You are on page 1of 13

REvIEwS

Immune monitoring using mass


cytometry and related high-
dimensional imaging approaches
Felix J. Hartmann    and Sean C. Bendall   *
Abstract | The cellular complexity and functional diversity of the human immune system
necessitate the use of high-dimensional single-cell tools to uncover its role in multifaceted
diseases such as rheumatic diseases, as well as other autoimmune and inflammatory disorders.
Proteomic technologies that use elemental (heavy metal) reporter ions, such as mass cytometry
(also known as CyTOF) and analogous high-dimensional imaging approaches (including
multiplexed ion beam imaging (MIBI) and imaging mass cytometry (IMC)), have been developed
from their low-dimensional counterparts, flow cytometry and immunohistochemistry , to meet
this need. A growing number of studies have been published that use these technologies to
identify functional biomarkers and therapeutic targets in rheumatic diseases, but the full
potential of their application to rheumatic disease research has yet to be fulfilled. This Review
introduces the underlying technologies for high-dimensional immune monitoring and discusses
aspects necessary for their successful implementation, including study design principles,
analytical tools and future developments for the field of rheumatology.

Technological advances have enabled scientific insights as well as in autoimmunity in general. The pathogenic
across many areas of research. Given the complexity of processes involved in rheumatic diseases, such as inter-
the human immune system, immunology has particu- actions between external factors and innate and adaptive
larly benefited from innovations in various single-cell immune responses, ongoing crosstalk and communica-
technologies1. These technologies facilitate in-depth tion within the immune system and the pathogenic prop-
study of the composition and activation of immune cell erties of certain subsets of immune cells, are well-suited
populations and their relation to disease. However, the to systems-level analyses3–5. Such analyses could enable
complex composition of the human immune system, the discovery of novel biomarkers to predict treatment
the numerous functional states of these cells and their response or to guide therapeutic decision making6,7
diverse localizations require high-dimensional method- and the identification of therapeutic targets that pres-
ologies for their assessment in comprehensive immune ent a reduced risk of infection compared with broadly
monitoring studies. In this context, ‘comprehensive immunosuppressive therapy, and could also address the
immune monitoring’ refers to the ability to robustly challenge presented by patients who do not respond to
identify cells from all of the main immune lineages currently available therapeutics. Importantly, biomarkers
(including T cells, B cells and monocytes) and their most identified by this approach are often not only predictive,
important functional subsets, and to assign all cells in but also intrinsically linked to disease mechanisms, and
a given sample to these lineages and subsets2. Notably, can therefore also be considered as therapeutic targets8–10.
robustly determining the absence of immune popula- Proteomic technologies that use elemental (heavy metal)
tions can also provide valuable insight, for example by reporter ions, such as mass cytometry (also known as
revealing populations of cells with altered trafficking cytometry by time-of-flight (CyTOF)) and the closely
Department of Pathology, properties or for monitoring the success of cell-depleting related imaging technologies multiplexed ion beam
School of Medicine, Stanford antibody therapies. imaging (MIBI) and imaging mass cytometry (IMC),
University, Palo Alto,
CA, USA.
Immune system dysregulation and aberrant acti- combine high-dimensional multiplexing capabilities
vation are thought to be crucial in the development of with high-throughput to facilitate the analysis of samples
*e-mail: bendall@
stanford.edu various rheumatic diseases, including rheumatoid arthri- across large cohorts, making these technologies particu-
https://doi.org/10.1038/ tis (RA), systemic lupus erythematosus (SLE), primary larly suited for the comprehensive immune monitoring
s41584-019-0338-z Sjögren syndrome (pSS) and systemic sclerosis (SSc), of biospecimens from patients with rheumatic diseases11.

Nature Reviews | Rheumatology


Reviews

Key points immune cell sub-classes, as well as important func-


tional cell states, including age-associated B cells
• Immune monitoring of human cells using systems immunology approaches has the (ABCs)13 and various disease-associated T cell subsets14.
potential to produce new insights into pathological processes and therapeutic However, the physical limitations of fluorescence-based
opportunities for rheumatic disease research. cytometry impair the addition of further experimen-
• Proteomic approaches that use elemental (heavy metal) reporter ions, such as mass tal parameters, thus prohibiting the comprehensive
cytometry and high-dimensional imaging techniques, might be of value for the study capturing of the whole immune state simultaneously.
of a wide variety of clinical samples.
One solution to this problem was found by substi-
• Mass cytometry enables in-depth analysis of the phenotype and functional state of tuting the fluorescence-based reporters used in flow
immune cells at the single-cell level.
cytometry for non-biological elemental isotopes and an
• High-dimensional imaging techniques use concepts analogous to mass cytometry to inductively coupled plasma (ICP) mass spectrometry-based
image cells in their histological context, providing spatial and cell–cell interaction
readout, creating a technique termed mass cytom­etry15–17.
information.
Some of the main characteristics of flow cytometry and
• A combination of these technologies with data-driven analytical approaches can give
mass cytometry are directly compared in Table 1.
predictive insights into disease mechanisms for rheumatic diseases.

How mass cytometry works. In mass cytometry, antibod-


In this Review, we introduce the underlying technol- ies are tagged with elemental (heavy metal) reporter ions
ogies of mass cytometry and related high-dimensional via covalent conjugation with chelating polymers18. After
imaging technologies, as well as improvements that staining with these tagged antibodies, single-cell suspen-
have accelerated their adoption in clinical settings. sions are introduced into the CyTOF analyser where they
Furthermore, we provide a practical guide to the design are first nebulized into droplets and subsequently intro-
of immune monitoring studies and the comprehensive duced into an ICP that vaporizes, atomizes and ionizes
and unbiased analysis of the datasets these studies pro- the sample (Fig. 1). The resulting ion cloud is then filtered
duce. We conclude by discussing examples of the success- by mass to remove biologically abundant, low-mass ion
ful application of these technologies in rheumatic disease species and finally analysed by time-of-flight (TOF) mass
research and the future possibilities and challenges spectrometry to quantify the abundance of all the iso-
involved in high-dimensional immune monitoring. topic reporter masses, which enables the quantification
of bound antibodies and therefore the expression of the
Mass cytometry markers of interest19–21.
For many years, immune monitoring, and much of Like conventional flow cytometry, mass cytometry
immunological research, has relied on flow cytometry is well suited to performing single-cell analysis of cell
and immunohistochemistry (IHC) or immunofluo- suspensions obtained from liquid samples such as blood
rescence imaging approaches to capture and quantify or urine, which are easily obtainable from patients with
cellular heterogeneity and its relationship to disease. rheumatic diseases. Although the staining procedures
The low number of parameters that could be analysed and sample handling in mass cytometry and flow cytom-
simultaneously by these methods forced restricted analy­ etry are comparable, issues related to spectral overlap
ses in terms of cellular properties and the composition between different analysis channels (Box 1) are reduced in
of cell populations. Consequently, in most experiments, mass cytometry as the heavy-metal isotopes used to tag
researchers could only target a single cell type, and addi- antibodies possess non-overlapping atomic masses that
tional answers could only be gained with the sacrifice of can be accurately resolved and quantified. In addition,
time and clinical material12. the availability of many different heavy metal isotopes
In flow cytometry, the development of novel fluoro- increases the multiplexing capacity of mass cytometry to,
phores and laser systems has enabled increased para­ theoretically, 120 separate analysis channels. Currently
meter­ization, which has led to the discovery of new available conjugation and chelation chemistry enables

Table 1 | Comparison of flow cytometry and mass cytometry


Characteristic Flow cytometry mass cytometry
Measurement basis Fluorescent probes Stable mass isotope probes
Sources of nonspecific signal (% of Spectral overlap (10–50%), autofluorescence Isotopic impurity , spectral
Inductively coupled plasma specific signal) (5–10%) and fluorophore degradation (5–10%) overlap and oxidation (all <5%)
A type of plasma in which the
energy is supplied through Maximum number of measurements 20 (theoretically ~40) 60 (theoretically ~120)
electromagnetic induction
(changes in magnetic fields).
Panel design complexity (number of Simple (<8), moderately complex (8–12) and Simple (<40) and moderately
probes) complex (>12) complex (40–60)
Time-of-flight Relative probe sensitivity (arbitrary 0.1–10 1–3
The time taken by a particle to units)
travel through a medium;
measuring the time-of-flight of Sampling efficiency >95% ~50%
ions in an electric field can be
used to infer the ions’
Number of cells measured 500–40,000 (5,000) 50–1,000 (500)
mass-to-charge ratio and
per second (typical)
therefore its identity. Data from REFS2,22,24,110,160–162.

www.nature.com/nrrheum
Reviews

antibodies to be tagged with ~60 of these heavy metal


isotopes22–24, and additional channels can be used for Antibody 1 Antibody 3
non-antibody probes (see below). Notably, the heavy
metal isotopes used in mass cytometry are not com- Antibody 2 Antibody N
monly found in unstained samples, thus eliminat-
ing biological background noise and issues related to Staining of single-cell suspensions with
antibodies that recognize markers of interest
auto­fluorescence17. Rare exceptions are samples from
patients with cancer who have been treated with the
platinum-containing cytostatic drug cisplatin, or from
individuals who have had medical imaging tests using
gadolinium-containing contrast agents. Interestingly,
residual gadolinium and platinum might be detectable
in cells isolated from these patients shortly after admin-
Introduction of stained single-cell
istration, a fact that has also been leveraged to study the suspension into the mass cytometer
biodistribution of these compounds in patients25–27.
Nebulizer Droplets
Use in immune monitoring. Mass cytometry can be
used to analyse a wide variety of cellular features of
interest. Antibodies specific for cell surface molecules
can be used to ascertain a cell’s lineage and to perform
in-depth phenotyping of heterogeneous cell popu­
Ion cloud ICP
lations2. Furthermore, combining mass cytometry with
metal-conjugated tetramers enables the multiplexed
analysis of a wide array of antigen-specific T cells28–32.
The chemically stable elemental reporters used in
mass cytometry are broadly compatible with com- Droplet nebulization and
plasma-based ionization
mon fixation and permeabilization procedures, which
facilitate the quantification of a variety of intracellular Quadrupole
targets in combination with a cell’s lineage. For exam-
ple, quantification of transcription factors can provide
insight into master regulators and their involvement
in cell fate decisions and the maintenance of lineages33, Orthogonal TOF
intracellular cytokine staining can be used to analyse mass spectrometer
high-dimensional patterns of cytokine co-production
Filtering of low mass ion species
across a wide variety of cell types34,35, and analysis of the and TOF-based quantification
phosphorylation of intracellular proteins provides infor-
mation on functional cell states and the responses of cells
to external stimuli36. Mass signal integration for each measured cell
In addition to the proteome, many other cellular
fea­tures can be assessed by mass cytometry. Cellu­
N-dimensional single-cell data
lar DNA content can be determined through the use
of metal-containing intercalators19 and cellular viabil-
ity through covalent binding of cisplatin or analogous Fig. 1 | analysis of single-cell suspensions by mass
palladium-containing compounds to membrane com- cytometry. In the first step, single-cell suspensions are
promised cells37,38. By making use of mRNA hybridi­ incubated with heavy metal-labelled antibodies and other
zation and rolling circle signal amplification, RNA reporter probes aimed at markers of interest (such as MHC
multimers to detect antigen-specific cells or probes that
transcripts can be analysed by mass cytometry along-
discriminate live cells from dead cells). Antibodies that
side proteins39,40. Mass cytometry can also be used to recognize intracellular proteins can be used in conjunction
reveal cell cycle states; antibodies targeting cyclin B1 and with common fixation and permeabilization protocols.
phosphorylated forms of histone H3 and retinoblastoma Following a series of washing steps, cells are introduced into
protein (Rb) can be used in combination with the incor- the mass cytometer, where the suspension is nebulized
poration of 5-iodo-2-deoxyuridine, which can be directly into small droplets and introduced into an inductively
analysed by CyTOF41. This technique can be extended coupled plasma (ICP), which breaks down the droplet
to simultaneously capture de novo transcriptional and contents into a cloud of elemental ions. Ion species with
translational activity in single cells using 5-bromo-2- low atomic masses (such as hydrogen and carbon) are
uridine and puromycin, respectively42. Reporter probes removed through a mass filter known as a quadrupole,
and the remaining ions (which were conjugated to the
for enzymatic activities43 or cellular states can also be
respective probes) are subsequently quantified using an
used with mass cytometry, such as tellurium-containing orthogonal time-of-flight (TOF) mass spectrometric
probes to detect hypoxia44. Furthermore, mass cytom- detection system. Ion counts are then integrated to derive
etry has been used to assess the phagocytic activity of quantities of bound antibodies, and thus single-cell
differentially polarized myeloid cells by quantifying the abundances of markers of interest. These high-dimensional
uptake of metal-labelled targets by the cells45. data are then exported for further downstream analysis.

Nature Reviews | Rheumatology


Reviews

Box 1 | Sources of nonspecific signal in flow cytometry and mass cytometry


In flow cytometry, a major source of nonspecific signal (in some cases >50% of the specific signal) is spectral overlap160,161.
Fluorophore emission spectra are usually broad and overlap into adjacent analysis channels, resulting in nonspecific signal
in this channel (see the figure). Additional sources of nonspecific signal are cellular autofluorescence and fluorophore
degradation161. Considerable effort has to be invested in panel design to account for these effects and signals have to be
corrected computationally. By contrast, in mass cytometry, antibodies are conjugated to elemental isotopes that have
non-overlapping masses, which can be resolved through their time-of-flight. Minor sources of overlap (usually ~1%) are
isotopic impurities in the metal stocks and oxidation of the metals20. Isotopic impurities that were not completely removed
during the purification process typically generate a signal in the mass + 1 channel, whereas oxidized elements generate
signal in the mass + 16 channel (see the figure). This lack of nonspecific signal facilitates the use of straightforward panel
designs of up to 60 antibodies in mass cytometry.

Flow cytometry Mass cytometry

Filter 1 Filter 2 Mass Mass + 1 Mass + 16

Isotopic impurity
into mass + 1 channel

Spectral overlap into Oxidation into


adjacent channel mass + 16 channel

Wavelength Time-of-flight

Since the inception of mass cytometry, there have paraffin-embedded (FFPE) samples has been the pre-
been numerous improvements that have increased the dominant methodology for the analysis of human tis-
usefulness of this technique in immune monitoring and sue samples, followed by immunofluorescence imaging.
cell phenotyping17. Normalization methods using bead Relying on chromogenic substrates and fluorescence
standards or biological control samples have improved emission, respectively, these technologies have a lim-
the comparability of mass cytometry results across time, ited multiplexing capacity of about four simultaneous
batches and study sites46–48. Similarly, the adoption of measurements under routine conditions. To address this
cell barcoding approaches has enabled the staining, pro- limitation, several approaches have been developed with
cessing and acquisition of multiple samples in a single the aim of increasing the number of parameters that can
composite sample, thus minimizing batch effects and be assessed in solid tissue samples.
enabling the conduction of large-scale screens36,49. Cell One type of approach relies on the serial acquisition
barcoding reduces technical variation between samples, of a small set of markers of interest through many cycles of
reagent consumption and analysis time, which is espe- imaging. Several technologies (such as histocytom­etry,
cially beneficial in clinical settings, in which large quan- multiplex immunofluorescence or tissue-based cyclic
tities of samples need to be compared. Furthermore, immunofluorescence) can be used for this approach,
barcoding improves the identification and exclusion of which usually use fluorescence-labelled antibodies that
cellular doublets, which is a crucial step for single-cell are read in multiple cycles of staining, imaging and
analyses49. Importantly, barcoding can be used with fixed quenching53–55. A variation of this approach that uses
or live samples, including cells of non-haematopoietic DNA barcoding, termed CODEX, enables the staining
origin38,50,51. of tissues with all antibodies at once, followed by several
Taken together, mass cytometry enables in-depth, rounds of imaging and fluorophore reporter removal56.
single-cell characterization of phenotypes and cellular Although these fluorescence-based technologies do have
functions across large groups of samples, which can be the ability to acquire high-dimensional images, certain
used to gain deep insights into the regulation of cellu- limitations and challenges remain. Cyclic staining, with
lar and systems-wide processes that are of relevance for repeated exposure to reporter stripping chemicals, can
disease. lead to changes in epitope accessibility and altered tissue
morphology, which makes the optimization of staining
High-dimensional imaging technologies order challenging and reduces the robustness of cyclic
Rheumatic diseases often manifest in solid tissues, such image stack reassembly. Tissue samples, particularly
Cell barcoding as the synovium in RA or kidney in SLE. Therefore archival FFPE samples, have background autofluores-
A method of labelling cells tissue-based imaging methods can offer a unique cence, which limits assay sensitivity and often requires
with a sample-specific opportunity to investigate cells in their native or patho- additional analytical compensation57. Additionally, chro-
signature that can be used logical context. Spatial analysis provides insight into mogenic and fluorophore dyes are often not chemically
to subsequently pool cells
from several samples for
both the cellular microenvironment and cell–cell inter- stable, which makes their long-term storage challeng-
downstream staining and actions, which are important contributors to cellular ing. The nature of cyclic methods is also incompatible
processing. identity and functional state52. IHC on formalin-fixed with subsequent re-acquisitions under different imaging

www.nature.com/nrrheum
Reviews

Rastering
conditions (such as a first-pass low-resolution scan How IMC and MIBI work. For both technologies, sam-
A pattern of scanning in which followed by a high-resolution acquisition). ples of either FFPE or cryopreserved tissues are mounted
an area is scanned (for example, To address these problems, two related technolo- onto a slide and stained with heavy metal isotope-tagged
with an ion beam) in lines from gies, IMC58 and MIBI59,60, have been developed in which antibodies or other reporter probes in a process simi­lar
side to side, starting at the top.
fluorophores are replaced with elemental mass reporters, to IHC or immunofluorescence protocols. In contrast to
analogous to mass cytometry. most of the cyclic approaches mentioned above, in IMC
or MIBI, samples can be stained with all antibodies and
reporter tags at once. The stained slides are then intro-
duced into the respective analysers. In IMC, a laser sys-
tem is used to ablate the stained tissue pixel-by-pixel58
(Fig. 2). The resulting ablated material is then introduced
Cryopreserved or FFPE tissue sectioning into a CyTOF analyser, where it is ionized through an
ICP and the elemental masses are quantified, similar to
Antibody 1 Antibody 3 the process for whole cells in mass cytometry. Currently
Antibody 2 Antibody N
available commercial IMC instruments provide an image
resolution of 1 µm and can acquire ~100 such 1 µm pix-
Simultaneous staining of tissue sections with all antibodies els every second61,62. In terms of sensitivity, a minimum
of ~50 copies of an epitope per pixel are required for
its detection.
Imaging mass cytometry Multiplexed ion beam imaging
In contrast to laser ablation-based IMC, MIBI utilizes
the principle of secondary ionization mass spectrometry.
For MIBI, stained slides are introduced into an analysis
Released Secondary ions chamber under vacuum, in which elemental reporters
Laser
particles Primary are released by rastering the tissue pixel-by-pixel with a
ion beam primary ion beam (Fig. 2). As the primary ions collide
with the sample, they liberate secondary ions (including
the heavy metal isotope reporters) from the tissue. These
Stained tissue
secondary ions are directly introduced into the detection
Ablation of tissue with laser beam Rastering of tissue with primary ion system without further ionization or transfer, which pro-
releases particles beam releases secondary ions duces a sensitivity of as little as a single copy of an epitope
per pixel. Whereas the first MIBI instruments made use
of a magnetic sector mass spectrometer59, next-generation
ICP Ion cloud
instruments make use of a full TOF detection system
(termed MIBI-TOF)60 that enables a large range of atomic
masses to be analysed, including naturally occurring ele-
ments such as phosphorus and iron, which have impor-
tant biological functions63. These current MIBI-TOF
Sample Orthogonal TOF Primary instruments can provide pixel resolutions of ~250 nm at
input mass spectrometer ion source Sample a rate as high as 10,000 pixels every second60,63.
Quantification of ions in ablated Pixel-based quantification of secondary
tissue via TOF mass spectrometry ions via TOF mass spectrometry Use in immune monitoring. A wide variety of the com-
mercially available antibodies used in IHC and immuno­
fluorescence staining procedures can be adapted for use
Mass signal integration for each pixel Mass signal integration for each pixel
in IMC and MIBI, as well as some of the alternative
reporter probes developed for mass cytometry such as
N-dimensional images N-dimensional images those that detect mRNA64. A ruthenium-based dye has
been used to identify tissue structures and extracellu-
Fig. 2 | high-dimensional imaging analysis of tissue sections. Tissue sections (from
formalin-fixed paraffin embedded (FFPE) or cryopreserved samples) are placed on a lar features, resulting in counterstaining analogous to
slide and subsequently stained with all heavy metal-tagged antibodies and other that produced by haematoxylin in IHC65. The in situ
reporter probes in a single staining step. In imaging mass cytometry (IMC), stained detection of antigen-specific T cells has been previously
sections are placed in the IMC analyser. A laser ablates the tissue pixel-by-pixel, demonstrated in both IHC and immunofluorescence
releasing particles that are then introduced into a mass cytometer analyser, where they imaging using tetramers66–68, so should therefore be also
are first ionized in an inductively coupled plasma (ICP) and then quantified in an feasible by IMC or MIBI. However, whereas laser-based
orthogonal time-of-flight (TOF) mass spectrometer. In multiplexed ion beam imaging IMC completely ablates the tissue at each pixel thus pre-
(MIBI), stained sections are placed into the MIBI analyser and introduced into a vacuum venting reanalysis, only a few hundred nanometres of
chamber. The MIBI instrument then rasters the tissue sections with a primary ion beam. tissue are liberated with each MIBI scan60. This minimal
Upon collision with the stained section, secondary ions (including those introduced
tissue destruction enables the same cells in the same tis-
through the binding of the heavy metal-tagged antibodies) are liberated from the
sample. This cloud of secondary ions is then focused through a series of lenses and sue section to be re-analysed multiple times; for example,
introduced into an orthogonal TOF mass spectrometer. For both techniques, the to create low-resolution overviews of large tissue areas,
detected signal is integrated and translated into ion counts per pixel, which represents identify specific regions of interest and subsequently
multidimensional images that can be directly visualized or further analysed using perform high-resolution analysis of these areas, or even
various image analysis pipelines. to create 3D reconstructions60.

Nature Reviews | Rheumatology


Reviews

Important factors for the implementation of these (such as calcium-containing crystals), and also subcellu-
technologies in clinical settings that involve large groups lar structures (including nuclei and other cell compart-
of patients are the throughput and robustness of these ments), can be automatically identified73. Furthermore,
platforms. Two 2019 publications reported the use of information about cellular location and proximity to
IMC to create image-based maps of the pancreas or other cells can be assigned to segmented data and lev-
pancreatic islets from patients with type 1 diabetes mel- eraged in the downstream analysis. Examples of the
litus61,62. In these studies, samples were acquired from application of such analyses to high-dimensional images
12 to 18 patients, and multiple areas within each sample include: the use of Delaunay triangulation79 to identify
were analysed, demonstrating the suitability of IMC for cellular neighbourhoods in a mouse model of lupus56;
such clinical studies. Similarly, a 2018 study demon- the use of spatial enrichment analysis to identify a hier-
strated the throughput and robustness of MIBI63. In this archical organization of tumour and immune cells in
study, 800 µm2 images (containing 2,048 × 2,048 pixels) triple-negative breast cancer63; and the use of cellular
of FFPE samples were acquired from each of 41 patients neighbourhood analysis on pancreatic islets to identify
with triple-negative breast cancer in less than 2 weeks63. disease stage-specific cellular interactions in patients
New MIBI-TOF instruments and ion sources further with type 1 diabetes mellitus61.
increase this throughput by another order of magni-
tude60. Overall, high-dimensional imaging methods such Visualization of high-dimensional single-cell data.
as IMC and MIBI provide interesting possibilities for the Single-cell proteomic data (either directly obtained
study of rheumatic diseases, including the potential to by mass cytometry or through image segmentation as
identify relevant cellular interactions and perturbations described above) is usually transformed using the hyper-
directly in the tissue microenvironment. bolic function arcsinh or a related logarithmic approach
to account for variance in protein expression between
Data analysis cell populations80–82. Following this pre-processing step,
The analysis of highly multiplexed datasets comprising high-dimensional datasets are often projected into an
dozens of measurements from large groups of samples interpretable low-dimensional space, with the aim of
provides a unique opportunity to gain insight into bio- preserving as much of the inherent high-dimensional
logical variation and its importance in a therapeutic structure as possible. An effective approach that is opti-
Image segmentation
context. However, such datasets also pose additional mized to group cells on the basis of the similarity of their
The process of identifying and
partitioning an image into challenges as their high-dimensional properties make high-dimensional expression profiles is the t-stochastic
meaningful objects (such as comprehensive manual analysis (such as analysis in 2D neighbour embedding (tSNE) algorithm83,84. tSNE proj­
cells) in order to facilitate their space using manual gating approaches) highly inefficient. ects high-dimensional data onto a (usually) 2D map,
downstream analysis. In the following section, we provide an introduction to thus providing a readily interpretable overview of the
Deep learning
some commonly employed analytic approaches that can cell populations contained in the dataset. The tSNE
A type of machine learning in be used as a starting point for the comprehensive analysis algorithm has been successfully applied to a wide range
which artificial neural of datasets from such high-dimensional immune mon- of single-cell measurements and is now integrated into
networks with multiple layers itoring studies69–72. The analytical tools discussed below many cytometry data-processing platforms85.
of adjustable nodes are used
should provide researchers with interpretable data rep- Several extensions and modifications of the original
to learn how to perform
specific tasks from large resentations and correlations with clinical features, thus tSNE algorithm have further broadened its applicabil-
amounts of data. enabling the identification of cellular disease signatures ity86–88. Real-time visualization of the two-dimensional
and potential novel therapeutic targets. projections provide users with feedback that can be
Classifier used to interactively optimize parameters87 and a hier-
An algorithm that has been
trained to predict the class of
Image segmentation and analysis of spatial relationships. archical step-wise application of tSNE can improve the
data points. As a first level of analysis, multiplexed imaging data can identification of rare cell populations88. Furthermore
be visualized in its native image format or as multi-colour artificial neural networks can approximate the tSNE
Artificial neural networks overlays to emphasize the spatial relationships between embedding function, thereby enabling the projection
A type of machine learning
different epitopes and cell types. Beyond this basic analy­ and comparison of additional data that were not part of
framework inspired by the
biological structure of sis, several approaches can be used to extract single-cell the initial embedding89. Alternatively, a novel embed-
the brain, in which (potentially data from high-dimensional images. Image segmentation ding technique, termed uniform manifold approxi-
many) layers of interconnected approaches that use deep learning techniques73 can be mation and projection (UMAP), has been proposed
nodes transmit information applied to MIBI-TOF and IMC images63,74. For example, for dimension reduction 90. Compared with tSNE,
to each other and apply
transformations to perform
a classifier can be trained to discriminate nuclear from UMAP improves the preservation of global structure in
classification or prediction non-nuclear pixels, which is then used to automatically high-dimensional datasets and provides shorter calcu-
tasks. identify nuclei and thus cells in a large set of images. lation times, factors that have contributed to the rapid
Other deep learning approaches, including tools such as adoption of UMAP90,91.
Minimum spanning tree
Ilastik75 and CellProfiler Analyst76, have been proposed A different approach that is particularly suited to
In a graph consisting of points
(nodes) connected through for cellular segmentation of high-dimensional image simultaneously comparing the characteristics of mul-
edges, the minimum spanning sets61,62,77,78. Once imaging data have been segmented, tiple cell populations within a set of samples is the
tree represents the subset of downstream analysis can be performed analogously to tool spanning-tree progression analysis of density-
the graph that connects all other types of single-cell data. Notably, image segmen- normalized events (SPADE)92. SPADE provides an over-
nodes with the minimum total
edge weight, usually
tation is not limited to the identification of single cells; view of all cell populations present in a sample as clusters
representing the length of large structures, including multicellular tissues (such as on a minimum spanning tree, which can be colour-coded
the edge. pancreatic islets) and extracellular pathological deposits for any marker of interest or for differential expression

www.nature.com/nrrheum
Reviews

when one or more reference samples are included85. Planning an immune monitoring study
Other data analysis and visualization tools make use of The conception and planning of a large immune moni-
force-directed layouts to provide low-dimensional rep- toring study involves several layers of complexity, includ-
resentations of a high-dimensional dataset. For exam- ing the choice of appropriately sized experimental and
ple, the VorteX clustering environment tool combines control groups. Much of this complexity is dependent on
a clustering algorithm with a single-cell representation the specific disease context and biological hypo­theses
of cellular heterogeneity through force-directed lay- being investigated. Given this context-dependency, such
outs93. The Flowmap tool follows a similar concept, considerations cannot be comprehensively covered in this
except that it incorporates the use of time-course data Review. Instead, we provide a set of practical considera-
when building the model94. Prior biological knowl- tions that cover questions that are commonly encountered
edge about lineage-defining expression patterns can be when planning studies involving mass cytometry (Fig. 3),
incorporated through the use of maps generated by the and that often also arise when using high-dimensional
SCAFFoLD tool33. In this approach, high-dimensional imaging techniques.
datasets are clustered and subsequently mapped around
previously defined landmark cell populations in a Sample collection. Studies involving patients and healthy
force-directed layout, which enables the integration of individuals often use existing collections of samples that
new data from different platforms and comparison to have been acquired over several years. Cell suspensions
previous analyses95. that have been viably cryopreserved can be directly
entered into the sample preparation pipeline for mass
Cell population identification and analysis. Many algo- cytometry. Similarly, archival FFPE tissue samples from
rithms have been developed for automated population standard pathology workflows are compatible with
identification through clustering96 to improve the repro- IMC and MIBI63. If sample collection is ongoing, sev-
ducibility of high-dimensional dataset analysis and eral aspects of storage and analysis should be considered.
reduce analysis-related variability. A prominent example Single-cell suspensions can be stored viably for use in
that has high performance and computational efficiency mass cytometry, which enables functional assays to be
is FlowSOM, an algorithm that uses an adaption of the performed, such as intracellular cytokine production
principle of self-organizing maps for cytometry data97. assays or protein phosphorylation assays. Fixation prior
Following population assignment, potential associations to storage can help preserve cryo-sensitive cells and pre-
of population frequencies or expression characteristics vent freeze–thaw-dependent changes in their functional
can be derived. To statistically compare cluster charac- state; however, some epitopes can become less accessible
teristics between clinical groups or biological scenarios, for antibody staining after fixation, thereby potentially
the CITRUS algorithm uses a hierarchical clustering reducing the overall analytical quality of a sample2. It is
step and subsequent regularized regression analysis to also advisable to anticipate the inclusion of a separate val-
identify features that can stratify cohorts98; a similar idation cohort by gathering samples not included in the
approach has also been integrated into complementary initial analysis, which can be subsequently used to inde-
visualization tools such as SCAFFoLD99. pendently confirm initial findings. Importantly, mass
Force-directed layouts
Although clustering can be a useful approach to cytometry is destructive and cells cannot be recovered
Graphical renderings that
assign spring-like (attractive identify groups of cells and cell populations, questions after acquisition; however, there are numerous examples
and repulsive) forces between arise as to the choice of clustering parameters, the of studies in which cell populations of interest have been
the edges and nodes of a definition and correct number of clusters in the data- identified in high-dimensional space by mass cytometry
graph to position them in sets and the potential loss of statistical power through and prospectively isolated by fluorescence-activated cell
2D space.
over-clustering. Therefore, several approaches have sorting for subsequent orthogonal analysis using a dif-
Clustering been developed to identify differentially abundant cells ferent set of methodologies8,102,107,108. Similarly for MIBI,
Grouping a set of points that or stratifying signatures without the need for clustering. serial sections of the same tissue samples could be used
are similar to each other. One solution is to assign cells to so-called hyperspheres, for confirmatory orthogonal assays such as traditional
which represent high-dimensional geometries, and IHC or bulk biomolecule analysis.
Self-organizing maps
A type of unsupervised then test for differential abundances for each hyper-
clustering and dimensionality sphere100. Alternatively, an approach termed CellCNN Antibody panel design. Once the samples are curated,
reduction approach that relies on representation learning to identify cell events questions around intra-assay comparison, panel design
preserves the topological associated with differential clinical outcomes or other and the choice of antibodies and other probes have to
information of the input data.
characteristics of interest101. be considered109. A 2019 study validated a commer-
Representation learning Cell development and lineage decisions can also be cially available set of antibodies that aims to capture and
The automated process of traced in mass cytometry data using trajectory algo- quantify a broad range of immune cell types beyond the
transforming raw data into rithms, which order cells according to progressive hypothesized target population(s), which could be easily
useful features that are
changes in their phenotype. Early examples of such algo- implemented for comprehensive immune monitoring in
subsequently used in other
machine learning applications. rithms for mass cytometry data were Wanderlust102 and rheumatic disease research2. Such a reference panel could
Wishbone103; however, a large number of such trajectory then be supplemented with further hypothesis-specific
Simpson’s diversity index algorithms are now available104. In addition, single-cell markers. Although the list of commercially available
A measurement of diversity regulatory networks can be modelled and quantified heavy metal-tagged antibodies is steadily growing, it is
that takes into account the
number of different groups
using a conditional density-based approach termed usually still necessary to conjugate some antibodies in-
present in a dataset, as well DREMI/DREVI105, and cellular diversity can be quanti- house. Protocols for the conjugation of antibodies with
as their relative abundance. fied by calculating an inverse Simpson’s diversity index106. heavy metals that can be used in mass cytometry or

Nature Reviews | Rheumatology


Reviews

in comparison with fluorescence-based signal over-


lap. Moreover, such contamination can be pra­ctically
resolved through panel design, reagent titration or
Patient cohort through bead-based compensation app­roaches (that
can be applied to both mass cytometry and high-
dimensional imaging approaches), further improving
data accuracy113.

Sample collection (different Reproducibility. As with many other biological assays,


treatments and time-points) antibody specificity should always be confirmed using
both positive and negative biological controls (samples
known to either express or not express the epitope of
Sample 1 Sample 2 Sample X
interest) and batch processing is recommended to min-
imize technical variation. For mass cytometry, the use
of barcoding approaches enables the combined stain-
ing and acquisition of multiple samples at the same
Single-cell suspensions
time36,38,49–51 (Fig. 3). Cells can then be assigned back to
their original sample in silico. An analogous approach
Sample 1 Sample 2 Sample X to barcoding for high-dimensional imaging is the use
of tissue microarrays, which combine small tissue areas
from multiple samples onto a single slide that is then
stained and processed as one composite sample.
Barcoding and pooling of multiple samples
If data are generated across long periods of time or on
multiple instruments, the use of standards can improve
sample comparability. Metal-containing beads can
normalize machine performance across time for mass
Antibody 1 Antibody 3 cytometry applications46, and repeat aliquots of stan­
dard sample(s) analysed with each run can be used in
Antibody 2 Antibody N
mass cytometry, as well as in high-dimensional imaging
Staining of combined sample technologies47. To improve reproducibility and acceler-
with heavy metal-tagged antibodies ate research efforts, raw data should be made available
in public repositories alongside published studies. Mass
cytometry raw data are often shared using platforms
Acquisition on mass cytometer such as Cytobank85, ImmPort114 and FlowRepository115.
The MIFlowCyt standard116, which provides crucial
information about experimental design, can be adopted
Data analysis for mass cytometry experiments, together enabling
re-analysis of the dataset, as well as large meta-analysis
Fig. 3 | Conducting large-scale immune-monitoring
approaches117. Analogous databases to those for mass
studies using mass cytometry. Patient cohorts are
selected depending on the experimental questions to be
cytometry data also exist for biomedical imaging data118.
addressed. Cells taken as samples at different time points
can be cryopreserved (as either live or fixed cells) and Immune monitoring in rheumatic diseases
collected prior to their analysis. To reduce technical Many rheumatic diseases share characteristics that
variation, single-cell suspensions from multiple donors can suggest that the immune system is important in their
be barcoded and pooled prior to staining and acquisition. aetiology. Firstly, genome-wide association studies have
In this process, a unique heavy metal tag (or a combination highlighted HLA alleles, which encode antigen presenta-
of several tags) is attached to all of the cells in a sample. tion machinery, as important genetic risk factors for the
These tagged samples can then be pooled and processed development of many rheumatic diseases119,120. Genes
(stained with heavy metal-tagged antibodies and acquired
related to other immune functions, such as cytokine
on a mass cytometer) as a single sample. Following
acquisition, cells can be assigned back to their original production121, are also associated with rheumatic dis-
sample and analysed further. eases. Genetic contributions to disease risk vary across
different rheumatic diseases, but in general, a combi-
nation of genetic predisposition and additional envi-
high-dimensional imaging are available24,110 and can be ronmental insults are thought to be triggering events in
performed in a few hours. rheumatic disease aetiology. For example, infection with
Antibodies specific for the chosen targets have to be Epstein–Barr virus and subsequent immune activation
distributed across the available mass reporter channels. are associated with an increased risk of developing many
Panel design considerations have been outlined else- rheumatic diseases119,122,123. Inflammation and other pro-
where111,112 but, briefly, minor sources of cross-channel cesses can in turn prompt epigenetic modifications120,
contamination can result from isotopic impurities as well as protein modifications such as citrullination121,
and the oxidation of elements (Box 1). Both phenom- which can potentially create novel autoantigens. In an
ena are often minor relative to technical variation and inflammatory microenvironment (as occurs in many

www.nature.com/nrrheum
Reviews

rheumatic diseases), antigen-presenting cells can display producing GM-CSF in these patients were CD4+ and
such modified antigens, thereby initiating an adaptive CD8+ T cells, γδ T cells and innate lymphoid cells.
immune response characterized by the interaction of Interestingly, GM-CSF expression by T cells is thought
T cells (particularly follicular helper T (TFH) cells) with to be an important mediator and potential therapeutic
B cells119,121,124. The production of autoantibodies by ter- target in other autoimmune diseases including multiple
minally differentiated plasma cells is a shared feature sclerosis130,131, suggesting that targeting GM-CSF might
of many rheumatic diseases that contributes to pathol­ also be an option in SpA. In addition to soluble medi­
ogy and is often used in their diagnosis119,121,122. Lastly, ators such as GM-CSF, the expression of surface epitopes
immune-targeted interventions (such as the modulation (such as programmed cell death protein 1 (PD1) on the
of TNF signalling or the depletion B cells) are therapeu- RA-associated cell population)127 could be targeted to
tic approaches in many rheumatic diseases125,126, again interfere with T cell–B cell interactions using existing
underlining the importance of the immune system in therapeutics.
these diseases.
Over the past few years, several studies have been Connective tissue diseases. In-depth functional and
published in which the authors have used mass cytom- pheno­typic analysis by mass cytometry has also been
etry to gain insights into the characteristics of various used to study the immune cell responses and cytokine
rheumatic diseases, some examples of which are dis- net­works that underlie connective tissue diseases such as
cussed in this section. Although clinical applicability has SLE and pSS. A comprehensive analysis of immune cell
been previously demonstrated in various contexts such subset responses to Toll-like receptor (TLR) ligands in
as cancer63 and autoimmunity61,62, we are not aware of cells from patients with SLE revealed a disease-associated
any published studies using MIBI or IMC in rheumatic immune signature that is triggered by TLR engagement132.
disease research. However, we expect important insights Specifically, CD14hi monocytes from patients with SLE
to be gained in the future by studying tissue-based had a pro-inflammatory chemokine and cytokine signa-
cellular interactions at sites of active disease. ture characterized by CC-chemokine ligand 2 (CCL2),
CCL4 and TNF. In a separate study, a similar immune
Inflammatory arthritis. Mass cytometry has been used signature, consisting of CCL2, CCL4 and IL-1 receptor
to study the activation and cytokine production profiles antagonist (IL-1RA), was found in monocytes from pae-
of T cells isolated from the peripheral blood, synovial diatric patients with SLE133. Interestingly, the presence of
fluid or inflamed tissues of patients with different types this immune signature correlated with clinical disease
of inflammatory arthritis, including RA and spondylo­ activity scores and could be abrogated through blockade
arthritis (SpA). The use of this high-dimensional of interferon α receptor (IFNAR) or Janus kinase (JAK)
approach enabled the identification of a pathologically inhibition133, thus providing mechanistic insights into the
expanded population of PD1hiCXCR5−CD4+ T cells in action of anifrolumab (an IFNAR inhibitor) and tofaci­
the synovium of patients with RA who were seroposi- tinib (a JAK inhibitor), emerging therapeutics for SLE
tive for rheumatoid factor or anti-citrullinated protein currently being evaluated in clinical trials134,135.
antibodies (ACPAs)127. In-depth phenotyping of these Focusing on B cell biology as a contributor to SLE, a
cells revealed the expression of several proteins involved mass cytometry study revealed dynamic changes in the
in B cell help, including IL-21, CXC-chemokine ligand frequency of several B cell subtypes during treatment
13 (CXCL13), inducible T cell co-stimulator (ICOS) and with belimumab136, a monoclonal antibody specific for
the transcription factor MAF, which enabled these cells B cell activating factor (BAFF) that has been approved
to be discriminated from traditional TFH cells, and mech- for the treatment of SLE. In this study, high-dimensional
anistic experiments demonstrated the ability of these phenotyping of B cells enabled the longitudinal moni-
cells to promote plasma cell differentiation. In a related toring of a broad range of B cell phenotypes, including
study from the same laboratories, peripheral blood from ABCs, a T-bet-dependent subset of B cells that display a
patients with RA and patients with osteoarthritis was distinct transcriptional profile compared with other B cell
analysed by mass cytometry128. Patients with RA had an subsets. ABCs increase in number with age, but also
expanded population of effector memory CD4+ T cells upon repeated viral infections and in patients with auto-
with a CD27−HLA-DR+ phenotype compared with immune diseases, including SLE137,138. A decrease in the
patients with osteoarthritis, suggesting chronic activa- number of ABCs (defined as CD11c+CD21− B cells) cor-
tion of these cells in RA. Together, these studies provide related with early clinical improvement during treatment
a mechanistic hypothesis for T cell-mediated patholog- with belimumab, and overall, pretreatment B cell counts
ical processes in RA in which tissue-infiltrating T cell were predictive of response to belimumab therapy136.
subsets might recruit additional immune cells via the Mass cytometry-based investigation of blood sam-
production of CXCL13 and IL-21. ples and salivary gland tissue from patients with pSS
The identification of such pathological processes can revealed a multi-population disease signature dominated
also provide potential targets for therapeutic interven- by activated CD8+ T cell populations and terminally dif-
tion. A mass cytometry study in SpA revealed increased ferentiated plasma B cells139. Importantly, this immuno­
numbers of a variety of granulocyte–macrophage logical signature could predict pSS diagnosis, as well
colony-stimulating factor (GM-CSF)-producing immune as stratify patients on the basis of clinical features and
cell subsets in the blood and joints of patients with disease activity, suggesting that these T cell and B cell
axial SpA compared with patients with RA and healthy populations might be potential targets for future clinical
individuals 129. Among the cell types identified as interventions.

Nature Reviews | Rheumatology


Reviews

Autoencoders
Insights across rheumatic diseases. These first mass For example, the use of autoencoders and related tools,
A type of artificial neural cytometry studies127–129,132,133,136,139 illustrate some of as already demonstrated on single-cell data141, could be
network that aims to learn the main advantages of this technique for in-depth directly applied to multiplexed images to enhance the
a lower-dimensional data phenotyping of immune cell subsets and how it can identification of relevant features142,143. Another excit-
representation from which
the original input can be
provide valuable insights into disease mechanisms in ing area of ongoing development for high-dimensional
reconstructed as closely rheumatic diseases. Multiple studies have provided evi- imaging is the transition from imaging a 2D plane
as possible. dence of chronic and/or increased T cell activation, such to imaging 3D structures, which would provide the
as the expansion of CD27− memory T cells in RA and opportunity to investigate spatial cellular information
multifaceted T cell activation in SpA and pSS128,129,139. in both health and disease. For example, the small
Unifying many of these findings across multiple auto- amount of tissue that is consumed with each MIBI scan
immune and rheumatic diseases, a 2019 study reported means that multiple scans of the same region could be
the use of mass cytometry and metal-conjugated MHC used to reconstruct 3D structures with highly resolved
class II tetramers to perform in-depth phenotyping on multiplexed infomation60,63,144,145.
antigen-specific autoimmune T cells from patients with The application of mass cytometry is ever-expanding
coeliac disease and other autoimmune and rheumatic into new research areas. For example, comprehensive
diseases140. The antigen-specific T cells in coeliac dis- immune phenotyping by mass cytometry has been used
ease140 had a phenotype that was strikingly similar to with as little as 100 µl of blood from newborn babies
that described in T cells in RA127, including the expres- to follow early immune development146. The authors
sion of multiple activation markers, PD1 and IL-21 and of this study performed additional analysis of plasma
the absence of CXC-chemokine receptor 5 (CXCR5). protein concentrations from these samples, highlighting
Interestingly, T cells with this phenotype were enriched the benefits of integrating multiple types of data. Such
in samples from individuals with many different auto­ multi-omics approaches combine data from transcrip-
immune diseases, including SSc and SLE140, thus posi- tomic, epigenetic, metabolomic and microbiome analy­
tioning these cells as important disease promoters in ses with clinical data, that together reveal otherwise
many autoimmune and rheumatic diseases. hidden correlations. Computational analysis of such
A unique advantage of defining high-dimensional integrated datasets remains challenging, but is an area
protein co-expression patterns via mass cytometry is of active research147–149. Using probes sensitive for chro-
the ability to precisely identify cellular signatures to bet- matin modification marks, researchers have also directly
ter understand the functional context of these cells and obtained single-cell epigenetic information using mass
to therapeutically target these specific cell populations. cytometry to investigate immunological changes that
In particular, several of the previously mentioned stud- occur with age150. Many autoimmune diseases, including
ies127,136,139 provide evidence that underlines the impor- rheumatic diseases such as SLE, RA and SSc, have sus-
tance of TFH cell–B cell interactions and plasma cell pected epigenetic contributions to their aetiology, and
differentiation, as well as defining the cell surface pheno- it would be of great interest to study epigenetic changes
types of the cell populations involved in these processes. at the single-cell level in combination with immune cell
Plasma cells ultimately contribute to autoantibody pro- phenotype(s) and cell population abundance120,151,152.
duction, an important pathogenic and diagnostic factor The ultimate goal of medical intervention in any
for many rheumatic diseases, and thus interfering with autoimmune disease is prevention of the disease before
plasma cell development might provide a therapeutic onset or, alternatively, early detection of damage to
strategy for these diseases. Another emerging theme from avoid disease exacerbation and the onset of comorbid-
these studies is altered cytokine expression profiles of ities153. In SLE, anti-nuclear antibodies can be detected
innate immune cells in SLE and of several populations up to 9 years before disease onset in some patients122,
of adaptive immune cells in RA and SpA, which point and similarly, the presence of ACPAs can precede the
towards the importance of immune communication onset of RA154. It would be of great interest to investi-
pathways that could be targeted therapeutically. gate if cellular signatures can be identified that predict
Together, these studies127–129,132,133,136,139 provide the imminent disease onset in individuals at risk of disease
first examples of how increased parametrization ena- development. A related research interest for rheumatic
bles the identification of complex cellular features that diseases is the identification of biomarkers that stratify
correlate with disease activity and clinical improvement, patients into clinically relevant groups. The early dis-
as well as the discovery of cellular signatures that could crimination of patients who are likely to respond well
be used diagnostically or that can serve as therapeutic to a given therapeutic from patients who are unlikely to
targets across a range of rheumatic diseases. benefit could contribute to the development of person-
alized therapeutic schemes. For example, a 2019 study
Future directions used mass cytometry to detect a T cell subset that could
The continued analytical and methodological develop- discriminate patients with juvenile idiopathic arthri-
ments in both mass cytometry and high-dimensional tis who were at risk of relapse upon anti-TNF therapy
imaging technologies make them core technologies for withdrawal155. Importantly, such in-depth monitoring
use in many immune-monitoring and related studies. The of patients receiving various therapies could addition-
ability to extract and recognize biologically informative ally pinpoint the mechanism of action of currently
but complex information from these high-dimensional approved drugs and thus open opportunities to refine
datasets will be further improved through developments their efficacy by directly modulating a specific cellular
in the fields of artificial intelligence and machine learning. function instead of broadly targeting complete immune

www.nature.com/nrrheum
Reviews

cell lineages. Furthermore, although many autoimmune offered by mass cytometry and high-dimensional
and rheumatic diseases are chronic, patients will often imaging technologies, in combination with innova-
experience short-term disease flares. The analysis and tive analytical approaches, provides an opportunity
comparison of affected tissues at different time points to elucidate these mechanisms at a high-dimensional
before, during and after disease flares could provide level. As illustrated in several studies over the past few
insight into the reservoir of pathological cells in these years, these technologies have enabled the discovery
patients and enable the identification of biomarkers that of disease-associated cellular signatures that provide
signify the timing of such flares. Novel microneedle sam- insight into pathology and ideas for future treatments
pling devices could facilitate such longitudinal studies across many diseases. Technologies such as mass cytom-
by potentially enabling at-home blood collections156,157, etry, IMC and MIBI offer the field of rheumatic disease
whereas skin or synovial tissue samples acquired using research an opportunity to comprehensively phenotype
sampling techniques such as fine-needle aspirates158 immune cells in a variety of diseases and conditions
or microneedle patches159 could be imaged directly or (including during and after flares or during treatment
analysed as a single-cell suspension by mass cytometry. with different therapies), enabling researchers to reveal
immune signatures for stratifying patients, to uncover
Conclusions novel therapeutic targets and to identify biomarkers for
A heterogeneous clinical presentation and complex therapeutic success.
pathobiology are characteristic of many rheumatic dis-
Published online xx xx xxxx
eases. The single-cell systems immunology perspective

1. Robinson, W. H. & Mao, R. Technological advances 19. Ornatsky, O. I. et al. Study of cell antigens and 35. Galli, E. et al. GM-CSF and CXCR4 define a T helper
transforming rheumatology. Nat. Rev. Rheumatol. 11, intracellular DNA by identification of element- cell signature in multiple sclerosis. Nat. Med. 25,
626–628 (2015). containing labels and metallointercalators using 1290–1300 (2019).
2. Hartmann, F. J. et al. Comprehensive immune inductively coupled plasma mass spectrometry. 36. Bodenmiller, B. et al. Multiplexed mass cytometry
monitoring of clinical trials to advance human Anal. Chem. 80, 2539–2547 (2008). profiling of cellular states perturbed by small-molecule
immunotherapy. Cell Rep. 28, 819–831.e4 (2019). 20. Ornatsky, O. I. et al. Development of analytical regulators. Nat. Biotechnol. 30, 858–867 (2012).
3. von Herrath, M. G. & Nepom, G. T. Lost in translation: methods for multiplex bio-assay with inductively 37. Fienberg, H. G., Simonds, E. F., Fantl, W. J., Nolan, G. P.
barriers to implementing clinical immunotherapeutics coupled plasma mass spectrometry. J. Anal. At. & Bodenmiller, B. A platinum-based covalent viability
for autoimmunity. J. Exp. Med. 202, 1159–1162 Spectrom. 23, 463 (2008). reagent for single-cell mass cytometry. Cytometry A
(2005). 21. Majonis, D. et al. Synthesis of a functional metal- 81A, 467–475 (2012).
4. Davis, M. M. A prescription for human immunology. chelating polymer and steps toward quantitative mass 38. Hartmann, F. J., Simonds, E. F. & Bendall, S. C.
Immunity 29, 835–838 (2008). cytometry bioassays. Anal. Chem. 82, 8961–8969 A universal live cell barcoding-platform for multiplexed
5. Davis, M. M. & Brodin, P. Rebooting human (2010). human single cell analysis. Sci. Rep. 8, 10770 (2018).
immunology. Annu. Rev. Immunol. 36, 843–864 22. Mei, H. E., Leipold, M. D. & Maecker, H. T. 39. Frei, A. P. et al. Highly multiplexed simultaneous
(2018). Platinum-conjugated antibodies for application detection of RNAs and proteins in single cells.
6. Robinson, W. H. & Mao, R. Biomarkers to guide in mass cytometry. Cytometry A 89, 292–300 Nat. Methods 13, 269–275 (2016).
clinical therapeutics in rheumatology? Curr. Opin. (2016). 40. Duckworth, A. D. et al. Multiplexed profiling of
Rheumatol. 28, 168–175 (2016). 23. Han, G. et al. Atomic mass tag of bismuth-209 for RNA and protein expression signatures in individual
7. Ermann, J., Rao, D. A., Teslovich, N. C., Brenner, M. B. increasing the immunoassay multiplexing capacity cells using flow or mass cytometry. Nat. Protoc. 14,
& Raychaudhuri, S. Immune cell profiling to guide of mass cytometry. Cytometry A 91, 1150–1163 901–920 (2019).
therapeutic decisions in rheumatic diseases. Nat. Rev. (2017). 41. Behbehani, G. K., Bendall, S. C., Clutter, M. R.,
Rheumatol. 11, 541–551 (2015). 24. Han, G., Spitzer, M. H., Bendall, S. C., Fantl, W. J. Fantl, W. J. & Nolan, G. P. Single-cell mass cytometry
8. Gaudillière, B. et al. Clinical recovery from surgery & Nolan, G. P. Metal-isotope-tagged monoclonal adapted to measurements of the cell cycle. Cytometry A
correlates with single-cell immune signatures. antibodies for high-dimensional mass cytometry. 81A, 552–566 (2012).
Sci. Transl Med. 6, 255ra131 (2014). Nat. Protoc. 13, 2121–2148 (2018). 42. Kimmey, S. C., Borges, L., Baskar, R. & Bendall, S. C.
9. Levine, J. H. et al. Data-driven phenotypic dissection 25. Wagner, J. et al. A single-cell atlas of the tumor and Parallel analysis of tri-molecular biosynthesis with cell
of AML reveals progenitor-like cells that correlate with immune ecosystem of human breast cancer. Cell 177, identity and function in single cells. Nat. Commun. 10,
prognosis. Cell 162, 184–197 (2015). 1330–1345.e18 (2019). 1185 (2019).
10. Good, Z. et al. Single-cell developmental classification 26. Chang, Q. et al. Single-cell measurement of the 43. Poreba, M. et al. The Activome: multiplexed probing
of B cell precursor acute lymphoblastic leukemia at uptake, intratumoral distribution and cell cycle effects of activity of proteolytic enzymes using mass cytometry-
diagnosis reveals predictors of relapse. Nat. Med. 24, of cisplatin using mass cytometry. Int. J. Cancer 136, compatible activity-based probes (TOF-probes).
474–483 (2018). 1202–1209 (2015). Preprint at bioRxiv https://doi.org/10.1101/775627
11. Nair, N. et al. Mass cytometry as a platform for the 27. Chang, Q. et al. Biodistribution of cisplatin revealed by (2019).
discovery of cellular biomarkers to guide effective imaging mass cytometry identifies extensive collagen 44. Edgar, L. J. et al. Identification of hypoxic cells using an
rheumatic disease therapy. Arthritis Res. Ther. 17, binding in tumor and normal tissues. Sci. Rep. 6, organotellurium tag compatible with mass cytometry.
127 (2015). 36641 (2016). Angew. Chem. Int. Ed. 53, 11473–11477 (2014).
12. Maecker, H. T., McCoy, J. P. & Nussenblatt, R. 28. Newell, E. W., Sigal, N., Bendall, S. C., Nolan, G. P. & 45. Schulz, D., Severin, Y., Zanotelli, V. R. T. &
Standardizing immunophenotyping for the Human Davis, M. M. Cytometry by time-of-flight shows Bodenmiller, B. In-depth characterization of
Immunology Project. Nat. Rev. Immunol. 12, combinatorial cytokine expression and virus-specific monocyte-derived macrophages using a mass
191–200 (2012). cell niches within a continuum of CD8+ T cell cytometry-based phagocytosis assay. Sci. Rep. 9,
13. Hao, Y., O’Neill, P., Naradikian, M. S., Scholz, J. L. & phenotypes. Immunity 36, 142–152 (2012). 1925 (2019).
Cancro, M. P. A B-cell subset uniquely responsive to 29. Newell, E. W. et al. Combinatorial tetramer staining 46. Finck, R. et al. Normalization of mass cytometry
innate stimuli accumulates in aged mice. Blood 118, and mass cytometry analysis facilitate T-cell epitope data with bead standards. Cytometry A 83, 483–494
1294–1304 (2011). mapping and characterization. Nat. Biotechnol. 31, (2013).
14. Chattopadhyay, P. K. & Roederer, M. Good cell, bad 623–629 (2013). 47. Kleinsteuber, K. et al. Standardization and quality
cell: flow cytometry reveals T-cell subsets important 30. Newell, E. W. & Davis, M. M. Beyond model antigens: control for high-dimensional mass cytometry studies
in HIV disease. Cytometry A 77, 614–622 (2010). high-dimensional methods for the analysis of of human samples. Cytometry A 89, 903–913 (2016).
15. Bandura, D. R. et al. Mass cytometry: technique for antigen-specific T cells. Nat. Biotechnol. 32, 149–157 48. Leipold, M. D. et al. Comparison of CyTOF assays
real time single cell multitarget immunoassay based (2014). across sites: results of a six-center pilot study.
on inductively coupled plasma time-of-flight mass 31. Leong, M. L. & Newell, E. W. in Single Cell Protein J. Immunol. Methods 453, 37–43 (2018).
spectrometry. Anal. Chem. 81, 6813–6822 (2009). Analysis (eds. Singh, A. K. & Chandrasekaran, A.) 49. Zunder, E. R. et al. Palladium-based mass tag cell
16. Ornatsky, O. et al. Highly multiparametric analysis by 115–131 (Humana, 2015). barcoding with a doublet-filtering scheme and
mass cytometry. J. Immunol. Methods 361, 1–20 32. Simoni, Y. et al. Bystander CD8+ T cells are abundant single-cell deconvolution algorithm. Nat. Protoc. 10,
(2010). and phenotypically distinct in human tumour 316–333 (2015).
17. Bendall, S. C. et al. Single-cell mass cytometry of infiltrates. Nature 557, 575–579 (2018). 50. Mei, H. E., Leipold, M. D., Schulz, A. R., Chester, C.
differential immune and drug responses across a 33. Spitzer, M. H. et al. An interactive reference framework & Maecker, H. T. Barcoding of live human peripheral
human hematopoietic continuum. Science 332, for modeling a dynamic immune system. Science 349, blood mononuclear cells for multiplexed mass
687–696 (2011). 1259425 (2015). cytometry. J. Immunol. 194, 2022–2031 (2015).
18. Lou, X. et al. Polymer-based elemental tags for 34. Hartmann, F. J. et al. High-dimensional single-cell 51. Lai, L., Ong, R., Li, J. & Albani, S. A CD45-based
sensitive bioassays. Angew. Chem. Int. Ed. 46, analysis reveals the immune signature of narcolepsy. barcoding approach to multiplex mass-cytometry
6111–6114 (2007). J. Exp. Med. 213, 2621–2633 (2016). (CyTOF). Cytometry A 87, 369–374 (2015).

Nature Reviews | Rheumatology


Reviews

52. Bodenmiller, B. Multiplexed epitope-based tissue 79. Gabriel, K. R. & Sokal, R. R. A new statistical approach 106. Strauss-Albee, D. M. et al. Human NK cell repertoire
imaging for discovery and healthcare applications. to geographic variation analysis. Syst. Zool. 18, diversity reflects immune experience and correlates
Cell Syst. 2, 225–238 (2016). 259–278 (1969). with viral susceptibility. Sci. Transl Med. 7, 297ra115
53. Gerner, M. Y., Kastenmuller, W., Ifrim, I., Kabat, J. & 80. Finak, G., Perez, J.-M., Weng, A. & Gottardo, R. (2015).
Germain, R. N. Histo-cytometry: a method for highly Optimizing transformations for automated, high 107. Good, Z. et al. Proliferation tracing with single-cell
multiplex quantitative tissue imaging analysis applied throughput analysis of flow cytometry data. mass cytometry optimizes generation of stem cell
to dendritic cell subset microanatomy in lymph nodes. BMC Bioinformatics 11, 546 (2010). memory-like T cells. Nat. Biotechnol. 37, 259–266
Immunity 37, 364–376 (2012). 81. Bagwell, C. B. Hyperlog?A flexible log-like transform (2019).
54. Gerdes, M. J. et al. Highly multiplexed single-cell for negative, zero, and positive valued data. 108. Aghaeepour, N. et al. GateFinder: projection-based
analysis of formalin-fixed, paraffin-embedded cancer Cytometry A 64A, 34–42 (2005). gating strategy optimization for flow and mass
tissue. Proc. Natl Acad. Sci. USA 110, 11982–11987 82. Parks, D. R., Roederer, M. & Moore, W. A. A new cytometry. Bioinformatics 34, 4131–4133
(2013). ‘Logicle’ display method avoids deceptive effects of (2018).
55. Lin, J.-R. et al. Highly multiplexed immunofluorescence logarithmic scaling for low signals and compensated 109. Spitzer, M. H. & Nolan, G. P. Mass cytometry:
imaging of human tissues and tumors using t-CyCIF data. Cytometry A 69, 541–551 (2006). single cells, many features. Cell 165, 780–791
and conventional optical microscopes. eLife 7, e31657 83. Maaten, L. Van Der & Hinton, G. Visualizing data using (2016).
(2018). t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008). 110. Hartmann, F. J. et al. Scalable conjugation and
56. Goltsev, Y. et al. Deep profiling of mouse splenic 84. Amir, E. D. et al. viSNE enables visualization of high characterization of immunoglobulins with stable
architecture with CODEX multiplexed imaging. Cell dimensional single-cell data and reveals phenotypic mass isotope reporters for single-cell mass cytometry
174, 968–981.e15 (2018). heterogeneity of leukemia. Nat. Biotechnol. 31, analysis. Methods Mol. Biol. 1989, 55–81 (2019).
57. Davis, A. S. et al. Characterizing and diminishing 545–552 (2013). 111. Leipold, M. D., Newell, E. W. & Maecker, H. T.
autofluorescence in formalin-fixed paraffin-embedded 85. Kotecha, N., Krutzik, P. O. & Irish, J. M. Web-based Multiparameter phenotyping of human PBMCs using
human respiratory tissue. J. Histochem. Cytochem. analysis and publication of flow cytometry experiments. mass cytometry. Methods Mol. Biol. 1343, 81–95
62, 405–423 (2014). Curr. Protoc. Cytom. 53, 10.17.1–10.17.24 (2010). (2015).
58. Giesen, C. et al. Highly multiplexed imaging of tumor 86. van der Maaten, L. Barnes-Hut-SNE. Preprint at arXiv 112. Takahashi, C. et al. Mass cytometry panel optimization
tissues with subcellular resolution by mass cytometry. https://arxiv.org/abs/1301.3342v2 (2013). through the designed distribution of signal interference.
Nat. Methods 11, 417–422 (2014). 87. Pezzotti, N. et al. Approximated and user steerable Cytometry A 91, 39–47 (2017).
59. Angelo, M. et al. Multiplexed ion beam imaging tSNE for progressive visual analytics. IEEE Trans. Vis. 113. Chevrier, S. et al. Compensation of signal spillover in
of human breast tumors. Nat. Med. 20, 436–442 Computer Graph. 23, 1739–1752 (2016). suspension and imaging mass cytometry. Cell Syst. 6,
(2014). 88. van Unen, V. et al. Visual analysis of mass cytometry 612–620.e5 (2018).
60. Keren, L. et al. MIBI-TOF: A multiplexed imaging data by hierarchical stochastic neighbour embedding 114. Bhattacharya, S. et al. ImmPort: disseminating data to
platform relates cellular phenotypes and tissue reveals rare cell types. Nat. Commun. 8, 1740 the public for the future of immunology. Immunol. Res.
structure. Sci. Adv. 5, eaax5851 (2019). (2017). 58, 234–239 (2014).
61. Damond, N. et al. A map of human type 1 diabetes 89. Cho, H., Berger, B. & Peng, J. Generalizable and 115. Spidlen, J., Breuer, K., Rosenberg, C., Kotecha, N.
progression by imaging mass cytometry. Cell Metab. scalable visualization of single-cell data using neural & Brinkman, R. R. FlowRepository: a resource of
29, 755–768.e5 (2019). networks. Cell Syst. 7, 185–191.e4 (2018). annotated flow cytometry datasets associated with
62. Wang, Y. J. et al. Multiplexed in situ imaging mass 90. McInnes, L. & Healy, J. UMAP: uniform manifold peer-reviewed publications. Cytometry A 81A,
cytometry analysis of the human endocrine pancreas approximation and projection for dimension reduction. 727–731 (2012).
and immune system in type 1 diabetes. Cell Metab. Preprint at arXiv https://arxiv.org/abs/1802.03426v2 116. Lee, J. A. et al. MIFlowCyt: the minimum information
29, 769–783.e4 (2019). (2018). about a flow cytometry experiment. Cytometry A 73A,
63. Keren, L. et al. A structured tumor-immune 91. Becht, E. et al. Dimensionality reduction for visualizing 926–930 (2008).
microenvironment in triple negative breast cancer single-cell data using UMAP. Nat. Biotechnol. 37, 117. Hu, Z. et al. MetaCyto: a tool for automated
revealed by multiplexed ion beam imaging. Cell 174, 38–44 (2019). meta-analysis of mass and flow cytometry data.
1373–1387.e19 (2018). 92. Qiu, P. et al. Extracting a cellular hierarchy from Cell Rep. 24, 1377–1388 (2018).
64. Schulz, D. et al. Simultaneous multiplexed imaging high-dimensional cytometry data with SPADE. 118. Williams, E. et al. Image Data Resource: a bioimage
of mRNA and proteins with subcellular resolution in Nat. Biotechnol. 29, 886–891 (2011). data integration and publication platform. Nat. Methods
breast cancer tissue samples by mass cytometry. 93. Samusik, N., Good, Z., Spitzer, M. H., Davis, K. L. & 14, 775–781 (2017).
Cell Syst. 6, 25–36.e5 (2018). Nolan, G. P. Automated mapping of phenotype space 119. Brito-Zerón, P. et al. Sjögren syndrome. Nat. Rev.
65. Catena, R., Montuenga, L. M. & Bodenmiller, B. with single-cell data. Nat. Methods 13, 493–496 Dis. Primers 2, 16047 (2016).
Ruthenium counterstaining for imaging mass (2016). 120. Angiolilli, C. et al. New insights into the genetics and
cytometry. J. Pathol. 244, 479–484 (2018). 94. Zunder, E. R., Lujan, E., Goltsev, Y., Wernig, M. & epigenetics of systemic sclerosis. Nat. Rev. Rheumatol.
66. Skinner, P. J., Daniels, M. A., Schmidt, C. S., Nolan, G. P. A continuous molecular roadmap to 14, 657–673 (2018).
Jameson, S. C. & Haase, A. T. Cutting edge: in situ iPSC reprogramming through progression analysis 121. Smolen, J. S. et al. Rheumatoid arthritis. Nat. Rev.
tetramer staining of antigen-specific T cells in tissues. of single-cell mass cytometry. Cell Stem Cell 16, Dis. Primers 4, 18001 (2018).
J. Immunol. 165, 613–617 (2000). 323–337 (2015). 122. Kaul, A. et al. Systemic lupus erythematosus. Nat. Rev.
67. Steinert, E. M. et al. Quantifying memory CD8 T cells 95. Mrdjen, D. et al. High-dimensional single-cell mapping Dis. Primers 2, 16039 (2016).
reveals regionalization of immunosurveillance. Cell of central nervous system immune cells reveals 123. Tsokos, G. C., Lo, M. S., Reis, P. C. & Sullivan, K. E.
161, 737–749 (2015). distinct myeloid subsets in health, aging, and disease. New insights into the immunopathogenesis of systemic
68. Li, S. et al. Simian immunodeficiency virus-producing Immunity 48, 380–395.e6 (2018). lupus erythematosus. Nat. Rev. Rheumatol. 12,
cells in follicles are partially suppressed by CD8+ cells 96. Weber, L. M. & Robinson, M. D. Comparison of 716–730 (2016).
in vivo. J. Virol. 90, 11168–11180 (2016). clustering methods for high-dimensional single-cell 124. Mavragani, C. P. & Moutsopoulos, H. M. Sjögren’s
69. Mair, F. et al. The end of gating? An introduction to flow and mass cytometry data. Cytometry A 89, syndrome. Annu. Rev. Pathol. Mech. Dis. 9, 273–285
automated analysis of high dimensional cytometry 1084–1096 (2016). (2014).
data. Eur. J. Immunol. 46, 34–43 (2016). 97. Van Gassen, S. et al. FlowSOM: using self-organizing 125. Sedger, L. M. & McDermott, M. F. TNF and
70. Saeys, Y., Gassen, S. Van & Lambrecht, B. N. maps for visualization and interpretation of cytometry TNF-receptors: from mediators of cell death
Computational flow cytometry: helping to make data. Cytometry A 87, 636–645 (2015). and inflammation to therapeutic giants – past,
sense of high-dimensional immunology data. Nat. Rev. 98. Bruggner, R. V., Bodenmiller, B., Dill, D. L., present and future. Cytokine Growth Factor. Rev. 25,
Immunol. 16, 449–462 (2016). Tibshirani, R. J. & Nolan, G. P. Automated 453–472 (2014).
71. Chester, C. & Maecker, H. T. Algorithmic tools for identification of stratifying signatures in cellular 126. Hofmann, K., Clauder, A.-K. & Manz, R. A. Targeting
mining high-dimensional cytometry data. J. Immunol. subpopulations. Proc. Natl Acad. Sci. USA 111, B cells and plasma cells in autoimmune diseases.
195, 773–779 (2015). E2770–E2777 (2014). Front. Immunol. 9, 835 (2018).
72. Newell, E. W. & Cheng, Y. Mass cytometry: blessed 99. Spitzer, M. H. et al. Systemic immunity is required for 127. Rao, D. A. et al. Pathologically expanded peripheral
with the curse of dimensionality. Nat. Immunol. 17, effective cancer immunotherapy. Cell 168, 487–502. T helper cell subset drives B cells in rheumatoid
890–895 (2016). e15 (2017). arthritis. Nature 542, 110–114 (2017).
73. Moen, E. et al. Deep learning for cellular image 100. Lun, A. T. L., Richard, A. C. & Marioni, J. C. Testing 128. Fonseka, C. Y. et al. Mixed-effects association of single
analysis. Nat. Methods 16, 1233–1246 (2019). for differential abundance in mass cytometry data. cells identifies an expanded effector CD4+ T cell
74. Van Valen, D. A. et al. Deep learning automates the Nat. Methods 14, 707–709 (2017). subset in rheumatoid arthritis. Sci. Transl Med. 10,
quantitative analysis of individual cells in live-cell 101. Arvaniti, E. & Claassen, M. Sensitive detection of rare eaaq0305 (2018).
imaging experiments. PLOS Comput. Biol. 12, disease-associated cell subsets via representation 129. Al-Mossawi, M. H. et al. Unique transcriptome
e1005177 (2016). learning. Nat. Commun. 8, 14825 (2017). signatures and GM-CSF expression in lymphocytes
75. Sommer, C., Straehle, C., Kothe, U. & Hamprecht, F. A. 102. Bendall, S. C. et al. Single-cell trajectory detection from patients with spondyloarthritis. Nat. Commun. 8,
in 2011 IEEE International Symposium on Biomedical uncovers progression and regulatory coordination 1510 (2017).
Imaging: From Nano to Macro 230–233 (IEEE, 2011). in human B cell development. Cell 157, 714–725 130. Noster, R. et al. IL-17 and GM-CSF expression are
76. Dao, D. et al. CellProfiler Analyst: interactive data (2014). antagonistically regulated by human T helper cells.
exploration, analysis and classification of large 103. Setty, M. et al. Wishbone identifies bifurcating Sci. Transl Med. 6, 241ra80 (2014).
biological image sets. Bioinformatics 32, 3210–3212 developmental trajectories from single-cell data. 131. Hartmann, F. J. et al. Multiple sclerosis-associated
(2016). Nat. Biotechnol. 34, 637–645 (2016). IL2RA polymorphism controls GM-CSF production in
77. Schapiro, D. et al. histoCAT: analysis of cell phenotypes 104. Saelens, W., Cannoodt, R., Todorov, H. & Saeys, Y. human TH cells. Nat. Commun. 5, 5056 (2014).
and interactions in multiplex image cytometry data. A comparison of single-cell trajectory inference 132. O’Gorman, W. E. et al. Single-cell systems-level
Nat. Methods 14, 873–876 (2017). methods. Nat. Biotechnol. 37, 547–554 (2019). analysis of human Toll-like receptor activation defines
78. Haberl, M. G. et al. CDeep3M–Plug-and-play 105. Krishnaswamy, S. et al. Systems biology. Conditional a chemokine signature in patients with systemic
cloud-based deep learning for image segmentation. density-based analysis of T cell signaling in single-cell lupus erythematosus. J. Allergy Clin. Immunol. 136,
Nat. Methods 15, 677–680 (2018). data. Science 346, 1250689 (2014). 1326–1336 (2015).

www.nature.com/nrrheum
Reviews

133. O’Gorman, W. E. et al. Mass cytometry identifies 147. Pedersen, H. K. et al. A computational framework to 159. Mandal, A. et al. Cell and fluid sampling
a distinct monocyte cytokine signature shared by integrate high-throughput ‘-omics’ datasets for the microneedle patches for monitoring skin-resident
clinically heterogeneous pediatric SLE patients. identification of potential mechanistic links. immunity. Sci. Transl Med. 10, eaar2227
J. Autoimmun. 81, 74–89 (2017). Nat. Protoc. 13, 2781–2800 (2018). (2018).
134. US National Library of Medicine. ClinicalTrials.gov 148. Ghaemi, M. S. et al. Multiomics modeling of the 160. Perfetto, S. P., Chattopadhyay, P. K. & Roederer, M.
https://clinicaltrials.gov/ct2/show/NCT02535689 immunome, transcriptome, microbiome, proteome and Seventeen-colour flow cytometry: unravelling the
(2018). metabolome adaptations during human pregnancy. immune system. Nat. Rev. Immunol. 4, 648–655
135. US National Library of Medicine. ClinicalTrials.gov Bioinformatics 35, 95–103 (2018). (2004).
https://clinicaltrials.gov/ct2/show/NCT02446899 149. Huang, S., Chaudhary, K. & Garmire, L. X. More is 161. Cossarizza, A. et al. Guidelines for the use of flow
(2019). better: recent progress in multi-omics data integration cytometry and cell sorting in immunological studies
136. Ramsköld, D. et al. B cell alterations during BAFF methods. Front. Genet. 8, 84 (2017). (second edition). Eur. J. Immunol. 49, 1457–1973
inhibition with belimumab in SLE. EBioMedicine 40, 150. Cheung, P. et al. Single-cell chromatin modification (2019).
517–527 (2019). profiling reveals increased epigenetic variations with 162. Bendall, S. C., Nolan, G. P., Roederer, M. &
137. Rubtsova, K., Rubtsov, A. V., Cancro, M. P. & aging. Cell 173, 1385–1397.e14 (2018). Chattopadhyay, P. K. A deep profiler’s guide to
Marrack, P. Age-associated B cells: a T-bet-dependent 151. Zhang, Z. & Zhang, R. Epigenetics in autoimmune cytometry. Trends Immunol. 33, 323–332 (2012).
effector with roles in protective and pathogenic diseases: pathogenesis and prospects for therapy.
immunity. J. Immunol. 195, 1933–1937 (2015). Autoimmun. Rev. 14, 854–863 (2015). Acknowledgements
138. Wang, S. et al. IL-21 drives expansion and plasma cell 152. Jeffries, M. A. & Sawalha, A. H. Autoimmune disease The work of F.J.H. is supported by the EMBO organization
differentiation of autoreactive CD11chiT-bet+ B cells in in the epigenetic era: how has epigenetics changed our (EMBO Long-Term Fellowship ALTF 1141-2017), the Novartis
SLE. Nat. Commun. 9, 1758 (2018). understanding of disease and how can we expect the Foundation for Medical-Biological Research (16C148) and
139. Mingueneau, M. et al. Cytometry by time-of-flight field to evolve? Expert. Rev. Clin. Immunol. 11, 45–58 the Swiss National Science Foundation (SNF Early Postdoc
immunophenotyping identifies a blood Sjögren’s (2015). Mobility P2ZHP3-171741). The work of S.C.B. is suppor­
signature correlating with disease activity and 153. Munroe, M. E. et al. Discerning risk of disease ted by the Damon Runyon Cancer Research Foundation
glandular inflammation. J. Allergy Clin. Immunol. 137, transition in relatives of systemic lupus erythematosus Fellowship (DRG-2017-09), the NIH (1DP2OD022550-01,
1809–1821.e12 (2016). patients utilizing soluble mediators and clinical 1 R01 AG 0 5 6 2 8 7 -01 , 1 R01 AG 0 5 7 91 5- 01 , 1 - R 00 -
140. Christophersen, A. et al. Distinct phenotype of features. Arthritis Rheumatol. 69, 630–642 GM104148-01, 1U24CA224309-01, 5U19AI116484-02
CD4+ T cells driving celiac disease identified in (2017). and U19 AI104209) and a Translational Research Award
multiple autoimmune conditions. Nat. Med. 25, 154. van der Woude, D. et al. Epitope spreading of the from the Stanford Cancer Institute.
734–737 (2019). anti-citrullinated protein antibody response occurs
141. Lopez, R., Regier, J., Cole, M. B., Jordan, M. I. & before disease onset and is associated with the Author contributions
Yosef, N. Deep generative modeling for single-cell disease course of early arthritis. Ann. Rheum. Dis. 69, The authors contributed equally to all aspects of the article.
transcriptomics. Nat. Methods 15, 1053–1058 1554–1561 (2010).
(2018). 155. Leong, J. Y. et al. Immunome perturbation is Competing interests
142. Falk, T. et al. U-Net: deep learning for cell counting, present in patients with juvenile idiopathic arthritis S.C.B. declares that he is an inventor of multiplexed ion beam
detection, and morphometry. Nat. Methods 16, who are in remission and will relapse upon anti-TNFα imaging technology and a scientific founder of IONpath Inc.,
67–70 (2019). withdrawal. Ann. Rheum. Dis. 78, 1712–1721 the company that commercialized this technology. F.J.H.
143. Gupta, A. et al. Deep learning in image cytometry: (2019). declares no competing interests.
a review. Cytometry A 95, 366–380 (2018). 156. Blicharz, T. M. et al. Microneedle-based device for the
144. Rovira-Clave, X. et al. Subcellular localization of drug one-step painless collection of capillary blood samples. Peer review information
distribution by super-resolution ion beam imaging. Nat. Biomed. Eng. 2, 151–157 (2018). Nature Reviews Rheumatology thanks J. Lederer, P. Brodin
Preprint at bioRxiv https://doi.org/10.1101/557603 157. Josyula, V. S., Lakshmikanth, T., Mikes, J., Chen, Y. & and the other, anonymous, reviewer(s) for their contribution
(2019). Brodin, P. Systems-level immunomonitoring using to the peer review of this work.
145. Coskun, A. F. et al. Ion beam subcellular tomography. self-sampled capillary blood. Preprint at bioRxiv
Preprint at bioRxiv https://doi.org/10.1101/557728 https://doi.org/10.1101/694521 (2019). Publisher’s note
(2019). 158. Tatovic, D. et al. Fine-needle aspiration biopsy of the Springer Nature remains neutral with regard to jurisdictional
146. Olin, A. et al. Stereotypic immune system development lymph node: a novel tool for the monitoring of immune claims in published maps and institutional affiliations.
in newborn children. Cell 174, 1277–1292.e14 responses after skin antigen delivery. J. Immunol.
(2018). 195, 386–392 (2015). © Springer Nature Limited 2019

Nature Reviews | Rheumatology

You might also like