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.

  • Review Article
  • Published:

The obese adipose tissue microenvironment in cancer development and progression

Abstract

Obesity is associated with both increased cancer incidence and progression in multiple tumour types, and is estimated to contribute to up to 20% of cancer-related deaths. These associations are driven, in part, by metabolic and inflammatory changes in adipose tissue that disrupt physiological homeostasis both within local tissues and systemically. However, the mechanisms underlying the obesity–cancer relationship are poorly understood. In this Review, we describe how the adipose tissue microenvironment (ATME) evolves during body-weight gain, and how these changes might influence tumour initiation and progression. We focus on multiple facets of ATME physiology, including inflammation, vascularity and fibrosis, and discuss therapeutic interventions that have the potential to normalize the ATME, which might be translationally relevant for cancer prevention and therapy. Given that the prevalence of obesity is increasing on an international scale, translational research initiatives are urgently needed to provide mechanistic explanations for the obesity–cancer relationship, and how to best identify high-risk individuals without relying on crude measures, such as BMI.

Key points

  • Obesity is associated with increased cancer incidence and mortality.

  • Substantial changes occur within the adipose tissue microenvironment (ATME) with body-weight gain.

  • Metabolic and inflammatory changes related to the obese ATME contribute to cancer development and progression.

  • Targeting adipose tissue dysfunction through pharmacological or lifestyle interventions might be useful for the prevention and treatment of cancer.

  • Given the limitations of BMI as a measurement of adiposity, finding novel ways to identify individuals who are metabolically unhealthy with excess adipose tissue will be critical to pinpoint those at risk who might benefit from weight loss or other personalized interventions.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Major adipose depots and anatomical locations in adult humans and mice.
Fig. 2: Evolution of the adipose tissue microenvironment during obesity.
Fig. 3: Interactions between tumour cells and cells within the obese adipose tissue microenvironment.
Fig. 4: Vascular inflammation and fibrosis in the obese adipose tissue microenvironment.

Similar content being viewed by others

References

  1. James, W. P. T. & McPherson, K. The costs of overweight. Lancet Public Health 2, e203–e204 (2017).

    PubMed  Google Scholar 

  2. Malik, V. S., Willett, W. C. & Hu, F. B. Global obesity: trends, risk factors and policy implications. Nat. Rev. Endocrinol. 9, 13–27 (2013).

    PubMed  Google Scholar 

  3. NCD Risk Factor Collaboration. Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2 million participants. Lancet 387, 1377–1396 (2016).

    Google Scholar 

  4. Ng, M. et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 384, 766–781 (2014).

    PubMed  PubMed Central  Google Scholar 

  5. Hales, C. M., Fryar, C. D., Carroll, M. D., Freedman, D. S. & Ogden, C. L. Trends in obesity and severe obesity prevalence in US youth and adults by sex and age, 2007–2008 to 2015–2016. JAMA 319, 1723–1725 (2018).

    PubMed  PubMed Central  Google Scholar 

  6. Cawley, J. & Meyerhoefer, C. The medical care costs of obesity: an instrumental variables approach. J. Health Econ. 31, 219–230 (2012).

    PubMed  Google Scholar 

  7. Lauby-Secretan, B. et al. Body fatness and cancer — viewpoint of the IARC Working Group. N. Engl. J. Med. 375, 794–798 (2016).

    PubMed  PubMed Central  Google Scholar 

  8. Calle, E. E., Rodriguez, C., Walker-Thurmond, K. & Thun, M. J. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U. S. adults. N. Engl. J. Med. 348, 1625–1638 (2003).

    PubMed  Google Scholar 

  9. Ligibel, J. A. et al. American Society of Clinical Oncology position statement on obesity and cancer. J. Clin. Oncol. 32, 3568–3574 (2014).

    PubMed  PubMed Central  Google Scholar 

  10. Calle, E. E. & Kaaks, R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 4, 579–591 (2004).

    CAS  PubMed  Google Scholar 

  11. Tao, W. & Lagergren, J. Clinical management of obese patients with cancer. Nat. Rev. Clin. Oncol. 10, 519–533 (2013).

    CAS  PubMed  Google Scholar 

  12. Rosenquist, K. J. et al. Fat quality and incident cardiovascular disease, all-cause mortality, and cancer mortality. J. Clin. Endocrinol. Metab. 100, 227–234 (2015).

    CAS  PubMed  Google Scholar 

  13. Iyengar, N. M. et al. Systemic correlates of white adipose tissue inflammation in early-stage breast cancer. Clin. Cancer Res. 22, 2283–2289 (2016).

    CAS  PubMed  Google Scholar 

  14. Rosen, E. D. & Spiegelman, B. M. What we talk about when we talk about fat. Cell 156, 20–44 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Wajchenberg, B. L. Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr. Rev. 21, 697–738 (2000).

    CAS  PubMed  Google Scholar 

  16. Neeland, I. J. et al. Associations of visceral and abdominal subcutaneous adipose tissue with markers of cardiac and metabolic risk in obese adults. Obesity 21, E439–E447 (2013).

    CAS  PubMed  Google Scholar 

  17. McLaughlin, T., Lamendola, C., Liu, A. & Abbasi, F. Preferential fat deposition in subcutaneous versus visceral depots is associated with insulin sensitivity. J. Clin. Endocrinol. Metab. 96, E1756–E1760 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Fox, C. S. et al. Abdominal visceral and subcutaneous adipose tissue compartments: association with metabolic risk factors in the Framingham Heart Study. Circulation 116, 39–48 (2007).

    PubMed  Google Scholar 

  19. Park, J., Morley, T. S., Kim, M., Clegg, D. J. & Scherer, P. E. Obesity and cancer — mechanisms underlying tumour progression and recurrence. Nat. Rev. Endocrinol. 10, 455–465 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Khandekar, M. J., Cohen, P. & Spiegelman, B. M. Molecular mechanisms of cancer development in obesity. Nat. Rev. Cancer 11, 886–895 (2011).

    CAS  PubMed  Google Scholar 

  21. Font-Burgada, J., Sun, B. & Karin, M. Obesity and cancer: the oil that feeds the flame. Cell Metab. 23, 48–62 (2016).

    CAS  PubMed  Google Scholar 

  22. Lengyel, E., Makowski, L., DiGiovanni, J. & Kolonin, M. G. Cancer as a matter of fat: the crosstalk between adipose tissue and tumors. Trends Cancer 4, 374–384 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Olson, O. C., Quail, D. F. & Joyce, J. A. Obesity and the tumor microenvironment. Science 358, 1130–1131 (2017).

    CAS  PubMed  Google Scholar 

  24. Borrud, L. G. et al. Body composition data for individuals 8 years of age and older: U.S. population, 1999–2004. Vital Health Stat. 250, 1–87 (2010).

    Google Scholar 

  25. Flegal, K. M. et al. Comparisons of percentage body fat, body mass index, waist circumference, and waist-stature ratio in adults. Am. J. Clin. Nutr. 89, 500–508 (2009).

    CAS  PubMed  Google Scholar 

  26. Brestoff, J. R. & Artis, D. Immune regulation of metabolic homeostasis in health and disease. Cell 161, 146–160 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Howe, L. R., Subbaramaiah, K., Hudis, C. A. & Dannenberg, A. J. Molecular pathways: adipose inflammation as a mediator of obesity-associated cancer. Clin. Cancer Res. 19, 6074–6083 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Crewe, C., An, Y. A. & Scherer, P. E. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J. Clin. Invest. 127, 74–82 (2017).

    PubMed  PubMed Central  Google Scholar 

  29. Osborn, O. & Olefsky, J. M. The cellular and signaling networks linking the immune system and metabolism in disease. Nat. Med. 18, 363–374 (2012).

    CAS  PubMed  Google Scholar 

  30. Reilly, S. M. & Saltiel, A. R. Adapting to obesity with adipose tissue inflammation. Nat. Rev. Endocrinol. 13, 633–643 (2017).

    CAS  PubMed  Google Scholar 

  31. Kanneganti, T. D. & Dixit, V. D. Immunological complications of obesity. Nat. Immunol. 13, 707–712 (2012).

    CAS  PubMed  Google Scholar 

  32. Giordano, A. et al. Obese adipocytes show ultrastructural features of stressed cells and die of pyroptosis. J. Lipid Res. 54, 2423–2436 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Cinti, S. et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res. 46, 2347–2355 (2005).

    CAS  PubMed  Google Scholar 

  34. Wen, H. et al. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 12, 408–415 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhou, R., Tardivel, A., Thorens, B., Choi, I. & Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat. Immunol. 11, 136–140 (2010).

    CAS  PubMed  Google Scholar 

  36. Mariathasan, S. et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 440, 228–232 (2006).

    CAS  PubMed  Google Scholar 

  37. Vandanmagsar, B. et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med. 17, 179–188 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Duewell, P. et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464, 1357–1361 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Youm, Y. H. et al. The Nlrp3 inflammasome promotes age-related thymic demise and immunosenescence. Cell Rep. 1, 56–68 (2012).

    CAS  PubMed  Google Scholar 

  40. Weisberg, S. P. et al. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest. 112, 1796–1808 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Nagareddy, P. R. et al. Adipose tissue macrophages promote myelopoiesis and monocytosis in obesity. Cell Metab. 19, 821–835 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Amano, S. U. et al. Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation. Cell Metab. 19, 162–171 (2014).

    CAS  PubMed  Google Scholar 

  43. Braune, J. et al. IL-6 regulates M2 polarization and local proliferation of adipose tissue macrophages in obesity. J. Immunol. 198, 2927–2934 (2017).

    CAS  PubMed  Google Scholar 

  44. Jenkins, S. J. et al. Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science 332, 1284–1288 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Haka, A. S. et al. Exocytosis of macrophage lysosomes leads to digestion of apoptotic adipocytes and foam cell formation. J. Lipid Res. 57, 980–992 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Kolb, R. et al. Obesity-associated NLRC4 inflammasome activation drives breast cancer progression. Nat. Commun. 7, 13007 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Arkan, M. C. et al. IKK-beta links inflammation to obesity-induced insulin resistance. Nat. Med. 11, 191–198 (2005).

    CAS  PubMed  Google Scholar 

  48. Solinas, G. et al. JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metab. 6, 386–397 (2007).

    CAS  PubMed  Google Scholar 

  49. Iyengar, N. M. et al. Metabolic obesity, adipose inflammation and elevated breast aromatase in women with normal body mass index. Cancer Prev. Res. 10, 235–243 (2017).

    CAS  Google Scholar 

  50. Koru-Sengul, T. et al. Breast cancers from black women exhibit higher numbers of immunosuppressive macrophages with proliferative activity and of crown-like structures associated with lower survival compared to non-black Latinas and Caucasians. Breast Cancer Res. Treat. 158, 113–126 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Carter, J. M. et al. Macrophagic “crown-like structures” are associated with an increased risk of breast cancer in benign breast disease. Cancer Prev. Res. 11, 113–119 (2018).

    Google Scholar 

  52. Diaz-Cruz, E. S., Sugimoto, Y., Gallicano, G. I., Brueggemeier, R. W. & Furth, P. A. Comparison of increased aromatase versus ERalpha in the generation of mammary hyperplasia and cancer. Cancer Res. 71, 5477–5487 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Gucalp, A. et al. Periprostatic adipose inflammation is associated with high-grade prostate cancer. Prostate Cancer Prostatic Dis. 20, 418–423 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Iyengar, N. M. et al. White adipose tissue inflammation and cancer-specific survival in patients with squamous cell carcinoma of the oral tongue. Cancer 122, 3794–3802 (2016).

    PubMed  Google Scholar 

  55. Quail, D. F. & Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Patsouris, D. et al. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab. 8, 301–309 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Nguyen, M. T. et al. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via toll-like receptors 2 and 4 and JNK-dependent pathways. J. Biol. Chem. 282, 35279–35292 (2007).

    CAS  PubMed  Google Scholar 

  58. Shaul, M. E., Bennett, G., Strissel, K. J., Greenberg, A. S. & Obin, M. S. Dynamic, M2-like remodeling phenotypes of CD11c+ adipose tissue macrophages during high-fat diet–induced obesity in mice. Diabetes 59, 1171–1181 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Franklin, R. A. et al. The cellular and molecular origin of tumor-associated macrophages. Science 344, 921–925 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Tkach, M. & Thery, C. Communication by extracellular vesicles: where we are and where we need to go. Cell 164, 1226–1232 (2016).

    CAS  PubMed  Google Scholar 

  62. Xu, X. et al. Obesity activates a program of lysosomal-dependent lipid metabolism in adipose tissue macrophages independently of classic activation. Cell Metab. 18, 816–830 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Kratz, M. et al. Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metab. 20, 614–625 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Wu, D. et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science 332, 243–247 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Coffelt, S. B., Wellenstein, M. D. & de Visser, K. E. Neutrophils in cancer: neutral no more. Nat. Rev. Cancer 16, 431–446 (2016).

    CAS  PubMed  Google Scholar 

  66. Talukdar, S. et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat. Med. 18, 1407–1412 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Liu, J. et al. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat. Med. 15, 940–945 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Olson, O. C. & Joyce, J. A. Cysteine cathepsin proteases: regulators of cancer progression and therapeutic response. Nat. Rev. Cancer 15, 712–729 (2015).

    CAS  PubMed  Google Scholar 

  69. Xia, S. et al. Gr-1+ CD11b+ myeloid-derived suppressor cells suppress inflammation and promote insulin sensitivity in obesity. J. Biol. Chem. 286, 23591–23599 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Clements, V. K. et al. Frontline science: high fat diet and leptin promote tumor progression by inducing myeloid-derived suppressor cells. J. Leukoc. Biol. 103, 395–407 (2018).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Yang, H. et al. Obesity accelerates thymic aging. Blood 114, 3803–3812 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Yang, H. et al. Obesity increases the production of proinflammatory mediators from adipose tissue T cells and compromises TCR repertoire diversity: implications for systemic inflammation and insulin resistance. J. Immunol. 185, 1836–1845 (2010).

    CAS  PubMed  Google Scholar 

  74. Canter, R. J. et al. Obesity results in higher PD-1-mediated T cell suppression but greater T cell effector functions following blockade. J. Clin. Oncol. 36, S65 (2018).

    Google Scholar 

  75. McQuade, J. L. et al. Association of body-mass index and outcomes in patients with metastatic melanoma treated with targeted therapy, immunotherapy, or chemotherapy: a retrospective, multicohort analysis. Lancet Oncol. 19, 310–322 (2018).

    PubMed  PubMed Central  Google Scholar 

  76. Winer, D. A. et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat. Med. 17, 610–617 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Nishimura, S. et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med. 15, 914–920 (2009).

    CAS  PubMed  Google Scholar 

  78. Cipolletta, D. et al. PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 486, 549–553 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Kolodin, D. et al. Antigen- and cytokine-driven accumulation of regulatory T cells in visceral adipose tissue of lean mice. Cell Metab. 21, 543–557 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Feuerer, M. et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat. Med. 15, 930–939 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  82. Elinav, E. et al. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat. Rev. Cancer 13, 759–771 (2013).

    CAS  PubMed  Google Scholar 

  83. Sung, H. K. et al. Adipose vascular endothelial growth factor regulates metabolic homeostasis through angiogenesis. Cell Metab. 17, 61–72 (2013).

    CAS  PubMed  Google Scholar 

  84. Lee, Y. S. et al. Increased adipocyte O2 consumption triggers HIF-1alpha, causing inflammation and insulin resistance in obesity. Cell 157, 1339–1352 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Arendt, L. M. et al. Obesity promotes breast cancer by CCL2-mediated macrophage recruitment and angiogenesis. Cancer Res. 73, 6080–6093 (2013).

    CAS  PubMed  Google Scholar 

  86. Shah, D. et al. Obesity-induced adipokine imbalance impairs mouse pulmonary vascular endothelial function and primes the lung for injury. Sci. Rep. 5, 11362 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Wang, L. et al. Enhancement of endothelial permeability by free fatty acid through lysosomal cathepsin B-mediated Nlrp3 inflammasome activation. Oncotarget 7, 73229–73241 (2016).

    PubMed  PubMed Central  Google Scholar 

  88. Incio, J. et al. Obesity promotes resistance to anti-VEGF therapy in breast cancer by up-regulating IL-6 and potentially FGF-2. Sci. Transl Med. 10, eaag0945 (2018).

    PubMed  PubMed Central  Google Scholar 

  89. Carmeliet, P. & Jain, R. K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 10, 417–427 (2011).

    CAS  PubMed  Google Scholar 

  90. Tammela, T. & Alitalo, K. Lymphangiogenesis: molecular mechanisms and future promise. Cell 140, 460–476 (2010).

    CAS  PubMed  Google Scholar 

  91. Greene, A. K., Grant, F. D. & Slavin, S. A. Lower-extremity lymphedema and elevated body-mass index. N. Engl. J. Med. 366, 2136–2137 (2012).

    CAS  PubMed  Google Scholar 

  92. McLaughlin, S. A. et al. Prevalence of lymphedema in women with breast cancer 5 years after sentinel lymph node biopsy or axillary dissection: objective measurements. J. Clin. Oncol. 26, 5213–5219 (2008).

    PubMed  PubMed Central  Google Scholar 

  93. Wong, B. W. et al. The role of fatty acid beta-oxidation in lymphangiogenesis. Nature 542, 49–54 (2017).

    CAS  PubMed  Google Scholar 

  94. Stacker, S. A. et al. Lymphangiogenesis and lymphatic vessel remodelling in cancer. Nat. Rev. Cancer 14, 159–172 (2014).

    CAS  PubMed  Google Scholar 

  95. Harvey, N. L. et al. Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity. Nat. Genet. 37, 1072–1081 (2005).

    CAS  PubMed  Google Scholar 

  96. Escobedo, N. et al. Restoration of lymphatic function rescues obesity in Prox1-haploinsufficient mice. JCI Insight 1, e85096 (2016).

    PubMed  PubMed Central  Google Scholar 

  97. Lund, A. W. Rethinking lymphatic vessels and antitumor immunity. Trends Cancer 2, 548–551 (2016).

    PubMed  Google Scholar 

  98. Hespe, G. E. et al. Exercise training improves obesity-related lymphatic dysfunction. J. Physiol. 594, 4267–4282 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Nitti, M. D. et al. Obesity-induced lymphatic dysfunction is reversible with weight loss. J. Physiol. 594, 7073–7087 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Escobedo, N. & Oliver, G. The lymphatic vasculature: its role in adipose metabolism and obesity. Cell Metab. 26, 598–609 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Cao, Y. Angiogenesis and vascular functions in modulation of obesity, adipose metabolism, and insulin sensitivity. Cell Metab. 18, 478–489 (2013).

    CAS  PubMed  Google Scholar 

  102. Levental, K. R. et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139, 891–906 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Paszek, M. J. et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005).

    CAS  PubMed  Google Scholar 

  104. Tanaka, M. et al. Macrophage-inducible C-type lectin underlies obesity-induced adipose tissue fibrosis. Nat. Commun. 5, 4982 (2014).

    CAS  PubMed  Google Scholar 

  105. Seo, B. R. et al. Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis. Sci. Transl Med. 7, 301ra130 (2015).

    PubMed  PubMed Central  Google Scholar 

  106. Iyengar, P. et al. Adipocyte-derived collagen VI affects early mammary tumor progression in vivo, demonstrating a critical interaction in the tumor/stroma microenvironment. J. Clin. Invest. 115, 1163–1176 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Khan, T. et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol. Cell. Biol. 29, 1575–1591 (2009).

    CAS  PubMed  Google Scholar 

  108. Halberg, N. et al. Hypoxia-inducible factor 1alpha induces fibrosis and insulin resistance in white adipose tissue. Mol. Cell. Biol. 29, 4467–4483 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Incio, J. et al. Obesity-induced inflammation and desmoplasia promote pancreatic cancer progression and resistance to chemotherapy. Cancer Discov. 6, 852–869 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Zhang, Z. & Scherer, P. E. Adipose tissue: the dysfunctional adipocyte - a cancer cell’s best friend. Nat. Rev. Endocrinol. 14, 132–134 (2018).

    CAS  PubMed  Google Scholar 

  111. Zhang, M. et al. Adipocyte-derived lipids mediate melanoma progression via FATP proteins. Cancer Discov. 8, 1006–1025 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Laurent, V. et al. Periprostatic adipocytes act as a driving force for prostate cancer progression in obesity. Nat. Commun. 7, 10230 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Kim, J. Y. et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J. Clin. Invest. 117, 2621–2637 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Yamauchi, T. et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. 7, 941–946 (2001).

    CAS  PubMed  Google Scholar 

  115. Zeng, W. et al. Sympathetic neuro-adipose connections mediate leptin-driven lipolysis. Cell 163, 84–94 (2015).

    CAS  PubMed  Google Scholar 

  116. Zhang, T. et al. CXCL1 mediates obesity-associated adipose stromal cell trafficking and function in the tumour microenvironment. Nat. Commun. 7, 11674 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Zhang, Y. et al. White adipose tissue cells are recruited by experimental tumors and promote cancer progression in mouse models. Cancer Res. 69, 5259–5266 (2009).

    CAS  PubMed  Google Scholar 

  118. Zhang, Y. et al. Stromal progenitor cells from endogenous adipose tissue contribute to pericytes and adipocytes that populate the tumor microenvironment. Cancer Res. 72, 5198–5208 (2012).

    CAS  PubMed  Google Scholar 

  119. Klopp, A. H. et al. Omental adipose tissue-derived stromal cells promote vascularization and growth of endometrial tumors. Clin. Cancer Res. 18, 771–782 (2012).

    CAS  PubMed  Google Scholar 

  120. Song, Y. H. et al. Breast cancer-derived extracellular vesicles stimulate myofibroblast differentiation and pro-angiogenic behavior of adipose stem cells. Matrix Biol. 60–61, 190–205 (2017).

    PubMed  Google Scholar 

  121. Strong, A. L. et al. Leptin produced by obese adipose stromal/stem cells enhances proliferation and metastasis of estrogen receptor positive breast cancers. Breast Cancer Res. 17, 112 (2015).

    PubMed  PubMed Central  Google Scholar 

  122. Daquinag, A. C. et al. Targeted proapoptotic peptides depleting adipose stromal cells inhibit tumor growth. Mol. Ther. 24, 34–40 (2016).

    CAS  PubMed  Google Scholar 

  123. Stern, J. H., Rutkowski, J. M. & Scherer, P. E. Adiponectin, leptin, and fatty acids in the maintenance of metabolic homeostasis through adipose tissue crosstalk. Cell Metab. 23, 770–784 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Quail, D. F. et al. Obesity alters the lung myeloid cell landscape to enhance breast cancer metastasis through IL5 and GM-CSF. Nat. Cell Biol. 19, 974–987 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Park, E. J. et al. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 140, 197–208 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Li, R. et al. Obesity, rather than diet, drives epigenomic alterations in colonic epithelium resembling cancer progression. Cell Metab. 19, 702–711 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Ericksen, R. E. et al. Obesity accelerates Helicobacter felis-induced gastric carcinogenesis by enhancing immature myeloid cell trafficking and TH17 response. Gut 63, 385–394 (2014).

    CAS  PubMed  Google Scholar 

  128. Nelson, E. R. et al. 27-Hydroxycholesterol links hypercholesterolemia and breast cancer pathophysiology. Science 342, 1094–1098 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Wu, Q. et al. 27-Hydroxycholesterol promotes cell-autonomous, ER-positive breast cancer growth. Cell Rep. 5, 637–645 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Baek, A. E. et al. The cholesterol metabolite 27 hydroxycholesterol facilitates breast cancer metastasis through its actions on immune cells. Nat. Commun. 8, 864 (2017).

    PubMed  PubMed Central  Google Scholar 

  131. Voisin, M. et al. Identification of a tumor-promoter cholesterol metabolite in human breast cancers acting through the glucocorticoid receptor. Proc. Natl Acad. Sci. USA 114, E9346–E9355 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Behan, J. W. et al. Adipocytes impair leukemia treatment in mice. Cancer Res. 69, 7867–7874 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Sheng, X. et al. Adipocytes sequester and metabolize the chemotherapeutic daunorubicin. Mol. Cancer Res. 15, 1704–1713 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Straussman, R. et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487, 500–504 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Olson, O. C., Kim, H., Quail, D. F., Foley, E. A. & Joyce, J. A. Tumor-associated macrophages suppress the cytotoxic activity of antimitotic agents. Cell Rep. 19, 101–113 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Zarrinpar, A., Chaix, A., Yooseph, S. & Panda, S. Diet and feeding pattern affect the diurnal dynamics of the gut microbiome. Cell Metab. 20, 1006–1017 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Panda, S. Circadian physiology of metabolism. Science 354, 1008–1015 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Marcheva, B. et al. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466, 627–631 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Costa, M. J. et al. Circadian rhythm gene period 3 is an inhibitor of the adipocyte cell fate. J. Biol. Chem. 286, 9063–9070 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Hatori, M. et al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 15, 848–860 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Sulli, G. et al. Pharmacological activation of REV-ERBs is lethal in cancer and oncogene-induced senescence. Nature 553, 351–355 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Joyce, J. A. Therapeutic targeting of the tumor microenvironment. Cancer Cell 7, 513–520 (2005).

    CAS  PubMed  Google Scholar 

  143. Miyazawa, M. et al. Pioglitazone inhibits periprostatic white adipose tissue inflammation in obese mice. Cancer Prev. Res. 11, 215–226 (2017).

    Google Scholar 

  144. Klil-Drori, A. J., Azoulay, L. & Pollak, M. N. Cancer, obesity, diabetes, and antidiabetic drugs: is the fog clearing? Nat. Rev. Clin. Oncol. 14, 85–99 (2017).

    CAS  PubMed  Google Scholar 

  145. Ghorpade, D. S. et al. Hepatocyte-secreted DPP4 in obesity promotes adipose inflammation and insulin resistance. Nature 555, 673–677 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Pietrocola, F. et al. Caloric restriction mimetics enhance anticancer immunosurveillance. Cancer Cell 30, 147–160 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Di Biase, S. et al. Fasting-mimicking diet reduces HO-1 to promote T cell-mediated tumor cytotoxicity. Cancer Cell 30, 136–146 (2016).

    PubMed  PubMed Central  Google Scholar 

  148. Cypess, A. M. et al. Activation of human brown adipose tissue by a beta3-adrenergic receptor agonist. Cell Metab. 21, 33–38 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Bhardwaj, P. et al. Estrogen protects against obesity-induced mammary gland inflammation in mice. Cancer Prev. Res. 8, 751–759 (2015).

    CAS  Google Scholar 

  150. Cancello, R. et al. Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes 54, 2277–2286 (2005).

    CAS  PubMed  Google Scholar 

  151. Adams, T. D. et al. Long-term mortality after gastric bypass surgery. N. Engl. J. Med. 357, 753–761 (2007).

    CAS  PubMed  Google Scholar 

  152. Sjostrom, L. et al. Effects of bariatric surgery on mortality in Swedish obese subjects. N. Engl. J. Med. 357, 741–752 (2007).

    PubMed  Google Scholar 

  153. Schauer, D. P. et al. Bariatric surgery and the risk of cancer in a large multisite cohort. Ann. Surg. https://doi.org/10.1097/SLA.0000000000002525 (2017).

    Article  PubMed  Google Scholar 

  154. Ligibel, J. A. et al. Randomized phase III trial evaluating the role of weight loss in adjuvant treatment of overweight and obese women with early breast cancer (Alliance A011401): study design. NPJ Breast Cancer 3, 37 (2017).

    PubMed  PubMed Central  Google Scholar 

  155. Clement, K. et al. Weight loss regulates inflammation-related genes in white adipose tissue of obese subjects. FASEB J. 18, 1657–1669 (2004).

    CAS  PubMed  Google Scholar 

  156. Aleman, J. O. et al. Effects of rapid weight loss on systemic and adipose tissue inflammation and metabolism in obese postmenopausal women. J. Endocr. Soc. 1, 625–637 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Wernstedt Asterholm, I. et al. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab. 20, 103–118 (2014).

    CAS  PubMed  Google Scholar 

  158. Bhardwaj, P. et al. Caloric restriction reverses obesity-induced mammary gland inflammation in mice. Cancer Prev. Res. 6, 282–289 (2013).

    CAS  Google Scholar 

  159. Kalaany, N. Y. & Sabatini, D. M. Tumours with PI3K activation are resistant to dietary restriction. Nature 458, 725–731 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Esposito, K. et al. Effect of a mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. JAMA 292, 1440–1446 (2004).

    CAS  PubMed  Google Scholar 

  161. Esposito, K. et al. Effect of weight loss and lifestyle changes on vascular inflammatory markers in obese women: a randomized trial. JAMA 289, 1799–1804 (2003).

    CAS  PubMed  Google Scholar 

  162. Toledo, E. et al. Mediterranean diet and invasive breast cancer risk among women at high cardiovascular risk in the PREDIMED trial: a randomized clinical trial. JAMA Intern. Med. 175, 1752–1760 (2015).

    PubMed  Google Scholar 

  163. Samaha, F. F. et al. A low-carbohydrate as compared with a low-fat diet in severe obesity. N. Engl. J. Med. 348, 2074–2081 (2003).

    CAS  PubMed  Google Scholar 

  164. Foster, G. D. et al. A randomized trial of a low-carbohydrate diet for obesity. N. Engl. J. Med. 348, 2082–2090 (2003).

    CAS  PubMed  Google Scholar 

  165. Shai, I. et al. Weight loss with a low-carbohydrate, mediterranean, or low-fat diet. N. Engl. J. Med. 359, 229–241 (2008).

    CAS  PubMed  Google Scholar 

  166. Koelwyn, G. J., Quail, D. F., Zhang, X., White, R. M. & Jones, L. W. Exercise-dependent regulation of the tumour microenvironment. Nat. Rev. Cancer 17, 620–632 (2017).

    PubMed  Google Scholar 

  167. Moore, S. C. et al. Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern. Med. 176, 816–825 (2016).

    PubMed  PubMed Central  Google Scholar 

  168. Dieli-Conwright, C. M. et al. Effects of aerobic and resistance exercise on metabolic syndrome, sarcopenic obesity, and circulating biomarkers in overweight or obese survivors of breast cancer: a randomized controlled trial. J. Clin. Oncol. 36, 875–883 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Playdon, M. C. et al. Weight gain after breast cancer diagnosis and all-cause mortality: systematic review and meta-analysis. J. Natl Cancer Inst. 107, djv275 (2015).

    PubMed  PubMed Central  Google Scholar 

  170. Demark-Wahnefried, W. et al. Weight management and physical activity throughout the cancer care continuum. CA Cancer J. Clin. 68, 64–89 (2018).

    PubMed  Google Scholar 

  171. Iyengar, N. M. et al. Body fat and risk of breast cancer in postmenopausal women with normal body mass index. JAMA Oncol. (in the press).

  172. Mirsoian, A. et al. Adiposity induces lethal cytokine storm after systemic administration of stimulatory immunotherapy regimens in aged mice. J. Exp. Med. 211, 2373–2383 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Gupta, S. Obesity: the fat advantage. Nature 537, S100–S102 (2016).

    CAS  PubMed  Google Scholar 

  174. Denis, G. V. & Obin, M. S. ‘Metabolically healthy obesity’: origins and implications. Mol. Aspects Med. 34, 59–70 (2013).

    CAS  PubMed  Google Scholar 

  175. Stefan, N., Schick, F. & Haring, H. U. Causes, characteristics, and consequences of metabolically unhealthy normal weight in humans. Cell Metab. 26, 292–300 (2017).

    CAS  PubMed  Google Scholar 

  176. Rubin, R. Postmenopausal women with a “normal” BMI might be overweight or even obese. JAMA 319, 1185–1187 (2018).

    PubMed  Google Scholar 

  177. WHO Expert Consultation. Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet 363, 157–163 (2004).

    Google Scholar 

  178. Naveiras, O. et al. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature 460, 259–263 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Lee, J. M. et al. Obesity alters the long-term fitness of the hematopoietic stem cell compartment through modulation of Gfi1 expression. J. Exp. Med. 215, 627–644 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Masamoto, Y. et al. Adiponectin enhances antibacterial activity of hematopoietic cells by suppressing bone marrow inflammation. Immunity 44, 1422–1433 (2016).

    CAS  PubMed  Google Scholar 

  181. Liu, A. et al. Bone marrow lympho-myeloid malfunction in obesity requires precursor cell-autonomous TLR4. Nat. Commun. 9, 708 (2018).

    PubMed  PubMed Central  Google Scholar 

  182. Trottier, M. D., Naaz, A., Li, Y. & Fraker, P. J. Enhancement of hematopoiesis and lymphopoiesis in diet-induced obese mice. Proc. Natl Acad. Sci. USA 109, 7622–7629 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Lu, Z. et al. Fasting selectively blocks development of acute lymphoblastic leukemia via leptin-receptor upregulation. Nat. Med. 23, 79–90 (2017).

    CAS  PubMed  Google Scholar 

  184. Yun, J. P. et al. Diet-induced obesity accelerates acute lymphoblastic leukemia progression in two murine models. Cancer Prev. Res. 3, 1259–1264 (2010).

    CAS  Google Scholar 

  185. Shafat, M. S. et al. Leukemic blasts program bone marrow adipocytes to generate a protumoral microenvironment. Blood 129, 1320–1332 (2017).

    CAS  PubMed  Google Scholar 

  186. Poynter, J. N. et al. Obesity over the life course and risk of acute myeloid leukemia and myelodysplastic syndromes. Cancer Epidemiol. 40, 134–140 (2016).

    PubMed  Google Scholar 

  187. Butturini, A. M. et al. Obesity and outcome in pediatric acute lymphoblastic leukemia. J. Clin. Oncol. 25, 2063–2069 (2007).

    PubMed  Google Scholar 

  188. Rossi, E. L. et al. Obesity-associated alterations in inflammation, epigenetics, and mammary tumor growth persist in formerly obese mice. Cancer Prev. Res. 9, 339–348 (2016).

    CAS  Google Scholar 

  189. Greenstein, A. S. et al. Local inflammation and hypoxia abolish the protective anticontractile properties of perivascular fat in obese patients. Circulation 119, 1661–1670 (2009).

    CAS  PubMed  Google Scholar 

  190. Rosen, C. J. & Bouxsein, M. L. Mechanisms of disease: is osteoporosis the obesity of bone? Nat. Clin. Pract. Rheumatol. 2, 35–43 (2006).

    CAS  PubMed  Google Scholar 

  191. Estruch, R. et al. Primary prevention of cardiovascular disease with a mediterranean diet supplemented with extra-virgin olive oil or nuts. N. Engl. J. Med. 378, e34 (2018).

    CAS  PubMed  Google Scholar 

  192. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01697566 (2018).

  193. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01101438 (2018).

  194. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02065687 (2018).

  195. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02040376 (2017).

  196. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01655719 (2017).

  197. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT00780234 (2016).

  198. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01838317 (2018).

  199. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT00099021 (2016).

  200. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT00427999 (2016).

  201. Irwin, M. L. et al. Randomized controlled trial of aerobic exercise on insulin and insulin-like growth factors in breast cancer survivors: the Yale Exercise and Survivorship study. Cancer Epidemiol. Biomarkers Prev. 18, 306–313 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Fairey, A. S. et al. Effects of exercise training on fasting insulin, insulin resistance, insulin-like growth factors, and insulin-like growth factor binding proteins in postmenopausal breast cancer survivors: a randomized controlled trial. Cancer Epidemiol. Biomarkers Prev. 12, 721–727 (2003).

    CAS  PubMed  Google Scholar 

  203. Spencer, M. et al. Pioglitazone treatment reduces adipose tissue inflammation through reduction of mast cell and macrophage number and by improving vascularity. PLOS ONE 9, e102190 (2014).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank Oakley C. Olson, Martin J. Richer and Azadeh Arabzadeh for their critical feedback on the manuscript. A.J.D. is supported by the Breast Cancer Research Foundation, the Botwinick-Wolfensohn Foundation (in memory of Mr and Mrs Benjamin Botwinick), NIH/NCI R01 CA215797 and NIH/NCI U54 CA210184. D.F.Q. is supported by Susan G. Komen CCR18548032 and Canadian Institutes of Health Research PJT-159742.

Author information

Authors and Affiliations

Authors

Contributions

D.F.Q and A.J.D. researched data for the article, contributed to the discussion of the content, wrote the article and reviewed and/or edited the manuscript before submission. Both authors contributed equally to this work.

Corresponding authors

Correspondence to Daniela F. Quail or Andrew J. Dannenberg.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Glossary

Adipocyte hypertrophy

Enlargement of adipocytes, which often occurs in association with obesity and increased numbers of crown-like structures in adipose tissue.

Metabolically obese normal-weight

(MONW). Individuals within a normal-range BMI category (18.5–24.9), yet with a high body fat composition, leading to qualitatively similar health risks as individuals who are obese.

Adipose tissue microenvironment

(ATME). The cellular and structural compartment of adipose tissue, including but not limited to the adipocyte.

Pyroptosis

Inflammatory programmed cell death, in which an immune cell bursts to release intracellular contents into the microenvironment to trigger a rapid immune response.

Myelopoiesis

Differentiation of haematopoetic progenitor cells within the bone marrow towards a myeloid lineage.

Monocytosis

Expansion of monocytes within the peripheral blood, which are precursors for macrophages and dendritic cells.

Crown-like structures

(CLS). A dying or dead adipocyte surrounded by a ‘crown’ of macrophages within adipose tissue; this structure is a histological biomarker of obesity-associated inflammation and the metabolic syndrome.

Metabo-inflammation

Inflammation within the adipose tissue microenvironment that has metabolic consequences irrespective of BMI status.

Benign breast disease

Heterogeneous group of lesions within the breast that might increase the risk of developing breast cancer.

CLS-associated macrophages

(CAMϕ). Macrophages that are directly associated with crown-like structures (CLS) in inflamed adipose tissue; these cells are phenotypically and transcriptionally distinct from other macrophages within the adipose tissue microenvironment.

Myofibroblasts

Contractile cells of the mesenchymal-fibroblast lineage that synthesize extracellular matrix and mediate tissue remodelling.

Adipose stromal cells

(ASCs). Multipotent mesenchymal progenitor cells found in adipose tissue that can differentiate into mesoderm lineages (such as adipocytes, myofibroblasts, chondrocytes and osteoblasts); several terms have been used in the literature to refer to these cells (for example, adipose-derived stem cells, pre-adipocytes, adipose mesenchymal stem cells or lipoblasts).

Neutrophilia

High number of mature neutrophils within the peripheral blood or within tissues, resulting from neutrophil leukocytosis.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Quail, D.F., Dannenberg, A.J. The obese adipose tissue microenvironment in cancer development and progression. Nat Rev Endocrinol 15, 139–154 (2019). https://doi.org/10.1038/s41574-018-0126-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41574-018-0126-x

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer