Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Metab ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951660

RESUMEN

As the microbiome field moves from descriptive and associative research to mechanistic and interventional studies, being able to account for all confounding variables in the experimental design, which includes the maternal effect1, cage effect2, facility differences3, as well as laboratory and sample handling protocols4, is critical for interpretability of results. Despite significant procedural and bioinformatic improvements, unexplained variability and lack of replicability still occur. One underexplored factor is that the microbiome is dynamic and exhibits diurnal oscillations that can change microbiome composition5-7. In this retrospective analysis of 16S amplicon sequencing studies in male mice, we show that sample collection time affects the conclusions drawn from microbiome studies and its effect size is larger than those of a daily experimental intervention or dietary changes. The timing of divergence of the microbiome composition between experimental and control groups is unique to each experiment. Sample collection times as short as only 4 hours apart can lead to vastly different conclusions. Lack of consistency in the time of sample collection may explain poor cross-study replicability in microbiome research. The impact of diurnal rhythms on the outcomes and study design of other fields is unknown but likely significant.

2.
J Clin Invest ; 133(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37712426

RESUMEN

Circadian rhythms govern glucose homeostasis, and their dysregulation leads to complex metabolic diseases. Gut microbes exhibit diurnal rhythms that influence host circadian networks and metabolic processes, yet underlying mechanisms remain elusive. Here, we showed hierarchical, bidirectional communication among the liver circadian clock, gut microbes, and glucose homeostasis in mice. To assess this relationship, we utilized mice with liver-specific deletion of the core circadian clock gene Bmal1 via Albumin-cre maintained in either conventional or germ-free housing conditions. The liver clock, but not the forebrain clock, required gut microbes to drive glucose clearance and gluconeogenesis. Liver clock dysfunctionality expanded proportions and abundances of oscillating microbial features by 2-fold relative to that in controls. The liver clock was the primary driver of differential and rhythmic hepatic expression of glucose and fatty acid metabolic pathways. Absent the liver clock, gut microbes provided secondary cues that dampened these rhythms, resulting in reduced lipid fuel utilization relative to carbohydrates. All together, the liver clock transduced signals from gut microbes that were necessary for regulating glucose and lipid metabolism and meeting energy demands over 24 hours.


Asunto(s)
Relojes Circadianos , Microbioma Gastrointestinal , Animales , Ratones , Glucosa , Metabolismo de los Lípidos , Hígado
3.
Science ; 381(6657): 502-508, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37535745

RESUMEN

The mammalian gut secretes a family of multifunctional peptides that affect appetite, intestinal secretions, and motility whereas others regulate the microbiota. We have found that peptide YY (PYY1-36), but not endocrine PYY3-36, acts as an antimicrobial peptide (AMP) expressed by gut epithelial paneth cells (PC). PC-PYY is packaged into secretory granules and is secreted into and retained by surface mucus, which optimizes PC-PYY activity. Although PC-PYY shows some antibacterial activity, it displays selective antifungal activity against virulent Candida albicans hyphae-but not the yeast form. PC-PYY is a cationic molecule that interacts with the anionic surfaces of fungal hyphae to cause membrane disruption and transcriptional reprogramming that selects for the yeast phenotype. Hence, PC-PYY is an antifungal AMP that contributes to the maintenance of gut fungal commensalism.


Asunto(s)
Antifúngicos , Péptidos Antimicrobianos , Candida , Células de Paneth , Fragmentos de Péptidos , Péptido YY , Animales , Antifúngicos/metabolismo , Péptidos Antimicrobianos/metabolismo , Candida/efectos de los fármacos , Candida/fisiología , Células de Paneth/metabolismo , Fragmentos de Péptidos/metabolismo , Péptido YY/metabolismo , Simbiosis , Humanos , Ratones
4.
Gut Microbes ; 15(1): 2231590, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37431867

RESUMEN

The gut microbiota affects hepatic drug metabolism. However, gut microbial factors modulating hepatic drug metabolism are largely unknown. In this study, using a mouse model of acetaminophen (APAP)-induced hepatotoxicity, we identified a gut bacterial metabolite that controls the hepatic expression of CYP2E1 that catalyzes the conversion of APAP to a reactive, toxic metabolite. By comparing C57BL/6 substrain mice from two different vendors, Jackson (6J) and Taconic (6N), which are genetically similar but harbor different gut microbiotas, we established that the differences in the gut microbiotas result in differential susceptibility to APAP-induced hepatotoxicity. 6J mice exhibited lower susceptibility to APAP-induced hepatotoxicity than 6N mice, and such phenotypic difference was recapitulated in germ-free mice by microbiota transplantation. Comparative untargeted metabolomic analysis of portal vein sera and liver tissues between conventional and conventionalized 6J and 6N mice led to the identification of phenylpropionic acid (PPA), the levels of which were higher in 6J mice. PPA supplementation alleviated APAP-induced hepatotoxicity in 6N mice by lowering hepatic CYP2E1 levels. Moreover, PPA supplementation also reduced carbon tetrachloride-induced liver injury mediated by CYP2E1. Our data showed that previously known PPA biosynthetic pathway is responsible for PPA production. Surprisingly, while PPA in 6N mouse cecum contents is almost undetectable, 6N cecal microbiota produces PPA as well as 6J cecal microbiota in vitro, suggesting that PPA production in the 6N gut microbiota is suppressed in vivo. However, previously known gut bacteria harboring the PPA biosynthetic pathway were not detected in either 6J or 6N microbiota, suggesting the presence of as-yet-unidentified PPA-producing gut microbes. Collectively, our study reveals a novel biological function of the gut bacterial metabolite PPA in the gut-liver axis and presents a critical basis for investigating PPA as a modulator of CYP2E1-mediated liver injury and metabolic diseases.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Microbioma Gastrointestinal , Ratones , Animales , Ratones Endogámicos C57BL , Acetaminofén/toxicidad , Citocromo P-450 CYP2E1/genética
5.
bioRxiv ; 2023 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-36712061

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is multifactorial in nature, affecting over a billion people worldwide. The gut microbiome has emerged as an associative factor in NAFLD, yet mechanistic contributions are unclear. Here, we show fast food (FF) diets containing high fat, added cholesterol, and fructose/glucose drinking water differentially impact short- vs. long-term NAFLD severity and progression in conventionally-raised, but not germ-free mice. Correlation and machine learning analyses independently demonstrate FF diets induce early and specific gut microbiota changes that are predictive of NAFLD indicators, with corresponding microbial community instability relative to control-fed mice. Shotgun metagenomics showed FF diets containing high cholesterol elevate fecal pro-inflammatory effectors over time, relating to a reshaping of host hepatic metabolic and inflammatory transcriptomes. FF diet-induced gut dysbiosis precedes onset and is highly predictive of NAFLD outcomes, providing potential insights into microbially-based pathogenesis and therapeutics.

6.
Biophys J ; 122(3): 577-594, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36528790

RESUMEN

Membrane transporters mediate the passage of molecules across membranes and are essential for cellular function. While the transmembrane region of these proteins is responsible for substrate transport, often the cytoplasmic regions are required for modulating their activity. However, it can be difficult to obtain atomic-resolution descriptions of these autoregulatory domains by classical structural biology techniques, especially if they lack a single, defined structure. The betaine permease, BetP, a homotrimer, is a prominent and well-studied example of a membrane protein whose autoregulation depends on cytoplasmic N- and C-terminal segments. These domains sense and transduce changes in K+ concentration and in lipid bilayer properties caused by osmotic stress. However, structural data for these terminal domains is incomplete, which hinders a clear description of the molecular mechanism of autoregulation. Here we used microsecond-scale molecular simulations of the BetP trimer to compare reported conformations of the 45-amino-acid long C-terminal tails. The simulations provide support for the idea that the conformation derived from electron microscopy (EM) data represents a more stable global orientation of the C-terminal segment under downregulating conditions while also providing a detailed molecular description of its dynamics and highlighting specific interactions with lipids, ions, and neighboring transporter subunits. A missing piece of the molecular puzzle is the N-terminal segment, whose dynamic nature has prevented structural characterization. Using Rosetta to generate ensembles of de novo conformations in the context of the EM-derived structure robustly identifies two features of the N-terminal tail, namely 1) short helical elements and 2) an orientation that would confine potential interactions to the protomer in the counterclockwise direction (viewed from the cytoplasm). Since each C-terminal tail only contacts the protomer in the clockwise direction, these results indicate an intricate interplay between the three protomers of BetP in the downregulated protein and a multidirectionality that may facilitate autoregulation of transport.


Asunto(s)
Simportadores , Subunidades de Proteína/metabolismo , Proteínas Bacterianas/química , Modelos Moleculares , Proteínas de la Membrana/metabolismo , Homeostasis
7.
Respir Res ; 23(1): 337, 2022 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-36496380

RESUMEN

BACKGROUND: Airway instillation of bleomycin (BLM) in mice is a widely used, yet challenging, model for acute lung injury (ALI) with high variability in treatment scheme and animal outcomes among investigators. Whether the gut microbiota plays any role in the outcome of BLM-induced lung injury is currently unknown. METHODS: Intratracheal instillation of BLM into C57BL/6 mice was performed. Fecal microbiomes were analyzed by 16s rRNA amplicon and metagenomic sequencing. Germ-free mice conventionalization and fecal microbiota transfer between SPF mice were performed to determine dominant commensal species that are associated with more severe BLM response. Further, lungs and gut draining lymph nodes of the mice were analyzed by flow cytometry to define immunophenotypes associated with the BLM-sensitive microbiome. RESULTS: Mice from two SPF barrier facilities at the University of Chicago exhibited significantly different mortality and weight loss during BLM-induced lung injury. Conventionalizing germ-free mice with SPF microbiota from two different housing facilities recapitulated the respective donors' response to BLM. Fecal microbiota transfer from the facility where the mice had worse mortality into the mice in the facility with more survival rendered recipient mice more susceptible to BLM-induced weight loss in a dominant negative manner. BLM-sensitive phenotype was associated with the presence of Helicobacter and Desulfovibrio in the gut, decreased Th17-neutrophil axis during steady state, and augmented lung neutrophil accumulation during the acute phase of the injury response. CONCLUSION: The composition of gut microbiota has significant impact on BLM-induced wasting and death suggesting a role of the lung-gut axis in lung injury.


Asunto(s)
Lesión Pulmonar Aguda , Bleomicina , Ratones , Animales , Bleomicina/toxicidad , ARN Ribosómico 16S , Ratones Endogámicos C57BL , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Pulmón/patología , Pérdida de Peso
9.
Int J Mol Sci ; 23(17)2022 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-36077073

RESUMEN

Studies have begun to reveal significant connections between the gut microbiome and various retinal diseases, including age-related macular degeneration (AMD). As critical supporting tissues of the retina, the retinal pigment epithelium (RPE) and underlying choroid play a critical role in retinal homeostasis and degeneration. However, the relationship between the microbiome and RPE/choroid remains poorly understood, particularly in animal models of AMD. In order to better elucidate this role, we performed high-throughput RNA sequencing of RPE/choroid tissue in germ-free (GF) and specific pathogen-free (SPF) mice. Furthermore, utilizing a specialized laser-induced choroidal neovascularization (CNV) model that we developed, we compared CNV size and inflammatory response between GF and SPF mice. After correction of raw data, 660 differentially expressed genes (DEGs) were identified, including those involved in angiogenesis regulation, scavenger and cytokine receptor activity, and inflammatory response-all of which have been implicated in AMD pathogenesis. Among lasered mice, the GF group showed significantly decreased CNV lesion size and microglial infiltration around CNV compared to the SPF group. Together, these findings provide evidence for a potential gut-RPE/choroidal axis as well as a correlation with neovascular features of AMD.


Asunto(s)
Neovascularización Coroidal , Microbioma Gastrointestinal , Degeneración Macular , Animales , Coroides/irrigación sanguínea , Neovascularización Coroidal/genética , Neovascularización Coroidal/patología , Degeneración Macular/genética , Degeneración Macular/patología , Ratones , Ratones Endogámicos C57BL , Epitelio Pigmentado de la Retina/patología , Transcriptoma
10.
Sci Rep ; 12(1): 14491, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-36008471

RESUMEN

Trillions of microbial oscillators reside throughout the mammalian body, yet their contributions toward fundamental features of host circadian rhythms (CRs) have not been characterized. Here, we demonstrate that the microbiome contributes to host CRs in activity and thermoregulation. Mice devoid of microbes (germ-free, GF) exhibited higher-amplitude CRs in a light-dark cycle and longer circadian periods in constant darkness. Circadian entrainment to food was greater in GF mice, but resetting responses to simulated jet-lag were unaffected. Microbial transplantation with cecal contents of conventionally-raised mice normalized CRs of GF mice, indicating that the concurrent activity of gut microbes modulates host circadian networks. Obesogenic effects of high-fat diet were absent in GF mice, but some circadian-disruptive effects persisted. Transkingdom (host-microbe) interactions affect circadian period and entrainment of CRs in diverse traits, and microbes alter interactions among light- and food-entrainable circadian processes in the face of environmental (light, diet) perturbations.


Asunto(s)
Ritmo Circadiano , Microbiota , Animales , Regulación de la Temperatura Corporal , Ritmo Circadiano/fisiología , Oscuridad , Luz , Mamíferos , Ratones , Fotoperiodo
11.
Cell Host Microbe ; 30(6): 809-823.e6, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35439436

RESUMEN

Gut microbial diurnal oscillations are important diet-dependent drivers of host circadian rhythms and metabolism ensuring optimal energy balance. However, the interplay between diet, microbes, and host factors sustaining intestinal oscillations is complex and poorly understood. Here, using a mouse model, we report the host C-type lectin antimicrobial peptide Reg3γ works with key ileal microbes to orchestrate these interactions in a bidirectional manner and does not correlate with the intestinal core circadian clock. High-fat diet is the primary driver of microbial oscillators that impair host metabolic homeostasis, resulting in arrhythmic host Reg3γ expression that secondarily drives abundance and oscillation of key gut microbes. This illustrates transkingdom coordination of biological rhythms primarily influenced by diet and reciprocal sensor-effector signals between host and microbial components, ultimately driving metabolism. Restoring the gut microbiota's capacity to sense dietary signals mediated by specific host factors such as Reg3γ could be harnessed to improve metabolic dysfunction.


Asunto(s)
Relojes Circadianos , Microbioma Gastrointestinal , Ritmo Circadiano , Dieta , Dieta Alta en Grasa/efectos adversos , Metabolismo de los Lípidos
12.
Cell Host Microbe ; 30(4): 458-462, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35421343

RESUMEN

Gut microbes are mediators of organismal-level circadian rhythms, responding to and transducing environmental cues. Gut microbes also exhibit rhythms, yet their contribution to a healthy microbiome remains unclear. We present our path to identifying host-microbe circadian dynamics related to health and outline a series of forward-thinking questions requiring further exploration.


Asunto(s)
Microbioma Gastrointestinal , Ritmo Circadiano
13.
Wiley Interdiscip Rev RNA ; 13(6): e1725, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35301791

RESUMEN

N6 -methyladenosine (m6 A) is the most prevalent posttranscriptional modification in eukaryotic mRNAs. Dynamic and reversible m6 A modification regulates gene expression to control cellular processes and diverse biological functions. Growing evidence indicated that m6 A modification is involved in the homeostasis of host and microbes (mostly viruses and bacteria). Disturbance of m6 A modification affects the life cycles of viruses and bacteria, however, these microbes could in turn change host m6 A modification leading to human disease including autoimmune diseases and cancer. Thus, we raise the concept that m6 A could be a "messenger" molecule to participate in the interactions between host and microbes. In this review, we summarize the regulatory mechanisms of m6 A modification on viruses and commensal microbiota, highlight the roles of m6 A methylation in the interaction of host and microbes, and finally discuss drugs development targeting m6 A modification. This article is categorized under: RNA in Disease and Development > RNA in Disease.


Asunto(s)
Adenosina , Virus , Humanos , Adenosina/metabolismo , Metilación , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Virus/genética , ARN/metabolismo
14.
Transl Vis Sci Technol ; 10(9): 14, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34388237

RESUMEN

Purpose: Compelling new evidence reveals a close link between the gut microbiome and the pathogenesis of neovascular age-related macular degeneration (nAMD). Germ-free (GF) animal models are the current gold standard for studying host the microbe interactions in vivo; yet, no GF animal models of nAMD are available today. This protocol describes gnotobiotic operations and assembly for a laser-induced choroidal neovascularization (CNV) model in GF mice to study the gut microbiome in neovascular AMD. Methods: We developed a step-wise approach to performing retinal laser photocoagulation in GF C57BL/6J mice that were bred and maintained at the gnotobiotic facility. Following a strict sterility protocol, we administered laser photocoagulation via an Argon 532-nm laser attached to a customized slit-lamp delivery system. Sterility was confirmed by weekly fecal cultures and reverse transcriptase-polymerase chain reaction. Results: The experiment was repeated twice at different time points using seven mice (14 eyes). Stool cultures and RT-PCR remained negative for 14 days post-procedure in all mice. Lectin immunostaining performed on choroidal flatmounts confirmed the presence of CNV lesions 2 weeks after laser treatment. Conclusions: We established a GF mouse model of nAMD with detailed guidelines to deliver retinal laser in GF mice maintaining sterility after the laser procedure. Translational Relevance: To our knowledge, this is the first protocol that describes a GF murine model of laser-induced CNV. In addition to nAMD, this animal model can be used to investigate host-microbial interactions in other eye diseases with laser-induced mouse models such as glaucoma and retinal vein occlusion.


Asunto(s)
Neovascularización Coroidal , Degeneración Macular Húmeda , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Neovascularización Coroidal/etiología , Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Rayos Láser , Ratones , Ratones Endogámicos C57BL , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Agudeza Visual
15.
Cells ; 10(8)2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34440888

RESUMEN

The relationship between retinal disease, diet, and the gut microbiome has shown increasing importance over recent years. In particular, high-fat diets (HFDs) are associated with development and progression of several retinal diseases, including age-related macular degeneration (AMD) and diabetic retinopathy. However, the complex, overlapping interactions between diet, gut microbiome, and retinal homeostasis are poorly understood. Using high-throughput RNA-sequencing (RNA-seq) of whole retinas, we compare the retinal transcriptome from germ-free (GF) mice on a regular diet (ND) and HFD to investigate transcriptomic changes without influence of gut microbiome. After correction of raw data, 53 differentially expressed genes (DEGs) were identified, of which 19 were upregulated and 34 were downregulated in GF-HFD mice. Key genes involved in retinal inflammation, angiogenesis, and RPE function were identified. Enrichment analysis revealed that the top 3 biological processes affected were regulation of blood vessel diameter, inflammatory response, and negative regulation of endopeptidase. Molecular functions altered include endopeptidase inhibitor activity, protease binding, and cysteine-type endopeptidase inhibitor activity. Human and mouse pathway analysis revealed that the complement and coagulation cascades are significantly affected by HFD. This study demonstrates novel data that diet can directly modulate the retinal transcriptome independently of the gut microbiome.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/fisiología , Retina/metabolismo , Animales , Degeneración Macular/metabolismo , Degeneración Macular/microbiología , Masculino , Ratones , Análisis de Secuencia de ARN , Transcriptoma/genética
16.
Gastroenterology ; 161(3): 940-952.e15, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34111469

RESUMEN

BACKGROUND & AIMS: Perturbations in the early-life gut microbiome are associated with increased risk for complex immune disorders like inflammatory bowel diseases. We previously showed that maternal antibiotic-induced gut dysbiosis vertically transmitted to offspring increases experimental colitis risk in interleukin (IL) 10 gene deficient (IL10-/-) mice, a finding that may result from the loss/lack of essential microbes needed for appropriate immunologic education early in life. Here, we aimed to identify key microbes required for proper development of the early-life gut microbiome that decrease colitis risk in genetically susceptible animals. METHODS: Metagenomic sequencing followed by reconstruction of metagenome-assembled genomes was performed on fecal samples of IL10-/- mice with and without antibiotic-induced dysbiosis to identify potential missing microbial members needed for immunologic education. One high-value target strain was then engrafted early and/or late into the gut microbiomes of IL10-/- mice with antibiotic-induced dysbiosis. RESULTS: Early-, but not late-, life engraftment of a single dominant Bacteroides strain of non-antibiotic-treated IL10-/- mice was sufficient to restore the development of the gut microbiome, promote immune tolerance, and prevent colitis in IL10-/- mice that had antibiotic-induced dysbiosis. CONCLUSIONS: Restitution of a keystone microbial strain missing in the early-life antibiotic-induced gut dysbiosis results in recovery of the microbiome, proper development of immune tolerance, and reduced risk for colitis in genetically prone hosts.


Asunto(s)
Bacteroides/crecimiento & desarrollo , Colitis/prevención & control , Colon/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Interleucina-10/deficiencia , Animales , Antibacterianos , Bacteroides/inmunología , Colitis/inmunología , Colitis/metabolismo , Colitis/microbiología , Colon/inmunología , Colon/metabolismo , Colon/patología , Modelos Animales de Enfermedad , Disbiosis , Heces/microbiología , Interacciones Huésped-Patógeno , Tolerancia Inmunológica , Interleucina-10/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Prueba de Estudio Conceptual , Factores de Tiempo
17.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33658361

RESUMEN

The human GlyT1 glycine transporter requires chloride for its function. However, the mechanism by which Cl- exerts its influence is unknown. To examine the role that Cl- plays in the transport cycle, we measured the effect of Cl- on both glycine binding and conformational changes. The ability of glycine to displace the high-affinity radioligand [3H]CHIBA-3007 required Na+ and was potentiated over 1,000-fold by Cl- We generated GlyT1b mutants containing reactive cysteine residues in either the extracellular or cytoplasmic permeation pathways and measured changes in the reactivity of those cysteine residues as indicators of conformational changes in response to ions and substrate. Na+ increased accessibility in the extracellular pathway and decreased it in the cytoplasmic pathway, consistent with stabilizing an outward-open conformation as observed in other members of this transporter family. In the presence of Na+, both glycine and Cl- independently shifted the conformation of GlyT1b toward an outward-closed conformation. Together, Na+, glycine, and Cl- stabilized an inward-open conformation of GlyT1b. We then examined whether Cl- acts by interacting with a conserved glutamine to allow formation of an ion pair that stabilizes the closed state of the extracellular pathway. Molecular dynamics simulations of a GlyT1 homolog indicated that this ion pair is formed more frequently as that pathway closes. Mutation of the glutamine blocked the effect of Cl-, and substituting it with glutamate or lysine resulted in outward- or inward-facing transporter conformations, respectively. These results provide an unexpected insight into the role of Cl- in this family of transporters.


