<iframe src="//www.googletagmanager.com/ns.html?id=GTM-5TSRKG" height="0" width="0" style="display: none; visibility: hidden">
Review Article
No access
Published Online: 27 February 2014

Redox-Mediated and Ionizing-Radiation-Induced Inflammatory Mediators in Prostate Cancer Development and Treatment

Publication: Antioxidants & Redox Signaling
Volume 20, Issue Number 9

Abstract

Significance: Radiation therapy is widely used for treatment of prostate cancer. Radiation can directly damage biologically important molecules; however, most effects of radiation-mediated cell killing are derived from the generated free radicals that alter cellular redox status. Multiple proinflammatory mediators can also influence redox status in irradiated cells and the surrounding microenvironment, thereby affecting prostate cancer progression and radiotherapy efficiency. Recent Advances: Ionizing radiation (IR)–generated oxidative stress can regulate and be regulated by the production of proinflammatory mediators. Depending on the type and stage of the prostate cancer cells, these proinflammatory mediators may lead to different biological consequences ranging from cell death to development of radioresistance. Critical Issues: Tumors are heterogeneous and dynamic communication occurs between stromal and prostate cancer cells, and complicated redox-regulated mechanisms exist in the tumor microenvironment. Thus, antioxidant and anti-inflammatory strategies should be carefully evaluated for each patient at different stages of the disease to maximize therapeutic benefits while minimizing unintended side effects. Future Directions: Compared with normal cells, tumor cells are usually under higher oxidative stress and secrete more proinflammatory mediators. Thus, redox status is often less adaptive in tumor cells than in their normal counterparts. This difference can be exploited in a search for new cancer therapeutics and treatment regimes that selectively activate cell death pathways in tumor cells with minimal unintended consequences in terms of chemo- and radio-resistance in tumor cells and toxicity in normal tissues. Antioxid. Redox Signal. 20, 1481–1500.

Get full access to this article

View all available purchase options and get full access to this article.

References

1.
Adler HL, McCurdy MA, Kattan MW, Timme TL, Scardino PT, and Thompson TC. Elevated levels of circulating interleukin-6 and transforming growth factor-beta1 in patients with metastatic prostatic carcinoma. J Urol 161: 182–187, 1999.
2.
Allen M and Louise Jones J. Jekyll and Hyde: the role of the microenvironment on the progression of cancer. J Pathol 223: 162–176, 2011.
3.
Alvarez B, Quinn LS, Busquets S, Quiles MT, Lopez-Soriano FJ, and Argiles JM. Tumor necrosis factor-alpha exerts interleukin-6-dependent and -independent effects on cultured skeletal muscle cells. Biochim Biophys Acta 1542: 66–72, 2002.
4.
Andarawewa KL, Erickson AC, Chou WS, Costes SV, Gascard P, Mott JD, Bissell MJ, and Barcellos-Hoff MH. Ionizing radiation predisposes nonmalignant human mammary epithelial cells to undergo transforming growth factor beta induced epithelial to mesenchymal transition. Cancer Res 67: 8662–8670, 2007.
5.
Andarawewa KL, Paupert J, Pal A, and Barcellos-Hoff MH. New rationales for using TGFbeta inhibitors in radiotherapy. Int J Radiat Biol 83: 803–811, 2007.
6.
Anscher MS, Thrasher B, Rabbani Z, Teicher B, and Vujaskovic Z. Antitransforming growth factor-beta antibody 1D11 ameliorates normal tissue damage caused by high-dose radiation. Int J Radiat Oncol Biol Phys 65: 876–881, 2006.
7.
Araki S, Omori Y, Lyn D, Singh RK, Meinbach DM, Sandman Y, Lokeshwar VB, and Lokeshwar BL. Interleukin-8 is a molecular determinant of androgen independence and progression in prostate cancer. Cancer Res 67: 6854–6862, 2007.
8.
Argiles JM, Busquets S, Toledo M, and Lopez-Soriano FJ. The role of cytokines in cancer cachexia. Curr Opin Support Palliat Care 3: 263–268, 2009.
9.
Azevedo A, Cunha V, Teixeira AL, and Medeiros R. IL-6/IL-6R as a potential key signaling pathway in prostate cancer development. World J Clin Oncol 2: 384–396, 2011.
10.
Baker AM, Oberley LW, and Cohen MB. Expression of antioxidant enzymes in human prostatic adenocarcinoma. Prostate 32: 229–233, 1997.
11.
Balkwill F. Tumour necrosis factor and cancer. Nat Rev Cancer 9: 361–371, 2009.
12.
Barcellos-Hoff MH and Dix TA. Redox-mediated activation of latent transforming growth factor-beta 1. Mol Endocrinol 10: 1077–1083, 1996.
13.
Barcellos-Hoff MH, Park C, and Wright EG. Radiation and the microenvironment—tumorigenesis and therapy. Nat Rev Cancer 5: 867–875, 2005.
14.
Basu HS, Thompson TA, Church DR, Clower CC, Mehraein-Ghomi F, Amlong CA, Martin CT, Woster PM, Lindstrom MJ, and Wilding G. A small molecule polyamine oxidase inhibitor blocks androgen-induced oxidative stress and delays prostate cancer progression in the transgenic adenocarcinoma of the mouse prostate model. Cancer Res 69: 7689–7695, 2009.
15.
Baud V and Karin M. Is NF-kappaB a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discov 8: 33–40, 2009.
16.
Bensalah K, Lotan Y, Karam JA, and Shariat SF. New circulating biomarkers for prostate cancer. Prostate Cancer Prostatic Dis 11: 112–120, 2008.
17.
Bierie B and Moses HL. TGF-beta and cancer. Cytokine Growth Factor Rev 17: 29–40, 2006.
18.
Bierie B and Moses HL. Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer 6: 506–520, 2006.
19.
Biswas S, Guix M, Rinehart C, Dugger TC, Chytil A, Moses HL, Freeman ML, and Arteaga CL. Inhibition of TGF-beta with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J Clin Invest 117: 1305–1313, 2007.
20.
Bonfil RD, Chinni S, Fridman R, Kim HR, and Cher ML. Proteases, growth factors, chemokines, and the microenvironment in prostate cancer bone metastasis. Urol Oncol 25: 407–411, 2007.
21.
Bostwick DG, Alexander EE, Singh R, Shan A, Qian J, Santella RM, Oberley LW, Yan T, Zhong W, Jiang X, and Oberley TD. Antioxidant enzyme expression and reactive oxygen species damage in prostatic intraepithelial neoplasia and cancer. Cancer 89: 123–134, 2000.
22.
Bostwick DG, Meiers I, and Shanks JH. Glutathione S-transferase: differential expression of alpha, mu, and pi isoenzymes in benign prostate, prostatic intraepithelial neoplasia, and prostatic adenocarcinoma. Hum Pathol 38: 1394–1401, 2007.
23.
Boudreau HE, Casterline BW, Rada B, Korzeniowska A, and Leto TL. Nox4 involvement in TGF-beta and SMAD3-driven induction of the epithelial-to-mesenchymal transition and migration of breast epithelial cells. Free Radic Biol Med 53: 1489–1499, 2012.
24.
Bouquet F, Pal A, Pilones KA, Demaria S, Hann B, Akhurst RJ, Babb JS, Lonning SM, DeWyngaert JK, Formenti SC, and Barcellos-Hoff MH. TGFbeta1 inhibition increases the radiosensitivity of breast cancer cells in vitro and promotes tumor control by radiation in vivo. Clin Cancer Res 17: 6754–6765, 2011.
25.
Bouraoui Y, Ricote M, Garcia-Tunon I, Rodriguez-Berriguete G, Touffehi M, Rais NB, Fraile B, Paniagua R, Oueslati R, and Royuela M. Pro-inflammatory cytokines and prostate-specific antigen in hyperplasia and human prostate cancer. Cancer Detect Prev 32: 23–32, 2008.
26.
Bower JE, Ganz PA, Tao ML, Hu W, Belin TR, Sepah S, Cole S, and Aziz N. Inflammatory biomarkers and fatigue during radiation therapy for breast and prostate cancer. Clin Cancer Res 15: 5534–5540, 2009.
27.
Bulua AC, Simon A, Maddipati R, Pelletier M, Park H, Kim KY, Sack MN, Kastner DL, and Siegel RM. Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). J Exp Med 208: 519–533, 2011.
28.
Byun HO, Jung HJ, Seo YH, Lee YK, Hwang SC, Hwang ES, and Yoon G. GSK3 inactivation is involved in mitochondrial complex IV defect in transforming growth factor (TGF) beta1-induced senescence. Exp Cell Res 318: 1808–1819, 2012.
29.
Carbo N, Busquets S, van Royen M, Alvarez B, Lopez-Soriano FJ, and Argiles JM. TNF-alpha is involved in activating DNA fragmentation in skeletal muscle. Br J Cancer 86: 1012–1016, 2002.
30.
Caruso DJ, Carmack AJ, Lokeshwar VB, Duncan RC, Soloway MS, and Lokeshwar BL. Osteopontin and interleukin-8 expression is independently associated with prostate cancer recurrence. Clin Cancer Res 14: 4111–4118, 2008.
31.
Charles KA, Kulbe H, Soper R, Escorcio-Correia M, Lawrence T, Schultheis A, Chakravarty P, Thompson RG, Kollias G, Smyth JF, Balkwill FR, and Hagemann T. The tumor-promoting actions of TNF-alpha involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J Clin Invest 119: 3011–3023, 2009.
32.
Chen Q, Chai YC, Mazumder S, Jiang C, Macklis RM, Chisolm GM, and Almasan A. The late increase in intracellular free radical oxygen species during apoptosis is associated with cytochrome c release, caspase activation, and mitochondrial dysfunction. Cell Death Differ 10: 323–334, 2003.
33.
Chendil D, Ranga RS, Meigooni D, Sathishkumar S, and Ahmed MM. Curcumin confers radiosensitizing effect in prostate cancer cell line PC-3. Oncogene 23: 1599–1607, 2004.
34.
Chiu HW, Chen YA, Ho SY, and Wang YJ. Arsenic trioxide enhances the radiation sensitivity of androgen-dependent and -independent human prostate cancer cells. PloS One 7: e31579, 2012.
