201
|
Abstract
Despite being one of the most common conditions leading to gastroenterological referral, irritable bowel syndrome (IBS) is poorly understood. However, recent years have seen major advances. These include new understanding of the role of both inflammation and altered microbiota as well as the impact of dietary intolerances as illuminated by magnetic resonance imaging (MRI), which has thrown new light on IBS. This article will review new data on how excessive bile acid secretion mediates diarrhea and evidence from post infectious IBS which has shown how gut inflammation can alter gut microbiota and function. Studies of patients with inflammatory bowel disease (IBD) have also shown that even when inflammation is in remission, the altered enteric nerves and abnormal microbiota can generate IBS-like symptoms. The efficacy of the low FODMAP diet as a treatment for bloating, flatulence, and abdominal discomfort has been demonstrated by randomized controlled trials. MRI studies, which can quantify intestinal volumes, have provided new insights into how FODMAPs cause symptoms. This article will focus on these areas together with recent trials of new agents, which this author believes will alter clinical practice within the foreseeable future.
Collapse
Affiliation(s)
- Robin Spiller
- Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
202
|
Kamada N, Rogler G. The Innate Immune System: A Trigger for Many Chronic Inflammatory Intestinal Diseases. Inflamm Intest Dis 2016; 1:70-77. [PMID: 29922660 DOI: 10.1159/000445261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 03/08/2016] [Indexed: 12/31/2022] Open
Abstract
Background Mononuclear phagocytes, such as monocytes, macrophages, and dendritic cells, are important cellular components of the innate immune system that contribute to the pathogenesis of many intestinal inflammatory diseases. Summary While mononuclear phagocytes play a key role in the induction of inflammation in many different tissues through production of pro-inflammatory cytokines and chemokines (such as IL-1, TNF, IL-6, IL-8 and MCP-1), free oxygen radicals (also termed 'oxidative burst'), proteases (such as cathepsins) and tissue-degrading enzymes (such as metalloproteinases), resident macrophages as well as dendritic cells in the intestine display an anergic and 'tolerogenic' phenotype mediating tolerance to commensal bacteria. In recent years many single nucleotide polymorphisms (SNPs) in genes mainly expressed in the above-mentioned cell types have been identified to convey an increased risk of autoimmune diseases. SNPs in the NOD2, ATG16L1 and TNFSF15 genes, which are involved in the function of the innate immune cells, are identified as risk factors for Crohn's disease (CD). Of note, these genes are involved in the different functions in the innate immune cells. For example, while NOD2 is required for intracellular recognition of microbial components, ATG16L1 is involved in autophagy responses against intracellular microbes. Likewise, TNFSF15 contributes to the induction of inflammatory responses by innate immune cells. Furthermore, the frequency of mutations in these genes differs by ethnicity. Genetic variations in the NOD2 and ATG16L1 genes are associated with CD in Caucasians but much less in Eastern Asian populations, whereas SNPs in TNFSF15 are dominated in Asian populations. Thus, different genetic risks may eventually lead to similar impairments in innate immune cells, thereby developing the same disease in Western and Asian patients with CD. Key Messages Despite differences in risk genes, similar mechanisms associated with the innate immune system may trigger autoimmune and chronic inflammatory intestinal diseases in East and West.
Collapse
Affiliation(s)
- Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Mich., USA
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
203
|
Montassier E, Al-Ghalith GA, Ward T, Corvec S, Gastinne T, Potel G, Moreau P, de la Cochetiere MF, Batard E, Knights D. Pretreatment gut microbiome predicts chemotherapy-related bloodstream infection. Genome Med 2016; 8:49. [PMID: 27121964 PMCID: PMC4848771 DOI: 10.1186/s13073-016-0301-4] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023] Open
Abstract
Background Bacteremia, or bloodstream infection (BSI), is a leading cause of death among patients with certain types of cancer. A previous study reported that intestinal domination, defined as occupation of at least 30 % of the microbiota by a single bacterial taxon, is associated with BSI in patients undergoing allo-HSCT. However, the impact of the intestinal microbiome before treatment initiation on the risk of subsequent BSI remains unclear. Our objective was to characterize the fecal microbiome collected before treatment to identify microbes that predict the risk of BSI. Methods We sampled 28 patients with non-Hodgkin lymphoma undergoing allogeneic hematopoietic stem cell transplantation (HSCT) prior to administration of chemotherapy and characterized 16S ribosomal RNA genes using high-throughput DNA sequencing. We quantified bacterial taxa and used techniques from machine learning to identify microbial biomarkers that predicted subsequent BSI. Results We found that patients who developed subsequent BSI exhibited decreased overall diversity and decreased abundance of taxa including Barnesiellaceae, Coriobacteriaceae, Faecalibacterium, Christensenella, Dehalobacterium, Desulfovibrio, and Sutterella. Using machine-learning methods, we developed a BSI risk index capable of predicting BSI incidence with a sensitivity of 90 % at a specificity of 90 % based only on the pretreatment fecal microbiome. Conclusions These results suggest that the gut microbiota can identify high-risk patients before HSCT and that manipulation of the gut microbiota for prevention of BSI in high-risk patients may be a useful direction for future research. This approach may inspire the development of similar microbiome-based diagnostic and prognostic models in other diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0301-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emmanuel Montassier
- Université de Nantes, EA 3826 Thérapeutiques cliniques et expérimentales des infections. Faculté de médecine, 1 Rue G Veil, Nantes, 44000, France.,Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Gabriel A Al-Ghalith
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, 55455, USA.,Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Tonya Ward
- Biotechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Stephane Corvec
- Université de Nantes, EA 3826 Thérapeutiques cliniques et expérimentales des infections. Faculté de médecine, 1 Rue G Veil, Nantes, 44000, France.,Nantes University Hospital, Microbiology Laboratory, Nantes, France
| | - Thomas Gastinne
- Hematology Department, Nantes University Hospital, Nantes, France
| | - Gilles Potel
- Université de Nantes, EA 3826 Thérapeutiques cliniques et expérimentales des infections. Faculté de médecine, 1 Rue G Veil, Nantes, 44000, France
| | - Phillipe Moreau
- Hematology Department, Nantes University Hospital, Nantes, France
| | - Marie France de la Cochetiere
- Université de Nantes, EA 3826 Thérapeutiques cliniques et expérimentales des infections. Faculté de médecine, 1 Rue G Veil, Nantes, 44000, France
| | - Eric Batard
- Université de Nantes, EA 3826 Thérapeutiques cliniques et expérimentales des infections. Faculté de médecine, 1 Rue G Veil, Nantes, 44000, France
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, 55455, USA. .,Biotechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA.
| |
Collapse
|
204
|
Enck P, Aziz Q, Barbara G, Farmer AD, Fukudo S, Mayer EA, Niesler B, Quigley EMM, Rajilić-Stojanović M, Schemann M, Schwille-Kiuntke J, Simren M, Zipfel S, Spiller RC. Irritable bowel syndrome. Nat Rev Dis Primers 2016; 2:16014. [PMID: 27159638 PMCID: PMC5001845 DOI: 10.1038/nrdp.2016.14] [Citation(s) in RCA: 633] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disease with a high population prevalence. The disorder can be debilitating in some patients, whereas others may have mild or moderate symptoms. The most important single risk factors are female sex, younger age and preceding gastrointestinal infections. Clinical symptoms of IBS include abdominal pain or discomfort, stool irregularities and bloating, as well as other somatic, visceral and psychiatric comorbidities. Currently, the diagnosis of IBS is based on symptoms and the exclusion of other organic diseases, and therapy includes drug treatment of the predominant symptoms, nutrition and psychotherapy. Although the underlying pathogenesis is far from understood, aetiological factors include increased epithelial hyperpermeability, dysbiosis, inflammation, visceral hypersensitivity, epigenetics and genetics, and altered brain-gut interactions. IBS considerably affects quality of life and imposes a profound burden on patients, physicians and the health-care system. The past decade has seen remarkable progress in our understanding of functional bowel disorders such as IBS that will be summarized in this Primer.
Collapse
Affiliation(s)
- Paul Enck
- Department of Internal Medicine VI (Psychosomatic Medicine and Psychotherapy), University Hospital Tübingen, Tübingen, Germany
| | - Qasim Aziz
- Wingate Institute of Neurogastroenterology, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, St. Orsola-Malpighi Hospital, Bologna, Italy
| | - Adam D Farmer
- Wingate Institute of Neurogastroenterology, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shin Fukudo
- Department of Behavioural Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Emeran A Mayer
- Oppenheimer Center for Neurobiology of Stress, Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Beate Niesler
- Department of Human Molecular Genetics, University of Heidelberg, Heidelberg, Germany
| | - Eamonn M M Quigley
- Lynda K and David M Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital, Weill Cornell Medical College, Houston, Texas, USA
| | - Mirjana Rajilić-Stojanović
- Department of Biochemical Engineering and Biotechnology, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Michael Schemann
- Department of Human Biology, Technical University Munich, Freising-Weihenstephan, Germany
| | - Juliane Schwille-Kiuntke
- Department of Internal Medicine VI (Psychosomatic Medicine and Psychotherapy), University Hospital Tübingen, Tübingen, Germany
| | - Magnus Simren
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stephan Zipfel
- Department of Internal Medicine VI (Psychosomatic Medicine and Psychotherapy), University Hospital Tübingen, Tübingen, Germany
| | - Robin C Spiller
- NIHR Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| |
Collapse
|
205
|
Fourie NH, Wang D, Abey SK, Sherwin LB, Joseph PV, Rahim-Williams B, Ferguson EG, Henderson WA. The microbiome of the oral mucosa in irritable bowel syndrome. Gut Microbes 2016; 7:286-301. [PMID: 26963804 PMCID: PMC4988452 DOI: 10.1080/19490976.2016.1162363] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a poorly understood disorder characterized by persistent symptoms, including visceral pain. Studies have demonstrated oral microbiome differences in inflammatory bowel diseases suggesting the potential of the oral microbiome in the study of non-oral conditions. In this exploratory study we examine whether differences exist in the oral microbiome of IBS participants and healthy controls, and whether the oral microbiome relates to symptom severity. The oral buccal mucosal microbiome of 38 participants was characterized using PhyloChip microarrays. The severity of visceral pain was assessed by orally administering a gastrointestinal test solution. Participants self-reported their induced visceral pain. Pain severity was highest in IBS participants (P = 0.0002), particularly IBS-overweight participants (P = 0.02), and was robustly correlated to the abundance of 60 OTUs, 4 genera, 5 families and 4 orders of bacteria (r2 > 0.4, P < 0.001). IBS-overweight participants showed decreased richness in the phylum Bacteroidetes (P = 0.007) and the genus Bacillus (P = 0.008). Analysis of β-diversity found significant separation of the IBS-overweight group (P < 0.05). Our oral microbial results are concordant with described fecal and colonic microbiome-IBS and -weight associations. Having IBS and being overweight, rather than IBS-subtypes, was the most important factor in describing the severity of visceral pain and variation in the microbiome. Pain severity was strongly correlated to the abundance of many taxa, suggesting the potential of the oral microbiome in diagnosis and patient phenotyping. The oral microbiome has potential as a source of microbial information in IBS.
Collapse
Affiliation(s)
- Nicolaas H. Fourie
- Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Dan Wang
- Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Sarah K. Abey
- Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - LeeAnne B. Sherwin
- Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Paule V. Joseph
- Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Bridgett Rahim-Williams
- National Institute on Minority Health and Health Disparities, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Eric G. Ferguson
- Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Wendy A. Henderson
- Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, DHHS, Bethesda, MD, USA
| |
Collapse
|
206
|
Quigley EMM. Overlapping irritable bowel syndrome and inflammatory bowel disease: less to this than meets the eye? Therap Adv Gastroenterol 2016; 9:199-212. [PMID: 26929782 PMCID: PMC4749858 DOI: 10.1177/1756283x15621230] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Though distinct in terms of pathology, natural history and therapeutic approach, irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD) have some features in common. These include shared symptomatology and largely similar demographics. However, in most instances, clinical presentation, together with laboratory, imaging and endoscopic findings will readily permit the differentiation of active IBD from IBS. More problematic is the situation where a subject with IBD, in apparent remission, continues to complain of symptoms which, in aggregate, satisfy commonly employed criteria for the diagnosis of IBS. Access to methodologies, such the assay for levels of calprotectin in feces, now allows identification of ongoing inflammation in some such individuals and prompts appropriate therapy. More challenging is the IBD patient with persisting symptoms and no detectable evidence of inflammation; is this coincident IBS, IBS triggered by IBD or an even more subtle level of IBD activity unrecognized by available laboratory or imaging methods? Arguments can be advanced for each of these proposals; lacking definitive data, this issue remains unresolved. The occurrence of IBS-type symptoms in the IBD patient, together with some data suggesting a very subtle level of 'inflammation' or 'immune activation' in IBS, raises other questions: is IBS a prodromal form of IBD; and are IBS and IBD part of the spectrum of the same disease? All of the available evidence indicates that the answer to both these questions should be a resounding 'no'. Indeed, the whole issue of overlap between IBS and IBD should be declared moot given their differing pathophysiologies, contrasting natural histories and divergent treatment paths. The limited symptom repertoire of the gastrointestinal tract may well be fundamental to the apparent confusion that has, of late, bedeviled this area.
Collapse
|
207
|
Altered gastrointestinal microbiota in irritable bowel syndrome and its modification by diet: probiotics, prebiotics and the low FODMAP diet. Proc Nutr Soc 2016; 75:306-18. [PMID: 26908093 DOI: 10.1017/s0029665116000021] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Irritable bowel syndrome (IBS) is a functional bowel disorder characterised by abdominal pain or discomfort with disordered defecation. This review describes the role of the gastrointestinal (GI) microbiota in the pathogenesis of IBS and how dietary strategies to manage symptoms impact on the microbial community. Evidence suggests a dysbiosis of the luminal and mucosal colonic microbiota in IBS, frequently characterised by a reduction in species of Bifidobacteria which has been associated with worse symptom profile. Probiotic supplementation trials suggest intentional modulation of the GI microbiota may be effective in treating IBS. A smaller number of prebiotic supplementation studies have also demonstrated effectiveness in IBS whilst increasing Bifidobacteria. In contrast, a novel method of managing IBS symptoms is the restriction of short-chain fermentable carbohydrates (low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet). Studies consistently demonstrate clinical effectiveness of the low FODMAP diet in patients with IBS. However, one unintentional consequence of this dietary intervention is its impact on the microbiota. This leads to an interesting paradox; namely, increasing luminal Bifidobacteria through probiotic supplementation is associated with a reduction in IBS symptoms while in direct conflict to this, the low FODMAP diet has clinical efficacy but markedly reduces luminal Bifidobacteria concentration. Given the multifactorial aetiology of IBS, the heterogeneity of symptoms and the complex and diverse nature of the microbiome, it is probable that both interventions are effective in patient subgroups. However combination treatment has never been explored and as such, presents an exciting opportunity for optimising clinical management, whilst preventing potentially deleterious effects on the GI microbiota.
Collapse
|
208
|
Abstract
Irritable bowel syndrome (IBS) is one of the most common gastrointestinal disorders worldwide. The economic impact of IBS on the health care system is substantial, as is the personal impact on patients. Patients with diarrhea-predominant IBS (IBS-D) comprise a substantial proportion of the overall IBS population. Primary care providers are often the first point of contact for patients with IBS-D and can accurately diagnose IBS after a careful history and examination without extensive diagnostic tests. Several pharmacologic treatments (eg, loperamide, alosetron, and antidepressants) and non-pharmacologic treatments (eg, dietary modification and probiotics) are available for IBS-D, but restrictions on use (eg, alosetron) or the lack of controlled trial data showing reductions in both global and individual IBS-D symptoms (eg, bloating, pain and stool frequency) emphasize the need for alternative treatment options. Two newer medications (eluxadoline and rifaximin) were approved in May 2015 for the treatment of IBS-D, and represent new treatment options for this common gastrointestinal condition.
Collapse
Affiliation(s)
- Brian E Lacy
- Division of Gastroenterology and Hepatology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| |
Collapse
|
209
|
Abstract
Irritable bowel syndrome (IBS) is one of the most common gastrointestinal disorders worldwide. The economic impact of IBS on the health care system is substantial, as is the personal impact on patients. Patients with diarrhea-predominant IBS (IBS-D) comprise a substantial proportion of the overall IBS population. Primary care providers are often the first point of contact for patients with IBS-D and can accurately diagnose IBS after a careful history and examination without extensive diagnostic tests. Several pharmacologic treatments (eg, loperamide, alosetron, and antidepressants) and non-pharmacologic treatments (eg, dietary modification and probiotics) are available for IBS-D, but restrictions on use (eg, alosetron) or the lack of controlled trial data showing reductions in both global and individual IBS-D symptoms (eg, bloating, pain and stool frequency) emphasize the need for alternative treatment options. Two newer medications (eluxadoline and rifaximin) were approved in May 2015 for the treatment of IBS-D, and represent new treatment options for this common gastrointestinal condition.
Collapse
Affiliation(s)
- Brian E Lacy
- Division of Gastroenterology and Hepatology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| |
Collapse
|
210
|
Moloney RD, Johnson AC, O'Mahony SM, Dinan TG, Greenwood‐Van Meerveld B, Cryan JF. Stress and the Microbiota-Gut-Brain Axis in Visceral Pain: Relevance to Irritable Bowel Syndrome. CNS Neurosci Ther 2016; 22:102-17. [PMID: 26662472 PMCID: PMC6492884 DOI: 10.1111/cns.12490] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 02/06/2023] Open
Abstract
Visceral pain is a global term used to describe pain originating from the internal organs of the body, which affects a significant proportion of the population and is a common feature of functional gastrointestinal disorders (FGIDs) such as irritable bowel syndrome (IBS). While IBS is multifactorial, with no single etiology to completely explain the disorder, many patients also experience comorbid behavioral disorders, such as anxiety or depression; thus, IBS is described as a disorder of the gut-brain axis. Stress is implicated in the development and exacerbation of visceral pain disorders. Chronic stress can modify central pain circuitry, as well as change motility and permeability throughout the gastrointestinal (GI) tract. More recently, the role of the gut microbiota in the bidirectional communication along the gut-brain axis, and subsequent changes in behavior, has emerged. Thus, stress and the gut microbiota can interact through complementary or opposing factors to influence visceral nociceptive behaviors. This review will highlight the evidence by which stress and the gut microbiota interact in the regulation of visceral nociception. We will focus on the influence of stress on the microbiota and the mechanisms by which microbiota can affect the stress response and behavioral outcomes with an emphasis on visceral pain.
Collapse
Affiliation(s)
- Rachel D. Moloney
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Present address:
Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Anthony C. Johnson
- Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Siobhain M. O'Mahony
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
| | - Timothy G. Dinan
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Psychiatry and Neurobehavioural ScienceUniversity College CorkCorkIreland
| | - Beverley Greenwood‐Van Meerveld
- Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
- V.A. Medical CenterOklahoma CityOKUSA
| | - John F. Cryan
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
| |
Collapse
|
211
|
Hod K, Ringel Y. Probiotics in functional bowel disorders. Best Pract Res Clin Gastroenterol 2016; 30:89-97. [PMID: 27048900 DOI: 10.1016/j.bpg.2016.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 01/07/2016] [Indexed: 01/31/2023]
Abstract
Functional bowel disorders (FBDs) are the most common gastrointestinal (GI) disorders seen by gastroenterologists and primary care physicians. The disorders affect patients functioning and quality of life (QOL) and are associated with significant healthcare burden. The current theory regarding the development of FBDs suggests brain-gut axis dysfunctions associated abnormal GI motility and sensation. Recent data suggest that alterations in the intestinal microbiota may have a role in the pathogenesis of FBDs; or at least have the potential to affect intestinal functions that are thought to be relevant to the development of functional GI symptoms. This has led to growing interest of healthcare providers and patients in targeting the intestinal microbiota for the treatment of FBDs. In this article we discuss the potential role probiotic interventions in the treatment of FBDs. We review the evidence from pre-clinical and clinical studies and discuss the current recommendations for the use of probiotics for FBDs in clinical practice.
Collapse
Affiliation(s)
- Keren Hod
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehuda Ringel
- Department of Gastroenterology, Rabin Medical Center, Petach Tikva, Israel; Department of Medicine, University of North Carolina School of Medicine at Chapel Hill, NC, USA
| |
Collapse
|
212
|
Andresen V, Löwe B, Broicher W, Riegel B, Fraedrich K, von Wulffen M, Gappmayer K, Wegscheider K, Treszl A, Rose M, Layer P, Lohse AW. Post-infectious irritable bowel syndrome (PI-IBS) after infection with Shiga-like toxin-producing Escherichia coli (STEC) O104:H4: A cohort study with prospective follow-up. United European Gastroenterol J 2016; 4:121-31. [PMID: 26966532 PMCID: PMC4766540 DOI: 10.1177/2050640615581113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In May/June 2011, the new Shiga-like toxin-producing Escherichia coli (STEC) strain O104:H4 caused the severest outbreak ever recorded of hemorrhagic enterocolitis in 3842 patients in Germany. OBJECTIVES As bacterial enterocolitis is an established risk factor of subsequent irritable bowel syndrome (IBS), we aimed to estimate prevalence and incidence of post-infectious (PI)-IBS after six and 12 months in a cohort of STEC O104:H4 patients and to prospectively identify associated somatic and psychometric risk factors. METHODS A total of 389 patients were studied prospectively at baseline and at six and 12 months after STEC infection using STEC disease-related questionnaires and validated instruments for IBS (Rome III) and psychological factors. Frequencies and logistic regression models using multiple imputations were applied to assess predictor variables. RESULTS Prevalence of IBS increased from 9.8% prior to STEC infection to 23.6% at six and 25.3% at 12 months after STEC infection. In patients without IBS symptoms prior to STEC infection, incidence of new IBS was 16.9%. Logistic regression models indicated higher somatization and anxiety scores as risk factors for, and mesalazine treatment during, STEC infection as the only significant protective factor against IBS. No other factor analyzed, including disease severity, showed an association. CONCLUSIONS PI-IBS rates following this unusually severe STEC outbreak were similar to what has been observed after other infectious gastroenteritis outbreaks. Our findings suggest that mesalazine may have reduced the risk of subsequent PI-IBS. As altered mucosal immune activity is a pivotal pathogenic factor in PI-IBS, our observation of a potential protective effect of mesalazine might be explained by its known modulatory action on mucosal immunity, and may warrant further investigation.
Collapse
Affiliation(s)
- Viola Andresen
- Department of Gastroenterology and Infectious Diseases
- Israelitic Hospital Hamburg and
| | - Bernd Löwe
- Department of Psychosomatic Medicine and Psychotherapy
- Schön Klinik Hamburg-Eilbek, Hamburg, Germany
| | - Wiebke Broicher
- Department of Psychosomatic Medicine and Psychotherapy
- Schön Klinik Hamburg-Eilbek, Hamburg, Germany
| | - Björn Riegel
- Department of Psychosomatic Medicine and Psychotherapy
- Schön Klinik Hamburg-Eilbek, Hamburg, Germany
| | | | | | - Kerrin Gappmayer
- Department of Psychosomatic Medicine and Psychotherapy
- Schön Klinik Hamburg-Eilbek, Hamburg, Germany
| | - Karl Wegscheider
- Department of Biostatistics and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - András Treszl
- Department of Biostatistics and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Rose
- Department of Psychosomatic Medicine, Center for Internal Medicine and Dermatology, Charité – Universitätsmedizin Berlin, Germany
- Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | |
Collapse
|
213
|
Abstract
Irritable bowel syndrome (IBS) is a common functional gastrointestinal (GI) disorder usually originated from gut dysfunction, and the mechanisms underlying IBS are not clear. IBS can seriously disrupt patient's normal routine, even though it is not life-threatening. With the development of high-throughput sequencing technology, a large number of studies have showed that intestinal flora imbalance does play an important role in the pathogenesis of IBS, especially Bifidobacterium. Bifidobacterium can resist the colonization and invasion of intestinal pathogenic bacteria, and enhance the intestinal epithelial barrier function. Besides, its metabolites also can improve the defense function of the intestinal tract. In the gut of patients with IBS, the number of Bifidobacteria is usually significantly reduced, suggesting that increasing the number of intestinal Bifidobacteria may play a positive role in the treatment of IBS. This paper summarizes the relationship between Bifidobacterium and IBS, and discusses the effect of Bifidobacterium in the adjuvant treatment of IBS.
Collapse
|
214
|
Abstract
Irritable bowel syndrome (IBS) is a multifactorial functional disorder with no clearly defined etiology or pathophysiology. Modern culture-independent techniques have improved the understanding of the gut microbiota’s composition and demonstrated that an altered gut microbiota profile might be found in at least some subgroups of IBS patients. Research on IBS from a microbial perspective is gaining momentum and advancing. This review will therefore highlight potential links between the gut microbiota and IBS by discussing the current knowledge of the gut microbiota; it will also illustrate bacterial-host interactions and how alterations to these interactions could exacerbate, induce or even help alleviate IBS.
Collapse
Affiliation(s)
- Sean M P Bennet
- Departments of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lena Ohman
- Departments of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Simren
- Departments of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
215
|
|
216
|
Abstract
PURPOSE OF REVIEW The study reviews recent publications that build on previous studies showing that acute enteric infection can produce persistent dysfunction in the lower gut (postinfectious irritable bowel syndrome) and proximal gut (postinfectious functional dyspepsia). The review addresses risk factors, the pathophysiological basis of persistent gut dysfunction, and the factors that initiate and maintain it. RECENT FINDINGS Recent work has identified several loci of host genetic predisposition to these syndromes that focus attention on host immune responses that may lead to gut dysfunction, including changes in intestinal barrier function and cytokine responses to the initial infection. Human and animal studies identify changes in the serotonergic and cannabinoid pathways regulating visceral pain responses and gut motility. Recent work has also focused attention on the putative role of the intestinal microbiota or dysbiosis in maintaining gut dysfunction and this is reviewed in depth. SUMMARY The development of long-term consequences following an acute episode of gastroenteritis reflects a convergence of host factors that include genetic predisposition and psychological factors, as well as the development of intestinal dysbiosis. It is anticipated that future research will generate biomarkers of susceptibility as well as novel microbiota-directed preventive and therapeutic strategies.
Collapse
|
217
|
Abstract
Irritable bowel syndrome (IBS) is the most prevalent and the best studied functional gastrointestinal disorder. The etiology and the pathogenesis of IBS are still not clear; however, recent studies have implicated a role for alterations in the intestinal microbiota (dysbiosis) in the pathophysiology of the disorder. Epidemiological observations have demonstrated that the development of IBS symptoms is often preceded by a disruption of the individual's normal intestinal microbiota, and microbiological studies have demonstrated compositional differences in the intestinal microbiota between patients with IBS patients and healthy controls. In addition, animal studies and a few recent human clinical studies have demonstrated that compositional changes in the intestinal microbiota in IBS are associated with relevant abnormal gastrointestinal and brain-gut axis functions that are often observed in patients with IBS. This article discusses points of interest from the current research on the microbiota-gut-brain interactions in IBS and highlights the relevance of the emerging data to our understanding of the disorder and the clinical implications for patients' care.
Collapse
|
218
|
Rangel I, Sundin J, Fuentes S, Repsilber D, de Vos WM, Brummer RJ. The relationship between faecal-associated and mucosal-associated microbiota in irritable bowel syndrome patients and healthy subjects. Aliment Pharmacol Ther 2015; 42:1211-21. [PMID: 26376728 DOI: 10.1111/apt.13399] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/02/2015] [Accepted: 08/19/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND The faecal-associated microbiota is commonly seen as a surrogate of the mucosal-associated microbiota. However, previous studies indicate that they are different. Furthermore, analyses of the mucosal microbiota are commonly done after standard bowel cleansing, affecting the microbial composition. AIM To compare the mucosal-associated microbiota, obtained from unprepared colon, with faecal-associated microbiota in healthy subjects and irritable bowel syndrome (IBS) patients. METHODS Faecal and mucosal biopsies were obtained from 33 IBS patients and 16 healthy controls. Of IBS patients, 49% belonged to the diarrhoea-predominant subgroup and 80% suffered from IBS symptoms during at least 5 years. Biopsies were collected from unprepared sigmoid colon and faecal samples a day before colonoscopy. Microbiota analyses were performed with a phylogenetic microarray and redundancy discriminant analysis. RESULTS The composition of the mucosal- and the faecal-associated microbiota in unprepared sigmoid colon differs significantly (P = 0.002). Clinical characteristics of IBS did not correlate with this difference. Bacteroidetes dominate the mucosal-associated microbiota. Firmicutes, Actinobacteria and Proteobacteria dominate the faecal-associated microbiota. Healthy subjects had a significantly higher (P < 0.005) abundance (1.9%) of the bacterial group uncultured Clostridiales I in the mucosal-associated microbiota than IBS patients (0.3%). Bacterial diversity was higher in faecal- compared with mucosal-associated microbiota in IBS patients (P < 0.005). No differences were found in healthy subjects. CONCLUSIONS Differences in the mucosal-associated microbiota between healthy individuals and IBS patients are minimal (one bacterial group) compared to differences in the faecal microbiota of both groups (53 bacterial groups). Microbial aberrations characterising IBS are more pronounced in the faeces than in the mucosa.
Collapse
Affiliation(s)
- I Rangel
- School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - J Sundin
- School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - S Fuentes
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - D Repsilber
- School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - W M de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands.,Departments of Bacteriology & Immunology and Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - R J Brummer
- School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
219
|
Abstract
Although probiotics have been used for many years by those who suffer from what would now be defined as irritable bowel syndrome (IBS), a scientific rationale for their use in this indication and clinical evidence to support their benefits have only emerged very recently. Evidence to support considering strategies, such as probiotics, that modulate the gut microbiome, in IBS, has been provided by laboratory studies implicating the microbiome and the host response to the enteric microenvironment in IBS, as well as in vitro and in vivo studies demonstrating the ability of various commensal bacteria to influence such relevant functions as motility, visceral sensation, gut barrier integrity, and brain-gut interactions. Clinical studies supporting a role for probiotics in the management of IBS predated such experimental data, and randomized controlled trials of probiotics in IBS continue to be reported. Their interpretation is hampered by the less than optimal quality of many studies; nevertheless, it is apparent that probiotics, as a category, do exert significant effects in IBS. Defining the optimal strain, dose, formulation, and duration of therapy is more challenging given the limitations of available data. There is also an urgent need for appropriately powered and rigorously designed clinical trials of appropriate duration of probiotics in IBS; such studies should also help to define those who are most likely to respond to probiotics. Future laboratory and translational research should attempt to define the mechanism(s) of action of probiotics in IBS and explore the response to bacterial components or products in this common and oftentimes troublesome disorder.
Collapse
Affiliation(s)
- Eamonn M M Quigley
- *Division of Gastroenterology and Hepatology and Department of Medicine, Houston Methodist Hospital †Department of Medicine, and Weill Cornell Medical College, Houston, TX
| |
Collapse
|
220
|
Paula H, Grover M, Halder SL, Locke GR, Schleck CD, Zinsmeister AR, Talley NJ. Non-enteric infections, antibiotic use, and risk of development of functional gastrointestinal disorders. Neurogastroenterol Motil 2015; 27:1580-6. [PMID: 26303310 PMCID: PMC4624515 DOI: 10.1111/nmo.12655] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 07/17/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gastrointestinal infections are risk factors for irritable bowel syndrome (IBS) and functional dyspepsia (FD). We investigated whether non-enteric infections and antibiotic exposure are also associated with the development of functional gastrointestinal disorders (FGIDs). METHODS In a nested case-control study, random samples of Olmsted County, MN, were mailed valid self-report questionnaires from 1988 through 1994, and then follow-up questionnaires from 1995 through 2003. Survey responders who did not report any FGID symptoms at baseline, but then reported such symptoms in at least one subsequent survey, were classified as new-onset cases. Age-matched controls were individuals who did not have symptoms at either the initial or subsequent surveys. KEY RESULTS The overall response rate was 78% to the initial survey and 52% to the follow-up survey. Based on the responses, 316 participants had a new onset of an FGID (43 IBS constipation, 95 IBS diarrhea, 25 IBS mixed, and 153 other FGIDs, including FD) and 250 did not (controls). Around 76% (241/316) of cases reported a non-enteric infection vs 66% (166/250) of the controls. The frequency of enteric infections was similar between the two groups. Of the new FGID cases, 83% had a non-enteric infection that was treated with antibiotic. In a logistic regression model, treatment with antibiotics for a non-gastrointestinal infection was associated with the development of an FGID (odds ratio = 1.90; 95% CI: 1.21-2.98; p = 0.005), after adjusting for age and sex. CONCLUSIONS & INFERENCES Based on a case-control study, treatment of a non-gastrointestinal infection with antibiotics appears to be a risk factor for development of an FGID.
Collapse
Affiliation(s)
- Helga Paula
- Medizinisch-Diagnostische Laboratorium West OG, Vienna, Austria
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Smita L. Halder
- Division of Gastroenterology, McMaster University, Hamilton, Ontario, Canada
| | - G. Richard Locke
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Cathy D. Schleck
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Alan R. Zinsmeister
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Nicholas J Talley
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA,Faculty of Health and Medicine, University of Newcastle, New Lambton, NSW, Australia
| |
Collapse
|
221
|
Jalanka J, Salonen A, Fuentes S, de Vos WM. Microbial signatures in post-infectious irritable bowel syndrome--toward patient stratification for improved diagnostics and treatment. Gut Microbes 2015; 6:364-9. [PMID: 26512631 PMCID: PMC4826089 DOI: 10.1080/19490976.2015.1096486] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a multifactorial and heterogeneous disorder estimated to affect over 10% of the Western population. A subset of the patients reports the start of the disease after an episode of gastroenteritis. The alterations in the intestinal microbiota of the post-infectious IBS (PI-IBS) patients were recently investigated in a British cohort and shown to differentiate from the healthy controls and resemble that of diarrhea-predominant IBS (IBS-D) patients. The altered 27 genus-like groups created a microbial signature, which could be used to objectively stratify patients and healthy controls. In this addendum, we combine the microbiota data derived from the British cohort with that of a recently reported Swedish PI-IBS cohort. Remarkably, robust and reproducible microbiota signatures were observed in these PI-IBS patients. We discuss these results with attention on the emerging role of microbiota in the classification, development and treatment of PI-IBS.
Collapse
Affiliation(s)
- Jonna Jalanka
- Department of Veterinary Biosciences; Microbiology; University of Helsinki; Helsinki, Finland,Correspondence to: Jonna Jalanka;
| | - Anne Salonen
- Department of Bacteriology and Immunology; Immunobiology Research Program; University of Helsinki; Helsinki, Finland
| | - Susana Fuentes
- Laboratory of Microbiology; Wageningen University; Wageningen, The Netherlands
| | - Willem M de Vos
- Department of Veterinary Biosciences; Microbiology; University of Helsinki; Helsinki, Finland,Department of Bacteriology and Immunology; Immunobiology Research Program; University of Helsinki; Helsinki, Finland,Laboratory of Microbiology; Wageningen University; Wageningen, The Netherlands
| |
Collapse
|
222
|
Sundin J, Rangel I, Repsilber D, Brummer RJ. Cytokine Response after Stimulation with Key Commensal Bacteria Differ in Post-Infectious Irritable Bowel Syndrome (PI-IBS) Patients Compared to Healthy Controls. PLoS One 2015; 10:e0134836. [PMID: 26366730 PMCID: PMC4569289 DOI: 10.1371/journal.pone.0134836] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/15/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Microbial dysbiosis and prolonged immune activation resulting in low-grade inflammation and intestinal barrier dysfunction have been suggested to be underlying causes of post-infectious irritable bowel syndrome (PI-IBS). The aim of this study was to evaluate the difference in cytokine response between mucosal specimens of PI-IBS patients and healthy controls (HC) after ex vivo stimulation with key anaerobic bacteria. METHODS Colonic biopsies from 11 PI-IBS patients and 10 HC were stimulated ex vivo with the commensal bacteria Bacteroides ovatus, Ruminococcus gnavus, Akkermansia muciniphila, Subdoligranulum variabile and Eubacterium limosum, respectively. The cytokine release (IL-1β, IL-2, IL-8, IL-10, IL-13, IL-17, TNF-α and IFN-γ) in stimulation supernatants was analyzed using the LUMINEX assay. Comparison of cytokine release between PI-IBS patients and healthy controls was performed taking both unstimulated and bacterially stimulated mucosal specimens into account. KEY RESULTS IL-13 release from mucosal specimens without bacterial stimulation was significantly lower in PI-IBS patients compared to HC (p < 0.05). After stimulation with Subdoligranulum variabile, IL-1β release from PI-IBS patients was significantly increased compared to HC (p < 0.05). Stimulation with Eubacterium limosum resulted in a significantly decreased IL-10 release in HC compared to PI-IBS patients (p < 0.05) and a tendency to decreased IL-13 release in HC compared to PI-IBS patients (p = 0.07). CONCLUSIONS & INFERENCES PI-IBS patients differ from HC with regard to cytokine release ex vivo after stimulation with selected commensal bacteria. Hence, our results support that the pathogenesis of PI-IBS comprises an altered immune response against commensal gut microbes.
Collapse
Affiliation(s)
- Johanna Sundin
- School of Health and Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ignacio Rangel
- School of Health and Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Dirk Repsilber
- School of Health and Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Robert-Jan Brummer
- School of Health and Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
223
|
Kolmeder CA, Ritari J, Verdam FJ, Muth T, Keskitalo S, Varjosalo M, Fuentes S, Greve JW, Buurman WA, Reichl U, Rapp E, Martens L, Palva A, Salonen A, Rensen SS, de Vos WM. Colonic metaproteomic signatures of active bacteria and the host in obesity. Proteomics 2015; 15:3544-52. [DOI: 10.1002/pmic.201500049] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/03/2015] [Accepted: 07/24/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Carolin A. Kolmeder
- Department of Veterinary Biosciences; University of Helsinki; Helsinki Finland
| | - Jarmo Ritari
- Department of Veterinary Biosciences; University of Helsinki; Helsinki Finland
| | - Froukje J. Verdam
- Department of General Surgery; NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University Medical Center; Maastricht The Netherlands
| | - Thilo Muth
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; Magdeburg Germany
| | - Salla Keskitalo
- Institute of Biotechnology; University of Helsinki; Helsinki Finland
| | - Markku Varjosalo
- Institute of Biotechnology; University of Helsinki; Helsinki Finland
| | - Susana Fuentes
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
| | - Jan Willem Greve
- Department of General Surgery; NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University Medical Center; Maastricht The Netherlands
| | - Wim A. Buurman
- Department of General Surgery; NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University Medical Center; Maastricht The Netherlands
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; Magdeburg Germany
- Chair of Bioprocess Engineering; Otto-von-Guericke University; Magdeburg Germany
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; Magdeburg Germany
| | - Lennart Martens
- Department of Biochemistry; Ghent University; Ghent Belgium
- VIB Medical Biotechnology Center; Department of Medical Protein Research; Ghent Belgium
| | - Airi Palva
- Department of Veterinary Biosciences; University of Helsinki; Helsinki Finland
| | - Anne Salonen
- Department of Bacteriology and Immunology; Immunobiology Research Program; University of Helsinki; Helsinki Finland
| | - Sander S. Rensen
- Department of General Surgery; NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University Medical Center; Maastricht The Netherlands
| | - Willem M. de Vos
- Department of Veterinary Biosciences; University of Helsinki; Helsinki Finland
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
- Department of Bacteriology and Immunology; Immunobiology Research Program; University of Helsinki; Helsinki Finland
| |
Collapse
|
224
|
Pozuelo M, Panda S, Santiago A, Mendez S, Accarino A, Santos J, Guarner F, Azpiroz F, Manichanh C. Reduction of butyrate- and methane-producing microorganisms in patients with Irritable Bowel Syndrome. Sci Rep 2015; 5:12693. [PMID: 26239401 PMCID: PMC4523847 DOI: 10.1038/srep12693] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/03/2015] [Indexed: 12/19/2022] Open
Abstract
The pathophysiology of irritable bowel syndrome (IBS) remains unclear. Here we investigated the microbiome of a large cohort of patients to identify specific signatures for IBS subtypes. We examined the microbiome of 113 patients with IBS and 66 healthy controls. A subset of these participants provided two samples one month apart. We analyzed a total of 273 fecal samples, generating more than 20 million 16S rRNA sequences. In patients with IBS, a significantly lower microbial diversity was associated with a lower relative abundance of butyrate-producing bacteria (P = 0.002; q < 0.06), in particular in patients with IBS-D and IBS-M. IBS patients who did not receive any treatment harboured a lower abundance of Methanobacteria compared to healthy controls (P = 0.005; q = 0.05). Furthermore, significant correlations were observed between several bacterial taxa and sensation of flatulence and abdominal pain (P < 0.05). Altogether, our findings showed that IBS-M and IBS-D patients are characterized by a reduction of butyrate producing bacteria, known to improve intestinal barrier function, and a reduction of methane producing microorganisms a major mechanism of hydrogen disposal in the human colon, which could explain excess of abdominal gas in IBS.
Collapse
Affiliation(s)
- Marta Pozuelo
- Digestive System Research Unit, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Suchita Panda
- Digestive System Research Unit, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Alba Santiago
- Digestive System Research Unit, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Sara Mendez
- Digestive Unit, University Hospital Vall d'Hebron, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Anna Accarino
- 1] Digestive Unit, University Hospital Vall d'Hebron, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain [2] Centro de Investigacion Biomedica en Red en el Área tematica de Enfermedades Heptaticas y Digestivas, CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Santos
- 1] Digestive System Research Unit, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain [2] Digestive Unit, University Hospital Vall d'Hebron, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain [3] Centro de Investigacion Biomedica en Red en el Área tematica de Enfermedades Heptaticas y Digestivas, CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Guarner
- 1] Digestive System Research Unit, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain [2] Digestive Unit, University Hospital Vall d'Hebron, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain [3] Centro de Investigacion Biomedica en Red en el Área tematica de Enfermedades Heptaticas y Digestivas, CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Fernando Azpiroz
- 1] Digestive System Research Unit, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain [2] Digestive Unit, University Hospital Vall d'Hebron, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain [3] Centro de Investigacion Biomedica en Red en el Área tematica de Enfermedades Heptaticas y Digestivas, CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Chaysavanh Manichanh
- 1] Digestive System Research Unit, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain [2] Centro de Investigacion Biomedica en Red en el Área tematica de Enfermedades Heptaticas y Digestivas, CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
225
|
Kump PK, Krause R, Allerberger F, Högenauer C. Faecal microbiota transplantation--the Austrian approach. Clin Microbiol Infect 2015; 20:1106-11. [PMID: 25274251 DOI: 10.1111/1469-0691.12801] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The intestinal microbiome is essential for maintaining human health and defending against intestinal pathogens. Alterations of the intestinal microbiota, also termed dysbiosis, play a pivotal role in the pathogenesis of various human diseases. Faecal microbiota transplantation (FMT) is aimed at correcting these alterations by delivering faecal microorganisms from a healthy person to the intestines of a patient. At present, recurrent Clostridium difficile infection is the only indication supported by solid scientific evidence, but many ongoing studies are investigating FMT in other dysbiosis-related diseases, such as inflammatory bowel disease. As there are no systematic methodological investigations, several questions about techniques, donor screening and safety issues remain. This shortage of evidence, especially on long-term safety concerns, is leading to worldwide controversy regarding the use of FMT. Regulations by healthcare authorities vary among different countries. This review reflects the Austrian situation and its FMT guidelines concerning indications, techniques and donor screening, recently developed by local scientific societies.
Collapse
Affiliation(s)
- P K Kump
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | | | | | | |
Collapse
|
226
|
Affiliation(s)
- Pamela J Hornby
- Janssen Research & Development, Cardiovascular and Metabolic Disease, Janssen Pharmaceutical Companies of Johnson and Johnson, SH42-2508-A, 1400 McKean Road, Spring House, PA 19477, USA
| |
Collapse
|
227
|
El-Salhy M. Recent developments in the pathophysiology of irritable bowel syndrome. World J Gastroenterol 2015; 21:7621-7636. [PMID: 26167065 PMCID: PMC4491952 DOI: 10.3748/wjg.v21.i25.7621] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 03/31/2015] [Accepted: 05/21/2015] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common gastrointestinal disorder, the pathophysiology of which is not completely known, although it has been shown that genetic/social learning factors, diet, intestinal microbiota, intestinal low-grade inflammation, and abnormal gastrointestinal endocrine cells play a major role. Studies of familial aggregation and on twins have confirmed the heritability of IBS. However, the proposed IBS risk genes are thus far nonvalidated hits rather than true predisposing factors. There is no convincing evidence that IBS patients suffer from food allergy/intolerance, with the effect exerted by diet seemingly caused by intake of poorly absorbed carbohydrates and fiber. Obesity is a possible comorbidity of IBS. Differences in the microbiota between IBS patients and healthy controls have been reported, but the association between IBS symptoms and specific bacterial species is uncertain. Low-grade inflammation appears to play a role in the pathophysiology of a major subset of IBS, namely postinfectious IBS. The density of intestinal endocrine cells is reduced in patients with IBS, possibly as a result of genetic factors, diet, intestinal microbiota, and low-grade inflammation interfering with the regulatory signals controlling the intestinal stem-cell clonogenic and differentiation activities. Furthermore, there is speculation that this decreased number of endocrine cells is responsible for the visceral hypersensitivity, disturbed gastrointestinal motility, and abnormal gut secretion seen in IBS patients.
Collapse
|
228
|
Camilleri M, Carlson P, Acosta A, Busciglio I. Colonic mucosal gene expression and genotype in irritable bowel syndrome patients with normal or elevated fecal bile acid excretion. Am J Physiol Gastrointest Liver Physiol 2015; 309:G10-20. [PMID: 25930081 PMCID: PMC4491506 DOI: 10.1152/ajpgi.00080.2015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/28/2015] [Indexed: 02/08/2023]
Abstract
The mucosal gene expression in rectosigmoid mucosa (RSM) in irritable bowel syndrome with diarrhea (IBS-D) is unknown. Our objectives were, first, to study mRNA expression [by RT(2) PCR of 19 genes pertaining to tight junctions, immune activation, intestinal ion transport and bile acid (BA) homeostasis] in RSM in IBS-D patients (n = 47) and healthy controls (n = 17) and study expression of a selected protein (PDZD3) in 10 IBS-D patients and 4 healthy controls; second, to assess RSM mRNA expression according to genotype and fecal BA excretion (high ≥ 2,337 μmol/48 h); and third, to determine whether genotype or mucosal mRNA expression is associated with colonic transit or BA parameters. Fold changes were corrected for false detection rate for 19 genes studied (P < 0.00263). In RSM in IBS-D patients compared with controls, mRNA expression of GUC2AB, PDZD3, and PR2Y4 was increased, whereas CLDN1 and FN1 were decreased. One immune-related gene was upregulated (C4BP4) and one downregulated (CCL20). There was increased expression of a selected ion transport protein (PDZD3) on immunohistochemistry and Western blot in IBS-D compared with controls (P = 0.02). There were no significant differences in mucosal mRNA in 20 IBS-D patients with high compared with 27 IBS-D patients with normal BA excretion. GPBAR1 (P < 0.05) was associated with colonic transit. We concluded that mucosal ion transport mRNA (for several genes and PDZD3 protein) is upregulated and barrier protein mRNA downregulated in IBS-D compared with healthy controls, independent of genotype. There are no differences in gene expression in IBS-D with high compared with normal fecal BA excretion.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| | | | | | | |
Collapse
|
229
|
Abstract
Tremendous progress has been made in characterizing the bidirectional interactions between the central nervous system, the enteric nervous system, and the gastrointestinal tract. A series of provocative preclinical studies have suggested a prominent role for the gut microbiota in these gut-brain interactions. Based on studies using rodents raised in a germ-free environment, the gut microbiota appears to influence the development of emotional behavior, stress- and pain-modulation systems, and brain neurotransmitter systems. Additionally, microbiota perturbations by probiotics and antibiotics exert modulatory effects on some of these measures in adult animals. Current evidence suggests that multiple mechanisms, including endocrine and neurocrine pathways, may be involved in gut microbiota-to-brain signaling and that the brain can in turn alter microbial composition and behavior via the autonomic nervous system. Limited information is available on how these findings may translate to healthy humans or to disease states involving the brain or the gut/brain axis. Future research needs to focus on confirming that the rodent findings are translatable to human physiology and to diseases such as irritable bowel syndrome, autism, anxiety, depression, and Parkinson's disease.
Collapse
|
230
|
Keller J, Andresen V. Darmmikrobiom und funktionelle gastrointestinale Erkrankungen. GASTROENTEROLOGE 2015. [DOI: 10.1007/s11377-014-0964-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
231
|
Schokker D, Zhang J, Vastenhouw SA, Heilig HGHJ, Smidt H, Rebel JMJ, Smits MA. Long-lasting effects of early-life antibiotic treatment and routine animal handling on gut microbiota composition and immune system in pigs. PLoS One 2015; 10:e0116523. [PMID: 25658611 PMCID: PMC4319779 DOI: 10.1371/journal.pone.0116523] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/22/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In intensive pig husbandry systems, antibiotics are frequently administrated during early life stages to prevent respiratory and gastro-intestinal tract infections, often in combination with stressful handlings. The immediate effects of these treatments on microbial colonization and immune development have been described recently. Here we studied whether the early life administration of antibiotics has long-lasting effects on the pig's intestinal microbial community and on gut functionality. METHODOLOGY/PRINCIPAL FINDINGS To investigate the long-lasting effect of early-life treatment, piglets were divided into three different groups receiving the following treatments: 1) no antibiotics and no stress, 2) antibiotics and no stress, and 3) antibiotics and stress. All treatments were applied at day four after birth. Sampling of jejunal content for community scale microbiota analysis, and jejunal and ileal tissue for genome-wide transcription profiling, was performed at day 55 (~8 weeks) and day 176 (~25 weeks) after birth. Antibiotic treatment in combination with or without exposure to stress was found to have long-lasting effects on host intestinal gene expression involved in a multitude of processes, including immune related processes. CONCLUSIONS/SIGNIFICANCE The results obtained in this study indicate that early life (day 4 after birth) perturbations have long-lasting effects on the gut system, both in gene expression (day 55) as well as on microbiota composition (day 176). At day 55 high variance was observed in the microbiota data, but no significant differences between treatment groups, which is most probably due to the newly acquired microbiota during and right after weaning (day 28). Based on the observed difference in gene expression at day 55, it is hypothesized that due to the difference in immune programming during early life, the systems respond differently to the post-weaning newly acquired microbiota. As a consequence, the gut systems of the treatment groups develop into different homeostasis.
Collapse
Affiliation(s)
- Dirkjan Schokker
- Wageningen UR Livestock Research, P.O. box 65, 8200 AB Lelystad, The Netherlands
- * E-mail:
| | - Jing Zhang
- Laboratory of Microbiology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, The Netherlands
| | | | - Hans G. H. J. Heilig
- Laboratory of Microbiology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, The Netherlands
| | - Johanna M. J. Rebel
- Central Veterinary Institute, P.O. box 65, 8200 AB Lelystad, The Netherlands
| | - Mari A. Smits
- Wageningen UR Livestock Research, P.O. box 65, 8200 AB Lelystad, The Netherlands
- Central Veterinary Institute, P.O. box 65, 8200 AB Lelystad, The Netherlands
| |
Collapse
|
232
|
Sundin J, Rangel I, Fuentes S, Heikamp-de Jong I, Hultgren-Hörnquist E, de Vos WM, Brummer RJ. Altered faecal and mucosal microbial composition in post-infectious irritable bowel syndrome patients correlates with mucosal lymphocyte phenotypes and psychological distress. Aliment Pharmacol Ther 2015; 41:342-51. [PMID: 25521822 DOI: 10.1111/apt.13055] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 09/30/2014] [Accepted: 11/26/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND A subset of irritable bowel syndrome (IBS) patients, denoted post-infectious IBS (PI-IBS), develop symptoms after an enteric infection. Bacterial dysbiosis and mucosal inflammation have been proposed to be involved in the pathophysiology of this entity. AIM To characterise the mucosal and faecal microbiota in PI-IBS, general IBS and healthy controls, and to investigate associations between the microbiota and the mucosal immune system. METHODS Mucosal biopsies and faeces were collected from 13 PI-IBS patients, 19 general IBS patients and 16 healthy controls. Global bacterial composition was determined by generating 16S rRNA amplicons that were examined by phylogenetic microarray hybridisation, principal component and redundancy analysis. We correlated previously reported lymphocyte proportions with the microbiota. RESULTS Faecal microbiota composition of PI-IBS patients differed significantly from both general IBS patients and healthy controls (P < 0.02). Both mucosal (P < 0.01) and faecal (P = 0.05) microbial diversity were reduced in PI-IBS compared to healthy controls. In the intraepithelial lymphocytes the previously published proportion of CD8(+) CD45RA(+) was negatively correlated with mucosal microbial diversity (P < 0.005). The previously published number of lamina propria lymphocytes was negatively correlated with mucosal microbial diversity (P < 0.05). Faecal microbial diversity was significantly negatively correlated with the Hospital Anxiety and Depression scale (P < 0.05). CONCLUSIONS We present data that distinguishes the intestinal microbiota of PI-IBS patients from that of both general IBS patients and HC. The microbial composition is significantly associated with the HADs score and alterations in lymphocyte subsets proportions.
Collapse
Affiliation(s)
- J Sundin
- School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | | | | | | | | | | | | |
Collapse
|
233
|
Rajilić-Stojanović M, Jonkers DM, Salonen A, Hanevik K, Raes J, Jalanka J, de Vos WM, Manichanh C, Golic N, Enck P, Philippou E, Iraqi FA, Clarke G, Spiller RC, Penders J. Intestinal microbiota and diet in IBS: causes, consequences, or epiphenomena? Am J Gastroenterol 2015; 110:278-87. [PMID: 25623659 PMCID: PMC4317767 DOI: 10.1038/ajg.2014.427] [Citation(s) in RCA: 265] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 12/11/2022]
Abstract
Irritable bowel syndrome (IBS) is a heterogeneous functional disorder with a multifactorial etiology that involves the interplay of both host and environmental factors. Among environmental factors relevant for IBS etiology, the diet stands out given that the majority of IBS patients report their symptoms to be triggered by meals or specific foods. The diet provides substrates for microbial fermentation, and, as the composition of the intestinal microbiota is disturbed in IBS patients, the link between diet, microbiota composition, and microbial fermentation products might have an essential role in IBS etiology. In this review, we summarize current evidence regarding the impact of diet and the intestinal microbiota on IBS symptoms, as well as the reported interactions between diet and the microbiota composition. On the basis of the existing data, we suggest pathways (mechanisms) by which diet components, via the microbial fermentation, could trigger IBS symptoms. Finally, this review provides recommendations for future studies that would enable elucidation of the role of diet and microbiota and how these factors may be (inter)related in the pathophysiology of IBS.
Collapse
Affiliation(s)
- Mirjana Rajilić-Stojanović
- Department of Biochemical Engineering and Biotechnology, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Daisy M Jonkers
- Division Gastroenterology-Hepatology, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Anne Salonen
- Immunobiology Research Programme, Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Kurt Hanevik
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jeroen Raes
- Department Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Jonna Jalanka
- Department of Veterinary Biosciences, Microbiology, University of Helsinki, Helsinki, Finland
| | - Willem M de Vos
- Immunobiology Research Programme, Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Microbiology, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Chaysavanh Manichanh
- Digestive System Research Unit, University Hospital Vall d'Hebron, Ciberehd, Barcelona, Spain
| | - Natasa Golic
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Paul Enck
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tuebingen, Tìbingen, Germany
| | - Elena Philippou
- Department of Life and Health Sciences, School of Sciences and Engineering, University of Nicosia, Nicosia, Cyprus
| | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gerard Clarke
- Department of Psychiatry and Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Robin C Spiller
- Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Queens Medical Centre, Nottingham, UK
| | - John Penders
- Department of Medical Microbiology, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
234
|
Joo YE. Alteration of fecal microbiota in patients with postinfectious irritable bowel syndrome. J Neurogastroenterol Motil 2015; 21:135-7. [PMID: 25611066 PMCID: PMC4288086 DOI: 10.5056/jnm14133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/28/2014] [Accepted: 12/28/2014] [Indexed: 12/16/2022] Open
Affiliation(s)
- Young Eun Joo
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
235
|
Aguilera M, Cerdà-Cuéllar M, Martínez V. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes 2015; 6:10-23. [PMID: 25531553 PMCID: PMC4615720 DOI: 10.4161/19490976.2014.990790] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Alterations in the composition of the commensal microbiota (dysbiosis) seem to be a pathogenic component of functional gastrointestinal disorders, mainly irritable bowel syndrome (IBS), and might participate in the secretomotor and sensory alterations observed in these patients.We determined if a state antibiotics-induced intestinal dysbiosis is able to modify colonic pain-related and motor responses and characterized the neuro-immune mechanisms implicated in mice. A 2-week antibiotics treatment induced a colonic dysbiosis (increments in Bacteroides spp, Clostridium coccoides and Lactobacillus spp and reduction in Bifidobacterium spp). Bacterial adherence was not affected. Dysbiosis was associated with increased levels of secretory-IgA, up-regulation of the antimicrobial lectin RegIIIγ, and toll-like receptors (TLR) 4 and 7 and down-regulation of the antimicrobial-peptide Resistin-Like Molecule-β and TLR5. Dysbiotic mice showed less goblet cells, without changes in the thickness of the mucus layer. Neither macroscopical nor microscopical signs of inflammation were observed. In dysbiotic mice, expression of the cannabinoid receptor 2 was up-regulated, while the cannabinoid 1 and the mu-opioid receptors were down-regulated. In antibiotic-treated mice, visceral pain-related responses elicited by intraperitoneal acetic acid or intracolonic capsaicin were significantly attenuated. Colonic contractility was enhanced during dysbiosis. Intestinal dysbiosis induce changes in the innate intestinal immune system and modulate the expression of pain-related sensory systems, an effect associated with a reduction in visceral pain-related responses. Commensal microbiota modulates gut neuro-immune sensory systems, leading to functional changes, at least as it relates to viscerosensitivity. Similar mechanisms might explain the beneficial effects of antibiotics or certain probiotics in the treatment of IBS.
Collapse
Key Words
- AMP, antimicrobial peptide
- CB1/2, cannabinoid receptor type 1 or 2
- FGD, functional gastrointestinal disorder
- FISH, fluorescent in situ hybridization
- GCM, gut commensal microbiota
- GI, gastrointestinal
- IBS, irritable bowel syndrome
- MOR, mu-opioid receptor
- NGF, nerve growth factor
- PPR, pattern recognition receptor
- RELMβ, resistin-like molecule-β
- RT-qPCR, reverse transcription quantitative polymerase chain reaction
- Reg3γ, regenerating islet-derived protein 3 gamma
- SFB, segmented filamentous bacteria
- TLR, toll-like receptor
- TPH 1/2, tryptophan hydroxylase isoforms 1 or 2
- TRPV1/3, transient receptor potential vanilloid types 1 or 3
- cannabinoid receptors
- colonic motility
- gut commensal microbiota
- iNOS, inducible nitric oxide synthase
- innate immune system
- intestinal dysbiosis
- opioid receptors
- sIgA, secretory IgA
- visceral sensitivity
Collapse
Affiliation(s)
- M Aguilera
- Department of Cell Biology; Physiology and Immunology; Universitat Autònoma de Barcelona; Barcelona, Spain,Neuroscience Institute; Universitat Autònoma de Barcelona; Barcelona, Spain
| | - M Cerdà-Cuéllar
- Centre de Recerca en Sanitat Animal (CReSA); UAB-IRTA; Universitat Autònoma de Barcelona, and Institut de Recerca i Tecnologia Agroalimentàries (IRTA); Barcelona, Spain
| | - V Martínez
- Department of Cell Biology; Physiology and Immunology; Universitat Autònoma de Barcelona; Barcelona, Spain,Neuroscience Institute; Universitat Autònoma de Barcelona; Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd); Instituto de Salud Carlos III; Madrid, Spain,Correspondence to: Vicente Martínez;
| |
Collapse
|
236
|
Abstract
The aetiology and pathology of IBS, a functional bowel disorder thought to lack an organic cause, is largely unknown. However, studies suggest that various features, such as altered composition of the gut microbiota, together with increased intestinal permeability, a changed balance in the enteroendocrine system and a dysregulated immune system in the gut, most likely have an important role in IBS. Exactly how these entities act together and give rise to symptoms is still unknown, but an altered gut microbiota composition could lead to dysregulation of the intestinal barrier as well as the enteroendocrine and the immune systems, which (through interactions with the nervous system) might generate symptoms. This Review highlights the crosstalk between the gut microbiota, the enteroendocrine system, the immune system and the role of intestinal permeability in patients with IBS.
Collapse
|
237
|
Dietary requirement for serum-derived bovine immunoglobulins in the clinical management of patients with enteropathy. Dig Dis Sci 2015; 60:13-23. [PMID: 25142170 PMCID: PMC4284400 DOI: 10.1007/s10620-014-3322-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 08/04/2014] [Indexed: 12/14/2022]
Abstract
A variety of human disease conditions are associated with chronic intestinal disorders or enteropathies that are characterized by intestinal inflammation, increased gut permeability, and reduced capacity to absorb nutrients. Such disruptions in the homeostasis of the gastrointestinal (GI) tract can lead to symptoms of abdominal pain and discomfort, bloating, abnormal bowel function, and malabsorption of nutrients. While significant advances have been made in understanding the factors that influence the complex and fragile balance between the gut microbiota, intestinal epithelial cell integrity, and the underlying immune system, effective therapies for restoring intestinal balance during enteropathy are still not available. Numerous studies have demonstrated the ability of oral immunoglobulins to improve weight gain, support gut barrier function, and reduce the severity of enteropathy in animals. More recently, studies in humans provide evidence that serum-derived bovine immunoglobulin/protein isolate is safe and improves nutritional status and GI symptoms in patients with enteropathy associated with irritable bowel syndrome or infection with the human immunodeficiency virus. This review summarizes studies showing the impact of enteropathy on nutritional status and how specially formulated bovine immunoglobulins may help restore intestinal homeostasis and nutritional status in patients with specific enteropathies. Such protein preparations may provide distinct nutritional support required for the dietary management of patients who, because of therapeutic or chronic medical needs, have limited or impaired capacity to digest, absorb, or metabolize ordinary foodstuffs or certain nutrients, or other special medically determined nutrient requirements that cannot be satisfied by changes to the normal diet alone.
Collapse
|
238
|
Abstract
Background Acute rejection (AR) remains a life-threatening complication after orthotopic liver transplantation (OLT) and there are few available diagnostic biomarkers clinically for AR. This study aims to identify intestinal microbial profile and explore potential application of microbial profile as a biomarker for AR after OLT. Methods The OLT models in rats were established. Hepatic graft histology, ultrastructure, function, and intestinal barrier function were tested. Ileocecal contents were collected for intestinal microbial analysis. Results Hepatic graft suffered from the ischemia-reperfusion (I/R) injury on day 1, initial AR on day 3, and severe AR on day 7 after OLT. Real-time quantitative polymerase chain reaction results showed that genus Faecalibacterium prausnitzii and Lactobacillus were decreased, whereas Clostridium bolteae was increased during AR. Notably, cluster analysis of denaturing gradient gel electrophoresis (DGGE) profiles showed the 7AR and 3AR groups clustered together with 73.4% similarity, suggesting that intestinal microbiota was more sensitive than hepatic function in responding to AR. Microbial diversity and species richness were decreased during AR. Phylogenetic tree analysis showed that most of the decreased key bacteria belonged to phylum Firmicutes, whereas increased key bacteria belonged to phylum Bacteroidetes. Moreover, intestinal microvilli loss and tight junction damage were noted, and intestinal barrier dysfunction during AR presented a decrease of fecal secretory immunoglobulin A (sIgA) and increase of blood bacteremia, endotoxin, and tumor necrosis factor-α. Conclusion We dynamically detail intestinal microbial characterization and find a high sensitivity of microbial change during AR after OLT, suggesting that intestinal microbial variation may predict AR in early phase and become an assistant therapeutic target to improve rejection after OLT. Supplemental digital content is available in the text.
Collapse
|
239
|
The future of probiotics for disorders of the brain-gut axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:417-32. [PMID: 24997045 DOI: 10.1007/978-1-4939-0897-4_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Probiotics, or at the very least products that might have probiotic properties, have been with us for decades, if not centuries, but it has only been in recent years that they have been subjected to serious scientific study. This surge in interest in probiotics has coincided with the era of the microbiome; as more and more is understood about the gut microbiota in health and disease, the therapeutic option of modulating the microbiota through the administration of probiotics has gained a more secure foundation. Regrettably, while a vast literature attests to the beneficial impact of probiotics in a variety of animal models and the mechanisms underlying such positive effects have been dissected in great detail, the data base on probiotics in man remains pretty slender.To make progress, a number of basic issues need to be addressed: strain characterization and other aspects of quality control need to be rigorously applied and additional steps such as dose optimization, definition of desired site of effect and tailoring of formulation accordingly accomplished before large scale trials, based on appropriately selected study endpoints and employing a clinically meaningful study duration, are embarked upon. Meantime, it is to be hoped that the regulatory climate will have been clarified and appropriate guidelines for the evaluation of probiotics, whether as food or drug, developed. Ultimately, the current terminology may have to be abandoned as evidence for biological and clinical activity for dead bacteria, bacterial components and bacterial products accumulates.
Collapse
|
240
|
Kennedy PJ, Cryan JF, Dinan TG, Clarke G. Irritable bowel syndrome: A microbiome-gut-brain axis disorder? World J Gastroenterol 2014; 20:14105-14125. [PMID: 25339800 PMCID: PMC4202342 DOI: 10.3748/wjg.v20.i39.14105] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/18/2014] [Accepted: 05/26/2014] [Indexed: 02/07/2023] Open
Abstract
Irritable bowel syndrome (IBS) is an extremely prevalent but poorly understood gastrointestinal disorder. Consequently, there are no clear diagnostic markers to help diagnose the disorder and treatment options are limited to management of the symptoms. The concept of a dysregulated gut-brain axis has been adopted as a suitable model for the disorder. The gut microbiome may play an important role in the onset and exacerbation of symptoms in the disorder and has been extensively studied in this context. Although a causal role cannot yet be inferred from the clinical studies which have attempted to characterise the gut microbiota in IBS, they do confirm alterations in both community stability and diversity. Moreover, it has been reliably demonstrated that manipulation of the microbiota can influence the key symptoms, including abdominal pain and bowel habit, and other prominent features of IBS. A variety of strategies have been taken to study these interactions, including probiotics, antibiotics, faecal transplantations and the use of germ-free animals. There are clear mechanisms through which the microbiota can produce these effects, both humoral and neural. Taken together, these findings firmly establish the microbiota as a critical node in the gut-brain axis and one which is amenable to therapeutic interventions.
Collapse
|
241
|
Rajilić-Stojanović M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev 2014; 38:996-1047. [PMID: 24861948 PMCID: PMC4262072 DOI: 10.1111/1574-6976.12075] [Citation(s) in RCA: 757] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/29/2014] [Accepted: 05/09/2014] [Indexed: 02/06/2023] Open
Abstract
The microorganisms that inhabit the human gastrointestinal tract comprise a complex ecosystem with functions that significantly contribute to our systemic metabolism and have an impact on health and disease. In line with its importance, the human gastrointestinal microbiota has been extensively studied. Despite the fact that a significant part of the intestinal microorganisms has not yet been cultured, presently over 1000 different microbial species that can reside in the human gastrointestinal tract have been identified. This review provides a systematic overview and detailed references of the total of 1057 intestinal species of Eukarya (92), Archaea (8) and Bacteria (957), based on the phylogenetic framework of their small subunit ribosomal RNA gene sequences. Moreover, it unifies knowledge about the prevalence, abundance, stability, physiology, genetics and the association with human health of these gastrointestinal microorganisms, which is currently scattered over a vast amount of literature published in the last 150 years. This detailed physiological and genetic information is expected to be instrumental in advancing our knowledge of the gastrointestinal microbiota. Moreover, it opens avenues for future comparative and functional metagenomic and other high-throughput approaches that need a systematic and physiological basis to have an impact.
Collapse
Affiliation(s)
- Mirjana Rajilić-Stojanović
- Department for Biotechnology and Biochemical Engineering, Faculty of Technology and Metallurgy, University of BelgradeBelgrade, Serbia
- Laboratory of Microbiology, Wageningen UniversityWageningen, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen UniversityWageningen, The Netherlands
- Departments of Bacteriology and Immunology, and Veterinary Biosciences, University of HelsinkiHelsinki, Finland
| |
Collapse
|
242
|
Affiliation(s)
- John K Marshall
- Division of Gastroenterology, Department of Medicine, McMaster University, Hamilton Ontario, Canada.
| |
Collapse
|
243
|
Dupont HL. Review article: evidence for the role of gut microbiota in irritable bowel syndrome and its potential influence on therapeutic targets. Aliment Pharmacol Ther 2014; 39:1033-42. [PMID: 24665829 DOI: 10.1111/apt.12728] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/18/2013] [Accepted: 03/06/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a prevalent gastrointestinal disease with a substantial social and economic burden. Treatment options remain limited and research on the aetiology and pathophysiology of this multifactorial disease is ongoing. AIM To discuss the potential role of gut microbiota in the pathophysiology of IBS and to identify possible interactions with pathophysiologic targets in IBS. METHODS Articles were identified via a PubMed database search ['irritable bowel syndrome' AND (anti-bacterial OR antibiotic OR flora OR microbiota OR microflora OR probiotic)]. English-language articles were screened for relevance. Full review of publications for the relevant studies was conducted, including additional publications that were identified from individual article reference lists. RESULTS The role of gut microbiota in IBS is supported by varying lines of evidence from animal and human studies. For example, post-infectious IBS in humans is well documented. In addition, certain probiotics and nonsystemic antibiotics appear to be efficacious in the treatment of IBS. Mechanisms involved in improving IBS symptoms likely go beyond mere changes in the composition of the gut microbiota, and accumulating animal data support the interplay of microbiota with other IBS targets, such as the gut-brain axis, visceral hypersensitivity, mucosal inflammation and motility. CONCLUSION The role of the gut microbiota is still being elucidated; however, it appears to be one of several important factors that contributes to the aetiology and pathophysiology of the irritable bowel syndrome.
Collapse
Affiliation(s)
- H L Dupont
- St Luke's Medical Center, and Baylor College of Medicine, The University of Texas School of Public Health, Houston, TX, USA
| |
Collapse
|
244
|
Theodorou V, Belgnaoui AA, Agostini S, Eutamene H. Effect of commensals and probiotics on visceral sensitivity and pain in irritable bowel syndrome. Gut Microbes 2014; 5:430-6. [PMID: 25184834 PMCID: PMC4153783 DOI: 10.4161/gmic.29796] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The last ten years' wide progress in the gut microbiota phylogenetic and functional characterization has been made evidencing dysbiosis in several gastrointestinal diseases including inflammatory bowel diseases and irritable bowel syndrome (IBS). IBS is a functional gut disease with high prevalence and negative impact on patient's quality of life characterized mainly by visceral pain and/or discomfort, representing a good paradigm of chronic gut hypersensitivity. The IBS features are strongly regulated by bidirectional gut-brain interactions and there is increasing evidence for the involvement of gut bacteria and/or their metabolites in these features, including visceral pain. Further, gut microbiota modulation by antibiotics or probiotics has been promising in IBS. Mechanistic data provided mainly by animal studies highlight that commensals or probiotics may exert a direct action through bacterial metabolites on sensitive nerve endings in the gut mucosa, or indirect pathways targeting the intestinal epithelial barrier, the mucosal and/or systemic immune activation, and subsequent neuronal sensitization and/or activation.
Collapse
Affiliation(s)
- Vassilia Theodorou
- INRA; UMR 1331 TOXALIM; Neuro-Gastroenterology and Nutrition Group; Toulouse, France,El-Purpan; UMR 1331 TOXALIM; Neuro-Gastroenterology and Nutrition Group; Toulouse, France,Correspondence to: Vassilia Theodorou,
| | - Afifa Ait Belgnaoui
- INRA; UMR 1331 TOXALIM; Neuro-Gastroenterology and Nutrition Group; Toulouse, France,El-Purpan; UMR 1331 TOXALIM; Neuro-Gastroenterology and Nutrition Group; Toulouse, France,Lallemand Health Solutions Inc; Montreal, Canada
| | - Simona Agostini
- INRA; UMR 1331 TOXALIM; Neuro-Gastroenterology and Nutrition Group; Toulouse, France,El-Purpan; UMR 1331 TOXALIM; Neuro-Gastroenterology and Nutrition Group; Toulouse, France
| | - Helene Eutamene
- INRA; UMR 1331 TOXALIM; Neuro-Gastroenterology and Nutrition Group; Toulouse, France,El-Purpan; UMR 1331 TOXALIM; Neuro-Gastroenterology and Nutrition Group; Toulouse, France
| |
Collapse
|