Single-nucleus cross-tissue molecular reference maps toward understanding disease gene function

Understanding gene function and regulation in homeostasis and disease requires knowledge of the cellular and tissue contexts in which genes are expressed. Here, we applied four single-nucleus RNA sequencing methods to eight diverse, archived, frozen tissue types from 16 donors and 25 samples, generating a cross-tissue atlas of 209,126 nuclei profiles, which we integrated across tissues, donors, and laboratory methods with a conditional variational autoencoder. Using the resulting cross-tissue atlas, we highlight shared and tissue-specific features of tissue-resident cell populations; identify cell types that might contribute to neuromuscular, metabolic, and immune components of monogenic diseases and the biological processes involved in their pathology; and determine cell types and gene modules that might underlie disease mechanisms for complex traits analyzed by genome-wide association studies. Description Cartography of human cells The function of disease genes active in different cell types is modulated to meet the needs of the different tissues and organs in which the cells reside. Resolving these differences is critical to understanding homeostasis and disease. However, single-cell atlases generated to date have largely focused on individual tissues. Eraslan et al. applied single-nucleus RNA sequencing to frozen, banked samples from eight healthy human organs from 16 donors and characterized cell populations across tissues, including tissue-resident myeloid and fibroblast populations, and their role in tissue support and immunity (see the Perspective by Liu and Zhang). Using this cross-tissue atlas, the authors linked specific cell populations to monogenic and polygenic diseases, suggesting cell- and tissue-specific programs. —LZ and DJ Optimized single-nucleus RNA sequencing of eight frozen, banked human tissues helps reveal cell types and mechanisms underlying monogenic diseases and complex traits. INTRODUCTION Understanding and treating disease requires deep, systematic characterization of different cells and their interactions across human tissues and organs, along with characterization of the genetic variants that causally contribute to disease risk. Recent studies have combined single-cell atlases of specific human tissues and organs with genes associated with human disease to relate risk variants to likely cells of action. However, it has been challenging to extend these studies to profile multiple tissues and organs across the body, conduct studies at population scale, and integrate cell atlases from multiple organs to yield unified insights. RATIONALE Because of the pleiotropy and specificity of disease-associated variants, systematically relating variants to cells and molecular processes requires analysis across multiple tissues and individuals. Prior cell atlases primarily relied on fresh tissue samples from a single organ or tissue. Single-nucleus RNA sequencing (snRNA-seq) can be applied to frozen, archived tissue and captures cell types that do not survive dissociation across many tissues. Deep learning methods can integrate data across individuals and tissues by controlling for batch effects while preserving biological variation. RESULTS We established a framework for multitissue human cell atlases and generated an atlas of 209,126 snRNA-seq profiles from eight tissue types across 16 individuals, archived as frozen tissue as part of the Genotype-Tissue Expression (GTEx) project. We benchmarked four protocols and show how to apply them in a pooled setting to enable larger studies. We integrated the cross-tissue atlas using a conditional variational autoencoder, annotated it with 43 broad and 74 fine categories, and demonstrated its use to decipher tissue residency, such as a macrophage dichotomy and lipid associations that are preserved across tissues, and tissue-specific fibroblast features, including lung alveolar fibroblasts with likely roles in mechanosensation. We relate cells to human disease biology and disease-risk genes for both rare and common diseases, including rare muscle disease gene groups enriched in distinct subsets of myonuclei and nonmyocytes, and cell type–specific enrichment of expression and splicing quantitative trait locus (QTL) target genes mapped to genome-wide association study loci. CONCLUSION Our framework will empower large, cross-tissue population and/or disease studies at single-cell resolution. These frameworks and the cross-tissue perspective provided here will form a basis for larger-scale future studies to improve our understanding of cross-tissue and cross-individual variation of cellular phenotypes in relation to disease-associated genetic variation. Cross-tissue snRNA-seq atlas in eight frozen, archived adult human tissues. Tissue sites and experimental pipeline (top row). The resulting atlas enables a cross-tissue census of tissue-specific and shared cell types (middle left). Differentiation trajectories and compositional analysis of dichotomous macrophage populations improve our understanding of tissue residency (middle center and right). Analyses of fibroblasts across tissues reveal tissue-specific and shared fibroblast features and their functional interpretation (bottom left). Robust and scalable computational methods enable comprehensive associations of monogenic and complex diseases to tissue-specific and shared cell populations (bottom center and right). E. mucosa, esophagus mucosa; E. muscularis, esophagus muscularis; Sk. muscle, skeletal muscle.

[1]  Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches , 2022, Cell.

[2]  B. Donadille,et al.  Molecular and Cellular Bases of Lipodystrophy Syndromes , 2022, Frontiers in Endocrinology.

[3]  A. Regev,et al.  ECLIPSER: identifying causal cell types and genes for complex traits through single cell enrichment of e/sQTL-mapped genes in GWAS loci , 2021, bioRxiv.

[4]  Ramkumar Veppathur Mohan,et al.  Identifying disease-critical cell types and cellular processes across the human body by integration of single-cell profiles and human genetics , 2021, bioRxiv.

[5]  A. Regev,et al.  Single-cell profiling of proteins and chromatin accessibility using PHAGE-ATAC , 2021, Nature Biotechnology.

[6]  Y. Saeys,et al.  Spatial proteogenomics reveals distinct and evolutionarily-conserved hepatic macrophage niches , 2021, bioRxiv.

[7]  D. Swandulla,et al.  Complexity of skeletal muscle degeneration: multi-systems pathophysiology and organ crosstalk in dystrophinopathy , 2021, Pflügers Archiv - European Journal of Physiology.

[8]  J. Todd,et al.  An open approach to systematically prioritize causal variants and genes at all published human GWAS trait-associated loci , 2021, Nature Genetics.

[9]  B. Garmy-Susini,et al.  Cardiac macrophage subsets differentially regulate lymphatic network remodeling during pressure overload , 2021, Scientific Reports.

[10]  Evan Z. Macosko,et al.  An atlas of healthy and injured cell states and niches in the human kidney , 2021, bioRxiv.

[11]  N. Rosenthal,et al.  Fibroblasts: Origins, definitions, and functions in health and disease , 2021, Cell.

[12]  E. Klechevsky,et al.  The cellular architecture of the antimicrobial response network in human leprosy granulomas , 2021, Nature Immunology.

[13]  Milena B. Furtado,et al.  Adult fibroblasts retain organ-specific transcriptomic identity , 2021, bioRxiv.

[14]  Bertrand Z. Yeung,et al.  Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells , 2021, Nature Biotechnology.

[15]  Tim O. Nieuwenhuis,et al.  Single cell RNA-seq analysis of the flexor digitorum brevis mouse myofibers , 2021, Skeletal muscle.

[16]  R. Bourgon,et al.  Cross-tissue organization of the fibroblast lineage , 2021, Nature.

[17]  Xuesong Shi,et al.  Single-cell RNA sequencing in cancer: Applications, advances, and emerging challenges , 2021, Molecular therapy oncolytics.

[18]  Timothy L. Tickle,et al.  COVID-19 tissue atlases reveal SARS-CoV-2 pathology and cellular targets , 2021, Nature.

[19]  M. Mayr,et al.  Fibroblast GATA-4 and GATA-6 promote myocardial adaptation to pressure overload by enhancing cardiac angiogenesis , 2021, Basic Research in Cardiology.

[20]  David S. Fischer,et al.  Single-cell meta-analysis of SARS-CoV-2 entry genes across tissues and demographics , 2021, Nature Medicine.

[21]  C. Bock,et al.  Spontaneously Resolved Atopic Dermatitis Shows Melanocyte and Immune Cell Activation Distinct From Healthy Control Skin , 2021, Frontiers in Immunology.

[22]  E. Mercuri,et al.  Duchenne muscular dystrophy , 2021, Nature Reviews Disease Primers.

[23]  G. Washko,et al.  Single Cell RNA-seq and Mass Cytometry Reveals a Novel and a Targetable Population of Macrophages in Idiopathic Pulmonary Fibrosis , 2021, bioRxiv.

[24]  LG Nord-Ost - Mecklenburg-Vorpommern , 2021, VPT Magazin.

[25]  Gautier Koscielny,et al.  Open Targets Genetics: systematic identification of trait-associated genes using large-scale genetics and functional genomics , 2020, Nucleic Acids Res..

[26]  Christopher D. Brown,et al.  Widespread dose-dependent effects of RNA expression and splicing on complex diseases and traits , 2019, bioRxiv.

[27]  N. Marquardt,et al.  Distinct developmental pathways from blood monocytes generate human lung macrophage diversity. , 2020, Immunity.

[28]  D. Pinto,et al.  Cell type-specific isolation and transcriptomic profiling informs glial pathology in human temporal lobe epilepsy , 2020, bioRxiv.

[29]  F. Rivier,et al.  The 2021 version of the gene table of neuromuscular disorders (nuclear genome) , 2020, Neuromuscular Disorders.

[30]  K. Plath,et al.  Female human primordial germ cells display X-chromosome dosage compensation despite the absence of X-inactivation , 2020, Nature Cell Biology.

[31]  A. Regev,et al.  snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis , 2020, Nature.

[32]  J. C. Love,et al.  Second-Strand Synthesis-Based Massively Parallel scRNA-Seq Reveals Cellular States and Molecular Features of Human Inflammatory Skin Pathologies , 2020, Immunity.

[33]  A. Hartner,et al.  A role for the alpha-8 integrin chain (itga8) in glomerular homeostasis of the kidney , 2020, Molecular and Cellular Pediatrics.

[34]  Catherine L. Worth,et al.  Cells of the adult human heart , 2020, Nature.

[35]  Michael J. Steinbaugh,et al.  Translocation of Viable Gut Microbiota to Mesenteric Adipose Drives Formation of Creeping Fat in Humans , 2020, Cell.

[36]  R. Guigó,et al.  Cell type–specific genetic regulation of gene expression across human tissues , 2020, Science.

[37]  Jacob C. Ulirsch,et al.  Leveraging polygenic enrichments of gene features to predict genes underlying complex traits and diseases , 2020, Nature Genetics.

[38]  Ramkumar Veppathur Mohan,et al.  Single-nucleus and spatial transcriptomics of archival pancreatic cancer reveals multi-compartment reprogramming after neoadjuvant treatment , 2020, bioRxiv.

[39]  A. Regev,et al.  The Human and Mouse Enteric Nervous System at Single-Cell Resolution , 2020, Cell.

[40]  C. Diorio,et al.  The Importance of Breast Adipose Tissue in Breast Cancer , 2020, International journal of molecular sciences.

[41]  Aviv Regev,et al.  Cumulus provides cloud-based data analysis for large-scale single-cell and single-nucleus RNA-seq , 2020, Nature Methods.

[42]  M. Wilkinson,et al.  Transcriptome profiling reveals signaling conditions dictating human spermatogonia fate in vitro , 2020, Proceedings of the National Academy of Sciences.

[43]  Aviv Regev,et al.  Chromatin Potential Identified by Shared Single-Cell Profiling of RNA and Chromatin , 2020, Cell.

[44]  I. Amit,et al.  The Physiology, Pathology, and Potential Therapeutic Applications of the TREM2 Signaling Pathway , 2020, Cell.

[45]  M Dugas,et al.  Benchmarking atlas-level data integration in single-cell genomics , 2020, Nature Methods.

[46]  G. Trynka,et al.  From GWAS to Function: Using Functional Genomics to Identify the Mechanisms Underlying Complex Diseases , 2020, Frontiers in Genetics.

[47]  Daniel J. Gaffney,et al.  Souporcell: robust clustering of single-cell RNA-seq data by genotype without reference genotypes , 2020, Nature Methods.

[48]  Michael T. Garcia,et al.  Single-cell resolution analysis of the human pancreatic ductal progenitor cell niche , 2020, Proceedings of the National Academy of Sciences.

[49]  M. Mildner,et al.  Single-cell transcriptomics combined with interstitial fluid proteomics defines cell-type-specific immune regulation in atopic dermatitis. , 2020, The Journal of allergy and clinical immunology.

[50]  A. Akalin,et al.  Single-nucleus transcriptomics reveals functional compartmentalization in syncytial skeletal muscle cells , 2020, Nature Communications.

[51]  M. Weirauch,et al.  Single-nucleus RNA-seq identifies transcriptional heterogeneity in multinucleated skeletal myofibers , 2020, Nature Communications.

[52]  Åsa K. Björklund,et al.  Characterization of the nuclear and cytosolic transcriptomes in human brain tissue reveals new insights into the subcellular distribution of RNA transcripts , 2020, Scientific Reports.

[53]  G. Frühbeck,et al.  Dermatopontin, A Novel Adipokine Promoting Adipose Tissue Extracellular Matrix Remodelling and Inflammation in Obesity , 2020, Journal of clinical medicine.

[54]  M. Ataollahi,et al.  Decreased serum levels of IL-27and IL-35 in patients with Graves disease. , 2020, Archives of endocrinology and metabolism.

[55]  B. Wessner,et al.  Skeletal Muscle Extracellular Matrix – What Do We Know About Its Composition, Regulation, and Physiological Roles? A Narrative Review , 2020, Frontiers in Physiology.

[56]  Alexander Yang,et al.  Adipocyte lipolysis: from molecular mechanisms of regulation to disease and therapeutics. , 2020, The Biochemical journal.

[57]  Matthias Heinig,et al.  The single-cell eQTLGen consortium , 2020, eLife.

[58]  A. Torkamani,et al.  Decoding the transcriptome of atherosclerotic plaque at single-cell resolution , 2020, bioRxiv.

[59]  Mirjana Efremova,et al.  CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes , 2020, Nature Protocols.

[60]  Christian H. Holland,et al.  Robustness and applicability of transcription factor and pathway analysis tools on single-cell RNA-seq data , 2020, Genome Biology.

[61]  L. Willemsen,et al.  Macrophage subsets in atherosclerosis as defined by single‐cell technologies , 2020, The Journal of pathology.

[62]  O. Wolkenhauer,et al.  Single-Nucleus Sequencing of an Entire Mammalian Heart: Cell Type Composition and Velocity , 2020, Cells.

[63]  O. Elemento,et al.  A reference single-cell transcriptomic atlas of human skeletal muscle tissue reveals bifurcated muscle stem cell populations , 2020, bioRxiv.

[64]  Aliza B. Rubenstein,et al.  Single-cell transcriptional profiles in human skeletal muscle , 2020, Scientific Reports.

[65]  K. Ardlie,et al.  Transcriptional and Cellular Diversity of the Human Heart , 2020, bioRxiv.

[66]  Z. Mo,et al.  Single-cell RNA sequencing of human kidney , 2020, Scientific Data.

[67]  T. Lassmann,et al.  Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows , 2020, Genome Biology.

[68]  Christopher D. Brown,et al.  The GTEx Consortium atlas of genetic regulatory effects across human tissues , 2019, Science.

[69]  Kerstin B. Meyer,et al.  BBKNN: fast batch alignment of single cell transcriptomes , 2019, Bioinform..

[70]  Enrico Petretto,et al.  A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation , 2019, Nature Neuroscience.

[71]  John C. Marioni,et al.  Unsupervised removal of systematic background noise from droplet-based single-cell experiments using CellBender , 2019, bioRxiv.

[72]  Randall J. Platt,et al.  Mapping human cell phenotypes to genotypes with single-cell genomics , 2019, Science.

[73]  Aviv Regev,et al.  A single-cell and single-nucleus RNA-Seq toolbox for fresh and frozen human tumors , 2019, Nature Medicine.

[74]  C. Ponting,et al.  Resolving the fibrotic niche of human liver cirrhosis at single cell level , 2019, Nature.

[75]  Irving L. Weissman,et al.  A molecular cell atlas of the human lung from single cell RNA sequencing , 2019, Nature.

[76]  Jiandie D. Lin,et al.  Landscape of Intercellular Crosstalk in Healthy and NASH Liver Revealed by Single-Cell Secretome Gene Analysis. , 2019, Molecular cell.

[77]  D. Beeson,et al.  The congenital myasthenic syndromes: expanding genetic and phenotypic spectrums and refining treatment strategies , 2019, Current opinion in neurology.

[78]  I. Amit,et al.  Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner , 2019, Cell.

[79]  Aviv Regev,et al.  Intra- and Inter-cellular Rewiring of the Human Colon during Ulcerative Colitis , 2019, Cell.

[80]  Benjamin J. Strober,et al.  Dynamic genetic regulation of gene expression during cellular differentiation , 2019, Science.

[81]  Masato Hoshi,et al.  A single-nucleus RNA-sequencing pipeline to decipher the molecular anatomy and pathophysiology of human kidneys , 2019, Nature Communications.

[82]  A. Shilatifard,et al.  Single-Cell Transcriptomic Analysis of Human Lung Provides Insights into the Pathobiology of Pulmonary Fibrosis , 2019, American journal of respiratory and critical care medicine.

[83]  Pere Roca-Cusachs,et al.  Integrins as biomechanical sensors of the microenvironment , 2019, Nature Reviews Molecular Cell Biology.

[84]  Clint L. Miller,et al.  Atheroprotective roles of smooth muscle cell phenotypic modulation and the TCF21 disease gene as revealed by single-cell analysis , 2019, Nature Medicine.

[85]  Christopher A. Lazarski,et al.  Fibroadipogenic progenitors are responsible for muscle loss in limb girdle muscular dystrophy 2B , 2019, Nature Communications.

[86]  Fabian J Theis,et al.  Current best practices in single‐cell RNA‐seq analysis: a tutorial , 2019, Molecular systems biology.

[87]  Davis J. McCarthy,et al.  Single-cell RNA-sequencing of differentiating iPS cells reveals dynamic genetic effects on gene expression , 2019, bioRxiv.

[88]  Y. Bossé,et al.  Benefits and limitations of genome-wide association studies , 2019, Nature Reviews Genetics.

[89]  Manolis Kellis,et al.  Single-cell transcriptomic analysis of Alzheimer’s disease , 2019, Nature.

[90]  F. Ginhoux,et al.  Two distinct interstitial macrophage populations coexist across tissues in specific subtissular niches , 2019, Science.

[91]  L. Coin,et al.  Genotype-free demultiplexing of pooled single-cell RNA-seq , 2019, Genome Biology.

[92]  Michael Brudno,et al.  Expanding the Boundaries of RNA Sequencing as a Diagnostic Tool for Rare Mendelian Disease , 2019, American journal of human genetics.

[93]  J. Dulak,et al.  Targeting angiogenesis in Duchenne muscular dystrophy , 2019, Cellular and Molecular Life Sciences.

[94]  R. Mirsky,et al.  The Success and Failure of the Schwann Cell Response to Nerve Injury , 2019, Front. Cell. Neurosci..

[95]  M. Mills,et al.  A scientometric review of genome-wide association studies , 2019, Communications Biology.

[96]  Haojia Wu,et al.  Advantages of Single-Nucleus over Single-Cell RNA Sequencing of Adult Kidney: Rare Cell Types and Novel Cell States Revealed in Fibrosis. , 2018, Journal of the American Society of Nephrology : JASN.

[97]  Alan F. Scott,et al.  OMIM.org: leveraging knowledge across phenotype–gene relationships , 2018, Nucleic Acids Res..

[98]  H. Esterbauer,et al.  Non-parenchymal TREM-2 protects the liver from immune-mediated hepatocellular damage , 2018, Gut.

[99]  Gerome Breen,et al.  Genetic identification of brain cell types underlying schizophrenia , 2017, Nature Genetics.

[100]  Sarah A Teichmann,et al.  A test metric for assessing single-cell RNA-seq batch correction , 2018, Nature Methods.

[101]  Fan Zhang,et al.  Fast, sensitive, and accurate integration of single cell data with Harmony , 2018, bioRxiv.

[102]  Kerstin B. Meyer,et al.  Single-cell reconstruction of the early maternal–fetal interface in humans , 2018, Nature.

[103]  Gary D Bader,et al.  Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations , 2018, Nature Communications.

[104]  K. Alitalo,et al.  Cardiac lymphatics in health and disease , 2018, Nature Reviews Cardiology.

[105]  Venkat S. Malladi,et al.  A Cellular Anatomy of the Normal Adult Human Prostate and Prostatic Urethra , 2018, bioRxiv.

[106]  S. Powers,et al.  The Renin-Angiotensin System and Skeletal Muscle. , 2018, Exercise and sport sciences reviews.

[107]  Burkhard Tümmler,et al.  Faculty Opinions recommendation of A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. , 2018, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[108]  Daniel J. Garry,et al.  Integrative effects of dystrophin loss on metabolic function of the mdx mouse , 2018, Scientific Reports.

[109]  J. Han Faculty Opinions recommendation of Single-cell epigenomics: Recording the past and predicting the future. , 2018, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[110]  Aviv Regev,et al.  A revised airway epithelial hierarchy includes CFTR-expressing ionocytes , 2018, Nature.

[111]  Jun Kit Wang,et al.  Hyaluronan Receptor LYVE‐1‐Expressing Macrophages Maintain Arterial Tone through Hyaluronan‐Mediated Regulation of Smooth Muscle Cell Collagen , 2018, Immunity.

[112]  S. Potter,et al.  Single-cell RNA sequencing for the study of development, physiology and disease , 2018, Nature Reviews Nephrology.

[113]  G. Booz,et al.  Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. , 2018, Physiological reviews.

[114]  Ayellet V. Segrè,et al.  Using an atlas of gene regulation across 44 human tissues to inform complex disease- and trait-associated variation , 2018, Nature Genetics.

[115]  Allon M. Klein,et al.  A single cell atlas of the tracheal epithelium reveals the CFTR-rich pulmonary ionocyte , 2018, Nature.

[116]  T. Einarson,et al.  Prevalence of cardiovascular disease in type 2 diabetes: a systematic literature review of scientific evidence from across the world in 2007–2017 , 2018, Cardiovascular Diabetology.

[117]  J. Sáez-Rodríguez,et al.  Benchmark and integration of resources for the estimation of human transcription factor activities , 2018, bioRxiv.

[118]  Matthew D. Young,et al.  SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data , 2018, bioRxiv.

[119]  Laleh Haghverdi,et al.  Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors , 2018, Nature Biotechnology.

[120]  Dennis Wolf,et al.  Single-Cell RNA-Seq Reveals the Transcriptional Landscape and Heterogeneity of Aortic Macrophages in Murine Atherosclerosis , 2018, Circulation research.

[121]  M. G. van der Wijst,et al.  Single-cell RNA sequencing identifies cell type-specific cis-eQTLs and co-expression QTLs , 2018, Nature Genetics.

[122]  H. Hammad,et al.  Langerhans Cells: Sensing the Environment in Health and Disease , 2018, Front. Immunol..

[123]  Chun Jimmie Ye,et al.  Multiplexed droplet single-cell RNA-sequencing using natural genetic variation , 2017, Nature Biotechnology.

[124]  Tiago P. Peixoto,et al.  A network approach to topic models , 2017, Science Advances.

[125]  Fabian J Theis,et al.  SCANPY: large-scale single-cell gene expression data analysis , 2018, Genome Biology.

[126]  Nicola J. Rinaldi,et al.  Genetic effects on gene expression across human tissues , 2017, Nature.

[127]  O. Stegle,et al.  Single-cell epigenomics: Recording the past and predicting the future , 2017, Science.

[128]  V. Olkkonen,et al.  Angiopoietin-like 8 (Angptl8) controls adipocyte lipolysis and phospholipid composition. , 2017, Chemistry and physics of lipids.

[129]  A. van Oudenaarden,et al.  Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations , 2017, Nature Methods.

[130]  Aviv Regev,et al.  Massively-parallel single nucleus RNA-seq with DroNc-seq , 2017, Nature Methods.

[131]  Amit A. Patel,et al.  The fate and lifespan of human monocyte subsets in steady state and systemic inflammation , 2017, The Journal of experimental medicine.

[132]  I. Amit,et al.  A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease , 2017, Cell.

[133]  Paul Flicek,et al.  Alignment of 1000 Genomes Project reads to reference assembly GRCh38 , 2017, GigaScience.

[134]  A. Rump,et al.  New gain-of-function mutation shows CACNA1D as recurrently mutated gene in autism spectrum disorders and epilepsy , 2017, Human molecular genetics.

[135]  Funded Statistical Methods groups-AWG,et al.  Improving genetic diagnosis in Mendelian disease with transcriptome sequencing , 2017 .

[136]  N. Hacohen,et al.  Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors , 2017, Science.

[137]  J. Knight,et al.  Pathogenic implications for autoimmune mechanisms derived by comparative eQTL analysis of CD4+ versus CD8+ T cells , 2017, PLoS genetics.

[138]  Rui B Chang,et al.  Piezo2 senses airway stretch and mediates lung inflation-induced apnoea , 2016, Nature.

[139]  A. Hofman,et al.  Identification of context-dependent expression quantitative trait loci in whole blood , 2016, Nature Genetics.

[140]  B. Wirth,et al.  Biallelic Loss of Proprioception-Related PIEZO2 Causes Muscular Atrophy with Perinatal Respiratory Distress, Arthrogryposis, and Scoliosis. , 2016, American journal of human genetics.

[141]  X. Wen,et al.  Integrating molecular QTL data into genome-wide genetic association analysis: Probabilistic assessment of enrichment and colocalization , 2016, bioRxiv.

[142]  Cynthia C. Hession,et al.  Div-Seq: Single-nucleus RNA-Seq reveals dynamics of rare adult newborn neurons , 2016, Science.

[143]  K. Inaba,et al.  Sperm dysfunction and ciliopathy , 2016, Reproductive medicine and biology.

[144]  M. Çakmak,et al.  Role of interlekin-35 as a biomarker in patients with newly diagnosed Hashimoto’s thyroiditis , 2016, Endocrine regulations.

[145]  Ellen T. Gelfand,et al.  A Novel Approach to High-Quality Postmortem Tissue Procurement: The GTEx Project , 2015, Biopreservation and biobanking.

[146]  Yakir A Reshef,et al.  Partitioning heritability by functional annotation using genome-wide association summary statistics , 2015, Nature Genetics.

[147]  I. Amit,et al.  Distinct Murine Mucosal Langerhans Cell Subsets Develop from Pre-dendritic Cells and Monocytes. , 2015, Immunity.

[148]  F. Rossi,et al.  Nilotinib reduces muscle fibrosis in chronic muscle injury by promoting TNF-mediated apoptosis of fibro/adipogenic progenitors , 2015, Nature Medicine.

[149]  R. Ransohoff,et al.  Nuclear Receptors License Phagocytosis by Trem2+ Myeloid Cells in Mouse Models of Alzheimer's Disease , 2015, The Journal of Neuroscience.

[150]  Steffen Jung,et al.  Macrophages: development and tissue specialization. , 2015, Annual review of immunology.

[151]  Ivana Y. Kuo,et al.  Signaling in muscle contraction. , 2015, Cold Spring Harbor perspectives in biology.

[152]  N. Friedman,et al.  High-Resolution Sequencing and Modeling Identifies Distinct Dynamic RNA Regulatory Strategies , 2014, Cell.

[153]  Chun Jimmie Ye,et al.  Intersection of population variation and autoimmunity genetics in human T cell activation , 2014, Science.

[154]  Xiuli Zuo,et al.  The Possible Role of the Novel Cytokines IL-35 and IL-37 in Inflammatory Bowel Disease , 2014, Mediators of inflammation.

[155]  L. Waddell,et al.  Recessive myosin myopathy with external ophthalmoplegia associated with MYH2 mutations , 2013, European Journal of Human Genetics.

[156]  Milena B. Furtado,et al.  Cardiogenic Genes Expressed in Cardiac Fibroblasts Contribute to Heart Development and Repair , 2014, Circulation research.

[157]  Je-Yoel Cho,et al.  ELK3 Suppresses Angiogenesis by Inhibiting the Transcriptional Activity of ETS-1 on MT1-MMP , 2014, International journal of biological sciences.

[158]  A. Heinzmann,et al.  Association of a FGFR-4 Gene Polymorphism with Bronchopulmonary Dysplasia and Neonatal Respiratory Distress , 2013, Disease markers.

[159]  J. Beckman,et al.  Diabetes and vascular disease: pathophysiology, clinical consequences, and medical therapy: part I , 2013, European heart journal.

[160]  C. L. Liang,et al.  Neuromuscular Junction Disorders , 2013, PM & R : the journal of injury, function, and rehabilitation.

[161]  R. Toes,et al.  Dysferlin Regulates Cell Adhesion in Human Monocytes* , 2013, The Journal of Biological Chemistry.

[162]  Comparison of total and cytoplasmic mRNA reveals global regulation by nuclear retention and miRNAs , 2012, BMC Genomics.

[163]  S. Bensinger,et al.  Liver X receptor and peroxisome proliferator‐activated receptor as integrators of lipid homeostasis and immunity , 2012, Immunological reviews.

[164]  K. Sekiguchi,et al.  Basement membrane assembly of the integrin α8β1 ligand nephronectin requires Fraser syndrome–associated proteins , 2012, The Journal of cell biology.

[165]  Andrew E. Jaffe,et al.  Bioinformatics Applications Note Gene Expression the Sva Package for Removing Batch Effects and Other Unwanted Variation in High-throughput Experiments , 2022 .

[166]  David G. Knowles,et al.  Fast Computation and Applications of Genome Mappability , 2012, PloS one.

[167]  W. Dolen,et al.  Allergy and the Allergic Diseases , 2012 .

[168]  N. Clarke Congenital fiber-type disproportion. , 2011, Seminars in pediatric neurology.

[169]  R. Lieber,et al.  Structure and function of the skeletal muscle extracellular matrix , 2011, Muscle & nerve.

[170]  C. Bönnemann The collagen VI-related myopathies: muscle meets its matrix , 2011, Nature Reviews Neurology.

[171]  C. Kimmel,et al.  Genetic Analysis of Fin Development in Zebrafish Identifies Furin and Hemicentin1 as Potential Novel Fraser Syndrome Disease Genes , 2010, PLoS genetics.

[172]  B. Halloran,et al.  The role of integrin alpha8beta1 in fetal lung morphogenesis and injury. , 2009, Developmental biology.

[173]  F. Alkuraya,et al.  FREM1 mutations cause bifid nose, renal agenesis, and anorectal malformations syndrome. , 2009, American journal of human genetics.

[174]  J. M. Aerts,et al.  Increased chitotriosidase activity in serum of leprosy patients: association with bacillary leprosy. , 2009, Clinical immunology.

[175]  K. Sekiguchi,et al.  Molecular Basis of the Recognition of Nephronectin by Integrin α8β1* , 2009, Journal of Biological Chemistry.

[176]  S. Hughes,et al.  Nerve‐dependent changes in skeletal muscle myosin heavy chain after experimental denervation and cross‐reinnervation and in a demyelinating mouse model of Charcot–Marie–Tooth disease type 1A , 2008, Muscle & nerve.

[177]  G. Bernardi,et al.  Preferential central nucleation of type 2 myofibers is an invariable feature of myotonic dystrophy type 2 , 2008, Muscle & nerve.

[178]  Stephen J. Galli,et al.  The development of allergic inflammation , 2008, Nature.

[179]  R. Vossen,et al.  Generation and Characterization of Transgenic Mice with the Full-length Human DMD Gene* , 2008, Journal of Biological Chemistry.

[180]  M. Zenker,et al.  Fraser syndrome due to homozygosity for a splice site mutation of FREM2 , 2008, American journal of medical genetics. Part A.

[181]  G. Narkis,et al.  Lethal congenital contractural syndrome type 2 (LCCS2) is caused by a mutation in ERBB3 (Her3), a modulator of the phosphatidylinositol-3-kinase/Akt pathway. , 2007, American journal of human genetics.

[182]  L. Reichardt,et al.  The ECM protein nephronectin promotes kidney development via integrinα 8β1-mediated stimulation of Gdnf expression , 2007, Development.

[183]  K. Sekiguchi,et al.  Breakdown of the reciprocal stabilization of QBRICK/Frem1, Fras1, and Frem2 at the basement membrane provokes Fraser syndrome-like defects , 2006, Proceedings of the National Academy of Sciences.

[184]  A. Yamauchi,et al.  Galectin-9 Induces Maturation of Human Monocyte-Derived Dendritic Cells , 2005, The Journal of Immunology.

[185]  G. Chalepakis,et al.  Basement Membrane Distortions Impair Lung Lobation and Capillary Organization in the Mouse Model for Fraser Syndrome* , 2005, Journal of Biological Chemistry.

[186]  M. Konishi,et al.  Fgf18 is required for embryonic lung alveolar development. , 2004, Biochemical and biophysical research communications.

[187]  S. Pikkarainen,et al.  GATA transcription factors in the developing and adult heart. , 2004, Cardiovascular research.

[188]  J. Beckman,et al.  Diabetes and vascular disease: pathophysiology, clinical consequences, and medical therapy: part I , 2013, European heart journal.

[189]  A. Paetau,et al.  Histopathological differences of myotonic dystrophy type 1 (DM1) and PROMM/DM2 , 2003, Neurology.

[190]  N. Prescott,et al.  Fraser syndrome and mouse blebbed phenotype caused by mutations in FRAS1/Fras1 encoding a putative extracellular matrix protein , 2003, Nature Genetics.

[191]  P. Scambler,et al.  Fras1 deficiency results in cryptophthalmos, renal agenesis and blebbed phenotype in mice , 2003, Nature Genetics.

[192]  Jiandie D. Lin,et al.  Transcriptional co-activator PGC-1α drives the formation of slow-twitch muscle fibres , 2002, Nature.

[193]  L. Reichardt,et al.  Identification and characterization of a novel extracellular matrix protein nephronectin that is associated with integrin α8β1 in the embryonic kidney , 2001, The Journal of cell biology.

[194]  A B Kay,et al.  Allergy and allergic diseases. First of two parts. , 2001, The New England journal of medicine.

[195]  G. D. Thomas,et al.  Functional muscle ischemia in neuronal nitric oxide synthase-deficient skeletal muscle of children with Duchenne muscular dystrophy. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[196]  W. Frontera,et al.  Contractile properties of single muscle fibers in myotonic dystrophy , 2000, Muscle & nerve.

[197]  C. Deng,et al.  FGFR-3 and FGFR-4 function cooperatively to direct alveogenesis in the murine lung. , 1998, Development.

[198]  R. Pedersen,et al.  Integrin α8β1 Is Critically Important for Epithelial–Mesenchymal Interactions during Kidney Morphogenesis , 1997, Cell.

[199]  O. Ibraghimov-Beskrovnaya,et al.  Primary structure of dystrophin-associated glycoproteins linking dystrophin to the extracellular matrix , 1992, Nature.