Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
2.
J Agric Food Chem ; 72(27): 15345-15356, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38932522

ABSTRACT

The human intestinal mucus layer protects against pathogenic microorganisms and harmful substances, whereas it also provides an important colonization niche for mutualistic microbes. The main functional components of mucus are heavily glycosylated proteins, called mucins. Mucins can be cleaved and utilized by intestinal microbes. The mechanisms between intestinal microbes and the regulation of mucin glycosylation are still poorly understood. In this study, in vitro mucus was produced by HT29-MTX-E12 cells under Semi-Wet interface with Mechanical Stimulation. Cells were exposed to pasteurized nonpathogenic bacteria Akkermansia muciniphila, Ruminococcus gnavus, and Bacteroides fragilis to evaluate influence on glycosylation patterns. Following an optimized protocol, O- and N-glycans were efficiently and reproducibly released, identified, and semiquantified using MALDI-TOF-MS and PGC-LC-MS/MS. Exposure of cells to bacteria demonstrated increased diversity of sialylated O-glycans and increased abundance of high mannose N-glycans in in vitro produced mucus. Furthermore, changes in glycan ratios were observed. It is speculated that bacterial components interact with the enzymatic processes in glycan production and that pasteurized bacteria influence glycosyltransferases or genes involved. These results highlight the influence of pasteurized bacteria on glycosylation patterns, stress the intrinsic relationship between glycosylation and microbiota, and show the potential of using in vitro produced mucus to study glycosylation behavior.


Subject(s)
Gastrointestinal Microbiome , Mucus , Polysaccharides , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Glycosylation , Humans , Tandem Mass Spectrometry/methods , Mucus/microbiology , Mucus/metabolism , Mucus/chemistry , Polysaccharides/metabolism , Polysaccharides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Mucins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , HT29 Cells , Chromatography, Liquid/methods , Bacteroides fragilis/metabolism , Bacteroides fragilis/chemistry , Bacteroides fragilis/physiology , Pasteurization , Akkermansia/metabolism , Liquid Chromatography-Mass Spectrometry
3.
J Microbiol Methods ; 223: 106975, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38889842

ABSTRACT

The mucin-degrading gut commensal Akkermansia muciniphila (A. muciniphila) negatively correlates with various diseases, including metabolic disorders, neurodegenerative disorders, and cancers, through interacting with host receptors by diverse molecules. Still, their exact metabolic capability within the nutrient-rich environment (such as in the human gut) is not fully characterized. Therefore, in the present study, we investigated the comprehensive metabolome and lipidome of A. muciniphila after supplementation of four major gut microbial nutrients: mucin, inorganic salts, bile salts, and short-chain fatty acids (SCFAs). Our results showed that mucin is the predominant driver of the different lipidomic and metabolomic profiles of A. muciniphila, and it promotes the overall growth of this bacteria. While the addition of inorganic salts, bile salts, and SCFAs was found to inhibit the growth of A. muciniphila. Interestingly, inorganic salts affected the purine metabolism in A. muciniphila cultures, while adding bile salts significantly increased the production of other bile acids and N-acyl amides. Lastly, SCFAs were identified to alter the A. muciniphila energy utilization of triglycerides, fatty acyls, and phosphatidylethanolamines. To our knowledge, this is the first study to examine the comprehensive lipidome and metabolome of A. muciniphila, which highlights the importance of nutritional impacts on the lipidome and metabolome of A. muciniphila and hence providing foundational knowledge to unveil the potential effects of A. muciniphila on host health.


Subject(s)
Akkermansia , Bile Acids and Salts , Gastrointestinal Microbiome , Lipidomics , Metabolomics , Probiotics , Akkermansia/metabolism , Akkermansia/growth & development , Metabolomics/methods , Bile Acids and Salts/metabolism , Lipidomics/methods , Probiotics/metabolism , Gastrointestinal Microbiome/physiology , Humans , Chromatography, Liquid/methods , Metabolome , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis , Mucins/metabolism , Mass Spectrometry/methods
4.
World J Microbiol Biotechnol ; 40(7): 199, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727988

ABSTRACT

Glucagon-like peptide-1(GLP-1) is an incretin hormone secreted primarily from the intestinal L-cells in response to meals. GLP-1 is a key regulator of energy metabolism and food intake. It has been proven that P9 protein from A. muciniphila could increase GLP-1 release and improve glucose homeostasis in HFD-induced mice. To obtain an engineered Lactococcus lactis which produced P9 protein, mature polypeptide chain of P9 was codon-optimized, fused with N-terminal signal peptide Usp45, and expressed in L. lactis NZ9000. Heterologous secretion of P9 by recombinant L. lactis NZP9 were successfully detected by SDS-PAGE and western blotting. Notably, the supernatant of L. lactis NZP9 stimulated GLP-1 production of NCI-H716 cells. The relative expression level of GLP-1 biosynthesis gene GCG and PCSK1 were upregulated by 1.63 and 1.53 folds, respectively. To our knowledge, this is the first report on the secretory expression of carboxyl-terminal processing protease P9 from A. muciniphila in L. lactis. Our results suggest that genetically engineered L. lactis which expressed P9 may have therapeutic potential for the treatment of diabetes, obesity and other metabolic disorders.


Subject(s)
Akkermansia , Glucagon-Like Peptide 1 , Lactococcus lactis , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/genetics , Akkermansia/genetics , Akkermansia/metabolism , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Humans , L Cells , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Animals , Mice , Cell Line , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
FEMS Microbiol Ecol ; 100(7)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38794902

ABSTRACT

Mucin is a glycoprotein secreted throughout the mammalian gastrointestinal tract that can support endogenous microorganisms in the absence of complex polysaccharides. While several mucin-degrading bacteria have been identified, the interindividual differences in microbial communities capable of metabolizing this complex polymer are not well described. To determine whether community assembly on mucin is deterministic across individuals or whether taxonomically distinct but functionally similar mucin-degrading communities are selected across fecal inocula, we used a 10-day in vitro sequential batch culture fermentation from three human donors with mucin as the sole carbon source. For each donor, 16S rRNA gene amplicon sequencing was used to characterize microbial community succession, and the short-chain fatty acid profile was determined from the final community. All three communities reached a steady-state by day 7 in which the community composition stabilized. Taxonomic comparisons amongst communities revealed that one of the final communities had Desulfovibrio, another had Akkermansia, and all three shared other members, such as Bacteroides. Metabolic output differences were most notable for one of the donor's communities, with significantly less production of acetate and propionate than the other two communities. These findings demonstrate the feasibility of developing stable mucin-degrading communities with shared and unique taxa. Furthermore, the mechanisms and efficiencies of mucin degradation across individuals are important for understanding how this community-level process impacts human health.


Subject(s)
Feces , Fermentation , Microbial Consortia , Mucins , RNA, Ribosomal, 16S , Humans , Mucins/metabolism , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome , Akkermansia/metabolism , Desulfovibrio/metabolism , Desulfovibrio/genetics , Desulfovibrio/classification , Bacteroides/metabolism , Bacteroides/genetics , Bacteroides/classification , Bacteroides/growth & development
6.
Nat Commun ; 15(1): 4582, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811534

ABSTRACT

The intestinal anaerobic bacterium Akkermansia muciniphila is specialized in the degradation of mucins, which are heavily O-glycosylated proteins that constitute the major components of the mucus lining the intestine. Despite that adhesion to mucins is considered critical for the persistence of A. muciniphila in the human intestinal tract, our knowledge of how this intestinal symbiont recognizes and binds to mucins is still limited. Here, we first show that the mucin-binding properties of A. muciniphila are independent of environmental oxygen concentrations and not abolished by pasteurization. We then dissected the mucin-binding properties of pasteurized A. muciniphila by use of a recently developed cell-based mucin array that enables display of the tandem repeats of human mucins with distinct O-glycan patterns and structures. We found that A. muciniphila recognizes the unsialylated LacNAc (Galß1-4GlcNAcß1-R) disaccharide selectively on core2 and core3 O-glycans. This disaccharide epitope is abundantly found on human colonic mucins capped by sialic acids, and we demonstrated that endogenous A. muciniphila neuraminidase activity can uncover the epitope and promote binding. In summary, our study provides insights into the mucin-binding properties important for colonization of a key mucin-foraging bacterium.


Subject(s)
Akkermansia , Mucins , Polysaccharides , Akkermansia/metabolism , Humans , Mucins/metabolism , Polysaccharides/metabolism , Neuraminidase/metabolism , Protein Binding , Glycosylation , Disaccharides/metabolism , Verrucomicrobia/metabolism , Epitopes/metabolism , Bacterial Adhesion
7.
Cell ; 187(11): 2687-2689, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38788691

ABSTRACT

In this issue of Cell, Nie and co-authors report that the microbe-derived bile acid (BA) 3-succinylated cholic acid protects against the progression of metabolic dysfunction-associated liver disease. Intriguingly, its protective mechanism does not involve traditional BA signaling pathways but is instead linked to the proliferation of the commensal microbe Akkermansia muciniphila.


Subject(s)
Akkermansia , Bile Acids and Salts , Periodicals as Topic , Animals , Humans , Mice , Akkermansia/metabolism , Bile Acids and Salts/metabolism , Cholic Acid/metabolism , Gastrointestinal Microbiome , Liver/metabolism , Liver Diseases/metabolism , Liver Diseases/microbiology , Verrucomicrobia/metabolism
8.
Adv Sci (Weinh) ; 11(28): e2306217, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38742466

ABSTRACT

Radiation proctopathy (RP) is a common complication of radiotherapy for pelvic malignancies with high incidence. RP accompanies by microbial dysbiosis. However, how the gut microbiota affects the disease remains unclear. Here, metabolomics reveals that the fecal and serous concentrations of microbiota-derived 3-hydroxybutyrate (3HB) are significantly reduced in RP mice and radiotherapeutic patients. Moreover, the concentration of 3HB is negatively associated with the expression of proinflammatory IL6 that is increased along with the severity of radiation damage. 3HB treatment significantly downregulates IL6 expression and alleviates IL6-mediated radiation damage. Irradiated cell-fecal microbiota co-culture experiments and in vivo assays show that such a radioprotection of 3HB is mediated by GPR43. Microbiome analysis reveals that radiation leads to a distinct bacterial community compared to untreated controls, in which Akkermansia muciniphila is significantly reduced in RP mice and radiotherapeutic patients and is associated with lower 3HB concentration. Gavage of A. muciniphila significantly increases 3HB concentration, downregulates GPR43 and IL6 expression, and ameliorates radiation damage. Collectively, these results demonstrate that the gut microbiota, including A. muciniphila, induce higher concentrations of 3HB to block GPR43-mediated IL6 signaling, thereby conferring radioprotection. The findings reveal a novel implication of the gut-immune axis in radiation pathophysiology, with potential therapeutic applications.


Subject(s)
3-Hydroxybutyric Acid , Gastrointestinal Microbiome , Interleukin-6 , Receptors, G-Protein-Coupled , Signal Transduction , Animals , Mice , Interleukin-6/metabolism , Interleukin-6/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , 3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/pharmacology , Humans , Radiation Injuries/metabolism , Disease Models, Animal , Proctitis/metabolism , Mice, Inbred C57BL , Male , Akkermansia/metabolism
9.
Cell ; 187(11): 2717-2734.e33, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38653239

