Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
1.
Sci Rep ; 14(1): 20056, 2024 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-39209875

RESUMEN

Although Akkermansia muciniphila (Am) plays a beneficial role as a probiotic in the treatment of metabolic syndrome, the mechanisms remain elusive. We tested the hypothesis that Am extracellular vesicles (AmEVs) protect against hypertension through modulation of gene expression in the kidneys of spontaneously hypertensive rats (SHRs). Extracellular vesicles purified from anaerobically cultured Am (1.0 × 108 or 1.0 × 109 particles/kg) or vehicles were injected into the tail veins of Wistar-Kyoto rats (WKYs) and SHRs weekly for 4 weeks. Renal cortical tissues isolated from both rat strains were analyzed by trichrome stain and RT-qPCR. AmEVs protect against the development of hypertension in SHRs without a serious adverse reaction. AmEVs increased the expression of vasocontracting Agt and At1ar as well as vasodilating At2r, Mas1 and Nos2 in the kidneys of both strains. These results indicate that AmEVs have a protective effect against hypertension without a serious adverse reaction. Therefore, it is foreseen that AmEVs may be utilized as a novel therapeutic for the treatment of hypertension.


Asunto(s)
Akkermansia , Vesículas Extracelulares , Hipertensión , Riñón , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Animales , Vesículas Extracelulares/metabolismo , Ratas , Riñón/metabolismo , Hipertensión/metabolismo , Hipertensión/genética , Masculino , Administración Intravenosa , Verrucomicrobia/genética , Regulación de la Expresión Génica , Probióticos/administración & dosificación
2.
Microbiologyopen ; 13(4): e1430, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39115291

RESUMEN

The gut microbiota significantly contributes to human health and well-being. The aim of this study was to evaluate the stability and resilience of a consortium composed of three next-generation probiotics (NGPs) candidates originally found in the human gut. The growth patterns of Akkermansia muciniphila, Bacteroides thetaiotaomicron, and Faecalibacterium prausnitzii were studied both individually and consortium. The growth kinetics of Akkermansia muciniphila (A. muciniphila), Bacteroides thetaiotaomicron (B. thetaiotaomicron), and Faecalibacterium prausnitzii (F. prausnitzii) were characterized both individually and in consortium using isothermal microcalorimetry and 16S ribosomal RNA next-generation sequencing. The consortium reached stability after three passages and demonstrated resilience to changes in its initial composition. The concentration of butyrate produced was nearly twice as high in the consortium compared to the monoculture of F. prausnitzii. The experimental conditions and methodologies used in this article are a solid foundation for developing further complex consortia.


Asunto(s)
Calorimetría , Microbioma Gastrointestinal , ARN Ribosómico 16S , Humanos , Microbioma Gastrointestinal/fisiología , ARN Ribosómico 16S/genética , Faecalibacterium prausnitzii/genética , Akkermansia/crecimiento & desarrollo , Akkermansia/fisiología , Consorcios Microbianos/fisiología , Consorcios Microbianos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Butiratos/metabolismo , Probióticos , Verrucomicrobia/genética , Verrucomicrobia/crecimiento & desarrollo , Bacteroides/genética , Bacteroides/crecimiento & desarrollo , ADN Bacteriano/genética
3.
Chem Commun (Camb) ; 60(68): 9089-9092, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39108142

RESUMEN

Herein, we have developed a non-enzymatic, isothermal amplification assay (NIA sensor) based on a catalytic hairpin assembly (CHA) reaction for quantifying the relative abundance of Akkermansia muciniphila. Through detection of the MUC-1437 gene (limit of detection: 8.3 fM) in a dynamic range from 10 fM to 1 nM, the NIA sensor shows high sensitivity and selectivity in preclinical models of mice fed a normal or high-fat diet (HFD), and treated with antibiotics (ATB). The NIA sensor, which operates without the use of any enzymes, leading to simplicity and cost-effectiveness, has great potential for biosensing research and clinical diagnostic applications.


Asunto(s)
Akkermansia , Técnicas de Amplificación de Ácido Nucleico , Animales , Ratones , Técnicas Biosensibles , Antibacterianos/farmacología , Dieta Alta en Grasa , Límite de Detección , Verrucomicrobia/genética , Verrucomicrobia/aislamiento & purificación
5.
Mol Syst Biol ; 20(6): 596-625, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38745106

RESUMEN

The erosion of the colonic mucus layer by a dietary fiber-deprived gut microbiota results in heightened susceptibility to an attaching and effacing pathogen, Citrobacter rodentium. Nevertheless, the questions of whether and how specific mucolytic bacteria aid in the increased pathogen susceptibility remain unexplored. Here, we leverage a functionally characterized, 14-member synthetic human microbiota in gnotobiotic mice to deduce which bacteria and functions are responsible for the pathogen susceptibility. Using strain dropouts of mucolytic bacteria from the community, we show that Akkermansia muciniphila renders the host more vulnerable to the mucosal pathogen during fiber deprivation. However, the presence of A. muciniphila reduces pathogen load on a fiber-sufficient diet, highlighting the context-dependent beneficial effects of this mucin specialist. The enhanced pathogen susceptibility is not owing to altered host immune or pathogen responses, but is driven by a combination of increased mucus penetrability and altered activities of A. muciniphila and other community members. Our study provides novel insights into the mechanisms of how discrete functional responses of the same mucolytic bacterium either resist or enhance enteric pathogen susceptibility.


Asunto(s)
Akkermansia , Citrobacter rodentium , Microbioma Gastrointestinal , Animales , Ratones , Citrobacter rodentium/patogenicidad , Humanos , Susceptibilidad a Enfermedades , Fibras de la Dieta/metabolismo , Vida Libre de Gérmenes , Dieta , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Verrucomicrobia/genética , Infecciones por Enterobacteriaceae/microbiología , Colon/microbiología , Ratones Endogámicos C57BL
6.
Gut Microbes ; 16(1): 2338947, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38717824

RESUMEN

The gut microbiota has coevolved with the host for hundreds of millions of years, playing a beneficial role in host health. Human parasitic helminths are widespread and pose a pervasive global public health issue. Although Type 2 immunity provides partial resistance to helminth infections, the composition of the gut microbiota can change correspondingly. Therefore, it raises the question of what role the gut microbiota plays during helminth infection. Akkermansia muciniphila has emerged as a notable representative of beneficial microorganisms in the gut microbiota. Recent studies indicate that A. muciniphila is not merely associated with helminth infection but is also causally linked to infection. Here, we provide an overview of the crosstalk between A. muciniphila and enteric helminth infection. Our goal is to enhance our understanding of the interplay among A. muciniphila, helminths, and their hosts while also exploring the potential underlying mechanisms.


Asunto(s)
Akkermansia , Microbioma Gastrointestinal , Animales , Humanos , Helmintiasis/inmunología , Helmintos/inmunología , Helmintos/genética , Verrucomicrobia/genética , Verrucomicrobia/inmunología
7.
Protein Expr Purif ; 219: 106483, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38609025

RESUMEN

Mussel foot proteins (Mfps) possess unique binding properties to various surfaces due to the presence of L-3,4-dihydroxyphenylalanine (DOPA). Mytilus edulis foot protein-3 (Mefp-3) is one of several proteins in the byssal adhesive plaque. Its localization at the plaque-substrate interface approved that Mefp-3 plays a key role in adhesion. Therefore, the protein is suitable for the development of innovative bio-based binders. However, recombinant Mfp-3s are mainly purified from inclusion bodies under denaturing conditions. Here, we describe a robust and reproducible protocol for obtaining soluble and tag-free Mefp-3 using the SUMO-fusion technology. Additionally, a microbial tyrosinase from Verrucomicrobium spinosum was used for the in vitro hydroxylation of peptide-bound tyrosines in Mefp-3 for the first time. The highly hydroxylated Mefp-3, confirmed by MALDI-TOF-MS, exhibited excellent adhesive properties comparable to a commercial glue. These results demonstrate a concerted and simplified high yield production process for recombinant soluble and tag-free Mfp3-based proteins with on demand DOPA modification.


Asunto(s)
Dihidroxifenilalanina , Mytilus edulis , Animales , Dihidroxifenilalanina/química , Dihidroxifenilalanina/metabolismo , Mytilus edulis/genética , Mytilus edulis/química , Mytilus edulis/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Verrucomicrobia/genética , Verrucomicrobia/metabolismo , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/metabolismo , Monofenol Monooxigenasa/química , Proteínas/genética , Proteínas/química , Proteínas/aislamiento & purificación , Hidroxilación , Escherichia coli/genética , Escherichia coli/metabolismo
8.
mBio ; 15(4): e0007824, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38470269

RESUMEN

Inflammatory bowel disease (IBD) is a group of inflammatory conditions of the gastrointestinal tract. The etiology of IBD remains elusive, but the disease is suggested to arise from the interaction of environmental and genetic factors that trigger inadequate immune responses and inflammation in the intestine. The gut microbiome majorly contributes to disease as an environmental variable, and although some causative bacteria are identified, little is known about which specific members of the microbiome aid in the intestinal epithelial barrier function to protect from disease. While chemically inducing colitis in mice from two distinct animal facilities, we serendipitously found that mice in one facility showed remarkable resistance to disease development, which was associated with increased markers of epithelial barrier integrity. Importantly, we show that Akkermansia muciniphila and Parabacteroides distasonis were significantly increased in the microbiota of resistant mice. To causally connect these microbes to protection against disease, we colonized susceptible mice with the two bacterial species. Our results demonstrate that A. muciniphila and P. distasonis synergistically drive a protective effect in both acute and chronic models of colitis by boosting the frequency of type 3 innate lymphoid cells in the colon and by improving gut epithelial integrity. Altogether, our work reveals a combined effort of commensal microbes in offering protection against severe intestinal inflammation by shaping gut immunity and by enhancing intestinal epithelial barrier stability. Our study highlights the beneficial role of gut bacteria in dictating intestinal homeostasis, which is an important step toward employing microbiome-driven therapeutic approaches for IBD clinical management. IMPORTANCE: The contribution of the gut microbiome to the balance between homeostasis and inflammation is widely known. Nevertheless, the etiology of inflammatory bowel disease, which is known to be influenced by genetics, immune response, and environmental cues, remains unclear. Unlocking novel players involved in the dictation of a protective gut, namely, in the microbiota component, is therefore crucial to develop novel strategies to tackle IBD. Herein, we revealed a synergistic interaction between two commensal bacterial strains, Akkermansia muciniphila and Parabacteroides distasonis, which induce protection against both acute and chronic models of colitis induction, by enhancing epithelial barrier integrity and promoting group 3 innate lymphoid cells in the colonic mucosa. This study provides a novel insight on how commensal bacteria can beneficially act to promote intestinal homeostasis, which may open new avenues toward the use of microbiome-derived strategies to tackle IBD.


Asunto(s)
Bacteroidetes , Colitis , Enfermedades Inflamatorias del Intestino , Animales , Ratones , Inmunidad Innata , Linfocitos , Colitis/microbiología , Enfermedades Inflamatorias del Intestino/microbiología , Inflamación , Verrucomicrobia/genética , Akkermansia
9.
Commun Biol ; 7(1): 192, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38365881

RESUMEN

The initial exposure to pathogens and commensals confers innate immune cells the capacity to respond distinctively upon a second stimulus. This training capacity might play key functions in developing an adequate innate immune response to the continuous exposure to bacteria. However, the mechanisms involved in induction of trained immunity by commensals remain mostly unexplored. A. muciniphila represents an attractive candidate to study the promotion of these long-term responses. Here, we show that priming of macrophages with live A. muciniphila enhances bacterial intracellular survival and decreases the release of pro- and anti-inflammatory signals, lowering the production of TNF and IL-10. Global transcriptional analysis of macrophages after a secondary exposure to the bacteria showed the transcriptional rearrangement underpinning the phenotype observed compared to acutely exposed cells, with the increased expression of genes related to phagocytic capacity and those involved in the metabolic adjustment conducing to innate immune training. Accordingly, key genes related to bacterial killing and pro-inflammatory pathways were downregulated. These data demonstrate the importance of specific bacterial members in the modulation of local long-term innate immune responses, broadening our knowledge of the association between gut microbiome commensals and trained immunity as well as the anti-inflammatory probiotic potential of A. muciniphila.


Asunto(s)
Inflamación , Verrucomicrobia , Humanos , Inflamación/genética , Verrucomicrobia/genética , Verrucomicrobia/metabolismo , Fenotipo , Antiinflamatorios/metabolismo , Akkermansia
10.
Neurogastroenterol Motil ; 36(3): e14745, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38263790

RESUMEN

BACKGROUND: Inflammatory bowel diseases (IBD) are chronic diseases that are not fully understood. Drugs in use can only be applied for a short time due to their side effects. Therefore, research is needed to develop new treatment approaches. In addition, it has been proven that IBD causes degeneration in the enteric nervous system (ENS). In recent years, it has been discussed that probiotics may have positive effects in the prevention and treatment of inflammatory enteric degeneration. Akkermansia muciniphila (A. muciniphila) is an anaerobic bacterium found in the mucin layer of the intestinal microbiota. It has been found that the population of A. muciniphila decreases in the case of different diseases. In light of this information, the curative effect of A. muciniphila application on colitis-induced inflammation and enteric degeneration was investigated. METHODS: In this study, 5 weeks of A. muciniphila treatment in Trinitro-benzene-sulfonic acid (TNBS)-induced chronic colitis model was investigated. Colon samples were examined at microscopic, biochemical, and molecular levels. Fecal samples were collected before, during, and after treatment to evaluate the population changes in the microbiota. Specific proteins secreted from the ENS were evaluated, and enteric degeneration was examined. RESULTS: As a result of the research, the ameliorative effects of A. muciniphila were shown in the TNBS colitis model-induced inflammation and ENS damage. DISCUSSION: In light of these results, A. muciniphila can potentially be evaluated as a microbiome-based treatment for IBD with further clinical and experimental studies.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Ratones , Animales , Enfermedades Neuroinflamatorias , Composición de Base , Análisis de Secuencia de ADN , ARN Ribosómico 16S , Filogenia , Colitis/inducido químicamente , Colitis/terapia , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Inflamatorias del Intestino/microbiología , Verrucomicrobia/genética , Inflamación , Enfermedad Crónica , Akkermansia
11.
Nat Commun ; 14(1): 7224, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37940686

RESUMEN

Planctopirus limnophila belongs to the bacterial phylum Planctomycetes, a relatively understudied lineage with remarkable cell biology features. Here, we report a genome-wide analysis of essential gene content in P. limnophila. We show that certain genes involved in peptidoglycan synthesis or cell division, which are essential in most other studied bacteria, are not essential for growth under laboratory conditions in this species. We identify essential genes likely involved in lipopolysaccharide biosynthesis, consistent with the view of Planctomycetes as diderm bacteria, and highlight other essential genes of unknown functions. Furthermore, we explore potential stages of evolution of the essential gene repertoire in Planctomycetes and the related phyla Verrucomicrobia and Chlamydiae. Our results provide insights into the divergent molecular and cellular biology of Planctomycetes.


Asunto(s)
Genes Esenciales , Planctomycetales , Planctomycetales/genética , Verrucomicrobia/genética
12.
Arch Microbiol ; 205(12): 366, 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37917352

RESUMEN

The PVC superphylum is a diverse group of prokaryotes that require stringent growth conditions. RNA is a fascinating molecule to find evolutionary relatedness according to the RNA World Hypothesis. We conducted tRNA gene analysis to find evolutionary relationships in the PVC phyla. The analysis of genomic data (P = 9, V = 4, C = 8) revealed that the number of tRNA genes varied from 28 to 90 in Planctomycetes and Chlamydia, respectively. Verrucomicrobia has whole genomes and the longest scaffold (3 + 1), with tRNA genes ranging from 49 to 53 in whole genomes and 4 in the longest scaffold. Most tRNAs in the E. coli genome clustered with homologs, but approximately 43% clustered with tRNAs encoding different amino acids. Planctomyces, Akkermansia, Isosphaera, and Chlamydia were similar to E. coli tRNAs. In a phylum, tRNAs coding for different amino acids clustered at a range of 8 to 10%. Further analysis of these tRNAs showed sequence similarity with Cyanobacteria, Proteobacteria, Viridiplantae, Ascomycota and Basidiomycota (Eukaryota). This indicates the possibility of horizontal gene transfer or, otherwise, a different origin of tRNA in PVC bacteria. Hence, this work proves its importance for determining evolutionary relatedness and potentially identifying bacteria using tRNA. Thus, the analysis of these tRNAs indicates that primitive RNA may have served as the genetic material of LUCA before being replaced by DNA. A quantitative analysis is required to test these possibilities that relate the evolutionary significance of tRNA to the origin of life.


Asunto(s)
Escherichia coli , ARN de Transferencia , Escherichia coli/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Verrucomicrobia/genética , Aminoácidos/metabolismo , Planctomicetos , Evolución Molecular
13.
PLoS Pathog ; 19(10): e1011683, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37788279

RESUMEN

Helminth Trichinella spiralis (Ts) is one of the major pathogens of human infective myocarditis that can lead to cardiac fibrosis (CF). The gut microbiota involved in this pathology are of interest. Here, we use mice infected with Ts as a model to examine the interactions between gut microbes and host protection to CF. Infected mice show enhanced CF severity. We find that antibiotics treatment to deplete the microbiota aggravates the disease phenotype. Attempts to restore microbiota using fecal microbiota transplantation ameliorates helminth-induced CF. 16S rRNA gene sequencing and metagenomics sequencing reveal a higher abundance of Akkermansia muciniphila in gut microbiomes of Ts-infected mice. Oral supplementation with alive or pasteurized A. muciniphila improves CF via TLR2. This work represents a substantial advance toward our understanding of causative rather than correlative relationships between the gut microbiota and CF.


Asunto(s)
Receptor Toll-Like 2 , Triquinelosis , Verrucomicrobia , Animales , Humanos , Ratones , Fibrosis , ARN Ribosómico 16S/genética , Receptor Toll-Like 2/genética , Verrucomicrobia/genética , Trichinella spiralis , Triquinelosis/inmunología
14.
Artículo en Inglés | MEDLINE | ID: mdl-37755432

RESUMEN

The thermo-acidophilic aerobic methanotrophic Verrucomicrobia bacterium, designated strain Kam1T was isolated from an acidic geothermal mud spring in Kamchatka, Russia. Kam1T is Gram-stain-negative, with non-motile cells and non-spore-forming rods, and a diameter of 0.45-0.65 µm and length of 0.8-1.0 µm. Its growth is optimal at the temperature of 55 °C (range, 37-60 °C) and pH of 2.5 (range, pH 1-6), and its maximal growth rate is ~0.11 h-1 (doubling time ~6.3 h). Its cell wall contains peptidoglycan with meso-diaminopimelic acid. In addition to growing on methane and methanol, strain Kam1T grows on acetone and 2-propanol. Phylogenetically, it forms a distinct group together with other Methylacidiphilum strains and with the candidate genus Methylacidimicrobium as a sister group. These findings support the classification of the strain Kam1T as a representative of a novel species and genus of the phylum Verrucomicrobiota. For this strain, we propose the name Methylacidiphilum kamchatkense sp. nov. as the type species within Methylacidiphilum gen. nov. Strain Kam1T (JCM 30608T=KCTC 4682T) is the type strain.


Asunto(s)
Ácidos Grasos , Verrucomicrobia , Ácidos Grasos/química , Análisis de Secuencia de ADN , Filogenia , ADN Bacteriano/genética , ARN Ribosómico 16S/genética , Composición de Base , Técnicas de Tipificación Bacteriana , Verrucomicrobia/genética
15.
Mol Metab ; 77: 101797, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37709134

RESUMEN

OBJECTIVE: Polyphenols have health-promoting effects, such as improving insulin resistance. Isoxanthohumol (IX), a prenylated flavonoid found in beer hops, has been suggested to reduce obesity and insulin resistance; however, the mechanism remains unknown. METHODS: High-fat diet-fed mice were administered IX. We analyzed glucose metabolism, gene expression profiles and histology of liver, epididymal adipose tissue and colon. Lipase activity, fecal lipid profiles and plasma metabolomic analysis were assessed. Fecal 16s rRNA sequencing was obtained and selected bacterial species were used for in vitro studies. Fecal microbiota transplantation and monocolonization were conducted to antibiotic-treated or germ-free (GF) mice. RESULTS: The administration of IX lowered weight gain, decreased steatohepatitis and improved glucose metabolism. Mechanistically, IX inhibited pancreatic lipase activity and lipid absorption by decreasing the expression of the fatty acid transporter CD36 in the small intestine, which was confirmed by increased lipid excretion in feces. IX administration increased markers of intestinal barrier function, including thickening the mucin layer and increasing caludin-1, a tight-junction related protein in the colon. In contrast, the effects of IX were nullified by antibiotics. As revealed using 16S rRNA sequencing, the microbial community structure changed with a significant increase in the abundance of Akkermansia muciniphila in the IX-treated group. An anaerobic chamber study showed that IX selectively promoted the growth of A. muciniphila while exhibiting antimicrobial activity against some Bacteroides and Clostridium species. To further explore the direct effect of A. muciniphila on lipid and glucose metabolism, we monocolonized either A. muciniphila or Bacteroides thetaiotaomicron to GF mice. A. muciniphila monocolonization decreased CD36 expression in the jejunum and improved glucose metabolism, with decreased levels of multiple classes of fatty acids determined using plasma metabolomic analysis. CONCLUSIONS: Our study demonstrated that IX prevents obesity and enhances glucose metabolism by inhibiting dietary fat absorption. This mechanism is linked to suppressing pancreatic lipase activity and shifts in microbial composition, notably an increase in A. muciniphila. These highlight new treatment strategies for preventing metabolic syndrome by boosting the gut microbiota with food components.


Asunto(s)
Resistencia a la Insulina , Animales , Ratones , ARN Ribosómico 16S/genética , Obesidad/tratamiento farmacológico , Obesidad/microbiología , Verrucomicrobia/genética , Verrucomicrobia/metabolismo , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta , Glucosa/metabolismo , Lipasa
16.
Nat Microbiol ; 8(8): 1450-1467, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37337046

RESUMEN

Akkermansia muciniphila, a mucophilic member of the gut microbiota, protects its host against metabolic disorders. Because it is genetically intractable, the mechanisms underlying mucin metabolism, gut colonization and its impact on host physiology are not well understood. Here we developed and applied transposon mutagenesis to identify genes important for intestinal colonization and for the use of mucin. An analysis of transposon mutants indicated that de novo biosynthesis of amino acids was required for A. muciniphila growth on mucin medium and that many glycoside hydrolases are redundant. We observed that mucin degradation products accumulate in internal compartments within bacteria in a process that requires genes encoding pili and a periplasmic protein complex, which we term mucin utilization locus (MUL) genes. We determined that MUL genes were required for intestinal colonization in mice but only when competing with other microbes. In germ-free mice, MUL genes were required for A. muciniphila to repress genes important for cholesterol biosynthesis in the colon. Our genetic system for A. muciniphila provides an important tool with which to uncover molecular links between the metabolism of mucins, regulation of lipid homeostasis and potential probiotic activities.


Asunto(s)
Intestinos , Mucinas , Verrucomicrobia , Animales , Ratones , Mucinas/metabolismo , Esteroles/biosíntesis , Verrucomicrobia/genética , Verrucomicrobia/crecimiento & desarrollo , Verrucomicrobia/metabolismo , Intestinos/microbiología , Organismos Libres de Patógenos Específicos , Elementos Transponibles de ADN/genética , Mutagénesis , Interacciones Microbiota-Huesped/genética , Espacio Intracelular/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transcripción Genética
18.
J Adv Res ; 52: 89-102, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36996967

RESUMEN

INTRODUCTION: The gut microbiome is vital for providing resistance against colonized pathogenicbacteria. Recently, specific commensal species have become recognized as important mediators of host defense against microbial infection by a variety of mechanisms. OBJECTIVES: To examine the contribution of live and pasteurized A. muciniphila to defend against the intestinal pathogen Salmonella Typhimurium in a streptomycin-treated mouse model of infection. METHODS: C57B6J mice were pretreated with phosphate-buffered saline (PBS), live Akkermansia muciniphila (AKK), and pasteurized A. muciniphila (pAKK) for two weeks, then mice were infected by S. Typhimurium SL 1344. 16S rRNA-based gut microbiota analysis was performed before and after infection. Bacterial counts in feces and tissues, histopathological analysis, gut barrier-related gene expression, and antimicrobial peptides were examined. Co-housing was performed to examine the role of microbiota in the change of susceptibility of mice to infection. RESULTS: AKK and pAKK markedly decreased Salmonella fecal and systemic burdens and reduced inflammation during infection. Notably, further characterization of AKK and pAKK protective mechanisms revealed different candidate protective pathways. AKK promoted gutbarrier gene expression and the secretion of antimicrobial peptides, and co-housing studies suggested that AKK-associated microbial community played a role in attenuating infection. Moreover, pAKK had a positive effect on NLRP3 in infected mice. We verified that pretreatment of pAKK could promote the expression of NLRP3, and enhance the antimicrobial activity of macrophage, likely through increasing the production of reactive oxygen (ROS), nitric oxide (NO), and inflammatory cytokines. CONCLUSION: Our study demonstrates that live or pasteurized A. muciniphila can be effective preventive measures for alleviating S. Typhimurium-induced disease, highlighting the potential of developing Akkermansia-based probiotics or postbiotics for the prevention of Salmonellosis.


Asunto(s)
Infecciones por Salmonella , Salmonella typhimurium , Ratones , Animales , Salmonella typhimurium/genética , ARN Ribosómico 16S/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Verrucomicrobia/química , Verrucomicrobia/genética , Verrucomicrobia/metabolismo , Péptidos Antimicrobianos
19.
Pharmacol Res ; 189: 106687, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36746362

RESUMEN

Accumulating evidence indicates gut microbiota contributes to aging-related disorders. However, the exact mechanism underlying gut dysbiosis-related pathophysiological changes during aging remains largely unclear. In the current study, we first performed gut microbiota remodeling on old mice by fecal microbiota transplantation (FMT) from young mice, and then characterized the bacteria signature that was specifically altered by FMT. Our results revealed that FMT significantly improved natural aging-related systemic disorders, particularly exerted hepatoprotective effects, and improved glucose sensitivity, hepatosplenomegaly, inflammaging, antioxidative capacity and intestinal barrier. Moreover, FMT particularly increased the abundance of fecal A.muciniphila, which was almost nondetectable in old mice. Interestingly, A.muciniphila supplementation also exerted similar benefits with FMT on old mice. Notably, targeted metabolomics on short chain fatty acids (SCFAs) revealed that only acetic acid was consistently reversed by FMT. Then, acetic acid intervention exerted beneficial actions on both Caenorhabditis elegans and natural aging mice. In conclusion, our current study demonstrated that gut microbiota remodeling improved natural aging-related disorders through A.muciniphila and its derived acetic acid, suggesting that interventions with potent stimulative capacity on A. muciniphila growth and production of acetic acid was alternative and effective way to maintain healthy aging. DATA AVAILABILITY STATEMENT: The data of RNAseq and 16 S rRNA gene sequencing can be accessed in NCBI with the accession number PRJNA848996 and PRJNA849355.


Asunto(s)
Microbioma Gastrointestinal , Ratones , Animales , Microbioma Gastrointestinal/genética , Ácido Acético , Verrucomicrobia/genética , Trasplante de Microbiota Fecal/métodos
20.
Nat Microbiol ; 8(3): 424-440, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36759753

RESUMEN

The molecular bases of how host genetic variation impacts the gut microbiome remain largely unknown. Here we used a genetically diverse mouse population and applied systems genetics strategies to identify interactions between host and microbe phenotypes including microbial functions, using faecal metagenomics, small intestinal transcripts and caecal lipids that influence microbe-host dynamics. Quantitative trait locus (QTL) mapping identified murine genomic regions associated with variations in bacterial taxa; bacterial functions including motility, sporulation and lipopolysaccharide production and levels of bacterial- and host-derived lipids. We found overlapping QTL for the abundance of Akkermansia muciniphila and caecal levels of ornithine lipids. Follow-up in vitro and in vivo studies revealed that A. muciniphila is a major source of these lipids in the gut, provided evidence that ornithine lipids have immunomodulatory effects and identified intestinal transcripts co-regulated with these traits including Atf3, which encodes for a transcription factor that plays vital roles in modulating metabolism and immunity. Collectively, these results suggest that ornithine lipids are potentially important for A. muciniphila-host interactions and support the role of host genetics as a determinant of responses to gut microbes.


Asunto(s)
Microbioma Gastrointestinal , Verrucomicrobia , Ratones , Animales , Verrucomicrobia/genética , Microbioma Gastrointestinal/genética , Akkermansia/genética , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...