Collagen and prostaglandin E2 regulate aromatase expression through the PI3K/AKT/IKK and the MAP kinase pathways in adipose stromal cells

  • Authors:
    • Ting Tan
    • Lie Wang
    • Bing Wang
  • View Affiliations

  • Published online on: June 9, 2015     https://doi.org/10.3892/mmr.2015.3901
  • Pages: 4766-4772
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Excessive local estrogen production in the breast promotes estrogen-dependent breast cancer. Aromatase is a key enzyme in estrogen biosynthesis. Aromatase inhibitors used in the treatment of breast cancer are very effective, but indiscriminately reduce estrogen synthesis in all tissues, causing major side‑effects. It is thus desirable to develop inhibitors that selectively block aromatase and estrogen production in breast cancer. To this end, it is important to identify the mechanisms by which aromatase is activated in the tumor microenvironment. Prostaglandin E2 (PGE2) and collagen are two important factors in the tumor microenvironment, which contribute to tumor development and progression. In this study, we show that collagen‑induced aromatase expression in adipose stromal cells (ASCs) was significantly reduced by inhibitors of phosphatidylinositide 3‑kinase (PI3K), IκB kinase (IKK), mitogen‑activated protein kinase kinase (MEK), c‑Jun NH2‑terminal kinase (JNK), protein kinase A (PKA), and by the knockdown of the JunB and AKT2 genes. In addition, PGE2‑induced aromatase expression was significantly inhibited by inhibitors of IKK, MEK, JNK, p38 and PKA. These results indicate that the PI3K/AKT/IKK and the mitogen‑activated protein (MAP) kinase pathways are involved in collagen‑ and PGE2‑induced aromatase expression, and also suggest that collagen and PGE2‑induced signaling pathways may crosstalk in regulating aromatase expression. This study enhances our understanding on the mechanism of regulation of aromatase expression by collagen and PGE2. Furthermore, this study provides a theoretical foundation for the development of specific inhibitors of aromatase by exploiting the signaling pathways identified herein in the context of breast cancer.

Introduction

Excessive estrogen exposure is a critical risk factor for breast cancer (17). While the ovary is the major site of estrogen biosynthesis in premenopausal women, adipose stromal cells (ASCs) in the breast are an important source of locally produced estrogen. Estrogens produced in distal adipose tissue and within the breast tissue influence the growth of breast epithelial cells (8). Especially, excessive local estrogen production in the breast promotes estrogen-dependent breast cancer. At the molecular level, tumor cells secrete factors such as prostaglandin E2 (PGE2). PGE2 stimulates stromal expression of aromatase, a key enzyme in estrogen biosynthesis (9). The breast quadrant bearing a malignant tumor shows consistently high levels of aromatase activity (10), and breast adipose tissue adjacent to the tumor shows a marked increase in aromatase expression and activity (1113). The clinically proven efficacy of aromatase inhibitors (AIs) in treating estrogen receptor-positive (ER+) post-menopausal breast cancer patients highlights the important role of excessive local estrogen production in breast cancer development. However, AIs indiscriminately reduce estrogen synthesis throughout the body, causing major side-effects including bone loss, increased fracture rates, and abnormal lipid metabolism (1416). Specific inhibitors that selectively inhibit aromatase expression in the tumor micro-environment would considerably benefit patients, by reducing side-effects. In addition, the aromatase promoters I.3 and PII have been reported to be activated in tumor, but not in healthy tissues (8). This implies that specific inhibition of pathways that lead to the activation of these promoters may specifically inhibit aromatase expression in the tumor tissue. In order to identify these pathways, the mechanism of regulation of aromatase expression needs to be elucidated.

Transcription of the aromatase gene is controlled by a number of tissue- and cell type-specific promoters that are located upstream of the aromatase coding region. The coding region of the aromatase transcripts is identical in different tissues, but a noncoding exon 1 is transcribed in a tissue-specific manner and spliced with the common coding exons (9,1719). In cancer-free breast adipose tissue, the aromatase gene is mainly transcribed under control of the relatively weak I.4 promoter, with a small amount of aromatase mRNA coming from ovary-specific promoters I.3 and PII. However, in ASCs adjacent to the breast tumor, aromatase expression is activated via the proximally-located promoters I.3 and PII (8). The switch in promoter utilization from the weak I.4 to the stronger I.3 and PII results in elevated aromatase expression and excessive production of local estrogen (20,21).

Several lines of evidence indicate that PGE2 is involved in breast cancer development and progression (2224). In the tumor microenvironment, PGE2 activates the protein kinase A and C (PKA and PKC) pathways and induces aromatase expression via promoters I.3 and PII in adjacent ASCs (25). In PGE2-treated ASCs, the aromatase mRNA level was found to be markedly increased, and phosphorylation of a number of signaling proteins was observed. These signaling proteins include PKA, PKC, transforming growth factor-β-activated kinase-1 (TAK1), mitogen-activated protein kinase kinase 4 (MMK4), c-Jun NH2-terminal kinase 1 (JNK-1) and mitogen-activated protein kinase (MAPK) p38 (26). This leads to phosphorylation of members of the Jun family of transcription factors and of the activating transcription factor (ATF) 2 protein. The phosphorylated Jun and ATF transcription factors directly bind to the promoter I.3 and PII to regulate aromatase gene expression (26,27). Inhibition of p38 or JNK1 by respective inhibitors effectively the blocked PGE2-induced aromatase expression. Consistently, knockdown of p38, JNK1, JunB or JunD by small interfering RNA (siRNA) also blocked PGE2-induced aromatase expression (28). Therefore, PGE2-induced pathways play important roles in regulation of aromatase expression in the tumor microenvironment.

It has been shown that mechanical changes, including elevated extracellular matrix (ECM) stiffness and increased interstitial pressures, are associated with epithelial carcinomas. Moreover, mechanical force due to altered architecture in the tissue microenvironment can affect gene expression patterns (2934). In breast cancer, mechanical force significantly affects the invasive behavior of tumor cells, as well as breast cancer incidence and mortality (3539).

In this study, we used a collagen 3D culture system to investigate the signaling pathways involved in regulation of aromatase expression. Mammographically dense breast tissue is linked to increased risk of breast carcinoma (40,41). Areas of increased breast density are not only associated with increased epithelial and stromal cellularity, but also significantly increased fibrillar collagen deposition (4245). It was also shown that increased stromal collagen in mouse mammary tissue significantly increases tumor formation and results in a significantly enhanced invasive phenotype (46). However, the mechanisms driving collagen-related breast tumor formation and progression remain largely unknown. Initiation of collagen-induced signals is mediated by a batch of cell-surface receptors, including integrins, discoidin domain receptors, glycoprotein VI, leukocyte-associated immunoglobin-like receptor-1, and members of the mannose receptor family (47). We are interested in two collagen receptors, α2β1 integrin and discoidin domain receptor 1. These two receptors propagate signals through separate, as well as shared pathways. One pathway is initiated by the integrin-linked kinase (ILK), which passes the signal to the IκB kinase (IKK) β via phosphatidylinositide 3-kinase (PI3K)/AKT (4851). Another pathway is the MAPK pathway, including extracellular-signal-regulated kinase (ERK)1/2, JNK and p38 (4855). It has been shown that IKKβ is involved in cell shape-induced aromatase expression, while MAPK pathways are involved in PGE2-induced aromatase expression (26,28,56). Based on this, we hypothesized that collagen may induce aromatase expression through these pathways and that there may be a crosstalk between collagen- and PGE2-induced signaling pathways. In this study, we investigated the signaling pathways involved in collagen- and PGE2-induced aromatase expression. This study will contribute to the understanding of the mechanism of regulation of aromatase expression by collagen and PGE2. Our study provides useful insights for the design of selective inhibitors of aromatase in the context of breast cancer via the inhibition of specific signaling pathways.

Materials and methods

Cell culture

Primary human ASCs were isolated from individuals undergoing elective surgical procedures at the Department of Breast Surgery, General Hospital of Nanjing Military Region (Fuzhou, China). Informed consent was obtained and this study was approved by the Ethics Committee of the General Hospital of Nanjing Military Region (Fuzhou, China) The cells were cultured in Dulbecco's modified Eagle's medium (DMEM)/nutrient mixture F-12 (Gibco Industries, Inc., Big Cabin, OK, USA), supplemented with 10% fetal bovine serum (FBS; Hyclone, Lawrenceville, GA, USA) and 1% antibiotic-antimycotic solution (Gibco Industries, Inc.). The 3D culture was performed in 24-well plates using collagen (collagen bovine type I; BD Biosciences, San Jose, CA, USA). Briefly, 2×105 cells were suspended in 125 µl medium and mixed with 125 µl collagen. Following the formation of a gel-like 3D structure, additional medium was added to the top.

Chemicals and treatment

PGE2 was purchased from Cayman Chemicals (Ann Arbor, MI, USA). The PI3K inhibitor LY-294002 was purchased from Sigma-Aldrich (St. Louis, MO, USA). The mitogen-activated protein kinase kinase (MEK)1/2 inhibitor U0126, JNK inhibitor AS601245, p38 inhibitor SB202190, and IKK inhibitor BAY11-7082 were purchased from Calbiochem (La Jolla, CA, USA). The PKA inhibitor H89 dihydrochloride was purchased from Cell Signaling Technology (Danvers, MA, USA). ASCs were serum-starved for 16 h and exposed to collagen or PGE2 with and without inhibitors for 12 h. Samples were harvested for RNA analysis. Chemical concentrations used in this study are shown in the figure legends.

Quantitative (q) and semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR)

Cells in collagen were released by using collagenase (Sigma-Aldrich). Total RNA was isolated using the Invitrogen™ TRIzol reagent (Thermo Fisher Scientific, Waltham, MA, USA) according to the manufacturer's instructions. The RNA concentration was measured using a NanoDrop2000 (Thermo Fisher Scientific), and RNA was reverse-transcribed using the ImPrompII™ kit (Promega, Madison, WI, USA). qPCR was carried out using the fluorescent dye SYBR-Green (Applied Biosystems, Foster City, CA, USA) on an ABI 7900 Real-Time PCR system (Applied Biosystems). The primers for amplification of aromatase were: forward, 5′-TGGAATTATGAGGGCACATCC-3′, and reverse, 5′-GTCCAATTCCCATGCAGTAGC-3′. Semi-quantitative RT-PCR was performed using primer pairs that are specific to the promoters I.4, I.3 and PII of the aromatase transcripts. The glyceraldehyde 3-phosphate dehydrogenase gene (GAPDH) served as an internal control. The forward primers used were: promoter I.3, 5′-CCTTGTTTTGACTTGTAACCA-3′; promoter I.4, 5′-GTAGAACGTGACCAACTGG-3′; and promoter PII, 5′-GCAACAGGAGCTATAGAT-3′. The reverse primer for all three promoters was 5′-ATTCCCATGCAGTAGCCAGG-3′. The GAPDH primers were: forward, 5′-CCATCAATGACCCCTTCATTG-3′, and reverse, 5′-GACGGTGCCATGGAATTT-3′. The PCR reaction was performed using reagents from the PCR Master Mix kit (Promega). The PCR cycling conditions were as follows: 94°C for 2 min (1 cycle), 94°C for 30 sec, 56°C for 30 sec, 72°C for 1 min (35 cycles), 72°C for 10 min (1 cycle), and hold at 4°C.

siRNA knockdown

Gene-specific knockdown by siRNA oligonucleotides was conducted using the Invitrogen™ Lipofectamine® RNAiMAX reagent (Thermo Fisher Scientific). Briefly, cells at an approximate density of 60% were transfected with siRNA oligos at a final concentration of 10 nM. Cells were trypsinized 24 h after transfection using 0.25% Trypsin-EDTA (Gibco Industries, Inc.), plated in subconfluent condition, and cultured in collagen. Cells were harvested for RNA analysis 12 h after plating. c-Jun, JunB, AKT2 and control siRNA oligos were purchased from Dharmacon, Inc. (Lafayette, CO, USA).

Statistics and data analysis

A paired t-test was used for pairwise comparisons of chemical-treated cells to control cells. Aromatase mRNA levels from independent measurements were collected for data analysis The data were analyzed using the SPSS 18.0 software (International Business Machines, Armonk, NY, USA).

Results

Collagen and PGE2 induce aromatase expression in adipose stromal cells

Collagen is a major component of the ECM and contributes to the formation of mechanical force in the tissue microenvironment. Mechanical force influences cell shape, as well as gene expression patterns (35,53,57,58). Furthermore, IKKβ is a downstream signaling molecule of collagen-induced signaling pathways that may play the same role in regulating aromatase expression. Based on these findings, we decided to test whether collagen can induce aromatase expression in ASCs.

Collagen-induced aromatase expression was revealed to be nearly 100-fold higher in ASCs compared to control ASCs that grew on 2D dishes (Fig. 1A). Data from semi-quantitative RT-PCR showed that collagen-induced aromatase expression involves the promoters I.3 and PII (Fig. 1C). PGE2-treated ASCs also showed induced aromatase expression through promoters I.3 and PII (Fig. 1B and D), in accordance with a previous study (26). In conclusion, both collagen and PGE2 can induce aromatase expression via promoters I.3 and PII in adipose stromal cells.

MAP kinase pathways are involved in collagen- and PGE2-induced aromatase expression

It has been shown that collagen activates MAP kinases, including ERK1/2, JNK and p38 (4855). In addition, MAP kinases are induced by PGE2 in ASCs (26). Furthermore, PGE2 induces aromatase expression in ASCs through JNK and p38 (26). Therefore, we hypothesized that collagen may induce aromatase expression by activating MAP kinase pathways. Collagen-induced aromatase expression was found significantly inhibited by the MEK inhibitor U0126, which leads to reduced ERK phosphorylation (Fig. 2A). Collagen-induced aromatase expression was also significantly inhibited by the JNK inhibitor AS601245, but not by the p38 inhibitor SB202190. PGE2-induced aromatase expression was significantly inhibited by all three inhibitors (Fig. 2B). Semi-quantitative RT-PCR confirmed these results (Fig. 2C and D). Therefore, we conclude that the MAP kinases ERK1/2 and JNK are involved in collagen-induced aromatase expression, while ERK1/2, JNK and p38 are all involved in PGE2-induced aromatase expression.

Since transcription factors of the Jun family act downstream of the MAP kinase pathways, and previous data from our laboratory, as well as other studies showed that the Jun family plays important roles in regulating aromatase expression (26,28,59,60), we knocked down c-Jun and JunB by siRNA to determine whether these molecules have an effect on aromatase expression. Knockdown of JunB significantly reduced collagen-induced aromatase expression. Knockdown of c-Jun did not reduce collagen-induced aromatase expression in our experiments (Fig. 2E). However, we observed that the c-Jun knockdown was not as efficient as the JunB one in terms of protein expression (data not shown). Semi-quantitative RT-PCR confirmed these results and showed that the JunB knockdown affects aromatase expression via both promoters I.3 and PII (Fig. 2F). In conclusion, JunB may be involved in collagen-induced aromatase expression by regulating the aromatase promoters I.3 and PII.

Cell-ECM communications are predominantly mediated by integrins, and integrin-mediated signaling events can activate PI3K/AKT (61,62). On the other hand, AKT is physically associated with IKKβ and can trigger phosphorylation of IKK in response to inflammatory factors such as tumor necrosis factor-α (63,64). We therefore hypothesized that AKT may play an important role in passing signals from ECM to downstream IKK, and thus, in activating aromatase expression. There are three AKT genes in the human genome: AKT1, AKT2 and AKT3. These genes code for enzymes that belong to the serine/threonine-specific protein kinase family (6567). Knockdown of AKT2 significantly reduced collagen-induced aromatase expression (Fig. 3E). Data from semi-quantitative RT-PCR confirmed these results and showed that knockdown of AKT2 affects both promoters I.3 and II (Fig. 3F). These data indicates that collagen induces aromatase expression at least partly via the PI3K/AKT/IKK pathway.

Next, we tested whether PKA is involved in collagen-induced aromatase expression, since it has been shown to be involved in PGE2-induced aromatase expression (26,68). Our data confirmed the important role of PKA in regulating PGE2-induced aromatase expression (Fig. 3B). In addition, our data showed that the PKA inhibitor H89 significantly inhibits collagen-induced aromatase expression (Fig. 3A). The RT-qPCR results were confirmed by semi-quantitative RT-PCR (Fig. 3C and D). Taken together, these findings strongly suggest that PKA plays an important role in PGE2-, as well as in collagen-induced aromatase expression.

Discussion

Excessive exposure to estrogen is a critical risk factor for breast cancer, while aromatase is a key enzyme controlling estrogen production. Currently, AIs are the most effective endocrine treatment for breast cancer (8,69). However, AIs indiscriminately reduce estrogen synthesis throughout the body, causing major side-effects (1416). To address this issue, new drug targets need to be identified, allowing to selectively block aromatase expression in the tumor microenvironment. To this end, it is important to dissect the mechanisms by which aromatase expression is regulated. Data from this study showed that collagen-induced aromatase expression in ASCs is significantly reduced by inhibitors of PI3K, IKK, MEK, JNK, PKA and the knockdown of the JunB and AKT2 genes. These findings indicate that the PI3K/AKT/IKK, as well as the MAP kinase pathways play important roles in collagen-induced aromatase expression. On the other hand, PGE2-induced aromatase expression was significantly reduced by inhibitors of IKK, MEK, JNK, p38 and PKA. Data from semi-quantitative RT-PCR showed that collagen and PGE2 induce aromatase expression through the aromatase promoters I.3 and PII. In addition, the semi-quantitative data were consistent with quantitative RT-PCR data in the drug treatment and the knockdown experiments.

Data from this study also indicated that there is a crosstalk between collagen and PGE2-induced signaling pathways in regulating aromatase expression. Signaling molecules that are responsible for this crosstalk could serve as potential drug targets. Signals induced by both PGE2 and collagen could be blocked by hitting the targets that are common to both pathways involved in regulating collagen and PGE2-induced expression, which may effectively silence the aromatase promoters. In addition to these shared-between-pathways molecules, our data also highlighted a number of proteins that are unique to one of the two aromatase-activating pathways. For example, p38 plays a critical role in PGE2-induced but not in collagen-induced aromatase expression. PI3K is another example, since this protein plays a critical role in collagen-induced, but not in PGE2-induced aromatase expression.

In the present study, PGE2-induced aromatase expression was inhibited by a MEK inhibitor. This indicates that the ERK pathway may be involved in regulating aromatase expression in response to PGE2 induction. Data from another study showed that the same MEK inhibitor has no effects on PGE2-induced aromatase expression (26). This discrepancy could be due to the use of different cells in the two studies. Since regulation of gene expression is complex, the signaling pathways involved in regulating expression are only one facet of the process. Other factors likely contribute to aromatase expression as well. For instance, the DNA methylation status of the aromatase promoter region also plays important roles in regulating aromatase expression. In another project of our research team, a DNA methylation assay was performed to test the methylation status of aromatase promoters in a batch of adipose stromal cells. Our data showed that the highest DNA methylation load of certain CpG sites corresponds to the lowest aromatase activation. Reduction of the DNA methylation load by drug treatment restored aromatase responsiveness to forskolin activation (unpublished data). Another study also showed that CpG dinucleotide methylation of the aromatase promoter modulates cAMP-stimulated aromatase activity (70).

This study elucidated the signaling pathways involved in collagen- and PGE2-induced aromatase expression in adipose stromal cells. It is thus expected to shed light on the mechanisms of aromatase activation in response to collagen, as well as PGE2. This study provides an opportunity to test whether breast cancer can be effectively treated by selectively blocking aromatase expression via the inhibition of the specific signaling pathways identified herein. Further investigation is however needed to identify the downstream transcription factors and additional molecular targets for selective inhibition of aromatase expression.

References

1 

Ewertz M: Influence of non-contraceptive exogenous and endogenous sex hormones on breast cancer risk in Denmark. Int J Cancer. 42:832–838. 1988. View Article : Google Scholar : PubMed/NCBI

2 

Nass SJ and Davidson NE: The biology of breast cancer. Hematol Oncol Clin North Am. 13:311–332. 1999. View Article : Google Scholar : PubMed/NCBI

3 

Okobia MN and Bunker CH: Estrogen metabolism and breast cancer risk - a review. Afr J Reprod Health. 10:13–25. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Meyer F, Brown JB, Morrison AS and MacMahon B: Endogenous sex hormones, prolactin, and breast cancer in premenopausal women. J Natl Cancer Inst. 77:613–616. 1986.PubMed/NCBI

5 

Key T, Appleby P, Barnes I and Reeves G; Endogenous Hormones and Breast Cancer Collaborative Group: Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. J Natl Cancer Inst. 94:606–616. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Onland-Moret NC, Kaaks R, van Noord PA, et al: Urinary endogenous sex hormone levels and the risk of postmenopausal breast cancer. Br J Cancer. 88:1394–1399. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Biggar RJ: Molecular pathways: digoxin use and estrogen-sensitive cancers - risks and possible therapeutic implications. Clin Cancer Res. 18:2133–2137. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Bulun SE, Lin Z, Imir G, et al: Regulation of aromatase expression in estrogen-responsive breast and uterine disease: from bench to treatment. Pharmacol Rev. 57:359–383. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Simpson ER and Davis SR: Minireview: aromatase and the regulation of estrogen biosynthesis - some new perspectives. Endocrinology. 142:4589–4594. 2001.PubMed/NCBI

10 

O'Neill JS, Elton RA and Miller WR: Aromatase activity in adipose tissue from breast quadrants: a link with tumour site. Br Med J (Clin Res Ed). 296:741–743. 1988. View Article : Google Scholar

11 

Bulun SE, Price TM, Aitken J, Mahendroo MS and Simpson ER: A link between breast cancer and local estrogen biosynthesis suggested by quantification of breast adipose tissue aromatase cytochrome P450 transcripts using competitive polymerase chain reaction after reverse transcription. J Clin Endocrinol Metab. 77:1622–1628. 1993.PubMed/NCBI

12 

Harada N: Aberrant expression of aromatase in breast cancer tissues. J Steroid Biochem Mol Biol. 61:175–184. 1997. View Article : Google Scholar : PubMed/NCBI

13 

Sasano H and Harada N: Intratumoral aromatase in human breast, endometrial, and ovarian malignancies. Endocr Rev. 19:593–607. 1998.PubMed/NCBI

14 

Morales L, Neven P and Paridaens R: Choosing between an aromatase inhibitor and tamoxifen in the adjuvant setting. Curr Opin Oncol. 17:559–565. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Umar A, Dunn BK and Greenwald P: Future directions in cancer prevention. Nat Rev Cancer. 12:835–848. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Tomao F, Spinelli G, Vici P, et al: Current role and safety profile of aromatase inhibitors in early breast cancer. Expert Rev Anticancer Ther. 11:1253–1263. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Kamat A, Hinshelwood MM, Murry BA and Mendelson CR: Mechanisms in tissue-specific regulation of estrogen biosynthesis in humans. Trends Endocrinol Metab. 13:122–128. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Bulun SE, Sebastian S, Takayama K, Suzuki T, Sasano H and Shozu M: The human CYP19 (aromatase P450) gene: update on physiologic roles and genomic organization of promoters. J Steroid Biochem Mol Biol. 86:219–224. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Stocco C: Tissue physiology and pathology of aromatase. Steroids. 77:27–35. 2012. View Article : Google Scholar :

20 

Meng L, Zhou J, Sasano H, Suzuki T, Zeitoun KM and Bulun SE: Tumor necrosis factor alpha and interleukin 11 secreted by malignant breast epithelial cells inhibit adipocyte differentiation by selectively down-regulating CCAAT/enhancer binding protein alpha and peroxisome proliferator-activated receptor gamma: mechanism of desmoplastic reaction. Cancer Res. 61:2250–2255. 2001.PubMed/NCBI

21 

Zhou J, Gurates B, Yang S, Sebastian S and Bulun SE: Malignant breast epithelial cells stimulate aromatase expression via promoter II in human adipose fibroblasts: an epithelial-stromal interaction in breast tumors mediated by CCAAT/enhancer binding protein beta. Cancer Res. 61:2328–2334. 2001.PubMed/NCBI

22 

Purohit A, Ghilchik MW, Duncan L, et al: Aromatase activity and interleukin-6 production by normal and malignant breast tissues. J Clin Endocrinol Metab. 80:3052–3058. 1995.PubMed/NCBI

23 

Schrey MP and Patel KV: Prostaglandin E2 production and metabolism in human breast cancer cells and breast fibroblasts. Regulation by inflammatory mediators. Br J Cancer. 72:1412–1419. 1995. View Article : Google Scholar : PubMed/NCBI

24 

Chang SH, Liu CH, Conway R, et al: Role of prostaglandin E2-dependent angiogenic switch in cyclooxygenase 2-induced breast cancer progression. Proc Natl Acad Sci USA. 101:591–596. 2004. View Article : Google Scholar :

25 

Zhao Y, Agarwal VR, Mendelson CR and Simpson ER: Estrogen biosynthesis proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, leading to activation of promoter II of the CYP19 (aromatase) gene. Endocrinology. 137:5739–5742. 1996.PubMed/NCBI

26 

Chen D, Reierstad S, Lin Z, et al: Prostaglandin E2 induces breast cancer related aromatase promoters via activation of p38 and c-Jun NH-terminal kinase in adipose fibroblasts. Cancer Res. 67:8914–8922. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Bulun SE, Lin Z, Zhao H, et al: Regulation of aromatase expression in breast cancer tissue. Ann NY Acad Sci. 1155:121–131. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Chen D, Reierstad S, Fang F and Bulun SE: JunD and JunB integrate prostaglandin E activation of breast cancer-associated proximal aromatase promoters. Mol Endocrinol. 25:767–775. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Avvisato CL, Yang X, Shah S, et al: Mechanical force modulates global gene expression and beta-catenin signaling in colon cancer cells. J Cell Sci. 120:2672–2682. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Shyy JY and Chien S: Role of integrins in endothelial mechanosensing of shear stress. Circ Res. 91:769–775. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Alenghat FJ and Ingber DE: Mechanotransduction: all signals point to cytoskeleton, matrix, and integrins. Sci STKE. 119:pe62002.

32 

Lopez JI, Mouw JK and Weaver VM: Biomechanical regulation of cell orientation and fate. Oncogene. 27:6981–6993. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Nelson CM and Bissell MJ: Of extracellular matrix, scaffolds, and signaling: tissue architecture regulates development, homeostasis, and cancer. Annu Rev Cell Dev Biol. 22:287–309. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Provenzano PP and Keely PJ: Mechanical signaling through the cytoskeleton regulates cell proliferation by coordinated focal adhesion and Rho GTPase signaling. J Cell Sci. 124:1195–1205. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Paszek MJ and Weaver VM: The tension mounts: mechanics meets morphogenesis and malignancy. J Mammary Gland Biol Neoplasia. 9:325–342. 2004. View Article : Google Scholar

36 

Ingber DE: Can cancer be reversed by engineering the tumor microenvironment? Semin Cancer Biol. 18:356–364. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Le Beyec J, Xu R, Lee SY, et al: Cell shape regulates global histone acetylation in human mammary epithelial cells. Exp Cell Res. 313:3066–3075. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Wiseman BS and Werb Z: Stromal effects on mammary gland development and breast cancer. Science. 296:1046–1049. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Plodinec M and Schoenenberger CA: Spatial organization acts on cell signaling: how physical force contributes to the development of cancer. Breast Cancer Res. 12:3082010. View Article : Google Scholar : PubMed/NCBI

40 

McCormack VA and dos Santos Silva I: Breast density and parenchymal patterns as markers of breast cancer risk: a meta-analysis. Cancer Epidemiol Biomarkers Prev. 15:1159–1169. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Boyd NF, Martin LJ, Stone J, Greenberg C, Minkin S and Yaffe MJ: Mammographic densities as a marker of human breast cancer risk and their use in chemoprevention. Curr Oncol Rep. 3:314–321. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Hawes D, Downey S, Pearce CL, et al: Dense breast stromal tissue shows greatly increased concentration of breast epithelium but no increase in its proliferative activity. Breast Cancer Res. 8:R242006. View Article : Google Scholar : PubMed/NCBI

43 

Guo YP, Martin LJ, Hanna W, et al: Growth factors and stromal matrix proteins associated with mammographic densities. Cancer Epidemiol Biomarkers Prev. 10:243–248. 2001.PubMed/NCBI

44 

Alowami S, Troup S, Al-Haddad S, Kirkpatrick I and Watson PH: Mammographic density is related to stroma and stromal proteoglycan expression. Breast Cancer Res. 5:R129–R135. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Li T, Sun L, Miller N, et al: The association of measured breast tissue characteristics with mammographic density and other risk factors for breast cancer. Cancer Epidemiol Biomarkers Prev. 14:343–349. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Provenzano PP, Inman DR, Eliceiri KW, et al: Collagen density promotes mammary tumor initiation and progression. BMC Med. 6:112008. View Article : Google Scholar : PubMed/NCBI

47 

Leitinger B and Hohenester E: Mammalian collagen receptors. Matrix Biol. 26:146–155. 2007. View Article : Google Scholar

48 

Shintani Y, Fukumoto Y, Chaika N, Svoboda R, Wheelock MJ and Johnson KR: Collagen I-mediated up-regulation of N-cadherin requires cooperative signals from integrins and discoidin domain receptor 1. J Cell Biol. 180:1277–1289. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Shintani Y, Maeda M, Chaika N, Johnson KR and Wheelock MJ: Collagen I promotes epithelial-to-mesenchymal transition in lung cancer cells via transforming growth factor-beta signaling. Am J Respir Cell Mol Biol. 38:95–104. 2008. View Article : Google Scholar

50 

Shintani Y, Hollingsworth MA, Wheelock MJ and Johnson KR: Collagen I promotes metastasis in pancreatic cancer by activating c-Jun NH2-terminal kinase 1 and up-regulating N-cadherin expression. Cancer Res. 66:11745–11753. 2006. View Article : Google Scholar : PubMed/NCBI

51 

Shintani Y, Wheelock MJ and Johnson KR: Phosphoinositide-3 kinase-Rac1-c-Jun NH -terminal kinase signaling mediates collagen I-induced cell scattering and up-regulation of N-cadherin expression in mouse mammary epithelial cells. Mol Biol Cell. 17:2963–2975. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Wheelock MJ and Johnson KR: Cadherin-mediated cellular signaling. Curr Opin Cell Biol. 15:509–514. 2003. View Article : Google Scholar : PubMed/NCBI

53 

Plant AL, Bhadriraju K, Spurlin TA and Elliott JT: Cell response to matrix mechanics: focus on collagen. Biochim Biophys Acta. 1793:893–902. 2009. View Article : Google Scholar

54 

Zhu X and Assoian RK: Integrin-dependent activation of MAP kinase: a link to shape-dependent cell proliferation. Mol Biol Cell. 6:273–282. 1995. View Article : Google Scholar : PubMed/NCBI

55 

Zhang L, Bewick M and Lafrenie RM: Role of Raf-1 and FAK in cell density-dependent regulation of integrin-dependent activation of MAP kinase. Carcinogenesis. 23:1251–1258. 2002. View Article : Google Scholar : PubMed/NCBI

56 

Ghosh S, Choudary A, Musi N, Hu Y and Li R: IKKbeta mediates cell shape-induced aromatase expression and estrogen biosynthesis in adipose stromal cells. Mol Endocrinol. 23:662–670. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Lee EY, Parry G and Bissell MJ: Modulation of secreted proteins of mouse mammary epithelial cells by the collagenous substrata. J Cell Biol. 98:146–155. 1984. View Article : Google Scholar : PubMed/NCBI

58 

Kass L, Erler JT, Dembo M and Weaver VM: Mammary epithelial cell: influence of extracellular matrix composition and organization during development and tumorigenesis. Int J Biochem Cell Biol. 39:1987–1994. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Ghosh S, Wu Y, Li R and Hu Y: Jun proteins modulate the ovary-specific promoter of aromatase gene in ovarian granulosa cells via a cAMP-responsive element. Oncogene. 24:2236–2246. 2005. View Article : Google Scholar : PubMed/NCBI

60 

Hu YF and Li R: JunB potentiates function of BRCA1 activation domain 1 (AD1) through a coiled-coil-mediated interaction. Genes Dev. 16:1509–1517. 2002. View Article : Google Scholar : PubMed/NCBI

61 

Basson MD: An intracellular signal pathway that regulates cancer cell adhesion in response to extracellular forces. Cancer Res. 68:2–4. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Brancaccio M, Hirsch E, Notte A, Selvetella G, Lembo G and Tarone G: Integrin signalling: the tug-of-war in heart hypertrophy. Cardiovasc Res. 70:422–433. 2006. View Article : Google Scholar : PubMed/NCBI

63 

Häcker H and Karin M: Regulation and function of IKK and IKK-related kinases. Sci STKE. 357:re132006.

64 

Bai D, Ueno L and Vogt PK: Akt-mediated regulation of NFkappaB and the essentialness of NFkappaB for the oncogenicity of PI3K and Akt. Int J Cancer. 125:2863–2870. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Staal SP, Hartley JW and Rowe WP: Isolation of transforming murine leukemia viruses from mice with a high incidence of spontaneous lymphoma. Proc Natl Acad Sci USA. 74:3065–3067. 1977. View Article : Google Scholar : PubMed/NCBI

66 

Staal SP: Molecular cloning of the akt oncogene and its human homologues AKT1 and AKT2: amplification of AKT1 in a primary human gastric adenocarcinoma. Proc Natl Acad Sci USA. 84:5034–5037. 1987. View Article : Google Scholar : PubMed/NCBI

67 

Yang ZZ, Tschopp O, Baudry A, Dümmler B, Hynx D and Hemmings BA: Physiological functions of protein kinase B/Akt. Biochem Soc Trans. 32:350–354. 2004. View Article : Google Scholar : PubMed/NCBI

68 

Bulun SE, Chen D, Lu M, et al: Aromatase excess in cancers of breast, endometrium and ovary. J Steroid Biochem Mol Biol. 106:81–96. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Baum M, Budzar AU, Cuzick J, et al: Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet. 359:2131–2139. 2002. View Article : Google Scholar : PubMed/NCBI

70 

Demura M and Bulun SE: CpG dinucleotide methylation of the CYP19 I.3/II promoter modulates cAMP-stimulated aromatase activity. Mol Cell Endocrinol. 283:127–132. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2015
Volume 12 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tan T, Wang L and Wang B: Collagen and prostaglandin E2 regulate aromatase expression through the PI3K/AKT/IKK and the MAP kinase pathways in adipose stromal cells. Mol Med Rep 12: 4766-4772, 2015
APA
Tan, T., Wang, L., & Wang, B. (2015). Collagen and prostaglandin E2 regulate aromatase expression through the PI3K/AKT/IKK and the MAP kinase pathways in adipose stromal cells. Molecular Medicine Reports, 12, 4766-4772. https://doi.org/10.3892/mmr.2015.3901
MLA
Tan, T., Wang, L., Wang, B."Collagen and prostaglandin E2 regulate aromatase expression through the PI3K/AKT/IKK and the MAP kinase pathways in adipose stromal cells". Molecular Medicine Reports 12.3 (2015): 4766-4772.
Chicago
Tan, T., Wang, L., Wang, B."Collagen and prostaglandin E2 regulate aromatase expression through the PI3K/AKT/IKK and the MAP kinase pathways in adipose stromal cells". Molecular Medicine Reports 12, no. 3 (2015): 4766-4772. https://doi.org/10.3892/mmr.2015.3901