Gut Microbiome and Its Role in the Pathophysiology of Irritable Bowel Syndrome

Giada De Palma ID· Premysl Bercik ID

Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada.

Acta Gastroenterol Latinoam 2021;51(4):378-384
Received:
29/09/2021 / Accepted: 26/10/2021 / Published in www.actagastro.org 13/12/2021 / https://doi.org/10.52787/DXFC9250

boton-pdf

Summary

Irritable bowel syndrome is the most common functional gastrointestinal disorder, affecting up to 9% individuals globally. Although the etiology of this syndrome is likely heterogenous, it presents with its hallmark symptoms of abdominal pain and altered intestinal motility. Moreover, it is considered to be a disorder of the gut-brain interaction, and the microbiome has often been implicated as a central player in its pathophysiology. Patients with irritable bowel syndrome display altered composition and function of the gut microbiota compared to healthy controls. Microbiome directed therapies, such as probiotics, antibiotics and fecal microbiome transplantation, appear to be beneficial for both gut symptoms and psychiatric comorbidities. This review aims to recapitulate the available literature on the microbiome contribution to the pathophysiology and symptoms presentation of irritable bowel syndrome, as well as the current literature on microbiome-targeted treatments for this disease.

Keywords. IBS, diet, microbiome, FMT.

El papel de la microbiota intestinal en la fisiopatología del síndrome de intestino irritable

Resumen

El síndrome de intestino irritable es el trastorno digestivo funcional más diagnosticado, el cual afecta hasta el 9% de la población mundial. Aunque la etiología y las manifestaciones clínicas de esta enfermedad son muy variables, se caracteriza por la presencia de dolor abdominal y alteraciones en la motilidad intestinal. Se considera un desorden del eje intestino-cerebro y se ha planteado que el microbioma intestinal juega un papel central en su fisiopatología. De hecho, los pacientes diagnosticados con síndrome de intestino irritable presentan alteraciones en la composición y función de la microbiota intestinal en comparación con controles sanos. En línea con esta hipótesis, varios estudios confirman que pacientes con este trastorno pueden beneficiarse, tanto a nivel gastrointestinal como psicológico, de intervenciones dietéticas y del uso de terapias dirigidas al microbioma como son el uso de probióticos, antibióticos y, más recientemente, del trasplante fecal. El objetivo de este artículo es llevar a cabo una revisión bibliográfica de la evidencia científica que apoya el papel de la microbiota en la fisiopatología y sintomatología de síndrome del intestino irritable, así como el uso de enfoques terapéuticos dietéticos o microbianos para el tratamiento de pacientes con esta enfermedad.

Palabras claves. SII, dieta, microbioma, transplante de materia fecal.

Abbreviations

IBS: Irritable bowel syndrome.
GI: Gastrointestinal.
IBS-C: Irritable bowel syndrome with constipation.
IBS-D: Irritable bowel syndrome with diarrhea.
ENS: Enteric nervous system.
5-HT: 5-hydroxytryptamine.
AHR: Aryl-hydrocarbon.
PI-IBS: Post-infectious IBS.
SIBO: Small intestinal bacterial overgrowth.
FMT: Fecal microbiome transplant.
FODMAPs: Fermentable oligo-, di- and monosaccharides and polyols.
SCFAs: Short chain fatty acids.
RCT: Randomized controlled trial.

Introduction

Irritable bowel syndrome (IBS) is the most common functional bowel disorder worldwide affecting between 5% and 10% individuals globally.1, 2 Its prevalence varies across the world according to the diagnostic definition used (Rome III vs. Rome IV), the population selected, and local factors.2 As there is no diagnostic biomarker for IBS, its diagnosis is based on symptom reporting. Rome IV is the latest iteration of the Rome Diagnostic criteria and with its rather strict approach, the global prevalence of IBS was found to be lower (4.1% pooled prevalence) than the one previously reported using the Rome III criteria (10.1% pooled prevalence).3 IBS is one of the most common reasons of healthcare seeking with significant socioeconomic impact.1

Despite being the most studied functional gastrointestinal (GI) disorder, its pathophysiology is incompletely understood, in part due to many factors involved in its genesis. It is now well accepted that IBS is a disorder of the gut-brain communication, presenting with visceral hypersensitivity, intestinal dysmotility, impaired central processing of stimuli arising from the GI tract, altered gut microbiota, as well as frequent psychiatric comorbidities, such as anxiety and depression. In addition, multiple dietary triggers are commonly reported by IBS patients.

IBS can be classified into 4 different categories according to bowel habits and stool form: IBS with constipation (IBS-C), IBS with diarrhea (IBS-D), Mixed IBS, and Unsubtyped IBS.4 IBS affects patients across the lifespan, but there is an overall strong female predominance, modulated by age and hormonal status.5, 6 Sex hormones also influence IBS symptom severity and subtype, with constipation being predominant in females, and diarrhea in males.6, 7 While all these factors play a role in IBS pathophysiology, this review will focus on the role of gut microbiota and the possible use of microbiota-directed therapies for symptoms’ mitigation.

Gut Microbiota and Healthy Gut

Mammals shelter in their body an incredibly complex and diverse community of microorganisms, collectively called microbiota (or microbiome if we refer to all the microorganisms and their genetic content), which comprises archaea, bacteria, viruses, fungi, and eukaryotes.8, 9 The gut microbiota evolves during early life until a unique, subject-specific (fingerprint) adult-like community arises, which is highly resilient and relatively stable throughout life,10-12 being dominated by few phyla only, mainly Firmicutes and Bacteroidetes, together with members of Actinobacteria, Verrucomicrobia, Proteobacteria, Fusobacteria, and Cyanobacteria.13 Diet, regional variability and ethnicity greatly impact gut microbiota composition.14, 15

Gut microbiota is thought to be functionally redundant, meaning that different bacterial consortia perform similar functions in different individuals.16 Indeed, gut microbiota carries out essential functions that the human body is unable to perform,17, 18 while occupying a unique, nutrient rich niche. The central role of the microbiome is highlighted by studies in germ-free or microbiome depleted animals, which demonstrated that gut microbiota is required for normal gut physiology, metabolism, a balanced immune system,19-25 regular development of the enteric nervous system (ENS),26, 27 and a normal perception of inflammatory, mechanical and visceral pain.28-30 In addition, gut microbiota affects GI motility31-34 due to its effects on the ENS, by modulating the expression of toll like receptors, serotonin (5-HT) release and activation of the aryl-hydrocarbon (AHR) and the 5-HT4 receptors.26, 35-37

The Role of Gut Microbiota in IBS

The implication of the gut microbiome in IBS is not a novel concept, as many studies have shown that infectious gastroenteritis is the most common trigger of IBS in previously healthy individuals.38-41 Post-infectious IBS (PI-IBS) can develop immediately after a bacterial, viral or protozoal infection, or chronic GI symptoms can worsen after an infectious gastroenteritis (for a detailed review see Berumen et al., 2021).42 The underlying mechanisms are still to be fully elucidated, although several studies have shown evidences of low-grade inflammation or immune activation in IBS patients.43 It has been proposed that transient inflammation leads to subtle but permanent changes in the structure and function of the gut, including increased infiltration with lymphocytes and mast cells, altered enteric nerves and enterochromaffin cells, that, in turn, induce GI symptoms.44

The structure and function of the gut microbiota is deeply perturbed at the site of the infection45 and it might act synergistically with ongoing inflammation and impaired epithelial permeability, increasing the risk of IBS development in susceptible individuals.40, 42 Another condition that has been often associated with IBS is the small intestinal bacterial overgrowth (SIBO), which may be responsible for symptom generation in some patients with IBS. SIBO is defined as a quantitative alteration of the small intestinal microbiota (reviewed in Bushyhead & Quigley, 2021).46 When the mechanisms in place to control and limit bacterial overgrowth in the small intestine (IgA secretion, gastric acid, bile acid and pancreatic secretions, as well as motor patterns) fail, pathological colonization occurs.46 Bacterial overgrowth results in unusual fermentation with increases in gas production, abdominal bloating, malabsorption, abdominal pain, diarrhea, and abnormal GI motility.46-49 There appears to be a link between SIBO and IBS;50 however, its role in IBS is controversial, in part due to the scientific community not reaching a consensus on the detection method to use. While the breath tests are not well validated for SIBO, the jejunal aspirates are not always accurate.50-54 Thus, it remains unclear whether SIBO is actually fundamental to the pathophysiology of IBS or is just a complicating phenomenon. However, several studies suggested that treatment of SIBO with non-absorbable antibiotics, rifaximin being the most commonly used, improves gut symptoms in a proportion of patients with IBS.46, 55-58

Alterations in the gut microbiota composition of IBS patients have been increasingly reported during the last decade, and while multiple studies have shown differences in the microbiota composition between IBS patients and healthy controls (recently reviewed by Pittayanon et al., 2019, and Duan et al., 2019),59,60 the results of these studies have been inconsistent and no unique IBS bacterial signature or profile has been identified. This could be partially due to the use of different detection methods, as well as to different patient populations. In general, there appear to be an increase in potentially pathogenic bacteria, often facultative anaerobes or aerobes, with a decrease in strict anaerobic bacteria, and a decrease in bacterial diversity in IBS patients compared to healthy controls.59, 61 It should be noted, however, that there is not a clear consensus on what constitutes a healthy microbiota.62, 63

A recent study reported that a great proportion of IBS patients (57% in this study) present with visible colonic biofilms, harbouring a less diverse microbiome with an overgrowth of Escherichia coli and Ruminococcus gnavus spp.64 These biofilms correlated with an altered gut microbiome composition and with bile acids malabsorption.64 While most IBS studies focused on the bacterial compartment of the gut microbiota, few studies that have researched its other components found differences in the mycobiome65, 66 and the virome,67 that were associated to those of the bacteriome.66, 67

However, it is now accepted that changes in microbial metabolic activity may have more impact on the host than the changes in microbial profiles. A recent study highlighted not only the importance of longitudinal sampling for IBS, given the fluctuating nature of IBS symptoms, but also the value in integrating different type of data, such as multiple-omics (metagenomics and metabolomics from host and microbes), as well as metadata on symptoms and gut physiology.68, 69 Indeed, these studies found that IBS symptom severity fluctuates in parallel with functional variations in the gut microbiota, as well as with altered bile acid and purine metabolism.69 Nevertheless, a unique and shared metabolomic dysfunction has yet to be discovered for IBS patients. Furthermore, it remains unclear whether the altered microbiome observed in clinical studies is a cause, or a consequence, of the gut dysfunction. Animal studies, employing gnotobiotic models in which germ-free mice are colonized with microbiota from patients with IBS or healthy controls, have proven the causal role of gut microbes in IBS pathophysiology, including altered motility, permeability, visceral hypersensitivity, immune activation, and psychiatric comorbidities.70-73 Nonetheless, further mechanistic studies are needed to advance the field into personalized medicine and microbiota-targeted therapies.

Microbiota-Directed Therapies of IBS

The growing body of evidence suggesting the key role of bacteria in IBS has led to the design of many interventional studies targeting the gut microbiota of IBS patients. Unfortunately, results of these studies have been rather inconsistent. Microbiome targeted approaches include the use of dietary interventions, probiotics and prebiotics (already discussed in depth by Valdovinos-Díaz M.A. in the previous issue of this journal),74 antibiotics, and, more recently, fecal microbiome transplantation (FMT). Dietary interventions are often a preferred, as non invasive, first line of treatement for IBS symptoms. Chronic diet is a major microbiome modulator, as our microbes eat what we eat. Dietary triggers, such as gluten or highly fermentable oligo-, di- and monosaccharides and polyols (FODMAPs), have been frequently reported to worsen symptoms in IBS patients (60%).75,76 Thus, dietary interventions are often proposed by clinicians or self-administered by patients as initial therapuetic approaches to curb IBS symptoms.

A systematic review for the American College of Gastroenterology found insufficient evidence to recommend excluding gluten to reduce IBS symptoms, due to paucity of randomized, placebo controlled trials, and a low quality evidence suggesting that reduction in FODMAPs intake reduces IBS symptoms.76 However, a more recent meta-analysis found that low FODMAP diet is indeed more effective at reducing GI symptoms, such as abdominal pain and abdominal bloating, as well as improving quality of life, than traditional dietary advice or control diets.77 Long-term effects of a low FODMAP diet, however, have been questioned, given that patients could develop nutritional deficits or detrimental loss of beneficial bacteria, such as Bifidobacteria.78 Staudacher and colleagues have recently reported the results of the first long-term personalized low FODMAP study, in which Bifidobacteria levels were unaffected, but a significant decrease in short chain fatty acids (SCFAs) was observed.79 The long-term consequences of this SCFAs impairment are unknown, and, as SCFAs have been implicated in regulation of GI motility and gut epithelial function,80, 81 this observation requires further studies. This study exemplifies very well the great conundrum behind diet-microbiome-directed therapies, as modifying one dietary component may have a temporal beneficial effect while possibly triggering a long-term ripple effect due to microbiome restructuring.

Another microbiome-targeted approach that has been employed for the treatement of IBS symptoms is rifaximin: a minimally absorbed antibiotic normally used for SIBO and as second-line treatement for IBS-D.58, 82 The evidence for its efficacy is, however, only modest.82, 83 Besides rifaximin, two other antibiotics have been tested in IBS patients, neomycin and norfloxacin83-85, with both medications being more effective than placebo at improving IBS symptoms83-85. However, repeated use of antibiotics in IBS is discouraged, and should not to be confused with its use for SIBO, as it could lead to increased microbial antibiotic resistance gene pool in IBS patients.

The last microbiome-targeted approach that has increasingly gained attention is the FMT. Seven randomized controlled trials (RCTs) have been performed up until now, with three of them reporting clear beneficial effects including reduction of IBS symptoms86-88 and improving quality of life.87, 88 Three RCTs found no clear efficacy of FMT in IBS89, 90, 91 and one found only a transient relief of symptoms.92 Two additional studies have investigated the long-term efficacy of FMT observing sustained efficacy93, 94 and safety,94 and changes to the gut microbiome that were more comprehensive that those observed at the end of the original RCT.93 All these studies, however, did not use a standardized method of delivery, with some administering the FMT into the cecum or the distal colon,86, 91, 92 some into the small bowel,87, 88 and others using oral capsules.89, 90 Similarly, these studies differed with the respect to the donors, with some using only one donor,87, 92 some multiple donors86, 88-91 and others pooling all donors together.86, 89 Furthermore, patient preparation was not identical, with some studies using bowel preparation or prescribing loperamide as pre-treatment.86, 88

Despite these differences in the study design and outcomes, some preliminary conclusions may be reached: donor selection appears to be crucial, and pooled microbiota from several donors may have worse outcomes than that of single donor. The amount of material (> 30g) and frequency of administration seems to play a major role, with repeated FMT having better efficacy.94, 95 Finally, donor microbiota engraftment does not appear to be necessary for the successful outcome. Based on gnotobiotic mouse models, the donor selection may be the key factor, as behavioral and physiological abnormalites seen in patients can be transferred into germ-free mice through microbiota transplantation,73 thus highlighting not only the necessity to screen in depth potential donors for physical and mental health, but also the potential for treating psychiatric comorbidities of IBS.96

In conclusion, accumulating data suggest that, in a significant proportion of patients, the microbiota plays an important role in the genesis and maintenance of IBS. The use of personalized dietary approaches, probiotics and other microbiota directed therapies, including FMT, appears to be of therapeutic value, although more clinical data are needed. We should strive to bridge the gap currently existing between preclinical and clinical research69 with further mechanistic translational and reverse translational studies to elucidate the complex interactions behind success and failure of these microbiome-directed therapies.

Intellectual Property. The authors declare that the data that appear in this article are original and were made in their belonging institutions.

Copyright

© 2021 Acta Gastroenterológica latinoamericana. This is an open-​access article released under the terms of the Creative Commons Attribution (CC BY-NC-SA 4.0) license, which allows non-commercial use, distribution, and reproduction, provided the original author and source are acknowledged.

Cite this article as: De Palma G, Bercik P. Gut Microbiome and Its Role in the Pathophysiology of Irritable Bowel Syndrome. Acta Gastroenterol Latinoam. 2021;51(4):378-4 https://doi.org/10.52787/DXFC9250

References

  1. Ford, A. C., Sperber, A. D., Corsetti, M. & Camilleri, M. Irritable bowel syndrome. Lancet 396, 1675-1688 (2020).
  2. Oka, P. et al. Global prevalence of irritable bowel syndrome according to Rome III or IV criteria: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 5, 908-917 (2020).
  3. Sperber, A. D. et al. Worldwide Prevalence and Burden of Functional Gastrointestinal Disorders, Results of Rome Foundation Global Study. Gastroenterology 160, 99-114 e113 (2021).
  4. Longstreth, G. F. et al. Functional bowel disorders. Gastroenterology 130, 1480-1491 (2006).
  5. Camilleri, M. Sex as a biological variable in irritable bowel syndrome. Neurogastroenterol Motil 32, e13802 (2020).
  6. So, S. Y. & Savidge, T. C. Sex-Bias in Irritable Bowel Syndrome: Linking Steroids to the Gut-Brain Axis. Front Endocrinol (Lausanne) 12, 684096 (2021).
  7. De Palma, G., Collins, S. M. & Bercik, P. The microbiota-gut-brain axis in functional gastrointestinal disorders. Gut Microbes 5, 419-429 (2014).
  8. Parfrey, L. W., Walters, W. A. & Knight, R. Microbial eukaryotes in the human microbiome: ecology, evolution, and future directions. Front Microbiol 2, 153 (2011).
  9. Dethlefsen, L., Eckburg, P. B., Bik, E. M. & Relman, D. A. Assembly of the human intestinal microbiota. Trends Ecol Evol 21, 517-523 (2006).
  10. Rajilic-Stojanovic, M., Heilig, H. G., Tims, S., Zoetendal, E. G. & de Vos, W. M. Long-term monitoring of the human intestinal microbiota composition. Environ Microbiol (2012).
  11. Costea, P. I. et al. Enterotypes in the landscape of gut microbial community composition. Nat Microbiol 3, 8-16 (2018).
  12. Gilbert, J. A. et al. Current understanding of the human microbiome. Nat Med 24, 392-400 (2018).
  13. Sommer, F. & Backhed, F. The gut microbiota–masters of host development and physiology. Nat Rev Microbiol. 11, 227-238 (2013).
  14. He, Y. et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat Med 24, 1532-1535 (2018).
  15. Deschasaux, M. et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nat Med 24, 1526-1531 (2018).
  16. Moya, A. & Ferrer, M. Functional Redundancy-Induced Stability of Gut Microbiota Subjected to Disturbance. Trends Microbiol 24 (2016).
  17. Nicholson, J. K. et al. Host-gut microbiota metabolic interactions. Science 336, 1262-1267 (2012).
  18. Fan, Y. & Pedersen, O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol 19, 55-71 (2021).
  19. Hansen, C. H. et al. Patterns of early gut colonization shape future immune responses of the host. PLoS.One. 7, e34043 (2012).
  20. Hapfelmeier, S. et al. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science 328, 1705-1709 (2010).
  21. Kunii, J. et al. Commensal bacteria promote migration of mast cells into the intestine. Immunobiology 216, 692-697 (2011).
  22. Olszak, T. et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336, 489-493 (2012).
  23. Slack, E. et al. Innate and adaptive immunity cooperate flexibly to maintain host-microbiota mutualism. Science 325, 617-620 (2009).
  24. Geuking, M. B. et al. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 34, 794-806 (2011).
  25. Macpherson, A. J., Geuking, M. B. & McCoy, K. D. Homeland security: IgA immunity at the frontiers of the body. Trends Immunol. 33, 160-167 (2012).
  26. De Vadder, F. et al. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc Natl Acad Sci U S A 115 (2018).
  27. Collins, J., Borojevic, R., Verdu, E. F., Huizinga, J. D. & Ratcliffe, E. M. Intestinal microbiota influence the early postnatal development of the enteric nervous system. Neurogastroenterol Motil 26, 98-107 (2014).
  28. Amaral, F. A. et al. Commensal microbiota is fundamental for the development of inflammatory pain. Proc.Natl.Acad.Sci.U.S.A 105, 2193-2197 (2008).
  29. Shen, S. et al. Gut microbiota is critical for the induction of chemotherapy-induced pain. Nat Neurosci 20, 1213-1216 (2017).
  30. Luczynski, P. et al. Microbiota regulates visceral pain in the mouse. Elife 6, doi:10.7554/eLife.25887 (2017).
  31. Abrams, G. D. & Bishop, J. E. Effect of the normal microbial flora on gastrointestinal motility. Proc.Soc.Exp.Biol.Med. 126, 301-304 (1967).
  32. Gustafsson, B. E., Midtvedt, T. & Strandberg, K. Effects of microbial contamination on the cecum enlargement of germfree rats. Scand.J.Gastroenterol. 5, 309-314 (1970).
  33. Wostmann, B. S. The germfree animal in nutritional studies. Annu.Rev.Nutr. 1, 257-279 (1981).
  34. Roager, H. M. et al. Colonic transit time is related to bacterial metabolism and mucosal turnover in the gut. Nat Microbiol 1, 16093 (2016).
  35. Anitha, M., Vijay-Kumar, M., Sitaraman, S. V., Gewirtz, A. T. & Srinivasan, S. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling. Gastroenterology 143, 1006-1016 e1004 (2012).
  36. Yarandi, S. S. et al. Intestinal Bacteria Maintain Adult Enteric Nervous System and Nitrergic Neurons via Toll-like Receptor 2-induced Neurogenesis in Mice. Gastroenterology 159, 200-213 e8 (2020).
  37. Obata, Y. et al. Neuronal programming by microbiota regulates intestinal physiology. Nature 578, 284-289 (2020).
  38. Marshall, J. K., Thabane, M., Borgaonkar, M. R. & James, C. Postinfectious irritable bowel syndrome after a food-borne outbreak of acute gastroenteritis attributed to a viral pathogen. Clin.Gastroenterol.Hepatol. 5, 457-460 (2007).
  39. Thabane, M., Kottachchi, D. T. & Marshall, J. K. Systematic review and meta-analysis: The incidence and prognosis of post-infectious irritable bowel syndrome. Aliment.Pharmacol.Ther. 26, 535-544 (2007).
  40. Thabane, M. & Marshall, J. K. Post-infectious irritable bowel syndrome. World J.Gastroenterol. 15, 3591-3596 (2009).
  41. Chaudhary, N. A. & Truelove, S. C. The irritable colon syndrome. A study of the clinical features, predisposing causes, and prognosis in 130 cases. Q J Med 31, 307-322 (1962).
  42. Berumen, A., Edwinson, A. L. & Grover, M. Post-infection Irritable Bowel Syndrome. Gastroenterol Clin North Am 50, 445-461 (2021).
  43. Spiller, R. & Lam, C. An Update on Post-infectious Irritable Bowel Syndrome: Role of Genetics, Immune Activation, Serotonin and Altered Microbiome. J.Neurogastroenterol.Motil. 18, 258-268 (2012).
  44. Mearin, F. Editorial: From the acute infection to the chronic disorder «Don’t worry it’s just a viral gastroenteritis». Am.J.Gastroenterol. 107, 900-901 (2012).
  45. Lupp, C. et al. Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae. Cell Host.Microbe 2, 204 (2007).
  46. Bushyhead, D. & Quigley, E. M. Small Intestinal Bacterial Overgrowth. Gastroenterol Clin North Am 50, 463-474 (2021).
  47. Dukowicz, A. C., Lacy, B. E. & Levine, G. M. Small intestinal bacterial overgrowth: a comprehensive review. Gastroenterol.Hepatol.(N.Y.) 3, 112-122 (2007).
  48. Pyleris, E. et al. The prevalence of overgrowth by aerobic bacteria in the small intestine by small bowel culture: relationship with irritable bowel syndrome. Dig.Dis.Sci. 57, 1321-1329 (2012).
  49. Ford, A. C., Spiegel, B. M., Talley, N. J. & Moayyedi, P. Small intestinal bacterial overgrowth in irritable bowel syndrome: systematic review and meta-analysis. Clin.Gastroenterol.Hepatol. 7, 1279-1286 (2009).
  50. Shah, A. et al. Small Intestinal Bacterial Overgrowth in Irritable Bowel Syndrome: A Systematic Review and Meta-Analysis of Case-Control Studies. Am J Gastroenterol 115, 190-201 (2020).
  51. Spiegel, B. M. Questioning the bacterial overgrowth hypothesis of irritable bowel syndrome: an epidemiologic and evolutionary perspective. Clin.Gastroenterol.Hepatol. 9, 461-469 (2011).
  52. Ghoshal, U. C. How to interpret hydrogen breath tests. J.Neurogastroenterol.Motil. 17, 312-317 (2011).
  53. Khoshini, R., Dai, S. C., Lezcano, S. & Pimentel, M. A systematic review of diagnostic tests for small intestinal bacterial overgrowth. Dig.Dis.Sci. 53, 1443-1454 (2008).
  54. Pimentel, M. Evaluating a bacterial hypothesis in IBS using a modification of Koch’s postulates: part 1. Am.J.Gastroenterol. 105, 718-721 (2010).
  55. Chey, W. D., Maneerattaporn, M. & Saad, R. Pharmacologic and complementary and alternative medicine therapies for irritable bowel syndrome. Gut Liver 5, 253-266(2011).
  56. Pimentel, M. et al. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N.Engl.J.Med. 364, 22-32 (2011).
  57. Pimentel, M. An evidence-based treatment algorithm for IBS based on a bacterial/SIBO hypothesis: Part 2. Am.J.Gastroenterol. 105, 1227-1230 (2010).
  58. Gatta, L. & Scarpignato, C. Systematic review with meta-analysis: rifaximin is effective and safe for the treatment of small intestine bacterial overgrowth. Aliment Pharmacol Ther 45, 604-616 (2017).
  59. Pittayanon, R. et al. Gut Microbiota in Patients With Irritable Bowel Syndrome-A Systematic Review. Gastroenterology 157, 97-108 (2019).
  60. Duan, R., Zhu, S., Wang, B. & Duan, L. Alterations of Gut Microbiota in Patients With Irritable Bowel Syndrome Based on 16S rRNA-Targeted Sequencing: A Systematic Review. Clin Transl Gastroenterol 10, e00012 (2019).
  61. Vich Vila, A. et al. Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome. Sci Transl Med 10 (2018).
  62. Lloyd-Price, J., Abu-Ali, G. & Huttenhower, C. The healthy human microbiome. Genome Med 8, 51 (2016).
  63. Shanahan, F., Ghosh, T. S. & O’Toole, P. W. The Healthy Microbiome-What Is the Definition of a Healthy Gut Microbiome? Gastroenterology 160, 483-494, doi:10.1053/j.gastro.2020.09.057 (2021).
  64. Baumgartner, M. et al. Mucosal Biofilms Are an Endoscopic Feature of Irritable Bowel Syndrome and Ulcerative Colitis. Gastroenterology 161, 1245-1256 e1220 (2021).
  65. Botschuijver, S. et al. Intestinal Fungal Dysbiosis Is Associated With Visceral Hypersensitivity in Patients With Irritable Bowel Syndrome and Rats. Gastroenterology 153, 1026-1039 (2017).
  66. Das, A., O’Herlihy, E., Shanahan, F., O’Toole, P. W. & Jeffery, I. B. The fecal mycobiome in patients with Irritable Bowel Syndrome. Scientific reports 11, 124 (2021).
  67. Coughlan, S. et al. The gut virome in Irritable Bowel Syndrome differs from that of controls. Gut Microbes 13, 1-15 (2021).
  68. Mars, R. A. T., Frith, M. & Kashyap, P. C. Functional Gastrointestinal Disorders and the Microbiome-What Is the Best Strategy for Moving Microbiome-based Therapies for Functional Gastrointestinal Disorders into the Clinic? Gastroenterology 160, 538-555 (2021).
  69. Mars, R. A. T. et al. Longitudinal Multi-omics Reveals Subset-Specific Mechanisms Underlying Irritable Bowel Syndrome. Cell 183, 1137-1140 (2020).
  70. Crouzet, L. et al. The hypersensitivity to colonic distension of IBS patients can be transferred to rats through their fecal microbiota. Neurogastroenterol Motil 25, e272-282 (2013).
  71. Touw, K. et al. Mutual reinforcement of pathophysiological host-microbe interactions in intestinal stasis models. Physiol Rep 5 (2017).
  72. Edogawa, S. et al. Serine proteases as luminal mediators of intestinal barrier dysfunction and symptom severity in IBS. Gut 69, 62-73 (2020).
  73. De Palma, G. et al. Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice. Sci Transl Med 9 (2017).
  74. Valdovinos-Díaz, M. A. Probióticos en Síndrome de Intestino Irritable: ¿Están listos para la práctica clínica? Acta Gastroenterologica Latinoamericana 51, 271-278 (2021).
  75. Pinto-Sanchez, M. I. & Verdu, E. F. Non-celiac gluten or wheat sensitivity: It’s complicated! Neurogastroenterol Motil 30, e13392 (2018).
  76. Dionne, J. et al. A Systematic Review and Meta-Analysis Evaluating the Efficacy of a Gluten-Free Diet and a Low FODMAPs Diet in Treating Symptoms of Irritable Bowel Syndrome. Am J Gastroenterol 113, 1290-1300 (2018).
  77. Black, C. J., Staudacher, H. M. & Ford, A. C. Efficacy of a low FODMAP diet in irritable bowel syndrome: systematic review and network meta-analysis. Gut (2021).
  78. McIntosh, K. et al. FODMAPs alter symptoms and the metabolome of patients with IBS: a randomised controlled trial. Gut (2016).
  79. Staudacher, H. M. et al. Long-term personalized low FODMAP diet improves symptoms and maintains luminal Bifidobacteria abundance in irritable bowel syndrome. Neurogastroenterol Motil, e14241 (2021).
  80. Banasiewicz, T. et al. Microencapsulated sodium butyrate reduces the frequency of abdominal pain in patients with irritable bowel syndrome. Colorectal Dis 15, 204-209 (2013).
  81. Lewis, S. J. & Heaton, K. W. Increasing butyrate concentration in the distal colon by accelerating intestinal transit. Gut 41, 245-251 (1997).
  82. Black, C. J. & Ford, A. C. Best management of irritable bowel syndrome. Frontline Gastroenterol 12, 303-315 (2021).
  83. Ford, A. C., Harris, L. A., Lacy, B. E., Quigley, E. M. M. & Moayyedi, P. Systematic review with meta-analysis: the efficacy of prebiotics, probiotics, synbiotics and antibiotics in irritable bowel syndrome. Aliment Pharmacol Ther 48, 1044-1060 (2018).
  84. Pimentel, M., Chow, E. J. & Lin, H. C. Normalization of lactulose breath testing correlates with symptom improvement in irritable bowel syndrome: a double-blind, randomized, placebo-controlled study. The American Journal of Gastroenterology 98, 412-419 (2003).
  85. Ghoshal, U. C., Srivastava, D., Misra, A. & Ghoshal, U. A proof-of-concept study showing antibiotics to be more effective in irritable bowel syndrome with than without small-intestinal bacterial overgrowth: a randomized, double-blind, placebo-controlled trial. Eur J Gastroenterol Hepatol 28, 281-289 (2016).
  86. Johnsen, P. H. et al. Faecal microbiota transplantation versus placebo for moderate-to-severe irritable bowel syndrome: a double-blind, randomised, placebo-controlled, parallel-group, single-centre trial. Lancet Gastroenterol Hepatol 3, 17-24 (2018).
  87. El-Salhy, M., Hatlebakk, J. G., Gilja, O. H., Brathen Kristoffersen, A. & Hausken, T. Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled study. Gut 69, 859-867 (2020).
  88. Holvoet, T. et al. Fecal Microbiota Transplantation Reduces Symptoms in Some Patients With Irritable Bowel Syndrome With Predominant Abdominal Bloating: Short- and Long-term Results From a Placebo-Controlled Randomized Trial. Gastroenterology 160, 145-157 e148 (2021).
  89. Halkjaer, S. I. et al. Faecal microbiota transplantation alters gut microbiota in patients with irritable bowel syndrome: results from a randomised, double-blind placebo-controlled study. Gut 67, 2107-2115 (2018).
  90. Aroniadis, O. C. et al. Faecal microbiota transplantation for diarrhoea-predominant irritable bowel syndrome: a double-blind, randomised, placebo-controlled trial. Lancet Gastroenterol Hepatol 4, 675-685 (2019).
  91. Holster, S. et al. The Effect of Allogenic Versus Autologous Fecal Microbiota Transfer on Symptoms, Visceral Perception and Fecal and Mucosal Microbiota in Irritable Bowel Syndrome: A Randomized Controlled Study. Clin Transl Gastroenterol 10, e00034 (2019).
  92. Lahtinen, P. et al. Randomised clinical trial: faecal microbiota transplantation versus autologous placebo administered via colonoscopy in irritable bowel syndrome. Aliment Pharmacol Ther 51, 1321-1331 (2020).
  93. El-Salhy, M. et al. Long-term effects of fecal microbiota transplantation (FMT) in patients with irritable bowel syndrome. Neurogastroenterol Motil, e14200 (2021).
  94. Cui, J. et al. Long-Term Follow-Up Results of Fecal Microbiota Transplantation for Irritable Bowel Syndrome: A Single-Center, Retrospective Study. Front Med (Lausanne) 8, 710452 (2021).
  95. El-Salhy, M., Hausken, T. & Hatlebakk, J. G. Current status of fecal microbiota transplantation for irritable bowel syndrome. Neurogastroenterol Motil, e14157 (2021).
  96. Collyer, R., Clancy, A. & Borody, T. Faecal microbiota transplantation alleviates symptoms of depression in individuals with irritable bowel syndrome: A case series. Medicine in Microecology 6, 100029 (2020).

Correspondence: Giada De Palma
Email: depalma@mcmaster.ca

Acta Gastroenterol Latinoam 2021;51(4):378-384

Otros Artículos

La era de la endoscopia inteligente: cómo la inteligencia artificial potencia la endoscopia digestiva

Jorge Baquerizo -Burgos ID· María Egas-Izquierdo ID· Doménica Cunto ID· Carlos Robles-Medranda ID Departamento de …