scholarly journals Melatonin and its-Remodeled Fecal Microbiota Improve Gut Health Through Inhibiting Oxidative Stress, Autophagy and Inflammation

Author(s):  
Yingqian Wang ◽  
Weike Shaoyong ◽  
Rui Cai ◽  
Xudong Yi ◽  
Wenlong Yong ◽  
...  

Abstract Background Gut health is involved in the nutrition absorption, reproduction and lactation in antenatal, perinatal and early weaned mammals. Recent literatures have demonstrated that melatonin functions in aging, cancer and obesity, but to date, few investigations toward exploring whether melatonin-reprogrammed fecal microbiota transplantation (FMT) and foster care (FC) affect gut health have been performed. Results Here, compared with the control group, melatonin and FMT increased intestinal villus height/crypt depth (V/C), inhibited gut oxidative stress, autophagy and inflammation in antenatal and perinatal rats. Likewise, not only melatonin and FMT, but also FC enhanced intestinal V/C through above parallel ways with decreasing intestinal permeability in early weaned rats. Mechanically, melatonin directly strengthened antioxidation, attenuated autophagy and apoptosis in H2O2-induced IEC6 intestinal epithelial cells. Furthermore, melatonin, FMT and FC reprogrammed intestinal microbiota in which more beneficial microbiota, including Allobaculum, Bifidobacterium and Faecalibaculum produced more metabolic short-chain fatty acids (SCFAs) including acetic acid and butyric acid to protect gut health. Most interestingly, compared with the control group, early weaned rats may get above probiotics via eating or licking the dung of the co-cage rats treated with melatonin in the FC group. Conclusions Overall, melatonin, FMT and FC improved gut health and the potential regulatory mechanism was associated with strengthening antioxidation, suppressing autophagy, inflammatory and apoptosis, as well as producing more SCFAs from reprogrammed gut microbiota. These findings suggest that melatonin, FMT and FC may be novel and effective methods to ameliorate gut health in antenatal, perinatal and weaned mammals.

2019 ◽  
Vol 97 (Supplement_3) ◽  
pp. 305-306
Author(s):  
Quanhang Xiang ◽  
Jian Peng

Abstract The objective of this study was to investigate the effects of early gut colonization by fecal microbiota transplantation and probiotics intervention on growth performance, immunity function, and gut health of piglets. A total of 121 pregnant sows were divided into 6 groups with average parity of 3.66 ± 1.34. After delivery, piglets of group AB were treated with antibiotics at age of 3-day. Piglets of group CON were gavaged with PBS. The remaining four treatment groups, FMT, FMT+C, FMT+S, and FMT+C+S, the piglets were gavaged with fecal suspension, fecal suspension with C. butyricum, fecal suspension with S. boulardii, and fecal suspension with C. butyricum and S.boulardii, respectively, with the frequency of once daily in the first 3 days. All the piglets were weaned at age of 21 day. The individual body weight of piglets were weighed weekly, blood samples and fecal samples were collected weekly. At the end of study, the ADG and diarrhea rate were caculated. FMT+C+S and FMT could increased piglets 21-day-old weight (P < 0.01), and FMT+C+S could increased ADG (P < 0.05) and decreased diarrhea rate (P < 0.05). Early antibiotics exposure for health care has no positive effect on growth performance and diarrhea. FMT, FMT+S and FMT+C+S improved fecal sIgA and plasma IgG of 14-day-old piglets (P < 0.05). FMT+C+S decreased the concentration of plasma DAO and D-LA, and increased fecal MUC2 content, so that the intestinal barrier was enhanced. The early intervention of FMT combined with C. butyricum and S. boulardii reduced the abundance of E. coli, and increased the abundance of Lactobacillus, Bifidobacterium and Faecalibacterium prausnitzii. In addition, it also increases the production of intestinal short-chain fatty acids. In conclusion, these data indicated that early intervention with FMT combined C. butyricum and S. boulardii could improve the growth performance, immune responses, and gut function of sucking piglets.


Nutrients ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 2180
Author(s):  
Jessica M. Moon ◽  
Peter Finnegan ◽  
Richard A. Stecker ◽  
Hanna Lee ◽  
Kayla M. Ratliff ◽  
...  

Glucosamine (GLU) is a natural compound found in cartilage, and supplementation with glucosamine has been shown to improve joint heath and has been linked to reduced mortality rates. GLU is poorly absorbed and may exhibit functional properties in the gut. The purpose of this study was to examine the impact of glucosamine on gastrointestinal function as well as changes in fecal microbiota and metabolome. Healthy males (n = 6) and females (n = 5) (33.4 ± 7.7 years, 174.1 ± 12.0 cm, 76.5 ± 12.9 kg, 25.2 ± 3.1 kg/m2, n = 11) completed two supplementation protocols that each spanned three weeks separated by a washout period that lasted two weeks. In a randomized, double-blind, placebo-controlled, crossover fashion, participants ingested a daily dose of GLU hydrochloride (3000 mg GlucosaGreen®, TSI Group Ltd., USA) or maltodextrin placebo. Study participants completed bowel habit and gastrointestinal symptoms questionnaires in addition to providing a stool sample that was analyzed for fecal microbiota and metabolome at baseline and after the completion of each supplementation period. GLU significantly reduced stomach bloating and showed a trend towards reducing constipation and hard stools. Phylogenetic diversity (Faith’s PD) and proportions of Pseudomonadaceae, Peptococcaceae, and Bacillaceae were significantly reduced following GLU consumption. GLU supplementation significantly reduced individual, total branched-chain, and total amino acid excretion, with no glucosamine being detected in any of the fecal samples. GLU had no effect on fecal short-chain fatty acids levels. GLU supplementation provided functional gut health benefits and induced fecal microbiota and metabolome changes.


2010 ◽  
Vol 1 (2) ◽  
pp. 189-196 ◽  
Author(s):  
U. Andersson ◽  
C. Bränning ◽  
S. Ahrné ◽  
G. Molin ◽  
J. Alenfall ◽  
...  

Today, the gut microbiota is considered a key organ in host nutritional metabolism and recent data have suggested that alterations in gut microbiota contribute to the development of type 2 diabetes and obesity. Accordingly, a whole range of beneficial effects relating to inflammation and gut health have been observed following administration of probiotics to both humans and different animal models. The objective of this study was to evaluate the metabolic effects of an oral probiotic supplement, Lactobacillus plantarum DSM 15313, to high-fat diet (HFD) fed C57BL/6J mice, a model of human obesity and early diabetes. The mice were fed the experimental diets for 20 weeks, after which the HFD had induced an insulin-resistant state in both groups compared to the start of the study. The increase in body weight during the HFD feeding was higher in the probiotic group than in the control group, however, there were no significant differences in body fat content. Fasting plasma glucose levels were lower in the group fed the probiotic supplement, whereas insulin and lipids were not different. Caecal levels of short-chain fatty acids were not significantly different between the groups. An oral glucose tolerance test showed that the group fed probiotics had a significantly lower insulin release compared to the control group, although the rate of glucose clearance was not different. Taken together, these data indicate that L. plantarum DSM 15313 has anti-diabetic properties when fed together with an HFD.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Chunchun Wang ◽  
Shuting Cao ◽  
Zhuojun Shen ◽  
Qihua Hong ◽  
Jie Feng ◽  
...  

Abstract Background The objective of this experiment was to investigate the influence of dietary tributyrin on intestinal mucosa development, oxidative stress, mitochondrial function and AMPK-mTOR signaling pathway. Methods Seventy-two pigs were divided into two treatments and received either a basal diet or the same diet supplemented with 750 mg/kg tributyrin. Each treatment has six replicates of six pigs. After 14 days, 6 pigs from each treatment were selected and the jejunal samples were collected. Results Results showed that supplemental tributyrin increased (P < 0.05) villus height and villus height: crypt depth of weaned pigs. Pigs fed tributyrin had greater (P < 0.05) RNA/DNA and protein/DNA ratios than pigs on the control group. The mRNA levels of sodium glucose transport protein-1 and glucose transporter-2 in the jejunum were upregulated (P < 0.05) in pigs fed the tributyrin diet. Dietary tributyrin supplementation lowered (P < 0.05) the malondialdehyde and hydrogen peroxide (H2O2) content in jejunum, enhanced (P < 0.05) the mitochondrial function, as demonstrated by decreased (P < 0.05) reactive oxygen species level and increased (P < 0.05) mitochondrial membrane potential. Furthermore, tributyrin increased (P < 0.05) mitochondrial DNA content and the mRNA abundance of genes related to mitochondrial functions, including peroxisomal proliferator-activated receptor-γ coactivator-1α, mitochondrial transcription factor A, nuclear respiratory factor-1 in the jejunum. Supplementation with tributyrin elevated (P < 0.05) the phosphorylation level of AMPK and inhibited (P < 0.05) the phosphorylation level of mTOR in jejunum compared with the control group. Conclusions These findings suggest that dietary supplementation with tributyrin promotes intestinal mucosa growth, extenuates oxidative stress, improves mitochondrial function and modulates the AMPK-mTOR signal pathway of weaned pigs.


Nutrients ◽  
2020 ◽  
Vol 12 (10) ◽  
pp. 3194
Author(s):  
Jing Wang ◽  
Yong Chen ◽  
Xiaosong Hu ◽  
Fengqin Feng ◽  
Luyun Cai ◽  
...  

The beneficial effects of ginger polyphenols have been extensively reported. However, their metabolic characteristics and health effects on gut microbiota are poor understood. The purpose of this study was to investigate the digestion stability of ginger polyphenols and their prebiotic effects on gut microbiota by simulating digestion and fermentation in vitro. Following simulated digestion in vitro, 85% of the polyphenols were still detectable, and the main polyphenol constituents identified in ginger extract are 6-, 8-, and 10-gingerols and 6-shogaol in the digestive fluids. After batch fermentation, the changes in microbial populations were measured by 16S rRNA gene Illumina MiSeq sequencing. In mixed-culture fermentation with fecal inoculate, digested ginger extract (GE) significantly modulated the fecal microbiota structure and promoted the growth of some beneficial bacterial populations, such as Bifidobacterium and Enterococcus. Furthermore, incubation with GE could elevate the levels of short-chain fatty acids (SCFAs) accompanied by a decrease in the pH value. Additionally, the quantitative PCR results showed that 6-gingerol (6G), as the main polyphenol in GE, increased the abundance of Bifidobacterium significantly. Therefore, 6G is expected to be a potential prebiotic that improves human health by promoting gut health.


2021 ◽  
Vol 7 ◽  
Author(s):  
Zhilong Tian ◽  
Xiaodan Wang ◽  
Yehui Duan ◽  
Yue Zhao ◽  
Wenming Zhang ◽  
...  

This study was conducted to investigate the effects of dietary supplementation with different types of Bacillus subtilis (B. subtilis) on the growth and gut health of weaned piglets. A total of 160 piglets were randomly assigned into four groups: control group (a basal diet), BS-A group (a basal diet supplemented with B. subtilis A at 1 × 106 CFU/g feed), BS-B group (a basal diet supplemented with B. subtilis B at 1 × 106 CFU/g feed), and BS-C group (a basal diet supplemented with B. subtilis C at 1 × 106 CFU/g feed). All groups had five replicates with eight piglets per replicate. On days 7, 21, and 42 of the trial, blood plasma and intestinal tissues and digesta samples were collected to determine plasma cytokine concentrations, intestinal morphology, gut microbiota community and metabolic activity, and the expression of genes related to gut physiology and metabolism. The results showed that dietary B. subtilis supplementation improved (P &lt; 0.05) the body weight and average daily gain (in BS-B and BS-C groups) of weaned piglets and decreased (P &lt; 0.05) the diarrhea rates (in BS-A, BS-B, and BS-C groups). In the intestinal morphology analysis, B. subtilis supplementation improved (P &lt; 0.05) the size of villus height and villus height to crypt depth ratio in the ileum of weaned piglets. Firmicutes, Bacteroidetes, and Tenericutes were the most dominant microflora in piglets' colon whatever the trial group and time of analysis. Dietary BS-C supplementation increased (P &lt; 0.05) the relative abundances of Anaerovibrio and Bulleidia and decreased (P &lt; 0.05) the relative abundances of Clostridium and Coprococcus compared with the control group. In addition, dietary B. subtilis supplementation increased (P &lt; 0.05) the indicators of intestinal health, including plasma levels of interleukin (IL)-2 and IL-10, as well as the colonic levels of short-chain fatty acids. Furthermore, dietary B. subtilis supplementation also up-regulated (P &lt; 0.05) the expression of genes involved in metabolic pathways related to intestinal microbiota maturation. In conclusion, these findings suggest that a diet containing BS-B or BS-C can efficiently promote growth performance, decrease diarrhea incidence, and ameliorate several indicators of intestinal health through the modulation of gut microbiota composition and metabolic activity in weaned piglets.


2020 ◽  
Vol 14 (10) ◽  
pp. 1074-1080
Author(s):  
Muluneh Ademe

A growing body of literatures showed the interaction of dysbiotic gut with a wide range of disorders, and the clinical use of fecal microbiota transplantation (FMT) shifted from infectious disease to non-communicable disorders. Despite the promising therapeutic benefits of FMT, the exact mechanisms through which fecal recipients benefit from the fecal intervention are not well understood. However, owing to the advantages of having a healthy gut microbiome, possible mechanisms of actions of FMT has been described. On the one hand, through direct ecological competition, FMT may potentially stimulate decolonization of pathogenic microorganisms and increase host resistance to pathogens. Moreover, following dysbiosis, abnormal microbial colonization of the gastrointestinal tract may also cause excessive or dysregulated immune response, resulting in chronic inflam­mation and the development of mucosal lesions. In this regard, repopulating gut microbiome through FMT helps to restore immune function and reduce host damage. On the other hand, FMT helps to restore essential metabolites used for host metabolism, including short-chain fatty acids (SCFA), antimicrobial peptides (AMP), bacteriocins and bile acids. Therefore, in this review, the existing evidences regarding the mechanisms of action, current opportunities and challenges of FMT will be described.


2021 ◽  
Vol 4 (2) ◽  
pp. e00151
Author(s):  
E.A. Sorokina ◽  
E.S. Zhgun ◽  
Yu.V. Kislun ◽  
E.A. Denisova ◽  
Yu.A. Bespyatykh ◽  
...  

Fecal microbiota transplantation (FMT) is now considered as an effective tool for the treatment of various GI pathologies. Fecal preparations are delivered both through the lower GIT (enema, colonoscopy) and upper (endoscopy, capsules). A common disadvantage of instrumental methods of administration is their high invasiveness associated with the risk of intestinal perforation and the use of anesthesia. Oral capsules are minimally invasive, comfortable and more aesthetic, so this method of drug delivery is gaining popularity. The main issue with the use of frozen feces (including the lyophilisate used in capsules) is its efficiency compared to the original material. During lyophilization, cells are exposed to stress factors such as low temperatures, water crystallization, osmotic stress, changes in pH, and dehydration. To reduce the likelihood of cell damage during lyophilization, protective media (lyo-protectants) are used. In this work sucrose, gelatin, and their combinations have been used as lyoprotectors. To estimate the number of microorganisms, a bacteriological study was carried out. The number of Bifidobacteria, Lactobacilli, and the total number of E.coli and Enterobacteriaceae was estimated. It was found that the lyophilized stool sample containing 10% sucrose as a protective medium had the highest number of viable cells. Also, the physical properties of the lyophilisate (its flowability) are convenient for preparing capsulated form. The molar ratios of short chain fatty acids (SCFAs) in the original fecal samples and lyophilisates were studied by gas chromatography. The molar ratios of major SCFAs (acetate, propionate and butyrate) were identical in the samples studied. The composition of the protective medium in which the lyophilized biomaterial corresponds to the original feces in terms of the number of "live" microorganisms has been proposed. According to its physical characteristics lyophilisate is convenient for capsules preparation.


2020 ◽  
Author(s):  
Yingli Jing ◽  
Yan Yu ◽  
Fan Bai ◽  
Limiao Wang ◽  
Degang Yang ◽  
...  

Abstract Background: Spinal cord injury (SCI) patients display disruption of gut microbiome and gut dysbiosis exacerbate neurological impairment in SCI models. Cumulative data support an important role of gut microbiome in SCI. Here, we investigated the hypothesis that fecal microbiota transplantation (FMT) may exert a neuroprotective effect on SCI mice. Results: We found that FMT facilitated functional recovery, promoted neural axonal regeneration, improved animal weight gain and metabolic profiling, and enhanced intestinal barrier integrity and GI motility. High-throughput sequencing revealed that levels of phylum Firmicutes, genus Blautia, Anaerostipes and Lactobacillus were reduced in fecal samples of SCI mice, and FMT remarkably reshaped gut microbiome. Also, FMT-treated SCI mice showed increased amount of fecal short-chain fatty acids (SCFAs), which correlated with alteration of intestinal permeability and locomotor recovery. Furthermore, FMT down-regulated IL-1β/NF-κB signaling in spinal cord and NF-κB signaling in gut. Conclusion: Our study demonstrates that reprogramming of gut microbiota by FMT improves locomotor and GI functions in SCI mice, possibly through the anti-inflammatory functions of SCFAs.


Sign in / Sign up

Export Citation Format

Share Document