Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.540
Filtrar
1.
Curr Microbiol ; 81(9): 267, 2024 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-39003673

RESUMEN

In this study, we evaluated the impact of human gut microbiota on the immune pathways in the respiratory tract using a gnotobiotic (Gn) piglet model. We humanized piglets with rural and urban infant fecal microbiota (RIFM and UIFM, respectively) and then infected them with a H1N1 swine influenza virus. We analyzed the microbial diversity and structure of the intestinal and respiratory tracts of the piglets before and after the influenza virus infection and measured the viral load and immune responses. We found that the viral load in the upper respiratory tract of UIFM transplanted piglets was higher than their rural cohorts (RIFM), while virus-specific antibody responses were comparable. The relative cytokine gene expression in the tracheobronchial (respiratory tract) and mesenteric (gastrointestinal) lymph nodes, lungs, blood, and spleen of RIFM and UIFM piglets revealed a trend in reciprocal regulation of proinflammatory, innate, and adaptive immune-associated cytokines as well as the frequency of T-helper/memory cells, cytotoxic T cells, and myeloid immune cell subsets. We also observed different phylum-level shifts of the fecal microbiota in response to influenza virus infection between the two piglet groups, suggesting the potential impact of the gut microbiota on the immune responses to influenza virus infection and lung microbiota. In conclusion, Gn piglets humanized with diverse infant fecal microbiota had differential immune regulation, with UIFM favoring the activation of proinflammatory immune mediators following an influenza virus infection compared to their rural RIFM cohorts. Furthermore, Gn piglets can be a useful model in investigating the impact of diverse human microbiota of the gastrointestinal tract, probably also the respiratory tract, on respiratory health and testing specific probiotic- or prebiotic-based therapeutics.


Asunto(s)
Citocinas , Modelos Animales de Enfermedad , Heces , Microbioma Gastrointestinal , Vida Libre de Gérmenes , Inmunidad Mucosa , Subtipo H1N1 del Virus de la Influenza A , Animales , Porcinos , Heces/microbiología , Heces/virología , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Citocinas/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Carga Viral , Lactante , Gripe Humana/inmunología , Gripe Humana/microbiología , Gripe Humana/virología
2.
Cell Host Microbe ; 32(7): 1163-1176.e6, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38906158

RESUMEN

Depletion of beneficial microbes by modern lifestyle factors correlates with the rising prevalence of food allergies. Re-introduction of allergy-protective bacteria may be an effective treatment strategy. We characterized the fecal microbiota of healthy and food-allergic infants and found that the anaerobe Anaerostipes caccae (A. caccae) was representative of the protective capacity of the healthy microbiota. We isolated a strain of A. caccae from the feces of a healthy infant and identified lactulose as a prebiotic to optimize butyrate production by A. caccae in vitro. Administration of a synbiotic composed of our isolated A. caccae strain and lactulose increased luminal butyrate in gnotobiotic mice colonized with feces from an allergic infant and in antibiotic-treated specific pathogen-free (SPF) mice, and prevented or treated an anaphylactic response to allergen challenge. The synbiotic's efficacy in two models and microbial contexts suggests that it may be a promising approach for the treatment of food allergy.


Asunto(s)
Heces , Hipersensibilidad a los Alimentos , Microbioma Gastrointestinal , Lactulosa , Simbióticos , Animales , Simbióticos/administración & dosificación , Hipersensibilidad a los Alimentos/prevención & control , Ratones , Humanos , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Lactante , Butiratos/metabolismo , Prebióticos/administración & dosificación , Femenino , Modelos Animales de Enfermedad , Organismos Libres de Patógenos Específicos , Vida Libre de Gérmenes , Masculino
3.
Cell Host Microbe ; 32(7): 1103-1113.e6, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38838675

RESUMEN

Antibiotic treatment promotes the outgrowth of intestinal Candida albicans, but the mechanisms driving this fungal bloom remain incompletely understood. We identify oxygen as a resource required for post-antibiotic C. albicans expansion. C. albicans depleted simple sugars in the ceca of gnotobiotic mice but required oxygen to grow on these resources in vitro, pointing to anaerobiosis as a potential factor limiting growth in the gut. Clostridia species limit oxygen availability in the large intestine by producing butyrate, which activates peroxisome proliferator-activated receptor gamma (PPAR-γ) signaling to maintain epithelial hypoxia. Streptomycin treatment depleted Clostridia-derived butyrate to increase epithelial oxygenation, but the PPAR-γ agonist 5-aminosalicylic acid (5-ASA) functionally replaced Clostridia species to restore epithelial hypoxia and colonization resistance against C. albicans. Additionally, probiotic Escherichia coli required oxygen respiration to prevent a post-antibiotic bloom of C. albicans, further supporting the role of oxygen in colonization resistance. We conclude that limited access to oxygen maintains colonization resistance against C. albicans.


Asunto(s)
Candida albicans , Oxígeno , Candida albicans/efectos de los fármacos , Animales , Ratones , Oxígeno/metabolismo , PPAR gamma/metabolismo , Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Candidiasis/microbiología , Anaerobiosis , Hipoxia/metabolismo , Ratones Endogámicos C57BL , Estreptomicina/farmacología , Humanos , Ciego/microbiología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Vida Libre de Gérmenes
4.
Methods Mol Biol ; 2820: 127-137, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38941020

RESUMEN

Intestinal fungi are a fundamental component of the gut microbiome and play important roles in mammalian host biology. At the same time, the contribution of gut fungi to host health and disease remains understudied due to their low abundance. In that respect, gnotobiotic animals with defined microbial populations of reduced complexity represent a well-suited model system that highlights the effects of low abundant gut fungi on host physiology and other members of the microbial community. In this chapter, a label-free quantitative metaproteomic approach for the characterization of simplified microbial communities in gnotobiotic mice is presented. The model allows for exploring various research questions on the role of gut fungi in disease pathogenesis, microbial ecosystem maturation, or host-microbiome crosstalk.


Asunto(s)
Hongos , Microbioma Gastrointestinal , Vida Libre de Gérmenes , Proteómica , Animales , Ratones , Proteómica/métodos , Hongos/metabolismo
5.
Gut Microbes ; 16(1): 2361660, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38935764

RESUMEN

The microbiota significantly impacts digestive epithelium functionality, especially in nutrient processing. Given the importance of iron for both the host and the microbiota, we hypothesized that host-microbiota interactions fluctuate with dietary iron levels. We compared germ-free (GF) and conventional mice (SPF) fed iron-containing (65 mg/Kg) or iron-depleted (<6 mg/Kg) diets. The efficacy of iron privation was validated by iron blood parameters. Ferritin and Dmt1, which represent cellular iron storage and transport respectively, were studied in tissues where they are abundant: the duodenum, liver and lung. When the mice were fed an iron-rich diet, the microbiota increased blood hemoglobin and hepcidin and the intestinal ferritin levels, suggesting that the microbiota helps iron storage. When iron was limiting, the microbiota inhibited the expression of the intestinal Dmt1 transporter, likely via the pathway triggered by Hif-2α. The microbiota assists the host in storing intestinal iron when it is abundant and competes with the host by inhibiting Dmt1 in conditions of iron scarcity. Comparison between duodenum, liver and lung indicates organ-specific responses to microbiota and iron availability. Iron depletion induced temporal changes in microbiota composition and activity, reduced α-diversity of microbiota, and led to Lactobacillaceae becoming particularly more abundant after 60 days of privation. By inoculating GF mice with a simplified bacterial mixture, we show that the iron-depleted host favors the gut fitness of Bifidobacterium longum.


Asunto(s)
Proteínas de Transporte de Catión , Duodeno , Microbioma Gastrointestinal , Hepcidinas , Hierro de la Dieta , Hígado , Animales , Ratones , Microbioma Gastrointestinal/fisiología , Hierro de la Dieta/metabolismo , Hierro de la Dieta/administración & dosificación , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Hígado/metabolismo , Hígado/microbiología , Duodeno/metabolismo , Duodeno/microbiología , Hepcidinas/metabolismo , Ferritinas/metabolismo , Vida Libre de Gérmenes , Interacciones Microbiota-Huesped , Pulmón/microbiología , Pulmón/metabolismo , Hierro/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/genética , Bacterias/aislamiento & purificación , Ratones Endogámicos C57BL , Hemoglobinas/metabolismo , Masculino
6.
Cell Host Microbe ; 32(6): 820-836, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38870899

RESUMEN

Microbial communities that colonize the human gastrointestinal (GI) tract defend against pathogens through a mechanism known as colonization resistance (CR). Advances in technologies such as next-generation sequencing, gnotobiotic mouse models, and bacterial cultivation have enhanced our understanding of the underlying mechanisms and the intricate microbial interactions involved in CR. Rather than being attributed to specific microbial clades, CR is now understood to arise from a dynamic interplay between microbes and the host and is shaped by metabolic, immune, and environmental factors. This evolving perspective underscores the significance of contextual factors, encompassing microbiome composition and host conditions, in determining CR. This review highlights recent research that has shifted its focus toward elucidating how these factors interact to either promote or impede enteric infections. It further discusses future research directions to unravel the complex relationship between host, microbiota, and environmental determinants in safeguarding against GI infections to promote human health.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Animales , Ratones , Interacciones Microbiota-Huesped , Tracto Gastrointestinal/microbiología , Bacterias/genética , Bacterias/clasificación , Interacciones Huésped-Patógeno , Vida Libre de Gérmenes , Interacciones Microbianas
7.
Gut Microbes ; 16(1): 2363015, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38845453

RESUMEN

Gut microbiota is responsible for essential functions in human health. Several communication axes between gut microbiota and other organs via neural, endocrine, and immune pathways have been described, and perturbation of gut microbiota composition has been implicated in the onset and progression of an emerging number of diseases. Here, we analyzed peripheral nerves, dorsal root ganglia (DRG), and skeletal muscles of neonatal and young adult mice with the following gut microbiota status: a) germ-free (GF), b) gnotobiotic, selectively colonized with 12 specific gut bacterial strains (Oligo-Mouse-Microbiota, OMM12), or c) natural complex gut microbiota (CGM). Stereological and morphometric analyses revealed that the absence of gut microbiota impairs the development of somatic median nerves, resulting in smaller diameter and hypermyelinated axons, as well as in smaller unmyelinated fibers. Accordingly, DRG and sciatic nerve transcriptomic analyses highlighted a panel of differentially expressed developmental and myelination genes. Interestingly, the type III isoform of Neuregulin1 (NRG1), known to be a neuronal signal essential for Schwann cell myelination, was overexpressed in young adult GF mice, with consequent overexpression of the transcription factor Early Growth Response 2 (Egr2), a fundamental gene expressed by Schwann cells at the onset of myelination. Finally, GF status resulted in histologically atrophic skeletal muscles, impaired formation of neuromuscular junctions, and deregulated expression of related genes. In conclusion, we demonstrate for the first time a gut microbiota regulatory impact on proper development of the somatic peripheral nervous system and its functional connection to skeletal muscles, thus suggesting the existence of a novel 'Gut Microbiota-Peripheral Nervous System-axis.'


Asunto(s)
Ganglios Espinales , Microbioma Gastrointestinal , Unión Neuromuscular , Animales , Unión Neuromuscular/microbiología , Ratones , Ganglios Espinales/metabolismo , Ganglios Espinales/microbiología , Vida Libre de Gérmenes , Nervios Periféricos/microbiología , Nervios Periféricos/crecimiento & desarrollo , Músculo Esquelético/microbiología , Ratones Endogámicos C57BL , Neurregulina-1/metabolismo , Neurregulina-1/genética , Masculino , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Células de Schwann/microbiología , Células de Schwann/metabolismo
8.
Int J Mol Sci ; 25(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38892368

RESUMEN

Intestinal epithelium renewal strictly depends on fine regulation between cell proliferation, differentiation, and apoptosis. While murine intestinal microbiota has been shown to modify some epithelial cell kinetics parameters, less is known about the role of the human intestinal microbiota. Here, we investigated the rate of intestinal cell proliferation in C3H/HeN germ-free mice associated with human flora (HFA, n = 8), and in germ-free (n = 15) and holoxenic mice (n = 16). One hour before sacrifice, all mice were intraperitoneally inoculated with 5-bromodeoxyuridine (BrdU), and the number of BrdU-positive cells/total cells (labelling index, LI), both in the jejunum and the colon, was evaluated by immunohistochemistry. Samples were also observed by scanning electron microscopy (SEM). Moreover, the microbiota composition in the large bowel of the HFA mice was compared to that of of human donor's fecal sample. No differences in LI were found in the small bowels of the HFA, holoxenic, and germ-free mice. Conversely, the LI in the large bowel of the HFA mice was significantly higher than that in the germ-free and holoxenic counterparts (p = 0.017 and p = 0.048, respectively). In the holoxenic and HFA mice, the SEM analysis disclosed different types of bacteria in close contact with the intestinal epithelium. Finally, the colonic microbiota composition of the HFA mice widely overlapped with that of the human donor in terms of dominant populations, although Bifidobacteria and Lactobacilli disappeared. Despite the small sample size analyzed in this study, these preliminary findings suggest that human intestinal microbiota may promote a high proliferation rate of colonic mucosa. In light of the well-known role of uncontrolled proliferation in colorectal carcinogenesis, these results may deserve further investigation in a larger population study.


Asunto(s)
Proliferación Celular , Colon , Microbioma Gastrointestinal , Mucosa Intestinal , Animales , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Ratones , Colon/microbiología , Colon/metabolismo , Masculino , Vida Libre de Gérmenes , Femenino , Ratones Endogámicos C3H , Heces/microbiología
9.
Artículo en Inglés | MEDLINE | ID: mdl-38901159

RESUMEN

Lipidomics is focusing on the screening of lipid species in complex mixtures using mass spectrometry-based approaches. In this work, we aim to enhance the intestinal lipidome coverage within the Oligo-Mouse-Microbiota (OMM12) colonized mouse model by testing eight mobile phase conditions on five reversed-phase columns. Our selected mobile phase modifiers included two ammonium salts, two concentrations, and the addition of respective acids at 0.1 %. We compared two columns with hybrid surface technology, two with ethylene bridged hybrid technology and one with core-shell particles. Best performance was attained for standards and intestinal lipidome, using either ammonium formate or acetate in ESI(+) or ammonium acetate in ESI(-) for all column technologies. Notably, a concentration of 5 mM ammonium salt showed optimal results for both modes, while the addition of acids had a negligible effect on lipid ionization efficiency. The HST BEH C18 column improved peak width and tailing factor parameters compared to other technologies. We achieved the highest lipid count in colon and ileum content, including ceramides, phosphatidylethanolamines and phosphatidylcholines, when using 5 mM ammonium acetate in ESI(-). Conversely, in ESI(+) 5 mM ammonium formate demonstrated superior coverage for diacylglycerols and triacylglycerols.


Asunto(s)
Vida Libre de Gérmenes , Lipidómica , Lípidos , Animales , Ratones , Cromatografía Líquida de Alta Presión/métodos , Lipidómica/métodos , Lípidos/análisis , Lípidos/química , Espectrometría de Masas/métodos , Microbioma Gastrointestinal , Intestinos/química
10.
FASEB J ; 38(11): e23648, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38822661

RESUMEN

Previous studies on germ-free (GF) animals have described altered anxiety-like and social behaviors together with dysregulations in brain serotonin (5-HT) metabolism. Alterations in circulating 5-HT levels and gut 5-HT metabolism have also been reported in GF mice. In this study, we conducted an integrative analysis of various behaviors as well as markers of 5-HT metabolism in the brain and along the GI tract of GF male mice compared with conventional (CV) ones. We found a strong decrease in locomotor activity, accompanied by some signs of increased anxiety-like behavior in GF mice compared with CV mice. Brain gene expression analysis showed no differences in HTR1A and TPH2 genes. In the gut, we found decreased TPH1 expression in the colon of GF mice, while it was increased in the cecum. HTR1A expression was dramatically decreased in the colon, while HTR4 expression was increased both in the cecum and colon of GF mice compared with CV mice. Finally, SLC6A4 expression was increased in the ileum and colon of GF mice compared with CV mice. Our results add to the evidence that the microbiota is involved in regulation of behavior, although heterogeneity among studies suggests a strong impact of genetic and environmental factors on this microbiota-mediated regulation. While no impact of GF status on brain 5-HT was observed, substantial differences in gut 5-HT metabolism were noted, with tissue-dependent results indicating a varying role of microbiota along the GI tract.


Asunto(s)
Conducta Animal , Vida Libre de Gérmenes , Serotonina , Animales , Serotonina/metabolismo , Ratones , Masculino , Microbioma Gastrointestinal/fisiología , Encéfalo/metabolismo , Triptófano Hidroxilasa/metabolismo , Triptófano Hidroxilasa/genética , Ansiedad/metabolismo , Ansiedad/microbiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Ratones Endogámicos C57BL , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT1A/genética , Colon/metabolismo , Colon/microbiología
11.
Cell Host Microbe ; 32(6): 925-944.e10, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38754417

RESUMEN

Hormones and neurotransmitters are essential to homeostasis, and their disruptions are connected to diseases ranging from cancer to anxiety. The differential reactivation of endobiotic glucuronides by gut microbial ß-glucuronidase (GUS) enzymes may influence interindividual differences in the onset and treatment of disease. Using multi-omic, in vitro, and in vivo approaches, we show that germ-free mice have reduced levels of active endobiotics and that distinct gut microbial Loop 1 and FMN GUS enzymes drive hormone and neurotransmitter reactivation. We demonstrate that a range of FDA-approved drugs prevent this reactivation by intercepting the catalytic cycle of the enzymes in a conserved fashion. Finally, we find that inhibiting GUS in conventional mice reduces free serotonin and increases its inactive glucuronide in the serum and intestines. Our results illuminate the indispensability of gut microbial enzymes in sustaining endobiotic homeostasis and indicate that therapeutic disruptions of this metabolism promote interindividual response variabilities.


Asunto(s)
Microbioma Gastrointestinal , Glucuronidasa , Homeostasis , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Glucuronidasa/metabolismo , Ratones Endogámicos C57BL , Serotonina/metabolismo , Glucurónidos/metabolismo , Humanos , Intestinos/microbiología , Masculino , Vida Libre de Gérmenes
12.
Infect Immun ; 92(6): e0006524, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38722167

RESUMEN

Giardia lamblia is an important protozoan cause of diarrheal disease worldwide, delayed development and cognitive impairment in children in low- and middle-income countries, and protracted post-infectious syndromes in developed regions. G. lamblia resides in the lumen and at the epithelial surface of the proximal small intestine but is not mucosa invasive. The protozoan parasite is genetically diverse with significant genome differences across strains and assemblages. Animal models, particularly murine models, have been instrumental in defining mechanisms of host defense against G. lamblia, but mice cannot be readily infected with most human pathogenic strains. Antibiotic pretreatment can increase susceptibility, suggesting that the normal microbiota plays a role in controlling G. lamblia infection in mice, but the broader implications on susceptibility to diverse strains are not known. Here, we have used gnotobiotic mice to demonstrate that robust intestinal infection can be achieved for a broad set of human-pathogenic strains of the genetic assemblages A and B. Furthermore, gnotobiotic mice were able to eradicate infection with a similar kinetics to conventional mice after trophozoite challenge. Germ-free mice could also be effectively immunized by the mucosal route with a protective antigen, α1-giardin, in a manner dependent on CD4 T cells. These results indicate that the gnotobiotic mouse model is powerful for investigating acquired host defenses in giardiasis, as the mice are broadly susceptible to diverse G. lamblia strains yet display no apparent defects in mucosal immunity needed for controlling and eradicating this lumen-dwelling pathogen.


Asunto(s)
Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Giardia lamblia , Giardiasis , Animales , Giardiasis/inmunología , Giardiasis/parasitología , Giardia lamblia/inmunología , Giardia lamblia/genética , Ratones , Vacunas Antiprotozoos/inmunología , Vacunación , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/parasitología , Humanos , Femenino
13.
Cell Host Microbe ; 32(6): 1025-1036.e5, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38795710

RESUMEN

The extent to which bacterial lipids produced by the gut microbiota penetrate host tissues is unclear. Here, we combined mass spectrometry approaches to identify lipids produced by the human gut symbiont Bacteroides thetaiotaomicron (B. theta) and spatially track these bacterial lipids in the mouse colon. We characterize 130 B. theta lipids by liquid chromatography-tandem mass spectrometry (LC-MS/MS), using wild-type and mutant B. theta strains to confidently identify lipid structures and their interconnected pathways in vitro. Of these, 103 B. theta lipids can be detected and spatially mapped in a single MALDI mass spectrometry imaging run. We map unlabeled bacterial lipids across colon sections of germ-free and specific-pathogen-free (SPF) mice and mice mono-colonized with wild-type or sphingolipid-deficient (BTMUT) B. theta. We observe co-localization of bacterially derived phosphatidic acid with host tissues in BTMUT mice, consistent with lipid penetration into host tissues. These results indicate limited and selective transfer of bacterial lipids to the host.


Asunto(s)
Bacteroides thetaiotaomicron , Colon , Microbioma Gastrointestinal , Lipidómica , Animales , Ratones , Bacteroides thetaiotaomicron/metabolismo , Microbioma Gastrointestinal/fisiología , Colon/microbiología , Colon/metabolismo , Lípidos/análisis , Espectrometría de Masas en Tándem , Cromatografía Liquida , Metabolismo de los Lípidos , Vida Libre de Gérmenes , Organismos Libres de Patógenos Específicos , Ácidos Fosfatidicos/metabolismo , Humanos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Esfingolípidos/metabolismo , Ratones Endogámicos C57BL , Femenino
14.
Cell Host Microbe ; 32(6): 887-899.e6, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38806059

RESUMEN

Inflammation boosts the availability of electron acceptors in the intestinal lumen, creating a favorable niche for pathogenic Enterobacteriaceae. However, the mechanisms linking intestinal inflammation-mediated changes in luminal metabolites and pathogen expansion remain unclear. Here, we show that mucosal inflammation induced by Salmonella enterica serovar Typhimurium (S. Tm) infection increases intestinal levels of the amino acid aspartate. S. Tm used aspartate-ammonia lyase (aspA)-dependent fumarate respiration for growth in the murine gut only during inflammation. AspA-dependent growth advantage was abolished in the gut of germ-free mice and restored in gnotobiotic mice colonized with members of the classes Bacteroidia and Clostridia. Reactive oxygen species (ROS) produced during the host response caused lysis of commensal microbes, resulting in the release of microbiota-derived aspartate that was used by S. Tm, in concert with nitrate-dependent anaerobic respiration, to outcompete commensal Enterobacteriaceae. Our findings demonstrate the role of microbiota-derived amino acids in driving respiration-dependent S. Tm expansion during colitis.


Asunto(s)
Ácido Aspártico , Microbioma Gastrointestinal , Especies Reactivas de Oxígeno , Salmonella typhimurium , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Ácido Aspártico/metabolismo , Colitis/microbiología , Colitis/metabolismo , Ratones Endogámicos C57BL , Enterobacteriaceae/metabolismo , Vida Libre de Gérmenes , Inflamación/microbiología , Inflamación/metabolismo , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/inmunología
15.
Pharmacol Res ; 205: 107231, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38815878

RESUMEN

We previously demonstrated that mice carrying natural mtDNA variants of the FVB/NJ strain (m.7778 G>T in the mt-Atp8 gene in mitochondrial complex V), namely C57BL/6 J-mtFVB/NJ (B6-mtFVB), exhibited (i) partial protection from experimental skin inflammatory diseases in an anti-murine type VII collagen antibody-induced skin inflammation model and psoriasiform dermatitis model; (ii) significantly altered metabolites, including short-chain fatty acids, according to targeted metabolomics of liver, skin and lymph node samples; and (iii) a differential composition of the gut microbiota according to bacterial 16 S rRNA gene sequencing of stool samples compared to wild-type C57BL/6 J (B6) mice. To further dissect these disease-contributing factors, we induced an experimental antibody-induced skin inflammatory disease in gnotobiotic mice. We performed shotgun metagenomic sequencing of caecum contents and untargeted metabolomics of liver, CD4+ T cell, and caecum content samples from conventional B6-mtFVB and B6 mice. We identified D-glucosamine as a candidate mediator that ameliorated disease severity in experimental antibody-induced skin inflammation by modulating immune cell function in T cells, neutrophils and macrophages. Because mice carrying mtDNA variants of the FVB/NJ strain show differential disease susceptibility to a wide range of experimental diseases, including diet-induced atherosclerosis in low-density lipoprotein receptor knockout mice and collagen antibody-induced arthritis in DBA/1 J mice, this experimental approach is valuable for identifying novel therapeutic options for skin inflammatory conditions and other chronic inflammatory diseases to which mice carrying specific mtDNA variants show differential susceptibility.


Asunto(s)
ADN Mitocondrial , Ratones Endogámicos C57BL , Animales , ADN Mitocondrial/genética , Microbioma Gastrointestinal , Ratones , Piel/metabolismo , Piel/microbiología , Piel/patología , Dermatitis/inmunología , Dermatitis/microbiología , Dermatitis/genética , Dermatitis/tratamiento farmacológico , Dermatitis/metabolismo , Inflamación/genética , Inflamación/inmunología , Modelos Animales de Enfermedad , Masculino , Vida Libre de Gérmenes , Psoriasis/tratamiento farmacológico , Psoriasis/inmunología , Psoriasis/genética , Ciego/microbiología , Enfermedad Crónica , Femenino
16.
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
17.
Viruses ; 16(5)2024 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-38793542

RESUMEN

The suboptimal performance of rotavirus (RV) vaccines in developing countries and in animals necessitates further research on the development of novel therapeutics and control strategies. To initiate infection, RV interacts with cell-surface O-glycans, including histo-blood group antigens (HBGAs). We have previously demonstrated that certain non-pathogenic bacteria express HBGA- like substances (HBGA+) capable of binding RV particles in vitro. We hypothesized that HBGA+ bacteria can bind RV particles in the gut lumen protecting against RV species A (RVA), B (RVB), and C (RVC) infection in vivo. In this study, germ-free piglets were colonized with HBGA+ or HBGA- bacterial cocktail and infected with RVA/RVB/RVC of different genotypes. Diarrhea severity, virus shedding, immunoglobulin A (IgA) Ab titers, and cytokine levels were evaluated. Overall, colonization with HBGA+ bacteria resulted in reduced diarrhea severity and virus shedding compared to the HBGA- bacteria. Consistent with our hypothesis, the reduced severity of RV disease and infection was not associated with significant alterations in immune responses. Additionally, colonization with HBGA+ bacteria conferred beneficial effects irrespective of the piglet HBGA phenotype. These findings are the first experimental evidence that probiotic performance in vivo can be improved by including HBGA+ bacteria, providing decoy epitopes for broader/more consistent protection against diverse RVs.


Asunto(s)
Antígenos de Grupos Sanguíneos , Vida Libre de Gérmenes , Infecciones por Rotavirus , Rotavirus , Animales , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/virología , Porcinos , Rotavirus/inmunología , Antígenos de Grupos Sanguíneos/metabolismo , Antígenos de Grupos Sanguíneos/inmunología , Diarrea/virología , Diarrea/microbiología , Diarrea/prevención & control , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/inmunología , Esparcimiento de Virus , Bacterias/clasificación , Inmunoglobulina A/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Citocinas/metabolismo
18.
Cell Host Microbe ; 32(5): 661-675.e10, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38657606

RESUMEN

The intestine and liver are thought to metabolize dietary nutrients and regulate host nutrient homeostasis. Here, we find that the gut microbiota also reshapes the host amino acid (aa) landscape via efficiently metabolizing intestinal aa. To identify the responsible microbes/genes, we developed a metabolomics-based assay to screen 104 commensals and identified candidates that efficiently utilize aa. Using genetics, we identified multiple responsible metabolic genes in phylogenetically diverse microbes. By colonizing germ-free mice with the wild-type strain and their isogenic mutant deficient in individual aa-metabolizing genes, we found that these genes regulate the availability of gut and circulatory aa. Notably, microbiota genes for branched-chain amino acids (BCAAs) and tryptophan metabolism indirectly affect host glucose homeostasis via peripheral serotonin. Collectively, at single-gene level, this work characterizes a microbiota-encoded metabolic activity that affects host nutrient homeostasis and provides a roadmap to interrogate microbiota-dependent activity to improve human health.


Asunto(s)
Aminoácidos de Cadena Ramificada , Aminoácidos , Microbioma Gastrointestinal , Homeostasis , Triptófano , Animales , Microbioma Gastrointestinal/fisiología , Ratones , Aminoácidos/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Triptófano/metabolismo , Ratones Endogámicos C57BL , Nutrientes/metabolismo , Intestinos/microbiología , Humanos , Metabolómica , Glucosa/metabolismo , Serotonina/metabolismo , Vida Libre de Gérmenes , Bacterias/metabolismo , Bacterias/genética , Bacterias/clasificación , Masculino
19.
J Vis Exp ; (206)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38682933

RESUMEN

Zebrafish serve as valuable models for research on growth, immunity, and gut microbiota due to their genomic similarities with mammals, transparent embryos developed in a relatively clean chorion environment, and extremely rapid development of larvae compared to rodent models. Germ-free (GF) zebrafish (Danio rerio) are crucial for evaluating pollutant toxicity and establishing human-like disease models related to microbial functions. In comparison to conventionally raised (CR) models (fish in common husbandry), GF zebrafish allow for more accurate manipulation of the host microbiota, aiding in determining the causal relationship between microorganisms and hosts. Consequently, they play a critical role in advancing our understanding of these relationships. However, GF zebrafish models are typically generated and researched during the early life stages (from embryos to larvae) due to limitations in immune function and nutrient absorption. This study optimizes the generation, maintenance, and identification of early GF zebrafish models without feeding and with long-term feeding using GF food (such as Artemia sp., brine shrimp). Throughout the process, daily sampling and culture were performed and identified through multiple detections, including plates and 16S rRNA sequencing. The aseptic rate, survival, and developmental indexes of GF zebrafish were recorded to ensure the quality and quantity of the generated models. Importantly, this study provides details on bacterial isolation and infection techniques for GF fish, enabling the efficient creation of GF fish models from larvae to juvenile stages with GF food support. By applying these procedures in biomedical research, scientists can better understand the relationships between intestinal bacterial functions and host health.


Asunto(s)
Vida Libre de Gérmenes , Larva , Modelos Animales , Pez Cebra , Animales , Pez Cebra/microbiología , Larva/microbiología , Larva/crecimiento & desarrollo , Femenino , Masculino
20.
Am J Physiol Renal Physiol ; 326(6): F1032-F1038, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38634136

RESUMEN

The gut microbiome regulates many important host physiological processes associated with cardiovascular health and disease; however, the impact of the gut microbiome on aldosterone is unclear. Investigating whether gut microbiota regulate aldosterone can offer novel insights into how the microbiome affects blood pressure. In this study, we aimed to determine whether gut microbiota regulate host aldosterone. We used enzyme-linked immunosorbent assays (ELISAs) to assess plasma aldosterone and plasma renin activity (PRA) in female and male mice in which gut microbiota are intact, suppressed, or absent. In addition, we examined urinary aldosterone. Our findings demonstrated that when the gut microbiota is suppressed following antibiotic treatment, there is an increase in plasma and urinary aldosterone in both female and male mice. In contrast, an increase in PRA is seen only in males. We also found that when gut microbiota are absent (germ-free mice), plasma aldosterone is significantly increased compared with conventional animals (in both females and males), but PRA is not. Understanding how gut microbiota influence aldosterone levels could provide valuable insights into the development and treatment of hypertension and/or primary aldosteronism. This knowledge may open new avenues for therapeutic interventions, such as probiotics or dietary modifications to help regulate blood pressure via microbiota-based changes to aldosterone.NEW & NOTEWORTHY We explore the role of the gut microbiome in regulating aldosterone, a hormone closely linked to blood pressure and cardiovascular disease. Despite the recognized importance of the gut microbiome in host physiology, the relationship with circulating aldosterone remains largely unexplored. We demonstrate that suppression of gut microbiota leads to increased levels of plasma and urinary aldosterone. These findings underscore the potential of the gut microbiota to influence aldosterone regulation, suggesting new possibilities for treating hypertension.


Asunto(s)
Aldosterona , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Renina , Animales , Aldosterona/sangre , Aldosterona/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Femenino , Masculino , Renina/sangre , Renina/metabolismo , Presión Sanguínea/efectos de los fármacos , Vida Libre de Gérmenes , Ratones , Antibacterianos/farmacología , Hipertensión/microbiología , Hipertensión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...