35.
Chou CH, Chen PJ, Lee PH, Cheng AL, Hsu HC, and Cheng JC. Radiation-induced hepatitis B virus reactivation in liver mediated by the bystander effect from irradiated endothelial cells. Clin Cancer Res 13: 851–857, 2007.
36.
Chun JY, Nadiminty N, Dutt S, Lou W, Yang JC, Kung HJ, Evans CP, and Gao AC. Interleukin-6 regulates androgen synthesis in prostate cancer cells. Clin Cancer Res 15: 4815–4822, 2009.
37.
Chung LW, Baseman A, Assikis V, and Zhau HE. Molecular insights into prostate cancer progression: the missing link of tumor microenvironment. J Urol 173: 10–20, 2005.
38.
Condeelis J and Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 124: 263–266, 2006.
39.
Coussens LM and Werb Z. Inflammation and cancer. Nature 420: 860–867, 2002.
40.
Culig Z and Puhr M. Interleukin-6: a multifunctional targetable cytokine in human prostate cancer. Mol Cell Endocrinol 360: 52–58, 2011.
41.
Culig Z, Steiner H, Bartsch G, and Hobisch A. Interleukin-6 regulation of prostate cancer cell growth. J Cell Biochem 95: 497–505, 2005.
42.
David SS, O'Shea VL, and Kundu S. Base-excision repair of oxidative DNA damage. Nature 447: 941–950, 2007.
43.
Deng X, Elzey BD, Poulson JM, Morrison WB, Ko SC, Hahn NM, Ratliff TL, and Hu CD. Ionizing radiation induces neuroendocrine differentiation of prostate cancer cells in vitro, in vivo and in prostate cancer patients. Am J Cancer Res 1: 834–844, 2011.
44.
Deng X, Liu H, Huang J, Cheng L, Keller ET, Parsons SJ, and Hu CD. Ionizing radiation induces prostate cancer neuroendocrine differentiation through interplay of CREB and ATF2: implications for disease progression. Cancer Res 68: 9663–9670, 2008.
45.
Dent P, Yacoub A, Fisher PB, Hagan MP, and Grant S. MAPK pathways in radiation responses. Oncogene 22: 5885–5896, 2003.
46.
Dhar SK, Tangpong J, Chaiswing L, Oberley TD, and St Clair DK. Manganese superoxide dismutase is a p53-regulated gene that switches cancers between early and advanced stages. Cancer Res 71: 6684–6695, 2011.
47.
Domingo-Domenech J, Mellado B, Ferrer B, Truan D, Codony-Servat J, Sauleda S, Alcover J, Campo E, Gascon P, Rovira A, Ross JS, Fernandez PL, and Albanell J. Activation of nuclear factor-kappaB in human prostate carcinogenesis and association to biochemical relapse. Br J Cancer 93: 1285–1294, 2005.
48.
Drabsch Y and ten Dijke P. TGF-beta signalling and its role in cancer progression and metastasis. Cancer Metastasis Rev 31: 553–568, 2012.
49.
Epperly MW, Osipov AN, Martin I, Kawai KK, Borisenko GG, Tyurina YY, Jefferson M, Bernarding M, Greenberger JS, and Kagan VE. Ascorbate as a “redox sensor” and protector against irradiation-induced oxidative stress in 32D CL 3 hematopoietic cells and subclones overexpressing human manganese superoxide dismutase. Int J Radiat Oncol Biol Phys 58: 851–861, 2004.
50.
Fiaschi T and Chiarugi P. Oxidative stress, tumor microenvironment, and metabolic reprogramming: a diabolic liaison. Int J Cell Biol 2012: 762825, 2012.
51.
Flechsig P, Dadrich M, Bickelhaupt S, Jenne J, Hauser K, Timke C, Peschke P, Hahn EW, Grone HJ, Yingling J, Lahn M, Wirkner U, and Huber PE. LY2109761 attenuates radiation-induced pulmonary murine fibrosis via reversal of TGF-beta and BMP-associated proinflammatory and proangiogenic signals. Clin Cancer Res 18: 3616–3627, 2012.
52.
Flores MB, Rocha GZ, Damas-Souza DM, Osorio-Costa F, Dias MM, Ropelle ER, Camargo JA, de Carvalho RB, Carvalho HF, Saad MJ, and Carvalheira JB. Obesity-induced increase in tumor necrosis factor-alpha leads to development of colon cancer in mice. Gastroenterology 143: 741–753 e1–e4, 2012.
53.
Gahan JC, Gosalbez M, Yates T, Young EE, Escudero DO, Chi A, Garcia-Roig M, Satyanarayana R, Soloway MS, Bird VG, and Lokeshwar VB. Chemokine and chemokine receptor expression in kidney tumors: molecular profiling of histological subtypes and association with metastasis. J Urol 187: 827–833, 2012.
54.
Gallet P, Phulpin B, Merlin JL, Leroux A, Bravetti P, Mecellem H, Tran N, and Dolivet G. Long-term alterations of cytokines and growth factors expression in irradiated tissues and relation with histological severity scoring. PloS One 6: e29399, 2011.
55.
Gannon PO, Godin-Ethier J, Hassler M, Delvoye N, Aversa M, Poisson AO, Peant B, Alam Fahmy M, Saad F, Lapointe R, and Mes-Masson AM. Androgen-regulated expression of arginase 1, arginase 2 and interleukin-8 in human prostate cancer. PloS One 5: e12107, 2010.
56.
Gladson CL and Welch DR. New insights into the role of CXCR4 in prostate cancer metastasis. Cancer Biol Ther 7: 1849–1851, 2008.
57.
Gonzalez-Ramos M, Mora I, de Frutos S, Garesse R, Rodriguez-Puyol M, Olmos G, and Rodriguez-Puyol D. Intracellular redox equilibrium is essential for the constitutive expression of AP-1 dependent genes in resting cells: studies on TGF-beta1 regulation. Int J Biochem Cell Biol 44: 963–971, 2012.
58.
Gough DJ, Corlett A, Schlessinger K, Wegrzyn J, Larner AC, and Levy DE. Mitochondrial STAT3 supports Ras-dependent oncogenic transformation. Science 324: 1713–1716, 2009.
59.
Govinden R and Bhoola KD. Genealogy, expression, and cellular function of transforming growth factor-beta. Pharmacol Ther 98: 257–265, 2003.
60.
Grady WM, Myeroff LL, Swinler SE, Rajput A, Thiagalingam S, Lutterbaugh JD, Neumann A, Brattain MG, Chang J, Kim SJ, Kinzler KW, Vogelstein B, Willson JK, and Markowitz S. Mutational inactivation of transforming growth factor beta receptor type II in microsatellite stable colon cancers. Cancer Res 59: 320–324, 1999.
61.
Guo G, Yan-Sanders Y, Lyn-Cook BD, Wang T, Tamae D, Ogi J, Khaletskiy A, Li Z, Weydert C, Longmate JA, Huang TT, Spitz DR, Oberley LW, and Li JJ. Manganese superoxide dismutase-mediated gene expression in radiation-induced adaptive responses. Mol Cell Biol 23: 2362–2378, 2003.
62.
Guo Y, Xu F, Lu T, Duan Z, and Zhang Z. Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat Rev 38: 904–910, 2012.
63.
Gupta SC, Hevia D, Patchva S, Park B, Koh W, and Aggarwal BB. Upsides and downsides of reactive oxygen species for cancer: the roles of reactive oxygen species in tumorigenesis, prevention, and therapy. Antioxid Redox Signal 16: 1295–1322, 2012.
64.
Gupta-Elera G, Garrett AR, Robison RA, and O'Neill KL. The role of oxidative stress in prostate cancer. Eur J Cancer Prev 21: 155–162, 2012.
65.
Hagemann T, Lawrence T, McNeish I, Charles KA, Kulbe H, Thompson RG, Robinson SC, and Balkwill FR. “Re-educating” tumor-associated macrophages by targeting NF-kappaB. J Exp Med 205: 1261–1268, 2008.
66.
Hall EJ. Radiobiology for the Radiologist. Philadelphia: JB Lippincott Company, 1994.
67.
Hanahan D and Weinberg RA. Hallmarks of cancer: the next generation. Cell 144: 646–674, 2011.
68.
Hardee ME, Marciscano AE, Medina-Ramirez CM, Zagzag D, Narayana A, Lonning SM, and Barcellos-Hoff MH. Resistance of glioblastoma-initiating cells to radiation mediated by the tumor microenvironment can be abolished by inhibiting transforming growth factor-beta. Cancer Res 72: 4119–4129, 2012.
69.
Hardmeier R, Hoeger H, Fang-Kircher S, Khoschsorur A, and Lubec G. Transcription and activity of antioxidant enzymes after ionizing irradiation in radiation-resistant and radiation-sensitive mice. Proc Natl Acad Sci U S A 94: 7572–7576, 1997.
70.
Hobisch A, Ramoner R, Fuchs D, Godoy-Tundidor S, Bartsch G, Klocker H, and Culig Z. Prostate cancer cells (LNCaP) generated after long-term interleukin 6 (IL-6) treatment express IL-6 and acquire an IL-6 partially resistant phenotype. Clin Cancer Res 7: 2941–2948, 2001.
71.
Holley AK, Xu Y, St Clair DK, and St Clair WH. RelB regulates manganese superoxide dismutase gene and resistance to ionizing radiation of prostate cancer cells. Ann N Y Acad Sci 1201: 129–136, 2010.
72.
Holmes WE, Lee J, Kuang WJ, Rice GC, and Wood WI. Structure and functional expression of a human interleukin-8 receptor. Science 253: 1278–1280, 1991.
73.
Hosoki A, Yonekura S, Zhao QL, Wei ZL, Takasaki I, Tabuchi Y, Wang LL, Hasuike S, Nomura T, Tachibana A, Hashiguchi K, Yonei S, Kondo T, and Zhang-Akiyama QM. Mitochondria-targeted superoxide dismutase (SOD2) regulates radiation resistance and radiation stress response in HeLa cells. J Radiat Res 53: 58–71, 2012.
74.
Huang LE, Arany Z, Livingston DM, and Bunn HF. Activation of hypoxia-inducible transcription factor depends primarily upon redox-sensitive stabilization of its alpha subunit. J Biol Chem 271: 32253–32259, 1996.
75.
Hunter NR, Valdecanas D, Liao Z, Milas L, Thames HD, and Mason KA. Mitigation and treatment of radiation-induced thoracic injury with a cyclooxygenase-2 inhibitor, celecoxib. Int J Radiat Oncol Biol Phys 85: 472–476, 2012.
76.
Husbeck B, Peehl DM, and Knox SJ. Redox modulation of human prostate carcinoma cells by selenite increases radiation-induced cell killing. Free Radic Biol Med 38: 50–57, 2005.
77.
Ivanovic V, Melman A, Davis-Joseph B, Valcic M, and Geliebter J. Elevated plasma levels of TGF-beta 1 in patients with invasive prostate cancer. Nat Med 1: 282–284, 1995.
78.
Iyer R, Lehnert BE, and Svensson R. Factors underlying the cell growth-related bystander responses to alpha particles. Cancer Res 60: 1290–1298, 2000.
79.
Josson S, Xu Y, Fang F, Dhar SK, St Clair DK, and St Clair WH. RelB regulates manganese superoxide dismutase gene and resistance to ionizing radiation of prostate cancer cells. Oncogene 25: 1554–1559, 2006.
80.
Kakkar P and Singh BK. Mitochondria: a hub of redox activities and cellular distress control. Mol Cell Biochem 305: 235–253, 2007.
81.
Kamata H, Honda S, Maeda S, Chang L, Hirata H, and Karin M. Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell 120: 649–661, 2005.
82.
Kang HY, Lin HK, Hu YC, Yeh S, Huang KE, and Chang C. From transforming growth factor-beta signaling to androgen action: identification of Smad3 as an androgen receptor coregulator in prostate cancer cells. Proc Natl Acad Sci U S A 98: 3018–3023, 2001.
83.
Kang MA, So EY, Simons AL, Spitz DR, and Ouchi T. DNA damage induces reactive oxygen species generation through the H2AX-Nox1/Rac1 pathway. Cell Death Dis 3: e249, 2012.
84.
Kang SK, Rabbani ZN, Folz RJ, Golson ML, Huang H, Yu D, Samulski TS, Dewhirst MW, Anscher MS, and Vujaskovic Z. Overexpression of extracellular superoxide dismutase protects mice from radiation-induced lung injury. Int J Radiat Oncol Biol Phys 57: 1056–1066, 2003.
85.
Kawada M, Inoue H, Arakawa M, and Ikeda D. Transforming growth factor-beta1 modulates tumor-stromal cell interactions of prostate cancer through insulin-like growth factor-I. Anticancer Res 28: 721–730, 2008.
86.
Khandrika L, Kumar B, Koul S, Maroni P, and Koul HK. Oxidative stress in prostate cancer. Cancer Lett 282: 125–136, 2009.
87.
Kil WJ, Tofilon PJ, and Camphausen K. Post-radiation increase in VEGF enhances glioma cell motility in vitro. Radiat Oncol 7: 25, 2012.
88.
Kim BY, Kim KA, Kwon O, Kim SO, Kim MS, Kim BS, Oh WK, Kim GD, Jung M, and Ahn JS. NF-kappaB inhibition radiosensitizes Ki-Ras-transformed cells to ionizing radiation. Carcinogenesis 26: 1395–1403, 2005.
89.
Kim MH, Minton AZ, and Agrawal V. C/EBPbeta regulates metastatic gene expression and confers TNF-alpha resistance to prostate cancer cells. Prostate 69: 1435–1447, 2009.
90.
Kim YS, Morgan MJ, Choksi S, and Liu ZG. TNF-induced activation of the Nox1 NADPH oxidase and its role in the induction of necrotic cell death. Mol Cell 26: 675–687, 2007.
91.
Kimura K, Bowen C, Spiegel S, and Gelmann EP. Tumor necrosis factor-alpha sensitizes prostate cancer cells to gamma-irradiation-induced apoptosis. Cancer Res 59: 1606–1614, 1999.
92.
Kishimoto T, Akira S, Narazaki M, and Taga T. Interleukin-6 family of cytokines and gp130. Blood 86: 1243–1254, 1995.
93.
Kitagawa Y, Dai J, Zhang J, Keller JM, Nor J, Yao Z, and Keller ET. Vascular endothelial growth factor contributes to prostate cancer-mediated osteoblastic activity. Cancer Res 65: 10921–10929, 2005.
94.
Ko S, Shi L, Kim S, Song CS, and Chatterjee B. Interplay of nuclear factor-kappaB and B-myb in the negative regulation of androgen receptor expression by tumor necrosis factor alpha. Mol Endocrinol 22: 273–286, 2008.
95.
Kong Z, Xie D, Boike T, Raghavan P, Burma S, Chen DJ, Habib AA, Chakraborty A, Hsieh JT, and Saha D. Downregulation of human DAB2IP gene expression in prostate cancer cells results in resistance to ionizing radiation. Cancer Res 70: 2829–2839, 2010.
96.
Koul HK, Kumar B, Koul S, Deb AA, Hwa JS, Maroni P, van Bokhoven A, Lucia MS, Kim FJ, and Meacham RB. The role of inflammation and infection in prostate cancer: importance in prevention, diagnosis and treatment. Drugs Today 46: 929–943, 2010.
97.
Kryston TB, Georgiev AB, Pissis P, and Georgakilas AG. Role of oxidative stress and DNA damage in human carcinogenesis. Mutat Res 711: 193–201, 2011.
98.
Kumar B, Koul S, Khandrika L, Meacham RB, and Koul HK. Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype. Cancer Res 68: 1777–1785, 2008.
99.
Larsen L and Ropke C. Suppressors of cytokine signalling: SOCS. APMIS 110: 833–844, 2002.
100.
Lejeune FJ. Clinical use of TNF revisited: improving penetration of anti-cancer agents by increasing vascular permeability. J Clin Invest 110: 433–435, 2002.
101.
Lenaz G and Genova ML. Structure and organization of mitochondrial respiratory complexes: a new understanding of an old subject. Antioxid Redox Signal 12: 961–1008, 2010.
102.
Levine AJ, Momand J, and Finlay CA. The p53 tumour suppressor gene. Nature 351: 453–456, 1991.
103.
Li F and Sethi G. Targeting transcription factor NF-kappaB to overcome chemoresistance and radioresistance in cancer therapy. Biochim Biophys Acta 1805: 167–180, 2010.
104.
Li JM, Fan LM, Christie MR, and Shah AM. Acute tumor necrosis factor alpha signaling via NADPH oxidase in microvascular endothelial cells: role of p47phox phosphorylation and binding to TRAF4. Mol Cell Biol 25: 2320–2330, 2005.
105.
Linard C, Ropenga A, Vozenin-Brotons MC, Chapel A, and Mathe D. Abdominal irradiation increases inflammatory cytokine expression and activates NF-kappaB in rat ileal muscularis layer. Am J Physiol Gastrointest Liver Physiol 285: G556–G565, 2003.
106.
Loetscher H, Pan YC, Lahm HW, Gentz R, Brockhaus M, Tabuchi H, and Lesslauer W. Molecular cloning and expression of the human 55 kd tumor necrosis factor receptor. Cell 61: 351–359, 1990.
107.
Lu L, Tang D, Wang L, Huang LQ, Jiang GS, Xiao XY, and Zeng FQ. Gambogic acid inhibits TNF-alpha-induced invasion of human prostate cancer PC3 cells in vitro through PI3K/Akt and NF-kappaB signaling pathways. Acta Pharmacol Sin 33: 531–541, 2012.
108.
Martin M, Lefaix J, and Delanian S. TGF-beta1 and radiation fibrosis: a master switch and a specific therapeutic target? Int J Radiat Oncol Biol Phys 47: 277–290, 2000.
109.
Martinez-Outschoorn UE, Balliet RM, Rivadeneira DB, Chiavarina B, Pavlides S, Wang C, Whitaker-Menezes D, Daumer KM, Lin Z, Witkiewicz AK, Flomenberg N, Howell A, Pestell RG, Knudsen ES, Sotgia F, and Lisanti MP. Oxidative stress in cancer associated fibroblasts drives tumor-stroma co-evolution: a new paradigm for understanding tumor metabolism, the field effect and genomic instability in cancer cells. Cell Cycle 9: 3256–3276, 2010.
110.
Masuda M, Wakasaki T, Suzui M, Toh S, Joe AK, and Weinstein IB. Stat3 orchestrates tumor development and progression: the Achilles' heel of head and neck cancers? Curr Cancer Drug Targets 10: 117–126, 2010.
111.
McBride WH, Chiang CS, Olson JL, Wang CC, Hong JH, Pajonk F, Dougherty GJ, Iwamoto KS, Pervan M, and Liao YP. A sense of danger from radiation. Radiat Res 162: 1–19, 2004.
112.
Mehrotra S, Pecaut MJ, Freeman TL, Crapo JD, Rizvi A, Luo-Owen X, Slater JM, and Gridley DS. Analysis of a metalloporphyrin antioxidant mimetic (MnTE-2-PyP) as a radiomitigator: prostate tumor and immune status. Technol Cancer Res Treat 11: 447–457, 2012.
113.
Mettler F and Upton A. Medical Effects of Ionizing Radiation. Philadelphia, PA: Saunders Elsvier, 2008.
114.
Meulmeester E and Ten Dijke P. The dynamic roles of TGF-beta in cancer. J Pathol 223: 205–218, 2011.
115.
Miao L and St Clair DK. Regulation of superoxide dismutase genes: implications in disease. Free Radic Biol Med 47: 344–356, 2009.
116.
Michaeloudes C, Sukkar MB, Khorasani NM, Bhavsar PK, and Chung KF. TGF-beta regulates Nox4, MnSOD and catalase expression, and IL-6 release in airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 300: L295–L304, 2011.
117.
Michalaki V, Syrigos K, Charles P, and Waxman J. Serum levels of IL-6 and TNF-alpha correlate with clinicopathological features and patient survival in patients with prostate cancer. Br J Cancer 90: 2312–2316, 2004.
118.
Mizokami A, Gotoh A, Yamada H, Keller ET, and Matsumoto T. Tumor necrosis factor-alpha represses androgen sensitivity in the LNCaP prostate cancer cell line. J Urol 164: 800–805, 2000.
119.
Mocellin S, Rossi CR, Pilati P, and Nitti D. Tumor necrosis factor, cancer and anticancer therapy. Cytokine Growth Factor Rev 16: 35–53, 2005.
120.
Morales A, Miranda M, Sanchez-Reyes A, Biete A, and Fernandez-Checa JC. Oxidative damage of mitochondrial and nuclear DNA induced by ionizing radiation in human hepatoblastoma cells. Int J Radiat Oncol Biol Phys 42: 191–203, 1998.
121.
Morton DM and Barrack ER. Modulation of transforming growth factor beta 1 effects on prostate cancer cell proliferation by growth factors and extracellular matrix. Cancer Res 55: 2596–2602, 1995.
122.
Mukherjee A and Martin SG. The thioredoxin system: a key target in tumour and endothelial cells. Br J Radiol 81 Spec No 1: S57–S68, 2008.
123.
Multhoff G and Radons J. Radiation, inflammation, and immune responses in cancer. Front Oncol 2: 58, 2012.
124.
Murley JS, Kataoka Y, Baker KL, Diamond AM, Morgan WF, and Grdina DJ. Manganese superoxide dismutase (SOD2)-mediated delayed radioprotection induced by the free thiol form of amifostine and tumor necrosis factor alpha. Radiat Res 167: 465–474, 2007.
125.
Naka K, Muraguchi T, Hoshii T, and Hirao A. Regulation of reactive oxygen species and genomic stability in hematopoietic stem cells. Antioxid Redox Signal 10: 1883–1894, 2008.
126.
Nakashima J, Tachibana M, Horiguchi Y, Oya M, Ohigashi T, Asakura H, and Murai M. Serum interleukin 6 as a prognostic factor in patients with prostate cancer. Clin Cancer Res 6: 2702–2706, 2000.
127.
Nakashima J, Tachibana M, Ueno M, Miyajima A, Baba S, and Murai M. Association between tumor necrosis factor in serum and cachexia in patients with prostate cancer. Clin Cancer Res 4: 1743–1748, 1998.
128.
Narayanan PK, LaRue KE, Goodwin EH, and Lehnert BE. Alpha particles induce the production of interleukin-8 by human cells. Radiat Res 152: 57–63, 1999.
129.
Nelson EC, Cambio AJ, Yang JC, Ok JH, Lara PN Jr., and Evans CP. Clinical implications of neuroendocrine differentiation in prostate cancer. Prostate Cancer Prostatic Dis 10: 6–14, 2007.
130.
Nguyen DH, Oketch-Rabah HA, Illa-Bochaca I, Geyer FC, Reis-Filho JS, Mao JH, Ravani SA, Zavadil J, Borowsky AD, Jerry DJ, Dunphy KA, Seo JH, Haslam S, Medina D, and Barcellos-Hoff MH. Radiation acts on the microenvironment to affect breast carcinogenesis by distinct mechanisms that decrease cancer latency and affect tumor type. Cancer Cell 19: 640–651, 2011.
131.
Oakley FD, Abbott D, Li Q, and Engelhardt JF. Signaling components of redox active endosomes: the redoxosomes. Antioxid Redox Signal 11: 1313–1333, 2009.
132.
Ogawa Y, Kobayashi T, Nishioka A, Kariya S, Hamasato S, Seguchi H, and Yoshida S. Radiation-induced reactive oxygen species formation prior to oxidative DNA damage in human peripheral T cells. Int J Mol Med 11: 149–152, 2003.
133.
Okamoto M, Lee C, and Oyasu R. Interleukin-6 as a paracrine and autocrine growth factor in human prostatic carcinoma cells in vitro. Cancer Res 57: 141–146, 1997.
134.
Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD, and Cunha GR. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res 59: 5002–5011, 1999.
135.
Oshikawa J, Urao N, Kim HW, Kaplan N, Razvi M, McKinney R, Poole LB, Fukai T, and Ushio-Fukai M. Extracellular SOD-derived H2O2 promotes VEGF signaling in caveolae/lipid rafts and post-ischemic angiogenesis in mice. PloS One 5: e10189, 2010.
136.
Park JI, Lee MG, Cho K, Park BJ, Chae KS, Byun DS, Ryu BK, Park YK, and Chi SG. Transforming growth factor-beta1 activates interleukin-6 expression in prostate cancer cells through the synergistic collaboration of the Smad2, p38-NF-kappaB, JNK, and Ras signaling pathways. Oncogene 22: 4314–4332, 2003.
137.
Park JS, Qiao L, Su ZZ, Hinman D, Willoughby K, McKinstry R, Yacoub A, Duigou GJ, Young CS, Grant S, Hagan MP, Ellis E, Fisher PB, and Dent P. Ionizing radiation modulates vascular endothelial growth factor (VEGF) expression through multiple mitogen activated protein kinase dependent pathways. Oncogene 20: 3266–3280, 2001.
138.
Pavlides S, Vera I, Gandara R, Sneddon S, Pestell RG, Mercier I, Martinez-Outschoorn UE, Whitaker-Menezes D, Howell A, Sotgia F, and Lisanti MP. Warburg meets autophagy: cancer-associated fibroblasts accelerate tumor growth and metastasis via oxidative stress, mitophagy, and aerobic glycolysis. Antioxid Redox Signal 16: 1264–1284, 2012.
139.
Perry KT, Anthony CT, Case T, and Steiner MS. Transforming growth factor beta as a clinical biomarker for prostate cancer. Urology 49: 151–155, 1997.
140.
Pfitzenmaier J, Vessella R, Higano CS, Noteboom JL, Wallace D Jr., and Corey E. Elevation of cytokine levels in cachectic patients with prostate carcinoma. Cancer 97: 1211–1216, 2003.
141.
Ping X, Junqing J, Junfeng J, and Enjin J. Radioprotective effects of troxerutin against gamma irradiation in mice liver. Int J Radiat Biol 88: 607–612, 2012.
142.
Prise KM and O'Sullivan JM. Radiation-induced bystander signalling in cancer therapy. Nat Rev Cancer 9: 351–360, 2009.
143.
Pu H, Collazo J, Jones E, Gayheart D, Sakamoto S, Vogt A, Mitchell B, and Kyprianou N. Dysfunctional transforming growth factor-beta receptor II accelerates prostate tumorigenesis in the TRAMP mouse model. Cancer Res 69: 7366–7374, 2009.
144.
Qiu Y, Robinson D, Pretlow TG, and Kung HJ. Etk/Bmx, a tyrosine kinase with a pleckstrin-homology domain, is an effector of phosphatidylinositol 3'-kinase and is involved in interleukin 6-induced neuroendocrine differentiation of prostate cancer cells. Proc Natl Acad Sci U S A 95: 3644–3649, 1998.
145.
Quiros-Gonzalez I, Sainz RM, Hevia D, and Mayo JC. MnSOD drives neuroendocrine differentiation, androgen independence, and cell survival in prostate cancer cells. Free Radic Biol Med 50: 525–536, 2011.
146.
Rabbani ZN, Anscher MS, Zhang X, Chen L, Samulski TV, Li CY, and Vujaskovic Z. Soluble TGFbeta type II receptor gene therapy ameliorates acute radiation-induced pulmonary injury in rats. Int J Radiat Oncol Biol Phys 57: 563–572, 2003.
147.
Reuter S, Gupta SC, Chaturvedi MM, and Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49: 1603–1616, 2010.
148.
Riley PA. Free radicals in biology: oxidative stress and the effects of ionizing radiation. Int J Radiat Biol 65: 27–33, 1994.
149.
Rivas MA, Carnevale RP, Proietti CJ, Rosemblit C, Beguelin W, Salatino M, Charreau EH, Frahm I, Sapia S, Brouckaert P, Elizalde PV, and Schillaci R. TNF alpha acting on TNFR1 promotes breast cancer growth via p42/P44 MAPK, JNK, Akt and NF-kappa B-dependent pathways. Exp Cell Res 314: 509–529, 2008.
150.
Rose-John S. Coordination of interleukin-6 biology by membrane bound and soluble receptors. Adv Exp Med Biol 495: 145–151, 2001.
151.
Rzymski P, Opala T, Wilczak M, Wozniak J, and Sajdak S. Serum tumor necrosis factor alpha receptors p55/p75 ratio and ovarian cancer detection. Int J Gynaecol Obstet 88: 292–298, 2005.
152.
Salama S, Diaz-Arrastia C, Patel D, Botting S, and Hatch S. 2-Methoxyestradiol, an endogenous estrogen metabolite, sensitizes radioresistant MCF-7/FIR breast cancer cells through multiple mechanisms. Int J Radiat Oncol Biol Phys 80: 231–239, 2011.
153.
Scheller J, Ohnesorge N, and Rose-John S. Interleukin-6 trans-signalling in chronic inflammation and cancer. Scand J Immunol 63: 321–329, 2006.
154.
Scherz-Shouval R and Elazar Z. ROS, mitochondria and the regulation of autophagy. Trends Cell Biol 17: 422–427, 2007.
155.
Schirmer MA, Brockmoller J, Rave-Frank M, Virsik P, Wilken B, Kuhnle E, Campean R, Hoffmann AO, Muller K, Goetze RG, Neumann M, Janke JH, Nasser F, Wolff HA, Ghadimi BM, Schmidberger H, Hess CF, Christiansen H, and Hille A. A putatively functional haplotype in the gene encoding transforming growth factor beta-1 as a potential biomarker for radiosensitivity. Int J Radiat Oncol Biol Phys 79: 866–874, 2011.
156.
Schreiber S, Nikolaus S, and Hampe J. Activation of nuclear factor kappa B inflammatory bowel disease. Gut 42: 477–484, 1998.
157.
Schumacker PT. Reactive oxygen species in cancer cells: live by the sword, die by the sword. Cancer Cell 10: 175–176, 2006.
158.
Seaton A, Maxwell PJ, Hill A, Gallagher R, Pettigrew J, Wilson RH, and Waugh DJ. Inhibition of constitutive and cxc-chemokine-induced NF-kappaB activity potentiates ansamycin-based HSP90-inhibitor cytotoxicity in castrate-resistant prostate cancer cells. Br J Cancer 101: 1620–1629, 2009.
159.
Seaton A, Scullin P, Maxwell PJ, Wilson C, Pettigrew J, Gallagher R, O'Sullivan JM, Johnston PG, and Waugh DJ. Interleukin-8 signaling promotes androgen-independent proliferation of prostate cancer cells via induction of androgen receptor expression and activation. Carcinogenesis 29: 1148–1156, 2008.
160.
Shamaladevi N, Lyn DA, Escudero DO, and Lokeshwar BL. CXC receptor-1 silencing inhibits androgen-independent prostate cancer. Cancer Res 69: 8265–8274, 2009.
161.
Shan W, Zhong W, Zhao R, and Oberley TD. Thioredoxin 1 as a subcellular biomarker of redox imbalance in human prostate cancer progression. Free Radic Biol Med 49: 2078–2087, 2010.
162.
Shariat SF, Shalev M, Menesses-Diaz A, Kim IY, Kattan MW, Wheeler TM, and Slawin KM. Preoperative plasma levels of transforming growth factor beta(1) (TGF-beta(1)) strongly predict progression in patients undergoing radical prostatectomy. J Clin Oncol 19: 2856–2864, 2001.
163.
Sharifi N, Hurt EM, Thomas SB, and Farrar WL. Effects of manganese superoxide dismutase silencing on androgen receptor function and gene regulation: implications for castration-resistant prostate cancer. Clin Cancer Res 14: 6073–6080, 2008.
164.
Sheets NC, Goldin GH, Meyer AM, Wu Y, Chang Y, Sturmer T, Holmes JA, Reeve BB, Godley PA, Carpenter WR, and Chen RC. Intensity-modulated radiation therapy, proton therapy, or conformal radiation therapy and morbidity and disease control in localized prostate cancer. JAMA 307: 1611–1620, 2012.
165.
Shiota M, Yokomizo A, and Naito S. Oxidative stress and androgen receptor signaling in the development and progression of castration-resistant prostate cancer. Free Radic Biol Med 51: 1320–1328, 2011.
166.
Shiota M, Yokomizo A, Tada Y, Inokuchi J, Kashiwagi E, Masubuchi D, Eto M, Uchiumi T, and Naito S. Castration resistance of prostate cancer cells caused by castration-induced oxidative stress through Twist1 and androgen receptor overexpression. Oncogene 29: 237–250, 2010.
167.
Siegel R, Naishadham D, and Jemal A. Cancer statistics, 2012. CA Cancer J Clin 62: 10–29, 2012.
168.
Singh RK and Lokeshwar BL. Depletion of intrinsic expression of interleukin-8 in prostate cancer cells causes cell cycle arrest, spontaneous apoptosis and increases the efficacy of chemotherapeutic drugs. Mol Cancer 8: 57, 2009.
169.
Singh RK and Lokeshwar BL. The IL-8-regulated chemokine receptor CXCR7 stimulates EGFR signaling to promote prostate cancer growth. Cancer Res 71: 3268–3277, 2011.
170.
Singh RK, Sudhakar A, and Lokeshwar BL. Role of chemokines and chemokine receptors in prostate cancer development and progression. J Cancer Sci Ther 2: 89–94, 2010.
171.
Skvortsova I, Skvortsov S, Stasyk T, Raju U, Popper BA, Schiestl B, von Guggenberg E, Neher A, Bonn GK, Huber LA, and Lukas P. Intracellular signaling pathways regulating radioresistance of human prostate carcinoma cells. Proteomics 8: 4521–4533, 2008.
172.
Slupphaug G, Kavli B, and Krokan HE. The interacting pathways for prevention and repair of oxidative DNA damage. Mutat Res 531: 231–251, 2003.
173.
Smith CA, Davis T, Anderson D, Solam L, Beckmann MP, Jerzy R, Dower SK, Cosman D, and Goodwin RG. A receptor for tumor necrosis factor defines an unusual family of cellular and viral proteins. Science 248: 1019–1023, 1990.
174.
Sorbara MT and Girardin SE. Mitochondrial ROS fuel the inflammasome. Cell Res 21: 558–560, 2011.
175.
Spiotto MT and Chung TD. STAT3 mediates IL-6-induced neuroendocrine differentiation in prostate cancer cells. Prostate 42: 186–195, 2000.
176.
Steuber T, O'Brien MF, and Lilja H. Serum markers for prostate cancer: a rational approach to the literature. Eur Urol 54: 31–40, 2008.
177.
Sun Y and Oberley LW. Redox regulation of transcriptional activators. Free Radic Biol Med 21: 335–348, 1996.
178.
Sun Y, St Clair DK, Xu Y, Crooks PA, and St Clair WH. A NADPH oxidase-dependent redox signaling pathway mediates the selective radiosensitization effect of parthenolide in prostate cancer cells. Cancer Res 70: 2880–2890, 2010.
179.
Tateishi Y, Sasabe E, Ueta E, and Yamamoto T. Ionizing irradiation induces apoptotic damage of salivary gland acinar cells via NADPH oxidase 1-dependent superoxide generation. Biochem Biophys Res Commun 366: 301–307, 2008.
180.
Tell G, Quadrifoglio F, Tiribelli C, and Kelley MR. The many functions of APE1/Ref-1: not only a DNA repair enzyme. Antioxid Redox Signal 11: 601–620, 2009.
181.
Thompson I, Thrasher JB, Aus G, Burnett AL, Canby-Hagino ED, Cookson MS, D'Amico AV, Dmochowski RR, Eton DT, Forman JD, Goldenberg SL, Hernandez J, Higano CS, Kraus SR, Moul JW, and Tangen CM. Guideline for the management of clinically localized prostate cancer: 2007 update. J Urol 177: 2106–2131, 2007.
182.
Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev 89: 381–410, 2009.
183.
Trachootham D, Alexandre J, and Huang P. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev Drug Discov 8: 579–591, 2009.
184.
Tsui KH, Lin YF, Chen YH, Chang PL, and Juang HH. Mechanisms by which interleukin-6 regulates prostate-specific antigen gene expression in prostate LNCaP carcinoma cells. J Androl 32: 383–393, 2011.
185.
Valerie NC, Casarez EV, Dasilva JO, Dunlap-Brown ME, Parsons SJ, Amorino GP, and Dziegielewski J. Inhibition of neurotensin receptor 1 selectively sensitizes prostate cancer to ionizing radiation. Cancer Res 71: 6817–6826, 2011.
186.
Vlahopoulos S, Boldogh I, Casola A, and Brasier AR. Nuclear factor-kappaB-dependent induction of interleukin-8 gene expression by tumor necrosis factor alpha: evidence for an antioxidant sensitive activating pathway distinct from nuclear translocation. Blood 94: 1878–1889, 1999.
187.
Wallace DC. Diseases of the mitochondrial DNA. Annu Rev Biochem 61: 1175–1212, 1992.
188.
Wang D, Lu S, and Dong Z. Regulation of TGF-beta1 gene transcription in human prostate cancer cells by nitric oxide. Prostate 67: 1825–1833, 2007.
189.
Waugh DJ and Wilson C. The interleukin-8 pathway in cancer. Clin Cancer Res 14: 6735–6741, 2008.
190.
Whitaker-Menezes D, Martinez-Outschoorn UE, Lin Z, Ertel A, Flomenberg N, Witkiewicz AK, Birbe RC, Howell A, Pavlides S, Gandara R, Pestell RG, Sotgia F, Philp NJ, and Lisanti MP. Evidence for a stromal-epithelial “lactate shuttle” in human tumors: MCT4 is a marker of oxidative stress in cancer-associated fibroblasts. Cell Cycle 10: 1772–1783, 2011.
191.
Wiegman EM, Blaese MA, Loeffler H, Coppes RP, and Rodemann HP. TGFbeta-1 dependent fast stimulation of ATM and p53 phosphorylation following exposure to ionizing radiation does not involve TGFbeta-receptor I signalling. Radiother Oncol 83: 289–295, 2007.
192.
Wikstrom P, Stattin P, Franck-Lissbrant I, Damber JE, and Bergh A. Transforming growth factor beta1 is associated with angiogenesis, metastasis, and poor clinical outcome in prostate cancer. Prostate 37: 19–29, 1998.
193.
Wilson C, Purcell C, Seaton A, Oladipo O, Maxwell PJ, O'Sullivan JM, Wilson RH, Johnston PG, and Waugh DJ. Chemotherapy-induced CXC-chemokine/CXC-chemokine receptor signaling in metastatic prostate cancer cells confers resistance to oxaliplatin through potentiation of nuclear factor-kappaB transcription and evasion of apoptosis. J Pharmacol Exp Ther 327: 746–759, 2008.
194.
Wilson C, Wilson T, Johnston PG, Longley DB, and Waugh DJ. Interleukin-8 signaling attenuates TRAIL- and chemotherapy-induced apoptosis through transcriptional regulation of c-FLIP in prostate cancer cells. Mol Cancer Ther 7: 2649–2661, 2008.
195.
Wolff JM, Fandel TH, Borchers H, and Jakse G. Serum concentrations of transforming growth factor-beta 1 in patients with benign and malignant prostatic diseases. Anticancer Res 19: 2657–2659, 1999.
196.
Xu Y, Fang F, St Clair DK, and St Clair WH. Inverse relationship between PSA and IL-8 in prostate cancer: an insight into a NF-kappaB-mediated mechanism. PloS One 7: e32905, 2012.
197.
Xu Y, Josson S, Fang F, Oberley TD, St Clair DK, Wan XS, Sun Y, Bakthavatchalu V, Muthuswamy A, and St Clair WH. RelB enhances prostate cancer growth: implications for the role of the nuclear factor-kappaB alternative pathway in tumorigenicity. Cancer Res 69: 3267–3271, 2009.
198.
Yoshida T, Goto S, Kawakatsu M, Urata Y, and Li TS. Mitochondrial dysfunction, a probable cause of persistent oxidative stress after exposure to ionizing radiation. Free Radic Res 46: 147–153, 2012.
199.
Yu H and Jove R. The STATs of cancer—new molecular targets come of age. Nat Rev Cancer 4: 97–105, 2004.
200.
Zhang S, Song C, Zhou J, Xie L, Meng X, Liu P, Cao J, Zhang X, Ding WQ, and Wu J. Amelioration of radiation-induced skin injury by adenovirus-mediated heme oxygenase-1 (HO-1) overexpression in rats. Radiat Oncol 7: 4, 2012.
201.
Zhou H, Ivanov VN, Gillespie J, Geard CR, Amundson SA, Brenner DJ, Yu Z, Lieberman HB, and Hei TK. Mechanism of radiation-induced bystander effect: role of the cyclooxygenase-2 signaling pathway. Proc Natl Acad Sci U S A 102: 14641–14646, 2005.
202.
Zhou J and Du Y. Acquisition of resistance of pancreatic cancer cells to 2-methoxyestradiol is associated with the upregulation of manganese superoxide dismutase. Mol Cancer Res 10: 768–777, 2012.
203.
Zhu ML and Kyprianou N. Androgen receptor and growth factor signaling cross-talk in prostate cancer cells. Endocr-Relat Cancer 15: 841–849, 2008.

Information & Authors

Information

Published In

cover image Antioxidants & Redox Signaling
Antioxidants & Redox Signaling
Volume 20Issue Number 9March 20, 2014
Pages: 1481 - 1500
PubMed: 24093432

History

Published in print: March 20, 2014
Published online: 27 February 2014
Published ahead of print: 22 January 2014
Published ahead of production: 5 October 2013
Accepted: 5 October 2013
Received: 16 September 2013

Permissions

Request permissions for this article.

Topics

    Authors

    Affiliations

    Lu Miao
    Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky.
    Aaron K. Holley
    Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky.
    Yanming Zhao
    Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky.
    William H. St. Clair
    Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky.
    Daret K. St. Clair
    Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky.

    Notes

    Address correspondence to:Dr. Daret K. St. ClairGraduate Center for ToxicologyUniversity of Kentucky1095 VA Drive, HSRB 454Lexington, KY 40536-0298E-mail: [email protected]

    Metrics & Citations

    Metrics

    Citations

    Export citation

    Select the format you want to export the citations of this publication.

    View Options

    Get Access

    Access content

    To read the fulltext, please use one of the options below to sign in or purchase access.

    Society Access

    If you are a member of a society that has access to this content please log in via your society website and then return to this publication.

    Restore your content access

    Enter your email address to restore your content access:

    Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

    View options

    PDF/EPUB

    View PDF/ePub

    Full Text

    View Full Text

    Media

    Figures

    Other

    Tables

    Share

    Share

    Copy the content Link

    Share on social media

    Back to Top