Next Article in Journal
BRCA Mutations and Breast Cancer Prevention
Next Article in Special Issue
Obesity-Induced TNFα and IL-6 Signaling: The Missing Link between Obesity and Inflammation—Driven Liver and Colorectal Cancers
Previous Article in Journal
NK Cell-Based Glioblastoma Immunotherapy
Previous Article in Special Issue
Lipid Metabolic Reprogramming in Hepatocellular Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Obesity-Linked Cancers: Current Knowledge, Challenges and Limitations in Mechanistic Studies and Rodent Models

1
Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
2
Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
*
Author to whom correspondence should be addressed.
Cancers 2018, 10(12), 523; https://doi.org/10.3390/cancers10120523
Submission received: 15 November 2018 / Revised: 9 December 2018 / Accepted: 15 December 2018 / Published: 18 December 2018
(This article belongs to the Special Issue Obesity as a Risk Factor for Cancer)

Abstract

:
The worldwide prevalence of obesity has doubled during the last 50 years, and according to the World Obesity Federation, one third of the people on Earth will be obese by the year 2025. Obesity is described as a chronic, relapsing and multifactorial disease that causes metabolic, biomechanical, and psychosocial health consequences. Growing evidence suggests that obesity is a risk factor for multiple cancer types and rivals smoking as the leading preventable cause for cancer incidence and mortality. The epidemic of obesity will likely generate a new wave of obesity-related cancers with high aggressiveness and shortened latency. Observational studies have shown that from cancer risk to disease prognosis, an individual with obesity is consistently ranked worse compared to their lean counterpart. Mechanistic studies identified similar sets of abnormalities under obesity that may lead to cancer development, including ectopic fat storage, altered adipokine profiles, hormone fluctuations and meta-inflammation, but could not explain how these common mechanisms produce over 13 different cancer types. A major hurdle in the mechanistic underpinning of obesity-related cancer is the lack of suitable pre-clinical models that spontaneously develop obesity-linked cancers like humans. Current approaches and animal models fall short when discerning the confounders that often coexist in obesity. In this mini-review, we will briefly survey advances in the different obesity-linked cancers and discuss the challenges and limitations in the rodent models employed to study their relationship. We will also provide our perspectives on the future of obesity-linked cancer research.

1. Introduction

Obesity, in simple terms, is an increase in the white adipose tissue (WAT) of the body. Over the years, layers of complexity have been added to obesity studies by incorporating location and functional consequences of different adipose depots, influence from heterogeneous cell populations and secreted molecules, and local and systemic immunometabolic disruptions. Today, we are in an era where overnutrition kills more than malnutrition [1,2]. What evolved to be an advantage in energy storage has become detrimental in the modern world. After obesity is diagnosed by a Body Mass Index (BMI) of ≥30 kg/m2, a hurricane of complications often ensues, including high blood sugar and cholesterol, insulin resistance, elevated blood pressure, cardiovascular complications, metabolic syndrome and increased risk of cancers [3,4]. Cancer, a seemingly unrelated manifestation, may not be the first to come to mind nor an immediate concern regarding obesity, but this demonstrated linkage cannot be overlooked. Recent data have linked obesity to more than 13 different cancer types in the body, including those in the digestive tract and accessory organs (esophagus, stomach, colon, liver, kidney, gallbladder, pancreas), the female reproductive system (ovary, endometrium, postmenopausal breast), the male reproductive system (prostate) and others (meningioma, thyroid, multiple myeloma, and non-Hodgkin lymphoma) [1,5,6]. Together, these cancers account for about 8% of all cancer cases in North America [1]. Available statistics also suggest that in many western countries, obesity may have surpassed smoking as the leading cause of preventable cancer deaths [7,8]. Common mechanisms such as ectopic fat accumulation, altered adipokine production, hormone fluctuations and meta-inflammation (defined as a low-grade chronic inflammatory state induced under metabolic dysregulations) have been proposed to either independently or synergistically increase cancer risks of all types. The majority of cancers develops and metastasizes in the vicinity of an adipose tissue-rich environment. Initially, local inflammation begins with adipocyte hypertrophy and increased cell necrosis, which attract resident macrophages and promote recruitment of circulating monocytes, as evidenced by crown-like structures (CLSs) in the obese WAT. Interactions between hypertrophied adipocytes and pro-inflammatory immune cells further exacerbate the inflamed microenvironment, leading to meta-inflammation and cellular transformation. In parallel, overproduction of insulin and insulin-like factors under a state of insulin resistance promotes the proliferation of all cells, including cancers. While most adipokines and growth factors released under obesity induce general pathogenesis, others like estrogens have a more targeted influence over the female reproductive system. However, it is puzzling to researchers how a similar set of abnormalities produced by obesity leads to different types of cancers (Figure 1), and the underlying mechanisms that produced these diversified cancers remain elusive.
Obesity-related cancer takes time to manifest; and precisely for this reason, solutions are needed for early intervention. Although there are strong evidence of a link between obesity and cancer, the mechanisms have not been elucidated due to limitations in teasing obesity apart from its associated abnormalities and a lack of suitable pre-clinical models to study the obesity-linked cancer development. A vast majority of data supporting the obesity-cancer link come from observational studies, and experimental studies have only emerged in certain cancer types. With these challenges ahead, solutions are urgently needed not only to reduce obesity but also to understand its relationship with each cancer type and prevent a serious burden on the health care system in the coming years.
In this mini-review, we will summarize the most recent studies that examined the links between obesity and several common cancer types. In addition, we will discuss the limitations associated with mechanistic studies and the emerging rodent models that may help us better understand obesity-linked cancers. A detailed literature search on PubMed and Google Scholar was done for original research articles since 2012, reviews, and meta-analysis using the following search combinations: adipocyte/adipose tissue/obesity plus the cancer type. The references of selected publications were further searched for relevant information.

2. Cancers in the Digestive Tract and Accessory Organs

Central adiposity, composed of both visceral (omental, mesenteric, retroperitoneal) and subcutaneous fat depots, surrounds the digestive tract and its accessory organs including the liver, pancreas, and the gallbladder. These organs participate in the initial energy intake and processing that eventually lead to the accumulation of WAT. Central obesity together with diet and microbiota takes the blame for cancers of the abdominal cavity [9,10]. These three factors are often interconnected as high fat diet (HFD) can directly induce obesity or shift gut microbial composition to enhanced energy harvest that have both immune and metabolic consequences [11,12]. For example, HFD containing saturated fat and trans-fat promotes metastatic prostate cancer [13]; and, obesity-induced gut bacterial metabolites promote perturbed senescence phenotype and pro-inflammatory and tumor-promoting secretome in liver cancer [14]. Their interactions invariably lead to tissue-specific inflammation, including gastritis, hepatitis, pancreatitis and inflammatory bowel diseases, all of which propel their respective cancer types. Furthermore, men are more than twice as likely to develop cancers near the abdomen as compared to premenopausal women [15]. This advantage then becomes insignificant as weight “shifts” from pear-shape to apple-shape in postmenopausal women [16]. The difference in cancer susceptibility between genders has been well documented in cancer epidemiology [17,18]. Central adiposity, thus, may be a potential driving factor for sex disparity observed in cancers of this category. Here, we focus on obesity-linked gastric, liver and pancreatic cancers.

2.1. Gastric Cancer

Gastric cancer (GC) is an old-age and male-predominant cancer with late-stage diagnosis [19]. A recent meta-analysis of 24 prospective studies have revealed a positive correlation between patients with obesity and gastric cardia adenocarcinoma (GCA), cancer of the upper stomach and the most prevalent GC [20]. Increased intra-abdominal pressure, gastroesophageal reflux and altered levels of adipokines and cytokines (leptin, TNFα, IL-6 and IL-17) from obesity have been proposed as potential GCA promoting factors and reviewed in depth [21,22]. However, reason(s) behind obesity predilection for promoting GCA as opposed to cancer of the lower stomach (non-GCA) have not been elucidated.
In vitro studies examined interactions between human stomach cancer cell lines of the MKN series (MKN-7, MKN-28, MKN-45) and gastric subserosal adipose tissue stromal cells (ASCs), and demonstrated that ASCs promoted growth and invasiveness of GC cells while decreased cancer apoptosis through COX-2 independent MAPK activation in co-culture experiments [23]. Another study found ASCs also promoted GC cell growth through the chemotactic SDF-1/CXCR4 axis [24]. Leptin is the satiety hormone that positively correlates with adipose tissue mass, which is highly elevated in the plasma of most obese patients. One study found that leptin promoted GC cell migration through the RhoA/ROCK pathway and enhanced expression of intercellular adhesion molecule-1 (ICAM-1) [25]. Other studies suggested that leptin induced GC cell proliferation through JAK2/STAT3 and ERK pathways [26], which occurred after EGFR transactivation [27]. Of note, both adipocytes and gastric mucosa produce leptin in response to food intake, it is thus difficult to discern the autocrine and paracrine effects of leptin from different sources [28]. Co-cultures of isolated omental adipocytes with GC cells increased oleic acid uptake and enhanced invasiveness of GC cells, demonstrating a preference for lipid-rich fuels [29]. Other elevated adipocytokines such as IL-1β, IL-6, IL-17, and MCP-1 promote tumorigenesis by stimulating the Src/PI3K, MAPK, STAT3, and PKC pathways, some of which overlap with activated GC pathways mentioned earlier [30]. One study also looked at the synergic effect of infection and obesity and found that Helicobacter felis-infected mice fed on HFD accelerated gastric carcinogenesis through elevated immune response induced by adipose-derived IL-6 and leptin, and heightened pro-survival gene expression in gastric tissue through STAT3 signaling [31]. However, reason(s) behind obesity predilection for promoting GCA as opposed to cancer of the lower stomach (non-GCA) have not been elucidated. Available data have only demonstrated association between obesity and GC metastasis, while no research has examined its role in GC development.

2.2. Liver Cancer

In the United States, liver cancer has tripled in the last four decades with extremely low 5-year survival rate [32]. While the highest incidence remains in less developed nations, liver cancer is expected to rise in developed countries parallel to the obesity epidemic [33]. In addition to excess alcohol intake and viral hepatitis infection, obesity has become another contributing risk factor to hepatocellular carcinoma (HCC), the most common primary liver cancer [34,35]. A recent case-control study stratified by birth cohort, race/ethnicity, and sex reaffirmed previous findings that there is a positive correlation between high BMI and increased liver cancer risks [36]. In fact, HCC is the cancer type most enhanced by obesity through its tie with non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH) and type 2 diabetes mellitus (T2DM) [37]. Typically, obesity induces ectopic fat deposition in the liver and an imbalance of fatty acid accumulation and oxidation, resulting in steatosis. Chronic steatosis is a precursor for NASH that causes irreversible fibrosis and cirrhosis, which play significant role in HCC occurrence. These pathological processes have complex interactions, and their relative contribution at various stages in the development of liver cancer remains to be determined.
In cell culture studies, hypertrophied adipocytes, free fatty acids (FFAs) and adipocytokines released from the adipose tissue have all been shown to increase liver pathology. Co-culture study between hepatocytes and adipose tissue explants found more pronounced cytotoxic response and insulin resistance in hepatocytes co-cultured with inguinal compared to epididymal depots, signifying depot-specific effect [38]. Several researches have demonstrated that FFA treatment induced lipotoxicity in cultured hepatocytes, such as enhanced expression of inflammatory markers (IL-1b, IL-6, IL-8, TNFα), increased reactive oxygen species and fibrogenic cytokine productions in a dose-dependent manner [39,40]. FFAs also reduced the activities of cytochromes P450 for drug metabolism and fibroblast growth factor 21 for glucose and lipid metabolism [41,42]. It is speculated that FFA-induced JNK activation together with hyperinsulinemia further enhances apoptosis of hepatocytes [43]. Besides adiponectin, most adipokines derived from the adipose tissue exert a negative effect on liver diseases [44,45]. Other less understood secretion such as the exosome-enclosed microRNA-27a has also been shown to promote liver cancer cell proliferation through FOXO1 downregulation and an increase in G1/S cell cycle transition [46]. In addition, the bidirectional communication allows exosomes from liver cancer cells to activate pro-inflammatory NF-κB signaling pathway and associated phospho-kinases (Akt, ERK1/2, and GSK3β) in adipocytes, contributing to a positive inflammatory feedback loop [47]. Another hepatic-derived protein, unconventional prefoldin RPB5 interactor (URI), regulated by the mTOR pathway, has proven crucial in the crosstalk between liver and WAT as its high expression under HFD induced both hepatic DNA damage and exacerbated adipose tissue lipolysis, immune infiltration and insulin resistance [44]. Still, more factors are being identified that keep fortifying the link between obesity and liver cancer.

2.3. Pancreatic Cancer

Pancreatic ductal adenocarcinoma (PDAC) is the major (~95%) exocrine pancreatic cancer and the most lethal form of all pancreatic cancers. Obesity and T2DM both have close association with PDAC [48]. Central adiposity and fatty infiltration in the pancreas parenchyma are risk factors for pancreatic precancerous lesions, and fatty infiltration is significantly higher in PDAC patients with high BMI [49]. Obesity also promotes chronic pancreatitis, another inducer of PDAC that increases its risk by tenfold [50,51]. Pancreatitis-induced tissue damage results in inflammatory response primarily mediated by macrophages and T cells during early stages of tumor development in PDAC models [52]. The mechanism by which inflammatory cells facilitate progression of pancreatic intraepithelial lesions into invasive PDAC tumors remains to be determined. In general, epidemiological studies do not report on the type of pancreatic cancers associated with obesity. Despite the relative prevalence of PDAC, it is arguable that obesity poses higher stress on endocrine β-cells in the pancreas. During obesity, there is an initial expansion in β-cell mass followed by defects in β-cell function resulting in insulin resistance. However, to the best of our knowledge, the link between obesity and malignancy in β-cells (insulinomas) has not been examined.
The crosstalk between adipocytes and PDAC cells has been evaluated in a number of studies [53,54]. Like many cancers in the abdominal cavity, PDAC preferentially metastasizes toward the omental adipose tissue. Based on Ingenuity Pathways Analysis, the human omental fat produces enriched extracellular molecules related to cellular growth, migration, invasion and chemoresistance, with which nine proteins (NGAL, FINC, ZA2G, PGS1, TIMP1, MSLN, IL-6, MMP8 and TSP1) have been reported to specifically promote pancreatic cancers [54]. In the same study, omental fat conditioned media treated-pancreatic cells and xenograft undergo profound cellular reprogramming, leading to a more aggressive tumor phenotype. Especially under nutrient-poor conditions, adipocytes were found to transfer increased glutamine to PDAC cells with the help of PDAC cell-induced glutaminase downregulation [55]. Okumura group found that genetically engineered mice with Pdx1, Kras or Trp53 mutations fed on HFD had significantly larger primary pancreatic tumors and higher distant organ metastasis; in vitro, FFAs from visceral adipose tissue-derived conditioned media also increased lipid droplets in the pancreatic cancer cells and enhanced cell invasiveness [56]. A recent study has emphasized that Kras mutation alone is not sufficiently oncogenic, and upstream stimuli are needed to amplify its activity in tumors [57]. In this context, pro-inflammatory and growth factors secreted under obesity provide the push for sustained Kras activation and its downstream NF-κB, COX2 and STAT3 signaling, which are corroborated by animal studies [58]. The impact of inflammatory milieu is supported by evidence that high intratumoral IL1-β and TNF-α were associated with poor response to neoadjuvant therapy, and high IL-8 and GM-CSF were predictive of tumor grade and size [59]. In particular, IL-1β facilitated a self-sustaining loop of crosstalk between adipocytes, tumor-associated neutrophils and fibrogenic pancreatic stellate cells that enhanced desmoplasia, limited vascular perfusion, impaired chemotherapeutics and accelerated tumor growth [60]. Interestingly, pancreas-associated adipose tissue contained more functional adipocytes and higher expressions of insulin receptor and adiponectin, which protected the pancreas against short-term lipotoxicity through its increased capacity for energy storage and reduced inflammation markers [61]. Still, animal models with early pancreatic tumor led to adipose tissue wasting and increased cancer survival, which are hypothesized to be driven by decreased pancreatic exocrine function [62].

3. Cancers in the Female Reproductive System

Obesity increases the risks of ovarian, endometrial and breast cancers [63]. Studies have shown that women with a high BMI have greater incidence of polycystic ovarian syndrome (PCOS), endometrial polyps and fibroids, all of which aggravate gynecological cancer occurrence [64,65,66]. PCOS is independently associated with insulin resistance and glucose intolerance, two conditions that are exacerbated by obesity [67]. It may also be an indirect link between obesity and increased risks of both endometrial and ovarian cancers in premenopausal women [68]. Most female dominant cancers have indisputable connections to the fluctuating estrogen levels throughout life. In postmenopausal women, estrogens are locally synthesized at peripheral sites, mainly the adipose tissue, and take over the loss of gonadal estrogens [69,70]. The ability of adipose tissue to convert androgens to estrogens through the action of aromatase contributes greatly to the estrogen-sensitive cancer types [71]. Moreover, obesity-linked meta-inflammation are found to be a major driver for reduced sex hormone-binding globulins [72], which may further increase the bioavailability of sex hormones for local action. Inheritable mutation of BRCA1 or BRCA2 gene has been associated with a lifetime risk of both ovarian and breast cancers. Ongoing study has reported that BRCA mutation carriers are more susceptible to DNA damage in the breast epithelial cells under an obese environment [73]. In this category, we focus on obesity-linked breast cancer for its high prevalence in women and for it being one of the best studied obesity-related cancers in experimental researches.

Breast Cancer

Breast cancer is the most common cancer in women, accounting for approximately one quarter (26%) of all new cancer cases and >12% cancer-related deaths in women worldwide [74]. Multiple studies have found that obese women with breast cancers have consistently poorer prognosis, larger tumors and lymph node metastasis [75,76,77,78,79]. The most common form develops from the lobules and ducts of the mammary gland surrounded largely by mature adipocytes and progenitors. While the relationship between local breast adipose tissue to metabolic risks and breast cancers has been conflicting and in need of standardization [80,81], it is apparent that the estrogen-rich microenvironment breast adipose tissue resides in and contributes to (through aromatase) plays a role in breast tumorigenesis [82]. Systemically, there is a strong link between obesity and breast cancer in postmenopausal women with inconsistent findings in premenopausal women [83,84,85]. A study on the Chinese population further incorporated the effect of adipose depots in breast cancer risk, showing premenopausal women with higher subcutaneous fat were more likely to develop hormone-receptor-positive breast cancer and postmenopausal women with higher visceral fat were more likely to develop hormone-receptor-negative breast cancer [86]. The vast heterogeneity in breast cancer etiology still remains elusive. In addition, it is worth noting that male breast cancer is on the rise with speculated connection to obesity and gynecomastia, the benign enlargement of the male breasts.
Increased production of estrogens promotes the development of breast cancer; together with progesterone, they are classified as the hormone-receptor positive subtypes. A study in postmenopausal women found that estradiol levels in breast adipose tissue were lower in breast cancer patients than in controls whereas serum concentrations did not differ, which pointed to the importance of tissue-specific estrogen regulation [87]. Animal and cell studies found that close proximity between adipocytes and breast cancer cells is required for tumor growth and their interaction depended on adipose tissue aromatase expression through leptin signaling [88]. Inflamed WAT has elevated pro-inflammatory mediators, enhanced aromatase expression and estrogen receptor-α (ER-α)-dependent gene expression [89]. Intriguingly, estradiol also promoted growth of estrogen receptor-negative cancer cells through the activation of ER-α in breast stroma, which prevented hypoxia and necrosis in grafted tumors and normalized tumor angiogenesis [90]. Despite the importance of estrogens, it has become evident that the estrogenic aspect of obesity alone cannot account for a large subgroup of obese women identified with ER-negative breast cancer [91]. In addition to estrogens, other molecules released from the adipose tissue such as insulin, IGF-I, inflammatory cytokines and adipokines also collectively promote breast cancer development and progression [92,93,94]. Analysis of triple-negative breast cancer models fed on HFD during metastatic progression showed metabolic perturbation in FA metabolism, olfactory transduction, mTOR signaling and autophagy [95]. Reciprocal metabolic interactions have been observed between adipocytes and breast cancer cells, which are reviewed previously [96,97,98,99]. Their crosstalk has been observed to change global gene expression pattern (upregulated pathways: TNF signaling, NF-κB signaling, senescence & autophagy and cytokines & inflammatory response; downregulated pathways: cell cycles) [100], interference with the anti-proliferative efficacy of tamoxifen [101], and antibody-dependent cytotoxicity of trastuzumab [102]. Whole-genome transcriptome microarrays and protein analyses revealed that obesity accelerated ER+ breast carcinogenesis and progression through increased PI3K/AKT/mTOR signaling and adipokine secretion [103]. When co-cultured with ASCs from obese models, breast cancer cells showed enhanced survival and radioresistance [104] in part through leptin signaling [105]. Production of paracrine cytokines from ASCs also stimulated breast cancer cell growth through upregulation of cancer cell calcium-binding protein, which promoted tumorigenesis [106]. CLSs, indicative of inflamed and necrotic adipocytes, were more frequently seen in breast adipose tissue of breast cancer patients and correlated with increased estrogen to androgen ratios in both breast adipose tissue and serum [107]. High CLS density in breast adipose tissue has now been shown to independently increase breast cancer risk [108]. Notably, the expression of fatty acid binding protein common to both adipocytes and macrophages (A-FABP) in tumor-associated macrophages facilitated breast cancer progression through IL-6/STAT3 signaling [109]. In particular, the prominent immune checkpoint molecule, programmed death-ligand 1 (PD-L1), was found to be markedly elevated in mature adipocytes, which may saturate anti-PD-L1 antibodies and prevent the intended function of T cell activation in immunotherapy [110]. In fact, current clinical success of cancer immunotherapy in breast cancers has been limited and could benefit from understanding of adipose-immune-tumor interactions [111].

4. Cancers in the Male Reproductive System

Low testosterone level in men correlates with increased central adiposity and reduced lean mass, energy imbalance and an overall impaired metabolism [112,113]. A decline in testosterone is attributed partly to a decrease in sex hormone-binding globulin associated with obesity and an increased aromatase conversion of estradiol in adipocytes [114,115]. Currently, the only male-specific cancer connected to obesity is prostate cancer. Despite controversies, low testosterone level is associated with a higher risk and worse outcomes for prostate cancer [116,117]. Several studies suggest an increased risk of aggressive prostate cancer in obese men, and possible links between obesity and prostate cancer have been reviewed in a dated publication [118]. In contrast, although obesity is shown to disrupt testicular morphology and spermatogenesis that cause infertility and induce chronic inflammation in the male reproductive system [119,120], no study has established a link between obesity and testicular cancer. For invasive penile cancer, obesity is included as a risk factor because of speculated and indirect mechanisms such as inflammation and obesity-induced buried penis [121,122,123]. For these andrological cancers, adipose tissue does not have direct contact and appears to facilitate primarily through ancillary inflammation, though further investigation is needed. In this category, we will survey the most recent findings on obesity-linked prostate cancer.

Prostate Cancer

The most common prostate cancer (PC) termed acinar adenocarcinoma develops from glandular cells surrounding the prostate. Current treatment for advanced PC is based on androgen deprivation to which tumor cells initially respond to, but eventually they become castrate-resistant by acquiring changes such as androgen-receptor overexpression [124]. Men with metastatic castrate-resistant PC have a median survival of less than 2 years and no standard therapeutic modality [125]. While only limited evidence has found a link between obesity and PC, their overlapping trend among older men suggests the likelihood that PC will continue to grow in light of the obesity pandemic. The pathophysiological and biological mechanisms underlying these associations are only beginning to be understood [126].
Periprostatic adipose tissue inflammation is commonly observed in PC patients [127]. Gene expression signature of periprostatic WAT in obese and overweight patients exhibited hypercellularity and reduced immunosurveillance [128]. Chemokine C-C motif ligand 7 (CCL7) produced in periprostatic WAT was found to stimulate the migration of chemokine receptor type 3-expressing tumor cells [129]. Multiple reviews proposed that obesity-induced inflammation is a major culprit in PC [130,131]. Within adipocytes, loss of p62 (a ubiquitin-binding protein in autophagy) shut-downed mTOC1-mediated FA metabolism, which increased FA availability for PC cells [132]. Interestingly, adipocyte specific p62-deficient mice developed obesity independent of diet and displayed enhanced synthesis/secretion of posteopontin from adipocytes and carnitine palmitoyltransferase Ia from tumor cells, both of which are predictors of poor PC prognosis [132]. PC exosomes also assisted adipocyte lipolysis through the p38 and ERK pathways and hormone-sensitive lipase activation, which were proposed as mechanisms to enhance pancreatic local invasiveness and metastasis [56,133]. Paracrine molecules from adipocytes were found to stimulate the non-canonical WNT signaling pathway in PC cells, leading to nuclear translocation of genes regulating epithelial-to-mesenchymal transition and cancer aggressiveness [134]. In addition, the periprostatic WAT also showed increased matrix metalloproteinase activity, which is essential in extracellular matrix remodeling that promotes PC cell proliferation and motility [135]. On the other hand, obesity is associated with decreased concentration of testosterone, which reduces the risk of hormone-sensitive PC but paradoxically increases the risk of aggressive PC [136]. Androgens can act both as a stimulator of lipolysis and regulator of several inflammatory genes [137,138]. The role of androgens in obesity and PC has been well established and independently studied, but their potential interplay has yet to be determined.

5. Current Approaches to Study Obesity-Linked Cancers and Their Limitations

The most difficult question in obesity-linked cancer research is to identify whether obesity independently increases the chance of cancer without its associated conditions. In vitro studies often examine interaction between adipocytes and cancer cells of interest using co-culture experiment or treatment with conditioned media from adipocytes. Emerging multicellular 3D models also more closely mimic the tumor microenvironment and its behavior. Direct cell-cell communication is vital in cancer proliferation and metastasis, but systemic interactions and hormone regulations from the adipose tissue require analysis in whole organism. In vivo approach combines oncogenic model [139] with obese model [140] in order to replicate obesity-linked cancer development and progression in humans (Figure 2). These combined models have provided valuable information on the effect of obesity on tumor progression as exemplified by some high impact studies from the previous sections. However, they fell short in a number of ways in elucidating the link between obesity and the onset of cancer. First, oncogenic models develop tumors regardless of an obese background. Most combined models observed an exacerbated tumor phenotype under an obese environment signifying aggressive progression of existing tumor but could not address whether obesity initiates tumorigenesis spontaneously. Second, combine models introduce confounding effects of diet, leptin deficiency and carcinogens in addition to obesity on tumor development/progression. For example, in DIO, the effect of HFD itself may be an independent risk factor for cancer. Other obese models such as the ob/ob and db/db, have a defect in leptin or the leptin receptor, which prevent leptin regulation on obesity-associated signaling pathways in cancer. Under an obese state in breast cancer, leptin interacts with molecular effectors such has estrogen, insulin, IGF-1 and inflammatory cytokines, which impacts various stages of breast cancer from initiation to metastatic progression [141]. Transplanted and carcinogen-induced tumors occur after obesity is established, and again do not replicate cancers that develop from a progressive and systemic change due to obesity. Although statistical techniques to adjust for confounding variables can improve epidemiological comparisons, there is still a lack of translational model to study the relationship between obesity, insulin resistance and tumor development, as well as the relative contribution of obesity-associated abnormalities to cancers. To the best of our knowledge, none of the combination model that pairs obesity to cancer develops obesity- or diabetes-linked cancer spontaneously and sequentially like those found in humans.

5.1. Tsumura Suziki Obese Diabetes (TSOD) Mouse Model

The male TSOD mouse model has been reported to spontaneously develop obesity-associated diabetes, NASH, and HCC by 10 months without additional gene manipulation [142,143]. The phenotypes of this inbred ddY strain have been attributed to three quantitative trait loci involved in blood glucose level and body weight: non-insulin-dependent diabetes (Nidd) 4 on mouse chromosome 11, Nidd5 on mouse chromosome 2, and Nidd6 on mouse chromosome 1 [144]. However, the gene(s) responsible for the observed hepatic tumorigenesis is currently unknown. At a young age (about 5 weeks), TSOD mice start gaining weight in both the visceral and subcutaneous adipose tissues, and develop high plasma triglyceride and cholesterol levels accompanied by oxidative stress marker, but not hyperglycemia [142,145]. Adipocyte hypertrophy, pro-inflammatory macrophage-induced CLSs and CD8-positive lymphoid aggregation were observed at 4 months, together with ballooning and microvesicular steatosis in the liver [146]. Multi-organ changes at advanced age were noticed, including severe hypertrophy of pancreatic islets due to B cell infiltration, thickening of the basement membrane in glomeruli, and motor and sensory neuropathies, closely mimicking the diabetic complications [147]. In TSOD mice that developed HCC, a high frequency of glutamine synthase expression and overloading of intrahepatic bile acids were observed, which resemble HCC in humans [143,148,149]. Of note, TSOD mice develop obesity due to disrupted control of hypothalamic-mediated hyperphagia that persist throughout diabetes and HCC, making it difficult to discern the effect of obesity on HCC from coexisting diet and diabetes [150]. Furthermore, TSOD mice also have both low sympathetic and adrenomedullary activities with high adrenocortical activity starting 4 months of age, which affect central energy regulation and hormone secretion and add extra linkages to HCC [151]. Since its creation, the TSOD model was only established and characterized in males; selective breeding in obese females also led to significant weight increase but showed no sign of urinary glucose [152]. This phenomenon is not unique to TSOD mice and has been reported in other polygenic mouse models [153]. It would be thus interesting to follow up on the male sex bias in metabolic syndrome severity. Nonetheless, the TSOD mouse model represents a promising tool to study the obesity-T2DM-NASH-HCC axis.

5.2. Mito-Ob and mMito-Ob Mouse Models

Irrespective of etiology, obesity is characterized by an increase in adipose tissue mass that requires corresponding changes in adipocytes, the structural and functional units of adipose tissue. Through adipocyte remodeling, a transgenic mouse model, Mito-Ob, developed obesity through an upregulation of prohibitin (PHB) under the adipocyte protein 2 (aP2, also known as Fabp4) promoter. PHB is a highly conserved and pleiotropic protein found in multiple subcellular compartments such as the mitochondria, cytosol and nucleus. Its predominant role in many cells has been in maintaining mitochondrial homeostasis and biogenesis, though it is also intricately involved in insulin sensitivity, proliferation and immune regulation in part through phosphorylation at tyrosine (Y)-114 [154,155]. Longitudinal studies on these mice uncovered obesity-related metabolic disruptions and a spontaneous development of HCC specifically in males [156,157]. A mutant (m)Mito-Ob mouse model that overexpresses a phospho-mutant PHB (Y114F) under the aP2 promoter also developed obesity and diabetic symptoms, but instead manifested lymphoproliferative disease (lymph node tumors) or adult onset autoimmune diabetes when fed on HFD [158]. It appears that mutation at a phosphorylation site in the adipose tissue-specific PHB gene altered the course of cancer development and produced multiple cancer types under the same obese background. Of note, since aP2 gene is expressed in both adipocytes and monocytic immune cells (macrophages and dendritic cells), PHB expression may be altered in both cell types. The distinct cancer types in Mito-Ob and mMito-Ob mice imply that PHB Y114F mutation may have activated an immune dysregulation mediated by enhanced PI3K-Akt signaling in monocytic cells, which increased the likelihood of immune-base complications such as the observed lymph node tumor and autoimmune diabetes [155]. These results suggest tissue- and compartment-specific functions of PHB, which are known in the literature [159]. It is speculated that PHB over-expression in adipocytes was sufficient to induce obesity, while Y11F mutant PHB over-expression preferentially affected monocytic cells. Research is currently underway to extrapolate the puzzling difference in mechanisms between the metabolic and immune functions of PHB. Both Mito-Ob and mMito-Ob mice have shown spontaneous development of cancers from an obesity background independent of diet and carcinogens, which may serve as a model to study obesity-cancer linkage. Furthermore, the metabolic dysregulations in female Mito-Ob mice appeared after ovariectomy, suggesting involvement of estrogens in regulating PHB [158]. These findings are consistent with recent WHO report proposing possible roles of sex steroids and chronic inflammation in obesity-linked cancers [5]. Thus, research going forward should be mindful of sex difference in the context of obesity and the development of all obesity-linked cancers.

6. Conclusions and Future Perspective

This review highlights the current knowledge on obesity-linked cancers, their speculated mechanisms and rodent models used to study the link. Now, 13 cancers have been confirmed to have an increased risk in obesity. It is likely that more cancer types will be added to the list given the systemic influence obesity and its complications have. To top it off, childhood and infant obesity are also on the rise, both of which are strong predictors of adult obesity [160,161,162]. While no evidence has linked obesity to any pediatric cancers, recent report has suggested that obesity accelerate cancer occurrence at a younger age [163]. Survivors of childhood cancer are also at increased risks for obesity, T2DM and cancers in adulthood [164]. In many cases, obesity-related abnormalities are the main facilitators of cancer development, not obesity per se. For example, the A-Zip/F1 mouse model, which has virtually no WAT, showed high serum glucose, insulin, IGF-1 and pro-inflammatory cytokine levels, and accelerated tumor formation [165]. Human studies have also identified a subset of up to 25% of obese individuals that are metabolically healthy, but little is known about their long-term metabolic outcome [166]. On the contrary, the metabolic syndrome has also been observed in lean individuals, pointing to the likelihood that obesity itself may not be sufficient for disrupted metabolism [167]. It would be interesting to extrapolate the differences between metabolically healthy obese and metabolically unhealthy lean individuals. In fact, the role of obesity-associated complications in cancer development has been strongly supported by the use of Metformin (an antidiabetic, glucose-lowering drug) and Aspirin (anti-inflammatory drug) in cancer prevention and treatment [168,169]. Obesity and related metabolic changes can be reversed through interventions, but whether obesity-induced cancer can be reversed is unknown. It raises the concern that losing weight in chronically obese patients may not be sufficient to reduce the risk of cancer development by significant amount. While obesity has been confirmed as an independent risk factor for cancers, it is extremely difficult to discern between obesity and its comorbidities in driving cancer. In this regard, monitoring the progression and metabolic profile at different stages of obesity (as proposed by the Edmonton Obesity Staging System [170]) may be useful in assessing obesity-related risks and prioritizing treatment. In any obesity-linked cancer, it is likely that dormant mutations are activated locally by inflamed adipose tissue or by secreted endocrine molecules at a distance. The proximity effect of obesity heavily depends on body adipose tissue depot distribution, composition and physiology to which tumorigenesis requires. The synergic effect of other carcinogenic factors such as diet in gastric cancer, alcohol in liver cancer, menopause in gynecological cancers and infections in general can further promote cancer of a particular type. Additionally, epigenetic modifications from obesity that are oncogenic cannot be ruled out. Studies on genetically identical mice and monozygotic twins found that changes in a chromatin-interacting protein, Trim28, resulted in an epigenetic binary switch (produced by a group of imprinted genes) of either a lean or obese phenotype [171]. Another study directly linked epigenetic changes during obesity to histone modifications and DNA methylation in the colonic epithelium, which preceded a tumor-prone gene signature that is reversible through weight loss [172,173]. Much of these findings still require rigorous follow-ups. Research carrying forward should build upon available pre-clinical models to examine the relationship between excess adiposity and the development and progression of individual cancer, while bearing in mind the need to distinguish potential comorbidities and advance our understanding for preventative and therapeutic strategies.

Author Contributions

Y.X.Z.X. and S.M. contributed to the literature reviews and writing of the manuscript.

Funding

S.M. is supported by funds from Natural Sciences and Engineering Research Council (RGPIN-2017-04962), Research Manitoba, and Health Sciences Centre Foundation. Y.X.Z.X. is a recipient of the University of Manitoba Graduate Fellowship (UMGF) and Alexander Graham Bell Canada Graduate Fellowship Doctoral (CGF D) from NSERC.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Arnold, M.; Pandeya, N.; Byrnes, G.; Renehan, P.A.G.; Stevens, G.A.; Ezzati, P.M.; Ferlay, J.; Miranda, J.J.; Romieu, I.; Dikshit, R.; et al. Global burden of cancer attributable to high body-mass index in 2012: A population-based study. Lancet Oncol. 2015, 16, 36–46. [Google Scholar] [CrossRef]
  2. Vos, T.; Abajobir, A.A.; Abate, K.H.; Abbafati, C.; Abbas, K.M.; Abd-Allah, F.; Abdulkader, R.S.; Abdulle, A.M.; Abebo, T.A.; Abera, S.F.; et al. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Lond. Engl. 2017, 390, 1211–1259. [Google Scholar] [CrossRef]
  3. Frayn, K.N. Obesity and metabolic disease: Is adipose tissue the culprit? Proc. Nutr. Soc. 2005, 64, 7–13. [Google Scholar] [CrossRef] [PubMed]
  4. Basen-Engquist, K.; Chang, M. Obesity and Cancer Risk: Recent Review and Evidence. Curr. Oncol. Rep. 2011, 13, 71–76. [Google Scholar] [CrossRef] [PubMed]
  5. 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] [PubMed]
  6. Steele, C.B.; Thomas, C.C.; Henley, S.J.; Massetti, G.M.; Galuska, D.A.; Agurs-Collins, T.; Puckett, M.; Richardson, L.C. Vital Signs: Trends in Incidence of Cancers Associated with Overweight and Obesity—United States, 2005–2014. MMWR Morbidity Mortality Wkly. Rep. 2017, 66, 1052–1058. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, Y.C.; McPherson, K.; Marsh, T.; Gortmaker, S.L.; Brown, M. Health and economic burden of the projected obesity trends in the USA and the UK. Lancet 2011, 378, 815–825. [Google Scholar] [CrossRef]
  8. Office of the Surgeon General (US); Office of Disease Prevention and Health Promotion (US); Centers for Disease Control and Prevention (US); National Institutes of Health (US). The Surgeon General’s Call To Action To Prevent and Decrease Overweight and Obesity; Publications and Reports of the Surgeon General; Office of the Surgeon General (US): Rockville, MD, USA, 2001.
  9. Castaner, O.; Goday, A.; Park, Y.-M.; Lee, S.-H.; Magkos, F.; Shiow, S.-A.T.E.; Schröder, H. The Gut Microbiome Profile in Obesity: A Systematic Review. Int. J. Endocrinol. 2018, 2018. [Google Scholar] [CrossRef] [PubMed]
  10. Pandey, A.; Patel, K.V.; Lavie, C.J. Obesity, Central Adiposity, and Fitness: Understanding the Obesity Paradox in the Context of Other Cardiometabolic Parameters. Mayo Clin. Proc. 2018, 93, 676–678. [Google Scholar] [CrossRef] [PubMed]
  11. Singh, R.K.; Chang, H.-W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [PubMed]
  12. Dalby, M.J.; Ross, A.W.; Walker, A.W.; Morgan, P.J. Dietary Uncoupling of Gut Microbiota and Energy Harvesting from Obesity and Glucose Tolerance in Mice. Cell Rep. 2017, 21, 1521–1533. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, M.; Zhang, J.; Sampieri, K.; Clohessy, J.G.; Mendez, L.; Gonzalez-Billalabeitia, E.; Liu, X.-S.; Lee, Y.-R.; Fung, J.; Katon, J.M.; et al. An aberrant SREBP-dependent lipogenic program promotes metastatic prostate cancer. Nat. Genet. 2018, 50, 206–218. [Google Scholar] [CrossRef] [PubMed]
  14. Yoshimoto, S.; Loo, T.M.; Atarashi, K.; Kanda, H.; Sato, S.; Oyadomari, S.; Iwakura, Y.; Oshima, K.; Morita, H.; Hattori, M.; et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013, 499, 97–101. [Google Scholar] [CrossRef] [PubMed]
  15. Valencak, T.G.; Osterrieder, A.; Schulz, T.J. Sex matters: The effects of biological sex on adipose tissue biology and energy metabolism. Redox Biol. 2017, 12, 806–813. [Google Scholar] [CrossRef] [PubMed]
  16. Lovejoy, J.; Champagne, C.; de Jonge, L.; Xie, H.; Smith, S. Increased visceral fat and decreased energy expenditure during the menopausal transition. Int. J. Obes. 2008, 32, 949–958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Dorak, M.T.; Karpuzoglu, E. Gender differences in cancer susceptibility: An inadequately addressed issue. Front. Genet. 2012, 3, 268. [Google Scholar] [CrossRef] [PubMed]
  18. Yuan, Y.; Liu, L.; Chen, H.; Wang, Y.; Xu, Y.; Mao, H.; Li, J.; Mills, G.B.; Shu, Y.; Li, L.; et al. Comprehensive Characterization of Molecular Differences in Cancer between Male and Female Patients. Cancer Cell 2016, 29, 711–722. [Google Scholar] [CrossRef] [PubMed]
  19. Stomach Cancer Risk Factors. Available online: https://www.cancer.org/cancer/stomach-cancer/causes-risks-prevention/risk-factors.html (accessed on 10 December 2018).
  20. Chen, Y.; Liu, L.; Wang, X.; Wang, J.; Yan, Z.; Cheng, J.; Gong, G.; Li, G. Body Mass Index and Risk of Gastric Cancer: A Meta-analysis of a Population with More Than Ten Million from 24 Prospective Studies. Cancer Epidemiol. Prev. Biomark. 2013, 22, 1395–1408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Li, Q.; Zhang, J.; Zhou, Y.; Qiao, L. Obesity and gastric cancer. Front. Biosci. Landmark Ed. 2012, 17, 2383–2390. [Google Scholar] [CrossRef] [PubMed]
  22. Olefson, S.; Moss, S.F. Obesity and related risk factors in gastric cardia adenocarcinoma. Gastric Cancer 2015, 18, 23–32. [Google Scholar] [CrossRef] [PubMed]
  23. Nomoto-Kojima, N.; Aoki, S.; Uchihashi, K.; Matsunobu, A.; Koike, E.; Ootani, A.; Yonemitsu, N.; Fujimoto, K.; Toda, S. Interaction between adipose tissue stromal cells and gastric cancer cells in vitro. Cell Tissue Res. 2011, 344, 287–298. [Google Scholar] [CrossRef] [PubMed]
  24. Zhao, B.-C.; Zhao, B.; Han, J.-G.; Ma, H.-C.; Wang, Z.-J. Adipose-derived stem cells promote gastric cancer cell growth, migration and invasion through SDF-1/CXCR4 axis. Hepatogastroenterology 2010, 57, 1382–1389. [Google Scholar] [PubMed]
  25. Dong, Z.; Fu, S.; Xu, X.; Yang, Y.; Du, L.; Li, W.; Kan, S.; Li, Z.; Zhang, X.; Wang, L.; et al. Leptin-mediated regulation of ICAM-1 is Rho/ROCK dependent and enhances gastric cancer cell migration. Br. J. Cancer 2014, 110, 1801–1810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Pai, R.; Lin, C.; Tran, T.; Tarnawski, A. Leptin activates STAT and ERK2 pathways and induces gastric cancer cell proliferation. Biochem. Biophys. Res. Commun. 2005, 331, 984–992. [Google Scholar] [CrossRef] [PubMed]
  27. Shida, D.; Kitayama, J.; Mori, K.; Watanabe, T.; Nagawa, H. Transactivation of Epidermal Growth Factor Receptor Is Involved in Leptin-Induced Activation of Janus-Activated Kinase 2 and Extracellular Signal–Regulated Kinase 1/2 in Human Gastric Cancer Cells. Cancer Res. 2005, 65, 9159–9163. [Google Scholar] [CrossRef] [PubMed]
  28. Cammisotto, P.G.; Levy, É.; Bukowiecki, L.J.; Bendayan, M. Cross-talk between adipose and gastric leptins for the control of food intake and energy metabolism. Prog. Histochem. Cytochem. 2010, 45, 143–200. [Google Scholar] [CrossRef] [PubMed]
  29. 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] [PubMed]
  30. Gislette, T.; Chen, J. The Possible Role of IL-17 in Obesity-Associated Cancer. Available online: https://www.hindawi.com/journals/tswj/2010/982036/abs/ (accessed on 10 December 2018).
  31. Ericksen, R.E.; Rose, S.; Westphalen, C.B.; Shibata, W.; Muthupalani, S.; Tailor, Y.; Friedman, R.A.; Han, W.; Fox, J.G.; Ferrante, A.W.; et al. Obesity accelerates Helicobacter felis-induced gastric carcinogenesis by enhancing immature myeloid cell trafficking and TH17 response. Gut 2014, 63, 385–394. [Google Scholar] [CrossRef] [PubMed]
  32. Liver Cancer Risk Factors. Available online: https://www.cancer.org/cancer/liver-cancer/causes-risks-prevention/risk-factors.html (accessed on 10 December 2018).
  33. Aleksandrova, K.; Stelmach-Mardas, M.; Schlesinger, S. Obesity and Liver Cancer. Recent Res. Cancer Res. Fortsch. Krebsforschung Progres Dans Rech. Cancer 2016, 208, 177–198. [Google Scholar]
  34. El-Serag, H.B. Hepatocellular carcinoma. N. Engl. J. Med. 2011, 365, 1118–1127. [Google Scholar] [CrossRef] [PubMed]
  35. Turati, F.; Talamini, R.; Pelucchi, C.; Polesel, J.; Franceschi, S.; Crispo, A.; Izzo, F.; La Vecchia, C.; Boffetta, P.; Montella, M. Metabolic syndrome and hepatocellular carcinoma risk. Br. J. Cancer 2013, 108, 222–228. [Google Scholar] [CrossRef] [PubMed]
  36. Petrick, J.L.; Freedman, N.D.; Demuth, J.; Yang, B.; Van Den Eeden, S.K.; Engel, L.S.; McGlynn, K.A. Obesity, diabetes, serum glucose, and risk of primary liver cancer by birth cohort, race/ethnicity, and sex: Multiphasic health checkup study. Cancer Epidemiol. 2016, 42, 140–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Tilg, H.; Moschen, A.R. Mechanisms behind the link between obesity and gastrointestinal cancers. Best Pract. Res. Clin. Gastroenterol. 2014, 28, 599–610. [Google Scholar] [CrossRef] [PubMed]
  38. Du, Z.-Y.; Ma, T.; Lock, E.-J.; Hao, Q.; Kristiansen, K.; Frøyland, L.; Madsen, L. Depot-Dependent Effects of Adipose Tissue Explants on Co-Cultured Hepatocytes. PLoS ONE 2011, 6, e20917. [Google Scholar] [CrossRef] [PubMed]
  39. Nov, O.; Kohl, A.; Lewis, E.C.; Bashan, N.; Dvir, I.; Ben-Shlomo, S.; Fishman, S.; Wueest, S.; Konrad, D.; Rudich, A. Interleukin-1β May Mediate Insulin Resistance in Liver-Derived Cells in Response to Adipocyte Inflammation. Endocrinology 2010, 151, 4247–4256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Yao, H.-R.; Liu, J.; Plumeri, D.; Cao, Y.-B.; He, T.; Lin, L.; Li, Y.; Jiang, Y.-Y.; Li, J.; Shang, J. Lipotoxicity in HepG2 cells triggered by free fatty acids. Am. J. Transl. Res. 2011, 3, 284–291. [Google Scholar] [PubMed]
  41. Asrih, M.; Montessuit, C.; Philippe, J.; Jornayvaz, F.R. Free Fatty Acids Impair FGF21 Action in HepG2 Cells. Cell. Physiol. Biochem. 2015, 37, 1767–1778. [Google Scholar] [CrossRef] [PubMed]
  42. Donato, M.T. Potential Impact of Steatosis on Cytochrome P450 Enzymes of Human Hepatocytes Isolated from Fatty Liver Grafts. Drug Metab. Dispos. 2006, 34, 1556–1562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Malhi, H.; Bronk, S.F.; Werneburg, N.W.; Gores, G.J. Free Fatty Acids Induce JNK-dependent Hepatocyte Lipoapoptosis. J. Biol. Chem. 2006, 281, 12093–12101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Roberts, A.A.; Hebbard, L.W. Molecular cross-talk between the liver and white adipose tissue links excessive noURIshment to hepatocellular carcinoma. Transl. Cancer Res. 2016, 5, S1222–S1226. [Google Scholar] [CrossRef]
  45. Buechler, C.; Haberl, E.M.; Rein-Fischboeck, L.; Aslanidis, C. Adipokines in Liver Cirrhosis. Int. J. Mol. Sci. 2017, 18, 1392. [Google Scholar] [CrossRef] [PubMed]
  46. Sun, B.; Li, J.; Shao, D.; Pan, Y.; Chen, Y.; Li, S.; Yao, X.; Li, H.; Liu, W.; Zhang, M.; et al. Adipose tissue-secreted miR-27a promotes liver cancer by targeting FOXO1 in obese individuals. OncoTargets Ther. 2015, 8, 735–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Wang, S.; Xu, M.; Li, X.; Su, X.; Xiao, X.; Keating, A.; Zhao, R.C. Exosomes released by hepatocarcinoma cells endow adipocytes with tumor-promoting properties. J. Hematol. Oncol. 2018, 11, 82. [Google Scholar] [CrossRef] [PubMed]
  48. Andersen, D.K.; Korc, M.; Petersen, G.M.; Eibl, G.; Li, D.; Rickels, M.R.; Chari, S.T.; Abbruzzese, J.L. Diabetes, Pancreatogenic Diabetes, and Pancreatic Cancer. Diabetes 2017, 66, 1103–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Rebours, V.; Gaujoux, S.; d’Assignies, G.; Sauvanet, A.; Ruszniewski, P.; Levy, P.; Paradis, V.; Bedossa, P.; Couvelard, A. Obesity and Fatty Pancreatic Infiltration Are Risk Factors for Pancreatic Precancerous Lesions (PanIN). Clin. Cancer Res. 2015, 21, 3522–3528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Malka, D.; Hammel, P.; Maire, F.; Rufat, P.; Madeira, I.; Pessione, F.; Lévy, P.; Ruszniewski, P. Risk of pancreatic adenocarcinoma in chronic pancreatitis. Gut 2002, 51, 849–852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Lowenfels, A.B.; Maisonneuve, P.; Cavallini, G.; Ammann, R.W.; Lankisch, P.G.; Andersen, J.R.; Dimagno, E.P.; Andren-Sandberg, A.; Domellof, L.; the International Pancreatitis Study Group. Pancreatitis and the Risk of Pancreatic Cancer. N. Engl. J. Med. 1993, 328, 1433–1437. [Google Scholar] [CrossRef] [PubMed]
  52. Guerra, C.; Barbacid, M. Genetically engineered mouse models of pancreatic adenocarcinoma. Mol. Oncol. 2013, 7, 232–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. White, P.B.; True, E.M.; Ziegler, K.M.; Wang, S.S.; Swartz-Basile, D.A.; Pitt, H.A.; Zyromski, N.J. Insulin, leptin, and tumoral adipocytes promote murine pancreatic cancer growth. J. Gastrointest. Surg. 2010, 14, 1888–1894. [Google Scholar] [CrossRef] [PubMed]
  54. Feygenzon, V.; Loewenstein, S.; Lubezky, N.; Pasmanic-Chor, M.; Sher, O.; Klausner, J.M.; Lahat, G. Unique cellular interactions between pancreatic cancer cells and the omentum. PLoS ONE 2017, 12, e0179862. [Google Scholar] [CrossRef] [PubMed]
  55. Meyer, K.A.; Neeley, C.K.; Baker, N.A.; Washabaugh, A.R.; Flesher, C.G.; Nelson, B.S.; Frankel, T.L.; Lumeng, C.N.; Lyssiotis, C.A.; Wynn, M.L.; et al. Adipocytes promote pancreatic cancer cell proliferation via glutamine transfer. Biochem. Biophys. Rep. 2016, 7, 144–149. [Google Scholar] [CrossRef] [PubMed]
  56. Okumura, T.; Ohuchida, K.; Sada, M.; Abe, T.; Endo, S.; Koikawa, K.; Iwamoto, C.; Miura, D.; Mizuuchi, Y.; Moriyama, T.; et al. Extra-pancreatic invasion induces lipolytic and fibrotic changes in the adipose microenvironment, with released fatty acids enhancing the invasiveness of pancreatic cancer cells. Oncotarget 2017, 8, 18280–18295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Huang, H.; Daniluk, J.; Liu, Y.; Chu, J.; Li, Z.; Ji, B.; Logsdon, C.D. Oncogenic K-Ras requires activation for enhanced activity. Oncogene 2014, 33, 532–535. [Google Scholar] [CrossRef] [PubMed]
  58. Philip, B.; Roland, C.L.; Daniluk, J.; Liu, Y.; Chatterjee, D.; Gomez, S.B.; Ji, B.; Huang, H.; Wang, H.; Fleming, J.B.; et al. A High-Fat Diet Activates Oncogenic Kras and COX2 to Induce Development of Pancreatic Ductal Adenocarcinoma in Mice. Gastroenterology 2013, 145, 1449–1458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Delitto, D.; Black, B.S.; Sorenson, H.L.; Knowlton, A.E.; Thomas, R.M.; Sarosi, G.A.; Moldawer, L.L.; Behrns, K.E.; Liu, C.; George, T.J.; et al. The inflammatory milieu within the pancreatic cancer microenvironment correlates with clinicopathologic parameters, chemoresistance and survival. BMC Cancer 2015, 15, 783. [Google Scholar] [CrossRef] [PubMed]
  60. Incio, J.; Liu, H.; Suboj, P.; Chin, S.M.; Chen, I.X.; Pinter, M.; Ng, M.R.; Nia, H.T.; Grahovac, J.; Kao, S.; et al. Obesity-induced inflammation and desmoplasia promote pancreatic cancer progression and resistance to chemotherapy. Cancer Discov. 2016. [Google Scholar] [CrossRef] [PubMed]
  61. Garcia, B.C.; Wu, Y.; Nordbeck, E.B.; Musovic, S.; Olofsson, C.S.; Rorsman, P.; Wernstedt, A. Pancreatic Adipose Tissue in Diet-Induced Type 2 Diabetes. Diabetes 2018, 67 (Suppl. 1). [Google Scholar] [CrossRef]
  62. Danai, L.V.; Babic, A.; Rosenthal, M.H.; Dennstedt, E.A.; Muir, A.; Lien, E.C.; Mayers, J.R.; Tai, K.; Lau, A.N.; Jones-Sali, P.; et al. Altered exocrine function can drive adipose wasting in early pancreatic cancer. Nature 2018, 558, 600–604. [Google Scholar] [CrossRef] [PubMed]
  63. Webb, P.M. Obesity and gynecologic cancer etiology and survival. Am. Soc. Clin. Oncol. Educ. Book Am. Soc. Clin. Oncol. Annu. Meet. 2013. [Google Scholar] [CrossRef] [PubMed]
  64. Carmina, E.; Lobo, R.A. Polycystic Ovary Syndrome (PCOS): Arguably the Most Common Endocrinopathy Is Associated with Significant Morbidity in Women. J. Clin. Endocrinol. Metab. 1999, 84, 1897–1899. [Google Scholar] [CrossRef] [PubMed]
  65. Serhat, E.; Cogendez, E.; Selcuk, S.; Asoglu, M.R.; Arioglu, P.F.; Eren, S. Is there a relationship between endometrial polyps and obesity, diabetes mellitus, hypertension? Arch. Gynecol. Obstet. 2014, 290, 937–941. [Google Scholar] [CrossRef] [PubMed]
  66. Sommer, E.M.; Balkwill, A.; Reeves, G.; Green, J.; Beral, D.V.; Coffey, K. Effects of obesity and hormone therapy on surgically-confirmed fibroids in postmenopausal women. Eur. J. Epidemiol. 2015, 30, 493–499. [Google Scholar] [CrossRef] [PubMed]
  67. Dunaif, A. Insulin resistance and the polycystic ovary syndrome: Mechanism and implications for pathogenesis. Endocr. Rev. 1997, 18, 774–800. [Google Scholar] [CrossRef] [PubMed]
  68. Yin, W.; Falconer, H.; Yin, L.; Xu, L.; Ye, W. Association Between Polycystic Ovary Syndrome and Cancer Risk. JAMA Oncol. 2018. [Google Scholar] [CrossRef] [PubMed]
  69. Bulun, S.E.; Zeitoun, K.; Sasano, H.; Simpson, E.R. Aromatase in aging women. Semin. Reprod. Endocrinol. 1999, 17, 349–358. [Google Scholar] [CrossRef] [PubMed]
  70. Simpson, E.R. Sources of estrogen and their importance. J. Steroid Biochem. Mol. Biol. 2003, 86, 225–230. [Google Scholar] [CrossRef]
  71. Cleary, M.P.; Grossmann, M.E. Obesity and Breast Cancer: The Estrogen Connection. Endocrinology 2009, 150, 2537–2542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Simó, R.; Sáez-López, C.; Barbosa-Desongles, A.; Hernández, C.; Selva, D.M. Novel insights in SHBG regulation and clinical implications. Trends Endocrinol. Metab. 2015, 26, 376–383. [Google Scholar] [CrossRef] [PubMed]
  73. In BRCA Mutation Carriers, Obesity is Linked with Increased DNA Damage in Normal Breast Gland Cells | Endocrine Society. Available online: https://www.endocrine.org/news-room/2018/in-brca-mutation-carriers-obesity-is-linked-with-increased-dna-damage-in-normal-breast-gland-cells (accessed on 10 December 2018).
  74. Canadian Cancer Statistics Publication—Canadian Cancer Society. Available online: http://www.cancer.ca/en/cancer-information/cancer-101/canadian-cancer-statistics-publication/?region=on (accessed on 10 December 2018).
  75. Daling, J.R.; Malone, K.E.; Doody, D.R.; Johnson, L.G.; Gralow, J.R.; Porter, P.L. Relation of body mass index to tumor markers and survival among young women with invasive ductal breast carcinoma. Cancer 2001, 92, 720–729. [Google Scholar] [CrossRef] [Green Version]
  76. De Azambuja, E.; McCaskill-Stevens, W.; Francis, P.; Quinaux, E.; Crown, J.P.A.; Vicente, M.; Giuliani, R.; Nordenskjöld, B.; Gutiérez, J.; Andersson, M.; et al. The effect of body mass index on overall and disease-free survival in node-positive breast cancer patients treated with docetaxel and doxorubicin-containing adjuvant chemotherapy: the experience of the BIG 02-98 trial. Breast Cancer Res. Treat. 2010, 119, 145–153. [Google Scholar] [CrossRef] [PubMed]
  77. Rasmussen, C.B.; Kjær, S.K.; Ejlertsen, B.; Andersson, M.; Jensen, M.-B.; Christensen, J.; Langballe, R.; Mellemkjær, L. Incidence of metachronous contralateral breast cancer in Denmark 1978–2009. Int. J. Epidemiol. 2014, 43, 1855–1864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Cantarero-Villanueva, I.; Galiano-Castillo, N.; Fernández-Lao, C.; Diaz-Rodríguez, L.; Fernández-Pérez, A.M.; Sánchez, M.J.; Arroyo-Morales, M. The influence of body mass index on survival in breast cancer patients. Clin. Breast Cancer 2015, 15, e117–e123. [Google Scholar] [CrossRef] [PubMed]
  79. Copson, E.R.; Cutress, R.I.; Maishman, T.; Eccles, B.K.; Gerty, S.; Stanton, L.; Altman, D.G.; Durcan, L.; Wong, C.; Simmonds, P.D.; et al. POSH Study Steering Group Obesity and the outcome of young breast cancer patients in the UK: The POSH study. Ann. Oncol. 2015, 26, 101–112. [Google Scholar] [CrossRef] [PubMed]
  80. Schautz, B.; Later, W.; Heller, M.; Müller, M.J.; Bosy-Westphal, A. Associations between breast adipose tissue, body fat distribution and cardiometabolic risk in women: Cross-sectional data and weight-loss intervention. Eur. J. Clin. Nutr. 2011, 65, 784–790. [Google Scholar] [CrossRef] [PubMed]
  81. Pettersson, A.; Tamimi, R.M. Breast fat and breast cancer. Breast Cancer Res. Treat. 2012, 135, 321–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Gérard, C.; Brown, K.A. Obesity and breast cancer—Role of estrogens and the molecular underpinnings of aromatase regulation in breast adipose tissue. Mol. Cell. Endocrinol. 2018, 466, 15–30. [Google Scholar] [CrossRef] [PubMed]
  83. Loi, S.; Milne, R.L.; Friedlander, M.L.; McCredie, M.R.E.; Giles, G.G.; Hopper, J.L.; Phillips, K.-A. Obesity and outcomes in premenopausal and postmenopausal breast cancer. Cancer Epidemiol. Biomark. Prev. 2005, 14, 1686–1691. [Google Scholar] [CrossRef] [PubMed]
  84. Dobbins, M.; Decorby, K.; Choi, B.C.K. The Association between Obesity and Cancer Risk: A Meta-Analysis of Observational Studies from 1985 to 2011. ISRN Prev. Med. 2013, 2013, 680536. [Google Scholar] [CrossRef] [PubMed]
  85. Chen, Y.; Liu, L.; Zhou, Q.; Imam, M.U.; Cai, J.; Wang, Y.; Qi, M.; Sun, P.; Ping, Z.; Fu, X. Body mass index had different effects on premenopausal and postmenopausal breast cancer risks: A dose-response meta-analysis with 3,318,796 subjects from 31 cohort studies. BMC Public Health 2017, 17, 936. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, F.; Liu, L.; Cui, S.; Tian, F.; Fan, Z.; Geng, C.; Cao, X.; Yang, Z.; Wang, X.; Liang, H.; et al. Distinct Effects of Body Mass Index and Waist/Hip Ratio on Risk of Breast Cancer by Joint Estrogen and Progestogen Receptor Status: Results from a Case-Control Study in Northern and Eastern China and Implications for Chemoprevention. Oncologist 2017, 22, 1431–1443. [Google Scholar] [CrossRef] [PubMed]
  87. Savolainen-Peltonen, H.; Vihma, V.; Leidenius, M.; Wang, F.; Turpeinen, U.; Hämäläinen, E.; Tikkanen, M.J.; Mikkola, T.S. Breast Adipose Tissue Estrogen Metabolism in Postmenopausal Women with or Without Breast Cancer. J. Clin. Endocrinol. Metab. 2014, 99, E2661–E2667. [Google Scholar] [CrossRef] [PubMed]
  88. Liu, E.; Samad, F.; Mueller, B.M. Local adipocytes enable estrogen-dependent breast cancer growth. Adipocyte 2013, 2, 165–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Iyengar, N.M.; Hudis, C.A.; Dannenberg, A.J. Obesity and inflammation: New insights into breast cancer development and progression. Am. Soc. Clin. Oncol. Educ. Book 2013, 46–51. [Google Scholar] [CrossRef] [PubMed]
  90. Péqueux, C.; Raymond-Letron, I.; Blacher, S.; Boudou, F.; Adlanmerini, M.; Fouque, M.-J.; Rochaix, P.; Noël, A.; Foidart, J.-M.; Krust, A.; et al. Stromal Estrogen Receptor-α Promotes Tumor Growth by Normalizing an Increased Angiogenesis. Cancer Res. 2012, 72, 3010–3019. [Google Scholar] [CrossRef] [PubMed]
  91. Maehle, B.O.; Tretli, S.; Thorsen, T. The associations of obesity, lymph node status and prognosis in breast cancer patients: Dependence on estrogen and progesterone receptor status. APMIS Acta Pathol. Microbiol. Immunol. Scand. 2004, 112, 349–357. [Google Scholar] [CrossRef] [PubMed]
  92. Rose, D.P.; Komninou, D.; Stephenson, G.D. Obesity, adipocytokines, and insulin resistance in breast cancer. Obes. Rev. 2004, 5, 153–165. [Google Scholar] [CrossRef] [PubMed]
  93. Hernandez, B.Y.; Wilkens, L.R.; LeMarchand, L.; Horio, D.; Chong, C.D.; Loo, L.W. Difference in IGF-axis protein expression and survival among multiethnic breast cancer patients. Cancer Med. 2015. [Google Scholar] [CrossRef] [PubMed]
  94. Madeddu, C.; Gramigano, G.; Floris, C.; Muenu, G.; Sollari, G.; Maccio, A. Role of inflammation and oxidative stress in post-menopausal oestrogen-dependent breast cancer. J. Cell. Mol. Med. 2014, 18, 2519–2529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. O’Flanagan, C.H.; Rossi, E.L.; McDonell, S.B.; Chen, X.; Tsai, Y.-H.; Parker, J.S.; Usary, J.; Perou, C.M.; Hursting, S.D. Metabolic reprogramming underlies metastatic potential in an obesity-responsive murine model of metastatic triple negative breast cancer. NPJ Breast Cancer. 2017, 3, 26. [Google Scholar] [CrossRef] [PubMed]
  96. Blücher, C.; Stadler, S.C. Obesity and Breast Cancer: Current Insights on the Role of Fatty Acids and Lipid Metabolism in Promoting Breast Cancer Growth and Progression. Front. Endocrinol. 2017, 8, 293. [Google Scholar] [CrossRef] [PubMed]
  97. Himbert, C.; Delphan, M.; Scherer, D.; Bowers, L.W.; Hursting, S.; Ulrich, C.M. Signals from the Adipose Microenvironment and the Obesity-Cancer Link—A Systematic Review. Cancer Prev. Res. 2017, 10, 494–506. [Google Scholar] [CrossRef] [PubMed]
  98. Hoy, A.J.; Balaban, S.; Saunders, D.N. Adipocyte–Tumor Cell Metabolic Crosstalk in Breast Cancer. Trends Mol. Med. 2017, 23, 381–392. [Google Scholar] [CrossRef] [PubMed]
  99. 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] [PubMed]
  100. Nickel, A.; Blücher, C.; Kadri, O.A.; Schwagarus, N.; Müller, S.; Schaab, M.; Thiery, J.; Burkhardt, R.; Stadler, S.C. Adipocytes induce distinct gene expression profiles in mammary tumor cells and enhance inflammatory signaling in invasive breast cancer cells. Sci. Rep. 2018, 8, 9482. [Google Scholar] [CrossRef] [PubMed]
  101. Bougaret, L.; Delort, L.; Billard, H.; Le Huede, C.; Boby, C.; De la Foye, A.; Rossary, A.; Mojallal, A.; Damour, O.; Auxenfans, C.; et al. Adipocyte/breast cancer cell crosstalk in obesity interferes with the anti-proliferative efficacy of tamoxifen. PLoS ONE 2018, 13. [Google Scholar] [CrossRef] [PubMed]
  102. Duong, M.N.; Cleret, A.; Matera, E.-L.; Chettab, K.; Mathé, D.; Valsesia-Wittmann, S.; Clémenceau, B.; Dumontet, C. Adipose cells promote resistance of breast cancer cells to trastuzumab-mediated antibody-dependent cellular cytotoxicity. Breast Cancer Res. 2015, 17, 57. [Google Scholar] [CrossRef] [PubMed]
  103. Fuentes-Mattei, E.; Velazquez-Torres, G.; Phan, L.; Zhang, F.; Chou, P.-C.; Shin, J.-H.; Choi, H.H.; Chen, J.-S.; Zhao, R.; Chen, J.; et al. Effects of obesity on transcriptomic changes and cancer hallmarks in estrogen receptor-positive breast cancer. J. Natl. Cancer Inst. 2014, 106. [Google Scholar] [CrossRef] [PubMed]
  104. Sabol, R.; Cote, A.; Bunnell, B. Adipose Stem Cells from Obese Individuals Promote Radioresistance of Estrogen Receptor Positive Breast Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, E615–E616. [Google Scholar] [CrossRef]
  105. 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] [PubMed]
  106. Sakurai, M.; Miki, Y.; Takagi, K.; Suzuki, T.; Ishida, T.; Ohuchi, N.; Sasano, H. Interaction with adipocyte stromal cells induces breast cancer malignancy via S100A7 upregulation in breast cancer microenvironment. Breast Cancer Res. 2017, 19, 70. [Google Scholar] [CrossRef] [PubMed]
  107. Mullooly, M.; Yang, H.P.; Falk, R.T.; Nyante, S.J.; Cora, R.; Pfeiffer, R.M.; Radisky, D.C.; Visscher, D.W.; Hartmann, L.C.; Carter, J.M.; et al. Relationship between crown-like structures and sex-steroid hormones in breast adipose tissue and serum among postmenopausal breast cancer patients. Breast Cancer Res. 2017, 19, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Carter, J.M.; Hoskin, T.L.; Pena, M.A.; Brahmbhatt, R.; Winham, S.J.; Frost, M.H.; Stallings-Mann, M.; Radisky, D.C.; Knutson, K.L.; Visscher, D.W.; et al. Macrophagic “Crown-like Structures” Are Associated with an Increased Risk of Breast Cancer in Benign Breast Disease. Cancer Prev. Res. 2018, 11, 113–119. [Google Scholar] [CrossRef] [PubMed]
  109. Hao, J.; Yan, F.; Zhang, Y.; Triplett, A.; Zhang, Y.; Schultz, D.A.; Sun, Y.; Zeng, J.; Silverstein, K.A.T.; Zheng, Q.; et al. Expression of adipocyte/macrophage fatty acid binding protein in tumor associated macrophages promotes breast cancer progression. Cancer Res. 2018. [Google Scholar] [CrossRef] [PubMed]
  110. Wu, B.; Sun, X.; Gupta, H.B.; Yuan, B.; Li, J.; Ge, F.; Chiang, H.-C.; Zhang, X.; Zhang, C.; Zhang, D.; et al. Adipose PD-L1 Modulates PD-1/PD-L1 Checkpoint Blockade Immunotherapy Efficacy in Breast Cancer. Oncoimmunology 2018, 7, e1500107. [Google Scholar] [CrossRef] [PubMed]
  111. Vonderheide, R.H.; Domchek, S.M.; Clark, A.S. Immunotherapy for Breast Cancer: What Are We Missing? Clin Cancer Res. 2017, 23, 2640–2646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Svartberg, J.; von Mühlen, D.; Sundsfjord, J.; Jorde, R. Waist circumference and testosterone levels in community dwelling men. The Tromsø study. Eur. J. Epidemiol. 2004, 19, 657–663. [Google Scholar] [CrossRef] [PubMed]
  113. Kelly, D.M.; Jones, T.H. Testosterone and obesity. Obes. Rev. 2015, 16, 581–606. [Google Scholar] [CrossRef] [PubMed]
  114. Cohen, P.G. Obesity in men: The hypogonadal-estrogen receptor relationship and its effect on glucose homeostasis. Med. Hypotheses 2008, 70, 358–360. [Google Scholar] [CrossRef] [PubMed]
  115. Ng Tang Fui, M.; Hoermann, R.; Prendergast, L.A.; Zajac, J.D.; Grossmann, M. Symptomatic response to testosterone treatment in dieting obese men with low testosterone levels in a randomized, placebo-controlled clinical trial. Int. J. Obes. 2017, 41, 420–426. [Google Scholar] [CrossRef] [PubMed]
  116. Michaud, J.E.; Billups, K.L.; Partin, A.W. Testosterone and prostate cancer: An evidence-based review of pathogenesis and oncologic risk. Ther. Adv. Urol. 2015, 7, 378–387. [Google Scholar] [CrossRef] [PubMed]
  117. Boyle, P.; Koechlin, A.; Bota, M.; d’Onofrio, A.; Zaridze, D.G.; Perrin, P.; Fitzpatrick, J.; Burnett, A.L.; Boniol, M. Endogenous and exogenous testosterone and the risk of prostate cancer and increased prostate-specific antigen (PSA) level: A meta-analysis. BJU Int. 2016, 118, 731–741. [Google Scholar] [CrossRef] [PubMed]
  118. Freedland, S.J.; Aronson, W.J. Examining the Relationship Between Obesity and Prostate Cancer. Rev. Urol. 2004, 6, 73–81. [Google Scholar] [PubMed]
  119. Campos-Silva, P.; Furriel, A.; Costa, W.S.; Sampaio, F.J.B.; Gregorio, B.M. Metabolic and testicular effects of the long-term administration of different high-fat diets in adult rats. Int. Braz. J. Urol. 2015, 41, 569–575. [Google Scholar] [CrossRef] [PubMed]
  120. Fan, W.; Xu, Y.; Liu, Y.; Zhang, Z.; Lu, L.; Ding, Z. Obesity or Overweight, a Chronic Inflammatory Status in Male Reproductive System, Leads to Mice and Human Subfertility. Front. Physiol. 2017, 8, 1117. [Google Scholar] [CrossRef] [PubMed]
  121. Barnes, K.T.; Smith, B.J.; Lynch, C.F.; Gupta, A. Obesity and invasive penile cancer. Eur. Urol. 2013, 63, 588–589. [Google Scholar] [CrossRef] [PubMed]
  122. Barnes, K.T.; McDowell, B.D.; Button, A.; Smith, B.J.; Lynch, C.F.; Gupta, A. Obesity is associated with increased risk of invasive penile cancer. BMC Urol. 2016, 16, 42. [Google Scholar] [CrossRef] [PubMed]
  123. Douglawi, A.; Masterson, T.A. Updates on the epidemiology and risk factors for penile cancer. Transl. Androl. Urol. 2017, 6, 785–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Zadra, G.; Photopoulos, C.; Loda, M. The fat side of prostate. Biochimica et Biophysica Acta 2013, 1831, 1518–1532. [Google Scholar] [CrossRef] [PubMed]
  125. Bracarda, S.; Logothetis, C.; Sternberg, C.N.; Oudard, S. Current and emerging treatment modalities for metastatic castration-resistant prostate cancer. BJU Int. 2011, 107 (Suppl. 2), 13–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Taylor, R.A.; Lo, J.; Ascui, N.; Watt, M.J. Linking obesogenic dysregulation to prostate cancer progression. Endocr. Connect. 2015, 4, R68–R80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Gucalp, A.; Iyengar, N.M.; Zhou, X.K.; Giri, D.D.; Falcone, D.J.; Wang, H.; Williams, S.; Krasne, M.D.; Yaghnam, I.; Kunzel, B.; et al. Periprostatic adipose inflammation is associated with high-grade prostate cancer. Prostate Cancer Prostatic Dis. 2017, 20, 418–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Ribeiro, R.; Monteiro, C.; Catalán, V.; Hu, P.; Cunha, V.; Rodríguez, A.; Gómez-Ambrosi, J.; Fraga, A.; Príncipe, P.; Lobato, C.; et al. Obesity and prostate cancer: Gene expression signature of human periprostatic adipose tissue. BMC Med. 2012, 10, 108. [Google Scholar] [CrossRef] [PubMed]
  129. Laurent, V.; Guérard, 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] [PubMed] [Green Version]
  130. De Marzo, A.M.; Platz, E.A.; Sutcliffe, S.; Xu, J.; Grönberg, H.; Drake, C.G.; Nakai, Y.; Isaacs, W.B.; Nelson, W.G. Inflammation in prostate carcinogenesis. Nat. Rev. Cancer 2007, 7, 256–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Sfanos, K.S.; De Marzo, A.M. Prostate cancer and inflammation: The evidence. Histopathology 2012, 60, 199–215. [Google Scholar] [CrossRef] [PubMed]
  132. Huang, J.; Duran, A.; Reina-Campos, M.; Valencia, T.; Castilla, E.A.; Müller, T.D.; Tschöp, M.H.; Moscat, J.; Diaz-Meco, M.T. Adipocyte p62/SQSTM1 Suppresses Tumorigenesis through Opposite Regulations of Metabolism in Adipose Tissue and Tumor. Cancer Cell 2018, 33, 770–784. [Google Scholar] [CrossRef] [PubMed]
  133. Sagar, G.; Sah, R.P.; Javeed, N.; Dutta, S.K.; Smyrk, T.C.; Lau, J.S.; Giorgadze, N.; Tchkonia, T.; Kirkland, J.L.; Chari, S.T.; et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut 2016, 65, 1165–1174. [Google Scholar] [CrossRef] [PubMed]
  134. Carbone, C.; Piro, G.; Gaianigo, N.; Ligorio, F.; Santoro, R.; Merz, V.; Simionato, F.; Zecchetto, C.; Falco, G.; Conti, G.; et al. Adipocytes sustain pancreatic cancer progression through a non-canonical WNT paracrine network inducing ROR2 nuclear shuttling. Int. J. Obes. 2018, 42, 334–343. [Google Scholar] [CrossRef] [PubMed]
  135. Ribeiro, R.; Monteiro, C.; Cunha, V.; Oliveira, M.J.; Freitas, M.; Fraga, A.; Príncipe, P.; Lobato, C.; Lobo, F.; Morais, A.; et al. Human periprostatic adipose tissue promotes prostate cancer aggressiveness in vitro. J. Exp. Clin. Cancer Res. 2012, 31, 32. [Google Scholar] [CrossRef] [PubMed]
  136. Collier, A.; Ghosh, S.; Mcglynn, B.; Hollins, G. Prostate Cancer, Androgen Deprivation Therapy, Obesity, the Metabolic Syndrome, Type 2 Diabetes, and Cardiovascular Disease: A Review. Am. J. Clin. Oncol. 2012, 35, 504–509. [Google Scholar] [CrossRef] [PubMed]
  137. O’Reilly, M.W.; House, P.J.; Tomlinson, J.W. Understanding androgen action in adipose tissue. J. Steroid Biochem. Mol. Biol. 2014, 143, 277–284. [Google Scholar]
  138. Meyer, T.E.; Chu, L.W.; Li, Q.; Yu, K.; Rosenberg, P.S.; Menashe, I.; Chokkalingam, A.P.; Quraishi, S.M.; Huang, W.-Y.; Weiss, J.M.; et al. The association between inflammation-related genes and serum androgen levels in men: The Prostate, Lung, Colorectal, and Ovarian Study. Prostate 2012, 72, 65–71. [Google Scholar] [CrossRef] [PubMed]
  139. Zitvogel, L.; Pitt, J.M.; Daillère, R.; Smyth, M.J.; Kroemer, G. Mouse models in oncoimmunology. Nat. Rev. Cancer 2016, 16, 759–773. [Google Scholar] [CrossRef] [PubMed]
  140. Kleinert, M.; Clemmensen, C.; Hofmann, S.M.; Moore, M.C.; Renner, S.; Woods, S.C.; Huypens, P.; Beckers, J.; de Angelis, M.H.; Schürmann, A.; et al. Animal models of obesity and diabetes mellitus. Nat. Rev. Endocrinol. 2018, 14, 140–162. [Google Scholar] [CrossRef] [PubMed]
  141. Saxena, N.K.; Sharma, D. Multifaceted Leptin network: The molecular connection between obesity and breast cancer. J. Mammary Gland Biol. Neoplasia 2013, 18, 309–320. [Google Scholar] [CrossRef] [PubMed]
  142. Akase, T.; Shimada, T.; Harasawa, Y.; Akase, T.; Ikeya, Y.; Nagai, E.; Iizuka, S.; Nakagami, G.; Iizaka, S.; Sanada, H.; et al. Preventive Effects of Salacia reticulata on Obesity and Metabolic Disorders in TSOD Mice. Evid.-Based Complement. Altern. Med. 2011, 2011, 484590. [Google Scholar] [CrossRef] [PubMed]
  143. Takahashi, T.; Deuschle, U.; Taira, S.; Nishida, T.; Fujimoto, M.; Hijikata, T.; Tsuneyama, K. Tsumura-Suzuki obese diabetic mice-derived hepatic tumors closely resemble human hepatocellular carcinomas in metabolism-related genes expression and bile acid accumulation. Hepatol. Int. 2018, 12, 254–261. [Google Scholar] [CrossRef] [PubMed]
  144. Hirayama, I.; Yi, Z.; Izumi, S.; Arai, I.; Suzuki, W.; Nagamachi, Y.; Kuwano, H.; Takeuchi, T.; Izumi, T. Genetic analysis of obese diabetes in the TSOD mouse. Diabetes 1999, 48, 1183–1191. [Google Scholar] [CrossRef] [PubMed]
  145. Murotomi, K.; Umeno, A.; Yasunaga, M.; Shichiri, M.; Ishida, N.; Abe, H.; Yoshida, Y.; Nakajima, Y. Type 2 diabetes model TSOD mouse is exposed to oxidative stress at young age. J. Clin. Biochem. Nutr. 2014, 55, 216–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Nishida, T.; Tsuneyama, K.; Fujimoto, M.; Nomoto, K.; Hayashi, S.; Miwa, S.; Nakajima, T.; Nakanishi, Y.; Sasaki, Y.; Suzuki, W.; et al. Spontaneous onset of nonalcoholic steatohepatitis and hepatocellular carcinoma in a mouse model of metabolic syndrome. Lab. Invest. 2013, 93, 230–241. [Google Scholar] [CrossRef] [PubMed]
  147. Iizuka, S.; Suzuki, W.; Tabuchi, M.; Nagata, M.; Imamura, S.; Kobayashi, Y.; Kanitani, M.; Yanagisawa, T.; Kase, Y.; Takeda, S.; et al. Diabetic complications in a new animal model (TSOD mouse) of spontaneous NIDDM with obesity. Exp. Anim. 2005, 54, 71–83. [Google Scholar] [CrossRef] [PubMed]
  148. Takahashi, T.; Nishida, T.; Baba, H.; Hatta, H.; Imura, J.; Sutoh, M.; Toyohara, S.; Hokao, R.; Watanabe, S.; Ogawa, H.; et al. Histopathological characteristics of glutamine synthetase-positive hepatic tumor lesions in a mouse model of spontaneous metabolic syndrome (TSOD mouse). Mol. Clin. Oncol. 2016, 5, 267–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Tsuneyama, K.; Nishitsuji, K.; Matsumoto, M.; Kobayashi, T.; Morimoto, Y.; Tsunematsu, T.; Ogawa, H. Animal models for analyzing metabolic syndrome-associated liver diseases. Pathol. Int. 2017, 67, 539–546. [Google Scholar] [CrossRef] [PubMed]
  150. Miyata, S.; Yamada, N.; Kwada, T. Possible involvement of hypothalamic nucleobindin-2 in hyperphagic feeding in Tsumara Suzuki Obese Diabetes mice. Biol. Pharm. Bull. 2012, 35, 1784–1793. [Google Scholar] [CrossRef] [PubMed]
  151. Takahashi, A.; Tabuchi, M.; Suzuki, W.; Iizuka, S.; Nagata, M.; Ikeya, Y.; Takeda, S.; Shimada, T.; Aburada, M. Insulin resistance and low sympathetic nerve activity in the Tsumura Suzuki obese diabetic mouse: A new model of spontaneous type 2 diabetes mellitus and obesity. Metabolism 2006, 55, 1664–1669. [Google Scholar] [CrossRef] [PubMed]
  152. Suzuki, W.; Iizuka, S.; Tabuchi, M.; Funo, S.; Yanagisawa, T.; Kimura, M.; Sato, T.; Endo, T.; Kawamura, H. A new mouse model of spontaneous diabetes derived from ddY strain. Exp. Anim. 1999, 48, 181–189. [Google Scholar] [CrossRef] [PubMed]
  153. Leiter, E.H. Selecting the “Right” Mouse Model for Metabolic Syndrome and Type 2 Diabetes Research. In Type 2 Diabetes: Methods and Protocols; Methods in Molecular Biology; Stocker, C., Ed.; Humana Press: Totowa, NJ, USA, 2009; pp. 1–17. ISBN 978-1-59745-448-3. [Google Scholar]
  154. Merkwirth, C.; Langer, T. Prohibitin function within mitochondria: Essential roles for cell proliferation and cristae morphogenesis. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2009, 1793, 27–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Ande, S.R.; Gu, Y.; Nyomba, B.L.G.; Mishra, S. Insulin induced phosphorylation of prohibitin at tyrosine 114 recruits Shp1. Biochim. Biophys. Acta 2009, 1793, 1372–1378. [Google Scholar] [CrossRef] [PubMed]
  156. Ande, S.R.; Nguyen, K.H.; Padilla-Meier, G.P.; Wahida, W.; Nyomba, B.L.G.; Mishra, S. Prohibitin overexpression in adipocytes induces mitochondrial biogenesis, leads to obesity development, and affects glucose homeostasis in a sex-specific manner. Diabetes 2014, 63, 3734–3741. [Google Scholar] [CrossRef] [PubMed]
  157. Ande, S.R.; Nguyen, K.H.; Grégoire Nyomba, B.L.; Mishra, S. Prohibitin-induced, obesity-associated insulin resistance and accompanying low-grade inflammation causes NASH and HCC. Sci. Rep. 2016, 6, 23608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Ande, S.R.; Nguyen, K.H.; Padilla-Meier, G.P.; Nyomba, B.L.G.; Mishra, S. Expression of a mutant prohibitin from the aP2 gene promoter leads to obesity-linked tumor development in insulin resistance-dependent manner. Oncogene 2016, 35, 4459–4470. [Google Scholar] [CrossRef] [PubMed]
  159. Mishra, S.; Murphy, L.C.; Murphy, L.J. The Prohibitins: Emerging roles in diverse functions. J. Cell. Mol. Med. 2006, 10, 353–363. [Google Scholar] [CrossRef] [PubMed]
  160. Rogers, P.C.; Meacham, L.R.; Oeffinger, K.C.; Henry, D.W.; Lange, B.J. Obesity in pediatric oncology. Pediatr. Blood Cancer 2005, 45, 881–891. [Google Scholar] [CrossRef] [PubMed]
  161. McCormick, D.P.; Sarpong, K.; Jordan, L.; Ray, L.A.; Jain, S. Infant obesity: Are we ready to make this diagnosis? J. Pediatr. 2010, 157, 15–19. [Google Scholar] [CrossRef] [PubMed]
  162. Sovio, U.; Kaakinen, M.; Tzoulaki, I.; Das, S.; Ruokonen, A.; Pouta, A.; Hartikainen, A.-L.; Molitor, J.; Järvelin, M.-R. How do changes in body mass index in infancy and childhood associate with cardiometabolic profile in adulthood? Findings from the Northern Finland Birth Cohort 1966 Study. Int. J. Obes. 2014, 38, 53–59. [Google Scholar] [CrossRef] [PubMed]
  163. Berger, N.A. Young Adult Cancer: Influence of the Obesity Pandemic. Obesity (Silver Spring) 2018, 26, 641–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Barnea, D.; Raghunathan, N.; Friedman, D.N.; Tonorezos, E.S. Obesity and Metabolic Disease After Childhood Cancer. Oncology (Williston Park) 2015, 29, 849–855. [Google Scholar] [PubMed]
  165. Hursting, S.D.; Nunez, N.P.; Varticovski, L.; Vinson, C. The Obesity-Cancer Link: Lessons Learned from a Fatless Mouse. Cancer Res. 2007, 67, 2391–2393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Roberson, L.L.; Aneni, E.C.; Maziak, W.; Agatston, A.; Feldman, T.; Rouseff, M.; Tran, T.; Blaha, M.J.; Santos, R.D.; Sposito, A.; et al. Beyond BMI: The “Metabolically healthy obese” phenotype & its association with clinical/subclinical cardiovascular disease and all-cause mortality—A systematic review. BMC Public Health 2014, 14, 14. [Google Scholar]
  167. Mathew, H.; Farr, O.M.; Mantzoros, C.S. Metabolic Health and Weight: Understanding metabolically unhealthy normal weight or metabolically healthy obese patients. Metabolism 2016, 65, 73–80. [Google Scholar] [CrossRef] [PubMed]
  168. Drew, D.A.; Cao, Y.; Chan, A.T. Aspirin and colorectal cancer: The promise of precision chemoprevention. Nat. Rev. Cancer 2016, 16, 173–186. [Google Scholar] [CrossRef] [PubMed]
  169. Chang, H.-H.; Moro, A.; Chou, C.E.N.; Dawson, D.W.; French, S.; Schmidt, A.I.; Sinnett-Smith, J.; Hao, F.; Hines, O.J.; Eibl, G.; et al. Metformin Decreases the Incidence of Pancreatic Ductal Adenocarcinoma Promoted by Diet-induced Obesity in the Conditional KrasG12D Mouse Model. Sci. Rep. 2018, 8, 5899. [Google Scholar] [CrossRef] [PubMed]
  170. Padwal, R.S.; Pajewski, N.M.; Allison, D.B.; Sharma, A.M. Using the Edmonton obesity staging system to predict mortality in a population-representative cohort of people with overweight and obesity. CMAJ 2011, 183, E1059–E1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Dalgaard, K.; Landgraf, K.; Heyne, S.; Lempradl, A.; Longinotto, J.; Gossens, K.; Ruf, M.; Orthofer, M.; Strogantsev, R.; Selvaraj, M.; et al. Trim28 Haploinsufficiency Triggers Bi-stable Epigenetic Obesity. Cell 2016, 164, 353–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Li, R.; Grimm, S.A.; Chrysovergis, K.; Kosak, J.; Wang, X.; Du, Y.; Burkholder, A.; Janardhan, K.; Mav, D.; Shah, R.; et al. Obesity, Rather Than Diet, Drives Epigenomic Alterations in Colonic Epithelium Resembling Cancer Progression. Cell Metab. 2014, 19, 702–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Li, R.; Grimm, S.A.; Mav, D.; Gu, H.; Djukovic, D.; Shah, R.; Merrick, B.A.; Raftery, D.; Wade, P.A. Transcriptome and DNA Methylome Analysis in a Mouse Model of Diet-Induced Obesity Predicts Increased Risk of Colorectal Cancer. Cell Rep. 2018, 22, 624–637. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Obesity has been linked to more than 13 types of cancers in the body. The white adipose tissue (WAT) is home to diverse cell types: adipocytes, adipose stem cells, endothelial cells and many resident and infiltrating immune cells. During obesity, WAT releases a plethora of molecules with autocrine, paracrine and endocrine functions, including growth factors, adipokines, pro-inflammatory molecules, fatty acids (FA) and lipid metabolites and many others, which create a favorable condition for cancer to develop. Obesity has been shown to increase the risk of 13 cancer types and 3 others with limited evidence (*) [5]. The link(s) between obesity and its complications to multiple cancer types observed in humans are currently unknown. GCA: gastric cardia adenocarcinoma; IGF: insulin-like growth factor; VEGF: vascular endothelial growth factor; IL: interleukin. TNF: tumor necrosis factor; MCP: monocyte chemoattractant protein.
Figure 1. Obesity has been linked to more than 13 types of cancers in the body. The white adipose tissue (WAT) is home to diverse cell types: adipocytes, adipose stem cells, endothelial cells and many resident and infiltrating immune cells. During obesity, WAT releases a plethora of molecules with autocrine, paracrine and endocrine functions, including growth factors, adipokines, pro-inflammatory molecules, fatty acids (FA) and lipid metabolites and many others, which create a favorable condition for cancer to develop. Obesity has been shown to increase the risk of 13 cancer types and 3 others with limited evidence (*) [5]. The link(s) between obesity and its complications to multiple cancer types observed in humans are currently unknown. GCA: gastric cardia adenocarcinoma; IGF: insulin-like growth factor; VEGF: vascular endothelial growth factor; IL: interleukin. TNF: tumor necrosis factor; MCP: monocyte chemoattractant protein.
Cancers 10 00523 g001
Figure 2. The development of obesity-linked tumors in combined rodent models, Tsumura Suzuki Obese Diabetes (TSOD) and Mito-Ob/mMito-Ob mouse models. Tsumura Suziki Obese Diabetes (TSOD) and Mito-Ob/mMito-Ob mouse models. The combined rodent models develop obesity and cancer through independent manipulations in genetics, diet, and inducible techniques [139,140]. They are excellent models to study cancer progression under an obese environment. Since the two morbidities can develop independent of each other, these models do not address whether obesity initiates tumorigenesis spontaneously. Both the male TSOD and Mito-Ob/mMito-Ob mouse models develop tumors spontaneously from genetic manipulations that led to an obese and diabetic phenotype, which may better explain the inducible cancer risks from obesity. For this mini-review, only rodent models are discussed, as they are the most commonly used animals. We acknowledge that other models exist and contribute significantly to obesity/cancer research but remain outside the scope of this mini-review. Nidd: non-insulin-dependent diabetes; aP2: adipocyte protein 2.
Figure 2. The development of obesity-linked tumors in combined rodent models, Tsumura Suzuki Obese Diabetes (TSOD) and Mito-Ob/mMito-Ob mouse models. Tsumura Suziki Obese Diabetes (TSOD) and Mito-Ob/mMito-Ob mouse models. The combined rodent models develop obesity and cancer through independent manipulations in genetics, diet, and inducible techniques [139,140]. They are excellent models to study cancer progression under an obese environment. Since the two morbidities can develop independent of each other, these models do not address whether obesity initiates tumorigenesis spontaneously. Both the male TSOD and Mito-Ob/mMito-Ob mouse models develop tumors spontaneously from genetic manipulations that led to an obese and diabetic phenotype, which may better explain the inducible cancer risks from obesity. For this mini-review, only rodent models are discussed, as they are the most commonly used animals. We acknowledge that other models exist and contribute significantly to obesity/cancer research but remain outside the scope of this mini-review. Nidd: non-insulin-dependent diabetes; aP2: adipocyte protein 2.
Cancers 10 00523 g002

Share and Cite

MDPI and ACS Style

Xu, Y.X.Z.; Mishra, S. Obesity-Linked Cancers: Current Knowledge, Challenges and Limitations in Mechanistic Studies and Rodent Models. Cancers 2018, 10, 523. https://doi.org/10.3390/cancers10120523

AMA Style

Xu YXZ, Mishra S. Obesity-Linked Cancers: Current Knowledge, Challenges and Limitations in Mechanistic Studies and Rodent Models. Cancers. 2018; 10(12):523. https://doi.org/10.3390/cancers10120523

Chicago/Turabian Style

Xu, Yang Xin Zi, and Suresh Mishra. 2018. "Obesity-Linked Cancers: Current Knowledge, Challenges and Limitations in Mechanistic Studies and Rodent Models" Cancers 10, no. 12: 523. https://doi.org/10.3390/cancers10120523

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