Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Publication year range
1.
Cell ; 187(10): 2343-2358, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38729109

ABSTRACT

As the number of single-cell datasets continues to grow rapidly, workflows that map new data to well-curated reference atlases offer enormous promise for the biological community. In this perspective, we discuss key computational challenges and opportunities for single-cell reference-mapping algorithms. We discuss how mapping algorithms will enable the integration of diverse datasets across disease states, molecular modalities, genetic perturbations, and diverse species and will eventually replace manual and laborious unsupervised clustering pipelines.


Subject(s)
Algorithms , Single-Cell Analysis , Single-Cell Analysis/methods , Humans , Computational Biology/methods , Data Analysis , Animals , Cluster Analysis
2.
Cell ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38925112

ABSTRACT

Most mammalian genes have multiple polyA sites, representing a substantial source of transcript diversity regulated by the cleavage and polyadenylation (CPA) machinery. To better understand how these proteins govern polyA site choice, we introduce CPA-Perturb-seq, a multiplexed perturbation screen dataset of 42 CPA regulators with a 3' scRNA-seq readout that enables transcriptome-wide inference of polyA site usage. We develop a framework to detect perturbation-dependent changes in polyadenylation and characterize modules of co-regulated polyA sites. We find groups of intronic polyA sites regulated by distinct components of the nuclear RNA life cycle, including elongation, splicing, termination, and surveillance. We train and validate a deep neural network (APARENT-Perturb) for tandem polyA site usage, delineating a cis-regulatory code that predicts perturbation response and reveals interactions between regulatory complexes. Our work highlights the potential for multiplexed single-cell perturbation screens to further our understanding of post-transcriptional regulation.

3.
Cell ; 184(13): 3573-3587.e29, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34062119

ABSTRACT

The simultaneous measurement of multiple modalities represents an exciting frontier for single-cell genomics and necessitates computational methods that can define cellular states based on multimodal data. Here, we introduce "weighted-nearest neighbor" analysis, an unsupervised framework to learn the relative utility of each data type in each cell, enabling an integrative analysis of multiple modalities. We apply our procedure to a CITE-seq dataset of 211,000 human peripheral blood mononuclear cells (PBMCs) with panels extending to 228 antibodies to construct a multimodal reference atlas of the circulating immune system. Multimodal analysis substantially improves our ability to resolve cell states, allowing us to identify and validate previously unreported lymphoid subpopulations. Moreover, we demonstrate how to leverage this reference to rapidly map new datasets and to interpret immune responses to vaccination and coronavirus disease 2019 (COVID-19). Our approach represents a broadly applicable strategy to analyze single-cell multimodal datasets and to look beyond the transcriptome toward a unified and multimodal definition of cellular identity.


Subject(s)
SARS-CoV-2/immunology , Single-Cell Analysis/methods , 3T3 Cells , Animals , COVID-19/immunology , Cell Line , Gene Expression Profiling/methods , Humans , Immunity/immunology , Leukocytes, Mononuclear/immunology , Lymphocytes/immunology , Mice , Sequence Analysis, RNA/methods , Transcriptome/immunology , Vaccination
4.
Nat Immunol ; 24(10): 1725-1734, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37735591

ABSTRACT

The immune response to SARS-CoV-2 antigen after infection or vaccination is defined by the durable production of antibodies and T cells. Population-based monitoring typically focuses on antibody titer, but there is a need for improved characterization and quantification of T cell responses. Here, we used multimodal sequencing technologies to perform a longitudinal analysis of circulating human leukocytes collected before and after immunization with the mRNA vaccine BNT162b2. Our data indicated distinct subpopulations of CD8+ T cells, which reliably appeared 28 days after prime vaccination. Using a suite of cross-modality integration tools, we defined their transcriptome, accessible chromatin landscape and immunophenotype, and we identified unique biomarkers within each modality. We further showed that this vaccine-induced population was SARS-CoV-2 antigen-specific and capable of rapid clonal expansion. Moreover, we identified these CD8+ T cell populations in scRNA-seq datasets from COVID-19 patients and found that their relative frequency and differentiation outcomes were predictive of subsequent clinical outcomes.


Subject(s)
CD8-Positive T-Lymphocytes , COVID-19 , Humans , COVID-19 Vaccines , SARS-CoV-2 , BNT162 Vaccine , COVID-19/prevention & control , Vaccination , Antibodies, Viral
5.
Nat Rev Mol Cell Biol ; 24(10): 695-713, 2023 10.
Article in English | MEDLINE | ID: mdl-37280296

ABSTRACT

Single-cell multi-omics technologies and methods characterize cell states and activities by simultaneously integrating various single-modality omics methods that profile the transcriptome, genome, epigenome, epitranscriptome, proteome, metabolome and other (emerging) omics. Collectively, these methods are revolutionizing molecular cell biology research. In this comprehensive Review, we discuss established multi-omics technologies as well as cutting-edge and state-of-the-art methods in the field. We discuss how multi-omics technologies have been adapted and improved over the past decade using a framework characterized by optimization of throughput and resolution, modality integration, uniqueness and accuracy, and we also discuss multi-omics limitations. We highlight the impact that single-cell multi-omics technologies have had in cell lineage tracing, tissue-specific and cell-specific atlas production, tumour immunology and cancer genetics, and in mapping of cellular spatial information in fundamental and translational research. Finally, we discuss bioinformatics tools that have been developed to link different omics modalities and elucidate functionality through the use of better mathematical modelling and computational methods.


Subject(s)
Computational Biology , Multiomics , Cell Lineage , Epigenome , Metabolome
6.
Cell ; 182(3): 641-654.e20, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32615085

ABSTRACT

Targeting glycolysis has been considered therapeutically intractable owing to its essential housekeeping role. However, the context-dependent requirement for individual glycolytic steps has not been fully explored. We show that CRISPR-mediated targeting of glycolysis in T cells in mice results in global loss of Th17 cells, whereas deficiency of the glycolytic enzyme glucose phosphate isomerase (Gpi1) selectively eliminates inflammatory encephalitogenic and colitogenic Th17 cells, without substantially affecting homeostatic microbiota-specific Th17 cells. In homeostatic Th17 cells, partial blockade of glycolysis upon Gpi1 inactivation was compensated by pentose phosphate pathway flux and increased mitochondrial respiration. In contrast, inflammatory Th17 cells experience a hypoxic microenvironment known to limit mitochondrial respiration, which is incompatible with loss of Gpi1. Our study suggests that inhibiting glycolysis by targeting Gpi1 could be an effective therapeutic strategy with minimum toxicity for Th17-mediated autoimmune diseases, and, more generally, that metabolic redundancies can be exploited for selective targeting of disease processes.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Glucose-6-Phosphate Isomerase/metabolism , Glycolysis/genetics , Oxidative Phosphorylation , Pentose Phosphate Pathway/physiology , Th17 Cells/metabolism , Animals , Cell Hypoxia/genetics , Cell Hypoxia/immunology , Chimera/genetics , Chromatography, Gas , Chromatography, Liquid , Clostridium Infections/immunology , Cytokines/deficiency , Cytokines/genetics , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Glucose-6-Phosphate Isomerase/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis/immunology , Homeostasis/genetics , Homeostasis/immunology , Inflammation/genetics , Inflammation/immunology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Pentose Phosphate Pathway/genetics , Pentose Phosphate Pathway/immunology , RNA-Seq , Single-Cell Analysis , Th17 Cells/immunology , Th17 Cells/pathology
7.
Cell ; 177(7): 1888-1902.e21, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31178118

ABSTRACT

Single-cell transcriptomics has transformed our ability to characterize cell states, but deep biological understanding requires more than a taxonomic listing of clusters. As new methods arise to measure distinct cellular modalities, a key analytical challenge is to integrate these datasets to better understand cellular identity and function. Here, we develop a strategy to "anchor" diverse datasets together, enabling us to integrate single-cell measurements not only across scRNA-seq technologies, but also across different modalities. After demonstrating improvement over existing methods for integrating scRNA-seq data, we anchor scRNA-seq experiments with scATAC-seq to explore chromatin differences in closely related interneuron subsets and project protein expression measurements onto a bone marrow atlas to characterize lymphocyte populations. Lastly, we harmonize in situ gene expression and scRNA-seq datasets, allowing transcriptome-wide imputation of spatial gene expression patterns. Our work presents a strategy for the assembly of harmonized references and transfer of information across datasets.


Subject(s)
Databases, Nucleic Acid , Gene Expression Profiling , Sequence Analysis, RNA , Single-Cell Analysis , Software , Transcriptome , Humans
8.
Cell ; 179(7): 1455-1467, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31835027

ABSTRACT

Understanding the genetic and molecular drivers of phenotypic heterogeneity across individuals is central to biology. As new technologies enable fine-grained and spatially resolved molecular profiling, we need new computational approaches to integrate data from the same organ across different individuals into a consistent reference and to construct maps of molecular and cellular organization at histological and anatomical scales. Here, we review previous efforts and discuss challenges involved in establishing such a common coordinate framework, the underlying map of tissues and organs. We focus on strategies to handle anatomical variation across individuals and highlight the need for new technologies and analytical methods spanning multiple hierarchical scales of spatial resolution.


Subject(s)
Anatomic Variation , Diagnostic Imaging/standards , Physical Examination/standards , Diagnostic Imaging/methods , Humans , Physical Examination/methods , Reference Standards
9.
Nat Immunol ; 22(6): 723-734, 2021 06.
Article in English | MEDLINE | ID: mdl-33958784

ABSTRACT

Continuous supply of immune cells throughout life relies on the delicate balance in the hematopoietic stem cell (HSC) pool between long-term maintenance and meeting the demands of both normal blood production and unexpected stress conditions. Here we identified distinct subsets of human long-term (LT)-HSCs that responded differently to regeneration-mediated stress: an immune checkpoint ligand CD112lo subset that exhibited a transient engraftment restraint (termed latency) before contributing to hematopoietic reconstitution and a primed CD112hi subset that responded rapidly. This functional heterogeneity and CD112 expression are regulated by INKA1 through direct interaction with PAK4 and SIRT1, inducing epigenetic changes and defining an alternative state of LT-HSC quiescence that serves to preserve self-renewal and regenerative capacity upon regeneration-mediated stress. Collectively, our data uncovered the molecular intricacies underlying HSC heterogeneity and self-renewal regulation and point to latency as an orchestrated physiological response that balances blood cell demands with preserving a stem cell reservoir.


Subject(s)
Cell Self Renewal/immunology , Hematopoietic Stem Cells/physiology , Immune Reconstitution , Multipotent Stem Cells/physiology , Stress, Physiological/immunology , Adult , Animals , Cell Self Renewal/genetics , Cells, Cultured , Epigenesis, Genetic/immunology , Female , Fetal Blood/cytology , Flow Cytometry , Gene Knockdown Techniques , Hematopoiesis , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunomagnetic Separation , Infant, Newborn , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Middle Aged , Nectins/metabolism , Primary Cell Culture , RNA-Seq , Single-Cell Analysis , Sirtuin 1/metabolism , Stress, Physiological/genetics , Transplantation, Heterologous , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
10.
Cell ; 174(3): 622-635.e13, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29909983

ABSTRACT

Transcription factors regulate the molecular, morphological, and physiological characteristics of neurons and generate their impressive cell-type diversity. To gain insight into the general principles that govern how transcription factors regulate cell-type diversity, we used large-scale single-cell RNA sequencing to characterize the extensive cellular diversity in the Drosophila optic lobes. We sequenced 55,000 single cells and assigned them to 52 clusters. We validated and annotated many clusters using RNA sequencing of FACS-sorted single-cell types and cluster-specific genes. To identify transcription factors responsible for inducing specific terminal differentiation features, we generated a "random forest" model, and we showed that the transcription factors Apterous and Traffic-jam are required in many but not all cholinergic and glutamatergic neurons, respectively. In fact, the same terminal characters often can be regulated by different transcription factors in different cell types, arguing for extensive phenotypic convergence. Our data provide a deep understanding of the developmental and functional specification of a complex brain structure.


Subject(s)
Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental/physiology , Neurogenesis/physiology , Animals , Cell Differentiation , Cholinergic Neurons/physiology , Cluster Analysis , Computer Simulation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Profiling/methods , Homeodomain Proteins , LIM-Homeodomain Proteins/metabolism , Maf Transcription Factors, Large/metabolism , Neuroglia/physiology , Neurons/physiology , Neurotransmitter Agents/genetics , Neurotransmitter Agents/physiology , Optic Lobe, Nonmammalian/physiology , Phenotype , Proto-Oncogene Proteins/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
11.
Cell ; 165(7): 1721-1733, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27212234

ABSTRACT

Plant roots can regenerate after excision of their tip, including the stem cell niche. To determine which developmental program mediates such repair, we applied a combination of lineage tracing, single-cell RNA sequencing, and marker analysis to test different models of tissue reassembly. We show that multiple cell types can reconstitute stem cells, demonstrating the latent potential of untreated plant cells. The transcriptome of regenerating cells prior to stem cell activation resembles that of an embryonic root progenitor. Regeneration defects are more severe in embryonic than in adult root mutants. Furthermore, the signaling domains of the hormones auxin and cytokinin mirror their embryonic dynamics and manipulation of both hormones alters the position of new tissues and stem cell niche markers. Our findings suggest that plant root regeneration follows, on a larger scale, the developmental stages of embryonic patterning and is guided by spatial information provided by complementary hormone domains.


Subject(s)
Plant Roots/physiology , Cytokinins/metabolism , Gene Expression Profiling , Indoleacetic Acids/metabolism , Plant Cells , Plant Growth Regulators/metabolism , Plant Roots/cytology , Seeds , Single-Cell Analysis , Stem Cell Niche , Stem Cells/cytology
12.
Cell ; 162(6): 1309-21, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26343579

ABSTRACT

Encounters between immune cells and invading bacteria ultimately determine the course of infection. These interactions are usually measured in populations of cells, masking cell-to-cell variation that may be important for infection outcome. To characterize the gene expression variation that underlies distinct infection outcomes and monitor infection phenotypes, we developed an experimental system that combines single-cell RNA-seq with fluorescent markers. Probing the responses of individual macrophages to invading Salmonella, we find that variation between individual infected host cells is determined by the heterogeneous activity of bacterial factors in individual infecting bacteria. We illustrate how variable PhoPQ activity in the population of invading bacteria drives variable host type I IFN responses by modifying LPS in a subset of bacteria. This work demonstrates a causative link between host and bacterial variability, with cell-to-cell variation between different bacteria being sufficient to drive radically different host immune responses. This co-variation has implications for host-pathogen dynamics in vivo.


Subject(s)
Host-Pathogen Interactions , Macrophages/immunology , Salmonella typhimurium/physiology , Animals , Interferon Type I/immunology , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Salmonella Infections/immunology , Salmonella Infections/microbiology , Specific Pathogen-Free Organisms
13.
Cell ; 163(6): 1400-12, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26607794

ABSTRACT

Extensive cellular heterogeneity exists within specific immune-cell subtypes classified as a single lineage, but its molecular underpinnings are rarely characterized at a genomic scale. Here, we use single-cell RNA-seq to investigate the molecular mechanisms governing heterogeneity and pathogenicity of Th17 cells isolated from the central nervous system (CNS) and lymph nodes (LN) at the peak of autoimmune encephalomyelitis (EAE) or differentiated in vitro under either pathogenic or non-pathogenic polarization conditions. Computational analysis relates a spectrum of cellular states in vivo to in-vitro-differentiated Th17 cells and unveils genes governing pathogenicity and disease susceptibility. Using knockout mice, we validate four new genes: Gpr65, Plzp, Toso, and Cd5l (in a companion paper). Cellular heterogeneity thus informs Th17 function in autoimmunity and can identify targets for selective suppression of pathogenic Th17 cells while potentially sparing non-pathogenic tissue-protective ones.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Sequence Analysis, RNA , Single-Cell Analysis , Th17 Cells/metabolism , Th17 Cells/pathology , Animals , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Profiling , Humans , Kruppel-Like Transcription Factors/metabolism , Lymph Nodes/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/metabolism , Peptide Fragments/metabolism , Promyelocytic Leukemia Zinc Finger Protein , Receptors, G-Protein-Coupled/metabolism , Receptors, Immunologic/metabolism , Receptors, Scavenger , Th17 Cells/immunology
14.
Cell ; 162(3): 675-86, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26189680

ABSTRACT

Finding the components of cellular circuits and determining their functions systematically remains a major challenge in mammalian cells. Here, we introduced genome-wide pooled CRISPR-Cas9 libraries into dendritic cells (DCs) to identify genes that control the induction of tumor necrosis factor (Tnf) by bacterial lipopolysaccharide (LPS), a key process in the host response to pathogens, mediated by the Tlr4 pathway. We found many of the known regulators of Tlr4 signaling, as well as dozens of previously unknown candidates that we validated. By measuring protein markers and mRNA profiles in DCs that are deficient in known or candidate genes, we classified the genes into three functional modules with distinct effects on the canonical responses to LPS and highlighted functions for the PAF complex and oligosaccharyltransferase (OST) complex. Our findings uncover new facets of innate immune circuits in primary cells and provide a genetic approach for dissection of mammalian cell circuits.


Subject(s)
CRISPR-Cas Systems , Genetic Techniques , Immunity, Innate , Animals , Bone Marrow Cells/immunology , Cell Differentiation , Cell Survival , Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Knockout Techniques , Gene Regulatory Networks , Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/immunology
15.
Cell ; 161(5): 1202-1214, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26000488

ABSTRACT

Cells, the basic units of biological structure and function, vary broadly in type and state. Single-cell genomics can characterize cell identity and function, but limitations of ease and scale have prevented its broad application. Here we describe Drop-seq, a strategy for quickly profiling thousands of individual cells by separating them into nanoliter-sized aqueous droplets, associating a different barcode with each cell's RNAs, and sequencing them all together. Drop-seq analyzes mRNA transcripts from thousands of individual cells simultaneously while remembering transcripts' cell of origin. We analyzed transcriptomes from 44,808 mouse retinal cells and identified 39 transcriptionally distinct cell populations, creating a molecular atlas of gene expression for known retinal cell classes and novel candidate cell subtypes. Drop-seq will accelerate biological discovery by enabling routine transcriptional profiling at single-cell resolution. VIDEO ABSTRACT.


Subject(s)
Gene Expression Profiling/methods , Genome-Wide Association Study , Microfluidic Analytical Techniques , Retina/cytology , Single-Cell Analysis , Animals , High-Throughput Nucleotide Sequencing , Mice , Sequence Analysis, RNA
16.
Cell ; 162(3): 607-21, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26232227

ABSTRACT

We identified a dominant missense mutation in the SCN transcription factor Zfhx3, termed short circuit (Zfhx3(Sci)), which accelerates circadian locomotor rhythms in mice. ZFHX3 regulates transcription via direct interaction with predicted AT motifs in target genes. The mutant protein has a decreased ability to activate consensus AT motifs in vitro. Using RNA sequencing, we found minimal effects on core clock genes in Zfhx3(Sci/+) SCN, whereas the expression of neuropeptides critical for SCN intercellular signaling was significantly disturbed. Moreover, mutant ZFHX3 had a decreased ability to activate AT motifs in the promoters of these neuropeptide genes. Lentiviral transduction of SCN slices showed that the ZFHX3-mediated activation of AT motifs is circadian, with decreased amplitude and robustness of these oscillations in Zfhx3(Sci/+) SCN slices. In conclusion, by cloning Zfhx3(Sci), we have uncovered a circadian transcriptional axis that determines the period and robustness of behavioral and SCN molecular rhythms.


Subject(s)
Circadian Rhythm , Gene Expression Regulation , Homeodomain Proteins/metabolism , Neuropeptides/genetics , Suprachiasmatic Nucleus/metabolism , Amino Acid Sequence , Animals , Down-Regulation , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Nucleotide Motifs , Promoter Regions, Genetic , Sequence Alignment , Transcription, Genetic
17.
Cell ; 159(1): 148-162, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25219674

ABSTRACT

Pseudouridine is the most abundant RNA modification, yet except for a few well-studied cases, little is known about the modified positions and their function(s). Here, we develop Ψ-seq for transcriptome-wide quantitative mapping of pseudouridine. We validate Ψ-seq with spike-ins and de novo identification of previously reported positions and discover hundreds of unique sites in human and yeast mRNAs and snoRNAs. Perturbing pseudouridine synthases (PUS) uncovers which pseudouridine synthase modifies each site and their target sequence features. mRNA pseudouridinylation depends on both site-specific and snoRNA-guided pseudouridine synthases. Upon heat shock in yeast, Pus7p-mediated pseudouridylation is induced at >200 sites, and PUS7 deletion decreases the levels of otherwise pseudouridylated mRNA, suggesting a role in enhancing transcript stability. rRNA pseudouridine stoichiometries are conserved but reduced in cells from dyskeratosis congenita patients, where the PUS DKC1 is mutated. Our work identifies an enhanced, transcriptome-wide scope for pseudouridine and methods to dissect its underlying mechanisms and function.


Subject(s)
Pseudouridine/analysis , RNA, Messenger/chemistry , RNA, Untranslated/chemistry , Animals , Candida albicans/genetics , Candida albicans/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/metabolism , Gene Expression Profiling , Humans , Intramolecular Transferases/chemistry , Intramolecular Transferases/metabolism , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pseudouridine/metabolism , RNA/chemistry , RNA/genetics , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Telomerase/chemistry , Telomerase/genetics
18.
Nature ; 619(7970): 585-594, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37468583

ABSTRACT

Understanding kidney disease relies on defining the complexity of cell types and states, their associated molecular profiles and interactions within tissue neighbourhoods1. Here we applied multiple single-cell and single-nucleus assays (>400,000 nuclei or cells) and spatial imaging technologies to a broad spectrum of healthy reference kidneys (45 donors) and diseased kidneys (48 patients). This has provided a high-resolution cellular atlas of 51 main cell types, which include rare and previously undescribed cell populations. The multi-omic approach provides detailed transcriptomic profiles, regulatory factors and spatial localizations spanning the entire kidney. We also define 28 cellular states across nephron segments and interstitium that were altered in kidney injury, encompassing cycling, adaptive (successful or maladaptive repair), transitioning and degenerative states. Molecular signatures permitted the localization of these states within injury neighbourhoods using spatial transcriptomics, while large-scale 3D imaging analysis (around 1.2 million neighbourhoods) provided corresponding linkages to active immune responses. These analyses defined biological pathways that are relevant to injury time-course and niches, including signatures underlying epithelial repair that predicted maladaptive states associated with a decline in kidney function. This integrated multimodal spatial cell atlas of healthy and diseased human kidneys represents a comprehensive benchmark of cellular states, neighbourhoods, outcome-associated signatures and publicly available interactive visualizations.


Subject(s)
Gene Expression Profiling , Kidney Diseases , Kidney , Single-Cell Analysis , Transcriptome , Humans , Cell Nucleus/genetics , Kidney/cytology , Kidney/injuries , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Transcriptome/genetics , Case-Control Studies , Imaging, Three-Dimensional
19.
Cell ; 155(6): 1409-21, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24269006

ABSTRACT

N(6)-methyladenosine (m(6)A) is the most ubiquitous mRNA base modification, but little is known about its precise location, temporal dynamics, and regulation. Here, we generated genomic maps of m(6)A sites in meiotic yeast transcripts at nearly single-nucleotide resolution, identifying 1,308 putatively methylated sites within 1,183 transcripts. We validated eight out of eight methylation sites in different genes with direct genetic analysis, demonstrated that methylated sites are significantly conserved in a related species, and built a model that predicts methylated sites directly from sequence. Sites vary in their methylation profiles along a dense meiotic time course and are regulated both locally, via predictable methylatability of each site, and globally, through the core meiotic circuitry. The methyltransferase complex components localize to the yeast nucleolus, and this localization is essential for mRNA methylation. Our data illuminate a conserved, dynamically regulated methylation program in yeast meiosis and provide an important resource for studying the function of this epitranscriptomic modification.


Subject(s)
Meiosis , RNA, Fungal/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces/cytology , Saccharomyces/metabolism , Adenosine/analogs & derivatives , Adenosine/analysis , Adenosine/metabolism , Cell Nucleolus/metabolism , Genome, Fungal , Methylation , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , tRNA Methyltransferases/metabolism
20.
Nature ; 597(7875): 196-205, 2021 09.
Article in English | MEDLINE | ID: mdl-34497388

ABSTRACT

The Human Developmental Cell Atlas (HDCA) initiative, which is part of the Human Cell Atlas, aims to create a comprehensive reference map of cells during development. This will be critical to understanding normal organogenesis, the effect of mutations, environmental factors and infectious agents on human development, congenital and childhood disorders, and the cellular basis of ageing, cancer and regenerative medicine. Here we outline the HDCA initiative and the challenges of mapping and modelling human development using state-of-the-art technologies to create a reference atlas across gestation. Similar to the Human Genome Project, the HDCA will integrate the output from a growing community of scientists who are mapping human development into a unified atlas. We describe the early milestones that have been achieved and the use of human stem-cell-derived cultures, organoids and animal models to inform the HDCA, especially for prenatal tissues that are hard to acquire. Finally, we provide a roadmap towards a complete atlas of human development.


Subject(s)
Cell Movement , Cell Tracking , Cells/cytology , Developmental Biology/methods , Embryo, Mammalian/cytology , Fetus/cytology , Information Dissemination , Organogenesis , Adult , Animals , Atlases as Topic , Cell Culture Techniques , Cell Survival , Data Visualization , Female , Humans , Imaging, Three-Dimensional , Male , Models, Animal , Organogenesis/genetics , Organoids/cytology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL