The Multi-Faceted Effect of Curcumin in Glioblastoma from Rescuing Cell Clearance to Autophagy-Independent Effects
Abstract
:1. Introduction
2. Pleiotropic Effects of Curcumin in the CNS: from Neuroprotection to Anti-Cancer Activities
3. Curcumin Suppresses GSCs’ Tumorigenicity through ATG Induction
4. Effects of Curcumin on GBM beyond ATG Induction
4.1. Curcumin Suppresses GBM Cell Proliferation and Survival via Inhibition of NF-κB and AP-1 Pathways
4.2. Curcumin Induces GBM Cell Cycle Arrest through the Modulation TP53 and RB Pathways
4.3. Curcumin Hampers GBM Aggressiveness through the Modulation of The JAK/STAT Pathway
4.4. Curcumin Induces Pro-Apoptotic Pathways in GBM Cells
5. Concluding Remarks
Outstanding Questions
Author Contributions
Funding
Conflicts of Interest
References
- Prasad, S.; Aggarwal, B.B. Turmeric, the Golden Spice: From Traditional Medicine to Modern Medicine. In Herbal Medicine: Biomolecular and Clinical Aspects, 2nd ed.; Benzie, I.F.F., Wachtel-Galor, S., Eds.; CRC Press/Taylor & Francis: Boca Raton, FL, USA, 2011. [Google Scholar]
- Aggarwal, B.B.; Sundaram, C.; Malani, N.; Ichikawa, H. Curcumin: The Indian solid gold. Adv. Exp. Med. Biol. 2007, 595, 1–75. [Google Scholar]
- Ammon, H.P.; Wahl, M.A. Pharmacology of Curcuma longa. Planta Med. 1991, 57, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Limanaqi, F.; Biagioni, F.; Busceti, C.L.; Ryskalin, L.; Polzella, M.; Frati, A.; Fornai, F. Phytochemicals Bridging Autophagy Induction and Alpha-Synuclein Degradation in Parkinsonism. Int. J. Mol. Sci. 2019, 20, 3274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miriyala, S.; Panchatcharam, M.; Rengarajulu, P. Cardioprotective effects of curcumin. Adv. Exp. Med. Biol. 2007, 595, 359–377. [Google Scholar] [PubMed]
- Jagtap, S.; Meganathan, K.; Wagh, V.; Winkler, J.; Hescheler, J.; Sachinidis, A. Chemoprotective mechanism of the natural compounds, epigallocatechin-3-O-gallate, quercetin and curcumin against cancer and cardiovascular diseases. Curr. Med. Chem. 2009, 16, 1451–1462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Funk, J.L.; Oyarzo, J.N.; Frye, J.B.; Chen, G.; Lantz, R.C.; Jolad, S.D.; Solyom, A.M.; Timmermann, B.N. Turmeric extracts containing curcuminoids prevent experimental rheumatoid arthritis. J. Nat. Prod. 2006, 69, 351–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aggarwal, B.B.; Sung, B. Pharmacological basis for the role of curcumin in chronic diseases: An age-old spice with modern targets. Trends Pharmacol. Sci. 2009, 30, 85–94. [Google Scholar] [CrossRef] [PubMed]
- Alappat, L.; Awad, A.B. Curcumin and obesity: Evidence and mechanisms. Nutr. Rev. 2010, 68, 729–738. [Google Scholar] [CrossRef]
- Singh, R.; Sharma, P. Hepatoprotective Effect of Curcumin on Lindane-induced Oxidative Stress in Male Wistar Rats. Toxicol. Int. 2011, 18, 124–129. [Google Scholar]
- Pellegrini, C.; Fornai, M.; Antonioli, L.; Blandizzi, C.; Calderone, V. Phytochemicals as Novel Therapeutic Strategies for NLRP3 Inflammasome-Related Neurological, Metabolic, and Inflammatory Diseases. Int. J. Mol. Sci. 2019, 20, 2876. [Google Scholar] [CrossRef] [Green Version]
- Darvesh, A.S.; Carroll, R.T.; Bishayee, A.; Novotny, N.A.; Geldenhuys, W.J.; Van der Schyf, C.J. Curcumin and neurodegenerative diseases: A perspective. Expert Opin. Investig. Drugs 2012, 21, 1123–1140. [Google Scholar]
- Maiti, P.; Dunbar, G.L. Use of Curcumin, a Natural Polyphenol for Targeting Molecular Pathways in Treating Age-Related Neurodegenerative Diseases. Int. J. Mol. Sci. 2018, 19, 1637. [Google Scholar]
- Goel, A.; Kunnumakkara, A.B.; Aggarwal, B.B. Curcumin as “Curecumin”: From kitchen to clinic. Biochem. Pharmacol. 2008, 75, 787–809. [Google Scholar] [PubMed] [Green Version]
- Aggarwal, B.B.; Shishodia, S.; Takada, Y.; Banerjee, S.; Newman, R.A.; Bueso-Ramos, C.E.; Price, J.E. Curcumin suppresses the paclitaxel-induced nuclear factor-kappaB pathway in breast cancer cells and inhibits lung metastasis of human breast cancer in nude mice. Clin. Cancer Res. 2005, 11, 7490–7498. [Google Scholar] [PubMed] [Green Version]
- Li, M.; Zhang, Z.; Hill, D.L.; Wang, H.; Zhang, R. Curcumin, a dietary component, has anticancer, chemosensitization, and radiosensitization effects by down-regulating the MDM2 oncogene through the PI3K/mTOR/ETS2 pathway. Cancer Res. 2007, 67, 1988–1996. [Google Scholar] [PubMed] [Green Version]
- Kunnumakkara, A.B.; Guha, S.; Krishnan, S.; Diagaradjane, P.; Gelovani, J.; Aggarwal, B.B. Curcumin potentiates antitumor activity of gemcitabine in an orthotopic model of pancreatic cancer through suppression of proliferation, angiogenesis, and inhibition of nuclear factor-kappaB-regulated gene products. Cancer Res. 2007, 67, 3853–3861. [Google Scholar]
- Yin, H.; Zhou, Y.; Wen, C.; Zhou, C.; Zhang, W.; Hu, X.; Wang, L.; You, C.; Shao, J. Curcumin sensitizes glioblastoma to temozolomide by simultaneously generating ROS and disrupting AKT/mTOR signaling. Oncol. Rep. 2014, 32, 1610–1616. [Google Scholar] [PubMed]
- Qian, Y.; Ma, J.; Guo, X.; Sun, J.; Yu, Y.; Cao, B.; Zhang, L.; Ding, X.; Huang, J.; Shao, J.F. Curcumin enhances the radiosensitivity of U87 cells by inducing DUSP-2 up-regulation. Cell. Physiol. Biochem. 2015, 35, 1381–1393. [Google Scholar]
- Gupta, S.C.; Prasad, S.; Kim, J.H.; Patchva, S.; Webb, L.J.; Priyadarsini, I.K.; Aggarwal, B.B. Multitargeting by curcumin as revealed by molecular interaction studies. Nat. Prod. Rep. 2011, 28, 1937–1955. [Google Scholar]
- Priyadarsini, K.I. Chemical and structural features influencing the biological activity of curcumin. Curr. Pharm. Des. 2013, 19, 2093–2100. [Google Scholar]
- Ryskalin, L.; Limanaqi, F.; Biagioni, F.; Frati, A.; Esposito, V.; Calierno, M.T.; Lenzi, P.; Fornai, F. The emerging role of m-TOR up-regulation in brain Astrocytoma. Histol. Histopathol. 2017, 32, 413–431. [Google Scholar]
- Ryskalin, L.; Lazzeri, G.; Flaibani, M.; Biagioni, F.; Gambardella, S.; Frati, A.; Fornai, F. mTOR-Dependent Cell Proliferation in the Brain. Biomed Res. Int. 2017, 2017, 7082696. [Google Scholar] [PubMed]
- Lenzi, P.; Lazzeri, G.; Biagioni, F.; Busceti, C.L.; Gambardella, S.; Salvetti, A.; Fornai, F. The Autophagoproteasome a Novel Cell Clearing Organelle in Baseline and Stimulated Conditions. Front. Neuroanat. 2016, 10, 78. [Google Scholar]
- Lazzeri, G.; Biagioni, F.; Fulceri, F.; Busceti, C.L.; Scavuzzo, M.C.; Ippolito, C.; Salvetti, A.; Lenzi, P.; Fornai, F. mTOR Modulates Methamphetamine-Induced Toxicity through Cell Clearing Systems. Oxid. Med. Cell. Longev. 2018, 2018, 6124745. [Google Scholar] [PubMed] [Green Version]
- Zhuang, X.X.; Wang, S.F.; Tan, Y.; Song, J.X.; Zhu, Z.; Wang, Z.Y.; Wu, M.Y.; Cai, C.Z.; Huang, Z.J.; Tan, J.Q.; et al. Pharmacological enhancement of TFEB-mediated autophagy alleviated neuronal death in oxidative stress-induced Parkinson’s disease models. Cell Death Dis. 2020, 11, 128. [Google Scholar] [PubMed] [Green Version]
- Wang, W.; Xu, J. Curcumin Attenuates Cerebral Ischemia-reperfusion Injury Through Regulating Mitophagy and Preserving Mitochondrial Function. Curr. Neurovasc. Res. 2020, 17, 113–122. [Google Scholar] [PubMed]
- Toti, L.; Bartalucci, A.; Ferrucci, M.; Fulceri, F.; Lazzeri, G.; Lenzi, P.; Soldani, P.; Gobbi, P.; La Torre, A.; Gesi, M. High-intensity exercise training induces morphological and biochemical changes in skeletal muscles. Biol. Sport. 2013, 30, 301–309. [Google Scholar]
- Davinelli, S.; De Stefani, D.; De Vivo, I.; Scapagnini, G. Polyphenols as Caloric Restriction Mimetics Regulating Mitochondrial Biogenesis and Mitophagy. Trends Endocrinol. Metab. 2020, 31, 536–550. [Google Scholar]
- Drion, C.M.; van Scheppingen, J.; Arena, A.; Geijtenbeek, K.W.; Kooijman, L.; van Vliet, E.A.; Aronica, E.; Gorter, J.A. Effects of rapamycin and curcumin on inflammation and oxidative stress in vitro and in vivo—in search of potential anti-epileptogenic strategies for temporal lobe epilepsy. J. NeuroInflamm. 2018, 15, 212. [Google Scholar]
- Li, W.; Yao, S.; Li, H.; Meng, Z.; Sun, X. Curcumin promotes functional recovery and inhibits neuronal apoptosis after spinal cord injury through the modulation of autophagy. J. Spinal Cord Med. 2019, 1–9. [Google Scholar] [CrossRef]
- Lim, G.P.; Chu, T.; Yang, F.; Beech, W.; Frautschy, S.A.; Cole, G.M. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J. Neurosci. 2001, 21, 8370–8377. [Google Scholar] [PubMed]
- Lin, L.; Li, C.; Zhang, D.; Yuan, M.; Chen, C.H.; Li, M. Synergic Effects of Berberine and Curcumin on Improving Cognitive Function in an Alzheimer’s Disease Mouse Model. Neurochem. Res. 2020, 45, 1130–1141. [Google Scholar] [PubMed]
- Maiti, P.; Rossignol, J.; Dunbar, G.L. Curcumin Modulates Molecular Chaperones and Autophagy-Lysosomal Pathways In Vitro after Exposure to Aβ42. J. Alzheimers Dis. Parkinsonism. 2017, 7, 299. [Google Scholar]
- Sharma, N.; Nehru, B. Curcumin affords neuroprotection and inhibits α-synuclein aggregation in lipopolysaccharide-induced Parkinson’s disease model. Inflammopharmacology 2018, 26, 349–360. [Google Scholar]
- Jiang, T.F.; Zhang, Y.J.; Zhou, H.Y.; Wang, H.M.; Tian, L.P.; Liu, J.; Ding, J.Q.; Chen, S.D. Curcumin ameliorates the neurodegenerative pathology in A53T α-synuclein cell model of Parkinson’s disease through the downregulation of mTOR/p70S6K signaling and the recovery of macroautophagy. J. Neuroimmune Pharmacol. 2013, 8, 356–369. [Google Scholar] [PubMed]
- Lin, C.F.; Yu, K.H.; Jheng, C.P.; Chung, R.; Lee, C.I. Curcumin reduces amyloid fibrillation of prion protein and decreases reactive oxidative stress. Pathogens 2013, 2, 506–519. [Google Scholar]
- Ferrucci, M.; Ryskalin, L.; Biagioni, F.; Gambardella, S.; Busceti, C.L.; Falleni, A.; Lazzeri, G.; Fornai, F. Methamphetamine increases Prion Protein and induces dopamine-dependent expression of protease resistant PrPsc. Arch. Ital. Biol. 2017, 155, 81–97. [Google Scholar]
- Ryskalin, L.; Busceti, C.L.; Biagioni, F.; Limanaqi, F.; Familiari, P.; Frati, A.; Fornai, F. Prion Protein in Glioblastoma Multiforme. Int. J. Mol. Sci. 2019, 20, 5107. [Google Scholar]
- Louis, N.; Perry, A.; Reifenberge, R.G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar]
- Ostrom, Q.T.; Gittleman, H.; Farah, P.; Ondracek, A.; Chen, Y.; Wolinsky, Y.; Stroup, N.E.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006–2010. Neuro Oncol. 2013, 15, ii1–ii56. [Google Scholar]
- Koshy, M.; Villano, J.L.; Dolecek, T.A.; Howard, A.; Mahmood, U.; Chmura, S.J.; Weichselbaum, R.R.; McCarthy, B.J. Improved survival time trends of glioblastoma using the SEER 17 population-based registries. Neuro Oncol. 2012, 107, 207–212. [Google Scholar]
- Van Meir, E.G.; Hadjipanayis, C.G.; Norden, A.D.; Shu, H.K.; Wen, P.Y.; Olson, J.J. Exciting new advances in neuro-oncology: The avenue to a cure for malignant glioma. CA Cancer J. Clin. 2010, 60, 166–193. [Google Scholar] [CrossRef] [PubMed]
- Mao, H.; Lebrun, D.G.; Yang, J.; Zhu, V.F.; Li, M. Deregulated signaling pathways in glioblastoma multiforme: Molecular mechanisms and therapeutic targets. Cancer Invest. 2012, 30, 48–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dhandapani, K.; Mahesh, V.; Brann, D. Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFkappaB transcription factors. J. Neurochem. 2007, 102, 522–538. [Google Scholar]
- Huang, T.; Tsai, T.; Hsu, C.; Hsu, Y. Curcuminoids suppress the growth and induce apoptosis through caspase-3-dependent pathways in glioblastoma multiforme (GBM) 8401 cells. J. Agric. Food Chem. 2010, 58, 10639–10645. [Google Scholar] [CrossRef]
- Liu, E.; Wu, J.; Cao, W.; Zhang, J.; Liu, W.; Jiang, X.; Zhang, X. Curcumin induces G2/M cell cycle arrest in a p53-dependent manner and upregulates ING4 expression in human glioma. J. Neurooncol. 2007, 85, 263–270. [Google Scholar] [CrossRef]
- Senft, C.; Polacin, M.; Priester, M.; Seifert, V.; Kogel, D.; Weissenberger, J. The nontoxic natural compound Curcumin exerts anti-proliferative, anti-migratory, and anti-invasive properties against malignant gliomas. BMC Cancer 2010, 10, 491. [Google Scholar] [CrossRef] [Green Version]
- Zanotto-Filho, A.; Braganhol, E.; Edelweiss, M.I.; Behr, G.A.; Zanin, R.; Schröder, R.; Simões-Pires, A.; Battastini, A.M.; Moreira, J.C. The curry spice curcumin selectively inhibits cancer cells growth in vitro and in preclinical model of glioblastoma. J. Nutr. Biochem. 2012, 23, 591–601. [Google Scholar] [CrossRef]
- Karmakar, S.; Banik, N.L.; Ray, S.K. Curcumin suppressed anti-apoptotic signals and activated cysteine proteases for apoptosis in human malignant glioblastoma U87MG cells. Neurochem. Res. 2007, 32, 2103–2113. [Google Scholar] [CrossRef]
- Gersey, Z.C.; Rodriguez, G.A.; Barbarite, E.; Sanchez, A.; Walters, W.M.; Ohaeto, K.C.; Komotar, R.J.; Graham, R.M. Curcumin decreases malignant characteristics of glioblastoma stem cells via induction of reactive oxygen species. BMC Cancer 2017, 17, 99. [Google Scholar] [CrossRef] [Green Version]
- Zhuang, W.; Long, L.; Zheng, B.; Ji, W.; Yang, N.; Zhang, Q.; Liang, Z. Curcumin promotes differentiation of glioma-initiating cells by inducing autophagy. Cancer Sci. 2012, 103, 684–690. [Google Scholar] [CrossRef] [PubMed]
- Shakeri, A.; Cicero, A.F.G.; Panahi, Y.; Mohajeri, M.; Sahebkar, A. Curcumin: A naturally occurring autophagy modulator. J. Cell. Physiol. 2019, 234, 5643–5654. [Google Scholar] [CrossRef] [PubMed]
- Shinojima, N.; Yokoyama, T.; Kondo, Y.; Kondo, S. Roles of the Akt/mTOR/p70S6K and ERK1/2 signaling pathways in curcumin-induced autophagy. Autophagy 2007, 3, 635–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sahab-Negah, S.; Ariakia, F.; Jalili-Nik, M.; Afshari, A.R.; Salehi, S.; Samini, F.; Rajabzadeh, G.; Gorji, A. Curcumin Loaded in Niosomal Nanoparticles Improved the Anti-tumor Effects of Free Curcumin on Glioblastoma Stem-like Cells: An In Vitro Study. Mol. Neurobiol. 2020, 57, 3391–3411. [Google Scholar] [CrossRef]
- Maiti, P.; Scott, J.; Sengupta, D.; Al-Gharaibeh, A.; Dunbar, G.L. Curcumin and Solid Lipid Curcumin Particles Induce Autophagy, but Inhibit Mitophagy and the PI3K-Akt/mTOR Pathway in Cultured Glioblastoma Cells. Int. J. Mol. Sci. 2019, 20, 399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, L.; Wu, Q.; Guryanova, O.A.; Huang, Z.; Huang, Q.; Rich, J.N.; Bao, S. Elevated invasive potential of glioblastoma stem cells. Biochem. Biophys. Res. Commun. 2011, 406, 643–648. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Liu, F.; Liao, W.; Yu, L.; Hu, Z.; Li, M.; Xia, H. Curcumin suppresses glioblastoma cell proliferation by p-AKT/mTOR pathway and increases the PTEN expression. Arch Biochem. Biophys. 2020, 689, 108412. [Google Scholar] [CrossRef]
- Du, W.Z.; Feng, Y.; Wang, X.F.; Piao, X.Y.; Cui, Y.Q.; Chen, L.C.; Lei, X.H.; Sun, X.; Liu, X.; Wang, H.B.; et al. Curcumin suppresses malignant glioma cells growth and induces apoptosis by inhibition of SHH/GLI1 signaling pathway in vitro and vivo. CNS Neurosci. Ther. 2013, 19, 926–936. [Google Scholar]
- Perry, M.C.; Demeule, M.; Régina, A.; Moumdjian, R.; Béliveau, R. Curcumin inhibits tumor growth and angiogenesis in glioblastoma xenografts. Mol. Nutr. Food Res. 2010, 54, 1192–1201. [Google Scholar] [CrossRef]
- Park, K.S.; Yoon, S.Y.; Park, S.H.; Hwang, J.H. Anti-migration and anti-invasion effects of curcumin via suppression of Fascin expression in glioblastoma cells. Brain Tumor Res. Treat. 2019, 7, 16–24. [Google Scholar] [CrossRef]
- Shi, L.; Sun, G. Low-dose DMC significantly enhances the effect of TMZ on glioma cells by targeting multiple signaling pathways both in vivo and in vitro. Neuromol. Med. 2015, 17, 431–442. [Google Scholar] [CrossRef] [PubMed]
- Leng, L.; Zhong, X.; Sun, G.; Qiu, W.; Shi, L. Demethoxycurcumin was superior to temozolomide in the inhibition of the growth of glioblastoma stem cells in vivo. Tumour Biol. 2016. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Zhu, Y.; Sun, X.; He, X.; Wang, M.; Wang, Z.; Wang, Q.; Zhu, R.; Wang, S. Curcumin-Loaded Layered Double Hydroxide Nanoparticles-Induced Autophagy for Reducing Glioma Cell Migration and Invasion. J. Biomed. Nanotechnol. 2016, 12, 2051–2062. [Google Scholar] [CrossRef] [PubMed]
- Shin, H.J.; Lee, S.; Jung, H.J. A curcumin derivative hydrazinobenzoylcurcumin suppresses stem-like features of glioblastoma cells by targeting Ca2+/calmodulin-dependent protein kinase II. J. Cell. Biochem. 2019, 120, 6741–6752. [Google Scholar] [CrossRef]
- Sansalone, L.; Veliz, E.A.; Myrthil, N.G.; Stathias, V.; Walters, W.; Torrens, I.I.; Schürer, S.C.; Vanni, S.; Leblanc, R.M.; Graham, R.M. Novel Curcumin Inspired Bis-Chalcone Promotes Endoplasmic Reticulum Stress and Glioblastoma Neurosphere Cell Death. Cancers 2019, 11, 357. [Google Scholar] [CrossRef] [Green Version]
- Corsaro, A.; Bajetto, A.; Thellung, S.; Begani, G.; Villa, V.; Nizzari, M.; Pattarozzi, A.; Solari, A.; Gatti, M.; Pagano, A.; et al. Cellular prion protein controls stem cell-like properties of human glioblastoma tumor-initiating cells. Oncotarget 2016, 7, 38638–38657. [Google Scholar] [CrossRef]
- Qazi, M.A.; Vora, P.; Venugopal, C.; Sidhu, S.S.; Moffat, J.; Swanton, C.; Singh, S.K. Intratumoral heterogeneity: Pathways to treatment resistance and relapse in human glioblastoma. Ann. Oncol. 2017, 28, 1448–1456. [Google Scholar] [CrossRef]
- Ryskalin, L.; Gaglione, A.; Limanaqi, F.; Biagioni, F.; Familiari, P.; Frati, A.; Esposito, V.; Fornai, F. The Autophagy Status of Cancer Stem Cells in Gliobastoma Multiforme: From Cancer Promotion to Therapeutic Strategies. Int. J. Mol. Sci. 2019, 20, 3824. [Google Scholar] [CrossRef] [Green Version]
- Huang, X.; Bai, H.M.; Chen, L.; Li, B.; Lu, Y.C. Reduced expression of LC3B-II and Beclin 1 in glioblastoma multiforme indicates a down-regulated autophagic capacity that relates to the progression of astrocytic tumors. J. Clin. Neurosci. 2010, 17, 1515–1519. [Google Scholar] [CrossRef]
- Li, X.Y.; Zhang, L.Q.; Zhang, X.G.; Li, X.; Ren, Y.B.; Ma, X.Y.; Li, X.G.; Wang, L.X. Association between AKT/mTOR signalling pathway and malignancy grade of human gliomas. J. Neurooncol. 2011, 103, 453–458. [Google Scholar] [CrossRef]
- Arcella, F.; Biagioni, M.; Oliva, A.; Bucci, D.; Frati, A.; Esposito, V.; Cantore, G.; Giangaspero, F.; Fornai, F. Rapamycin inhibits the growth of glioblastoma. Brain Res. 2013, 1495, 37–51. [Google Scholar] [CrossRef] [PubMed]
- Shukla, S.; Patric, I.R.; Patil, V.; Shwetha, S.D.; Hegde, A.S.; Chandramouli, B.A.; Arivazhagan, A.; Santosh, V.; Somasundaram, K. Methylation silencing of ULK2, an autophagy gene, is essential for astrocyte transformation and tumor growth. J. Biol. Chem. 2014, 289, 22306–22318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iwamaru, A.; Kondo, Y.; Iwado, E.; Aoki, H.; Fujiwara, K.; Yokoyama, T.; Mills, G.B.; Kondo, S. Silencing mammalian target of rapamycin signaling by small interfering RNA enhances rapamycin-induced autophagy in malignant glioma cells. Oncogene 2007, 26, 1840–1851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, H.; White, E.J.; Conrad, C.; Gomez-Manzano, C.; Fueyo, J. Autophagy pathways in glioblastoma. Methods Enzymol. 2009, 453, 273–286. [Google Scholar]
- Zhao, Y.; Huang, Q.; Yang, J.; Lou, M.; Wang, A.; Dong, J.; Qin, Z.; Zhang, T. Autophagy impairment inhibits differentiation of glioma stem/progenitor cells. Brain Res. 2010, 1313, 250–258. [Google Scholar] [CrossRef]
- Ferrucci, M.; Biagioni, F.; Lenzi, P.; Gambardella, S.; Ferese, R.; Calierno, M.T.; Falleni, A.; Grimaldi, A.; Frati, A.; Esposito, V.; et al. Rapamycin promotes differentiation increasing βIII-tubulin, NeuN, and NeuroD while suppressing nestin expression in glioblastoma cells. Oncotarget 2017, 8, 29574–29599. [Google Scholar] [CrossRef] [Green Version]
- Jiang, H.; Gomez-Manzano, C.; Aoki, H.; Alonso, M.M.; Kondo, S.; McCormick, F.; Xu, J.; Kondo, Y.; Bekele, B.N.; Colman, H.; et al. Examination of the therapeutic potential of Delta-24-RGD in brain tumor stem cells: Role of autophagic cell death. J. Natl. Cancer Inst. 2007, 99, 1410–1414. [Google Scholar] [CrossRef] [Green Version]
- Liu, R.; Li, J.; Zhang, T.; Zou, L.; Chen, Y.; Wang, K.; Lei, Y.; Yuan, K.; Li, Y.; Lan, J.; et al. Itraconazole suppresses the growth of glioblastoma through induction of autophagy: Involvement of abnormal cholesterol trafficking. Autophagy 2014, 10, 1241–1255. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.E.; Yoon, S.S.; Moon, E.Y. Curcumin-Induced Autophagy Augments Its Antitumor Effect against A172 Human Glioblastoma Cells. Biomol. Ther. (Seoul) 2019, 27, 484–491. [Google Scholar] [CrossRef]
- Aoki, H.; Takada, Y.; Kondo, S.; Sawaya, R.; Aggarwal, B.B.; Kondo, Y. Evidence that curcumin suppresses the growth of malignant gliomas in vitro and in vivo through induction of autophagy: Role of Akt and extracellular signal-regulated kinase signaling pathways. Mol. Pharmacol. 2007, 72, 29–39. [Google Scholar] [CrossRef] [Green Version]
- Zhuang, W.; Li, B.; Long, L.; Chen, L.; Huang, Q.; Liang, Z. Induction of autophagy promotes differentiation of glioma-initiating cells and their radiosensitivity. Int. J. Cancer 2011, 129, 2720–2731. [Google Scholar] [CrossRef] [PubMed]
- Bharti, A.C.; Donato, N.; Singh, S.; Aggarwal, B.B. Curcumin (diferuloylmethane) down-regulates the constitutive activation of nuclear factor-kappa B and IkappaBalpha kinase in human multiple myeloma cells, leading to suppression of proliferation and induction of apoptosis. Blood 2003, 101, 1053–1062. [Google Scholar] [CrossRef] [PubMed]
- Xiao, K.; Jiang, J.; Guan, C.; Dong, C.; Wang, G.; Bai, L.; Sun, J.; Hu, C.; Bai, C. Curcumin induces autophagy via activating the AMPK signaling pathway in lung adenocarcinoma cells. J. Pharmacol. Sci. 2013, 123, 102–109. [Google Scholar] [CrossRef] [Green Version]
- Su, J.; Zhou, X.; Yin, X.; Wang, L.; Zhao, Z.; Hou, Y.; Zheng, N.; Xia, J.; Wang, Z. The effects of curcumin on proliferation, apoptosis, invasion, and NEDD4 expression in pancreatic cancer. Biochem. Pharmacol. 2017, 140, 28–40. [Google Scholar] [CrossRef]
- Beevers, C.S.; Chen, L.; Liu, L.; Luo, Y.; Webster, N.J.; Huang, S. Curcumin disrupts the Mammalian target of rapamycin-raptor complex. Cancer Res. 2009, 69, 1000–1008. [Google Scholar] [CrossRef] [Green Version]
- Fong, D.; Yeh, A.; Naftalovich, R.; Choi, T.H.; Chan, M.M. Curcumin inhibits the side population (SP) phenotype of the rat C6 glioma cell line: Towards targeting of cancer stem cells with phytochemicals. Cancer Lett. 2010, 293, 65–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Ying, X.; Xu, H.; Yan, H.; Li, X.; Tang, H. The functional curcumin liposomes induce apoptosis in C6 glioblastoma cells and C6 glioblastoma stem cells in vitro and in animals. Int. J. Nanomed. 2017, 12, 1369–1384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zanotto-Filho, A.; Coradini, K.; Braganhol, E.; Schroder, R.; de Oliveira, C.M.; Simoes-Pires, A.; Battastini, A.M.O.; Pohlmann, A.R.; Guterres, S.S.; Forcelini, C.M.; et al. Curcumin-loaded lipid-core nanocapsules as a strategy to improve pharmacological efficacy of curcumin in glioma treatment. Eur. J. Pharm. Biopharm. 2013, 83, 156–167. [Google Scholar] [CrossRef]
- Tao, Z.; Li, T.; Ma, H.; Yang, Y.; Zhang, C.; Hai, L.; Liu, P.; Yuan, F.; Li, J.; Yi, L.; et al. Autophagy suppresses self-renewal ability and tumorigenicity of glioma-initiating cells and promotes Notch1 degradation. Cell Death Dis. 2018, 9, 1063. [Google Scholar] [CrossRef]
- Wang, X.; Deng, J.; Yuan, J.; Tang, X.; Wang, Y.; Chen, H.; Liu, Y.; Zhou, L. Curcumin exerts its tumor suppressive function via inhibition of NEDD4 oncoprotein in glioma cancer cells. Int. J. Oncol. 2017, 51, 467–477. [Google Scholar] [CrossRef]
- Wang, X.; Trotman, L.C.; Koppie, T.; Alimonti, A.; Chen, Z.; Gao, Z.; Wang, J.; Erdjument-Bromage, H.; Tempst, P.; Cordon-Cardo, C.; et al. NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 2007, 128, 129–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, L.; Fei, X.; Wang, Z. Demethoxycurcumin was prior to temozolomide on inhibiting proliferation and induced apoptosis of glioblastoma stem cells. Tumour Biol. 2015, 36, 7107–7119. [Google Scholar] [CrossRef] [PubMed]
- Lim, K.J.; Bisht, S.; Bar, E.E.; Maitra, A.; Eberhart, C.G. A polymeric nanoparticle formulation of curcumin inhibits growth, clonogenicity and stem-like fraction in malignant brain tumors. Cancer Biol. Ther. 2011, 11, 464–473. [Google Scholar] [CrossRef] [Green Version]
- Zhuang, W.; Qin, Z.; Liang, Z. The role of autophagy in sensitizing malignant glioma cells to radiation therapy. Acta Biochim. Biophys. Sin. 2009, 41, 341–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, H.; Chen, L.; Li, J.J.; Zhou, Q.; Huang, A.; Liu, W.W.; Wang, K.; Gao, L.; Qi, S.T.; Lu, Y.T. miR-519a enhances chemosensitivity and promotes autophagy in glioblastoma by targeting STAT3/Bcl2 signaling pathway. J. Hematol. Oncol. 2018, 11, 70. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Peng, L.; Liu, A.; Ji, J.; Zhao, L.; Zhai, G. The enhanced effect of tetrahydrocurcumin on radiosensitivity of glioma cells. J. Pharm. Pharmacol. 2018, 70, 749–759. [Google Scholar] [CrossRef]
- Castonguay, A.; Doucet, C.; Juhas, M.; Maysinger, D. New ruthenium(II)-letrozole complexes as anticancer therapeutics. J. Med. Chem. 2012, 55, 8799–8806. [Google Scholar] [CrossRef] [Green Version]
- Ramachandran, C.; Nair, S.M.; Escalon, E.; Melnick, S.J. Potentiation of etoposide and temozolomide cytotoxicity by curcumin and turmeric force™ in brain tumor cell lines. J. Complement. Integr. Med. 2012, 9, 20. [Google Scholar] [CrossRef]
- Fratantonio, D.; Molonia, M.S.; Bashllari, R.; Muscara, C.; Ferlazzo, G.; Costa, G.; Saija, A.; Cimino, F.; Speciale, A. Curcumin potentiates the antitumor activity of Paclitaxel in rat glioma C6 cells. Phytomedicine 2019, 55, 23–30. [Google Scholar] [CrossRef]
- Zhao, J.; Zhu, J.; Lv, X.; Xing, J.; Liu, S.; Chen, C.; Xu, Y. Curcumin potentiates the potent antitumor activity of ACNU against glioblastoma by suppressing the PI3K/AKT and NF-kappaB/COX-2 signaling pathways. OncoTargets Ther. 2017, 10, 5471–5482. [Google Scholar] [CrossRef] [Green Version]
- Galan-Moya, E.M.; Le Guelte, A.; Lima Fernandes, E.; Thirant, C.; Dwyer, J.; Bidere, N.; Couraud, P.O.; Scott, M.G.; Junier, M.P.; Chneiweiss, H.; et al. Secreted factors from brain endothelial cells maintain glioblastoma stem-like cell expansion through the mTOR pathway. EMBO Rep. 2011, 12, 470–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryskalin, L.; Biagioni, F.; Lenzi, P.; Frati, A.; Fornai, F. mTOR Modulates Intercellular Signals for Enlargement and Infiltration in Glioblastoma Multiforme. Cancers 2020, 12, 2486. [Google Scholar] [CrossRef] [PubMed]
- Poehler, A.-M.; Xiang, W.; Spitzer, P.; May, V.E.L.; Meixner, H.; Rockenstein, E.; Chutna, O.; Outeiro, T.F.; Winkler, J.; Masliah, E.; et al. Autophagy modulates SNCA/α-synuclein release, thereby generating a hostile microenvironment. Autophagy 2014, 10, 2171–2192. [Google Scholar] [CrossRef] [Green Version]
- Xu, J.; Camfield, R.; Gorski, S.M. The interplay between exosomes and autophagy-partners in crime. J. Cell Sci. 2018, 131, jcs215210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oshima, M.; Seki, T.; Kurauchi, Y.; Hisatsune, A.; Katsuki, H. Reciprocal Regulation of Chaperone-Mediated Autophagy/Microautophagy and Exosome Release. Biol. Pharm. Bull. 2019, 42, 1394–1401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skog, J.; Wurdinger, T.; van Rijn, S.; Meijer, D.H.; Gainche, L.; Sena-Esteves, M.; Curry, W.T., Jr.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef] [PubMed]
- Nakano, I.; Garnier, D.; Minata, M.; Rak, J. Extracellular vesicles in the biology of brain tumour stem cells--Implications for inter-cellular communication, therapy and biomarker development. Semin. Cell Dev. Biol. 2015, 40, 17–26. [Google Scholar] [CrossRef]
- De Vrij, J.; Maas, S.L.; Kwappenberg, K.M.; Schnoor, R.; Kleijn, A.; Dekker, L.; Luider, T.M.; de Witte, L.D.; Litjens, M.; van Strien, M.E.; et al. Glioblastoma-derived extracellular vesicles modify the phenotype of monocytic cells. Int. J. Cancer 2015, 137, 1630–1642. [Google Scholar] [CrossRef] [Green Version]
- Gabrusiewicz, K.; Li, X.; Wei, J.; Hashimoto, Y.; Marisetty, A.L.; Ott, M.; Wang, F.; Hawke, D.; Yu, J.; Healy, L.M.; et al. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology 2018, 7, e1412909. [Google Scholar] [CrossRef]
- Mukherjee, S.; Baidoo, J.N.E.; Sampat, S.; Mancuso, A.; David, L.; Cohen, L.S.; Zhou, S.; Banerjee, P. Liposomal TriCurin, A Synergistic Combination of Curcumin, Epicatechin Gallate and Resveratrol, Repolarizes Tumor-Associated Microglia/Macrophages, and Eliminates Glioblastoma (GBM) and GBM Stem Cells. Molecules 2018, 23, 201. [Google Scholar] [CrossRef] [Green Version]
- Aggarwal, B.B.; Kumar, A.; Bharti, A. Anticancer potential of curcumin: Preclinical and clinical studies. Anticancer Res. 2003, 23, 363–398. [Google Scholar] [PubMed]
- Su, C.; Wang, M.; Chiu, T. The anti-cancer efficacy of curcumin scrutinized through core signaling pathways in glioblastoma. Int. J. Mol. Med. 2010, 26, 217–224. [Google Scholar]
- Chen, Q.G.; Zhou, W.; Han, T.; Du, S.Q.; Li, Z.H.; Zhang, Z.; Shan, G.Y.; Kong, C.Z. MiR-378 suppresses prostate cancer cell growth through downregulation of MAPK1 in vitro and in vivo. Tumour Biol. 2016, 37, 2095–2103. [Google Scholar] [PubMed]
- Kuo, C.L.; Wu, S.Y.; Ip, S.W.; Wu, P.P.; Yu, C.S.; Yang, J.S.; Chen, P.Y.; Wu, S.H.; Chung, J.G. Apoptotic death in curcumin-treated NPC-TW human nasopharyngeal carcinoma cells is mediated through the ROS, mitochondrial depolarization and caspase-3-dependent signaling responses. Int. J. Oncol. 2011, 39, 319–328. [Google Scholar]
- Liontas, A.; Yeger, H. Curcumin and resveratrol induce apoptosis and nuclear translocation and activation of p53 in human neuroblastoma. Anticancer Res. 2004, 24, 987–998. [Google Scholar] [PubMed]
- Choi, B.; Kim, C.; Bae, Y.; Lim, Y.; Lee, Y.; Shin, S. p21 Waf1/Cip1 expression by curcumin in U-87MG human glioma cells: Role of early growth response-1 expression. Cancer Res. 2008, 68, 1369–1377. [Google Scholar]
- Oeckinghaus, A.; Ghosh, S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb. Perspect. Biol. 2009, 1, a000034. [Google Scholar]
- Wang, H.; Wang, H.; Zhang, W.; Huang, H.J.; Liao, W.S.; Fuller, G.N. Analysis of the activation status of Akt, NFkappaB, and Stat3 in human diffuse gliomas. Lab Invest. 2004, 84, 941–951. [Google Scholar]
- Dan, H.C.; Cooper, M.J.; Cogswell, P.C.; Duncan, J.A.; Ting, J.P.; Baldwin, A.S. Akt-dependent regulation of NF-{kappa}B is controlled by mTOR and Raptor in association with IKK. Genes Dev. 2008, 22, 1490–1500. [Google Scholar]
- Puliyappadamba, V.T.; Hatanpaa, K.J.; Chakraborty, S.; Habib, A.A. The role of NF-κB in the pathogenesis of glioma. Mol. Cell. Oncol. 2014, 1, e963478. [Google Scholar]
- Kim, S.Y.; Jung, S.H.; Kim, H.S. Curcumin is a potent broad spectrum inhibitor of matrix metalloproteinase gene expression in human astroglioma cells. Biochem. Biophys. Res. Commun. 2005, 337, 510–516. [Google Scholar]
- Kumar, R.; Lal, N.; Nemaysh, V.; Luthra, P.M. Demethoxycurcumin mediated targeting of MnSOD leading to activation of apoptotic pathway and inhibition of Akt/NF-kappaB survival signalling in human glioma U87 MG cells. Toxicol. Appl. Pharmacol. 2018, 345, 75–93. [Google Scholar]
- Hesari, A.; Rezaei, M.; Rezaei, M.; Dashtiahangar, M.; Fathi, M.; Rad, J.G.; Momeni, F.; Avan, A.; Ghasemi, F. Effect of curcumin on glioblastoma cells. J. Cell. Physiol. 2019, 234, 10281–10288. [Google Scholar] [CrossRef] [PubMed]
- Woo, M.S.; Jung, S.H.; Kim, S.Y.; Hyun, J.W.; Ko, K.H.; Kim, W.K.; Kim, H.S. Curcumin suppresses phorbol ester-induced matrix metalloproteinase-9 expression by inhibiting the PKC to MAPK signaling pathways in human astroglioma cells. Biochem. Biophys. Res. Commun. 2005, 335, 1017–1025. [Google Scholar] [PubMed]
- Luthra, P.; Kumar, R.; Prakash, A. Demethoxycurcumin induces Bcl-2 mediated G2/M arrest and apoptosis in human glioma U87 cells. Biochem. Biophys. Res. Commun. 2009, 384, 420–425. [Google Scholar]
- Wu, B.; Yao, H.; Wang, S.; Xu, R. DAPK1 modulates a curcumin-induced G2/M arrest and apoptosis by regulating STAT3, NF-κB, and caspase-3 activation. Biochem. Biophys. Res. Commun. 2013, 434, 75–80. [Google Scholar]
- Weissenberger, J.; Priester, M.; Bernreuther, C.; Rakel, S.; Glatzel, M.; Seifert, V.; Kogel, D. Dietary curcumin attenuates glioma growth in a syngeneic mouse model by inhibition of the JAK1,2/STAT3 signaling pathway. Clin. Cancer Res. 2010, 16, 5781–5795. [Google Scholar]
- Nagai, S.; Kurimoto, M.; Washiyama, K.; Hirashima, Y.; Kumanishi, T.; Endo, S. Inhibition of cellular proliferation and induction of apoptosis by curcumin in human malignant astrocytoma cell lines. J. Neurooncol. 2005, 74, 105–111. [Google Scholar]
- Khaw, A.K.; Hande, M.P.; Kalthur, G.; Hande, M.P. Curcumin inhibits telomerase and induces telomere shortening and apoptosis in brain tumor cells. J. Cell. Biochem. 2013, 114, 1257–1270. [Google Scholar]
- Zhang, Y.; Tu, L.; Zhou, X.; Li, B. Curcumin-Mediated Induction of Apoptosis in Human Glioma CHME Cells. Med. Sci. Monit. Basic Res. 2018, 24, 216–224. [Google Scholar] [PubMed]
- Prasad, S.; Tyagi, A.K.; Aggarwal, B.B. Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: The golden pigment from golden spice. Cancer Res. Treat. 2014, 46, 2–18. [Google Scholar] [CrossRef] [Green Version]
- Dei Cas, M.; Ghidoni, R. Dietary Curcumin: Correlation between Bioavailability and Health Potential. Nutrients 2019, 11, 2147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsai, Y.-M.; Chien, C.-F.; Lin, L.-C.; Tsai, T.-H. Curcumin and its nano-formulation: the kinetics of tissue distribution and blood-brain barrier penetration. Int. J. Pharm. 2011, 416, 331–338. [Google Scholar] [CrossRef] [PubMed]
- Zhao, M.; Zhao, M.; Fu, C.; Yu, Y.; Fu, A. Targeted therapy of intracranial glioma model mice with curcumin nanoliposomes. Int. J. Nanomed. 2018, 13, 1601–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dutzmann, S.; Schiborr, C.; Kocher, A.; Pilatus, U.; Hattingen, E.; Weissenberger, J.; Gessler, F.; Quick-Weller, J.; Franz, K.; Seifert, V.; et al. Intratumoral concentrations and effects of orally administered micellar curcuminoids in glioblastoma patients. Nutr. Cancer 2016, 68, 943–948. [Google Scholar] [CrossRef] [PubMed]
- Jayaprakasha, G.; Rao, L.J.; Sakariah, K. Antioxidant activities of curcumin, demethoxycurcumin and bisdemethoxycurcumin. Food Chem. 2006, 98, 720–724. [Google Scholar] [CrossRef]
- Huang, T.Y.; Hsu, C.W.; Chang, W.C.; Wang, M.Y.; Wu, J.F.; Hsu, Y.C. Demethoxycurcumin Retards Cell Growth and Induces Apoptosis in Human Brain Malignant Glioma GBM 8401 Cells. Evid. Based Complement Altern. Med. 2012, 2012, 396573. [Google Scholar] [CrossRef] [Green Version]
Cell Line (s) | Dose (s) | Molecular Target (s) | Effect (s) | Reference |
---|---|---|---|---|
T98G *, U87MG *, T67 *, C6 ‡ | 25–50 μmol/L | AP-1, NFκB | Reduces cell survival; suppresses chemotherapy resistance | [44] |
U251 * | 10 μM | p53, ING4, p21 WAF-1/CIP-1 | Inhibits cell growth, induces G2/M cell cycle arrest | [46] |
A172 *, MZ-18 *, MZ-54 §, MZ-256 §, MZ-304 § | 10 μM, 20 μM and 50 μM | JAK/STAT3 | Inhibits cell proliferation, migration, and invasion | [47] |
U251 *, SNB19 * | 10 μM, 15 μM | pAkt, p57, Skp2 | Inhibits cell proliferation, migration, and invasion, induces cell cycle arrest and apoptosis | [48] |
U87MG* | 25 μM, 50 μM | NFκB, IAPs, Smac/Diablo, Bax, Bcl-2, caspase-3 | Decreases cell viability and induces apoptosis | [49] |
U87 *, U251 * | 2.5 μM (IC50 25 μM) | STAT3, MAPK IAP, ROS, | Decreases cell viability, inhibits proliferation, sphere-forming ability, and colony-forming potential of glioblastoma stem cells | [50] |
SU-2 *, SU-3 * | 2 μM | GFAP, Tuj1, Olig2, βIIItubulin, LC3 | Induces ATG and differentiation, while inhibiting self-renewal in glioma-initiating cells (GICs) | [51] |
U87 * | 10 µM/L, 20 µM/L | STAT3 | Inhibits cell migration and invasion | [52] |
A172 * | 10 μM | Atg5, Beclin-1 | Induces ATG | [53] |
U87-MG *, U373-MG * | 20 μM, 40 μM | Akt/mTOR/p70S6K, ERK1/2 | Induces G2/M arrest, inhibits cell growth, induces ATG | [54] |
U-87MG *, GL261 †, F98 ‡, C6 ‡ | 25 μM | Atg5, Atg7, Beclin-1, LC3A/B, p62, PI3K/Akt/mTOR | Induces ATG | [55] |
U87 *, U373 *, U138MG *, C6 ‡ | 7.5 μM, 10 μM and 15 μM (IC50 19–28 μM) | PI3K/Akt, NFκB, caspase-3 | Induces G2/M cell cycle arrest, inhibits cell proliferation | [56] |
U251 *, U87 * | 10 μM, 20 μM and 40 μM | p-Akt, p-mTOR, PTEN, p53 | Inhibits cell proliferation, migration, and invasion, while inducing apoptosis | [57] |
SNB19 *, A1207 * | 10 μM, 15 μM and 20 μM | PI3K/Akt, Notch1, NEDD4 | Inhibits cell proliferation, induces cell cycle arrest, inhibits cell migration and invasion | [58] |
Model (s) | Cell line (s) and Injection Site | Dose (s) | Effect (s) | Reference |
---|---|---|---|---|
Intracranial xenograft | U-87; caudate-putamen | i.p. injection (120 mg/kg) | Increases survival of curcumin-treated mice | [59] |
Intracranial xenograft | SU-2 and SU-3; caudate nucleus | i.p. injection (300 mg/kg) | Increases survival of curcumin-treated mice | [51] |
Subcutaneous injection | U87MG; right flank | Intratumoral injection (100 mg/kg) | Inhibits tumor growth and induces ATG | [54] |
Intracranial xenograft | C6; striatum | i.p. injection (50 mg/kg) | Inhibits tumor growth | [56] |
Subcutaneous injection | U87; flank | i.p. injection (60 mg/kg) | Decreases tumor volume | [57] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ryskalin, L.; Biagioni, F.; Busceti, C.L.; Lazzeri, G.; Frati, A.; Fornai, F. The Multi-Faceted Effect of Curcumin in Glioblastoma from Rescuing Cell Clearance to Autophagy-Independent Effects. Molecules 2020, 25, 4839. https://doi.org/10.3390/molecules25204839
Ryskalin L, Biagioni F, Busceti CL, Lazzeri G, Frati A, Fornai F. The Multi-Faceted Effect of Curcumin in Glioblastoma from Rescuing Cell Clearance to Autophagy-Independent Effects. Molecules. 2020; 25(20):4839. https://doi.org/10.3390/molecules25204839
Chicago/Turabian StyleRyskalin, Larisa, Francesca Biagioni, Carla L. Busceti, Gloria Lazzeri, Alessandro Frati, and Francesco Fornai. 2020. "The Multi-Faceted Effect of Curcumin in Glioblastoma from Rescuing Cell Clearance to Autophagy-Independent Effects" Molecules 25, no. 20: 4839. https://doi.org/10.3390/molecules25204839
APA StyleRyskalin, L., Biagioni, F., Busceti, C. L., Lazzeri, G., Frati, A., & Fornai, F. (2020). The Multi-Faceted Effect of Curcumin in Glioblastoma from Rescuing Cell Clearance to Autophagy-Independent Effects. Molecules, 25(20), 4839. https://doi.org/10.3390/molecules25204839