You are on page 1of 178

pub_NanoSIMS_cell-press2_Cell press selection 03/11/2020 14:24 Page 4

Upgrade your knowledge on


microanalytical techniques!

The Essential Guide on


Dynamic Secondary Ion Mass
Spectrometry (SIMS)
Still the most sensitive microanalytical technique available, Dynamic
SIMS provides localized elemental, isotopic and molecular characterization
of the sample surface.
Our free Dynamic SIMS guide provides a simple introduction to the basic principles of this essential
microanalytical technique, outlining its advantages over alternative surface analysis techniques. Four
case studies allow readers to explore current practices and applications of the NanoSIMS and other
dynamic SIMS instruments including research conducted at Harvard Medical School (see extract below).

Scan the code or visit


cameca.com/focus/sims-tuto
to download a free guide
on Dynamic SIMS!

Extract from Dynamic SIMS guide, case study Nr 4:


Protein turnover quantification by feeding animals with a 15N-labeled
precursor amino acid to measure appearance of new protein.
In contrast to previous studies, direct measurement with NanoSIMS
multi-isotope imaging mass spectrometry revealed unexpectedly low
turnover of protein in hair-cell stereocili. Measured volume: 8x8x2μm.

From: Multi-isotope imaging mass spectrometry reveals slow


protein turnover in hair-cell stereocilia. Duan-Sun Zhang et al.
NATURE, vol 481, 26 January 2012

cameca.info@ametek.com
www.cameca.com
pub_NanoSIMS_cell-press2_Cell press selection 03/11/2020 14:24 Page 1

NanoSIMS 50L
Quantitative measurement
of metabolic activity and
cellular exchanges with
sub-cellular resolution
The CAMECA NanoSIMS 50L is a unique imaging Secondary Ion Mass Spectrometer enabling
quantitative measurements as well as 2D and 3D imaging of molecular distribution, kinetics and fluxes
in tissues or at intra-cellular level through a strategy of stable isotope or rare element labelling.
The NanoSIMS 50L delivers simultaneously:
• 2D & 3D imaging of molecular incorporation with 50nm resolution;
• Mapping of stable isotopes and/or trace elements tagged molecules;
• Up to 7 isotopes or masses recorded in parallel at high mass resolution.

Metabolic origin of glial lipid droplets revealed with NanoSIMS multi-isotope imaging mass spectrometry.

fold change
12
C / 13C 12
C / 13C 12
C / 13C above natural
ratio

Drosophilia larvae were raised on diets containing 13C-labeled glucose, acetate or linoleate, major carbon
sources for lipogenesis. Glial lipid droplets were then induced with 22 hr hypoxia.
NanoSIMS analyses reveal strong 13C incorporation of labeled fatty acids into the core of hypoxic lipid droplets.
De novo fatty acid synthesis contributes neutral lipid cargo to glial lipid droplets.
From: Antioxidant Role for Lipid Droplets in a Stem Cell Niche of Drosophila. Andrew P. Bailey et al.
CELL 163, 340–353, 8 October 2015

cameca.info@ametek.com
www.cameca.com
Foreword
The ability of cells to undergo metabolic reprogramming in response to their changing environment is crucial to their
function, and disruption of these complex responses underlies a growing number of metabolic diseases. However,
until recently, research has relied on bulk measurements to understand a cell’s underlying metabolic status and its
response to and interaction with its environment.

With the emergence of ever-more-sophisticated single-cell techniques, we are now beginning to get a glimpse of
the metabolic status of individual cells within a heterogeneous population and to understand the dynamics of these
cell populations in space and time. These technologies have painted a broader picture of how cells interact within
tissues and with their environment during health and disease.

In this Cell Press Selection, Metabolism at the Single-Cell Level, we showcase diverse cutting-edge techniques,
including live-cell imaging, chemical imaging, and single-cell transcriptomic technologies that have been harnessed
for understanding metabolism at the single-cell and even sub-cellular level. These technologies have been used
in diverse ways and have challenged us to ask better questions and get more complete answers to how biology
works.

This compilation represents a snapshot of the exciting research in this area. We hope you will visit cell.com and cell.
com/cell-metabolism to keep up with the ever-changing landscape.

Lastly, we are grateful for the support of Cameca, who made the publication of this collection possible.

Allyson Evans
Editor-in-Chief, Cell Metabolism

For more information about Cell Press Selections:


Gordon Sheffield
Program Director, Cell Press Selections
g.sheffield@cell.com
617-386-2189 ll
pub_NanoSIMS_cell-press2_Cell press selection 03/11/2020 14:24 Page 2

NanoSIMS 50L
A revolutionary imaging
and analytical instrument
serving medical research
and drug discovery
The CAMECA NanoSIMS 50L is a unique imaging Secondary Ion Mass Spectrometer capable of
tracking molecules and quantifying biological processes at subcellular level. It has been successfully
applied to numerous life science research projects: molecular mechanisms of lipid transport, action
mechanisms of pharmaceutical compounds, investigation of the toxicology of nanomaterials...

Multiplexed antibody imaging:


the NanoSIMS 50L provides new
insights into disease pathogenesis
that will be valuable for
basic research and
drug discovery.

False color rendering of


multiplexed antibody imaging.
Ten different rare earth
elements were conjugated
with ten different antibodies.
From: Multiplexed ion beam
imaging of human breast tumor.
M. Angelo et al. NATURE
MEDECINE, 2014 Apr, 20 (4).

Scan the code


or visit cameca.com/go/nanosims-bio
to download your free copy of our booklet
“NanoSIMS 50L, a revolution in cellular biology”

cameca.info@ametek.com
www.cameca.com
Metabolism at the Single-Cell Level
Techniques for Linking Inter- and Intra-cellular Responses to Environment

Perspectives and Reviews


Immunometabolism in the Single-Cell Era Maxim N. Artyomov and Jan Van den Bossche

Single-Cell RNA Sequencing in Cancer: Lessons Learned Mario L. Suvá and Itay Tirosh
and Emerging Challenges

Subcellular Chemical Imaging: New Avenues Johan Decelle, Giulia Veronesi, Benoit Gallet,
in Cell Biology Hryhoriy Stryhanyuk, Pietro Benettoni, Matthias Schmidt,
Rémi Tucoulou, Melissa Passarelli, Sylvain Bohic, Peta Clode,
and Niculina Musat

GPIHBP1 and Lipoprotein Lipase, Partners in Plasma Stephen G. Young, Loren G. Fong, Anne P. Beigneux,
Triglyceride Metabolism Christopher M. Allan, Cuiwen He, Haibo Jiang,
Katsuyuki Nakajima, Muthuraman Meiyappan, Gabriel Birrane,
and Michael Ploug

Short Article
Age Mosaicism across Multiple Scales in Adult Tissues Rafael Arrojo e Drigo, Varda Lev-Ram, Swati Tyagi,
Ranjan Ramachandra, Thomas Deerinck, Eric Bushong,
Sebastien Phan, Victoria Orphan, Claude Lechene,
Mark H. Ellisman, and Martin W. Hetzer

Articles
Patch-Seq Links Single-Cell Transcriptomes to Human Joan Camunas-Soler, Xiao-Qing Dai, Yan Hang,
Islet Dysfunction in Diabetes Austin Bautista, James Lyon, Kunimasa Suzuki, Seung K. Kim,
Stephen R. Quake, and Patrick E. MacDonald

ll
Niche-Specific Reprogramming of Epigenetic Jason S. Seidman, Ty D. Troutman, Mashito Sakai,
Landscapes Drives Myeloid Cell Diversity in Nonalcoholic Anita Gola, Nathanael J. Spann, Hunter Bennett,
Steatohepatitis Cassi M. Bruni, Zhengyu Ouyang, Rick Z. Li, Xiaoli Sun,
BaoChau T. Vu, Martina P. Pasillas, Kaori M. Ego,
David Gosselin, Verena M. Link, Ling-Wa Chong,
Ronald M. Evans, Bonne M. Thompson, Jeffrey G. McDonald,
Mojgan Hosseini, Joseph L. Witztum, Ronald N. Germain,
and Christopher K. Glass

NanoSIMS Analysis of Intravascular Lipolysis and Lipid Cuiwen He, Thomas A. Weston, Rachel S. Jung, Patrick Heizer,
Movement across Capillaries and into Cardiomyocytes Mikael Larsson, Xuchen Hu, Christopher M. Allan,
Peter Tontonoz, Karen Reue, Anne P. Beigneux, Michael Ploug,
Andrea Holme, Matthew Kilburn, Paul Guagliardo, David A. Ford,
Loren G. Fong, Stephen G. Young, and Haibo Jiang

TREM2 Regulates Microglial Cholesterol Metabolism Alicia A. Nugent, Karin Lin, Bettina van Lengerich,
upon Chronic Phagocytic Challenge Steve Lianoglou, Laralynne Przybyla, Sonnet S. Davis,
Ceyda Llapashtica, Junhua Wang, Do Jin Kim, Dan Xia,
Anthony Lucas, Sulochanadevi Baskaran, Patrick C.G. Haddick,
Melina Lenser, Timothy K. Earr, Ju Shi, Jason C. Dugas,
Benjamin J. Andreone, Todd Logan, Hilda O. Solanoy,
Hang Chen, Ankita Srivastava, Suresh B. Poda,
Pascal E. Sanchez, Ryan J. Watts, Thomas Sandmann,
Giuseppe Astarita, Joseph W. Lewcock, Kathryn M. Monroe,
and Gilbert Di Paolo

Oxidative Metabolism Drives Immortalization of Neural François Bonnay, Ana Veloso, Victoria Steinmann,
Stem Cells during Tumorigenesis Thomas Köcher, Merve Deniz Abdusselamoglu, Sunanjay Bajaj,
Elisa Rivelles, Lisa Landskron, Harald Esterbauer,
Robert P. Zinzen, and Juergen A. Knoblich
Resource
Single-Cell RNA Sequencing Maps Endothelial Metabolic Katerina Rohlenova, Jermaine Goveia, Melissa García-Caballero,
Plasticity in Pathological Angiogenesis Abhishek Subramanian, Joanna Kalucka, Lucas Treps,
Kim D. Falkenberg, Laura P.M.H. de Rooij, Yingfeng Zheng,
Lin Lin, Liliana Sokol, Laure-Anne Teuwen, Vincent Geldhof,
Federico Taverna, Andreas Pircher, Lena-Christin Conradi,
Shawez Khan, Steve Stegen, Dena Panovska,
Frederik De Smet, Frank J.T. Staal, Rene J. Mclaughlin,
Stefan Vinckier, Tine Van Bergen, Nadine Ectors,
Patrik De Haes, Jian Wang, Lars Bolund, Luc Schoonjans,
Tobias K. Karakach, Huanming Yang, Geert Carmeliet,
Yizhi Liu, Bernard Thienpont, Mieke Dewerchin, Guy Eelen,
Xuri Li, Yonglun Luo, and Peter Carmeliet

On the cover: Increasingly sophisticated imaging techniques now allow researchers to better
understand metabolism at the single-cell level. Pictured on the cover is an image, created with
NanoSIMS, showing transport of deuterium-labeled lipids (red) across a capillary endothelial
cell (yellow) into surrounding cardiomyocytes (blue). Image credit: Haibo Jiang, Stephen Young,
and Cuiwen He.
INSIGHTS DRIVE
DRUG DISCOVERY
Are you keeping pace with peer-reviewed research
that will affect the future of therapeutics? Are
you up to speed on the latest trends, delivery
mechanisms, and policies?

Register today to receive alerts from Research Arc, a resource for R&D
professionals that draws on the vast collection of top-tier life science
review journals published by Cell Press. It’s the perfect companion
for anyone in the biopharmaceutical community looking for targeted
scientific insights that mirror the trajectory of drug development.

cell.com/research-arc
ll

Perspective
Immunometabolism in the Single-Cell Era
Maxim N. Artyomov1,* and Jan Van den Bossche2,*
1Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
2Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular
Sciences, Cancer Center Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
*Correspondence: martyomov@wustl.edu (M.N.A.), j.vandenbossche@amsterdamumc.nl (J.V.d.B.)
https://doi.org/10.1016/j.cmet.2020.09.013

SUMMARY

Emerging research has identified metabolic pathways that are crucial for the proper regulation of immune
cells and how, when deranged, they can cause immune dysfunction and disease progression. However,
due to technical limitations such insights have relied heavily on bulk measurements in immune cells, often
activated in vitro. But with the emergence of single-cell applications, researchers can now estimate the meta-
bolic state of individual immune cells in clinical samples. Here, we review these single-cell techniques and
their ability to validate common principles in immunometabolism, while also revealing context-dependent
metabolic heterogeneity within the immune cell compartment. We also discuss current gaps and limitations,
as well as identify future opportunities to move the field forward toward the development of therapeutic tar-
gets and improved diagnostic capabilities.

INTRODUCTION sion, researchers need to be able to resolve the trajectory by


which immune cells metabolically transit into their effector
Metabolism is at the core of all biological processes and can be phenotype. Arguably, single-cell profiling technology possesses
broadly subdivided into anabolic processes that use energy and the power to address all of the above questions, even at the cur-
building blocks for biosynthesis, and catabolic processes that rent state of development.
provide the raw material for anabolism as well as produce energy In this Perspective, we explore how current and emerging
to fuel biological functions. In addition to these functions, meta- state-of-the-art technologies contribute to our understanding
bolic enzymes and intermediates have important regulatory roles of metabolic regulation of immunity and discuss the transition
and are major controllers of cellular behavior. Recent studies in from bulk to single-cell immunometabolic profiling (Ahl et al.,
the field of immunometabolism have demonstrated a close 2020; Hartmann et al., 2020; Levine et al., 2020; Miller et al.,
connection between metabolic programs and the specific im- 2017; Xiao et al., 2019).
mune functions they support during both health and disease
(Ayres, 2020; Buck et al., 2017; Makowski et al., 2020; O’Neill COMMON BULK METABOLIC ANALYSES
et al., 2016). Dysregulation of these core metabolic programs
is linked to many modern diseases, including cancer and chronic Extracellular Flux Analysis
inflammatory metabolic diseases such as diabetes, obesity, Extracellular flux analyzers, such as the Seahorse apparatus,
atherosclerosis, and rheumatoid arthritis (Ketelhuth et al., perform real-time recordings of the extracellular acidification
2019; Koelwyn et al., 2018; Lim et al., 2020; Tabas and Bornfeldt, rate (ECAR) and oxygen consumption rate (OCR) as key read-
2020; Turbitt et al., 2020). Yet a detailed understanding of the im- outs of metabolism, and as such provide indirect measurements
munometabolic remodeling that occurs in pathogenic settings is of glycolysis and mitochondrial respiration, respectively. While
mostly lacking, largely due to the complex nature of the diseases the use of specific substrates and inhibitors allows one to inves-
and involved cellular phenotypes. tigate the relative preference of a cell for glucose, glutamine, or
Alterations in nutrient levels, oxygen availability, the presence fatty acids, extracellular flux analysis does not yield detailed in-
of signaling factors, and crosstalk with neighboring cells induce formation about the activity of metabolic pathways outside of
metabolic changes that allow the cell to function in its specific glycolysis and the TCA cycle within the mitochondria. Neverthe-
tissue microenvironment (Caputa et al., 2019; Van den Bossche less, the fact that this technology allows the easy, fast, and
and Saraber, 2018). As every cell resides in a unique environ- reasonably affordable profiling of cells in a 96-well format
ment, no single cell within our body is completely identical meta- strongly contributed to the expansion of the immunometabolism
bolically, phenotypically, and functionally. Addressing the field (Pelgrom et al., 2016; Van den Bossche et al., 2015; van der
spatiotemporal aspect of immunometabolism and unravelling Windt et al., 2016). Some of the key findings obtained using the
this complexity at a single-cell resolution will move the immuno- Seahorse instrument include the glycolytic switch and mitochon-
metabolism field forward, allowing translation to clinical applica- drial dysfunction in inflammatory macrophages, increased
tions and understanding the fundamentals of in vivo immu- glycolysis in effector T cells and activated dendritic cells (DCs),
nology. Indeed, to fully understand the mechanisms by which and the heightened mitochondrial respiration in T memory cells
cellular metabolism regulates immunity and disease progres- and IL-4-activated macrophages (Everts et al., 2014; Huang

Cell Metabolism 32, November 3, 2020 ª 2020 Elsevier Inc. 1


ll
Perspective

et al., 2014; Van den Bossche et al., 2016; van der Windt et al., article. For more details on immunometabolism, we refer to
2012). Noteworthy, ECAR is a surrogate marker for glycolysis, excellent and more comprehensive reviews on this subject in
and it is important to stress that acidification may not always diverse immune cells and diseases (Ayres, 2020; Buck et al.,
result from enhanced glycolysis. Indeed, CO2 production within 2017; Makowski et al., 2020; O’Neill et al., 2016).
respiring mitochondria and/or extracellular release of TCA cycle
intermediates (e.g., succinate) also acidifies the culture medium. Glycolysis Supports Diverse Immune Effector Functions
This limitation can be overcome by performing targeted metab- Immune activation is an energy-demanding process that is typi-
olomics or fluxomics for the glycolysis pathway. cally accompanied by an increase in glycolytic flux. Glycolysis
starts with the uptake of glucose by glucose transporters (e.g.,
Steady-State Metabolomics GLUT1, which is predominant in immune cells) and subsequent
For a given time point, metabolomics measures steady-state processing in the cytosol to yield pyruvate, generating ATP and
levels of a broad range of metabolites via liquid or gas chroma- reducing NAD+ to NADH during the process (Figure 1A). Elegant
tography-mass spectrometry (LC-MS or GC-MS). While untar- research highlights the direct control of immune effector func-
geted metabolomics measures hundreds of metabolites in bio- tions by the glycolytic enzymes hexokinase 1 and 2 (HK1 and
logical samples, targeted metabolomics assesses preselected HK2), glyceraldehyde-3-phosphate dehydrogenase (GAPDH),
metabolites and yields data with higher sensitivity and allows enolase, and the pyruvate kinase isoenzyme M2 (PKM2) (Chang
for accurate absolute quantification of metabolite concentra- et al., 2013; Galván-Peña et al., 2019; Moon et al., 2015; Pals-
tions. In addition to measuring small metabolites, high-resolution son-McDermott et al., 2015). To maintain glycolytic flux in the
MS-based profiling has been recently applied in ‘‘shotgun’’ lipi- absence of mitochondrial oxidative phosphorylation (OXPHOS),
domics to profile the lipidome of differentially activated macro- cells can reduce pyruvate to lactate via lactate dehydrogenase
phages (Dennis et al., 2010; Hsieh et al., 2020). Most MS-based (LDH) to recycle NADH into NAD+. This fermentation reaction
approaches still require relatively high levels of input material (on can be measured as ECAR in Seahorse analysis when protons,
the order of 100,000s of cells), yet recent developments provide along with glycolysis-derived lactate, are exported from the
an avenue to spatially measure metabolites at a single-cell reso- cell by monocarboxylate transporter MCT1. While glycolysis is
lution. For more in-depth information on the recent develop- most prominent in inflammatory immune effector cells (Krawczyk
ments in the field of spatial and single-cell metabolomics, we et al., 2010; Michalek et al., 2011), it is critical for diverse modes
refer to excellent reviews on key emerging methods, including of immune activation, including alternative macrophage activa-
matrix-assisted laser desorption ionization (MALDI)-MS imaging tion and the induction of T regulatory (Treg) cells, and glycolytic
and secondary ion mass spectrometry (SIMS) (Duncan et al., rates differ between B and T lymphocytes (Caro-Maldonado
2019; Gilmore et al., 2019). Combining such analysis with the sin- et al., 2014; De Rosa et al., 2015; Huang et al., 2016; Van den
gle-cell approaches discussed in this Perspective will certainly Bossche et al., 2016) (Figure 1B).
help to unravel the immunometabolism at single-cell resolution
within complex tissue microenvironments in vivo. The Pentose Phosphate Pathway Is Particularly
Important in Inflammatory Myeloid Cells
Fluxomics The pentose phosphate pathway (PPP) branches off from the
Altered flow through a given pathway does not necessarily corre- glycolytic pathway when hexokinase-derived glucose-6-phos-
late directly with metabolite levels within this pathway as phate is oxidized by G6P dehydrogenase (G6PD), the rate-
measured by metabolomic profiling at a set time point. The use limiting step of the PPP (Figure 1A). This oxidative branch gen-
of stable isotope 13C-labeled or 15N-labeled substrates and erates reducing equivalents of NADPH that have multiple roles
measurement of isotopologues through targeted MS at serial in immune cells. Neutrophils and inflammatory macrophages
time points allows one to determine metabolic fluxes and use the PPP and especially NADPH oxidase to generate reac-
pathway dynamics. While metabolomics provides a ‘‘snapshot’’ tive oxygen species (ROS) to combat infectious agents
of the metabolites present at one particular moment, 13C/15N la- (Figure 1B). Moreover, NADPH is used to generate fatty acids
bel-tracing (or so-called fluxomics) could be regarded as a (through fatty acid synthesis, FAS) and anti-oxidants, such as
‘‘movie’’ that yields insight into the relative flux through distinct glutathione, that prevent excessive tissue damage. Nucleotide
metabolic paths. The use of such analyses is still in its infancy and amino acid precursors needed to support cell proliferation
at single-cell resolution but can be readily used by the commu- are produced in the non-oxidative branch of the PPP. Together,
nity to quantify the rates of specific metabolic reactions in bulk both branches of the PPP support anabolic programs and im-
approaches such as the mass isotopomer multi-ordinate spec- mune effector functions (Baardman et al., 2018; Haschemi
tral analysis (MIMOSA fluxomics) as developed in the Kibbey et al., 2012).
Lab (Alves et al., 2015).
The TCA Cycle and ETC as a Central Hub
KEY IMMUNOMETABOLISM CONCEPTS DERIVED The TCA cycle and mitochondrial electron transport chain (ETC)-
FROM BULK ANALYSES based energy production are key elements of life as most cells
rely on energy production via OXPHOS, which is typically evalu-
The descriptions of the distinct metabolic pathways below pro- ated measuring OCR. Compared to glycolysis, OXPHOS is much
vide minimal immunometabolism background for the distinct more efficient in generating energy, and thus this pathway is typi-
metabolic proteins that are most frequently targeted in the sin- cally associated with the longevity of homeostatic immune cells
gle-cell metabolic profiling approaches discussed later in this such as alternatively activated macrophages or B and T memory

2 Cell Metabolism 32, November 3, 2020


ll
Perspective

Figure 1. Key Metabolic Pathways in Immune Cell Subtypes


(A) Overview of the most important metabolic pathways regulating immune cells, including the metabolic targets that were used in recent single-cell profiling
approaches.
(B) Summary of the metabolic characteristics and needs of various immune cell subsets as detailed in the text and as reviewed earlier elsewhere (O’Neill et al.,
2016; Van den Bossche et al., 2017).

cells (Lam et al., 2016; van der Windt et al., 2013). Yet during pro- Fatty Acid Metabolism Supports Immune Cell
inflammatory activation of macrophages, the TCA cycle is signif- Phenotypes and Functions
icantly remodeled via downregulation of IDH and SDH activity Fatty acid oxidation (FAO) is a catabolic pathway converting
(Jha et al., 2015). The latter is achieved through direct inhibition fatty acids into products like acetyl-CoA, NADH, and FADH2,
by the immunoregulatory metabolite itaconate that is specifically which are used in the mitochondria to generate energy. Carni-
produced by ACOD1/IRG1 in activated myeloid cells (Lampro- tine palmitoyl transferase 1 and 2 (CPT1 and CPT2) shuttle
poulou et al., 2016; O’Neill and Artyomov, 2019; Swain long-chain fatty acids into the mitochondrial matrix for subse-
et al., 2020). quent oxidation to acetyl-CoA by hydroxyacyl-CoA

Cell Metabolism 32, November 3, 2020 3


ll
Perspective

dehydrogenase (HADHA). Experiments using often too high LIMITATIONS OF CURRENT APPROACHES AND
concentrations of the CPT1-inhibitor etomoxir suggested that ASSOCIATED GAPS IN OUR KNOWLEDGE
FAO is particularly important in IL-4-induced macrophages,
memory T cells, and Tregs (Michalek et al., 2011; van der Windt The Need for In Vitro to In Vivo Validation
et al., 2012; Vats et al., 2006). Yet recent literature employing Given the importance of environmental factors, immune cell
cell-specific genetic knockouts for CPT1 and CPT2 reports metabolism in laboratory cell culture is obviously different than
that FAO is mostly dispensable for these processes (Divakaruni in vivo. Indeed, Ma et al. recently used a 13C-glucose infusion
et al., 2018; Nomura et al., 2016; Raud et al., 2018; Van den method in mice to investigate the metabolism of CD8+ T cells
Bossche and van der Windt, 2018). in response to Listeria infection and observed fundamentally
As opposed to FAO, FAS is an anabolic pathway converting different metabolic profiles for activated T cells in vivo (Ma
cytosolic acetyl-CoA into lipids. Acetyl-CoA carboxylase (ACC) et al., 2019). While in vitro-activated T cells show a shift away
first carboxylates acetyl-CoA to malonyl-CoA, which is subse- from OXPHOS toward glycolysis, CD8+ T cells activated in vivo
quently elongated by fatty acid synthase (FASN). FAS supports display higher rates of oxidative metabolism and a reliance on
cellular proliferation of effector T cells and is key to configuring, glucose-dependent biosynthesis of the amino acid serine. The
the plasma membrane for inflammatory signaling in macro- approach developed in this work requires delicate experimental
phages, and for endoplasmic reticulum synthesis to allow cyto- implementation and is not readily scalable or applicable to hu-
kine secretion by activated DCs (Everts et al., 2014; Wang man samples. This translation to the human setting is important
et al., 2011). since animal models have their limitations and do not always
replicate human immunology and biology. Generally, the further
Amino Acid Metabolism Is Differentially Regulated in development of in vivo (or ex vivo) assessments of cellular meta-
Distinct Immune Cells bolism with these and other techniques has a high priority in the
Immune activation of T cells is associated with an increased field. Meanwhile, the validation of key concepts in immunome-
demand for amino acid metabolism, as exemplified by the tabolism research in vivo, especially in humans, requires alter-
importance of L-type amino acid transporter 1 (LAT1, CD98, nate options for metabolic profiling.
and SLC7A5) and the serine pathway for successful signaling
upon TCR engagement (Hayashi et al., 2013; Ma et al., 2017,
2019). Furthermore, amino acids play a role in fine-tuning the The Need to Understand the Spatiotemporal Aspects of
specific direction of immune responses. For instance, both Immunometabolism
Th1 and Th17 cells increase glutamine usage via the trans- Immune cells heavily rely on a timely supply of nutrients, which
porter protein ASCT2 in response to antigen receptor stimula- can differ depending on the location of the cell within the micro-
tion, whereas anti-inflammatory Tregs are not affected by environment of a tumor or inflammation site (Caputa et al., 2019;
altered glutamine supply (Nakaya et al., 2014). Glutaminase Van den Bossche and Saraber, 2018). Sorting and bulk measure-
(GLS) converts glutamine into glutamate to fuel the TCA cycle, ments inevitably average profiles over cell populations, which
and this pathway promotes Th17 differentiation while diminish- obscures the effects of timing and environmental variables. As
ing Th1 and cytotoxic T lymphocyte differentiation (Johnson such, understanding the cell’s metabolic state at single-cell res-
et al., 2018). olution can explain the molecular mechanisms underlying their
In macrophages, amino acids also play important functional phenotypic heterogeneity within complex tissue microenviron-
and regulatory roles. Glutamine metabolism has been associ- ments in vivo. This could help to understand the reasons behind
ated with anti-inflammatory polarization programs, and serine their functioning, or their inability to function properly in patho-
metabolism was implicated in the regulation of IL-1b produc- logical settings. Most recent transcriptional and imaging tech-
tion (Jha et al., 2015; Rodriguez et al., 2019). Furthermore, nologies can resolve the spatial arrangement of individual cells
amino acids can also be used for the production of effector or even metabolites, and their further development will be critical
molecules in macrophages. LPS(+IFNg)-induced macro- to establish the next level of understanding of immunometabolic
phages mainly convert arginine into nitric oxide (NO) via remodeling in human samples and in in vivo settings (Mazumdar
inducible NO synthase (iNOS), while IL-4-stimulated macro- et al., 2020; Van den Bossche et al., 2017). Timing is another
phages predominantly metabolize arginine into ornithine and aspect that is often overlooked in the immunometabolism field.
polyamines (Munder et al., 1998; Van den Bossche et al., We know most immune cells rapidly ramp up glycolysis upon
2012). For more information on how amino acids regulate im- activation, and this ultimately leads to the increased metabolic
munity, we refer to an excellent recent review on this topic fluxes and abundance of specific metabolites in their effector
(Kelly and Pearce, 2020). state (typically assessed after 24 h). Yet how this happens over
Importantly, most of the concepts described so far were time and how it supports the acquisition of the effector pheno-
derived from bulk measurements in immune cells that were type and disease progression is not well understood. Resolving
cultured and activated in vitro. As discussed below, tech- the kinetics by which immune cells metabolically transit into their
nological limitations largely prevented the validation of cur- effector phenotype is essential to define which events are the
rent dogmas in vivo and the determination of the spatio- cause and what are the subsequent consequences. Together,
temporal facets of immunometabolism. Next, we describe understanding this spatiotemporal aspect of immunometabo-
the single-cell approaches addressing these key open lism will not only revolutionize our knowledge of fundamental
questions to push the immunometabolism field to the biology, but also aid the rational design of new therapeutic ap-
next level. proaches.

4 Cell Metabolism 32, November 3, 2020


ll
Perspective

Cell Numbers and Sorting Time Limitation METABOLIC ANALYSIS BASED ON SINGLE-CELL
The lack of in vitro to in vivo transitions and mouse-to-human TRANSCRIPTOMICS
validations can be at least partially explained by the techno-
logical limitations of the commonly used bulk analysis tech- With the advent of scRNA-seq, one can resolve transcriptional
niques in immunometabolism research. A major caveat of profiles of individual cells within complex multicellular ecosys-
both metabolomics and extracellular flux analysis is the tems, which is critically important in an immunological context.
need for relatively high cell numbers that are often not present Using the common 10x Genomics pipeline, scRNA-seq exper-
in human clinical samples. For example, the Seahorse XF iments nowadays profile up to ten thousand cells per sample,
analyzer measures extracellular fluxes in tens to hundreds of routinely detecting two to four thousand genes per cell. Even
thousands of cells per well and typically requires 4–5 replicate direct examination of scRNA-seq data in an immunological
wells per condition. For metabolomics, researchers typically context can be very illuminating with regard to the key meta-
measure half a million pooled cells per replicate, and even bolic processes that occur in vivo. For example, we can use
with this high input, the metabolic coverage is limited as we data on the immune cells within murine sarcomas that were
typically measure a few hundred metabolites out of the esti- treated either with immune checkpoint blockade (ICB) anti-
mated thousands present in the cell. Another drawback of PD-1/anti-CTLA-4 antibodies or with control antibodies (Gubin
extracellular flux analysis and metabolomics is the fact that et al., 2018). In this system, ICB treatment leads to tumor
both assess metabolic features that are not very stable. As rejection, which is paralleled by overall activation of the im-
such, the obtained results can be strongly affected by lengthy mune compartment and the appearance of activated cells
and harsh tissue digestion and fluorescence-activated cell within the myeloid compartment (Gubin et al., 2018). As illus-
sorting protocols (Llufrio et al., 2018). Therefore, new meta- trated in Figures 2A and 2B, we created a visualization of
bolic profiling methods accompanied by novel, optimized the information from this scRNA-seq dataset to demonstrate
cell isolation approaches are needed to measure stable meta- the behavior of some key metabolic genes in the immune
bolic features within small quantities of immune cells ex vivo. cell subsets, notably:

(i) Genes associated with increased glycolytic activity like


Leveraging Different ‘omics Approaches for Indirect Glut1 are enriched in the activated macrophages that
Metabolic Assessment appear after ICB treatment.
The instability of metabolite levels can be at least partially (ii) Nos2, a metabolic gene characteristic of inflammatory
overcome by integrating metabolomics with other ‘omics macrophage activation, is enriched in macrophages that
data that are more stable during processing such as tran- are specific to the ICB treatment condition.
scriptomics and proteomics. Metabolic fluxes can be re- (iii and iv) Phgdh expression is associated with Mki67 ex-
garded as the net result of metabolite levels and the pres- pressing highly proliferative T cells, which is
ence and activity of the enzymes that metabolize them. consistent with bulk ex vivo analyses describing
Gene expression analysis and proteomics can already give the role of serine metabolism in T cell proliferation
some insight into the regulation of metabolic pathways but (Ma et al., 2017, 2019).
become particularly powerful when integrated with metabo- (v and vi) Levels of NRF2 transcript (Nfe2l2), a key factor in
lomics data. Using a network integration approach, Jha oxidative stress responses, are generally enriched
et al. identified an important metabolic break in the TCA cy- in macrophages relative to T cells and do not
cle that supports inflammatory macrophage responses (Jha appear to depend on the treatment condition.
et al., 2015). Baardman et al. also combined metabolomics However, its direct targets such as Hmox1 do in-
and transcriptomics data to demonstrate that a defective crease in subpopulations of inflammatory macro-
PPP is associated with decreased inflammatory responses phages, consistent with potential oxidative/elec-
in lipid-laden macrophages (Baardman et al., 2018). Simi- trophilic stress from NO and itaconate. Together,
larly, integration of bulk proteomics and metabolomics re- this is in line with the fact that NRF2 is mainly regu-
vealed the metabolic requirements for T cell activation (Gei- lated on the protein level (Taguchi et al., 2011) and
ger et al., 2016). These distinct ‘omics approaches, highlights the need to profile protein rather than
especially when integrated, can reveal (de)regulated meta- gene expression for this key metabolic regulator.
bolic hubs that can be functionally tested using genetic
tools or pharmacological compounds. A similar approach of direct examination of specific key meta-
Single-cell metabolomics is currently too premature for large- bolic genes in scRNA-seq data proved to be powerful in
scale applications, and thus alternate approaches are needed describing the metabolic aspects of apoptosis during Drosophila
for immunometabolism research to enter the single-cell era. Pre- eye development (Ariss et al., 2018). Together, these examples
vious successes using bulk transcriptomics and proteomics to illustrate the potential utility of scRNA-seq data for studying
delineate cellular metabolism show that using the related sin- metabolism. Yet methods to comprehensively map the meta-
gle-cell ‘omics approaches is the way to go at the moment. To bolic landscape at single-cell resolution remain relatively scarce
this end, the latest single-cell RNA sequencing (scRNA-seq) and/or are in the early stages of development. Most approaches
and cytometry-based approaches that map the metabolic land- for metabolic analysis of scRNA-seq data arise from bulk RNA-
scape at single-cell resolution are discussed in the next parts of seq analysis techniques, and considerable challenges have to
this article. be overcome when making this transition.

Cell Metabolism 32, November 3, 2020 5


ll
Perspective
Figure 2. Expression of Some Key
Metabolic Genes in Intratumoral Immune
Cells Based on Published Single-Cell
Transcriptomics Data
The behavior of some key metabolic genes is
shown in immune cells of mouse sarcomas that
were treated with either anti-PD1/anti-CTLA4 im-
mune checkpoint blockade antibodies or control
antibody (Gubin et al., 2018) (A). To do so, we
created a direct visualization of the indicated
genes from the associated published dataset
(GEO: GSE119352) (B).

RNA content per cell, which results in un-


even coverage between cell types. For
example, a smaller number of total tran-
scripts is typically detected in neutrophils
as compared to T cells or macrophages.
Thus, care is required when comparing
pathway enrichments between different
cells since lower coverage of some clus-
ters can lead to false-negative results.
Most of the approaches for metabolic
analysis of scRNA-seq data can be
broadly classified into two main cate-
gories that are briefly described below:
(1) pathway-based analysis and (2) flux
balance analysis (FBA)-based methods.

Pathway-Based Approaches
Genes composing distinct metabolic
pathways are fairly well annotated (Kane-
hisa and Goto, 2000) and can be directly
probed as gene sets using standard bio-
Indeed, bulk transcriptional profiling provides an appealing informatics pathway enrichment techniques. This simple yet
source of data as it yields exhaustive information about tran- powerful approach can reveal novel biological insights. For
script levels within the sequenced cells. As such, the absence example, the role of serine metabolism in T cells has been inves-
of a signal typically implies absence of the corresponding tran- tigated based on the initial observation of the transcriptional
script. This is in contrast to metabolomics profiling, where enrichment of this pathway in published bulk RNA-seq data
coverage is limited and absence of a signal often arises due to (Ma et al., 2017). As Figure 2 shows, this pathway is enriched
technological limitations rather than due to absence of the in the proliferating T cell cluster within the scRNA-seq data.
metabolite itself. Yet this advantage of transcriptional profiling Recently, Xiao et al. demonstrated how pathway enrichment
is not as pronounced in scRNA-seq data. Due to generally lower pipelines can be adjusted for scRNA-seq data analysis and
depth of sequencing and drop-out events, expression levels of used it to characterize the metabolic diversity within the tumor
individual genes can appear zero in some cells even though microenvironment. Using a similar analysis strategy, Miragaia
these genes are actually expressed. Typical depth of sequencing et al. (2019) highlighted the metabolic heterogeneity among
is in the order of 25,000–50,000 reads per cell, which yields Tregs across different tissues using pathway enrichments.
confident detection of 3,000–4,000 genes per cell. Shallow
sequencing (i.e., on the order of 1,000 genes per cell) might result Flux Balance Analysis-Based Approaches
in detection of mostly common and housekeeping genes and FBA is an elegant way to compute the steady-state distribution of
only very few subpopulation-specific genes, which would lead metabolic fluxes in the system that would satisfy constraints on
to significant difficulties when comparing the data with other input and output fluxes given a specific network architecture
published signatures. Still, even the detection of 3,000–4,000 (Orth et al., 2010). Simply put, FBA views metabolic wiring in a
genes per cell is significantly lower than the estimated 12,000– cell as a system of pipes with a limited number of input and output
15,000 genes expressed in purified cell populations based on faucets and requires that there is steady-state equilibrium estab-
the bulk RNA-seq data. In these situations, techniques such as lished at every node of a system. In other words, incoming meta-
data imputations have to be used to post-process the data bolic flow toward each reaction should equal metabolic flow going
and to obtain smooth transcriptional profiles. Within the same away from that reaction. As such, specific expression levels of in-
scRNA-seq dataset different cell clusters can have very different dividual enzymes are not directly participating in modeling and the

6 Cell Metabolism 32, November 3, 2020


ll
Perspective

results are mostly dependent on network topology and require- metabolism research yet, recent literature showcases the use
ments on input and output reactions of the model. Inputs are typi- of mass cytometry (cytometry by time of flight, CyTOF) to esti-
cally the most common substrate uptake reactions (e.g., glucose, mate the metabolic configurations of enzymes within single cells.
glutamine), and outputs (usually referred as objective function) are This technology allows for simultaneous quantification of 40
associated with the specific studied cell behavior. For instance, an parameters at single-cell resolution (Hartmann and Bendall,
objective can be to maximize the production of biomass for 2020). In the context of immunometabolism, one should
growing cells or the production of certain metabolites such as consider the inclusion of antibodies targeting the main metabolic
NO for activated macrophages (Bordbar et al., 2012). features described above (Figure 1). Not surprisingly, the meta-
An important advantage of the FBA-based approaches is that bolic panels that were independently designed by three distinct
they rely on network analysis that is not associated with prede- research groups show considerable overlap (Table 1) (Ahl et al.,
fined pathway knowledge and therefore can reveal novel biolog- 2020; Hartmann et al., 2020; Levine et al., 2020).
ical concepts. As such, it provides a powerful tool to understand To design an immunometabolic CyTOF panel, Hartmann et al.
changes in metabolic flux upon changes in network topologies, first screened over 100 commercial antibodies against a broad
e.g., due to gene knockout or tissue-specific pattern of expres- range of metabolic factors and came to a selection of 41 meta-
sion. Indeed, Zhang et al. have demonstrated scRNA-seq-based bolic antibodies that passed all biological controls and yielded
analysis of NAD+ biosynthesis in different tissues using an FBA- robust reproducible results in a proof-of-concept study of hu-
based approach (Zhang et al., 2020). Conceptually, this work fol- man white blood cells (Hartmann et al., 2020). After identifying
lows from the approach of studying tissue-specific metabolism distinct immune cell subsets based on their lineage marker
based on bulk RNA-seq data from different tissues (Bordbar expression pattern, the metabolic states of all immune cell sub-
et al., 2011). In addition to looking at the dramatically different sets could be estimated and compared without the need for prior
network topologies, the FBA framework allows investigation of sorting. The metabolic state profiles acquired by mass cytometry
metabolic remodeling within the same network, but upon the were in agreement with their described roles in specific immune
change of the required cellular output induced by a stimulation. functions as exemplified by high expression levels of targets
This is feasible when at least some key metabolic features are associated with glucose and fatty acid metabolism in plasmacy-
known. For instance, using this approach, Bordbar et al. toid DCs, and high levels of the rate-limiting PPP enzyme G6PD
modeled activation of macrophages using increase in NO pro- in neutrophils (O’Neill et al., 2016). Furthermore, Hartman et al.
duction as a key objective of inflammatory macrophages (Bord- have demonstrated a high degree of consistency between meta-
bar et al., 2012). Key objectives can also be defined based on the bolic changes in T cells measured by Seahorse and the changes
differential expression between conditions. This approach for observed at a single-cell resolution on the protein level. This
bulk data has previously been adopted to reveal itaconate as highlights that CyTOF-based single-cell metabolic profiling is a
an SDH inhibitor (Lampropoulou et al., 2016) and has recently powerful tool to gain new insights into the concepts of metabolic
been translated to scRNA-seq data to investigate metabolic dif- remodeling established by bulk measurements.
ferences associated with pathogenic/non-pathogenic Th17
cells, as well as their difference with Tregs (Wagner et al., 2020). Application of Metabolic CyTOF in the Ex Vivo Clinical
One significant caveat of the current approaches is that they Context
typically focus on differences between two distinct cell popula- Comparing tumor and healthy adjacent tissue from colorectal
tions. In these situations, a more straightforward approach carcinoma patients, along with healthy donor peripheral blood
would be to simply perform differential expression analysis be- mononuclear cells and lymph nodes, Hartmann et al. (2020) re-
tween the two populations and perform either pathway enrich- vealed specific metabolic phenotypes that were enriched in tu-
ment analysis or metabolic subnetwork analysis. We argue that mor-associated CD8+ T cells. These cells showed increased
it is critical to move past the one-to-one comparison of known expression of the large neutral amino acid transporter 1 (LAT1,
cell types and to learn to capture metabolic diversity within the CD98) and decreased expression of a broad range of enzymes
whole dataset. Thus, the next generation metabolic scRNA- across distinct metabolic pathways. These so-called metalow
seq analysis approaches is expected to reveal major metabolic cells displayed distinct signs of T cell exhaustion, including
axes of variation independent of the assignments of the cellular increased expression of PD1 and CD39, reduced mitochondrial
identities. While it is feasible at the moment to perform such clus- capacity, and low levels of TCF1. Of note, a fraction of the met-
tering simply in the space of all metabolic genes or using anno- alow subset did not express PD1 or CD39, indicating that
tated metabolic pathways as principal components, the most combining metabolic and immune cell markers provides addi-
powerful clustering approaches would incorporate network- tional dimensions to phenotypically and functionally define tu-
based analysis with no a priori knowledge of individual path- mor-associated immune cells in relation to human disease.
ways. This would allow for unbiased definition of individual cells
as distinct metabolic entities and will provide an unbiased view of Use of CyTOF-Based Metabolic Profiling in Animal
the whole cellular ecosystem from a metabolic perspective. Model Research
In addition to human samples, metabolic CyTOF panels can be
PROTEIN-BASED SINGLE-CELL METABOLIC ANALYSIS useful to dissect metabolic remodeling in vivo in animal models.
Levine et al. used CyTOF-based metabolic profiling of T cell re-
Single-Cell Metabolic Profiling by Mass Cytometry sponses during Listeria monocytogenes infection as a well-
While untargeted analysis of proteins within single cells is still in described model of CD8 T cell differentiation (Levine et al.,
its infancy (Slavov, 2020) and has not been applied in immuno- 2020). The metabolic panel configured here was largely based

Cell Metabolism 32, November 3, 2020 7


ll
Perspective

Table 1. Metabolic Targets Used in Different Single-Cell Profiling Approaches


Metabolic Pathway and Inclusion in Panels
Target Function Hartmann Levine Ahl Miller Corea
Glycolysis
GLUT1 glucose uptake x x x x
HK first glycolytic enzyme x x (x)
PFK2 glycolytic enzyme and x
regulator
GAPDH glycolytic enzyme/ x x X x
immune regulator
PKM2 glycolysis/transcriptional x
regulator
Fermentation
LDH pyruvate to lactate x x (x)
conversion
MCT1 lactate export x (x)
PPP
G6PD G6P dehydrogenase, x x x x
rate-limiting PPP step
TCA/ETC
CS citrate synthase x x (x)
IDH isocitrate x x x x
dehydrogenase
OGDH alpha-ketoglutarate x (x)
dehydrogenase
SDH succinate x x
dehydrogenase/ETC
complex II
ATP5A ATP synthase/ETC x x x x
complex V
CytC cytochrome c, essential x x (x)
for ETC function
Mitochondrial Regulation
PGC1a mitochondrial biogenesis x
VDAC1 outer mitochondrial x x (x)
membrane porin
OPA1 mitochondrial fusion x
Fatty Acid Metabolism
CD36/FAT fatty acid transporter x
CPT1A fatty acid shuttling into x x x
mitochondria
HADHA long-chain fatty acid x
oxidation
ACADM medium-chain acyl-CoA x x
dehydrogenase
ACC/ACAC acetyl-CoA carboxylase/ x x x
fatty acid synthesis
Amino Acid Metabolism
CD98/LAT1 essential amino acid x x x
transporter
GLUD glutamate x
dehydrogenase
ASCT2 alanine/serine/cysteine- x
preferring amino acid
transporter 2
(Continued on next page)

8 Cell Metabolism 32, November 3, 2020


ll
Perspective

Table 1. Continued
Metabolic Pathway and Inclusion in Panels
Target Function Hartmann Levine Ahl Miller Corea
GLS glutamine to glutamate x
conversion
GOT2 glutamic-oxaloacetic x
transaminase/malate-
aspartate shuttle
ASS1 arginosuccinate x
synthase
Metabolic Regulation/Signaling
NRF1 anti-oxidant transcription x x (x)
factor
NRF2 anti-oxidant transcription x
factor
KEAP1 NRF2 degradation x
PRDX2 peroxiredoxin 2 anti- x
oxidant enzyme
HIF1A hypoxia and x x x
inflammation-induced
transcription factor
mTOR central hub of nutrient x
signaling/anabolic
metabolism
S6-P ribosomal protein x x x
controlled by mTOR
XBP1 ER stress transcription x
factor regulating diverse
immune cells
Functional Output
CyclinB1 proliferation/clonal x
expansion
Ki-67 proliferation/clonal x x
expansion
BrU RNA biosynthesis x
IbU DNA biosynthesis x
Puromycin protein biosynthesis x
a
A core metabolic cytometry panel of 10 targets is proposed, along with additional targets (x) for further insight

on earlier bulk metabolic measurements and previous literature, This newfound subset appeared 4 days after infection, was high-
and as such, the obtained mass cytometry data nicely recapitu- ly proliferative, showed strongly induced expression of both
lated established concepts in the field. Indeed, effector T cells glycolysis and mitochondrial oxidation markers, and appeared
showed an expected increase in glycolytic activity (Michalek to be fueled by both fatty acids and amino acids based on the
et al., 2011) as evident by GLUT1 and GAPDH induction in Cy- expression of CTP1a and CD98, respectively. Having detected
TOF analysis and as validated by an increased ECAR by Sea- this unknown subset of transitional cells by CyTOF, the authors
horse analysis. Conversely, memory T cells showed induction next sorted these cells as CD62Llo CD44hi CD25hi to confirm their
of CPT1a, a key player in FAO that was previously associated increased glycolytic and oxidative metabolism through extracel-
with CD8 memory T cells (van der Windt et al., 2012). This meta- lular flux analysis. This is an excellent example of how single-cell
bolic rewiring occurred in a stepwise manner that could be fol- technologies provide new insights and how established bulk
lowed over time at single-cell resolution. This metabolic change analysis can be used to validate such new discoveries.
included a uniform glycolytic switch early after activation, fol-
lowed by heterogeneous GAPDH expression after 2 days of acti- Met-Flow: Single-Cell Metabolic Profiling by Flow
vation. These two GAPDH populations showed functional differ- Cytometry
ences, which would not have been revealed in bulk approaches. By using fluorescently labeled antibodies instead of heavy-
Furthermore, this approach revealed a transitional pool of metal-conjugated ones, single-cell metabolic profiling can also
effector T cells that could not be detected when solely using be performed by ‘‘regular’’ flow cytometry (with some advan-
common antibodies against lineage markers and T cell subsets. tages and disadvantages; see later). Ahl et al. configured a

Cell Metabolism 32, November 3, 2020 9


ll
Perspective

flow cytometry-based method called Met-Flow to interrogate the the detection of 40 proteins, including cell lineage markers,
metabolic state of immune cells using 10 fluorochrome-conju- as well as selected metabolic enzymes such as the ones in
gated antibodies targeting rate-limiting enzymes and key pro- Figure 1A and Table 1. Therefore, utilization of cytometry ap-
teins in distinct metabolic pathways, combined with phenotypic proaches requires reasonably specific hypotheses to design
markers to generate a 27-color flow cytometry panel (Ahl et al., the antibody panel.
2020). To acquire this extensive panel, the authors used an X- At the same time, the typical number of cells that is profiled by
30 FACSymphony cytometer from BD. Using the expression pro- the two approaches is very different, as scRNA-seq is currently
files of 10 metabolic markers allowed Ahl et al. to cluster and limited to about 10,000 cells per sample, while cytometry can
separate blood leukocyte subsets into distinct metabolic islands. routinely process millions of cells and can be readily scaled up
Akin to the CyTOF analysis by Hartmann et al., CD4+ and CD8+ if needed. This distinction becomes critical when minor cellular
T cell subsets were not separated on metabolic proteins alone, populations like innate lymphoid cells or DCs need to be consid-
but most other immune cell subsets were quite efficiently defined ered. Furthermore, the cost of single-cell transcriptional profiling
solely based on their metabolic profiles. This indicates that it is is nearly an order of magnitude larger than that of a cytometry
probably more beneficial to extend our conventional cytometry run, and therefore it is not feasible to profile either large numbers
panels with antibodies against key metabolic enzymes that are of replicates or to increase statistical power by profiling many
coupled to specific immune effector functions, instead of stain- cells from the same samples. Additionally, scRNA-seq is more
ing additional immune cell surface markers for which the actual sensitive to cellular morphology compared to cytometry ap-
function is often unknown. Even when using relatively small cy- proaches. A number of cell types, such as myocytes or neurons,
tometry panels, including a few metabolic antibodies, it will prob- cannot be reliably profiled due to incompatibility of cellular
ably be highly informative to further dissect the phenotype, func- shapes and instrumental design. In other cases, such as neutro-
tion, and metabolism of immune cells using equipment that is phils, transcriptional profiling is complicated by fragility of the
present in virtually every institute. This also supports the idea cells and overall lower RNA content of the cells, which introduces
of Poznanski and Ashkar, who previously addressed the ques- significant biases into the data.
tion, ‘‘If an NK Cell Cannot be Defined by How It Looks, Could On the other side, one significant advantage of transcriptional
It be Defined by How It Is Fueled?’’ in an elegant review on which profiling is rooted in the utilization of single-nucleus RNA-seq. In
factors regulate NK cell function (Poznanski and Ashkar, 2019). this approach, one can isolate individual nuclei and profile corre-
Of note, Met-Flow with only 10 metabolic proteins identified sponding transcripts (Mereu et al., 2020). This approach has
blood leukocyte subsets with a resolution comparable to that of several important advantages, even though the number of de-
+500 genes by scRNA-seq analysis, whereas solely analyzing tected transcripts is typically much smaller due to the fact that
the expression of the same 10 genes did not resolve the immune most RNA is localized in the cytosol and not in the nucleus
populations. Additionally, Met-Flow demonstrated the well-docu- (Ding et al., 2020). First, this approach avoids morphological
mented upregulation of glycolysis, OXPHOS, and FAS upon T cell complications and all cell types within the sample can be profiled
activation, and this metabolic rewiring was confirmed by bulk on the same footing. But most importantly, this approach allows
extracellular flux analysis. Follow-up single-cell Met-Flow analysis profiling of frozen samples, including ones that have been bio-
indicated that most cells relied on glucose for their metabolic banked years before. This opens up the ability to study cohorts
switch, but also revealed a subset of CD4 central memory cells of tissues from clinical samples that typically have to be
with an alternative metabolic fuel. As such, single-cell, but not collected across extended periods of time.
bulk, metabolic profiling is able to identify specific metabolic char- In spite of these advantages, transcriptional data provide only
acteristics and requirements of individual T cell subsets. a ‘‘twice-removed’’ proxy to the metabolic measurements, and
in that respect, cytometry is a more direct approach that yields
PROS AND CONS OF DISTINCT SINGLE-CELL protein-level measurements. Antibody-based approaches
METABOLIC PROFILING TECHNIQUES AND directly measure levels of enzymes, including their post-transla-
DEVELOPMENTS tional modifications (and the modification of their key regulators).
An additional benefit of using cytometry-based approaches is
Single-Cell Transcriptomics versus Cytometry that it allows inclusion of non-protein-based markers of cellular
Single-cell transcriptomics and multicolor cytometry are com- phenotypes. For instance, by including tagged IdU, BrU, and pu-
plementary approaches in terms of information acquired and romycin substrates, metabolic profiles and cellular phenotypes
experimental set-up required for profiling. As such they can be can be functionally linked to anabolic process like DNA, RNA,
used simultaneously for maximal resolution, but at the same and protein synthesis (Kimmey et al., 2019).
time either approach can be more accessible or informative. Altogether, in the setting of controlled experimental design we
Here, we describe some basic considerations for selecting the would advocate for a combinatorial approach where scRNA-seq
most optimal approach (Table 2). is run on a limited scale to explore the broad landscape of the
scRNA-seq typically yields 3,000–5,000 genes detected in changes, followed by large-scale cytometry profiling using a hy-
each cell. This allows for unbiased clustering of the data and po- pothesis-driven antibody panel for validation at the protein level.
tential identification of novel subpopulations that have not been
described before. From the metabolic perspective, it allows Mass versus Flow Cytometry for Single-Cell
simultaneous detection of multiple genes across many pathways Immunometabolism Research
and therefore yields a global picture of metabolic changes. On Flow cytometry is still the most commonly used approach for im-
the other side, cytometry approaches are currently limited to mune cell profiling, but the number of parameters that can be

10 Cell Metabolism 32, November 3, 2020


ll
Perspective

Table 2. Strengths and Weaknesses of the Techniques


Strengths Weaknesses
Extracellular flux analysis (indirect) functional readout high cell number and replicates needed
availability of apparatus limited to glycolysis and mitochondrial
respiration
affordable and fast affected by cell isolation and sorting
Bulk metabolomics and fluxomics info on a broad range of pathways instability of metabolome
most direct assessment of metabolism high levels of input material needed
applicable in vivo/ex vivo (advanced) costly
Single-cell transcriptomics Unbiased limited to 10,000 cells/sample
combined metabolic and phenotypic info limited sequencing depth/coverage
generation of new hypotheses costly
Flow cytometry combination with metabolic dyes targets need to be selected carefully
(biased)
machinery available in most institutes fluorescent spill-over and background
affordable and fast indirect metabolic measurements
Mass cytometry (CyTOF) largest antibody panel size only available in high-end (core) facilities/
costly
barcoding of multiple samples lower sensitivity
stability of signal indirect metabolic measurements
MIBI-TOF (indirect) spatial assessment of metabolism advanced high-end technique (availability/
cost/time)
large antibody panels indirect metabolic measurements
can be performed on biobanked material
In situ activity assays activity measurements in situ limited to dehydrogenases
measures selected enzymes in key measured at saturated substrate levels
pathways
specialized technique
MS imaging/single-cell metabolomics direct in situ measurements of metabolism advanced high-end technique (availability)
broad range of metabolites/lipids measured to be combined with phenotypic/metabolic
readouts

simultaneously tested is relatively limited due to fluorescence handled together once the clinical trial or experiment is finalized.
spillover. This occurs when the fluorescence emission of one Indeed, unique barcoding of up to 20 samples allows one to
fluorochrome is sensed by the detector of another fluorochrome. combine and subsequently stain, process, and acquire them
Therefore, flow cytometry data need to be carefully compen- as one multiplexed sample. These advantages come with a
sated, and this can be particularly challenging for large panels. cost since CyTOF antibodies are more expensive than fluoro-
Nevertheless, the number of colors and antibodies used in flow chrome-coupled ones, and the instrument used to acquire the
cytometry is growing fast and panels of 20 and more markers labeled cells is more costly and advanced in the case of mass cy-
are now common. So far, it does not reach the +40 parameters tometry. While multi-color flow cytometers are the workhorses of
that can be assessed in CyTOF, but at this point flow cytometry virtually every lab, mass cytometers are less common and
is clearly catching up. Opposed to fluorescence detectors, Cy- restricted to high-end core facilities.
TOF uses a mass cytometer to detect the TOF of each metal Nowadays, spectral analyzers are pushing the boundaries of
that is set by its mass (Spitzer and Nolan, 2016). As such, both flow cytometry. Employing 5 lasers to detect the full emission
the detection overlap and background (autofluorescence in spectra over 64 channels, instruments like the Cytek Aurora
flow cytometry) are very low in CyTOF analyses because heavy are now able to detect 30+ colors in a sensitive, fast, and acces-
metals do not naturally occur in cells. Designing ideal CyTOF sible manner (Ferrer-Font et al., 2020). This is particularly valu-
panels is still key to minimize ‘‘spillover’’ due to impurities of able for (immuno)metabolism research since it can combine fluo-
metal tags and to take differences in signal intensity of distinct rochrome-tagged antibodies against metabolic proteins and
metals into account. Also, no mass channel in CyTOF is as sen- immune lineage markers with fluorescent metabolic tools to pro-
sitive as bright fluorochromes such as PE in flow cytometry. vide an extra layer of metabolic information (Scharping et al.,
While stained cells need to be acquired fast in flow cytometry 2016). For example, expression of the glucose transporter
to prevent photobleaching, metal-tagged samples can be GLUT1 can be directly related to the uptake of fluorescently
(cryo)preserved for weeks and acquired simultaneously when labeled 2NBDG. Similarly, increased expression of proteins
all samples are collected over time during a clinical trial. Alterna- that suggest increased mitochondrial oxidation can be directly
tively, cells can be isolated, fixed, and stored over time and linked to mitochondrial mass and membrane potential using

Cell Metabolism 32, November 3, 2020 11


ll
Perspective

fluorescent dyes such as MitoTracker and TMRE (or related mitochondrial respiration, or amino acid metabolism were often
TMRM), respectively. As such, a clear advantage of fluores- surrounded by neighboring cells expressing the same metabolic
cence-based flow cytometry is the applicability of fluorescent target. This microenvironment-driven metabolic polarization was
metabolic dyes, whereas the largest benefits of mass cytometry especially apparent when comparing immunometabolic profiles
are panel size, signal stability, and barcoding possibilities that of cells near the tumor border with the ones positioned farther
allow in-depth single-cell metabolic profiling of immune cells in away from the boundary. Typically, metabolically suppressed
patient biopsies obtained during clinical trials. cells were farther away from the tumor border, while metaboli-
cally active cells resided at the tumor-immune edge. Since the
SPATIALLY RESOLVED IMMUNOMETABOLISM MIBI-TOF approach can be performed on existing FFPE material
obtained from biobanks, it offers the opportunity to link immuno-
A major drawback of all approaches listed so far is that they metabolic profiles and locations to clinical outcome and thera-
require cells in suspension and as such lack spatial information. peutic success. As such, single-cell metabolic profiling is an
The spatial aspect is particularly important since specific micro- important next step to translate immunometabolism research to-
environmental factors, including the complex mixture of stimuli, ward clinical diagnostics and therapy. Yet it should be noted that
availability of nutrients, and interactions with neighboring cells, the presence of a particular enzyme does not always correlate
are key drivers of cellular metabolic profiles in tissues and criti- with its activity, and this aspect should be taken into account
cally shape immunity and disease progression. Another draw- when drawing conclusions from these antibody-based ap-
back of conventional single-cell profiling approaches is that the proaches.
specific protocols used to digest tissues into single cells induce
cell death in some, but not all, cells and might favor the isolation Mapping Metabolism within Microenvironments In Situ
of specific immune cell subsets with associated metabolic pro- Using Dehydrogenase Activity Assays
files. Together, these limitations underscore the need for the vali- Miller et al. previously configured an alternate method to assess
dation of key observations of immunometabolic features of cells the metabolic configuration of cells in their tissue microenviron-
within their tissue microenvironment, taking into account their ment (Miller et al., 2017). Their approach relies on enzyme activ-
spatial organization and avoiding problems associated with ity measurements at saturating substrate and co-factor availabil-
cell sorting. New methods that instantaneously capture spatial ity in combination with staining of multiple immune cell markers
context and single-cell transcriptome profiles are now being on consecutive tissue sections. Dehydrogenase activities are
applied, but their use in immunometabolism research has yet measured for five enzymes catalyzing key steps in major meta-
to be described. Nevertheless, recent publications highlight bolic pathways: G6PD in the PPP, GAPDH in glycolysis, LDH in
how the metabolic configuration of single cells within their micro- lactate fermentation, and IDH and SDH in the TCA cycle. When
environment can be assessed at the protein level in a highly mul- these dehydrogenases are active, nitroblue tetrazolium chloride
tiplexed manner. (NBT) is reduced by NAD(P)H into a strongly colored state that
can be quantified and related to the acquired immune cell marker
MIBI-TOF as Protein-Based Spatial Single-Cell Immune/ expression on consecutive sections. The authors employed this
Metabolic Analysis technique to interrogate the metabolic signatures of macro-
Hartmann et al. took an important step toward spatial metabolic phage and T cell subsets in human cancerous and control colon.
profiling by transferring their established metabolic CyTOF panel Macrophages producing IL-6 and TNF within the tumor showed
to the multiplexed ion beam imaging (MIBI-TOF) platform. This increased glycolytic GADPH activity in comparison with non-in-
recently developed approach couples single-cell metabolic pro- flammatory macrophages within the TME, but overall tumor-
files, immune cell phenotypes, and functional states to cell-cell associated macrophages appeared metabolically repressed in
interactions and location within tissues (Hartmann and Bendall, comparison to macrophages in healthy tissue. It is therefore
2020; Keren et al., 2019). To do so, tissue sections are stained tempting to speculate that an impaired metabolic fitness of mac-
with heavy-metal-coupled antibodies (that are also used in Cy- rophages in tumors might prevent their antitumor activities.
TOF), and then scanned with an ion beam to release the heavy Moreover, Tregs showed cancer-specific metabolic features
metals from antibodies recognizing specific metabolic and im- with suppressed glycolytic GAPDH activity and increased mito-
mune targets. For each acquired pixel, the released ions are chondrial SDH compared to Tregs in healthy tissues. Identifying
quantified by TOF-MS as in CyTOF, with the important advan- such tumor-specific metabolic properties of immune cell sub-
tage that the obtained high-dimensional data obtained by sets could aid the development of new therapeutic approaches.
MIBI-TOF also contain spatial information. Of note, the activities of these distinct dehydrogenases are
Staining colorectal carcinoma and control tissue sections with measured at saturated substrate concentrations and are thus
lineage markers in combination with a broad range of metabolic estimates of the optimal in vivo scenario rather than a precise
antibodies, followed by segmentation of individual cells in the ac- reflection of their activity in a given context such as a hypoxia
quired images and subsequent clustering of cells into cell line- TME in which cells compete for nutrients.
ages, allowed researchers to couple immune cell phenotypes
to metabolic characteristics that are driven by their specific tis- CONCLUSIONS AND FUTURE DIRECTIONS
sue microenvironment (Hartmann et al., 2020). Metabolic profiles
were spatially organized in environmental niches with similar It is clear that a technological transition toward understanding
metabolic characteristics irrespective of cell type. Cells express- immunometabolism at the single-cell level is inevitable, although
ing high levels of metabolic factors associated with glycolysis, not yet complete. Technologies developed for transcriptomic

12 Cell Metabolism 32, November 3, 2020


ll
Perspective

Figure 3. Hypothetical Technology Development Perspective in the Context of Single-Cell Immunometabolism Applications

and proteomic profiling at single-cell resolution demonstrate a but rather in MS-based imaging approaches to spatially resolve
sufficient level of maturity to establish the current frontier in the the metabolic configuration of single cells within their tissue
field of immunometabolism—initial assessment and focused microenvironment. For detailed information on emerging tech-
probing of the metabolic landscape within clinical samples and niques such as MALDI-MS for single-cell and even subcellular
in vivo models of inflammation. As discussed above, specific analysis, we refer the reader to excellent recent reviews (Gilmore
metabolic features can be informed by scRNA-seq profiling or c
et al., 2019; S upáková et al., 2020).
even bulk observations from mixed cell cultures/ex vivo samples Resolving immunometabolism at single-cell resolution is
and then dissected through dedicated protein-targeting panels. certainly not the end stage as cells are not homogeneous bags
For instance, based on the overlap between the recently pub- but rather highly compartmentalized structures with a heteroge-
lished panels and previous literature, one can compile a core neous distribution of metabolites with different functions at
metabolic panel consisting of 10 metabolic antibodies that is different subcellular locations. For example, acetyl-CoA feeds
also applicable in multicolor flow cytometry and a more exten- the TCA cycle within the mitochondria, while fueling fatty acid
sive set panel including an additional 10 antibodies to gain and cholesterol biosynthesis in the cytosol and histone acetyla-
more detailed and functional insight through CyTOF analysis (Ta- tion within the nucleus. As such, improving the subcellular reso-
ble 1). lution of metabolic readouts will provide an additional layer of
While methods such as CyTOF and scRNA-seq can be insight- insight into the immunometabolism field and to science in gen-
ful with respect to metabolic remodeling within the diverse cell eral (Alexandrov, 2020; Pareek et al., 2020).
subpopulations, the primary data obtained with these methods In the context of complementary ‘omics profiling, fusion of
remain either indirect or limited in scope. Therefore, the techno- protein and transcriptomic level measurements along the lines
logical advances that can address these challenges will build up implemented in CITE-seq approaches bears significant promise
a foundation for the next generation of immunometabolism for the immunometabolism field (Stoeckius et al., 2017). Specif-
research (Figure 3). Two key directions would be (1) improving ically, extension of the CITE-seq-based approach to robust
depth and quality of data for the single-cell level profiling of measurement of intracellular proteins would allow for measuring
ex vivo cell suspensions, and (2) improving our ability to resolve significantly larger numbers of enzyme and their post-transla-
the spatial distribution of metabolic features (e.g., within tissue tional modifications since antibodies are conjugated to nucleic
slices). acid barcodes and therefore 100–200 proteins can feasibly be
The next generation approaches would include direct single- measured simultaneously (Hwang et al., 2020; Stoeckius et al.,
cell metabolite profiling (Duncan et al., 2019), which should be 2017). This will significantly improve the resolution compared
accompanied by the development of robust methods to isolate to the current 40 antibodies possible with CyTOF, especially
individual cells without significant metabolic perturbations (Fig- considering the fact that almost half of the panel is typically
ures 3). Such an approach would not only allow direct evaluation devoted to cell-type-specific markers.
of metabolite levels, but also provide fluxomics at a single-cell Furthermore, adopting new data generation techniques will
level, when cell mixtures, or even animals and people, are sup- demand development of a portfolio of novel computational ap-
plemented with labeled substrates. Given the unstable nature proaches that could integrate the network-based nature of the
of the metabolome and the fact that isolation of cells affects data with corresponding technological outputs in terms of spatial
the readout, the future of single-cell metabolomics is probably location or potential temporal aspects of immune responses.
not in measuring metabolites in isolated cells in suspension, The use of computational algorithms such as SCORPIUS

Cell Metabolism 32, November 3, 2020 13


ll
Perspective

(Cannoodt et al., 2016) is promising to deduce a pseudotime Baardman, J., Verberk, S.G.S., Prange, K.H.M., van Weeghel, M., van der Vel-
den, S., Ryan, D.G., Wu €st, R.C.I., Neele, A.E., Speijer, D., Denis, S.W., et al.
ordering to investigate the presumed temporal sequence of (2018). A defective pentose phosphate pathway reduces inflammatory macro-
metabolic rewiring. Understanding the kinetics of phenotypic, phage responses during hypercholesterolemia. Cell Rep. 25, 2044–2052.e5.
functional, and metabolic changes can reveal which metabolic
Bordbar, A., Feist, A.M., Usaite-Black, R., Woodcock, J., Palsson, B.O., and
events are causal and what are mere bystanders of a particular Famili, I. (2011). A multi-tissue type genome-scale metabolic network for anal-
immune state. Understanding the spatiotemporal aspect of im- ysis of whole-body systems physiology. BMC Syst. Biol. 5, 180.
munometabolism can help to rationally design new therapeutic Bordbar, A., Mo, M.L., Nakayasu, E.S., Schrimpe-Rutledge, A.C., Kim, Y.M.,
approaches as one would have a better understanding of the Metz, T.O., Jones, M.B., Frank, B.C., Smith, R.D., Peterson, S.N., et al.
drivers of disease versus bystander effects. In particular, target- (2012). Model-driven multi-omic data analysis elucidates metabolic immuno-
modulators of macrophage activation. Mol. Syst. Biol. 8, 558.
ing the metabolic changes that kinetically precede, and thus
likely also regulate, the phenotypic alterations in immune cells Buck, M.D., Sowell, R.T., Kaech, S.M., and Pearce, E.L. (2017). Metabolic in-
struction of immunity. Cell 169, 570–586.
could improve immune cell function and disease outcome.
Moreover, physiological immune response occurs in multicel- Cannoodt, R., Saelens, W., Sichien, D., Tavernier, S., Janssens, S., Guilliams,
lular environments that are highly organized spatially. Given M., Lambrecht, B., Preter, K.D., and Saeys, Y. (2016). SCORPIUS improves
trajectory inference and identifies novel modules in dendritic cell development.
that our current understanding of the immunometabolic regula- bioRxiv. https://doi.org/10.1101/079509.
tion is obtained using highly controlled in vitro settings (typically
Caputa, G., Castoldi, A., and Pearce, E.J. (2019). Metabolic adaptations of tis-
with purified cell types), the design of metabolic interventions for
sue-resident immune cells. Nat. Immunol. 20, 793–801.
in vivo phenotypes relies as much on chance as it does on knowl-
edge-based considerations. We envision that understanding the Caro-Maldonado, A., Wang, R., Nichols, A.G., Kuraoka, M., Milasta, S., Sun,
L.D., Gavin, A.L., Abel, E.D., Kelsoe, G., Green, D.R., and Rathmell, J.C.
fundamental principles of multicellular immunometabolic re- (2014). Metabolic reprogramming is required for antibody production that is
modeling will pave the way to deductively transition from pertur- suppressed in anergic but exaggerated in chronically BAFF-exposed B cells.
bation of key intracellular pathways toward the successful ther- J. Immunol. 192, 3626–3636.

apies on the whole-body scale. Yet it is clear that additional Chang, C.H., Curtis, J.D., Maggi, L.B., Jr., Faubert, B., Villarino, A.V., O’Sulli-
analytical tools are required to fully grasp the spatiotemporal van, D., Huang, S.C., van der Windt, G.J., Blagih, J., Qiu, J., et al. (2013). Post-
transcriptional control of T cell effector function by aerobic glycolysis. Cell 153,
aspect of immunometabolism and for tasks such as metabolic 1239–1251.
clustering on the single-cell level.
Altogether, immunometabolism is quickly becoming a sys- De Rosa, V., Galgani, M., Porcellini, A., Colamatteo, A., Santopaolo, M., Zu-
chegna, C., Romano, A., De Simone, S., Procaccini, C., La Rocca, C., et al.
tems-level science where multidisciplinary expertise is required (2015). Glycolysis controls the induction of human regulatory T cells by modu-
for making the fullest use of the novel technologies. Therefore, lating the expression of FOXP3 exon 2 splicing variants. Nat. Immunol. 16,
1174–1184.
computational advances would be important not only in terms
of data analysis itself but also in terms of their usability and inter- Dennis, E.A., Deems, R.A., Harkewicz, R., Quehenberger, O., Brown, H.A.,
pretability by experimental immunologists in order to success- Milne, S.B., Myers, D.S., Glass, C.K., Hardiman, G., Reichart, D., et al.
(2010). A mouse macrophage lipidome. J. Biol. Chem. 285, 39976–39985.
fully transit to the single-cell era of immunometabolism.
Ding, J., Adiconis, X., Simmons, S.K., Kowalczyk, M.S., Hession, C.C., Marja-
novic, N.D., Hughes, T.K., Wadsworth, M.H., Burks, T., Nguyen, L.T., et al.
ACKNOWLEDGMENTS (2020). Systematic comparison of single-cell and single-nucleus RNA-
sequencing methods. Nat. Biotechnol. 38, 737–746.
We thank Sjoerd Schetters, Menno de Winther, and Felix Hartmann for insight- Divakaruni, A.S., Hsieh, W.Y., Minarrieta, L., Duong, T.N., Kim, K.K.O., Des-
ful feedback; Sanne Verberk, Kyra de Goede, and Luc Magré for proofreading; ousa, B.R., Andreyev, A.Y., Bowman, C.E., Caradonna, K., Dranka, B.P.,
and Amanda Swain and Karl Harber for edits and comments on the manu- et al. (2018). Etomoxir inhibits macrophage polarization by disrupting CoA ho-
script. M.N.A. is supported by R01-AI125618 from National Institute of Allergy meostasis. Cell Metab. 28, 490–503.e7.
and Infectious Diseases (NIAID). J.V.d.B. received a VENI grant from ZonMW
(91615052), a Netherlands Heart Foundation Junior Postdoctoral grant Duncan, K.D., Fyrestam, J., and Lanekoff, I. (2019). Advances in mass spec-
(2013T003) and Senior Fellowship (2017T048), an NWO ENW-KLEIN-1 grant trometry based single-cell metabolomics. Analyst (Lond.) 144, 782–793.
(268), and a CCA PhD grant from Cancer Center Amsterdam.
Everts, B., Amiel, E., Huang, S.C., Smith, A.M., Chang, C.H., Lam, W.Y., Red-
mann, V., Freitas, T.C., Blagih, J., van der Windt, G.J., et al. (2014). TLR-driven
REFERENCES early glycolytic reprogramming via the kinases TBK1-IKKe supports the
anabolic demands of dendritic cell activation. Nat. Immunol. 15, 323–332.
Ahl, P.J., Hopkins, R.A., Xiang, W.W., Au, B., Kaliaperumal, N., Fairhurst, A.M., Ferrer-Font, L., Pellefigues, C., Mayer, J.U., Small, S.J., Jaimes, M.C., and
and Connolly, J.E. (2020). Met-Flow, a strategy for single-cell metabolic anal- Price, K.M. (2020). Panel design and optimization for high-dimensional immu-
ysis highlights dynamic changes in immune subpopulations. Commun Biol nophenotyping assays using spectral flow cytometry. Curr. Protoc. Cytom.
3, 305. 92, e70.

Alexandrov, T. (2020). Probing metabolism in time and space. Science 368, Galván-Peña, S., Carroll, R.G., Newman, C., Hinchy, E.C., Palsson-McDer-
241–242. mott, E., Robinson, E.K., Covarrubias, S., Nadin, A., James, A.M., Haneklaus,
M., et al. (2019). Malonylation of GAPDH is an inflammatory signal in macro-
Alves, T.C., Pongratz, R.L., Zhao, X., Yarborough, O., Sereda, S., Shirihai, O., phages. Nat. Commun. 10, 338.
Cline, G.W., Mason, G., and Kibbey, R.G. (2015). Integrated, step-wise, mass-
isotopomeric flux analysis of the TCA cycle. Cell Metab. 22, 936–947. Geiger, R., Rieckmann, J.C., Wolf, T., Basso, C., Feng, Y., Fuhrer, T., Koga-
deeva, M., Picotti, P., Meissner, F., Mann, M., et al. (2016). L-arginine modu-
Ariss, M.M., Islam, A.B.M.M.K., Critcher, M., Zappia, M.P., and Frolov, M.V. lates T cell metabolism and enhances survival and anti-tumor activity. Cell
(2018). Single cell RNA-sequencing identifies a metabolic aspect of apoptosis 167, 829–842.e13.
in Rbf mutant. Nat. Commun. 9, 5024.
Gilmore, I.S., Heiles, S., and Pieterse, C.L. (2019). Metabolic imaging at the sin-
Ayres, J.S. (2020). Immunometabolism of infections. Nat. Rev. Immunol. gle-cell scale: recent advances in mass spectrometry imaging. Annu. Rev.
20, 79–80. Anal. Chem. (Palo Alto, Calif.) 12, 201–224.

14 Cell Metabolism 32, November 3, 2020


ll
Perspective
Gubin, M.M., Esaulova, E., Ward, J.P., Malkova, O.N., Runci, D., Wong, P., No- Lam, W.Y., Becker, A.M., Kennerly, K.M., Wong, R., Curtis, J.D., Llufrio, E.M.,
guchi, T., Arthur, C.D., Meng, W., Alspach, E., et al. (2018). High-dimensional McCommis, K.S., Fahrmann, J., Pizzato, H.A., Nunley, R.M., et al. (2016). Mito-
analysis delineates myeloid and lymphoid compartment remodeling during chondrial pyruvate import promotes long-term survival of antibody-secreting
successful immune-checkpoint cancer therapy. Cell 175, 1014–1030.e19. plasma cells. Immunity 45, 60–73.

Hartmann, F.J., and Bendall, S.C. (2020). Immune monitoring using mass cy- Lampropoulou, V., Sergushichev, A., Bambouskova, M., Nair, S., Vincent,
tometry and related high-dimensional imaging approaches. Nat. Rev. Rheu- E.E., Loginicheva, E., Cervantes-Barragan, L., Ma, X., Huang, S.C., Griss, T.,
matol. 16, 87–99. et al. (2016). Itaconate links inhibition of succinate dehydrogenase with macro-
phage metabolic remodeling and regulation of inflammation. Cell Metab. 24,
Hartmann, F.J., Mrdjen, D., McCaffrey, E., Glass, D.R., Greenwald, N.F., Bhar- 158–166.
adwaj, A., Khair, Z., Verberk, S.G.S., Baranski, A., Baskar, R., et al. (2020). Sin-
gle-cell metabolic profiling of human cytotoxic T cells. Nat. Biotechnol. Pub- Levine, L.S., Hiam, K.J., Marquez, D.M., Tenvooren, I., Contreras, D.C., Rath-
lished online August 31, 2020. https://doi.org/10.1038/s41587-020-0651-8. mell, J.C., and Spitzer, M.H. (2020). Single-cell metabolic analysis by mass cy-
tometry reveals distinct transitional states of CD8 T cell differentiation. bioRxiv.
Haschemi, A., Kosma, P., Gille, L., Evans, C.R., Burant, C.F., Starkl, P., Knapp, https://doi.org/10.1101/2020.01.21.911545.
B., Haas, R., Schmid, J.A., Jandl, C., et al. (2012). The sedoheptulose kinase
CARKL directs macrophage polarization through control of glucose meta- Lim, A.R., Rathmell, W.K., and Rathmell, J.C. (2020). The tumor microenviron-
bolism. Cell Metab. 15, 813–826. ment as a metabolic barrier to effector T cells and immunotherapy. eLife 9,
e55185.
Hayashi, K., Jutabha, P., Endou, H., Sagara, H., and Anzai, N. (2013). LAT1 is a
critical transporter of essential amino acids for immune reactions in activated
Llufrio, E.M., Wang, L., Naser, F.J., and Patti, G.J. (2018). Sorting cells alters
human T cells. J. Immunol. 191, 4080–4085.
their redox state and cellular metabolome. Redox Biol. 16, 381–387.
Hsieh, W.Y., Zhou, Q.D., York, A.G., Williams, K.J., Scumpia, P.O., Kronen-
berger, E.B., Hoi, X.P., Su, B., Chi, X., Bui, V.L., et al. (2020). Toll-like receptors Ma, E.H., Bantug, G., Griss, T., Condotta, S., Johnson, R.M., Samborska, B.,
induce signal-specific reprogramming of the macrophage lipidome. Cell Mainolfi, N., Suri, V., Guak, H., Balmer, M.L., et al. (2017). Serine is an essential
Metab. 32, 128–143.e5. metabolite for effector T cell expansion. Cell Metab. 25, 345–357.

Huang, S.C., Everts, B., Ivanova, Y., O’Sullivan, D., Nascimento, M., Smith, Ma, E.H., Verway, M.J., Johnson, R.M., Roy, D.G., Steadman, M., Hayes, S.,
A.M., Beatty, W., Love-Gregory, L., Lam, W.Y., O’Neill, C.M., et al. (2014). Williams, K.S., Sheldon, R.D., Samborska, B., Kosinski, P.A., et al. (2019).
Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macro- Metabolic profiling using stable isotope tracing reveals distinct patterns of
phages. Nat. Immunol. 15, 846–855. glucose utilization by physiologically activated CD8+ T cells. Immunity 51,
856–870.e5.
Huang, S.C., Smith, A.M., Everts, B., Colonna, M., Pearce, E.L., Schilling, J.D.,
and Pearce, E.J. (2016). Metabolic reprogramming mediated by the mTORC2- Makowski, L., Chaib, M., and Rathmell, J.C. (2020). Immunometabolism: from
IRF4 signaling axis is essential for macrophage alternative activation. Immunity basic mechanisms to translation. Immunol. Rev. 295, 5–14.
45, 817–830.
Mazumdar, C., Driggers, E.M., and Turka, L.A. (2020). The untapped opportu-
Hwang, B., Lee, D.S., Tamaki, W., Sun, Y., Ogorodnikov, A., Hartoularos, G., nity and challenge of immunometabolism: a new paradigm for drug discovery.
Winters, A., Song, Y.S., Chow, E.D., Spitzer, M.H., et al. (2020). SCITO-seq: Cell Metab. 31, 26–34.
single-cell combinatorial indexed cytometry sequencing. bioRxiv. https://doi.
org/10.1101/2020.03.27.012633. Mereu, E., Lafzi, A., Moutinho, C., Ziegenhain, C., McCarthy, D.J., Álvarez-Var-
€n, D., Lau, J.K., et al. (2020). Benchmarking single-
ela, A., Batlle, E., Sagar, Gru
Jha, A.K., Huang, S.C., Sergushichev, A., Lampropoulou, V., Ivanova, Y., Log- cell RNA-sequencing protocols for cell atlas projects. Nat. Biotechnol. 38,
inicheva, E., Chmielewski, K., Stewart, K.M., Ashall, J., Everts, B., et al. (2015). 747–755.
Network integration of parallel metabolic and transcriptional data reveals
metabolic modules that regulate macrophage polarization. Immunity 42, Michalek, R.D., Gerriets, V.A., Jacobs, S.R., Macintyre, A.N., MacIver, N.J.,
419–430. Mason, E.F., Sullivan, S.A., Nichols, A.G., and Rathmell, J.C. (2011). Cutting
edge: distinct glycolytic and lipid oxidative metabolic programs are essential
Johnson, M.O., Wolf, M.M., Madden, M.Z., Andrejeva, G., Sugiura, A., Contre- for effector and regulatory CD4+ T cell subsets. J. Immunol. 186, 3299–3303.
ras, D.C., Maseda, D., Liberti, M.V., Paz, K., Kishton, R.J., et al. (2018). Distinct
regulation of Th17 and Th1 cell differentiation by glutaminase-dependent Miller, A., Nagy, C., Knapp, B., Laengle, J., Ponweiser, E., Groeger, M., Starkl,
metabolism. Cell 175, 1780–1795.e19. P., Bergmann, M., Wagner, O., and Haschemi, A. (2017). Exploring metabolic
configurations of single cells within complex tissue microenvironments. Cell
Kanehisa, M., and Goto, S. (2000). KEGG: Kyoto encyclopedia of genes and Metab. 26, 788–800.e6.
genomes. Nucleic Acids Res. 28, 27–30.
Miragaia, R.J., Gomes, T., Chomka, A., Jardine, L., Riedel, A., Hegazy, A.N.,
Kelly, B., and Pearce, E.L. (2020). Amino assets: how amino acids support im- Whibley, N., Tucci, A., Chen, X., Lindeman, I., et al. (2019). Single-cell tran-
munity. Cell Metab. 32, 154–175. scriptomics of regulatory T cells reveals trajectories of tissue adaptation. Im-
munity 50, 493–504.e7.
Keren, L., Bosse, M., Thompson, S., Risom, T., Vijayaragavan, K., McCaffrey,
E., Marquez, D., Angoshtari, R., Greenwald, N.F., Fienberg, H., et al. (2019).
Moon, J.S., Hisata, S., Park, M.A., DeNicola, G.M., Ryter, S.W., Nakahira, K.,
MIBI-TOF: a multiplexed imaging platform relates cellular phenotypes and tis-
and Choi, A.M.K. (2015). mTORC1-induced HK1-dependent glycolysis regu-
sue structure. Sci. Adv. 5, x5851.
lates NLRP3 inflammasome activation. Cell Rep. 12, 102–115.
Ketelhuth, D.F.J., Lutgens, E., Ba €ck, M., Binder, C.J., Van den Bossche, J.,
Daniel, C., Dumitriu, I.E., Hoefer, I., Libby, P., O’Neill, L., et al. (2019). Immuno- Munder, M., Eichmann, K., and Modolell, M. (1998). Alternative metabolic
metabolism and atherosclerosis: perspectives and clinical significance: a po- states in murine macrophages reflected by the nitric oxide synthase/arginase
sition paper from the Working Group on Atherosclerosis and Vascular Biology balance: competitive regulation by CD4+ T cells correlates with Th1/Th2
of the European Society of Cardiology. Cardiovasc. Res. 115, 1385–1392. phenotype. J. Immunol. 160, 5347–5354.

Kimmey, S.C., Borges, L., Baskar, R., and Bendall, S.C. (2019). Parallel anal- Nakaya, M., Xiao, Y., Zhou, X., Chang, J.H., Chang, M., Cheng, X., Blonska,
ysis of tri-molecular biosynthesis with cell identity and function in single cells. M., Lin, X., and Sun, S.C. (2014). Inflammatory T cell responses rely on amino
Nat. Commun. 10, 1185. acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase
activation. Immunity 40, 692–705.
Koelwyn, G.J., Corr, E.M., Erbay, E., and Moore, K.J. (2018). Regulation of
macrophage immunometabolism in atherosclerosis. Nat. Immunol. 19, Nomura, M., Liu, J., Rovira, I.I., Gonzalez-Hurtado, E., Lee, J., Wolfgang, M.J.,
526–537. and Finkel, T. (2016). Fatty acid oxidation in macrophage polarization. Nat. Im-
munol. 17, 216–217.
Krawczyk, C.M., Holowka, T., Sun, J., Blagih, J., Amiel, E., DeBerardinis, R.J.,
Cross, J.R., Jung, E., Thompson, C.B., Jones, R.G., and Pearce, E.J. (2010). O’Neill, L.A.J., and Artyomov, M.N. (2019). Itaconate: the poster child of meta-
Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic bolic reprogramming in macrophage function. Nat. Rev. Immunol. 19,
cell activation. Blood 115, 4742–4749. 273–281.

Cell Metabolism 32, November 3, 2020 15


ll
Perspective
O’Neill, L.A., Kishton, R.J., and Rathmell, J. (2016). A guide to immunometab- Turbitt, W.J., Buchta Rosean, C., Weber, K.S., and Norian, L.A. (2020). Obesity
olism for immunologists. Nat. Rev. Immunol. 16, 553–565. and CD8 T cell metabolism: implications for anti-tumor immunity and cancer
immunotherapy outcomes. Immunol. Rev. 295, 203–219.
Orth, J.D., Thiele, I., and Palsson, B.O. (2010). What is flux balance analysis?
Nat. Biotechnol. 28, 245–248. Van den Bossche, J., and Saraber, D.L. (2018). Metabolic regulation of macro-
phages in tissues. Cell. Immunol. 330, 54–59.
Palsson-McDermott, E.M., Curtis, A.M., Goel, G., Lauterbach, M.A., Sheedy,
F.J., Gleeson, L.E., van den Bosch, M.W., Quinn, S.R., Domingo-Fernandez, Van den Bossche, J., and van der Windt, G.J.W. (2018). Fatty acid oxidation in
R., Johnston, D.G., et al. (2015). Pyruvate kinase M2 regulates Hif-1a activity macrophages and T cells: time for reassessment? Cell Metab. 28, 538–540.
and IL-1b induction and is a critical determinant of the warburg effect in
LPS-activated macrophages. Cell Metab. 21, 65–80.
Van den Bossche, J., Lamers, W.H., Koehler, E.S., Geuns, J.M., Alhonen, L.,
Pareek, V., Tian, H., Winograd, N., and Benkovic, S.J. (2020). Metabolomics Uimari, A., Pirnes-Karhu, S., Van Overmeire, E., Morias, Y., Brys, L., et al.
and mass spectrometry imaging reveal channeled de novo purine synthesis (2012). Pivotal Advance: Arginase-1-independent polyamine production stim-
in cells. Science 368, 283–290. ulates the expression of IL-4-induced alternatively activated macrophage
markers while inhibiting LPS-induced expression of inflammatory genes.
Pelgrom, L.R., van der Ham, A.J., and Everts, B. (2016). Analysis of TLR- J. Leukoc. Biol. 91, 685–699.
induced metabolic changes in dendritic cells using the Seahorse XF(e)96
extracellular flux analyzer. Methods Mol. Biol. 1390, 273–285. Van den Bossche, J., Baardman, J., and de Winther, M.P. (2015). Metabolic
characterization of polarized M1 and M2 bone marrow-derived macrophages
Poznanski, S.M., and Ashkar, A.A. (2019). What defines NK cell functional fate: using real-time extracellular flux analysis. J. Vis. Exp. 105.
phenotype or metabolism? Front. Immunol. 10, 1414.
Van den Bossche, J., Baardman, J., Otto, N.A., van der Velden, S., Neele, A.E.,
Raud, B., Roy, D.G., Divakaruni, A.S., Tarasenko, T.N., Franke, R., Ma, E.H., van den Berg, S.M., Luque-Martin, R., Chen, H.J., Boshuizen, M.C., Ahmed,
Samborska, B., Hsieh, W.Y., Wong, A.H., Stu €ve, P., et al. (2018). Etomoxir ac- M., et al. (2016). Mitochondrial dysfunction prevents repolarization of inflam-
tions on regulatory and memory T cells are independent of Cpt1a-mediated matory macrophages. Cell Rep. 17, 684–696.
fatty acid oxidation. Cell Metab. 28, 504–515.e7.
Van den Bossche, J., O’Neill, L.A., and Menon, D. (2017). Macrophage immu-
Rodriguez, A.E., Ducker, G.S., Billingham, L.K., Martinez, C.A., Mainolfi, N., nometabolism: where are we (going)? Trends Immunol. 38, 395–406.
Suri, V., Friedman, A., Manfredi, M.G., Weinberg, S.E., Rabinowitz, J.D., and
Chandel, N.S. (2019). Serine metabolism supports macrophage IL-1b produc- van der Windt, G.J., Everts, B., Chang, C.H., Curtis, J.D., Freitas, T.C., Amiel,
tion. Cell Metab. 29, 1003–1011.e4.
E., Pearce, E.J., and Pearce, E.L. (2012). Mitochondrial respiratory capacity is
a critical regulator of CD8+ T cell memory development. Immunity 36, 68–78.
Scharping, N.E., Menk, A.V., Moreci, R.S., Whetstone, R.D., Dadey, R.E., Wat-
kins, S.C., Ferris, R.L., and Delgoffe, G.M. (2016). The tumor microenvironment
represses T cell mitochondrial biogenesis to drive intratumoral T cell metabolic van der Windt, G.J., O’Sullivan, D., Everts, B., Huang, S.C., Buck, M.D., Curtis,
insufficiency and dysfunction. Immunity 45, 374–388. J.D., Chang, C.H., Smith, A.M., Ai, T., Faubert, B., et al. (2013). CD8 memory
T cells have a bioenergetic advantage that underlies their rapid recall ability.
c
S upáková, K., Balluff, B., Tressler, C., Adelaja, T., Heeren, R.M.A., Glunde, K., Proc. Natl. Acad. Sci. USA 110, 14336–14341.
and Ertaylan, G. (2020). Cellular resolution in clinical MALDI mass spectrom-
etry imaging: the latest advancements and current challenges. Clin. Chem. van der Windt, G.J.W., Chang, C.H., and Pearce, E.L. (2016). Measuring bio-
Lab. Med. 58, 914–929. energetics in T cells using a Seahorse extracellular flux analyzer. Curr. Protoc.
Immunol. 113, 1–10, 14.
Slavov, N. (2020). Unpicking the proteome in single cells. Science 367,
512–513. Vats, D., Mukundan, L., Odegaard, J.I., Zhang, L., Smith, K.L., Morel, C.R.,
Wagner, R.A., Greaves, D.R., Murray, P.J., and Chawla, A. (2006). Oxidative
Spitzer, M.H., and Nolan, G.P. (2016). Mass cytometry: single cells, many fea- metabolism and PGC-1beta attenuate macrophage-mediated inflammation.
tures. Cell 165, 780–791. Cell Metab. 4, 13–24.
Stoeckius, M., Hafemeister, C., Stephenson, W., Houck-Loomis, B., Chatto- Wagner, A., Wang, C., DeTomaso, D., Avila-Pacheco, J., Zaghouani, S., Fess-
padhyay, P.K., Swerdlow, H., Satija, R., and Smibert, P. (2017). Simultaneous ler, J., Akama-Garren, E., Pierce, K., Ron-Harel, N., Douglas, V.P., et al. (2020).
epitope and transcriptome measurement in single cells. Nat. Methods 14, In silico modeling of metabolic state in single Th17 cells reveals novel regula-
865–868. tors of inflammation and autoimmunity. bioRxiv. https://doi.org/10.1101/2020.
01.23.912717.
Swain, A., Bambouskova, M., Kim, H., Andhey, P.S., Duncan, D., Auclair, K.,
Chubukov, V., Simons, D.M., Roddy, T.P., Stewart, K.M., and Artyomov,
M.N. (2020). Comparative evaluation of itaconate and its derivatives reveals Wang, R., Dillon, C.P., Shi, L.Z., Milasta, S., Carter, R., Finkelstein, D., McCor-
divergent inflammasome and type I interferon regulation in macrophages. mick, L.L., Fitzgerald, P., Chi, H., Munger, J., and Green, D.R. (2011). The tran-
Nat Metab 2, 594–602. scription factor Myc controls metabolic reprogramming upon T lymphocyte
activation. Immunity 35, 871–882.
Tabas, I., and Bornfeldt, K.E. (2020). Intracellular and intercellular aspects of
macrophage immunometabolism in atherosclerosis. Circ. Res. 126, Xiao, Z., Dai, Z., and Locasale, J.W. (2019). Metabolic landscape of the tumor
1209–1227. microenvironment at single cell resolution. Nat. Commun. 10, 3763.

Taguchi, K., Motohashi, H., and Yamamoto, M. (2011). Molecular mechanisms Zhang, Y., Kim, M.S., Nguyen, E., and Taylor, D.M. (2020). Modeling metabolic
of the Keap1–Nrf2 pathway in stress response and cancer evolution. Genes variation with single-cell expression data. bioRxiv. https://doi.org/10.1101/
Cells 16, 123–140. 2020.01.28.923680.

16 Cell Metabolism 32, November 3, 2020


Molecular Cell

Perspective

Single-Cell RNA Sequencing in Cancer:


Lessons Learned and Emerging Challenges
Mario L. Suvà1,2,* and Itay Tirosh3,*
1Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston,

MA 02114, USA
2Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
3Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 761001, Israel

*Correspondence: suva.mario@mgh.harvard.edu (M.L.S.), itay.tirosh@weizmann.ac.il (I.T.)


https://doi.org/10.1016/j.molcel.2019.05.003

Bulk genomic analyses and expression profiling of clinical specimens have shaped much of our understand-
ing of cancer in patients. However, human tumors are intricate ecosystems composed of diverse cells,
including malignant, immune, and stromal subsets, whose precise characterization is masked by bulk
genomic methods. Single-cell genomic techniques have emerged as powerful approaches to dissect human
tumors at the resolution of individual cells, providing a compelling approach to deciphering cancer biology.
Here, we discuss some of the common themes emerging from initial studies of single-cell RNA sequencing in
cancer and then highlight challenges in cancer biology for which emerging single-cell genomics methods
may provide a compelling approach.

Although cancer is a genetic disease, malignancies are associ- may be considered as ‘‘cell types’’ (e.g., malignant cells,
ated with aberrations in gene expression related to both cancer T cells, and fibroblasts) and further diversity within each of those
cell-intrinsic and extrinsic factors. Genomic profiling of biological clusters that may be considered as ‘‘cell states,’’ such as pro-
samples has revolutionized our understanding of cancer in pa- gression along the cell cycle, different metabolic states, and
tients. Yet most datasets and analyses of gene expression are other dynamic programs. Cell types and states may then be an-
dominated by bulk profiling, in which an entire biological sample notated based on the identity of preferentially expressed genes
(e.g., tumor) is profiled as a single entity, thus averaging the (in each cluster or subcluster) along with genetic classification,
expression profiles of the constituent cells. Expression intra-tu- including large-scale copy number alterations (CNAs), point mu-
mor heterogeneity (eITH) governs many critical facets of tumor tations, and fusion proteins, that can be inferred from scRNA-
biology that are driven by subsets of tumor cells, such as tumor seq data and help to resolve malignant from non-malignant cells
growth, metastasis, and resistance to treatment. eITH of malig- (Filbin et al., 2018; Jerby-Arnon et al., 2018; Patel et al., 2014;
nant cells is governed by at least three determinants: (1) genetic Puram et al., 2017; Tirosh et al., 2016a, 2016b; Venteicher
heterogeneity, due to subclonal mutations that arise during tu- et al., 2017). An emerging theme from recent scRNA-seq studies
mor evolution, drives diverse cellular programs; (2) epigenetic is that malignant cells tend to cluster in their expression profiles
and developmental programs, such as those of tissue stem cells primarily by patient sample, and non-malignant cells cluster in
and their differentiated progeny, ascribe cancer cells with an their expression profiles by cell type, somewhat independently
(often aberrant) cell identity and in some cases enable cellular of the patient of origin (Filbin et al., 2018; Jerby-Arnon et al.,
plasticity; and (3) extrinsic and spatial factors that determine ox- 2018; Tirosh et al., 2016a, 2016b; Venteicher et al., 2017). This
ygen, nutrient availability, and cell-cell interactions. Single-cell indicates that (1) inter-tumor heterogeneity is typically larger for
RNA sequencing (scRNA-seq) techniques measure the tran- malignant cells than for any particular type of non-malignant
scriptional output of cells directly in tumor samples; the cells. (2) For malignant cells, inter-tumor heterogeneity is much
measured cellular programs represent an integration of its ge- larger than intra-tumor heterogeneity. These results highlight
netic, epigenetic, and environmental cues and determine cancer the considerable degree of inter-tumor heterogeneity and might
cells fitness, behavior, and response to therapies. As such, be misinterpreted as reflecting a limited role of intra-tumor het-
scRNA-seq studies place cellular biology at the center of cancer erogeneity of cancer cells. However, the power of single-cell
biology and offer both different resolution and perspective to methods is that they now further allow us to interrogate the pat-
bulk genomic studies. We have recently reviewed initial studies terns of intra-tumor heterogeneity, which have been difficult to
of single-cell expression profiling in human cancer (Tirosh and assess with previous approaches and are often continuous
Suvà, 2019). Here, we focus on emerging themes and on future rather than discrete.
challenges for the field.
Recurrent Patterns of eITH among Cancer Cells
Cell Types, Cell States, and Malignant and Non- What then are the patterns of heterogeneity observed among the
malignant Cells malignant cells in an individual tumor? First of all, the level of any
Analysis of the expression diversity within tumors (or other tis- individual gene varies between cells, partially due to the stochas-
sues) typically reveals two layers: highly distinct clusters that tic and noisy nature of gene expression regulation. In addition to

Molecular Cell 75, July 11, 2019 ª 2019 Elsevier Inc. 7


Molecular Cell

Perspective

Figure 1. Recurrent Patterns of eITH among Malignant Cells


(A–D) IDH mutant glioma (A; Tirosh et al., 2016b; Venteicher et al., 2017), H3-K27M glioma (B; Filbin et al., 2018), melanoma (C; Tirosh et al., 2016a), and head and
neck cancer (D; Puram et al., 2017). Images represent a scatter-plot in which cells (dots) are scored by the expression of particular programs identified in that
cancer type (x and y axes). In each case, a similar pattern of eITH was identified across multiple patients. Plots were either reproduced (A–C) or generated from the
data (D) of the original publication. Also highlighted in red are the associations between certain subpopulations and important clinical features, as described in the
main text and the original publications.

this stochasticity, however, sets of genes vary coherently be- 2017). In glioblastoma, IDH mutant gliomas, and histone H3-
tween cells, reflecting different cellular states that might have a K27M mutant gliomas, eITH patterns were dominated by glial
more significant biological implication. For example, concomi- differentiation and neurodevelopmental programs. These pro-
tant upregulation of dozens of cell-cycle-related genes indicates grams highlighted putative differentiation hierarchies, with
that certain cells have entered the cell cycle and others were not stem-like cells that were linked to self-renewal and increased
cycling at the moment in which they were profiled (Tirosh et al., aggressiveness (Filbin et al., 2018; Patel et al., 2014; Tirosh
2016a, 2016b). Similarly, concomitant upregulation of many et al., 2016b; Venteicher et al., 2017).
genes associated with hypoxia and/or stress responses indicate These initial results suggest two important conclusions. First,
that certain cells respond to their immediate microenvironment the patterns of eITH are of considerable significance for the
and to the lack of nutrients or oxygen by mounting such re- maintenance and progression of tumors, warranting further anal-
sponses (Patel et al., 2014; Tirosh et al., 2016a, 2016b). These ysis and future approaches to target particular tumor subpopu-
two processes—cell cycle and stress responses—reflect com- lations. Second, the consistency of these patterns across pa-
mon patterns of cellular heterogeneity within tumors, which are tients of the same cancer type or subtype suggests that these
observed within different patients and across different can- reflect fundamental properties of the corresponding cells from
cer types. which the cancer originates. For example, gliomas likely origi-
In addition to those ‘‘generic’’ patterns of heterogeneity, there nate from neural stem-like cells, which have an intrinsic capacity
are also patterns that are context specific and hence may be to differentiate toward astrocytes and oligodendrocytes; thus,
more informative about the specific biology of a given tumor the resulting cancer cells display an aberrant differentiation to-
type. Multiple previous studies found that apart from cell cycle ward astrocyte-like and oligodendrocyte-like cells that dominate
and stress there are particular intra-tumor expression programs the cellular diversity within gliomas. Similarly, epithelial cells
(i.e., sets of genes that coherently vary among malignant cells in have the intrinsic capacity to activate mesenchymal programs
the same tumor) that are found within multiple patients of a that might normally be required during development and wound
particular cancer type, but not in other cancer types (Figure 1; healing; thus, the resulting cancer cells may also activate those
Tirosh and Suvà, 2019). Strikingly, in each of those cases, these programs in response to cues, such as transforming growth fac-
were large-scale expression programs that were uncovered by tor b (TGF-b), produced by surrounding fibroblasts.
unbiased analysis, and examination of the associated genes Thus, these patterns might be thought of as reflecting the
suggested that they impact on crucial aspects of tumor progres- ‘‘epigenetic landscape’’ of the cancer cells: only particular pro-
sion, including drug resistance, metastasis, and self-renewal. In grams may be accessible to cells in each cancer type, based
melanoma, those programs highlighted distinct MITF and AXL on the nature of the cell of origin or the impact of oncogenic
expression levels of cells within the same tumor, with suggested transformation. Accordingly, cells may dynamically activate
implications for resistance to BRAF inhibition (Rambow et al., those ‘‘accessible’’ programs, resulting in their variability within
2018; Shaffer et al., 2017; Tirosh et al., 2016a). More recently, an individual tumor. This model suggests that it will be important
a cancer cell state in melanoma that promotes immune evasion to define such programs of variability in each tumor type (or sub-
and resistance to immune checkpoint inhibitors was identified by type) and subsequently examine its impact on the functional
scRNA-seq analysis of 33 melanoma samples (Jerby-Arnon properties of the cancer cells and their vulnerabilities.
et al., 2018). In head and neck cancer, malignant cells differed
in their expression of epithelial differentiation programs and in Integrating Bulk with Single-Cell Tumor Profiles
the expression of epithelial-to-mesenchymal transition pro- These initial studies provide a proof of concept for the utility of
grams, with important implications for metastasis (Puram et al., single-cell expression profiling of tumors. However, it is also

8 Molecular Cell 75, July 11, 2019


Molecular Cell

Perspective
Figure 2. Questions in Cancer Biology
recurrence rare tumor
and MRD subpopulations Addressed by Emerging Directions with
scRNA-Seq
See main text for further description of each of
3 4 the biological questions and the emerging ap-
frozen
samples increased
throughput proaches to address it. MRD, minimal residual
disease.

microenvironmental cues cell-of-origin


and cell-cell interactions and dev. pathways

2 spatial 5
profiling HCA sion by the cancer cells would need to
be contrasted with the alternative option
scRNA-seq of abundant ECM-producing fibroblasts.
tumor profiling More generally, any subpopulation of
cells that is identified within tumors
through single-cell approaches could
multi-omics large cohorts
model systems then be investigated for its estimated
abundance and associations across
large cohorts of bulk samples, thereby
1 N-of-1
studies
partially circumventing the limited size
genetic vs. 6 of single-cell cohorts.
non-genetic significance of
mechanisms subpopulations
Emerging Challenges and
7 exceptional Directions in Single-Cell Cancer
responders Analysis
In the next few years, the approach of
investigating tumors through single-cell
important to realize the limitations of this approach. First, RNA-seq will be further integrated into cancer research and
scRNA-seq provides only a partial sampling of the transcriptome will be used to address an increasing number of questions about
of cells, with a bias for more highly expressed genes over mid-to- tumor biology (Figure 2). Below, we briefly describe some of the
lowly expressed ones. Second, scRNA-seq is very sensitive to main questions that are likely to be addressed and point to
sample quality and as such is not suited for the profiling of emerging technologies in the area of scRNA-seq that would
sub-optimally preserved or handled clinical specimen. Third, facilitate these studies.
high cost limits the ability to profile large cohorts of tumors,
and only few to a few dozens of samples are typically profiled Distinguishing the Contributions of Genetic versus Non-
in each study. Given these challenges with single-cell ap- genetic Mechanisms: Single-Cell Multi-omics Analyses
proaches and the comprehensive bulk databases that already Genetic heterogeneity is a prominent mechanism that generates
exist, such as The Cancer Genome Atlas (TCGA), we anticipate diversity between and within tumors. However, an increasing
that single-cell datasets will not supplant bulk tumor profiling amount of evidence points to the significance of non-genetic
but instead will be integrated with existing and new bulk profiles mechanisms. For example, in the context of eITH, we have
to advance our understanding of tumor biology. recently shown that cellular hierarchies in IDH mutant and his-
In the past, bulk profiles were analyzed in a somewhat naive tone H3-K27M mutant glioma appear to be largely independent
way, ignoring the fact that each bulk profile reflects a composite of genetics (Filbin et al., 2018; Tirosh et al., 2016b; Venteicher
of many distinct populations and sometimes misinterpreting sig- et al., 2017). Thus, an important challenge is to map both cell
nals of the tumor microenvironment as those of the cancer cells. state and genetic state in the same cells and begin to distinguish
Single-cell genomics, along with advances in cancer immuno- between these mechanisms. Notably, therapeutic approaches
therapies, have emphasized the significance of considering not for dealing with genetic versus non-genetic ITH may be quite
just the cancer cells but also the various non-cancer populations. distinct, underscoring the significance of this question. To
As a result, many recent methods and studies have focused on date, most scRNA-seq studies either ignored the genetic states
inferring the composition of tumors through deconvolution of of cells or had a partial capacity to evaluate genetic states, by (1)
their bulk profiles (Aran et al., 2017; Newman et al., 2015; Puram detecting mutations in the RNA with limited sensitivity, (2) tar-
et al., 2017; Tirosh et al., 2016a). Such methods typically require geted amplification of individual mutations, or (3) inferring
a prior definition of potential tumor components (i.e., cell types), CNAs (Filbin et al., 2018; Giustacchini et al., 2017; Tirosh and
which can now be derived directly and accurately from scRNA- Suvà, 2019). However, multiple technologies are being devel-
seq studies. In the next few years, we expect this trend to oped (Macaulay et al., 2015, 2016, 2017) and improved to enable
expand and that it will become standard to analyze bulk profiles such joint profiling of cell state and genetics, and these are ex-
in light of their measured or inferred composition. For example, if pected to provide a more integrated view in the future (Nam
certain tumors have high expression of extracellular matrix et al., 2018; Rodriguez-Meira et al., 2019; Velten et al., 2018).
(ECM) genes, the naive possibility of an aberrant ECM expres- For example, a recent study in acute myelogenous leukemia

Molecular Cell 75, July 11, 2019 9


Molecular Cell

Perspective

(AML) highlighted that cell type composition correlated with pro- for scRNA-seq, creating a severe bottleneck for processing of
totypic genetic alterations (van Galen et al., 2019). More gener- matched primary and recurrent samples given the time until
ally, ‘‘multi-OMICs’’ approaches are being developed to recurrence. However, multiple approaches may enable profiling
combine the measurement of mRNA levels with various other of frozen samples and thereby open the door to characterizing
features, such as protein levels, DNA methylation, chromatin collections of samples that are stored in hospitals and labs,
accessibility, clonal expansion, etc. These approaches will pro- including longitudinal cohorts. For example, frozen or fixed sam-
vide the ability to identify mechanisms that generate eITH and ples may be analyzed by isolation and sequencing of single
advance scRNA-seq toward more detailed mechanisms. nuclei (instead of single cells), as recently demonstrated (Gao
et al., 2017; Habib et al., 2016, 2017). In addition to recurrent tu-
Cell-Cell Interactions and Spatial Location: Spatially mors, scRNA-seq provides a compelling approach to directly
Resolved Single-Cell Technologies characterize the rare residual cells that survive through treat-
Each tumor is analogous to an ecosystem, and thus, the state of ments and ultimately underlie tumor recurrence. Such rare cells,
each cell may be highly dependent on its exact spatial location typically referred to as minimal residual disease (MRD), may be
and interaction with adjacent cells. These complex interactions profiled in animal models (Rambow et al., 2018) and compared
and their impact on cancer cell biology remain poorly under- to both primary and recurrent tumors, providing additional in-
stood. Coupling transcriptomes with spatial information will sights to their capacity to evade treatments, remain dormant,
dramatically improve the ability to predict and further charac- and finally give rise to recurrent tumors.
terize such interactions, and accordingly, many approaches
are being developed to couple scRNA-seq with spatial informa- Tumor Subpopulations: Their Detection, Functional
tion. To date, most scRNA-seq studies have measured the state Significance, and Origin
of cells without preserving any information on their location. Tumor progression is an evolutionary process, and hence, only
Other studies have measured or inferred the spatial location of few cells are sufficient for a new phenotype to gradually emerge
cells but typically characterized the cells by either multiplexed through selection. Consequently, critical subpopulations, such
markers (Chen et al., 2015) or in lower (non-cellular) resolution as cancer stem cells, drug-resistant cells, and migratory cells
(Chen et al., 2017) or relied on a stereotypical tissue structure, that underlie metastasis, might reflect a very rare subpopulation
which is a more effective strategy for normal than for tumor tis- (e.g., less than 0.1%) that escapes detection by most methods,
sues (Halpern et al., 2017; Satija et al., 2015). Continuous im- including current scRNA-seq approaches. Importantly, although
provements in spatially resolved technologies for tumor profiling, previous scRNA-seq studies were performed with platforms that
as well as the integration of data from disparate technologies, measure limited cell numbers (e.g., tens to hundreds), more
will dramatically advance these directions over the next few recent and ongoing studies now leverage droplet microfluidics
years and improve our understanding of cell-cell interactions or other technologies (Gierahn et al., 2017; Jerby- Arnon et al.,
and spatial effects. 2018; Cao et al., 2017) that enable an order of magnitude
Apart from spatial profiling, the study of cell-cell interactions more cells in each batch (e.g., thousands), and future technolo-
within tumors will advance through increased focus on the gies are likely to further expand this capacity into tens of thou-
expression of ligands and receptors as central mediators of sands to even millions of cells, which would help uncover very
cellular communication. Databases of ligand-receptor com- rare subpopulations.
plexes, coupled with a definition of tumor cell subpopulations Once unique subpopulations have been uncovered, their func-
by scRNA-seq data, highlight potential cell-cell interactions, in tional and clinical significance remains difficult to ascertain. Two
which one population produces a ligand that signals to another main approaches enable further analysis of such subpopula-
population expressing the corresponding receptor (Puram tions. First, studies of large cohorts would uncover statistical as-
et al., 2017; Vento-Tormo et al., 2018). The complexity of the tu- sociations between the presence and/or frequency of subpopu-
mor microenvironment implies that the number of such potential lations and clinical features, such as survival, metastasis, and
interactions would be large, warranting follow-up studies that will drug responses. Given the cost and labor associated with
determine the significance of individual interactions. scRNA-seq of large cohorts, we anticipate that many of these
analyses will leverage available bulk RNA-seq datasets and
Residual Disease and Recurrences: Leveraging Single- use computational approaches for their deconvolution and the
Nucleus Profiling (Frozen) inference of tumor composition, as described above. However,
In many cancer types, initial therapeutic responses are typically whether these would be based on single-cell or bulk RNA-seq
followed by the recurrence of tumors with increased aggressive- profiles, such correlative studies will require mechanistic
ness and drug resistance, highlighting the significance of both follow-ups. Thus, a second approach will rely on the detailed
the residual disease and its evolution and growth toward estab- characterization of such subpopulations by scRNA-seq to iden-
lishing the recurrent tumors. Previous studies compared primary tify markers that would enable isolation of these subpopulations
and recurrent tumors and revealed various genetic causes of and further functional studies in animal and culture models.
drug resistance. Future studies will extend this approach to com- As described above, recent scRNA-seq studies uncovered
parison by scRNA-seq and provide a more detailed view of remarkable similarities between tumor expression profiles and
recurrent tumors and their difference from the corresponding pri- those of normal developmental cell types, raising specific hy-
mary tumors (Brady et al., 2017; Kim et al., 2018). An important potheses about the origin of cancers and the ongoing differenti-
challenge is that current protocols require fresh tumor samples ation processes that occur within them. Through extensive

10 Molecular Cell 75, July 11, 2019


Molecular Cell

Perspective

scRNA-seq of tumors, of normal tissues, and of developmental of human cancer; they are paving the way for many more discov-
processes, for example, through the human cell atlas (HCA) eries that will come from improved technologies, resolution,
initiative, the number of such observations is expected to computational methods, and broader clinical settings.
dramatically increase over the next few years, including refine-
ment of many previous observations. For example, scRNA-seq ACKNOWLEDGMENTS
of H3-K27M midline gliomas (Filbin et al., 2018) highlighted the
similarity of malignant cells to oligodendrocytic precursor cells We thank Ania Hupalowska for help with graphic design. This work was sup-
ported by grants from the Howard Goodman Fellowship at MGH (M.L.S.),
(OPCs), supporting previous literature that suggested that the Merkin Institute Fellowship at the Broad Institute of MIT and Harvard
OPCs are a candidate cell of origin for this tumor type (Gibson (M.L.S.), the Sontag Foundation (M.L.S.), the Zuckerman STEM Leadership
et al., 2018). A caveat with these inferences is that scRNA-seq Program (I.T.), the Human Frontiers Science Program (I.T.), the Mexican
Friends New Generation (I.T.), the Benoziyo Endowment Fund for the
of an established tumor cannot identify the cell in which the mu-
Advancement of Science (I.T.), and start-up funds from the MGH Department
tation first occurred (‘‘cell of mutation’’), which can be distinct of Pathology (M.L.S.) and the Weizmann Institute of Science (I.T.). I.T. is an
from the cell that gives rise to the tumor. Additionally, compari- incumbent of the Dr. Celia Zwillenberg-Fridman and Dr. Lutz Zwillenberg
sons between malignant cells and normal cells not only highlight Career Development Chair.
similarities but also provide a more detailed view of the differ-
REFERENCES
ences between tumor cells and their presumed cells of origin
and thereby increase our understanding of the oncogenic
Aran, D., Hu, Z., and Butte, A.J. (2017). xCell: digitally portraying the tissue
process. cellular heterogeneity landscape. Genome Biol. 18, 220.

Brady, S.W., McQuerry, J.A., Qiao, Y., Piccolo, S.R., Shrestha, G., Jenkins,
Unique Tumor Phenotypes and Exceptional D.F., Layer, R.M., Pedersen, B.S., Miller, R.H., Esch, A., et al. (2017).
Responders: Single-Cell Analysis in N = 1 Cohorts Combating subclonal evolution of resistant cancer phenotypes. Nat. Commun.
Although therapeutic decisions are based on statistical patterns 8, 1231.

in large cohorts, outliers are quite common and present impor- Cao, J., Packer, J.S., Ramani, V., Cusanovich, D.A., Huynh, C., Daza, R., Qiu,
tant conundrums. For example, ‘‘exceptional responders’’ illus- X., Lee, C., Furlan, S.N., Steemers, F.J., et al. (2017). Comprehensive single-
cell transcriptional profiling of a multicellular organism. Science 357, 661–667.
trate our limited understanding of treatment response and the
theoretical possibility to improve patient stratifications (Ferreri Chen, K.H., Boettiger, A.N., Moffitt, J.R., Wang, S., and Zhuang, X. (2015). RNA
imaging. Spatially resolved, highly multiplexed RNA profiling in single cells.
et al., 2010). In the past, it has been difficult to understand the
Science 348, aaa6090.
unique phenotypes of such cases, which were studied primarily
by bulk genetic analysis. However, scRNA-seq, along with the Chen, J., Suo, S., Tam, P.P., Han, J.J., Peng, G., and Jing, N. (2017). Spatial
transcriptomic analysis of cryosectioned tissue samples with Geo-seq. Nat.
associated technological improvements described above (e.g., Protoc. 12, 566–580.
spatial profiling and multi-OMIC techniques), enable character-
Ferreri, A.J., Illerhaus, G., Zucca, E., and Cavalli, F.; International Extranodal
ization of tumor ecosystems at unprecedented depth and Lymphoma Study Group (2010). Flows and flaws in primary central nervous
breadth. We propose that these would be ideal to begin to iden- system lymphoma. Nat. Rev. Clin. Oncol. 7, 472.
tify the basis for unique phenotypes and accordingly that N-of-1
Filbin, M.G., Tirosh, I., Hovestadt, V., Shaw, M.L., Escalante, L.E., Mathewson,
scRNA-seq studies may be particularly informative. Because N.D., Neftel, C., Frank, N., Pelton, K., Hebert, C.M., et al. (2018). Develop-
extreme phenotypes are often detected late, this approach will mental and oncogenic programs in H3K27M gliomas dissected by single-
cell RNA-seq. Science 360, 331–335.
also likely depend on analysis of frozen samples that have
been archived. Gao, R., Kim, C., Sei, E., Foukakis, T., Crosetto, N., Chan, L.K., Srinivasan, M.,
Zhang, H., Meric-Bernstam, F., and Navin, N. (2017). Nanogrid single-nucleus
RNA sequencing reveals phenotypic diversity in breast cancer. Nat. Commun.
Concluding Remarks 8, 228.
Tumor heterogeneity has puzzled cancer biologists and clini-
Gibson, E.M., Geraghty, A.C., and Monje, M. (2018). Bad wrap: myelin and
cians for a long time. As genomic tools were lacking to accu- myelin plasticity in health and disease. Dev. Neurobiol. 78, 123–135.
rately measure individual cells in a tumor, we relied on average
measurements or single markers, masking many critical aspects Gierahn, T.M., Wadsworth, M.H., 2nd, Hughes, T.K., Bryson, B.D., Butler, A.,
Satija, R., Fortune, S., Love, J.C., and Shalek, A.K. (2017). Seq-Well: portable,
of ITH and hindering our understanding of tumor biology. The low-cost RNA sequencing of single cells at high throughput. Nat. Methods 14,
precise characterization of single-cell programs in tumors places 395–398.
cellular biology at the center of cancer biology. By measuring the Giustacchini, A., Thongjuea, S., Barkas, N., Woll, P.S., Povinelli, B.J., Booth,
transcriptional output as a function of both cell type and ge- C.A.G., Sopp, P., Norfo, R., Rodriguez-Meira, A., Ashley, N., et al. (2017). Sin-
netics, these efforts lay the foundation for renewed understand- gle-cell transcriptomics uncovers distinct molecular signatures of stem cells in
chronic myeloid leukemia. Nat. Med. 23, 692–702.
ing of cancer initiation and evolution. The comprehensive
profiling of single-cell programs in cancer also sheds light on tu- Habib, N., Li, Y., Heidenreich, M., Swiech, L., Avraham-Davidi, I., Trombetta,
J.J., Hession, C., Zhang, F., and Regev, A. (2016). Div-seq: single-nucleus
mor classification based on bulk measurements by enabling the RNA-seq reveals dynamics of rare adult newborn neurons. Science 353,
deconvolution of signal and highlighting unanticipated patterns 925–928.
of ITH. Although the increased understanding of ITH might at first
Habib, N., Avraham-Davidi, I., Basu, A., Burks, T., Shekhar, K., Hofree, M.,
seem daunting, common cellular states and programs are Choudhury, S.R., Aguet, F., Gelfand, E., Ardlie, K., et al. (2017). Massively par-
emerging and are offering new candidate vulnerabilities that allel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958.
could be exploited for therapeutics. The initial studies discussed Halpern, K.B., Shenhav, R., Matcovitch-Natan, O., Toth, B., Lemze, D., Go-
here are the beginning of an exciting era of single-cell genomics lan, M., Massasa, E.E., Baydatch, S., Landen, S., Moor, A.E., et al. (2017).

Molecular Cell 75, July 11, 2019 11


Molecular Cell

Perspective
Single-cell spatial reconstruction reveals global division of labour in the Rodriguez-Meira, A., Buck, G., Clark, S.-A., Povinelli, B.J., Alcolea, V., Louka,
mammalian liver. Nature 542, 352–356. E., McGowan, S., Hamblin, A., Sousos, N., Barkas, N., et al. (2019). Unraveling
intratumoral heterogeneity through high-sensitivity single-cell mutational anal-
Jerby-Arnon, L., Shah, P., Cuoco, M.S., Rodman, C., Su, M.J., Melms, J.C., ysis and parallel RNA sequencing. Mol. Cell 73, 1292–1305.E8.
Leeson, R., Kanodia, A., Mei, S., Lin, J.R., et al. (2018). A cancer cell program
promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, Satija, R., Farrell, J.A., Gennert, D., Schier, A.F., and Regev, A. (2015). Spatial
984–997.e24. reconstruction of single-cell gene expression data. Nat. Biotechnol 33,
495–502.
Kim, C., Gao, R., Sei, E., Brandt, R., Hartman, J., Hatschek, T., Crosetto, N.,
Foukakis, T., and Navin, N.E. (2018). Chemoresistance evolution in triple-
negative breast cancer delineated by single-cell sequencing. Cell 173, 879– Shaffer, S.M., Dunagin, M.C., Torborg, S.R., Torre, E.A., Emert, B., Krepler, C.,
893.e13. Beqiri, M., Sproesser, K., Brafford, P.A., Xiao, M., et al. (2017). Rare cell vari-
ability and drug-induced reprogramming as a mode of cancer drug resistance.
Macaulay, I.C., Haerty, W., Kumar, P., Li, Y.I., Hu, T.X., Teng, M.J., Goolam, Nature 546, 431–435.
M., Saurat, N., Coupland, P., Shirley, L.M., et al. (2015). G&T-seq: parallel
sequencing of single-cell genomes and transcriptomes. Nat. Methods 12, Tirosh, I., and Suvà, M.L. (2019). Deciphering Human Tumor Biology by Single-
519–522. Cell Expression Profiling. Annu. Rev. Cancer Biol 3, 151–166.

Macaulay, I.C., Teng, M.J., Haerty, W., Kumar, P., Ponting, C.P., and Voet, T. Tirosh, I., Izar, B., Prakadan, S.M., Wadsworth, M.H., 2nd, Treacy, D., Trom-
(2016). Separation and parallel sequencing of the genomes and transcrip- betta, J.J., Rotem, A., Rodman, C., Lian, C., Murphy, G., et al. (2016a). Dissect-
tomes of single cells using G&T-seq. Nat. Protoc 11, 2081–2103. ing the multicellular ecosystem of metastatic melanoma by single-cell RNA-
seq. Science 352, 189–196.
Macaulay, I.C., Ponting, C.P., and Voet, T. (2017). Single-cell multiomics: mul-
tiple measurements from single cells. Trends Genet. 33, 155–168.
Tirosh, I., Venteicher, A.S., Hebert, C., Escalante, L.E., Patel, A.P., Yizhak, K.,
Nam, A.S., Kim, K.-T., Chaligne, R., Izzo, F., Ang, C., Abu-Zeinah, G., Omans, Fisher, J.M., Rodman, C., Mount, C., Filbin, M.G., et al. (2016b). Single-cell
N.D., Taylor, J., Pastore, A., Alonso, A., et al. (2018). High throughput droplet RNA-seq supports a developmental hierarchy in human oligodendroglioma.
single-cell Genotyping of Transcriptomes (GoT) reveals the cell identity depen- Nature 539, 309–313.
dency of the impact of somatic mutations. bioRxiv. https://doi.org/10.1101/
444687. van Galen, P., Hovestadt, V., Wadsworth Ii, M.H., Hughes, T.K., Griffin, G.K.,
Battaglia, S., Verga, J.A., Stephansky, J., Pastika, T.J., Lombardi Story, J.,
Newman, A.M., Liu, C.L., Green, M.R., Gentles, A.J., Feng, W., Xu, Y., Hoang, et al. (2019). Single-cell RNA-seq reveals AML hierarchies relevant to disease
C.D., Diehn, M., and Alizadeh, A.A. (2015). Robust enumeration of cell subsets progression and immunity. Cell 176, 1265–1281.e24.
from tissue expression profiles. Nat. Methods 12, 453–457.
Velten, L., Story, B.A., Hernandez-Malmierca, P., Milbank, J., Paulsen, M.,
Patel, A.P., Tirosh, I., Trombetta, J.J., Shalek, A.K., Gillespie, S.M., Wakimoto, Lutz, C., Nowak, D., Jann, J.-C., Pabst, C., Boch, T., et al. (2018). MutaSeq re-
H., Cahill, D.P., Nahed, B.V., Curry, W.T., Martuza, R.L., et al. (2014). Single- veals the transcriptomic consequences of clonal evolution in acute myeloid
cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. leukemia. bioRxiv. https://doi.org/10.1101/500108.
Science 344, 1396–1401.

Puram, S.V., Tirosh, I., Parikh, A.S., Patel, A.P., Yizhak, K., Gillespie, S., Venteicher, A.S., Tirosh, I., Hebert, C., Yizhak, K., Neftel, C., Filbin, M.G., Hov-
Rodman, C., Luo, C.L., Mroz, E.A., Emerick, K.S., et al. (2017). Single-cell tran- estadt, V., Escalante, L.E., Shaw, M.L., Rodman, C., et al. (2017). Decoupling
scriptomic analysis of primary and metastatic tumor ecosystems in head and genetics, lineages, and microenvironment in IDH-mutant gliomas by single-
neck cancer. Cell 171, 1611–1624.e24. cell RNA-seq. Science 355, eaai8478.

Rambow, F., Rogiers, A., Marin-Bejar, O., Aibar, S., Femel, J., Dewaele, M., Vento-Tormo, R., Efremova, M., Botting, R.A., Turco, M.Y., Vento-Tormo, M.,
Karras, P., Brown, D., Chang, Y.H., Debiec-Rychter, M., et al. (2018). Toward Meyer, K.B., Park, J.E., Stephenson, E., Polanski, K., Goncalves, A., et al.
minimal residual disease-directed therapy in melanoma. Cell 174, 843– (2018). Single-cell reconstruction of the early maternal-fetal interface in
855.e19. humans. Nature 563, 347–353.

12 Molecular Cell 75, July 11, 2019


Trends in Cell Biology

Review

Subcellular Chemical Imaging: New Avenues in


Cell Biology
Johan Decelle,1,* Giulia Veronesi,2,3 Benoit Gallet,4 Hryhoriy Stryhanyuk,5 Pietro Benettoni,5 Matthias Schmidt,5
Rémi Tucoulou,3 Melissa Passarelli,6 Sylvain Bohic,3,7 Peta Clode,8,9 and Niculina Musat5

To better understand the physiology and acclimation capability of the cell, one of Highlights
the great challenges of the future is to access the interior of a cell and unveil its Chemical imaging instruments have
chemical landscape (composition and distribution of elements and molecules). substantially improved in sensitivity
and spatial resolution and have become
Chemical imaging has greatly improved in sensitivity and spatial resolution to
the method of choice to visualize and
visualize and quantify nutrients, metabolites, toxic elements, and drugs in single quantify elements, isotopes, and mole-
cells at the subcellular level. This review aims to present the current potential of cules at the subcellular level in various
these emerging imaging technologies and to guide biologists towards a strategy disciplines, such as cell biology, eco-
physiology, and toxicology.
for interrogating biological processes at the nanoscale. We also describe various
solutions to combine multiple imaging techniques in a correlative way and pro- Since chemical imaging provides low
vide perspectives and future directions for integrative subcellular imaging across contrast, it is often combined with elec-
different disciplines. tron microscopy in a correlative way to
unambiguously identify the localization
of an element or molecule in the cell.
New Avenues for the Subcellular Exploration of the Cell
The advent of electron microscopy (EM) in the mid-20th century was a formidable tool for the de- Different complementary chemical imag-
ing instruments (e.g., SIMS, X-ray fluo-
tailed exploration of a cell’s structure at nanoscale resolution. Nowadays, a key challenge in cell
rescence) can be combined to correlate
biology is to understand the activity and function of organelles and cellular compartments and elemental, isotopic, and molecular infor-
their role in the metabolism and physiology of a cell. Omics bulk analyses (e.g., transcriptomics, mation from a region of the cell or from
metabolomics) have greatly improved our understanding of cellular mechanisms but provide the entire cell in two or three dimensions.
only averaged information on extracted molecules from numerous lysed cells. Hence, spatial
New sample preparation methods and
information at the subcellular level is a missing dimension to fully interpret the phenotypic state analytical instruments enable indirect or
of a cell and assess heterogeneity in a population. Chemical imaging (see Glossary) techniques direct correlative subcellular imaging
are now able to reveal the chemical landscape of cells (i.e., the composition and distribution of from cells in their close-to-native state.

elements and molecules) at the subcellular level without the need to add or genetically encode
fluorescent labels. Probing the elemental and molecular composition of organelles and subcellular
structures can reveal fundamental information about the function and physiology of a cell in re- 1
Cell and Plant Physiology Laboratory,
sponse to different conditions. The subcellular distribution of some elements (e.g., the macronu- Université Grenoble Alpes, CNRS, CEA,
INRAE, IRIG, 38000, Grenoble, France
trients N, P, and S), which are essential building blocks of biomolecules (e.g., DNA, proteins, 2
Chemistry and Biology of Metals
lipids), can reflect the metabolic roles and needs of organelles [1]. Trace metals (e.g., Fe, Cu, Laboratory, Université Grenoble Alpes,
Zn) play a fundamental role in various biochemical functions of the cell and their homeostasis CNRS, CEA, IRIG, Grenoble, France
3
ESRF – The European Synchrotron,
and compartmentalization need to be tightly controlled to avoid cell death and severe patholo- Grenoble, France
gies. More particularly, metals are key players in parasitic and viral infections, cancer cells, and 4
Institut de Biologie Structurale,
neurodegenerative diseases [2]. In the biomedical field, the increasing human exposure to Université Grenoble Alpes, CNRS, CEA,
Grenoble, France
exogenous compounds (e.g., metal-based nanoparticles, toxic elements) and the use of thera- 5
Helmholtz Centre for Environmental
peutic drugs call for imaging techniques to visualize their fate in tissues and cells and to assess Research – UFZ, Department of Isotope
their toxicity and impact on the homeostasis of native elements [3,4]. In addition to elements, Biogeochemistry, Leipzig, Germany
6
École Polytechnique Fédérale de
the localization of metabolites (e.g., sugars, lipids) in cells is essential to fully understand metabolic Lausanne (EPFL), Laboratory for
processes. Therefore, subcellular mapping of elements and metabolites is becoming indispensable Biological Geochemistry,
to investigate the physiology and metabolism of healthy and diseased cell types, to understand Lausanne, Switzerland
7
INSERM – UA7 – Synchrotron Radiation
cellular interactions in tissues or with beneficial cells (e.g., symbioses) and pathogens (e.g., viral for Biomedicine, STROBE, Université
or bacterial infection), and their adaptive response to abiotic stresses. Grenoble Alpes, Grenoble, France

Trends in Cell Biology, March 2020, Vol. 30, No. 3 https://doi.org/10.1016/j.tcb.2019.12.007 173
© 2020 Elsevier Ltd. All rights reserved.
Trends in Cell Biology

8
Recent technological progress in chemical imaging has substantially improved the sensitivity and Centre for Microscopy Characterisation
and Analysis, The University of Western
spatial resolution, allowing the disentangling of cellular compartments in a single cell. However, Australia, Crawley, WA, Australia
9
the multiplicity of these complex imaging techniques requires guidance for nonspecialists. An UWA School of Biological Sciences, The
overview of the chemical imaging techniques currently available is therefore needed to help University of Western Australia, Crawley,
WA, Australia
biologists integrate the subcellular scale in their studies while being aware of its potential and
limitations. Each chemical imaging platform presents experimental specifications that make
them more sensitive to some elements or molecules, so different platforms need to be combined *Correspondence:
to have a comprehensive view of the chemical landscape of a cell. Moreover, since chemical johan.decelle@univ-grenoble-alpes.fr
imaging generally provides limited information on cell ultrastructure, EM is often required to inter- (J. Decelle).

pret the intracellular localization of elements and molecules. Correlation between light microscopy
and EM (CLEM) is well established [5,6], but correlation between EM and chemical imaging is less
developed. Bridging the data acquired with different high-resolution imaging strategies is the next
challenge and will make correlative subcellular imaging a new, powerful research tool towards
integrative cell biology.

This review aims to present the potential and limitations of state-of-the-art chemical imaging
techniques for nonspecialists who seek to obtain chemical information at the subcellular level.
We aim to guide biologists to the appropriate imaging technique and associated sample prepa-
ration to visualize and quantify elements or biomolecules in cells. We also summarize the new
developments in correlative subcellular imaging (Figure 1, Key Figure), highlight the role of such
combinatory techniques in disentangling the biochemical processes of a cell, and discuss future
challenges and directions in the field.

Potential and Limitations of Subcellular Chemical Imaging Platforms and


Required Sample Preparation
Multiple chemical imaging instruments are capable of visualizing the molecular, elemental, and
isotopic composition of a cell with high lateral resolution [7]. These microscopes are generally
equipped with a focused high-energy primary beam of electrons, protons, photons, or ions that
scans the surface of the sample and obtain quantitative information in a spatially
resolved manner (Box 1). The instrument and experimental setup need to be carefully chosen
according to the research question and the target elements or molecules of interest. Here we
focus on the key methodologies that are routinely used to provide subcellular information.
Lower-resolution technologies (e.g., MALDI, LA-ICP-MS) are not discussed in detail here
(see the review in [8]).

X-ray fluorescence (XRF) microscopy relies on the excitation of core electrons of atoms that
leads to X-ray emissions, which are specific to elements in the sample (Box 1). The primary probe
determines the technique: electrons in energy dispersive X-ray spectrometry [scanning/transmission
EM (S/TEM)-EDS]; protons in particle-induced X-ray emission (PIXE); and synchrotron-generated
photons in synchrotron XRF (S-XRF) imaging. These analytical techniques can be used to visualize
and quantify the distribution of macronutrients (e.g., P, S), key trace elements (e.g., Mn, Fe, Cu, Zn,
Se), toxic heavy metals (e.g., Hg, Pb), and pharmacological compounds (e.g., organometallic
compounds based on Pt, Ir, Os, or Ru). S/TEM-EDS can provide the highest spatial resolution
(subnanometer in TEM) with sensitivity of ~1000 ppm (1 mg g−1) [9]. Compared with this, PIXE is
less spatially resolved (submicron) but is more sensitive (ppm range) [7]. However, overall, S-XRF
provides arguably the best combination of high-spatial-resolution capabilities (down to a few
tens of nanometers) and high sensitivity (sub-ppm) to light and heavy elements (Figure 2 and
Box 1) [10,11]. S-XRF has allowed the mapping and quantification of metals such as Fe, Zn, and
Cu in microalgal and human cells [12–14] as well as silica, drugs, organometallic molecules,
and titanium oxide nanoparticles in cancer cells [2,15–19]. In combination with XRF imaging,

174 Trends in Cell Biology, March 2020, Vol. 30 , No. 3


Trends in Cell Biology

Key Figure Glossary


Outline of Correlative Multimodal Subcellular Imaging Workflow Including Chemical imaging: spatial
characterization of the chemical
Electron and Chemical Imaging composition of a sample (isotopes,
elements, molecules). This can be
achieved by multiple high-resolution
imaging platforms using different
physical processes to interrogate
subcellular information (e.g. XRF,
ionization).
Chemical landscape: composition
and distribution patterns of elements,
isotopes, and molecules in a sample
(e.g., cell). Its visualization cannot be
obtained with a single imaging platform
but different techniques need to be used
in a correlative way.
Correlative EM and chemical
imaging: workflow to prepare samples,
obtain micrographs from different
complementary imaging platforms, and
overlap morphological and chemical
(elements/molecules) information from
the same region of a specimen.
Hybrid SIMS: [Bi]n (n = 1, 3, 5, 7) or [Ar]
n (n ~ 1000) gas-cluster ion source used
for the analysis of large molecular ions.
The OrbiTrap analyzer provides a MRP
of ~10E5 that enables precise
compound identification.
Multimodal imaging: microscopy
observations of the same sample using
two or more imaging platforms to obtain
complementary morphological and
chemical information (e.g., light and EM,
nanoSIMS and S-XRF).
NanoSIMS: single-atomic Cs+ or O-
primary ions are used for both sputtering
and analysis; the ionized material is then
analyzed by a Mattauch–Herzog mass
spectrometer that allows the parallel
detection of a maximum of seven
masses. The high energy of the primary
ions causes strong fragmentation of
molecules down to single-atomic ions,
allowing quantitation of changes in
isotopic composition.
Secondary ion mass spectrometry
(SIMS): secondary ions are sputtered
away from the topmost layer of a sample
by a focused primary ion beam and
analyzed in a mass spectrometer.
NanoSIMS, ToF-SIMS, and hybrid SIMS
are SIMS imaging techniques with
primary ion beams of different sources
and energies and with different mass
spectrometers to probe elements,
isotopes, and small molecules.
Time-of-flight (ToF)-SIMS: pulsed [Bi]
n (n = 1, 3, 5, 7) or [Ar]n (n ~ 1000) cluster
Trends in Cell Biology
ion sources are mainly used for analysis
(See figure legend at the bottom of the next page.) while Ar[n] clusters, Cs+ or O- can be

Trends in Cell Biology, March 2020, Vol. 30, No. 3 175


Trends in Cell Biology

X-ray absorption spectroscopy (XAS) can be performed to reveal the chemical speciation of a used as sputtering sources. The use of
cluster ions for analysis reduces the
target element. XAS has disclosed the chemical transformations of indium-based nanocrystals
fragmentation on impact, leading to the
and of osmium-based anticancer drugs in cancer cells [20,21]. The modulations of the preservation of molecular species. The
XAS spectra also allowed mapping of the distribution of the different chemical states of ToF mass spectrometer allows the
S (e.g., sulfate esters, inorganic sulfate) in a biological tissue, to understand their role in cell simultaneous detection of all masses.
Vitreous cell: frozen hydrated cell with
differentiation [22]. amorphous ice (i.e., without crystals that
can alter the ultrastructure and chemical
Secondary ion mass spectrometry (SIMS) instruments are based on the analysis of the mass composition of cells). Vitreous cells can
of elements and molecules. Secondary ions are sputtered away from the topmost layer of a be obtained using high-pressure
freezing or plunge-freezing machines.
sample by a focused primary ion beam and analyzed in a mass spectrometer (Box 1). NanoSIMS X-ray fluorescence (XRF): physical
is a SIMS instrument particularly suitable for probing macronutrients and metals in cells at a lateral process comprising the emission of X-
resolution down to 50 nm (Figure 2) [23–26]. For instance, P, S, Ca, Fe, Zn, Mn, and Cu have been rays from a specimen following the
excitation of core electrons of atoms;
mapped in cells [12,25,27,28]. Morphological features of the cell can be revealed by a secondary
analysis of the emitted X-rays allows the
electron signal (only in negative extraction mode) and from different secondary ions, such as identification of the elemental content of
cyanide (12C14N−) and phosphorous (31P−) showing the overall shape and internal compartments the specimen. The excitation of
of the cell and the nucleus, respectively. The high mass-resolving power of nanoSIMS can also electrons can be achieved by a beam of
electrons, protons, or photons.
unveil the isotopic composition of a cell (i.e., being able to distinguish between 12C15N and
13 14 X-ray phase-contrast tomography:
C N) and so is highly suitable for stable isotope probing (SIP) [29,30]. SIP-nanoSIMS allows tomographic technique sensitive to the
quantitation of metabolic activities at the subcellular level (e.g., 50–100 nm), such as C and N refraction of X-rays in matter, leading to
assimilation [31–34]. This technique has been used to understand nutrient exchange between phase variations of the X-rays
depending on the sample’s electron
cells in symbiotic, pathogenic, and virus–host interactions [35–40] and the localization of drug
density, and particularly adapted to
compounds in human cells [41] (Figure 2). reveal weakly absorbing features like
those present in biological samples.
With time-of-flight (ToF)-SIMS (Box 1), molecular information can be obtained since the ion
probe (polyatomic or gas cluster) is less destructive than in nanoSIMS (monoatomic). The softer
ionization conditions compared with nanoSIMS allow spatially resolved analysis of large molecular
fragment species in the range 1 to ~1000 Da with a typical lateral resolution of 100 nm to 5 μm.
Different analysis modes exist: spectrometry mode to obtain high mass resolution, imaging mode
to obtain high lateral resolution, and delayed-extraction mode to combine high mass resolution
(10 000 MRP) with high lateral resolution (400 nm) [42]. Delayed extraction is now widely used
to image organic samples and recently was shown to achieve a 108-nm lateral resolution to
visualize single particles in algal biofilms [43,44]. ToF-SIMS is a useful method for studying
small molecules [45–47] and lipids in cells, especially in lipid-related diseases, such as cancer,
Duchenne muscular dystrophy, and atherosclerosis (Figure 2) [48,49].

Seeing is believing, but what we see critically depends on the sample preparation. Sample
preparation is one of the most fundamental steps and should aim to preserve cells as close as
possible to their native state – the Holy Grail in cell biology. The ideal method is one that fixes
and conserves both the ultrastructure of the cell and its native chemical composition (Box 2).
However, sample preparation is highly specific to both the sample and the instrument used
and compromises must be made at each experiment (Table 1).

Figure 1. Individual cells can be isolated from a population in a tissue, culture, or the environment and observed in vivo using
light/fluorescence microscopy for dynamic and functional imaging. After sample preparation (fixation and sectioning/milling),
a cell can be analyzed by different high-resolution imaging platforms in a correlative way. Electron microscopy can unveil
detailed ultrastructure of the cell while chemical imaging platforms [nano-secondary ion mass spectrometry (nanoSIMS),
X-ray fluorescence microscopy, time-of-flight (ToF)-SIMS, hybrid SIMS] enable the visualization and quantification of
elements, isotopes, and molecules at the subcellular level. Finally, image processing allows the correlation between
multimodal micrographs that contain complementary information on the cell. This workflow still requires some
methodological developments at various steps, from sample preparation to image processing, to further understand the
metabolism and physiology of a cell at the nanoscale in its close-to-native state.

176 Trends in Cell Biology, March 2020, Vol. 30 , No. 3


Trends in Cell Biology

Box 1. Principles of Subcellular Chemical Imaging Techniques


XRF relies on the photoelectric effect occurring when an X-ray photon is absorbed by an atom: a photoelectron is ejected from a core orbital of the excited atom leaving a
vacancy, which is then filled by another electron from a higher orbital. This is followed by the emission of a fluorescent X-ray photon whose energy is characteristic of the
element. XRF acquisition is often performed by scanning the sample and energy dispersive detectors collect and measure the emitted fluorescence spectrum in each
pixel, unveiling the elemental composition. The beam size determines the spatial resolution, from 10 nm to 1 μm. For a thin sample, the intensity of the fluorescence is
proportional to the concentration of the elements, which can be calculated using thin standards of known concentration (Figure I).

In SIMS, on the impact of a focused ion beam, sample material is sputtered and about 1% of ejected material is ionized. In nanoSIMS, these ions are extracted in negative
or positive modes into a mass spectrometer and separated according to their mass-to-charge ratio (m/z) in the magnetic sector of the mass analyzer. Simultaneous
detection of up to seven secondary ion species (monoatomic ions or small molecular fragments of up to four to six atoms) can be achieved. The ion beam can scan
a predefined surface area of the cell, therefore providing color-coded cartography of ion counts per pixel. Note that the charge compensation (i.e., compensating the
buildup of charge on nonconductive surfaces) is available only in negative extraction mode. Therefore, coating the sample surface with a conductive metal (~10 nm)
is mandatory to overcome this limitation in positive extraction mode.

In ToF-SIMS, the ejection and ionization of material relies on a lower ion beam fluence and less destructive cluster ion sources compared with nanoSIMS. With new
cluster ion sources (Bin, Arn, Aun, C60), ToF-SIMS provides the possibility of minimizing molecular damage while maximizing molecular ion yields. Small organic molecules
in the range of 1 to ~1000 Da can be detected with a lateral resolution of around a micron on biological material. The pulsed operation mode of the primary ion gun and its
45° mounting geometry allows charge compensation in both extraction polarities while employing the same primary ion species. However, the 45° geometry can cause
a shadowing and lateral displacement effect on depth profiling or when analyzing a surface with a pronounced topography.

Trends in Cell Biology

Figure I. Principles of Subcellular Chemical Imaging Techniques.

Correlated Subcellular Imaging towards Integrative Cell Biology


Correlation between Morphology and Chemical Imaging
Since chemical imaging provides little morphological information, combination with light microscopy/
EM is required to unambiguously elucidate the localization of elements and molecules in a cell.
However, the challenge is the trade-off between ultrastructure and chemistry preservation of the
cell during the sample preparation and the ability to transfer and analyze the same cells on different

Trends in Cell Biology, March 2020, Vol. 30 , No. 3 177


Trends in Cell Biology

Trends in Cell Biology

Figure 2. The Potential of Chemical Imaging to Unveil the Chemical Landscape of a Cell: Composition and Distribution of Elements, Isotopes, and
Molecules at the Nanoscale. (A–C) Nano-secondary ion mass spectrometry (nanoSIMS) images showing the distribution of the macronutrients nitrogen [(A)
12
C14N−], phosphorous [(B) 31P−], and sulfur [(C) 32S−] inside a microalgal cell. (D) NanoSIMS image showing the uptake of 13C incorporated in proteins (13C14N) in cells
after incubation in 13C-labeled bicarbonate [stable isotope probing (SIP)-nanoSIMS]. (E) NanoSIMS image acquired on macrophages treated with the drug iodine-
containing amiodarone. The overlay of 31P− (blue) and 127I− (purple) secondary ion maps provides morphological information (localization of the nucleus) and shows the
specific localization of the drug within the lysosomes. Reproduced, with permission, from [84]. (F) Specific labeling of proteins for correlated fluorescence microscopy
and nanoSIMS using FluorLink–nanobody anti-GFP and direct immunostaining strategies. NanoSIMS image of 19F/12C14N ratio shows the presence of the targeted
protein in specific cellular areas. Reproduced, with permission, from [65]. (G) Visualization of antibiotic in cells. Overlay of nanoSIMS and electron microscopy images
showing the accumulation of the antibiotic in lipid droplets (LDs). The bromine-containing antibiotic (bedaquiline) can be detected and semiquantified by nanoSIMS
(Figure legend continued at the bottom of the next page.)

178 Trends in Cell Biology, March 2020, Vol. 30 , No. 3


Trends in Cell Biology

Box 2. Sample Preparation for Subcellular Imaging


The sample preparation workflow depends on the sample, imaging platform, and targeted compound (e.g., molecule, element, isotope) (Figure I). Because of the high-
vacuum conditions in most EM and chemical imaging instruments, live cells cannot be directly analyzed but generally need to be fixed, dehydrated, and sectioned/milled
to access subcellular structures (Table 1). For fixation, the use of aldehydes at room temperature can alter the ultrastructure and chemistry of the cell since diffusible
mobile ions and small molecules (e.g., free amino acids, lipids) can be redistributed and/or washed out from the cell. Therefore, rapid freezing methods (high-pressure
freezing and plunge freezing) are superior in preserving native-state cell structure and chemistry at a fast rate (a scale of milliseconds compared with minutes for chemical
fixation). Then, freeze substitution is a dehydration step where the ice of the vitreous cell is slowly replaced by an organic solvent at a very low temperature (from −90°C
to −30°C) with retention of elements and metabolites possible, provided the substitution protocol is chosen carefully [13,50]. Chemical fixatives, such as osmium, tannic
acid, or aldehydes, can be used during this step to stabilize cellular structures [51]. A resin-embedding step is then necessary to obtain thin sections and access the
cell’s interior. Cryodehydration can also be performed by freeze drying, but this method can cause ultrastructural modification and elemental redistribution. However,
freeze drying is particularly powerful for drying ultrathin frozen-hydrated sections for room-temperature analysis. After sample preparation, chemical preservation of
the cell can be assessed by visualizing the most diffusible elements (K+, Na+, Ca2+, Cl−) that move rapidly across membranes and within the cytoplasm, thus
representing a relevant rule-of-thumb criterion for chemical preservation [52]. Overall, it remains difficult to be certain that the chemical environment in a cell is a true
representation of normal physiology, and each step of the preparation can be debatable. We recommend more methodological development and comparisons in
the future to optimize sample preparation and assess putative artefacts. This is a challenge since access to cutting-edge microscopes is generally difficult for that meth-
odological purpose. Compared with freeze-substituted and resin-embedded cells, analysis of frozen-hydrated cells (or vitreous cells) is obviously superior for chemical
preservation (Table 1). However, this leads to many challenges that need to be tackled in the future (further discussed in this review): (i) cryosectioning; (ii) the need for a
cryotransfer system and a cryostage in the imaging platform; (iii) difficulty in undertaking correlative studies across different platforms; and (iv) inherent lack of contrast for
sample visualization.

Trends in Cell Biology

Figure I. Sample Preparation for Subcellular Imaging.

through the 79Br ions (red signal). Reproduced, with permission, from [41]. (H) Time-of-flight (ToF)-SIMS images showing accumulation of phosphates (red; PO3−) in biofilm
of algal cells (green; CN−) growing in cotton (blue; CH4S−). Reproduced, with permission, from [47]. (I) Synchrotron X-ray fluorescence image showing the distribution of
indium phosphide-based nanocrystals in a frozen section of Hydra vulgaris. Nanocrystals detected by indium X-ray fluorescence (in red) are mainly internalized in the
ectoderm layer (ect). The natural macronutrients phosphorous (blue) and sulfur (green) provide the morphological context. Reproduced, with permission, from [21].
(J) Synchrotron X-ray fluorescence image showing the presence of silver nanowires in a fibroblast cell (S in green, Ag in red). Yellow regions indicate colocalization of
Ag and S inside the cell. Reproduced, with permission, from [4]. Copyright 2019 National Academy of Sciences. (K) NanoSIMS image showing the distribution of Ca
(40Ca+) and Fe (56Fe+) in a microalga. Calcium mapping unveils the overall morphology of the cells, with high concentration in the nucleus (nucl). Iron is mostly
contained in the plastids (pla). (L,M) Synchrotron X-ray fluorescence images showing the subcellular distribution and quantification of the trace metals Fe (red), Co
(blue), and Os (green) in microalgal cells. Pla, plastid of microalgal cell; Cyt, cytoplasm; Nucl, nucleus; LD, lipid droplet; ect, ectoderm; end, endoderm.

Trends in Cell Biology, March 2020, Vol. 30 , No. 3 179


Trends in Cell Biology

Table 1. Detailed Procedures for Sample Preparation for EM and Chemical Imaging and Considerations (Pros and Cons) for Each Step
Sample preparation Strategy Pros Cons
step
Fixation of cell Chemical fixation Easy to use in the field or for pathogens (human Ultrastructure and chemical composition can be
parasites) greatly modified
Cryofixation under high Excellent preservation of the ultrastructure and Thickness of the sample must be less than 200 μm,
pressure chemistry of the cell requires bulky laboratory-based equipment
Cryofixation with plunge Can be done in the field or the laboratory Maximum sample thickness to maintain vitreous
freezing in a liquid cryogen ice formation is ~5 μm, some ice crystal
formation in thicker samples
Dehydration Chemical dehydration at Can be done in the field or the laboratory Structural and chemical preservation are not
room temperature guaranteed
Freeze drying No use of chemicals or solvents, ideal for drying Likelihood of movement of target ions (particularly
ultrathin frozen-hydrated sections for diffusible elements) or metabolites, especially in
room-temperature analysis highly vacuolated tissues
Freeze substitution Dehydration at very low temperature allows good Use of solvents can extract materials of interest,
structural and chemical preservation long process (days to weeks)
Use of chemical Osmium Membranes are fixed and osmium provides contrast Highly toxic, interferes with some molecules and
fixative during the for EM investigation and structural information in XRF elements for XRF and ToF-SIMS analysis
freeze substitution
Aldehydes Proteins are fixed, maintaining structural preservation Toxic, no contrast for EM investigation, cannot
be prepared as anhydrous (therefore loss of any
water-soluble material)
Acrolein Crosslinks at low temperatures making it highly suited Hazardous material
for use with freeze substitution, can be anhydrous
Resin embedding Plastic epoxy resin Good structural preservation and contrast Often contains Cl, requires solvents for good
infiltration
Methacrylate resins Preservation of antigenicity, low viscosity ideal for Poor stability under an ion beam, usually require
difficult-to-embed samples O-free environment to cure
Sectioning Wet sectioning Easy to collect the sections Highly diffusible molecules can be washed out
Dry sectioning Avoidance of water/liquids allowing retainment of Difficult to cut sections thinner than 500 nm, difficult
water-soluble ions and molecules to obtain flat/uncompressed sections for analysis
Cryoanalysis Frozen-hydrated cells The best structural and chemical preservation close Lack of contrast and structural information,
to the native state, whole cells or cross sections of correlative approaches across platforms
cells can be analyzed currently difficult, cryosectioning (microtomy or
cryo-FIB) highly specialized

imaging platforms. Some imaging techniques, such as SIMS, are destructive, meaning that morpho-
logical imaging must generally take place beforehand. Here we propose various strategies that can
be adopted to analyze the same cellular region of interest with multimodal imaging (Figure 1).

Organelles can be labeled and observed with fluorescence microscopy before the entire cell is
subjected to chemical imaging. For instance, the accumulation of Mn in the Golgi apparatus of
dopaminergic cells was revealed using GFPs targeting the organelle followed by S-XRF imaging
under cryogenic conditions [53–55]. More recently, correlation between super-resolution stimu-
lated emission depletion microscopy of proteins and S-XRF imaging of trace metals was per-
formed with 40-nm spatial resolution on neurons [56].

To obtain a high-resolution cellular context, it is also possible to analyze cell sections in S/TEM
followed by S-XRF, providing an unambiguous spatial origin of elements in subcellular compart-
ments (Figures 3 and 4) [57]. Osmium tetroxide (OsO4) can be used to fix and stain cellular com-
partments (Table 1), providing morphological contrast not only on EM, but also on S-XRF, where
Os fluorescence reveals the ultrastructure of the cell (Figures 2 and 3; [12]). The drawback is that

180 Trends in Cell Biology, March 2020, Vol. 30 , No. 3


Trends in Cell Biology

Trends in Cell Biology

Figure 3. Examples of Correlated Electron Microscopy (EM) and Chemical Imaging. (A) Correlation between EM
(left images) and nano-secondary ion mass spectrometry (nanoSIMS) (middle images) showing the sulfur (32S−; upper image)
and nitrogen (12C14N−; lower image) content of a microalgal cell. Right images show the overlay of macronutrient mapping
and ultrastructure obtained from consecutive sections or the same section. Image courtesy of Charlotte Lekieffre.
(B) Correlation between scanning EM (SEM) and synchrotron X-ray fluorescence microscopy (S-XRF). SEM observation
has been performed on the same cell section after S-XRF analysis. S-XRF mapping of phosphorous and iron (green) and
sulfur (red) unveils numerous hotspots (one example highlighted by the yellow circle) where sulfur and iron are colocalized
at high concentration in the cell. (C) Transmission EM (TEM) image (left) with corresponding energy-filtered TEM (EFTEM)
Fe map (right) showing aggregated ferritin molecules in a cell. By courtesy of Jeremy Shaw and David Keays. Bar,
100 nm. (D) Correlation between SEM, time-of-flight (ToF)-SIMS, and nanoSIMS showing the cell ultrastructure and
(Figure legend continued at the bottom of the next page.)

Trends in Cell Biology, March 2020, Vol. 30 , No. 3 181


Trends in Cell Biology

Trends in Cell Biology

Figure 4. How to Visualize Metals in Cells. Flowchart to guide users when probing metals in a single cell at high
resolution and sensitivity.

distribution of sulfur molecules (CHSN−, S−, HS−) and sulfur (32S−), respectively. These multimodal images were acquired
from consecutive thin sections. (E) EM hybrid SIMS image showing the 3D distribution in a single cell of the drug
amiodarone (m/z 646; green) and biomolecules at m/z 157 (purple) and m/z 184 (green) (upper image). Lower image
obtained from the hybrid SIMS instrument shows that C24:1 sulfatides (m/z 888.62; green) are localized to the corpus
collosum. The DNA base adenine (red; m/z 134.05), a nuclear marker, shows that neurons are densely packed in the
pyramidal layer and sparsely packed in the striatum oriens, where phosphoinositol is located (m/z 241.01; blue). Adapted,
with permission, from [75]. (F) Direct correlation between X-ray phase-contrast and X-ray fluorescence tomography on a
malaria-infected cell. The 3D mass density volume is obtained after tomographic reconstruction (on the left). Subsequent
X-ray fluorescence scanning measurements were performed on the same sample showing the 3D mass concentration
volumes of iron, sulfur, and phosphorous (on the right). Reproduced, with permission, from [70].

182 Trends in Cell Biology, March 2020, Vol. 30 , No. 3


Trends in Cell Biology

the XRF emission lines of Os interfere with those of phosphorous and some trace metals (e.g., Cu,
Zn), increasing their detection limit. To obtain structural and elemental/isotopic information from a
single cellular region, it is possible to perform TEM followed by nanoSIMS (Figure 3). Usually it
would require the use of a specific TEM grid with coordinates or fiducial markers to find the
same regions of interest in the two instruments (Figures 3 and 4) [35,37,58,59]. With the recent
advent of sensitive backscatter detectors in modern SEM, it is also possible to acquire structural
information from sections using SEM before nanoSIMS and S-XRF analyses, on the same sample
or on consecutive sections [12,41,60].

Correlation between Fluorescence and NanoSIMS Using Element Labeling


The coupling between fluorescence microscopy and nanoSIMS can be a powerful approach
to unveil the functional identity of a cell and organelles. This relies on targeted probing of
DNA, RNA, and proteins by coupling fluorescent dyes to elements usually absent from the
cell’s natural composition, such as halogens (fluorine, bromine), gold, and boron. A specific
exogenous element (detectable by nanoSIMS) can be associated with/linked to fluorescent
dyes, antibodies, or nanobodies, allowing correlative microscopy between fluorescence
in situ hybridization (FISH), immunocytochemistry, or click chemistry approaches and
nanoSIMS [23,61,62]. For example, the nucleotide analog bromodeoxyuridine (BrdU), which
is incorporated into replicating DNA during cell division, can be detected by fluorescence
immunohistochemistry but also as bromine ions (e.g., 79Br, 81Br) by nanoSIMS in the same
cells [63]. Fluorine (19F) labeling of proteins using a 19F-azide probe [64] or by conjugation to
nanobodies (e.g., GFP-like proteins and antibodies) can enable correlated fluorescence
and nanoSIMS imaging [65] (Figure 2). Similarly, 197Au can be conjugated to an antibody
to recognize cellular actin and synaptophysin proteins by coupled immunofluorescence
and nanoSIMS [66], while antibodies tagged with isotopically pure elemental metal reporters
(i.e., lanthanides) have been utilized to image protein expression in human breast tumor tissue
sections [67]. Recently, boron linked to proteins or to nanobodies binding to proteins has been
used for simultaneous protein identification and elemental mapping by correlative fluorescence
microscopy and nanoSIMS [68]. These studies suggest that exogenous elements of small
mass and size are suitable for probing DNA and proteins in the complex cellular environment
and can be used in correlated nanoSIMS studies.

Correlation between More Than Two Subcellular Imaging Platforms


The combination of multiple subcellular imaging platforms can provide a comprehensive view of
the ultrastructure, concentration, and distribution of elements (macronutrients and metals) and
isotopic ratios, and molecules from a single region of interest in the cell (Figure 3). This can be per-
formed by analyzing different consecutive resin sections from the same cell on different platforms
[12,13,57]. This flexible strategy allows one to choose the required thickness and support
(e.g., wafer, grid, Si window) for each section, and detailed ultrastructure can be obtained with
EM. Thus, the morphological and various chemical features can be superimposed. However,
collecting serial sections of different thicknesses to target the same cellular region or organelle is
a challenge.

Integrated Correlative Instruments


A correlative approach can be facilitated when morphological and chemical information from exactly
the same area can be acquired in the same instrument. One of the best examples is TEM-EDS and
EFTEM. TEM offers the highest possible resolution for both imaging and element analysis. Provided
that samples are thin (usually ~150 nm at 200 kV) and the elements of interest can be preserved,
simultaneous structural and elemental information can be obtained at the nanoscale (Figure 3)
[69]. However, molecular information is not available and absolute quantitation can be difficult.

Trends in Cell Biology, March 2020, Vol. 30, No. 3 183


Trends in Cell Biology

Using synchrotron X-ray nanoprobes, the combination of X-ray phase-contrast tomography and
XRF microscopy can provide the morphological information and quantification of elements,
respectively (Figure 3). This has been recently performed on a freeze-dried human phagocytic cell
[16] and human red blood cells infected with the malaria parasite Plasmodium falciparum [70]. An
alternative to phase-contrast imaging is ptychography [71], which can be combined with XRF
tomography to obtain the 3D localization of elements in cells, such as bacteria in [72].

New instruments that offer simultaneous morphological and molecular information are emerging
but are yet to be applied to biological specimens. For instance, SIMS in helium-ion microscopy
(HIM-SIMS) can combine high-resolution morphological images with elemental and isotopic
maps from SIMS [73]. In contrast to EDS on SEM, HIM-SIMS provides better detection limits
for elements (including the very light ones) and differentiation between isotopes. However,
using HIM-SIMS for stable-isotope labeling experiments will require a significant improvement
of the mass resolution. Overall, the combination of high-resolution secondary electron images
and mass-separated sputtered ion distributions has high potential to answer open questions in
cell biology.

Image Processing for Multimodal Correlative Imaging


On their acquisition on different imaging platforms, micrographs need to be processed to corre-
late or even overlap information from the same cellular region. Multimodal 2D microscopy requires
registration algorithms that can analyze the same region but at different fields of view and resolu-
tions from images acquired with different excitation probes (e.g., photons, electrons, ions) as well
as different detectors. Compared with monomodal image processing, more sophisticated
approaches are needed for multimodal images since the pixel intensities of features are not com-
parable or do not even occur in both images. The additional challenge in multimodal data sets is
that the shape of the object might differ because of preparation steps or the different probing
depths of the microscopes. Distortions may also be introduced when subsequent sections,
representing different depth layers of the sample, are used for correlative imaging. In this context,
the ImageJ-based software Correlia has recently been developed for the registration of 2D–2D
multimodal microscopy data sets (available on request).

Perspectives and Future Challenges for Multimodal Subcellular Imaging


Subcellular Mapping of Metabolites in Cells
Visualization of the compartmentalized distribution of metabolites in cells is one of the most
promising research avenues in biology. Recently, laser-based ionization mass spectrometry
techniques have been successfully used in single-cell metabolomics profiling experiments,
but the lateral resolution precludes imaging of subcellular chemistry [74]. Mapping metabolites
at subcellular level can be now envisioned with the revolutionary hybrid SIMS instrument (3D
OrbiSIMS, IONTOF) that combines the speed and high-lateral-resolution imaging capabilities of
ToF-SIMS with the unprecedented mass resolution (240K), mass accuracy (sub-ppm), dynamic
range (S/N ratio ~ 105), and MS/MS capabilities of the Orbitrap Q-Exactive [75]. The instrument
has been used to map the distribution of lipids and neurotransmitters in the hippocampal region
of the mouse brain at the cellular and subcellular level. The 3D imaging capabilities were used to
visualize the accumulation of amiodarone in single lung macrophage cells and to assess its toxic
effect by correlating drug concentration with the levels of phospholipids and cholesterol [75].

Future Improvements Needed to Probe Cells in Their Native State and in Four Dimensions
The future of chemical imaging largely relies in the development of cryoanalyses and associated
correlative workflows to allow multiscale chemical analysis of cells in their native state across mul-
tiple instruments. Currently, cryoanalyses present limitations with regard to the suitability of

184 Trends in Cell Biology, March 2020, Vol. 30 , No. 3


Trends in Cell Biology

samples for cryopreservation and their transfer between different platforms without ice contami- Outstanding Questions
nation. In most cases, frozen cells must be sectioned/milled to visualize the interior. Future expan- • Is it possible to preserve the native
sion in this area will require improvements in the preparation of quality cryosections (e.g., cryo- physiological state of a cell from
sample preparation to imaging?
FIB-SEM), the development of cryoenabled instrument platforms (e.g., nanoSIMS), and the ability
How can structural and chemical
to transfer samples seamlessly between these. Additional challenges associated with preservation of the cell be carefully
cryoimaging are the low contrast of vitrified cells and possible devitrification under irradiation assessed and artifacts identified to
from the probe (e.g., ions, X-rays). avoid misleading results?
• Can we resolve the chemical landscape
(composition and distribution of
Chemical information in 2D is insufficient to fully describe the compartmentalization of an element/
elements and molecules) of a cell
molecule throughout an entire cell, especially when information comes from a single thin section with high spatial resolution, in three
(60–300 nm thickness) of a cell. Techniques to acquire 3D structural information at the subcellular dimensions, and over a relevant
level are readily available (e.g., FIB-SEM) but it is difficult to couple these with analytical informa- temporal scale? What would be the
analytical workflow to implement for
tion. Synchrotron-based coherent X-ray scattering techniques such as holotomography or 4D subcellular imaging?
ptychography have both demonstrated constant improvements towards the ultrastructural 3D • What are the methodological and
characterization of a cell architecture [76]. We therefore foresee a bright future in the coupling technological strategies to analyze
between these techniques and XRF tomography for 3D elemental imaging. the same subcellular region of
interest across different subcellular
imaging techniques?
The downside of high-resolution chemical imaging is the low throughput, which precludes
• How can we include cryoanalysis in the
robust statistical analyses. We can expect that ultrahigh-speed scanning strategy and highly workflow of correlative subcellular
efficient detection will be the next steps to foster on the instrumentation side. For example, imaging, from sample preparation and
extremely brilliant X-ray synchrotron sources and high-rate scanning strategies have become obtaining vitreous cells to transfer and
imaging across different platforms?
reality with the upgrade of synchrotrons such as the European Synchrotron Radiation Facility Can it be applied to different types of
(ESRF) [77]. cells from a tissue or isolated in culture
or the environment?
Finally, future developments need to integrate a biologically relevant temporal dimension into the • How can the throughput of chemical
imaging techniques be increased to
correlative imaging workflow towards 4D imaging. Because probing elements and molecules of
observe large numbers of biological
live cells at the nanoscale level remain challenging, there is a need to capture multiple snapshots samples and perform statistical
of the phenotypic state of a cell over time to follow and better understand dynamic cellular pro- comparisons?
cesses following exposure to abiotic or biotic stress. A single snapshot may provide a biased • Can we enhance the mass resolution
vision of the phenotypic response of a cell if imaging analysis occurs on an inappropriate timescale. and spatial resolution of SIMS imaging
instruments to visualize a large number
Temporal resolution can be obtained by coupling with live imaging (fluorescence and super- of different metabolites in the cell?
resolution microscopy) and the development of microfluidic devices [78] or rapid cryofixation
strategies [79]. For instance, dynamic fluorescence light microscopy can be rapidly followed by
cryoimmobilization in few seconds for CLEM studies thanks to new tools (e.g., CryoCapsule)
and could be extended to chemical imaging in the future [80]. In addition, recent advances in
super-resolution fluorescence microscopy [e.g., stimulated emission depletion (STED) microscopy]
[81] and the future development of new fluorescent, element-specific probes will potentially provide
a dynamic view of labile elements (e.g., Ca2+, Fe2+/3+, Zn2+) in live cells at 10–50 nm resolution [82].
Correlation with chemical imaging will open new perspectives bridging temporal and spatial
resolution [83].

Thus, the development of time-resolved 3D subcellular imaging under cryoconditions will be a


breakthrough in cell biology that will capture dynamic subcellular processes in a native-state cell.

Concluding Remarks
Subcellular chemical imaging techniques are constantly improving and becoming ever-more-
powerful tools for quantitative visualization of elements, isotopes, and molecules in cells. How-
ever, untangling their complex requirements and capabilities is a vital step in ensuring that
researchers can apply such methods to outstanding research questions and problems (see
Outstanding Questions). With this, appropriate sample preparation and suitable imaging

Trends in Cell Biology, March 2020, Vol. 30 , No. 3 185


Trends in Cell Biology

platforms need to be selected according to the sample, spatial resolution, and targeted ele-
ments/molecules. In this review, we have outlined the principles of key analytical instrumentation,
discussed strategies for sample preparation, and highlighted the potential for correlative EM and
chemical imaging to accumulate structural and chemical information from a single region of a cell.
Correlated morphological and chemical imaging has the potential to spur a rapid expansion in
various fields, such as cell biology, biomedicine, ecophysiology, pharmacology, toxicology, and
biogeochemistry. In the near future, we do not foresee a technique that would encompass all of
the capabilities to explore the chemical/molecular/isotopic composition and ultrastructure of a
cell. Thus, the development of integrative studies and dedicated analytical correlative workflows,
from sample preparation to multimodal imaging and image processing, will be a major contribution
towards a full comprehension of the physiology of the cell at the subcellular level.

Acknowledgments
J.D. was supported by the LabEx GRAL (ANR-10-LABX-49-01), financed within the University Grenoble Alpes graduate
school (Ecoles Universitaires de Recherche) CBH-EUR-GS (ANR-17-EURE-0003), and Défi X-Life grant from CNRS. The
authors acknowledge the support and use of resources from Instruct (a Landmark ESFRI project) and the ESRF for pro-
viding beamtime. We are thankful for the use of the analytical facilities of the Centre for Chemical Microscopy (ProVIS) at
UFZ Leipzig, which is supported by European Regional Development Funds (EFRE – Europe Funds Saxony) and the Helmholtz
Association. This research is also supported by EMBRC-France, whose French state funds are managed by the ANR within the
Investments of the Future program under reference ANR-10-INBS-02. We also thank Yannick Schwab for critically reading the
manuscript and suggesting improvements. We are grateful to colleagues for sharing microscopy images.

References
1. Salt, D.E. et al. (2008) Ionomics and the study of the plant combined with X-ray phase contrast nanotomography. Appl.
ionome. Annu. Rev. Plant Biol. 59, 709–733 Phys. Lett. 112, 053701
2. Hackett, M.J. et al. (2019) Elemental characterisation of the py- 17. Brown, K. et al. (2018) Intracellular in situ labeling of TiO2 nano-
ramidal neuron layer within the rat and mouse hippocampus. particles for fluorescence microscopy detection. Nano Res. 11,
Metallomics 11, 151–165 464–476
3. Veronesi, G. et al. (2016) Visualization, quantification and coordi- 18. Fus, F. et al. (2019) Intracellular localization of an osmocenyl-
nation of Ag+ ions released from silver nanoparticles in hepato- tamoxifen derivative in breast cancer cells revealed by synchro-
cytes. Nanoscale 8, 17012–17021 tron radiation X-ray fluorescence nanoimaging. Angew. Chem.
4. Lehmann, S.G. et al. (2019) Crumpling of silver nanowires by Int. Ed. Engl. 58, 3461–3465
endolysosomes strongly reduces toxicity. Proc. Natl. Acad. 19. Kapishnikov, S. et al. (2019) Mode of action of quinoline antima-
Sci. U. S. A. 116, 14893–14898 larial drugs in red blood cells infected by Plasmodium falciparum
5. de Boer, P. et al. (2015) Correlated light and electron microscopy: revealed in vivo. Proc. Natl. Acad. Sci. U. S. A. 116,
ultrastructure lights up! Nat. Methods 12, 503–513 22946–22952
6. Karreman, M.A. et al. (2016) Intravital correlative microscopy: 20. Sanchez-Cano, C. et al. (2019) Nanofocused synchrotron X-ray
imaging life at the nanoscale. Trends Cell Biol. 26, 848–863 absorption studies of the intracellular redox state of an organo-
7. da Cunha, M.M.L. et al. (2016) Overview of chemical imaging metallic complex in cancer cells. Chem. Commun. 55,
methods to address biological questions. Micron 84, 23–36 7065–7068
8. Baker, T.C. et al. (2017) Recent advancements in matrix-assisted 21. Veronesi, G. et al. (2019) In vivo biotransformations of indium
laser desorption/ionization mass spectrometry imaging. Curr. phosphide quantum dots revealed by X-ray microspectroscopy.
Opin. Biotechnol. 43, 62–69 ACS Appl. Mater. Interfaces 11, 35630–35640
9. Lauwers, M. et al. (2013) An iron-rich organelle in the cuticular 22. Hackett, M.J. et al. (2016) Chemical biology in the embryo: in situ
plate of avian hair cells. Curr. Biol. 23, 924–929 imaging of sulfur biochemistry in normal and proteoglycan-
10. Cotte, M. et al. (2017) The ID21 X-ray and infrared microscopy deficient cartilage matrix. Biochemistry 55, 2441–2451
beamline at the ESRF: status and recent applications to artistic 23. Gyngard, F. and Steinhauser, M.L. (2019) Biological explorations
materials. J. Anal. At. Spectrom. 32, 477–493 with nanoscale secondary ion mass spectrometry. J. Anal. At.
11. Pushie, M.J. et al. (2014) Elemental and chemically specific X-ray Spectrom. 34, 1534–1545
fluorescence imaging of biological systems. Chem. Rev. 114, 24. Agüi-Gonzalez, P. et al. (2019) SIMS imaging in neurobiology
8499–8541 and cell biology. J. Anal. At. Spectrom. 34, 1355–1368
12. Decelle, J. et al. (2019) Algal remodeling in a ubiquitous planktonic 25. Malherbe, J. et al. (2016) A new RF plasma oxygen primary ion
photosymbiosis. Curr. Biol. 29, 968–978.e4 source on nanoSIMS for improved lateral resolution and detec-
13. Kashiv, Y. et al. (2016) Imaging trace element distributions in single tion of electropositive elements at single cell level. Anal. Chem.
organelles and subcellular features. Sci. Rep. 6, 21437 88, 7130–7136
14. Adams, M.S. et al. (2016) Copper uptake, intracellular localization, 26. Nuñez, J. et al. (2018) NanoSIMS for biological applications:
and speciation in marine microalgae measured by synchrotron ra- current practices and analyses. Biointerphases 13, 03B301
diation X- ray fluorescence and absorption microspectroscopy. 27. Weng, N. et al. (2017) In situ subcellular imaging of copper
Environ. Sci. Technol. 50, 8827–8839 and zinc in contaminated oysters revealed by nanoscale
15. Ciccotosto, G.D. et al. (2014) Quantitation and localization of secondary ion mass spectrometry. Environ. Sci. Technol.
intracellular redox active metals by X-ray fluorescence micros- 51, 14426–14435
copy in cortical neurons derived from APP and APLP2 knockout 28. Tsednee, M. et al. (2019) Manganese co-localizes with calcium
tissue. Metallomics 6, 1894–1904 and phosphorus in Chlamydomonas acidocalcisomes and is
16. Gramaccioni, C. et al. (2018) Nanoscale quantification of intra- mobilized in Mn-deficient conditions. J. Biol. Chem. 294,
cellular element concentration by X-ray fluorescence microscopy 17626–17641

186 Trends in Cell Biology, March 2020, Vol. 30 , No. 3


Trends in Cell Biology

29. Mayali, X. (2020) NanoSIMS: microscale quantification of synchrotron radiation focused beams. J. Anal. At. Spectrom.
biogeochemical activity with large-scale impacts. Annu. Rev. 30, 2525–2532
Mar. Sci. 12, 449–467 53. Das, S. et al. (2019) Manganese mapping using a fluorescent
30. Musat, N. et al. (2016) Tracking microbial interactions with Mn2+ sensor and nanosynchrotron X-ray fluorescence reveals
nanoSIMS. Curr. Opin. Biotechnol. 41, 114–121 the role of the Golgi apparatus as a manganese storage site.
31. Stryhanyuk, H. et al. (2018) Calculation of single cell assimilation Inorg. Chem. 58, 13724–13732
rates from SIP-nanoSIMS-derived isotope ratios: a comprehen- 54. Carmona, A. et al. (2019) Mapping chemical elements and iron
sive approach. Front. Microbiol. 9, 2342 oxidation states in the substantia nigra of 6-hydroxydopamine le-
32. Berthelot, H. et al. (2018) NanoSIMS single cell analyses reveal sioned rats using correlative immunohistochemistry with proton
the contrasting nitrogen sources for small phytoplankton. ISME and synchrotron micro-analysis. Front. Neurosci. 13, 1014
J. 13, 651–662 55. Carmona, A. et al. (2014) Environmental manganese com-
33. Terrado, R. et al. (2017) Autotrophic and heterotrophic acquisi- pounds accumulate as Mn(II) within the Golgi apparatus of
tion of carbon and nitrogen by a mixotrophic chrysophyte dopamine cells: relationship between speciation, subcellular dis-
established through stable isotope analysis. ISME J. 11, tribution, and cytotoxicity. Metallomics 6, 822
2022–2034 56. Domart, F. et al. (2019) Correlating STED and synchrotron XRF
34. He, C. et al. (2018) NanoSIMS imaging reveals unexpected nano-imaging unveils the co-segregation of metals and cytoskele-
heterogeneity in nutrient uptake by brown adipocytes. Biochem. ton proteins in dendrites. bioRxiv Published online October 21,
Biophys. Res. Commun. 504, 899–902 2019. https://www.biorxiv.org/content/10.1101/810754v1
35. Lekieffre, C. et al. (2018) Inorganic carbon and nitrogen assimilation 57. Sanchez-Cano, C. et al. (2017) Synchrotron X-ray fluorescence
in cellular compartments of a benthic kleptoplastic foraminifer. Sci. nanoprobe reveals target sites for organo-osmium complex in
Rep. 8, 10140 human ovarian cancer cells. Chemistry 23, 2512–2516
36. Volland, J.M. et al. (2018) NanoSIMS and tissue autoradiography 58. Clode, P.L. et al. (2009) In situ mapping of nutrient uptake in the
reveal symbiont carbon fixation and organic carbon transfer to rhizosphere using nanoscale secondary ion mass spectrometry.
giant ciliate host. ISME J. 12, 714–727 Plant Physiol. 151, 1751–1757
37. Gibbin, E. et al. (2019) Vibrio coralliilyticus infection triggers a be- 59. Kopp, C. et al. (2015) Subcellular investigation of photosynthesis-
havioural response and perturbs nutritional exchange and tissue driven carbon assimilation in the symbiotic reef coral Pocillopora
integrity in a symbiotic coral. ISME J. 13, 989–1003 damicornis. MBio 6, e02299-14
38. Martínez-Pérez, C. et al. (2016) The small unicellular diazotrophic 60. Hu, X. et al. (2019) Release of cholesterol-rich particles from the
symbiont, UCYN-A, is a key player in the marine nitrogen cycle. macrophage plasma membrane during movement of filopodia
Nat. Microbiol. 1, 16163 and lamellipodia. Elife 8, e50231
39. Pasulka, A.L. et al. (2018) Interrogating marine virus–host inter- 61. Musat, N. et al. (2012) Detecting metabolic activities in single
actions and elemental transfer with BONCAT and nanoSIMS- cells, with emphasis on nanoSIMS. FEMS Microbiol. Rev. 36,
based methods. Environ. Microbiol. 20, 671–692 486–511
40. Worrich, A. et al. (2017) Mycelium-mediated transfer of water 62. Saka, S.K. et al. (2014) Correlated optical and isotopic
and nutrients stimulates bacterial activity in dry and oligotrophic nanoscopy. Nat. Commun. 5, 3664
environments. Nat. Commun. 8, 15472 63. Lau, K.H. et al. (2010) Development of a new bimodal imaging
41. Greenwood, D.J. et al. (2019) Subcellular antibiotic visualization re- methodology: a combination of fluorescence microscopy and
veals a dynamic drug reservoir in infected macrophages. Science high-resolution secondary ion mass spectrometry. J. Microsc.
364, 1279–1282 240, 21–31
42. Vanbellingen, Q.P. et al. (2015) Time-of-flight secondary ion 64. Vreja, I.C. et al. (2015) Secondary-ion mass spectrometry of
mass spectrometry imaging of biological samples with delayed genetically encoded targets. Angew. Chem. Int. Ed. Engl. 54,
extraction for high mass and high spatial resolutions. Rapid 5784–5788
Commun. Mass Spectrom. 29, 1187–1195 65. Kabatas, S. et al. (2019) Fluorinated nanobodies for targeted
43. Benettoni, P. et al. (2019) Identification of nanoparticles and their molecular imaging of biological samples using nanoscale
localization in algal biofilm by 3D-imaging secondary ion mass secondary ion mass spectrometry. J. Anal. At. Spectrom. 34,
spectrometry. J. Anal. At. Spectrom. 34, 1098–1108 1083–1087
44. Henss, A. et al. (2018) High resolution imaging and 3D analysis 66. Thiery-Lavenant, G. et al. (2014) Detection of immunolabels with
of Ag nanoparticles in cells with ToF-SIMS and delayed extrac- multi-isotope imaging mass spectrometry. Surf. Interface Anal.
tion. Biointerphases 13, 03B410 46, 147–149
45. Raina, J.-B. et al. (2017) Subcellular tracking reveals the location 67. Angelo, M. et al. (2014) Multiplexed ion beam imaging of human
of dimethylsulfoniopropionate in microalgae and visualises its up- breast tumors. Nat. Med. 20, 436–442
take by marine bacteria. Elife 6, e23008 68. Kabatas, S. et al. (2019) Boron-containing probes for non-
46. Dowlatshahi Pour, M. et al. (2019) Mass spectrometry imaging optical high-resolution imaging of biological samples. Angew.
as a novel approach to measure hippocampal zinc. J. Anal. At. Chem. Int. Ed. Engl. 58, 3438–3443
Spectrom. 34, 1581–1587 69. Ismagulova, T. et al. (2018) A new simple method for quantifica-
47. Osorio, J.H.M. et al. (2019) Investigation of architecture tion and locating P and N reserves in microalgal cells based on
development and phosphate distribution in Chlorella biofilm energy-filtered transmission electron microscopy (EFTEM) ele-
by complementary microscopy techniques. FEMS Microbiol. mental maps. PLoS One 13, e0208830
Ecol. 95, fiz029 70. Yang, Y. et al. (2019) Three-dimensional correlative imaging of a
48. Sämfors, S. et al. (2019) Localised lipid accumulation detected in malaria-infected cell with a hard X-ray nanoprobe. Anal. Chem.
infarcted mouse heart tissue using ToF-SIMS. Int. J. Mass 91, 6549–6554
Spectrom. 437, 77–86 71. Dierolf, M. et al. (2010) Ptychographic X-ray computed tomogra-
49. Passarelli, M.K. et al. (2013) Single-cell lipidomics: characterizing phy at the nanoscale. Nature 467, 436–439
and imaging lipids on the surface of individual Aplysia californica 72. Victor, T.W. et al. (2018) X-ray fluorescence nanotomography
neurons with cluster secondary ion mass spectrometry. Anal. of single bacteria with a sub-15 nm beam. Sci. Rep. 8,
Chem. 85, 2231–2238 13415
50. Moore, K.L. et al. (2014) Combined nanoSIMS and synchrotron 73. Wirtz, T. et al. (2019) Imaging and analytics on the helium ion
X-ray fluorescence reveal distinct cellular and subcellular microscope. Annu. Rev. Anal. Chemi. 12, 523–543
distribution patterns of trace elements in rice tissues. New 74. Baumeister, T.U.H. et al. (2019) Live single-cell metabolo-
Phytol. 201, 104–115 mics with matrix-free laser/desorption ionization mass spec-
51. Zhou, J. et al. (2011) Reproducibility and quantitation trometry to address microalgal physiology. Front. Plant Sci.
of amplicon sequencing-based detection. ISME J. 5, 10, 172
1303–1313 75. Passarelli, M.K. et al. (2017) The 3D OrbiSIMS – label-free meta-
52. Perrin, L. et al. (2015) Evaluation of sample preparation methods bolic imaging with subcellular lateral resolution and high mass-
for single cell quantitative elemental imaging using proton or resolving power. Nat. Methods 14, 1175–1183

Trends in Cell Biology, March 2020, Vol. 30 , No. 3 187


Trends in Cell Biology

76. Deng, J. et al. (2018) Correlative 3D X-ray fluorescence and 81. Sigal, Y.M. et al. (2018) Visualizing and discovering cellular struc-
ptychographic tomography of frozen-hydrated green algae. tures with super-resolution microscopy. Science 361, 880–887
Sci. Adv. 4, eaau4548 82. Ackerman, C.M. et al. (2017) Analytical methods for imaging
77. Deng, J. et al. (2019) The Velociprobe: an ultrafast hard X-ray metals in biology: from transition metal metabolism to transition
nanoprobe for high-resolution ptychographic imaging. Rev. Sci. metal signaling. Anal. Chem. 89, 22–41
Instrum. 90 83. Bernhardt, M. et al. (2018) Correlative microscopy approach for
78. Gibbin, E. et al. (2018) Using nanoSIMS coupled with biology using X-ray holography, X-ray scanning diffraction and
microfluidics to visualize the early stages of coral infection by STED microscopy. Nat. Commun. 9, 3641
Vibrio coralliilyticus. BMC Microbiol. 18, 39 84. Jiang, H. et al. (2017) High-resolution sub-cellular imaging
79. Kontziampasis, D. et al. (2019) A cryo-EM grid preparation by correlative nanoSIMS and electron microscopy of amioda-
device for time-resolved structural studies. IUCrJ 6, 1024–1031 rone internalisation by lung macrophages as evidence
80. Heiligenstein, X. et al. (2014) The CryoCapsule: simplifying for drug-induced phospholipidosis. Chem. Commun. 53,
correlative light to electron microscopy. Traffic 15, 700–716 1506–1509

188 Trends in Cell Biology, March 2020, Vol. 30 , No. 3


Cell Metabolism

Review

GPIHBP1 and Lipoprotein Lipase,


Partners in Plasma Triglyceride Metabolism
Stephen G. Young,1,2,* Loren G. Fong,1,* Anne P. Beigneux,1 Christopher M. Allan,1 Cuiwen He,1 Haibo Jiang,1,3
Katsuyuki Nakajima,4 Muthuraman Meiyappan,5 Gabriel Birrane,6 and Michael Ploug7,8,*
1Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
2Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
3School of Molecular Sciences, University of Western Australia, Crawley 6009, Australia
4Department of Clinical Laboratory Medicine, Gunma University Graduate School of Department of Medicine, Maebashi,

Gunma 371-0805, Japan


5Discovery Therapeutics, Takeda Pharmaceutical Company Ltd., Cambridge, MA 02142, USA
6Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
7Finsen Laboratory, Rigshospitalet, Copenhagen DK–2200, Denmark
8Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen DK–2200, Denmark

*Correspondence: sgyoung@mednet.ucla.edu (S.G.Y.), lfong@mednet.ucla.edu (L.G.F.), m-ploug@finsenlab.dk (M.P.)


https://doi.org/10.1016/j.cmet.2019.05.023

Lipoprotein lipase (LPL), identified in the 1950s, has been studied intensively by biochemists, physiologists,
and clinical investigators. These efforts uncovered a central role for LPL in plasma triglyceride metabolism
and identified LPL mutations as a cause of hypertriglyceridemia. By the 1990s, with an outline for plasma
triglyceride metabolism established, interest in triglyceride metabolism waned. In recent years, however, in-
terest in plasma triglyceride metabolism has awakened, in part because of the discovery of new molecules
governing triglyceride metabolism. One such protein—and the focus of this review—is GPIHBP1, a protein of
capillary endothelial cells. GPIHBP1 is LPL’s essential partner: it binds LPL and transports it to the capillary
lumen; it is essential for lipoprotein margination along capillaries, allowing lipolysis to proceed; and it pre-
serves LPL’s structure and activity. Recently, GPIHBP1 was the key to solving the structure of LPL. These
developments have transformed the models for intravascular triglyceride metabolism.

The identification of lipoprotein lipase (LPL) as an intravascular crona et al., 1985; Osborne et al., 1985; Peterson et al., 2002;
triglyceride hydrolase (Korn, 1955; Korn and Quigley, 1955) Vannier and Ailhaud, 1989; Wong et al., 1994, 1997). The cloning
was a momentous discovery, one that suggested a mechanism of the LPL cDNA in the 1980s (Enerba €ck et al., 1987; Kirchgess-
by which triglycerides in the bloodstream are converted to fatty ner et al., 1987; Wion et al., 1987) triggered studies on LPL
acids for utilization by vital tissues (Havel, 2010). This discovery sequences required for substrate binding, catalytic activity,
triggered interest in triglyceride metabolism by scientists around and heparin binding (Emmerich et al., 1992; Lookene et al.,
the globe. Soon thereafter, a deficiency of LPL was shown to 1996, 1997a, 1997b; Ma et al., 1994; Sendak et al., 1998), and
cause familial chylomicronemia (Havel and Gordon, 1960), the also opened the door to understanding LPL mutations underly-
first example of an inborn error in lipoprotein metabolism (Havel, ing the familial chylomicronemia syndrome (Mailly et al., 1997;
2010). LPL-mediated processing of triglyceride-rich lipoproteins Peterson et al., 2002). Together, these efforts created an outline
was shown to be important for utilization of dietary lipids, for for intravascular lipoprotein processing—one that was taught to
generating atherogenic remnant lipoproteins, and for contrib- a generation of students and practicing physicians. The outline
uting to the biogenesis of high-density lipoproteins (HDLs) changed little over several decades, contributing to a percep-
(Havel, 2010; Havel and Kane, 2001; Rader, 2006; Rye and tion, occasionally voiced at conferences during the 1990s, that
Barter, 2014). The observation that LPL could be released into the field of plasma triglyceride metabolism was approaching
the bloodstream with an injection of heparin (Korn, 1955), com- maturity.
bined with the fact that LPL binds to heparan sulfate proteogly- For the scientists who were actively working on plasma triglyc-
cans (HSPGs) in vitro (Lookene et al., 1997b; Simionescu and eride metabolism, any suggestion that the field was close to
Simionescu, 1986; Vijayagopal et al., 1980), led to the proposal maturity would have made little sense. After all, no one under-
that the LPL inside blood vessels is bound to HSPGs within the stood how LPL, an enzyme that is secreted by adipocytes and
glycocalyx lining of blood vessels (Blanchette-Mackie et al., myocytes, reached its site of action in the capillary lumen; the
1989; Olivecrona and Bengtsson-Olivecrona, 1993). LPL was regulation of LPL activity was not understood; the structure of
shown to be activated by apolipoprotein (apo) CII (Bengtsson LPL remained elusive; and many cases of chylomicronemia,
and Olivecrona, 1979; Breckenridge et al., 1978; Havel et al., both genetic and acquired, were unexplained. Also, some fea-
1970; LaRosa et al., 1970), and several lines of evidence sug- tures within the outline of triglyceride metabolism needed more
gested that LPL is active as a homodimer (Garfinkel et al., investigation. For example, it was unclear why the LPL secreted
1983; Iverius and Ostlund-Lindqvist, 1976; Kobayashi et al., by parenchymal cells would end up being attached to HSPGs in
2002; Lookene and Bengtsson-Olivecrona, 1993; Lookene the lumen of blood vessels, given that HSPGs are abundant
et al., 1994; Olivecrona and Bengtsson-Olivecrona, 1990; Olive- within the interstitial spaces around parenchymal cells. Also,

Cell Metabolism 30, July 2, 2019 ª 2019 Elsevier Inc. 51


Cell Metabolism

Review

Figure 1. A Cartoon Representation of GPIHBP1’s Functions in Lipoprotein Lipase (LPL)-Mediated Lipoprotein Processing
GPIHBP1 serves four important functions in plasma triglyceride processing. The first two are illustrated in the left panel. First, GPIHBP1 on the abluminal plasma
membrane of capillary endothelial cells captures LPL (yellow) from heparin-sulfate proteoglycan (HSPG)-binding sites (green, tree-like structures) within the
subendothelial spaces (Davies et al., 2010; Kristensen et al., 2018). GPIHBP1’s intrinsically disordered acidic domain (green), which projects from GPIHBP1’s
three-fingered LU domain (blue), is important for capturing LPL and bringing it into high-affinity interactions with GPIHBP1’s LU domain (Kristensen et al., 2018).
Second, GPIHBP1 stabilizes the structure of LPL and protects it from unfolding, even in the face of physiologic inhibitor proteins such as ANGPTL4 (Kristensen
et al., 2018; Mysling et al., 2016a, 2016b). The preservation of LPL structure is illustrated by changing the color of LPL from yellow to purple. The middle panel
depicts the third function of GPIHBP1 in LPL biology, which is to transport LPL to its site of action in the capillary lumen (Davies et al., 2010). The movement of
GPIHBP1 across endothelial cells is bidirectional (Davies et al., 2012); thus, GPIHBP1 is able to return to the abluminal surface of endothelial cells, pick up more
LPL, and then replenish LPL stores along the capillary lumen. The right panel depicts the fourth function of GPIHBP1, which is to mediate the margination of
triglyceride-rich lipoproteins (TRLs) along the luminal surface of capillaries. GPIHBP1-bound LPL is required for TRL margination (Goulbourne et al., 2014),
allowing triglyceride hydrolysis to proceed.

the idea that LPL is only active as a homodimer needed more negatively charged domain at the amino terminus of GPIHBP1)
study. Finally, the mechanisms by which specific apolipopro- (Gin et al., 2008, 2011).
teins affected the efficiency of triglyceride hydrolysis were The first clue that GPIHBP1 was important for plasma triglyc-
incompletely understood. eride metabolism was finding severe hypertriglyceridemia in
Over the past decade, there has been considerable progress Gpihbp1 knockout mice (Gpihbp1 ⁄ ) (Beigneux et al., 2007;
in understanding plasma triglyceride metabolism. Much of that Weinstein et al., 2010a, 2010b). That discovery, combined with
progress has centered around GPIHBP1, a GPI-anchored pro- the finding that GPIHBP1 binds LPL and is expressed by endo-
tein of capillary endothelial cells (Beigneux et al., 2007). thelial cells, suggested that GPIHBP1 could be the binding site
GPIHBP1 is a dedicated partner for LPL: it captures LPL from for LPL on endothelial cells (i.e., the ‘‘platform’’ for LPL-mediated
within the subendothelial spaces and shuttles it to the capillary lipoprotein processing) (Beigneux et al., 2007).
lumen (Davies et al., 2012, 2010); it is essential for the margin- Subsequent studies showed that GPIHBP1’s function is more
ation of lipoproteins along capillaries, allowing LPL-mediated tri- substantial than simply serving as a binding site for LPL within
glyceride processing to proceed (Goulbourne et al., 2014); and it capillaries. GPIHBP1 is a partner for LPL in four ways (Figure 1).
stabilizes the structure and catalytic activity of LPL (Mysling First, the GPIHBP1 on the abluminal surface of capillary
et al., 2016a, 2016b). Recently, GPIHBP1 proved to be crucial endothelial cells is important for capturing LPL from within
for solving the structure of LPL (Birrane et al., 2019; Horton, the subendothelial spaces (Davies et al., 2010; Kristensen
2019; Arora et al., 2019). Here, we review recent insights into et al., 2018). Second, GPIHBP1 binding stabilizes the structure
the functions of GPIHBP1 and LPL in intravascular lipolysis, and activity of LPL (Kristensen et al., 2018; Mysling et al.,
including insights derived from the structures of those proteins. 2016a, 2016b). Third, GPIHBP1 shuttles LPL across capillary
endothelial cells to its site of action in the capillary lumen (Davies
GPIHBP1 and Plasma Triglyceride Metabolism et al., 2010). Fourth, GPIHBP1-bound LPL is critical for the
GPIHBP1 was initially identified during a screen of a mouse liver margination of lipoproteins in the bloodstream (Goulbourne
cDNA library for clones that render CHO cells capable of binding et al., 2014), allowing LPL-mediated lipoprotein processing to
HDL (Ioka et al., 2003). The screen uncovered SR-B1, long proceed.
recognized as an HDL-binding protein (Acton et al., 1996), and GPIHBP1’s function in transporting LPL across capillaries was
GPIHBP1, a GPI-anchored Ly6/uPAR (LU) protein with a long evident from the distribution of LPL in the tissues of wild-type
stretch of negatively charged amino acids at its amino terminus and Gpihbp1 ⁄ mice (Davies et al., 2010). In wild-type mice,
(Ioka et al., 2003). To this day, it is unclear why GPIHBP1 ap- most of the LPL in tissues is bound to GPIHBP1 on capillary
peared in that screen, given that Gin and colleagues found little endothelial cells (Figure 2A), where it is distributed evenly be-
evidence of HDL, apo-AI, or apo-E binding to GPIHBP1-ex- tween the luminal and abluminal plasma membranes (Davies
pressing cells (Gin et al., 2011). Our best guess is that the HDL et al., 2012, 2010). In Gpihbp1 ⁄ mice, LPL never reaches the
used in the initial screen contained small amounts of LPL (which capillary lumen (Davies et al., 2010). Instead, the LPL in
binds to GPIHBP1 avidly) or was enriched in apo-AV (a ‘‘heparin- Gpihbp1 ⁄ mice remains stranded within the interstitial spaces,
binding’’ apolipoprotein that has been reported to bind to the presumably bound to HSPGs in close proximity to the surface of

52 Cell Metabolism 30, July 2, 2019


Cell Metabolism

Review
Figure 2. Localization of GPIHBP1 and
Lipoprotein Lipase (LPL) in Brown Adipose
Tissue (BAT)
(A) Mislocalization of LPL in the BAT of Gpihbp1 ⁄
mice. Sections of BAT from a Gpihbp1+/+ and
Gpihbp1 ⁄ mouse were stained with antibodies
against CD31 (green), GPIHBP1 (magenta), and
LPL (red), and imaged by confocal fluorescence
microscopy. DNA was stained with DAPI (blue).
Scale bar, 50 mm.
(B) GPIHBP1 is expressed in endothelial cells of
capillaries, but not in endothelial cells of larger
blood vessels. Sections of BAT were stained with
antibodies against CD31 (green) and GPIHBP1
(red). GPIHBP1 expression in endothelial cells
decreases as capillaries merge into a venule
(arrow). Scale bar, 50 mm.

GPIHBP1’s transport function was


established by studies showing that
GPIHBP1 transports a GPIHBP1-specific
monoclonal antibody from one side of an
endothelial cell to the other (Davies
et al., 2010). The movement of GPIHBP1
across endothelial cells is bidirectional
(Davies et al., 2012). When wild-type
mice are injected intravenously with a flu-
cells (both parenchymal cells and capillary endothelial cells) orescently labeled GPIHBP1-specific monoclonal antibody, the
(Davies et al., 2010) (Figure 2A). antibody initially binds to GPIHBP1 within the capillary lumen
By immunohistochemistry, GPIHBP1 is expressed solely in but then quickly appears on the abluminal surface of capillaries.
capillary endothelial cells and is absent in endothelial cells of ve- Conversely, when the antibody is injected into tissues, it first
nules, arterioles, and larger blood vessels (Beigneux et al., 2007; associates with GPIHBP1 on the outside of capillaries but then
Davies et al., 2010) (Figure 2B). GPIHBP1 is found in capillaries quickly appears in the capillary lumen. Antibody transport did
of all peripheral tissues, with particularly high levels in the heart not occur in Gpihbp1 ⁄ mice. GPIHBP1 appears to be a long-
and adipose tissue (where LPL-mediated processing of lipopro- lived protein (Olafsen et al., 2010), implying that GPIHBP1 could
teins is robust), but it is absent from capillaries of the brain make multiple trips across endothelial cells, with each trip
(Beigneux, 2010; Beigneux et al., 2007; Davies et al., 2010; Olafsen providing an opportunity to capture LPL and bring it to the capil-
et al., 2010), which uses glucose for fuel (Mergenthaler et al., 2013). lary lumen.
Consistent with immunohistochemistry studies, Gpihbp1 tran- The intravascular processing of triglyceride-rich lipoproteins
scripts are absent from capillary endothelial cells of the brain, as by LPL requires that the lipoproteins in the bloodstream stop
judged by single-cell RNA sequencing (RNA-seq) studies (He (marginate) along capillaries. For years, lipoprotein margination
et al., 2018b; Vanlandewijck et al., 2018). Interestingly, high levels was assumed to involve electrostatic interactions between
of GPIHBP1 expression are observed in capillaries of the lung, a positively charged apolipoproteins on lipoproteins and the nega-
tissue where LPL expression is low (Olafsen et al., 2010). GPIHBP1 tively charged HSPGs that line the luminal surface of endothelial
in lung capillaries appears to play a role in scavenging LPL that cells (de Beer et al., 1999; Lookene et al., 1997b; Wang and
escapes into the plasma compartment (Goulbourne et al., 2014), Eckel, 2009). More recent studies revealed that lipoprotein
but the physiologic importance of that role is unclear. GPIHBP1 margination is utterly dependent on GPIHBP1—and more spe-
deficiency does not appear to affect pulmonary function. cifically on the GPIHBP1-LPL complex (Goulbourne et al.,
Why GPIHBP1 expression is restricted to the endothelial cells 2014). GPIHBP1 expression alone is not sufficient for lipoprotein
of the capillaries of peripheral tissues and why GPIHBP1 is ex- margination along capillaries; lipoprotein margination is negli-
pressed at higher levels in some tissues than others are poorly gible in lung capillaries, where GPIHBP1 is abundant but LPL
understood. An enhancer element was recently identified expression is low. However, when the GPIHBP1 in lung capil-
3.6 kb upstream of exon 1 of Gpihbp1 (Allan et al., 2019). De- laries was artificially loaded with exogenous bovine LPL, lipopro-
leting that enhancer reduced Gpihbp1 transcripts in heart and tein margination along lung capillaries was robust (Goulbourne
brown adipose tissue by 50% but nearly eliminated transcripts et al., 2014).
in liver and kidney (Allan et al., 2019). Fluorescently labeled lipoproteins, when injected intrave-
By immunohistochemistry, the tissue distribution of LPL in pe- nously into wild-type mice, rapidly marginate along capillaries
ripheral tissues mirrors that of GPIHBP1. Even though LPL is of the heart, co-localizing with LPL and GPIHBP1. In contrast,
synthesized and secreted by parenchymal cells, the vast major- lipoprotein margination is negligible along capillaries in hearts
ity of the LPL in tissues is bound to GPIHBP1 on capillary endo- of Gpihbp1 ⁄ mice, which are devoid of GPIHBP1-LPL com-
thelial cells (Beigneux et al., 2007; Davies et al., 2010). plexes (Goulbourne et al., 2014). In vitro studies indicated that

Cell Metabolism 30, July 2, 2019 53


Cell Metabolism

Review
Figure 3. NanoSIMS Imaging to Examine
Intravascular Lipolysis
(A) 2H/1H ratio image of a capillary in a mouse heart
2 min after an intravenous injection of 2H-TRLs
(triglyceride-rich lipoproteins enriched in 2H-tri-
glycerides). The 2H/1H image shows 2H-TRLs that
have marginated along the luminal surface of capil-
lary endothelial cells and entry of 2H-enriched lipids
into endothelial cells as well as the mitochondria and
lipid droplets of cardiomyocytes. The 2H/1H ratio is
multiplied by 10,000; the range spans from slightly
higher than 2H natural abundance to approximately
six times natural abundance. Scale bar, 1 mm.
(B) Composite 12C14N and 32S /12C14N images of
mouse heart capillaries. The signal from 12C14N
secondary ions (blue), reflecting 14N distribution,
was robust in both capillary endothelial cells (EC)
and cardiomyocytes. The 32S /12C14N ratio (green)
was set from 0.05 to 0.06. Pixels with a 32S /12C14N
ratio above 0.05 are shown, revealing features rich
in 32S, relative to 14N, on the surface of endothelial
cells and cardiomyocytes. Scale bar, 1 mm.

lipoprotein binding to the GPIHBP1-LPL complex depends on a Properties of GPIHBP1 Required for LPL Binding
tryptophan-rich loop within the carboxy-terminal domain of LPL Mature GPIHBP1 contains two key domains, a disordered
(Goulbourne et al., 2014). amino-terminal acidic domain and an LU domain containing 10
Because the GPIHBP1-LPL complex is anchored to the cysteines, all arranged in a characteristic spacing pattern and
plasma membrane of endothelial cells and because the luminal all disulfide bonded (Beigneux et al., 2011; Mysling et al.,
surface of endothelial cells is covered by an HSPG-rich glycoca- 2016a). The five disulfide bonds stabilize the assembly of three
lyx, it was initially unclear how the GPIHBP1-LPL complex would loops and form the hallmark three-fingered scaffold of the LU
be able to participate in lipoprotein margination. However, elec- domain (Leth et al., 2019). The acidic domain of human GPIHBP1
tron microscopy (EM) studies revealed that the glycocalyx on the is impressive, with 21 acidic amino acids (glutamates or aspar-
surface of capillaries is patchy, with tufts of glycocalyx interrup- tates) in 26 consecutive residues. A sulfated tyrosine, located
ted by ‘‘meadows’’ where the plasma membrane is exposed in the middle of the acidic domain (Kristensen et al., 2018),
(Goulbourne et al., 2014). EM tomography suggested that lipo- adds to the negative charge. The GPIHBP1 in every mammalian
proteins bind to capillaries within the gaps in the glycocalyx species contains an acidic domain, but the precise sequence
(Goulbourne et al., 2014). and length of the domain is variable. For example, the acidic
domain in opossum GPIHBP1 contains 32 aspartates or gluta-
Relevance of GPIHBP1 Expression to Uptake of mates in 39 consecutive residues, including a stretch of 23
Lipoprotein-Derived Nutrients by Tissues consecutive aspartates (Fong et al., 2016). Like all GPI-anchored
The turnover of triglyceride-rich lipoproteins in the bloodstream proteins, GPIHBP1 contains a carboxy-terminal hydrophobic
is quite rapid (Havel, 2010). Recent NanoSIMS imaging studies signal peptide that triggers the addition of a GPI anchor within
revealed that the movement of the fatty acid products of lipolysis the endoplasmic reticulum (Ferguson et al., 2009).
across endothelial cells and into surrounding parenchymal cells GPIHBP1’s LU domain is required for the stability of LPL bind-
is also rapid (He et al., 2018a). After administering [2H]triglycer- ing, contributing to a long half-life for the GPIHBP1-LPL complex
ide-enriched lipoproteins (2H-TRLs) to wild-type mice by an (t½  35 s). When GPIHBP1’s LU domain is replaced with an LU
intravenous injection, it was possible to visualize the margination domain from another member of the LU family (Gin et al., 2008) or
of 2H-TRLs in heart capillaries as early as 30 s after the injection when any of the 10 cysteines in GPIHBP1’s LU domain is
(He et al., 2018a). As early as 2 min after the injection, substantial mutated (Beigneux et al., 2009b), stable interactions with LPL
amounts of deuterium had already entered the mitochondria and are abolished. Aside from the 10 cysteines, alanine-scanning
cytosolic lipid droplets of cardiomyocytes (He et al., 2018a) mutagenesis of GPIHBP1’s LU domain uncovered 12 additional
(Figure 3A). Interestingly, deuterium enrichment in capillary residues required for LPL binding, with the majority located in
endothelial cells and cardiomyocytes was similar (i.e., there conserved segments within the second finger of the LU domain
was little evidence that endothelial cells play a gatekeeper role (amino acids 89–110) (Beigneux et al., 2011). Many mutations in
in the movement of fatty acids into cardiomyocytes). When iso- GPIHBP1’s LU domain interfered with disulfide bonding and pro-
lated hearts from wild-type mice were perfused with 2H-TRLs, moted the formation of disulfide-linked GPIHBP1 dimers and
similar findings were observed—rapid margination of 2H-TRLs multimers (Beigneux et al., 2015; Plengpanich et al., 2014), but
along capillaries and rapid uptake of fatty acids into cytosolic there was one noteworthy exception. Mutations in GPIHBP1
lipid droplets (He et al., 2018a). When isolated hearts from Trp-109 abolished LPL binding without interfering with disulfide
Gpihbp1 ⁄ mice were perfused with 2H-TRLs, the margination bonding or promoting protein multimerization, suggesting that
of 2H-TRLs was absent and 2H enrichment in the cytosolic tri- Trp-109 plays a direct role in the GPIHBP1-LPL binding interface
glyceride droplets of cardiomyocytes was markedly reduced (Beigneux et al., 2015). Consistent with that idea, hydrogen-
(He et al., 2018a). deuterium exchange mass spectrometry (HDX-MS) studies

54 Cell Metabolism 30, July 2, 2019


Cell Metabolism

Review

showed that the binding of LPL to GPIHBP1 protects GPIHBP1 2018). Finally, they documented, by SPR, large differences in
residues 104–128 from solvent exposure and reduces deuterium the abilities of full-length GPIHBP1 and Dacidic-GPIHBP1 to
uptake into GPIHBP1 (Mysling et al., 2016a). dislodge LPL when it was bound to a sensor chip containing im-
Studies by Gin and coworkers (Gin et al., 2012) revealed that mobilized short-chain heparin moieties. Full-length GPIHBP1
GPIHBP1 binds to the carboxy-terminal domain of LPL (residues dislodged heparin-bound LPL efficiently, whereas Dacidic-
325–475). Consistent with those findings, an LPL-specific mono- GPIHBP1 did not (despite the fact that Dacidic-GPIHBP1 was
clonal antibody with an epitope within the carboxy-terminal able to bind to the LPL) (Kristensen et al., 2018).
domain of LPL, 88B8, blocks LPL binding to GPIHBP1 (Allan The fact that full-length GPIHBP1 effectively captures LPL
et al., 2016). Also, two different missense mutations within the from heparin is probably relevant to LPL trafficking to capillaries.
carboxy-terminal domain of LPL, C445Y and E448K, first identi- As noted earlier, the LPL in Gpihbp1 ⁄ mice is stranded within
fied in patients with severe hypertriglyceridemia (Henderson the interstitial spaces, bound to HSPGs near the surface of cells
et al., 1996, 1998), abolish LPL binding to GPIHBP1 (Voss (Davies et al., 2010). The interactions between LPL and HSPGs
et al., 2011). Finally, HDX-MS studies revealed that when within the interstitium are transient (i.e., with a high koff), but the
GPIHBP1 is bound to LPL, residues 429–446 in LPL’s carboxy- abundance of HSPG-binding sites means that LPL in Gpihbp1 ⁄
terminal domain are protected from solvent exposure and deute- tissues is retained within the interstitium rather than escaping
rium uptake, implying that those residues are involved in into the lymphatic circulation. The same interstitial HSPG-bind-
GPIHBP1 binding (Mysling et al., 2016a). ing sites exist in wild-type mice; however, the dynamic nature
of LPL-HSPG interactions allows LPL to move to more stable
Deciphering Functions for GPIHBP1’s Intrinsically GPIHBP1-binding sites on capillaries. The ability of GPIHBP1
Disordered Acidic Domain to recruit LPL away from interstitial HSPG-binding sites has
To explore the contributions of GPIHBP1’s LU and acidic been documented experimentally. When GPIHBP1-coated
domains to the formation of the GPIHBP1-LPL complex, the ki- agarose beads were implanted into the brown adipose tissue
netics of LPL binding to full-length GPIHBP1 and ‘‘Dacidic- of Gpihbp1 ⁄ mice, HSPG-bound LPL within the interstitial
GPIHBP1’’ (a GPIHBP1 mutant lacking the amino-terminal acidic spaces moved to the GPIHBP1-coated beads, depleting LPL
domain) were analyzed by surface plasmon resonance (SPR). In stores within the interstitium (Allan et al., 2017b).
these studies, the two different GPIHBP1 proteins were passed Recent immunohistochemical studies on LPL localization in
over a sensor chip coated with LPL (which had been tethered tissues of Gpihbp1 ⁄ mice yielded further insights into the traf-
to the sensor chip with the LPL-specific monoclonal antibody ficking of LPL within the interstitium (Kristensen et al., 2018). As
5D2) (Kristensen et al., 2018; Mysling et al., 2016a). Both full- expected, the LPL in the heart and skeletal muscle of Gpihbp1 ⁄
length GPIHBP1 and Dacidic-GPIHBP1 bound LPL with nano- mice was mislocalized to the interstitium. Interestingly, however,
molar affinities, but the binding of full-length GPIHBP1 was the amount of LPL adjacent to endothelial cells was greater than
stronger. Interestingly, the stability of the GPIHBP1-LPL the amount adjacent to myocytes (Kristensen et al., 2018). Thus,
complex, as judged by the dissociation rate constant (koff), was even in the absence of GPIHBP1, the LPL within the interstitium
nearly identical for full-length GPIHBP1 and Dacidic-GPIHBP1 appears to associate preferentially with HSPGs on the outside of
(2 3 10 2 s 1) (Mysling et al., 2016a). However, the association capillary endothelial cells. We propose that ‘‘directed diffusion’’
rate constants (kon) for Dacidic-GPIHBP1 (0.1 3 106 M 1s 1) (Duchesne et al., 2012) along an HSPG electrostatic gradient is
and full-length GPIHBP1 (2.6 3 107 M 1s 1) were very different, responsible for the flow of LPL from the HSPGs near myocytes
highlighting a key role for GPIHBP1’s acidic domain in to HSPGs on the outer surface of endothelial cells. The same
accelerating the encounter rate and promoting the formation directional LPL movement presumably exists in tissues of wild-
of the GPIHBP1-LPL complex (Kristensen et al., 2018). Further- type mice, generating a pool of LPL for capture by GPIHBP1
more, the kon for full-length GPIHBP1-LPL binding was sensitive on endothelial cells. Small-angle X-ray scattering (SAXS) studies
to the ionic strength of the buffer and was as high as showed that GPIHBP1’s disordered acidic domain, as it projects
3 3 108 M 1s 1 under physiological conditions, underscoring from the LU domain, occupies a large mushroom-shaped space
the importance of electrostatic steering in the initial interaction with a diameter of 112 Å (Kristensen et al., 2018). In tissues,
between LPL and full-length GPIHBP1 (Kristensen et al., 2018). GPIHBP1’s acidic domain likely projects at least 100 Å from
Under physiologic conditions, GPIHBP1’s disordered acidic the basolateral plasma membrane of endothelial cells and prob-
domain increases the velocity of LPL binding by >2,500-fold. ably functions in a fly casting-like mechanism (Shoemaker et al.,
These observations are likely relevant to the ability of GPIHBP1 2000) to engage the LPL that is bound in a dynamic fashion to
to capture LPL from within the interstitial spaces along the baso- HSPGs and drive it into a stable complex with GPIHBP1’s LU
lateral surface of capillaries. domain.
Kristensen and coworkers recently investigated GPIHBP1- The explanation for the preferential association of interstitial
LPL interactions in more detail (Kristensen et al., 2018). They es- LPL with endothelial cells in Gpihbp1 ⁄ mice is unknown, but
tablished that the stoichiometry of GPIHBP1 binding to LPL is 1:1 the existence of HSPGs on endothelial cells is well documented.
(Kristensen et al., 2018). Remarkably, a peptide corresponding Collagen XVIII, a basement membrane HSPG, is found on capil-
to GPIHBP1’s acidic domain also bound to LPL with 1:1 stoichi- lary endothelial cells, as judged by immunohistochemistry
ometry. They also found, by SPR, that the sulfated tyrosine (Tyr- (Bishop et al., 2010). Also, NanoSIMS analyses of 32S distribution
38) in GPIHBP1’s acidic domain contributes to the strength of in tissues appear consistent with the existence of HSPGs adja-
LPL binding. When Tyr-38 was replaced with Phe, the KD for cent to cardiomyocytes and endothelial cells (Figure 3B). 32S is
GPIHBP1-LPL binding increased 3-fold (Kristensen et al., found in methionines and cysteines of all cellular proteins, but

Cell Metabolism 30, July 2, 2019 55


Cell Metabolism

Review
Figure 4. Inherent Instability of LPL, as
Judged by HDX-MS Studies
Bovine LPL was incubated at 25 C in deuterium
oxide.
(A) The emergence of a bimodal isotope envelope
of deuterium uptake in a peptic peptide covering
the catalytic triad of LPL (from Mysling et al.,
2016a). Bimodality of the isotope envelope is a
hallmark of protein unfolding (Mysling et al., 2016a,
2016b). Substantial portions of the amino-terminal
catalytic domain of LPL (NTD) exhibited the
bimodal isotope envelope, while peptides from
the carboxy-terminal lipid-binding domain (CTD)
exhibit a unimodal exchange pattern, reflecting
greater stability.
(B) Position of the catalytic triad peptic peptide
(green) in the crystal structure of human LPL
(modified from the crystal structure by Birrane and
coworkers [Birrane et al., 2019]). The structure of
human LPL is shown as a cartoon representation,
with a helices in red and b strands in cyan.
The location of the three catalytic triad residues
(S159, D183, and H268) are noted. The structure
was visualized with PyMol (Schrödinger) using
PDB: 6E7K.

the amount of 32S (relative to 14N) is greater within the interstitial The spontaneous unfolding of LPL, as judged by HDX-MS
spaces near the surface of cardiomyocytes and endothelial cells studies, is more than an in vitro biochemical curiosity. In
than in the 14N-rich cytoplasm of those cells. Larger amounts follow-up studies, Mysling and coworkers (Mysling et al.,
of 32S, relative to 14N, are also observed in the capillary lumen, 2016b) showed that ANGPTL4, a physiologic inhibitor of LPL, in-
likely because of HSPGs in the glycocalyx of endothelial cells activates LPL by catalyzing LPL unfolding. Spontaneous unfold-
(Figure 3B). ing of LPL, as judged by HDX-MS, is relatively slow, with 7%
unfolding after 5 min at 25 C and 29% unfolding after 30 min.
GPIHBP1 Stabilizes the Structure and Activity of LPL, In contrast, incubating LPL with sub-stoichiometric amounts of
even in the Presence of Endogenous LPL Inhibitors ANGPTL4 (a 10:1 LPL to ANGPTL4 molar ratio) resulted in
For years, biochemists and physiologists have noted that cata- 70% unfolding of LPL in 5 min. Mysling and coworkers also
lytic activity disappears rapidly when purified preparations of tested the capacity of an ANGPTL4 polymorphic variant,
LPL are incubated at room temperature. HDX-MS studies by ANGPTL4E40K, to catalyze LPL unfolding (Mysling et al.,
Mysling and coworkers (Mysling et al., 2016a) provided a likely 2016b). The ANGPTL4E40K variant, present in 3% of Caucasians,
explanation. When LPL is incubated at 25 C with deuterated is associated with lower plasma triglyceride levels and reduced
water (2H2O), deuterium uptake into specific regions of LPL re- risk of coronary disease (Dewey et al., 2016; Helgadottir et al.,
sults in bimodal isotope envelopes, reflecting progressive un- 2016; Myocardial Infarction Genetics and CARDIoGRAM
folding of those regions (Figure 4A). Bimodal isotope envelopes Exome Consortia Investigators et al., 2016). The capacity of
were observed in peptides corresponding to LPL’s amino-termi- ANGPTL4E40K to catalyze LPL unfolding was 60% lower than
nal hydrolase domain, including in a peptide spanning LPL’s cat- that of wild-type ANGPTL4. Consistent with those findings,
alytic triad (Figure 4B), but they were nearly absent in peptides LPL incubated with ANGPTL4E40K retained 40%–50% more cat-
from LPL’s carboxy-terminal domain, which has a more stable alytic activity than LPL incubated with wild-type ANGPTL4. The
conformation. The appearance of bimodal deuterium signatures reduced capacity of ANGPTL4E40K to inactivate LPL, which is
in the amino-terminal domain was accompanied by a parallel fall almost certainly due to instability of a functionally important
in LPL catalytic activity (Mysling et al., 2016a). The binding of a-helix in ANGPTL4 (Mysling et al., 2016b), presumably explains
GPIHBP1 to LPL markedly reduced both the unfolding of LPL’s the reduced plasma triglyceride levels in ANGPTL4E40K carriers.
amino-terminal domain and the loss of catalytic activity, demon- The fact that LPL activity is regulated by an inhibitory protein
strating that GPIHBP1 preserves the structural integrity and cat- (i.e., ANGPTL4) that promotes unfolding is, to the best of our
alytic activity of LPL. Interestingly, the protective effect of knowledge, a novel mechanism for controlling the activity of an
GPIHBP1 was largely due to GPIHBP1’s intrinsically disordered enzyme. Mysling and colleagues (Mysling et al., 2016b) pro-
acidic domain (Mysling et al., 2016a). First, the preservation posed that ANGPTL4 interacts transiently with LPL and pro-
of LPL structure and catalytic activity was much greater with motes an unstable conformation that leads inexorably to enzyme
full-length GPIHBP1 than with Dacidic-GPIHBP1 (Mysling inactivation. Another group reported that inactivation of LPL by
et al., 2016a). Second, a synthetic peptide corresponding to ANGPTL4 is reversible when the inactivation of LPL is performed
GPIHBP1’s acidic domain reduced both LPL unfolding and the in the presence of deoxycholate (Gutgsell et al., 2019; Lafferty
loss of triglyceride hydrolase activity. et al., 2013), a detergent that stabilizes LPL activity (Bengtsson

56 Cell Metabolism 30, July 2, 2019


Cell Metabolism

Review

and Olivecrona, 1979). The significance of this finding is unclear, mia was caused by the introduction of an unpaired cysteine into
in part because the relevance of deoxycholate-stabilized LPL to the LU domain (S107C) (Plengpanich et al., 2014). Chylomicro-
the normal physiology of intravascular lipolysis is open to ques- nemia was also observed in the setting of a T80K mutation,
tion. Also, the LPL activity in those studies was assessed with a which interferes with N-linked glycosylation and trafficking of
soluble substrate rather than with an emulsified triglyceride sub- GPIHBP1 to the plasma membrane (Ariza et al., 2016). A
strate. Finally, the ANGPTL4 proved to be a relatively inefficient G175R substitution has been observed in several patients with
inhibitor under the conditions that were studied, with a Ki of hypertriglyceridemia (Beigneux et al., 2017; Charrière et al.,
0.86–1.7 mM (Gutgsell et al., 2019; Lafferty et al., 2013). 2011; Chyzhyk et al., 2019) but is unlikely to be causative.
In the studies by Mysling and coworkers, full-length GPIHBP1 G175 is located in the signal peptide that is normally replaced
protected LPL from ANGPTL4-mediated unfolding, but there by the GPI anchor (i.e., it is not present in mature GPIHBP1).
was little protection by Dacidic-GPIHBP1 or an acidic domain The G175R substitution has no significant effect on the addition
peptide (Mysling et al., 2016b). Of note, the inactivation of LPL of the GPI anchor (Beigneux et al., 2017). No one has yet uncov-
by ANGPTL4 was not reversible; the loss of LPL activity with ered a GPIHBP1 mutation that results in the deletion of all or part
ANGPTL4 could not be reversed by adding GPIHBP1 to the incu- of GPIHBP1’s acidic domain.
bation mixture (Mysling et al., 2016b). Mysling and coworkers Low levels of LPL in the pre- and post-heparin plasma are a
proposed that the anchoring of GPIHBP1’s LU domain to LPL re- hallmark of GPIHBP1 deficiency and presumably reflect reduced
sults in optimal positioning of GPIHBP1’s acidic domain, allow- amounts of LPL within capillaries (Beigneux et al., 2009a; Olive-
ing it to stabilize LPL structure and activity (Mysling et al., crona et al., 2010; Plengpanich et al., 2014). Low levels of
2016b). From the standpoint of plasma triglyceride physiology, GPIHBP1 in the plasma are another hallmark (Beigneux et al.,
the ability of GPIHBP1 to preserve LPL structure and activity 2017). Heterozygous carriers of GPIHBP1 mutations have
(even in the presence of inhibitory proteins) probably serves to half-normal plasma levels of GPIHBP1 (Beigneux et al., 2017;
focus catalytically active LPL where it needs to be for lipoprotein Miyashita et al., 2018b).
processing (i.e., in the capillary lumen). It seems likely that at GPIHBP1 missense mutations causing chylomicronemia
least some of the regulation of LPL by ANGPTL4 occurs before abolish the capacity of GPIHBP1 to bind LPL (Franssen et al.,
LPL reaches the capillary lumen. Kersten and collaborators 2010; Olivecrona et al., 2010; Plengpanich et al., 2014). In cell
showed that ANGPTL4 can inactivate LPL in the secretory transfection studies, missense mutations involving cysteine
pathway of adipocytes (Dijk et al., 2016). Also, the LPL in the residues (e.g., C65Y, C65S, and C68G) interfere with disulfide
interstitial spaces, as it moves toward endothelial cells, could bonding and result in the appearance of disulfide-linked
be susceptible to inhibition by ANGPTL4. Nilsson and coworkers GPIHBP1 dimers and multimers on the surface of cells (Beigneux
suggested that the interstitial spaces may be a site for et al., 2015). The GPIHBP1 dimers and multimers have no capac-
ANGPTL4-mediated regulation of LPL (Nilsson et al., 2012). ity to bind LPL (Beigneux et al., 2015; Plengpanich et al., 2014).
ANGPTL3, another physiologic inhibitor of LPL, also catalyzes The fact that dimeric and multimeric GPIHBP1 proteins were
LPL unfolding, but it is less potent than ANGPTL4—at least un- able to reach the surface of the transfected cells was somewhat
der the experimental conditions that were tested (Mysling surprising, given that a cysteine mutation in CD59 (another LU
et al., 2016b). ANGPTL3-catalyzed LPL unfolding is also in- protein) abolished CD59 trafficking to the plasma membrane
hibited by the binding of GPIHBP1. In the case of ANGPTL3, (Nevo et al., 2013). Given that observation, it seemed possible
several studies have demonstrated that the functional inhibitory that the appearance of GPIHBP1 dimers and multimers on the
unit is actually a complex of ANGPTL3 and ANGPTL8 (Chi et al., surface of transfected cells was simply an artifact of protein
2015; Haller et al., 2017; Quagliarini et al., 2012) and that this overexpression. To explore that possibility, Allan and coworkers
complex is as efficient as ANGPTL4 in inactivating LPL (Kovrov (Allan et al., 2017a) created knockin mice harboring a cysteine
et al., 2019). substitution in the LU domain (C63Y, corresponding to a muta-
tion observed in humans; Franssen et al., 2010). Homozygotes
Familial Chylomicronemia from GPIHBP1 Mutations exhibited severe chylomicronemia, even on a low-fat chow
Soon after the discovery of chylomicronemia in Gpihbp1 ⁄ diet. GPIHBP1-C63Y reached the surface of endothelial cells
mice, GPIHBP1 mutations were identified in patients with familial and could be easily detectable by immunohistochemistry, but
chylomicronemia syndrome (Ariza et al., 2016; Beigneux et al., the amount was reduced by 70% (Allan et al., 2017a). No LPL
2009a; Charrière et al., 2011; Coca-Prieto et al., 2011; Franssen was bound to GPIHBP1-C63Y in the capillary lumen, demon-
et al., 2010; Gonzaga-Jauregui et al., 2014; Olivecrona et al., strating that the mutant protein had no capacity to bind LPL.
2010; Plengpanich et al., 2014; Rabacchi et al., 2016; Rios Interestingly, most of the GPIHBP1-C63Y on the surface of
et al., 2012; Yamamoto et al., 2013). The chylomicronemia that endothelial cells in vivo was monomeric; only small amounts of
results from GPIHBP1 mutations presents in infancy (Rios disulfide-linked dimers were detected. Thus, the capillary endo-
et al., 2012) and persists lifelong, with plasma triglycerides often thelial cells in vivo appear to be more efficient than transfected
exceeding 2,000 mg/dL. Two mutant GPIHBP1 alleles are CHO cells in removing misfolded GPIHBP1 proteins (including
required for disease; the plasma triglyceride levels in heterozy- dimers and multimers), thereby limiting the amounts of GPIHBP1
gotes are normal. A variety of different GPIHBP1 mutations that reach the surface of cells.
have been uncovered but the majority has involved one of the
cysteines in the LU domain (e.g., C65Y, C65S, C68Y, C68G, Chylomicronemia from GPIHBP1 Autoantibodies
C68R, C83R, and C89F) or a residue in close proximity to a Chylomicronemia occasionally appears spontaneously, and
cysteine (e.g., Q115P and T111P). In one kindred, chylomicrone- some of these ‘‘acquired’’ cases are caused by GPIHBP1

Cell Metabolism 30, July 2, 2019 57


Cell Metabolism

Review

autoantibodies (‘‘GPIHBP1 autoantibody syndrome’’) (Beigneux patient invariably has severe chylomicronemia. We have no
et al., 2017; Eguchi et al., 2019; Hu et al., 2017a; Kersten, 2017). evidence that GPIHBP1 autoantibodies explain cases of mild-
This disorder was discovered fortuitously while characterizing a to-moderate hypertriglyceridemia. In one patient, the chylomi-
monoclonal antibody-based ELISA designed to measure plasma cronemia resulting from GPIHBP1 autoantibodies resolved
levels of GPIHBP1 (Beigneux et al., 2017; Miyashita et al., spontaneously (Hu et al., 2017a). In two cases, the chylomicro-
2018a). In those studies, two plasma samples, both from pa- nemia resolved after initiating immunosuppressive treatment,
tients with chylomicronemia, had abnormally low GPIHBP1 and in one of those cases, the normalization of triglyceride levels
levels, and the GPIHBP1 levels remained low even after was accompanied by the disappearance of GPIHBP1 autoanti-
‘‘spiking’’ the plasma samples with recombinant GPIHBP1. The bodies (Beigneux et al., 2017). More experience is needed to
latter finding strongly suggested ‘‘immunoassay interference,’’ establish the natural history and treatment strategies for the
which can be caused by autoantibodies. Additional studies re- GPIHBP1 autoantibody syndrome.
vealed that the plasma from the two chylomicronemia patients
did indeed contain GPIHBP1 autoantibodies (Beigneux et al., A Crystal Structure for the GPIHBP1-LPL Complex
2017). Of note, the autoantibodies abolished the capacity of The structure of LPL remained elusive for decades, forcing in-
GPIHBP1 to bind LPL. Patients with the GPIHBP1 autoantibody vestigators to rely on a homology model (Kobayashi et al.,
syndrome invariably have low plasma levels of LPL, consistent 2002) derived from the structure of pancreatic lipase, a distantly
with the inability of GPIHBP1 to bind LPL and transport it to related lipase family member (Winkler et al., 1990). The absence
the capillary lumen (Beigneux et al., 2017). Subsequent studies of a structure for LPL was not for lack of trying; several labora-
revealed that GPIHBP1 autoantibodies are directed against tories attempted to generate LPL crystals, but none succeeded,
GPIHBP1’s LU domain, not the acidic domain (Kristensen likely because of the propensity of LPL to unfold (Mysling et al.,
et al., 2018). 2016a). Inspired by the discovery that GPIHBP1 stabilizes LPL
The plasma triglyceride levels in patients with the GPIHBP1 structure and activity, Birrane and coworkers solved the crystal
autoantibody syndrome are extremely high, typically exceeding structure of a GPIHBP1-LPL complex at 2.8-Å resolution (Birrane
1,500 mg/dL, and many of the patients had experienced an et al., 2019; Horton, 2019).
episode of acute pancreatitis. In some cases, GPIHBP1 autoan- The crystallographic unit contained two LPL-GPIHBP1 com-
tibodies were the sole manifestation of autoimmune disease, but plexes, with the two LPL molecules arranged in a head-to-tail
the majority of patients had clinical or serological evidence of an orientation, with the Trp-rich loop in LPL’s carboxy-terminal
autoimmune disease (e.g., systemic lupus erythematosus; SLE) domain interacting with the catalytic pocket in the LPL’s
(Beigneux et al., 2017). One patient with SLE and GPIHBP1 amino-terminal domain. The same conformation was observed
autoantibodies became pregnant and delivered a baby girl. for the complex in solution by SAXS studies (Birrane et al.,
The newborn infant manifested severe chylomicronemia that 2019). The structure of LPL contained an amino-terminal hydro-
resolved after several weeks, presumably coinciding with disap- lase domain with six a helices and ten b strands and a carboxy-
pearance of maternal GPIHBP1 autoantibodies (Beigneux et al., terminal flattened b-barrel domain containing twelve b strands
2017). In another patient with autoimmune thyroid disease and (Birrane et al., 2019). LPL contained two N-linked glycans and
multiple sclerosis (Kawahara et al., 2017), GPIHBP1 autoanti- one calcium ion in the amino-terminal catalytic domain.
bodies appeared after initiating b-interferon treatment (Eguchi Portions of the Trp-rich loop and the lid covering the catalytic
et al., 2019). (b-interferon can, in some patients, cause wors- pocket were not well defined in the structure by Birrane and co-
ening of autoimmune diseases; Banchereau and Pascual, workers (Birrane et al., 2019). However, very recently, Arora and
2006; Di Domizio and Cao, 2013; Silva, 2012.) In that patient, coworkers solved the crystal structure for a GPIHBP1-LPL com-
the GPIHBP1 autoantibodies disappeared and the plasma tri- plex in the presence of a weak competitive inhibitor of LPL (Arora
glycerides normalized after b-interferon therapy was discontin- et al., 2019). That LPL structure was similar to the one reported
ued (Eguchi et al., 2019). earlier (Birrane et al., 2019); however, the presence of the inhib-
In the patient treated with b-interferon, the GPIHBP1 autoanti- itor in LPL’s active site permitted a higher definition of the Trp-
bodies were largely subclass IgG4 (Eguchi et al., 2019). IgG4 rich loop and the lid domain.
autoantibodies have been reported to cause a variety of autoim- The GPIHBP1 was bound to the carboxy-terminal domain of
mune diseases, typically by disrupting protein-protein interac- LPL and, as expected, contained a classic LU fold with three
tions rather than by fixing complement and inducing tissue injury loops stabilized by five disulfide bonds (Leth et al., 2019). Elec-
(Huijbers et al., 2018; Koneczny, 2018). Whether most patients tron densities corresponding to GPIHBP1’s acidic domain could
with the GPIHBP1 autoantibody syndrome have IgG4 autoanti- not be identified in the GPIHBP1-LPL crystal structures, which
bodies remains to be determined. It will also be important to was not surprising, given that the acidic domain is disordered
determine if immune complexes are present in the plasma of and binds LPL in a transient and dynamic fashion (Kristensen
affected patients. The presence of immune complexes could et al., 2018).
have been overlooked by the ELISAs used to detect GPIHBP1 The interface between GPIHBP1 and LPL is large (940 Å2)
autoantibodies (Beigneux et al., 2017; Miyashita et al., 2018a). and mediated largely by hydrophobic contacts (Birrane et al.,
The frequency of the GPIHBP1 autoantibody syndrome 2019). Consistent with results from mutagenesis and HDX-MS
among patients with acquired forms of chylomicronemia is un- studies (Beigneux et al., 2011, 2015; Mysling et al., 2016a),
clear, but our experience, while limited, suggests that the syn- Trp-109 and adjacent residues in the second finger of GPIHBP1
drome may not be particularly rare (Beigneux et al., 2017). In were directly involved in the binding interface. Extensive se-
our experience, if any GPIHBP1 autoantibody is detected, the quences within the carboxy-terminal domain of LPL, including

58 Cell Metabolism 30, July 2, 2019


Cell Metabolism

Review

cysteine-knot protein stabilized by five disulfide bonds (van


Tilbeurgh et al., 1992). A comparison of crystal structures re-
vealed that GPIHBP1 and colipase bind to opposite sides of
the carboxy-terminal domains of LPL and PL, respectively (Bir-
rane et al., 2019). Also, colipase-PL interactions were discrete
and mediated by electrostatic interactions (van Tilbeurgh et al.,
1992), whereas GPIHBP1-LPL interactions were extensive and
driven primarily by hydrophobic contacts (Birrane et al., 2019).
In hindsight, the existence of distinct binding sites for GPIHBP1
and colipase was not surprising. While the two proteins have su-
perficial similarities, they are evolutionarily distinct and play very
different functions, with colipase serving to activate triglyceride
hydrolysis and GPIHBP1 serving to transport LPL and anchor it
to capillary endothelial cells.

LPL Contains a Single Large Basic Patch


LPL contains several positively charged heparin-binding motifs,
scattered within the primary sequence, and their functional
importance has been investigated by several groups (Hata
et al., 1993; Hill et al., 1998; Ma et al., 1994; Sendak et al.,
1998). Interestingly, an electrostatic surface potential map,
based on the GPIHBP1-LPL crystal structure, revealed that the
various heparin-binding motifs converge to create a single large
contiguous basic patch (covering 2,400 Å2) (Birrane et al.,
2019) (Figures 5A and 5B). This patch extends into both the
amino- and carboxy-terminal domains of LPL and spans the nar-
row hinge region connecting the two domains. Of note, two
lysines (K472 and K473) located within the last five residues of
LPL were not visualized in the electron density map (Birrane
et al., 2019). Had they been visualized, the size of LPL’s basic
patch would have appeared even larger.
As noted earlier, GPIHBP1’s intrinsically disordered acidic
Figure 5. Three-Dimensional Structure of the Human LPL-GPIHBP1 domain increases the initial encounter rate with LPL and stabi-
Complex lizes LPL’s structure. These effects are mediated by transient in-
The crystal structure of LPL as a surface representation colored by its elec-
trostatic potential (acidic [red], neutral [white], and basic [blue]) and human teractions (Kristensen et al., 2018; Mysling et al., 2016a, 2016b),
GPIHBP1 in a cartoon representation with b strands colored green (modified resulting in the formation of a ‘‘fuzzy complex’’ (Borgia et al.,
from the crystal structure by Birrane and coworkers [Birrane et al., 2019]). (A) 2018; Ivic et al., 2019). Given the transient nature of the interac-
Electrostatic potential map, revealing the binding of GPIHBP1’s LU domain to
tions, it was not surprising that the acidic domain could not be
the carboxy- terminal domain (CTD) of LPL.
(B) Electrostatic potential map (rotated 90 degrees compared to A), revealing a visualized in the crystal structure. However, it is noteworthy
large basic patch one surface of LPL (blue), spanning both the amino-terminal that GPIHBP1 was positioned such that its acidic domain,
domain (NTD) and the CTD. The amino- and carboxy-terminal regions of >60 Å in length (Kristensen et al., 2018), would be expected to
GPIHBP1 are marked by N and C, respectively.
(C) A model of GPIHBP1 in solution, as defined by SAXS analyses, illustrating project toward—and potentially interact with—the entirety of
the large space (112 Å) occupied by GPIHBP1’s intrinsically disordered acidic LPL’s basic patch (Figures 5C and 5D). It seems likely that these
domain (modified from Kristensen et al., 2018). Different ensembles from the interactions stabilize LPL structure and activity (Birrane et al.,
modeling of the disordered acidic domain are illustrated by colored spheres,
while the folded LU domain (located below) is represented by a cartoon rep-
2019). GPIHBP1’s acidic domain could provide a ‘‘backbone’’
resentation. The acidic domain of GPIHBP1 is absent in the models shown in spanning LPL’s amino- and carboxy-terminal domains—one
(A) and (B) because the acidic domain is disordered and was not visualized in that provides sufficient flexibility for enzyme function but also
the crystal structure.
the rigidity required to prevent the unfolding that might otherwise
(D) Schematic representation of the GPIHBP1-LPL complex, depicting a po-
tential interaction between GPIHBP1’s intrinsically disordered acidic domain be initiated by torsion of LPL’s hinge region (or collapse of LPL’s
(the semi-transparent ‘‘red cloud’’) and LPL’s large basic patch (blue). two domains upon each other).

Structural Insights into LPL Mutations Causing


sequences that had been implicated by mutagenesis and HDX- Chylomicronemia
MS studies (Hu et al., 2017b; Mysling et al., 2016a; Voss et al., Because the interface between GPIHBP1’s LU domain and
2011), participated in the GPIHBP1-LPL binding interface. the carboxy-terminal domain of LPL is large, Birrane and
With the GPIHBP1-LPL crystal structure in hand, one of our coworkers predicted that it would be possible to identify muta-
early questions was whether the binding site for GPIHBP1 on tions that would disrupt GPIHBP1-LPL interactions (Birrane
LPL might resemble the binding site for colipase on pancreatic et al., 2019). They were intrigued by a missense mutation in
lipase (PL). Like GPIHBP1, colipase is a small, multi-fingered LPL, M404R, identified in a Middle Eastern patient with

Cell Metabolism 30, July 2, 2019 59


Cell Metabolism

Review

chylomicronemia. That mutation was reported to abolish LPL proteins. The conformation of the GPIHBP1-LPL complex in the
secretion from cells (Pingitore et al., 2016), but the crystal struc- presence of lipoproteins remains to be determined.
ture revealed that M404 is located within the GPIHBP1-LPL In recent studies, Beigneux and coworkers revisited accepted
binding interface—implying that the mutation might actually dogma holding that LPL is only active as a homodimer (Beigneux
cause disease by interfering with LPL’s ability to bind to et al., 2019). When they subjected freshly secreted human LPL to
GPIHBP1. Indeed, GPIHBP1-LPL binding assays revealed that density gradient ultracentrifugation, they found that the LPL
the M404R mutation abolishes LPL binding to GPIHBP1 but mass and activity peaks corresponded to the size of monomers
does not affect LPL secretion from cells (Birrane et al., 2019). (55 kDa)—overlapping with or slightly smaller than the 66-kDa
The GPIHBP1-LPL crystal structure also yielded insights into a albumin standard and with minimal overlap with a 97-kDa phos-
D201V mutation in LPL, identified in two Lebanese families with phorylase b standard. Incubating the LPL with guanidine hydro-
chylomicronemia (Abifadel et al., 2004). The crystal structure re- chloride inactivated catalytic activity but did not alter the
vealed that D201 participates in the coordination of LPL’s sole apparent size of LPL, as judged by density gradient ultracentrifu-
calcium ion. A valine cannot fulfill that function. Birrane and co- gation. Also, the GPIHBP1-LPL complex, expected to have a
workers suspected that the inability of LPL-D201V to coordinate size of 75 kDa, was slightly larger than the albumin standard
calcium would interfere with LPL folding and abolish LPL by density gradient ultracentrifugation, with minimal overlap
secretion from cells. Indeed, when LPL-D201V was introduced with the phosphorylase b standard (Beigneux et al., 2019).
into CHO cells, the mutant LPL was produced in normal When LPL is loaded onto a heparin-Sepharose column and
amounts, but none was secreted from cells, providing a clear then eluted with a linear gradient of sodium chloride, there
explanation for the chylomicronemia phenotype. A mutation in are two distinct LPL peaks—a low-salt peak with little catalytic
D202, another residue involved in coordinating the calcium ion, activity and a high-salt peak containing large amounts of cata-
also abolished LPL secretion from cells (Birrane et al., 2019). lytic activity. For years, the low-salt peak was presumed to
Insights from the GPIHBP1-LPL crystal structure, along with contain inactive LPL monomers, whereas the high-salt peak
straightforward biochemical assays, should make it possible to was assumed to contain active homodimers. However, when
understand and classify most LPL mutations associated with Beigneux and coworkers subjected the active LPL in the high-
chylomicronemia. salt peak to density gradient ultracentrifugation, they found
that it exhibited the size of a monomer (i.e., slightly smaller
Relevance of the Head-to-Tail LPL Homodimer than the albumin standard) (Beigneux et al., 2019). Most of the
Conformation in the Crystal Structure LPL in the low-salt peak was inactive and in the form of aggre-
The fact that the crystal structure revealed two LPL molecules in gates at the bottom of the density gradient tube. Beigneux and
a head-to-tail orientation appeared, at least at first glance, to coworkers also examined the LPL in the low- and high-salt peaks
support long-held assumptions about the structure of LPL. For with a sandwich ELISA in which a single LPL-specific mono-
decades, the functional unit of active LPL has been assumed clonal antibody (either 5D2 or 88B8) was used to both capture
to be a homodimer (Berryman and Bensadoun, 1995; Garfinkel and detect the LPL. This single-antibody sandwich ELISA format
et al., 1983; Iverius and Ostlund-Lindqvist, 1976; Krapp et al., had been touted as being specific for catalytically active LPL
1995; Lookene and Bengtsson-Olivecrona, 1993; Lookene homodimers (Peterson et al., 2002). However, Beigneux and
et al., 1994; Olivecrona and Bengtsson-Olivecrona, 1990; Olive- coworkers found that the catalytically active LPL in the high-
crona et al., 1985; Osborne et al., 1985; Peterson et al., 2002; salt peak exhibited little reactivity in single-antibody sandwich
Vannier and Ailhaud, 1989; Wong et al., 1994; Zhang et al., ELISAs, whereas the aggregate-containing LPL species in the
2005), and homodimer assembly has been considered essential low-salt peak yielded a robust signal (Beigneux et al., 2019).
for LPL secretion from cells (Doolittle et al., 2010; Doolittle and Importantly, the LPL in both the low- and high-salt fractions
Péterfy, 2010). Several studies concluded that active LPL homo- yielded robust signals in sandwich ELISAs employing two
dimers have a head-to-tail orientation (Kobayashi et al., 2002; different monoclonal antibodies (one to capture the LPL and
Wong et al., 1997), with the Trp-rich loop in the carboxy-terminal the other to detect the LPL). The two-antibody sandwich ELISAs
domain of one monomer supplying substrates to the catalytic are sensitive in detecting LPL monomers as well as aggregated
pocket in the amino-terminal domain of the partner monomer. LPL species. The findings by Beigneux and coworkers implied
However, in the crystal structures for the GPIHBP1-LPL complex that the catalytically active LPL in the high-salt peak, long pre-
(Birrane et al., 2019; Horton, 2019; Arora et al., 2019), the Trp- sumed to be in the form of homodimers, is largely in the form
rich loop in the carboxy-terminal domain of one monomer inter- of monomers (Beigneux et al., 2019).
acted with the catalytic pocket of the partner monomer, raising Beigneux and coworkers also tested whether CHO cells, when
doubts about whether that particular conformation would be co-transfected with two LPL constructs harboring different
compatible with either lipoprotein binding or the hydrolysis of epitope tags, would secrete catalytically active ‘‘mixed homo-
triglyceride substrates. One possibility is that the reciprocal in- dimers’’ (i.e., LPL species containing both epitope tags)
teractions between the two LPL monomers do not represent (Beigneux et al., 2019). The medium of the co-transfected cells
physiologic interactions and instead reflect a stable conforma- contained large amounts of both differentially tagged LPL pro-
tion that LPL adopts at high protein concentrations—one in teins, and both LPL proteins were catalytically active. However,
which the catalytic pocket shields the hydrophobic Trp-rich the amount of mixed LPL species in the medium was very low, as
loop from the aqueous environment. It is important to empha- judged by specific ELISAs, but still detectable. Interestingly,
size, however, that all of the GPIHBP1-LPL crystal structures the mixed species in the medium were primarily located in
were generated in the absence of triglyceride substrates or lipo- the inactive low-salt peak, as judged by heparin-Sepharose

60 Cell Metabolism 30, July 2, 2019


Cell Metabolism

Review

chromatography, not in the catalytically active high-salt peak ACKNOWLEDGMENTS


(Beigneux et al., 2019). These studies provided further support
for the notion that freshly secreted, catalytically active LPL is pri- Supported by grants (HL090553, HL087228, and HL125335) from the National
Heart, Lung, and Blood Institute; a Transatlantic Network grant (12CVD04)
marily in the form of monomers. The concept that LPL can be from the Leducq Foundation; and Novo Nordisk Foundation grants
active as a monomer (Beigneux et al., 2019) was endorsed by (NNF17OC0026868 and NNF18OC0033864).
Arora and coworkers (Arora et al., 2019), based in part on their
suspicion that the head-to-tail LPL homodimers in their crystal DECLARATION OF INTERESTS
structure were unlikely to be physiologically active in binding li-
poproteins or hydrolyzing triglycerides. M.M. is an employee of Shire/Takeda and holds stock and stock options in the
The view that LPL can be active as a monomer is not inconsis- company. K.N. holds stock in Immunobiologic Laboratories and serves as a
consultant for Skylight and Sysmex. S.G.Y., A.P.B., L.G.F., and K.N. and their
tent with the existence of LPL homodimers in specific settings. respective institutions have applied for a patent on an assay for a diagnostic
LPL homodimers can exist, as documented by the crystallog- test for the GPIHBP1 autoantibody syndrome.
raphy and SAXS studies (Arora et al., 2019; Birrane et al.,
2019), but the relevance of those conformations to the physio- REFERENCES
logic function of LPL is definitely open to question. It is important
to emphasize that the LPL homodimers in the crystallization and Abifadel, M., Jambart, S., Allard, D., Rabès, J.P., Varret, M., Derré, A., Chou-
ery, E., Salem, N., Junien, C., Aydénian, H., et al. (2004). Identification of the
SAXS studies were observed when the protein concentrations first Lebanese mutation in the LPL gene and description of a rapid detection
were high, whereas the density gradient and immunochemical method. Clin. Genet. 65, 158–161.
studies supporting the existence of active LPL monomers were
Acton, S., Rigotti, A., Landschulz, K.T., Xu, S., Hobbs, H.H., and Krieger, M.
performed at far lower concentrations of LPL (Beigneux et al., (1996). Identification of scavenger receptor SR-BI as a high density lipoprotein
2019). It is possible that LPL homodimers that appear at high receptor. Science 271, 518–520.
protein concentrations dissociate into active monomers at lower Allan, C.M., Larsson, M., Hu, X., He, C., Jung, R.S., Mapar, A., Voss, C., Miya-
protein concentrations. However, at this point, the existence of shita, K., Machida, T., Murakami, M., et al. (2016). An LPL-specific monoclonal
stable and catalytically active LPL homodimers appears ques- antibody, 88B8, that abolishes the binding of LPL to GPIHBP1. J. Lipid Res. 57,
1889–1898.
tionable. Moreover, dogma that has accompanied the notion
that LPL is active only when it is in the form of homodimers Allan, C.M., Jung, C.J., Larsson, M., Heizer, P.J., Tu, Y., Sandoval, N.P., Dang,
T.L.P., Jung, R.S., Beigneux, A.P., de Jong, P.J., et al. (2017a). Mutating a
may need additional scrutiny. For example, the notion that
conserved cysteine in GPIHBP1 reduces amounts of GPIHBP1 in capillaries
ANGPTL4 functions by converting catalytically active LPL homo- and abolishes LPL binding. J. Lipid Res. 58, 1453–1461.
dimers into inactive LPL monomers (Sukonina et al., 2006) may
Allan, C.M., Larsson, M., Jung, R.S., Ploug, M., Bensadoun, A., Beigneux,
need to be revisited. Also, the concept that lipase maturation A.P., Fong, L.G., and Young, S.G. (2017b). Mobility of "HSPG-bound" LPL ex-
factor 1 (LMF-1) (an integral membrane protein of the ER) func- plains how LPL is able to reach GPIHBP1 on capillaries. J. Lipid Res. 58,
tions to convert newly synthesized LPL monomers into catalyti- 216–225.

cally active and secretion-competent homodimers (Doolittle Allan, C.M., Heizer, P.J., Tu, Y., Sandoval, N.P., Jung, R.S., Morales, J.E., Sajti,
et al., 2010; Doolittle and Péterfy, 2010) may also require addi- E., Troutman, T.D., Saunders, T.L., Cusanovich, D.A., et al. (2019). An up-
stream enhancer regulates Gpihbp1 expression in a tissue-specific manner.
tional scrutiny. J. Lipid Res. 60, 869–879.

Concluding Remarks Almeida, K.A., Strunz, C.M., Maranhão, R.C., and Mansur, A.P. (2007). The
S447X polymorphism of lipoprotein lipase: effect on the incidence of prema-
Studies over the past decade have established that GPIHBP1 is ture coronary disease and on plasma lipids. Arq. Bras. Cardiol. 88, 297–303.
LPL’s partner—required for transporting LPL into capillaries, li-
Ariza, M.J., Martı́nez-Hernández, P.L., Ibarretxe, D., Rabacchi, C., Rioja, J.,
poprotein margination, and for preserving LPL structure and ac- Grande-Aragón, C., Plana, N., Tarugi, P., Olivecrona, G., Calandra, S., et al.
tivity. Impaired GPIHBP1-LPL interactions, whether from genetic (2016). Novel mutations in the GPIHBP1 gene identified in 2 patients with
defects or autoantibodies, cause chylomicronemia. Biophysical recurrent acute pancreatitis. J. Clin. Lipidol. 10, 92–100.
studies have demonstrated that GPIHBP1’s acidic domain stabi- Arora, R., Nimonkar, A.V., Baird, D., Wang, C., Chiu, C.H., Horton, P.A., Han-
lizes LPL structure and activity, likely by creating a fuzzy complex rahan, S., Cubbon, R., Weldon, S., Tschantz, W.R., et al. (2019). Structure of
lipoprotein lipase in complex with GPIHBP1. Proc. Natl. Acad. Sci. U.S.A.
with LPL’s large basic patch. For years, the properties of LPL 116, 10360–10365.
have been analyzed primarily with purified preparations of LPL,
but future research will need to focus on the properties of the Banchereau, J., and Pascual, V. (2006). Type I interferon in systemic lupus
erythematosus and other autoimmune diseases. Immunity 25, 383–392.
GPIHBP1-LPL complex. We need a better understanding of lipo-
protein interactions with the GPIHBP1-LPL complex and how Beigneux, A.P. (2010). GPIHBP1 and the processing of triglyceride-rich lipo-
proteins. J. Clin. Lipidol. 5, 575–582.
regulators of lipolysis (e.g., apo-CII and apo-AV) influence the
activity of the GPIHBP1-LPL complex. We also need a clear un- Beigneux, A.P., Davies, B.S., Gin, P., Weinstein, M.M., Farber, E., Qiao, X.,
derstanding of how the ‘‘ANGPTL proteins’’ initiate unfolding of Peale, F., Bunting, S., Walzem, R.L., Wong, J.S., et al. (2007). Glycosylphos-
phatidylinositol-anchored high density lipoprotein–binding protein 1 plays a
LPL and a more precise molecular understanding of the interac- critical role in the lipolytic processing of chylomicrons. Cell Metab. 5, 279–291.
tions between GPIHBP1’s acidic domain and LPL’s basic patch.
Beigneux, A.P., Franssen, R., Bensadoun, A., Gin, P., Melford, K., Peter, J.,
Finally, commonly occurring LPL polymorphisms are known to
Walzem, R.L., Weinstein, M.M., Davies, B.S., Kuivenhoven, J.A., et al.
influence plasma triglyceride levels (Almeida et al., 2007; Jensen (2009a). Chylomicronemia with a mutant GPIHBP1 (Q115P) that cannot bind
et al., 2009; Rip et al., 2006); the impact of those polymorphisms lipoprotein lipase. Arterioscler. Thromb. Vasc. Biol. 29, 956–962.
needs to be investigated in the context of the GPIHBP1-LPL Beigneux, A.P., Gin, P., Davies, B.S.J., Weinstein, M.M., Bensadoun, A., Fong,
complex. L.G., and Young, S.G. (2009b). Highly conserved cysteines within the Ly6

Cell Metabolism 30, July 2, 2019 61


Cell Metabolism

Review
domain of GPIHBP1 are crucial for the binding of lipoprotein lipase. J. Biol. Davies, B.S., Goulbourne, C.N., Barnes, R.H., 2nd, Turlo, K.A., Gin, P.,
Chem. 284, 30240–30247. Vaughan, S., Vaux, D.J., Bensadoun, A., Beigneux, A.P., Fong, L.G., et al.
(2012). Assessing mechanisms of GPIHBP1 and lipoprotein lipase movement
Beigneux, A.P., Davies, B.S., Tat, S., Chen, J., Gin, P., Voss, C.V., Weinstein, across endothelial cells. J. Lipid. Res. 53, 2690–2697.
M.M., Bensadoun, A., Pullinger, C.R., Fong, L.G., et al. (2011). Assessing the
role of the glycosylphosphatidylinositol-anchored high density lipoprotein- de Beer, F., Hendriks, W.L., van Vark, L.C., Kamerling, S.W., van Dijk, K.W.,
binding protein 1 (GPIHBP1) three-finger domain in binding lipoprotein lipase. Hofker, M.H., Smelt, A.H., and Havekes, L.M. (1999). Binding of beta-VLDL
J. Biol. Chem. 286, 19735–19743. to heparan sulfate proteoglycans requires lipoprotein lipase, whereas ApoE
only modulates binding affinity. Arterioscler. Thromb. Vasc. Biol. 19, 633–637.
Beigneux, A.P., Fong, L.G., Bensadoun, A., Davies, B.S., Oberer, M., Gårds-
voll, H., Ploug, M., and Young, S.G. (2015). GPIHBP1 missense mutations Dewey, F.E., Gusarova, V., O’Dushlaine, C., Gottesman, O., Trejos, J., Hunt,
often cause multimerization of GPIHBP1 and thereby prevent lipoprotein C., Van Hout, C.V., Habegger, L., Buckler, D., Lai, K.M., et al. (2016). Inactivat-
lipase binding. Circ. Res. 116, 624–632. ing variants in ANGPTL4 and risk of coronary artery disease. N. Engl. J. Med.
374, 1123–1133.
Beigneux, A.P., Miyashita, K., Ploug, M., Blom, D.J., Ai, M., Linton, M.F., Kho-
vidhunkit, W., Dufour, R., Garg, A., McMahon, M.A., et al. (2017). Autoanti- Di Domizio, J., and Cao, W. (2013). Fueling autoimmunity: type I interferon in
bodies against GPIHBP1 as a cause of hypertriglyceridemia. N. Engl. J. autoimmune diseases. Expert Rev. Clin. Immunol. 9, 201–210.
Med. 376, 1647–1658.
Dijk, W., Beigneux, A.P., Larsson, M., Bensadoun, A., Young, S.G., and Ker-
Beigneux, A.P., Allan, C.M., Sandoval, N.P., Cho, G.W., Heizer, P.J., Jung, sten, S. (2016). Angiopoietin-like 4 promotes intracellular degradation of lipo-
R.S., Stanhope, K.L., Havel, P.J., Birrane, G., Meiyappan, M., et al. (2019). Li- protein lipase in adipocytes. J. Lipid. Res. 57, 1670–1683.
poprotein lipase is active as a monomer. Proc. Natl. Acad. Sci. U.S.A. 116,
6319–6328. Doolittle, M.H., and Péterfy, M. (2010). Mechanisms of lipase maturation.
J. Clin. Lipidol. 5, 71–85.
Bengtsson, G., and Olivecrona, T. (1979). Apolipoprotein CII enhances hydro-
lysis of monoglycerides by lipoprotein lipase, but the effect is abolished by Doolittle, M.H., Ehrhardt, N., and Péterfy, M. (2010). Lipase maturation factor
fatty acids. FEBS Lett. 106, 345–348. 1: structure and role in lipase folding and assembly. Curr. Opin. Lipidol. 21,
198–203.
Berryman, D.E., and Bensadoun, A. (1995). Heparan sulfate proteoglycans are
primarily responsible for the maintenance of enzyme activity, binding, and Duchesne, L., Octeau, V., Bearon, R.N., Beckett, A., Prior, I.A., Lounis, B., and
degradation of lipoprotein lipase in Chinese hamster ovary cells. J. Biol. Fernig, D.G. (2012). Transport of fibroblast growth factor 2 in the pericellular
Chem. 270, 24525–24531. matrix is controlled by the spatial distribution of its binding sites in heparan sul-
fate. PLoS Biol. 10, e1001361.
Birrane, G., Beigneux, A.P., Dwyer, B., Strack-Logue, B., Kristensen, K.K.,
Francone, O.L., Fong, L.G., Mertens, H.D.T., Pan, C.Q., Ploug, M., et al. Eguchi, J., Miyashita, K., Fukamachi, I., Nakajima, K., Murakami, M., Kawa-
(2019). Structure of the lipoprotein lipase-GPIHBP1 complex that mediates hara, Y., Yamashita, T., Ohta, Y., Abe, K., Nakatsuka, A., et al. (2019). GPIHBP1
plasma triglyceride hydrolysis. Proc. Natl. Acad. Sci. U.S.A. 116, 1723–1732. autoantibody syndrome during interferon beta1a treatment. J. Clin. Lipidol.
13, 62–69.
Bishop, J.R., Passos-Bueno, M.R., Fong, L., Stanford, K.I., Gonzales, J.C.,
Yeh, E., Young, S.G., Bensadoun, A., Witztum, J.L., Esko, J.D., et al. (2010).
Emmerich, J., Beg, O.U., Peterson, J., Previato, L., Brunzell, J.D., Brewer,
Deletion of the basement membrane heparan sulfate proteoglycan type XVIII
H.B., Jr., and Santamarina-Fojo, S. (1992). Human lipoprotein lipase. Analysis
collagen causes hypertriglyceridemia in mice and humans. PLoS One 5,
of the catalytic triad by site-directed mutagenesis of Ser-132, Asp-156, and
e13919.
His-241. J. Biol. Chem. 267, 4161–4165.
Blanchette-Mackie, E.J., Masuno, H., Dwyer, N.K., Olivecrona, T., and Scow,
Enerba€ck, S., Semb, H., Bengtsson-Olivecrona, G., Carlsson, P., Hermans-
R.O. (1989). Lipoprotein lipase in myocytes and capillary endothelium of heart:
immunocytochemical study. Am. J. Physiol. 256, E818–E828. son, M.L., Olivecrona, T., and Bjursell, G. (1987). Molecular cloning and
sequence analysis of cDNA encoding lipoprotein lipase of guinea pig. Gene
Borgia, A., Borgia, M.B., Bugge, K., Kissling, V.M., Heidarsson, P.O., Fer- 58, 1–12.
nandes, C.B., Sottini, A., Soranno, A., Buholzer, K.J., Nettels, D., et al.
(2018). Extreme disorder in an ultrahigh-affinity protein complex. Nature Ferguson, M.A.J., Kinoshita, T., and Hart, G.W. (2009). Glycosylphosphatidy-
555, 61–66. linositol anchors. In Essentials of Glycobiology, A. Varki, R.D. Cummings, J.D.
Esko, H.H. Freeze, P. Stanley, C.R. Bertozzi, G.W. Hart, and M.E. Etzler, eds.
Breckenridge, W.C., Little, J.A., Steiner, G., Chow, A., and Poapst, M. (1978). (Cold Spring Harbor).
Hypertriglyceridemia associated with deficiency of apolipoprotein C-II.
N. Engl. J. Med. 298, 1265–1273. Fong, L.G., Young, S.G., Beigneux, A.P., Bensadoun, A., Oberer, M., Jiang, H.,
and Ploug, M. (2016). GPIHBP1 and plasma triglyceride metabolism. Trends
Charrière, S., Peretti, N., Bernard, S., Di Filippo, M., Sassolas, A., Merlin, M., Endocrinol. Metab. 27, 455–469.
Delay, M., Debard, C., Lefai, E., Lachaux, A., et al. (2011). GPIHBP1 C89F
neomutation and hydrophobic C-terminal domain G175R mutation in two ped- Franssen, R., Young, S.G., Peelman, F., Hertecant, J., Sierts, J.A., Schimmel,
igrees with severe hyperchylomicronemia. J. Clin. Endocrinol. Metab. 96, A.W.M., Bensadoun, A., Kastelein, J.J.P., Fong, L.G., Dallinga-Thie, G.M.,
E1675–E1679. et al. (2010). Chylomicronemia with low postheparin lipoprotein lipase levels
in the setting of GPIHBP1 defects. Circ. Cardiovasc. Genet. 3, 169–178.
Chi, X., Shetty, S.K., Shows, H.W., Hjelmaas, A.J., Malcolm, E.K., and Davies,
B.S. (2015). Angiopoietin-like 4 modifies the interactions between lipoprotein Garfinkel, A.S., Kempner, E.S., Ben-Zeev, O., Nikazy, J., James, S.J., and
lipase and its endothelial cell transporter GPIHBP1. J. Biol. Chem. 290, Schotz, M.C. (1983). Lipoprotein lipase: size of the functional unit determined
11865–11877. by radiation inactivation. J. Lipid Res. 24, 775–780.

Chyzhyk, V., Kozmic, S., Brown, A.S., Hudgins, L.C., Starc, T.J., Davila, A.D., Gin, P., Yin, L., Davies, B.S., Weinstein, M.M., Ryan, R.O., Bensadoun, A.,
Blevins, T.C., Diffenderfer, M.R., He, L., Geller, A.S., et al. (2019). Extreme hy- Fong, L.G., Young, S.G., and Beigneux, A.P. (2008). The acidic domain of
pertriglyceridemia: genetic diversity, pancreatitis, pregnancy, and prevalence. GPIHBP1 is important for the binding of lipoprotein lipase and chylomicrons.
J. Clin. Lipidol. 13, 89–99. J. Biol. Chem. 283, 29554–29562.

Coca-Prieto, I., Kroupa, O., Gonzalez-Santos, P., Magne, J., Olivecrona, G., Gin, P., Beigneux, A.P., Voss, C., Davies, B.S., Beckstead, J.A., Ryan, R.O.,
Ehrenborg, E., and Valdivielso, P. (2011). Childhood-onset chylomicronaemia Bensadoun, A., Fong, L.G., and Young, S.G. (2011). Binding preferences for
with reduced plasma lipoprotein lipase activity and mass: identification of a GPIHBP1, a glycosylphosphatidylinositol-anchored protein of capillary endo-
novel GPIHBP1 mutation. J. Intern. Med. 270, 224–228. thelial cells. Arterioscler. Thromb. Vasc. Biol. 31, 176–182.

Davies, B.S., Beigneux, A.P., Barnes, R.H., Tu, Y., Gin, P., Weinstein, M.M., Gin, P., Goulbourne, C.N., Adeyo, O., Beigneux, A.P., Davies, B.S., Tat, S.,
Nobumori, C., Nyrén, R., Goldberg, I., Olivecrona, G., et al. (2010). GPIHBP1 Voss, C.V., Bensadoun, A., Fong, L.G., and Young, S.G. (2012). Chylomicrone-
is responsible for the entry of lipoprotein lipase into capillaries. Cell Metab. mia mutations yield new insights into interactions between lipoprotein lipase
12, 42–52. and GPIHBP1. Hum. Mol. Genet. 21, 2961–2972.

62 Cell Metabolism 30, July 2, 2019


Cell Metabolism

Review
Gonzaga-Jauregui, C., Mir, S., Penney, S., Jhangiani, S., Midgen, C., Finegold, Ioka, R.X., Kang, M.J., Kamiyama, S., Kim, D.H., Magoori, K., Kamataki, A., Ito,
M., Muzny, D.M., Wang, M., Bacino, C.A., Gibbs, R.A., et al. (2014). Whole- Y., Takei, Y.A., Sasaki, M., Suzuki, T., et al. (2003). Expression cloning and
exome sequencing reveals GPIHBP1 mutations in infantile colitis with severe characterization of a novel glycosylphosphatidylinositol-anchored high den-
hypertriglyceridemia. J. Pediatr. Gastroenterol. Nutr. 59, 17–21. sity lipoprotein-binding protein, GPI-HBP1. J. Biol. Chem. 278, 7344–7349.

Goulbourne, C.N., Gin, P., Tatar, A., Nobumori, C., Hoenger, A., Jiang, H., Iverius, P.H., and Ostlund-Lindqvist, A.M. (1976). Lipoprotein lipase from
Grovenor, C.R., Adeyo, O., Esko, J.D., Goldberg, I.J., et al. (2014). The bovine milk. Isolation procedure, chemical characterization, and molecular
GPIHBP1-LPL complex is responsible for the margination of triglyceride-rich weight analysis. J. Biol. Chem. 251, 7791–7795.
lipoproteins in capillaries. Cell Metab. 19, 849–860.
Ivic, N., Potocnjak, M., Solis-Mezarino, V., Herzog, F., Bilokapic, S., and Halic,
Gutgsell, A.R., Ghodge, S.V., Bowers, A.A., and Neher, S.B. (2019). Mapping M. (2019). Fuzzy interactions form and shape the histone transport complex.
the sites of the lipoprotein lipase (LPL)-angiopoietin-like protein 4 (ANGPTL4) Mol. Cell 73, 1191–1203.
interaction provides mechanistic insight into LPL inhibition. J. Biol. Chem. 294,
2678–2689. Jensen, M.K., Rimm, E.B., Rader, D., Schmidt, E.B., Sørensen, T.I., Vogel, U.,
Overvad, K., and Mukamal, K.J. (2009). S447X variant of the lipoprotein lipase
Haller, J.F., Mintah, I.J., Shihanian, L.M., Stevis, P., Buckler, D., Alexa-Braun, gene, lipids, and risk of coronary heart disease in 3 prospective cohort studies.
C.A., Kleiner, S., Banfi, S., Cohen, J.C., Hobbs, H.H., et al. (2017). ANGPTL8 Am. Heart J. 157, 384–390.
requires ANGPTL3 to inhibit lipoprotein lipase and plasma triglyceride clear-
ance. J. Lipid. Res. 58, 1166–1173. Kawahara, Y., Yamashita, T., Ohta, Y., Sato, K., Tsunoda, K., Takemoto, M.,
Hishikawa, N., Eguchi, J., and Abe, K. (2017). Marked hypertriglyceridemia
Hata, A., Ridinger, D.N., Sutherland, S., Emi, M., Shuhua, Z., Myers, R.L., Ren, induced by interferon-beta1a therapy in a clinically isolated syndrome patient.
K., Cheng, T., Inoue, I., and Wilson, D.E. (1993). Binding of lipoprotein lipase to J. Neurol. Sci. 373, 144–146.
heparin. Identification of five critical residues in two distinct segments of the
amino-terminal domain. J. Biol. Chem. 268, 8447–8457. Kersten, S. (2017). Triglyceride metabolism under attack. Cell Metab. 25,
1209–1210.
Havel, R.J. (2010). Triglyceride-rich lipoproteins and plasma lipid transport.
Arterioscler. Thromb. Vasc. Biol. 30, 9–19. Kirchgessner, T.G., Svenson, K.L., Lusis, A.J., and Schotz, M.C. (1987). The
sequence of cDNA encoding lipoprotein lipase. A member of a lipase gene
Havel, R.J., and Gordon, R.S., Jr. (1960). Idiopathic hyperlipemia: metabolic family. J. Biol. Chem. 262, 8463–8466.
studies in an affected family. J. Clin. Invest. 39, 1777–1790.
Kobayashi, Y., Nakajima, T., and Inoue, I. (2002). Molecular modeling of the
Havel, R.J., and Kane, J.P. (2001). Introduction: structure and metabolism of dimeric structure of human lipoprotein lipase and functional studies of the
plasma lipoproteins. In The metabolic and molecular bases of inherited dis- carboxyl-terminal domain. Eur. J. Biochem. 269, 4701–4710.
ease, C.R. Scriver, A.L. Beaudet, W.S. Sly, D. Valle, B. Childs, K.W. Kinzler,
and B. Vogelstein, eds. (McGraw-Hill), pp. 2705–2716. Koneczny, I. (2018). A new classification system for IgG4 autoantibodies.
Front. Immunol. 9, 97.
Havel, R.J., Shore, V.G., Shore, B., and Bier, D.M. (1970). Role of specific gly-
copeptides of human serum lipoproteins in the activation of lipoprotein lipase. Korn, E.D. (1955). Clearing factor, a heparin-activated lipoprotein lipase. I.
Circ. Res. 27, 595–600. Isolation and characterization of the enzyme from normal rat heart. J. Biol.
Chem. 215, 1–14.
He, C., Weston, T.A., Jung, R.S., Heizer, P., Larsson, M., Hu, X., Allan, C.M.,
Tontonoz, P., Reue, K., Beigneux, A.P., et al. (2018a). NanoSIMS analysis of Korn, E.D., and Quigley, T.W., Jr. (1955). Studies on lipoprotein lipase of rat
intravascular lipolysis and lipid movement across capillaries and into cardio- heart and adipose tissue. Biochim. Biophys. Acta 18, 143–145.
myocytes. Cell Metab. 27, 1055–1066.e3.
Kovrov, O., Kristensen, K.K., Larsson, E., Ploug, M., and Olivecrona, G. (2019).
He, L., Vanlandewijck, M., Ma €e, M.A., Andrae, J., Ando, K., Del Gaudio, F., Na- On the mechanism of angiopoietin-like protein 8 for control of lipoprotein
har, K., Lebouvier, T., Laviña, B., Gouveia, L., et al. (2018b). Single-cell RNA lipase activity. J. Lipid Res. 60, 783–793.
sequencing of mouse brain and lung vascular and vessel-associated cell
types. Sci. Data 5. Krapp, A., Zhang, H., Ginzinger, D., Liu, M.S., Lindberg, A., Olivecrona, G.,
Hayden, M.R., and Beisiegel, U. (1995). Structural features in lipoprotein lipase
Helgadottir, A., Gretarsdottir, S., Thorleifsson, G., Hjartarson, E., Sigurdsson, necessary for the mediation of lipoprotein uptake into cells. J. Lipid Res. 36,
A., Magnusdottir, A., Jonasdottir, A., Kristjansson, H., Sulem, P., Oddsson, A., 2362–2373.
et al. (2016). Variants with large effects on blood lipids and the role of choles-
terol and triglycerides in coronary disease. Nat. Genet. 48, 634–639. Kristensen, K.K., Midtgaard, S.R., Mysling, S., Kovrov, O., Hansen, L.B., Skar-
Gislinge, N., Beigneux, A.P., Kragelund, B.B., Olivecrona, G., Young, S.G.,
Henderson, H.E., Hassan, F., Marais, D., and Hayden, M.R. (1996). A new mu- et al. (2018). A disordered acidic domain in GPIHBP1 harboring a sulfated
tation destroying disulphide bridging in the C-terminal domain of lipoprotein tyrosine regulates lipoprotein lipase. Proc. Natl. Acad. Sci. U.S.A. 115,
lipase. Biochem. Biophys. Res. Commun. 227, 189–194. E6020–E6029.

Henderson, H., Leisegang, F., Hassan, F., Hayden, M., and Marais, D. (1998). A Lafferty, M.J., Bradford, K.C., Erie, D.A., and Neher, S.B. (2013). Angiopoietin-
novel Glu421Lys substitution in the lipoprotein lipase gene in pregnancy- like protein 4 inhibition of lipoprotein lipase: evidence for reversible complex
induced hypertriglyceridemic pancreatitis. Clin. Chim. Acta 269, 1–12. formation. J. Biol. Chem. 288, 28524–28534.

Hill, J.S., Yang, D., Nikazy, J., Curtiss, L.K., Sparrow, J.T., and Wong, H. LaRosa, J.C., Levy, R.I., Herbert, P., Lux, S.E., and Fredrickson, D.S. (1970). A
(1998). Subdomain chimeras of hepatic lipase and lipoprotein lipase. Localiza- specific apoprotein activator for lipoprotein lipase. Biochem. Biophys. Res.
tion of heparin and cofactor binding. J. Biol. Chem. 273, 30979–30984. Commun. 41, 57–62.

Horton, J.D. (2019). Intravascular triglyceride lipolysis becomes crystal clear. Leth, J., Leth-Espensen, K., Kristensen, K., Kumari, A., Winther, A., Young, S.,
Proc. Natl. Acad. Sci. U.S.A. 116, 1480–1482. and Ploug, M. (2019). Evolution and medical significance of LU domain
containing proteins. Int. J. Mol. Sci. 20, E2760, https://doi.org/10.3390/
Hu, X., Dallinga-Thie, G.M., Hovingh, G.K., Chang, S.Y., Sandoval, N.P., Dang, ijms20112760.
T.L.P., Fukamachi, I., Miyashita, K., Nakajima, K., Murakami, M., et al. (2017a).
GPIHBP1 autoantibodies in a patient with unexplained chylomicronemia. Lookene, A., and Bengtsson-Olivecrona, G. (1993). Chymotryptic cleavage of
J. Clin. Lipidol. 11, 964–971. lipoprotein lipase. Identification of cleavage sites and functional studies of the
truncated molecule. Eur. J. Biochem. 213, 185–194.
Hu, X., Sleeman, M.W., Miyashita, K., Linton, M.F., Allan, C.M., He, C., Lars-
son, M., Tu, Y., Sandoval, N.P., Jung, R.S., et al. (2017b). Monoclonal anti- Lookene, A., Skottova, N., and Olivecrona, G. (1994). Interactions of lipopro-
bodies that bind to the Ly6 domain of GPIHBP1 abolish the binding of LPL. tein lipase with the active-site inhibitor tetrahydrolipstatin (orlistat). Eur. J. Bio-
J. Lipid Res. 58, 208–215. chem. 222, 395–403.

Huijbers, M.G., Plomp, J.J., van der Maarel, S.M., and Verschuuren, J.J. Lookene, A., Chevreuil, O., Ostergaard, P., and Olivecrona, G. (1996). Interac-
(2018). IgG4-mediated autoimmune diseases: a niche of antibody-mediated tion of lipoprotein lipase with heparin fragments and with heparan sulfate: stoi-
disorders. Ann. N. Y. Acad. Sci. 1413, 92–103. chiometry, stabilization, and kinetics. Biochemistry 35, 12155–12163.

Cell Metabolism 30, July 2, 2019 63


Cell Metabolism

Review
Lookene, A., Groot, N.B., Kastelein, J.J., Olivecrona, G., and Bruin, T. (1997a). Osborne, J.C., Jr., Bengtsson-Olivecrona, G., Lee, N.S., and Olivecrona, T.
Mutation of tryptophan residues in lipoprotein lipase. Effects on stability, (1985). Studies on inactivation of lipoprotein lipase: role of the dimer to mono-
immunoreactivity, and catalytic properties. J. Biol. Chem. 272, 766–772. mer dissociation. Biochemistry 24, 5606–5611.

Lookene, A., Savonen, R., and Olivecrona, G. (1997b). Interaction of lipopro- Peterson, J., Ayyobi, A.F., Ma, Y., Henderson, H., Reina, M., Deeb, S.S., San-
teins with heparan sulfate proteoglycans and with lipoprotein lipase. Studies tamarina-Fojo, S., Hayden, M.R., and Brunzell, J.D. (2002). Structural and
by surface plasmon resonance technique. Biochemistry 36, 5267–5275. functional consequences of missense mutations in exon 5 of the lipoprotein
lipase gene. J. Lipid Res. 43, 398–406.
Ma, Y., Henderson, H.E., Liu, M.S., Zhang, H., Forsythe, I.J., Clarke-Lewis, I.,
Hayden, M.R., and Brunzell, J.D. (1994). Mutagenesis in four candidate hepa- Pingitore, P., Lepore, S.M., Pirazzi, C., Mancina, R.M., Motta, B.M., Valenti, L.,
rin binding regions (residues 279–282, 291–304, 390–393, and 439–448) and Berge, K.E., Retterstøl, K., Leren, T.P., Wiklund, O., et al. (2016). Identification
identification of residues affecting heparin binding of human lipoprotein lipase. and characterization of two novel mutations in the LPL gene causing type I hy-
J. Lipid Res. 35, 2049–2059. perlipoproteinemia. J. Clin. Lipidol. 10, 816–823.

Mailly, F., Palmen, J., Muller, D.P., Gibbs, T., Lloyd, J., Brunzell, J., Durrington, Plengpanich, W., Young, S.G., Khovidhunkit, W., Bensadoun, A., Karnman, H.,
P., Mitropoulos, K., Betteridge, J., Watts, G., et al. (1997). Familial lipoprotein Ploug, M., Gårdsvoll, H., Leung, C.S., Adeyo, O., Larsson, M., et al. (2014).
lipase (LPL) deficiency: a catalogue of LPL gene mutations identified in 20 pa- Multimerization of glycosylphosphatidylinositol-anchored high density lipo-
tients from the UK, Sweden, and Italy. Hum. Mutat. 10, 465–473. protein-binding protein 1 (GPIHBP1) and familial chylomicronemia from a
serine-to-cysteine Substitution in GPIHBP1 Ly6 domain. J. Biol. Chem. 289,
Mergenthaler, P., Lindauer, U., Dienel, G.A., and Meisel, A. (2013). Sugar for 19491–19499.
the brain: the role of glucose in physiological and pathological brain function.
Trends Neurosci. 36, 587–597. Quagliarini, F., Wang, Y., Kozlitina, J., Grishin, N.V., Hyde, R., Boerwinkle, E.,
Valenzuela, D.M., Murphy, A.J., Cohen, J.C., and Hobbs, H.H. (2012). Atypical
Miyashita, K., Fukamachi, I., Machida, T., Nakajima, K., Young, S.G., Mura- angiopoietin-like protein that regulates ANGPTL3. Proc. Natl. Acad. Sci.
kami, M., Beigneux, A.P., and Nakajima, K. (2018a). An ELISA for quantifying U.S.A. 109, 19751–19756.
GPIHBP1 autoantibodies and making a diagnosis of the GPIHBP1 autoanti-
body syndrome. Clin. Chim. Acta 487, 174–178. Rabacchi, C., D’Addato, S., Palmisano, S., Lucchi, T., Bertolini, S., Calandra, S.,
and Tarugi, P. (2016). Clinical and genetic features of 3 patients with familial chy-
Miyashita, K., Fukamachi, I., Nagao, M., Ishida, T., Kobayashi, J., Machida, T., lomicronemia due to mutations in GPIHBP1 gene. J. Clin. Lipidol. 10, 915–921.
Nakajima, K., Murakami, M., Ploug, M., Beigneux, A.P., et al. (2018b). An
enzyme-linked immunosorbent assay for measuring GPIHBP1 levels in human Rader, D.J. (2006). Molecular regulation of HDL metabolism and function: im-
plasma or serum. J. Clin. Lipidol. 12, 203–210. plications for novel therapies. J. Clin. Invest. 116, 3090–3100.

Myocardial Infarction Genetics; CARDIoGRAM Exome Consortia Investiga- Rios, J.J., Shastry, S., Jasso, J., Hauser, N., Garg, A., Bensadoun, A., Cohen,
tors, Stitziel, N.O., Stirrups, K.E., Masca, N.G., Erdmann, J., Ferrario, P.G., J.C., and Hobbs, H.H. (2012). Deletion of GPIHBP1 causing severe chylomi-
König, I.R., Weeke, P.E., Webb, T.R., Auer, P.L., et al. (2016). Coding variation cronemia. J. Inherit. Metab. Dis. 35, 531–540.
in ANGPTL4, LPL, and SVEP1 and the risk of coronary disease. N. Engl. J.
Med. 374, 1134–1144. Rip, J., Nierman, M.C., Ross, C.J., Jukema, J.W., Hayden, M.R., Kastelein,
J.J., Stroes, E.S., and Kuivenhoven, J.A. (2006). Lipoprotein lipase S447X:
Mysling, S., Kristensen, K.K., Larsson, M., Beigneux, A.P., Gårdsvoll, H., Fong, a naturally occurring gain-of-function mutation. Arterioscler. Thromb. Vasc.
L.G., Bensadouen, A., Jørgensen, T.J., Young, S.G., and Ploug, M. (2016a). Biol. 26, 1236–1245.
The acidic domain of the endothelial membrane protein GPIHBP1 stabilizes li-
Rye, K.A., and Barter, P.J. (2014). Regulation of high-density lipoprotein meta-
poprotein lipase activity by preventing unfolding of its catalytic domain. eLife 5,
bolism. Circ. Res. 114, 143–156.
e12095.
Sendak, R.A., Melford, K., Kao, A., and Bensadoun, A. (1998). Identification of
Mysling, S., Kristensen, K.K., Larsson, M., Kovrov, O., Bensadouen, A., Jør-
the epitope of a monoclonal antibody that inhibits heparin binding of lipopro-
gensen, T.J., Olivecrona, G., Young, S.G., and Ploug, M. (2016b). The angio-
tein lipase: new evidence for a carboxyl-terminal heparin-binding domain.
poietin-like protein ANGPTL4 catalyzes unfolding of the hydrolase domain in
J. Lipid Res. 39, 633–646.
lipoprotein lipase and the endothelial membrane protein GPIHBP1 counter-
acts this unfolding. eLife 5, e20958. Shoemaker, B.A., Portman, J.J., and Wolynes, P.G. (2000). Speeding molec-
ular recognition by using the folding funnel: the fly-casting mechanism. Proc.
Nevo, Y., Ben-Zeev, B., Tabib, A., Straussberg, R., Anikster, Y., Shorer, Z., Natl. Acad. Sci. U.S.A. 97, 8868–8873.
Fattal-Valevski, A., Ta-Shma, A., Aharoni, S., Rabie, M., et al. (2013). CD59
deficiency is associated with chronic hemolysis and childhood relapsing im- Silva, M.O. (2012). Risk of autoimmune complications associated with inter-
mune-mediated polyneuropathy. Blood 121, 129–135. feron therapy. Gastroenterol. Hepatol. (NY) 8, 540–542.
Nilsson, S.K., Anderson, F., Ericsson, M., Larsson, M., Makoveichuk, E., Loo- Simionescu, M., and Simionescu, N. (1986). Functions of the endothelial cell
kene, A., Heeren, J., and Olivecrona, G. (2012). Triacylglycerol-rich lipopro- surface. Annu. Rev. Physiol. 48, 279–293.
teins protect lipoprotein lipase from inactivation by ANGPTL3 and ANGPTL4.
Biochim. Biophys. Acta 1821, 1370–1378. Sukonina, V., Lookene, A., Olivecrona, T., and Olivecrona, G. (2006). Angio-
poietin-like protein 4 converts lipoprotein lipase to inactive monomers and
Olafsen, T., Young, S.G., Davies, B.S., Beigneux, A.P., Kenanova, V.E., Voss, modulates lipase activity in adipose tissue. Proc. Natl. Acad. Sci. U.S.A.
C., Young, G., Wong, K.P., Barnes, R.H., 2nd, Tu, Y., et al. (2010). Unexpected 103, 17450–17455.
expression pattern for glycosylphosphatidylinositol-anchored HDL-binding
protein 1 (GPIHBP1) in mouse tissues revealed by positron emission tomogra- van Tilbeurgh, H., Sarda, L., Verger, R., and Cambillau, C. (1992). Structure of
phy scanning. J. Biol. Chem. 285, 39239–39248. the pancreatic lipase–procolipase complex. Nature 359, 159–162.

Olivecrona, T., and Bengtsson-Olivecrona, G. (1990). Lipoprotein lipase and Vanlandewijck, M., He, L., Ma €e, M.A., Andrae, J., Ando, K., Del Gaudio, F., Na-
hepatic lipase. Curr. Opin. Lipidol. 1, 222–230. har, K., Lebouvier, T., Laviña, B., Gouveia, L., et al. (2018). A molecular atlas of
cell types and zonation in the brain vasculature. Nature 554, 475–480.
Olivecrona, T., and Bengtsson-Olivecrona, G. (1993). Lipoprotein lipase and
hepatic lipase. Curr. Opin. Lipidol. 4, 187–196. Vannier, C., and Ailhaud, G. (1989). Biosynthesis of lipoprotein lipase in
cultured mouse adipocytes. II. Processing, subunit assembly, and intracellular
Olivecrona, T., Bengtsson-Olivecrona, G., Osborne, J.C., Jr., and Kempner, transport. J. Biol. Chem. 264, 13206–13216.
E.S. (1985). Molecular size of bovine lipoprotein lipase as determined by radi-
ation inactivation. J. Biol. Chem. 260, 6888–6891. Vijayagopal, P., Radhakrishnamurthy, B., Srinivasan, S.R., and Berenson, G.S.
(1980). Studies of biologic properties of proteoglycans from bovine aorta. Lab.
Olivecrona, G., Ehrenborg, E., Semb, H., Makoveichuk, E., Lindberg, A., Hay- Invest. 42, 190–196.
den, M.R., Gin, P., Davies, B.S., Weinstein, M.M., Fong, L.G., et al. (2010).
Mutation of conserved cysteines in the Ly6 domain of GPIHBP1 in familial chy- Voss, C.V., Davies, B.S., Tat, S., Gin, P., Fong, L.G., Pelletier, C., Mottler, C.D.,
lomicronemia. J. Lipid Res. 51, 1535–1545. Bensadoun, A., Beigneux, A.P., and Young, S.G. (2011). Mutations in

64 Cell Metabolism 30, July 2, 2019


Cell Metabolism

Review
lipoprotein lipase that block binding to the endothelial cell transporter Wion, K.L., Kirchgessner, T.G., Lusis, A.J., Schotz, M.C., and Lawn, R.M.
GPIHBP1. Proc. Natl. Acad. Sci. U.S.A. 108, 7980–7984. (1987). Human lipoprotein lipase complementary DNA sequence. Science
235, 1638–1641.
Wang, H., and Eckel, R.H. (2009). Lipoprotein lipase: from gene to obesity. Am.
J. Physiol. Endocrinol. Metab. 297, E271–E288. Wong, H., Davis, R.C., Thuren, T., Goers, J.W., Nikazy, J., Waite, M., and
Schotz, M.C. (1994). Lipoprotein lipase domain function. J. Biol. Chem. 269,
10319–10323.
Weinstein, M.M., Tu, Y., Beigneux, A.P., Davies, B.S., Gin, P., Voss, C., Wal-
zem, R.L., Reue, K., Tontonoz, P., Bensadoun, A., et al. (2010a). Cholesterol
Wong, H., Yang, D., Hill, J.S., Davis, R.C., Nikazy, J., and Schotz, M.C. (1997).
intake modulates plasma triglyceride levels in glycosylphosphatidylinositol
A molecular biology-based approach to resolve the subunit orientation of lipo-
HDL-binding protein 1-deficient mice. Arterioscler. Thromb. Vasc. Biol. 30,
protein lipase. Proc. Natl. Acad. Sci. U.S.A. 94, 5594–5598.
2106–2113.
Yamamoto, H., Onishi, M., Miyamoto, N., Oki, R., Ueda, H., Ishigami, M., Hir-
Weinstein, M.M., Yin, L., Tu, Y., Wang, X., Wu, X., Castellani, L.W., Walzem, aoka, H., Matsuzawa, Y., and Kihara, S. (2013). Novel combined GPIHBP1
R.L., Lusis, A.J., Fong, L.G., Beigneux, A.P., et al. (2010b). Chylomicronemia mutations in a patient with hypertriglyceridemia associated with CAD.
elicits atherosclerosis in mice–brief report. Arterioscler. Thromb. Vasc. Biol. J. Atheroscler. Thromb. 20, 777–784.
30, 20–23.
Zhang, L., Lookene, A., Wu, G., and Olivecrona, G. (2005). Calcium triggers
Winkler, F.K., D’Arcy, A., and Hunziker, W. (1990). Structure of human pancre- folding of lipoprotein lipase into active dimers. J. Biol. Chem. 280,
atic lipase. Nature 343, 771–774. 42580–42591.

Cell Metabolism 30, July 2, 2019 65


Lead
with
purpose
:HSXWVFLHQFHðUVW(YHU\WKLQJZHGRDLPVWRGULYHUHVHDUFKIRUZDUGò
ZKHWKHULWâVZRUNLQJZLWKDXWKRUVHQKDQFLQJWKHSHHUUHYLHZSURFHVV
RUGHYHORSLQJLQQRYDWLRQVLQSXEOLVKLQJ
:HFKDPSLRQWUDQVIRUPDWLYHDQGJURXQGEUHDNLQJLGHDV7KHDXWKRUDQG
WKHLUUHVHDUFKDUHDWRXUFHQWHU2XUHGLWRULDOWHDPLVNQRZOHGJHDEOHDQG
HQJDJHG2XUSURFHVVHVDUHWKRXJKWIXODQGIDLU
:HEHOLHYHWKDWLI\RXOHDGZLWKSXUSRVH\RXFDQLQVSLUHIXWXUHGLUHFWLRQV
:HKRSH\RXâOOMRLQXV/HWâVPDNHVFLHQFHEHWWHU

John Pham is the new Editor-in-Chief of Cell, the fourth in the journal’s 44-year-old history. He sees strong opportunities for Cell
and Cell Press to attract and publish the best science and to help push science forward.
Cell Metabolism

Short Article

Age Mosaicism across Multiple


Scales in Adult Tissues
Rafael Arrojo e Drigo,1 Varda Lev-Ram,2 Swati Tyagi,1 Ranjan Ramachandra,3 Thomas Deerinck,3 Eric Bushong,3
Sebastien Phan,3 Victoria Orphan,4 Claude Lechene,5 Mark H. Ellisman,2,6,7 and Martin W. Hetzer1,7,8,*
1Salk Institute for Biological Studies, Molecular and Cell Biology Laboratory (MCBL), La Jolla, CA, USA
2Department of Pharmacology, University of California, San Diego School of Medicine (UCSD), La Jolla, CA, USA
3National Center for Microscopy and Imaging Research (NCMIR), University of California, San Diego (UCSD), La Jolla, CA, USA
4Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, CA, USA
5Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
6Department of Neurosciences, University of California, San Diego School of Medicine (UCSD), La Jolla, CA, USA
7These authors contributed equally
8Lead Contact

*Correspondence: hetzer@salk.edu
https://doi.org/10.1016/j.cmet.2019.05.010

SUMMARY time for the majority of neurons, cardiomyocytes, and all inner
hear hair cells (De Anda et al., 2016; Brann and Firestein,
Most neurons are not replaced during an animal’s 2014; Foglia and Poss, 2016; Steinhauser et al., 2012; Zhang
lifetime. This nondividing state is characterized by et al., 2012). In some cases, somatic stem cells can respond
extreme longevity and age-dependent decline of to tissue damage and proliferate according to tissue-specific
key regulatory proteins. To study the lifespans of needs, as in the striated muscle, which can somewhat regen-
cells and proteins in adult tissues, we combined erate after wound because of activation of its satellite stem
cells (Blau et al., 2015). A similar pattern is also observed at
isotope labeling of mice with a hybrid imaging
the proteome level, where proteins have different lifespans,
method (MIMS-EM). Using 15N mapping, we show
ranging from hours and days to years (Ori et al., 2015; Toyama
that liver and pancreas are composed of cells with et al., 2013). Like stressed and/or damaged cells, misfolded
vastly different ages, many as old as the animal. and damaged proteins must be degraded and replaced
Strikingly, we also found that a subset of fibroblasts with new and functional versions (Taylor and Dillin, 2011).
and endothelial cells, both known for their replicative Over the last decades these insights steered biomedical
potential, are characterized by the absence of cell di- research toward focusing on cellular and molecular replace-
vision during adulthood. In addition, we show that ment processes.
the primary cilia of beta cells and neurons contains It remains poorly understood how neurons, cardiomyocytes,
different structural regions with vastly different life- and potentially other long-lived cells (LLCs), maintain functional
spans. Based on these results, we propose that integrity and protein homeostasis over the span of several
decades. Because these cells are almost never replaced, they
age mosaicism across multiple scales is a funda-
are essentially as old as the animal itself and must function
mental principle of adult tissue, cell, and protein
properly throughout life, which in humans can be more than a
complex organization. century (De Anda et al., 2016). Understanding how functionality
is maintained in LLCs is important given that aging is associated
with physiological impairments in these kinds of cells (e.g.,
INTRODUCTION neurons and cardiomyocytes) (D’Angelo et al., 2009; Mattson
and Magnus, 2006).
The lifespan of a terminally differentiated cell is quite variable Recent advances in whole-animal metabolic-labeling strate-
among organs: 3 to 4 days for epithelial intestinal cells, to ol- gies and quantitative mass spectrometry (MS/MS) have enabled
factory neuronal cells replaced from basal stem cells, to a life system-wide, high-resolution analyses of global protein turnover

Context and Significance

Some cells in the body, such as skin, are constantly being replaced during one’s lifetime, while most brain cells are not. Cells
with minimal to no turnover contain long-lived proteins whose function declines with age. Researchers at the Salk Institute
and University of California, San Diego, in California used a detailed isotope-tracing method to measure and map the age of
cells and protein in different mouse organs. Unexpectedly, most organs are a mix of cells and proteins of vastly different
ages, regardless of whether they are slow or quick to regenerate. Some cells also contain proteins of different ages. Under-
standing the principles of cellular aging will be important in helping combat the age-associated decline in organ function.

Cell Metabolism 30, 343–351, August 6, 2019 ª 2019 Elsevier Inc. 343
rates. However, imaging approaches for studying extreme cell have been replaced through cell division during adulthood. In
longevity are not well developed. We recently combined stable the liver, contrary to what has been inferred from liver regenera-
isotope metabolic pulse-chase labeling of rats using 15N (herein tion models, we discovered that most hepatocytes, bile duct
called 15N-SILAM [stable isotope labeling in mammals]) with cells, and stellate-like cells (i.e., hepatic stellate cells and Kupffer
quantitative MS/MS to discover a class of long-lived proteins cells) are LLCs. Age mosaicism can also be observed among
(LLPs) in neurons (Toyama et al., 2013). These neuronal LLPs capillary endothelial cells, which exhibit vastly different lifespans
localize primarily to the nuclear pore complex (NPC) and chro- in different tissue such as the brain, pancreas, and liver. Further-
matin, and are maintained with no or limited turnover, in striking more, age mosaicism also occurs at the molecular level, where
contrast to the majority of the proteome, which is renewed within proteins with vastly different lifespans coexist at the supramolec-
hours or days (Ori et al., 2015; Schoenheimer, 1942). Only a few ular level in the basal body and axoneme of the primary cilium of
other LLPs have been previously identified, including lens crys- neurons and beta cells.
talline, collagen, and myelin basic protein (Fischer and Morell,
1974; Lynnerup et al., 2008; Verzijlbergen et al., 2010). These RESULTS AND DISCUSSION
proteins deteriorate with age and, with the exception of myelin,
are unlikely to contribute to cellular aging because they typically Identification of LLCs and Structures in the Central
reside in cells with minimal metabolic activity and/or play struc- Nervous System (CNS) with MIMS-EM
tural roles (Lynnerup et al., 2008). In contrast, the neuronal LLPs Information about the lifespan of different cells is still lacking for
that we identified play critical roles in gene regulation and nuclear many adult tissues, largely because of technical limitations. To
trafficking pathways and age-dependent loss of these LLPs im- determine which and whether cells divide or persist in a nondi-
pairs nuclear function (D’Angelo et al., 2009; Ibarra et al., 2016; viding state throughout life we combined 15N metabolic labeling
Toyama et al., 2013). of mice with MIMS-EM (Steinhauser et al., 2012). In brief, we
LLCs face a constant lifelong demand for performance to used animals initially intended for protein MS/MS as in our
maintain organ function and homeostasis and are constantly previous studies (Toyama et al., 2013). As such, molecular com-
exposed to drivers of molecular and cellular damage. In fact, ponents of these animals (i.e., nucleic and amino acids) were
the accumulation of such damage over time may eventually alter labeled with 15N throughout their embryonic development and
gene expression and protein homeostasis pathways, leading to fed an 15N-rich diet until the 21st or 45th day of post-natal devel-
impaired cellular function and disease (D’Angelo et al., 2009; opment (see details in STAR Methods). We refer to these mice as
Ibarra et al., 2016). Understanding these mechanisms requires P21- or P45-15N-SILAM mice, respectively. After the 15N-label-
the identity and distribution pattern of LLCs in different organs. ing period, 15N-animals were chased with a normal diet with
However, lifelong cell persistence has not been analyzed in a the natural 99.4% content of 14N for 6, 18, or 26 months of
systematic and quantitative manner, and therefore fundamental age. During the chase period, when 15N-labeled cells are
questions about cell type-specific turnover rates remains poorly replaced by newly synthesized cells, 15N is replaced by 14N.
characterized. In addition, while implementation of 15N-SILAM Therefore, the rate of cell (and protein turnover) can be quantified
together with quantitative MS/MS allowed us to identify LLPs with MIMS by simultaneously imaging 15N and 14N in a target re-
(Toyama et al., 2013), this approach lacks spatial information gion and determining the 15N/14N ratio (i.e., old-to-young ratio).
(other than the region of tissue that was dissected) and does Importantly, our previous work has shown that the relative
not provide any information regarding cell identities. To over- amount of nuclear 15N-labeled protein represents less than 2%
come this challenge, we made use of 15N-SILAM-labeled of the whole nuclear proteome after 12 months of chase (Toyama
animals initially labeled for MS/MS to develop a new imaging et al., 2013). Therefore, the vast majority of the nuclear 15N is
pipeline for correlated scanning electron microscopy (SEM) expected to stem from 15N-DNA and which decreases by 50%
and multi-isotope imaging mass spectroscopy (MIMS) (herein after each cell division, as expected (Steinhauser et al., 2012).
called MIMS-electron microscopy [EM]). MIMS is a mapping Since our SILAM approach cannot distinguish between 15N-
technique compatible with electron microscopy that can deter- DNA and rare 15N-proteins in the nucleus, we used MIMS-EM
mine the relative amount of different stable isotopes incorpo- to quantitatively determine the 15N content of random sections
rated into nucleic acids and proteins in cells and tissues of neuronal nuclei in different regions of the CNS. This allowed
(Lechene et al., 2006; Steinhauser et al., 2012; Zhang et al., us to establish a mean baseline and range of the nuclear 15N
2012). Given the high spatial resolution and sensitivity of signal that is characteristic for nondividing cells (De Anda
EM and MIMS, we used MIMS-EM to visualize and quantify et al., 2016; D’Angelo et al., 2009). We targeted neurons in the
cell and protein turnover in the brain, liver, and pancreas in young mouse layer-2 (L2) in the motor cortex and in the rat cerebellum
and old 15N-SILAM mice and rats. from animals labeled with 15N until P45 and chased for 6 months.
Using MIMS-EM on tissue samples from exceptionally long- MIMS-EM of the mouse L2 cortex showed that most of the 15N
lived mice initially labeled for stable isotope mass spectrometry signal was nuclear or overlapping with myelin sheaths, as ex-
experiments (Toyama et al., 2013), we found that neurons and pected (Figure S1C) (Toyama et al., 2013). We measured whole
cardiomyocytes are not the only cell types with exceptional nuclei 15N/14N ratios and found that they were markedly higher
longevity. Much to our surprise, adult mouse liver and pancreas than the natural occurrence (15N/14N = 37 3 104) because of a
contain populations of cells with vastly different lifespans, many significant retention of 15N in all neuronal nuclei mapped (Figures
of which are as old as neurons. The pancreas serves a paradigm 1A–1I and S1A–S1C). Since we observed species-specific differ-
for the observed cellular age mosaicism, in that a subset of ences (Figure S1E), we therefore decided to focus the rest of our
insulin-beta cells are as old as cortical neurons, while others analysis on mouse tissues.

344 Cell Metabolism 30, 343–351, August 6, 2019


Figure 1. MIMS-EM of LLCs and Structures in the CNS
(A and B) SEM (A) and MIMS (B) of two capillaries in the optic nerve head (ONH) of a 6-month chase mouse. An endothelial cell nucleus (yellow arrow) and myelin
sheaths (pink arrows) are indicated. A pericyte nucleus is visible to the left the capillary lumen.
(C and D) SEM (C) and MIMS (D) of a capillary in the ONH. An old endothelial cell nucleus (yellow arrow), a fibroblast (pink arrow) and 15N-rich extracellular matrix
(ECM) (white arrow) are indicated.
(E and F) SEM (E) and MIMS (F) of a capillary in the ONH, with an old fibroblast (top) and a young endothelial cell (nucleus delineated in white and indicated by the
yellow asterisk).
(G and H) SEM (G) and MIMS (H) of an L2 neuron (top) and an endothelial cell.
(I) Close-up of granular cells in the rat cerebellum with an old endothelial cell visible. Full mosaic is shown in Figure S1B.
Scale bars: 5 mm (A, C, E, and G) (SEM). At the bottom of the MIMS images, the heatmap shows the 15N/14N 3 104 and scaled with a hue saturation intensity (HSI).

It is well established that cortical neurons do not divide during namely 15N/14N ratios at least 5.5-fold over the natural ratio
adulthood, thus we decided to use the mean 15N/14N ratio from (i.e., 15N/14N > 208, Figure S1F). Importantly, the relative
random nuclear sections of L2 neurons as baseline level for the age of non-neuronal cells in nervous and somatic organs
identification of nondividing cells. We observed that the mean was determined by comparing their 15N/14N with that of L2
15
N/14N in the L2 neuron population analyzed varied between neurons from P45- or P21-SILAM (e.g., cells from P45-SILAM
5- and 19-fold higher than the natural ratio (Figure S3D). This animals were benchmarked against P45-SILAM L2 neurons).
variation is likely explained by the random occurrence of dense Of note, while this classification approach can determine the
heterochromatin patches in the nuclear sections imaged with relative longevity of ‘‘old’’ cells after 6, 18, or 26 months of
MIMS-EM, whereby sections with a higher density of hetero- chase, it cannot determine their proliferative potential during
chromatin in a given section are expected to yield higher 15N the pulse period or how many times a cell has divided during
signals due to a relatively higher concentration of 15N-DNA the chase period (see below, Limitations of Study). Neverthe-
in comparison with euchromatin-rich sections. In fact, we less, we found that cells in the inner nuclear layer of the retina;
observed a maximum level of 15N labeling in intranuclear regions and most endothelial cells in the mouse optic nerve head
that correlate with dense patches of heterochromatin, which (ONH) and all in the cerebellum were LLCs and as old as
may also contain rare 15N-proteins in addition to 15N-DNA (Fig- neurons, and did not show any sign of turnover (Figures 1A–
ures 1G, 1H, S1A, S1D, and S1E). 1F and S1A–S1D). In fact, we observed retention of 15N in
Accordingly, we decided to classify cells as ‘‘old’’ when endothelial cells located in the L2 cortical layer of a
their nuclear mean 15N/14N ratio was similar to L2 neurons, P21-15N-SILAM mouse chased for almost 26 months (Figures

Cell Metabolism 30, 343–351, August 6, 2019 345


1G and 1H). In addition, the nuclear 15N/14N ratio in peri- estimate that hepatocytes could indeed last for at least 1
vascular fibroblasts and an oligodendrocyte was >20-fold year (Magami et al., 2002; Malato et al., 2011).
higher than the natural 15N/14N ratio, similar to neurons (Fig-
ures 1A–1F and 4D). These data suggest that lifelong cell Adult Pancreas is Composed of Cells with Vastly
persistence extends to non-neuronal cells, is more prominent Different Lifespans
than previously thought and also confirms a recent report Having identified blood vessels containing endothelial cells with
regarding oligodendrocyte longevity (Tripathi et al., 2017). In different ages, we asked whether age mosaicism is a more gen-
contrast, we found one example of a capillary endothelial eral principle and turned our attention to the pancreas, which
cell in the ONH that had close-to-background 15N levels contains exocrine and endocrine cells. Development and matu-
(Figures 1E and 1F), which indicates that turnover of endothe- ration of pancreatic endocrine and exocrine cells extends to the
lial cells in the brain can happen, although it may be a rare early postnatal period when cells are mostly generated by self-
event. Together, these data from these exceptionally long- duplication (Dor et al., 2004; Houbracken and Bouwens, 2017).
lived SILAM mice provides the first evidence that the Here, we used MIMS-EM to determine the longevity and turnover
neurovascular unit, and, more specifically, endothelial cells of the endocrine and exocrine pancreas in adult animals labeled
in the brain, are mostly old. However, it also suggests that until two different time points in early life (post-natal day 21 [P21]
some vessels are mosaics, built of cells with vastly different or P45, shown in Figures 3A and 3B) and chased for almost
lifespans (Figures 1A–1F). We conclude that MIMS-EM is a 2 years. First, we analyzed islets from a P21-15N-SILAM mouse
powerful tool to study cellular lifespan and we decided to chased with 14N for 26 months and observed that the turnover
explore the concept of age mosaicism in other tissues rate of alpha, beta, and delta cells is not uniform. As expected,
(see below). we observed a large number of alpha and beta cells exhibiting
background 15N (representing 77% and 76%, respectively) (Fig-
Cellular Age Mosaicism in the Liver ures 3F and S3D), likely reflecting the wave of alpha and beta cell
Several studies in the context of tissue injury have led to the proliferation that takes place in neonates and is important for
well-established concept that the liver has a high regenerative beta cell maturation (Stolovich-Rain et al., 2015). Strikingly,
capacity (Malato et al., 2011). In line with this concept, our pre- almost a quarter of alpha and beta cells (representing 23% and
vious 15N-SILAM MS/MS experiments in rats did not identify 24%, respectively) and all delta cells had 15N levels similar to
15
N-labeled peptides in the liver after only a 4-month chase, neurons (Figure 3F), and therefore suggest that a subpopulation
which further suggested that these cells are replaced within of islet endocrine cells can become LLCs before or at the wean-
months (Toyama et al., 2013). However, little is known about ing stage and may not proliferate for an entire lifetime.
the longevity of hepatocytes and other key liver cell types in Next, we analyzed islets from a P45-15N-SILAM mouse
healthy adult mice (Magami et al., 2002; Malato et al., 2011). chased for 18 months and observed that most beta cells (61%)
We applied MIMS-EM to map cells located near bile ducts, had 15N levels approximately 10-fold higher than the natural ra-
portal or central veins, and sinusoid capillaries (Figures 2A tio, similar to L2 neurons after 18 months of chase (Figure S3D),
and 2B), and found that most hepatocytes in P45-15N-SILAM while 39% of beta cells had lower 15N levels and therefore had
mice were as old as neurons. Long-lived hepatocytes were proliferated during adulthood (Figures 3B–3G and S3D). Recent
observed in the vicinity of central veins, portal veins, and capil- data indicate that different types of beta cells exist in the islet,
lary sinusoids after 6 and 18 months of chase (92% and 95%, including proliferation-prone and/or immature beta cells located
respectively) (Figures 2A, 2B, and S2A–S2D). Similarly, MIMS- in a neogenic niche at the islet periphery (Bader et al., 2016; van
EM revealed that most cholangiocytes were old as neurons der Meulen et al., 2017). We mapped the location of proliferative
and, therefore, are also LLCs (red arrows in Figures 2A, 2B, beta cells in relation to the islet periphery and found no correla-
2D, 2F, and S2A–S2D). In contrast, while endothelial cells in tion between beta cell age and intraislet cellular location (Fig-
portal and central veins did not divide (Figures 2A–2C, white ar- ure S3E), thus suggesting that beta cell proliferation is random
rows; Figures S2A–S2D), most sinusoid capillary endothelial within the islet ultrastructure. Similar results were observed for
cells contained 15N levels at the 18-month time point that alpha cells, pancreatic endothelial cells, and stellate cells, which
were lower than at the 6-month time point (Figures 2A, 2B, were also found to be mostly LLCs (Figures 3B–3G and S3D–
and 2E, white arrows; Figures S2A–S2D). A similar phenotype S3F). In striking contrast, all delta cells and the majority of ductal
was observed in stellate-like cells (Figures 2A, 2B, and 2F, cells did not show age mosaicism, and their 15N levels were
green arrows; Figures S2A–S2D). These results indicate that, similar to neurons (Figures 3A–3G and S3B–S3D). In contrast,
while hepatocytes remain largely quiescent throughout life, most acinar cells we observed had close to or background 15N
endothelial cells in hepatic sinusoids and most stellate-like cells levels and thus have clearly undergone significant turnover
undergo a major turnover event between 6 and 18 months of during adulthood (Figures 3A and 3B, white arrow; Figure S3D),
life. Lastly, the exceptional longevity of liver cells was associ- as expected (Houbracken and Bouwens, 2017). Notably, the
ated with old extracellular matrix elements located to the peri- observation of long-lived acinar cells suggests that some acinar
vascular space of portal and central veins and interstitial space cells become long-lived in early adult life (Figure 3G, pink arrow),
(Figures 2A–2D and S2B). These data suggest that the aging which could have a role in pancreatic cancer (Pan et al., 2013;
liver has a tissue architecture characterized by an intercellular Westphalen et al., 2016; Wollny et al., 2016).
age mosaicism, where a young sinusoid vasculature would Together, these data indicate that the islet is a mosaic
support the function of relatively older cells, a notion that is composed of cells with vastly different lifespans, where most
further supported by hepatocyte lineage tracing studies that alpha and beta cells assume a quiescent and LLC phenotype

346 Cell Metabolism 30, 343–351, August 6, 2019


Figure 2. LLCs in the Liver
(A and B) SEM and MIMS of cells near bile ducts, central veins or capillary sinusoids from a 15N-SILAM P45 mouse chased for 6 months (A) or 18 months (B). ECM
(yellow arrowheads), hepatocytes (pink arrowheads), cholangiocytes (red arrowheads), stellate-like (green arrowheads), and endothelial cells (white arrowheads)
are indicated. Cv, central vein; Bd, bile duct; c, capillary.
(C–F) Relative turnover of liver hepatocytes (C), cholangiocytes (D), sinusoid endothelial cells (E), and hepatic stellate-like cells (HSCs) (F) after a 6- or 18-month
(mo) chase. The total number of cells analyzed for each cell type is indicated underneath each pie chart. At the bottom of the MIMS images, the heatmap shows
the 15N/14N 3 104 and scaled with a HSI. Scale bar, 10 mm.

between weaning (P21) and the first month (P45) of adult life. Visualization of LLP Assemblies In Vivo
Moreover, it suggests that proliferation of alpha and beta cells Our previous SILAM MS/MS experiments revealed that LLCs do
is not uniform throughout adulthood, contrary to what earlier contain a subset of proteins that persist throughout life. Specif-
studies have indicated (Brennand et al., 2007; Dor et al., 2004). ically, we found that scaffold NPC proteins are maintained in
While the specific differences between young and old beta or the nuclear envelope throughout adulthood in neurons (Savas
alpha cells remains to be understood, our data adds to recent et al., 2012; Toyama et al., 2013). However, it was unclear
studies that have begun to investigate the cellular identity, whether this exceptional longevity extended to other subcellular
the heterogeneity and aging of beta cell subpopulations and components. While studying long-lived beta cells, we observed
its potential role in age-associated type II diabetes onset the retention of significant 15N signal by a small, ‘‘star-shaped’’
(Aguayo-Mazzucato et al., 2017; Almaça et al., 2014; Basu structure within the cytoplasm of a beta cell from a P45-15N-
et al., 2003; Johnston et al., 2016; van der Meulen et al., 2017; SILAM mouse (data not shown). We hypothesized that this signal
Segerstolpe et al., 2016). was associated with the basal body (Bb) of the cell’s primary

Cell Metabolism 30, 343–351, August 6, 2019 347


Figure 3. LLCs in the Pancreas
(A and B) SEM (A) and MIMS (B) of a cross-section of the islets of Langerhans. Old and young acinar cells are indicated by pink and white arrow, respectively.
Yellow dotted box highlights cells shown in (C).
(C) Enlarged view of boxed region in (A) and (B). SEM and MIMS of beta cells (yellow arrows) and an old alpha cell (pink arrow).
(D) An old delta cell (left) next to a younger beta cell (top right).
(E) SEM and MIMS of a young (bottom) and an old (top right) endothelial cell. Old pancreatic stellate cells are seen in the top and lower right corners.

(legend continued on next page)

348 Cell Metabolism 30, 343–351, August 6, 2019


Basal Basal
Tz body Tz body
A Pm B C

Tz Basal 1.5-2x 1.5-2x 2-3x


body
basal root
microtubules
transition fibers
D 5-10 10-25 25-50 50-100x 0nm 80nm 160nm 240nm

320nm 400nm 480nm 560nm

Figure 4. Longevity of Primary Cilia in LLCs


(A) Cartoon of a primary cilium Bb. Microtubules (black), transition fibers (cyan), basal root (pink), transition zone (Tz), axoneme, and plasma membrane (Pm)
are shown.
(B and C) Two different sections of a beta cell primary cilium from a P21-15N-SILAM mouse chased for 26 months and imaged with MIMS-EM. Top rows: SEM
micrographs with visible transition zone, Bb, transition fibers (cyan arrows), and basal root (pink arrows). Bottom rows: overlay of SEM micrographs and 15N/14N
thresholds: 1.53–23 (green) and 23–33 (cyan) the natural 15N/14N of 37 3 104.
(D) SEM micrograph overlaid with thresholded 15N/14N of an L2 neuron from a P45-15N-SILAM mouse chased for 6 months. The yellow box indicates the Bb.
On the right are serial sections of the neuron Bb and primary cilium. 15N/14N thresholds: 53–103 (green), 103–253 (cyan), 253–503 (yellow), and 503–1003
(magenta) the natural 15N/14N. At the bottom of the MIMS images, the heatmap shows the 15N/14N 3 104 and scaled with a HSI.
Scale bars: 300 nm (B), 1 mm (D), 500 nm (D, inset).

cilium (Figure 4A), which is a signaling organelle important these long-lived components remains to be discovered, it is note-
for beta-cell function (Gerdes et al., 2014). We tested this worthy that long-lived nucleoporins, including Nup93, are known
hypothesis by imaging different beta-cell BBs from P21- and to form a permeability barrier in the ciliary gate in the cilium tran-
P45-15N-SILAM mice chased for 18 and 26 months, respectively. sition zone (McClure-Begley and Klymkowsky, 2017). These
We applied MIMS-EM to serial cross-sections of different primary exciting findings raise the question of whether aging is associated
cilia within beta cells and discovered that, indeed, the Bb con- with impaired primary cilium function due to loss of LLPs, as seen
tained high 15N levels, while the rest of the cilium has been re- for NPCs in old neurons (D’Angelo et al., 2009).
placed by new components (Figures 4B, 4C, S4C, and S4D). Analogous to genetic mosaicism, which refers to the presence
We expanded this analysis to L2 neurons, where we observed of two or more populations of cells with different genotypes in one
that the 15N signal was not limited to the Bb and was also found individual, we propose the concept of age mosaicism as a key
in the axoneme, decreasing as the cilium projected toward the feature of adult tissue organization. Our data indicate that non-
extracellular space (Figure 4D). These findings provide evidence uniform turnover of cells and proteins gives rise to tissue- and
of the occurrence of protein age mosaicism within one subcellular cell-specific aging architectures characterized by a mosaic orga-
structure, while suggesting that long-lived structures are present nization of young and old elements at the cell and protein level.
in the primary cilium and that their turnover could be different in These data provide insight into an organization pattern in biology
beta cells and neurons (Figures 4B–4D). While the identity of that forms an age mosaic across the mesoscale, where the close

(F) Relative turnover in percentages of cells that are as old (gray) or younger (white) than L2 neurons from 15N-SILAM P21 mouse chased for 26 months.
(G) Same as in (F), but from a 15N-SILAM P45 mouse chased for 18 months. The total number of cells analyzed for each cell type is listed underneath each pie
chart. At the bottom of the MIMS images, the heatmap shows the 15N/14N 3 104 and scaled with an HSI.
Scale bars: 5 mm (A, C, and E) and 2.5 mm (D).

Cell Metabolism 30, 343–351, August 6, 2019 349


relationship of young and old components is found in protein su- B Processing of Samples for Electron Microscopy
per complexes and organelles (i.e., Bb in the primary cilium), to B X-Ray Microscopy (XRM)
cells (e.g., beta cells and hepatocytes) and tissues (e.g., brain, B Correlative Electron Microscopy and Multi-isotope
liver, and pancreas). This study raises the exciting possibility Mass Spectroscopy (MIMS)
that the age-dependent decline in LLPs is not limited to neurons, B MIMS-EM Image Analysis and Display
but may be a widespread phenomenon for all tissues rich in LLCs d QUANTIFICATION AND STATISTICAL ANALYSIS
15 14
and links the loss of LLPs with the onset of age-related disease B Quantification of N/ N Ratio in Tissues and Cell Age
such as diabetes (Basu et al., 2003). More broadly, future SILAM Classification
studies will contain the use of different stable isotope-labeled B Cell Type Classification in Electron Micrographs
molecules (i.e., 15N-thymidine for DNA and 18O-Leucine for pro- d DATA AND SOFTWARE AVAILABILITY
teins) to help identify and functionally characterize other exam- d ADDITIONAL RESOURCES
ples of heterogeneity in the aging of cells and proteins in different
organs and in health and disease. SUPPLEMENTAL INFORMATION

Limitations of Study Supplemental Information can be found online at https://doi.org/10.1016/j.


cmet.2019.05.010.
Our pulse-chase labeling approach using 15N-enriched food can
provide information about protein identity using MS/MS ana-
lyses (Toyama et al., 2013). However, our experimental set up ACKNOWLEDGMENTS

using MIMS-EM cannot distinguish between 15N-labeled nucleic


This work was supported by grants to M.W.H. from the NIH Transformative
acids and amino acids in the regions we investigated. Although Research Award (R01 NS096786), the Keck Foundation, and the NOMIS Foun-
most of the 15N signal shown in brain, liver, and pancreatic cells dation. It was also supported by grants to M.H.E. and V.L.-R. from the NIH
is most likely due to 15N-DNA, we cannot rule out a potential NINDS (NIH RO1 NS027177-30), which supported the creation of the 15N/14N
small contribution of 15N-peptides from intranuclear LLPs mice; and the NIH NIGMS (5P41 GM103412-29), which supports the advanced
(Toyama et al., 2013). However, regardless of the nature of the technologies and instrument development activities of the NCMIR, most heavily
15 used, and whose new methods developments were partly driven by this work.
N signal our results suggest that these cells are long-lived.
V.O. and M.H.E. are also supported to develop and carry out correlated light
Second, in its current stage, MIMS-EM is not a high-throughput and electron microscopy with MIMS under an award from the US Department
imaging technique as it is time and labor intensive, requiring cells of Energy, Office of Science, Office of Biological and Environmental Research
and tissues to be cut in thin sections (50–250 nm in thickness) (DE-SC0016469). R.A.eD. is supported by an American Diabetes Association
and long acquisition times (e.g., 0.5–48 h for a 30 3 30-mm postdoctoral fellowship (1-18-PMF-007) and benefitted from assistance from
raster), thus making volumetric imaging of any number of cells Daniela Rhodes and the Nanyang Institute of Structural Biology (NISB) (at the
Nanyang Technological University, Singapore) from a career development sti-
very challenging. As a result, this study was limited by the acqui-
pend. The authors are also thankful to Yunbin Guan, Ph.D., from the Caltech
sition of random nuclear sections, which can add variability to
Microanalysis Center in the Division of Geological and Planetary Sciences, Cal-
the 15N measurements by the random distribution of heterochro- ifornia Institute of Technology for technical support and assistance with MIMS
matin-rich domains in the sections analyzed. Nevertheless, we image acquisition; to Dr. Ting-Di Wu and Dr. Jean-Luc Guerquin-Kern from the
have imaged >700 cells in total with MIMS-EM to provide an INSERM, Université Paris-Sud, Paris; and the Cell and Tissue Imaging Facility of
important insight into the location and identity of LLCs outside the Institut Curie (PICT), a member of the France BioImaging National Infrastruc-
the brain. MIMS-EM is a very sensitive and quantitative tech- ture (ANR-10-INBS-04), for the use of Curie NanoSIMS instrument; Greg
McMahon from the National Center of Excellence in Mass Spectrometry Imag-
nique with multiplexed detection capabilities, and therefore
ing, National Physical Laboratory (NPL), UK; and to David O’Keefe (Salk Insti-
future applications of MIMS-EM will take advantage of a tute) for critical inputs on manuscript editing and revision.
SILAM approach that labels nucleic acids, amino acids, and fatty
acids with different stable isotopes (15N, 18O, and 13C, for
AUTHOR CONTRIBUTIONS
example). Together with improved tools for cell/tissue mapping
and data acquisition pipelines, MIMS-EM can be used to inves- R.A.eD. collected and analyzed the data and wrote the article. V.L.-E., S.T.,
tigate a large number of cells and intracellular components from R.R., T.D., E.B., S.P., and C.L. collected the data. V.O., M.H.E., and M.W.H.
designed the study and wrote the article.
cells in virtually any SILAM tissue, including humans (Stein-
hauser et al., 2012).
DECLARATION OF INTERESTS

STAR+METHODS The authors declare no competing interests.

Detailed methods are provided in the online version of this paper Received: June 14, 2018
Revised: October 18, 2018
and include the following:
Accepted: May 11, 2019
Published: June 6, 2019
d KEY RESOURCES TABLE
d CONTACT FOR REAGENT AND RESOURCE SHARING
REFERENCES
d EXPERIMENTAL MODEL AND SUBJECT DETAILS
B Animal Studies
Adams, S.R., Mackey, M.R., Ramachandra, R., Palida Lemieux, S.F.,
d METHOD DETAILS Steinbach, P., Bushong, E.A., Butko, M.T., Giepmans, B.N.G., Ellisman,
B Stable Isotope Metabolic Labeling of Mam- M.H., and Tsien, R.Y. (2016). Multicolor electron microscopy for simultaneous
mals (SILAM) visualization of multiple molecular species. Cell Chem. Biol. 23, 1417–1427.

350 Cell Metabolism 30, 343–351, August 6, 2019


Aguayo-Mazzucato, C., van Haaren, M., Mruk, M., Lee, T.B., Crawford, C., €rmann, N., Ng, R., Wang, B., Zape, J., Kay, M.A.,
Malato, Y., Naqvi, S., Schu
Hollister-Lock, J., Sullivan, B.A., Johnson, J.W., Ebrahimi, A., Dreyfuss, Grimm, D., and Willenbring, H. (2011). Fate tracing of mature hepatocytes in
J.M., et al. (2017). Beta cell aging markers have heterogeneous distribution mouse liver homeostasis and regeneration. J. Clin. Invest. 121, 4850–4860.
and are induced by insulin resistance. Cell Metab. 25, 898–910. Mattson, M.P., and Magnus, T. (2006). Ageing and neuronal vulnerability. Nat.
Almaça, J., Molina, J., Arrojo e Drigo, R., Abdulreda, M.H., Jeon, W.B., Berggren, Rev. Neurosci. 7, 278–294.
P.O., Caicedo, A., and Nam, H.G. (2014). Young capillary vessels rejuvenate McClure-Begley, T.D., and Klymkowsky, M.W. (2017). Nuclear roles for cilia-
aged pancreatic islets. Proc. Natl. Acad. Sci. USA 111, 17612–17617. associated proteins. Cilia 6, 8.
Bader, E., Migliorini, A., Gegg, M., Moruzzi, N., Gerdes, J., Roscioni, S.S., Ori, A., Toyama, B.H., Harris, M.S., Bock, T., Iskar, M., Bork, P., Ingolia, N.T.,
Bakhti, M., Brandl, E., Irmler, M., Beckers, J., et al. (2016). Identification of pro- Hetzer, M.W., and Beck, M. (2015). Integrated transcriptome and proteome
liferative and mature b-cells in the islets of Langerhans. Nature 535, 430–434. analyses reveal organ-specific proteome deterioration in old rats. Cell Syst.
Basu, R., Breda, E., Oberg, A.L., Powell, C.C., Dalla Man, C., Basu, A., Vittone, 1, 224–237.
J.L., Klee, G.G., Arora, P., Jensen, M.D., et al. (2003). Mechanisms of the age- Pan, F.C., Bankaitis, E.D., Boyer, D., Xu, X., Van de Casteele, M., Magnuson,
associated deterioration in glucose tolerance: contribution of alterations in in- M.A., Heimberg, H., and Wright, C.V.E. (2013). Spatiotemporal patterns of mul-
sulin secretion, action, and clearance. Diabetes 52, 1738–1748. tipotentiality in Ptf1a-expressing cells during pancreas organogenesis and
Blau, H.M., Cosgrove, B.D., and Ho, A.T.V. (2015). The central role of muscle injury-induced facultative restoration. Development 140, 751–764.
stem cells in regenerative failure with aging. Nat. Med. 21, 854–862. Savas, J.N., Toyama, B.H., Xu, T., Yates, J.R., and Hetzer, M.W. (2012).
Brann, J.H., and Firestein, S.J. (2014). A lifetime of neurogenesis in the olfac- Extremely long-lived nuclear pore proteins in the rat brain. Science 335, 942.
tory system. Front. Neurosci. 13, 112. Schoenheimer, R. (1942). The dynamic state of body constituents. Cancer
Brennand, K., Huangfu, D., and Melton, D. (2007). All b cells contribute equally Res. 2, 810.
to islet growth and maintenance. PLOS Biol. Published May 29, 2007. https:// Segerstolpe, Å., Palasantza, A., Eliasson, P., Andersson, E.M., Andréasson,
doi.org/10.1371/journal.pbio.0050163. A.C., Sun, X., Picelli, S., Sabirsh, A., Clausen, M., Bjursell, M.K., et al. (2016).
D’Angelo, M.A., Raices, M., Panowski, S.H., and Hetzer, M.W. (2009). Age- Single-cell transcriptome profiling of human pancreatic islets in health and
dependent deterioration of nuclear pore complexes causes a loss of nuclear Type 2 diabetes. Cell Metab. 24, 593–607.
integrity in postmitotic cells. Cell 136, 284–295. Sosinsky, G.E., Deerinck, T.J., Greco, R., Buitenhuys, C.H., Bartol, T.M., and
De Anda, F.C., Madabhushi, R., Rei, D., Meng, J., Gra €ff, J., Durak, O., Meletis, Ellisman, M.H. (2005). Development of a model for microphysiological simula-
K., Richter, M., Schwanke, B., Mungenast, A., et al. (2016). Cortical neurons tions: small nodes of Ranvier from peripheral nerves of mice reconstructed by
gradually attain a post-mitotic state. Cell Res. 26, 1033–1047. electron tomography. Neuroinformatics 3, 133–162.
Dor, Y., Brown, J., Martinez, O.I., and Melton, D.A. (2004). Adult pancreatic Steinhauser, M.L., Bailey, A.P., Senyo, S.E., Guillermier, C., Perlstein, T.S.,
beta-cells are formed by self-duplication rather than stem-cell differentiation. Gould, A.P., Lee, R.T., and Lechene, C.P. (2012). Multi-isotope imaging
Nature 429, 41–46. mass spectrometry quantifies stem cell division and metabolism. Nature
Fischer, C.A., and Morell, P. (1974). Turnover of proteins in myelin and myelin- 481, 516–519.
like material of mouse brain. Brain Res. 74, 51–65. Stolovich-Rain, M., Enk, J., Vikesa, J., Nielsen, F.C., Saada, A., Glaser, B., and
Foglia, M.J., and Poss, K.D. (2016). Building and re-building the heart by car- Dor, Y. (2015). Weaning triggers a maturation step of pancreatic b cells. Dev.
diomyocyte proliferation. Development 143, 729–740. Cell 32, 535–545.
Gerdes, J.M., Christou-Savina, S., Xiong, Y., Moede, T., Moruzzi, N., Karlsson- Taylor, R.C., and Dillin, A. (2011). Aging as an event of proteostasis collapse.
Edlund, P., Leibiger, B., Leibiger, I.B., Östenson, C.G., Beales, P.L., et al. Cold Spring Harb. Perspect. Biol. 3, 1–17.
(2014). Ciliary dysfunction impairs beta-cell insulin secretion and promotes Thévenaz, P., Ruttimann, U.E., and Unser, M. (1998). A pyramid approach to
development of type 2 diabetes in rodents. Nat. Commun. 5, 5308. subpixel registration based on intensity. IEEE Trans. Image Process 7, 27–41.
Goldstein, J.I., Lyman, C.E., Newbury, D.E., Lifshin, E., Echlin, P., Sawyer, L., Toyama, B.H., Savas, J.N., Park, S.K., Harris, M.S., Ingolia, N.T., Yates, J.R.,
Joy, D.C., and Micheal, J.R. (2003). Scanning electron microscopy and micro- and Hetzer, M.W. (2013). Identification of long-lived proteins reveals excep-
analysis. Scanning Electron Microsc. X-Ray Microanal. 34, 690. tional stability of essential cellular structures. Cell 154, 971–982.
Herrmann, A.M., Ritz, K., Nunan, N., Clode, P.L., Pett-Ridge, J., Kilburn, M.R., Tripathi, R.B., Jackiewicz, M., McKenzie, I.A., Kougioumtzidou, E., Grist, M.,
Murphy, D.V., O’Donnell, A.G., and Stockdale, E.A. (2007). Nano-scale sec- and Richardson, W.D. (2017). Remarkable stability of myelinating oligodendro-
ondary ion mass spectrometry - a new analytical tool in biogeochemistry cytes in mice. Cell Rep. 21, 316–323.
and soil ecology: a review article. Soil Biol. Biochem. 39, 1835–1850.
van der Meulen, T., Mawla, A.M., DiGruccio, M.R., Adams, M.W., Nies, V.,
Houbracken, I., and Bouwens, L. (2017). Acinar cells in the neonatal pancreas Dólleman, S., Liu, S., Ackermann, A.M., Cáceres, E., Hunter, A.E., et al.
grow by self-duplication and not by neogenesis from duct cells. Sci. Rep. 7. (2017). Virgin beta cells persist throughout life at a neogenic niche within
Ibarra, A., Benner, C., Tyagi, S., Cool, J., and Hetzer, M.W. (2016). pancreatic islets. Cell Metab. 25, 911–926.
Nucleoporin-mediated regulation of cell identity genes. Genes Dev. 30, Verzijlbergen, K.F., Menendez-Benito, V., van Welsem, T., van Deventer, S.J.,
2253–2258. Lindstrom, D.L., Ovaa, H., Neefjes, J., Gottschling, D.E., and van Leeuwen, F.
Johnston, N.R., Mitchell, R.K., Haythorne, E., Pessoa, M.P., Semplici, F., (2010). Recombination-induced tag exchange to track old and new proteins.
Ferrer, J., Piemonti, L., Marchetti, P., Bugliani, M., Bosco, D., et al. (2016). Proc. Natl. Acad. Sci. USA 107, 64–68.
Beta cell hubs dictate pancreatic islet responses to glucose. Cell Metab. 24, Westphalen, C.B., Takemoto, Y., Tanaka, T., Macchini, M., Jiang, Z., Renz,
389–401. B.W., Chen, X., Ormanns, S., Nagar, K., Tailor, Y., et al. (2016). Dclk1 defines
Lechene, C., Hillion, F., McMahon, G., Benson, D., Kleinfeld, A.M., Kampf, quiescent pancreatic progenitors that promote injury-induced regeneration
J.P., Distel, D., Luyten, Y., Bonventre, J., Hentschel, D., et al. (2006). High-res- and tumorigenesis. Cell Stem Cell 18, 441–455.
olution quantitative imaging of mammalian and bacterial cells using stable €ne, D., Ziebell, F.,
Wollny, D., Zhao, S., Everlien, I., Lun, X., Brunken, J., Bru
isotope mass spectrometry. J. Biol. 5, 20. Tabansky, I., Weichert, W., Marciniak-Czochra, A., et al. (2016). Single-cell
Lynnerup, N., Kjeldsen, H., Heegaard, S., Jacobsen, C., and Heinemeier, J. analysis uncovers clonal acinar cell heterogeneity in the adult pancreas.
(2008). Radiocarbon dating of the human eye lens crystallines reveal proteins Dev. Cell 39, 289–301.
without carbon turnover throughout life. PLoS One 3, e1529. Zhang, D.S., Piazza, V., Perrin, B.J., Rzadzinska, A.K., Poczatek, J.C., Wang,
Magami, Y., Azuma, T., Inokuchi, H., Kokuno, S., Moriyasu, F., Kawai, K., and M., Prosser, H.M., Ervasti, J.M., Corey, D.P., and Lechene, C.P. (2012). Multi-
Hattori, T. (2002). Cell proliferation and renewal of normal hepatocytes and bile isotope imaging mass spectrometry reveals slow protein turnover in hair-cell
duct cells in adult mouse liver. Liver 22, 419–425. stereocilia. Nature 481, 520–524.

Cell Metabolism 30, 343–351, August 6, 2019 351


Where big ideas multiply
Cell Reports Physical Science—a new broad-scope, open access journal
from Cell Press—publishes cutting-edge research across the full spectrum
of the physical sciences, including chemistry, physics, materials science,
energy science, engineering, and related interdisciplinary work.

Submit your paper today.

www.cell.com/cell-reports-physical-science
ll

Resource
Patch-Seq Links Single-Cell Transcriptomes
to Human Islet Dysfunction in Diabetes
Joan Camunas-Soler,1,2,8 Xiao-Qing Dai,3,4,8 Yan Hang,5 Austin Bautista,3,4 James Lyon,4 Kunimasa Suzuki,4
Seung K. Kim,5,6,* Stephen R. Quake,1,2,6,7,9,10,* and Patrick E. MacDonald3,4,9,*
1Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
2Chan Zuckerberg Biohub, San Francisco, CA 94518, USA
3Department of Pharmacology, University of Alberta, Edmonton, AB T6G 2E1, Canada
4Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
5Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
6Stanford Diabetes Research Center, Stanford University, Stanford, CA 94305, USA
7Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
8These authors contributed equally
9Senior author
10Lead Contact

*Correspondence: seungkim@stanford.edu (S.K.K.), steve@quake-lab.org (S.R.Q.), pmacdonald@ualberta.ca (P.E.M.)


https://doi.org/10.1016/j.cmet.2020.04.005

SUMMARY

Impaired function of pancreatic islet cells is a major cause of metabolic dysregulation and disease in hu-
mans. Despite this, it remains challenging to directly link physiological dysfunction in islet cells to precise
changes in gene expression. Here we show that single-cell RNA sequencing combined with electrophys-
iological measurements of exocytosis and channel activity (patch-seq) can be used to link endocrine
physiology and transcriptomes at the single-cell level. We collected 1,369 patch-seq cells from the
pancreata of 34 human donors with and without diabetes. An analysis of function and gene
expression networks identified a gene set associated with functional heterogeneity in b cells that can
be used to predict electrophysiology. We also report transcriptional programs underlying dysfunction
in type 2 diabetes and extend this approach to cryopreserved cells from donors with type 1 diabetes,
generating a valuable resource for understanding islet cell heterogeneity in health and disease.

INTRODUCTION derstand the connection between dynamic physiological pro-


cesses such as exocytosis with the generally slower changes
Cells use exocytosis to secrete a wide variety of molecules, in the transcriptomes of cells, which reflect the cell’s gene
including proteins, hormones, and neurotransmitters. From islet expression and genome regulatory processes. Single-cell RNA
cells in the pancreas to follicle cells in the reproductive system, sequencing (scRNA-seq) allows us to profile the molecular
endocrine cells secrete hormones that regulate metabolism, identity of individual cells and detect changes in gene expression
growth, and development (Burgoyne and Morgan, 2003; that arise in pathological settings. This technique has pro-
Williams, 2016). Exocytosis can be monitored at the single-cell vided increasingly accurate cell-type classifications in diverse
level by using patch-clamp electrophysiology to measure organs based on their transcriptional fingerprint (La Manno
changes in membrane capacitance as vesicles fuse with the cell et al., 2016; Tabula Muris Consortium et al., 2018; Villani et al.,
membrane and release their content. It is of great interest to un- 2017). However, it remains challenging to directly attribute

Context and Significance

Understanding the molecular mechanisms behind the dysfunction of pancreatic islet cells during diabetes could lead to bet-
ter therapies. Recently, key cell types in the islets have been found to exist as different subpopulations; for example, not all b
cells (the source of insulin) are the same in a given pancreas. But how these subpopulations may be functionally different has
been unclear. Here Camunas-Soler, Dai et al. address this issue by combining scRNA-seq (as a measure of molecular
phenotype) with single-cell electrophysiological measurements (as a measure of functional phenotype) in islet cells from
individuals with or without diabetes. The authors report induction of genes suggestive of compensation mechanisms,
and identify transcriptomic patterns associated with dysfunction, in b cells during T1D and T2D.

Cell Metabolism 31, 1017–1031, May 5, 2020 ª 2020 Published by Elsevier Inc. 1017
ll
Resource

physiological properties in a cell to its measured transcriptomic RESULTS


phenotype, and to establish unambiguous links with cellular
dysfunction in disease (Stuart and Satija, 2019; Wang and Kaest- Patch-Seq in Endocrine Cells to Simultaneously Assess
ner, 2019). Physiology and the Transcriptome
The islets of Langerhans control circulating glucose levels in To achieve patch-clamp followed by scRNA-seq (patch-seq) in
the body through regulated exocytosis of the two principal glucor- individual human islet cells, we initially isolated cells from 28 do-
egulatory hormones, insulin and glucagon. Several single-cell ge- nors, including 18 with no diabetes (ND), 7 with T2D, and 3 addi-
nomics studies revealed significant variability across islet cell tional donors that were excluded from further analysis due to the
types, as well as the existence of transcriptionally distinct cellular reasons specified in Table S1. We established patch-seq as a
subpopulations (Baron et al., 2016; Enge et al., 2017; Muraro two-step process: (1) we performed electrophysiological mea-
et al., 2016; Segerstolpe et al., 2016). This heterogeneity has surements using whole-cell patch-clamp, and (2) within 5 min
been confirmed through identification of surface markers and from ‘‘break-in’’ we collected cellular content using a larger sec-
mass spectrometry signatures (Bader et al., 2016; van der Meulen ondary pipette filled with lysis buffer (Figure 1A; STAR Methods).
et al., 2017; Wang et al., 2016a). Prior seminal physiologic studies This allowed intracellular access for whole-cell recording and
established that islet cells, and especially insulin-producing b cells, exocytosis measurements and was followed by recovery of
have heterogeneous function (Pipeleers, 1992; Salomon and full-length transcriptomes using Smart-seq2 that were
Meda, 1986; Stefan et al., 1987), while impaired b cell function sequenced to an average depth of 1–2 million reads (Figures
and reduced exocytosis are hallmark features of early-stage type S1A–S1C; STAR Methods). A total of 1,021 cells (80%) passed
2 diabetes (T2D) (Alejandro et al., 2015; Ashcroft and Rorsman, quality control for both electrophysiology and sequencing and
2012; Kahn et al., 2006). However, the ability to link islet cell molec- were classified into major cell types based on the expression
ular heterogeneity directly to such physiologic properties remains of key marker genes in a tSNE projection (Figure 1B; STAR
limited (Bakhti et al., 2019; Wang and Kaestner, 2019). Moreover, Methods). We obtained representatives of all major islet cell
the connection between genes up- or downregulated in T2D (Se- types (a, b, d, and g cells) and non-islet types such as acinar cells
gerstolpe et al., 2016) to functional consequences in islets remains (Figures 1C and 1D). For each cell we measured parameters rep-
unclear, and major gaps persist in our mechanistic understanding resenting cell size, exocytosis, Na+ channel currents, and Ca2+
of T2D ‘‘risk’’ candidate genes identified by genome-wide associ- channel currents (Figures 1E and 1F). Thus, we obtained a broad
ation studies (Mahajan et al., 2018; Prasad and Groop, 2016; survey of electrophysiological activity of all major islet cell types
Tritschler et al., 2017). in both ND and T2D settings (Figures S1D–S1K). In addition to
To address such limitations, we combine here the use of expected cell-type differences in size measured by whole-cell
whole-cell patch-clamp measurements and scRNA-seq capacitance (Figures 1C and 1F), these data demonstrate sub-
(together referred to as patch-seq; Cadwell et al., 2016; Földy stantial variation in exocytosis and channel activity in different
et al., 2016; Fuzik et al., 2016) in dispersed islet cells. This pancreatic cell types and disease states, including altered
approach enabled us to simultaneously measure the cellular exocytotic function in both a and b cells in T2D (Figures
function and transcriptomes of human endocrine cells, allowing S1D–S1K).
us to link gene expression and quantitative physiologic measure- To rigorously assess the robustness of patch-seq, we also
ments of vesicle exocytosis and ion-channel activity. We collected an additional 3,518 cells by fluorescence-activated
collected 1,021 patch-seq cells from 28 donors with no diabetes cell sorting (FACS) for scRNA-seq without patch clamping
(ND) or with T2D, most with short disease duration, and from 14 of these donors (8 ND, 6 T2D; Figure S1A; STAR
dissected function-to-gene expression relationships across islet Methods). The transcriptomes of cells after patch-seq or
cell types. We discovered a subset of 484 genes that predicts FACS purification from the same donors led to comparable
multiple b cell electrophysiological phenotypes, and we vali- quality metrics (Figure S2A). While we observed a difference
dated putative novel regulators of b cell physiology through ge- in the number of genes with detectable transcripts, the values
netic loss-of-function experiments. We also identified pathways from cells after patch-seq or FACS were within the range of
associated with reduced exocytosis in b cells from donors with previously published datasets (Figure S2B; Segerstolpe et al.,
T2D, and found that these are distinct from those observed in 2016). Analysis of genes required to maintain fundamental
nondiabetic donors. In b cells from individuals with T2D we cellular functions (hereafter, ‘‘housekeeping’’), islet identity,
also identified an upregulation of genes linked to a high exocy- and immediate early genes (IEGs) showed that most differ-
tosis phenotype, compared to b cells from nondiabetic donors, ences are driven by (1) varying sensitivity to genes expressed
and propose a genetic mechanism underlying physiological at low levels (drop-out of genes with low expression), and (2)
dysfunction of b cells during T2D. For example, we show that varying expression of stress-response genes likely reflecting
the transcription factor ETV1 likely plays a major role in b cell steps like islet shipping and dispersion (Figure S2C). Gene
dysfunction in humans with T2D. Finally, we demonstrate that expression in patch-clamped cells showed a high number of
our approach can also be applied to cryopreserved human islet islet-specific transcripts and low IEG activation (Figure S2C)
samples. Using 348 cells from rare cryopreserved islets of do- like in prior scRNA-seq studies of human islets (Segerstolpe
nors with T1D, we obtained profiles from a, g, d, duct, and surviv- et al., 2016). A pseudo-bulk average analysis of our patch-
ing b cells. Our work maps islet cell function and its transcrip- seq and FACS dataset confirmed these results (Figures S2D
tome at the single-cell level, providing a valuable resource for and S2E). We further investigated the effects of cell culture
exploring islet physiological and genetic dysfunction in health by collecting a subset of patch-seq cells on the same day of
and disease. dispersion. We found an initial increase in IEG transcripts that

1018 Cell Metabolism 31, 1017–1031, May 5, 2020


ll
Resource

A B C 10
Cell Size (pF)
0
Patch-clamp Cell collection β-cells γ-cells 16

GCG
for scRNAseq α-cells δ-cells 8
acinar 0
16

INS
log2 (CPM)
8

tSNE2
0
Measurements: Lysis 16

SST
Cell Size buffer 8
Exocytosis 0
Ca2+ Currents 16

PPY
Na+ Currents
8
secreted 0
endocrine cell

PRSS1
granules 16
tSNE1 8
0
cells
D E
α-cells, 689
0 mV 1s 30 mV
-70 mV -20 mV 50 ms
1.0 -70 mV 500 ms

Na+ Current (I/Imax)


-120 mV
ND acinar, 18 b Membrane 0.8

T2D d 500 ms
γ-cells, 21
capacitance 0.6 h
other a
δ-cells, 24 0.4
g
0.2
e
c 0.0
-150 -100 -50 0
f
Current Voltage (mV)

β-cells, 268

F
Cell size Exocytosis Na+ currents Ca2+ currents
e
Relative Frequency (%)

Relative Frequency (%)

Relative Frequency (%)

Relative Frequency (%)

Relative Frequency (%)


50 30
Relative Frequency (%)

Relative Frequency (%)


30 30 25 20 f 25
a d g h
α-cells 25 40
20 20
β-cells 15
20 20 20
15 30 15
15 10
10 20 10
10 10 10
5 10
5 5 5

0 0 0 0 0 0 0
0 2 4 6 8 10 12 0 10 20 30 40 -20 0 20 40 60 80 10 -80 -60 -40 -20 0 10 20 30 40 0 5 10 15 -2 0 2 4 6
Capacitance(pF) Early Exocytosis (fF/pF) Total Exocytosis (fF/pF) Na+ Current Half- Peak Na+ Current (pA/pF) Early Ca2+ Current (pA/pF) Late Ca2+ Current (pA/pF)
Inactivation (mV)

Figure 1. Patch-Seq Measurements in Endocrine Cells


(A) Schematic of patch-seq.
(B) tSNE projection of patch-seq cells clustered by expression of over-dispersed genes.
(C) Cell size (as membrane capacitance) for each cell and expression of key marker genes. Color code as in (B). Dashed line indicates average value per cell type.
(D) Patch-seq cells collected in this study.
(E) In each cell we measured the following: a, early exocytosis; b, late exocytosis; c, Ca2+ integral; d, total exocytosis; e, Na+ current half-inactivation; f, peak Na+
current; g, early Ca2+ current; h, late Ca2+ current (not shown: cell size, exocytosis normalized to Ca2+, reversal potential, and Na+ and Ca2+ conductance).
(F) Distribution of selected parameters demonstrating functional heterogeneity of a (red) and b (black) cells. Inset letters (a–h) correspond to parameters in (E).
Distribution of exocytotic responses in a and d at 1 mM glucose (a cells) or 5–10 mM glucose (b cells).
See also Figures S1 and S2 and Table S1.

decreased after 1 day in culture, while transcripts encoding is representative of the total secretory capacity of a b cell and a
islet-specific genes recovered (Figures S2F and S2G). This key indicator of b cell function and dysfunction in T2D (Fer-
recovery was also observed for cells obtained from donors daoussi et al., 2015; Gembal et al., 1992). In this way, we found
with T2D (Figure S2H). Finally, patch-seq itself did not genes positively or negatively associated with b cell exocytotic
appear to impact gene expression since cells collected with capacity (Figure 2B). Among top correlates, we found several
or without patch-clamp overlapped in a tSNE representation genes linked to pathways thought to regulate insulin secretion,
(Figure S2I). including b cell transcription factors (MAFA and ETV1), mole-
cules associated with insulin granules (SLC30A8, VAMP2,
Patch-Seq Identifies Novel Genetic Regulators of SCG2, and INS), metabolic enzymes (PDK4, PDHA1, and
Human b Cell Physiology GYG1), and ion channels (ABCC9, KCNH2, KCNMB2, and
We applied the patch-seq approach to find genes associated NALCN) including the L-type Ca2+ channel encoded by
with electrical and exocytotic function in b cells. In these cells, CACNA1C (Rorsman and Braun, 2013; Lu et al., 2002; Ait-Lounis
we found that electrophysiological properties cluster by ‘‘func- et al., 2010; Zhang et al., 2005; Chimienti et al., 2004). Gene set
tional group’’ (exocytosis, Ca2+, and Na+ currents) and are enrichment analysis (GSEA) using correlation scores confirmed
uncorrelated with other parameters such as donor age, sex, or many of these pathways and revealed additional enriched
BMI (Figures 2A and S3A). We first correlated the transcriptome categories, such as neuronal regulators, transcription fac-
of b cells to total exocytosis at stimulatory glucose levels, which tors, and regulators of cell polarity or stress (Figures 2C and

Cell Metabolism 31, 1017–1031, May 5, 2020 1019


ll
Resource

A B
Total Exocytosis
1.0 Cell Size
spearman r

0.8
Exocytosis VAMP2 4.0
0.6
Ca2+ channels

Total Exocytosis
0.4 MAFA 3.2
Na+ channels

(fF/pF, log)
0.2 SCG2 2.4
FAM159B 1.6
CACNA1C

Correlated
Cell size 0.8
INS
GPRASP1 0.0
0.71 0.58 0.52 0.38 Exocytosis Norm Ca2+
RGS9
Exocytosis

OGDHL 1.5
0.71 0.85 0.76 0.33 Early Exocytosis

Gene Expression
KCNH2
1.0
0.58 0.85 0.98 0.35 Total Exocytosis SLC16A9

(z-score)
0.5
SLC30A8
KCNMB2 0.0
0.52 0.76 0.98 0.34 Late Exocytosis
−0.5
-0.91 0.33 Peak Na+ Current −1.0
Na+

Anticorrelated
−1.5
-0.91 Na+ Conductance

0.33 0.35 0.34 -0.62 -0.59 -0.68 Late Ca2+ Conductance 36


RBP4
32
0.38 -0.62 0.73 0.72 Ca2+ Charge Entry PDHA1
Ca2+

BMI
GYG1 28
0.33 -0.59 0.73 0.77 Early Ca2+ current (pA/pF) 24
DLD
20
-0.68 0.72 0.77 Late Ca2+ current (pA/pF)
ID2
ME1
BMI
Donor
Sex

Other ion channel


glucose and related:
ATP1B1
C D KCNH2
KCNMB2
GYG1
glucose-6-phosphate glycogen ABCC9
NAGK NALCN
pyruvate Nucleus:
MAFA
ME1 ETV1
CELL ADHESION MOLECULES PC PDHA1
PDK4 NKX6.1
DLD CACNA1C
REGULATION OF BETA CELL DEVELOPMENT malate TCA
SLC25A6
ATP CACNA2D2
ADP
ION CHANNEL TRANSPORT OGDHL

NOTCH SIGNALING PATHWAY Ca2+

REGULATION OF GENE EXPRESSION IN BETA CELLS Granule


proteins:
ION TRANSPORT BY P TYPE ATPASES GPCR signalling: INS
TYPE II DIABETES MELLITUS GPRASP1 SCG2 Vesicular Membrane fusion:
RGS9 membrane: AP3B1
ALDOSTERONE REGULATED SODIUM REABSORPTION SLC30A8
SSTR5-AS1 SNAP25
NEURONAL SYSTEM PRKACB SYN1 VAMP2
PROTEASOME PRKAR1A NAPA
STX3
GLYCOLYSIS GLUCONEOGENESIS AP2A1
PATHOGENIC ESCHERICHIA COLI INFECTION
n.s.
SIGNALING BY WNT
****
MITOCHONDRIAL PROTEIN IMPORT E
DESTABILIZATION OF MRNA BY AUF1 HNRNP 100
Total Exocytosis (fF/pF)

APOPTOSIS
IL1 SIGNALING 75
RESPONSE ELEVATED PLATELET CYTOSOLIC CA2+
-2 -1 0 1 2 50

Enrichment (NES)
25

0
l

9b

S9

1
L
tro

YG
H

15

G
PA
D
on

G
G

m
C

TS

si

si
O

Fa
si

si

si

Figure 2. Correlation of b Cell Exocytosis to Single-Cell Gene Expression and Pathway Analysis
(A) Spearman correlation of electrophysiological parameters shows clustering of each functional group and low cross-correlation across clusters. All parameters
are normalized to cell size.
(B) Heatmap of top genes correlated and anticorrelated to total exocytosis in b cells from ND at 5–10 mM glucose. Cells are sorted by exocytotic response from
highest (left, dark green) to lowest (right, yellow). Gene expression is shown as a Z score after smoothing (n = 20 cells). Metadata for each cell are shown at bottom
(BMI, donor, sex).
(C) Top enriched pathways in genes correlated (green) and anticorrelated (red) to total exocytosis (KEGG and Reactome databases, false discovery rate
[FDR] < 0.1).
(D) Summarized map of cellular location and pathways with genes correlated (green) and anticorrelated (red) to exocytosis.
(E) Exocytosis in b cells measured at 10 mM glucose following target gene knockdown compared to control cells from same donors (348 cells, n S 3 donors per
knockdown experiment).
**p < 0.01, ****p < 0.0001 (Mann-Whitney U test and Benjamini-Hochberg [BH] correction). See also Figure S3 and Tables S2, S3, and S4.

S3B). Islet transcription factors with weaker but significant asso- permitting visualization and analysis of gene expression signifi-
ciation to exocytosis included PAX6, FOXO1, and NKX6-1 cantly correlated with exocytotic function in b cells.
(Figure S3B). This analysis nominated multiple genes—without previously
The majority of genes whose expression correlated known roles in b cells—as candidate regulators of physiological
significantly with b cell exocytosis are islet specific and included function, including OGDHL, FAM159B, TSPAN1, RGS9, and
candidate T2D risk genes like YWHAG (Fernández-Tajes et al., GYG1. Prior studies have linked OGDHL to metabolism, while
2019). Top gene correlates to exocytosis also included regula- FAM159B and TSPAN1 encode membrane proteins and RGS9
tors of oxidation and detoxification that could mitigate T2D- specifies a regulator of G-protein signaling (Bunik et al., 2008;
associated stress (Otter and Lammert, 2016), such as Danielsson et al., 2014; Uhlén et al., 2015). To test our predic-
glutathione peroxidase and transferases (GPX3, GSTK1, and tions, we performed small interfering RNA (siRNA) knockdown
GSTA4; Table S2). We integrated these results into a map that followed by patch-clamp for this set of genes in islets from an
combined correlations of transcript levels with four exocytosis additional 8 ND donors (Table S3; Figure 2E). Knockdown of
measures (early, total, late, and normalized to Ca2+; Figure 2D), each gene, confirmed by qPCR (Table S4), was followed by

1020 Cell Metabolism 31, 1017–1031, May 5, 2020


ll
Resource

A Exocytosis Ca2+ Na+ % expressed


positively
correlated
negatively
correlated
B
ABCC9
ACIN1
AP3B1 Exocytosis Ca2+ Na+ % expressed
Exocytosis BMP5 RBP4 1

Norm. ephys (log) Norm. expression


ATP1B1 ADK
BIRC2 ADSL
BMP5
C21orf91
ANP32B
ATP5B
ρBMP5=0.28(8)
C9orf142
CACNA1C
CHCHD7 ρRBP4=-0.26(8)
COPS3
CAMK2B COQ6 ρSYN1=0.17(8)
CCDC57 CTSB
CFC1 CYC1 ρMAFA=0.24(8) 0
CHGA DHPS
CLSTN1 DLD
CMC1 DNAJC8 Peak Na+ current SYN1 MAFA
COTL1
DMKN
DNTTIP2
EIF4EBP1 1
FAM159B
F3 GET4
GSTO1
ρBMP5=0.14(9)
GALK2 GYG1 ρRBP4=-0.44(7)

tsne 2
GPRASP1 H1F0
GPRIN1 HGS ρSYN1=0.28(8)
GPX3 ID2
GRIA4 ILF3-AS1 ρMAFA=0.17(8)
GSTK1 IMP3 0
IFI6 JTB 100 tsne 1
KCNH2 LMBRD2
KCNMB2 MAGEH1
LRCH4 80
MRFAP1L1

% cells expressed
MAFA MTHFD2
MDK MTND4P12 60
MRAP2 NDUFA9
MT-ND1
NAXD
NOL7
PCGF1 40
C
NEO1 PFN1
NPTX2 PNPO
OGDHL 20
POLR2G
PAXBP1 PSMB7
PDK4 RBP4 0
PIGX RSF1
PLEKHF2 RUVBL2
RAI1 SARS
SCG2 SCAND1
SEPT3 SEC13
SIN3A SIGMAR1
SLC16A9 SLC25A6
SLC30A8 SLC3A2
SNX27 SLC9A3R1
STAG2 SNF8
STAT3 SNRNP40
SYN1 SNRPB
TPPP SRXN1
TRIP12 STARD3NL
TSPAN2 STK25
VWDE TBCC
WDFY2 TCEB2
WDR59 UGDH
La Exo Size
Ex Ea Exo osis

Ca 2osis xocy is
Ea ha rm C is
rly rge a 2+
y
C nt

Na + Na + tan t
Co Cu e
uc nt
ce

La Exo Size
Ex Ea Exoc osis

Ca 2osis xocy is
Ea ha rm C is
rly rge a 2+

C ry
C nt

Na + Na + tan t
Co Cu e
uc nt
ce
uc n
E ytos
tos

La La a 2+ Entr

uc n
c

E ytos
tos

c
e
Pe ond urre

nd rre
tan

t
e
Pe ond urre

nd rre
tan
Ca 2 En
Ca 2 Ca 2 urr
t

Ca 2 Ca 2 urr
t
cy
tal Cell

cy
tal Cell
c

C
+
C o

+
C o
n

n
te
oc rly

te
oc rly
C

C
ak

ak
te te
To

te te
To

+
yt

yt

La La

D E
Total Exocytosis Late Ca2+ Conductance Peak Na+ Current Cell Size
= 0.59, P = 2e-10 100 30
= 0.54, P = 8e-09 = 0.51, P = 9e-08 0.8
= 0.46, P = 0.02 = 0.41, P = 0.02 = 0.64, P = 0.0001 Na+ Total
* *
Predicted k-NN

0.6
0 6
Predicted k-NN

Predicted k-NN

80 Conductance Exocytosis Cell Size Exocytosis


20 20 Ca2+ genes Na+ genes
0.4
4 genes genes
60
Peak Na +
0
0.2 Early
10 40 10 Current * 0 Exocytosis
20
0 0
0 Late Ca2+ * * Late
Conductance Exocytosis
0 20 40 0 50 100 0 20 40 Early Ca 2+
Measured patch-clamp Measured patch-clamp Measured patch-clamp Predictive Set genes
Ca2+ Current Charge Entry
random genes
(95% CI)

Figure 3. Gene Networks Associated with b Cell Functional Heterogeneity


(A) Genes with significant correlation to electrophysiology (Z score > 2 for n > 5 parameters; STAR Methods). Significant positive and negative correlations are
indicated in red and blue, respectively.
(B) tSNE projection of b cells from ND using genes in (A). Spearman rank correlation between each parameter and gene is shown as mean and error in last digit
(STAR Methods).
(C) Network of genes connecting different functional groups (‘‘PS genes’’). Genes (small dots) connecting different functional groups (large dots) are selected if
they show significant correlations (Z score > 2) to at least one functional parameter in each group. Edge color indicates positive/negative correlations (red/blue),
and gene color identifies clusters connected to the same functional groups.
(D) Predictions of the k-NN model using ‘‘PS genes’’ for training (black) and validation (red) sets. Spearman correlation and p for each parameter are indicated
as inset.
(E) Performance of the model using the ‘‘PS genes’’ (dark gray), measured as Spearman correlation between experimental and predicted values. Comparison to
model using 484 random genes of equivalent expression in b cells (10,000 permutations, 95% CI interval in light gray). Asterisk indicates parameters with p < 0.05
in validation set. Smaller spider plots show performance of the ‘‘PS gene set’’ (gray) versus top correlated genes to exocytosis (pink), Ca2+ (green), Na+ (orange),
and cell size (blue).
See also Figure S4 and Table S5.

electrophysiological characterization at 10 mM glucose and in- stimulated at these high-glucose conditions. Together, these
sulin immunostaining to confirm cell type. In 4/4 cases of genes data show that patch-seq analysis robustly identified previously
positively correlated to exocytosis (OGDHL, FAM159B, unknown regulators of human b cell physiology.
TSPAN1, and RGS9), we observed significant reduction of the
exocytotic responses after knockdown (Figure 2E). By contrast, Prediction of b Cell Electrophysiology from Gene
knockdown of GYG1, whose transcript levels are anticorrelated Expression
with exocytosis, did not lead to significant changes of exocy- To identify and investigate additional genes linked by patch-seq
tosis, possibly reflecting that b cell activity is already maximally to b cell excitability, we broadened our analysis to include

Cell Metabolism 31, 1017–1031, May 5, 2020 1021


ll
Resource

additional electrophysiological parameters (Ca2+ currents and dented algorithms that reliably link gene expression to b cell
Na+ currents) and selected transcripts showing consistent posi- function.
tive or negative correlations to multiple parameters as likely reg-
ulators of b cell physiology in nondiabetic states (Figure 3A). This Markers of b Cell Heterogeneity Correlate to b Cell
analysis identified several key genes, including genes previously Excitability
associated with b cell transcriptional heterogeneity (Baron et al., Within the identified ‘‘PS gene set’’ we found that transcripts en-
2016; Dorrell et al., 2016; Rui et al., 2017; Segerstolpe et al., coding RBP4 are significantly correlated with b cell functional het-
2016). For instance, transcripts encoding retinol binding protein erogeneity. Prior studies have noted heterogeneous expression of
4 (RBP4) correlated negatively with cell size, Na+ currents, and RBP4 in b cell subsets, but did not establish direct links to func-
total exocytosis, while transcripts encoding the b cell surface tional heterogeneity (Baron et al., 2016; Dorrell et al., 2016; Rui
protein FAM159B correlated positively to exocytosis, Ca2+ entry, et al., 2017; Segerstolpe et al., 2016). Other studies show that
and Na+ currents. A tSNE projection using these highly corre- RBP4 is an adipokine with roles in homeostatic regulation of meta-
lated genes shows a gradient of functional measures across b bolism (Broch et al., 2007; Tritschler et al., 2017). We observe that
cells that overlaps with patterns of gene expression (Figure 3B). RBP4 transcript levels have significant correlation with multiple b
To understand further how genetic pathways are connected to cell functional phenotypes; for example, it is the ‘‘PS gene’’ with
each major group of physiological function (exocytosis, Ca2+ strongest anti-correlation to b cell Na+ channel activity. RBP4+ b
currents, and Na+ currents), we repeated GSEA on their aver- cells have decreased exocytosis and Na+ currents despite having
aged correlation scores (Figure S3C). Results for overall exocy- normal Ca2+ current activity (Figure 4A). Consistent with this, we
tosis are consistent with those reported above (Figure 2C); by observed that RBP4+ b cells also have significantly reduced
contrast, GSEA further identified that Na+ and Ca2+ currents expression of key regulators of stimulus-secretion coupling, like
are linked to pathways related to increased excitability and circa- KCNJ8, ABCC9, and SCN3A (Figure 4B). In rodent b cells,
dian rhythms, respectively (Perelis et al., 2015). Several of the Scn3a encodes the principal physiologically relevant Na+ channel
observed pathways identified by GSEA on averaged correlation (Zhang et al., 2003). Together, these results provide evidence that
scores also overlap with those recently implicated in islet RBP4 expression is a marker of b cells with reduced function and
dysfunction in T2D using genomic data (Fernández-Tajes show that previously identified markers of b cell transcriptomic
et al., 2019). heterogeneity identify subpopulations with heterogeneous func-
We then asked whether genes that correlate to multiple groups tion (Dorrell et al., 2016; Johnston et al., 2016).
of physiological function (‘‘functional groups’’) could be used to
develop predictive algorithms that link transcriptional signatures In T2D, b Cells Induce Genes Linked to Increased and
to b cell function. To do so, we generated a network of genes with Decreased Exocytosis
significant correlation to more than one functional group (e.g., Islets from the 7 donors with T2D (Table S1) showed reduced in-
Ca2+ and exocytosis) and selected those with highest median sulin content and secretion (Figure 5A), and impaired b cell
expression (Figure 3C; Table S5). This list of highly connected exocytosis (Figures 5B and S5A). All b cells were obtained from
genes, which we termed our predictive set (PS), contains genes donors with recent T2D onset (<9 years), and who were not
(1) previously linked to b cell excitability, (2) with heterogenous receiving insulin treatment (as we did not record any b cells for
expression in b cell subpopulations and in T2D (FFAR4, R241, the only donor on insulin treatment; STAR Methods). To
FXYD2, and ID4), and (3) with unknown function in b cells (Table identify genetic drivers of b cell dysfunction in T2D, we first as-
S5). We then attempted to predict the measured electrophysi- sessed transcripts that we previously identified to be significantly
ology of each cell from the average values of its nearest neigh- correlated to exocytosis in b cells from ND (Figure 2B; Table S2).
bors (NN) in PS gene expression (n = 484 genes, k-NN with 5 Unexpectedly, we found that genes that positively correlate with
neighbors) (STAR Methods). We obtained significant correla- exocytosis in b cells from ND are upregulated in T2D, while genes
tions between the experimentally measured patch-clamp pa- that negatively correlate with exocytosis in b cells from ND had
rameters and the predictions from the k-NN model (Figures 3D reduced expression in T2D (Figure 5C). We found that this tran-
and S4A, black). We compared the performance of PS genes scriptomic shift is also observed in b cells from obese ND individ-
to randomly selected genes of equivalent expression (n = uals (Figure S5B). These data suggest that b cells attempt to alter
10,000 permutations) and found that the PS genes performed transcript levels to increase functionality, perhaps in response to
significantly better for all parameters (Figure 3E). We also tested increased insulin demand associated with T2D and obesity. But
our model by comparing its predictive power to that obtained us- in T2D this ultimately fails to maintain an increase of b cell func-
ing the genes most correlated to each functional group. We tion, since their exocytotic response is significantly impaired
found that PS genes showed consistent performance across compared to ND controls (Figure 5B).
all measured parameters, whereas group-specific gene sets To find genes that could underlie this effect, we performed
only performed well for their subset of parameters (Figures 3E correlation analysis in cells from donors with T2D and integrated
and S4B). Finally, we validated the predictive power of PS genes the ND and T2D results in a correlation map of exocytosis with
with a ‘‘test’’ set of data including gene expression and functional the overall changes in gene expression (Figure 5D; Table S2).
parameters of cells withheld from the analysis that generated the Next, we performed a pathway analysis for each of four gene
PS gene set. This analysis demonstrated appropriate recovery of subsets (ND/T2D and correlated/anticorrelated to exocytosis)
significant correlations for 5 of the measured functional parame- (Figures 5E and S5C). Our data show that the pathways associ-
ters (Figures 3D, S4A, red, and 3E). Thus, patch-seq and network ated with reduced exocytosis in T2D are distinct from those
analysis combined with machine learning generated unprece- associated with low exocytosis in ND. While we found that low

1022 Cell Metabolism 31, 1017–1031, May 5, 2020


ll
Resource

A
Na+ currents Ca2+ currents Exocytosis
*** * * * *
17.5

Late Ca2+ Conductance (pS/pF)


60 60
3.0

Ca2+ Charge Entry (pC/pF)

Early Ca2+ Current (pA/pF)


1.50 8 120
Peak Na+ Current (pA/pF)

Na+ Conductance (pS/pF)

Late Ca2+ Current (pA/pF)


1000 40

Early Exocytosis (fF/pF)


Total Exocytosis (fF/pF)
15.0

Late Exocytosis (fF/pF)


50 50
1.25 2.5 100
800 6 12.5
40 40 30
1.00 2.0 80 10.0
30 600 1.5 30
0.75 4 60 20 7.5
20 400 0.50 1.0 40 20 5.0
2 10
10 200 0.25 0.5 20 10 2.5
0 0.00 0 0.0 0 0 0 0.0
0
RBP4+
B Na+ channels ATP-sensitive K+ channel Ca2+ channels RBP4-
log2(CPM+1)

** ** ***
10
5
0
100
75
% cells

50

25

0
KCNJ11
SCN1B

SCN2A

SCN3A

SCNM1

SCN3B

SCN9A

KCNJ8

ABCC8

ABCC9

CACNA1A

CACNA1C

CACNA1D

CACNA1H

CACNA2D1

CACNA2D2

CACNB2
Figure 4. Functional and Transcriptomic Differences in b Cell Subpopulations
(A) Electrophysiological function in RBP4 subpopulations of b cells. Significantly lower Na+ currents and exocytosis is observed for RBP4+ cells.
(B) Gene expression values (top) and % b cells with detectable expression (bottom) of Na+, ATP-sensitive K+ channels, and Ca2+ channels.
***p < 0.001, **p < 0.01, *p < 0.05 (Mann-Whitney U test with BH correction).

exocytosis in b cells from ND is linked to metabolic pathways and STAT3. These results indicate that optimal insulin secretion
glucose metabolism, the dysfunction in T2D is related to immune may require balanced expression of ETV1 and its downstream
response, cell-cycle pathways, and altered transcription factor targets, and that an imbalance may lead to dysregulated secre-
expression. For instance, we observe induction of pathways tion during early T2D (Figure 5G). To test the differential role of
like NFkB signaling, cell-cycle checkpoints, and auto-degrada- ETV1 in b cell physiology, we performed siRNA knockdown of
tion of the ubiquitin ligase COP1. Genes in T2D that show ETV1 followed by patch-clamp in an additional set of human is-
increased expression associated with reduced b cell exocytosis lets. We found that knockdown of ETV1 rescued exocytosis in b
included known regulators of immune and inflammatory path- cells from donors with T2D, whereas it did not alter exocytosis in
ways like NFKBIA, IL6R, and IRAK1, and the transcription factors b cells from ND (Figures 5H and S5G), further supporting a signif-
ETV1 and STAT3 (Figures 5D–5F and S5D). In particular, we icant role for ETV1 in b cell dysfunction in T2D.
found ETV1 to be significantly enriched in b cells from donors
with T2D, and to show ‘‘reversed’’ correlations to exocytosis in Transcriptional and Physiological Heterogeneity
T2D and ND. Overall, we found that in T2D, increased ETV1 Observed in a Cells
mRNA levels were associated with reduced b cell exocytosis Cell size and Na+ channel properties are often used to identify a
(Figure 5D, yellow box). These results were consistent across and b cells in rodents (Briant et al., 2017; Zhang et al., 2014);
several measured exocytosis parameters (Figures S5E and however, this is not generally applicable to humans, making it
S5F). Hence, our results suggest that dysfunctional cells in difficult to distinguish islet cell types at the time of patch-clamp-
T2D express higher levels of ETV1 than functional ones, whereas ing. For example, a cells (definitively identified post hoc from
the opposite happens in ND. scRNA-seq) were frequently mis-classified as possible b cells
The ETV transcription factors impair insulin secretion in hyper- during our initial patch-clamp measures of capacitance that indi-
glycemic mouse models and are negatively regulated through cated cell size (>4 pF; Figure 1F). To improve cell-type identifica-
COP1-dependent targeted degradation (Figure 5G) (Suriben tion prior to scRNA-seq, we implemented a machine learning
et al., 2015). We tested genes previously reported to show algorithm based in ‘‘random forests’’ (Breiman, 2001) to classify
ETV-dependent expression in mouse islets (Suriben et al., cell types from their electrophysiological profile. We trained and
2015) and found several human orthologs with correlating validated the model in cells from ND donors using 10-fold cross-
expression in single b cells, including MAFA, SLC30A8, and validation, obtaining an AUC of 0.95 in the validation dataset

Cell Metabolism 31, 1017–1031, May 5, 2020 1023


ll
Resource

A *
B ** ** n.s. C
4000 * 2.5 ND 40 40 10
***

Early Exocytosis (fF/pF)


Total Exocytosis (fF/pF)
35 35

Late Exocytosis (fF/pF)


Insulin Content (ng/ml)

T2D
2.0 8
3000 Insulin Secretion 30 30
(% content) genes anticorrelated
25 25 6
1.5 to exocytosis in ND
2000 20 20
genes correlated
1.0 15 15 4 to exocytosis in ND

1000 10 10
0.5 2 −0.4 −0.2 0.0 0.2
5 5 median logFC (T2D vs ND)
0 0.0 0 0 0
ND T2D 1 mM 10 mM ND T2D ND T2D ND T2D
Glucose

Increased β-cell exocytosis in ND


D E
Pathways
log2FC (T2D vs ND)

1.5 Relative diff. in GPD2


% cells expressing
gene (normalized):
ND high exocytosis genes (top level) i ii iii iv
0.0
0.0 5 SNARE interactions in vesicular transport
0.4 DDX46 CTSF PDK4 RGS9
Regulation of beta-cell development
0.25

-log10 (FDR )
−1.5 0.5 4 Type II diabetes mellitus
FXYD6 Cell adhesion molecules (CAMs)
STXBP5-AS1 MRAP2
3
Glycosaminoglycan biosynthesis
SCG5
2 and heparan sulfate
0.2 1 Pyruvate metabolism

Decreased β-cell exocytosis in T2D


RBP4 TSPAN2
Correlation to Exocytosis (T2D)

CACNA1C
TCF7L2
TSPAN1 0
MAFA

KCNH2
Vesicle-mediated
SLC30A8
transport Metabolism of carbohydrates
0.0 Developmental Biology
Metabolism Glycolysis
PDHA1 Disease
TNS2 Neuronal System
ENO1 GUCY1A3
ID2 Signaling molecules
−0.2 and interaction Autodegradation of the E3
ADAM10 ITGB1
VAMP2
Transcription / ubiquitin ligase COP1
CTNNAL1
IL6R TXNDC11
DNA replication Cell Cycle Checkpoints
Immune System
Cell Cycle Activation of NF-κβ in B Cells
−0.4 STAT3
Transport of small
T2D low IRAK1
ETV1
exocytosis genes AP1S2
IFNAR1 genes with molecules ND T2D ND T2D
reversed correlation Hemostasis
PRELID1 NFKBIA high low
exocytosis exocytosis
−0.6
−0.4 −0.3 −0.2 −0.1 0.0 0.1 0.2 0.3 0.4
Correlation to Exocytosis (ND)
H 100
Total Exocytosis (fF/pF)

F ETV1 G Increased
75
insulin demand
**
1
optimal
range
***
10
log2 FC (T2D-ND)

Function

dysfunction dysfunction 50
log2(CPM+1)

ETV1 STAT3
5 0 ***
ETV1 / 25
STAT3 / COP1 inflammation /
0 −1 immune genes β-cell
dysregulation 0
STAT3

ND T2D
RBP4
ETV1

IL6R

Expression
ctrl siETV1 ctrl siETV1

ND T2D

Figure 5. Functional and Transcriptomic Changes in b Cells Early in T2D


(A) Insulin content and secretion (as % content) for donors included in this study. *p < 0.05, ***p < 0.001 (Student’s t test; two-way ANOVA and Tukey post test).
(B) Measured exocytosis for b cells in ND and T2D. **p < 0.01 (Mann-Whitney U test and BH correction).
(C) Enrichment analyis in T2D of genes found to be correlated (red) or anticorrelated (blue) to exocytosis in cells from ND. Data show median log fold change in
gene expression between T2D and ND for each subset of genes. Error is SEM. Central bar shows range of log2 fold change obtained by sampling random genes of
equivalent expression (10,000 iterations, 400 genes, 5%–95% range).
(D) Gene correlation map of exocytosis. Scatterplot shows correlation to exocytosis in ND (x axis) and T2D (y axis) for each gene. Genes with significant cor-
relations (Z score > 2) are colored according to their fold enrichment in T2D cells (red, upregulated in T2D; blue, downregulated in T2D), and size is proportional to
relative change in % expression. Regions of interest are highlighted with dotted boxes. Genes with non-significant correlations are shown in gray.
(E) Enriched pathways for genes correlated to exocytosis in ND (1) and T2D (2), and genes anticorrelated to exocytosis in ND (3) and T2D (4). Enrichment is shown
as log10(FDR). Left bar indicates top category of each pathway.
(F) Distribution of ETV1 expression (left) and enrichment of a subset of genes that change between ND and T2D.
(G) Model showing the hypothesized role of ETV1, STAT3, and immune pathways in b cell dysfunction in early T2D. Based on Suriben et al. (2015).
(H) Exocytosis in b cells measured at 5 mM glucose following ETV1 knockdown compared to control cells from same donors (n = 3 donors both for ND and T2D,
***p < 0.001 Kruskal-Wallis test and Dunn’s post test).
See also Figure S5.

(Figure 6A). This model correctly identified 85% (92%) of a cells in cells from T2D donors (AUC = 0.93) (Figure 6A). Compared
(b cells), misidentifying 15% of a cells and 8% of b cells (Fig- with a capacitance-based cell size cut-off at the time of patch-
ure 6B). Features from every functional group contributed to clamping, our model significantly improved the a priori identifica-
the classifier (Figure 6C), and the model also performed robustly tion of a cells and reduced their mis-classification as b cells

1024 Cell Metabolism 31, 1017–1031, May 5, 2020


ll
Resource

A B D

0.8

0.6

0.4

0.2
1.0 Original prediction at

α-cells
0.85 0.15 the time of patch-clamp
True Positive Rate

True
0.8

α-cells
β-cells
0.08 0.92
0.6
α-cells β-cells
ND α-cells, validation Predicted
0.4 ND β-cells, validation
Predicted by model
AUC = 0.95 Feature importance
C

α-cells
0.2 T2D α-cells 0 0.2 0.4
T2D β-cells Cell Size
AUC = 0.93 Peak Na+ Current
Early Exocytosis
0.0 Total Exocytosis
0.0 0.2 0.4 0.6 0.8 1.0 Na+ Conductance correctly predicted
False Positive Rate Late Exocytosis misidentified as β-cells
Early Ca2+ Current
Late Ca2+ Current
remaining unclassified
Ca2+ Charge Entry

E
Capacitance (Cell Size) DDIT3 PPP1R15A SQSTM1 XBP1
ρPPP1R15A=-0.44(3)
ρLOXL4=0.40(3)
1

Norm. expression
ρFEV=0.37(4)
ρSCG2=0.57(3)
ρFFAR1=0.44(4)

Peak Na+ Current ARX LOXL4 FEV MAFB


ρPPP1R15A=-0.24(4) 0
ρLOXL4=0.32(4)
ρFEV=0.26(4)
1

Norm. ephys (log)


ρSCG2=0.33(4)
ρFFAR1=0.33(4)
Na+ Conductance SCG2 FAP FFAR1 GPR119
ρPPP1R15A=-0.22(4)
ρLOXL4=0.32(4)
ρFEV=0.26(4) 0
tSNE2

ρSCG2=0.30(4)
ρFFAR1=0.23(4)
tSNE1

Figure 6. Transcriptomic and Electrophysiological Heterogeneity in a Cells


(A) ROC curve of cell-type prediction using random forests for the validation dataset in ND (red, blue) and in T2D (green, purple).
(B) Confusion matrix showing accuracy of predictions in ND validation dataset.
(C) Contribution of each feature to the random forest model.
(D) Comparison of a cell identification from predictions at the time of patch-clamping (simple cell size cut-off) versus the model.
(E) tSNE plots showing heterogeneity in gene expression of a cells using over-dispersed genes and normalized electrophysiological measurements.
See also Figure S6.

(Figure 6D). Analysis of a cell transcriptomes revealed significant identification of patch-clamped human a cells, our studies pro-
transcriptional heterogeneity in islets from multiple human do- vide evidence for molecular heterogeneity associated with func-
nors, including those with T2D. In particular, a subset of cells tional heterogeneity in a cells.
showed enrichment of regulators of a cell maturation (LOXL4,
MAFB, and ARX), regulators of reduced ER stress (DDIT3, Application of Patch-Seq to Rare Cryopreserved
XBP1, and PPP1R15A), transcription factors governing endo- Samples: Islet Cells in T1D
crine fate (FEV and ISL1), receptors involved in glucose homeo- Tissue banking programs can provide unique access to cells from
stasis (FFAR1 and GPAR119), and secretory pathways (SCG2) subjects with specific disorders. Hence, we investigated whether
(Figures 6E, S6A, and S6B). This transcriptional heterogeneity the patch-seq approach could be extended to rare cryopre-
correlated to electrophysiological features including Na+ cur- served islet samples (Manning Fox et al., 2015). We found that
rents and cell size (Figure 6E); further work will be required to un- single-cell transcriptomes from cryo-banked islets had compara-
derstand the relationship of this heterogeneity to a cell dysfunc- ble quality metrics to those of fresh tissue (Figure S7A), allowing
tion in T2D. Thus, in addition to improving prospective us to investigate cells from three donors with T1D and three

Cell Metabolism 31, 1017–1031, May 5, 2020 1025


ll
Resource

A B

α-cells

β-cells

δ-cells
γ-cells
acinar

ductal
PSCs
β-cells γ-cells acinar T1D N total
α-cells δ-cells ductal ND PSCs 16
acinar
PSCs 133 215
1.0 12

0.8 α-cells

Fraction of cells

log2 (CPM+1)
8
0.6
tSNE2

0.4
β-cells 4
0.2
ductal
0.0
ND 1

T1D
γ-cells
tSNE1
δ-cells 0

CXCL8
MMP2
SPARC
DCN
NNMT
MSC
ANXA1
COL1A2
PLIN2
IL24
PRSS2
CTRB2
CTRB1
SPINK1
REG1A
BCAT1
PRSS1
MGST1
AKR1C3
CPA2
GCG
TTR
LOXL4
VIM
GC
RGS4
IGFBP2
HIGD1A
RSAD2
GPX3
INS
IAPP
MEG3
RBP4
HADH
NPTX2
ERO1B
IGF2
PDX1
GSN
TINAGL1
ANXA3
TFPI2
KRT19
CAV2
KRT7
BICC1
PMEPA1
TACSTD2
CRIM1
PPY
GPC5-AS1
ETV1
AQP3
GCNT3
CARD11
FXYD2
PXK

SST
THSD7A
SLITRK6
HHEX
RBP4
LEPR
FAM84B
EDN3
CD9
MS4A8
SLC38A1
HAP1
C β-cells

T1D D
ND 100% β-cells α-cells ND
% cells

75% 20 20
log2 (CPM+1)

log2 (CPM+1)
T1D
CHGA
CHGB

NKX2-2

NKX6-1
PDX1

MAFB
MAFA
GCG

ETV1
INS

50% 15 15
25% 10 10
5 5
0 0
α-cells
mean expression

0.8
0.7
F P4

-D 4

S
A

M K1

M 3
C 1
G 7
G 2
3
EM 163

FK B
LD IA

AD 6

PS A1
H H

3
S1 A

N A
LA R

1
C
4
PX

TM PX

G 00A

D
M
FA

N 76

G 45

P
(norm.)

T1D

B
C

D
0.6
B

IN

PD
H FA

AC
TM EM

D
U
R

SP
ND 0.5
0.4
0.3
CHGA
CHGB

NKX6-1
NKX2-2
MAFB
GCG

ARX
INS

FEV

ETV1

0.2
F ** *
3.5
5

Early Ca2+ Current (pA/pF)

Late Ca2+ Current (pA/pF)


T1D β-cells T1D α-cells 3.0
E 4
ADAPTIVE IMMUNE SYSTEM IMMUNE SYSTEM 2.5
TYPE I DIABETES MELLITUS ADAPTIVE IMMUNE SYSTEM
IMMUNE SYSTEM INTERFERON GAMMA SIGNALING 2.0
GRAFT VERSUS HOST DISEASE ER PHAGOSOME PATHWAY 3
ALLOGRAFT REJECTION ANTIGEN PROCESSING CROSS PRESENTATION 1.5
ENDOSOMAL VACUOLAR PATHWAY ENDOSOMAL VACUOLAR PATHWAY 2
AUTOIMMUNE THYROID DISEASE CLASS I MHC ANTIGEN PROCESSING PRESENTATION 1.0
NK CELL MEDIATED CYTOTOXICITY TCR SIGNALING
INTERFERON GAMMA SIGNALING APOPTOSIS 1 0.5
ER PHAGOSOME PATHWAY INTERFERON SIGNALING
0 1 2 3 4 0 1 2 3 0 0.0
-log10(FDR) -log10(FDR) ND T1D ND T1D

Figure 7. Patch-Seq in Cells from Cryopreserved T1D Islets


(A) Left: tSNE projection of patch-seq cells clustered by expression of over-dispersed genes. Right: cell types and total number of cells obtained for ND and T1D.
(B) Marker genes for each cell type.
(C) Expression of key identity genes on a and b cells from donors with T1D and ND matched controls.
(D) Representative genes obtained in a differential expression analysis between T1D and ND for b and a cells.
(E) Pathways enriched in upregulated genes in T1D a and b cells.
(F) Distribution of Ca2+ parameters showing statistically significant differences between a cells of donors with T1D and ND.
**p < 0.01, *p < 0.05 (Mann-Whitney U test with BH correction). See also Figure S7 and Tables S6 and S7.

controls matched for BMI, age, sex, and storage time (Table S6). script enrichment of genes related to immune activation and allo-
We performed patch-seq in 348 cells from these samples (Fig- graft rejection (HLA-DMA and FAS) (Figures 7D and 7E). In a cells
ure 7A; Table S6) and used a logistical regression model to iden- from donors with T1D, we found increased NKX6.1 and
tify marker genes for each population (Figures 7B and S7B). Sam- decreased NKX2.2 mRNA (Figures 7C and S7E) along with
ples from donors with T1D contained an unanticipated variety of decreased Ca2+ channel activity (Figures 7F and S7D), in general
cell types (Figure 7B), including an enrichment for pancreatic agreement with recent reports using bulk RNA-seq and immuno-
polypeptide-secreting g cells and ductal cells (Figure S7C). histochemistry (Brissova et al., 2018; Chakravarthy et al., 2017),
We obtained 11 b cells from two donors with T1D, consistent although we did not detect a consistent decrease in Ca2+ channel
with prior observations of b cell survival years after T1D onset gene expression as previously reported (Figure S7E). Our anal-
(Keenan et al., 2010; Morgan and Richardson, 2018) (Figure S7C). ysis of a cells in T1D also showed transcript enrichment of mucin
These b cells had similar electrophysiological profiles as cryopre- (MUC) and other genes typically associated with ductal cells, and
served b cells from ND controls (Figure S7D) and appeared to FEV1, a reported endocrine progenitor cell marker in mice (Fig-
maintain equivalent expression of hallmark b cell genes (INS, ures 7C and 7D; Table S7) (Byrnes et al., 2018; Liu et al., 2019).
PDX1, and MAFB) (Figure 7C). Differential expression analysis Together, our data demonstrate that patch-seq can be used in
showed decreased expression of RBP4 and FFAR4 and tran- rare cryo-stored tissue types, and supports the view that the

1026 Cell Metabolism 31, 1017–1031, May 5, 2020


ll
Resource

transcriptomic and functional signatures of surviving b cells may RGS9, and MRAP2 in retinal tissue; GPRIN1 in brain; and
be preserved in T1D, while a cells lose characteristic functional KCNH2 in cardiomyocytes) (Uhlén et al., 2015), indicating shared
and transcriptomic phenotypes, consistent with the observation mechanisms of signal transduction and showing that the analyt-
of impaired glucagon regulation in T1D. ical tools developed here could be applicable to patch-seq
studies in other excitable cells and endocrine tissues.
DISCUSSION Regulation of islet exocytosis by metabolism is complex (New-
gard, 2017). We observed that multiple expected metabolic
Success in identifying the genetic basis of islet function and pathways positively correlated with b cell functional measure-
dysfunction in diabetes has been limited by an inability to con- ments (e.g., glucose flux and metabolism, circadian rhythm),
nect islet cell physiologic function with transcriptome regulation. but also observed unexpected negative correlations of b cell
Elucidating mechanisms underlying diabetes also suffers from exocytosis to pathways like glycolysis and pyruvate metabolism.
limited human cell and tissue availability that hinders single- We integrated several of the observed metabolic correlations to
cell-based investigations, particularly when considering T1D. b cell exocytosis in a functional map (Figure 2D), providing sup-
Multi-modal single-cell technologies will help address these is- port for the hypothesis that glucose is preferentially used for TCA
sues by increasing the depth of available data and by making it anaplerosis through pyruvate carboxylase to facilitate insulin
possible to directly link transcriptomes to additional readouts secretion (Alves et al., 2015). These findings are also sustained
in individual cells (Macaulay et al., 2017; Stuart and Satija, by measures of CPT1A and SLC16A9 expression, which
2019). One integrated approach previously used in studies of ro- mediate the rate-limiting step of fatty acid oxidation and carnitine
dent brain slices is patch-seq (Cadwell et al., 2016; Földy et al., efflux, respectively (Aichler et al., 2017; Newgard, 2017). Overall,
2016; Fuzik et al., 2016). Like neurons, endocrine cell secretion is the observation of glucose homeostasis pathways across our
coupled to dynamic electrophysiological mechanisms that analysis also resonates with the importance of metabolites in
culminate in regulated exocytosis of secretory vesicles. Methods shifting transcriptional states of b cells, recently dubbed ‘‘meta-
that allow simultaneous measurements of transcriptomes and bolic memory’’ (Rosen et al., 2018; Sharma and Rando, 2017).
secretory capacity hold promise for understanding the mecha- Another useful outcome here is our development of a network
nisms by which gene expression enables physiological function analysis tool that links a PS of 484 genes with applied machine-
in these cells. learning techniques. We used this to predict b cell function solely
Here we describe the development of patch-seq for endocrine from RNA transcript abundance. Genetic regulators of b cell ac-
cell types and report paired physiologic-transcriptomic data for tivity, including many uncharacterized in islet biology, were iden-
more than 1,300 human islet cells. This depth of data allowed tified in the predictive set, and included antioxidant molecules
us to link transcriptional and physiological heterogeneity at (GPX3 and GSTK1), surface proteins (TSPAN1 and TSPAN2),
single-cell resolution, and to identify genes and pathways regu- and b cell enriched molecules (NPTX2 and FAM159B). To vali-
lating exocytosis of human islet b and a cells. With a patch-seq- date predictions from patch-seq correlations and this network
derived minimal PS of genes and machine-based learning tools, analysis, we used genetic loss-of-function approaches, which
we developed algorithms that correctly predicted hallmark phys- provided evidence that previously uncharacterized genes like
iological functions like exocytosis from gene expression, and OGDHL, RGS9, TSPAN1, and FAM159B are positive regulators
accurately classified live isolated islet cell types. For studies of of b cell exocytosis. Prior studies have shown that FAM159B
islets from T1D and T2D donors, we used patch-seq to identify may be regulated by the b cell transcription factor SIX3 (Arda
molecular mechanisms underlying b and a cell dysfunction in et al., 2016), and is a marker of b cell subsets (Dorrell et al.,
these diseases. Thus, our study provides a powerful resource 2016). Like FAM159B, other genes linked by our analysis to dif-
and toolset for simultaneous multiplex phenotyping of human ferential b cell function were also found to be expressed in b cell
islet cells, in health or disease. sub-populations (ID2, ID4, and RBP4) (Baron et al., 2016; Dorrell
Patch-seq analysis identified key genes and pathways regu- et al., 2016; Rui et al., 2017; Segerstolpe et al., 2016). For RBP4,
lating b cell exocytosis, including both known and previously un- we provide evidence that ND RBP4- b cells, compared to ND
characterized regulators and mechanisms controlling insulin RBP4+ b cells, have relatively superior electrophysiological func-
secretion such as cell adhesion molecules and neuroactive tion (also see discussion of T1D below). Overall, our data show
ligand receptors. Adhesion molecules have previously demon- that b cells exhibit significant transcriptomic and electrophysio-
strated roles in b cell attachment and motility (Dahl et al., 1996; logical heterogeneity, supporting the idea that b cells comprise a
Kaido et al., 2004), and are thought to govern b cell polarity spectrum of cellular states. Further studies are needed to
and function through control of directed insulin exocytosis into address whether this heterogeneity corresponds to dynamic or
the vascular axis (Geron et al., 2015; Parnaud et al., 2015; Gan static states and the existence of mechanisms allowing cells to
et al., 2018). Among neuronal regulators, we identified pairs of switch between them, or possibly to a hierarchical physiological
factors such as neurexin 1 (NRXN1) and neuroligin 1 (NLGN1), organization of b cells (Johnston et al., 2016). Similarly, further
which are known to bind in a heteromeric complex (Suckow application of the PS gene signature to FACS datasets in disease
et al., 2008). The identification of binding partners in our gene- settings (T2D) would enable high-throughput functional pheno-
function correlation analysis confirms the robustness of this typing of b cells.
approach to identify unanticipated candidate regulators of islet Understanding the basis of islet adaptation or dysfunction in
cell function. Similarly, examination of tissue specificity reveals diabetes is another important result described here. Our work
that several of the encoded proteins correlating to b cell exocy- shows that the pathways associated with reduced b cell exocy-
tosis are also enriched in other excitable cell types (GPRASP1, tosis in T2D significantly differ from those correlated to low

Cell Metabolism 31, 1017–1031, May 5, 2020 1027


ll
Resource

exocytosis in cells from ND. Furthermore, the observation that b comparable to that of control fresh islet cells. Moreover, our
cells from donors with T2D or obesity show increased expres- detection of a cell dysfunction and normal function of surviving
sion of genes that are correlated to a high exocytosis phenotype b cells using patch-seq with T1D islets correlates well with find-
in ND suggests molecular mechanisms for functional compensa- ings from a recent study using different methods (Brissova et al.,
tion by b cells in early-stage T2D. However, our analysis also re- 2018). It is important to note that b cells from donors with T1D
veals as-yet unexplained complexity in b cell responses; for studied here were from islets obtained >11 years (and up to 31
example, ETV1 and STAT3 mRNA are positively correlated years) following diagnosis. Thus, further studies are needed to
with ND b cell functional phenotypes, and also with b cell investigate potential b cell dysfunction that may occur at earlier
dysfunction in T2D. To address this, we performed genetic stages of disease progression. Patch-seq revealed greater a
loss-of-function experiments that showed recovery of exocytotic cell heterogeneity in T1D compared to ND islets, including evi-
capacity after knockdown of ETV1 in b cells from donors with dence of impaired maintenance of a cell fate, and we speculate
T2D. Recent studies suggest that b cell ETV1 and STAT3 protein that this might contribute to the well-recognized impairment of
degradation is regulated by the ubiquitin ligase complex (Dalla- glucagon secretion observed in T1D (Unger and Cherrington,
valle et al., 2016; Suriben et al., 2015). Our work suggests that 2012). Moreover, transcriptomic profiling here reveals that func-
ubiquitination and proteosomal degradation pathways in b cells, tional preservation in b cells from donors with T1D corresponds
including a pathway stimulating COP1 auto-degradation, are to undetectable expression of RBP4, consistent with work by
associated with dysfunction in T2D. Thus, in addition to tran- others (Brissova et al., 2018; Rui et al., 2017). By contrast, we
script-based mechanisms, our results suggest that post-transla- show that ductal cells in T1D, which some suggest represent a
tional mechanisms involving factors like COP1, ETV1, and potential source of new b cells (Corritore et al., 2016), lack
STAT3 may govern b cell dysfunction or responses in T2D detectable endocrine physiological phenotypes. Whether from
(O’Shea and Plenge, 2012; Saarima €ki-Vire et al., 2017; Suriben pancreatic cell reprogramming or other sources (Chakravarthy
et al., 2015). et al., 2017; Thorel et al., 2010), rigorous evaluation of ‘‘replace-
Patch-seq also advances our understanding and ability to ment’’ b cells with patch-seq should emerge as a new bench-
study human a cells. Cell size and Na+ channel properties are mark to assess functional and transcriptional resemblance to
key identifiers used to distinguish rodent a and b cells (Briant bona fide b cells.
et al., 2017; Zhang et al., 2014), but human a cell heterogeneity In conclusion, the approaches presented here enable a precise
results in overlap of these electrophysiological properties. Using molecular characterization of endocrine cell physiology and
patch-seq data, we developed additional models for improved represent significant advances in the development of multimodal
cell-type identification of live human islet a and b cells. We scRNA-seq technologies by increasing cell throughput and ex-
also show that specific phenotypes like Na+ current activities tending analyses of simultaneously measured transcriptomic
and cell size can vary significantly in a cells, and that this data and physiological features. Our results help clarify observed
functional heterogeneity corresponds well with expression of inherent islet cellular heterogeneity and provide valuable tools for
transcripts specifying islet cell lineage (ARX, MAFB, and FEV) understanding the molecular mechanisms underlying normal islet
or governing cellular stress (DDIT3 and PPP1R15A). Prior studies function and dysfunction in diabetes. This work also sets a frame-
have reported the impact of the ER stress response in b cell sub- work that could be applied to other cell types where regulated
populations (Baron et al., 2016; Muraro et al., 2016) and in a cells exocytosis and secretion are hallmark features of cellular identity.
(Akiyama et al., 2013; Burcelin et al., 2008; Korsunsky et al.,
2019), suggesting that varying levels of ER stress could drive Limitations of Study
altered or dysfunctional states in both islet cell types. We noted Our patch-seq characterization is performed after islet isolation
a cell subpopulations expressing markers indicating either low or and dispersion into single cells. Although we performed controls
high ER stress, both in ND and in T2D settings. Thus, patch-seq to assess the effect of single-cell dispersion and patch-seq,
merges electrophysiological and transcriptomic data to docu- translation of our findings to in vivo conditions will require further
ment, and suggest a basis for, heterogeneous function and tran- studies. Similarly, by single-cell dissociation, contextual infor-
scriptome regulation in human a cells. mation from neighboring cells and tissue structure is lost.
Phenotyping live islet cells from donors with T1D and other, Patch-seq directly in live pancreas slices could bridge our results
rarer forms of diabetes is a significant challenge that stems to islet structure, although this still poses significant technical
from the paucity of available tissue and low islet cell recovery; challenges in a ribonuclease-rich tissue such as the pancreas.
this has retarded understanding of islet and diabetes biology Throughout this work, we used voltage-clamp recordings of
(Brissova et al., 2018; Wang et al., 2016b). Likewise, procuring ion channels and measurements of exocytosis as a proxy for islet
and studying appropriate control tissue for studies of T1D islets, cell function. Although these are established markers of islet cell
matched in key properties like donor age, is an enduring chal- excitability and function, further experiments recording action
lenge. Electrophysiological studies of islets from only one donor potential firing, Ca2+ signaling, and hormone secretion assays
with T1D have been reported, suggesting that surviving b cells would strengthen gene candidates identified here and their
have normal function and a cells are dysfunctional (Walker mechanistic role in b cell physiology. The PS gene model is
et al., 2011). Similarly, to date only islets from one donor with derived using patch-seq cells from nondiabetic donors. Its
T1D have been studied using scRNA-seq (Wang et al., 2016b). refinement through larger datasets that include more diabetic
Thus, our extension of patch-seq to studies of cryo-stored islets donors should improve the generality of the model and applica-
from ND and T1D donors represents a significant innovation. bility for b cell benchmarking in biomedical settings. For T1D
Here we show that library quality from cryo-preserved islets is studies we used cryopreserved islets. An advantage to this is

1028 Cell Metabolism 31, 1017–1031, May 5, 2020


ll
Resource

that it allowed us to perform all experiments using matched AUTHOR CONTRIBUTIONS


controls to minimize confounding effects; however, it would be J.C.-S. and X.-Q.D. developed the pancreas patch-seq pipeline; X.-Q.D. per-
of interest to confirm these results in independent studies using formed and analyzed patch-clamp data; J.C.-S. performed scRNA-seq and
fresh tissue. analyzed patch-seq data; X.-Q.D. and K.S. performed knockdown experi-
ments; J.C.-S. and Y.H. performed FACS; J.L. and A.B. performed islet isola-
tions; A.B. performed insulin measurements; J.L. performed cryopreservation;
STAR+METHODS J.C.-S., X.-Q.D., Y.H., S.K.K., S.R.Q., and P.E.M. discussed data and inter-
preted results; S.K.K., S.R.Q., and P.E.M. supervised the project; and all au-
Detailed methods are provided in the online version of this paper thors contributed to writing the manuscript.
and include the following:
DECLARATION OF INTERESTS
d KEY RESOURCES TABLE
The authors disclose no conflicts of interest.
d RESOURCE AVAILABILITY
B Lead Contact and Materials Availability Received: November 12, 2019
B Data and Code Availability Revised: January 23, 2020
d EXPERIMENTAL MODEL AND SUBJECT DETAILS Accepted: April 2, 2020
B Islet Isolation, Cryopreservation, and Insulin Secretion Published: April 16, 2020
d METHOD DETAILS
REFERENCES
B Electrophysiological Phenotyping
B siRNA Transfection Aichler, M., Borgmann, D., Krumsiek, J., Buck, A., MacDonald, P.E., Manning
B Islet Dispersion and FACS Fox, J.E., Lyon, J., Light, P.E., Keipert, S., Jastroch, M., et al. (2017). N-acyl
B Single-cell RNA-Seq taurines and acylcarnitines cause an imbalance in insulin synthesis and secre-
d QUANTIFICATION AND STATISTICAL ANALYSIS tion provoking b cell dysfunction in type 2 diabetes. Cell Metab. 25, 1334–
1347.e4.
B Processing and Quality Control of Single-cell RNA-
Ait-Lounis, A., Bonal, C., Seguı́n-Estévez, Q., Schmid, C.D., Bucher, P.,
seq Data
Herrera, P.L., Durand, B., Meda, P., and Reith, W. (2010). The transcription fac-
B Clustering and Cell Type Determination
tor Rfx3 regulates beta-cell differentiation, function, and glucokinase expres-
B Correlation between Electrophysiology and Gene sion. Diabetes 59, 1674–1685.
Expression Akiyama, M., Liew, C.W., Lu, S., Hu, J., Martinez, R., Hambro, B., Kennedy,
B Electrophysiological Predictions Using PS Gene Set R.T., and Kulkarni, R.N. (2013). X-box binding protein 1 is essential for insulin
B Cell Type Identification from Electrophysiological Data regulation of pancreatic a-cell function. Diabetes 62, 2439–2449.
B Statistical Analysis Alejandro, E.U., Gregg, B., Blandino-Rosano, M., Cras-Méneur, C., and
Bernal-Mizrachi, E. (2015). Natural history of b-cell adaptation and failure in
type 2 diabetes. Mol. Aspects Med. 42, 19–41.
SUPPLEMENTAL INFORMATION
Alves, T.C., Pongratz, R.L., Zhao, X., Yarborough, O., Sereda, S., Shirihai, O.,
Cline, G.W., Mason, G., and Kibbey, R.G. (2015). Integrated, step-wise, mass-
Supplemental Information can be found online at https://doi.org/10.1016/j.
isotopomeric flux analysis of the TCA cycle. Cell Metab. 22, 936–947.
cmet.2020.04.005.
Anders, S., Pyl, P.T., and Huber, W. (2015). HTSeq–a Python framework to
work with high-throughput sequencing data. Bioinformatics 31, 166–169.
ACKNOWLEDGMENTS Arda, H.E., Li, L., Tsai, J., Torre, E.A., Rosli, Y., Peiris, H., Spitale, R.C., Dai, C.,
Gu, X., Qu, K., et al. (2016). Age-dependent pancreatic gene regulation reveals
This work was supported by funding from the Chan Zuckerberg Biohub and mechanisms governing human b cell function. Cell Metab. 23, 909–920.
the California Institute for Regenerative Medicine to S.R.Q., a Foundation
Ashcroft, F.M., and Rorsman, P. (2012). Diabetes mellitus and the b cell: the
Grant from the Canadian Institutes of Health Research (CIHR: 148451) to
last ten years. Cell 148, 1160–1171.
P.E.M., grants from the U.S. National Institutes of Health (1U01DK10830001,
1R01DK107507, 1R01DK108817, and P30 DK116074 to S.K.K. and Bader, E., Migliorini, A., Gegg, M., Moruzzi, N., Gerdes, J., Roscioni, S.S.,
1U01DK120447-01 to S.K.K., S.R.Q., and P.E.M.), and JDRF (2-SRA-2019- Bakhti, M., Brandl, E., Irmler, M., Beckers, J., et al. (2016). Identification of pro-
698-S-B) to S.K.K. and P.E.M. Support from the Human Islet Research Core liferative and mature b-cells in the islets of Langerhans. Nature 535, 430–434.
in the Stanford Diabetes Research Center is gratefully acknowledged. Human Bakhti, M., Böttcher, A., and Lickert, H. (2019). Modelling the endocrine
islet isolation at the Alberta Diabetes Institute IsletCore was initially subsidized pancreas in health and disease. Nat. Rev. Endocrinol. 15, 155–171.
by the Alberta Diabetes Foundation. We thank Dr. S.H. Kim (Kim lab, Stanford) Baron, M., Veres, A., Wolock, S.L., Faust, A.L., Gaujoux, R., Vetere, A., Ryu,
and Dylan Hendersson (Chan Zuckerberg Biohub) for help in sample process- J.H., Wagner, B.K., Shen-Orr, S.S., Klein, A.M., et al. (2016). A single-cell tran-
ing, and Dr. Norma Neff (Chan Zuckerberg Biohub) and Jennifer Okamoto scriptomic map of the human and mouse pancreas reveals inter- and intra-cell
(Chan Zuckerberg Biohub) for sequencing expertise. We thank Dr. Linford population structure. Cell Syst. 3, 346–360.e4.
Briant (University of Oxford) for helpful discussions on islet cell-type modeling, Breiman, L. (2001). Random Forests. Mach. Learn. 45, 5–32.
and Dr. Jocelyn Manning Fox (University of Alberta) for critical reading of the
Briant, L.J.B., Zhang, Q., Vergari, E., Kellard, J.A., Rodriguez, B., Ashcroft,
manuscript. We thank the organ procurement organizations across Canada,
F.M., and Rorsman, P. (2017). Functional identification of islet cell types by
particularly the Human Organ Procurement and Exchange (HOPE) program
electrophysiological fingerprinting. J. R. Soc. Interface 14, 20160999.
in Edmonton and the Trillium Gift of Life Network (TGLN) in Ontario, for their
work in obtaining human pancreas for research. We thank Drs. Rita Bottino (Al- Brissova, M., Haliyur, R., Saunders, D., Shrestha, S., Dai, C., Blodgett, D.M.,
legheny Health Network) and Alvin C. Powers (Vanderbilt University) for kindly Bottino, R., Campbell-Thompson, M., Aramandla, R., Poffenberger, G., et al.
providing islets used in validation studies. We also thank Dr. Manning Fox and (2018). a cell function and gene expression are compromised in type 1 dia-
Mrs. Nancy Smith (University of Alberta) for their contributions to human islet betes. Cell Rep. 22, 2667–2676.
isolations. Finally, we are indebted to organ donors and their families for their Broch, M., Vendrell, J., Ricart, W., Richart, C., and Fernández-Real, J.-M.
generous support of scientific research. (2007). Circulating retinol-binding protein-4, insulin sensitivity, insulin

Cell Metabolism 31, 1017–1031, May 5, 2020 1029


ll
Resource
secretion, and insulin disposition index in obese and nonobese subjects. Gan, W.J., Do, O.H., Cottle, L., Ma, W., Kosobrodova, E., Cooper-White, J.,
Diabetes Care 30, 1802–1806. Bilek, M., and Thorn, P. (2018). Local integrin activation in pancreatic b cells
Bunik, V., Kaehne, T., Degtyarev, D., Shcherbakova, T., and Reiser, G. (2008). targets insulin secretion to the vasculature. Cell Rep. 24, 2819–2826.e3.
Novel isoenzyme of 2-oxoglutarate dehydrogenase is identified in brain, but Gembal, M., Gilon, P., and Henquin, J.C. (1992). Evidence that glucose can
not in heart. FEBS J. 275, 4990–5006. control insulin release independently from its action on ATP-sensitive K+ chan-
nels in mouse B cells. J. Clin. Invest. 89, 1288–1295.
Burcelin, R., Knauf, C., and Cani, P.D. (2008). Pancreatic a-cell dysfunction in
diabetes. Diabetes Metab. 34 (Suppl 2 ), S49–S55. Geron, E., Boura-Halfon, S., Schejter, E.D., and Shilo, B.-Z. (2015). The edges
of pancreatic islet b cells constitute adhesive and signaling microdomains. Cell
Burgoyne, R.D., and Morgan, A. (2003). Secretory granule exocytosis. Physiol.
Rep. 10, 317–325.
Rev. 83, 581–632.
Johnston, N.R., Mitchell, R.K., Haythorne, E., Pessoa, M.P., Semplici, F.,
Byrnes, L.E., Wong, D.M., Subramaniam, M., Meyer, N.P., Gilchrist, C.L.,
Ferrer, J., Piemonti, L., Marchetti, P., Bugliani, M., Bosco, D., et al. (2016).
Knox, S.M., Tward, A.D., Ye, C.J., and Sneddon, J.B. (2018). Lineage dy-
Beta cell hubs dictate pancreatic islet responses to glucose. Cell Metab. 24,
namics of murine pancreatic development at single-cell resolution. Nat.
389–401.
Commun. 9, 3922.
Kahn, S.E., Hull, R.L., and Utzschneider, K.M. (2006). Mechanisms linking
Cadwell, C.R., Palasantza, A., Jiang, X., Berens, P., Deng, Q., Yilmaz, M.,
obesity to insulin resistance and type 2 diabetes. Nature 444, 840–846.
Reimer, J., Shen, S., Bethge, M., Tolias, K.F., et al. (2016).
Electrophysiological, transcriptomic and morphologic profiling of single neu- Kaido, T., Perez, B., Yebra, M., Hill, J., Cirulli, V., Hayek, A., and Montgomery,
rons using Patch-seq. Nat. Biotechnol. 34, 199–203. A.M. (2004). Alphav-integrin utilization in human beta-cell adhesion,
spreading, and motility. J. Biol. Chem. 279, 17731–17737.
Chakravarthy, H., Gu, X., Enge, M., Dai, X., Wang, Y., Damond, N., Downie, C.,
Liu, K., Wang, J., Xing, Y., et al. (2017). Converting adult pancreatic islet a cells Keenan, H.A., Sun, J.K., Levine, J., Doria, A., Aiello, L.P., Eisenbarth, G.,
into b cells by targeting both Dnmt1 and Arx. Cell Metab. 25, 622–634. Bonner-Weir, S., and King, G.L. (2010). Residual insulin production and
pancreatic ß-cell turnover after 50 years of diabetes: Joslin Medalist Study.
Chimienti, F., Devergnas, S., Favier, A., and Seve, M. (2004). Identification and Diabetes 59, 2846–2853.
cloning of a beta-cell-specific zinc transporter, ZnT-8, localized into insulin
Korsunsky, I., Millard, N., Fan, J., Slowikowski, K., Zhang, F., Wei, K.,
secretory granules. Diabetes 53, 2330–2337.
Baglaenko, Y., Brenner, M., Loh, P.R., and Raychaudhuri, S. (2019). Fast, sen-
Corritore, E., Lee, Y.-S., Sokal, E.M., and Lysy, P.A. (2016). b-cell replacement sitive and accurate integration of single-cell data with Harmony. Nat. Methods
sources for type 1 diabetes: a focus on pancreatic ductal cells. Ther. Adv. 16, 1289–1296.
Endocrinol. Metab. 7, 182–199.
La Manno, G., Gyllborg, D., Codeluppi, S., Nishimura, K., Salto, C., Zeisel, A.,
Dahl, U., Sjødin, A., and Semb, H. (1996). Cadherins regulate aggregation of Borm, L.E., Stott, S.R.W., Toledo, E.M., Villaescusa, J.C., et al. (2016).
pancreatic beta-cells in vivo. Development 122, 2895–2902. Molecular diversity of midbrain development in mouse, human, and stem cells.
Dallavalle, C., Albino, D., Civenni, G., Merulla, J., Ostano, P., Mello-Grand, M., Cell 167, 566–580.e19.
Rossi, S., Losa, M., D’Ambrosio, G., Sessa, F., et al. (2016). MicroRNA-424 im- Liu, R., Mignardi, M., Jones, R., Enge, M., Kim, S.K., Quake, S.R., and Zou, J.
pairs ubiquitination to activate STAT3 and promote prostate tumor progres- (2019). Modeling spatial correlation of transcripts with application to devel-
sion. J. Clin. Invest. 126, 4585–4602. oping pancreas. Sci. Rep. 9, 5592.
Danielsson, A., Pontén, F., Fagerberg, L., Hallström, B.M., Schwenk, J.M., Lu, D., Mulder, H., Zhao, P., Burgess, S.C., Jensen, M.V., Kamzolova, S.,
Uhlén, M., Korsgren, O., and Lindskog, C. (2014). The human pancreas prote- Newgard, C.B., and Sherry, A.D. (2002). 13C NMR isotopomer analysis reveals
ome defined by transcriptomics and antibody-based profiling. PLoS ONE 9, a connection between pyruvate cycling and glucose-stimulated insulin secre-
e115421. tion (GSIS). Proc. Natl. Acad. Sci. USA 99, 2708–2713.
Dobin, A., Davis, C.A., Schlesinger, F., Drenkow, J., Zaleski, C., Jha, S., Batut, Lyon, J., Spigelman, A., MacDonald, P., and Manning, J. (2019). ADI IsletCore
P., Chaisson, M., and Gingeras, T.R. (2013). STAR: ultrafast universal RNA-seq protocols for the isolation, assessment and cryopreservation of human
aligner. Bioinformatics 29, 15–21. pancreatic islets of Langerhans for research purposes. Published online
Dorrell, C., Schug, J., Canaday, P.S., Russ, H.A., Tarlow, B.D., Grompe, M.T., February 12, 2019. https://doi.org/10.17504/protocols.io.x3mfqk6.
Horton, T., Hebrok, M., Streeter, P.R., Kaestner, K.H., and Grompe, M. (2016). Macaulay, I.C., Ponting, C.P., and Voet, T. (2017). Single-cell multiomics: mul-
Human islets contain four distinct subtypes of b cells. Nat. Commun. 7, 11756. tiple measurements from single cells. Trends Genet. 33, 155–168.
Enge, M., Arda, H.E., Mignardi, M., Beausang, J., Bottino, R., Kim, S.K., and Mahajan, A., Taliun, D., Thurner, M., Robertson, N.R., Torres, J.M., Rayner,
Quake, S.R. (2017). Single-cell analysis of human pancreas reveals transcrip- N.W., Payne, A.J., Steinthorsdottir, V., Scott, R.A., Grarup, N., et al. (2018).
tional signatures of aging and somatic mutation patterns. Cell 171, 321– Fine-mapping type 2 diabetes loci to single-variant resolution using high-den-
330.e14. sity imputation and islet-specific epigenome maps. Nat. Genet. 50,
1505–1513.
Ferdaoussi, M., Dai, X., Jensen, M.V., Wang, R., Peterson, B.S., Huang, C.,
Ilkayeva, O., Smith, N., Miller, N., Hajmrle, C., et al. (2015). Isocitrate-to- Manning Fox, J.E., Lyon, J., Dai, X.Q., Wright, R.C., Hayward, J., van de Bunt,
SENP1 signaling amplifies insulin secretion and rescues dysfunctional b cells. M., Kin, T., Shapiro, A.M.J., McCarthy, M.I., Gloyn, A.L., et al. (2015). Human
J. Clin. Invest. 125, 3847–3860. islet function following 20 years of cryogenic biobanking. Diabetologia 58,
1503–1512.
Fernández-Tajes, J., Gaulton, K.J., van de Bunt, M., Torres, J., Thurner, M.,
Mahajan, A., Gloyn, A.L., Lage, K., and McCarthy, M.I. (2019). Developing a Morgan, N.G., and Richardson, S.J. (2018). Fifty years of pancreatic islet pa-
network view of type 2 diabetes risk pathways through integration of genetic, thology in human type 1 diabetes: insights gained and progress made.
genomic and functional data. Genome Med. 11, 19. Diabetologia 61, 2499–2506.
€dhof, T.C.
Földy, C., Darmanis, S., Aoto, J., Malenka, R.C., Quake, S.R., and Su Muraro, M.J., Dharmadhikari, G., Gru€n, D., Groen, N., Dielen, T., Jansen, E.,
(2016). Single-cell RNAseq reveals cell adhesion molecule profiles in electro- van Gurp, L., Engelse, M.A., Carlotti, F., de Koning, E.J.P., and van
physiologically defined neurons. Proc. Natl. Acad. Sci. USA 113, Oudenaarden, A. (2016). A single-cell transcriptome atlas of the human
E5222–E5231. pancreas. Cell Syst. 3, 385–394.e3.

Fuzik, J., Zeisel, A., Máté, Z., Calvigioni, D., Yanagawa, Y., Szabó, G., Newgard, C.B. (2017). Metabolomics and metabolic diseases: where do we
Linnarsson, S., and Harkany, T. (2016). Integration of electrophysiological re- stand? Cell Metab. 25, 43–56.
cordings with single-cell RNA-seq data identifies neuronal subtypes. Nat. O’Shea, J.J., and Plenge, R. (2012). JAK and STAT signaling molecules in
Biotechnol. 34, 175–183. immunoregulation and immune-mediated disease. Immunity 36, 542–550.

1030 Cell Metabolism 31, 1017–1031, May 5, 2020


ll
Resource
Otter, S., and Lammert, E. (2016). Exciting times for pancreatic islets: gluta- Tabula Muris Consortium; Overall coordination; Logistical coordination; Organ
mate signaling in endocrine cells. Trends Endocrinol. Metab. 27, 177–188. collection and processing; Library preparation and sequencing;
Parnaud, G., Lavallard, V., Bedat, B., Matthey-Doret, D., Morel, P., Berney, T., Computational data analysis; Cell type annotation; Writing group;
and Bosco, D. (2015). Cadherin engagement improves insulin secretion of sin- Supplemental text writing group; Principal investigators (2018). Single-cell
gle human b-cells. Diabetes 64, 887–896. transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562,
367–372.
Pedregosa, F., Varoquaux, G., Gramfort, A., Michel, V., Thirion, B., Grisel, O.,
Blondel, M., Prettenhofer, P., Weiss, R., Dubourg, V., et al. (2011). Scikit-learn: Thorel, F., Népote, V., Avril, I., Kohno, K., Desgraz, R., Chera, S., and Herrera,
machine learning in Python. J. Mach. Learn. Res. 12, 2825–2830. P.L. (2010). Conversion of adult pancreatic a-cells to b-cells after extreme
Perelis, M., Marcheva, B., Ramsey, K.M., Schipma, M.J., Hutchison, A.L., b-cell loss. Nature 464, 1149–1154.
Taguchi, A., Peek, C.B., Hong, H., Huang, W., Omura, C., et al. (2015). Tritschler, S., Theis, F.J., Lickert, H., and Böttcher, A. (2017). Systematic sin-
Pancreatic b cell enhancers regulate rhythmic transcription of genes control- gle-cell analysis provides new insights into heterogeneity and plasticity of the
ling insulin secretion. Science 350, aac4250. pancreas. Mol. Metab. 6, 974–990.
Picelli, S., Faridani, O.R., Björklund, Å.K., Winberg, G., Sagasser, S., and Uhlén, M., Fagerberg, L., Hallström, B.M., Lindskog, C., Oksvold, P.,
Sandberg, R. (2014). Full-length RNA-seq from single cells using Smart- Mardinoglu, A., Sivertsson, Å., Kampf, C., Sjöstedt, E., Asplund, A., et al.
seq2. Nat. Protoc. 9, 171–181. (2015). Proteomics. Tissue-based map of the human proteome. Science
Pipeleers, D.G. (1992). Heterogeneity in pancreatic beta-cell population. 347, 1260419.
Diabetes 41, 777–781. Unger, R.H., and Cherrington, A.D. (2012). Glucagonocentric restructuring of
Prasad, R.B., and Groop, L. (2016). Single-cell sequencing of human pancre- diabetes: a pathophysiologic and therapeutic makeover. J. Clin. Invest.
atic islets-new kids on the block. Cell Metab. 24, 523–524. 122, 4–12.
Rorsman, P., and Braun, M. (2013). Regulation of insulin secretion in human van der Meulen, T., Mawla, A.M., DiGruccio, M.R., Adams, M.W., Nies, V.,
pancreatic islets. Annu. Rev. Physiol. 75, 155–179. Dólleman, S., Liu, S., Ackermann, A.M., Cáceres, E., Hunter, A.E., et al.
Rosen, E.D., Kaestner, K.H., Natarajan, R., Patti, M.-E., Sallari, R., Sander, M., (2017). Virgin beta cells persist throughout life at a neogenic niche within
and Susztak, K. (2018). Epigenetics and epigenomics: implications for dia- pancreatic islets. Cell Metab. 25, 911–926.e6.
betes and obesity. Diabetes 67, 1923–1931. Villani, A.-C., Satija, R., Reynolds, G., Sarkizova, S., Shekhar, K., Fletcher, J.,
Rui, J., Deng, S., Arazi, A., Perdigoto, A.L., Liu, Z., and Herold, K.C. (2017). b Griesbeck, M., Butler, A., Zheng, S., Lazo, S., et al. (2017). Single-cell RNA-seq
cells that resist immunological attack develop during progression of autoim- reveals new types of human blood dendritic cells, monocytes, and progeni-
mune diabetes in NOD mice. Cell Metab. 25, 727–738. tors. Science 356, 1–10.
Saarima €ki-Vire, J., Balboa, D., Russell, M.A., Saarikettu, J., Kinnunen, M., Walker, J.N., Johnson, P.R.V., Shigeto, M., Hughes, S.J., Clark, A., and
Keskitalo, S., Malhi, A., Valensisi, C., Andrus, C., Eurola, S., et al. (2017). An Rorsman, P. (2011). Glucose-responsive beta cells in islets isolated from a pa-
activating STAT3 mutation causes neonatal diabetes through premature in- tient with long-standing type 1 diabetes mellitus. Diabetologia 54, 200–202.
duction of pancreatic differentiation. Cell Rep. 19, 281–294. Wang, Y.J., and Kaestner, K.H. (2019). Single-cell RNA-seq of the pancreatic
Salomon, D., and Meda, P. (1986). Heterogeneity and contact-dependent islets–a promise not yet fulfilled? Cell Metab. 29, 539–544.
regulation of hormone secretion by individual B cells. Exp. Cell Res. 162,
Wang, Y.J., Golson, M.L., Schug, J., Traum, D., Liu, C., Vivek, K., Dorrell, C.,
507–520.
Naji, A., Powers, A.C., Chang, K.-M., et al. (2016a). Single-cell mass cytometry
Segerstolpe, Å., Palasantza, A., Eliasson, P., Andersson, E.-M., Andréasson, analysis of the human endocrine pancreas. Cell Metab. 24, 616–626.
A.-C., Sun, X., Picelli, S., Sabirsh, A., Clausen, M., Bjursell, M.K., et al.
Wang, Y.J., Schug, J., Won, K.-J., Liu, C., Naji, A., Avrahami, D., Golson, M.L.,
(2016). Single-cell transcriptome profiling of human pancreatic islets in health
and Kaestner, K.H. (2016b). Single-cell transcriptomics of the human endo-
and type 2 diabetes. Cell Metab. 24, 593–607.
crine pancreas. Diabetes 65, 3028–3038.
Sharma, U., and Rando, O.J. (2017). Metabolic inputs into the epigenome. Cell
Williams, R. (2016). Williams Textbook of Endocrinology (Elsevier).
Metab. 25, 544–558.
Stefan, Y., Meda, P., Neufeld, M., and Orci, L. (1987). Stimulation of insulin Wolf, F.A., Angerer, P., and Theis, F.J. (2018). SCANPY: large-scale single-cell
secretion reveals heterogeneity of pancreatic B cells in vivo. J. Clin. Invest. gene expression data analysis. Genome Biol. 19, 15.
80, 175–183. Zhang, Q., Chibalina, M.V., Bengtsson, M., Groschner, L.N., Ramracheya, R.,
Stuart, T., and Satija, R. (2019). Integrative single-cell analysis. Nat. Rev. Rorsman, N.J.G., Leiss, V., Nassar, M.A., Welling, A., Gribble, F.M., et al.
Genet. 20, 257–272. (2014). Na+ current properties in islet a- and b-cells reflect cell-specific
Scn3a and Scn9a expression. J. Physiol. 592, 4677–4696.
Suckow, A.T., Comoletti, D., Waldrop, M.A., Mosedale, M., Egodage, S.,
Taylor, P., and Chessler, S.D. (2008). Expression of neurexin, neuroligin, and Zhang, M., Goforth, P., Bertram, R., Sherman, A., and Satin, L. (2003). The
their cytoplasmic binding partners in the pancreatic b-cells and the involve- Ca2+ dynamics of isolated mouse b-cells and islets: implications for mathe-
ment of neuroligin in insulin secretion. Endocrinology 149, 6006–6017. matical models. Biophys. J. 84, 2852–2870.
Suriben, R., Kaihara, K.A., Paolino, M., Reichelt, M., Kummerfeld, S.K., Zhang, C., Moriguchi, T., Kajihara, M., Esaki, R., Harada, A., Shimohata, H.,
Modrusan, Z., Dugger, D.L., Newton, K., Sagolla, M., Webster, J.D., et al. Oishi, H., Hamada, M., Morito, N., Hasegawa, K., et al. (2005). MafA is a key
(2015). b-cell insulin secretion requires the ubiquitin ligase COP1. Cell 163, regulator of glucose-stimulated insulin secretion. Mol. Cell. Biol. 25,
1457–1467. 4969–4976.

Cell Metabolism 31, 1017–1031, May 5, 2020 1031


The science of data

We believe: Data are boundless. Big ideas deserve a big audience.


Insights fuel action.
Patterns, a new open access data science journal from Cell Press, promotes all
types of research outputs and facilitates sharing and collaboration to solve key
scientific problems and aid in the development of solutions for practice, policies,
and management.
The research we publish is both theoretical and practical. We’re committed to
innovations that make research actionable for humans and machines alike.

Share your data story. Submit your paper to cell.com/patterns


ll

Article
Niche-Specific Reprogramming of Epigenetic
Landscapes Drives Myeloid Cell Diversity
in Nonalcoholic Steatohepatitis
Jason S. Seidman,1,10 Ty D. Troutman,1,2,10,* Mashito Sakai,1,10 Anita Gola,3 Nathanael J. Spann,1 Hunter Bennett,1
Cassi M. Bruni,1 Zhengyu Ouyang,1 Rick Z. Li,1 Xiaoli Sun,2 BaoChau T. Vu,1 Martina P. Pasillas,1 Kaori M. Ego,1
David Gosselin,4 Verena M. Link,1,5 Ling-Wa Chong,6 Ronald M. Evans,6,7 Bonne M. Thompson,8 Jeffrey G. McDonald,8
Mojgan Hosseini,9 Joseph L. Witztum,2 Ronald N. Germain,3 and Christopher K. Glass1,2,11,*
1Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
2Department of Medicine, University of California, San Diego, La Jolla, CA, USA
3Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes

of Health, Bethesda, MD 201892, USA


4Department of Molecular Medicine, Université Laval, Quebec City, QC, Canada
5Faculty of Biology, Division of Evolutionary Biology, Ludwig-Maximilian University of Munich, Munich, Germany
6Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
7Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA, USA
8Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX, USA
9Department of Pathology, University of California, San Diego, La Jolla, CA, USA
10These authors contributed equally
11Lead Contact

*Correspondence: ttroutman@health.ucsd.edu (T.D.T.), ckg@ucsd.edu (C.K.G.)


https://doi.org/10.1016/j.immuni.2020.04.001

SUMMARY

Tissue-resident and recruited macrophages contribute to both host defense and pathology. Multiple macro-
phage phenotypes are represented in diseased tissues, but we lack deep understanding of mechanisms con-
trolling diversification. Here, we investigate origins and epigenetic trajectories of hepatic macrophages dur-
ing diet-induced non-alcoholic steatohepatitis (NASH). The NASH diet induced significant changes in Kupffer
cell enhancers and gene expression, resulting in partial loss of Kupffer cell identity, induction of Trem2 and
Cd9 expression, and cell death. Kupffer cell loss was compensated by gain of adjacent monocyte-derived
macrophages that exhibited convergent epigenomes, transcriptomes, and functions. NASH-induced
changes in Kupffer cell enhancers were driven by AP-1 and EGR that reprogrammed LXR functions required
for Kupffer cell identity and survival to instead drive a scar-associated macrophage phenotype. These find-
ings reveal mechanisms by which disease-associated environmental signals instruct resident and recruited
macrophages to acquire distinct gene expression programs and corresponding functions.

INTRODUCTION Resident macrophages and infiltrating monocyte-derived cells


contribute to a diverse array of human diseases (Holtman et al.,
Tissue-resident macrophages exhibit different transcriptomes in 2017; Kiss et al., 2018; Pirzgalska and Domingos, 2018). Nonal-
response to environmental signals, enabling tissue-specific coholic steatohepatitis (NASH) is a form of nonalcoholic fatty
functions (Gautier et al., 2012; Gosselin et al., 2014; Lavin liver disease (NAFLD) and develops through combinatorial ac-
et al., 2014; Mass et al., 2016). Most tissues acquire resident tions of steatosis and growing inflammation in response to
macrophage populations early in development from yolk sac or cellular stresses of the perturbed fatty liver environment (Haas
fetal liver erythromyeloid progenitor (EMP) cells (Ginhoux et al., et al., 2016; Hardy et al., 2016; Machado and Diehl, 2016; Musso
2010; Gomez Perdiguero et al., 2015; McGrath et al., 2015; et al., 2016; Rinella and Sanyal, 2016; Younossi et al., 2016). Dur-
Schulz et al., 2012). In the brain and liver, EMP-derived microglia ing NASH or acute liver injury, new macrophages are derived
and Kupffer cells (KCs) continue as exclusive, self-renewing from circulating monocytes (Karlmark et al., 2009; Seki et al.,
populations. In other tissues, EMP-derived macrophages are 2009a, 2009b; Tacke and Zimmermann, 2014; Zigmond et al.,
partially or completely replaced over time by hematopoietic 2014). These recruited monocyte-derived cells can have both
stem cell (HSC)-derived cells (Bain et al., 2014; De Schepper detrimental and supportive roles, contributing to increases in pa-
et al., 2018; Shaw et al., 2018; Yona et al., 2013). thology during fibrosis onset, but hastening recovery when the

Immunity 52, 1057–1074, June 16, 2020 ª 2020 Elsevier Inc. 1057
ll
Article

damage-evoking agent is removed (Ju and Tacke, 2016; Karl- as Ly6Clo-RM) highly expressed Cd209a, Cd7, and Itgax
mark et al., 2009; Mitchell et al., 2009; Seki et al., 2009a, (Figure S1E).
2009b; Zigmond et al., 2014). These observations raise ques- Using flow cytometry, we observed both Tim4+ and Tim4
tions as to the mechanisms underlying the phenotypic diversity cells in the CD11bloF4/80hi KC gate during NASH in similar
of disease-associated macrophages. proportions to KC-N and KN-RM and collected them for bulk
Here, we define the identities, developmental origins, and mi- RNA-seq and epigenomic profiling (Figure 1D). We also
cro-anatomic locations of the major myeloid populations in collected CD11bhiF4/80lo-Ly6Chi-RM and Ly6Clo-RM for RNA-
response to a NASH-inducing diet. By sorting these cell popula- seq and assay for transposase-accessible chromatin (ATAC-
tions and performing deep transcriptomic and epigenomic anal- seq) (Figure 1E; Buenrostro et al., 2013). RNA-seq analysis of
ysis, we provide evidence for combinatorial effects of diet and these populations yielded concordant expression profiles to
anatomic location on regulatory pathways and transcription fac- scRNA-seq when comparing Seurat marker genes, indicating
tors (TFs) that explain the emergence of disease-associated that we isolated the corresponding macrophage populations
macrophage phenotypes. defined by scRNA-seq (Figure 1F).
We next compared these mouse macrophages to human
RESULTS scRNA-seq populations of non-parenchymal liver cells from in-
dividuals with either advanced cirrhosis or without evident liver
Single-Cell RNA-Seq Defines Myeloid Diversity During disease (Ramachandran et al., 2019). Using orthogonal genes
Dietary NASH to cluster the combined set of human and mouse cells under
To investigate immune cell heterogeneity during NASH, we per- control and disease conditions resulted in eight clusters (0–7,
formed single-cell RNA sequencing (scRNA-seq) on liver non- Figures S1F and S1G). Human livers exhibited a greater diver-
parenchymal cells from healthy mice fed a control diet or sity of myeloid cells and, in contrast to the mouse model, all cell
NASH diet. As noted in previous studies (Hall et al., 2010; Pous- populations were present to some extent in individuals with or
sin et al., 2011), C57BL/6J mice exhibited rapid weight gain without cirrhosis. Importantly, mouse KC-H cells overlapped
when fed the NASH diet (Figure S1A) and developed steatosis, with human KCs (clusters 0 and 1). A key finding of the human
inflammation, and a modest degree of fibrosis (Figure S1B). studies was the increased presence of a ‘‘scar-associated
Non-parenchymal cells from control mice and mice fed the macrophage’’ (SAM) population in cirrhotic livers (Ramachan-
NASH diet for 30 weeks were purified using fluorescence-acti- dran et al., 2019). These cells were found to reside in close
vated cell sorting (FACS) (Figure S1C). Over 6,000 scRNA-seq li- proximity to areas of fibrosis and were characterized by
braries that passed quality thresholds were created from these increased expression of TREM2 and CD9. Importantly, the
isolated cells. cluster containing SAMs also contained substantial fractions
The NASH diet induced qualitative transcriptional differences of the NASH-associated KC-N and KN-RM cells (cluster 3),
in cell clustering between cells from control livers and NASH but not KC-H cells, consistent with the former cell type’s
livers (Figure 1A left, Figure 1B; Table S1). Further, three of 17 increased expression of Trem2 and Cd9 (Figure 1F). Thus,
clusters were primarily derived from cells from control diet despite relatively sparse fibrosis, a SAM phenotype emerges
mice while eight clusters were primarily derived from NASH in this model.
diet mice (Figures 1A and S1D; Table S1). Interrogation of the
most abundant and most differentially expressed (DE) tran- Ontogeny and Environment Contribute to Diversity of
scripts in each cluster allowed a cell identity to be readily as- Myeloid Cells During NASH
signed to most clusters. Further studies were focused on the We hypothesized that, similar to experimental KC ablation (Bon-
five most abundant macrophage clusters. (Figure S1E; nardel et al., 2019; Sakai et al., 2019; Scott et al., 2016), Tim4
Table S1). KC-like macrophages that arise during NASH (KN-RM) may be
The three most abundant clusters (KC-H, healthy KC; ontogenically distinct from the embryonically derived Tim4+
KC-N, NASH KC; and KN-RM, named for being a recruited KCs (KC-N). To test this hypothesis, we performed lineage-
macrophage occupying the KC niche, as described later) tracing experiments using Cx3cr1CreERT2; Rosa26tdTomato/+
had high expression of Adgre1, encoding the tissue macro- mice (Figure S2A; Theurl et al., 2016; Yona et al., 2013). This
phage marker F4/80, and high expression of putative KC model labels tissue macrophages if tamoxifen is administered
lineage-determining transcription factors (LDTFs) Mafb perinatally, while only certain macrophages such as microglia,
and Maf, and KC-specific genes, as well as low expression but not KCs, are labeled if mice are pulsed during adulthood
of Itgam, encoding CD11b (Figures 1C and S1E; Lavin et al., (Dick et al., 2019; Theurl et al., 2016; Yona et al., 2013). In
2014). KC-H consisted almost entirely of cells from controls, contrast, during adulthood, tamoxifen labels peripheral mono-
while KC-N and KN-RM consisted of cells from the NASH cytes and thus monocyte-derived macrophages.
diet animals (Figures 1A and 1B; Table S1). The other two Tamoxifen administration on days 1 and 2 post-parturition re-
major macrophage clusters were predominantly found in sulted in ~50% labeling of embryonic KCs (Figures S2B–S2D).
NASH livers and had characteristic gene expression of After 20 weeks on the NASH diet, nearly all tdTomato+ cells
previously described monocyte-derived liver macrophages were Tim4+ KCs (KC-N) (Figures S2B–S2D), indicating that
(Figures 1C and S1E; Krenkel et al., 2018). Cluster 4 (hereafter Tim4 KC-like macrophages (KN-RM) are not long-lived daugh-
referred to as Ly6Chi-RM) expressed higher amounts of ters of embryonic KCs. Conversely, when adult mice fed the
transcripts typical of Ly6Chi monocytes such as Ly6c2, NASH diet for 20 weeks were given tamoxifen 4 or 1 week prior
Chil3, F13a1, and Fn1, while cluster 9 (hereafter referred to to sacrifice, ~90% of circulating monocytes were TdTomato+

1058 Immunity 52, 1057–1074, June 16, 2020


ll
Article

A F

D E

Figure 1. Transcriptional Diversity of Hepatic Macrophages during NASH


(A) tSNE projections of identified graph-based cell clusters from scRNA-seq data derived from hepatic CD45+CD146 cells from control mice or mice fed a NASH
diet for 30 weeks. (Left) cells are colored based on dietary condition. (Right) cells are colored based on cell identity of the five major myeloid cells identified. Data
represent two independent donor mice per group.
(B) Macrophage proportions from (A) for control mice (left) or mice with NASH (right).
(C) Gene expression of normalized scRNA-seq data of genes supporting cluster identities. Boxes denote quartiles, and whiskers denote data range excluding
outliers.
(D) Terminal FACS gates for purification of KCs from control mice (left), or CD11bloF4/80hiTim4+ and Tim4 cells from mice with NASH (right).
(E) Terminal FACS gates for purification of Ly6Chi and Ly6Clo CD11bhiF4/80loLy6G CX3CR1+ recruited hepatic macrophages (RM). ‘‘PMN’’ stands for poly-
morphonuclear leukocyte.
(F) Comparison of myeloid clusters defined by scRNA-seq (left) and bulk RNA-seq (right) for the corresponding sorted populations. Left heatmap (100 cells per
column) depicts normalized and scaled expression values for marker genes identified using a Wilcoxon rank sum test through Seurat. Right heatmap shows z-
normalized row expression of each gene for RNA-seq from bulk purified cell populations from independent biological duplicates.
Please see also Figure S1.

2 days after the final tamoxifen injection (data not shown), and could be due to low expression of Cx3cr1 in Tim4+ KC-N cells
more than 10% of Tim4 KC-like macrophages (KN-RM) were or to the gradual upregulation of Tim4 by recruited cells (Scott
TdTomato+ (Figures S2C and S2D). We also observed a small et al., 2016). Overall, these results indicate that embryonic KCs
degree of labeling of Tim4+ KCs (Figures S2C and S2D), which remain in the liver during development of NASH and are Tim4+,

Immunity 52, 1057–1074, June 16, 2020 1059


ll
Article

A B
NASH diet
E-cadherin tdTomato+ F4/80+

Ly6Chi&lo-RM KN-RM Ly6Chi&lo-RM Mgl2+


17% KN-RM (Ly6Clo-RM)
KC-N
PV

tdTomato

Mgl2
MFI
6.94%

cv
75.3%
PV Mgl2–
KC-N
(Ly6Chi-RM)
PV

F4/80 Tim4 F4/80

C D F
E-cadherin Ly6Chi&lo-RM KN-RM KC-N 1000 ***
*** E-cadherin Mgl2+ Mgl2–

neighbor distance (μm)


750

KC−N nearest
PV

500
CV PV
***
250

CV 0

M M M
−R hi −R lo −R
cv KN y6C y6C
L L
E G
PV PV ***
Distance to nearest

Distance to nearest
125 *** *** ***
large vessel (μm)

800

CV or PV (μm)
**
100 *** ***
600
75
400
50
PV
200
25
PV
0 0
CV PV CV PV
−N RM M
KC KN− hi&lo −R Ly6Chi−RM Ly6Clo−RM
6 C
H Ly
F4/80 TUNEL Tim4 F4/80 TUNEL Tim4
KC-N

KN-RM
NASH diet

KN-RM

KN-RM

KC-N

I J K
TUNEL nearest neighbor
Total TUNEL+ cell count

***
100 *** 100
per area 1,000 μm2

Percent KN-RM

***
1000
distance (μm)

75
1 100
50

10 25
0.01

1 0
KC−N KN−RM Ly6Chi&lo−RM 0 10 20 30
−H RM RM −N RM RM Weeks on AMLN NASH diet
KC KN− hi&lo − KC KN− hi&lo −
6C 6C
Ly Ly
Control NASH

(legend on next page)


1060 Immunity 52, 1057–1074, June 16, 2020
ll
Article

while most recruited CD11bloF4/80hi macrophages are Tim4 . sinusoids, but were highly enriched around both portal and cen-
These results also indicate that recruited KN-RM cells can tral vein vasculature identifiable by the large vessel diameter
survive at least 4 weeks in the NASH liver while Ly6Chi&lo-RMs (15 mm or larger) (Figures 2D and 2E). Further, Ly6Chi-RM
are shorter lived. (Mgl2 ) cells were bi-modally distributed around portal and
To determine the localization of myeloid cells in the liver during central veins, while Ly6Clo-RM (Mgl2+) cells were more uniformly
NASH, immunofluorescence (IF) microscopy was performed on distributed in closer proximity to central veins (Figures 2F and
livers from Cx3cr1CreERT2; Rosa26tdTomato/+ mice pulsed with 2G). Together, these findings further supported Ly6Chi&lo-RM
tamoxifen 7 days prior to tissue collection (Figures 2A, 2B, cells as being positionally separated from KC-N and KN-RM
S2A, S2E, and S3A). As expected, tdTomato+ monocyte-derived cells based on measurements that were independent of
cells were abundant in NASH livers but nearly absent in control anatomical landmarks. These results provide evidence that
livers (Figure S2E). Using multi-parameter imaging and histo-cy- KC-N and KN-RM cells reside in a similar niche within the liver
tometry (Gerner et al., 2012), we distinguished KC-H or KC-N, sinusoids, which is spatially distinct from the anatomic positions
KN-RM, and Ly6Chi&lo-RM in the control and NASH livers based occupied by Ly6Chi&lo-RMs.
on Tim4, F4/80, and tdTomato expression. As Ly6C histological Previous work found that DLL4 expression by liver sinusoidal
staining could not be performed, Mgl2 was used as a surrogate endothelial cells (LSECs) is graded toward higher expression
marker for Ly6Chi&lo-RM, with the Ly6Chi fraction being Mgl2 patterns in periportal zones (Halpern et al., 2018). Further,
and Ly6Clo being represented by Mgl2+ (Figures 2A–2C, see Fig- DLL4 mediated-activation of RBPJ participates in driving differ-
ure 3G for Ly6Clo-specific expression of Mgl2). Following in situ entiation of monocytes to Kupffer-like cells in a repopulation
confirmation of these myeloid cell subsets, we assessed their model (Sakai et al., 2019). During NASH, we confirmed the
spatial distribution using the positional data preserved in histo- peri-portal (E-cadherin+) enrichment of DLL4 on LSECs (Figures
cytometry and nearest-neighbor distance analyses. The two S3D and S3E). Further, KC-N and KN-RM cells were significantly
subsets (KN-RM and Ly6Chi&lo-RM) that were increased during closer to DLL4+ CD138+ LSECs compared to Ly6Chi&lo-RMs
NASH displayed significant differences in their spatial distribu- (Figures S3F and S3G). These results provide support for DLL4
tions. By comparing nearest-neighbor distances from KC-N, as a factor that may support niche specialization of distinct
KN-RM cells were found to be distributed significantly closer macrophage populations during NASH, consistent with its
to KC-N cells than Ly6Chi&lo-RM cells (Figure 2D). In addition, known function in instructing a KC program in recruited
the small number KN-RM cells observed in control livers were monocytes.
also in close proximity to KC-H cells (Figure S3B and data
not shown). NASH Diet Induces KC Death and Replacement
KCs reside within the hepatic sinusoids, but the sub-anatom- KCs are considered a self-renewing population that is normally
ical organization of other liver macrophages during NASH is closed to HSC-derived cells (Perdiguero and Geissmann,
less certain (Ju and Tacke, 2016). High-magnification imaging 2016). However, the observation that the NASH diet resulted in
using Collagen IV to visualize the endothelial basement mem- recruitment of HSC-derived KN-RM cells raised the question of
brane demonstrated that KC-N and KN-RM cells resided within whether the NASH diet induces loss of embryonic KCs, providing
hepatic sinusoids in this model (Figure S3C). In contrast, Ly6- an open niche. TUNEL staining of NASH livers showed apoptosis
Chi&lo-RM cells were predominantly not found within the liver of Tim4+ KC-Ns during NASH diet, but not of KC-H cells from

Figure 2. Expanded Macrophage Diversity during NASH Is Supported by Monocyte Recruitment and Occupancy of Distinct Anatomical
Niches
(A) Confocal image of NASH liver showing E-cadherin (to demark the peri-portal regions) and tdTomato F4/80+ summed channel. Highlighted in the center by
white circles are the central veins (CV). Demarked at the periphery of the liver lobule, portal-venous or arterial vessels (PV). Data from n = 3–4 mice per condition.
Scale: 50 microns.
(B) Histo-cytometry analysis of NASH liver sample. Statistical information of segmented objects (F4/80+tdTomato+ surfaces) was imported into FlowJo and
subsequently gated on F4/80 and tdTomato expression to quantify KC-N, KN-RM, and Ly6Chi&lo-RM cells. Tim4 mean fluorescence intensity (MFI) for each gated
population are shown in the middle panel, and Mgl2 expression on Ly6Chi&lo-RM cells macrophage is shown at right to distinguish Ly6Chi (Mgl2 ) or Ly6Clo
(Mgl2+) cells.
(C) Confocal image of NASH liver showing distribution of rendered surfaces of KC-N, KN-RM, and Ly6Chi&lo-RM (Rec) cells. Dashed lines denote peri-portal
regions as in (A). Scale: 50 microns.
(D) Distance and phenotype (KN-RM, Mgl2lo-RM, or Mgl2hi-RM) of closest neighbor to KC-N cells in NASH livers. Data pooled from n = 4 mice. Wilcox two-sided
test; p < 0.001(***).
(E) Distance to nearest portal or central vein vasculature (large diameter vessels defined to be greater than 15 mm in diameter) of KC-N, KN-RM, and Ly6Chi&lo-RM
cells. Kruskal-Wallis with Dunn test; p < 0.001(***).
(F) Zoom-in representative confocal image of NASH liver showing distribution of rendered surfaces of Mgl2lo (Ly6Chi) and Mgl2hi (Ly6Clo)-RMs. E-cadherin
demarks the peri-portal regions, and highlighted by white circles are CV and PV blood vessels. Scale: 30 microns.
(G) Distance to nearest center of CV or PV of Ly6Chi and Ly6Clo RM cells (mm). Kruskal-Wallis with Dunn test; p < 0.01(**), p < 0.001(***).
(H) Immunofluorescence (IF) image assessing in situ cell death via TUNEL staining in addition to Tim4 and F4/80 staining. Image from NASH mice; maximum
intensity projection (MIP) of a 20-mm z stack. Scale: 5 microns for the top row and 10 microns for the bottom row.
(I) Quantification of total TUNEL+ hepatic macrophages per area (1000 mm2) in Ctrl and NASH mouse livers.
(J) Nearest neighbor distance of all TUNEL+ cells to KN-RM, Ly6Chi or lo-RM, and KC-N cells in NASH livers, data pooled from n = 4 mice. Kruskal-Wallis with Dunn
test; p < 0.001(***).
(K) Temporal assessment of percentage of total CD11bloF4/80+CD146 cells that are Tim4 KN-RM from mice fed a NASH diet as indicated.
Please see also Figure S2 and Figure S3.

Immunity 52, 1057–1074, June 16, 2020 1061


ll
Article

A EMP Blood
C Mean expression
D Mean expression
Ly6Chi (log2(TPM+1)) (log2(TPM+1))
15 15
316 53

KN-RM
10 10

KC-H
Ly6Chi Ly6Clo 5 5
KC-H KC-N KN-RM -RM -RM

Ctrl NASH 521 40


0 0
Sinusoids Large vessels 0 5 10 15 0 5 10 15
KC-N KC-N
Cluster 1

E F

2,210 genes; Log2FC > 2; p.adj < 0.05; TPM > 4


15 15
1046 1442

Ly6CHi-RM
10 10

KN-RM
Cluster 3 Cluster 4

5 5

0 1193 0 685

0 5 10 15 0 5 10 15
Ly6Chi-RM Blood Ly6Chi Mono
Cluster 2

G Kupffer cell signature ROS metab. signature


C6 Arg2
1500
Cd163 200 Txnrd1
1000
100
500
Mean Expression (TPM)

z-scaled (Log2(TPM+1)) 0 0
−2 −1 0 1 2
K H
KN C-N
6C -RM

K H
Ly N- -N
6C RM
6C M

6C RM

o
6C M

6C M
-

-
-M

-M
KC

KC

K C
Ly h-iR

Ly -R

Ly -R
hi

hi
Ly h-i
lo

lo
B Ly6Chi-RM
Ly

30
Ly6Clo-RM Scar-assoc. mac (SAM) Response to wounding
20 800
RLM 12h Cd9 Clec10a
75
Trem2 Mgl2
PC2 (19%)

10 RLM 1d 600
Blood 50
0 KN-RM 400
Ly6Chi
200 25
−10 KC-N Monocytes
(Ctrl and NASH)
0 0
−20 RLM 14d
-H

Ly KN -N
Ly h-i M

-H

Ly KN -N
Ly h-i M
6C RM

6C RM

o
o
6C M

6C M
6C -R

6C -R

-M
-M
KC
KC

KC
KC
Ly -R

Ly -R

−30 KC-H
hi
hi
lo

lo

−20 0 20 40 60
PC1 (57%)
H I Mean expression J K NASH CRN
Clec4f+Tim+ Clec4f+Tim– (log2(TPM+1)) Score
15 6
DTR–

Total KC KC KC

6
DTR+, n= 12

10 3 10 4
Percent CD45+

4 2
5
DTR+

5 2
2 1 DTR–
0
DTR+
DTR –
0 5 10 15 0
0 DTR+ 0 0 DTR–, n = 10

(legend on next page)

1062 Immunity 52, 1057–1074, June 16, 2020


ll
Article

mice fed a control diet (Figures 2H and 2I). In both control and KN-RM cells exhibited more than 2,200 DE genes in compar-
NASH livers, TUNEL+ KN-RM and Ly6Chi&lo-RM cells were rare ison to Ly6Chi-RM, despite both cells originating from an HSC
(Figure 2I). By nearest-neighbor analysis of TUNEL+ cells, KN- precursor (Figure 3E). The divergent differentiation programs
RM cells were significantly enriched in nearby areas as of KN-RM and Ly6Chi-RM were further reinforced by pairwise
compared to Ly6Chi&lo-RM and KC-N cells (Figure 2J). Time comparisons with gene expression in Ly6Chi circulating
course experiments indicated that KN-RM cells were detected monocytes, indicating more than 2,000 DE genes in each
by 10 weeks after initiation of the NASH diet and progressively case (Figures 3F and S4A).
accumulated to account for more than 50% of the KC population Prior studies defined a set of KC identity genes that distinguish
at 30 weeks (Figure 2K). These results indicate that Tim4+ KCs the KC from other tissue-resident macrophages (Lavin et al.,
undergo cell death during NASH, potentially resulting in partial 2014). Twenty-eight of the genes downregulated during the tran-
opening of the niche and enabling repopulation by HSC-derived sition of KC-H cells to KC-N cells in the NASH model are among
KN-RM cells. this set, exemplified by Cd163 and C6 (Figures 3G and S4B).
However, many KC-specific genes maintained similar expres-
Effect of NASH Diet on Resident and Recruited Myeloid sion in cells from mice on the NASH diet, including C1qa, Cd5l,
Cell Transcriptomes Id3, and Il18bp, indicating that only a subset of the KC-specific
The ability to sort distinct populations of hepatic myeloid cells gene expression program is altered during NASH. Furthermore,
defined by scRNA-seq analysis enabled deep transcriptomic KC identity genes such as Clec4f, Vsig4, and Cdh5 were highly
profiling of each population (Table S2). Unsupervised hierar- expressed in KN-RM, but neither Ly6Chi&lo-RM population.
chical clustering of 2,210 DE genes among KC-H, KC-N, Considering the differential spatial distribution of these cell pop-
KN-RM, Ly6Chi-RM, and Ly6Clo-RM tightly grouped KC-N ulations, such findings suggest that the KC niche is necessary to
and KN-RM and distinguished KC-H, KC-N, and KN-RM cells promote induction of these genes (Figure S4B).
from Ly6Chi&lo-RMs (Figure 3A). With inclusion of DE genes in Gene ontology analyses of genes whose expression distin-
blood monocytes, principal component analysis (PCA) also guishes KC-H, KC-N, and KN-RM cells from Ly6Chi&lo-RM
indicated that KC-N and KN-RM cells from NASH mice group- (clusters 1 and 2, Figure 3A) were consistent with a stronger
ed most closely with KC-H cells from healthy mice (Figure 3B). pro-inflammatory and wound repair phenotype of the Ly6-
After experimental KC ablation, blood Ly6Chi monocytes are Chi&lo-RMs (Figure S4C), in agreement with prior studies (Hey-
recruited to the liver and rapidly differentiate into KC-like liver mann and Tacke, 2016). Examples of DE genes associated
macrophages (Bonnardel et al., 2019; Sakai et al., 2019; Scott with the functional categories of ‘‘ROS metabolism’’ and
et al., 2016). At 14 days after KC ablation, these repopulating ‘‘response to wounding’’ are illustrated in Figure 3G. Ly6Clo-
macrophages (GEO: GSE128662) also grouped closely with RM preferentially expressed Mgl2 (CD301b), a gene recently re-
KC-H, KC-N, and KN-RM (Figure 3B). Collectively, these re- ported to be expressed in skin macrophages that activates a
sults suggest that monocyte-derived KN-RMs follow a similar specific myofibroblast population implicated in tissue repair
developmental trajectory in the context of NASH to that of re- and aging (Shook et al., 2018). In addition, we observed
populating liver macrophages (RLMs) in the KC abla- increased expression of Trem2 and Cd9 in both KC-N and
tion model. KN-RM cells, consistent with recent observations identifying
Pairwise comparisons of KC-H and KC-N cells indicated these genes as markers for macrophages associated with
that the NASH diet had a large impact on resident KC gene increased lipid burden and the presence of tissue scarring
expression (>800 DE genes), consistent with scRNA-seq (Hill et al., 2018; Jaitin et al., 2019; Ramachandran et al.,
data (Figure 3C). Notably, fewer than 100 genes were identi- 2019; Xiong et al., 2019). Collectively, these results indicate
fied as significantly altered when comparing KC-N and KN- that the NASH diet acts to reprogram the endogenous KC pop-
RM KCs from mice with NASH (Figure 3D), despite their ulation and induce divergent programs of differentiation of KN-
distinct origins (EMP versus HSC). In contrast, during NASH, RM cells and Ly6Chi&lo-RMs.

Figure 3. Highly Divergent Gene Expression Patterns across Myeloid Populations in NASH
(A) Unsupervised hierarchical clustering of DE genes in the indicated cell types in control and NASH liver.
(B) PCA of 2,000 most variable genes in RNA-seq data from myeloid cells in liver and blood from healthy and NASH-diet-fed mice, or recruited liver macrophages
from Sakai et al. (2019), n = 2–3 per group.
(C) Scatterplot of RNA-seq data in healthy (KC-H) or NASH (KC-N) Tim4+ KCs. DE genes identified by DESeq2 (FC > 2, p-adj < 0.05) are colored in red.
(D) Comparison of NASH Tim4+ KCs (KC-N) and Tim4 Kupffer niche recruited macrophages (KN-RM).
(E) Comparison of NASH Tim4 Kupffer niche recruited macrophages (KN-RM) and Ly6Chi-RM.
(F) Comparison of NASH blood Ly6Chi monocytes and Ly6Chi-RM.
(G) RNA-seq expression (mean TPM ± SD) of representative genes. TPM = transcripts per kilobase million).
(H) Percentage of hepatic CD45+F4/80hiCD11bloLiveSinglets from Clec4f-cre R26-iDTR / (n = 6) or Clec4f-cre R26i-DTR+ (n = 6) mouse livers. Mice were treated
with DT, allowed to rest for 4 weeks, then fed the choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) for 4 weeks.
(I) RNA-seq from whole liver tissue from Clec4f-cre R26-iDTR / (n = 10) or Clec4f-cre R26-iDTR+ mice (n = 12) as in (H). Zero DE genes (DESeq2). Boxes denote
quartiles, and whiskers denote data range.
(J) Representative (n > 5) H&E staining of liver tissue.
(K) NASH Clinical Research Network (CRN) scoring of stained liver sections. Scores > 5 are indicative of steatohepatitis. No significant difference was identified
using Kruskal-Wallis rank sum test (p = 0.71). Boxes denote quartiles, and whiskers denote data range.
Please see also Figure S4.

Immunity 52, 1057–1074, June 16, 2020 1063


ll
Article

Functional Convergence of Embryonic KCs and KN-RMs RM cells were highly enriched for motifs recognized by LXR
in NASH and members of the MAF and TFE families (Figure 4E). In
To clarify the functional biological roles of embryonic KCs and contrast, ATAC-seq peaks specific for Ly6Clo-RM cells were
monocyte origin KN-RM cells, we used the Clec4f-cre R26- highly enriched for NF-kB motifs and for motifs recognized by
iDTR system (Sakai et al., 2019). We treated DTR+ or DTR RUNX, ZEB, and KLF TFs. Open chromatin regions lost from
mice with diphtheria toxin (DT) to ablate embryonic KCs and blood monocytes during acquisition of either the KC or Ly6Chi&lo
then allowed the KC niche to repopulate with monocyte precur- -RM niche signatures (Figures 4B and 4C, blue points) were en-
sors. After a 4-week recovery period, the mice were subjected to riched for KLF and C/EBP motifs (Figures S5B and S5C). The
ad libitum feeding for 4 weeks with a rapid NASH-inducing model open chromatin regions lost during acquisition of the KC niche
(Matsumoto et al., 2013). Ablation of embryonic (KC-H) KCs additionally included motifs for RUNX and CTCF (Figure S5B).
before feeding mice the NASH diet led to increased proportions The pattern of motif enrichment in KC-N and KN-RM cells im-
of Tim4 KN-RM cells, as expected (Figure 3H). To assess the plies that liver niche signals increase the expression and/or activ-
global molecular consequence of replacing embryonic KCs ities of TFs that bind to LXR, MAF, and TFE motifs. Consistent
with monocyte-derived KN-RMs, we performed RNA-seq on to- with this possibility, Nr1h3 (encoding LXRa), Mafb, Tfec, and
tal liver tissue (Figure 3I). Notably, we found no DE genes be- Id3 (Lavin et al., 2014; Mass et al., 2016) are among a set of
tween the two groups. We also found no differences in 23 circu- KC LDTFs higher expressed in KC-N and KN-RM cells than in
lating inflammatory cytokines (Figure S4D). Finally, quantitative Ly6Chi&lo-RM cells (Figure 4F; Table S2). Importantly, these
histological assessment of liver sections indicated no effect of TFs are highly induced in RLMs within 12 h of entry into the
replacing KCs with KN-RM. These results are consistent with open KC niche of the liver (Sakai et al., 2019). These findings
the largely convergent transcriptomes observed in comparison further support a model in which KN-RM cells largely follow the
of KC-N and KN-RM (Figures 3J and 3K). developmental program taken by RLMs following KC depletion
upon adherence to LSECs. In contrast, the lack of induction of
Niche Occupancy Reprograms the Epigenetic KC LDTFs in Ly6Chi&lo-RM cells is consistent with their location
Landscapes outside of the sinusoidal space. NF-kB motifs are enriched in
To investigate mechanisms responsible for environment-spe- KC enhancers in comparison to other tissue-resident macro-
cific programs of gene expression, we identified accessible phages (Sakai et al., 2019), but the particularly strong enrichment
chromatin defined by ATAC-seq in five populations of cells for this motif in Ly6Clo-RM cells implies that the niche occupied
from mice with NASH: KC-N and KN-RM cells, Ly6Chi&lo-RMs, by these cells provides additional signals that activate NF-kB. In
and Ly6Chi blood monocytes. Examples of ATAC-seq peaks in addition, while RUNX factors are lower in KC-N and KN-RM cells
these five populations of cells from mice fed the NASH diet in in comparison to blood Ly6Chi monocytes, their expression is
the vicinity of the Clec4f and Itgam genes, which are highly differ- maintained or increased in Ly6Chi&lo-RMs (Figure S5D). In con-
entially expressed in KCs compared to Ly6Chi&lo-RMs, are illus- cert, analysis of open chromatin provides evidence that the
trated in Figure 4A. Genome-wide comparisons of ATAC-seq divergent patterns of gene expression observed in HSC-derived
peak tag counts for Ly6Clo-RM versus Ly6Chi blood monocytes KN-RM and Ly6Chi&lo-RM cells are in part determined by
are illustrated in Figure 4B, and the same comparison for KN-RM whether or not they receive niche-specific signals necessary to
and Ly6Chi blood monocytes is shown in Figure 4C. Differential adequately induce KC LDTFs.
regions from the five myeloid populations in NASH mice, as
well as healthy KC-H cells and 24- and 48-h RLMs after KC abla- NASH Diet Reprograms the KC Enhancer Landscape
tion (Sakai et al., 2019), were used for PCA illustrated as in We next sought to understand the basis for the altered expres-
Figure 4D. PC1, accounting for ~65% of variance, primarily sion of the more than 900 mRNAs during the transition of KC-H
distinguished KC-H, KC-N, and KN-RM populations from Ly6Chi cells in the healthy liver to KC-N cells in the NASH liver (Figures
blood monocytes and Ly6Chi&lo-RM. RLMs 24 and 48 h after KC 3A and 3C). Corresponding changes in the regulatory land-
ablation became incrementally closer to KCs along PC1, reflect- scapes during this transition would potentially enable inference
ing chromatin remodeling after arriving at the KC niche. PC2, ac- of TFs and upstream signaling pathways that mediate re-
counting for ~13% of variance, primarily separated Ly6Chi blood sponses to the NASH diet. In contrast to changes in open chro-
monocytes from Ly6Chi&lo-RM. matin observed in transition of Ly6Chi blood monocytes to KN-
Hierarchical clustering of ATAC-seq data further support the RM cells (Figures 4C and 4D), relatively few differences in open
relationships suggested by PCA, with KC-N and KN-RM cells ex- chromatin were observed comparing KC-H and KC-N cells
hibiting highly similar patterns of open chromatin that are distinct (Figure 5A, left). The enriched motifs associated with the
from the transitional patterns observed in Ly6Chi&lo-RM (Fig- increased chromatin accessibility included sites for AP1 and
ure S5A). Based on these findings and the results of lineage- EGR factor binding, whereas sites of decreased chromatin
tracing experiments, we considered the open chromatin regions accessibility were enriched for PU.1 and SpiC motifs (Figure 5A
of KN-RM and Ly6Clo-RM populations as divergent endpoints of right).
chromatin remodeling events following entry of Ly6Chi blood To investigate potential changes in the transcriptional func-
monocytes into the NASH liver. ATAC-seq peaks specific for tions of these regions, we performed chromatin immunoprecip-
KN-RM or Ly6Clo-RM in comparison to circulating Ly6Chi mono- itation sequencing (ChIP-seq) for acetylation of histone H3 lysine
cytes (red data points in Figures 4B and 4C, respectively) indi- 27 (H3K27ac) in KC-H cells in the healthy liver and KC-N cells in
cated that approximately 75% of these peaks were specific to the NASH liver. H3K27ac is deposited by histone acetyltrans-
KN-RM or Ly6Clo-RM cells (Figure 4E). Peaks specific for KN- ferases (HATs) associated with transcriptional co-activators

1064 Immunity 52, 1057–1074, June 16, 2020


ll
Article

A ATAC-seq RNA ATAC-seq RNA B C


ATAC-seq ATAC-seq
Clec4f Itgam (log2(norm. tags +1)) (log2(norm. tags +1))
5 kb 10 kb 14 14
Ly6Chi 3e3
4e2
2e3
Blood 1e3 2e2
12 8507 12 6732
0 0
3e3
Ly6Chi 2e3
4e2 10 10

Ly6Clo-RM
-RM 1e3 2e2

KN-RM
0 0 8 8
Ly6Clo 3e3
4e2
2e3 6 6
-RM 1e3 2e2
0 0 4
3e3
4
4e2
KC-N 2e3
2 2
1e3 2e2 3509 2458
0 0
3e3 0 0
KN-RM 4e2
2e3
1e3 2e2 0 2 4 6 8 10 12 14 0 2 4 6 8 10 12 14
0 0
Ly6Chi Blood Ly6Chi Blood

D Ly6Chi E
Blood
Control Gained during niche acquisition versus
40
KC-H ATAC-seq peaks Ly6Chi peripheral blood monocytes
hi
Ly6C
20 Blood Motif P- %targ/ Best
NASH value %bkgd Match
KC-N
0 GG
AA
GTCACA G
TGT AC GT 1e-101 30/17 TFE/MITF
PC2 (13%)

C A
TTAA
TG

RLM
T
CTG
CC G T A G T
C
G
AC G
CC
T A CA

48hr RLM TGACCT TGTGACCCT 1e-91


ATTG
C
AGATCG
AA
CGC
T
TT
CGAA
C
A
A
5.1/0.9 LXRE
24hr
G
CC T G A G
A
C G
C A T G T
C G T G T A G A

−20 4,500 only


C
G
AGTCAGCA 1e-79 33/21 MAF
KN-RM T C
A T G
C AG T
AG
C T C
T
G T A
A G
C T

Ly6Chi-RM up in KN-RM CGTGGGAGAGC


G
T
A
CC
T
T
C
A
G A
A
C
A
TTC
C
A
G
TT
CT
A C
A
TG
1e-22 12/7.6 RBPJ
−40 GGG T C
AATTCC C
A
GAG
TAC
TGC 1e-91 20/6.8 NF-κB
2,226 up C
AGT
A C T A C TT
C
GG
CCG A
G AG T AA G T G
ATA
TCCT

−60 in both GAAAAACGATGGAAAAT


C T
A G
C T G
C
T
C T GG T
C G
C TG T
A C T C
G AC A T C T A G T
C G T
C G T
C G T
C G
C A
1e-79 2.1/.02 NFAT
6,279 only up CA
G

C
T
G
TGACGTCATC
AC
AT

C
T

G T
C

G
C A
GCAA
G
C
TA
A G
C
G
C

G
C A
GC
T

G T
G

G T
C
T
GT
CG
AA
A G
C A G
C A G
C A G
C A G T A G
C A G
C A
1e-75 31/16 JUN-CRE
Ly6Clo-RM in Ly6Clo GG T CCCC
−80 AAAAC
T
A GT
TTCG
AT
AA
T
A
1e-522 34/10 NF-κB
-RM
C
TCCGC G GT GT
G
A
G

GG
C
CA
TA
AAGC
TCT
TC
GC
T
C
GTAT
C
G
AA
T
C
AGGTGA G
C
A
T 1e-148 37/23 ZEB
−100 −50 0 50 100 CAA
A ACCT
ACAG 1e-102 39/27 RUNX
GGC TG
TG
A
T
TC C TG A T G T AG C G T A G
C T C

PC1 (65%)

F Nr1h3 Mafb Tfec Id3


Mean Expression (TPM)

800 400 400


60
600 300 300

40
400 200 200

200 100 20 100

0 0 0 0
KC-H
KC-N
KN−RM
Ly6Chi-RM
Ly6Clo-RM
Ly6Chi Blood

KC-H
KC-N
KN−RM
Ly6Chi-RM
Ly6Clo-RM
Ly6Chi Blood

KC-H
KC-N
KN−RM
Ly6Chi-RM
Ly6Clo-RM
Ly6Chi Blood

KC-H
KC-N
KN−RM
Ly6Chi-RM
Ly6Clo-RM
Ly6Chi Blood

Figure 4. Niche-Specific Reprogramming of Epigenetic Landscapes


(A) UCSC genome browser tracks of ATAC-seq signals in the vicinities of the Clec4f and Itgam genes in the indicated cell types. Bar plots to the right of each track
represent the RNA-seq gene expression (mean TPM ± SD) in each population.
(B) Genome-wide comparison of normalized ATAC-seq peak tags at enhancer-like regions (>3 kb removed from transcription start site [TSS]) comparing Ly6Chi
blood monocytes and Ly6Clo-RM during NASH. Differential regions were identified using DESeq2 (FC > 2 and p-adj < 0.05 using independent biological du-
plicates).
(C) Genome-wide comparison of normalized ATAC-seq peak tags at enhancer-like regions (>3 kb removed from TSS) comparing open chromatin in Ly6Chi blood
monocytes and KN-RM.
(D) PCA of ATAC-seq datasets (n = 2–3) for the top 10,000 most variable distal (>3 kb from TSS) regions in myeloid cell populations during NASH, or recruited liver
macrophages (RLM) from Sakai et al. (2019).
(E) De novo motifs enriched in distal open chromatin regions (>3 kb from TSS) enriched in KN-RM (top), Ly6Clo-RM (bottom), or enriched in both populations
(middle) compared to Ly6Chi blood monocytes during NASH. The background for motif enrichment analysis is the distal open chromatin from Ly6Chi blood
monocytes.
(F) Expression (mean TPM ± SD) of KC LDTFs in the indicated cell types.
Please see also Figure S5.

Immunity 52, 1057–1074, June 16, 2020 1065


ll
Article

A D
ATAC-seq Differential Kupffer ATAC-seq
(log2(norm. tags +1)) RNA- vs H3K27ac ChIP-seq
14 P- %targ/ Best
402 Motif 10 R = 0.70
value %bkgd match

log2(KC-N/KC-H) RNA-seq
12
G
CTGAGTCA
T
A G
C A C T A C
G T
C T
A G
C A G T AG T
C
1e-87 69/22 AP1/ATF
ACCCACGCG
CG
10 T 1e-15 19/7 EGR2 5
TT AAA
TTGC
ACGATCG
G C A T A G T G A G T T

8
ACTTCCTCTT
KC-N

G G
A
T
AA 1e-24 60/13 PU.1/SpiC
0
CG T T A G
C A G
C A G T
A G A
C
TCGG A C
G

6
4 C −5
2 Effect of NASH on H3K27ac
90 All enhancers KC signature
0
Up 10%
0 2 4 6 8 10 12 14 Up 3.5% −6 −4 −2 0 2 4 6
KC-H Down Down log2(KC-N/KC-H) H3K27ac ChIP-seq
8% 24%
B E
H3K27ac ChIP-seq
(log2(norm. tags +1)) Egr2 ***
Differential Kupffer H3K27ac ChIP-seq
43352 total 10452 KC sig Atf3 ***
14 P- %targ/ Best Fos
4201 Motif value %bkgd match Srebf1
365 KC sig ***
12 Jun *
GA
T
CC
TTGA TCA
AG
T
C
G
A
T A
C
G
A
T
1e-137 35/19 AP1/ATF Irf8 ***
TTTCTAATATTTTCC
G
C A C A G T
C G
C A G T G T
C

10 A
A

1e-25 0.7/.04 NFAT Runx1


G
C TGCCG CC AT

***
TGTGGTTA
G
C A A G
C A G A C G T G T C T G A G A C
G A G
C A G T G A

C A 1e-20 11/7.3 RUNX Mafb **


8 CGCCCACC
G
C A C T A G
C A C T A C T A G A G
C G
C
KC-N

G
T
A
C T
A G T A G A
T
G G
TTTTA G
A CG A
1e-19 16/11 EGR Nr1h2 ***
Mitf *
6
Irf7 *
A AGGTCA
TCG
A
1e-33 49/39 NR half-site
4
G
C G
T C T C T A C T A G
C A G T A G T
C

Nr1h3
AGTGGTCACAGGAC 1e-25 AATAT
T
AG C
T
G
A
0.4/0.0 MITF Irf5
TCTGTCAGCCTATCTG
G T
C C T A G
C C T C A G
C G A G T
C G T C C T A T A G T
C

C 1e-19 0.3/0.0 MAF


2 CA C G GAG GG Maf
GAAAGTAAAAGCAC 1e-17
G A G T G
C A T A G
C A T A G T
C C T A T A G T C A C T C A G T A G
C A T A

3583 C T A G T
C G T
C G T
C C
C
TAC A G G T G T
C G T
C G T
C CG
T A T A G T
C G T A
0.3/0.0 IRF Tfec **
0 2553 KC sig Irf1 **
G GGTTACTA AGGTCA 1e-15 3.5/1.6 LXRE
C
CG
A C T
TCG G T
Spic
0 2 4 6 8 10 12 14
C TC
T
ATCGTA
G
G
A C AG A
A G
C AC TC T
A
C
A TG
C ATG G A
T
C

(known) ***
KC-H 0 2 4
log2(NASH/Ctrl)
F G RNA-seq (TPM)
C6 Cd9
5 kb ATAC 20 kb ATAC
KC-H
RNA-seq (TPM)

KC-N 100 Atf3


Egr1
H3K27ac H3K27ac 50 Egr2
Spic
0

Cd163 Trem2
10 kb ATAC 5 kb ATAC
800 C6
RNA-seq (TPM)

Cd163
400 Cd9
H3K27ac H3K27ac
Trem2
0

0 1 4 10 20 30
Weeks on AMLN diet

Figure 5. The NASH Diet Alters the Activity States of Resident KC Enhancers
(A) Left: Normalized ATAC-seq signal at all distal open chromatin regions (>3 kb from TSS) in Tim4+ KCs from healthy mice (KC-H) or Tim4+ KCs from mice on
NASH diet (KC-N). Regions with significantly more chromatin accessibility during NASH are colored in red, while regions with less accessibility during NASH are
colored blue. Right: De novo motif enrichment from differentially accessible chromatin region shown at right.
(B) Left: H3K27ac ChIP-seq signal around distal (>3 kb from TSS) ATAC-seq peaks in a 2,000 bp window. Differentially acetylated regions were determined using
DESeq2 (FC > 2, p-adj < 0.05). Regions overlapping with KC signature enhancers (Figure S6A) are colored green. Enhancers with more acetylation during NASH
are colored red, or orange if also a KC signature enhancer. Enhancers with less acetylation during NASH are colored blue, or purple if also a KC signature
enhancer. Right: Representative motif enrichment from differentially acetylated chromatin regions.

(legend continued on next page)


1066 Immunity 52, 1057–1074, June 16, 2020
ll
Article

and is highly correlated with regulatory element activity motifs matching AP1, NFAT, RUNX, and EGR TFs (Fig-
(Creyghton et al., 2010). Out of 43,352 total distal open chro- ure 5B right).
matin regions defined by ATAC-seq, 4,201 putative enhancers Comparison of KC-H to KC-N cells indicated significant induc-
gained H3K27ac and 3,583 putative enhancers lost H3K27ac tion of Atf3, Fos, Jun, Egr2, and Runx1 mRNAs (Figures 5E and
in response to the NASH diet (Figure 5B), affecting 18% of the 5G), suggesting that increased expression of these TFs during
enhancer-like regions overall (Figure 5C). NASH may contribute to activation of enhancers with corre-
A comparison of H3K27ac associated with ATAC-seq peaks in sponding DNA binding elements (Figure 5B). Conversely,
KC-H cells versus in other macrophage populations enabled mRNAs encoding a subset of KC LDTFs were downregulated
definition of a set of 10,452 putative KC-H signature enhancers in response to the NASH diet, including Spic, Irf1, and Tfec.
(Figure S6A; Sakai et al., 2019). A profound overlap was Notably, SpiC (encoded by Spic) binds to a motif nearly identical
observed between KC signature enhancers and enhancers to that recognized by PU.1 that is enriched at genomic regions
downregulated by the NASH diet, in total identifying 2,553 KC exhibiting loss of open chromatin in NASH. In contrast, while
signature downregulated enhancers (24% of KC signature en- LXR motifs were highly enriched at genomic regions exhibiting
hancers, 71% of all downregulated enhancers, Fisher’s exact loss of open chromatin, expression of Nr1h3, encoding LXRa,
test p value < 0.001) (Figures 5B and 5C). Conversely, only 365 was not affected by the NASH diet, and expression of Nr1h2, en-
KC signature enhancers were upregulated by NASH diet (3.5% coding LXRb, was slightly increased (Figure 5E).
of KC signature enhancers, 8.7% of all upregulated enhancers, To relate changes in TF expression to global changes in gene
Fisher’s exact test p value = 1) (Figures 5B and 5C). Changes expression, we performed RNA-seq analysis of KCs following
in H3K27ac were highly correlated with changes in expression 1, 4, 10, 20, and 30 weeks of the NASH diet. Coordinated
of the nearest gene (Figure 5D). These findings suggest a prefer- changes in gene expression began to occur between 4 and
ential suppressive effect of the NASH diet on gene regulatory 10 weeks of the NASH diet, with upregulation of Atf3 and
networks governing the function of KC enhancers, in line with Egr2 associated with increases in Trem2 and Cd9 expression
the corresponding downregulation of KC identity genes and downregulation of Spic associated with decreases in
(Figure S4B). Cd163 and C6 expression (Figure 5G). These findings indicate
ATAC-seq and H3K27ac ChIP-seq tracks associated with the that acquisition of features corresponding to the LAM or SAM
downregulated KC identity genes C6 and Cd163 illustrated sub- phenotype characterized by high expression of Trem2 and
stantial loss of H3K27ac at their respective promoters and distal Cd9 requires prolonged exposure to the NASH diet, which ulti-
regions of open chromatin (Figure 5F). The opposite pattern was mately results in substantial reprogramming of the KC regula-
observed at the upregulated lipid-associated macrophage (LAM) tory landscape.
and SAM-defining genes Trem2 and Cd9 (Jaitin et al., 2019;
Ramachandran et al., 2019). In addition, Trem2 and Cd9 both NASH Diet Selectively Impairs LXR Regulation of KC
provided examples of genes in which new regions of open chro- Identity Genes
matin are established that are associated with H3K27ac (Fig- The observation that LXR binding motifs were highly enriched in
ure 5F), representing putative NASH-dependent enhancers. genomic regions exhibiting diet-induced loss of H3K27ac led to
Both pre-existing and NASH-induced regions associated with a focused analysis of LXR function. Comparison of effects of
Trem2 were shifted but conserved at the TREM2 locus in human LXR deletion with effects of the NASH diet on KC gene expres-
microglia (Figure S6C). Additional examples of NASH-induced sion indicated that a subset of LXR target genes associated
genes in KCs and associated gene ontologies are shown in with its role as a KC LDTF were strongly downregulated by
Figure S6B. the NASH diet. For example, Cd5l, Timd4, Cd209l, Pcolce2,
Motif enrichment analysis of regions of open chromatin and Plac8 require LXRa for expression (Sakai et al., 2019),
exhibiting loss of H3K27ac in response to the NASH diet and all but Cd5l exhibited significantly reduced expression in
showed enrichment for binding sites recognized by LXR, response to the NASH diet (Figure 6A). However, many canon-
MAF, and IRF TFs (Figure 5B right). Each of these motifs are ical LXR target genes such as Abca1, Abcg1, Mylip, and Srebf1
recognized by TFs that are established or proposed to drive were significantly upregulated in response to the NASH diet
KC identity (Lavin et al., 2014; Mass et al., 2016; Sakai (Figure 6B; Table S3), arguing against a general loss of LXR
et al., 2019; Scott et al., 2018). In contrast, open chromatin function. Targeted lipidomic analysis of known LXR ligands
regions with gained H3K27ac were enriched for de novo (Peet et al., 1998) in liver indicated that the concentration of

(C) Summaries of percent representation of (left) differentially activated enhancers between control and NASH KCs, or (right) differentially acetylated enhancers
intersected with the KC signature enhancers.
(D) Ratio-ratio plot depicting fold change in H3K27ac ChIP-seq signal at enhancers (2,000 bp window centered on ATAC-seq peaks >3 kb from TSS) compared to
fold change in mRNA expression of closest gene annotated to enhancer region in Tim4+ KCs in NASH diet mice (KC-N) versus healthy mice (KC-H). Points colored
in blue are significantly different (FC > 2, p-adj < 0.05) for both H3K27ac ChIP-seq signal at enhancers and closest mRNA. Pearson correlation of 0.70 denotes
relationship between the highlighted differential data points.
(E) Log2FC of candidate TFs known to bind DNA elements found enriched in (B) for KCs from healthy mice (KC-H) and NASH mice (KC-N). *p-adj < 0.05, **p-adj <
0.01, and ***p-adj < 0.001 using DESeq2.
(F) UCSC genome browser tracks of ATAC-seq or H3K27ac ChIP-seq signals in the vicinities of the indicated genes.
(G) Mean TPM (LOESS fit) of the indicated genes in Tim4+ KCs from mice fed the AMLN NASH diet as indicated (0 week: n = 3; 1 week: n = 2; 4 week: n = 2; 10
week: n = 3; 20 week: n = 3; 30 week: n = 4).
Please see also Figure S6.

Immunity 52, 1057–1074, June 16, 2020 1067


ll
Article

A B C

D E

Figure 6. Genome-wide Occupancy of LXR Binding in KCs during NASH


(A) RNA-seq expression (mean TPM ± SD) of LXR-dependent KC signature genes. *p-adj < 0.05, **p-adj < 0.01, and ***p-adj < 0.001 using DESeq2.
(B) RNA-seq expression (mean TPM ± SD) of canonical LXR target genes.
(C) Quantification of desmosterol, 24-, 25-, and 27-OHC and 24,25-EC in livers from control or NASH mice. *** = p < 0.001.
(D) Mean LXR ChIP-seq signal at merged irreproducibility discovery rate (IDR) peaks from KC nuclei. Nuclei were sorted from Clec4f-Cre-NLS-TdTomato mice fed
either a control diet (n = 2) or the NASH diet for 20 weeks (n = 2). Peaks with significantly altered LXR binding were determined using DESeq2 (FC > 2, p-adj < 0.05).
(E) ATAC-seq, H3K27ac ChIP-seq, and LXR ChIP-seq signals in the vicinities of the indicated genes.
(F) Normalized distribution of LXR ChIP-seq density or H3K27ac ChIP-seq density at enhancers with either significantly altered LXR binding during NASH or all
LXR bound enhancers.
(G) Left: De novo motif enrichment for NASH-gained LXR peaks (right) or NASH-lost LXR peaks (right) shown in Figure 7D.
Please see also Figure S7.

1068 Immunity 52, 1057–1074, June 16, 2020


ll
Article

A B ATF3 ChIP-seq
IDR peaks
LXR gained LXR lost LXR total Control NASH
AP1/ATF3 7
Motifs per bp per peak

0.008 NR/LXR

Log2(Tags+1)
6
Egr2 0.004 0.003 5
PU1 25,507 4
0.006
0.003 3
0.002 2
0.004 0.002 1
2,808 0
0.002 0.001 0.001
13,291
0.000
0.000 0.000
−200 −100 0 100 200 −200 −100 0 100 200 −200 −100 0 100 200
1,000 bp
Distance from peak (bp)

C D
NASH specific ATF3 vs all ATF3 peaks NASH TF ocupancy at
P- %targ/ Best NASH gained enhancers
Motif value %bkgd match None 32%
ATF3 10%
1e-671 49.26/23.38 AP1/ATF3 LXR 3%

1e-111 11.06/5.27 Egr2/Egr1

1e-69 42.57/33.94 MITF/USF1


Both 54%

E
ChIP-seq tag densities centered on genomic regions co-bound by LXRs and ATF3
ATF3 LXR P300 H3K27ac
ChIP fragment depth

12 8 Control
20 10.0
per bp per peak

15 9 6 NASH
7.5

10 5.0 6 4

5 2.5 3
2
0
0 0 0 00 00 0 0 0 00 00 0 0 0 00 00 0 0 0 00 00
00 00 10 20 00 00 10 20 00 00 10 20 00 00 10 20
−2 −1 −2 −1 −2 −1 −2 −1
Distance from enhancer (bp)

F Control NASH
Trem2 Cd9 Arhgap22 Kcnn4
5 kb 10 kb 20 kb 10 kb
P300 ATF3 LXR

G H I
Mean expression Mean expression Arhgap22 Cd9 Itgax
(log2(TPM+1)) (log2(TPM+1))
*** * ***
12.5 80
10.0 43 significant 300
Mean Expression (TPM)

Gpnmb Mmp12 150 60


Clec4f-Cre+ − KN-RM

Itgax Trem2 100 40


200
(LXRα/β KC-DKO)

Arhgap22 10.0
Cd9 50 100
NASH − KC-N

7.5 20
0 0 0
7.5
146 significant

5.0 Kcnn4 Tgfb3 Trem2


5.0 *** ** n.s.
25 80
2.5 30 20
2.5 60
20 15
10 40
10 5 20
0.0 0.0 0 0 0
0.0 2.5 5.0 7.5 10.0 0.0 2.5 5.0 7.5 10.0 12.5 - + - + - +
re re re re re re
Control − KC-H Clec4f-Cre- − KN-RM f-C f-C f-C f-C f-C f-C
l e c4 lec4 l e c4 lec4 l e c4 lec4
(LXRα/β WT) C C C C C C

Figure 7. Combinatorial Actions of LXR and ATF3 Coordinate NASH Responsive Gene Expression in KCs during NASH
(A) Motif frequency within 500 bp of LXR binding sites described in Figure 6F.
(B) Normalized ATF3 ChIP-seq signal at merged IDR peaks from KC nuclei. Nuclei were sorted from Clec4f-Cre-NLS-TdTomato mice fed either a control diet or
the NASH diet for 20 weeks (n = 2). Significant ATF3 peaks had Poisson enrichment p value < 0.0001 and FC > 4.
(legend continued on next page)
Immunity 52, 1057–1074, June 16, 2020 1069
ll
Article

desmosterol, which is by far the most abundant endogenous induced changes in the binding and function of LXRs were the
LXR ligand in the liver (Sakai et al., 2019; Yang et al., 2006), consequence of altered collaborative binding interactions with
was significantly increased in the setting of the cholesterol- AP-1 factors, EGRs, and members of the ETS TF family PU.1
rich NASH diet in comparison to the control diet (Figure 6C). and/or SpiC. To examine this possibility, we focused on ATF3,
All other natural LXR agonists were present at much lower con- as it was the most highly induced AP-1 factor in response to
centrations (data not shown). The increase in desmosterol is the NASH diet (Figure 5E) and recognized the AP-1 motif en-
consistent with the upregulation of general LXR target genes riched in NASH-specific enhancers (Figure 5B). Similar to
involved in cholesterol homeostasis. A corresponding increase expression amounts of Atf3, we detected ~2.5 times as many
in desmosterol was reported in livers of human subjects with peaks in nuclei from mice with NASH (data not shown). This
NASH and cirrhosis (Gorden et al., 2015). observation correlated with an overall increased magnitude of
To investigate whether changes in gene expression are asso- ATF3 ChIP-seq signal during NASH (Figures 7B and S7D). En-
ciated with corresponding changes in LXR binding, we per- hancers with gained ATF3 binding during NASH were enriched
formed ChIP-seq for LXRa + LXRb in the combination of KC- for de novo motifs corresponding to AP-1, EGR, and MITF2
N and KN-RM cells marked by sorting nuclei prepared from TFs (Figures 7C and S7C).
rapidly fixed control or NASH livers for nuclear Clec4f-tdTo- Intersection of the LXR and ATF3 ChIP-seq datasets indicated
mato expression (Figure S7A). These studies yielded high qual- ~28,000 instances of overlapping peaks (Figure S7E). Of the
ity datasets that could not be obtained using conventionally 1,793 LXR binding sites increased by >2-fold in NASH, ~80%
sorted cells and indicated that the NASH diet induced LXR overlapped with ATF3 peaks, consistent with a collaborative
binding by more than 2-fold at more than 1,700 locations and binding relationship. Notably, the combination of LXR and
resulted in a loss of binding at more than 1,000 locations (Fig- ATF3 was observed at 54% of NASH-induced enhancers (Fig-
ure 6D). Notably, DNA binding was relatively unchanged at ure 7D). The majority of these sites were occupied by LXRs under
most of the LXR binding sites associated with canonical LXR control conditions, but exhibited substantial increases in
activity (e.g., Abca1), which are highly LXR-dependent but not H3K27ac in the context of NASH (Figure 7E). This result sug-
affected at the mRNA level by the NASH diet (Figure 6E). In gested that diet-induced binding of ATF3 to these sites resulted
contrast, LXR binding in the vicinity of Timd4, Pcolce2, and in the subsequent recruitment of HATs. We therefore performed
Plac8 was markedly reduced. Overall, LXR ChIP-seq peaks ChIP-seq for the HAT P300 and found its recruitment to the LXR
were preferentially depleted from KC signature enhancers and ATF3 peaks in response to the NASH diet, indicating a likely
(405/1046, or ~39% of total downregulated LXR peaks in en- role in contribution to acetylation of H3K27 and subsequent
hancers, blue points, Fisher’s exact test p value < 0.001) enhancer function (Figures 7E and S7F–S7H). Representative
compared to LXR signal gained at KC signature enhancers examples of NASH-induced co-binding of LXRs and ATF3 or
(128/1795, or ~7% of total upregulated LXR peaks in en- for NASH-induced binding of ATF3 at pre-existing LXR binding
hancers, red points, Fisher’s exact test p value = 1) (Figures sites are illustrated for Trem2, Cd9, Arhgap22, and Kcnn4 in
6D and S7B). Sites at which LXR binding was gained were Figure 7F.
associated with increased H3K27ac, whereas the opposite Analysis of the genes nearest to enhancers bound by ATF3
pattern was observed at sites at which LXR binding was lost, and LXR in the context of the NASH diet revealed a shift in
consistent with a positive regulatory function of LXR at these lo- expression distribution toward a higher mean during NASH (Fig-
cations (Figure 6F). ure 7G). To directly evaluate NASH-specific functions of LXRs re-
sulting from these induced binding sites, we crossed
NASH-Induced Collaborative Binding Partners LXRafl/flLXRbfl/fl mice to the Clec4f-cre-TdTomato mouse to
Repurpose LXRs generate KC-N and KN-RM-specific LXRa / LXRb / mice.
Motif analysis of genomic regions exhibiting gain or loss of LXR The progeny were then fed either a control diet or the NASH
binding in response to the NASH diet indicated enrichment for diet for 20 weeks. Due to the near complete absence of KC-N
AP-1 or ATF, EGR, and MITF motifs at gained sites, and PU.1 cells in the Clec4f-cre-TdTomato LXRafl/flLXRbfl/fl mice, compar-
or SPIC, GLIS3, and nuclear receptor half site motifs at lost sites isons were made of control and Clec4f-cre-TdTomato
(Figure 6G). The distance relationships of these motifs to the cen- LXRafl/flLXRbfl/fl KN-RMs. By comparing LXR-dependent gene
ter of the LXR peak were consistent with collaborative binding in- expression in KN-RMs under these two conditions, we identified
teractions observed for other LDTFs (Figure 7A; Heinz et al., a set of 146 NASH-specific LXR-dependent genes (Figures 7H
2015). These findings suggested the possibility that NASH- and 7I), 33 of which were part of the set defined in Figure 7G.

(C) De novo motif enrichment for NASH-specific ATF3 peaks as in (B).


(D) Intersection of ATF3 and/or LXR IDR ChIP-seq peaks from KC nuclei isolated from NASH-diet-fed mice with enhancers with increased activity during NASH
(from Figure 5B).
(E) Normalized distribution of ChIP-seq tag density at sites co-bound by LXRs and ATF3 indicated in (D).
(F) UCSC genome browser tracks of LXR, ATF3, and P300 ChIP-seq signals in the vicinities of the indicated genes. Peaks with NASH-gained (HOMER) binding for
both LXR and ATF3 are shaded yellow, and peaks with only gained ATF3 are shaded green.
(G) Mean expression (TPM) for genes nearest to NASH-specific LXR peaks co-bound by ATF3. DE genes identified using p-adj < 0.05 and FC > 2.
(H) RNA-seq (n = 2 per group) of KN-RM from control mice or Clec4fCre-LXRafl/flLXRbfl/fl mice fed the NASH diet for 20 weeks. DE genes identified as in (G).
(I) As in (H) for selected genes. **p-adj < 0.01; ***p-adj < 0.001.
Please see also Figure S7.

1070 Immunity 52, 1057–1074, June 16, 2020


ll
Article

Notably, LXR dependency was observed both at genes populations provides a basis for understanding how disease-
exhibiting NASH-induced binding of LXR (e.g., the SAM-associ- promoting environmental signals instruct resident and recruited
ated gene Cd9) and genes exhibiting recruitment of ATF3 to pre- macrophages to acquire distinct pathogenic programs of gene
existing LXR binding sites (e.g., Arhgap22 and Kcnn4). Trem2 expression. Application of the methods utilized in this study
expression was reduced in LXRa / LXRb / KN-RMs, but did should prove valuable in gaining a better understanding of
not retain statistical significance after correction for multiple how distinct myeloid phenotypes are established in other dis-
testing. Collectively, these findings provide evidence that ease contexts.
NASH-induced changes in collaborative TFs result in altered
binding and function of LXRs, contributing to the NASH-specific STAR+METHODS
program of KC gene expression.
Detailed methods are provided in the online version of this paper
DISCUSSION and include the following:

Here, investigation of genomic and anatomic relationships d KEY RESOURCES TABLE


among phenotypically distinct macrophage populations in a d RESOURCE AVAILABILITY
mouse model of NASH revealed a strong association of niche B Lead Contact
occupancy with the transcriptional and phenotypic states of B Materials Availability
different EMP- and HSC-derived myeloid cells. HSC-derived B Data and Code Availability
macrophages observed in this model of NASH reside in at least d EXPERIMENTAL MODEL AND SUBJECT DETAILS
two distinct niches associated with divergent pathways of dif- B Mice
ferentiation. One pathway corresponds to the development of B NASH-Model Diets
Ly6Chi&lo-RMs, which reside proximal to large portal and cen- B Diphtheria toxin (DT)-mediated depletion of
tral vein vessels in contact with conventional (but most likely in- Kupffer cells
flamed) vascular endothelium. An alternative pathway results in d METHOD DETAILS
the accumulation of KN-RM cells, which colocalized with KC-N B Microscopy and Histo-Cytometry
in contact with the LSECs and adopted a similar pattern of B Nuclei Sorting
gene expression. This process occurred concordantly with pro- B Cell Sorting and Flow Cytometry
gressive loss of resident KC-Ns, most likely due to apoptosis. B Single-Cell RNA-Seq
These findings provide strong evidence for instructive roles of B ATAC-Seq
the LSECs and the NASH diet in determining the molecular B Poly A RNA-Seq
phenotypes of both KN-RM and KC-N cells. Of particular inter- B Chromatin Immunoprecipitation
est, we observed that a subset of KC-N and KN-RM cells ac- B ChIP-Seq Library Preparation
quire similarities to the recently identified SAM that resides in B Lipid Measurements
areas of fibrosis in human cirrhotic livers (Ramachandran d QUANTIFICATION AND STATISTICAL ANALYSIS
et al., 2019). B Sequencing Data Analysis
The transitions of open chromatin, H3K27ac and genome-
wide locations of LXRs and ATF3 provide evidence for SUPPLEMENTAL INFORMATION
NASH-induced alterations in expression and activities of
ATF3, resulting in reprogramming KC-specific functions of Supplemental Information can be found online at https://doi.org/10.1016/j.
LXRs. In the context of relatively constant LXR expression, immuni.2020.04.001.

increased ATF3 and likely other AP-1 TFs are suggested to


drive redistribution of LXR to NASH-specific enhancers as ACKNOWLEDGMENTS

well as activate poised enhancers bound by LXRs under control


These studies were supported by NIH grants DK091183, HL088083,
conditions. The decreased expression of Spic in the context of DK063491, and GM085764 and Fondation Leducq grant 16CVD01. J.S.S.
the NASH diet may also contribute to loss of collaborative LXR was supported by American Heart Association Fellowship 16PRE30980030
binding at a subset of KC-specific enhancers. The delineation and NIH Predoctoral Training grant 5T32DK007541. T.D.T. was supported
of NASH-induced regulatory elements associated with the by P30 DK063491, T32DK007044, and NRSA T32CA009523. M.S. was sup-
Cd9 and Trem2 genes that are occupied by LXRs and ATF3 ported by the Manpei Suzuki Diabetes Foundation of Tokyo, Japan and the
Osamu Hayaishi Memorial Scholarship for Study Abroad, Japan. X.S. was
suggest roles of these TFs in establishing the SAM and/or
supported by the American Heart Association grant 18POST34060088.
LAM phenotypes. A key remaining question is to determine This work was also supported in part by the Intramural Research program
whether the altered functions of LXRs in KCs are pathogenic of NIAID, NIH.
or represent an adaptive response to the NASH diet.
Overall, these studies are consistent with a model in which AUTHOR CONTRIBUTIONS
distinct microenvironments within the NASH liver drive diver-
Conceptualization, J.S.S., T.D.T., M.S., A.G., R.N.G, and C.K.G.; Methodol-
gent patterns of differentiation of resident and infiltrating cells
ogy, J.S.S., T.D.T., M.S., A.G. and N.J.S.; Formal Analysis, J.S.S, T.D.T.,
by remodeling the open chromatin landscapes of recruited
A.G., Z.O., R.Z.L., and M.H.; Investigation, J.S.S., T.D.T., M.S., A.G.,
monocytes and altering the activities of pre-existing enhancers C.M.B., N.J.S., H.B., B.T.V., M.P.P., K.M.E., D.G., and L.-W.C.; Data Curation,
of the resident KC population. The inference of TFs and up- J.S.S., T.D.T, and Z.O; Writing – Original Draft, J.S.S., T.D.T, A.G., R.N.G, and
stream signaling pathways associated with these distinct cell C.K.G.; Writing – Review & Editing, J.S.S, T.D.T., M.S., A.G., V.M.L., R.N.G.,

Immunity 52, 1057–1074, June 16, 2020 1071


ll
Article
and C.K.G.; Funding Acquisition, J.S.S., T.D.T., R.N.G, and C.K.G.; Visualiza- Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S., Mehler,
tion, J.S.S., T.D.T., A.G., and Z.O. M.F., Conway, S.J., Ng, L.G., Stanley, E.R., et al. (2010). Fate mapping analysis
reveals that adult microglia derive from primitive macrophages. Science 330,
DECLARATION OF INTERESTS 841–845.
Gomez Perdiguero, E., Klapproth, K., Schulz, C., Busch, K., Azzoni, E., Crozet,
J.L.W. and X.S. are named inventors on patent applications or patents
L., Garner, H., Trouillet, C., de Bruijn, M.F., Geissmann, F., and Rodewald, H.R.
related to the use of oxidation-specific antibodies that are held by UCSD.
(2015). Tissue-resident macrophages originate from yolk-sac-derived erythro-
J.L.W. is a consultant to Ionis Pharm and is a scientific founder of Oxi-
myeloid progenitors. Nature 518, 547–551.
tope, Inc.
Gorden, D.L., Myers, D.S., Ivanova, P.T., Fahy, E., Maurya, M.R., Gupta, S.,
Received: February 3, 2020 Min, J., Spann, N.J., McDonald, J.G., Kelly, S.L., et al. (2015). Biomarkers of
Revised: April 1, 2020 NAFLD progression: a lipidomics approach to an epidemic. J. Lipid Res. 56,
Accepted: April 8, 2020 722–736.
Published: May 1, 2020 Gosselin, D., Link, V.M., Romanoski, C.E., Fonseca, G.J., Eichenfield, D.Z.,
Spann, N.J., Stender, J.D., Chun, H.B., Garner, H., Geissmann, F., and
REFERENCES Glass, C.K. (2014). Environment drives selection and function of
enhancers controlling tissue-specific macrophage identities. Cell 159,
Baddeley, A., Rubak, E., and Turner, R. (2016). Spatial point patterns: method- 1327–1340.
ology and applications with R (Boca Raton, London, New York: CRC Press,
Gosselin, D., Skola, D., Coufal, N.G., Holtman, I.R., Schlachetzki, J.C.M., Sajti,
Taylor & Francis Group).
E., Jaeger, B.N., O’Connor, C., Fitzpatrick, C., Pasillas, M.P., et al. (2017). An
Bain, C.C., Bravo-Blas, A., Scott, C.L., Perdiguero, E.G., Geissmann, F., Henri, environment-dependent transcriptional network specifies human microglia
S., Malissen, B., Osborne, L.C., Artis, D., and Mowat, A.M. (2014). Constant identity. Science 356, https://doi.org/10.1126/science.aal3222.
replenishment from circulating monocytes maintains the macrophage pool in
Haas, J.T., Francque, S., and Staels, B. (2016). Pathophysiology and
the intestine of adult mice. Nat. Immunol. 15, 929–937.
Mechanisms of Nonalcoholic Fatty Liver Disease. Annu. Rev. Physiol. 78,
Bonnardel, J., T’Jonck, W., Gaublomme, D., Browaeys, R., Scott, C.L.,
181–205.
Martens, L., Vanneste, B., De Prijck, S., Nedospasov, S.A., Kremer, A., et al.
(2019). Stellate Cells, Hepatocytes, and Endothelial Cells Imprint the Kupffer Hall, D., Poussin, C., Velagapudi, V.R., Empsen, C., Joffraud, M., Beckmann,
Cell Identity on Monocytes Colonizing the Liver Macrophage Niche. J.S., Geerts, A.E., Ravussin, Y., Ibberson, M., Oresic, M., and Thorens, B.
Immunity 51, 638–654.e9. (2010). Peroxisomal and microsomal lipid pathways associated with resis-
tance to hepatic steatosis and reduced pro-inflammatory state. J. Biol.
Buch, T., Heppner, F.L., Tertilt, C., Heinen, T.J., Kremer, M., Wunderlich, F.T.,
Chem. 285, 31011–31023.
Jung, S., and Waisman, A. (2005). A Cre-inducible diphtheria toxin receptor
mediates cell lineage ablation after toxin administration. Nat. Methods 2, Halpern, K.B., Shenhav, R., Massalha, H., Toth, B., Egozi, A., Massasa, E.E.,
419–426. Medgalia, C., David, E., Giladi, A., Moor, A.E., et al. (2018). Paired-cell
sequencing enables spatial gene expression mapping of liver endothelial cells.
Buenrostro, J.D., Giresi, P.G., Zaba, L.C., Chang, H.Y., and Greenleaf, W.J.
Nat. Biotechnol. 36, 962–970.
(2013). Transposition of native chromatin for fast and sensitive epigenomic
profiling of open chromatin, DNA-binding proteins and nucleosome position. Hardy, T., Oakley, F., Anstee, Q.M., and Day, C.P. (2016). Nonalcoholic Fatty
Nat. Methods 10, 1213–1218. Liver Disease: Pathogenesis and Disease Spectrum. Annu. Rev. Pathol. 11,
Creyghton, M.P., Cheng, A.W., Welstead, G.G., Kooistra, T., Carey, B.W., 451–496.
Steine, E.J., Hanna, J., Lodato, M.A., Frampton, G.M., Sharp, P.A., et al. Heinz, S., Benner, C., Spann, N., Bertolino, E., Lin, Y.C., Laslo, P., Cheng, J.X.,
(2010). Histone H3K27ac separates active from poised enhancers and Murre, C., Singh, H., and Glass, C.K. (2010). Simple combinations of lineage-
predicts developmental state. Proc. Natl. Acad. Sci. USA 107, determining transcription factors prime cis-regulatory elements required for
21931–21936. macrophage and B cell identities. Mol. Cell 38, 576–589.
De Schepper, S., Verheijden, S., Aguilera-Lizarraga, J., Viola, M.F., Boesmans, Heinz, S., Romanoski, C.E., Benner, C., and Glass, C.K. (2015). The selection
W., Stakenborg, N., Voytyuk, I., Schmidt, I., Boeckx, B., Dierckx de Casterle, I., and function of cell type-specific enhancers. Nat. Rev. Mol. Cell Biol. 16,
et al. (2018). Self-Maintaining Gut Macrophages Are Essential for Intestinal 144–154.
Homeostasis. Cell 175, 400–415.e13.
Heinz, S., Texari, L., Hayes, M.G.B., Urbanowski, M., Chang, M.W., Givarkes,
Dick, S.A., Macklin, J.A., Nejat, S., Momen, A., Clemente-Casares, X., N., Rialdi, A., White, K.M., Albrecht, R.A., Pache, L., et al. (2018).
Althagafi, M.G., Chen, J., Kantores, C., Hosseinzadeh, S., Aronoff, L., et al. Transcription Elongation Can Affect Genome 3D Structure. Cell 174, 1522–
(2019). Self-renewing resident cardiac macrophages limit adverse remodeling 1536.e22.
following myocardial infarction. Nat. Immunol. 20, 29–39.
Heymann, F., and Tacke, F. (2016). Immunology in the liver–from homeostasis
Dobin, A., Davis, C.A., Schlesinger, F., Drenkow, J., Zaleski, C., Jha, S., Batut, to disease. Nat. Rev. Gastroenterol. Hepatol. 13, 88–110.
P., Chaisson, M., and Gingeras, T.R. (2013). STAR: ultrafast universal RNA-seq
aligner. Bioinformatics 29, 15–21. Hill, D.A., Lim, H.W., Kim, Y.H., Ho, W.Y., Foong, Y.H., Nelson, V.L., Nguyen,
H.C.B., Chegireddy, K., Kim, J., Habertheuer, A., et al. (2018). Distinct macro-
Eichenfield, D.Z., Troutman, T.D., Link, V.M., Lam, M.T., Cho, H., Gosselin, D.,
phage populations direct inflammatory versus physiological changes in adi-
Spann, N.J., Lesch, H.P., Tao, J., Muto, J., et al. (2016). Tissue damage drives
pose tissue. Proc. Natl. Acad. Sci. USA 115, E5096–E5105.
co-localization of NF-kB, Smad3, and Nrf2 to direct Rev-erb sensitive wound
repair in mouse macrophages. eLife 5, 554–562. Holtman, I.R., Skola, D., and Glass, C.K. (2017). Transcriptional control of
microglia phenotypes in health and disease. J. Clin. Invest. 127,
Gautier, E.L., Shay, T., Miller, J., Greter, M., Jakubzick, C., Ivanov, S., Helft, J.,
3220–3229.
Chow, A., Elpek, K.G., Gordonov, S., et al.; Immunological Genome
Consortium (2012). Gene-expression profiles and transcriptional regulatory Jaitin, D.A., Adlung, L., Thaiss, C.A., Weiner, A., Li, B., Descamps, H.,
pathways that underlie the identity and diversity of mouse tissue macro- Lundgren, P., Bleriot, C., Liu, Z., Deczkowska, A., et al. (2019). Lipid-
phages. Nat. Immunol. 13, 1118–1128. Associated Macrophages Control Metabolic Homeostasis in a Trem2-
Gerner, M.Y., Kastenmuller, W., Ifrim, I., Kabat, J., and Germain, R.N. (2012). Dependent Manner. Cell 178, 686–698.e14.
Histo-cytometry: a method for highly multiplex quantitative tissue imaging Ju, C., and Tacke, F. (2016). Hepatic macrophages in homeostasis and liver
analysis applied to dendritic cell subset microanatomy in lymph nodes. diseases: from pathogenesis to novel therapeutic strategies. Cell. Mol.
Immunity 37, 364–376. Immunol. 13, 316–327.

1072 Immunity 52, 1057–1074, June 16, 2020


ll
Article
Karlmark, K.R., Weiskirchen, R., Zimmermann, H.W., Gassler, N., Ginhoux, F., Resolution of Pro-inflammatory TLR4 Signaling by Reprogramming Fatty
Weber, C., Merad, M., Luedde, T., Trautwein, C., and Tacke, F. (2009). Hepatic Acid Metabolism. Cell Metab. 25, 412–427.
recruitment of the inflammatory Gr1+ monocyte subset upon liver injury pro- Peet, D.J., Janowski, B.A., and Mangelsdorf, D.J. (1998). The LXRs: a new
motes hepatic fibrosis. Hepatology 50, 261–274. class of oxysterol receptors. Curr. Opin. Genet. Dev. 8, 571–575.
Kent, W.J., Sugnet, C.W., Furey, T.S., Roskin, K.M., Pringle, T.H., Zahler, A.M., Perdiguero, E.G., and Geissmann, F. (2016). The development and mainte-
and Haussler, D. (2002). The human genome browser at UCSC. Genome Res. nance of resident macrophages. Nat. Immunol. 17, 2–8.
12, 996–1006.
Pirzgalska, R.M., and Domingos, A.I. (2018). Macrophages in obesity. Cell.
Kiss, M., Van Gassen, S., Movahedi, K., Saeys, Y., and Laoui, D. (2018). Immunol. 330, 183–187.
Myeloid cell heterogeneity in cancer: not a single cell alike. Cell. Immunol.
330, 188–201. Poussin, C., Ibberson, M., Hall, D., Ding, J., Soto, J., Abel, E.D., and Thorens,
B. (2011). Oxidative phosphorylation flexibility in the liver of mice resistant to
Kleiner, D.E., Brunt, E.M., Van Natta, M., Behling, C., Contos, M.J., Cummings,
high-fat diet-induced hepatic steatosis. Diabetes 60, 2216–2224.
O.W., Ferrell, L.D., Liu, Y.C., Torbenson, M.S., Unalp-Arida, A., et al.;
Nonalcoholic Steatohepatitis Clinical Research Network (2005). Design and Ramachandran, P., Dobie, R., Wilson-Kanamori, J.R., Dora, E.F., Henderson,
validation of a histological scoring system for nonalcoholic fatty liver disease. B.E.P., Luu, N.T., Portman, J.R., Matchett, K.P., Brice, M., Marwick, J.A., et al.
Hepatology 41, 1313–1321. (2019). Resolving the fibrotic niche of human liver cirrhosis at single-cell level.
Nature 575, 512–518.
Krenkel, O., Puengel, T., Govaere, O., Abdallah, A.T., Mossanen, J.C.,
Kohlhepp, M., Liepelt, A., Lefebvre, E., Luedde, T., Hellerbrand, C., et al. Rinella, M.E., and Sanyal, A.J. (2016). Management of NAFLD: a stage-based
(2018). Therapeutic inhibition of inflammatory monocyte recruitment reduces approach. Nat. Rev. Gastroenterol. Hepatol. 13, 196–205.
steatohepatitis and liver fibrosis. Hepatology 67, 1270–1283. Sakai, M., Troutman, T.D., Seidman, J.S., Ouyang, Z., Spann, N.J., Abe, Y.,
Langmead, B., and Salzberg, S.L. (2012). Fast gapped-read alignment with Ego, K.M., Bruni, C.M., Deng, Z., Schlachetzki, J.C.M., et al. (2019). Liver-
Bowtie 2. Nat. Methods 9, 357–359. Derived Signals Sequentially Reprogram Myeloid Enhancers to Initiate and
Maintain Kupffer Cell Identity. Immunity 51, 655–670.e8.
Lavin, Y., Winter, D., Blecher-Gonen, R., David, E., Keren-Shaul, H., Merad,
M., Jung, S., and Amit, I. (2014). Tissue-resident macrophage enhancer land- Satija, R., Farrell, J.A., Gennert, D., Schier, A.F., and Regev, A. (2015). Spatial
scapes are shaped by the local microenvironment. Cell 159, 1312–1326. reconstruction of single-cell gene expression data. Nat. Biotechnol. 33,
495–502.
Li, Q., Brown, J.B., Huang, H., and Bickel, J. (2011). Measuring reproducibility
of high-throughput experiments. The Annals of Applied Statistics 5, Schulz, C., Gomez Perdiguero, E., Chorro, L., Szabo-Rogers, H., Cagnard, N.,
1752–1779. Kierdorf, K., Prinz, M., Wu, B., Jacobsen, S.E.W., Pollard, J.W., et al. (2012). A
lineage of myeloid cells independent of Myb and hematopoietic stem cells.
Link, V.M., Duttke, S.H., Chun, H.B., Holtman, I.R., Westin, E., Hoeksema,
Science 336, 86–90.
M.A., Abe, Y., Skola, D., Romanoski, C.E., Tao, J., et al. (2018). Analysis of
Genetically Diverse Macrophages Reveals Local and Domain-wide Scott, C.L., T’Jonck, W., Martens, L., Todorov, H., Sichien, D., Soen, B.,
Mechanisms that Control Transcription Factor Binding and Function. Cell Bonnardel, J., De Prijck, S., Vandamme, N., Cannoodt, R., et al. (2018). The
173, 1796–1809.e17. Transcription Factor ZEB2 Is Required to Maintain the Tissue-Specific
Love, M.I., Huber, W., and Anders, S. (2014). Moderated estimation of fold Identities of Macrophages. Immunity 49, 312–325.e5.
change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550. Scott, C.L., Zheng, F., De Baetselier, P., Martens, L., Saeys, Y., De Prijck, S.,
Machado, M.V., and Diehl, A.M. (2016). Pathogenesis of Nonalcoholic Lippens, S., Abels, C., Schoonooghe, S., Raes, G., et al. (2016). Bone marrow-
Steatohepatitis. Gastroenterology 150, 1769–1777. derived monocytes give rise to self-renewing and fully differentiated Kupffer
cells. Nat. Commun. 7, 10321.
€nther, P., Crozet, L.,
Mass, E., Ballesteros, I., Farlik, M., Halbritter, F., Gu
Jacome-Galarza, C.E., Ha €ndler, K., Klughammer, J., Kobayashi, Y., et al. Seki, E., De Minicis, S., Osterreicher, C.H., Kluwe, J., Osawa, Y., Brenner, D.A.,
(2016). Specification of tissue-resident macrophages during organogenesis. and Schwabe, R.F. (2007). TLR4 enhances TGF-beta signaling and hepatic
Science 353, https://doi.org/10.1126/science.aaf4238. fibrosis. Nat. Med. 13, 1324–1332.

Matsumoto, M., Hada, N., Sakamaki, Y., Uno, A., Shiga, T., Tanaka, C., Ito, T., Seki, E., De Minicis, S., Gwak, G.-Y., Kluwe, J., Inokuchi, S., Bursill, C.A.,
Katsume, A., and Sudoh, M. (2013). An improved mouse model that rapidly de- Llovet, J.M., Brenner, D.A., and Schwabe, R.F. (2009a). CCR1 and CCR5 pro-
velops fibrosis in non-alcoholic steatohepatitis. Int. J. Exp. Pathol. 94, 93–103. mote hepatic fibrosis in mice. J. Clin. Invest. 119, 1858–1870.

McGrath, K.E., Frame, J.M., Fegan, K.H., Bowen, J.R., Conway, S.J., Seki, E., de Minicis, S., Inokuchi, S., Taura, K., Miyai, K., van Rooijen, N.,
Catherman, S.C., Kingsley, P.D., Koniski, A.D., and Palis, J. (2015). Distinct Schwabe, R.F., and Brenner, D.A. (2009b). CCR2 promotes hepatic fibrosis
Sources of Hematopoietic Progenitors Emerge before HSCs and Provide in mice. Hepatology 50, 185–197.
Functional Blood Cells in the Mammalian Embryo. Cell Rep. 11, 1892–1904. Shaw, T.N., Houston, S.A., Wemyss, K., Bridgeman, H.M., Barbera, T.A.,
Mederacke, I., Dapito, D.H., Affò, S., Uchinami, H., and Schwabe, R.F. (2015). Zangerle-Murray, T., Strangward, P., Ridley, A.J.L., Wang, P.,
High-yield and high-purity isolation of hepatic stellate cells from normal and Tamoutounour, S., et al. (2018). Tissue-resident macrophages in the intestine
fibrotic mouse livers. Nat. Protoc. 10, 305–315. are long lived and defined by Tim-4 and CD4 expression. J. Exp. Med. 215,
1507–1518.
Mitchell, C., Couton, D., Couty, J.-P., Anson, M., Crain, A.-M., Bizet, V., Rénia,
L., Pol, S., Mallet, V., and Gilgenkrantz, H. (2009). Dual role of CCR2 in the Shook, B.A., Wasko, R.R., Rivera-Gonzalez, G.C., Salazar-Gatzimas, E.,
constitution and the resolution of liver fibrosis in mice. Am. J. Pathol. 174, López-Giráldez, F., Dash, B.C., Muñoz-Rojas, A.R., Aultman, K.D., Zwick,
1766–1775. R.K., Lei, V., et al. (2018). Myofibroblast proliferation and heterogeneity are
Muse, E.D., Yu, S., Edillor, C.R., Tao, J., Spann, N.J., Troutman, T.D., supported by macrophages during skin repair. Science 362, https://doi.org/
Seidman, J.S., Henke, A., Roland, J.T., Ozeki, K.A., et al. (2018). Cell-specific 10.1126/science.aar2971.
discrimination of desmosterol and desmosterol mimetics confers selective Tacke, F., and Zimmermann, H.W. (2014). Macrophage heterogeneity in liver
regulation of LXR and SREBP in macrophages. Proc. Natl. Acad. Sci. USA injury and fibrosis. J. Hepatol. 60, 1090–1096.
115, E4680–E4689. Theurl, I., Hilgendorf, I., Nairz, M., Tymoszuk, P., Haschka, D., Asshoff, M., He,
Musso, G., Cassader, M., and Gambino, R. (2016). Non-alcoholic steatohepa- S., Gerhardt, L.M.S., Holderried, T.A.W., Seifert, M., et al. (2016). On-demand
titis: emerging molecular targets and therapeutic strategies. Nat. Rev. Drug erythrocyte disposal and iron recycling requires transient macrophages in the
Discov. 15, 249–274. liver. Nat. Med. 22, 945–951.
Oishi, Y., Spann, N.J., Link, V.M., Muse, E.D., Strid, T., Edillor, C., Kolar, M.J., Tripathi, S., Pohl, M.O., Zhou, Y., Rodriguez-Frandsen, A., Wang, G., Stein,
Matsuzaka, T., Hayakawa, S., Tao, J., et al. (2017). SREBP1 Contributes to D.A., Moulton, H.M., DeJesus, P., Che, J., Mulder, L.C.F., et al. (2015).

Immunity 52, 1057–1074, June 16, 2020 1073


ll
Article
Meta- and Orthogonal Integration of Influenza ‘‘OMICs’’ Data Defines a Role origins and dynamics of monocytes and tissue macrophages under homeo-
for UBR4 in Virus Budding. Cell Host Microbe 18, 723–735. stasis. Immunity 38, 79–91.
Xiong, X., Kuang, H., Ansari, S., Liu, T., Gong, J., Wang, S., Zhao, X.Y., Ji, Y., Li,
C., Guo, L., et al. (2019). Landscape of Intercellular Crosstalk in Healthy and Younossi, Z.M., Blissett, D., Blissett, R., Henry, L., Stepanova, M., Younossi,
NASH Liver Revealed by Single-Cell Secretome Gene Analysis. Mol. Cell 75, Y., Racila, A., Hunt, S., and Beckerman, R. (2016). The economic and clinical
644–660.e5. burden of nonalcoholic fatty liver disease in the United States and Europe.
Yang, C., McDonald, J.G., Patel, A., Zhang, Y., Umetani, M., Xu, F., Westover, Hepatology 64, 1577–1586.
E.J., Covey, D.F., Mangelsdorf, D.J., Cohen, J.C., and Hobbs, H.H. (2006).
Sterol intermediates from cholesterol biosynthetic pathway as liver X receptor Zigmond, E., Samia-Grinberg, S., Pasmanik-Chor, M., Brazowski, E., Shibolet,
ligands. J. Biol. Chem. 281, 27816–27826. O., Halpern, Z., and Varol, C. (2014). Infiltrating monocyte-derived macro-
Yona, S., Kim, K.W., Wolf, Y., Mildner, A., Varol, D., Breker, M., Strauss-Ayali, phages and resident kupffer cells display different ontogeny and functions in
D., Viukov, S., Guilliams, M., Misharin, A., et al. (2013). Fate mapping reveals acute liver injury. J. Immunol. 193, 344–353.

1074 Immunity 52, 1057–1074, June 16, 2020


Cell Metabolism

Article

NanoSIMS Analysis of Intravascular


Lipolysis and Lipid Movement
across Capillaries and into Cardiomyocytes
Cuiwen He,1 Thomas A. Weston,1 Rachel S. Jung,1 Patrick Heizer,1 Mikael Larsson,1 Xuchen Hu,1 Christopher M. Allan,1
Peter Tontonoz,3,4 Karen Reue,2 Anne P. Beigneux,1 Michael Ploug,5,6 Andrea Holme,7 Matthew Kilburn,7
Paul Guagliardo,7 David A. Ford,8 Loren G. Fong,1 Stephen G. Young,1,2,9,10,* and Haibo Jiang1,7,9,*
1Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
2Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
3Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
4Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
5Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
6Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
7Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia
8Department of Biochemistry and Molecular Biology, Center for Cardiovascular Research, Saint Louis University, St. Louis, MO 63104, USA
9Senior author
10Lead Contact

*Correspondence: sgyoung@mednet.ucla.edu (S.G.Y.), haibo.jiang@uwa.edu.au (H.J.)


https://doi.org/10.1016/j.cmet.2018.03.017

SUMMARY et al., 2016). LPL is synthesized by parenchymal cells and


secreted into the interstitial spaces, where it is captured by gly-
The processing of triglyceride-rich lipoproteins cosylphosphatidylinositol-anchored high-density lipoprotein-
(TRLs) in capillaries provides lipids for vital tissues, binding protein 1 (GPIHBP1) on endothelial cells and transported
but our understanding of TRL metabolism is limited, to the lumen of capillaries (Beigneux et al., 2007; Davies et al.,
in part because TRL processing and lipid movement 2010). GPIHBP1 moves bidirectionally across endothelial cells
have never been visualized. To investigate the move- (Davies et al., 2012), with each excursion providing an opportu-
nity to pick up LPL and move it to the capillary lumen. In the
ment of TRL-derived lipids in the heart, mice were
setting of GPIHBP1 deficiency, LPL is stranded within the inter-
given an injection of [2H]triglyceride-enriched TRLs,
stitial spaces (Davies et al., 2010), where it is useless for intravas-
and the movement of 2H-labeled lipids across capil- cular TRL processing. GPIHBP1- and GPIHBP1-bound LPL are
laries and into cardiomyocytes was examined by located only on capillary endothelial cells and are absent from
NanoSIMS. TRL processing and lipid movement in endothelial cells of arterioles and venules (Beigneux et al.,
tissues were extremely rapid. Within 30 s, TRL- 2007; Davies et al., 2010).
derived lipids appeared in the subendothelial spaces Aside from shuttling LPL to the capillary lumen, GPIHBP1
and in the lipid droplets and mitochondria of cardio- plays two additional functions in intravascular lipolysis. First,
myocytes. Enrichment of 2H in capillary endothelial the GPIHBP1-LPL complex is crucial for the margination of
cells was not greater than in cardiomyocytes, TRLs along capillaries (Goulbourne et al., 2014), making it
implying that endothelial cells may not be a control possible for LPL-mediated TRL processing to proceed. Second,
the binding of LPL to GPIHBP1 stabilizes the structural integrity
point for lipid movement into cardiomyocytes.
of LPL’s hydrolase domain and preserves its enzymatic activity,
Remarkably, a deficiency of the putative fatty acid
even in the presence of physiologic inhibitors (e.g., ANGPTL4),
transport protein CD36, which is expressed highly and by doing so GPIHBP1 serves to focus LPL activity in capil-
in capillary endothelial cells, did not impede entry laries (Fong et al., 2016; Mysling et al., 2016a, 2016b).
of TRL-derived lipids into cardiomyocytes. Although recent studies have clarified functions of GPIHBP1
and LPL in TRL processing, other topics in the realm of intravas-
cular lipolysis have remained enigmatic. In particular, no one
INTRODUCTION understands the mechanisms by which the fatty acid products
of TRL processing move across endothelial cells and into paren-
The lipolytic processing of triglyceride-rich lipoproteins (TRLs) by chymal cells (Hagberg et al., 2013). An early electron microscopy
lipoprotein lipase (LPL) is the central event in plasma lipid meta- study (Scow et al., 1976) suggested that the products of lipolysis
bolism, providing lipid nutrients for adipose tissue, heart, skeletal move across endothelial cells in ‘‘channels,’’ but the existence of
muscle, and other tissues (Havel, 2010; Havel and Kane, 2001; such channels is questionable and has never been substanti-
Kane and Havel, 2001). The processing of TRLs has been stud- ated. In recent years, several groups have proposed that fatty
ied for decades (Havel and Gordon, 1960; Korn, 1955a, 1955b), acid movement across endothelial cells is regulated and requires
but only recently have key mechanisms fallen into place (Fong transporter proteins (e.g., CD36 and fatty acid transporters)

Cell Metabolism 27, 1055–1066, May 1, 2018 ª 2018 Elsevier Inc. 1055
(Hagberg et al., 2013). The movement of fatty acids across cap- were on the bottom of the ultracentrifuge tube when spun at
illaries has been proposed to involve fatty acid entry into the d = 1.006 g/mL). Quantitative lipidomics studies confirmed that
endothelial cell cytoplasm and formation of acyl-coenzyme A large percentages of the fatty acyl chains in the 2H-TRLs were
(acyl-CoA) derivatives (Hagberg et al., 2013). Also, endothelial deuterated (Table S1). Indeed, the majority of the linoleic acid
cells have been proposed to be a ‘‘control point’’ for regulating was deuterated. We observed consistent findings by NanoSIMS.
fatty acid movement, protecting parenchymal cells from exces- When the 2H-TRL particles were analyzed by NanoSIMS, the
sive lipid uptake (Hagberg et al., 2013). According to the latter 2
H/1H ratio in the larger TRLs was 3,000–6,000 times the natu-
view, one could easily imagine that TRL-derived lipids might ral abundance of 2H, such that about one-third of all hydrogens in
accumulate in endothelial cells before moving to parenchymal the lipoprotein particles were 2H (Figure S1).
cells. On the other hand, others have proposed that fatty acids The 2H-TRLs (40 mg triglycerides in a volume of 200 mL) were
simply diffuse freely across mammalian cells without assistance injected intravenously into wild-type mice. After 30 s, 2 min, or
from protein transporters (Guo et al., 2006; Hamilton et al., 2001; 30 min, the mice were euthanized, perfused extensively with
Xu et al., 2013). Another enigma has been the itinerary of TRL- PBS, and then perfusion-fixed with carbodiimide/glutaralde-
derived fatty acids after entering parenchymal cells. It remains hyde. (Carbodiimide is effective in fixing carboxylate-containing
controversial whether fatty acids can enter mitochondria directly small molecules in cells or tissues [Dallwig and Deitmer, 2002;
or whether they first must enter cytosolic triglyceride droplets. Takei et al., 2012; Tymianski et al., 1997], and our studies, in
Understanding how lipids move across capillaries and into which [3H]oleic acid was added to CHO-K1 cells, revealed
surrounding parenchymal cells represents an important issue 60%–75% greater [3H]oleic acid fixation with carbodiimide/
in lipoprotein physiology, but thus far the insights into this glutaraldehyde than with glutaraldehyde alone.) After fixation,
process have been limited and have depended on cell culture the left ventricular apex was embedded in epoxy resin,
models or indirect evidence (e.g., drawing inferences from tran- sectioned, and analyzed by NanoSIMS. Images from 12C14N–
script levels for putative lipid-transport proteins or measuring ions (a cluster secondary ion that reveals tissue nitrogen content)
lipid stores within tissue extracts) (Bharadwaj et al., 2010; were useful for visualizing tissue morphology; composite
12 14 –
Hagberg et al., 2010, 2013; Jang et al., 2016). We suspected C N and 2H/1H images identified regions of 2H enrichment
that it would be possible to gain additional insights into TRL pro- (Figure 1). In the composite images, the 2H/1H scale ranges
cessing and lipid movement by imaging TRL processing within from 0.0002 to 0.01 (from slightly above 2H natural abundance
capillaries. to 70 times natural abundance). Margination of 2H-TRLs along
In the current studies, we used nanoscale secondary ion mass the luminal surface of capillary endothelial cells was detectable
spectrometry (NanoSIMS) to visualize both TRL processing in 30 s and 2 min after the injection of 2H-TRLs (Figures 1A and
heart capillaries and the fate of TRL-derived lipids in adjacent 1B, white arrows), but ‘‘marginated TRLs’’ were not encountered
cardiomyocytes. NanoSIMS imaging uses a Cs+ beam to in capillaries at the 30-min time point. The 2H/1H ratio in the
bombard a tissue section, releasing secondary ions that can marginated lipoproteins at 30 s and 2 min ranged from 15 to
be analyzed by mass spectrometry and used to create images 450 times natural abundance, suggesting that processing had
based solely on the isotopic content of the tissue (He et al., removed some of the 2H-labeled triglycerides from the particles.
2017a, 2017b; Jiang et al., 2014a, 2014b). After giving mice an Enrichment of 2H within cardiomyocyte lipid droplets (40–50
injection of TRLs enriched in [2H]triglycerides, NanoSIMS imag- times natural abundance) was observed as early as 30 s after
ing makes it possible to visualize the 2H-labeled products of TRL injecting 2H-TRLs (Figure 1, yellow arrows).
processing in endothelial cells, the subendothelial spaces, the The margination of TRLs along capillaries is mediated by the
mitochondria, and/or cytosolic lipid droplets of cardiomyocytes. LPL-GPIHBP1 complex (Goulbourne et al., 2014); thus, there is
In NanoSIMS imaging studies, all of the secondary ion data are little doubt that the marginated TRLs in Figure 1 were undergoing
available for quantitative analyses. In the current studies, we processing by LPL. Interestingly, we were unable to document
show that the movement of TRL-derived lipids across endothe- streaming of 2H-labeled lipids away from marginated TRLs and
lial cells and into cytosolic lipid droplets and mitochondria of into endothelial cells or cardiomyocytes (Figure 2). When we
cardiomyocytes is extraordinarily rapid, occurring within sec- quantified the 2H content of marginated TRLs and the 2H content
onds. We found no evidence that capillary endothelial cells are of immediately adjacent regions of endothelial cells and cardio-
a substantial barrier to the movement of TRL-derived lipids myocytes, we observed a ‘‘fall-off’’ in 2H enrichment with
into cardiomyocytes. Also, we found no evidence that CD36 increasing distance from the TRL (Figure 2). One might have
deficiency impedes the entry of TRL-derived lipids into easily jumped to the conclusion that the fall-off in 2H enrichment
cardiomyocytes. represented streaming of fatty acids away from the TRL, but we
do not believe that this was the case, simply because we
RESULTS observed a similar fall-off in 2H enrichment with increasing dis-
tance into the capillary lumen. In our tissue sections, the capillary
The Lipolytic Processing of TRLs and Lipid Movement lumen is filled with epoxy resin, which is rich in 12C but devoid of
14
into Cardiomyocytes Are Rapid N. We suspect that the appearance of 2H enrichment in the
2
H-Enriched TRLs (2H-TRLs) were harvested from the plasma of lumen of heart capillaries (which had been thoroughly perfused
Gpihbp1 / mice after administering 2H-labeled fatty acids by before fixation) was the result of short-distance diffusion of tri-
gastric gavage. The 2H content of TRLs harvested from glycerides into the resin-filled capillary lumen during tissue pro-
Gpihbp1 / mice was quite high, as judged by the fact that their cessing. Because of this observation, we do not believe that we
density exceeded 1.006 g/mL (i.e., virtually all of the 2H-TRLs observed bona fide streaming of fatty acids away from TRLs and

1056 Cell Metabolism 27, 1055–1066, May 1, 2018


Figure 1. NanoSIMS Images of TRLs along
Heart Capillary Endothelial Cells
Four-month-old wild-type female mice were fas-
ted for 4 hr and then injected with 200 mL 2H-TRLs
containing 40 mg triglycerides (produced in
Gpihbp1 / mice after administering 2H-labeled
fatty acids by gavage). After 30 s, 2 min, or 30 min,
mice were euthanized and perfusion-fixed with
carbodiimide/glutaraldehyde; sections were pre-
pared for NanoSIMS imaging. Three mice were
used in this experiment, one for each time point.
(A) 12C14N– NanoSIMS images were generated to
visualize tissue morphology; composite 12C14N–
(blue) and 2H/1H ratio (red) images revealed
2
H-TRLs that had marginated along capillary
endothelial cells (white arrows) and 2H-enriched
cytosolic lipid droplets in cardiomyocytes (yellow
arrows). The scale shows the 2H/1H ratio multiplied
by 10,000. The 2H natural abundance, relative to
1
H, is 0.000156. Scale bars, 6 mm.
(B) The boxed areas in (A) images are shown at
higher magnification to reveal capillary endothelial
cells. Composite 12C14N– (gray) and 2H/1H ratio
(red) images show marginated lipoproteins (ar-
rows) in the capillary lumen and 2H-enriched
cytosolic lipid droplets in cardiomyocytes. Scale
bars, 2 mm. See also Figure S1.

into endothelial cells or cardiomyocytes. We suspect that the reduced TRL processing (Figure 3B). In both wild-type and
streaming of fatty acids away from TRLs, which obviously must Gpihbp1 / mice, the 2H/1H ratio in endothelial cells was similar
occur physiologically, is so rapid that it was not detectable by to the 2H/1H ratio in surrounding cardiomyocytes (Figure 3B).
NanoSIMS at the 30-s or 2-min time points. In support of that
reasoning, we never observed higher levels of 2H enrichment in Investigating the Fate of TRL-Derived Lipids within
segments of cardiomyocytes that were immediately adjacent Cardiomyocytes
to capillaries (Figure 1), implying that movement of 2H-labeled The mitochondria of cardiomyocytes could be identified in back-
lipids into and across cardiomyocytes is quite rapid. scattered electron (BSE) images and in 12C14N–, 16O–, and 31P–
NanoSIMS images (Figure 4A). Visualizing mitochondria with
The Entry of 2H-TRL-Derived Lipids into Cardiomyocytes 12 14 –
C N images was particularly helpful because the NanoSIMS
Depends on Intravascular Processing of 2H-TRLs by LPL 50L instrument is capable of recording 12C14N–, 2H–, and 1H– ions
To be confident that the rapid appearance of 2H in cytosolic lipid simultaneously. In Figure 4B, we show high-resolution 12C14N–
droplets of cardiomyocytes (Figure 1) was secondary to LPL- and 2H/1H NanoSIMS images of the heart after injecting
mediated 2H-TRL processing, we performed experiments in 2
H-TRLs. The 2H/1H scale in these images differs from those in
isolated, perfused mouse hearts, which allowed us to compare Figure 1, ranging from slightly above 2H natural abundance to
2
H entry into cardiomyocytes in the presence or absence of an 7 times natural abundance. When the mice were killed 30 s
LPL inhibitor. We also examined 2H uptake into hearts of or 2 min after the injection of 2H-TRLs, marginated TRLs could
Gpihbp1 / mice (where LPL is absent from the luminal surface be identified in the capillary lumen, and there was 2H enrichment
of capillaries) (Davies et al., 2010, 2012). In perfused hearts of in both mitochondria and cytosolic lipid droplets of cardiomyo-
wild-type mice treated with a nonspecific lipase inhibitor (tetra- cytes (Figure 4C). At the 30-min time point, we did not find
hydrolipistatin), the appearance of 2H into cardiomyocytes was marginated TRLs in capillaries; 2H enrichment persisted in cardi-
lower than in the absence of the inhibitor (Figures 3A and 3B). omyocyte mitochondria and lipid droplets, but the amount of 2H
Enrichment of 2H in cardiomyocytes was also lower in the setting enrichment in mitochondria was lower in the 30-min images than
of Gpihbp1 deficiency (Figures 3A and 3B). In wild-type hearts in the 30-s or 2-min images (Figures 4C and 4D), almost certainly
that had been treated with the LPL inhibitor, numerous reflecting mitochondrial catabolism of 2H-labeled fatty acids in
2
H-TRLs were found along endothelial cells, reflecting an accu- the 30-min time span after the injection of 2H-TRLs. We found
mulation of marginated but unprocessed 2H-TRLs in capillaries no evidence, at any of the time points, that 2H enrichment in
(Figure 3A). As expected, 2H enrichment was lower in endothelial endothelial cells was greater than in adjacent cardiomyocytes
cells of Gpihbp1 / hearts (Figure 3), where TRL margination (Figures 1, 2, and 3; data not shown), implying that TRL-derived
does not occur (Goulbourne et al., 2014). When we measured lipids do not accumulate in endothelial cells before moving into
the 2H/1H ratio in endothelial cells of wild-type mice (focusing parenchymal cells.
exclusively on segments devoid of marginated TRLs) with the In some experiments, 13C-labeled fatty acids were given to
2
H/1H ratio in endothelial cells of Gpihbp1 / mice, we found mice by gastric gavage for several days before administering
a lower 2H/1H ratio in Gpihbp1 / endothelial cells, reflecting the intravenous injection of 2H-TRLs. In these studies, we once

Cell Metabolism 27, 1055–1066, May 1, 2018 1057


Figure 2. Movement of Lipids from 2H-TRLs to Surrounding Cardiomyocytes
Four-month-old female wild-type mice were fasted for 4 hr and then given an intravenous injection of 200 mL of 2H-TRLs containing 40 mg triglycerides. After 30 s
or 2 min, mice were euthanized and perfusion-fixed with carbodiimide/glutaraldehyde; tissue sections were prepared for NanoSIMS. Images from three regions
(I, II, III) of the left ventricular apex were recorded. Two mice were used in this experiment.
(A) 12C14N– NanoSIMS images to show tissue morphology. Section I was prepared from tissue that had been stained with imidazole.
(B and C) Composite 12C14N–, blue in (B) and gray in (C), and 2H/1H ratio (red) images were generated to visualize 2H-enriched TRLs in the capillary lumen and
2
H-enriched lipid droplets within cardiomyocytes. The boxed area of the composite image is shown at higher magnification in (C). 12C14N– ions and the 2H/1H ratio
were measured in a TRL particle (purple circle) along the luminal surface of a capillary endothelial cell and in regions (white circles) extending into the endothelial
cell (e-1, e-2), an adjacent cardiomyocyte (c), or into the capillary lumen (l). Scale bars, 2 mm.
(D) Graph showing a progressive decrease in the 2H/1H ratio—starting with the TRL particle in the capillary lumen and extending into the endothelial cell, the
adjacent cardiomyocyte, and the capillary lumen.
(E) Graph showing 12C14N– ions in the TRL particle and in regions extending into the endothelial cell, the cardiomyocyte, and the capillary lumen.

again observed 2H-TRLs margination in endothelial cells and 2H was also enriched in 13C, but to a lesser degree than cytosolic
enrichment in cardiomyocyte lipid droplets 30 s after injecting lipid droplets (Figure S2).
2
H-TRLs (Figure 5A). 2H-Labeled lipids invariably entered cyto- The adult mouse brain primarily uses glucose for fuel, and TRL
solic lipid droplets that were already labeled with 13C (Figure 5A), margination is absent in the capillaries of the brain (Goulbourne
and the levels of 2H and 13C enrichment in lipid droplets were et al., 2014). For those reasons, we were skeptical that we would
positively correlated (p < 0.001) (Figure 5B). Of note, however, detect significant amounts of 13C or 2H enrichment in the brain af-
the percentages of the labels in cytosolic lipid droplets were ter administering 13C-labeled fatty acids or 2H-TRLs. Indeed, after
different. Approximately 50% of cardiomyocyte 2H was located administering 13C-labeled fatty acids by gastric gavage and sub-
in cytosolic lipid droplets, whereas only 5% of the 13C was in sequently administering 2H-TRLs intravenously, we were unable
lipid droplets (Figures 5C and 5D). This difference was not partic- to detect 13C or 2H enrichment in the cerebral cortex (Figure S3A).
ularly surprising because 13C-labeled fatty acids (administered a NanoSIMS instruments have extraordinarily accurate mass
day earlier) would be expected to contribute to membrane phos- resolution, making it nearly impossible to mistake one isotope
pholipids. Also, fatty acid metabolites are used for synthesis of for another. However, to be confident that we identified 2H–
nonessential amino acids (Sidossis et al., 1995). Indeed, the and 13C– ions correctly, we created NanoSIMS images from
fact that metabolites from 13C-labeled fatty acids contribute to the heart of a mouse that had not been given 13C-labeled fatty
protein synthesis was illustrated by NanoSIMS images of brown acids or 2H-TRLs. We had no difficulty in generating 12C14N–
adipose tissue. As expected, the cytosolic lipid droplets in brown NanoSIMS images of heart tissue, but as expected there was
adipocytes were enriched in 13C but devoid of nitrogen (i.e., as no enrichment in 13C or 2H (Figure S3B).
judged by the 12C14N– images) (Figure S2). Interestingly, the he-
moglobin in one erythrocyte (almost certainly a newly released Detecting 2H-Labeled Lipids in the Subendothelial
erythrocyte) was enriched in 13C, but 13C enrichment was negli- Spaces
gible in older blood cells (two leukocytes and one erythrocyte) The TRL-derived lipids that enter cardiomyocytes obviously
(Figure S2). The nitrogen-rich cytoplasm of brown adipocytes must have traversed the subendothelial spaces around

1058 Cell Metabolism 27, 1055–1066, May 1, 2018


Figure 3. The 2H-Enriched Lipids that Enter Cardiomyocytes Are Derived from LPL-Mediated Intravascular Lipolysis, as Judged by Studies in
Isolated, Perfused Hearts
Hearts from 4-month-old female wild-type or Gpihbp1 / mice were isolated and cannulated. Beating hearts were perfused with Tyrode’s buffer containing CaCl2
and either an LPL inhibitor (1 mM tetrahydrolipistatin) or vehicle (DMSO) alone. Next, hearts were perfused with 2H-TRLs. After 5 min, the hearts were perfused
with PBS (to remove nonmarginated TRLs) and then perfusion-fixed with carbodiimide/glutaraldehyde. Sections of hearts were prepared for NanoSIMS. Three
mice were used in this experiment, one for each time point.
(A) 12C14N– images were created to visualize morphology (left panels); composite 12C14N– (blue) and 2H/1H ratio (red) images (right panels) were used
to visualize 2H-TRLs in capillaries and 2H-enriched lipid droplets in cardiomyocytes. The scale shows the 2H/1H ratio multiplied by 10,000. Scale
bars, 2 mm.
(B) Bar graphs show 2H/1H ratios in cardiomyocytes, the sum of 2H/1H ratios in cytosolic lipid droplets, 2H/1H ratios in capillary endothelial cells, 2H/1H ratios in
regions of cardiomyocytes devoid of lipid droplets (LD), and 2H/1H ratios in regions of endothelial cells devoid of marginated lipoproteins (LP). In each graph, the y
axis starts at 0.00015 (natural abundance of 2H). *p < 0.05; **p < 0.01; ***p < 0.0005. Each graph represent quantification of four 30 3 30 mm images. Data are
presented as means ± SD and were analyzed with one-way ANOVA with multiple comparisons.

capillaries. After administering an intravenous injection of TRLs Staining of lipids by imidazole was also evident in NanoSIMS
and then staining heart tissue with OsO4 and imidazole, we images of cardiomyocytes from mice that had been given
13
observed irregular, darkly stained structures in the subendo- C-labeled fatty acids by gastric gavage (Figure S4B). Normally,
13
thelial spaces around capillaries. (Imidazole is a nitrogen-rich C-enriched cytosolic lipid droplets in cardiomyocytes are
mordant that increases OsO4 binding to lipids [Angermu €ller devoid of nitrogen and appear ‘‘black’’ in a 12C14N– NanoSIMS
and Fahimi, 1982].) The irregular, darkly stained structures image. When the same tissue was stained with imidazole, the
were frequent at the 30-s and 2-min time points but were cytosolic lipid droplets became nitrogen rich and were ‘‘white’’
uncommon at the 30-min time point (Figure 6A). To determine in a 12C14N– image (Figure S4B).
if the darkly stained subendothelial structures contained TRL- The irregular OsO4/imidazole-stained structures were less
derived lipids, we performed correlative BSE/NanoSIMS imag- common around larger blood vessels, where GPIHBP1 and
ing on OsO4/imidazole-stained heart tissue harvested 30 s after LPL are absent (Davies et al., 2010; Fong et al., 2016). In the
an intravenous injection of 2H-TRLs. BSE images revealed electron micrograph shown in Figure 6C, many irregular, darkly
irregular, darkly stained structures in the subendothelial stained structures were found in and around a capillary, but
spaces (Figures 6B and S4A); NanoSIMS imaging of the not in the vicinity of an arteriole (Figures 6C and S4C). By
same sections revealed that these structures were enriched NanoSIMS, we could not detect 2H enrichment in the subendo-
in 2H (Figures 6B and S4A) from 2H-TRL processing. Because thelial spaces surrounding arterioles, even though 2H-labeled
these structures were stained with imidazole, they were en- lipids from TRL processing were present in adjacent cardio-
riched in nitrogen, as judged by 12C14N– images (Figures 6B myocytes and in the smooth muscle cells of nearby arterioles
and S4A). (Figures 6D).

Cell Metabolism 27, 1055–1066, May 1, 2018 1059


Figure 4. NanoSIMS Imaging to Visualize Lipids Released from 2H-Enriched TRLs
Three-month-old female wild-type mice were fasted for 4 hr and then given an intravenous injection of 200 mL 2H-TRLs (40 mg triglycerides). After 30 s, 2 min, or
30 min, the mice were euthanized and perfusion-fixed with carbodiimide/glutaraldehyde. Tissue sections were prepared for NanoSIMS and backscattered
electron (BSE) imaging. Three mice were used in this experiment, one for each time point.
(A) NanoSIMS images (12C14N–, 16O–, 31P–) from tissue not stained with imidazole. A BSE image of the same region was useful for morphology. Several mito-
chondria are outlined in red.
(B) NanoSIMS images of heart sections 30 s, 2 min, or 30 min after the injection of 2H-TRLs. 12C14N– images to visualize cell and tissue morphology (top panels);
2
H/1H ratio images (bottom panels) to visualize the fate of lipids released from 2H-TRLs. LP, lipoproteins; L, capillary lumen; M, mitochondria; LD, lipid droplets.
The scale shows the 2H/1H ratio multiplied by 10,000.
(C) Bar graph showing the 2H/1H ratio in endothelial cells, mitochondria, myocytes, regions of myocytes lacking lipid droplets (LD), and lipid droplets at different
time points. Three 30 3 30 mm images (each containing a minimum of two to three cardiomyocytes) were analyzed at each time point (endothelial cells, n = 8–13;
mitochondria, n = 24–73; cardiomyocytes, n = 7–18; regions of myocytes lacking lipid droplets, n = 40; lipid droplets, n = 40–90). Data are presented as means ±
SD Differences were assessed with one-way ANOVA. *p < 0.05; **p < 0.01; ns, nonsignificant.
(D) Bar graph depicting 2H/1H in lipid droplets divided by 2H/1H in mitochondria at each time point (mean ± SD). Each graph represent data from two 10 3 10 mm
images and three 30 3 30 mm images. Data are presented as means ± SD and were analyzed with one-way ANOVA with multiple comparisons. **p < 0.01. See
also Figure S2.

Assessing the Impact of CD36 Deficiency on the Entry of 7A and 7C). When we administered 13C-labeled fatty acids to
TRL-Derived Lipids into Cardiomyocytes wild-type and Cd36 / mice by gastric gavage, we observed
NanoSIMS imaging opens the door to examining the functional consistent findings—similar amounts of 13C in wild-type and
relevance of specific proteins in the movement of TRL-derived Cd36 / cardiomyocytes (Figures 7D and 7E), but reduced
lipids into and across capillaries. CD36, a putative fatty acid amounts of 13C in cytosolic lipid droplets of Cd36 / cardiomyo-
transporter, is expressed at high levels in heart capillary endo- cytes (as a percentage of total 13C in cells) (Figures 7D and 7F). In
thelial cells (Greenwalt et al., 1995) (Figure S5) and has been addition to reduced amounts of total 13C in lipid droplets, the
13
hypothesized to play a role in the movement of fatty acids into C/12C ratio in lipid droplets was lower in Cd36 / cardiomyo-
cardiomyocytes (Bharadwaj et al., 2010; Hagberg et al., 2013). cytes (Figure 7G). Interestingly, the 13C/12C ratio was slightly
In the current study, we focused on testing whether a deficiency higher in Cd36 / mitochondria (Figure 7G). In another series
of CD36 might impede the movement of 2H-TRL-derived lipids of NanoSIMS studies, we once again found similar amounts of
into cardiomyocytes. Wild-type and Cd36 / mice were injected 13
C enrichment in wild-type and Cd36 / cardiomyocytes (Fig-
intravenously with equivalent amounts of 2H-TRLs, and heart ures S6A and S6B), but the percentage of total 13C in lipid drop-
sections were prepared for NanoSIMS imaging. At 1.5- and lets was lower in Cd36 / cardiomyocytes (Figure S6C). CD36
2-min time points, 2H enrichment in cardiomyocytes was equiv- deficiency has been reported to be associated with lower levels
alent in wild-type and Cd36 / mouse hearts (Figures 7A and of acyl-CoA in cells (Bharadwaj et al., 2010), but whether that
7B), but the amount of 2H in cytosolic lipid droplets was lower finding is due to the reduced amounts of 2H in cytosolic lipid
in Cd36 / hearts (as a percentage of total 2H in cells) (Figures droplets of Cd36 / mice is unknown. We considered the

1060 Cell Metabolism 27, 1055–1066, May 1, 2018


Figure 5. NanoSIMS Images of Heart Tissue from Mice that Had Been Given 13C-Labeled Fatty Acids by Gastric Gavage and on the Following
Day an Intravenous Injection of 2H-TRLs
13
C-Labeled fatty acids were administered to 3- to 4-month-old female wild-type mice by gastric gavage (two 80-mg doses 12 hr apart). At 22 hr after the second
dose, mice were fasted for 4 hr and then injected with 200 mL 2H-TRLs containing 40 mg triglycerides. After periods of 30 s or 2 min post-injection, the mice were
euthanized and perfusion-fixed with carbodiimide/glutaraldehyde; sections of heart were prepared for NanoSIMS imaging. Two mice were used in this exper-
iment, one for each time point.
(A) NanoSIMS images of heart tissues 30 s after the injection of 2H-TRLs. 12C14N– images were useful for morphology; 2H/1H ratio images revealed marginated
2
H-TRLs in capillaries (white arrows) and 2H-enriched lipids in lipid droplets of cardiomyocytes (boxed regions). 13C/12C ratio images show 13C distribution. 2H/1H
and 13C/12C ratio scales are multiplied by 10,000. Scale bars, 2 mm.
(B) Scatterplots comparing 2H/1H and 13C/12C ratios in the center of cardiomyocyte lipid droplets. Data were obtained from lipid droplets (n = 50–60) in two
NanoSIMS images (31 3 31 mm).
(C) NanoSIMS images showing 2H enrichment in marginated lipoproteins in capillaries (white arrows) and cardiomyocyte lipid droplets (several indicated by
yellow arrows). 12C14N– images show tissue morphology, 2H/1H ratio images show the distribution of 2H 30 s or 2 min after the injection of 2H-TRLs, and 13C/12C
ratio images show 13C distribution 1 day after giving 13C-labeled fatty acids by gastric gavage. The same lipid droplets were identified in both 2H/1H and 13C/12C
images. 2H/1H and 13C/12C ratio scales are multiplied by 10,000. Scale bars, 5 mm.
(D) Plot depicting the percentage of cellular 2H and 13C in cytosolic lipid droplets, relative to 1H and 12C, respectively (two 35 3 35 mm NanoSIMS images analyzed
at each time point). See also Figure S3.

possibility that the lipid droplets in Cd36 / cardiomyocytes DISCUSSION


were simply too small to be visualized by NanoSIMS (where
the lateral resolution is 40–50 nm). By electron microscopy, We used NanoSIMS imaging along with electron microscopy to
the average area of cytosolic lipid droplets in Cd36 / cardio- visualize intravascular TRL processing and the fate of TRL-
myocytes was lower than in wild-type cardiomyocytes (Fig- derived lipids in cardiomyocytes. We gleaned three insights
ure S7), but this difference was small, probably too small to from our studies. First, TRL processing and the uptake of TRL
explain the paucity of lipid droplets in the NanoSIMS images of lipids by cardiomyocytes are extremely rapid—occurring within
Cd36 / cardiomyocytes. seconds after TRLs enter the bloodstream. Second, during

Cell Metabolism 27, 1055–1066, May 1, 2018 1061


Figure 6. The Lipid Products of TRL Processing Can Be Identified in the Subendothelial Spaces
(A) Transmission electron micrographs of the left ventricular apex from 4-month-old female wild-type mice 30 s, 2 min, or 30 min after an intravenous injection of
200 mL TRLs. Irregular, darkly stained structures were observed in the subendothelial spaces (red arrows) in OsO4/imidazole-stained tissue; these structures were
more common at the 30-s and 2-min time points. Scale bars, 1 mm. 2H/1H ratio scales were multiplied by 10,000. Three mice were used in this experiment, one for
each time point.
(B) Visualizing irregular, darkly stained structures in the subendothelial spaces by NanoSIMS and BSE imaging. A 4-month-old female wild-type mouse was
fasted for 4 hr, injected with 200 mL 2H-TRLs, and killed 30 s later. Heart tissue was stained with OsO4/imidazole and processed for NanoSIMS and BSE imaging.
The irregular, darkly stained subendothelial structures (white arrows) in the BSE images (left panel) were enriched in nitrogen (12C14N– ions, middle panels),
reflecting the high nitrogen content of imidazole, and were enriched in 2H, as judged by composite 12C14N– (blue) and 2H/1H ratio (red) images (right panels).
(C) The irregular, darkly stained subendothelial structures were found in the subendothelial spaces around capillaries (a), but not in the vicinity of a large blood
vessel (b and c).
(D) NanoSIMS images of an arteriole and an adjacent cardiomyocyte 2 min after an injection of 2H-TRLs. Enrichment of 2H was observed in mitochondria and
cytosolic lipid droplets of cardiomyocytes and in the mitochondria of arteriolar smooth muscle cells, but 2H enrichment was not observed in the interstitial spaces
around the arteriole. See also Figure S4.

active TRL processing, we observed no evidence that 2H-TRL- to capillaries. Also, 2H rapidly entered arteriolar smooth muscle
derived lipids accumulate to higher levels in capillary endothelial cells, despite the fact that TRL processing in arterioles is almost
cells than in adjacent cardiomyocytes. Third, we found no evi- certainly negligible, owing to an absence of GPIHBP1 and LPL
dence that CD36 deficiency substantially impedes entry of (Davies et al., 2010). All of these observations appear to be
TRL-derived lipids into cardiomyocytes. consistent with the proposal that fatty acids diffuse rapidly
For decades, the lipid metabolism field has recognized across cells (Guo et al., 2006; Hamilton et al., 2001; Xu et al.,
that the half-life of TRLs is measured in minutes (Havel and 2013) or, alternatively, that there is a yet-to-be-identified abun-
Kane, 2001), but we were nevertheless quite surprised by how dant high-capacity transport system for fatty acid movement
rapidly TRL-derived lipids traverse capillary endothelial cells across endothelial cells. Before reaching cardiomyocytes, the
and enter cardiomyocytes. NanoSIMS imaging showed that products of lipolysis obviously must traverse subendothelial
2
H-TRL-derived lipids appear in cardiomyocytes within seconds spaces. In imidazole-stained heart tissue, we observed darkly
(preferentially in cytosolic lipid droplets but also in mitochondria). staining structures in the subendothelial spaces, and those
Other features of the NanoSIMS images were consistent with structures contained 2H-labeled lipids from 2H-TRL processing.
extremely rapid movement of fatty acids into and across tissues. An attractive feature of NanoSIMS analysis is that the second-
For example, we never observed more 2H enrichment in capillary ary ion data are available for quantitative analyses, pixel by pixel,
endothelial cells than in cardiomyocytes, nor did we observe with >260,000 pixels/image. In our studies, quantitative analyses
greater 2H enrichment in regions of cardiomyocytes adjacent revealed that an LPL inhibitor drug or a deficiency of GPIHBP1 is

1062 Cell Metabolism 27, 1055–1066, May 1, 2018


Figure 7. TRL-Derived Lipids Move across Endothelial Cells and Enter Cardiomyocytes in Both Wild-Type and Cd36-Deficient Mice
NanoSIMS analyses of 2H– ions in the heart after administering an intravenous injection of 200 mL 2H-TRLs to 4-month-old female wild-type (Cd36+/+) and Cd36 /
mice. At 1.5 or 2 min after the injection, hearts were harvested and sections were prepared for NanoSIMS. Differences in quantitative NanoSIMS data were
analyzed with a Student’s t test with Welch’s correction. Four mice were used in this experiment, one for each time point.
(A) 12C14N– images (left) were used to define tissue morphology; the 2H/1H ratio images (middle) and the 2H/1H hue saturation images (right) show sites of
2
H enrichment. White arrows indicate examples of cytosolic lipid droplets. Ratio scales were multiplied by 10,000. Scale bars, 10 mm.
(B) 2H/1H ratios in cardiomyocytes of Cd36+/+ and Cd36 / mice. Each data point represents one cardiomyocyte in two or three 30 3 30 mm NanoSIMS images.
*p < 0.05; ns, nonsignificant.
(C) 2H in cytosolic lipid droplets as a percentage of total 2H in cardiomyocytes. Graphs were generated by quantifying two or three 30 3 30 mm NanoSIMS images
for each time point. NanoSIMS analyses of 13C– ions in the heart after feeding 13C-labeled fatty acids by gavage (three 80-mg doses 12 hr apart). Twenty-four
hours after the last dose, mice were fasted for 4 hr, and hearts were harvested and prepared for NanoSIMS. *p < 0.05; ns, nonsignificant.
(D) 12C14N– NanoSIMS images were used to visualize morphology; 31P images were used to visualize mitochondria; 13C/12C ratio images show sites of
13
C enrichment. Arrows indicate examples of cytosolic lipid droplets. Ratio scales were multiplied by 10,000. Scale bars, 10 mm.
(legend continued on next page)
Cell Metabolism 27, 1055–1066, May 1, 2018 1063
accompanied by reduced uptake of 2H-TRL-derived lipids into changes in the distribution of 2H-TRL-derived lipids within
cardiomyocytes. Quantitative analyses also allowed us to quan- cardiomyocytes.
tify the distribution of 2H-TRL-derived lipids within cardiomyo- NanoSIMS analyses are particularly helpful when, as in the
cytes. Approximately 50% of the 2H in cardiomyocytes was current study, there are anatomical questions to address (i.e.,
located in cytosolic lipid droplets 30 s or 2 min after an injection an interest in defining the distribution of an isotope in different
of 2H-TRLs—4- to 6-fold more than in mitochondria. After cell types or different subcellular compartments). NanoSIMS
30 min, 2H enrichment was 12-fold greater in lipid droplets imaging will continue to be a useful approach in sorting out fac-
than in mitochondria, likely reflecting intervening oxidation of tors that regulate lipolysis and lipid movement within tissues. In
2
H-labeled fatty acids in mitochondria. We also examined the past, others have concluded, based largely on perturbations
13
C distribution in cardiomyocytes 1 day after administering of lipid stores in knockout mice, that vascular endothelial growth
13
C-labeled fatty acids by gavage. Only 5% of the 13C in cardi- factor-B is crucial for regulating fatty acid transport across capil-
omyocytes was located in cytosolic lipid droplets, likely reflect- lary endothelial cells (Hagberg et al., 2010). Fatty acid-binding
ing the use of fatty acids for membrane phospholipids and the proteins and fatty acyl-CoA synthetases have also been pro-
use of fatty acid metabolites in the synthesis of nonessential posed to play a role in the delivery of lipids to myocytes (Hagberg
amino acids. et al., 2010, 2013). Recently, a valine metabolite, 3-hydroxyiso-
CD36 is expressed at high levels in capillary endothelial cells butyrate, was proposed to regulate fatty acid transport across
(Greenwalt et al., 1995), and is often assumed to play a role in endothelial cells (Jang et al., 2016), based largely on in vitro
uptake of fatty acids by cells and tissues (Tanaka et al., 1997; models and measurements of lipid stores in tissues. NanoSIMS
Yang et al., 2007). When radioiodinated fatty acid derivatives imaging could be useful for defining the impact of both specific
are injected intravenously into humans, they are taken up by genes and small molecules in lipid movement into and across
the heart. Uptake of those radiopharmaceuticals by the heart capillaries. The utility of NanoSIMS imaging for metabolism
is reduced in subjects with CD36 deficiency (Hwang et al., research will extend well beyond TRL metabolism. For example,
1998; Tanaka et al., 1997). Also, when Cd36 / mice are NanoSIMS could simultaneously quantify the uptake and sub-
fasted, fatty acids accumulate in the plasma, and triglyceride cellular distribution of 2H-labeled glucose (or 2-deoxyglucose),
15
accumulation in the heart is reduced (Bharadwaj et al., 2010). N-labeled glutamine, and 13C-labeled fatty acids in solid
These observations suggest that CD36 could be important for tumors, making it possible to define metabolic heterogeneity in
the uptake of free fatty acids in the plasma compartment, but tumor cells, stromal cells, and endothelial cells.
it is unclear whether CD36 deficiency influences uptake of
TRL-derived nutrients by cardiomyocytes. Bharadwaj et al. Limitations of Study
(2010) loaded human VLDL with 14C-labeled triglycerides There are certain limitations to our study. One is that the number
in vitro and then injected the TRLs into wild-type and of mice that we studied was low, and the number of images
Cd36 / mice; they found reduced amounts of 14C in heart tis- generated per mouse was rather limited. NanoSIMS imaging is
sue extracts from Cd36 / mice. On the other hand, Cd36 defi- very expensive, slow, and technically demanding. In a 24-hr
ciency appeared to have little effect on the uptake of lipids from period, only 8 to 12 images could be obtained from one very small
TRLs that had been endogenously labeled with 14C (14C-TRLs region of the heart. Although each image is generated from an
prepared in Lpl-deficient mice). In the current study, we used extraordinary amount of quantitative data on secondary ions, it
NanoSIMS to assess the effect of Cd36 deficiency on the fate is simply not feasible to image many mice per group, many
of 2H-TRL-derived lipids in the heart. After 1.5 or 2 min, 2H entry different lines of mice, or many different tissues. That is why we
into wild-type and Cd36 / cardiomyocytes was nearly iden- believe that NanoSIMS is most suitable for addressing an
tical, indicating that CD36 deficiency has little effect on the anatomical issue and less suitable when scientific questions
movement of TRL-derived lipids into cardiomyocytes. (This can be addressed by measuring tritium-labeled lipids in tissue
observation seems consistent with an absence of impaired extracts. Another limitation of our study is that it is conceivable
cardiac performance in Cd36 / mice [Koonen et al., 2007].) that there was some fatty acid diffusion within tissues during
However, we observed reduced entry of 2H into cardiomyocyte the PBS perfusion (prior to perfusion-fixation with carbodiimide
lipid droplets in Cd36 / mice. The cytosolic lipid droplets were and glutaraldehyde) or some fatty acid diffusion after fixation dur-
slightly smaller in Cd36 / mice, but we are skeptical that their ing the preparation of tissues for NanoSIMS imaging. However,
smaller size prevented us from detecting them by NanoSIMS. this type of lipid diffusion could not explain the rapid entry of
Cd36 deficiency has complex effects on cardiomyocytes, deuterium into neutral lipids of cytosolic lipid droplets of
including effects on insulin and calcium signaling, fatty acyl- cardiomyocytes. Our NanoSIMS images uncovered deuterium-
CoA levels, and fatty acid esterification (Abumrad and Gold- labeled lipids in the subendothelial spaces of the heart, but it
berg, 2016; Bharadwaj et al., 2010; Xu et al., 2013), and those is likely that the patchy localization of those lipids was due
effects presumably could contribute, directly or indirectly, to to imidazole and osmium binding. Also, even though the

(E) 13C/12C ratio in cardiomyocytes of Cd36+/+ and Cd36 / mice. Each data point represents one cardiomyocyte in six 30 3 30 mm NanoSIMS images.
***p < 0.0005.
(F) 13C enrichment in cardiomyocyte lipid droplets as a percentage of total 13C in cardiomyocytes. Graphs were generated by quantifying six 30 3 30 mm images
for each sample. **p < 0.01. See also Figures S5–S7.
(G) 13C/12C ratio in mitochondria and lipid droplets of Cd36+/+ and Cd36 / mice. Six 30 3 30 mm NanoSIMS images were analyzed. Each data point shows the
mean 13C/12C ratio in the mitochondria and lipid droplets in each image (>50 mitochondria and 5–50 lipid droplets were analyzed per image). ***p < 0.0005.

1064 Cell Metabolism 27, 1055–1066, May 1, 2018


deuterium-enriched, imidazole-staining patches were primarily DECLARATION OF INTERESTS
located around small capillaries, we cannot fully exclude the pos- The authors declare no competing interests.
sibility that some of the deuterium-labeled lipids moved into the
subendothelial spaces during tissue processing. Finally, we Received: December 14, 2017
would point out that the 13C NanoSIMS images (obtained at least Revised: February 16, 2018
22 hr after the administration of 13C-labeled fatty acids) should Accepted: March 24, 2018
not be interpreted as exclusively showing lipid distribution, given Published: May 1, 2018

that some of the 13C-labeled fatty acids were metabolized and


REFERENCES
used to generate amino acid substrates for protein synthesis.
Abumrad, N.A., and Goldberg, I.J. (2016). CD36 actions in the heart: lipids,
STAR+METHODS calcium, inflammation, repair and more? Biochim. Biophys. Acta 1861,
1442–1449.
Detailed methods are provided in the online version of this paper Angermu €ller, S., and Fahimi, H.D. (1982). Imidazole-buffered osmium tetrox-
ide: an excellent stain for visualization of lipids in transmission electron micro-
and include the following:
scopy. Histochem. J. 14, 823–835.
d KEY RESOURCES TABLE Beigneux, A.P., Davies, B., Gin, P., Weinstein, M.M., Farber, E., Qiao, X.,
d CONTACT FOR REAGENT AND RESOURCE SHARING Peale, P., Bunting, S., Walzem, R.L., Wong, J.S., et al. (2007).
Glycosylphosphatidylinositol-anchored high density lipoprotein-binding
d EXPERIMENTAL MODEL AND SUBJECT DETAILS
protein 1 plays a critical role in the lipolytic processing of chylomicrons. Cell
B Mice
Metab. 5, 279–291.
d METHOD DETAILS
2 Beigneux, A.P., Gin, P., Davies, B.S.J., Weinstein, M.M., Bensadoun, A., Fong,
B H-labeled Triglyceride-rich Lipoproteins L.G., and Young, S.G. (2009). Highly conserved cysteines within the Ly6
13 2
B Administration of [ C]Fatty Acids and H-TRLs to Mice domain of GPIHBP1 are crucial for the binding of lipoprotein lipase. J. Biol.
B Isolated Mouse Heart Experiments Chem. 284, 30240–30247.
B Preparation of Tissue Sections for Electron Micro- Bharadwaj, K.G., Hiyama, Y., Hu, Y., Huggins, L.A., Ramakrishnan, R.,
scopy and NanoSIMS Imaging Abumrad, N.A., Shulman, G.I., Blaner, W.S., and Goldberg, I.J. (2010).
B NanoSIMS and Backscattered Electron (BSE) Imaging Chylomicron- and VLDL-derived lipids enter the heart through different path-
ways: in vivo evidence for receptor- and non-receptor-mediated fatty acid
B Transmission Electron Microscopy
uptake. J. Biol. Chem. 285, 37976–37986.
B Transmission Electron Microscopy of Negatively
Bligh, E.G., and Dyer, W.J. (1959). A rapid method of total lipid extraction and
Stained Lipoproteins
purification. Can. J. Biochem. Physiol. 37, 911–917.
B Immunohistochemistry
Dallwig, R., and Deitmer, J.W. (2002). Cell-type specific calcium responses in
B Statistics
acute rat hippocampal slices. J. Neurosci. Methods 116, 77–87.
Davies, B.S., Goulbourne, C.N., Barnes, R.H., 2nd, Turlo, K.A., Gin, P.,
SUPPLEMENTAL INFORMATION Vaughan, S., Vaux, D.J., Bensadoun, A., Beigneux, A.P., Fong, L.G., et al.
(2012). Assessing mechanisms of GPIHBP1 and lipoprotein lipase movement
Supplemental Information includes seven figures and one table and can be across endothelial cells. J. Lipid Res. 53, 2690–2697.
found with this article online at https://doi.org/10.1016/j.cmet.2018.03.017.
Davies, B.S.J., Beigneux, A.P., Barnes, R.H., II, Tu, Y., Gin, P., Weinstein,
M.M., Nobumori, C., Nyrén, R., Goldberg, I.J., Olivecrona, G., et al. (2010).
ACKNOWLEDGMENTS GPIHBP1 is responsible for the entry of lipoprotein lipase into capillaries.
Cell Metab. 12, 42–52.
We are grateful for the NanoSIMS facilities at California Institute of Technology
Febbraio, M., Abumrad, N.A., Hajjar, D.P., Sharma, K., Cheng, W., Pearce,
(Pasadena, CA) and the University of Western Australia (Perth). We acknowl-
S.F.A., and Silverstein, R.L. (1999). A null mutation in murine CD36 reveals
edge support from the Leducq Foundation Transatlantic Network grant
an important role in fatty acid and lipoprotein metabolism. J. Biol. Chem.
(12CVD04) (to S.G.Y.), the NIH (P01 HL090553, R01 HL087228, and
274, 19055–19062.
HL125335 to S.G.Y.), and a Ruth L. Kirschstein National Research Service
Award, F32 HL132471 (to C.H.). We also acknowledge the Australian Micro- Fong, L.G., Young, S.G., Beigneux, A.P., Bensadoun, A., Oberer, M., Jiang, H.,
scopy & Microanalysis Research Facility and the Science and Industry Endow- and Ploug, M. (2016). GPIHBP1 and plasma triglyceride metabolism. Trends
ment Fund for supporting the Ion Probe Facility at the Centre for Microscopy, Endocrinol. Metab. 27, 455–469.
Characterisation and Analysis at the University of Western Australia. We thank Goulbourne, C.N., Gin, P., Tatar, A., Nobumori, C., Hoenger, A., Jiang, H.,
Dr. Marianne Cilluffo for help in preparing tissue sections. Grovenor, C.R., Adeyo, O., Esko, J.D., Goldberg, I.J., et al. (2014). The
GPIHBP1-LPL complex is responsible for the margination of triglyceride-rich
AUTHOR CONTRIBUTIONS lipoproteins in capillaries. Cell Metab. 19, 849–860.
Greenwalt, D.E., Scheck, S.H., and Rhinehart-Jones, T. (1995). Heart CD36
C.H. designed, performed, and analyzed the experiments, and prepared fig- expression is increased in murine models of diabetes and in mice fed a high
ures and the initial draft of the paper. T.A.W. performed electron microscopy fat diet. J. Clin. Invest. 96, 1382–1388.
and prepared tissue sections for NanoSIMS. R.S.J. performed the experi-
Guo, W., Huang, N., Cai, J., Xie, W., and Hamilton, J.A. (2006). Fatty acid trans-
ments and analyzed NanoSIMS data. P.H. managed the mouse colony and
port and metabolism in HepG2 cells. Am. J. Physiol. Gastrointest. Liver
performed mouse experiments. M.L. performed isolated heart studies. X.H.
Physiol. 290, G528–G534.
performed NanoSIMS studies. C.M.A. performed immunohistochemistry
studies. P.T., K.R., M.P., and A.P.B. advised on experimental design and Hagberg, C., Mehlem, A., Falkevall, A., Muhl, L., and Eriksson, U. (2013).
data analysis. A.H., M.K., and P.G. assisted with NanoSIMS. D.A.F. performed Endothelial fatty acid transport: role of vascular endothelial growth factor B.
lipidomics studies. L.G.F. and S.G.Y. analyzed the data and prepared the Physiology (Bethesda) 28, 125–134.
paper. H.J. performed NanoSIMS and BSE imaging, analyzed the data, and Hagberg, C.E., Falkevall, A., Wang, X., Larsson, E., Huusko, J., Nilsson, I., van
assisted with figures and the paper. Meeteren, L.A., Samen, E., Lu, L., Vanwildemeersch, M., et al. (2010). Vascular

Cell Metabolism 27, 1055–1066, May 1, 2018 1065


endothelial growth factor B controls endothelial fatty acid uptake. Nature 464, Mysling, S., Kristensen, K.K., Larsson, M., Beigneux, A.P., Gardsvoll, H., Fong,
917–921. L.G., Bensadouen, A., Jorgensen, T.J., Young, S.G., and Ploug, M. (2016a).
Hamilton, J.A., Johnson, R.A., Corkey, B., and Kamp, F. (2001). Fatty acid The acidic domain of the endothelial membrane protein GPIHBP1 stabilizes
transport: the diffusion mechanism in model and biological membranes. lipoprotein lipase activity by preventing unfolding of its catalytic domain.
J. Mol. Neurosci. 16, 99–108, discussion 151–107. eLife 5, e12095.
Havel, R.J. (2010). Triglyceride-rich lipoproteins and plasma lipid transport. Mysling, S., Kristensen, K.K., Larsson, M., Kovrov, O., Bensadouen, A.,
Arterioscler. Thromb. Vasc. Biol. 30, 9–19. Jorgensen, T.J., Olivecrona, G., Young, S.G., and Ploug, M. (2016b). The
Havel, R.J., and Gordon, R.S., Jr. (1960). Idiopathic hyperlipemia: metabolic angiopoietin-like protein ANGPTL4 catalyzes unfolding of the hydrolase
studies in an affected family. J. Clin. Invest. 39, 1777–1790. domain in lipoprotein lipase and the endothelial membrane protein GPIHBP1
counteracts this unfolding. eLife 5, e20958.
Havel, R.J., and Kane, J.P. (2001). Introduction: structure and metabolism of
plasma lipoproteins. In The Metabolic and Molecular Bases of Inherited Quehenberger, O., Armando, A.M., Brown, A.H., Milne, S.B., Myers, D.S.,
Disease, C.R. Scriver, A.L. Beaudet, W.S. Sly, D. Valle, B. Childs, K.W. Merrill, A.H., Bandyopadhyay, S., Jones, K.N., Kelly, S., Shaner, R.L., et al.
Kinzler, and B. Vogelstein, eds. (McGraw-Hill), pp. 2705–2716. (2010). Lipidomics reveals a remarkable diversity of lipids in human plasma.
He, C., Fong, L.G., Young, S.G., and Jiang, H. (2017a). NanoSIMS imaging: an J. Lipid Res. 51, 3299–3305.
approach for visualizing and quantifying lipids in cells and tissues. J. Investig. Scow, R.O., Blanchette-Mackie, E.J., and Smith, L.C. (1976). Role of capillary
Med. 65, 669–672. endothelium in the clearance of chylomicrons. A model for lipid transport from
He, C., Hu, X., Jung, R.S., Weston, T.A., Sandoval, N.P., Tontonoz, P., Kilburn, blood by lateral diffusion in cell membranes. Circ. Res. 39, 149–162.
M.R., Fong, L.G., Young, S.G., and Jiang, H. (2017b). High-resolution imaging
Shao, F., and Ford, D.A. (2014). Elaidic acid increases hepatic lipogenesis by
and quantification of plasma membrane cholesterol by NanoSIMS. Proc. Natl.
mediating sterol regulatory element binding protein-1c activity in huh-7 cells.
Acad. Sci. USA 114, 2000–2005.
Lipids 49, 403–413.
Hwang, E.H., Taki, J., Yasue, S., Fujimoto, M., Taniguchi, M., Matsunari, I.,
Sidossis, L.S., Coggan, A.R., Gastaldelli, A., and Wolfe, R.R. (1995). Pathway
Nakajima, K., Shiobara, S., Ikeda, T., and Tonami, N. (1998). Absent myocar-
of free fatty acid oxidation in human subjects. Implications for tracer studies.
dial iodine-123-BMIPP uptake and platelet/monocyte CD36 deficiency.
J. Clin. Invest. 95, 278–284.
J. Nucl. Med. 39, 1681–1684.
Jang, C., Oh, S.F., Wada, S., Rowe, G.C., Liu, L., Chan, M.C., Rhee, J., Takei, S., Hasegawa-Ishii, S., Uekawa, A., Chiba, Y., Umegaki, H., Hosokawa, M.,
Hoshino, A., Kim, B., Ibrahim, A., et al. (2016). A branched-chain amino acid Woodward, D.F., Watanabe, K., and Shimada, A. (2012). Immunohistochemical
metabolite drives vascular fatty acid transport and causes insulin resistance. demonstration of increased prostaglandin F(2)alpha levels in the rat hippocam-
Nat. Med. 22, 421–426. pus following kainic acid-induced seizures. Neuroscience 218, 295–304.
Jiang, H., Favaro, E., Goulbourne, C.N., Rakowska, P.D., Hughes, G.M., Tanaka, T., Okamoto, F., Sohmiya, K., and Kawamura, K. (1997). Lack of
Ryadnov, M.G., Fong, L.G., Young, S.G., Ferguson, D.J., Harris, A.L., et al. myocardial iodine-123 15-(p-iodiphenyl)-3-R,S-methylpentadecanoic acid
(2014a). Stable isotope imaging of biological samples with high resolution sec- (BMIPP) uptake and CD36 abnormality–CD36 deficiency and hypertrophic
ondary ion mass spectrometry and complementary techniques. Methods 68, cardiomyopathy. Jpn. Circ. J. 61, 724–725.
317–324.
Tymianski, M., Bernstein, G.M., Abdel-Hamid, K.M., Sattler, R., Velumian, A.,
Jiang, H., Goulbourne, C.N., Tatar, A., Turlo, K., Wu, D., Beigneux, A.P., Carlen, P.L., Razavi, H., and Jones, O.T. (1997). A novel use for a carbodiimide
Grovenor, C.R., Fong, L.G., and Young, S.G. (2014b). High-resolution imaging compound for the fixation of fluorescent and non-fluorescent calcium indica-
of dietary lipids in cells and tissues by NanoSIMS analysis. J. Lipid Res. 55, tors in situ following physiological experiments. Cell Calcium 21, 175–183.
2156–2166.
Weinstein, M.M., Yin, L., Tu, Y., Wang, X., Wu, X., Castellani, L.W., Walzem,
Kane, J.P., and Havel, R.J. (2001). Disorders of the biogenesis and secretion of
R.L., Lusis, A.J., Fong, L.G., Beigneux, A.P., et al. (2010). Chylomicronemia
lipoproteins containing the B apolipoproteins. In The Metabolic and Molecular
elicits atherosclerosis in mice. Arterioscler. Thromb. Vasc. Biol. 30, 20–23.
Bases of Inherited Disease, C.R. Scriver, A.L. Beaudet, W.S. Sly, D. Valle, B.
Childs, K.W. Kinzler, and B. Vogelstein, eds. (McGraw-Hill), pp. 2717–2752. Xu, S., Jay, A., Brunaldi, K., Huang, N., and Hamilton, J.A. (2013). CD36
enhances fatty acid uptake by increasing the rate of intracellular esterification
Koonen, D.P., Febbraio, M., Bonnet, S., Nagendran, J., Young, M.E.,
but not transport across the plasma membrane. Biochemistry 52, 7254–7261.
Michelakis, E.D., and Dyck, J.R. (2007). CD36 expression contributes to
age-induced cardiomyopathy in mice. Circulation 116, 2139–2147. Yang, J., Sambandam, N., Han, X., Gross, R.W., Courtois, M., Kovacs, A.,
Korn, E.D. (1955a). Clearing factor, a heparin-activated lipoprotein lipase. I. Febbraio, M., Finck, B.N., and Kelly, D.P. (2007). CD36 deficiency rescues
Isolation and characterization of the enzyme from normal rat heart. J. Biol. lipotoxic cardiomyopathy. Circ. Res. 100, 1208–1217.
Chem. 215, 1–14. Yao, G. (2000). Effect of LPL activity on fatty acid composition in mouse
Korn, E.D. (1955b). Clearing factor, a heparin-activated lipoprotein lipase. II. plasma, liver, skeletal and cardiac muscles, adipose and brain tissues,
Substrate specificity and activation of coconut oil. J. Biol. Chem. 215, 15–26. M.Sc. thesis (Université Laval).

1066 Cell Metabolism 27, 1055–1066, May 1, 2018


Neuron

Article

TREM2 Regulates Microglial Cholesterol Metabolism


upon Chronic Phagocytic Challenge
Alicia A. Nugent,1,2 Karin Lin,1,2 Bettina van Lengerich,1 Steve Lianoglou,1 Laralynne Przybyla,1 Sonnet S. Davis,1
Ceyda Llapashtica,1 Junhua Wang,1 Do Jin Kim,1 Dan Xia,1 Anthony Lucas,1 Sulochanadevi Baskaran,1,3
Patrick C.G. Haddick,1 Melina Lenser,1 Timothy K. Earr,1 Ju Shi,1,4 Jason C. Dugas,1 Benjamin J. Andreone,1 Todd Logan,1
Hilda O. Solanoy,1 Hang Chen,1,5 Ankita Srivastava,1 Suresh B. Poda,1 Pascal E. Sanchez,1 Ryan J. Watts,1
Thomas Sandmann,1 Giuseppe Astarita,1 Joseph W. Lewcock,1 Kathryn M. Monroe,1,* and Gilbert Di Paolo1,6,*
1Denali Therapeutics, South San Francisco, CA 94080, USA
2These authors contributed equally
3Current address: Frontier Medicines, South San Francisco, CA 94080, USA
4Current address: Jazz Pharmaceuticals, Palo Alto, CA 94304, USA
5Current address: BridGene Biosciences, Sunnyvale, CA 94089, USA
6Lead Contact

*Correspondence: monroe@dnli.com (K.M.M.), dipaolo@dnli.com (G.D.P.)


https://doi.org/10.1016/j.neuron.2019.12.007

SUMMARY ceptors, and enhanced cytokine secretion (Deczkowska et al.,


2018; Hammond et al., 2018). Recent advances in single-cell
Loss-of-function (LOF) variants of TREM2, an im- RNA sequencing (scRNA-seq) have revealed multiple states of
mune receptor expressed in microglia, increase microglia. These studies suggest that functional sub-popula-
Alzheimer’s disease risk. TREM2 senses lipids and tions of microglia co-exist within the brain in various mouse
mediates myelin phagocytosis, but its role in micro- models of neurodegenerative diseases, including homeostatic
glial lipid metabolism is unknown. Combining and disease-associated microglia (DAM), the microglial neuro-
degenerative phenotype (MGnD), and activated response micro-
chronic demyelination paradigms and cell sorting
glia (ARM) (Deczkowska et al., 2018; Keren-Shaul et al., 2017;
with RNA sequencing and lipidomics, we find that
Krasemann et al., 2017; Sala Frigerio et al., 2019). Collectively,
wild-type microglia acquire a disease-associated we refer to responsive microglial states as damage-associated
transcriptional state, while TREM2-deficient micro- microglia, because different disease or aging models show sig-
glia remain largely homeostatic, leading to neuronal nificant overlap but distinct transcriptional profiles. Whether
damage. TREM2-deficient microglia phagocytose transition to damage-associated microglia states is beneficial
myelin debris but fail to clear myelin cholesterol, re- or deleterious remains unclear and likely depends on the disease
sulting in cholesteryl ester (CE) accumulation. CE in- type, stage, and inherent pathology. Identifying the molecular
crease is also observed in APOE-deficient glial cells, and cellular basis underlying microglial states in healthy and dis-
reflecting impaired brain cholesterol transport. This ease conditions may facilitate the development of immune ther-
finding replicates in myelin-treated TREM2-deficient apies for the treatment of neurodegenerative diseases.
Microglial dysfunction appears central to the etiology of late-
murine macrophages and human iPSC-derived mi-
onset Alzheimer’s disease (LOAD), as large-scale genetic
croglia, where it is rescued by an ACAT1 inhibitor
studies have uncovered variants in LOAD risk-associated genes
and LXR agonist. Our studies identify TREM2 as a that are highly expressed in microglia (Carmona et al., 2018;
key transcriptional regulator of cholesterol transport Efthymiou and Goate, 2017). One of the LOAD genes encodes
and metabolism under conditions of chronic myelin triggering receptor expressed on myeloid cells 2 (TREM2), a sin-
phagocytic activity, as TREM2 LOF causes patho- gle-pass transmembrane immune receptor selectively ex-
genic lipid accumulation in microglia. pressed in microglia within the CNS. Individuals carrying rare
heterozygous variants of TREM2, such as R47H, have higher
LOAD risk (average odds ratio [OR] 4.5) (Guerreiro et al.,
INTRODUCTION 2013; Jonsson et al., 2013; Ulland and Colonna, 2018). The
immunoglobulin-like ectodomain (ECD) of TREM2 binds various
Microglia constantly surveil the brain parenchyma to eliminate ligands, including lipids, and the AD variant R47H has reduced
dead cells, dysfunctional synapses, and other cellular debris affinity for apolipoproteins (e.g., APOE) or lipid ligands, suggest-
(Deczkowska et al., 2018; Hammond et al., 2018; Ransohoff, ing that the increased LOAD risk reflects a partial loss of function
2016). With aging or in pathological conditions, parenchyma (LOF) in lipid recognition and/or lipid-induced signaling (Atagi
damage can facilitate the transition of microglia from homeostat- et al., 2015; Bailey et al., 2015; Sudom et al., 2018; Ulland and
ic into reactive states. This transition profoundly alters the Colonna, 2018; Wang et al., 2016; Yeh et al., 2016). Trem2–/–
microglial transcriptome, leading to morphological changes, and Trem2R47H mutant microglia fail to surround and clear
increased phagocytic activity, expression of various immune re- amyloid plaques in vivo, resulting in accumulation of dystrophic

Neuron 105, 837–854, March 4, 2020 ª 2019 Elsevier Inc. 837


A C F
Trem2 + /+ + /- -/- Microglial DAM genes
Control CPZ Control CPZ Control CPZ
+/+
5 week
+/- -/- +/+
12 week
+/- -/-
Ctl CPZ Ctl CPZ Ctl CPZ Ctl CPZ Ctl CPZ Ctl CPZ

5 week CPZ

Down
Trem2 + /+ + /- -/-
Ctl CPZ Control CPZ Control CPZ
12 week CPZ

Trem2 +/+ Control


Trem2 +/- Cuprizone
Trem2 -/-

B Trem2 genotype effect 12 wk Cuprizone


effect
+/+ vs -/- +/+ vs +/- +/+ +/-

6 0 0 199 159 740

Up
0 8
0 0 1 6
0 2

+/- vs -/- -/-


D E
Lysosomal function Lipid metabolism
control treatment (log2)
control treatment (log2)

Change from +/+


Change from +/+

+/+ +/- -/- +/+ +/- -/- +/+ +/- -/- +/+ +/- -/-
Trem2 genotype Trem2 genotype log2 FC
Cst7 Ctse Galns Hexa Apoc1
A Fabp3 Lpl Olr1 4 2 0 -2 -4
Ctsb Ctsl Gla Prcp Apoe Fabp5
F Nceh1 Soat1
Ctsd Ctsz Gusb Ch25h Lipa Npc2
G H I
Homeostatic genes Stage 1 DAM genes Stage 2 DAM genes
control treatment (log2)

control treatment (log2)

control treatment (log2)


Change from +/+

Change from +/+

Change from +/+

+/+ +/- -/- +/+ +/- -/- +/+ +/- -/- +/+ +/- -/- +/+ +/- -/- +/+ +/- -/-
Trem2 genotype Trem2 genotype Trem2 genotype
Cx3cr1 P2ry12 Tmem119
T Apoe Ctsb Fth1 Tyrobp Axl Cd9 Csf1 Ctsl Lpl
B2m Ctsd Lyz2 Ccl6 Clec7a Csf7 Itgax Timp2

Figure 1. TREM2 Deficiency Prevents DAM Conversion during Chronic Demyelination


(A) Principal-component analysis of top 500 differentially expressed genes in bulk microglia isolated from brains of control versus 5 week and control versus
12 week cuprizone (CPZ)-treated Trem2+/+, Trem2+/–, and Trem2–/– mice (n = 2–4 mice per condition).
(B) Number of differentially expressed genes in bulk microglia after control (left) or 12 week CPZ diet (right). ANOVA; FDR < 0.1; absolute log2 FC > 0.5.
(C) Heatmap of top gene expression changes (log2 fold change) in bulk microglia after 5 (top) and 12 (bottom) week CPZ diet. Orange marks indicate previously
identified DAM genes. ANOVA, FDR < 0.1, absolute log2 FC > 0.5; columns represent individual mice.

(legend continued on next page)

838 Neuron 105, 837–854, March 4, 2020


neurites near diffuse plaques (Jay et al., 2017; Parhizkar et al., lipidomic techniques revealed elevation of CE in Trem2–/– mi-
2019; Ulland and Colonna, 2018; Wang et al., 2015, 2016; croglia, a phenotype also observed in Apoe–/– microglia after
Yuan et al., 2016). Additionally, TREM2 LOF modulates tau seed- CPZ diet. This could be reproduced in vitro, where CE accu-
ing, spreading, and tau-associated neuroinflammation and mulation occurs in Trem2–/– bone marrow-derived macro-
neurotoxicity (Bemiller et al., 2017; Leyns et al., 2017, 2019; phages (BMDMs) and human induced pluripotent stem cell
Sayed et al., 2018). Trem2 overexpression reduces amyloid (iPSC)-derived microglia subjected to myelin challenge. CE
burden and neuritic dystrophy in an Alzheimer’s disease (AD) accumulation was rescued by inhibition of acetyl-CoA acetyl-
mouse model (Lee et al., 2018). Rare homozygous TREM2 LOF transferase 1 (ACAT1), which converts cholesterol to CE, and
mutations cause Nasu-Hakola disease, a syndrome with recur- by a liver X receptor (LXR) agonist, suggesting that TREM2
rent bone fractures, myelin loss, neurodegeneration, and early- LOF causes an impairment in cholesterol efflux. Our results
onset dementia, likely resulting from microglial dysfunction establish a key role for TREM2 in the control of microglial
(Paloneva et al., 2002). gene expression and cholesterol transport upon chronic
Consistent with a role in microglia proliferation and survival, myelin phagocytosis. Failure to properly execute this program
TREM2 signals via receptor tyrosine kinases DNAX-activation results in extensive neuronal damage in the brain. Because CE
protein 10 (DAP10) and DAP12 to modulate proliferation, sur- is known to accumulate in AD patient brain and AD mouse
vival, immune response, calcium mobilization, cytoskeletal dy- models (Astarita et al., 2011; Chan et al., 2012; Morel et al.,
namics, mTOR signaling, autophagy, and energy metabolism 2013; Shibuya et al., 2015), and LOAD-linked TREM2 variants
(Ulland and Colonna, 2018; Ulland et al., 2017; Yeh et al., result in a partial LOF (Ulland and Colonna, 2018), our study
2017). A proposed function of TREM2 is to mediate microglial strongly suggests that enhancing TREM2 function may be
response and transition to a damage-associated microglia state beneficial in AD, in part by facilitating lipid clearance in
via control of gene expression (Deczkowska et al., 2018; Ulland microglia.
and Colonna, 2018). scRNA-seq analyses of TREM2-deficient
5XFAD microglia revealed the inability to acquire a late-stage RESULTS
DAM profile (DAM stage 2), instead halting at an intermediate
DAM state (DAM stage 1) (Keren-Shaul et al., 2017). Other TREM2 Deficiency Prevents DAM Conversion during
studies showed a key role for TREM2 and APOE in the transition Chronic Demyelination
to damage-associated microglia states (Krasemann et al., 2017; To characterize the effects of acute and chronic demyelination
Götzl et al., 2019). Microarray studies in Trem2–/– mice subjected on TREM2-dependent gene expression in microglia, Trem2+/+,
to a demyelinating cuprizone (CPZ) diet also showed that TREM2 Trem2+/–, and Trem2–/– mice were fed a 0.2% CPZ diet for 5 or
controls expression of many microglial genes controlling lipid 12 weeks. CD11b+ microglia were isolated from hemibrain using
transport (Apoe) or catabolism (Lpl) (Poliani et al., 2015). FACS (Figures S1A–S1E). CPZ induced transcriptional changes
Trem2–/– mice showed normal demyelination in the acute phase in Trem2+/+ and Trem2+/– microglia, whereas CPZ-challenged
but partial remyelination during the recovery phase, suggesting Trem2–/– microglia clustered with those of untreated mice (prin-
that TREM2 may bind to myelin debris for engulfment and clear- cipal-component analysis [PCA]; Figure 1A). Few genotype-
ance, a process required for proper remyelination (Cantoni et al., dependent differences were found under normal diet conditions
2015; Poliani et al., 2015). However, whether TREM2 mediates (Figure 1B), but CPZ caused significant changes in hundreds of
myelin lipid processing in microglia is unknown. genes after 5 and 12 weeks (Figures 1B and 1C; absolute log2
We sought to assess the physiological role of TREM2 in fold change [FC] > 0.5, false discovery rate [FDR] < 0.2; Data
microglial lipid metabolism by subjecting mice to a demyelin- S1). These changes were restricted primarily to Trem2+/+ and
ating CPZ diet (Praet et al., 2014). Myelin contains the majority Trem2+/– microglia, while Trem2–/– microglia largely failed to
(>80%) of the brain’s free cholesterol (Martı́n et al., 2014), respond to CPZ (Figures 1B and 1C).
allowing the assessment of microglial response to a chronic Analysis of gene sets from the Reactome database (Fabregat
and extensive cholesterol challenge that is selective to et al., 2018) revealed Trem2-dependent upregulation of genes
the brain and independent of peripheral lipid metabolism. involved in lysosome and phagosome function, AD, oxidative
CPZ triggered the TREM2-dependent transition from a phosphorylation, and cholesterol metabolism. Key lysosome
homeostatic to damage-associated microglia state with cathepsin genes, such as Ctse and Ctsl, were upregulated
upregulation of genes controlling lipid transport and meta- 2-fold upon chronic demyelination in Trem2+/+ and Trem2+/– mi-
bolism. Chronic myelin phagocytosis caused accumulation croglia (FDR < 0.05) but unchanged in Trem2–/– microglia (Fig-
of cholesteryl ester (CE) and oxidized CE (oxCE) in Trem2–/– ure 1D; Data S1; interaction p < 0.05). Previous microarray
brain. Fluorescence-activated cell sorting (FACS)-based studies reported a failure of Trem2–/– microglia to upregulate

(D and E) Expression changes in individual genes associated with (D) lysosomal function and (E) lipid metabolism in bulk microglia with control (left inset) or CPZ
diet (right inset) for 5 (top) or 12 (bottom) weeks.
(F) Heatmap of bulk microglial expression changes in the top 69 DAM genes downregulated (top) or upregulated (bottom) in 5XFAD compared with wild-type
microglia from Karen-Shaul et al. 2017 after 5 and 12 week control versus CPZ diet. Camera; p < 1 3 10 41.
(G–I) Expression changes in individual (G) homeostatic, (H) stage 1, and (I) stage 2 DAM genes (identified in Keren-Shaul et al., 2017) in Trem2+/+, Trem2+/–, and
Trem2–/– bulk microglia with control (left inset) or CPZ diet (right inset) for 5 (top) or 12 (bottom) weeks.
See also Figure S1 and Data S1.

Neuron 105, 837–854, March 4, 2020 839


A B
All cells Sample enrichment DAM activity
1
Cluster
1 5
2 30
6 20
3
10
4 2 0
5 S
6 8
7
8 4

3
0 5 10 15 20 Trem2 Trem2 Trem2 Trem2 DAM1 DAM1 DAM2
% of sample +/+ +/+ +/- -/- down up up
(average) Ctl 12wk 12wk 12wk
CPZ CPZ CPZ
C D
Cluster gene markers
Cluster gene marker expression
3000
Apoe

2000
1000
1 down

0
50
40
30
Lpl

20
10
0
1 up

200
Ctsb

100
0
600
2 up

Ctsd

400
200
0
90
Ctsz
3 down

60
30
0
100
P2ry12

75
50
25
3 up

0
200
150
B2m

100
50
0

Normalized UMI
4 down

40
Cd68

20
0
100
Cx3cr1

75
50
4 up

25
0
60
Lgals3

40
5u 5d

20
0
600
Spp1

400
200
6d

0
50
6u

40
Cst7

30
20
10
7d

0
120
Cd63

80
40
7 up

0
20
Capg

15
10
5
0
8 up

300
Fth1

200
100
0

Figure 2. scRNA-Seq Confirms that Trem2–/– Microglia Exhibit Attenuated Transition to DAM upon Demyelination
(A) tSNE (t-distributed stochastic neighbor embedding) plot of the 3,023 single microglia sorted from Trem2+/+ with control diet and Trem2+/+, Trem2+/–, and
Trem2–/– with 12 week CPZ, colored by cluster assignment (n = 2 pooled mice per condition).
(B) Percentage composition of all cells within each cluster across all samples (left), within each cluster per condition (middle), and aggregated scores of DAM-
related gene sets per cluster (right). Gene set scores per cell are normalized to zero mean and unit variance and visualized in the heatmap as their average over all
cells per cluster.
(legend continued on next page)

840 Neuron 105, 837–854, March 4, 2020


genes involved in phagocytosis and lipid metabolism (Poliani 2017). Unlike with CPZ, microglia from aged mice only showed
et al., 2015). We confirmed these results and expanded the list a mild increase in expression of cholesterol metabolism-related
of lipid metabolism-related genes (Figure 1E). Importantly, genes compared with young mice, suggesting that aged micro-
several Trem2-dependent genes control cholesterol transport glia face only a minor cholesterol burden. Thus, gene expression
and metabolism, including Apoe, Apoc1, Ch25h, Lipa, Nceh1, changes in microglia derived from CPZ-challenged Trem2–/–
Npc2, and Soat1. Gene set enrichment analysis also showed mice are also present in aged Trem2–/– microglia, although the
that DAM genes (Keren-Shaul et al., 2017) were significantly up- latter are less striking.
regulated with CPZ in Trem2+/+ and Trem2+/– microglia at both
time points but attenuated in Trem2–/– microglia (Figures 1C, scRNA-Seq Confirms that Trem2–/– Microglia Exhibit
1F, and S1F). This suggests that chronic demyelination induces Attenuated Transition to DAM upon Demyelination
expression changes related to those observed in 5XFAD and To determine if the population-based CPZ-induced transcrip-
SOD1 microglia (Keren-Shaul et al., 2017). tional changes observed were homogeneous across microglia,
Keren-Shaul et al. (2017) described the microglial transition we conducted scRNA-seq on CD11b+/CD45low microglia iso-
from homeostasis into DAM as a two-step process: a lated from CPZ-treated mice (Figures S2A–S2D). We used a
TREM2-independent transition to DAM stage 1, followed by a graph-based clustering approach (Xu and Su, 2015) to divide
TREM2-dependent transition to DAM stage 2. We compared 3,023 cells into eight microglia sub-populations (Figure 2A),
our TREM2 CPZ dataset with published homeostatic, DAM 1, each accounting for 2%–19% of analyzed cells (Figure 2B,
and DAM 2 gene profiles. Homeostatic genes P2ry12 and left). Quantification of cluster membership across groups (Fig-
Tmem119 were significantly downregulated in Trem2+/+ and ure 2B, middle) identified two treatment- and genotype-depen-
Trem2+/– (FDR < 0.01) but not Trem2–/– microglia in response dent clusters (4 and 8). Cluster 4 mainly contained microglia
to CPZ (Figure 1G; genotype-diet interaction p < 0.05; Data exposed to CPZ with at least one copy of Trem2 (1% of
S1). Although described as a TREM2-independent phenome- Trem2+/+ controls; 16%–19% of Trem2+/+ CPZ and Trem2+/–
non in an AD model, we observed reduced induction of DAM CPZ; 1.5% of Trem2–/– CPZ). Microglia from cluster 8 were
1 genes such as Apoe (interaction p < 0.001) and Fth1 (interac- largely absent in Trem2+/+ controls (< 2.3%), mildly increased
tion p < 0.005) in Trem2–/– compared with Trem2+/+ and in the Trem2+/+ CPZ and Trem2+/– CPZ mice (10%), and
Trem2+/– microglia following CPZ treatment (Figure 1H). Apoe most abundant in the Trem2–/– CPZ mice (20%) (Data S2).
expression was 8-fold higher with CPZ treatment of Trem2+/+ Cluster 3 consists of very few cells and is discussed in STAR
and Trem2+/– mice after 12 weeks but was attenuated in Methods.
Trem2–/– mice, similar to DAM 2 genes, such as Axl (interaction To further characterize cells within each cluster, we identified
p < 0.05) and Cd9 (interaction p < 0.001) (Figure 1I; Data S1). marker genes with cluster-specific over- and under-expression
Our data indicate a role of TREM2 in endolysosomal function (Data S2). Relative expression of the top 15 up- and downregu-
and lipid metabolism, with a clear implication of cholesterol lated genes per cluster confirmed that these clusters are distinct
transport and metabolism. Additionally, they suggest that (Figure 2C), although genes exclusive to a single cluster are rare.
CPZ elicits a damage-associated microglia state that fails to Consistent with bulk RNA-seq data, the top upregulated marker
be initiated in Trem2–/– microglia. genes in the Trem2+/+ CPZ and Trem2+/– CPZ-enriched cluster
4 are lysosomal genes (e.g., Ctsb, Ctsd, Ctsz) and genes
TREM2 Deficiency Reduces Age-Dependent Conversion involved in lipid metabolism (e.g., Apoe, Lpl) (Figure 2D). The
to DAM downregulated marker genes in cluster 4 include homeostatic
Microglial dysfunction is a hallmark of the aging brain, as cells genes, such as P2ry12 and Tmem119, suggesting microglia in
accumulate excessive myelin debris and acquire a DAM tran- this cluster are in a reactive state (Figures 2D and S2E). Cluster
scriptional state (Mecca et al., 2018; Safaiyan et al., 2016; 8 similarly consisted of cells with upregulated lysosome- and
Keren-Shaul et al., 2017). We sorted microglia derived from lipid metabolism-related genes, such as Ctsb, Ctsd, and Apoe,
young (2-month-old) and aged (15- to 17-month-old) wild-type but their expression was lower than that seen in cluster 4 (Figures
and Trem2–/– mice to assess whether aged Trem2–/– microglia 2D and S2F). About 20% of the total microglia population upre-
exhibit a DAM-like state. Aged wild-type microglia downregu- gulate the above genes; thus not all Trem2+/+ and Trem2+/–
lated a number of homeostatic genes and upregulated DAM 1 microglia alter gene expression with CPZ.
and DAM 2 genes compared to young microglia, albeit to a lesser Consistent with our bulk microglial findings, the transcriptome
degree than modulation in the CPZ model (Figures S1G and from Trem2+/+ and Trem2+/– CPZ cluster 4 largely contained up-
S1H). As with CPZ, activation of the DAM 2 gene set was atten- regulated marker genes that previously have been characterized
uated in aged Trem2–/– microglia compared with aged controls in DAM 2 expression (Keren-Shaul et al., 2017), including Lgals3,
(Figures S1G and S1H). This effect was most striking for Lpl Cd63, Spp1, Cst7, Cd68, Capg, and Fth1 (Figure 2D). To deter-
and Spp1, consistent with the Trem2 dependency of the DAM mine if clusters 4 and 8 relate to DAM stages, we aggregated
2 profile exhibited in 5XFAD microglia (Keren-Shaul et al., the expression of established DAM stage 1 and 2 marker genes

(C) Heatmap showing the relative gene expression profiles (normalized to zero mean and unit variance) of the top 15 up- and downregulated genes per cluster.
(D) Expression profiles for selected marker genes plotted as normalized counts per cell. Left legend denotes upregulated (up arrow) versus downregulated (down
arrow) marker genes in indicated clusters.
See also Figure S2 and Data S2.

Neuron 105, 837–854, March 4, 2020 841


(Data S2) for each cluster. Cluster 4 showed the highest enrich- CE22:6 showed the most striking increase in Trem2–/– brain
ment for DAM 2 gene expression, followed by cluster 8 (Fig- with chronic demyelination, upward of 38-fold compared with
ure 2B, right), suggesting that Trem2+/+ CPZ and Trem2+/– CPZ Trem2–/– control brain and 2.5-fold compared with Trem2+/+
microglia exhibit a DAM 2-like transition in response to CPZ 12 week CPZ brain (Figure 3C). Likewise, oxCE species, previ-
that is attenuated in Trem2–/– CPZ microglia. As in Keren-Shaul ously reported only in atherosclerotic lesions (Choi et al., 2017;
et al. (2017), DAM 1 genes were upregulated in Trem2+/+ Hutchins et al., 2011), were significantly enhanced in Trem2–/–
CPZ, Trem2+/– CPZ, and Trem2–/– CPZ microglia, although brain compared with Trem2+/+ with chronic demyelination,
Trem2–/– showed reduced upregulation. Thus, upon chronic although they were found at much lower levels than CE
demyelination, Trem2–/– microglia appear to be arrested in their (Figure 3D; interaction p < 0.01 for 12 week CPZ, two-way
transition to expression states seen in Trem2+/+ and Trem2+/– mi- ANOVA). Despite the significant accumulation of CE, brain
croglia and are unable to upregulate transcription of DAM genes, cholesterol levels remained unaltered (Figure 3E). Other neutral
including lysosome- and lipid metabolism-related genes, similar lipids, such as triacylglycerol (TG), were elevated in Trem2–/–
to reports of Trem2–/–:5XFAD microglia (Keren-Shaul et al., brain upon CPZ treatment (Figure 3F; interaction p < 0.05, two-
2017). Proper conversion to reactive states during chronic way ANOVA). Ganglioside GM3d38:1 and d40:1 were also
demyelination may enable lysosomal degradation and/or efflux increased in Trem2–/– brain with chronic demyelination (Fig-
of myelin lipids. ure 3G; interaction p < 0.05, two-way ANOVA), reminiscent of
lysosomal storage disorders. LCMS of mouse plasma did not
TREM2 Deficiency Causes Neuronal Damage during reveal any genotype- or CPZ-specific differences in cholesterol
Chronic Demyelination and CE levels (Data S3; two-way ANOVA). These data indicate
Neurite dystrophy occurs in CPZ-treated Trem2–/– mice (Cantoni that chronic demyelination causes a profound alteration of
et al., 2015; Poliani et al., 2015). Likewise, we found accumula- cholesterol metabolism selectively in Trem2–/– CNS.
tion of dystrophic APP-positive puncta in the hippocampus
and corpus callosum of Trem2–/– mice after 5 or 12 weeks of TREM2 Deficiency Causes CE Accumulation in Isolated
CPZ (Figures S3A and S3B). Quantification of APP-positive Microglia
dystrophic neurite puncta number, intensity, or area showed a The increase in CE found in Trem2–/– brain upon chronic demy-
significant interaction between genotype and treatment for the elination may be primarily intracellular, resulting from phagocy-
5 and/or 12 week CPZ diet (Figure S3C). Accordingly, plasma tosis of cholesterol-rich myelin by brain phagocytes and storage
neurofilament-light chain (Nf-L) levels were higher in CPZ- in lipid droplets after conversion of cholesterol to CE by ACAT.
treated Trem2–/– mice (Figure 3A; 12 week CPZ treatment-geno- Alternatively, if Trem2–/– phagocytes are unable to properly
type interaction p < 0.001, two-way ANOVA). Similarly, aged engulf myelin debris, the latter may accumulate in the interstitial
Trem2–/– plasma showed higher Nf-L levels (Figure S3D; age-ge- space and/or the cerebral spinal fluid (CSF), potentially gener-
notype interaction p < 0.05, two-way ANOVA), indicating that ating CE extracellularly. To test this, we developed cell-type-
TREM2 is neuroprotective. specific lipidomics via isolation of microglia (CD11b+/CD45low)
and astrocytes (ACSA2+) using FACS (Figure S4A) and collected
TREM2 Deficiency Causes CE Accumulation in the Brain CSF to assess circulating levels of lipids in the CNS.
Lipid metabolism-related genes are strongly induced upon As observed in forebrain, 12 week CPZ increased levels of
chronic demyelination in wild-type but not Trem2–/– microglia certain lipid species in microglia of all genotypes compared
(Figures 1E and 2D), including seven genes coding for proteins with untreated genotype controls, but Trem2–/– microglia ex-
directly involved in cholesterol transport (Apoe, Apoc1), hydroly- hibited more dramatic increases compared with Trem2+/+ and
sis of CE in lysosomes (Lipa), egress of cholesterol from lyso- Trem2+/– microglia (Figure 4A; Data S3; two-way ANOVA,
somes (Npc2), CE synthesis and storage in lipid droplets FDR < 0.05). There were no genotype effects in microglia without
(Soat1), CE hydrolysis in lipid droplets (Nceh1), and 25-hydroxyl- CPZ. Lipid changes were unique to microglia, as astrocytes or
ation (Ch25h). Thus, both intracellular and extracellular choles- CSF did not display any genotype- or CPZ treatment-specific al-
terol transport may be defective in Trem2–/– microglia with terations (Figures 4B and 4C; two-way ANOVA, FDR < 0.05).
CPZ. To test this, we conducted liquid chromatography-mass Lipids selectively increased in Trem2–/– microglia upon chronic
spectrometry (LCMS) analysis of lipids from coronal forebrain demyelination included many species dysregulated in the fore-
sections containing the corpus callosum. Brain lipidomic profiles brain (e.g., CE20:4, CE22:6) (Figures 4D and 4E; interaction p <
of the three genotypes under control diet were similar (Figure 3B; 0.001 for 12 week CPZ, two-way ANOVA), which were elevated
Data S3). Upon 5 week CPZ, minimal changes were detected, 8- to 20-fold in Trem2–/– microglia upon 12 week CPZ diet
with an enhancement in CE and oxCE levels in all three geno- compared with controls. Myelin-enriched lipids (Podbielska
types (Figure S3E). With 12 week CPZ, CE and oxCE lipid spe- et al., 2011) or metabolites thereof, such as hexosylceramide
cies were significantly elevated in Trem2–/– brain compared (HexCer) d18:1/22:0, d18:1/24:0, and d18:1/24:1, galactosylcer-
with Trem2+/+ and Trem2+/– brain (Figures 3B–3D; FDR < 0.05; amide (GalCer) d18:1/22:0 and d18:1/24:0, ceramide (Cer)
interaction p < 0.01 for 12 week CPZ, two-way ANOVA). Further d18:1/24:1, and sulfatide d18:1/24:0, d18:1/24:0h, and d18:1/
comparison revealed that Trem2–/– brain significantly accumu- 24:1, were significantly elevated upon 5 week CPZ treatment
lated CE species containing polyunsaturated fatty acids, such without genotype effects, suggesting comparable microglial
as CE22:6 (docosahexaenoic acid) and CE20:4 (arachidonic uptake of myelin upon acute demyelination (treatment effect
acid) (Figure 3C; interaction p < 0.0001, two-way ANOVA). p < 0.05 for 5 week CPZ, two-way ANOVA; Data S3). HexCer

842 Neuron 105, 837–854, March 4, 2020


A C
Cholesteryl esters
Plasma neurofilament light chain
100 *** *** ** **
1000 ***
Plasma Nf-L (pg/mL)

10

ng lipid/ug protein
Trem2 +/+
100 Trem2 +/-
1
Trem2 -/-

0.1
10
Control 5 week CPZ 12 week CPZ
0.01
B Trem2 +/+ +/- -/- Z-score
CE 18:1 CE 20:4 CE 20:5 CE 22:6
Control CPZ Control CPZ Control CPZ
D
Oxidized cholesteryl esters
100
*** ** **** **

pg lipid/ug protein 10

0.1

0.01
CE oxoODE CE HODE CE HpODE CE oxoHETE

E F
Cholesterol Triacylglycerols
1000 1
*
ng lipid/ug protein
ng lipid/ug protein

0.1

100 0.01
Cholesterol TG 58:8/22:6

G Gangliosides
100
* * **
Trem2 +/+ Control
Trem2 +/+ 5 wk CPZ
pg lipid/ug protein

10 Trem2 +/+ 12 wk CPZ


Trem2 +/- Control
Trem2 +/- 5 wk CPZ
1 Trem2 +/- 12 wk CPZ
Trem2 -/- Control
Trem2 -/- 5 wk CPZ
0.1
Trem2 -/- 12 wk CPZ
GM3 d36:1 GM3 d38:1 GM3 d40:1

Figure 3. TREM2 Deficiency Causes CE Accumulation in the Brain


(A) Neurofilament light chain (Nf-L) levels in plasma isolated from Trem2+/+, Trem2+/–, and Trem2–/– mice with control, 5 week, or 12 week CPZ diet. Genotype-
treatment interaction with 12 week CPZ: ***p < 0.001, two-way ANOVA; n = 7–16 mice per condition. Data represent median ± max/min.
(B) Heatmap of lipids significantly altered by genotype and/or 12 week CPZ in mouse forebrain. Two-way ANOVA, FDR < 0.05; columns represent individual mice;
n = 3–7 mice per condition.
(C–G) Concentration of (C) CE, (D) oxidized CE, (E) cholesterol, (F) triacylglycerol (TG), and (G) GM3 lipid species from mouse forebrain with control, 5 week, or
12 week CPZ diet. Data represent median ± max/min. Two-way ANOVA fitted for 5 week and 12 week CPZ diet; genotype-treatment interaction with 12 week
CPZ: *p < 0.05, **p < 0.01, and ***p < 0.001. No significant genotype-treatment interaction with 5 week CPZ diet.
See also Figure S3 and Data S3.

Neuron 105, 837–854, March 4, 2020 843


Microglia lipidomics Trem2 +/+ Control Trem2 +/- Control Trem2 -/- Control
A Trem2 +/+ +/- -/- Trem2 +/+ 5 wk CPZ Trem2 +/- 5 wk CPZ Trem2 -/- 5 wk CPZ
Ctl 5wk 12wk Ctl 5wk 12wk Ctl 5wk 12wk Z-score
Trem2 +/+ 12 wk CPZ Trem2 +/- 12 wk CPZ Trem2 -/- 12 wk CPZ

Microglia sterols Astrocyte sterols


D +
E
10 **** *** 10

pg lipid/cell

pg lipid/cell
1
1

0.1
0.1

0.01
0.01
10-5

10-10
0.001
Cholesterol CE 18:1 CE 20:4 CE 22:6 Cholesterol CE 18:1 CE 20:4 CE 22:6

F Microglia hexosylceramides G Astrocyte hexosylceramides


100 1000
*** **** **

100

fg lipid/cell
fg lipid/cell

10

10
B Astrocyte lipidomics
Trem2 +/+ +/- -/-
Ctl 5wk 12wk Ctl 5wk 12wk Ctl 5wk 12wk Z-score
1 1
d18:1/22:0 d18:1/24:0 d18:1/24:1 d18:1/22:0 d18:1/24:0 d18:1/24:1
H Microglia galactosylceramides I Astrocyte galactosylceramides
100 100
* *
10

fg lipid/cell
10
fg lipid/cell

1
0.1

0.01 0.1
d18:1/22:0 d18:1/24:0 d18:1/24:1 d18:1/22:0 d18:1/24:0 d18:1/24:1
J 100 Microglia ceramides
K Astrocyte ceramides
100
*** ***

10
fg lipid/cell
fg lipid/cell

10

C CSF lipidomics
Trem2 +/+ +/- -/- 1 0.1
Ctl 5wk 12wk Ctl 5wk 12wk Ctl 5wk 12wk d18:1/18:0 d18:1/24:0 d18:1/24:1 d18:1/18:0 d18:1/24:0 d18:1/24:1
L M
1000 Microglia sulfatides Astrocyte sulfatides
+ + 1000
** ** *
fg lipid/cell

100
fg lipid/cell

100

10
10

1 1
d18:1/24:0 d18:1/24:0h d18:1/24:1 d18:1/24:0 d18:1/24:0h d18:1/24:1

N Microglia gangliosides O Astrocyte gangliosides


100
* * 1000
fg lipid/cell

100
fg lipid/cell

10

10

1 1
Z-score GM3 d36:1 GM3 d38:1 GM3 d40:1 GM3 d36:1 GM3 d38:1 GM3 d40:1

Figure 4. TREM2 Deficiency Causes CE Accumulation in Isolated Microglia


(A) Heatmap of lipids significantly altered by treatment and/or genotype in microglia isolated from Trem2+/+, Trem2+/–, and Trem2–/– mouse brain upon control,
5 week, or 12 week CPZ diet. Two-way ANOVA, FDR < 0.05; columns represent individual mice; n = 6 mice per condition.
(B and C) Heatmap comparison of lipids detected in (B) astrocytes or (C) cerebral spinal fluid (CSF) isolated from Trem2 mice upon control, 5 week, or 12 week
CPZ diet. Columns represent individual mice; n = 5 or 6 mice per condition.
(D–O) Concentrations of significantly altered lipid species in (A) from microglia or astrocytes isolated from mouse brain with control, 5 week, or 12 week CPZ diet:
(D and E) sterols, including free cholesterol and CE, from (D) microglia or (E) astrocytes; (F and G) HexCer from (F) microglia or (G) astrocytes; (H and I) GalCer from

(legend continued on next page)

844 Neuron 105, 837–854, March 4, 2020


species, GalCer d18:1/24:0 and d18:1/24:1, Cer d18:1/18:0 and and 2.3-fold, respectively, compared with wild-type microglia,
d18:1/24:1, and sulfatides were further enriched in Trem2–/– mi- suggesting that CPZ increases the levels of most CE species
croglia compared with Trem2+/+ upon 12 week CPZ (Figures 4F– similarly in wild-type and Apoe–/– microglia, thus preserving ge-
4M; genotype-treatment interaction p < 0.05, two-way ANOVA). notype differences. Genotype-specific increases in CE were pre-
Additionally, GM3d36:1, GM3d38:1, and BMP36:2 were sent in sorted astrocytes (Figures 5E and 5F; Data S4; p < 0.001,
elevated in Trem2–/– microglia upon 12 week CPZ diet (Figures two-way ANOVA), with 2.1- and 4.5-fold increases in CE18:1 and
4N, 4O, S4B, and S4C), potentially indicating lysosomal dysfunc- CE22:6, respectively, in Apoe–/– astrocytes relative to wild-type
tion (Bissig and Gruenberg, 2013; Miranda et al., 2018). LCMS astrocytes with CPZ. Interaction between genotype and CPZ
analysis of CSF from the three genotypes with or without CPZ was observed for CE18:1 (p < 0.05). Remarkably, no changes
did not show any lipid changes (Figures S4D–S4H), suggesting were found for cholesterol in the forebrain or sorted glial cells
that lipid accumulation observed in bulk forebrain tissue does (Figures 5B, 5D, and 5F). Myelin lipids HexCer and sulfatides
not reflect extracellular accumulation, although changes in inter- were similarly increased in wild-type and Apoe–/– microglia
stitial fluid cannot be ruled out. Thus, Trem2–/– microglia are able with 12 week CPZ, but there were no treatment effects in astro-
to phagocytose myelin debris during demyelination but are un- cytes (Figures S5B and S5C; Data S4). Unlike in Trem2–/– CSF,
able to properly metabolize or mediate the efflux of myelin lipids. CEs were elevated in Apoe–/– CSF, pointing to a widespread in-
Microglia sorted from the brain of aged Trem2+/+ and Trem2–/– crease of these sterols in mutant brain (Figures S5D and S5E).
mice were also analyzed for lipid content, but no genotype-spe- Therefore, impaired cholesterol transport resulting from APOE
cific alteration of CE was found, consistent with minor alterations deficiency causes massive, primarily CPZ-independent, accu-
in cholesterol metabolism-related genes compared with CPZ- mulation of CE in forebrain, glial cells, and CSF. The fact that
treated Trem2–/– microglia (Figures S1G and S1H). Trem2–/– microglia exhibit similar CE storage and have lower
APOE levels suggests that the lipid phenotype stems from
APOE Deficiency Results in a Broader CE Accumulation cholesterol transport defects.
in the Brain
Apoe is one of the most striking TREM2-dependent damage- Myelin Sulfatide Binds TREM2 and Promotes
associated microglia genes (Götzl et al., 2019; Krasemann Downstream Signaling
et al., 2017; Parhizkar et al., 2019) and is commonly believed Next, we sought to delineate the mechanisms underlying lipid
to be the main cholesterol transporter in the CNS. Additionally, dysregulation in Trem2–/– cells in vitro. Myelin lipid accumulation
microglial responses in the Apoe–/– phenocopy those of in mutant cells could result from either increased phagocytosis
Trem2–/– in specific paradigms, such as phagocytosis of or reduced lipid clearance post-phagocytosis. Because
apoptotic neurons (Krasemann et al., 2017). APOE deficiency Trem2–/– myeloid cells generally show reduced phagocytosis
may thus also produce microglial CE accumulation with CPZ, re- (Kleinberger et al., 2014; Poliani et al., 2015), Trem2–/– microglia
flecting reduced CNS cholesterol transport. However, unlike lipid phenotypes are unlikely to reflect increased myelin phago-
TREM2, APOE is expressed in microglia and astrocytes, raising cytosis. Prior to investigating myelin phagocytosis in Trem2–/–
the possibility that APOE deficiency may more broadly affect cells, we assessed the ability of specific myelin lipids to bind
brain cholesterol metabolism. We subjected Apoe+/+ and and signal via TREM2, which may in turn regulate the phagocytic
Apoe–/– mice to a control or 12 week CPZ diet prior to LCMS an- clearance of myelin.
alyses of forebrain, isolated microglia, astrocytes, and CSF. Lack TREM2 binds to various lipids, including phosphatidylcholine
of APOE generally increased brain CE levels (Figure 5A; Data S4). (PC), sphingomyelin (SM), phosphatidylserine (PS), phosphati-
Levels of CE18:1 and CE22:6 were increased by 2.7- and 4-fold, dylinositol (PI), and sulfatide (Kober et al., 2016; Sudom et al.,
respectively, in Apoe–/– forebrain relative to wild-type forebrain 2018; Wang et al., 2015), some of which (e.g., sulfatide) are
with control diet. CPZ led to an increase in CE18:1, 20:4, and myelin enriched (Du and Grabowski, 2004). We generated lipo-
22:6 species in Apoe–/– versus Apoe+/+ forebrain (Figure 5B; somes harboring candidate TREM2 ligands composed of 70
main effect from two-way ANOVA, p < 0.001; FCs were 6.6, molar % PC with 30 molar % of a test lipid to assess TREM2
1.4, and 6.7, respectively), and APOE deficiency significantly stimulation. Upon lipid ligand binding, TREM2 recruits SYK via
exacerbated the treatment effects for CE18:1 and CE20:4 (geno- DAP12’s ITAM domain, leading to SYK phosphorylation (Hamer-
type-treatment interaction p < 0.05). In addition, levels of BMP man et al., 2006). We stably overexpressed human DAP12 in
40:8 and 44:12 were higher in the Apoe–/– forebrain, consistent the presence or absence of human TREM2 (hTREM2) in
with lysosomal defects (Figure S5A; p < 0.05). There was a geno- HEK293 cells (Figure S6A) and monitored phospho-SYK
type-specific increase in all CE species in sorted microglia (pSYK) levels as a readout for receptor activation, using a
(Figures 5C and 5D; Data S4; p < 0.0001, two-way ANOVA), TREM2 agonist antibody as a positive control. PS-, PI-,
with 3.2-, 6.9-, and 2.5-fold increases in CE18:1, CE20:4, and GalCer-, and sulfatide-containing liposomes increased pSYK
CE22:6, respectively, in Apoe–/– microglia relative to wild-type levels in TREM2/DAP12 HEK293 cells (Figure 6A). To examine
microglia on control diet. With CPZ, changes were 2.8-, 4.2-, lipid signaling via endogenous TREM2, we differentiated human

(H) microglia or (I) astrocytes; (J and K) ceramides from (J) microglia or (K) astrocytes; (L and M) sulfatides from (L) microglia or (M) astrocytes; and (N and O)
gangliosides from (N) microglia or (O) astrocytes. Data represent median ± max/min. Two-way ANOVA fitted for 5 week and 12 week CPZ diet; genotype-
treatment interaction with 5 (+p < 0.05) or 12 week CPZ diet (*p < 0.05, **p < 0.01, and ***p < 0.001). Independent genotype or treatment effects noted in Data S3.
See also Figure S4.

Neuron 105, 837–854, March 4, 2020 845


A Forebrain lipidomics C Microglia lipidomics E Astrocyte lipidomics
Apoe +/+ -/- Apoe +/+ -/- Apoe +/+ -/-
Ctl 12wk Ctl 12wk Z-score Ctl 12wk Ctl 12wk Z-score Z-score
3
Ctl 12wk Ctl 12wk 3
Sulfatide d181/ CE161 Sulfatide d181/240h
LPC180 2 CE226 2
Sulfatide d181/240
PCO 180/20 CE182 Sulfatide d181/241
PE361 1 CE204 1
LacCerd181/180
PG160_181 CE181 HexCerd181/241
PG181/181 0 PE386 0
HexCerd181/240
Sulfatide d181/ PE406 SMd181/240
HexCerd181/160 1 PE364 1
PEP 180/181
PA180_204 PE384 Sulfatide d181/180
PA181/181 CE205
2 HexCerd181/180 2
PA160_181 LacCerd181/241 PE386
PA180_181 HexCerd181/180
3 PE364 3
DG181/81 HexCerd181/241 PC361
PI180_181 LacCerd181/180 PEP 180/204
DG180_181 HexCerd181/240 CE204
PS180_181 Sulfatide d181/240 TG 587/204
Sulfatide d181/ Sulfatide d181/241 TG 546/204
PEP 180/181 Sulfatide d181/160 Stearic acid
PI180_204 Sulfatide d181/180h Palmitic acid
PI160_181 Sulfatide d181/180 Palmitoleic acid
PI181/181 Sulfatide d181/240h PA181_181
PC361 Sulfatide d181/241h PC386
PC362 Cerd181/160 PI180_181
Cholesteryl glucos SMd181/160 TG 585/204
SMd181/241 HexCerd181/160 LPG180
Cholesterol PG160_181 GM3d361
PEP 181/204 PI204/204 Sulfatide d181/180h
Cerd181/240 PI180_181 DG180/204
GM3d361 LBPA181/181 Sulfatide d181/241h
GM3d341 PG180_204 PI181/181
GM3d401 Cerd181/241 TG 542/180
PC406 PI181/181 TG 523/181
PC386 LPEP 180 CE HpODE
BMP204/204 PC386 PA180_204
CE204 PC364 DG181/181
PS180_204 TG 587/204 CE205
SMd181/240 LPC204 CE226
SMd181/160 LPC181 CE181
SMd181/180 TG 524/181 CE161
CE182 LPE181 CE182
CE181 LPC226 LBPA181/181
CE161 LPG180 GM3d381
CE205 PEP 180/181 PCO 180/20
CE226 PC361 PI160_226
BMP226/226 GM3d381 PEP 160/226
TG564/204 GM3d361 LPC180
LPI160 PEP 180/182 LacCerd181/241
LPI180 TG 546/204 LPC260
Sitosteryl glucosi PI160_226 Cerd181/240
DG160_181 PG180_181 Cerd181/160

B Forebrain sterols D Microglia sterols F Astrocyte sterols


1000 * * 10 * 10 *
#### ### #### #### #### ### ###
ng lipid/mg tissue

100
pg lipid/cell

1 pg lipid/cell 1

10

1 0.1 0.1
Cholesterol CE 18:1 CE 20:4 CE 22:6 Cholesterol CE 18:1 CE 20:4 CE 22:6 Cholesterol CE 18:1 CE 20:4 CE 22:6

Apoe +/+ Control Apoe -/- Control


Apoe +/+ 12 wk CPZ Apoe -/- 12 wk CPZ

Figure 5. APOE Deficiency Causes CE Accumulation in the Brain, Sorted Microglia, and Astrocytes
(A) Heatmap of top 50 lipid species (ranked by ANOVA p value) altered by genotype and/or 12 week CPZ diet in Apoe+/+ and Apoe–/– mouse forebrain.
(B) Concentration of free cholesterol and CE species from Apoe+/+ and Apoe–/– mouse forebrain extracts with control or 12 week CPZ diet.
(C) Heatmap of top 50 lipid species (ranked by ANOVA p value) altered by genotype and/or 12 week CPZ diet in Apoe+/+ and Apoe–/– sorted microglia.
(D) Concentration of free cholesterol and CE species from Apoe+/+ and Apoe–/– sorted microglia with control or 12 week CPZ diet.
(E) Heatmap of top 50 lipid species (ranked by ANOVA p value) altered by genotype and/or 12 week CPZ diet in Apoe+/+ and Apoe–/– sorted astrocytes.
(F) Concentration of free cholesterol and CE species from Apoe+/+ and Apoe–/– sorted astrocytes with control or 12 week CPZ diet.
In (B), (D), and (F), data represent median ± max/min (n = 6). Two-way ANOVA; genotype effects: ###p < 0.001 and ####p < 0.0001; genotype-treatment in-
teractions: *p < 0.05. Treatment effects are noted in Data S4. See also Figure S5.

peripheral blood monocytes into macrophages (Figure S6B). (RU) = 704, and KD = 5.6 mM, RU = 631, respectively (Figures
Cells showed liposome dose-dependent increases in pSYK 6D and 6E). In comparison, hTREM2 R47H showed reduced
levels selectively for sulfatide and PS (Figure 6B). Increased affinity and lower binding response (i.e., RU) for sulfatide
pSYK response to sulfatide liposomes was abolished by addition and PS liposomes: KD = 20 mM, RU = 191, and KD = 14 mM,
of recombinant hTREM2 but not hTREM1 ECD, confirming RU = 267, respectively, suggesting ligand specificity (Figures
TREM2 binding and signaling specificity (Figure 6C). 6D and 6E). Lower binding response was due to a faster off-
Certain TREM2 LOAD variants, including R47H, show rate of the interaction, leading to shorter residency of mutant
reduced affinity to lipids (Kober et al., 2016; Ulland and TREM2 on the lipid surface. Decreased affinity and response
Colonna, 2018; Wang et al., 2015). We characterized the bind- values observed with the R47H variant were accounted for
ing affinity and kinetics of sulfatide and PS to the ECD of wild- by 5-fold (sulfatide) and 2.4-fold (PS) faster off-rates and rela-
type and mutant TREM2 R47H (Figure S6C) with surface plas- tively similar on-rates (Figures S6D–S6G). These data show
mon resonance. hTREM2 exhibited similar binding affinity and that sulfatide binds and signals via TREM2 and that the
response at the highest analyte concentration to sulfatide- R47H LOAD variant is significantly impaired in its interaction
and PS-containing liposomes: KD = 6.8 mM, response units with this lipid.

846 Neuron 105, 837–854, March 4, 2020


A Liposome stimulation B Liposome titration
8 TREM2/DAP12 HEK293 cells 6
human macrophage
TREM2/DAP12
Sulfatide
6 DAP12 PS
pSyk FOB

4 PI

pSyk FOB
GalCer
4 IgG3
TREM2 agonist
2
2

0 0
-10 -5 0

t
is
PC

ol

SM

PE

PS

PI

er

3
tid

on
er

C
Log [mg/mL]

Ig
al

lfa
st

ag
G
le

Su

2
ho

EM
C

TR
C TREM2 ECD competition D Sulfatide liposome binding E PS liposome binding
8 human macrophage
* 800 800
* hTREM2 hTREM2
Binding response (RU)

Binding response (RU)


6 Stimulated hTREM2 R47H
600 KD 7uM 600 hTREM2 R47H
pSyk FOB

TREM2-ECD KD 6uM
4 400
TREM1-ECD 400

2 200 200 KD 14uM


KD 20uM
0 0 0
0 5 10 15 0 5 10 15
e

PS

PC

r
ffe
tid

TREM2 concentration (uM) TREM2 concentration (uM)


Bu
lfa
Su

5ng/mL M-CSF
F G
Vehicle pHrodo-myelin pHrodo-myelin phagocytosis
1.5
5ng/mL M-CSF
(normalized to WT 180 min)
Trem2 +/+

pHrodo intensity / cell

Trem2 +/+
1.0 Trem2 -/-

0.5
*
Trem2 -/-

0.0
0 50 100 150
Time (minutes)

pHrodo-myelin/cell mask/Hoechst

50ng/mL M-CSF
H I
Vehicle pHrodo-myelin pHrodo-myelin phagocytosis
50ng/mL M-CSF
1.5
(normalized to WT 180 min)
Trem2 +/+

pHrodo intensity / cell

Trem2 +/+
1.0 Trem2 -/-

0.5
Trem2 -/-

0.0
0 50 100 150
Time (minutes)

pHrodo-myelin/cell mask/Hoechst

(legend on next page)

Neuron 105, 837–854, March 4, 2020 847


High M-CSF Rescues Myelin Phagocytosis Defects in Next, we tested if CE accumulation in Trem2–/– BMDMs is due
Trem2–/– BMDMs to excess storage of myelin cholesterol by ACAT1, which con-
Because TREM2 binds to myelin lipids such as sulfatide, Trem2–/– verts free cholesterol to CE in the endoplasmic reticulum.
cells may be impaired in myelin uptake, although our in vivo CPZ Because myelin debris contain high levels of cholesterol but
data suggest that myelin lipid uptake is unchanged in Trem2–/– mi- low levels of CE, CE can accumulate in lipid droplets only if
croglia. To evaluate acute myelin uptake in vitro, we treated myelin-derived cholesterol escapes from lysosomes post-
Trem2+/+ and Trem2–/– BMDMs with pHrodo-conjugated myelin, phagocytosis. BMDMs were treated for 48 h with myelin and
after verifying loss of TREM2 protein in null cells (Figures S6H co-dosed with ACAT1 inhibitor (500 nM K604) (Ikenoya et al.,
and S6I). At low concentrations of pro-survival factor M-CSF 2007). LCMS analysis showed that K604 selectively rescues
(5 ng/mL), pHrodo-myelin phagocytosis was reduced in Trem2–/– the accumulation of most CE species in response to myelin in
BMDMs compared with Trem2+/+, consistent with previous both genotypes, suggesting myelin cholesterol is indeed stored
studies (Poliani et al., 2015) (Figures 6F and 6G). However, at as an esterified form in lipid droplets (Figure 7C; Data S5).
high concentrations of M-CSF (50 ng/mL), pHrodo-myelin phago- We subsequently asked if CE accumulation in Trem2–/–
cytosis was comparable for both genotypes (Figures 6H and 6I). BMDMs is specific to myelin phagocytosis or if other physiolog-
Thus, high levels of M-CSF may provide compensatory upregula- ically relevant uptake mechanisms, such as endocytosis of
tion of phagocytic pathways in Trem2–/– BMDMs and reveal that CE-containing lipoproteins, may cause a similar CE increase.
the phagocytosis defects in TREM2-deficient cells can be context First, we tested if oxidized low-density lipoprotein (oxLDL) binds
dependent. Importantly, although they are obtained from BMDMs, and stimulates TREM2, using low-density lipoprotein (LDL) as a
these data do not support a mechanism by which increased myelin control. Using our hTREM2/hDAP12 HEK293 line, we found that
binding or phagocytosis causes lipid accumulation in Trem2–/– mi- oxLDL elevates pSYK levels (Figure S7A). A dose-dependent
croglia in vivo, prompting further mechanistic analyses. increase in pSYK levels was found in oxLDL-treated human mac-
rophages (Figure S7B). The increase trended toward attenuation
CE Accumulation in Trem2–/– Cells Is Rescued by ACAT1 by pre-incubating oxLDL with high concentrations of recombi-
Inhibitor and LXR Agonist In Vitro nant hTREM2 ECD at 9 mM (p = 0.1, two-way ANOVA, Tukey
Because Trem2–/– BMDMs can normally phagocytose myelin in test) but not at lower concentrations such as those used in lipo-
high M-CSF, we tested if accumulation of neutral lipids, including some/hTREM2 competition experiments (3 mM) (Figures S7C
CE, occurs downstream of myelin phagocytosis in null cells. and 6C), consistent with the fact that oxLDL is known to bind
BMDMs were treated with 25 mg/mL myelin over 48 h, then and stimulate multiple immune receptors (Choi et al., 2017).
stained with Nile red to assess neutral lipid storage with fluores- When treated chronically with 50 mg/mL oxLDL, Trem2–/–
cence microscopy. Trem2–/– BMDMs exhibited neutral lipid BMDM exhibited an increase in Nile red stain (Figures S7D and
accumulation upon myelin treatment, as shown by an 3-fold in- S7E), which did not stem from increased oxLDL uptake by
crease in Nile red stain (Figures 7A and 7B). Lipidomics also re- Trem2–/– BMDMs, as indicated by comparable internalization
vealed prominent genotype-specific accumulation of CE18:2, of DiI-labeled oxLDL (Figure S7F). By LCMS, certain species of
CE20:4, and CE20:5 (Figure 7C; Data S5; p < 0.01, two-way CE, TG, HexCer, and cholesterol displayed genotype-specific in-
ANOVA). Cholesterol and various species of TG, diacylglycerol creases in both vehicle and oxLDL-treated conditions (Fig-
(DG), and myelin-derived HexCer also accumulated in Trem2–/– ure S7G; Data S5). K604 reduced levels of CE20:5 and CE22:6
BMDMs (Figure 7C). These changes were reminiscent of those in Trem2–/– BMDMs upon oxLDL exposure (Figure S7G; p <
observed in Trem2–/– microglia in vivo with CPZ diet (Figure 4). 0.05, Student’s t test), albeit less substantially than seen in

Figure 6. Myelin Sulfatide Binds TREM2 and Promotes Downstream Signaling


(A) Phospho-SYK (pSYK) fold change in TREM2/DAP12-HEK293 cells stimulated with indicated liposomes, normalized to buffer control (dotted line) and
compared with TREM2 agonist antibody and isotype control. N R 2 experimental replicates from two or more averaged technical replicates. PE, phosphati-
dylethanolamine. Data represent mean ± SEM.
(B) Liposome titration curve of pSYK fold changes in human macrophage cells from two to four donors upon stimulation with indicated test lipid, normalized to
buffer control, and compared with TREM2 antibody and isotype control.
(C) Liposome stimulation of indicated test lipid in human macrophage cells from four or five donors with liposomes only (stimulated) or liposomes with 3 mM
recombinant TREM2- or TREM1-extracellular domain (ECD) protein normalized to buffer control (dotted line). *p < 0.05, two-way ANOVA, Tukey test. Data
represent median ± max/min.
(D and E) Surface plasmon resonance binding response of increasing concentrations of wild-type (gray) and mutant R47H (orange) hTREM2 protein to (D) 30%
sulfatide/70%PC or (E) 30% PS/70% PC 100 nm liposomes.
(F) Trem2+/+ and Trem2–/– BMDM vehicle or pHrodo-myelin (red) phagocytosis with 5 ng/mL M-CSF. Green, cell membrane stain; blue, Hoechst. Scale
bar: 20 mm.
(G) Intensity of pHrodo-myelin (5 mg/mL) phagocytosis in Trem2+/+ and Trem2–/– BMDMs with 5 ng/mL M-CSF. Data represent mean ± SEM (n = 3 biological
replicates); *p < 0.05 comparing Trem2+/+ and Trem2–/– AUC, two-tailed t test.
(H) Trem2+/+ and Trem2–/– BMDM vehicle or pHrodo-myelin (red) phagocytosis with 50 ng/mL M-CSF. Green, cell membrane stain; blue, Hoechst. Scale
bar: 20 mm.
(I) Intensity of pHrodo-myelin (5 mg/mL) phagocytosis in Trem2+/+ and Trem2–/– BMDM with 50 ng/mL M-CSF. Data represent mean ± SEM (n = 3 biological
replicates).
See also Figure S6.

848 Neuron 105, 837–854, March 4, 2020


A BMDM Nile Red/DAPI B BMDM
Vehicle Myelin

*
Trem2 +/+
2500

Nile Red Spot Area


2000 Vehicle
Myelin
1500

1000
Trem2 -/-

500

0
Trem2 +/+ Trem2 -/-

C BMDM Sterols
CE 18:1 CE 18:2 CE 20:4 CE 20:5
## ## ##

10000 1000 100000 10000


** ** *
Adjusted Mean Abundance

Adjusted Mean Abundance

Adjusted Mean Abundance


Adjusted Mean Abundance
** 10000 1000 Vehicle
1000 100
**
1000 100 Myelin
100 10
100 10
Myelin + K604

10 1 10 1
+/+ -/- +/+ -/- +/+ -/- +/+ -/-

Triacyglycerols
CE 22:6 Cholesterol TG (52:3/18:1) TG (54:2/18:0)
### ###
###
10 10
Adjusted Mean Abundance

Adjusted Mean Abundance


100000 10000
**
Adjusted Mean Abundance

Adjusted Mean Abundance

10000
**
1000
1000 1 1
100

10
Interaction p-value: 0.017
1 100 0.1 0.1
+/+ -/- +/+ -/- +/+ -/- +/+ -/-

Diacyglycerols
DG (16:0/18:1) DG (18:0/18:1) DG (18:0/20:4) DG (18:1/18:1)
# ## ### #

10 10 100 10
Adjusted Mean Abundance

Adjusted Mean Abundance


Adjusted Mean Abundance

Adjusted Mean Abundance

10 *
1 1 1

0.1 0.1 0.1 0.1


+/+ -/- +/+ -/- +/+ -/- +/+ -/-

Hexosylceramides
HexCer (d18:1/16:0) HexCer (d18:1/18:0) HexCer (d18:1/24:0) HexCer (d18:1/24:1)
# ### ### ###

10 10 10 100
Adjusted Mean Abundance

Adjusted Mean Abundance

Adjusted Mean Abundance


Adjusted Mean Abundance

1 10

1 1

0.1 1

Interaction p-value: 0.029


0.1 0.01 0.1 0.1
+/+ -/- +/+ -/- +/+ -/- +/+ -/-
D iMG Sterols
CE 18:1 CE 20:4 CE 22:6 Cholesterol
# # ##

10000 10000
*
Adjusted Mean Abundance

* * *
Adjusted Mean Abundance
Adjusted Mean Abundance

Adjusted Mean Abundance

* * * * Vehicle
** * 10000 ** 10000 **
1000 Myelin
1000 1000 Myelin + K604
1000

100
100 100
Myelin + GW

10 10 10 100
+/+ -/- +/+ -/- +/+ -/- +/+ -/-

Figure 7. TREM2 Deficiency-Associated CE Accumulation Is Rescued by ACAT1 Inhibitor and LXR Agonist In Vitro
(A) Nile red stain of neutral lipids in BMDMs cultured from Trem2+/+ and Trem2–/– mice, treated with vehicle or 25 mg/mL purified myelin for 48 h. Scale bar: 50 mm.
(B) Quantification of total spot area of Nile red stain in vehicle or 25 mg/mL myelin-treated Trem2+/+ and Trem2–/– BMDMs. Data represent mean + SEM (n = 5
biological replicates); *p < 0.05; one-tailed t test for comparison between Trem2+/+ with myelin versus Trem2–/– with myelin.
(C and D) Quantification of Trem2+/+ and Trem2–/– (C) sterols, TG, diacylglycerols (DG), and HexCer from cultured BMDMs and (D) sterols from cultured iPSC-
derived microglia (iMG) treated with vehicle, myelin, or myelin with ACAT1 inhibitor K604 (500 nM) or LXR agonist GW3965 (10 mM) for 48 h. Batch-adjusted mean

(legend continued on next page)

Neuron 105, 837–854, March 4, 2020 849


myelin uptake experiments (Figure 7C), without altering choles- and metabolism. TREM2 LOF caused robust intracellular accu-
terol or other lipid levels (Figure S7G; Data S5). In contrast to mulation of a storage form of cholesterol, CE, which is also
the myelin uptake paradigm, ACAT1 is responsible for only a observed in glial cells lacking APOE, the main cholesterol trans-
fraction of CE accumulation in oxLDL-treated Trem2–/– BMDMs, porter in the CNS. This function of TREM2 was revealed by a
suggesting that a pool of CE accumulates in organelles other chronic CPZ challenge that exposed microglia to myelin debris,
than lipid droplets, likely lysosomes. which are enriched for cholesterol and signaling lipids, such as
Next, we sought a relevant in vitro model to extend our findings sulfatides. Lipid ligands are recognized by the TREM2 ECD
to human cells. We differentiated human iPSCs into microglia and mediate downstream signaling, which we hypothesize may
and established that the transcriptome is similar to that of pri- enable resolution of disease pathology within the damaged
mary human microglia (Figure S7H). We then genetically ablated CNS parenchyma.
TREM2 using CRISPR (Figure S7I) and found that TREM2–/– As shown by scRNA-seq analysis of sorted microglia, CPZ
iPSC-derived microglia (iMG) have a 20% decrease in leads to TREM2-dependent gene expression changes in a sub-
pHrodo-myelin uptake relative to wild-type iMG after a 4 h incu- set of microglia as part of a transition to DAM-like states related
bation (n = 4 technical replicates). Despite reduced myelin to those found in other disease models (Keren-Shaul et al.,
phagocytosis, there was a genotype-specific increase in 2017). This transcriptional program encompasses a gene
CE20:4, CE22:6, and cholesterol (Figure 7D; Data S5; CE, p < ‘‘toolbox’’ that may enable microglia to enhance not only their
0.05; cholesterol, p < 0.01 [two-way ANOVA]). As in BMDMs, chemotactic and phagocytic properties but also their lysosomal
the increase in CE, but not cholesterol, was abolished by the degradative capacity and ability to facilitate intracellular and
ACAT1 inhibitor (Figure 7D; p < 0.05, Student’s t test). extracellular cholesterol transport, including efflux from micro-
Drugs such as LXR agonists can reduce CE stores by upregu- glia. Failure to induce this transcriptional program in Trem2–/–
lating the expression of ABCA1 and ABCG1 transporters (Moore microglia results in lipid accumulation rather than phagocytic de-
and Tabas, 2011). We thus treated iMG with the LXR agonist fects per se, suggesting that impaired lipid metabolism may
GW3695 which enhanced the mRNA expression of ABCA1 (by represent a critical aspect of TREM2 LOF in chronic diseases,
10.3 ± 0.2 and 14.8 ± 2.1 fold in wild-type and knockout iMG, including Nasu-Hakola disease and LOAD. This maladaptive
respectively) and ABCG1 (by 46.6 ± 5.0 and 53.8 ± 2.9 fold in state leads to neuritic dystrophy and Nf-L release, a phenome-
wild-type and knockout iMG, respectively; n = 2 biological repli- non generally reflective of neurodegeneration (Blennow and Zet-
cates). GW3695 rescued CE accumulation in myelin-treated terberg, 2018). Whether LOAD-associated LOF TREM2 variants
TREM2+/+ and TREM2–/– iMG (Figure 7D; Data S5; TREM2+/+ (e.g., R47H) phenocopy the complete LOF remains to be
iMG, p < 0.01; TREM2–/– iMG, p < 0.05 [Student’s t test]). Our established.
data suggest that TREM2 deficiency causes cholesterol efflux Trem2–/– macrophages and microglia exhibit a severe CE
defects, leading to accumulation of an ACAT1 inhibitor-sensitive storage disorder reminiscent of macrophage foam cells in
pool of CE in human iMG. atherosclerotic lesions (Moore and Tabas, 2011). Cholesterol
dyshomeostasis in foam cells is generally associated with pro-
DISCUSSION inflammatory responses (Choi et al., 2017; Heneka et al., 2018;
Moore and Tabas, 2011). Familial cases of atherosclerosis linked
Growing evidence indicates that TREM2 signaling is necessary to mutations in the LIPA gene (encoding the lysosomal acid
for normal microglial functions, including control of proliferation, lipase) lead to a CE storage disorder, suggesting that accumula-
survival, energy metabolism, and gene expression (Deczkowska tion of this lipid is pathogenic, at least in lysosomes (Du and
et al., 2018; Ulland and Colonna, 2018; Yeh et al., 2017). Grabowski, 2004). CE accumulation may mediate toxicity via
Although a link has been established between TREM2 and lipid enzymatic or non-enzymatic generation of oxidized metabolites
metabolism, previous studies have focused primarily on lipids and lipid peroxidation products from the mono- or polyunsatu-
as candidate TREM2 ligands, in the form of either lipoprotein rated fatty acyl chains it harbors (Choi et al., 2017; Hutchins
particles or cell surface-exposed signals (Atagi et al., 2015; et al., 2011). Most of the brain cholesterol is found in myelin,
Bailey et al., 2015; Sudom et al., 2018; Ulland and Colonna, and thus excessive release of myelin debris in disease situations
2018; Wang et al., 2016; Yeh et al., 2016). Further clues that may cause cholesterol- or CE-induced toxicity, because choles-
TREM2 mediates microglial lipid metabolism stemmed from mi- terol cannot be efficiently catabolized and is deleterious to cells
croarray studies (Poliani et al., 2015). Recently, TREM2 was at high concentrations. Our in vivo studies indicate that the
shown to control blood cholesterol metabolism in obese mice cholesterol burden imposed upon microglia by chronic demye-
by modulating the macrophage transcriptome in adipose tissue, lination leads to severe CE storage in Trem2–/– microglia, which
further linking TREM2 to lipid metabolism (Jaitin et al., 2019). likely reflects impairment in the metabolic flux of cholesterol in
Here, we combined FACS-based CNS cell type isolation with these cells. By failing to upregulate multiple genes involved in
RNA-seq, scRNA-seq, and lipidomics to establish a key role of cholesterol transport (Apoe, Apoc1, Npc2) and metabolism
TREM2 signaling in the control of microglial cholesterol transport (Lipa, Nceh1, Ch25h) upon chronic phagocytic challenge,

and 95% confidence interval for each group, n = 3 biological replicates. Significant genotype effect (two-way ANOVA, comparing vehicle and myelin treatments in
Trem2+/+ and Trem2–/– cells): #p < 0.05, ##p < 0.01, and ###p < 0.001. Significant drug treatment effects within each genotype (Student’s t test): *p < 0.05
and **p < 0.01.
See also Figure S7 and Data S5.

850 Neuron 105, 837–854, March 4, 2020


Trem2–/– cells accumulate intracellular CE to the detriment of mi- benefits in LOAD and other diseases associated with lipid
croglial function. Reduced metabolic flux of CE may expose it to dysregulation.
oxidation, resulting in secondary accumulation of oxCE species,
as observed in Trem2–/– mouse brain. Whether oxCE species are STAR+METHODS
neurotoxic or contribute to inflammation as seen in atheroscle-
rotic lesions remains to be determined (Choi et al., 2017). Detailed methods are provided in the online version of this paper
Our in vitro studies showed that post-phagocytic conversion and include the following:
of myelin-derived cholesterol to CE by ACAT1, followed by its
storage in lipid droplets, underlies the molecular basis for CE d KEY RESOURCES TABLE
accumulation in Trem2–/– cells. Further mechanistic dissection d LEAD CONTACT AND MATERIALS AVAILABILITY
indicated that TREM2–/– iMG are defective in cholesterol efflux, d EXPERIMENTAL MODEL AND SUBJECT DETAILS
on the basis of the ability of an LXR agonist to fully alleviate B Animals
CE accumulation. Because TREM2 regulates microglial expres- B Cell Lines
sion of APOE, an apolipoprotein that can function both in lipid B Primary Cell Cultures
intake and lipid efflux (Shi and Holtzman, 2018; Tall, 2008), a d METHOD DETAILS
likely mechanism accounting for CE accumulation in Trem2–/– B Immunofluorescence
cells is reduced efflux of cholesterol onto APOE-containing B Neurofilament light detection
lipoproteins. This is consistent with our Apoe–/– CPZ studies, B Fluorescence activated cell sorting (FACS)
showing CE accumulation in mutant brain, sorted astrocytes, B RNA isolation, qPCR, QuantSeq library preparation,
and microglia. Trem2–/– and Apoe–/– mice thus share phenotypic and analysis
similarities, although APOE, unlike TREM2, is expressed in astro- B Single cell RNAseq library preparation
cytes and thus controls CNS cholesterol metabolism more B Single cell RNAseq data processing
broadly. APOE isoform-specific differences in the handling of B Single cell RNAseq cluster and expression analysis
lipids by glial cells may also account for the lower and higher B Brain sample collection and preparation for LCMS
genetic risk of APOE2 and APOE4 (relative to APOE3) to analysis
LOAD, respectively. The fact that human iPSC-derived APOE4/ B FACS lipid extraction
E4 astrocytes were recently reported to accumulate endolysoso- B CSF sample collection and preparation for LCMS
mal cholesterol suggests that cholesterol metabolism in glial analysis
cells could be implicated in LOAD (Lin et al., 2018). As several B LCMS analysis of lipids
other LOAD genes expressed in microglia mediate lipid meta- B Liposome preparation
bolism (e.g., CLU, ABCA7, SORL1), alteration of cholesterol B pSYK AlphaLISA
and CE metabolism in microglia may be a unifying mechanism B Recombinant purification of His tagged hTREM2 and
accounting for disease risk, perhaps via impacts on Ab, tau, hTREM2 R47H ECD
inflammation, or other AD-relevant pathogenic processes. B Lipid binding analysis using surface plasmon reso-
The initial characterization of AD pathology by Dr. Alzheimer nance (SPR)
noted accumulation of ‘‘adipose inclusions,’’ likely neutral lipids, B Myelin purification and phagocytosis
in glial cells from postmortem brain samples of patients with de- B Lipid challenge experiments in BMDMs and human
mentia (Foley, 2010). More recent studies have indicated the rele- iPSC-derived microglia
vance of CE in AD, as it accumulates in postmortem brain tissue d QUANTIFICATION AND STATISTICAL ANALYSIS
from patients and in the brain of PSEN1/APP mutant mice (Astarita d DATA AND CODE AVAILABILITY
et al., 2011; Chan et al., 2012; Morel et al., 2013) and other neuro-
degenerative models (Miranda et al., 2018). Importantly, genetic SUPPLEMENTAL INFORMATION

ablation and pharmacological inhibition of ACAT1, which lowers


Supplemental Information can be found online at https://doi.org/10.1016/j.
CE levels, is protective in amyloid and tauopathy models through neuron.2019.12.007.
various mechanisms, including autophagy activation (Bhattachar-
yya and Kovacs, 2010; Bryleva et al., 2010; Di Paolo and Kim, 2011; ACKNOWLEDGMENTS
Hutter-Paier et al., 2004; Puglielli et al., 2001; Shibuya et al., 2015).
However, with the exception of Shibuya et al. (2014), these studies We thank Rishi Rahkit and Nathan Moerke for help developing the pSYK assay
primarily examined the role of ACAT1 in neurons and did not test if and Connie Ha for help with RNA-seq. We thank Kimberly Scearce-Levie,
Zachary Sweeney, Nga Bien-Ly, and Jeff Kessler for critical reading of the
ACAT1 inhibitors decrease CE burden in microglia. Brain-pene-
manuscript. This study was funded by Denali Therapeutics.
trant LXR agonists have also been considered as therapeutics
on the basis of anti-inflammatory and amyloid-lowering properties, AUTHOR CONTRIBUTIONS
resulting from their ability to enhance cholesterol efflux. However,
they may present safety liabilities from broad impact on all cell K.M.M. and G.D.P. conceived the study. A.A.N., K.L., B.V.L., S.L., D.J.K., D.X.,
H.C., P.C.G.H., P.E.S., T.S., G.A., J.W.L., K.M.M., and G.D.P. designed exper-
types and peripheral lipid metabolism (Moutinho and Landreth,
iments. A.A.N., K.L., B.V.L., J.W., L.P., S.S.D., C.L., D.J.K., D.X., A.L., S.B.,
2017). Our study raises the possibility that enhancing TREM2 T.K.E., P.C.G.H., M.L., J.S., B.J.A., T.L., H.O.S., A.S., and G.A. performed ex-
function may selectively alleviate cholesterol and CE burden in periments. T.S. performed the QuantSeq analyses. S.L. performed the scRNA-
myeloid cells, including microglia, potentially providing therapeutic seq analyses. A.A.N., K.L., B.V.L., S.L., J.W., L.P., S.S.D., D.J.K., J.C.D.,

Neuron 105, 837–854, March 4, 2020 851


B.J.A., T.S., and G.A. analyzed data. L.P., T.L., J.W.L., P.E.S., S.B.P., and Deczkowska, A., Amit, I., and Schwartz, M. (2018). Microglial immune check-
R.J.W. edited the manuscript. A.A.N., K.L., B.V.L., S.L., T.S., G.A., K.M.M., point mechanisms. Nat. Neurosci. 21, 779–786.
and G.D.P. wrote the manuscript. Di Paolo, G., and Kim, T.W. (2011). Linking lipids to Alzheimer’s disease:
cholesterol and beyond. Nat. Rev. Neurosci. 12, 284–296.
DECLARATION OF INTERESTS Dobin, A., Davis, C.A., Schlesinger, F., Drenkow, J., Zaleski, C., Jha, S., Batut,
P., Chaisson, M., and Gingeras, T.R. (2013). STAR: ultrafast universal RNA-seq
All authors are paid employees and shareholders of Denali Therapeutics.
aligner. Bioinformatics 29, 15–21.
R.J.W. is a founder and member of the Board of Directors of Denali. Denali
has filed patent applications related to the subject matter of this paper. Doench, J.G., Fusi, N., Sullender, M., Hegde, M., Vaimberg, E.W., Donovan,
K.F., Smith, I., Tothova, Z., Wilen, C., Orchard, R., et al. (2016). Optimized
Received: December 24, 2018 sgRNA design to maximize activity and minimize off-target effects of
Revised: August 7, 2019 CRISPR-Cas9. Nat. Biotechnol. 34, 184–191.
Accepted: December 4, 2019 Du, H., and Grabowski, G.A. (2004). Lysosomal acid lipase and atheroscle-
Published: January 2, 2020 rosis. Curr. Opin. Lipidol. 15, 539–544.
Durinck, S., Moreau, Y., Kasprzyk, A., Davis, S., De Moor, B., Brazma, A., and
REFERENCES Huber, W. (2005). BioMart and Bioconductor: a powerful link between biolog-
ical databases and microarray data analysis. Bioinformatics 21, 3439–3440.
Astarita, G., Jung, K.M., Vasilevko, V., Dipatrizio, N.V., Martin, S.K., Cribbs,
Efthymiou, A.G., and Goate, A.M. (2017). Late onset Alzheimer’s disease ge-
D.H., Head, E., Cotman, C.W., and Piomelli, D. (2011). Elevated stearoyl-
netics implicates microglial pathways in disease risk. Mol. Neurodegener.
CoA desaturase in brains of patients with Alzheimer’s disease. PLoS ONE 6,
12, 43.
e24777.
Fabregat, A., Jupe, S., Matthews, L., Sidiropoulos, K., Gillespie, M., Garapati,
Atagi, Y., Liu, C.C., Painter, M.M., Chen, X.F., Verbeeck, C., Zheng, H., Li, X.,
P., Haw, R., Jassal, B., Korninger, F., May, B., et al. (2018). The Reactome
Rademakers, R., Kang, S.S., Xu, H., et al. (2015). Apolipoprotein E is a ligand
Pathway Knowledgebase. Nucleic Acids Res. 46 (D1), D649–D655.
for triggering receptor expressed on myeloid cells 2 (TREM2). J. Biol. Chem.
Foley, P. (2010). Lipids in Alzheimer’s disease: A century-old story.
290, 26043–26050.
Biochimica. et. Biophysica. Acta. 1801, 750–753.
Bailey, C.C., DeVaux, L.B., and Farzan, M. (2015). The triggering receptor ex-
Götzl, J.K., Brendel, M., Werner, G., Parhizkar, S., Sebastian Monasor, L.,
pressed on myeloid cells 2 binds apolipoprotein E. J. Biol. Chem. 290,
Kleinberger, G., Colombo, A.V., Deussing, M., Wagner, M., Winkelmann, J.,
26033–26042.
et al. (2019). Opposite microglial activation stages upon loss of PGRN or
Bemiller, S.M., McCray, T.J., Allan, K., Formica, S.V., Xu, G., Wilson, G.,
TREM2 result in reduced cerebral glucose metabolism. EMBO Mol. Med.
Kokiko-Cochran, O.N., Crish, S.D., Lasagna-Reeves, C.A., Ransohoff, R.M.,
11, e9711.
et al. (2017). TREM2 deficiency exacerbates tau pathology through dysregu-
Guerreiro, R., Wojtas, A., Bras, J., Carrasquillo, M., Rogaeva, E., Majounie, E.,
lated kinase signaling in a mouse model of tauopathy. Mol. Neurodegener
Cruchaga, C., Sassi, C., Kauwe, J.S., Younkin, S., et al.; Alzheimer Genetic
12, 74.
Analysis Group (2013). TREM2 variants in Alzheimer’s disease. N. Engl. J.
Benjamini, Y., and Hochberg, Y. (1995). Controlling the false discovery rate: a Med. 368, 117–127.
practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. A Stat.
Hamerman, J.A., Jarjoura, J.R., Humphrey, M.B., Nakamura, M.C., Seaman,
Soc. 57, 289–300.
W.E., and Lanier, L.L. (2006). Cutting edge: inhibition of TLR and FcR re-
Bhattacharyya, R., and Kovacs, D.M. (2010). ACAT inhibition and amyloid beta sponses in macrophages by triggering receptor expressed on myeloid cells
reduction. Biochim. Biophys. Acta 1801, 960–965. (TREM)-2 and DAP12. J. Immunol. 177, 2051–2055.
Bissig, C., and Gruenberg, J. (2013). Lipid sorting and multivesicular endo- Hammond, T.R., Robinton, D., and Stevens, B. (2018). Microglia and the brain:
some biogenesis. Cold Spring Harb. Perspect. Biol. 5, a016816. complementary partners in development and disease. Annu. Rev. Cell Dev.
Blennow, K., and Zetterberg, H. (2018). Biomarkers for Alzheimer’s disease: Biol. 34, 523–544.
current status and prospects for the future. J. Intern. Med. 284, 643–663. Heneka, M.T., McManus, R.M., and Latz, E. (2018). Inflammasome signalling in
Blondel, V.D., Guillaume, J.L., Lambiotte, R., and Lefebvre, E. (2008). Fast un- brain function and neurodegenerative disease. Nat. Rev. Neurosci. 19,
folding of communities in large networks. J. Stat. Mech. 2008, P1008. 610–621.
Bryleva, E.Y., Rogers, M.A., Chang, C.C., Buen, F., Harris, B.T., Rousselet, E., Hutchins, P.M., Moore, E.E., and Murphy, R.C. (2011). Electrospray MS/MS
Seidah, N.G., Oddo, S., LaFerla, F.M., Spencer, T.A., et al. (2010). ACAT1 gene reveals extensive and nonspecific oxidation of cholesterol esters in human pe-
ablation increases 24(S)-hydroxycholesterol content in the brain and amelio- ripheral vascular lesions. J. Lipid Res. 52, 2070–2083.
rates amyloid pathology in mice with AD. Proc. Natl. Acad. Sci. U S A 107, Hutter-Paier, B., Huttunen, H.J., Puglielli, L., Eckman, C.B., Kim, D.Y.,
3081–3086. Hofmeister, A., Moir, R.D., Domnitz, S.B., Frosch, M.P., Windisch, M., and
Cantoni, C., Bollman, B., Licastro, D., Xie, M., Mikesell, R., Schmidt, R., Yuede, Kovacs, D.M. (2004). The ACAT inhibitor CP-113,818 markedly reduces amy-
C.M., Galimberti, D., Olivecrona, G., Klein, R.S., et al. (2015). TREM2 regulates loid pathology in a mouse model of Alzheimer’s disease. Neuron 44, 227–238.
microglial cell activation in response to demyelination in vivo. Acta Ikenoya, M., Yoshinaka, Y., Kobayashi, H., Kawamine, K., Shibuya, K., Sato,
Neuropathol. 129, 429–447. F., Sawanobori, K., Watanabe, T., and Miyazaki, A. (2007). A selective
Carmona, S., Hardy, J., and Guerreiro, R. (2018). The genetic landscape of ACAT-1 inhibitor, K-604, suppresses fatty streak lesions in fat-fed hamsters
Alzheimer disease. Handb. Clin. Neurol. 148, 395–408. without affecting plasma cholesterol levels. Atherosclerosis 191, 290–297.
Chan, R.B., Oliveira, T.G., Cortes, E.P., Honig, L.S., Duff, K.E., Small, S.A., Ilicic, T., Kim, J.K., Kolodziejczyk, A.A., Bagger, F.O., McCarthy, D.J., Marioni,
Wenk, M.R., Shui, G., and Di Paolo, G. (2012). Comparative lipidomic analysis J.C., and Teichmann, S.A. (2016). Classification of low quality cells from single-
of mouse and human brain with Alzheimer disease. J. Biol. Chem. 287, cell RNA-seq data. Genome Biol. 17, 29.
2678–2688. Jaitin, D.A., Adlung, L., Thaiss, C.A., Weiner, A., Li, B., Descamps, H.,
Choi, S.H., Sviridov, D., and Miller, Y.I. (2017). Oxidized cholesteryl esters and Lundgren, P., Bleriot, C., Liu, Z., Deczkowska, A., et al. (2019). Lipid-associ-
inflammation. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 1862, 393–397. ated macrophages control metabolic homeostasis in a Trem2-dependent
Chong, J., Soufan, O., Li, C., Caraus, I., Li, S., Bourque, G., Wishart, D.S., and manner. Cell 178, 686–698.e14.
Xia, J. (2018). MetaboAnalyst 4.0: towards more transparent and integrative Jay, T.R., Hirsch, A.M., Broihier, M.L., Miller, C.M., Neilson, L.E., Ransohoff,
metabolomics analysis. Nucl. Acids. Res. 46, 486–494. R.M., Lamb, B.T., and Landreth, G.E. (2017). Disease progression-dependent

852 Neuron 105, 837–854, March 4, 2020


effects of TREM2 deficiency in a mouse model of Alzheimer’s disease. McCarthy, D.J., Campbell, K.R., Lun, A.T., and Wills, Q.F. (2017). Scater: pre-
J. Neurosci. 37, 637–647. processing, quality control, normalization and visualization of single-cell RNA-
Jiang, H., Lei, R., Ding, S.W., and Zhu, S. (2014). Skewer: a fast and accurate seq data in R. Bioinformatics 33, 1179–1186.
adapter trimmer for next-generation sequencing paired-end reads. BMC Mecca, C., Giambanco, I., Donato, R., and Arcuri, C. (2018). Microglia and ag-
Bioinformatics 15, 182. ing: the role of the TREM2-DAP12 and CX3CL1-CX3CR1 axes. Int. J. Mol. Sci.
Jonsson, T., Stefansson, H., Steinberg, S., Jonsdottir, I., Jonsson, P.V., 19, E318.
Snaedal, J., Bjornsson, S., Huttenlocher, J., Levey, A.I., Lah, J.J., et al. Miranda, A.M., Lasiecka, Z.M., Xu, Y., Neufeld, J., Shahriar, S., Simoes, S.,
(2013). Variant of TREM2 associated with the risk of Alzheimer’s disease. Chan, R.B., Oliveira, T.G., Small, S.A., and Di Paolo, G. (2018). Neuronal lyso-
N. Engl. J. Med. 368, 107–116. somal dysfunction releases exosomes harboring APP C-terminal fragments
Keren-Shaul, H., Spinrad, A., Weiner, A., Matcovitch-Natan, O., Dvir- and unique lipid signatures. Nat. Commun. 9, 291.
Szternfeld, R., Ulland, T.K., David, E., Baruch, K., Lara-Astaiso, D., Toth, B., Moore, K.J., and Tabas, I. (2011). Macrophages in the pathogenesis of athero-
et al. (2017). A unique microglia type associated with restricting development sclerosis. Cell 145, 341–355.
of Alzheimer’s disease. Cell 169, 1276–1290.e17. Morel, E., Chamoun, Z., Lasiecka, Z.M., Chan, R.B., Williamson, R.L.,
Kleinberger, G., Yamanishi, Y., Suárez-Calvet, M., Czirr, E., Lohmann, E., Vetanovetz, C., Dall’Armi, C., Simoes, S., Point Du Jour, K.S., McCabe,
Cuyvers, E., Struyfs, H., Pettkus, N., Wenninger-Weinzierl, A., Mazaheri, F., B.D., et al. (2013). Phosphatidylinositol-3-phosphate regulates sorting and
et al. (2014). TREM2 mutations implicated in neurodegeneration impair cell processing of amyloid precursor protein through the endosomal system.
surface transport and phagocytosis. Sci. Transl. Med. 6, 243ra86. Nat. Commun. 4, 2250.
Kober, D.L., Alexander-Brett, J.M., Karch, C.M., Cruchaga, C., Colonna, M., Moutinho, M., and Landreth, G.E. (2017). Therapeutic potential of nuclear re-
Holtzman, M.J., and Brett, T.J. (2016). Neurodegenerative disease mutations ceptor agonists in Alzheimer’s disease. J. Lipid Res. 58, 1937–1949.
in TREM2 reveal a functional surface and distinct loss-of-function mecha- Muffat, J., Li, Y., Yuan, B., Mitalipova, M., Omer, A., Corcoran, S., Bakiasi, G.,
nisms. eLife 5, e20391. Tsai, L.H., Aubourg, P., Ransohoff, R.M., and Jaenisch, R. (2016). Efficient
Krasemann, S., Madore, C., Cialic, R., Baufeld, C., Calcagno, N., El Fatimy, R., derivation of microglia-like cells from human pluripotent stem cells. Nat.
Beckers, L., O’Loughlin, E., Xu, Y., Fanek, Z., et al. (2017). The TREM2-APOE Med. 22, 1358–1367.
pathway drives the transcriptional phenotype of dysfunctional microglia in Paloneva, J., Manninen, T., Christman, G., Hovanes, K., Mandelin, J.,
neurodegenerative diseases. Immunity 47, 566–581.e9. Adolfsson, R., Bianchin, M., Bird, T., Miranda, R., Salmaggi, A., et al. (2002).
Kucukural, A., Yukselen, O., Ozata, D.M., Moore, M.J., and Garber, M. (2019). Mutations in two genes encoding different subunits of a receptor signaling
DEBrowser: interactive differential expression analysis and visualization tool complex result in an identical disease phenotype. Am. J. Hum. Genet. 71,
for count data. BMC Genomics 20, 6. 656–662.
Lee, C.Y.D., Daggett, A., Gu, X., Jiang, L.L., Langfelder, P., Li, X., Wang, N., Pandya, H., Shen, M.J., Ichikawa, D.M., Sedlock, A.B., Choi, Y., Johnson,
Zhao, Y., Park, C.S., Cooper, Y., et al. (2018). Elevated TREM2 gene dosage K.R., Kim, G., Brown, M.A., Elkahloun, A.G., Maric, D., et al. (2017).
reprograms microglia responsivity and ameliorates pathological phenotypes Differentiation of human and murine induced pluripotent stem cells to micro-
in Alzheimer’s disease models. Neuron 97, 1032–1048.e5. glia-like cells. Nat. Neurosci. 20, 753–759.
Leyns, C.E.G., Gratuze, M., Narasimhan, S., Jain, N., Koscal, L.J., Jiang, H., Parhizkar, S., Arzberger, T., Brendel, M., Kleinberger, G., Deussing, M., Focke,
Manis, M., Colonna, M., Lee, V.M.Y., Ulrich, J.D., and Holtzman, D.M. C., Nuscher, B., Xiong, M., Ghasemigharagoz, A., Katzmarski, N., et al. (2019).
(2019). TREM2 function impedes tau seeding in neuritic plaques. Nat. Loss of TREM2 function increases amyloid seeding but reduces plaque-asso-
Neurosci. 22, 1217–1222. ciated ApoE. Nat. Neurosci. 22, 191–204.
Leyns, C.E.G., Ulrich, J.D., Finn, M.B., Stewart, F.R., Koscal, L.J., Serrano, Podbielska, M., Levery, S.B., and Hogan, E.L. (2011). The structural and func-
J.R., Robinson, G.O., Anderson, E., Colonna, M., and Holtzman, D.M. tional role of myelin fast-migrating cerebrosides: pathological importance in
(2017). TREM2 deficiency attenuates neuroinflammation and protects against multiple sclerosis. Clin. Lipidol. 6, 159–179.
neurodegeneration in a mouse model of tauopathy. PNAS 114, 11524–11529. Poliani, P.L., Wang, Y., Fontana, E., Robinette, M.L., Yamanishi, Y., Gilfillan, S.,
Liao, Y., Smyth, G.K., and Shi, W. (2014). featureCounts: an efficient general and Colonna, M. (2015). TREM2 sustains microglial expansion during aging
purpose program for assigning sequence reads to genomic features. and response to demyelination. J. Clin. Invest. 125, 2161–2170.
Bioinformatics 30, 923–930. Praet, J., Guglielmetti, C., Berneman, Z., Van der Linden, A., and Ponsaerts, P.
Lin, Y.T., Seo, J., Gao, F., Feldman, H.M., Wen, H.L., Penney, J., Cam, H.P., (2014). Cellular and molecular neuropathology of the cuprizone mouse model:
Gjoneska, E., Raja, W.K., Cheng, J., et al. (2018). APOE4 causes widespread clinical relevance for multiple sclerosis. Neurosci. Biobehav. Rev. 47, 485–505.
molecular and cellular alterations associated with Alzheimer’s disease pheno- Puglielli, L., Konopka, G., Pack-Chung, E., Ingano, L.A., Berezovska, O.,
types in human iPSC-derived brain cell types. Neuron 98, 1141–1154.e7. Hyman, B.T., Chang, T.Y., Tanzi, R.E., and Kovacs, D.M. (2001). Acyl-coen-
Liu, L., Zhang, K., Sandoval, H., Yamamoto, S., Jaiswal, M., Sanz, E., Li, Z., zyme A: cholesterol acyltransferase modulates the generation of the amyloid
Hui, J., Graham, B.H., Quintana, A., and Bellen, H.J. (2015). Glial lipid droplets beta-peptide. Nat. Cell Biol. 3, 905–912.
and ROS induced by mitochondrial defects promote neurodegeneration. Cell Ransohoff, R.M. (2016). How neuroinflammation contributes to neurodegener-
160, 177–190. ation. Science 353, 777–783.
Lun, A.T., Bach, K., and Marioni, J.C. (2016a). Pooling across cells to normalize Robinson, M.D., and Oshlack, A. (2010). A scaling normalization method for
single-cell RNA sequencing data with many zero counts. Genome Biol. 17, 75. differential expression analysis of RNA-seq data. Genome Biol. 11, R25.
Lun, A.T., McCarthy, D.J., and Marioni, J.C. (2016b). A step-by-step workflow Safaiyan, S., Kannaiyan, N., Snaidero, N., Brioschi, S., Biber, K., Yona, S.,
for low-level analysis of single-cell RNA-seq data with Bioconductor. Edinger, A.L., Jung, S., Rossner, M.J., and Simons, M. (2016). Age-related
F1000Res. 5, 2122. myelin degradation burdens the clearance function of microglia during aging.
Martı́n, M.G., Pfrieger, F., and Dotti, C.G. (2014). Cholesterol in brain disease: Nat. Neurosci. 19, 995–998.
sometimes determinant and frequently implicated. EMBO Rep. 15, Sala Frigerio, C., Wolfs, L., Fattorelli, N., Thrupp, N., Voytyuk, I., Schmidt, I.,
1036–1052. Mancuso, R., Chen, W.T., Woodbury, M.E., Srivastava, G., et al. (2019). The
Masuda, T., Sankowski, R., Staszewski, O., Böttcher, C., Amann, L., Sagar, major risk factors for Alzheimer’s disease: age, sex, and genes modulate the
Scheiwe, C., Nessler, S., Kunz, P., van Loo, G., et al. (2019). Author correction: microglia response to Abeta plaques. Cell Rep. 27, 1293–1306.e6.
spatial and temporal heterogeneity of mouse and human microglia at single- Sayed, F.A., Telpoukhovskaia, M., Kodama, L., Li, Y., Zhou, Y., Le, D., Hauduc,
cell resolution. Nature 568, E4. A., Ludwig, C., Gao, F., Clelland, C., et al. (2018). Differential effects of partial

Neuron 105, 837–854, March 4, 2020 853


and complete loss of TREM2 on microglial injury response and tauopathy. maintains microglial metabolic fitness in Alzheimer’s disease. Cell 170, 649–
PNAS 115, 10172–10177. 663.e13.
Shi, Y., and Holtzman, D.M. (2018). Interplay between innate immunity and Wang, Y., Cella, M., Mallinson, K., Ulrich, J.D., Young, K.L., Robinette, M.L.,
Alzheimer disease: APOE and TREM2 in the spotlight. Nat. Rev. Immunol. Gilfillan, S., Krishnan, G.M., Sudhakar, S., Zinselmeyer, B.H., et al. (2015).
18, 759–772. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s dis-
ease model. Cell 160, 1061–1071.
Shibuya, Y., Chang, C.C., Huang, L.H., Bryleva, E.Y., and Chang, T.Y. (2014).
Inhibiting ACAT1/SOAT1 in microglia stimulates autophagy-mediated lyso- Wang, Y., Ulland, T.K., Ulrich, J.D., Song, W., Tzaferis, J.A., Hole, J.T., Yuan,
somal proteolysis and increases Ab1-42 clearance. J. Neurosci. 34, P., Mahan, T.E., Shi, Y., Gilfillan, S., et al. (2016). TREM2-mediated early micro-
14484–14501. glial response limits diffusion and toxicity of amyloid plaques. J. Exp. Med.
213, 667–675.
Shibuya, Y., Chang, C.C., and Chang, T.Y. (2015). ACAT1/SOAT1 as a thera-
Wickham, H. (2016). ggplot2: elegant graphics for data analysis (New York:
peutic target for Alzheimer’s disease. Future Med. Chem. 7, 2451–2467.
Springer).
Simes, R.J. (1986). An improved Bonferroni procedure for multiple tests of sig-
Wu, D., and Smyth, G.K. (2012). Camera: a competitive gene set test account-
nificance. Biometrika 73, 751–754.
ing for inter-gene correlation. Nucleic Acids Res. 40, e133.
Sudom, A., Talreja, S., Danao, J., Bragg, E., Kegel, R., Min, X., Richardson, J.,
Xu, C., and Su, Z. (2015). Identification of cell types from single-cell transcrip-
Zhang, Z., Sharkov, N., Marcora, E., et al. (2018). Molecular basis for the loss-
tomes using a novel clustering method. Bioinformatics 31, 1974–1980.
of-function effects of the Alzheimer’s disease-associated R47H variant of the
Yeh, F.L., Wang, Y., Tom, I., Gonzalez, L.C., and Sheng, M. (2016). TREM2
immune receptor TREM2. J. Biol. Chem. 293, 12634–12646.
binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facili-
Tall, A.R. (2008). Cholesterol efflux pathways and other potential mechanisms tates uptake of amyloid-beta by microglia. Neuron 91, 328–340.
involved in the athero-protective effect of high density lipoproteins. J. Intern.
Yeh, F.L., Hansen, D.V., and Sheng, M. (2017). TREM2, microglia, and neuro-
Med. 263, 256–273.
degenerative diseases. Trends Mol. Med. 23, 512–533.
Ulland, T.K., and Colonna, M. (2018). TREM2—a key player in microglial Yuan, P., Condello, C., Keene, C.D., Wang, Y., Bird, T.D., Paul, S.M., Luo, W.,
biology and Alzheimer disease. Nat. Rev. Neurol. 14, 667–675. Colonna, M., Baddeley, D., and Grutzendler, J. (2016). TREM2 haplodeficiency
Ulland, T.K., Song, W.M., Huang, S.C., Ulrich, J.D., Sergushichev, A., Beatty, in mice and humans impairs the microglia barrier function leading to decreased
W.L., Loboda, A.A., Zhou, Y., Cairns, N.J., Kambal, A., et al. (2017). TREM2 amyloid compaction and severe axonal dystrophy. Neuron 90, 724–739.

854 Neuron 105, 837–854, March 4, 2020


KEEP
YOUR
FINGER
ON
THE
PULSE

With Cell Press Reviews


Looking for authoritative reviews on the To propel your research forward, faster, turn to
forefront of science? Turn to Cell Press. Cell Press Reviews. Our insightful, authoritative
Our editors stay abreast of the latest reviews—published across the life sciences in
developments so they can commission our primary research and Trends journals—go
expert, cutting-edge reviews. beyond synthesis and offer a point of view.

Find your way


with Cell Press Reviews
www.cell.com/reviews
ll

Article
Oxidative Metabolism Drives Immortalization
of Neural Stem Cells during Tumorigenesis
François Bonnay,1 Ana Veloso,2 Victoria Steinmann,1 Thomas Köcher,3 Merve Deniz Abdusselamoglu,1,5 Sunanjay Bajaj,1
Elisa Rivelles,4,6 Lisa Landskron,1,7 Harald Esterbauer,4 Robert P. Zinzen,2 and Juergen A. Knoblich1,8,*
1Instituteof Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030 Vienna, Austria
2Systems Biology of Neurogenesis, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the
Helmholtz Association (MDC), 13125 Berlin, Germany
3Vienna Biocenter Core Facilities (VBCF), 1030 Vienna, Austria
4Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
5Present address: Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The

Rockefeller University, New York, NY, USA


6Present address: IHR Labor, Medical Diagnostic Laboratory, 1220 Vienna, Austria
7Present address: Netherlands Cancer Institute (NKI), 1066 CX Amsterdam, the Netherlands
8Lead Contact

*Correspondence: juergen.knoblich@imba.oeaw.ac.at
https://doi.org/10.1016/j.cell.2020.07.039

SUMMARY

Metabolic reprogramming is a key feature of many cancers, but how and when it contributes to tumorigenesis
remains unclear. Here we demonstrate that metabolic reprogramming induced by mitochondrial fusion can
be rate-limiting for immortalization of tumor-initiating cells (TICs) and trigger their irreversible dedication to
tumorigenesis. Using single-cell transcriptomics, we find that Drosophila brain tumors contain a rapidly
dividing stem cell population defined by upregulation of oxidative phosphorylation (OxPhos). We combine
targeted metabolomics and in vivo genetic screening to demonstrate that OxPhos is required for tumor
cell immortalization but dispensable in neural stem cells (NSCs) giving rise to tumors. Employing an in vivo
NADH/NAD+ sensor, we show that NSCs precisely increase OxPhos during immortalization. Blocking Ox-
Phos or mitochondrial fusion stalls TICs in quiescence and prevents tumorigenesis through impaired
NAD+ regeneration. Our work establishes a unique connection between cellular metabolism and immortali-
zation of tumor-initiating cells.

INTRODUCTION demonstrate defects in the tricarboxyic acid (TCA) cycle and


increased glycolytic metabolism in clear-cell renal cell carci-
Tumor cells are immortalized and display a number of character- noma (ccRCC) (Courtney et al., 2018). However, the same study
istic features that distinguish them from normal cells. Among also confirmed that, unlike ccRCC, brain- and lung-derived tu-
those, reprogramming of metabolic pathways is indisputable mors retain fully functional and efficient glucose oxidation
but only partially understood. Since Otto Warburg demonstrated (Courtney et al., 2018), suggesting that mitochondrion-depen-
that tumor cells preferentially ferment glucose into lactate in lieu dent bioenergetics can also be a key contributor of tumorigen-
of oxygen-enabled mitochondrial oxidative phosphorylation (the esis. It is now also well established that oxidative metabolism
Warburg effect) (Warburg, 1956; Warburg et al., 1927), energy can contribute to tumor cell proliferation by promoting biosyn-
metabolism in tumor cells has been under growing scrutiny. thesis of aspartate and subsequent metabolism of nucleotides
Although widely accepted for multiple tumor types and animal (Birsoy et al., 2015; Sullivan et al., 2015). However, when these
models (Flaveny et al., 2015; Patra et al., 2013), the Warburg ef- metabolic changes are required during tumorigenesis and how
fect is not as universal as assumed previously. In fact, carcinoma they can contribute to the immortalization process is unknown.
subtypes derived from the brain (Janiszewska et al., 2012), leu- Because the complex microenvironment and heterogeneity of
kocytes (Kuntz et al., 2017), pancreas (Franco et al., 2016), mammalian tumors make it difficult to address the precise timing
skin (Haq et al., 2013; Vazquez et al., 2013), lungs (Rao et al., and function of metabolic changes during the tumorigenesis pro-
2019; Ye et al., 2011), and ovaries (Pastò et al., 2014) rely on cess, we used a genetically defined Drosophila neural stem cell
oxidative phosphorylation (OxPhos) for retaining stemness of (NSC)-derived tumor model to overcome these caveats.
their cancerous cells. The complexity of metabolic reprogram- Drosophila NSCs and neuroblasts (NBs) have proven to be an
ming is illustrated by a recent report using isotope tracing to important model for understanding key developmental

1490 Cell 182, 1490–1507, September 17, 2020 ª 2020 Elsevier Inc.
ll
Article

processes like asymmetric cell division and temporal patterning duction and acidity rates, hallmarks of the Warburg effect, are
(Homem and Knoblich, 2012). During the larval stages of devel- only minimally changed. By single-cell RNA sequencing
opment, NBs form neurons and glia cells following highly repro- (scRNA-seq) profiling of brat tumors, we identified a distinct sub-
ducible lineages. NBs and their progeny do not migrate and are population of tumor cells with higher expression of respiratory
surrounded by a glial sheath, so they can be easily recognized enzymes. Our genetic data suggest that the oxidative population
and tracked in clonal analysis. Multiple cellular markers allow un- drives tumor formation, whereas the glycolytic population is sec-
ambiguous tracking of maturation in progenitors and differentia- ondary. We generated an in vivo metabolic nicotinamide adenine
tion in the daughter cells so NBs are particularly suitable for dinucleotide (NAD+) sensor to demonstrate that uncoupling of
studying aberrant cell behavior such as tumorigenesis (Jiang transcriptional and metabolic changes in the brat mutant type
and Reichert, 2014). Changes in metabolism profoundly influ- II NB population is rate limiting for their irreversible commitment
ence NB behavior (Homem et al., 2014), suggesting that bioen- to tumorigenesis. Finally, we show that uncontrolled mitochon-
ergetics and cell fate are tightly connected. drial fusion causes the rate-limiting metabolic changes observed
Based on their lineages, NBs are subdivided into type 0, type I, in the tumor population. Our data reveal an unexpected connec-
and type II. Type 0 NBs divide asymmetrically into one NB and tion between mitochondrial metabolism and cellular immortali-
one post-mitotic neuron (Karcavich and Doe, 2005; Ulvklo et al., zation and show that precise coordination between cell fate
2012). Type I NBs (NBIs) produce a ganglion mother cell (GMC) and metabolic changes is essential to avoid tumorigenesis.
that again divides into two post-mitotic neurons or glia cells
(Buescher et al., 1998). Type II NBs (NBIIs) generate transit-ampli- RESULTS
fying intermediate neural progenitors (INPs) (Bello et al., 2008;
Boone and Doe, 2008; Bowman et al., 2008) that divide asymmet- brat tNBs Undergo Oxidative Metabolism
rically 5–6 times, each time generating a GMC that ultimately Metabolic regulation is critical for the onset and termination of
gives rise to two neurons or glia cells. NBIIs can be uniquely iden- proliferation in Drosophila NSCs (Homem et al., 2014; Otsuki
tified based on their expression of the Ets transcription factor and Brand, 2018). To determine whether immortalization of
Pointed (PntP1) (Zhu et al., 2011) and by the absence of Asense Drosophila NSCs is associated with alterations of metabolic
(Ase), a neural precursor marker present in all other NB subtypes pathways, we re-analyzed the transcriptome of NBIIs and their
(Brand et al., 1993; Jarman et al., 1993). Right after division, each tumorigenic bratRNAi counterparts (tNBs) (Landskron et al.,
INP undergoes a stereotypical maturation process that can be 2018). We determined mRNA expression changes between
tracked by sequential expression of unique markers. In newly tNBs and NBIIs for a wide range of enzymes catalyzing the reac-
born INPs, the asymmetrically segregating RNA binding protein tions of gluconeogenesis, glycolysis, fermentation, the pentose
Brain tumor (Brat) post-transcriptionally inhibits the general NB phosphate pathway (PPP), the TCA cycle, OxPhos, and nucleo-
marker Deadpan (Dpn) (Reichardt et al., 2018). In a stereotypical tide and amino acid biosynthesis (Figure S1A). Interestingly, four
fashion, the INP then turns on Ase, followed by re-expressing glycolytic enzymes (Hex-A, Pfk, Ald, and Pgm) and the two
Dpn, before it re-enters the cell cycle. The presence of a transit- fermentation-promoting enzymes lactate dehydrogenase (LDH)
amplifying population greatly increases the number of neurons and pyruvate dehydrogenase kinase (PDK) were increased
generated by each NBII, and this might explain why NBIIs are more than 4-fold in tNBs. In contrast, most OxPhos-related en-
particularly susceptible to tumorigenesis when the asymmetric zymes were unchanged or downregulated. These gene expres-
cell division machinery or differentiation programs are altered sion data could be interpreted to mean that bratRNAi brain tumors
(Kelsom and Lu, 2012; Knoblich, 2010). show enhanced glycolytic activity at the expense of OxPhos, as
NBs mutant for brat give rise to abnormal INPs that constitu- already suggested for epithelial tumors in imaginal discs (Eichen-
tively express the Brat targets dpn and zelda (Reichardt et al., laub et al., 2018; de la Cova et al., 2014; Wang et al., 2016).
2018). Instead of initiating Ase expression and re-entering We challenged our observations by directly measuring meta-
mitosis, tumor-initiating cells enter a long transient cell cycle ar- bolic changes in brain tumors (Figure 1A). We measured the ox-
rest (Bowman et al., 2008). Only after 24–48 h do they undergo ygen consumption rate (OCR) and extracellular acidification rate
mitosis and irrevocably convert into immortal tumor NBs (tNBs) (ECAR) of bratRNAi and control brain cells (Figures 1B–1D; Fig-
(Caussinus and Gonzalez, 2005) that will form huge lethal and ures S1C and S1D). Although the OCR and ECAR were increased
transplantable brain tumors. Although all brain NBs become in brain tumor cells, surprisingly, the OCR/ECAR ratio was also
quiescent and differentiate or die during pupariation, tNBs can strongly increased, which is indicative of increased oxidative
sustain proliferation until adulthood (Betschinger et al., 2006; metabolism (Figure 1D). To confirm these initial results, we per-
Landskron et al., 2018), indicating that they lack major prolifera- formed a targeted metabolomics analysis by mass spectrom-
tion control mechanisms. How these tumor cells become etry. Among 22 measured intermediates of glycolysis, the TCA
immortal and why they resist metamorphosis-induced differenti- cycle, and PPP or amino acids (Figure 1E), we found six signifi-
ation remains a mystery. cantly increased metabolites in brat tumors. Three were interme-
Because our previous results indicated a strong connection diates of the TCA cycle (citrate, alpha-ketoglutarate, and ma-
between metabolism and growth control in the Drosophila brain late), whereas none of the eight glycolytic intermediates were
(Homem et al., 2014), we used the NBII lineage to investigate increased significantly. Lactate was increased slightly, but pyru-
metabolic reprogramming during tumorigenesis. Our results vate, the pivotal final glycolytic product, was the most upregu-
reveal that brat NSC-derived tumors exhibit increased TCA inter- lated metabolite we observed. Although bratRNAi tumors mostly
mediates and higher oxygen consumption, whereas lactate pro- consist of tNBs, whereas control brains contain a majority of

Cell 182, 1490–1507, September 17, 2020 1491


ll
Article

A B C D E

F
H

G
I

K L
J

M N O

(legend on next page)

1492 Cell 182, 1490–1507, September 17, 2020


ll
Article

neurons, the observed metabolic changes are most likely due to NAD(H) redox (SoNar) (Zhao et al., 2016). SoNar can bind to
alterations in the NB population. In mammals and Drosophila either NAD+ or NADH with specific conformations and excitatory
(Hall et al., 2012; Volkenhoff et al., 2015), neurons predominantly wavelengths, allowing us to follow the NAD+/NADH ratio by live
rely on OxPhos; therefore, observing higher TCA cycle interme- imaging. Because NADH consumption and NAD+ production are
diates and oxygen consumption in stem-cell-enriched tumor not specific for fermentation or OxPhos, we first determined their
brains is particularly intriguing. relative contribution to the SoNar readout. We used oxamate to
We next investigated the origin of TCA cycle intermediates in inhibit LDH, koningic acid to inhibit glyceraldehyde 3-phosphate
brat tumors. Normally, glucose is metabolized into TCA cycle in- dehydrogenase (GAPDH), and rotenone and antimycin A to
termediates via glycolysis and subsequent import of pyruvate inhibit ETC complex I and III on dissociated tNBs from bratRNAi
into mitochondria and its transformation into acetyl-coenzyme SoNar-expressing tNBs (Videos S1, S2, and S3; Figures 1J–
A (CoA). Tumor cells, however, can use other carbon sources, 1O). Although OxPhos inhibitors induced a clear drop in NAD+/
such as glutamine (DeBerardinis et al., 2007) or acetate (Ma- NADH ratio, oxamate and koningic acid had no visible effect (Fig-
shimo et al., 2014), via generation of alpha-ketoglutarate and ures 1J–1M, 1O). Interestingly, however, not all tNBs decreased
acetyl-CoA, respectively. their NAD+/NADH ratio upon rotenone or antimycin A treatment
We independently performed [U-13C]glucose, [U-13C]L-Gln, (Figure 1J, white arrow; Videos S2 and S3), indicating metabolic
and [U-13C]acetate labeling in control and tumor brains and heterogeneity among tumor cells. Our measurements were
analyzed the subsequent integration of 13C into glycolysis, confirmed by fluorescence-activated cell sorting (FACS) analysis
fermentation, and TCA cycle metabolites (Figures 1F–1I; Figures of SoNar activity after treating cells with LDH and OxPhos inhib-
S1E and S1F). Interestingly, incorporation of glucose into the itors (Figure S1B; Figures 1K, 1L, and 1O). Finally, we used oxa-
TCA cycle (citrate [M2]) was lower in tumor brains than in control mate, koningic acid, and rotenone on SoNar-expressing NBIs to
brains, whereas incorporation of glucose into lactate was higher measure the tumor specificity of these responses (Figures 1N
(Figures 1F and 1G). This is consistent with the higher LDH and and 1O); as expected, normal NBs decreased their NAD+/
PDK levels found in tNBs in bulk RNA sequencing (RNA-seq) NADH ratio upon oxamate but not rotenone treatment. Alto-
data (Figure S1A). Acetate was incorporated into the TCA cycle gether, these results indicate that most brat tNBs shift to OxPhos
only to a minor extent in control and brat tumor brains (citrate and, unlike their non-tumorigenic counterparts, do not rely on
[M2]) and therefore does not seem to be a main contributor to fermentation for their bioenergetic needs.
OxPhos in Drosophila brains (Figures S1E and S1F). Strikingly
however, Gln was predominantly incorporated into the TCA cy- Dividing tNBs Display an Enhanced Oxidative
cle in control and tumor brains (malate [M4]) (Figures 1H and Metabolism Transcriptomic Signature
1I). In control brains, 13C from Gln was rapidly diluted during a Mammalian tumors are known to be dynamic and increase their
second TCA cycle (citrate [M4] = 25%, citrate [M2] = 4%), indi- heterogeneity with growth (Lawson et al., 2018). In particular,
cating that these TCA intermediates were diverted toward other bioenergetic heterogeneity is considered to be essential for tu-
metabolic paths. In brat tumors, however, labeled TCA interme- mor progression and adaptability (Nakajima and Van Houten,
diates originating from 13C-Gln were recycled to a higher degree 2013). To test the heterogeneity of brat tumors, we performed
(citrate [M4] = 46%, citrate [M2] = 10%), suggesting that, in single-cell Switch Mechanism At the 5’ end of RNA Templates
brains tumors, Gln-derived TCA cycle intermediates play a 2 (Smart-Seq2) RNA-seq on 157 bratRNAi tumor and 93 control
more active role in OxPhos. central brain NBs sorted by FACS (STAR Methods; Figure S2A).
Altogether, these results indicate that brat tumors rely on Gln Although the sorting gate of control NBs was stringent enough to
rather than glucose to fuel OxPhos and that Gln-generated only collect NBIs and NBIIs (the largest and brightest cells), the
TCA cycle intermediates are better recycled in the TCA cycle sorting gate for tNBs was enlarged to account for the range of
in brat tumors than in control brains, further suggesting their pref- tNB cell sizes within the tumor (Figure S2B). Subsequently, 29
erential use of OxPhos. non-tumorigenic cells originating from NBI lineages were
To determine bioenergetics in brat tNBs at cellular resolution, sequenced but excluded from tumor samples based on NBI/
we generated transgenic flies expressing the sensor for the co- GMC markers (asehigh) or absence of the proliferative marker
enzymes NAD+ and NADH (reduced form of NAD+), sensor of (dpnlow) (Figure S2A).

Figure 1. brat tNBs Exhibit an Oxidative Metabolism


(A) Processing of control and tumor third-instar larva whole brains (WB) for metabolic profiling.
(B–D) Oxygen consumption rate (OCR; B), extracellular acidification rate (ECAR; C), and OCR/ECAR ratio of control and tumor brains (D).
(E) Fold changes of metabolites from glycolysis, the TCA cycle, and PPP or amino acids (AA) metabolic pathways between tumor and control brains, normalized
by all analyzed metabolites as measured by liquid chromatography (LC)/mass spectrometry (MS). Metabolites highlighted in boldface are significantly higher in
tumor versus control samples (unpaired [two-tailed] t test, p < 0.05).
(F and H) Schematic view of [U-13C]glucose-originated (F) and [U-13C]glutamine-originated (H) 13C incorporation into the TCA cycle.
(G and I) 13C incorporation of [U-13C]glucose (G) and [U-13C]glutamine (I) into glycolytic (G) and TCA cycle intermediates (G and I) in control (gray bars) and tumor
(blue bars) third-instar larva WBs.
(J–N) Live-imaging of type II (J and M) or type I (N) NB-expressed SoNar and RFPnuclear and (K and L) FACS analysis of dissociated tNBs from third-instar larva
tumor (J–M) or control (N) brains before and after treatment with oxamate, rotenone, or koningic acid.
(O) Quantification of the NAD+/NADH ratio from individual control and tNBs 10 min after rotenone or oxamate treatment.
Scale bars, 20 mm. See also Figure S1, Table S2, and Videos S1, S2, and S3.

Cell 182, 1490–1507, September 17, 2020 1493


ll
Article

A B

(legend on next page)

1494 Cell 182, 1490–1507, September 17, 2020


ll
Article

Initially, tNBs were analyzed separately using the Pseudotime already undergone changes to adapt to the hypoxic tumor
analytical tool (Monocle R package) and separated by unsuper- environment.
vised clustering into three clusters of 14 (cluster 1 [c1]), 49 (c2), To understand how OxPhos was maintained in c1 cells, we
and 94 (c3) cells (Figure 2A). Interestingly, c1 was distinguished analyzed the expression pattern of all genes annotated as ‘‘tran-
from all other tNBs by its proliferative signature: the G2 > M cell scription factors’’ (TFs) or ‘‘DNA_binding’’ in pseudotime across
cycle inducer string (stg) was among the top 10 clustering genes. all tNBs and displayed candidates with cluster-specific expres-
Furthermore, c1 tNBs also expressed higher levels of several key sion changes (Figure S2D). We tested the role of 26 candidate
OxPhos components (CG5214 [a-ketoglutarate dehydrogenase TFs with a clear association with c1 in bratRNAi tumorigenesis
E2], Mdh2, and the mitochondrial phosphate carrier [Mpcp]) (Figure S2E) and found that RNAi of the TFs E2F transcription
and two enzymes catalyzing formation of a-ketoglutarate (Fig- factor 1 (E2f1) and PAR-domain protein 1 (pdp1) led to partial
ure 2B; Figure S2C) from lysine (Sccpdh1) and glutamate rescue of the tumor, as monitored by the tumor-specific RFP
(Gdh). This analysis suggests that c1 tNBs are the most prolifer- signal in adult flies. Although E2f1 is a known cell cycle inducer
ative and associated with higher levels of enzymes involved in (Duronio et al., 1995), pdp1 is mostly known to regulate tran-
OxPhos (Figures 2D, 2E, and 2G). Additionally, the inducers of scriptional activity in muscle and circadian clock neurons (Cyran
G1 > S transition cyclin-E and E2f1 were less differentially ex- et al., 2003), and its RNAi did not affect normal NBII proliferation
pressed between the three clusters (Figures 2G–2I), suggesting (Figure S2F). These data suggest that pdp1 acts as a tumor-spe-
that increased OxPhos is specifically correlated with the G2 > cific transcriptional regulator of the most oxidative and prolifera-
M phase transition in tNBs. tive subpopulation of brat tumors.
c3, the cluster most transcriptionally distinct from c1, was
separated from other tNBs by its higher expression of gluta- brat Tumorigenesis Requires Aerobic Metabolism
thione-producing enzymes (GstD9 and GstE1) and unfolded pro- To test whether the observed metabolic changes are biologically
tein response chaperones (Hsp26 and Hsp22) (Figure 2B), sug- relevant for tumor growth, we designed a genetic screen for
gesting higher stress levels in these cells. Furthermore, LDH genes that are essential for tumor-induced adult lethality (Fig-
was higher in c3 compared with c1 and c2, probably indicating ure 3A). Compared with control flies, bratRNAi tumor-bearing flies
a distinct bioenergetic profile (Figure 2F). Finally, c2 expressed have a dramatically shorter lifespan (15 days after hatching).
intermediate levels of stg and a-KGDH-E2, indicating an inter- Depleting genes essential for NSC proliferation, such as dpn,
mediate or transient state between c1 and c3 (Figures 2D, 2E, within the bratRNAi tumor restores the lifespan almost completely
and 2G). Overall, these results reveal a striking heterogeneity to that of control flies. Therefore, we used the time when 50% of
among bratRNAi tumor cells and identify a minor population of the flies had died (LT50) as a readout for the degree of tumor
highly proliferative cells with higher OxPhos among a majority rescue that can be achieved by depletion of individual metabolic
of cells that appear to be less proliferative and high in reactive enzymes (Figure 3B).
oxygen species (ROS) detoxification and endoplasmic reticulum Using this assay, we performed an F2 genetic screen (Fig-
(ER)/mitochondrial stress. ure S3A) to test RNAi constructs targeting nearly all known en-
To compare the three tNB clusters with untransformed control zymes involved in gluconeogenesis, glycolysis, fermentation,
NBs, we analyzed tNBs together with normal control NBs using the TCA cycle, ETC, and PPP (Figures 3C, 3D, and 3F; Fig-
Monocle (Figure 2C). Intriguingly, cells in c1, which express high ure S3B) for their ability to rescue brat tumor-induced lethality.
levels of cell cycle regulators and OxPhos enzymes, were tran- Strikingly, no RNAi construct targeting gluconeogenic, glyco-
scriptionally closest to the majority of the control NBs, whereas lytic, or fermenting enzymes led to significant lifespan rescue.
c3 was furthest away. A small minority of control NBs did not In stark contrast, depletion of 10 TCA enzymes (Idh, Pdhb,
follow this general trend and clustered in close proximity to c3 Acon, aKGDH-E1, E2, E3, Mdh2, Scs-a, Scs-b, and Fum1) and
tNBs (Figure 2C). This subpopulation is not identified when four of the five electron transport chain (ETC) complexes (com-
NBs are clustered without tNBs and shows significant expres- plex I, III, IV, and V) (Figures S4C–S4H) each led to significant
sion differences in only three genes (bru3, troll, and Hr4), sug- rescue of the tumor-induced lethality.
gesting that this is a clustering artifact generated by the pres- To further show that a rescued lifespan was indeed correlated
ence of residual non-tNBs in the bratRNAi population. with tumor prevention, we assessed the tumor burden by visual-
Taken together, our data indicate that proliferative and oxida- izing the tumor-specific RFP signal of adult flies. As expected,
tive tNBs in c1 are generated first during tumorigenesis, whereas tumors depleted of glycolytic and fermentation enzymes had
the glycolytic cells in c2 and c3 represent later cells that have largely similar or even increased tumor burdens compared with

Figure 2. Dividing tNBs Display an Enhanced Oxidative Metabolism Transcriptomic Signature


(A) t-distributed Stochastic Neighbor Embedding (tSNE) plot of the 157 confirmed tNBs.
(A and B) Unsupervised clustering of tNBs using Monocle v2. Three clusters could be defined: c1 (14 cells, red), c2 (49 cells, blue), and c3 (94 cells, green). Shown
are the top 10 differentially expressed genes as determined by contrasting each cluster with all other cells in the sample.
(C) tSNE plot of the 157 confirmed tNBs (red, blue, and green dots) and the 93 confirmed control NBs (purple) together. The 4 NBIIs were determined by high
expression levels of pnt.
(D) Relative expression of lactate LDH, stg, and CG5214 among tumor and control NBs.
(E–I) Relative expression of CG5214, LDH, stg, cyclin-E, and E2f1 along pseudotime, as defined by the Monocle function plot_genes_in_pseudotime.
See also Figure S2.

Cell 182, 1490–1507, September 17, 2020 1495


ll
Article

A B C

D E

G
F

H I

J K

(legend on next page)

1496 Cell 182, 1490–1507, September 17, 2020


ll
Article

control tumors (with the notable exception of AldolaseRNAi of key TCA cycle and ETC enzymes were unchanged or even
tumors, which showed partial rescue) (Figure 3E; Figures S4A downregulated in tNBs (Figure S1A), we again analyzed our tran-
and S4B). In contrast, dpn- and OxPhos-deficient tumors scriptome data for other causes of metabolic reprogramming
showed no or very low tumor-specific signal, similar to control (Landskron et al., 2018) and focused on markers of key mito-
conditions, where no tumorigenesis was induced (Figures 3E chondrial functions (Figure 4A). Strikingly, the only mitochondrial
and 3G; Figures S4A and S4B). markers upregulated in tNBs were the Dynamin-like GTPases
These results could be explained if OxPhos enzymes were marf and opa1. Marf and Opa1 are required for mitochondrial
generally required for all NBII fitness and/or proliferation and, fusion and regulate fusion of the inner and outer mitochondrial
therefore, depleting them from the tumor would prevent its membranes, respectively (Westermann, 2010). Because mito-
growth. To rule out this possibility, we tested the same RNAi con- chondrial fusion is well known to increase OxPhos (Mishra and
structs for their effect on normal, non-transformed NBII lineages Chan, 2016) and causes cells to rely more heavily on OxPhos
(Figures S3C and S3D). As expected (Zacharioudaki et al., 2012), for energy production (Rossignol et al., 2004), we wanted to
dpn RNAi strongly reduced NBII progeny numbers. Interestingly, find out whether deregulation of these enzymes could explain
AldolaseRNAi led to the same phenotype, indicating that glycol- the observed metabolic transformations.
ysis is required for normal NBII lineage proliferation and explain- We investigated mitochondrial morphology in control NBIIs
ing its partial rescue of tumor formation. In striking contrast, RNAi and tNBs at the L3 stage using a mitochondrial GFP reporter
of the three TCA cycle enzymes aKGDH-E2, Scs-a, and Fum1 (GFPCOX8A) (Figure 4B). Mitochondria appeared to be numerous,
led to a non-significant slight increase in NBII progeny, and small, and round in control NBIIs and their immediate progeny,
RNAi of the ETC complex I, III, and IV top tumor-rescuing indicative of fragmented mitochondria. However, in most tNBs,
enzymes led to stable NBII progeny numbers, indicating that mitochondria were thicker and more elongated, indicating sus-
OxPhos is specifically required for tNB but not normal NBII pro- tained mitochondrial fusion events. A few tNBs, however, also
liferation. These results are in line with our previous work showed fragmented mitochondria, even within the tumor tissue.
showing that OxPhos is not required for non-tumorigenic NB It is well known that mitochondria are fragmented during mitosis
proliferation (Homem et al., 2014). Altogether, these results indi- (Yamano and Youle, 2011), and because those cells often occur
cate that tNBs, unlike healthy NBs, rely on oxygen-dependent in pairs, they seem to be early post-mitotic cells. To confirm the
bioenergetics to proliferate . presence of fused mitochondria in tNBs, we performed transmis-
To further demonstrate that brat tumors rely on oxygen to pro- sion electron microscopy (TEM) on dorsal sections of control and
liferate, we raised bratRNAi tumor-forming larvae under hypoxic bratRNAi central brains (Figure 4C). bratRNAi tNBs contained
conditions (5% O2) for 24, 48, or 72 h after 3 days of develop- longer mitochondria (2.3-fold in length and 2.8-fold in area)
ment. Strikingly, hypoxic incubation for 2 and 3 days led to compared with NBIIs (Figures 4D and 4E). Interestingly, fused
54% and 70% reduced tumor brain volume, respectively mitochondria were also observed in numbRNAi larval brain tu-
(Figures 3H and 3I). This oxygen dependence was tumor specific mors (Figure 4B, rightmost panels). Like bratRNAi, numbRNAi re-
because control NBIIs grown under the same conditions did not sults in formation of large, lethal, transplantable brain tumors
alter their progeny numbers. Thus, reduced oxygen levels do not (Bowman et al., 2008; Caussinus and Gonzalez, 2005). However,
impair NBII proliferation but can significantly impair brain tumor unlike brat tumors, numb tumors arise because of unrestricted,
growth (Figures 3J and 3K). enhanced Notch pathway activation (Bowman et al., 2008).
OxPhos can contribute to tumor cell proliferation by promoting Thus, formation of enlarged, elongated mitochondria is a general
biosynthesis of aspartate and subsequent metabolism of nucle- feature of Drosophila NSC-derived tumors that is independent of
otides (Birsoy et al., 2015; Sullivan et al., 2015) by the enzymes the tumor-inducing signaling pathway.
glutamate oxaloacetate transaminase 1 and 2 (Got1 and Got2). Next, to determine whether bratRNAi tNBs mitochondrial fusion
RNAi of both enzymes, however, did not rescue brat tumor for- resulted in increased OxPhos, we labeled dissociated control
mation (Figure S3E), suggesting that brat tumor OxPhos is not and tNBs with tetramethylrhodamine (TMRM), whose fluores-
required for this purpose. cence intensity correlates with mitochondrial membrane poten-
tial (Figures S6D–S6F). Expectedly, TMRM staining showed that
tNBs Undergo and Depend on Mitochondrial Fusion tNBs have significantly increased mitochondrial potential
Our data indicate a metabolic shift from glycolysis to OxPhos compared with control NBs, indicating that bratRNAi tNB-fused
when NBs become tumorigenic. Because the transcript levels mitochondria display increased ETC activity.

Figure 3. The TCA Cycle, OxPhos, and Normoxia Are Necessary for brat Tumorigenesis
(A and B) Lifespan and (B) 50% lethality time (LT50) of flies with no tumor (mCherryRNAi) and tumor-bearing (bratRNAi, mCherryRNAi) and rescued tumor-bearing
(bratRNAi, dpnRNAi) flies.
(C, D, and F) Lifespan and (D and F) LT50 of metabolic pathway-deficient tumor-bearing flies. Genes depletions highlighted in boldface significantly rescued
tumor-induced lethality versus control tumors (unpaired [two-tailed] t test, p < 0.05).
(E and G) Tumor-specific RFP signal of flies bearing glycolysis pathway-, PPP-, glutaminolysis-, TCA-, or ETC-deficient tumors.
(H and I) RFPnuclear, Miranda, and PH3 immunostaining and (I) tumor lobe volume estimation of third-instar larva tumor brains incubated for 24, 48, or 72 h in 5%
O2. Scale bars, 50 mm.
(J) RFPnuclear, Miranda, and PH3 immunostaining and (K) NBII progeny count (RFP+ Miranda+) of third-instar larva control brains incubated for 24, 48, or 72 h in 5%
O2. Scale bars, 10 mm.
Each data point corresponds to one independent brain lobe in (I) and (K). See also Figures S3 and S4 and Table S1.

Cell 182, 1490–1507, September 17, 2020 1497


ll
Article

A B

C
D

F
G

H I

(legend on next page)

1498 Cell 182, 1490–1507, September 17, 2020


ll
Article

To analyze the role of marf, opa1, and other mitochondrial reg- after brat depletion, imINPs already began to regrow and reiniti-
ulators in tumorigenesis, we analyzed their effect on brat tumor ate cell division (Bowman et al., 2008). At this point, restoring brat
lethality (Figures 4F and 4G) and tumor burden (Figures 4H and expression can no longer prevent tumor formation, indicating
4I; Figures S5D and S5E). Strikingly, marfRNAi was sufficient to that cell cycle re-entry irreversibly commits the abnormal INPs
fragment tNB mitochondria (Figure S5A), completely abrogate to tumorigenesis.
the tumor, and rescue tumor-induced lethality, whereas overex- To correlate metabolic changes with tumor commitment, we
pressing marf slightly increased the tumor burden. Depleting reversibly depleted Brat by RNAi for 24 h in flies also expressing
opa1 abrogated the tumor but did not rescue tumor-induced SoNar (Figure 5A). For this, we first used the detection wave-
lethality. Because adult escapers did not harbor a detectable tu- length for SoNar-NAD+ because the SoNar-NADH signal was
mor, this early lethality is likely due to effects of opa1 RNAi very sensitive to photobleaching and harder to detect (Figures
outside of the NBII lineage. In contrast, depleting drp1, a regu- 5A, 5B, and 5D, left panels). However, SoNar-NAD+ levels
lator of mitochondrial fission, did not affect tumor growth, indi- were robustly detected (second left panels) and allowed us to
cating that mitochondrial fragmentation is not essential for the distinguish two cell populations with a remarkably sharp bound-
tumorigenesis process. Interestingly, we also found that ses-B, ary in between. Although NBs, imINPs (white arrowheads), and
an ATP:ADP antiporter, and tko, a component of the mitochon- the first mature INPs (mINPs; yellow arrowheads) show relatively
drial small ribosomal subunit are essential for tumor growth (Fig- low NAD+ levels, later mINPs and all more differentiated progeny
ures 4F and 4G; Figures S5D and S5E). have higher NAD+ levels (Figure 5A, top panels). Interestingly,
We also analyzed the role of marf, opa1, and ses-B in normal 24 h bratRNAi NBII lineages showed the exact same profile,
NBIIs (Figures S5B and S5C). RNAi of any of these genes led with a sharp boundary between young and older tumor-initiating
to a similar or higher number of INPs compared with control cells, despite the fact that all of them mis-expressed dpn (red
NBII lineages. Altogether, these results indicate that mitochon- arrowheads). However, after 48 h and 72 h of bratRNAi, a strong
drial fusion is an essential component of tumorigenesis in the increase in NAD+ levels occurred in the oldest tumor-initiating
Drosophila brain and the key mechanism underlying metabolic cells (TICs) furthest away from the NB (Figures 5B, bottom
reprograming in tNBs. panels, blue arrowheads, and 5D, 5E, and 5G, blue arrowheads).
Importantly, NAD+ and NADH levels increased in 48 h and
Metabolic Reprogramming Coincides with 72 h bratRNAi TICs, as revealed by careful scanning of both
Immortalization in tNBs SoNar excitation lengths (Figure 5C). Furthermore, these bio-
To determine the precise role of metabolic reprogramming dur- energetically abnormal (NAD+very high) cells contained higher
ing tumor cell immortalization, we used SoNar to determine the levels of marf compared with the other TICs (Figure 5F; Fig-
metabolic state of each cell in the NBII lineage during tumorigen- ure S6A) and were the first TICs harboring fused mitochondria
esis. For this, we first determined the point of no return for tumor (Figure 5G, blue arrowheads). Importantly, marfRNAi almost
formation using temperature-sensitive brat RNAi. We used the completely abrogated formation of these bioenergetically
Temporal And Regional Gene Expression Targeting (TARGET) abnormal TICs (Figure S6B), suggesting that mitochondrial
system (McGuire et al., 2003), where expression of the upstream fusion generated this metabolic state. Finally, pdp1 was also
activating sequence (UAS)/Gal4 system is prevented by a ther- significantly enriched in NAD+very high cells (Figure S6C), indi-
mosensitive Gal80 (Gal80TS) inhibitor. This allowed us to induce cating that this TF may already be required for increasing
bratRNAi by a temperature shift and then re-express the Brat pro- OxPhos during tumor initiation. Altogether, these results suggest
tein by shifting back to the permissive temperature. that mitochondrial fusion and a bioenergetic switch precisely
After 24 h of brat RNAi (induced by inactivating Gal80TS at coincide with tumor cell immortalization in brat-deficient TICs.
29 C), NBIIs had generated several abnormal immature INPs
(imINPs) that continued to express Dpn because dpn translation OxPhos and Mitochondrial Fusion Are Required for
is not repressed by Brat (Figure 5A). However, at this stage, Tumor Cell Immortalization
these dysfunctional imINPs are cell cycle arrested in G2 To test whether the shift to OxPhos and the underlying fusion of
(Bowman et al., 2008) and will not form a tumor when brat mitochondria are indeed required for TIC immortalization, we
expression is restored (by restoring Gal80TS by shifting the larvae measured how a-KGDH-E2, UQCR-Q, and marf RNAi affected
back to 18 C) (Figures S5F and S5G). However, 48 h and 72 h the different stages of tumor initiation. After 24 h of bratRNAi,

Figure 4. tNBs Undergo and Depend on Mitochondrial Fusion


(A) Transcriptional changes between tumor and control NBIIs (extracted from Landskron et al., 2018). Gene transcripts highlighted in boldface and blue are
significantly higher in tumor versus control NBIIs (unpaired [two-tailed] t test, p < 0.05).
(B) Immunostaining of NBII-expressed mitochondrion-addressed GFP (GFPCOX8A), RFPnuclear, and PH3 in control NBIIs and brat and numb tNBs. Yellow and
white arrows label tNBs with fused and fragmented mitochondria, respectively. Scale bars, 10 mm.
(C) TEM of control NBIIs and tNBs. Mitochondria are highlighted in blue (NBs) or in green (INPs). Scale bars, 2 mm.
(D and E) Mitochondrial surface and length from one control and one tNBII.
(F) Lifespan and (G) LT50 of mitochondrial biology gene-deficient tumor-bearing flies, representative of three independent experiments. Gene depletions
highlighted in boldface significantly rescued tumor-induced lethality versus control tumors (unpaired [two-tailed] t test, p < 0.05).
(H and I) Tumor-specific RFP signal of flies bearing mitochondrial fusion-deficient or -enhanced tumors.
(I) Head tumor burden quantification. Each dot corresponds to the average of 5–10 heads. n = 3 except for opa1RNAi (n = 1) because of developmental lethality.
See also Figures S5A–S5E and Table S1.

Cell 182, 1490–1507, September 17, 2020 1499


ll
Article

A C

E F

Figure 5. tNB Start Changing Their Bioenergetics When They Immortalize


(A–C and G) Confocal images of NBII-expressed SoNar showing Dpn and ATP5A immunostaining in the control and bratRNAi NBII lineage after (A) 24 h, (B) 48 h,
and (D) 72 h of RNAi. Yellow stars, NBIIs; white and yellow arrowheads, imINPs and mINPs; red and blue arrowheads, young and older tumor-initiating cells (TICs);
white arrows, NAD+- and NADH-high cells. Scale bars, 10 mm.

(legend continued on next page)

1500 Cell 182, 1490–1507, September 17, 2020


ll
Article

a-KGDH-E2RNAi, UQCR-QRNAi, and marfRNAi, TICs behaved signaling pathways (Feitelson et al., 2015), inactivated cell cycle
similarly as control TICs and failed to repress dpn (Figure 6A). Af- checkpoints (Bertoli et al., 2013; Burkhart and Sage, 2008; Ho
ter 48 h of bratRNAi, control TICs immortalized and started prolif- and Dowdy, 2002), and evasion of apoptosis by expressing
erating (Figure 6B, second column, yellow arrowheads). In sharp anti-apoptotic factors (Fernald and Kurokawa, 2013). Metabolic
contrast, at the same time point, OxPhos- and marf-deficient reprogramming has been widely considered an adaptation to the
TICs stacked but failed to start proliferation as efficiently (Figures environment when the tumor has been formed or during metas-
6B–6D). After 72 h of bratRNAi, control tumor cells were largely tasis but has not been described as a primary feature of tumor
proliferative and had expanded beyond their original lineage in cell immortalization (DeBerardinis and Chandel, 2016). Our re-
the brain (Figure 6C). At the same time point, a-KGDH-E2-, sults indicate that a metabolic switch toward OxPhos and
UQCR-Q-, and marf-deficient TICs kept stacking and mostly increased NAD+ biogenesis driven by glutamine incorporation
failed to re-initiate cell division (Figures 6C and 6D). is rate limiting for the immortalization of stem cells during tumor
Finally, unrestrained bratRNAi-mediated tumorigenesis led to initiation in Drosophila brain tumors. Moreover, extensive fusion
highly proliferative (PH3+) gigantic brain expansion (Figure 6E). of mitochondria induces a change in OxPhos in TICs to trigger
In contrast, a-KGDH-E2-, UQCR-Q-, marf-, and sesB-deficient transition into tumor cells. Our data support the idea that, like
bratRNAi NBII lineages kept producing intermediate progenitors other changes of fate (Tatapudy et al., 2017), immortalization is
that mostly failed to re-initiate cell division (PH3 ) and did not a dramatic and irreversible event in a cell that requires abrupt
affect overall brain size. Unlike healthy NBII lineages, however, metabolic changes.
these NBII lineages failed to generate fully differentiated Unlike other described Drosophila tumor models, brat NSC-
(Miranda-negative) smaller-sized progeny. Instead, nonprolifer- derived tumors originate from a glycolytic lineage and adapt
ative TICs kept stacking in these NBII lineages that were no into oxidative cells strictly relying on oxygen and mitochondrial
longer clearly separated. NAD+ production. There is strong evidence supporting a key
Finally, to understand how increased OxPhos promotes tumor role of OxPhos in various mammalian tumor types (Franco
cell immortalization, we tested the possibility that release of ROS et al., 2016; Haq et al., 2013; Kuntz et al., 2017; Pastò et al.,
generated by OxPhos could be required to initiate tumorigen- 2014; Rao et al., 2019; Vazquez et al., 2013; Ye et al., 2011),
esis. Indeed, brat tumors exhibit significantly more ROS than including brain-derived tumors (Janiszewska et al., 2012), indi-
normal NBIIs (Figures S7A and S7B). Importantly however, cating that this tumorigenic feature is evolutionarily conserved
depleting ROS by treating larvae initiating brat tumors with the from arthropods to humans. The key role of OxPhos in tumor
ROS scavenger N-acetylcysteine for 72 h did not alter or pro- cell proliferation in Drosophila NSC-derived tumors is also sup-
mote tumor initiation (Figures S7C and S7D). Furthermore, over- ported by a recent report (van den Ameele and Brand, 2019).
expressing the Superoxide dismutase 1 and 2 similarly did not Although this work did not specifically address the role of meta-
affect bratRNAi tumor formation (Figures S7E and S7F). To further bolic reprogramming during tumor cell immortalization, together
decipher OxPhos function during tumorigenesis, we investigated our conclusions indicate a fundamental role of OxPhos in dereg-
whether OxPhos-dependent NAD+ metabolism was sufficient to ulation of cell proliferation programs.
sustain tumorigenesis independent of ATP synthesis. Strikingly, Intriguingly, a molecularly distinct switch to oxidative meta-
overexpressing the proton-pump-independent yeast mitochon- bolism has been described in Drosophila NSCs but in a very
drial NADH dehydrogenase Ndi1 (Sanz et al., 2010) in ATP syn- different context: when NSCs exit proliferation (Homem et al.,
thase-reduced brat tumors was sufficient to fully restore tumor- 2014). It has been described previously that NSCs transcription-
igenesis (Figures 7A–7D). These results suggest that NAD+ ally upregulate TCA and ETC enzymes during pupal stages and
metabolism, rather than ATP or ROS production from this that this is required for timely exit from the cell cycle. Although
increased oxidative metabolism, is primarily required by bratRNAi superficially similar, the metabolic switch during pupal stages
TICs to immortalize. Altogether, these results indicate that a is not accompanied by altered mitochondrial morphology and
metabolic switch to OxPhos and higher NAD+ metabolism is instead triggered by increased levels of TCA or ETC enzymes.
induced by uncontrolled mitochondrial fusion is the rate-limiting Furthermore, it has requirements for metabolic pathways that
step for irreversible immortalization of NSCs during tumor forma- are different from the one we describe here; although complex
tion (Figure 7E). II and flavoproteins fueling the ETC with reduced forms of flavin
adenine dinucleotide (FADH) are key to prevent NB regrowth and
DISCUSSION to end proliferation in normal NBs, they are fully dispensable for
tNB proliferation. Overall, these combined observations suggest
The potential to divide indefinitely and to ignore organismal pro- that two distinct pathways can increase OxPhos in NSCs and
liferation control signals is a key hallmark of tumor cells. Several that their relative role is context dependent. When NSCs exit
cellular processes have been shown to play a crucial role in the proliferation, they do so by increasing the levels of respiratory
transition: telomere stabilization by telomerase (Shay and enzymes and utilizing the complete ETC to increase ATP produc-
Wright, 2011), sustained and deregulated proliferation control tion. When immature TICs are immortalized, however, they do so

(D) SoNar-NAD+ intensity quantification of successive INPs or TICs relative to their originating NBIIs.
(E) Schematic representation of (B).
(F) qPCR analysis of dpn, marf, stg, alpha-KGDH-E2, and LDH in control INPs and in NAD+-low and NAD+-high TICs.
See also Figures S5F and S5G.

Cell 182, 1490–1507, September 17, 2020 1501


ll
Article

A NB24h > CD8::GFP,


mCherryRNAi bratRNAi, bratRNAi, bratRNAi, bratRNAi,
mCherryRNAi KGDH-E2RNAi marfRNAi UQCR-QRNAi
PH3 GFP Dpn PH3 GFP Dpn PH3 GFP Dpn PH3 GFP Dpn PH3 GFP Dpn

*
*

B NB48h > CD8::GFP,


mCherryRNAi bratRNAi, bratRNAi, bratRNAi, bratRNAi,
mCherryRNAi KGDH-E2RNAi marfRNAi UQCR-QRNAi
PH3 GFP Dpn PH3 GFP Dpn PH3 GFP Dpn PH3 GFP Dpn

C NB72h > CD8::GFP,


bratRNAi, bratRNAi, bratRNAi, bratRNAi,
mCherryRNAi KGDH-E2RNAi marfRNAi UQCR-QRNAi
PH3 GFP Dpn PH3 GFP Dpn PH3 GFP Dpn PH3 GFP Dpn D
15 p=0.0075
48h of RNAi
Dpn+ PH3+ per lineage

12 72h of RNAi

9
p=0.0029
6
p<0.0001 p=0.022
p=0.021
3 p=0.008 p<0.0001

0
mCherry KGDH-E2 marf UQCR-Q
NB II > RNAi-brat + RNAi-

E
NBII > RFPnuclear,
mCherryRNAi bratRNAi, bratRNAi, bratRNAi, bratRNAi, bratRNAi,
mCherryRNAi KGDH-E2RNAi marfRNAi UQCR-QRNAi sesBRNAi
PH3 RFP Mira

(legend on next page)

1502 Cell 182, 1490–1507, September 17, 2020


ll
Article

by enhancing mitochondrial membrane fusion and shortcutting transcriptionally upregulated genes in tNBs and their preferential
ETC complex II to critically increase mitochondrial NAD+ synthe- use of pyruvate for fermentation. Interestingly, the dispensability
sis. Interestingly, complex II has already been described to be of high LDH levels in tumors has also been observed recently in
non-essential for respirasome formation in other contexts squamous cell carcinoma (Flores et al., 2019). This raises the
(Jang and Javadov, 2018). question of why such high levels of tumor-expressed glycolytic
Our discovery that mitochondrial fusion and increased mito- and fermenting enzymes (HexA and LDH) transcript levels do
chondrial NAD+ production are key for triggering tumor cell not result in lactate accumulation or increased acidity. One pos-
immortalization and inducing mitosis is fundamentally unex- sibility suggested by other tumor models (Brisson et al., 2016;
pected and surprising. In fact, multiple previous reports have Faubert et al., 2017; Hui et al., 2017) is that a fraction of the
suggested the opposite (Lee et al., 2013; Peiris-Pagès et al., tNB LDH levels would catalyze the opposite reaction to create
2018; Senft and Ronai, 2016). However, mitochondrial dynamics pyruvate out of lactate and fuel the TCA cycle. Furthermore,
(fragmentation and fusion) have already been shown in a very the elevated levels of the PPP intermediates D-erythrose-4-
different system to be instructive and critical for cell fate decision phosphate and phosphoribosyl pyrophosphate observed in
between effector (fragmented mitochondria) and memory (fused brat tumors could reveal an enhanced nucleotide biosynthesis
mitochondria) T cells (Buck et al., 2016). In this case, however, in strategy to which high levels of glycolytic enzymes and PDK
memory T cells as well as in other biological contexts, such as in may also contribute.
neurons (Beckervordersandforth, 2017) or oxidative muscle fi- Our results shed light on uncharacterized events during the
bers (Mishra and Chan, 2016), the combination of mitochondrial long transition of a TIC converting into an ectopic tNB. Other
fusion and increased OxPhos has been associated with longer- NB factors have been implicated in the earliest stages that
lived and non-dividing cells. Only a recent report in fibroblasts initiate the tumorigenesis process (such as dpn and zld in a
(Yao et al., 2019) associated mitochondrial fusion and increased brat context; Reichardt et al., 2018). Although these factors
OxPhos with actively dividing cells, which, in this case, provides can undoubtedly affect tumorigenesis, they also have important
enhanced aspartate production. Therefore, our study provides functions in normal NSC proliferation and development and,
the first strong evidence showing that OxPhos and mitochondrial therefore, cannot fully and unambiguously explain the tumori-
fusion can be potent proliferative mechanisms in multiple cellular genic process. In contrast, our results show a strictly tumor-spe-
contexts. cific process that is independent of the tumor-triggering muta-
How could this increased oxidative and NAD+ metabolism tion and that regulates the later step of tumor initiation.
contribute to tumor cell immortalization? First, our results indi- OxPhos is not required for dpn mis-expression in brat-deficient
cate that Got1 and Got2 are not required for tumorigenesis. TICs; therefore, inhibiting OxPhos did not rescue the accumula-
This argues against a key role of TICs’ increased OxPhos and tion of TICs. Instead, TICs switched to OxPhos only later, when it
NAD+ pool to promote biosynthesis of aspartate and subsequent was required for their immortalization and conversion into tNBs.
anabolism of nucleotides. More generally, it seems unlikely that Is OxPhos only required during tumor initiation or can it also
the main role of increased OxPhos in TICs is to promote anabo- play a broader role during tumorigenesis? Our results indicate
lism because brat tumor cells divide at a similar or even slower that established tumors are oxidative even at later stages and
pace than normal type II NBs (Homem et al., 2014). that reducing oxygen concentration can prevent the growth of
Instead, it is tempting to speculate that the increased NAD+ pool already established tumors. Furthermore, scRNA-seq clearly
observed in TICs is required for signaling during the long quiescent associated the most proliferative cell population with higher tran-
period enabling TIC immortalization. Indeed, NAD+ is a well- scriptional levels of OxPhos-associated genes. This strongly ar-
described co-factor regulating poly(ADP-ribose) polymerase gues that this bioenergetic switch has to be maintained long after
(PARP)-mediated poly-ADP ribosylation and Sirtuin-mediated de- tumor initiation to sustain growth of the tumor.
acetylation, which are known to influence chromatin plasticity, Finally, what are the molecular mechanisms driving mitochon-
genome stability, and cell fate (Hottiger, 2015). It will therefore drial fusion and the oxidative metabolic switch of the immortal-
be of utmost interest to investigate the potential role of NAD+ in izing tumor cells? It seems unlikely that Brat or Notch pathway
these pathways specifically during tumor cell immortalization. direct transcriptional targets are responsible for these changes;
Importantly, our data based on RNAi cannot completely most of them are widely expressed and required in NBs, which
exclude a role of glycolysis and fermentation in the tumor pro- undertake very distinct metabolic routes. Instead, it seems
cess. However, the higher tumor burden observed upon more likely that TICs develop their own transcriptional program
GAPDH, PGK, or LDH depletion argues that they would be detri- during their long quiescent phase of cellular transformation to
mental to tumor development despite LDH being one of the most induce this metabolic change. Identifying these regulatory

Figure 6. OxPhos and Mitochondrial Fusion Are Required For Tumor Cell Immortalization
(A–C) Immunostaining of GFP, Dpn, and PH3 in mCherryRNAi, bratRNAi and TCA cycle-deficient, marf-deficient and ETC-deficient bratRNAi NBII lineages after (A) 24
h, (B) 48 h, and (C) 72 h of RNAi. Red arrowheads, Dpn misrepression; yellow arrowheads, PH3+ TICs.
(D) Quantification of PH3+ cells per NBII lineage after 48 h (orange) and 72 h (red) of TCA cycle-deficient, marf-deficient, and ETC-deficient bratRNAi. Scale
bars, 10 mm.
(E) WB lobe imaging of RFPnuclear, Miranda, and PH3 in the no-tumor control, TCA cycle-deficient tumors, and marf-deficient, ETC-deficient, and sesB-deficient
tumors. Yellow dashed line, RFP+ tumor area.
Scale bars, 50 mm. See also Figure S6.

Cell 182, 1490–1507, September 17, 2020 1503


ll
Article

B C

(legend on next page)

1504 Cell 182, 1490–1507, September 17, 2020


ll
Article

mechanisms and analyzing their significance for mammalian tu- in the J.A. Knoblich laboratory is supported by the Austrian Federal Ministry of
mor cell immortalization will be of utmost interest. Education, Science and Research; the Austrian Academy of Sciences, the
Austrian Science Fund (Z_153_B09), and the City of Vienna and The Austrian
Science Fund (grants SFB F778 and DOC 72-B27, respectively). This project
STAR+METHODS has received funding from the European Research Council under the European
Union Horizon 2020 Research and Innovation Program (grants 695642,
Detailed methods are provided in the online version of this paper 693184, and 874769) and under the FP7 Program (grant 250342). F.B. was
and include the following: supported by a European Molecular Biology Organization long-term fellowship
(LTF 1280-2014). The VBCF Metabolomics Facility is funded by the City of
d KEY RESOURCES TABLE Vienna through the Vienna Business Agency.
d RESOURCE AVAILABILITY
B Lead Contact AUTHOR CONTRIBUTIONS

B Materials Availability
Conceptualization, F.B., A.V., R.P.Z., and J.A.K.; Methodology, F.B., A.V.,
B Data and Code availability V.S., L.L., T.K., R.P.Z., and J.A.K.; Investigation, F.B., A.V., V.S., M.D.A.,
d EXPERIMENTAL MODEL AND SUBJECT DETAILS S.B., E.R., T.K., and R.P.Z.; Resources, T.K., R.P.Z., and H.E.; Writing – Orig-
B Fly lines inal Draft, F.B. and J.A.K.; Writing – Review & Editing, F.B., R.P.Z., and J.A.K.;
B Fly husbandry Supervision, R.P.Z. and J.A.K.
d METHOD DETAILS
B Cloning DECLARATION OF INTERESTS
B Seahorse metabolic analysis
The authors declare no competing interests.
B Mass-spectrometry targeted metabolomics
B Ex vivo live imaging of SoNar Received: June 17, 2020
B Single-cell RNA-sequencing Revised: June 24, 2020
B RNA extraction, reverse-transcription and qPCR Accepted: July 28, 2020
analysis Published: September 10, 2020
B Lifespan and in vivo tumor burden analysis
REFERENCES
B Hypoxic incubations
B N-acetylcysteine treatments
Beckervordersandforth, R. (2017). Mitochondrial Metabolism-Mediated Regu-
B Immunofluorescence lation of Adult Neurogenesis. Brain Plast. 3, 73–87.
B ROS detection (Cell-ROX) Bello, B.C., Izergina, N., Caussinus, E., and Reichert, H. (2008). Amplification
B Mitochondrial membrane potential assay of neural stem cell proliferation by intermediate progenitor cells in Drosophila
B Transmitted electron microscopy brain development. Neural Dev. 3, 5.
d QUANTIFICATIONS AND STATISTICAL ANALYSIS Bertoli, C., Skotheim, J.M., and de Bruin, R.A.M. (2013). Control of cell cycle
transcription during G1 and S phases. Nat. Rev. Mol. Cell Biol. 14, 518–528.
SUPPLEMENTAL INFORMATION Betschinger, J., Mechtler, K., and Knoblich, J.A. (2006). Asymmetric segrega-
tion of the tumor suppressor brat regulates self-renewal in Drosophila neural
Supplemental Information can be found online at https://doi.org/10.1016/j. stem cells. Cell 124, 1241–1253.
cell.2020.07.039. Birsoy, K., Wang, T., Chen, W.W., Freinkman, E., Abu-Remaileh, M., and Sa-
batini, D.M. (2015). An Essential Role of the Mitochondrial Electron Transport
ACKNOWLEDGMENTS Chain in Cell Proliferation Is to Enable Aspartate Synthesis. Cell 162, 540–551.
Boone, J.Q., and Doe, C.Q. (2008). Identification of Drosophila type II neuro-
We would like to dedicate this paper to Suzanne Eaton, one of the greatest blast lineages containing transit amplifying ganglion mother cells. Dev. Neuro-
Drosophila biologists, who inspired many of the described experiments in fruit- biol. 68, 1185–1195.
ful discussions during the early stages of this work. We thank all Knoblich lab
Bowman, S.K., Rolland, V., Betschinger, J., Kinsey, K.A., Emery, G., and Kno-
members for support and discussions; Joshua A. Bagley, Christopher Esk,
blich, J.A. (2008). The tumor suppressors Brat and Numb regulate transit-
and Chong Li for comments on the manuscript; Peter Duchek, Joseph Gokce-
amplifying neuroblast lineages in Drosophila. Dev. Cell 14, 535–546.
zade, Elke Kleiner, the IMP/IMBA Biooptics Facility, the Next Generation
Sequencing Unit, and the Electron Microscopy Unit of the Vienna Biocenter Brand, M., Jarman, A.P., Jan, L.Y., and Jan, Y.N. (1993). asense is a Drosophila
Core Facilities (VBCF) for assistance; and the Harvard TRiP collection, the neural precursor gene and is capable of initiating sense organ formation.
Bloomington Drosophila Stock Center, and the Vienna Drosophila Resource Development 119, 1–17.
Center (VDRC) from the Vienna Biocenter Core Facilities (VBCF) for reagents. Brisson, L., Banski, P., Sboarina, M., Dethier, C., Danhier, P., Fontenille, M.-J.,
We thank the BIMSB Genomics Core Facility, especially Bora Uyar and Vedran Van Hée, V.F., Vazeille, T., Tardy, M., Falces, J., et al. (2016). Lactate Dehydro-
Franke, for supplying the PiGx pipeline. We also thank Dr. Yi Yang (East China genase B Controls Lysosome Activity and Autophagy in Cancer. Cancer Cell
University of Science and Technology) for providing the SoNar construct. Work 30, 418–431.

Figure 7. NAD+ Regeneration Is the Key Output of OxPhos during brat Tumorigenesis
(A) RFP-specific tumor burden analysis of flies bearing control tumors, ATP synthase-deficient tumors, and ATP synthase-deficient tumors rescued by over-
expression of Ndi1 bratRNAi.
(B) Head tumor burden quantification from (A). Each dot corresponds to one head.
(C) WB lobe imaging of RFPnuclear, Miranda, and PH3 in control, ATPsynd-deficient, and Ndi1-rescued ATPsynd-deficient tumors.
(D) Schematic of the experiments in (A)–(C) and their outcomes.
(E) Proposed mechanism of tumor cell immortalization during bratRNAi tumorigenesis. Scale bars, 50 mm. See also Figure S7.

Cell 182, 1490–1507, September 17, 2020 1505


ll
Article
Buck, M.D., O’Sullivan, D., Klein Geltink, R.I., Curtis, J.D., Chang, C.-H., Sanin, Franco, J., Balaji, U., Freinkman, E., Witkiewicz, A.K., and Knudsen, E.S.
D.E., Qiu, J., Kretz, O., Braas, D., van der Windt, G.J.W., et al. (2016). Mito- (2016). Metabolic Reprogramming of Pancreatic Cancer Mediated by CDK4/
chondrial Dynamics Controls T Cell Fate through Metabolic Programming. 6 Inhibition Elicits Unique Vulnerabilities. Cell Rep. 14, 979–990.
Cell 166, 63–76. Hall, C.N., Klein-Flügge, M.C., Howarth, C., and Attwell, D. (2012). Oxidative
Buescher, M., Yeo, S.L., Udolph, G., Zavortink, M., Yang, X., Tear, G., and phosphorylation, not glycolysis, powers presynaptic and postsynaptic mech-
Chia, W. (1998). Binary sibling neuronal cell fate decisions in the Drosophila anisms underlying brain information processing. J. Neurosci. 32, 8940–8951.
embryonic central nervous system are nonstochastic and require inscute- Haq, R., Shoag, J., Andreu-Perez, P., Yokoyama, S., Edelman, H., Rowe, G.C.,
able-mediated asymmetry of ganglion mother cells. Genes Dev. 12, Frederick, D.T., Hurley, A.D., Nellore, A., Kung, A.L., et al. (2013). Oncogenic
1858–1870. BRAF regulates oxidative metabolism via PGC1a and MITF. Cancer Cell 23,
Burkhart, D.L., and Sage, J. (2008). Cellular mechanisms of tumour suppres- 302–315.
sion by the retinoblastoma gene. Nat. Rev. Cancer 8, 671–682. Ho, A., and Dowdy, S.F. (2002). Regulation of G(1) cell-cycle progression by
Butler, A., Hoffman, P., Smibert, P., Papalexi, E., and Satija, R. (2018). Inte- oncogenes and tumor suppressor genes. Curr. Opin. Genet. Dev. 12, 47–52.
grating single-cell transcriptomic data across different conditions, technolo- Homem, C.C.F., and Knoblich, J.A. (2012). Drosophila neuroblasts: a model for
gies, and species. Nat. Biotechnol. 36, 411–420. stem cell biology. Development 139, 4297–4310.
Caussinus, E., and Gonzalez, C. (2005). Induction of tumor growth by altered Homem, C.C.F., Steinmann, V., Burkard, T.R., Jais, A., Esterbauer, H., and
stem-cell asymmetric division in Drosophila melanogaster. Nat. Genet. 37, Knoblich, J.A. (2014). Ecdysone and mediator change energy metabolism to
1125–1129. terminate proliferation in Drosophila neural stem cells. Cell 158, 874–888.
Courtney, K.D., Bezwada, D., Mashimo, T., Pichumani, K., Vemireddy, V., Hottiger, M.O. (2015). Nuclear ADP-Ribosylation and Its Role in Chromatin
Funk, A.M., Wimberly, J., McNeil, S.S., Kapur, P., Lotan, Y., et al. (2018). Plasticity, Cell Differentiation, and Epigenetics. Annu. Rev. Biochem. 84,
Isotope Tracing of Human Clear Cell Renal Cell Carcinomas Demonstrates 227–263.
Suppressed Glucose Oxidation In Vivo. Cell Metab. 28, 793–800.e2. Hui, S., Ghergurovich, J.M., Morscher, R.J., Jang, C., Teng, X., Lu, W., Es-
Cyran, S.A., Buchsbaum, A.M., Reddy, K.L., Lin, M.-C., Glossop, N.R.J., Har- parza, L.A., Reya, T., Le Zhan, Yanxiang Guo, J., et al. (2017). Glucose feeds
din, P.E., Young, M.W., Storti, R.V., and Blau, J. (2003). vrille, Pdp1, and the TCA cycle via circulating lactate. Nature 551, 115–118.
dClock form a second feedback loop in the Drosophila circadian clock. Cell Jang, S., and Javadov, S. (2018). Elucidating the contribution of ETC com-
112, 329–341. plexes I and II to the respirasome formation in cardiac mitochondria. Sci.
Rep. 8, 17732.
de la Cova, C., Senoo-Matsuda, N., Ziosi, M., Wu, D.C., Bellosta, P., Quinzii,
C.M., and Johnston, L.A. (2014). Supercompetitor status of Drosophila Myc Janiszewska, M., Suvà, M.L., Riggi, N., Houtkooper, R.H., Auwerx, J.,
cells requires p53 as a fitness sensor to reprogram metabolism and promote Clément-Schatlo, V., Radovanovic, I., Rheinbay, E., Provero, P., and Stamen-
viability. Cell Metab. 19, 470–483. kovic, I. (2012). Imp2 controls oxidative phosphorylation and is crucial for pre-
serving glioblastoma cancer stem cells. Genes Dev. 26, 1926–1944.
DeBerardinis, R.J., and Chandel, N.S. (2016). Fundamentals of cancer meta-
bolism. Sci. Adv. 2, e1600200. Jarman, A.P., Brand, M., Jan, L.Y., and Jan, Y.N. (1993). The regulation and
function of the helix-loop-helix gene, asense, in Drosophila neural precursors.
DeBerardinis, R.J., Mancuso, A., Daikhin, E., Nissim, I., Yudkoff, M., Wehrli, S.,
Development 119, 19–29.
and Thompson, C.B. (2007). Beyond aerobic glycolysis: transformed cells can
engage in glutamine metabolism that exceeds the requirement for protein and Jiang, Y., and Reichert, H. (2014). Drosophila neural stem cells in brain devel-
nucleotide synthesis. Proc. Natl. Acad. Sci. USA 104, 19345–19350. opment and tumor formation. J. Neurogenet. 28, 181–189.
Karcavich, R., and Doe, C.Q. (2005). Drosophila neuroblast 7-3 cell lineage: a
Duronio, R.J., O’Farrell, P.H., Xie, J.E., Brook, A., and Dyson, N. (1995). The
model system for studying programmed cell death, Notch/Numb signaling,
transcription factor E2F is required for S phase during Drosophila embryogen-
and sequential specification of ganglion mother cell identity. J. Comp. Neurol.
esis. Genes Dev. 9, 1445–1455.
481, 240–251.
Eichenlaub, T., Villadsen, R., Freitas, F.C.P., Andrejeva, D., Aldana, B.I.,
Kelsom, C., and Lu, W. (2012). Uncovering the link between malfunctions in
Nguyen, H.T., Petersen, O.W., Gorodkin, J., Herranz, H., and Cohen, S.M.
Drosophila neuroblast asymmetric cell division and tumorigenesis. Cell Biosci.
(2018). Warburg Effect Metabolism Drives Neoplasia in a Drosophila Genetic
2, 38.
Model of Epithelial Cancer. Curr. Biol. 28, 3220–3228.e6.
Knoblich, J.A. (2010). Asymmetric cell division: recent developments and their
Eroglu, E., Burkard, T.R., Jiang, Y., Saini, N., Homem, C.C.F., Reichert, H., and
implications for tumour biology. Nat. Rev. Mol. Cell Biol. 11, 849–860.
Knoblich, J.A. (2014). SWI/SNF complex prevents lineage reversion and in-
duces temporal patterning in neural stem cells. Cell 156, 1259–1273. Kuntz, E.M., Baquero, P., Michie, A.M., Dunn, K., Tardito, S., Holyoake, T.L.,
Helgason, G.V., and Gottlieb, E. (2017). Targeting mitochondrial oxidative
Faubert, B., Li, K.Y., Cai, L., Hensley, C.T., Kim, J., Zacharias, L.G., Yang, C., phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem
Do, Q.N., Doucette, S., Burguete, D., et al. (2017). Lactate Metabolism in Hu- cells. Nat. Med. 23, 1234–1240.
man Lung Tumors. Cell 171, 358–371.e9.
Landskron, L., Steinmann, V., Bonnay, F., Burkard, T.R., Steinmann, J., Reich-
Feitelson, M.A., Arzumanyan, A., Kulathinal, R.J., Blain, S.W., Holcombe, R.F., ardt, I., Harzer, H., Laurenson, A.-S., Reichert, H., and Knoblich, J.A. (2018).
Mahajna, J., Marino, M., Martinez-Chantar, M.L., Nawroth, R., Sanchez-Gar- The asymmetrically segregating lncRNA cherub is required for transforming
cia, I., et al. (2015). Sustained proliferation in cancer: Mechanisms and novel stem cells into malignant cells. eLife 7, R106.
therapeutic targets. Semin. Cancer Biol. 35 (Suppl ), S25–S54.
Lawson, D.A., Kessenbrock, K., Davis, R.T., Pervolarakis, N., and Werb, Z.
Fernald, K., and Kurokawa, M. (2013). Evading apoptosis in cancer. Trends (2018). Tumour heterogeneity and metastasis at single-cell resolution. Nat.
Cell Biol. 23, 620–633. Cell Biol. 20, 1349–1360.
Flaveny, C.A., Griffett, K., El-Gendy, Bel.-D., Kazantzis, M., Sengupta, M., Lee, K.S., Wu, Z., Song, Y., Mitra, S.S., Feroze, A.H., Cheshier, S.H., and Lu, B.
Amelio, A.L., Chatterjee, A., Walker, J., Solt, L.A., Kamenecka, T.M., and Bur- (2013). Roles of PINK1, mTORC2, and mitochondria in preserving brain tumor-
ris, T.P. (2015). Broad Anti-tumor Activity of a Small Molecule that Selectively forming stem cells in a noncanonical Notch signaling pathway. Genes Dev. 27,
Targets the Warburg Effect and Lipogenesis. Cancer Cell 28, 42–56. 2642–2647.
Flores, A., Sandoval-Gonzalez, S., Takahashi, R., Krall, A., Sathe, L., Wei, L., Mashimo, T., Pichumani, K., Vemireddy, V., Hatanpaa, K.J., Singh, D.K., Sira-
Radu, C., Joly, J.H., Graham, N.A., Christofk, H.R., and Lowry, W.E. (2019). sanagandla, S., Nannepaga, S., Piccirillo, S.G., Kovacs, Z., Foong, C., et al.
Increased lactate dehydrogenase activity is dispensable in squamous carci- (2014). Acetate is a bioenergetic substrate for human glioblastoma and brain
noma cells of origin. Nat. Commun. 10, 91. metastases. Cell 159, 1603–1614.

1506 Cell 182, 1490–1507, September 17, 2020


ll
Article
McGuire, S.E., Le, P.T., Osborn, A.J., Matsumoto, K., and Davis, R.L. (2003). Tumanov, S., Bulusu, V., Gottlieb, E., and Kamphorst, J.J. (2016). A rapid
Spatiotemporal rescue of memory dysfunction in Drosophila. Science 302, method for quantifying free and bound acetate based on alkylation and GC-
1765–1768. MS analysis. Cancer Metab. 4, 17.
Mishra, P., and Chan, D.C. (2016). Metabolic regulation of mitochondrial dy- Ugrankar, R., Theodoropoulos, P., Akdemir, F., Henne, W.M., and Graff, J.M.
namics. J. Cell Biol. 212, 379–387. (2018). Circulating glucose levels inversely correlate with Drosophila larval
Nakajima, E.C., and Van Houten, B. (2013). Metabolic symbiosis in cancer: re- feeding through insulin signaling and SLC5A11. Commun. Biol. 1, 110–115.
focusing the Warburg lens. Mol. Carcinog. 52, 329–337. Ulvklo, C., MacDonald, R., Bivik, C., Baumgardt, M., Karlsson, D., and Thor, S.
Neumüller, R.A., Richter, C., Fischer, A., Novatchkova, M., Neumüller, K.G., (2012). Control of neuronal cell fate and number by integration of distinct
and Knoblich, J.A. (2011). Genome-wide analysis of self-renewal in Drosophila daughter cell proliferation modes with temporal progression. Development
neural stem cells by transgenic RNAi. Cell Stem Cell 8, 580–593. 139, 678–689.
Otsuki, L., and Brand, A.H. (2018). Cell cycle heterogeneity directs the timing of van den Ameele, J., and Brand, A.H. (2019). Neural stem cell temporal
neural stem cell activation from quiescence. Science 360, 99–102. patterning and brain tumour growth rely on oxidative phosphorylation. eLife
Pastò, A., Bellio, C., Pilotto, G., Ciminale, V., Silic-Benussi, M., Guzzo, G., Ra- 8, 859.
sola, A., Frasson, C., Nardo, G., Zulato, E., et al. (2014). Cancer stem cells from Vazquez, F., Lim, J.-H., Chim, H., Bhalla, K., Girnun, G., Pierce, K., Clish, C.B.,
epithelial ovarian cancer patients privilege oxidative phosphorylation, and Granter, S.R., Widlund, H.R., Spiegelman, B.M., and Puigserver, P. (2013).
resist glucose deprivation. Oncotarget 5, 4305–4319. PGC1a expression defines a subset of human melanoma tumors with
Patra, K.C., Wang, Q., Bhaskar, P.T., Miller, L., Wang, Z., Wheaton, W., Chan- increased mitochondrial capacity and resistance to oxidative stress. Cancer
del, N., Laakso, M., Muller, W.J., Allen, E.L., et al. (2013). Hexokinase 2 is Cell 23, 287–301.
required for tumor initiation and maintenance and its systemic deletion is ther- Volkenhoff, A., Weiler, A., Letzel, M., Stehling, M., Klämbt, C., and Schirmeier,
apeutic in mouse models of cancer. Cancer Cell 24, 213–228. S. (2015). Glial Glycolysis Is Essential for Neuronal Survival in Drosophila. Cell
Peiris-Pagès, M., Bonuccelli, G., Sotgia, F., and Lisanti, M.P. (2018). Mito- Metab. 22, 437–447.
chondrial fission as a driver of stemness in tumor cells: mDIVI1 inhibits mito- Wang, C.-W., Purkayastha, A., Jones, K.T., Thaker, S.K., and Banerjee, U.
chondrial function, cell migration and cancer stem cell (CSC) signalling. Onco- (2016). In vivo genetic dissection of tumor growth and the Warburg effect. eLife
target 9, 13254–13275. 5, 11.
Piyankarage, S.C., Augustin, H., Grosjean, Y., Featherstone, D.E., and Shippy, Warburg, O. (1956). On respiratory impairment in cancer cells. Science 124,
S.A. (2008). Hemolymph amino acid analysis of individual Drosophila larvae. 269–270.
Anal. Chem. 80, 1201–1207.
Warburg, O., Wind, F., and Negelein, E. (1927). THE METABOLISM OF TU-
Rao, S., Mondragón, L., Pranjic, B., Hanada, T., Stoll, G., Köcher, T., Zhang, P., MORS IN THE BODY. J. Gen. Physiol. 8, 519–530.
Jais, A., Lercher, A., Bergthaler, A., et al. (2019). AIF-regulated oxidative phos-
Westermann, B. (2010). Mitochondrial fusion and fission in cell life and death.
phorylation supports lung cancer development. Cell Res. 29, 579–591.
Nat. Rev. Mol. Cell Biol. 11, 872–884.
Reichardt, I., Bonnay, F., Steinmann, V., Loedige, I., Burkard, T.R., Meister, G.,
Wurmus, R., Uyar, B., Osberg, B., Franke, V., Gosdschan, A., Wreczycka, K.,
and Knoblich, J.A. (2018). The tumor suppressor Brat controls neuronal stem
Ronen, J., and Akalin, A. (2018). PiGx: reproducible genomics analysis pipe-
cell lineages by inhibiting Deadpan and Zelda. EMBO Rep. 19, 102–117.
lines with GNU Guix. Gigascience 7, 1226.
Rossignol, R., Gilkerson, R., Aggeler, R., Yamagata, K., Remington, S.J., and
Capaldi, R.A. (2004). Energy substrate modulates mitochondrial structure and Yamano, K., and Youle, R.J. (2011). Coupling mitochondrial and cell division.
oxidative capacity in cancer cells. Cancer Res. 64, 985–993. Nat. Cell Biol. 13, 1026–1027.

Sanz, A., Soikkeli, M., Portero-Otı́n, M., Wilson, A., Kemppainen, E., McIlroy, Yao, C.-H., Wang, R., Wang, Y., Kung, C.-P., Weber, J.D., and Patti, G.J.
G., Ellilä, S., Kemppainen, K.K., Tuomela, T., Lakanmaa, M., et al. (2010). (2019). Mitochondrial fusion supports increased oxidative phosphorylation
Expression of the yeast NADH dehydrogenase Ndi1 in Drosophila confers during cell proliferation. eLife 8, 279.
increased lifespan independently of dietary restriction. Proc. Natl. Acad. Sci. Ye, X.-Q., Li, Q., Wang, G.-H., Sun, F.-F., Huang, G.-J., Bian, X.-W., Yu, S.-C.,
USA 107, 9105–9110. and Qian, G.-S. (2011). Mitochondrial and energy metabolism-related proper-
Senft, D., and Ronai, Z.A. (2016). Regulators of mitochondrial dynamics in can- ties as novel indicators of lung cancer stem cells. Int. J. Cancer 129, 820–831.
cer. Curr. Opin. Cell Biol. 39, 43–52. Zacharioudaki, E., Magadi, S.S., and Delidakis, C. (2012). bHLH-O proteins are
Shay, J.W., and Wright, W.E. (2011). Role of telomeres and telomerase in can- crucial for Drosophila neuroblast self-renewal and mediate Notch-induced
cer. Semin. Cancer Biol. 21, 349–353. overproliferation. Development 139, 1258–1269.
Sullivan, L.B., Gui, D.Y., Hosios, A.M., Bush, L.N., Freinkman, E., and Vander Zhao, Y., Wang, A., Zou, Y., Su, N., Loscalzo, J., and Yang, Y. (2016). In vivo
Heiden, M.G. (2015). Supporting Aspartate Biosynthesis Is an Essential Func- monitoring of cellular energy metabolism using SoNar, a highly responsive
tion of Respiration in Proliferating Cells. Cell 162, 552–563. sensor for NAD(+)/NADH redox state. Nat. Protoc. 11, 1345–1359.
Tatapudy, S., Aloisio, F., Barber, D., and Nystul, T. (2017). Cell fate decisions: Zhu, S., Lin, S., Kao, C.-F., Awasaki, T., Chiang, A.-S., and Lee, T. (2006). Gra-
emerging roles for metabolic signals and cell morphology. EMBO Rep. 18, dients of the Drosophila Chinmo BTB-zinc finger protein govern neuronal tem-
2105–2118. poral identity. Cell 127, 409–422.
Trapnell, C., Cacchiarelli, D., Grimsby, J., Pokharel, P., Li, S., Morse, M., Len- Zhu, S., Barshow, S., Wildonger, J., Jan, L.Y., and Jan, Y.-N. (2011). Ets tran-
non, N.J., Livak, K.J., Mikkelsen, T.S., and Rinn, J.L. (2014). The dynamics and scription factor Pointed promotes the generation of intermediate neural pro-
regulators of cell fate decisions are revealed by pseudotemporal ordering of genitors in Drosophila larval brains. Proc. Natl. Acad. Sci. USA 108,
single cells. Nat. Biotechnol. 32, 381–386. 20615–20620.

Cell 182, 1490–1507, September 17, 2020 1507


Cell Metabolism

Resource

Single-Cell RNA Sequencing Maps Endothelial


Metabolic Plasticity in Pathological Angiogenesis
Katerina Rohlenova,1,19 Jermaine Goveia,1,19 Melissa Garcı́a-Caballero,1,19 Abhishek Subramanian,1,19
Joanna Kalucka,1,16 Lucas Treps,1 Kim D. Falkenberg,1 Laura P.M.H. de Rooij,1 Yingfeng Zheng,2 Lin Lin,3,4 Liliana Sokol,1
Laure-Anne Teuwen,1,5,6 Vincent Geldhof,1 Federico Taverna,1 Andreas Pircher,1,14 Lena-Christin Conradi,1,15
Shawez Khan,1 Steve Stegen,7 Dena Panovska,8 Frederik De Smet,8 Frank J.T. Staal,9 Rene J. Mclaughlin,9

(Author list continued on next page)

1Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU

Leuven, Leuven 3000, Belgium


2State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, China
3Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark
4Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, Qingdao 266555, China
5Translational Cancer Research Unit, GZA Hospitals Sint-Augustinus, Antwerp 2610, Belgium
6Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
7Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging, KU Leuven, Leuven 3000,

Belgium
8Laboratory for Precision Cancer Medicine, Translational Cell & Tissue Research, Department of Imaging & Pathology, KU Leuven, Leuven

3000, Belgium
9Department of Immunology and Blood Transfusion, Leiden University Medical Center, Leiden 2300 RC, the Netherlands
10OXURION NV, Leuven 3001, Belgium
11BGI-Shenzhen, Shenzhen 518083, China

(Affiliations continued on next page)

SUMMARY cycle progression and in quiescence. Hypothesizing


that conserved genes are important, we used an inte-
Endothelial cell (EC) metabolism is an emerging grated analysis, based on congruent transcriptome
target for anti-angiogenic therapy in tumor angiogen- analysis, CEC-tailored genome-scale metabolic
esis and choroidal neovascularization (CNV), but little modeling, and gene expression meta-analysis in
is known about individual EC metabolic transcrip- cross-species datasets, followed by in vitro and
tomes. By single-cell RNA sequencing 28,337 murine in vivo validation, to identify SQLE and ALDH18A1
choroidal ECs (CECs) and sprouting CNV-ECs, we as previously unknown metabolic angiogenic targets.
constructed a taxonomy to characterize their hetero-
geneity. Comparison with murine lung tumor ECs
(TECs) revealed congruent marker gene expression
INTRODUCTION
by distinct EC phenotypes across tissues and dis-
eases, suggesting similar angiogenic mechanisms. Endothelial cell (EC) metabolism regulates angiogenesis, and is
Trajectory inference predicted that differentiation of an emerging target for anti-angiogenic therapy (AAT) in cancer
venous to angiogenic ECs was accompanied by and wet age-related macular degeneration (AMD) (Eelen et al.,
metabolic transcriptome plasticity. ECs displayed 2018). The design of new AATs by targeting EC metabolism
metabolic transcriptome heterogeneity during cell- would benefit from a better understanding of individual EC

Context and Significance

Targeting the metabolism of endothelial cells (ECs) is a promising strategy to block pathological blood vessel growth, or
angiogenesis, for the treatment of diseases like cancer. Understanding the landscape of metabolic gene expression at
the single-cell level will aid in identifying novel angiogenic targets. Here, researchers in Belgium and their colleagues sur-
veyed thousands of ECs in pre-clinical models of age-related macular degeneration and lung cancer. Their comprehensive
investigation identified genes and metabolic pathways that are congruently upregulated across diseases and tissues during
angiogenesis. Using an integrated analysis, the researchers generated a list of prioritized metabolic candidates and vali-
dated the importance of two candidates, SQLE and ALDH18A1, in pathological angiogenesis, supporting their potential
as therapeutic targets.

862 Cell Metabolism 31, 862–877, April 7, 2020 ª 2020 Elsevier Inc.
Stefan Vinckier,1 Tine Van Bergen,10 Nadine Ectors,8 Patrik De Haes,10 Jian Wang,11,12 Lars Bolund,3,4 Luc Schoonjans,1,2
Tobias K. Karakach,1,17,18 Huanming Yang,11,12 Geert Carmeliet,7 Yizhi Liu,2 Bernard Thienpont,13 Mieke Dewerchin,1
Guy Eelen,1 Xuri Li,2,20,* Yonglun Luo,3,4,11,12,20,* and Peter Carmeliet1,2,20,21,*
12China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
13Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
14Present address: Department of Hematology and Oncology, Internal Medicine V, Medical University Innsbruck, Innsbruck, Austria
15Present address: Clinic of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
16Present address: Aarhus Institute of Advanced Studies (AIAS), Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
17Present address: Bioinformatics Core Laboratory, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
18Present address: Rady Faculty of Health Sciences, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB,

Canada
19These authors contributed equally
20Senior author
21Lead Contact

*Correspondence: lixr6@mail.sysu.edu.cn (X.L.), alun@biomed.au.dk (Y.L.), peter.carmeliet@kuleuven.vib.be (P.C.)


https://doi.org/10.1016/j.cmet.2020.03.009

metabolism, but it remains unknown if ECs express a heteroge- 6 mice and repeated this procedure 3 times, using choroids from
neous metabolic gene signature and how single ECs reprogram healthy mice as controls (6 mice per sample, in triplicate) (Figures
their metabolic transcriptome signature when forming new ves- 1A and 1B). For comparative analysis, we generated a pooled
sels in disease. However, metabolomics (measuring metabolite sample of two choroids from one healthy human donor (see
levels or metabolic fluxes) is insufficiently sensitive to determine below). Single-cell suspensions were MACS-enriched for
single EC metabolism. Since we documented that changes in CD45 /CD31+ ECs (Cantelmo et al., 2016) and subjected to
metabolic gene expression signatures at the bulk population scRNA-seq. After quality filtering (Table S1), batch correction,
level can be predictive of changes in metabolism in ECs (Bruning and in silico EC selection, graph-based clustering was per-
et al., 2018; Cantelmo et al., 2016; Kalucka et al., 2018; Vande- formed to group a total of 28,337 ECs according to their gene
keere et al., 2018), we analyzed the metabolic transcriptome of expression profile. Clusters were annotated based on marker
ECs at the single-cell level. genes (Tables S2 and S3) and results were visualized using
During vessel sprouting, a navigating tip EC leads the way, t-distributed stochastic neighbor embedding (t-SNE) (Figures
while proliferating stalk cells elongate the vessel sprout (Po- 1C, 1D, S1A, and S1B).
tente et al., 2011); once newly formed vessels become CECs from control mice were indistinguishable from healthy
perfused, ECs adopt a quiescent phalanx phenotype (Welti peripheral CECs from lasered mice and clustered together (Fig-
et al., 2013). ECs rely on metabolic reprogramming when ures 1C–1F). We detected a new separate population in lasered
switching from quiescence to vessel sprouting (Eelen et al., mice, not present in healthy CECs, representing CNV-ECs (Fig-
2018; Li et al., 2019; Sawada and Arany, 2017; Yu et al., ure 1D). Compared to healthy CECs, CNV-ECs expressed acti-
2018). In tumors, bulk metabolic gene expression profiling vation markers associated with response to injury such as Sparc
identified metabolic targets in tumor ECs (Cantelmo et al., (Bradshaw and Sage, 2001) and Col18a1, a source of the angio-
2016). AMD is a common blinding disease of elderly people, static endostatin, previously used for CNV treatment (Marneros
characterized by ocular neovascularization. Laser-induced et al., 2007), a finding confirmed at the protein level by quantita-
choroid neovascularization (CNV) is a preclinical model of tive mass cytometry (CyTOF) (Figures 1G, S1C, and S1D;
AMD (Ambati and Fowler, 2012). Since angiogenic ECs in Table S2).
AMD/CNV have not been studied at the single-cell level, we
used single-cell RNA sequencing (scRNA-seq) to profile their Taxonomy of CECs and CNV-ECs
(metabolic) transcriptome heterogeneity. In CECs, we identified previously unknown sublineages of the
Anti-VEGF drugs are used for the treatment of cancer and classical arterial, capillary, and venous EC phenotypes (Figures
AMD, but resistance limits their efficacy (Jain, 2014; Yang 1E–1G; for more complete description of marker genes and puta-
et al., 2016). Hence, there is an unmet clinical need to identify tive inferred biological activity, see Table S3). For instance, we
novel angiogenic targets. scRNA-seq is a powerful technology identified large supply arteries (P1 on Figure 1D), smaller ramifying
to identify such candidates, but an outstanding challenge is to arterioles (P2) and arterial ECs expressing the shear stress marker
prioritize targets for further clinical translation. Here, we present (Pi16) (tentatively coined shear-stress induced arterial ECs [P3]),
a strategy, starting from scRNA-seq and complemented with and a laser-induced arterial subpopulation (activated arterial
orthogonal techniques, to prioritize metabolic targets that con- CEC) that upregulated activation markers and matricellular pro-
trol angiogenesis. teins (P4) (Figure 1H). Activated arterial CECs clustered together
with other arterial phenotypes (Figure 1D), suggesting a relatively
RESULTS normal transcriptome. Capillary CEC phenotypes expressed sig-
natures of the outer (P5) and inner choriocapillaries (P6), charac-
Identification and Characterization of CNV-ECs by terized by the differential expression of genes involved in fenestra-
scRNA-Seq tion and VEGF signaling (Blaauwgeers et al., 1999; McLeod et al.,
To model CNV in mice, we laser-induced 10 lesions per eye and 1995) (Table S2). Venous subclusters included cells expressing
microdissected choroids 7 days later. We pooled choroids from markers of large caliber vessels (P7), venules (P8), shear stress

Cell Metabolism 31, 862–877, April 7, 2020 863


A C D

G H I J

Figure 1. Heterogeneity of Choroid NEC and CNV-EC Phenotypes


(A) Schematic overview of the choroidal vasculature.
(B) Schematic overview of the study design.
(C) t-SNE plot, color-coded for the sample type. ECs isolated from healthy choroid are depicted in gray (top); ECs isolated from laser-injured choroids in red
(bottom). Red arrowhead and dotted circles indicate an emerging population of laser-injured specific CNV-EC phenotypes.
(D) t-SNE visualization of EC subpopulations in healthy and laser-injured choroids. CNV-EC phenotypes (red arrowhead in C) are boxed in the global t-SNE plot
and shown in a separate t-SNE plot on the lower left. Subclusters of peripheral (p) and intralesional CNV-ECs (c) are numbered.
(legend continued on next page)

864 Cell Metabolism 31, 862–877, April 7, 2020


(P9), post-capillary venules (pcvs) (P10) that upregulated a previ- from differentiation of activated pcv CECs to transitioning CNV-
ously identified CEC signature of resident endothelial stem cells ECs, then to immature CNV-ECs, which thereafter differenti-
(ESCs) (Naito et al., 2012; Wakabayashi et al., 2013), and an acti- ated to tip cells and finally to more mature neophalanx CNV-
vated pcv CEC phenotype (p11) (Figures 1I and S1E). We ECs (Figure 2B). This prediction extends previous morpholog-
observed two putative lymphatic EC phenotypes (LEC [P12] and ical evidence that neovessels may originate from pcvs (Folk-
LEC-like [P13]) that differentially expressed Lyve-1 (Figure 1D; Ta- man, 1982). Since pcv CECs expressed a previously validated
ble S2). The existence of lymphatics in the choroid remains signature of resident ESCs, our analysis provides further sug-
debated (Heindl et al., 2015; Koina et al., 2015). gestion that ESCs might contribute to new vessel sprouting,
Angiogenic CNV-ECs were distinct from normal CECs and as previously established by lineage tracing (Corey et al.,
included proliferating ECs (C1 in Figure 1D) and tip ECs (C2), 2016; Manavski et al., 2018; McDonald et al., 2018; Mondor
but also 3 previously unknown phenotypes that expressed sig- et al., 2016; Red-Horse et al., 2010; Wakabayashi et al.,
natures associated with transitioning from pcv to angiogenic 2013, 2018).
EC phenotypes (transitioning CNV-ECs [C3]), and immature Interestingly, when focusing on metabolic genes and path-
(immature [C4]) and maturing (neophalanx [C5]) neovasculature ways, we noted that membrane transport, ATP synthase, and
(Figure 1J). Tip cells upregulated transcripts of the disease- glycolysis gene signatures were dynamically regulated during
restricted angiogenic factor Pgf (encoding placental growth fac- differentiation from quiescent vein to angiogenic ECs (Figure 2B).
tor, Plgf) (Figures 1J and S1F). Immature ECs were characterized Maximal differences in metabolic gene expression of central car-
by the lack of specific marker gene expression, but expressed bon metabolism were observed in the most angiogenic EC phe-
activation markers and upregulated ribosomal gene expression notypes (immature and tip ECs), possibly suggesting that these
consistent with an activated intermediate phenotype. Neopha- ECs had higher metabolic demands to execute their biological
lanx ECs expressed markers of mature capillaries and arteries, functions (Figure 2B).
and were characterized by upregulation of a Notch signaling
gene signature (Figure S1G). Congruency Analysis of Metabolic Transcriptome
Interestingly, transcription factor activity analysis using single- Reprogramming
cell regulatory network inference and clustering (SCENIC) (Aibar We explored whether metabolic transcriptome reprogramming
et al., 2017) indicated differential transcription factor activity in was specific to CNV-ECs or a more general hallmark of the
EC subtypes (Figure 2A). Consistent with previous reports, Nr2f2 angiogenic switch in pathological angiogenesis (such as in tu-
expression was induced in activated pcvs and transitioning ECs mors), as this would address a fundamental question in vascular
(Jeong et al., 2017), while Sox17 expression was highest in arterial biology of whether vessels in different tissues and diseases form
ECs (Corada et al., 2013; You et al., 2005). SCENIC analysis of via similar or different mechanisms. We therefore explored to
CNV-ECs also identified transcription factors not previously impli- which extent CNV and tumors contained similar EC phenotypes,
cated in EC specification, such as in tip (Tgif1), immature (Smad1 and whether they expressed congruent genes.
and Sox4), and proliferating (Trp53) CNV-ECs (Figure 2A). Similarity and Congruency Analysis
We validated the taxonomy using orthogonal in situ localization We analyzed a publicly available, previously in-house gener-
techniques. Quantitative RNAscope to count transcript numbers, ated dataset of murine lung tumor ECs (TECs) (Goveia et al.,
combined with staining for the EC marker CD105, confirmed that 2020), which comprised largely similar EC phenotypes as
arterial (Gja4) and venous (Nr2f2) marker transcripts did not coloc- CNV-ECs. However, in addition, murine lung TECs contained
alize in the same CNV-ECs (Figure S1H). We confirmed by immu- breach and pre-breach ECs (that expressed both tip cell
nostaining of healthy choroids the expression of the following EC and podosome rosette markers, presumably involved in
markers: (1) artery ECs (ELN) and arteriole ECs (CXCL12) (Figures vessel sprouting initiation), and interferon ECs (displaying a
S2A–S2C), (2) capillary ECs (VEGFR2) (Figure S2D), and (3) transcriptome response to interferon, possibly involved in im-
venous ECs (VWF and SELP) (Figures S2E and S2F). Immuno- mune surveillance) (Goveia et al., 2020). Focusing on all de-
staining of CNV lesions confirmed the expression of a marker of tected genes, we explored whether similar EC phenotypes
immature ECs (APLNR), tip ECs (PlGF, LXN, and CXCR4), and could be detected in these diseases, and whether they ex-
pcv ECs (SPARCL1) (Figures S2G–S2J). pressed congruent genes.
We performed differential gene expression and gene set
Metabolic Transcriptome Reprogramming during enrichment analysis to determine which processes were upregu-
Pathological Vessel Sprouting lated in CNV-ECs and TECs versus CECs and NECs, respec-
We explored if ECs underwent a differentiation trajectory during tively (Figures 2C and 2D). Gene sets associated with prolifera-
vessel sprouting and if EC differentiation was associated with tion, hypoxia signaling, and extracellular matrix formation were
metabolic transcriptome changes. Trajectory inference analysis commonly upregulated (Figure 2C). Consistently, many of the
predicted that the hierarchy of angiogenic phenotypes resulted 175 commonly upregulated genes were involved in extracellular

(E and F) t-SNE plots, color-coded for the expression of the indicated marker genes (E) or gene sets (F). Red arrowheads indicate cells with high expression of the
indicated marker gene. Scale: white/gray is low expression; black (gene) or red (gene sets) is high gene expression.
(G–J) Heatmap of gene expression levels of the top 50 marker genes for broad categories of EC phenotypes (G), artery subclusters (H), vein subclusters (I), and
CNV-EC subclusters (J). In this and all further heatmaps depicting marker genes, colors represent row-wise scaled gene expression with a mean of 0 and SD of 1
(Z scores).
c.c., choriocapillaris; r.p.e., retinal pigment epithelium. See also Figures S1 and S2 and Tables S1, S2, and S3.

Cell Metabolism 31, 862–877, April 7, 2020 865


A B

E F

Figure 2. Gene Expression Signatures in CNV and TEC Subtypes


(A) Dot plot heatmap of the inferred activity of the indicated transcription factors. Scale: white/gray is low expression; black is high gene expression; dot size
corresponds to the fraction of cells in each cluster that have higher than average activity of the indicated transcription factor.
(B) Pseudotime trajectory of the indicated CNV-EC phenotypes (left) and Loess regression-smoothened gene expression of the indicated genes and metabolic
gene sets in pseudotime (right).
(C) Vascular gene sets upregulated in TECs (versus lung NECs) and in CNV-ECs (versus choroid CECs). Gene sets congruently upregulated in TECs and CNV-ECs
are summarized on the right.
(D) Genes upregulated in TECs versus NECs and choroid CECs versus CNV-ECs. A selection of genes upregulated in lung TECs versus NECs (left) and in CNV-
ECs versus choroid CECs (right) are listed. Genes congruently upregulated in TECs and CNV-ECs are listed on the right. Genes encoding ribosomal proteins, ATP
synthase subunits, and elongation factors are not listed individually; instead, their total number is displayed.

(legend continued on next page)

866 Cell Metabolism 31, 862–877, April 7, 2020


matrix remodeling, cytoskeleton, glycolysis, EC activation, and their presumed role in vessel sprouting initiation (Goveia et al.,
others. Interestingly, Aplnr, an angiogenic and vasculoprotective 2020) (Figures 3C and S3C).
gene that regulates EC metabolism (Apostolidis et al., 2018; Correlation with Transcription Factor Expression
Hwangbo et al., 2017), was identified as a congruent marker of Consistent with literature reports (Kanda et al., 2009), correlating
CNV-ECs and TECs (Figure 2D). the gene signatures with inferred transcription factor activity
Since differential analysis of pooled populations may not scores revealed that Pparg correlated with the induction of a tri-
adequately discover genes with restricted expression in small EC glyceride catabolism signature. Interestingly, hypoxia, as indicated
subpopulations such as tip and proliferating cells, we determined by the inferred activity of HIF-1a, correlated with glycolysis and OX-
whether the same EC subpopulations were present in CNV-ECs PHOS gene expression in TECs (Figure S3D). While expected for
and TECs. We used the Jaccard similarity index to score the simi- glycolysis (given that hypoxia upregulates glycolysis; Eales et al.,
larity of marker gene sets of all EC subpopulations, and observed 2016), the upregulation of OXPHOS genes in hypoxic conditions
that marker gene sets across CNV-ECs and TECs were relatively was surprising (given that hypoxia suppresses OXPHOS; Eales
similar for several EC subpopulations (Figures 2E and S3A). et al., 2016; Semenza, 2011), but in line with findings that HIF-1a
Further, TECs and CNV-ECs of the same phenotype expressed activation can occur at oxygen levels that are not sufficiently low
congruent marker genes (Figure 2F). Similar to CNV-ECs, trajectory to suppress mitochondrial respiration (Eales et al., 2016). Thus,
inference analysis predicted that the hierarchy of TEC phenotypes while proliferating CNV-ECs and TECs upregulate OXPHOS and
originated in veins that expressed resident ESC markers (Goveia glycolysis gene signatures, the metabolic transcriptome profile of
et al., 2020; Wakabayashi et al., 2018) differentiating to postcapil- other angiogenic ECs is rather diverse across diseases.
lary veins and further to an immature TEC phenotype, tip cells, neo-
phalanx TECs, and activated arteries (Figure S3B). Analysis of Metabolic Transcriptome Reprogramming
Metabolic Transcriptome Signatures during the Cell Cycle
Focusing on metabolic genes, we observed that proliferating Since our congruency analysis showed that proliferating CNV-ECs
ECs in both disease models upregulated the expression of meta- and TECs have similar gene expression profiles, we analyzed their
bolic genes involved in one-carbon metabolism, nucleotide syn- (metabolic) signature and transcription factor regulation in more
thesis, tricarboxylic acid (TCA) cycle, and oxidative phosphoryla- detail. We therefore developed a prediction model to reconstruct
tion (OXPHOS) (Figure 3A). In contrast, glycolytic gene continuous cell-cycle pseudotime in proliferating murine CNV-
expression was upregulated in proliferating, tip, and immature ECs and TECs based on periodically expressed genes identified
ECs in tumors, and was elevated in CNV in proliferating ECs, in cultured TECs (Figures S3E–S3I; STAR Methods). Comparative
but less in tip and immature ECs (Figure 3B). These observations analysis of the top 1% most periodic genes in proliferating TECs
might suggest that the metabolic demands of proliferating ECs and CNV-ECs revealed 97 genes that were periodically expressed
are disease- or tissue-type independent, while metabolic adap- during the cell cycle in both models, 43 of which were not previ-
tations of other subtypes may be more plastic. ously described in reference databases (Santos et al., 2015). Inter-
The metabolic gene expression signatures between the estingly, in both models, several metabolic genes ranked in the top
different TEC phenotypes were more outspoken, possibly re- 1% most periodic genes (Rrm2, Tk1, Tyms, Dut, Rrm1, and
flecting the harsh nutrient-deprived micro-environment in tu- Dctpp1) (Figure 3D; Table S4). Further, gene set variation analysis
mors and the fact that TECs grow in an uncontrolled, non- revealed that various metabolic pathways involved in nucleotide
resolving manner. Indeed, heatmap analysis revealed that biosynthesis (e.g., one-carbon metabolism and pyrimidine nucle-
most TECs exhibited a different metabolic transcriptome signa- oside biosynthesis) ranked among the top 5% most periodically
ture (Figure S3C). Subsequent analysis at the gene level showed expressed gene sets (Figure 3E; Table S4). Overall, our scRNA-
that capillary TECs upregulated the expression of genes control- seq offers new possibilities to study metabolic gene expression ki-
ling lipid uptake (Figures 3C and S3C), raising the question of netics during the cell cycle.
whether they need lipids for internal use when switching to
quiescence (Kalucka et al., 2018) and/or for trans-EC transport Metabolic Angiogenic Target Identification
to cancer cells for energy production or lipogenesis (Santos Identification of Commonly Upregulated Metabolic Genes
and Schulze, 2012). Venous TECs upregulated transcripts of Since OXPHOS and glycolysis are validated metabolic angiogenic
genes involved in prostaglandin metabolism (Figures 3C and targets (Cantelmo et al., 2016; De Bock et al., 2013; Diebold et al.,
S3C), suggesting a role in vasoregulation, sprouting, or vascular 2019), we designed an integrated analysis to identify other previ-
inflammation (Félétou et al., 2011; Iñiguez et al., 2003). Interferon ously unrecognized angiogenic candidates regulating EC meta-
(IFN)-activated TECs upregulated genes involved in nucleotide bolism. We thus performed differential gene expression analysis
catabolism to salvage/lower nucleotide content (Figure 3C) (Bar- to determine which metabolic genes and gene sets were
ankiewicz et al., 1986). In turn, breach TECs upregulated genes commonly upregulated in TECs and CNV-ECs, versus normal
involved in extracellular matrix production the most, in line with ECs. Pathway mapping of gene transcripts involved in central

(E) Three-dimensional principal component analysis (PCA) on the pairwise Jaccard similarity coefficients of marker gene sets between subpopulations in TECs
and CNV-ECs. Squares denote CNV-EC phenotypes; circles denote TEC phenotypes. Note that equivalents of breach, pre-breach, and interferon TEC phe-
notypes were not present in CNV-ECs.
(F) Heatmap of expression levels of congruent genes in TEC and CNV-EC phenotypes (all genes analyzed). Note that the TEC and CNV-EC heatmaps show the
same set of congruent genes.
Comp, component; PC, principal component. See also Figure S3.

Cell Metabolism 31, 862–877, April 7, 2020 867


A serine, one-carbon B glycolysis C lipid metabolism
& nucleotide synthesis

TEC CNV-EC TEC CNV-EC


serine Slc2a1 Lpl
1C
Hk1 Lipe
cycle biosynthesis
TCA nucleotide

Pfkl Mgll
Pfkm Fabp4
Pfkp Fabp5
2
Aldoa Pparg 1
Tpi1 Cd36 0
Gapdh Gpihbp1 -1

Pgk1
Pgam1 prostaglandin metabolism
OXPHOS

Eno1 Ptgs1
2 2
2 1
Eno1b 1 Ptgs2 1
Pkm Ptgis
0 0 0 0
Ldha 0
-1 Slco2a1
-2 Slc16a3 -1
-2
tip cell
proliferating

transitioning
tip cell
proliferating

breach cell
pre-breach cell

vein

interferon
large vein

immature
immature

neophalanx
postcapillary

activated artery

artery
neophalanx

TEC capillary

tip cell
proliferating

transitioning
tip cell

breach cell

interferon
proliferating

pre-breach cell

vein
large vein

immature
immature

neophalanx
postcapillary

activated artery

artery
neophalanx

TEC capillary
nucleotide breakdown
Cmpk2
2
Upp1
1
Nt5c3 0
Pnp -1

ECM biosynthesis
Chst7
D expression in cell cycle E expression in cell cycle Mgat5
Stt3a
metabolic genes metabolic gene sets Ganab
Colgalt1 2

100 0 100 0 Plod1


0
Plod3
M M P4ha1
88 88 -2
87 87

tip cell
proliferating

breach cell

interferon
pre-breach cell

vein
large vein
immature
postcapillary

activated artery

artery
neophalanx

TEC capillary
G2M

G2M
G1

G1
TECs

TECs

75 75
74 74
G2

G2

64 64
63 S 63 S
G1S 42 G1S 42
43 Tk1 43 G1/S specific transcription
53 52 53 52
Rrm2 kinesins
Tyms one-carbon pool by folate
100 0 100 0
Dut deoxyribonucleotide biosynthesis
88
M Dctpp1 88
M pyrimidine nucleoside biosynthesis
87 87
G2M

G2M
G1

G1
CNV-ECs

CNV-ECs

75 75
74 74
G2

G2

64 64
63 S 63 S
G1S 42 G1S 42
43 43
53 52 53 52

Figure 3. Metabolic Heterogeneity in TECs and CNV-ECs


(A) Heatmap of the gene expression levels of the indicated metabolic pathways in TEC and CNV-EC subpopulations. Genes were grouped according to metabolic
pathways and ordered so that the most discriminative genes for proliferating ECs are depicted first.
(B) Heatmap of gene expression levels of the indicated glycolytic genes in TEC and CNV-EC subpopulations.
(C) Heatmap of gene expression levels of the indicated genes in TECs.
(D and E) Circos plot representation of Loess regression-smoothened gene expression of the indicated genes (D) and gene sets (E) during the cell cycle in TECs
(top) and CNV-ECs (bottom). Cell-cycle phase assignment, indicated as color-coded sectors, was based on the known periodicity of cyclins. Cell-cycle
pseudotime is represented as a percentage and indicated outside of the circos plot, starting (t = 0%) from cytokinesis (start of G1). The thickness of the line
corresponds to the expression levels.
1C, one-carbon. See also Figure S3 and Table S4.

carbon metabolism confirmed that CNV-ECs upregulated tran- also involved in other metabolic/biological activities, such as redox
scripts of metabolic pathways supporting biomass synthesis, and energy homeostasis (Eelen et al., 2018). Compared to NECs,
including glycolysis, nucleotide synthesis, TCA cycle, OXPHOS, TECs showed a qualitatively largely similar upregulation of
and others (Figures 4A and 4B). Several of these pathways are anabolic pathways (Figures 4A and S4A).

868 Cell Metabolism 31, 862–877, April 7, 2020


A B Figure 4. Global Metabolic Reprogramming
in Pathological Angiogenesis
(A) Dot plot heatmap of the indicated glycolytic
genes. The dot size corresponds to the fraction of
cells that have higher than average activity of the
indicated genes. Scale: white/gray is low expres-
sion; black is high gene expression.
(B) Map of upregulated central carbon metabolic
pathways in CNV-ECs versus peripheral CECs.
Blue color indicates downregulated expression,
red upregulated expression, and gray unchanged
expression.
(C and D) Metabolic gene set enrichment analysis
in NEC versus TECs (C) and CEC versus CNV-ECs
(D) (q < 0.25 for all gene sets). Numbers between
parentheses indicate alternative gene sets per-
taining to the same biological function or signaling
pathway.
(E) Metabolic gene sets, upregulated in TECs
(versus lung NECs) and in CNV-ECs (versus
choroid CECs). Congruent upregulated metabolic
pathways are listed underneath.
See also Figure S4.

biosynthesis, and TCA cycle, compared


to NECs and peripheral ECs (Figures 4C–
4E). Notably, transcripts of genes involved
in collagen synthesis were also highly upre-
gulated in angiogenic ECs in both diseases
(Figure 4E). Thus, based on transcriptome
C D E analysis, angiogenic ECs have at least
two prominent metabolic gene expression
signatures, i.e., that of biomass production
for proliferation and of collagen biosyn-
thesis for extracellular matrix remodeling.
Prediction of Candidates with
Functionally Relevant Role in
Metabolism by GEMs
Given that changes in transcript levels of
metabolic genes alone may not relate to
changes in metabolic fluxes, we used
genome-scale metabolic models (GEMs)
to in silico prioritize metabolic candidates.
The upregulation of OXPHOS and TCA cycle gene expression GEMs are mathematical representations of a network of active
signatures in proliferating ECs was noteworthy, when consid- metabolic enzymes (pathways) (Kim and Lun, 2014; Ryu et al.,
ering that ECs are glycolysis-addicted (Cantelmo et al., 2016; 2015) and represent computational tools to predict the impor-
De Bock et al., 2013), but is in line with reports showing the tance of metabolic reactions for biological responses (Pagliarini
importance of OXPHOS for EC proliferation (Diebold et al., et al., 2016; Thiele and Palsson, 2010). We constructed a CEC-
2019; Ying et al., 2017). The upregulation of a glycolysis tran- tailored GEM based on the generic human metabolic recon-
scriptome signature in TECs has been validated by functional ev- struction RECON1 and scRNA-seq data from 1,670 freshly iso-
idence that genetic deletion in ECs or pharmacological blockade lated human CECs, which recapitulated murine CEC pheno-
of the glycolytic activator PFKFB3 reduces TEC proliferation and types but with lower resolution (Figures 5A, S4B, and S4C),
induces tumor vessel normalization (Cantelmo et al., 2016), and and optimized it for biomass (as a proxy for proliferation) or
may also explain why treatment with a PFKFB3 blocker inhibited collagen production using two distinct EC-specific objective
CNV in the mouse (Schoors et al., 2014). functions.
An unbiased metabolic gene set enrichment analysis, combined Using the CEC-tailored GEM, we identified 288 essential
with a congruency analysis to identify commonly upregulated genes for biomass synthesis (Table S5), involved in glycolysis,
metabolic pathways, revealed that genes involved in pathways in TCA cycle, pentose phosphate pathway, OXPHOS, fatty acid
central carbon metabolism were among the most upregulated in oxidation, nucleotide synthesis and salvage, cholesterol biosyn-
TECs and CNV-ECs, including glycolysis, OXPHOS, nucleotide thesis, sphingolipid metabolism, and amino acid metabolism

Cell Metabolism 31, 862–877, April 7, 2020 869


A B

C D

Figure 5. Metabolic Target Prediction


(A) Schematic representation of GEM reconstruction.
(B) Venn diagrams indicating metabolic genes that encode rate-limiting enzymes and are predicted to be essential for biomass production (top) and collagen
biosynthesis (bottom) by four different methods (see STAR Methods for details).
(C) Upset plot visualization of the results of a differential gene set variation enrichment meta-analysis of nine bulk transcriptomics datasets, showing the number of
genes that were more highly expressed in TECs than NECs isolated from the indicated tumor type. The bar graph represents the number of gene sets detected in
the tumor type(s) indicated by the dot plot panel below. Five gene sets (displayed on the figure; involved in the displayed processes) were consistently higher
expressed in TECs than NECs (red bar and intersection). HCC, hepatocellular carcinoma; CRCLM, colorectal cancer liver metastasis; CRC, colorectal cancer;
medullo Wnt, Wnt-driven medulloblastoma; medullo Shh, sonic hedgehog driven medulloblastoma; RCC, renal cell carcinoma.
(D) Gene expression meta-analysis of the nine NEC versus TEC datasets shown in (C). The S-curve has 10,850 dots, representing genes that were detected in all
nine datasets. x axis, rank numbers from 1 to 10,850 (consistently overexpressed genes in TECs have a low rank number; consistently downregulated genes have
a high rank number); y axis, the scaled meta-analysis score (consistently overexpressed genes in TECs have a low meta-analysis score; consistently down-
regulated genes have a high meta-analysis score). SQLE and ALDH18A1 are shown as red dots and listed on the left.
See also Figure S5 and Table S5.

(Table S5). The roles of glycolysis (De Bock et al., 2013), Integrated Analysis to Prioritize Metabolic Genes for
OXPHOS (Diebold et al., 2019), fatty acid oxidation (Schoors Functional Validation
et al., 2015), serine metabolism (Vandekeere et al., 2018), and To identify functionally relevant metabolic angiogenic targets in
glutamine metabolism (Huang et al., 2017) in biomass synthesis CNV-ECs, we performed an integrated analysis (Figure S5A).
and EC proliferation have been established, thus validating the Specifically, we focused on the subset of rate-limiting single
predictive potential of the CEC-tailored GEM. However, a gene encoded metabolic enzymes, predicted by GEM to be
possible role of cholesterol synthesis in EC growth has only mini- essential for biomass production or collagen biosynthesis (Fig-
mally been studied, without conclusive results. Further, consis- ure 5B; Table S5). We selected candidates that were more highly
tent with previous reports (Phang, 2019), the CEC-tailored expressed in CNV-ECs than CECs, yielding 30 genes for
GEM predicted the essentiality of proline biosynthesis for biomass production and 4 for collagen synthesis (Table S5). Sec-
collagen production. In addition, glutamine and glutamate trans- ond, we reasoned that genes that were conserved across
porters, and enzymes involved in alanine, glycine, and serine models, diseases, and species represent robust targets. We
metabolism, were also predicted to be essential, reflecting that thus filtered for genes consistently upregulated also in tumor
the molecular composition of collagen consists of >70% of (hy- angiogenesis, resulting in a set of 17 candidate genes. Finally,
droxy)proline, glycine, glutamate, and alanine (Eastoe, 1955). unbiased meta-analysis across 9 different murine and human

870 Cell Metabolism 31, 862–877, April 7, 2020


A B C

E F G

Figure 6. Metabolic Target Validation


(A) 3H-thymidine incorporation in DNA assay upon SQLE (mean ± SEM, n = 16, *p < 0.05, unpaired two-tailed t test) or ALDH18A1 (mean ± SEM, n = 3, *p < 0.05,
unpaired two-tailed t test) silencing (KD denotes shRNA knockdown).
(B) Scratch wound migration assay with control and SQLE (mean ± SEM, n = 7, *p < 0.05, unpaired two-tailed t test) or ALDH18A1 (mean ± SEM, n = 3, *p < 0.05,
unpaired two-tailed t test) silenced ECs.
(legend continued on next page)
Cell Metabolism 31, 862–877, April 7, 2020 871
datasets revealed that Aldh18a1 and Sqle were among the (Goveia et al., 2020), which we now used to analyze metabolic
most consistently induced genes in TECs (Figures 5C and 5D; gene expression, enabled us to characterize metabolic gene
Table S5). Aldh18a1 encodes pyrroline-5-carboxylate-synthase expression heterogeneity in sprouting ECs across two tissues
(P5CS), the rate-controlling enzyme of proline and collagen and diseases.
biosynthesis, a pathway that was consistently upregulated in The metabolic transcriptome diversity was most outspoken for
angiogenic ECs in tumors and CNV (Figures S5B and S5C). TECs and, remarkably, TEC phenotypes expressed distinct
Sqle encodes squalene monooxygenase, a rate-limiting enzyme metabolic transcriptome signatures. Presumably, this complex
in cholesterol biosynthesis (Cerqueira et al., 2016). metabolic transcriptome heterogeneity is required for EC pheno-
Functional Validation of Selected Targets types to execute their specialized functions in different vascular
To functionally validate the role of ALDH18A1 and SQLE in vessel compartments. However, given that the micro-environment can
sprouting, we silenced these genes (Figures S5D–S5G), which influence cellular metabolism (Muir et al., 2018), part of the het-
impaired EC proliferation and migration, vessel sprouting (Fig- erogeneity can possibly also be attributed to different environ-
ures 6A–6D), and EC tip cell competitivity (Figures 6E and 6F). mental signals in distinct vascular compartments. This may
To explore global expression changes induced by SQLE and partly explain why tip and immature ECs had a stronger glyco-
ALDH18A1 silencing, we performed multi-omics analysis. Tran- lytic gene signature in tumors than in CNV, possibly evoked by
scriptomics analysis confirmed decreased expression of genes the harsher nutrient-deprived environment and larger abun-
involved in cell proliferation and DNA replication in silenced cells dance of metabolism-altering signals in the tumor as compared
(Figures S6A and S6B). These results were corroborated at the to the CNV lesions (Lyssiotis and Kimmelman, 2017).
protein level by proteomics analysis (Figure S6C). Targeted me- The EC taxonomies in the eye and lung offer integrated com-
tabolomics showed that SQLE and ALDH18A1 silencing did not parison of EC phenotypes across tissues and diseases. Even
affect the energy charge (Figure S6D), but confirmed that though tissue-type-specific EC phenotypes were recognized,
ALDH18A1 knockdown lowered the levels of proline and hy- angiogenic ECs in CNV and tumors seemed to form and maintain
droxyproline, a surrogate for collagen content (Stoilov et al., neovessels by developing similar EC phenotypes, including
2018) (Figures S6E and S6F). Finally, in vivo intraocular treatment proliferating, tip, immature, and neophalanx ECs. Across dis-
with siRNAs against Sqle and Aldh18a1 decreased the expres- eases and tissues, angiogenic ECs congruently upregulated
sion levels of these targets in CNV-ECs (Figure S6G) and in- the expression of non-metabolic marker genes, as well as meta-
hibited neovascularization of laser-induced choroid (Figure 6G). bolic genes involved in OXPHOS, glycolysis, TCA cycle, nucleo-
To explore potential therapeutic relevance, we used NB-598 to tide biosynthesis, and mRNA metabolism. Hence, blood vessels
pharmacologically inhibit SQLE in vivo and observed a reduction seem to sprout by inducing the differentiation of largely similar
in both corneal angiogenesis and CNV (Figure 6H). EC phenotypes across diseases and tissues. Tumors have
been named ‘‘non-healing wounds’’ and angiogenesis of tumors
DISCUSSION may thus build on similar principles as a CNV wound (Dvorak,
1986). Nonetheless, tissue- and disease-specific differences in
Objectives of this study were (1) to construct a taxonomy of metabolic transcriptome profiles were observed, likely in part
CECs and CNV-ECs, (2) to characterize the heterogeneity of due to differences in micro-environmental conditions.
metabolic gene expression signatures of different ECs at the sin- Trajectory inference analysis predicted that the hierarchy of
gle-cell level, (3) to explore if and how angiogenic ECs reprogram angiogenic phenotypes is derived from pcv ECs and that pheno-
their metabolic transcriptome signature during pathological typic differentiation is associated with plastic metabolic gene
angiogenesis, and (4) to design an integrated analytical expression reprogramming, suggesting that metabolic plasticity
approach to identify angiogenic targets regulating EC meta- supports phenotypic plasticity. Early morphological studies sug-
bolism, not previously known to co-determine pathological gested that vessel sprouting originates from this venous vascular
angiogenesis. bed (Folkman, 1982), but using in silico lineage tracing, we iden-
We constructed a CEC/CNV-EC taxonomy and identified 18 tified a differentiation trajectory of venous ECs over intermediate
choroidal EC phenotypes, of which 8 were previously not recog- transitioning immature ECs to tip ECs and later to more mature
nized, and characterized transcription factors, predicted to be neophalanx ECs.
involved in the differentiation of different choroidal ECs. The Another objective of this study was to explore whether meta-
availability of this choroidal CEC/CNV-EC taxonomy, as well as bolic signatures of individual ECs could be utilized for the discov-
a previously in-house constructed lung NEC/TEC taxonomy ery of metabolic genes, previously not recognized to fuel vessel

(C) Bright field photographs of control, ALDH18A1KD, and SQLEKD EC spheroids. Scale bar, 100 mm.
(D) Morphometric quantification of the number of sprouts, average, and cumulative sprout length for control, SQLEKD, and ALDH18A1KD spheroids with or without
MitoC treatment (mean ± SEM, n = 3 for all parameters; ns, p > 0.05; *p < 0.05, unpaired two-tailed t test).
(E and F) Mosaic EC spheroid competition of control (red), SQLEKD (E), or ALDH18A1KD (F) (green) ECs. Quantification of the fraction of tip cells with the indicated
genotype. Data are mean ± SEM, n = 3, *p < 0.05 by unpaired two-tailed t test.
(G) Quantification of CNV blood vessel area in mice treated with control siRNA (CTRL) or siRNA against murine Sqle or Aldh18a1. Representative images are
shown on the right. Scale bars, 75 mm. Data are mean ± SEM, n = 3 independent experiments each using six mice per group, *p < 0.05, unpaired two-tailed t test.
(H) Quantification of CNV (left) and corneal angiogenesis upon corneal cauterization-induced injury (right) in mice treated with vehicle (CTRL) or NB-598 (an SQLE
blocker). Representative micrographs are shown on the right. Scale bars, 75 mm (CNV) and 500 mm (cornea). Data are mean ± SEM, n = 3 independent ex-
periments each using six mice per group, *p < 0.05, unpaired two-tailed t test.
See also Figures S5 and S6.

872 Cell Metabolism 31, 862–877, April 7, 2020


sprouting. Hypothesizing that genes that were conservedly upre- B Mouse model of Lewis Lung Carcinoma
gulated across diseases and species represent robust candi- B Cell Lines and Primary Cell Culture
dates, we performed an integrated multi-layered approach d METHOD DETAILS
combining scRNA-seq of ECs from different tissues/diseases, B In Vitro Functional Assays
congruency transcriptome analysis, genome-scale metabolic B Knockdown Strategy
modeling, and cross-species meta-analysis to identify B Treatment with Inhibitor in the Murine Ocular Models
conserved angiogenic metabolic targets, i.e., Sqle for biomass B Murine Choroid Immunostaining
synthesis and Aldh18a1 for collagen synthesis. Functional vali- B RNA Isolation and Quantitative RT-PCR
dation revealed that silencing of these targets impaired vessel B Single-cell Droplet-based RNA Sequencing
sprouting in vitro and inhibited pathological ocular angiogenesis B Single-cell Transcriptomics Analysis
in vivo. While it was not the primary goal to develop new AAT B Quality Control and Data Normalization
strategies, but rather to provide proof of principle of the inte- B In Silico EC Selection
grated approach, the identified metabolic targets might none- B Batch Effect Correction
theless deserve further attention for AAT development, though B Feature Selection and Dimensionality Reduction
an EC-selective drug delivery approach would then be desirable. B EC Cluster Identification
B Marker Gene Analysis
Limitations of Study B Cluster Annotation
We acknowledge limitations of our study. First, the inferred biolog- B Gene Set Variation Analysis
ical role and topographical localization in the vasculature of each B Evaluation of Dissociation Artifacts
EC phenotype and the computational trajectory inference analysis B Heatmap Analysis
are putative/predicted and require additional marker gene identi- B Trajectory Inference
fication and functional validation to probe their biological roles/ B Jaccard Similarity Analysis
principles and anatomical topography. Second, the contribution B Gene Set Enrichment Analysis
of the resident ESCs to angiogenesis and the precise mechanisms B Metabolic Gene Expression Analysis and Pathway
of how ALDH18A1 and SQLE regulate EC migration/proliferation Mapping
and EC metabolism require further study. Third, transcript levels B Cell Cycle Pseudotime Analysis
and metabolic modeling by GEMs do not fully capture the B Development of a Multivariate Model to Predict Cell
complexity of metabolism (metabolic fluxes, enzyme activity, Cycle Pseudotime
and metabolite levels). However, gene signatures and GEM B Pathway and Transcription Factor Activity Analysis
modeling have been proven to be predictive of the metabolic B Bulk RNA-sequencing Analysis
flux changes in ECs (Bruning et al., 2018; Cantelmo et al., 2016; B Proteomics Sample Preparation
Kalucka et al., 2018; McGarrity et al., 2018; Patella et al., 2015; B Proteomics LC-MS/MS and Data Analysis
Vandekeere et al., 2018). Fourth, we expect that future exploration B Metabolomics and Data Analysis
of our dataset using newly developed pre-processing and analysis B Meta-analysis of Transcriptomics Data
algorithms might further fine-tune our analysis and interpretation. B RNAscope In Situ Hybridization and Quantification
Finally, the current data await future development of novel tech- B Cytometry by Time of Flight Mass Cytometry (CyTOF)
nology to quantify metabolism at the single-cell level in ECs, but B CyTOF Data Analysis
the potential of using an integrated analysis to overcome technical B Curation of a GEM Model
limitations in metabolic target prioritization is demonstrated by the B Development of an EC-tailored GEM
validation of the functional role of the selected candidates B Development of EC-Tailored Objective Functions
(ALDH18A1 and SQLE) in vessel sprouting in vivo. B Development of an Angiogenic CEC-GEM Model
To maximize resource value, we provide accompanying data B Prediction of Essential Genes in the Angiogenic
exploration web tools—available at https://www.vibcancer.be/ CEC Model
software-tools/Murine_ECTax and https://www.vibcancer.be/ d QUANTIFICATION AND STATISTICAL ANALYSIS
software-tools/scCycle, and via the publicly available added- d DATA AND CODE AVAILABILITY
value database EndoDB (Khan et al., 2019). B Data Resources
B Software

STAR+METHODS
SUPPLEMENTAL INFORMATION
Detailed methods are provided in the online version of this paper
Supplemental Information can be found online at https://doi.org/10.1016/j.
and include the following:
cmet.2020.03.009.
d KEY RESOURCES TABLE
d LEAD CONTACT AND MATERIALS AVAILABILITY ACKNOWLEDGMENTS
d EXPERIMENTAL MODEL AND SUBJECT DETAILS
We acknowledge A. Bouché, T. Van Brussel, R. Schepers, M. Pellemans, L.
B Patient Material and Choroid EC Isolation
Roussel, and I. Van Hove for technical assistance. We also gratefully acknowl-
B Mice edge the expert advice and help of J. Qian, B. Boeckx, and D. Lambrechts.
B Mouse Model of Choroidal Neovascularization Metabolomics analysis was done at the Metabolomics Expertise Center,
B Murine Choroid Endothelial Cell Isolation VIB, Leuven, Belgium. Proteomics analysis was done at the Proteomics

Cell Metabolism 31, 862–877, April 7, 2020 873


Expertise Center, VIB, Ghent, Belgium. Funding was provided by Fonds voor Apostolidis, S.A., Stifano, G., Tabib, T., Rice, L.M., Morse, C.M., Kahaleh, B.,
Wetenschappelijk Onderzoek (FWO; to K.R., J.G., J.K., and L.T.); a Marie Cu- and Lafyatis, R. (2018). Single cell RNA sequencing identifies HSPG2 and
rie-IEF Fellowship (to K.D.F.); Bettencourt Schueller Foundation (to L.T.); the APLNR as markers of endothelial cell injury in systemic sclerosis skin. Front.
Lundbeck Foundation (R219–2016-1375) and DFF Sapere Aude Starting Grant Immunol. 9, 2191.
(8048-00072A) (to L.L.); University of Antwerp (to L.-A.T., in part); Strategisch €ppel-like transcription factors in
Atkins, G.B., and Jain, M.K. (2007). Role of Kru
Basisonderzoek FWO-Vlaanderen (to V.G.); the Austrian Science Fund (FWF) endothelial biology. Circ. Res. 100, 1686–1695.
(J3730-B26; to A.P.); the Else Kröner-Fresenius-Stiftung and the Fritz Thyssen
Aurich, M.K., Fleming, R.M.T., and Thiele, I. (2016). MetaboTools: a compre-
Stiftung (10.16.2.017MN; to L.-C.C.); Kom op Tegen Kanker (to S.K.); the
hensive toolbox for analysis of genome-scale metabolic models. Front.
Sanming Project of Medicine in Shenzhen (SZSM201612074), BGI-Research,
Physiol. 7, 327.
the Danish Research Council for Independent Research (DFF–1337–00128),
the Sapere Aude Young Research Talent Prize (DFF-1335–00763A), and Aar- Barankiewicz, J., Kaplinsky, C., and Cohen, A. (1986). Modification of ribonu-
hus University Strategic Grant (AU-iCRISPR) (to L.B. and Y. Luo); the State Key cleotide and deoxyribonucleotide metabolism in interferon-treated human B-
Laboratory of Ophthalmology, Zhongshan Ophthalmic Center at the Sun Yat- lymphoblastoid cells. J. Interferon Res. 6, 717–727.
Sen University, China, and the National Natural Science Foundation of China Baydoun, A.R., Emery, P.W., Pearson, J.D., and Mann, G.E. (1990). Substrate-
(81670855), a Guangdong Province Leading Expert Program grant, and the dependent regulation of intracellular amino acid concentrations in cultured
Key Program of Guangzhou Scientific Research Plan (3030901006074) (to bovine aortic endothelial cells. Biochem. Biophys. Res. Commun. 173,
X.L.); and the VIB TechWatch Program, long-term structural Methusalem fund- 940–948.
ing from the Flemish Government, FWO, Foundation against Cancer (2016-
Becker, S.A., and Palsson, B.O. (2008). Context-specific metabolic networks
078), Kom op Tegen Kanker, ERC Proof of Concept (ERC-713758), and ERC
are consistent with experiments. PLoS Comput. Biol. 4, e1000082.
Advanced Research Grant (EU-ERC743074) (to P.C.).
Bjorklund, G., Svanberg, E., Dadar, M., Card, D.J., Chirumbolo, S., Harrington,
D.J., and Aaseth, J. (2018). The role of matrix Gla protein (MGP) in vascular
AUTHOR CONTRIBUTIONS
calcification. Curr. Med. Chem. Published July 15, 2018. https://doi.org/10.
K.R. and J.G. designed and analyzed all experiments. M.G.-C. performed 2174/0929867325666180716104159.
in vivo experiments. A.S. developed GEMs. J.G., J.K., M.G.-C., L.T., A.P., Blaauwgeers, H.G., Holtkamp, G.M., Rutten, H., Witmer, A.N., Koolwijk, P.,
and L.-C.C. set up endothelial cell isolation protocols. L.T. and K.D.F. per- Partanen, T.A., Alitalo, K., Kroon, M.E., Kijlstra, A., van Hinsbergh, V.W., and
formed in vitro experiments. F.T. performed bioinformatics analysis and imple- Schlingemann, R.O. (1999). Polarized vascular endothelial growth factor
mented the online databases. N.E. provided human choroid samples. J.K., secretion by human retinal pigment epithelium and localization of vascular
L.P.M.H.d.R., Y.Z., L. Sokol, V.G., D.P., F.D.S., F.J.T.S., R.J.M., and Y. Liu per- endothelial growth factor receptors on the inner choriocapillaris. Evidence
formed CyTOF measurements. K.R., J.K., M.G.-C., L.T., L. Sokol, L.-A.T., V.G., for a trophic paracrine relation. Am. J. Pathol. 155, 421–428.
and L.-C.C. prepared scRNA-seq samples. L.L., B.T., and Y. Luo performed
Bradshaw, A.D., and Sage, E.H. (2001). SPARC, a matricellular protein that
10x single-cell sequencing. S.K. processed scRNA-seq data. S.S. and G.C.
functions in cellular differentiation and tissue response to injury. J. Clin.
performed hydroxyproline measurements. F.D.S., S.V., T.V.B., G.E., P.D.H.,
Invest. 107, 1049–1054.
M.D., L. Schoonjans, J.W., L.B., H.Y., B.T., X.L., and Y. Luo provided advice
and discussed results. K.R., J.G., and P.C. wrote the manuscript. P.C. Bruning, U., Morales-Rodriguez, F., Kalucka, J., Goveia, J., Taverna, F.,
conceptualized the study. All authors discussed results and commented on Queiroz, K.C.S., Dubois, C., Cantelmo, A.R., Chen, R., Loroch, S., et al.
the manuscript. (2018). Impairment of angiogenesis by fatty acid synthase inhibition involves
mTOR malonylation. Cell Metab. 28, 866–880.e15.
Cannoodt, R., Saelens, W., Sichien, D., Tavernier, S., Janssens, S., Guilliams,
DECLARATION OF INTERESTS
M., Lambrecht, B., Preter, K.D., and Saeys, Y. (2016). SCORPIUS improves
The authors declare no competing interests. trajectory inference and identifies novel modules in dendritic cell development.
bioRxiv. https://doi.org/10.1101/079509.
Received: July 11, 2019 Cantelmo, A.R., Conradi, L.C., Brajic, A., Goveia, J., Kalucka, J., Pircher, A.,
Revised: December 20, 2019 Chaturvedi, P., Hol, J., Thienpont, B., Teuwen, L.A., et al. (2016). Inhibition
Accepted: March 9, 2020 of the glycolytic activator PFKFB3 in endothelium induces tumor vessel
Published: April 7, 2020 normalization, impairs metastasis, and improves chemotherapy. Cancer Cell
30, 968–985.
SUPPORTING CITATIONS Carmeliet, P., and Jain, R.K. (2011). Molecular mechanisms and clinical appli-
cations of angiogenesis. Nature 473, 298–307.
The following references appear in the Supplemental Information: Atkins and
Cerqueira, N.M.F.S.A., Oliveira, E.F., Gesto, D.S., Santos-Martins, D., Moreira,
Jain (2007); Bjorklund et al. (2018); Carmeliet and Jain (2011); Chen (2009);
C., Moorthy, H.N., Ramos, M.J., and Fernandes, P.A. (2016). Cholesterol
del Toro et al. (2010); dela Paz and D’Amore (2009); Epstein (2016); Hazell
biosynthesis: a mechanistic overview. Biochemistry 55, 5483–5506.
et al. (2016); McCandless et al. (2008); McCormick et al. (2001); Naschberger
et al. (2016); Neve et al. (2014); Patel et al. (2017); Pusztaszeri et al. (2006); Chambers, M.C., Maclean, B., Burke, R., Amodei, D., Ruderman, D.L.,
Roca and Adams (2007); Sardana et al. (2017); Strasser et al. (2010); Takaba- Neumann, S., Gatto, L., Fischer, B., Pratt, B., Egertson, J., et al. (2012). A
take et al. (2009); Thiriot et al. (2017); Vanlandewijck et al. (2018); Vartanian and cross-platform toolkit for mass spectrometry and proteomics. Nat.
Weidner (1994); Wagenseil and Mecham (2012); Zanetta et al. (2000); Zhao Biotechnol. 30, 918–920.
et al. (2018). Chen, L. (2009). Ocular lymphatics: state-of-the-art review. Lymphology
42, 66–76.
REFERENCES Corada, M., Orsenigo, F., Morini, M.F., Pitulescu, M.E., Bhat, G., Nyqvist, D.,
Breviario, F., Conti, V., Briot, A., Iruela-Arispe, M.L., et al. (2013). Sox17 is
Aibar, S., González-Blas, C.B., Moerman, T., Huynh-Thu, V.A., Imrichova, H., indispensable for acquisition and maintenance of arterial identity. Nat.
Hulselmans, G., Rambow, F., Marine, J.C., Geurts, P., Aerts, J., et al. (2017). Commun. 4, 2609.
SCENIC: single-cell regulatory network inference and clustering. Nat. Corey, D.M., Rinkevich, Y., and Weissman, I.L. (2016). Dynamic patterns of
Methods 14, 1083–1086. clonal evolution in tumor vasculature underlie alterations in lymphocyte-endo-
Ambati, J., and Fowler, B.J. (2012). Mechanisms of age-related macular thelial recognition to foster tumor immune escape. Cancer Res. 76,
degeneration. Neuron 75, 26–39. 1348–1353.

874 Cell Metabolism 31, 862–877, April 7, 2020


Cox, J., and Mann, M. (2008). MaxQuant enables high peptide identification Haraldsdóttir, H.S., Cousins, B., Thiele, I., Fleming, R.M.T., and Vempala, S.
rates, individualized p.p.b.-range mass accuracies and proteome-wide pro- (2017). CHRR: coordinate hit-and-run with rounding for uniform sampling of
tein quantification. Nat. Biotechnol. 26, 1367–1372. constraint-based models. Bioinformatics 33, 1741–1743.
Creemers, L.B., Jansen, D.C., van Veen-Reurings, A., van den Bos, T., and Hart, T., Komori, H.K., LaMere, S., Podshivalova, K., and Salomon, D.R.
Everts, V. (1997). Microassay for the assessment of low levels of hydroxypro- (2013). Finding the active genes in deep RNA-seq gene expression studies.
line. Biotechniques 22, 656–658. BMC Genomics 14, 778.
De Bock, K., Georgiadou, M., Schoors, S., Kuchnio, A., Wong, B.W., Hazell, G.G., Peachey, A.M., Teasdale, J.E., Sala-Newby, G.B., Angelini, G.D.,
Cantelmo, A.R., Quaegebeur, A., Ghesquière, B., Cauwenberghs, S., Eelen, Newby, A.C., and White, S.J. (2016). PI16 is a shear stress and inflammation-
G., et al. (2013). Role of PFKFB3-driven glycolysis in vessel sprouting. Cell regulated inhibitor of MMP2. Sci. Rep. 6, 39553.
154, 651–663. Heindl, L.M., Kaser-Eichberger, A., Schlereth, S.L., Bock, F., Regenfuss, B.,
de Lichtenberg, U., Jensen, L.J., Fausbøll, A., Jensen, T.S., Bork, P., and Reitsamer, H.A., McMenamin, P., Lutty, G.A., Maruyama, K., Chen, L., et al.
Brunak, S. (2005). Comparison of computational methods for the identification (2015). Sufficient evidence for lymphatics in the developing and adult human
of cell cycle-regulated genes. Bioinformatics 21, 1164–1171. choroid? Invest. Ophthalmol. Vis. Sci. 56, 6709–6710.
del Toro, R., Prahst, C., Mathivet, T., Siegfried, G., Kaminker, J.S., Larrivee, B., Heirendt, L., Arreckx, S., Pfau, T., Mendoza, S.N., Richelle, A., Heinken, A.,
Breant, C., Duarte, A., Takakura, N., Fukamizu, A., et al. (2010). Identification Haraldsdóttir, H.S., Wachowiak, J., Keating, S.M., Vlasov, V., et al. (2019).
and functional analysis of endothelial tip cell-enriched genes. Blood 116, Creation and analysis of biochemical constraint-based models using the
4025–4033. COBRA Toolbox v.3.0. Nat. Protoc. 14, 639–702.
dela Paz, N.G., and D’Amore, P.A. (2009). Arterial versus venous endothelial Hong, F., Breitling, R., McEntee, C.W., Wittner, B.S., Nemhauser, J.L., and
cells. Cell Tissue Res. 335, 5–16. Chory, J. (2006). RankProd: a bioconductor package for detecting differentially
Desouki, A.A., Jarre, F., Gelius-Dietrich, G., and Lercher, M.J. (2015). expressed genes in meta-analysis. Bioinformatics 22, 2825–2827.
CycleFreeFlux: efficient removal of thermodynamically infeasible loops from Huang, H., Vandekeere, S., Kalucka, J., Bierhansl, L., Zecchin, A., Bru €ning, U.,
flux distributions. Bioinformatics 31, 2159–2165. Visnagri, A., Yuldasheva, N., Goveia, J., Cruys, B., et al. (2017). Role of gluta-
Diebold, L.P., Gil, H.J., Gao, P., Martinez, C.A., Weinberg, S.E., and Chandel, mine and interlinked asparagine metabolism in vessel formation. EMBO J. 36,
N.S. (2019). Mitochondrial complex III is necessary for endothelial cell prolifer- 2334–2352.
ation during angiogenesis. Nat Metab 1, 158–171. Hwangbo, C., Wu, J., Papangeli, I., Adachi, T., Sharma, B., Park, S., Zhao, L.,
Duarte, N.C., Becker, S.A., Jamshidi, N., Thiele, I., Mo, M.L., Vo, T.D., Srivas, Ju, H., Go, G.W., Cui, G., et al. (2017). Endothelial APLNR regulates tissue fatty
R., and Palsson, B.Ø. (2007). Global reconstruction of the human metabolic acid uptake and is essential for apelin’s glucose-lowering effects. Sci. Transl.
network based on genomic and bibliomic data. Proc. Natl. Acad. Sci. USA Med. 9, https://doi.org/10.1126/scitranslmed.aad4000.
104, 1777–1782. Iñiguez, M.A., Rodrı́guez, A., Volpert, O.V., Fresno, M., and Redondo, J.M.
Dvorak, H.F. (1986). Tumors: wounds that do not heal. Similarities between tu- (2003). Cyclooxygenase-2: a therapeutic target in angiogenesis. Trends Mol.
mor stroma generation and wound healing. N. Engl. J. Med. 315, 1650–1659. Med. 9, 73–78.

Eales, K.L., Hollinshead, K.E., and Tennant, D.A. (2016). Hypoxia and meta- Jain, R.K. (2014). Antiangiogenesis strategies revisited: from starving tumors
bolic adaptation of cancer cells. Oncogenesis 5, e190. to alleviating hypoxia. Cancer Cell 26, 605–622.

Eastoe, J.E. (1955). The amino acid composition of mammalian collagen and Jeong, H.W., Hernández-Rodrı́guez, B., Kim, J., Kim, K.P., Enriquez-Gasca,
gelatin. Biochem. J. 61, 589–600. R., Yoon, J., Adams, S., Schöler, H.R., Vaquerizas, J.M., and Adams, R.H.
(2017). Transcriptional regulation of endothelial cell behavior during sprouting
Eelen, G., de Zeeuw, P., Treps, L., Harjes, U., Wong, B.W., and Carmeliet, P.
angiogenesis. Nat. Commun. 8, 726.
(2018). Endothelial cell metabolism. Physiol. Rev. 98, 3–58.
€ning, U.,
Kalucka, J., Bierhansl, L., Conchinha, N.V., Missiaen, R., Elia, I., Bru
Epstein, M. (2016). Matrix Gla-protein (MGP) not only inhibits calcification in
Scheinok, S., Treps, L., Cantelmo, A.R., Dubois, C., et al. (2018). Quiescent
large arteries but also may be renoprotective: connecting the dots.
endothelial cells upregulate fatty acid b-oxidation for vasculoprotection via
EBioMedicine 4, 16–17.
redox homeostasis. Cell Metab. 28, 881–894.e13.
Félétou, M., Huang, Y., and Vanhoutte, P.M. (2011). Endothelium-mediated
Kanda, T., Brown, J.D., Orasanu, G., Vogel, S., Gonzalez, F.J., Sartoretto, J.,
control of vascular tone: COX-1 and COX-2 products. Br. J. Pharmacol. 164,
Michel, T., and Plutzky, J. (2009). PPARgamma in the endothelium regulates
894–912.
metabolic responses to high-fat diet in mice. J. Clin. Invest. 119, 110–124.
Fitzgerald, G., Soro-Arnaiz, I., and De Bock, K. (2018). The Warburg effect in
Khan, S., Taverna, F., Rohlenova, K., Treps, L., Geldhof, V., de Rooij, L., Sokol,
endothelial cells and its potential as an anti-angiogenic target in cancer.
L., Pircher, A., Conradi, L.C., Kalucka, J., et al. (2019). EndoDB: a database of
Front. Cell Dev. Biol. 6, 100.
endothelial cell transcriptomics data. Nucleic Acids Res. 47, D736–D744.
Folkman, J. (1982). Angiogenesis: initiation and control. Ann. N Y Acad. Sci.
Kim, M.K., and Lun, D.S. (2014). Methods for integration of transcriptomic data
401, 212–227.
in genome-scale metabolic models. Comput. Struct. Biotechnol. J. 11, 59–65.
Garcı́a-Caballero, M., Zecchin, A., Souffreau, J., Truong, A.-C.K., Teuwen,
Koina, M.E., Baxter, L., Adamson, S.J., Arfuso, F., Hu, P., Madigan, M.C., and
L.-A., Vermaelen, W., Martı́n-Pérez, R., de Zeeuw, P., Bouché, A., Vinckier,
Chan-Ling, T. (2015). Evidence for lymphatics in the developing and adult hu-
S., et al. (2019). Role and therapeutic potential of dietary ketone bodies in
man choroid. Invest. Ophthalmol. Vis. Sci. 56, 1310–1327.
lymph vessel growth. Nature Metabolism 1, 666–675.
Goveia, J., Rohlenova, K., Taverna, F., Treps, L., Conradi, L.C., Pircher, A., Lambert, V., Lecomte, J., Hansen, S., Blacher, S., Gonzalez, M.L., Struman, I.,
Geldhof, V., de Rooij, L.P.M.H., Kalucka, J., Sokol, L., et al. (2020). An inte- Sounni, N.E., Rozet, E., de Tullio, P., Foidart, J.M., et al. (2013). Laser-induced
grated gene expression landscape profiling approach to identify lung tumor choroidal neovascularization model to study age-related macular degenera-
endothelial cell heterogeneity and angiogenic candidates. Cancer Cell 37, tion in mice. Nat. Protoc. 8, 2197–2211.
21–36.e13. Langmead, B., and Salzberg, S.L. (2012). Fast gapped-read alignment with
Haghverdi, L., Lun, A.T.L., Morgan, M.D., and Marioni, J.C. (2018). Batch ef- Bowtie 2. Nat. Methods 9, 357–359.
fects in single-cell RNA-sequencing data are corrected by matching mutual Levandowsky, M., and Winter, D. (1971). Distance between Sets. Nature
nearest neighbors. Nat. Biotechnol. 36, 421–427. 234, 34.
€nzelmann, S., Castelo, R., and Guinney, J. (2013). GSVA: gene set variation
Ha Li, X., Kumar, A., and Carmeliet, P. (2019). Metabolic pathways fueling the
analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7. endothelial cell drive. Annu. Rev. Physiol. 81, 483–503.

Cell Metabolism 31, 862–877, April 7, 2020 875


Luo, W., and Brouwer, C. (2013). Pathview: an R/Bioconductor package for progenitors versus mature endothelial cells reveals key SoxF-dependent dif-
pathway-based data integration and visualization. Bioinformatics 29, ferentiation process. Circulation 135, 786–805.
1830–1831. Patella, F., Schug, Z.T., Persi, E., Neilson, L.J., Erami, Z., Avanzato, D.,
Lyssiotis, C.A., and Kimmelman, A.C. (2017). Metabolic interactions in the tu- Maione, F., Hernandez-Fernaud, J.R., Mackay, G., Zheng, L., et al. (2015).
mor microenvironment. Trends Cell Biol. 27, 863–875. Proteomics-based metabolic modeling reveals that fatty acid oxidation
Mahadevan, R., and Schilling, C.H. (2003). The effects of alternate optimal so- (FAO) controls endothelial cell (EC) permeability. Mol. Cell. Proteomics 14,
lutions in constraint-based genome-scale metabolic models. Metab. Eng. 5, 621–634.
264–276. Perez-Riverol, Y., Csordas, A., Bai, J., Bernal-Llinares, M., Hewapathirana, S.,
Manavski, Y., Lucas, T., Glaser, S.F., Dorsheimer, L., Gu€nther, S., Braun, T., Kundu, D.J., Inuganti, A., Griss, J., Mayer, G., Eisenacher, M., et al. (2019). The
Rieger, M.A., Zeiher, A.M., Boon, R.A., and Dimmeler, S. (2018). Clonal expan- PRIDE database and related tools and resources in 2019: improving support
sion of endothelial cells contributes to ischemia-induced neovascularization. for quantification data. Nucleic Acids Res. 47 (D1), D442–D450.
Circ. Res. 122, 670–677. Phang, J.M. (2019). Proline metabolism in cell regulation and cancer biology:
Marneros, A.G., She, H., Zambarakji, H., Hashizume, H., Connolly, E.J., Kim, I., recent advances and hypotheses. Antioxid. Redox Signal. 30, 635–649.
Gragoudas, E.S., Miller, J.W., and Olsen, B.R. (2007). Endogenous endostatin Potente, M., Gerhardt, H., and Carmeliet, P. (2011). Basic and therapeutic as-
inhibits choroidal neovascularization. FASEB J. 21, 3809–3818. pects of angiogenesis. Cell 146, 873–887.
McCandless, E.E., Piccio, L., Woerner, B.M., Schmidt, R.E., Rubin, J.B.,
Pusztaszeri, M.P., Seelentag, W., and Bosman, F.T. (2006).
Cross, A.H., and Klein, R.S. (2008). Pathological expression of CXCL12 at
Immunohistochemical expression of endothelial markers CD31, CD34, von
the blood-brain barrier correlates with severity of multiple sclerosis. Am. J.
Willebrand factor, and Fli-1 in normal human tissues. J. Histochem.
Pathol. 172, 799–808.
Cytochem. 54, 385–395.
McCormick, S.M., Eskin, S.G., McIntire, L.V., Teng, C.L., Lu, C.M., Russell,
Rastogi, B.K., and Nordøy, A. (1980). Lipid composition of cultured human
C.G., and Chittur, K.K. (2001). DNA microarray reveals changes in gene
endothelial cells. Thromb. Res. 18, 629–641.
expression of shear stressed human umbilical vein endothelial cells. Proc.
Natl. Acad. Sci. USA 98, 8955–8960. Red-Horse, K., Ueno, H., Weissman, I.L., and Krasnow, M.A. (2010). Coronary
arteries form by developmental reprogramming of venous cells. Nature 464,
McDonald, A.I., Shirali, A.S., Aragón, R., Ma, F., Hernandez, G., Vaughn, D.A.,
549–553.
Mack, J.J., Lim, T.Y., Sunshine, H., Zhao, P., et al. (2018). Endothelial regen-
eration of large vessels is a biphasic process driven by local cells with distinct Ritchie, M.E., Phipson, B., Wu, D., Hu, Y., Law, C.W., Shi, W., and Smyth, G.K.
proliferative capacities. Cell Stem Cell 23, 210–225.e6. (2015). limma powers differential expression analyses for RNA-sequencing
and microarray studies. Nucleic Acids Res. 43, e47.
McGarrity, S., Anuforo, Ó., Halldórsson, H., Bergmann, A., Halldórsson, S.,
Palsson, S., Henriksen, H.H., Johansson, P.I., and Rolfsson, Ó. (2018). Roca, C., and Adams, R.H. (2007). Regulation of vascular morphogenesis by
Metabolic systems analysis of LPS induced endothelial dysfunction applied Notch signaling. Genes Dev. 21, 2511–2524.
to sepsis patient stratification. Sci. Rep. 8, 6811. Ryu, J.Y., Kim, H.U., and Lee, S.Y. (2015). Reconstruction of genome-scale
McLeod, D.S., Lefer, D.J., Merges, C., and Lutty, G.A. (1995). Enhanced human metabolic models using omics data. Integr. Biol. 7, 859–868.
expression of intracellular adhesion molecule-1 and P-selectin in the diabetic Santos, C.R., and Schulze, A. (2012). Lipid metabolism in cancer. FEBS J. 279,
human retina and choroid. Am. J. Pathol. 147, 642–653. 2610–2623.
Mondor, I., Jorquera, A., Sene, C., Adriouch, S., Adams, R.H., Zhou, B.,
Santos, A., Wernersson, R., and Jensen, L.J. (2015). Cyclebase 3.0: a multi-or-
Wienert, S., Klauschen, F., and Bajénoff, M. (2016). Clonal proliferation and
ganism database on cell-cycle regulation and phenotypes. Nucleic Acids Res.
stochastic pruning orchestrate lymph node vasculature remodeling.
43, D1140–D1144.
Immunity 45, 877–888.
Sardana, M., Vasim, I., Varakantam, S., Kewan, U., Tariq, A., Koppula, M.R.,
Muir, A., Danai, L.V., and Vander Heiden, M.G. (2018). Microenvironmental
Syed, A.A., Beraun, M., Drummen, N.E., Vermeer, C., et al. (2017). Inactive ma-
regulation of cancer cell metabolism: implications for experimental design
trix Gla-protein and arterial stiffness in type 2 diabetes mellitus. Am. J.
and translational studies. Dis. Model. Mech. 11, https://doi.org/10.1242/
Hypertens. 30, 196–201.
dmm.035758.
Satija, R., Farrell, J.A., Gennert, D., Schier, A.F., and Regev, A. (2015). Spatial
Murphy, E.J., Joseph, L., Stephens, R., and Horrocks, L.A. (1992).
reconstruction of single-cell gene expression data. Nat. Biotechnol. 33,
Phospholipid composition of cultured human endothelial cells. Lipids 27,
495–502.
150–153.
Sawada, N., and Arany, Z. (2017). Metabolic regulation of angiogenesis in dia-
Naito, H., Kidoya, H., Sakimoto, S., Wakabayashi, T., and Takakura, N. (2012).
betes and aging. Physiology (Bethesda) 32, 290–307.
Identification and characterization of a resident vascular stem/progenitor cell
population in preexisting blood vessels. EMBO J. 31, 842–855. Schoors, S., De Bock, K., Cantelmo, A.R., Georgiadou, M., Ghesquière, B.,
€tz, M., Britzen-Laurent, N.,
Naschberger, E., Liebl, A., Schellerer, V.S., Schu Cauwenberghs, S., Kuchnio, A., Wong, B.W., Quaegebeur, A., Goveia, J.,
Kölbel, P., Schaal, U., Haep, L., Regensburger, D., Wittmann, T., et al. et al. (2014). Partial and transient reduction of glycolysis by PFKFB3 blockade
(2016). Matricellular protein SPARCL1 regulates tumor microenvironment- reduces pathological angiogenesis. Cell Metab. 19, 37–48.
dependent endothelial cell heterogeneity in colorectal carcinoma. J. Clin. Schoors, S., Bruning, U., Missiaen, R., Queiroz, K.C., Borgers, G., Elia, I.,
Invest. 126, 4187–4204. Zecchin, A., Cantelmo, A.R., Christen, S., Goveia, J., et al. (2015). Fatty acid
Neve, A., Cantatore, F.P., Maruotti, N., Corrado, A., and Ribatti, D. (2014). carbon is essential for dNTP synthesis in endothelial cells. Nature 520,
Extracellular matrix modulates angiogenesis in physiological and pathological 192–197.
conditions. BioMed Res. Int. 2014, 756078. Scialdone, A., Natarajan, K.N., Saraiva, L.R., Proserpio, V., Teichmann, S.A.,
O’Brien, E.J., Monk, J.M., and Palsson, B.O. (2015). Using genome-scale Stegle, O., Marioni, J.C., and Buettner, F. (2015). Computational assignment
models to predict biological capabilities. Cell 161, 971–987. of cell-cycle stage from single-cell transcriptome data. Methods 85, 54–61.
Pagliarini, R., Castello, R., Napolitano, F., Borzone, R., Annunziata, P., Semenza, G.L. (2011). Hypoxia-inducible factor 1: regulator of mitochondrial
Mandrile, G., De Marchi, M., Brunetti-Pierri, N., and di Bernardo, D. (2016). metabolism and mediator of ischemic preconditioning. Biochim. Biophys.
In silico modeling of liver metabolism in a human disease reveals a key enzyme Acta 1813, 1263–1268.
for histidine and histamine homeostasis. Cell Rep. 15, 2292–2300. Shlomi, T., Benyamini, T., Gottlieb, E., Sharan, R., and Ruppin, E. (2011).
Patel, J., Seppanen, E.J., Rodero, M.P., Wong, H.Y., Donovan, P., Neufeld, Z., Genome-scale metabolic modeling elucidates the role of proliferative adapta-
Fisk, N.M., Francois, M., and Khosrotehrani, K. (2017). Functional definition of tion in causing the Warburg effect. PLoS Comput. Biol. 7, e1002018.

876 Cell Metabolism 31, 862–877, April 7, 2020


Stoilov, I., Starcher, B.C., Mecham, R.P., and Broekelmann, T.J. (2018). Wakabayashi, T., Naito, H., Takara, K., Kidoya, H., Sakimoto, S., Oshima, Y.,
Measurement of elastin, collagen, and total protein levels in tissues. Nishida, K., and Takakura, N. (2013). Identification of vascular endothelial side
Methods Cell Biol. 143, 133–146. population cells in the choroidal vessels and their potential role in age-related
Strasser, G.A., Kaminker, J.S., and Tessier-Lavigne, M. (2010). Microarray macular degeneration. Invest. Ophthalmol. Vis. Sci. 54, 6686–6693.
analysis of retinal endothelial tip cells identifies CXCR4 as a mediator of tip Wakabayashi, T., Naito, H., Suehiro, J.I., Lin, Y., Kawaji, H., Iba, T., Kouno, T.,
cell morphology and branching. Blood 115, 5102–5110. Ishikawa-Kato, S., Furuno, M., Takara, K., et al. (2018). CD157 marks tissue-
resident endothelial stem cells with homeostatic and regenerative properties.
Swainston, N., Smallbone, K., Hefzi, H., Dobson, P.D., Brewer, J., Hanscho,
Cell Stem Cell 22, 384–397.e6.
M., Zielinski, D.C., Ang, K.S., Gardiner, N.J., Gutierrez, J.M., et al. (2016).
Recon 2.2: from reconstruction to model of human metabolism. Welti, J., Loges, S., Dimmeler, S., and Carmeliet, P. (2013). Recent molecular
Metabolomics 12, 109. discoveries in angiogenesis and antiangiogenic therapies in cancer. J. Clin.
Invest. 123, 3190–3200.
Takabatake, Y., Sugiyama, T., Kohara, H., Matsusaka, T., Kurihara, H., Koni,
P.A., Nagasawa, Y., Hamano, T., Matsui, I., Kawada, N., et al. (2009). The Whitfield, M.L., Sherlock, G., Saldanha, A.J., Murray, J.I., Ball, C.A., Alexander,
CXCL12 (SDF-1)/CXCR4 axis is essential for the development of renal vascu- K.E., Matese, J.C., Perou, C.M., Hurt, M.M., Brown, P.O., and Botstein, D.
lature. J. Am. Soc. Nephrol. 20, 1714–1723. (2002). Identification of genes periodically expressed in the human cell cycle
and their expression in tumors. Mol. Biol. Cell 13, 1977–2000.
Thiele, I., and Palsson, B.O. (2010). A protocol for generating a high-quality
Yang, S., Zhao, J., and Sun, X. (2016). Resistance to anti-VEGF therapy in neo-
genome-scale metabolic reconstruction. Nat. Protoc. 5, 93–121.
vascular age-related macular degeneration: a comprehensive review. Drug
Thiriot, A., Perdomo, C., Cheng, G., Novitzky-Basso, I., McArdle, S., Des. Devel. Ther. 10, 1857–1867.
Kishimoto, J.K., Barreiro, O., Mazo, I., Triboulet, R., Ley, K., et al. (2017).
Ying, Y., Ueta, T., Jiang, S., Lin, H., Wang, Y., Vavvas, D., Wen, R., Chen, Y.G.,
Differential DARC/ACKR1 expression distinguishes venular from non-venular
and Luo, Z. (2017). Metformin inhibits ALK1-mediated angiogenesis via activa-
endothelial cells in murine tissues. BMC Biol. 15, 45.
tion of AMPK. Oncotarget 8, 32794–32806.
van den Brink, S.C., Sage, F., Vértesy, Á., Spanjaard, B., Peterson-Maduro, J.,
You, L.R., Lin, F.J., Lee, C.T., DeMayo, F.J., Tsai, M.J., and Tsai, S.Y. (2005).
Baron, C.S., Robin, C., and van Oudenaarden, A. (2017). Single-cell
Suppression of Notch signalling by the COUP-TFII transcription factor regu-
sequencing reveals dissociation-induced gene expression in tissue subpopu-
lates vein identity. Nature 435, 98–104.
lations. Nat. Methods 14, 935–936.
Yu, G., Wang, L.G., Han, Y., and He, Q.Y. (2012). clusterProfiler: an R package
Vandekeere, S., Dubois, C., Kalucka, J., Sullivan, M.R., Garcı́a-Caballero, M., for comparing biological themes among gene clusters. OMICS 16, 284–287.
Goveia, J., Chen, R., Diehl, F.F., Bar-Lev, L., Souffreau, J., et al. (2018). Serine
Yu, P., Alves, T.C., Kibbey, R.G., and Simons, M. (2018). Metabolic analysis of
synthesis via PHGDH is essential for heme production in endothelial cells. Cell
lymphatic endothelial cells. Methods Mol. Biol. 1846, 325–334.
Metab. 28, 573–587.e13.
Zanetta, L., Marcus, S.G., Vasile, J., Dobryansky, M., Cohen, H., Eng, K.,
Vanlandewijck, M., He, L., Ma €e, M.A., Andrae, J., Ando, K., Del Gaudio, F.,
Shamamian, P., and Mignatti, P. (2000). Expression of Von Willebrand factor,
Nahar, K., Lebouvier, T., Laviña, B., Gouveia, L., et al. (2018). A molecular atlas
an endothelial cell marker, is up-regulated by angiogenesis factors: a potential
of cell types and zonation in the brain vasculature. Nature 554, 475–480.
method for objective assessment of tumor angiogenesis. Int. J. Cancer 85,
Vartanian, R.K., and Weidner, N. (1994). Correlation of intratumoral endothelial 281–288.
cell proliferation with microvessel density (tumor angiogenesis) and tumor cell Zhao, Q., Eichten, A., Parveen, A., Adler, C., Huang, Y., Wang, W., Ding, Y.,
proliferation in breast carcinoma. Am. J. Pathol. 144, 1188–1194. Adler, A., Nevins, T., Ni, M., et al. (2018). Single-cell transcriptome analyses
Wagenseil, J.E., and Mecham, R.P. (2012). Elastin in large artery stiffness and reveal endothelial cell heterogeneity in tumors and changes following antian-
hypertension. J. Cardiovasc. Transl. Res. 5, 264–273. giogenic treatment. Cancer Res. 78, 2370–2382.

Cell Metabolism 31, 862–877, April 7, 2020 877


DON’T BE THE
LAST TO KNOW

Give your research a boost with alerts


from Cell Press. You’ll be glad you did.
Get first access with immediate, regular Exciting extra features like video abstracts,
electronic table of contents (eToCs) alerts podcasts, and blog posts give you additional
delivered directly to your desktop, free depth and context and you can access news
of charge, that keep you informed of and commentary, normally not accessible
breakthroughs in your field. online, in advance of publication.

Register today
Visit cell.com/alerts
Viruses in Health and Disease
October 24–26, 2021 — Sitges, Spain

This meeting will bring together research that encompasses basic and translational
Abstract virology. We envision this meeting showcasing the ubiquity of viruses while highlighting
submission commonalities among researchers, from technology through a standard goal of
deadline: understanding how viruses behave and adapt and how this may be exploited for
prediction of the next outbreak or leveraged for therapeutic development. The scientific
June 18, response to SARS-CoV-2/COVID-19 continues to showcase how science has
2021 progressed, as the rapidity of basic and translational research has been second to
none, while also revealing our weaknesses as a population. This meeting is both a
Early celebration of how far virology has come and a forecast of what is next and the inroads
registration to get us there.
deadline:
Our topics will include:
August 13, 1) Viruses Within: defining the virome; interaction with the microbiome;
2021 endogenous retroviruses in health and disease; tools and technologies
2) Viral Pathogens: evolution, genomics, spread, prediction, and pandemic
preparedness of emerging and re-emerging viruses
3) Virus-Host Interaction: immunology, genetics, and host factors
4) Viruses as Therapeutics: diagnostics, phage therapy, viral vectors, and
disease therapy
5) Targeting Viruses: Antivirals, Vaccines, Antibodies: lessons from the past;
successes versus failures

Organizers
Erica Saphire, La Jolla Institute of Immunology, USA
Kanta Subbarao, Doherty Institute, Australia
Amanda Monahan, Scientific Editor, Cell Host & Microbe
Sri Narasimhan, Deputy Editor, Cell

cell-symposia.com/Viruses-2021
cell.com/symposia
stands for interdisciplinary

iScience is an interdisciplinary
open access journal from Cell Press
iScienceW\ISPZOLZIHZPJHUKHWWSPLKYLZLHYJO[OH[HK]HUJLZHZWLJPÄJÄLSKHJYVZZSPMLWO`ZPJHS
HUKLHY[OZJPLUJLZ0[»ZHUVWLUHJJLZZQV\YUHS^P[OJVU[PU\V\ZW\ISPJH[PVUZVYLZLHYJOPZ
PTTLKPH[LS`HJJLZZPISL6\YUVUVUZLUZLHWWYVHJO[VZ\ITPZZPVUZPZZPTWSLMHZ[HUKMHPYHUK
V\YJVTTP[TLU[[VPU[LNYP[`TLHUZ^LW\ISPZO[YHUZWHYLU[TL[OVKZ^P[OOPNOLKP[VYPHSZ[HUKHYKZ

So if you’re looking for a place to be inspired by science, welcome home.


Submit your paper to iScience.

cell.com/iscience
pub_NanoSIMS_cell-press2_Cell press selection 03/11/2020 14:24 Page 3

Cutting-Edge Research
Instruments for Elemental
& Isotopic Microanalysis
IMS xf series SIMS NanoSIMS 50L LEAP 5000 APT
®

CAMECA IMS 7f series Secondary Ion The CAMECA NanoSIMS 50L is a The LEAP 5000 is CAMECA’s state-of-
Mass Spectrometers (SIMS) can map unique imaging Secondary Ion Mass the-art Atom Probe Tomography (APT)
major and trace species in biological Spectrometer (SIMS) enabling quanti- microscope delivering 3D sub-nanometer
samples with high sensitivity and a tative measurements as well as 2D and chemical analysis of wide variety of
lateral resolution reaching ~0.5 μm. 3D imaging of molecular distribution, samples. Specific specimen preparation
In a recent study, an IMS 7f was used to kinetics and fluxes in tissues or at intra- methods open the path to a large range
localize uranium at very low concen- cellular level through a strategy of stable of APT applications in life sciences.
tration levels within cells, showing for the isotope or rare element labelling.
first time the presence of soluble
uranium within the nuclei after 15 min 14
to 24 hours exposure to concentrations N
lower than 100 μM.

10 µm
Analysis of impurities in chiton teeth: APT
15
N/14N allows 3D atomic-scale analysis of these
impurities in the surrounding matrix in 3D
providing unique insights into the
arrangement of the atoms.
From D. Joester, et al. Organic Materials and Organic /
norganic Heterostructures in Atom Probe Tomography (2012)
Microscopy Today. Vol.20, Issue 3

10 µm

Cardiomyocyte cell cycle activity in the


exercised heart: 15N-thymidine (labelling
DNA) was administered for 8 weeks to
adult mice undergoing voluntary wheel
Uranium distribution inside HepG2 cells running versus sedentary activity. Mass 14N
after 30 min. exposure to 100 μM of uranyl image shows histological details, while
nitrate. Superposition of 238U+, 23Na+ and 15
N/14N image demonstrates nuclear 15N
40
Ca+ SIMS ion images and 238U mass labeling (red, 150% above natural ratio) of Scan the code
spectra. Red arrows indicate 238U a cardio-myocyte while the cytoplasm and or visit
precipitates. White circles delimit the other cells are at natural abundance level
nucleus. Field of view: 50 x 50 μm2. (blue, 0% above natural ratio).
cameca.com/focus/tuto
From from D. Suhard et al., From A. Vujic et al. Exercise induces new cardiomyocyte for free guides on
Microsc Res Tech. (2018) generation in the adult mammalian heart. (2018) SIMS & APT
DOI: 10.1002/jemt.23047 Nat Commun 2018;9:1659

www.cameca.com cameca.info@ametek.com
pub_NanoSIMS_cell-press2_Cell press selection 03/11/2020 14:24 Page 4

Upgrade your knowledge on


microanalytical techniques!

The Essential Guide on


Dynamic Secondary Ion Mass
Spectrometry (SIMS)
Still the most sensitive microanalytical technique available, Dynamic
SIMS provides localized elemental, isotopic and molecular characterization
of the sample surface.
Our free Dynamic SIMS guide provides a simple introduction to the basic principles of this essential
microanalytical technique, outlining its advantages over alternative surface analysis techniques. Four
case studies allow readers to explore current practices and applications of the NanoSIMS and other
dynamic SIMS instruments including research conducted at Harvard Medical School (see extract below).

Scan the code or visit


cameca.com/focus/sims-tuto
to download a free guide
on Dynamic SIMS!

Extract from Dynamic SIMS guide, case study Nr 4:


Protein turnover quantification by feeding animals with a 15N-labeled
precursor amino acid to measure appearance of new protein.
In contrast to previous studies, direct measurement with NanoSIMS
multi-isotope imaging mass spectrometry revealed unexpectedly low
turnover of protein in hair-cell stereocili. Measured volume: 8x8x2μm.

From: Multi-isotope imaging mass spectrometry reveals slow


protein turnover in hair-cell stereocilia. Duan-Sun Zhang et al.
NATURE, vol 481, 26 January 2012

cameca.info@ametek.com
www.cameca.com

You might also like