ABSTRACT

The gut microbiota has been found to play an important role in the progression of metabolic dysfunction-associated steatohepatitis (MASH), but the mechanisms have not been established. Here, by developing a click-chemistry-based enrichment strategy, we identified several microbial-derived bile acids, including the previously uncharacterized 3-succinylated cholic acid (3-sucCA), which is negatively correlated with liver damage in patients with liver-tissue-biopsy-proven metabolic dysfunction-associated fatty liver disease (MAFLD). By screening human bacterial isolates, we identified Bacteroides uniformis strains as effective producers of 3-sucCA both in vitro and in vivo. By activity-based protein purification and identification, we identified an enzyme annotated as ß-lactamase in B. uniformis responsible for 3-sucCA biosynthesis. Furthermore, we found that 3-sucCA is a lumen-restricted metabolite and alleviates MASH by promoting the growth of Akkermansia muciniphila. Together, our data offer new insights into the gut microbiota-liver axis that may be leveraged to augment the management of MASH.


Subject(s)
Akkermansia , Bacteroides , Bile Acids and Salts , Gastrointestinal Microbiome , Non-alcoholic Fatty Liver Disease , Symbiosis , Animals , Humans , Male , Mice , Akkermansia/metabolism , Bacteroides/metabolism , beta-Lactamases/metabolism , Bile Acids and Salts/metabolism , Biosynthetic Pathways/genetics , Fatty Liver/metabolism , Liver/metabolism , Mice, Inbred C57BL , Verrucomicrobia/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/microbiology
10.
J Bacteriol ; 206(2): e0033423, 2024 02 22.
Article in English | MEDLINE | ID: mdl-38299857

ABSTRACT

Among the first microorganisms to colonize the human gut of breastfed infants are bacteria capable of fermenting human milk oligosaccharides (HMOs). One of the most abundant HMOs, 2'-fucosyllactose (2'-FL), may specifically drive bacterial colonization of the intestine. Recently, differential growth has been observed across multiple species of Akkermansia on various HMOs including 2'-FL. In culture, we found growth of two species, A. muciniphila MucT and A. biwaensis CSUN-19,on HMOs corresponded to a decrease in the levels of 2'-FL and an increase in lactose, indicating that the first step in 2'-FL catabolism is the cleavage of fucose. Using phylogenetic analysis and transcriptional profiling, we found that the number and expression of fucosidase genes from two glycoside hydrolase (GH) families, GH29 and GH95, vary between these two species. During the mid-log phase of growth, the expression of several GH29 genes was increased by 2'-FL in both species, whereas the GH95 genes were induced only in A. muciniphila. We further show that one putative fucosidase and a ß-galactosidase from A. biwaensis are involved in the breakdown of 2'-FL. Our findings indicate that the plasticity of GHs of human-associated Akkermansia sp. enables access to additional growth substrates present in HMOs, including 2'-FL. Our work highlights the potential for Akkermansia to influence the development of the gut microbiota early in life and expands the known metabolic capabilities of this important human symbiont.IMPORTANCEAkkermansia are mucin-degrading specialists widely distributed in the human population. Akkermansia biwaensis has recently been observed to have enhanced growth relative to other human-associated Akkermansia on multiple human milk oligosaccharides (HMOs). However, the mechanisms for enhanced growth are not understood. Here, we characterized the phylogenetic diversity and function of select genes involved in the growth of A. biwaensis on 2'-fucosyllactose (2'-FL), a dominant HMO. Specifically, we demonstrate that two genes in a genomic locus, a putative ß-galactosidase and α-fucosidase, are likely responsible for the enhanced growth on 2'-FL. The functional characterization of A. biwaensis growth on 2'-FL delineates the significance of a single genomic locus that may facilitate enhanced colonization and functional activity of select Akkermansia early in life.


Subject(s)
Akkermansia , Trisaccharides , alpha-L-Fucosidase , Infant , Humans , Akkermansia/metabolism , alpha-L-Fucosidase/genetics , alpha-L-Fucosidase/metabolism , Phylogeny , Oligosaccharides/metabolism , beta-Galactosidase/genetics
11.
mSystems ; 9(1): e0104923, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38095419

ABSTRACT

The gut microbiota is a key player in the host metabolism. Some bacteria are able to ferment non-digestible compounds and produce short-chain fatty acids that the host can later transform and accumulate in tissue. In this study, we aimed to better understand the relationships between the microorganisms and the short-chain fatty acid composition of the rectal content, including the possible linkage with the fatty acid composition in backfat and muscle of the pig. We studied a Duroc × Iberian crossbred population, and we found significant correlations between different bacterial and archaeal genera and the fatty acid profile. The abundance of n-butyric acid in the rectal content was positively associated with Prevotella spp. and negatively associated with Akkermansia spp., while conversely, the abundance of acetic acid was negatively and positively associated with the levels of Prevotella spp. and Akkermansia spp., respectively. The most abundant genus, Rikenellaceae RC9 gut group, had a positive correlation with palmitic acid in muscle and negative correlations with stearic acid in backfat and oleic acid in muscle. These results suggest the possible role of Prevotella spp. and Akkermansia spp. as biomarkers for acetic and n-butyric acids, and the relationship of Rikenellaceae RC9 gut group with the lipid metabolism, building up the potential, although indirect, role of the microbiota in the modification of the backfat and muscle fatty acid composition of the host.IMPORTANCEThe vital role of the gut microbiota on its host metabolism makes it essential to know how its modulation is mirrored on the fatty acid composition of the host. Our findings suggest Prevotella spp. and Akkermansia spp. as potential biomarkers for the levels of beneficial short-chain fatty acids and the possible influence of Rikenellaceae RC9 gut group in the backfat and muscle fatty acid composition of the pig.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Swine , Animals , Fatty Acids , Fatty Acids, Volatile/metabolism , Bacteria , Butyric Acid , Akkermansia/metabolism , Bacteroidetes/metabolism , Biomarkers
13.
Int Immunopharmacol ; 121: 110548, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37356123

ABSTRACT

Acute lung injury (ALI) is a life-threatening disease with high morbidity and mortality. Our previous results demonstrated that Ficolin A (FcnA) protected against lipopolysaccharide (LPS)-induced mild ALI via activating complement, however the mechanism of severe lung damage caused by sepsis remains unclear. This study aimed to investigate whether FcnA modulated gut microbiota to affect the progression of sepsis-induced severe ALI. Fcna-/- and Fcnb-/- C57BL/6 mice were applied to establish the ALI model by injection of LPS intraperitoneally. Mice were treated with antibiotics, fecal microbiota transplantation (FMT), and intratracheal administration of recombinant protein S100A4. Changes in body weight of mice were recorded, and lung injury were assessed. Then lung tissue wet/dry weight was calculated. We found knockout of FcnA, but not FcnB, alleviated sepsis-induced severe ALI evidenced by increased body weight change, decreased wet/dry weight of lung tissue, reduced inflammatory infiltration, decreased lung damage score, decreased Muc-2, TNF-α, IL-1ß, IL-6, and Cr levels, and increased sIgA levels. Furthermore, knockout of FcnA restored gut microbiota homeostasis in mice. Correlation analysis showed that Akkermansia was significantly negatively associated with TNF-α, IL-1ß, and IL-6 levels in serum and bronchoalveolar lavage fluid (BALF). Moreover, knockout of FcnA regulated gut microbiota to protect ALI through S100A4. Finally, we found knockout of FcnA alleviated ALI by inhibiting S100A4 via gut Akkermansia in mice, which may provide further insights and new targets into treating sepsis-induced severe lung injury.


Subject(s)
Acute Lung Injury , Sepsis , Mice , Animals , Akkermansia/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Mice, Inbred C57BL , Lung/metabolism , Acute Lung Injury/chemically induced , Sepsis/metabolism , Ficolins
14.
Crit Rev Microbiol ; 49(1): 82-100, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35603929

ABSTRACT

Akkermansia muciniphila, an intestinal microorganism, belongs to Verrucomicrobia, one of the most abundant microorganisms in the mammalian gut. It is a mucin-degrading bacterium that can colonise intestines of mammals such as humans and mice by utilising mucin as the only nitrogen and carbon source. When A. muciniphila colonises the intestine, its metabolites interact with the intestinal barrier, affecting host health by consolidating the intestinal barrier, regulating metabolic functions of the intestinal and circulatory systems, and regulating immune functions. This review summarised the mechanisms of A. muciniphila-host interactions that are relevant to host health. We focussed on characteristics of A. muciniphila in relation to its metabolites to provide a comprehensive understanding of A. muciniphila and its effects on host health and disease processes.


Subject(s)
Akkermansia , Verrucomicrobia , Humans , Animals , Mice , Verrucomicrobia/genetics , Verrucomicrobia/metabolism , Akkermansia/metabolism , Mucins/metabolism , Mammals/metabolism
15.
Physiol Behav ; 258: 114023, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36336146

ABSTRACT

Akkermansia muciniphila is present in the mucus layer of its host gut, and its outer membrane protein Amuc_1100 has a significant ameliorative effect on metabolic disorders and emotional memory aspects of enteritis, obesity, depression, and anxiety in the host. Antibiotics affect gut microbial composition, leading to imbalance and behavioral changes in the gut-brain axis, while probiotics have a protective effect against behavioral changes caused by gut flora disorders. In the present study, a depressed mouse model using a broad-spectrum cocktail mixture resulted in increased anxiety and depression-like behavior, decreased serum and hippocampal levels of 5-hydroxytryptamine (5-HT), and increased serum corticosterone (cort) levels. After application of A. muciniphila and Amuc_1100, anxiety and depression-like behavior in antibiotic-treated mice were significantly alleviated. In addition, the brain derived neurotrophic factor / Tropomyosin receptor kinase B (BDNF/TrkB) signaling pathway was altered, glial fibrillary acidic protein (GFAP) expression increased, and c-Fos protein expression decreased in the hippocampus of antibiotic-treated mice. After treatment with A. muciniphila and Amuc_1100, BDNF and TrkB levels were restored in the hippocampus and cortex. These results suggest that A. muciniphila and Amuc_1100 may alleviate antibiotic-induced anxiety and depression by affecting the BDNF/TrkB signaling pathway.


Subject(s)
Anxiety , Bacterial Outer Membrane Proteins , Depression , Animals , Mice , Anti-Bacterial Agents/adverse effects , Anxiety/chemically induced , Anxiety/drug therapy , Brain-Derived Neurotrophic Factor/metabolism , Depression/chemically induced , Depression/drug therapy , Depression/metabolism , Serotonin/metabolism , Akkermansia/metabolism , Bacterial Outer Membrane Proteins/pharmacology
16.
Sci Rep ; 12(1): 10031, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35705595

ABSTRACT

This study aimed to investigate the effects of active and heat-inactivated forms of Akkermansia muciniphila, bacterium-derived outer membrane vesicles (OMVs), and cell-free supernatant on the transcription of endocannabinoid system (ECS) members, including cannabinoid receptors 1 and 2 (CB1 and CB2), fatty acid amide hydrolase (FAAH), and peroxisome proliferator-activated receptors (PPARs) genes (i.e., α, ß/δ, and δ) in Caco-2 and HepG-2 cell lines. After the inoculation of A. muciniphila in brain heart infusion enriched medium, OMVs and cell-free supernatant were extracted. For the investigation of the effects of bacteria and its derivatives on the expression of ECS and PPARs genes, the aforementioned cells were treated by active and heat-inactivated bacteria, OMVs, and cell-free supernatant. Quantitative real-time polymerase chain reaction analysis revealed that both forms of the bacterium, bacterial-derived OMVs, and cell-free supernatant could affect the expression of CB1, CB2, FAAH, and PPARs genes (i.e., α, ß/δ, and δ) significantly (P < 0.05). Considering the engagement of the aforementioned genes in metabolic pathways, it might be suggested that both forms of the bacterium, OMVs, and cell-free supernatant might have the potential to serve as a probiotic, paraprobiotic, and postbiotic candidate to prevent obesity, metabolic disorders, and liver diseases.


Subject(s)
Akkermansia , Endocannabinoids , Peroxisome Proliferator-Activated Receptors , Akkermansia/metabolism , Caco-2 Cells , Endocannabinoids/metabolism , Humans , Peroxisome Proliferator-Activated Receptors/genetics , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics
17.
Int J Mol Sci ; 23(4)2022 Feb 19.
Article in English | MEDLINE | ID: mdl-35216439

ABSTRACT

The development of non-alcoholic steatohepatitis (NASH) has been associated with alterations in gut microbiota composition and reduced gut barrier function. Akkermansia muciniphila is a gut microbe that is thought to have health-promoting properties, including the ability to improve gut barrier function and host metabolism, both when administered live and after heat-inactivation. We questioned whether heat-inactivated A. muciniphila may reduce NASH development. Ldlr-/-.Leiden mice, a translational, diet-induced model for NASH, were fed a NASH-inducing high-fat diet (HFD) supplemented with heat-inactivated A. muciniphila. After 28 weeks, effects of the treatment on obesity and associated metabolic dysfunction in the gut (microbiota composition and permeability), adipose tissue, and liver were studied relative to an untreated HFD control. Treatment with heat-inactivated A. muciniphila did not affect body weight or adiposity and had no effect on plasma lipids, blood glucose, or plasma insulin. Heat-inactivated A. muciniphila had some minor effects on mucosal microbiota composition in ileum and colon and improved gut barrier function, as assessed by an in vivo functional gut permeability test. Epidydimal white adipose tissue (WAT) hypertrophy and inflammation were not affected, but heat-inactivated A. muciniphila did reduce hypertrophy in the mesenteric WAT which is in close proximity to the intestine. Heat-inactivated A. muciniphila did not affect the development of NASH or associated fibrosis in the liver and did not affect circulating bile acids or markers of liver fibrosis, but did reduce PRO-C4, a type IV collagen synthesis marker, which may be associated with gut integrity. In conclusion, despite beneficial effects in the gut and mesenteric adipose tissue, heat-inactivated A. muciniphila did not affect the development of NASH and fibrosis in a chronic disease setting that mimics clinically relevant disease stages.


Subject(s)
Intestinal Mucosa/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, LDL/metabolism , Adipose Tissue/metabolism , Akkermansia/metabolism , Animals , Diet, High-Fat/methods , Gastrointestinal Microbiome/physiology , Hot Temperature , Inflammation/metabolism , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Mice , Mice, Obese , Obesity/metabolism , Permeability
18.
Cell Mol Life Sci ; 79(2): 76, 2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35043293

ABSTRACT

Probiotics currently available on the market generally belong to a narrow range of microbial species. However, recent studies about the importance of the gut microbial commensals on human health highlighted that the gut microbiome is an unexplored reservoir of potentially beneficial microbes. For this reason, academic and industrial research is focused on identifying and testing novel microbial strains of gut origin for the development of next-generation probiotics. Although several of these are promising for the prevention and treatment of many chronic diseases, studies on human subjects are still scarce and approval from regulatory agencies is, therefore, rare. In addition, some issues need to be overcome before implementing their wide application on the market, such as the best methods for cultivation and storage of these oxygen-sensitive taxa. This review summarizes the most recent evidence related to NGPs and provides an outlook to the main issues that still limit their wide employment.


Subject(s)
Bacteria/classification , Bacteria/metabolism , Gastrointestinal Microbiome/physiology , Probiotics/pharmacology , Akkermansia/metabolism , Bacterial Physiological Phenomena , Clostridiales/metabolism , Dysbiosis/microbiology , Faecalibacterium prausnitzii/metabolism , Humans , Prevotella/metabolism
19.
PLoS One ; 16(12): e0260757, 2021.
Article in English | MEDLINE | ID: mdl-34855864

ABSTRACT

Akkermansia muciniphila is a Gram-negative bacterium that resides within the gut mucus layer, and plays an important role in promoting gut barrier integrity, modulating the immune response and inhibiting gut inflammation. Growth stimulation of A. muciniphila by polyphenols including epigallocatechin-3-gallate (EGCG) from difference sources is well-documented. However, no published in vitro culture data on utilization of polyphenols by A. muciniphila are available, and the mechanism of growth-stimulating prebiotic effect of polyphenols on it remains unclear. Here in vitro culture studies have been carried out on the metabolism of EGCG by A. muciniphila in the presence of either mucin or glucose. We found that A. muciniphila did not metabolize EGCG alone but could co-metabolize it together with both these substrates in the presence of mineral salts and amino acids for mucin and protein sources for glucose. Our metabolomic data show that A. muciniphila converts EGCG to gallic acid, epigallocatechin, and (-)-epicatechin through ester hydrolysis. The (-)-epicatechin formed is then further converted to hydroxyhydroquinone. Co-metabolism of A. muciniphila of EGCG together with either mucin or glucose promoted substantially its growth, which serves as a further demonstration of the growth-promoting effect of polyphenols on A. muciniphila and provides an important addition to the currently available proposed mechanisms of polyphenolic prebiotic effects on A. muciniphila.


Subject(s)
Catechin/analogs & derivatives , Glucose/metabolism , Metabolome , Mucins/metabolism , Akkermansia/growth & development , Akkermansia/metabolism , Catechin/metabolism , In Vitro Techniques
20.
BMC Microbiol ; 21(1): 298, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34715771

ABSTRACT

BACKGROUND: Akkermansia muciniphila is a member of the human gut microbiota where it resides in the mucus layer and uses mucin as the sole carbon, nitrogen and energy source. A. muciniphila is the only representative of the Verrucomicrobia phylum in the human gut. However, A. muciniphila 16S rRNA gene sequences have also been found in the intestines of many vertebrates. RESULTS: We detected A. muciniphila-like bacteria in the intestines of animals belonging to 15 out of 16 mammalian orders. In addition, other species belonging to the Verrucomicrobia phylum were detected in fecal samples. We isolated 10 new A. muciniphila strains from the feces of chimpanzee, siamang, mouse, pig, reindeer, horse and elephant. The physiology and genome of these strains were highly similar in comparison to the type strain A. muciniphila MucT. Overall, the genomes of the new strains showed high average nucleotide identity (93.9 to 99.7%). In these genomes, we detected considerable conservation of at least 75 of the 78 mucin degradation genes that were previously detected in the genome of the type strain MucT. CONCLUSIONS: The low genomic divergence observed in the new strains may indicate that A. muciniphila favors mucosal colonization independent of the differences in hosts. In addition, the conserved mucus degradation capability points towards a similar beneficial role of the new strains in regulating host metabolic health.


Subject(s)
Genome, Bacterial/genetics , Mammals/microbiology , Akkermansia/classification , Akkermansia/genetics , Akkermansia/isolation & purification , Akkermansia/metabolism , Animals , Feces/microbiology , Gastrointestinal Tract/microbiology , Genetic Variation , Genomics , Humans , Mammals/classification , Mice , Mucins/metabolism , Phylogeny , RNA, Ribosomal, 16S/genetics , Verrucomicrobia/classification , Verrucomicrobia/genetics , Verrucomicrobia/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL