Hostname: page-component-8448b6f56d-sxzjt Total loading time: 0 Render date: 2024-04-23T23:42:10.867Z Has data issue: false hasContentIssue false

Irritable bowel syndrome: the problem and the problem of treating it – is there a role for probiotics?

Published online by Cambridge University Press:  22 August 2014

Antonieta R. Santos
Affiliation:
Neurogastroenterology Unit, University Hospital of South Manchester, Manchester, UK
P. J. Whorwell*
Affiliation:
Neurogastroenterology Unit, University Hospital of South Manchester, Manchester, UK
*
*Corresponding author: Professor P. J. Whorwell, fax +(0)161 291 5813, email peter.whorwell@uhsm.nhs.uk
Rights & Permissions [Opens in a new window]

Abstract

The aim of this review is to highlight the impact of irritable bowel syndrome (IBS) in those patients who consult the medical profession and examine the therapeutic potential of probiotics in this condition, where there is a strong need for new treatment options. Traditionally, IBS is frequently regarded as a trivial condition which is certainly not life threatening and mainly psychological in origin. However, these preconceptions are misplaced, as in some patients the condition can be devastating with the pain being as severe as that of childbirth coupled with incapacitating bowel dysfunction. In addition, patients suffer from a variety of non-colonic symptoms such as low backache, constant lethargy, nausea and genito-urinary problems, all of which lead to these patients having extremely poor quality of life. Unfortunately, the treatment of IBS is very unsatisfactory with only one new medication being developed for this condition in the last 25 years. It is now recognised that IBS is a multifactorial condition with symptoms being triggered by a variety of factors, some of which appear to be influenced by probiotics, resulting in speculation that they may have therapeutic potential in this condition. There have been over thirty controlled clinical trials of probiotics in IBS with approximately two-thirds of these studies showing evidence of an improvement in symptoms. However, not all probiotics appear to be effective with different symptoms being improved by different strains and some improving symptoms more than others. Consequently, the ideal probiotic for the treatment of IBS has yet to be defined, but the evidence is good enough to encourage further research with the aim of identifying an optimal strain or strains.

Type
Conference on ‘Diet, gut microbiology and human health’
Copyright
Copyright © The Authors 2014 

The problem

Irritable bowel syndrome (IBS) is often viewed as a relatively mild condition which is psychological in origin and certainly not life threatening. Approximately 10–15 % of the population are affected( Reference Wilson, Roberts and Roalfe 1 ) and it is certainly true that in a proportion of these individuals the condition is more of a nuisance than being anything serious and they seldom, if ever, have to consult their doctor. However, the condition can be much more severe in a substantial minority necessitating repetitive consultation both in the primary and secondary care setting and placing a significant burden on healthcare systems because the treatment is so unsatisfactory( Reference Spiegel 2 , Reference Maxion-Bergemann, Thielecke and Abel 3 ). The principal symptoms are abdominal pain, abdominal bloating and some form of bowel dysfunction which can be either diarrhoea (IBS-D), constipation (IBS-C) or an alternation between the two (IBS-A). Over the years, a number of diagnostic criteria for IBS have been developed of which the Rome III criteria( Reference Drossman, Corazziari and Delvaux 4 ) are the most widely used, although a further update is in development. These criteria are extremely useful for ensuring homogeneity of patient groups for research purposes but rather cumbersome for use in the clinical setting. In the more severe cases of IBS, many women liken the pain to that of childbirth and can experience this on a regular, sometimes daily, basis( Reference Agrawal and Whorwell 5 ). In the diarrhoea group of patients, there can be significant urgency and some patients experience faecal incontinence which can have an extremely restricting effect on their lifestyle( Reference Agrawal and Whorwell 5 ). The constipation variety is characterised by infrequent defecation and it is not uncommon for some patients to fail to open their bowels for up to 1–2 weeks. The gastrocolonic reflex is a response to eating where a healthy individual experiences a desire to open their bowels shortly after eating their first meal of the day and not with any subsequent intake of food. In IBS this reflex is exaggerated, so that patients with the diarrhoea predominant form of the condition often need to open their bowels after every meal whereas in those with constipation, abdominal pain or bloating is made worse by eating. Bloating is another extremely common feature of IBS which can take the form of a feeling of pressure within the abdomen which, in some individuals, can be accompanied by an actual increase in girth usually referred to as distension( Reference Agrawal and Whorwell 6 , Reference Whorwell 7 ). In addition, IBS patients often suffer from a variety of non-colonic symptoms such as low backache, constant lethargy, nausea and a range of urological and gynaecological symptoms such as frequency of micturition and pain on intercourse( Reference Whorwell, McCallum and Creed 8 ). Not surprisingly, the latter can profoundly affect sexual activity in women suffering from this problem( Reference Guthrie, Creed and Whorwell 9 ). These non-colonic symptoms are important as on some occasions the patients find them more intrusive than the traditional symptoms of IBS. Furthermore, they can lead to inappropriate referral to gynaecological, urological and even orthopaedic clinics where the outcome is poor because they actually have IBS rather than a condition relating to these specialities( Reference Prior and Whorwell 10 , Reference Francis, Duffy and Whorwell 11 ). Altogether, these features of IBS lead to the erosion of the quality of life of sufferers to the extent that it can be equal to or worse than that of diseases such as diabetes and chronic renal disease( Reference Gralnek, Hays and Kilbourne 12 ). All these issues coupled with the inadequacies of current treatment options can lead to a sense of hopelessness and it has been shown that suicidal ideation is worryingly high in many of these patients( Reference Miller, Hopkins and Whorwell 13 ). Consequently, there is a strong need for the development of new treatment strategies for IBS.

The pathophysiology of irritable bowel syndrome and the influence of probiotics

It is now recognised that there is no single cause of IBS with disturbances of gastrointestinal motility and sensitivity, the central processing of painful stimuli from the gut, inflammation, diet, genetic and psychological factors all being important( Reference Drossman, Camilleri and Mayer 14 ). Furthermore, there is an accumulating body of evidence that the microbiota of the gut is disturbed (dysbiosis) in at least a proportion of patients( Reference Simren, Barbara and Flint 15 ) although no particular phenotype characteristic of IBS has yet been identified. The presence of dysbiosis, coupled with accumulating evidence that some of the pathophysiological mechanisms involved in IBS can be modified by probiotics, has led to speculation that probiotics might be effective in treating the condition( Reference Whorwell 16 ).

The administration of probiotics can influence motility( Reference Verdu, Bercik and Collins 17 , Reference Quigley and Flourie 18 ) which is known to be abnormal in IBS( Reference Hasler 19 ), and in particular, there is convincing evidence that gastrointestinal transit can be accelerated by some organisms (Fig. 1)( Reference Agrawal, Houghton and Morris 20 Reference Waller, Gopal and Leyer 30 ) suggesting those that have this effect would be better suited to IBS-C( Reference Miller and Ouwehand 31 ). Visceral hypersensitivity of the gastrointestinal tract is one of the most consistent abnormalities in patients with IBS( Reference Ritchie 32 , Reference Azpiroz, Bouin and Camilleri 33 ) and is usually assessed by balloon distension of the rectum. Although the effect of probiotics on visceral hypersensitivity has not yet been assessed in IBS, it has been shown that they can reduce this abnormality in experimental animals( Reference Johnson, Greenwood-Van Meerveld and McRorie 34 , Reference Agostini, Goubern and Tondereau 35 ) using balloon distension techniques. Brain scanning techniques, such as positron emission tomography or functional MRI, have been utilised to provide good evidence that the central processing of painful stimuli applied to the gut is abnormal in IBS( Reference Naliboff, Derbyshire and Munakata 36 , Reference Mertz, Morgan and Tanner 37 ). It is therefore of interest that it has been shown that the administration of a probiotic can reduce the reactivity of areas in the brain associated with this abnormal processing( Reference Tillisch, Labus and Kilpatrick 38 ). It has been known for many years that some patients with IBS date the onset of their symptoms to an episode of gastroenteritis( Reference Spiller and Lam 39 ) and this condition is sometimes referred to as post-infective IBS. It remains to be determined whether IBS-D and post-infective IBS are similar entities, although a recent study has suggested that the microbiota in these two conditions is very similar( Reference Jalanka-Tuovinen, Salojärvi and Salonen 40 ). It has been shown that in a proportion of individuals with post-infective IBS and other forms of IBS, there is evidence of a persisting, low grade, inflammation in the gastrointestinal mucosa( Reference Gwee, Leong and Graham 41 , Reference Feng, La and Schwartz 42 ). This abnormality might be amenable to modulation by some species of probiotic organisms that have been shown to elaborate a variety of proteins and metabolites that have anti-inflammatory and immunomodulatory activity( Reference O'Mahony, McCarthy and Kelly 43 , Reference Barbara, Zecchi and Barbaro 44 ). For instance, it has been shown that in some patients with IBS the ratio between IL10 (an anti-inflammatory cytokine) and IL12 (a proinflammatory cytokine) is in a pro-inflammatory state and this can be normalised by the administration of Bifidobacterium infantis 35 624( Reference O'Mahony, McCarthy and Kelly 43 ). In addition to having dysbiosis( Reference Simren, Barbara and Flint 15 ), it is a common clinical observation that patients with IBS frequently give a history of repeated or prolonged antibiotic consumption. This is supported by a prospective study showing that 4 months after receiving antibiotics, 48 % of patients experienced functional bowel symptoms compared with only 22 % of controls not receiving such a medication( Reference Maxwell, Rink and Kumar 45 ). There is also some evidence that a proportion of patients with IBS have small-bowel bacterial overgrowth( Reference Pimentel, Kong and Park 46 ) although this finding remains somewhat controversial because of dispute over what is the optimum method for detecting this abnormality( Reference Simren, Barbara and Flint 15 ). Furthermore, some of the medications that are commonly used in IBS can affect the microbiota and this is particularly true for proton pump inhibitors and laxatives or anti-diarrhoeal medications( Reference Simren, Barbara and Flint 15 ). Consequently, the observation that probiotics have been shown to be beneficial in antibiotic diarrhoea and that some species even have anti-microbial activity( Reference Hempel, Newberry and Maher 47 , Reference Heineman, Bubenik and McClave 48 ) suggests a possible role in targeting the dysbiosis in IBS. There is growing evidence that a defect of the gastrointestinal barrier, which helps to confine a variety of bacterial and other antigens to the gut lumen, plays an important role in the pathogenesis of a number of gastrointestinal diseases, including IBS( Reference Camilleri, Madsen and Spiller 49 ). Consequently, there is considerable interest in understanding how this barrier is maintained coupled with the fact that there is experimental support for the notion that probiotic bacteria enhance barrier function( Reference Natividad and Verdu 50 , Reference Madsen, Cornish and Soper 51 ). We have shown in a number of studies that the prevalence of depression is not especially high in patients with IBS, although anxiety is a consistently common finding in this condition( Reference Gonsalkorale, Cooper and Cruickshanks 52 ). Therefore, it is noteworthy that the administration of probiotics has been shown to lead to a reduction in anxiety and depression behaviour in animals( Reference Desbonnet, Garrett and Clarke 53 ), as well as reduced anxiety scores in human subjects( Reference Benton, Williams and Brown 54 , Reference Messaoudi, Lalonde and Violle 55 ).

Fig. 1. A comparison of studies measuring the effect of a variety of different probiotics on gastrointestinal transit time. A positive value equates to an acceleration of transit.

Despite the fact that a substantial proportion of patients with IBS give a history of antibiotic usage, it is interesting to note that some non-absorbable antibiotics may actually have a beneficial effect. Originally this observation was noted for neomycin but this medication can have side effects and more recently, attention has focused on rifaximin( Reference Menees, Maneerattannaporn and Kim 56 ). The beneficial effects of this antibiotic have been well documented in large-scale clinical trials although some questions remain about how often such an antibiotic should be given and whether repeated courses could lead to drug resistance. However, the observation that this antibiotic can have beneficial effects in IBS lends further support to the notion that targeting the microbiota in this condition has therapeutic potential.

Treating irritable bowel syndrome with probiotics

Most of the drugs in development for the treatment of IBS target a specific receptor and consequently are aimed at a particular subgroup of the condition, such as IBS-C or IBS-D, whereas the more traditional medications, such as antispasmodics, tend to be given to all patients irrespective of the pattern of their symptomatology. Similarly, because probiotics potentially modulate a number of the pathophysiological mechanisms involved in IBS, they are usually given to all subtypes of the condition both in the clinical setting as well as in therapeutic trials. There have been twenty-eight controlled trials assessing the efficacy of probiotics in adults suffering from IBS with twenty (71 %) showing a positive effect( Reference O'Mahony, McCarthy and Kelly 43 , Reference Gade and Thorn 57 Reference Hong, Kang and Im 75 ), seven showing no effect( Reference O'Mahony, McCarthy and Kelly 43 , Reference Sen, Mullan and Parker 76 Reference Sondergaard, Olsson and Ohlson 81 ) and one leading to a deterioration of symptoms( Reference Ligaarden, Axelsson and Naterstad 82 ). In addition, four trials in children all gave positive results( Reference Bausserman and Michail 83 Reference Guandalini, Magazzu and Chiaro 86 ). However, the quality of some of these trials has not been optimal and there is always the problem of the possibility of publication bias. In addition, formulation is critical with efficacy not necessarily improving with a higher concentration of bacteria. For instance, a liquid probiotic preparation containing 1010 colony-forming units of B. infantis was effective in IBS, whereas the same dose in a capsule had no effect because in this form, it solidified on contact with moisture( Reference Whorwell, Altringer and Morel 61 ). Although many trials show an improvement in symptoms, different preparations appear to improve different symptoms with some helping pain, others reducing flatulence and some relieving a range of symptoms( Reference Drossman, Camilleri and Mayer 14 ). A meta analysis of probiotic trials would be inappropriate, as for instance, different strains of bifidobacteria will have unique properties not necessarily shared by other closely related organisms. However, it is entirely acceptable to compare different therapeutic trials assessing the efficacy of the same organism or product.

As the majority of studies have been undertaken on IBS, irrespective of bowel habit subtype, it is difficult to know whether some may be more suited to one subtype or another. In one trial on B. infantis, although all subtypes were included( Reference Whorwell, Altringer and Morel 61 ), the results were broken down according to subtype (Table 1) and it can be seen that the preparation was equally effective in all three groups. However, in those studies where transit was accelerated (Fig. 1), it might be anticipated that those products might be more suited to constipation type IBS. An increase in abdominal girth, sometimes referred to as distension, is an extremely common feature of IBS and has been shown to be associated with constipation and delayed gastrointestinal transit( Reference Houghton, Lea and Agrawal 87 , Reference Agrawal, Houghton and Reilly 88 ). Consequently, it might be expected that this symptom might be improved by relieving constipation or accelerating gastrointestinal transit and this has been confirmed using a probiotic yoghurt that has been shown to reduce transit times( Reference Agrawal, Houghton and Morris 20 ).

Table 1. Comparison of the change in symptom scores (negative value equals improvement) in various irritable bowel syndrome (IBS) bowel habit subtypes* following treatment with Bifidobacterium infantis or placebo

* The principal symptoms of IBS are abdominal pain, abdominal bloating and some form of bowel dysfunction. The latter can take the form of diarrhoea, constipation or an alternation between the two and these different types of IBS are usually referred to as IBS-D, IBS-C and IBS-A respectively.

Despite the evidence that probiotics may be potentially useful in the treatment of IBS, a number of questions remain to be answered. For instance, it is still not known whether preparations containing single organisms or mixtures are preferable or whether there are any contraindications to their use, such as administration to immunocompromised patients. Those patients most likely to respond, such as possibly individuals with a history of excessive antibiotic consumption, have also yet to be defined but it might be anticipated that these products are going to be more effective in combination with other approaches such as dietary manipulation, rather than being stand-alone treatments. Their beneficial effects seem to evolve quite slowly and symptoms start to return when treatment is discontinued( Reference Whorwell, Altringer and Morel 61 ) indicating that it is likely that long-term maintenance treatment will be required when a particular preparation is found to be effective.

In conclusion, there is reasonably good evidence that probiotic organisms have activity in the treatment of IBS but the best organism(s) for this purpose have yet to be defined. The availability of products for which there are data to support their use in IBS is patchy and varies from country to country. Consequently the best advice that can be given at the present time, is to try and source an evidence-based product and take it for at least 4 weeks.

Acknowledgements

Antonieta R Santos is a visiting clinical fellow funded by the Gastroenterology Department at the Hospital Amato Lusitano in Castelo Branco, Portugal.

Financial Support

None.

Conflicts of Interest

P. J. W. has acted as a consultant for, or received research grant support from the following pharmaceutical companies: Almirall Pharma, Boehringer–Ingelheim, Chr Hansen, Abbott, Danone Research, Ironwood Pharmaceuticals, Norgine, Proctor and Gamble, Shire UK and Sucampo Pharmaceuticals.

Authorship

Both authors reviewed the literature and worked on the manuscript, agreeing on the final version.

References

1. Wilson, S, Roberts, L, Roalfe, A et al. (2004) Prevalence of irritable bowel syndrome: a community survey. Br J Gen Pract 54, 495502.Google Scholar
2. Spiegel, BM (2009) The burden of IBS: looking at metrics. Curr Gastroenterol Rep 11, 265269.Google Scholar
3. Maxion-Bergemann, S, Thielecke, F, Abel, F et al. (2006) Costs of irritable bowel syndrome in the UK and US. Pharmacoeconomics 24, 2137.CrossRefGoogle ScholarPubMed
4. Drossman, DA, Corazziari, E, Delvaux, M et al. (2006) Rome III: The Functional Gastrointestinal Disorders, 3rd ed. McLean, Virginia, USA: Degnon Associated.Google Scholar
5. Agrawal, A & Whorwell, PJ (2006) Irritable bowel syndrome: diagnosis and management. Br Med J 332, 280283.Google Scholar
6. Agrawal, A & Whorwell, PJ (2008) Review article: abdominal bloating and distension in functional gastrointestinal disorders – epidemiology and exploration of possible mechanisms. Aliment Pharmacol Ther 27, 210.Google Scholar
7. Whorwell, PJ (2012) Unraveling functional abdominal bloating and distension: the role of thoraco-abdominal accommodation and a physical sign to aid its detection. Neurogastroenterol Motil 24, 301304.Google Scholar
8. Whorwell, PJ, McCallum, M, Creed, FH et al. (1986) Non-colonic features of irritable bowel syndrome. Gut 27, 3740.Google Scholar
9. Guthrie, E, Creed, FH & Whorwell, PJ (1987) Severe sexual dysfunction in women with the irritable bowel syndrome: comparison with inflammatory bowel disease and duodenal ulceration. Br Med J 295, 577578.Google Scholar
10. Prior, A & Whorwell, PJ (1989) Gynaecological consultation in patients with the irritable bowel syndrome. Gut 30, 996998.Google Scholar
11. Francis, CY, Duffy, JN, Whorwell, PJ et al. (1997) High prevalence of irritable bowel syndrome in patients attending urological outpatient departments. Dig Dis Sci 42, 404407.Google Scholar
12. Gralnek, IM, Hays, RD, Kilbourne, A et al. (2000) The impact of irritable bowel syndrome on health-related quality of life. Gastroenterology 119, 654660.CrossRefGoogle ScholarPubMed
13. Miller, V, Hopkins, L & Whorwell, PJ (2004) Suicidal ideation in patients with irritable bowel syndrome. Clin Gastroenterol Hepatol 2, 10641068.CrossRefGoogle ScholarPubMed
14. Drossman, DA, Camilleri, M, Mayer, EA et al. (2002) AGA technical review on irritable bowel syndrome. Gastroenterology 123, 21082131.Google Scholar
15. Simren, M, Barbara, G, Flint, HJ et al. (2013) Intestinal microbiota in functional bowel disorders: a Rome foundation report. Gut 62, 159176.Google Scholar
16. Whorwell, PJ (2009) Do probiotics improve symptoms in patients with irritable bowel syndrome? Therap Adv Gastroenterol 2, 3744.Google Scholar
17. Verdu, EF, Bercik, P & Collins, SM (2009) Effect of probiotics on gastrointestinal function: evidence from animal models. Therap Adv Gastroenterol 2, 3135.Google Scholar
18. Quigley, EM & Flourie, B (2007) Probiotics and irritable bowel syndrome: a rationale for their use and an assessment of the evidence to date. Neurogastroenterol Motil 19, 166172.Google Scholar
19. Hasler, WL (2011) Traditional thoughts on the pathophysiology of irritable bowel syndrome. Gastroenterol Clin North Am 40, 2143.Google Scholar
20. Agrawal, A, Houghton, LA, Morris, J et al. (2009) Clinical trial: the effects of a fermented milk product containing Bifidobacterium lactis DN-173 010 on abdominal distension and gastrointestinal transit in irritable bowel syndrome with constipation. Aliment Pharmacol Ther 29, 104114.Google Scholar
21. Bartram, HP, Scheppach, W, Gerlach, S et al. (1994) Does yogurt enriched with Bifidobacterium longum affect colonic microbiology and fecal metabolites in health subjects? Am J Clin Nutr 59, 428432.Google Scholar
22. Bouvier, M, Meance, S, Bouley, C et al. (2001) Effects of consumption of a milk fermented by the probiotic strain Bifidobacterium animalis DN-173 010 on colonic transit time in healthy humans. Biosci Microflora 20, 4348.Google Scholar
23. Holma, R, Hongisto, SM, Saxelin, M et al. (2010) Constipation is relieved more by rye bread than wheat bread or laxatives without increased adverse gastrointestinal effects. J Nutr 140, 534541.Google Scholar
24. Hongisto, SM, Paajanen, L, Saxelin, M et al. (2006) A combination of fibre-rich rye bread and yoghurt containing Lactobacillus GG improves bowel function in women with self-reported constipation. Eur J Clin Nutr 60, 319324.Google Scholar
25. Krammer, HJ, Seggem, HV, Schaumburg, J et al. (2011) Effect of Lactobacillus casei Shirota on colonic transit time in patients with chronic constipation. Coloproctology 33, 109113.Google Scholar
26. Malpeli, A, González, S, Vicentin, D et al. (2012) Randomised, double-blind and placebo-controlled study of he effect of a synbiotic dairy product on orocecal transit time in healthy adult women. Nutr Hosp 27, 13141319.Google Scholar
27. Marteau, P, Cuillerier, E, Meance, S et al. (2002) Bifidobacterium animalis strain DN-173 010 shortens the colonic transit time in healthy women: a double-blind, randomized, controlled study. Aliment Pharmacol Ther 16, 587593.Google Scholar
28. Rosenfeldt, V, Paerregaard, A, Nexmann, CL et al. (2003) Faecal recovery, mucosal adhesion, gastrointestinal effects and tolerance of mixed cultures of potential prebiotic lactobacilli. Microb Ecol Health Dis 15, 29.Google Scholar
29. Sairanen, U, Piirainen, L, Gråsten, S et al. (2007) The effect of probiotic fermented milk and inulin on the functions and microecology of the intestine. J Dairy Res 74, 367373.Google Scholar
30. Waller, PA, Gopal, PK, Leyer, GJ et al. (2011) Dose-response effect of Bifidobacterium lactis HN019 on whole gut transit time and functional gastrointestinal symptoms in adults. Scand J Gastroenterol 46, 10571064.Google Scholar
31. Miller, LE & Ouwehand, AC (2013) Probiotic supplementation decreases intestinal transit time: meta-analysis of randomized controlled trials. World J Gastroenterol 19, 47184725.Google Scholar
32. Ritchie, J (1973) Pain from distension of the pelvic colon by inflating a balloon in the irritable colon syndrome. Gut 14, 125132.Google Scholar
33. Azpiroz, F, Bouin, M, Camilleri, M et al. (2007) Mechanisms of hypersensitivity in IBS and functional disorders. Neurogastroenterol Motil 19, 6288.CrossRefGoogle ScholarPubMed
34. Johnson, AC, Greenwood-Van Meerveld, B & McRorie, J (2011) Effects of Bifidobacterium infantis 35624 on post-inflammatory visceral hypersensitivity in the rat. Dig Dis Sci 56, 31793186.Google Scholar
35. Agostini, S, Goubern, M, Tondereau, V et al. (2012) A marketed fermented dairy product containing Bifidobacterium lactis CNCM I-2494 suppresses gut hypersensitivity and colonic barrier disruption induced by acute stress in rats. Neurogastroenterol Motil 24, 376–e172.Google Scholar
36. Naliboff, BD, Derbyshire, SW, Munakata, J et al. (2001) Cerebral activation in patients with irritable bowel syndrome and control subjects during rectosigmoid stimulation. Psychosom Med 63, 365375.Google Scholar
37. Mertz, H, Morgan, V, Tanner, G et al. (2000) Regional cerebral activation in irritable bowel syndrome and control subjects with painful and nonpainful rectal distention. Gastroenterology 118, 842848.Google Scholar
38. Tillisch, K, Labus, J, Kilpatrick, L et al. (2013) Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 144, 13941401, e1391–1394.Google Scholar
39. Spiller, R & Lam, C (2012) An update on post-infectious irritable bowel syndrome: role of genetics, immune activation, serotonin and altered microbiome. J Neurogastroenterol 18, 258268.CrossRefGoogle ScholarPubMed
40. Jalanka-Tuovinen, J, Salojärvi, J, Salonen, A et al. (2013) Faecal microbiota composition and host-microbe cross-talk following gastroenteritis and in postinfectious irritable bowel syndrome. Gut (Epublication ahead of print version).Google Scholar
41. Gwee, KA, Leong, YL, Graham, C et al. (1999) The role of psychological and biological factors in postinfective gut dysfunction. Gut 44, 400406.Google Scholar
42. Feng, B, La, JH, Schwartz, ES et al. (2012) Irritable bowel syndrome: methods, mechanisms, and pathophysiology. Neural and neuro-immune mechanisms of visceral hypersensitivity in irritable bowel syndrome. Am J Psysiol Gastrointest Liver Physiol 302, G1085G1098.Google Scholar
43. O'Mahony, L, McCarthy, J, Kelly, P et al. (2005) Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology 128, 541551.Google Scholar
44. Barbara, G, Zecchi, L, Barbaro, R et al. (2012) Mucosal permeability and immune activation as potential therapeutic targets of probiotics in irritable bowel syndrome. J Clin Gastroenterol 46, Suppl., S52S55.CrossRefGoogle ScholarPubMed
45. Maxwell, PR, Rink, E, Kumar, D et al. (2002) Antibiotics increase functional abdominal symptoms. Am J Gastroenterol 97, 104108.Google Scholar
46. Pimentel, M, Kong, Y & Park, S (2003) Breath testing to evaluate lactose intolerance in irritable bowel syndrome correlates with lactulose testing and may not reflect true lactose malabsorption. Am J Gastroenterol 98, 27002704.Google Scholar
47. Hempel, S, Newberry, SJ, Maher, AR et al. (2012) Probiotics for the prevention and treatment of antibiotic-associated diarrhea: a systematic review and meta-analysis. J Am Med Assoc 307, 19591969.Google Scholar
48. Heineman, J, Bubenik, S, McClave, S et al. (2012) Fighting fire with fire: is it time to use probiotics to manage pathogenic bacterial diseases? Curr Gastroenterol Rep 14, 343348.Google Scholar
49. Camilleri, M, Madsen, K, Spiller, R et al. (2012) Intestinal barrier function in health and gastrointestinal disease. Neurogastroenterol Motil 24, 503512.Google Scholar
50. Natividad, JM & Verdu, EF (2013) Modulation of intestinal barrier by intestinal microbiota: pathological and therapeutic implications. Pharmacol Res 69, 4251.Google Scholar
51. Madsen, K, Cornish, A, Soper, P et al. (2001) Probiotic bacteria enhance murine and human intestinal epithelial barrier function. Gastroenterology 121, 580591.Google Scholar
52. Gonsalkorale, WM, Cooper, P, Cruickshanks, PB et al. (2002) Hypnotherapy in irritable bowel syndrome: a large scale audit of a clinical service with examination of factors influencing responsiveness. Am J Gastroenterol 97, 954961.Google Scholar
53. Desbonnet, L, Garrett, L, Clarke, G et al. (2008) The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J Psychiatr Res 43, 164174.Google Scholar
54. Benton, D, Williams, C & Brown, A (2007) Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur J Clin Nutr 61, 355361.CrossRefGoogle ScholarPubMed
55. Messaoudi, M, Lalonde, R, Violle, N et al. (2011) Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br J Nutr 105, 755764.Google Scholar
56. Menees, SB, Maneerattannaporn, M, Kim, HM et al. (2012) The efficacy and safety of rifaximin for the irritable bowel syndrome: a systematic review and meta-analysis. Am J Gastroenterol 107, 2835. quiz 36.Google Scholar
57. Gade, J & Thorn, P (1989) Paraghurt for patients with irritable bowel syndrome. A controlled clinical investigation from general practice. Scand J Prim Health Care 7, 2326.CrossRefGoogle ScholarPubMed
58. Halpern, GM, Prindiville, T, Blankenburg, M et al. (1996) Treatment of irritable bowel syndrome with lacteol fort: a randomized, double-blind, cross-over trial. Am J Gastroenterol 91, 15791585.Google Scholar
59. Nobaek, S, Johansson, ML, Molin, G et al. (2000) Alteration of intestinal microflora is associated with reduction in abdominal bloating and pain in patients with irritable bowel syndrome. Am J Gastroenterol 95, 12311238.Google Scholar
60. Niedzielin, K, Kordecki, H & Birkenfeld, B (2001) A controlled, double-blind, randomized study on the efficacy of Lactobacillus plantarum 299 V in patients with irritable bowel syndrome. Eur J Gastroenterol Hepatol 13, 11431147.Google Scholar
61. Whorwell, PJ, Altringer, L, Morel, J et al. (2006) Efficacy of an encapsulated probiotic Bifidobacterium infantis 35624 in women with irritable bowel syndrome. Am J Gastroenterol 101, 15811590.Google Scholar
62. Guyonnet, D, Chassany, O, Ducrotte, P et al. (2007) Effect of a fermented milk containing Bifidobacterium animalis DN-173 010 on the health-related quality of life and symptoms in irritable bowel syndrome in adults in primary care: a multicentre, randomized, double-blind, controlled trial. Aliment Pharmacol Ther 26, 475486.Google Scholar
63. Agrawal, A, Houghton, LA, Morris, J et al. (2008) Clinical trial: the effects of a fermented milk product containing Bifidobacterium lactis DN-173-010 on abdominal distension and gastrointestinal transit in irritable bowel syndrome with constipation. Aliment Pharmacol Ther 29, 104114.Google Scholar
64. Guglielmetti, S, Mora, D, Gschwender, M et al. (2011) Randomised clinical trial: Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome and improves quality of life, a double-blind, placebo-controlled study. Aliment Pharmacol Ther 33, 11231132.Google Scholar
65. Dolin, BJ (2009) Effects of a proprietary Bacillus coagulans preparation on symptoms of diarrhea-predominant irritable bowel syndrome. Methods Find Exp Clin Pharmacol 31, 655659.CrossRefGoogle ScholarPubMed
66. Kruis, W, Chrubasik, S, Boehm, S et al. (2012) A double-blind placebo-controlled trial to study therapeutic effects of probiotic Escherichia coli nissle 1917 in subgroups of patients with irritable bowel syndrome. Int J Colorectal Dis 27, 467474.Google Scholar
67. Kim, HJ, Camilleri, M, McKinzie, S et al. (2003) A randomized controlled trial of a probiotic, VSL#3, on gut transit and symptoms in diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther 17, 895904.Google Scholar
68. Kim, HJ, Vazquez Roque, MI, Camilleri, M et al. (2005) A randomized controlled trial of a probiotic combination VSL# 3 and placebo in irritable bowel syndrome with bloating. Neurogastroenterol Motil 17, 687696.Google Scholar
69. Kim, YG, Moon, JT, Lee, KM et al. (2006) The effects of probiotics on symptoms of irritable bowel syndrome. Korean J Gastroenterol 47, 413419.Google Scholar
70. Kajander, K, Hatakka, K, Poussa, T et al. (2005) A probiotic mixture alleviates symptoms in irritable bowel syndrome patients: a controlled 6-month intervention. Aliment Pharmacol Ther 22, 387394.Google Scholar
71. Kajander, K, Myllyluoma, E, Rajilic-Stojanovic, M et al. (2008) Clinical trial: multispecies probiotic supplementation alleviates the symptoms of irritable bowel syndrome and stabilizes intestinal microbiota. Aliment Pharmacol Ther 27, 4857.Google Scholar
72. Williams, E, Stimpson, J, Wang, D et al. (2009) Clinical trial: a multistrain probiotic preparation significantly reduces symptoms of irritable bowel syndrome in a double-blind placebo-controlled study. Aliment Pharmacol Ther 29, 97103.Google Scholar
73. Sinn, DH, Song, JH, Kim, HJ et al. (2008) Therapeutic effect of Lactobacillus acidophilus-SDC 2012, 2013 in patients with irritable bowel syndrome. Dig Dis Sci 53, 27142718.Google Scholar
74. Enck, P, Zimmermann, K, Menke, G et al. (2008) A mixture of Escherichia coli (DSM 17252) and Enterococcus faecalis (DSM 16440) for treatment of the irritable bowel syndrome – a randomized controlled trial with primary care physicians. Neurogastroenterol Motil 20, 11031109.Google Scholar
75. Hong, KS, Kang, HW, Im, JP et al. (2009) Effect of probiotics on symptoms in Korean adults with irritable bowel syndrome. Gut Liver 3, 101107.CrossRefGoogle ScholarPubMed
76. Sen, S, Mullan, MM, Parker, TJ et al. (2002) Effect of Lactobacillus plantarum 299v on colonic fermentation and symptoms of irritable bowel syndrome. Dig Dis Sci 47, 26152620.CrossRefGoogle ScholarPubMed
77. O'Sullivan, MA & O'Morain, CA (2000) Bacterial supplementation in the irritable bowel syndrome. A randomised double-blind placebo-controlled crossover study. Dig Liver Dis 32, 294301.Google Scholar
78. Niv, E, Naftali, T, Hallak, R et al. (2005) The efficacy of Lactobacillus reuteri ATCC 55730 in the treatment of patients with irritable bowel syndrome – a double blind, placebo-controlled, randomized study. Clin Nutr 24, 925931.Google Scholar
79. Drouault-Holowacz, S, Bieuvelet, S, Burckel, A et al. (2008) A double blind randomized controlled trial of a probiotic combination in 100 patients with irritable bowel syndrome. Gastroenterol Clin Biol 32, 147152.Google Scholar
80. Simren, M, Ohman, L, Olsson, J et al. (2010) Clinical trial: the effects of a fermented milk containing three probiotic bacteria in patients with irritable bowel syndrome – randomized, double-blind, controlled study. Aliment Pharmacol Ther 31, 218227.Google Scholar
81. Sondergaard, B, Olsson, J, Ohlson, K et al. (2011) Effects of probiotic fermented milk on symptoms and intestinal flora in patients with irritable bowel syndrome: a randomized, placebo-controlled trial. Scand J Gastroenterol 46, 663672.Google Scholar
82. Ligaarden, SC, Axelsson, L, Naterstad, K et al. (2010) A candidate probiotic with unfavourable effects in subjects with irritable bowel syndrome: a randomised controlled trial. BMC Gastroenterol 10, 16.Google Scholar
83. Bausserman, M & Michail, S (2005) The use of Lactobacillus GG in irritable bowel syndrome in children: a double-blind randomized control trial. J Pediatr 147, 197201.Google Scholar
84. Gawronska, A, Dziechciarz, P, Horvath, A et al. (2007) A randomized double-blind placebo-controlled trial of Lactobacillus GG for abdominal pain disorders in children. Aliment Pharmacol Ther 25, 177184.Google Scholar
85. Francavilla, R, Miniello, V, Magista, AM et al. (2010) A randomized controlled trial of Lactobacillus GG in children with functional abdominal pain. Pediatrics 126, 14451452.Google Scholar
86. Guandalini, S, Magazzu, G, Chiaro, A et al. (2010) VSL#3 improves symptoms in children with irritable bowel syndrome: a multicenter, randomized, placebo-controlled, double-blind, crossover study. J Pediatr Gastroenterol Nutr 51, 2430.Google Scholar
87. Houghton, LA, Lea, R, Agrawal, A et al. (2006) Relationship of abdominal bloating to distension in irritable bowel syndrome and effect of bowel habit. Gastroenterology 131, 10031010.Google Scholar
88. Agrawal, A, Houghton, LA, Reilly, B et al. (2009) Bloating and distension in irritable bowel syndrome: the role of gastrointestinal transit. Am J Gastroenterol 104, 19982004.Google Scholar
Figure 0

Fig. 1. A comparison of studies measuring the effect of a variety of different probiotics on gastrointestinal transit time. A positive value equates to an acceleration of transit.

Figure 1

Table 1. Comparison of the change in symptom scores (negative value equals improvement) in various irritable bowel syndrome (IBS) bowel habit subtypes* following treatment with Bifidobacterium infantis or placebo