Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Biochem J. Author manuscript; available in PMC 2021 Feb 10.
Published in final edited form as:
PMCID: PMC7875330
NIHMSID: NIHMS1665107
PMID: 32026949

Single Cell Approaches to Address Adipose Tissue Stromal Cell Heterogeneity

Associated Data

Supplementary Materials

Abstract

A central function of adipose tissue is in the management of systemic energy homeostasis that is achieved through the coordinated regulation of energy storage and mobilization, adipokine release, and immune functions. With the dramatic increase in the prevalence of obesity and obesity-related metabolic disease over the past 30 years, there has been extensive interest in targeting adipose tissue for therapeutic benefit. However, in order for this goal to be achieved it is essential to establish a comprehensive atlas of adipose tissue cellular composition and define mechanisms of intercellular communication that mediate pathologic and therapeutic responses. While traditional methods, such as fluorescence-activated cell sorting (FACS) and genetic lineage tracing, have greatly advanced the field, these approaches are inherently limited by the choice of markers and the ability to comprehensively identify and characterize dynamic interactions among stromal cells within the tissue microenvironment. Single cell RNA sequencing (scRNAseq) has emerged as a powerful tool for deconvolving cellular heterogeneity and holds promise for understanding the development and plasticity of adipose tissue under normal and pathological conditions. scRNAseq has recently been used to characterize adipose stem cell (ASC) populations and has provided new insights into subpopulations of macrophages that arise during anabolic and catabolic remodeling in white adipose tissue. The current review summarizes recent findings that use this technology to explore adipose tissue heterogeneity and plasticity.

INTRODUCTION

We are in the midst of an epidemic of obesity-related diseases including diabetes, cardiovascular disease, and cancer (14). It is now clear that obesity itself, i.e., the excessive accumulation of fat tissue, is not the specific cause of obesity-related disease. Rather, obesity-related diseases occur when the environmental demands to store excess energy disrupts the integrative metabolic, endocrine and immune functions of adipose tissue (reviewed in (59)). Viewed from this perspective, an important therapeutic goal is to improve functioning of adipose tissue by augmenting its energy buffering capacity, improving the mixture of released adipokines, and/or reducing levels of persistent inflammation (7, 1017). Thus, central to the ability to therapeutically target adipose tissue is a more detailed understanding of the cellular composition (i.e., adipocytes, stromal cells and immune cells) among tissue depots and how this composition changes in response to physiological, nutritional, and environmental challenges.

Adipocyte cell number is thought to be determined by early adulthood, with homeostatic turnover estimated at ~8% per year in humans (18) and ~18% per month in mice (19). However, white adipose tissue (WAT) is highly plastic and can undergo extensive remodeling in response to metabolic, nutritional, and pharmacological challenges (reviewed in (2023)). During periods of excess energy intake, WAT expands by both hypertrophy (increase in cell size) and/or hyperplasia (increase in cell number) (2426), and differences in the anatomical localization and type of adipose tissue expansion has profound effects on metabolic health (2731). In general, hypertrophy in visceral adipose tissue (VAT) is linked more strongly to inflammation and insulin resistance than that in subcutaneous depots (3236). In rodent models, high fat feeding leads to adipocyte hypertrophy in both depots, whereas adipogenesis is thought to occur primarily in VAT (2426, 37). The ability of adipose tissue to expand by hyperplasia confers a protective advantage on insulin sensitivity and metabolic disease risk (3336, 38). During chronic overnutrition, adipocyte death coupled with accumulation of pro-inflammatory macrophages and fibrotic changes eventually leads to ectopic fat accumulation in muscle and liver with systemic insulin resistance (reviewed in (7, 20, 39, 40)). In addition to remodeling during overnutrition, exposure to cold temperatures, β-adrenergic receptor agonists, and a number of hormones and metabolites stimulate the appearance of thermogenic brown adipocytes (“beige” or brown in white, “brite”) within WAT (4154). Brown adipocytes are thought to exert a protective role on metabolism through increased fatty acid and glucose utilization (55). Brown adipocytes can arise from either existing white adipocytes in which the brown adipocyte program is reinstated (43, 44, 47, 48, 54), or via differentiation of new brown/beige adipocytes from progenitors, depending on the location, duration, and type of inductive stimuli (26, 46, 49, 54, 56).

Given its central role in metabolism, there is substantial interest in better defining cellular subtypes involved in adipose tissue homeostasis and the mechanisms that regulate in vivo adipogenesis, plasticity, and inflammation for therapeutic strategies to treat metabolic disease (22, 57). While adipocytes are the major functional cell type in adipose tissue and occupy the greatest mass and volume of the tissue (owing to stored triglyceride), these cells comprise fewer than ~20% of total cells (58). The remaining ~80% of cells is a complex mixture of poorly defined stromal cells (fibroblasts, vascular cells, adipocyte stem cells) and immune cells (5861). Importantly, these stromal and immune cell types play critical roles in the establishment and maintenance of parenchymal cell function. The composition of stromal cells varies across fat depots (24, 59), likely reflecting tissue specialization and differences in energy storage, vascularization, innervation, and metabolism (62). Traditional methods of addressing heterogeneity such as antibody-based cell sorting approaches (e.g., FACS) have provided useful information for some cellular populations (46, 63); however, this approach is limited by the number of antibodies available to characterize cell populations. Additionally, surface marker expression can be altered during distinct biological processes, as is the case with Pdgfra, which decreases in cells undergoing active adipogenesis (64). Similar challenges exist for immune cells that are dynamic, and whose marker expression can change depending on differentiation and activation states (65, 66). Other methods, such as bulk RNA-seq analysis of whole tissue and/or sorted cells provides a greater depth in gene expression profiles. However, this technology does not completely discriminate differences in gene expression due to shifts in cellular composition versus changes in cellular states (67, 68). Additionally, relatively minor or undefined cell types can be underrepresented or missed entirely (65, 67, 69). Therefore, an important barrier to understanding adipose tissue biology is the ability to define and characterize cellular subtypes and expression profiles in a comprehensive and unbiased fashion. In this regard, single cell RNA sequencing (scRNAseq) technology has recently emerged as a powerful tool to address tissue heterogeneity (65). This review highlights recent findings on the use of scRNAseq to address adipose tissue stromal and immune cell complexity in WAT, with a focus on studies in mouse models.

OVERVIEW OF scRNASEQ

High throughput single cell RNA technology has evolved as a powerful tool to deconvolve tissue heterogeneity (65, 68, 7074). The concept of scRNAseq is to characterize individual cells based on the expression profile of thousands of genes compared to only a handful of surface markers commonly used in FACS-based sorting (68, 75). One of the benefits of this technique is that no prior knowledge of tissue composition is required. Although subject to some technical noise, scRNAseq provides crucial information about cellular populations, gene expression profiles, as well as changes in cellular states that may arise under different physiological conditions (65, 75). Available scRNAseq platforms differ slightly in design (droplet vs plate-based), throughput, and information acquired (e.g., full length mRNA vs 5′ or 3′ digital end sequencing); for recent reviews see (65, 70, 7577). Droplet-based microfluidic technology, such as inDrop (71, 72), Drop-seq (73), and 10X Genomics (74) are among the most widely used scRNAseq platforms due to increased throughput and more automated protocols (77, 78). The technical aspects of droplet generation are similar among methods (78). Briefly, cells and barcoded beads are partitioned into separate channels of a microfluidic device, and each bead/cell complex is then mixed with oil to generate an emulsion containing nanoliter-sized single cell droplets (Fig. 1) (7174, 78). Each bead contains primers, cellular barcodes, and a unique molecular identifier (UMI) to provide transcript counts per cell (7174, 7880). Reverse transcription either takes place within the droplets (inDrop and 10X Genomics) or following demulsification and cell lysis (Drop-seq) (7174, 78). cDNA libraries are then prepared for sequencing through preamplification, tagmentation/fragmentation, and PCR indexing (Drop-seq, 10X Genomics) or by linear amplification (in vitro transcription, RNA fragmentation, and RT-PCR) for inDrop (7174, 78). A comparison among the different droplet-based technologies has recently been presented (78).

An external file that holds a picture, illustration, etc.
Object name is nihms-1665107-f0001.jpg
Schematic Diagram of Basic Steps in Droplet-Based Microfluidic scRNAseq.

Cells and DNA-barcoded beads are partitioned into a microfluidic device and mixed with oil to generate nanoliter-sized single cell droplets. Each bead contains primers, cellular barcodes, and a unique molecular identifier (UMI). Cells are lysed within droplets, and reverse transcription either takes place within the droplets or following primer annealing and demulsification. Downstream steps differ slightly between platforms (not shown) to generate libraries for sequencing. Following sequencing, reads are demultiplexed, aligned, and then UMI counts per cell are quantified. Modified from (78).

Numerous bioinformatic tools are available and continuously emerging for analyzing scRNAseq output (77, 79, 81). Standardized pipelines involve initial preprocessing of reads (demultiplexing, alignment, and quantification), normalization as well as filtering to remove technical noise, such as cells containing high mitochondrial counts and doublets (75, 77, 79, 82). This is followed by dimensionality reduction, visualization, and identification of cell clusters based on differential gene expression (75, 77, 79, 82). In addition to identifying major and minor cell types within complex tissues, advanced bioinformatic programs are available for analysis of cellular transitions/hierarchies (83, 84), inference of gene regulatory networks (85, 86), predictions of cell-cell interactions (87), and integrative analyses of diverse datasets (88, 89).

ADIPOSE STEM CELL POPULATIONS IN MAJOR WAT DEPOTS AS IDENTIFIED BY scRNASEQ

There is a general consensus that adipocyte progenitors within adult WAT express the mesenchymal surface markers Pdgfrα, CD34, and Sca-1 (Ly6a) (46, 63, 9092). Based on previous work, these progenitors are located in the interstitial and perivascular space and have a dendritic morphology with long processes that extend and contact several cells (46, 92). Lineage tracing experiments with constitutive and inducible Pdgfrα-Cre drivers indicates that most adipocytes arising in adult tissues are derived from Pdgfrα progenitors (90) and contribute to de novo adipogenesis both following high fat diets (HFD) and during catabolic tissue remodeling (46, 92). However, additional populations of adipocyte progenitors have also been described (38, 49, 9397). For example, adipogenesis is recognized to be tightly linked to vasculature development (93, 98101) and some, but not all, studies indicate adipocytes can be derived from endothelial or mural cells that also express Pparγ or Pdgfrβ (38, 49, 90, 9395). Hematopoietic-derived progenitor cells have also been shown to contribute to neogenesis as well as the heterogeneity of adipocytes within adipose tissue (96, 97).

More recent studies have used scRNAseq to deconvolve progenitor heterogeneity in adipose tissue during development and in adults, under basal conditions (64, 102104) and during catabolic remodeling induced by β3 adrenergic receptor (ADRB3) activation (64). A summary of the basic designs, including mouse strain, sex, and depots examined are presented in Table 1. In order to characterize the major cell populations present within non-immune stromal cells across studies, we integrated these diverse datasets using canonical correlation analysis (CCA) (89, 105). CCA is designed to reduce variability that can arise under different experimental conditions (e.g., cell isolation, mice strains, sex, and treatment) and clusters cell populations based on common or shared characteristics (89, 105). Approximately 11, 000 cells from eWAT and 28, 000 from iWAT were included in the integration (64, 102104). Where appropriate, selection markers that were used to further characterize ASC subtypes with cell sorting along with the original cluster identity from each study are also presented (Table 1). A proposed nomenclature for adipocyte stem cells (ASC) based on marker gene expression is also included to facilitate comparisons between studies (Table 1). Additionally, individual findings from each study are summarized and discussed separately below.

Table 1.

Summary of scRNAseq Studies Performed on Non-Immune Stromal Cells from White Adipose Tissue.

ReferenceWAT DepotSexAgeStrainStimulusscRNA Cell Sorting TechniqueASC Populations (identified by authors)Proposed ASC NomenclatureCell Sorting Strategy (Functional Assays)**
Burl et al. (64)eWATM8–10 WkC57BL6/JSHAM or 3 days CL-316,243
(0.75 nmol/hr)
Lineage Negative
(CD45-, CD45R-, CD11b-, Gr-1 (Ly-6G/C)-, 7–4-, and Ter-119-)
ASC1
ASC2
ASC1a
ASC2
ND
Hepler et al. (104)eWATM8 WkPdgfrbrtTA (C57BL6/J)None“Muralchaser”
tdTomato- mGFP+ SVC
APC
FIPs
ASC1a
ASC2
Pdgfrβ+Ly6C-CD9-Pdgfrβ+Ly6C+

Burl et al. (64)iWATM8–10 WkC57BL6/JSHAM or 3 days CL-316,243
(0.75 nmol/hr)
Lineage Negative
(CD45-, CD45R-, CD11b-, Gr-1 (Ly-6G/C)-, 7–4-, and Ter-119-)
ASC1
ASC1
ASC2
ASC1a
ASC1b
ASC2
ND
Merrick et al. (102)iWATM/FPND12C57BL6/JNoneLineage Negative
(CD45-)
Group2
Group3
Group1
ASC1a
ASC1b
ASC2
Icam+CD142-
CD142+
DPP4+CD142-
Merrick et al. (102)iWATM10 Wk129S6/SvEvNoneLineage Negative
(CD45-)
Group2
Group3
Group1
ASC1a
ASC1b
ASC2
Icam+CD142-
CD142+
DPP4+CD142-
Schwalie et al. (103)iWATM/F8 WkTg(Pref1-CreER)426 Biat (Pref1-CreER)NoneLineage Negative (CD31−CD45−Ter119−)G2 (P2*)
G3 (P3*)
G1/G4 (P1*)
ASC1a
ASC1b
ASC2
Vap1+Adam12+ CD142+Abcg1+ CD55+IL13ra+
*ASC groups in parenthesis refer to the author’s nomenclature for scRNA groups identified using the Fluidigm C1.
**Cell sorting strategy for Hepler et al. (104) and Merrick et al. (102) were selection markers after depleting CD45+CD31+ stromal vascular cells. See text for details of additional sorting strategies used by Schwalie et al. (103).

Abbr: Adipose stem cells (ASC), adipose progenitor cells (APC), fibro-inflammatory progenitors (FIP), stromal vascular cells (SVC).

Across studies, a majority of the non-immune stromal cell population detected by scRNAseq from each depot (>70%) were identified as ASC cells and express genes for previously identified surface markers including Pdgfrα (Pdgfra), CD29 (Itgb1), CD34 (Cd34), and Sca-1 (Ly6a) (Figs. 2B, ,3B)3B) (64, 102104). Two main populations of ASC (ASC1a and 2) are further distinguishable by scRNAseq, with an additional subpopulation (ASC1b) present in iWAT (64, 102104). Some of the ASC2 genes commonly expressed within both adipose depots include those associated with inflammation/prostaglandin synthesis (Ptgs2), angiogenesis/endothelial cell migration (Anxa3), proteolysis (Pi16, Dpp4), as well as immune signaling (Cd55) (Supplemental Figs. 2,3). Genes commonly expressed within the ASC1a population include those involved in lipid uptake/metabolism (Plpp1, G0S2, and Apoe) and the metalloreductase Steap4 (Supplemental Figs. 2,3). Several genes encoding extracellular matrix (ECM) components are also strongly expressed in both ASC2 and ACS1, however the type of ECM differs between populations (Supplemental Figs. 2,3). For example, Fn1 and Col14a1 are more enriched in ASC2 whereas Col15a1 and Col4a2 are enriched in ASC1 progenitors (Supplemental Figs. 2, 3) (102104). The relative proportion of total ACS1 to ASC2 in adult animals is also higher in eWAT than that observed in iWAT (Figs. 2A, ,3A),3A), and functional assays suggest that the ASC1a population is more poised for adipogenesis (102104).

An external file that holds a picture, illustration, etc.
Object name is nihms-1665107-f0002.jpg
Integrated scRNAseq of Immune-Depleted Stromal Cells Isolated from eWAT of Adult Mice.

Publicly available scRNAseq datasets (64, 104) from immune-depleted stromal cells isolated from eWAT were integrated using Seurat (105). (A) tSNE plots displaying the overlay of integrated datasets (left) and cell cluster identification (right). (B) tSNE plots displaying the log2 expression levels for select genes commonly used to purify adipose stem cells by FACS and/or used in lineage tracing experiments. (C) Violin plots displaying relative log2 expression levels of select marker genes in individual clusters. Abbr: Ly6a (stem cell antigen-1 (Sca-1)), Itgb1 (CD29).

An external file that holds a picture, illustration, etc.
Object name is nihms-1665107-f0003.jpg
Integrated scRNAseq of Immune-Depleted Stromal Cells Isolated from iWAT of Adult Mice.

Publicly available scRNAseq datasets (64, 102, 103) from immune-depleted stromal cells isolated from iWAT were integrated using Seurat (105). (A) tSNE plots displaying the overlay of integrated datasets (left) and cell cluster identification (right). (B) tSNE plots displaying the log2 expression levels for select genes commonly used to purify adipose stem cells by FACS and/or used in lineage tracing experiments. (C) Violin plots displaying relative log2 expression levels of select marker genes in individual clusters. Abbr: Ly6a (stem cell antigen-1 (Sca-1)), Itgb1 (CD29).

As expected, very few actively differentiating preadipocytes were detected in either depot (Figs. 2, ,3).3). In the study by Burl et al. (64), the proportion of proliferating (Birc5, Cdca8, Top2a) and differentiating preadipocytes (Cebpa, Adig, Plin1, Scd1) was drastically increased in response to ADRB3 stimulation in eWAT, and readily resolved as a separate cluster (Figs. 2A, ,2C)2C) (64). A population of mesothelial-like cells present only in eWAT was also distinguished by scRNAseq (Fig. 2A) (64, 104). The mesothelium is a thin membrane containing a layer of epithelial-like cells that surrounds some internal organs, including visceral adipose depots, and has been proposed to be a source of adipocyte progenitors during development (106109). This cluster shares some similarity with marker genes for ASC2 (Dpp4, Ackr3, Smpd3), expresses low levels of Pdgfra/Pdgfrb, and highly express genes characteristic of mesothelial/epithelial cells (Msln, Krt8, and Krt14) (104). Wt1, a developmental marker (109), is also strongly expressed in these cells, as well as the other ASC clusters in eWAT, but not in iWAT (Fig. 2B). Other subpopulations present to varying degrees in each depot were identified based on marker gene expression, and included endothelial cells (Plvap, Flt1, Tie1), smooth muscle cells (SMC; Tagln, Acta2, Myh11), and subpopulations of contaminating immune cells tentatively identified as macrophages, NKT cells, B lymphocytes, and dendritic cells (DC) that were somewhat variable across studies (Figs. 2A, ,3A3A).

A number of different genetic tracers have been used to determine progenitor fate both during development and in adult tissue (46, 49, 9092, 94, 109112). During development, adipocytes that arise in eWAT have a history of Wt1 and Pax3 (in males) expression, whereas subcutaneous depots are thought to be derived from cells that once expressed Prx1 (91, 109, 111, 112). Among these, only Wt1 was detectable in ASC from adults and expression was restricted to eWAT (Fig. 2B). Dlk1 (Pref-1) also marks mesenchymal ASC progenitors (110) and was largely absent in ASC from adults, but present earlier in development and more strongly in ASC1a (unpublished observations). In addition to Pdgfra, which was broadly expressed in both ASC populations, other markers that have been used to trace the adipocyte lineage in adult tissue including Pdgfrb and Pparg are more strongly expressed in ASC1a. However, these genes are also co-expressed in endothelial/SMC clusters that are positive for Cdh5 and Acta2, but generally negative for Pdgfra (Figs. 2B, ,3B)3B) (64).

In Vivo and In Vitro Functional Characteristics of eWAT Progenitors

Many of the known transcriptional and morphological changes that occur during adipogenesis have been derived from in vitro studies, either using preadipocyte cell lines or progenitors isolated from adipose tissue (reviewed in (113116)). One of the benefits of scRNAseq, aside from detection of major cell populations, is the ability to detect changes in cellular states that occur in vivo (67). The ADRB3 agonist CL-316,243 (CL) is a potent inducer of brown adipogenesis in white adipose tissue; however, the mechanisms through which CL induces brown adipocyte formation is depot specific (46, 92, 117). In a series of studies, Lee et al. (45, 46, 92, 117) demonstrated that de novo brown adipogenesis in epididymal WAT (eWAT) is dependent on recruitment of alternatively activated (M2) macrophages that proliferate to form “crown like structures” (CLS) involved in the clearance of dead adipocytes (117, 118). The local release of lipid metabolites (e.g., 9-hydroxyoctadecadienoic acid) and other chemoattractants (e.g., OPN) by macrophages that comprise the CLS then potentiates the proliferation, migration, and differentiation of Pdgfrα/CD44+ progenitors into brown adipocytes (45, 46, 92, 117). In iWAT, CLS are rarely induced by CL treatment (92), and most brown adipocytes arise from existing white adipocytes that transform into a brown/beige phenotype (46, 54, 64, 92).

Burl et al. (64) profiled non-immune stromal cells in eWAT with scRNAseq 3 days following CL treatment to further deconvolve the recruited Pdgfrα+ progenitors and identify temporal changes in ASC that occur during brown adipogenesis in vivo. Two major clusters of quiescent ASC (ASC1, ASC2) were identified from both depots (Figs. 2A, ,3A)3A) (64). An additional cluster of proliferating and differentiating preadipocytes was further resolved in the CL-treated group, comprising ~10% of the total ASC population in eWAT (Fig. 2A) (64). Pseudotime analysis mapped a differentiation trajectory of cells undergoing brown adipogenesis into distinct cellular states, spanning proliferating to late differentiating preadipocytes (64). In general, differentiating preadipocytes lost expression of proliferation markers (Birc5, Ccnb2) and progressively gained expression in transcriptional regulators of adipogenesis as well as metabolic markers of maturing adipocytes (e.g., Dgat1/2, Pnpla2, Lipe, Acot5, Agpat2) (64). Several ECM remodeling proteins were also downregulated (Mmp23) whereas Ucp1, a marker of brown adipocytes, was upregulated in late differentiating preadipocytes (64). Moreover, scRNAseq provided better resolution of these “cellular state” changes compared to bulk RNA sequencing of FACS-isolated Pdgfrα cells (64). A number of marker genes defining ASC1a (Agt, Col15a1) were also co-expressed within the proliferating and differentiating cluster, indicating that ASC1a (Pdgfra+) cells are a more committed adipocyte progenitor (64). Similar to previous findings (46, 92), progenitors in iWAT were largely refractory to the effects of ADRB3 agonism (Fig. 3A), with few transcriptional changes detected amongst the ASC clusters (64).

Perivascular cells expressing the surface marker Pdgfrβ are enriched in the adipogenic transcription factor Pparγ and have been suggested to be a source of progenitors in white adipocyte tissue (38, 49, 94). Disrupting Pparg within this subtype impairs, while overexpression promotes, hyperplastic VAT expansion and improves insulin sensitivity during diet-induced obesity (38). Hepler et al. (104) performed scRNAseq on FACS-sorted cells isolated from eWAT of “Muralchaser” mice to further characterize the Pdgfrβ progenitor populations (Table 1). In the Muralchaser mouse model, treatment with doxycycline constitutively activates a membrane green fluorescent protein (mGFP) reporter in a Pdgfrb- and Cre-dependent manner, allowing purification of cells with a history of Pdgfrb expression at the time of pulsing (described in (49, 104)). Using this model, mGFP+ cells were isolated from the total stromal population and used as input for scRNAseq (104). Despite a more selective cell enrichment strategy, distinct clusters of adipocyte progenitor cells (ASC1a, FIPs/ASC2, preadipocytes) and a small population of mesothelial cells (Fig. 2A) were resolved that exhibited an expression profile similar to those found by Burl et al. (64) (Figs. 2B, ,C;C; Supplemental Fig. 1). Hepler et al. (104) further devised a cell sorting strategy to purify and functionally evaluate these populations (Table 1, Fig. 2B). In cell culture, ASC1a cells (Pdgfrβ+CD9-Ly6c-) were found to be more adipogenic than FIPs/ASC2 (Pdgfrβ+Ly6c+) or mesothelial cells (Pdgfrβ+CD9+Ly6c-) (104). Further, ASC1a, but not FIP/ASC2 cells differentiated into adipocytes when transplanted into the subcutaneous fat pad of lypodystrophic mice, providing further evidence that ASC1a is a direct adipogenic precursor (64, 104). In comparison, cell sorted FIPs/ASC2 inhibited the differentiation of ASC1a and displayed a higher pro-inflammatory/pro-fibrotic phenotype in vitro (104). Interestingly, FIPs/ASC2 were also more proliferative than ASC1a in response to a high fat diet in vivo (104). These findings are consistent with those of Marcelin et al. (119) who described a Pdgfrα+CD9high population in eWAT with a similar profile to ASC2 (Fig. 2B) that were also pro-fibrotic and more resistant to adipogenesis in vitro.

In Vivo and In Vitro Functional Characteristics of iWAT Progenitors

Adipose progenitors in the subcutaneous iWAT are generally less sensitive to de novo adipogenesis following ADRB3 stimulation or high fat feeding than progenitors in eWAT (2426, 46, 64, 92). Two additional studies conducted scRNAseq on non-immune stromal cells isolated from iWAT under basal conditions (103) and during the period of perinatal tissue expansion (102) (Table 1). Schwalie et al. (103) performed scRNAseq on stromal cells from adult mice and (in addition to ASC1 and ASC2) identified a subset of ASC1 cells they named “Aregs” (adipogenesis-regulatory cells; ASC1b). This subpopulation was distinguished by the expression of several gene markers including Fmo2, F3 (CD142), and Gdf10 (Fig. 3), and co-localized to perivascular regions within the parenchyma body (103). In cell culture, FACS-sorted ASC1b cells (Lin-Sca1+CD142+Abcg1+) were more refractory to adipogenesis when compared to total ASC (Lin-Sca1+) or total ASC depleted of ASC1b (Lin-Sca1+CD142-Abcg1-) (103). Further, FACS-isolated ASC1b cells inhibited the differentiation capacity of ASC (Lin-Sca1+CD142-Abcg1-) in vitro, potentially through a paracrine effect (103). FACS-isolated ASC1b cells were also detected in higher proportions within the subcutaneous and visceral depots of ob/ob mice, a genetically obese model, although a similar cluster based on scRNAseq was not readily identifiable in eWAT of mice (104) or in subcutaneous abdominal fat from humans (102). In Matrigel transplantation experiments, depletion of ASC1b from the total ASC population also led to a greater number of mature adipocytes following high fat feeding, suggesting that this subtype maintains regulatory function in vivo (103).

Merrick et al. (102) evaluated immune-depleted stromal cells from iWAT during a period of perinatal adipose tissue expansion (postnatal day (PND) 12) (Table 1). Similar to findings by Schwalie et al. (103), they identified an ASC2 (DPP4+CD142-) and two ASC1 populations (Icam+CD142-(ASC1a) and CD142+ (ASC1b); Table 1), with a proportionally higher number of total ASC1 cells during the period of perinatal adipose tissue growth (102). However, in contrast to the reported inhibitory role of ASC1b, all 3 cell sorted ASC subpopulations were adipogenic both in vitro and in vivo (102). Additional functional assays indicate that ASC2 are more resistant to in vitro adipogenesis when insulin concentrations are reduced, are capable of multilineage differentiation, and are more sensitive to the anti-adipogenic effects of transforming growth factor beta (TGFβ) than ASC1a or ASC1b (102). In Matrigel transplantation studies of isolated populations in vivo, ASC2 cells were found to transition into either ASC1a or ASC1b prior to becoming adipocytes, suggesting ASC2 are a less committed progenitor (102). It is noteworthy that although all ASC progenitors from iWAT were adipogenic in this study, ASC2 cells isolated from eWAT and transplanted into iWAT of lipodystrophic mice failed to develop into adipocytes and were more resistant to in vitro adipogenesis (104, 119). Therefore, despite similarities in gene expression profiles (Figs. 2, ,3),3), at least some characteristics of ASC2 appear to be depot specific.

Immunohistochemical Localization of ASC Subpopulations in iWAT and eWAT

Cluster analysis indicates the major cell populations in eWAT or iWAT exhibit similar marker gene profiles, as well as other functional characteristics. For example, a pro-adipogenic phenotype was observed for ASC1a from both WAT depots (102104), and an inflammatory, fibrotic, or mesenchymal phenotype for ASC2 (102, 104, 119). Merrick et al. (102) evaluated the distribution of ASC progenitors during perinatal adipose tissue development and found ASC subtypes to be compartmentally localized in iWAT. ASC2 (Pref-1- DPP4+) cells were mainly confined to the collagen-rich fascia (“reticular interstitium”), whereas ASC1a progenitors (Pref-1+DPP4-) were located within the parenchyma body, closely associated with adipocytes (102). To determine whether these subtypes share a similar distribution in eWAT, we evaluated total ASC (Pdgfrα) and ASC2 (DPP4, Pi16) markers in both depots from adult mice by immunohistochemistry (Fig. 4). Similar to findings from Merrick et al. (102), ASC2 (Pdgfrα+DPP4+/Pi16+) cells were restricted mainly to the fascia (extracellular connective tissue) in iWAT, which encapsulates the adipose depot and is several cell layers thick (Fig. 4B). Double positive cells also could be found in a thin connective layer lining individual fat lobules (Fig. 4B). By comparison, ASC1 cells (Pdgfrα+DPP4-) were found throughout the parenchyma body and in areas surrounding vessels, as previously observed (46).

An external file that holds a picture, illustration, etc.
Object name is nihms-1665107-f0004.jpg
Immunohistochemical Staining of Total ASC (Pdgfrα) and ASC2 (DPP4, Pi16) in eWAT and iWAT of Adult Male Mice.

(A, B) Immunofluorescence for Pdgfrα (green) and either DPP4 (red, top panel) or Pi16 (red, bottom panel) was evaluated in formaldehyde-fixed tissue sections (10 μM) from (A) eWAT and (B) iWAT. (A) ASC2 cells (Pdgfrα+DPP4+ or PI16+) cells are localized in areas underlying the mesothelium in eWAT. Note the strong immunoreactivity of DPP4 surrounding eWAT, which is also expressed in mesothelial cells from scRNAseq (Fig. 2C). (B) ASC2 progenitors are localized primarily within the fascia that encapsulates the inguinal adipose depot and is also present in a thin layer of connective tissue lining individual fat lobules. ASC1 cells (Pdgfrα+DPP4-) are localized within the parenchyma body in association with mature adipocytes and surrounding vessels. DAPI was used for nuclear staining as indicated in blue. Orange arrows denote some areas immunoreactive for both Pdgfrα and either DPP4 or Pi16. Abbr: Mesothelium (Mesoth.).

In eWAT, a layer of cells that stained strongly for DPP4 surrounded the entire adipose depot (Fig. 4A). This distribution is consistent with the visceral mesothelium previously described (Fig. 4A) (106, 109). Some double-labeled cells (ASC2; Pdgfrα+DPP4+) were observed underlying the mesothelial layer but were mostly absent within fat lobules (Fig. 4A). To better distinguish cells, we additionally evaluated distribution of Pi16, a marker more specific for this ASC cell type in eWAT (Fig. 2C). Doubly labeled cells (ASC2; Pdgfrα+Pi16+) were localized to areas directly underlying the mesothelium but were generally fewer in number compared to ASC1a cells (Pdgfrα+Pi16-) (Fig. 4A). Pi16+ cells were also found in areas surrounding large vessels in eWAT (not shown). In comparison, ASC1a cells were located near the mesothelium and dispersed throughout the fat lobules. Mesothelial cells are thought to serve as a protective barrier, with additional roles including immunomodulatory functions as well as tissue repair (107, 108, 120123). It is interesting that ASC2 cells were closely associated with this cell type in eWAT and may be related to the higher pro-inflammatory and fibrotic phenotypes observed in vitro and in vivo (104, 119). Whether ASC2 can transition into ASC1a as suggested for iWAT (102) or if there are distinct progenitor lineages for each population is currently under investigation.

ADIPOSE STEM CELL POPULATIONS IN DERMAL WAT (dWAT) AND THE LACTATING MAMMARY GLAND AS IDENTIFIED BY scRNASEQ

In addition to ASC identified within the major WAT depots, Scherer and colleagues (124) recently used scRNAseq to address plasticity of dermal white adipose tissue (dWAT) during hair follicle cycling and in the mammary gland during pregnancy and lactation (125). The dermal adipocyte layer expands by both hypertrophy and hyperplasia during active periods of hair follicle growth (anagen) and then diminishes in thickness during the regression of follicles and rest (catagen/telogen) (124, 126). Using the “AdipoChaser-mT/mG” model, whereby mGFP is expressed on the membrane of cells with a history of adiponectin expression at the time of pulsing (second anagen), dermal adipocytes (GFP+) were found to dedifferentiate into Pdgfrα/GFP+ progenitor cells following hair follicle regression/rest (second telogen) (124). FACS-isolated Pdgfrα (CD45-/CD31-) progenitors were then profiled with scRNAseq during the second telogen to evaluate transcriptomic differences between dedifferentiated (GFP+) and unlabeled (GFP-) progenitors (124). Using this model, two main populations of ASC were identified that share some similarity in marker gene profiles with those observed in the other major WAT depots (ASC2, Ptgs2; ASC1a, Apoe) (64, 102104, 124). Although few transcriptional changes were observed between dedifferentiated (GFP+) and GFP- ASC, dedifferentiated cells were slightly more adipogenic in vitro (124). Similar to the above, the fate of adipocytes during lactation and recurrence at involution were evaluated in the “Adipochaser-mT/mG” model (125). Adipocytes (GFP+) labeled prior to pregnancy were found to dedifferentiate into Pdgfrα+/GFP+ SVC during lactation and re-differentiate into adipocytes upon weaning of pups (125). Using scRNAseq, the Pdgfrα+/GFP+ (CD31-/CD45-) progenitors were found to have a similar expression profile to Pdgfrα progenitors isolated from virgin females (125). However, due to limitations in sample size, it is unknown if the dedifferentiated cells represent more than one ASC subtype (125). Nonetheless, these findings highlight the cellular plasticity of adipose tissue and further suggest that adipocytes can fully dedifferentiate into ASC under certain physiological conditions (124, 125).

IMMUNE CELL POPULATIONS IN eWAT DETECTED WITH scRNASEQ

Visceral adipose tissue is known to have differential susceptibility to inflammation and neogenesis compared to subcutaneous fat (2426, 37, 46, 92, 127). Infiltration and local proliferation of adipose tissue macrophages (ATMs) is frequently observed in obesity and contributes to insulin resistance and other metabolic derangements through local secretion of pro-inflammatory mediators (16, 37, 128131). In obese states, a large proportion of ATMs in the visceral depot surround dead adipocytes, forming “crown-like structures” (CLS) (37, 118, 127), that are thought to be an important source of these mediators (37). The recruitment and proliferation of macrophages is also tightly linked with de novo adipogenesis and is important for initiating WAT remodeling in response to energy excess, β-adrenergic stimulation, and following tissue injury (45, 92, 117). Recruited macrophages also exhibit heterogeneity in phenotypes depending on the type and duration of the stimulus (37, 45, 92, 130), although the distinct populations involved and activation states have not been thoroughly characterized. Several studies have used scRNAseq to characterize immune cell populations during WAT remodeling, either following ADRB3 stimulation or during diet-induced obesity (64, 132134). However, due to the extent of data evaluated, results are only briefly summarized below.

Immune Cell Populations Associated with ADRB3-Induced Remodeling

ADRB3-induced remodeling in eWAT is associated with the rapid accumulation of adipose tissue macrophages that are involved in phagocytosis and lipid clearance (45, 46, 92, 117). Compared to other types of WAT remodeling (92), ADRB3-induced remodeling involves an alternatively activated (M2-polarized, non-inflammatory) macrophage response that is dependent on the chemokine osteopontin (OPN; Spp1) (45, 92, 117, 135). Burl et al. (64) used scRNAseq to profile immune cell populations following CL treatment. Adipose tissue macrophages (ATM) were found to be the most prominent immune cell type present (~58%), and the relative proportion of macrophages expanded by ~2.5 fold following CL treatment (64). Clustering of ATM populations based on transcriptional profiles identified at least 6 different subgroups (64). Among these, CL modestly expanded a subset that was enriched for proliferation markers and whose gene expression profile was associated with processes including lipid uptake and metabolism and ECM remodeling (64). Osteopontin (OPN, Spp1), a chemoattractant that is highly upregulated in CLS macrophages (92, 117), was also enriched within this subpopulation (64). OPN is a chemoattractant for ASC1a progenitors and would likely explain their recruitment to CLS and formation of an “adipogenic niche” previously described during catabolic tissue remodeling (45, 46, 64, 92, 117).

Immune Cell Populations Associated with HFD-Induced Remodeling

Jaitin et al. (133) performed scRNAseq on isolated lineage positive (CD45+) cells to identify immune cell populations driving WAT remodeling during diet-induced obesity. Cells were isolated from eWAT at 6, 12, and 18 weeks following HFD and cell populations (“metacells”) were classified into 15 different immune cell types (133). Between 6 and 12 weeks following high fat feeding, there was a dramatic shift in immune cell populations, including a substantial increase in the proportion of macrophages and concomitant decrease in monocytes (133). Aside from resident/perivascular macrophages, they identified two populations of macrophages (“Mac2” and “Mac3”), characterized by differential expression of Cd9 and Nceh1, that were prominent only in obese animals, derived from recruited monocytes, and by histochemical analyses, accumulated in CLS (133). Consistent with localization to the CLS, the Mac3 population was enriched for genes involved in lipid metabolism and phagocytosis, and the receptor Trem2 was found to be exclusive for this immune cell population (133). In loss of function studies, Trem2-deficiecy prevented the appearance of the Mac3 subset during obesity, without affecting monocyte/macrophage infiltration (133). Further histochemical and metabolic analyses revealed an important role for Trem2 signaling in preventing adipose tissue dysfunction by promoting the formation of CLS to initiate remodeling and limit/regulate adipocyte size (133).

Two other studies evaluated immune cell heterogeneity in eWAT (132, 134). Hill et al. (132) profiled CD11b+Ly6c- ATMs and identified 2 distinct populations with scRNAseq. One subset was enriched for Cd9 and found to be the main population of Ly6c- ATMs that accumulate during diet-induced obesity (132). Similar to findings from Jaitin et al. (133), both Cd9+Ly6c- and Cd9-Ly6c+ cells were recruited from bone marrow-derived monocytes, and Cd9+Ly6c- cells localized to CLS in obese states (132). The transcriptional profile and chromatin landscape of Cd9+Ly6c- macrophages further suggested a proinflammatory role compared to that of the Cd9-Ly6c+ subset, which were implicated more in tissue maintenance/homeostasis (132). Interestingly, the subset of CLS macrophages identified by Burl et al. (64) was also enriched for the genes Trem2 and Cd9. Therefore, this transcriptional “signature” appears to be a conserved pathway for CLS-associated macrophages during WAT remodeling.

Weinstock et al. (134) profiled immune cell changes that occur in eWAT during high fat feeding and following 2 weeks of caloric restriction (CR; 70% kcal) and compared these profiles to that of lean mice (64). They identified 15 distinct immune cell populations in eWAT with scRNAseq (134). Seven of the clusters were categorized as macrophages and collectively represented more than one-half of the total immune cell types present (134). HFD was found to expand the “major macrophage” population by ~50% and also enriched for a subset of “phagocytic macrophages” (7%) that were not present in lean control mice (64, 134). Following a two week period of CR, this phagocytic population further increased to 30% of the total immune population, whereas the major macrophage population decreased to proportions observed in lean mice (134). The increase in phagocytic macrophages following CR was suggested to enhance clearance of lipid and adipocyte remnants to restore ATM homeostasis (134).

SUMMARY OF scRNASEQ STUDIES IN WAT

Collectively, studies exploring adipose tissue composition using scRNAseq have provided relatively consistent data regarding WAT progenitor populations, expression profiles, and insight into immune cell phenotypes during catabolic and anabolic tissue remodeling. The adipocyte progenitors identified from subcutaneous and visceral depots express several of the previously identified genes for stem cells including Pdgfra, Itgb1, Cd34, and Ly6a; however, 2–3 distinct subpopulations could be further resolved by scRNAseq. Based on both in vitro characterization (102104) and in vivo trajectories (64), ASC1a appears to represent an immediate adipocyte progenitor. Further, immunochemical evidence indicates this cell type is localized in the parenchyma body in association with adipocytes (102).

Some inconsistencies exist regarding ASC1b in iWAT, as both pro-adipogenic and anti-adipogenic roles have been reported (102, 103). The reason for this discrepancy is currently unknown but may be due to the different selection markers used to purify cell populations, as suggested (102). ASC2 appears to be a relatively new defined progenitor subset, and available data suggest a pro-inflammatory role (104, 119), at least in eWAT. This subpopulation is detected by immunohistochemistry in the collagen-rich fascia encapsulating iWAT (102) and in close contact with mesothelial cells in eWAT. ASC2 are further characterized by a higher expression of Wnt genes generally associated with a less committed, multi-lineage mesenchymal stem cell, and have been hypothesized to be a direct precursor for both ASC1a and b in iWAT (102). Lineage tracing using cell-selective markers identified through these studies will help further delineate a potential hierarchal role for this subtype in vivo (102).

In visceral depots, the most prominent immune cell population is monocyte/macrophages. In separate models of tissue remodeling, scRNAseq was used to detect population and phenotype changes in response to either HFD or ARDB3 stimuli (64, 132134). A shift in macrophage populations as well as gene expression patterns were resolved using scRNAseq. Further, a subset of macrophages associated with “crown-like structures” were distinguishable based on gene profiling and further confirmed with histochemical analyses (64, 132, 133). Cd9, Spp1, and Trem2 were identified as genes enriched within this subset. Additionally, Trem2 signaling appears to drive the transcriptional and functional responses of this subgroup in obesity, which is suggested to promote formation of CLS to control adipocyte size (133). These findings highlight an important role for CLS-associated macrophages in initiating adipose tissue remodeling and provide new insights into the complexity of immune activation states in adipose tissue.

Although the current review aimed to summarize findings available from preclinical mouse models, a few comparable studies addressing heterogeneity of stromal cells with scRNAseq in humans have also been reported (102, 132, 133). For example, Merrick et al. (102) found similar populations of the two major ASC (ASC1a, ASC2), in human abdominal subcutaneous adipose tissue as those identified in mice, with a proportionally higher number of ASC1a cells. Conversely, ASC1b markers were more diffusely expressed (102). FACS-sorted ASC populations also exhibited similar functional properties in adipogenic assays, with ASC1a being more poised for adipogenesis than the ASC2 subtype (102). The presence of an analogous, lipid-laden CD9/TREM2+ macrophage population associated with CLS in obese human visceral fat was also identified using scRNAseq (133) and by histological and FACS-based analyses (132). Importantly, a positive association between the accumulation of CD9+ macrophages in VAT and obesity was reported by both Jaiten et al. (133) and Hill et al. (132). These studies suggest at least some conserved cell types and cellular responses between species. Nonetheless, further integration and interrogation of single cell datasets will lead to a better understanding of species-dependent differences among adipose tissue depots and potentially aid in more directed treatments for obesity-related metabolic dysfunction.

FUTURE DIRECTIONS

It is evident that scRNAseq differentiates stromal cell populations and cellular states with high resolution, providing a powerful tool for addressing adipose tissue heterogeneity. One limitation from the available studies is that all have enriched for lineage selection, therefore the relative composition of stromal cells within the different depots and proportional changes in cell types under different conditions are not well defined. Cellular input and capture efficiencies differ to some degree among different scRNAseq platforms (65, 78). As scRNAseq becomes increasingly streamlined, the capacity to detect wider range of cells is feasible and should be considered in future studies.

Although the focus of this review was on stromal cell heterogeneity, adipocytes arise from distinct developmental lineages and depot specific differences in adipokine release, lipolysis, and inflammation are well established (reviewed in (60, 136, 137)). Heterogeneity among adipocytes within the same depot have also been reported by both genetic and histological criteria (138, 139). For example, adjacent brown adipocytes within the interscapular brown adipose tissue (iBAT) of mice often express substantially different levels of uncoupling protein 1 (UCP1), yielding a ‘harlequin’ type appearance upon histological staining (138). Due to their fragility and natural buoyancy in suspension, and for brown adipocytes, high mitochondrial content, examining isolated adipocytes by scRNAseq is technically challenging (64). Methods for single nucleus RNA-seq (snRNAseq) are available (140, 141) and results from other tissues suggest good correlation to profiles generated with scRNAseq that are less affected by cell isolation methods (142, 143). Two recent studies have begun to address adipocyte heterogeneity, either in isolated brown adipocytes or from inguinal white adipocyte nuclei using sc/snRNAseq, respectively (139, 144). These studies reported some evidence of heterogeneity within adipocyte gene-expressing clusters (139, 144). Nonetheless, both studies also identified additional clusters of putative adipocytes that co-express markers prominent in other cell types, including endothelial cells (e.g., Flt1, Pecam1) (139, 144) as well as ASC subtypes (e.g., Pi16, Pdgfra, Gdf10) (144). Indeed, the profile of putative brown adipocytes expressing low levels of UCP1 reported by Song et al. (139) is more similar to capillary endothelial cells than brown adipocytes when integrated with multiple independent scRNAseq data sets. Whether these cells represent non-classical adipocytes or contaminating cells present in the floating adipocyte fraction is worthy of future validation.

While scRNAseq provides deep information regarding cell types and states, it does not provide information on localization of cellular populations within adipose tissue. Nonetheless, scRNAseq data provide additive and informative markers that can be used to localize cells within the tissue microenvironment and confirm potential cell-cell interactions. In this way, ASC2 has been localized to the fascia or “reticular interstitium” (102) in iWAT, and Spp1/Trem2 macrophages and activated ASC progenitors to the CLS of visceral depots (46, 64, 92, 117, 132, 133). We anticipate that additional exploration of markers genes identified through current scRNAseq studies as well as future developments in spatial transcriptomics (145) will provide a more complete picture of adipose tissue heterogeneity, cellular interactions, and help dissect mechanisms of tissue plasticity.

Supplementary Material

Acknowledgements

The authors thank members of CIMER for useful suggestions and Dr. Roger Pique-Regi for guidance in scRNAseq data analysis.

Funding

This work was supported by the National Institute of Diabetes and Digestive and Kidney Disease (NIDDK) [JGG; R01DK062292] and the National Institutes of Health National Institute of Environmental Health Sciences [Center Grant P30 ES020957].

Abbreviations:

(Aregs)Adipogenesis-regulatory cells
(ASC)adipose stem cell
(ATMs)adipose tissue macrophages
(ADRB3)β3 adrenergic receptor
(BAT)brown adipose tissue
(CCA)canonical correlation analysis
(CL)CL316,243
(cDNA)complementary DNA
(CLS)crown-like structures
(eWAT)epidydimal white adipose tissue
(ECM)extracellular matrix
(DC)dendritic cells
(dWAT)dermal white adipose tissue
(DPP4)dipeptidyl peptidase 4
(ECM)extracellular matrix
(FIP)fibro-inflammatory progenitor
(FACS)fluorescence-activated cell sorting
(HFD)high fat diet
(iBAT)interscapular brown adipose tissue
(iWAT)inguinal white adipose tissue
(mGFP)membrane green fluorescent protein
(NKT)natural killer T cells
(OPN)osteopontin
(Prx1)paired related homeobox transcription factor 1
(Pi16)peptidase inhibitor 16
(PPARγ)peroxisome proliferator-activated receptor gamma
(Pdgfrα)platelet-derived growth factor alpha
(Pdgfrβ)platelet-derived growth factor beta
(PCR)polymerase chain reaction
(PND)postnatal day
(Pref-1)preadipocyte factor-1
(RT-PCR)reverse transcription-polymerase chain reaction
(scRNAseq)single cell RNA-sequencing
(snRNAseq)single nucleus RNA-sequencing
(Sca-1)stem cell antigen-1
(t-SNE)t-distributed stochastic neighbor embedding
(UCP1)uncoupling protein 1
(VAT)visceral adipose tissue
(WAT)white adipose tissue
(UMI)unique molecule identifier
(Wt1)Wilms tumor 1

Footnotes

Competing Interests

The authors declare that there are no competing interests.

REFERENCES

1. Collaborators GBDO, Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, et al. Health Effects of Overweight and Obesity in 195 Countries over 25 Years. N Engl J Med. 2017;377(1):13–27. [PMC free article] [PubMed] [Google Scholar]
2. Collaboration NCDRF. Worldwide trends in diabetes since 1980: a pooled analysis of 751 population-based studies with 4.4 million participants. Lancet. 2016;387(10027):1513–30. [PMC free article] [PubMed] [Google Scholar]
3. Sung H, Siegel RL, Rosenberg PS, Jemal A. Emerging cancer trends among young adults in the USA: analysis of a population-based cancer registry. Lancet Public Health. 2019;4(3):e137–e47. [PubMed] [Google Scholar]
4. Steele CB, Thomas CC, Henley SJ, Massetti GM, Galuska DA, Agurs-Collins T, et al. Vital Signs: Trends in Incidence of Cancers Associated with Overweight and Obesity - United States, 2005–2014. MMWR Morb Mortal Wkly Rep. 2017;66(39):1052–8. [PMC free article] [PubMed] [Google Scholar]
5. Stolarczyk E Adipose tissue inflammation in obesity: a metabolic or immune response? Curr Opin Pharmacol. 2017;37:35–40. [PubMed] [Google Scholar]
6. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–7. [PubMed] [Google Scholar]
7. Ravussin E, Smith SR. Increased fat intake, impaired fat oxidation, and failure of fat cell proliferation result in ectopic fat storage, insulin resistance, and type 2 diabetes mellitus. Ann N Y Acad Sci. 2002;967:363–78. [PubMed] [Google Scholar]
8. Kalupahana NS, Moustaid-Moussa N, Claycombe KJ. Immunity as a link between obesity and insulin resistance. Mol Aspects Med. 2012;33(1):26–34. [PubMed] [Google Scholar]
9. Russo L, Lumeng CN. Properties and functions of adipose tissue macrophages in obesity. Immunology. 2018;155(4):407–17. [PMC free article] [PubMed] [Google Scholar]
10. Franckhauser S, Munoz S, Pujol A, Casellas A, Riu E, Otaegui P, et al. Increased fatty acid re-esterification by PEPCK overexpression in adipose tissue leads to obesity without insulin resistance. Diabetes. 2002;51(3):624–30. [PubMed] [Google Scholar]
11. Zeve D, Tang W, Graff J. Fighting fat with fat: the expanding field of adipose stem cells. Cell Stem Cell. 2009;5(5):472–81. [PMC free article] [PubMed] [Google Scholar]
12. Khan T, Muise ES, Iyengar P, Wang ZV, Chandalia M, Abate N, et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol Cell Biol. 2009;29(6):1575–91. [PMC free article] [PubMed] [Google Scholar]
13. Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116(6):1494–505. [PMC free article] [PubMed] [Google Scholar]
14. Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest. 2006;116(1):115–24. [PMC free article] [PubMed] [Google Scholar]
15. Tran TT, Yamamoto Y, Gesta S, Kahn CR. Beneficial effects of subcutaneous fat transplantation on metabolism. Cell Metab. 2008;7(5):410–20. [PMC free article] [PubMed] [Google Scholar]
16. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112(12):1821–30. [PMC free article] [PubMed] [Google Scholar]
17. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, et al. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007;447(7148):1116–20. [PMC free article] [PubMed] [Google Scholar]
18. Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453(7196):783–7. [PubMed] [Google Scholar]
19. Rigamonti A, Brennand K, Lau F, Cowan CA. Rapid cellular turnover in adipose tissue. PLoS One. 2011;6(3):e17637. [PMC free article] [PubMed] [Google Scholar]
20. Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin Invest. 2011;121(6):2094–101. [PMC free article] [PubMed] [Google Scholar]
21. Choe SS, Huh JY, Hwang IJ, Kim JI, Kim JB. Adipose Tissue Remodeling: Its Role in Energy Metabolism and Metabolic Disorders. Front Endocrinol (Lausanne). 2016;7:30. [PMC free article] [PubMed] [Google Scholar]
22. Lee YH, Mottillo EP, Granneman JG. Adipose tissue plasticity from WAT to BAT and in between. Biochim Biophys Acta. 2014;1842(3):358–69. [PMC free article] [PubMed] [Google Scholar]
23. Hepler C, Gupta RK. The expanding problem of adipose depot remodeling and postnatal adipocyte progenitor recruitment. Mol Cell Endocrinol. 2017;445:95–108. [PMC free article] [PubMed] [Google Scholar]
24. van Beek L, van Klinken JB, Pronk AC, van Dam AD, Dirven E, Rensen PC, et al. The limited storage capacity of gonadal adipose tissue directs the development of metabolic disorders in male C57Bl/6J mice. Diabetologia. 2015;58(7):1601–9. [PMC free article] [PubMed] [Google Scholar]
25. Jeffery E, Church CD, Holtrup B, Colman L, Rodeheffer MS. Rapid depot-specific activation of adipocyte precursor cells at the onset of obesity. Nat Cell Biol. 2015;17(4):376–85. [PMC free article] [PubMed] [Google Scholar]
26. Wang QA, Tao C, Gupta RK, Scherer PE. Tracking adipogenesis during white adipose tissue development, expansion and regeneration. Nat Med. 2013;19(10):1338–44. [PMC free article] [PubMed] [Google Scholar]
27. Fox CS, Massaro JM, Hoffmann U, Pou KM, Maurovich-Horvat P, Liu CY, et al. Abdominal visceral and subcutaneous adipose tissue compartments: association with metabolic risk factors in the Framingham Heart Study. Circulation. 2007;116(1):39–48. [PubMed] [Google Scholar]
28. Carey DG, Jenkins AB, Campbell LV, Freund J, Chisholm DJ. Abdominal fat and insulin resistance in normal and overweight women: Direct measurements reveal a strong relationship in subjects at both low and high risk of NIDDM. Diabetes. 1996;45(5):633–8. [PubMed] [Google Scholar]
29. Koster A, Stenholm S, Alley DE, Kim LJ, Simonsick EM, Kanaya AM, et al. Body fat distribution and inflammation among obese older adults with and without metabolic syndrome. Obesity (Silver Spring). 2010;18(12):2354–61. [PMC free article] [PubMed] [Google Scholar]
30. Zhang M, Hu T, Zhang S, Zhou L. Associations of Different Adipose Tissue Depots with Insulin Resistance: A Systematic Review and Meta-analysis of Observational Studies. Sci Rep. 2015;5:18495. [PMC free article] [PubMed] [Google Scholar]
31. McLaughlin T, Lamendola C, Liu A, Abbasi F. Preferential fat deposition in subcutaneous versus visceral depots is associated with insulin sensitivity. J Clin Endocrinol Metab. 2011;96(11):E1756–60. [PMC free article] [PubMed] [Google Scholar]
32. Lessard J, Laforest S, Pelletier M, Leboeuf M, Blackburn L, Tchernof A. Low abdominal subcutaneous preadipocyte adipogenesis is associated with visceral obesity, visceral adipocyte hypertrophy, and a dysmetabolic state. Adipocyte. 2014;3(3):197–205. [PMC free article] [PubMed] [Google Scholar]
33. Arner E, Westermark PO, Spalding KL, Britton T, Ryden M, Frisen J, et al. Adipocyte turnover: relevance to human adipose tissue morphology. Diabetes. 2010;59(1):105–9. [PMC free article] [PubMed] [Google Scholar]
34. Bjorntorp P Sjostrom L,+SJOSTROM L: Number and size of adipose tissue fat cells in relation to metabolism in human obesity. Metabolism. 1971;20(7):703–13. [PubMed] [Google Scholar]
35. Bjorntorp P, Bengtsson C, Blohme G, Jonsson A, Sjostrom L, Tibblin E, et al. Adipose tissue fat cell size and number in relation to metabolism in randomly selected middle-aged men and women. Metabolism. 1971;20(10):927–35. [PubMed] [Google Scholar]
36. Ktotkiewski M, Sjostrom L, Bjorntorp P, Smith U. Regional adipose tissue cellularity in relation to metabolism in young and middle-aged women. Metabolism. 1975;24(6):703–10. [PubMed] [Google Scholar]
37. Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW 2nd, DeFuria J, Jick Z, et al. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes. 2007;56(12):2910–8. [PubMed] [Google Scholar]
38. Shao M, Vishvanath L, Busbuso NC, Hepler C, Shan B, Sharma AX, et al. De novo adipocyte differentiation from Pdgfrbeta(+) preadipocytes protects against pathologic visceral adipose expansion in obesity. Nat Commun. 2018;9(1):890. [PMC free article] [PubMed] [Google Scholar]
39. Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell. 2012;148(5):852–71. [PMC free article] [PubMed] [Google Scholar]
40. Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, et al. Adipose Tissue Dysfunction as Determinant of Obesity-Associated Metabolic Complications. Int J Mol Sci. 2019;20(9). [PMC free article] [PubMed] [Google Scholar]
41. Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, et al. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature. 2012;481(7382):463–8. [PMC free article] [PubMed] [Google Scholar]
42. Velazquez-Villegas LA, Perino A, Lemos V, Zietak M, Nomura M, Pols TWH, et al. TGR5 signalling promotes mitochondrial fission and beige remodelling of white adipose tissue. Nat Commun. 2018;9(1):245. [PMC free article] [PubMed] [Google Scholar]
43. Barbatelli G, Murano I, Madsen L, Hao Q, Jimenez M, Kristiansen K, et al. The emergence of cold-induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation. Am J Physiol Endocrinol Metab. 2010;298(6):E1244–53. [PubMed] [Google Scholar]
44. Lee YH, Petkova AP, Konkar AA, Granneman JG. Cellular origins of cold-induced brown adipocytes in adult mice. FASEB J. 2015;29(1):286–99. [PMC free article] [PubMed] [Google Scholar]
45. Lee YH, Kim SN, Kwon HJ, Maddipati KR, Granneman JG. Adipogenic role of alternatively activated macrophages in beta-adrenergic remodeling of white adipose tissue. Am J Physiol Regul Integr Comp Physiol. 2016;310(1):R55–65. [PMC free article] [PubMed] [Google Scholar]
46. Lee YH, Petkova AP, Mottillo EP, Granneman JG. In vivo identification of bipotential adipocyte progenitors recruited by beta3-adrenoceptor activation and high-fat feeding. Cell Metab. 2012;15(4):480–91. [PMC free article] [PubMed] [Google Scholar]
47. Himms-Hagen J, Melnyk A, Zingaretti MC, Ceresi E, Barbatelli G, Cinti S. Multilocular fat cells in WAT of CL-316243-treated rats derive directly from white adipocytes. Am J Physiol Cell Physiol. 2000;279(3):C670–81. [PubMed] [Google Scholar]
48. Rosenwald M, Perdikari A, Rulicke T, Wolfrum C. Bi-directional interconversion of brite and white adipocytes. Nat Cell Biol. 2013;15(6):659–67. [PubMed] [Google Scholar]
49. Vishvanath L, MacPherson KA, Hepler C, Wang QA, Shao M, Spurgin SB, et al. Pdgfrbeta+ Mural Preadipocytes Contribute to Adipocyte Hyperplasia Induced by High-Fat-Diet Feeding and Prolonged Cold Exposure in Adult Mice. Cell Metab. 2016;23(2):350–9. [PMC free article] [PubMed] [Google Scholar]
50. Carriere A, Jeanson Y, Berger-Muller S, Andre M, Chenouard V, Arnaud E, et al. Browning of white adipose cells by intermediate metabolites: an adaptive mechanism to alleviate redox pressure. Diabetes. 2014;63(10):3253–65. [PubMed] [Google Scholar]
51. Lin JZ, Martagon AJ, Cimini SL, Gonzalez DD, Tinkey DW, Biter A, et al. Pharmacological Activation of Thyroid Hormone Receptors Elicits a Functional Conversion of White to Brown Fat. Cell Rep. 2015;13(8):1528–37. [PMC free article] [PubMed] [Google Scholar]
52. Fisher FM, Kleiner S, Douris N, Fox EC, Mepani RJ, Verdeguer F, et al. FGF21 regulates PGC-1alpha and browning of white adipose tissues in adaptive thermogenesis. Genes Dev. 2012;26(3):271–81. [PMC free article] [PubMed] [Google Scholar]
53. Lee YH, Kim SN, Kwon HJ, Granneman JG. Metabolic heterogeneity of activated beige/brite adipocytes in inguinal adipose tissue. Sci Rep. 2017;7:39794. [PMC free article] [PubMed] [Google Scholar]
54. Jiang Y, Berry DC, Graff JM. Distinct cellular and molecular mechanisms for beta3 adrenergic receptor-induced beige adipocyte formation. Elife. 2017;6. [PMC free article] [PubMed] [Google Scholar]
55. Sidossis L, Kajimura S. Brown and beige fat in humans: thermogenic adipocytes that control energy and glucose homeostasis. J Clin Invest. 2015;125(2):478–86. [PMC free article] [PubMed] [Google Scholar]
56. Berry DC, Jiang Y, Graff JM. Mouse strains to study cold-inducible beige progenitors and beige adipocyte formation and function. Nat Commun. 2016;7:10184. [PMC free article] [PubMed] [Google Scholar]
57. Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat Rev Drug Discov. 2016;15(9):639–60. [PubMed] [Google Scholar]
58. Eto H, Suga H, Matsumoto D, Inoue K, Aoi N, Kato H, et al. Characterization of structure and cellular components of aspirated and excised adipose tissue. Plast Reconstr Surg. 2009;124(4):1087–97. [PubMed] [Google Scholar]
59. Prunet-Marcassus B, Cousin B, Caton D, Andre M, Penicaud L, Casteilla L. From heterogeneity to plasticity in adipose tissues: site-specific differences. Exp Cell Res. 2006;312(6):727–36. [PubMed] [Google Scholar]
60. Lee MJ, Wu Y, Fried SK. Adipose tissue heterogeneity: implication of depot differences in adipose tissue for obesity complications. Mol Aspects Med. 2013;34(1):1–11. [PMC free article] [PubMed] [Google Scholar]
61. Bora P, Majumdar AS. Adipose tissue-derived stromal vascular fraction in regenerative medicine: a brief review on biology and translation. Stem Cell Res Ther. 2017;8(1):145. [PMC free article] [PubMed] [Google Scholar]
62. Cinti S The adipose organ: morphological perspectives of adipose tissues. Proc Nutr Soc. 2001;60(3):319–28. [PubMed] [Google Scholar]
63. Rodeheffer MS, Birsoy K, Friedman JM. Identification of white adipocyte progenitor cells in vivo. Cell. 2008;135(2):240–9. [PubMed] [Google Scholar]
64. Burl RB, Ramseyer VD, Rondini EA, Pique-Regi R, Lee YH, Granneman JG. Deconstructing Adipogenesis Induced by beta3-Adrenergic Receptor Activation with Single-Cell Expression Profiling. Cell Metab. 2018;28(2):300–9 e4. [PMC free article] [PubMed] [Google Scholar]
65. Kalisky T, Oriel S, Bar-Lev TH, Ben-Haim N, Trink A, Wineberg Y, et al. A brief review of single-cell transcriptomic technologies. Brief Funct Genomics. 2018;17(1):64–76. [PubMed] [Google Scholar]
66. Fang P, Li X, Dai J, Cole L, Camacho JA, Zhang Y, et al. Immune cell subset differentiation and tissue inflammation. J Hematol Oncol. 2018;11(1):97. [PMC free article] [PubMed] [Google Scholar]
67. Trapnell C Defining cell types and states with single-cell genomics. Genome Res. 2015;25(10):1491–8. [PMC free article] [PubMed] [Google Scholar]
68. Stegle O, Teichmann SA, Marioni JC. Computational and analytical challenges in single-cell transcriptomics. Nat Rev Genet. 2015;16(3):133–45. [PubMed] [Google Scholar]
69. Nguyen A, Khoo WH, Moran I, Croucher PI, Phan TG. Single Cell RNA Sequencing of Rare Immune Cell Populations. Front Immunol. 2018;9:1553. [PMC free article] [PubMed] [Google Scholar]
70. Prakadan SM, Shalek AK, Weitz DA. Scaling by shrinking: empowering single-cell ‘omics’ with microfluidic devices. Nat Rev Genet. 2017;18(6):345–61. [PMC free article] [PubMed] [Google Scholar]
71. Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell. 2015;161(5):1187–201. [PMC free article] [PubMed] [Google Scholar]
72. Zilionis R, Nainys J, Veres A, Savova V, Zemmour D, Klein AM, et al. Single-cell barcoding and sequencing using droplet microfluidics. Nat Protoc. 2017;12(1):44–73. [PubMed] [Google Scholar]
73. Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell. 2015;161(5):1202–14. [PMC free article] [PubMed] [Google Scholar]
74. Zheng GX, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, et al. Massively parallel digital transcriptional profiling of single cells. Nat Commun. 2017;8:14049. [PMC free article] [PubMed] [Google Scholar]
75. AlJanahi AA, Danielsen M, Dunbar CE. An Introduction to the Analysis of Single-Cell RNA-Sequencing Data. Mol Ther Methods Clin Dev. 2018;10:189–96. [PMC free article] [PubMed] [Google Scholar]
76. Ziegenhain C, Vieth B, Parekh S, Reinius B, Guillaumet-Adkins A, Smets M, et al. Comparative Analysis of Single-Cell RNA Sequencing Methods. Mol Cell. 2017;65(4):631–43 e4. [PubMed] [Google Scholar]
77. Chen G, Ning B, Shi T. Single-Cell RNA-Seq Technologies and Related Computational Data Analysis. Front Genet. 2019;10:317. [PMC free article] [PubMed] [Google Scholar]
78. Zhang X, Li T, Liu F, Chen Y, Yao J, Li Z, et al. Comparative Analysis of Droplet-Based Ultra-High-Throughput Single-Cell RNA-Seq Systems. Mol Cell. 2019;73(1):130–42 e5. [PubMed] [Google Scholar]
79. Hwang B, Lee JH, Bang D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med. 2018;50(8):96. [PMC free article] [PubMed] [Google Scholar]
80. Kivioja T, Vaharautio A, Karlsson K, Bonke M, Enge M, Linnarsson S, et al. Counting absolute numbers of molecules using unique molecular identifiers. Nat Methods. 2011;9(1):72–4. [PubMed] [Google Scholar]
81. Andrews TS, Hemberg M. Identifying cell populations with scRNASeq. Mol Aspects Med. 2018;59:114–22. [PubMed] [Google Scholar]
82. Luecken MD, Theis FJ. Current best practices in single-cell RNA-seq analysis: a tutorial. Mol Syst Biol. 2019;15(6):e8746. [PMC free article] [PubMed] [Google Scholar]
83. Svensson V, Pachter L. RNA Velocity: Molecular Kinetics from Single-Cell RNA-Seq. Mol Cell. 2018;72(1):7–9. [PubMed] [Google Scholar]
84. La Manno G, Soldatov R, Zeisel A, Braun E, Hochgerner H, Petukhov V, et al. RNA velocity of single cells. Nature. 2018;560(7719):494–8. [PMC free article] [PubMed] [Google Scholar]
85. Aibar S, Gonzalez-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulselmans G, et al. SCENIC: single-cell regulatory network inference and clustering. Nat Methods. 2017;14(11):1083–6. [PMC free article] [PubMed] [Google Scholar]
86. Fiers M, Minnoye L, Aibar S, Bravo Gonzalez-Blas C, Kalender Atak Z, Aerts S. Mapping gene regulatory networks from single-cell omics data. Brief Funct Genomics. 2018;17(4):246–54. [PMC free article] [PubMed] [Google Scholar]
87. Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB v2.0: Inferring cell-cell communication from combined expression of multi-subunit receptor-ligand complexes. bioRxiv. 2019. [PubMed] [Google Scholar]
88. Stuart T, Satija R. Integrative single-cell analysis. Nat Rev Genet. 2019;20(5):257–72. [PubMed] [Google Scholar]
89. Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36(5):411–20. [PMC free article] [PubMed] [Google Scholar]
90. Berry R, Rodeheffer MS. Characterization of the adipocyte cellular lineage in vivo. Nat Cell Biol. 2013;15(3):302–8. [PMC free article] [PubMed] [Google Scholar]
91. Krueger KC, Costa MJ, Du H, Feldman BJ. Characterization of Cre recombinase activity for in vivo targeting of adipocyte precursor cells. Stem Cell Reports. 2014;3(6):1147–58. [PMC free article] [PubMed] [Google Scholar]
92. Lee YH, Petkova AP, Granneman JG. Identification of an adipogenic niche for adipose tissue remodeling and restoration. Cell Metab. 2013;18(3):355–67. [PMC free article] [PubMed] [Google Scholar]
93. Tran KV, Gealekman O, Frontini A, Zingaretti MC, Morroni M, Giordano A, et al. The vascular endothelium of the adipose tissue gives rise to both white and brown fat cells. Cell Metab. 2012;15(2):222–9. [PMC free article] [PubMed] [Google Scholar]
94. Tang W, Zeve D, Suh JM, Bosnakovski D, Kyba M, Hammer RE, et al. White fat progenitor cells reside in the adipose vasculature. Science. 2008;322(5901):583–6. [PMC free article] [PubMed] [Google Scholar]
95. Gupta RK, Mepani RJ, Kleiner S, Lo JC, Khandekar MJ, Cohen P, et al. Zfp423 expression identifies committed preadipocytes and localizes to adipose endothelial and perivascular cells. Cell Metab. 2012;15(2):230–9. [PMC free article] [PubMed] [Google Scholar]
96. Gavin KM, Gutman JA, Kohrt WM, Wei Q, Shea KL, Miller HL, et al. De novo generation of adipocytes from circulating progenitor cells in mouse and human adipose tissue. FASEB J. 2016;30(3):1096–108. [PMC free article] [PubMed] [Google Scholar]
97. Majka SM, Fox KE, Psilas JC, Helm KM, Childs CR, Acosta AS, et al. De novo generation of white adipocytes from the myeloid lineage via mesenchymal intermediates is age, adipose depot, and gender specific. Proc Natl Acad Sci U S A. 2010;107(33):14781–6. [PMC free article] [PubMed] [Google Scholar]
98. Wassermann F, Mc DT. Electron microscopic investigation on the surface membrane structures of the fat-cell and of their changes during depletion of the cell. Z Zellforsch Mikrosk Anat. 1960;52:778–800. [PubMed] [Google Scholar]
99. Cinti S, Cigolini M, Bosello O, Bjorntorp P. A morphological study of the adipocyte precursor. J Submicrosc Cytol. 1984;16(2):243–51. [PubMed] [Google Scholar]
100. Crandall DL, Hausman GJ, Kral JG. A review of the microcirculation of adipose tissue: anatomic, metabolic, and angiogenic perspectives. Microcirculation. 1997;4(2):211–32. [PubMed] [Google Scholar]
101. Poissonnet CM, Burdi AR, Bookstein FL. Growth and development of human adipose tissue during early gestation. Early Hum Dev. 1983;8(1):1–11. [PubMed] [Google Scholar]
102. Merrick D, Sakers A, Irgebay Z, Okada C, Calvert C, Morley MP, et al. Identification of a mesenchymal progenitor cell hierarchy in adipose tissue. Science. 2019;364(6438). [PMC free article] [PubMed] [Google Scholar]
103. Schwalie PC, Dong H, Zachara M, Russeil J, Alpern D, Akchiche N, et al. A stromal cell population that inhibits adipogenesis in mammalian fat depots. Nature. 2018;559(7712):103–8. [PubMed] [Google Scholar]
104. Hepler C, Shan B, Zhang Q, Henry GH, Shao M, Vishvanath L, et al. Identification of functionally distinct fibro-inflammatory and adipogenic stromal subpopulations in visceral adipose tissue of adult mice. Elife. 2018;7. [PMC free article] [PubMed] [Google Scholar]
105. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, et al. Comprehensive Integration of Single-Cell Data. Cell. 2019;177(7):1888–902 e21. [PMC free article] [PubMed] [Google Scholar]
106. Chau YY, Hastie N. Wt1, the mesothelium and the origins and heterogeneity of visceral fat progenitors. Adipocyte. 2015;4(3):217–21. [PMC free article] [PubMed] [Google Scholar]
107. Mutsaers SE. The mesothelial cell. Int J Biochem Cell B. 2004;36(1):9–16. [PubMed] [Google Scholar]
108. Gupta OT, Gupta RK. Visceral Adipose Tissue Mesothelial Cells: Living on the Edge or Just Taking Up Space? Trends Endocrinol Metab. 2015;26(10):515–23. [PubMed] [Google Scholar]
109. Chau YY, Bandiera R, Serrels A, Martinez-Estrada OM, Qing W, Lee M, et al. Visceral and subcutaneous fat have different origins and evidence supports a mesothelial source. Nat Cell Biol. 2014;16(4):367–75. [PMC free article] [PubMed] [Google Scholar]
110. Hudak CS, Gulyaeva O, Wang Y, Park SM, Lee L, Kang C, et al. Pref-1 marks very early mesenchymal precursors required for adipose tissue development and expansion. Cell Rep. 2014;8(3):678–87. [PMC free article] [PubMed] [Google Scholar]
111. Sanchez-Gurmaches J, Hsiao WY, Guertin DA. Highly selective in vivo labeling of subcutaneous white adipocyte precursors with Prx1-Cre. Stem Cell Reports. 2015;4(4):541–50. [PMC free article] [PubMed] [Google Scholar]
112. Sanchez-Gurmaches J, Guertin DA. Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed. Nat Commun. 2014;5:4099. [PMC free article] [PubMed] [Google Scholar]
113. Ruiz-Ojeda FJ, Ruperez AI, Gomez-Llorente C, Gil A, Aguilera CM. Cell Models and Their Application for Studying Adipogenic Differentiation in Relation to Obesity: A Review. Int J Mol Sci. 2016;17(7). [PMC free article] [PubMed] [Google Scholar]
114. Poulos SP, Dodson MV, Culver MF, Hausman GJ. The increasingly complex regulation of adipocyte differentiation. Exp Biol Med (Maywood). 2016;241(5):449–56. [PMC free article] [PubMed] [Google Scholar]
115. Kuri-Harcuch W, Velez-delValle C, Vazquez-Sandoval A, Hernandez-Mosqueira C, Fernandez-Sanchez V. A cellular perspective of adipogenesis transcriptional regulation. J Cell Physiol. 2019;234(2):1111–29. [PubMed] [Google Scholar]
116. Rosen ED, MacDougald OA. Adipocyte differentiation from the inside out. Nat Rev Mol Cell Biol. 2006;7(12):885–96. [PubMed] [Google Scholar]
117. Lee YH, Thacker RI, Hall BE, Kong R, Granneman JG. Exploring the activated adipogenic niche: interactions of macrophages and adipocyte progenitors. Cell Cycle. 2014;13(2):184–90. [PMC free article] [PubMed] [Google Scholar]
118. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res. 2005;46(11):2347–55. [PubMed] [Google Scholar]
119. Marcelin G, Ferreira A, Liu Y, Atlan M, Aron-Wisnewsky J, Pelloux V, et al. A PDGFRalpha-Mediated Switch toward CD9(high) Adipocyte Progenitors Controls Obesity-Induced Adipose Tissue Fibrosis. Cell Metab. 2017;25(3):673–85. [PubMed] [Google Scholar]
120. Kato S, Yuzawa Y, Tsuboi N, Maruyama S, Morita Y, Matsuguchi T, et al. Endotoxin-induced chemokine expression in murine peritoneal mesothelial cells: the role of toll-like receptor 4. J Am Soc Nephrol. 2004;15(5):1289–99. [PubMed] [Google Scholar]
121. Colmont CS, Raby AC, Dioszeghy V, Lebouder E, Foster TL, Jones SA, et al. Human peritoneal mesothelial cells respond to bacterial ligands through a specific subset of Toll-like receptors. Nephrol Dial Transplant. 2011;26(12):4079–90. [PMC free article] [PubMed] [Google Scholar]
122. Mutsaers SE, Whitaker D, Papadimitriou JM. Stimulation of mesothelial cell proliferation by exudate macrophages enhances serosal wound healing in a murine model. Am J Pathol. 2002;160(2):681–92. [PMC free article] [PubMed] [Google Scholar]
123. Whitaker D, Papadimitriou J. Mesothelial healing: morphological and kinetic investigations. J Pathol. 1985;145(2):159–75. [PubMed] [Google Scholar]
124. Zhang Z, Shao M, Hepler C, Zi Z, Zhao S, An YA, et al. Dermal adipose tissue has high plasticity and undergoes reversible dedifferentiation in mice. J Clin Invest. 2019;129(12):5327–42. [PMC free article] [PubMed] [Google Scholar]
125. Wang QA, Song A, Chen W, Schwalie PC, Zhang F, Vishvanath L, et al. Reversible De-differentiation of Mature White Adipocytes into Preadipocyte-like Precursors during Lactation. Cell Metab. 2018;28(2):282–8 e3. [PMC free article] [PubMed] [Google Scholar]
126. Festa E, Fretz J, Berry R, Schmidt B, Rodeheffer M, Horowitz M, et al. Adipocyte lineage cells contribute to the skin stem cell niche to drive hair cycling. Cell. 2011;146(5):761–71. [PMC free article] [PubMed] [Google Scholar]
127. Murano I, Barbatelli G, Parisani V, Latini C, Muzzonigro G, Castellucci M, et al. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. J Lipid Res. 2008;49(7):1562–8. [PubMed] [Google Scholar]
128. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–808. [PMC free article] [PubMed] [Google Scholar]
129. Han MS, Jung DY, Morel C, Lakhani SA, Kim JK, Flavell RA, et al. JNK expression by macrophages promotes obesity-induced insulin resistance and inflammation. Science. 2013;339(6116):218–22. [PMC free article] [PubMed] [Google Scholar]
130. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117(1):175–84. [PMC free article] [PubMed] [Google Scholar]
131. Fujisaka S, Usui I, Bukhari A, Ikutani M, Oya T, Kanatani Y, et al. Regulatory mechanisms for adipose tissue M1 and M2 macrophages in diet-induced obese mice. Diabetes. 2009;58(11):2574–82. [PMC free article] [PubMed] [Google Scholar]
132. Hill DA, Lim HW, Kim YH, Ho WY, Foong YH, Nelson VL, et al. Distinct macrophage populations direct inflammatory versus physiological changes in adipose tissue. Proc Natl Acad Sci U S A. 2018;115(22):E5096–E105. [PMC free article] [PubMed] [Google Scholar]
133. Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B, Descamps H, et al. Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner. Cell. 2019. [PMC free article] [PubMed] [Google Scholar]
134. Weinstock A, Brown EJ, Garabedian ML, Pena S, Sharma M, Lafaille J, et al. Single-Cell RNA Sequencing of Visceral Adipose Tissue Leukocytes Reveals that Caloric Restriction Following Obesity Promotes the Accumulation of a Distinct Macrophage Population with Features of Phagocytic Cells. Immunometabolism. 2019;1. [PMC free article] [PubMed] [Google Scholar]
135. Granneman JG, Li P, Zhu Z, Lu Y. Metabolic and cellular plasticity in white adipose tissue I: effects of beta3-adrenergic receptor activation. Am J Physiol Endocrinol Metab. 2005;289(4):E608–16. [PubMed] [Google Scholar]
136. Tchkonia T, Thomou T, Zhu Y, Karagiannides I, Pothoulakis C, Jensen MD, et al. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab. 2013;17(5):644–56. [PMC free article] [PubMed] [Google Scholar]
137. Schoettl T, Fischer IP, Ussar S. Heterogeneity of adipose tissue in development and metabolic function. J Exp Biol. 2018;221(Pt Suppl 1). [PubMed] [Google Scholar]
138. Cinti S, Cancello R, Zingaretti MC, Ceresi E, De Matteis R, Giordano A, et al. CL316,243 and cold stress induce heterogeneous expression of UCP1 mRNA and protein in rodent brown adipocytes. J Histochem Cytochem. 2002;50(1):21–31. [PubMed] [Google Scholar]
139. Song A, Dai W, Jang MJ, Medrano L, Li Z, Zhao H, et al. Low- and high-thermogenic brown adipocyte subpopulations coexist in murine adipose tissue. J Clin Invest. 2019. [PMC free article] [PubMed] [Google Scholar]
140. Habib N, Avraham-Davidi I, Basu A, Burks T, Shekhar K, Hofree M, et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat Methods. 2017;14(10):955–8. [PMC free article] [PubMed] [Google Scholar]
141. Habib N, Li Y, Heidenreich M, Swiech L, Avraham-Davidi I, Trombetta JJ, et al. Div-Seq: Single-nucleus RNA-Seq reveals dynamics of rare adult newborn neurons. Science. 2016;353(6302):925–8. [PMC free article] [PubMed] [Google Scholar]
142. Bakken TE, Hodge RD, Miller JA, Yao Z, Nguyen TN, Aevermann B, et al. Single-nucleus and single-cell transcriptomes compared in matched cortical cell types. PLoS One. 2018;13(12):e0209648. [PMC free article] [PubMed] [Google Scholar]
143. Wu H, Kirita Y, Donnelly EL, Humphreys BD. Advantages of Single-Nucleus over Single-Cell RNA Sequencing of Adult Kidney: Rare Cell Types and Novel Cell States Revealed in Fibrosis. J Am Soc Nephrol. 2019;30(1):23–32. [PMC free article] [PubMed] [Google Scholar]
144. Rajbhandari P, Arneson D, Hart SK, Ahn IS, Diamante G, Santos LC, et al. Single cell analysis reveals immune cell-adipocyte crosstalk regulating the transcription of thermogenic adipocytes. Elife. 2019;8. [PMC free article] [PubMed] [Google Scholar]
145. Eng CL, Lawson M, Zhu Q, Dries R, Koulena N, Takei Y, et al. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH. Nature. 2019;568(7751):235–9. [PMC free article] [PubMed] [Google Scholar]
146. Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1):W90–7. [PMC free article] [PubMed] [Google Scholar]
-