Activation of peroxisome proliferator-activated receptor gamma is crucial for antitumoral effects of 6-iodolactone

Mario Nava-Villalba, Rosa E Nuñez-Anita, Alexander Bontempo, Carmen Aceves, Mario Nava-Villalba, Rosa E Nuñez-Anita, Alexander Bontempo, Carmen Aceves

Abstract

Background: Molecular iodine (I2) exhibits antiproliferative and apoptotic effects on in vivo and in vitro cancer models. These effects are thought to be mediated by an iodinated arachidonic acid derivative, 6-iodolactone (6IL), and one of the proposed mechanisms is that 6IL activates Peroxisome Proliferator-Activated Receptors type gamma (PPARG). These receptors have been implicated in the inhibition of carcinogenic processes, in addition to their classical role in maintaining lipid and glucose homeostasis. The aim of this study was to determine whether PPARG participates in the 6IL antiproliferative and apoptotic effects on the mammary cancer cell line MCF-7.

Methods: The 6IL/PPARG complex was inhibited by the PPARG antagonist GW9662, in both an endogenous and overexpressed (adenoviral vector infection) context, and stable PPARG-knockdown MCF-7 cells (RNA interference, confirmed with hydrolysis probes and Western blot), were used to corroborate the PPARG participation. 6IL effects on proliferation (measured by Trypan Blue exclusion) and apoptosis (phosphatidylserine identification by flow cytometer) were evaluated in conditions of chemical inhibition (GW9662) and silencing (RNA interference). A wound-healing assay was conducted on wild-type and stable PPARG-knockdown MCF-7 cells to evaluate the antimigrational effect of 6IL. Caspase-8 activity was evaluated to determine if the extrinsic pathway is involved in the effects of 6IL and I2 treatment.

Results: Antiproliferative and pro-apoptotic 6IL effects require the activation of PPARG. In addition, wound-healing assays show that 6IL is able to inhibit MCF-7 cell migration and that PPARG plays a role in this phenomenon. Finally, the data exclude the participation of the extrinsic apoptotic pathway in 6IL- and I2-induced apoptosis.

Conclusions: These results support the previously proposed mechanism, in which the I2 effects are mediated by 6IL, and they provide further support for the use of I2 as coadjuvant in breast cancer treatment.

Figures

Fig. 1
Fig. 1
Antiproliferative and apoptotic effect of 6IL is blocked by GW9662 (GW). a MCF-7 cells were treated with increasing GW9662 concentrations to determine the optimal concentration that did not inhibit proliferation; 0.5 μM was selected for use in subsequent experiments. b Proliferation in the presence of 10 μM 6IL and after GW pretreatment was analyzed by the Trypan Blue exclusion assay. c Apoptosis was analyzed by flow cytometry (c, d). Results are expressed as percent change with respect to the control. Data are expressed as mean ± SD (n = 4 independent assays), and the asterisk indicates a significant difference with respect to the control (P < 0.05)
Fig. 2
Fig. 2
Overexpression of PPARG enhances antiproliferative and apoptotic effects of 6IL, and these enhancements are partially prevented by GW9662 (GW). a Representative panels of the infection-efficiency assay. MCF-7 cells were infected (24 h) with AdGFP at increasing multiplicities of infection (MOIs). GFP expression is reported as a percentage of the number of events (%). bPPARG expression in AdPPARG-infected cells was analyzed by qPCR and normalized to ACTB expression. c A range of MOIs was analyzed to determine the optimal level with no effect on proliferation; an MOI of 50 was selected for subsequent experiments. d Fatty acid synthase (FASN) expression was analyzed by the qPCR assay, and the results were normalized to ACTB expression. e Proliferation in the presence of 10 μM 6IL and after GW pretreatment was analyzed by the Trypan Blue exclusion assay. f The apoptotic effect of 6IL was analyzed by flow cytometry, and the results are expressed as percent change with respect to the control. Data are expressed as mean ± SD (n = 4 independent assays); the asterisks indicate significant differences with respect to the control (P < 0.05), and different letters indicate significant differences between groups (P < 0.05)
Fig. 3
Fig. 3
Knockdown of PPARG blocks the 6IL effects. MCF-7 cells were transfected with a retroviral silencing plasmid (HuSH™) containing a short, specific hairpin with RNA interference against PPARG (RNAi) or containing a scramble sequence (Scramble). a Representative microphotographs of GFP expression in stably transfected MCF-7 cells (20× objective, scale bar 50 μm). bPPARG expression was analyzed by qPCR and normalized to ACTB expression. c PPARG protein was quantified by Western blot and densitometry and is reported as percent change with respect to wild-type (WT) cells. d Proliferation in the presence of 10 μM 6IL was analyzed by Trypan Blue exclusion. e The apoptotic effect of 6IL was analyzed by flow cytometry, and the results are expressed as percent change with respect to the corresponding control. Data are expressed as mean ± SD (n = 4 independent assays), and different letters indicate significant differences between groups (P < 0.05)
Fig. 4
Fig. 4
6IL inhibits MCF-7 migration and this effect is partially reverted by PPARG knockdown. Wild type, Scramble- and RNAi-PPARG transfected MCF-7 cells were treated with 10 μM 6IL for 24 h, and the number of cells that had migrated into the scratch during this time was determined (10× objective, scale bar 200 μm, average scratch width 377 μm). a Representative microphotographs of wound-healing assays. b Migrating cells (total number). Data are expressed as mean ± SD (n = 3 independent assays), two-way ANOVA and Tukey’s multiple comparison tests were performed; different letters indicate significant differences between groups (P < 0.05)
Fig. 5
Fig. 5
Extrinsic apoptotic pathway is not involved in the induction of apoptosis by 6IL or I2. MCF-7 cells were treated with 10 μM 6IL or 100 μM I2 for 48 h. Caspase-8 activity was analyzed by a colorimetric assay and normalized for the amount of protein. The results are reported as percent change with respect to the control. Data are expressed as mean ± SD (n = 3 independent assays). There were no significant differences between groups

References

    1. Arroyo-Helguera O, Rojas E, Delgado G, Aceves C. Signaling pathways involved in the antiproliferative effect of molecular iodine in normal and tumoral breast cells: evidence that 6-iodolactone mediates apoptotic effects. Endocr Relat Cancer. 2008;15:1003–11. doi: 10.1677/ERC-08-0125.
    1. Aceves C, García-Solís P, Arroyo-Helguera O, Vega-Riveroll L, Delgado G, Anguiano B. Antineoplastic effect of iodine in mammary cancer: participation of 6-iodolactone (6-IL) and peroxisome proliferator-activated receptors (PPAR) Mol Cancer. 2009;8:33. doi: 10.1186/1476-4598-8-33.
    1. Nava-Villalba M, Aceves C. 6-Iodolactone, key mediator of antitumoral properties of iodine. Prostaglandins Other Lipid Mediat. 2014;112C:27–33. doi: 10.1016/j.prostaglandins.2014.07.001.
    1. García-Solís P, Alfaro Y, Anguiano B, Delgado G, Guzman RC, Nandi S, et al. Inhibition of N-methyl-N-nitrosourea-induced mammary carcinogenesis by molecular iodine (I2) but not by iodide (I-) treatment evidence that I2 prevents cancer promotion. Mol Cell Endocrinol. 2005;236:49–57. doi: 10.1016/j.mce.2005.03.001.
    1. Arroyo-Helguera O, Anguiano B, Delgado G, Aceves C. Uptake and antiproliferative effect of molecular iodine in the MCF-7 breast cancer cell line. Endocr Relat Cancer. 2006;13:1147–58. doi: 10.1677/erc.1.01250.
    1. Rösner H, Torremante P, Möller W, Gärtner R. Antiproliferative/cytotoxic activity of molecular iodine and iodolactones in various human carcinoma cell lines. No interfering with EGF-signaling, but evidence for apoptosis. Exp Clin Endocrinol Diabetes. 2010;118:410–9. doi: 10.1055/s-0029-1225615.
    1. Gärtner R, Rank P, Ander B. The role of iodine and delta-iodolactone in growth and apoptosis of malignant thyroid epithelial cells and breast cancer cells. Horm (Athens) 2010;9:60–6. doi: 10.14310/horm.2002.1254.
    1. Aranda N, Sosa S, Delgado G, Aceves C, Anguiano B. Uptake and antitumoral effects of iodine and 6-iodolactone in differentiated and undifferentiated human prostate cancer cell lines. Prostate. 2013;73:31–41. doi: 10.1002/pros.22536.
    1. Shrivastava A, Tiwari M, Sinha RA, Kumar A, Balapure AK, Bajpai VK, et al. Molecular iodine induces caspase-independent apoptosis in human breast carcinoma cells involving the mitochondria-mediated pathway. J Biol Chem. 2006;281:19762–71. doi: 10.1074/jbc.M600746200.
    1. Nuñez-Anita RE, Arroyo-Helguera O, Cajero-Juárez M, López-Bojorquez L, Aceves C. A complex between 6-iodolactone and the peroxisome proliferator-activated receptor type gamma may mediate the antineoplastic effect of iodine in mammary cancer. Prostaglandins Other Lipid Mediat. 2009;89:34–42. doi: 10.1016/j.prostaglandins.2009.04.001.
    1. Michalik L, Desvergne B, Wahli W. Peroxisome-proliferator-activated receptors and cancers: complex stories. Nat Rev Cancer. 2004;4:61–70. doi: 10.1038/nrc1254.
    1. Tachibana K, Yamasaki D, Ishimoto K, Doi T. The role of PPARs in cancer. PPAR Res. 2008;2008:102737. doi: 10.1155/2008/102737.
    1. Nunez-Anita RE, Cajero-Juarez M, Aceves C. Peroxisome proliferator-activated receptors: role of isoform gamma in the antineoplastic effect of iodine in mammary cancer. Curr Cancer Drug Targets. 2011;11:775–86. doi: 10.2174/156800911796798931.
    1. Roberts-Thomson SJ, Snyderwine EG. Characterization of peroxisome proliferator-activated receptor alpha in normal rat mammary gland and 2-amino-l-methyl-6-phenylimidazo[4, 5-b]pyridine-induced mammary gland tumors from rats fed high and low fat diets. Toxicol Lett. 2000;118:79–86. doi: 10.1016/S0378-4274(00)00265-4.
    1. Suchanek KM, May FJ, Robinson JA, Lee WJ, Holman NA, Monteith GR, et al. Peroxisome proliferator-activated receptor alpha in the human breast cancer cell lines MCF-7 and MDA-MB-231. Mol Carcinog. 2002;34:165–71. doi: 10.1002/mc.10061.
    1. Germain P, Staels B, Dacquet C, Spedding M, Laudet V. Overview of nomenclature of nuclear receptors. Pharmacol Rev. 2006;58:685–704. doi: 10.1124/pr.58.4.2.
    1. Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, et al. International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev. 2006;58:726–41. doi: 10.1124/pr.58.4.5.
    1. Elstner E, Muller C, Koshizuka K, Williamson EA, Park D, Asou H, et al. Ligands for peroxisome proliferator-activated receptor gamma and retinoic acid receptor inhibit growth and induce apoptosis of human breast cancer cells in vitro and in BNX mice. Proc Natl Acad Sci U S A. 1998;95:8806–11. doi: 10.1073/pnas.95.15.8806.
    1. Mueller E, Sarraf P, Tontonoz P, Evans RM, Martin KJ, Zhang M, et al. Terminal differentiation of human breast cancer through PPAR gamma. Mol Cell. 1998;1:465–70. doi: 10.1016/S1097-2765(00)80047-7.
    1. Yin F, Wakino S, Liu Z, Kim S, Hsueh WA, Collins AR, et al. Troglitazone inhibits growth of MCF-7 breast carcinoma cells by targeting G1 cell cycle regulators. Biochem Biophys Res Commun. 2001;286:916–22. doi: 10.1006/bbrc.2001.5491.
    1. Lapillonne H, Konopleva M, Tsao T, Gold D, McQueen T, Sutherland RL, et al. Activation of peroxisome proliferator-activated receptor gamma by a novel synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid induces growth arrest and apoptosis in breast cancer cells. Cancer Res. 2003;63:5926–39.
    1. Yin Y, Russell RG, Dettin LE, Bai R, Wei ZL, Kozikowski AP, et al. Peroxisome proliferator-activated receptor delta and gamma agonists differentially alter tumor differentiation and progression during mammary carcinogenesis. Cancer Res. 2005;65:3950–7. doi: 10.1158/0008-5472.CAN-04-3990.
    1. Papi A, Guarnieri T, Storci G, Santini D, Ceccarelli C, Taffurelli M, et al. Nuclear receptors agonists exert opposing effects on the inflammation dependent survival of breast cancer stem cells. Cell Death Differ. 2012;19:1208–19. doi: 10.1038/cdd.2011.207.
    1. Papi A, Storci G, Guarnieri T, De Carolis S, Bertoni S, Avenia N, et al. Peroxisome proliferator activated receptor-α/hypoxia inducible factor-1α interplay sustains carbonic anhydrase IX and apoliprotein E expression in breast cancer stem cells. PLoS One. 2013;8:e54968. doi: 10.1371/journal.pone.0054968.
    1. Seargent JM, Yates EA, Gill JH. GW9662, a potent antagonist of PPARgamma, inhibits growth of breast tumour cells and promotes the anticancer effects of the PPARgamma agonist rosiglitazone, independently of PPARgamma activation. Br J Pharmacol. 2004;143:933–7. doi: 10.1038/sj.bjp.0705973.
    1. Eskin BA, Grotkowski CE, Connolly CP, Ghent WR. Different tissue responses for iodine and iodide in rat thyroid and mammary glands. Biol Trace Elem Res. 1995;49:9–19. doi: 10.1007/BF02788999.
    1. Funahashi H, Imai T, Tanaka Y, Tobinaga J, Wada M, Morita T, et al. Suppressive effect of iodine on DMBA-induced breast tumor growth in the rat. J Surg Oncol. 1996;61:5. doi: 10.1002/(SICI)1096-9098(199603)61:3<209::AID-JSO9>;2-F.
    1. Soriano O, Delgado G, Anguiano B, Petrosyan P, Molina-Servín ED, Gonsebatt ME, et al. Antineoplastic effect of iodine and iodide in dimethylbenz[a]anthracene-induced mammary tumors: association between lactoperoxidase and estrogen-adduct production. Endocr Relat Cancer. 2011;18:529–39. doi: 10.1530/ERC-11-0065.
    1. Alfaro Y, Delgado G, Cárabez A, Anguiano B, Aceves C. Iodine and doxorubicin, a good combination for mammary cancer treatment: antineoplastic adjuvancy, chemoresistance inhibition, and cardioprotection. Mol Cancer. 2013;12:45. doi: 10.1186/1476-4598-12-45.
    1. Ghent WR, Eskin BA, Low DA, Hill LP. Iodine replacement in fibrocystic disease of the breast. Can J Surg. 1993;36:453–60.
    1. Kessler JH. The effect of supraphysiologic levels of iodine on patients with cyclic mastalgia. Breast J. 2004;10:328–36. doi: 10.1111/j.1075-122X.2004.21341.x.
    1. Anguiano B, Ledezma O, Juarez M, Nuñez F, Aceves C. Therapeutic effect of iodine on benign human prostatic hyperplasia. In: 14th International Thyroid Congress. Paris, France: American Thyroid Association; 2010. p. 0051.
    1. Peralta G, Torres J, Delgado G, Domınguez A, De Obaldıa R, Duarte L, et al. 102nd Annual Meeting, AACR. Orlando, Florida: AACR; 2011. Iodine exhibits dual effects on breast cancer as a co-treatment with anthracyclines: anti- neoplastic synergy and cardioprotector; p. 3509/16.
    1. Cann SA, van Netten JP, van Netten C. Hypothesis: iodine, selenium and the development of breast cancer. Cancer Causes Control. 2000;11:121–7. doi: 10.1023/A:1008925301459.
    1. Venturi S, Donati FM, Venturi A, Venturi M, Grossi L, Guidi A. Role of iodine in evolution and carcinogenesis of thyroid, breast and stomach. Adv Clin Path. 2000;4:11–7.
    1. Venturi S. Is there a role for iodine in breast diseases? Breast. 2001;10:379–82. doi: 10.1054/brst.2000.0267.
    1. Olvera-Caltzontzin P, Delgado G, Aceves C, Anguiano B. Iodine uptake and prostate cancer in the TRAMP mouse model. Mol Med. 2013;19:409–16. doi: 10.2119/molmed.2013.00093.
    1. Dugrillon A, Bechtner G, Uedelhoven WM, Weber PC, Gärtner R. Evidence that an iodolactone mediates the inhibitory effect of iodide on thyroid cell proliferation but not on adenosine 3′,5′-monophosphate formation. Endocrinology. 1990;127:337–43. doi: 10.1210/endo-127-1-337.
    1. Dugrillon A, Uedelhoven WM, Pisarev MA, Bechtner G, Gärtner R. Identification of delta-iodolactone in iodide treated human goiter and its inhibitory effect on proliferation of human thyroid follicles. Horm Metab Res. 1994;26:465–9. doi: 10.1055/s-2007-1001734.
    1. Leesnitzer LM, Parks DJ, Bledsoe RK, Cobb JE, Collins JL, Consler TG, et al. Functional consequences of cysteine modification in the ligand binding sites of peroxisome proliferator activated receptors by GW9662. Biochemistry. 2002;41:6640–50. doi: 10.1021/bi0159581.
    1. Hondares E, Mora O, Yubero P, Rodriguez de la Concepción M, Iglesias R, Giralt M, et al. Thiazolidinediones and rexinoids induce peroxisome proliferator-activated receptor-coactivator (PGC)-1alpha gene transcription: an autoregulatory loop controls PGC-1alpha expression in adipocytes via peroxisome proliferator-activated receptor-gamma coactivation. Endocrinology. 2006;147:2829–38. doi: 10.1210/en.2006-0070.
    1. Wakabayashi K, Okamura M, Tsutsumi S, Nishikawa NS, Tanaka T, Sakakibara I, et al. The peroxisome proliferator-activated receptor gamma/retinoid X receptor alpha heterodimer targets the histone modification enzyme PR-Set7/Setd8 gene and regulates adipogenesis through a positive feedback loop. Mol Cell Biol. 2009;29:3544–55. doi: 10.1128/MCB.01856-08.
    1. Koirala K, Wan Y, Liu Y, Wang X, Cui W, Wang C, et al. Effects of peroxisome proliferator-activated receptor-gamma ligand troglitazone on colon cancer cell growth. Beijing Da Xue Xue Bao. 2006;38(4):385–8.
    1. Fujii D, Yoshida K, Tanabe K, Hihara J, Toge T. The ligands of peroxisome proliferator-activated receptor (PPAR) gamma inhibit growth of human esophageal carcinoma cells through induction of apoptosis and cell cycle arrest. Anticancer Res. 2004;24:1409–16.
    1. Viswakarma N, Jia Y, Bai L, Vluggens A, Borensztajn J, Xu J. et al. Coactivators in PPAR-Regulated Gene Expression. PPAR Res. 2010; doi:10.1155/2010/250126.
    1. Kadegowda AKG, Bionaz M, Piperova LS, Erdman RA, Loor JJ. Peroxisome proliferator-activated receptor-gamma activation and long-chain fatty acids alter lipogenic gene networks in bovine mammary epithelial cells to various extents. J Dairy Sci. 2009;92:4276–89. doi: 10.3168/jds.2008-1932.
    1. Bogacka I, Xie H, Bray GA, Smith SR. The effect of pioglitazone on peroxisome proliferator-activated receptor-gamma target genes related to lipid storage in vivo. Diabetes Care. 2004;27:1660–7. doi: 10.2337/diacare.27.7.1660.
    1. Garcia-Bates TM, Peslak SA, Baglole CJ, Maggirwar SB, Bernstein SH, Phipps RP. Peroxisome proliferator-activated receptor gamma overexpression and knockdown: impact on human B cell lymphoma proliferation and survival. Cancer Immunol Immunother. 2009;58:1071–83. doi: 10.1007/s00262-008-0625-z.
    1. Qin L, Gong C, Chen AM, Guo FJ, Xu F, Ren Y, et al. Peroxisome proliferator activated receptor γ agonist rosiglitazone inhibits migration and invasion of prostate cancer cells through inhibition of the CXCR4/CXCL12 axis. Mol Med Rep. 2014;10:695–700.
    1. Reka AK, Kurapati H, Narala VR, Bommer G, Chen J, Standiford TJ, et al. Peroxisome proliferator-activated receptor-gamma activation inhibits tumor metastasis by antagonizing Smad3-mediated epithelial-mesenchymal transition. Mol Cancer Ther. 2010;9:3221–32. doi: 10.1158/1535-7163.MCT-10-0570.
    1. Bonofiglio D, Gabriele S, Aquila S, Qi H, Belmonte M, Catalano S, et al. Peroxisome proliferator-activated receptor gamma activates fas ligand gene promoter inducing apoptosis in human breast cancer cells. Breast Cancer Res Treat. 2009;113:423–34. doi: 10.1007/s10549-008-9944-1.
    1. Yao Y, Li L, Huang X, Gu X, Xu Z, Zhang Y, et al. SERPINA3K induces apoptosis in human colorectal cancer cells via activating the Fas/FasL/caspase-8 signaling pathway. FEBS J. 2013;280:3244–55. doi: 10.1111/febs.12303.
    1. Monteagudo ES, Caro HN, Veleiro AS, Pisarev MA, Burton G. Synthesis and characterization of iodinated derivatives of arachidonic acid. An Asoc Quím Argent. 1990;78:31–6.

Source: PubMed

3
Abonner