Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Resource
  • Published:

Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity

Abstract

White adipose tissue (WAT) is an essential regulator of energy storage and systemic metabolic homeostasis. Regulatory networks consisting of immune and structural cells are necessary to maintain WAT metabolism, which can become impaired during obesity in mammals. Using single-cell transcriptomics and flow cytometry, we unveil a large-scale comprehensive cellular census of the stromal vascular fraction of healthy lean and obese human WAT. We report new subsets and developmental trajectories of adipose-resident innate lymphoid cells, dendritic cells and monocyte-derived macrophage populations that accumulate in obese WAT. Analysis of cell–cell ligand–receptor interactions and obesity-enriched signaling pathways revealed a switch from immunoregulatory mechanisms in lean WAT to inflammatory networks in obese WAT. These results provide a detailed and unbiased cellular landscape of homeostatic and inflammatory circuits in healthy human WAT.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Single-cell sequencing reveals the cellular heterogeneity of the SVF of human WAT.
Fig. 2: Single-cell analysis unveils unique human WAT-resident ILC subsets.
Fig. 3: RNA-velocity and CytoTRACE analyses identify a shared ILC precursor to mature adipose ILC1s and ILC3s.
Fig. 4: Single-cell analysis identifies unique cell lineages within human WAT myeloid populations.
Fig. 5: RNA-velocity analysis uncovers a distinct monocyte state upstream of adipose IMs in obese individuals.
Fig. 6: CellphoneDB analysis reveals the lean human WAT interactome.
Fig. 7: CellphoneDB analysis predicts a dramatic remodeling of the human WAT interactome in obesity.
Fig. 8: Analysis of putative upstream regulators uncovers a distinct obese human WAT-enriched signalome.

Similar content being viewed by others

Data availability

The scRNA-seq datasets are accessible from the Gene Expression Omnibus with accession nos. GSE155960 and GSE156110.

References

  1. Christ, A., Lauterbach, M. & Latz, E. Western diet and the immune system: an inflammatory connection. Immunity 51, 794–811 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Roden, M. & Shulman, G. I. The integrative biology of type 2 diabetes. Nature 576, 51–60 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Lee, Y. S., Wollam, J. & Olefsky, J. M. An integrated view of immunometabolism. Cell 172, 22–40 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lackey, D. E. & Olefsky, J. M. Regulation of metabolism by the innate immune system. Nat. Rev. Endocrinol. 12, 15–28 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Pollack, R. M., Donath, M. Y., LeRoith, D. & Leibowitz, G. Anti-inflammatory agents in the treatment of diabetes and its vascular complications. Diabetes Care 39, S244–S252 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Ward, Z. J. et al. Projected U.S. state-level prevalence of adult obesity and severe obesity. N. Engl. J. Med. 381, 2440–2450 (2019).

    Article  PubMed  Google Scholar 

  7. Vijay, J. et al. Single-cell analysis of human adipose tissue identifies depot- and disease-specific cell types. Nat. Metab. 2, 97–109 (2020).

    Article  PubMed  Google Scholar 

  8. Jaitin, D. A. et al. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell 178, 686–698.e14 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sun, W. et al. snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis. Nature https://doi.org/10.1038/s41586-020-2856-x (2020).

  10. Dutertre, C.-A. et al. Single-cell analysis of human mononuclear phagocytes reveals subset-defining markers and identifies circulating inflammatory dendritic cells. Immunity 51, 573–589.e8 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Raredon, M. S. B. et al. Single-cell connectomic analysis of adult mammalian lungs. Sci. Adv. 5, eaaw3851 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Stewart, B. J. et al. Spatiotemporal immune zonation of the human kidney. Science 365, 1461–1466 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Merrick, D. et al. Identification of a mesenchymal progenitor cell hierarchy in adipose tissue. Science 364, eaav2501 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Vieira Braga, F. A. et al. A cellular census of human lungs identifies novel cell states in health and in asthma. Nat. Med. 25, 1153–1163 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Vivier, E. et al. Innate lymphoid cells: 10 years on. Cell 174, 1054–1066 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Weizman, O.-E. et al. ILC1 confer early host protection at initial sites of viral infection. Cell 171, 795–808.e12 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nagasawa, M. et al. KLRG1 and NKp46 discriminate subpopulations of human CD117+CRTH2 ILCs biased toward ILC2 or ILC3. J. Exp. Med. 216, 1762–1776 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Dogra, P. et al. Tissue determinants of human NK cell development, function, and residence. Cell 180, 749–763.e13 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhao, J. et al. Single-cell RNA sequencing reveals the heterogeneity of liver-resident immune cells in human. Cell Discov. 6, 22 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cuff, A. O. et al. Eomeshi NK cells in human liver are long-lived and do not recirculate but can be replenished from the circulation. J. Immunol. 197, 4283–4291 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Brestoff, J. R. et al. Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature 519, 242–246 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Mjösberg, J. M. et al. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat. Immunol. 12, 1055–1062 (2011).

    Article  PubMed  CAS  Google Scholar 

  23. Simoni, Y. et al. Human innate lymphoid cell subsets possess tissue-type based heterogeneity in phenotype and frequency. Immunity 46, 148–161 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. O’Sullivan, T. E. et al. Adipose-resident group 1 innate lymphoid cells promote obesity-associated insulin resistance. Immunity 45, 428–441 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Bar-Ephraim, Y. E. et al. Cross-tissue transcriptomic analysis of human secondary lymphoid organ-residing ILC3s reveals a quiescent state in the absence of inflammation. Cell Rep. 21, 823–833 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Villanova, F. et al. Characterization of innate lymphoid cells in human skin and blood demonstrates increase of NKp44+ ILC3 in psoriasis. J. Invest. Dermatol. 134, 984–991 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Lim, A. I. et al. Systemic human ILC precursors provide a substrate for tissue ILC differentiation. Cell 168, 1086–1100.e10 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Bar-Ephraim, Y. E. et al. CD62L is a functional and phenotypic marker for circulating innate lymphoid cell precursors. J. Immunol. 202, 171–182 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Gulati, G. S. et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science 367, 405–411 (2020).

  31. Villani, A.-C. et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Chakarov, S. et al. Two distinct interstitial macrophage populations coexist across tissues in specific subtissular niches. Science 363, eaau0964 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Hill, D. A. et al. Distinct macrophage populations direct inflammatory versus physiological changes in adipose tissue. Proc. Natl Acad. Sci. USA 115, E5096–E5105 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Brown, C. C. et al. Transcriptional basis of mouse and human dendritic cell heterogeneity. Cell 179, 846–863.e24 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kim, J. et al. Silencing CCR2 in macrophages alleviates adipose tissue inflammation and the associated metabolic syndrome in dietary obese mice. Mol. Ther. Nucleic Acids 5, e280 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Weisberg, S. P. et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J. Clin. Invest. 116, 115–124 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Hotamisligil, G. S. Inflammation, metaflammation and immunometabolic disorders. Nature 542, 177–185 (2017).

    Article  CAS  PubMed  Google Scholar 

  38. Hume, D. A. The many alternative faces of macrophage activation. Front. Immunol. 6, 370 (2015).

  39. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Mittelsteadt, K. L. & Campbell, D. J. ICOS signaling in visceral adipose tissue regulatory T cell homeostasis and function. J. Immunol. 202 (Suppl.), 128.4 (2019).

    Google Scholar 

  41. Joller, N. et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity 40, 569–581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Snelgrove, R. J. et al. A critical function for CD200 in lung immune homeostasis and the severity of influenza infection. Nat. Immunol. 9, 1074–1083 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Shafiei-Jahani, P. et al. DR3 stimulation of adipose resident ILC2s ameliorates type 2 diabetes mellitus. Nat. Commun. 11, 4718 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Maazi, H. et al. ICOS:ICOS–ligand interaction is required for type 2 innate lymphoid cell function, homeostasis, and induction of airway hyperreactivity. Immunity 42, 538–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Holmes, T. D. et al. Licensed human natural killer cells aid dendritic cell maturation via TNFSF14/LIGHT. Proc. Natl Acad. Sci. USA 111, E5688–E5696 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kim, H.-M., Jeong, C.-S., Choi, H.-S., Kawada, T. & Yu, R. LIGHT/TNFSF14 enhances adipose tissue inflammatory responses through its interaction with HVEM. FEBS Lett. 585, 579–584 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. O’Rourke, R. W. et al. Systemic inflammation and insulin sensitivity in obese IFN-γ knockout mice. Metabolism 61, 1152–1161 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Böttcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037.e14 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Macdougall, C. E. et al. Visceral adipose tissue immune homeostasis is tegulated by the crosstalk between adipocytes and dendritic cell subsets. Cell Metab. 27, 588–601.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Maier, B. et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 580, 257–262 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Koay, H.-F. et al. A divergent transcriptional landscape underpins the development and functional branching of MAIT cells. Sci. Immunol. 4, eaay6039 (2019).

    Article  CAS  PubMed  Google Scholar 

  53. Szabo, P. A. et al. Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease. Nat. Commun. 10, 4706 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wong, K. L. et al. Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood 118, e16–e31 (2011).

    Article  CAS  PubMed  Google Scholar 

  56. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. https://doi.org/10.1038/s41587-020-0591-3 (2020).

  58. Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the following labs for helpful discussions: A. Freud, T.E.O., M.P. and M. Su. We thank D. Barrett for discarded surgical subcutaneous adipose tissue samples. We thank R. Modlin for assistance with IPA. A.D.H was supported by the Ruth L. Kirschstein National Research Service (award no. AI007323). T.E.O. was supported by the National Institutes of Health (grant nos. P30DK063491 and AI145997). F.M. was supported by the Whitcome Fellowship at UCLA.

Author information

Authors and Affiliations

Authors

Contributions

A.D.H. and T.E.O. designed the study. F.M. and M.P. performed scRNA-seq bioinformatics. A.D.H, YY.W. and R.S. performed all other experiments. A.D.H. and T.E.O. wrote the manuscript.

Corresponding author

Correspondence to Timothy E. O’Sullivan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Jorge Moscat, Sarah Teichmann, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Lineage-associated gene signatures of CD45+ and CD45 SVF cells from healthy human WAT.

a, Dot plot showing selected top differentially expressed marker genes for each cluster, supporting assignment of clusters to compartments shown in Fig. 1b. Color saturation indicates the strength of expression in positive cells, while dot size reflects the percentage of each cell cluster expressing the gene. b, Violin plots showing expression levels of additional cluster markers for the indicated structural cell populations. c, Representative gating strategy for scRNAseq-defined human WAT non-immune cell populations (CD45): Endothelial cell: CD31+, Smooth muscle cell (SMC): CD31CD34CD29, Adipocyte precursor cell (APC): CD31CD34CD29+, Preadipocyte: CD31CD34+CD29intICAM-1+CD26, Interstitial progenitor cell: CD31CD34+CD29intICAM-1+CD26+. d, Density correlation analysis of the indicated non-immune subsets with patient BMI. Line of best fit and 95% confidence intervals are shown for each plot. Each point represents an individual patient. Linear regression and two-tailed Pearson Correlation analysis with 95% confidence intervals were conducted. p < 0.05 was considered significant.

Extended Data Fig. 2 Single cell analysis reveals heterogeneous adaptive lymphocyte populations in healthy human WAT.

a, UMAP plot of 36,601 subclustered human adipose effector lymphocytes from Fig. 1. Cluster analysis yields 9 distinct clusters comprising of T cell subsets, ILCs and NK cells. b, Unbiased heatmap of gene expression of the top 6 unique cluster marker genes for each T cell cluster. Cluster identities are shown above the heatmap. Color saturation indicates the strength of expression. c, Violin plots showing RNA expression of additional cluster markers for the indicated T cell populations. d, Representative gating strategy for scRNAseq-defined human WAT T cell populations (CD45+Lin(CD34+CD19+CD14+): γδT cell (γδ T): TCRγδ+Vα24-Jα18Vα7.2, NKT cell (NKT): TCRγδVα24-Jα18+Vα7.2, MAIT cell (MAIT): TCRγδVα24-Jα18Vα7.2+TCRαβ+, CD8+ T cell (CD8 T): TCRγδVα24-Jα18Vα7.2TCRαβ+CD8α+CD4, CD4+ T cell (CD4 T): TCRγδVα24-Jα18Vα7.2TCRαβ+CD8αCD4+, Regulatory T cell (Treg): TCRγδVα24-Jα18Vα7.2TCRαβ+CD8αCD4+FoxP3+. e, Representative histogram of KLRB1 (CD161) expression on human WAT T cell subsets. f, Representative flow cytometry plot of CD8α and CD4 expression on human WAT γδ T cells. e,f, Data is representative of 4 individual patient samples. g, Density correlation analysis of the indicated T cell subsets with patient BMI. Line of best fit and 95% confidence intervals are shown for each plot. Each point represents an individual patient. Linear regression and two-tailed Pearson Correlation analysis with 95% confidence intervals were conducted. p < 0.05 was considered significant.

Extended Data Fig. 3 Single cell analysis identifies unique human WAT-resident ILCs.

a, Representative sorting strategy for LinCD7+ cell populations based on expression of CD200R1; used in Fig. 2. b, Unsupervised heatmap of the top 5 differentially expressed cluster marker genes for each indicated innate lymphoid cell cluster. Cluster identities are shown above the heatmap. Color saturation indicates the strength of expression. c, Selected UMAP feature plots showing RNA expression of additional cluster markers, based on the UMAP shown in Fig. 2a. d, UMAP of sorted innate lymphoid cell populations denoted by the source of the sorted sample (CD200R1+ vs CD200R1) and patient source classification as lean (red, green) or obese (blue, purple); based on the UMAP shown in Fig. 2a. e, Bar plots showing the proportion of innate lymphoid cells derived from 7 lean (blue) and 5 obese (red) patients.

Extended Data Fig. 4 Flow cytometry analysis of scRNAseq-identified human WAT ILCs.

a,b, Representative gating strategies for the scRNAseq-defined human WAT (a) NK cell and (b) ILC populations identified in Fig. 2. Human WAT ILC populations are defined as CD45+Lin(CD3+TCRαβ+CD19+CD34+CD14+CD5+TCRγδ+EOMES+)CD7+CD200R1+); ILC1: TBET+, ILC2: TBETCRTH2+NKp44, ILC3: TBETCRTH2NKp44+, ILCP-like: TBETCRTH2NKp44CD62L+/–. c, Representative histogram of IFN-γ by human WAT NK cell subsets. Unstim refers to CD200R1 cells cultured without PMA and Ionomycin. d, Representative histograms of TBET, CRTH2, NKp44, and RORC expression on human WAT ILC subsets. e, Analysis of RORC MFI values from human WAT ILC subsets. Each point represents an individual patient (n = 3). ILCP-like: p = 0.0198, ILC1: p = 0.0313, ILC2: p = 0.0194. f, Representative flow cytometry plots of CD7+CD200R1+ cells isolated from human PBMC. g, Density correlation analysis of ILC2 with patient BMI. Each point represents an individual patient. Line of best fit and 95% confidence intervals are shown for the plot. h,i, Density of indicated ILCs by BMI classification. Each point represents an individual patient; (h) n = 8 lean and n = 7 obese patients. i, ILC1: n = 10 lean and n = 7 obese patients; ILC2, ILC3, ILCP-like: n = 5 lean and n = 3 obese patients. ILC2: p = 0.0293, ILC3: p = 0.0094, ILCP-like: p = 0.022. c,d,f, Data is representative of 3 individual patient samples. Samples were compared using two-tailed Student’s t test with Welch’s correction, assuming unequal SD, and data are presented as individual points with the mean ± SEM (*p < 0.05, **p < 0.01). Linear regression and two-tailed Pearson Correlation analysis with 95% confidence intervals were conducted. p < 0.05 was considered significant.

Extended Data Fig. 5 Analysis of ILC1 and ILC3 fate DEGs suggests a clear developmental bifurcation.

Bifurcation heatmap of enriched genes for ILC1 (left), ILCP-like (middle) and ILC3 (right). Color indicates increased (red) or decreased (blue) expression.

Extended Data Fig. 6 Single cell analysis identifies unique myeloid populations within healthy human WAT.

a, Representative sorting strategies for CD11b+CD14+ macrophage and HLA-DR+CD11c+ dendritic cell populations indicated in Fig. 4. b, Heatmap shows the top 4 differentially expressed cluster marker genes for each indicated myeloid cell cluster. Cluster identities are shown above the heatmap. Color saturation indicates the strength of expression. c,d, Violin plots showing RNA expression levels of cluster markers for (c) myeloid (d) and monocyte populations. e, Classical, Intermediate, and Nonclassical Monocyte gene module score analysis for the indicated monocyte populations based on comparison of signature genes for each cell type from previously defined datasets to DEGs within each cluster. f, Neutrophil gene module score analysis for the indicated myeloid populations based on comparison of signature genes for each cell type from previously defined datasets to DEGs within each cluster. g, Bar plots showing the proportion of myeloid cells derived from 7 lean (blue) and 5 obese (red) patients.

Extended Data Fig. 7 Flow cytometry analysis of scRNAseq-identified human WAT macrophage and dendritic cell populations.

a, Representative gating strategy for scRNAseq-defined human WAT macrophage and DC populations identified in Fig. 4. b, Representative histograms of CD64, CD68, CD88, and CD15 expression on human WAT macrophage cell subsets, CD14+ monocytes and CD15+ neutrophils isolated from human PBMC. c, Flow cytometry analysis of endogenous TNF-α production by human WAT macrophage subsets from n = 4 lean, n = 2 overweight, and n = 4 obese patients. Each point represents an individual patient. d, Representative flow cytometry histograms of CLEC10A and FCER1A expression on human WAT DC subsets. e, Density of the indicated DC populations by BMI classification, n = 9 lean and n = 7 obese patients. Each point represents an individual patient (f) Representative flow cytometry plots of human WAT macrophage populations from lean (left) and obese (right) patients. g, Density of the indicated macrophage populations by BMI classification, n = 7 lean and n = 7 obese patients. Each point represents an individual patient. b,d, Data is representative of 3 individual patient samples. Samples were compared using two-tailed Student’s t test with Welch’s correction, assuming unequal SD, and data are presented as individual points with the mean ± SEM (*p < 0.05, **p < 0.01).

Extended Data Fig. 8 Analysis of the DEGs between PVM and IM fates shows a clear bifurcation in gene expression programs.

Bifurcation heatmap of enriched genes for PVM (left), Mo-1 (middle) and IM (right). Color indicates increased (red) or decreased (blue) expression.

Extended Data Fig. 9 Novel human WAT cell types contribute to obesity-associated inflammatory networks.

a, Dot plots showing expression of ligands (left) and receptors (right) in novel human WAT cells; only ligands and receptors from cell types with detected expression (>25%) are shown. Implicated chemokines can be found in the lower panel. Color saturation indicates the strength of expression in positive cells, while dot size reflects the percentage of each cell cluster expressing the gene. b-e, GOSt analysis of obese-enriched differentially expressed genes for the indicated novel human WAT cell types. b, GOSt analysis of trNK. c, GOSt analysis of ILC3. d, GOSt analysis of IM. e, GOSt analysis of cDC2B. Terms were considered statistically significantly enriched if -log10(Padj)<0.05.

Extended Data Fig. 10 Proposed model of cell-cell interactions in healthy lean and obese human WAT.

In lean WAT, IL-15 expression from cDC2A coupled with IL-15RA expression from APCs may support the viability of IL-15RB-expressing ILCs and NK cell populations. CSF1 (M-CSF) expression from APCs and IL-13 from ILC2s likely drives the Mo-1 transition to PVMs, while CSF2 from APCs may support dendritic cell homeostasis. cDC2A-derived CD200 could suppress ILC activation at steady-state. TGFβ1, PDGF, AREG, and GAS6 signaling from dendritic cells and PVM to APCs may promote tissue homeostasis. During obesity, IL-23 from an unknown source could drive the differentiation and accumulation of WAT ILC3s from ILCP-like cells. cDC2B-derived IL-18 and potentially IL-12 might stimulate the production of IFNγ by trNK and ILC1 subsets and contribute to the development of LAM from PVM. Increased CCL2 production from hypoxia-sensing PVM and SMC could recruit circulating Mo-1 into the WAT where MIF, LIF, and IFN-γ signaling from ILCs and NKs, as well as IL-1β, OSM and TNF-α signaling from IM, LAM, and cDC2B could polarize Mo-1 to the IM fate. trNK production of TNFSF14 (LIGHT) may further promote activation of cDC2B and IM. Together, these interactions suggest a cell type specific positive feedback loop whereby accumulation and polarization of WAT-resident lymphoid and myeloid cell types potentiate inflammation during human obesity.

Supplementary information

Reporting Summary

Supplementary Tables 1–18

Supplementary tables 1–18, containing patient data, gene lists, DEGs, IPA and gene set enrichment analyses.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hildreth, A.D., Ma, F., Wong, Y.Y. et al. Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nat Immunol 22, 639–653 (2021). https://doi.org/10.1038/s41590-021-00922-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00922-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing