Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
1.
Cell Host Microbe ; 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38917808

RÉSUMÉ

Gut bacteria are implicated in inflammatory bowel disease (IBD), but the strains driving these associations are unknown. Large-scale studies of microbiome evolution could reveal the imprint of disease on gut bacteria, thus pinpointing the strains and genes that may underlie inflammation. Here, we use stool metagenomes of thousands of IBD patients and healthy controls to reconstruct 140,000 strain genotypes, revealing hundreds of lineages enriched in IBD. We demonstrate that these strains are ancient, taxonomically diverse, and ubiquitous in humans. Moreover, disease-associated strains outcompete their healthy counterparts during inflammation, implying long-term adaptation to disease. Strain genetic differences map onto known axes of inflammation, including oxidative stress, nutrient biosynthesis, and immune evasion. Lastly, the loss of health-associated strains of Eggerthella lenta was predictive of fecal calprotectin, a biomarker of disease severity. Our work identifies reservoirs of strain diversity that may impact inflammatory disease and can be extended to other microbiome-associated diseases.

2.
Nat Immunol ; 25(2): 330-342, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38172260

RÉSUMÉ

Antibody-secreting plasma cells (PCs) are generated in secondary lymphoid organs but are reported to reside in an emerging range of anatomical sites. Analysis of the transcriptome of different tissue-resident (Tr)PC populations revealed that they each have their own transcriptional signature indicative of functional adaptation to the host tissue environment. In contrast to expectation, all TrPCs were extremely long-lived, regardless of their organ of residence, with longevity influenced by intrinsic factors like the immunoglobulin isotype. Analysis at single-cell resolution revealed that the bone marrow is unique in housing a compendium of PCs generated all over the body that retain aspects of the transcriptional program indicative of their tissue of origin. This study reveals that extreme longevity is an intrinsic property of TrPCs whose transcriptome is imprinted by signals received both at the site of induction and within the tissue of residence.


Sujet(s)
Moelle osseuse , Plasmocytes , Cellules de la moelle osseuse
3.
Nat Genet ; 55(8): 1267-1276, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37443254

RÉSUMÉ

Genome-wide association studies (GWASs) are a valuable tool for understanding the biology of complex human traits and diseases, but associated variants rarely point directly to causal genes. In the present study, we introduce a new method, polygenic priority score (PoPS), that learns trait-relevant gene features, such as cell-type-specific expression, to prioritize genes at GWAS loci. Using a large evaluation set of genes with fine-mapped coding variants, we show that PoPS and the closest gene individually outperform other gene prioritization methods, but observe the best overall performance by combining PoPS with orthogonal methods. Using this combined approach, we prioritize 10,642 unique gene-trait pairs across 113 complex traits and diseases with high precision, finding not only well-established gene-trait relationships but nominating new genes at unresolved loci, such as LGR4 for estimated glomerular filtration rate and CCR7 for deep vein thrombosis. Overall, we demonstrate that PoPS provides a powerful addition to the gene prioritization toolbox.


Sujet(s)
Hérédité multifactorielle , Locus de caractère quantitatif , Humains , Hérédité multifactorielle/génétique , Locus de caractère quantitatif/génétique , Étude d'association pangénomique/méthodes , Prédisposition génétique à une maladie/génétique , Phénotype , Polymorphisme de nucléotide simple/génétique
4.
Cell ; 186(4): 877-891.e14, 2023 02 16.
Article de Anglais | MEDLINE | ID: mdl-36708705

RÉSUMÉ

We introduce BacDrop, a highly scalable technology for bacterial single-cell RNA sequencing that has overcome many challenges hindering the development of scRNA-seq in bacteria. BacDrop can be applied to thousands to millions of cells from both gram-negative and gram-positive species. It features universal ribosomal RNA depletion and combinatorial barcodes that enable multiplexing and massively parallel sequencing. We applied BacDrop to study Klebsiella pneumoniae clinical isolates and to elucidate their heterogeneous responses to antibiotic stress. In an unperturbed population presumed to be homogeneous, we found within-population heterogeneity largely driven by the expression of mobile genetic elements that promote the evolution of antibiotic resistance. Under antibiotic perturbation, BacDrop revealed transcriptionally distinct subpopulations associated with different phenotypic outcomes including antibiotic persistence. BacDrop thus can capture cellular states that cannot be detected by bulk RNA-seq, which will unlock new microbiological insights into bacterial responses to perturbations and larger bacterial communities such as the microbiome.


Sujet(s)
Analyse de profil d'expression de gènes , Analyse de l'expression du gène de la cellule unique , Analyse de séquence d'ARN , RNA-Seq , Bactéries/génétique , Analyse sur cellule unique
5.
Gastroenterology ; 162(3): 859-876, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34780721

RÉSUMÉ

BACKGROUND & AIMS: Monogenic forms of inflammatory bowel disease (IBD) illustrate the essential roles of individual genes in pathways and networks safeguarding immune tolerance and gut homeostasis. METHODS: To build a taxonomy model, we assessed 165 disorders. Genes were prioritized based on penetrance of IBD and disease phenotypes were integrated with multi-omics datasets. Monogenic IBD genes were classified by (1) overlapping syndromic features, (2) response to hematopoietic stem cell transplantation, (3) bulk RNA-sequencing of 32 tissues, (4) single-cell RNA-sequencing of >50 cell subsets from the intestine of healthy individuals and patients with IBD (pediatric and adult), and (5) proteomes of 43 immune subsets. The model was validated by addition of newly identified monogenic IBD defects. As a proof-of-concept, we explore the intersection between immunometabolism and antimicrobial activity for a group of disorders (G6PC3/SLC37A4). RESULTS: Our quantitative integrated taxonomy defines the cellular landscape of monogenic IBD gene expression across 102 genes with high and moderate penetrance (81 in the model set and 21 genes in the validation set). We illustrate distinct cellular networks, highlight expression profiles across understudied cell types (e.g., CD8+ T cells, neutrophils, epithelial subsets, and endothelial cells) and define genotype-phenotype associations (perianal disease and defective antimicrobial activity). We illustrate processes and pathways shared across cellular compartments and phenotypic groups and highlight cellular immunometabolism with mammalian target of rapamycin activation as one of the converging pathways. There is an overlap of genes and enriched cell-specific expression between monogenic and polygenic IBD. CONCLUSION: Our taxonomy integrates genetic, clinical and multi-omic data; providing a basis for genomic diagnostics and testable hypotheses for disease functions and treatment responses.


Sujet(s)
Maladies inflammatoires intestinales/classification , Maladies inflammatoires intestinales/génétique , Âge de début , Antiports/génétique , Cellules cultivées , Classification , Analyse de profil d'expression de gènes , Études d'associations génétiques , Génotype , Glucosephosphatase/génétique , Glucose-6-phosphate/métabolisme , Humains , Maladies inflammatoires intestinales/métabolisme , Macrophages , Métabolomique , Transporteurs de monosaccharides/génétique , Pénétrance , Phénotype , Transduction du signal/génétique
6.
Cell ; 184(18): 4734-4752.e20, 2021 09 02.
Article de Anglais | MEDLINE | ID: mdl-34450029

RÉSUMÉ

Immune responses to cancer are highly variable, with mismatch repair-deficient (MMRd) tumors exhibiting more anti-tumor immunity than mismatch repair-proficient (MMRp) tumors. To understand the rules governing these varied responses, we transcriptionally profiled 371,223 cells from colorectal tumors and adjacent normal tissues of 28 MMRp and 34 MMRd individuals. Analysis of 88 cell subsets and their 204 associated gene expression programs revealed extensive transcriptional and spatial remodeling across tumors. To discover hubs of interacting malignant and immune cells, we identified expression programs in different cell types that co-varied across tumors from affected individuals and used spatial profiling to localize coordinated programs. We discovered a myeloid cell-attracting hub at the tumor-luminal interface associated with tissue damage and an MMRd-enriched immune hub within the tumor, with activated T cells together with malignant and myeloid cells expressing T cell-attracting chemokines. By identifying interacting cellular programs, we reveal the logic underlying spatially organized immune-malignant cell networks.


Sujet(s)
Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Protéines morphogénétiques osseuses/métabolisme , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Compartimentation cellulaire , Lignée cellulaire tumorale , Chimiokines/métabolisme , Études de cohortes , Tumeurs colorectales/génétique , Réparation de mésappariement de l'ADN/génétique , Cellules endothéliales/métabolisme , Régulation de l'expression des gènes tumoraux , Humains , Immunité , Inflammation/anatomopathologie , Monocytes/anatomopathologie , Cellules myéloïdes/anatomopathologie , Granulocytes neutrophiles/anatomopathologie , Cellules stromales/métabolisme , Lymphocytes T/métabolisme , Transcription génétique
7.
Cell Rep Med ; 2(5): 100287, 2021 05 18.
Article de Anglais | MEDLINE | ID: mdl-33969320

RÉSUMÉ

Mechanisms underlying severe coronavirus disease 2019 (COVID-19) disease remain poorly understood. We analyze several thousand plasma proteins longitudinally in 306 COVID-19 patients and 78 symptomatic controls, uncovering immune and non-immune proteins linked to COVID-19. Deconvolution of our plasma proteome data using published scRNA-seq datasets reveals contributions from circulating immune and tissue cells. Sixteen percent of patients display reduced inflammation yet comparably poor outcomes. Comparison of patients who died to severely ill survivors identifies dynamic immune-cell-derived and tissue-associated proteins associated with survival, including exocrine pancreatic proteases. Using derived tissue-specific and cell-type-specific intracellular death signatures, cellular angiotensin-converting enzyme 2 (ACE2) expression, and our data, we infer whether organ damage resulted from direct or indirect effects of infection. We propose a model in which interactions among myeloid, epithelial, and T cells drive tissue damage. These datasets provide important insights and a rich resource for analysis of mechanisms of severe COVID-19 disease.

8.
bioRxiv ; 2020 Nov 03.
Article de Anglais | MEDLINE | ID: mdl-33173871

RÉSUMÉ

COVID-19 has caused over 1 million deaths globally, yet the cellular mechanisms underlying severe disease remain poorly understood. By analyzing several thousand plasma proteins in 306 COVID-19 patients and 78 symptomatic controls over serial timepoints using two complementary approaches, we uncover COVID-19 host immune and non-immune proteins not previously linked to this disease. Integration of plasma proteomics with nine published scRNAseq datasets shows that SARS-CoV-2 infection upregulates monocyte/macrophage, plasmablast, and T cell effector proteins. By comparing patients who died to severely ill patients who survived, we identify dynamic immunomodulatory and tissue-associated proteins associated with survival, providing insights into which host responses are beneficial and which are detrimental to survival. We identify intracellular death signatures from specific tissues and cell types, and by associating these with angiotensin converting enzyme 2 (ACE2) expression, we map tissue damage associated with severe disease and propose which damage results from direct viral infection rather than from indirect effects of illness. We find that disease severity in lung tissue is driven by myeloid cell phenotypes and cell-cell interactions with lung epithelial cells and T cells. Based on these results, we propose a model of immune and epithelial cell interactions that drive cell-type specific and tissue-specific damage in severe COVID-19.

9.
Cell ; 182(6): 1606-1622.e23, 2020 09 17.
Article de Anglais | MEDLINE | ID: mdl-32888429

RÉSUMÉ

The enteric nervous system (ENS) coordinates diverse functions in the intestine but has eluded comprehensive molecular characterization because of the rarity and diversity of cells. Here we develop two methods to profile the ENS of adult mice and humans at single-cell resolution: RAISIN RNA-seq for profiling intact nuclei with ribosome-bound mRNA and MIRACL-seq for label-free enrichment of rare cell types by droplet-based profiling. The 1,187,535 nuclei in our mouse atlas include 5,068 neurons from the ileum and colon, revealing extraordinary neuron diversity. We highlight circadian expression changes in enteric neurons, show that disease-related genes are dysregulated with aging, and identify differences between the ileum and proximal/distal colon. In humans, we profile 436,202 nuclei, recovering 1,445 neurons, and identify conserved and species-specific transcriptional programs and putative neuro-epithelial, neuro-stromal, and neuro-immune interactions. The human ENS expresses risk genes for neuropathic, inflammatory, and extra-intestinal diseases, suggesting neuronal contributions to disease.


Sujet(s)
Système nerveux entérique/cytologie , Système nerveux entérique/métabolisme , Régulation de l'expression des gènes au cours du développement/génétique , Neurones/métabolisme , Corps de Nissl/métabolisme , ARN messager/métabolisme , Analyse sur cellule unique/méthodes , Vieillissement/génétique , Vieillissement/métabolisme , Animaux , Horloges circadiennes/génétique , Côlon/cytologie , Côlon/métabolisme , Réticulum endoplasmique rugueux/génétique , Réticulum endoplasmique rugueux/métabolisme , Réticulum endoplasmique rugueux/ultrastructure , Cellules épithéliales/métabolisme , Femelle , Prédisposition génétique à une maladie/génétique , Humains , Iléum/cytologie , Iléum/métabolisme , Inflammation/génétique , Inflammation/métabolisme , Maladies intestinales/génétique , Maladies intestinales/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Microscopie électronique à transmission , Maladies du système nerveux/génétique , Maladies du système nerveux/métabolisme , Névroglie/cytologie , Névroglie/métabolisme , Neurones/cytologie , Corps de Nissl/génétique , Corps de Nissl/ultrastructure , ARN messager/génétique , RNA-Seq , Ribosomes/métabolisme , Ribosomes/ultrastructure , Cellules stromales/métabolisme
10.
ISME J ; 14(9): 2347-2357, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32514119

RÉSUMÉ

While horizontal gene transfer is prevalent across the biosphere, the regulatory features that enable expression and functionalization of foreign DNA remain poorly understood. Here, we combine high-throughput promoter activity measurements and large-scale genomic analysis of regulatory regions to investigate the cross-compatibility of regulatory elements (REs) in bacteria. Functional characterization of thousands of natural REs in three distinct bacterial species revealed distinct expression patterns according to RE and recipient phylogeny. Host capacity to activate foreign promoters was proportional to their genomic GC content, while many low GC regulatory elements were both broadly active and had more transcription start sites across hosts. The difference in expression capabilities could be explained by the influence of the host GC content on the stringency of the AT-rich canonical σ70 motif necessary for transcription initiation. We further confirm the generalizability of this model and find widespread GC content adaptation of the σ70 motif in a set of 1,545 genomes from all major bacterial phyla. Our analysis identifies a key mechanism by which the strength of the AT-rich σ70 motif relative to a host's genomic GC content governs the capacity for expression of acquired DNA. These findings shed light on regulatory adaptation in the context of evolving genomic composition.


Sujet(s)
Bactéries , Transfert horizontal de gène , Bactéries/génétique , Composition en bases nucléiques , ADN , Génome bactérien/génétique , Site d'initiation de la transcription
11.
Cell ; 178(3): 714-730.e22, 2019 07 25.
Article de Anglais | MEDLINE | ID: mdl-31348891

RÉSUMÉ

Genome-wide association studies (GWAS) have revealed risk alleles for ulcerative colitis (UC). To understand their cell type specificities and pathways of action, we generate an atlas of 366,650 cells from the colon mucosa of 18 UC patients and 12 healthy individuals, revealing 51 epithelial, stromal, and immune cell subsets, including BEST4+ enterocytes, microfold-like cells, and IL13RA2+IL11+ inflammatory fibroblasts, which we associate with resistance to anti-TNF treatment. Inflammatory fibroblasts, inflammatory monocytes, microfold-like cells, and T cells that co-express CD8 and IL-17 expand with disease, forming intercellular interaction hubs. Many UC risk genes are cell type specific and co-regulated within relatively few gene modules, suggesting convergence onto limited sets of cell types and pathways. Using this observation, we nominate and infer functions for specific risk genes across GWAS loci. Our work provides a framework for interrogating complex human diseases and mapping risk variants to cell types and pathways.


Sujet(s)
Rectocolite hémorragique/anatomopathologie , Côlon/métabolisme , Adulte , Sujet âgé , Anticorps monoclonaux/usage thérapeutique , Bestrophines/métabolisme , Antigènes CD8/métabolisme , Études cas-témoins , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/métabolisme , Côlon/anatomopathologie , Entérocytes/cytologie , Entérocytes/métabolisme , Femelle , Locus génétiques , Étude d'association pangénomique , Humains , Interleukine-17/métabolisme , Mâle , Adulte d'âge moyen , Facteurs de risque , Lymphocytes T/cytologie , Lymphocytes T/métabolisme , Thrombospondines/métabolisme , Facteur de nécrose tumorale alpha/immunologie , Facteur de nécrose tumorale alpha/métabolisme , Jeune adulte
12.
Nat Methods ; 15(5): 323-329, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-30052624

RÉSUMÉ

Robust and predictably performing synthetic circuits rely on the use of well-characterized regulatory parts across different genetic backgrounds and environmental contexts. Here we report the large-scale metagenomic mining of thousands of natural 5' regulatory sequences from diverse bacteria, and their multiplexed gene expression characterization in industrially relevant microbes. We identified sequences with broad and host-specific expression properties that are robust in various growth conditions. We also observed substantial differences between species in terms of their capacity to utilize exogenous regulatory sequences. Finally, we demonstrate programmable species-selective gene expression that produces distinct and diverse output patterns in different microbes. Together, these findings provide a rich resource of characterized natural regulatory sequences and a framework that can be used to engineer synthetic gene circuits with unique and tunable cross-species functionality and properties, and also suggest the prospect of ultimately engineering complex behaviors at the community level.


Sujet(s)
Régulation de l'expression des gènes/physiologie , Métagénomique/méthodes , Éléments de régulation transcriptionnelle/physiologie , Fouille de données , Escherichia coli/génétique , Escherichia coli/métabolisme , Génie génétique/méthodes , Génie métabolique , Voies et réseaux métaboliques , Spécificité d'espèce , Biologie synthétique/méthodes
13.
Cell Host Microbe ; 23(2): 229-240.e5, 2018 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-29447696

RÉSUMÉ

Fecal microbiota transplantation (FMT) from healthy donor to patient is a treatment for microbiome-associated diseases. Although the success of FMT requires donor bacteria to engraft in the patient's gut, the forces governing engraftment in humans are unknown. Here we use an ongoing clinical experiment, the treatment of recurrent Clostridium difficile infection, to uncover the rules of engraftment in humans. We built a statistical model that predicts which bacterial species will engraft in a given host, and developed Strain Finder, a method to infer strain genotypes and track them over time. We find that engraftment can be predicted largely from the abundance and phylogeny of bacteria in the donor and the pre-FMT patient. Furthermore, donor strains within a species engraft in an all-or-nothing manner and previously undetected strains frequently colonize patients receiving FMT. We validated these findings for metabolic syndrome, suggesting that the same principles of engraftment extend to other indications.


Sujet(s)
Clostridioides difficile/croissance et développement , Infections à Clostridium/prévention et contrôle , Transplantation de microbiote fécal/méthodes , Microbiome gastro-intestinal/physiologie , Tube digestif/microbiologie , Prévention secondaire/méthodes , Biodiversité , Infections à Clostridium/thérapie , Humains , Modèles biologiques , Récidive
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...