Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nature ; 586(7831): 785-789, 2020 10.
Article in English | MEDLINE | ID: mdl-33057196

ABSTRACT

In the mammalian lung, an apparently homogenous mesh of capillary vessels surrounds each alveolus, forming the vast respiratory surface across which oxygen transfers to the blood1. Here we use single-cell analysis to elucidate the cell types, development, renewal and evolution of the alveolar capillary endothelium. We show that alveolar capillaries are mosaics; similar to the epithelium that lines the alveolus, the alveolar endothelium is made up of two intermingled cell types, with complex 'Swiss-cheese'-like morphologies and distinct functions. The first cell type, which we term the 'aerocyte', is specialized for gas exchange and the trafficking of leukocytes, and is unique to the lung. The other cell type, termed gCap ('general' capillary), is specialized to regulate vasomotor tone, and functions as a stem/progenitor cell in capillary homeostasis and repair. The two cell types develop from bipotent progenitors, mature gradually and are affected differently in disease and during ageing. This cell-type specialization is conserved between mouse and human lungs but is not found in alligator or turtle lungs, suggesting it arose during the evolution of the mammalian lung. The discovery of cell type specialization in alveolar capillaries transforms our understanding of the structure, function, regulation and maintenance of the air-blood barrier and gas exchange in health, disease and evolution.


Subject(s)
Capillaries/cytology , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/cytology , Pulmonary Gas Exchange , Aging , Alligators and Crocodiles/anatomy & histology , Animals , Biological Evolution , Capillaries/metabolism , Cell Division , Cell Self Renewal , Cellular Senescence , Humans , Male , Mice , Pulmonary Alveoli/metabolism , Stem Cells/classification , Stem Cells/cytology , Turtles/anatomy & histology
2.
Nature ; 587(7835): 619-625, 2020 11.
Article in English | MEDLINE | ID: mdl-33208946

ABSTRACT

Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1-9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.


Subject(s)
Cells/classification , Cells/metabolism , Immunity , Lung/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome/genetics , Aged , Animals , Atlases as Topic , Biomarkers , Cell Communication , Cells/immunology , Chemokines/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Female , Humans , Lung/immunology , Male , Mice , Middle Aged , Receptors, Lymphocyte Homing/metabolism , Signal Transduction , Stromal Cells/metabolism
4.
J Biol Chem ; 289(44): 30511-30524, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25231983

ABSTRACT

Methylation of various components of the translational machinery has been shown to globally affect protein synthesis. Little is currently known about the role of lysine methylation on elongation factors. Here we show that in Saccharomyces cerevisiae, the product of the EFM3/YJR129C gene is responsible for the trimethylation of lysine 509 on elongation factor 2. Deletion of EFM3 or of the previously described EFM2 increases sensitivity to antibiotics that target translation and decreases translational fidelity. Furthermore, the amino acid sequences of Efm3 and Efm2, as well as their respective methylation sites on EF2, are conserved in other eukaryotes. These results suggest the importance of lysine methylation modification of EF2 in fine tuning the translational apparatus.


Subject(s)
Methyltransferases/physiology , Peptide Elongation Factor 2/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Catalytic Domain , Conserved Sequence , Methylation , Methyltransferases/chemistry , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Biosynthesis , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry
5.
Biochem Biophys Res Commun ; 455(3-4): 382-9, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25446118

ABSTRACT

The elongation factors of Saccharomyces cerevisiae are extensively methylated, containing a total of ten methyllysine residues. Elongation factor methyltransferases (Efm1, Efm2, Efm3, and Efm4) catalyze at least four of these modifications. Here we report the identification of a new type of protein lysine methyltransferase, Efm5 (Ygr001c), which was initially classified as N6-adenine DNA methyltransferase-like. Efm5 is required for trimethylation of Lys-79 on EF1A. We directly show the loss of this modification in efm5Δ strains by both mass spectrometry and amino acid analysis. Close homologs of Efm5 are found in vertebrates, invertebrates, and plants, although some fungal species apparently lack this enzyme. This suggests possible unique functions of this modification in S. cerevisiae and higher eukaryotes. The misannotation of Efm5 was due to the presence of a DPPF sequence in post-Motif II, typically associated with DNA methylation. Further analysis of this motif and others like it demonstrates a potential consensus sequence for N-methyltransferases.


Subject(s)
Gene Deletion , Histone-Lysine N-Methyltransferase/chemistry , Lysine/chemistry , Peptide Elongation Factor 1/chemistry , Saccharomyces cerevisiae/genetics , Amino Acid Motifs , Amino Acid Sequence , Computational Biology , Evolution, Molecular , Genotype , Lysine/analogs & derivatives , Mass Spectrometry , Protein Processing, Post-Translational , S-Adenosylmethionine/chemistry , Saccharomyces cerevisiae Proteins/chemistry
6.
bioRxiv ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38915664

ABSTRACT

Throughout an organism's life, a multitude of biological systems transition through complex biophysical processes. These processes serve as indicators of the underlying biological states. Inferring these latent unobserved states is a key problem in modern biology and neuroscience. Unfortunately, in many experimental setups we can at best obtain snapshots of the system at different times for different individuals, and one major challenge is the one of reconciling those measurements. This formalism is particularly relevant in the study of Alzheimer's Disease (AD) progression, in which we observe in brain donors the aggregation of pathological proteins but the underlying disease state is unknown. The progression of AD can be modeled by assigning a latent score - termed pseudotime - to each pathological state, creating a pseudotemporal ordering of donors based on their pathological burden. This paper proposes a hierarchical Bayesian framework to model AD progression using detailed quantification of multiple AD pathological proteins from the Seattle AD Brain Cell Atlas consortium (SEA-AD). Inspired by biophysical models, we model pathological burden as an exponential process. Theoretical properties of the model are studied, by using linearization to reveal convergence and identifiability properties. We provide Markov chain Monte Carlo estimation algorithms, and show the effectiveness of our approach with multiple simulation studies across data conditions. Applying the methodology to SEA-AD brain data, we infer pseudotime for each donor and order them by pathological burden. Finally, we analyze the information within each pathological feature and utilize it to refine the model by focusing on the most informative pathologies. This lays the groundwork for suggesting future experimental design approaches.

7.
J Exp Med ; 221(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38597954

ABSTRACT

Early stages of deadly respiratory diseases including COVID-19 are challenging to elucidate in humans. Here, we define cellular tropism and transcriptomic effects of SARS-CoV-2 virus by productively infecting healthy human lung tissue and using scRNA-seq to reconstruct the transcriptional program in "infection pseudotime" for individual lung cell types. SARS-CoV-2 predominantly infected activated interstitial macrophages (IMs), which can accumulate thousands of viral RNA molecules, taking over 60% of the cell transcriptome and forming dense viral RNA bodies while inducing host profibrotic (TGFB1, SPP1) and inflammatory (early interferon response, CCL2/7/8/13, CXCL10, and IL6/10) programs and destroying host cell architecture. Infected alveolar macrophages (AMs) showed none of these extreme responses. Spike-dependent viral entry into AMs used ACE2 and Sialoadhesin/CD169, whereas IM entry used DC-SIGN/CD209. These results identify activated IMs as a prominent site of viral takeover, the focus of inflammation and fibrosis, and suggest targeting CD209 to prevent early pathology in COVID-19 pneumonia. This approach can be generalized to any human lung infection and to evaluate therapeutics.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Macrophages , Inflammation , RNA, Viral , Lung
8.
Science ; 382(6667): eadf2359, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824649

ABSTRACT

Single-cell transcriptomic studies have identified a conserved set of neocortical cell types from small postmortem cohorts. We extended these efforts by assessing cell type variation across 75 adult individuals undergoing epilepsy and tumor surgeries. Nearly all nuclei map to one of 125 robust cell types identified in the middle temporal gyrus. However, we found interindividual variance in abundances and gene expression signatures, particularly in deep-layer glutamatergic neurons and microglia. A minority of donor variance is explainable by age, sex, ancestry, disease state, and cell state. Genomic variation was associated with expression of 150 to 250 genes for most cell types. This characterization of cellular variation provides a baseline for cell typing in health and disease.


Subject(s)
Temporal Lobe , Transcriptome , Adult , Humans , Epilepsy/metabolism , Gene Expression Profiling , Neurons/metabolism , Temporal Lobe/cytology , Temporal Lobe/metabolism , Nervous System Diseases/genetics , Mental Disorders/genetics
9.
Nat Med ; 29(6): 1563-1577, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37291214

ABSTRACT

Single-cell technologies have transformed our understanding of human tissues. Yet, studies typically capture only a limited number of donors and disagree on cell type definitions. Integrating many single-cell datasets can address these limitations of individual studies and capture the variability present in the population. Here we present the integrated Human Lung Cell Atlas (HLCA), combining 49 datasets of the human respiratory system into a single atlas spanning over 2.4 million cells from 486 individuals. The HLCA presents a consensus cell type re-annotation with matching marker genes, including annotations of rare and previously undescribed cell types. Leveraging the number and diversity of individuals in the HLCA, we identify gene modules that are associated with demographic covariates such as age, sex and body mass index, as well as gene modules changing expression along the proximal-to-distal axis of the bronchial tree. Mapping new data to the HLCA enables rapid data annotation and interpretation. Using the HLCA as a reference for the study of disease, we identify shared cell states across multiple lung diseases, including SPP1+ profibrotic monocyte-derived macrophages in COVID-19, pulmonary fibrosis and lung carcinoma. Overall, the HLCA serves as an example for the development and use of large-scale, cross-dataset organ atlases within the Human Cell Atlas.


Subject(s)
COVID-19 , Lung Neoplasms , Pulmonary Fibrosis , Humans , Lung , Lung Neoplasms/genetics , Macrophages
10.
Science ; 382(6667): eade9516, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824638

ABSTRACT

The cognitive abilities of humans are distinctive among primates, but their molecular and cellular substrates are poorly understood. We used comparative single-nucleus transcriptomics to analyze samples of the middle temporal gyrus (MTG) from adult humans, chimpanzees, gorillas, rhesus macaques, and common marmosets to understand human-specific features of the neocortex. Human, chimpanzee, and gorilla MTG showed highly similar cell-type composition and laminar organization as well as a large shift in proportions of deep-layer intratelencephalic-projecting neurons compared with macaque and marmoset MTG. Microglia, astrocytes, and oligodendrocytes had more-divergent expression across species compared with neurons or oligodendrocyte precursor cells, and neuronal expression diverged more rapidly on the human lineage. Only a few hundred genes showed human-specific patterning, suggesting that relatively few cellular and molecular changes distinctively define adult human cortical structure.


Subject(s)
Cognition , Hominidae , Neocortex , Temporal Lobe , Animals , Humans , Gene Expression Profiling , Gorilla gorilla/genetics , Hominidae/genetics , Hominidae/physiology , Macaca mulatta/genetics , Pan troglodytes/genetics , Phylogeny , Transcriptome , Neocortex/physiology , Species Specificity , Temporal Lobe/physiology
11.
Science ; 382(6667): eadf6812, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824655

ABSTRACT

Variation in cytoarchitecture is the basis for the histological definition of cortical areas. We used single cell transcriptomics and performed cellular characterization of the human cortex to better understand cortical areal specialization. Single-nucleus RNA-sequencing of 8 areas spanning cortical structural variation showed a highly consistent cellular makeup for 24 cell subclasses. However, proportions of excitatory neuron subclasses varied substantially, likely reflecting differences in connectivity across primary sensorimotor and association cortices. Laminar organization of astrocytes and oligodendrocytes also differed across areas. Primary visual cortex showed characteristic organization with major changes in the excitatory to inhibitory neuron ratio, expansion of layer 4 excitatory neurons, and specialized inhibitory neurons. These results lay the groundwork for a refined cellular and molecular characterization of human cortical cytoarchitecture and areal specialization.


Subject(s)
Neocortex , Humans , Neocortex/metabolism , Neocortex/ultrastructure , Neurons/classification , Neurons/metabolism , Transcriptome , Single-Cell Gene Expression Analysis , Phylogeny
12.
Res Sq ; 2023 May 23.
Article in English | MEDLINE | ID: mdl-37292694

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia in older adults. Neuropathological and imaging studies have demonstrated a progressive and stereotyped accumulation of protein aggregates, but the underlying molecular and cellular mechanisms driving AD progression and vulnerable cell populations affected by disease remain coarsely understood. The current study harnesses single cell and spatial genomics tools and knowledge from the BRAIN Initiative Cell Census Network to understand the impact of disease progression on middle temporal gyrus cell types. We used image-based quantitative neuropathology to place 84 donors spanning the spectrum of AD pathology along a continuous disease pseudoprogression score and multiomic technologies to profile single nuclei from each donor, mapping their transcriptomes, epigenomes, and spatial coordinates to a common cell type reference with unprecedented resolution. Temporal analysis of cell-type proportions indicated an early reduction of Somatostatin-expressing neuronal subtypes and a late decrease of supragranular intratelencephalic-projecting excitatory and Parvalbumin-expressing neurons, with increases in disease-associated microglial and astrocytic states. We found complex gene expression differences, ranging from global to cell type-specific effects. These effects showed different temporal patterns indicating diverse cellular perturbations as a function of disease progression. A subset of donors showed a particularly severe cellular and molecular phenotype, which correlated with steeper cognitive decline. We have created a freely available public resource to explore these data and to accelerate progress in AD research at SEA-AD.org.

13.
Science ; 376(6594): eabl4896, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35549404

ABSTRACT

Molecular characterization of cell types using single-cell transcriptome sequencing is revolutionizing cell biology and enabling new insights into the physiology of human organs. We created a human reference atlas comprising nearly 500,000 cells from 24 different tissues and organs, many from the same donor. This atlas enabled molecular characterization of more than 400 cell types, their distribution across tissues, and tissue-specific variation in gene expression. Using multiple tissues from a single donor enabled identification of the clonal distribution of T cells between tissues, identification of the tissue-specific mutation rate in B cells, and analysis of the cell cycle state and proliferative potential of shared cell types across tissues. Cell type-specific RNA splicing was discovered and analyzed across tissues within an individual.


Subject(s)
Atlases as Topic , Cells , Organ Specificity , RNA Splicing , Single-Cell Analysis , Transcriptome , B-Lymphocytes/metabolism , Cells/metabolism , Humans , Organ Specificity/genetics , T-Lymphocytes/metabolism
14.
Elife ; 52016 12 20.
Article in English | MEDLINE | ID: mdl-27996937

ABSTRACT

The comprehensive understanding of cellular signaling pathways remains a challenge due to multiple layers of regulation that may become evident only when the pathway is probed at different levels or critical nodes are eliminated. To discover regulatory mechanisms in canonical WNT signaling, we conducted a systematic forward genetic analysis through reporter-based screens in haploid human cells. Comparison of screens for negative, attenuating and positive regulators of WNT signaling, mediators of R-spondin-dependent signaling and suppressors of constitutive signaling induced by loss of the tumor suppressor adenomatous polyposis coli or casein kinase 1α uncovered new regulatory features at most levels of the pathway. These include a requirement for the transcription factor AP-4, a role for the DAX domain of AXIN2 in controlling ß-catenin transcriptional activity, a contribution of glycophosphatidylinositol anchor biosynthesis and glypicans to R-spondin-potentiated WNT signaling, and two different mechanisms that regulate signaling when distinct components of the ß-catenin destruction complex are lost. The conceptual and methodological framework we describe should enable the comprehensive understanding of other signaling systems.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Genetic Testing/methods , Wnt Signaling Pathway , Casein Kinase I/deficiency , Cytoskeletal Proteins/deficiency , Genes, Reporter , Haploidy , Humans , Wnt Proteins/genetics , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL