Zinc-α2-glycoprotein: A proliferative factor for breast cancer? In vitro study and molecular mechanisms

  • Authors:
    • Laetitia Delort
    • Stéphane Perrier
    • Virginie Dubois
    • Hermine Billard
    • Tomas Mracek
    • Chen Bing
    • Marie-Paule Vasson
    • Florence Caldefie-Chézet
  • View Affiliations

  • Published online on: February 27, 2013     https://doi.org/10.3892/or.2013.2311
  • Pages: 2025-2029
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Zinc-α2-glycoprotein (ZAG) is a new adipokine whose gene expression is downregulated in obese patients. We recently reported ZAG expression in breast tumor or healthy breast tissue and detected this expression at high levels in ductal carcinoma and in normal epithelial adjacent tissue but not in normal tissue of healthy women. In the present study, we used two human breast tumor cell lines (MCF-7 and MDA-MB‑231) and one fibrocystic breast cell line (MCF‑10a) to examine whether recombinant ZAG has an effect on proliferative/apoptotic response in breast cancer cell lines. ZAG seemed to exert a proliferative effect on breast cancer cell proliferation [+11 to 27% in MCF-7 with (ZAG) = 5-20 µg/ml; +13% in MDA-MB-231 with (ZAG) = 5 µg/ml] and, on the contrary, an anti-proliferative effect in the fibrocystic breast cell line [-5 to -8% in MCF-10a with (ZAG) = 5-10 µg/ml]. ZAG was able to modulate gene and protein expression involved in the apoptotic response. However, further studies are required to fully elucidate the effects of ZAG on the proliferation of mammary cells.

Introduction

Obesity is related to several metabolic disorders such as type 2 diabetes mellitus, coronary heart disease and hypertension, and is associated with cancer development in different tissues including colon, prostate and breast (1). It has been clearly demonstrated that obesity is a risk factor for breast cancer development in postmenopausal women (2,3). Moreover, an excess of adipose tissue favors breast cancer recurrence and is associated with higher mortality (4). Thus, overweight or obese women with breast carcinoma have a 2.5-fold increased risk of mortality within five years of diagnosis compared with lean women (5). Numerous factors have been proposed to explain the relationship between obesity and breast cancer (68), however, none has been completely conclusive.

Emerging data suggest that adipose tissue, which is an endocrine organ producing a large range of factors, may interfere with cancer development. These factors, mainly secreted by the adipose tissue, known as adipokines, include angiogenic factors, paracrine mitogens and anti-mitogens, growth factors and pro-inflammatory cytokines (IL-1, TNF-α and IL-6) involved in the mediation or the coordination of inflammatory diseases and obesity (9,10). Adipokines are produced by different fat depots, including subcutaneous, visceral and mammary adipose tissue. Of note, adipokines may act on breast tissue in an endocrine manner (via external adipose sources), in a paracrine pathway (via mammary adipose tissue secretion and non adipose sources including stromal cells and inflammatory cells) and in an autocrine manner (via the mammary tumor by itself). The structure of the mammary gland favors a close interaction between mammary adipose tissue and breast tissue, and suggests that adipokines produced by mammary adipose tissue and the tumor microenvironment may be the major link between obesity and disease progression and metastasis (1114). We previously investigated simultaneously the in vitro and in vivo molecular mechanisms by which leptin induces and, conversely, adiponectin suppresses, tumor proliferation in breast cancer cells (15,16). We suggested that these two adipokines have antagonistic properties in breast cancer development by modulating differentially both proliferative and apoptotic signaling pathways (17).

Zinc-α2-glycoprotein (ZAG) is a new adipokine whose gene expression is reduced in subcutaneous fat of obese patients (18,19). This expression is positively correlated with adiponectin expression and negatively correlated with leptin expression, suggesting a protective role for ZAG in breast cancer (20). ZAG is also a sound immunohistochemical marker of breast cell differentiation since ZAG tissue levels are associated with histological grades of tumors and vary from 4.6 μg/mg in well-differentiated tumors to 0.9 μg/mg in poorly differentiated tumors (21,22). However, other studies found that circulating levels of ZAG are significantly higher in cancer patients (51 mg/l) as compared with levels in healthy women (44 mg/l), particularly in patients with advanced and node-positive breast cancer (23). Moreover, Bing et al(18) showed that ZAG expression is upregulated in mice with cancer cachexia. Overexpression in white adipose tissue of tumor-bearing mice suggests that ZAG plays a role in the substantial reduction of adiposity in cancer cachexia. ZAG is also considered a prognostic marker in breast cancer (24). We recently reported ZAG expression in breast tumor or healthy breast tissue and detected this expression at high levels in ductal carcinoma and in normal epithelial adjacent tissue, but not in normal tissue of healthy women (25). We also found ZAG expression was positively correlated to leptin receptor and negatively correlated to adiponectin receptor in cancer tissues. Our previous data suggested both a potential prognostic role for ZAG in breast cancer and a close interaction between ZAG and other major adipokine pathways.

The aim of the present study was to characterize the involvement of ZAG in breast cancer proliferation. Thus, we explored the in vitro potential effect of human recombinant ZAG on i) proliferative/apoptotic response, and ii) the modifications of gene expression in different breast epithelial cell lines.

Materials and methods

Cell culture

The human breast cancer cell lines MCF-7 and MDA-MB-231, and the human fibrocystic breast cell line MCF-10a were obtained from the American Type Culture Collection (ATCC). MCF-7 cells were routinely cultured in RPMI-1640 medium supplemented with 10% heat-inactivated fetal calf serum (FCS), L-glutamine (2 mM) and gentamicin (50 μg/ml) at 37°C under a 5% CO2 atmosphere. MDA-MB-231 cells were cultured in Leibovitz’s L-15 medium with 15% heat-inactivated FCS, L-glutamine (2 mM) and gentamicin (50 μg/ml) at 37°C in humidified conditions. MCF-10a cells were cultured in DMEM HAM’s F12 medium supplemented with 10% heat-inactivated horse serum (HS), EGF (0.02 μg/ml), cholera toxin (0.1 μg/ml), hydrocortisone (0.5 μg/ml), insulin (0.25 UI/ml) and L-glutamine (2 mM) at 37°C under a 5% CO2 atmosphere (16).

Recombinant human ZAG (rh-ZAG)

Recombinant human (rh)-ZAG was kindly provided by Mracek et al(20) (Obesity Biology Research Unit, School of Clinical Sciences, University of Liverpool) and its production was recently described.

Proliferation assay

The human mammary cell lines (MCF-7, MDA-MB-231 and MCF-10a) were seeded at the density of 5×103 cells in 96-well plates in a complete medium. After 24 h of incubation, cells were washed with PBS and exposed to fresh medium (control) or to fresh medium containing different concentrations of rh-ZAG: 1.2, 2.5, 5, 10 and 20 μg/ml. After 96 h, cells were washed with PBS and incubated with 200 μl of a 25 μg/ml solution of resazurin in RPMI-1640 medium for 2 h at 37°C. Fluorescence was then measured on an automated 96-well plate reader (Fluoroskan Ascent FL; Thermo Fisher Scientific, Wilmington, DE, USA) using an excitation wavelength of 530 nm and an emission wavelength of 590 nm. Under these conditions, fluorescence was proportional to the number of living cells in the well (16). The cell proliferation assay was performed three times in triplicates for each concentration tested.

Analysis of gene expression

Total RNA was extracted with Trizol according to the manufacturer’s recommendations (Invitrogen). The quantity and quality of RNA was assessed by 260/280 ratio using a NanoDrop 8000 Spectrophotometer (Thermo Fisher Scientific). cDNAs were obtained with HighCap cDNA RT kit RNAse inhibitor (Applied Biosystems).

Real-time PCR assays of BAG1, BAG3, MX1, TP53 and 18S mRNA expression levels were performed using the StepOne instrument (Applied Biosystems) with Power SYBR-Green (Applied Biosystems) following the manufacturer’s instructions. The cDNAs were amplified using the primers summarized in Table I. The PCRs were set up in 48-well plates in a total volume of 20 μl and 20 ng of cDNA. The PCR conditions were as follows: 2 min at 50°C, 10 min at 95°C, 40 cycles of 15 sec at 95°C and 30 sec at 62°C.

Table I

Primers used in qRT-PCR assays.

Table I

Primers used in qRT-PCR assays.

BAG1F: 5′-CACAGCAATGAGAAGCACG-3′
BAG1R: 5′-GTGTTTCCATTTCCTTCAGAG-3
BAG3F: 5′-ATGACCCATCGAGAAACTGC-3′
BAG3R: 5′-AATTGGGATGTGTCCAGGAG-3′
MX1F: 5′-AGCTCGGCAACAGACTCTTC-3′
MX1R: 5′-GGATGATCAAAGGGATGTGG-3′
TP53F: 5′-GCGCACAGAGGAAGAGAATC-3′
TP53R: 5′-AGAGGAGCTGGTGTTGTTGG-3′
18SF: 5′-GTCTGTGATGCCCTTAGATG-3′
18SR: 5′-AGCTTATGACCCGCACTTAC-3′

[i] F, forward; R, reverse.

The comparative cycle threshold (CT) method (2−ΔΔCT) was used to calculate the relative gene expression of ZAG-treated cells normalized within the sample to an endogenous reference gene (18S), and relative to the expression of the same gene in untreated cells: 2−ΔΔCT method with ΔΔCT = [ΔCT (ZAG-treated cells) − ΔCT (untreated cells)] and ΔCT = [CT (target gene) − CT (reference gene)].

Statistical analysis was performed using the paired Student’s t-test. A P-value <0.05 was considered to indicate a statistically significant difference.

Western blotting

Cells lysates (containing 10 μg of total proteins) were separated on 4–12% SDS-PAGE gels (Invitrogen), transferred to nitrocellulose and blotted with various antibodies. We used primary antibodies (Santa Cruz Biotechnology, Inc.,) against the following proteins: p53 (DO-1), p21, Bax, c-Myc and cyclin D1. Secondary horseradish peroxidase (HRP)-conjugated antibodies were obtained from Santa Cruz Biotechnology, Inc., (anti-rabbit) or Dako (anti-mouse). Immunoreactive bands were visualized by incubation with DURA Western blotting detection system from Thermo Fisher Scientific. Glyceraldehyde-3-phosphate-dehydrogenase (GAPDH) monoclonal antibody was used as a loading control. Developed films were scanned as JPEG images, and the pixel intensities within a band were measured with ImageJ software. The intensity of each gene was then reported to GAPDH intensity.

Results

Mammary cell proliferation

The proliferation of estrogen receptor positive (ER+) mammary cancer cells MCF-7 was increased at the highest concentrations tested [11 to 27% with (ZAG) = 5 to 20 μg/ml] (Fig. 1). In ER- mammary cancer cells MDA-MB-231, rh-ZAG had a proliferative effect at the lowest concentrations tested [+24% with (ZAG) = 1.2 μg/ml; +13% with (ZAG) = 5 μg/ml]. By contrast, rh-ZAG had an anti-proliferative effect on fibrocystic breast cells MCF-10a when used at the concentrations of 5 and 10 μg/ml (−5 and −8% in proliferation decrease).

Gene expression

qRT-PCR was performed to study the effects of a 72-h rh-ZAG treatment on the expression of two pro-apoptotic genes (p53, mx1) and two anti-apoptotic genes (Bag1 and Bag3) in the three studied breast cell lines. We used rh-ZAG at the respective concentration points leading to an optimal effect on cell growth: 20 μg/ml for MCF-7 cells, 1 μg/ml for MDA-MB-231 cells and 10 μg/ml for MCF-10 cells.

Bag1 and Bag3 expressions were downregulated by rh-ZAG in MCF-7 and MDA-MB-231 cells, but remained unaltered in MCF-10a cells (Fig. 2). Concomitantly, Mx1 gene expression was upregulated by rh-ZAG in MCF-7 and MCF-10a and more strongly in MCF-7 cells.

Protein expression

Fig. 3 shows the results obtained in cells treated for 24, 48 or 72 h with rh-ZAG (0, 1, 10 ng/ml). In MCF-7 cells treated with rh-ZAG, an increase in p53 protein expression at 24 h [+100% with (ZAG) = 1 ng/ml; +300% with (ZAG) = 10 ng/ml] but a decrease at 72 h (-32% with (ZAG) = 10 ng/ml) were observed. The expression of c-Myc was increased only at 24 h (+160%) whereas the expression of p21 and Bax slightly increased during the experiments (p21: +21, +39 and 28% at 24, 48 and 72 h, respectively; Bax: +58, +37 and +64% at 24, 48 and 72 h, respectively). In MDA-MB-231 cells, there was no evidence of ZAG-modulated variations in the expression of the different proteins tested. In MCF-10a cells, rh-ZAG enhanced c-Myc expression at 72 h [+80% with (ZAG) = 10 ng/ml] while downregulating both p53 (−95%) and Bax (−90%) protein expression at the same incubation time.

Discussion

ZAG, a well-known lipid mobilizing factor, which is downregulated in obesity, is now considered as an adipokine. We previously reported in vivo ZAG expression in breast malignant cells and in normal epithelial adjacent tissue (25). The present study showed ZAG had a proliferative activity on the two human breast tumor cell lines (MCF-7 and MDA-MB-231), but an anti-proliferative effect on MCF-10a cells derived from a non-cancerous fibrocystic tissue.

Tumor growth should be viewed as the result of a balance between cell proliferation and cell death. Our aim was to assess whether ZAG can regulate mammary cancer proliferation not only through an inhibitory pathway but also by triggering a programmed cell death. Markedly, ZAG had a proliferative effect in MCF-7 cells while upregulating pro-apoptotic Mx1 and downregulating the gene expression of anti-apoptotic genes Bag1 and Bag3. Such a modulation on apoptotic markers should result in a slowing process of cell proliferation.

Our data also suggest that ZAG-induced cell-growth inhibition in MCF-10a breast cancer cells could be mediated through the c-Myc pathway. Indeed, we found ZAG upregulates c-Myc protein expression in these cells. Despite its role in promoting tumorigenesis, there is increasing evidence that c-Myc also induces apoptosis in cancer cells (26,27). Epithelial cells have also been shown to be susceptible to apoptosis by c-Myc (28). In addition, it was reported that MCF-7 breast carcinoma cells deprived of glucose exhibit both c-Myc elevation and significant cell death, which can be blocked by the addition of antisense c-Myc oligonucleotides (29).

We previously reported that endogenous ZAG was not associated with Bax and Bcl2 apoptosis biomarkers in breast cancer tissues (25). In the present study, we found exogenous ZAG did not consistently affect Bax expression in the two human breast tumor cell lines (MCF-7 and MDA-MB-231). By contrast, ZAG downregulated the expression of both pro-apoptotic proteins p53 and Bax in MCF-10a cells. We also observed an anti-proliferative effect for ZAG in these cells, therefore, apoptosis may not be the only mechanism to explain the resulting inhibitory effect on cell proliferation. In squamous carcinoma, ZAG inhibits cell proliferation by downregulating cdc2 expression and affecting cell cycle (30). In this study, ZAG did not affect the expression of the genes involved in apoptosis or differentiation (PCNA, p53, c-Myc and Bcl-2). Of note, if adipokines generally modulate the apoptotic response in vitro, they may exert their activities through different pathways from one cell type to another since breast cancer cell lines display distinct patterns of apoptosis-regulatory genes (31). For example, MCF-7 cells are caspase-3 negative, ZR-75 cells are Bcl-2 negative and the p53 status is either wild or mutant, depending on the cell line.

Thus, our in vitro approach does not seem to reflect the complex regulation that may occur in vivo. Metabolic dysregulations associated with obesity (such as hypoadiponectinemia and hyperleptinemia) are likely to promote cancer cell growth via both systemic and local mechanisms. Furthermore, when mammary cells are engaged in the process of carcinogenesis, they produce adipokines (mainly leptin) able to act on surrounding cancer cells in a paracrine and/or autocrine manner. Adipokines act via their receptors on mammary tumor cells to i) influence tumor cell proliferation, migration and invasion in breast cancer; ii) regulate the production of epithelial-derived proteins, angiogenic proteins and growth factors; iii) stimulate other cells in the tumor microenvironment to invade and proliferate. For example, a study reported a proliferative effect of adipocyte-secreted factors on the MCF-7 breast cancer cell line, through the regulation of genes involved in cell motility, migration, survival, apoptosis and angiogenesis (32). In the same way, Celis et al(33) identified in mammary adipose tissue 359 protein components and excreted factors that may provide insight into the close interplay between mammary epithelium, stroma and fat tissue. Among these proteins, they identified several cell cycle regulators, including p53 and p21. Perera et al(34) used a proteomic approach and described for the first time the secretion of epithelial-derived proteins in MCF-7 cells in response to leptin. The secretion of such proteins in breast cancer cells in response to ZAG has yet to be reported.

In summary, these preliminary data show that recombinant ZAG has a pro-carcinogenic effect on breast cancer cells and conversely an anti-carcinogenic effect on non malignant breast cells. ZAG clearly modulates signaling pathways involved in proliferation and apoptosis but this modulation cannot fully explain the effects we previously observed with ZAG in breast cancer tissues.

References

1 

Klein S, Wadden T and Sugerman HJ: AGA technical review on obesity. Gastroenterology. 123:882–932. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Lorincz AM and Sukumar S: Molecular links between obesity and breast cancer. Endocr Relat Cancer. 13:279–292. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Wolk A, Gridley G, Svensson M, Nyren O, McLaughlin JK, Fraumeni JF and Adam HO: A prospective study of obesity and cancer risk (Sweden). Cancer Causes Control. 12:13–21. 2001. View Article : Google Scholar

4 

Chlebowski RT, Aiello E and McTiernan A: Weight loss in breast cancer patient management. J Clin Oncol. 20:1128–1143. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Daling JR, Malone KE, Doody DR, Johnson LG, Gralow JR and Porter PL: Relation of body mass index to tumor markers and survival among young women with invasive ductal breast carcinoma. Cancer. 92:720–729. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Belfiore A and Frasca F: IGF and insulin receptor signaling in breast cancer. J Mammary Gland Biol Neoplasia. 13:381–406. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Cold S, Hansen S, Overvad K and Rose C: A woman’s build and the risk of breast cancer. Eur J Cancer. 34:1163–1174. 1998.

8 

Pischon T, Nothlings U and Boeing H: Obesity and cancer. Proc Nutr Soc. 67:128–145. 2008. View Article : Google Scholar

9 

Fantuzzi G: Adipose tissue, adipokines, and inflammation. J Allergy Clin Immunol. 115:911–919. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Tilg H and Moschen AR: Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. 6:772–783. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Perrier S, Caldefie-Chezet F and Vasson MP: IL-1 family in breast cancer: potential interplay with leptin and other adipocytokines. FEBS Lett. 583:259–265. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Rose DP, Komninou D and Stephenson GD: Obesity, adipocytokines, and insulin resistance in breast cancer. Obes Rev. 5:153–165. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Schaffler A, Scholmerich J and Buechler C: Mechanisms of disease: adipokines and breast cancer - endocrine and paracrine mechanisms that connect adiposity and breast cancer. Nat Clin Pract Endocrinol Metab. 3:345–354. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Vona-Davis L, Howard-McNatt M and Rose DP: Adiposity, type 2 diabetes and the metabolic syndrome in breast cancer. Obes Rev. 8:395–408. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Jarde T, Caldefie-Chezet F, Damez M, Mishellany F, Perrone D, Penault-Llorca F, Guillot J and Vasson MP: Adiponectin and leptin expression in primary ductal breast cancer and in adjacent healthy epithelial and myoepithelial tissue. Histopathology. 53:484–487. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Jarde T, Caldefie-Chezet F, Goncalves-Mendes N, Mishellany F, Buechler C, Penault-Llorca F and Vasson MP: Involvement of adiponectin and leptin in breast cancer: clinical and in vitro studies. Endocr Relat Cancer. 16:1197–1210. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Jarde T, Perrier S, Vasson MP and Caldefie-Chezet F: Molecular mechanisms of leptin and adiponectin in breast cancer. Eur J Cancer. 47:33–43. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Bing C, Bao Y, Jenkins J, Sanders P, Manieri M, Cinti S, Tisdale MJ and Trayhurn P: Zinc-alpha2-glycoprotein, a lipid mobilizing factor, is expressed in adipocytes and is up-regulated in mice with cancer cachexia. Proc Natl Acad Sci USA. 101:2500–2505. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Marrades MP, Martinez JA and Moreno-Aliaga MJ: ZAG, a lipid mobilizing adipokine, is downregulated in human obesity. J Physiol Biochem. 64:61–66. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Mracek T, Ding Q, Tzanavari T, Kos K, Pinkney J, Wilding J, Trayhurn P and Bing C: The adipokine zinc-alpha2-glycoprotein (ZAG) is downregulated with fat mass expansion in obesity. Clin Endocrinol. 72:334–341. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Bundred NJ, Miller WR and Walker RA: An immunohistochemical study of the tissue distribution of the breast cyst fluid protein, zinc alpha 2 glycoprotein. Histopathology. 11:603–610. 1987. View Article : Google Scholar : PubMed/NCBI

22 

Diez-Itza I, Sanchez LM, Allende MT, Vizoso F, Ruibal A and Lopez-Otin C: Zn-alpha 2-glycoprotein levels in breast cancer cytosols and correlation with clinical, histological and biochemical parameters. Eur J Cancer. 29A:1256–1260. 1993. View Article : Google Scholar : PubMed/NCBI

23 

Bundred NJ, Scott WN, Davies SJ, Miller WR and Mansel RE: Zinc alpha-2 glycoprotein levels in serum and breast fluids: a potential marker of apocrine activity. Eur J Cancer. 27:549–552. 1991. View Article : Google Scholar : PubMed/NCBI

24 

Sanchez LM, Vizoso F, Diez-Itza I and Lopez-Otin C: Identification of the major protein components in breast secretions from women with benign and malignant breast diseases. Cancer Res. 52:95–100. 1992.PubMed/NCBI

25 

Dubois V, Delort L, Mishellany F, Jarde T, Billard H, Lequeux C, Damour O, Penault-Llorca F, Vasson MP and Caldefie-Chezet F: Zinc-alpha2-glycoprotein: a new biomarker of breast cancer? Anticancer Res. 30:2919–2925. 2010.PubMed/NCBI

26 

Meyer N, Kim SS and Penn LZ: The Oscar-worthy role of Myc in apoptosis. Semin Cancer Biol. 16:275–287. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Prendergast GC: Mechanisms of apoptosis by c-Myc. Oncogene. 18:2967–2987. 1999. View Article : Google Scholar : PubMed/NCBI

28 

Sakamuro D, Eviner V, Elliott KJ, Showe L, White E and Prendergast GC: c-Myc induces apoptosis in epithelial cells by both p53-dependent and p53-independent mechanisms. Oncogene. 11:2411–2418. 1995.PubMed/NCBI

29 

Lee YJ, Galoforo SS, Berns CM, Tong WP, Kim HR and Corry PM: Glucose deprivation-induced cytotoxicity in drug resistant human breast carcinoma MCF-7/ADR cells: role of c-myc and bcl-2 in apoptotic cell death. J Cell Sci. 110:681–686. 1997.PubMed/NCBI

30 

He N, Brysk H, Tyring SK, Ohkubo I and Brysk MM: Zinc-alpha(2)-glycoprotein hinders cell proliferation and reduces cdc2 expression. J Cell Biochem Suppl. 36:162–169. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Zapata JM, Krajewska M, Krajewski S, Huang RP, Takayama S, Wang HG, Adamson E and Reed JC: Expression of multiple apoptosis-regulatory genes in human breast cancer cell lines and primary tumors. Breast Cancer Res Treat. 47:129–140. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Iyengar P, Combs TP, Shah SJ, Gouon-Evans V, Pollard JW, Albanese C, Flanagan L, Tenniswood MP, Guha C, Lisanti MP, Pestell RG and Scherer PE: Adipocyte-secreted factors synergistically promote mammary tumorigenesis through induction of anti-apoptotic transcriptional programs and proto-oncogene stabilization. Oncogene. 22:6408–6423. 2003. View Article : Google Scholar

33 

Celis JE, Moreira JM, Cabezon T, Gromov P, Friis E, Rank F and Gromova I: Identification of extracellular and intracellular signaling components of the mammary adipose tissue and its interstitial fluid in high risk breast cancer patients: toward dissecting the molecular circuitry of epithelial-adipocyte stromal cell interactions. Mol Cell Proteomics. 4:492–522. 2005. View Article : Google Scholar

34 

Perera CN, Spalding HS, Mohammed SI and Camarillo IG: Identification of proteins secreted from leptin stimulated MCF-7 breast cancer cells: a dual proteomic approach. Exp Biol Med. 233:708–720. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May 2013
Volume 29 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Delort L, Perrier S, Dubois V, Billard H, Mracek T, Bing C, Vasson M and Caldefie-Chézet F: Zinc-α2-glycoprotein: A proliferative factor for breast cancer? In vitro study and molecular mechanisms. Oncol Rep 29: 2025-2029, 2013
APA
Delort, L., Perrier, S., Dubois, V., Billard, H., Mracek, T., Bing, C. ... Caldefie-Chézet, F. (2013). Zinc-α2-glycoprotein: A proliferative factor for breast cancer? In vitro study and molecular mechanisms. Oncology Reports, 29, 2025-2029. https://doi.org/10.3892/or.2013.2311
MLA
Delort, L., Perrier, S., Dubois, V., Billard, H., Mracek, T., Bing, C., Vasson, M., Caldefie-Chézet, F."Zinc-α2-glycoprotein: A proliferative factor for breast cancer? In vitro study and molecular mechanisms". Oncology Reports 29.5 (2013): 2025-2029.
Chicago
Delort, L., Perrier, S., Dubois, V., Billard, H., Mracek, T., Bing, C., Vasson, M., Caldefie-Chézet, F."Zinc-α2-glycoprotein: A proliferative factor for breast cancer? In vitro study and molecular mechanisms". Oncology Reports 29, no. 5 (2013): 2025-2029. https://doi.org/10.3892/or.2013.2311