Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Research Article

Genipin Inhibits the Development of Osteosarcoma through PI3K/AKT Signaling Pathway

Author(s): Xiongjie Huang, Habu Jiwa, Jingtao Xu, Jun Zhang, Yanran Huang and Xiaoji Luo*

Volume 29, Issue 16, 2023

Published on: 31 May, 2023

Page: [1300 - 1310] Pages: 11

DOI: 10.2174/1381612829666230508145533

Price: $65

Abstract

Background: Osteosarcoma is a highly invasive and early metastatic tumor. At present, the toxic and side effects of chemotherapy affect the quality of life of cancer patients to varying degrees. Genipin is an extract of the natural medicine gardenia with various pharmacological activities.

Objective: The purpose of this study was to investigate the effect of Genipin on osteosarcoma and its potential mechanism of action.

Methods: Crystal violet staining, MTT assay and colony formation assay were used to detect the effect of genipin on the proliferation of osteosarcoma. The effects of vitexin on migration and invasion of osteosarcoma were detected by scratch healing assay and transwell assay. Hoechst staining and flow cytometry were used to detect the effect of genipin on apoptosis of osteosarcoma cells. The expression of related proteins was detected by Western blot. An orthotopic tumorigenic animal model was used to verify the effect of genipin on osteosarcoma in vivo.

Results: The results of crystal violet staining, MTT method and colony formation method proved that genipin significantly inhibited the proliferation of osteosarcoma cells. The results of the scratch healing assay and transwell assay showed that gen significantly inhibited the migration and invasion of osteosarcoma cells. The results of Hoechst staining and flow cytometry showed that genipin significantly promoted the apoptosis of osteosarcoma cells. The results of animal experiments show that genipin has the same anti-tumor effect in vivo. Genipin may inhibit the growth of osteosarcoma through PI3K/AKT signaling.

Conclusion: Genipin can inhibit the growth of human osteosarcoma cells, and its mechanism may be related to the regulation of PI3K/AKT signaling pathway.

Keywords: Genipin, osteosarcoma, proliferation, migration, apoptosis, PI3K/AKT signaling.

« Previous
[1]
Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin 2015; 65(2): 87-108.
[http://dx.doi.org/10.3322/caac.21262] [PMID: 25651787]
[2]
Song QH, Guo MJ, Zheng JS, Zheng XH, Ye ZH, Wei P. Study on targeting relationship between miR-320b and FGD5-AS1 and its effect on biological function of osteosarcoma cells. Cancer Manag Res 2020; 12: 13589-98.
[http://dx.doi.org/10.2147/CMAR.S264682] [PMID: 33408528]
[3]
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020; 70(1): 7-30.
[http://dx.doi.org/10.3322/caac.21590] [PMID: 31912902]
[4]
Berhe S, Danzer E, Meyers PA, Behr G, LaQuaglia MP, Price AP. Unusual abdominal metastases in osteosarcoma. J Pediatr Surg Case Rep 2018; 28: 13-6.
[http://dx.doi.org/10.1016/j.epsc.2017.09.022] [PMID: 29085778]
[5]
Chen C, Xie L, Ren T, Huang Y, Xu J, Guo W. Immunotherapy for osteosarcoma: Fundamental mechanism, rationale, and recent breakthroughs. Cancer Lett 2021; 500: 1-10.
[http://dx.doi.org/10.1016/j.canlet.2020.12.024] [PMID: 33359211]
[6]
Wang L, Yuan Q, Tan M, et al. Evaluation of efficacy and nephrotoxicity during vancomycin therapy: A retrospective study in China. Exp Ther Med 2019; 17(3): 2389-96.
[http://dx.doi.org/10.3892/etm.2019.7188] [PMID: 30867724]
[7]
Harrison RT, DeBacker JR, Bielefeld EC. A low-dose regimen of cisplatin before high-dose cisplatin potentiates ototoxicity. Laryngoscope 2015; 125(2): E78-83.
[http://dx.doi.org/10.1002/lary.24948] [PMID: 25267530]
[8]
Khasabova IA, Khasabov S, Paz J, Harding-Rose C, Simone DA, Seybold VS. Cannabinoid type-1 receptor reduces pain and neurotoxicity produced by chemotherapy. J Neurosci 2012; 32(20): 7091-101.
[http://dx.doi.org/10.1523/JNEUROSCI.0403-12.2012] [PMID: 22593077]
[9]
Zhang CY, Parton LE, Ye CP, et al. Genipin inhibits UCP2-mediated proton leak and acutely reverses obesity- and high glucose-induced β cell dysfunction in isolated pancreatic islets. Cell Metab 2006; 3(6): 417-27.
[http://dx.doi.org/10.1016/j.cmet.2006.04.010] [PMID: 16753577]
[10]
Kimura Y, Okuda H, Arichi S. Effects of geniposide isolated from Gardenia jasminoides on metabolic alterations in high sugar diet-fed rats. Chem Pharm Bull 1982; 30(12): 4444-7.
[http://dx.doi.org/10.1248/cpb.30.4444] [PMID: 7168870]
[11]
Miyasita S. A historical study of Chinese drugs for the treatment of Jaundice. Am J Chin Med 1976; 4(3): 239-43.
[http://dx.doi.org/10.1142/S0192415X76000317] [PMID: 788496]
[12]
Tseng TH, Chu CY, Huang JM, Shiow SJ, Wang CJ. Crocetin protects against oxidative damage in rat primary hepatocytes. Cancer Lett 1995; 97(1): 61-7.
[http://dx.doi.org/10.1016/0304-3835(95)03964-X] [PMID: 7585479]
[13]
Shan M, Yu S, Yan H, et al. A review on the phytochemistry, pharmacology, pharmacokinetics and toxicology of geniposide, a natural product. Molecules 2017; 22(10): 1689.
[http://dx.doi.org/10.3390/molecules22101689] [PMID: 28994736]
[14]
Tan HY, Wang N, Tsao SW, Che CM, Yuen MF, Feng Y. IRE1α inhibition by natural compound genipin on tumour associated macrophages reduces growth of hepatocellular carcinoma. Oncotarget 2016; 7(28): 43792-804.
[http://dx.doi.org/10.18632/oncotarget.9696] [PMID: 27270308]
[15]
Wang N, Zhu M, Tsao SW, Man K, Zhang Z, Feng Y. Up-regulation of TIMP-1 by genipin inhibits MMP-2 activities and suppresses the metastatic potential of human hepatocellular carcinoma. PLoS One 2012; 7(9): e46318.
[http://dx.doi.org/10.1371/journal.pone.0046318] [PMID: 23029478]
[16]
Cao H, Feng Q, Xu W, et al. Genipin induced apoptosis associated with activation of the c-Jun NH2-terminal kinase and p53 protein in HeLa cells. Biol Pharm Bull 2010; 33(8): 1343-8.
[http://dx.doi.org/10.1248/bpb.33.1343] [PMID: 20686229]
[17]
Czarnecka AM, Synoradzki K, Firlej W, et al. Molecular biology of osteosarcoma. Cancers 2020; 12(8): 2130.
[http://dx.doi.org/10.3390/cancers12082130] [PMID: 32751922]
[18]
Liu Q, Wang Z, Zhou X, et al. miR-485-5p/HSP90 axis blocks Akt1 phosphorylation to suppress osteosarcoma cell proliferation and migration via PI3K/AKT pathway. J Physiol Biochem 2020; 76(2): 279-90.
[http://dx.doi.org/10.1007/s13105-020-00730-8] [PMID: 32100243]
[19]
Liu BO, Wu YI, Peng DAN. Astrocyte elevated gene-1 regulates osteosarcoma cell invasion and chemoresistance via endothelin-1/endothelin A receptor signaling. Oncol Lett 2013; 5(2): 505-10.
[http://dx.doi.org/10.3892/ol.2012.1056] [PMID: 23420812]
[20]
Durfee RA, Mohammed M, Luu HH. Review of osteosarcoma and current management. Rheumatol Ther 2016; 3(2): 221-43.
[http://dx.doi.org/10.1007/s40744-016-0046-y] [PMID: 27761754]
[21]
Strauss SJ, Whelan JS. Current questions in bone sarcomas. Curr Opin Oncol 2018; 30(4): 252-9.
[http://dx.doi.org/10.1097/CCO.0000000000000456] [PMID: 29782347]
[22]
Esmaeili J, Barati A, Ai J, Nooshabadi VT, Mirzaei Z. Employing hydrogels in tissue engineering approaches to boost conventional cancer-based research and therapies. RSC Advances 2021; 11(18): 10646-69.
[http://dx.doi.org/10.1039/D1RA00855B] [PMID: 35423538]
[23]
Was H, Borkowska A, Bagues A, et al. Mechanisms of chemotherapy-induced neurotoxicity. Front Pharmacol 2022; 13: 750507.
[http://dx.doi.org/10.3389/fphar.2022.750507]
[24]
Parekh HS, Liu G, Wei MQ. A new dawn for the use of traditional Chinese medicine in cancer therapy. Mol Cancer 2009; 8(1): 21.
[http://dx.doi.org/10.1186/1476-4598-8-21] [PMID: 19298677]
[25]
Wu B, Cui J, Zhang C, Li Z. A polysaccharide from Agaricus blazei inhibits proliferation and promotes apoptosis of osteosarcoma cells. Int J Biol Macromol 2012; 50(4): 1116-20.
[http://dx.doi.org/10.1016/j.ijbiomac.2012.02.023] [PMID: 22390851]
[26]
Zhang L, Wei W. Anti-inflammatory and immunoregulatory effects of paeoniflorin and total glucosides of paeony. Pharmacol Ther 2020; 207: 107452.
[http://dx.doi.org/10.1016/j.pharmthera.2019.107452] [PMID: 31836457]
[27]
Evan GI, Vousden KH. Proliferation, cell cycle and apoptosis in cancer. Nature 2001; 411(6835): 342-8.
[http://dx.doi.org/10.1038/35077213] [PMID: 11357141]
[28]
Pistritto G, Trisciuoglio D, Ceci C, Garufi A, D’Orazi G. Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging 2016; 8(4): 603-19.
[http://dx.doi.org/10.18632/aging.100934] [PMID: 27019364]
[29]
Gandour-Edwards R, Mack PC, deVere-White RW, Gumerlock PH. Abnormalities of apoptotic and cell cycle regulatory proteins in distinct histopathologic components of benign prostatic hyperplasia. Prostate Cancer Prostatic Dis 2004; 7(4): 321-6.
[http://dx.doi.org/10.1038/sj.pcan.4500749] [PMID: 15314639]
[30]
Peralta-Leal A, Rodríguez-Vargas JM, Aguilar-Quesada R, et al. PARP inhibitors: New partners in the therapy of cancer and inflammatory diseases. Free Radic Biol Med 2009; 47(1): 13-26.
[http://dx.doi.org/10.1016/j.freeradbiomed.2009.04.008] [PMID: 19362586]
[31]
Van Opdenbosch N, Lamkanfi M. Caspases in cell death, inflammation, and disease. Immunity 2019; 50(6): 1352-64.
[http://dx.doi.org/10.1016/j.immuni.2019.05.020] [PMID: 31216460]
[32]
Kale J, Osterlund EJ, Andrews DW. BCL-2 family proteins: Changing partners in the dance towards death. Cell Death Differ 2018; 25(1): 65-80.
[http://dx.doi.org/10.1038/cdd.2017.186] [PMID: 29149100]
[33]
Pastushenko I, Blanpain C. Transition states during tumor progression and metastasis. Trends Cell Biol 2019; 29: 212-26.
[34]
Serrano-Gomez SJ, Maziveyi M, Alahari SK. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol Cancer 2016; 15(1): 18.
[http://dx.doi.org/10.1186/s12943-016-0502-x] [PMID: 26905733]
[35]
Satelli A, Li S. Vimentin in cancer and its potential as a molecular target for cancer therapy. Cell Mol Life Sci 2011; 68(18): 3033-46.
[http://dx.doi.org/10.1007/s00018-011-0735-1] [PMID: 21637948]
[36]
Wang Y, Shi J, Chai K, Ying X, Zhou B. The role of snail in EMT and tumorigenesis. Curr Cancer Drug Targets 2013; 13(9): 963-72.
[http://dx.doi.org/10.2174/15680096113136660102] [PMID: 24168186]
[37]
Najafi M, Farhood B, Mortezaee K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J Cell Biochem 2019; 120(3): 2782-90.
[http://dx.doi.org/10.1002/jcb.27681] [PMID: 30321449]
[38]
Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell 2010; 141(1): 52-67.
[http://dx.doi.org/10.1016/j.cell.2010.03.015] [PMID: 20371345]
[39]
Kuijjer ML, van den Akker BEWM, Hilhorst R, et al. Kinome and mRNA expression profiling of high-grade osteosarcoma cell lines implies Akt signaling as possible target for therapy. BMC Med Genomics 2014; 7(1): 4.
[http://dx.doi.org/10.1186/1755-8794-7-4] [PMID: 24447333]
[40]
He JP, Hao Y, Wang XL, et al. Review of the molecular pathogenesis of osteosarcoma. Asian Pac J Cancer Prev 2014; 15(15): 5967-76.
[http://dx.doi.org/10.7314/APJCP.2014.15.15.5967] [PMID: 25124559]
[41]
Xu Z, Han X, Ou D, et al. Targeting PI3K/AKT/mTOR-mediated autophagy for tumor therapy. Appl Microbiol Biotechnol 2020; 104(2): 575-87.
[http://dx.doi.org/10.1007/s00253-019-10257-8] [PMID: 31832711]
[42]
Song M, Bode AM, Dong Z, Lee MH. AKT as a therapeutic target for cancer. Cancer Res 2019; 79(6): 1019-31.
[http://dx.doi.org/10.1158/0008-5472.CAN-18-2738] [PMID: 30808672]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy