101
|
Tuck CJ, De Palma G, Takami K, Brant B, Caminero A, Reed DE, Muir JG, Gibson PR, Winterborn A, Verdu EF, Bercik P, Vanner S. Nutritional profile of rodent diets impacts experimental reproducibility in microbiome preclinical research. Sci Rep 2020; 10:17784. [PMID: 33082369 PMCID: PMC7575541 DOI: 10.1038/s41598-020-74460-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
The lack of reproducibility of animal experimental results between laboratories, particularly in studies investigating the microbiota, has raised concern among the scientific community. Factors such as environment, stress and sex have been identified as contributors, whereas dietary composition has received less attention. This study firstly evaluated the use of commercially available rodent diets across research institutions, with 28 different diets reported by 45 survey respondents. Secondly, highly variable ingredient, FODMAP (Fermentable Oligo-, Di-, Mono-saccharides And Polyols) and gluten content was found between different commercially available rodent diets. Finally, 40 mice were randomized to four groups, each receiving a different commercially available rodent diet, and the dietary impact on cecal microbiota, short- and branched-chain fatty acid profiles was evaluated. The gut microbiota composition differed significantly between diets and sexes, with significantly different clusters in β-diversity. Total BCFA were highest (p = 0.01) and SCFA were lowest (p = 0.03) in mice fed a diet lower in FODMAPs and gluten. These results suggest that nutritional composition of commercially available rodent diets impact gut microbiota profiles and fermentation patterns, with major implications for the reproducibility of results across laboratories. However, further studies are required to elucidate the specific dietary factors driving these changes.
Collapse
Affiliation(s)
- C J Tuck
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada.
- Department of Dietetics, Nutrition and Sport, La Trobe University, Bundoora, VIC, Australia.
| | - G De Palma
- Farncombe Institute, McMaster University, Hamilton, ON, Canada
| | - K Takami
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - B Brant
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - A Caminero
- Farncombe Institute, McMaster University, Hamilton, ON, Canada
| | - D E Reed
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - J G Muir
- Department of Gastroenterology, Monash University, Melbourne, Australia
| | - P R Gibson
- Department of Gastroenterology, Monash University, Melbourne, Australia
| | - A Winterborn
- Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - E F Verdu
- Farncombe Institute, McMaster University, Hamilton, ON, Canada
| | - P Bercik
- Farncombe Institute, McMaster University, Hamilton, ON, Canada
| | - S Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| |
Collapse
|
102
|
Camilleri M, Chedid V. Actionable biomarkers: the key to resolving disorders of gastrointestinal function. Gut 2020; 69:1730-1737. [PMID: 32269066 DOI: 10.1136/gutjnl-2019-320325] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/12/2020] [Accepted: 02/15/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Michael Camilleri
- Division of Gastroenterology, Mayo Clinic, Rochester, Minnesota, USA
| | - Victor Chedid
- Division of Gastroenterology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
103
|
Predisposition of Neonatal Maternal Separation to Visceral Hypersensitivity via Downregulation of Small-Conductance Calcium-Activated Potassium Channel Subtype 2 (SK2) in Mice. Neural Plast 2020; 2020:8876230. [PMID: 33029124 PMCID: PMC7528131 DOI: 10.1155/2020/8876230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 12/13/2022] Open
Abstract
Background Visceral hypersensitivity is a common occurrence of gastrointestinal diseases such as irritable bowel syndrome (IBS), wherein early-life stress (ELS) may have a high predisposition to the development of visceral hypersensitivity in adulthood, with the specific underlying mechanism still elusive. Herein, we assessed the potential effect of small-conductance calcium-activated potassium channel subtype 2 (SK2) in the spinal dorsal horn (DH) on the pathogenesis of visceral hypersensitivity induced by maternal separation (MS) in mice. Methods Neonatal mice were subjected to the MS paradigm, an established ELS model. In adulthood, the visceral pain threshold and the abdominal withdrawal reflex (AWR) were measured with an inflatable balloon. The elevated plus maze, open field test, sucrose preference test, and forced swim test were employed to evaluate the anxiety- and depression-like behaviors. The expression levels of SK2 in the spinal DH were determined by immunofluorescence and western blotting. The mRNA of SK2 and membrane palmitoylated protein 2 (MPP2) were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Electrophysiology was applied to evaluate the neuronal firing rates and SK2 channel-mediated afterhyperpolarization current (I AHP). The interaction between MPP2 and SK2 was validated by coimmunoprecipitation. Results In contrast to the naïve mice, ethological findings in MS mice revealed lowered visceral pain threshold, more evident anxiety- and depression-like behaviors, and downregulated expression of membrane SK2 protein and MPP2 protein. Moreover, electrophysiological results indicated increased neuronal firing rates and decreased I AHP in the spinal DH neurons. Nonetheless, intrathecal injection of the SK2 channel activator 1-ethyl-2-benzimidazolinone (1-EBIO) in MS mice could reverse the electrophysiological alterations and elevate the visceral pain threshold. In the naïve mice, administration of the SK2 channel blocker apamin abated I AHP and elevated spontaneous neuronal firing rates in the spinal DH neurons, reducing the visceral pain threshold. Finally, disruption of the MPP2 expression by small interfering RNA (siRNA) could amplify visceral hypersensitivity in naïve mice. Conclusions ELS-induced visceral pain and visceral hypersensitivity are associated with the underfunction of SK2 channels in the spinal DH.
Collapse
|
104
|
Lin B, Wang Y, Zhang P, Yuan Y, Zhang Y, Chen G. Gut microbiota regulates neuropathic pain: potential mechanisms and therapeutic strategy. J Headache Pain 2020; 21:103. [PMID: 32807072 PMCID: PMC7433133 DOI: 10.1186/s10194-020-01170-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/10/2020] [Indexed: 02/08/2023] Open
Abstract
Neuropathic pain (NP) is a sustained and nonreversible condition characterized by long-term devastating physical and psychological damage. Therefore, it is urgent to identify an effective treatment for NP. Unfortunately, the precise pathogenesis of NP has not been elucidated. Currently, the microbiota-gut-brain axis has drawn increasing attention, and the emerging role of gut microbiota is investigated in numerous diseases including NP. Gut microbiota is considered as a pivotal regulator in immune, neural, endocrine, and metabolic signaling pathways, which participates in forming a complex network to affect the development of NP directly or indirectly. In this review, we conclude the current understanding of preclinical and clinical findings regarding the role of gut microbiota in NP and provide a novel therapeutic method for pain relief by medication and dietary interventions.
Collapse
Affiliation(s)
- Binbin Lin
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qingchun East Road, Zhejiang, 310016, Hangzhou, China
| | - Yuting Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qingchun East Road, Zhejiang, 310016, Hangzhou, China
| | - Piao Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qingchun East Road, Zhejiang, 310016, Hangzhou, China
| | - Yanyan Yuan
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qingchun East Road, Zhejiang, 310016, Hangzhou, China
| | - Ying Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qingchun East Road, Zhejiang, 310016, Hangzhou, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qingchun East Road, Zhejiang, 310016, Hangzhou, China.
| |
Collapse
|
105
|
Yoshida N, Yamashita T, Kishino S, Watanabe H, Sasaki K, Sasaki D, Tabata T, Sugiyama Y, Kitamura N, Saito Y, Emoto T, Hayashi T, Takahashi T, Shinohara M, Osawa R, Kondo A, Yamada T, Ogawa J, Hirata KI. A possible beneficial effect of Bacteroides on faecal lipopolysaccharide activity and cardiovascular diseases. Sci Rep 2020; 10:13009. [PMID: 32747669 PMCID: PMC7398928 DOI: 10.1038/s41598-020-69983-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Faecal lipopolysaccharides (LPS) have attracted attention as potent elements to explain a correlation between the gut microbiota and cardiovascular disease (CVD) progression. However, the underlying mechanism of how specific gut bacteria contribute to faecal LPS levels remains unclear. We retrospectively analysed the data of 92 patients and found that the abundance of the genus Bacteroides was significantly and negatively correlated with faecal LPS levels. The controls showed a higher abundance of Bacteroides than that in the patients with CVD. The endotoxin units of the Bacteroides LPS, as determined by the limulus amoebocyte lysate (LAL) tests, were drastically lower than those of the Escherichia coli LPS; similarly, the Bacteroides LPS induced relatively low levels of pro-inflammatory cytokine production and did not induce sepsis in mice. Fermenting patient faecal samples in a single-batch fermentation system with Bacteroides probiotics led to a significant increase in the Bacteroides abundance, suggesting that the human gut microbiota could be manipulated toward decreasing the faecal LPS levels. In the clinical perspective, Bacteroides decrease faecal LPS levels because of their reduced LAL activity; therefore, increasing Bacteroides abundance might serve as a novel therapeutic approach to prevent CVD via reducing faecal LPS levels and suppressing immune responses.
Collapse
Affiliation(s)
- Naofumi Yoshida
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 6500017, Japan
| | - Tomoya Yamashita
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 6500017, Japan.
| | - Shigenobu Kishino
- Division of Applied Life Science, Graduate School of Agriculture, Kyoto University, Kyoto, 6068502, Japan
| | - Hikaru Watanabe
- School and Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Tokyo, 1528550, Japan
| | - Kengo Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, 6578501, Japan
| | - Daisuke Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, 6578501, Japan
| | - Tokiko Tabata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 6500017, Japan
| | - Yuta Sugiyama
- Division of Applied Life Science, Graduate School of Agriculture, Kyoto University, Kyoto, 6068502, Japan
| | - Nahoko Kitamura
- Division of Applied Life Science, Graduate School of Agriculture, Kyoto University, Kyoto, 6068502, Japan
| | - Yoshihiro Saito
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 6500017, Japan
| | - Takuo Emoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 6500017, Japan
| | - Tomohiro Hayashi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 6500017, Japan
| | - Tomoya Takahashi
- School and Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Tokyo, 1528550, Japan
| | - Masakazu Shinohara
- Division of Epidemiology, Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe, 6500017, Japan
| | - Ro Osawa
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe, 6578501, Japan
| | - Akihiko Kondo
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, 6578501, Japan
| | - Takuji Yamada
- School and Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Tokyo, 1528550, Japan
| | - Jun Ogawa
- Division of Applied Life Science, Graduate School of Agriculture, Kyoto University, Kyoto, 6068502, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 6500017, Japan
| |
Collapse
|
106
|
The Effectiveness of Synbiotic Preparation Containing Lactobacillus and Bifidobacterium Probiotic Strains and Short Chain Fructooligosaccharides in Patients with Diarrhea Predominant Irritable Bowel Syndrome-A Randomized Double-Blind, Placebo-Controlled Study. Nutrients 2020; 12:nu12071999. [PMID: 32635661 PMCID: PMC7400954 DOI: 10.3390/nu12071999] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/29/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
The purpose of the randomized double-blind placebo-controlled trial was to assess the effectiveness of synbiotic preparation containing probiotic Lactobacillus rhamnosus FloraActive™ 19070-2, Lactobacillus acidophilus DSMZ 32418, Bifidobacterium lactis DSMZ 32269, Bifidobacterium longum DSMZ 32946, Bifidobacterium bifidum DSMZ 32403 and fructooligosaccharides in adult patients with diarrhea-dominant IBS (IBS-D). The study included eighty patients with moderate and severe IBS-D who were randomized to receive synbiotics or placebo for eight weeks. Finally, a total of sixty-eight patients finished the study. The primary endpoints included the assessment of the symptoms’ severity with IBS symptom severity scale (IBS-SSS), an improvement of IBS global symptoms with Global Improvement Scale (IBS-GIS) and adequate relief of symptoms after four and eight weeks of therapy. Secondary endpoints, which were collected by telephone interviewers three times a week included the assessment of individual IBS symptoms and adverse events. Synbiotic treatment in comparison to placebo significantly improved IBS-GIS (p = 0.043), and IBS-SSS score inducing a decrease in the total IBS-SSS (p = 0.042) and in domain-specific scores related to flatulence (p = 0.028) and bowel habit (p = 0.028) after four and eight weeks. Patients treated with synbiotics reported in weekly observations a significant amelioration in a feeling of incomplete bowel movements, flatulence, pain, stool pressure and diarrheal stools compared to those receiving placebo. There were no differences in adverse events between both groups. Concluding, the multi-strain synbiotic preparation was associated with a significant improvement in symptoms in IBS-D patients and was well-tolerated. These results suggest that the use of synbiotics offers a benefit for IBS-D patients. [Clinicaltrials.gov NCT04206410 registered 20 December 2019].
Collapse
|
107
|
Gibson PR, Halmos EP, Muir JG. Review article: FODMAPS, prebiotics and gut health-the FODMAP hypothesis revisited. Aliment Pharmacol Ther 2020; 52:233-246. [PMID: 32562590 DOI: 10.1111/apt.15818] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/27/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Restriction of dietary FODMAP intake can alleviate symptoms in patients with irritable bowel syndrome. Because many FODMAPs have prebiotic actions, there is concern that their dietary restriction leads to dysbiosis with health consequences, and their intake is being encouraged by addition to foods and via supplements. AIMS To examine the hazards and benefits of high and low FODMAP intake. METHODS Current literature was reviewed and alternative hypotheses formulated. RESULTS Low FODMAP intake reduces abundance of faecal Bifidobacteria without known adverse outcomes and has no effect on diversity, but the reduction in bacterial density may potentially be beneficial to gut health. Supplementary prebiotics can markedly elevate the intake of FODMAPs over levels consumed in the background diet. While this increases the abundance of Bifidobacteria, it adversely affects gut health in animal studies by inducing colonic mucosal barrier dysfunction, mucosal inflammation and visceral hypersensitivity. Rapid colonic fermentation is central to the identified mechanisms that include injury from high luminal concentrations of short-chain fatty acids and low pH, and inflammatory effects of increased endotoxin load and glycation of macromolecules. Whether these observations translate into humans requires further study. Opposing hypotheses are presented whereby excessive intake of FODMAPs might have health benefits via prebiotic effects, but might also be injurious and contribute to the apparent increase in functional intestinal disorders. CONCLUSIONS Reduced FODMAP intake has few deleterious effects on gut microbiota. Consequences (both positive and negative) of excessive carbohydrate fermentation in the human intestines from elevated FODMAP intake require more attention.
Collapse
Affiliation(s)
- Peter R Gibson
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Vic., Australia
| | - Emma P Halmos
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Vic., Australia
| | - Jane G Muir
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Vic., Australia
| |
Collapse
|
108
|
Moayyedi P, Simrén M, Bercik P. Evidence-based and mechanistic insights into exclusion diets for IBS. Nat Rev Gastroenterol Hepatol 2020; 17:406-413. [PMID: 32123377 DOI: 10.1038/s41575-020-0270-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
Abstract
Exclusion diets are becoming increasingly popular in the management of irritable bowel syndrome (IBS). Several mechanisms exist by which food items might cause gastrointestinal symptoms, such as direct osmotic effects of food in the gut lumen, changes to the gut microbiota and immune activation. These effects have been demonstrated in animal models and in human studies, particularly in the case of gluten-free diets and diets low in fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAPs). Indeed, randomized controlled trials (RCTs) suggest that gluten-free diets and low-FODMAP diets improve IBS symptoms, and guidelines recommend the latter approach for treating symptoms in some patients with IBS. Designing such RCTs is challenging as participants need to eat so an 'inert' placebo is not an option. Blinding is also an issue with these studies; in the future, new exclusion diets should not advertise what the diet consists of until it is proved to reduce symptoms. In this Review, we outline the advantages and disadvantages of each choice of control group and emphasize the importance of collecting mechanistic data (regarding direct effects of food on the gut lumen, changes in gut microbiota and intestinal inflammation) as well as symptom data in RCTs of exclusion diets in IBS.
Collapse
Affiliation(s)
- Paul Moayyedi
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.
| | - Magnus Simrén
- Department of Internal Medicine & Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
109
|
Zhao T, Zhang H, Yin X, Zhao H, Ma L, Yan M, Peng L, Wang Q, Dong X, Li P. Tangshen formula modulates gut Microbiota and reduces gut-derived toxins in diabetic nephropathy rats. Biomed Pharmacother 2020; 129:110325. [PMID: 32535383 DOI: 10.1016/j.biopha.2020.110325] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/19/2020] [Accepted: 05/23/2020] [Indexed: 12/12/2022] Open
Abstract
Growing evidence shows that diabetic kidney disease (DKD) is linked with intestinal dysbiosis from gut-derived toxins. Tangshen Formula (TSF) is a traditional Chinese herbal medicine that has been used to treat DKD. In this study, streptozotocin injection and uninephrectomy-induced diabetic nephropathy (DN) rat model was established to explore the impact of TSF on gut microbiota composition, gut-derived toxins, and the downstream inflammatory pathway of urotoxins in the kidney. TSF treatment for 12 weeks showed significant attenuation of both renal histologic injuries and urinary excretion of albumin compared with DN rats without treatment. TSF treatment also reconstructed gut dysbiosis and reduced levels of indoxyl sulfate and metabolic endotoxemia/lipopolysaccharide. MCP-1 and TNF-α were decreased by TSF both in the serum and kidney. In addition, we revealed that the inhibitory effect of TSF on renal inflammation was associated with the inhibition of aryl hydrocarbon, a receptor of indoxyl sulfate, and TLR4, thereby inhibiting JNK and NF-κB signaling in the kidney. Spearman correlation analysis found that a cluster of gut bacterial phyla and genera were significantly correlated with renal pathology, renal function, and systemic inflammation. In conclusion, orally administered TSF significantly inhibited diabetic renal injury, and modulated gut microbiota, which decreased levels of lipopolysaccharide and indoxyl sulfate, and attenuated renal inflammation. Our results indicate that TSF may be used as an agent in the prevention of gut dysbiosis and elimination of intestinal toxins in DN individuals.
Collapse
Affiliation(s)
- TingTing Zhao
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Department of Pharmacology, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - HaoJun Zhang
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Department of Pharmacology, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Xingbin Yin
- Beijing University of Chinese Medicine, No. 11 Beisanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - HaiLing Zhao
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Department of Pharmacology, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Liang Ma
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Department of Pharmacology, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - MeiHua Yan
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Department of Pharmacology, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Liang Peng
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Department of Pharmacology, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Qian Wang
- Beijing University of Chinese Medicine, No. 11 Beisanhuan Donglu, Chaoyang District, Beijing 100029, China
| | - Xi Dong
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Department of Pharmacology, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Department of Pharmacology, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, PR China.
| |
Collapse
|
110
|
Grabauskas G, Wu X, Gao J, Li JY, Turgeon DK, Owyang C. Prostaglandin E 2, Produced by Mast Cells in Colon Tissues From Patients With Irritable Bowel Syndrome, Contributes to Visceral Hypersensitivity in Mice. Gastroenterology 2020; 158:2195-2207.e6. [PMID: 32084424 PMCID: PMC7282934 DOI: 10.1053/j.gastro.2020.02.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 01/31/2020] [Accepted: 02/06/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Visceral hypersensitivity is common in patients with irritable bowel syndrome (IBS). We investigated whether inflammatory molecules, such as histamine and proteases, activate prostaglandin-endoperoxide synthase 2 (also called COX2) to increase the synthesis of prostaglandin E2 (PGE2) by mast cells, which activates the receptor PTGER2 (also called EP2) in the dorsal root ganglia to promote visceral hypersensitivity. METHODS We used an enzyme-linked immunosorbent assay to measure levels of spontaneous release of molecules from mast cells in colonic mucosa from patients with IBS with diarrhea (IBS-D; 18 women and 5 men; aged 28-60 years), healthy individuals (controls, n = 24), mice, and rats. We measured visceromotor responses to colorectal distension in rodents after intracolonic administration of colon biopsy supernatants, histamine, PGE2, a small interfering RNA against EP2, or an agonist of F2R like trypsin receptor 1 (F2RL1, also called protease-activated receptor 2 [PAR2]). We investigated the role of COX2, produced by mast cells, in mediation of visceral hypersensitivity using mice with the Y385F substitution in Ptgs2 (Ptgs2Y385F mice), mast cell-deficient (W/WV) mice, and W/WV mice given injections of mast cells derived from wild-type or Ptgs2Y385F mice. RESULTS Colon biopsies from patients with IBS-D had increased levels of PGE2, based on enzyme-linked immunosorbent assay, and COX2 messenger RNA and protein, compared with control biopsies. Immunohistochemistry showed that most of the COX2 was in mast cells. Intracolonic infusions of rats with IBS-D biopsy supernatants generated a 3- to 4-fold increase in visceromotor responses to colorectal distension; this was associated with significant increases in PGE2, histamine, and tryptase in the colonic mucosa. These increases were prevented by a mast cell stabilizer, COX2 inhibitor, or knockdown of EP2. Intracolonic administration of supernatants from biopsies of patients with IBS-D failed to induce visceral hypersensitivity or increase the level of PGE2 in W/WV and Ptgs2Y385Fmice. Reconstitution of mast cells in W/WV mice restored the visceral hypersensitivity response. CONCLUSIONS Abnormal synthesis of PGE2 by colonic mast cells appears to induce visceral hypersensitivity in patients with IBS-D.
Collapse
Affiliation(s)
- Gintautas Grabauskas
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Xiaoyin Wu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jun Gao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Ji-Yao Li
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Danielle Kim Turgeon
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Chung Owyang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
111
|
van Thiel IAM, de Jonge WJ, Chiu IM, van den Wijngaard RM. Microbiota-neuroimmune cross talk in stress-induced visceral hypersensitivity of the bowel. Am J Physiol Gastrointest Liver Physiol 2020; 318:G1034-G1041. [PMID: 32308040 PMCID: PMC7642838 DOI: 10.1152/ajpgi.00196.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Visceral hypersensitivity of the lower gastrointestinal tract, defined as an increased response to colorectal distension, frequently prompts episodes of debilitating abdominal pain in irritable bowel syndrome (IBS). Although the pathophysiology of IBS is not yet fully elucidated, it is well known that stress is a major risk factor for development and acts as a trigger of pain sensation. Stress modulates both immune responses as well as the gut microbiota and vice versa. Additionally, either microbes themselves or through involvement of the immune system, activate or sensitize afferent nociceptors. In this paper, we review current knowledge on the influence of stress along the gut-brain-microbiota axis and exemplify relevant neuroimmune cross talk mechanisms in visceral hypersensitivity, working toward understanding how gut microbiota-neuroimmune cross talk contributes to visceral pain sensation in IBS patients.
Collapse
Affiliation(s)
- Isabelle A. M. van Thiel
- 1Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,2Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Wouter J. de Jonge
- 1Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,2Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands,3Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands,4Department of General, Visceral, Thoracic, and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Isaac M. Chiu
- 5Department of Immunology, Harvard Medical School. Boston, Massachusetts
| | - Rene M. van den Wijngaard
- 1Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,2Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands,3Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
112
|
Abstract
Irritable bowel syndrome (IBS) is a chronic disorder characterised by recurrent abdominal pain or discomfort and transit disturbances with heterogeneous pathophysiological mechanisms. The link between food and gastrointestinal (GI) symptoms is often reported by patients with IBS and the role of fructose has recently been highlighted. Fructose malabsorption can easily be assessed by hydrogen and/or methane breath test in response to 25 g fructose; and its prevalence is about 22 % in patients with IBS. The mechanism of fructose-related symptoms is incompletely understood. Osmotic load, fermentation and visceral hypersensitivity are likely to participate in GI symptoms in the IBS population and may be triggered or worsened by fructose. A low-fructose diet could be integrated in the overall treatment strategy, but its role and implication in the improvement of IBS symptoms should be evaluated. In the present review, we discuss fructose malabsorption in adult patients with IBS and the interest of a low-fructose diet in order to underline the important role of fructose in IBS.
Collapse
|
113
|
Li M, Li S, Zhou H, Tang X, Wu Y, Jiang W, Tian Z, Zhou X, Yang X, Wang Y. Chemotaxis-driven delivery of nano-pathogenoids for complete eradication of tumors post-phototherapy. Nat Commun 2020; 11:1126. [PMID: 32111847 PMCID: PMC7048836 DOI: 10.1038/s41467-020-14963-0] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
The efficacy of nano-mediated drug delivery has been impeded by multiple biological barriers such as the mononuclear phagocyte system (MPS), as well as vascular and interstitial barriers. To overcome the abovementioned obstacles, we report a nano-pathogenoid (NPN) system that can in situ hitchhike circulating neutrophils and supplement photothermal therapy (PTT). Cloaked with bacteria-secreted outer membrane vesicles inheriting pathogen-associated molecular patterns of native bacteria, NPNs are effectively recognized and internalized by neutrophils. The neutrophils migrate towards inflamed tumors, extravasate across the blood vessels, and penetrate through the tumors. Then NPNs are rapidly released from neutrophils in response to inflammatory stimuli and subsequently taken up by tumor cells to exert anticancer effects. Strikingly, due to the excellent targeting efficacy, cisplatin-loaded NPNs combined with PTT completely eradicate tumors in all treated mice. Such a nano-platform represents an efficient and generalizable strategy towards in situ cell hitchhiking as well as enhanced tumor targeted delivery. The presence of several biological barriers impede the efficacy of nano-mediated drug delivery for solid cancer therapy. Here, the authors develop a nano-pathogenoid system that targets circulating neutrophils and show that it overcomes these biological barriers and improves tumour targeting and efficacy.
Collapse
Affiliation(s)
- Min Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China.,Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, Anhui, China.,College of Chemistry and Environmental Engineering, Shenzhen University, 518060, Shenzhen, Guangdong, China
| | - Shuya Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Han Zhou
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xinfeng Tang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Yi Wu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Wei Jiang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Xuechang Zhou
- College of Chemistry and Environmental Engineering, Shenzhen University, 518060, Shenzhen, Guangdong, China
| | - Xianzhu Yang
- Institutes for Life Sciences, School of Medicine and National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, 510006, Guangzhou, China
| | - Yucai Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, Anhui, China. .,Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, Anhui, China.
| |
Collapse
|
114
|
Milajerdi A, Sadeghi O, Siadat SD, Keshavarz SA, Sima A, Vahedi H, Adibi P, Esmaillzadeh A. A randomized controlled trial investigating the effect of a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols on the intestinal microbiome and inflammation in patients with ulcerative colitis: study protocol for a randomized controlled trial. Trials 2020; 21:201. [PMID: 32070388 PMCID: PMC7029518 DOI: 10.1186/s13063-020-4108-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/26/2020] [Indexed: 12/12/2022] Open
Abstract
Background No conclusive treatment is available for irritable bowel disease (IBD). Adherence to a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) might alleviate clinical symptoms of IBD. However, no study has investigated the effect of low FODMAPs diet on the intestinal microbiota and inflammatory biomarkers in patients with IBD. The aim of current study is to examine the effect a low FODMAP diet on IBD symptoms, inflammation, and the intestinal microbiota in patients with ulcerative colitis. Methods and analysis This study is a randomized clinical trial. Thirty patients with mild to moderate ulcerative colitis will be randomly allocated to receive a low FODMAP diet (n = 15) or to continue their usual diet as control (n = 15), for 4 weeks. The quantity of intestinal microbiota including Clostridium cluster IV, Faecalibacterium prausnitzii, Rosburia spp., Lactobacillus spp., Bifidobacteria spp., Akkermansia muciniphila, Bacteroides fragilis, and Ruminococcus spp., and the Firmicutes to Bacteroidetes ratio and calprotectin and lactoferrin levels will be explored in fecal samples from patients. In addition, anthropometric measures and biochemical assessments including serum concentrations of highly sensitive-C reactive protein (hs-CRP), tumour necrosis factor-α (TNF-α) and IL-1β will be taken from patients at baseline and end of the study. The study has been registered in IRCT (IRCT20181126041763N1; registration date: 2019-01-18). Discussion Consumption of a low-FODMAP diet might decrease systemic and intestinal inflammation, change the bacterial population in the gut, and modulate clinical symptoms in patients with ulcerative colitis. Further studies investigating the effect of such a diet on other variables, including other bacterial species and inflammatory cytokines, are required to confirm future findings of this trial.
Collapse
Affiliation(s)
- Alireza Milajerdi
- Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, PO Box 81745, Tehran, Iran
| | - Omid Sadeghi
- Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, PO Box 81745, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Ali Keshavarz
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Sima
- Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Homayoon Vahedi
- Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Peyman Adibi
- Integrative Functional Gastroenterology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Esmaillzadeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, PO Box 81745, Tehran, Iran. .,Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
115
|
Holma R, Laatikainen R, Orell H, Joensuu H, Peuhkuri K, Poussa T, Korpela R, Österlund P. Consumption of Lactose, Other FODMAPs and Diarrhoea during Adjuvant 5-Fluorouracil Chemotherapy for Colorectal Cancer. Nutrients 2020; 12:nu12020407. [PMID: 32033152 PMCID: PMC7071323 DOI: 10.3390/nu12020407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy-induced mucosal injury of the small intestine may interfere with the enzymes and transporters responsible for the hydrolysis and absorption of dietary carbohydrates causing diarrhoea, abdominal discomfort and pain. The aim of this study was to investigate the association between the consumption of foods rich in FODMAPs (fermentable oligo-, di- and monosaccharides and polyols) and gastrointestinal symptoms in patients receiving adjuvant therapy for colorectal cancer. The patients (n = 52) filled in a 4-day food diary at baseline and during therapy and kept a symptom diary. The intakes of FODMAP-rich foods were calculated as portions and the intakes were divided into two consumption categories. Patients with high consumption of FODMAP-rich foods had diarrhoea more frequently than those with low consumption (for lactose-rich foods the odds ratio (OR) was 2.63, P = 0.03; and for other FODMAP-rich foods 1.82, P = 0.20). Patients with high consumption of both lactose-rich and other FODMAP-rich foods had an over 4-fold risk of developing diarrhoea as compared to those with low consumption of both (OR, 4.18; P = 0.02). These results were confirmed in multivariate models. Conclusion: Consumption of lactose-rich foods results in an increased risk of diarrhoea during adjuvant therapy for colorectal cancer, especially when the consumption of other FODMAP-rich foods is also high.
Collapse
Affiliation(s)
- Reetta Holma
- Faculty of Medicine, Pharmacology, Medical Nutrition Physiology and Human Microbe Research Program, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland; (R.H.); (R.L.); (R.K.)
| | - Reijo Laatikainen
- Faculty of Medicine, Pharmacology, Medical Nutrition Physiology and Human Microbe Research Program, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland; (R.H.); (R.L.); (R.K.)
- Booston Oy Ltd., Viikinkaari 6, FI-00790 Helsinki, Finland
| | - Helena Orell
- Department of Clinical Nutrition Therapy, Helsinki University Hospital, P.O. Box 100, FI-00029 HUS, Finland;
| | - Heikki Joensuu
- Department of Oncology, Helsinki University Hospital and University of Helsinki, P.O. Box 180, FI-00029 HUS, Finland;
| | - Katri Peuhkuri
- Social Services and Health Care Division, P.O. Box 6230, FI-00099 City of Helsinki, Finland;
| | - Tuija Poussa
- STAT-Consulting, Vahverokatu 6, FI-37130 Nokia, Finland;
| | - Riitta Korpela
- Faculty of Medicine, Pharmacology, Medical Nutrition Physiology and Human Microbe Research Program, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland; (R.H.); (R.L.); (R.K.)
| | - Pia Österlund
- Department of Oncology, Helsinki University Hospital and University of Helsinki, P.O. Box 180, FI-00029 HUS, Finland;
- Department of Oncology, Tampere University Hospital and Tampere university, P.O. Box 2000, FI-33521 Tampere, Finland
- Correspondence: or or ; Tel.: +358-9-4711
| |
Collapse
|
116
|
Kamphuis JBJ, Guiard B, Leveque M, Olier M, Jouanin I, Yvon S, Tondereau V, Rivière P, Guéraud F, Chevolleau S, Noguer-Meireles MH, Martin JF, Debrauwer L, Eutamène H, Theodorou V. Lactose and Fructo-oligosaccharides Increase Visceral Sensitivity in Mice via Glycation Processes, Increasing Mast Cell Density in Colonic Mucosa. Gastroenterology 2020; 158:652-663.e6. [PMID: 31711923 DOI: 10.1053/j.gastro.2019.10.037] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 10/28/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Irritable bowel syndrome (IBS) is characterized by abdominal pain, bloating, and erratic bowel habits. A diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) can reduce symptoms of IBS, possibly by reducing microbial fermentation products. We investigated whether ingestion of FODMAPs can induce IBS-like visceral hypersensitivity mediated by fermentation products of intestinal microbes in mice. METHODS C57Bl/6 mice were gavaged with lactose, with or without the antiglycation agent pyridoxamine, or saline (controls) daily for 3 weeks. A separate group of mice were fed a diet containing fructo-oligosaccharides, with or without pyridoxamine in drinking water, or a normal chow diet (controls) for 6 weeks. Feces were collected and analyzed by 16S ribosomal RNA gene sequencing and bacterial community analyses. Abdominal sensitivity was measured by electromyography and mechanical von Frey filament assays. Colon tissues were collected from some mice and analyzed by histology and immunofluorescence to quantify mast cells and expression of advanced glycosylation end-product specific receptor (AGER). RESULTS Mice gavaged with lactose or fed fructo-oligosaccharides had increased abdominal sensitivity compared with controls, associated with increased numbers of mast cells in colon and expression of the receptor for AGER in proximal colon epithelium. These effects were prevented by administration of pyridoxamine. Lactose and/or pyridoxamine did not induce significant alterations in the composition of the fecal microbiota. Mass spectrometric analysis of carbonyl compounds in fecal samples identified signatures associated with mice given lactose or fructo-oligosaccharides vs controls. CONCLUSIONS We found that oral administration of lactose or fructo-oligosaccharides to mice increases abdominal sensitivity, associated with increased numbers of mast cells in colon and expression of AGER; these can be prevented with an antiglycation agent. Lactose and/or pyridoxamine did not produce alterations in fecal microbiota of mice. Our findings indicate that preventing glycation reactions might reduce abdominal pain in patients with IBS with sensitivity to FODMAPs.
Collapse
Affiliation(s)
- Jasper B J Kamphuis
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Bruno Guiard
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Mathilde Leveque
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Maiwenn Olier
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Isabelle Jouanin
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Sophie Yvon
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Valerie Tondereau
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Pauline Rivière
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Françoise Guéraud
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Prevention and Promotion of Carcinogenesis by Food team, Toxalim, Toulouse, France
| | - Sylvie Chevolleau
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Maria-Helena Noguer-Meireles
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Jean-François Martin
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Laurent Debrauwer
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Helene Eutamène
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse.
| | - Vassilia Theodorou
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| |
Collapse
|
117
|
Chen Y, Wu G, Zhao Y. Gut Microbiota and Alimentary Tract Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1238:11-22. [PMID: 32323177 DOI: 10.1007/978-981-15-2385-4_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gastrointestinal (GI) tract is inhabited by a diverse array of microbes, which play crucial roles in health and disease. Dysbiosis of microbiota has been tightly linked to gastrointestinal inflammatory and malignant diseases. Here we highlight the role of Helicobacter pylori alongside gastric microbiota associated with gastric inflammation and cancer. We summarize the taxonomic and functional aspects of intestinal microbiota linked to inflammatory bowel diseases (IBD), irritable bowel syndrome (IBS), and colorectal cancer in clinical investigations. We also discuss microbiome-related animal models. Nevertheless, there are tremendous opportunities to reveal the causality of microbiota in health and disease and detailed microbe-host interaction mechanisms by which how dysbiosis is causally linked to inflammatory disease and cancer, in turn, potentializing clinical interventions with a personalized high efficacy.
Collapse
Affiliation(s)
- Ye Chen
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangyan Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yongzhong Zhao
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
118
|
Heenan PE, Keenan JI, Bayer S, Simon M, Gearry RB. Irritable bowel syndrome and the gut microbiota. J R Soc N Z 2019. [DOI: 10.1080/03036758.2019.1695635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Phoebe E. Heenan
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | | | - Simone Bayer
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Myrthe Simon
- Department of Medicine, Vrije Universiteit, Amsterdam, Netherlands
| | - Richard B. Gearry
- Department of Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
119
|
Pensabene L, Salvatore S, Turco R, Tarsitano F, Concolino D, Baldassarre ME, Borrelli O, Thapar N, Vandenplas Y, Staiano A, Saps M. Low FODMAPs diet for functional abdominal pain disorders in children: critical review of current knowledge. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2019. [DOI: 10.1016/j.jpedp.2019.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
120
|
Pensabene L, Salvatore S, Turco R, Tarsitano F, Concolino D, Baldassarre ME, Borrelli O, Thapar N, Vandenplas Y, Staiano A, Saps M. Low FODMAPs diet for functional abdominal pain disorders in children: critical review of current knowledge. J Pediatr (Rio J) 2019; 95:642-656. [PMID: 31028745 DOI: 10.1016/j.jped.2019.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE This narrative review aimed to provide practitioners a synthesis of the current knowledge on the role of a low Fermentable Oligosaccharides Disaccharides Monosaccharides and Polyols diet in reducing symptoms associated with functional abdominal pain disorders in children. This review is focused on the pathophysiology, efficacy and criticism of low Fermentable Oligosaccharides Disaccharides Monosaccharides and Polyols diet in children. SOURCES Cochrane Database, Pubmed and Embase were searched using specific terms for Fermentable Oligosaccharides Disaccharides Monosaccharides and Polyols diet interventions and functional abdominal pain disorders. SUMMARY OF THE FINDINGS In children, only one Randomized Control Trial and one open-label study reported positive results of low Fermentable Oligosaccharides Disaccharides Monosaccharides and Polyols diet; one Randomized Control Trial showed exacerbation of symptoms with fructans in children with Irritable Bowel Syndrome; no effect was found for the lactose-free diet whilst fructose-restricted diets were effective in 5/6 studies. CONCLUSIONS In children there are few trials evaluating low Fermentable Oligosaccharides Disaccharides Monosaccharides and Polyols in functional abdominal pain disorders, with encouraging data on the therapeutic efficacy particularly of fructose-restricted diet. Additional efforts are still needed to fill this research gap and clarify the most efficient way for tailoring dietary restrictions based on the patient's tolerance and/or identification of potential biomarkers of low Fermentable Oligosaccharides Disaccharides Monosaccharides and Polyols efficacy, to maintain nutritional adequacy and to simplify the adherence to diet by labeling Fermentable Oligosaccharides Disaccharides Monosaccharides and Polyols content in commercial products.
Collapse
Affiliation(s)
- Licia Pensabene
- University "Magna Graecia" of Catanzaro, Pediatric Unit, Department of Medical and Surgical Sciences, Catanzaro, Italy.
| | - Silvia Salvatore
- University of Insubria, Section of Pediatrics, Department of Medicine and Surgery, Varese, Italy
| | - Rossella Turco
- University of Naples "Federico II", Section of Pediatrics, Department of Translational Medical Science, Naples, Italy
| | - Flora Tarsitano
- University "Magna Graecia" of Catanzaro, Pediatric Unit, Department of Medical and Surgical Sciences, Catanzaro, Italy
| | - Daniela Concolino
- University "Magna Graecia" of Catanzaro, Pediatric Unit, Department of Medical and Surgical Sciences, Catanzaro, Italy
| | | | - Osvaldo Borrelli
- Great Ormond Street Hospital for Children, Neurogastroenterology and Motility Unit, Department of Gastroenterology, London, United Kingdom
| | - Nikhil Thapar
- Great Ormond Street Hospital for Children, Neurogastroenterology and Motility Unit, Department of Gastroenterology, London, United Kingdom
| | - Yvan Vandenplas
- Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, KidZ Health Castle, Brussels, Belgium
| | - Annamaria Staiano
- University of Naples "Federico II", Section of Pediatrics, Department of Translational Medical Science, Naples, Italy
| | - Miguel Saps
- University of Miami, Holtz Children's Hospital, Miller School of Medicine, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Miami, United States
| |
Collapse
|
121
|
[Diagnosis and treatment of interstitial cystitis (IC/PBS) : S2k guideline of the German Society of Urology]. Urologe A 2019. [PMID: 31659368 DOI: 10.1007/s00120-019-01054-2.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
In this review article, the authors describe all relevant aspects of the new S2k guideline from the German Society of Urology (Deutschen Gesellschaft für Urologie, DGU) for the diagnosis and treatment of IC/PBS (interstitial cystitis/painful bladder syndrome). A list of necessary and optional examinations and the necessity of diagnosis of exclusion are summarized and evaluated. The treatment options listed (ranging from conservative, oral drug, and complementary medicine to interventional surgical procedures) also give the reader a good overview of the contents of the guideline and possible therapeutic approaches. Finally, the recommendations including consensus of the guideline group are also summarized in various information boxes.
Collapse
|
122
|
[Diagnosis and treatment of interstitial cystitis (IC/PBS) : S2k guideline of the German Society of Urology]. Urologe A 2019; 58:1313-1323. [PMID: 31659368 DOI: 10.1007/s00120-019-01054-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In this review article, the authors describe all relevant aspects of the new S2k guideline from the German Society of Urology (Deutschen Gesellschaft für Urologie, DGU) for the diagnosis and treatment of IC/PBS (interstitial cystitis/painful bladder syndrome). A list of necessary and optional examinations and the necessity of diagnosis of exclusion are summarized and evaluated. The treatment options listed (ranging from conservative, oral drug, and complementary medicine to interventional surgical procedures) also give the reader a good overview of the contents of the guideline and possible therapeutic approaches. Finally, the recommendations including consensus of the guideline group are also summarized in various information boxes.
Collapse
|
123
|
Pain regulation by gut microbiota: molecular mechanisms and therapeutic potential. Br J Anaesth 2019; 123:637-654. [PMID: 31551115 DOI: 10.1016/j.bja.2019.07.026] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
The relationship between gut microbiota and neurological diseases, including chronic pain, has received increasing attention. The gut microbiome is a crucial modulator of visceral pain, whereas recent evidence suggests that gut microbiota may also play a critical role in many other types of chronic pain, including inflammatory pain, headache, neuropathic pain, and opioid tolerance. We present a narrative review of the current understanding on the role of gut microbiota in pain regulation and discuss the possibility of targeting gut microbiota for the management of chronic pain. Numerous signalling molecules derived from gut microbiota, such as by-products of microbiota, metabolites, neurotransmitters, and neuromodulators, act on their receptors and remarkably regulate the peripheral and central sensitisation, which in turn mediate the development of chronic pain. Gut microbiota-derived mediators serve as critical modulators for the induction of peripheral sensitisation, directly or indirectly regulating the excitability of primary nociceptive neurones. In the central nervous system, gut microbiota-derived mediators may regulate neuroinflammation, which involves the activation of cells in the blood-brain barrier, microglia, and infiltrating immune cells, to modulate induction and maintenance of central sensitisation. Thus, we propose that gut microbiota regulates pain in the peripheral and central nervous system, and targeting gut microbiota by diet and pharmabiotic intervention may represent a new therapeutic strategy for the management of chronic pain.
Collapse
|
124
|
Lomax AE, Pradhananga S, Sessenwein JL, O'Malley D. Bacterial modulation of visceral sensation: mediators and mechanisms. Am J Physiol Gastrointest Liver Physiol 2019; 317:G363-G372. [PMID: 31290688 DOI: 10.1152/ajpgi.00052.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The potential role of the intestinal microbiota in modulating visceral pain has received increasing attention during recent years. This has led to the identification of signaling pathways that have been implicated in communication between gut bacteria and peripheral pain pathways. In addition to the well-characterized impact of the microbiota on the immune system, which in turn affects nociceptor excitability, bacteria can modulate visceral afferent pathways by effects on enterocytes, enteroendocrine cells, and the neurons themselves. Proteases produced by bacteria, or by host cells in response to bacteria, can increase or decrease the excitability of nociceptive dorsal root ganglion (DRG) neurons depending on the receptor activated. Short-chain fatty acids generated by colonic bacteria are involved in gut-brain communication, and intracolonic short-chain fatty acids have pronociceptive effects in rodents but may be antinociceptive in humans. Gut bacteria modulate the synthesis and release of enteroendocrine cell mediators, including serotonin and glucagon-like peptide-1, which activate extrinsic afferent neurons. Deciphering the complex interactions between visceral afferent neurons and the gut microbiota may lead to the development of improved probiotic therapies for visceral pain.
Collapse
Affiliation(s)
- Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Sabindra Pradhananga
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Jessica L Sessenwein
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Dervla O'Malley
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
125
|
Wang XJ, Camilleri M, Vanner S, Tuck C. Review article: biological mechanisms for symptom causation by individual FODMAP subgroups - the case for a more personalised approach to dietary restriction. Aliment Pharmacol Ther 2019; 50:517-529. [PMID: 31309595 DOI: 10.1111/apt.15419] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/16/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Due to the paucity of targeted therapy for irritable bowel syndrome (IBS), many patients turn to dietary modifications for symptom management. The combination of five subgroups of poorly absorbed and rapidly fermented carbohydrates-fructans, galacto-oligosaccharides, lactose, excess fructose and polyols-are thought to trigger gastrointestinal symptoms and are referred to collectively as "FODMAPs". AIMS To examine the biological plausibility and mechanisms by which foods high in specific FODMAP subgroups cause symptoms, and to use this information to explore the possibility of targeting select dietary components to allow for a more personalised approach to dietary adjustment METHODS: Recent literature was analysed via search databases including Medline, PubMed and Scopus. RESULTS Lactose, fructans and galacto-oligosaccharides have strong biologic plausibility for symptom generation due to lack of hydrolases resulting in distention from osmosis and rapid fermentation. However, excess fructose and polyols may only cause symptoms in specific individuals when consumed in high doses, but this remains to be established. There is evidence to suggest that certain patient characteristics such as ethnicity may predict response to lactose, but differentiation of other subgroups is difficult prior to dietary manipulation. CONCLUSIONS While some clear mechanisms of action for symptom generation have been established, further research is needed to understand which patients will respond to specific FODMAP subgroup restriction. We suggest that clinicians consider in some patients a tailored, personalised "bottom-up" approach to the low-FODMAP diet, such as dietary restriction relevant to the patients' ethnicity, symptom profile and usual dietary intake.
Collapse
Affiliation(s)
- Xiao Jing Wang
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Camilleri
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Caroline Tuck
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
126
|
Uno Y. Hypothesis: Mechanism of irritable bowel syndrome in inflammatory bowel disease. Med Hypotheses 2019; 132:109324. [PMID: 31421429 DOI: 10.1016/j.mehy.2019.109324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/23/2019] [Indexed: 02/08/2023]
Abstract
Functional bowel symptoms can be occurred during remission from inflammatory bowel disease. In this case, a low fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet is effective for the amelioration or prevention of symptoms. However, the reason is not fully explained. This report proposes a hypothesis regarding the entire process in which inflammatory bowel disease with IBS-like symptoms (IBD-IBS) causes symptoms. A detailed process was assumed, starting from high pressure in the lumen and finally to abdominal symptoms. In this process, relationships were linked based on interactions such as ischemia, compliance, pain threshold, visceral hypersensitivity, mast cells, and permeability reported in IBD-IBS. In the process mapping, to understand the relationship between the amount of gas increased by FODMAP and ischemia, the hydrodynamic hypothesis and Ritchie's hypothesis were adapted. Ischemia in dilated intestines due to an increase in gas volume can induce excessive spasms via the mast cells and show the whole process of lowering the pain threshold. From the standpoint of the mechanism of IBD-IBS, the origin trigger may be FODMAP. Therefore, a low-FODMAP diet is recommended to relieve and prevent IBD-IBS symptoms.
Collapse
Affiliation(s)
- Yoshiharu Uno
- Office Uno Column, 419-2, Yota, Onoe-Cho, Kakogawa, Hyogo, Japan.
| |
Collapse
|
127
|
Halmos EP, Gibson PR. Controversies and reality of the FODMAP diet for patients with irritable bowel syndrome. J Gastroenterol Hepatol 2019; 34:1134-1142. [PMID: 30945376 DOI: 10.1111/jgh.14650] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/05/2019] [Indexed: 12/12/2022]
Abstract
Since its first trial showing evidence of efficacy for managing symptoms of irritable bowel syndrome, the fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet has been gaining popularity but not without criticism. Application of the diet has changed from a rigid list of "allowed" and "not allowed" foods to a structured program of initial FODMAP restriction followed by food reintroduction and finally personalization so that patients are empowered to adjust their diet themselves to achieve good predictability of symptoms. Safety concerns of the diet have centered around its initial elimination leading to compromise of nutritional and psychological health, but careful patient assessment and management, preferably through a FODMAP-trained dietitian, will reduce the risk of such negative health outcomes. Most negative attention for the FODMAP diet has been the notion that it will ruin the microbiota. Controlled studies have indicated that reducing FODMAP intake has no effects on bacterial diversity but will reduce total bacterial abundance, and higher FODMAP intakes will increase health-promoting bacteria, supporting the concept of the full FODMAP program, including attaining a minimal "maintenance" level of FODMAP restriction. This review addresses all these concerns in detail and how to overcome them, including the use of a "FODMAP-gentle" diet, describing restriction of a select few foods very concentrated in FODMAPs. This version of the diet is commonly applied in practice by experienced FODMAP-trained dietitians but is not clearly described in literature. Careful direction and assessment of response or nonresponse will decrease the risks of over-restriction and under-restriction of diet.
Collapse
Affiliation(s)
- Emma P Halmos
- Department of Gastroenterology, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Peter R Gibson
- Department of Gastroenterology, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
128
|
Panacer K, Whorwell PJ. Dietary Lectin exclusion: The next big food trend? World J Gastroenterol 2019; 25:2973-2976. [PMID: 31293334 PMCID: PMC6603809 DOI: 10.3748/wjg.v25.i24.2973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
Until recently, with the exception of coeliac disease, gastroenterologists have not been particularly interested in the role of diet in the management of gastrointestinal disorders. However, patients have always felt that diet must play a part in their symptoms and, in the absence of any medical interest, have turned to alternative dietary practitioners for help, which can often have no evidence base. Fortunately, with the advent of the FODMAP diet (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) and the realisation that diet can have a profound effect on the microbiome, medical opinion is now changing. Nevertheless, research on the various diets that are now available is often completely lacking. Lectins are carbohydrate binding proteins which are widely distributed in nature and are found in a whole variety of commonly consumed foods. It seems likely that the exclusion of lectins from the diet could become the next “food fashion” for alternative practitioners to promote, especially as there is some evidence to suggest that certain lectins may be harmful to health. It is, therefore, the purpose of this viewpoint to try and stimulate research on the dietary effects of lectins, which is currently minimal, so that we can pre-empt a situation where we are unable to give patients or the public evidence based advice on this topic.
Collapse
Affiliation(s)
- Kirpal Panacer
- University of Manchester Medical Student, Stopford Building, University of Manchester, Manchester M13 9PG, United Kingdom
| | - Peter J Whorwell
- Neurogastroenterology Unit, Wythenshawe Hospital, Manchester M23 9LT, United Kingdom
| |
Collapse
|
129
|
Gajula P, Quigley EM. Overlapping irritable bowel syndrome and inflammatory bowel disease. MINERVA GASTROENTERO 2019; 65:107-115. [PMID: 30746927 DOI: 10.23736/s1121-421x.19.02559-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of irritable bowel-type symptoms occurring in patients with inflammatory bowel disease who are in apparent remission continues to generate scientific controversy and the interpretation and management of these symptoms, so distressing to the sufferer, represent major challenges for the clinician. On the one hand, these symptoms often satisfy Rome IV criteria for IBS and their occurrence correlates highly with anxiety, a known trigger for IBS. On the other hand, recent studies have shown that many of these patients exhibit subtle inflammatory changes. These observations beg the question: are these symptoms "true" IBS superimposed on IBD, or an active but subclinical form of IBD? While it is certain that earlier studies failed to detect subclinical inflammation, it is also evident that even with the use of sensitive biomarkers for inflammation, such as calprotectin and lactoferrin backed up by pan-endoscopy and biopsy to exclude ongoing inflammatory activity in its most subtle form, the prevalence of IBS-type symptoms remains higher than expected in the IBD patient. Pending further definition of its etiology and pathology, we coined the term irritable inflammatory bowel syndrome (IIBS) to refer to this phenomenon. Here we explore the risk factors for this entity, sift through clues to its pathogenesis and attempt to provide, albeit bereft of a robust evidence base, an approach to its management.
Collapse
Affiliation(s)
- Prianka Gajula
- Department of Medicine, Houston Methodist Hospital and Weill Cornell Medical College, Houston, TX, USA
| | - Eamonn M Quigley
- Division of Gastroenterology and Hepatology, Lynda K. and David M. Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, TX, USA -
| |
Collapse
|
130
|
Ooi SL, Correa D, Pak SC. Probiotics, prebiotics, and low FODMAP diet for irritable bowel syndrome - What is the current evidence? Complement Ther Med 2019; 43:73-80. [PMID: 30935559 DOI: 10.1016/j.ctim.2019.01.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/15/2019] [Indexed: 11/26/2022] Open
Abstract
Irritable bowel syndrome (IBS) is one of the most common functional gastrointestinal disorders worldwide. While the pathogenesis is not clearly understood, current research points to the role of the gut microbiome and alterations in the diversity of the microbiota. Probiotics, prebiotics, and low FODMAP diet are therapeutic means associated with modification of the gut microbiome for the alleviation of IBS symptoms. This narrative review assesses the current evidence on the efficacy of these treatment options based on findings from recent systematic reviews and meta-analyses published from October 2013 to October 2018. There is a general agreement in the 11 included systematic reviews and meta-analyses that probiotic therapy is safe and can be effective in improving overall IBS symptom scores and abdominal pain in the general IBS population. Nonetheless, conflicting findings remain and no recommendation on the specific species/strains or combination can be made. Short-term restriction of FODMAP in the diet can improve IBS symptoms as per the findings of 7 systematic reviews and meta-analyses, even though the quality of the evidence remains questionable. Inappropriate use of the low FODMAP diet can potentially impact health negatively. As such, a low FODMAP diet is only recommended as a second line treatment guided by qualified clinicians with specialized training. Despite preclinical studies of some prebiotics demonstrated the potential use in improving gut microbiome and intestinal inflammatory response, the beneficial effect of prebiotics for IBS remains theoretical. Two systematic reviews found no evidence to support the clinical use of prebiotics for IBS.
Collapse
Affiliation(s)
- Soo Liang Ooi
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia
| | - Dianne Correa
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia
| | - Sok Cheon Pak
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia.
| |
Collapse
|
131
|
Kortlever TL, Ten Bokkel Huinink S, Offereins M, Hebblethwaite C, O'Brien L, Leeper J, Mulder CJJ, Barrett JS, Gearry RB. Low-FODMAP Diet Is Associated With Improved Quality of Life in IBS Patients-A Prospective Observational Study. Nutr Clin Pract 2019; 34:623-630. [PMID: 30644587 DOI: 10.1002/ncp.10233] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The low fermentable oligosaccharide, disaccharide, monosaccharide, and polyol (FODMAP) diet is effectively manages irritable bowel syndrome (IBS) symptoms. Long-term low-FODMAP studies rarely report quality of life (QoL). We aimed to determine the effect of low-FODMAP diet on long-term QoL, gastrointestinal (GI) and non-GI symptoms in IBS patients. METHODS A prospective observational study of IBS patients referred for low-FODMAP dietary advice was performed. The primary outcome of QoL and secondary outcomes of GI symptoms, anxiety/depression, fatigue, sleep quality, and happiness were obtained at baseline, 6 weeks (T6), and 6 months (T26). RESULTS 111 patients were recruited. 91.0%, 71.6%, and 50.5% of participants completed baseline, T6, and T26 assessments, respectively. There were significant improvements in QoL from baseline at T6 and T26 (both P < 0.001). Significant reductions were seen in GI symptoms at T6 and T26 (both P < 0.001), fatigue at T6 and T26 (both P < 0.003), and anxiety at T6 and T26 (both P < 0.007), compared with baseline. A significant reduction was seen for depression (P < 0.010) from baseline at T26, and a significant increase was seen for both happiness and vitality (both P < 0.04) from baseline at T26. There was a significant correlation between GI symptom response and change in QoL, anxiety, depression, and fatigue (all P < 0.034). CONCLUSION Low-FODMAP diet was associated with improved long-term QoL and GI symptoms, reduced fatigue and anxiety/depression, and increased happiness and vitality. These data support a wider range of benefits for IBS patients consuming a low-FODMAP diet.
Collapse
Affiliation(s)
- Tim L Kortlever
- Department of Medicine, University of Otago, Christchurch, New Zealand.,Department of Gastroenterology, VU Medical Centre, Amsterdam, the Netherlands
| | - Sebastiaan Ten Bokkel Huinink
- Department of Medicine, University of Otago, Christchurch, New Zealand.,Department of Gastroenterology, VU Medical Centre, Amsterdam, the Netherlands
| | - Marleen Offereins
- Department of Medicine, University of Otago, Christchurch, New Zealand.,Department of Gastroenterology, VU Medical Centre, Amsterdam, the Netherlands
| | | | | | - Julie Leeper
- Dietary Specialists Ltd, Christchurch, New Zealand
| | - Chris J J Mulder
- Department of Gastroenterology, VU Medical Centre, Amsterdam, the Netherlands
| | - Jacqueline S Barrett
- Department of Gastroenterology, Monash University, The Alfred Hospital, Melbourne, Australia
| | - Richard B Gearry
- Department of Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
132
|
Sugihara K, Morhardt TL, Kamada N. The Role of Dietary Nutrients in Inflammatory Bowel Disease. Front Immunol 2019; 9:3183. [PMID: 30697218 PMCID: PMC6340967 DOI: 10.3389/fimmu.2018.03183] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/27/2018] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disease of the gastrointestinal tract. Although the precise etiology of IBD remains incompletely understood, accumulating evidence suggests that various environmental factors, including dietary nutrients, contribute to its pathogenesis. Dietary nutrients are known to have an impact on host physiology and diseases. The interactions between dietary nutrients and intestinal immunity are complex. Dietary nutrients directly regulate the immuno-modulatory function of gut-resident immune cells. Likewise, dietary nutrients shape the composition of the gut microbiota. Therefore, a well-balanced diet is crucial for good health. In contrast, the relationships among dietary nutrients, host immunity and/or the gut microbiota may be perturbed in the context of IBD. Genetic predispositions and gut dysbiosis may affect the utilization of dietary nutrients. Moreover, the metabolism of nutrients in host cells and the gut microbiota may be altered by intestinal inflammation, thereby increasing or decreasing the demand for certain nutrients necessary for the maintenance of immune and microbial homeostasis. Herein, we review the current knowledge of the role dietary nutrients play in the development and the treatment of IBD, focusing on the interplay among dietary nutrients, the gut microbiota and host immune cells. We also discuss alterations in the nutritional metabolism of the gut microbiota and host cells in IBD that can influence the outcome of nutritional intervention. A better understanding of the diet-host-microbiota interactions may lead to new therapeutic approaches for the treatment of IBD.
Collapse
Affiliation(s)
- Kohei Sugihara
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Tina L Morhardt
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.,Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
133
|
Mitchell H, Porter J, Gibson PR, Barrett J, Garg M. Review article: implementation of a diet low in FODMAPs for patients with irritable bowel syndrome-directions for future research. Aliment Pharmacol Ther 2019; 49:124-139. [PMID: 30589971 DOI: 10.1111/apt.15079] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 10/27/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Despite the efficacy of a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) for patients with irritable bowel syndrome, many questions remain unanswered with respect to its clinical implementation. AIM To review literature to identify, synthesise, and provide direction for future research in the implementation and evaluation of the low FODMAP diet. METHODS Bibliographical searches were performed in Ovid Medline, CINAHL, Scopus and PubMed from database commencement until September 2018, with search terms focused on the population (irritable bowel syndrome) and intervention of interest (FODMAP). RESULTS Predictors of response to a low FODMAP diet remain under investigation, with preliminary data supporting faecal microbiota or faecal volatile organic compound profiling. Training of clinicians, and standards for the education of patients about the phases of a low FODMAP diet, as well as the role of Apps, require formal evaluation. There are limited data on the longer term efficacy and safety of the low FODMAP diet with respect to sustained symptom control, effect on quality of life and healthcare utilisation, nutritional adequacy, precipitation of disordered eating behaviours, effects on faecal microbiota and metabolomic markers, and subsequent translation to clinical effects. CONCLUSIONS Many gaps in implementation of the low FODMAP diet in clinical practice, as well as long-term safety and efficacy, remain for further investigation.
Collapse
Affiliation(s)
- Hannah Mitchell
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Gastroenterology, Eastern Health, Melbourne, Victoria, Australia.,Allied Health, Eastern Health, Melbourne, Victoria, Australia
| | - Judi Porter
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia.,Allied Health, Eastern Health, Melbourne, Victoria, Australia
| | - Peter R Gibson
- Department of Gastroenterology, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| | - Jacqueline Barrett
- Department of Gastroenterology, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| | - Mayur Garg
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Gastroenterology, Eastern Health, Melbourne, Victoria, Australia
| |
Collapse
|
134
|
Cao W, Wang C, Chin Y, Chen X, Gao Y, Yuan S, Xue C, Wang Y, Tang Q. DHA-phospholipids (DHA-PL) and EPA-phospholipids (EPA-PL) prevent intestinal dysfunction induced by chronic stress. Food Funct 2019; 10:277-288. [DOI: 10.1039/c8fo01404c] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
DHA-PL and EPA-PL may effectively protect mice against intestinal dysfunction under chronic stress exposure.
Collapse
Affiliation(s)
- Wanxiu Cao
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Chengcheng Wang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Yaoxian Chin
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Xin Chen
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Yuan Gao
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Shihan Yuan
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Changhu Xue
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Yuming Wang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Qingjuan Tang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| |
Collapse
|
135
|
A Retrospective Study on Dietary FODMAP Intake in Celiac Patients Following a Gluten-Free Diet. Nutrients 2018; 10:nu10111769. [PMID: 30445688 PMCID: PMC6265860 DOI: 10.3390/nu10111769] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022] Open
Abstract
Our aim was to evaluate the intake of foods containing fermentable oligo/di/mono-saccharides and polyols (FODMAP) as a possible factor that induces gastrointestinal symptoms in treated celiac disease (CD) patients. We collected seven-day weighed food records for 104 CD patients and 91 healthy volunteers. All evaluated food items were from sources with high and low content of FODMAP, which were divided into cereals and sweets, sweeteners and soft drinks, fruits, dried fruits, and vegetables. Nutrient intake was calculated using the food database of the European Institute of Oncology. The symptoms reported were assessed by a Rome IV Irritable bowel syndrome (IBS) diagnostic questionnaire and by specific questions for the evaluation of functional gastrointestinal disorders (FGIDs). The 12% of CD patients met IBS symptoms criteria as opposed to 6% of controls (p = 0.09) and 27% of patients reported FGIDs symptoms vs. 22% of healthy controls (p = 0.42). The intake by CD patients was significantly higher than healthy volunteers for: sweeteners and sugars with low content of FODMAP (p = 0.0007), fruits, dried fruits, and vegetables high in FODMAP (p = 0.003) and low in FODMAP (p = 0.04) when compared to controls. CD patients had a lower intake of cereals and sweets with a high content of FODMAP (p = 0.00001). Healthy volunteers consumed significantly higher alcoholic beverages and fats high in FODMAP (both p < 0.044). The mean daily intake of other food categories did not differ between both groups. Even though CD patients had a low intake of gluten-free cereals high in FODMAP, they still consumed a significant amount of fruits and vegetables high in FODMAP. The clinical effect of a concomitant gluten-free diet and low-FODMAP diet should be prospectively evaluated as a supportive therapy in CD patients.
Collapse
|
136
|
Han X, Lee A, Huang S, Gao J, Spence JR, Owyang C. Lactobacillus rhamnosus GG prevents epithelial barrier dysfunction induced by interferon-gamma and fecal supernatants from irritable bowel syndrome patients in human intestinal enteroids and colonoids. Gut Microbes 2018; 10:59-76. [PMID: 30040527 PMCID: PMC6363076 DOI: 10.1080/19490976.2018.1479625] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Disruption of intestinal barrier homeostasis is an important pathogenic factor in conditions such as irritable bowel syndrome (IBS). Lactobacillus rhamnosus GG (LGG) improves IBS symptoms through unclear mechanisms. Previous studies utilizing colorectal adenocarcinoma cell lines showed that LGG metabolites prevented interferon gamma (IFN-gamma) induced barrier damage but the model employed limited these findings. We aimed to interrogate the protective effects of LGG on epithelial barrier function using human intestinal epithelial cultures (enteroids and colonoids) as a more physiologic model. To investigate how LGG affects epithelial barrier function, we measured FITC-Dextran (FD4) flux across the epithelium as well as tight junction zonula occludens 1 (ZO-1) and occludin (OCLN) expression. Colonoids were incubated with fecal supernatants from IBS patients (IBS-FSN) and healthy controls in the presence or absence of LGG to examine changes in gut permeability. Enteroids incubated with IFN-gamma demonstrated a downregulation of OCLN and ZO-1 expression by 67% and 50%, respectively (p<0.05). This was accompanied by increased paracellular permeability as shown by leakage of FD4. Pretreatment of enteroids with LGG prevented these changes and normalized OCLN and ZO-1 to control levels. These actions were independent of its action against apoptosis. However, these protective effects were not seen with LGG cell wall extracts, LGG DNA, or denatured (boiled) LGG. Intriguingly, IBS-FSN injected into colonoids increased paracellular permeability, which was prevented by LGG. LGG, likely due to secreted proteins, protects against epithelial barrier dysfunction. Bacterial-derived factors to modulate gut barrier function may be a treatment option in disorders such as IBS.
Collapse
Affiliation(s)
- Xu Han
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology and Hepatology, Ann Arbor, MI, USA
| | - Allen Lee
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology and Hepatology, Ann Arbor, MI, USA
| | - Sha Huang
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology and Hepatology, Ann Arbor, MI, USA
| | - Jun Gao
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology and Hepatology, Ann Arbor, MI, USA
| | - Jason R. Spence
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology and Hepatology, Ann Arbor, MI, USA
| | - Chung Owyang
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology and Hepatology, Ann Arbor, MI, USA.,CONTACT Chung Owyang 3912 Taubman Center, 1500 E. Medical Center Dr., SPC 5362, Ann Arbor, MI 48109-5362
| |
Collapse
|
137
|
Vakil N. Dietary Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols (FODMAPs) and Gastrointestinal Disease. Nutr Clin Pract 2018; 33:468-475. [PMID: 29870082 DOI: 10.1002/ncp.10108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
FODMAP is an acronym for fermentable oligosaccharides, disaccharides, monosaccharides, and polyols. Dietary modification of FODMAPs has been shown to have significant effects on the physiology of the gastrointestinal tract and improves symptoms of abdominal pain, distention, and bloating in patients with irritable bowel syndrome. Structured withdrawal and reintroduction of FODMAPs supervised by a dietitian is the optimal practice for dietary FODMAP modification in irritable bowel syndrome. FODMAPs are present in enteral feeding formulas and may have a role in diarrhea and bloating in tube-fed patients. Emerging areas of research include the effects of dietary modification of FODMAPs on the microbiome, micronutrient absorption, and caloric intake. FODMAP dietary modification is an emerging area in other gastrointestinal disorders and is of relevance to all practicing dietitians.
Collapse
Affiliation(s)
- Nimish Vakil
- University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Aurora Health Care, Summit, Wisconsin, USA
| |
Collapse
|
138
|
IBS: High FODMAP diet induces LPS-derived intestinal inflammation and visceral hypersensitivity. Nat Rev Gastroenterol Hepatol 2018; 15:68. [PMID: 29300051 DOI: 10.1038/nrgastro.2017.187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
139
|
Harper A, Naghibi MM, Garcha D. The Role of Bacteria, Probiotics and Diet in Irritable Bowel Syndrome. Foods 2018; 7:E13. [PMID: 29373532 PMCID: PMC5848117 DOI: 10.3390/foods7020013] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome is a highly prevalent gastrointestinal disorder that threatens the quality of life of millions and poses a substantial financial burden on healthcare systems around the world. Intense research into the human microbiome has led to fascinating discoveries which directly and indirectly implicate the diversity and function of this occult organ in irritable bowel syndrome (IBS) pathophysiology. The benefit of manipulating the gastrointestinal microbiota with diet and probiotics to improve symptoms has been demonstrated in a wealth of both animal and human studies. The positive and negative mechanistic roles bacteria play in IBS will be explored and practical probiotic and dietary choices offered.
Collapse
Affiliation(s)
- Ashton Harper
- Protexin, Medical Affairs, Probiotics International Ltd., Lopen Head, Somerset TA13 5JH, UK.
| | - Malwina M Naghibi
- Protexin, Medical Affairs, Probiotics International Ltd., Lopen Head, Somerset TA13 5JH, UK.
| | - Davinder Garcha
- Protexin, Medical Affairs, Probiotics International Ltd., Lopen Head, Somerset TA13 5JH, UK.
| |
Collapse
|