scholarly journals Recovery strategies intestinal mucosal barrier

2021 ◽  
Vol 1 (6) ◽  
pp. 88-95
Author(s):  
A. A. Khryanin ◽  
M. F. Osipenko ◽  
O. B. Nemchaninova ◽  
O. N. Pozdnyakova ◽  
T. B. Reshetnikova ◽  
...  

The surface of the intestinal tract in healthy people is free of bacteria in all segments of the intestine. Thus, the attachment of bacteria to epithelial cells is a sign of infection. Unlike the mucous membrane, the intestinal lumen is never sterile. The reason for this is the polymicrobial nature of the gut microbiota. In the intestine, segments such as the stomach or small intestine, where bacteria are actively suppressed, the microbiota is random in appearance, composition, and concentration. However, the situation is completely different in the colon, where the growth of bacteria increases and their suppression is suspended. The concentration and diversity of bacteria in the colon reaches astronomical numbers. Some of these bacteria are required for the colon to function. Many of the local bacteria in the colon are potential pathogens: Bacteroides, Enterobacteriaceae, Enterococci, and Clostridium histolyticum. Control of pathogens in the colon is achieved through an impenetrable mucus layer. Inflammatory bowel disease is a polymicrobial infection characterized by persistent disruption of the mucosal barrier, subsequent migration of bacteria to the mucous membrane, and overgrowth of a complex bacterial biofilm on the surface of the epithelium, resulting in invasive and cytopathological effects. As long as the mucosal barrier function is impaired, the inflammatory process cannot successfully remove bacteria from the mucosal surface, and inflammation itself is detrimental. Due to the inflammatory reaction, the composition and structure of the fecal microbiota changes. Based on the biostructure of the fecal casts, active Crohn’s disease and ulcerative colitis can be distinguished from each other and from other gastrointestinal diseases. The relationship between the gut microbiome and various dermatological diseases (psoriasis, acne, rosacea, atopic dermatitis) is discussed.

2018 ◽  
Vol 5 (3) ◽  
pp. 180107 ◽  
Author(s):  
Sophie C. Payne ◽  
Robert K. Shepherd ◽  
Alicia Sedo ◽  
James B. Fallon ◽  
John B. Furness

Inflammatory damage to the bowel, as occurs in inflammatory bowel disease (IBD), is debilitating to patients. In both patients and animal experimental models, histological analyses of biopsies and endoscopic examinations are used to evaluate the disease state. However, such measurements often have delays and are invasive, while endoscopy is not quantitatively objective. Therefore, a real-time quantitative method to assess compromised mucosal barrier function is advantageous. We investigated the correlation of in vivo changes in electrical transmural impedance with histological measures of inflammation. Four platinum (Pt) ball electrodes were placed in the lumen of the rat small intestine, with a return electrode under the skin. Electrodes placed within the non-inflamed intestine generated stable impedances during the 3 h testing period. Following an intraluminal injection of 2,4,6-trinitrobenzene sulfonic acid (TNBS), an established animal model of IBD, impedances in the inflamed region significantly decreased relative to a region not exposed to TNBS ( p  < 0.05). Changes in intestinal transmural impedance were correlated ( p  < 0.05) with histologically assessed damage to the mucosa and increases in neutrophil, eosinophil and T-cell populations at 3 h compared with tissue from control regions. This quantitative, real-time assay may have application in the diagnosis and clinical management of IBD.


Nutrients ◽  
2020 ◽  
Vol 12 (4) ◽  
pp. 896 ◽  
Author(s):  
Binning Wu ◽  
Rohil Bhatnagar ◽  
Vijaya V. Indukuri ◽  
Shara Chopra ◽  
Kylie March ◽  
...  

Inflammatory bowel disease (IBD), a chronic intestinal inflammatory condition, awaits safe and effective preventive strategies. Naturally occurring flavonoid compounds are promising therapeutic candidates against IBD due to their great antioxidant potential and ability to reduce inflammation and improve immune signaling mediators in the gut. In this study, we utilized two maize near-isogenic lines flavan-4-ols-containing P1-rr (F+) and flavan-4-ols-lacking p1-ww (F−) to investigate the anti-inflammatory property of flavan-4-ols against carboxymethylcellulose (CMC)-induced low-grade colonic inflammation. C57BL/6 mice were exposed to either 1% CMC (w/v) or water for a total of 15 weeks. After week six, mice on CMC treatment were divided into four groups. One group continued on the control diet. The second and third groups were supplemented with F+ at 15% or 25% (w/w). The fourth group received diet supplemented with F− at 15%. Here we report that mice consuming F+(15) and F+(25) alleviated CMC-induced increase in epididymal fat-pad, colon histology score, pro-inflammatory cytokine interleukin 6 expression and intestinal permeability compared to mice fed with control diet and F−(15). F+(15) and F+(25) significantly enhanced mucus thickness in CMC exposed mice (p < 0.05). These data collectively demonstrated the protective effect of flavan-4-ol against colonic inflammation by restoring intestinal barrier function and provide a rationale to breed for flavan-4-ols enriched cultivars for better dietary benefits.


Acta Medica ◽  
2021 ◽  
pp. 1-9
Author(s):  
Ayşe Buruş ◽  
Başak Çeltikçi ◽  
Yasemin Aksoy

Microbiome studies have shown alterations in bacterial communities in the state of many diseases, including inflammatory bowel disease, metabolic disorders, autoimmune diseases, neurodegenerative diseases, and cancer. Chronic inflammation is a common promoter of many of these pathological processes. Shifting in the microbial diversity is also known as dysbiosis. Dysbiosis, increased detrimental bacterial products, decreased favorable microbial metabolites, interrupted tissue barriers, and bacterial translocation cause excessive immune response and inflammation. Several mechanisms play a role to maintain intestinal homeostasis by limiting bacterial translocation from the intestinal lumen into the lamina propria. Among these mechanisms, most importantly, the mucosal barrier that consists of the antimicrobial peptides, mucus, and immunoglobulin A is fundamental to protect epithelial barrier integrity to reduce the excessive immune response. Moreover, recognizing bacteria and metabolites through receptors results in T cell regulation and immune modulation, which is the keystone of the controlled immune response. This review summarizes the anti-inflammatory and pro-inflammatory mechanisms driven by gastrointestinal microbiota, and it also highlights the recent approaches, including epigenetics and precision medicine.


F1000Research ◽  
2015 ◽  
Vol 4 ◽  
pp. 1146 ◽  
Author(s):  
Jonathan D. Kaunitz ◽  
Piyush Nayyar

The annual incidence of the inflammatory bowel diseases (IBDs) ulcerative colitis and Crohn’s disease has increased at an alarming rate. Although the specific pathophysiology underlying IBD continues to be elusive, it is hypothesized that IBD results from an aberrant and persistent immune response directed against microbes or their products in the gut, facilitated by the genetic susceptibility of the host and intrinsic alterations in mucosal barrier function. In this review, we will describe advances in the understanding of how the interaction of host genetics and the intestinal microbiome contribute to the pathogenesis of IBD, with a focus on bacterial metabolites such as short chain fatty acids (SCFAs) as possible key signaling molecules.  In particular, we will describe alterations of the intestinal microbiota in IBD, focusing on how genetic loci affect the gut microbial phylogenetic distribution and the production of their major microbial metabolic product, SCFAs. We then describe how enteroendocrine cells and myenteric nerves express SCFA receptors that integrate networks such as the cholinergic and serotonergic neural systems and the glucagon-like peptide hormonal pathway, to modulate gut inflammation, permeability, and growth as part of an integrated model of IBD pathogenesis.  Through this integrative approach, we hope that novel hypotheses will emerge that will be tested in reductionist, hypothesis-driven studies in order to examine the interrelationship of these systems in the hope of better understanding IBD pathogenesis and to inform novel therapies.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Weiwei Liang ◽  
Xinjian Peng ◽  
Qingqing Li ◽  
Pingzhang Wang ◽  
Ping Lv ◽  
...  

AbstractThe physiological homeostasis of gut mucosal barrier is maintained by both genetic and environmental factors and its impairment leads to pathogenesis such as inflammatory bowel disease. A cytokine like molecule, FAM3D (mouse Fam3D), is highly expressed in mouse gastrointestinal tract. Here, we demonstrate that deficiency in Fam3D is associated with impaired integrity of colonic mucosa, increased epithelial hyper-proliferation, reduced anti-microbial peptide production and increased sensitivity to chemically induced colitis associated with high incidence of cancer. Pretreatment of Fam3D−/− mice with antibiotics significantly reduces the severity of chemically induced colitis and wild type (WT) mice co-housed with Fam3D−/− mice phenocopy Fam3D-deficiency showing increased sensitivity to colitis and skewed composition of fecal microbiota. An initial equilibrium of microbiota in cohoused WT and Fam3D−/− mice is followed by an increasing divergence of the bacterial composition after separation. These results demonstrate the essential role of Fam3D in colon homeostasis, protection against inflammation associated cancer and normal microbiota composition.


2017 ◽  
Vol 8 (4) ◽  
pp. 605-614 ◽  
Author(s):  
J.R. Bailey ◽  
V. Vince ◽  
N.A. Williams ◽  
T.A. Cogan

Treatment of inflammatory bowel disease (IBD) is mainly based on suppression of symptoms, often with numerous side effects. Trials of probiotics in IBD have frequently produced disappointing results. The majority of probiotics are unusual, since they do not require iron for growth, unlike many bacteria resident in the intestine. The IBD intestine is iron-rich due to bleeding and use of oral iron supplements; conventional probiotics would be rapidly outcompeted. We have evaluated an iron-responsive Streptococcus thermophilus strain for its potential to reduce signs of colitis. Efficacy of S. thermophilus was evaluated in the dextran sodium sulphate mouse model of colitis. Treated animals were given 1×108 cfu S. thermophilus per day and clinical observations were taken daily. At termination, gross and histopathological signs of disease, cellular infiltration, location of bacteria, and cytokine expression in the intestine were determined. S. thermophilus delayed onset of colitis and reduced clinical signs of disease, including bodyweight loss and gastrointestinal bleeding. It reduced bacterial translocation into the colonic tissue. Increased numbers of CD8+ intraepithelial lymphocytes were seen in control animals treated with S. thermophilus. S. thermophilus had no effect on gross pathology, histopathology or cytokine production in either colitic or control animals. We propose that S. thermophilus promotes maintenance of mucosal barrier function which reduces bacterial translocation, thereby reducing immune stimulation and associated inflammation. This allows mucosal healing, reducing gastrointestinal bleeding and weight loss. This could be studied as a locally-acting adjunct or alternative to current IBD treatments.


2020 ◽  
Vol 21 (8) ◽  
pp. 777-784
Author(s):  
Manyi Jia ◽  
Yucheng Zhang ◽  
Yuqi Gao ◽  
Xi Ma

Medium-chain fatty acids (MCFAs) are the main form of Medium Chain Triglycerides (MCTs) utilized by monogastric animals. MCFAs can be directly absorbed and supply rapid energy to promote the renewal and repair of intestinal epithelial cells, maintain the integrity of intestinal mucosal barrier function, and reduce inflammation and stress. In our review, we pay more attention to the role of MCFAs on intestinal microbiota and mucosa immunity to explore MCFA's positive effect. It was found that MCFAs and their esterified forms can decrease pathogens while increasing probiotics. In addition, being recognized via specific receptors, MCFAs are capable of alleviating inflammation to a certain extent by regulating inflammation and immune-related pathways. MCFAs may also have a certain value to relieve intestinal allergy and inflammatory bowel disease (IBD). Unknown mechanism of various MCFA characteristics still causes dilemmas in the application, thus MCFAs are used generally in limited dosages and combined with short-chain organic acids (SOAs) to attain ideal results. We hope that further studies will provide guidance for the practical use of MCFAs in animal feed.


2016 ◽  
Vol 30 (1) ◽  
pp. 191-231 ◽  
Author(s):  
Lauren E. Hudson ◽  
Sarah E. Anderson ◽  
Anita H. Corbett ◽  
Tracey J. Lamb

SUMMARY Beneficial microorganisms hold promise for the treatment of numerous gastrointestinal diseases. The transfer of whole microbiota via fecal transplantation has already been shown to ameliorate the severity of diseases such as Clostridium difficile infection, inflammatory bowel disease, and others. However, the exact mechanisms of fecal microbiota transplant efficacy and the particular strains conferring this benefit are still unclear. Rationally designed combinations of microbial preparations may enable more efficient and effective treatment approaches tailored to particular diseases. Here we use an infectious disease, C. difficile infection, and an inflammatory disorder, the inflammatory bowel disease ulcerative colitis, as examples to facilitate the discussion of how microbial therapy might be rationally designed for specific gastrointestinal diseases. Fecal microbiota transplantation has already shown some efficacy in the treatment of both these disorders; detailed comparisons of studies evaluating commensal and probiotic organisms in the context of these disparate gastrointestinal diseases may shed light on potential protective mechanisms and elucidate how future microbial therapies can be tailored to particular diseases.


2021 ◽  
Vol 5 (1) ◽  
pp. 004-009
Author(s):  
Ohara Tadashi

Currently, the emergence of highly virulent mutants in Europe and the United States has caused refractory recurrent Clostridium difficile infection (RCDI) to be a problem in clinical practice. In 2013, the Netherland group demonstrated breakthrough therapeutic efficacy in fecal microbial transplant (FMT) treatment clinical trials for RCDI, and FMT treatment is rapidly gaining attention. In addition to RCDI, FMT treatment has been attempted in various gastrointestinal diseases such as inflammatory bowel disease, irritable bowel syndrome and chronic constipation, as well as extragastrointestinal diseases. In this review, I would like to describe the current status, complications and prospects of FMT treatment.


Sign in / Sign up

Export Citation Format

Share Document