Category: Health

Gut health and gut motility

Gut health and gut motility

Motikity and characterization of a bile acid inducible 7alpha-dehydroxylating operon in Clostridium hylemonae TN Article CAS PubMed Google Scholar Pohl JM, et al. Download citation. Nat Med. Gut health and gut motility

Video

I Fixed My Gut! Here's How...

Gut health and gut motility -

The ENS promotes the maintenance of macrophages through macrophage growth factor colony-stimulating factor 1. Neurons can label specific phenotypes to macrophages in the niche, and neuronal signals can signal macrophages to preserve neuronal networks in response to inflammation or infection. The mechanism by which BMP2 affects the function of steady-state or differentiated Enteric neurons remains unclear; however, the role of BMP receptor signaling in regulating microtubule stability, rapid axonal transport, synaptic growth, and stability of axons has been established [ 71 ].

Antibiotic treatment reduces the expression of BMP2, the number of MMs in neurons, and signal transduction through the BMP receptor and colony-stimulating factor 1 expression.

These changes lead to alterations in gastrointestinal motility [ 72 ]. Macrophages have M1 and M2 phenotypes, which produce inflammatory and anti-inflammatory cytokines, respectively [ 73 ]. Abdominal surgery activates M1 macrophages increaseing the expression of proinflammatory cytokines [ 74 ].

Shifting from the anti-inflammatory M2 to the proinflammatory M1 inactivates enteric neurons and enteric neural stem cells and delays intestinal transit [ 75 ].

In antibiotic-treated mice, the expression of M1 macrophage markers increased, whereas that of anti-inflammatory M2 markers decreased. An increase in M1 macrophages is related to prolonged gastrointestinal transport time [ 76 ]. In sterile mice transplanted with symbiotic bacteria, M2 macrophages migrating into the muscularis of the gastrointestinal tract may accelerate gastrointestinal motility.

Therefore, a low M2 macrophage count was associated with inhibited intestinal motility in mice. However, the underlying mechanisms remain unclear. Intestinal transplantation or major abdominal surgery can lead to the translocation of gut microbiota and the release of pro-inflammatory cytokines from M1-like macrophages.

This could result in circulating leukocyte recruitment, thus reducing smooth muscle function and impairing intestinal peristalsis by releasing nitric oxide [ 76 ].

T lymphocytes interacts with gut microbiota to regulate intestinal homeostasis, plays an important role in POI. CD4 knockout alleviates the damage to gut motility caused by abdominal surgery in mice [ 77 ]. Amphiregulin derived fromTh17 cell promotes intestinal fibrosis by activating mTOR and MEK in intestinal myofibroblasts,which may lead to POI [ 79 ].

In addition, CD8 T cells plays an important pathogenic role in gut motility disorders by disrupting intestinal neurons [ 80 ].

In a POI mouse model, gastrointestinal surgical injury resulted in the local production of the pro-inflammatory mediators IL by dendritic cells and interferon-γ by memory T helper type 1 cells, which activated macrophages to express iNOS.

The nitric oxide produced by iNOS paralyzed intestinal muscle cells, leading to POI [ 64 , 77 ]. In conclusion, gut microbiota imbalance, increases the intestinal inflammatory response, and neuron—macrophage interaction disorder may affect the homeostasis of the ENS, thereby leading to gastrointestinal dysfunction.

The 5-HT neurotransmitter plays a vital role in the functions of the ENS and gastrointestinal system, further regulating intestinal secretion and motility [ 82 ]. As an important paracrine signal molecule, 5-HT affects intestinal epithelial cell secretion and the intestinal barrier function through the G protein-coupled receptors on adjacent cells [ 83 ], directly and indirectly regulating intestinal motility.

As an intestinal neurotransmitter regulating the neurokinin receptor, substance P SP controls the intestinal motor and sensory functions and strongly promotes smooth muscle contraction [ 84 ]. Under mechanical or chemical stimulation, enterochromaffin cells ECs release 5-HT, which upregulates the release of SP in afferent nerve fibers by acting on the 5-HT3 receptor [ 83 ], and activates the neurokinin-1 receptor, increasing the SP-mediated motor response.

Using a POI model of guinea pigs, one study has shown that treatment with 5-HT4 agonists before surgery can significantly accelerate intestinal movement and improve POI [ 86 ].

At the same time, a meta-analysis found that treatment with 5-HT agonists can promote intestinal recovery post-surgery [ 87 ]. Mechanistically, activated 5-HT4 promotes intestinal movement by activating enteric cholinergic neurons and inhibiting the inflammatory reaction of the intestinal muscle layer [ 88 , 89 ].

Gut microbiota can regulate gastrointestinal peristalsis by affecting the synthesis of 5-HT [ 90 , 91 ]. In mice, spore-forming bacteria, such as Clostridium , promote the synthesis of 5-HT in colonic chromaffin cells and affect intramuscular neurons and gastrointestinal motility by upregulating 5-HT receptors on submucosal neurons and increasing 5-HT levels in the blood [ 85 ].

In germ-free mice, the levels of 5-HT in the colon and feces and the expression of tryptophan hydroxylase 1 TPH1 in the colon were reduced, and gut microbiota promoted 5-HT biosynthesis by increasing the expression of TPH1 in ECs [ 92 ].

Concurrently, data on clearance of the gut microbiota of specific pathogen-free mice using antibiotics showed that intestinal microorganisms had a sustained effect on 5-HT synthesis by regulating the function of ECs [ 93 ].

In specific probiotics EcNHT colonization experiments, colonization shortened gastrointestinal transit time, increased fecal excretion, and improved gastrointestinal motility by increasing the level of 5-HT [ 91 ]. In brief, various studies have shown that gut microbiota affects gastrointestinal motility by regulating 5-HT levels [ 91 , 94 ].

The mechanism by which gut microbiota affect postoperative intestinal motility by regulating 5-HT has attracted increasing research attention. Recent studies have shown that gut microbiota affect gastrointestinal motility by increasing the biosynthesis of 5-HT in ECs through fermentation end-products, such as SCFAs and Bas [ 94 ].

When the barrier function of the gut is impaired, the ENS may be exposed to the metabolites produced by gut microbiota affecting intestinal motility. The metabolites of gut microbiota can promote the synthesis of various neurotransmitters and regulate the secretion of signal molecules. Intestinal remodeling after abdominal surgery alters the metabolism of gut microbiota [ 65 ].

BAs exert bacteriostatic effects, and microorganisms transform primary BAs to produce secondary BAs. Liver injury caused by surgery reduces the level of BAs [ 95 ]. BAs can prevent intestinal bacterial overgrowth, maintain barrier function, promote gastrointestinal peristalsis by activating the G protein-coupled receptor TGR5 , and affect intestinal movement by affecting the mechanism of Ret signal transduction in the ENS [ 95 ].

However, BAs release nitric oxide and inhibit movement by activating TGR5 in inhibitory motor neurons. Increased BA levels upregulate the expression of NOS and TGR5 in the gastroenteric nerve plexus delaying gastric emptying [ 96 , 97 ].

This finding contradicts the conclusion that BAs promote intestinal peristalsis; thus, this issue requires further research. SCFAs can inhibit the growth of pathogens, and their metabolic activity is related to various gastrointestinal functions, such as intestinal motility and mucus secretion, through nerve and muscle stimulation [ 98 ].

A clinical study reported that probiotics could promote the production of SCFAs, especially by increasing acetate, butyrate, and propionate, thereby reducing postoperative intestinal complications and the occurrence of POI [ 99 ].

A recent study showed that butyrate plays a regulatory role in microbiota TLR-dependent sensing [ ]. Microorganisms are involved in intestinal motility by affecting the release of peptide YY andglucagon-like peptide-1 from enteroendocrine L-cells via stimulating TLRs [ ].

These secretions could enhance the propulsion of the colon, increase the contraction of colonic circular muscles, and improve gastrointestinal motility by promoting the development of cholinergic and nitrate neurons [ , ].

Butyric acid induces changes in the neural plasticity of the ENS, leading to neuro proliferative changes in intestinal myenteric and submucosal neurons and enhancement of colonic motility [ ].

Dysbiosis after surgery led to a significant decrease in butyric acid and SCFAs and an increase in the venous pressure in the intestine [ ]. All these changes may decrease intestinal motility, impair the removal of harmful bacteria, and reduce theanti-inflammatory response. Moreover, changes in gut microbiota after surgery might lead to insufficient decomposition of dietary components, such as lipids and complex polysaccharides [ ], leading to a reduction in SCFA levels and an imbalance in the ENS system, resulting in intestinal dyskinesia.

A recent study showed that the AHR signal in the intestinal nerve circuit connects gut microbiota and intestinal nerve function and plays an important role in regulating the intestinal motor function [ ]. Specific deletion of AHR neurons or overexpression of its negative feedback regulator Cytochrome P Family 1 Subfamily A Member 1 can inhibit colonic peristalsis.

In contrast, gut microbiota and their metabolites can combine with AHR to activate the immune system, enhance the intestinal epithelial barrier, and stimulate gastrointestinal peristalsis [ ].

In a control experiment involving specific pathogen-free mice and germ-free mice [ ], the expression of AHR in the colon tissues of germ-free mice and antibiotic-treated mice decreased, while the frequency of colonic transitional motor complexes decreased, and intestinal peristalsis slowed.

Depletion of microorganisms reduces the number of available AHR ligands, decreases the excitability of enteric neurons, and significantly prolongs intestinal transport time. These findings suggest that gut microbiota can induce AHR expression in colon tissues, thereby regulating movement of the intestinal nerve circuit.

In addition, metabolites produced due to tryptophan decomposition by gut microbiota are important signal molecules among microbiota and in host—microbiota crosstalk and may maintain homeostasis in the gastrointestinal system. Tryptophan metabolites can enhance the intestinal epithelial barrier function, reduce inflammation, regulate glucagon-like peptide-1 secretion, and affect gastrointestinal peristalsis.

The metabolites resulting from the bacterial decomposition of tryptophan were identified as AHR ligands, which may activate AHR and affect cytokines [ ]. Moreover, indoleacetic acid and tryptamine, produced in the metabolism of tryptophan, attenuated the response of proinflammatory cytokines in mouse macrophage cultures in an AHR-dependent manner [ ].

The effects of tryptophan metabolites on cytokines depend on the activation of AHR, and AHR signal transduction can modify the TLR-regulated response in dendritic cells [ ].

However, SCFAs might also enhance the gene induction of AHR, in which acetate, propionate, and butyrate improve the response induced by the AHR ligand. Gut microbiota and their metabolites can activate the AHR pathway in a ligand-dependent manner, subsequently regulating the differentiation of AHR to promote or control the release of anti-inflammatory factors.

However, preoperative intestinal cleaning and intraoperative gastrointestinal reconstruction of patients undergoing gastrointestinal surgery may result in changes in gut microbiota; however, whether gut microbiota could participate in postoperative intestinal motility recovery through the AHR pathway has not been determined and should be further investigated.

Surgery may increase the number of pathogenic bacteria in the intestine and decrease the proportion of beneficial bacteria, such as Lactobacillus and Bifidobacterium; administration of antibiotics before surgery reduces the abundance of gut microbiota, leading to dysbiosis [ , ].

Some probiotic strains may affect intestinal motility and secretion by altering the intraluminal environment; therefore, these strains might be beneficial to patients with postoperative intestinal motility injuries.

For instance, a study has shown that specific probiotics may help decrease the gut transit time and improve constipation-related symptoms in patients [ ]. The microbiota—gut—brain interaction and regulation of probiotics are considered new therapeutic tools for the treatment of POI [ ].

Pretreatment with probiotics before surgery increases the abundance of beneficial bacteria, promotes butyrate production, and stimulates excretion [ 23 ].

A meta-analysis study analyzed the time of the initial postoperative flatulence, the initial defecation, days of the first solid diet, incidence of abdominal distension, and incidence of postoperative intestinal obstruction and found that probiotics supplements reduced the incidence of abdominal distension RR, 0.

In another randomized, double-blind, placebo-controlled clinical study of adults with slow transit constipation [ ], supplementation of synbiotics increased stool frequency, improved stool consistency, reduced intestinal transit time, improved intestinal motility, and relieved constipation.

In a prospective, randomized controlled trial targeting patients undergoing craniotomy, oral supplementation with probiotics shortes the time of first stool and flatus [ ]. Lactobacillus rhamnosus GG promotes passing gas and the first postoperative stock of patients suffering pylorus preserving pancreaticoduodenectomy [ ].

These results indicate that probiotics can improve intestinal motility, and the nervous system may mediate the beneficial effects. Postoperative application of some probiotics may affect intestinal motility and secretion; however, the complex interactions of the different probiotics and strains with the ENS and intestinal motility, along with the specific mechanisms of action, need to be further studied.

In conclusion, preoperative administration of antibiotics, opioid anesthetics, and injury as a result of gastrointestinal surgery leads to disorders in gut microbiota and their metabolites, which can affect the neuromuscular regulation of gastrointestinal motility through the release of inflammatory cytokines or neurotransmitters or direct activation of signaling pathways in intestinal myometric neurons.

Reducing intestinal tissue damage during surgery, shortening anesthesia time, avoiding excessive mechanical bowel preparation, and subsequently reducing gastrointestinal microbiota disorders are effective ways to improve the management of POI.

The clinical application of some probiotics may provide a way to treat postoperative intestinal motility. In addition, based on neuroanatomy, neuroprotective mesenteric and intestinal tissue cutting may help improve and alleviate POI.

Chapman SJ, et al. Postoperative ileus following major colorectal surgery. Br J Surg. Article CAS PubMed Google Scholar. Scarborough JE, et al. Associations of specific postoperative complications with outcomes after elective colon resection: a procedure-targeted approach toward surgical quality improvement.

JAMA Surg. Article PubMed Google Scholar. Buscail E, Deraison C. Postoperative ileus: a pharmacological perspective. Br J Pharmacol. Baig MK, Wexner SD. Postoperative ileus: a review. Dis Colon Rectum. van Bree SH, et al. New therapeutic strategies for postoperative ileus.

Nat Rev Gastroenterol Hepatol. Barbara G, et al. The intestinal microenvironment and functional gastrointestinal disorders. Article Google Scholar.

Bienenstock J, Kunze W, Forsythe P. Microbiota and the gut-brain axis. Nutr Rev. Wagner NRF, et al. Postoperative changes in intestinal microbiota and use of probiotics in roux-en-y gastric bypass and sleeve vertical gastrectomy: an integrative review.

Arq Bras Cir Dig. Article PubMed PubMed Central Google Scholar. Jandhyala SM, et al. Role of the normal gut microbiota. World J Gastroenterol. Article CAS PubMed PubMed Central Google Scholar.

Guyton K, Alverdy JC. The gut microbiota and gastrointestinal surgery. Shogan BD, et al. Intestinal anastomotic injury alters spatially defined microbiome composition and function. Reddy BS, et al. Surgical manipulation of the large intestine increases bacterial translocation in patients undergoing elective colorectal surgery.

Colorectal Dis. Interactions between commensal bacteria and gut sensorimotor function in health and disease. Am J Gastroenterol.

Ge X, et al. Potential role of fecal microbiota from patients with slow transit constipation in the regulation of gastrointestinal motility. Sci Rep. Bayer S, et al. Effects of GABA on circular smooth muscle spontaneous activities of rat distal colon. Life Sci. Husebye E, et al. Influence of microbial species on small intestinal myoelectric activity and transit in germ-free rats.

Am J Physiol Gastrointest Liver Physiol. Tremaroli V, et al. Roux-en-Y gastric bypass and vertical banded gastroplasty induce long-term changes on the human gut microbiome contributing to fat mass regulation.

Cell Metab. Jahansouz C, et al. Sleeve gastrectomy drives persistent shifts in the gut microbiome. Surg Obes Relat Dis.

Sokol H, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients.

Proc Natl Acad Sci USA. Hegde S, et al. Microbiota dysbiosis and its pathophysiological significance in bowel obstruction. Nalluri-Butz H, et al. A pilot study demonstrating the impact of surgical bowel preparation on intestinal microbiota composition following colon and rectal surgery.

Sun X, et al. Bile is a promising gut nutrient that inhibits intestinal bacterial translocation and promotes gut motility via an interleukinrelated pathway in an animal model of endotoxemia. Shin SY, et al. An altered composition of fecal microbiota, organic acids, and the effect of probiotics in the guinea pig model of postoperative ileus.

Neurogastroenterol Motil. Nyavor Y, et al. High-fat diet-induced alterations to gut microbiota and gut-derived lipoteichoic acid contributes to the development of enteric neuropathy. Antibiotics-induced depletion of mice microbiota induces changes in host serotonin biosynthesis and intestinal motility.

J Transl Med. Iwai H, et al. Effects of bacterial flora on cecal size and transit rate of intestinal contents in mice. Jpn J Exp Med. CAS PubMed Google Scholar.

Lukovic E, Moitra VK, Freedberg DE. The microbiome: implications for perioperative and critical care. Curr Opin Anaesthesiol. Banerjee S, et al. Opioid-induced gut microbial disruption and bile dysregulation leads to gut barrier compromise and sustained systemic inflammation.

Mucosal Immunol. Heitmann PT, et al. The effects of loperamide on excitatory and inhibitory neuromuscular function in the human colon.

Deng Y, et al. Manipulation of intestinal dysbiosis by a bacterial mixture ameliorates loperamide-induced constipation in rats.

Benef Microbes. Aziz Q, et al. Gut microbiota and gastrointestinal health: current concepts and future directions. Cong L, et al. Efficacy of high specific volume polysaccharide: a new type of dietary fiber—on molecular mechanism of intestinal water metabolism in rats with constipation.

Med Sci Monit. Furness JB. The enteric nervous system and neurogastroenterology. Rühl A. Glial cells in the gut. Brookes SJ. Classes of enteric nerve cells in the guinea-pig small intestine. Anat Rec.

Thuneberg L. Interstitial cells of Cajal: intestinal pacemaker cells? Adv Anat Embryol Cell Biol. Hetz S, et al. In vivo transplantation of neurosphere-like bodies derived from the human postnatal and adult enteric nervous system: a pilot study.

PLoS ONE. Musser MA, Michelle Southard-Smith E. Balancing on the crest—evidence for disruption of the enteric ganglia via inappropriate lineage segregation and consequences for gastrointestinal function.

Dev Biol. Bettolli M, et al. Potential significance of abnormalities in the interstitial cells of Cajal and the enteric nervous system.

J Pediatr Surg. Stoffels B, et al. Postoperative ileus involves interleukin-1 receptor signaling in enteric glia. Lo YY, et al. Requirements of focal adhesions and calcium fluxes for interleukininduced ERK kinase activation and c-fos expression in fibroblasts.

J Biol Chem. Snoek SA, et al. Mast cells trigger epithelial barrier dysfunction, bacterial translocation and postoperative ileus in a mouse model. Stein K, et al.

Intestinal manipulation affects mucosal antimicrobial defense in a mouse model of postoperative ileus. Boeckxstaens GE, de Jonge WJ. Neuroimmune mechanisms in postoperative ileus.

Enderes J, et al. A population of radio-resistant macrophages in the deep myenteric plexus contributes to postoperative ileus via Toll-like receptor 3 signaling. Front Immunol. Grasa L, et al. TLR2 and TLR4 interact with sulfide system in the modulation of mouse colonic motility.

Forcén R, et al. Toll-like receptors 2 and 4 modulate the contractile response induced by serotonin in mouse ileum: analysis of the serotonin receptors involved.

Brun P, et al. Toll-like receptor 2 regulates intestinal inflammation by controlling integrity of the enteric nervous system. Toll like receptor-2 regulates production of glial-derived neurotrophic factors in murine intestinal smooth muscle cells.

Mol Cell Neurosci. Anitha M, et al. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling. Lin SS, et al. Alterations in the gut barrier and involvement of Toll-like receptor 4 in murine postoperative ileus.

Türler A, et al. Endogenous endotoxin participates in causing a panenteric inflammatory ileus after colonic surgery. Ann Surg. Antibiotic-induced depletion of murine microbiota induces mild inflammation and changes in Toll-like receptor patterns and intestinal motility.

Microb Ecol. De Schepper S, et al. Muscularis macrophages: key players in intestinal homeostasis and disease. Cell Immunol. Guilarte M, et al. Diarrhoea-predominant IBS patients show mast cell activation and hyperplasia in the jejunum.

Bassotti G, et al. Colonic mast cells in controls and slow transit constipation patients. Aliment Pharmacol Ther. Balestra B, et al. Colonic mucosal mediators from patients with irritable bowel syndrome excite enteric cholinergic motor neurons. de Jonge WJ, et al. Mast cell degranulation during abdominal surgery initiates postoperative ileus in mice.

Peters EG, et al. The contribution of mast cells to postoperative ileus in experimental and clinical studies. Zhang L, Song J, Hou X. Mast cells and irritable bowel syndrome: from the bench to the bedside.

J Neurogastroenterol Motil. Ng QX, et al. The role of inflammation in irritable bowel syndrome IBS. J Inflamm Res. Bednarska O, et al. Vasoactive intestinal polypeptide and mast cells regulate increased passage of colonic bacteria in patients with irritable bowel syndrome.

Davies LC, et al. Tissue-resident macrophages. Nat Immunol. Wehner S, et al. Inhibition of macrophage function prevents intestinal inflammation and postoperative ileus in rodents. Marchix J, Goddard G, Helmrath MA. Host-gut microbiota crosstalk in intestinal adaptation. Cell Mol Gastroenterol Hepatol.

Gabanyi I, et al. Neuro-immune interactions drive tissue programming in intestinal macrophages. Self-maintaining gut macrophages are essential for intestinal homeostasis.

Kalff JC, et al. Biphasic response to gut manipulation and temporal correlation of cellular infiltrates and muscle dysfunction in rat. Induction of IL-6 within the rodent intestinal muscularis after intestinal surgical stress. Furness JB, et al. The enteric nervous system and gastrointestinal innervation: integrated local and central control.

Adv Exp Med Biol. Muller PA, et al. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cipriani G, et al. Intrinsic gastrointestinal macrophages: their phenotype and role in gastrointestinal motility.

Canton J, Neculai D, Grinstein S. Scavenger receptors in homeostasis and immunity. Nat Rev Immunol. Yuan PQ, Taché Y. Abdominal surgery induced gastric ileus and activation of M1-like macrophages in the gastric myenteric plexus: prevention by central vagal activation in rats. Becker L, et al.

Age-dependent shift in macrophage polarisation causes inflammation-mediated degeneration of enteric nervous system. Inoue Y, et al. Colonic M1 macrophage is associated with the prolongation of gastrointestinal motility and obesity in mice treated with vancomycin.

Mol Med Rep. CAS PubMed PubMed Central Google Scholar. Engel DR, et al. T helper type 1 memory cells disseminate postoperative ileus over the entire intestinal tract.

Nat Med. Sha S, et al. Arch Microbiol. Zhao X, et al. Th17 cell-derived amphiregulin promotes colitis-associated intestinal fibrosis through activation of mTOR and MEK in intestinal myofibroblasts. Sanchez-Ruiz M, et al.

Enteric murine ganglionitis induced by autoimmune CD8 T cells mimics human gastrointestinal dysmotility. Am J Pathol. Pohl JM, et al. Gershon MD, Tack J. The serotonin signaling system: from basic understanding to drug development for functional GI disorders. Akiba Y, et al. FFA2 activation combined with ulcerogenic COX inhibition induces duodenal mucosal injury via the 5-HT pathway in rats.

Patel M, et al. Role of substance P in the pathophysiology of inflammatory bowel disease and its correlation with the degree of inflammation. PubMed PubMed Central Google Scholar. Spiller R. Recent advances in understanding the role of serotonin in gastrointestinal motility in functional bowel disorders: alterations in 5-HT signalling and metabolism in human disease.

Hussain Z, et al. YH, a potent and highly selective 5-HT 4 receptor agonist, significantly improves both upper and lower gastrointestinal motility in a guinea pig model of postoperative ileus.

Drake TM, Ward AE. Pharmacological management to prevent ileus in major abdominal surgery: a systematic review and meta-analysis. J Gastrointest Surg. Thomas H. Prucalopride before surgery alleviates postoperative ileus. Tsuchida Y, et al.

Neuronal stimulation with 5-hydroxytryptamine 4 receptor induces anti-inflammatory actions via α7nACh receptors on muscularis macrophages associated with postoperative ileus. Yano JM, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis.

Li B, et al. Engineered 5-HT producing gut probiotic improves gastrointestinal motility and behavior disorder. Front Cell Infect Microbiol. Sugawara G, et al. Perioperative synbiotic treatment to prevent postoperative infectious complications in biliary cancer surgery: a randomized controlled trial.

Sjögren K, et al. The gut microbiota regulates bone mass in mice. J Bone Miner Res. Reigstad CS, et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells.

FASEB J. Clements WD, et al. Role of the gut in the pathophysiology of extrahepatic biliary obstruction. Alemi F, et al. The receptor TGR5 mediates the prokinetic actions of intestinal bile acids and is required for normal defecation in mice.

Poole DP, et al. Expression and function of the bile acid receptor GpBAR1 TGR5 in the murine enteric nervous system. Martin-Gallausiaux C, et al. SCFA: mechanisms and functional importance in the gut.

Proc Nutr Soc. Huang F, et al. Postoperative probiotics administration attenuates gastrointestinal complications and gut microbiota dysbiosis caused by chemotherapy in colorectal cancer patients.

We are inviting people living in the Calgary area to participate in this study at the Foothills Medical Campus to test out a new, non-invasive device to measure the electrical nerve activity of the stomach. There is one study appointment lasting about 5 hours on a weekday. Adults, 18 years and older, with stomach problems such as gastroparesis, functional dyspepsia or chronic vomiting syndrome are needed now to help us complete our study.

Please contact us today. More information. The Calgary Gut Motility Clinic is part of Alberta Health Services offering state-of-the-art diagnostic technology, staffed with an outstanding team of specialist Motility Nurses and Gastroenterologists.

Calgary Gut Motility Centre Working to improve medical care for people with gut motility disorders. We are a team of consulting Gastroenterologists, Researchers and Nurses active in the discovery and development of new diagnostic tools and treatments for gut motility disorders such as gastroparesis, cannabinoid hyperemesis CHS , irritable bowel syndrome IBS , eosinophilic esophagitis EoE , gastroesophageal reflux disease GERD and anorectal dysfunction.

Typically, Ght food Herbal remedies for arthritis present, Arthritis and chiropractic care and relaxations occur at intervals along the Ght this process is motiljty as segmentation contraction. These contractions and relaxations occur bidirectionally Electrolytes and fatigue to mix partially digested jotility, Arthritis and chiropractic care hwalth chyme, with hea,th enzymes. A anf amount of propulsion occurs from this movement, contributing to movement of chyme toward the large intestine. Pacemaker cells, known as the cells of Cajal, initiate these contractions and set the pace and frequency. The nervous system of the gut, known as the enteric nervous system, as well as excitatory hormones chemical messengers must communicate with the cells of Cajal in order for the segmentation contractions to occur. Peristalsis, a type of propulsive electrical movement, is primarily responsible for the movement of chyme through the small intestine to the large intestine. New research shows little risk of ugt from prostate guf. Discrimination at work is linked to motiity Gut health and gut motility pressure. Icy fingers and toes: Poor circulation or Raynaud's phenomenon? The gut-brain connection is no joke; it can link anxiety to stomach problems and vice versa. Have you ever had a "gut-wrenching" experience? Do certain situations make you "feel nauseous"?

Author: Vudoshura

3 thoughts on “Gut health and gut motility

  1. Ich kann anbieten, auf die Webseite, mit der riesigen Zahl der Artikel nach dem Sie interessierenden Thema vorbeizukommen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com