Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 456
Filter
1.
Cell ; 185(14): 2495-2509.e11, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35764090

ABSTRACT

Plant fibers in byproduct streams produced by non-harsh food processing methods represent biorepositories of diverse, naturally occurring, and physiologically active biomolecules. To demonstrate one approach for their characterization, mass spectrometry of intestinal contents from gnotobiotic mice, plus in vitro studies, revealed liberation of N-methylserotonin from orange fibers by human gut microbiota members including Bacteroides ovatus. Functional genomic analyses of B. ovatus strains grown under permissive and non-permissive N-methylserotonin "mining" conditions revealed polysaccharide utilization loci that target pectins whose expression correlate with strain-specific liberation of this compound. N-methylserotonin, orally administered to germ-free mice, reduced adiposity, altered liver glycogenesis, shortened gut transit time, and changed expression of genes that regulate circadian rhythm in the liver and colon. In human studies, dose-dependent, orange-fiber-specific fecal accumulation of N-methylserotonin positively correlated with levels of microbiome genes encoding enzymes that digest pectic glycans. Identifying this type of microbial mining activity has potential therapeutic implications.


Subject(s)
Citrus sinensis , Gastrointestinal Microbiome , Animals , Citrus sinensis/metabolism , Dietary Fiber , Gastrointestinal Microbiome/physiology , Germ-Free Life , Humans , Mice , Pectins/metabolism , Polysaccharides/metabolism , Serotonin/analogs & derivatives
2.
Cell ; 179(1): 59-73.e13, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31539500

ABSTRACT

Development of microbiota-directed foods (MDFs) that selectively increase the abundance of beneficial human gut microbes, and their expressed functions, requires knowledge of both the bioactive components of MDFs and the mechanisms underlying microbe-microbe interactions. Here, gnotobiotic mice were colonized with a defined consortium of human-gut-derived bacterial strains and fed different combinations of 34 food-grade fibers added to a representative low-fiber diet consumed in the United States. Bioactive carbohydrates in fiber preparations targeting particular Bacteroides species were identified using community-wide quantitative proteomic analyses of bacterial gene expression coupled with forward genetic screens. Deliberate manipulation of community membership combined with administration of retrievable artificial food particles, consisting of paramagnetic microscopic beads coated with dietary polysaccharides, disclosed the contributions of targeted species to fiber degradation. Our approach, including the use of bead-based biosensors, defines nutrient-harvesting strategies that underlie, as well as alleviate, competition between Bacteroides and control the selectivity of MDF components.


Subject(s)
Bacteroides/genetics , Dietary Fiber/pharmacology , Gastrointestinal Microbiome/drug effects , Germ-Free Life/physiology , Microbial Interactions/drug effects , Polysaccharides/pharmacology , Proteomics/methods , Animals , Diet/methods , Dietary Fiber/metabolism , Feces/microbiology , Gastrointestinal Microbiome/physiology , Gene Expression Regulation, Bacterial/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Polysaccharides/metabolism
4.
Cell ; 167(5): 1339-1353.e21, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863247

ABSTRACT

Despite the accepted health benefits of consuming dietary fiber, little is known about the mechanisms by which fiber deprivation impacts the gut microbiota and alters disease risk. Using a gnotobiotic mouse model, in which animals were colonized with a synthetic human gut microbiota composed of fully sequenced commensal bacteria, we elucidated the functional interactions between dietary fiber, the gut microbiota, and the colonic mucus barrier, which serves as a primary defense against enteric pathogens. We show that during chronic or intermittent dietary fiber deficiency, the gut microbiota resorts to host-secreted mucus glycoproteins as a nutrient source, leading to erosion of the colonic mucus barrier. Dietary fiber deprivation, together with a fiber-deprived, mucus-eroding microbiota, promotes greater epithelial access and lethal colitis by the mucosal pathogen, Citrobacter rodentium. Our work reveals intricate pathways linking diet, the gut microbiome, and intestinal barrier dysfunction, which could be exploited to improve health using dietary therapeutics.


Subject(s)
Dietary Fiber/administration & dosage , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Animals , Citrobacter rodentium/physiology , Colitis/microbiology , Colon/microbiology , Disease Susceptibility , Enterobacteriaceae Infections/microbiology , Escherichia coli , Female , Germ-Free Life , Humans , Male , Mice , Mucin-2/genetics
5.
Nature ; 625(7993): 157-165, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38093016

ABSTRACT

Evidence is accumulating that perturbed postnatal development of the gut microbiome contributes to childhood malnutrition1-4. Here we analyse biospecimens from a randomized, controlled trial of a microbiome-directed complementary food (MDCF-2) that produced superior rates of weight gain compared with a calorically more dense conventional ready-to-use supplementary food in 12-18-month-old Bangladeshi children with moderate acute malnutrition4. We reconstructed 1,000 bacterial genomes (metagenome-assembled genomes (MAGs)) from the faecal microbiomes of trial participants, identified 75 MAGs of which the abundances were positively associated with ponderal growth (change in weight-for-length Z score (WLZ)), characterized changes in MAG gene expression as a function of treatment type and WLZ response, and quantified carbohydrate structures in MDCF-2 and faeces. The results reveal that two Prevotella copri MAGs that are positively associated with WLZ are the principal contributors to MDCF-2-induced expression of metabolic pathways involved in utilizing the component glycans of MDCF-2. The predicted specificities of carbohydrate-active enzymes expressed by their polysaccharide-utilization loci are correlated with (1) the in vitro growth of Bangladeshi P. copri strains, possessing varying degrees of polysaccharide-utilization loci and genomic conservation with these MAGs, in defined medium containing different purified glycans representative of those in MDCF-2, and (2) the levels of faecal carbohydrate structures in the trial participants. These associations suggest that identifying bioactive glycan structures in MDCFs metabolized by growth-associated bacterial taxa will help to guide recommendations about their use in children with acute malnutrition and enable the development of additional formulations.


Subject(s)
Food , Gastrointestinal Microbiome , Malnutrition , Polysaccharides , Humans , Infant , Bacteria/genetics , Bangladesh , Body Weight/genetics , Feces/microbiology , Gastrointestinal Microbiome/physiology , Genome, Bacterial/genetics , Malnutrition/microbiology , Metagenome/genetics , Polysaccharides/metabolism , Weight Gain
6.
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
7.
Nature ; 595(7865): 91-95, 2021 07.
Article in English | MEDLINE | ID: mdl-34163075

ABSTRACT

Changing food preferences brought about by westernization that have deleterious health effects1,2-combined with myriad forces that are contributing to increased food insecurity-are catalysing efforts to identify more nutritious and affordable foods3. Consumption of dietary fibre can help to prevent cardiovascular disease, type 2 diabetes and obesity4-6. A substantial number of reports have explored the effects of dietary fibre on the gut microbial community7-9. However, the microbiome is complex, dynamic and exhibits considerable intra- and interpersonal variation in its composition and functions. The large number of potential interactions between the components of the microbiome makes it challenging to define the mechanisms by which food ingredients affect community properties. Here we address the question of how foods containing different fibre preparations can be designed to alter functions associated with specific components of the microbiome. Because a marked increase in snack consumption is associated with westernization, we formulated snack prototypes using plant fibres from different sustainable sources that targeted distinct features of the gut microbiomes of individuals with obesity when transplanted into gnotobiotic mice. We used these snacks to supplement controlled diets that were consumed by adult individuals with obesity or who were overweight. Fibre-specific changes in their microbiomes were linked to changes in their plasma proteomes indicative of an altered physiological state.


Subject(s)
Dietary Fiber/pharmacology , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Snacks , Adolescent , Adult , Animals , Bacteroides/drug effects , Bacteroides/isolation & purification , Blood Proteins/analysis , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/microbiology , Overweight/microbiology , Proteome/analysis , Proteome/drug effects , Young Adult
8.
Nature ; 588(7839): 688-692, 2020 12.
Article in English | MEDLINE | ID: mdl-33268895

ABSTRACT

Inflammasomes are important sentinels of innate immune defence that are activated in response to diverse stimuli, including pathogen-associated molecular patterns (PAMPs)1. Activation of the inflammasome provides host defence against aspergillosis2,3, which is a major health concern for patients who are immunocompromised. However, the Aspergillus fumigatus PAMPs that are responsible for inflammasome activation are not known. Here we show that the polysaccharide galactosaminogalactan (GAG) of A. fumigatus is a PAMP that activates the NLRP3 inflammasome. The binding of GAG to ribosomal proteins inhibited cellular translation machinery, and thus activated the NLRP3 inflammasome. The galactosamine moiety bound to ribosomal proteins and blocked cellular translation, which triggered activation of the NLRP3 inflammasome. In mice, a GAG-deficient Aspergillus mutant (Δgt4c) did not elicit protective activation of the inflammasome, and this strain exhibited enhanced virulence. Moreover, administration of GAG protected mice from colitis induced by dextran sulfate sodium in an inflammasome-dependent manner. Thus, ribosomes connect the sensing of this fungal PAMP to the activation of an innate immune response.


Subject(s)
Aspergillosis/prevention & control , Aspergillus fumigatus/metabolism , Inflammasomes/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Polysaccharides/metabolism , Animals , Aspergillosis/immunology , Aspergillosis/microbiology , Aspergillus fumigatus/immunology , Biofilms , Colitis/chemically induced , Colitis/prevention & control , Dextran Sulfate , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Immunity, Innate , Inflammasomes/immunology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polysaccharides/immunology , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism
9.
Proc Natl Acad Sci U S A ; 120(39): e2311422120, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37733741

ABSTRACT

Understanding how members of the human gut microbiota prioritize nutrient resources is one component of a larger effort to decipher the mechanisms defining microbial community robustness and resiliency in health and disease. This knowledge is foundational for development of microbiota-directed therapeutics. To model how bacteria prioritize glycans in the gut, germfree mice were colonized with 13 human gut bacterial strains, including seven saccharolytic Bacteroidaceae species. Animals were fed a Western diet supplemented with pea fiber. After community assembly, an inducible CRISPR-based system was used to selectively and temporarily reduce the absolute abundance of Bacteroides thetaiotaomicron or B. cellulosilyticus by 10- to 60-fold. Each knockdown resulted in specific, reproducible increases in the abundances of other Bacteroidaceae and dynamic alterations in their expression of genes involved in glycan utilization. Emergence of these "alternate consumers" was associated with preservation of community saccharolytic activity. Using an inducible system for CRISPR base editing in vitro, we disrupted translation of transporters critical for utilizing dietary polysaccharides in Phocaeicola vulgatus, a B. cellulosilyticus knockdown-responsive taxon. In vitro and in vivo tests of the resulting P. vulgatus mutants allowed us to further characterize mechanisms associated with its increased fitness after knockdown. In principle, the approach described can be applied to study utilization of a range of nutrients and to preclinical efforts designed to develop therapeutic strategies for precision manipulation of microbial communities.


Subject(s)
Bacteroides thetaiotaomicron , Bacteroides , Humans , Animals , Mice , Bacteroides/genetics , Polysaccharides , Bacteroides thetaiotaomicron/genetics , Biological Assay , Diet, Western
10.
Proc Natl Acad Sci U S A ; 120(10): e2214076120, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36848567

