Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Infect Immun ; 91(2): e0057022, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36692308

ABSTRACT

A disrupted "dysbiotic" gut microbiome engenders susceptibility to the diarrheal pathogen Clostridioides difficile by impacting the metabolic milieu of the gut. Diet, in particular the microbiota-accessible carbohydrates (MACs) found in dietary fiber, is one of the most powerful ways to affect the composition and metabolic output of the gut microbiome. As such, diet is a powerful tool for understanding the biology of C. difficile and for developing alternative approaches for coping with this pathogen. One prominent class of metabolites produced by the gut microbiome is short-chain fatty acids (SCFAs), the major metabolic end products of MAC metabolism. SCFAs are known to decrease the fitness of C. difficile in vitro, and high intestinal SCFA concentrations are associated with reduced fitness of C. difficile in animal models of C. difficile infection (CDI). Here, we use controlled dietary conditions (8 diets that differ only by MAC composition) to show that C. difficile fitness is most consistently impacted by butyrate, rather than the other two prominent SCFAs (acetate and propionate), during murine model CDI. We similarly show that butyrate concentrations are lower in fecal samples from humans with CDI than in those from healthy controls. Finally, we demonstrate that butyrate impacts growth in diverse C. difficile isolates. These findings provide a foundation for future work which will dissect how butyrate directly impacts C. difficile fitness and will lead to the development of diverse approaches distinct from antibiotics or fecal transplant, such as dietary interventions, for mitigating CDI in at-risk human populations. IMPORTANCE Clostridioides difficile is a leading cause of infectious diarrhea in humans, and it imposes a tremendous burden on the health care system. Current treatments for C. difficile infection (CDI) include antibiotics and fecal microbiota transplant, which contribute to recurrent CDIs and face major regulatory hurdles, respectively. Therefore, there is an ongoing need to develop new ways to cope with CDI. Notably, a disrupted "dysbiotic" gut microbiota is the primary risk factor for CDI, but we incompletely understand how a healthy microbiota resists CDI. Here, we show that a specific molecule produced by the gut microbiota, butyrate, is negatively associated with C. difficile burdens in humans and in a mouse model of CDI and that butyrate impedes the growth of diverse C. difficile strains in pure culture. These findings help to build a foundation for designing alternative, possibly diet-based, strategies for mitigating CDI in humans.


Subject(s)
Clostridioides difficile , Clostridium Infections , Humans , Animals , Mice , Butyrates , Permissiveness , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Fatty Acids, Volatile
2.
Nat Commun ; 14(1): 512, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36720857

ABSTRACT

The human gut microbiota produces dozens of small molecules that circulate in blood, accumulate to comparable levels as pharmaceutical drugs, and influence host physiology. Despite the importance of these metabolites to human health and disease, the origin of most microbially-produced molecules and their fate in the host remains largely unknown. Here, we uncover a host-microbe co-metabolic pathway for generation of hippuric acid, one of the most abundant organic acids in mammalian urine. Combining stable isotope tracing with bacterial and host genetics, we demonstrate reduction of phenylalanine to phenylpropionic acid by gut bacteria; the host re-oxidizes phenylpropionic acid involving medium-chain acyl-CoA dehydrogenase (MCAD). Generation of germ-free male and female MCAD-/- mice enabled gnotobiotic colonization combined with untargeted metabolomics to identify additional microbial metabolites processed by MCAD in host circulation. Our findings uncover a host-microbe pathway for the abundant, non-toxic phenylalanine metabolite hippurate and identify ß-oxidation via MCAD as a novel mechanism by which mammals metabolize microbiota-derived metabolites.


Subject(s)
Hippurates , Metabolomics , Animals , Female , Humans , Male , Mice , Acyl-CoA Dehydrogenase , Phenylalanine
3.
Nat Microbiol ; 7(5): 695-706, 2022 05.
Article in English | MEDLINE | ID: mdl-35505245

ABSTRACT

Gut bacteria face a key problem in how they capture enough energy to sustain their growth and physiology. The gut bacterium Clostridium sporogenes obtains its energy by utilizing amino acids in pairs, coupling the oxidation of one to the reduction of another-the Stickland reaction. Oxidative pathways produce ATP via substrate-level phosphorylation, whereas reductive pathways are thought to balance redox. In the present study, we investigated whether these reductive pathways are also linked to energy generation and the production of microbial metabolites that may circulate and impact host physiology. Using metabolomics, we find that, during growth in vitro, C. sporogenes produces 15 metabolites, 13 of which are present in the gut of C. sporogenes-colonized mice. Four of these compounds are reductive Stickland metabolites that circulate in the blood of gnotobiotic mice and are also detected in plasma from healthy humans. Gene clusters for reductive Stickland pathways suggest involvement of electron transfer proteins, and experiments in vitro demonstrate that reductive metabolism is coupled to ATP formation and not just redox balance. Genetic analysis points to the broadly conserved Rnf complex as a key coupling site for energy transduction. Rnf complex mutants show aberrant amino acid metabolism in a defined medium and are attenuated for growth in the mouse gut, demonstrating a role of the Rnf complex in Stickland metabolism and gut colonization. Our findings reveal that the production of circulating metabolites by a commensal bacterium within the host gut is linked to an ATP-yielding redox process.


Subject(s)
Clostridium , Metabolomics , Adenosine Triphosphate/metabolism , Animals , Bacteria/metabolism , Clostridium/genetics , Clostridium/metabolism , Fermentation , Mice
4.
Nat Metab ; 4(1): 19-28, 2022 01.
Article in English | MEDLINE | ID: mdl-34992297

ABSTRACT

The enteric pathogen Clostridioides difficile (Cd) is responsible for a toxin-mediated infection that causes more than 200,000 recorded hospitalizations and 13,000 deaths in the United States every year1. However, Cd can colonize the gut in the absence of disease symptoms. Prevalence of asymptomatic colonization by toxigenic Cd in healthy populations is high; asymptomatic carriers are at increased risk of infection compared to noncolonized individuals and may be a reservoir for transmission of Cd infection2,3. Elucidating the molecular mechanisms by which Cd persists in the absence of disease is necessary for understanding pathogenesis and developing refined therapeutic strategies. Here, we show with gut microbiome metatranscriptomic analysis that mice recalcitrant to Cd infection and inflammation exhibit increased community-wide expression of arginine and ornithine metabolic pathways. To query Cd metabolism specifically, we leverage RNA sequencing in gnotobiotic mice infected with two wild-type strains (630 and R20291) and isogenic toxin-deficient mutants of these strains to differentiate inflammation-dependent versus -independent transcriptional states. A single operon encoding oxidative ornithine degradation is consistently upregulated across non-toxigenic Cd strains. Combining untargeted and targeted metabolomics with bacterial and host genetics, we demonstrate that both diet- and host-derived sources of ornithine provide a competitive advantage to Cd, suggesting a mechanism for Cd persistence within a non-inflammatory, healthy gut.


Subject(s)
Clostridioides difficile/physiology , Clostridium Infections/metabolism , Clostridium Infections/microbiology , Host-Pathogen Interactions , Ornithine/metabolism , Oxidation-Reduction , Amino Acids/metabolism , Animals , Energy Metabolism , Gastrointestinal Microbiome , Humans , Metabolic Networks and Pathways , Metabolome , Metabolomics/methods , Mice , Nitric Oxide Synthase/metabolism , Oxidative Stress
5.
Cell ; 184(16): 4137-4153.e14, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34256014

ABSTRACT

Diet modulates the gut microbiome, which in turn can impact the immune system. Here, we determined how two microbiota-targeted dietary interventions, plant-based fiber and fermented foods, influence the human microbiome and immune system in healthy adults. Using a 17-week randomized, prospective study (n = 18/arm) combined with -omics measurements of microbiome and host, including extensive immune profiling, we found diet-specific effects. The high-fiber diet increased microbiome-encoded glycan-degrading carbohydrate active enzymes (CAZymes) despite stable microbial community diversity. Although cytokine response score (primary outcome) was unchanged, three distinct immunological trajectories in high-fiber consumers corresponded to baseline microbiota diversity. Alternatively, the high-fermented-food diet steadily increased microbiota diversity and decreased inflammatory markers. The data highlight how coupling dietary interventions to deep and longitudinal immune and microbiome profiling can provide individualized and population-wide insight. Fermented foods may be valuable in countering the decreased microbiome diversity and increased inflammation pervasive in industrialized society.


Subject(s)
Diet , Gastrointestinal Microbiome , Immunity , Biodiversity , Dietary Fiber/pharmacology , Feeding Behavior , Female , Fermented Foods , Gastrointestinal Microbiome/drug effects , Humans , Inflammation/pathology , Male , Middle Aged , Signal Transduction/drug effects
7.
Cell Host Microbe ; 28(3): 371-379.e5, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32652063

ABSTRACT

Our emerging view of the gut microbiome largely focuses on bacteria, while less is known about other microbial components, such as bacteriophages (phages). Though phages are abundant in the gut, very few phages have been isolated from this ecosystem. Here, we report the genomes of 27 phages from the United States and Bangladesh that infect the prevalent human gut bacterium Bacteroides thetaiotaomicron. These phages are mostly distinct from previously sequenced phages with the exception of two, which are crAss-like phages. We compare these isolates to existing human gut metagenomes, revealing similarities to previously inferred phages and additional unexplored phage diversity. Finally, we use host tropisms of these phages to identify alleles of phage structural genes associated with infectivity. This work provides a detailed view of the gut's "viral dark matter" and a framework for future efforts to further integrate isolation- and sequencing-focused efforts to understand gut-resident phages.


Subject(s)
Bacteriophages/classification , Bacteriophages/genetics , Bacteroides thetaiotaomicron/virology , Host Specificity/genetics , Viral Tropism/genetics , Bacteriophages/isolation & purification , Bacteroides thetaiotaomicron/genetics , Bangladesh , Biodiversity , Gastrointestinal Microbiome , Genome, Viral , Genomics , Humans , Metagenome/genetics , Phylogeny , Sequence Analysis , United States , Whole Genome Sequencing
8.
Cell Rep Med ; 1(1)2020 04 21.
Article in English | MEDLINE | ID: mdl-32483557

ABSTRACT

Clostridium difficile infection (CDI) is an enteric bacterial disease that is increasing in prevalence worldwide. C. difficile capitalizes on gut inflammation and microbiome dysbiosis to establish infection, with symptoms ranging from watery diarrhea to toxic megacolon. We reported that the safe-in-human clinical drug ebselen (ClinicalTrials.gov: NCT03013400, NCT01452607, NCT00762671, and NCT02603081) has biochemical, cell-based, and in vivo efficacy against the toxins of C. difficile. Here, we show that ebselen treatment reduces recurrence rates and decreases colitis in a hamster model of relapsing CDI. Furthermore, ebselen treatment does not alter microbiome diversity and promotes recovery back to that of healthy controls after antibiotic-induced dysbiosis in healthy and C. difficile-infected mice. This increased microbiome recovery upon ebselen treatment correlates with a decrease in host-derived inflammatory markers, suggesting that the anti-inflammatory properties of ebselen, combined with its anti-toxin function, help to mitigate the major clinical challenges of CDI, including recurrence, microbial dysbiosis, and colitis.


Subject(s)
Clostridium Infections/drug therapy , Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , Inflammation/drug therapy , Isoindoles/therapeutic use , Organoselenium Compounds/therapeutic use , Animals , Clostridioides difficile/drug effects , Clostridioides difficile/physiology , Clostridium Infections/complications , Cricetinae , Disease Models, Animal , Dysbiosis/microbiology , Enterocolitis, Pseudomembranous/complications , Enterocolitis, Pseudomembranous/drug therapy , Female , Gastrointestinal Microbiome/physiology , Inflammation/microbiology , Male , Mesocricetus , Mice
9.
Annu Rev Pathol ; 15: 345-369, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31622559

ABSTRACT

The human gastrointestinal tract is home to an incredibly dense population of microbes. These microbes employ unique strategies to capture energy in this largely anaerobic environment. In the process of breaking down dietary- and host-derived substrates, the gut microbiota produce a broad range of metabolic products that accumulate to high levels in the gut. Increasingly, studies are revealing that these chemicals impact host biology, either by acting on cells within the gastrointestinal tract or entering circulation and exerting their effects at distal sites within the body. Given the high level of functional diversity in the gut microbiome and the varied diets that we consume, the repertoire of microbiota-derived molecules within our bodies varies dramatically across individuals. Thus, the microbes in our gut and the metabolic end products they produce represent a phenotypic lever that we can potentially control to develop new therapeutics for personalized medicine. Here, we review current understanding of how microbes in the gastrointestinal tract contribute to the molecules within our gut and those that circulate within our bodies. We also highlight examples of how these molecules affect host physiology and discuss potential strategies for controlling their production to promote human health and to treat disease.


Subject(s)
Gastrointestinal Microbiome/physiology , Health , Metabolic Diseases/microbiology , Metabolome/physiology , Diet , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Humans , Metabolic Diseases/etiology
10.
Cell Host Microbe ; 26(5): 650-665.e4, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31726029

ABSTRACT

Antibiotics alter microbiota composition and increase infection susceptibility. However, the generalizable effects of antibiotics on and the contribution of environmental variables to gut commensals remain unclear. To address this, we tracked microbiota dynamics with high temporal and taxonomic resolution during antibiotic treatment in a controlled murine system by isolating variables such as diet, treatment history, and housing co-inhabitants. Human microbiotas were remarkably resilient and recovered during antibiotic treatment, with transient dominance of resistant Bacteroides and taxa-asymmetric diversity reduction. In certain cases, in vitro sensitivities were not predictive of in vivo responses, underscoring the significance of host and community context. A fiber-deficient diet exacerbated microbiota collapse and delayed recovery. Species replacement through cross housing after ciprofloxacin treatment established resilience to a second treatment. Single housing drastically disrupted recovery, highlighting the importance of environmental reservoirs. Our findings highlight deterministic microbiota adaptations to perturbations and the translational potential for modulating diet, sanitation, and microbiota composition during antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Load/drug effects , Bacteroides/growth & development , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/microbiology , Animals , Bacteroides/classification , Bacteroides/isolation & purification , Biodiversity , Ciprofloxacin/pharmacology , Diet , Female , Gastrointestinal Tract/drug effects , Germ-Free Life , Humans , Male , Mice , Rifaximin/pharmacology , Streptomycin/pharmacology
11.
Gut Microbes ; 10(2): 216-227, 2019.
Article in English | MEDLINE | ID: mdl-30118385

ABSTRACT

The study of traditional populations provides a view of human-associated microbes unperturbed by industrialization, as well as a window into the microbiota that co-evolved with humans. Here we discuss our recent work characterizing the microbiota from the Hadza hunter-gatherers of Tanzania. We found seasonal shifts in bacterial taxa, diversity, and carbohydrate utilization by the microbiota. When compared to the microbiota composition from other populations around the world, the Hadza microbiota shares bacterial families with other traditional societies that are rare or absent from microbiotas of industrialized nations. We present additional observations from the Hadza microbiota and their lifestyle and environment, including microbes detected on hands, water, and animal sources, how the microbiota varies with sex and age, and the short-term effects of introducing agricultural products into the diet. In the context of our previously published findings and of these additional observations, we discuss a path forward for future work.


Subject(s)
Diet/ethnology , Environmental Microbiology , Gastrointestinal Microbiome , Life Style/ethnology , Age Factors , Animals , Biodiversity , Dietary Carbohydrates/metabolism , Feces/microbiology , Female , Humans , Male , Seasons , Tanzania/ethnology
12.
Nat Microbiol ; 3(6): 662-669, 2018 06.
Article in English | MEDLINE | ID: mdl-29686297

ABSTRACT

Clostridium difficile is an opportunistic diarrhoeal pathogen, and C. difficile infection (CDI) represents a major health care concern, causing an estimated 15,000 deaths per year in the United States alone 1 . Several enteric pathogens, including C. difficile, leverage inflammation and the accompanying microbial dysbiosis to thrive in the distal gut 2 . Although diet is among the most powerful available tools for affecting the health of humans and their relationship with their microbiota, investigation into the effects of diet on CDI has been limited. Here, we show in mice that the consumption of microbiota-accessible carbohydrates (MACs) found in dietary plant polysaccharides has a significant effect on CDI. Specifically, using a model of antibiotic-induced CDI that typically resolves within 12 days of infection, we demonstrate that MAC-deficient diets perpetuate CDI. We show that C. difficile burdens are suppressed through the addition of either a diet containing a complex mixture of MACs or a simplified diet containing inulin as the sole MAC source. We show that switches between these dietary conditions are coincident with changes to microbiota membership, its metabolic output and C. difficile-mediated inflammation. Together, our data demonstrate the outgrowth of MAC-utilizing taxa and the associated end products of MAC metabolism, namely, the short-chain fatty acids acetate, propionate and butyrate, are associated with decreased C. difficile fitness despite increased C. difficile toxin expression in the gut. Our findings, when placed into the context of the known fibre deficiencies of a human Western diet, provide rationale for pursuing MAC-centric dietary strategies as an alternate line of investigation for mitigating CDI.


Subject(s)
Anti-Bacterial Agents/adverse effects , Clostridium Infections/diet therapy , Dietary Carbohydrates/administration & dosage , Dysbiosis/diet therapy , Plants/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridium Infections/chemically induced , Clostridium Infections/complications , Dietary Carbohydrates/pharmacology , Disease Models, Animal , Dysbiosis/etiology , Gastrointestinal Microbiome/drug effects , Humans , Inulin/administration & dosage , Inulin/pharmacology , Mice , Treatment Outcome
13.
Nature ; 551(7682): 648-652, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29168502

ABSTRACT

The human gut microbiota produces dozens of metabolites that accumulate in the bloodstream, where they can have systemic effects on the host. Although these small molecules commonly reach concentrations similar to those achieved by pharmaceutical agents, remarkably little is known about the microbial metabolic pathways that produce them. Here we use a combination of genetics and metabolic profiling to characterize a pathway from the gut symbiont Clostridium sporogenes that generates aromatic amino acid metabolites. Our results reveal that this pathway produces twelve compounds, nine of which are known to accumulate in host serum. All three aromatic amino acids (tryptophan, phenylalanine and tyrosine) serve as substrates for the pathway, and it involves branching and alternative reductases for specific intermediates. By genetically manipulating C. sporogenes, we modulate serum levels of these metabolites in gnotobiotic mice, and show that in turn this affects intestinal permeability and systemic immunity. This work has the potential to provide the basis of a systematic effort to engineer the molecular output of the gut bacterial community.


Subject(s)
Amino Acids, Aromatic/metabolism , Closterium/metabolism , Gastrointestinal Microbiome/physiology , Metabolic Networks and Pathways , Metabolome/physiology , Serum/chemistry , Serum/metabolism , Amino Acids, Aromatic/blood , Animals , Blood Chemical Analysis , Closterium/genetics , Gastrointestinal Microbiome/genetics , Germ-Free Life , Humans , Immunity , Indoles/blood , Indoles/metabolism , Intestinal Mucosa/metabolism , Male , Metabolic Networks and Pathways/genetics , Metabolomics , Mice , Multigene Family/genetics , Permeability , Phenylalanine/metabolism , Tryptophan/metabolism , Tyrosine/metabolism
14.
Front Microbiol ; 7: 333, 2016.
Article in English | MEDLINE | ID: mdl-27014249

ABSTRACT

Skin bacterial communities can protect amphibians from a fungal pathogen; however, little is known about how these communities are maintained. We used a neutral model of community ecology to identify bacteria that are maintained on salamanders by selection or by dispersal from a bacterial reservoir (soil) and ecological drift. We found that 75% (9/12) of bacteria that were consistent with positive selection, <1% of bacteria that were consistent with random dispersal and none of the bacteria that were consistent under negative selection had a 97% or greater match to antifungal isolates. Additionally we performed an experiment where salamanders were either provided or denied a bacterial reservoir and estimated immigration and loss (emigration and local extinction) rates of bacteria on salamanders in both treatments. Loss was strongly related to bacterial richness, suggesting competition is important for structuring the community. Bacteria closely related to antifungal isolates were more likely to persist on salamanders with or without a bacterial reservoir, suggesting they had a competitive advantage. Furthermore, over-represented and under-represented operational taxonomic units (OTUs) had similar persistence on salamanders when a bacterial reservoir was present. However, under-represented OTUs were less likely to persist in the absence of a bacterial reservoir, suggesting that the over-represented and under-represented bacteria were selected against or for on salamanders through time. Our findings from the neutral model, migration and persistence analyses show that bacteria that exhibit a high similarity to antifungal isolates persist on salamanders, which likely protect hosts against pathogens and improve fitness. This research is one of the first to apply ecological theory to investigate assembly of host associated-bacterial communities, which can provide insights for probiotic bioaugmentation as a conservation strategy against disease.

15.
Cell ; 159(4): 789-99, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25417156

ABSTRACT

Host genetics and the gut microbiome can both influence metabolic phenotypes. However, whether host genetic variation shapes the gut microbiome and interacts with it to affect host phenotype is unclear. Here, we compared microbiotas across >1,000 fecal samples obtained from the TwinsUK population, including 416 twin pairs. We identified many microbial taxa whose abundances were influenced by host genetics. The most heritable taxon, the family Christensenellaceae, formed a co-occurrence network with other heritable Bacteria and with methanogenic Archaea. Furthermore, Christensenellaceae and its partners were enriched in individuals with low body mass index (BMI). An obese-associated microbiome was amended with Christensenella minuta, a cultured member of the Christensenellaceae, and transplanted to germ-free mice. C. minuta amendment reduced weight gain and altered the microbiome of recipient mice. Our findings indicate that host genetics influence the composition of the human gut microbiome and can do so in ways that impact host metabolism.


Subject(s)
Bacteria/classification , Bacteria/isolation & purification , Feces/microbiology , Microbiota , Animals , Bacteria/metabolism , Body Mass Index , Female , Gastrointestinal Tract/microbiology , Germ-Free Life , Humans , Male , Mice , Obesity/microbiology , Twins, Dizygotic , Twins, Monozygotic
16.
Cell ; 159(2): 253-66, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25284151

ABSTRACT

To study how microbes establish themselves in a mammalian gut environment, we colonized germ-free mice with microbial communities from human, zebrafish, and termite guts, human skin and tongue, soil, and estuarine microbial mats. Bacteria from these foreign environments colonized and persisted in the mouse gut; their capacity to metabolize dietary and host carbohydrates and bile acids correlated with colonization success. Cohousing mice harboring these xenomicrobiota or a mouse cecal microbiota, along with germ-free "bystanders," revealed the success of particular bacterial taxa in invading guts with established communities and empty gut habitats. Unanticipated patterns of ecological succession were observed; for example, a soil-derived bacterium dominated even in the presence of bacteria from other gut communities (zebrafish and termite), and human-derived bacteria colonized germ-free bystander mice before mouse-derived organisms. This approach can be generalized to address a variety of mechanistic questions about succession, including succession in the context of microbiota-directed therapeutics.


Subject(s)
Bacteria/classification , Bacteria/growth & development , Gastrointestinal Tract/microbiology , Mice/microbiology , Animals , Bacteria/metabolism , Ecosystem , Estuaries , Germ-Free Life , Humans , Isoptera/microbiology , Microbial Interactions , Skin/microbiology , Soil Microbiology , Symbiosis , Tongue/microbiology , Zebrafish/microbiology
17.
Microbiome ; 2: 12, 2014.
Article in English | MEDLINE | ID: mdl-24739094

ABSTRACT

To augment capacity-building for microbiome and probiotic research in Africa, a workshop was held in Nairobi, Kenya, at which researchers discussed human, animal, insect, and agricultural microbiome and probiotics/prebiotics topics. Five recommendations were made to promote future basic and translational research that benefits Africans.

18.
Science ; 341(6150): 1241214, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-24009397

ABSTRACT

The role of specific gut microbes in shaping body composition remains unclear. We transplanted fecal microbiota from adult female twin pairs discordant for obesity into germ-free mice fed low-fat mouse chow, as well as diets representing different levels of saturated fat and fruit and vegetable consumption typical of the U.S. diet. Increased total body and fat mass, as well as obesity-associated metabolic phenotypes, were transmissible with uncultured fecal communities and with their corresponding fecal bacterial culture collections. Cohousing mice harboring an obese twin's microbiota (Ob) with mice containing the lean co-twin's microbiota (Ln) prevented the development of increased body mass and obesity-associated metabolic phenotypes in Ob cage mates. Rescue correlated with invasion of specific members of Bacteroidetes from the Ln microbiota into Ob microbiota and was diet-dependent. These findings reveal transmissible, rapid, and modifiable effects of diet-by-microbiota interactions.


Subject(s)
Adiposity , Bacteroidetes/physiology , Gastrointestinal Tract/microbiology , Metagenome/physiology , Obesity/metabolism , Adult , Animals , Bacteroidetes/genetics , Cecum/metabolism , Cecum/microbiology , Diet, Fat-Restricted , Feces/microbiology , Female , Germ-Free Life , Humans , Metabolome , Metagenome/genetics , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Thinness/microbiology , Twins , Weight Gain , Young Adult
19.
Bioessays ; 35(9): 810-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23836415

ABSTRACT

Large-scale characterization of the human microbiota has largely focused on Western adults, yet these populations may be uncharacteristic because of their diets and lifestyles. In particular, the rise of "Western diseases" may in part stem from reduced exposure to, or even loss of, microbes with which humans have coevolved. Here, we review beneficial microbes associated with pathogen resistance, highlighting the emerging role of complex microbial communities in protecting against disease. We discuss ways in which modern lifestyles and practices may deplete physiologically important microbiota, and explore prospects for reintroducing or encouraging the growth of beneficial microbes to promote the restoration of healthy microbial ecosystems.


Subject(s)
Gastrointestinal Tract/microbiology , Metagenome , Microbiota , Animals , Disease , Ecosystem , Gastrointestinal Tract/immunology , Host-Pathogen Interactions/immunology , Humans , Probiotics/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...