Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cell ; 185(18): 3307-3328.e19, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35987213

ABSTRACT

Non-nutritive sweeteners (NNS) are commonly integrated into human diet and presumed to be inert; however, animal studies suggest that they may impact the microbiome and downstream glycemic responses. We causally assessed NNS impacts in humans and their microbiomes in a randomized-controlled trial encompassing 120 healthy adults, administered saccharin, sucralose, aspartame, and stevia sachets for 2 weeks in doses lower than the acceptable daily intake, compared with controls receiving sachet-contained vehicle glucose or no supplement. As groups, each administered NNS distinctly altered stool and oral microbiome and plasma metabolome, whereas saccharin and sucralose significantly impaired glycemic responses. Importantly, gnotobiotic mice conventionalized with microbiomes from multiple top and bottom responders of each of the four NNS-supplemented groups featured glycemic responses largely reflecting those noted in respective human donors, which were preempted by distinct microbial signals, as exemplified by sucralose. Collectively, human NNS consumption may induce person-specific, microbiome-dependent glycemic alterations, necessitating future assessment of clinical implications.


Subject(s)
Microbiota , Non-Nutritive Sweeteners , Adult , Animals , Aspartame/pharmacology , Blood Glucose , Humans , Mice , Non-Nutritive Sweeteners/analysis , Non-Nutritive Sweeteners/pharmacology , Saccharin/pharmacology
2.
Cell ; 185(16): 2879-2898.e24, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35931020

ABSTRACT

Human gut commensals are increasingly suggested to impact non-communicable diseases, such as inflammatory bowel diseases (IBD), yet their targeted suppression remains a daunting unmet challenge. In four geographically distinct IBD cohorts (n = 537), we identify a clade of Klebsiella pneumoniae (Kp) strains, featuring a unique antibiotics resistance and mobilome signature, to be strongly associated with disease exacerbation and severity. Transfer of clinical IBD-associated Kp strains into colitis-prone, germ-free, and colonized mice enhances intestinal inflammation. Stepwise generation of a lytic five-phage combination, targeting sensitive and resistant IBD-associated Kp clade members through distinct mechanisms, enables effective Kp suppression in colitis-prone mice, driving an attenuated inflammation and disease severity. Proof-of-concept assessment of Kp-targeting phages in an artificial human gut and in healthy volunteers demonstrates gastric acid-dependent phage resilience, safety, and viability in the lower gut. Collectively, we demonstrate the feasibility of orally administered combination phage therapy in avoiding resistance, while effectively inhibiting non-communicable disease-contributing pathobionts.


Subject(s)
Bacteriophages , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Animals , Colitis/therapy , Humans , Inflammation/therapy , Inflammatory Bowel Diseases/therapy , Klebsiella pneumoniae , Mice
3.
Nat Immunol ; 24(4): 585-594, 2023 04.
Article in English | MEDLINE | ID: mdl-36941399

ABSTRACT

Unlike other nucleotide oligomerization domain-like receptors, Nlrp10 lacks a canonical leucine-rich repeat domain, suggesting that it is incapable of signal sensing and inflammasome formation. Here we show that mouse Nlrp10 is expressed in distal colonic intestinal epithelial cells (IECs) and modulated by the intestinal microbiome. In vitro, Nlrp10 forms an Apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC)-dependent, m-3M3FBS-activated, polyinosinic:polycytidylic acid-modulated inflammasome driving interleukin-1ß and interleukin-18 secretion. In vivo, Nlrp10 signaling is dispensable during steady state but becomes functional during autoinflammation in antagonizing mucosal damage. Importantly, whole-body or conditional IEC Nlrp10 depletion leads to reduced IEC caspase-1 activation, coupled with enhanced susceptibility to dextran sodium sulfate-induced colitis, mediated by altered inflammatory and healing programs. Collectively, understanding Nlrp10 inflammasome-dependent and independent activity, regulation and possible human relevance might facilitate the development of new innate immune anti-inflammatory interventions.


Subject(s)
Apoptosis Regulatory Proteins , Inflammasomes , Mice , Humans , Animals , Inflammasomes/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Apoptosis , Caspase 1/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-1beta/metabolism , Adaptor Proteins, Signal Transducing/metabolism
4.
Cell ; 182(6): 1441-1459.e21, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32888430

ABSTRACT

Throughout a 24-h period, the small intestine (SI) is exposed to diurnally varying food- and microbiome-derived antigenic burdens but maintains a strict immune homeostasis, which when perturbed in genetically susceptible individuals, may lead to Crohn disease. Herein, we demonstrate that dietary content and rhythmicity regulate the diurnally shifting SI epithelial cell (SIEC) transcriptional landscape through modulation of the SI microbiome. We exemplify this concept with SIEC major histocompatibility complex (MHC) class II, which is diurnally modulated by distinct mucosal-adherent SI commensals, while supporting downstream diurnal activity of intra-epithelial IL-10+ lymphocytes regulating the SI barrier function. Disruption of this diurnally regulated diet-microbiome-MHC class II-IL-10-epithelial barrier axis by circadian clock disarrangement, alterations in feeding time or content, or epithelial-specific MHC class II depletion leads to an extensive microbial product influx, driving Crohn-like enteritis. Collectively, we highlight nutritional features that modulate SI microbiome, immunity, and barrier function and identify dietary, epithelial, and immune checkpoints along this axis to be potentially exploitable in future Crohn disease interventions.


Subject(s)
Crohn Disease/microbiology , Epithelial Cells/metabolism , Gastrointestinal Microbiome , Histocompatibility Antigens Class II/metabolism , Intestine, Small/immunology , Intestine, Small/microbiology , Transcriptome/genetics , Animals , Anti-Bacterial Agents/pharmacology , Circadian Clocks/physiology , Crohn Disease/immunology , Crohn Disease/metabolism , Diet , Epithelial Cells/cytology , Epithelial Cells/immunology , Flow Cytometry , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Gene Expression Profiling , Histocompatibility Antigens Class II/genetics , Homeostasis , In Situ Hybridization, Fluorescence , Interleukin-10/metabolism , Interleukin-10/pharmacology , Intestine, Small/physiology , Lymphocytes , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Periodicity , T-Lymphocytes/immunology , Transcriptome/physiology
5.
Cell ; 178(3): 686-698.e14, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31257031

ABSTRACT

Immune cells residing in white adipose tissue have been highlighted as important factors contributing to the pathogenesis of metabolic diseases, but the molecular regulators that drive adipose tissue immune cell remodeling during obesity remain largely unknown. Using index and transcriptional single-cell sorting, we comprehensively map all adipose tissue immune populations in both mice and humans during obesity. We describe a novel and conserved Trem2+ lipid-associated macrophage (LAM) subset and identify markers, spatial localization, origin, and functional pathways associated with these cells. Genetic ablation of Trem2 in mice globally inhibits the downstream molecular LAM program, leading to adipocyte hypertrophy as well as systemic hypercholesterolemia, body fat accumulation, and glucose intolerance. These findings identify Trem2 signaling as a major pathway by which macrophages respond to loss of tissue-level lipid homeostasis, highlighting Trem2 as a key sensor of metabolic pathologies across multiple tissues and a potential therapeutic target in metabolic diseases.


Subject(s)
Macrophages/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Diet, High-Fat , Glucose Intolerance , Humans , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Lipid Metabolism/genetics , Lipids/analysis , Macrophages/cytology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/metabolism , Obesity/metabolism , Obesity/pathology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Signal Transduction , Single-Cell Analysis
6.
Cell ; 174(6): 1388-1405.e21, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193112

ABSTRACT

Empiric probiotics are commonly consumed by healthy individuals as means of life quality improvement and disease prevention. However, evidence of probiotic gut mucosal colonization efficacy remains sparse and controversial. We metagenomically characterized the murine and human mucosal-associated gastrointestinal microbiome and found it to only partially correlate with stool microbiome. A sequential invasive multi-omics measurement at baseline and during consumption of an 11-strain probiotic combination or placebo demonstrated that probiotics remain viable upon gastrointestinal passage. In colonized, but not germ-free mice, probiotics encountered a marked mucosal colonization resistance. In contrast, humans featured person-, region- and strain-specific mucosal colonization patterns, hallmarked by predictive baseline host and microbiome features, but indistinguishable by probiotics presence in stool. Consequently, probiotics induced a transient, individualized impact on mucosal community structure and gut transcriptome. Collectively, empiric probiotics supplementation may be limited in universally and persistently impacting the gut mucosa, meriting development of new personalized probiotic approaches.


Subject(s)
Gastrointestinal Microbiome , Probiotics/administration & dosage , Adolescent , Adult , Aged , Animals , Bacteria/genetics , Bacteria/isolation & purification , Feces/microbiology , Female , Gastric Mucosa/microbiology , Humans , Intestinal Mucosa/microbiology , Male , Metagenomics , Mice , Mice, Inbred C57BL , Middle Aged , Placebo Effect , Principal Component Analysis , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Transcriptome , Young Adult
7.
Cell ; 174(6): 1406-1423.e16, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193113

ABSTRACT

Probiotics are widely prescribed for prevention of antibiotics-associated dysbiosis and related adverse effects. However, probiotic impact on post-antibiotic reconstitution of the gut mucosal host-microbiome niche remains elusive. We invasively examined the effects of multi-strain probiotics or autologous fecal microbiome transplantation (aFMT) on post-antibiotic reconstitution of the murine and human mucosal microbiome niche. Contrary to homeostasis, antibiotic perturbation enhanced probiotics colonization in the human mucosa but only mildly improved colonization in mice. Compared to spontaneous post-antibiotic recovery, probiotics induced a markedly delayed and persistently incomplete indigenous stool/mucosal microbiome reconstitution and host transcriptome recovery toward homeostatic configuration, while aFMT induced a rapid and near-complete recovery within days of administration. In vitro, Lactobacillus-secreted soluble factors contributed to probiotics-induced microbiome inhibition. Collectively, potential post-antibiotic probiotic benefits may be offset by a compromised gut mucosal recovery, highlighting a need of developing aFMT or personalized probiotic approaches achieving mucosal protection without compromising microbiome recolonization in the antibiotics-perturbed host.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Probiotics/administration & dosage , Adolescent , Adult , Aged , Animals , Fecal Microbiota Transplantation , Feces/microbiology , Female , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Lactobacillus/drug effects , Lactobacillus/genetics , Lactobacillus/isolation & purification , Lactococcus/genetics , Lactococcus/isolation & purification , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA, Ribosomal, 16S/analysis , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Young Adult
8.
Cell ; 166(5): 1231-1246.e13, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27545347

ABSTRACT

Innate lymphoid cells (ILCs) are critical modulators of mucosal immunity, inflammation, and tissue homeostasis, but their full spectrum of cellular states and regulatory landscapes remains elusive. Here, we combine genome-wide RNA-seq, ChIP-seq, and ATAC-seq to compare the transcriptional and epigenetic identity of small intestinal ILCs, identifying thousands of distinct gene profiles and regulatory elements. Single-cell RNA-seq and flow and mass cytometry analyses reveal compartmentalization of cytokine expression and metabolic activity within the three classical ILC subtypes and highlight transcriptional states beyond the current canonical classification. In addition, using antibiotic intervention and germ-free mice, we characterize the effect of the microbiome on the ILC regulatory landscape and determine the response of ILCs to microbial colonization at the single-cell level. Together, our work characterizes the spectrum of transcriptional identities of small intestinal ILCs and describes how ILCs differentially integrate signals from the microbial microenvironment to generate phenotypic and functional plasticity.


Subject(s)
Gastrointestinal Microbiome , Immunity, Innate/genetics , Intestines/immunology , Intestines/microbiology , Lymphocytes/immunology , Lymphocytes/microbiology , Animals , Base Sequence , Chromatin/metabolism , Cytokines/immunology , Epigenesis, Genetic , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Single-Cell Analysis , Transcription, Genetic
9.
Cell ; 167(6): 1495-1510.e12, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912059

ABSTRACT

The intestinal microbiota undergoes diurnal compositional and functional oscillations that affect metabolic homeostasis, but the mechanisms by which the rhythmic microbiota influences host circadian activity remain elusive. Using integrated multi-omics and imaging approaches, we demonstrate that the gut microbiota features oscillating biogeographical localization and metabolome patterns that determine the rhythmic exposure of the intestinal epithelium to different bacterial species and their metabolites over the course of a day. This diurnal microbial behavior drives, in turn, the global programming of the host circadian transcriptional, epigenetic, and metabolite oscillations. Surprisingly, disruption of homeostatic microbiome rhythmicity not only abrogates normal chromatin and transcriptional oscillations of the host, but also incites genome-wide de novo oscillations in both intestine and liver, thereby impacting diurnal fluctuations of host physiology and disease susceptibility. As such, the rhythmic biogeography and metabolome of the intestinal microbiota regulates the temporal organization and functional outcome of host transcriptional and epigenetic programs.


Subject(s)
Circadian Rhythm , Colon/microbiology , Gastrointestinal Microbiome , Transcriptome , Animals , Chromatin/metabolism , Colon/metabolism , Germ-Free Life , Liver/metabolism , Mice , Microscopy, Electron, Scanning
10.
Cell ; 163(6): 1428-43, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26638072

ABSTRACT

Host-microbiome co-evolution drives homeostasis and disease susceptibility, yet regulatory principles governing the integrated intestinal host-commensal microenvironment remain obscure. While inflammasome signaling participates in these interactions, its activators and microbiome-modulating mechanisms are unknown. Here, we demonstrate that the microbiota-associated metabolites taurine, histamine, and spermine shape the host-microbiome interface by co-modulating NLRP6 inflammasome signaling, epithelial IL-18 secretion, and downstream anti-microbial peptide (AMP) profiles. Distortion of this balanced AMP landscape by inflammasome deficiency drives dysbiosis development. Upon fecal transfer, colitis-inducing microbiota hijacks this microenvironment-orchestrating machinery through metabolite-mediated inflammasome suppression, leading to distorted AMP balance favoring its preferential colonization. Restoration of the metabolite-inflammasome-AMP axis reinstates a normal microbiota and ameliorates colitis. Together, we identify microbial modulators of the NLRP6 inflammasome and highlight mechanisms by which microbiome-host interactions cooperatively drive microbial community stability through metabolite-mediated innate immune modulation. Therefore, targeted "postbiotic" metabolomic intervention may restore a normal microenvironment as treatment or prevention of dysbiosis-driven diseases.


Subject(s)
Colon/immunology , Colon/microbiology , Inflammasomes/immunology , Microbiota , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Antimicrobial Cationic Peptides , Colitis/chemically induced , Colitis/drug therapy , Colon/metabolism , Dysbiosis/metabolism , Germ-Free Life , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Interleukin-18/immunology , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/genetics , Taurine/administration & dosage
11.
Cell ; 159(3): 514-29, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417104

ABSTRACT

All domains of life feature diverse molecular clock machineries that synchronize physiological processes to diurnal environmental fluctuations. However, no mechanisms are known to cross-regulate prokaryotic and eukaryotic circadian rhythms in multikingdom ecosystems. Here, we show that the intestinal microbiota, in both mice and humans, exhibits diurnal oscillations that are influenced by feeding rhythms, leading to time-specific compositional and functional profiles over the course of a day. Ablation of host molecular clock components or induction of jet lag leads to aberrant microbiota diurnal fluctuations and dysbiosis, driven by impaired feeding rhythmicity. Consequently, jet-lag-induced dysbiosis in both mice and humans promotes glucose intolerance and obesity that are transferrable to germ-free mice upon fecal transplantation. Together, these findings provide evidence of coordinated metaorganism diurnal rhythmicity and offer a microbiome-dependent mechanism for common metabolic disturbances in humans with aberrant circadian rhythms, such as those documented in shift workers and frequent flyers.


Subject(s)
Circadian Clocks , Circadian Rhythm , Glucose Intolerance , Microbiota , Animals , Dysbiosis/microbiology , Dysbiosis/physiopathology , Feeding Behavior , Homeostasis , Humans , Jet Lag Syndrome/physiopathology , Metabolic Diseases/microbiology , Metabolic Diseases/physiopathology , Mice , Obesity/metabolism , Sleep
12.
Nature ; 624(7992): 645-652, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38093014

ABSTRACT

People with diabetes feature a life-risking susceptibility to respiratory viral infection, including influenza and SARS-CoV-2 (ref. 1), whose mechanism remains unknown. In acquired and genetic mouse models of diabetes, induced with an acute pulmonary viral infection, we demonstrate that hyperglycaemia leads to impaired costimulatory molecule expression, antigen transport and T cell priming in distinct lung dendritic cell (DC) subsets, driving a defective antiviral adaptive immune response, delayed viral clearance and enhanced mortality. Mechanistically, hyperglycaemia induces an altered metabolic DC circuitry characterized by increased glucose-to-acetyl-CoA shunting and downstream histone acetylation, leading to global chromatin alterations. These, in turn, drive impaired expression of key DC effectors including central antigen presentation-related genes. Either glucose-lowering treatment or pharmacological modulation of histone acetylation rescues DC function and antiviral immunity. Collectively, we highlight a hyperglycaemia-driven metabolic-immune axis orchestrating DC dysfunction during pulmonary viral infection and identify metabolic checkpoints that may be therapeutically exploited in mitigating exacerbated disease in infected diabetics.


Subject(s)
Dendritic Cells , Diabetes Complications , Diabetes Mellitus , Disease Susceptibility , Hyperglycemia , Lung , Virus Diseases , Animals , Mice , Acetyl Coenzyme A/metabolism , Acetylation , Chromatin/genetics , Chromatin/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Diabetes Complications/immunology , Diabetes Complications/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Glucose/metabolism , Histones/metabolism , Hyperglycemia/complications , Hyperglycemia/immunology , Hyperglycemia/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , T-Lymphocytes/immunology , Virus Diseases/complications , Virus Diseases/immunology , Virus Diseases/mortality , Viruses/immunology , Disease Models, Animal , Humans
13.
Nature ; 600(7890): 713-719, 2021 12.
Article in English | MEDLINE | ID: mdl-34880502

ABSTRACT

Cigarette smoking constitutes a leading global cause of morbidity and preventable death1, and most active smokers report a desire or recent attempt to quit2. Smoking-cessation-induced weight gain (SCWG; 4.5 kg reported to be gained on average per 6-12 months, >10 kg year-1 in 13% of those who stopped smoking3) constitutes a major obstacle to smoking abstinence4, even under stable5,6 or restricted7 caloric intake. Here we use a mouse model to demonstrate that smoking and cessation induce a dysbiotic state that is driven by an intestinal influx of cigarette-smoke-related metabolites. Microbiome depletion induced by treatment with antibiotics prevents SCWG. Conversely, fecal microbiome transplantation from mice previously exposed to cigarette smoke into germ-free mice naive to smoke exposure induces excessive weight gain across diets and mouse strains. Metabolically, microbiome-induced SCWG involves a concerted host and microbiome shunting of dietary choline to dimethylglycine driving increased gut energy harvest, coupled with the depletion of a cross-regulated weight-lowering metabolite, N-acetylglycine, and possibly by the effects of other differentially abundant cigarette-smoke-related metabolites. Dimethylglycine and N-acetylglycine may also modulate weight and associated adipose-tissue immunity under non-smoking conditions. Preliminary observations in a small cross-sectional human cohort support these findings, which calls for larger human trials to establish the relevance of this mechanism in active smokers. Collectively, we uncover a microbiome-dependent orchestration of SCWG that may be exploitable to improve smoking-cessation success and to correct metabolic perturbations even in non-smoking settings.


Subject(s)
Gastrointestinal Microbiome , Smoking Cessation , Weight Gain , Animals , Cross-Sectional Studies , Dysbiosis/etiology , Dysbiosis/metabolism , Dysbiosis/pathology , Mice , Models, Animal , Smoking/metabolism , Smoking/pathology
14.
Nature ; 572(7770): 474-480, 2019 08.
Article in English | MEDLINE | ID: mdl-31330533

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disorder, in which the clinical manifestations may be influenced by genetic and unknown environmental factors. Here we show that ALS-prone Sod1 transgenic (Sod1-Tg) mice have a pre-symptomatic, vivarium-dependent dysbiosis and altered metabolite configuration, coupled with an exacerbated disease under germ-free conditions or after treatment with broad-spectrum antibiotics. We correlate eleven distinct commensal bacteria at our vivarium with the severity of ALS in mice, and by their individual supplementation into antibiotic-treated Sod1-Tg mice we demonstrate that Akkermansia muciniphila (AM) ameliorates whereas Ruminococcus torques and Parabacteroides distasonis exacerbate the symptoms of ALS. Furthermore, Sod1-Tg mice that are administered AM are found to accumulate AM-associated nicotinamide in the central nervous system, and systemic supplementation of nicotinamide improves motor symptoms and gene expression patterns in the spinal cord of Sod1-Tg mice. In humans, we identify distinct microbiome and metabolite configurations-including reduced levels of nicotinamide systemically and in the cerebrospinal fluid-in a small preliminary study that compares patients with ALS with household controls. We suggest that environmentally driven microbiome-brain interactions may modulate ALS in mice, and we call for similar investigations in the human form of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/microbiology , Amyotrophic Lateral Sclerosis/physiopathology , Gastrointestinal Microbiome/physiology , Niacinamide/metabolism , Akkermansia , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Dysbiosis , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Humans , Longevity , Male , Mice , Mice, Transgenic , Niacinamide/biosynthesis , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Survival Rate , Symbiosis/drug effects , Verrucomicrobia/metabolism , Verrucomicrobia/physiology
16.
Clin Sci (Lond) ; 136(18): 1371-1387, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36156126

ABSTRACT

Smoking is associated with an increased risk of cancer, pulmonary and cardiovascular diseases, but the precise mechanisms by which such risk is mediated remain poorly understood. Additionally, smoking can impact the oral, nasal, oropharyngeal, lung and gut microbiome composition, function, and secreted molecule repertoire. Microbiome changes induced by smoking can bear direct consequences on smoking-related illnesses. Moreover, smoking-associated dysbiosis may modulate weight gain development following smoking cessation. Here, we review the implications of cigarette smoking on microbiome community structure and function. In addition, we highlight the potential impacts of microbial dysbiosis on smoking-related diseases. We discuss challenges in studying host-microbiome interactions in the context of smoking, such as the correlations with smoking-related disease severity versus causation and mechanism. In all, understanding the microbiome's role in the pathophysiology of smoking-related diseases may promote the development of rational therapies for smoking- and smoking cessation-related disorders, as well as assist in smoking abstinence.


Subject(s)
Cardiovascular Diseases , Gastrointestinal Microbiome , Microbiota , Cardiovascular Diseases/complications , Dysbiosis/complications , Humans , Smoking/adverse effects
17.
Nature ; 540(7634): 544-551, 2016 Dec 22.
Article in English | MEDLINE | ID: mdl-27906159

ABSTRACT

In tackling the obesity pandemic, considerable efforts are devoted to the development of effective weight reduction strategies, yet many dieting individuals fail to maintain a long-term weight reduction, and instead undergo excessive weight regain cycles. The mechanisms driving recurrent post-dieting obesity remain largely elusive. Here we identify an intestinal microbiome signature that persists after successful dieting of obese mice and contributes to faster weight regain and metabolic aberrations upon re-exposure to obesity-promoting conditions. Faecal transfer experiments show that the accelerated weight regain phenotype can be transmitted to germ-free mice. We develop a machine-learning algorithm that enables personalized microbiome-based prediction of the extent of post-dieting weight regain. Additionally, we find that the microbiome contributes to diminished post-dieting flavonoid levels and reduced energy expenditure, and demonstrate that flavonoid-based 'post-biotic' intervention ameliorates excessive secondary weight gain. Together, our data highlight a possible microbiome contribution to accelerated post-dieting weight regain, and suggest that microbiome-targeting approaches may help to diagnose and treat this common disorder.

18.
Gastroenterology ; 159(5): 1807-1823, 2020 11.
Article in English | MEDLINE | ID: mdl-32653496

ABSTRACT

BACKGROUND & AIMS: The intestinal barrier protects intestinal cells from microbes and antigens in the lumen-breaches can alter the composition of the intestinal microbiota, the enteric immune system, and metabolism. We performed a screen to identify molecules that disrupt and support the intestinal epithelial barrier and tested their effects in mice. METHODS: We performed an imaging-based, quantitative, high-throughput screen (using CaCo-2 and T84 cells incubated with lipopolysaccharide; tumor necrosis factor; histamine; receptor antagonists; and libraries of secreted proteins, microbial metabolites, and drugs) to identify molecules that altered epithelial tight junction (TJ) and focal adhesion morphology. We then tested the effects of TJ stabilizers on these changes. Molecules we found to disrupt or stabilize TJs were administered mice with dextran sodium sulfate-induced colitis or Citrobacter rodentium-induced intestinal inflammation. Colon tissues were collected and analyzed by histology, fluorescence microscopy, and RNA sequencing. RESULTS: The screen identified numerous compounds that disrupted or stabilized (after disruption) TJs and monolayers of epithelial cells. We associated distinct morphologic alterations with changes in barrier function, and identified a variety of cytokines, metabolites, and drugs (including inhibitors of actomyosin contractility) that prevent disruption of TJs and restore TJ integrity. One of these disruptors (putrescine) disrupted TJ integrity in ex vivo mouse colon tissues; administration to mice exacerbated colon inflammation, increased gut permeability, reduced colon transepithelial electrical resistance, increased pattern recognition receptor ligands in mesenteric lymph nodes, and decreased colon length and survival times. Putrescine also increased intestine levels and fecal shedding of viable C rodentium, increased bacterial attachment to the colonic epithelium, and increased levels of inflammatory cytokines in colon tissues. Colonic epithelial cells from mice given putrescine increased expression of genes that regulate metal binding, oxidative stress, and cytoskeletal organization and contractility. Co-administration of taurine with putrescine blocked disruption of TJs and the exacerbated inflammation. CONCLUSIONS: We identified molecules that disrupt and stabilize intestinal epithelial TJs and barrier function and affect development of colon inflammation in mice. These agents might be developed for treatment of barrier intestinal impairment-associated and inflammatory disorders in patients, or avoided to prevent inflammation.


