Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Circulation ; 148(6): 459-472, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37435755

ABSTRACT

BACKGROUND: Gut microbiota have been implicated in atherosclerotic disease, but their relation with subclinical coronary atherosclerosis is unclear. This study aimed to identify associations between the gut microbiome and computed tomography-based measures of coronary atherosclerosis and to explore relevant clinical correlates. METHODS: We conducted a cross-sectional study of 8973 participants (50 to 65 years of age) without overt atherosclerotic disease from the population-based SCAPIS (Swedish Cardiopulmonary Bioimage Study). Coronary atherosclerosis was measured using coronary artery calcium score and coronary computed tomography angiography. Gut microbiota species abundance and functional potential were assessed with shotgun metagenomics sequencing of fecal samples, and associations with coronary atherosclerosis were evaluated with multivariable regression models adjusted for cardiovascular risk factors. Associated species were evaluated for association with inflammatory markers, metabolites, and corresponding species in saliva. RESULTS: The mean age of the study sample was 57.4 years, and 53.7% were female. Coronary artery calcification was detected in 40.3%, and 5.4% had at least 1 stenosis with >50% occlusion. Sixty-four species were associated with coronary artery calcium score independent of cardiovascular risk factors, with the strongest associations observed for Streptococcus anginosus and Streptococcus oralis subsp oralis (P<1×10-5). Associations were largely similar across coronary computed tomography angiography-based measurements. Out of the 64 species, 19 species, including streptococci and other species commonly found in the oral cavity, were associated with high-sensitivity C-reactive protein plasma concentrations, and 16 with neutrophil counts. Gut microbial species that are commonly found in the oral cavity were negatively associated with plasma indole propionate and positively associated with plasma secondary bile acids and imidazole propionate. Five species, including 3 streptococci, correlated with the same species in saliva and were associated with worse dental health in the Malmö Offspring Dental Study. Microbial functional potential of dissimilatory nitrate reduction, anaerobic fatty acid ß-oxidation, and amino acid degradation were associated with coronary artery calcium score. CONCLUSIONS: This study provides evidence of an association of a gut microbiota composition characterized by increased abundance of Streptococcus spp and other species commonly found in the oral cavity with coronary atherosclerosis and systemic inflammation markers. Further longitudinal and experimental studies are warranted to explore the potential implications of a bacterial component in atherogenesis.


Subject(s)
Atherosclerosis , Coronary Artery Disease , Humans , Female , Middle Aged , Male , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/epidemiology , Cross-Sectional Studies , Calcium , Atherosclerosis/epidemiology , Streptococcus
3.
Nat Commun ; 13(1): 5370, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36151114

ABSTRACT

Human gut microbiota produce a variety of molecules, some of which enter the bloodstream and impact health. Conversely, dietary or pharmacological compounds may affect the microbiota before entering the circulation. Characterization of these interactions is an important step towards understanding the effects of the gut microbiota on health. In this cross-sectional study, we used deep metagenomic sequencing and ultra-high-performance liquid chromatography linked to mass spectrometry for a detailed characterization of the gut microbiota and plasma metabolome, respectively, of 8583 participants invited at age 50 to 64 from the population-based Swedish CArdioPulmonary bioImage Study. Here, we find that the gut microbiota explain up to 58% of the variance of individual plasma metabolites and we present 997 associations between alpha diversity and plasma metabolites and 546,819 associations between specific gut metagenomic species and plasma metabolites in an online atlas ( https://gutsyatlas.serve.scilifelab.se/ ). We exemplify the potential of this resource by presenting novel associations between dietary factors and oral medication with the gut microbiome, and microbial species strongly associated with the uremic toxin p-cresol sulfate. This resource can be used as the basis for targeted studies of perturbation of specific metabolites and for identification of candidate plasma biomarkers of gut microbiota composition.


Subject(s)
Gastrointestinal Microbiome , Biomarkers , Cross-Sectional Studies , Gastrointestinal Microbiome/genetics , Humans , Metabolome , Metabolomics/methods , Middle Aged , Uremic Toxins
4.
Sci Rep ; 11(1): 13252, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168163

ABSTRACT

Knowledge about in vivo effects of human circulating C-6 hydroxylated bile acids (BAs), also called muricholic acids, is sparse. It is unsettled if the gut microbiome might contribute to their biosynthesis. Here, we measured a range of serum BAs and related them to markers of human metabolic health and the gut microbiome. We examined 283 non-obese and obese Danish adults from the MetaHit study. Fasting concentrations of serum BAs were quantified using ultra-performance liquid chromatography-tandem mass-spectrometry. The gut microbiome was characterized with shotgun metagenomic sequencing and genome-scale metabolic modeling. We find that tauro- and glycohyocholic acid correlated inversely with body mass index (P = 4.1e-03, P = 1.9e-05, respectively), waist circumference (P = 0.017, P = 1.1e-04, respectively), body fat percentage (P = 2.5e-03, P = 2.3e-06, respectively), insulin resistance (P = 0.051, P = 4.6e-4, respectively), fasting concentrations of triglycerides (P = 0.06, P = 9.2e-4, respectively) and leptin (P = 0.067, P = 9.2e-4). Tauro- and glycohyocholic acids, and tauro-a-muricholic acid were directly linked with a distinct gut microbial community primarily composed of Clostridia species (P = 0.037, P = 0.013, P = 0.027, respectively). We conclude that serum conjugated C-6-hydroxylated BAs associate with measures of human metabolic health and gut communities of Clostridia species. The findings merit preclinical interventions and human feasibility studies to explore the therapeutic potential of these BAs in obesity and type 2 diabetes.


Subject(s)
Bile Acids and Salts/blood , Clostridium/metabolism , Gastrointestinal Microbiome , Adiposity , Body Mass Index , Cholic Acids/blood , Chromatography, High Pressure Liquid , Clostridium/genetics , Deoxycholic Acid/blood , Female , Gastrointestinal Microbiome/genetics , Humans , Logistic Models , Male , Metagenomics , Middle Aged , Obesity/blood , Obesity/microbiology , Tandem Mass Spectrometry , Taurocholic Acid/blood , Waist Circumference
5.
Gastroenterology ; 160(7): 2423-2434.e5, 2021 06.
Article in English | MEDLINE | ID: mdl-33662387

ABSTRACT

BACKGROUND & AIMS: IgA exerts its primary function at mucosal surfaces, where it binds microbial antigens to regulate bacterial growth and epithelial attachment. One third of individuals with IgA deficiency (IgAD) suffers from recurrent mucosal infections, possibly related to an altered microbiota. We aimed to delineate the impact of IgAD and the IgA-autoantibody status on the composition and functional capacity of the gut microbiota. METHODS: We performed a paired, lifestyle-balanced analysis of the effect of IgA on the gut microbiota composition and functionality based on fecal samples from individuals with IgAD and IgA-sufficient household members (n = 100), involving quantitative shotgun metagenomics, species-centric functional annotation of gut bacteria, and strain-level analyses. We supplemented the data set with 32 individuals with IgAD and examined the influence of IgA-autoantibody status on the composition and functionality of the gut microbiota. RESULTS: The gut microbiota of individuals with IgAD exhibited decreased richness and diversity and was enriched for bacterial species encoding pathogen-related functions including multidrug and antimicrobial peptide resistance, virulence factors, and type III and VI secretion systems. These functional changes were largely attributed to Escherichia coli but were independent of E coli strain variations and most prominent in individuals with IgAD with IgA-specific autoreactive antibodies. CONCLUSIONS: The microbiota of individuals with IgAD is enriched for species holding increased proinflammatory potential, thereby potentially decreasing the resistance to gut barrier-perturbing events. This phenotype is especially pronounced in individuals with IgAD with IgA-specific autoreactive antibodies, thus warranting a screening for IgA-specific autoreactive antibodies in IgAD to identify patients with IgAD with increased risk for gastrointestinal implications.


Subject(s)
Autoantibodies/metabolism , Gastrointestinal Microbiome/immunology , IgA Deficiency/immunology , IgA Deficiency/microbiology , Immunoglobulin A/metabolism , Adult , Aged , Case-Control Studies , Feces/microbiology , Female , Humans , Male , Middle Aged
6.
PLoS One ; 15(9): e0238648, 2020.
Article in English | MEDLINE | ID: mdl-32947608

ABSTRACT

Elevated postprandial plasma glucose is a risk factor for development of type 2 diabetes and cardiovascular disease. We hypothesized that the inter-individual postprandial plasma glucose response varies partly depending on the intestinal microbiome composition and function. We analyzed data from Danish adults (n = 106), who were self-reported healthy and attended the baseline visit of two previously reported randomized controlled cross-over trials within the Gut, Grain and Greens project. Plasma glucose concentrations at five time points were measured before and during three hours after a standardized breakfast. Based on these data, we devised machine learning algorithms integrating bio-clinical, as well as shotgun-sequencing-derived taxa and functional potentials of the intestinal microbiome to predict individual postprandial glucose excursions. In this post hoc study, we found microbial and clinical features, which predicted up to 48% of the inter-individual variance of postprandial plasma glucose responses (Pearson correlation coefficient of measured vs. predicted values, R = 0.69, 95% CI: 0.45 to 0.84, p<0.001). The features were age, fasting serum triglycerides, systolic blood pressure, BMI, fasting total serum cholesterol, abundance of Bifidobacterium genus, richness of metagenomics species and abundance of a metagenomic species annotated to Clostridiales at order level. A model based only on microbial features predicted up to 14% of the variance in postprandial plasma glucose excursions (R = 0.37, 95% CI: 0.02 to 0.64, p = 0.04). Adding fasting glycaemic measures to the model including microbial and bio-clinical features increased the predictive power to R = 0.78 (95% CI: 0.59 to 0.89, p<0.001), explaining more than 60% of the inter-individual variance of postprandial plasma glucose concentrations. The outcome of the study points to a potential role of the taxa and functional potentials of the intestinal microbiome. If validated in larger studies our findings may be included in future algorithms attempting to develop personalized nutrition, especially for prediction of individual blood glucose excursions in dys-glycaemic individuals.


Subject(s)
Blood Glucose/metabolism , Gastrointestinal Microbiome , Postprandial Period , Algorithms , Fasting/blood , Female , Humans , Life Style , Male , Middle Aged , Models, Biological , Phenomics
8.
Nat Commun ; 9(1): 4630, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30425247

ABSTRACT

Adherence to a low-gluten diet has become increasingly common in parts of the general population. However, the effects of reducing gluten-rich food items including wheat, barley and rye cereals in healthy adults are unclear. Here, we undertook a randomised, controlled, cross-over trial involving 60 middle-aged Danish adults without known disorders with two 8-week interventions comparing a low-gluten diet (2 g gluten per day) and a high-gluten diet (18 g gluten per day), separated by a washout period of at least six weeks with habitual diet (12 g gluten per day). We find that, in comparison with a high-gluten diet, a low-gluten diet induces moderate changes in the intestinal microbiome, reduces fasting and postprandial hydrogen exhalation, and leads to improvements in self-reported bloating. These observations suggest that most of the effects of a low-gluten diet in non-coeliac adults may be driven by qualitative changes in dietary fibres.


Subject(s)
Diet , Gastrointestinal Microbiome , Glutens/administration & dosage , Glutens/adverse effects , Adult , Aged , Body Mass Index , Creatinine/urine , Cross-Over Studies , Cytokines/blood , DNA, Bacterial/analysis , Denmark , Fasting , Feces/microbiology , Female , Fermentation , Gastrointestinal Microbiome/genetics , Humans , Hydrogen , Intestines/microbiology , Male , Metabolomics , Metagenomics , Middle Aged , Postprandial Period , Self Report , Young Adult
10.
Mol Cell Endocrinol ; 444: 9-18, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28131743

ABSTRACT

Specific inbred strains and transgenic inhibin-α Simian Virus 40 T antigen (inhα/Tag) mice are genetically susceptible to gonadectomy-induced adrenocortical neoplasias. We identified altered gene expression in prepubertally gonadectomized (GDX) inhα/Tag and wild-type (WT) mice. Besides earlier reported Gata4 and Lhcgr, we found up-regulated Esr1, Prlr-rs1, and down-regulated Grb10, Mmp24, Sgcd, Rerg, Gnas, Nfatc2, Gnrhr, Igf2 in inhα/Tag adrenal tumors. Sex-steroidogenic enzyme genes expression (Srd5a1, Cyp19a1) was up-regulated in tumors, but adrenal-specific steroidogenic enzyme (Cyp21a1, Cyp11b1, Cyp11b2) down-regulated. We localized novel Lhcgr transcripts in adrenal cortex parenchyma and in non-steroidogenic A cells, in GDX WT and in intact WT mice. We identified up-regulated Esr1 as a potential novel biomarker of gonadectomy-induced adrenocortical tumors in inhα/Tag mice presenting with an inverted adrenal-to-gonadal steroidogenic gene expression profile. A putative normal adrenal remodeling or tumor suppressor role of the down-regulated genes (e.g. Grb10, Rerg, Gnas, and Nfatc2) in the tumors remains to be addressed.


Subject(s)
Adrenal Gland Neoplasms/genetics , Genes, Neoplasm , Gonadotropins/pharmacology , Animals , Biomarkers, Tumor/metabolism , GATA Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Ontology , Male , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Steroids/biosynthesis
11.
Nat Microbiol ; 1(11): 16152, 2016 Aug 26.
Article in English | MEDLINE | ID: mdl-27564131

ABSTRACT

The human gastrointestinal (GI) tract is the habitat for hundreds of microbial species, of which many cannot be cultivated readily, presumably because of the dependencies between species1. Studies of microbial co-occurrence in the gut have indicated community substructures that may reflect functional and metabolic interactions between cohabiting species2,3. To move beyond species co-occurrence networks, we systematically identified transcriptional interactions between pairs of coexisting gut microbes using metagenomics and microarray-based metatranscriptomics data from 233 stool samples from Europeans. In 102 significantly interacting species pairs, the transcriptional changes led to a reduced expression of orthologous functions between the coexisting species. Specific species-species transcriptional interactions were enriched for functions important for H2 and CO2 homeostasis, butyrate biosynthesis, ATP-binding cassette (ABC) transporters, flagella assembly and bacterial chemotaxis, as well as for the metabolism of carbohydrates, amino acids and cofactors. The analysis gives the first insight into the microbial community-wide transcriptional interactions, and suggests that the regulation of gene expression plays an important role in species adaptation to coexistence and that niche segregation takes place at the transcriptional level.


Subject(s)
Gastrointestinal Microbiome/genetics , Gene Expression Profiling , Metagenome , Microbial Interactions , ATP-Binding Cassette Transporters/genetics , Bifidobacterium bifidum/genetics , Bifidobacterium bifidum/metabolism , Butyrates/metabolism , Carbon Dioxide/metabolism , Denmark , Feces/microbiology , Gastrointestinal Microbiome/physiology , Humans , Metabolic Networks and Pathways/genetics , Microbial Interactions/genetics , Microbial Interactions/physiology , Spain , Systems Analysis
12.
Nat Microbiol ; 1(9): 16093, 2016 Jun 27.
Article in English | MEDLINE | ID: mdl-27562254

ABSTRACT

Little is known about how colonic transit time relates to human colonic metabolism and its importance for host health, although a firm stool consistency, a proxy for a long colonic transit time, has recently been positively associated with gut microbial richness. Here, we show that colonic transit time in humans, assessed using radio-opaque markers, is associated with overall gut microbial composition, diversity and metabolism. We find that a long colonic transit time associates with high microbial richness and is accompanied by a shift in colonic metabolism from carbohydrate fermentation to protein catabolism as reflected by higher urinary levels of potentially deleterious protein-derived metabolites. Additionally, shorter colonic transit time correlates with metabolites possibly reflecting increased renewal of the colonic mucosa. Together, this suggests that a high gut microbial richness does not per se imply a healthy gut microbial ecosystem and points at colonic transit time as a highly important factor to consider in microbiome and metabolomics studies.


Subject(s)
Gastrointestinal Microbiome , Gastrointestinal Transit , Metabolome , Adult , Aged , Biomarkers/metabolism , Carbohydrate Metabolism , Colon/metabolism , Feces/microbiology , Female , Fermentation , Humans , Male , Metabolism , Middle Aged , Mucous Membrane/metabolism , Proteins/metabolism , Reproducibility of Results , Young Adult
13.
Nature ; 528(7581): 262-266, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26633628

ABSTRACT

In recent years, several associations between common chronic human disorders and altered gut microbiome composition and function have been reported. In most of these reports, treatment regimens were not controlled for and conclusions could thus be confounded by the effects of various drugs on the microbiota, which may obscure microbial causes, protective factors or diagnostically relevant signals. Our study addresses disease and drug signatures in the human gut microbiome of type 2 diabetes mellitus (T2D). Two previous quantitative gut metagenomics studies of T2D patients that were unstratified for treatment yielded divergent conclusions regarding its associated gut microbial dysbiosis. Here we show, using 784 available human gut metagenomes, how antidiabetic medication confounds these results, and analyse in detail the effects of the most widely used antidiabetic drug metformin. We provide support for microbial mediation of the therapeutic effects of metformin through short-chain fatty acid production, as well as for potential microbiota-mediated mechanisms behind known intestinal adverse effects in the form of a relative increase in abundance of Escherichia species. Controlling for metformin treatment, we report a unified signature of gut microbiome shifts in T2D with a depletion of butyrate-producing taxa. These in turn cause functional microbiome shifts, in part alleviated by metformin-induced changes. Overall, the present study emphasizes the need to disentangle gut microbiota signatures of specific human diseases from those of medication.


Subject(s)
Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Metformin/pharmacology , Biodiversity , Diabetes Mellitus, Type 2/drug therapy , Female , Gastrointestinal Microbiome/genetics , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Metagenome/drug effects , Metagenome/physiology , Metformin/therapeutic use , RNA, Ribosomal, 16S/genetics
14.
Nat Biotechnol ; 33(10): 1103-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26414350

ABSTRACT

We established a catalog of the mouse gut metagenome comprising ∼2.6 million nonredundant genes by sequencing DNA from fecal samples of 184 mice. To secure high microbiome diversity, we used mouse strains of diverse genetic backgrounds, from different providers, kept in different housing laboratories and fed either a low-fat or high-fat diet. Similar to the human gut microbiome, >99% of the cataloged genes are bacterial. We identified 541 metagenomic species and defined a core set of 26 metagenomic species found in 95% of the mice. The mouse gut microbiome is functionally similar to its human counterpart, with 95.2% of its Kyoto Encyclopedia of Genes and Genomes (KEGG) orthologous groups in common. However, only 4.0% of the mouse gut microbial genes were shared (95% identity, 90% coverage) with those of the human gut microbiome. This catalog provides a useful reference for future studies.


Subject(s)
Bacteria/genetics , Chromosome Mapping/methods , Databases, Genetic , Genome, Bacterial/genetics , Intestines/microbiology , Microbiota/genetics , Animals , Bacterial Proteins/genetics , Catalogs as Topic , Humans , Intestinal Mucosa/metabolism , Species Specificity
15.
Nat Biotechnol ; 32(8): 822-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997787

ABSTRACT

Most current approaches for analyzing metagenomic data rely on comparisons to reference genomes, but the microbial diversity of many environments extends far beyond what is covered by reference databases. De novo segregation of complex metagenomic data into specific biological entities, such as particular bacterial strains or viruses, remains a largely unsolved problem. Here we present a method, based on binning co-abundant genes across a series of metagenomic samples, that enables comprehensive discovery of new microbial organisms, viruses and co-inherited genetic entities and aids assembly of microbial genomes without the need for reference sequences. We demonstrate the method on data from 396 human gut microbiome samples and identify 7,381 co-abundance gene groups (CAGs), including 741 metagenomic species (MGS). We use these to assemble 238 high-quality microbial genomes and identify affiliations between MGS and hundreds of viruses or genetic entities. Our method provides the means for comprehensive profiling of the diversity within complex metagenomic samples.


Subject(s)
Metagenomics , Cluster Analysis , Databases, Genetic
16.
Nature ; 473(7346): 174-80, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21508958

ABSTRACT

Our knowledge of species and functional composition of the human gut microbiome is rapidly increasing, but it is still based on very few cohorts and little is known about variation across the world. By combining 22 newly sequenced faecal metagenomes of individuals from four countries with previously published data sets, here we identify three robust clusters (referred to as enterotypes hereafter) that are not nation or continent specific. We also confirmed the enterotypes in two published, larger cohorts, indicating that intestinal microbiota variation is generally stratified, not continuous. This indicates further the existence of a limited number of well-balanced host-microbial symbiotic states that might respond differently to diet and drug intake. The enterotypes are mostly driven by species composition, but abundant molecular functions are not necessarily provided by abundant species, highlighting the importance of a functional analysis to understand microbial communities. Although individual host properties such as body mass index, age, or gender cannot explain the observed enterotypes, data-driven marker genes or functional modules can be identified for each of these host properties. For example, twelve genes significantly correlate with age and three functional modules with the body mass index, hinting at a diagnostic potential of microbial markers.


Subject(s)
Bacteria/classification , Intestines/microbiology , Metagenome , Bacteria/genetics , Bacterial Typing Techniques , Biodiversity , Biomarkers/analysis , Europe , Feces/microbiology , Female , Humans , Male , Metagenomics , Phylogeny
17.
PLoS Pathog ; 6(10): e1001137, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20949080

ABSTRACT

Certain pathogens deliver effectors into plant cells to modify host protein targets and thereby suppress immunity. These target modifications can be detected by intracellular immune receptors, or Resistance (R) proteins, that trigger strong immune responses including localized host cell death. The accelerated cell death 11 (acd11) "lesion mimic" mutant of Arabidopsis thaliana exhibits autoimmune phenotypes such as constitutive defense responses and cell death without pathogen perception. ACD11 encodes a putative sphingosine transfer protein, but its precise role during these processes is unknown. In a screen for lazarus (laz) mutants that suppress acd11 death we identified two genes, LAZ2 and LAZ5. LAZ2 encodes the histone lysine methyltransferase SDG8, previously shown to epigenetically regulate flowering time via modification of histone 3 (H3). LAZ5 encodes an RPS4-like R-protein, defined by several dominant negative alleles. Microarray and chromatin immunoprecipitation analyses showed that LAZ2/SDG8 is required for LAZ5 expression and H3 lysine 36 trimethylation at LAZ5 chromatin to maintain a transcriptionally active state. We hypothesize that LAZ5 triggers cell death in the absence of ACD11, and that cell death in other lesion mimic mutants may also be caused by inappropriate activation of R genes. Moreover, SDG8 is required for basal and R protein-mediated pathogen resistance in Arabidopsis, revealing the importance of chromatin remodeling as a key process in plant innate immunity.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Arabidopsis Proteins/genetics , Arabidopsis/immunology , Epigenesis, Genetic/physiology , Membrane Transport Proteins/genetics , Receptors, Immunologic/genetics , Apoptosis Regulatory Proteins/physiology , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/physiology , Arabidopsis Proteins/physiology , Autoimmunity/physiology , Cell Death/genetics , Cell Death/immunology , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/immunology , Chromatin Assembly and Disassembly/physiology , Epigenesis, Genetic/immunology , Gene Expression Regulation, Plant/physiology , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/physiology , Immunity, Innate/genetics , Membrane Transport Proteins/physiology , Receptors, Immunologic/physiology
18.
PLoS One ; 5(9): e12586, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20830211

ABSTRACT

BACKGROUND: Programmed cell death (PCD) is a necessary part of the life of multi-cellular organisms. A type of plant PCD is the defensive hypersensitive response (HR) elicited via recognition of a pathogen by host resistance (R) proteins. The lethal, recessive accelerated cell death 11 (acd11) mutant exhibits HR-like accelerated cell death, and cell death execution in acd11 shares genetic requirements for HR execution triggered by one subclass of R proteins. METHODOLOGY/PRINCIPAL FINDINGS: To identify genes required for this PCD pathway, we conducted a genetic screen for suppressors of acd11, here called lazarus (laz) mutants. In addition to known suppressors of R protein-mediated HR, we isolated 13 novel complementation groups of dominant and recessive laz mutants. Here we describe laz1, which encodes a protein with a domain of unknown function (DUF300), and demonstrate that LAZ1 contributes to HR PCD conditioned by the Toll/interleukin-1 (TIR)-type R protein RPS4 and by the coiled-coil (CC)-type R protein RPM1. Using a yeast-based topology assay, we also provide evidence that LAZ1 is a six transmembrane protein with structural similarities to the human tumor suppressor TMEM34. Finally, we demonstrate by transient expression of reporter fusions in protoplasts that localization of LAZ1 is distributed between the cytosol, the plasma membrane and FM4-64 stained vesicles. CONCLUSIONS/SIGNIFICANCE: Our findings indicate that LAZ1 functions as a regulator or effector of plant PCD associated with the HR, in addition to its role in acd11-related death. Furthermore, the similar topology of a plant and human DUF300 proteins suggests similar functions in PCD across the eukaryotic kingdoms, although a direct role for TMEM34 in cell death control remains to be established. Finally, the subcellular localization pattern of LAZ1 suggests that it may have transport functions for yet unknown, death-related signaling molecules at the plasma membrane and/or endosomal compartments. In summary, our results validate the utility of the large-scale suppressor screen to identify novel components with functions in plant PCD, which may also have implications for deciphering cell death mechanisms in other organisms.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Arabidopsis Proteins/immunology , Arabidopsis Proteins/metabolism , Arabidopsis/immunology , Membrane Transport Proteins/metabolism , Plant Diseases/immunology , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/microbiology , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Cytosol/chemistry , Cytosol/metabolism , Host-Pathogen Interactions , Membrane Transport Proteins/genetics , Plant Diseases/microbiology , Protein Structure, Tertiary , Protein Transport , Pseudomonas syringae/immunology , Pseudomonas syringae/physiology
19.
Nature ; 464(7285): 59-65, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20203603

ABSTRACT

To understand the impact of gut microbes on human health and well-being it is crucial to assess their genetic potential. Here we describe the Illumina-based metagenomic sequencing, assembly and characterization of 3.3 million non-redundant microbial genes, derived from 576.7 gigabases of sequence, from faecal samples of 124 European individuals. The gene set, approximately 150 times larger than the human gene complement, contains an overwhelming majority of the prevalent (more frequent) microbial genes of the cohort and probably includes a large proportion of the prevalent human intestinal microbial genes. The genes are largely shared among individuals of the cohort. Over 99% of the genes are bacterial, indicating that the entire cohort harbours between 1,000 and 1,150 prevalent bacterial species and each individual at least 160 such species, which are also largely shared. We define and describe the minimal gut metagenome and the minimal gut bacterial genome in terms of functions present in all individuals and most bacteria, respectively.


Subject(s)
Gastrointestinal Tract/microbiology , Genomics , Metagenome/genetics , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Cohort Studies , Contig Mapping , Denmark , Feces/microbiology , Genes, Bacterial/genetics , Genes, Essential/genetics , Genome, Bacterial/genetics , Health , Humans , Inflammatory Bowel Diseases/genetics , Obesity/genetics , Open Reading Frames/genetics , Overweight/genetics , Sequence Analysis, DNA , Spain
20.
Mol Ecol ; 18(14): 3062-75, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19538339

ABSTRACT

We investigated variation in transcript abundance in the scleractinian coral, Acropora millepora, within and between populations characteristically exposed to different turbidity regimes and hence different levels of light and suspended particulate matter. We examined phenotypic plasticity by comparing levels of gene expression between source populations and following 10 days of acclimatization to a laboratory environment. Analyses of variance revealed that 0.05% of genes were differentially expressed between source populations, 1.32% following translocation into a common laboratory and 0.07% in the interaction (source population-dependent responses to translocation). Functional analyses identified an over-representation of differentially expressed genes associated with metabolism and fluorescence categories (primarily downregulated), and environmental information processing (primarily upregulated) following translocation to a lower light and turbidity environment. Such metabolic downregulation may indicate nonoxidative stress, hibernation or caloric restriction associated with the changed environmental conditions. Green fluorescent protein-related genes were the most differentially expressed and were exclusively downregulated; however, green fluorescent protein levels remained unchanged following translocation. Photophysiological responses of corals from both locations were characterized by a decline when introduced to the common laboratory environment but remained healthy (F(v)/F(m) > 0.6). Declines in total lipid content following translocation were the greatest for inshore corals, suggesting that turbid water corals have a strong reliance on heterotrophic feeding.


Subject(s)
Anthozoa/genetics , Environment , Gene Expression Profiling , Acclimatization/genetics , Animals , Anthozoa/physiology , Anthozoa/radiation effects , Cluster Analysis , Gene Expression Regulation , Genetics, Population , Green Fluorescent Proteins/genetics , Light , Oligonucleotide Array Sequence Analysis , Phenotype , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...