Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 3479, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38347087

ABSTRACT

Reduced butyrate-production capacity has been reported in fecal microbial communities in patients with active ulcerative colitis. However, the butyrate-production capacity of the mucosal microbiome from active vs quiescent mucosa in ulcerative colitis has been unexplored. We sought to determine the diversity and relative abundance of mucosal bacterial and fungal communities from endoscopically active vs quiescent mucosa in patients with UC, and aimed to predict contributions of mucosal microbial communities to butyrate synthesis. Systematic, segmental right- and left-sided biopsies were obtained from endoscopically active (n = 13) or quiescent (n = 17) colonic mucosa, among 15 patients with pan-colonic ulcerative colitis. Dietary fiber intake of patients was performed using the validated five-item FiberScreen questionnaire. Amplicon sequencing of mucosal bacteria and fungi was performed. The diversity and relative abundance of mucosal bacterial and fungal taxa were quantified, and predicted contributions to butyrate synthesis were ascertained. Bacterial alpha and beta diversity were similar between active vs quiescent mucosa. Butyrogenic taxa were significantly increased in quiescence, including Butyricimonas, Subdoligranulum, and Alistipes. Predicted butyrate kinase activity was significantly and concomitantly increased in quiescent mucosa. Fiber intake was positively correlated with butyrogenic microbes. Compared to mucosal bacterial prevalence, mucosal fungi were detected in low prevalence. Butyrogenic microbes are relatively increased in quiescent mucosa in ulcerative colitis, and may be related to increased fiber intake during quiescence. Manipulation of the mucosal microbiome towards butyrate-producing bacteria may be associated with endoscopic quiescence.


Subject(s)
Colitis, Ulcerative , Humans , Colitis, Ulcerative/pathology , Butyrates , Colon/pathology , Biopsy , Intestinal Mucosa/pathology , Bacteria/genetics
2.
bioRxiv ; 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37808673

ABSTRACT

Dietary lipids play an essential role in regulating the function of the gut microbiota and gastrointestinal tract, and these luminal interactions contribute to mediating host metabolism. PAHSAs are a class of lipids with anti-diabetic and anti-inflammatory properties, but whether the gut microbiota contributes to their beneficial effects on host metabolism is unknown. Here, we report that treating high fat diet (HFD)-fed germ-free mice with PAHSAs does not improve insulin sensitivity. However, transfer of feces from PAHSA-treated, but not Vehicle-treated, chow-fed mice increases insulin-sensitivity in HFD-fed germ free mice. Thus, the gut microbiota is necessary for and can transmit the insulin-sensitizing effects of PAHSAs in HFD-fed germ-free mice. Functional analyses of the cecal metagenome and lipidome of PAHSA-treated mice identified multiple lipid species that associate with the gut commensal Bacteroides thetaiotaomicron ( Bt ) and with insulin sensitivity resulting from PAHSA treatment. Bt supplementation in HFD-fed female mice prevented weight gain, reduced adiposity, improved glucose tolerance, fortified the colonic mucus barrier and reduced systemic inflammation versus chow-fed controls, effects that were not observed in HFD-fed male mice. Furthermore, ovariectomy partially reversed the beneficial Bt effects on host metabolism, indicating a role for sex hormones in mediating probiotic effects. Altogether, these studies highlight the fact that lipids can modulate the gut microbiota resulting in improvement in host metabolism and that PAHSA-induced changes in the microbiota result in at least some of their insulin-sensitizing effects in female mice.

3.
Ann Surg ; 278(6): e1164-e1174, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37185230

ABSTRACT

OBJECTIVE: The aim was to determine preoperative gut microbiota metabolites that may be associated with postoperative delirium (POD) development in patients and further study in rodents. SUMMARY BACKGROUND DATA: POD occurs in 9% to 50% of older patients undergoing anesthesia/surgery but lacks effective treatments or prevention. High-throughput metabolomics using liquid chromatography with tandem mass spectrometry has accelerated disease-related biomarkers discovery. We performed metabolomic studies in humans to identify potential metabolite biomarkers linked to POD and examined potential mechanisms in rodents. METHODS: We performed a prospective observational cohort study to examine the metabolomic changes that were associated with the development of POD. Then the gut microbiota-related metabolomic changes were recapitulated by gut microbiota perturbation in rodents. POD was assessed in mice using a battery of behavioral tests including novel objective test, Y-maze test, open-field test, and buried food test. The mechanisms through which gut microbiota-related metabolomic changes influenced POD were examined using chemogenetics. RESULTS: Indole-3-propionic acid (IPA) is a gut microbiota metabolite that belongs to the indole family. Baseline plasma levels of IPA were significantly inversely correlated with the onset of POD in 103 (17 cases) human individuals. This relationship was validated in preclinical mouse models for POD: reducing IPA levels through gut microbiota perturbation promoted POD-like behavior. More importantly, IPA administration deterred POD-like behavior. Colonization of germ-free mice with mutant Clostridium sporogenes that did not produce IPA-promoted POD-like behavior. Chemogenetic studies revealed that the protective effect of IPA in mice was mediated, in part, by peroxisome proliferator-activated receptor gamma coactivator 1-alpha in hippocampal interneurons. CONCLUSIONS: Gut microbiota-derived IPA is an important molecule implicated in the pathogenesis of POD, which could potentially be harnessed for POD prevention.


Subject(s)
Emergence Delirium , Gastrointestinal Microbiome , Humans , Mice , Animals , Prospective Studies , Indoles/metabolism , Indoles/pharmacology , Biomarkers
4.
Cell Rep ; 40(7): 111187, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977507

ABSTRACT

Dietary protein restriction (PR) has rapid effects on metabolism including improved glucose and lipid homeostasis, via multiple mechanisms. Here, we investigate responses of fecal microbiome, hepatic transcriptome, and hepatic metabolome to six diets with protein from 18% to 0% of energy in mice. PR alters fecal microbial composition, but metabolic effects are not transferable via fecal transplantation. Hepatic transcriptome and metabolome are significantly altered in diets with lower than 10% energy from protein. Changes upon PR correlate with calorie restriction but with a larger magnitude and specific changes in amino acid (AA) metabolism. PR increases steady-state aspartate, serine, and glutamate and decreases glucose and gluconeogenic intermediates. 13C6 glucose and glycerol tracing reveal increased fractional enrichment in aspartate, serine, and glutamate. Changes remain intact in hepatic ATF4 knockout mice. Together, this demonstrates an ATF4-independent shift in gluconeogenic substrate utilization toward specific AAs, with compensation from glycerol to promote a protein-sparing response.


Subject(s)
Glucose , Glycerol , Animals , Aspartic Acid/metabolism , Dietary Proteins/metabolism , Gluconeogenesis , Glucose/metabolism , Glutamic Acid/metabolism , Glycerol/metabolism , Liver/metabolism , Mice , Serine/metabolism
5.
STAR Protoc ; 3(1): 101211, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35257115

ABSTRACT

Here, we present a protocol for the use of negative pressure isolator systems to maintain defined association and contain BSL-2 pathogens in germ-free and gnotobiotic mouse studies. We describe setup and operation of negative pressure isolators with integrated microbiologic procedures, using the BSL-2 pathogen Clostridioides difficile as a working example. This approach supports experimental systems with defined-association mice and enables high-resolution mechanistic studies of pathogen-commensal interactions and their impacts on host phenotypes. For complete details on the use and execution of this protocol, please refer to Girinathan et al. (2021).


Subject(s)
Germ-Free Life , Symbiosis , Animals , Disease Models, Animal , Mice , Microbiological Techniques
6.
Nat Commun ; 13(1): 136, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013263

ABSTRACT

Emerging research supports that triclosan (TCS), an antimicrobial agent found in thousands of consumer products, exacerbates colitis and colitis-associated colorectal tumorigenesis in animal models. While the intestinal toxicities of TCS require the presence of gut microbiota, the molecular mechanisms involved have not been defined. Here we show that intestinal commensal microbes mediate metabolic activation of TCS in the colon and drive its gut toxicology. Using a range of in vitro, ex vivo, and in vivo approaches, we identify specific microbial ß-glucuronidase (GUS) enzymes involved and pinpoint molecular motifs required to metabolically activate TCS in the gut. Finally, we show that targeted inhibition of bacterial GUS enzymes abolishes the colitis-promoting effects of TCS, supporting an essential role of specific microbial proteins in TCS toxicity. Together, our results define a mechanism by which intestinal microbes contribute to the metabolic activation and gut toxicity of TCS, and highlight the importance of considering the contributions of the gut microbiota in evaluating the toxic potential of environmental chemicals.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Carcinogens/antagonists & inhibitors , Colitis/prevention & control , Colorectal Neoplasms/prevention & control , Glucuronidase/antagonists & inhibitors , Glycoside Hydrolase Inhibitors/pharmacology , Triclosan/antagonists & inhibitors , Animals , Anti-Infective Agents, Local/chemistry , Anti-Infective Agents, Local/metabolism , Anti-Infective Agents, Local/toxicity , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biotransformation , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogens/chemistry , Carcinogens/metabolism , Carcinogens/toxicity , Colitis/chemically induced , Colitis/enzymology , Colitis/microbiology , Colon/drug effects , Colon/microbiology , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/drug effects , Gene Expression , Glucuronidase/chemistry , Glucuronidase/genetics , Glucuronidase/metabolism , Glycoside Hydrolase Inhibitors/chemistry , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Triclosan/chemistry , Triclosan/metabolism , Triclosan/toxicity
7.
Microb Cell ; 8(6): 131-142, 2021 Apr 29.
Article in English | MEDLINE | ID: mdl-34055966

ABSTRACT

Cranberry consumption has numerous health benefits, with experimental reports showing its anti-inflammatory and anti-tumor properties. Importantly, microbiome research has demonstrated that the gastrointestinal bacterial community modulates host immunity, raising the question of whether the cranberry-derived effect may be related to its ability to modulate the microbiome. Only a few studies have investigated the effect of cranberry products on the microbiome to date. Especially because cranberries are rich in dietary fibers, the extent of microbiome modulation by polyphenols, particularly proanthocyanidins (PACs), remains to be shown. Since previous work has only focused on long-term effects of cranberry extracts, in this study we investigated the effect of a water-soluble, PAC-rich cranberry juice extract (CJE) on the short-term dynamics of a human-derived bacterial community in a gnotobiotic mouse model. CJE characterization revealed a high enrichment in PACs (57%), the highest ever utilized in a microbiome study. In a 37-day experiment with a ten-day CJE intervention and 14-day recovery phase, we profiled the microbiota via 16S rRNA sequencing and applied diverse time-series analytics methods to identify individual bacterial responses. We show that daily administration of CJE induces distinct dynamic patterns in bacterial abundances during and after treatment, before recovering resiliently to pre-treatment levels. Specifically, we observed an increase of Akkermansia muciniphila and Clostridium hiranonis at the expense of Bacteroides ovatus after the offset of the selection pressure imposed by the PAC-rich CJE. This demonstrates that termination of an intervention with a cranberry product can induce changes of a magnitude as high as the intervention itself.

8.
Sci Total Environ ; 787: 147677, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34004538

ABSTRACT

Metabolic transformations play critical roles in the bioavailability and toxicities of environmental pollutants and toxicants. However, most previous research has focused on the metabolic reactions in host tissues, the gut microbiota-mediated biotransformation of environmental compounds is understudied. Using triclocarban (TCC) as a model environmental compound, here we study the metabolic fate of TCC in gut tissues and determine the roles of gut microbiota involved. We find that compared with other tissues, the colon tissue has a unique metabolic profile of TCC, with high abundance of the parent compound TCC and its free-form metabolites. Using a variety of approaches including antibiotic-mediated suppression of gut bacteria in vivo, germ-free mice, and in vitro culture of fecal bacteria, we found that the unique metabolic profile of TCC in the colon is mediated by the actions of gut microbiota. Overall, our findings support that gut microbiota plays important roles in colonic metabolism of TCC, highlighting the importance to consider the contributions of gut microbiota in toxicology evaluation of environmental compounds.


Subject(s)
Carbanilides , Gastrointestinal Microbiome , Animals , Carbanilides/toxicity , Colon , Feces , Mice
9.
Physiol Rep ; 8(19): e14584, 2020 10.
Article in English | MEDLINE | ID: mdl-33052618

ABSTRACT

In mice, both androgens and the gut microbiota modify pulmonary responses to ozone. We hypothesized that androgens affect gut microbiota and thereby impact pulmonary responses to ozone. To address this hypothesis, we transferred cecal microbiota from male castrated or sham castrated C57BL/6J mice into female germ-free recipient C57BL/6J mice. Four weeks later mice were exposed to ozone (2 ppm) or room air for 3 hr. The gut microbiomes of castrated versus sham castrated donors differed, as did those of recipients of microbiota from castrated versus sham castrated donors. In recipients, ozone-induced airway hyperresponsiveness was not affected by donor castration status. However, compared to mice receiving microbiota from sham castrated donors, mice receiving microbiota from castrated donors had elevated numbers of bronchoalveolar lavage (BAL) neutrophils despite evidence of reduced lung injury as measured by BAL protein. Serum concentrations of IL-17A and G-CSF were significantly greater in recipients of castrated versus sham castrated microbiota. Furthermore, BAL neutrophils correlated with both serum IL-17A and serum G-CSF. Our data indicate that androgen-mediated effects on the gut microbiota modulate pulmonary inflammatory responses to ozone and suggest a role for circulating IL-17A and G-CSF in these events.


Subject(s)
Androgens/pharmacology , Bronchoalveolar Lavage Fluid/microbiology , Gastrointestinal Microbiome/drug effects , Ozone/adverse effects , Animals , Interleukin-17/metabolism , Lung/drug effects , Lung/metabolism , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Respiratory Hypersensitivity/physiopathology
10.
Proc Natl Acad Sci U S A ; 117(5): 2570-2578, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31964813

ABSTRACT

The thymus generates cells of the T cell lineage that seed the lymphatic and blood systems. Transcription factor regulatory networks control the lineage programming and maturation of thymic precursor cells. Whether extrathymic antigenic events, such as the microbial colonization of the mucosal tract also shape the thymic T cell repertoire is unclear. We show here that intestinal microbes influence the thymic homeostasis of PLZF-expressing cells in early life. Impaired thymic development of PLZF+ innate lymphocytes in germ-free (GF) neonatal mice is restored by colonization with a human commensal, Bacteroides fragilis, but not with a polysaccharide A (PSA) deficient isogenic strain. Plasmacytoid dendritic cells influenced by microbes migrate from the colon to the thymus in early life to regulate PLZF+ cell homeostasis. Importantly, perturbations in thymic PLZF+ cells brought about by alterations in early gut microbiota persist into adulthood and are associated with increased susceptibility to experimental colitis. Our studies identify a pathway of communication between intestinal microbes and thymic lymphocytes in the neonatal period that can modulate host susceptibility to immune-mediated diseases later in life.


Subject(s)
Gastrointestinal Microbiome , Lymphocytes/immunology , Thymus Gland/growth & development , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteroides fragilis/physiology , Cell Differentiation , Colitis/genetics , Colitis/immunology , Colitis/microbiology , Colon/microbiology , Humans , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein/genetics , Promyelocytic Leukemia Zinc Finger Protein/immunology , Thymus Gland/cytology , Thymus Gland/immunology
11.
Cell Host Microbe ; 25(6): 803-814.e5, 2019 Jun 12.
Article in English | MEDLINE | ID: mdl-31175044

ABSTRACT

The human gut microbiome is comprised of densely colonizing microorganisms including bacteriophages, which are in dynamic interaction with each other and the mammalian host. To address how bacteriophages impact bacterial communities in the gut, we investigated the dynamic effects of phages on a model microbiome. Gnotobiotic mice were colonized with defined human gut commensal bacteria and subjected to predation by cognate lytic phages. We found that phage predation not only directly impacts susceptible bacteria but also leads to cascading effects on other bacterial species via interbacterial interactions. Metabolomic profiling revealed that shifts in the microbiome caused by phage predation have a direct consequence on the gut metabolome. Our work provides insight into the ecological importance of phages as modulators of bacterial colonization, and it additionally suggests the potential impact of gut phages on the mammalian host with implications for their therapeutic use to precisely modulate the microbiome.


Subject(s)
Bacteriolysis , Bacteriophages/growth & development , Feces/chemistry , Gastrointestinal Microbiome , Metabolome , Animals , Germ-Free Life , Mice , Microbial Interactions
12.
Am J Respir Cell Mol Biol ; 61(6): 702-712, 2019 12.
Article in English | MEDLINE | ID: mdl-31144984

ABSTRACT

Obesity is a risk factor for asthma, especially nonatopic asthma, and attenuates the efficacy of standard asthma therapeutics. Obesity also augments pulmonary responses to ozone, a nonatopic asthma trigger. The purpose of this study was to determine whether obesity-related alterations in gut microbiota contribute to these augmented responses to ozone. Ozone-induced increases in airway responsiveness, a canonical feature of asthma, were greater in obese db/db mice than in lean wild-type control mice. Depletion of gut microbiota with a cocktail of antibiotics attenuated obesity-related increases in the response to ozone, indicating a role for microbiota. Moreover, ozone-induced airway hyperresponsiveness was greater in germ-free mice that had been reconstituted with colonic contents of db/db than in wild-type mice. In addition, compared with dietary supplementation with the nonfermentable fiber cellulose, dietary supplementation with the fermentable fiber pectin attenuated obesity-related increases in the pulmonary response to ozone, likely by reducing ozone-induced release of IL-17A. Our data indicate a role for microbiota in obesity-related increases in the response to an asthma trigger and suggest that microbiome-based therapies such as prebiotics may provide an alternative therapeutic strategy for obese patients with asthma.


Subject(s)
Gastrointestinal Microbiome/physiology , Obesity/complications , Ozone/toxicity , Respiratory Hypersensitivity/etiology , Airway Resistance , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Asthma/etiology , Asthma/therapy , Cellulose/administration & dosage , Dietary Fiber/administration & dosage , Fecal Microbiota Transplantation , Female , Fermentation , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Obesity/microbiology , Obesity/physiopathology , Pectins/administration & dosage , Pectins/therapeutic use , Receptors, Leptin/deficiency , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/diet therapy , Respiratory Hypersensitivity/microbiology
13.
Curr Protoc Immunol ; 121(1): e48, 2018 04.
Article in English | MEDLINE | ID: mdl-30008984

ABSTRACT

Animal models are essential to dissect host-microbiota interactions that impact health and the development of disease. In addition to providing pre-clinical models for the development of novel therapeutics and diagnostic biomarkers, mouse systems actively support microbiome studies by defining microbial contributions to normal development and homeostasis, and as well as their role in promoting diseases such as inflammatory auto-immune diseases, diabetes, metabolic syndromes, and susceptibilities to infectious agents. Mice provide a genetically tenable host that can be reared under gnotobiotic (germfree) conditions, allowing colonization studies with human or mouse-origin defined or complex microbial communities to define specific in vivo effects. The protocols and background information detail key aspects to consider in designing host-microbiome experiments with mouse models, and to develop robust systems that leverage gnotobiotic mice, microbial consortia, and specific environmental perturbations to identify causal effects in vivo.


Subject(s)
Germ-Free Life , Microbiological Techniques , Animals , Feces/microbiology , Humans , Mice , Microbiota , Sterilization
14.
Genome Biol ; 17(1): 121, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27259475

ABSTRACT

Predicting dynamics of host-microbial ecosystems is crucial for the rational design of bacteriotherapies. We present MDSINE, a suite of algorithms for inferring dynamical systems models from microbiome time-series data and predicting temporal behaviors. Using simulated data, we demonstrate that MDSINE significantly outperforms the existing inference method. We then show MDSINE's utility on two new gnotobiotic mice datasets, investigating infection with Clostridium difficile and an immune-modulatory probiotic. Using these datasets, we demonstrate new capabilities, including accurate forecasting of microbial dynamics, prediction of stable sub-communities that inhibit pathogen growth, and identification of bacteria most crucial to community integrity in response to perturbations.


Subject(s)
Clostridioides difficile/genetics , Host-Pathogen Interactions/genetics , Microbiota/genetics , Models, Theoretical , Algorithms , Animals , Clostridioides difficile/growth & development , Clostridioides difficile/pathogenicity , Mice
15.
PLoS One ; 9(7): e95534, 2014.
Article in English | MEDLINE | ID: mdl-25014551

ABSTRACT

Longitudinal studies of the microbiota are important for discovering changes in microbial communities that affect the host. The complexity of these ecosystems requires rigorous integrated experimental and computational methods to identify temporal signatures that promote physiologic or pathophysiologic responses in vivo. Employing a murine model of infectious colitis with the pathogen Citrobacter rodentium, we generated a 2-month time-series of 16S rDNA gene profiles, and quantitatively cultured commensals, from multiple intestinal sites in infected and uninfected mice. We developed a computational framework to discover time-varying signatures for individual taxa, and to automatically group signatures to identify microbial sub-communities within the larger gut ecosystem that demonstrate common behaviors. Application of this model to the 16S rDNA dataset revealed dynamic alterations in the microbiota at multiple levels of resolution, from effects on systems-level metrics to changes across anatomic sites for individual taxa and species. These analyses revealed unique, time-dependent microbial signatures associated with host responses at different stages of colitis. Signatures included a Mucispirillum OTU associated with early disruption of the colonic surface mucus layer, prior to the onset of symptomatic colitis, and members of the Clostridiales and Lactobacillales that increased with successful resolution of inflammation, after clearance of the pathogen. Quantitative culture data validated findings for predominant species, further refining and strengthening model predictions. These findings provide new insights into the complex behaviors found within host ecosystems, and define several time-dependent microbial signatures that may be leveraged in studies of other infectious or inflammatory conditions.


Subject(s)
Citrobacter rodentium/genetics , Colitis/microbiology , Enterobacteriaceae Infections/microbiology , Genes, Bacterial , Microbiota/genetics , RNA, Ribosomal, 16S/genetics , Animals , Bacterial Load , Citrobacter rodentium/growth & development , Enterobacter/genetics , Enterobacter/isolation & purification , Host-Pathogen Interactions , Intestinal Mucosa/microbiology , Intestines/microbiology , Lactobacillus/genetics , Lactobacillus/isolation & purification , Metagenome , Mice , Molecular Sequence Annotation , Phylogeny , Proteus vulgaris/genetics , Proteus vulgaris/isolation & purification , RNA, Ribosomal, 16S/classification
SELECTION OF CITATIONS
SEARCH DETAIL
...