Journal of Obesity & Metabolic Syndrome

Search

Article

J Obes Metab Syndr 2023; 32(4): 312-321

Published online December 30, 2023 https://doi.org/10.7570/jomes23044

Copyright © Korean Society for the Study of Obesity.

Fat Biology in Triple-Negative Breast Cancer: Immune Regulation, Fibrosis, and Senescence

Chae Min Lee1,2, Sungsoon Fang1,2,3,4,*

1Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul; 2Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul; 3Chronic Intractable Disease for Systems Medicine Research Center, 4Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea

Correspondence to:
Sungsoon Fang
https://orcid.org/0000-0003-0201-5567
Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Korea
Tel: +82-2-2019-5406
Fax: +82-2-2227-8306
E-mail: sfang@yuhs.ac

Received: July 21, 2023; Reviewed : September 18, 2023; Accepted: November 21, 2023

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Obesity, now officially recognized as a disease requiring intervention, has emerged as a significant health concern due to its strong association with elevated susceptibility to diverse diseases and various types of cancer, including breast cancer. The link between obesity and cancer is intricate, with obesity exerting a significant impact on cancer recurrence and elevated mortality rates. Among the various subtypes of breast cancer, triple-negative breast cancer (TNBC) is the most aggressive, accounting for 15% to 20% of all cases. TNBC is characterized by low expression of estrogen receptors and progesterone receptors as well as the human epidermal growth factor 2 receptor protein. This subtype poses distinct challenges in terms of treatment response and exhibits strong invasiveness. Furthermore, TNBC has garnered attention because of its association with obesity, in which excess body fat and reduced physical activity have been identified as contributing factors to the increased incidence of this aggressive form of breast cancer. In this comprehensive review, the impact of obesity on TNBC was explored. Specifically, we focused on the three key mechanisms by which obesity affects TNBC development and progression: modification of the immune profile, facilitation of fibrosis, and initiation of senescence. By comprehensively examining these mechanisms, we illuminated the complex interplay between TNBC and obesity, facilitating the development of novel approaches for prevention, early detection, and effective management of this challenging disease.

Keywords: Triple-negative breast cancer, Obesity, Tumor microenvironment, Immunity, Fibrosis, Senescence

Obesity, which is becoming increasingly prevalent, has gained official recognition as a ‘disease’ requiring intervention. According to the World Health Organization, obesity is defined as a body mass index equal to or greater than 30 kg/m2.1 This condition is closely linked to susceptibility to metabolic diseases, cardiovascular diseases, musculoskeletal diseases, depression, Alzheimer’s disease, and various types of cancer, including breast cancer.2-8 The link between obesity and cancer is complex, with obesity being associated with increased risk of cancer.9 Furthermore, obesity is associated with increased risk of cancer recurrence and higher mortality rates.10 Therefore, management of obesity is important for improving cancer outcomes, emphasizing the need for early intervention in patients with early-stage cancer.

Triple-negative breast cancer (TNBC) has low expression of estrogen receptor and progesterone receptor as well as the human epidermal growth factor 2 receptor protein, and it accounts for 10% to 15% of breast cancer cases.11,12 It is the most aggressive type of breast cancer and is characterized by a low response to treatment and strong invasive properties; few studies have been performed in this type breast cancer.13 While there are many complexities yet to be uncovered, current research has revealed a multitude of factors influencing the development of TNBC. Age, race, genetic variations, family history, oral contraceptive use, and weight, including obesity or overweight, have emerged as notable contributors.14-18

An abundance of research has delved into the intricate correlations between obesity and TNBC. It has been firmly established that a TNBC diagnosis is notably more prevalent among individuals classified as obese, as opposed to those who are not obese. Recent investigations have studied distinct clinical disparities within TNBC that are intimately linked to obesity.19,20 Notably, obese TNBC patients frequently present with more substantial tumor dimensions and elevated T-stage categorizations and exhibit a propensity for higher tumor grades.21 Nevertheless, the exact influence of metabolic changes in adipose tissue on the progression of TNBC remains a mystery, underscoring the need for additional research.

Therefore, we aimed to investigate the influence of obesity on TNBC based on existing research. We focused on how obesity impacts TNBC development through three mechanisms: modification of the immune profile, facilitation of fibrosis, and initiation of senescence (Fig. 1).

Recent studies have revealed that obese animals exhibit a state of hypoxia in adipose tissue.22,23 This hypoxia leads to interactions between adipose cells and stromal vascular cells, driving tumor initiation and progression.24 Within adipose tissue, hypoxia triggers increased insulin resistance; decreased adiponectin; and elevated leptin, adipocyte apoptosis, and endoplasmic reticulum (ER) stress.25-28 Ultimately, these changes result in favorable microenvironment alterations for tumor development, such as enhanced vascularization and extracellular matrix (ECM) remodeling.

In summary, the impact of obesity on tumorigenesis can be summarized in three main facets. First, there is an increase in inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6). This cytokine upregulation leads to chronic inflammation, primarily associated with breast cancer and colorectal cancer.29,30 Second, the elevated level of leptin activates oncogenic transcription factors, linked to breast cancer and prostate cancer.31,32 Last, the reduction in adiponectin interferes with proliferation and mutagenesis, potentially leading to breast cancer and lung cancer.33 As a result of these characteristics, adipose tissue has emerged as a crucial component in the tumor microenvironment (TME).

Through these insights, it has become evident that obesity significantly influences various cancers, with a pronounced impact on breast cancer. In TNBC with obesity, these insights are aligned with the previously mentioned effects of obesity on tumor development. These cancer-associated adipocytes release free fatty acids into breast cancer cells,34 triggering the secretion of pro-inflammatory cytokines and growth factors, including interferon-gamma, insulin-like growth factor (IGF)-1, IL-6, IL-8, leptin, and TNF-α (Fig. 2).35-37 These insights are consistent with research indicating that obese TNBC patients have elevated levels of inflammatory cytokines TNF-α and IL-6 and lower level of adiponectin compared to that in non-obese patients.20 Remarkably, these inflammatory cytokines accelerate tissue inflammation and activate signaling pathways, contributing to the aggressive nature of TNBC.

In TNBC, adipose tissue stem cells play a significant role in facilitating tumor growth, epithelial-mesenchymal transition (EMT), and invasion of breast cancer cells, which ultimately lead to changes in the composition of the ECM. Specifically, obesity contributes to increased secretion of leptin into TNBC cells. This mechanism converts adipose stem cells and triggers EMT in cancer cells,38 modifying the TNBC microenvironment and amplifying metastatic potential through the leptin-mediated pathway. Research has shown that when leptin is combined with cyclic adenosine monophosphate-elevating agents, it has the capability of inducing apoptosis in TNBC.39 Conversely, adiponectin exerts a negative influence on breast tumor formation.20 Adiponectin expression is notably lower in breast cancer tissue from obese individuals than in individuals that are not obese. The impact of adiponectin varies based on estrogen receptor status, with an elevated leptin-to-adiponectin ratio showing a more pronounced association with TNBC than human epidermal growth factor 2 receptor -positive breast cancer.40 Furthermore, adiponectin hinders cell proliferation, invasion, and migration, while also triggering apoptosis and autophagic cell death in estrogen receptor and progesterone receptor-negative breast cancer.41

In conclusion, adipose tissue impacts TNBC in multiple ways. Cytokines derived from adipose tissue promote the migration and proliferation of TNBC cells, disrupting normal adipokine and hormone levels. This disruption fosters the promotion of mitogenic and mutagenic pathways, advancing tumor progression in obese TNBC patients.

Numerous studies have emphasized the impact of obesity on the immune landscape, often referred to as “meta-inflammation.”42,43 With the development of obesity, there is an increase in immune infiltration and angiogenesis, leading to increased inflammation.42 These findings have been emphasized in multiple studies, highlighting the significance of this phenomenon in the context of obesity-related pathophysiology. Therefore, there should be a focus on the influence of obesity on the heterogeneity of the immune system in TNBC.

The expression of the immune checkpoint ligand programmed death ligand-1 (PD-L1) in obesity is associated with immune suppression, and the expression of the programmed death-1 receptor (PD-1) and PD-L1 in TNBC shows an intermediate level of variability.44,45 In TNBC tumors, PD-L1 expression inhibits the interaction between PD-L1 and the PD-1 receptor, restoring T-cell activation and supporting antitumor immunity.46 Interestingly, several studies have indicated that TNBC patients with obesity, characterized by an exhausted immune response, may have impaired checkpoint function.47,48

Moreover, insulin signaling plays a critical role in the development of obesity-related cancers, including TNBC. The IGF system is closely associated with tumorigenesis and the development and progression of tumor cells.49 Leptin directly affects the activation of the IGF-1 receptor, creating mutual activation between leptin and the IGF-1 receptor.50 This phenomenon leads to migration and proliferation of TNBC cells, as higher level of IGF-1 receptors is observed in TNBC. In addition, TNBC exhibits rapid growth that is dependent on glucose utilization and aerobic glycolysis.51 This rapid glycolytic growth and increased glucose uptake create an environment rich in anabolic precursors, promoting accelerated growth and contributing to mitochondrial dysfunction which, in turn, enhances resistance to cancer cell apoptosis.52

Furthermore, obesity has a significant impact on both the activity and numbers of natural killer (NK) and natural killer T (NKT) cells.53 Obesity induces alterations in the phenotype and functionality of NK cells. These changes, along with obesity-induced shifts in the function and number of T-cells, collectively contribute to increased cancer susceptibility in obese individuals.54 In addition, there is an increase in immature, non-cytotoxic NK cells and a decrease in mature, cytotoxic NK cells in obese patients with TNBC.55,56 These findings suggest that a reduction in both the number and function of NK cells can be anticipated in TNBC patients who are obese.

In summary, TNBC is characterized by enhanced fatty acid synthase and glycolysis,51 whereas obesity is associated with immune paralysis of NK cells. Specifically, obesity exerts an inhibitory effect on NK cell function, which normally contributes to the control of tumor cell growth and metastasis. However, in the presence of breast cancer cells, NK cell function is impaired, resulting in reduced antitumor activity.57 Collectively, the interplay between obesity and TNBC triggers a complex immune response that promotes cancer cell proliferation.

ECM proteins serve a dual role in regulating the mechanical properties of adipose tissue and influencing adipogenesis.58 When obesity leads to an increase in adipocyte volume and tissue mass, it disrupts the normal functioning of adipose tissue. This disruption triggers inflammation and eventually results in adipose tissue fibrosis.59 In addition, homeostasis and organization of the epithelial tissue are affected by the ECM in adipocytes.60 This process highlights the critical role of ECM proteins in adipose tissue homeostasis and the pathogenesis of adipose tissue fibrosis associated with obesity. Stiff and increased ECM contributes to the local and systemic pathologies associated with obesity, promoting collagen formation around adipocytes, which leads to fibrosis.61 In addition, high ECM stiffness leads to EMT in cancer cells, ultimately resulting in local migration, invasion, metastasis, and the loss of epithelial polarity.62-65 These conditions are particularly notable in mammary tissue, where collagen deposition and alignment increase around mammary ducts and pre-neoplastic lesions.66 Specific changes are characterized by increased expression of collagen VI and reduced elastin in subcutaneous fat, signifying alterations in ECM properties.67 These changes affect tissue ability to retain secreted molecules, with transforming growth factor beta 1 (TGFβ1) expression being a notable example. TGFβ1 is known to induce EMT in tumor cells and to foster the growth of cancer-associated fibroblasts (CAFs) within the TME.68 In obese adipose tissue, TGFβ1 expression is upregulated.69 Initially, it is produced in an inactive or latent form, requiring extracellular activation before it can bind to receptors. Consequently, latent TGFβ1 present in the ECM binds to latent TGFβ1 proteins and matrix components, including decorin, which sequesters inactive TGFβ1 until activation. Within the mammary ECM of obese patients, decorin becomes enriched and forms a complex with latent TGFβ1, resulting in increased TGFβ1 storage.70 This influences both tumor cell proliferation and CAF development. Specifically, ECM components participate in TNBC progression by influencing the activation of the signaling pathways that govern the properties of TNBC cells.67 An interesting observation is the increased presence of collagen VI in both TNBC and obese ECM. This increase is mediated through ECM, growth factors, and mitogen-activated protein kinase (MAPK) signaling pathway interactions. Specifically, collagen VI has been found to impact TNBC cell adhesion, two-dimensional migration, and three-dimensional invasion.71 In simpler terms, obesity can profoundly alter the composition of ECM in TNBC tissue, potentially enhancing its local invasiveness and metastatic potential.

Hypoxia, characterized by insufficient tissue oxygenation due to reduced blood supply, has been extensively studied as a major contributor to fibrosis in obese adipose tissue.72 In adipose tissue, there is a notable increase in the expression of hypoxia-induced factor-1 alpha (HIF-1α).73 Furthermore, the growth of tumors triggers intra-tumoral hypoxia, leading to concurrent activation of the HIF-1α pathway within the TME.74 Elevated HIF-1α in breast tumors, including TNBC, can cause metabolic changes in tumor cells and promote metastasis.75,76 These findings suggest that upregulation of HIF-1α stimulates tumor formation and contributes to aggressive tumor growth.

The impact of aging in the microenvironment on obese TNBC patients has not yet been clearly elucidated. However, the influence of obesity and breast cancer, especially TNBC, on cellular aging has been well-established. Therefore, we were focused on the effects of obesity and TNBC on cellular aging.

The increase in inflammatory markers in obese patients might be due to certain cell populations undergoing senescence.77 Several studies have inferred that adipose tissue serves as a primary reservoir of senescent cells.78,79 Cellular senescence can be induced by multiple factors, such as DNA damage, telomere attrition, oxidative damage, mitotic stress, mitochondrial dysfunction, ER stress, and oncogene activation.80-86 DNA damage, which is frequently induced by telomere shortening and early onset cellular senescence, is the key factor in triggering senescence among these factors. Similarly, activation of oncogenes, such as DNA replication stress, can lead to DNA damage in TNBC. Consequently, there is a significant increase in the expression of DNA damage markers, dysregulated checkpoints, and suppressed DNA repair pathways in TNBC.87

Senescent cells also release a set of molecules known as senescence-associated secretory phenotype (SASP). This group of molecules is comprised of inflammatory factors, proteases, and growth factors, all of which foster an inflammatory environment and contribute to tumor development and progression.88,89 SASP factors are classified into three groups: soluble signaling factors (chemokines, growth factors, and interleukins), secreted proteases, and secreted insoluble protein and ECM components.90 The most important element here is the ECM. According to the results of these studies, obesity in TNBC promotes the expression of cancer stem cell and MET genes through leptin.91 Concomitantly, senescent cells exhibit an increased adenosine monophosphate:adenosine triphosphate ratio.92 adenosine monophosphate-activated protein kinase (AMPK), a key regulator of growth and cellular processes such as autophagy and cell polarity, is activated by the reduced intracellular adenosine triphosphate level.93

Furthermore, senescent cells impact ER stress and the unfolded protein response, leading to activation of these pathways, increasing the demand for SASP production.94 This activation triggers a heightened demand for SASP production and results in alterations in cell morphology through increased activity of the mammalian target of rapamycin (mTOR) in senescent cells.95 The composition of the SASP is influenced by various signaling pathways, including the mTOR pathway. Secretion by senescent cells plays a significant role in SASP.96 In obesity, the expression levels of ER stress and SASP increase, and these increases are associated with senescence. Furthermore, there is a noticeable connection between the functional aspects of invasion, migration, and ER stress in TNBC.97,98 Evidence is emerging to suggest that various cellular stresses associated with obesity and tumors play a role in cellular senescence, resulting in accumulation of senescent cells in adipose tissue and various types of cancer.

Overall, we sought to clarify how obesity affects the growth and advancement of this aggressive type of cancer. We explored three main mechanisms: changes in the immune system, promotion of fibrosis, and onset of senescence. Each of these mechanisms significantly impacts the challenging clinical landscape of TNBC in individuals dealing with obesity.

Considering the complexity of these interactions, a multidisciplinary approach is essential when devising personalized treatment strategies. These strategies should address not only tumor-related factors, but also the metabolic effects of obesity. Such a comprehensive approach holds promise for improving outcomes and prognoses in TNBC patients who are dealing with obesity.

Subsequent research should concentrate on specific intervention points within the intricate interplay of obesity and TNBC. Targeting these points could disrupt the cancer-promoting effects of obesity, potentially leading to more refined treatment strategies and ultimately better outcomes for those with obesity and TNBC.

Fig. 1. The immune profile, fibrosis, and senescence in adipocytes and triple-negative breast cancer (TNBC). The similarities between the tumor microenvironment in TNBC and obesity and their effects on the development of cancer. Natural killer (NK), natural killer T (NKT) cells, and lymphocytes indicate modification of the immune network, fibroblast and collagen participate in the facilitation of fibrosis, and senescent cells and senescence-associated secretory phenotype are related to initiation of senescence.
Fig. 2. The impact of obesity on triple-negative breast cancer (TNBC) tumor development. Adipocytes secrete free fatty acids (FFA) toward TNBC cells, and both TNBC cells and stromal cells are influenced by FFA. TNBC cells respond to these signals by releasing cytokines and growth factors, acting in tandem with stromal cells to promote TNBC tumor progression.
  1. National Institutes of Health. Clinical guidelines on the identification, evaluation, and treatment of overweight and obesity in adults: the evidence report. Obes Res 1998;6 Suppl 2:51S-209S.
  2. Jee SH, Kim HJ, Lee J. Obesity, insulin resistance and cancer risk. Yonsei Med J 2005;46:449-55.
    Pubmed KoreaMed CrossRef
  3. Calle EE, Thun MJ, Petrelli JM, Rodriguez C, Heath CW Jr. Body-mass index and mortality in a prospective cohort of U.S. adults. N Engl J Med 1999;341:1097-105.
    Pubmed CrossRef
  4. Blüher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol 2019;15:288-98.
    Pubmed CrossRef
  5. Karimi S, Pasdar Y, Hamzeh B, Ayenehpour A, Heydarpour F, Goudarzi F. Obesity phenotypes related to musculoskeletal disorders; a cross-sectional study from RaNCD cohort. Arch Public Health 2022;80:185.
    Pubmed KoreaMed CrossRef
  6. Blasco BV, García-Jiménez J, Bodoano I, Gutiérrez-Rojas L. Obesity and depression: its prevalence and influence as a prognostic factor: a systematic review. Psychiatry Investig 2020;17:715-24.
    Pubmed KoreaMed CrossRef
  7. Kang SY, Kim YJ, Jang W, Son KY, Park HS, Kim YS. Body mass index trajectories and the risk for Alzheimer's disease among older adults. Sci Rep 2021;11:3087.
    Pubmed KoreaMed CrossRef
  8. Yang YS, Han BD, Han K, Jung JH, Son JW; Taskforce Team of the Obesity Fact Sheet of the Korean Society for the Study of Obesity. Obesity fact sheet in Korea, 2021: trends in obesity prevalence and obesity-related comorbidity incidence stratified by age from 2009 to 2019. J Obes Metab Syndr 2022;31:169-77.
    Pubmed KoreaMed CrossRef
  9. Avgerinos KI, Spyrou N, Mantzoros CS, Dalamaga M. Obesity and cancer risk: emerging biological mechanisms and perspectives. Metabolism 2019;92:121-35.
    Pubmed CrossRef
  10. Petrelli F, Cortellini A, Indini A, Tomasello G, Ghidini M, Nigro O, et al. Association of obesity with survival outcomes in patients with cancer: a systematic review and meta-analysis. JAMA Netw Open 2021;4:e213520.
    Pubmed KoreaMed CrossRef
  11. Sun H, Zou J, Chen L, Zu X, Wen G, Zhong J. Triple-negative breast cancer and its association with obesity. Mol Clin Oncol 2017;7:935-42.
    Pubmed KoreaMed CrossRef
  12. Ma C, Zu X, Liu K, Bode AM, Dong Z, Liu Z, et al. Knockdown of pyruvate kinase M inhibits cell growth and migration by reducing NF-kB activity in triple-negative breast cancer cells. Mol Cells 2019;42:628-36.
  13. Cho S, Joo B, Park M, Ahn SJ, Suh SH, Park YW, et al. A radiomics-based model for potentially more accurate identification of subtypes of breast cancer brain metastases. Yonsei Med J 2023;64:573-80.
    Pubmed KoreaMed CrossRef
  14. Plasilova ML, Hayse B, Killelea BK, Horowitz NR, Chagpar AB, Lannin DR. Features of triple-negative breast cancer: analysis of 38,813 cases from the national cancer database. Medicine (Baltimore) 2016;95:e4614.
    Pubmed KoreaMed CrossRef
  15. Stevens KN, Vachon CM, Couch FJ. Genetic susceptibility to triple-negative breast cancer. Cancer Res 2013;73:2025-30.
    Pubmed KoreaMed CrossRef
  16. Phipps AI, Buist DS, Malone KE, Barlow WE, Porter PL, Kerlikowske K, et al. Family history of breast cancer in first-degree relatives and triple-negative breast cancer risk. Breast Cancer Res Treat 2011;126:671-8.
    Pubmed KoreaMed CrossRef
  17. Li L, Zhong Y, Zhang H, Yu H, Huang Y, Li Z, et al. Association between oral contraceptive use as a risk factor and triple-negative breast cancer: a systematic review and meta-analysis. Mol Clin Oncol 2017;7:76-80.
    Pubmed KoreaMed CrossRef
  18. Bao PP, Cai H, Peng P, Gu K, Su Y, Shu XO, et al. Body mass index and weight change in relation to triple-negative breast cancer survival. Cancer Causes Control 2016;27:229-36.
    Pubmed KoreaMed CrossRef
  19. Gourgue F, Mignion L, Van Hul M, Dehaen N, Bastien E, Payen V, et al. Obesity and triple-negative-breast-cancer: is apelin a new key target? J Cell Mol Med 2020;24:10233-44.
    Pubmed KoreaMed CrossRef
  20. Zhang K, Chen L, Zheng H, Zeng Y. Cytokines secreted from adipose tissues mediate tumor proliferation and metastasis in triple negative breast cancer. BMC Cancer 2022;22:886.
    Pubmed KoreaMed CrossRef
  21. Hao S, Liu Y, Yu KD, Chen S, Yang WT, Shao ZM. Overweight as a prognostic factor for triple-negative breast cancers in Chinese women. PLoS One 2015;10:e0129741.
    Pubmed KoreaMed CrossRef
  22. Todorčević M, Manuel AR, Austen L, Michailidou Z, Hazlehurst JM, Neville M, et al. Markers of adipose tissue hypoxia are elevated in subcutaneous adipose tissue of severely obese patients with obesity hypoventilation syndrome but not in the moderately obese. Int J Obes (Lond) 2021;45:1618-22.
    Pubmed KoreaMed CrossRef
  23. Alcalá M, Calderon-Dominguez M, Bustos E, Ramos P, Casals N, Serra D, et al. Increased inflammation, oxidative stress and mitochondrial respiration in brown adipose tissue from obese mice. Sci Rep 2017;7:16082.
    Pubmed KoreaMed CrossRef
  24. Almendros I, Gileles-Hillel A, Khalyfa A, Wang Y, Zhang SX, Carreras A, et al. Adipose tissue macrophage polarization by intermittent hypoxia in a mouse model of OSA: effect of tumor microenvironment. Cancer Lett 2015;361:233-9.
    Pubmed CrossRef
  25. Arcidiacono B, Chiefari E, Foryst-Ludwig A, Currò G, Navarra G, Brunetti FS, et al. Obesity-related hypoxia via miR-128 decreases insulin-receptor expression in human and mouse adipose tissue promoting systemic insulin resistance. EBioMedicine 2020;59:102912.
    Pubmed KoreaMed CrossRef
  26. Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab 2007;293:E1118-28.
    Pubmed CrossRef
  27. Alkhouri N, Gornicka A, Berk MP, Thapaliya S, Dixon LJ, Kashyap S, et al. Adipocyte apoptosis, a link between obesity, insulin resistance, and hepatic steatosis. J Biol Chem 2010;285:3428-38.
    Pubmed KoreaMed CrossRef
  28. Mondal AK, Das SK, Varma V, Nolen GT, McGehee RE, Elbein SC, et al. Effect of endoplasmic reticulum stress on inflammation and adiponectin regulation in human adipocytes. Metab Syndr Relat Disord 2012;10:297-306.
    Pubmed KoreaMed CrossRef
  29. Incio J, Ligibel JA, McManus DT, Suboj P, Jung K, Kawaguchi K, et al. Obesity promotes resistance to anti-VEGF therapy in breast cancer by up-regulating IL-6 and potentially FGF-2. Sci Transl Med 2018;10:eaag0945.
    Pubmed KoreaMed CrossRef
  30. Wunderlich CM, Ackermann PJ, Ostermann AL, Adams-Quack P, Vogt MC, Tran ML, et al. Obesity exacerbates colitis-associated cancer via IL-6-regulated macrophage polarisation and CCL-20/CCR-6-mediated lymphocyte recruitment. Nat Commun 2018;9:1646.
    Pubmed KoreaMed CrossRef
  31. Hosney M, Sabet S, El-Shinawi M, Gaafar KM, Mohamed MM. Leptin is overexpressed in the tumor microenvironment of obese patients with estrogen receptor positive breast cancer. Exp Ther Med 2017;13:2235-46.
    Pubmed KoreaMed CrossRef
  32. Noda T, Kikugawa T, Tanji N, Miura N, Asai S, Higashiyama S, et al. Long‑term exposure to leptin enhances the growth of prostate cancer cells. Int J Oncol 2015;46:1535-42.
    Pubmed CrossRef
  33. Di Zazzo E, Polito R, Bartollino S, Nigro E, Porcile C, Bianco A, et al. Adiponectin as link factor between adipose tissue and cancer. Int J Mol Sci 2019;20:839.
    Pubmed KoreaMed CrossRef
  34. Gyamfi J, Yeo JH, Kwon D, Min BS, Cha YJ, Koo JS, et al. Interaction between CD36 and FABP4 modulates adipocyte-induced fatty acid import and metabolism in breast cancer. NPJ Breast Cancer 2021;7:129.
    Pubmed KoreaMed CrossRef
  35. Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res 2011;71:2455-65.
    Pubmed CrossRef
  36. Sánchez-Jiménez F, Pérez-Pérez A, de la Cruz-Merino L, Sánchez-Margalet V. Obesity and breast cancer: role of leptin. Front Oncol 2019;9:596.
    Pubmed KoreaMed CrossRef
  37. Wu Q, Li B, Li Z, Li J, Sun S, Sun S. Cancer-associated adipocytes: key players in breast cancer progression. J Hematol Oncol 2019;12:95.
    Pubmed KoreaMed CrossRef
  38. Sabol RA, Bowles AC, Côté A, Wise R, O'Donnell B, Matossian MD, et al. Leptin produced by obesity-altered adipose stem cells promotes metastasis but not tumorigenesis of triple-negative breast cancer in orthotopic xenograft and patient-derived xenograft models. Breast Cancer Res 2019;21:67.
    Pubmed KoreaMed CrossRef
  39. Naviglio S, Di Gesto D, Romano M, Sorrentino A, Illiano F, Sorvillo L, et al. Leptin enhances growth inhibition by cAMP elevating agents through apoptosis of MDA-MB-231 breast cancer cells. Cancer Biol Ther 2009;8:1183-90.
    Pubmed CrossRef
  40. Sultana R, Kataki AC, Borthakur BB, Basumatary TK, Bose S. Imbalance in leptin-adiponectin levels and leptin receptor expression as chief contributors to triple negative breast cancer progression in Northeast India. Gene 2017;621:51-8.
    Pubmed CrossRef
  41. Oh SW, Park CY, Lee ES, Yoon YS, Lee ES, Park SS, et al. Adipokines, insulin resistance, metabolic syndrome, and breast cancer recurrence: a cohort study. Breast Cancer Res 2011;13:R34.
    Pubmed KoreaMed CrossRef
  42. Russo S, Kwiatkowski M, Govorukhina N, Bischoff R, Melgert BN. Meta-inflammation and metabolic reprogramming of macrophages in diabetes and obesity: the importance of metabolites. Front Immunol 2021;12:746151.
    Pubmed KoreaMed CrossRef
  43. Festa A, D'Agostino R Jr, Williams K, Karter AJ, Mayer-Davis EJ, Tracy RP, et al. The relation of body fat mass and distribution to markers of chronic inflammation. Int J Obes Relat Metab Disord 2001;25:1407-15.
    Pubmed CrossRef
  44. Le CT, Khuat LT, Caryotakis SE, Wang M, Dunai C, Nguyen AV, et al. PD-1 blockade reverses obesity-mediated T cell priming impairment. Front Immunol 2020;11:590568.
    Pubmed KoreaMed CrossRef
  45. Vonderheide RH, Domchek SM, Clark AS. Immunotherapy for breast cancer: what are we missing? Clin Cancer Res 2017;23:2640-6.
    Pubmed KoreaMed CrossRef
  46. Qiu Y, Yang Y, Yang R, Liu C, Hsu JM, Jiang Z, et al. Activated T cell-derived exosomal PD-1 attenuates PD-L1-induced immune dysfunction in triple-negative breast cancer. Oncogene 2021;40:4992-5001.
    Pubmed KoreaMed CrossRef
  47. Mittendorf EA, Philips AV, Meric-Bernstam F, Qiao N, Wu Y, Harrington S, et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res 2014;2:361-70.
    Pubmed KoreaMed CrossRef
  48. Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P, et al. RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors. Clin Cancer Res 2016;22:1499-509.
    Pubmed KoreaMed CrossRef
  49. Denduluri SK, Idowu O, Wang Z, Liao Z, Yan Z, Mohammed MK, et al. Insulin-like growth factor (IGF) signaling in tumorigenesis and the development of cancer drug resistance. Genes Dis 2015;2:13-25.
    Pubmed KoreaMed CrossRef
  50. Min DY, Jung E, Kim J, Lee YH, Shin SY. Leptin stimulates IGF-1 transcription by activating AP-1 in human breast cancer cells. BMB Rep 2019;52:385-90.
    Pubmed KoreaMed CrossRef
  51. Shen L, O'Shea JM, Kaadige MR, Cunha S, Wilde BR, Cohen AL, et al. Metabolic reprogramming in triple-negative breast cancer through Myc suppression of TXNIP. Proc Natl Acad Sci U S A 2015;112:5425-30.
    Pubmed KoreaMed CrossRef
  52. Han J, Zhang L, Guo H, Wysham WZ, Roque DR, Willson AK, et al. Glucose promotes cell proliferation, glucose uptake and invasion in endometrial cancer cells via AMPK/mTOR/S6 and MAPK signaling. Gynecol Oncol 2015;138:668-75.
    Pubmed KoreaMed CrossRef
  53. Michelet X, Dyck L, Hogan A, Loftus RM, Duquette D, Wei K, et al. Metabolic reprogramming of natural killer cells in obesity limits antitumor responses. Nat Immunol 2018;19:1330-40.
    Pubmed CrossRef
  54. Bähr I, Jahn J, Zipprich A, Pahlow I, Spielmann J, Kielstein H. Impaired natural killer cell subset phenotypes in human obesity. Immunol Res 2018;66:234-44.
    Pubmed KoreaMed CrossRef
  55. Thacker G, Henry S, Nandi A, Debnath R, Singh S, Nayak A, et al. Immature natural killer cells promote progression of triple-negative breast cancer. Sci Transl Med 2023;15:eabl4414.
    Pubmed CrossRef
  56. Park IH, Yang HN, Lee KJ, Kim TS, Lee ES, Jung SY, et al. Tumor-derived IL-18 induces PD-1 expression on immunosuppressive NK cells in triple-negative breast cancer. Oncotarget 2017;8:32722-30.
    Pubmed KoreaMed CrossRef
  57. Mamessier E, Sylvain A, Thibult ML, Houvenaeghel G, Jacquemier J, Castellano R, et al. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest 2011;121:3609-22.
    Pubmed KoreaMed CrossRef
  58. Nakajima I, Yamaguchi T, Ozutsumi K, Aso H. Adipose tissue extracellular matrix: newly organized by adipocytes during differentiation. Differentiation 1998;63:193-200.
    Pubmed CrossRef
  59. García-Alonso V, Titos E, Alcaraz-Quiles J, Rius B, Lopategi A, López-Vicario C, et al. Prostaglandin E2 exerts multiple regulatory actions on human obese adipose tissue remodeling, inflammation, adaptive thermogenesis and lipolysis. PLoS One 2016;11:e0153751.
    Pubmed KoreaMed CrossRef
  60. de Sousa Neto IV, Durigan JL, da Silva AS, de Cássia Marqueti R. Adipose tissue extracellular matrix remodeling in response to dietary patterns and exercise: molecular landscape, mechanistic insights, and therapeutic approaches. Biology (Basel) 2022;11:765.
    Pubmed KoreaMed CrossRef
  61. Seo BR, Bhardwaj P, Choi S, Gonzalez J, Andresen Eguiluz RC, Wang K, et al. Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis. Sci Transl Med 2015;7:301ra130.
    CrossRef
  62. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 2009;139:891-906.
    Pubmed KoreaMed CrossRef
  63. Tharp KM, Weaver VM. Modeling tissue polarity in context. J Mol Biol 2018;430:3613-28.
    Pubmed KoreaMed CrossRef
  64. Wang M, Yang Y, Han L, Han S, Liu N, Xu F, et al. Effect of three-dimensional ECM stiffness on cancer cell migration through regulating cell volume homeostasis. Biochem Biophys Res Commun 2020;528:459-65.
    Pubmed CrossRef
  65. Acerbi I, Cassereau L, Dean I, Shi Q, Au A, Park C, et al. Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr Biol (Camb) 2015;7:1120-34.
    Pubmed KoreaMed CrossRef
  66. Chamberlin T, Clack M, Silvers C, Kuziel G, Thompson V, Johnson H, et al. Targeting obesity-induced macrophages during preneoplastic growth promotes mammary epithelial stem/progenitor activity, DNA damage, and tumor formation. Cancer Res 2020;80:4465-75.
    Pubmed KoreaMed CrossRef
  67. Wishart AL, Conner SJ, Guarin JR, Fatherree JP, Peng Y, McGinn RA, et al. Decellularized extracellular matrix scaffolds identify full-length collagen VI as a driver of breast cancer cell invasion in obesity and metastasis. Sci Adv 2020;6:eabc3175.
    Pubmed KoreaMed CrossRef
  68. Li W, Zhang X, Wang J, Li M, Cao C, Tan J, et al. TGFβ1 in fibroblasts-derived exosomes promotes epithelial-mesenchymal transition of ovarian cancer cells. Oncotarget 2017;8:96035-47.
    Pubmed KoreaMed CrossRef
  69. Felicidade I, Bocchi M, Ramos MR, Carlos LO, Wagner NR, Campos AC, et al. Transforming growth factor beta 1 (TGFβ1) plasmatic levels and haplotype structures in obesity: a role for TGFβ1 in steatosis development. Mol Biol Rep 2021;48:6401-11.
    Pubmed CrossRef
  70. Liu Y, Wang X, Wang Z, Ju W, Wang D. Decorin inhibits the proliferation of HepG2 cells by elevating the expression of transforming growth factor-β receptor II. Exp Ther Med 2016;12:2191-5.
    Pubmed KoreaMed CrossRef
  71. Wishart A, Peng Y, Conner S, Guarin J, Crews R, Fatherree J, et al. Obesity-driven changes in ECM composition promote local invasion and metastasis of breast tumors. bioRxiv [Preprint] 2020 Jan 29. https://doi.org/10.1101/2020.01.29.924431
    CrossRef
  72. Reggio S, Pellegrinelli V, Clément K, Tordjman J. Fibrosis as a cause or a consequence of white adipose tissue inflammation in obesity. Curr Obes Rep 2013;2:1-9.
    CrossRef
  73. He Q, Gao Z, Yin J, Zhang J, Yun Z, Ye J. Regulation of HIF-1{alpha} activity in adipose tissue by obesity-associated factors: adipogenesis, insulin, and hypoxia. Am J Physiol Endocrinol Metab 2011;300:E877-85.
    Pubmed KoreaMed CrossRef
  74. Cha YJ, Youk JH, Kim BG, Jung WH, Cho NH. Lymphangiogenesis in breast cancer correlates with matrix stiffness on shear-wave elastography. Yonsei Med J 2016;57:599-605.
    Pubmed KoreaMed CrossRef
  75. Ebright RY, Zachariah MA, Micalizzi DS, Wittner BS, Niederhoffer KL, Nieman LT, et al. HIF1A signaling selectively supports proliferation of breast cancer in the brain. Nat Commun 2020;11:6311.
    Pubmed KoreaMed CrossRef
  76. Montagner M, Enzo E, Forcato M, Zanconato F, Parenti A, Rampazzo E, et al. SHARP1 suppresses breast cancer metastasis by promoting degradation of hypoxia-inducible factors. Nature 2012;487:380-4.
    Pubmed CrossRef
  77. Fournier F, Diaz-Marin R, Pilon F, Neault M, Juneau R, Girouard G, et al. Obesity triggers tumoral senescence and renders poorly immunogenic malignancies amenable to senolysis. Proc Natl Acad Sci U S A 2023;120:e2209973120.
    Pubmed KoreaMed CrossRef
  78. Abid S, Adnot S, Agusti A, Beaulieu D, Bhushan A, Bird TG, et al. Contributors. In: Serrano M, Muñoz-Espín D, editors. Cellular senescence in disease. Academic Press; 2022. p. xv-xix.
    CrossRef
  79. Espinosa De Ycaza AE, Søndergaard E, Morgan-Bathke M, Carranza Leon BG, Lytle KA, Ramos P, et al. Senescent cells in human adipose tissue: a cross-sectional study. Obesity (Silver Spring) 2021;29:1320-7.
    Pubmed KoreaMed CrossRef
  80. Georget M, Defois A, Guiho R, Bon N, Allain S, Boyer C, et al. Development of a DNA damage-induced senescence model in osteoarthritic chondrocytes. Aging (Albany NY) 2023;15:8576-93.
    Pubmed KoreaMed CrossRef
  81. Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of human fibroblasts. Nature 1990;345:458-60.
    Pubmed CrossRef
  82. Faraonio R. Oxidative stress and cell senescence process. Antioxidants (Basel) 2022;11:1718.
    Pubmed KoreaMed CrossRef
  83. Goutas A, Outskouni Z, Papathanasiou I, Georgakopoulou A, Karpetas GE, Gonos ES, et al. The establishment of mitotic errors-driven senescence depends on autophagy. Redox Biol 2023;62:102701.
    Pubmed KoreaMed CrossRef
  84. Velarde MC, Demaria M, Melov S, Campisi J. Pleiotropic age-dependent effects of mitochondrial dysfunction on epidermal stem cells. Proc Natl Acad Sci U S A 2015;112:10407-12.
    Pubmed KoreaMed CrossRef
  85. Ziegler DV, Martin N, Bernard D. Cellular senescence links mitochondria-ER contacts and aging. Commun Biol 2021;4:1323.
    Pubmed KoreaMed CrossRef
  86. Bartkova J, Rezaei N, Liontos M, Karakaidos P, Kletsas D, Issaeva N, et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 2006;444:633-7.
    Pubmed CrossRef
  87. Lee KJ, Mann E, Wright G, Piett CG, Nagel ZD, Gassman NR. Exploiting DNA repair defects in triple negative breast cancer to improve cell killing. Ther Adv Med Oncol 2020;12:1758835920958354.
    Pubmed KoreaMed CrossRef
  88. Budamagunta V, Manohar-Sindhu S, Yang Y, He Y, Traktuev DO, Foster TC, et al. Senescence-associated hyper-activation to inflammatory stimuli in vitro. Aging (Albany NY) 2021;13:19088-107.
    Pubmed KoreaMed CrossRef
  89. Basisty N, Kale A, Jeon OH, Kuehnemann C, Payne T, Rao C, et al. A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol 2020;18:e3000599.
    Pubmed KoreaMed CrossRef
  90. Coppé JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 2010;5:99-118.
    Pubmed KoreaMed CrossRef
  91. Bowers LW, Rossi EL, McDonell SB, Doerstling SS, Khatib SA, Lineberger CG, et al. Leptin signaling mediates obesity-associated CSC enrichment and EMT in preclinical TNBC models. Mol Cancer Res 2018;16:869-79.
    Pubmed KoreaMed CrossRef
  92. Wang W, Yang X, López de Silanes I, Carling D, Gorospe M. Increased AMP:ATP ratio and AMP-activated protein kinase activity during cellular senescence linked to reduced HuR function. J Biol Chem 2003;278:27016-23.
    Pubmed CrossRef
  93. Guo L, Xie B, Mao Z. Autophagy in premature senescent cells is activated via AMPK pathway. Int J Mol Sci 2012;13:3563-82.
    Pubmed KoreaMed CrossRef
  94. Matos L, Gouveia AM, Almeida H. ER stress response in human cellular models of senescence. J Gerontol A Biol Sci Med Sci 2015;70:924-35.
    Pubmed CrossRef
  95. Xu S, Cai Y, Wei Y. mTOR signaling from cellular senescence to organismal aging. Aging Dis 2013;5:263-73.
    Pubmed KoreaMed CrossRef
  96. Herranz N, Gallage S, Mellone M, Wuestefeld T, Klotz S, Hanley CJ, et al. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat Cell Biol 2015;17:1205-17.
    Pubmed KoreaMed CrossRef
  97. Fang J, Li L, Cao X, Yue H, Fu W, Chen Y, et al. Transmissible endoplasmic reticulum stress mediated by extracellular vesicles from adipocyte promoting the senescence of adipose-derived mesenchymal stem cells in hypertrophic obesity. Oxid Med Cell Longev 2022;2022:7175027.
    Pubmed KoreaMed CrossRef
  98. Nan J, Hu X, Guo B, Xu M, Yao Y. Inhibition of endoplasmic reticulum stress alleviates triple-negative breast cancer cell viability, migration, and invasion by syntenin/SOX4/Wnt/β-catenin pathway via regulation of heat shock protein A4. Bioengineered 2022;13:10564-77.
    Pubmed KoreaMed CrossRef