Asunto(s)
Cloruros/química , Proteínas de Transporte de Glicina en la Membrana Plasmática/química , Simulación de Dinámica Molecular , Línea Celular , Cloruros/metabolismo , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Humanos , Transporte Iónico , Conformación Proteica , Sodio/química , Sodio/metabolismo
18.
Cell Mol Gastroenterol Hepatol ; 11(2): 491-502, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32835897

RESUMEN

BACKGROUND & AIMS: Inflammatory bowel diseases (IBD) are chronic inflammatory disorders where predictive biomarkers for the disease development and clinical course are sorely needed for development of prevention and early intervention strategies that can be implemented to improve clinical outcomes. Since gut microbiome alterations can reflect and/or contribute to impending host health changes, we examined whether gut microbiota metagenomic profiles would provide more robust measures for predicting disease outcomes in colitis-prone hosts. METHODS: Using the interleukin (IL) 10 gene-deficient (IL10 KO) murine model where early life dysbiosis from antibiotic (cefoperozone [CPZ]) treated dams vertically transferred to pups increases risk for colitis later in life, we investigated temporal metagenomic profiles in the gut microbiota of post-weaning offspring and determined their relationship to eventual clinical outcomes. RESULTS: Compared to controls, offspring acquiring maternal CPZ-induced dysbiosis exhibited a restructuring of intestinal microbial membership in both bacteriome and mycobiome that was associated with alterations in specific functional subsystems. Furthermore, among IL10 KO offspring from CPZ-treated dams, several functional subsystems, particularly nitrogen metabolism, diverged between mice that developed spontaneous colitis (CPZ-colitis) versus those that did not (CPZ-no-colitis) at a time point prior to eventual clinical outcome. CONCLUSIONS: Our findings provide support that functional metagenomic profiling of gut microbes has potential and promise meriting further study for development of tools to assess risk and manage human IBD.


Asunto(s)
Colitis/diagnóstico , Disbiosis/complicaciones , Microbioma Gastrointestinal/inmunología , Interleucina-10/deficiencia , Animales , Antibacterianos/administración & dosificación , Cefoperazona/administración & dosificación , Colitis/inmunología , Colitis/microbiología , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Disbiosis/inmunología , Disbiosis/microbiología , Heces/microbiología , Femenino , Microbioma Gastrointestinal/genética , Humanos , Interleucina-10/genética , Mucosa Intestinal/inmunología , Masculino , Metagenoma , Metagenómica , Ratones , Ratones Noqueados , Pronóstico
19.
Biology (Basel) ; 9(12)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255707

RESUMEN

Circadian rhythms are essential for nearly all life forms, mediated by a core molecular gene network that drives downstream molecular processes involved in immune function and metabolic regulation. These biological rhythms serve as the body's metronome in response to the 24-hour light:dark cycle and other timed stimuli. Disrupted circadian rhythms due to drastic lifestyle and environmental shifts appear to contribute to the pathogenesis of metabolic diseases, although the mechanisms remain elusive. Gut microbiota membership and function are also key mediators of metabolism and are highly sensitive to environmental perturbations. Recent evidence suggests rhythmicity of gut microbes is essential for host metabolic health. The key molecular mediators that transmit rhythmic signals between microbes and host metabolic networks remain unclear, but studies suggest the host immune system may serve as a conduit between these two systems, providing homeostatic signals to maintain overall metabolic health. Despite this knowledge, the precise mechanism and communication modalities that drive these rhythms remain unclear, especially in humans. Here, we review the current literature examining circadian dynamics of gut microbes, the immune system, and metabolism in the context of metabolic dysregulation and provide insights into gaps and challenges that remain.

20.
Cell Host Microbe ; 28(2): 157-159, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32791108

RESUMEN

Gut microbes exhibit diurnal rhythmicity, and disruptions in this rhythmicity potentially impact host health. In this issue of Cell Host & Microbe, Reitmeier et al. (2020) employ timestamped gut microbiome sequencing data from human subjects coupled with machine learning to identify microbial rhythmicity patterns that predict Type 2 Diabetes incidence.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Microbiota , Ritmo Circadiano , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...