Next Article in Journal
Effects on Bioaccumulation, Growth Performance, Hematological Parameters, Plasma Components, and Antioxidant Responses in Starry Flounder (Platichthys stellatus) Exposed to Dietary Cadmium and Ascorbic Acid
Next Article in Special Issue
Study of Ferroptosis Transmission by Small Extracellular Vesicles in Epithelial Ovarian Cancer Cells
Previous Article in Journal
Reduced Ribose-5-Phosphate Isomerase A-1 Expression in Specific Neurons and Time Points Promotes Longevity in Caenorhabditis elegans
Previous Article in Special Issue
The Protective Anticancer Effect of Natural Lycopene Supercritical CO2 Watermelon Extracts in Adenocarcinoma Lung Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Adipocyte–Macrophage Relationship in Cancer: A Potential Target for Antioxidant Therapy

by
Sofía Sanhueza
1,2,3,†,
Layla Simón
1,2,4,†,
Mariana Cifuentes
2,3,* and
Andrew F. G. Quest
1,2,*
1
Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380492, Chile
2
Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago 8380492, Chile
3
Laboratory of Obesity and Metabolism in Geriatrics and Adults (OMEGA), Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago 7830490, Chile
4
Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501015, Chile
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2023, 12(1), 126; https://doi.org/10.3390/antiox12010126
Submission received: 16 November 2022 / Revised: 20 December 2022 / Accepted: 22 December 2022 / Published: 4 January 2023
(This article belongs to the Special Issue Oxidative Stress and Inflammation in Cancer)

Abstract

:
Obesity has emerged as a major public health concern with a staggering 39% worldwide prevalence as of 2021. Given the magnitude of the problem and considering its association with chronic low-grade systemic inflammation, it does not come as a surprise that obesity is now considered one of the major risk factors for the development of several chronic diseases, such as diabetes, cardiovascular problems, and cancer. Adipose tissue dysfunction in obesity has taken center stage in understanding how changes in its components, particularly adipocytes and macrophages, participate in such processes. In this review, we will initially focus on how changes in adipose tissue upon excess fat accumulation generate endocrine signals that promote cancer development. Moreover, the tumor microenvironment or stroma, which is also critical in cancer development, contains macrophages and adipocytes, which, in reciprocal paracrine communication with cancer cells, generate relevant signals. We will discuss how paracrine signaling in the tumor microenvironment between cancer cells, macrophages, and adipocytes favors cancer development and progression. Finally, as reactive oxygen species participate in many of these signaling pathways, we will summarize the information available on how antioxidants can limit the effects of endocrine and paracrine signaling due to dysfunctional adipose tissue components in obesity.

1. Introduction

1.1. Obesity: A Major Worldwide Health Problem

Obesity is characterized by an excessive accumulation of white adipose tissue that elevates health risks and is considered a severe public health problem. The already epidemic proportions continue to grow throughout the world and are of great concern given that numerous diseases are either triggered or aggravated by this disorder. Indeed, the COVID-19 pandemic revealed that people with obesity were at greater risk for unfavorable outcomes related to disease severity and death [1]. Obesity is a complex multifactorial disease with etiological components that range from molecular to social factors, and given the well-established connection to the development of multiple chronic diseases, it is of utmost importance to better understand the molecular mechanisms involved in order to prevent or revert related comorbidities.

1.2. White Adipose Tissue Dysfunction and Disease Development: Role of Macrophages and Adipocytes

For a long time, adipose tissue was considered a simple energy reservoir; however, after the first reports in the 1990s on its ability to secrete numerous key regulatory factors, this tissue has come to be recognized as a major endocrine organ [2,3]. Besides the secretion of regulatory proteins, adipose tissue communicates with other organs and aids in maintaining homeostasis through the release of many different mediators, including lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs) [4]. Importantly, obesity is frequently characterized by adipose tissue dysfunction; it is often associated with infiltration by macrophages, ensuing changes that result in the liberation of many proinflammatory factors, insulin resistance, and impaired lipid metabolism. Local changes in adipose tissue are transmitted to various organs via this altered secretome and thereby contribute to deleterious forms of organ crosstalk typically associated with the development of chronic diseases, such as nonalcoholic fatty liver disease, cardiovascular disease, type 2 diabetes, and cancer.
Adipose tissue expansion due to excessive energy intake is accompanied by numerous changes in tissue function and composition [5]. Particularly, dysfunctional fat is characterized by inflammation, driven mainly by the infiltration of proinflammatory CD11c-expressing M1-polarized macrophages. Once the tissue has been infiltrated, M1 macrophages and adipocytes establish a complex reciprocal communication network involving cytokines, chemokines, and microRNA-containing exosomes, among others, that precipitate functional dysregulation [6]. In addition, one key hallmark of adipose tissue inflammation is the presence of histological markers, termed crown-like structures (CLS), generated by proinflammatory macrophages surrounding dead or dying adipocytes [7].
In the context of this review, the fact that obesity has been associated with the development of several types of cancer, including endometrial, breast, ovarian, prostate, liver, gallbladder, kidney, and colon cancer, is of particular interest. Indeed, the aforementioned inflammatory changes that adipose tissue undergoes in obesity have been mechanistically linked to multiple aspects of cancer development.

1.3. Obesity-Associated Cancer

A 2016 report that analyzed the results from over 1000 epidemiological studies [8] determined that there is sufficient evidence in humans to conclude that the absence of excess body fat lowers the risk for thirteen sites/types of cancer, among which are esophagus adenocarcinoma, colon and rectum, liver, pancreas, breast (postmenopausal), and corpus uteri cancer. The study of the mechanisms involved in this relationship, which may be relevant to all cancers and/or particular to specific cases, is extremely important for identifying therapeutic targets and developing novel treatment strategies. However, given the wide variety of alterations involved in obesity-related pathophysiology on the one hand and the complexity of the mechanisms that are altered during different carcinogenic processes on the other, it is difficult to address all aspects in a single model. For instance, the development of some types of cancer may be more specifically favored by obesity-related alterations in sex hormone metabolism while others are stimulated by enhanced insulin and insulin growth factor signaling, altered adipokine levels, or elevated proinflammatory and/or oxidative status [9,10,11].
Interestingly, adipocytes in obesity suffer several subcellular/metabolic alterations, such as mitochondrial dysfunction and endoplasmic reticulum (ER) stress. These changes result in an increased production of reactive oxygen species (ROS) as a consequence of excess food consumption and the ensuing overload of nutrients that need to be processed [12,13,14,15]. In turn, such constantly elevated ROS production is expected to further damage cell components and threaten cell viability, exacerbating the aforementioned formation of CLS, thereby generating a microenvironment favorable for tumor progression due to its effects on cell proliferation, survival, invasion, and metastasis [16]. In addition, obesity leads to an excess of adipose tissue, which in turn favors inflammation, dyslipidemia, hyperglycemia, and, consequently, oxidative stress. For instance, obesity promotes oxidative stress and favors tumor development by inducing DNA damage and genomic instability. Oxidative stress is further increased by mitochondrial alterations and potentiated by obesity-induced chronic inflammation. Moreover, obesity-induced dyslipidemia promotes lipid peroxidation and DNA mutations. Finally, obesity-induced hyperglycemia increases insulin signaling, glycolytic pathway activity, oxidative stress, and DNA alterations (reviewed in [17,18]).

1.4. Tumor Microenvironment: Focus on Adipocytes and Macrophages

As mentioned above, adipose tissue in obesity may undergo several structural, metabolic, and functional changes that create a proinflammatory and -oxidative milieu within the tissue microenvironment. These local changes involving cell–cell and cell–extracellular matrix crosstalk are thought to be particularly relevant in tumors that are in close proximity to adipose tissue, as is the case for breast cancer [19].
Multiple players in the obese adipose tissue microenvironment promote insulin resistance and elevate local insulin levels, which then can enhance phosphoinositide 3-kinase (PI3K) signaling and promote tumorigenesis [20,21]. Moreover, increased vascular endothelial growth factor (VEGF) production associated with adipose extracellular matrix remodeling likely facilitates tumor angiogenesis [22]. This matrix remodeling together with interstitial fibrosis in mammary adipose tissue has been shown to promote breast tumorigenesis by altering mammary epithelial cell malignancy and matrix mechanics [23]. In addition, factors secreted from adipose tissue that are elevated in obesity, such as leptin, visfatin, or those contained in EVs, have been shown to promote processes associated with tumor development, such as angiogenesis, invasion, viability, metalloproteinase activation, and ROS production, and do so by elevating ERK and PI3K/Akt signaling among other mechanisms [24,25,26].

1.5. Adipose Derangements and the Tumor Microenvironment as Potential Therapeutic Targets

Many of the proposed mechanisms linking obesity to cancer development involve well-known adipose tissue derangements that occur during excess weight gain. Consequently, therapeutic approaches aimed at decreasing adipose dysfunction are promising in order to uncouple these events. In particular, supplementation with natural or synthetic antioxidants is one of these strategies. For instance, obesity-induced metabolic disorders in rat models have been prevented or ameliorated by treatments with natural antioxidants, such as rutin [27], which also suppressed the formation of CLS and macrophage ROS production. Another study showed that dietary supplementation with oleuropein (an abundant phenolic compound in olives) in diet-induced obese mice decreased the levels of the angiogenic factor VEGF-A and suppressed the induction of solid tumor growth as well as lymph node metastasis [28]. In addition, the natural polyphenolic compound resveratrol has been shown to reduce or prevent many cancer-inducing effects related to the hyperglycemic conditions frequently observed in obesity, such as cell proliferation, ROS production, signaling cascades involved in tumor migration and invasion (ERK and p38 MAPK) as well as enhanced expression of factors relevant to cell migration, invasion, and metastasis [29]. Moreover, treatment of Glut1-overexpressing macrophages from diet-induced obese mice with N-acetylcysteine to quench ROS abolished the upregulation of the proinflammatory plasminogen activator inhibitor 1 (PAI-1) [30]. These few examples highlight the utility of antioxidants as potential therapeutic tools to prevent obesity-induced inflammation in cancer development. In the following sections, we will review the currently available information relating to endocrine and paracrine communication between adipose tissue and macrophages with a focus on their importance in tumor growth and the potential of different therapeutic approaches using antioxidants for delaying or preventing cancer development. The aspects that will be covered in this review are graphically summarized in Figure 1.

2. Endocrine Communication between Adipose Tissue and the Tumor Microenvironment

Obesity is associated with an increased risk of developing several types of cancer due to, among other mechanisms, chronic inflammation and metabolic dysregulation. Inflammation-related cytokines, such as interleukin-6 (IL-6) and C-reactive protein (CRP), as well as changes in metabolism-related adipokines, such as adiponectin and leptin, have been shown to reflect a proinflammatory microenvironment that promotes tumor development and progression. Patients with a normal weight tend to have higher levels of the anti-inflammatory and insulin-sensitizing adipokine adiponectin while those who are overweight or obese have higher leptin, IL-6 and CRP levels. In addition, elevated IL-6 is associated with increased cancer mortality in patients with obesity [31]. In the following paragraphs, we will focus on the mechanisms connecting adipose tissue to the tumor microenvironment as summarized in Table 1.
Obese CLS+ tissue donors showed increased liver fat deposition and circulating tumor necrosis factor-alpha (TNFα) levels as well as insulin resistance, which are all important systemic consequences of disrupted adipose tissue homeostasis. Pathway analysis of differentially expressed genes in adipose tissue revealed the upregulation of various molecules belonging to the proinflammatory nuclear factor kappa B (NF-κB) signaling pathway in CLS+ explants [7]. Overweight and obese breast cancer patients develop CLS+ and adipose tissue inflammation, which is associated with markers of metabolic derangements, such as increased lipid, glucose and CRP levels in serum [42]. Moreover, breast cancer patients with adipose inflammation have increased insulin, glucose, leptin, triglycerides, CRP, and IL-6 levels as well as decreased HDL and adiponectin levels. Furthermore, obesity, metabolic syndrome and the circulation of proinflammatory molecules are associated with poorer prognosis in breast cancer patients. Indeed, adipose tissue inflammation reduces the distant recurrence-free survival of these patients [43].
In lung cancer, there is also a strong association between overweight/obesity, lipid alterations, and oxidative stress. Indeed, the total cholesterol/HDL ratio, triglyceride levels, lipid peroxidation and oxidative stress are increased in lung cancer patients compared to healthy people. Moreover, these alterations increase in lung cancer patients who are overweight and obese. These data suggest that obesity could enhance lung cancer-induced systemic changes, such as lipid and redox imbalance [47].
A high-fat diet (HFD) increases the myeloid-derived suppressor cell (MDSC) fraction, a heterogeneous population of cells of myeloid origin that mediates the suppression of T cells and contributes to the inhibition of immune responses during cancer [48,50]. In addition, a HFD enhances the M2/M1 macrophage ratio and accelerates prostate cancer tumor growth via IL-6/STAT3 signaling [48]. Furthermore, adipose tissue-derived factors observed in mice on a HFD stimulate the progression of prostate cancer via endocrine and paracrine mechanisms. For instance, HFD-induced obese mice showed increases in VEGF, IL-6, chemokine ligand 2 (CCL2), and C-X-C motif chemokine ligand 1/2/13 (CXCL1/2/13), which promote angiogenesis and the infiltration of immune cells that favor tumor development and metastasis [49].
Interestingly, statistical modeling of microRNA-mediated gene regulation identified common interactions in obesity, liver and uterine cancers. For instance, the upregulation of genes encoding for proteins involved in key metabolic processes, such as fatty acid and arachidonic acid metabolism, as well as signaling pathways related to cell growth and inflammation, are observed in all cases [35]. Furthermore, in endometrial cancer patients, body mass index (BMI), leptin, IL-6 and oxidative stress correlate with pathogenesis and poor prognosis [36]. Likewise, in breast cancer, several factors secreted by adipose tissue augment cancer cell aggressiveness. For instance, leptin secreted by adipose stromal/stem cells isolated from women with obesity enhances the expression of epithelial-to-mesenchymal transition (EMT) and metastasis-related genes, such as serpine 1, matrix metalloproteinase 2 (MMP-2) and IL-6 [37]. In addition, the reciprocal relationship between adipocytes, immune cells and cancer cells induces a systemically immunosuppressive macroenvironment that promotes breast cancer development. Moreover, breast cancer cells implanted in a mouse model of diet-induced obesity modified the visceral adipose tissue, spleen and tumor microenvironment in order to create an immunosuppressive milieu that promoted cancer development. The characteristic increase in the M1/M2 macrophage ratio reported in the visceral adipose tissue in obesity was completely reversed in tumor-bearing mice, resulting in a predominantly M2-polarized profile typically observed in the tumor microenvironment. Moreover, Tregs were decreased in obese mice, but increased in visceral adipose tissue in the presence of tumors. Altogether, these results suggest the existence of a regulatory feedback mechanism between the growing tumor and adipose tissue, which generates local and systemic immunosuppressive conditions that promote tumor development [38].
In addition to the aforementioned mechanisms, there is a link between obesity and breast cancer that includes a connection between adipocytes, endothelial and immune cells. The obese tumor microenvironment recruits macrophages that activate the NOD-like receptor 4 (NLRC4) inflammasome, leading to interleukin-1β (IL-1β) activation. IL-1β promotes breast cancer progression by increasing adipocyte VEGF secretion and angiogenesis [39]. Moreover, angiopoietin-like 4 (ANGPTL4), a known proangiogenic factor in cancer, is induced by IL-1β from adipocytes in a manner dependent on NF-κB and MAPK. This mechanism is further stimulated by hypoxia. In this way, adipocyte-secreted ANGPTL4 promoted angiogenesis and breast cancer progression in obese mice [40]. Furthermore, factors secreted by adipose tissue from HFD-induced obese mice and the fat tissue of female patients with obesity led to the upregulation of genes involved in inflammation and lipid metabolism, such as PPARα, IL-1β and ANGPTL4 in triple-negative breast cancer cells. Moreover, focal adhesion kinase (FAK) activation increased in these cancer cells, resulting in elevated lipid metabolism, wound healing, proliferation, and invasion [41].
As discussed in the previous paragraphs of this section, obesity and inflammation promote not only postmenopausal breast cancer progression but also metastasis. Adipocyte progenitors (CD45-CD34+) in xenograft and breast cancer models secrete granulocyte-macrophage colony-stimulating factor (GM-CSF) and matrix metalloproteinase 9 (MMP-9), thereby inducing immunosuppression, intratumor vascularization, as well as local and metastatic tumor growth [44]. Moreover, obesity increases lung neutrophilia and breast cancer metastasis to the lung in a GM-CSF and IL-5-dependent manner [45]. In addition, obesity induces the expression of sphingosine-1-phosphate (S1P), a bioactive sphingolipid mediator of breast cancer pathogenesis. Obesity enhances the expression of sphingosine kinase 1 (SphK1), the enzyme that produces S1P and its receptor S1PR1. Indeed, obesity-induced breast tumors increase SphK1 and secrete S1P, which promotes macrophage recruitment through S1PR1 in lung premetastatic niches. Macrophages secrete S1P, IL-6 and TNFα, thereby promoting breast cancer metastasis [46].
Regarding the role of specific proinflammatory cytokines and adipokines, a study in a mouse model of chemically induced colitis-associated colorectal cancer (CAC) showed that diet-induced obesity increased IL-6 levels, thereby favoring macrophage polarization towards tumor-promoting M2 macrophages. Moreover, M2 macrophages produced CC-chemokine-ligand-20 (CCL-20) that promoted the recruitment of CC-chemokine-receptor-6 (CCR-6)-expressing B and T cells. The recruited B cells promoted further macrophage polarization and the T cells inhibited other immune cells, thereby favoring CAC progression [32]. Survivin is also secreted by adipose tissue and drives protumoral macrophage polarization in colon cancer, colorectal adenocarcinoma and hepatocellular carcinoma. Indeed, adipose-derived stem cells (ASCs) from subjects with obesity release survivin that induces tumor-associated macrophage (TAM) reprogramming coincident with increased survivin expression. Thus, survivin produced by ASCs and macrophages together promotes the malignancy of colon and liver cancer cells [33]. Obesity has also been associated with the secretion of adipokines and cytokines in the liver tumor microenvironment. The proinflammatory adipokines visfatin and resistin are increased in the serum and liver tumors of patients with obesity. Moreover, visfatin and resistin increase cell viability, invasion, lipogenesis, fatty acid synthase (FASN) protein levels, MMP-9 activity, as well as Akt and ERK phosphorylation in liver cancer cells [11]. In a mouse model of hepatocellular carcinoma initiated by diethylnitrosamine administration and promoted by dietary or genetic obesity, PI3Kγ ablation reduced tumor growth, insulinemia, and steatosis [34], suggesting a preponderant role for PI3K/Akt signaling in obesity-induced cancer.

3. Paracrine Communication between Adipocytes, Macrophages and Cancer Cells in the Tumor Microenvironment

Tumors are now recognized as containing many different cell populations beyond cancer cells, including fibroblasts, immune cells (macrophages, lymphocytes, neutrophils), endothelial cells and adipocytes, among others [51,52]. Moreover, it is increasingly evident that these additional populations of tumor stromal cells promote extracellular matrix remodeling, cell migration, angiogenesis, invasion, metastasis, and drug resistance by producing a variety of signaling molecules that include extracellular matrix constituents, growth factors, metabolites, chemokines and cytokines [53]. Therefore, to determine what drives tumor development, it is necessary to understand how the complex communication between tumor cells and the surrounding stroma components conspires to produce “factors” that augment inflammation in the tumor microenvironment (TME), thereby favoring tumor progression and metastasis. Note that the mechanisms described in the following paragraphs are summarized in Table 2.
Adipocytes play a fundamental role in the TME of multiple types of cancer, especially in those with significant adipocyte content in the stromal microenvironment. Available evidence indicates that dynamic communication between tumor cells and adipocytes leads to phenotypic and functional changes in both cell types that can drive tumor progression [64,66,88]. This section will consider some of the crosstalk mechanisms between adipocytes and cancer cells in the TME.

3.1. Effects of Adipocytes on Cancer Cells

As mentioned above, communication between cancer cells and adipocytes is reciprocal. Adipocytes have been shown to produce factors involved in matrix remodeling, metabolic reprogramming, EMT, angiogenesis, invasion and the survival of cancer cells as well as drug resistance [56,68,76,79,89].
Cancer-associated adipocytes (CAAs) produce elevated insulin-like growth factor binding protein 2 (IGFBP-2) levels in the tumor microenvironment, which upregulates MMP-2 and then promotes migration, invasion, and metastasis in breast cancer [58]. Likewise, CAAs cocultured with breast cancer cells have been shown to increase their expression of MMP-11 as well as proinflammatory cytokines (IL-6 and IL-1β) and thereby promote tumor cell invasion and metastasis [64]. Other investigators demonstrated that cancer cells cultured with in vitro differentiated adipocytes or proinflammatory cytokines (IL-6, IL-8, CXCL10, CCL2 or CCL5) activate Src to upregulate Sox2 and induce miR-302b, generating feed-forward loops that contribute to mammary cancer stem cell self-renewal and drive metastatic tumor progression [61]. Moreover, the interaction between CAAs and breast cancer cells involves a positive feedback loop between the cytokine leukemia inhibitory factor (LIF) and CXCL subfamily chemokines, which promotes breast cancer invasion and metastasis [55]. Furthermore, the expression of factors involved in cancer development, such as MMP-9 [17] and interferon γ-inducible protein 30 [90], show the highest upregulation in CLS+ as compared to CLS- human adipose tissue explants [7]. Moreover, studies in high-fat diet-fed mice have shown that secreted factors from their CLS+ adipose tissue, which is rich in proinflammatory cytokines, induce ROS production, changes in cell morphology, loss of E-cadherin, proliferation and neoplastic transformation in the epidermal JB6 P+ cell line [75].
The evidence also suggests that adipocytes mediate proangiogenic events in several tumors. For instance, IL-1β secreted by macrophages induced ANGPTL4 expression in primary mouse adipocytes through activation of NF-κB and MAPKs, which is further enhanced by hypoxia. Consequently, adipocyte-derived ANGPTL4 promotes obesity-driven breast cancer progression and angiogenesis [39,40]. Additionally, CAAs secrete high levels of IL-8, which increases angiogenesis at the primary tumor site, induces a protumoral neutrophil phenotype, modifies the expression of cell-adhesion molecules in tumor cells and favors early dissemination of breast cancer [59]. Furthermore, Al-Khalaf et al. reported that CAAs secreted higher levels of IL-8, which was crucial for enhancing the proangiogenic effects of breast adipocytes [60].
Interestingly, it has been suggested that adipocytes induce metabolic reprogramming in tumor cells. Peritoneum-derived adipocytes induce lipid droplet accumulation and fatty acid oxidation (FAO) in gastric cancer cells through the upregulation of diacylglycerol acyltransferase 2 (DGAT2) in a CCAAT/enhancer-binding protein alpha (C/EBPα)-dependent manner [78]. The lipid-rich environment stimulates nicotinamide adenine dinucleotide phosphate (NADPH) production, promotes resistance to anoikis through ROS-dependent mechanisms, and supports HFD-induced peritoneal dissemination and lung metastasis in vivo [78]. In an adipose triglyceride lipase/hormone-sensitive lipase (ATGL/HSL-dependent manner, adipocytes release fatty acids to boost fatty acid metabolism in breast cancer cells through the increased expression of carnitine palmitoyl transferase I (CPT1A) together with proteins of the electron transport chain, resulting in increased proliferation, migration and invasion of breast cancer cells [65,66]. Mammary adipocytes stimulate metabolic remodeling in cancer cells. Indeed, fatty acid metabolism in cancer cells is uncoupled from ATP production, leading to the promotion of the Warburg effect and increased glycolysis, thereby promoting the proliferation, migration and invasion of cancer cells [66]. Consistently, CPT1A is upregulated in colon cancer cells following coculture with mature human adipocytes or exposure to fatty acids. Elevated CPT1A allows cancer cells to produce increasing levels of Ac-CoA through the mitochondrial FAO to promote activation of the β-catenin/Wnt signaling and cancer stem cell function [80]. Furthermore, human omental adipocytes may act as a source of oleic acid for gastric cancer cells in omental gastric cancer metastases. Oleic acid enhances the invasiveness of gastric cancer cells, as well as the expression of MMP-2, by activating the PI3K-Akt signaling pathway in a PTEN-independent manner [77]. Primary mammary gland-derived adipocytes promote the tumorigenicity of cancer cells that express monocarboxylate transporter 2 (MCT2) via β-hydroxybutyrate. In detail, β-hydroxybutyrate secreted from adipocytes increases histone H3K9 acetylation and induces IL-1β and lipocalin 2 (LCN2) expression to enhance tumorigenicity in MCT2-expressing breast cancer cells, which in turn is linked to poor prognosis in breast cancer patients [54].
The fatty acid receptor CD36 is overexpressed in many metastatic tumors, such as oral squamous cell carcinomas (OSCC), melanoma and breast cancers. In OSCC cell lines or patient-derived cells with low metastatic potential, high levels of CD36 significantly increased lymph node metastasis. Furthermore, metastatic potential depends on the increased expression of FAO-related genes and is boosted by an HFD in a CD36-dependent manner. Consistent with these data, CD36 expression strongly correlates with poor prognosis in lung, bladder and breast cancer patients [82]. Later, the same group observed that ovarian cancer cells cocultured with primary human omental adipocytes expressed high levels of CD36, thereby facilitating exogenous fatty acid uptake and ovarian cancer metastasis. Conversely, CD36 inhibition reduced microenvironment-derived fatty acid uptake in ovarian cancer cells, decreasing adipocyte-stimulated invasion and migration in vitro, as well as tumor growth in vivo [69]. These observations highlight the crucial role of lipid metabolism in tumor progression and metastasis.
Additionally, adipocytes can induce EMT in several types of cancer. In breast cancer cells, mature adipocytes induce the EMT phenotype and promote cancer cell migration, invasion and proliferation, which could be associated with the upregulation of MMP-9 and twist-related protein 1 (TWIST1) [57]. Melanoma cells cocultured with differentiated 3T3-L1 adipocyte cells increased their invasive ability by promoting the expression of EMT genes (SNAI1, MMP-9, TWIST, and vimentin) and decreased the expression of genes, such as E-cadherin and Kiss1 [73]. Similarly, the conditioned medium of CAAs has been shown to improve migration/invasion and chemoresistance, as well as to promote EMT in pancreatic cancer cells by the upregulation of serum amyloid A1 (SAA1) [81], which has been associated with poor prognosis in breast cancer [91].
Other studies related to therapy resistance established that the conditioned media from human adipocytes promote resistance of melanoma cells to chemotherapeutic drugs (cisplatin and docetaxel) and therapeutic agents targeting the PI3K/Akt and MEK/ERK pathways [74]. Similarly, another study showed that human adipocytes of both subcutaneous and visceral/omental origin secrete soluble factors that increase resistance to chemotherapeutic drugs in ovarian cancer cells by activating the Akt pathway [70]. Moreover, primary human omental adipocytes induce FABP4 expression in ovarian cancer cells and promote metastasis and Carboplatin resistance in ovarian cancer cells [67]. On the other hand, for breast cancer cells cocultured with in vitro differentiated murine adipocytes, a significant increase in IL-6 expression was observed, which elevated checkpoint kinase 1 (Chk1) phosphorylation and promoted radioresistance in tumor cells [62]. Together, these studies highlight the role of adipocytes in inducing responses in tumor cells that enhance therapy resistance.
Finally, recent studies showed that murine and human adipocytes release EVs that deliver enzymes and substrates (fatty acids) involved in FAO to melanoma cells, which subsequently improves mitochondrial metabolism and remodels the mitochondrial network to support melanoma migration and invasion [71,72]. Another study reported that EVs from CAAs delivered microRNA-21 (miR21) to ovarian cancer cells, where it suppresses apoptosis and induces chemoresistance to paclitaxel through the downregulation of miR21’s direct target apoptotic peptidase activator factor 1 (APAF1) [68]. Moreover, it has been reported that in vitro differentiated adipocytes as well as primary mammary adipocytes from patients with normal weight or with obesity confer a multidrug resistance phenotype to breast cancer cells by increasing the nuclear efflux of doxorubicin through a major vault protein (MVP)-dependent process as well as by expulsion from breast cancer cells via EVs [63].

3.2. Effects of Tumor-Associated Macrophages on Cancer Cells

As mentioned above, the communication between cancer cells and TAMs is bidirectional. In the TME, TAMs typically promote cancer cell proliferation, angiogenesis, metastasis, and other processes through various anti-inflammatory mechanisms [92].
TAMs are known to be strongly involved in tumor angiogenesis. For instance, it has been reported that the presence of M2-polarized TAMs and Tie2-expressing monocytes is associated with angiogenesis, as well as decreased overall and recurrence-free survival in human PDAC patients [86]. In pancreatic islet cancers, breast tumors and lung metastases, interleukin-4 (IL-4) induces cathepsin protease activity in macrophages. Consequently, cathepsins B and S supplied by TAMs promote tumor growth, angiogenesis and invasion in vivo, as well as markedly increase the invasiveness of cancer cells in vitro [85]. Others reported that the development of human breast cancer is strongly associated with overexpression of the Wnt family ligand Wnt7B in the tumor stroma and that isolated human breast carcinoma TAMs express Wnt7B. Furthermore, in mice, Wnt7b was shown to play a crucial role in the malignant progression of luminal breast cancer by promoting angiogenesis, tumor growth, progression, invasion and metastasis. Additionally, the authors attributed the failure to induce the angiogenic switch to the reduced VEGF-A mRNA and protein levels in vascular endothelial cells [83].
M2-like TAMs reportedly promote glycolysis and hypoxia in cancer cells, leading to changes in cellular metabolism that are necessary for tumor initiation and progression. In non-small cell lung cancer, TAMs secrete TNFα to promote glycolysis in tumor cells and activate both AMPK and peroxisome proliferator-activated coactivator gamma 1-alpha, which together augment tumor hypoxia by promoting mitochondrial oxygen consumption and increasing mitochondrial membrane potential [87]. Furthermore, in this same study, TAMs were found to significantly reduce T cell infiltration by upregulating programmed death ligand 1 (PD-L1) expression in tumors. Therefore, the immune mechanisms that are associated with PD-L1 expression and controlled by TAMs can be considered critical for tumor immune escape [93]. Importantly, lactate efflux plays a critical role in tumor–stroma communication. Cancer cell-derived lactate increases the secretion of CCL5 through Notch signaling in TAMs, and CCL5 in turn induces EMT and aerobic glycolysis in breast cancer cells. In addition, TGF-β signaling regulates the expression of the CCL5-CCR5 axis components, which induce aerobic glycolysis in cancer cells by activating AMPK signaling. These observations point towards a pivotal role for the CCL5-CCR5 axis in metabolic communication between cancer cells and macrophages [84].
Finally, differences between TAMs from primary and metastatic cancers have been reported. In the primary tumor, TNFα induces EMT and the expression of IL12Rβ, a subunit of the IL-35 receptor, to promote tumor cell migration and invasion. Instead, at metastatic sites, TAMs secrete interleukin-35 (IL-35) to activate the JAK2–STAT6–GATA3 signaling axis in cancer cells, which reverses EMT to facilitate tissue colonization by cancer cells [94]. Additionally, Morrissey et al. reported that tumor-derived EVs polarize premetastatic niche macrophages towards an immunosuppressive phenotype through NF-kB-dependent glycolytic reprogramming to favor tumor cell metastasis. These prometastatic macrophages are characterized by increased de novo synthesis of PD-L1, increased glucose uptake, and GLUT-1 expression as well as the increased conversion of pyruvate to lactate, which subsequently augments further PD-L1 expression [95].

4. Paracrine Communication between Cancer Cells, Adipocytes and Macrophages in the Tumor Microenvironment

Macrophages in the tumor microenvironment, termed TAMs, represent the largest immune cell population in most human solid tumors [96,97,98]. TAMs are implicated in tumor progression through various mechanisms, such as promoting tumor growth, angiogenesis, invasion and metastasis as well as mediating resistance to drug therapy and suppressing antitumor immune responses [99,100]. Cancer cells and macrophages communicate with one another to determine the local organization of the TME. Therefore, in this section, we will discuss studies that explain how cancer cells recruit macrophages and change their phenotype to TAMs and how TAMs then modulate the characteristics of cancer cells to favor tumor progression. Note that the mechanisms described in the following paragraphs are summarized in Table 3.

4.1. Effects of Cancer Cells on Adipocytes in the Tumor Microenvironment

In this section, we will focus on recent studies that explain how normal adipocytes are “transformed’ into CAAs by tumor cells and how adipocytes regulate cancer cell behavior in the TME, thereby supporting tumor progression. Several reports suggest that invasive tumor cells modify the phenotype of adipocytes by reprogramming them to become CAAs, which in turn stimulate the aggressive behavior of cancer cells [64,125]. CAAs are located primarily around the invasive front of tumors, that is, adjacent to cancer cells. Morphologically, CAAs are smaller in size than normal adipocytes and have irregular shapes and smaller lipid droplets [64,126]. Thus, CAAs exhibit a modified phenotype characterized by the loss of lipid content, a decrease in mature adipocyte markers, as well as changes in the expression of adipokines, cytokines, and inflammatory factors, such as leptin, matrix metalloproteinase-11 (MMP-11), CCL2, chemokine ligand 5 (CCL5), IL-6 and IL-1β [64,127]. Compared to normal mature adipocytes, CAAs possess biological characteristics that promote metabolic reprogramming of cancer cells, extracellular matrix remodeling, tumor growth, progression and metastasis. Importantly, the acquisition of many of these characteristics is often also linked to changes in the adipocyte secretome [64,66,125]. Moreover, Zoico et al. demonstrated that pancreatic tumors induce the dedifferentiation of adipocytes into fibroblast-like cells through a Wnt5a-dependent signaling pathway. Specifically, the coculture of pancreatic cancer cells and adipocytes induces the JAK/STAT3 pathway, leading to increased Wnt5a expression and adipocyte reprogramming [107]. Likewise, breast cancer cells secrete Wnt3a, which activates the Wnt/β-catenin pathway and induces the dedifferentiation of mouse and human adipocytes, which then acquire a CAA-like phenotype [103].
Wei et al. proposed that the interaction between CAAs and breast cancer cells promotes the reorganization of adipocyte-derived collagen in a procollagen-lysine 2-oxoglutarate 5-dioxygenase 2 (PLOD2)-dependent manner, facilitating breast cancer migration along aligned and clustered collagen fibers. Mechanistically, PAI-1 is secreted by breast cancer cells and promotes the expression of PLOD2 in adipocytes by activating the PI3K/AKT-FOXP1 axis. Consequently, PLOD2 activation induces the linear organization of collagen derived from CAAs, which favors tumor cell metastasis [102].
The tumor microenvironment may alter adipose lipid metabolism. Dirat et al. demonstrated that mature adipocytes cocultured with breast cancer cells exhibited an altered phenotype associated with a reduced lipid content and a decrease in adipocyte markers together with an overexpression of MMP-11 and proinflammatory cytokines (IL-6 and IL-1β) [64]. Additionally, breast cancer cells have been reported to drive lipolysis in adipocytes by stimulating the ATGL/HSL pathway [65], thus elevating local free fatty acids. Similarly, in the presence of ovarian cancer cells, adipocytes release significantly more free fatty acids than adipocytes growing alone, thereby increasing the availability of lipids in the tumor microenvironment. As a result, adipocytes provide fatty acids to cancer cells and drive β-oxidation, suggesting that they are converted into CAAs to provide metabolites for cancer cells. Moreover, hypoxic cancer cell-conditioned media potentiate the metabolic shifts in adipocytes by increasing lipolysis in a HIF-1α dependent manner. Thereby, adipocytes transfer lipids to breast and ovarian cancer cells and consequently increase cancer cell proliferation [128]. In addition, adipocytes cocultured with breast cancer cells increase the expression of HIF-1α and TGF-β. Cancer cells cocultured with adipocytes increase the levels of EMT-inducing transcription factors FOXC2 and TWIST1 in a HIF-1α adipocyte expression-dependent manner. Taken together, these results suggest that adipocytes under hypoxic conditions promote cancer occurrence, migration, invasion and metastasis [129]. Additionally, upregulation of fatty acid-binding protein 4 (FABP4, also known as aP2) was observed in omental metastases compared to primary ovarian tumors. This suggests that FABP4 could be key in the ovarian cancer cell–adipocyte interaction, resulting in elevated lipid availability to favor metastasis [106]. In patients with colorectal cancer, a significant reduction in gene expression and activity levels of lipoprotein lipase (LPL) and FASN was detected in adipose tissue adjacent to the tumor lesion compared to tissue distant from the neoplasm [108]. Taken together, these results highlight the importance of the tumor microenvironment in adipose tissue lipid metabolism, as they provide evidence for tumor-induced deterioration in the lipid storage capacity of adipose tissue in patients with colorectal cancer.
In the last decades, the phenomenon of white adipose tissue browning has gained increasing interest in the obesity field. In this process, white adipocytes acquire a thermogenic phenotype in response to stimuli, such as cold or beta-adrenergic agonists [130]. A few studies support the idea that adipocytes adjacent to cancer cells display higher browning activity, which may contribute to tumorigenesis. Specifically, it has been demonstrated that local activation of tumor-adjacent thermogenic adipocytes could promote cancer cell proliferation and invasion by providing fuel sources [131]. Master et al. demonstrated that the mammary gland contains brown adipose tissue, which is potentially implicated in adaptive thermogenesis [132]. Other studies have shown that the expression of genes encoding for markers of fat browning, such as uncoupling protein 1 (UCP1), PR domain-containing 16 (PRDM16), cell death-inducing DFFA-like effector protein A (CIDEA), cytochrome c oxidase subunit VIIa polypeptide 1 (COX7A1), peroxisome proliferator-activated receptor gamma coactivator 1α, transmembrane protein 26 (TMEM26), and T-box 1 (TBX1), were upregulated in adipose tissue adjacent to breast tumors. Furthermore, the browning activity of adipocytes adjacent to breast cancer tissues was higher compared to those adjacent to benign breast lesions [104]. More recently, tumor cells have been shown to induce beige/brown phenotype and remodel metabolism in resident adipocytes by EVs with elevated miRNA-144 and miRNA-126 content. miRNA-144 promotes the beige/brown phenotype in adipocytes by downregulating the MAP3K8/ERK1/2/PPARγ axis, while miRNA-126 remodels metabolism by disrupting IRS/Glut-4 signaling, activating the AMP-activated protein kinase (AMPK)/autophagy pathway and stabilizing HIF1α expression [101]. Likewise, miR-155 derived from breast cancer cell exosomes promotes browning and metabolic remodeling of adipocytes through the downregulation of PPARγ, which triggers cancer-associated cachexia and facilitates tumor progression [105].

4.2. Effects of Cancer Cells on Macrophages in the Tumor Microenvironment

TAMs originate from two primary sources: (a) tissue-resident macrophages that develop from embryonic precursors (fetal yolk sack or fetal liver progenitors) and (b) macrophages derived from circulating monocytes that originate from bone marrow hemopoietic cell progenitors [133]. The TME affects the programming of both resident and infiltrating macrophages such that they acquire functionally distinct phenotypes [134,135]. As mentioned in previous sections, two well-established polarized phenotypes are the classically activated (M1) and the alternatively activated (M2) macrophages [136].
The M1-like polarized macrophages are activated by Th1 cytokines, such as interferon-γ (IFN-γ), or by exposure to lipopolysaccharides (LPS). Classically activated macrophages are responsible for generating a proinflammatory response directed against pathogens by incrementing phagocytic activity combined with the secretion of proinflammatory cytokines (IL-6, interleukin-12 (IL-12), interleukin-23 (IL-23), TNF-α) [137], as well as chemokines (CXCL1–3, CXCL-5, CXCL8–10) [138]. Moreover, they are characterized by a high antigen presentation capacity and produce proinflammatory mediators, such as nitric oxide and ROS [139,140]. After recognition by M1 macrophages, tumor cells can be killed through several mechanisms, including contact-dependent phagocytosis and cytotoxicity [141]. In contrast, M2 macrophages are activated by cytokines, such as IL-4, IL-13, glucocorticoids and immunoglobulin complexes that are anti-inflammatory and regulate tissue remodeling, angiogenesis, wound healing, parasite clearance and immunosuppression [100,142]. The transition of macrophages to the M2 phenotype facilitates tumor development, specifically through the production of anti-inflammatory cytokines (IL-10, arginase-1, TGF-β), as well as proangiogenic factors (CCL2, placental growth factor (PGF) and VEGF-A), which together favor tumor growth and metastasis [92,143,144].
Various studies have demonstrated that TAMs in the TME develop tumor-favoring activities similar to those of M2 macrophages [100], including promoting angiogenesis and matrix remodeling, as well as suppressing adaptive immunity. Moreover, they facilitate tumor progression through the secretion of several pro-angiogenic factors, extracellular matrix remodeling molecules, and anti-inflammatory cytokines [145,146]. Notably, the tumor promoting role of TAMs in cancer is supported by clinical studies that observed a positive correlation between the extent of macrophage presence in tumors and poor patient prognosis [147,148].
Several microenvironment signals derived from tumor and stromal cells directly affect monocyte recruitment and macrophage polarization. For instance, hypoxia can be a crucial driver of macrophage recruitment and the tumor-supporting transformation of TAMs [149]. In this context, it has been reported that hypoxia increases ZEB1 expression in cervical cancer cells, which directly promotes CCL8 production and induces macrophage infiltration, mainly into hypoxic areas, via the CCR2–NF-κB pathway. On the other hand, CCL8-mediated TAM infiltration contributes to hypoxic ZEB1-related cancer progression and poor prognosis in cervical cancer [114]. Similarly, it has been shown that hypoxic cervical cancer cells instruct the recruited macrophages to transform into M2-like TAMs by upregulating Neuropilin-1 (Nrp-1) in hypoxic regions. The presence of Nrp-1 and M2-like TAMs correlated with an increase in the malignant properties of cervical cancer cells, such as lymph node metastasis and poor tumor differentiation [113]. In breast cancer, hypoxic cancer cells reportedly attract macrophages and induce their M2 polarization by secreting eotaxin and oncostatin M [110]. Damage-associated molecular pattern (DAMP) and high-mobility group box 1 (HMGB1) proteins are also associated with hypoxia-induced macrophage polarization. Huber et al. reported that HMGB1 is released by melanomas in hypoxia and promotes the M2-like TAM phenotype, as well as IL-10 accumulation within a tumor [121]. In addition, 5-lipoxygenase (5-LOX), a member of the lipoxygenase gene family, is a crucial enzyme which converts arachidonic acid to 5-hydroxyeicosatetraenoic acid (5-HETE) and leukotrienes, lipid mediators with essential roles in tumor-associated inflammation [150]. Wen et al. found that the expression of 5-LOX strongly correlated with the density of TAMs in hypoxic areas of human ovarian tumors and provided in vitro evidence that increased 5-LOX metabolites from hypoxic ovarian cancer cells promote TAM infiltration through the upregulation of MMP-7 [112]. Finally, in hypoxic tumor microenvironments, various chemoattractants, such as endothelin-2 [151], VEGF-A [152], and endothelial-monocyte activating polypeptide (EMAPII) [153], also play a potential role in regulating macrophage recruitment and differentiation into TAMs [100,154].
Alterations of metabolic pathways in the tumor microenvironment also regulate macrophage polarization and function. During anaerobic glycolysis, tumor cells secrete lactate, thereby favoring macrophage differentiation to the M2 phenotype, which in turn promotes tumor growth by liberating VEGF-A and arginase-1 [120]. Interestingly, M2 polarization of melanoma-associated TAMs seems to be promoted by a mechanism involving a G-protein-coupled receptor that senses TME acidification induced by enhanced cancer cell glycolysis [119]. Moreover, α-ketoglutarate (αKG) produced via glutaminolysis in bone marrow-derived macrophages (BMDMs) is an anti-inflammatory metabolite that promotes M2 activation and controls metabolic reprogramming of M2 macrophages through jumonji domain-containing protein 3 (Jmjd3)-dependent mechanisms. Furthermore, αKG limits M1 polarization by reducing NF-κB pathway activity via αKG–prolyl hydroxylase (PHD)-dependent control of IKKβ activity [155].
In pancreatic ductal adenocarcinoma (PDAC), G12D is the most frequent KRAS mutation (referred to as KRASG12D) and the presence of this oncogene correlates with distinctive features of the TME [156]. Notably, autophagy-dependent ferroptosis favors the EV-mediated release of KRASG12D protein from PDAC cells during oxidative stress. Upon EV uptake by macrophages via the receptor for advanced glycation end products (RAGE), KRASG12D induces M2-like TAM polarization via STAT3-dependent fatty acid oxidation. Importantly, the abundance of KRASG12D in TAMs is associated with poor prognosis of PDAC patients [115].
Chemokines, cytokines and products of the complement cascade are significant determinants of macrophage recruitment and accumulation in tumors [157,158]. For instance, elevated levels of tumor-derived chemotactic protein CCL2 have been correlated with high numbers of TAMs and poor patient prognosis in various cancer types [118,123,124]. Moreover, NADPH oxidase 4 (NOX4) in tumors reportedly mediates macrophage chemotaxis and M2 polarization in lung cancer by stimulating the expression of various cytokines, including CCL7, IL-8, CSF-1 and VEGF-C via ROS/PI3K/AKT signaling. Likewise, NOX4-educated M2 macrophages exhibit elevated JNK activity and release heparin-binding EGF (HB-EGF) related growth factor, thus contributing to non-small cell lung cancer growth [122]. Another study revealed that the IL-6 receptor gp130 is present in breast cancer cell-derived EVs and stimulates STAT3 signaling in BMDMs. The survival of BMDMs increases in response to EV exposure, also upregulating the expression of protumoral cytokines, thereby converting them into potentially cancer-promoting macrophages [109].
As previously anticipated, several studies suggest that EVs might participate in the communication between cancer cells and macrophages. For instance, several miRNAs (miR-25-3p, miR-130b-3p, miR-425-5p) that are upregulated in colorectal cancer cells by activating the CXCL12/CXCR4 axis, are transferred to macrophages via EVs and induce their polarization. Specifically, exosomal miRNAs induce M2 polarization by activating the PI3K/AKT signaling pathway and inhibiting PTEN. In turn, M2-polarized macrophages promote liver metastasis of colorectal cancer by enhancing EMT and secreting VEGF [117]. Similarly, epithelial ovarian cancer-derived EVs contain high levels of miR-222-3p that can polarize macrophages to M2-like TAMs and thereby favor tumor progression by inducing SOCS3/STAT3 signaling [111]. Additionally, miR-145 secreted from colorectal cancer cells via EVs is taken up by macrophages where it promotes polarization to the M2-like phenotype through the downregulation of histone deacetylase 11 (HDAC11). Therefore, TAMs polarized by miR-145 contained in EVs contribute to the modulation of the tumor microenvironment and tumor progression [116].

5. Antioxidants as Potential Therapeutic Tools Targeting Adipocyte–Macrophage Crosstalk in Cancer

5.1. Effects on Obesity and Cancer

ROS participate in many physiological and pathological processes. For example, oxidative stress modulates the differentiation of adipocytes in physiological conditions and obesity. With this in mind, one might anticipate that antioxidants could be employed for the development of therapeutic strategies to preclude obesity-related oxidative stress (reviewed in [159]).
The main source of ROS are the mitochondria due to the leakage of electrons from the respiratory chain. In addition, ROS are produced by the endoplasmic reticulum, lysosomes, peroxisomes and cytosolic enzymes [159]. ROS regulate signaling pathways, gene expression and cell death in normal and cancer cells. In normal cells, a balance exists between ROS formation and antioxidant activity that maintains low ROS levels, thereby limiting cell proliferation and survival. Cancer cells increase ROS generation and also antioxidant capacity, which allows them to adapt to elevated ROS levels and pro-tumorigenic signaling without suffering cytotoxic effects. Through the activation of MAPK/ERK and PI3K/Akt signaling pathways, ROS enhance cell proliferation and cell survival. In addition, oxidative stress enhances HIF-1α expression and allows cancer cell adaptation. Furthermore, ROS induce drug resistance by activating PI3K/Akt and HIF-1α. Exacerbated increments in ROS levels to toxic concentrations will also promote cancer cell death (reviewed in [160]). For this reason, increases in ROS and their reduction using antioxidants can be associated with both beneficial and detrimental effects in cancer cells (reviewed in [161]). For instance, some chemotherapies increase ROS to levels that promote oxidative stress-induced tumor cell death while others suppress ROS levels to reduce cancer cell proliferation (reviewed in [160]).
In previous sections, we have summarized information linking obesity-induced inflammation to cancer development and progression. Indeed, obesity promotes cancer through mechanisms involving ROS and is associated with the presence of adipokines, VEGF, TNFα and IL-6 (reviewed in [162]). For that reason, the coming sections will discuss how antioxidants may protect against adverse obesity-induced effects both at the systemic (Section 5.2) and local levels (Section 5.3).

5.2. Systemic Effects

As we previously described, an intricate connection exists between obesity, inflammation and cancer, whereby several systemic mediators and pathways are involved, including but not limited to ROS, cytokines, adipokines, Akt, STAT3 and several metabolites. Table 4 summarizes antioxidants known to prevent cancer associated with obesity and inflammation by controlling these systemic mechanisms.

Plant-Derived Antioxidants

Curcumin, a polyphenol with antioxidant and anti-inflammatory properties found primarily in turmeric, has been ascribed interesting beneficial systemic effects. For instance, curcumin combined with the anti-inflammatory drug salsalate reportedly prevents the development of colon cancer in mice fed a HFD and exposed to the colon-specific carcinogen azoxymethane. The combination of curcumin/salsalate elicited such effects by reducing proinflammatory cytokine levels and diminishing activation of the PI3K/Akt/mTOR/NF-κB/Wnt pathway [163,164]. Importantly, curcumin alone has antitumor effects too. For example, curcumin prevents obesity-related colorectal cancer by attenuating chronic inflammation and improving adipokine imbalance, reducing cyclooxygenase-2 (COX-2), AMPK, NF-κB, TNFα, IL-6 and leptin levels, while increasing adiponectin levels [165]. In addition, curcumin prevents accelerated polyp development associated with a HFD by enhancing apoptosis, reducing oxidation-induced DNA damage, and increasing the efficiency of DNA repair [166].
Similar to curcumin, epigallocatechin gallate (EGCG) is an antioxidant that seems to be effective in reducing obesity and tumor growth. Specifically, EGCG has been shown to suppress azoxymethane-induced premalignant colon lesions in obese mice and these beneficial effects of EGCG were associated with reductions in IGF-IR, p-GSK-3β, β-catenin, COX2 and cyclin D1 protein levels as well as IGF-I, insulin, triglyceride, cholesterol and leptin serum levels [167]. Silibinin, a polyphenolic flavonoid found in silymarin extracted from milk thistle, is another antioxidant that reduces the protumoral properties associated with obesity. In fact, silibinin prevents lipogenesis and obesity-induced tumor progression. Moreover, in hepatocellular carcinoma HepG2 cells, silibinin reduces invasion by decreasing ROS, MMP-9, FASN, IL-6, IL-1β and Erk phosphorylation [168].
Some flavonoids have both antioxidant and antitumoral effects. For instance, obese ovariectomized mice on a HFD supplemented with naringenin have less adipose mass, reduced adipocyte size, as well as inflammatory cytokine levels, in both mammary and perigonadal adipose tissues than their counterparts without supplementation. Moreover, naringenin delays the growth of tumors at the early stages of breast cancer development [169]. Taxifolin is another flavonoid that can prevent obesity-induced breast cancer [170]. In fact, a study in African American women with obesity showed that adipose tissue secretes hepatocyte growth factor (HGF) with an endocrine effect on breast cancer cells. Specifically, HGF activates c-Met, thereby increasing SOS1-Grb2-CCL2 signaling. CCL2, in turn, induces macrophage polarization to favor the development of an anti-inflammatory microenvironment. Likewise, taxifolin inhibits SOS1 signaling by blocking the interaction between SOS1 and Grb2. Thus, taxifolin can prevent the systemic effects of obesity in cancer cells by reducing signaling due to adipose-secreted HGF. In addition, taxifolin prevents the local effects of obesity by inhibiting SOS1 signaling, thereby reducing macrophage polarization in the tumor microenvironment (see Table 5, next section [170]).
Resveratrol is another antioxidant that reduces the endocrine effects of obese adipose tissue on breast cancer cells. More specifically, subcutaneous adipose tissue from obese rats promotes cell cycle entry in MCF7 breast cancer cells. Resveratrol supplementation can prevent this by increasing the adiponectin/leptin ratio as well as promoting AMPK and inhibiting Akt signaling [172]. In addition, resveratrol inhibits obesity-associated adipose tissue dysfunction and tumor growth. In a mouse model of diet-induced obesity, abnormal adipose expansion was associated with a deregulated COX-2/PPARγ ratio, increased levels of proinflammatory markers, macrophage infiltration and tumor growth. Supplementation with resveratrol attenuates the effects of diet-induced obesity by inhibiting adipocyte hypertrophy and dysregulation and also prevents breast cancer development [171]. Of note, resveratrol not only has systemic but also local effects and modulates adipocyte function in the tumor microenvironment, as summarized in Table 5.

5.3. Local Effects

In the previous section, we summarized some examples of the impact obesity has on the tumor microenvironment, underscoring the relevance of systemic and local events in the development of cancer. Within the tumor microenvironment, cancer cell interactions with adipocytes and macrophages promote cancer progression through a variety of mechanisms involving oxidative stress, IL-6, IL-1β, STAT3, ERK and CLS. In the following paragraphs, we summarize some of the available literature evidencing the efficacy of antioxidant treatments in reverting/preventing cancer by remodeling the tumor microenvironment and regulating the mechanisms summarized in Table 5.
As may be anticipated, some antioxidants, such as taxifolin and resveratrol, have systemic and local effects that contribute to preventing obesity-induced inflammation and cancer [170,171]. For instance, taxifolin inhibits the stimulation of cancer cells by HGF secreted by adipocytes and does so by preventing activation of the SOS1-Grb2-CCL2 pathway. In this way, taxifolin reduces CCL2 in cancer cells and modulates macrophage polarization, thereby preventing cancer progression [170]. Another example is resveratrol, which reduces the median area of intratumoral adipocytes and the number of TAMs that increase as a consequence of diet-induced obesity. Thus, resveratrol modifies the tumor microenvironment by preventing the formation of CLS that appear as a consequence of adipocyte dysfunction and macrophage infiltration. In doing so, resveratrol reduces obesity-related cancer development [171].
Recently, Stassi and collaborators reviewed novel strategies targeting adipose tissue-released factors such as lipids, glucose, adipocytokines and miRNA [185]. For instance, targeting lipid biosynthesis through FASN inhibition results in effective antitumoral effects [185]. Indeed, silibinin reduces FASN protein expression, thereby decreasing the impact of obesity in liver cancer [168]. In addition, PPARγ regulates lipid and glucose homeostasis, as well as the expression of tumor suppressor genes, such as BRCA1 and PTEN. In this regard, PPARγ antagonists impair cell proliferation and mammary tumor growth [185]. In fact, in silico analysis demonstrates that quercetin and rutin target PPARγ and thereby have potential anti-obesity effects [186]. Targeting MTC1 (a lactate transporter) or lactate dehydrogenase, and thereby controlling glucose metabolism, reduces the occurrence and progression of non-small cell lung cancer [185]. Moreover, resveratrol increases insulin signaling and glucose uptake by adipocytes [187], indicative of its potential therapeutic benefits in treating adipocyte-altered glucose metabolism. Finally, antioxidants play a relevant role in the treatment of obesity. For instance, rosiglitazone combined with manganese tetroxide antioxidant nanoparticles synergistically reduces obesity via locally promoting beige adipogenesis, reducing fat accumulation and increasing antioxidative defense in adipose tissue. The mechanisms implicated involve the inhibition of PPARγ and ROS scavenging [188]. Similarly, the antioxidant arctigenin reduced the weight gain induced by HFDs in obese mice through the inhibition of adipogenesis-related factors, including PPARγ, C/EBPα, fatty acid synthase, adipocyte fatty acid-binding protein and lipoprotein lipase [189].
In colon cancer, nanoparticles containing vitamin E succinate inhibit cancer cell proliferation and dissemination as well as improve the intraperitoneal microenvironment by decreasing the levels of VEGF, IL-10 and the presence of M2-like TAMs [173]. Furthermore, the polyphenolic isoflavone found in soybeans, genistein, regulates the tumor microenvironment and exhibits an anticancer effect in oxidative stress-induced experimental colon carcinogenesis [174]. In addition, berberine is another antioxidant that inhibits the development of colitis-associated colorectal cancer by interfering with TNFα and IL-6-induced inflammatory responses in colonic macrophages, thereby inhibiting EGFR/ERK signaling in tumor cells [175]. Moreover, berberine downregulates TNFα and other inflammatory and procarcinogenic markers (IL-1β, Ccl1, Cxcl9, NF-κB, COX-2, JNK/STAT3, β-catenin, c-Myc and CylinD1), as well as modulates gut microbiota. In fact, berberine increases the abundance of beneficial bacteria, such as Roseburia, Eubacterium, Ruminococcaceae and Firmicutes, while reducing detrimental Odoribacter, Muribaculum, Mucispirillum and Parasutterella. These changes aid in preventing tumor development by decreasing the release of proinflammatory and carcinogenic factors [176,177]. Berberine, likewise, reverts breast cancer by attenuating the inflammatory response. Specifically, berberine reduces the expression of TNFα and IL-6 as well as inhibits c-Jun, c-Fos and NF-κB signaling in breast cancer cells [178].
EGCG is another antioxidant that is suggested to prevent breast cancer. First, EGCG was reported to inhibit adipocyte/cancer cell STAT3-mediated oncogenic paracrine control, thereby preventing adipogenesis and the obesogenic environment that favors breast cancer development [179]. Furthermore, EGCG was found to modulate not only adipogenesis but also inflammation. Breast cancer cells induce the expression of cytokines (CCL2, CCL5, IL-1β, and IL-6) and immunomodulators (COX2, HIF-1α, VEGFα, and PD-L1) by cancer-associated adipocytes. EGCG prevents this inflammatory response due to the cancer cell–adipocyte connection and also modulates Snail, Smad2 and NF-κB [180]. Moreover, EGCG has been successfully employed in the release of nanochemical drug combinations. For instance, the EGCG/taxane combination promotes the internalization of the nanoparticles into prostate cancer cells through CD44 and the inhibition of cell growth [181].
Among other antioxidants, quercetin exerts beneficial effects in a xenograft model of liver cancer via the remodeling of the tumor microenvironment in a manner that improves tumor permeation by quercetin/doxorubicin-loaded nanoparticles [182]. Additionally, extracellular microparticles encapsulated with the organic compound diallyl trisulfide found in garlic prevent the prometastatic, inflammatory microenvironment in lung metastatic niches. These extracellular microparticles are able to fuse with cancer and normal lung cells and inhibit tumor cell migration, as well as reduce inflammation in the tumor microenvironment by decreasing the release of S100A8/A9 and IL-6 [183]. Similarly, the synthetic antioxidant nicaraven prevents the growth of inflamed lung tumors by reducing the recruitment of macrophages and neutrophils. Indeed, Nicaraven reduces IL-2 and MIP-2 levels in serum, as well as CXCL10 and SDF-1 levels in tumors [184].

6. Conclusions

Obesity is a complex multifactorial disease that is associated with the development of multiple comorbidities and, as such, it has become of great importance to better understand the molecular underpinnings involved in these processes. In the context of this review, the connection that exists between obesity and the development of several types of cancer is of particular interest (see Figure 1). Specifically, inflammatory changes that adipose tissue undergoes in obesity are linked to multiple effects on cancer cell behavior. These changes can be triggered by systemic “endocrine” signaling from adipose tissue to the tumor or by more intimate “paracrine” mechanisms in the tumor microenvironment. The prevalent macrophages and mechanisms are summarized showing how the presence of tumor-associated adipocytes “convert” these macrophages into a population of cells that very actively promote tumor growth. Importantly, the signaling between these two cell populations is bidirectional and serves to generate an amplification loop that exacerbates inflammation and oxidative stress in the tumor environment. This in turn favors cancer cell progression to an ever more aggressive phenotype and ultimately promotes metastasis, the leading cause of death in cancer patients. Fortunately, antioxidants of vastly different chemical natures have shown some promise in blocking several of these mechanisms, and one may anticipate that gaining greater insight into how precisely such agents function in the aforementioned context could yield key “reagents” to detain tumor development.

Author Contributions

Conceptualization, S.S., L.S., M.C. and A.F.G.Q.; investigation and writing—original draft preparation, S.S., L.S. and M.C.; writing—review and editing, S.S., L.S., M.C. and A.F.G.Q.; resources, project administration, and funding acquisition, M.C. and A.F.G.Q. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by CONICYT-FONDAP [15130011] (A.F.G.Q., M.C.), FONDECYT [1210644] (AFGQ), FONDECYT [1211477] (M.C.), FONDECYT post-doctoral fellowship [3190330] (L.S.), ANID Ph.D. fellowship [21211248] (S.S.), Concurso de Investigación con Colaboración Internacional de la Universidad Finis Terrae (L.S.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All the data are contained within the article.

Acknowledgments

Alejandra Sandoval is gratefully acknowledged for providing access with her license to BIORENDERS which was used to generate Figure 1.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hendren, N.S.; de Lemos, J.A.; Ayers, C.; Das, S.R.; Rao, A.; Carter, S.; Rosenblatt, A.; Walchok, J.; Omar, W.; Khera, R.; et al. Association of Body Mass Index and Age with Morbidity and Mortality in Patients Hospitalized with COVID-19: Results from the American Heart Association COVID-19 Cardiovascular Disease Registry. Circulation 2021, 143, 135–144. [Google Scholar] [CrossRef] [PubMed]
  2. Hotamisligil, G.S.; Shargill, N.S.; Spiegelman, B.M. Adipose expression of tumor necrosis factor-alpha: Direct role in obesity-linked insulin resistance. Science 1993, 259, 87–91. [Google Scholar] [CrossRef] [PubMed]
  3. Zhang, Y.; Proenca, R.; Maffei, M.; Barone, M.; Leopold, L.; Friedman, J.M. Positional cloning of the mouse obese gene and its human homologue. Nature 1994, 372, 425–432. [Google Scholar] [CrossRef] [PubMed]
  4. Funcke, J.B.; Scherer, P.E. Beyond adiponectin and leptin: Adipose tissue-derived mediators of inter-organ communication. J. Lipid Res. 2019, 60, 1648–1684. [Google Scholar] [CrossRef] [PubMed]
  5. Cox, A.R.; Chernis, N.; Masschelin, P.M.; Hartig, S.M. Immune Cells Gate White Adipose Tissue Expansion. Endocrinology 2019, 160, 1645–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Cai, Z.; Huang, Y.; He, B. New Insights into Adipose Tissue Macrophages in Obesity and Insulin Resistance. Cells 2022, 11, 1424. [Google Scholar] [CrossRef] [PubMed]
  7. Le, K.A.; Mahurkar, S.; Alderete, T.L.; Hasson, R.E.; Adam, T.C.; Kim, J.S.; Beale, E.; Xie, C.; Greenberg, A.S.; Allayee, H.; et al. Subcutaneous adipose tissue macrophage infiltration is associated with hepatic and visceral fat deposition, hyperinsulinemia, and stimulation of NF-kappaB stress pathway. Diabetes 2011, 60, 2802–2809. [Google Scholar] [CrossRef] [Green Version]
  8. Lauby-Secretan, B.; Scoccianti, C.; Loomis, D.; Grosse, Y.; Bianchini, F.; Straif, K.; International Agency for Research on Cancer Handbook Working Group. Body Fatness and Cancer—Viewpoint of the IARC Working Group. N. Engl. J. Med. 2016, 375, 794–798. [Google Scholar] [CrossRef] [Green Version]
  9. Frisch, C.M.; Zimmermann, K.; Zillessen, P.; Pfeifer, A.; Racke, K.; Mayer, P. Non-small cell lung cancer cell survival crucially depends on functional insulin receptors. Endocr. Relat. Cancer 2015, 22, 609–621. [Google Scholar] [CrossRef] [Green Version]
  10. Slabuszewska-Jozwiak, A.; Lukaszuk, A.; Janicka-Kosnik, M.; Wdowiak, A.; Jakiel, G. Role of Leptin and Adiponectin in Endometrial Cancer. Int. J. Mol. Sci. 2022, 23, 5307. [Google Scholar] [CrossRef]
  11. Miethe, C.; Zamora, M.; Torres, L.; Raign, K.G.; Groll, C.J.; Price, R.S. Characterizing the differential physiological effects of adipocytokines visfatin and resistin in liver cancer cells. Horm. Mol. Biol. Clin. Investig. 2019, 38, 20180068. [Google Scholar] [CrossRef] [PubMed]
  12. Cunarro, J.; Casado, S.; Lugilde, J.; Tovar, S. Hypothalamic Mitochondrial Dysfunction as a Target in Obesity and Metabolic Disease. Front. Endocrinol. 2018, 9, 283. [Google Scholar] [CrossRef] [PubMed]
  13. Assumpcao, J.A.F.; Pasquarelli-do-Nascimento, G.; Duarte, M.S.V.; Bonamino, M.H.; Magalhaes, K.G. The ambiguous role of obesity in oncology by promoting cancer but boosting antitumor immunotherapy. J. Biomed. Sci. 2022, 29, 12. [Google Scholar] [CrossRef] [PubMed]
  14. Hauck, A.K.; Huang, Y.; Hertzel, A.V.; Bernlohr, D.A. Adipose oxidative stress and protein carbonylation. J. Biol. Chem. 2019, 294, 1083–1088. [Google Scholar] [CrossRef] [Green Version]
  15. Jackisch, L.; Murphy, A.M.; Kumar, S.; Randeva, H.; Tripathi, G.; McTernan, P.G. Tunicamycin-Induced Endoplasmic Reticulum Stress Mediates Mitochondrial Dysfunction in Human Adipocytes. J. Clin. Endocrinol. Metab. 2020, 105, 2905–2918. [Google Scholar] [CrossRef]
  16. Nishikawa, M.; Hashida, M.; Takakura, Y. Catalase delivery for inhibiting ROS-mediated tissue injury and tumor metastasis. Adv. Drug. Deliv. Rev. 2009, 61, 319–326. [Google Scholar] [CrossRef]
  17. Mondal, S.; Adhikari, N.; Banerjee, S.; Amin, S.A.; Jha, T. Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: A minireview. Eur. J. Med. Chem. 2020, 194, 112260. [Google Scholar] [CrossRef]
  18. Usman, M.; Volpi, E.V. DNA damage in obesity: Initiator, promoter and predictor of cancer. Mutat. Res. Rev. Mutat. Res. 2018, 778, 23–37. [Google Scholar] [CrossRef]
  19. Quail, D.F.; Dannenberg, A.J. The obese adipose tissue microenvironment in cancer development and progression. Nat. Rev. Endocrinol. 2019, 15, 139–154. [Google Scholar] [CrossRef]
  20. Lu, Y.; Zhou, X.Y.; Zhou, C.L.; Liu, J.; Yong, T.; Fan, Y.; Wang, C. Insulin receptor tyrosine kinase substrate (IRTKS) promotes the tumorigenesis of pancreatic cancer via PI3K/AKT signaling. Hum. Cell 2022, 35, 1885–1899. [Google Scholar] [CrossRef]
  21. Hu, D.X.; Sun, Q.F.; Xu, L.; Lu, H.D.; Zhang, F.; Li, Z.M.; Zhang, M.Y. Knockdown of DEAD-box 51 inhibits tumor growth of esophageal squamous cell carcinoma via the PI3K/AKT pathway. World J. Gastroenterol. 2022, 28, 464–478. [Google Scholar] [CrossRef] [PubMed]
  22. Gu, J.W.; Young, E.; Patterson, S.G.; Makey, K.L.; Wells, J.; Huang, M.; Tucker, K.B.; Miele, L. Postmenopausal obesity promotes tumor angiogenesis and breast cancer progression in mice. Cancer Biol. 2011, 11, 910–917. [Google Scholar] [CrossRef]
  23. Seo, B.R.; Bhardwaj, P.; Choi, S.; Gonzalez, J.; Andresen Eguiluz, R.C.; Wang, K.; Mohanan, S.; Morris, P.G.; Du, B.; Zhou, X.K.; et al. Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis. Sci. Transl. Med. 2015, 7, 301ra130. [Google Scholar] [CrossRef] [Green Version]
  24. Gonzalez-Perez, R.R.; Lanier, V.; Newman, G. Leptin’s Pro-Angiogenic Signature in Breast Cancer. Cancers 2013, 5, 1140–1162. [Google Scholar] [CrossRef] [Green Version]
  25. Miethe, C.; Torres, L.; Zamora, M.; Price, R.S. Inhibition of PI3K/Akt and ERK signaling decreases visfatin-induced invasion in liver cancer cells. Horm. Mol. Biol. Clin. Investig. 2021, 42, 357–366. [Google Scholar] [CrossRef] [PubMed]
  26. Ramos-Andrade, I.; Moraes, J.; Brandao-Costa, R.M.; Vargas da Silva, S.; de Souza, A.; da Silva, C.; Renovato-Martins, M.; Barja-Fidalgo, C. Obese adipose tissue extracellular vesicles raise breast cancer cell malignancy. Endocr. Relat. Cancer 2020, 27, 571–582. [Google Scholar] [CrossRef]
  27. Gao, M.; Ma, Y.; Liu, D. Rutin suppresses palmitic acids-triggered inflammation in macrophages and blocks high fat diet-induced obesity and fatty liver in mice. Pharm. Res. 2013, 30, 2940–2950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Song, H.; Lim, D.Y.; Jung, J.I.; Cho, H.J.; Park, S.Y.; Kwon, G.T.; Kang, Y.H.; Lee, K.W.; Choi, M.S.; Park, J.H.Y. Dietary oleuropein inhibits tumor angiogenesis and lymphangiogenesis in the B16F10 melanoma allograft model: A mechanism for the suppression of high-fat diet-induced solid tumor growth and lymph node metastasis. Oncotarget 2017, 8, 32027–32042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Cao, L.; Chen, X.; Xiao, X.; Ma, Q.; Li, W. Resveratrol inhibits hyperglycemia-driven ROS-induced invasion and migration of pancreatic cancer cells via suppression of the ERK and p38 MAPK signaling pathways. Int. J. Oncol. 2016, 49, 735–743. [Google Scholar] [CrossRef] [Green Version]
  30. Freemerman, A.J.; Johnson, A.R.; Sacks, G.N.; Milner, J.J.; Kirk, E.L.; Troester, M.A.; Macintyre, A.N.; Goraksha-Hicks, P.; Rathmell, J.C.; Makowski, L. Metabolic reprogramming of macrophages: Glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. J. Biol. Chem. 2014, 289, 7884–7896. [Google Scholar] [CrossRef]
  31. Dibaba, D.T.; Judd, S.E.; Gilchrist, S.C.; Cushman, M.; Pisu, M.; Safford, M.; Akinyemiju, T. Association between obesity and biomarkers of inflammation and metabolism with cancer mortality in a prospective cohort study. Metabolism 2019, 94, 69–76. [Google Scholar] [CrossRef] [PubMed]
  32. Wunderlich, C.M.; Ackermann, P.J.; Ostermann, A.L.; Adams-Quack, P.; Vogt, M.C.; Tran, M.L.; Nikolajev, A.; Waisman, A.; Garbers, C.; Theurich, S.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Benaiges, E.; Ceperuelo-Mallafre, V.; Madeira, A.; Bosch, R.; Nunez-Roa, C.; Ejarque, M.; Maymo-Masip, E.; Huber-Ruano, I.; Lejeune, M.; Vendrell, J.; et al. Survivin drives tumor-associated macrophage reprogramming: A novel mechanism with potential impact for obesity. Cell Oncol. 2021, 44, 777–792. [Google Scholar] [CrossRef]
  34. Becattini, B.; Breasson, L.; Sardi, C.; Zani, F.; Solinas, G. PI3Kgamma promotes obesity-associated hepatocellular carcinoma by regulating metabolism and inflammation. JHEP Rep. 2021, 3, 100359. [Google Scholar] [CrossRef] [PubMed]
  35. Dogan, H.; Shu, J.; Hakguder, Z.; Xu, Z.; Cui, J. Elucidation of molecular links between obesity and cancer through microRNA regulation. BMC Med. Genom. 2020, 13, 161. [Google Scholar] [CrossRef] [PubMed]
  36. Madeddu, C.; Sanna, E.; Gramignano, G.; Tanca, L.; Cherchi, M.C.; Mola, B.; Petrillo, M.; Maccio, A. Correlation of Leptin, Proinflammatory Cytokines and Oxidative Stress with Tumor Size and Disease Stage of Endometrioid (Type I) Endometrial Cancer and Review of the Underlying Mechanisms. Cancers 2022, 14, 268. [Google Scholar] [CrossRef] [PubMed]
  37. Strong, A.L.; Ohlstein, J.F.; Biagas, B.A.; Rhodes, L.V.; Pei, D.T.; Tucker, H.A.; Llamas, C.; Bowles, A.C.; Dutreil, M.F.; Zhang, S.; et al. Leptin produced by obese adipose stromal/stem cells enhances proliferation and metastasis of estrogen receptor positive breast cancers. Breast Cancer Res. 2015, 17, 112. [Google Scholar] [CrossRef] [Green Version]
  38. Nunez-Ruiz, A.; Sanchez-Brena, F.; Lopez-Pacheco, C.; Acevedo-Dominguez, N.A.; Soldevila, G. Obesity modulates the immune macroenvironment associated with breast cancer development. PLoS ONE 2022, 17, e0266827. [Google Scholar] [CrossRef]
  39. Kolb, R.; Phan, L.; Borcherding, N.; Liu, Y.; Yuan, F.; Janowski, A.M.; Xie, Q.; Markan, K.R.; Li, W.; Potthoff, M.J.; et al. Obesity-associated NLRC4 inflammasome activation drives breast cancer progression. Nat. Commun. 2016, 7, 13007. [Google Scholar] [CrossRef] [Green Version]
  40. Kolb, R.; Kluz, P.; Tan, Z.W.; Borcherding, N.; Bormann, N.; Vishwakarma, A.; Balcziak, L.; Zhu, P.; Davies, B.S.; Gourronc, F.; et al. Obesity-associated inflammation promotes angiogenesis and breast cancer via angiopoietin-like 4. Oncogene 2019, 38, 2351–2363. [Google Scholar] [CrossRef]
  41. Blucher, C.; Iberl, S.; Schwagarus, N.; Muller, S.; Liebisch, G.; Horing, M.; Hidrobo, M.S.; Ecker, J.; Spindler, N.; Dietrich, A.; et al. Secreted Factors from Adipose Tissue Reprogram Tumor Lipid Metabolism and Induce Motility by Modulating PPARalpha/ANGPTL4 and FAK. Mol. Cancer Res. 2020, 18, 1849–1862. [Google Scholar] [CrossRef] [PubMed]
  42. Zhao, Y.X.; Sun, Y.L.; Ye, J.H.; Zhang, Y.; Shi, X.B.; Wang, J.M.; Wu, H.Y.; Zhang, W.J.; Yao, Y.Z. The Relationship Between white Adipose Tissue Inflammation and Overweight/Obesity in Chinese Female Breast Cancer: A Retrospective Study. Adv. Ther. 2020, 37, 2734–2747. [Google Scholar] [CrossRef] [PubMed]
  43. Iyengar, N.M.; Zhou, X.K.; Gucalp, A.; Morris, P.G.; Howe, L.R.; Giri, D.D.; Morrow, M.; Wang, H.; Pollak, M.; Jones, L.W.; et al. Systemic Correlates of White Adipose Tissue Inflammation in Early-Stage Breast Cancer. Clin. Cancer Res. 2016, 22, 2283–2289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Reggiani, F.; Labanca, V.; Mancuso, P.; Rabascio, C.; Talarico, G.; Orecchioni, S.; Manconi, A.; Bertolini, F. Adipose Progenitor Cell Secretion of GM-CSF and MMP9 Promotes a Stromal and Immunological Microenvironment That Supports Breast Cancer Progression. Cancer Res. 2017, 77, 5169–5182. [Google Scholar] [CrossRef] [Green Version]
  45. Quail, D.F.; Olson, O.C.; Bhardwaj, P.; Walsh, L.A.; Akkari, L.; Quick, M.L.; Chen, I.C.; Wendel, N.; Ben-Chetrit, N.; Walker, J.; et al. Obesity alters the lung myeloid cell landscape to enhance breast cancer metastasis through IL5 and GM-CSF. Nat. Cell Biol. 2017, 19, 974–987. [Google Scholar] [CrossRef] [PubMed]
  46. Nagahashi, M.; Yamada, A.; Katsuta, E.; Aoyagi, T.; Huang, W.C.; Terracina, K.P.; Hait, N.C.; Allegood, J.C.; Tsuchida, J.; Yuza, K.; et al. Targeting the SphK1/S1P/S1PR1 Axis That Links Obesity, Chronic Inflammation, and Breast Cancer Metastasis. Cancer Res. 2018, 78, 1713–1725. [Google Scholar] [CrossRef] [Green Version]
  47. Zablocka-Slowinska, K.; Placzkowska, S.; Skorska, K.; Prescha, A.; Pawelczyk, K.; Porebska, I.; Kosacka, M.; Grajeta, H. Oxidative stress in lung cancer patients is associated with altered serum markers of lipid metabolism. PLoS ONE 2019, 14, e0215246. [Google Scholar] [CrossRef]
  48. Hayashi, T.; Fujita, K.; Nojima, S.; Hayashi, Y.; Nakano, K.; Ishizuya, Y.; Wang, C.; Yamamoto, Y.; Kinouchi, T.; Matsuzaki, K.; et al. High-Fat Diet-Induced Inflammation Accelerates Prostate Cancer Growth via IL6 Signaling. Clin. Cancer Res. 2018, 24, 4309–4318. [Google Scholar] [CrossRef] [Green Version]
  49. Cho, H.J.; Kwon, G.T.; Park, H.; Song, H.; Lee, K.W.; Kim, J.I.; Park, J.H. A high-fat diet containing lard accelerates prostate cancer progression and reduces survival rate in mice: Possible contribution of adipose tissue-derived cytokines. Nutrients 2015, 7, 2539–2561. [Google Scholar] [CrossRef] [Green Version]
  50. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  51. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
  52. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  53. Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Huang, C.K.; Chang, P.H.; Kuo, W.H.; Chen, C.L.; Jeng, Y.M.; Chang, K.J.; Shew, J.Y.; Hu, C.M.; Lee, W.H. Adipocytes promote malignant growth of breast tumours with monocarboxylate transporter 2 expression via beta-hydroxybutyrate. Nat. Commun. 2017, 8, 14706. [Google Scholar] [CrossRef] [Green Version]
  55. Zhou, C.; He, X.; Tong, C.; Li, H.; Xie, C.; Wu, Y.; Wang, L.; Yan, X.; Luo, D.; Tang, Y.; et al. Cancer-associated adipocytes promote the invasion and metastasis in breast cancer through LIF/CXCLs positive feedback loop. Int. J. Biol. Sci. 2022, 18, 1363–1380. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, L.; Wu, Y.; Zhang, C.; Zhou, C.; Li, Y.; Zeng, Y.; Zhang, C.; Li, R.; Luo, D.; Wang, L.; et al. Cancer-associated adipocyte-derived G-CSF promotes breast cancer malignancy via Stat3 signaling. J. Mol. Cell. Biol. 2020, 12, 723–737. [Google Scholar] [CrossRef] [Green Version]
  57. Lee, Y.; Jung, W.H.; Koo, J.S. Adipocytes can induce epithelial-mesenchymal transition in breast cancer cells. Breast Cancer Res. Treat. 2015, 153, 323–335. [Google Scholar] [CrossRef]
  58. Wang, C.; Gao, C.; Meng, K.; Qiao, H.; Wang, Y. Human adipocytes stimulate invasion of breast cancer MCF-7 cells by secreting IGFBP-2. PLoS ONE 2015, 10, e0119348. [Google Scholar] [CrossRef] [Green Version]
  59. Vazquez Rodriguez, G.; Abrahamsson, A.; Jensen, L.D.E.; Dabrosin, C. Adipocytes Promote Early Steps of Breast Cancer Cell Dissemination via Interleukin-8. Front. Immunol. 2018, 9, 1767. [Google Scholar] [CrossRef] [Green Version]
  60. Al-Khalaf, H.H.; Al-Harbi, B.; Al-Sayed, A.; Arafah, M.; Tulbah, A.; Jarman, A.; Al-Mohanna, F.; Aboussekhra, A. Interleukin-8 Activates Breast Cancer-Associated Adipocytes and Promotes Their Angiogenesis- and Tumorigenesis-Promoting Effects. Mol. Cell Biol. 2019, 39, e00332-18. [Google Scholar] [CrossRef]
  61. Picon-Ruiz, M.; Pan, C.; Drews-Elger, K.; Jang, K.; Besser, A.H.; Zhao, D.; Morata-Tarifa, C.; Kim, M.; Ince, T.A.; Azzam, D.J.; et al. Interactions between Adipocytes and Breast Cancer Cells Stimulate Cytokine Production and Drive Src/Sox2/miR-302b-Mediated Malignant Progression. Cancer Res. 2016, 76, 491–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Bochet, L.; Meulle, A.; Imbert, S.; Salles, B.; Valet, P.; Muller, C. Cancer-associated adipocytes promotes breast tumor radioresistance. Biochem. Biophys. Res. Commun. 2011, 411, 102–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lehuede, C.; Li, X.; Dauvillier, S.; Vaysse, C.; Franchet, C.; Clement, E.; Esteve, D.; Longue, M.; Chaltiel, L.; Le Gonidec, S.; et al. Adipocytes promote breast cancer resistance to chemotherapy, a process amplified by obesity: Role of the major vault protein (MVP). Breast Cancer Res. 2019, 21, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Dirat, B.; Bochet, L.; Dabek, M.; Daviaud, D.; Dauvillier, S.; Majed, B.; Wang, Y.Y.; Meulle, A.; Salles, B.; Le Gonidec, S.; et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011, 71, 2455–2465. [Google Scholar] [CrossRef] [Green Version]
  65. Balaban, S.; Shearer, R.F.; Lee, L.S.; van Geldermalsen, M.; Schreuder, M.; Shtein, H.C.; Cairns, R.; Thomas, K.C.; Fazakerley, D.J.; Grewal, T.; et al. Adipocyte lipolysis links obesity to breast cancer growth: Adipocyte-derived fatty acids drive breast cancer cell proliferation and migration. Cancer Metab. 2017, 5, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Wang, Y.Y.; Attane, C.; Milhas, D.; Dirat, B.; Dauvillier, S.; Guerard, A.; Gilhodes, J.; Lazar, I.; Alet, N.; Laurent, V.; et al. Mammary adipocytes stimulate breast cancer invasion through metabolic remodeling of tumor cells. JCI Insight 2017, 2, e87489. [Google Scholar] [CrossRef] [Green Version]
  67. Mukherjee, A.; Chiang, C.Y.; Daifotis, H.A.; Nieman, K.M.; Fahrmann, J.F.; Lastra, R.R.; Romero, I.L.; Fiehn, O.; Lengyel, E. Adipocyte-Induced FABP4 Expression in Ovarian Cancer Cells Promotes Metastasis and Mediates Carboplatin Resistance. Cancer Res. 2020, 80, 1748–1761. [Google Scholar] [CrossRef] [Green Version]
  68. Au Yeung, C.L.; Co, N.N.; Tsuruga, T.; Yeung, T.L.; Kwan, S.Y.; Leung, C.S.; Li, Y.; Lu, E.S.; Kwan, K.; Wong, K.K.; et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 2016, 7, 11150. [Google Scholar] [CrossRef] [Green Version]
  69. Ladanyi, A.; Mukherjee, A.; Kenny, H.A.; Johnson, A.; Mitra, A.K.; Sundaresan, S.; Nieman, K.M.; Pascual, G.; Benitah, S.A.; Montag, A.; et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 2018, 37, 2285–2301. [Google Scholar] [CrossRef]
  70. Yang, J.; Zaman, M.M.; Vlasakov, I.; Roy, R.; Huang, L.; Martin, C.R.; Freedman, S.D.; Serhan, C.N.; Moses, M.A. Adipocytes promote ovarian cancer chemoresistance. Sci. Rep. 2019, 9, 13316. [Google Scholar] [CrossRef]
  71. Lazar, I.; Clement, E.; Dauvillier, S.; Milhas, D.; Ducoux-Petit, M.; LeGonidec, S.; Moro, C.; Soldan, V.; Dalle, S.; Balor, S.; et al. Adipocyte Exosomes Promote Melanoma Aggressiveness through Fatty Acid Oxidation: A Novel Mechanism Linking Obesity and Cancer. Cancer Res. 2016, 76, 4051–4057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Clement, E.; Lazar, I.; Attane, C.; Carrie, L.; Dauvillier, S.; Ducoux-Petit, M.; Esteve, D.; Menneteau, T.; Moutahir, M.; Le Gonidec, S.; et al. Adipocyte extracellular vesicles carry enzymes and fatty acids that stimulate mitochondrial metabolism and remodeling in tumor cells. EMBO J. 2020, 39, e102525. [Google Scholar] [CrossRef]
  73. Kushiro, K.; Chu, R.A.; Verma, A.; Nunez, N.P. Adipocytes Promote B16BL6 Melanoma Cell Invasion and the Epithelial-to-Mesenchymal Transition. Cancer Microenviron. 2012, 5, 73–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Chi, M.; Chen, J.; Ye, Y.; Tseng, H.Y.; Lai, F.; Tay, K.H.; Jin, L.; Guo, S.T.; Jiang, C.C.; Zhang, X.D. Adipocytes contribute to resistance of human melanoma cells to chemotherapy and targeted therapy. Curr. Med. Chem. 2014, 21, 1255–1267. [Google Scholar] [CrossRef] [PubMed]
  75. Bernard, J.J.; Lou, Y.R.; Peng, Q.Y.; Li, T.; Vakil, P.R.; Ding, N.; Laskin, J.D.; Dong, Z.; Conney, A.H.; Lu, Y.P. Parametrial fat tissue from high fat diet-treated SKH-1 mice stimulates transformation of mouse epidermal JB6 cells. J. Carcinog. Mutagen 2014, 5, 2157–2518. [Google Scholar] [CrossRef] [Green Version]
  76. Laurent, V.; Guerard, A.; Mazerolles, C.; Le Gonidec, S.; Toulet, A.; Nieto, L.; Zaidi, F.; Majed, B.; Garandeau, D.; Socrier, Y.; et al. Periprostatic adipocytes act as a driving force for prostate cancer progression in obesity. Nat. Commun. 2016, 7, 10230. [Google Scholar] [CrossRef] [Green Version]
  77. Xiang, F.; Wu, K.; Liu, Y.; Shi, L.; Wang, D.; Li, G.; Tao, K.; Wang, G. Omental adipocytes enhance the invasiveness of gastric cancer cells by oleic acid-induced activation of the PI3K-Akt signaling pathway. Int. J. Biochem. Cell. Biol. 2017, 84, 14–21. [Google Scholar] [CrossRef]
  78. Li, S.; Wu, T.; Lu, Y.X.; Wang, J.X.; Yu, F.H.; Yang, M.Z.; Huang, Y.J.; Li, Z.J.; Wang, S.L.; Huang, L.; et al. Obesity promotes gastric cancer metastasis via diacylglycerol acyltransferase 2-dependent lipid droplets accumulation and redox homeostasis. Redox Biol. 2020, 36, 101596. [Google Scholar] [CrossRef]
  79. Wen, Y.A.; Xing, X.; Harris, J.W.; Zaytseva, Y.Y.; Mitov, M.I.; Napier, D.L.; Weiss, H.L.; Mark Evers, B.; Gao, T. Adipocytes activate mitochondrial fatty acid oxidation and autophagy to promote tumor growth in colon cancer. Cell Death Dis. 2017, 8, e2593. [Google Scholar] [CrossRef] [Green Version]
  80. Xiong, X.; Wen, Y.A.; Fairchild, R.; Zaytseva, Y.Y.; Weiss, H.L.; Evers, B.M.; Gao, T. Upregulation of CPT1A is essential for the tumor-promoting effect of adipocytes in colon cancer. Cell Death Dis. 2020, 11, 736. [Google Scholar] [CrossRef]
  81. Takehara, M.; Sato, Y.; Kimura, T.; Noda, K.; Miyamoto, H.; Fujino, Y.; Miyoshi, J.; Nakamura, F.; Wada, H.; Bando, Y.; et al. Cancer-associated adipocytes promote pancreatic cancer progression through SAA1 expression. Cancer Sci. 2020, 111, 2883–2894. [Google Scholar] [CrossRef] [PubMed]
  82. Pascual, G.; Avgustinova, A.; Mejetta, S.; Martin, M.; Castellanos, A.; Attolini, C.S.; Berenguer, A.; Prats, N.; Toll, A.; Hueto, J.A.; et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 2017, 541, 41–45. [Google Scholar] [CrossRef]
  83. Yeo, E.J.; Cassetta, L.; Qian, B.Z.; Lewkowich, I.; Li, J.F.; Stefater, J.A., 3rd; Smith, A.N.; Wiechmann, L.S.; Wang, Y.; Pollard, J.W.; et al. Myeloid WNT7b mediates the angiogenic switch and metastasis in breast cancer. Cancer Res. 2014, 74, 2962–2973. [Google Scholar] [CrossRef] [Green Version]
  84. Lin, S.; Sun, L.; Lyu, X.; Ai, X.; Du, D.; Su, N.; Li, H.; Zhang, L.; Yu, J.; Yuan, S. Lactate-activated macrophages induced aerobic glycolysis and epithelial-mesenchymal transition in breast cancer by regulation of CCL5-CCR5 axis: A positive metabolic feedback loop. Oncotarget 2017, 8, 110426–110443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Gocheva, V.; Wang, H.W.; Gadea, B.B.; Shree, T.; Hunter, K.E.; Garfall, A.L.; Berman, T.; Joyce, J.A. IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 2010, 24, 241–255. [Google Scholar] [CrossRef] [Green Version]
  86. Atanasov, G.; Potner, C.; Aust, G.; Schierle, K.; Dietel, C.; Benzing, C.; Krenzien, F.; Bartels, M.; Eichfeld, U.; Schmelzle, M.; et al. TIE2-expressing monocytes and M2-polarized macrophages impact survival and correlate with angiogenesis in adenocarcinoma of the pancreas. Oncotarget 2018, 9, 29715–29726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Jeong, H.; Kim, S.; Hong, B.J.; Lee, C.J.; Kim, Y.E.; Bok, S.; Oh, J.M.; Gwak, S.H.; Yoo, M.Y.; Lee, M.S.; et al. Tumor-Associated Macrophages Enhance Tumor Hypoxia and Aerobic Glycolysis. Cancer Res. 2019, 79, 795–806. [Google Scholar] [CrossRef] [Green Version]
  88. Fujisaki, K.; Fujimoto, H.; Sangai, T.; Nagashima, T.; Sakakibara, M.; Shiina, N.; Kuroda, M.; Aoyagi, Y.; Miyazaki, M. Cancer-mediated adipose reversion promotes cancer cell migration via IL-6 and MCP-1. Breast Cancer Res. Treat. 2015, 150, 255–263. [Google Scholar] [CrossRef]
  89. Arendt, L.M.; McCready, J.; Keller, P.J.; Baker, D.D.; Naber, S.P.; Seewaldt, V.; Kuperwasser, C. Obesity promotes breast cancer by CCL2-mediated macrophage recruitment and angiogenesis. Cancer Res. 2013, 73, 6080–6093. [Google Scholar] [CrossRef] [Green Version]
  90. Fan, Y.; Wang, X.; Li, Y. IFI30 expression predicts patient prognosis in breast cancer and dictates breast cancer cells proliferation via regulating autophagy. Int. J. Med. Sci. 2021, 18, 3342–3352. [Google Scholar] [CrossRef]
  91. Yang, M.; Liu, F.; Higuchi, K.; Sawashita, J.; Fu, X.; Zhang, L.; Zhang, L.; Fu, L.; Tong, Z.; Higuchi, K. Serum amyloid A expression in the breast cancer tissue is associated with poor prognosis. Oncotarget 2016, 7, 35843–35852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef]
  93. Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr. Opin. Immunol. 2012, 24, 207–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lee, C.C.; Lin, J.C.; Hwang, W.L.; Kuo, Y.J.; Chen, H.K.; Tai, S.K.; Lin, C.C.; Yang, M.H. Macrophage-secreted interleukin-35 regulates cancer cell plasticity to facilitate metastatic colonization. Nat. Commun. 2018, 9, 3763. [Google Scholar] [CrossRef] [Green Version]
  95. Morrissey, S.M.; Zhang, F.; Ding, C.; Montoya-Durango, D.E.; Hu, X.; Yang, C.; Wang, Z.; Yuan, F.; Fox, M.; Zhang, H.G.; et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021, 33, 2040–2058 e2010. [Google Scholar] [CrossRef] [PubMed]
  96. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Vitale, I.; Manic, G.; Coussens, L.M.; Kroemer, G.; Galluzzi, L. Macrophages and Metabolism in the Tumor Microenvironment. Cell Metab. 2019, 30, 36–50. [Google Scholar] [CrossRef]
  98. Wagner, J.; Rapsomaniki, M.A.; Chevrier, S.; Anzeneder, T.; Langwieder, C.; Dykgers, A.; Rees, M.; Ramaswamy, A.; Muenst, S.; Soysal, S.D.; et al. A Single-Cell Atlas of the Tumor and Immune Ecosystem of Human Breast Cancer. Cell 2019, 177, 1330–1345 e1318. [Google Scholar] [CrossRef] [Green Version]
  99. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
  100. Chanmee, T.; Ontong, P.; Konno, K.; Itano, N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers 2014, 6, 1670–1690. [Google Scholar] [CrossRef]
  101. Wu, Q.; Li, J.; Li, Z.; Sun, S.; Zhu, S.; Wang, L.; Wu, J.; Yuan, J.; Zhang, Y.; Sun, S.; et al. Exosomes from the tumour-adipocyte interplay stimulate beige/brown differentiation and reprogram metabolism in stromal adipocytes to promote tumour progression. J. Exp. Clin. Cancer Res. 2019, 38, 223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Wei, X.; Li, S.; He, J.; Du, H.; Liu, Y.; Yu, W.; Hu, H.; Han, L.; Wang, C.; Li, H.; et al. Tumor-secreted PAI-1 promotes breast cancer metastasis via the induction of adipocyte-derived collagen remodeling. Cell Commun. Signal 2019, 17, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Gustafson, B.; Smith, U. Activation of canonical wingless-type MMTV integration site family (Wnt) signaling in mature adipocytes increases beta-catenin levels and leads to cell dedifferentiation and insulin resistance. J. Biol. Chem. 2010, 285, 14031–14041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wang, F.; Gao, S.; Chen, F.; Fu, Z.; Yin, H.; Lu, X.; Yu, J.; Lu, C. Mammary fat of breast cancer: Gene expression profiling and functional characterization. PLoS ONE 2014, 9, e109742. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, Q.; Sun, S.; Li, Z.; Yang, Q.; Li, B.; Zhu, S.; Wang, L.; Wu, J.; Yuan, J.; Yang, C.; et al. Tumour-originated exosomal miR-155 triggers cancer-associated cachexia to promote tumour progression. Mol. Cancer 2018, 17, 155. [Google Scholar] [CrossRef] [Green Version]
  106. Nieman, K.M.; Kenny, H.A.; Penicka, C.V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M.R.; Romero, I.L.; Carey, M.S.; Mills, G.B.; Hotamisligil, G.S.; et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011, 17, 1498–1503. [Google Scholar] [CrossRef] [Green Version]
  107. Zoico, E.; Darra, E.; Rizzatti, V.; Budui, S.; Franceschetti, G.; Mazzali, G.; Rossi, A.P.; Fantin, F.; Menegazzi, M.; Cinti, S.; et al. Adipocytes WNT5a mediated dedifferentiation: A possible target in pancreatic cancer microenvironment. Oncotarget 2016, 7, 20223–20235. [Google Scholar] [CrossRef] [Green Version]
  108. Notarnicola, M.; Miccolis, A.; Tutino, V.; Lorusso, D.; Caruso, M.G. Low levels of lipogenic enzymes in peritumoral adipose tissue of colorectal cancer patients. Lipids 2012, 47, 59–63. [Google Scholar] [CrossRef]
  109. Ham, S.; Lima, L.G.; Chai, E.P.Z.; Muller, A.; Lobb, R.J.; Krumeich, S.; Wen, S.W.; Wiegmans, A.P.; Moller, A. Breast Cancer-Derived Exosomes Alter Macrophage Polarization via gp130/STAT3 Signaling. Front. Immunol. 2018, 9, 871. [Google Scholar] [CrossRef] [Green Version]
  110. Tripathi, C.; Tewari, B.N.; Kanchan, R.K.; Baghel, K.S.; Nautiyal, N.; Shrivastava, R.; Kaur, H.; Bhatt, M.L.; Bhadauria, S. Macrophages are recruited to hypoxic tumor areas and acquire a pro-angiogenic M2-polarized phenotype via hypoxic cancer cell derived cytokines Oncostatin M and Eotaxin. Oncotarget 2014, 5, 5350–5368. [Google Scholar] [CrossRef]
  111. Ying, X.; Wu, Q.; Wu, X.; Zhu, Q.; Wang, X.; Jiang, L.; Chen, X.; Wang, X. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget 2016, 7, 43076–43087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Wen, Z.; Liu, H.; Li, M.; Li, B.; Gao, W.; Shao, Q.; Fan, B.; Zhao, F.; Wang, Q.; Xie, Q.; et al. Increased metabolites of 5-lipoxygenase from hypoxic ovarian cancer cells promote tumor-associated macrophage infiltration. Oncogene 2015, 34, 1241–1252. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, X.J.; Wu, S.; Yan, R.M.; Fan, L.S.; Yu, L.; Zhang, Y.M.; Wei, W.F.; Zhou, C.F.; Wu, X.G.; Zhong, M.; et al. The role of the hypoxia-Nrp-1 axis in the activation of M2-like tumor-associated macrophages in the tumor microenvironment of cervical cancer. Mol. Carcinog 2019, 58, 388–397. [Google Scholar] [CrossRef]
  114. Chen, X.J.; Deng, Y.R.; Wang, Z.C.; Wei, W.F.; Zhou, C.F.; Zhang, Y.M.; Yan, R.M.; Liang, L.J.; Zhong, M.; Liang, L.; et al. Hypoxia-induced ZEB1 promotes cervical cancer progression via CCL8-dependent tumour-associated macrophage recruitment. Cell Death Dis. 2019, 10, 508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Dai, E.; Han, L.; Liu, J.; Xie, Y.; Kroemer, G.; Klionsky, D.J.; Zeh, H.J.; Kang, R.; Wang, J.; Tang, D. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy 2020, 16, 2069–2083. [Google Scholar] [CrossRef] [PubMed]
  116. Shinohara, H.; Kuranaga, Y.; Kumazaki, M.; Sugito, N.; Yoshikawa, Y.; Takai, T.; Taniguchi, K.; Ito, Y.; Akao, Y. Regulated Polarization of Tumor-Associated Macrophages by miR-145 via Colorectal Cancer-Derived Extracellular Vesicles. J. Immunol. 2017, 199, 1505–1515. [Google Scholar] [CrossRef] [Green Version]
  117. Wang, D.; Wang, X.; Si, M.; Yang, J.; Sun, S.; Wu, H.; Cui, S.; Qu, X.; Yu, X. Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer Lett. 2020, 474, 36–52. [Google Scholar] [CrossRef]
  118. Zou, K.; Wang, Y.; Hu, Y.; Zheng, L.; Xu, W.; Li, G. Specific tumor-derived CCL2 mediated by pyruvate kinase M2 in colorectal cancer cells contributes to macrophage recruitment in tumor microenvironment. Tumour Biol. 2017, 39, 1010428317695962. [Google Scholar] [CrossRef] [Green Version]
  119. Bohn, T.; Rapp, S.; Luther, N.; Klein, M.; Bruehl, T.J.; Kojima, N.; Aranda Lopez, P.; Hahlbrock, J.; Muth, S.; Endo, S.; et al. Tumor immunoevasion via acidosis-dependent induction of regulatory tumor-associated macrophages. Nat. Immunol. 2018, 19, 1319–1329. [Google Scholar] [CrossRef]
  120. Colegio, O.R.; Chu, N.Q.; Szabo, A.L.; Chu, T.; Rhebergen, A.M.; Jairam, V.; Cyrus, N.; Brokowski, C.E.; Eisenbarth, S.C.; Phillips, G.M.; et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014, 513, 559–563. [Google Scholar] [CrossRef]
  121. Huber, R.; Meier, B.; Otsuka, A.; Fenini, G.; Satoh, T.; Gehrke, S.; Widmer, D.; Levesque, M.P.; Mangana, J.; Kerl, K.; et al. Tumour hypoxia promotes melanoma growth and metastasis via High Mobility Group Box-1 and M2-like macrophages. Sci. Rep. 2016, 6, 29914. [Google Scholar] [CrossRef] [Green Version]
  122. Zhang, J.; Li, H.; Wu, Q.; Chen, Y.; Deng, Y.; Yang, Z.; Zhang, L.; Liu, B. Tumoral NOX4 recruits M2 tumor-associated macrophages via ROS/PI3K signaling-dependent various cytokine production to promote NSCLC growth. Redox Biol. 2019, 22, 101116. [Google Scholar] [CrossRef] [PubMed]
  123. Arakaki, R.; Yamasaki, T.; Kanno, T.; Shibasaki, N.; Sakamoto, H.; Utsunomiya, N.; Sumiyoshi, T.; Shibuya, S.; Tsuruyama, T.; Nakamura, E.; et al. CCL2 as a potential therapeutic target for clear cell renal cell carcinoma. Cancer. Med. 2016, 5, 2920–2933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Yang, H.; Zhang, Q.; Xu, M.; Wang, L.; Chen, X.; Feng, Y.; Li, Y.; Zhang, X.; Cui, W.; Jia, X. CCL2-CCR2 axis recruits tumor associated macrophages to induce immune evasion through PD-1 signaling in esophageal carcinogenesis. Mol. Cancer 2020, 19, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. 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. [Google Scholar] [CrossRef]
  126. Suarez-Najera, L.E.; Chanona-Perez, J.J.; Valdivia-Flores, A.; Marrero-Rodriguez, D.; Salcedo-Vargas, M.; Garcia-Ruiz, D.I.; Castro-Reyes, M.A. Morphometric study of adipocytes on breast cancer by means of photonic microscopy and image analysis. Microsc. Res. Tech. 2018, 81, 240–249. [Google Scholar] [CrossRef]
  127. Choi, J.; Cha, Y.J.; Koo, J.S. Adipocyte biology in breast cancer: From silent bystander to active facilitator. Prog. Lipid Res. 2018, 69, 11–20. [Google Scholar] [CrossRef]
  128. Aird, R.; Wills, J.; Roby, K.F.; Benezech, C.; Stimson, R.H.; Wabitsch, M.; Pollard, J.W.; Finch, A.; Michailidou, Z. Hypoxia-driven metabolic reprogramming of adipocytes fuels cancer cell proliferation. Front. Endocrinol. 2022, 13, 989523. [Google Scholar] [CrossRef]
  129. Yao-Borengasser, A.; Monzavi-Karbassi, B.; Hedges, R.A.; Rogers, L.J.; Kadlubar, S.A.; Kieber-Emmons, T. Adipocyte hypoxia promotes epithelial-mesenchymal transition-related gene expression and estrogen receptor-negative phenotype in breast cancer cells. Oncol. Rep. 2015, 33, 2689–2694. [Google Scholar] [CrossRef] [Green Version]
  130. Bartelt, A.; Heeren, J. Adipose tissue browning and metabolic health. Nat. Rev. Endocrinol. 2014, 10, 24–36. [Google Scholar] [CrossRef]
  131. Yin, X.; Chen, Y.; Ruze, R.; Xu, R.; Song, J.; Wang, C.; Xu, Q. The evolving view of thermogenic fat and its implications in cancer and metabolic diseases. Signal. Transduct. Target 2022, 7, 324. [Google Scholar] [CrossRef] [PubMed]
  132. Master, S.R.; Hartman, J.L.; D’Cruz, C.M.; Moody, S.E.; Keiper, E.A.; Ha, S.I.; Cox, J.D.; Belka, G.K.; Chodosh, L.A. Functional microarray analysis of mammary organogenesis reveals a developmental role in adaptive thermogenesis. Mol. Endocrinol. 2002, 16, 1185–1203. [Google Scholar] [CrossRef] [PubMed]
  133. Bleriot, C.; Chakarov, S.; Ginhoux, F. Determinants of Resident Tissue Macrophage Identity and Function. Immunity 2020, 52, 957–970. [Google Scholar] [CrossRef] [PubMed]
  134. Lawrence, T.; Natoli, G. Transcriptional regulation of macrophage polarization: Enabling diversity with identity. Nat. Rev. Immunol. 2011, 11, 750–761. [Google Scholar] [CrossRef]
  135. Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995. [Google Scholar] [CrossRef]
  136. Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar] [CrossRef] [Green Version]
  137. Fujiwara, N.; Kobayashi, K. Macrophages in inflammation. Curr. Drug Targets Inflamm. Allergy 2005, 4, 281–286. [Google Scholar] [CrossRef]
  138. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
  139. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef]
  140. Atri, C.; Guerfali, F.Z.; Laouini, D. Role of Human Macrophage Polarization in Inflammation during Infectious Diseases. Int. J. Mol. Sci. 2018, 19, 1801. [Google Scholar] [CrossRef]
  141. Sinha, P.; Clements, V.K.; Ostrand-Rosenberg, S. Reduction of myeloid-derived suppressor cells and induction of M1 macrophages facilitate the rejection of established metastatic disease. J. Immunol. 2005, 174, 636–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Gordon, S.; Martinez, F.O. Alternative activation of macrophages: Mechanism and functions. Immunity 2010, 32, 593–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Hao, N.B.; Lu, M.H.; Fan, Y.H.; Cao, Y.L.; Zhang, Z.R.; Yang, S.M. Macrophages in tumor microenvironments and the progression of tumors. Clin. Dev. Immunol. 2012, 2012, 948098. [Google Scholar] [CrossRef] [Green Version]
  144. Sica, A.; Saccani, A.; Bottazzi, B.; Polentarutti, N.; Vecchi, A.; van Damme, J.; Mantovani, A. Autocrine production of IL-10 mediates defective IL-12 production and NF-kappa B activation in tumor-associated macrophages. J. Immunol. 2000, 164, 762–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Hourani, T.; Holden, J.A.; Li, W.; Lenzo, J.C.; Hadjigol, S.; O’Brien-Simpson, N.M. Tumor Associated Macrophages: Origin, Recruitment, Phenotypic Diversity, and Targeting. Front. Oncol. 2021, 11, 788365. [Google Scholar] [CrossRef] [PubMed]
  146. Chen, Y.; Song, Y.; Du, W.; Gong, L.; Chang, H.; Zou, Z. Tumor-associated macrophages: An accomplice in solid tumor progression. J. Biomed. Sci. 2019, 26, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Jeong, H.; Hwang, I.; Kang, S.H.; Shin, H.C.; Kwon, S.Y. Tumor-Associated Macrophages as Potential Prognostic Biomarkers of Invasive Breast Cancer. J. Breast Cancer 2019, 22, 38–51. [Google Scholar] [CrossRef] [PubMed]
  148. Ruffell, B.; Coussens, L.M. Macrophages and therapeutic resistance in cancer. Cancer Cell 2015, 27, 462–472. [Google Scholar] [CrossRef] [Green Version]
  149. Henze, A.T.; Mazzone, M. The impact of hypoxia on tumor-associated macrophages. J. Clin. Investig. 2016, 126, 3672–3679. [Google Scholar] [CrossRef]
  150. Haeggstrom, J.Z.; Funk, C.D. Lipoxygenase and leukotriene pathways: Biochemistry, biology, and roles in disease. Chem. Rev. 2011, 111, 5866–5898. [Google Scholar] [CrossRef]
  151. Grimshaw, M.J.; Wilson, J.L.; Balkwill, F.R. Endothelin-2 is a macrophage chemoattractant: Implications for macrophage distribution in tumors. Eur. J. Immunol. 2002, 32, 2393–2400. [Google Scholar] [CrossRef] [PubMed]
  152. Korsisaari, N.; Kasman, I.M.; Forrest, W.F.; Pal, N.; Bai, W.; Fuh, G.; Peale, F.V.; Smits, R.; Ferrara, N. Inhibition of VEGF-A prevents the angiogenic switch and results in increased survival of Apc+/min mice. Proc. Natl. Acad. Sci. USA 2007, 104, 10625–10630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Matschurat, S.; Knies, U.E.; Person, V.; Fink, L.; Stoelcker, B.; Ebenebe, C.; Behrensdorf, H.A.; Schaper, J.; Clauss, M. Regulation of EMAP II by hypoxia. Am. J. Pathol. 2003, 162, 93–103. [Google Scholar] [CrossRef] [Green Version]
  154. Guo, C.; Buranych, A.; Sarkar, D.; Fisher, P.B.; Wang, X.Y. The role of tumor-associated macrophages in tumor vascularization. Vasc. Cell 2013, 5, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Liu, P.S.; Wang, H.; Li, X.; Chao, T.; Teav, T.; Christen, S.; Di Conza, G.; Cheng, W.C.; Chou, C.H.; Vavakova, M.; et al. alpha-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 2017, 18, 985–994. [Google Scholar] [CrossRef] [PubMed]
  156. Waters, A.M.; Der, C.J. KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb Perspect Med 2018, 8, a031435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [Green Version]
  158. Weitzenfeld, P.; Ben-Baruch, A. The chemokine system, and its CCR5 and CXCR4 receptors, as potential targets for personalized therapy in cancer. Cancer Lett. 2014, 352, 36–53. [Google Scholar] [CrossRef] [PubMed]
  159. Perez-Torres, I.; Castrejon-Tellez, V.; Soto, M.E.; Rubio-Ruiz, M.E.; Manzano-Pech, L.; Guarner-Lans, V. Oxidative Stress, Plant Natural Antioxidants, and Obesity. Int. J. Mol. Sci. 2021, 22, 1786. [Google Scholar] [CrossRef]
  160. Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 2018, 80, 50–64. [Google Scholar] [CrossRef]
  161. Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef] [PubMed]
  162. Rani, V.; Deep, G.; Singh, R.K.; Palle, K.; Yadav, U.C. Oxidative stress and metabolic disorders: Pathogenesis and therapeutic strategies. Life Sci. 2016, 148, 183–193. [Google Scholar] [CrossRef] [PubMed]
  163. Wu, X.; Pfalzer, A.C.; Koh, G.Y.; Tang, S.; Crott, J.W.; Thomas, M.J.; Meydani, M.; Mason, J.B. Curcumin and Salsalate Suppresses Colonic Inflammation and Procarcinogenic Signaling in High-Fat-Fed, Azoxymethane-Treated Mice. J. Agric. Food Chem. 2017, 65, 7200–7209. [Google Scholar] [CrossRef] [PubMed]
  164. Wu, X.; Koh, G.Y.; Huang, Y.; Crott, J.W.; Bronson, R.T.; Mason, J.B. The Combination of Curcumin and Salsalate is Superior to Either Agent Alone in Suppressing Pro-Cancerous Molecular Pathways and Colorectal Tumorigenesis in Obese Mice. Mol. Nutr. Food Res. 2019, 63, e1801097. [Google Scholar] [CrossRef] [PubMed]
  165. Kubota, M.; Shimizu, M.; Sakai, H.; Yasuda, Y.; Terakura, D.; Baba, A.; Ohno, T.; Tsurumi, H.; Tanaka, T.; Moriwaki, H. Preventive effects of curcumin on the development of azoxymethane-induced colonic preneoplastic lesions in male C57BL/KsJ-db/db obese mice. Nutr. Cancer 2012, 64, 72–79. [Google Scholar] [CrossRef]
  166. Pettan-Brewer, C.; Morton, J.; Mangalindan, R.; Ladiges, W. Curcumin suppresses intestinal polyps in APC Min mice fed a high fat diet. Pathobiol. Aging Age Relat. Dis. 2011, 1, 7013. [Google Scholar] [CrossRef] [Green Version]
  167. Shimizu, M.; Shirakami, Y.; Sakai, H.; Adachi, S.; Hata, K.; Hirose, Y.; Tsurumi, H.; Tanaka, T.; Moriwaki, H. (-)-Epigallocatechin gallate suppresses azoxymethane-induced colonic premalignant lesions in male C57BL/KsJ-db/db mice. Cancer Prev. Res. 2008, 1, 298–304. [Google Scholar] [CrossRef] [Green Version]
  168. Miethe, C.; Nix, H.; Martin, R.; Hernandez, A.R.; Price, R.S. Silibinin Reduces the Impact of Obesity on Invasive Liver Cancer. Nutr. Cancer 2017, 69, 1272–1280. [Google Scholar] [CrossRef]
  169. Ke, J.Y.; Banh, T.; Hsiao, Y.H.; Cole, R.M.; Straka, S.R.; Yee, L.D.; Belury, M.A. Citrus flavonoid naringenin reduces mammary tumor cell viability, adipose mass, and adipose inflammation in obese ovariectomized mice. Mol. Nutr. Food Res. 2017, 61, 1600934. [Google Scholar] [CrossRef]
  170. Xing, F.; Zhao, D.; Wu, S.Y.; Tyagi, A.; Wu, K.; Sharma, S.; Liu, Y.; Deshpande, R.; Wang, Y.; Cleary, J.; et al. Epigenetic and Posttranscriptional Modulation of SOS1 Can Promote Breast Cancer Metastasis through Obesity-Activated c-Met Signaling in African-American Women. Cancer Res. 2021, 81, 3008–3021. [Google Scholar] [CrossRef]
  171. Rossi, E.L.; Khatib, S.A.; Doerstling, S.S.; Bowers, L.W.; Pruski, M.; Ford, N.A.; Glickman, R.D.; Niu, M.; Yang, P.; Cui, Z.; et al. Resveratrol inhibits obesity-associated adipose tissue dysfunction and tumor growth in a mouse model of postmenopausal claudin-low breast cancer. Mol. Carcinog. 2018, 57, 393–407. [Google Scholar] [CrossRef]
  172. Theriau, C.F.; Sauve, O.S.; Beaudoin, M.S.; Wright, D.C.; Connor, M.K. Proliferative endocrine effects of adipose tissue from obese animals on MCF7 cells are ameliorated by resveratrol supplementation. PLoS ONE 2017, 12, e0183897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Hama, S.; Nishi, T.; Isono, E.; Itakura, S.; Yoshikawa, Y.; Nishimoto, A.; Suzuki, S.; Kirimura, N.; Todo, H.; Kogure, K. Intraperitoneal administration of nanoparticles containing tocopheryl succinate prevents peritoneal dissemination. Cancer Sci. 2022, 113, 1779–1788. [Google Scholar] [CrossRef] [PubMed]
  174. Sekar, V.; Anandasadagopan, S.K.; Ganapasam, S. Genistein regulates tumor microenvironment and exhibits anticancer effect in dimethyl hydrazine-induced experimental colon carcinogenesis. Biofactors 2016, 42, 623–637. [Google Scholar] [CrossRef] [PubMed]
  175. Li, D.; Zhang, Y.; Liu, K.; Zhao, Y.; Xu, B.; Xu, L.; Tan, L.; Tian, Y.; Li, C.; Zhang, W.; et al. Berberine inhibits colitis-associated tumorigenesis via suppressing inflammatory responses and the consequent EGFR signaling-involved tumor cell growth. Lab. Investig. 2017, 97, 1343–1353. [Google Scholar] [CrossRef] [Green Version]
  176. Chen, H.; Ye, C.; Cai, B.; Zhang, F.; Wang, X.; Zhang, J.; Zhang, Z.; Guo, Y.; Yao, Q. Berberine inhibits intestinal carcinogenesis by suppressing intestinal pro-inflammatory genes and oncogenic factors through modulating gut microbiota. BMC Cancer 2022, 22, 566. [Google Scholar] [CrossRef]
  177. Deng, J.; Zhao, L.; Yuan, X.; Li, Y.; Shi, J.; Zhang, H.; Zhao, Y.; Han, L.; Wang, H.; Yan, Y.; et al. Pre-Administration of Berberine Exerts Chemopreventive Effects in AOM/DSS-Induced Colitis-Associated Carcinogenesis Mice via Modulating Inflammation and Intestinal Microbiota. Nutrients 2022, 14, 726. [Google Scholar] [CrossRef]
  178. Zhao, L.; Zhang, C. Berberine Inhibits MDA-MB-231 Cells by Attenuating Their Inflammatory Responses. Biomed. Res. Int. 2020, 2020, 3617514. [Google Scholar] [CrossRef] [Green Version]
  179. Gonzalez Suarez, N.; Rodriguez Torres, S.; Ouanouki, A.; El Cheikh-Hussein, L.; Annabi, B. EGCG Inhibits Adipose-Derived Mesenchymal Stem Cells Differentiation into Adipocytes and Prevents a STAT3-Mediated Paracrine Oncogenic Control of Triple-Negative Breast Cancer Cell Invasive Phenotype. Molecules 2021, 26, 1506. [Google Scholar] [CrossRef]
  180. Gonzalez Suarez, N.; Fernandez-Marrero, Y.; Torabidastgerdooei, S.; Annabi, B. EGCG Prevents the Onset of an Inflammatory and Cancer-Associated Adipocyte-like Phenotype in Adipose-Derived Mesenchymal Stem/Stromal Cells in Response to the Triple-Negative Breast Cancer Secretome. Nutrients 2022, 14, 1099. [Google Scholar] [CrossRef]
  181. Chen, M.L.; Lai, C.J.; Lin, Y.N.; Huang, C.M.; Lin, Y.H. Multifunctional nanoparticles for targeting the tumor microenvironment to improve synergistic drug combinations and cancer treatment effects. J. Mater. Chem. B 2020, 8, 10416–10427. [Google Scholar] [CrossRef] [PubMed]
  182. Li, M.; Yue, X.; Wang, Y.; Zhang, J.; Kan, L.; Jing, Z. Remodeling the tumor microenvironment to improve drug permeation and antitumor effects by co-delivering quercetin and doxorubicin. J. Mater. Chem. B 2019, 7, 7619–7626. [Google Scholar] [CrossRef] [PubMed]
  183. Liu, Y.; Fu, R.; Tu, S.; Qu, D.; Liu, C.; Li, X.; Zhao, Y.; Ma, C.; Chen, Y.; Tao, L. Extracellular Microparticles Encapsulated with Diallyl Trisulfide Interfere with the Inflammatory Tumor Microenvironment and Lung Metastasis of Invasive Melanoma. Mol. Pharm. 2021, 18, 822–835. [Google Scholar] [CrossRef]
  184. Abdelghany, L.; Zhang, X.; Kawabata, T.; Goto, S.; El-Mahdy, N.; Jingu, K.; Li, T.S. Nicaraven prevents the fast growth of inflamed tumors by an anti-inflammatory mechanism. Med. Oncol. 2021, 39, 7. [Google Scholar] [CrossRef] [PubMed]
  185. Lo Iacono, M.; Modica, C.; Porcelli, G.; Brancato, O.R.; Muratore, G.; Bianca, P.; Gaggianesi, M.; Turdo, A.; Veschi, V.; Todaro, M.; et al. Targeting of the Peritumoral Adipose Tissue Microenvironment as an Innovative Antitumor Therapeutic Strategy. Biomolecules 2022, 12, 702. [Google Scholar] [CrossRef] [PubMed]
  186. Mandal, S.K.; Kumar, B.K.; Sharma, P.K.; Murugesan, S.; Deepa, P.R. In silico and in vitro analysis of PPAR—Alpha/gamma dual agonists: Comparative evaluation of potential phytochemicals with anti-obesity drug orlistat. Comput. Biol. Med. 2022, 147, 105796. [Google Scholar] [CrossRef]
  187. Chen, S.; Zhao, Z.; Ke, L.; Li, Z.; Li, W.; Zhang, Z.; Zhou, Y.; Feng, X.; Zhu, W. Resveratrol improves glucose uptake in insulin-resistant adipocytes via Sirt1. J. Nutr. Biochem. 2018, 55, 209–218. [Google Scholar] [CrossRef]
  188. Ding, Z.; Chen, M.; Tao, X.; Liu, Y.; He, J.; Wang, T.; Li, X. Synergistic Treatment of Obesity via Locally Promoting Beige Adipogenesis and Antioxidative Defense in Adipose Tissues. ACS Biomater. Sci. Eng. 2021, 7, 727–738. [Google Scholar] [CrossRef]
  189. Han, Y.H.; Kee, J.Y.; Park, J.; Kim, H.L.; Jeong, M.Y.; Kim, D.S.; Jeon, Y.D.; Jung, Y.; Youn, D.H.; Kang, J.; et al. Arctigenin Inhibits Adipogenesis by Inducing AMPK Activation and Reduces Weight Gain in High-Fat Diet-Induced Obese Mice. J. Cell Biochem. 2016, 117, 2067–2077. [Google Scholar] [CrossRef]
Figure 1. Endocrine and paracrine communication between adipocytes, macrophages, and cancer cells. Obesity-induced changes in the adipose tissue result in increased expression of cytokines, adipokines, metabolites, and extracellular vesicles (EVs) that influence cancer cell behavior and promote tumor development. In addition, in the tumor microenvironment (TME), adipocytes and macrophages are key components that interact with cancer cells by secreting a variety of signaling molecules, including, adipokines, cytokines, metabolites, fatty acids and EVs, which increase local inflammation and oxidative stress to generate a favorable microenvironment for tumor progression. Some antioxidants discussed in Section 5 and the pathways they control are also indicated.
Figure 1. Endocrine and paracrine communication between adipocytes, macrophages, and cancer cells. Obesity-induced changes in the adipose tissue result in increased expression of cytokines, adipokines, metabolites, and extracellular vesicles (EVs) that influence cancer cell behavior and promote tumor development. In addition, in the tumor microenvironment (TME), adipocytes and macrophages are key components that interact with cancer cells by secreting a variety of signaling molecules, including, adipokines, cytokines, metabolites, fatty acids and EVs, which increase local inflammation and oxidative stress to generate a favorable microenvironment for tumor progression. Some antioxidants discussed in Section 5 and the pathways they control are also indicated.
Antioxidants 12 00126 g001
Table 1. Endocrine communication between adipose tissue and the tumor microenvironment.
Table 1. Endocrine communication between adipose tissue and the tumor microenvironment.
Cancer TypeMechanismRefs.
Increased cancer-associated mortalityInflammation and metabolism[31]
Colitis-associated cancerIL-6, CCL-20/CCR-6[32]
Colon cancer, colorectal adenocarcinoma, and hepatocellular carcinomaSurvivin[33]
Liver cancerVisfatin and Resistin
Akt and ERK, ROS, MMP-9
[11]
Hepatocellular carcinomaPI3Kγ, insulinemia and steatosis[34]
Liver and uterine cancermicroRNA, fatty acid and arachidonic acid metabolism, cell growth, and inflammation[35]
Endometrial CancerLeptin, proinflammatory cytokines (IL-6) and oxidative stress[36]
Breast cancerLeptin[37]
Breast cancerRelationship between adipocytes, immune cells and the tumor[38]
Breast cancerNLRC4, IL-1β, VEGF, angiogenesis[39]
Breast cancerANGPTL4, IL-1β, angiogenesis, macrophage tumor infiltration[40]
Breast cancerPPARα and FAK-signaling (inflammation and lipid metabolism, such as PPARα, IL-1β, and ANGPTL4)[41]
Breast cancerWhite adipose tissue inflammation, higher serum lipid, glucose, C-reactive protein levels and CLS[42]
Breast cancerWhite adipose tissue inflammation, CLS, insulin, glucose, triglycerides, HDL, leptin, adiponectin, C-reactive protein, and IL-6[43]
Breast cancerGM-CSF and MMP-9[44]
Breast cancerIL-5 and GM-CSF[45]
Breast cancerIL-6, SphK1/S1P/S1PR1[46]
Lung cancerLipid metabolism and oxidative stress[47]
Prostate CancerIL-6, MDSCs, M2/M1 tumor infiltration, pSTAT3[48]
Prostate CancerIL-6, VEGF, CXCL1/2/13[49]
ANGPTL4: angiopoietin-like 4; CCL-20: CC-chemokine-ligand-20; CCR-6: CC-chemokine-receptor-6; CLS: crown-like structures; CXCL1/2/13: C-X-C motif chemokine ligand 1/2/13; FAK: focal adhesion kinase; GM-CSF: granulocyte-macrophage colony-stimulating factor; IL-1β: interleukin-1β; IL-5: interleukin-5; IL-6: interleukin-6; MDSCs: myeloid-derived suppressor cells; MMP-9: matrix metalloproteinase 9; NLRC4: NOD-like receptor 4; PI3K: phosphoinositide 3-kinase; PPARα: peroxisome proliferator-activated receptor alpha; ROS: reactive oxygen species; S1P: sphingosine-1-phosphate; S1PR1: sphingosine-1-phosphate receptor 1; SphK1: sphingosine kinase 1; STAT3: signal transducer and activator of transcription 3; VEGF: vascular endothelial growth factor.
Table 2. Paracrine communication between adipocytes, macrophages, and cancer cells in the tumor microenvironment.
Table 2. Paracrine communication between adipocytes, macrophages, and cancer cells in the tumor microenvironment.
Signals from Adipocytes to Cancer Cells
Cancer TypeMechanismsEffect on TMERefs.
Breast cancerβ-hydroxybutyrate increases histone acetylation and upregulates the expression of tumor-promoting genes (IL-1β and LCN2)Tumor growth and poor prognosis[54]
Breast cancerA positive feedback loop between the cytokines LIF and CXCLsInvasion and metastasis[55]
Breast cancerG-CSF/Stat3EMT, migration, and invasion[56]
Breast cancerUpregulation of MMP9, TWIST1 and VimentinEMT, migration, and invasion[57]
Breast cancerIGFBP-2 and MMP-2Adipocytes acquire CAA-like phenotype. Migration, invasion, and metastasis.[58]
Breast cancerIL-1β/ANGPTL4Tumor growth and angiogenesis[40]
Breast cancerIL-8Angiogenesis and breast cancer cell dissemination[59]
Breast cancerIL-8/STAT3Angiogenesis and tumorigenesis[60]
Breast cancerCytokines (CCL2, CCL5, IL-6, IL-8, CXCL10) activate Src to upregulate Sox2 and induce miR-302bCancer stem cell self-renewal and metastasis[61]
Breast cancerIL-6/Chk1 phosphorylationRadioresistant phenotype[62]
Breast cancerMajor vault proteinResistance to Doxorubicin[63]
Breast cancerIncrease expression of MMP-11, IL-6, and IL-1βInvasion and metastasis[64]
Breast cancerATGL/HSL-mediated fatty acids released by adipocytes.
FAO induction through increased CPT1A in cancer cells.
Proliferation and migration[65]
Breast cancerATGL-dependent lipolysis and uncoupled FAOInvasion[66]
Breast cancerNLRC4 inflammasome/IL-1β/VEGFAAngiogenesis[39]
Ovarian cancerFABP4Metastasis and Carboplatin resistance[67]
Ovarian cancerExosomal miR21/APAF1Invasion and Paclitaxel resistance[68]
Ovarian cancerCD36/Exogenous fatty acid uptakeMigration, invasion, and metastasis[69]
Ovarian cancerArachidonic acid/Akt pathwayCisplatin resistance[70]
MelanomaAdipocyte-EVs transfer proteins implicated in FAOMetabolic reprogramming, migration and invasion[71]
MelanomaAdipocyte-EVs transfer proteins required for FAOMigration[72]
MelanomaExpression of EMT genes (SNAI1, MMP-9, TWIST and Vimentin)Invasion[73]
MelanomaPI3K/Akt and MEK/ERK pathwaysResistance to chemotherapeutic drugs (Cisplatin and Docetaxel)[74]
Skin cancerAdipokine, ROS, E-cadherin, TWISTProliferation and tumor growth[75]
Prostate cancerCCR3/CCL7 axisMigration[76]
Gastric cancerOleic acid/PI3K/Akt/MMP2Increase in lipid uptake and invasion[77]
Gastric cancerDGAT2-dependent lipid droplet accumulation and redox homeostasisProliferation, migration and invasion[78]
Colon cancerUpregulation of autophagy and mitochondrial FAO via AMPKMigration and EMT[79]
Colon cancerCPT1A-dependent FAO promote the acetylation and nuclear translocation of β-cateninPromote cancer stem cell functions, cell proliferation and tumor growth[80]
Pancreatic cancerSAA1Adipocytes acquire CAA-like phenotype and induce migration, invasion, and EMT in cancer cells[81]
Oral squamous cell carcinoma, melanoma, breast cancerCD36/FAO-related genesPoor prognosis. Lymph node metastasis[82]
Signals from Macrophages to Cancer Cells
Cancer TypeMechanismsEffect on TMERefs.
Breast cancerWNT7B /VEGFAAngiogenesis, tumor growth, invasion and metastasis[83]
Breast cancerLactate and CCL5-CCR5 axisAerobic glycolysis and EMT[84]
Pancreatic, breast, and lung cancerIL-4-inducing cathepsin activity in macrophagesTumor growth, angiogenesis and invasion[85]
Pancreatic ductal adenocarcinomaM2-polarized TAMs, angiopoietin-axis and TIE2-expressing monocytesAngiogenesis and metastasis[86]
Lung cancerAMPK/PGC-1α, TNFα, PD-L1Aerobic glycolysis, tumor hypoxia and resistance to anticancer therapies[87]
AMPK: AMP-activated protein kinase; ANGPTL4: angiopoietin-like 4; APAF1: apoptotic peptidase activator factor 1; ATGL: adipose triglyceride lipase; CCL2: CC-chemokine-ligand-2; CCL5: CC-chemokine-ligand-5; CCR3: CC-chemokine-receptor-3; CCR5: CC-chemokine-receptor-5; Chk1: checkpoint kinase 1; CPT1A: carnitine palmitoyl transferase I; CXCLs: C-X-C motif chemokine ligands; DGAT2: diacylglycerol acyltransferase 2; EMT: epithelial-to-mesenchymal transition; EVs: extracellular vesicles; G-CSF: granulocyte colony-stimulating factor; IGFBP-2: insulin-like growth factor binding protein 2; IL-1β: interleukin-1β; IL-6: interleukin-6; IL-8: interleukin-8; LCN2: lipocalin 2; LIF: leukemia inhibitory factor; MMPs: matrix metalloproteinase; NLRC4: NOD-like receptor 4; PD-1: programmed cell death-1; PD-L1: programmed death ligand 1; PGC-1α: peroxisome proliferator-activated coactivator gamma 1-alpha; PPARγ: peroxisome proliferator-activated receptor gamma; SAA1: serum amyloid A1; TNFα: tumor necrosis factor alpha; TWIST1: twist-related protein 1; VEGFA: vascular endothelial growth factor A.
Table 3. Paracrine communication between cancer cells on adipocytes and macrophages in the tumor microenvironment.
Table 3. Paracrine communication between cancer cells on adipocytes and macrophages in the tumor microenvironment.
Signals from Cancer Cells to Adipocytes
Cancer TypeMechanismsEffect on TMERefs.
Breast cancerLower C/EBPα expression
IL-6/CCL2
Adipocytes revert to an immature and proliferative phenotype. Proliferation, migration and EMT[88]
Breast cancerIL-6Adipocytes acquire CAA-like phenotype and promote invasion in cancer cells[64]
Breast cancerATGL-dependent lipolysis and uncoupled FAOInduce a lipolytic process in adipocytes, which then become CAA. Invasion and metastasis[66]
Breast cancerExosomal miRNA-144 and miRNA-126Induce beige/brown differentiation in adipocytes. Metabolic reprogramming and tumor progression[101]
Breast cancerPAI-1, PLOD2, and PI3K/AKT-FOXP1 axisAdipocyte-derived collagen reorganization, which leads to invasion and metastasis[102]
Breast cancerWnt/β-catenin pathwayAdipocytes acquire CAA-like phenotype[103]
Breast cancerIncreases expression of fatty browning marker genes in adipose tissue adjacent to tumorsBrowning activity of adipocytes adjacent to cancer tissues[104]
Breast cancerExosomal miR-155/PPARγBrowning and metabolic remodeling of adipocytes. Cancer-associated cachexia and tumor progression[105]
Ovarian cancerHSL and FABP4Adipocyte lipolysis. Tumor growth and metastasis[106]
Pancreatic cancerWNT5aAdipocytes acquire CAA-like phenotype[107]
Colorectal cancerReduction of LPL and FASN in adipose tissue adjacent to tumorsImpairs lipid storage capacity of adipose tissue[108]
Signals from Cancer Cells to Macrophages
Cancer typeMechanismsEffect on TMERefs.
Breast cancerBreast cancer-derived EVs /IL-6/gp130/STAT3Induce TAM polarization[109]
Breast cancerHypoxia/Eotaxin and OncostatinMacrophage recruitment, M2 polarization and angiogenesis[110]
Ovarian cancerOvarian cancer-derived exosomes /SOCS3/STAT3 pathwayActivate macrophages to a TAM phenotype. Tumor growth and metastasis.[111]
Ovarian cancer5-LOX metabolites enhance the expression of MMP-7 through activation of the p38 pathwayMacrophage infiltration. Metastasis.[112]
Cervical cancerHypoxia/Nrp-1Macrophage recruitment and M2 polarization[113]
Cervical cancerHypoxia increases ZEB1 expression, which promotes CCL8 production via the CCR2–NF-κB pathwayMacrophage infiltration into hypoxic areas[114]
Pancreatic ductal adenocarcinomaEV release of KRASG12D/STAT3-dependent FAOM2 polarization[115]
Colorectal cancerExosomal miR-145/HDAC11 downregulationM2 polarization. Tumor progression[116]
Liver metastasis of colorectal cancerExosomal miRNAs/CXCL12/CXCR4/PTEN/PI3K/AktM2 polarization. EMT and metastasis[117]
Colorectal cancerPKM2/NF-κB p65–mediated CCL2 gene expressionRecruitment of macrophages[118]
MelanomaAcidification of TME induces GPCR-dependent expression of ICER in TAMsM2 polarization[119]
Lung carcinoma and melanomaAnaerobic glycolysis/Lactate/VEGF-A and Arginase-1M2 polarization and tumor growth[120]
MelanomaHypoxia-induced HMGB1 and RAGE-dependent IL-10 productionM2-like TAM polarization. Tumor growth and metastasis[121]
Nonsmall cell lung cancerNOX4, ROS/PI3K and cytokine production (CCL7, IL-8, CSF-1, VEGF-C)Macrophage recruitment, M2 polarization and tumor growth[122]
Renal cell carcinomaCCL2Macrophage infiltration. Tumor growth and angiogenesis.[123]
Esophageal squamous cell carcinomaCCL2-CCR2 axis and PD-1 signaling pathwayImmune evasion[124]
5-LOX: 5-lipoxygenase; C/EBPα: CCAAT/enhancer-binding protein alpha; CCL2: CC-chemokine-ligand-2; CCL7: CC-chemokine-ligand-7; CCL8: CC-chemokine-ligand-8; CCR2: CC-chemokine-receptor-2; CSF-1: colony stimulating factor 1; CXCLs: C-X-C motif chemokine ligands; EMT: epithelial-to-mesenchymal transition; FABP4: fatty acid-binding protein 4; FAO: fatty acid oxidation; FASN: fatty acid synthase; FOXP1: forkhead box P1; gp130: glycoprotein 130; GPCR: G-protein-coupled receptor; HDAC11: histone deacetylase 11; HMGB1: high-mobility group box 1; HSL: hormone-sensitive lipase; IL-10: interleukin-10; IL-4: interleukin-4; IL-6: interleukin-6; IL-8: interleukin-8; LPL: lipoprotein lipase; MMPs: matrix metalloproteinases; NF-κB: nuclear factor kappa B; NOX4: NADPH oxidase 4; Nrp-1: neuropilin-1; PAI-1: plasminogen activator inhibitor 1; PI3K: phosphoinositide 3-kinase; PKM2: pyruvate kinase M2; PLOD2: procollagen-lysine,2-oxoglutarate 5-dioxygenase 2; RAGE: receptor for advanced glycation end products; ROS: reactive oxygen species; SOCS3: suppressor of cytokine signaling 3; STAT3: signal transducer and activator of transcription 3; VEGF: vascular endothelial growth factor; ZEB1: zinc finger E-box binding homeobox 1.
Table 4. Antioxidants preventing cancer associated with obesity and systemic inflammation.
Table 4. Antioxidants preventing cancer associated with obesity and systemic inflammation.
DrugMechanismCancer TypeRefs.
Curcumin and SalsalateIL-1β, IL-6, Akt, NF-κBColon cancer[163]
Curcumin and SalsalatePI3K/Akt/mTOR/NF-κB/Wnt
cytokines
Colorectal cancer[164]
CurcuminAMPK, COX-2, NF-κB, TNF, IL-6, adiponectin, leptinColonic preneoplastic lesions[165]
CurcuminApoptosis, oxidation-induced DNA damage and DNA repairColorectal cancer[166]
Epigallocatechin gallateProtein levels of IGF-IR, p- GSK3β, beta-catenin, cyclooxygenase-2 and cyclin D1
Serum levels of IGF-I, insulin, triglyceride, cholesterol, and leptin
Colonic premalignant lesions[167]
SilibininObesity-induced cancer cells growth, ROS, lipogenesis, MMP-9 and cancer cell invasion
FASN, IL-6, IL-1β and Erk
Liver Cancer[168]
NaringeninAdiposity and inflammation
AMPK
Breast cancer[169]
TaxifolinAdipocyte, HGF, SOS1, Grb2, CCL2, macrophagesBreast cancer[170]
ResveratrolAdipocyte hypertrophy
macrophage infiltration
COX-2, PPARγ
Breast cancer[171]
ResveratrolProliferation of cancer cells
AMPK, Akt
Breast cancer[172]
AMPK: AMP-activated protein kinase; CCL2: CC-chemokine-ligand-2; COX-2: cyclooxygenase-2; FASN: fatty acid synthase; Grb2: growth factor receptor bound protein 2; GSK3β: glycogen synthase kinase-3 beta; HGF: hepatocyte growth factor; IGF-I: insulin-like growth factor I; IGF-IR: insulin-like growth factor I receptor; IL-1β: interleukin-1β; IL-6: interleukin-6; MMP-9: matrix metalloproteinase-9; mTOR: mammalian target of rapamycin; NF-κB: nuclear factor kappa B; PI3K: phosphoinositide 3-kinase; PPARγ: peroxisome proliferator-activated receptor gamma; ROS: reactive oxygen species; TNF: tumor necrosis factor.
Table 5. Antioxidant effects in the tumor microenvironment.
Table 5. Antioxidant effects in the tumor microenvironment.
DrugMechanismCancer TypeRefs.
TaxifolinAdipocyte, HGF, SOS1, Grb2, CCL2, macrophagesBreast cancer[170]
ResveratrolAdipocyte hypertrophy
Macrophage infiltration
COX-2, PPARγ
Breast cancer[171]
Vitamin E succinateVascular endothelial growth factor A, IL-10, and M2-like phenotype of tumor-associated macrophagesColon cancer[173]
GenisteinReverts oxidative stress-induced cancer
CD133, CD44, and β-catenin
Colon cancer[174]
BerberineEGFR-ERK
TNF-α and IL-6
Colitis-associated tumorigenesis[175]
BerberineIL-1β, TNF, NF-κBIntestinal carcinogenesis[176]
BerberineIL-6, IL-1β, COX-2 and TNF-α
Microbiota
STAT3, JNK and β-catenin
Colitis-associated carcinogenesis[177]
BerberineTNF-α and IL-6Breast cancer[178]
Epigallocatechin-3-gallateAdipogenesis, STAT3Breast cancer[179]
Epigallocatechin-3-gallatePrevents cancer to TME effects.
CCL2, CCL5, IL-1β and IL-6
COX2, HIF-1α, VEGFα, and PD-L1
Smad2 and NF-κB
Breast cancer[180]
Epigallocatechin-3-gallateCD44Prostate cancer[181]
Quercetin and doxorubicinCD44Liver cancer[182]
Diallyl trisulfideS100A8/A9, serum amyloid A (SAA), IL-6, fibronectin, MRP8, myeloperoxidase (MPO) and TLR4-Myd88Melanoma and lung metastasis[183]
NicaravenCXCL10, SDF-1, IL-2 and MIP-2
MAPK
Lung carcinoma[184]
CCL2: CC-chemokine-ligand-2; COX-2: cyclooxygenase-2; CCL5:CC-chemokine-ligand-5; CXCL10: C-X-C motif chemokine ligand-10; EGFR: epidermal growth factor receptor; Grb2: growth factor receptor bound protein 2; HGF: hepatocyte growth factor; HIF-1α: hypoxia-inducible factor 1 subunit alpha; IL-10: interleukin-10; IL-1β: interleukin-1β; IL-6: interleukin-6; JNK: c-Jun N-terminal kinase; MAPK: mitogen-activated protein kinase; MIP-2: macrophage inflammatory protein-2; MPO: myeloperoxidase; MRP8: myeloid-related protein 8; Myd88: myeloid differentiation primary response 88; NF-κB: nuclear factor kappa B; PD-L1: programmed death ligand 1; PPARγ: peroxisome proliferator-activated receptor gamma; SAA: serum amyloid A; SDF-1: stromal cell-derived factor 1; Smad2: small mother against decapentaplegic 2; STAT3: signal transducer and activator of transcription 3; TLR4: toll like receptor 4; TNF-α: tumor necrosis factor-alpha; VEGFα: vascular endothelial growth factor alpha.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sanhueza, S.; Simón, L.; Cifuentes, M.; Quest, A.F.G. The Adipocyte–Macrophage Relationship in Cancer: A Potential Target for Antioxidant Therapy. Antioxidants 2023, 12, 126. https://doi.org/10.3390/antiox12010126

AMA Style

Sanhueza S, Simón L, Cifuentes M, Quest AFG. The Adipocyte–Macrophage Relationship in Cancer: A Potential Target for Antioxidant Therapy. Antioxidants. 2023; 12(1):126. https://doi.org/10.3390/antiox12010126

Chicago/Turabian Style

Sanhueza, Sofía, Layla Simón, Mariana Cifuentes, and Andrew F. G. Quest. 2023. "The Adipocyte–Macrophage Relationship in Cancer: A Potential Target for Antioxidant Therapy" Antioxidants 12, no. 1: 126. https://doi.org/10.3390/antiox12010126

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop