Nitric oxide synthase and breast cancer: role of TIMP-1 in NO-mediated Akt activation

Lisa A Ridnour, Kimberly M Barasch, Alisha N Windhausen, Tiffany H Dorsey, Michael M Lizardo, Harris G Yfantis, Dong H Lee, Christopher H Switzer, Robert Y S Cheng, Julie L Heinecke, Ernst Brueggemann, Harry B Hines, Chand Khanna, Sharon A Glynn, Stefan Ambs, David A Wink, Lisa A Ridnour, Kimberly M Barasch, Alisha N Windhausen, Tiffany H Dorsey, Michael M Lizardo, Harris G Yfantis, Dong H Lee, Christopher H Switzer, Robert Y S Cheng, Julie L Heinecke, Ernst Brueggemann, Harry B Hines, Chand Khanna, Sharon A Glynn, Stefan Ambs, David A Wink

Abstract

Prediction of therapeutic response and cancer patient survival can be improved by the identification of molecular markers including tumor Akt status. A direct correlation between NOS2 expression and elevated Akt phosphorylation status has been observed in breast tumors. Tissue inhibitor matrix metalloproteinase-1 (TIMP-1) has been proposed to exert oncogenic properties through CD63 cell surface receptor pathway initiation of pro-survival PI3k/Akt signaling. We employed immunohistochemistry to examine the influence of TIMP-1 on the functional relationship between NOS2 and phosphorylated Akt in breast tumors and found that NOS2-associated Akt phosphorylation was significantly increased in tumors expressing high TIMP-1, indicating that TIMP-1 may further enhance NO-induced Akt pathway activation. Moreover, TIMP-1 silencing by antisense technology blocked NO-induced PI3k/Akt/BAD phosphorylation in cultured MDA-MB-231 human breast cancer cells. TIMP-1 protein nitration and TIMP-1/CD63 co-immunoprecipitation was observed at NO concentrations that induced PI3k/Akt/BAD pro-survival signaling. In the survival analysis, elevated tumor TIMP-1 predicted poor patient survival. This association appears to be mainly restricted to tumors with high NOS2 protein. In contrast, TIMP-1 did not predict poor survival in patient tumors with low NOS2 expression. In summary, our findings suggest that tumors with high TIMP-1 and NOS2 behave more aggressively by mechanisms that favor Akt pathway activation.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Expression of TIMP-1 and CD63…
Figure 1. Expression of TIMP-1 and CD63 in human breast tumors.
Shown is immunohistochemical staining for TIMP-1 (A–C) and CD63 (D–F). (A) Strong staining of TIMP-1 is shown in the tumor epithelial cells. The surrounding tissue consisting of stromal cells and adipocytes is negative for TIMP-1. (B) Moderate staining of TIMP-1 in tumor epithelial cells is shown in the lower center and to the right. Normal epithelial cells are negative for TIMP-1 (upper left corner). (C) Normal epithelial cells in surrounding non-tumor tissue of an invasive breast tumor are negative for TIMP-1. (D) Strong staining of CD63 in the tumor epithelium. The surrounding tissue consisting of stromal cells and a benign breast duct is mostly negative for CD63. (E) Moderate staining of CD63 in tumor epithelial cells (center). (F) Moderate to strong staining of CD63 in tumor epithelial cells (lower and upper center). Normal epithelial cells to the left and stromal cells are negative for CD63. Magnification: 100× for A, C, D, F; 200× for B and E. Counterstain hematoxylin.
Figure 2. TIMP-1 predicts poor breast cancer…
Figure 2. TIMP-1 predicts poor breast cancer survival in patients whose tumors express elevated NOS2 but not in those with low NOS2 expression.
Kaplan-Meier survival analysis demonstrating reduced breast cancer specific survival in A) patients whose tumors express elevated TIMP-1 protein and B) a similar trend when stratified for tumors with high TIMP-1 and high NOS2 protein levels. C) In contrast, TIMP-1 did not predict poor survival of patients with low NOS2 tumor expression.
Figure 3. Transfection of NOS2 expression plamid…
Figure 3. Transfection of NOS2 expression plamid enhances pAkt.
NOS2 overexpressing MDA-MB-231 breast cancer cells treated with L-Arginine show increased pAkt. Treatment of the same NOS2 transfected cells with the NOS2 inhibitor aminoguanidine (AG) show no increase in pAkt.
Figure 4. Temporal steady state NO flux…
Figure 4. Temporal steady state NO flux generated by the NO Donor DETA/NO.
Steady state nM NO levels distributed evenly throughout cell culture media as a function of NO donor (DETA/NO) concentration measured through 24 hr.
Figure 5. Western blots demonstrate the requirement…
Figure 5. Western blots demonstrate the requirement of TIMP-1 for NO-induced Akt phosphorylation in MDA-MB-231 breast cancer cells.
A) Suppression of TIMP-1 protein translation using anti-sense TIMP-1 morpholino through 72 hr. B) TIMP-1 suppression (TIMP-1kd) abolishes NO-induced Akt phosphorylation and reduces the extent of NO-mediated phosphorylation of Akt downstream pro-apoptotic target BAD. The cells were incubated with TIMP-1 morpholino as described in the methods section, serum starved, and then treated with increasing concentrations of DETA/NO for 24 hr. The western blots shown were stripped and reblotted for total Akt are representative of two independent experiments. C–D) Densitometry measurements of pAkt (C) and pBAD (D) relative to total Akt loading control were plotted vs. DETA/NO concentration. Linear regression pAkt/total Akt: control, r2 = 0.974, y-intercept = 90.40+/−10.35, slope = 0.2187+/−0.021 p = 0.0018; TIMP-1 kd, r2 = 0.573, y-intercept = 76.04+/−14.61, slope = 0.0583+/−0.029, p = 0.1381. Linear regression pBAD/total Akt: control, r2 = 0.988, y-intercept = 91.45+/−29.29, slope = 0.9053+/−0.058, p = 0.0006; TIMP-1 kd, r2 = 0.953, y-intercept = 44.60+/−24.30, slope = 0.3747+/−0.048, p = 0.0027.
Figure 6. NO modulation of TIMP-1 protein…
Figure 6. NO modulation of TIMP-1 protein levels.
A) NO does not significantly effect steady state TIMP-1 mRNA as shown by real time PCR (n = 5). B) NO modulates TIMP-1 protein in the media from MDA-MB-231 breast cancer cells as shown by ELISA assay. The results are presented as mean+/−SEM of n = 4 samples. C) NO modulates TIMP-1 protein in the media from human fibroblasts as shown by western blot.
Figure 7. Confocal image microscopy shows effects…
Figure 7. Confocal image microscopy shows effects of NO on CD63 and TIMP-1 protein co-localization in MDA-MB-231 breast cancer cells treated with DETA/NO.
CD63 (red fluorescence) and TIMP-1 (green fluorescence) in MDA-MB-231 breast cancer cells treated for 24 hr with the NO donor DETA/NO at the following concentrations; A) control, B) 100 µM, C) 500 µM, D) 1000 µM. Panels E and F are the enlarged areas circled in panels C and D, respectively, which show areas of CD63 and TIMP-1 protein co-localization (yellow/orange fluorescence). Cellular nuclei are visualized as blue fluorescence (DAPI). Scale bar = 20 µm for all panels.
Figure 8. Mass spectrum showing nitration of…
Figure 8. Mass spectrum showing nitration of two tyrosine residues (Y95, Y143) in human recombinant TIMP-1 protein treated with 100, 500, or 1000 µM DETA/NO.
A) amino acid sequence of human recombinant TIMP-1 protein (NCBI Reference Sequence: NP_003245.1) and location of tyrosine nitration (Y95, Y143). B) representative mass spectrum of a nitrated peptide from human TIMP-1, illustrating the mass shift of 45 daltons that accompanied nitration.
Figure 9. TIMP-1 immunoprecipitation of DETA/NO treated…
Figure 9. TIMP-1 immunoprecipitation of DETA/NO treated MDA-MB-231 cell lysates.
A) Western blot showing TIMP-1 protein expression in the lysates of DETA/NO treated MDA-MB-231 breast cancer cells. B) TIMP-1 immunoprecipitated from DETA/NO treated MDA-MB-231 cells and immunoblotted for 3′NT and CD63 demonstrates enhanced TIMP-1 tyrosine nitration and CD63 co-immunoprecipitation associated with DETA/NO exposure.
Figure 10. DETA/NO induced PI3k p85 phosphorylation…
Figure 10. DETA/NO induced PI3k p85 phosphorylation is suppressed in TIMP-1 knockdown cells.
Figure 11. NOS2/NO promotes TIMP-1-dependent pro-survival signaling…
Figure 11. NOS2/NO promotes TIMP-1-dependent pro-survival signaling through Akt activation in the breast tumor microenvironment.

References

    1. American Cancer Society Facts and Figures (2010) Available: .
    1. Lackner MR (2010) Prospects for personalized medicine with inhibitors targeting the RAS and PI3K pathways. Expert Rev Mol Diagn 10: 75–87.
    1. Steelman LS, Stadelman KM, Chappell WH, Horn S, Bäsecke J, et al. (2008) Akt as a therapeutic target in cancer. Expert Opin Ther Targets 12: 1139–65.
    1. Zhou X, Tan M, Stone Hawthorne V, Klos KS, Lan KH, et al. (2004) Activation of the Akt/mammalian target of rapamycin/4E-BP1 pathway by ErbB2 overexpression predicts tumor progression in breast cancers. Clin Cancer Res 10: 6779–88.
    1. Pérez-Tenorio G, Stål O (2002) Activation of AKT/PKB in breast cancer predicts a worse outcome among endocrine treated patients. Br J Cancer 86: 540–5.
    1. Lambert E, Boudot C, Kadri Z, Soula-Rothhut M, Sowa ML, et al. (2003) Tissue inhibitor of metalloproteinases-1 signaling pathway leading to erythroid cell survival. Biochem J 372: 767–74.
    1. Chirco R, Liu XW, Jung KK, Choi Kim HR (2006) Novel functions of TIMPs in cell signaling. Cancer Met Rev 25: 99–113.
    1. Würtz SO, Schrohl AS, Mouridsen H, Brünner N (2008) TIMP-1 as a tumor marker in breast cancer–an update. Acta Oncol 47: 580–90.
    1. Jung KK, Liu XW, Chirco R, Fridman R, Kim HR (2006) Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein. EMBO J 25: 3934–42.
    1. Lis S, Hua ZC (2008) FAK expression regulation and therapeutic potential. Adv Cancer Res 101: 45–61.
    1. Zhao J, Guan JL (2009) Signal transduction by focal adhesion kinase in cancer. Cancer Met Rev 28: 35–49.
    1. Schrohl AS, Meijer-van Gelder ME, Holten-Andersen MN, Christensen IJ, Look MP, et al. (2006) Primary tumor levels of tissue inhibitor of metalloproteinases-1 are predictive of resistance to chemotherapy in patients with metastatic breast cancer. Clin Cancer Res 12: 7054–8.
    1. Sørensen NM, Byström P, Christensen IJ, Berglund A, Nielsen HJ, et al. (2007) TIMP-1 is significantly associated with objective response and survival in metastatic colorectal cancer patients receiving combination of irinotecan, 5-fluorouracil, and folinic acid. Clin Cancer Res 13: 4117–22.
    1. Lipton A, Leitzel K, Chaudri-Ross HA, Evans DB, Ali SM, et al. (2008) Serum TIMP-1 and Response to the Aromatase Inhibitor Letrozole versus Tamoxifen in Metastatic Breast Cancer. J Clin Oncol 26: 2653–58.
    1. Lipton A, Ali SM, Leitzel K, Demers L, Evans DB, et al. (2007) Elevated plasma tissue inhibitor of metalloproteinase-1 level predicts decreased response and survival in metastatic breast cancer. Cancer 109: 1933–39.
    1. Davidsen ML, Würtz SO, Romer MU, Sorensen NM, Johansen SK, et al. (2006) TIMP-1 gene deficiency increases tumour cell sensitivity to chemotherapy-induced apoptosis. Br J Cancer 95: 1114–20.
    1. Liu XW, Taube ME, Jung KK, Dong Z, Lee YJ, et al. (2005) Tissue inhibitor of metalloproteinase-1 protects human breast epithelial cells from extrinsic cell death: a potential oncogenic activity of tissue inhibitor of metalloproteinase-1. Cancer Res 65: 898–906.
    1. Bigelow RL, Williams BJ, Carroll JL, Daves LK, Cardelli JA (2009) TIMP-1 overexpression promotes tumorigenesis of MB-231 breast cancer cells and alters expression of a subset of cancer promoting genes in vivo distinct fro those observed in vitro. Breast Cancer Res Treat 117: 31–44.
    1. Kundu JK, Surh YJ (2008) Inflammation: gearing the journey to cancer. Mut Res 659: 15–30.
    1. Wink DA, Ridnour LA, Hussain PS, Harris CC (2008) The reemergence of nitric oxide and cancer. Nitric oxide 19: 65–67.
    1. Ridnour LA, Thomas DD, Switzer C, Flores-Santana W, Isenberg JS, et al. (2008) Molecular mechanisms for discrete nitric oxide levels in cancer. Nitric oxide 19: 73–76.
    1. Loibl S, Buck A, Strank C, von Minckwitz G, Roller M, et al. (2005) The role of early expression of inducible nitric oxide synthase in human breast cancer. Eur J Cancer 41: 265–71.
    1. Ekmekcioglu S, Ellerhorst J, Smid CM, Prieto VG, Munsell M, et al. (2000) Inducible nitric oxide synthase and nitrotyrosine in human metastatic melanoma tumors correlate with poor survival. Clin Cancer Res 6: 4768–75.
    1. Raspollini MR, Amunni G, Villanucci A, Boddi V, Baroni G, et al. (2004) Expression of inducible nitric oxide synthase and cyclooxygenase-2 in ovarian cancer: correlation with clinical outcome. Gynecol Oncol 92: 806–12.
    1. Connelly ST, Macabeo-Ong M, Dekker N, Jordan RC, Schmidt BL (2005) Increased nitric oxide levels and iNOS over-expression in oral squamous cell carcinoma. Oral Oncol 41: 261–267.
    1. Brennan PA, Dennis S, Poller D, Quintero M, Puxeddu R, et al. (2008) Inducible nitric oxide synthase: correlation with extracapsular spread and enhancement of tumor cell invasion in head and neck squamous cell carcinoma. Head Neck 30: 208–14.
    1. Cronauer MV, Ince Y, Engers R, Rinnab L, Weidemann W, et al. (2007) Nitric oxide-mediated inhibition of androgen receptor activity: possible implications for prostate cancer progression. Oncogene 26: 1875–84.
    1. Glynn SA, Boersma BJ, Dorsey TH, Yi M, Yfantis HG, et al. (2010) Increased NOS2 predicts poor survival in estrogen receptor-negative breast cancer patients. J Clin Invest 120: 3843–54.
    1. Prueitt RL, Boersma BJ, Howe TM, Goodman JE, Thomas DD, et al. (2006) Inflammation and IGF-I activate the Akt pathway in breast cancer. Int J Cancer 120: 796–805.
    1. Ridnour LA, Windhausen AN, Isenberg JS, Yeung N, Thomas DD, et al. (2007) Nitric oxide regulates matrix metalloproteinase-9 activity by guanylyl-cyclase-dependent and –independent pathways. Proc Natl Acad Sci U S A 104: 16898–903.
    1. Death AK, Nakhla S, McGrath KC, Martell S, Yue DK, et al. (2002) Nitroglycerin upregulates matrix metalloproteinase expression by human macrophages. J Am Coll Cardiol 39: 1943–50.
    1. Clemons NJ, Shannon NB, Abeyratne LR, Walker CE, Saadi A, et al. (2010) Nitric oxide-mediated invasion in Barrett's high-grade dysplasia and adenocarcinoma. Carcinogenesis 31: 1669–75.
    1. Hsu YC, Wang LF, Chien YW, Lee WR (2007) Induction of TIMP-1 and HSP47 synthesis in primary keloid fibroblasts by exogenous nitric oxide. J Dermatological Science 45: 37–44.
    1. Pervin S, Singh R, Hernandez E, Wu G, Chaudhuri G (2007) Nitric oxide in physiologic concentrations targets the translational machinery to increase the proliferation of human breast cancer cells: involvement of mammalian target of rapamycin/elF4E pathway. Cancer Res 67: 289–99.
    1. Switzer CH, Ridnour LA, Cheng RY, Sparatore A, Del Soldato P, et al. (2009) Dithiolethione compounds inhibit Akt signaling in human breast and lung cancer cells by increasing PP2A activity. Oncogene 28: 3837–46.
    1. Switzer CH, Cheng RYS, Ridnour LA, Murray MC, Tazzari V, et al. (2012) Dithiolethiones inhibit NF-kB activity via covalent modification in human estrogen receptor-negative breast cancer. Cancer Res 72: 2394–404.
    1. Nakagawa SA, Lopes A, Lopes de Carvalho A, Rossi BM, Werneck da Cunha I, et al. (2010) Nitric oxide synthases, cyclooxygenase-2, nitrotyrosine, and angiogenesis in chondrosarcoma and their relation to prognosis. J Bone Joint Surg Am 92: 1738–46.
    1. Campos AH, Aldred VL, Ribeiro KC, Vassallo J, Soares FA (2009) Role of immunoexpression of nitric oxide synthases by Hodgkin and Reed-Sternberg cells on apoptosis deregulation and on clinical outcome of classical Hodgkin lymphoma. Mol Cell Biochem 321: 95–102.
    1. Ambs S, Glynn SA (2011) Candidate pathways linking inducible nitric oxide synthase to a basal-like transcription pattern and tumor progression in human breast cancer. Cell Cycle 10: 619–624.
    1. Lambert E, Bridoux L, Devy J, Dassé E, Sowa ML, et al. (2009) TIMP-1 binding to proMMP-9/CD44 complex localized at the cell surface promotes erythroid cell survival. Int J Biochem Cell Biol 41: 1102–15.
    1. Charafe-Jauffret E, Ginestier C, Monville F, Finetti P, Adélaïde J, et al. (2006) Gene expression profiling of breast cell lines identifies potential new basal markers. Oncogene 25: 2273–84.
    1. Espey MG, Miranda KM, Pluta RM, Wink DA (2000) Nitrosative capacity of macrophages is dependent on nitric-oxide synthase induction signals. J Biol Chem 275: 11341–7.
    1. Thomas DD, Espey MG, Ridnour LA, Hofseth LJ, Mancardi D, et al. (2004) Hypoxic inducible factor 1 alpha, extracellular signal-regulated kinase, and p53 are regulated by distinct threshold concentrations of nitric oxide. Proc Natl Acad Sci U S A 101: 8894–9.
    1. Ridnour LA, Isenberg JS, Espey MG, Thomas DD, Roberts DD, et al. (2005) Nitric oxide regulates angiogenesis through a functional switch involving thrombospondin-1. Proc Natl Acad Sci U S A 102: 13147–52.
    1. Simon DI, Mullins ME, Jia L, Gaston B, Singel DJ, et al. (1996) Polynitrosylated proteins: Characterization, bioactivity, and functional consequences. Proc Natl Acad Sci U S A 93: 4736–41.
    1. Wink DA, Nims RW, Darbyshire JF, Christodololou D, Handbauer I, et al. (1994) Reaction kinetics for nitrosation of cysteine and glutathione in aerobic nitric oxide solutions at neutral pH. Insights into the fate and physiological effects of intermediates generated in the NO/O2 reaction. Chem Res Tox 7: 519–25.
    1. Patruno A, Pesce M, Marrone A, Speranza L, Grilli A, et al. (2012) Activity of matrix metalloproteinases (MMPs) and the tissue inhibitor of MMP (TIMP-1) in electromagnetic field-exposed THP-1 cells. J Cell Physiol 227: 2767–74.
    1. Kim JE, Tannenbaum SR (2004) S-Nitrosation regulates the activation of endogenous procaspase-9 in HT-29 human colon carcinoma cells. J Biol Chem 279: 9758–64.
    1. Sumbayev VV (2008) PI3 kinase and direct S-nitrosation are involved in down regulation of apoptosis signal-regulating kinase 1 during LPS-induced Toll-like receptor 4 signaling. Immunology Letters 115: 126–30.
    1. Li G, Fridman R, Kim HRC (1999) Tissue inhibitor of metalloproteinase-1 inhibits apoptosis of human breast epithelial cells. Cancer Res 59: 6267–75.
    1. Duarte S, Hamada T, Kuriyama N, Busuttil RW, Coito AJ (2012) Tissue inhibitor of metalloproteinases-1 (TIMP-1) deficiency leads to lethal partial hepatic ischemia and reperfusion injury. Hepatology In press.
    1. Krejci P, Prochazkova J, Smutny J, Chlebova K, Lin P, et al. (2010) FGFR3 signaling induces a reversible senescence phenotype in chondrocytes similar to oncogene-induced premature senescence. Bone 47: 102–10.
    1. Sohn JJ, Schetter AJ, Yfantis HG, Ridnour LA, Horikawa I, et al. (2012) Macrophages, nitric oxide and microRNAs are associated with DNA damage response pathway and senescence in inflammatory bowel disease. PLoS ONE In press.
    1. Stetler-Stevenson WG (2008) Tissue inhibitors of metalloproteinases in cell signaling: metalloproteinase-independent biological activities. Sci Signal 1: 1–9.
    1. Dilley TK, Bowden T, Chen QM (2003) Novel mechanisms of sublethal oxidant toxicity: induction of premature senescence in human fibroblasts confers tumor promoter activity. Exp Cell Research 290: 38–48.
    1. Martin DN, Boersma BJ, Yi M, Reimers M, Howe TM, et al. (2009) Differences in the Tumor Microenvironment between African-American and European-American Breast Cancer Patients. Plos ONE 4: 1–14.
    1. Abramoff MD, Magalhaes PJ, Ram SJ (2004) Image Processing with ImageJ. Biophotonics Interactional 11: 36–42.

Source: PubMed

3
Prenumerera