Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Dec 1.
Published in final edited form as: Breast Cancer Res Treat. 2013 Nov 12;142(3):489–503. doi: 10.1007/s10549-013-2741-5

Role of HGF in obesity-associated tumorigenesis: C3(1)-TAg mice as a model for human basal-like breast cancer

Sneha Sundaram *, Alex J Freemerman *, Amy R Johnson *, J Justin Milner *, Kirk K McNaughton $, Joseph A Galanko %, Katharine M Bendt +, David B Darr +, Charles M Perou +,@,^, Melissa A Troester ~,&,^, Liza Makowski ~,^,*,%,#
PMCID: PMC3904507  NIHMSID: NIHMS539617  PMID: 24218051

Abstract

Obesity is associated with basal-like breast cancer (BBC), an aggressive breast cancer subtype. The objective of this study was to determine whether obesity promotes BBC onset in adulthood and to evaluate the role of stromal-epithelial interactions in obesity-associated tumorigenesis. We hypothesized that hepatocyte growth factor (HGF) plays a promoting role in BBC, which express the HGF receptor, c-Met. In C3(1)-Tag mice, a murine model of BBC, we demonstrated that obesity leads to a significant increase in HGF secretion and an associated decrease in tumor latency. By immunohistochemical analysis, normal mammary gland exhibited obesity-induced HGF, c-Met and phospho-c-Met, indicating that activation of the cascade was obesity-driven. HGF secretion was also increased from primary mammary fibroblasts isolated from normal mammary glands and tumors of obese mice compared to lean. These results demonstrate that obesity-induced elevation of HGF expression is a stable phenotype, maintained after several passages, and after removal of dietary stimulation. Conditioned media from primary tumor fibroblasts from obese mice drove tumor cell proliferation. In co-culture, neutralization of secreted HGF blunted tumor cell migration, further linking obesity-mediated HGF-dependent effects to in vitro measures of tumor aggressiveness. In sum, these results demonstrate that HGF/c-Met plays an important role in obesity-associated carcinogenesis. Understanding the effects of obesity on risk and progression is important given that epidemiologic studies imply a portion of BBC could be eliminated by reducing obesity.

Keywords: Basal-like breast cancer, tumor latency, microenvironment, normal mammary gland, high fat diet-induced obesity, fibroblast

INTRODUCTION

Obesity is associated with an increased risk of postmenopausal breast cancer in several population-based studies, but links between obesity and premenopausal breast cancer have been controversial [1,2]. The Breast Cancer Prevention Trial suggested an elevated risk of premenopausal breast cancer among obese women, in contrast to several previous studies suggesting a protective effect of obesity in this group [3,4]. The Breast Cancer Association Consortium reported that triple negative tumors are specifically associated with obesity in younger women [5,6], which is in agreement with several smaller studies that have also linked obesity or high waist-to-hip ratio with triple negative or basal-like breast cancer (BBC) [7-9,1]. These studies emphasize that considering tumor subtype is essential when reporting obesity-related breast cancer risk, and that, taken together, strong associations with obesity and BBC exist.

BBC is a triple-negative subtype characterized by the lack of expression of estrogen receptor (ER), progesterone receptor (PR), and type-2 human epidermal growth factor receptor (HER2) and as such, BBCs are the only breast cancer subtype without a targeted therapy [10]. BBC disproportionately affects young (< 50 years old) and African-American women [11,12]. Given that African-Americans experience higher rates of pubertal and adult obesity [13], and retain more weight after pregnancy [14], it is important to better understand the biological mechanisms by which obesity, a modifiable risk factor, increases the risk of BBC.

Adding to the complexity of the relationship between obesity and differing breast cancer subtypes, the effects of obesity on the breast may depend upon the timing of exposure. Animal studies suggest that early life obesity is a risk factor for breast cancer, showing that high fat diet-induced obesity alters puberty onset, mammary gland development and morphology [15,16], as well as decreased tumor latency [17]. However, there has been limited study on the effects of obesity on mammary carcinogenesis later in life. Of particular relevance to human cancer, the effect of diet-induced obesity during adulthood has not been evaluated. Adult obesity is an epidemic in the US and is increasing worldwide [18], hence understanding the role of obesity exposure during adulthood is a question with high public health impact.

Stromal-epithelial interactions are known to modulate breast cancer [19]. Stromal remodeling is important for understanding BBC specifically, because this subtype has unique interactions with surrounding stroma. Although changes to the stroma are less dramatic once maturity is reached, recent data has suggested that effects of obesity on mammary stroma are striking [20-22]. Co-culture of BBC epithelial cells with fibroblasts upregulates a wide range of cytokines, immune response pathways, and hepatic fibrosis-associated genes [23]. This is important because stroma-derived hepatocyte growth factor (HGF) can interact with the c-Met receptor, an important pathway in basal-like carcinogenesis examined herein [24,25]. HGF levels are frequently elevated in obese patients [26] and reduced in patients with weight loss [27]. Elevations of plasma HGF are correlated with breast cancer, specifically ER-negative [28-30]. We hypothesized that, similar to results during puberty, obesity would lead to mammary tumorigenesis in adulthood, potentially through HGF/c-Met-mediated signaling. We selected a preclinical model focusing on the BBC subtype, since obesity is an important risk factor for triple negative disease [12,31,11]. Our data demonstrate that in C3(1)-TAg mice associations between obesity and tumorigenesis mirrored human epidemiologic findings. Furthermore, our results indicate that HGF may play a role in mediating the onset of obesity-associated BBCs and suggest that HGF/c-Met signaling is a plausible molecular target for prevention of obesity-associated breast cancer.

METHODS

Reagents and antibodies

Details are provided in the supplementary materials.

C3(1)-TAg Mouse Model

Animals

C3(1)-TAg mice (described previously [32]) were used to study the role of obesity on BBC, as these mice were shown to be highly representative of human BBC [33]. Studies were performed with approval and in accordance with guidelines of the Institutional Animal Care and Use Committee at the University of North Carolina at Chapel Hill (UNC-CH, NC). Female C3(1)-TAg mice were obtained through a collaboration with the UNC Lineberger Comprehensive Cancer Center (LCCC) Mouse Phase I Unit (MP1U). Nulliparous female C3(1)-TAg mice were randomly assigned to various diet groups at adulthood, 10 weeks of age, after the end of puberty (Model of study design, Supplementary (Suppl). Figure 1). Diet details are provided in the supplementary materials and (Suppl. Table 1).

Tumor latency, number, growth and volume

Following initiation of diets at 10 weeks of age, mice were monitored for tumor development by palpating thrice weekly. Tumor latency was defined as the age at detection of the first tumor. After detection of the first tumor, tumor volumes were measured weekly over 3 weeks using ultrasound measurements with the Visualsonics 2000 (Toronto, Canada). Details are provided in the supplementary material.

Body Weight & Composition

Prior to starting mice on diet and weekly until sacrifice, body weight was measured. Body composition including lean mass, fat mass, free water content and total water content of non-anesthetized mice was also measured at 0, 4 and 8 weeks on diet using the EchoMRI-100 quantitative magnetic resonance whole body composition analyzer (Echo Medical Systems, Houston, TX). Fat mass is presented as percent fat mass over total body weight. There were no significant declines in absolute lean mass in grams (data not shown).

Blood Glucose

Blood glucose was measured prior to start of diet and at sacrifice following a 6h fast using a Bayer Contour Blood Glucose Monitor (Bayer HealthCare LLC, Tarrytown, NY).

Tissue Harvest

Details are provided in the supplementary materials.

Plasma and Tissue Cytokines Panel

Details are provided in the supplementary materials.

Quantitative PCR (qPCR)

Total RNA from normal mammary gland and tumor was isolated as in Sampey et. al. [34]. Real-time quantitative PCR for HGF, c-Met and aromatase (Cyp19a1) was performed [34].

Immunohistochemistry/Immunofluorescence of HGF, c-Met, pc-Met, -SMA and/or SV40-TAg in Normal Mammary Glands and Tumors

Details of immunohistochemistry (IHC) and immunofluorescence (IF) staining and methods for scanning slides and analysis are provided in the supplementary materials.

Co-culture studies of stromal-epithelial interactions

Animals

Details are provided in the supplementary materials.

Fibroblast isolation

Normal associated fibroblasts (“NAF”) were isolated from normal inguinal mammary glands with no evidence of cancer, while cancer associated fibroblasts (“CAF”) were isolated from tumors from abdominal or inguinal mammary glands with tumors using methods from Fleming, et. al. [35]. Details are provided in the supplementary materials.

Co-culture

All culture experiments were completed in triplicate using NAFs and CAFs from mouse diet groups A, B, and C. 4T1 basal-like breast cancer-like (CRL-2539) cell line was obtained from ATCC (Manassas, VA). Details of the co-culture studies are provided in the supplementary materials. The media from these studies was centrifuged at 1620 × g and supernatant was assayed for HGF concentrations by ELISA (Abcam, Cambridge, MA) using a Bio-Rad Model 680 Microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA). Western immunoblot analysis in 4T1 cells treated as described in supplementary materials for phospho- and total c-Met expression was performed as previously described [36].

Cell Proliferation and Wound Migration Assay

Details of the proliferation study are provided in the supplementary materials. Wound migration assay was performed as described previously by Camp et al. [23]. Percent wound closure was calculated by measuring the width of scratch at 0h (time point of scratch) and at 12h (end point of study) using NIH ImageJ 1.46 software and the following formula: (0h width – 12h width * 100)/0h width.

Statistical Analysis

Data are expressed as mean ± standard error of the mean (SEM). All analysis of variance (ANOVA) with Tukey’s post-hoc test for statistical differences were performed using SAS version 9.2 (SAS Institute, Cary, NC) or SPSS (version 20) software (IBM SPSS Statistic 20.0, Armonk, NY). P-values < 0.05 were considered statistically significant.

RESULTS

C3(1)-TAg basal-like tumor latency is decreased by obesity: an association with HGF

Mice were fed control or obesogenic diets in adulthood and tumor and metabolic parameters were evaluated as indicated in Suppl. Figure 1. Mice made obese by exposure to obesogenic diets (45% and 60%) exhibited significant decreases in tumor latency compared with lean controls (10%). The hazard ratios comparing either 45% or 60% to 10% were 2.41 or 2.87, respectively (Figure 1A). Median times to onset of 10%, 45%, and 60%-fed mice were 19.14, 17.29, and 16.57 weeks, respectively. Obese mice displayed significantly earlier median latencies (10% vs. 45% p=0.0236 and 10% vs. 60% p=0.0091) but there was no significant difference between 45% and 60%-fed median latencies (Figure 1A). Using a chi-square tests with a degree of freedom of 2, 10% vs. 60% equaled 6.801, 10% vs. 45% equaled 5.121, and 45% vs. 60% was 0.193. Tumor burden (Figure 1B) or progression (Figure 1C) were not significantly altered. We next confirmed that expression of the transgene driving tumorigenesis in C3(1)-TAg mice was not responsible for obesity-mediated alterations in latency. Indeed, SV40 TAg staining exhibited no significant obesity-induced elevations in normal mammary gland (Suppl. Figures 2A and B) or tumors (Suppl. Figures 2C and D). The lack of change in tumor burden in the C3(1)-TAg mice suggested an effect of obesity on early tumor etiology, rather than on tumor progression.

Figure 1. Obesity-mediated decrease in latency correlates with elevated HGF expression.

Figure 1

A) Upon initiation of 10, 45 and 60% kcal derived from fat diets at 10 weeks of age, mice were palpated three times weekly for tumor onset. Hazard ratios comparing either 45% or 60% to 10% were 2.41 or 2.87, respectively. Median latencies are indicated ((10% vs. 45% p=0.0236 and 10% vs. 60% p=0.0091). B) Number of tumors was detected at sacrifice (n=15 mice). C) Tumor volume was measured by ultrasound (n=15 mice). D) HGF mRNA expression was determined using qPCR analysis in n=8 normal mammary glands. * 10% vs. 60% (P = 0.025).

Obesity strongly regulated HGF/c-Met signaling in normal mammary gland

HGF is a potential mediator of obesity-induced alterations in breast tumor onset since it is regulated by obesity [26,25,24,2] and could alter tumor etiology. The association between obesity, HGF, and tumorigenesis has not been investigated to date. Hence we examined HGF/c-Met signaling in both normal mammary gland and tumors. Obesity significantly elevated HGF mRNA levels in the normal mammary gland (Figure 1D, P=0.025 10% vs. 60%-fed).

To examine protein expression and localization in tissues, immunohistochemistry (IHC) was undertaken. HGF expression was primarily localized to stroma with additional staining in epithelium in normal mammary glands (Figure 2A). Quantification demonstrated that HGF protein concentrations measured in the stroma were significantly increased in obese 60%-fed mice compared with 10%-fed lean controls (Figure 2B; P = 0.011). HGF mediates its effects via its cognate receptor c-Met, a proto-oncogenic tyrosine kinase receptor involved in BBC [37,25,24]. c-Met mRNA expression did not exhibit significant obesity-induced differences (Figure 2C). Since c-Met mRNA is degraded rapidly [38], we quantified protein concentrations through IHC. Representative photomicrographs of cell membrane c-Met expression in normal mammary gland demonstrated primarily epithelial localization (Figure 2D). Quantification revealed a strong obesity-induced upregulation of c-Met protein expression (Figure 2E). c-Met protein was significantly increased in obese 45% (P=0.04) and 60% (P=0.001)-fed mice compared with 10%-fed controls, and between obese 60% (P=0.025)-fed mice compared with 45%-fed mice. To examine activation of the HGF/c-Met cascade, phospho-c-Met (pc-Met) expression was quantified. Representative photomicrographs of normal mammary gland demonstrated both membrane and cytoplasmic (internalized c-Met [39,40]) staining (Figure 2F). Obesity upregulated expression of pc-Met (Figure 2G) and significant increases were observed in 45% (P = 0.011) and 60% (P = 0.0001) compared with 10%-fed controls, and between obese 60%-fed (P = 0.0125) mice and 45%-fed mice. Tumors were next evaluated for obesity-mediated alterations in HGF/c-Met signaling. In tumors, HGF protein concentrations were not significantly regulated by obesity (Figure 3A and B). Similarly, neither c-Met mRNA expression nor protein expression was significantly regulated by diet, although obesity-induced trends existed (Figure 3C-E). Western immunoblotting of tumors demonstrated variable c-Met protein expression, with no significant differences detected between groups (Figure 3F). In addition, no significant differences were observed in the levels of pc-Met in the tumors (Figures 3G and 3H). Thus, in normal mammary glands obesity positively regulated HGF and c-Met expression and signaling in obese mice compared to lean. However, significant obesity-mediated HGF/c-Met regulation in tumors was not evident.

Figure 2. Obesity upregulated HGF, c-Met, and pc-Met expression in normal mammary glands.

Figure 2

Figure 2

Figure 2

A) Representative photomicrographs (40X) of HGF staining in negative control (secondary only), 10%, 45% and 60% diet groups in normal (unaffected) mammary glands. Arrows indicate positive HGF staining in the stroma. B) Total HGF protein levels of the normal mammary glands were quantified using the Aperio ImageScope color deconvolution algorithm for n = 2 sections from 5 mice per diet group. * 10% vs. 60% (P = 0.011). C) c-Met mRNA expression was determined using qPCR analysis in normal mammary glands for n = 7. D) Representative photomicrographs (40X) of the IHC analysis of negative control and membrane localized c-Met staining in 10%, 45%, and 60% diet groups (arrows). E) Total c-Met protein levels in normal mammary glands were measured using the Aperio ImageScope membrane algorithm and quantified as above. * 10% vs. 45% (P = 0.04) and 10% vs. 60% (P = 0.001); ^ 45% vs. 60% (P = 0.025). F) Representative photomicrographs (40X) of the IHC analysis of negative control and pc-Met staining in 10%, 45%, and 60% diet groups. G) Total pc-Met protein levels in normal mammary glands were measured using the Aperio ImageScope color deconvolution algorithm and quantified as above. * 10% vs. 45% (P = 0.011) and 10% vs. 60% (P = 0.0001); ^ 45% vs. 60% (P = 0.0125).

Figure 3. Obesity minimally modified HGF, c-Met, and pc-Met expression in tumors.

Figure 3

Figure 3

Figure 3

Figure 3

A) Representative photomicrographs (40X) of the IHC analysis of tumors showing HGF staining in negative control and 10%, 45%, and 60% diet groups. B) HGF protein levels in tumors were quantified using the Aperio ImageScope color deconvolution algorithm for n = 2 sections from 5 mice per diet group. C) c-Met mRNA expression was determined using qPCR analysis in tumor tissues for n = 7. D) Representative photomicrographs (40X) of the IHC tumor analysis of membrane localized c-Met staining in negative controls and 10%, 45%, and 60% diet groups (arrows). E) Total c-Met protein levels in tumors were measured using the Aperio ImageScope membrane algorithm and quantified as above. F) Western immunoblot expression of c-Met in n=3 tumors per diet group and quantification (c-Met normalized to amido black staining loading control). G) Representative photomicrographs (40X) of the IHC tumor analysis of pc-Met staining in negative controls and 10%, 45%, and 60% diet groups. H) Total pc-Met protein levels in tumors were measured using the Aperio ImageScope color deconvolution algorithm and quantified as above.

Analysis of metabolic parameters revealed that mice fed 45% and 60% diets were significantly heavier compared to mice on 10% diet (Figure 4A, P<0.05). Significant differences in weight could not be detected after week 12 on diet because the sample size was reduced as mice were sacrificed after tumor formation. Obese mice had greater fat mass compared to mice fed 10% at 4 weeks on diet (Figure 4B; 45% P=0.009 and 60% P=0.007 versus 10%), and at 8 weeks (45% P=0.0001 and 60% P=0.0002 versus 10%). Leptin is a marker of adiposity and increases with fat accretion [41,2]. Mice fed 45% (P=0.005) and 60% (P=0.003) diets exhibited elevated leptin concentrations measured at sacrifice compared to mice on 10% diet (Figure 4C). Of note, decreased leptin concentrations of obese mice fed 60% diets compared to 45%-fed mice may be due to disease status of the mice as these levels were measured at sacrifice after tumor onset, when tumor-induced cachexia may occur [42-44].

Figure 4. Diet affects body weight, body composition, and plasma leptin concentration but not metabolic parameters.

Figure 4

A) Body weight increased in proportion with fat content in diet. * 10% vs. 45% (P <0.05) over weeks 3 - 12 (solid line) and ^ 10% vs. 60% (P < 0.05) over weeks 2 - 12 (dashed line). N= 15 mice per group at start of diet. B) Percent fat content relative to total body mass is shown for n = 15. At 4 weeks on diet * 10% vs. 45% (P=0.009) and 10% vs. 60% (P=0.007) and at 8 weeks on diets ^ 10% vs. 45% (P=0.0001) and 10% vs. 60% (P=0.0002). C) Leptin levels in plasma at sacrifice were measured (n = 12). * 10% vs. 45% (P=0.005) and 10% vs. 60% (P=0.003). D) Blood glucose concentrations prior to start of diet and at sacrifice were measured in 6 hour fasted mice (n = 15 mice). E) Insulin in plasma at sacrifice was measured (n = 12). F) Homeostasis model assessment of insulin resistance (HOMAIR) at sacrifice (n = 12).

Alterations in tumor onset are likely not due to changes in metabolic parameters since glucose, insulin or HOMAIR did not vary by obesity (Figures 4D-F). Inflammatory proteins were measured in plasma, normal mammary, and tumors. Obese mice fed 45% (P=0.023) and 60% (P=0.048) diets exhibited an increase in plasma TNF-α concentrations compared to mice on 10% diet (Figure 5A). However, no significant differences were observed in TNF-α concentrations in the normal mammary gland or tumors. Similarly, plasma and tissue concentrations of IL-6 and MCP-1 were not significantly modified by obesity (Figures 5B-C). In addition, plasma concentrations of hormones including estrogen and progesterone were not altered by obesity (Figures 5 D-E). Finally, aromatase (Cyp19a1) expression was not detectable by qPCR in any of the diet groups in normal mammary gland or in tumors (data not shown). Epididymal adipose tissue was used as a positive control: obese adipose expressed high levels of aromatase compared to lean controls (data not shown).

Figure 5. Plasma levels of TNF-α, but not IL-6 or MCP-1, nor tissue levels of cytokines are regulated by obesity.

Figure 5

Figure 5

Cytokine and chemokine levels of A) TNF-α, B) IL-6 and C) MCP-1 in plasma, normal mammary gland, and tumor tissues; or levels of hormones D) estrogen and E) progesterone were measured at sacrifice (n=12 for plasma samples, n=4 for normal mammary glands and n=5 for tumors). * TNF-α plasma levels: 10% vs. 45% (P = 0.023 and 10% vs. 60% (P = 0.048).

Obesity and tumor associated fibroblast-derived HGF drove basal-like cell proliferation and migration

HGF was detected in tissue fibroblasts from normal mammary glands and tumor tissue (Figure 6A). HGF secretion and function was next investigated in primary fibroblasts. Secretion of HGF over various time points from 4T1 cells remained low over 24 hours of mono-culture (Figure 6A). Secretion of HGF from all primary fibroblasts groups at 20 and 24 hours in mono-culture was significantly elevated over earlier time points and 4T1 cells. Interestingly, fibroblasts isolated from obese mice secreted significantly greater HGF compared to fibroblasts isolated from lean mice. In addition, within a given diet, CAFs secreted greater HGF compared to NAFs. HGF secreted from 10%-fed NAFs was significantly lower than all other groups (P=0.001), and secretion from 60%-fed CAFs was elevated compared to 10% NAF, 60% NAF, and 10% CAF (P=0.0001, P=0.005, and P=0.043, respectively). HGF secreted from fibroblasts activated the c-Met signaling cascade in 4T1 cells similar to recombinant murine HGF (Figure 6C).

Figure 6. HGF mediates wound closure driven by obese and/or cancer-derived fibroblasts.

Figure 6

Figure 6

A) Representative confocal photomicrographs (63X) of HGF (green) co-stained with fibroblast marker - smooth muscle actin (SMA, red) in normal mammary and tumor. Nucleus (blue) is stained with DAPI. B) HGF was quantified in conditioned media at indicated time points from mono-cultures of 4T1 basal like epithelial cells, as well as primary normal associated fibroblasts (NAF) and cancer associated fibroblasts (CAF) isolated from lean (10%-fed mice) and obese (60%-fed mice) for n=3 experiments. *P = 0.0001 vs. all other groups; + vs. all other groups (P=0.001); # vs. 10% NAF (P=0.0001), 60% NAF (P=0.005), and 10% CAF (P=0.043). C) phospho-c-Met and total c-Met expression in 4T1 cells was determined using Western immunoblot analysis after 15 min treatment with conditioned media (CM) from primary 60% CAFs or recombinant mouse HGF (50 ng/ml). Loading control is non-specific band from c-Met immunoblot. D) Proliferation index measured using BrdU incorporation is shown for n = 3 experimental groups. * vs. HGF (50 ng/ml) (P = 0.024); # vs. 60%-CAF with 10%-NAF (P = 0.003) and 60% NAF (P = 0.02). E) Fluorescently labeled 4T1 cells were co-cultured with NAFs or CAFs isolated from lean (10% diet) and obese (60% diet) mice, with or without HGF neutralizing antibody during the 24 hour co-culture period and the 12 hour scratch test. Representative photomicrographs of wound migration assay at 0h (time point of scratch) and 12h (end of study). F) Percent wound closure is shown for n = 3 experiments. * vs. control (no anti-HGF) for 10% CAF (P=0.0001) and 60% CAF (P=0.0004). + vs. 10% NAF (P=0.0002) and vs. 60%-NAF (P=0.006) controls. # vs. all groups (P = 0.0001).

Based on an elevation of HGF with obesity in vivo and ex vivo, we assayed cellular proliferation and migration associated with fibroblast-derived HGF signaling [23]. In parallel with HGF concentrations from primary NAF and CAFs (Figure 6B), conditioned media from NAFs and CAFs drove 4T1 proliferation as measured by BrdU incorporation. The greatest proliferation was driven by fibroblasts isolated from tumors from 60%-fed mice (Figure 6D, P=0.003 versus 10% NAFs and P=0.02 versus 60% NAFs). The extent of proliferation was similar to 50ng/ml recombinant HGF-stimulated 4T1 proliferation. Cell counts revealed identical proliferative capacity (data not shown).

Finally, to test the effect of stromal-epithelial interactions, primary fibroblasts were cocultured with 4T1 cells and a scratch test “wound assay” was conducted to examine migratory capacity. In parallel with elevated HGF levels in fibroblast-conditioned media (Figure 6B) and effects on proliferation (Figure 6D), CAFs demonstrated a significantly greater effect on wound closure than NAFs in both cases where fibroblasts were isolated from lean and obese mice (Figure 6E and F). The greatest percent wound closure occurred when 4T1 cells were co-cultured with 60% CAFs compared to all other groups (60% CAF vs. 10% NAF: P=0.0001 and 60% NAF: P=0.006). Effects of fibroblasts-epithelial coculture were significantly reversed in the CAF-4T1 model by an HGF neutralizing antibody (Figure 6E and F, 10% CAF: P=0.0001 and 60% CAF: P=0.000004 versus untreated). Anti-HGF antibody treatment demonstrated the greatest inhibition of wound closure in 4T1s co-cultured with 60% CAFs (P=0.0001 vs. all other groups).

DISCUSSION

Genetic characteristics of BBC have been well-defined, including loss of retinoblastoma (Rb), p53 mutation, BRCA1 mutation, and signaling defects [10]. However, stromal interactions likely play a contributing role in BBC etiology, and may underlie higher rates of local recurrence for this subtype. Fibroblasts are an abundant stromal cell type in the tumor microenvironment and play a fundamental role in the etiology and progression of tumors [19,45]. Stromal cell interactions are important in tumor cell proliferation, macrophage polarization, as well as in reorganization of the extracellular matrix which contributes to metastasis [46,47,23,48,49,45]. While our previous work using ex vivo co-culture models suggest unique interactions between human fibroblasts and BBC cells [23,49,50], stromal-epithelial interactions early in carcinogenesis are poorly understood. Herein we demonstrate a plausible role of obesity-modulated fibroblast-derived growth factor expression in normal mammary gland, with implications for etiology of BBC.

Both in vivo and ex vivo, we demonstrated that stroma-derived HGF expression is associated with obesity in a pre-clinical model of BBC. Normal mammary glands exhibit obesity-induced increases in HGF and c-Met expression as well as phosphorylated c-Met, providing an optimal microenvironment for tumorigenesis. Indeed, obesity significantly decreased tumor latency. Interestingly, decreases in latency occurred in the absence of changes in either tumor burden or tumor volume. It is possible that the increased HGF and c-Met expression detected in the normal mammary glands prior to tumor onset act to mediate tumor etiology in C3(1)-TAg mice. c-Met signaling causes luminal progenitors to preferentially undergo basal lineage commitment at the expense of luminal cell-fate specification [25]. There is little work completed on basal-like murine models and to date only one prior publication from our group has indicated c-Met in basal-like murine tumors [24]. Evidence presented herein that obesity strongly regulates HGF/c-Met suggests a novel targetable pathway.

Our data suggest that the effects on BBC etiology include a role for HGF/c-Met and ruled out metabolic effects as significant mediators of this association. This is important because obesity has been hypothesized to play a role in carcinogenesis through altered metabolic homeostasis, including hyperglycemia and hyperinsulinemia secondary to insulin resistance [51]. Although we observed increased body weight and adiposity in the obese groups relative to lean controls, obese mice did not develop hyperglycemia, hyperinsulinemia, or evidence of insulin resistance. Leptin was elevated by obesity in C3(1)-TAg mice. Leptin is mitogenic, anti-apoptotic, pro-angiogenic, and pro-inflammatory, and, thus, is implicated in the stimulation, migration, and invasion of tumor cells [52,53,2]. Further studies to determine the role of leptin in BBC etiology need to be completed. Although significant increases in systemic TNF- in obese mice were observed, local levels of all cytokines and chemokines measured indicated that important pro-inflammatory mediators (TNF-α, IL-6 and MCP-1) were not altered by obesity, hence did not contribute to tumor onset in in C3(1)-TAg mice in this experimental design. However, our previous work and A.J. Dannenberg’s studies have demonstrated that both obese women and murine models display elevated macrophage infiltration in normal breast tissue [54,55,22]. Future studies examining the effects of local inflammation and macrophages on BBC are needed to clarify the time course of microenvironmental alterations and obesity-mediated tumor onset.

In epidemiologic studies that have examined obesity risk among all breast cancers or among cancers stratified by ER status, risk of breast cancer was elevated only among postmenopausal women [6] leading some to hypothesize that elevated aromatase may play a role. However, several more recent studies have shown that obesity and adiposity may increase risk for triple negative breast cancers [4]. In the triple negative subgroup, a prominent role for aromatase and estrogen signaling is less likely; however we investigated systemic hormones concentrations and aromatase expression in our model to rule out any hormonal effects. The lack of obesity-mediated differences in plasma estrogen and progesterone concentrations, and absence of aromatase expression in the mammary gland or tumors, suggests that the latency was not driven by systemic or local hormone concentrations. Furthermore, it has been reported that estrogen levels in C3(1)-TAg mice do not affect the tumor growth and progression as shown by Yoshidome et. al. [56]. Similarly, we demonstrated a lack of significant elevations in SV40 TAg levels, indicating that obesity-mediated alterations in transgene expression could not explain detected reductions in tumor latency. Therefore, metabolic and hormonal action did not appear to be the primary drivers of obesity-mediated alterations in latency.

An intriguing finding was the persistent ex vivo release of HGF from fibroblasts. In vivo, HGF was localized to fibroblasts in both normal mammary glands and tumor tissues. HGF secretion was significantly increased from NAFs from obese mice compared to lean, demonstrating that in vivo obesity-induced elevation of HGF expression is a phenotype that is conserved in primary culture. In addition, CAFs derived from mice on either diet secreted significantly greater HGF than NAFs, demonstrating that the tumor microenvironment primes for elevated HGF release from fibroblasts. Conditioned media from fibroblasts induced cell proliferation in direct correlation with HGF concentrations secreted from the NAFs and CAFs, with obese-derived tumor fibroblast conditioned media being as effective as recombinant HGF in driving epithelial cell proliferation. Obesity regulation of HGF/c-Met driven proliferation is one mechanism demonstrated in vitro that may be an underlying mechanism in vivo.

To probe the HGF/c-Met mechanism in a faithful model of the mammary microenvironment, we cocultured primary fibroblasts from C3(1)-TAg diet-exposed animals with 4T1 BBC-like epithelial cancer cells [23,46,54] and completed a scratch test wound response assay. Earlier work by our group demonstrated a strong wound response signature in more aggressive and basal-like breast cancer samples from patients, as well as in the normal mammary gland extra-tumoral microenvironment [45,48,47,50.]. In fibroblasts/4T1 co-culture herein, completeness of wound closure was elevated in association with HGF expression increases, such that fibroblasts isolated from obese mice and/or from tumors demonstrated the greatest extent of wound closure. Since HGF is the only known ligand of c-Met, inhibiting HGF signaling using a neutralizing antibody is effective at blunting c-Met activation [57,49,50]. Our results demonstrated that neutralization of HGF had the most dramatic effects in conditions where fibroblast-driven proliferation was the greatest, demonstrating specificity for HGF/c-Met signaling. The persistence of HGF secretion from primary fibroblasts ex vivo is consistent with work demonstrating that HGF is secreted in greater amounts from primary adipocytes isolated from obese versus lean subjects [27]. Our work presented here suggest that fibroblast-derived HGF may be regulated by obesity through epigenetic means [58], which could have long-lasting effects on the mammary gland stroma.

CONCLUSIONS

In sum, our data demonstrate that obesity reduces BBC latency in C3(1)-TAg mice. HGF may be a potential mediator of tumor onset: expression is elevated by obesity in normal mammary glands and persists in isolated primary fibroblasts. Indeed, in vitro modeling of the normal and tumor microenvironment demonstrate that fibroblasts derived from tumors were an important regulator of proliferation and wound response, specifically through HGF/c-Met signaling. Increased proliferation and motility induced by HGF allows for cells with already elevated proliferative rates (such as the TAg-overexpressing epithelium of this model) to locally invade and develop tumors. c-Met inhibitors are currently in clinical trials for basal-like breast cancer [59]. Our findings support a novel role for obesity-mediated HGF/c-Met signaling as modifiable risk factor for basal-like tumor onset, which has important public health implications with regard to BBC risk. Whether increased risk of breast cancer associated with obesity is reversible with weight loss is still an important area of uncertainty.

Supplementary Material

10549_2013_2741_MOESM1_ESM

ACKNOWLEDGEMENTS

We thank the directors and personnel of the UNC LCCC Mouse Phase One Unit (MP1U) for assistance with animal handling, therapeutic studies, and tumor serial assessment. Work in the MP1U was supported by the University Cancer Research Fund. This publication was made possible by the Breast Cancer and the Environment Research Program (BCERP) award number U01ES019472 from the National Institute of Environmental Health Sciences (NIEHS) and the National Cancer Institute (NCI), NIH, DHHS.

FINANCIAL SUPPORT: LM was supported by UNC University Cancer Research Fund, NIH AA017376; NIH ES019472; NIH P30DK056350-Nutrition Obesity Research Consortium (NORC); NIH P30DK034987 Center for GI Biology and Disease. MAT was supported by NIH ES019472 and RO1-CA138255. JAG was supported by NIH grants DK034987 and DK056350. CMP was supported by funds from the NCI Breast SPORE program (P50-CA58223-09A1), by RO1-CA138255 and RO1-CA148761

LIST OF ABBREVIATIONS

BBC

Basal-like breast cancer

HGF

Hepatocyte growth factor

BMI

Body mass index

ER

estrogen receptor

PR

progesterone receptor

HER2

Human epidermal growth factor – 2

NAF

Normal associated fibroblasts

CAF

Cancer associated fibroblasts

Footnotes

CONFLICT OF INTEREST: None

REFERENCES

  • 1.Thiebaut AC, Kipnis V, Chang SC, Subar AF, Thompson FE, Rosenberg PS, Hollenbeck AR, Leitzmann M, Schatzkin A. Dietary fat and postmenopausal invasive breast cancer in the National Institutes of Health-AARP Diet and Health Study cohort. Journal of the National Cancer Institute. 2007;99(6):451–462. doi: 10.1093/jnci/djk094. doi:10.1093/jnci/djk094. [DOI] [PubMed] [Google Scholar]
  • 2.Sundaram S, Johnson AR, Makowski L. Obesity, metabolism and the microenvironment: Links to cancer. J Carcinog. 2013;12(19) doi: 10.4103/1477-3163.119606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cecchini RS, Costantino JP, Cauley JA, Cronin WM, Wickerham DL, Land SR, Weissfeld JL, Wolmark N. Body mass index and the risk for developing invasive breast cancer among high-risk women in NSABP P-1 and STAR breast cancer prevention trials. Cancer Prev Res (Phila) 2012;5(4):583–592. doi: 10.1158/1940-6207.CAPR-11-0482. doi:10.1158/1940-6207.CAPR-11-0482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Anderson GL, Neuhouser ML. Obesity and the Risk for Premenopausal and Postmenopausal Breast Cancer. Cancer Prevention Research. 2012;5(4):515–521. doi: 10.1158/1940-6207.CAPR-12-0091. doi:10.1158/1940-6207.capr-12-0091. [DOI] [PubMed] [Google Scholar]
  • 5.Yang XR, Chang-Claude J, Goode EL, Couch FJ, Nevanlinna H, Milne RL, Gaudet M, Schmidt MK, Broeks A, Cox A, Fasching PA, Hein R, Spurdle AB, Blows F, Driver K, Flesch-Janys D, Heinz J, Sinn P, Vrieling A, Heikkinen T, Aittomaki K, Heikkila P, Blomqvist C, Lissowska J, Peplonska B, Chanock S, Figueroa J, Brinton L, Hall P, Czene K, Humphreys K, Darabi H, Liu J, Van ’t Veer LJ, van Leeuwen FE, Andrulis IL, Glendon G, Knight JA, Mulligan AM, O’Malley FP, Weerasooriya N, John EM, Beckmann MW, Hartmann A, Weihbrecht SB, Wachter DL, Jud SM, Loehberg CR, Baglietto L, English DR, Giles GG, McLean CA, Severi G, Lambrechts D, Vandorpe T, Weltens C, Paridaens R, Smeets A, Neven P, Wildiers H, Wang X, Olson JE, Cafourek V, Fredericksen Z, Kosel M, Vachon C, Cramp HE, Connley D, Cross SS, Balasubramanian SP, Reed MW, Dork T, Bremer M, Meyer A, Karstens JH, Ay A, Park-Simon TW, Hillemanns P, Arias Perez JI, Menendez Rodriguez P, Zamora P, Benitez J, Ko YD, Fischer HP, Hamann U, Pesch B, Bruning T, Justenhoven C, Brauch H, Eccles DM, Tapper WJ, Gerty SM, Sawyer EJ, Tomlinson IP, Jones A, Kerin M, Miller N, McInerney N, Anton-Culver H, Ziogas A, Shen CY, Hsiung CN, Wu PE, Yang SL, Yu JC, Chen ST, Hsu GC, Haiman CA, Henderson BE, Le Marchand L, Kolonel LN, Lindblom A, Margolin S, Jakubowska A, Lubinski J, Huzarski T, Byrski T, Gorski B, Gronwald J, Hooning MJ, Hollestelle A, van den Ouweland AM, Jager A, Kriege M, Tilanus-Linthorst MM, Collee M, Wang-Gohrke S, Pylkas K, Jukkola-Vuorinen A, Mononen K, Grip M, Hirvikoski P, Winqvist R, Mannermaa A, Kosma VM, Kauppinen J, Kataja V, Auvinen P, Soini Y, Sironen R, Bojesen SE, Orsted DD, Kaur-Knudsen D, Flyger H, Nordestgaard BG, Holland H, Chenevix-Trench G, Manoukian S, Barile M, Radice P, Hankinson SE, Hunter DJ, Tamimi R, Sangrajrang S, Brennan P, McKay J, Odefrey F, Gaborieau V, Devilee P, Huijts PE, Tollenaar RA, Seynaeve C, Dite GS, Apicella C, Hopper JL, Hammet F, Tsimiklis H, Smith LD, Southey MC, Humphreys MK, Easton D, Pharoah P, Sherman ME, Garcia-Closas M. Associations of breast cancer risk factors with tumor subtypes: a pooled analysis from the Breast Cancer Association Consortium studies. Journal of the National Cancer Institute. 2011;103(3):250–263. doi: 10.1093/jnci/djq526. doi:10.1093/jnci/djq526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kapur K, Jiang H, Xing Y, Wong W. Cross-hybridization modeling on Affymetrix exon arrays. Bioinformatics. 2008;24:2887–2893. doi: 10.1093/bioinformatics/btn571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Calle EE, Kaaks R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nature reviews Cancer. 2004;4(8):579–591. doi: 10.1038/nrc1408. doi:10.1038/nrc1408. [DOI] [PubMed] [Google Scholar]
  • 8.Reeves KW, Faulkner K, Modugno F, Hillier TA, Bauer DC, Ensrud KE, Cauley JA. Body mass index and mortality among older breast cancer survivors in the Study of Osteoporotic Fractures. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2007;16(7):1468–1473. doi: 10.1158/1055-9965.EPI-07-0051. doi:10.1158/1055-9965.EPI-07-0051. [DOI] [PubMed] [Google Scholar]
  • 9.Sieri S, Krogh V, Ferrari P, Berrino F, Pala V, Thiebaut AC, Tjonneland A, Olsen A, Overvad K, Jakobsen MU, Clavel-Chapelon F, Chajes V, Boutron-Ruault MC, Kaaks R, Linseisen J, Boeing H, Nothlings U, Trichopoulou A, Naska A, Lagiou P, Panico S, Palli D, Vineis P, Tumino R, Lund E, Kumle M, Skeie G, Gonzalez CA, Ardanaz E, Amiano P, Tormo MJ, Martinez-Garcia C, Quiros JR, Berglund G, Gullberg B, Hallmans G, Lenner P, Bueno-de-Mesquita HB, van Duijnhoven FJ, Peeters PH, van Gils CH, Key TJ, Crowe FL, Bingham S, Khaw KT, Rinaldi S, Slimani N, Jenab M, Norat T, Riboli E. Dietary fat and breast cancer risk in the European Prospective Investigation into Cancer and Nutrition. The American journal of clinical nutrition. 2008;88(5):1304–1312. doi: 10.3945/ajcn.2008.26090. [DOI] [PubMed] [Google Scholar]
  • 10.Toft DJ, Cryns VL. Minireview: Basal-Like Breast Cancer: From Molecular Profiles to Targeted Therapies. Molecular Endocrinology. 2011;25(2):199–211. doi: 10.1210/me.2010-0164. doi:10.1210/me.2010-0164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Carey LA, Perou CM, Livasy CA, Dressler LG, Cowan D, Conway K, Karaca G, Troester MA, Tse CK, Edmiston S, Deming SL, Geradts J, Cheang MC, Nielsen TO, Moorman PG, Earp HS, Millikan RC. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA: the journal of the American Medical Association. 2006;295(21):2492–2502. doi: 10.1001/jama.295.21.2492. doi:10.1001/jama.295.21.2492. [DOI] [PubMed] [Google Scholar]
  • 12.Millikan RC, Newman B, Tse CK, Moorman PG, Conway K, Dressler LG, Smith LV, Labbok MH, Geradts J, Bensen JT, Jackson S, Nyante S, Livasy C, Carey L, Earp HS, Perou CM. Epidemiology of basal-like breast cancer. Breast cancer research and treatment. 2008;109(1):123–139. doi: 10.1007/s10549-007-9632-6. doi:10.1007/s10549-007-9632-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Boggs DA, Rosenberg L, Cozier YC, Wise LA, Coogan PF, Ruiz-Narvaez EA, Palmer JR. General and abdominal obesity and risk of death among black women. The New England journal of medicine. 2011;365(10):901–908. doi: 10.1056/NEJMoa1104119. doi:10.1056/NEJMoa1104119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Keppel KG, Taffel SM. Pregnancy-related weight gain and retention: implications of the 1990 Institute of Medicine guidelines. American journal of public health. 1993;83(8):1100–1103. doi: 10.2105/ajph.83.8.1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Olson LK, Tan Y, Zhao Y, Aupperlee MD, Haslam SZ. Pubertal exposure to high fat diet causes mouse strain-dependent alterations in mammary gland development and estrogen responsiveness. Int J Obes (Lond) 2010;34(9):1415–1426. doi: 10.1038/ijo.2010.51. doi:10.1038/ijo.2010.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hue-Beauvais C, Chavatte-Palmer P, Aujean E, Dahirel M, Laigre P, Pechoux C, Bouet S, Devinoy E, Charlier M. An obesogenic diet started before puberty leads to abnormal mammary gland development during pregnancy in the rabbit. Developmental dynamics: an official publication of the American Association of Anatomists. 2011;240(2):347–356. doi: 10.1002/dvdy.22536. doi:10.1002/dvdy.22536. [DOI] [PubMed] [Google Scholar]
  • 17.Gordon RR, Hunter KW, La Merrill M, Sorensen P, Threadgill DW, Pomp D. Genotype X diet interactions in mice predisposed to mammary cancer: II. Tumors and metastasis. Mammalian genome: official journal of the International Mammalian Genome Society. 2008;19(3):179–189. doi: 10.1007/s00335-008-9096-y. doi:10.1007/s00335-008-9096-y. [DOI] [PubMed] [Google Scholar]
  • 18.Flegal KM, Carroll MD, Kit BK, Ogden CL. Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999-2010. JAMA: the journal of the American Medical Association. 2012;307(5):491–497. doi: 10.1001/jama.2012.39. doi:10.1001/jama.2012.39. [DOI] [PubMed] [Google Scholar]
  • 19.Casbas-Hernandez P, Fleming JM, Troester M. Gene Expression Analysis of In Vitro Cocultures to Study Interactions between Breast Epithelium and Stroma. Journal of Biomedicine and Biotechnology. 2011 doi: 10.1155/2011/520987. 2011. doi:10.1155/2011/520987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Le TT, Rehrer CW, Huff TB, Nichols MB, Camarillo IG, Ji-Xin C. Nonlinear Optical Imaging to Evaluate the Impact of Obesity on Mammary Gland and Tumor Stroma. Molecular Imaging. 2007;6(3):205–204. doi:10.2310/7290.2007.00018. [PMC free article] [PubMed] [Google Scholar]
  • 21.Sinicrope FA, Dannenberg AJ. Obesity and breast cancer prognosis: weight of the evidence. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2011;29(1):4–7. doi: 10.1200/JCO.2010.32.1752. doi:10.1200/JCO.2010.32.1752. [DOI] [PubMed] [Google Scholar]
  • 22.Subbaramaiah K, Howe LR, Bhardwaj P, Du B, Gravaghi C, Yantiss RK, Zhou XK, Blaho VA, Hla T, Yang P, Kopelovich L, Hudis CA, Dannenberg AJ. Obesity is associated with inflammation and elevated aromatase expression in the mouse mammary gland. Cancer Prev Res (Phila) 2011;4(3):329–346. doi: 10.1158/1940-6207.CAPR-10-0381. doi:10.1158/1940-6207.CAPR-10-0381. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 23.Camp JT, Elloumi F, Roman-Perez E, Rein J, Stewart DA, Harrell JC, Perou CM, Troester MA. Interactions with fibroblasts are distinct in Basal-like and luminal breast cancers. Molecular cancer research: MCR. 2011;9(1):3–13. doi: 10.1158/1541-7786.MCR-10-0372. doi:10.1158/1541-7786.MCR-10-0372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Xu K, Usary J, Kousis Philaretos C, Prat A, Wang D-Y, Adams Jessica R, Wang W, Loch Amanda J, Deng T, Zhao W, Cardiff Robert D, Yoon K, Gaiano N, Ling V, Beyene J, Zacksenhaus E, Gridley T, Leong Wey L, Guidos Cynthia J, Perou Charles M, Egan Sean E. Lunatic Fringe Deficiency Cooperates with the Met/Caveolin Gene Amplicon to Induce Basal-like Breast Cancer. Cancer cell. 2012;21(5):626–641. doi: 10.1016/j.ccr.2012.03.041. doi:10.1016/j.ccr.2012.03.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gastaldi S, Sassi F, Accornero P, Torti D, Galimi F, Migliardi G, Molyneux G, Perera T, Comoglio PM, Boccaccio C, Smalley MJ, Bertotti A, Trusolino L. Met signaling regulates growth, repopulating potential and basal cell-fate commitment of mammary luminal progenitors: implications for basal-like breast cancer. Oncogene. 2012 doi: 10.1038/onc.2012.154. doi: http://www.nature.com/onc/journal/vaop/ncurrent/suppinfo/onc2012154s1.html. [DOI] [PubMed] [Google Scholar]
  • 26.Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, Pell CL, Johnstone BH, Considine RV, March KL. Secretion of Angiogenic and Antiapoptotic Factors by Human Adipose Stromal Cells. Circulation. 2004;109(10):1292–1298. doi: 10.1161/01.CIR.0000121425.42966.F1. doi:10.1161/01.cir.0000121425.42966.f1. [DOI] [PubMed] [Google Scholar]
  • 27.Bell LN, Ward JL, Degawa-Yamauchi M, Bovenkerk JE, Jones R, Cacucci BM, Gupta CE, Sheridan C, Sheridan K, Shankar SS, Steinberg HO, March KL, Considine RV. Adipose tissue production of hepatocyte growth factor contributes to elevated serum HGF in obesity. American Journal of Physiology - Endocrinology And Metabolism. 2006;291(4):E843–E848. doi: 10.1152/ajpendo.00174.2006. doi:10.1152/ajpendo.00174.2006. [DOI] [PubMed] [Google Scholar]
  • 28.Toi M, Taniguchi T, Ueno T, Asano M, Funata N, Sekiguchi K, Iwanari H, Tominaga T. Significance of circulating hepatocyte growth factor level as a prognostic indicator in primary breast cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 1998;4(3):659–664. [PubMed] [Google Scholar]
  • 29.Sheen-Chen SM, Liu YW, Eng HL, Chou FF. Serum levels of hepatocyte growth factor in patients with breast cancer. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2005;14(3):715–717. doi: 10.1158/1055-9965.EPI-04-0340. doi:10.1158/1055-9965.EPI-04-0340. [DOI] [PubMed] [Google Scholar]
  • 30.Taniguchi T, Toi M, Inada K, Imazawa T, Yamamoto Y, Tominaga T. Serum concentrations of hepatocyte growth factor in breast cancer patients. Clinical cancer research: an official journal of the American Association for Cancer Research. 1995;1(9):1031–1034. [PubMed] [Google Scholar]
  • 31.Maiti B, Kundranda MN, Spiro TP, Daw HA. The association of metabolic syndrome with triple-negative breast cancer. Breast cancer research and treatment. 2010;121(2):479–483. doi: 10.1007/s10549-009-0591-y. doi:10.1007/s10549-009-0591-y. [DOI] [PubMed] [Google Scholar]
  • 32.Green JE, Shibata MA, Yoshidome K, Liu ML, Jorcyk C, Anver MR, Wigginton J, Wiltrout R, Shibata E, Kaczmarczyk S, Wang W, Liu ZY, Calvo A, Couldrey C. The C3(1)/SV40 T-antigen transgenic mouse model of mammary cancer: ductal epithelial cell targeting with multistage progression to carcinoma. Oncogene. 2000;19(8):1020–1027. doi: 10.1038/sj.onc.1203280. doi:10.1038/sj.onc.1203280. [DOI] [PubMed] [Google Scholar]
  • 33.Herschkowitz JI, Simin K, Weigman VJ, Mikaelian I, Usary J, Hu Z, Rasmussen KE, Jones LP, Assefnia S, Chandrasekharan S, Backlund MG, Yin Y, Khramtsov AI, Bastein R, Quackenbush J, Glazer RI, Brown PH, Green JE, Kopelovich L, Furth PA, Palazzo JP, Olopade OI, Bernard PS, Churchill GA, Van Dyke T, Perou CM. Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome biology. 2007;8(5):R76. doi: 10.1186/gb-2007-8-5-r76. doi:10.1186/gb-2007-8-5-r76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sampey BP, Vanhoose AM, Winfield HM, Freemerman AJ, Muehlbauer MJ, Fueger PT, Newgard CB, Makowski L. Cafeteria diet is a robust model of human metabolic syndrome with liver and adipose inflammation: comparison to high-fat diet. Obesity (Silver Spring) 2011;19(6):1109–1117. doi: 10.1038/oby.2011.18. doi:10.1038/oby.2011.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fleming JM, Miller TC, Kidacki M, Ginsburg E, Stuelten CH, Stewart DA, Troester MA, Vonderhaar BK. Paracrine interactions between primary human macrophages and human fibroblasts enhance murine mammary gland humanization in vivo. Breast cancer research: BCR. 2012;14(3):R97. doi: 10.1186/bcr3215. doi:10.1186/bcr3215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sampey BP, Freemerman AJ, Zhang J, Kuan PF, Galanko JA, O’Connell TM, Ilkayeva OR, Muehlbauer MJ, Stevens RD, Newgard CB, Brauer HA, Troester MA, Makowski L. Metabolomic profiling reveals mitochondrial-derived lipid biomarkers that drive obesity-associated inflammation. PloS one. 2012;7(6):e38812. doi: 10.1371/journal.pone.0038812. doi:10.1371/journal.pone.0038812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ponzo MG, Park M. The Met receptor tyrosine kinase and basal breast cancer. Cell Cycle. 2010;9(6):1043–1050. doi: 10.4161/cc.9.6.11033. [DOI] [PubMed] [Google Scholar]
  • 38.Moghul A, Lin L, Beedle A, Kanbour-Shakir A, DeFrances MC, Liu Y, Zarnegar R. Modulation of c-MET proto-oncogene (HGF receptor) mRNA abundance by cytokines and hormones: evidence for rapid decay of the 8 kb c-MET transcript. Oncogene. 1994;9(7):2045–2052. [PubMed] [Google Scholar]
  • 39.Blumenschein GR, Mills GB, Gonzalez-Angulo AM. Targeting the Hepatocyte Growth Factor-cMET Axis in Cancer Therapy. Journal of Clinical Oncology. 2012;30(26):3287–3296. doi: 10.1200/JCO.2011.40.3774. doi:10.1200/jco.2011.40.3774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jagadeeswaran R, Ma PC, Seiwert TY, Jagadeeswaran S, Zumba O, Nallasura V, Ahmed S, Filiberti R, Paganuzzi M, Puntoni R, Kratzke RA, Gordon GJ, Sugarbaker DJ, Bueno R, Janamanchi V, Bindokas VP, Kindler HL, Salgia R. Functional Analysis of c-Met/Hepatocyte Growth Factor Pathway in Malignant Pleural Mesothelioma. Cancer research. 2006;66(1):352–361. doi: 10.1158/0008-5472.CAN-04-4567. doi:10.1158/0008-5472.can-04-4567. [DOI] [PubMed] [Google Scholar]
  • 41.Johnson AR, Justin Milner J, Makowski L. The inflammation highway: metabolism accelerates inflammatory traffic in obesity. Immunological Reviews. 2012;249(1):218–238. doi: 10.1111/j.1600-065X.2012.01151.x. doi:10.1111/j.1600-065X.2012.01151.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wolf I, Sadetzki S, Kanety H, Kundel Y, Pariente C, Epstein N, Oberman B, Catane R, Kaufman B, Shimon I. Adiponectin, ghrelin, and leptin in cancer cachexia in breast and colon cancer patients. Cancer. 2006;106(4):966–973. doi: 10.1002/cncr.21690. doi:10.1002/cncr.21690. [DOI] [PubMed] [Google Scholar]
  • 43.Engineer DR, Garcia JM. Leptin in Anorexia and Cachexia Syndrome. International Journal of Peptides. 2012 doi: 10.1155/2012/287457. 2012. doi:10.1155/2012/287457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Smiechowska J, Utech A, Taffet G, Hayes T, Marcelli M, Garcia JM. Adipokines in patients with cancer anorexia and cachexia. Journal of investigative medicine: the official publication of the American Federation for Clinical Research. 2010;58(3):554–559. doi: 10.231/JIM.0b013e3181cf91ca. doi:10.231/JIM.0b013e3181cf91ca. [DOI] [PubMed] [Google Scholar]
  • 45.Troester MA, Lee MH, Carter M, Fan C, Cowan DW, Perez ER, Pirone JR, Perou CM, Jerry DJ, Schneider SS. Activation of Host Wound Responses in Breast Cancer Microenvironment. Clinical Cancer Research. 2009;15(22):7020–7028. doi: 10.1158/1078-0432.CCR-09-1126. doi:10.1158/1078-0432.ccr-09-1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Stewart DA, Yang Y, Makowski L, Troester MA. Basal-like Breast Cancer Cells Induce Phenotypic and Genomic Changes in Macrophages. Molecular Cancer Research. 2012;10(6):727–738. doi: 10.1158/1541-7786.MCR-11-0604. doi:10.1158/1541-7786.mcr-11-0604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sun X, Gierach GL, Sandhu R, Williams T, Midkiff BR, Lissowska J, Wesolowska E, Boyd NF, Johnson NB, Figueroa JD, Sherman ME, Troester MA. Relationship of Mammographic Density and Gene Expression: Analysis of Normal Breast Tissue Surrounding Breast Cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2013;19(18):4972–4982. doi: 10.1158/1078-0432.CCR-13-0029. doi:10.1158/1078-0432.CCR-13-0029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Roman-Perez E, Casbas-Hernandez P, Pirone JR, Rein J, Carey LA, Lubet RA, Mani SA, Amos KD, Troester MA. Gene expression in extratumoral microenvironment predicts clinical outcome in breast cancer patients. Breast cancer research: BCR. 2012;14(2):R51. doi: 10.1186/bcr3152. doi:10.1186/bcr3152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Casbas-Hernandez P, DArcy M, Roman-Perez E, Brauer H, McNaughton K, Miller S, Chhetri R, Oldenburg A, Fleming J, Amos K, Makowski L, Troester M. Role of HGF in epithelial-stromal cell interactions during progression from benign breast disease to ductal carcinoma in situ. Breast Cancer Research. 2013;15(5):R82. doi: 10.1186/bcr3476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Brauer HA, Makowski L, Hoadley KA, Casbas-Hernandez P, Lang LJ, Roman-Perez E, D’Arcy M, Freemerman AJ, Perou CM, Troester MA. Impact of tumor microenvironment and epithelial phenotypes on metabolism in breast cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2013;19(3):571–585. doi: 10.1158/1078-0432.CCR-12-2123. doi:10.1158/1078-0432.CCR-12-2123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Davis AA, Kaklamani VG. Metabolic Syndrome and Triple-Negative Breast Cancer: A New Paradigm. International Journal of Breast Cancer. 2012 doi: 10.1155/2012/809291. 2012. doi:10.1155/2012/809291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Paz-Filho G, Lim EL, Wong ML, Licinio J. Associations between adipokines and obesity-related cancer. Frontiers in bioscience: a journal and virtual library. 2011;16:1634–1650. doi: 10.2741/3810. [DOI] [PubMed] [Google Scholar]
  • 53.Saxena NK, Taliaferro-Smith L, Knight BB, Merlin D, Anania FA, O’Regan RM, Sharma D. Bidirectional crosstalk between leptin and insulin-like growth factor-I signaling promotes invasion and migration of breast cancer cells via transactivation of epidermal growth factor receptor. Cancer research. 2008;68(23):9712–9722. doi: 10.1158/0008-5472.CAN-08-1952. doi:10.1158/0008-5472.CAN-08-1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sun X, Casbas-Hernandez P, Bigelow C, Makowski L, Joseph Jerry D, Smith Schneider S, Troester MA. Normal breast tissue of obese women is enriched for macrophage markers and macrophage-associated gene expression. Breast cancer research and treatment. 2012;131(3):1003–1012. doi: 10.1007/s10549-011-1789-3. doi:10.1007/s10549-011-1789-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Morris PG, Hudis CA, Giri D, Morrow M, Falcone DJ, Zhou XK, Du B, Brogi E, Crawford CB, Kopelovich L, Subbaramaiah K, Dannenberg AJ. Inflammation and Increased Aromatase Expression Occur in the Breast Tissue of Obese Women with Breast Cancer. Cancer Prevention Research. 2011;4(7):1021–1029. doi: 10.1158/1940-6207.CAPR-11-0110. doi:10.1158/1940-6207.capr-11-0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yoshidome K, Shibata M-A, Couldrey C, Korach KS, Green JE. Estrogen Promotes Mammary Tumor Development in C3(1)/SV40 Large T-Antigen Transgenic Mice: Paradoxical Loss of Estrogen Receptorα Expression during Tumor Progression. Cancer research. 2000;60(24):6901–6910. [PubMed] [Google Scholar]
  • 57.Cao B, Su Y, Oskarsson M, Zhao P, Kort EJ, Fisher RJ, Wang L-M, Vande Woude GF. Neutralizing monoclonal antibodies to hepatocyte growth factor/scatter factor (HGF/SF) display antitumor activity in animal models. Proceedings of the National Academy of Sciences. 2001;98(13):7443–7448. doi: 10.1073/pnas.131200498. doi:10.1073/pnas.131200498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Martinez-Lopez N, Varela-Rey M, Ariz U, Embade N, Vazquez-Chantada M, Fernandez-Ramos D, Gomez-Santos L, Lu SC, Mato JM, Martinez-Chantar ML. S-adenosylmethionine and proliferation: new pathways, new targets. Biochemical Society transactions. 2008;36(Pt 5):848–852. doi: 10.1042/BST0360848. doi:10.1042/BST0360848. [DOI] [PubMed] [Google Scholar]
  • 59.Clinical Trials Involving HGF/SF-Met Inhibitors The data currently includes three drug categories: anti-HGF/SF antibodies, anti-Met antibodies and Met kinase inhibitors. 2013 http://www.vai.org/metclinicaltrials.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

10549_2013_2741_MOESM1_ESM

RESOURCES