Skip to main content

Single-cell transcriptomics of clinical grade adipose-derived regenerative cells reveals consistency between donors independent of gender and BMI

Abstract

Adipose-derived regenerative cells (ADRCs) also referred to as the stromal vascular fraction, provide an ample source of stem cells with widespread regenerative therapeutic use. Being heterogenous in nature, possibly affecting the clinical outcome after stem cell treatment, the ADRC- donor, -BMI, and -gender may have a large impact on ADRC composition and quality but this remains largely unexplored. Herein, we provide a comprehensive single-cell RNA sequencing ADRC mapping across two cell trial intervention studies but found no gender- or BMI-related variations, except for a minor female increase in PI16/CD55-expressing stem cells. Indeed, ADRC heterogeneity was surprisingly minimal between donors. This provides important decision-making support on adipose stem cell donor selection for stem cell treatments, and suggest that donor, gender and BMI should be regarded as less influential.

Introduction

Subcutaneous adipose tissue (SAT) remains an inexhaustible source of stem cells that are used for various stem cell interventions [1]. Adipose-derived regenerative cells (ADRCs, a mixture of regenerative cells also referred to as stromal vascular fraction (SVF)) remain the cellular origin of SAT homeostasis and -expansion, and may be isolated for immediately use in autologous cell transplantation [2,3,4]. Alternatively, ADRCs can be expanded in culture to obtain adipose-derived stromal/stem cells (ASCs) before stem cell transplantations into patients in an autologous or allogeneic design and currently, more than 350 trials using adipose tissue-derived cells are registered as either completed, or recruiting at Clinical.trials.gov. However, since the ADRCs encompass all the cell types present in adipose tissue except mature adipocytes, the specific cell composition and heterogeneity may contribute significantly to the clinical outcome after stem cell treatments [1, 5,6,7]. For therapeutic use, several attempts have been utilized to obtain a more homogenous population of the stem cells such as adherence [5] and pre-/deselection of ADRC subpopulations based on e.g. their expression of membrane proteins (e.g. PECAM1-positive endothelial- [8] or Integrin α10β1-selected mesenchymal stem cells [9]). To define ADRC or ASC heterogeneity, Kostecka and colleagues [1], most recently reviewed ADRC single-cell RNA sequencing (scRNAseq) studies and reported that fat deposit, donor-age and health status such as obesity, Type 2 diabetes (T2D) etc., as well as ADRC isolation and expansion protocols may impact heterogeneity and also clinical stem cell intervention outcome. Yet, studies in the field of metabolomics [10] have suggested that ADRC cell composition indeed is similar between fat deposits, despite that certain CD55/PI16-progenitors may correlate with the metabolic phenotype [10]. Such discrepancies may be explained by the small scRNAseq sample- and cell size [1, 10]. However, another caveat to these previous studies is the numerous approaches used for isolating ADRCs [10], procedures that are generally not compatible with clinical stem cell production. Thus, despite some data exist, ADRC heterogeneity between donors remains relatively uncertain. Moreover, it is surprising that no previous investigations focus on gender as a component underlying ADRC heterogeneity. To stem cell therapy, donor cell diversity and its relation to gender is a very important issue when selecting the most proper ADRC donor to benefit clinical outcome.

Herein we therefore performed scRNAseq of ADRCs isolated by the same Good manufacturing practice (GMP) compatible and reproducible approach across samples from two clinical stem cell interventions. With this, we investigated overall and individual cell composition heterogeneity, with a subsequent focus on variations in cell composition between men and women to aid in donor selection.

Results and discussion

GMP compatible ADRCs for cell therapeutics reveal high quality scRNAseq data

ADRCs were isolated from SAT obtained in relation to two phase II randomized, controlled trials (RCTs), conducted at Odense University Hospital, Denmark. The RCTs were designed to test the efficacy of autologous non-cultured ADRC therapy versus placebo (randomization ratio 1:1) in relation to treatment of breast cancer-related secondary lymphedema (BCRL, n = 80, NCT03776721 [11]) and post-radical prostatectomy related erectile dysfunction (pRP-ED, n = 70, EudraCT 2015–005140-33), respectively. From each trial, samples from 7 individuals were randomly selected (BCRL, females, cell therapy n = 7 and pRP-ED, males, cell therapy n = 4; placebo: n = 3) upon availability, for further scRNAseq processing. As previously exploited by us and others [2, 11,12,13,14] in several cell therapy trials, the CE-marked, GMP-compliant device (Cytori Celution IV) cell isolation platform was used for isolation of the ADRCs. Cell numbers and -sizes were similar between donors (data not shown) and in agreement with our previous studies [2, 11,12,13,14]. Immediately after isolation, we applied the ADRCs to a recently in-house validated methanol-fixation and freezing step [8, 15, 16] based scRNAseq protocol. With this, we facilitated high RNA integrity of the ADRCs (Supplementary Table 1, Additional File 1) while preserving in situ conditions as closely as possible. High-throughput scRNA-seq using 10X Genomics combined with NovaSeq was followed by quality controlling (Supplementary Table 1, Additional File 1) and data integration. Resulting quality-filtered, single-cell data (Supplementary Table 1, Additional File 1) from the 14 patients included a total of 111,983 human ADRCs that were integrated. This number of high quality GMP compatible ADRCs exceeded previous scRNAseq datasets from trials, thus providing an opportunity to investigate clinically relevant ADRC heterogeneity in detail between donors and genders.

ADRCs enclose a mixture of regenerative vascular cell types

Prior to donor and gender comparison, we initially performed unbiased scRNAseq clustering based on ADRC expression of the canonical markers PECAM1, PTPRC, TAGLN and CFD, respectively (Supplementary Figs. 14, Additional File 2). As such, we identified 24 cell clusters (Fig. 1A–E and Supplementary Table 2, Additional File 1) falling into four major well-separated subgroups of endothelial-, immune-perivascular mural- and adipose stem/progenitor cells. In line with Massier and colleagues [10], as well as Gene Ontology enrichment analyses (Supplementary Fig. 24, Additional File 2 and Supplementary Table 3, Additional File 1 using ShinyGO [17]) and selected cell type annotation tools (Supplementary Table 4, Additional File 1: CellMarker2024, Tabula Sapiens [18], Tabula Muris [19], using Enrichr [20]) we annotated the ADRC clusters.

Fig. 1
figure 1

Single-cell RNA sequencing landscape of ADRCs integrated from 14 patients. A Left; Uniform manifold approximation and projection (UMAP) 2D visualization of 111,983 ADRCs in 24 clusters falling into four major and one minor cell categories: endothelial- (EC), immune- (IC), perivascular mural- (PC), adipose stem/progenitor- (AC) and red blood (RBC) cells. Right; Dendrogram showing the hierarchical relationship between the 24 ADRC clusters. BE DimPlot UMAP with subcluster annotation,—fractions (%) and violin plot of selected genes for each of the major cell types i.e. B endothelial- (EC1-4), C perivascular mural- (PC1-3), D immune- (IC1-9), and E adipose stem/progenitor (AC1-7) cells. MyoFCs: myofibrocytes, vSMC: vascular smooth muscle cells, Mϕ: Macrophages, Mo/DC: Monocyte/dendritic cells, LAM: lipid associated macrophages, NKT: natural killer T-cells, APC: adipose precursor cells, CPA: committed preadipocytes, FAP: fibroblast and adipogenic progenitor cells

A major subgroup of ADRCs includes endothelial cells (EC), which are considered to hold the high angiogenic potential of ADRCs [8]. The top-three genes separating ECs from the remaining major subgroups were VWF [21], CLDN5 [22] and ADGRL4 [23, 24] (Fig. 1B and Supplementary Fig. 1C, Additional File 2). ECs could however, be further branched into 4 subsets classified according to known endothelial cell types along the arteriovenous axis of the microvasculature (Fig. 1B and Supplementary Fig. 2A, Additional File 2). The largest EC clusters, EC1 and EC2 expressed arterial- (SEMA3G, HEY1, EFNB2), and to a greater extent, capillary (RGCC, CA4, CD36, FABP4, GPIHBP1) marker genes. PLVAP (endothelial cell-specific plasmalemma vesicle-associated protein) [25] was expressed by all EC-clusters although at higher levels in the minor EC clusters, EC3 and EC4. PLVAP thus marks a transition to post-capillary venules represented by these two subsets that were also characterized by their expression of additional postcapillary venule-associated genes i.e. SELE, ACKR1, ICAM1 and VCAM1. The EC progenitor markers CD34 and ENG, appeared to be uniformly expressed across all 4 EC clusters although the expression pattern of angiogenic and tip-cell EC markers suggests that the majority of potential tissue-resident endothelial progenitors [26] are found within the EC1 and EC2 subsets. The arterial/capillary/venous EC features were similar to those reported by others [10] however, as previously with CD31/PECAM1-pre-selected ADRCs from SAT [8], we failed to identify any LYVE1/PROX1 expressing ECs. Notably the gene signature of our SAT-originating ECs was very similar to ECs from visceral adipose tissue (VAT) [27] (Supplementary Fig. 4B, Additional File 2).

Besides endothelial cells, perivascular mural cells (PC) exhibit vast regenerative potential [28]. Among all ADRCs in our dataset, the PC subgroup was characterized by high expression of RGS5 [29, 30], NDUFA4L2 [31] and ACTA2 (Fig. 1C and Supplementary Fig. 1C, Additional File 2) and encompassed 3 subclusters, PC1-3 with PC1 and PC2 being closest neighbors (Fig. 1A,C). PC2 consists of pericytes defined by the genes PDGFRB, CSPG4 (NG2), KCNJ8, ABCC9, NOTCH3 and RGS5 [31] and as for cluster EC1 and EC2, FABP4, CD36 and PPARG transcripts are abundant suggesting involvement in fatty acid transport across vessels as previously described in human skin [32]. Of the mural cells, PC2 expresses the highest levels of NDUFA4L2 and COX4I2, both encoding subunits of mitochondrial proteins reported to mark human brain pericytes and vascular smooth muscle cells (vSMC) [33] and which decline along the arteriole-arterial smooth muscle cell axis [33]. Thus, the lower NDUFA4L2- and COX4I2-expression in the mural cluster PC1 along with higher levels of genes involved in vSMC contraction including ACTA2, TAGLN, MYH11, MYLK, and CNN1 [31], suggest that PC1 contains cells representing vSMCs. LMOD1- and SYNPO2 have both previously been associated with vSMC maturity [34] and while SYNPO2 is slightly more pronounced in PC3 than in PC2, PC3 can however, be further distinguished by high levels of the chemokines CCL21, CCL19 and CCL2 (top-3 genes) identifying this cluster as myofibrocytes [35] (Fig. 1C).

Altogether, these data for EC and PC subtypes underscores that ADRCs in its fresh form contain a very defined vascular potential, which may be exploited when replenishing ischemic tissues upon ADRC treatment. It will be interesting in the future to further delineate which of these EC and PC clusters that embrace the highest regenerative potential or whether they work in synergy to accomplish revascularization of ischemic tissues.

Stem- and immunomodulatory cells constitute substantial fractions of ADRCs

A large number of studies have reported an immunomodulatory mode of action underlying SVF/ADRC function in stem cell treatments [36]. This capacity is thought to be enclosed mainly within the mesenchymal cell-like population (below) but also the ADRC-derived immune cells (IC) seem involved in this function [37, 38]. In our data, the transmembrane Immune Signaling Adaptor TYROBP (TYROBP), CD45 (PTPRC) and Serglycin (SRGN) separated the IC subgroup from the remaining ADRCs by being the top-3 highest expressed genes (Supplementary Fig. 1C, Additional File 2). The ICs further branched into 9 clusters of immune cells expressing markers of various known immune cells of both myeloid and lymphoid lineages (Fig. 1D and Supplementary Fig. 3, Additional File 2). Most of the identified ICs are of the myeloid lineage (20% of all ADRC, 71% of all ICs) with further specification into monocyte/dendritic/macrophage (IC1,-2,-4) subtypes as well as a neutrophil-like cluster (IC3), mast cells (IC5) and a small cluster of contaminating red blood cells (RBC). A mixed cell cluster (IC9) appeared related to the RBC cluster, both being involved in oxygen transport (GO-biological function, Supplementary Fig. 3, Additional File 2 and Supplementary Table 34, Additional File 1).

Finally, three clusters belonging to the lymphoid lineage, encompassed T/NKT-cells (IC 6 + IC7) and B-cells/plasma cells (IC8). Of note, the frequency distribution and absolute cell counts of circulating immune cells will depend on initial blood infiltration and following washing steps (if any) and may, from a clinical perspective be important [39]. The differences in number- and subtypes of infiltrating immune cells can simply also reflect an actual disease state like in the case of diabetic HIV patients [40]. It is however still speculative, which IC subtypes that facilitates the immunomodulatory action, but both Tregs and monocytes have been suggested to enclose this activity [37, 38]. Besides ICs, also the mesenchymal stem cells in ADRCs possess immunomodulatory action in addition to other functions such as regeneration and wound healing [41]. Our data are in agreement with other studies [10] and showed that a rather large fraction (43%) of ADRCs were classified as adipose stem/progenitor cells (AC) distinguished herein from remaining ADRCs by their high expression of C1S, MFAP4 and NOVA1 (Fig. 1e and Supplementary Fig. 1C, Additional file 2). While Complement C1S, C3, CFD and Microfibril Associated Protein 4, MFAP4 mRNAs are known to mark adipogenic/fibroblast progenitors, the expression of NOVA1 [42, 43] has only very recently been highlighted in human adipose tissue and the stromal vascular fraction (SVF, equal to ADRCs) hereof [42]. Interestingly, Yang and colleagues recognized NOVA1 as a regulator of chromatin-remodeling during- and a prerequisite for adipogenesis in humans [42]. This function appeared to be species-specific and could not be recapitulated in mice, which can possibly add to explain the lack of NOVA1-observations in adipose tissue-derived sc/snRNAseq data that are dominated by mouse data. GEO Profile data (NCBI, GEO Profile ID: 111023595; DataSet ID: 5056 [44] and GEO Profile ID: 83214995; DataSet ID: 4276 [45]) indicate NOVA1 expression to be more pronounced in subcutaneous than visceral fat and that NOVA1 mRNA levels may be higher in human adipose tissue or -derived ASCs from lean as compared to obese individuals. From a large set of integrated human sc- and snRNAseq data [10] NOVA1 was noted in one of the minor omental- but not subcutaneous fat clusters. Herein, we reinspected their original data, and found that NOVA1 indeed was the second highest ranking gene (fold-change/adj. p-value ranking) in one of the largest subcutaneous fat clusters (sfC01 [10]). Our AC class of cells further segregated into 6 common (AC1-6) clusters while one (AC7) was specific to a single individual (male, #M2. Fig. 2C, D and Supplementary Fig. 1A, Additional File 2). Subclusters AC1 and AC3-5 appeared closely related as did subclusters AC2, AC6 and AC7 (Fig. 1A) and all are enriched for Gene Ontology (GO) terms and/or genes related to ECM/fibroblast/stromal cells (Supplementary Fig. 4A, Additional File 2 and Supplementary Table 34, Additional File 1). As noted by several groups [10, 46], the nomenclature for the adipose progenitor/stem cell class (termed fibroblast and adipogenic progenitors (FAPs)) is not well-established possibly due to heterogeneity between species and fat deposits. Despite of this heterogeneity, Massier et al. [10] reported the VAT and SAT clusters from their large integrated sc/snRNAseq object to be very similar. Also our SAT derived AC clusters recapitulate the expression of genes used for clustering visceral fat FAPs [27] including BMPER, recently recognized as a positive modulator of adipogenesis and highly expressed in VAT [27] (Supplementary Fig. 4B, Additional File 2). Jaccard similarity scores between our AC clusters and the described 17 SAT-derived FAPs [10] (Supplementary Fig. 4C, Additional File 2), suggests that clusters AC2, AC6 and to a less degree AC7 represent a central APC cluster, marked by the genes PI16/CD55 characteristic of non-committed APCs. AC1 and AC4-5 confine cells moving towards a committed preadipocyte (CPA) phenotype, while AC3 appears to represent terminally differentiated fibroblast/stromal cells likely arising from a different cell type other than PI16/CD55 stem cells [10]. Finally, we did a comprehensive comparison of our AC data to the AC-marker lists from other substantial human scRNAseq adipose tissue studies (Supplementary Figs, 4–5, Additional File 2). These analyses demonstrated varying degrees of overlap for the AC clusters between studies, which further underscored the design of our clustering strategy. Still, we would like to emphasize a clear importance of input data quality and differences in initial data resolution which may explain reported differences in gene enrichment profiles. Moreover, the selected markers and lack of consensus regarding nomenclature in the field complicate comparisons and cell type annotations especially in the case of the stromal cell types [46].

Fig. 2
figure 2

Comparative analyses of ADRCs between donors, gender and BMI. A Baseline characteristics (age and BMI) were similar between the analyzed male and female subjects (2way ANOVA with Šídák's multiple comparisons test). B The 24 ADRC clusters (AC1-7, EC1-4, IC1-9, PC1-3 and RBC) depicted for each individual (male, black circle and female, open circle) as the fraction (%) of all ADRCs. Multiple Mann–Whitney tests revealed no differences between genders. C, D Distribution of C clusters (% of ADRCs) and D subclusters (as % of their respective major cell class) in each individual (male M1–M7 and female, F1–F7). Overall, cluster distribution was similar between donors. E Comparison of subclusters (as % of their respective major cell class) between male and female individuals showing only adipose stem/progenitor cluster AC6 to be significantly different (Multiple Mann–Whitney test, q = 0.003531). F Distribution the major cell classes AC, EC, IC and PC (% of ADRC) as a function patient BMI. No correlation was observed (Pearson). G Nebulosa UMAP showing ADRCs co-expressing PI16 and CD55. H Comparison of the percentage of CD55/PI16 co-expressing cells (% of ADRCs, left and % of ACs, right) showing significantly higher numbers in females (unpaired t-tests, p = 0.0002 in both cases). I Analysis of BMI (kg/m2) and percentage of PI16/CD55 co-expressing ADRCs or ACs revealed no correlation (Pearson, p = 0.9879 and p = 0.8544, respectively). For multiple Mann–Whitney tests the FDR was controlled using the Two-stage step-up method of Benjamini, Krieger and Yekutieli

Donor and gender dependent ADRC heterogeneity is minimal

With the established ADRC sub-clustering design described above, we next investigated donor and gender impact on ADRC composition. Our scRNAseq data were derived from 7 males and 7 females that aside from gender, were comparable regarding age and BMI (Fig. 2A). The impact of gender on the cluster distribution was initially tested using all clusters individually (Fig. 2B-C) and subsequently for each of the four major cell classes: EC, IC, PC, and AC (Supplementary Fig. 6A, Additional File 2). Overall, these analyses, did not reveal significant differences in ADRC composition between genders. Indeed, the variation in subcluster size was minimal between donors (Fig. 2C, D). Also, the distribution of subclusters within each cell class (Fig. 2D, E) appeared similar between genders except for AC6, that constituted a higher fraction of ACs in females versus males (Multiple Mann–Whitney test, q value = 0.0035). The latter seems to be explained by a significantly higher number of CD55/PI16 co-expressing stem cells in females than in males, but this difference was independent of BMI (Fig. 2G–I). This contradicts previous bulk RNAseq studies [10] where the PI16/CD55-stem cell cluster was suggested to positively correlate to BMI (and a negative metabolic status in general). In fact, we observed no BMI-related differences in the distribution of cell classes (Fig. 2F) or subclusters among the donors in general (Supplementary Fig. 6A, Additional File 2). This may possibly be due to the rather discrete variation in BMI (28.7 ± 2.8 kg/m2 (mean ± SD); range: 24.7–34.7 kg/m2) or because of a more metabolically healthy status of our patient groups (Fig. 2F) as compared to those previously reported [10]. This was also true when comparing cell class distributions in patients having the significantly (t-test, p = 0.0003) lowest versus highest BMI’s (Supplementary Fig. 6C, Additional File 2). Thus, despite a small difference in CD55/PI16 co-expressing stem cells between genders, our data suggest that heterogeneity in ADRC cluster distribution is minimal between cell donors, at least within the discrete, but clinically relevant BMI and age ranges investigated herein. We can, however, not exclude that the difference in CD55/PI16 co-expressing stem cells could be due to the given disease, which differs between genders. Nonetheless, as a direct cell therapeutic product, the smaller very low-frequency populations often described and emphasized in scRNAseq data such as the CD55/PI16 co-expressing stem cells, may seem less physiologically relevant although the interplay between minor and major populations could potentially be important. Previously, Noreen-Thorsen et al. used RNAseq of unfractionated human adipose tissue and observed no obvious gender-related gene expression differences in SAT [47] underscoring the importance of scRNAseq data to identify even small differences as seen herein. However, this issue awaits to be further addressed in the context of clinical efficacy. For instance, one clinical study did report that the injected hematopoietic stem cell (HSC)/ASC ratio along with the presence of endothelial cells was important for a beneficial clinical outcome and that efficacy apparently was not linked to gender, ethnicity, age or BMI although the yield of SVF cells was higher in females than in males. Higher yields from females were also reported by Cremona et al. who analyzed the composition of uncultured ADRCs/SVF isolated in an ATMP-compliant setting from 300+ patients treated at their hospital. Using an enzyme-based method similar to ours [48], they too observed higher ADRC/SVF yields from females versus males, while the distribution of cell types (surface marker by flow cytometry) and other parameters were similar between genders as also was the case with the patient reported outcome measures. Whether the higher percentage of CD55/PI16 co-expressing stem cells observed here for female donors could impact expansion and the cell composition after ADRC culture, when aiming for allogeneic ADRC transplantations, also remains speculative, but is an interesting point to follow in the future as well. For freshly isolated ADRCs, we recently reported that, at the single-cell level, individual autologous cell therapeutic products were indistinguishable regardless of the clinical response [49]. While this supports small variations in ADRC composition to have less impact on the clinical outcome, it may be limited to the specific method of isolation, disease and patient groups among other factors [1]. Therefore a more general knowledge on how the composition of cell therapeutic products may affect clinical outcomes are highly demanded to develop the stem cell field further.

Conclusion

In conclusion, despite minor individual variations may exist, the ADRC cell therapeutic product our patients have received is very similar and with no or very limited impact of gender and BMI on the distribution of cell types. Indeed, the variation was low between donors in general. This is important for regenerative medicine because it suggests that males and females and donors overall have the same prerequisites for a successful treatment when it comes to autologous cell therapy as well as to be equally suited as cell donors for allogeneic cell therapy using cultured ASCs. However, the finding that PI16 and CD55 co-expression is significantly higher in females versus males may potentially impact therapeutic efficacy and should therefore be considered in mixed gender trials using ADRCs/ASCs.

Methods

Human samples

Lipoaspirates for ADRC isolation (n = 14) were obtained from patients enrolled in two clinical RCT studies using non-cultured ADRCs for cell therapy of 1) male participants suffering from erectile dysfunction (ED) following radical prostatectomy (Clinical trials.gov ID:NCT02240823 [50]) and female participants with breast cancer related lymphedema (BCRL). All patients gave written informed consent before participation.

Regulatory approvals

The pRP-ED-trial followed ATMP guidelines and was approved by The Danish Health and Medicines Authority (2015-005140-33), the Danish National Ethics Committee (2017070103) as well as the Danish Data Protection Agency (no 16/2816). The BCRL-trial was approved by The Regional Committees on Health Research Ethics for Southern Denmark (S-20180117) and registered with the Danish Data Protection Agency (18/51767). Both studies were registered at ClinicalTrials.gov (pRP-ED-trial: NCT02240823 and BCRL-trial: NCT03776721).and performed in accordance with the Declaration of Helsinki and ICH-GCP guidelines. All patients gave written informed consent before participation.

Isolation and preparation of ADRCs for single-cell RNA sequencing.

Adipose derived regenerative cells (ADRCs) were isolated from lipoaspirates utilizing a CE-marked, GMP-compliant device (Cytori Celution IV, Lorem Cytori) as described in details [12, 13, 51]. Excess cell material not being returned to the patient was immediately processed for futher downstream single-cell RNA sequencing. To this end and as recently reported, single cell suspensions were obtained by addition of PBS containing 1% ultrapure BSA and 1 U/µl RNAsin PLUS RNase Inhibitor (Promega, Cat.no. N2615) and passage through a 40 µm Flowmi® Cell strainer (VWR, cat.no. 734-5950) before fixation in methanol and storage at − 80 °C until further processing [8, 15]. In both trials, all cell isolations and preparation of ADRCs for scRNAseq were performed by the same staff using the same procedures.

Single-cell RNA sequencing

Prior to sequencing, fixed cells were thawed, rehydrated and immediately loaded onto a 10 × Genomics Chromium controller (10X Genomics, PN110203). Libraries were prepared following the manufacturer instructions using the 10 × Genomics Single-Cell 3’ v2/v3, Chromium Single Cell B Chip Kit, 48 runs (10X Genomics, 10X Genomics, PN-1000073) and sequenced on an Illumina NovaSeq 6000 System (10X Genomics, 20012850). Cell samples were kept for RNA purification, and RIN-values were measured using a RNA 6000 Nano kit (Agilent, cat no. 5067-1512) (Supplementary Table 1, Addional File 1).

scRNAseq data analysis

Single-cell feature count matrices were created using Cell Ranger software (v. 2.1.1 or 3.1.0), for demultiplexing the raw sequencing output files and aligning to the GRCh38 human reference genome. The output metrics generated by Cell Ranger are listed in Extended Data Table 1. Overall, an average sequence depth of 195 million total reads per sample equalling 32,796 reads per cell was obtained. All subsequent analyses were performed using R Version 4.2.3 and the Seurat R software package (Version 4.3.0) in a standard workflow as previously described [8]. Initially, data was converted to single-cell Seurat objects in which genes were expressed in at least five cells, followed by a filtering step excluding low-quality cells with mitochondrial RNA contents above 15%, or expressing less than 200 genes. Next, doublets were predicted and filtered out using DoubletFinder [52], following the authors' standard workflow with an estimated doublet formation rate of 5%. A total of 111,983 cells passed these quality filtering steps and all datasets merged, normalized, scaled and the top 3000 most variable genes identified for use in the following dimensional reduction step. Possible batch-effects were corrected by Harmony integration using the top 30 principal components, before computing UMAP embedding and clustered using Louvain clustering with the resolution parameter set to 0.5. For patients represented by two samples (resuspension in PBS and SSC following methanol fixation), the average number of cells within each cluster was calculated and used for further statistical analyses.

Differential gene expression analysis was performed for each individual cluster using the FindMarkers-command in Seurat and alldifferentially expressed genes are listed in Extended Data Table 2. Differentially expressed genes for each cluster were analyzed for significantly enriched gene ontology (GO) terms using ShinyGO [17] version 0.77 (and beta-version 0.80) (Extended Data Table 3). GO-terms were also extracted from Enrichr [20] as was cell identity predictions from the CellMarker2024-, Tabula Sapiens [18]- and Tabula Muris databases [19] (Extended Data Table 4). For comparing data sets using Jaccard index, the marker genes were selected based on a log2 fold-change > 0.5, and adjusted p value < 0.05 [10]. Data visualization and analysis of scRNAseq data included the use of scCustomize [53] and Nebulosa [54] packages for R.

Statistics

Statistical analyses were performed using the GraphPad Prism software version 10. Statistical significance was set at p < 0.05. and in each case determined using the appropriate statistical method following normality testing, as denoted in the corresponding figure legends.

Availability of data and materials

Following exclusion of sensitive genetic information from RNA-seq raw files using BAMboozle [55], all scRNA-seq data were deposited in the GEO database under the accession code: GSE286321.

References

  1. Kostecka A, et al. Adipose-derived mesenchymal stromal cells in clinical trials: Insights from single-cell studies. Life Sci. 2024;351:122761. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2024.122761.

    Article  PubMed  CAS  Google Scholar 

  2. Sørensen KM, Jensen CH, Sheikh SP, Qvist N, Sørensen JA. Treatment of fistulizing perianal crohn’s disease by autologous microfat enriched with adipose-derived regenerative cells. Inflamm Bowel Dis. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ibd/izab276.

    Article  PubMed Central  Google Scholar 

  3. Jørgensen MG, et al. Adipose-derived regenerative cells and lipotransfer in alleviating breast cancer-related lymphedema: an open-label phase I trial with 4 years of follow-up. Stem Cell Transl Med. 2021;10:844–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/sctm.20-0394.

    Article  CAS  Google Scholar 

  4. Haahr MK, et al. A 12-Month follow-up after a single intracavernous injection of autologous adipose-derived regenerative cells in patients with erectile dysfunction following radical prostatectomy: an open-label phase i clinical trial. Urology. 2018;121:203 e206-203 e213. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.urology.2018.06.018.

    Article  Google Scholar 

  5. Brooks AES, et al. Ex vivo human adipose tissue derived mesenchymal stromal cells (ASC) are a heterogeneous population that demonstrate rapid culture-induced changes. Front Pharmacol. 2019;10:1695. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2019.01695.

    Article  PubMed  CAS  Google Scholar 

  6. Wu L, et al. Chondrocytes cocultured with stromal vascular fraction of adipose tissue present more intense chondrogenic characteristics than with adipose stem cells. Tissue Eng Pt A. 2016;22:336–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/ten.tea.2015.0269.

    Article  CAS  Google Scholar 

  7. Zhang HZ, Chae D-S, Kim S-W. ASC and SVF cells synergistically induce neovascularization in ischemic hindlimb following cotransplantation. Int J Mol Sci. 2021;23:185. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms23010185.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Dhumale P, et al. CD31 defines a subpopulation of human adipose-derived regenerative cells with potent angiogenic effects. Sci Rep-Uk. 2023;13:14401. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-023-41535-1.

    Article  CAS  Google Scholar 

  9. Andersen C, et al. Intraarticular treatment with integrin alpha10beta1-selected mesenchymal stem cells affects microRNA expression in experimental post-traumatic osteoarthritis in horses. Front Vet Sci. 2024;11:1374681. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fvets.2024.1374681.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Massier L, et al. An integrated single cell and spatial transcriptomic map of human white adipose tissue. Nat Commun. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-023-36983-2.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Jørgensen MG, et al. No clinical efficacy of adipose-derived regenerative cells and lipotransfer in breast cancer-related lymphedema: a double-blinded placebo-controlled phase-II trial. Plast Reconstr Surg. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/prs.0000000000011343.

    Article  PubMed  Google Scholar 

  12. Haahr MK, et al. Safety and potential effect of a single intracavernous injection of autologous adipose-derived regenerative cells in patients with erectile dysfunction following radical prostatectomy: an open-label phase I clinical trial. EBioMedicine. 2016;5:204–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ebiom.2016.01.024.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Toyserkani NM, Jensen CH, Andersen DC, Sheikh SP, Sørensen JA. Treatment of breast cancer-related lymphedema with adipose-derived regenerative cells and fat grafts: a feasibility and safety study. Stem Cell Transl Med. 2017;6:1666–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/sctm.17-0037.

    Article  Google Scholar 

  14. Wiborg MH, et al. Treatment with autologous adipose-derived regenerative cells for peyronie’s disease in men: the straight @head pilot study. Eur Urol Open Sci. 2025;71:180–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.euros.2024.12.005.

    Article  PubMed  Google Scholar 

  15. Chen J, et al. PBMC fixation and processing for Chromium single-cell RNA sequencing. J Transl Med. 2018;16:198. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-018-1578-4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Ellman DG, et al. Protocol to achieve high-resolution single-cell transcriptomics of cardiomyocytes in multiple species. STAR Protocols. 2024;5:103194. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.xpro.2024.103194.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Ge SX, Jung D, Yao R. ShinyGO: a graphical gene-set enrichment tool for animals and plants. Bioinformatics. 2020;36:2628–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/btz931.

    Article  PubMed  CAS  Google Scholar 

  18. Consortium, T. T. S. et al. The Tabula Sapiens: A multiple-organ, single-cell transcriptomic atlas of humans. Science. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.abl4896

  19. Consortium, T. T. M. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature. 2018;562:367–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-018-0590-4.

    Article  CAS  Google Scholar 

  20. Kuleshov MV, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44:W90–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gkw377.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Wang X, Starodubtseva MN, Kapron CM, Liu J. Cadmium, von Willebrand factor and vascular aging. npj Aging. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41514-023-00107-3.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Richards M, et al. Claudin5 protects the peripheral endothelial barrier in an organ and vessel-type-specific manner. Elife. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.78517.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Al Mahri S, et al. Profiling of G-protein coupled receptors in adipose tissue and differentiating adipocytes offers a translational resource for obesity/metabolic research. Cells. 2023;12:377. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells12030377.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Favara DM, et al. Elevated expression of the adhesion GPCR ADGRL4/ELTD1 promotes endothelial sprouting angiogenesis without activating canonical GPCR signalling. Sci Rep-Uk. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-021-85408-x.

    Article  Google Scholar 

  25. Guo L, Zhang H, Hou Y, Wei T, Liu J. Plasmalemma vesicle-associated protein: a crucial component of vascular homeostasis. Exp Ther Med. 2016;12:1639–44. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/etm.2016.3557.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Chambers SEJ, et al. Current concepts on endothelial stem cells definition, location, and markers. Stem Cell Transl Med. 2021;10:S54–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/sctm.21-0022.

    Article  Google Scholar 

  27. Garritson JD, et al. BMPER is a marker of adipose progenitors and adipocytes and a positive modulator of adipogenesis. Commun Biol. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42003-023-05011-w.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Ahmed TA, El-Badri N. Pericytes: the role of multipotent stem cells in vascular maintenance and regenerative medicine. Adv Exp Med Biol. 2018;1079:69–86. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/5584_2017_138.

    Article  PubMed  CAS  Google Scholar 

  29. Frommer ML, et al. Single-cell analysis of ADSC interactions with fibroblasts and endothelial cells in scleroderma skin. Cells. 2023;12:1784. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells12131784.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Holm A, Heumann T, Augustin HG. Microvascular mural cell organotypic heterogeneity and functional plasticity. Trends Cell Biol. 2018;28:302–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tcb.2017.12.002.

    Article  PubMed  Google Scholar 

  31. Van Splunder H, Villacampa P, Martínez-Romero A, Graupera M. Pericytes in the disease spotlight. Trends Cell Biol. 2024;34:58–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tcb.2023.06.001.

    Article  PubMed  PubMed Central  Google Scholar 

  32. He H, et al. Single-cell transcriptome analysis of human skin identifies novel fibroblast subpopulation and enrichment of immune subsets in atopic dermatitis. J Allergy Clin Immunol. 2020;145:1615–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2020.01.042.

    Article  PubMed  CAS  Google Scholar 

  33. Mesa-Ciller C, et al. Unique expression of the atypical mitochondrial subunit NDUFA4L2 in cerebral pericytes fine tunes HIF activity in response to hypoxia. J Cereb Blood Flow Metab. 2023;43:44–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0271678X221118236.

    Article  PubMed  CAS  Google Scholar 

  34. Kumar A, et al. Specification and diversification of pericytes and smooth muscle cells from mesenchymoangioblasts. Cell Rep. 2017;19:1902–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2017.05.019.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Chen C, et al. Single-cell and spatial transcriptomics reveal POSTN+ cancer-associated fibroblasts correlated with immune suppression and tumour progression in non-small cell lung cancer. Clin Transl Med. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ctm2.1515.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Al-Ghadban S, Bunnell BA. Adipose tissue-derived stem cells: immunomodulatory effects and therapeutic potential. Physiology. 2020;35:125–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/physiol.00021.2019.

    Article  PubMed  CAS  Google Scholar 

  37. Bensemmane L, Milliat F, Treton X, Linard C. Systemically delivered adipose stromal vascular fraction mitigates radiation-induced gastrointestinal syndrome by immunomodulating the inflammatory response through a CD11b+ cell-dependent mechanism. Stem Cell Res Ther. 2023;14:325. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-023-03562-7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Gandolfi S, et al. Stromal vascular fraction in the treatment of myositis. Cell Death Discov. 2023;9:346. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41420-023-01605-9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Aguilo-Seara G, et al. Extent of tissue washing can significantly alter the composition of adipose-derived stromal vascular fraction cell preparations: implications for clinical translation. Stem Cell Transl Med. 2023;12:391–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/stcltm/szad025.

    Article  CAS  Google Scholar 

  40. Wanjalla CN, et al. Single-cell analysis shows that adipose tissue of persons with both HIV and diabetes is enriched for clonal, cytotoxic, and CMV-specific CD4+ T cells. Cell Reports Medicine. 2021;2:100205. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.xcrm.2021.100205.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Zhidu S, Ying T, Rui J, Chao Z. Translational potential of mesenchymal stem cells in regenerative therapies for human diseases: challenges and opportunities. Stem Cell Res Ther. 2024;15:266. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-024-03885-z.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Yang Z, et al. NOVA1 prevents overactivation of the unfolded protein response and facilitates chromatin access during human white adipogenesis. Nucleic Acids Res. 2023;51:6981–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gkad469.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Vernia S, et al. An alternative splicing program promotes adipose tissue thermogenesis. Elife. 2016;5:e17672. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.17672.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Oñate B, et al. Stem cells isolated from adipose tissue of obese patients show changes in their transcriptomic profile that indicate loss in stemcellness and increased commitment to an adipocyte-like phenotype. BMC Genomics. 2013;14:625. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2164-14-625.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Tam CS, et al. An early inflammatory gene profile in visceral adipose tissue in children. Int J Pediatr Obes. 2011;6:e360–3. https://doiorg.publicaciones.saludcastillayleon.es/10.3109/17477166.2011.575152.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Emont MP, et al. A single-cell atlas of human and mouse white adipose tissue. Nature. 2022;603:926. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-022-04518-2.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Norreen-Thorsen M, et al. A human adipose tissue cell-type transcriptome atlas. Cell Rep. 2022;40:111046. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2022.111046.

    Article  PubMed  CAS  Google Scholar 

  48. Cremona M, et al. Processing adipose tissue samples in a GMP environment standardizes the use of SVF in cell therapy treatments: data on 302 patients. Biomedicines. 2023;11:2533. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biomedicines11092533.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Andersen DC, Bjerre FA, Jorgensen MG, Sorensen JA, Jensen CH. Clinical outcome is unlinked to injection of adipose-derived regenerative cells in the axilla of breast cancer-related lymphedema patients. Stem Cell Res Ther. 2024;15:426. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-024-04037-z.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Hansen, S. T. et al. A Randomized double-blind, placebo-controlled trial of adipose-derived regenerative cells injected into corpora cavernosum following radical prostatectomy. EBioMedicine;2025.

  51. Hansen ST, Jensen CH, Sørensen JA, Sheikh SP, Lund L. Isolation of adipose derived regenerative cells for the treatment of erectile dysfunction following radical prostatectomy. J Vis Exp. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3791/59183.

    Article  PubMed  Google Scholar 

  52. Mcginnis CS, Murrow LM, Gartner ZJ. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 2019;8:329-337.e324. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cels.2019.03.003.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. scCustomize: custom visualizations & functions for streamlined analyses of single cell sequencing. (2021).

  54. Alquicira-Hernandez J, Powell JE. Nebulosa recovers single-cell gene expression signals by kernel density estimation. Bioinformatics. 2021;37:2485–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/btab003.

    Article  PubMed  CAS  Google Scholar 

  55. Ziegenhain C, Sandberg R. BAMboozle removes genetic variation from human sequence data for open data sharing. Nat Commun. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-021-26152-8.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Bäckdahl J, et al. Spatial mapping reveals human adipocyte subpopulations with distinct sensitivities to insulin. Cell Metab. 2021;33:1869-1882.e1866. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2021.07.018.

    Article  PubMed  CAS  Google Scholar 

  57. Hildreth AD, et al. Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nat Immunol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41590-021-00922-4.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Vijay J, et al. Single-cell analysis of human adipose tissue identifies depot- and disease-specific cell types. Nat Metab. 2020;2:97–109. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42255-019-0152-6.

    Article  PubMed  Google Scholar 

  59. Rondini EA, Granneman JG. Single cell approaches to address adipose tissue stromal cell heterogeneity. Biochem J. 2020;477:583–600. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/bcj20190467.

    Article  PubMed  CAS  Google Scholar 

  60. Rondini EA, Ramseyer VD, Burl RB, Pique-Regi R, Granneman JG. Single cell functional genomics reveals plasticity of subcutaneous white adipose tissue (WAT) during early postnatal development. Mol Metab. 2021;53:101307. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molmet.2021.101307.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Raajendiran A, et al. Identification of metabolically distinct adipocyte progenitor cells in human adipose tissues. Cell Rep. 2019;27:1528-1540 e1527. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2019.04.010.

    Article  PubMed  CAS  Google Scholar 

  62. Oguri Y, et al. CD81 controls beige fat progenitor cell growth and energy balance via FAK signaling. Cell. 2020;182:563-577 e520. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2020.06.021.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Ramirez AK, et al. Single-cell transcriptional networks in differentiating preadipocytes suggest drivers associated with tissue heterogeneity. Nat Commun. 2020;11:2117. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-020-16019-9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank Tina Kjærgaard Andersen, Tonja Lyngse Jørgensen and Christina Fenger (Amplexa, Odense, DK) for excellent technical assistance.

Funding

This study was supported by the Innovation Fund Denmark (#7051-00001A), the Novo Nordisk Foundation (#NNF21OC0071847, #NNF19OC0055353), and Research funding from Odense University Hospital, the Region of Southern Denmark, and Clinical Institute at the University of Southern Denmark.

Author information

Authors and Affiliations

Authors

Contributions

FAB: assembly of data, data analysis, manuscript editing, final approval of manuscript; JVN: conception and design, collection and/or assembly of data, data analysis and interpretation, final approval of manuscript; MB: collection and/or assembly of data, data analysis and interpretation, final approval of manuscript; PD: collection and assembly of data, final approval of manuscript; MGJ and STH: provision of study patient materials, final approval of manuscript; MT: collection and/or assembly of data, final approval of manuscript; LL and JAS: conception and design, provision of study patient materials, financial support, final approval of manuscript; DCA: conception and design, data interpretation, manuscript writing, financial support, final approval of manuscript; CHJ: conception and design, data analysis and interpretation, manuscript writing, financial support, final approval of manuscript.

Corresponding author

Correspondence to Charlotte Harken Jensen.

Ethics declarations

Ethics approval and consent to participate

The pRP-ED-trial (entitled “Can fat derived stem cells (SVF) be used in the treatment of erectile dysfunction after prostatectomy—a randomized, placebo-controlled, double-blind clinical trial”) was approved on January 29th, 2018 by the Danish National Ethics Committee (no. 2017070103) and the BCRL-trial (entitled “Treatment of breast cancer-related lymphedema with stem cells and fat grafting”) by The Regional Committees on Health Research Ethics for Southern Denmark on December 7th, 2018 (no. S-20180117). Both studies were performed in accordance with the Declaration of Helsinki and ICH-GCP guidelines and all patients gave written informed consent before participation.

Consent for publication

The informed consent in both trials included a section regarding publication of study data and was signed by all patients.

Competing interests

The authors declare no competing interests.

Artificial intelligence

The authors declare that they have not used Artificial Intelligence in this study.

Additional information

Publisher's Note

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

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bjerre, F.A., Nielsen, J.V., Burton, M. et al. Single-cell transcriptomics of clinical grade adipose-derived regenerative cells reveals consistency between donors independent of gender and BMI. Stem Cell Res Ther 16, 109 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04234-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04234-4

Keywords