1932

Abstract

Adipose tissue depots in distinct anatomical locations mediate key aspects of metabolism, including energy storage, nutrient release, and thermogenesis. Although adipocytes make up more than 90% of adipose tissue volume, they represent less than 50% of its cellular content. Here, I review recent advances in genetic lineage tracing and transcriptomics that reveal the identities of the heterogeneous cell populations constituting mouse and human adipose tissues. In addition to mature adipocytes and their progenitors, these include endothelial and various immune cell types that together orchestrate adipose tissue development and functions. One salient finding is the identification of progenitor subtypes that can modulate adipogenic capacity through paracrine mechanisms. Another is the description of fate trajectories of monocyte/macrophages, which can respond maladaptively to nutritional and thermogenic stimuli, leading to metabolic disease. These studies have generated an extraordinary source of publicly available data that can be leveraged to explore commonalities and differences among experimental models, providing new insights into adipose tissues and their role in metabolic disease.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-physiol-031620-095446
2021-02-10
2024-04-18
Loading full text...

Full text loading...

/deliver/fulltext/physiol/83/1/annurev-physiol-031620-095446.html?itemId=/content/journals/10.1146/annurev-physiol-031620-095446&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Ottaviani E, Malagoli D, Franceschi C 2011. The evolution of the adipose tissue: a neglected enigma. Gen. Comp. Endocrinol. 174:1–4
    [Google Scholar]
  2. 2. 
    Gesta S, Tseng YH, Kahn CR 2007. Developmental origin of fat: tracking obesity to its source. Cell 131:242–56
    [Google Scholar]
  3. 3. 
    Gimble J, Guilak F. 2003. Adipose-derived adult stem cells: isolation, characterization, and differentiation potential. Cytotherapy 5:362–69
    [Google Scholar]
  4. 4. 
    Yamamoto N, Akamatsu H, Hasegawa S, Yamada T, Nakata S et al. 2007. Isolation of multipotent stem cells from mouse adipose tissue. J. Dermatol. Sci 48:43–52
    [Google Scholar]
  5. 5. 
    Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW et al. 2001. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 7:211–28
    [Google Scholar]
  6. 6. 
    Ferrante AW Jr 2013. Macrophages, fat, and the emergence of immunometabolism. J. Clin. Investig. 123:4992–93
    [Google Scholar]
  7. 7. 
    Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr 2003. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 112:1796–808
    [Google Scholar]
  8. 8. 
    Eto H, Suga H, Matsumoto D, Inoue K, Aoi N et al. 2009. Characterization of structure and cellular components of aspirated and excised adipose tissue. Plast. Reconstr. Surg. 124:1087–97
    [Google Scholar]
  9. 9. 
    Chen G, Ning B, Shi T 2019. Single-cell RNA-seq technologies and related computational data analysis. Front. Genet. 10:317
    [Google Scholar]
  10. 10. 
    Efremova M, Vento-Tormo R, Park JE, Teichmann SA, James KR 2020. Immunology in the era of single-cell technologies. Annu. Rev. Immunol. 38:727–57
    [Google Scholar]
  11. 11. 
    Natarajan KN, Miao Z, Jiang M, Huang X, Zhou H et al. 2019. Comparative analysis of sequencing technologies for single-cell transcriptomics. Genome Biol 20:70
    [Google Scholar]
  12. 12. 
    Shinde P, Mohan L, Kumar A, Dey K, Maddi A et al. 2018. Current trends of microfluidic single-cell technologies. Int. J. Mol. Sci. 19:3143
    [Google Scholar]
  13. 13. 
    Schaum N, Karkanias J, Neff NF, May AP, Quake SR et al. 2018. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. . Nature 562:367–72
    [Google Scholar]
  14. 14. 
    Napolitano L. 1963. The differentiation of white adipose cells. an electron microscope study. J. Cell Biol. 18:663–79
    [Google Scholar]
  15. 15. 
    Napolitano L, Gagne HT. 1963. Lipid-depleted white adipose cells: an electron microscope study. Anat. Rec. 147:273–93
    [Google Scholar]
  16. 16. 
    Napolitano L, McNary JE, Kloep LP 1965. The release of free fatty acids from brown and white adipose tissues after incubation with ACTH or epinephrine. Metabolism 14:1076–83
    [Google Scholar]
  17. 17. 
    Napolitano LM. 1965. Observations on the fine structure of adipose cells. Ann. N. Y. Acad. Sci. 131:34–42
    [Google Scholar]
  18. 18. 
    Slavin BG, Ballard KW. 1978. Morphological studies on the adrenergic innervation of white adipose tissue. Anat. Rec. 191:377–89
    [Google Scholar]
  19. 19. 
    Alexander G, Stevens D. 1980. Sympathetic innervation and the development of structure and function of brown adipose tissue: studies on lambs chemically sympathectomized in utero with 6-hydroxydopamine. J. Dev. Physiol. 2:119–37
    [Google Scholar]
  20. 20. 
    Davis V. 1980. The structure and function of brown adipose tissue in the neonate. J. Obstet. Gynecol. Neonatal Nurs. 9:368–72
    [Google Scholar]
  21. 21. 
    Hausman GJ, Richardson RL. 1983. Cellular and vascular development in immature rat adipose tissue. J. Lipid Res. 24:522–32
    [Google Scholar]
  22. 22. 
    Pond CM, Mattacks CA. 1985. Cellular structure of adipose tissue in birds. J. Morphol. 185:195–202
    [Google Scholar]
  23. 23. 
    Marchington JM, Mattacks CA, Pond CM 1989. Adipose tissue in the mammalian heart and pericardium: structure, foetal development and biochemical properties. Comp. Biochem. Physiol. B 94:225–32
    [Google Scholar]
  24. 24. 
    Cinti S. 1999. Adipose tissues and obesity. Ital. J. Anat. Embryol. 104:37–51
    [Google Scholar]
  25. 25. 
    Suter ER. 1969. The fine structure of brown adipose tissue. I. Cold-induced changes in the rat. J. Ultrastruct. Res. 26:216–41
    [Google Scholar]
  26. 26. 
    Salans LB, Dougherty JW. 1971. The effect of insulin upon glucose metabolism by adipose cells of different size. Influence of cell lipid and protein content, age, and nutritional state. J. Clin. Investig. 50:1399–410
    [Google Scholar]
  27. 27. 
    Gliemann J, Vinten J. 1974. Lipogenesis and insulin sensitivity of single fat cells. J. Physiol. 236:499–516
    [Google Scholar]
  28. 28. 
    Varlamov O, Somwar R, Cornea A, Kievit P, Grove KL, Roberts CT Jr 2010. Single-cell analysis of insulin-regulated fatty acid uptake in adipocytes. Am. J. Physiol. Endocrinol. Metab. 299:E486–96
    [Google Scholar]
  29. 29. 
    Hagberg CE, Li Q, Kutschke M, Bhowmick D, Kiss E et al. 2018. Flow cytometry of mouse and human adipocytes for the analysis of browning and cellular heterogeneity. Cell Rep 24:2746–56.e5
    [Google Scholar]
  30. 30. 
    Sanchez-Gurmaches J, Guertin DA. 2014. Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed. Nat. Commun. 5:4099
    [Google Scholar]
  31. 31. 
    Chi J, Wu Z, Choi CHJ, Nguyen L, Tegegne S et al. 2018. Three-dimensional adipose tissue imaging reveals regional variation in beige fat biogenesis and PRDM16-dependent sympathetic neurite density. Cell Metab 27:226–36.e3
    [Google Scholar]
  32. 32. 
    Ebke LA, Nestor-Kalinoski AL, Slotterbeck BD, Al-Dieri AG, Ghosh-Lester S et al. 2014. Tight association between macrophages and adipocytes in obesity: implications for adipocyte preparation. Obesity 22:1246–55
    [Google Scholar]
  33. 33. 
    Tran KV, Gealekman O, Frontini A, Zingaretti MC, Morroni M et al. 2012. The vascular endothelium of the adipose tissue gives rise to both white and brown fat cells. Cell Metab 15:222–29
    [Google Scholar]
  34. 34. 
    Rajbhandari P, Arneson D, Hart SK, Ahn IS, Diamante G et al. 2019. Single cell analysis reveals immune cell–adipocyte crosstalk regulating the transcription of thermogenic adipocytes. eLife 8:e49501
    [Google Scholar]
  35. 35. 
    Janke J, Engeli S, Gorzelniak K, Luft FC, Sharma AM 2002. Mature adipocytes inhibit in vitro differentiation of human preadipocytes via angiotensin type 1 receptors. Diabetes 51:1699–707
    [Google Scholar]
  36. 36. 
    Min SY, Kady J, Nam M, Rojas-Rodriguez R, Berkenwald A et al. 2016. Human ‘brite/beige’ adipocytes develop from capillary networks, and their implantation improves metabolic homeostasis in mice. Nat. Med. 22:312–18
    [Google Scholar]
  37. 37. 
    Rojas-Rodriguez R, Lujan-Hernandez J, Min SY, DeSouza T, Teebagy P et al. 2019. Generation of functional human adipose tissue in mice from primed progenitor cells. Tissue Eng. A 25:842–54
    [Google Scholar]
  38. 38. 
    Song A, Dai W, Jang MJ, Medrano L, Li Z et al. 2020. Low- and high-thermogenic brown adipocyte subpopulations coexist in murine adipose tissue. J. Clin. Investig. 130:247–57
    [Google Scholar]
  39. 39. 
    Lee KY, Luong Q, Sharma R, Dreyfuss JM, Ussar S, Kahn CR 2019. Developmental and functional heterogeneity of white adipocytes within a single fat depot. EMBO J 38:e99291
    [Google Scholar]
  40. 40. 
    Chau YY, Bandiera R, Serrels A, Martinez-Estrada OM, Qing W et al. 2014. Visceral and subcutaneous fat have different origins and evidence supports a mesothelial source. Nat. Cell Biol. 16:367–75
    [Google Scholar]
  41. 41. 
    Czech MP. 2020. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol. Metab. 34:27–42
    [Google Scholar]
  42. 42. 
    Spaethling JM, Sanchez-Alavez M, Lee J, Xia FC, Dueck H et al. 2016. Single-cell transcriptomics and functional target validation of brown adipocytes show their complex roles in metabolic homeostasis. FASEB J 30:81–92
    [Google Scholar]
  43. 43. 
    Kazak L, Chouchani ET, Jedrychowski MP, Erickson BK, Shinoda K et al. 2015. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell 163:643–55
    [Google Scholar]
  44. 44. 
    Kazak L, Chouchani ET, Lu GZ, Jedrychowski MP, Bare CJ et al. 2017. Genetic depletion of adipocyte creatine metabolism inhibits diet-induced thermogenesis and drives obesity. Cell Metab 26:660–71.e3
    [Google Scholar]
  45. 45. 
    Sun W, Dong H, Balaz M, Slyper M, Drokhlyansky E et al. 2020. Single-nucleus RNA-Seq reveals a new type of brown adipocyte regulating thermogenesis. bioRxiv 2020.01.20.890327. https://doi.org/10.1101/2020.01.20.890327
    [Crossref]
  46. 46. 
    Lake BB, Codeluppi S, Yung YC, Gao D, Chun J et al. 2017. A comparative strategy for single-nucleus and single-cell transcriptomes confirms accuracy in predicted cell-type expression from nuclear RNA. Sci. Rep. 7:6031
    [Google Scholar]
  47. 47. 
    Wu H, Kirita Y, Donnelly EL, Humphreys BD 2019. Advantages of single-nucleus over single-cell RNA sequencing of adult kidney: rare cell types and novel cell states revealed in fibrosis. J. Am. Soc. Nephrol. 30:23–32
    [Google Scholar]
  48. 48. 
    Roesler A, Kazak L. 2020. UCP1-independent thermogenesis. Biochem. J. 477:709–25
    [Google Scholar]
  49. 49. 
    Ikeda K, Kang Q, Yoneshiro T, Camporez JP, Maki H et al. 2017. UCP1-independent signaling involving SERCA2b-mediated calcium cycling regulates beige fat thermogenesis and systemic glucose homeostasis. Nat. Med. 23:1454–65
    [Google Scholar]
  50. 50. 
    Zheng B, Cao B, Li G, Huard J 2006. Mouse adipose-derived stem cells undergo multilineage differentiation in vitro but primarily osteogenic and chondrogenic differentiation in vivo. Tissue Eng 12:1891–901
    [Google Scholar]
  51. 51. 
    Tang W, Zeve D, Suh JM, Bosnakovski D, Kyba M et al. 2008. White fat progenitor cells reside in the adipose vasculature. Science 322:583–86
    [Google Scholar]
  52. 52. 
    Rodeheffer MS, Birsoy K, Friedman JM 2008. Identification of white adipocyte progenitor cells in vivo. Cell 135:240–49
    [Google Scholar]
  53. 53. 
    Smith NC, Fairbridge NA, Pallegar NK, Christian SL 2015. Dynamic upregulation of CD24 in pre-adipocytes promotes adipogenesis. Adipocyte 4:89–100
    [Google Scholar]
  54. 54. 
    Schwalie PC, Dong H, Zachara M, Russeil J, Alpern D et al. 2018. A stromal cell population that inhibits adipogenesis in mammalian fat depots. Nature 559:103–8
    [Google Scholar]
  55. 55. 
    Min SY, Desai A, Yang Z, Sharma A, DeSouza T et al. 2019. Diverse repertoire of human adipocyte subtypes develops from transcriptionally distinct mesenchymal progenitor cells. PNAS 116:17970–79
    [Google Scholar]
  56. 56. 
    Burl RB, Ramseyer VD, Rondini EA, Pique-Regi R, Lee YH, Granneman JG 2018. Deconstructing adipogenesis induced by β3-adrenergic receptor activation with single-cell expression profiling. Cell Metab 28:300–9.e4
    [Google Scholar]
  57. 57. 
    Merrick D, Sakers A, Irgebay Z, Okada C, Calvert C et al. 2019. Identification of a mesenchymal progenitor cell hierarchy in adipose tissue. Science 364:eaav2501
    [Google Scholar]
  58. 58. 
    Rennert RC, Januszyk M, Sorkin M, Rodrigues M, Maan ZN et al. 2016. Microfluidic single-cell transcriptional analysis rationally identifies novel surface marker profiles to enhance cell-based therapies. Nat. Commun. 7:11945
    [Google Scholar]
  59. 59. 
    Hepler C, Shan B, Zhang Q, Henry GH, Shao M et al. 2018. Identification of functionally distinct fibro-inflammatory and adipogenic stromal subpopulations in visceral adipose tissue of adult mice. eLife 7:e39636
    [Google Scholar]
  60. 60. 
    Vishvanath L, MacPherson KA, Hepler C, Wang QA, Shao M et al. 2016. Pdgfrβ+ mural preadipocytes contribute to adipocyte hyperplasia induced by high-fat-diet feeding and prolonged cold exposure in adult mice. Cell Metab 23:350–59
    [Google Scholar]
  61. 61. 
    Marcelin G, Ferreira A, Liu Y, Atlan M, Aron-Wisnewsky J et al. 2017. A PDGFRα-mediated switch toward CD9high adipocyte progenitors controls obesity-induced adipose tissue fibrosis. Cell Metab 25:673–85
    [Google Scholar]
  62. 62. 
    Kim YJ, Yu JM, Joo HJ, Kim HK, Cho HH et al. 2007. Role of CD9 in proliferation and proangiogenic action of human adipose-derived mesenchymal stem cells. Pflügers Arch 455:283–96
    [Google Scholar]
  63. 63. 
    Cho DS, Lee B, Doles JD 2019. Refining the adipose progenitor cell landscape in healthy and obese visceral adipose tissue using single-cell gene expression profiling. Life Sci. Alliance 2: https://doi.org/10.26508/lsa.201900561
    [Crossref] [Google Scholar]
  64. 64. 
    Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI et al. 2002. Human adipose tissue is a source of multipotent stem cells. Mol. Biol. Cell 13:4279–95
    [Google Scholar]
  65. 65. 
    Schaffler A, Buchler C. 2007. Concise review: adipose tissue-derived stromal cells—basic and clinical implications for novel cell-based therapies. Stem Cells 25:818–27
    [Google Scholar]
  66. 66. 
    Osathanon T, Subbalekha K, Sastravaha P, Pavasant P 2012. Notch signalling inhibits the adipogenic differentiation of single-cell-derived mesenchymal stem cell clones isolated from human adipose tissue. Cell Biol. Int. 36:1161–70
    [Google Scholar]
  67. 67. 
    Acosta JR, Joost S, Karlsson K, Ehrlund A, Li X et al. 2017. Single cell transcriptomics suggest that human adipocyte progenitor cells constitute a homogeneous cell population. Stem Cell Res. Ther. 8:250
    [Google Scholar]
  68. 68. 
    Vijay J, Gauthier MF, Biswell RL, Louiselle DA, Johnston JJ et al. 2020. Single-cell analysis of human adipose tissue identifies depot and disease specific cell types. Nat. Metab. 2:97–109
    [Google Scholar]
  69. 69. 
    Tchkonia T, Lenburg M, Thomou T, Giorgadze N, Frampton G et al. 2007. Identification of depot-specific human fat cell progenitors through distinct expression profiles and developmental gene patterns. Am. J. Physiol. Endocrinol. Metab. 292:E298–307
    [Google Scholar]
  70. 70. 
    Zhang Z, Shao M, Hepler C, Zi Z, Zhao S et al. 2019. Dermal adipose tissue has high plasticity and undergoes reversible dedifferentiation in mice. J. Clin. Investig. 129:5327–42
    [Google Scholar]
  71. 71. 
    Maurizi G, Poloni A, Mattiucci D, Santi S, Maurizi A et al. 2017. Human white adipocytes convert into “rainbow” adipocytes in vitro. J. Cell Physiol. 232:2887–99
    [Google Scholar]
  72. 72. 
    Wolock SL, Krishnan I, Tenen DE, Matkins V, Camacho V et al. 2019. Mapping distinct bone marrow niche populations and their differentiation paths. Cell Rep 28:302–11.e5
    [Google Scholar]
  73. 73. 
    Zhong L, Yao L, Tower RJ, Wei Y, Miao Z et al. 2020. Single cell transcriptomics identifies a unique adipose lineage cell population that regulates bone marrow environment. eLife 9:e54695
    [Google Scholar]
  74. 74. 
    Baccin C, Al-Sabah J, Velten L, Helbling PM, Grunschlager F et al. 2020. Combined single-cell and spatial transcriptomics reveal the molecular, cellular and spatial bone marrow niche organization. Nat. Cell Biol. 22:38–48
    [Google Scholar]
  75. 75. 
    Chang L, Milton H, Eitzman DT, Chen YE 2013. Paradoxical roles of perivascular adipose tissue in atherosclerosis and hypertension. Circ. J. 77:11–18
    [Google Scholar]
  76. 76. 
    Chang L, Villacorta L, Li R, Hamblin M, Xu W et al. 2012. Loss of perivascular adipose tissue on peroxisome proliferator-activated receptor-gamma deletion in smooth muscle cells impairs intravascular thermoregulation and enhances atherosclerosis. Circulation 126:1067–78
    [Google Scholar]
  77. 77. 
    Gu W, Nowak WN, Xie Y, Le Bras A, Hu Y et al. 2019. Single-cell RNA-sequencing and metabolomics analyses reveal the contribution of perivascular adipose tissue stem cells to vascular remodeling. Arterioscler. Thromb. Vasc. Biol. 39:2049–66
    [Google Scholar]
  78. 78. 
    Pan XX, Ruan CC, Liu XY, Kong LR, Ma Y et al. 2019. Perivascular adipose tissue-derived stromal cells contribute to vascular remodeling during aging. Aging Cell 18:e12969
    [Google Scholar]
  79. 79. 
    Tran KV, Fitzgibbons T, Min SY, DeSouza T, Corvera S 2018. Distinct adipocyte progenitor cells are associated with regional phenotypes of perivascular aortic fat in mice. Mol. Metab. 9:199–206
    [Google Scholar]
  80. 80. 
    Lindhorst A, Bechmann I, Gericke M 2020. Unspecific DNA recombination in AdipoqCre-ERT2-mediated knockout approaches in transgenic mice is sex-, age- and genotype-dependent. Adipocyte 9:1–6
    [Google Scholar]
  81. 81. 
    Jiang Y, Berry DC, Jo A, Tang W, Arpke RW et al. 2017. A PPARγ transcriptional cascade directs adipose progenitor cell-niche interaction and niche expansion. Nat. Commun. 8:15926
    [Google Scholar]
  82. 82. 
    Park J, Kim M, Sun K, An YA, Gu X, Scherer PE 2017. VEGF-A-expressing adipose tissue shows rapid beiging and enhanced survival after transplantation and confers IL-4-independent metabolic improvements. Diabetes 66:1479–90
    [Google Scholar]
  83. 83. 
    Hill DA, Lim HW, Kim YH, Ho WY, Foong YH et al. 2018. Distinct macrophage populations direct inflammatory versus physiological changes in adipose tissue. PNAS 115:E5096–105
    [Google Scholar]
  84. 84. 
    Sharma M, Schlegel M, Brown EJ, Sansbury BE, Weinstock A et al. 2019. Netrin-1 alters adipose tissue macrophage fate and function in obesity. Immunometabolism 1:e190010
    [Google Scholar]
  85. 85. 
    Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B et al. 2019. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell 178:686–98.e14
    [Google Scholar]
  86. 86. 
    Weinstock A, Brown EJ, Garabedian ML, Pena S, Sharma M et al. 2019. Single-cell RNA sequencing of visceral adipose tissue leukocytes reveals that caloric restriction following obesity promotes the accumulation of a distinct macrophage population with features of phagocytic cells. Immunometabolism 1:e190008
    [Google Scholar]
  87. 87. 
    Li C, Menoret A, Farragher C, Ouyang Z, Bonin C et al. 2019. Single cell transcriptomics-based MacSpectrum reveals novel macrophage activation signatures in diseases. JCI Insight 5:e126453
    [Google Scholar]
  88. 88. 
    Liu LF, Craig CM, Tolentino LL, Choi O, Morton J et al. 2017. Adipose tissue macrophages impair preadipocyte differentiation in humans. PLOS ONE 12:e0170728
    [Google Scholar]
  89. 89. 
    Cipolletta D, Kolodin D, Benoist C, Mathis D 2011. Tissular T(regs): a unique population of adipose-tissue-resident Foxp3+CD4+ T cells that impacts organismal metabolism. Semin. Immunol. 23:431–37
    [Google Scholar]
  90. 90. 
    Vasanthakumar A, Chisanga D, Blume J, Gloury R, Britt K et al. 2020. Sex-specific adipose tissue imprinting of regulatory T cells. Nature 579:581–85
    [Google Scholar]
  91. 91. 
    Kalin S, Becker M, Ott VB, Serr I, Hosp F et al. 2017. A Stat6/Pten axis links regulatory T cells with adipose tissue function. Cell Metab 26:475–92.e7
    [Google Scholar]
  92. 92. 
    Setiady YY, Coccia JA, Park PU 2010. In vivo depletion of CD4+FOXP3+ Treg cells by the PC61 anti-CD25 monoclonal antibody is mediated by FcγRIII+ phagocytes. Eur. J. Immunol. 40:780–86
    [Google Scholar]
  93. 93. 
    Crewe C, Joffin N, Rutkowski JM, Kim M, Zhang F et al. 2018. An endothelial-to-adipocyte extracellular vesicle axis governed by metabolic state. Cell 175:695–708.e13
    [Google Scholar]
  94. 94. 
    Romacho T, Sell H, Indrakusuma I, Roehrborn D, Castaneda TR et al. 2020. DPP4 deletion in adipose tissue improves hepatic insulin sensitivity in diet-induced obesity. Am. J. Physiol. Endocrinol. Metab. 318:E59099
    [Google Scholar]
  95. 95. 
    Zhong J, Rao X, Rajagopalan S 2013. An emerging role of dipeptidyl peptidase 4 (DPP4) beyond glucose control: potential implications in cardiovascular disease. Atherosclerosis 226:305–14
    [Google Scholar]
  96. 96. 
    Glastonbury CA, Couto Alves A, El-Sayed Moustafa JS, Small KS 2019. Cell-type heterogeneity in adipose tissue is associated with complex traits and reveals disease-relevant cell-specific eQTLs. Am. J. Hum. Genet. 104:1013–24
    [Google Scholar]
/content/journals/10.1146/annurev-physiol-031620-095446
Loading
/content/journals/10.1146/annurev-physiol-031620-095446
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error