ABSTRACT

Lentinula is a broadly distributed group of fungi that contains the cultivated shiitake mushroom, L. edodes. We sequenced 24 genomes representing eight described species and several unnamed lineages of Lentinula from 15 countries on four continents. Lentinula comprises four major clades that arose in the Oligocene, three in the Americas and one in Asia-Australasia. To expand sampling of shiitake mushrooms, we assembled 60 genomes of L. edodes from China that were previously published as raw Illumina reads and added them to our dataset. Lentinula edodes sensu lato (s. lat.) contains three lineages that may warrant recognition as species, one including a single isolate from Nepal that is the sister group to the rest of L. edodes s. lat., a second with 20 cultivars and 12 wild isolates from China, Japan, Korea, and the Russian Far East, and a third with 28 wild isolates from China, Thailand, and Vietnam. Two additional lineages in China have arisen by hybridization among the second and third groups. Genes encoding cysteine sulfoxide lyase (lecsl) and γ-glutamyl transpeptidase (leggt), which are implicated in biosynthesis of the organosulfur flavor compound lenthionine, have diversified in Lentinula. Paralogs of both genes that are unique to Lentinula (lecsl 3 and leggt 5b) are coordinately up-regulated in fruiting bodies of L. edodes. The pangenome of L. edodes s. lat. contains 20,308 groups of orthologous genes, but only 6,438 orthogroups (32%) are shared among all strains, whereas 3,444 orthogroups (17%) are found only in wild populations, which should be targeted for conservation.


Subject(s)
Lentinula , Phylogeny , Asia, Eastern , Thailand
11.
Semin Cell Dev Biol ; 134: 112-124, 2023 Jan 30.
Article in English | MEDLINE | ID: mdl-35307283

ABSTRACT

In brown algae, the extracellular matrix (ECM) and its constitutive polymers play crucial roles in specialized functions, including algal growth and development. In this review we offer an integrative view of ECM construction in brown algae. We briefly report the chemical composition of its main constituents, and how these are interlinked in a structural model. We examine the ECM assembly at the tissue and cell level, with consideration on its structure in vivo and on the putative subcellular sites for the synthesis of its main constituents. We further discuss the biosynthetic pathways of two major polysaccharides, alginates and sulfated fucans, and the progress made beyond the candidate genes with the biochemical validation of encoded proteins. Key enzymes involved in the elongation of the glycan chains are still unknown and predictions have been made at the gene level. Here, we offer a re-examination of some glycosyltransferases and sulfotransferases from published genomes. Overall, our analysis suggests novel investigations to be performed at both the cellular and biochemical levels. First, to depict the location of polysaccharide structures in tissues. Secondly, to identify putative actors in the ECM synthesis to be functionally studied in the future.


Subject(s)
Phaeophyceae , Phaeophyceae/genetics , Phaeophyceae/chemistry , Phaeophyceae/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Genome , Extracellular Matrix/metabolism
12.
Proc Natl Acad Sci U S A ; 119(20): e2123411119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35533274

ABSTRACT

Increases in snack consumption associated with Westernized lifestyles provide an opportunity to introduce nutritious foods into poor diets. We describe two 10-wk-long open label, single group assignment human studies that measured the effects of two snack prototypes containing fiber preparations from two sustainable and scalable sources; the byproducts remaining after isolation of protein from the endosperm of peas and the vesicular pulp remaining after processing oranges for the manufacture of juices. The normal diets of study participants were supplemented with either a pea- or orange fiber-containing snack. We focused our analysis on quantifying the abundances of genes encoding carbohydrate-active enzymes (CAZymes) (glycoside hydrolases and polysaccharide lyases) in the fecal microbiome, mass spectrometric measurements of glycan structures (glycosidic linkages) in feces, plus aptamer-based assessment of levels of 1,300 plasma proteins reflecting a broad range of physiological functions. Computational methods for feature selection identified treatment-discriminatory changes in CAZyme genes that correlated with alterations in levels of fiber-associated glycosidic linkages; these changes in turn correlated with levels of plasma proteins representing diverse biological functions, including transforming growth factor type ß/bone morphogenetic protein-mediated fibrosis, vascular endothelial growth factor-related angiogenesis, P38/MAPK-associated immune cell signaling, and obesity-associated hormonal regulators. The approach used represents a way to connect changes in consumer microbiomes produced by specific fiber types with host responses in the context of varying background diets.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Dietary Fiber/metabolism , Gastrointestinal Microbiome/physiology , Humans , Polysaccharides/metabolism , Proteome
13.
BMC Genomics ; 25(1): 643, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937673

ABSTRACT

BACKGROUND: The CBM13 family comprises carbohydrate-binding modules that occur mainly in enzymes and in several ricin-B lectins. The ricin-B lectin domain resembles the CBM13 module to a large extent. Historically, ricin-B lectins and CBM13 proteins were considered completely distinct, despite their structural and functional similarities. RESULTS: In this data mining study, we investigate structural and functional similarities of these intertwined protein groups. Because of the high structural and functional similarities, and differences in nomenclature usage in several databases, confusion can arise. First, we demonstrate how public protein databases use different nomenclature systems to describe CBM13 modules and putative ricin-B lectin domains. We suggest the introduction of a novel CBM13 domain identifier, as well as the extension of CAZy cross-references in UniProt to guard the distinction between CAZy and non-CAZy entries in public databases. Since similar problems may occur with other lectin families and CBM families, we suggest the introduction of novel CBM InterPro domain identifiers to all existing CBM families. Second, we investigated phylogenetic, nomenclatural and structural similarities between putative ricin-B lectin domains and CBM13 modules, making use of sequence similarity networks. We concluded that the ricin-B/CBM13 superfamily may be larger than initially thought and that several putative ricin-B lectin domains may display CAZyme functionalities, although biochemical proof remains to be delivered. CONCLUSIONS: Ricin-B lectin domains and CBM13 modules are associated groups of proteins whose database semantics are currently biased towards ricin-B lectins. Revision of the CAZy cross-reference in UniProt and introduction of a dedicated CBM13 domain identifier in InterPro may resolve this issue. In addition, our analyses show that several proteins with putative ricin-B lectin domains show very strong structural similarity to CBM13 modules. Therefore ricin-B lectin domains and CBM13 modules could be considered distant members of a larger ricin-B/CBM13 superfamily.


Subject(s)
Lectins , Phylogeny , Protein Domains , Ricin , Ricin/chemistry , Ricin/genetics , Lectins/chemistry , Lectins/genetics , Lectins/metabolism , Databases, Protein , Amino Acid Sequence , Sequence Homology, Amino Acid
14.
Mol Biol Evol ; 40(3)2023 03 04.
Article in English | MEDLINE | ID: mdl-36811946

ABSTRACT

The mutualistic ectomycorrhizal (ECM) fungal genus Pisolithus comprises 19 species defined to date which colonize the roots of >50 hosts worldwide suggesting that substantial genomic and functional evolution occurred during speciation. To better understand this intra-genus variation, we undertook a comparative multi-omic study of nine Pisolithus species sampled from North America, South America, Asia, and Australasia. We found that there was a small core set of genes common to all species (13%), and that these genes were more likely to be significantly regulated during symbiosis with a host than accessory or species-specific genes. Thus, the genetic "toolbox" foundational to the symbiotic lifestyle in this genus is small. Transposable elements were located significantly closer to gene classes including effector-like small secreted proteins (SSPs). Poorly conserved SSPs were more likely to be induced by symbiosis, suggesting that they may be a class of protein that tune host specificity. The Pisolithus gene repertoire is characterized by divergent CAZyme profiles when compared with other fungi, both symbiotic and saprotrophic. This was driven by differences in enzymes associated with symbiotic sugar processing, although metabolomic analysis suggest that neither copy number nor expression of these genes is sufficient to predict sugar capture from a host plant or its metabolism in fungal hyphae. Our results demonstrate that intra-genus genomic and functional diversity within ECM fungi is greater than previously thought, underlining the importance of continued comparative studies within the fungal tree of life to refine our focus on pathways and evolutionary processes foundational to this symbiotic lifestyle.


Subject(s)
Basidiomycota , Mycorrhizae , Mycorrhizae/genetics , Symbiosis/genetics , Basidiomycota/genetics , Plant Roots , Sugars
15.
New Phytol ; 242(4): 1676-1690, 2024 May.
Article in English | MEDLINE | ID: mdl-38148573

ABSTRACT

Soil fungi belonging to different functional guilds, such as saprotrophs, pathogens, and mycorrhizal symbionts, play key roles in forest ecosystems. To date, no study has compared the actual gene expression of these guilds in different forest soils. We used metatranscriptomics to study the competition for organic resources by these fungal groups in boreal, temperate, and Mediterranean forest soils. Using a dedicated mRNA annotation pipeline combined with the JGI MycoCosm database, we compared the transcripts of these three fungal guilds, targeting enzymes involved in C- and N mobilization from plant and microbial cell walls. Genes encoding enzymes involved in the degradation of plant cell walls were expressed at a higher level in saprotrophic fungi than in ectomycorrhizal and pathogenic fungi. However, ectomycorrhizal and saprotrophic fungi showed similarly high expression levels of genes encoding enzymes involved in fungal cell wall degradation. Transcripts for N-related transporters were more highly expressed in ectomycorrhizal fungi than in other groups. We showed that ectomycorrhizal and saprotrophic fungi compete for N in soil organic matter, suggesting that their interactions could decelerate C cycling. Metatranscriptomics provides a unique tool to test controversial ecological hypotheses and to better understand the underlying ecological processes involved in soil functioning and carbon stabilization.


Subject(s)
Forests , Fungi , Soil Microbiology , Transcriptome , Fungi/genetics , Fungi/physiology , Transcriptome/genetics , Mycorrhizae/physiology , Mycorrhizae/genetics , Gene Expression Profiling , Gene Expression Regulation, Fungal , Nitrogen/metabolism , Soil/chemistry , Ecosystem , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Nucleic Acids Res ; 50(D1): D571-D577, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34850161

ABSTRACT

Thirty years have elapsed since the emergence of the classification of carbohydrate-active enzymes in sequence-based families that became the CAZy database over 20 years ago, freely available for browsing and download at www.cazy.org. In the era of large scale sequencing and high-throughput Biology, it is important to examine the position of this specialist database that is deeply rooted in human curation. The three primary tasks of the CAZy curators are (i) to maintain and update the family classification of this class of enzymes, (ii) to classify sequences newly released by GenBank and the Protein Data Bank and (iii) to capture and present functional information for each family. The CAZy website is updated once a month. Here we briefly summarize the increase in novel families and the annotations conducted during the last 8 years. We present several important changes that facilitate taxonomic navigation, and allow to download the entirety of the annotations. Most importantly we highlight the considerable amount of work that accompanies the analysis and report of biochemical data from the literature.


Subject(s)
Carbohydrates/chemistry , Databases, Nucleic Acid , Databases, Protein , Enzymes/chemistry , Carbohydrates/classification , Enzyme Activation/genetics , Enzymes/classification , Humans
17.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34001614

ABSTRACT

The concept that gut microbiome-expressed functions regulate ponderal growth has important implications for infant and child health, as well as animal health. Using an intergenerational pig model of diet restriction (DR) that produces reduced weight gain, we developed a feature-selection algorithm to identify representative characteristics distinguishing DR fecal microbiomes from those of full-fed (FF) pigs as both groups consumed a common sequence of diets during their growth cycle. Gnotobiotic mice were then colonized with DR and FF microbiomes and subjected to controlled feeding with a pig diet. DR microbiomes have reduced representation of genes that degrade dominant components of late growth-phase diets, exhibit reduced production of butyrate, a key host-accessible energy source, and are causally linked to reduced hepatic fatty acid metabolism (ß-oxidation) and the selection of alternative energy substrates. The approach described could aid in the development of guidelines for microbiome stewardship in diverse species, including farm animals, in order to support their healthy growth.


Subject(s)
Butyrates/metabolism , Gastrointestinal Microbiome/physiology , Lipid Metabolism/physiology , Malnutrition/metabolism , Phosphoric Monoester Hydrolases/metabolism , alpha-Glucosidases/metabolism , Algorithms , Animals , Body Weight , Diet/methods , Diet Therapy/methods , Disease Models, Animal , Feces/microbiology , Germ-Free Life , Liver/metabolism , Male , Malnutrition/physiopathology , Mice , Mice, Inbred C57BL , Starch/metabolism , Sucrose/metabolism , Swine , Taurocholic Acid/metabolism
18.
BMC Biol ; 21(1): 99, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37143068

ABSTRACT

BACKGROUND: Diplonemid flagellates are among the most abundant and species-rich of known marine microeukaryotes, colonizing all habitats, depths, and geographic regions of the world ocean. However, little is known about their genomes, biology, and ecological role. RESULTS: We present the first nuclear genome sequence from a diplonemid, the type species Diplonema papillatum. The ~ 280-Mb genome assembly contains about 32,000 protein-coding genes, likely co-transcribed in groups of up to 100. Gene clusters are separated by long repetitive regions that include numerous transposable elements, which also reside within introns. Analysis of gene-family evolution reveals that the last common diplonemid ancestor underwent considerable metabolic expansion. D. papillatum-specific gains of carbohydrate-degradation capability were apparently acquired via horizontal gene transfer. The predicted breakdown of polysaccharides including pectin and xylan is at odds with reports of peptides being the predominant carbon source of this organism. Secretome analysis together with feeding experiments suggest that D. papillatum is predatory, able to degrade cell walls of live microeukaryotes, macroalgae, and water plants, not only for protoplast feeding but also for metabolizing cell-wall carbohydrates as an energy source. The analysis of environmental barcode samples shows that D. papillatum is confined to temperate coastal waters, presumably acting in bioremediation of eutrophication. CONCLUSIONS: Nuclear genome information will allow systematic functional and cell-biology studies in D. papillatum. It will also serve as a reference for the highly diverse diplonemids and provide a point of comparison for studying gene complement evolution in the sister group of Kinetoplastida, including human-pathogenic taxa.


Subject(s)
Eukaryota , Kinetoplastida , Humans , Eukaryota/genetics , Meiotic Prophase I , Euglenozoa/genetics , Kinetoplastida/genetics , Multigene Family , Phylogeny
20.
Nature ; 544(7648): 65-70, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28329766

ABSTRACT

The metabolism of carbohydrate polymers drives microbial diversity in the human gut microbiota. It is unclear, however, whether bacterial consortia or single organisms are required to depolymerize highly complex glycans. Here we show that the gut bacterium Bacteroides thetaiotaomicron uses the most structurally complex glycan known: the plant pectic polysaccharide rhamnogalacturonan-II, cleaving all but 1 of its 21 distinct glycosidic linkages. The deconstruction of rhamnogalacturonan-II side chains and backbone are coordinated to overcome steric constraints, and the degradation involves previously undiscovered enzyme families and catalytic activities. The degradation system informs revision of the current structural model of rhamnogalacturonan-II and highlights how individual gut bacteria orchestrate manifold enzymes to metabolize the most challenging glycan in the human diet.


Subject(s)
Bacteroides thetaiotaomicron/enzymology , Bacteroides thetaiotaomicron/metabolism , Biocatalysis , Gastrointestinal Tract/microbiology , Glycoside Hydrolases/metabolism , Pectins/chemistry , Pectins/metabolism , Bacteroides thetaiotaomicron/growth & development , Borates/chemistry , Borates/metabolism , Catalytic Domain , Gastrointestinal Microbiome , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/classification , Humans , Models, Molecular , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL