Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
PeerJ ; 12: e17583, 2024.
Article in English | MEDLINE | ID: mdl-38948211

ABSTRACT

Background: Recent studies suggest that gut microbiota composition, abundance and diversity can influence many chronic diseases such as type 2 diabetes. Modulating gut microbiota through targeted nutrition can provide beneficial effects leading to the concept of personalized nutrition for health improvement. In this prospective clinical trial, we evaluated the impact of a microbiome-based targeted personalized diet on hyperglycaemic and hyperlipidaemic individuals. Specifically, BugSpeaks®-a microbiome profile test that profiles microbiota using next generation sequencing and provides personalized nutritional recommendation based on the individual microbiota profile was evaluated. Methods: A total of 30 participants with type 2 diabetes and hyperlipidaemia were recruited for this study. The microbiome profile of the 15 participants (test arm) was evaluated using whole genome shotgun metagenomics and personalized nutritional recommendations based on their microbiota profile were provided. The remaining 15 participants (control arm) were provided with diabetic nutritional guidance for 3 months. Clinical and anthropometric parameters such as HbA1c, systolic/diastolic pressure, c-reactive protein levels and microbiota composition were measured and compared during the study. Results: The test arm (microbiome-based nutrition) showed a statistically significant decrease in HbA1c level from 8.30 (95% confidence interval (CI), [7.74-8.85]) to 6.67 (95% CI [6.2-7.05]), p < 0.001 after 90 days. The test arm also showed a 5% decline in the systolic pressure whereas the control arm showed a 7% increase. Incidentally, a sub-cohort of the test arm of patients with >130 mm Hg systolic pressure showed a statistically significant decrease of systolic pressure by 14%. Interestingly, CRP level was also found to drop by 19.5%. Alpha diversity measures showed a significant increase in Shannon diversity measure (p < 0.05), after the microbiome-based personalized dietary intervention. The intervention led to a minimum two-fold (Log2 fold change increase in species like Phascolarctobacterium succinatutens, Bifidobacterium angulatum, and Levilactobacillus brevis which might have a beneficial role in the current context and a similar decrease in species like Alistipes finegoldii, and Sutterella faecalis which have been earlier shown to have some negative effects in the host. Overall, the study indicated a net positive impact of the microbiota based personalized dietary regime on the gut microbiome and correlated clinical parameters.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Hyperglycemia , Hypertension , Precision Medicine , Humans , Male , Hypertension/diet therapy , Hypertension/microbiology , Female , Middle Aged , Prospective Studies , Diabetes Mellitus, Type 2/diet therapy , Diabetes Mellitus, Type 2/microbiology , Hyperglycemia/diet therapy , Hyperglycemia/microbiology , Precision Medicine/methods , Inflammation/diet therapy , Proof of Concept Study , Glycated Hemoglobin/metabolism , Glycated Hemoglobin/analysis , Aged , Hyperlipidemias/diet therapy , Hyperlipidemias/blood , Hyperlipidemias/microbiology , Adult , C-Reactive Protein/analysis , C-Reactive Protein/metabolism
2.
Pharmacol Res ; 205: 107232, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38825157

ABSTRACT

Type 3 resistant starch from Canna edulis (Ce-RS3) is an insoluble dietary fiber which could improve blood lipids in animals, but clinically robust evidence is still lacking. We performed a double-blind randomized controlled trial to assess the effects of Ce-RS3 on lipids in mild hyperlipidemia. One hundred and fifteen patients were included followed the recruitment criteria, and were randomly allocated to receive Ce-RS3 or placebo (native starch from Canna edulis) for 12 weeks (20 g/day). In addition to serum lipids, complete blood counts, serum inflammatory factors, antioxidant indexes, and dietary survey, 16 S rRNA sequencing technique was utilized to analyze the gut microbiota alterations. Targeted quantitative metabolomics (TQM) was used to detect metabolite changes. Compared with the placebo, Ce- RS3 significantly decreased levels of total cholesterol, lowdensity lipoprotein cholesterol, and non-high-density lipoprotein cholesterol, and increased the glutathione peroxidase. Based on the 16 S rRNA sequencing, TQM, the correlation analysis, as well as the Kyoto Encyclopedia of Genes (KEGG) and Genomes and Human Metabolome Database (HMDB) analysis, we found that Ce-RS3 could increase the abundances of genera Faecalibacterium and Agathobacter, while reduce the abundances of genera norank_f_Ruminococcaceae and Christensenellaceae_R-7_ group to regulate phenylalanine metabolism, which could reduce the fatty acid biosynthesis and fatty acid elongation in the mitochondria to lower blood lipids. Conclusively, we firstly confirmed the feasibility of Ce-RS3 for clinical application, which presents a novel, effective therapy for the mild hyperlipidemia. (Chictr. org. cn. Clinical study on anti-mild hyperlipidemia of Canna edulis RS3 resistant starch, ID Number: ChiCTR2200062871).


Subject(s)
Gastrointestinal Microbiome , Hyperlipidemias , Humans , Gastrointestinal Microbiome/drug effects , Double-Blind Method , Male , Middle Aged , Hyperlipidemias/drug therapy , Hyperlipidemias/blood , Hyperlipidemias/microbiology , Female , Adult , Lipids/blood , Resistant Starch , Starch , Hypolipidemic Agents/therapeutic use , Hypolipidemic Agents/pharmacology , Aged
3.
Front Cell Infect Microbiol ; 14: 1333145, 2024.
Article in English | MEDLINE | ID: mdl-38812752

ABSTRACT

Objective: To investigate the structure, composition, and functions of the gut microbiota in elderly patients with hyperlipidemia. Methods: Sixteen older patients diagnosed with hyperlipidemia (M group) and 10 healthy, age-matched normal volunteers (N group) were included. These groups were further subdivided by sex into the male normal (NM, n = 5), female normal (NF, n = 5), male hyperlipidemia (MM, n = 8), and female hyperlipidemia (MF, n = 8) subgroups. Stool samples were collected for high-throughput sequencing of 16S rRNA genes. Blood samples were collected for clinical biochemical index testing. Results: Alpha- and beta-diversity analyses revealed that the structure and composition of the gut microbiota were significantly different between the M and N groups. The relative abundances of Bacteroides, Parabacteroides, Blautia, Peptococcus, and Bifidobacterium were significantly decreased, while those of Lactobacillus, Helicobacter, and Desulfovibrio were significantly higher in the M group. There were also significant sex-related differences in microbial structure between the NM and NF groups, and between the MM and MF groups. Through functional prediction with PICRUSt 2, we observed distinct between-group variations in metabolic pathways associated with the gut microbiota and their impact on the functionality of the nervous system. Pearson's correlation coefficient was used as a distance metric to build co-abundance networks. A hypergeometric test was used to detect taxonomies with significant enrichment in specific clusters. We speculated that modules with Muribaculaceae and Lachnospiraceae as the core microbes play an important ecological role in the intestinal microbiota of the M group. The relative intestinal abundances of Agathobacter and Faecalibacterium in the M group were positively correlated with serum triglyceride and low-density lipoprotein levels, while the relative abundance of Bifidobacterium was negatively correlated with the serum lipoprotein a level.


Subject(s)
Bacteria , Feces , Gastrointestinal Microbiome , Hyperlipidemias , RNA, Ribosomal, 16S , Humans , Gastrointestinal Microbiome/genetics , Male , Female , Aged , Hyperlipidemias/microbiology , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , High-Throughput Nucleotide Sequencing , Middle Aged , Aged, 80 and over
4.
Food Chem ; 404(Pt A): 134599, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36444019

ABSTRACT

Ordinary and hyperlipidemic rats were gavaged with lotus seed resistant starch (LRS), and the structure of the small intestinal flora and bile acids composition were determined for four groups of rats to construct a relationship network diagram between different bacterial genera, bile acids and blood lipid profiles, revealing a microbial mechanism for the lipid-lowering effect of LRS in hyperlipidemic rats. LRS inhibited the growth of Romboutsia, Bacillus, Blautia, norank_f__Muribaculaceae and norank_f__Eubacterium_coprostanoligenes_group in hyperlipidemic rats. Meanwhile LRS promoted the production of primary bile acids (CA, CDCA, ß-MCA) and secondary bile acids (LCA, UDCA), and reduced the contents of TCA, Dehydro-LCA, isoLCA, LCA-3-S and THDCA in hyperlipidemic rats. Furthermore, Blautia, norank_f__Muribaculaceae and norank_f__Eubacterium_coprostanoligenes_group were positively correlated with Dehydro-LCA, isoLCA, TCA, LCA-3-S, TCHO, TG and LDL-C. In summary, LRS improves blood lipid levels by regulating small intestinal flora and accelerating the breakdown of cholesterol into bile acids in the liver.


Subject(s)
Hyperlipidemias , Lotus , Resistant Starch , Seeds , Animals , Rats , Bacteroidetes , Bile Acids and Salts , Clostridiales , Gastrointestinal Microbiome/drug effects , Resistant Starch/pharmacology , Seeds/chemistry , Hyperlipidemias/microbiology , Hyperlipidemias/therapy
5.
Gut Microbes ; 14(1): 2003176, 2022.
Article in English | MEDLINE | ID: mdl-34923903

ABSTRACT

Non-fasting lipidemia (nFL), mainly contributed by postprandial lipidemia (PL), has recently been recognized as an important cardiovascular disease (CVD) risk as fasting lipidemia (FL). PL serves as a common feature of dyslipidemia in Type 2 Diabetes (T2D), albeit effective therapies targeting on PL were limited. In this study, we aimed to evaluate whether the therapy combining probiotics (Prob) and berberine (BBR), a proven antidiabetic and hypolipidemic regimen via altering gut microbiome, could effectively reduce PL in T2D and to explore the underlying mechanism. Blood PL (120 min after taking 100 g standard carbohydrate meal) was examined in 365 participants with T2D from the Probiotics and BBR on the Efficacy and Change of Gut Microbiota in Patients with Newly Diagnosed Type 2 Diabetes (PREMOTE study), a random, placebo-controlled, and multicenter clinical trial. Prob+BBR was superior to BBR or Prob alone in improving postprandial total cholesterol (pTC) and low-density lipoprotein cholesterol (pLDLc) levels with decrement of multiple species of postprandial lipidomic metabolites after 3 months follow-up. This effect was linked to the changes of fecal Bifidobacterium breve level responding to BBR alone or Prob+BBR treatment. Four fadD genes encoding long-chain acyl-CoA synthetase were identified in the genome of this B. breve strain, and transcriptionally activated by BBR. In vitro BBR treatment further decreased the concentration of FFA in the culture medium of B. breve compared to vehicle. Thus, the activation of fadD by BBR could enhance FFA import and mobilization in B. breve and diliminish the intraluminal lipids for absorption to mediate the effect of Prob+BBR on PL. Our study confirmed that BBR and Prob (B. breve) could exert a synergistic hypolipidemic effect on PL, acting as a gut lipid sink to achieve better lipidemia and CVD risk control in T2D.


Subject(s)
Berberine/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Hyperlipidemias/drug therapy , Probiotics/administration & dosage , Adult , Animals , Cholesterol/blood , Cholesterol, LDL/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/microbiology , Double-Blind Method , Drug Therapy, Combination , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Humans , Hyperlipidemias/blood , Hyperlipidemias/microbiology , Male , Middle Aged , Postprandial Period/drug effects
6.
Nutrients ; 13(11)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34836140

ABSTRACT

Postprandial hyperglycaemia is associated with increased risk of cardiovascular disease. Recent studies highlight the role of the gut microbiome in influencing postprandial glycaemic (PPG) and lipidaemic (PPL) responses. The authors of this review sought to address the question: "To what extent does individual gut microbiome diversity and composition contribute to PPG and PPL responses?". CINAHL Plus, PubMed, Web of Science, and the Cochrane Central Register of Controlled Trials (CENTRAL) databases were searched from January 2010 to June 2020. Following screening, 22 studies were eligible to be included in the current review. All trials reported analysis of gut microbiome diversity and composition and PPG and/or PPL. Results were reported according to the 'Preferred Reporting Items for Systematic Reviews and Meta-Analysis' (PRISMA) statement. Individual microbiota structure was found to play a key role in determining postprandial metabolic responses in adults and is attributed to a complex interplay of diet, microbiota composition, and metagenomic activity, which may be predicted by metagenomic analysis. Alterations of gut microbiota, namely relative abundance of bacterial phylum Actinobacteria and Proteobacteria, along with Enterobacteriaceae, were associated with individual variation in postprandial glycaemic response in adults. The findings of the current review present new evidence to support a personalised approach to nutritional recommendations and guidance for optimal health, management, and treatment of common metabolic disorders. In conclusion, personalised nutrition approaches based on individual microbial composition may improve postprandial regulation of glucose and lipids, providing a potential strategy to ameliorate cardiometabolic health outcomes.


Subject(s)
Gastrointestinal Microbiome/physiology , Hyperglycemia/microbiology , Hyperlipidemias/microbiology , Nutritional Physiological Phenomena/physiology , Postprandial Period/physiology , Humans
7.
Front Endocrinol (Lausanne) ; 12: 722055, 2021.
Article in English | MEDLINE | ID: mdl-34707567

ABSTRACT

The aim of the study was to investigate the regulatory effects of policosanol on hyperlipidemia, gut microbiota and metabolic status in a C57BL/6 mouse model. A total of 35 C57BL/6 mice were assigned to 3 groups, chow (n=12), high fat diet (HFD, n=12) and HFD+policosanol (n=11), then treated for 18 weeks. Policosanol supplementation significantly reduced serum triglycerides and total cholesterol, as well as the weight of brown adipose tissue (BAT) (p<0.05), without affecting body weight in HFD-fed mice (p>0.05). Combined 16S rRNA gene sequencing and untargeted metabolomic analysis demonstrated that policosanol had regulatory effects on gut microbiota and serum metabolism in mice. In obese mice, policosanol increased the proportion of Bacteroides, decreased the proportion of Firmicutes, and increased the ratio of Bacteroides to Firmicutes (p<0.05). Policosanol promoted lipolysis and thermogenesis process, including tricarboxylic acid (TCA) cycle and pyruvate cycle, correlated with the increasing level of Bacteroides, Parasutterella, and decreasing level of Lactobacillus and Candidatus_Saccharimonas. Moreover, policosanol decreased fatty acid synthase (FAS) in the iWAT of obese mice. Policosanol also increased peroxisome proliferators-activated receptor-γ (PPARγ), uncoupling Protein-1 (UCP-1), peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and PR domain containing 16 (PRDM16) in brown adipose tissue (BAT) obese mice (p<0.05). This study presents the new insight that policosanol may inhibit the synthesis of fatty acids, and promote lipolysis, thermogenesis related gene expression and regulate gut microbiota constituents, which provides potential for policosanol as an antihyperlipidemia functional food additive and provide new evidence for whole grain food to replace refined food.


Subject(s)
Fatty Alcohols/pharmacology , Gastrointestinal Microbiome/drug effects , Hyperlipidemias , Lipid Metabolism/drug effects , Animals , Diet, High-Fat , Gastrointestinal Microbiome/genetics , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Hyperlipidemias/microbiology , Hyperlipidemias/pathology , Lipid Metabolism/genetics , Male , Metabolomics/methods , Metagenomics/methods , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Obesity/metabolism , Obesity/microbiology , Obesity/pathology , Systems Integration , Thermogenesis/drug effects , Thermogenesis/genetics
8.
Sci Rep ; 11(1): 14993, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294791

ABSTRACT

The treatment of periodontitis has numerous positive effects on established chronic health conditions, including cardiovascular disease and diabetes. However, ethical considerations do limit the establishment of human trials to investigate whether periodontitis promotes the early stages of chronic conditions. Therefore, the aim of this study was to investigate whether periodontitis induces endothelial dysfunction in hyperlipidemic apolipoprotein E gene-deficient (ApoE-/-) mice. Forty-five 8-week-old ApoE-/- mice were challenged by oral lavage with Porphyromonas gingivalis and Streptococcus gordonii for 4 weeks. A subgroup of animals (n = 15-17/group) was placed in a metabolic chamber immediately before euthanasia at 4 weeks to measure VO2/CO2 concentrations and voluntary locomotion. In infected and control animals alveolar bone levels were measured by x-ray imaging and endothelial function was determined by measuring endothelial-dependent vasorelaxation of aortic rings. The mRNA expression levels of serum amyloid A and tumor necrosis factor were determined in liver tissues by qRT PCR and protein concentrations in serum by ELISA. Caecal contents were analysed by sequencing to determine changes to the gut microbiota to investigate linkages between microbiome and systemic changes. The results showed that oral lavage of P. gingivalis and S. gordonii for 4 weeks, initiated periodontitis in ApoE-/- mice, similar to the human situation. The oral inflammation was accompanied by a significant increase in mRNA expression of pro-inflammatory mediators serum amyloid A1 and tumor necrosis factor in the liver. Mice with periodontitis also exhibited impaired endothelial-dependent vasorelaxation responses to acetylcholine. This systemic response was connected to increased energy expenditure, locomotion and respiratory quotient. No differences were detected in caecal microbiota between the infected and control animals. Overall, this is the first report that provide evidence that periodontitis induces endothelial dysfunction in mice. Other systemic responses observed in response to the local reaction need further investigation. The study suggests that early prevention of periodontitis may help limit the early stages of endothelial dysfunction that is linked to atherogenesis in humans.


Subject(s)
Apolipoproteins E/genetics , Bacteroidaceae Infections/diagnostic imaging , Hyperlipidemias/genetics , Periodontitis/microbiology , Plaque, Atherosclerotic/diagnostic imaging , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Disease Models, Animal , Energy Metabolism , Gastrointestinal Microbiome , Gene Knockout Techniques , Hyperlipidemias/microbiology , Male , Mice , Periodontitis/diagnostic imaging , Periodontitis/genetics , Phylogeny , Plaque, Atherosclerotic/microbiology , Porphyromonas gingivalis/pathogenicity , Sequence Analysis, RNA , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism , Streptococcus gordonii/pathogenicity , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , X-Rays
9.
Phytomedicine ; 83: 153477, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33545549

ABSTRACT

BACKGROUND: Our previous clinical research showed that the interaction between gut microbiota and bile acids (BAs) in patients with type 2 diabetes mellitus (T2DM) changed significantly. We hypothesized that T2DM could be improved by adjusting this interaction mediated by farnesoid X receptor (FXR). T2DM belongs to the category of "xiaoke" in traditional Chinese medicine. Radix scutellariae has the effects of clearing away heat and eliminating dampness, curing jaundice and quenching thirst and is widely used alone or in combination with other medicines for the treatment of T2DM in China and throughout Asia. Additionally, the interaction between Radix scutellariae and gut microbiota may influence its efficacy in the treatment of T2DM. PURPOSE: This study chose Radix scutellariae to validate that T2DM could improve by adjusting the interaction between gut microbiota and bile acid metabolism. STUDY DESIGN AND METHODS: Radix scutellariae water extract (WESB) was administered to a T2DM rat model established by a high-fat diet combined with streptozotocin. The body weight and blood glucose and insulin levels were measured. The levels of serum lipids, creatinine, uric acid, albumin and total bile acid were also detected. Changes in the pathology and histology of the pancreas, liver and kidney were observed by haematoxylin-eosin staining. The 16S rRNAs of gut microbiota were sequenced, and the faecal and serum BAs were determined by liquid chromatography tandem mass spectrometry. The expression levels of BA metabolism-associated proteins in the liver and intestine were evaluated by immunoblot analysis. RESULTS: The results showed that WESB improved hyperglycaemia, hyperlipaemia, and liver and kidney damage in T2DM rats. In addition, the abundances of key gut microbiota and the concentrations of certain secondary BAs in faeces and serum were restored. Moreover, there was a significant correlation between the restored gut microbiota and BAs, which might be related to the activation of liver cholesterol 7α-hydroxylase (CYP7A1) and the inhibition of FXR expression in the intestine rather than the liver. CONCLUSIONS: This study provided new ideas for the prevention or treatment of clinical diabetes and its complications by adjusting the interaction between gut microbiota and bile acid metabolism.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Microbiome/drug effects , Hyperglycemia/drug therapy , Hyperlipidemias/drug therapy , Scutellaria baicalensis/chemistry , Animals , Cholesterol 7-alpha-Hydroxylase/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/physiology , Hyperglycemia/metabolism , Hyperglycemia/microbiology , Hyperlipidemias/metabolism , Hyperlipidemias/microbiology , Intestines/drug effects , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Rats, Sprague-Dawley
10.
Biomed Pharmacother ; 133: 111014, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33246225

ABSTRACT

BACKGROUND: Intestinal microbiota is a novel drug target of metabolic diseases, especially for those with poor oral bioavailability. Nuciferine, with poor bioavailability, has an anti-hyperlipidemic effect at low dosages. PURPOSE: In the present study, we aimed to explore the role of intestinal microbiota in the anti-hyperlipidemic function of nuciferine and identify the key bacterial targets that might confer the therapeutic actions. METHODS: The contribution of gut microbes in the anti-hyperlipidemic effect of nuciferine was evaluated by conventional and antibiotic-established pseudo-sterile mice. Whole-metagenome shotgun sequencing was used to characterize the changes in microbial communities by various agents. RESULTS: Nuciferine exhibited potent anti-hyperlipidemic and liver steatosis-alleviating effects at the doses of 7.5-30 mg/kg. The beneficial effects of nuciferine were substantially abolished when combined with antibiotics. Metagenomic analysis showed that nuciferine significantly shifted the microbial structure, and the enrichment of Akkermansia muciniphila was closely related to the therapeutic effect of nuciferine. CONCLUSIONS: Our results revealed that gut microbiota played an essential role in the anti-hyperlipidemic effect of nuciferine, and enrichment of Akkermansia muciniphila represented a key mechanism through which nuciferine exerted its therapeutic effects.


Subject(s)
Aporphines/pharmacology , Gastrointestinal Microbiome/drug effects , Hyperlipidemias/drug therapy , Hypolipidemic Agents/pharmacology , Intestines/microbiology , Lipids/blood , Akkermansia/drug effects , Akkermansia/genetics , Akkermansia/growth & development , Animals , Anti-Bacterial Agents/pharmacology , Bacteroides/drug effects , Bacteroides/genetics , Bacteroides/growth & development , Biomarkers/blood , Diet, High-Fat , Disease Models, Animal , Hyperlipidemias/blood , Hyperlipidemias/microbiology , Male , Metagenome , Metagenomics , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/blood , Obesity/microbiology , Obesity/prevention & control , RNA-Seq
11.
J Microbiol Biotechnol ; 31(2): 181-188, 2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33144552

ABSTRACT

Bacillus subtilis and Enterococcus faecium are commonly used probiotics. This study aimed to identify the effect of live combined Bacillus subtilis R0179 and Enterococcus faecium R0026 (LCBE) on obesityassociated hyperlipidemia and gut microbiota in C57BL/6 mice. Forty male C57BL/6 mice were divided into four groups: normal group (N group), model group (M group), low-dose group (L group), and high-dose group (H group). Mice were gavaged with LCBE at 0.023 g/mice/day (L group) or 0.23 g/mice/day (H group) and fed with a high-fat diet for 8 weeks. In vitro E. faecium R0026 showed an ability to lower the low-concentration of cholesterol by 46%, and the ability to lower the highconcentration of cholesterol by 58%. LCBE significantly reduced the body weight gain, Lee index, brown fat index and body mass index of mice on a high-fat diet. Moreover, LCBE markedly improved serum lipids (including serum triglyceride, total cholesterol, low-density lipoprotein and highdensity lipoprotein) while also significantly reducing liver total cholesterol. Serum lipopolysaccharide and total bile acid in L and H groups decreased significantly compared with M group. PCR-DGGE analysis showed that the composition of gut microbiota in the treatment groups was improved. Akkermansia muciniphila was found in H group. The PCA result indicated a similar gut microbiota structure between LCBE treatment groups and normal group while the number of bands and Shannon diversity index increased significantly in the LCBE treatment groups. Finally, qPCR showed Bifidobacterium spp. increased significantly in H group compared with M group, LCBE alleviated liver steatosis and improved brown adipose tissue index.


Subject(s)
Bacillus subtilis/physiology , Enterococcus faecium/physiology , Gastrointestinal Microbiome/drug effects , Hyperlipidemias/prevention & control , Obesity/complications , Probiotics/administration & dosage , Animals , Cholesterol/metabolism , Humans , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/microbiology , Lipid Metabolism/drug effects , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Weight Gain/drug effects
12.
Nutrients ; 12(10)2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33007825

ABSTRACT

The human gut microbiota is involved in host health and disease development. Therefore, lifestyle-related diseases such as hypertension (HT), hyperlipidemia (HL), and type 2 diabetes mellitus (T2D) may alter the composition of gut microbiota. Here, we investigated gut microbiota changes related to these diseases and their coexistence. This study involved 239 Japanese subjects, including healthy controls (HC). The fecal microbiota was analyzed through the isolation of bacterial genomic DNA obtained from fecal samples. Although there were no significant differences in the microbial structure between groups, there was a significant difference in the α-diversity between HC and the patients in whom two diseases coexisted. Moreover, Actinobacteria levels were significantly increased, whereas Bacteroidetes levels were significantly decreased in all disease groups. At the genus level, Bifidobacterium levels were significantly increased in the HL and T2D groups, as were those of Collinsella in all disease groups. In contrast, Alistipes levels were significantly lower in the HL group. Furthermore, metabolic enzyme families were significantly increased in all disease groups. Interestingly, the structure and function of the gut microbiota showed similar profiles in all the studied diseases. In conclusion, several changes in the structure of the gut microbiota are associated with T2D, HT, and HL in Japanese subjects.


Subject(s)
Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome , Hyperlipidemias/microbiology , Hypertension/microbiology , Actinobacteria/isolation & purification , Adolescent , Adult , Aged , Bacteroidetes/isolation & purification , Bifidobacterium/isolation & purification , Case-Control Studies , DNA, Bacterial/isolation & purification , Feces/microbiology , Female , Humans , Japan , Male , Middle Aged , Young Adult
13.
J Agric Food Chem ; 68(42): 11735-11746, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-32985184

ABSTRACT

The mechanisms of highland barley whole grain (BWG) with rich phenolics on obese db/db mice were investigated in this study. Oral consumption of BWG reduced food intake, body weight, organ/body weight indexes of liver and fat, levels of serum and hepatic lipids, liver injury, and oxidative stress. Furthermore, BWG recovered the disorder of cecal microbiota by augmenting the Bacteroidetes/Firmicutes ratio and Alistipes abundance and decreasing the abundances of Bacteroides and Desulfovibrionaceae to modulate lipid metabolism-related genes. BWG inhibited fatty acid biosynthesis via upregulating the phosphorylation of AMP-activated protein kinase α, while downregulating sterol regulatory element binding protein-1c, fatty acid synthase (FAS), and stearoyl-CoA desaturase 1 levels. BWG also significantly downregulated miRNA-122, miRNA-33, miRNA-34a, and miRNA-206 levels. Accordingly, BWG exhibited hypolipidemic potential through modulating cecal microbiota, AMPK/SREBP-1c/FAS pathway, and related miRNAs, triggering the alleviation of dyslipidemia. These findings suggested BWG as an effective candidate to ameliorate the symptoms of hyperlipidemia.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cecum/microbiology , Gastrointestinal Microbiome , Hordeum/metabolism , Hyperlipidemias/diet therapy , Receptors, Leptin/deficiency , AMP-Activated Protein Kinases/genetics , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Cecum/metabolism , Disease Models, Animal , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Humans , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Hyperlipidemias/microbiology , Liver/metabolism , Male , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Receptors, Leptin/genetics , Seeds/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
14.
J Agric Food Chem ; 68(24): 6530-6543, 2020 Jun 17.
Article in English | MEDLINE | ID: mdl-32383865

ABSTRACT

This study aimed to investigate the protective mechanism of common buckwheat (Fagopyrum esculentum Moench.) against nonalcoholic fatty liver disease (NAFLD) associated with dyslipidemia in mice that were fed a high-fat and high-cholesterol diet (HFD). Results showed that oral supplementation of common buckwheat significantly improved physiological indexes and biochemical parameters related to dyslipidemia and NAFLD in mice fed with HFD. Furthermore, the HFD-induced reductions in fecal short-chain fatty acids were reversed by common buckwheat intervention, which also increased the fecal bile acid (BA) abundance compared with HFD-induced hyperlipidemic mice. Liver metabolomics based on ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry demonstrated that common buckwheat supplementation made significant regulatory effects on the pentose phosphate pathway, starch and sucrose metabolism, primary BA biosynthesis, and so forth. The results of high-throughput sequencing revealed that common buckwheat supplementation significantly altered the structure of the intestinal microbiota in mice fed with HFD. The correlations between lipid metabolic parameters and intestinal microbial phylotypes were also revealed by the heatmap and network. Additionally, common buckwheat intervention regulated the mRNA expressions of genes responsible for liver lipid metabolism and BA homeostasis, thus promoting BA synthesis and excretion. These findings confirmed that common buckwheat has the outstanding ability of improving lipid metabolism and could be used as a potential functional food for the prevention of NAFLD and hyperlipidemia.


Subject(s)
Cholesterol/metabolism , Fagopyrum/metabolism , Hyperlipidemias/diet therapy , Non-alcoholic Fatty Liver Disease/diet therapy , Animals , Bile Acids and Salts/metabolism , Cholesterol/adverse effects , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome , Humans , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/microbiology , Liver/metabolism , Male , Mice , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/microbiology
15.
Int J Mol Med ; 45(5): 1601-1615, 2020 May.
Article in English | MEDLINE | ID: mdl-32323749

ABSTRACT

Hyperlipidemia is a major risk factor for cardiovascular diseases. Simvastatin (SV), a cholesterol­lowering agent, has been widely used in the treatment of hyperlipidemia. Gut microbiota is known to influence drug response, including that to statins. However, the effect of SV on the gut microbiota of hyperlipidemic rats is not fully understood. To investigate the influence of SV on gut microbiota in hyperlipidemic rats, the molecular characterization of gut microbiota and the potential functions of genes involved in the downstream metabolic pathways were analyzed using high­throughput sequencing technology and the Phylogenetic Investigation of Communities by Reconstruction of Unobserved States approach. The results revealed that SV treatment could reduce the gut microbial diversity and drive marked remodeling of the fecal bacterial community composition. At the phylum level, the relative abundance of Firmicutes and Actinobacteria was decreased following SV therapy, whereas that of Bacteroidetes was elevated. At the genus level, the percentage of the genera Bacteroides, Sutterella and Phascolarctobacterium was significantly increased, but that of Bifidobacterium, Ruminococcaceae_NK4A214, Ruminococcaceae_UCG­009, Intestinimonas and Tyzzerella was significantly decreased. Additionally, functional prediction analysis indicated that in the SV­associated microbiota, genes involved in energy, carbohydrate, amino acid and nucleotide metabolism likely exhibited enrichment. Briefly, to the best of our knowledge, the present study was the first to establish a profound and comprehensive association between the SV­induced alterations of the gut flora and the consequent influences of downstream metabolic pathways by gut microbiota. These findings suggested that the gut microbiota may contribute to the SV hypolipidemic efficacy in the progression of hyperlipidemia, which could provide insights for the prevention and treatment of hyperlipidemia.


Subject(s)
Gastrointestinal Microbiome/drug effects , Hyperlipidemias/drug therapy , Hyperlipidemias/microbiology , Simvastatin/pharmacology , Animals , Diet , Feces/microbiology , Male , Metabolic Networks and Pathways/drug effects , RNA, Ribosomal, 16S/metabolism , Rats , Rats, Sprague-Dawley
16.
J Agric Food Chem ; 68(1): 147-159, 2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31826616

ABSTRACT

This study was aimed at investigating the hypoglycemic and hypolipidemic effects of a polysaccharide (RTFP) isolated from Rosa roxburghii Tratt fruit on type-2 diabetic db/db mice. The results indicated that the oral administration of RTFP could significantly decrease the body weight, fat, and liver hypertrophy and the levels of fasting blood glucose, serum insulin, and serum lipids of the db/db mice. Histopathological observation showed that RTFP could effectively protect the pancreas, liver, and epididymal fat against damage and dysfunction. Real-time quantitative polymerase chain reaction analysis confirmed that the gene expression levels of peroxisome proliferator-activated receptors-γ (PPAR-γ), sterol regulatory element-binding protein-1 (SREBP-1c), acetyl-CoA carboxylase-1 (ACC-1), fatty acid synthase (FAS), and glucose-6-phosphatase (G6 Pase) were significantly down-regulated in the liver of db/db mice after treatment with RTFP. Moreover, RTFP treatment reversed gut dysbiosis by lowering the Firmicutes-to-Bacteroidetes ratio and enhancing the relative abundances of beneficial bacteria including Bacteroidaceae, Bacteroidaceae S24-7 group, and Lactobacillaceae. These findings suggest that RTFP can be used as a promising functional supplement for the prevention and treatment of type-2 diabetes mellitus.


Subject(s)
Colon/microbiology , Hyperglycemia/drug therapy , Hyperlipidemias/drug therapy , Hypoglycemic Agents/administration & dosage , Hypolipidemic Agents/administration & dosage , Plant Extracts/administration & dosage , Polysaccharides/administration & dosage , Rosa/chemistry , Animals , Colon/metabolism , Disease Models, Animal , Fruit/chemistry , Gastrointestinal Microbiome , Humans , Hyperglycemia/metabolism , Hyperglycemia/microbiology , Hyperlipidemias/metabolism , Hyperlipidemias/microbiology , Liver/drug effects , Liver/metabolism , Male , Mice , Triglycerides/metabolism
17.
J Agric Food Chem ; 68(5): 1186-1197, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31855431

ABSTRACT

A bioactive polysaccharide from microalga Chlorella pyrenoidosa (CPP) was successively prepared via DEAE-52 and G-100 columns. Nuclear magnetic resonance analysis showed that the main glycosidic bonds were composed of 1,2-linked-α-l-Fucp, 1,4-linked-α-l-Rhap, 1,4-linked-ß-l-Araf, 1-linked-α-d-Glcp, 1,3-linked-ß-d-GlcpA, 1,4-linked-ß-d-Xylp, and 1,3,6-linked-ß-d-Manp. Its molecular weight was 5.63 × 106 Da. The hypolipidemic effect and intestinal flora regulation of CPP on diet-induced rats were evaluated through histopathology and biochemistry analyses. CPP could improve plasma and liver lipid metabolism and accelerate the metabolism of the cecal total bile acids and short-chain fatty acids. CPP has also upregulated the adenosine-monophosphate-activated protein kinase α and downregulated the acetyl-CoA carboxylase, sterol regulatory element-binding protein 1c, and ß-hydroxy ß-methylglutaryl-CoA expressions. Moreover, with the 16S rRNA gene sequencing, it was revealed that the composition of intestinal flora changed drastically after treatment, such as the bloom of Coprococcus_1, Lactobacillus, and Turicibacter, whereas there was a strong reduction of the [Ruminococcus]_gauvreauii_group. The above results illustrated that CPP might be served as an effective ingredient to ameliorate lipid metabolism disorders and intestinal flora in hyperlipidemia rats.


Subject(s)
Chlorella/chemistry , Gastrointestinal Microbiome/drug effects , Hyperlipidemias/drug therapy , Microalgae/chemistry , Plant Extracts/administration & dosage , Polysaccharides/administration & dosage , Animals , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Humans , Hyperlipidemias/metabolism , Hyperlipidemias/microbiology , Lipid Metabolism/drug effects , Male , Plant Extracts/chemistry , Polysaccharides/chemistry , Rats , Rats, Wistar
18.
Biochem Biophys Res Commun ; 520(3): 507-513, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31610917

ABSTRACT

Atherosclerosis is exacerbated by periodontal pathogens, which induce vascular inflammation after entering the bloodstream. Among oral indigenous bacteria, Streptococcus sanguinis and S. anginosus are related to systemic disorders, such as infective endocarditis and abscess, and are sometimes detected in human atherosclerotic plaques or blood. Thus, these oral streptococci may contribute to the progression of atherosclerosis. To test this hypothesis, apolipoprotein E-deficient spontaneously hyperlipidemic mice were intraorally challenged with S. sanguinis or S. anginosus. Atherosclerotic plaque formation increased significantly in the S. sanguinis-challenged group compared with the carboxymethylcellulose-treated control group. Expression levels of mRNAs of proinflammatory cytokines in the aorta and levels of atherosclerosis-related mediators in blood increased upon S. sanguinis challenge. Adaptor molecule TNF receptor-associated factor 6 was also enhanced in the aorta when mice were challenged with S. sanguinis. Furthermore, challenge with S. anginosus induced systemic inflammation, but inflammation-related mRNA expression levels in the aorta only increased slightly and were accompanied by minimal expansion of the lesion area. By contrast, with the exception of IL-1α, the expression levels of inflammation-related genes did not change in gingival tissues of both bacteria- and sham-challenged groups. These results reveal that S. sanguinis causes aortic inflammation that leads to accelerated progression of atherosclerosis.


Subject(s)
Aorta/microbiology , Atherosclerosis/microbiology , Hyperlipidemias/microbiology , Inflammation/microbiology , Streptococcal Infections/physiopathology , Streptococcus , Administration, Oral , Animals , Aorta/physiopathology , Cytokines/metabolism , Disease Progression , Gingiva/microbiology , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1alpha/metabolism , Male , Mice , Mice, Inbred BALB C , Mouth/microbiology , Plaque, Atherosclerotic/microbiology , Streptococcus anginosus , Streptococcus sanguis , TNF Receptor-Associated Factor 6/metabolism
19.
J Oleo Sci ; 68(9): 909-922, 2019.
Article in English | MEDLINE | ID: mdl-31484903

ABSTRACT

The objective of this research was to evaluate the effect of wheat gluten on gut microbiota from hamsters and also analyse whether alterations in microbiota could result in wheat gluten's lipid-lowering properties. Four weeks male hamsters were divided into 3 groups (n=10). Two hypercholesterolemic groups were fed for 35 days with hypercholesterolemic diet, containing 20% (w/w) wheat gluten or casein. Wheat gluten significantly reduced serum total cholesterol (TC), low density lipoprotein cholesterol (LDL-C) concentrations, and also decreased the liver total cholesterol (TC), free cholesterol (FC), cholesterol ester (CE), triglycerides (TG) concentrations. Wheat gluten group had a higher fecal lipids, total cholesterol (TC) and bile acids (BA) than that of casein group (p < 0.05). Moreover, wheat gluten significantly increased total short-chain fatty acids (SCFA) concentrations in feces. Sequencing of 16S rRNA gene revealed that intake of wheat gluten decreased the relative abundances of Firmicutes and Erysipelotrichaceae, but to increased the relative abundances of Bateroidetes, Bacteroidales_S24-7_group and Ruminococcaceae. The lipid lowering properties of wheat gluten was associated with the lower ratio of Firmicutes/Bateroidetes, the lower of the bacterial taxa Erysipelotrichaceae and the higher of the bacterial taxa Bacteroidales_S24-7_group and Ruminococcaceae. These results suggest that wheat gluten modulate cholesterol metabolism by altering intestinal microflora.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol, LDL/metabolism , Gastrointestinal Microbiome/drug effects , Glutens/pharmacology , Hyperlipidemias/microbiology , Triticum/chemistry , Animals , Cholesterol, LDL/blood , Fatty Acids, Volatile/metabolism , Feces/microbiology , Gastrointestinal Microbiome/genetics , Male , Mesocricetus , RNA, Ribosomal, 16S/analysis
20.
PLoS One ; 14(7): e0219010, 2019.
Article in English | MEDLINE | ID: mdl-31269076

ABSTRACT

Medicine-food homology is a long-standing concept in traditional Chinese medicine. YiNianKangBao (YNKB) tea is a medicine-food formulation based on Sichuan dark tea (Ya'an Tibetan tea), which is traditionally used for its lipid-lowering properties. In this study, we evaluated the effects of YNKB on dyslipidemia and investigated the mechanism underlying its correlation with gut microbiota and serum metabolite regulation. Wild-type mice were fed a normal diet as a control. Male ApoE-/- mice were randomly divided into three high-fat diet (HFD) groups, a model group, and two treated groups (100, 400 mg/kg/d for low, high-dose), and fed by gavage for 12 weeks. Serum lipid levels, composition of gut microbiota, and serum metabolites were then analyzed before treatment with YNKB. We extracted the ingredients of YNKB in boiled water for one hour. YNKB supplementation at a high dose of 400 mg/kg/day reduced bodyweight gains (relative epididymal fat pad and liver weight), and markedly attenuated serum lipid profiles and atherosclerosis index, with no significant differences present between the low-dose treatment and HFD groups. Gut microbiota and serum metabolic analysis indicated that significant differences were observed between normal, HFD, and YNKB treatment groups. These differences in gut microbiota exhibited strong correlations with dyslipidemia-related indexes and serum metabolite levels. Oral administration of high-dose YNKB also showed significant lipid-lowering activity against hyperlipidemia in apoE-deficient mice, which might be associated with composition alterations of the gut microbiota and changes in serum metabolite abundances. These findings highlight that YNKB as a medicine-food formulation derived from Sichuan dark tea could prevent dyslipidemia and improve the understanding of its mechanisms and the pharmacological rationale for preventive use.


Subject(s)
Gastrointestinal Microbiome , Hyperlipidemias/diet therapy , Teas, Herbal , Teas, Medicinal , Animals , Body Weight , Diet, High-Fat/adverse effects , Drugs, Chinese Herbal , Gastrointestinal Microbiome/genetics , Humans , Hyperlipidemias/blood , Hyperlipidemias/microbiology , Lipids/blood , Male , Metabolomics , Mice , Mice, Knockout, ApoE , Non-alcoholic Fatty Liver Disease/diet therapy , Non-alcoholic Fatty Liver Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL