Skip to main content

Dissecting human adipose tissue heterogeneity using single‐cell omics technologies

Abstract

Single-cell omics technologies that profile genes (genomic and epigenomic) and determine the abundance of mRNA (transcriptomic), protein (proteomic and secretomic), lipids (lipidomic), and extracellular matrix (matrisomic) support the dissection of adipose tissue heterogeneity at unprecedented resolution in a temporally and spatially defined manner. In particular, cell omics technologies may provide innovative biomarkers for the identification of rare specific progenitor cell subpopulations, assess transcriptional and proteomic changes affecting cell proliferation and immunomodulatory potential, and accurately define the lineage hierarchy and differentiation status of progenitor cells. Unraveling adipose tissue complexity may also provide for the precise assessment of a dysfunctional state, which has been associated with cancer, as cancer-associated adipocytes play an important role in shaping the tumor microenvironment supporting tumor progression and metastasis, obesity, metabolic syndrome, and type 2 diabetes mellitus. The information collected by single-cell omics has relevant implications for regenerative medicine because adipose tissue is an accessible source of multipotent cells; alternative cell-free approaches, including the use of adipose tissue stromal cell-conditioned medium, extracellular vesicles, or decellularized extracellular matrix, are clinically valid options. Subcutaneous white adipose tissue, which is generally harvested via liposuction, is highly heterogeneous because of intrinsic biological variability and extrinsic inconsistencies in the harvesting and processing procedures. The current limited understanding of adipose tissue heterogeneity impinges on the definition of quality standards appropriate for clinical translation, which requires consistency and uniformity of the administered product. We review the methods used for dissecting adipose tissue heterogeneity and provide an overview of advances in omics technology that may contribute to the exploration of heterogeneity and dynamics of adipose tissue at the single-cell level.

Introduction

Over the last decades, the therapeutic potential of autologous adipose tissue-derived material transplantation has been explored in several regenerative medicine clinical trials [1]. Most of the preclinical and translational research employed adipose tissue-derived stromal cells isolated by enzymatic digestion of white adipose tissue (WAT), mainly obtained from subcutaneous fat depots [2, 3]; the regenerative potential of mechanically manipulated adipose tissue has also been evaluated [4]; moreover, adipose tissue paracrine factors, including extracellular vesicles (EVs) and cell-conditioned medium, and decellularized extracellular matrix, have also been considered as cell-free therapeutics [5], in order to circumvent the limited transplanted cell viability in the hypoxic microenvironment at the administration site [6, 7]. Therapeutic efficacy of adipose tissue derivatives in regenerative medicine, dermatology, plastic and aesthetic surgery procedures has been ascribed to both the action of resident multipotent stromal cells and paracrine signals. Nonetheless, inconsistencies in the methods used for adipose tissue manipulation currently limit the reproducible therapeutic efficacy of adipose tissue derivatives, but progressive efforts in the standardization of the procedures will eventually address this issue. Another source of variability is the intrinsic biological heterogeneity, which requires extensive studies at the cellular level to unravel the biological properties, function and clinical potential of adipose tissue derivatives [8]. Single-cell technologies measure the abundance of molecules in individual cells and represent a unique tool to finely dissect tissue cellular and functional heterogeneity, which has been more extensively used for the analysis of the human tumor microenvironment [9]. Single-cell omics analysis of adipose tissue heterogeneity may support the identification and isolation of more potent cellular subpopulations and paracrine factors with preferred regenerative properties [10,11,12]. Therefore, the exploitation of technologies for single-cell analysis can be instrumental in more precisely elucidating the mechanisms underlying stem cell differentiation potentials, possibly finding druggable targets to promote tissue regeneration [13] and clinical translation [14].

Here, we review the current methods and provide an overview of the advances in omics technology used to better define and refine the regenerative potential of adipose tissue derivatives.

Intrinsic and extrinsic factors affecting adipose tissue derivative heterogeneity

The comprehensive evaluation of adipose tissue is challenging because is a heterogeneous and dynamic organ. Adipose tissue is the main reservoir of energy substrates and endocrine mediators, including growth factors, hormones, cytokines, and microRNAs, which contribute to whole-body energy metabolism and homeostasis. Moreover, adipose tissue has a key role in immune response, mechanical protection, thermogenesis, and thermal insulation. This variety of functions is reflected in its heterogeneous composition: mature adipocytes are the distinctive cell type and encompass up to 80% of the volume of a white adipose tissue depot; however, multiple additional cytotypes are also present, including preadipocytes, endothelial cells, pericytes, immune cells such as T cells and macrophages, and mesenchymal/stromal progenitor/stem cells, which collectively form the adipose tissue stromal vascular fraction (SVF) [15]. Interestingly, although SVF occupies only a minor part of the volume of an adipose tissue depot, it comprises approximately 60–80% of the total cell number. In addition, an extracellular matrix (ECM) network, mainly composed of collagens and proteoglycans, comprises the adipose tissue niche, which exerts biomechanical functions and serves as a reservoir of cytokines, growth factors, and extracellular vesicles that influence cell shape and metabolism. Cross-talk between the different components regulates both adipocyte fate and function and supports tissue dynamic plasticity to respond to the energy needs of the organism through adipocyte hypertrophy, proliferation of precursor cells, and ECM remodeling.

The adipose tissue composition of humans cannot be predicted from studies on mice [16] and other vertebrates [17, 18] due to significant differences in their metabolism and physiology. In humans, adipose tissue depots are highly dynamic and variable in distribution and dimension, depending on several factors, including donor genetics, epigenetics, gender, age, ethnic background, metabolism and nutritional status, and response to hormones or drugs. As a result, there is a substantial donor-to-donor variation in adipose tissue composition; moreover, different cell populations are present in different fat depots and local cellular composition variety exists within the same white adipose tissue depot, resulting in intraindividual variation; furthermore, cell populations respond to their specific tissue microenvironment and neighboring cells. Therefore, it is possible that adipose tissue heterogenicity may arise, at least in part, by cell transition in different states in response to intra- and inter-tissue cellular crosstalk (Fig. 1). Health status strongly affects adipose tissue endocrine and paracrine functions, as dysfunctional adipose tissue has been associated with obesity, metabolic syndrome, type 2 diabetes mellitus and cancer-associated cachexia [19, 20]. Active cross talk between adipose tissue and cancer exists, as some tumors (including, prostate, breast, and ovary) invade the surrounding adipose tissue, whereas adipose tissue can promote carcinogenesis via paracrine and metabolic modulation [21]. Furthermore, the stromal component of the tumor microenvironment is characterized by the presence of cancer-associated adipocytes, which play an active role in supporting tumor progression and metastasis, and therapy resistance via metabolic reprogramming and paracrine-mediated crosstalk with tumor cells [22, 23]. Cancer-associated adipocytes are characterized by an activated phenotype, morphological and functional changes, reduced lipid content and expression of adipocyte markers [24]; specific classes of tumor‑promoting adipocytes characterized by expression of dipeptidyl peptidase-4 (DPP4) and adiponectin (ADIPOQ) have been recently identified [25]. In addition, adipose stromal cells shape the tumor immune microenvironment originating collagen 11A-expressing cancer-associated fibroblasts that promote tumor ECM remodeling and modulate antitumor immune response [26]. The contribution of adipocytes in cancer has often been underestimated, as well as their potential role as therapeutic targets [27]; however, recently, Sato defined as "adipo-oncology" a new area of research to improve our understanding of the role of adipocytes to increase the effectiveness of metastatic tumor therapy [23].

Fig. 1
figure 1

Major determinants affecting adipose tissue heterogeneity. BMI: body mass index; ECM: extracellular matrix. Templates were adapted from Servier Medical Art (https://smart.servier.com/, accessed: 05 September 2024), available under a Creative Commons Attribution 3.0 Unported License

Based on tissue anatomical location, white adipose tissue (WAT) is subcategorized into subcutaneous (sWAT or SAT) and visceral or omental (vWAT or VAT) fat; differences in the anatomy and function between sWAT and vWAT [28] distinctively affect the corresponding cell gene and protein expression profiles [29,30,31,32]. In humans, distinct subcutaneous and dermal adipose tissue depots are located beneath the skin, mainly in the abdominal, femoral and gluteal regions. Subcutaneous WAT can be easily collected by liposuction, digested by collagenase, followed by washing, filtration, and centrifugation steps to collect the cellular SVF (cSVF) fraction encompassing multipotent cells able to differentiate into adipogenic, chondrogenic, and osteogenic cells [33]. Freshly isolated cSVF can be used either for therapeutic purposes or for in vitro expansion. Adipose tissue-derived stromal cells (ASCs) are isolated by culturing cSVF and maintaining only plastic-adherent cells. The yield of ASCs depends on several parameters, including the harvesting procedure, isolation protocol, and cell counting method [34, 35]. Moreover, the conditions used for ex vivo cell culture amplification influence various aspects of ASC gene expression, functional, proliferative and differentiative potentials [36, 37]. Alternatively, the adipose tissue stromal vascular fraction (tSVF) can be obtained by minimal, nonenzymatic manipulation of lipoaspirate [4]. This procedure eliminates the prevalent adipocytic fraction and preserves, in addition to the SVF cellular components, the ECM, which has biomechanical functions and can store soluble factors with immune regulatory, angiogenic and tissue reparative capabilities. Non-enzymatic manipulation can be performed using different devices and protocols; however, standardization of the procedures is needed for a reliable prediction of the treatment outcome [38].

Methods for dissecting adipose tissue heterogeneity

Several methods have been used to study adipose tissue morphological, cellular and functional heterogeneity (Table 1). Some technologies, mainly based on histological examination, can be performed on adipose tissue samples and allow for the analysis of tissue morphology and three-dimensional (3D) structure. Other applications analyze mixtures of intact cells, either freshly isolated or upon ex vivo culture, after adipose tissue disaggregation. Innovative transcriptomic techniques aim to provide a more comprehensive understanding of the contribution of each cell type by single-cell analysis [12, 39]. In the following sections, we briefly review the main methods used for dissecting adipose tissue heterogeneity.

Table 1 Approaches for the study of adipose tissue heterogeneity

Adipose tissue histology

Earlier attempts to characterize white adipose tissue cellular components have mostly relied on histology to study the morphology of the cells, their anatomical localization [40] and quantify adipocyte cellularity [41]. Subcutaneous white adipose tissue, which represents over 80% of the total fat, is mainly organized into mature unilocular adipocytes and connective tissue septa. In unilocular adipocytes, a large lipid-filled vacuole occupies most of the cytoplasm and the nucleus is localized in the periphery. Under light microscope examination of routine hematoxylin and eosin-stained adipose tissue sections, adipocytes appear as ‘empty cells’ since their typical lipid droplets are extracted by the organic solvents used for tissue processing. The collagen fibers in the septa of loose connective tissue that separate the adipose tissue lobules can be visualized by Picrosirius red or Masson’s trichrome staining. Paraffin-embedded sections of adipose tissue can also be used for immunofluorescence and immunohistochemistry-based approaches to detect the expression of specific adipose tissue markers, including CCAAT enhancer binding protein beta (C/EBP β), peroxisome proliferator-activated receptor-gamma (PPAR-γ), caveolin-1, perilipin-1 and preadipocyte factor-1 (Pref-1) to label progenitor and mature adipocytes [42]. Nonetheless, frozen sectioning generally allows for more effective antigen labeling with fluorochrome-conjugated antibodies compared with formalin-included sections.

Electron microscopy (transmission and scanning) techniques achieve considerably higher resolution than optical histologic analysis. Using electron microscopy, Sbarbati et al. characterized subcutaneous WAT from several depots, highlighting specific 3D morphological and ultrastructural differences mainly related to the composition of the stromal and microvascular compartments of adipose tissue [43]. Accordingly, sWAT harvested from areas including trochanters, inner faces of the knees, thighs, and hips, in virtue of their strong vascular and stromal fraction, may represent the optimal source of adipose tissue to be used for regenerative procedures.

Whole mounted adipose tissue imaging

Adipose tissue specimens stained with fluorophores can be visualized by whole mount imaging using confocal microscopy [44]. The processing of adipose tissue for whole mounted imaging does not require fixation, embedding, or sectioning; thus, adipose tissue morphology is preserved, allowing for 3D visualization of tissue architecture and providing indications on possible interactions between different cellular components. Furthermore, organotypic slice cultures of human adipose tissue represent a suitable model in which tissue morphology and metabolism are preserved for up to 2 weeks in culture [45]. The major obstacle is that fluorescently conjugated antibodies or fluorescent stains do not fully permeate the tissue. Notwithstanding this limitation, whole mount imaging has been instrumental for the identification of the adipocyte cellular lineage in vivo in murine models [46].

Adipose tissue stromal cell functional and differentiation assays

The study of adipose tissue-derived cells obtained by enzymatic dissociation may provide more in-depth insights into the contribution of different cell types to adipose tissue function. Adipose tissue-derived stromal cells (ASCs) have a strong potential impact on regenerative cell therapy. These cells are plastic-adherent when maintained in standard culture conditions and should be characterized by functional assays to assess self-renewal and differentiation potentials before clinical use [47].

Colony-forming unit-fibroblastoid assay

The colony-forming unit-fibroblastoid (CFU-F) assay assesses ASC self-renewal potential by defining the number of progenitor cells. In this assay, ASCs are seeded at low density (40–400 cells/cm2), cultured for 10–14 days, and their clonal expansion is determined by counting the colonies after fixation and staining with crystal violet [47]. The self-renewal potential of ASCs is affected by donor age, body mass index and fat localization; moreover, ASCs at higher passage of culture tend to be senescent and show decreased proliferation rate.

Differentiation assays

In the presence of well-defined culture conditions in vitro, isolated ASCs undergo mesenchymal tri-lineage (adipocytes, osteoblasts, and chondrocytes) differentiation as well as into various nonmesodermal lineages (including cardiomyocytes, myocytes, endothelial cells, neurons, insulin producing cells, and hepatocytes) [33, 48,49,50,51,52].

According to the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT) guidelines for proper ASC characterization, adipocytic, chondrogenic and osteoblastic differentiation assays should be performed [47]. Accordingly, cells are maintained in culture in the appropriate inducing medium for approximately 2–4 weeks, and differentiation is then demonstrated by specific staining or by evaluating the expression of distinctive gene markers by reverse transcription quantitative polymerase chain reaction (RT-qPCR) (Table 2). The differentiation potential may be affected by cell culture passage and confluency. Slightly dissimilar differentiation protocols have been published, and some selective media and kits for confirmatory assays are commercially available [33, 53].

Table 2 Adipose tissue-derived stromal cells mesenchymal tri-lineage differentiation

The adipogenic induction medium typically contains glucocorticoids, 1-methyl-3-isobutylxanthine and insulin; rosiglitazone or other thiazolidinediones can be included as peroxisome proliferator-activated receptor-gamma (PPAR-γ) agonists. After approximately 2–3 weeks of culture, the formation of lipid droplets can be observed by phase-contrast microscopy after staining with Oil Red O or Nile red. Semi-quantification of the stained lipid droplets can be performed by measuring the absorbance of the eluted Oil red O stain. Recently, a deep learning-based method was developed to more accurately assess the kinetics of adipogenic differentiation of human ASCs [54]. Expression assessment of adipogenic transcription factors (PPAR γ, C/EBP α/β) and adipocyte genes (adiponectin, fatty acid binding protein 4, lipoprotein lipase, leptin, perilipin, adipose triglyceride lipase) can further validate adipogenic differentiation [55].

Different protocols for the chondrogenic differentiation of ASCs have been described [56]; the most common inducing medium comprises dexamethasone, ascorbate-2-phosphate, transforming growth factor beta 1 (TGF-β1), and insulin [57]. After 3–4 weeks of culture, the production of cartilage-like, sulfated proteoglycan-rich matrix can be detected using alcian blue, toluidine blue, or safranin-O staining. The presence of collagen (manly type II and X) can be assessed using Picrosirius red staining. The differentiation of ASCs to chondrocytes can be determined by the expression analysis of collagen type II and X, aggrecan, Sox-9, decorin, and biglycan.

Osteogenic induction medium comprises dexamethasone, ascorbate, beta-glycerophosphate, and 1,25 dihydroxy vitamin D3 [58]. After 2–4 weeks of culture, differentiated ASCs deposit calcium phosphate in the extracellular matrix, which can be detected by Alizarin red or von Kossa staining. The expression of osteogenic markers, including alkaline phosphatase, bone morphogenic proteins, and their receptors, can be assessed.

Phenotypic characterization of adipose tissue cells by flow cytometry

Collagenase digestion of adipose tissue permits the separation of adipocytes from the cellular stromal vascular fraction, although the isolation of lipid-filled adipocytes is technically difficult because of their fragility and high buoyancy. Isolated cells can then be characterized by flow cytometry (fluorescence-activated cell sorting—FACS), which allows the assessment of cell size and granularity, and the detection of multiple putative candidate lineage markers via fluorescent antibody staining. Limitations of flow cytometry techniques include the fact that lineage markers are mainly cell surface antigens, are not highly specific, and have heterogeneous expression. Thus, multiple markers, which can differ across studies, may be needed to detect specific cell populations; moreover, many cell surface proteins remain undetectable due to the limited commercial availability of specific antibodies. Nonetheless, FACS evaluation has critically contributed to the definition of the stromal cells isolated from adipose tissue that, according to the IFATS and ISCT guidelines, should express stromal cell surface markers (CD13, CD29, CD44, CD73, CD90, CD105), and should not express hematopoietic and endothelial antigens (CD31, CD45, CD235a) [47]. However, it should be noted that even ASCs that meet these criteria are heterogeneous, as variations in the phenotype might be observed at the origin or acquired during cell culture [59]. Accordingly, phenotypic characterization by FACS analysis should be performed to confirm ASC identity at different passages during in vitro expansion for the manufacture of cell-based therapies.

Purification/enrichment of specific ASC subpopulations based on their phenotype can be performed using immunoaffinity methods that rely on the binding of surface markers by specific monoclonal antibodies linked to fluorescent dyes or magnetic microbeads for fluorescent- or magnetic–activated cell sorting, FACS or MACS, respectively [60, 61]. It is also possible to perform negative selection of cells expressing ASC markers to isolate and characterize adipocytes even if fragility and large size make their purification challenging [62], or immune populations [63] within adipose tissue. Recently, the development of a microfluidic device for high-throughput, label-free enrichment of ASCs has been described [64].

Adipose tissue transcriptomics

The suffix “omic” defines new branches of research aiming at the identification and quantification of biomolecules that contribute to the molecular complexity and heterogeneity of biological samples. Single-cell and spatial omics technologies, in single-modality or in combination (multi-omics approach) [9, 65], as well as the ability of computational tools for data analysis and integration are rapidly evolving. These cutting edge techniques including genomic and epigenomic for the assessment of the profile of genes, proteomic and secretomic to detect the abundance of proteins, lipidomic to identify lipids, matrisomic for detection of extracellular matrix, metabolomics for metabolites, and transcriptomic for analysis of mRNAs, offer the unique opportunity to in-depth characterization of adipose tissue samples under physiological or pathological condition [10].

Currently, it is not possible to identify ASCs using a single marker. The progressive development of microarrays that enabled genome-scale expression analysis, improved high-throughput sequencing capabilities, and, more recently, single-cell RNA sequencing technologies provide an unprecedented opportunity to more accurately elucidate the phenotype, status and function of adipose tissue cellular components by simultaneous assessment of thousands of marker genes at as low as the cellular level. To collect the cellular transcriptomic signature, RNA is extracted and mRNA is selectively reverse-transcribed to generate a cDNA library that can be subsequently sequenced using high-throughput platforms. Then, sequencing data are mapped, the expression level of each gene is determined, and comparative transcriptome evaluation is performed. This analysis allows the identification of known and novel cell populations defining their unique transcriptomic profiles by unsupervised cell clustering. Moreover, using bioinformatics tools it is possible to gain a more comprehensive view of gene expression through the classification and organization into physiologically or pathologically significant networks. Bulk RNA transcriptome data average gene expression across all the cells within a tissue sample; thus, cellular heterogeneity cannot be precisely defined. Both bulk and single-cell RNA-sequencing require tissue disaggregation for efficient RNA extraction; thus, critical spatial information about adipose tissue is lost in the process. Spatial transcriptomics, which combines transcriptional analysis and in situ hybridization, has been developed to provide whole expression data with spatial information, ideally at a single-cell resolution. The transcriptome is highly dynamic because cells may respond to various environmental and pathological stimuli by modulating gene expression. Therefore, transcriptomic analysis may capture adipose tissue dysfunctional biological pathways associated with pathological states such as obesity and diabetes. In the following section, we review how the exploitation of innovative transcriptomics techniques has expanded our knowledge of adipose tissue.

Bulk RNA transcriptome analysis of adipose tissue and adipose tissue-derived stromal cells

Bulk RNA transcriptomics enables gene expression analysis in adipose tissue samples or pooled cell populations. In this approach, the entire biological sample is processed and RNA extracted to study gene expression using whole genome expression microarrays or RNA-sequencing. Thus, the results reveal the presence of specific RNAs and define the average spectrum of expression of gene markers across all the cells in the sample, yet the contribution of rare cell populations can be unnoticed. Comparison of bulk transcriptomic profiles is informative for assessing global variability among fat depots [66, 67] and mesenchymal stromal cells of different origin [68]; for instance, a higher aggregated profile of expression of genes important in endocrine function has been assessed in visceral compared with subcutaneous adipose tissue, in agreement with known functional differences between the two depots [69]. Bulk RNA transcriptomics can also be used to define gene expression dynamics during ASC isolation, expansion and differentiation. In particular, four distinct adipocyte subtypes have been identified by bulk transcriptomic analysis of human ASC clones before and after adipogenic differentiation, thus confirming adipocyte heterogeneity, which can be traced to specific progenitor cells [70].

Bulk transcriptomics cannot distinguish expression profiles in unique cell populations; nevertheless, computational approaches of deconvolution, which use expression datasets from known cell types as reference, allow for an estimate of cell type-specific gene expression from bulk RNA-seq data [71]. Performing deconvolution assessment of bulk human SAT transcriptome datasets, Glastonbury et al. characterized subcutaneous adipose tissue heterogeneity by estimating the proportions of adipocytes, macrophages, CD4+ T cells, and microvascular endothelial cells [72]. Moreover, a bulk RNA-seq deconvoluting algorithm that relies on single-nucleus RNA sequencing (snRNA-seq) data has been recently developed to estimate the composition of over 13 distinct cell types in human SAT and VAT [73]. Furthermore, by gene expression deconvolution of publicly available bulk transcriptomics data, Lenz et al. [74] estimated that the median fraction of adipocytes in different adipose tissue depots is within the range of 55–80%. Interestingly, the average percentage of ASCs is approximately 15% in SAT, making adipose tissue the richest source of multipotent stromal cells in the body, even if a large variability (from 3% to over 30%) has been observed across individuals. Recently, on the bases of deconvoluted bulk RNA-seq data, Li et al. compared the SVF cellular composition between Coleman fat and ECM/SVF gel obtained by mechanical processing, showing that the proportion of the stromal cells was not modified by the procedure, while a significant loss of M2 macrophages was observed in the ECM/SVF-gel [75].

Integrative co-expression analysis of publicly available adipose tissue bulk RNA-seq data was used to implementing a human adipose tissue cell-type transcriptome atlas and define the expression profile of genes differentially expressed between SAT and VAT [76]. Collectively, these studies highlighted depot-specific gene expression profiles and were instrumental in the identification of putative innovative markers suitable for detecting distinct cell populations. In particular, adipose progenitor cells have been shown to specifically express higher levels of mannose receptor C type 2 (MRC2, aliases ENDO180, UPARAP, CD280), which plays a role in extracellular matrix remodeling, and cadherin 11 (CDH11), which is involved in calcium-dependent cell–cell adhesion (Fig. 2). Similarly, the immunoglobulin superfamily member CD26 (alias B7 Homolog 3, B7-H3) is one of the 11 genes with the highest predicted specificity for mesenchymal cells within VAT [76].

Fig. 2
figure 2

Adipose tissue cell type enriched transcriptome. Genes with predicted adipose progenitor cell specificity within subcutaneous (top panels) or visceral adipose tissue (bottom panels). MRC2: mannose receptor C type 2; CDH11: cadherin 11; (image credit: courtesy of Human Protein Atlas, www.proteinatlas.org, accessed: 05 September 2024 [136]. Image available at the following URL: v23.proteinatlas.org/humancell [76, 137]

Single-cell RNA-sequencing and single-nucleus RNA sequencing of adipose tissue cells

Over the past few years, the progression of molecular biology techniques with improved resolution, such as single-cell real-time quantitative PCR (RT-qPCR), digital droplet PCR (ddPCR) and, particularly, single-cell RNA sequencing (scRNA-seq), has provided the possibility of dissecting adipose tissue heterogeneity through the transcriptional evaluation of individual cells. While PCR-based techniques are limited to the study of a relatively small number of genes, high-throughput scRNA-seq offers a holistic view of the adipose tissue transcriptome, capturing the entire spectrum of cell states and enabling the identification of novel markers and cellular populations. In virtue of its sensitivity, scRNA-seq can also investigate low-copy mRNAs. Single-cell suspensions are needed for scRNA-seq analysis; thus, after extensive adipose tissue disaggregation or cultured cell trypsinization, each cell is isolated by different methods, including fluorescence-activated cell sorting, magnetic-activated cell sorting, and microfluidic systems, and the RNA is amplified and sequenced from each cell separately.

Single-cell transcriptome investigation provides a snapshot of the gene expression profile of an individual cell; nonetheless, by sequencing several cells at a given time point, it is possible to infer the transcriptome dynamics of biological processes such as cell differentiation, development, and disease progression. In particular, assuming a continuous spectrum of transcriptional changes between more or less differentiated conditions during cell differentiation, computational evaluation of RNA-seq data makes it possible to associate distinctive cells, characterized by their representative transcriptome, with different states in a pseudo-temporal trajectory of cell differentiation, generating a presumed cell lineage tree. This approach has been successfully used to establish a lineage hierarchy of several distinct subtypes of ASC [77]. Conversely, it is difficult to define the precise trajectory of adipogenesis because adipocytes have a slow turnover rate [78] and a single sample cannot always comprise the complete transcriptional changes associated with all the differentiation steps [79].

To obtain a comprehensive view of adipose tissue cellular and temporal heterogeneity, a relatively large number of individual cells must be isolated, processed and the RNA sequenced. Single-cell isolation of adipocytes for scRNA-seq studies is challenging because of their buoyancy and fragility, which are attributable to their high lipid content and large size. To circumvent this problem, in vitro differentiation studies on preadipocytes have been performed [80], but this approach cannot precisely recapitulate all physiological differentiation cues. Therefore, although scRNA-seq approaches have been extensively used to study the adipose tissue stromal-vascular cell fraction, scRNA-seq data for adipocytes are limited.

Single-nucleus RNA sequencing (snRNA-seq), which works on nuclei rather than intact cells, is an alternative approach to scRNA-seq. The snRNA-seq procedure does not require tissue enzymatic digestion, which can lead to critical loss of some cell types; therefore, it is suitable for the analysis of the transcriptome of all cell populations within an adipose tissue sample, including fragile adipocytes and adipogenic precursors [81]. Moreover, nuclei can be isolated from both fresh and fixed or frozen archived tissues. Although scRNA-seq enables the analysis of both nuclear and cytoplasmic transcripts, whereas snRNA-seq mostly measures nuclear transcripts, both approaches yield similar expression profiles of relevant genes in adipose tissue samples [82, 83].

Several studies, preferentially using scRNA-seq for characterizing human stromal vascular fraction cells, and snRNA-seq to characterize mature adipocytes, have been performed, as reported in Table 3. We mainly focused on studies performed on human-derived samples because, due to substantial intra-species differences in adipose tissue structure, function and metabolism [16], cell populations identified in murine models do not exactly map those in humans [25]. Nonetheless, some cell populations identified by scRNA-seq in mice were confirmed in humans; for instance, the detection of adipogenesis-regulatory cells refractory to adipogenesis, named Areg, using scRNA-seq on murine sWAT led to the identification of a functionally conserved CD142+, SCA1+ cell population in humans [84]; the dipeptidyl peptidase-4 expressing (DPP4+) mesenchymal cell population, which in virtue of its highly proliferative potential may be used for regenerative medicine applications, was identified in both murine and human adipose tissue [77].

Table 3 Transcriptomics studies of human white adipose tissue

Some scRNA-seq studies have been performed on cultured cells, whereas others on freshly isolated uncultured cells. Large-scale scRNA-seq of cultured human ASCs from three donors demonstrated that over 96% of cells adhere to the IFATS and ISCT recommendations for ASC marker expression. Nonetheless, even in an almost pure cell population, gene expression levels may still be heterogeneous, mainly because of cell cycle influence [85]. In addition, in primary, FACS-sorted, human preadipocytes acquired from a commercial source and subjected to adipogenic culture conditions, at least two distinct cell populations can be identified by scRNA-seq analysis [80]. Palani et al. recently isolated human adipocyte progenitors from subcutaneous and visceral white adipose tissue (WAT), which were then assessed by scRNA-seq at different timepoints during in vitro differentiation. The authors delineated human adipocyte single-cell trajectory (available at https://cphbat.shinyapps.io/adipodiff/. Accessed: 05 September 2024) showing that when cultured in adipogenic differentiating medium, WAT cells separate from a common progenitor into two main cell fates, an adipogenic and a structural branch, defined on the basis of an extracellular matrix and developmental gene signature [86].

Single-cell transcriptomic sequencing assessment of ASCs isolated from one healthy donor performed after 3 passages of culture revealed 9 cell clusters; moreover, the authors, using a computational approach, demonstrated that the ASC cluster with the maximum stemness was characterized by the highest expression of genes involved in cytoskeleton structure and regulation of cell cycle transition, such as tropomyosin 1 (TPM1) and cyclin D1 (CCND1) [87].

The first scRNA-seq study performed on uncultured cells from human sWAT of 4 healthy women suggested that ASCs constitute a single and homogeneous population [88]. In 2020, Vijay et al. used single-cell RNA-seq to analyze the non-adipocyte fraction of adipose samples from 14 obese donors. This study assessed that in the stromal vascular fraction of human WAT, the average proportion of adipocyte progenitors and stem cells is approximately 55%, divided into several subpopulations with different commitment but all CD45, CD34+, CD31 [89]. In particular, within sWAT, pre-adipocyte/adipose stem cells express matrix Gla protein (MGP), apolipoprotein D (APOD), C-X-C Motif Chemokine Ligand 14 (CXCL14), WNT1-inducible signaling pathway protein 2 (WISP2, alias CCN5) and complement factor D (CDF), whereas the cluster of more mature adipocyte progenitor cells express apolipoprotein E (APOE), fatty acid binding protein 4 (FABP4), C/EBP β, CD36 antigen (collagen type I receptor, thrombospondin receptor) and share CDF expression; an additional stromal cell cluster, mainly encompassing fibroblasts, is characterized by ECM remodeling and fibrosis markers such as diverse types of collagens (COL3A1, COL6A3, COL1A1 and COL6A1). The study also highlighted the presence of 37% immune cells and 8% endothelial cells, including microvascular and lymphatic endothelial cells [89]. In line with these results, high PR domain zinc finger protein 16 (PRDM16) expression has been identified as a possible marker for an endothelial progenitor cell population that is enriched in sWAT compared with vWAT, in agreement with an increased sWAT angiogenic potential [90].

Hildreth et al. performed scRNA-seq analysis on CD45+-sorted uncultured SVF isolated from abdominal sWAT from lean and obese patients, thereby identifying 28 distinct structural and immune cell populations [91]. In particular, according to this study, WAT contains a wide array of immune cells, including neutrophils, monocytes, macrophages, different classes of T cells (CD4 + T cells, naive CD8 + T cells, cytotoxic CD8 + T cells, CD8 + γδ T cells, regulatory T cells, mucosa-associated invariant T cells), B lymphocytes, mature NK cells, myeloid-like cells, and NK-like cells. Interestingly, while lean WAT contains mainly noninflammatory cells, in obese WAT the immune profile shifts to a proinflammatory state [92]. Recently, Gu et al. analyzed the cell subpopulations in WAT of different anatomical origins using scRNA-seq data from public databases and one de novo dataset from subcutaneous leg adipose tissues and found that sWAT of the legs is rich in ASCs [93]. According to single-cell RNA seq analysis of SVF cells isolated from lipoaspirate obtained from 2 healthy donors, Whytock et al. estimated that one stem cell cluster (PTPRC/PECAM1/CD34+/PDGFRA+/PDGFRB+) represents approximately 28% of the total SVF and 7% of the total sWAT cells [82].

Emont et al. generated a comprehensive atlas of human subcutaneous and visceral WAT on the basis of scRNA and snRNA-seq data [94]. All the main cell populations were represented, to a different extent, in both sWAT and vWAT, excluding mesothelial and endometrial cell clusters present only in visceral fat (Fig. 3). In particular, human white adipocytes cluster into six subpopulations, whereas adipose stem and progenitor cells cluster into seven subpopulations that express the common marker gene platelet-derived growth factor receptor alpha (PDGFRA). Interestingly, no significant difference was observed among the overall proportions of adipocytes and adipose stem and progenitor cells between vWAT and sWAT. Based on this study [94], a reference dataset for the analysis and interpretation of single-cell RNA-seq experiments has been developed and is available on Azimuth (https://app.azimuth.hubmapconsortium.org/app/human-adipose, accessed: 05 September 2024) [95].

Fig. 3
figure 3

A single cell atlas of adipose tissue. Uniform manifold approximation and projection maps (UMAP) embedding coordinates for human subcutaneous and visceral white adipose tissue (sWAT and vWAT). Image credit: courtesy of Single Cell Portal, www.singlecell.broadinstitute.org/single_cell [138]. The study is available at the following URL: https://singlecell.broadinstitute.org/single_cell/study/SCP1376, accessed: 05 September 2024 [94]

More recently, Massier et al. generated a high-resolution map of human subcutaneous, omental, and perivascular WAT by analyzing publicly available and newly generated scRNA-seq, snRNA-seq, and spatial transcriptomic data integrated with deconvoluted bulk transcriptomic datasets [96]. This study, which included over 400,000 quality-filtered cells/nuclei obtained from 83 donors, identified more than 60 cell subpopulations, providing a comprehensive overview of the cells residing in human WAT. Remarkably, most of the cell subpopulations were present in all analyzed WAT depots (subcutaneous, omental, and perivascular), as previously observed [94]. Stromal cells, including fibroblast and adipogenic progenitors at different stages of commitment, constituted approximately 40% of the total cell populations, adipocytes were ~ 20%, immune cells ~ 20% and vascular cells less than 15%. Recently, a reference atlas of white adipose tissue including 43 cell subtypes was generated by integrating scRNA-seq and snRNA-seq datasets from 6 independent studies, further supported by deconvoluted bulk RNA-seq data [97]. This study established that, compared with vWAT, sWAT is enriched in vascular endothelial cells, fibro-adipogenic progenitors and T cells.

Single-nucleus RNA sequencing has also been instrumental in defining cell-type specific composition of tumor-adjacent and distal adipose tissue in patients with breast cancer, highlighting the increased expression of inflammatory genes in tumor-associated adipocytes and macrophages [98].

Adipose tissue spatial transcriptomics

The behavior of adipose tissue mesenchymal stromal cells depends on the local tissue environment, or “niche”, that is characterized by a unique combination of local gradients of signaling factors, nutrients and oxygen, neighboring cell cross-talk, and ECM-derived biomechanical forces. Single-cell omics studies contribute to the accurate definition of gene expression within each cell but cannot capture the cell location information that is lost during tissue disaggregation required for sample processing. Spatial transcriptomics combines RNA sequencing and immunohistological analysis of intact tissue sections, mapping transcriptional data to their original spatial location within the tissue, ideally at single-cell resolution. Thus, spatial transcriptomics allows the investigation of tissue spatial heterogeneity and provides insights into cell–cell interactions and tissue functional organization.

Three distinct adipocyte and two adipocyte progenitor subpopulations were identified in human WAT; interestingly, spatial transcriptomics analysis confirmed that collagen-rich adipocyte progenitors are frequently positioned over fibrous and vascular structures and close to M2-like macrophages [99], organizing adipogenic niches [100]. More recently, Massier et al. described an integrated single cell and spatial transcriptomic map of human subcutaneous, omental, and perivascular WAT [96]. In particular, the authors identified in scWAT 17 clusters of fibroblasts and adipogenic stem/progenitor cells at different stages of commitment (FAPs) with specific tissue distributions, commonly near to vessels or macrophages, forming distinct putative regenerative niches.

Adipose tissue proteomics and secretomics

Transcriptomic data cannot precisely predict cellular protein content because multiple regulatory processes in transcription, translation, post-translational modification, mRNA and protein degradation affect protein production and stability. Characterization of adipose tissue, ASCs and their secretome using high-throughput mass spectrometry-based proteomics has been performed, as previously reviewed [32, 101]. Some studies have analyzed fully digested protein preparations from adipose tissue and have demonstrated that the levels of relevant metabolic pathways, including carbohydrate and lipid metabolism, protein synthesis, and inflammation, are increased in VAT compared with SAT, in agreement with an higher cardiovascular risk associated with visceral adiposity [102, 103]. Other studies have investigated the proteome in isolated cellular fractions [104] and have shown that mature adipocytes and SVF cells share a common proteomic profile [105]. In particular, adipokines are produced both by adipocytes and SVF cells, whereas inflammatory mediators, such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), are mainly produced by SVF cells [106]. Moreover, proteomic analysis highlighted significant differences between uncultured adipocytes isolated from different obese donor-matched adipose tissue depots; in particular, the vWAT adipocyte proteome has an increased content of proteins involved in glycolysis, lipogenesis, and mitochondrial function compared with sWAT [107]. Approximately 22% of the total number of proteins in adipocytes have mitochondrial origin, compared with 4.8% in the whole human proteome [108], confirming the important role of mitochondria in key pathways such as lipid and glucose homeostasis and oxidative capacity [109]. More recently, comparison between sWAT and vWAT proteomes from lean and obese individuals and patients with type 2 diabetes revealed specific down-regulation of mitochondrial and up-regulation of ECM and immune response-related pathways associated with type 2 diabetes [110].

Adaptation to cell culture involves proteome changes, as assessed during amplification under good manufacturing practices of ASCs that otherwise maintain a homogeneous immunophenotypic profile up to seven passages [111]. Dynamic proteomic changes in ASCs undergoing adipocyte differentiation indicate modulation of Wnt and transforming growth factor-β (TGF-β) signaling pathways [112], whereas osteogenic differentiation is associated with increased expression of integrins and genes involved in ECM organization [113].

Adipose tissue releases hundreds of bioactive molecules, commonly referred to as adipokines, that contribute to the regulation of whole-body energy metabolism and mediate ASCs regenerative and immunomodulatory potential. The adipose tissue secretome comprises a wide array of proteins, peptides, metabolites, lipids (lipokines), and extracellular vesicle cargoes, including non-coding RNAs [114, 115].

The use of ASC conditioned medium, EVs and decellularized lipoaspirate has gained attention as a potential cell-free alternative to cell therapy [116, 117]; before clinical translation, precise assessment of adipose tissue/ASC secretome is mandatory and standardization is required [118]. Transcript levels do not correlate with secreted protein levels because proteins may be stored in secretory vesicles and released upon stimulation; therefore, adipose tissue transcriptomics should be integrated with proteomic studies of secreted proteins (secretomics). The secretome collected from in vitro cell cultures only partially reflects the in vivo complexity, whereas the adipose tissue secretome contains proteins released by both adipocytes and SVF cells. Adipokines released by vWAT explants include adiponectin, interleukin-6, and proteins involved in ECM organization [119]. Proteomic evaluation of extracellular vesicles produced in vitro by human primary adipocytes identified over 880 proteins, among which fatty acid binding protein 4 (FABP4) has been proposed as a tissue-specific marker [120].

Cellular protein content is highly dynamic; although the average protein copy number per cell is higher than the corresponding number of RNA copies, proteins cannot be amplified like nucleic acids, making the assessment of the proteome in individual cells difficult. Nonetheless, recent technical developments have rapidly moved the field toward single-cell proteomics through liquid chromatography mass spectrometry and next-generation sequencing, single-cell spatial proteomics [121], and time-resolved assessment of protein secretion from single cells by sequencing (TRAPS-seq) [122]. Therefore, single-cell-based proteomic studies of human adipose tissue will soon contribute in deciphering adipose tissue heterogeneity.

Adipose tissue lipidomics

Adipocytes exhibit highly dynamic lipid metabolism that involves the production, storage, and release of bioactive lipids (lipokines) that modulate immunological function and tissue homeostasis. In 2021, Lange et al. [123] presented a mass spectrometry-based atlas of the lipidome of human adipose tissue that captures the lipid diversity of over 1600 lipid molecular species, mainly triacylglycerols, whose physiological contributions remain largely unexplored. Lipid composition is depot-specific and heterogeneous [124]; for instance, differences in plasmalogen content in sWAT and vWAT affect cell membrane fluidity and, in turn, adipocyte remodeling and pathological fat mass expansion [123]. Despite the progress of mass spectrometry methods, the identification of lipids at the single-cell level remains difficult and, to the best of our knowledge, analysis of human adipose tissue has not been performed yet [125].

Adipose tissue matrisomics

Adipose tissue extracellular matrix (ECM) is a complex meshwork mainly composed of collagens, fibronectin, laminin and glycosaminoglycans. ECM organizes tissue 3D architecture, supports tissue growth and remodeling in response to nutritional status, and provides biomechanical, biochemical, and biophysical cues to coordinate cell proliferation, migration and differentiation during tissue repair. Decellularized adipose tissue/ASC ECM-derived products have reduced immunogenicity and may be employed for the fabrication of biocompatible scaffolds for regenerative medicine using innovative three-dimensional bioprinting techniques [126, 127]. To obtain a decellularized matrix, cellular, genetic, and lipidic components of adipose tissue are removed by enzymatic, physical, or chemical procedures, while the native 3D configuration of the ECM is preserved [128]. No consensus has been reached regarding the best procedure for obtaining an adipose tissue-derived material that preserves the native microenvironment to support in situ tissue regeneration. Standardization of the procedures and characterization of the adipose tissue matrisome are essential to develop a decellularized ECM suitable for translational procedures [129]. In particular, significant matrisome differences were detected between suction and power-assisted lipoaspirates. The most abundant proteins in the adipose tissue matrisome are structural collagens and heparan sulphate proteoglycan, whereas the content of some proteins, such as fibronectin and vitronectin, varies according to donor age and depot location [129].

Clinical use of off-the-shelf decellularized adipose-derived products in regenerative medicine procedures is ongoing; precise definition of the ECM components, their biomechanical proprieties, involvement in tissue repair, degradation time, immune profile, and standardization of product development will further promote clinical translation [130].

Perspectives and conclusions

In recent years, omics-based approaches at single-cell and spatial resolutions have been applied to dissect human adipose tissue heterogeneity and provide insights into ASC differentiation trajectories [11]. Technologies are being further refined to increase the throughput requirements needed to characterize rare subpopulations, reduce costs, and minimize sample bias. Currently, several methods are under investigation to reduce batch effects by integrating scRNA-seq and snRNA-seq datasets acquired with different technologies and annotations [97]. Moreover, computational advances, artificial intelligence integration, and improved spatial transcriptome deconvolution algorithms will allow the extension of data analysis of spatial single-cell heterogeneity. For instance, spatial transcriptomic and proteomic data across several tissue slices can be analyzed utilizing deep learning approaches to generate multiomic 3D maps. The next frontier is currently represented by the integration of simultaneous multimodal spatial single-cell omics approaches [9] and the development of technologies with improved resolution for subcellular omics [131] to elucidate the key role of mitochondria in human WAT function [109]. Combined simultaneous application of different omics technologies on human adipose tissue is increasing. Of particular relevance, are studies that associate transcriptome/genome and methylome analyses to identify possible epigenetic differences in different adipose tissue depots under pathological condition [69, 132].

However, several critical points need to be further addressed: in general, single-cell analysis describes a snapshot of the cellular gene expression profile, whereas the adipose tissue cellular landscape is highly dynamic in response to different stimuli. Therefore, adipose tissue heterogenicity may arise, at least in part, by cell transition into different functional states, which require extensive studies to be fully captured. In addition, the tissue microenvironment clearly plays a critical role in regulating the ASC state; therefore, 3D information should be preserved for understanding possible cell–cell interactions in the context of tissue structural organization.

Notwithstanding these limitations, single-cell omics studies have provided formal evidence of the extensive cellular heterogeneity of adipose tissue (Table 3) encompassing over 60 different subpopulations of adipocytes, fibroblast (stromal) and adipogenic progenitors, vascular, and immune cells [96]. Single-nucleus RNA sequencing studies confirmed observations from morphological studies, indicating that, despite occupying most of the tissue volume, adipocytes represent only approximately 20% of the total WAT cell number. Different mature adipocyte subclusters have been identified in each depot, possibly associated with heterogeneous functional differences [133]. Conversely, according to both snRNA-seq and scRNA-seq data, fibroblast and adipogenic precursors numerically represent the largest class of WAT cell populations. Overall, all cell subpopulations were present in both sWAT and vWAT, but some depot-specific differences were observed. In particular, vWAT is enriched with mesothelial cells and macrophages, whereas sWAT is enriched with vascular, fibro-adipogenic progenitors, and endothelial cells [97], suggesting that WAT is a preferable source of progenitor cells for regenerative applications. Moreover, sWAT spatial transcriptomic data indicate that fibro-adipogenic progenitors are located in proximity to vessels, in accordance with the previously established association of mesenchymal cells with perivascular niches [134].

In conclusion, recent studies using single‐cell multiomic approaches have significantly advanced our understanding of the cellular heterogeneity within the human white adipose tissue stromal vascular fraction. Even if the assessment of the clinical relevance of the identified cell types needs to be fully established, these studies may lead to the potential selection of unique subpopulations with optimized regenerative potentials, paving the way to more reproducible and effective clinical applications using adipose tissue derivatives.

Availability of data and materials

Not applicable.

Abbreviations

ASC:

Adipose tissue-derived stromal cell

ECM:

Extracellular matrix

EV:

Extracellular vesicle

FACS:

Fluorescence-activated cell sorting

MSC:

Mesenchymal stromal cell

PPAR-γ:

Peroxisome proliferator-activated receptor-gamma

scRNA-seq:

Single-cell RNA sequencing

snRNA-seq:

Single-nucleus RNA sequencing

SAT or sWAT:

Subcutaneous (white) adipose tissue

SVF:

Stromal vascular fraction

VAT or vWAT:

Visceral (white) adipose tissue

WAT:

White adipose tissue

References

  1. Dong L, Li X, Leng W, Guo Z, Cai T, Ji X, et al. Adipose stem cells in tissue regeneration and repair: From bench to bedside. Regen Ther. 2023;24:547–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Trivisonno A, Abecassis M, Monti M, Toietta G, Bachir A. Adipose tissue: from energy reservoir to a source of cells for epithelial tissue engineering. In: Shiffman MA, Di Giuseppe A, Bassetto F, editors. Stem cells in aesthetic procedures: art, science, and clinical techniques. Berlin: Springer; 2014. p. 303–26.

    Chapter  Google Scholar 

  3. Qin Y, Ge G, Yang P, Wang L, Qiao Y, Pan G, et al. An update on adipose-derived stem cells for regenerative medicine: where challenge meets opportunity. Adv Sci (Weinh). 2023;10: e2207334.

    Article  PubMed  Google Scholar 

  4. Trivisonno A, Alexander RW, Baldari S, Cohen SR, Di Rocco G, Gentile P, et al. Intraoperative strategies for minimal manipulation of autologous adipose tissue for cell- and tissue-based therapies: concise review. Stem Cells Transl Med. 2019;8:1265–71.

    Article  PubMed  PubMed Central  Google Scholar 

  5. He C, Dai M, Zhou X, Long J, Tian W, Yu M. Comparison of two cell-free therapeutics derived from adipose tissue: small extracellular vesicles versus conditioned medium. Stem Cell Res Ther. 2022;13:86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and strategies for improving the regenerative effects of mesenchymal stromal cell-based therapies. Int J Mol Sci. 2017;18:2087.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Baldari S, Di Rocco G, Trivisonno A, Samengo D, Pani G, Toietta G. Promotion of survival and engraftment of transplanted adipose tissue-derived stromal and vascular cells by overexpression of manganese superoxide dismutase. Int J Mol Sci. 2016;17:1082.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Gui C, Parson J, Meyer GA. Harnessing adipose stem cell diversity in regenerative medicine. APL Bioeng. 2021;5: 021501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Baysoy A, Bai Z, Satija R, Fan R. The technological landscape and applications of single-cell multi-omics. Nat Rev Mol Cell Biol. 2023;24:695–713.

    Article  CAS  PubMed  Google Scholar 

  10. Avtanski D, Hadzi-Petrushev N, Josifovska S, Mladenov M, Reddy V. Emerging technologies in adipose tissue research. Adipocyte. 2023;12:2248673.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Maniyadath B, Zhang Q, Gupta RK, Mandrup S. Adipose tissue at single-cell resolution. Cell Metab. 2023;35:386–413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Duerre DJ, Galmozzi A. Deconstructing adipose tissue heterogeneity one cell at a time. Front Endocrinol (Lausanne). 2022;13: 847291.

    Article  PubMed  Google Scholar 

  13. Li H, Wang Y, Zhu G, Ma Q, Huang S, Guo G, et al. Application progress of single-cell sequencing technology in mesenchymal stem cells research. Front Cell Dev Biol. 2024;11:1336482.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Kostecka A, Kalamon N, Skoniecka A, Koczkowska M, Skowron PM, Piotrowski A, et al. Adipose-derived mesenchymal stromal cells in clinical trials: insights from single-cell studies. Life Sci. 2024;351: 122761.

    Article  CAS  PubMed  Google Scholar 

  15. Corvera S. Cellular heterogeneity in adipose tissues. Annu Rev Physiol. 2021;83:257–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Börgeson E, Boucher J, Hagberg CE. Of mice and men: pinpointing species differences in adipose tissue biology. Front Cell Dev Biol. 2022;10:1003118.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Ford H, Liu Q, Fu X, Strieder-Barboza C. White adipose tissue heterogeneity in the single-cell era: from mice and humans to cattle. Biology (Basel). 2023;12:1289.

    CAS  PubMed  Google Scholar 

  18. Liu P, Li D, Zhang J, He M, Li Y, Liu R, et al. Transcriptomic and lipidomic profiling of subcutaneous and visceral adipose tissues in 15 vertebrates. Sci Data. 2023;10:453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Auger C, Kajimura S. Adipose tissue remodeling in pathophysiology. Annu Rev Pathol. 2023;18:71–93.

    Article  CAS  PubMed  Google Scholar 

  20. Fang R, Yan L, Liao Z. Abnormal lipid metabolism in cancer-associated cachexia and potential therapy strategy. Front Oncol. 2023;13:1123567.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lengyel E, Makowski L, DiGiovanni J, Kolonin MG. Cancer as a matter of fat: the crosstalk between adipose tissue and tumors. Trends Cancer. 2018;4:374–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yao H, He S. Multi-faceted role of cancer-associated adipocytes in the tumor microenvironment (Review). Mol Med Rep. 2021;24:866.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sato S. Adipo-oncology: adipocyte-derived factors govern engraftment, survival, and progression of metastatic cancers. Cell Commun Signal. 2024;22:52.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011;71:2455–65.

    Article  CAS  PubMed  Google Scholar 

  25. Liu SQ, Chen DY, Li B, Gao ZJ, Feng HF, Yu X, et al. Single-cell analysis of white adipose tissue reveals the tumor-promoting adipocyte subtypes. J Transl Med. 2023;21:470.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhu K, Cai L, Cui C, de Los Toyos JR, Anastassiou D. Single-cell analysis reveals the pan-cancer invasiveness-associated transition of adipose-derived stromal cells into COL11A1-expressing cancer-associated fibroblasts. PLoS Comput Biol. 2021;17: e1009228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bouche C, Quail DF. Fueling the tumor microenvironment with cancer-associated adipocytes. Cancer Res. 2023;83:1170–2.

    Article  CAS  PubMed  Google Scholar 

  28. Ibrahim MM. Subcutaneous and visceral adipose tissue: structural and functional differences. Obes Rev. 2010;11:11–8.

    Article  PubMed  Google Scholar 

  29. Baglioni S, Francalanci M, Squecco R, Lombardi A, Cantini G, Angeli R, et al. Characterization of human adult stem-cell populations isolated from visceral and subcutaneous adipose tissue. FASEB J. 2009;23:3494–505.

    Article  CAS  PubMed  Google Scholar 

  30. Hocking SL, Wu LE, Guilhaus M, Chisholm DJ, James DE. Intrinsic depot-specific differences in the secretome of adipose tissue, preadipocytes, and adipose tissue-derived microvascular endothelial cells. Diabetes. 2010;59:3008–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mariani S, Di Rocco G, Toietta G, Russo MA, Petrangeli E, Salvatori L. Sirtuins 1–7 expression in human adipose-derived stem cells from subcutaneous and visceral fat depots: influence of obesity and hypoxia. Endocrine. 2017;57:455–63.

    Article  CAS  PubMed  Google Scholar 

  32. Peinado JR, Pardo M, de la Rosa O, Malagón MM. Proteomic characterization of adipose tissue constituents, a necessary step for understanding adipose tissue complexity. Proteomics. 2012;12:607–20.

    Article  CAS  PubMed  Google Scholar 

  33. Gimble J, Guilak F. Adipose-derived adult stem cells: isolation, characterization, and differentiation potential. Cytotherapy. 2003;5:362–9.

    Article  PubMed  Google Scholar 

  34. Trivisonno A, Di Rocco G, Cannistra C, Finocchi V, Torres Farr S, Monti M, et al. Harvest of superficial layers of fat with a microcannula and isolation of adipose tissue-derived stromal and vascular cells. Aesthet Surg J. 2014;34:601–13.

    Article  PubMed  Google Scholar 

  35. Jurgens WJFM, Oedayrajsingh-Varma MJ, Helder MN, Zandiehdoulabi B, Schouten TE, Kuik DJ, et al. Effect of tissue-harvesting site on yield of stem cells derived from adipose tissue: implications for cell-based therapies. Cell Tissue Res. 2008;332:415–26.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Riis S, Zachar V, Boucher S, Vemuri MC, Pennisi CP, Fink T. Critical steps in the isolation and expansion of adipose-derived stem cells for translational therapy. Expert Rev Mol Med. 2015;17: e11.

    Article  CAS  PubMed  Google Scholar 

  37. Brooks AES, Iminitoff M, Williams E, Damani T, Jackson-Patel V, Fan V, et al. Ex vivo human adipose tissue derived mesenchymal stromal cells (ASC) are a heterogeneous population that demonstrate rapid culture-induced changes. Front Pharmacol. 2020;10:1695.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Greenwood V, Clausen P, Matuska AM. Micro-fragmented adipose tissue cellular composition varies by processing device and analytical method. Sci Rep. 2022;12:16107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dewal RS, Wolfrum C. Master of disguise: deconvoluting adipose tissue heterogeneity and its impact on metabolic health. Curr Opin Genet Dev. 2023;81: 102085.

    Article  CAS  PubMed  Google Scholar 

  40. Berry R, Church CD, Gericke MT, Jeffery E, Colman L, Rodeheffer MS. Imaging of adipose tissue. Meth Enzymol. 2014;537:47–73.

    Article  CAS  Google Scholar 

  41. Galarraga M, Campión J, Muñoz-Barrutia A, Boqué N, Moreno H, Martínez JA, et al. Adiposoft: automated software for the analysis of white adipose tissue cellularity in histological sections. J Lipid Res. 2012;53:2791–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Nicu C, Pople J, Bonsell L, Bhogal R, Ansell DM, Paus R. A guide to studying human dermal adipocytes in situ. Exp Dermatol. 2018;27:589–602.

    Article  CAS  PubMed  Google Scholar 

  43. Sbarbati A, Accorsi D, Benati D, Marchetti L, Orsini G, Rigotti G, et al. Subcutaneous adipose tissue classification. Eur J Histochem. 2010;54: e48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jiang Y, Yeung JLH, Lee JH, An J, Steadman PE, Kim JR, et al. Visualization of 3D white adipose tissue structure using whole-mount staining. J Vis Exp. 2018;141:58683.

    Google Scholar 

  45. Schopow N, Kallendrusch S, Gong S, Rapp F, Körfer J, Gericke M, et al. Examination of ex-vivo viability of human adipose tissue slice culture. PLoS ONE. 2020;15: e0233152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Berry R, Rodeheffer MS. Characterization of the adipocyte cellular lineage in vivo. Nat Cell Biol. 2013;15:302–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bourin P, Bunnell BA, Casteilla L, Dominici M, Katz AJ, March KL, et al. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International federation for adipose therapeutics and science (IFATS) and the international society for cellular therapy (ISCT). Cytotherapy. 2013;15:641–8.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Di Rocco G, Iachininoto MG, Tritarelli A, Straino S, Zacheo A, Germani A, et al. Myogenic potential of adipose-tissue-derived cells. J Cell Sci. 2006;119(Pt 14):2945–52.

    Article  PubMed  Google Scholar 

  49. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Planat-Benard V, Silvestre J-S, Cousin B, André M, Nibbelink M, Tamarat R, et al. Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation. 2004;109:656–63.

    Article  PubMed  Google Scholar 

  51. Di Rocco G, Gentile A, Antonini A, Truffa S, Piaggio G, Capogrossi MC, et al. Analysis of biodistribution and engraftment into the liver of genetically modified mesenchymal stromal cells derived from adipose tissue. Cell Transpl. 2012;21:1997–2008.

    Article  Google Scholar 

  52. Jang S, Cho H-H, Cho Y-B, Park J-S, Jeong H-S. Functional neural differentiation of human adipose tissue-derived stem cells using bFGF and forskolin. BMC Cell Biol. 2010;11:25.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Robert AW, Marcon BH, Dallagiovanna B, Shigunov P. Adipogenesis, osteogenesis, and chondrogenesis of human mesenchymal stem/stromal cells: a comparative transcriptome approach. Front Cell Dev Biol. 2020;8:561.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Brooks PT, Munthe-Fog L, Rieneck K, Banch Clausen F, Rivera OB, Kannik Haastrup E, et al. Application of a deep learning-based image analysis and live-cell imaging system for quantifying adipogenic differentiation kinetics of adipose-derived stem/stromal cells. Adipocyte. 2021;10:621–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Lee M-J, Fried SK. Optimal protocol for the differentiation and metabolic analysis of human adipose stromal cells. Meth Enzymol. 2014;538:49–65.

    Article  CAS  Google Scholar 

  56. Stromps J-P, Paul NE, Rath B, Nourbakhsh M, Bernhagen J, Pallua N. Chondrogenic differentiation of human adipose-derived stem cells: a new path in articular cartilage defect management? Biomed Res Int. 2014;2014: 740926.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–28.

    Article  CAS  PubMed  Google Scholar 

  58. Halvorsen YD, Franklin D, Bond AL, Hitt DC, Auchter C, Boskey AL, et al. Extracellular matrix mineralization and osteoblast gene expression by human adipose tissue-derived stromal cells. Tissue Eng. 2001;7:729–41.

    Article  CAS  PubMed  Google Scholar 

  59. Ong WK, Chakraborty S, Sugii S. Adipose tissue: understanding the heterogeneity of stem cells for regenerative medicine. Biomolecules. 2021;11:918.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Feng X, Qi F, Wang H, Li W, Gan Y, Qi C, et al. sorting technology for mesenchymal stem cells from a single tissue source. Stem Cell Rev and Rep. 2024;20:524–37.

    Article  PubMed  Google Scholar 

  61. Hearnden R, Sandhar B, Vyas V, Longhi MP. Isolation of stromal vascular fraction cell suspensions from mouse and human adipose tissues for downstream applications. STAR Prot. 2021;2: 100422.

    Article  CAS  Google Scholar 

  62. Majka SM, Miller HL, Helm KM, Acosta AS, Childs CR, Kong R, et al. Analysis and isolation of adipocytes by flow cytometry. Meth Enzymol. 2014;537:281–96.

    Article  CAS  Google Scholar 

  63. Delaney KZ, Dam V, Murphy J, Morais JA, Denis R, Atlas H, et al. A reliable, reproducible flow cytometry protocol for immune cell quantification in human adipose tissue. Anal Biochem. 2021;613: 113951.

    Article  CAS  PubMed  Google Scholar 

  64. Lee LM, Klarmann GJ, Haithcock DW, Wang Y, Bhatt KH, Prabhakarpandian B, et al. Label-free enrichment of human adipose-derived stem cells using a continuous microfluidic sorting cascade. Lab Chip. 2023;23:2131–40.

    Article  CAS  PubMed  Google Scholar 

  65. Dai X, Shen L. Advances and trends in omics technology development. Front Med (Lausanne). 2022;9: 911861.

    Article  PubMed  Google Scholar 

  66. Vohl M-C, Sladek R, Robitaille J, Gurd S, Marceau P, Richard D, et al. A survey of genes differentially expressed in subcutaneous and visceral adipose tissue in men. Obes Res. 2004;12:1217–22.

    Article  CAS  PubMed  Google Scholar 

  67. Schleinitz D, Krause K, Wohland T, Gebhardt C, Linder N, Stumvoll M, et al. Identification of distinct transcriptome signatures of human adipose tissue from fifteen depots. Eur J Hum Genet. 2020;28:1714–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Roson-Burgo B, Sanchez-Guijo F, Del Cañizo C, De Las RJ. Insights into the human mesenchymal stromal/stem cell identity through integrative transcriptomic profiling. BMC Genom. 2016;17:944.

    Article  Google Scholar 

  69. Bradford ST, Nair SS, Statham AL, van Dijk SJ, Peters TJ, Anwar F, et al. Methylome and transcriptome maps of human visceral and subcutaneous adipocytes reveal key epigenetic differences at developmental genes. Sci Rep. 2019;9:9511.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Min SY, Desai A, Yang Z, Sharma A, DeSouza T, Genga RMJ, et al. Diverse repertoire of human adipocyte subtypes develops from transcriptionally distinct mesenchymal progenitor cells. Proc Natl Acad Sci USA. 2019;116:17970–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Momeni K, Ghorbian S, Ahmadpour E, Sharifi R. Unraveling the complexity: understanding the deconvolutions of RNA-seq data. Transl Med Commun. 2023;8:21.

    Article  Google Scholar 

  72. Glastonbury CA, Couto Alves A, El-Sayed Moustafa JS, Small KS. Cell-type heterogeneity in adipose tissue Is associated with complex traits and reveals disease-relevant cell-specific eQTLs. Am J Hum Genet. 2019;104:1013–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Sorek G, Haim Y, Chalifa-Caspi V, Lazarescu O, Ziv-Agam M, Hagemann T, et al. sNucConv: A bulk RNA-seq deconvolution method trained on single-nucleus RNA-seq data to estimate cell-type composition of human adipose tissues. iScience. 2024;27:110368

  74. Lenz M, Arts ICW, Peeters RLM, de Kok TM, Ertaylan G. Adipose tissue in health and disease through the lens of its building blocks. Sci Rep. 2020;10:10433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Li X, Zhang G, Wang M, Lu C, Zhang G, Chen Z, et al. Comparison of stromal vascular fraction cell composition between coleman fat and extracellular matrix/stromal vascular fraction gel. Adipocyte. 2024;13:2360037.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Norreen-Thorsen M, Struck EC, Öling S, Zwahlen M, Von Feilitzen K, Odeberg J, et al. A human adipose tissue cell-type transcriptome atlas. Cell Rep. 2022;40: 111046.

    Article  CAS  PubMed  Google Scholar 

  77. 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.

    Article  Google Scholar 

  78. Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453:783–7.

    Article  CAS  PubMed  Google Scholar 

  79. Yang Loureiro Z, Solivan-Rivera J, Corvera S. Adipocyte heterogeneity underlying adipose tissue functions. Endocrinology. 2022;163:bqab138.

    Article  PubMed  Google Scholar 

  80. Ramirez AK, Dankel SN, Rastegarpanah B, Cai W, Xue R, Crovella M, et al. Single-cell transcriptional networks in differentiating preadipocytes suggest drivers associated with tissue heterogeneity. Nat Commun. 2020;11:2117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Whytock KL, Divoux A, Sun Y, Hopf M, Yeo RX, Pino MF, et al. Isolation of nuclei from frozen human subcutaneous adipose tissue for full-length single-nuclei transcriptional profiling. STAR Prot. 2023;4: 102054.

    Article  CAS  Google Scholar 

  82. Whytock KL, Sun Y, Divoux A, Yu G, Smith SR, Walsh MJ, et al. Single cell full-length transcriptome of human subcutaneous adipose tissue reveals unique and heterogeneous cell populations. iScience. 2022;25: 104772.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Gupta A, Shamsi F, Altemose N, Dorlhiac GF, Cypess AM, White AP, et al. Characterization of transcript enrichment and detection bias in single-nucleus RNA-seq for mapping of distinct human adipocyte lineages. Genome Res. 2022;32:242–57.

    Article  PubMed  PubMed Central  Google Scholar 

  84. 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;559:103–8.

    Article  CAS  PubMed  Google Scholar 

  85. Liu X, Xiang Q, Xu F, Huang J, Yu N, Zhang Q, et al. Single-cell RNA-seq of cultured human adipose-derived mesenchymal stem cells. Sci Data. 2019;6: 190031.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Palani NP, Horvath C, Timshel PN, Folkertsma P, Grønning AGB, Henriksen TI, et al. Adipogenic and SWAT cells separate from a common progenitor in human brown and white adipose depots. Nat Metab. 2023;5:996–1013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Liu Q, Zhang P, Yuan X, Ya O, Li Q, Li J, et al. Investigate the stemness of adult adipose-derived stromal cells based on single-cell RNA-sequencing. Cell Biol Int. 2022;46:2118–31.

    Article  CAS  PubMed  Google Scholar 

  88. Acosta JR, Joost S, Karlsson K, Ehrlund A, Li X, Aouadi M, et al. Single cell transcriptomics suggest that human adipocyte progenitor cells constitute a homogeneous cell population. Stem Cell Res Ther. 2017;8:250.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Vijay J, Gauthier M-F, Biswell RL, Louiselle DA, Johnston JJ, Cheung WA, et al. Single-cell analysis of human adipose tissue identifies depot and disease specific cell types. Nat Metab. 2020;2:97–109.

    Article  PubMed  Google Scholar 

  90. Strieder-Barboza C, Flesher C, Geletka L, Delproposto J, Eichler T, Akinleye O, et al. Single-nuclei transcriptome of human adipose tissue reveals metabolically distinct depot-specific adipose progenitor subpopulations. BioRxiv. [Preprint]. 2022. https://doi.org/10.1101/2022.06.29.496888

  91. Hildreth AD, Ma F, Wong YY, Sun R, Pellegrini M, O’Sullivan TE. Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nat Immunol. 2021;22:639–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Michailidou Z, Gomez-Salazar M, Alexaki VI. Innate immune cells in the adipose tissue in health and metabolic disease. J Innate Immun. 2022;14:4–30.

    Article  CAS  PubMed  Google Scholar 

  93. Gu S, Gong Z, Liu S, Lu G, Ling Y, Wei Y, et al. Global single-cell sequencing landscape of adipose tissue of different anatomical site origin in humans. Stem Cells Int. 2023;2023:8282961.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Emont MP, Jacobs C, Essene AL, Pant D, Tenen D, Colleluori G, et al. A single-cell atlas of human and mouse white adipose tissue. Nature. 2022;603:926–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184:3573–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Massier L, Jalkanen J, Elmastas M, Zhong J, Wang T, Nono Nankam PA, et al. An integrated single cell and spatial transcriptomic map of human white adipose tissue. Nat Commun. 2023;14:1438.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Møller AF, Madsen JGS. JOINTLY: interpretable joint clustering of single-cell transcriptomes. Nat Commun. 2023;14:8473.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Tang L, Li T, Xie J, Huo Y. Diversity and heterogeneity in human breast cancer adipose tissue revealed at single-nucleus resolution. Front Immunol. 2023;14:1158027.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Bäckdahl J, Franzén L, Massier L, Li Q, Jalkanen J, Gao H, et al. Spatial mapping reveals human adipocyte subpopulations with distinct sensitivities to insulin. Cell Metab. 2021;33:1869-1882.e6.

    Article  PubMed  Google Scholar 

  100. Lee YH, Petkova AP, Granneman JG. Identification of an adipogenic niche for adipose tissue remodeling and restoration. Cell Metab. 2013;18:355–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Kapur SK, Katz AJ. Review of the adipose derived stem cell secretome. Biochimie. 2013;95:2222–8.

    Article  CAS  PubMed  Google Scholar 

  102. Insenser M, Montes-Nieto R, Vilarrasa N, Lecube A, Simó R, Vendrell J, et al. A nontargeted proteomic approach to the study of visceral and subcutaneous adipose tissue in human obesity. Mol Cell Endocrinol. 2012;363:10–9.

    Article  CAS  PubMed  Google Scholar 

  103. Pérez-Pérez R, Ortega-Delgado FJ, García-Santos E, López JA, Camafeita E, Ricart W, et al. Differential proteomics of omental and subcutaneous adipose tissue reflects their unalike biochemical and metabolic properties. J Proteome Res. 2009;8:1682–93.

    Article  PubMed  Google Scholar 

  104. Hruska P, Kucera J, Pekar M, Holéczy P, Mazur M, Buzga M, et al. Proteomic signatures of human visceral and subcutaneous adipocytes. J Clin Endocrinol Metab. 2022;107:755–75.

    Article  PubMed  Google Scholar 

  105. Kheterpal I, Ku G, Coleman L, Yu G, Ptitsyn AA, Floyd ZE, et al. Proteome of human subcutaneous adipose tissue stromal vascular fraction cells versus mature adipocytes based on DIGE. J Proteome Res. 2011;10:1519–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Peinado JR, Jimenez-Gomez Y, Pulido MR, Ortega-Bellido M, Diaz-Lopez C, Padillo FJ, et al. The stromal-vascular fraction of adipose tissue contributes to major differences between subcutaneous and visceral fat depots. Proteomics. 2010;10:3356–66.

    Article  CAS  PubMed  Google Scholar 

  107. Raajendiran A, Krisp C, Souza DPD, Ooi G, Burton PR, Taylor RA, et al. Proteome analysis of human adipocytes identifies depot-specific heterogeneity at metabolic control points. Am J Physiol Endocrinol Metab. 2021;320:E1068–84.

    Article  CAS  PubMed  Google Scholar 

  108. Xie X, Yi Z, Bowen B, Wolf C, Flynn CR, Sinha S, et al. Characterization of the human adipocyte proteome and reproducibility of protein abundance by one-dimensional gel electrophoresis and HPLC-ESI-MS/MS. J Proteome Res. 2010;9:4521–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Cai J, Wang F, Shao M. The emerging importance of mitochondria in white adipocytes: neither last nor least. Endocrinol Metab (Seoul). 2023;38:493–503.

    Article  CAS  PubMed  Google Scholar 

  110. Larsen JK, Kruse R, Sahebekhtiari N, Moreno-Justicia R, Gomez Jorba G, Petersen MH, et al. High-throughput proteomics uncovers exercise training and type 2 diabetes-induced changes in human white adipose tissue. Sci Adv. 2023;9:eadi7548.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Capra E, Beretta R, Parazzi V, Viganò M, Lazzari L, Baldi A, et al. Changes in the proteomic profile of adipose tissue-derived mesenchymal stem cells during passages. Proteome Sci. 2012;10:46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Kim J, Choi YS, Lim S, Yea K, Yoon JH, Jun D-J, et al. Comparative analysis of the secretory proteome of human adipose stromal vascular fraction cells during adipogenesis. Proteomics. 2010;10:394–405.

    Article  CAS  PubMed  Google Scholar 

  113. Dadras M, May C, Wagner JM, Wallner C, Becerikli M, Dittfeld S, et al. Comparative proteomic analysis of osteogenic differentiated human adipose tissue and bone marrow-derived stromal cells. J Cell Mol Med. 2020;24:11814–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Navarro-Perez J, Vidal-Puig A, Carobbio S. Recent developments in adipose tissue-secreted factors and their target organs. Curr Opin Genet Dev. 2023;80: 102046.

    Article  CAS  PubMed  Google Scholar 

  115. Alonso-Alonso ML, García-Posadas L, Diebold Y. Extracellular vesicles from human adipose-derived mesenchymal stem cells: a review of common cargos. Stem Cell Rev Rep. 2022;18:854–901.

    Article  CAS  PubMed  Google Scholar 

  116. Cai Y, Li J, Jia C, He Y, Deng C. Therapeutic applications of adipose cell-free derivatives: a review. Stem Cell Res Ther. 2020;11:312.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Shukla L, Yuan Y, Shayan R, Greening DW, Karnezis T. Fat therapeutics: The clinical capacity of adipose-derived stem cells and exosomes for human disease and tissue regeneration. Front Pharmacol. 2020;11:158.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Pinheiro-Machado E, Getova VE, Harmsen MC, Burgess JK, Smink AM. Towards standardization of human adipose-derived stromal cells secretomes. Stem Cell Rev Rep. 2023;19:2131–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Alvarez-Llamas G, Szalowska E, de Vries MP, Weening D, Landman K, Hoek A, et al. Characterization of the human visceral adipose tissue secretome. Mol Cell Proteom. 2007;6:589–600.

    Article  CAS  Google Scholar 

  120. Hartwig S, De Filippo E, Göddeke S, Knebel B, Kotzka J, Al-Hasani H, et al. Exosomal proteins constitute an essential part of the human adipose tissue secretome. Biochim Biophys Acta Proteins Proteom. 2019;1867: 140172.

    Article  CAS  PubMed  Google Scholar 

  121. Bennett HM, Stephenson W, Rose CM, Darmanis S. Single-cell proteomics enabled by next-generation sequencing or mass spectrometry. Nat Methods. 2023;20:363–74.

    Article  CAS  PubMed  Google Scholar 

  122. Wu T, Womersley HJ, Wang JR, Scolnick J, Cheow LF. Time-resolved assessment of single-cell protein secretion by sequencing. Nat Methods. 2023;20:723–34.

    Article  CAS  PubMed  Google Scholar 

  123. Lange M, Angelidou G, Ni Z, Criscuolo A, Schiller J, Blüher M, et al. AdipoAtlas: a reference lipidome for human white adipose tissue. Cell Rep Med. 2021;2: 100407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Hou B, Zhao Y, He P, Xu C, Ma P, Lam SM, et al. Targeted lipidomics and transcriptomics profiling reveal the heterogeneity of visceral and subcutaneous white adipose tissue. Life Sci. 2020;245: 117352.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Wang Z, Cao M, Lam SM, Shui G. Embracing lipidomics at single-cell resolution: promises and pitfalls. TrAC Trends Anal Chem. 2023;160: 116973.

    Article  CAS  Google Scholar 

  126. Mohiuddin OA, Campbell B, Poche JN, Thomas-Porch C, Hayes DA, Bunnell BA, et al. Decellularized adipose tissue: biochemical composition, in vivo analysis and potential clinical applications. Adv Exp Med Biol. 2020;1212:57–70.

    Article  CAS  PubMed  Google Scholar 

  127. Riis S, Hansen AC, Johansen L, Lund K, Pedersen C, Pitsa A, et al. Fabrication and characterization of extracellular matrix scaffolds obtained from adipose-derived stem cells. Methods. 2020;171:68–76.

    Article  CAS  PubMed  Google Scholar 

  128. Thomas-Porch C, Li J, Zanata F, Martin EC, Pashos N, Genemaras K, et al. Comparative proteomic analyses of human adipose extracellular matrices decellularized using alternative procedures. J Biomed Mater Res A. 2018;106:2481–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Major G, Simcock J, Kumar A, Kleffmann T, Woodfield TBF, Lim KS. Comprehensive matrisome profiling of human adipose tissue for soft tissue reconstruction. Adv Biol. 2024;8: e2300448.

    Article  Google Scholar 

  130. Peña AN, Garcia JA, Elisseeff JH. Translational considerations for adipose-derived biological scaffolds for soft tissue repair. Curr Opin Biomed Eng. 2021;20: 100321.

    Article  Google Scholar 

  131. Christopher JA, Geladaki A, Dawson CS, Vennard OL, Lilley KS. Subcellular transcriptomics and proteomics: a comparative methods review. Mol Cell Proteom. 2022;21: 100186.

    Article  CAS  Google Scholar 

  132. McAllan L, Baranasic D, Villicaña S, Brown S, Zhang W, Lehne B, et al. Integrative genomic analyses in adipocytes implicate DNA methylation in human obesity and diabetes. Nat Commun. 2023;14:2784.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bilson J, Sethi JK, Byrne CD. Heterogeneity of white adipocytes in metabolic disease. Curr Opin Clin Nutr Metab Care. 2023;26:72–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Zannettino ACW, Paton S, Arthur A, Khor F, Itescu S, Gimble JM, et al. Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J Cell Physiol. 2008;214:413–21.

    Article  CAS  PubMed  Google Scholar 

  135. Raajendiran A, Ooi G, Bayliss J, O’Brien PE, Schittenhelm RB, Clark AK, et al. Identification of metabolically distinct adipocyte progenitor cells in human adipose tissues. Cell Rep. 2019;27:1528-1540.e7.

    Article  CAS  PubMed  Google Scholar 

  136. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Tissue-based map of the human proteome. Science. 2015;347:1260419.

    Article  PubMed  Google Scholar 

  137. Karlsson M, Zhang C, Méar L, Zhong W, Digre A, Katona B, et al. A single-cell type transcriptomics map of human tissues. Sci Adv. 2021;7:eabh2169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Tarhan L, Bistline J, Chang J, Galloway B, Hanna E, Weitz E. Single cell portal: an interactive home for single-cell genomics data. BioRxiv. [Preprint]. 2023. https://doi.org/10.1101/2023.07.13.5488862023.

Download references

Acknowledgements

Not applicable.

Funding

No external funding was received to assist with the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, A.T., Ga.T., G.D.R.; writing—original draft preparation, Ga.T.; writing—review and editing A.T., G.D.R., Gi.T., Ga.T. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Gabriele Toietta.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interest

The authors declare that they have no competing of interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Di Rocco, G., Trivisonno, A., Trivisonno, G. et al. Dissecting human adipose tissue heterogeneity using single‐cell omics technologies. Stem Cell Res Ther 15, 322 (2024). https://doi.org/10.1186/s13287-024-03931-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-024-03931-w

Keywords