The effect of adipocyte-macrophage crosstalk in obesity-related breast cancer


Creative Commons License

ENGİN A. B., Engin A., IŞIK GÖNÜL İ.

JOURNAL OF MOLECULAR ENDOCRINOLOGY, cilt.62, sa.3, 2019 (SCI-Expanded) identifier identifier identifier

  • Yayın Türü: Makale / Derleme
  • Cilt numarası: 62 Sayı: 3
  • Basım Tarihi: 2019
  • Doi Numarası: 10.1530/jme-18-0252
  • Dergi Adı: JOURNAL OF MOLECULAR ENDOCRINOLOGY
  • Derginin Tarandığı İndeksler: Science Citation Index Expanded (SCI-EXPANDED), Scopus
  • Anahtar Kelimeler: obesity, breast cancer, aromatase activity, TSC1-TSC2 complex, estrogen receptor, ADIPOSE-TISSUE MACROPHAGES, ENDOPLASMIC-RETICULUM STRESS, ESTROGEN-RECEPTOR-ALPHA, BODY-MASS INDEX, INDUCED INFLAMMATORY CHANGES, UNFOLDED PROTEIN RESPONSE, ARYL-HYDROCARBON RECEPTOR, TUMOR-NECROSIS-FACTOR, IKK-BETA SUPPRESSION, FACTOR-KAPPA-B
  • Gazi Üniversitesi Adresli: Evet

Özet

Adipose tissue is the primary source of many pro-inflammatory cytokines in obesity. Macrophage numbers and pro-inflammatory gene expression are positively associated with adipocyte size. Free fatty acid and tumor necrosis factor-alpha involve in a vicious cycle between adipocytes and macrophages aggravating inflammatory changes. Thereby, M1 macrophages form a characteristic 'crown-like structure (CLS)' around necrotic adipocytes in obese adipose tissue. In obese women, CLSs of breast adipose tissue are responsible for both increase in local aromatase activity and aggressive behavior of breast cancer cells. Interlinked molecular mechanisms between adipocyte-macrophage-breast cancer cells in obesity involve seven consecutive processes: Excessive release of adipocyte- and macrophage-derived inflammatory cytokines, TSC1-TSC2 complex-mTOR crosstalk, insulin resistance, endoplasmic reticulum (ER) stress and excessive oxidative stress generation, uncoupled respiration and hypoxia, SIRT1 controversy, the increased levels of aromatase activity and estrogen production. Considering elevated risks of estrogen receptor (E2R)-positive postmenopausal breast cancer growth in obesity, adipocytemacrophage crosstalk is important in the aforementioned issues. Increased mTORC1 signaling in obesity ensures the strong activation of oncogenic signaling in E2R alpha-positive breast cancer cells. Since insulin and insulin-like growth factors have been identified as tumor promoters, hyperinsulinemia is an independent risk factor for poor prognosis in breast cancer despite peripheral insulin resistance. The unpredictable effects of adipocyte-derived leptin-estrogen-macrophage axis, and sirtuin 1 (SIRT1)-adipose-resident macrophage axis in obese postmenopausal patients with breast cancer are unresolved mechanistic gaps in the molecular links between the tumor growth and adipocytokines.