purehealth research

Free Shipping & Tracking in the USA

365 Day Money Back Guarantee

Premium-Quality Supplements

24/7 Customer Service

Free Shipping & Tracking in the USA

365 Day Money Back Guarantee

Premium-Quality Supplements

24/7 Customer Service

ImproBiome

SCIENTIFIC RESEARCH ON THE FOLLOWING INGREDIENTS:

Inulin from Jerusalem Artichokes and Chicory Root (Lactobacillus)

Effect of Lactobacillus sakei, a Probiotic Derived from Kimchi, on Body Fat in Koreans with Obesity: A Randomized Controlled Study

Abstract

Background: The increased prevalence of obesity has led to increases in the prevalence of chronic diseases worldwide. There is interest whether probiotics have an effect on obesity, but the effectiveness and safety of only a few probiotics for the treatment of obesity have been reported. The purpose of this study was to investigate whether ingestion of Lactobacillus sakei (CJLS03) derived from kimchi causes weight loss in people with obesity.

Methods: This randomized, double-blind, placebo-controlled, clinical trial involved 114 adults with a body mass index (BMI) ≥25 kg/m2 who were assigned randomly to a CJLS03 or placebo group. The groups received two allocations of either 5×109 colony-forming units of CJLS03/allocation or the equivalent vehicle for 12 weeks. Demographic and biochemical parameters, and body composition including fat and muscle mass were measured at baseline and after 12 weeks. Changes in body fat, weight, and waist circumference were compared between the two treatment groups. Adverse events were monitored during study period.

Results: Body fat mass decreased by 0.2 kg in the CJLS03 group and increased by 0.6 kg in the placebo group (0.8 kg difference, P=0.018). After the 12 weeks, waist circumference was 0.8 cm smaller in the CJLS03 group than in the placebo group (P=0.013). BMI and body weight did not change after the 12 weeks. Adverse events were mild and did not differ between the two groups.

Conclusion: These data suggest that L. sakei (CJLS03) might help people with obesity reduce body fat mass without serious side effects (ClinicalTrials.gov: NCT03248414).

Source: Lim S, Moon JH, Shin CM, Jeong D, Kim B. “Effect of Lactobacillus sakei, a Probiotic Derived from Kimchi, on Body Fat in Koreans with Obesity: A Randomized Controlled Study.” Endocrinol Metab (Seoul). (2020);35(2):425-434.

Gut microbiota from twins discordant for obesity modulate metabolism in mice

Abstract

The role of specific gut microbes in shaping body composition remains unclear. We transplanted fecal microbiota from adult female twin pairs discordant for obesity into germ-free mice fed low-fat mouse chow, as well as diets representing different levels of saturated fat and fruit and vegetable consumption typical of the U.S. diet. Increased total body and fat mass, as well as obesity-associated metabolic phenotypes, were transmissible with uncultured fecal communities and with their corresponding fecal bacterial culture collections. Cohousing mice harboring an obese twin's microbiota (Ob) with mice containing the lean co-twin's microbiota (Ln) prevented the development of increased body mass and obesity-associated metabolic phenotypes in Ob cage mates. Rescue correlated with invasion of specific members of Bacteroidetes from the Ln microbiota into Ob microbiota and was diet-dependent. These findings reveal transmissible, rapid, and modifiable effects of diet-by-microbiota interactions.

Source: Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, Muehlbauer MJ, Ilkayeva O, Semenkovich CF, Funai K, Hayashi DK, Lyle BJ, Martini MC, Ursell LK, Clemente JC, Van Treuren W, Walters WA, Knight R, Newgard CB, Heath AC, Gordon JI. “Gut microbiota from twins discordant for obesity modulate metabolism in mice.” Science (2013);341(6150):1241214.

Beneficial metabolic effects of a probiotic via butyrate-induced GLP-1 hormone secretion

Abstract

Obesity and diabetes are associated with excess caloric intake and reduced energy expenditure resulting in a negative energy balance. The incidence of diabetes has reached epidemic proportions, and childhood diabetes and obesity are increasing alarmingly. Therefore, it is important to develop safe, easily deliverable, and economically viable treatment alternatives for these diseases. Here, we provide data supporting the candidacy of probiotics as such a therapeutic modality against obesity and diabetes. Probiotics are live bacteria that colonize the gastrointestinal tract and impart beneficial effects for health. However, their widespread prescription as medical therapies is limited primarily because of the paucity of our understanding of their mechanism of action. Here, we demonstrate that the administration of a probiotic, VSL#3, prevented and treated obesity and diabetes in several mouse models. VSL#3 suppressed body weight gain and insulin resistance via modulation of the gut flora composition. VSL#3 promoted the release of the hormone GLP-1, resulting in reduced food intake and improved glucose tolerance. The VSL#3-induced changes were associated with an increase in the levels of a short chain fatty acid (SCFA), butyrate. Using a cell culture system, we demonstrate that butyrate stimulated the release of GLP-1 from intestinal L-cells, thereby providing a plausible mechanism for VSL#3 action. These findings suggest that probiotics such as VSL#3 can modulate the gut microbiota-SCFA-hormone axis. Moreover, our results indicate that probiotics are of potential therapeutic utility to counter obesity and diabetes.

Source: Yadav H, Lee JH, Lloyd J, Walter P, Rane SG. “Beneficial metabolic effects of a probiotic via butyrate-induced GLP-1 hormone secretion.” J Biol Chem. (2013);288(35):25088-25097.

Decreased fat storage by Lactobacillus paracasei is associated with increased levels of angiopoietin-like 4 protein (ANGPTL4)

Abstract

Background: Intervention strategies for obesity are global issues that require immediate attention. One approach is to exploit the growing consensus that beneficial gut microbiota could be of use in intervention regimes. Our objective was to determine the mechanism by which the probiotic bacteria Lactobacillus paracasei ssp paracasei F19 (F19) could alter fat storage. Angiopoietin-like 4 (ANGPTL4) is a circulating lipoprotein lipase (LPL) inhibitor that controls triglyceride deposition into adipocytes and has been reported to be regulated by gut microbes.

Methodology/principal findings: A diet intervention study of mice fed high-fat chow supplemented with F19 was carried out to study potential mechanistic effects on fat storage. Mice given F19 displayed significantly less body fat, as assessed by magnetic resonance imaging, and a changed lipoprotein profile. Given that previous studies on fat storage have identified ANGPTL4 as an effector, we also investigated circulating levels of ANGPTL4, which proved to be higher in the F19-treated group. This increase, together with total body fat and triglyceride levels told a story of inhibited LPL action through ANGPTL4 leading to decreased fat storage. Co-culture experiments of colonic cell lines and F19 were set up in order to monitor any ensuing alterations in ANGPTL4 expression by qPCR. We observed that potentially secreted factors from F19 can induce ANGPTL4 gene expression, acting in part through the peroxisome proliferator activated receptors alpha and gamma. To prove validity of in vitro findings, germ-free mice were monocolonized with F19. Here we again found changes in serum triglycerides as well as ANGPTL4 in response to F19.

Conclusions/significance: Our results provide an interesting mechanism whereby modifying ANGPTL4, a central player in fat storage regulation, through manipulating gut flora could be an important gateway upon which intervention trials of weight management can be based.

Source: Aronsson L, Huang Y, Parini P, Korach-André M, Håkansson J, Gustafsson JÅ, Pettersson S, Arulampalam V, Rafter J. “Decreased fat storage by Lactobacillus paracasei is associated with increased levels of angiopoietin-like 4 protein (ANGPTL4).” PLoS One. (2010);5(9):e13087.

Short-chain fatty acids suppress food intake by activating vagal afferent neurons

Abstract

Fermentable carbohydrates including dietary fibers and resistant starch produce short-chain fatty acids (SCFAs), including acetate, propionate and butyrate, through microbial fermentation in the intestine of rodents and humans. Consumption of fermentable carbohydrate and SCFAs suppress food intake, an effect involving the brain. However, their signaling pathway to the brain remains unclear. Vagal afferents serve to link intestinal information to the brain. In the present study, we explored possible role of vagal afferents in the anorexigenic effect of SCFAs. Intraperitoneal (ip) injection of three SCFA molecules (6 mmol/kg) suppressed food intake in fasted mice with the rank order of butyrate > propionate > acetate. The suppressions of feeding by butyrate, propionate and acetate were attenuated by vagotomy of hepatic branch and blunted by systemic treatment with capsaicin that denervates capsaicin-sensitive sensory nerves including vagal afferents. Ip injection of butyrate induced significant phosphorylation of extracellular-signal-regulated kinase 1/2, cellular activation markers, in nodose ganglia and their projection site, medial nucleus tractus solitaries. Moreover, butyrate directly interacted with single neurons isolated from nodose ganglia and induced intracellular Ca2+ signaling. The present results identify the vagal afferent as the novel pathway through which exogenous SCFAs execute the remote control of feeding behavior and possibly other brain functions. Vagal afferents might participate in suppression of feeding by intestine-born SCFAs.

Source: Goswami C, Iwasaki Y, Yada T. “Short-chain fatty acids suppress food intake by activating vagal afferent neurons.” J Nutr Biochem. (2018);57:130-135.

Effect of Lactobacillus rhamnosus CGMCC1.3724 supplementation on weight loss and maintenance in obese men and women

Abstract

The present study investigated the impact of a Lactobacillus rhamnosus CGMCC1.3724 (LPR) supplementation on weight loss and maintenance in obese men and women over 24 weeks. In a double-blind, placebo-controlled, randomised trial, each subject consumed two capsules per d of either a placebo or a LPR formulation (1.6 × 10(8) colony-forming units of LPR/capsule with oligofructose and inulin). Each group was submitted to moderate energy restriction for the first 12 weeks followed by 12 weeks of weight maintenance. Body weight and composition were measured at baseline, at week 12 and at week 24. The intention-to-treat analysis showed that after the first 12 weeks and after 24 weeks, mean weight loss was not significantly different between the LPR and placebo groups when all the subjects were considered. However, a significant treatment × sex interaction was observed. The mean weight loss in women in the LPR group was significantly higher than that in women in the placebo group (P = 0.02) after the first 12 weeks, whereas it was similar in men in the two groups (P= 0.53). Women in the LPR group continued to lose body weight and fat mass during the weight-maintenance period, whereas opposite changes were observed in the placebo group. Changes in body weight and fat mass during the weight-maintenance period were similar in men in both the groups. LPR-induced weight loss in women was associated not only with significant reductions in fat mass and circulating leptin concentrations but also with the relative abundance of bacteria of the Lachnospiraceae family in faeces. The present study shows that the Lactobacillus rhamnosus CGMCC1.3724 formulation helps obese women to achieve sustainable weight loss.

Source: Sanchez M, Darimont C, Drapeau V, Emady-Azar S, Lepage M, Rezzonico E, Ngom-Bru C, Berger B, Philippe L, Ammon-Zuffrey C, Leone P, Chevrier G, St-Amand E, Marette A, Doré J, Tremblay A. “Effect of Lactobacillus rhamnosus CGMCC1.3724 supplementation on weight loss and maintenance in obese men and women.” Br J Nutr. (2014);111(8):1507-19.

Effect of Lactobacillus gasseri SBT2055 in fermented milk on abdominal adiposity in adults in a randomised controlled trial

Abstract

Consumption of fermented milk (FM) containing a probiotic, Lactobacillus gasseri SBT2055 (LG2055), previously showed a reduction in abdominal adiposity in a randomised controlled trial (RCT) using FM with 10(8) colony-forming units (cfu) of LG2055/g. However, whether the effectiveness is observed at lower concentrations, the recommended minimum or intermediate levels of probiotics (10(6) or 10(7) cfu/g, respectively), remains to be examined. A multi-centre, double-blind, parallel-group RCT was conducted using 210 healthy Japanese adults with large visceral fat areas (80·2 - 187·8 cm(2)). They were balanced for their baseline characteristics and randomly assigned to three groups receiving FM containing 10(7), 10(6) or 0 (control) cfu LG2055/g of FM, and were asked to consume 200 g FM/d for 12 weeks. Abdominal visceral fat areas, which were determined by computed tomography, at week 12, changed from baseline by an average of -8·5 % (95 % CI -11·9, -5·1; P < 0·01) in the 10(7) dose group, and by -8·2 % (95 % CI -10·8, -5·7; P < 0·01) in the 10(6) dose group. Other measures including BMI, waist and hip circumferences, and body fat mass were also significantly decreased from baseline at week 12 in both groups; interestingly, the cessation of taking FM for 4 weeks attenuated these effects. In the control group, none of these parameters significantly decreased from baseline. These findings demonstrate that consumption of LG2055 at doses as low as the order of 10(8) cfu/d exhibited a significant lowering effect on abdominal adiposity, and suggest that constant consumption might be needed to maintain the effect.

Source: Kadooka Y, Sato M, Ogawa A, Miyoshi M, Uenishi H, Ogawa H, Ikuyama K, Kagoshima M, Tsuchida T. “Effect of Lactobacillus gasseri SBT2055 in fermented milk on abdominal adiposity in adults in a randomised controlled trial.” Br J Nutr. (2013);110(9):1696-703.

Effect of Lactobacillus sakei, a Probiotic Derived from Kimchi, on Body Fat in Koreans with Obesity: A Randomized Controlled Study

Abstract

Background: The increased prevalence of obesity has led to increases in the prevalence of chronic diseases worldwide. There is interest whether probiotics have an effect on obesity, but the effectiveness and safety of only a few probiotics for the treatment of obesity have been reported. The purpose of this study was to investigate whether ingestion of Lactobacillus sakei (CJLS03) derived from kimchi causes weight loss in people with obesity.

Methods: This randomized, double-blind, placebo-controlled, clinical trial involved 114 adults with a body mass index (BMI) ≥25 kg/m2 who were assigned randomly to a CJLS03 or placebo group. The groups received two allocations of either 5×109 colony-forming units of CJLS03/allocation or the equivalent vehicle for 12 weeks. Demographic and biochemical parameters, and body composition including fat and muscle mass were measured at baseline and after 12 weeks. Changes in body fat, weight, and waist circumference were compared between the two treatment groups. Adverse events were monitored during study period.

Results: Body fat mass decreased by 0.2 kg in the CJLS03 group and increased by 0.6 kg in the placebo group (0.8 kg difference, P=0.018). After the 12 weeks, waist circumference was 0.8 cm smaller in the CJLS03 group than in the placebo group (P=0.013). BMI and body weight did not change after the 12 weeks. Adverse events were mild and did not differ between the two groups.

Conclusion: These data suggest that L. sakei (CJLS03) might help people with obesity reduce body fat mass without serious side effects (ClinicalTrials.gov: NCT03248414).

Source: Lim S, Moon JH, Shin CM, Jeong D, Kim B. “Effect of Lactobacillus sakei, a Probiotic Derived from Kimchi, on Body Fat in Koreans with Obesity: A Randomized Controlled Study.” Endocrinol Metab (Seoul). (2020);35(2):425-434.

Single-component versus multicomponent dietary goals for the metabolic syndrome: a randomized trial

Abstract

Background: Few studies have compared diets to determine whether a program focused on 1 dietary change results in collateral effects on other untargeted healthy diet components.

Objective: To evaluate a diet focused on increased fiber consumption versus the multicomponent American Heart Association (AHA) dietary guidelines.

Design: Randomized, controlled trial from June 2009 to January 2014. (ClinicalTrials.gov: NCT00911885).

Setting: Worcester, Massachusetts.

Participants: 240 adults with the metabolic syndrome.

Intervention: Participants engaged in individual and group sessions.

Measurements: Primary outcome was weight change at 12 months.

Results: At 12 months, mean change in weight was -2.1 kg (95% CI, -2.9 to -1.3 kg) in the high-fiber diet group versus -2.7 kg (CI, -3.5 to -2.0 kg) in the AHA diet group. The mean between-group difference was 0.6 kg (CI, -0.5 to 1.7 kg). During the trial, 12 (9.9%) and 15 (12.6%) participants dropped out of the high-fiber and AHA diet groups, respectively (P = 0.55). Eight participants developed diabetes (hemoglobin A1c level ≥6.5%) during the trial: 7 in the high-fiber diet group and 1 in the AHA diet group (P = 0.066).

Limitations: Generalizability is unknown. Maintenance of weight loss after cessation of group sessions at 12 months was not assessed. Definitive conclusions cannot be made about dietary equivalence because the study was powered for superiority.

Conclusion: The more complex AHA diet may result in up to 1.7 kg more weight loss; however, a simplified approach to weight reduction emphasizing only increased fiber intake may be a reasonable alternative for persons with difficulty adhering to more complicated diet regimens.

Source: Ma Y, Olendzki BC, Wang J, Persuitte GM, Li W, Fang H, Merriam PA, Wedick NM, Ockene IS, Culver AL, Schneider KL, Olendzki GF, Carmody J, Ge T, Zhang Z, Pagoto SL. “Single-component versus multicomponent dietary goals for the metabolic syndrome: a randomized trial.” Ann Intern Med. (2015);162(4):248-57.

Synbiotic effect of various prebiotics on in vitro activities of probiotic lactobacilli

Abstract

In the present study, five Lactobacillus strains were evaluated for their viability in presence of different prebiotics viz. inulin, oligofructose, lactulose, raftilose, and honey. The viability of lactobacilli was observed before and after 5 weeks of refrigerated storage. The doubling time varied from 5.2 hrs to 9.6 hrs. The lowest doubling time was for Lactobacillus plantarum M5 followed by L. plantarum Ch1 with inulin. Viability of lactobacilli was greatest with inulin. The growth and viability in presence of prebiotics were found to be strain-specific. Hence, it could be concluded that the addition of prebiotics have a significant effect on probiotics, and hence, a combination of suitable Lactobacillus strain(s) with a specific prebiotic could be a viable probiotic-based functional food approach in administering the beneficial bacteria in-vivo.

Source: Nagpal R, Kaur A. “Synbiotic effect of various prebiotics on in vitro activities of probiotic lactobacilli.” Ecol Food Nutr. (2011);50(1):63-8.

Xylooligosaccharides (XOS) (Bifidobacteria)

Richness of human gut microbiome correlates with metabolic markers

Abstract

We are facing a global metabolic health crisis provoked by an obesity epidemic. Here we report the human gut microbial composition in a population sample of 123 non-obese and 169 obese Danish individuals. We find two groups of individuals that differ by the number of gut microbial genes and thus gut bacterial richness. They contain known and previously unknown bacterial species at different proportions; individuals with a low bacterial richness (23% of the population) are characterized by more marked overall adiposity, insulin resistance and dyslipidaemia and a more pronounced inflammatory phenotype when compared with high bacterial richness individuals. The obese individuals among the lower bacterial richness group also gain more weight over time. Only a few bacterial species are sufficient to distinguish between individuals with high and low bacterial richness, and even between lean and obese participants. Our classifications based on variation in the gut microbiome identify subsets of individuals in the general white adult population who may be at increased risk of progressing to adiposity-associated co-morbidities.

Source: Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, Almeida M, Arumugam M, Batto JM, Kennedy S, Leonard P, Li J, Burgdorf K, Grarup N, Jørgensen T, Brandslund I, Nielsen HB, Juncker AS, Bertalan M, Levenez F, Pons N, Rasmussen S, Sunagawa S, Tap J, Tims S, Zoetendal EG, Brunak S, Clément K, Doré J, Kleerebezem M, Kristiansen K, Renault P, Sicheritz-Ponten T, de Vos WM, Zucker JD, Raes J, Hansen T; MetaHIT consortium, Bork P, Wang J, Ehrlich SD, Pedersen O. “Richness of human gut microbiome correlates with metabolic markers.” Nature. (2013);500(7464):541-6.

Gut Bifidobacteria Populations in Human Health and Aging

Abstract

The intestinal microbiota has increasingly been shown to have a vital role in various aspects of human health. Indeed, several studies have linked alterations in the gut microbiota with the development of different diseases. Among the vast gut bacterial community, Bifidobacterium is a genus which dominates the intestine of healthy breast-fed infants whereas in adulthood the levels are lower but relatively stable. The presence of different species of bifidobacteria changes with age, from childhood to old age. Bifidobacterium longum, B. breve, and B. bifidum are generally dominant in infants, whereas B. catenulatum, B. adolescentis and, as well as B. longum are more prevalent in adults. Increasingly, evidence is accumulating which shows beneficial effects of supplementation with bifidobacteria for the improvement of human health conditions ranging from protection against infection to different extra- and intra-intestinal positive effects. Moreover, bifidobacteria have been associated with the production of a number of potentially health promoting metabolites including short chain fatty acids, conjugated linoleic acid and bacteriocins. The aim of this mini-review is to describe the bifidobacteria compositional changes associated with different stages in life, highlighting their beneficial role, as well as their presence or absence in many disease states.

Source: Arboleya S, Watkins C, Stanton C, Ross RP. “Gut Bifidobacteria Populations in Human Health and Aging.” Front Microbiol. 2016 Aug 19;7:1204.

Effects of daily consumption of the probiotic Bifidobacterium animalis subsp. lactis CECT 8145 on anthropometric adiposity biomarkers in abdominally obese subjects: a randomized controlled trial

Abstract

Background: The effects of probiotic Bifidobacterium animalis subsp. lactis CECT 8145 (Ba8145) and those of its heat-killed form (h-k Ba8145) on human anthropometric adiposity biomarkers are unknown.

Objective: To assess the effect of Ba8145 and h-k Ba8145 ingestion on anthropometric adiposity biomarkers.

Design: Randomized, parallel, double-blind, placebo-controlled trial with abdominally obese individuals. Participants (n = 135) consumed 1 capsule/day containing 1010 colony forming unit (CFU) of Ba8145, 1010 CFU of h-k Ba8145, or placebo (maltodextrin) for 3 months.

Results: Ba8145 ingestion decreased waist circumference, waist circumference/height ratio, and Conicity index (P < 0.05) versus its baseline. Changes versus the placebo group reached significance (P < 0.05) after the h-k Ba8145 treatment. Ba8145 decreased the body mass index compared with baseline and placebo group (P < 0.05). The decrease in visceral fat area after Ba8145 treatments reached significance (P < 0.05) only after h-k Ba8145. When analyses by gender were performed, significance remained only for women. Diastolic blood pressure and HOMA index decreased (P < 0.05) after h-k Ba8145. Gut microbiome analyses showed an increase in Akkermansia spp. after Ba8145 treatment, particularly in the live form, which was inversely related to weight (P = 0.003).

Conclusions: In abdominally obese individuals, consumption of Ba8145, both as viable and mainly as heat-killed cells, improves anthropometric adiposity biomarkers, particularly in women. An increase in the gut Akkermansia genus appears as a possible mechanism involved. Our results support Ba8145 probiotic as a complementary strategy in obesity management.

Source: Pedret A, Valls RM, Calderón-Pérez L, Llauradó E, Companys J, Pla-Pagà L, Moragas A, Martín-Luján F, Ortega Y, Giralt M, Caimari A, Chenoll E, Genovés S, Martorell P, Codoñer FM, Ramón D, Arola L, Solà R. “Effects of daily consumption of the probiotic Bifidobacterium animalis subsp. lactis CECT 8145 on anthropometric adiposity biomarkers in abdominally obese subjects: a randomized controlled trial.” Int J Obes (Lond). (2019);43(9):1863-1868.

A randomised controlled study shows supplementation of overweight and obese adults with lactobacilli and bifidobacteria reduces bodyweight and improves well-being

Abstract

In an exploratory, block-randomised, parallel, double-blind, single-centre, placebo-controlled superiority study (ISRCTN12562026, funded by Cultech Ltd), 220 Bulgarian participants (30 to 65 years old) with BMI 25-34.9 kg/m2 received Lab4P probiotic (50 billion/day) or a matched placebo for 6 months. Participants maintained their normal diet and lifestyle. Primary outcomes were changes in body weight, BMI, waist circumference (WC), waist-to-height ratio (WtHR), blood pressure and plasma lipids. Secondary outcomes were changes in plasma C-reactive protein (CRP), the diversity of the faecal microbiota, quality of life (QoL) assessments and the incidence of upper respiratory tract infection (URTI). Significant between group decreases in body weight (1.3 kg, p < 0.0001), BMI (0.045 kg/m2, p < 0.0001), WC (0.94 cm, p < 0.0001) and WtHR (0.006, p < 0.0001) were in favour of the probiotic. Stratification identified greater body weight reductions in overweight subjects (1.88%, p < 0.0001) and in females (1.62%, p = 0.0005). Greatest weight losses were among probiotic hypercholesterolaemic participants (-2.5%, p < 0.0001) alongside a significant between group reduction in small dense LDL-cholesterol (0.2 mmol/L, p = 0.0241). Improvements in QoL and the incidence rate ratio of URTI (0.60, p < 0.0001) were recorded for the probiotic group. No adverse events were recorded. Six months supplementation with Lab4P probiotic resulted in significant weight reduction and improved small dense low-density lipoprotein-cholesterol (sdLDL-C) profiles, QoL and URTI incidence outcomes in overweight/obese individuals.

Source: Michael DR, Jack AA, Masetti G, Davies TS, Loxley KE, Kerry-Smith J, Plummer JF, Marchesi JR, Mullish BH, McDonald JAK, Hughes TR, Wang D, Garaiova I, Paduchová Z, Muchová J, Good MA, Plummer SF. “A randomised controlled study shows supplementation of overweight and obese adults with lactobacilli and bifidobacteria reduces bodyweight and improves well-being.” Sci Rep. (2020);10(1):4183.

Xylooligosaccharides Increase Bifidobacteria and Lachnospiraceae in Mice on a High-Fat Diet, with a Concomitant Increase in Short-Chain Fatty Acids, Especially Butyric Acid

Abstract

Effects of xylooligosaccharides (XOSs) as well as a mixture of XOS, inulin, oligofructose, and partially hydrolyzed guar gum (MIX) in mice fed a high-fat diet (HFD) were studied. Control groups were fed an HFD or a low-fat diet. Special attention was paid to the cecal composition of the gut microbiota and formation of short-chain fatty acids, but metabolic parameters were also documented. The XOS group had significantly higher cecum levels of acetic, propionic, and butyric acids than the HFD group, and the butyric acid content was higher in the XOS than in the MIX group. The cecum microbiota of the XOS group contained more Bifidobacteria, Lachnospiraceae, and S24-7 bacteria than the HFD group. A tendency of lower body weight gain was observed on comparing the XOS and HFD groups. In conclusion, the XOS was shown to be a promising prebiotic candidate. The fiber diversity in the MIX diet did not provide any advantages compared to the XOS diet.

Source: Berger K, Burleigh S, Lindahl M, Bhattacharya A, Patil P, Stålbrand H, Nordberg Karlsson E, Hållenius F, Nyman M, Adlercreutz P. “Xylooligosaccharides Increase Bifidobacteria and Lachnospiraceae in Mice on a High-Fat Diet, with a Concomitant Increase in Short-Chain Fatty Acids, Especially Butyric Acid.” J Agric Food Chem. (2021);69(12):3617-3625.

Effect of dietary xylooligosaccharides on intestinal characteristics, gut microbiota, cecal short-chain fatty acids, and plasma immune parameters of laying hens

Abstract

This study examined the prebiotic effects of xylooligosaccharides (XOS) on intestinal characteristics, gut microbiota, cecal short-chain fatty acids, plasma calcium metabolism, and immune parameters of laying hens. A total of 1,080 White Lohmann laying hens (28 wk of age) was assigned to 6 dietary treatments that included XOS at concentrations of 0, 0.01, 0.02, 0.03, 0.04, or 0.05% for 8 weeks. Each treatment had 6 replicates with 10 cages (3 birds/cage). Blood, intestinal tissues, and cecal digesta samples were collected from chickens at the end of the experiment. Villus height, crypt depth, the villus to crypt (VH: CD) ratio, and the relative length of different intestinal sections were evaluated. Additionally, the number of microorganisms and the content of short-chain fatty acids in cecal digesta samples were determined. Plasma concentrations of immunoglobulin A (IgA), immunoglobulin G, immunoglobulin M (IgM), interleukin 2 (IL-2), tumor necrosis factor-α(TNF-α), 1, 25-dihydroxyvitamin D3 (1,25(OH)2D3), calcitonin (CT), and parathyroid hormone (PTH) were also determined. The results showed that villus height and the VH: CD ratio of the jejunum were increased (linear, P < 0.01) with the increase in dietary XOS concentration, and the relative length of the jejunum (P = 0.03) was increased significantly in XOS diets. Dietary supplementation of XOS significantly increased (linear, P < 0.01) the number of Bifidobacteria in the cecum; however, total bacteria count, Lactobacillus, and Escherichia coli in the cecum were not affected by XOS supplementation. In addition, inclusion of XOS increased (linear, P < 0.01) the content of butyrate in the cecum; and the content of acetic acid showed a linear increasing trend (P = 0.053) with increasing concentration of XOS in the diets. Supplementation with XOS increased (quadratic, P < 0.05) the content of 1,25(OH)2D3 in plasma. There were no significant differences (P > 0.05) in the content of CT and PTH among dietary treatments. Furthermore, dietary XOS increased contents of IgA (linear, P < 0.05), TNF-α (linear, P < 0.05), IgM (linear, P < 0.05; quadratic, P < 0.05), and IL-2 (quadratic, P < 0.05). Taken together, it was suggested that supplemental XOS can enhance the intestinal health and immune function of laying hens by positively influencing the intestinal characteristics, gut microbiota, cecal short-chain fatty acids, and immune parameters.

Source: Ding XM, Li DD, Bai SP, Wang JP, Zeng QF, Su ZW, Xuan Y, Zhang KY. “Effect of dietary xylooligosaccharides on intestinal characteristics, gut microbiota, cecal short-chain fatty acids, and plasma immune parameters of laying hens.” Poult Sci. (2018);97(3):874-881.

Apple Pectin

Soluble Fermentable Dietary Fibre (Pectin) Decreases Caloric Intake, Adiposity and Lipidaemia in High-Fat Diet-Induced Obese Rats

Abstract

Consumption of a high fat diet promotes obesity and poor metabolic health, both of which may be improved by decreasing caloric intake. Satiety-inducing ingredients such as dietary fibre may be beneficial and this study investigates in diet-induced obese (DIO) rats the effects of high or low fat diet with or without soluble fermentable fibre (pectin). In two independently replicated experiments, young adult male DIO rats that had been reared on high fat diet (HF; 45% energy from fat) were given HF, low fat diet (LF; 10% energy from fat), HF with 10% w/w pectin (HF+P), or LF with 10% w/w pectin (LF+P) ad libitum for 4 weeks (n = 8/group/experiment). Food intake, body weight, body composition (by magnetic resonance imaging), plasma hormones, and plasma and liver lipid concentrations were measured. Caloric intake and body weight gain were greatest in HF, lower in LF and HF+P, and lowest in the LF+P group. Body fat mass increased in HF, was maintained in LF, but decreased significantly in LF+P and HF+P groups. Final plasma leptin, insulin, total cholesterol and triglycerides were lower, and plasma satiety hormone PYY concentrations were higher, in LF+P and HF+P than in LF and HF groups, respectively. Total fat and triglyceride concentrations in liver were greatest in HF, lower in LF and HF+P, and lowest in the LF+P group. Therefore, the inclusion of soluble fibre in a high fat (or low fat) diet promoted increased satiety and decreased caloric intake, weight gain, adiposity, lipidaemia, leptinaemia and insulinaemia. These data support the potential of fermentable dietary fibre for weight loss and improving metabolic health in obesity.

Source: Adam CL, Thomson LM, Williams PA, Ross AW. “Soluble Fermentable Dietary Fibre (Pectin) Decreases Caloric Intake, Adiposity and Lipidaemia in High-Fat Diet-Induced Obese Rats.” PLoS One. 2015 Oct 8;10(10):e0140392.

Apple-Derived Pectin Modulates Gut Microbiota, Improves Gut Barrier Function, and Attenuates Metabolic Endotoxemia in Rats with Diet-Induced Obesity

Abstract

This study was aimed at determining potential effects of apple-derived pectin on weight gain, gut microbiota, gut barrier and metabolic endotoxemia in rat models of diet-induced obesity. The rats received a standard diet (control; Chow group; n = 8) or a high-fat diet (HFD; n = 32) for eight weeks to induce obesity. The top 50th percentile of weight-gainers were selected as diet induced obese rats. Thereafter, the Chow group continued on chow, and the diet induced obese rats were randomly divided into two groups and received HFD (HF group; n = 8) or pectin-supplemented HFD (HF-P group; n = 8) for six weeks. Compared to the HF group, the HF-P group showed attenuated weight gain (207.38 ± 7.96 g vs. 283.63 ± 10.17 g, p < 0.01) and serum total cholesterol level (1.46 ± 0.13 mmol/L vs. 2.06 ± 0.26 mmol/L, p < 0.01). Compared to the Chow group, the HF group showed a decrease in Bacteroidetes phylum and an increase in Firmicutes phylum, as well as subordinate categories (p < 0.01). These changes were restored to the normal levels in the HF-P group. Furthermore, compared to the HF group, the HF-P group displayed improved intestinal alkaline phosphatase (0.57 ± 0.20 vs. 0.30 ± 0.19, p < 0.05) and claudin 1 (0.76 ± 0.14 vs. 0.55 ± 0.18, p < 0.05) expression, and decreased Toll-like receptor 4 expression in ileal tissue (0.76 ± 0.58 vs. 2.04 ± 0.89, p < 0.01). The HF-P group also showed decreased inflammation (TNFα: 316.13 ± 7.62 EU/mL vs. 355.59 ± 8.10 EU/mL, p < 0.01; IL-6: 51.78 ± 2.35 EU/mL vs. 58.98 ± 2.59 EU/mL, p < 0.01) and metabolic endotoxemia (2.83 ± 0.42 EU/mL vs. 0.68 ± 0.14 EU/mL, p < 0.01). These results suggest that apple-derived pectin could modulate gut microbiota, attenuate metabolic endotoxemia and inflammation, and consequently suppress weight gain and fat accumulation in diet induced obese rats.

Source: Jiang T, Gao X, Wu C, Tian F, Lei Q, Bi J, Xie B, Wang HY, Chen S, Wang X. “Apple-Derived Pectin Modulates Gut Microbiota, Improves Gut Barrier Function, and Attenuates Metabolic Endotoxemia in Rats with Diet-Induced Obesity.” Nutrients. (2016);8(3):126.

References:
  1. https://pubmed.ncbi.nlm.nih.gov/32615727/
  2. https://pubmed.ncbi.nlm.nih.gov/24009397/
  3. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3757173/
  4. https://pubmed.ncbi.nlm.nih.gov/20927337/
  5. https://pubmed.ncbi.nlm.nih.gov/29702431/
  6. https://pubmed.ncbi.nlm.nih.gov/24299712/
  7. https://pubmed.ncbi.nlm.nih.gov/23614897/
  8. https://pubmed.ncbi.nlm.nih.gov/32615727/
  9. https://pubmed.ncbi.nlm.nih.gov/25686165/
  10. https://pubmed.ncbi.nlm.nih.gov/21888588/
  11. https://pubmed.ncbi.nlm.nih.gov/23985870/
  12. https://pubmed.ncbi.nlm.nih.gov/27594848/
  13. https://pubmed.ncbi.nlm.nih.gov/30262813/
  14. https://pubmed.ncbi.nlm.nih.gov/32144319/
  15. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8041301/
  16. https://pubmed.ncbi.nlm.nih.gov/29294100/
  17. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4598151/
  18. https://pubmed.ncbi.nlm.nih.gov/26938554/