[1]
|
Butler MS. Natural products to drugs:natural product derived compounds in clinical trials[J]. Nat Prod Rep, 2005, 22(2):162-195. |
[2]
|
Gurib-Fakim A. Medicinal plants:traditions of yesterday and drugs of tomorrow[J]. Mol Aspects Med, 2006, 27(1):1-93. |
[3]
|
Man S, Gao W, Zhang Y, et al. Chemical study and medical application of saponins as anti-cancer agents[J]. Fitoterapia, 2010, 81(7):703-714. |
[4]
|
Shi J, Arunasalam K, Yeung D, et al. Saponins from edible legumes:chemistry, processing, and health benefits[J]. J Med Food, 2004, 7(1):67-78. |
[5]
|
Djerassi C, Rosenkranz G, Pataki J, et al. Steroids, XXVII. Synthesis of allopregnane-3beta, 11beta, 17alpha-, 20beta, 21-pentol from cortisone and diosgenin[J]. J Biol Chem, 1952, 194(1):115-118. |
[6]
|
Raju J, Mehta R. Cancer chemopreventive and therapeutic effects of diosgenin, a food saponin[J]. Nutr Cancer, 2008, 61(1):27-35. |
[7]
|
Yue L, Chen L, Kou JP, et al. Recent advances of diosgenin in its pharmacological activities and mechanism[J]. Chin J Clin Pharmacol Ther, 2010, 15(2):233-237. |
[8]
|
Corbiere C, Liagre B, Bianchi A, et al. Different contribution of apoptosis to the antiproliferative effects of diosgenin and other plant steroids, hecogenin and tigogenin, on human 1547 osteosarcoma cells[J]. Int J Oncol, 2003, 22(4):899-906. |
[9]
|
Corbiere C, Liagre B, Terro F, et al. Induction of antiproliferative effect by diosgenin through activation of p53, release of apoptosis-inducing factor (AIF) and modulation of caspase-3 activity in different human cancer cells[J]. Cell Res, 2004, 14(3):188-196 |
[10]
|
Wang WC, Liu SF, Chang WT, et al. The effects of diosgenin in the regulation of renal proximal tubular fibrosis[J]. Exp Cell Res, 2014, 323(2):255-262. |
[11]
|
Chen PS, Shih YW, Huang HC, et al. Diosgenin, a steroidal saponin, inhibits migration and invasion of human prostate cancer PC-3 cells by reducing matrix metalloproteinases expression[J]. PLos One, 2011, 6(5):e20164. |
[12]
|
He Z, Chen H, Li G, et al. Diosgenin inhibits the migration of human breast cancer MDA-MB-231 cells by suppressing Vav2 activity[J]. Phytomedicine, 2014, 21(6):871-876. |
[13]
|
Mao ZJ, Tang QJ, Zhang CA, et al. Anti-proliferation and anti-invasion effects of diosgenin on gastric cancer BGC-823 cells with HIF-1 shRNAs[J]. Int J Mol Sci, 2012, 13(5):6521-6533. |
[14]
|
Evan GI, Vousden KH. Proliferation, cell cycle and apoptosis in cancer[J]. Nature, 2001, 411(6835):342-348. |
[15]
|
Shishodia S, Aggarwal BB. Diosgenin inhibits osteoclastogenesis, invasion, and proliferation through the downregulation of Akt, IB kinase activation and NF-B-regulated gene expression[J]. Oncogene, 2005, 25(10):1463-1473. |
[16]
|
Chiang CT, Way TD, Tsai SJ, et al. Diosgenin, a naturally occurring steroid, suppresses fatty acid synthase expression in HER2-overexpressing breast cancer cells through modulating Akt, mTOR and JNK phosphorylation[J]. Febs Lett, 2007, 581(30):5735-5742. |
[17]
|
Cailleteau C, Liagre B, Beneytout JL. A proteomic approach to the identification of molecular targets in subsequent apoptosis of HEL cells after diosgenin-induced megakaryocytic differentiation[J]. J Cell Biochem, 2009, 107(4):785-796. |
[18]
|
Raju J, Patlolla JM, Swamy MV, et al. Diosgenin, a steroid saponin of Trigonella foenum graecum (Fenugreek), inhibits azoxymethane-induced aberrant crypt foci formation in F344 rats and induces apoptosis in HT-29 human colon cancer cells[J]. Cancer Epidemiol Biomarkers Prev, 2004, 13(8):1392-1398. |
[19]
|
Mohammad RY, Somayyeh G, Gholamreza H, et al. Diosgenin inhibits hTERT gene expression in the A549 lung cancer cell line[J]. Asian Pac J Cancer Prev, 2013, 14(11):6945-6948. |
[20]
|
Bian D, Li Z, Ma H, et al. Effects of diosgenin on cell proliferation induced by IGF-1 in primary human thyrocytes[J]. Arch Pharm Res, 2011, 34(6):997-1005. |
[21]
|
Mu S, Tian X, Ruan Y, et al. Diosgenin induces apoptosis in IGF-1-stimulated human thyrocytes through two caspasedependent pathways[J]. Biochem Biophys Res Commun, 2012, 418(2):347-352. |
[22]
|
Cai H, Wang Z, Zhang H, et al. Diosgenin relieves goiter via the inhibition of thyrocyte proliferation in a mouse model of Graves' disease[J]. Acta Pharmacol Sini, 2014, 35(1):65-73. |
[23]
|
Mani SA, Guo W, Liao MJ, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells[J]. Cell, 2008, 133(4):704-715. |
[24]
|
Zhou X, Zhang H, Han X. Role of epithelial to mesenchymal transition proteins in gynecological cancers:pathological and therapeutic perspectives[J]. Tumor Biology, 2014, 35(10):9523-9530. |
[25]
|
Chang HY, Kao MC, Way TD, et al. Diosgenin suppresses hepatocyte growth factor (HGF)-induced epithelialmesenchymal transition by down-regulation of Mdm2 and Vimentin[J]. J Agric Food Chem, 2011, 59(10):5357-5363. |
[26]
|
Gialeli C, Theocharis AD, Karamanos NK. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting[J]. FEBS J, 2011, 278(1):16-27. |
[27]
|
Wang W, Goswami S, Sahai E, et al. Tumor cells caught in the act of invading:their strategy for enhanced cell motility[J]. Trends Cell Biol, 2005, 15(3):138-145. |
[28]
|
Le Clainche C, Carlier MF. Regulation of actin assembly associated with protrusion and adhesion in cell migration[J]. Physiol Rev, 2008, 88(2):489-513. |
[29]
|
Rathinam R, Berrier A, Alahari SK. Role of Rho GTPases and their regulators in cancer progression[J]. Front Biosci, 2011, 16(1):2561-2571. |
[30]
|
Lin M, van Golen KL. Rho-regulatory proteins in breast cancer cell motility and invasion[J]. Breast Cancer Res Treat, 2004, 84(1):49-60. |
[31]
|
Mihalache A, Rogoveanu I. Angiogenesis factors involved in the pathogenesis of colorectal cancer[J]. Curr Health Sci J, 2014, 40(1):5-11. |
[32]
|
Shimizu T, Tolcher AW, Papadopoulos KP, et al. The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer[J]. Clin Cancer Res, 2012, 18(8):2316-2325. |
[33]
|
Das S, Dey KK, Dey G, et al. Antineoplastic and apoptotic potential of traditional medicines thymoquinone and diosgenin in squamous cell carcinoma[J]. PLos One, 2012, 7(10):e46641 |
[34]
|
Srinivasan S, Koduru S, Kumar R, et al. Diosgenin targets Akt-mediated prosurvival signaling in human breast cancer cells[J]. Int J Cancer, 2009, 125(4):961-967. |
[35]
|
Kim DS, Jeon BK, Lee YE, et al. Diosgenin induces apoptosis in HepG2 cells through generation of reactive oxygen species and mitochondrial pathway[J]. Evid Based Complement Alternat Med, 2012, 2012:981675. |
[36]
|
Liagre B, Bertrand J, Leger DY, et al. Diosgenin, a plant steroid, induces apoptosis in COX-2 deficient K562 cells with activation of the p38 MAP kinase signalling and inhibition of NF-B binding[J]. Int J Mol Med, 2005, 16(6):1095-1101. |
[37]
|
Bertrand J, Liagre B, Bgaud-Grimaud G, et al. Study of diosgenin-induced apoptosis kinetics in K562 cells by sedimentation field flow fractionation[J]. J Chromatogr B, 2008, 869(1):75-83. |
[38]
|
Yadav VR, Prasad S, Sung B, et al. Targeting inflammatory pathways by triterpenoids for prevention and treatment of cancer[J]. Toxins, 2010, 2(10):2428-2466. |
[39]
|
Shishodia S, Aggarwal BB. Diosgenin inhibits osteoclastogenesis, invasion, and proliferation through the downregulation of Akt, IB kinase activation and NF-B-regulated gene expression[J]. Oncogene, 2006, 25(10):1463-1473. |
[40]
|
Li F, Fernandez PP, Rajendran P, et al. Diosgenin, a steroidal saponin, inhibits STAT3 signaling pathway leading to suppression of proliferation and chemosensitization of human hepatocellular carcinoma cells[J]. Cancer Lett, 2010, 292(2):197-207. |
[41]
|
Khan Z, Khan N, Tiwari RP, et al. Biology of Cox-2:an application in cancer therapeutics[J]. Curr Drug Targets, 2011, 12(7):1082-1093. |
[42]
|
Hsu AL, Ching TT, Wang DS, et al. The cyclooxygenase-2 inhibitor celecoxib induces apoptosis by blocking Akt activation in human prostate cancer cells independently of Bcl-2[J]. J Biol Chem, 2000, 275(15):11397-11403. |
[43]
|
Cailleteau C, Liagre B, Battu S, et al. Increased cyclooxygenase-2 and thromboxane synthase expression is implicated in diosgenin-induced megakaryocytic differentiation in human erythroleukemia cells[J]. Anal Biochem, 2008, 380(1):26-34. |
[44]
|
Lepage C, Liagre B, Cook-Moreau J, et al. Cyclooxygenase-2 and 5-lipoxygenase pathways in diosgenin-induced apoptosis in HT-29 and HCT-116 colon cancer cells[J]. Int J Oncol, 2010, 36(5):1183-1191. |
[45]
|
Lepage C, Lger DY, Bertrand J, et al. Diosgenin induces death receptor-5 through activation of p38 pathway and promotes TRAIL-induced apoptosis in colon cancer cells[J]. Cancer Lett, 2011, 301(2):193-202. |
[46]
|
Miyoshi N, Nagasawa T, Mabuchi R, et al. Chemoprevention of azoxymethane/dextran sodium sulfate-induced mouse colon carcinogenesis by freeze-dried Yam Sanyaku and its constituent diosgenin[J]. Cancer Prev Res, 2011, 4(6):924-934. |
[47]
|
Knudson AG. Two genetic hits (more or less) to cancer[J]. Nat Rev Cancer, 2001, 1(2):157-162. |
[48]
|
Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis[J]. Nat Rev Cancer, 2007,7(10):763-777. |
[49]
|
Baron A, Migita T, Tang D, et al. Fatty acid synthase:a metabolic oncogene in prostate cancer[J]. J Cell Biochem, 2004, 91(1):47-53. |
[50]
|
Lee BH, Taylor MG, Robinet P, et al. Dysregulation of cholesterol homeostasis in human prostate cancer through loss of ABCA1[J]. Cancer Res, 2013, 73(3):1211-1218. |
[51]
|
Guruswamy S, Rao CV. Multi-target approaches in colon cancer chemoprevention based on systems biology of tumor cell-signaling[J]. Gene Regul Syst Biol, 2008, 2:163-176. |
[52]
|
DeBose-Boyd RA. Feedback regulation of cholesterol synthesis:sterol-accelerated ubiquitination and degradation of HMG CoA reductase[J]. Cell Res, 2008, 18(6):609-621. |
[53]
|
Raju J, Bird RP. Diosgenin, a naturally occurring furostanol saponin suppresses 3-hydroxy-3-methylglutaryl CoA reductase expression and induces apoptosis in HCT-116 human colon carcinoma cells[J]. Cancer Lett, 2007, 255(2):194-204. |
[54]
|
Ebrahimi H, Badalzadeh R, Mohammadi M, et al. Diosgenin attenuates inflammatory response induced by myocardial reperfusion injury:role of mitochondrial ATP-sensitive potassium channels[J]. J Physiol Biochem, 2014, 70(2):435-432. |
[55]
|
Salimeh A, Mohammadi M, Rashidi B. Preconditioning with diosgenin and treadmill exercise preserves the cardiac toxicity of isoproterenol in rats[J]. J Physiol Biochem, 2013, 69(2):255-265. |
[56]
|
Jayachandran KS, Vasanthi HR, Rajamanickam GV. Antilipoperoxidative and membrane stabilizing effect of diosgenin, in experimentally induced myocardial infarction[J]. Mol Cell Biochem, 2009, 327(1-2):203-210. |
[57]
|
Gutirrez E, Flammer AJ, Lerman LO, et al. Endothelial dysfunction over the course of coronary artery disease[J]. Eur Heart J, 2013, 34(41):3175-3181. |
[58]
|
McVeigh GE, Brennan GM, Johnston GD, et al. Impaired endothelium-dependent and independent vasodilation in patients with type 2(non-insulin-dependent) diabetes mellitus[J]. Diabetologia, 1992, 35(8):771-776. |
[59]
|
Manivannan J, Balamurugan E, Silambarasan T, et al. Diosgenin improves vascular function by increasing aortic eNOS expression, normalize dyslipidemia and ACE activity in chronic renal failure rats[J]. Mol Cell Biochem, 2013, 384(1-2):113-120. |
[60]
|
Liu K, Zhao W, Gao X, et al. Diosgenin ameliorates palmitate-induced endothelial dysfunction and insulin resistance via blocking IKK and IRS-1 pathways[J]. Atherosclerosis, 2012, 223(2):350-358. |
[61]
|
Dias KL, Correia NA, Pereira KK, et al. Mechanisms involved in the vasodilator effect induced by diosgenin in rat superior mesenteric artery[J]. Eur J Pharmacol, 2007, 574(2):172-178. |
[62]
|
Gong G, Qin Y, Huang W, et al. Protective effects of diosgenin in the hyperlipidemic rat model and in human vascular endothelial cells against hydrogen peroxide-induced apoptosis[J]. Chem-Biol Interact, 2010, 184(3):366-375. |
[63]
|
Au AL, Kwok CC, Lee AT, et al. Activation of iberiotoxin-sensitive, Ca2+ activated K+ channels of porcine isolated left anterior descending coronary artery by diosgenin[J]. Eur J Pharmacol, 2004, 502(1):123-133. |
[64]
|
Esfandiarei M, Lam JT, Yazdi SA, et al. Diosgenin modulates vascular smooth muscle cell function by regulating cell viability, migration, and calcium homeostasis[J]. J Pharmacol Exp Ther, 2011, 336(3):925-939. |
[65]
|
Mayer K, Merfels M, Muhly-Reinholz M, et al. -3 Fatty acids suppress monocyte adhesion to human endothelial cells:role of endothelial PAF generation[J]. Am J Physiol-Heart C, 2002, 283(2):H811-H818. |
[66]
|
Liu YC, Zou XB, Chai YF, et al. Macrophage polarization in inflammatory diseases[J]. Int J Biol Sci, 2014, 10(5):520-529. |
[67]
|
Carlos TM, Schwartz BR, Kovach NL, et al. Vascular cell adhesion molecule-1 mediates lymphocyte adherence to cytokine-activated cultured human endothelial cells[J]. Blood, 1990, 76(5):965-970. |
[68]
|
Ling S, Nheu L, A Komesaroff P. Cell adhesion molecules as pharmaceutical target in atherosclerosis[J]. Mini-Rev Med Chem, 2012, 12(2):175-183. |
[69]
|
Choi KW, Park HJ, Jung DH, et al. Inhibition of TNF--induced adhesion molecule expression by diosgenin in mouse vascular smooth muscle cells via down regulation of the MAPK, Akt and NF-B signaling pathways[J]. Vasc Pharmacol, 2010, 53(5):273-280. |
[70]
|
Song JX, Ma L, Kou JP, et al. Diosgenin reduces leukocytes adhesion and migration linked with inhibition of intercellular adhesion molecule-1 expression and NF-B p65 activation in endothelial cells[J]. Chin J Nat Med, 2012, 10(2):142-149. |
[71]
|
Drozdowska J. Tissue factor in endothelial cells--its structure and function according to the current literature[J]. Postepy Biochem, 2011, 58(3):273-280. |
[72]
|
berg M, Siegbahn A. Tissue factor non-coagulant signaling-molecular mechanisms and biological consequences with a focus on cell migration and apoptosis[J]. J Thromb Haemost, 2013, 11(5):817-825. |
[73]
|
Yang HP, Yue L, Jiang WW, et al. Diosgenin inhibits tumor necrosis factor-induced tissue factor activity and expression in THP-1 cells via down-regulation of the NF-B, Akt, and MAPK signaling pathways[J]. Chin J Nat Med, 2013, 11(6):608-615. |
[74]
|
Gong G, Qin Y, Huang W. Anti-thrombosis effect of diosgenin extract from Dioscorea zingiberensis CH Wright in vitro and in vivo[J]. Phytomedicine, 2011, 18(6):458-463. |
[75]
|
Yan MX, Li YQ, Meng M, et al. Long-term high-fat diet induces pancreatic injuries via pancreatic microcirculatory disturbances and oxidative stress in rats with hyperlipidemia[J]. Biochem Biophys Res Commun, 2006, 347(1):192-199. |
[76]
|
Scheuer H, Gwinner W, Hohbach J, et al. Oxidant stress in hyperlipidemia-induced renal damage[J]. Am J Physiol-Renal, 2000, 278(1):F63-F74. |
[77]
|
Frohnert BI, Bernlohr DA. Protein carbonylation, mitochondrial dysfunction, and insulin resistance[J]. Adv Nutr, 2013, 4(2):157-163. |
[78]
|
Argani H, Ghorbani A, Rashtchizade N, et al. Effect of lovastatin on lipid peroxidation and total antioxidant concentrations in hemodialysis patients[J]. Lipids Health Dis, 2004, 3:6. |
[79]
|
Kregel KC, Zhang HJ. An integrated view of oxidative stress in aging:basic mechanisms, functional effects, and pathological considerations[J]. Am J Physiol-Reg I, 2007, 292(1):R18-R36. |
[80]
|
Czak L, Szabolcs A, Vajda , et al. Hyperlipidemia induced by a cholesterol-rich diet aggravates necrotizing pancreatitis in rats[J]. Eur J Pharmacol, 2007, 572(1):74-81. |
[81]
|
Son IS, Kim JH, Sohn HY, et al. Antioxidative and hypolipidemic effects of diosgenin, a steroidal saponin of yam (Dioscorea spp.), on high-cholesterol fed rats[J]. Biosci Biotech Biochem, 2007, 71(12):3063-3071. |
[82]
|
Manivannan J, Barathkumar TR, Sivasubramanian J, et al. Diosgenin attenuates vascular calcification in chronic renal failure rats[J]. Mol Cell Biochem, 2013, 378(1-2):9-18. |
[83]
|
Esser N, Legrand-Poels S, Piette J, et al. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes[J]. Diabetes Res Clin Pract, 2014, 105(2):141-150. |
[84]
|
Uemura T, Goto T, Kang MS, et al. Diosgenin, the main aglycon of fenugreek, inhibits LXR activity in HepG2 cells and decreases plasma and hepatic triglycerides in obese diabetic mice[J]. J Nutr, 2011, 141(1):17-23. |
[85]
|
Sangeetha MK, ShriShri Mal N, Atmaja K, et al. PPAR's and diosgenin a chemico biological insight in NIDDM[J]. Chem-Biol Interact, 2013, 206(2):403-410. |
[86]
|
Pari L, Monisha P, Mohamed Jalaludeen A. Beneficial role of diosgenin on oxidative stress in aorta of streptozotocin induced diabetic rats[J]. Eur J Pharmacol, 2012, 691(1):143-150. |
[87]
|
Uemura T, Hirai S, Mizoguchi N, et al. Diosgenin present in fenugreek improves glucose metabolism by promoting adipocyte differentiation and inhibiting inflammation in adipose tissues[J]. Mol Nutr Food Res, 2010, 54(11):1596-1608. |
[88]
|
Hirai S, Uemura T, Mizoguchi N, et al. Diosgenin attenuates inflammatory changes in the interaction between adipocytes and macrophages[J]. Mol Nutr Food Res, 2010, 54(6):797-804. |
[89]
|
Xiao L, Guo D, Hu C, et al. Diosgenin promotes oligodendrocyte progenitor cell differentiation through estrogen receptor mediated ERK1/2 activation to accelerate remyelination[J]. Glia, 2012, 60(7):1037-1052. |
[90]
|
Girach A, Vignati L. Diabetic microvascular complications-can the presence of one predict the development of another[J]. J Diabetes Complicat, 2006, 20(4):228-237. |
[91]
|
Cheng HT, Dauch JR, Hayes JM, et al. Nerve growth factor mediates mechanical allodynia in a mouse model of type 2 diabetes[J]. J Neuropath Exp Neur, 2009, 68(11):1229-1243. |
[92]
|
Kang TH, Moon E, Hong BN, et al. Diosgenin from Dioscorea nipponica ameliorates diabetic neuropathy by inducing nerve growth factor[J]. Biol Pharm Bull, 2011, 34(9):1493-1498. |
[93]
|
Kondeva-Burdina M, Lopez MU, Mitcheva M, et al. In vitro effect of diosgenin, isolated from Asparagus officinalis, in a dopamine model of oxidative stress in isolated rat synaptosomes[J]. Toxicol Lett, 2007, 172(s7):S68. |
[94]
|
Chiu CS, Chiu YJ, Wu LY, et al. Diosgenin ameliorates cognition deficit and attenuates oxidative damage in senescent mice induced by D-galactose[J]. Am J Chin Med, 2011, 39(3):551-563. |
[95]
|
Tohda C, Lee YA, Goto Y, et al. Diosgenin-induced cognitive enhancement in normal mice is mediated by 1, 25D3-MARRS[J]. Sci Rep, 2013, 3:3395. |
[96]
|
Wang YJ, Liu YC, Chang HD, et al. Diosgenin, a plant-derived sapogenin, stimulates Ca2+ -activated K+ current in human cortical HCN-1A neuronal cells[J]. Planta Med, 2006, 72(5):430-436. |
[97]
|
Huang CH, Liu DZ, Jan TR. Diosgenin, a plant-derived sapogenin, enhances regulatory T-cell immunity in the intestine of mice with food allergy[J]. J Nat Prod, 2010, 73(6):1033-1037. |
[98]
|
Huang CH, Ku CY, Jan TR. Diosgenin attenuates allergen-induced intestinal inflammation and IgE production in a murine model of food allergy[J]. Planta Med, 2009, 75(12):1300-1305. |
[99]
|
Jan TR, Wey SP, Kuan CC, et al. Diosgenin, a steroidal sapogenin, enhances antigen-specific IgG2a and interferon- expression in ovalbumin-sensitized BALB/c mice[J]. Planta Med, 2007, 73(5):421-426. |
[100]
|
Ku CM, Lin JY. Anti-inflammatory effects of 27 selected terpenoid compounds tested through modulating Th1/Th2 cytokine secretion profiles using murine primary splenocytes[J]. Food Chem, 2013, 141(2):1104-1113. |
[101]
|
Gao M, Chen L, Yu H, et al. Diosgenin down-regulates NF-B p65/p50 and p38MAPK pathways and attenuates acute lung injury induced by lipopolysaccharide in mice[J]. Int Immunopharmacol, 2013, 15(2):240-245. |
[102]
|
Jung DH, Park HJ, Byun HE, et al. Diosgenin inhibits macrophage-derived inflammatory mediators through downregulation of CK2, JNK, NF-B and AP-1 activation[J]. Int Immunopharmacol, 2010, 10(9):1047-1054. |
[103]
|
He Z, Tian Y, Zhang X, et al. Anti-tumour and immunomodulating activities of diosgenin, a naturally occurring steroidal saponin[J]. Nat Prod Res, 2012, 26(23):2243-2246. |
[104]
|
Conner P. Breast response to menopausal hormone therapy-aspects on proliferation, apoptosis and mammographic density[J]. Ann Med, 2007, 39(1):28-41. |
[105]
|
Komesaroff PA, Black CV, Cable V, et al. Effects of wild yam extract on menopausal symptoms, lipids and sex hormones in healthy menopausal women[J]. Climacteric, 2001, 4(2):144-150. |
[106]
|
Accatino L, Pizarro M, Sols N, et al. Effects of diosgenin, a plant-derived steroid, on bile secretion and hepatocellular cholestasis induced by estrogens in the rat[J]. Hepatology, 1998, 28(1):129-140. |
[107]
|
Rao AR, Kale RK. Diosgenin-a growth stimulator of mammary gland of ovariectomized mouse[J]. Indian J Exp Biol, 1992, 30(5):367-370. |
[108]
|
Chiang SS, Chang SP, Pan TM. Osteoprotective effect of monascus-fermented dioscorea in ovariectomized rat model of postmenopausal osteoporosis[J]. J Agric Food Chem, 2011, 59(17):9150-9157. |
[109]
|
Higdon K, Scott A, Tucci M, et al. The use of estrogen, DHEA, and diosgenin in a sustained delivery setting as a novel treatment approach for osteoporosis in the ovariectomized adult rat model[J]. Biomed Sci Instrum, 2001, 37:281-286. |
[110]
|
Yen ML, Su JL, Chien CL, et al. Diosgenin induces hypoxia-inducible factor-1 activation and angiogenesis through estrogen receptor-related phosphatidylinositol 3-kinase/Akt and p38 mitogen-activated protein kinase pathways in osteoblasts[J]. Mol Pharmacol, 2005, 68(4):1061-1073. |
[111]
|
Medigović I, Ristić N, Živanović J, et al. Diosgenin does not express estrogenic activity:a uterotrophic assay[J]. Can J Physiol Pharm, 2014, 92(4):292-298. |
[112]
|
Hsu KH, Chang CC, Tsai HD, et al. Effects of yam and diosgenin on calpain systems in skeletal muscle of ovariectomized rats[J]. Taiwan J Obstet Gynecol, 2008, 47(2):180-186. |
[113]
|
Tada Y, Kanda N, Haratake A, et al. Novel effects of diosgenin on skin aging[J]. Steroids, 2009, 74(6):504-511. |
[114]
|
Lee J, Jung K, Kim YS, et al. Diosgenin inhibits melanogenesis through the activation of phosphatidylinositol-3-kinase pathway (PI3K) signaling[J]. Life Sci, 2007, 81(3):249-254. |
[115]
|
Zheng L, Zhou Y, Zhang JY, et al. Two new steroidal saponins from the rhizomes of Dioscorea zingiberensis[J]. Chin J Nat Med, 2014, 12(2):142-147. |
[116]
|
Noguchi E, Fujiwara Y, Matsushita S, et al. Metabolism of tomato steroidal glycosides in humans[J]. Chem Pharm Bull, 2006, 54(9):1312-1314. |