Matsumoto S, Ohashi Y, Imaoka A, Setoyama H, Umesaki Y, Tanaka R, Otani T. Beneficial effects of probiotic bifidobacterium and galacto-oligosaccharide in patients with ulcerative colitis: a randomized controlled study. Digestion. 2011;84:128-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 125]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
58.  Wildt S, Nordgaard I, Hansen U, Brockmann E, Rumessen JJ. A randomised double-blind placebo-controlled trial with Lactobacillus acidophilus La-5 and Bifidobacterium animalis subsp. lactis BB-12 for maintenance of remission in ulcerative colitis. J Crohns Colitis. 2011;5:115-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 93]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
59.  Matthes H, Krummenerl T, Giensch M, Wolff C, Schulze J. Clinical trial: probiotic treatment of acute distal ulcerative colitis with rectally administered Escherichia coli Nissle 1917 (EcN). BMC Complement Altern Med. 2010;10:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 126]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
60.  Sanders ME, Guarner F, Guerrant R, Holt PR, Quigley EM, Sartor RB, Sherman PM, Mayer EA. An update on the use and investigation of probiotics in health and disease. Gut. 2013;62:787-796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 345]  [Cited by in F6Publishing: 315]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
61.  Kang SS, Bloom SM, Norian LA, Geske MJ, Flavell RA, Stappenbeck TS, Allen PM. An antibiotic-responsive mouse model of fulminant ulcerative colitis. PLoS Med. 2008;5:e41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 104]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
62.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 704]  [Cited by in F6Publishing: 708]  [Article Influence: 44.3]  [Reference Citation Analysis (0)]
63.  Ohkusa T, Nomura T, Terai T, Miwa H, Kobayashi O, Hojo M, Takei Y, Ogihara T, Hirai S, Okayasu I. Effectiveness of antibiotic combination therapy in patients with active ulcerative colitis: a randomized, controlled pilot trial with long-term follow-up. Scand J Gastroenterol. 2005;40:1334-1342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 69]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
64.  Nomura T, Ohkusa T, Okayasu I, Yoshida T, Sakamoto M, Hayashi H, Benno Y, Hirai S, Hojo M, Kobayashi O. Mucosa-associated bacteria in ulcerative colitis before and after antibiotic combination therapy. Aliment Pharmacol Ther. 2005;21:1017-1027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 34]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
65.  Ohkusa T, Sato N, Ogihara T, Morita K, Ogawa M, Okayasu I. Fusobacterium varium localized in the colonic mucosa of patients with ulcerative colitis stimulates species-specific antibody. J Gastroenterol Hepatol. 2002;17:849-853.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Ohkusa T, Kato K, Terao S, Chiba T, Mabe K, Murakami K, Mizokami Y, Sugiyama T, Yanaka A, Takeuchi Y. Newly developed antibiotic combination therapy for ulcerative colitis: a double-blind placebo-controlled multicenter trial. Am J Gastroenterol. 2010;105:1820-1829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 92]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
68.  Uehara T, Kato K, Ohkusa T, Sugitani M, Ishii Y, Nemoto N, Moriyama M. Efficacy of antibiotic combination therapy in patients with active ulcerative colitis, including refractory or steroid-dependent cases. J Gastroenterol Hepatol. 2010;25 Suppl 1:S62-S66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
69.  Elliott DE, Weinstock JV. Helminthic therapy: using worms to treat immune-mediated disease. Adv Exp Med Biol. 2009;666:157-166.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Chu KM, Watermeyer G, Shelly L, Janssen J, May TD, Brink K, Benefeld G, Li X. Childhood helminth exposure is protective against inflammatory bowel disease: a case control study in South Africa. Inflamm Bowel Dis. 2013;19:614-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 35]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
71.  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;16:1841-1849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 241]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
72.  Berrilli F, Di Cave D, Cavallero S, D’Amelio S. Interactions between parasites and microbial communities in the human gut. Front Cell Infect Microbiol. 2012;2:141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 89]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
73.  Weinstock JV, Elliott DE. Helminths and the IBD hygiene hypothesis. Inflamm Bowel Dis. 2009;15:128-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 154]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
74.  Summers RW, Elliott DE, Urban JF, Thompson RA, Weinstock JV. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology. 2005;128:825-832.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  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;98:2034-2041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 325]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
76.  Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4098]  [Cited by in F6Publishing: 4107]  [Article Influence: 315.9]  [Reference Citation Analysis (0)]
77.  Selma MV, Espín JC, Tomás-Barberán FA. Interaction between phenolics and gut microbiota: role in human health. J Agric Food Chem. 2009;57:6485-6501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 908]  [Cited by in F6Publishing: 847]  [Article Influence: 56.5]  [Reference Citation Analysis (0)]
78.  Jin JS, Touyama M, Hisada T, Benno Y. Effects of green tea consumption on human fecal microbiota with special reference to Bifidobacterium species. Microbiol Immunol. 2012;56:729-739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 106]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
79.  Vendrame S, Guglielmetti S, Riso P, Arioli S, Klimis-Zacas D, Porrini M. Six-week consumption of a wild blueberry powder drink increases bifidobacteria in the human gut. J Agric Food Chem. 2011;59:12815-12820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 206]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
80.  Tzounis X, Rodriguez-Mateos A, Vulevic J, Gibson GR, Kwik-Uribe C, Spencer JP. Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study. Am J Clin Nutr. 2011;93:62-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 380]  [Cited by in F6Publishing: 340]  [Article Influence: 26.2]  [Reference Citation Analysis (0)]
81.  Larrosa M, González-Sarrías A, Yáñez-Gascón MJ, Selma MV, Azorín-Ortuño M, Toti S, Tomás-Barberán F, Dolara P, Espín JC. Anti-inflammatory properties of a pomegranate extract and its metabolite urolithin-A in a colitis rat model and the effect of colon inflammation on phenolic metabolism. J Nutr Biochem. 2010;21:717-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 313]  [Cited by in F6Publishing: 324]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
82.  Bialonska D, Ramnani P, Kasimsetty SG, Muntha KR, Gibson GR, Ferreira D. The influence of pomegranate by-product and punicalagins on selected groups of human intestinal microbiota. Int J Food Microbiol. 2010;140:175-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 163]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
83.  Larrosa M, Yañéz-Gascón MJ, Selma MV, González-Sarrías A, Toti S, Cerón JJ, Tomás-Barberán F, Dolara P, Espín JC. Effect of a low dose of dietary resveratrol on colon microbiota, inflammation and tissue damage in a DSS-induced colitis rat model. J Agric Food Chem. 2009;57:2211-2220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 238]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
84.  Lee HC, Jenner AM, Low CS, Lee YK. Effect of tea phenolics and their aromatic fecal bacterial metabolites on intestinal microbiota. Res Microbiol. 2006;157:876-884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 474]  [Cited by in F6Publishing: 460]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]