scholarly journals Functional enrichment of gut microbiome by early supplementation of Bacillus based probiotic in cage free hens: a field study

2021 ◽  
Vol 3 (1) ◽  
Author(s):  
Samiullah Khan ◽  
Kapil K. Chousalkar

Abstract Background The chicken gut microbiota passes through different stages of maturation; therefore, strengthening it with well characterised probiotics increases its resilience required for optimum gut health and wellbeing. However, there is limited information on the interaction of Bacillus based probiotics with gut microbial community members in cage free laying chickens both in rearing and production phases of life. In the current study, we investigated the changes in the gut microbiome of free range hens in the field after Bacillus based probiotic supplementation. Results Overall, at phylum level, probiotic supplementation increased the populations of Bacteroidetes and Proteobacteria mainly at the expense of Firmicutes. The population of Bacteroidetes significantly increased during the production as compared to the rearing phase, and its higher population in the probiotic-supplemented chickens reflects the positive role of Bacillus based probiotic in gut health. Core differences in the beta diversity suggest that probiotic supplementation decreased microbial compositionality. The non-significant difference in alpha diversity between the probiotic and control chickens showed that the composition of community structure did not change. No Salmonella spp. were isolated from the probiotic supplemented birds. Egg internal quality was significantly higher, while egg production and body weight did not differ. Functional prediction data showed that probiotic supplementation enriched metabolic pathways, such as vitamin B6 metabolism, phenylpropanoid biosynthesis, monobactam biosynthesis, RNA degradation, retinol metabolism, pantothenate and CoA biosynthesis, phosphonate and phosphinate metabolism, AMPK signaling pathway, cationic antimicrobial peptide (CAMP) resistance and tyrosine metabolism. Conclusions Overall, age was the main factor affecting the composition and diversity of gut microbiota, where probiotic supplementation improved the abundance of many useful candidates in the gut microbial communities. The generated baseline data in the current study highlights the importance of the continuous use of Bacillus based probiotic for optimum gut health and production.

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Anne-Sophie Alvarez ◽  
Julien Tap ◽  
Isabelle Chambaud ◽  
Stéphanie Cools-Portier ◽  
Laurent Quinquis ◽  
...  

Abstract Many clinical studies have evaluated the effect of probiotics, but only a few have assessed their dose effects on gut microbiota and host. We conducted a randomized, double-blind, controlled intervention clinical trial to assess the safety (primary endpoint) of and gut microbiota response (secondary endpoint) to the daily ingestion for 4 weeks of two doses (1 or 3 bottles/day) of a fermented milk product (Test) in 96 healthy adults. The Test product is a multi-strain fermented milk product, combining yogurt strains and probiotic candidate strains Lactobacillus paracasei subsp. paracasei CNCM I-1518 and CNCM I-3689 and Lactobacillus rhamnosus CNCM I-3690. We assessed the safety of the Test product on the following parameters: adverse events, vital signs, hematological and metabolic profile, hepatic, kidney or thyroid function, inflammatory markers, bowel habits and digestive symptoms. We explored the longitudinal gut microbiota response to product consumption and dose, by 16S rRNA gene sequencing and functional contribution by shotgun metagenomics. Safety results did not show any significant difference between the Test and Control products whatever the parameters assessed, at the two doses ingested daily over a 4-week-period. Probiotic candidate strains were detected only during consumption period, and at a significantly higher level for the three strains in subjects who consumed 3 products bottles/day. The global structure of the gut microbiota as assessed by alpha and beta-diversity, was not altered by consumption of the product for four weeks. A zero-inflated beta regression model with random effects (ZIBR) identified a few bacterial genera with differential responses to test product consumption dose compared to control. Shotgun metagenomics analysis revealed a functional contribution to the gut microbiome of probiotic candidates.


2021 ◽  
Vol 8 ◽  
Author(s):  
Yu Tian ◽  
Kai-yi Sun ◽  
Tian-qing Meng ◽  
Zhen Ye ◽  
Shi-meng Guo ◽  
...  

Coronavirus disease 2019 (COVID-19) has infected over 124 million people worldwide. In addition to the development of therapeutics and vaccines, the evaluation of the sequelae in recovered patients is also important. Recent studies have indicated that COVID-19 has the ability to infect intestinal tissues and to trigger alterations of the gut microbiota. However, whether these changes in gut microbiota persist into the recovery stage remains largely unknown. Here, we recruited seven healthy Chinese men and seven recovered COVID-19 male patients with an average of 3-months after discharge and analyzed their fecal samples by 16S rRNA sequencing analysis to identify the differences in gut microbiota. Our results suggested that the gut microbiota differed in male recovered patients compared with healthy controls, in which a significant difference in Chao index, Simpson index, and β-diversity was observed. And the relative abundance of several bacterial species differed clearly between two groups, characterized by enrichment of opportunistic pathogens and insufficiency of some anti-inflammatory bacteria in producing short chain fatty acids. The above findings provide preliminary clues supporting that the imbalanced gut microbiota may not be fully restored in recovered patients, highlighting the importance of continuous monitoring of gut health in people who have recovered from COVID-19.


2020 ◽  
Vol 70 (1) ◽  
Author(s):  
Yinlong Cheng ◽  
Yining Li ◽  
Yonghong Xiong ◽  
Yixin Zou ◽  
Siyu Chen ◽  
...  

Abstract Purpose To investigate the effect of liver-specific knockdown of ANGPTL8 on the structure of the gut microbiota. Methods We constructed mice with liver-specific ANGPTL8 knockdown by using an adeno-associated virus serotype 8 (AAV8) system harbouring an ANGPTL8 shRNA. We analysed the structure and function of the gut microbiome through pyrosequencing and KEGG (Kyoto Encyclopedia of Genes and Genomes) functional prediction. Results Compared with controls, ANGPTL8 shRNA reduced the Simpson index and Shannon index (p < 0.01) of the gut microbiota in mice. At the phylum level, the sh-ANGPTL8 group showed a healthier gut microbiota composition than controls (Bacteroidetes: controls 67.52%, sh-ANGPTL8 80.75%; Firmicutes: controls 10.96%, sh-ANGPTL8 8.58%; Proteobacteria: controls 9.29%, sh-ANGPTL8 0.98%; F/B ratio: controls 0.16, sh-ANGPTL8 0.11). PCoA and UPGMA analysis revealed a significant difference in microbiota composition, while KEGG analysis revealed a significant difference in microbiota function between controls and the sh-ANGPTL8 group. Conclusion Our results revealed that inhibition of ANGPTL8 signalling altered the structure of the gut microbiome, which might further affect the metabolism of mice. We have thus identified ANGPTL8 as a novel hepatogenic hormone potentially involving the liver-gut axis and regulating the structure of the gut microbiota.


2014 ◽  
Vol 112 (S2) ◽  
pp. S44-S49 ◽  
Author(s):  
Devin J. Rose

The gut microbiota plays important roles in proper gut function and can contribute to or help prevent disease. Whole grains, including oats, constitute important sources of nutrients for the gut microbiota and contribute to a healthy gut microbiome. In particular, whole grains provide NSP and resistant starch, unsaturated TAG and complex lipids, and phenolics. The composition of these constituents is unique in oats compared with other whole grains. Therefore, oats may contribute distinctive effects on gut health relative to other grains. Studies designed to determine these effects may uncover new human-health benefits of oat consumption.


2019 ◽  
Author(s):  
Sofie Ingdam Halkjær ◽  
Victoria Elisabeth de Knegt ◽  
Bobby Lo ◽  
Lisbeth Nilas ◽  
Dina Cortes ◽  
...  

Abstract Background: Maternal obesity is associated with adverse pregnancy outcomes. Probiotic supplementation during pregnancy may have positive effects on blood glucose, gestational weight gain (GWG), and the risk of gestational diabetes mellitus (GDM). The primary aim was to determine the feasibility of probiotic intervention in obese pregnant women from the early second trimester until delivery. The secondary aim was to investigate the effect of daily probiotic supplementation on GWG, maternal glucose homeostasis, infant birthweight, and maternal gut microbiota. We carried out a randomized double-blinded placebo-controlled study in 50 obese pregnant women. Participants were randomly allocated to two treatment groups, multi-strain probiotic [Vivomixx®] or placebo at 14–20 weeks of gestation until delivery. Participants were followed with two pre-delivery visits at gestational week 27-30 and 36-37 and with one post-delivery visit 2-3 days after birth. All visits included blood and fecal sampling. An oral glucose tolerance test was performed at inclusion and gestational week 27-30. Results: Forty-nine participants completed the study. Thirty-eight participants took more than 80% of the Vivomixx® capsules (n=21), placebo (n=17). There was no significant difference in HbA1c levels and the occurrence of GDM between groups. There was no significant difference in GWG and infant birth weight between groups in intention to treat analysis. There was, however, a lower mean GWG (11.9 vs 13.0 kg) and lower mean infant birthweight (3554 vs 3658 g) in the probiotic group in the per protocol analysis, due to sample size this difference did not reach statistical significance. Fecal microbiota analyses showed an overall increase in α-diversity over time in the Vivomixx® group only (p=0.016). Conclusions: Administration of probiotics during pregnancy is feasible in obese women. Multi-strain probiotic can modulate the gut microbiota in obese women during pregnancy. A larger study population is needed to uncover whether the results regarding lower GWG and infant birth weight after probiotic supplementation are significant. Trial registration: ClincalTrials.gov Identifier: NCT02508844, registered on May 11, 2015.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ashish Kumar Srivastava ◽  
Vishwajeet Rohil ◽  
Brij Bhushan ◽  
Malleswara Rao Eslavath ◽  
Harshita Gupta ◽  
...  

AbstractShip voyage to Antarctica is a stressful journey for expedition members. The response of human gut microbiota to ship voyage and a feasible approach to maintain gut health, is still unexplored. The present findings describe a 24-day long longitudinal study involving 19 members from 38th Indian Antarctic Expedition, to investigate the impact of ship voyage and effect of probiotic intervention on gut microbiota. Fecal samples collected on day 0 as baseline and at the end of ship voyage (day 24), were analyzed using whole genome shotgun sequencing. Probiotic intervention reduced the sea sickness by 10% compared to 44% in placebo group. The gut microbiome in placebo group members on day 0 and day 24, indicated significant alteration compared to a marginal change in the microbial composition in probiotic group. Functional analysis revealed significant alterations in carbohydrate and amino acid metabolism. Carbohydrate-active enzymes analysis represented functional genes involved in glycoside hydrolases, glycosyltransferases and carbohydrate binding modules, for maintaining gut microbiome homeostasis. Suggesting thereby the possible mechanism of probiotic in stabilizing and restoring gut microflora during stressful ship journey. The present study is first of its kind, providing a feasible approach for protecting gut health during Antarctic expedition involving ship voyage.


2021 ◽  
Vol 8 ◽  
Author(s):  
Gil Zandani ◽  
Sarit Anavi-Cohen ◽  
Nina Tsybina-Shimshilashvili ◽  
Noa Sela ◽  
Abraham Nyska ◽  
...  

Nonalcoholic fatty liver disease (NAFLD) is linked to obesity, type 2 diabetes, hyperlipidemia, and gut dysbiosis. Gut microbiota profoundly affects the host energy homeostasis, which, in turn, is affected by a high-fat diet (HFD) through the liver-gut axis, among others. Broccoli contains beneficial bioactive compounds and may protect against several diseases. This study aimed to determine the effects of broccoli supplementation to an HFD on metabolic parameters and gut microbiome in mice. Male (7–8 weeks old) C57BL/J6 mice were divided into four groups: normal diet (ND), high-fat diet (HFD), high-fat diet+10% broccoli florets (HFD + F), and high-fat diet + 10% broccoli stalks (HFD + S). Liver histology and serum biochemical factors were evaluated. Alterations in protein and gene expression of the key players in lipid and carbohydrate metabolism as well as in gut microbiota alterations were also investigated. Broccoli florets addition to the HFD significantly reduced serum insulin levels, HOMA-IR index, and upregulated adiponectin receptor expression. Conversely, no significant difference was found in the group supplemented with broccoli stalks. Both broccoli stalks and florets did not affect fat accumulation, carbohydrate, or lipid metabolism-related parameters. Modifications in diversity and in microbial structure of proteobacteria strains, Akermansia muciniphila and Mucispirillum schaedleri were observed in the broccoli-supplemented HFD-fed mice. The present study suggests that dietary broccoli alters parameters related to insulin sensitivity and modulates the intestinal environment. More studies are needed to confirm the results of this study and to investigate the mechanisms underlying these beneficial effects.


Author(s):  
Anna L. Beale ◽  
Joanne A. O’Donnell ◽  
Michael E. Nakai ◽  
Shane Nanayakkara ◽  
Donna Vizi ◽  
...  

Background Risk factors for heart failure with preserved ejection fraction (HFpEF) include hypertension, age, sex, and obesity. Emerging evidence suggests that the gut microbiota independently contributes to each one of these risk factors, potentially mediated via gut microbial‐derived metabolites such as short‐chain fatty acids. In this study, we determined whether the gut microbiota were associated with HFpEF and its risk factors. Methods and Results We recruited 26 patients with HFpEF and 67 control participants from 2 independent communities. Patients with HFpEF were diagnosed by exercise right heart catheterization. We assessed the gut microbiome by bacterial 16S rRNA sequencing and food intake by the food frequency questionnaire. There was a significant difference in α‐diversity (eg, number of microbes) and β‐diversity (eg, type and abundance of microbes) between both cohorts of controls and patients with HFpEF ( P =0.001). We did not find an association between β‐diversity and specific demographic or hemodynamic parameters or risk factors for HFpEF. The Firmicutes to Bacteroidetes ratio, a commonly used marker of gut dysbiosis, was lower, but not significantly so ( P =0.093), in the patients with HFpEF. Compared with controls, the gut microbiome of patients with HFpEF was depleted of bacteria that are short‐chain fatty acid producers. Consistent with this, participants with HFpEF consumed less dietary fiber (17.6±7.7 versus 23.2±8.8 g/day; P =0.016). Conclusions We demonstrate key changes in the gut microbiota in patients with HFpEF, including the depletion of bacteria that generate metabolites known to be important for cardiovascular homeostasis. Further studies are required to validate the role of these gut microbiota and metabolites in the pathophysiology of HFpEF.


Nutrients ◽  
2019 ◽  
Vol 11 (12) ◽  
pp. 3006 ◽  
Author(s):  
Silvia Lai ◽  
Alessio Molfino ◽  
Massimo Testorio ◽  
Adolfo M. Perrotta ◽  
Annachiara Currado ◽  
...  

Introduction: The gut microbiota has coevolved with humans for a mutually beneficial coexistence and plays an important role in health and disease. A dysbiotic gut microbiome may contribute to progression to chronic kidney disease (CKD) and CKD-related complications such as cardiovascular disease. Microbiota modulation through the administration of prebiotics may represent an important therapeutic target. Aim: We sought to evaluate the effects of a low-protein diet (LPD) (0.6 g/kg/day) with or without the intake of the prebiotic inulin (19 g/day) on microbiota and clinical parameters in CKD patients. Materials and Methods: We performed a longitudinal, prospective, controlled, and interventional study on 16 patients: 9 patients treated with LPD (0.6 g/kg/day) and inulin (19 g/day) and 7 patients (control group) treated only with LPD (0.6 g/kg/day). Clinical evaluations were performed and fecal samples were collected for a subsequent evaluation of the intestinal microbiota in all patients. These tests were carried out before the initiation of LPD, with or without inulin, at baseline (T0) and at 6 months (T2). The microbiota of 16 healthy control (HC) subjects was also analyzed in order to identify potential dysbiosis between patients and healthy subjects. Results: Gut microbiota of CKD patients was different from that of healthy controls. The LPD was able to significantly increase the frequencies of Akkermansiaceae and Bacteroidaceae and decrease the frequencies of Christensenellaceae, Clostridiaceae, Lactobacillaceae, and Pasteurellaceae. Only Bifidobacteriaceae were increased when the LPD was accompanied by oral inulin intake. We showed a significant reduction of serum uric acid (SUA) and C-reactive protein (CRP) in patients treated with LPD and inulin (p = 0.018 and p = 0.003, respectively), an improvement in SF-36 (physical role functioning and general health perceptions; p = 0.03 and p = 0.01, respectively), and a significant increase of serum bicarbonate both in patients treated with LPD (p = 0.026) or with LPD and inulin (p = 0.01). Moreover, in patients treated with LPD and inulin, we observed a significant reduction in circulating tumor necrosis factor alpha (TNF-α) (p = 0.041) and plasma nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX2) (p = 0.027) levels. We did not find a significant difference in the circulating levels of Interleukin (IL)-1β (p = 0.529) and IL-6 (p = 0.828) in the two groups. Conclusions: LPD, associated or not with inulin, modified gut microbiota and modulated inflammatory and metabolic parameters in patients with CKD. Our results suggest that interventions attempting to modulate the gut microbiome may represent novel strategies to improve clinical outcomes in CKD patients and may provide useful therapeutic effects.


2020 ◽  
Vol 65 (4) ◽  
pp. 278-280 ◽  
Author(s):  
Aadil Bharwani ◽  
Asem Bala ◽  
Michael Surette ◽  
John Bienenstock ◽  
Simone N. Vigod ◽  
...  

Objectives: Compelling animal data exists examining the impact of the gut microbiome on the brain, but work is required to translate these findings in a clinical population. We sought to do this by exploring the effects of antidepressant medications on the gut microbiota, and establishing a baseline Major Depressive Disorder (MDD) gut phenotype. Methods: Participants with a primary diagnosis of MDD (n = 15) who were nonmedicated were recruited and followed over 6 months. Stool samples were collected prior to treatment initiation and 3 and 6 months following treatment. 16S rRNA sequencing was employed in order to analyze the gut microbial community profile. Symptom severity was measured by the Beck Depression Inventory. Alpha diversity metrics revealed no significant difference in the community diversity across any of the time-points. Results: Comparison of within-group versus between-group distances revealed a lack of clustering of samples based on time-point, suggesting no significant change in the microbiota across treatment duration. When analyzed based on treatment response, however, patients in the responder group exhibited greater phylogenetic diversity than non-responders (Mann-Whitney U = 5, p = 0.026). At 3-months, 35 Operational Taxonomic Units (OTUs) were significantly different between groups and at 6-months, 42 OTUs were significantly different between responders and non-responders. Conclusions: These observations indicate that antidepressant medications alter the gut microbiota of patients with MDD, with disparate effects in responders versus non responders. This supports the concept of a microbiota phenotype associate with treatment response in MDD.


Sign in / Sign up

Export Citation Format

Share Document