Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(25): 4841-4860.e25, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36493756

ABSTRACT

We present a multiomic cell atlas of human lung development that combines single-cell RNA and ATAC sequencing, high-throughput spatial transcriptomics, and single-cell imaging. Coupling single-cell methods with spatial analysis has allowed a comprehensive cellular survey of the epithelial, mesenchymal, endothelial, and erythrocyte/leukocyte compartments from 5-22 post-conception weeks. We identify previously uncharacterized cell states in all compartments. These include developmental-specific secretory progenitors and a subtype of neuroendocrine cell related to human small cell lung cancer. Our datasets are available through our web interface (https://lungcellatlas.org). To illustrate its general utility, we use our cell atlas to generate predictions about cell-cell signaling and transcription factor hierarchies which we rigorously test using organoid models.


Subject(s)
Fetus , Lung , Humans , Cell Differentiation , Gene Expression Profiling , Lung/cytology , Organogenesis , Organoids , Atlases as Topic , Fetus/cytology
2.
Nat Immunol ; 21(3): 343-353, 2020 03.
Article in English | MEDLINE | ID: mdl-32066951

ABSTRACT

Gastrointestinal microbiota and immune cells interact closely and display regional specificity; however, little is known about how these communities differ with location. Here, we simultaneously assess microbiota and single immune cells across the healthy, adult human colon, with paired characterization of immune cells in the mesenteric lymph nodes, to delineate colonic immune niches at steady state. We describe distinct helper T cell activation and migration profiles along the colon and characterize the transcriptional adaptation trajectory of regulatory T cells between lymphoid tissue and colon. Finally, we show increasing B cell accumulation, clonal expansion and mutational frequency from the cecum to the sigmoid colon and link this to the increasing number of reactive bacterial species.


Subject(s)
Colon/immunology , Colon/microbiology , Gastrointestinal Microbiome/immunology , Adult , B-Lymphocytes/immunology , Colon/cytology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation , Organ Specificity , RNA-Seq , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Transcriptome
3.
Nature ; 631(8019): 189-198, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38898278

ABSTRACT

The COVID-19 pandemic is an ongoing global health threat, yet our understanding of the dynamics of early cellular responses to this disease remains limited1. Here in our SARS-CoV-2 human challenge study, we used single-cell multi-omics profiling of nasopharyngeal swabs and blood to temporally resolve abortive, transient and sustained infections in seronegative individuals challenged with pre-Alpha SARS-CoV-2. Our analyses revealed rapid changes in cell-type proportions and dozens of highly dynamic cellular response states in epithelial and immune cells associated with specific time points and infection status. We observed that the interferon response in blood preceded the nasopharyngeal response. Moreover, nasopharyngeal immune infiltration occurred early in samples from individuals with only transient infection and later in samples from individuals with sustained infection. High expression of HLA-DQA2 before inoculation was associated with preventing sustained infection. Ciliated cells showed multiple immune responses and were most permissive for viral replication, whereas nasopharyngeal T cells and macrophages were infected non-productively. We resolved 54 T cell states, including acutely activated T cells that clonally expanded while carrying convergent SARS-CoV-2 motifs. Our new computational pipeline Cell2TCR identifies activated antigen-responding T cells based on a gene expression signature and clusters these into clonotype groups and motifs. Overall, our detailed time series data can serve as a Rosetta stone for epithelial and immune cell responses and reveals early dynamic responses associated with protection against infection.


Subject(s)
COVID-19 , Nasopharynx , SARS-CoV-2 , Single-Cell Analysis , T-Lymphocytes , Humans , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Nasopharynx/virology , Nasopharynx/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Interferons/immunology , Interferons/metabolism , Male , Female , Macrophages/immunology , Macrophages/virology , Virus Replication , Epithelial Cells/virology , Epithelial Cells/immunology , Time Factors , Adult
4.
Nature ; 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38057666

ABSTRACT

Human limbs emerge during the fourth post-conception week as mesenchymal buds, which develop into fully formed limbs over the subsequent months1. This process is orchestrated by numerous temporally and spatially restricted gene expression programmes, making congenital alterations in phenotype common2. Decades of work with model organisms have defined the fundamental mechanisms underlying vertebrate limb development, but an in-depth characterization of this process in humans has yet to be performed. Here we detail human embryonic limb development across space and time using single-cell and spatial transcriptomics. We demonstrate extensive diversification of cells from a few multipotent progenitors to myriad differentiated cell states, including several novel cell populations. We uncover two waves of human muscle development, each characterized by different cell states regulated by separate gene expression programmes, and identify musculin (MSC) as a key transcriptional repressor maintaining muscle stem cell identity. Through assembly of multiple anatomically continuous spatial transcriptomic samples using VisiumStitcher, we map cells across a sagittal section of a whole fetal hindlimb. We reveal a clear anatomical segregation between genes linked to brachydactyly and polysyndactyly, and uncover transcriptionally and spatially distinct populations of the mesenchyme in the autopod. Finally, we perform single-cell RNA sequencing on mouse embryonic limbs to facilitate cross-species developmental comparison, finding substantial homology between the two species.

5.
Nature ; 619(7971): 801-810, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37438528

ABSTRACT

The function of a cell is defined by its intrinsic characteristics and its niche: the tissue microenvironment in which it dwells. Here we combine single-cell and spatial transcriptomics data to discover cellular niches within eight regions of the human heart. We map cells to microanatomical locations and integrate knowledge-based and unsupervised structural annotations. We also profile the cells of the human cardiac conduction system1. The results revealed their distinctive repertoire of ion channels, G-protein-coupled receptors (GPCRs) and regulatory networks, and implicated FOXP2 in the pacemaker phenotype. We show that the sinoatrial node is compartmentalized, with a core of pacemaker cells, fibroblasts and glial cells supporting glutamatergic signalling. Using a custom CellPhoneDB.org module, we identify trans-synaptic pacemaker cell interactions with glia. We introduce a druggable target prediction tool, drug2cell, which leverages single-cell profiles and drug-target interactions to provide mechanistic insights into the chronotropic effects of drugs, including GLP-1 analogues. In the epicardium, we show enrichment of both IgG+ and IgA+ plasma cells forming immune niches that may contribute to infection defence. Overall, we provide new clarity to cardiac electro-anatomy and immunology, and our suite of computational approaches can be applied to other tissues and organs.


Subject(s)
Cellular Microenvironment , Heart , Multiomics , Myocardium , Humans , Cell Communication , Fibroblasts/cytology , Glutamic Acid/metabolism , Heart/anatomy & histology , Heart/innervation , Ion Channels/metabolism , Myocardium/cytology , Myocardium/immunology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Neuroglia/cytology , Pericardium/cytology , Pericardium/immunology , Plasma Cells/immunology , Receptors, G-Protein-Coupled/metabolism , Sinoatrial Node/anatomy & histology , Sinoatrial Node/cytology , Sinoatrial Node/physiology , Heart Conduction System/anatomy & histology , Heart Conduction System/cytology , Heart Conduction System/metabolism
6.
Nature ; 602(7896): 321-327, 2022 02.
Article in English | MEDLINE | ID: mdl-34937051

ABSTRACT

It is not fully understood why COVID-19 is typically milder in children1-3. Here, to examine the differences between children and adults in their response to SARS-CoV-2 infection, we analysed paediatric and adult patients with COVID-19 as well as healthy control individuals (total n = 93) using single-cell multi-omic profiling of matched nasal, tracheal, bronchial and blood samples. In the airways of healthy paediatric individuals, we observed cells that were already in an interferon-activated state, which after SARS-CoV-2 infection was further induced especially in airway immune cells. We postulate that higher paediatric innate interferon responses restrict viral replication and disease progression. The systemic response in children was characterized by increases in naive lymphocytes and a depletion of natural killer cells, whereas, in adults, cytotoxic T cells and interferon-stimulated subpopulations were significantly increased. We provide evidence that dendritic cells initiate interferon signalling in early infection, and identify epithelial cell states associated with COVID-19 and age. Our matching nasal and blood data show a strong interferon response in the airways with the induction of systemic interferon-stimulated populations, which were substantially reduced in paediatric patients. Together, we provide several mechanisms that explain the milder clinical syndrome observed in children.


Subject(s)
COVID-19/blood , COVID-19/immunology , Dendritic Cells/immunology , Interferons/immunology , Killer Cells, Natural/immunology , SARS-CoV-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Bronchi/immunology , Bronchi/virology , COVID-19/pathology , Chicago , Cohort Studies , Disease Progression , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Humans , Immunity, Innate , London , Male , Nasal Mucosa/immunology , Nasal Mucosa/virology , SARS-CoV-2/growth & development , Single-Cell Analysis , Trachea/virology , Young Adult
7.
Nature ; 597(7875): 250-255, 2021 09.
Article in English | MEDLINE | ID: mdl-34497389

ABSTRACT

The cellular landscape of the human intestinal tract is dynamic throughout life, developing in utero and changing in response to functional requirements and environmental exposures. Here, to comprehensively map cell lineages, we use single-cell RNA sequencing and antigen receptor analysis of almost half a million cells from up to 5 anatomical regions in the developing and up to 11 distinct anatomical regions in the healthy paediatric and adult human gut. This reveals the existence of transcriptionally distinct BEST4 epithelial cells throughout the human intestinal tract. Furthermore, we implicate IgG sensing as a function of intestinal tuft cells. We describe neural cell populations in the developing enteric nervous system, and predict cell-type-specific expression of genes associated with Hirschsprung's disease. Finally, using a systems approach, we identify key cell players that drive the formation of secondary lymphoid tissue in early human development. We show that these programs are adopted in inflammatory bowel disease to recruit and retain immune cells at the site of inflammation. This catalogue of intestinal cells will provide new insights into cellular programs in development, homeostasis and disease.


Subject(s)
Aging , Enteric Nervous System/cytology , Fetus/cytology , Health , Intestines/cytology , Intestines/growth & development , Lymph Nodes/cytology , Lymph Nodes/growth & development , Adult , Animals , Child , Crohn Disease/pathology , Datasets as Topic , Enteric Nervous System/anatomy & histology , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Epithelial Cells/cytology , Female , Fetus/anatomy & histology , Fetus/embryology , Humans , Intestines/embryology , Intestines/innervation , Lymph Nodes/embryology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Organogenesis , Receptors, IgG/metabolism , Signal Transduction , Spatio-Temporal Analysis , Time Factors
8.
Nature ; 588(7838): 466-472, 2020 12.
Article in English | MEDLINE | ID: mdl-32971526

ABSTRACT

Cardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and therapeutic strategies require a deeper understanding of the molecular processes involved in the healthy heart. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavour. Here, using state-of-the-art analyses of large-scale single-cell and single-nucleus transcriptomes, we characterize six anatomical adult heart regions. Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes and fibroblasts, and reveal distinct atrial and ventricular subsets of cells with diverse developmental origins and specialized properties. We define the complexity of the cardiac vasculature and its changes along the arterio-venous axis. In the immune compartment, we identify cardiac-resident macrophages with inflammatory and protective transcriptional signatures. Furthermore, analyses of cell-to-cell interactions highlight different networks of macrophages, fibroblasts and cardiomyocytes between atria and ventricles that are distinct from those of skeletal muscle. Our human cardiac cell atlas improves our understanding of the human heart and provides a valuable reference for future studies.


Subject(s)
Myocardium/cytology , Single-Cell Analysis , Transcriptome , Adipocytes/classification , Adipocytes/metabolism , Adult , Angiotensin-Converting Enzyme 2/analysis , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Epithelial Cells/classification , Epithelial Cells/metabolism , Epithelium , Female , Fibroblasts/classification , Fibroblasts/metabolism , Gene Expression Profiling , Genome-Wide Association Study , Heart Atria/anatomy & histology , Heart Atria/cytology , Heart Atria/innervation , Heart Ventricles/anatomy & histology , Heart Ventricles/cytology , Heart Ventricles/innervation , Homeostasis/immunology , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myocytes, Cardiac/classification , Myocytes, Cardiac/metabolism , Neurons/classification , Neurons/metabolism , Pericytes/classification , Pericytes/metabolism , Receptors, Coronavirus/analysis , Receptors, Coronavirus/genetics , Receptors, Coronavirus/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Stromal Cells/classification , Stromal Cells/metabolism
9.
Nature ; 574(7778): 365-371, 2019 10.
Article in English | MEDLINE | ID: mdl-31597962

ABSTRACT

Definitive haematopoiesis in the fetal liver supports self-renewal and differentiation of haematopoietic stem cells and multipotent progenitors (HSC/MPPs) but remains poorly defined in humans. Here, using single-cell transcriptome profiling of approximately 140,000 liver and 74,000 skin, kidney and yolk sac cells, we identify the repertoire of human blood and immune cells during development. We infer differentiation trajectories from HSC/MPPs and evaluate the influence of the tissue microenvironment on blood and immune cell development. We reveal physiological erythropoiesis in fetal skin and the presence of mast cells, natural killer and innate lymphoid cell precursors in the yolk sac. We demonstrate a shift in the haemopoietic composition of fetal liver during gestation away from being predominantly erythroid, accompanied by a parallel change in differentiation potential of HSC/MPPs, which we functionally validate. Our integrated map of fetal liver haematopoiesis provides a blueprint for the study of paediatric blood and immune disorders, and a reference for harnessing the therapeutic potential of HSC/MPPs.


Subject(s)
Fetus/cytology , Hematopoiesis , Liver/cytology , Liver/embryology , Blood Cells/cytology , Cellular Microenvironment , Female , Fetus/metabolism , Flow Cytometry , Gene Expression Profiling , Humans , Liver/metabolism , Lymphoid Tissue/cytology , Single-Cell Analysis , Stem Cells/metabolism
10.
Nature ; 563(7731): 347-353, 2018 11.
Article in English | MEDLINE | ID: mdl-30429548

ABSTRACT

During early human pregnancy the uterine mucosa transforms into the decidua, into which the fetal placenta implants and where placental trophoblast cells intermingle and communicate with maternal cells. Trophoblast-decidual interactions underlie common diseases of pregnancy, including pre-eclampsia and stillbirth. Here we profile the transcriptomes of about 70,000 single cells from first-trimester placentas with matched maternal blood and decidual cells. The cellular composition of human decidua reveals subsets of perivascular and stromal cells that are located in distinct decidual layers. There are three major subsets of decidual natural killer cells that have distinctive immunomodulatory and chemokine profiles. We develop a repository of ligand-receptor complexes and a statistical tool to predict the cell-type specificity of cell-cell communication via these molecular interactions. Our data identify many regulatory interactions that prevent harmful innate or adaptive immune responses in this environment. Our single-cell atlas of the maternal-fetal interface reveals the cellular organization of the decidua and placenta, and the interactions that are critical for placentation and reproductive success.


Subject(s)
Cell Communication , Fetus/cytology , Histocompatibility, Maternal-Fetal/immunology , Placenta/cytology , Placenta/metabolism , Pregnancy/immunology , Single-Cell Analysis , Cell Communication/immunology , Cell Differentiation/genetics , Decidua/cytology , Decidua/immunology , Decidua/metabolism , Female , Fetus/immunology , Fetus/metabolism , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Ligands , Placenta/immunology , RNA, Small Cytoplasmic/genetics , Sequence Analysis, RNA , Stromal Cells/cytology , Stromal Cells/metabolism , Transcriptome , Trophoblasts/cytology , Trophoblasts/immunology , Trophoblasts/metabolism
11.
Bioinformatics ; 36(3): 964-965, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31400197

ABSTRACT

MOTIVATION: Increasing numbers of large scale single cell RNA-Seq projects are leading to a data explosion, which can only be fully exploited through data integration. A number of methods have been developed to combine diverse datasets by removing technical batch effects, but most are computationally intensive. To overcome the challenge of enormous datasets, we have developed BBKNN, an extremely fast graph-based data integration algorithm. We illustrate the power of BBKNN on large scale mouse atlasing data, and favourably benchmark its run time against a number of competing methods. AVAILABILITY AND IMPLEMENTATION: BBKNN is available at https://github.com/Teichlab/bbknn, along with documentation and multiple example notebooks, and can be installed from pip. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Algorithms , Transcriptome , Animals , Mice , RNA-Seq , Exome Sequencing
13.
Bioinformatics ; 34(5): 884-886, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29126246

ABSTRACT

Summary: Every year, a large number of novel algorithms are introduced to the scientific community for a myriad of applications, but using these across different research groups is often troublesome, due to suboptimal implementations and specific dependency requirements. This does not have to be the case, as public cloud computing services can easily house tractable implementations within self-contained dependency environments, making the methods easily accessible to a wider public. We have taken 14 popular methods, the majority related to expression data or promoter analysis, developed these up to a good implementation standard and housed the tools in isolated Docker containers which we integrated into the CyVerse Discovery Environment, making these easily usable for a wide community as part of the CyVerse UK project. Availability and implementation: The integrated apps can be found at http://www.cyverse.org/discovery-environment, while the raw code is available at https://github.com/cyversewarwick and the corresponding Docker images are housed at https://hub.docker.com/r/cyversewarwick/. Contact: info@cyverse.warwick.ac.uk or D.L.Wild@warwick.ac.uk. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Cloud Computing , Computational Biology/methods , Gene Expression Regulation , Promoter Regions, Genetic , Software , Algorithms , Gene Expression Profiling/methods , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods
14.
Plant Cell ; 27(11): 3038-64, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26566919

ABSTRACT

Transcriptional reprogramming is integral to effective plant defense. Pathogen effectors act transcriptionally and posttranscriptionally to suppress defense responses. A major challenge to understanding disease and defense responses is discriminating between transcriptional reprogramming associated with microbial-associated molecular pattern (MAMP)-triggered immunity (MTI) and that orchestrated by effectors. A high-resolution time course of genome-wide expression changes following challenge with Pseudomonas syringae pv tomato DC3000 and the nonpathogenic mutant strain DC3000hrpA- allowed us to establish causal links between the activities of pathogen effectors and suppression of MTI and infer with high confidence a range of processes specifically targeted by effectors. Analysis of this information-rich data set with a range of computational tools provided insights into the earliest transcriptional events triggered by effector delivery, regulatory mechanisms recruited, and biological processes targeted. We show that the majority of genes contributing to disease or defense are induced within 6 h postinfection, significantly before pathogen multiplication. Suppression of chloroplast-associated genes is a rapid MAMP-triggered defense response, and suppression of genes involved in chromatin assembly and induction of ubiquitin-related genes coincide with pathogen-induced abscisic acid accumulation. Specific combinations of promoter motifs are engaged in fine-tuning the MTI response and active transcriptional suppression at specific promoter configurations by P. syringae.


Subject(s)
Arabidopsis/immunology , Immunosuppression Therapy , Pathogen-Associated Molecular Pattern Molecules/metabolism , Plant Immunity/genetics , Plant Leaves/immunology , Pseudomonas syringae/physiology , Transcription, Genetic , Arabidopsis/genetics , Arabidopsis/microbiology , Base Sequence , Chromatin/metabolism , Gene Expression Profiling , Gene Expression Regulation, Plant , Gene Ontology , Gene Regulatory Networks , Genes, Plant , Molecular Sequence Data , Nucleotide Motifs/genetics , Plant Diseases/genetics , Plant Diseases/immunology , Plant Diseases/microbiology , Plant Leaves/genetics , Plant Leaves/microbiology , Promoter Regions, Genetic/genetics , Pseudomonas syringae/growth & development , Transcription Factors/metabolism
16.
Bioinformatics ; 30(7): 962-70, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24351708

ABSTRACT

MOTIVATION: Identification of modules of co-regulated genes is a crucial first step towards dissecting the regulatory circuitry underlying biological processes. Co-regulated genes are likely to reveal themselves by showing tight co-expression, e.g. high correlation of expression profiles across multiple time series datasets. However, numbers of up- or downregulated genes are often large, making it difficult to discriminate between dependent co-expression resulting from co-regulation and independent co-expression. Furthermore, modules of co-regulated genes may only show tight co-expression across a subset of the time series, i.e. show condition-dependent regulation. RESULTS: Wigwams is a simple and efficient method to identify gene modules showing evidence for co-regulation in multiple time series of gene expression data. Wigwams analyzes similarities of gene expression patterns within each time series (condition) and directly tests the dependence or independence of these across different conditions. The expression pattern of each gene in each subset of conditions is tested statistically as a potential signature of a condition-dependent regulatory mechanism regulating multiple genes. Wigwams does not require particular time points and can process datasets that are on different time scales. Differential expression relative to control conditions can be taken into account. The output is succinct and non-redundant, enabling gene network reconstruction to be focused on those gene modules and combinations of conditions that show evidence for shared regulatory mechanisms. Wigwams was run using six Arabidopsis time series expression datasets, producing a set of biologically significant modules spanning different combinations of conditions. AVAILABILITY AND IMPLEMENTATION: A Matlab implementation of Wigwams, complete with graphical user interfaces and documentation, is available at: warwick.ac.uk/wigwams. .


Subject(s)
Gene Expression Profiling/methods , Gene Expression , Software , Arabidopsis/genetics , Gene Expression Regulation, Plant , Gene Regulatory Networks
17.
Front Oncol ; 14: 1323961, 2024.
Article in English | MEDLINE | ID: mdl-38410100

ABSTRACT

Background: Neoadjuvant radiotherapy (neo-RT) is widely used in locally advanced rectal cancer (LARC) as a component of radical treatment. Despite the advantages of neo-RT, which typically improves outcomes in LARC patients, the lack of reliable biomarkers that predict response and monitor the efficacy of therapy, can result in the application of unnecessary aggressive therapy affecting patients' quality of life. Hence, the search for molecular biomarkers for assessing the radio responsiveness of this cancer represents a relevant issue. Methods: Here, we combined proteomic and metabolomic approaches to identify molecular signatures, which could discriminate LARC tumors with good and poor responses to neo-RT. Results: The integration of data on differentially accumulated proteins and metabolites made it possible to identify disrupted metabolic pathways and signaling processes connected with response to irradiation, including ketone bodies synthesis and degradation, purine metabolism, energy metabolism, degradation of fatty acid, amino acid metabolism, and focal adhesion. Moreover, we proposed multi-component panels of proteins and metabolites which could serve as a solid base to develop biomarkers for monitoring and predicting the efficacy of preoperative RT in rectal cancer patients. Conclusion: We proved that an integrated multi-omic approach presents a valid look at the analysis of the global response to cancer treatment from the perspective of metabolomic reprogramming.

18.
Nat Biotechnol ; 42(1): 40-51, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37055623

ABSTRACT

Assessment of single-cell gene expression (single-cell RNA sequencing) and adaptive immune receptor (AIR) sequencing (scVDJ-seq) has been invaluable in studying lymphocyte biology. Here we introduce Dandelion, a computational pipeline for scVDJ-seq analysis. It enables the application of standard V(D)J analysis workflows to single-cell datasets, delivering improved V(D)J contig annotation and the identification of nonproductive and partially spliced contigs. We devised a strategy to create an AIR feature space that can be used for both differential V(D)J usage analysis and pseudotime trajectory inference. The application of Dandelion improved the alignment of human thymic development trajectories of double-positive T cells to mature single-positive CD4/CD8 T cells, generating predictions of factors regulating lineage commitment. Dandelion analysis of other cell compartments provided insights into the origins of human B1 cells and ILC/NK cell development, illustrating the power of our approach. Dandelion is available at https://www.github.com/zktuong/dandelion .


Subject(s)
Taraxacum , Humans , T-Lymphocytes , Single-Cell Analysis
19.
Sci Rep ; 14(1): 7756, 2024 04 02.
Article in English | MEDLINE | ID: mdl-38565965

ABSTRACT

SAG21/LEA5 is an unusual late embryogenesis abundant protein in Arabidopsis thaliana, that is primarily mitochondrially located and may be important in regulating translation in both chloroplasts and mitochondria. SAG21 expression is regulated by a plethora of abiotic and biotic stresses and plant growth regulators indicating a complex regulatory network. To identify key transcription factors regulating SAG21 expression, yeast-1-hybrid screens were used to identify transcription factors that bind the 1685 bp upstream of the SAG21 translational start site. Thirty-three transcription factors from nine different families bound to the SAG21 promoter, including members of the ERF, WRKY and NAC families. Key binding sites for both NAC and WRKY transcription factors were tested through site directed mutagenesis indicating the presence of cryptic binding sites for both these transcription factor families. Co-expression in protoplasts confirmed the activation of SAG21 by WRKY63/ABO3, and SAG21 upregulation elicited by oligogalacturonide elicitors was partially dependent on WRKY63, indicating its role in SAG21 pathogen responses. SAG21 upregulation by ethylene was abolished in the erf1 mutant, while wound-induced SAG21 expression was abolished in anac71 mutants, indicating SAG21 expression can be regulated by several distinct transcription factors depending on the stress condition.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Transcription Factors/metabolism , Arabidopsis/metabolism , Gene Expression Regulation, Plant , Arabidopsis Proteins/metabolism , Oxidation-Reduction , Plant Proteins/genetics , Plant Proteins/metabolism , Stress, Physiological
20.
Nat Aging ; 4(5): 727-744, 2024 May.
Article in English | MEDLINE | ID: mdl-38622407

ABSTRACT

Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species.


Subject(s)
Aging , Muscle, Skeletal , Humans , Aging/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Animals , Mice , Adult , Aged , Sarcopenia/pathology , Sarcopenia/metabolism , Male , Neuromuscular Junction/metabolism , Middle Aged , Female
SELECTION OF CITATIONS
SEARCH DETAIL