ArticlePDF AvailableLiterature Review

Towards a Human Cell Atlas: Taking Notes from the Past

Authors:

Abstract

Comprehensively characterizing the cellular composition and organization of tissues has been a long-term scientific challenge that has limited our ability to study fundamental and clinical aspects of human physiology. The Human Cell Atlas (HCA) is a global collaborative effort to create a reference map of all human cells as a basis for both understanding human health and diagnosing, monitoring, and treating disease. Many aspects of the HCA are analogous to the Human Genome Project (HGP), whose completion presents a major milestone in modern biology. To commemorate the HGP’s 20-year anniversary of completion, we discuss the launch of the HCA in light of the HGP, and highlight recent progress by the HCA consortium.
Series: Celebrating the Human Genome Project and Its Outcomes
Opinion
Towards a Human Cell Atlas: Taking Notes
from the Past
Rik G.H. Lindeboom ,
1,
*Aviv Regev,
2,4
and Sarah A. Teichmann
1,3
Comprehensively characterizing the cellular composition and organization of
tissues has been a long-term scientic challenge that has limited our ability to
study fundamental and clinical aspects of human physiology. The Human Cell
Atlas (HCA) is a global collaborative effort to create a reference map of all
human cells as a basis for both understanding human health and diagnosing,
monitoring, and treating disease. Many aspects of the HCA are analogous to
the Human Genome Project (HGP), whose completion presents a major mile-
stone in modern biology. To commemorate the HGPs 20-year anniversary of
completion, we discuss the launch of the HCA in light of the HGP, and highlight
recent progress by the HCA consortium.
Building a Reference Map of the Human Body
In the past decade, new methods have emerged for single-cell genomics that have revolutionized
our ability to identify and characterize the cells that comprise complex tissues. With these tools at
hand, the HCA project was launched as an international collaborative effort to create comprehensive
reference maps of all human cells the fundamental units of life as a basis for both understanding
human health and diagnosing, monitoring, and treating disease [1]. The foundation for organizing
large-scale consortium efforts such as the HCA leads back to the HGP. To commemorate the
completion of the HGP 20 years ago, we lay out organizational considerations and the latest
progress of the HCA community.
Lessons from the HGP
The HGP was launched in 1990 as a scientic effort of unprecedented magnitude to create a
reference map of the human genome. The success of this ambitious project depended on an inter-
disciplinary approach that bridged teams specialized in computation, engineering, and biology, in
an international collaboration between institutions in the USA, Europe, and Asia. The focus on in-
ternational and interdisciplinary collaboration inspired numerous large-scale consortium-based re-
search ventures that followed, including the HCA initiative. Recognizing the broad importance of
ethics by dedicating 5% of its funding to ensure proper ethical practice and explore its societal im-
pact, the HGP contributed an important aspect that inspired many large-scale biological projects.
While it was not fully clear at the time of the launch how the HGP would succeed in its ambitious
goal, the focus on intermediate milestones and technology development eventually led to a nished
human genome reference 2 years ahead of schedule and with budget to spare. This achievement
underlines the importance of phasing long-term initiatives into graspable intermediate goals to re-
ne future plans and exploit the inevitable increase in throughput and resolution that technological
advances bring.
Organizing the HCA
Similar to the aim of the HGP to build a reference map of the genome, the goal of the HCA initiative
is to create reference maps that chart the cells in human tissues and organs. Building such maps
Highlights
The Human Cell Atlas (HCA) consortium
was founded as a collaborative and
open effort to create a reference map of
the cells in the human body.
Organizing a large-scale project such as
the HCA draws inspiration from the
Human Genome Project (HGP) that
was completed 20 years ago.
Signicant progress has been made by
the HCA community, including proling
more than 39 million cells from 15 major
organs to date.
The expected impact of the HCA is illus-
trated by its use during the coronavirus
disease 2019 (COVID-19) pandemic.
1
Wellcome Sanger Institute, Wellcome
Genome Campus, Hinxton, Cambridge,
UK
2
Klarman Cel l Observatory , Broad
Institute of MIT and Harvard, Cambridge,
MA, USA
3
Cavendish Laboratory, JJ Thomson
Avenue, University o f Cambridge,
Cambridge, UK
4
Current Address: Genentech, South
San Francisco,
CA, USA
*Correspondence:
rl21@sanger.ac.uk (R.G.H. Lindeboom).
Trends in Genetics, Month 2021, Vol. xx, No. xx https://doi.org/10.1016/j.tig.2021.03.007 1
© 2021 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Trends in
Genetics OPEN ACCESS
TIGS 1799 No. of Pages 6
requires collaboration between research groups and institutes, so the HCA was launched as an
open international initiative. To maximize the benets from collaboration and data sharing, the
HCA is organized as an open and community-driven venture with more than 2000 members to
date, and growing. Any scientist that shares its ambitions, goals, and values can become a mem-
ber at any point by registering online
i
.
To ensure scientic leadership and deep engagement from the broad scienticcommunity,
HCAs working groups take on its core challenges
ii
, and include the Biological Networks, each
taking on a specic tissue, organ, or system, the Analysis Working Group, focused on computa-
tional and analytical challenges, the Standards and Technologies Working Group, focused on the
needed experimental assays. Notably, when creating a human reference resource such as the
HCA, it is essential to ensure an equal benet is gained worldwide, to both the participating
scientists and the representation of humanity, requiring the incorporation of extensive diversity
in sex and ethnicity. HCA engaged in this goal early and in an ongoing manner through the Ethics
Working Group, and more recently the Equity Working Group [2].
The relatively late realization of the importance of data analysis in the HGP presented a bottleneck
for the HGP to construct a genome reference [3]. Learning from this past experience, the
continuous development of computational approaches has thus been a major area of focus of
the HCA community
iii
, where the Analysis Working Group the rst working group of HCA is
dedicated to the key computational challenge of building and querying and atlas. While the
core product of the HGP was essentially a single DNA sequence, the multimodal and complex
nature of the data generated within the HCA will require a modular and multifaceted approach
to standardize, integrate, and share data. To this end, the HCA Data Coordination Platform
was established in 2017, and is under continuous development to accommodate standardized
processing and broad access to HCA data through both graphic and programming interfaces
iv
.
In addition, a burgeoning community of tertiary data portals now enable users to easily access
and analyze HCA data without the need for sophisticated bioinformatics expertise. Examples of
these portals include the cellxgene software [4], EBIs Single Cell Expression Atlas
v
,the
Cambridge Portal
vi
, the Broad Single Cell Portal
vii
,andtheUCSCCellPortal
viii
, each offering
interactive access to datasets from a wide range of HCA studies, with distinct analysis features.
Other dedicated portals, such as the COVID-19 Cell Atlas
ix
, Developmental Cell Atlas
x
, and the
Human Tumor Atlas
xi
data portals, are HCA-related portals dedicated to specic aspects of
human biology and/or disease.
Unlike the single coordinated funding structure for the HGP, the HCA involves a more distributed
structure, reecting the democratization of technology, computation, and growth of the biomed-
ical scientic community itself, especially in genomics and computational biology, over the past
decades. At the time of the HGP, only a few, large, and heavily funded centers could perform
the needed work, but both single cell genomics technologies and associated computation has
become much more broadly accessible, partly due to the innovation and efforts of HCA
members. As a result, the funding and organizational structure is distinct: scientists participate
in HCA irrespective of their specic funding source, many funders support atlas construction
activities, and HCA is allied with several formally funded consortia focused on specic aspects,
providing a scientic community open to all.
Progress towards a Draft of the HCA
The HCA has already made signicant progress towards the goal set for the rst draft of a cell
atlas proling the common cell types in tissues from the major human organs
xii
. Currently,
HCA scientists have proled more than 39 million cells using suspension cell genomics from 15
Trends in Genetics
OPEN ACCESS
2Trends in Genetics, Month 2021, Vol. xx, No. xx
major organ systems, including for example 11.1 million nervous system cells, 5.8 million embry-
onic and fetal cells, 3.4 million lung cells and 7.2 million immune cells. These atlases also cover
important human diseases, including nearly 4.8 million cells derived from severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2)-infected individuals. These statistics are collected by the
HCA Executive Ofce through regular quarterly surveys of its members, and thus reect data cur-
rently available in a multiple range of sources, including data consented for open access stored at
the HCA Data Coordination Platform
iv
, and HCA data generated under consent for data sharing
by managed or controlled access only, which are currently distributed across various databases
such as dbGAP, DUOS, and EGA. These cell counts also include unpublished datasets for which
the cell numbers have been shared with us by HCA members.
The data collected across HCA is leading to exciting scientic discoveries. For example, a recently
published cardiac single cell reference highlights the cellular heterogeneity of the atrial and ven-
tricular chambers, and gender-specic differences in the cellular composition of the heart [5].
The emerging lung atlas discovered a host of new cell types, from the ionocyte, a new cell type
expressing the cystic brosis gene CFTR [6], to endothelial cell subsets that may play a role in
COVID-19 [7]. The gut atlas is recovering many dozens of cell types in the small and large intestine
[8,9], including rare cells such as enteric neurons [10]. Similarly, multiple atlases of the human
liver, an organ historically known for its homogenous cell type composition, reveal heterogeneity
in epithelial progenitors [11] and provide broad insights in hematopoietic development that
occurs in the fetal liver [12]. Single-nucleus RNA-sequencing (seq) of the neurons of the cerebral
cortex uncovered extensive differences in the cellular composition and characteristics between
human and mouse models, highlighting the importance of generating a cell atlas for humans
[13]. A spatial cell atlas of healthy and diseased pancreas tissues reveals how the morphological
organization of this organ features cell-type-specic neighborhoods and unexpected cellcell
interactions [14]. In the thymus, the dynamic cellular composition across the human lifespan
unraveled the development and repertoire of T cells and thymic stroma at unprecedented
detail [15]. Proling the cellular composition of the maternalfetal interface of the placenta
unveiled many regulatory interactions that govern the cellular organization during early human
pregnancy [16].
While the rst steps towards a HCA are to create a reference of healthy cells, many efforts have
also already examined implications in disease. A human single-cell atlas of the lung has identied
novel epithelial cell types, including asthma-related cell populations [17], and similar atlases in the
gut have helped understand cells related to inammatory bowel disease [8,9]. Charting the dy-
namic cellular composition of the fetal, pediatric, and adult human kidney has uncovered that pe-
diatric and adult kidney cancers originate from different and previously little-known cell types [18].
Systematic interrogation of tumor cell landscapes with complementary single-cell RNA-seq tech-
niques has furthermore enabled scientists to study single-cell biology at a pancancer scale [19].
Several cell atlases have detailed organ-specic subsets of tissue-resident immune cells
[5,12,15,18], underscoring the impact of spatial and environmental inuences of cells for their
identity and function. In disease, the HCA approach has inspired dedicated initiatives such as
the Human Tumor Atlas Network [20], whereas efforts such as the Kidney Precision Medicine
Program (KPMP) are tackling multiple kidney diseases. Similarly, the COVID-19 pandemic
sparked a large-scale joint effort of HCA scientists to shed light on this new pathology at
single-cell resolution (see later).
Impact of the HCA
As highlighted earlier, contributions by the HCA community have already led to numerous insights
ranging from basic human physiology and fundamental biology to discoveries with direct clinical
Trends in Genetics OPEN ACCESS
Trends in Genetics, Month 2021, Vol. xx, No. xx 3
applications such as pinpointing disease-associated cell types and pathology-induced cell states
(Figure 1). Beyond these direct insights that individual studies bring, the long-term goal of the
HCA is to provide a comprehensive reference of the identities and characteristics of the cells in
ahumanbody(seeOutstanding Questions). The HGP provided a reference where biologists
could look up the origin of, for example, their isolated fragment of DNA, RNA, or protein, and
how this differed in a disease context. The HCA aspires to become a similar tool to accelerate
both fundamental and translational science. Equivalent look-ups for HCA will include determining
which cells express a gene of interest, what cell types are present in a tissue/organ, and which cell
types co-occur in close spatial proximity. In addition, key marker genes that identify a cell type of
interest can be derived from the HCA, which can be the starting point for numerous experimental
assays.
The impact of the HCA on understanding human disease was powerfully illustrated during the
dawn of the COVID-19 pandemic. To obtain early insights into the pathology of COVID-19, the
HCA community used the existing single-cell RNA-seq data in the atlas to study important as-
pects of viral infection and responses at single cell resolution [21]. This quickly led to a compre-
hensive overview of the cells and organs that express key viral entry genes, such as ACE2, and
are therefore susceptible to SARS-CoV-2 infection. HCA scientists have since turned to studying
samples from COVID-19 patients and deceased donors directly, to shed light on disease pathol-
ogy at the single cell and spatial level. This work presents a clear case-in-point of how the HCA will
be useful for biologists and society.
The HCA also holds a translational promise, where the cell atlas can be applied to a range of
medical questions related to, for example, disease mechanisms, diagnostics, regenerative
medicine, and drug discovery and toxicity (Figure 1). Here, the HCA can be used to identify
disease-associated cell phenotypes that represent drug targets, predict cell-type specic effects
and understand on-target effects in other cell types. In regenerative medicine, the cell atlas can
provide guidance on how to steer differentiation into desired cell fates. In the clinic, new
biomarkers for disease could be identied and interpreted at unprecedented resolution.
Disease mechanisms
Diagnostics
Drug development
Regenerative medicineReference for spatial and
molecular cell characteristics
Foundation for
future consortia
Human
Genome
Project
Trends
Trends
in
in
Genetics
Genetics
Figure 1. Schematic Overview of the Impact of the Human Cell Atlas (HCA). Different biomedical elds that are
expected to benet from the HCA are highlighted around the HCA logo. These elds (indirectly) also greatly beneted from
the Human Genome Project, which has provided insights and inspiration for organizing and predicting the impact of
consortia such as the HCA.
Trends in Genetics
OPEN ACCESS
4Trends in Genetics, Month 2021, Vol. xx, No. xx
While the earlier-mentioned examples provide insights in the direct and future utility of the HCA,
we can take notes from other consortia such as the HGP to predict the longer-term future impact
of generating the HCA (Figure 1). The impact of the HGP was much greater than the direct discov-
eries about the human genome. With the genome reference as a basis, many new large-scale
consortia such as HapMAP, 1000 Genomes, GWAS, ENCODE, and TCGA/ICGC projects
were launched, resulting in unprecedented scientic achievements. We expect that the HCA
can have a similar impact, where the fundamental knowledge about the cellular organization of
our body will act as a foundation for future projects and consortia to investigate human physiology
and disease at even higher resolution and in the spatial context of the human body.
Concluding Remarks and Future Perspectives
As we progress towards a rst draft of the HCA, exciting technological advances are enabling the
community to characterize cells at higher throughput, and in a more detailed and comprehensive
manner. While the cellular maps highlighted earlier are mostly based on single-cell RNA-seq,
these are now complemented with chromatin proles, protein proles, and spatial information.
Simultaneous measurements of multiple modalities in the same single cell,such as the proteome,
transcriptome, and epigenome, will greatly advance our understanding of cell identities and phe-
notypic characteristics.
As laid out in the HCA White Paper [22], the rst draft of the HCA aims to prole 30100 million
human cells from all major organs in ethnically diverse males and females. In addition to cell
suspension-based transcriptome proling which is currently the most mature technology to
prole cells for the HCA these tissues will also be proled with spatial proling technologies to
map identied cell types onto the tissue architecture of the human body. Rapid developments
in the throughput and molecular resolution of spatial transcriptomics and other spatial proling
methods have enabled the establishment of the spatial branch of the HCA.
Lastly, mirroring the advances in sequencing technologies at the time of the HGP, there is a
sustained dramatic increase in throughput of single-cell proling techniques; a trend that will
allow HCA to reach its ambitious goal to prole and ultimately characterize billions of cells from
human organs in health, as a reference map of the human body.
Acknowledgments
We gratefully acknowledge Jennifer E. Rood for critical reading and editingof this manuscript, and all authors of HCA papers
that we have not been able to cite due to space constraints.
Declaration of Interests
In the last 3 years, S.A.T. has consulted for Genentech and Roche, and is a member of SABs of Biogen, GlaxoSmithKline,
and Foresite Labs. A.R. is a cofounder and equity holder of Celsius Therapeutics, an equity holder of Immunitasand was an
SAB member of Neogene Therapeutics, Thermo Fisher Scientic, Asimov, and Syros Pharmaceuticals until July 31, 2020.
Since August 1, 2020, A.R. is an employee of Genentech, a mem ber of the Roche group. A.R. is an inventor on multiple
patents to the Broad Institute in the area of single cell genomics. R.G.H.L. has no interests to declare.
Resources
i
www.humancellatlas.org/join-hca/
ii
www.humancellatlas.org/learn-more/working-groups/
iii
https://openproblems.bio/
iv
https://data.humancellatlas.org
v
www.ebi.ac.uk/gxa/sc/home
vi
www.cambridgecellatlas.org
vii
https://singlecell.broadinstitute.org/single_cell
Trends in Genetics OPEN ACCESS
Trends in Genetics, Month 2021, Vol. xx, No. xx 5
Outstanding Questions
What is the comprehensive compendium
of cell types and cell states in the human
body?
Are there differences in tissue architecture
between males and females (besides
reproductive tissues)?
Are there stereotypical patterns of
ageing in human tissues?
Can HCA data reveal new units of
tissue architecture (i.e., recurring 3D
motifs of cell types) that are hitherto
unappreciated?
Can HCA data reveal new, previously
unknown adult stem cells and pinpoint
regeneration occurring as part of ho-
meostasis within adult tissues?
How similar are immune, broblast,
and vascular lineage cells across
tissues and organs?
What is the impact of gradients of
signaling molecules (either proteins or
small molecules, including, e.g., oxygen
gradients) in development and in adult
tissues?
Are cellular responses to gradients and
other challenges (e.g., oxidative stress)
restricted to a single cellular compart-
ment or concerted across, for exam-
ple, immune, epithelial, and vascular
compartments?
viii
https://cells.ucsc.edu
ix
www.covid19cellatlas.org
x
https://developmentcellatlas.ncl.ac.uk
xi
https://data.humantumoratlas.org
xii
www.humancellatlas.org/publications/
References
1. Regev, A. et al. (2017) The Human Cell Atlas. eLife 6, e27041
2. Majumder, P.P. et al. (2020) The Human Cell Atlas and equity:
lessons learned. Nat. Med. 26, 15091511
3. Green, E.D. et al. (2015) Human Genome Project: twenty-ve
years of big biology. Nature 526, 2931
4. Li, K. et al. (2020) cellxgene VIP unleashes full power of interac-
tive visualization, plotting and analysis of scRNA-seq data in the
scale of millions of cells. bioRxiv Publ ished online A ugust 31,
2020. https://doi.org/10.1101/2020.08.28.270652
5. Litviňuková, M. et al. (2020) Cells of the adult human heart.
Nature 588, 466472
6. Montoro, D.T. et al. (2018) A revised airway epithelial hierarchy
includes CFTR-expressing ionocytes. Nature 560, 319324
7. Travaglini, K.J. et al. (2020) A molecular cell atlas of the human
lung from single-cell RNA sequencing. Nature 587, 619625
8. Smillie, C.S. et al. (2019) Intra- and inter-cellular rewiring of the
human colon during ulcerative colitis. Cell 178, 714730.e22
9. Martin, J.C. et al. (2019) Single-cell analysis of Crohns disease
lesions identies a pathogenic c ellular module associated with
resistance to anti-TNF therapy. Cell 178, 14931508
10. Drokhlyansky, E. et al. (2020) The human and mouse enteric ner-
vous system at single-cell resolution. Cell 182, 16061622.e23
11. Aizarani, N. et al. (2019) A human liver cell atlas reveals heteroge-
neity and epithelial progenitors. Nature 572, 199204
12. Popescu, D.M. et al. (2019) Decoding human fetal liver
haematopoiesis. Nature 574, 365371
13. Hodge, R.D. et al. (2019) Conserved cell t ypes with diverge nt
features in human versus mouse cortex. Nature 573, 6168
14. Tosti,L. et al. (2020) Singlenucleus and in situRNA sequencingre-
veals cell topographies in the human pancreas. Gastroenterology
160, 13301344.e11
15. Park, J.E. et al. (2020) A cell atlas of human thymic development
denes T cell repertoire formation. Science 367, eaay3224
16. Vento-Tormo, R. et al. (2018 ) Single-cell recon struction of the
early maternalfetal interface in humans. Nature 563, 347353
17. Vieira Braga, F.A. et al. (2019) A cellular census of human lungs
identies novel cell states in health and in asthma. Nat. Med. 25,
11531163
18. Young, M.D. et al. (2018) Single-cell transcriptomes from human
kidneys reveal the cellular identity of renal tumors. Science 361,
594599
19. Slyper, M. et al. (2020) A single-cell and single-nucleus RNA-Seq tool-
box for fresh and frozen human tumors. Nat. Med. 26, 792802
20. Rozenblatt-Rosen, O. et al. (2020) The Human Tumor Atlas
Network: chart ing tumor transit ions across space a nd time at
single-cell resolution. Cell 181, 236249
21. Teichmann, S. and Regev, A. (2020) The network effect:
studying COVID-19 patholog y with the Human Cell Atla s. Nat.
Rev. Mol. Cell Biol. 21, 415416
22. Regev, A. et al. (2018) The Human Cell Atlas White Paper. arXiv
Published online October 11, 2018. http://arxiv.org/abs/
1810.05192
Trends in Genetics
OPEN ACCESS
6Trends in Genetics, Month 2021, Vol. xx, No. xx
... The first will feature single-cell and single-nucleus transcriptomic data from healthy tissues, combined with spatial analysis. HCA researchers have profiled 58.5 million cells across 15 organ systems to date [13]. Commensurate with their objectives; however, the HCA's Biological Network atlases map discrete tissues, organ systems, or focus areas such that they, with the limited exceptions described here, do not yield cross-tissue single-cell atlases. ...
Article
Full-text available
The observation that certain therapeutic strategies for targeting inflammation benefit patients with distinct immune-mediated inflammatory diseases (IMIDs) is exemplified by the success of TNF blockade in conditions including rheumatoid arthritis, ulcerative colitis, and skin psoriasis, albeit only for subsets of individuals with each condition. This suggests intersecting “nodes” in inflammatory networks at a molecular and cellular level may drive and/or maintain IMIDs, being “shared” between traditionally distinct diagnoses without mapping neatly to a single clinical phenotype. In line with this proposition, integrative tumour tissue analyses in oncology have highlighted novel cell states acting across diverse cancers, with important implications for precision medicine. Drawing upon advances in the oncology field, this narrative review will first summarise learnings from the Human Cell Atlas in health as a platform for interrogating IMID tissues. It will then review cross-disease studies to date that inform this endeavour before considering future directions in the field.
... The first will feature single-cell and single-nucleus transcriptomic data from healthy tissues, combined with spatial analysis. HCA researchers have profiled 58.5 million cells across 15 organ systems to date [13]. Commensurate with their objectives, however, the HCA's Biological Network atlases map discrete tissues, organ systems or focus areas such that, with limited exceptions described here, they do not yield cross-tissue single cell atlases. ...
Preprint
Full-text available
The observation that certain therapeutic strategies for targeting inflammation benefit patients with distinct immune-mediated inflammatory diseases (IMIDs) is exemplified by the success of TNF blockade in conditions including rheumatoid arthritis, ulcerative colitis and skin psoriasis, albeit only for subsets of individuals with each condition. This suggests intersecting "nodes" in inflammatory networks at a molecular and cellular level may drive and/or maintain IMIDs, being “shared” between traditionally distinct diagnoses without mapping neatly to a single clinical phenotype. In line with this proposition, integrative tumour tissue analyses in oncology have highlighted novel cell states acting across diverse cancers, with important implications for precision medicine. Drawing upon advances in the oncology field, this narrative review will first summarise learnings from the Human Cell Atlas in health as a platform for interrogating IMID tissues. It will then review cross-disease studies to date that inform this endeavour, before considering future directions in the field.
... The first will feature single-cell and single-nucleus transcriptomic data from healthy tissues, combined with spatial analysis. HCA researchers have profiled 58.5 million cells across 15 organ systems to date [13]. Commensurate with their objectives, however, the HCA's Biological Network atlases map discrete tissues, organ systems or focus areas such that, with limited exceptions described here, they do not yield cross-tissue single cell atlases. ...
Preprint
Full-text available
The observation that certain therapeutic strategies for targeting inflammation benefit patients with distinct immune-mediated inflammatory diseases (IMIDs) is exemplified by the success of TNF blockade in conditions including rheumatoid arthritis, ulcerative colitis and skin psoriasis, albeit only for subsets of individuals with each condition. This suggests intersecting "nodes" in inflammatory networks at a molecular and cellular level may drive and/or maintain IMIDs, being “shared” between traditionally distinct diagnoses without mapping neatly to a single clinical phenotype. In line with this proposition, integrative tumour tissue analyses in oncology have highlighted novel cell states acting across diverse cancers, with important implications for precision medicine. Drawing upon advances in the oncology field, this narrative review will first summarise learnings from the Human Cell Atlas in health as a platform for interrogating IMID tissues. It will then review cross-disease studies to date that inform this endeavour, before considering future directions in the field.
... Public datasets of scRNA-seq are increasing through international collaborative efforts such as the Human Cell Atlas (HCA) charting the cell types in the healthy body, across time from fetal development to adulthood, and eventually to old age 3 . There has been little attempt to integrate the scRNA-seq data from different sources 4 . ...
Article
Full-text available
Despite the wealth of publicly available single-cell datasets, our understanding of distinct resident immune cells and their unique features in diverse human organs remains limited. To address this, we compiled a meta-analysis dataset of 114,275 CD45+ immune cells sourced from 14 organs in healthy donors. While the transcriptome of immune cells remains relatively consistent across organs, our analysis has unveiled organ-specific gene expression differences (GTPX3 in kidney, DNTT and ACVR2B in thymus). These alterations are linked to different transcriptional factor activities and pathways including metabolism. TNF-α signaling through the NFkB pathway was found in several organs and immune compartments. The presence of distinct expression profiles for NFkB family genes and their target genes, including cytokines, underscores their pivotal role in cell positioning. Taken together, immune cells serve a dual role: safeguarding the organs and dynamically adjusting to the intricacies of the host organ environment, thereby actively contributing to its functionality and overall homeostasis.
... The building block of complex tissues is the diverse range of cell types [1][2][3][4]. Knowing the identity and spatial location of cells from different cell types is the key for understanding how they communicate with each other to carry out specific functions and how diseases emerge when this complex network of interactions goes awry [5][6][7][8][9][10][11]. Single-cell RNA sequencing (scRNA-seq) provides a powerful tool to study the identity of cell types and cell states [12][13][14][15][16][17]. However, the spatial information is lost due to cell disassociation during library preparation. ...
Article
Full-text available
Targeted spatial transcriptomics hold particular promise in analyzing complex tissues. Most such methods, however, measure only a limited panel of transcripts, which need to be selected in advance to inform on the cell types or processes being studied. A limitation of existing gene selection methods is their reliance on scRNA-seq data, ignoring platform effects between technologies. Here we describe gpsFISH, a computational method performing gene selection through optimizing detection of known cell types. By modeling and adjusting for platform effects, gpsFISH outperforms other methods. Furthermore, gpsFISH can incorporate cell type hierarchies and custom gene preferences to accommodate diverse design requirements. Supplementary Information The online version contains supplementary material available at 10.1186/s13059-024-03174-1.
... As these technologies became more broadly available to researchers worldwide, they have led to tremendous biological insights into cardiovascular diseases, including atherosclerosis [16,17], vascular calcification [18], kidney [19][20][21], and heart disease [11-13, 15, 22, 23] and transformed our understanding of cellular heterogeneity, differentiation trajectories, and plasticity. The impact of these technologies is highlighted by the initiation of consortium-based research projects like the Human Cell Atlas (HCA) [24,25] or the Human BioMolecular Atlas Program (HuBMAP) [26], which primarily focus on creating a comprehensive cellular map of the human body detailing the location, function, and characteristics of each cell type in the different tissues. However, it has become clear that for a comprehensive understanding of intrinsic and extrinsic factors which control cell fate decisions in tissues, it is crucial to consider and include spatial molecular information. ...
Article
Full-text available
Spatial multi-omic studies have emerged as a promising approach to comprehensively analyze cells in tissues, enabling the joint analysis of multiple data modalities like transcriptome, epigenome, proteome, and metabolome in parallel or even the same tissue section. This review focuses on the recent advancements in spatial multi-omics technologies, including novel data modalities and computational approaches. We discuss the advancements in low-resolution and high-resolution spatial multi-omics methods which can resolve up to 10,000 of individual molecules at subcellular level. By applying and integrating these techniques, researchers have recently gained valuable insights into the molecular circuits and mechanisms which govern cell biology along the cardiovascular disease spectrum. We provide an overview of current data analysis approaches, with a focus on data integration of multi-omic datasets, highlighting strengths and weaknesses of various computational pipelines. These tools play a crucial role in analyzing and interpreting spatial multi-omics datasets, facilitating the discovery of new findings, and enhancing translational cardiovascular research. Despite nontrivial challenges, such as the need for standardization of experimental setups, data analysis, and improved computational tools, the application of spatial multi-omics holds tremendous potential in revolutionizing our understanding of human disease processes and the identification of novel biomarkers and therapeutic targets. Exciting opportunities lie ahead for the spatial multi-omics field and will likely contribute to the advancement of personalized medicine for cardiovascular diseases.
... In addition, global scientific communities and consortia such as The Cancer Genome Atlas (TCGA) (Tomczak et al., 2015), the International Cancer Genome Consortium (ICGC) (International Cancer Genome et al., 2010), BLUEPRINT (Martens and Stunnenberg, 2013), Human Cell Atlas (HCA) (Lindeboom et al., 2021), etc., make relevant results available to everyone by publishing omics data and metadata, giving the opportunity for further exploration and data integration. This vast amount of complex omics data can be analyzed with machine learning (ML) algorithms to uncover biomarkers or predictive signatures for better patient stratification and treatment selection. ...
Article
Full-text available
Chronic lymphocytic leukemia is a complex and heterogeneous hematological malignancy. The advance of high-throughput multi-omics technologies has significantly influenced chronic lymphocytic leukemia research and paved the way for precision medicine approaches. In this review, we explore the role of machine learning in the analysis of multi-omics data in this hematological malignancy. We discuss recent literature on different machine learning models applied to single omic studies in chronic lymphocytic leukemia, with a special focus on the potential contributions to precision medicine. Finally, we highlight the recently published machine learning applications in multi-omics data in this area of research as well as their potential and limitations.
Article
Single-cell T cell and B cell antigen receptor-sequencing data analysis can potentially perform in-depth assessments of adaptive immune cells that inform on understanding immune cell development to tracking clonal expansion in disease and therapy. However, it has been extremely challenging to analyze and interpret T cells and B cells and their adaptive immune receptor repertoires at the single-cell level due to not only the complexity of the data but also the underlying biology. In this Review, we delve into the computational breakthroughs that have transformed the analysis of single-cell T cell and B cell antigen receptor-sequencing data.
Article
Full-text available
Recent advancements in single‐cell technologies have led to rapid developments in the construction of cell atlases. These atlases have the potential to provide detailed information about every cell type in different organisms, enabling the characterization of cellular diversity at the single‐cell level. Global efforts in developing comprehensive cell atlases have profound implications for both basic research and clinical applications. This review provides a broad overview of the cellular diversity and dynamics across various biological systems. In addition, the incorporation of machine learning techniques into cell atlas analyses opens up exciting prospects for the field of integrative biology.
Article
Full-text available
Background & aims: Molecular evidence of cellular heterogeneity in the human exocrine pancreas has not been yet established because of the local concentration and cascade of hydrolytic enzymes that can rapidly degrade cells and RNA upon pancreatic resection. We sought to better understand the heterogeneity and cellular composition of the pancreas in neonates and adults in healthy and diseased conditions using single-cell-sequencing approaches. Methods: We innovated single-nucleus RNA-sequencing protocols and profiled more than 120,000 cells from pancreata of adult and neonatal human donors. We validated the single-nucleus findings using RNA fluorescence in situ hybridization, in situ sequencing, and computational approaches. Results: We created the first comprehensive atlas of human pancreas cells, to our knowledge, including epithelial and nonepithelial constituents, and uncovered 3 distinct acinar cell types, with possible implications for homeostatic and inflammatory processes of the pancreas. The comparison with neonatal single-nucleus-sequencing data showed a different cellular composition of the endocrine tissue, highlighting the tissue dynamics occurring during development. By applying spatial cartography, involving cell proximity mapping through in situ sequencing, we found evidence of specific cell type neighborhoods, dynamic topographies in the endocrine and exocrine pancreas, and principles of morphologic organization of the organ. Furthermore, similar analyses in chronic pancreatitis biopsy samples showed the presence of acinar-REG+ cells, a reciprocal association between macrophages and activated stellate cells, and a new potential role of tuft cells in this disease. Conclusions: Our human pancreas cell atlas can be interrogated to understand pancreatic cell biology and provides a crucial reference set for comparisons with diseased tissue samples to map the cellular foundations of pancreatic diseases.
Article
Full-text available
Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1–9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.
Article
Full-text available
Cardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and therapeutic strategies require deeper understanding of the healthy heart's molecular processes. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavor. Here, using state-of-the-art analyses of large-scale single-cell and nuclei transcriptomes, we characterise six anatomical adult heart regions. Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes, and fibroblasts, revealing distinct atrial and ventricular subsets with diverse developmental origins and specialized properties. We define the complexity of the cardiac vasculature and its changes along the arterio-venous axis. In the immune compartment we identify cardiac resident macrophages with inflammatory and protective transcriptional signatures. Further, inference of cell-cell interactions highlight different macrophage-fibroblast-cardiomyocyte networks between atria and ventricles that are distinct from skeletal muscle. Our human cardiac cell atlas improves our understanding of the human heart and provides a healthy reference for future studies.
Preprint
Full-text available
To meet the growing demands from scientists to effectively extract deep insights from single cell RNA-seq datasets, we developed cellxgene VIP, a frontend interactive visualization plugin to cellxgene framework, which directly interacts with in-memory data to generate a comprehensive set of plots in high resolution, perform advanced analysis, and make data downloadable for further analysis. It makes large scale scRNA-seq data visualization and analysis more accessible and reproducible with the potential to become an ecosystem for the scientific community to contribute even more modules to the Swiss knife of scRNA-seq data exploration tool.
Article
Full-text available
Single-cell genomics is essential to chart tumor ecosystems. Although single-cell RNA-Seq (scRNA-Seq) profiles RNA from cells dissociated from fresh tumors, single-nucleus RNA-Seq (snRNA-Seq) is needed to profile frozen or hard-to-dissociate tumors. Each requires customization to different tissue and tumor types, posing a barrier to adoption. Here, we have developed a systematic toolbox for profiling fresh and frozen clinical tumor samples using scRNA-Seq and snRNA-Seq, respectively. We analyzed 216,490 cells and nuclei from 40 samples across 23 specimens spanning eight tumor types of varying tissue and sample characteristics. We evaluated protocols by cell and nucleus quality, recovery rate and cellular composition. scRNA-Seq and snRNA-Seq from matched samples recovered the same cell types, but at different proportions. Our work provides guidance for studies in a broad range of tumors, including criteria for testing and selecting methods from the toolbox for other tumors, thus paving the way for charting tumor atlases. A set of ready-to-use tools for profiling fresh and frozen clinical tumor samples using scRNA-Seq and snRNA-Seq facilitates the implementation of single-cell technologies in clinical settings and the construction of single-cell tumor atlases.
Article
The Human Cell Atlas has been undergoing a massive effort to support global scientific equity. The co-leaders of its Equity Working Group share some lessons learned in the process.
Article
The enteric nervous system (ENS) coordinates diverse functions in the intestine but has eluded comprehensive molecular characterization because of the rarity and diversity of cells. Here we develop two methods to profile the ENS of adult mice and humans at single-cell resolution: RAISIN RNA-seq for profiling intact nuclei with ribosome-bound mRNA and MIRACL-seq for label-free enrichment of rare cell types by droplet-based profiling. The 1,187,535 nuclei in our mouse atlas include 5,068 neurons from the ileum and colon, revealing extraordinary neuron diversity. We highlight circadian expression changes in enteric neurons, show that disease-related genes are dysregulated with aging, and identify differences between the ileum and proximal/distal colon. In humans, we profile 436,202 nuclei, recovering 1,445 neurons, and identify conserved and species-specific transcriptional programs and putative neuro-epithelial, neuro-stromal, and neuro-immune interactions. The human ENS expresses risk genes for neuropathic, inflammatory, and extra-intestinal diseases, suggesting neuronal contributions to disease.
Article
The SARS coronavirus 2 (SARS-CoV-2) pandemic is a global challenge, which the scientific community has tackled by mounting extensive global collaborations, combining expertise in diverse areas and applying cutting-edge techniques, to dissect mechanisms underlying coronavirus disease-19 (COVID-19) pathology. The Human Cell Atlas consortium is emerging as an important contributor to furthering this progress. The Human Cell Atlas consortium in collaborative work on COVID-19
Article
Crucial transitions in cancer—including tumor initiation, local expansion, metastasis, and therapeutic resistance—involve complex interactions between cells within the dynamic tumor ecosystem. Transformative single-cell genomics technologies and spatial multiplex in situ methods now provide an opportunity to interrogate this complexity at unprecedented resolution. The Human Tumor Atlas Network (HTAN), part of the National Cancer Institute (NCI) Cancer Moonshot Initiative, will establish a clinical, experimental, computational, and organizational framework to generate informative and accessible three-dimensional atlases of cancer transitions for a diverse set of tumor types. This effort complements both ongoing efforts to map healthy organs and previous large-scale cancer genomics approaches focused on bulk sequencing at a single point in time. Generating single-cell, multiparametric, longitudinal atlases and integrating them with clinical outcomes should help identify novel predictive biomarkers and features as well as therapeutically relevant cell types, cell states, and cellular interactions across transitions. The resulting tumor atlases should have a profound impact on our understanding of cancer biology and have the potential to improve cancer detection, prevention, and therapeutic discovery for better precision-medicine treatments of cancer patients and those at risk for cancer.
Article
Thymus development, cell by cell The human thymus is the organ responsible for the maturation of many types of T cells, which are immune cells that protect us from infection. However, it is not well known how these cells develop with a full immune complement that contains the necessary variation to protect us from a variety of pathogens. By performing single-cell RNA sequencing on more than 250,000 cells, Park et al. examined the changes that occur in the thymus over the course of a human life. They found that development occurs in a coordinated manner among immune cells and with their developmental microenvironment. These data allowed for the creation of models of how T cells with different specific immune functions develop in humans. Science , this issue p. eaay3224