12
The Human Lung Cell Atlas: A High-Resolution Reference Map of the Human Lung in Health and Disease The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Schiller, Herbert B. et al. “The Human Lung Cell Atlas: A High- Resolution Reference Map of the Human Lung in Health and Disease.” American journal of respiratory cell and molecular biology 61 (2019): 31-41 © 2019 The Author(s) As Published 10.1165/rcmb.2018-0416tr Publisher American Thoracic Society Version Final published version Citable link https://hdl.handle.net/1721.1/125014 Terms of Use Creative Commons Attribution-NonCommercial-NoDerivs License Detailed Terms http://creativecommons.org/licenses/by-nc-nd/4.0/

The Human Lung Cell Atlas: A High-Resolution Reference Map

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The Human Lung Cell Atlas: A High-Resolution Reference Map

The Human Lung Cell Atlas: A High-ResolutionReference Map of the Human Lung in Health and Disease

The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters.

Citation Schiller, Herbert B. et al. “The Human Lung Cell Atlas: A High-Resolution Reference Map of the Human Lung in Health andDisease.” American journal of respiratory cell and molecular biology61 (2019): 31-41 © 2019 The Author(s)

As Published 10.1165/rcmb.2018-0416tr

Publisher American Thoracic Society

Version Final published version

Citable link https://hdl.handle.net/1721.1/125014

Terms of Use Creative Commons Attribution-NonCommercial-NoDerivs License

Detailed Terms http://creativecommons.org/licenses/by-nc-nd/4.0/

Page 2: The Human Lung Cell Atlas: A High-Resolution Reference Map

TRANSLATIONAL REVIEW

The Human Lung Cell Atlas: A High-Resolution Reference Map of theHuman Lung in Health and DiseaseHerbert B. Schiller1, Daniel T. Montoro2,3, Lukas M. Simon4, Emma L. Rawlins5, Kerstin B. Meyer6, Maximilian Strunz1,Felipe A. Vieira Braga6, Wim Timens7,8, Gerard H. Koppelman8,9, G. R. Scott Budinger10, Janette K. Burgess7,8,Avinash Waghray2,3, Maarten van den Berge8,11, Fabian J. Theis4,12, Aviv Regev13,14, Naftali Kaminski15,Jayaraj Rajagopal2,3, Sarah A. Teichmann6, Alexander V. Misharin10*, and Martijn C. Nawijn7,8*1Helmholtz Zentrum Munchen, Institute of Lung Biology and Disease, Group Systems Medicine of Chronic Lung Disease, Member of theGerman Center for Lung Research (DZL), Munich, Germany; 2Harvard Stem Cell Institute, Cambridge, Massachusetts; 3Center forRegenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts; 4Helmholtz Zentrum Munchen, German ResearchCenter for Environmental Health, Institute of Computational Biology, Neuherberg, Germany; 5Wellcome Trust/Cancer Research UKGurdon Institute, University of Cambridge, Cambridge, United Kingdom; 6Wellcome Sanger Institute, Hinxton, Cambridge, UnitedKingdom; 7Department of Pathology and Medical Biology, 9Department of Pediatric Pulmonology and Pediatric Allergology, BeatrixChildren’s Hospital, and 11Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen,the Netherlands; 8Groningen Research Institute for Asthma and COPD at the University of Groningen, University Medical CenterGroningen, Groningen, the Netherlands; 10Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois;12Department of Mathematics, Technische Universitat Munchen, Munich, Germany; 13Klarman Cell Observatory, Broad Instituteof Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts; 14Department of Biology, Howard Hughes MedicalInstitute and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts;and 15Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut

ORCID IDs: 0000-0002-6222-2149 (D.T.M.); 0000-0001-7426-3792 (E.L.R.); 0000-0001-5906-1498 (K.B.M.); 0000-0002-4146-6363 (W.T.);0000-0001-8567-3252 (G.H.K.); 0000-0003-3372-6521 (M.C.N.).

Abstract

Lung disease accounts for every sixth death globally. Profiling themolecular state of all lung cell types in health anddisease is currentlyrevolutionizing the identification of disease mechanisms and willaid the design of novel diagnostic and personalized therapeuticregimens. Recent progress in high-throughput techniques forsingle-cell genomic and transcriptomic analyses has openedup new possibilities to study individual cells within a tissue,classify these into cell types, and characterize variations intheir molecular profiles as a function of genetics, environment,cell–cell interactions, developmental processes, aging, or

disease. Integration of these cell state definitions with spatialinformation allows the in-depth molecular description of cellularneighborhoods and tissue microenvironments, including the tissueresident structural and immune cells, the tissue matrix,and the microbiome. The Human Cell Atlas consortium aims tocharacterize all cells in the healthy human body and has prioritizedlung tissue as one of the flagship projects. Here, we present therationale, the approach, and the expected impact of a Human LungCell Atlas.

Keywords: Human Cell Atlas; single-cell RNA sequencing; spatialtranscriptomics; systems biology

(Received in original form December 21, 2018; accepted in final form April 17, 2019 )

This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0(http://creativecommons.org/licenses/by-nc-nd/4.0/). For commercial usage and reprints, please contact Diane Gern ([email protected]).

*Shared senior authors.

Supported by the Helmholtz Association and the German Center for Lung Research (DZL) (H.B.S.); the European Union’s Horizon 2020 research andinnovation programme under the Marie Sklodowska-Curie grant agreement 753039 (L.M.S.); U.K. Medical Research Council grant G0900424 (E.L.R.);National Institutes of Health (NIH) grants ES013995, HL071643, and AG049665, and Veterans Administration grant BX000201 and Department of Defensegrant PR141319 (G.R.S.B.); NIH grants HL135124 and AI135964 and Department of Defense grant PR141319 (A.V.M.); NIH grants R01HL141852,R01HL127349, UHHL3123886, U01HL122626, and UG3TR002445, and Department of Defence grant PR151124 (N.K.); and the Netherlands LungFoundation grants 5.1.14.020 and 4.1.18.226 (M.C.N.).

Author Contributions: Conception and outline: H.B.S., A.R., N.K., J.R., S.A.T., A.V.M., and M.C.N.; drafting the manuscript for important intellectual content:all authors.

Correspondence and requests for reprints should be addressed to Martijn C. Nawijn, Ph.D., University Medical Center Groningen–GRIAC Research Institute,Department of Pathology and Medical Biology, Hanzeplein 1, Groningen NL-9700-RB, the Netherlands. E-mail: [email protected].

Am J Respir Cell Mol Biol Vol 61, Iss 1, pp 31–41, Jul 2019

Copyright © 2019 by the American Thoracic Society

Originally Published in Press as DOI: 10.1165/rcmb.2018-0416TR on April 17, 2019

Internet address: www.atsjournals.org

Translational Review 31

Page 3: The Human Lung Cell Atlas: A High-Resolution Reference Map

Lung disease is a leading cause of mortalityin the United States and worldwide, withmore than 7 million deaths attributed tolung disease annually (1). Although the lunghas been reported to harbor at least40 discrete cell types (2), the recentidentification of the ionocyte as a novelepithelial cell type in human airway wall(Figure 1) shows that our knowledge of thecells in human lung is incomplete (3, 4).Therefore, further studies to systematicallyand comprehensively characterize all celltypes in human lung tissue and the changesin cellular composition and functionassociated with initiation, progression, andresolution of human lung disease areurgently needed.

The cell is the fundamental unit of allliving organisms (5). The cumulativefunction of all cells and their interactionswith each other and with noncellulartissue components determines thephysiology of any tissue or organ. Diseaseis believed to be a consequence of cell-intrinsic changes, changes in response toenvironmental insults, altered cell–cellcommunication, disbalance of cell typeproportions, and/or perturbed architecture

of tissues. Recent progress in single-cellanalyses and accompanying computationalmethodologies provides an extraordinaryopportunity to characterize individual cellsand their spatial organization on the basisof their RNA or protein expression profile,allowing de novo classification of cell typesand a comprehensive description of theirdynamic phenotype. Combining these withrapidly evolving spatially resolving methodsfor the analysis of DNA, RNA, or proteinprofiles allows the interpretation orannotation of the cellular heterogeneity in atissue context.

The Human Cell Atlas (HCA)Consortium is an international,collaborative effort aiming to define allhuman cell types in terms of their distinctivepatterns of gene expression, physiologicalstates, developmental trajectories, andspatial relationships in tissue (6). As part ofthis overarching effort, we intend to build acomprehensive atlas of human lung in astaged and integrated approach. Molecularprofiles of single cells will be mappedcomputationally on a common coordinateframework of the entire organ in relation tospatial landmarks of both micro- and

macro-anatomy, starting from thehistological structure of cell neighborhoods,building up layers of increasingly largerunits of spatial organization, up to the fullorgan.

The Lung Cell Atlas will revealunprecedented insights about the identities,activities, and lineage relationships of all cells inhealthy human lung, enabling the modelingof lung homeostatic circuitry (6). The LungCell Atlas will then serve as a reference pointfor the analysis of diseased lung tissue atsingle-cell resolution and will allowidentification of the shifts in cellular repertoire,the changes in cellular states and phenotypes,and the altered cell–cell interactions thatdisrupt normal lung homeostasis andconstitute disease. Comparing lung diseasedata with the lung development referencewithin the Human Lung Cell Atlas may revealwhether cellular changes in certain chroniclung diseases represent a failed recapitulationof organogenesis or the acquisition of entirelynew pathologic programs governing cell fateand behavior. Thus, the generation of a high-resolution, comprehensive catalog of thechanges in lung cellular composition andfunction in health and disease is expected to

CFTR-high

FOXI1+ Pulmonary lonocyte

Figure 1. The discovery of the pulmonary ionocyte. The first systematic analysis of the diversity of epithelial cells lining the airways by single-celltechnology was recently reported in the mouse (3). In addition to finding an unexpected diversity of club, tuft, and goblet cells in the murine airway, a newairway epithelial cell type was discovered. This novel cell type specifically expresses the transcription factor Foxi1, and its composite gene expressionprofile resembles those of specialized ion-transporting cells in fish gills, frog skin, and mammalian kidney. As these diverse cell types are collectivelyreferred to as ionocytes, the new pulmonary epithelial cell has been coined a “pulmonary ionocyte.” Surprisingly, these cells also expressed the majority ofCftr (cystic fibrosis transmembrane conductance regulator gene), whose mutation causes cystic fibrosis (CF). The investigators found that deletion of Foxi1in mice resulted in the loss of mature ionocytes and significantly altered mucus viscosity. FOXI11 pulmonary ionocytes were also found to line the airwaysof human lung and express high levels of CFTR. Their role in ion transport, regulation of mucus, and high CFTR expression suggests that pulmonaryionocytes play a critical role in CF biology and disease.

TRANSLATIONAL REVIEW

32 American Journal of Respiratory Cell and Molecular Biology Volume 61 Number 1 | July 2019

Page 4: The Human Lung Cell Atlas: A High-Resolution Reference Map

lead to development of novel cell- and disease-specific biomarkers and advancements intherapeutic strategies for lung disease.

Cellular and AnatomicalComplexity of the HumanLung

The primary function of lung tissue—gas exchange between the body andits environment—is dependent on aspecialized anatomy involving numerousdistinct epithelial and endothelial cellpopulations, supported by specific tissue-resident leukocyte populations, and variousmesenchymal cell types providingstructural support. The unique anatomy ofthe lung is established during developmentby branching morphogenesis resulting in atree of airways and alveoli, mirrored withtrees of blood and lymphatic vessels. Airflows from the nasal cavity into theconducting airways, involving trachea andthe main bronchi, which iterativelybifurcate into the branching bronchial tree.The human trachea and airways arecomposed of a pseudostratified epitheliumon the luminal side, with a basal laminalined with mesenchyme, cartilage, smoothmuscle, blood vessels, immune cells, andother rare cell types (7). The airways end interminal bronchioles, which then lead intocomposite respiratory units including therespiratory bronchioles, the alveolar duct,and the alveolar sac, where gas exchangeoccurs (7). The airways serve as a conduitfor gases from the atmosphere to the alveolibut also have a role in filtering the inhaledair for solid particles and removing thesein a process called mucociliary clearance,thereby protecting the distal saccularstructures of the alveoli. Gas exchangehappens in the alveolar unit, featuringultrathin type-1 pneumocytes wrapped by alayer of capillaries, supported by interstitialcells such as matrix-producing fibroblastsand the pulmonary surfactant layer (8).Type-2 pneumocytes produce and secretesurfactant, maintain the fluid balance of thealveolar unit, and serve as local facultativeprogenitor cells for type-1 cells (9–11).Resident immune cells, particularly alveolarmacrophages and peribronchial andperivascular “interstitial” macrophages,play important roles in orchestration of theimmune response and in the maintenanceof lung homeostasis (12, 13). Alveolarmacrophages and alveolar epithelial cells

express reciprocal sets of ligands andreceptors, which prevent an excessiveinflammatory response to inhaled particles.Several studies in mice have demonstratedthat peribronchial macrophages consist ofmultiple ontologically and anatomicallydistinct subsets (14); however, little isknown about their human counterparts.

Single-cell molecular studies profiledfrom a comprehensive sampling of humanlungs will empower the mapping of allpossible cellular identities (or states) onto aframework of a high-dimensionalphenotypic cellular (gene and proteinexpression) landscape. Such a phenotypiclandscape will describe cell type identity asstable intermediates within a continuumof possible states, including lineagerelationships and potential transitory pathsbetween different states. The power of thisapproach is elegantly shown in a recentreview on the cellular and molecularpathways involved in lung organogenesis,where single-cell datasets from mouse lungdevelopment are used to identify and mapcellular trajectories during the differentstages of organogenesis (15). The excitingrecent demonstration of genetic lineagetracing in human tissues usingmitochondrial DNA mutations will nowalso enable the combination of lineageinference with gene expression orchromatin state profiles in human tissues(16). Thus, the use of mitochondrial DNAmutations as natural genetic barcodes mayreveal the clonal architecture in humanlung health and disease. The multiplesnapshots of molecular identities presentalong the high-dimensional gene expressionlandscape can also result in the moleculardescription of unknown cellular states,which may have functions distinct from thetraditional cell type definitions currentlyrecognized. Similarly, we will likelyencounter gradients of cellular phenotypesalong spatial trajectories, as these“specialized” cellular states are driven bytheir microenvironment, as was recentlyreported for enterocytes in the mouseintestine (17).

All lung cells are contained within aregionally tailored and highly complexextracellular matrix that forms the basis ofvarious distinct microenvironments, whichis further defined by (the signals from) othercells in close proximity and potentially localvariations in the microbiome. The effect ofthe microenvironment on the molecularphenotype of a cell has not been explored

into great detail, although diseased humanlung extracellular matrix, for instance, hasbeen shown to drive a specific cellulartranscriptome (18). Whether the identity ofcells within the lung is determined by theirspecific location and function within thetissue and local microenvironment, by theirdevelopmental origin, or by a combinationof both, is one of the key questions for theHuman Lung Cell Atlas. Interestingly,it was recently reported that distinctdevelopmental lineages can produce thesame anatomical cell type by converging toa homogenous transcriptomic state inthe final cell divisions of development,indicating that distinct developmentallineages can assume very similar molecularphenotypes as a function of their localniche (19). Location-dependent variation inthe (molecular) state of cells in human lunghas not been studied in detail yet, althoughin the mouse model, club cells have beenshown to have a transcriptomic phenotypethat is dependent on their position alongthe respiratory tract (3). The relativefrequency of epithelial airway cell typesvaries along the respiratory tract (20).Submucosal glands are present fromtrachea to the small airways in human lung,but whether their composition is constantalong the respiratory tract remains to beestablished. Similarly, it is unknownwhether location within the lung affects themolecular phenotype of the cells that makeup the alveolar unit. A unique feature of thelung is that it is a constantly moving organ,with dynamic changes in pressure andshape leading to presumably similar cellsbeing exposed to very distinct mechanicalcues—apical versus basal, central versusperipheral—and the cellular transcriptomicadaptations to these variables are completelyunknown.

A complete atlas of the healthy humanlung must sample the entire coordinateframework of the organ, both macro- andmicro-anatomically, and capture the naturalvariation of cellular states within the healthypopulation (Figure 2). A comprehensivemap of molecular cell states and thepossible transitions between themwill provide mechanistic insight intodevelopmental processes and regenerativeand repair capacity in the lung and theirunderlying regulatory mechanisms. TheHuman Lung Cell Atlas will map thislandscape of molecular cell states onto aframework of spatial landmarks and enablethe direct comparison of samples between

TRANSLATIONAL REVIEW

Translational Review 33

Page 5: The Human Lung Cell Atlas: A High-Resolution Reference Map

multiple healthy and, eventually, diseasedindividuals.

A Cellular Reference Map ofHuman Lung Development

The process of building the lung structure,and maturing the cells it contains,occurs over many weeks of embryonicdevelopment and beyond in humans and isbelieved to require numerous differentcell–cell and cell–matrix interactions(15, 21). Many of these signaling andphysical interactions occur betweenmultiple progenitor cell types, which arespecific to a developmental time windowand do not exist in the adult organ. Theepithelium is the best-characterized partof the developing lung, and at least three,apparently embryonic-specific, progenitorcell types have been detected. For example,the budding tip of the developing humanlung epithelium was shown to be analogousto that of the mouse and to contain a majorepithelial progenitor population requiredduring branching morphogenesis (22, 23).However, there is no evidence thatsuch cells exist in the mature lung.Characterization of the development-specific, or possibly developmentallyenriched, progenitor cell states, and their(developmental) trajectories in thetranscriptomic landscape, within theHuman Cell Atlas Project will first andforemost provide insight into the basicmechanisms of lung development. Inaddition, it will provide specific andcritical data allowing long-standinghypotheses regarding the recapitulation ofdevelopmental mechanisms in repair anddisease states to be tested. Although theinitial focus of the Human Lung Cell Atlasis on adult lung, it will integrate data fromthe Pediatric Cell Atlas (24) and initiativesfocused on lung development, such asLungMAP or the recent National Institutesof Health (NIH)-funded DEVMAP.

Because of the progressive nature ofbranching morphogenesis, the developinglung epithelium contains a continuum ofcells along the proximal–distal axis inprogressive states of maturity frommultipotent progenitor to fully maturemucociliary, or alveolar, epithelium. This isparticularly significant given the low ratesof cell turnover in the adult organ (25)and means that analysis of embryoniclung development will allow states of

unsupervised clustering of cell states in lung parenchyma

AT2 cells

cells

genes

single cell multi-omics data (e.g., scRNA-seq, scATAC-seq)+ spatial transcriptomics (e.g., MERFISH, Slide-seq)

SPACE(anatomical

location)

Hierarchical approachto CELL TYPE IDENTITY

CELL STATE IDENTITY

TIME(development,perturbation,

age)

MODELS(gene regulation, upstreamregulators, receptor–ligand,

transcription factors,epigenetic marks)

UM

AP

2

con

stit

uti

ve

constitutivegene modules

facultativegene modules

facu

ltat

ive

Cryobiopsy 01Donor 01Donor 02Donor 03Donor 04Donor 05Donor 06Donor 07Donor 08

Donor 1

Donor 2

Donor 3

Donor 4

Donor 5

Donor 6

Donor 7

Donor 8

HP 01IPF 01IPF 02IPF 03IPF 04Myositis–ILD 01SSc–ILD 01

UM

AP

2

UMAP1

UMAP1

SFTPC (AT2)

A

B

SSc–ILD 02

Figure 2. Cell types and cell states in the human lung cell atlas. (A) SFTPC-positive alveolar type-2(AT2) cell clusters observed in lung parenchyma are shown in a dimension-reduced UMAP plot(left upper and lower panel). The heat map in the right panel shows constitutive genes defining AT2cell type identity across eight healthy donors and clusters of genes that have been found tosignificantly differ between AT2 cells of different individuals. Hence, the constant features definethe cell type, which can adopt multiple molecular phenotypes, or cell states, on the basis of thevariable features. Adapted from Reference 53. (B) Data collected in the Human Lung Cell Atlasproject will be used to define cell type identity in a hierarchical approach, which defines cell type bysimilarity metrics of their marker genes across many individuals. On the basis of these cell typeidentities and observed variation in facultative gene modules within the different cell types, we willgenerate predictive models of gene regulation that can be experimentally tested. These models willput (disease) genes in the context of their cell type–specific “gene regulatory environments” andpredict the appearance of both constitutive and facultative gene modules as a function of timeand space as occurring during lung development or regenerative and immune responses.HP = hypersensitivity pneumonitis; ILD = interstitial lung disease; IPF = idiopathic pulmonaryfibrosis; MERFISH=multiplexed error-robust fluorescence in situ hybridization; scATAC-seq = single-cell Assay for Transposase-Accessible Chromatin sequencing; scRNA-seq = single-cell RNA sequencing; SSc = systemic sclerosis; UMAP= uniform manifold approximation andprojection.

TRANSLATIONAL REVIEW

34 American Journal of Respiratory Cell and Molecular Biology Volume 61 Number 1 | July 2019

Page 6: The Human Lung Cell Atlas: A High-Resolution Reference Map

differentiation, particularly intermediateforms, to be analyzed transcriptionallyin a way that will be very difficult in theadult lung. In addition, the developinghuman lung mesenchymal and leukocytecell lineages are almost completelyuncharacterized. For example, it isunknown when artery-venous fate arises inthe lung endothelium, or when alveolarmacrophages first become resident in thehuman lungs. A Developmental Lung CellAtlas will provide baseline data describingthe molecular phenotype of the cells indeveloping lung, facilitating their futurefunctional characterization. It is importantto note that the human lung continues togrow after birth well into puberty andbeyond. Although the cellular changes duringthis period are completely unknown, they areconsidered to be of critical importance todevelopment of lung disease later in life (26).The presence of bronchial-associatedlymphoid tissue is rarely observed duringfetal lung development and infrequently innewborns and infants in the absence ofinfection (27), has been reported to increasewith age during the first few years of life (28,29), and is absent in healthy adults after theage of 20 years (30) except in the context ofspecific inflammatory conditions (31). Thisillustrates the intimate interactions between adeveloping immune system, environmentalchallenges, and a maturing lung duringearly life, further emphasizing the need forinclusion of neonatal and pediatric studies inthe Human Lung Cell Atlas.

The Human Lung Cell Atlas ofHealthy Aging and Disease

The aging process manifests itself in age-related changes in body composition, theendocrine system, and vasculature, whichare initially subclinical (32). The lung growsprogressively until early adulthood, andlung function peaks between 18 and 25years of age. After this peak, lung functiondeclines, associated with structuralremodeling of the lung, progressive lossof alveolar surface area, and enlargementof alveolar size (33, 34). In addition,mucociliary clearance is reduced with age inboth upper and lower airways (35, 36). Atthe cellular level, hallmarks of aging includegenomic instability, telomere attrition,epigenetic alterations, loss of proteostasis,deregulated nutrient-sensing mitochondrialdysfunction, stem-cell exhaustion,

cellular senescence, and altered cell–cellcommunication (37), which is reflected in(bulk) gene expression patterns from lung(38). Cellular senescence contributes toalterations in extracellular matrix and isassociated with physiological remodeling ofthe aging lung (39), and mitochondrialdysfunction has been observed in age-related lung disorders, including chronicobstructive pulmonary disease andidiopathic pulmonary fibrosis (40).Integrating deep-tissue proteomics andsingle-cell transcriptomics, a first draft of anaging mouse lung atlas reported increasedtranscriptional noise, indicating loss ofepigenetic control (41). Furthermore, age-associated changes in composition of theextracellular matrix, ciliated cell frequency,and a proinflammatory microenvironmentwere evident in both proteome andtranscriptome (41).

To systematically and comprehensivelyanalyze the effect of age on human lungcells, tissue sources used for the HumanLung Cell Atlas should therefore be collectedover the entire adult age range from 20 to 80years old. To distinguish natural variationfrom subclinical disease and to incorporate ahistory of relevant environmental andinfectious exposures, we will need to obtaindetailed clinical data, and potentially need touse biological characteristics of aging, suchas telomere length (42) and the epigeneticclock (43), to calibrate the findingsand understand natural variation andboundaries of health and disease. Thepromise of single-cell molecular profiling isto deliver unprecedented insight into thecellular origins of human disease. Thehuman genome project and decadesof genomic research have enabledinvestigations into the association of genepolymorphisms with disease. Using single-cell transcriptomics, we can now movefrom genetic and genomic associations tomore mechanistic questions about diseaseand cell type–associated gene networksand reveal the cellular context of disease-associated polymorphisms (44).

The Approach of the HumanLung Cell Atlas: Toolbox andResources

Single-Cell TranscriptomicsSingle-cell RNA sequencing (scRNA-seq)aims to comprehensively capture andsequence the RNA contents of a single cell,

allowing a description of transcriptionalheterogeneity in cell populations. Stabletranscriptional phenotypes can becontrasted with differentiation trajectoriesof cells by inferring an ordered sequence oftranscriptional states in pseudotime. Thekey to the utility of scRNA-seq in tissueprofiling has been the massive increasefrom 10s to more than 100,000 cells perexperiment in less than a decade (45, 46),due to technological advances as recentlyreviewed (47). Comprehensive profilingof a tissue under specific (developmental,diseased) conditions requires a highthroughput, capturing all cell types andstates—including putative novel ones—at athreshold of sensitivity that is determinedby both cell input and the observedvariation in transcriptional profiles betweendiscrete cell states. Rare cell types with atranscriptional phenotype that is notsufficiently discrete can be missed inunsupervised clustering, as exemplified bythe neuroendocrine cells in the recent cellcensus of airway wall tissue (48). Althoughenrichment or purification of these celltypes before scRNA-seq analysis can beapplied, this does require prior knowledgeof their identity. Atlasing studies thereforeoften combine a high-throughput approachfor cell type and cell marker identificationwith in-depth profiling of purifiedsubpopulations of special interest. To assesscompleteness of the single-cell sampling insilico, bulk profiles from scRNA-seq can becompared with true bulk RNA-seq datagenerated from adjacent regions of tissue.Another attractive alternative is the use ofnew high-density spatial transcriptomictools, such as Slide-seq or High-DensitySpatial Transcriptomics (HDST) (49, 50).These spatial tools can identify themicroanatomical niches that aresystematically undersampled and guideefforts for targeted enrichment.

Currently, sequencing of single cellsfrom freshly dissociated tissue yields thegreatest number of genes identified percell. However, this comes at the cost ofintroducing potential dissociation biases,such as a stress response induced byenzymatic tissue digestion (51) or a bias incell type composition, as cell types differ intheir optimal conditions for release fromthe tissue (52, 53). An alternative techniqueis the isolation of nuclei from snap-frozentissue, which are then assayed by singlenuclear RNAseq protocols (snRNA-seq).In general, snRNA-seq can be performed

TRANSLATIONAL REVIEW

Translational Review 35

Page 7: The Human Lung Cell Atlas: A High-Resolution Reference Map

on cryostat sections of lung tissue, with theadvantage of improved tissue processingwith less biased isolation, and enablingprofiling of adjacent tissue or serialsections using targeted imaging techniquesto put the snRNA-seq data into spatialcontext (49, 50, 54–57). Although single-nucleus RNA-seq detects fewer genes andunique transcripts than scRNA-seq, itallows the use of archived samples(58–61). On the whole, scRNA-seq andsnRNA-seq data correlate well (62) andallow robust cell type identification aswell as the derivation of developmentaltrajectories for the different cell typespresent in a tissue.

Beyond Single-Cell TranscriptomicsAlthough much of single-cell biology hasfocused on the transcriptome, epigeneticprofiling of single cells can provideinformation on a cells’ past history andpotential future states. Of the variousepigenetic profiling methods available,Assay for Transposase-AccessibleChromatin (ATAC)-seq uses transposasetagging to identify regions of openchromatin and regulatory elements withinthe genome. Recently, single-cell ATAC-seq(scATAC-seq) has become available withthe development of upfront bulktagmentation, followed by DNAfragmentation at the single-cell level (63,64). scATAC-seq has been shown toprovide mechanistic insight into theregulatory mechanisms underlyingcell states and their transitions (65).Differentiation trajectories of blood cellsdetected with scATAC-seq were similar tothose from RNA but with earlier branchingpoints, reflecting that the chromatinchanges detected by ATAC accessibilityprecede expression at the RNA level (66).Thus, scATAC-seq and scRNA-seq willprovide complementary insights into lung-cell states and the trajectories betweenthem. As the cost of high-throughputscATAC protocols continues to reduce(67), we expect that this approach willcontribute greatly to our understanding oflung development, cell type specification,and abnormality in disease.

Additional single-cell “omics”approaches, such as genome, methylome,proteome, and metabolome, are becomingavailable to complement transcriptomicmeasurements (68). The true size andshape of the cellular phenotypic landscapeof any organ will only be revealed in a

multiomic description of the cells andthe noncellular components. Severalmultiomics single-cell approaches havebeen explored, including integratedsingle-cell measurement of chromatinaccessibility; DNA methylation andtranscriptome (69); integratedgenetic, epigenetic, and transcriptomicmeasurement (70); and recentlyintegrated scRNA-seq with genotypedata to discover cell type–specificexpression quantitative trait loci (71).Recent progress in single-cell profilingtechnologies has made it possible tomeasure protein and RNA levels withinthe same cell simultaneously (72–74),which enhances discovery of subtlecellular phenotypes, such as cell typesubpopulations (75).

Imaging and Spatial TranscriptomicsDespite the advance of single-cellprofiling, the function of individual cellswithin a tissue ultimately depends on theirlocation within the tissue architecture andtheir interactions with the extracellularmatrix and neighboring cells. Consideringthe morphological diversity of the lung,characterization of the exact location andlocal microenvironment where cells areobtained from is critically important.Techniques for measuring gene expressionwithin lung tissue context range fromspatial transcriptomics, microdissectionapproaches such as laser-capturemicrodissection, and in situ RNAhybridization, to in situ RNA sequencing.The main limitations on spatial mappingof cell states defined by high-dimensionalgene expression patterns, however, are thenumber of genes detected on the sametissue sample and the shift from targetedto unbiased detection of gene expression.Spatial transcriptomic approachesmeasure gene expression in small voxelsof tissue, with recent High-DensitySpatial Transcriptomics (HDST) andSlide-seq technologies bringing theresolution down to around 1 to 10 mm(49, 50). A single-molecule fluorescencein situ hybridization (FISH)-basedprotocol to achieve high throughput is themultiplexed error-robust FISH (orMERFISH) technology (76), althoughapplication to the lung tissue mightrequire further optimization (77).Another protocol suitable for multiplexedRNA species detection is the proximityligation and in situ hybridization

technology (78). Interestingly, proximityligation and in situ hybridization iscompatible with immunohistochemicalapplications and the use of antibodieson archived formalin-fixed paraffinembedded (FFPE) tissue. Similarly,detection of proteins and their post-translational modification with antibodiesin combination with mRNA transcripts intissue sections was also achieved using amass cytometric approach (79). Finally,in situ RNA sequencing using padlockprobes (77, 80) or fluorescence in situRNA sequencing technology (81) isof interest. The recently developedSTARmap method combined in situamplification of mRNA transcripts withhydrogel tissue chemistry, allowingdetection of around a thousand genesin millimeter scale tissue blocks afterclearing of cells and lipids, whileretaining relative location within thetissue blocks’ coordinate system (82).Integration of such methods with high-resolution transcriptomic single-cellprofiling of matched tissue will likelybe key to establish a true atlas of thecellular landscape of healthy or diseasedhuman lung. A final technologicalchallenge is to integrate proteomic dataand transcriptomic data from singlecells, which requires development ofproteomic methods with single-cellsensitivity. Currently, a combinationof antibody-labeling technologies ormultiomics approaches, such ascombining mass cytometry by time-of-flight (CyTOF) with spatially resolvingFISH methodologies, remains theapproach of choice.

Sources of Lung TissueLung tissue for the Human Lung Cell Atlasis typically received from (routine) surgicalresections, from biopsy extraction in routineclinical care, or in a research setting wheninvolving healthy volunteers or lungtransplantation programs. In this context, itis important to highlight the difficulty ofobtaining normal lung parenchyma. Themost common sources for lung parenchymaare either uninvolved edges of lungresections (mostly for cancer) or tissueobtained from deceased donors throughorgan procurement organizations, which allcan be archived or processed fresh.Common sources of archived lung tissue arelarge tissue banks and repositories that havebeen established in many centers, mostly

TRANSLATIONAL REVIEW

36 American Journal of Respiratory Cell and Molecular Biology Volume 61 Number 1 | July 2019

Page 8: The Human Lung Cell Atlas: A High-Resolution Reference Map

associated with academic, medical, orgovernmental organizations (83). Biobanksor core tissue facilities take care of patientinformation, obtain informed writtenconsent for tissue donations and biopsycollections, and process and store tissue inan appropriate manner. They organize andmonitor tissue distribution for scientificresearch, store relevant clinical informationwith respect to preserving patientanonymity, and provide centralized datastorage. Specimens are collected accordingto site-specific protocols and legalregulations. To minimize tissue collectionvariability between different centers,common standard principles for tissueprocessing and collection are beingdeveloped. Live tissue from bronchoscopicbiopsies, organ procurement organizations,or surgical resection needs special attentionafter collection. These specimens need to betransported immediately after acquisitionto the research site for preparation ofsingle-cell suspensions and downstreamprocessing. Protocols for acquisition,transport, and processing of fresh tissuesamples are harmonized between the HCALung working group members and areavailable through online platforms (such ashttps://www.protocols.io/groups/hca/publications).

Computational ChallengesThe number of observations in a typicalsingle-cell experiment is generally muchlarger than bulk RNA-seq data. Therefore,the underlying gene expression matrices,often encoded with genes as rows and cellsas columns, pose novel and complexcomputational challenges because of theirsize. Moreover, so-called dropout events, inwhich an expressed gene is simply notdetected because of lack of sensitivity,introduce high levels of sparsity into thedata. Third, the increased resolution enablesmore precise capture of biologicalheterogeneity, which in turn leads toincreased complexity in the data. Altogether,scalability, sparsity, and complexity insingle-cell data pose major challengeswhen analyzing single-cell expressionprofiles.

Some popular scRNA-seq analysisframeworks, such as Scanpy (84), aredesigned to provide high scalability.Moreover, dimension reductionapplications in molecular biology haveevolved with the increased size andcomplexity of single-cell data. Linear

dimension reduction by principalcomponent analysis is commonly used forthe visualization of bulk and initial scRNA-seq datasets. More recent approaches, usingnonlinear two-dimensional embeddings,such as t-distributed stochastic neighborembedding (t-SNE) and uniform manifoldapproximation and projection (UMAP),have helped visualize single-cell dataand have recently been discussed withrespect to quality as well as scalability(85, 86). These nonlinear approaches areparticularly suitable for highly complextissues, such as the lung, where a largenumber of different cell types exist(41, 53).

The increased number of cells profiledin a single experiment represents acomputational challenge but also anopportunity for the application of moresophisticated modeling approaches,including machine learning (46). Deeplearning, a subfield of machine learning,which has for instance revolutionizedimage analysis, is particularly powerfulwhen large training datasets exist (87).Some of the recent, larger datasets nowopen up the possibility to apply deep-learning methods to scRNA-seq data.First, deep-learning algorithms have beendeveloped and tailored toward scRNA-seqdata with, for example, the goal ofdenoising scRNA-seq expression data (88).Machine learning approaches could bevery useful for the detection of robustcell type–specific signatures and theintegration of datasets generated ondifferent platforms, where sequencingdepth and transcript capture bias can varydramatically depending on the protocoland experimental setup. As more databecome available and datasets grow in size,we anticipate more machine learningapplications for the analysis of scRNA-seqdata.

Unbiased clustering approaches can beapplied not only to scRNA-seq or scATAC-seq data but also to spatial techniques touncover interactions between the celltypes forming lung microniches, as wasrecently demonstrated for epithelial andmesenchymal cells (10, 78, 89). These toolswill be particularly helpful for integration ofhigh-throughput sequencing and spatialdatasets. In combination with neuralnetwork–based approaches, we expectthat additional data features such asmorphometry—which can for example beused to robustly predict cell cycle stage

(90)—will be integrated into spatial analysisto learn about combined cell state andcommunication.

Another promising computationalapproach is the statistical deconvolution ofbulk RNA-seq data using scRNA-seq data toinfer cell type proportions of a complextissue sample (measured with bulk RNA-seq) by leveraging information fromscRNA-seq data (91). It was recently shownin pancreatic datasets that cell-typeheterogeneity represents a major covariatein bulk differential expression analysis (92).Lung tissue biopsies can show very highcell-type heterogeneity depending on theprecise location of surgery (93). Therefore,publicly available lung bulk data frompatients and control subjects is aparticularly promising candidatefor reanalysis using deconvolutiontechniques.

Opportunities to Build a Human LungCell AtlasThe opportunity to build a high-resolutionsingle-cell and spatial atlas of the humanbody has generated enormous excitement inthe scientific community. The globalinitiative to build the Human Cell Atlas,launched in 2016, now involves scientistsfrom 62 countries, a number likely toincrease. The HCA community is supportedby many funding organizations andinitiatives and organized into biologicalnetworks, including the Human LungCell Atlas. For example, in the specificarea of the lung, the NIH has activelysupported these efforts through programs,including LungMAP (94) in the healthylung, and disease-focused programs, inparticular the Cancer Moonshot HumanTumor Atlas Network (http://bit.ly/NCI-HumanTumorAtlas). Moreover,recognizing these emerging initiativesand the importance of supporting thisambitious new vision, the NIH CommonFund recently launched the HumanBioMolecular Atlas Program (HuBMAP),which includes the lung among its organsof focus (https://commonfund.nih.gov/hubmap). Other relevant efforts aresupported by the Medical Research Council(http://bit.ly/HCALungMRC) andWellcome Trust (http://bit.ly/HCALungW)in the United Kingdom and by the ChanZuckerberg Initiative (http://bit.ly/HCALungCZI1). The Human Lung CellAtlas will be built by integration of thedatasets provided by these and other

TRANSLATIONAL REVIEW

Translational Review 37

Page 9: The Human Lung Cell Atlas: A High-Resolution Reference Map

initiatives. Working within the HCA thusensures integration and internationalcoordination across multiple fundedefforts. Indeed, the HCA design isinherently inclusive and open, allowingparallel initiatives to contribute to theHuman Cell Atlas, leveraging expertise,data, and technological developmentsbetween projects, organ systems, institutes,and countries. Consequently, theinfrastructure that is under developmentwithin the HCA consortium will have ascope and reach beyond any of the organ-specific atlases, making the HCA wellsuited for a truly community-wideeffort toward establishing a HumanCell Atlas.

Next Steps Toward Establishing aHuman Lung Cell AtlasThe first cell censuses of upper airwaysand lung tissue have been published (48, 53,95, 96), and more will follow with bettercoverage of regional differences andrare cell types, as evident from theprogress meetings at the HCA meetings(http://bit.ly/HCALungTV1, http://bit.ly/HCALungTV2). These studies onsingle-cell profiling of healthy and diseasedlung tissue mainly report scRNA-seq andATAC-seq data (48, 53, 95, 96). With theanticipated increase in studies reportingmolecular phenotypes of lung cells, theefforts of the Lung Biological Networkwithin the HCA consortium is rapidlyshifting from reporting on the progress ofindividual groups to coordinating sharedand integrated analysis of the cumulativedata within the HCA data coordinationplatform. The next breakthrough inestablishing a Human Lung Cell Atlas willalso come from a detailed spatial mappingof cell types onto the tissue architecture,the description of molecular cell states as afunction of relative position in the lungalong one of the many gradients present inthe tissue or as a function of local cellularneighborhood, and the visualization of thecellular profiles in a spatial frameworkreflecting the structure of the lung. Inaddition, novel technological advances,especially in spatial mapping andcomputational approaches (e.g., towardmultimodal data integration andcombined visualization of profiling andspatial data) will remain an important

topic for future meetings of the HCAconsortium.

Joining the HCA LungBiological Network

The HCA consortium is open for anyonein the respiratory community to join(https://www.humancellatlas.org/joinHCA), and open sharing of bestpractices and proven productiveapproaches to tissue acquisition,processing, data generation, and analysisas well as technological innovations willhelp bring the field forward. Progress willbe presented at the HCA meetings butalso at the annual meetings of theAmerican Thoracic Society and theEuropean Respiratory Society. Lung hasbeen identified as one of the 12 priorityorgans and systems within the HCAinitiative, with an initial focus on upperand lower airways as well as lungparenchyma (6). To achieve our ambitionof establishing the Human Lung CellAtlas, we call on widespread involvementof the respiratory community tocontribute to this effort. This will allowthe HCA Lung Biological Network toachieve the depth and breadth necessaryto obtain a meaningful reference for basic,translational, and clinical respiratoryscientists that may serve as an examplefor other less readily accessible organsystems within the HCA. To facilitateintegration of datasets acquiredby different research groups atgeographically distinct sites, in multipleethnicities, age groups, and sexes, weurgently need standardization in tissueprocurement, processing, and storage, aswell as in data generation, handling,storage, and analysis pipelines. For tissueacquisition, we have developed a seriesof standardized protocols specificallytested and validated for lung tissue andcontinue to expand these, making themavailable to the community through openaccess platforms such as protocols.io(https://www.protocols.io/groups/hca/publications). For processing andintegration of data originating frommultiple centers and divergent technologicalplatforms, methodologies are beingtested using the datasets from the

initial lung scRNA-seq efforts. Thecomputational approaches are alsoshared with the community, for instancethrough platforms such as Github(https://github.com/HumanCellAtlas).Methods for integration of genomic,transcriptomic, and proteomic datasetsfrom the cellular branch of the HumanLung Cell Atlas with those acquired in thespatial branch will need to be furtherdeveloped. For data storage andaccessibility, the HCA lung project willtap into the HCA data coordinationplatform infrastructure that is currentlyin beta testing. One critical aspect for theHuman Lung Cell Atlas to achieve its fullimpact will be the development of aninteractive, tertiary HCA data portal (6)that allows exploration of the data by therespiratory and single-cell community. Atthis moment, interim data portals allowvisualization and limited exploration ofthe currently available datasets (seefor instance http://bit.ly/LungMap-scRNASeq [94], https://lungcellatlas.org [48], https://theislab.github.io/LungAgingAtlas [41], and https://www.nupulmonary.org/resources/ [53]).Incorporation of detailed demographicand clinical metadata from each tissuesource in the HCA data coordinationportal will be critical to allow theidentification of individual and combinedcontributions of sex, ethnicity, age,respiratory health, and disease to theposition of each cell type in thetranscriptomic and proteomic landscapethat defines the Human Lung CellAtlas.

The Human Lung cell atlas holds thepromise of becoming a resource ofunsurpassed scale and detail, allowing therespiratory community to acquire novelinsights into the biology of the lung,including—but not restricted to—itsdevelopment, anatomy, and physiology inhealth and the changes thereof in lungdisease. n

Author disclosures are available with the textof this article at www.atsjournals.org.

Acknowledgment: The authors thank DanaPe9er and Jennifer Rood for discussions onthe paper outline and feedback on themanuscript.

TRANSLATIONAL REVIEW

38 American Journal of Respiratory Cell and Molecular Biology Volume 61 Number 1 | July 2019

Page 10: The Human Lung Cell Atlas: A High-Resolution Reference Map

References

1. Gibson GJ, Loddenkemper R, Lundback B, Sibille Y. Respiratory healthand disease in Europe: the new European Lung White Book. EurRespir J 2013;42:559–563.

2. Franks TJ, Colby TV, Travis WD, Tuder RM, Reynolds HY, Brody AR,et al. Resident cellular components of the human lung: currentknowledge and goals for research on cell phenotyping and function.Proc Am Thorac Soc 2008;5:763–766.

3. Montoro DT, Haber AL, Biton M, Vinarsky V, Lin B, Birket SE, et al.A revised airway epithelial hierarchy includes CFTR-expressingionocytes. Nature 2018;560:319–324.

4. Plasschaert LW, Zilionis R, Choo-Wing R, Savova V, Knehr J, Roma G,et al. A single-cell atlas of the airway epithelium reveals the CFTR-richpulmonary ionocyte. Nature 2018;560:377–381.

5. Regev A, Teichmann SA, Lander ES, Amit I, Benoist C, Birney E, et al.;Human Cell Atlas Meeting Participants. The Human Cell Atlas. eLife2017;6:e27041.

6. Regev A, Teichmann S, Rozenblatt-Rosen O, Stubbington M, Ardlie K,Amit I, et al. The Human Cell Atlas White Paper. 2018 [accessed 2018Nov 1]. Available from: http://arxiv.org/abs/1810.05192.

7. Hogan BLM, Barkauskas CE, Chapman HA, Epstein JA, Jain R, HsiaCCW, et al. Repair and regeneration of the respiratory system:complexity, plasticity, and mechanisms of lung stem cell function. CellStem Cell 2014;15:123–138.

8. Weibel ER. On the tricks alveolar epithelial cells play to make a goodlung. Am J Respir Crit Care Med 2015;191:504–513.

9. Whitsett JA, Wert SE, Weaver TE. Diseases of pulmonary surfactanthomeostasis. Annu Rev Pathol 2015;10:371–393.

10. Nabhan AN, Brownfield DG, Harbury PB, Krasnow MA, Desai TJ.Single-cell Wnt signaling niches maintain stemness of alveolar type 2cells. Science 2018;359:1118–1123.

11. Zacharias WJ, Frank DB, Zepp JA, Morley MP, Alkhaleel FA, Kong J,et al. Regeneration of the lung alveolus by an evolutionarilyconserved epithelial progenitor. Nature 2018;555:251–255.

12. Lim HY, Lim SY, Tan CK, Thiam CH, Goh CC, Carbajo D, et al.Hyaluronan receptor LYVE-1-expressing macrophages maintainarterial tone through hyaluronan-mediated regulation of smoothmuscle cell collagen. Immunity 2018;49:326–341, e7. [Publishederratum appears in Immunity 49:1191.]

13. Gibbings SL, Thomas SM, Atif SM, McCubbrey AL, Desch AN, DanhornT, et al. Three unique interstitial macrophages in the murine lung atsteady state. Am J Respir Cell Mol Biol 2017;57:66–76.

14. Tan SYS, Krasnow MA. Developmental origin of lung macrophagediversity. Development 2016;143:1318–1327.

15. Whitsett JA, Kalin TV, Xu Y, Kalinichenko VV. Building and regeneratingthe lung cell by cell. Physiol Rev 2019;99:513–554.

16. Ludwig LS, Lareau CA, Ulirsch JC, Christian E, Muus C, Li LH, et al.Lineage tracing in humans enabled by mitochondrial mutations andsingle-cell genomics. Cell 2019;176:1325–1339, e22.

17. Moor AE, Harnik Y, Ben-Moshe S, Massasa EE, Rozenberg M, Eilam R,et al. Spatial reconstruction of single enterocytes uncovers broadzonation along the intestinal villus axis. Cell 2018;175:1156–1167,e15.

18. Parker MW, Rossi D, Peterson M, Smith K, Sikstrom K, White ES, et al.Fibrotic extracellular matrix activates a profibrotic positive feedbackloop. J Clin Invest 2014;124:1622–1635.

19. Packer JS, Zhu Q, Huynh C, Sivaramakrishnan P, Preston E, Dueck H,et al. A lineage-resolved molecular atlas of C. elegansembryogenesis at single cell resolution [preprint]. bioRxiv 2019[accessed 2019 Mar 11]. Available from: https://www.biorxiv.org/content/10.1101/565549v2.abstract.

20. Tata PR, Rajagopal J. Plasticity in the lung: making and breaking cellidentity. Development 2017;144:755–766.

21. Nikolic MZ, Sun D, Rawlins EL. Human lung development: recentprogress and new challenges. Development 2018;145:dev163485.

22. Nikolic MZ, Johnson J-A, Sun D, Caritg O, Laresgoiti U, Brady J, et al.Development of a genetically modifiable epithelial in-vitro culturesystem from human embryonic lung epithelial stem cells: towardshuman lung regeneration in end-stage respiratory failure. Lancet2017;389:S74.

23. Miller AJ, Hill DR, Nagy MS, Aoki Y, Dye BR, Chin AM, et al. In vitroinduction and in vivo engraftment of lung bud tip progenitor cellsderived from human pluripotent stem cells. Stem Cell Reports 2018;10:101–119.

24. Taylor DM, Aronow BJ, Tan K, Bernt K, Salomonis N, Greene CS, et al.The pediatric cell atlas: defining the growth phase of humandevelopment at single-cell resolution. Dev Cell 2019;49:10–29.

25. Teixeira VH, Nadarajan P, Graham TA, Pipinikas CP, Brown JM, FalzonM, et al. Stochastic homeostasis in human airway epithelium isachieved by neutral competition of basal cell progenitors. eLife 2013;2:e00966.

26. Bush A. Lung development and aging. Ann Am Thorac Soc 2016;13:S438–S446.

27. Gould SJ, Isaacson PG. Bronchus-associated lymphoid tissue (BALT)in human fetal and infant lung. J Pathol 1993;169:229–234.

28. Tschernig T, Kleemann WJ, Pabst R. Bronchus-associated lymphoidtissue (BALT) in the lungs of children who had died from suddeninfant death syndrome and other causes. Thorax 1995;50:658–660.

29. Heier I, Malmstrom K, Sajantila A, Lohi J, Makela M, Jahnsen FL.Characterisation of bronchus-associated lymphoid tissue andantigen-presenting cells in central airway mucosa of children. Thorax2011;66:151–156.

30. Hiller AS, Tschernig T, Kleemann WJ, Pabst R. Bronchus-associatedlymphoid tissue (BALT) and larynx-associated lymphoid tissue(LALT) are found at different frequencies in children, adolescents andadults. Scand J Immunol 1998;47:159–162.

31. Tschernig T, Pabst R. Bronchus-associated lymphoid tissue (BALT)is not present in the normal adult lung but in different diseases.Pathobiology 2000;68:1–8.

32. Partridge L, Deelen J, Slagboom PE. Facing up to the global challengesof ageing. Nature 2018;561:45–56.

33. Ochs M, Nyengaard JR, Jung A, Knudsen L, Voigt M, Wahlers T, et al.The number of alveoli in the human lung. Am J Respir Crit Care Med2004;169:120–124.

34. Hecker L. Mechanisms and consequences of oxidative stress in lungdisease: therapeutic implications for an aging populace. Am JPhysiol Lung Cell Mol Physiol 2018;314:L642–L653.

35. Proença de Oliveira-Maul J, Barbosa de Carvalho H, Goto DM, MaiaRM, Flo C, Barnabe V, et al. Aging, diabetes, and hypertension areassociated with decreased nasal mucociliary clearance. Chest 2013;143:1091–1097.

36. Svartengren M, Falk R, Philipson K. Long-term clearance from smallairways decreases with age. Eur Respir J 2005;26:609–615.

37. Lopez-Otın C, Blasco MA, Partridge L, Serrano M, Kroemer G. Thehallmarks of aging. Cell 2013;153:1194–1217.

38. de Vries M, Faiz A, Woldhuis RR, Postma DS, de Jong TV, Sin DD, et al.Lung tissue gene-expression signature for the ageing lung in COPD.Thorax [online ahead of print]; 6 Dec 2017 DOI: 10.1136/thoraxjnl-2017-210074.

39. Calhoun C, Shivshankar P, Saker M, Sloane LB, Livi CB, Sharp ZD,et al. Senescent cells contribute to the physiological remodeling ofaged lungs. J Gerontol A Biol Sci Med Sci 2016;71:153–160.

40. Meiners S, Eickelberg O, Konigshoff M. Hallmarks of the ageing lung.Eur Respir J 2015;45:807–827.

41. Angelidis I, Simon LM, Fernandez IE, Strunz M, Mayr CH, Greiffo FR,et al. An atlas of the aging lung mapped by single cell transcriptomicsand deep tissue proteomics. Nat Commun 2019;10:963.

42. Everaerts S, Lammertyn EJ, Martens DS, De Sadeleer LJ, Maes K, vanBatenburg AA, et al. The aging lung: tissue telomere shortening inhealth and disease. Respir Res 2018;19:95.

43. Horvath S, Raj K. DNA methylation-based biomarkers and theepigenetic clock theory of ageing. Nat Rev Genet 2018;19:371–384.

44. Hemberg M. Summing up the parts of the hypothalamus. Nat Neurosci2017;20:378–379.

45. Svensson V, Vento-Tormo R, Teichmann SA. Exponential scaling ofsingle-cell RNA-seq in the past decade. Nat Protoc 2018;13:599–604.

46. Angerer P, Simon L, Tritschler S, Alexander Wolf F, Fischer D, Theis FJ.Single cells make big data: new challenges and opportunities intranscriptomics. Curr Opin Syst Biol 2017;4:85–91.

47. Chen X, Teichmann SA, Meyer KB. From tissues to cell types and back:single-cell gene expression analysis of tissue architecture. Annu RevBiomed Data Sci 2018;1:29–51.

TRANSLATIONAL REVIEW

Translational Review 39

Page 11: The Human Lung Cell Atlas: A High-Resolution Reference Map

48. Vieira Braga FA, Kar G, Berg M, Carpaij OA, Polanski K, Simon LM, et al.A cellular census of healthy lung and asthmatic airway wall identifiesnovel cell states in health and disease [preprint]. bioRxiv 2019.[accessed 2019 Mar 11]. Available from: https://www.biorxiv.org/content/10.1101/527408v1.abstract.

49. Vickovic S, Eraslan G, Salmen F, Klughammer J, Stenbeck L, Aijo T, et al.High-density spatial transcriptomics arrays for in situ tissue profiling[preprint]. bioRxiv 2019 [accessed 2019 Mar 11]. Available from:https://www.biorxiv.org/content/10.1101/563338v1.abstract.

50. Rodriques SG, Stickels RR, Goeva A, Martin CA, Murray E, VanderburgCR, et al. Slide-seq: a scalable technology for measuring genome-wideexpression at high spatial resolution. Science 2019;363:1463–1467.

51. van den Brink SC, Sage F, Vertesy A, Spanjaard B, Peterson-Maduro J,Baron CS, et al. Single-cell sequencing reveals dissociation-inducedgene expression in tissue subpopulations. Nat Methods 2017;14:935–936.

52. Cohen M, Giladi A, Gorki A-D, Solodkin DG, Zada M, Hladik A, et al.Lung single-cell signaling interaction map reveals basophil role inmacrophage imprinting. Cell 2018;175:1031–1044, e18.

53. Reyfman PA, Walter JM, Joshi N, Anekalla KR, McQuattie-Pimentel AC,Chiu S, et al. Single-cell transcriptomic analysis of human lungprovides insights into the pathobiology of pulmonary fibrosis. Am JRespir Crit Care Med [online ahead of print] 15 Dec 2018; DOI:10.1164/rccm.201712-2410OC.

54. Stahl PL, Salmen F, Vickovic S, Lundmark A, Navarro JF, Magnusson J,et al. Visualization and analysis of gene expression in tissue sectionsby spatial transcriptomics. Science 2016;353:78–82.

55. Pinskiy V, Jones J, Tolpygo AS, Franciotti N, Weber K, Mitra PP. High-throughput method of whole-brain sectioning, using the tape-transfer technique. PLoS One 2015;10:e0102363.

56. Carow B, Hauling T, Qian X, Kramnik I, Nilsson M, Rottenberg ME.Spatial and temporal localization of immune transcripts defineshallmarks and diversity in the tuberculosis granuloma [preprint].bioRxiv 2019 [accessed 2019 Mar 11]. Available from:https://www.biorxiv.org/content/10.1101/509547v1.abstract.

57. Ziegenhain C, Vieth B, Parekh S, Hellmann I, Enard W. Quantitativesingle-cell transcriptomics. Brief Funct Genomics 2018;17:220–232.

58. Lacar B, Linker SB, Jaeger BN, Krishnaswami SR, Barron JJ, KelderMJE, et al. Nuclear RNA-seq of single neurons reveals molecularsignatures of activation. Nat Commun 2016;7:11022.

59. Habib N, Avraham-Davidi I, Basu A, Burks T, Shekhar K, Hofree M, et al.Massively parallel single-nucleus RNA-seq with DroNc-seq. NatMethods 2017;14:955–958.

60. Habib N, Li Y, Heidenreich M, Swiech L, Avraham-Davidi I, TrombettaJJ, et al. Div-seq: single-nucleus RNA-seq reveals dynamics of rareadult newborn neurons. Science 2016;353:925–928.

61. Krishnaswami SR, Grindberg RV, Novotny M, Venepally P, Lacar B,Bhutani K, et al. Using single nuclei for RNA-seq to capture thetranscriptome of postmortem neurons. Nat Protoc 2016;11:499–524.

62. Lake BB, Codeluppi S, Yung YC, Gao D, Chun J, Kharchenko PV,et al. A comparative strategy for single-nucleus and single-celltranscriptomes confirms accuracy in predicted cell-type expressionfrom nuclear RNA. Sci Rep 2017;7:6031.

63. Chen X, Miragaia RJ, Natarajan KN, Teichmann SA. A rapid and robustmethod for single cell chromatin accessibility profiling. Nat Commun2018;9:5345.

64. Chen X, Litzenburger U, Wei Y, Schep AN, LaGory EL, Choudhry H,et al. Joint single-cell DNA accessibility and protein epitope profilingreveals environmental regulation of epigenomic heterogeneity. NatCommun 2018;9:4590.

65. Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, SnyderMP, et al. Single-cell chromatin accessibility reveals principles ofregulatory variation. Nature 2015;523:486–490.

66. Buenrostro JD, Corces MR, Lareau CA, Wu B, Schep AN, Aryee MJ,et al. Integrated single-cell analysis maps the continuous regulatorylandscape of human hematopoietic differentiation. Cell 2018;173:1535–1548, e16.

67. Mezger A, Klemm S, Mann I, Brower K, Mir A, Bostick M, et al. High-throughput chromatin accessibility profiling at single-cell resolution.Nat Commun 2018;9:3647.

68. Colome-Tatche M, Theis FJ. Statistical single cell multi-omicsintegration. Curr Opin Syst Biol 2018;7:54–59.

69. Clark SJ, Argelaguet R, Kapourani C-A, Stubbs TM, Lee HJ, Alda-Catalinas C, et al. scNMT-seq enables joint profiling of chromatinaccessibility DNA methylation and transcription in single cells. NatCommun 2018;9:781.

70. Hou Y, Guo H, Cao C, Li X, Hu B, Zhu P, et al. Single-cell tripleomics sequencing reveals genetic, epigenetic, and transcriptomicheterogeneity in hepatocellular carcinomas. Cell Res 2016;26:304–319.

71. van der Wijst MGP, Brugge H, de Vries DH, Deelen P, Swertz MA,Franke L; LifeLines Cohort Study; BIOS Consortium. Single-cell RNAsequencing identifies celltype-specific cis-eQTLs and co-expressionQTLs. Nat Genet 2018;50:493–497.

72. Valdes AM, Glass D, Spector TD. Omics technologies and the study ofhuman ageing. Nat Rev Genet 2013;14:601–607.

73. Darmanis S, Gallant CJ, Marinescu VD, Niklasson M, Segerman A,Flamourakis G, et al. Simultaneous multiplexed measurement of RNAand proteins in single cells. Cell Rep 2016;14:380–389.

74. Peterson VM, Zhang KX, Kumar N, Wong J, Li L, Wilson DC, et al.Multiplexed quantification of proteins and transcripts in single cells.Nat Biotechnol 2017;35:936–939.

75. Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B,Chattopadhyay PK, Swerdlow H, et al. Simultaneous epitope andtranscriptome measurement in single cells. Nat Methods 2017;14:865–868.

76. Moffitt JR, Hao J, Wang G, Chen KH, Babcock HP, Zhuang X. High-throughput single-cell gene-expression profiling with multiplexederror-robust fluorescence in situ hybridization. Proc Natl Acad SciUSA 2016;113:11046–11051.

77. Moor AE, Itzkovitz S. Spatial transcriptomics: paving the way fortissue-level systems biology. Curr Opin Biotechnol 2017;46:126–133.

78. Nagendran M, Riordan DP, Harbury PB, Desai TJ. Automated cell-typeclassification in intact tissues by single-cell molecular profiling. eLife2018;7:e30510.

79. Schulz D, Zanotelli VRT, Fischer JR, Schapiro D, Engler S, Lun X-K,et al. Simultaneous multiplexed imaging of mRNA and proteins withsubcellular resolution in breast cancer tissue samples by masscytometry. Cell Syst 2018;6:531.

80. Ke R, Mignardi M, Pacureanu A, Svedlund J, Botling J, Wahlby C, et al.In situ sequencing for RNA analysis in preserved tissue and cells. NatMethods 2013;10:857–860.

81. Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Ferrante TC, Terry R, et al.Fluorescent in situ sequencing (FISSEQ) of RNA for gene expressionprofiling in intact cells and tissues. Nat Protoc 2015;10:442–458.

82. Wang X, Allen WE, Wright MA, Sylwestrak EL, Samusik N, Vesuna S,et al. Three-dimensional intact-tissue sequencing of single-celltranscriptional states. Science 2018;361:eaat5691.

83. Haga SB, Beskow LM. Ethical, legal, and social implications ofbiobanks for genetics research. Adv Genet 2008;60:505–544.

84. Wolf FA, Angerer P, Theis FJ. SCANPY: large-scale single-cell geneexpression data analysis. Genome Biol 2018;19:15.

85. Becht E, McInnes L, Healy J, Dutertre C-A, Kwok IWH, Ng LG,et al. Dimensionality reduction for visualizing single-cell data usingUMAP. Nat Biotechnol [online ahead of print] 3 Dec 2018; DOI:10.1038/nbt.4314.

86. Kobak D, Berens P. The art of using t-SNE for single-celltranscriptomics [preprint]. bioRxiv 2018. [accessed 2019 Mar 11].Available from: https://www.biorxiv.org/content/10.1101/453449v1.abstract.

87. LeCun Y, Bengio Y, Hinton G. Deep learning. Nature 2015;521:436–444.88. Eraslan G, Simon LM, Mircea M, Mueller NS, Theis FJ. Single-cell RNA-

seq denoising using a deep count autoencoder. Nat Commun 2019;10:390.

89. Zepp JA, Zacharias WJ, Frank DB, Cavanaugh CA, Zhou S, Morley MP,et al. Distinct mesenchymal lineages and niches promote epithelialself-renewal and myofibrogenesis in the lung. Cell 2017;170:1134–1148, e10.

90. Eulenberg P, Kohler N, Blasi T, Filby A, Carpenter AE, Rees P, et al.Reconstructing cell cycle and disease progression using deeplearning. Nat Commun 2017;8:463.

TRANSLATIONAL REVIEW

40 American Journal of Respiratory Cell and Molecular Biology Volume 61 Number 1 | July 2019

Page 12: The Human Lung Cell Atlas: A High-Resolution Reference Map

91. Shen-Orr SS, Gaujoux R. Computational deconvolution: extracting celltype-specific information from heterogeneous samples. Curr OpinImmunol 2013;25:571–578.

92. Baron M, Veres A, Wolock SL, Faust AL, Gaujoux R, Vetere A, et al. Asingle-cell transcriptomic map of the human and mouse pancreasreveals inter- and intra-cell population structure. Cell Syst 2016;3:346–360, e4.

93. Baharom F, Thomas S, Rankin G, Lepzien R, Pourazar J, Behndig AF,et al. Dendritic cells and monocytes with distinct inflammatoryresponses reside in lung mucosa of healthy humans. J Immunol2016;196:4498–4509.

94. Ardini-Poleske ME, Clark RF, Ansong C, Carson JP, Corley RA,Deutsch GH, et al.; LungMAP Consortium. LungMAP: the molecularatlas of lung development program. Am J Physiol Lung Cell MolPhysiol 2017;313:L733–L740.

95. Ordovas-Montanes J, Dwyer DF, Nyquist SK, Buchheit KM, Vukovic M,Deb C, et al. Allergic inflammatory memory in human respiratoryepithelial progenitor cells. Nature 2018;560:649–654.

96. Xu Y, Mizuno T, Sridharan A, Du Y, Guo M, Tang J, et al. Single-cell RNA sequencing identifies diverse roles of epithelialcells in idiopathic pulmonary fibrosis. JCI Insight 2016;1:e90558.

TRANSLATIONAL REVIEW

Translational Review 41