Molecular Insights into the Multifunctional Role of Natural Compounds: Autophagy Modulation and Cancer Prevention
Abstract
:1. Introduction
2. Mechanisms of Autophagy Signaling Pathway
3. Molecular Mechanism of Autophagy Signaling in Cancer Pathogenesis
3.1. Autophagy Signaling Roles in Cancer Inhibition
3.2. Autophagy Signaling Roles in Cancer Promotion
4. Natural Compound Triggers in Autophagy Modulation
4.1. Inhibition of Autophagy by Natural Compounds for Cancer Therapy
4.2. Induction of Autophagy by Natural Compounds as Potential Cancer Therapy
5. Role of Natural Compounds in Autophagy Modulation of Neurodegenerative Diseases
6. Effect of Natural Compounds on Solid Tumors and Lymphomas
7. Therapeutic View of Autophagy in Heart/Cardiovascular Diseases
8. Perspectives of Naturally Occurring Autophagy Modulators in Cancer Therapy
9. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Rahman, M.A.; Rhim, H. Therapeutic implication of autophagy in neurodegenerative diseases. BMB Rep. 2017, 50, 345–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Corti, O.; Blomgren, K.; Poletti, A.; Beart, P.M. Autophagy in neurodegeneration: New insights underpinning therapy for neurological diseases. J. Neurochem. 2020, e15002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uddin, M.S.; Stachowiak, A.; Al Mamun, A.; Tzvetkov, N.T.; Takeda, S.; Atanasov, A.G.; Bergantin, L.B.; Abdel-Daim, M.M.; Stankiewicz, A.M. Autophagy and Alzheimer’s Disease: From Molecular Mechanisms to Therapeutic Implications. Front. Aging Neurosci. 2018, 10. [Google Scholar] [CrossRef] [PubMed]
- Arroyo, D.S.; Gaviglio, E.A.; Ramos, J.M.P.; Bussi, C.; Rodriguez-Galan, M.C.; Iribarren, P. Autophagy in inflammation, infection, neurodegeneration and cancer. Int. Immunopharmacol. 2014, 18, 55–65. [Google Scholar] [CrossRef] [Green Version]
- Perry, R.J.; Shulman, G.I. Mechanistic Links between Obesity, Insulin, and Cancer. Trends Cancer 2020, 6, 75–78. [Google Scholar] [CrossRef]
- Ling, Y.; Perez-Soler, R. Disruption of autophagic and autolysosomal signaling pathways leads to synergistic augmentation of erlotinib-induced apoptosis in wild type EGFR human non-small cell lung cancer cell lines. EJC Suppl. 2010, 8, 183. [Google Scholar] [CrossRef]
- Towers, C.G.; Wodetzki, D.; Thorburn, A. Autophagy and cancer: Modulation of cell death pathways and cancer cell adaptations. J. Cell Biol. 2020, 219. [Google Scholar] [CrossRef]
- Manic, G.; Obrist, F.; Kroemer, G.; Vitale, I.; Galluzzi, L. Chloroquine and hydroxychloroquine for cancer therapy. Mol. Cell. Oncol. 2014, 1, e29911. [Google Scholar] [CrossRef]
- Khandia, R.; Dadar, M.; Munjal, A.; Dhama, K.; Karthik, K.; Tiwari, R.; Yatoo, M.I.; Iqbal, H.M.N.; Singh, K.P.; Joshi, S.K.; et al. A Comprehensive Review of Autophagy and Its Various Roles in Infectious, Non-Infectious, and Lifestyle Diseases: Current Knowledge and Prospects for Disease Prevention, Novel Drug Design, and Therapy. Cells 2019, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bialik, S.; Dasari, S.K.; Kimchi, A. Autophagy-dependent cell death—Where, how and why a cell eats itself to death. J. Cell Sci. 2018, 131. [Google Scholar] [CrossRef] [Green Version]
- Lagoa, R.; Silva, J.; Rodrigues, J.R.; Bishayee, A. Advances in phytochemical delivery systems for improved anticancer activity. Biotechnol. Adv. 2020, 38. [Google Scholar] [CrossRef]
- Tavakol, S.; Ashrafizadeh, M.; Deng, S.; Azarian, M.; Abdoli, A.; Motavaf, M.; Poormoghadam, D.; Khanbabaei, H.; Afshar, E.G.; Mandegary, A.; et al. Autophagy Modulators: Mechanistic Aspects and Drug Delivery Systems. Biomolecules 2019, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirata, E.; Ohya, Y.; Suzuki, K. Atg4 plays an important role in efficient expansion of autophagic isolation membranes by cleaving lipidated Atg8 in Saccharomyces cerevisiae. PLoS ONE 2017, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; He, J.; Tian, M.; Zhang, S.Y.; Guo, M.R.; Kasimu, R.; Wang, J.H.; Ouyang, L. UNC51-like kinase 1, autophagic regulator and cancer therapeutic target. Cell Proliferat. 2014, 47, 494–505. [Google Scholar] [CrossRef] [PubMed]
- Rahman, M.A.; Rahman, M.R.; Zaman, T.; Uddin, M.S.; Islam, R.; Abdel-Daim, M.M.; Rhim, H. Emerging Potential of Naturally Occurring Autophagy Modulators Against Neurodegeneration. Curr. Pharm. Des. 2020, 26, 772–779. [Google Scholar] [CrossRef]
- Nakamura, S.; Yoshimori, T. New insights into autophagosome-lysosome fusion. J. Cell Sci. 2017, 130, 1209–1216. [Google Scholar] [CrossRef] [Green Version]
- Schulze, H.; Kolter, T.; Sandhoff, K. Principles of lysosomal membrane degradation Cellular topology and biochemistry of lysosomal lipid degradation. BBA-Mol. Cell Res. 2009, 1793, 674–683. [Google Scholar] [CrossRef] [Green Version]
- Shpilka, T.; Weidberg, H.; Pietrokovski, S.; Elazar, Z. Atg8: An autophagy-related ubiquitin-like protein family. Genome Biol. 2011, 12. [Google Scholar] [CrossRef]
- Agrotis, A.; von Chamier, L.; Oliver, H.; Kiso, K.; Singh, T.; Ketteler, R. Human ATG4 autophagy proteases counteract attachment of ubiquitin-like LC3/GABARAP proteins to other cellular proteins. J. Biol. Chem. 2019, 294, 12610–12621. [Google Scholar] [CrossRef] [Green Version]
- Lystad, A.H.; Simonsen, A. Mechanisms and Pathophysiological Roles of the ATG8 Conjugation Machinery. Cells 2019, 8. [Google Scholar] [CrossRef] [Green Version]
- Ward, C.; Martinez-Lopez, N.; Otten, E.G.; Carroll, B.; Maetzel, D.; Singh, R.; Sarkar, S.; Korolchuk, V.I. Autophagy, lipophagy and lysosomal lipid storage disorders. BBA-Mol. Cell Biol. Lipids 2016, 1861, 269–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galluzzi, L.; Pietrocola, F.; Bravo-San Pedro, J.M.; Amaravadi, R.K.; Baehrecke, E.H.; Cecconi, F.; Codogno, P.; Debnath, J.; Gewirtz, D.A.; Karantza, V.; et al. Autophagy in malignant transformation and cancer progression. Embo J. 2015, 34, 856–880. [Google Scholar] [CrossRef]
- Desantis, V.; Saltarella, I.; Lamanuzzi, A.; Mariggio, M.A.; Racanelli, V.; Vacca, A.; Frassanito, M.A. Autophagy: A New Mechanism of Prosurvival and Drug Resistance in Multiple Myeloma. Transl. Oncol. 2018, 11, 1350–1357. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.N.J.; Chee, C.E.; Huang, S.B.; Sinicrope, F.A. The Role of Autophagy in Cancer: Therapeutic Implications. Mol. Cancer Ther. 2011, 10, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
- Ricci, M.S.; Zong, W.X. Chemotherapeutic approaches for targeting cell death pathways. Oncologist 2006, 11, 342–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mowers, E.E.; Sharifi, M.N.; Macleod, K.F. Autophagy in cancer metastasis. Oncogene 2017, 36, 1619–1630. [Google Scholar] [CrossRef] [PubMed]
- Altman, B.J.; Rathmell, J.C. Metabolic Stress in Autophagy and Cell Death Pathways. CSH Perspect. Biol. 2012, 4. [Google Scholar] [CrossRef] [PubMed]
- Mowers, E.E.; Sharifi, M.N.; Macleod, K.F. Functions of autophagy in the tumor microenvironment and cancer metastasis. FEBS J. 2018, 285, 1751–1766. [Google Scholar] [CrossRef] [Green Version]
- Park, S.A.; Surh, Y.J. Modulation of tumor microenvironment by chemopreventive natural products. Ann. N. Y. Acad. Sci. 2017, 1401, 65–74. [Google Scholar] [CrossRef]
- Zhang, B.; Tian, L.; Xie, J.; Chen, G.; Wang, F. Targeting miRNAs by natural products: A new way for cancer therapy. Biomed. Pharmacother. 2020, 130, 110546. [Google Scholar] [CrossRef]
- Jiang, J.W.; Zhang, L.; Chen, H.N.; Lei, Y.L.; Zhang, T.; Wang, Y.L.; Jin, P.; Lan, J.; Zhou, L.; Huang, Z.; et al. Regorafenib induces lethal autophagy arrest by stabilizing PSAT1 in glioblastoma. Autophagy 2020, 16, 106–122. [Google Scholar] [CrossRef] [PubMed]
- Cuomo, F.; Altucci, L.; Cobellis, G. Autophagy Function and Dysfunction: Potential Drugs as Anti-Cancer Therapy. Cancers 2019, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Livesey, K.M.; Tang, D.L.; Zeh, H.J.; Lotze, M.T. Autophagy inhibition in combination cancer treatment. Curr. Opin. Investig. Drugs 2009, 10, 1269–1279. [Google Scholar] [PubMed]
- Wu, Y.T.; Tan, H.L.; Shui, G.H.; Bauvy, C.; Huang, Q.; Wenk, M.R.; Ong, C.N.; Codogno, P.; Shen, H.M. Dual Role of 3-Methyladenine in Modulation of Autophagy via Different Temporal Patterns of Inhibition on Class I and III Phosphoinositide 3-Kinase. J. Biol. Chem. 2010, 285, 10850–10861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vazquez-Martin, A.; Oliveras-Ferraros, C.; Menendez, J.A. Autophagy Facilitates the Development of Breast Cancer Resistance to the Anti-HER2 Monoclonal Antibody Trastuzumab. PLoS ONE 2009, 4. [Google Scholar] [CrossRef] [Green Version]
- Rothe, K.; Porter, V.; Jiang, X.Y. Current Outlook on Autophagy in Human Leukemia: Foe in Cancer Stem Cells and Drug Resistance, Friend in New Therapeutic Interventions. Int. J. Mol. Sci. 2019, 20. [Google Scholar] [CrossRef] [Green Version]
- Jo, G.H.; Bogler, O.; Chwae, Y.J.; Yoo, H.; Lee, S.H.; Park, J.B.; Kim, Y.J.; Kim, J.H.; Gwak, H.S. Radiation-Induced Autophagy Contributes to Cell Death and Induces Apoptosis Partly in Malignant Glioma Cells. Cancer Res. Treat. 2015, 47, 221–241. [Google Scholar] [CrossRef]
- He, S.S.; Zhao, Z.; Yang, Y.F.; O’Connell, D.; Zhang, X.W.; Oh, S.; Ma, B.Y.; Lee, J.H.; Zhang, T.; Varghese, B.; et al. Truncating mutation in the autophagy gene UVRAG confers oncogenic properties and chemosensitivity in colorectal cancers. Nat. Commun. 2015, 6. [Google Scholar] [CrossRef]
- Yue, Z.Y.; Jin, S.K.; Yang, C.W.; Levine, A.J.; Heintz, N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. USA 2003, 100, 15077–15082. [Google Scholar] [CrossRef] [Green Version]
- Takamura, A.; Komatsu, M.; Hara, T.; Sakamoto, A.; Kishi, C.; Waguri, S.; Eishi, Y.; Hino, O.; Tanaka, K.; Mizushima, N. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 2011, 25, 795–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, X.; Yu, D.D.; Yan, F.; Jing, Y.Y.; Han, Z.P.; Sun, K.; Liang, L.; Hou, J.; Wei, L.X. The role of autophagy induced by tumor microenvironment in different cells and stages of cancer. Cell Biosci. 2015, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rahman, M.A.; Bishayee, K.; Habib, K.; Sadra, A.; Huh, S.O. 18alpha-Glycyrrhetinic acid lethality for neuroblastoma cells via de-regulating the Beclin-1/Bcl-2 complex and inducing apoptosis. Biochem. Pharmacol. 2016, 117, 97–112. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.J.; Lee, C.C.; Shih, Y.L.; Lin, T.Y.; Wang, S.H.; Lin, Y.F.; Shih, C.M. Resveratrol enhances the therapeutic effect of temozolomide against malignant glioma in vitro and in vivo by inhibiting autophagy. Free Radi.c Biol. Med. 2012, 52, 377–391. [Google Scholar] [CrossRef] [PubMed]
- Zanotto-Filho, A.; Braganhol, E.; Klafke, K.; Figueiro, F.; Terra, S.R.; Paludo, F.J.; Morrone, M.; Bristot, I.J.; Battastini, A.M.; Forcelini, C.M.; et al. Autophagy inhibition improves the efficacy of curcumin/temozolomide combination therapy in glioblastomas. Cancer Lett. 2015, 358, 220–231. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.A.; Hwang, H.; Nah, S.Y.; Rhim, H. Gintonin stimulates autophagic flux in primary cortical astrocytes. J. Ginseng Res. 2020, 44, 67–78. [Google Scholar] [CrossRef]
- Kim, H.; Moon, J.Y.; Ahn, K.S.; Cho, S.K. Quercetin induces mitochondrial mediated apoptosis and protective autophagy in human glioblastoma U373MG cells. Oxid. Med. Cell. Longev. 2013, 2013, 596496. [Google Scholar] [CrossRef] [Green Version]
- Xi, G.; Hu, X.; Wu, B.; Jiang, H.; Young, C.Y.; Pang, Y.; Yuan, H. Autophagy inhibition promotes paclitaxel-induced apoptosis in cancer cells. Cancer Lett. 2011, 307, 141–148. [Google Scholar] [CrossRef]
- Suzuki, R.; Kang, Y.; Li, X.; Roife, D.; Zhang, R.; Fleming, J.B. Genistein potentiates the antitumor effect of 5-Fluorouracil by inducing apoptosis and autophagy in human pancreatic cancer cells. Anticancer Res. 2014, 34, 4685–4692. [Google Scholar]
- Lv, X.Q.; Liu, F.; Shang, Y.; Chen, S.Z. Honokiol exhibits enhanced antitumor effects with chloroquine by inducing cell death and inhibiting autophagy in human non-small cell lung cancer cells. Oncol. Rep. 2015, 34, 1289–1300. [Google Scholar] [CrossRef] [Green Version]
- Oh, J.M.; Kim, E.; Chun, S. Ginsenoside Compound K Induces Ros-Mediated Apoptosis and Autophagic Inhibition in Human Neuroblastoma Cells In Vitro and In Vivo. Int. J. Mol. Sci. 2019, 20. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.A.; Bishayee, K.; Sadra, A.; Huh, S.O. Oxyresveratrol activates parallel apoptotic and autophagic cell death pathways in neuroblastoma cells. Biochim. Biophys. Acta Gen Subj. 2017, 1861, 23–36. [Google Scholar] [CrossRef] [PubMed]
- Russo, M.; Russo, G.L. Autophagy inducers in cancer. Biochem. Pharmacol. 2018, 153, 51–61. [Google Scholar] [CrossRef] [PubMed]
- Nunez-Olvera, S.I.; Gallardo-Rincon, D.; Puente-Rivera, J.; Salinas-Vera, Y.M.; Marchat, L.A.; Morales-Villegas, R.; Lopez-Camarillo, C. Autophagy Machinery as a Promising Therapeutic Target in Endometrial Cancer. Front. Oncol. 2019, 9. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.Y.; Wu, H.C.; Wu, J.E.; Huang, K.Y.; Yang, S.C.; Chen, S.X.; Tsao, C.J.; Hsu, K.F.; Chen, Y.L.; Hong, T.M. The dual PI3K/mTOR inhibitor BEZ235 restricts the growth of lung cancer tumors regardless of EGFR status, as a potent accompanist in combined therapeutic regimens. J. Exp. Clin. Cancer Res. 2019, 38. [Google Scholar] [CrossRef] [PubMed]
- Rizzieri, D.A.; Feldman, E.; DiPersio, J.F.; Gabrail, N.; Stock, W.; Strair, R.; Rivera, V.M.; Albitar, M.; Bedrosian, C.L.; Giles, F.J. A phase 2 clinical trial of deforolimus (AP23573, MK-8669), a novel mammalian target of rapamycin inhibitor, in patients with relapsed or refractory hematologic malignancies. Clin. Cancer Res. 2008, 14, 2756–2762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Lu, Y.; Pan, T.; Fan, Z. Roles of autophagy in cetuximab-mediated cancer therapy against EGFR. Autophagy 2010, 6, 1066–1077. [Google Scholar] [CrossRef] [Green Version]
- Lauzier, A.; Normandeau-Guimond, J.; Vaillancourt-Lavigueur, V.; Boivin, V.; Charbonneau, M.; Rivard, N.; Scott, M.S.; Dubois, C.M.; Jean, S. Colorectal cancer cells respond differentially to autophagy inhibition in vivo. Sci. Rep. 2019, 9, 11316. [Google Scholar] [CrossRef] [Green Version]
- Strohecker, A.M.; White, E. Autophagy promotes BrafV600E-driven lung tumorigenesis by preserving mitochondrial metabolism. Autophagy 2014, 10, 384–385. [Google Scholar] [CrossRef] [Green Version]
- Avalos, Y.; Canales, J.; Bravo-Sagua, R.; Criollo, A.; Lavandero, S.; Quest, A.F. Tumor suppression and promotion by autophagy. BioMed Res. Int. 2014, 2014, 603980. [Google Scholar] [CrossRef]
- Shteingauz, A.; Porat, Y.; Voloshin, T.; Schneiderman, R.S.; Munster, M.; Zeevi, E.; Kaynan, N.; Gotlib, K.; Giladi, M.; Kirson, E.D.; et al. AMPK-dependent autophagy upregulation serves as a survival mechanism in response to Tumor Treating Fields (TTFields). Cell. Death Dis. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.A.; Rahman, M.S.; Uddin, M.J.; Mamum-Or-Rashid, A.N.M.; Pang, M.G.; Rhim, H. Emerging risk of environmental factors: Insight mechanisms of Alzheimer’s diseases. Environ. Sci. Pollut. Res. 2020. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.H.W.; Kuo, M.T. Improving radiotherapy in cancer treatment: Promises and challenges. Oncotarget 2017, 8, 62742–62758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, H.; Vong, C.T.; Chen, H.B.; Gao, Y.; Lyu, P.; Qiu, L.; Zhao, M.M.; Liu, Q.; Cheng, Z.H.; Zou, J.; et al. Naturally occurring anti-cancer compounds: Shining from Chinese herbal medicine. Chin. Med. 2019, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, P.Q.; Zhu, L.J.; Sun, D.J.; Gan, F.H.; Gao, S.Y.; Yin, Y.Y.; Chen, L.X. Natural products as modulator of autophagy with potential clinical prospects. Apoptosis 2017, 22, 325–356. [Google Scholar] [CrossRef]
- Choi, E.J.; Kim, G.H. Apigenin Induces Apoptosis through a Mitochondria/Caspase-Pathway in Human Breast Cancer MDA-MB-453 Cells. J. Clin. Biochem. Nutr. 2009, 44, 260–265. [Google Scholar] [CrossRef] [Green Version]
- Mohan, N.; Banik, N.L.; Ray, S.K. Combination of N-(4-hydroxyphenyl) retinamide and apigenin suppressed starvation-induced autophagy and promoted apoptosis in malignant neuroblastoma cells. Neurosci. Lett. 2011, 502, 24–29. [Google Scholar] [CrossRef] [Green Version]
- Nakamura, Y.; Yogosawa, S.; Izutani, Y.; Watanabe, H.; Otsuji, E.; Sakai, T. A combination of indol-3-carbinol and genistein synergistically induces apoptosis in human colon cancer HT-29 cells by inhibiting Akt phosphorylation and progression of autophagy. Mol. Cancer 2009, 8, 100. [Google Scholar] [CrossRef] [Green Version]
- Gan, M.L.; Zheng, T.; Shen, L.Y.; Tan, Y.; Fan, Y.; Shuai, S.R.; Bai, L.; Li, X.W.; Wang, J.Y.; Zhang, S.H.; et al. Genistein reverses isoproterenol-induced cardiac hypertrophy by regulating miR-451/TIMP2. Biomed. Pharmacother. 2019, 112. [Google Scholar] [CrossRef]
- Ma, C.H.; Zhang, Y.X.; Tang, L.H.; Yang, X.J.; Cui, W.M.; Han, C.C.; Ji, W.Y. MicroRNA-1469, a p53-responsive microRNA promotes Genistein induced apoptosis by targeting Mcl1 in human laryngeal cancer cells. Biomed. Pharmacother. 2018, 106, 665–671. [Google Scholar] [CrossRef]
- Kuo, P.L.; Hsu, Y.L.; Cho, C.Y. Plumbagin induces G2-M arrest and autophagy by inhibiting the AKT/mammalian target of rapamycin pathway in breast cancer cells. Mol. Cancer Ther. 2006, 5, 3209–3221. [Google Scholar] [CrossRef] [Green Version]
- Thiyagarajan, V.; Tsai, M.J.; Weng, C.F. Antroquinonol Targets FAK-Signaling Pathway Suppressed Cell Migration, Invasion, and Tumor Growth of C6 Glioma. PLoS ONE 2015, 10, e0141285. [Google Scholar] [CrossRef] [PubMed]
- Yu, C.C.; Chiang, P.C.; Lu, P.H.; Kuo, M.T.; Wen, W.C.; Chen, P.N.; Guh, J.H. Antroquinonol, a natural ubiquinone derivative, induces a cross talk between apoptosis, autophagy and senescence in human pancreatic carcinoma cells. J. Nutr. Biochem. 2012, 23, 900–907. [Google Scholar] [CrossRef] [PubMed]
- Chiang, P.C.; Lin, S.C.; Pan, S.L.; Kuo, C.H.; Tsai, I.L.; Kuo, M.T.; Wen, W.C.; Chen, P.; Guh, J.H. Antroquinonol displays anticancer potential against human hepatocellular carcinoma cells: A crucial role of AMPK and mTOR pathways. Biochem. Pharmacol. 2010, 79, 162–171. [Google Scholar] [CrossRef] [PubMed]
- Chiou, J.F.; Wu, A.T.H.; Wang, W.T.; Kuo, T.H.; Gelovani, J.G.; Lin, I.H.; Wu, C.H.; Chiu, W.T.; Deng, W.P. A Preclinical Evaluation of Antrodia camphorata Alcohol Extracts in the Treatment of Non-Small Cell Lung Cancer Using Non-Invasive Molecular Imaging. Evid.-Based Complement. Altern. Med. 2011, 1–12. [Google Scholar] [CrossRef]
- Kannaiyan, R.; Manu, K.A.; Chen, L.X.; Li, F.; Rajendran, P.; Subramaniam, A.; Lam, P.; Kumar, A.P.; Sethi, G. Celastrol inhibits tumor cell proliferation and promotes apoptosis through the activation of c-Jun N-terminal kinase and suppression of PI3 K/Akt signaling pathways. Apoptosis 2011, 16, 1028–1041. [Google Scholar] [CrossRef]
- Wang, W.B.; Feng, L.X.; Yue, Q.X.; Wu, W.Y.; Guan, S.H.; Jiang, B.H.; Yang, M.; Liu, X.; Guo, D.A. Paraptosis accompanied by autophagy and apoptosis was induced by celastrol, a natural compound with influence on proteasome, ER stress and Hsp90. J. Cell. Physiol. 2012, 227, 2196–2206. [Google Scholar] [CrossRef]
- Li, H.; Zhang, Y.Y.; Tan, H.W.; Jia, Y.F.; Li, D. Therapeutic effect of tripterine on adjuvant arthritis in rats. J. Ethnopharmacol. 2008, 118, 479–484. [Google Scholar] [CrossRef] [Green Version]
- Dyshlovoy, S.A.; Hauschild, J.; Amann, K.; Tabakmakher, K.M.; Venz, S.; Walther, R.; Guzii, A.G.; Makarieva, T.N.; Shubina, L.K.; Fedorov, S.N.; et al. Marine alkaloid Monanchocidin a overcomes drug resistance by induction of autophagy and lysosomal membrane permeabilization. Oncotarget 2015, 6, 17328–17341. [Google Scholar] [CrossRef]
- Dyshlovoy, S.A.; Venz, S.; Hauschild, J.; Tabakmakher, K.M.; Otte, K.; Madanchi, R.; Walther, R.; Guzii, A.G.; Makarieva, T.N.; Shubina, L.K.; et al. Anti-migratory activity of marine alkaloid monanchocidin A—Proteomics-based discovery and confirmation. Proteomics 2016, 16, 1590–1603. [Google Scholar] [CrossRef]
- McGuire, W.P.; Blessing, J.A.; Moore, D.; Lentz, S.S.; Photopulos, G. Paclitaxel has moderate activity in squamous cervix cancer: A gynecologic oncology group study. J. Clin. Oncol. 1996, 14, 792–795. [Google Scholar] [CrossRef]
- Kampan, N.C.; Madondo, M.T.; McNally, O.M.; Quinn, M.; Plebanski, M. Paclitaxel and Its Evolving Role in the Management of Ovarian Cancer. Biomed. Res. Int. 2015, 2015, 413076. [Google Scholar] [CrossRef] [PubMed]
- Qiu, Y.; Li, P.; Ji, C.Y. Cell Death Conversion under Hypoxic Condition in Tumor Development and Therapy. Int. J. Mol. Sci. 2015, 16, 25536–25551. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.; Na, J.; Lee, M.S.; Cha, E.Y.; Sul, J.Y.; Park, J.B.; Lee, J.S. Combination of pristimerin and paclitaxel additively induces autophagy in human breast cancer cells via ERK1/2 regulation. Mol. Med. Rep. 2018, 18, 4281–4288. [Google Scholar] [CrossRef] [PubMed]
- Veldhoen, R.A.; Banman, S.L.; Hemmerling, D.R.; Odsen, R.; Simmen, T.; Simmonds, A.J.; Underhill, D.A.; Goping, I.S. The chemotherapeutic agent paclitaxel inhibits autophagy through two distinct mechanisms that regulate apoptosis. Oncogene 2013, 32, 736–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.; Hu, X.J.; Jin, B.; Qu, X.J.; Hou, K.Z.; Liu, Y.P. ss-Elemene induces apoptosis as well as protective autophagy in human non-small-cell lung cancer A549 cells. J. Pharm. Pharmacol. 2012, 64, 146–153. [Google Scholar] [CrossRef]
- Zhan, Y.H.; Liu, J.; Qu, X.J.; Hou, K.Z.; Wang, K.F.; Liu, Y.P.; Wu, B. beta-Elemene Induces Apoptosis in Human Renal-cell Carcinoma 786-0 Cells through Inhibition of MAPK/ERK and PI3K/Akt/mTOR Signalling Pathways. Asian Pac. J. Cancer P 2012, 13, 2739–2744. [Google Scholar] [CrossRef] [Green Version]
- Saiki, S.; Sasazawa, Y.; Imamichi, Y.; Kawajiri, S.; Fujimaki, T.; Tanida, I.; Kobayashi, H.; Sato, F.; Sato, S.; Ishikawa, K.I.; et al. Caffeine induces apoptosis by enhancement of autophagy via PI3K/Akt/mTOR/p70S6K inhibition. Autophagy 2011, 7, 176–187. [Google Scholar] [CrossRef] [Green Version]
- Lopez-Lazaro, M. Anticancer and carcinogenic properties of curcumin: Considerations for its clinical development as a cancer chemopreventive and chemotherapeutic agent. Mol. Nutr. Food Res. 2008, 52, S103–S127. [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]
- Klinger, N.V.; Mittal, S. Therapeutic Potential of Curcumin for the Treatment of Brain Tumors. Oxid. Med. Cell. Longev. 2016, 2016, 9324085. [Google Scholar] [CrossRef]
- Masuelli, L.; Benvenuto, M.; Di Stefano, E.; Mattera, R.; Fantini, M.; De Feudis, G.; De Smaele, E.; Tresoldi, I.; Giganti, M.G.; Modesti, A.; et al. Curcumin blocks autophagy and activates apoptosis of malignant mesothelioma cell lines and increases the survival of mice intraperitoneally transplanted with a malignant mesothelioma cell line. Oncotarget 2017, 8, 34405–34422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buhrmann, C.; Kraehe, P.; Lueders, C.; Shayan, P.; Goel, A.; Shakibaei, M. Curcumin Suppresses Crosstalk between Colon Cancer Stem Cells and Stromal Fibroblasts in the Tumor Microenvironment: Potential Role of EMT. PLoS ONE 2014, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, R.; Li, H.B.; Wu, S.Q.; Qu, J.; Yuan, H.Y.; Zhou, Y.G.; Lu, Q. MicroRNA-1246 regulates the radio-sensitizing effect of curcumin in bladder cancer cells via activating P53. Int. Urol. Nephrol. 2019, 51, 1771–1779. [Google Scholar] [CrossRef] [PubMed]
- Aggarwal, B.B.; Deb, L.; Prasad, S. Curcumin Differs from Tetrahydrocurcumin for Molecular Targets, Signaling Pathways and Cellular Responses. Molecules 2015, 20, 185–205. [Google Scholar] [CrossRef] [Green Version]
- Wu, J.C.; Lai, C.S.; Badmaev, V.; Nagabhushanam, K.; Ho, C.T.; Pan, M.H. Tetrahydrocurcumin, a major metabolite of curcumin, induced autophagic cell death through coordinative modulation of PI3K/Akt-mTOR and MAPK signaling pathways in human leukemia HL-60 cells. Mol. Nutr. Food Res. 2011, 55, 1646–1654. [Google Scholar] [CrossRef] [PubMed]
- Hu, F.; Wei, F.; Wang, Y.L.; Wu, B.B.; Fang, Y.; Xiong, B. EGCG synergizes the therapeutic effect of cisplatin and oxaliplatin through autophagic pathway in human colorectal cancer cells. J. Pharmacol. Sci. 2015, 128, 27–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Irimie, A.I.; Braicu, C.; Zanoaga, O.; Pileczki, V.; Gherman, C.; Berindan-Neagoe, I.; Campian, R.S. Epigallocatechin-3-gallate suppresses cell proliferation and promotes apoptosis and autophagy in oral cancer SSC-4 cells. Oncotargets Ther. 2015, 8, 461–470. [Google Scholar] [CrossRef] [Green Version]
- Zhou, J.; Farah, B.L.; Sinha, R.A.; Wu, Y.; Singh, B.K. Epigallocatechin-3-Gallate (EGCG), a Green Tea Polyphenol, Stimulates Hepatic Autophagy and Lipid Clearance (vol 9, e87161, 2014). PLoS ONE 2014, 9. [Google Scholar] [CrossRef]
- Gray, A.L.; Stephens, C.A.; Bigelow, R.L.H.; Coleman, D.T.; Cardelli, J.A. The Polyphenols (-)-Epigallocatechin-3-Gallate and Luteolin Synergistically Inhibit TGF-beta-Induced Myofibroblast Phenotypes through RhoA and ERK Inhibition. PLoS ONE 2014, 9, e109208. [Google Scholar] [CrossRef] [Green Version]
- Chen, A.P.; Zhang, L. The antioxidant (-)-epigallocatechin-3-gallate inhibits rat hepatic stellate cell proliferation in vitro by blocking the tyrosine phosphorylation and reducing the gene expression of platelet-derived growth factor-beta receptor. J. Biol. Chem. 2003, 278, 23381–23389. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.L.; Xu, Y. Epigallocatechin gallate attenuates uric acid-induced injury in rat renal interstitial fibroblasts NRK-49F by up-regulation of miR-9. Eur. Rev. Med. Pharmacol. 2018, 22, 7458–7469. [Google Scholar]
- Adhami, V.M.; Syed, D.N.; Khan, N.; Mukhtar, H. Dietary flavonoid fisetin: A novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management. Biochem. Pharmacol. 2012, 84, 1277–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, X.; Ma, X.M.; Li, Q.W.; Yang, Y.; Xu, X.L.; Sun, J.Q.; Yu, M.W.; Cao, K.X.; Yang, L.; Yang, G.W.; et al. Anti-cancer effects of fisetin on mammary carcinoma cells via regulation of the PI3K/Akt/mTOR pathway: In vitro and in vivo studies. Int. J. Mol. Med. 2018, 42, 811–820. [Google Scholar] [CrossRef] [PubMed]
- Khan, N.; Afaq, F.; Khusro, F.H.; Adhami, V.M.; Suh, Y.; Mukhtar, H. Dual inhibition of phosphatidylinositol 3-kinase/Akt and mammalian target of rapamycin signaling in human nonsmall cell lung cancer cells by a dietary flavonoid fisetin. Int. J. Cancer 2012, 130, 1695–1705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suh, Y.; Afaq, F.; Khan, N.; Johnson, J.J.; Khusro, F.H.; Mukhtar, H. Fisetin induces autophagic cell death through suppression of mTOR signaling pathway in prostate cancer cells. Carcinogenesis 2010, 31, 1424–1433. [Google Scholar] [CrossRef] [Green Version]
- Yang, P.M.; Tseng, H.H.; Peng, C.W.; Chen, W.S.; Chiu, S.J. Dietary flavonoid fisetin targets caspase-3-deficient human breast cancer MCF-7 cells by induction of caspase-7-associated apoptosis and inhibition of autophagy. Int. J. Oncol. 2012, 40, 469–478. [Google Scholar] [CrossRef] [Green Version]
- Tiwari, R.V.; Parajuli, P.; Sylvester, P.W. gamma-Tocotrienol-induced endoplasmic reticulum stress and autophagy act concurrently to promote breast cancer cell death. Biochem. Cell. Biol. 2015, 93, 306–320. [Google Scholar] [CrossRef]
- Deng, S.; Shanmugam, M.K.; Kumar, A.P.; Yap, C.T.; Sethi, G.; Bishayee, A. Targeting autophagy using natural compounds for cancer prevention and therapy. Cancer-Am. Cancer Soc. 2019, 125, 1228–1246. [Google Scholar] [CrossRef]
- Jiang, Q.; Rao, X.Y.; Kim, C.Y.; Freiser, H.; Zhang, Q.B.; Jiang, Z.Y.; Li, G.L. Gamma-tocotrienol induces apoptosis and autophagy in prostate cancer cells by increasing intracellular dihydrosphingosine and dihydroceramide. Int. J. Cancer 2012, 130, 685–693. [Google Scholar] [CrossRef] [Green Version]
- Tiwari, R.V.; Parajuli, P.; Sylvester, P.W. gamma-Tocotrienol-induced autophagy in malignant mammary cancer cells. Exp. Biol. Med. 2014, 239, 33–44. [Google Scholar] [CrossRef]
- Kim, S.H.; Park, E.J.; Lee, C.R.; Chun, J.N.; Cho, N.H.; Kim, I.G.; Lee, S.; Kim, T.W.; Park, H.H.; So, I.; et al. Geraniol induces cooperative interaction of apoptosis and autophagy to elicit cell death in PC-3 prostate cancer cells. Int. J. Oncol. 2012, 40, 1683–1690. [Google Scholar] [CrossRef] [PubMed]
- Trzoss, L.; Fukuda, T.; Costa-Lotufo, L.V.; Jimenez, P.; La Clair, J.J.; Fenical, W. Seriniquinone, a selective anticancer agent, induces cell death by autophagocytosis, targeting the cancer-protective protein dermcidin. Proc. Natl. Acad. Sci. USA 2014, 111, 14687–14692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chu, S.C.; Hsieh, Y.S.; Yu, C.C.; Lai, Y.Y.; Chen, P.N. Thymoquinone Induces Cell Death in Human Squamous Carcinoma Cells via Caspase Activation-Dependent Apoptosis and LC3-II Activation-Dependent Autophagy. PLoS ONE 2014, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Racoma, I.O.; Meisen, W.H.; Wang, Q.E.; Kaur, B.; Wani, A.A. Thymoquinone Inhibits Autophagy and Induces Cathepsin-Mediated, Caspase-Independent Cell Death in Glioblastoma Cells. PLoS ONE 2013, 8. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.C.; Lee, N.H.; Hsu, H.H.; Ho, T.J.; Tu, C.C.; Hsieh, D.J.Y.; Lin, Y.M.; Chen, L.M.; Kuo, W.W.; Huang, C.Y. Thymoquinone Induces Caspase-Independent, Autophagic Cell Death in CPT-11-Resistant LoVo Colon Cancer via Mitochondrial Dysfunction and Activation of JNK and p38. J. Agric. Food Chem. 2015, 63, 1540–1546. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.L.; Tang, W.; Du, G.H.; Kokudo, N. Targeting apoptosis pathways in cancer with magnolol and honokiol, bioactive constituents of the bark of Magnolia officinalis. Drug Discov. Ther. 2011, 5, 202–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, H.B.; Yi, X.; Gao, J.M.; Ying, X.X.; Guan, H.Q.; Li, J.C. Magnolol-induced H460 cells death via autophagy but not apoptosis. Arch. Pharm. Res. 2007, 30, 1566–1574. [Google Scholar] [CrossRef]
- Rasul, A.; Yu, B.; Khan, M.; Zhang, K.; Iqbal, F.; Ma, T.H.; Yang, H. Magnolol, a natural compound, induces apoptosis of SGC-7901 human gastric adenocarcinoma cells via the mitochondrial and PI3K/Akt signaling pathways. Int. J. Oncol. 2012, 40, 1153–1161. [Google Scholar] [CrossRef] [Green Version]
- Bae, U.J.; Jung, E.S.; Jung, S.J.; Chae, S.W.; Park, B.H. Mulberry leaf extract displays antidiabetic activity in db/db mice via Akt and AMP-activated protein kinase phosphorylation. Food Nutr. Res. 2018, 62. [Google Scholar] [CrossRef] [Green Version]
- Cheng, K.C.; Wang, C.J.; Chang, Y.C.; Hung, T.W.; Lai, C.J.; Kuo, C.W.; Huang, H.P. Mulberry fruits extracts induce apoptosis and autophagy of liver cancer cell and prevent hepatocarcinogenesis in vivo. J. Food Drug Anal. 2020, 28, 84–93. [Google Scholar] [CrossRef] [Green Version]
- Kwon, Y.H.; Bishayee, K.; Rahman, M.A.; Hong, J.S.; Lim, S.S.; Huh, S.O. Morus alba Accumulates Reactive Oxygen Species to Initiate Apoptosis via FOXO-Caspase 3-Dependent Pathway in Neuroblastoma Cells. Mol. Cells 2015, 38, 630–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lao, Y.Z.; Wan, G.; Liu, Z.Y.; Wang, X.Y.; Ruan, P.; Xu, W.; Xu, D.Q.; Xie, W.D.; Zhang, Y.; Xu, H.X.; et al. The natural compound oblongifolin C inhibits autophagic flux and enhances antitumor efficacy of nutrient deprivation. Autophagy 2014, 10, 736–749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, M.; Lao, Y.Z.; Tan, H.S.; Lu, G.; Ren, Y.; Zheng, Z.Q.; Yi, J.; Fu, W.W.; Shen, H.M.; Xu, H.X. Oblongifolin C suppresses lysosomal function independently of TFEB nuclear translocation. Acta Pharmacol. Sin. 2019, 40, 929–937. [Google Scholar] [CrossRef] [PubMed]
- Acharya, B.R.; Bhattacharyya, S.; Choudhury, D.; Chakrabarti, G. The microtubule depolymerizing agent naphthazarin induces both apoptosis and autophagy in A549 lung cancer cells. Apoptosis 2011, 16, 924–939. [Google Scholar] [CrossRef]
- Herr, I.; Lozanovski, V.; Houben, P.; Schemmer, P.; Buchler, M.W. Sulforaphane and related mustard oils in focus of cancer prevention and therapy. Wien. Med. Wochenschr. 2013, 163, 80–88. [Google Scholar] [CrossRef]
- Herman-Antosiewicz, A.; Johnson, D.E.; Singh, S.V. Sulforaphane causes autophagy to inhibit release of cytochrome C and apoptosis in human prostate cancer cells. Cancer Res. 2006, 66, 5828–5835. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Z.; Lin, K.; Hu, Y.; Zhou, Y.; Ding, X.; Wang, Y.; Wu, W. Sulforaphane metabolites inhibit migration and invasion via microtubule-mediated Claudins dysfunction or inhibition of autolysosome formation in human non-small cell lung cancer cells. Cell Death Dis. 2019, 10, 259. [Google Scholar] [CrossRef] [Green Version]
- Chaudhuri, D.; Orsulic, S.; Ashok, B.T. Antiproliferative activity of sulforaphane in Akt-overexpressing ovarian cancer cells. Mol. Cancer Ther. 2007, 6, 334–345. [Google Scholar] [CrossRef] [Green Version]
- Uddin, M.S.; Mamun, A.A.; Jakaria, M.; Thangapandiyan, S.; Ahmad, J.; Rahman, M.A.; Mathew, B.; Abdel-Daim, M.M.; Aleya, L. Emerging promise of sulforaphane-mediated Nrf2 signaling cascade against neurological disorders. Sci. Total Environ. 2020, 707, 135624. [Google Scholar] [CrossRef]
- Kim, S.M.; Park, H.S.; Jun, D.Y.; Woo, H.J.; Woo, M.H.; Yang, C.H.; Kim, Y.H. Mollugin induces apoptosis in human Jurkat T cells through endoplasmic reticulum stress-mediated activation of JNK and caspase-12 and subsequent activation of mitochondria-dependent caspase cascade regulated by Bcl-xL. Toxicol. Appl. Pharm. 2009, 241, 210–220. [Google Scholar] [CrossRef]
- Zhang, L.; Wang, H.D.; Zhu, J.H.; Xu, J.G.; Ding, K. Mollugin induces tumor cell apoptosis and autophagy via the PI3K/AKT/mTOR/p70S6K and ERK signaling pathways. Biochem. Biophys Res. Commun. 2014, 450, 247–254. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.Y.; Lee, S.Y.; Kang, S.S.; Kim, Y.S. Antitumor Activity of Jujuboside B and the Underlying Mechanism via Induction of Apoptosis and Autophagy. J. Nat. Prod. 2014, 77, 370–376. [Google Scholar] [CrossRef] [PubMed]
- Lu, J.; Sun, D.P.; Gao, S.; Gao, Y.; Ye, J.T.; Liu, P.Q. Cyclovirobuxine D Induces Autophagy-Associated Cell Death via the Akt/mTOR Pathway in MCF-7 Human Breast Cancer Cells. J. Pharmacol. Sci. 2014, 125, 74–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ouyang, L.; Chen, Y.; Wang, X.Y.; Lu, R.F.; Zhang, S.Y.; Tian, M.; Xie, T.; Liu, B.; He, G. Polygonatum odoratum lectin induces apoptosis and autophagy via targeting EGFR-mediated Ras-Raf-MEK-ERK pathway in human MCF-7 breast cancer cells. Phytomedicine 2014, 21, 1658–1665. [Google Scholar] [CrossRef]
- Li, C.Y.; Chen, J.; Lu, B.M.; Shi, Z.; Wang, H.L.; Zhang, B.; Zhao, K.L.; Qi, W.; Bao, J.K.; Wang, Y. Molecular Switch Role of Akt in Polygonatum odoratum Lectin-Induced Apoptosis and Autophagy in Human Non-Small Cell Lung Cancer A549 Cells. PLoS ONE 2014, 9, e101526. [Google Scholar] [CrossRef]
- Jiang, H.; Zhang, L.; Kuo, J.; Kuo, K.; Gautam, S.C.; Groc, L.; Rodriguez, A.I.; Koubi, D.; Hunter, T.J.; Corcoran, G.B.; et al. Resveratrol-induced apoptotic death in human U251 glioma cells. Mol. Cancer Ther. 2005, 4, 554–561. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.A.; Kim, N.H.; Kim, S.H.; Oh, S.M.; Huh, S.O. Antiproliferative and Cytotoxic Effects of Resveratrol in Mitochondria-Mediated Apoptosis in Rat B103 Neuroblastoma Cells. Korean J. Physiol. Pharm. 2012, 16, 321–326. [Google Scholar] [CrossRef]
- Lang, F.F.; Qin, Z.Y.; Li, F.; Zhang, H.L.; Fang, Z.H.; Hao, E.K. Apoptotic Cell Death Induced by Resveratrol Is Partially Mediated by the Autophagy Pathway in Human Ovarian Cancer Cells. PLoS ONE 2015, 10. [Google Scholar] [CrossRef]
- Garg, T.; Yadav, V.K. Effects of Resveratrol as an anticancer agent. A Systematic Review and Meta-Analysis. Indian J. Pharmacol. 2013, 45, S206. [Google Scholar]
- Gong, C.H.; Xia, H.L. Resveratrol suppresses melanoma growth by promoting autophagy through inhibiting the PI3K/AKT/mTOR signaling pathway. Exp. Ther. Med. 2020, 19, 1878–1886. [Google Scholar] [CrossRef] [Green Version]
- Liu, Q.J.; Fang, Q.; Ji, S.Q.; Han, Z.X.; Cheng, W.L.; Zhang, H.J. Resveratrol-mediated apoptosis in renal cell carcinoma via the p53/AMP-activated protein kinase/mammalian target of rapamycin autophagy signaling pathway. Mol. Med. Rep. 2018, 17, 502–508. [Google Scholar] [CrossRef] [PubMed]
- Godichaud, S.; Si-Tayeb, K.; Auge, N.; Desmouliere, A.; Balabaud, C.; Payrastre, B.; Negre-Salvayre, A.; Rosenbaum, J. The grape-derived polyphenol resveratrol differentially affects epidermal and platelet-derived growth factor signaling in human liver myofibroblasts. Int. J. Biochem. Cell Biol. 2006, 38, 629–637. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.D.; Liu, L.; Steinle, J.J. miRNA15a regulates insulin signal transduction in the retinal vasculature. Cell. Signal. 2018, 44, 28–32. [Google Scholar] [CrossRef]
- Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of Biological Importance of Quercetin: A Bioactive Flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, B.N.; Kumar, D.; Shankar, S.; Srivastava, R.K. Rottlerin induces autophagy which leads to apoptotic cell death through inhibition of PI3K/Akt/mTOR pathway in human pancreatic cancer stem cells. Biochem. Pharmacol. 2012, 84, 1154–1163. [Google Scholar] [CrossRef]
- Rahman, M.A.; Kim, N.H.; Yang, H.; Huh, S.O. Angelicin induces apoptosis through intrinsic caspase-dependent pathway in human SH-SY5Y neuroblastoma cells. Mol. Cell. Biochem. 2012, 369, 95–104. [Google Scholar] [CrossRef]
- Rahman, M.A.; Bishayee, K.; Huh, S.O. Angelica polymorpha Maxim Induces Apoptosis of Human SH-SY5Y Neuroblastoma Cells by Regulating an Intrinsic Caspase Pathway. Mol. Cells 2016, 39, 119–128. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.R.; Chen, Y.Q.; Chen, X.D.; Liang, Y.; Yang, D.P.; Dong, J.; Yang, N.; Liang, Z.Q. Angelicin inhibits the malignant behaviours of human cervical cancer potentially via inhibiting autophagy. Exp. Ther. Med. 2019, 18, 3365–3374. [Google Scholar] [CrossRef] [Green Version]
- Uddin, M.S.; Rahman, M.A.; Kabir, M.T.; Behl, T.; Mathew, B.; Perveen, A.; Barreto, G.E.; Bin-Jumah, M.N.; Abdel-Daim, M.M.; Ashraf, G.M. Multifarious roles of mTOR signaling in cognitive aging and cerebrovascular dysfunction of Alzheimer’s disease. IUBMB Life 2020. [Google Scholar] [CrossRef]
- Leng, S.; Hao, Y.; Du, D.; Xie, S.; Hong, L.; Gu, H.; Zhu, X.; Zhang, J.; Fan, D.; Kung, H.F. Ursolic acid promotes cancer cell death by inducing Atg5-dependent autophagy. Int. J. Cancer 2013, 133, 2781–2790. [Google Scholar] [CrossRef]
- Xavier, C.P.R.; Lima, C.F.; Pedro, D.F.N.; Wilson, J.M.; Kristiansen, K.; Pereira-Wilson, C. Ursolic acid induces cell death and modulates autophagy through JNK pathway in apoptosis-resistant colorectal cancer cells. J. Nutr. Biochem. 2013, 24, 706–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, C.; Yin, S.T.; Dong, Y.H.; Guo, X.; Fan, L.H.; Ye, M.; Hu, H.B. Autophagy-dependent EIF2AK3 activation compromises ursolic acid-induced apoptosis through upregulation of MCL1 in MCF-7 human breast cancer cells. Autophagy 2013, 9, 196–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shin, S.W.; Park, J.W. Autophagy inhibition enhances ursolic acid-induced apoptosis in PC3 cells. Free Radic. Biol. Med. 2012, 53, S116. [Google Scholar] [CrossRef]
- Wang, S.Y.; Yu, Q.J.; Bao, J.K.; Liu, B. Polygonatum cyrtonema lectin, a potential antineoplastic drug targeting programmed cell death pathways. Biochem. Biophys Res. Commun. 2011, 406, 497–500. [Google Scholar] [CrossRef]
- Liu, B.; Cheng, Y.; Bian, H.J.; Bao, J.K. Molecular mechanisms of Polygonatum cyrtonema lectin-induced apoptosis and autophagy in cancer cells. Autophagy 2009, 5, 253–255. [Google Scholar] [CrossRef] [Green Version]
- Liu, B.; Cheng, Y.; Zhang, B.; Bian, H.J.; Bao, J.K. Polygonatum cyrtonema lectin induces apoptosis and autophagy in human melanoma A375 cells through a mitochondria-mediated ROS-p38-p53 pathway. Cancer Lett. 2009, 275, 54–60. [Google Scholar] [CrossRef]
- Duan, W.J.; Li, Q.S.; Xia, M.Y.; Tashiro, S.I.; Onodera, S.; Ikejima, T. Silibinin Activated p53 and Induced Autophagic Death in Human Fibrosarcoma HT1080 Cells via Reactive Oxygen Species-p38 and c-Jun N-Terminal Kinase Pathways. Biol. Pharm. Bull. 2011, 34, 47–53. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.A.; Yang, H.; Kim, N.H.; Huh, S.O. Induction of apoptosis by Dioscorea nipponica Makino extracts in human SH-SY5Y neuroblastoma cells via mitochondria-mediated pathway. Anim. Cells Syst. 2014, 18, 41–51. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.A.; Yang, H.; Lim, S.S.; Huh, S.O. Apoptotic Effects of Melandryum firmum Root Extracts in Human SH-SY5Y Neuroblastoma Cells. Exp. Neurobiol. 2013, 22, 208–213. [Google Scholar] [CrossRef] [Green Version]
- Bassham, D.C.; Crespo, J.L. Autophagy in plants and algae. Front. Plant Sci. 2014, 5. [Google Scholar] [CrossRef] [Green Version]
- Uddin, M.S.; Mamun, A.A.; Rahman, M.A.; Kabir, M.T.; Alkahtani, S.; Alanazi, I.S.; Perveen, A.; Ashraf, G.M.; Bin-Jumah, M.N.; Abdel-Daim, M.M. Exploring the Promise of Flavonoids to Combat Neuropathic Pain: From Molecular Mechanisms to Therapeutic Implications. Front. Neurosci. 2020, 14, 478. [Google Scholar] [CrossRef]
- Rahman, M.A.; Hong, J.S.; Huh, S.O. Antiproliferative properties of Saussurea lappa Clarke root extract in SH-SY5Y neuroblastoma cells via intrinsic apoptotic pathway. Anim. Cells Syst. 2015, 19, 119–126. [Google Scholar] [CrossRef] [Green Version]
- Tian, X.; Song, H.S.; Cho, Y.M.; Park, B.; Song, Y.J.; Jang, S.; Kang, S.C. Anticancer effect of Saussurea lappa extract via dual control of apoptosis and autophagy in prostate cancer cells. Medicine 2017, 96. [Google Scholar] [CrossRef]
- Ryoo, N.; Rahman, M.A.; Hwang, H.; Ko, S.K.; Nah, S.Y.; Kim, H.C.; Rhim, H. Ginsenoside Rk1 is a novel inhibitor of NMDA receptors in cultured rat hippocampal neurons. J. Ginseng Res. 2020, 44, 490–495. [Google Scholar] [CrossRef]
- Ko, H.; Kim, Y.J.; Park, J.S.; Park, J.H.; Yang, H.O. Autophagy Inhibition Enhances Apoptosis Induced by Ginsenoside Rk1 in Hepatocellular Carcinoma Cells. Biosci. Biotechnol. Biochem. 2009, 73, 2183–2189. [Google Scholar] [CrossRef] [Green Version]
- Zecchini, S.; Serafini, F.P.; Catalani, E.; Giovarelli, M.; Coazzoli, M.; Di Renzo, I.; De Palma, C.; Perrotta, C.; Clementi, E.; Buonanno, F.; et al. Dysfunctional autophagy induced by the pro-apoptotic natural compound climacostol in tumour cells. Cell Death Dis. 2018, 10. [Google Scholar] [CrossRef] [Green Version]
- Saha, S.; Panigrahi, D.P.; Patil, S.; Bhutia, S.K. Autophagy in health and disease: A comprehensive review. Biomed. Pharmacother. 2018, 104, 485–495. [Google Scholar] [CrossRef]
- Grossi, C.; Rigacci, S.; Ambrosini, S.; Ed Dami, T.; Luccarini, I.; Traini, C.; Failli, P.; Berti, A.; Casamenti, F.; Stefani, M. The polyphenol oleuropein aglycone protects TgCRND8 mice against Ass plaque pathology. PLoS ONE 2013, 8, e71702. [Google Scholar] [CrossRef]
- Luccarini, I.; Grossi, C.; Rigacci, S.; Coppi, E.; Pugliese, A.M.; Pantano, D.; la Marca, G.; Ed Dami, T.; Berti, A.; Stefani, M.; et al. Oleuropein aglycone protects against pyroglutamylated-3 amyloid-ss toxicity: Biochemical, epigenetic and functional correlates. Neurobiol. Aging 2015, 36, 648–663. [Google Scholar] [CrossRef]
- Zhu, Z.; Yan, J.; Jiang, W.; Yao, X.G.; Chen, J.; Chen, L.; Li, C.; Hu, L.; Jiang, H.; Shen, X. Arctigenin effectively ameliorates memory impairment in Alzheimer’s disease model mice targeting both beta-amyloid production and clearance. J. Neurosci. 2013, 33, 13138–13149. [Google Scholar] [CrossRef]
- Lu, C.; Guo, Y.; Yan, J.; Luo, Z.; Luo, H.B.; Yan, M.; Huang, L.; Li, X. Design, synthesis, and evaluation of multitarget-directed resveratrol derivatives for the treatment of Alzheimer’s disease. J. Med. Chem. 2013, 56, 5843–5859. [Google Scholar] [CrossRef] [PubMed]
- de Oliveira, M.R.; Nabavi, S.F.; Manayi, A.; Daglia, M.; Hajheydari, Z.; Nabavi, S.M. Resveratrol and the mitochondria: From triggering the intrinsic apoptotic pathway to inducing mitochondrial biogenesis, a mechanistic view. BBA-Gen. Subj. 2016, 1860, 727–745. [Google Scholar] [CrossRef] [PubMed]
- Juhasz, B.; Varga, B.; Gesztelyi, R.; Kemeny-Beke, A.; Zsuga, J.; Tosaki, A. Resveratrol: A Multifunctional Cytoprotective Molecule. Curr. Pharm. Biotechnol. 2010, 11, 810–818. [Google Scholar] [CrossRef] [PubMed]
- Filomeni, G.; Graziani, I.; De Zio, D.; Dini, L.; Centonze, D.; Rotilio, G.; Ciriolo, M.R. Neuroprotection of kaempferol by autophagy in models of rotenone-mediated acute toxicity: Possible implications for Parkinson’s disease. Neurobiol. Aging 2012, 33, 767–785. [Google Scholar] [CrossRef]
- Lu, J.H.; Tan, J.Q.; Durairajan, S.S.; Liu, L.F.; Zhang, Z.H.; Ma, L.; Shen, H.M.; Chan, H.Y.; Li, M. Isorhynchophylline, a natural alkaloid, promotes the degradation of alpha-synuclein in neuronal cells via inducing autophagy. Autophagy 2012, 8, 98–108. [Google Scholar] [CrossRef] [Green Version]
- Sasazawa, Y.; Sato, N.; Umezawa, K.; Simizu, S. Conophylline protects cells in cellular models of neurodegenerative diseases by inducing mammalian target of rapamycin (mTOR)-independent autophagy. J. Biol. Chem. 2015, 290, 6168–6178. [Google Scholar] [CrossRef] [Green Version]
- 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 alpha-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] [CrossRef]
- Guo, Y.J.; Dong, S.Y.; Cui, X.X.; Feng, Y.; Liu, T.; Yin, M.; Kuo, S.H.; Tan, E.K.; Zhao, W.J.; Wu, Y.C. Resveratrol alleviates MPTP-induced motor impairments and pathological changes by autophagic degradation of alpha-synuclein via SIRT1-deacetylated LC3. Mol. Nutr. Food Res. 2016, 60, 2161–2175. [Google Scholar] [CrossRef]
- Ferretta, A.; Gaballo, A.; Tanzarella, P.; Piccoli, C.; Capitanio, N.; Nico, B.; Annese, T.; Di Paola, M.; Dell’aquila, C.; De Mari, M.; et al. Effect of resveratrol on mitochondrial function: Implications in parkin-associated familiar Parkinson’s disease. Biochim. Biophys. Acta 2014, 1842, 902–915. [Google Scholar] [CrossRef] [Green Version]
- Cai, C.Z.; Zhou, H.F.; Yuan, N.N.; Wu, M.Y.; Lee, S.M.; Ren, J.Y.; Su, H.X.; Lu, J.J.; Chen, X.P.; Li, M.; et al. Natural alkaloid harmine promotes degradation of alpha-synuclein via PKA-mediated ubiquitin-proteasome system activation. Phytomedicine 2019, 61, 152842. [Google Scholar] [CrossRef]
- Wong, V.K.; Wu, A.G.; Wang, J.R.; Liu, L.; Law, B.Y. Neferine attenuates the protein level and toxicity of mutant huntingtin in PC-12 cells via induction of autophagy. Molecules 2015, 20, 3496–3514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarkar, S.; Davies, J.E.; Huang, Z.; Tunnacliffe, A.; Rubinsztein, D.C. Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein. J. Biol. Chem. 2007, 282, 5641–5652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, W.; Wei, W.; Gaertig, M.A.; Li, S.; Li, X.J. Therapeutic Effect of Berberine on Huntington’s Disease Transgenic Mouse Model. PLoS ONE 2015, 10, e0134142. [Google Scholar] [CrossRef] [PubMed]
- Castillo, K.; Nassif, M.; Valenzuela, V.; Rojas, F.; Matus, S.; Mercado, G.; Court, F.A.; van Zundert, B.; Hetz, C. Trehalose delays the progression of amyotrophic lateral sclerosis by enhancing autophagy in motoneurons. Autophagy 2013, 9, 1308–1320. [Google Scholar] [CrossRef] [Green Version]
- Menzies, F.M.; Huebener, J.; Renna, M.; Bonin, M.; Riess, O.; Rubinsztein, D.C. Autophagy induction reduces mutant ataxin-3 levels and toxicity in a mouse model of spinocerebellar ataxia type 3. Brain 2010, 133, 93–104. [Google Scholar] [CrossRef] [Green Version]
- Singhal, J.; Nagaprashantha, L.D.; Vatsyayan, R.; Awasthi, S.; Singhal, S.S. Didymin induces apoptosis by inhibiting N-Myc and upregulating RKIP in neuroblastoma. Cancer Prev. Res. 2012, 5, 473–483. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.A.; Kim, N.H.; Huh, S.O. Cytotoxic effect of gambogic acid on SH-SY5Y neuroblastoma cells is mediated by intrinsic caspase-dependent signaling pathway. Mol. Cell. Biochem. 2013, 377, 187–196. [Google Scholar] [CrossRef]
- Vengoji, R.; Macha, M.A.; Batra, S.K.; Shonka, N.A. Natural products: A hope for glioblastoma patients. Oncotarget 2018, 9, 22194–22219. [Google Scholar] [CrossRef] [Green Version]
- Yuan, Y.; Xue, X.; Guo, R.B.; Sun, X.L.; Hu, G. Resveratrol Enhances the Antitumor Effects of Temozolomide in Glioblastoma via ROS-dependent AMPK-TSC-mTOR Signaling Pathway. CNS. Neurosci. Ther. 2012, 18, 536–546. [Google Scholar] [CrossRef]
- Shah, S.H. Carbonic anhydrase, net photosynthetic rate and yield of black cumin (Nigella sativa) plants sprayed with kinetin. Acta Bot. Croat. 2008, 67, 63–68. [Google Scholar]
- Hussain, A.R.; Uddin, S.; Ahmed, M.; Al-Dayel, F.; Bavi, P.P.; Al-Kuraya, K.S. Phosphorylated IkappaBalpha predicts poor prognosis in activated B-cell lymphoma and its inhibition with thymoquinone induces apoptosis via ROS release. PLoS ONE 2013, 8, e60540. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, K.; Ishikawa, C.; Katano, H.; Yasumoto, T.; Mori, N. Fucoxanthin and its deacetylated product, fucoxanthinol, induce apoptosis of primary effusion lymphomas. Cancer Lett. 2011, 300, 225–234. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Q.; Guan, J.W.; Qin, Y.H.; Ren, P.; Zhang, Z.W.; Lv, J.; Sun, S.J.; Zhang, C.L.; Mao, W.F. Curcumin sensitizes lymphoma cells to DNA damage agents through regulating Rad51-dependent homologous recombination. Biomed. Pharmacother. 2018, 97, 115–119. [Google Scholar] [CrossRef] [PubMed]
- Shi, L.Y.; Qin, H.H.; Jin, X.D.; Yang, X.X.; Lu, X.; Wang, H.G.; Wang, R.Y.; Yu, D.Y.; Feng, B.M. The natural phenolic peperobtusin A induces apoptosis of lymphoma U937 cells via the Caspase dependent and p38 MAPK signaling pathways. Biomed. Pharmacother. 2018, 102, 772–781. [Google Scholar] [CrossRef] [PubMed]
- Xiao, X.H.; Li, H.L.; Jin, H.Z.; Jin, J.; Yu, M.; Ma, C.M.; Tong, Y.; Zhou, L.; Lei, H.; Xu, H.Z.; et al. Identification of 11(13)-dehydroivaxillin as a potent therapeutic agent against non-Hodgkin’s lymphoma. Cell Death Dis. 2017, 8. [Google Scholar] [CrossRef] [PubMed]
- Martino, R.; Beer, M.F.; Elso, O.; Donadel, O.; Sulsen, V.; Anesini, C. Sesquiterpene lactones from Ambrosia spp. are active against a murine lymphoma cell line by inducing apoptosis and cell cycle arrest. Toxicol. In Vitro 2015, 29, 1529–1536. [Google Scholar] [CrossRef]
- Leidal, A.M.; Levine, B.; Debnath, J. Autophagy and the cell biology of age-related disease. Nat. Cell Biol. 2018, 20, 1338–1348. [Google Scholar] [CrossRef]
- Sciarretta, S.; Maejima, Y.; Zablocki, D.; Sadoshima, J. The Role of Autophagy in the Heart. Annu. Rev. Physiol. 2018, 80, 1–26. [Google Scholar] [CrossRef]
- Meyer, G.; Czompa, A.; Reboul, C.; Csepanyi, E.; Czegledi, A.; Bak, I.; Balla, G.; Balla, J.; Tosaki, A.; Lekli, I. The cellular autophagy markers Beclin-1 and LC3B-II are increased during reperfusion in fibrillated mouse hearts. Curr. Pharm. Des. 2013, 19, 6912–6918. [Google Scholar] [CrossRef]
- Shirakabe, A.; Zhai, P.; Ikeda, Y.; Saito, T.; Maejima, Y.; Hsu, C.P.; Nomura, M.; Egashira, K.; Levine, B.; Sadoshima, J. Drp1-Dependent Mitochondrial Autophagy Plays a Protective Role Against Pressure Overload-Induced Mitochondrial Dysfunction and Heart Failure. Circulation 2016, 133, 1249–1263. [Google Scholar] [CrossRef] [Green Version]
- Matsui, Y.; Takagi, H.; Qu, X.P.; Abdellatif, M.; Sakoda, H.; Asano, T.; Levine, B.; Sadoshima, J. Distinct roles of autophagy in the heart during ischemia and reperfusion—Roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ. Res. 2007, 100, 914–922. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Yao, X.; Zhang, Q.J.; Zhu, M.; Liu, Z.P.; Ci, B.; Xie, Y.; Carlson, D.; Rothermel, B.A.; Sun, Y.; et al. Beclin-1-Dependent Autophagy Protects the Heart During Sepsis. Circulation 2018, 138, 2247–2262. [Google Scholar] [CrossRef] [PubMed]
- Gyongyosi, A.; Szoke, K.; Fenyvesi, F.; Fejes, Z.; Debreceni, I.B.; Nagy, B.; Tosaki, A.; Lekli, I. Inhibited autophagy may contribute to heme toxicity in cardiomyoblast cells. Biochem. Biophys Res. Commun. 2019, 511, 732–738. [Google Scholar] [CrossRef] [PubMed]
- Zheng, Y.T.; Shi, B.H.; Ma, M.Q.; Wu, X.Q.; Lin, X.H. The novel relationship between Sirt3 and autophagy in myocardial ischemia-reperfusion. J. Cell Physiol. 2019, 234, 5488–5495. [Google Scholar] [CrossRef] [PubMed]
- Zilinyi, R.; Czompa, A.; Czegledi, A.; Gajtko, A.; Pituk, D.; Lekli, I.; Tosaki, A. The Cardioprotective Effect of Metformin in Doxorubicin-Induced Cardiotoxicity: The Role of Autophagy. Molecules 2018, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gyongyosi, A.; Zilinyi, R.; Czegledi, A.; Tosaki, A.; Tosaki, A.; Lekli, I. The Role of Autophagy and Death Pathways in Dose-dependent Isoproterenolinduced Cardiotoxicity. Curr. Pharm. Des. 2019, 25, 2192–2198. [Google Scholar] [CrossRef]
- Dong, Y.; Chen, H.W.; Gao, J.L.; Liu, Y.M.; Li, J.; Wang, J. Molecular machinery and interplay of apoptosis and autophagy in coronary heart disease. J. Mol. Cell. Cardiol. 2019, 136, 27–41. [Google Scholar] [CrossRef] [Green Version]
- Lekli, I.; Haines, D.D.; Balla, G.; Tosaki, A. Autophagy: An adaptive physiological countermeasure to cellular senescence and ischaemia/reperfusion-associated cardiac arrhythmias. J. Cell. Mol. Med. 2017, 21, 1058–1072. [Google Scholar] [CrossRef]
- Rahman, M.A.; Saha, S.K.; Rahman, M.S.; Uddin, M.J.; Uddin, M.S.; Pang, M.G.; Rhim, H.; Cho, S.G. Molecular Insights Into Therapeutic Potential of Autophagy Modulation by Natural Products for Cancer Stem Cells. Front. Cell Dev. Biol. 2020, 8. [Google Scholar] [CrossRef]
- Sohn, E.J.; Park, H.T. Natural agents mediated autophagic signal networks in cancer. Cancer Cell Int. 2017, 17. [Google Scholar] [CrossRef] [Green Version]
Natural Compound/Chemical | Cell Model | Molecular Mechanisms | Combination with Autophagy Inhibitors | References |
---|---|---|---|---|
18α-Glycyrrhetinic acid | Neuroblastoma | Autophagy and apoptosis induction | CQ combination induces cell death | [42] |
Resveratrol | Glioblastomas | Induction of apoptosis and autophagy via reduction of ROS/MAPK pathway | Baf A1 combination augments apoptotic cell death | [43] |
Curcumin | Glioblastomas | Induction of autophagy and apoptosis | CQ combination rises apoptotic cell death | [44] |
Gintonin | Cortical astrocytes and glioblastoma cells | Induction of autophagy | CQ and Baf A1 combination stimulates autophagy | [45] |
Quercetin | U373MG cells | PI3K/Akt inhibition and causes apoptosis and autophagy | CQ combination increases apoptotic cell death | [46] |
Paclitaxel | Human lung carcinoma A549 cells | Induction of autophagy and apoptosis | 3-MA combination enhances apoptotic cell death | [47] |
Genistein | MIA PaCa-2 human pancreatic cancer cell | Autophagy and apoptosis induction | CQ combination stimulates apoptotic cell death | [48] |
Honokiol | Non-small cell lung cancer A549 and H460 cells | p62 and LC3 induces autophagy induction | Combination of CQ triggered apoptosis | [49] |
Ginsenoside Compound K | Neuroblastoma SK-N-BE(2) and SH-SY5Y cells | ROS-mediated autophagy inhibition and apoptosis induction | Combination of CQ inhibit autophagy and induces apoptosis | [50] |
Oxyresveratrol | Neuroblastoma SK-N-BE(2) and SH-SY5Y cells | PI3K/AKT/mTOR pathway independent autophagy and apoptosis | 3-MA combination enhances apoptotic cell death | [51] |
Natural Compounds | Cellular/Animal Models | Concentrations/Doses | Effects and Mechanisms | References |
---|---|---|---|---|
Thymoquinone from Nigella sativa | Activated B cell lymphoma cell lines | 5–10 mM | ROS production and apoptosis | [191] |
Resveratrol | Neuroblastoma | 30 µM | Apoptosis induction and antiproliferation | [137] |
Fuxocanthinol | Primary effusion lymphomas BCBL-1 and TY-1 | 1.3–5 µM | Apoptosis induction and cell cycle arrest | [192] |
Curcumin | CH12F3 lymphoma cells | 5 µM | Caspase-3 dependent apoptosis and DNA damage | [193] |
Peperobtusin A | Lymphoma U937 cells | 25, 50, 100 μM | Caspase-3, 8, 9 dependent apoptosis and p38 MAPK activation | [194] |
11(13)-dehydroivaxillin (DHI) | Lymphoid malignancies of NHL cells xenografts | 5, 7, 10 μM | Induction of NF-κB and apoptosis | [195] |
Psilostachyin C | Murine lymphoma cell line BW5147 | 0.01–50 μg/mL | Induction of apoptosis, necrosis, and ROS generation | [196] |
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
Rahman, M.A.; Rahman, M.H.; Hossain, M.S.; Biswas, P.; Islam, R.; Uddin, M.J.; Rahman, M.H.; Rhim, H. Molecular Insights into the Multifunctional Role of Natural Compounds: Autophagy Modulation and Cancer Prevention. Biomedicines 2020, 8, 517. https://doi.org/10.3390/biomedicines8110517
Rahman MA, Rahman MH, Hossain MS, Biswas P, Islam R, Uddin MJ, Rahman MH, Rhim H. Molecular Insights into the Multifunctional Role of Natural Compounds: Autophagy Modulation and Cancer Prevention. Biomedicines. 2020; 8(11):517. https://doi.org/10.3390/biomedicines8110517
Chicago/Turabian StyleRahman, Md. Ataur, MD. Hasanur Rahman, Md. Shahadat Hossain, Partha Biswas, Rokibul Islam, Md Jamal Uddin, Md. Habibur Rahman, and Hyewhon Rhim. 2020. "Molecular Insights into the Multifunctional Role of Natural Compounds: Autophagy Modulation and Cancer Prevention" Biomedicines 8, no. 11: 517. https://doi.org/10.3390/biomedicines8110517