Browse > Article
http://dx.doi.org/10.22034/APJCP.2016.17.12.5087

New Insights into mTOR Signal Pathways in Ovarian-Related Diseases: Polycystic Ovary Syndrome and Ovarian Cancer  

Liu, Ai Ling (Institute of Biological Science, The Key Laboratory of Biological Toxicology and Ecological Restoration of Hengyang City, School of Pharmaceutical and Biological Science, University of South China)
Liao, Hong Qing (Reproductive Health Hospital of Nanhua-Xinghui in Hengyang City)
Li, Zhi Liang (Institute of Biological Science, The Key Laboratory of Biological Toxicology and Ecological Restoration of Hengyang City, School of Pharmaceutical and Biological Science, University of South China)
Liu, Jun (Institute of Biological Science, The Key Laboratory of Biological Toxicology and Ecological Restoration of Hengyang City, School of Pharmaceutical and Biological Science, University of South China)
Zhou, Cui Lan (Department of Anatomy, University of South China)
Guo, Zi Fen (Institute of Pharmacy and Pharmacology, University of South China)
Xie, Hong Yan (Institute of Biological Science, The Key Laboratory of Biological Toxicology and Ecological Restoration of Hengyang City, School of Pharmaceutical and Biological Science, University of South China)
Peng, Cui Ying (Institute of Biological Science, The Key Laboratory of Biological Toxicology and Ecological Restoration of Hengyang City, School of Pharmaceutical and Biological Science, University of South China)
Publication Information
Asian Pacific Journal of Cancer Prevention / v.17, no.12, 2016 , pp. 5087-5094 More about this Journal
Abstract
mTOR, the mammalian target of rapamycin, is a conserved serine/threonine kinase which belongs to the phosphatidyl-linositol kinase-related kinase (PIKK) family. It has two complexes called mTORC1 and mTORC2. It is well established that mTOR plays important roles in cell growth, proliferation and differentiation. Over-activation of the mTOR pathway is considered to have a relationship with the development of many types of diseases, including polycystic ovary syndrome (PCOS) and ovarian cancer (OC). mTOR pathway inhibitors, such as rapamycin and its derivatives, can directly or indirectly treat or relieve the symptoms of patients suffering from PCOS or OC. Moreover, mTOR inhibitors in combination with other chemical-molecular agents may have extraordinary efficacy. This paper will discuss links between mTOR signaling and PCOS and OC, and explore the mechanisms of mTOR inhibitors in treating these two diseases, with conclusions regarding the most effective therapeutic approaches.
Keywords
mTOR signaling pathway; PCOS; ovarian cancer;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Montero JC, Chen X, Ocana A, Pandiella A (2012). Predominance of mTORC1 over mTORC2 in the regulation of proliferation of ovarian cancer cells: therapeutic implications. Mol Cancer Ther, 11, 1342-52.   DOI
2 Muralidhar GG, Barbolina MV (2015). The miR-200 family: versatile players in epithelial ovarian cancer. Int J Mol Sci, 16, 16833-47.   DOI
3 Mallon R, Feldberg LR, Lucas J, et al (2011). Antitumor efficacy of PKI-587, a highly potent dual PI3K/mTOR kinase inhibitor. Clin Cancer Res, 17, 3193-203.   DOI
4 McCartney CR, Eagleson CA, Marshall JC (2002). Regulation of gonadotropin secretion: implications for polycystic ovary syndrome. Semin Reprod Med, 20, 317-26.   DOI
5 Sancak Y, Thoreen CC, Peterson TR, et al (2007). PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase. Mol Cell, 25, 903-15.   DOI
6 Ohta T, Ohmichi M, Hayasaka T, et al (2006). Inhibition of phosphatidylinositol 3-kinase increases efficacy of cisplatin in in vivo ovarian cancer models. Endocrinology, 147, 1761-9.   DOI
7 Rescigno P, Cerillo I, Ruocco R, et al (2013). New hypothesis on pathogenesis of ovarian cancer lead to future tailored approaches. Biomed Res Int, 2013, 852839.
8 Roa J, Garcia-Galiano D, Varela L, et al (2009). The mammalian target of rapamycin as novel central regulator of puberty onset via modulation of hypothalamic Kiss1 system. Endocrinology, 150, 5016-26.   DOI
9 Schlosshauer PW, Li W, Lin KT, Chan JL, Wang LH (2009). Rapamycin by itself and additively in combination with carboplatin inhibits the growth of ovarian cancer cells. Gynecol Oncol, 114, 516-22.   DOI
10 Sciarretta S, Volpe M, Sadoshima J (2014). Mammalian target of rapamycin signaling in cardiac physiology and disease. Circ Res, 114, 549-64.   DOI
11 Shah KN, Patel SS (2016). Phosphatidylinositide 3-kinase inhibition: A new potential target for the treatment of polycystic ovarian syndrome. Pharm Biol, 54, 975-83.   DOI
12 Mok SC, Kwong J, Welch WR, et al (2007). Etiology and pathogenesis of epithelial ovarian cancer. Dis Markers, 23, 367-76.   DOI
13 Tao K, Yin Y, Shen Q, et al (2016). Akt inhibitor MK-2206 enhances the effect of cisplatin in gastric cancer cells. Biomed Rep, 4, 365-68.   DOI
14 Sivalingam VN, Myers J, Nicholas S, Balen AH, Crosbie EJ (2014). Metformin in reproductive health, pregnancy and gynaecological cancer: established and emerging indications. Hum Reprod Update, 20, 853-68.   DOI
15 Swaroop A, Jaipuriar AS, Gupta SK, et al (2015). Efficacy of a novel fenugreek seed extract (Trigonella foenum-graecum, Furocyst) in polycystic ovary syndrome (PCOS). Int J Med Sci, 12, 825-31.   DOI
16 Takano M, Kikuchi Y, Kudoh K, et al (2011). Weekly administration of temsirolimus for heavily pretreated patients with clear cell carcinoma of the ovary: a report of six cases. Int J Clin Oncol, 16, 605-9.   DOI
17 Tessaro I, Modina SC, Franciosi F, et al (2015). Effect of oral administration of low-dose follicle stimulating hormone on hyperandrogenized mice as a model of polycystic ovary syndrome. J Ovarian Res, 8, 64.   DOI
18 Tsikouras P, Spyros L, Manav B, et al (2015). Features of polycystic Ovary Syndrome in adolescence. J Med Life, 8, 291-6.
19 Yaba A, Bianchi V, Borini A, Johnson J (2008). A putative mitotic checkpoint dependent on mTOR function controls cell proliferation and survival in ovarian granulosa cells. Reprod Sci, 15, 128-38.   DOI
20 Yaba A, Demir N (2012). The mechanism of mTOR (mammalian target of rapamycin) in a mouse model of polycystic ovary syndrome (PCOS). J Ovarian Res, 5, 38.   DOI
21 Yang YM, Choi EJ (2015). Efficacy and safety of metformin or oral contraceptives, or both in polycystic ovary syndrome. Ther Clin Risk Manag, 11, 1345-53.
22 Zhang Y, Zheng Y, Faheem A, et al (2016). A novel AKT inhibitor, AZD5363, inhibits phosphorylation of AKT downstream molecules, and activates phosphorylation of mTOR and SMG-1 dependent on the liver cancer cell type. Oncol Lett, 11, 1685-92.   DOI
23 Yap TA, Garrett MD, Walton MI, et al (2008). Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises. Curr Opin Pharmacol, 8, 393-412.   DOI
24 Yuan R, Kay A, Berg WJ, Lebwohl D (2009). Targeting tumorigenesis: development and use of mTOR inhibitors in cancer therapy. J Hematol Oncol, 2, 45.   DOI
25 Zhang HY, Zhang YF, Han YK, et al (2012). Activation and significance of the PI3K/Akt pathway in endometrium with polycystic ovary syndrome patients. Zhonghua Fu Chan Ke Za Zhi, 47, 19-23.
26 Zhang S, Lu Z, Mao W, et al (2015). CDK5 regulates paclitaxel sensitivity in ovarian cancer cells by modulating AKT Activation, p21Cip1- and p27Kip1-mediated G1 cell cycle arrest and apoptosis. PLoS One, 10, e0131833   DOI
27 Zhang D, Xia H, Zhang W, Fang B (2016). The anti-ovarian cancer activity by WYE-132, a mTORC1/2 dual inhibitor. Tumour Biol, 37, 1327-36.   DOI
28 Zhao Y, Zhang C, Huang Y, et al (2015). Up-regulated expression of WNT5a increases inflammation and oxidative stress via PI3K/AKT/NF-kappaB signaling in the granulosa cells of PCOS patients. J Clin Endocrinol Meta, 100, 201-11.   DOI
29 Zinzalla V, Stracka D, Oppliger W, Hall MN (2011). Activation of mTORC2 by association with the ribosome. Cell, 144, 757-68.   DOI
30 Zhu T, Yuan J, Wang Y, et al (2015). MiR-661 contributed to cell proliferation of human ovarian cancer cells by repressing INPP5J expression. Biomed Pharmacother, 75, 123-8.   DOI
31 Yip PY (2015). Phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin (PI3K-Akt-mTOR) signaling pathway in non-small cell lung cancer. Transl Lung Cancer Res, 4, 165-76.
32 Huang H, Chen AY, Rojanasakul Y, et al (2015). Dietary compounds galangin and myricetin suppress ovarian cancer cell angiogenesis. J Funct Foods, 15, 464-75.   DOI
33 Hong Z, Xiao M, Yang Y, et al (2011). Arsenic disulfide synergizes with the phosphoinositide 3-kinase inhibitor PI-103 to eradicate acute myeloid leukemia stem cells by inducing differentiation. Carcinogenesis, 32, 1550-8.   DOI
34 Hsu MI (2015). Clinical characteristics in Taiwanese women with polycystic ovary syndrome. Clin Exp Reprod Med, 42, 86-93.   DOI
35 Huang S, Houghton PJ (2003). Targeting mTOR signaling for cancer therapy. Curr Opin Pharmacol, 3, 371-7.   DOI
36 Inoki K, Ouyang H, Zhu T, et al (2006). TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell, 126, 955-68.   DOI
37 Jebahi A, Villedieu M, Petigny-Lechartier C, et al (2014). PI3K/mTOR dual inhibitor NVP-BEZ235 decreases Mcl-1 expression and sensitizes ovarian carcinoma cells to Bcl-xL-targeting strategies, provided that Bim expression is induced. Cancer Lett, 348, 38-49.   DOI
38 Johnson SC, Rabinovitch PS, Kaeberlein M (2013). mTOR is a key modulator of ageing and age-related disease. Nature, 493, 338-45.   DOI
39 Kim SH, Juhnn YS, Song YS (2007). Akt involvement in paclitaxel chemoresistance of human ovarian cancer cells. Ann N Y Acad Sci, 1095, 82-9.   DOI
40 Kinross KM, Montgomery KG, Kleinschmidt M, et al (2012). An activating Pi3kca mutation coupled with Pten loss is sufficient to initiate tumorigenesis in mice. J Clin Invest, 122, 553-7   DOI
41 Badura S, Tesanovic T, Pfeifer H, et al (2013). Differential effects of selective inhibitors targeting the PI3K/AKT/mTOR pathway in acute lymphoblastic leukemia. PLoS One, 8, e80070.   DOI
42 Alam H, Maizels ET, Park Y, et al (2004). Follicle-stimulating hormone activation of hypoxia-inducible factor-1 by the phosphatidylinositol 3-kinase/AKT/Ras homolog enriched in brain (Rheb)/mammalian target of rapamycin (mTOR) pathway is necessary for induction of select protein markers of follicular differentiation. J Biol Chem, 279, 19431-40.   DOI
43 Allemand MC, Irving BA, Asmann YW, et al (2009). Effect of testosterone on insulin stimulated IRS1 Ser phosphorylation in primary rat myotubes--a potential model for PCOS-related insulin resistance. PLoS One, 4, e4274.   DOI
44 Ashworth RE, Wu J (2014). Mammalian target of rapamycin inhibition in hepatocellular carcinoma. World J Hepatol, 6, 776-82.   DOI
45 Cheaib B, Auguste A, Leary A (2015). The PI3K/Akt/mTOR pathway in ovarian cancer: therapeutic opportunities and challenges. Chin J Cancer, 34, 4-16.
46 Koizume S, Miyagi Y (2015). Tissue factor-factor VII complex as a key regulator of ovarian cancer phenotypes. Biomark Cancer, 7, 1-13.
47 Bai H, Li H, Li W, et al (2015). The PI3K/AKT/mTOR pathway is a potential predictor of distinct invasive and migratory capacities in human ovarian cancer cell lines. Oncotarget, 6, 25520-32.   DOI
48 Behbakht K, Sill MW, Darcy KM, et al (2011). Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent and recurrent epithelial ovarian and primary peritoneal malignancies: a Gynecologic Oncology Group study. Gynecol Oncol, 123,19-26.   DOI
49 Brugarolas J, Lei K, Hurley RL, et al (2004). Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev, 18, 2893-904.   DOI
50 Campone M, Levy V, Bourbouloux E, et al (2009). Safety and pharmacokinetics of paclitaxel and the oral mTOR inhibitor everolimus in advanced solid tumours. Br J Cancer, 100, 315-21.   DOI
51 Cheng Z, Guo J, Chen L, et al (2015). A long noncoding RNA AB073614 promotes tumorigenesis and predicts poor prognosis in ovarian cancer. Oncotarget, 6, 25381-9.   DOI
52 Chon HJP, Apte S, Lee JH (2014). Phase I study of combination carboplatin, paclitaxel and ridaforolimus in patients with solid, endometrial and ovarian cancers, J Clin Oncol, 32, 2614.
53 Gwinn DM, Shackelford DB, Egan DF, et al (2008). AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell, 30, 214-26.   DOI
54 Dibble CC, Asara JM, Manning BD (2009). Characterization of Rictor phosphorylation sites reveals direct regulation of mTOR complex 2 by S6K1. Mol Cell Biol, 29, 5657-70.   DOI
55 Domecq JP, Prutsky G, Mullan RJ, et al (2013). Adverse effects of the common treatments for polycystic ovary syndrome: a systematic review and meta-analysis. J Clin Endocrinol Metab, 98, 4646-54.   DOI
56 Gao N, Flynn DC, Zhang Z, et al (2004). G1 cell cycle progression and the expression of G1 cyclins are regulated by PI3K/AKT/mTOR/p70S6K1 signaling in human ovarian cancer cells. Am J Physiol Cell Physiol, 287, 281-91.
57 Heitman J, Movva NR, Hall MN (1991). Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science, 253, 905-9.   DOI
58 He Z, Chen AY, Rojanasakul Y, Rankin GO, Chen YC (2016). Gallic acid, a phenolic compound, exerts anti-angiogenic effects via the PTEN/AKT/HIF-1alpha/VEGF signaling pathway in ovarian cancer cells. Oncol Rep, 35, 291-7.   DOI
59 Laplante M, Sabatini DM (2012). mTOR signaling in growth control and disease. Cell, 149, 274-93.   DOI
60 Kollmannsberger C, Hirte H, Siu LL, et al (2012). Temsirolimus in combination with carboplatin and paclitaxel in patients with advanced solid tumors: a NCIC-CTG, phase I, open-label dose-escalation study (IND 179). Ann Oncol, 23, 238-44.   DOI
61 Lee d Y (2015). Roles of mTOR Signaling in Brain Development. Exp Neurobiol, 24, 177-85.   DOI
62 Lengyel E (2010). Ovarian cancer development and metastasis. Am J Pathol, 177, 1053-64.   DOI
63 Li C, Liu VW, Chan DW, Yao KM, Ngan HY (2012). LY294002 and metformin cooperatively enhance the inhibition of growth and the induction of apoptosis of ovarian cancer cells. Int J Gynecol Cancer, 22, 15-22.   DOI
64 Lipton JO, Sahin M (2014). The neurology of mTOR. Neuron, 84, 275-91.   DOI
65 Liu M, Qi Z, Liu B, et al (2015). RY-2f, an isoflavone analog, overcomes cisplatin resistance to inhibit ovarian tumorigenesis via targeting the PI3K/AKT/mTOR signaling pathway. Oncotarget, 6, 25281-94.   DOI
66 Li X, Cui P, Jiang HY, et al (2015). Reversing the reduced level of endometrial GLUT4 expression in polycystic ovary syndrome: a mechanistic study of metformin action. Am J Transl Res, 7, 574-86.
67 Lyu D, Yu W, Tang N, et al (2013). The mTOR signaling pathway regulates pain-related synaptic plasticity in rat entorhinal-hippocampal pathways. Mol Pain, 9, 64.