Subject(s)
Colitis/drug therapy , Colon/drug effects , Enterobacteriaceae Infections/drug therapy , Epithelial Cells/drug effects , Gastrointestinal Agents/pharmacology , High-Throughput Screening Assays , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Tight Junctions/drug effects , Animals , Caco-2 Cells , Citrobacter rodentium/pathogenicity , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Colon/metabolism , Colon/microbiology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastrointestinal Microbiome , Host-Pathogen Interactions , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice, Inbred C57BL , Permeability , Putrescine/pharmacology , Taurine/pharmacology , Tight Junctions/metabolism , Tight Junctions/microbiology , Tight Junctions/pathology
19.
Trends Immunol ; 38(4): 248-260, 2017 04.
Article in English | MEDLINE | ID: mdl-28214100

ABSTRACT

NLRP6, a member of the nucleotide-binding domain, leucine-rich repeat-containing (NLR) innate immune receptor family, regulates inflammation and host defense against microorganisms. Similar to other NLRs, NLRP6 not only participates in inflammasome formation, but is also involved in nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling regulation and facilitation of gastrointestinal antiviral effector functions. Additionally, NLRP6 contributes to the regulation of mucus secretion and antimicrobial peptide production, thereby impacting intestinal microbial colonization and associated microbiome-related infectious, autoinflammatory, metabolic, and neoplastic diseases. However, several of the mechanisms attributed to the functions of NLRP6 remain debatable, leaving open questions as to the relevant molecular mechanisms and interacting partners, and putative human relevance. We herein discuss recent findings related to NLRP6 activity, while highlighting outstanding questions and future perspectives in elucidating its roles in health and disease.


Subject(s)
Immunity, Innate , Inflammasomes/metabolism , Inflammation/immunology , Intestines/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions , Humans , Intestines/microbiology , Intracellular Signaling Peptides and Proteins/immunology , MAP Kinase Signaling System , Mice , NF-kappa B/metabolism
20.
Nature ; 514(7521): 181-6, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25231862

ABSTRACT

Non-caloric artificial sweeteners (NAS) are among the most widely used food additives worldwide, regularly consumed by lean and obese individuals alike. NAS consumption is considered safe and beneficial owing to their low caloric content, yet supporting scientific data remain sparse and controversial. Here we demonstrate that consumption of commonly used NAS formulations drives the development of glucose intolerance through induction of compositional and functional alterations to the intestinal microbiota. These NAS-mediated deleterious metabolic effects are abrogated by antibiotic treatment, and are fully transferrable to germ-free mice upon faecal transplantation of microbiota configurations from NAS-consuming mice, or of microbiota anaerobically incubated in the presence of NAS. We identify NAS-altered microbial metabolic pathways that are linked to host susceptibility to metabolic disease, and demonstrate similar NAS-induced dysbiosis and glucose intolerance in healthy human subjects. Collectively, our results link NAS consumption, dysbiosis and metabolic abnormalities, thereby calling for a reassessment of massive NAS usage.


Subject(s)
Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Glucose Intolerance/chemically induced , Glucose Intolerance/microbiology , Microbiota/drug effects , Sweetening Agents/adverse effects , Animals , Anti-Bacterial Agents/pharmacology , Aspartame/adverse effects , Body Weight/drug effects , Diet, High-Fat , Dietary Fats/pharmacology , Feces/microbiology , Female , Germ-Free Life , Glucose/metabolism , Glucose Intolerance/metabolism , Humans , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Mice , Mice, Inbred C57BL , Saccharin/administration & dosage , Saccharin/adverse effects , Sucrose/adverse effects , Sucrose/analogs & derivatives , Waist-Hip Ratio
SELECTION OF CITATIONS
SEARCH DETAIL