Join Us | Latest Articles | Contact

Journal Home


Editorial Board


Archive


Submit to this journal


Current issue

Journal of Infectious Diseases and Epidemiology





DOI: 10.23937/2474-3658/1510025



The Intestinal Microbial Community and Inflammatory Bowel Diseases

Hubert E Blum*


Department of Medicine II, University Hospital Freiburg, Germany


*Corresponding author: Prof. Hubert E Blum, Department of Medicine II, University Hospital Freiburg, Hugstetterstrasse 55, D-79106 Freiburg, Germany, Tel: +0049-761-27018116, Fax: +0049-761-27018117, E-mail: hubert.blum@uniklinik-freiburg.de
J Infect Dis Epidemiol, JIDE-3-025, (Volume 3, Issue 1), Review Article; ISSN: 2474-3658
Received: May 07, 2016 | Accepted: March 04, 2017 | Published: March 06, 2017
Citation: Blum HE (2017) The Intestinal Microbial Community and Inflammatory Bowel Diseases. J Infect Dis Epidemiol 3:025. 10.23937/2474-3658/1510025
Copyright: © 2017 Blum HE. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.



Abstract

Based on molecular, genetic, epigenetic, biochemical and microbiological analyses it is increasingly possible to identify individual disease-related characteristics that define disease pathogenesis, disease disposition or prognosis as well as the efficacy of therapeutic strategies ('personalized medicine/precision medicine'). In this context, the global human microbiome project, aimed at deciphering the complete set of genes of the microbiota, i.e., the individual's complete microbial community, was established in 2007 and has meanwhile developed into a major field of biomedical research. In particular, the intestinal microbial community has turned out to play a major role in human health and disease. In this context, the following comment addresses novel insights into its contribution to the pathogenesis, prevention and treatment of inflammatory bowel diseases (IBD). Beyond IBD, the intestinal microbial community is involved in numerous other, common, non-gastrointestinal diseases, such as obesity/metabolic syndrome and atherosclerosis as well as in health, in particular in immunity, making it one of the most dynamic current topics in biomedical research.


Keywords

Genome-wide association studies, Human microbiome project, Microbiota, Personalized medicine


Introduction

The basic aspects of molecular and cell biology are not only integral part of biomedical research but are also increasingly translated into 'personalized/precision medicine' with clinical relevance for the diagnosis, treatment and prevention of human diseases. Apart from the international human genome organization (HUGO) project that identified the complete sequence of the human genome about 15 years ago [1,2] and the international haplotype map (HapMap) project initiated in 2005 to identify, based on genome-wide association studies (GWAS) in ethnically different populations, single nucleotide polymorphisms (SNPs) and their association with specific human diseases and individual phenotypic characteristics, respectively [3,4], a third global consortium, the human microbiome project (HMP), was established in 2007 [5-9]. The HMP and the 'Metagenomics of the Human Intestinal Tract (Meta-HIT) Consortium Europe' aim at the sequencing of all microbes (eukaryotes, archaea, bacteria and viruses) that inhabit specific body sites (Table 1). Recent data demonstrate that specific compositions of the microbial community are associated with health and disease [5-9] and suggest that the detailed characterization, function and variation of the microbial community will reveal important commensal host-microbe as well as microbe-microbe interactions with diagnostic, therapeutic and preventive implications [10,11].



Table 1: Human microbial communities. View Table 1


Intestinal microbial community

In recent years the intestinal microbial community has been studied in great detail. The colonization of the human gut begins at birth, is characterized by a successively changing composition and eventually becomes relatively stable in adulthood [12]. Important factors for the composition of the intestinal microbial community are, among others, diet, lifestyle and exposure to drugs/antibiotics. In this context it appears that the administration of low-dose penicillin early in life has lasting effects on the body mass index (obesity) through alteration of the intestinal microbial community [13,14]. Recent evidence further suggests that human genetic variation also influences the abundance of specific members of the intestinal microbial community [15].

The intestinal microbial community is highly variable from person to person but family members tend to have more similar microbes than unrelated individuals, possibly due to shared environmental factors and genetic similarities. Recent studies indicate that certain intestinal microbial communities are not only associated with different chronic diseases but may play a causative role in disease pathogenesis [5-9]. This is, last but not least, supported by the fact that the transplantation of intestinal microbes from diseased animals/humans to healthy recipients results in phenotypic disease characteristics, e.g., in kwashiorkor [16].

A causal relationship between the intestinal microbial community and an increasing number of human diseases, have been identified to date (Table 2). In addition, intestinal microbes play a central role in drug metabolism, e.g., of sulfasalazine, levodopa and irinotecan, both with respect to efficacy and to undesired side effects.



Table 2: Intestinal microbial community in health and diseases (examples). View Table 2


The inflammatory bowel diseases (IBD) in humans include ulcerative colitis (UC) and Crohn disease (CD).These are characterized by inflammation limited to the mucosal layer of the colon in UC and the transmural involvement of the gastrointestinal tract, including extraintestinal sites in CD. While the pathogenesis of IBD is not fully understood [17], it is clear that its pathology depends among others on the intestinal microbial community [18,19]. Further, a case-control study identified 'IBD-specific' alterations of the intestinal microbiota that may serve as biomarkers for the prediction of disease predisposition, activity/severity and responsiveness to therapy [20,21].

Host genes with effects on the composition of the intestinal microbiota are the IgA locus and the HLA genes as well as the defensin genes, the NOD2 gene, the resistin-like molecule beta gene, the apolipoprotein I gene, the MEFV gene and the myeloid differentiation primary response protein 88 gene. The three components -environment, host genetics and the microbial community- interact to maintain homeostasis in the intestine [6]. The disruption of the stability of this interaction may be a trigger for disease development. Two recent publications shed a new light on the pathogenesis of IBD through the change of the intestinal microbial composition involving two different pathways: helminth infection [22] and lipocalin-2 expression [23], respectively.


Helminth infection, microbial community and IBD

Epidemiologic studies demonstrated a major increase of the incidence of IBD in the developed world, suggesting a change in the environment, including an alteration of the intestinal microbiome [24] and a decreased exposure to intestinal parasites, such as helminths [25]. In mice deficient for the CD susceptibility gene Nod2 (Nod2-/-/knockout) [26], it could be demonstrated that small intestinal abnormalities develop in the face of a sustained colonization with the inflammatory bacterium Bacteroides vulgatus, an ubiquitous member of the intestinal microbial community [27]. Chronic infection of Nod2 mice with the parasitic worm Trichuris muris, however, inhibited colonization with inflammatory Bacteroides species and promoted the establishment of a protective microbial environment enriched in Clostridiales [22]. Further, the authors demonstrated that individuals from helminth-endemic regions harbour a similar protective microbial community and deworming treatment reduced Clostridiales and increased Bacteriodales, resulting in an increased IBD incidence. These data support the hygiene hypothesis whereby certain individuals are genetically susceptible to the consequences of a changing intestinal microbial community that favours IBD development.


Lipocalin-2 protection from IBD and colon cancer

Lipocalin-2 (Lcn2) is an antimicrobial peptide with high mucosal and fecal concentrations in patients with IBD. It is produced by various cell types, including epithelial cells, and acts as an antimicrobial defence mediator by binding to a subset of bacterial siderophores, thereby preventing bacterial iron acquisition and growth of siderophore-dependent strains. While it has been implicated in several biologic processes, such as acute phase response, erythropoiesis and iron metabolism, its functional role in contributing to IBD development remained unclear.

To decipher the role of Lcn2 in colon inflammation, mice double deficient in Lcn2 and IL-10 Lcn2-/-/IL10-/- double knockout) were generated and compared to single knockouts and wild-type animals. The experimental data indicate that Lcn2 expression protects from early onset colitis and the spontaneous emergence of right-sided colonic tumours that result from IL-10 deficiency. The inflammation is driven by IL-6 which also controls tumorigenesis. The Lcn2-/-/IL10-/- double knockout mice showed major alterations of their intestinal microbial community, especially with respect to the facultative pathogenic Alistipes spp. These contribute to inflammation and tumorigenesis as shown by the transmissibility of the phenotype and the protection by antibiotic therapy. Taken together, the authors demonstrate that Lcn2 protects against intestinal inflammation and tumorigenesis in the face of an altered intestinal microbial composition [23].


Conclusions and Perspectives

Recent advances in cell and molecular biology resulted in an increasingly detailed understanding of the pathogenesis of gastrointestinal diseases, including IBD. Apart from an increasing number of host genetic susceptibility loci and environmental factors, the individual microbial community is central for the barrier between microbes and hosts and for the mucosal homeostasis. In this context the mucosal immune responses to luminal dietary and/or microbial antigens play a major role. A thorough understanding of IBD pathogenesis depends on the one hand on the detailed characterization of the host's mucosal defence and response mechanisms and on the other hand on the careful analysis of the complex dynamics of the intestinal microbes.

The two recent publications discussed above [22,23], give a first glimpse into the complex mechanisms underlying the protection from IBD by the intestinal microbial community, i.e., by its modification through chronic helminth infection in susceptible hosts or by changing the microbial composition that results in a reduction of bacterial species with inflammatory and tumorigenic potential thereby increasing Lcn2 expression. These findings suggest a highly intricate interplay between the host and the microbial community in IBD pathogenesis and may eventually contribute to IBD prevention and therapy in humans.

Beyond IBD, the intestinal microbial community is involved in numerous other, common, non-gastrointestinal diseases, such as obesity/metabolic syndrome and atherosclerosis as well as in health, in particular in immunity, making it to one of the most dynamic areas of biomedical research.


References
  1. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, et al. (2001) Initial sequencing and analysis of the human genome. Nature 409: 860-921.

  2. Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, et al. (2001) The sequence of the human genome. Science 291: 1304-1351.

  3. Manolio TA, Brooks LD, Collins FS (2008) A HapMap harvest of insights into the genetics of common disease. J Clin Invest 118: 1590-1605.

  4. Manolio TA, Collins FS (2009) The HapMap and genome-wide association studies in diagnosis and therapy. Annu Rev Med 60: 443-456.

  5. Proctor LM (2011) The Human Microbiome Project in 2011 and beyond. Cell Host Microbe 10: 287-291.

  6. Spor A, Koren O, Ley R (2011) Unravelling the effects of the environment and host genotype on the gut microbiome. Nat Rev Microbiol 9: 279-290.

  7. Human Microbiome Project Consortium (2012) Structure, function and diversity of the healthy human microbiome. Nature 486: 207-214.

  8. Human Microbiome Project Consortium (2012) A framework for human microbiome research. Nature 486: 215-221.

  9. Gevers D, Knight R, Petrosino JF, Huang K, McGuire AL, et al. (2012) The Human Microbiome Project: a community resource for the healthy human microbiome. PLoS Biol 10: e1001377.

  10. Morgan XC, Segata N, Huttenhower C (2013) Biodiversity and functional genomics in the human microbiome. Trends Genet 29: 51-58.

  11. Rossen NG, Fuentes S, van der Spek MJ, Tijssen JG, Hartman JH, et al. (2015) Findings From a Randomized Controlled Trial of Fecal Transplantation for Patients With Ulcerative Colitis. Gastroenterology 149: 110-118.

  12. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, et al. (2012) Human gut microbiome viewed across age and geography. Nature 486: 222-227.

  13. Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, et al. (2014) Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 158: 705-721.

  14. Jess T (2014) Microbiota, antibiotics, and obesity. N Engl J Med 371: 2526-2528.

  15. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, et al. (2014) Human genetics shape the gut microbiome. Cell 159: 789-799.

  16. Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, et al. (2013) Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science 339: 548-554.

  17. Xavier RJ, Podolsky DK (2007) Unravelling the pathogenesis of inflammatory bowel disease. Nature 448: 427-434.

  18. Hall LJ, Walshaw J, Watson AJ (2014) Gut microbiome in new-onset Crohn's disease. Gastroenterology 147: 932-934.

  19. Sha S, Xu B, Wang X, Zhang Y, Wang H, et al. (2013) The biodiversity and composition of the dominant fecal microbiota in patients with inflammatory bowel disease. Diagn Microbial Infect Dis 75: 245-251.

  20. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, et al. (2007) Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci USA 104: 13780-13785.

  21. Peterson DA, Frank DN, Pace NR, Gordon JI (2008) Metagenomic approaches for defining the pathogenesis of inflammatory bowel diseases. Cell Host Microbe 3: 417-427.

  22. Ramanan D, Bowcutt R, Lee SC, Tang MS, Kurtz ZD, et al. (2016) Helminth infection promotes colonization resistance via type 2 immunity. Science 352: 608-612.

  23. Moschen AR, Gerner RR, Wang J, Klepsch V, Adolph TE, et al. (2016) Lipocalin 2 protects from inflammation and tumorigenesis associated with gut microbiota alterations. Cell Host Microbe 19: 455-469.

  24. Belkaid Y, Hand TW (2014) Role of the microbiota in immunity and inflammation. Cell 157: 121-141.

  25. Weinstock JV, Elliott DE (2009) Helminths and the IBD hygiene hypothesis. Inflamm Bowel Dis 15: 128-133.

  26. Cleynen I, Boucher G, Jostins L, Schumm LP, Zeissig S, et al. (2016) Inherited determinants of Crohn's disease and ulcerative colitis phenotypes: a genetic association study. Lancet 387: 156-167.

  27. Ramanan D, Tang MS, Bowcutt R, Loke P, Cadwell K (2014) Bacterial sensor Nod2 prevents inflammation of the small intestine by restricting the expansion of the commensal Bacteroides vulgatus. Immunity 41: 311-324.

  28. Honda K, Littman DR (2016) The microbiota in adaptive immune homeostasis and disease. Nature 535: 75-84.

  29. Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, et al. (2016) Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk Cell 165: 111-124.

  30. Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, et al. (2015) Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 163: 1585-1595.

  31. Jonsson AL, Backhed F (2015) Drug the Bug! Cell 163: 1565-1566.

  32. Tilg H (2016) A gut feeling about thrombosis. N Engl J Med 374: 2494-2496.

  33. Vatanen T, Kostic AD, d'Hennezel E, Siljander H, Franzosa EA, et al. (2016) Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 165: 842-853.

  34. Feng Q, Liang S, Jia H, Stadlmayr A, Tang L, et al. (2015) Gut microbiome development along the colorectal adenoma-carcinoma sequence. Nat commun 6: 6528.

  35. Forbes JD, Van Domselaar G, Bernstein CN (2016) Microbiome survey of the inflamed and noninflamed gut at different compartments within the gastrointestinal tract of inflammatory bowel disease patients. Inflamm Bowel Dis 22: 817-825.

  36. Forbes JD, Van Domselaar G, Bernstein CN (2016) The Gut Microbiota in Immune-Mediated Inflammatory Diseases. Front microbial 7: 1081.

  37. Cantarel BL, Waubant E, Chehoud C, Kuczynski J, DeSantis TZ, et al. (2015) The gut microbiota in immune-mediated inflammatory diseases. J Investing Med 63: 729-734.

  38. Chen J, Wright K, Davis JM, Jeraldo P, Marietta EV, et al. (2016) An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med 8: 43.

  39. Scher JU, Ubeda C, Artacho A, Attur M, Isaac S, et al. (2015) Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis Rheumatol 67: 128-139.

  40. Thaiss CA, Zmora N, Levy M, Elinav E (2016) The microbiome and innate immunity. Nature 535: 65-74.

  41. Haque TR, Barritt AS 4th(2016) Intestinal microbiota in liver disease. Best Pract Res Clin Gastroenterol 30: 133-142.

  42. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, et al. (2009) A core gut microbiome in obese and lean twins. Nature 457: 480-484.

  43. Shen J, Obin MS and Zhao L (2013) The gut microbiota, obesity and insulin resistance. Mol Aspects Medicine 34: 39-58.

  44. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, et al. (2013) Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 341: 1241214.

  45. Mazidi M, Rezaie P, Kengne AP, Mobarhan MG, Ferns GA (2016) Gut microbiome and metabolic syndrome. Diabetes Metab Syndr 10: S150-S157.

  46. Sonnenburg JL, Backhed F (2016) Diet-microbiota interactions as moderators of human metabolism. Nature 535: 56-64.

  47. Fung TC, Olson CA, Hsiao EY (2017) Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci 20: 145-155.

  48. Choi GB, Yim YS, Wong H, Kim S, Kim H, et al. (2016) The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science 351: 933-939.

  49. Aizawa E, Tsuji H, Asahara T, Takahashi T, Teraishi T, et al. (2016) Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. J Affect Disord 202: 254-257.

  50. Bu XL, Yao XQ, Jiao SS, Zeng F, Liu YH, et al. (2015) A study on the association between infectious burden and Alzheimer's disease. Eur J Neurol 22: 1519-1525.

  51. Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, et al. (2015) Gut microbiota are related to Parkinson's disease and clinical phenotype. Mov Disord 30: 350-358.

International Journal of Anesthetics and Anesthesiology (ISSN: 2377-4630)
International Journal of Blood Research and Disorders   (ISSN: 2469-5696)
International Journal of Brain Disorders and Treatment (ISSN: 2469-5866)
International Journal of Cancer and Clinical Research (ISSN: 2378-3419)
International Journal of Clinical Cardiology (ISSN: 2469-5696)
Journal of Clinical Gastroenterology and Treatment (ISSN: 2469-584X)
Clinical Medical Reviews and Case Reports (ISSN: 2378-3656)
Journal of Dermatology Research and Therapy (ISSN: 2469-5750)
International Journal of Diabetes and Clinical Research (ISSN: 2377-3634)
Journal of Family Medicine and Disease Prevention (ISSN: 2469-5793)
Journal of Genetics and Genome Research (ISSN: 2378-3648)
Journal of Geriatric Medicine and Gerontology (ISSN: 2469-5858)
International Journal of Immunology and Immunotherapy (ISSN: 2378-3672)
International Journal of Medical Nano Research (ISSN: 2378-3664)
International Journal of Neurology and Neurotherapy (ISSN: 2378-3001)
International Archives of Nursing and Health Care (ISSN: 2469-5823)
International Journal of Ophthalmology and Clinical Research (ISSN: 2378-346X)
International Journal of Oral and Dental Health (ISSN: 2469-5734)
International Journal of Pathology and Clinical Research (ISSN: 2469-5807)
International Journal of Pediatric Research (ISSN: 2469-5769)
International Journal of Respiratory and Pulmonary Medicine (ISSN: 2378-3516)
Journal of Rheumatic Diseases and Treatment (ISSN: 2469-5726)
International Journal of Sports and Exercise Medicine (ISSN: 2469-5718)
International Journal of Stem Cell Research & Therapy (ISSN: 2469-570X)
International Journal of Surgery Research and Practice (ISSN: 2378-3397)
Trauma Cases and Reviews (ISSN: 2469-5777)
International Archives of Urology and Complications (ISSN: 2469-5742)
International Journal of Virology and AIDS (ISSN: 2469-567X)
More Journals

Contact Us

ClinMed International Library | Science Resource Online LLC
3511 Silverside Road, Suite 105, Wilmington, DE 19810, USA
Email: contact@clinmedlib.org
 

Feedback

Get Email alerts
 
Creative Commons License
Open Access
by ClinMed International Library is licensed under a Creative Commons Attribution 4.0 International License based on a work at https://clinmedjournals.org/.
Copyright © 2017 ClinMed International Library. All Rights Reserved.