Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell ; 173(7): 1742-1754.e17, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29906449

ABSTRACT

Osmotic diarrhea is a prevalent condition in humans caused by food intolerance, malabsorption, and widespread laxative use. Here, we assess the resilience of the gut ecosystem to osmotic perturbation at multiple length and timescales using mice as model hosts. Osmotic stress caused reproducible extinction of highly abundant taxa and expansion of less prevalent members in human and mouse microbiotas. Quantitative imaging revealed decimation of the mucus barrier during osmotic perturbation, followed by recovery. The immune system exhibited temporary changes in cytokine levels and a lasting IgG response against commensal bacteria. Increased osmolality prevented growth of commensal strains in vitro, revealing one mechanism contributing to extinction. Environmental availability of microbiota members mitigated extinction events, demonstrating how species reintroduction can affect community resilience. Our findings (1) demonstrate that even mild osmotic diarrhea can cause lasting changes to the microbiota and host and (2) lay the foundation for interventions that increase system-wide resilience.


Subject(s)
Diarrhea/pathology , Gastrointestinal Microbiome/drug effects , Polyethylene Glycols/pharmacology , Animals , Bacteroidetes/drug effects , Bacteroidetes/genetics , Bacteroidetes/isolation & purification , Cecum/chemistry , Cecum/metabolism , Cecum/microbiology , Cecum/pathology , Colon/chemistry , Colon/microbiology , Colon/pathology , Cytokines/metabolism , Diarrhea/immunology , Diarrhea/microbiology , Diarrhea/veterinary , Feces/microbiology , Glycoside Hydrolases/metabolism , Humans , Immunity, Humoral/drug effects , Immunoglobulin G/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Metagenomics , Mice , Osmolar Concentration , Polyethylene Glycols/metabolism , Proteome/analysis , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/genetics , Verrucomicrobia/drug effects , Verrucomicrobia/genetics , Verrucomicrobia/isolation & purification
2.
FASEB J ; 33(6): 7274-7288, 2019 06.
Article in English | MEDLINE | ID: mdl-30857422

ABSTRACT

Alcoholic beverages, which are consumed widely in most parts of the world, have long been identified as a major risk factor for all liver diseases, particularly alcoholic liver disease (ALD). Recent compositional analyses suggest that Chinese baijiu (CB), a clear alcoholic liquid distilled from fermented grains, contains large amounts of small molecule bioactive compounds in addition to a significant amount of ethanol (EtOH). Here, in an experimental mouse model, we show that CB caused lower degrees of liver injury than pure EtOH by protecting against the decrease of the relative abundance of Akkermansia and increase of the relative abundance of Prevotella in the gut, thereby preventing the destruction of the intestinal barrier. Furthermore, we demonstrated that EtOH-induced alteration of the gut microbiota profoundly affected the host metabolome. Compared with EtOH feeding, CB feeding resulted in higher concentrations of functional saturated long-chain fatty acids and short-chain fatty acids. The additional mouse models of low dosages of EtOH and of blending baijiu validated that volatile compounds in CB can attenuate EtOH-induced liver damages. Our results provide supporting evidence that ALD was profoundly influenced by host-gut microbiota metabolic interactions and that small molecule organic compounds in CB could attenuate ALD.-Fang, C., Du, H., Zheng, X., Zhao, A., Jia, W., Xu, Y. Solid-state fermented Chinese alcoholic beverage (baijiu) and ethanol resulted in distinct metabolic and microbiome responses.


Subject(s)
Alcoholic Beverages , Dysbiosis/chemically induced , Ethanol/pharmacology , Gastrointestinal Microbiome/drug effects , Liver Diseases, Alcoholic/etiology , Liver/drug effects , Metabolome/drug effects , Alcoholic Beverages/toxicity , Animals , Bacterial Translocation/drug effects , Distillation , Dysbiosis/metabolism , Dysbiosis/microbiology , Ethanol/toxicity , Fatty Acids/metabolism , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/microbiology , Fermentation , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/microbiology , Male , Mice , Mice, Inbred C57BL , Random Allocation , Ribotyping , Specific Pathogen-Free Organisms , Thiobarbituric Acid Reactive Substances/analysis , Verrucomicrobia/drug effects , Verrucomicrobia/isolation & purification
3.
Molecules ; 25(3)2020 Feb 05.
Article in English | MEDLINE | ID: mdl-32033507

ABSTRACT

The allicin diallyldisulfid-S-oxide, a major garlic organosulfur compound (OSC) in crushed garlic (Allium sativum L.), possesses antibacterial effects, and influences gut bacteria. In this study, we made allicin-free garlic (AFG) extract and investigated its effects on gut microbiome. C57BL/6N male mice were randomly divided into 6 groups and fed normal diet (ND) and high-fat diet (HFD) supplemented with or without AFG in concentrations of 1% and 5% for 11 weeks. The genomic DNAs of feces were used to identify the gut microbiome by sequencing 16S rRNA genes. The results revealed that the ratio of p-Firmicutes to p-Bacteroidetes increased by aging and HFD was reduced by AFG. In particular, the f-Lachnospiraceae, g-Akkermansia, and g-Lactobacillus decreased by aging and HFD was enhanced by AFG. The g-Dorea increased by aging and HFD decreased by AFG. In addition, the ratio of glutamic-pyruvic transaminase to glutamic-oxaloacetic transaminase (GPT/GOT) in serum was significantly increased in the HFD group and decreased by AFG. In summary, our data demonstrated that dietary intervention with AFG is a potential way to balance the gut microbiome disturbed by a high-fat diet.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dietary Supplements , Garlic/chemistry , Gastrointestinal Microbiome/drug effects , Plant Extracts/pharmacology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bacteroidetes/drug effects , Bacteroidetes/isolation & purification , Diet, High-Fat , Disulfides , Firmicutes/drug effects , Firmicutes/isolation & purification , Garlic/genetics , Male , Mice , Mice, Inbred C57BL , Sulfinic Acids/analysis , Verrucomicrobia/drug effects , Verrucomicrobia/isolation & purification
4.
Ecotoxicol Environ Saf ; 171: 92-98, 2019 Apr 30.
Article in English | MEDLINE | ID: mdl-30597321

ABSTRACT

Cadmium (Cd) is an environmental pollutant that poses serious health hazards. Due to the increasing contamination of aquatic systems with Cd, the increased accumulation of Cd in fish has become a food safety and public health concern. The present study was conducted to investigate the effects of waterborne Cd exposure on the microbial community composition and diversity in the gut of common carp. Common carp were exposed to three waterborne Cd concentrations (0, 50 and 500 µg Cd L-1) for 4 weeks. Our results indicated that Cd exposure profoundly affected the composition of the gut microbiota in the common carp. At the phylum level, Saccharibacteria were detected in only the 0 µg and 50 µg Cd L-1 exposure groups, and the abundance of Fusobacteria decreased with increasing Cd concentration, while the abundance of Firmicutes increased with increasing Cd concentration. At the genus level, Cetobacterium was the dominant group in the gut of the common carp, and the abundance of Cetobacterium decreased after Cd exposure. Notably, the abundance of Akkermansia muciniphila, a probiotic, was found to decrease after Cd exposure, and the proportions of some Cd-resistant bacteria were found to increase following Cd exposure. Our results also demonstrated that Cd exposure decreased the community diversity of the gut microbiota. These results suggest that Cd exposure may impact the gut homeostasis of common carp and further affect the health of the organism.


Subject(s)
Bacteria/drug effects , Cadmium/toxicity , Carps , Gastrointestinal Microbiome/drug effects , Water Pollutants, Chemical/toxicity , Animals , Biodiversity , Firmicutes/drug effects , Fusobacteria/drug effects , Intestines/microbiology , Verrucomicrobia/drug effects
5.
Gut ; 67(5): 891-901, 2018 05.
Article in English | MEDLINE | ID: mdl-28550049

ABSTRACT

OBJECTIVE: Alcoholic liver disease (ALD) is a global health problem with limited therapeutic options. Intestinal barrier integrity and the microbiota modulate susceptibility to ALD. Akkermansia muciniphila, a Gram-negative intestinal commensal, promotes barrier function partly by enhancing mucus production. The aim of this study was to investigate microbial alterations in ALD and to define the impact of A. muciniphila administration on the course of ALD. DESIGN: The intestinal microbiota was analysed in an unbiased approach by 16S ribosomal DNA (rDNA) sequencing in a Lieber-DeCarli ALD mouse model, and faecal A. muciniphila abundance was determined in a cohort of patients with alcoholic steatohepatitis (ASH). The impact of A. muciniphila on the development of experimental acute and chronic ALD was determined in a preventive and therapeutic setting, and intestinal barrier integrity was analysed. RESULTS: Patients with ASH exhibited a decreased abundance of faecal A. muciniphila when compared with healthy controls that indirectly correlated with hepatic disease severity. Ethanol feeding of wild-type mice resulted in a prominent decline in A. muciniphila abundance. Ethanol-induced intestinal A. muciniphila depletion could be restored by oral A. muciniphila supplementation. Furthermore, A. muciniphila administration when performed in a preventive setting decreased hepatic injury, steatosis and neutrophil infiltration. A. muciniphila also protected against ethanol-induced gut leakiness, enhanced mucus thickness and tight-junction expression. In already established ALD, A. muciniphila used therapeutically ameliorated hepatic injury and neutrophil infiltration. CONCLUSION: Ethanol exposure diminishes intestinal A. muciniphila abundance in both mice and humans and can be recovered in experimental ALD by oral supplementation. A. muciniphila promotes intestinal barrier integrity and ameliorates experimental ALD. Our data suggest that patients with ALD might benefit from A. muciniphila supplementation.


Subject(s)
Ethanol/adverse effects , Gastrointestinal Microbiome/physiology , Liver Diseases, Alcoholic/microbiology , Verrucomicrobia/drug effects , Adult , Aged , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Feces/microbiology , Female , Fluorescent Antibody Technique , Gastrointestinal Microbiome/genetics , Humans , Immunohistochemistry , Liver , Male , Mice , Mice, Inbred C57BL , Middle Aged , Verrucomicrobia/physiology
6.
Mar Drugs ; 16(5)2018 May 17.
Article in English | MEDLINE | ID: mdl-29772753

ABSTRACT

Recently, accumulating evidence has suggested that Enteromorpha clathrata polysaccharide (ECP) could contribute to the treatment of diseases. However, as a promising candidate for marine drug development, although ECP has been extensively studied, less consideration has been given to exploring its effect on gut microbiota. In this light, given the critical role of gut microbiota in health and disease, we investigated here the effect of ECP on gut microbiota using 16S rRNA high-throughput sequencing. As revealed by bioinformatic analyses, ECP considerably changed the structure of the gut microbiota and significantly promoted the growth of probiotic bacteria in C57BL/6J mice. However, interestingly, ECP exerted different effects on male and female microbiota. In females, ECP increased the abundances of Bifidobacterium spp. and Akkermansia muciniphila, a next-generation probiotic bacterium, whereas in males, ECP increased the population of Lactobacillus spp. Moreover, by shaping a more balanced structure of the microbiota, ECP remarkably reduced the antigen load from the gut in females. Altogether, our study demonstrates for the first time a prebiotic effect of ECP on gut microbiota and forms the basis for the development of ECP as a novel gut microbiota modulator for health promotion and disease management.


Subject(s)
Aquatic Organisms/metabolism , Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , Polysaccharides/pharmacology , Ulva/metabolism , Acute-Phase Proteins/immunology , Administration, Oral , Animals , Bifidobacterium/drug effects , Bifidobacterium/isolation & purification , Carrier Proteins/blood , Carrier Proteins/immunology , Computational Biology , Dietary Supplements , Disease Models, Animal , Dysbiosis/blood , Dysbiosis/immunology , Female , Humans , Lactobacillus/drug effects , Lactobacillus/isolation & purification , Male , Membrane Glycoproteins/blood , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Polysaccharides/isolation & purification , Polysaccharides/therapeutic use , Specific Pathogen-Free Organisms , Verrucomicrobia/drug effects , Verrucomicrobia/isolation & purification
7.
BMC Genomics ; 18(1): 800, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-29047329

ABSTRACT

BACKGROUND: Akkermansia muciniphila is one of the most dominant bacteria that resides on the mucus layer of intestinal tract and plays key role in human health, however, little is known about its genomic content. RESULTS: Herein, we for the first time characterized the genomic architecture of A. muciniphila based on whole-genome sequencing, assembling, and annotating of 39 isolates derived from human and mouse feces. We revealed a flexible open pangenome of A. muciniphila currently consisting of 5644 unique proteins. Phylogenetic analysis identified three species-level A. muciniphila phylogroups exhibiting distinct metabolic and functional features. Based on the comprehensive genome catalogue, we reconstructed 106 newly A. muciniphila metagenome assembled genomes (MAGs) from available metagenomic datasets of human, mouse and pig gut microbiomes, revealing a transcontinental distribution of A. muciniphila phylogroups across mammalian gut microbiotas. Accurate quantitative analysis of A. muciniphila phylogroups in human subjects further demonstrated its strong correlation with body mass index and anti-diabetic drug usage. Furthermore, we found that, during their mammalian gut evolution history, A. muciniphila acquired extra genes, especially antibiotic resistance genes, from symbiotic microbes via recent lateral gene transfer. CONCLUSIONS: The genome repertoire of A. muciniphila provided insights into population structure, evolutionary and functional specificity of this significant bacterium.


Subject(s)
Gastrointestinal Microbiome/genetics , Mammals/microbiology , Verrucomicrobia/genetics , Verrucomicrobia/physiology , Whole Genome Sequencing , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Evolution, Molecular , Humans , Mice , Molecular Sequence Annotation , Verrucomicrobia/drug effects
8.
Microb Pathog ; 106: 171-181, 2017 May.
Article in English | MEDLINE | ID: mdl-26875998

ABSTRACT

Akkermansia muciniphila is an intestinal bacterium that was isolated a decade ago from a human fecal sample. Its specialization in mucin degradation makes it a key organism at the mucosal interface between the lumen and host cells. Although it was isolated quite recently, it has rapidly raised significant interest as A. muciniphila is the only cultivated intestinal representative of the Verrucomicrobia, one of the few phyla in the human gut that can be easily detected in phylogenetic and metagenome analyses. There has also been a growing interest in A. muciniphila, due to its association with health in animals and humans. Notably, reduced levels of A. muciniphila have been observed in patients with inflammatory bowel diseases (mainly ulcerative colitis) and metabolic disorders, which suggests it may have potential anti-inflammatory properties. The aims of this review are to summarize the existing data on the intestinal distribution of A. muciniphila in health and disease, to provide insight into its ecology and its role in founding microbial networks at the mucosal interface, as well as to discuss recent research on its role in regulating host functions that are disturbed in various diseases, with a specific focus on metabolic disorders in both animals and humans.


Subject(s)
Gastrointestinal Microbiome , Intestines/microbiology , Verrucomicrobia/physiology , Animals , Anti-Bacterial Agents/therapeutic use , Clinical Studies as Topic , Feces/microbiology , Gastrointestinal Microbiome/immunology , Humans , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/physiology , Metabolic Diseases/microbiology , Metagenome , Mice , Models, Animal , Obesity/microbiology , Phylogeny , Verrucomicrobia/classification , Verrucomicrobia/drug effects , Verrucomicrobia/growth & development
9.
Nutr Cancer ; 69(6): 943-951, 2017.
Article in English | MEDLINE | ID: mdl-28718724

ABSTRACT

Natural compounds can alter the diversity and composition of the gut microbiome, potentially benefiting our health. We previously demonstrated chemopreventive effects of black raspberries (BRBs) in colorectal cancer, which is associated with gut dysbiosis. To investigate the effects of whole BRBs and their fractions on gut microbiota, we fed F-344 rats a control diet, 5% BRBs, the BRB anthocyanin fraction, or the BRB residue fraction for 6 weeks. Feces were collected at baseline and at weeks 3 and 6, and bacterial sequence counts were analyzed. We observed distinct patterns of microbiota from different diet groups. Beta diversity analysis suggested that all diet groups exerted time-dependent changes in the bacterial diversity. Hierarchical clustering analysis revealed that post-diet fecal microbiota was segregated from baseline fecal microbiota within each diet. It is interesting to note that fractions of BRBs induced different changes in gut bacteria compared to whole BRBs. The abundance of specific microbial species known to have anti-inflammatory effects, such as Akkermansia and Desulfovibrio, was increased by whole BRBs and their residue. Further, butyrate-producing bacteria, e.g., Anaerostipes, were increased by whole BRBs. Our results suggest that whole BRBs and their fractions alter the gut microbiota in ways that could significantly influence human health.


Subject(s)
Anthocyanins/pharmacology , Dietary Fiber/pharmacology , Gastrointestinal Microbiome/drug effects , Rubus/chemistry , Animals , Anthocyanins/analysis , Clostridiales/drug effects , Clostridiales/isolation & purification , Desulfovibrio/drug effects , Desulfovibrio/isolation & purification , Diet , Dietary Fiber/analysis , Feces/microbiology , Fruit/chemistry , Rats , Rats, Inbred F344 , Verrucomicrobia/drug effects , Verrucomicrobia/isolation & purification
10.
Gut ; 64(6): 872-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25080446

ABSTRACT

OBJECTIVE: The increasing prevalence of obesity and type 2 diabetes (T2D) demonstrates the failure of conventional treatments to curb these diseases. The gut microbiota has been put forward as a key player in the pathophysiology of diet-induced T2D. Importantly, cranberry (Vaccinium macrocarpon Aiton) is associated with a number of beneficial health effects. We aimed to investigate the metabolic impact of a cranberry extract (CE) on high fat/high sucrose (HFHS)-fed mice and to determine whether its consequent antidiabetic effects are related to modulations in the gut microbiota. DESIGN: C57BL/6J mice were fed either a chow or a HFHS diet. HFHS-fed mice were gavaged daily either with vehicle (water) or CE (200 mg/kg) for 8 weeks. The composition of the gut microbiota was assessed by analysing 16S rRNA gene sequences with 454 pyrosequencing. RESULTS: CE treatment was found to reduce HFHS-induced weight gain and visceral obesity. CE treatment also decreased liver weight and triglyceride accumulation in association with blunted hepatic oxidative stress and inflammation. CE administration improved insulin sensitivity, as revealed by improved insulin tolerance, lower homeostasis model assessment of insulin resistance and decreased glucose-induced hyperinsulinaemia during an oral glucose tolerance test. CE treatment was found to lower intestinal triglyceride content and to alleviate intestinal inflammation and oxidative stress. Interestingly, CE treatment markedly increased the proportion of the mucin-degrading bacterium Akkermansia in our metagenomic samples. CONCLUSIONS: CE exerts beneficial metabolic effects through improving HFHS diet-induced features of the metabolic syndrome, which is associated with a proportional increase in Akkermansia spp.


Subject(s)
Enteritis/drug therapy , Enteritis/microbiology , Insulin Resistance , Obesity, Abdominal/prevention & control , Plant Extracts/pharmacology , Vaccinium macrocarpon/chemistry , Verrucomicrobia/drug effects , Animals , Diet, High-Fat/adverse effects , Endotoxemia/etiology , Endotoxemia/prevention & control , Hepatitis/prevention & control , Homeostasis/drug effects , Intestines/microbiology , Lipid Metabolism/drug effects , Lipids/blood , Lipopolysaccharides/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Obesity, Abdominal/etiology , Organ Size/drug effects , Polyphenols/analysis , Polyphenols/pharmacology , Triglycerides/metabolism , Verrucomicrobia/isolation & purification
11.
Carcinogenesis ; 36(11): 1388-96, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26320104

ABSTRACT

Gastrointestinal tumor growth is thought to be promoted by gastrointestinal bacteria and their inflammatory products. We observed that intestine-specific conditional Apc mutant mice (FabplCre;Apc (15lox/+)) developed many more colorectal tumors under conventional than under pathogen-low housing conditions. Shotgun metagenomic sequencing plus quantitative PCR analysis of feces DNA revealed the presence of two bacterial species in conventional mice, absent from pathogen-low mice. One, Helicobacter typhlonius, has not been associated with cancer in man, nor in immune-competent mice. The other species, mucin-degrading Akkermansia muciniphila, is abundantly present in healthy humans, but reduced in patients with inflammatory gastrointestinal diseases and in obese and type 2 diabetic mice. Eradication of H.typhlonius in young conventional mice by antibiotics decreased the number of intestinal tumors. Additional presence of A.muciniphila prior to the antibiotic treatment reduced the tumor number even further. Colonization of pathogen-low FabplCre;Apc (15lox/+) mice with H.typhlonius or A.muciniphila increased the number of intestinal tumors, the thickness of the intestinal mucus layer and A.muciniphila colonization without H.typhlonius increased the density of mucin-producing goblet cells. However, dual colonization with H.typhlonius and A.muciniphila significantly reduced the number of intestinal tumors, the mucus layer thickness and goblet cell density to that of control mice. By global microbiota composition analysis, we found a positive association of A.muciniphila, and of H.typhlonius, and a negative association of unclassified Clostridiales with increased tumor burden. We conclude that A.muciniphila and H.typhlonius can modulate gut microbiota composition and intestinal tumor development in mice.


Subject(s)
Anti-Bacterial Agents/pharmacology , Helicobacter Infections/complications , Helicobacter/drug effects , Intestinal Neoplasms/microbiology , Verrucomicrobia/drug effects , Amoxicillin/pharmacology , Animals , Carcinogenesis , Clarithromycin/pharmacology , Drug Therapy, Combination , Female , Gastrointestinal Microbiome , Goblet Cells/microbiology , Helicobacter Infections/drug therapy , Intestinal Neoplasms/prevention & control , Intestines/microbiology , Intestines/pathology , Male , Metronidazole/pharmacology , Mice, Inbred C57BL , Omeprazole/pharmacology
12.
Appl Environ Microbiol ; 80(19): 5935-43, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25038099

ABSTRACT

Metformin is commonly used as the first line of medication for the treatment of metabolic syndromes, such as obesity and type 2 diabetes (T2D). Recently, metformin-induced changes in the gut microbiota have been reported; however, the relationship between metformin treatment and the gut microbiota remains unclear. In this study, the composition of the gut microbiota was investigated using a mouse model of high-fat-diet (HFD)-induced obesity with and without metformin treatment. As expected, metformin treatment improved markers of metabolic disorders, including serum glucose levels, body weight, and total cholesterol levels. Moreover, Akkermansia muciniphila (12.44%±5.26%) and Clostridium cocleatum (0.10%±0.09%) abundances increased significantly after metformin treatment of mice on the HFD. The relative abundance of A. muciniphila in the fecal microbiota was also found to increase in brain heart infusion (BHI) medium supplemented with metformin in vitro. In addition to the changes in the microbiota associated with metformin treatment, when other influences were controlled for, a total of 18 KEGG metabolic pathways (including those for sphingolipid and fatty acid metabolism) were significantly upregulated in the gut microbiota during metformin treatment of mice on an HFD. Our results demonstrate that the gut microbiota and their metabolic pathways are influenced by metformin treatment.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diet, High-Fat/adverse effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Microbiota/drug effects , Animals , Biomarkers/metabolism , Blood Glucose/drug effects , Body Weight/drug effects , Clostridium/drug effects , Clostridium/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 2/drug therapy , Feces/microbiology , Female , Gastrointestinal Tract/microbiology , Male , Mice , Mice, Inbred C57BL , Microbiota/physiology , Obesity/drug therapy , Verrucomicrobia/drug effects , Verrucomicrobia/metabolism
13.
Nutrients ; 13(11)2021 Nov 06.
Article in English | MEDLINE | ID: mdl-34836220

ABSTRACT

Grapes provide a rich source of polyphenols and fibers. This study aimed to evaluate the effect of the daily consumption of 46 g of whole grape powder, providing the equivalent of two servings of California table grapes, on the gut microbiome and cholesterol/bile acid metabolism in healthy adults. This study included a 4-week standardization to a low-polyphenol diet, followed by 4 weeks of 46 g of grape powder consumption while continuing the low-polyphenol diet. Compared to the baseline, 4 weeks of grape powder consumption significantly increased the alpha diversity index of the gut microbiome. There was a trend of increasing Verrucomicrobia (p = 0.052) at the phylum level, and a significant increase in Akkermansia was noted. In addition, there was an increase in Flavonifractor and Lachnospiraceae_UCG-010, but a decrease in Bifidobacterium and Dialister at the genus level. Grape powder consumption significantly decreased the total cholesterol by 6.1% and HDL cholesterol by 7.6%. There was also a trend of decreasing LDL cholesterol by 5.9%, and decreasing total bile acid by 40.9%. Blood triglyceride levels and body composition were not changed by grape powder consumption. In conclusion, grape powder consumption significantly modified the gut microbiome and cholesterol/bile acid metabolism.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol/metabolism , Gastrointestinal Microbiome/drug effects , Plant Extracts/administration & dosage , Vitis/chemistry , Adult , Akkermansia/drug effects , Bifidobacterium/drug effects , Cholesterol/blood , Female , Healthy Volunteers , Humans , Male , Middle Aged , Pilot Projects , Polyphenols/metabolism , Powders , Triglycerides/blood , Verrucomicrobia/drug effects , Young Adult
14.
Nat Commun ; 11(1): 4822, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32973149

ABSTRACT

Abiraterone acetate (AA) is an inhibitor of androgen biosynthesis, though this cannot fully explain its efficacy against androgen-independent prostate cancer. Here, we demonstrate that androgen deprivation therapy depletes androgen-utilizing Corynebacterium spp. in prostate cancer patients and that oral AA further enriches for the health-associated commensal, Akkermansia muciniphila. Functional inferencing elucidates a coinciding increase in bacterial biosynthesis of vitamin K2 (an inhibitor of androgen dependent and independent tumor growth). These results are highly reproducible in a host-free gut model, excluding the possibility of immune involvement. Further investigation reveals that AA is metabolized by bacteria in vitro and that breakdown components selectively impact growth. We conclude that A. muciniphila is a key regulator of AA-mediated restructuring of microbial communities, and that this species may affect treatment response in castrate-resistant cohorts. Ongoing initiatives aimed at modulating the colonic microbiota of cancer patients may consider targeted delivery of poorly absorbed selective bacterial growth agents.


Subject(s)
Abiraterone Acetate/pharmacology , Gastrointestinal Microbiome/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms/drug therapy , Verrucomicrobia/drug effects , Abiraterone Acetate/metabolism , Abiraterone Acetate/therapeutic use , Akkermansia , Androgen Antagonists/pharmacology , Androgens/metabolism , Bacteria/metabolism , Feces/microbiology , Humans , Male , RNA, Ribosomal, 16S/genetics , Verrucomicrobia/genetics , Verrucomicrobia/metabolism , Vitamin K 2/metabolism , Vitamin K 2/pharmacology
15.
Int J Biol Macromol ; 163: 1393-1402, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32755709

ABSTRACT

In this study, the effect of Ziziphus jujuba Mill var. spinosa seeds (ZSS) polysaccharides on the bioavailability of spinosin in mice and its molecular mechanism were investigated. After continuously fed with ZSS polysaccharides 100 mg/kg·d-1 for 28 consecutive days, the C57BL/6 mice absorbed spinosin at an obvious lower level compared with the control group. The expression levels of P-gp, MRP2 and Occludin in the colon were significantly increased. ZSS polysaccharides significantly regulated the composition of the gut microbiota, reducing the abundance of Bacteroidetes, and increasing the richness of Firmicutes and Verrucomicrobia. Moreover, ZSS polysaccharides can significantly regulate the expression levels of tight junction proteins and efflux transporters in Caco-2 cells. However, the gut microbiota culture supernatant showed no obvious biological activity in this regard. Furthermore, histopathological analysis revealed ZSS polysaccharides can alleviate TNBS-induced colitis, reduced inflammatory cell infiltration in mice. This immune regulation was related to the NF-κB and MAPK pathways in RAW264.7 cells.


Subject(s)
Colitis/prevention & control , Flavonoids/metabolism , Polysaccharides/pharmacology , Seeds/chemistry , Ziziphus/chemistry , Animals , Bacteroidetes/drug effects , Biological Availability , Caco-2 Cells , Cell Line, Tumor , Colitis/microbiology , Firmicutes/drug effects , Gastrointestinal Microbiome/drug effects , Humans , Inflammation/microbiology , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Verrucomicrobia/drug effects
16.
Sci Rep ; 10(1): 15054, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32929122

ABSTRACT

This study examined the genetic mutation and toxicant exposure in producing gut microbiota alteration and neurotoxicity. Homozygous α-synuclein mutant (SNCA) mice that overexpress human A53T protein and littermate wild-type mice received a single injection of LPS (2 mg/kg) or a selective norepinephrine depleting toxin DSP-4 (50 mg/kg), then the motor activity, dopaminergic neuron loss, colon gene expression and gut microbiome were examined 13 months later. LPS and DSP-4 decreased rotarod and wirehang activity, reduced dopaminergic neurons in substantia nigra pars compacta (SNpc), and SNCA mice were more vulnerable. SNCA mice had 1,000-fold higher human SNCA mRNA expression in the gut, and twofold higher gut expression of NADPH oxidase (NOX2) and translocator protein (TSPO). LPS further increased expression of TSPO and IL-6 in SNCA mice. Both LPS and DSP-4 caused microbiome alterations, and SNCA mice were more susceptible. The altered colon microbiome approximated clinical findings in PD patients, characterized by increased abundance of Verrucomicrobiaceae, and decreased abundance of Prevotellaceae, as evidenced by qPCR with 16S rRNA primers. The Firmicutes/Bacteroidetes ratio was increased by LPS in SNCA mice. This study demonstrated a critical role of α-synuclein and toxins interactions in producing gut microbiota disruption, aberrant gut pro-inflammatory gene expression, and dopaminergic neuron loss.


Subject(s)
Benzylamines/toxicity , Gastrointestinal Microbiome/drug effects , Lipopolysaccharides/toxicity , Neurotoxins/toxicity , alpha-Synuclein/genetics , Animals , Bacteroidetes/drug effects , Benzylamines/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Motor Activity , Mutation, Missense , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Neurotoxins/pharmacology , Norepinephrine/metabolism , Receptors, GABA/genetics , Receptors, GABA/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Verrucomicrobia/drug effects , alpha-Synuclein/metabolism
17.
Gut Microbes ; 11(5): 1348-1361, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32372706

ABSTRACT

The understanding of the effects of compounds on the gut microbiome is limited. In particular, it is unclear whether structurally similar compounds would have similar or distinct effects on the gut microbiome. Here, we selected berberine (BBR), an isoquinoline quaternary alkaloid, and 16 structural analogs and evaluated their effects on seven individual gut microbiomes cultured in vitro. The responses of the individual microbiomes were evaluated by metaproteomic profiles and by assessing butyrate production. We show that both interindividual differences and compound treatments significantly contributed to the variance of metaproteomic profiles. BBR and eight analogs led to changes in proteins involved in microbial defense and stress responses and enrichment of proteins from Verrucomicrobia, Proteobacteria, and Bacteroidetes phyla. It also led to a decrease in proteins from the Firmicutes phylum and its Clostridiales order which correlated to decrease proteins involved in the butyrate production pathway and butyrate concentration. Three of the compounds, sanguinarine, chelerythrine, and ethoxysanguinarine, activated bacterial protective mechanisms, enriched Proteobacteria, increased opacity proteins, and markedly reduced butyrate production. Dihydroberberine had a similar function to BBR in enriching the Akkermansia genus. In addition, it showed less overall adverse impacts on the functionality of the gut microbiome, including a better maintenance of the butyrate level. Our study shows that ex vivo microbiome assay can assess differential regulating effects of compounds with subtle differences and reveals that compound analogs can have distinct effects on the microbiome.


Subject(s)
Bacteroidetes/drug effects , Berberine/analogs & derivatives , Berberine/pharmacology , Firmicutes/drug effects , Gastrointestinal Microbiome/drug effects , Proteobacteria/drug effects , Verrucomicrobia/drug effects , Bacterial Proteins/metabolism , Bacteroidetes/growth & development , Bacteroidetes/metabolism , Berberine/chemistry , Biological Variation, Population , Butyrates/metabolism , Firmicutes/growth & development , Firmicutes/metabolism , Gastrointestinal Microbiome/physiology , Humans , Metabolic Networks and Pathways , Molecular Structure , Proteobacteria/growth & development , Proteobacteria/metabolism , Proteomics , Verrucomicrobia/growth & development , Verrucomicrobia/metabolism
18.
J Ethnopharmacol ; 247: 112299, 2020 Jan 30.
Article in English | MEDLINE | ID: mdl-31606537

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Hua-Feng-Dan (HFD) is a traditional Chinese medicine used for neurological disorders. HFD contains cinnabar (HgS) and realgar (As4S4). The ethnopharmacological basis of cinnabar and realgar in HFD is not known. AIM OF THE STUDY: To address the role of cinnabar and realgar in HFD-produced neuroprotection against neurodegenerative diseases and disturbance of gut microbiota. MATERIALS AND METHODS: Lipopolysaccharide (LPS) plus rotenone (ROT)-elicited rat dopaminergic (DA) neuronal damage loss was performed as a Parkinson's disease animal model. Rats were given a single injection of LPS. Four months later, rats were challenged with the threshold dose of ROT. The clinical dose of HFD was administered via feed, starting from ROT administration for 46 days. Behavioral dysfunction was detected by rotarod and Y-maze tests. DA neuron loss and microglial activation were assessed via immunohistochemical staining and western bolt analysis. The colon content was collected to extract bacterial DNA followed by real-time PCR analysis with 16S rRNA primers. RESULTS: LPS plus ROT induced neurotoxicity, as evidenced by DA neuron loss in substantia nigra, impaired behavioral functions and increased microglial activation. HFD-original (containing 10% cinnabar and 10% realgar) rescued loss of DA neurons, improved behavioral dysfunction and attenuated microglial activation. Compared with HFD-original, HFD-reduced (3% cinnabar and 3% realgar) was also effective, but to be a less extent, while HFD-removed (without cinnabar and realgar) was ineffective. In analysis of gut microbiome, the increased Verrucomicrobiaceae and Lactobacteriaceae, and the decreased Enterobacteeriaceae by LPS plus ROT were ameliorated by HFD-original, and to be the less extent by HFD-reduced. CONCLUSION: Cinnabar and realgar are active ingredients in HFD to exert beneficial effects in a neurodegenerative model and gut microbiota.


Subject(s)
Arsenicals/pharmacology , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Mercury Compounds/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Sulfides/pharmacology , Animals , Arsenicals/chemistry , Arsenicals/therapeutic use , DNA, Bacterial/isolation & purification , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Enterobacteriaceae/drug effects , Enterobacteriaceae/genetics , Enterobacteriaceae/isolation & purification , Ethnopharmacology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Humans , Inflammation Mediators/metabolism , Lactobacillaceae/drug effects , Lactobacillaceae/genetics , Lactobacillaceae/isolation & purification , Lipopolysaccharides/toxicity , Male , Mercury Compounds/chemistry , Mercury Compounds/therapeutic use , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Nerve Degeneration , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , RNA, Ribosomal, 16S/genetics , Rats , Rotenone/toxicity , Sulfides/chemistry , Sulfides/therapeutic use , Verrucomicrobia/drug effects , Verrucomicrobia/genetics , Verrucomicrobia/isolation & purification
19.
Nutrients ; 11(7)2019 Jul 11.
Article in English | MEDLINE | ID: mdl-31336737

ABSTRACT

Akkermansia muciniphila and Faecalibacterium prausnitzii are highly abundant human gut microbes in healthy individuals, and reduced levels are associated with inflammation and alterations of metabolic processes involved in the development of type 2 diabetes. Dietary factors can influence the abundance of A. muciniphila and F. prausnitzii, but the evidence is not clear. We systematically searched PubMed and Embase to identify clinical trials investigating any dietary intervention in relation to A. muciniphila and F. prausnitzii. Overall, 29 unique trials were included, of which five examined A. muciniphila, 19 examined F. prausnitzii, and six examined both, in a total of 1444 participants. A caloric restriction diet and supplementation with pomegranate extract, resveratrol, polydextrose, yeast fermentate, sodium butyrate, and inulin increased the abundance of A. muciniphila, while a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols decreased the abundance of A. muciniphila. For F. prausnitzii, the main studied intervention was prebiotics (e.g. fructo-oligosaccharides, inulin type fructans, raffinose); seven studies reported an increase after prebiotic intervention, while two studies reported a decrease, and four studies reported no difference. Current evidence suggests that some dietary factors may influence the abundance of A. muciniphila and F. prausnitzii. However, more research is needed to support these microflora strains as targets of microbiome shifts with dietary intervention and their use as medical nutrition therapy in prevention and management of chronic disease.


Subject(s)
Diet , Faecalibacterium prausnitzii/drug effects , Gastrointestinal Microbiome , Verrucomicrobia/drug effects , Akkermansia , Humans
20.
Nutrients ; 11(12)2019 Dec 07.
Article in English | MEDLINE | ID: mdl-31817899

ABSTRACT

Estrogen decline during menopause is associated with altered metabolism, weight gain and increased risk of cardiometabolic diseases. The gut microbiota also plays a role in the development of cardiometabolic dysfunction and is also subject to changes associated with age-related hormone changes. Phytoestrogens are plant-based estrogen mimics that have gained popularity as dietary supplements for the treatment or prevention of menopause-related symptoms. These compounds have the potential to both modulate and be metabolized by the gut microbiota. Hops (Humulus lupulus L.) contain potent phytoestrogen precursors, which rely on microbial biotransformation in the gut to estrogenic forms. We supplemented ovariectomized (OVX) or sham-operated (SHAM) C57BL/6 mice, with oral estradiol (E2), a flavonoid-rich extract from hops, or a placebo carrier oil, to observe effects on adiposity, inflammation, and gut bacteria composition. Hops extract (HE) and E2 protected against increased visceral adiposity and liver triglyceride accumulation in OVX animals. Surprisingly, we found no evidence of OVX having a significant impact on the overall gut bacterial community structure. We did find differences in the abundance of Akkermansia muciniphila, which was lower with HE treatment in the SHAM group relative to OVX E2 treatment and to placebo in the SHAM group.


Subject(s)
Estrogens/pharmacology , Flavonoids/pharmacology , Gastrointestinal Microbiome , Humulus/chemistry , Plant Extracts/pharmacology , Adiposity/drug effects , Akkermansia , Animals , Dietary Supplements/microbiology , Estradiol/pharmacology , Female , Flavanones , Mice , Mice, Inbred C57BL , Models, Animal , Ovariectomy , Phytoestrogens/pharmacology , Triglycerides/metabolism , Verrucomicrobia/drug effects , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL