scholarly journals The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation

Nutrients ◽  
2018 ◽  
Vol 10 (8) ◽  
pp. 988 ◽  
Author(s):  
Kaisa Hiippala ◽  
Hanne Jouhten ◽  
Aki Ronkainen ◽  
Anna Hartikainen ◽  
Veera Kainulainen ◽  
...  

The intestinal microbiota, composed of pro- and anti-inflammatory microbes, has an essential role in maintaining gut homeostasis and functionality. An overly hygienic lifestyle, consumption of processed and fiber-poor foods, or antibiotics are major factors modulating the microbiota and possibly leading to longstanding dysbiosis. Dysbiotic microbiota is characterized to have altered composition, reduced diversity and stability, as well as increased levels of lipopolysaccharide-containing, proinflammatory bacteria. Specific commensal species as novel probiotics, so-called next-generation probiotics, could restore the intestinal health by means of attenuating inflammation and strengthening the epithelial barrier. In this review we summarize the latest findings considering the beneficial effects of the promising commensals across all major intestinal phyla. These include the already well-known bifidobacteria, which use extracellular structures or secreted substances to promote intestinal health. Faecalibacterium prausnitzii, Roseburia intestinalis, and Eubacterium hallii metabolize dietary fibers as major short-chain fatty acid producers providing energy sources for enterocytes and achieving anti-inflammatory effects in the gut. Akkermansia muciniphila exerts beneficial action in metabolic diseases and fortifies the barrier function. The health-promoting effects of Bacteroides species are relatively recently discovered with the findings of excreted immunomodulatory molecules. These promising, unconventional probiotics could be a part of biotherapeutic strategies in the future.

2019 ◽  
Vol 56 (4) ◽  
pp. 439
Author(s):  
P. Meena Kumari ◽  
S. P. Muthukumar

Due to its grave pathological role of obesity, comprehensive research is being continued to find out the causative factors involved in it. Recent advances in this field are increasingly recognized that there is a connection between diet, gut microbiota, intestinal barrier function and the low-grade inflammation that characterize the progression from obesity to metabolic disturbances, making dietary strategies to modulate the intestinal environment is important. In this context, the ability of some Gram-positive anaerobic bacteria to produce the shortchain fatty acid butyrate is impressive. A lower abundance of butyrate-producing bacteria has been associated with metabolic risk in humans. Recent studies suggest that butyrate might have been linked with metabolic risk in humans, and recommend that butyrate might have an anti-inflammatory mediator in metabolic diseases, and the potential of butyrate can alleviate obesity-related metabolic complications, possibly due to its ability to enhance the intestinal barrier function. Endogenous butyrate synthesis, delivery, and absorption by colonocytes have been well studied. Butyrate exerts its function by serving as a histone deacetylase (HDAC) inhibitor or signaling through several G Protein-Coupled Receptors (GPCRs). Latterly butyrate has gained selective attention for its favorable effects on intestinal homeostasis and energy metabolism. With anti-inflammatory properties, butyrate improves intestinal barrier function and mucosal immunity. Growing proof has highlighted the influence of butyrate on obesity. In this review the current knowledge on the features of butyrate, especially its potential effects and mechanisms involved in intestinal health and obesity. Here we review and discuss the potentials of butyrate as an anti-inflammatory mediator in obesity and the potential for dietary interventions increasing intestinal availability of butyrate.


2020 ◽  
Vol 11 (12) ◽  
pp. 10839-10851
Author(s):  
Zhi-jie Ma ◽  
Huan-jun Wang ◽  
Xiao-jing Ma ◽  
Yue Li ◽  
Hong-jun Yang ◽  
...  

Ginger extract showed beneficial effects on rats with antibiotic-associated diarrhea, and the underlying mechanism might be associated with the recovery of gut microbiota and intestinal barrier function.


Nutrients ◽  
2019 ◽  
Vol 11 (7) ◽  
pp. 1464 ◽  
Author(s):  
Yala Stevens ◽  
Evelien Van Rymenant ◽  
Charlotte Grootaert ◽  
John Van Camp ◽  
Sam Possemiers ◽  
...  

Citrus flavanones, with hesperidin and naringin as the most abundant representatives, have various beneficial effects, including anti-oxidative and anti-inflammatory activities. Evidence also indicates that they may impact the intestinal microbiome and are metabolized by the microbiota as well, thereby affecting their bioavailability. In this review, we provide an overview on the current evidence on the intestinal fate of hesperidin and naringin, their interaction with the gut microbiota, and their effects on intestinal barrier function and intestinal inflammation. These topics will be discussed as they may contribute to gastrointestinal health in various diseases. Evidence shows that hesperidin and naringin are metabolized by intestinal bacteria, mainly in the (proximal) colon, resulting in the formation of their aglycones hesperetin and naringenin and various smaller phenolics. Studies have also shown that citrus flavanones and their metabolites are able to influence the microbiota composition and activity and exert beneficial effects on intestinal barrier function and gastrointestinal inflammation. Although the exact underlying mechanisms of action are not completely clear and more research in human subjects is needed, evidence so far suggests that citrus flavanones as well as their metabolites have the potential to contribute to improved gastrointestinal function and health.


2017 ◽  
Vol 312 (4) ◽  
pp. C438-C445 ◽  
Author(s):  
Jing Wang ◽  
Siddhartha S. Ghosh ◽  
Shobha Ghosh

Association between circulating lipopolysaccharide (LPS) and metabolic diseases (such as type 2 diabetes and atherosclerosis) has shifted the focus from high-fat high-cholesterol containing Western-type diet (WD)-induced changes in gut microbiota per se to release of gut bacteria-derived products (e.g., LPS) into circulation due to intestinal barrier dysfunction as the possible mechanism for the chronic inflammatory state underlying the development of these diseases. We demonstrated earlier that oral supplementation with curcumin attenuates WD-induced development of type 2 diabetes and atherosclerosis. Poor bioavailability of curcumin has precluded the establishment of a causal relationship between oral supplementation and it is in vivo effects. We hypothesized that curcumin attenuates WD-induced chronic inflammation and associated metabolic diseases by modulating the function of intestinal epithelial cells (IECs) and the intestinal barrier function. The objective of the present study was to delineate the underlying mechanisms. The human IEC lines Caco-2 and HT-29 were used for these studies and modulation of direct as well as indirect effects of LPS on intracellular signaling as well as tight junctions were examined. Pretreatment with curcumin significantly attenuated LPS-induced secretion of master cytokine IL-1β from IECs and macrophages. Furthermore, curcumin also reduced IL-1β-induced activation of p38 MAPK in IECs and subsequent increase in expression of myosin light chain kinase involved in the phosphorylation of tight junction proteins and ensuing disruption of their normal arrangement. The major site of action of curcumin is, therefore, likely the IECs and the intestinal barrier, and by reducing intestinal barrier dysfunction, curcumin modulates chronic inflammatory diseases despite poor bioavailability.


Foods ◽  
2021 ◽  
Vol 10 (7) ◽  
pp. 1595
Author(s):  
Borkwei Ed Nignpense ◽  
Nidhish Francis ◽  
Christopher Blanchard ◽  
Abishek Bommannan Santhakumar

Cereal bioactive compounds, especially polyphenols, are known to possess a wide range of disease preventive properties that are attributed to their antioxidant and anti-inflammatory activity. However, due to their low plasma concentrations after oral intake, there is controversy regarding their therapeutic benefits in vivo. Within the gastrointestinal tract, some cereal polyphenols are absorbed in the small intestine, with the majority accumulating and metabolised by the colonic microbiota. Chemical and enzymatic processes occurring during gastrointestinal digestion modulate the bioactivity and bioaccessibility of phenolic compounds. The interactions between the cereal polyphenols and the intestinal epithelium allow the modulation of intestinal barrier function through antioxidant, anti-inflammatory activity and mucin production thereby improving intestinal health. The intestinal microbiota is believed to have a reciprocal interaction with polyphenols, wherein the microbiome produces bioactive and bioaccessible phenolic metabolites and the phenolic compound, in turn, modifies the microbiome composition favourably. Thus, the microbiome presents a key link between polyphenol consumption and the health benefits observed in metabolic conditions in numerous studies. This review will explore the therapeutic value of cereal polyphenols in conjunction with their bioaccessibility, impact on intestinal barrier function and interaction with the microbiome coupled with plasma anti-inflammatory effects.


2020 ◽  
Author(s):  
Qianqian Xu ◽  
Xiaoting Zou ◽  
Xinyang Dong

Abstract BackgroundLinoleic acid (LA) is predominantly essential for poultry. Deficiency of LA in poultry were manifested in various aspects such as retarded growth and reduced resistance to disease. The effects of LA on intestinal health in vitro and in mammals has been studied, whereas research related to the effects of LA on intestine health in poultry was scanty. Intestinal health and immune function play an important role in pigeon squab growth. Considering squabs are fed by their parents, the purpose of this study was to explore the effects of maternal dietary LA on intestinal barrier function in squabs by determining intestinal morphology, gene expression of tight junction protein, immune cytokines, and microbial flora.ResultsA completely randomized design with a control group, 1% dietary LA supplementation group, 2% dietary LA supplementation group, and 4% dietary LA supplementation group was used. Six squabs from each treatment were randomly sampled at 21 posthatch. Results indicated that LA supplementation improved intestinal morphology as reflected by increased villus height, villus area and the ratio of villus to crypt, and the promotion at dosage of 1% was most significant. Besides, 1% LA supplementation elevated distribution density of goblet cell in intestine, and strengthened tight junction between enterocytes by up-regulating claudin3 and occludin gene expression, but down-regulating claudin2 gene expression. Moreover, 1% LA supplementation reduced secretion of pro-inflammation cytokines and increased anti-inflammation cytokines partly. The diversity index Chao1 of intestinal microbiota in 1% LA supplementation group was higher than other groups. And Butyrivibrio as beneficial bacteria was the biomarker of LA1%. However, excessive (4%) LA supplementation led to adverse impact on intestinal immunity and microbiota.ConclusionsMaternal dietary LA in three levels all could improve intestinal morphology in squabs. Therein, appropriate dosage (1%) supplementation might enhance mucosal protection and epithelium barrieer in squabs, and furthermore consolidated intestine immunity and luminal microbial environment. However, excessive (4%) LA supplementation might lead to adverse impact on immunity and microbiota. Maternal dietary LA might alter intestinal barrier function in pigeon squabs in a dose-dependent manner.


2015 ◽  
Vol 6 (8) ◽  
pp. 2568-2577 ◽  
Author(s):  
Yuan Yue ◽  
Shuangchan Wu ◽  
Zhike Li ◽  
Jian Li ◽  
Xiaofei Li ◽  
...  

Dietary polysaccharides provide various beneficial effects for our health.


2020 ◽  
Author(s):  
Xue Song ◽  
Lugen Zuo ◽  
Luyao Wang ◽  
Zihan Zhu ◽  
Jing Tao ◽  
...  

ABSTRACTOBJECTIVESRottlerin, a pan PDE inhibitor, has a variety of pharmacological activities, including enhancing barrier function and mediating anti-inflammatory activity by changing the distribution of occludin and ZO-1. Nevertheless, the function of rottlerin on Crohn disease (CD) keep unknown. Our aim of the study is to investigate the role of rottlerin on CD-like colitis and its mechanism.METHODSWild-type mice which were 8-10 weeks old were randomly divided into three treatment groups: (i) the normal feeding, no administration (control) group, (ii) the group administered 3% dextran sodium sulfate (DSS) alone, and (iii) the group administered rottlerin (100 mg/kg) and 3% DSS. In this study, the effect of rottlerin on the function and structure of the intestinal barrier was investigated, and the possible mechanism was discussed. We performed signaling pathway analysis and flow cytometry to identify the detailed mechanisms by which rottlerin (10 μg/mL) treatment inhibits cell growth arrest and the attenuation of TJ proteins in LPS-treated FHs 74 int cells.RESULTSRottlerin treatment significantly ameliorated colitis induced by DSS in WT mice, which was manifested by a decrease in inflammation score, the attenuation of inflammatory factors and the inhibition of destruction on intestinal barrier structure. Rottlerin enhanced the levels of occludin and ZO-1, and improved the function of intestinal barrier, which may have been why rottlerin ameliorated colitis in WT mice. The anti-inflammatory effect of rottlerin may be partly due to the activation of Epac-2/Rap-1 signaling.CONCLUSIONSRottlerin may treat CD in humans via enhancing TJ proteins expression and improving the function of intestinal barrier.


2018 ◽  
Vol 6 (1) ◽  
pp. e1425085 ◽  
Author(s):  
Siddhartha S. Ghosh ◽  
Hongliang He ◽  
Jing Wang ◽  
Todd W. Gehr ◽  
Shobha Ghosh

Sign in / Sign up

Export Citation Format

Share Document