Mitochondrial Metabolism in Melanoma
Abstract
:1. Introduction
2. BRAF—A Major Driver towards Glycolysis
RAF1 (CRAF)-Bypassing BRAF
3. SOX2—Mediating a Shift towards Oxidative Phosphorylation
4. MCL1—Maintaining Integrity of Mitochondria in Melanoma Cells
5. TRAP1—Stabilizing Mitochondrial Proteins and Inhibiting Oxidative Phosphorylation
6. RHOA and Serum Response Factor (SRF)—Regulating Mitochondrial Fusion versus Fission and Glutaminolysis
7. SIRT3—Inducing Oxidative Phosphorylation in Melanoma Cells
8. PTEN and AKT1—Regulating Both Glycolysis and Oxidative Phosphorylation
9. Discussion
10. Conclusions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Faries, M.B.; Steen, S.; Ye, X.; Sim, M.; Morton, D.L. Late Recurrence in Melanoma: Clinical Implications of Lost Dormancy. J. Am. Coll. Surg. 2013, 217, 27–34. [Google Scholar] [CrossRef] [Green Version]
- Ossowski, L.; Aguirre-Ghiso, J. Dormancy of metastatic melanoma. Pigment. Cell Melanoma Res. 2009, 23, 41–56. [Google Scholar] [CrossRef]
- Liu, W.; Dowling, J.P.; Murray, W.K.; McArthur, G.A.; Thompson, J.F.; Wolfe, R.; Kelly, J.W. Rate of Growth in Melanomas. Arch. Dermatol. 2006, 142, 1551–1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zbytek, B.; Carlson, J.A.; Granese, J.; Ross, J.; Mihm, M.C., Jr.; Slominski, A. Current concepts of metastasis in melanoma. Expert Rev. Dermatol. 2008, 3, 569–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Avagliano, A.; Fiume, G.; Pelagalli, A.; Sanità, G.; Ruocco, M.R.; Montagnani, S.; Arcucci, A. Metabolic Plasticity of Melanoma Cells and Their Crosstalk With Tumor Microenvironment. Front. Oncol. 2020, 10, 722. [Google Scholar] [CrossRef]
- Potter, M.; Newport, E.; Morten, K.J. The Warburg effect: 80 years on. Biochem. Soc. Trans. 2016, 44, 1499–1505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goyal, M.S.; Hawrylycz, M.; Miller, J.A.; Snyder, A.Z.; Raichle, M.E. Aerobic Glycolysis in the Human Brain Is Associated with Development and Neotenous Gene Expression. Cell Metab. 2014, 19, 49–57. [Google Scholar] [CrossRef] [Green Version]
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
- López-Lázaro, M. The warburg effect: Why and how do cancer cells activate glycolysis in the presence of oxygen? Anti-Cancer Agents Med. Chem. 2008, 8, 305–312. [Google Scholar] [CrossRef]
- Kim, J.; DeBerardinis, R.J. Mechanisms and Implications of Metabolic Heterogeneity in Cancer. Cell Metab. 2019, 30, 434–446. [Google Scholar] [CrossRef]
- Chaube, B.; Malvi, P.; Vikram Singh, S.; Muhammad, N.; Singh Meena, A.; Bhat, M.K. Targeting metabolic flexibility by simultaneously inhibiting respiratory complex I and lactate generation retards melanoma progression. Oncotarget 2015, 6, 37281–37299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fischer, G.M.; Vashisht Gopal, Y.N.; McQuade, J.L.; Peng, W.; DeBerardinis, R.J.; Davies, M.A. Metabolic strategies of melanoma cells: Mechanisms, interactions with the tumor microenvironment, and therapeutic implications. Pigment. Cell Melanoma Res. 2018, 31, 11–30. [Google Scholar] [CrossRef]
- Andreucci, E.; Peppicelli, S.; Ruzzolini, J.; Bianchini, F.; Biagioni, A.; Papucci, L.; Magnelli, L.; Mazzanti, B.; Stecca, B.; Calorini, L. The acidic tumor microenvironment drives a stem-like phenotype in melanoma cells. J. Mol. Med. 2020, 98, 1431–1446. [Google Scholar] [CrossRef]
- Muz, B.; de la Puente, P.; Azab, F.; Azab, A.K. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia 2015, 3, 83–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ho, J.; de Moura, M.B.; Lin, Y.; Vincent, G.; Thorne, S.; Duncan, L.M.; Hui-Min, L.; Kirkwood, J.M.; Becker, D.; Van Houten, B.; et al. Importance of glycolysis and oxidative phosphorylation in advanced melanoma. Mol. Cancer 2012, 11, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, P.R.; Moore, J.A.; Bowles, K.M.; Rushworth, S.A.; Moncrieff, M.D. Mitochondrial oxidative phosphorylation in cutaneous melanoma. Br. J. Cancer 2021, 124, 115–123. [Google Scholar] [CrossRef]
- Graham, K.; Unger, E. Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment. Int. J. Nanomed. 2018, 13, 6049–6058. [Google Scholar] [CrossRef] [Green Version]
- Arbiser, J.L.; Bonner, M.Y.; Gilbert, L.C. Targeting the duality of cancer. NPJ Precis. Oncol. 2017, 1, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Peppicelli, S.; Toti, A.; Giannoni, E.; Bianchini, F.; Margheri, F.; Del Rosso, M.; Calorini, L. Metformin is also effective on lactic acidosis-exposed melanoma cells switched to oxidative phosphorylation. Cell Cycle 2016, 15, 1908–1918. [Google Scholar] [CrossRef]
- Andreucci, E.; Pietrobono, S.; Peppicelli, S.; Ruzzolini, J.; Bianchini, F.; Biagioni, A.; Stecca, B.; Calorini, L. SOX2 as a novel contributor of oxidative metabolism in melanoma cells. Cell Commun. Signal. 2018, 16, 87. [Google Scholar] [CrossRef] [Green Version]
- Bollag, G.; Hirth, P.; Tsai, J.; Zhang, J.; Ibrahim, P.N.; Cho, H.; Spevak, W.; Zhang, C.; Zhang, Y.; Habets, G.; et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nat. Cell Biol. 2010, 467, 596–599. [Google Scholar] [CrossRef] [PubMed]
- Poulikakos, P.I.; Persaud, Y.; Janakiraman, M.; Kong, X.; Ng, C.; Moriceau, G.; Shi, H.; Atefi, M.; Titz, B.; Gabay, M.T.; et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nat. Cell Biol. 2011, 480, 387–390. [Google Scholar] [CrossRef] [Green Version]
- Quintás-Cardama, A.; Cortes, J. Molecular biology of bcr-abl1–positive chronic myeloid leukemia. Blood 2009, 113, 1619–1630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Proietti, I.; Skroza, N.; Bernardini, N.; Tolino, E.; Balduzzi, V.; Marchesiello, A.; Michelini, S.; Volpe, S.; Mambrin, A.; Mangino, G.; et al. Mechanisms of Acquired BRAF Inhibitor Resistance in Melanoma: A Systematic Review. Cancers 2020, 12, 2801. [Google Scholar] [CrossRef]
- Wu, S.; Singh, R. Resistance to chemotherapy and molecularly targeted therapies: Rationale for combination therapy in malignant melanoma. Curr. Mol. Med. 2011, 11, 553–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuan, L.; Mishra, R.; Patel, H.; Alanazi, S.; Wei, X.; Ma, Z.; Garrett, J.T. BRAF Mutant Melanoma Adjusts to BRAF/MEK Inhibitors via Dependence on Increased Antioxidant SOD2 and Increased Reactive Oxygen Species Levels. Cancers 2020, 12, 1661. [Google Scholar] [CrossRef]
- Fan, J.; Kamphorst, J.J.; Mathew, R.; Chung, M.K.; White, E.; Shlomi, T.; Rabinowitz, J.D. Glutamine-driven oxidative phosphorylation is a major ATP source in transformed mammalian cells in both normoxia and hypoxia. Mol. Syst. Biol. 2013, 9, 712. [Google Scholar] [CrossRef]
- Koundouros, N.; Poulogiannis, G. Reprogramming of fatty acid metabolism in cancer. Br. J. Cancer 2020, 122, 4–22. [Google Scholar] [CrossRef] [Green Version]
- Scott, D.; Richardson, A.; Filipp, F.; Knutzen, C.A.; Chiang, G.; Ronai, Z.; Osterman, A.L.; Smith, J.W. Comparative Metabolic Flux Profiling of Melanoma Cell Lines. J. Biol. Chem. 2011, 286, 42626–42634. [Google Scholar] [CrossRef] [Green Version]
- Vyas, S.; Zaganjor, E.; Haigis, M.C. Mitochondria and Cancer. Cell 2016, 166, 555–566. [Google Scholar] [CrossRef]
- Perciavalle, R.M.; Stewart, D.P.; Koss, B.; Lynch, J.; Milasta, S.; Bathina, M.; Temirov, J.; Cleland, M.M.; Pelletier, S.; Schuetz, J.D.; et al. Anti-apoptotic MCL-1 localizes to the mitochondrial matrix and couples mitochondrial fusion to respiration. Nat. Cell Biol. 2012, 14, 575–583. [Google Scholar] [CrossRef] [Green Version]
- Gonzalez Herrera, K.N.; Lee, J.; Haigis, M.C. Intersections between mitochondrial sirtuin signaling and tumor cell metabolism. Crit. Rev. Biochem. Mol. Biol. 2015, 50, 242–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- George, J.; Nihal, M.; Singh, C.K.; Zhong, W.; Liu, X.; Ahmad, N. Pro-Proliferative Function of Mitochondrial Sirtuin Deacetylase SIRT3 in Human Melanoma. J. Investig. Dermatol. 2016, 136, 809–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haq, R.; Shoag, J.; Andreu-Perez, P.; Yokoyama, S.; Edelman, H.; Rowe, G.; Frederick, D.T.; Hurley, A.D.; Nellore, A.; Kung, A.; et al. Oncogenic BRAF Regulates Oxidative Metabolism via PGC1α and MITF. Cancer Cell 2013, 23, 302–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holderfield, M.; Deuker, M.M.; McCormick, F.; McMahon, M. Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat. Rev. Cancer 2014, 14, 455–467. [Google Scholar] [CrossRef] [Green Version]
- Sharma, A.; Shah, S.R.; Illum, H.; Dowell, J. Vemurafenib. Drugs 2012, 72, 2207–2222. [Google Scholar] [CrossRef] [PubMed]
- Bowyer, S.; Lee, R.; Fusi, A.; Lorigan, P. Dabrafenib and its use in the treatment of metastatic melanoma. Melanoma Manag. 2015, 2, 199–208. [Google Scholar] [CrossRef]
- Tran, B.; Cohen, M.S. The discovery and development of binimetinib for the treatment of melanoma. Expert Opin. Drug Discov. 2020, 15, 745–754. [Google Scholar] [CrossRef]
- Kazandjian, D.; Blumenthal, G.M.; Yuan, W.; He, K.; Keegan, P.; Pazdur, R. FDA Approval of Gefitinib for the Treatment of Patients with Metastatic EGFR Mutation–Positive Non–Small Cell Lung Cancer. Clin. Cancer Res. 2016, 22, 1307–1312. [Google Scholar] [CrossRef] [Green Version]
- Tron, A.E.; Belmonte, M.A.; Adam, A.; Aquila, B.M.; Boise, L.; Chiarparin, E.; Cidado, J.; Embrey, K.J.; Gangl, E.; Gibbons, F.D.; et al. Discovery of Mcl-1-specific inhibitor AZD5991 and preclinical activity in multiple myeloma and acute myeloid leukemia. Nat. Commun. 2018, 9, 1–14. [Google Scholar] [CrossRef]
- Masgras, I.; Laquatra, C.; Cannino, G.; Serapian, S.A.; Colombo, G.; Rasola, A. The molecular chaperone TRAP1 in cancer: From the basics of biology to pharmacological targeting. Semin. Cancer Biol. 2021, in press. [Google Scholar] [CrossRef]
- Condelli, V.; Piscazzi, A.; Sisinni, L.; Matassa, D.S.; Maddalena, F.; Lettini, G.; Simeon, V.; Palladino, G.; Amoroso, M.R.; Trino, S.; et al. TRAP1 Is Involved in BRAF Regulation and Downstream Attenuation of ERK Phosphorylation and Cell-Cycle Progression: A Novel Target for BRAF-Mutated Colorectal Tumors. Cancer Res. 2014, 74, 6693–6704. [Google Scholar] [CrossRef] [Green Version]
- Kang, B.H.; Siegelin, M.D.; Plescia, J.; Raskett, C.M.; Garlick, D.S.; Dohi, T.; Lian, J.B.; Stein, G.S.; Languino, L.; Altieri, D.C. Preclinical Characterization of Mitochondria-Targeted Small Molecule Hsp90 Inhibitors, Gamitrinibs, in Advanced Prostate Cancer. Clin. Cancer Res. 2010, 16, 4779–4788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haikala, H.M.; Marques, E.; Turunen, M.; Klefström, J. Myc requires RhoA/SRF to reprogram glutamine metabolism. Small GTPases 2016, 9, 274–282. [Google Scholar] [CrossRef] [Green Version]
- Santiago-Lopez, A.J.; Gutekunst, C.-A.; Gross, R.E. C3 Transferase Gene Therapy for Continuous RhoA Inhibition. In Rho GTPases: Methods and Protocols; Rivero, F., Ed.; Springer: New York, NY, USA, 2018; pp. 267–281. [Google Scholar] [CrossRef]
- Finley, L.W.; Carracedo, A.; Lee, J.; Souza, A.; Egia, A.; Zhang, J.; Teruya-Feldstein, J.; Moreira, P.; Cardoso, S.M.; Clish, C.; et al. SIRT3 Opposes Reprogramming of Cancer Cell Metabolism through HIF1α Destabilization. Cancer Cell 2011, 19, 416–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akamata, K.; Wei, J.; Bhattacharyya, M.; Cheresh, P.; Bonner, M.Y.; Arbiser, J.L.; Raparia, K.; Gupta, M.P.; Kamp, D.W.; Varga, J. SIRT3 is attenuated in systemic sclerosis skin and lungs, and its pharmacologic activation mitigates organ fibrosis. Oncotarget 2016, 7, 69321–69336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, H.; Goel, V.; Haluska, F.G. PTEN signaling pathways in melanoma. Oncogene 2003, 22, 3113–3122. [Google Scholar] [CrossRef] [Green Version]
- Haq, R.; Fisher, D.E.; Widlund, H.R. Molecular Pathways: BRAF Induces Bioenergetic Adaptation by Attenuating Oxidative Phosphorylation. Clin. Cancer Res. 2014, 20, 2257–2263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robey, R.B.; Hay, N. Is Akt the “Warburg kinase”?—Akt-energy metabolism interactions and oncogenesis. Semin. Cancer Biol. 2009, 19, 25–31. [Google Scholar] [CrossRef] [Green Version]
- Houghton, P.J. Everolimus: Fig. 1. Clin. Cancer Res. 2010, 16, 1368–1372. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Kim, S.G.; Blenis, J. Rapamycin: One Drug, Many Effects. Cell Metab. 2014, 19, 373–379. [Google Scholar] [CrossRef] [Green Version]
- Chapuis, N.; Tamburini, J.; Green, A.S.; Vignon, C.; Bardet, V.; Neyret, A.; Pannetier, M.; Willems, L.; Park, S.; Macone, A.; et al. Dual Inhibition of PI3K and mTORC1/2 Signaling by NVP-BEZ235 as a New Therapeutic Strategy for Acute Myeloid Leukemia. Clin. Cancer Res. 2010, 16, 5424–5435. [Google Scholar] [CrossRef] [Green Version]
- Hussain, S.S.; Patel, D.; Ghosh, R.; Kumar, A.P. Extracting the Benefit of Nexrutine® for Cancer Prevention. Curr. Pharmacol. Rep. 2015, 1, 365–372. [Google Scholar] [CrossRef] [Green Version]
- Hambright, H.G.; Meng, P.; Kumar, A.P.; Ghosh, R. Inhibition of PI3K/AKT/mTOR axis disrupts oxidative stress-mediated survival of melanoma cells. Oncotarget 2015, 6, 7195–7208. [Google Scholar] [CrossRef] [Green Version]
- Roberts, P.J.; Der, C.J. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene 2007, 26, 3291–3310. [Google Scholar] [CrossRef] [Green Version]
- Ascierto, P.; Kirkwood, J.M.; Grob, J.-J.; Simeone, E.; Grimaldi, A.M.; Maio, M.; Palmieri, G.; Testori, A.; Marincola, F.M.; Mozzillo, N. The role of BRAF V600 mutation in melanoma. J. Transl. Med. 2012, 10, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bai, R.; Chen, N.; Li, L.; Du, N.; Bai, L.; Lv, Z.; Tian, H.; Cui, J. Mechanisms of Cancer Resistance to Immunotherapy. Front. Oncol. 2020, 10, 1290. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.-W.; Tchernyshyov, I.; Semenza, G.L.; Dang, C.V. HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006, 3, 177–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, S.M.; Yu, H.; Edwards, R.; Chen, L.; Kazianis, S.; Brafford, P.; Acs, G.; Herlyn, M.; Xu, X. Mutant V600E BRAF Increases Hypoxia Inducible Factor-1α Expression in Melanoma. Cancer Res. 2007, 67, 3177–3184. [Google Scholar] [CrossRef] [Green Version]
- Mills, C.N.; Joshi, S.S.; Niles, R.M. Expression and function of hypoxia inducible factor-1 alpha in human melanoma under non-hypoxic conditions. Mol. Cancer 2009, 8, 104. [Google Scholar] [CrossRef] [Green Version]
- Hanna, S.C.; Krishnan, B.; Bailey, S.T.; Moschos, S.J.; Kuan, P.-F.; Shimamura, T.; Osborne, L.D.; Siegel, M.B.; Duncan, L.M.; O’Brien, E.T., III; et al. HIF1α and HIF2α independently activate SRC to promote melanoma metastases. J. Clin. Investig. 2013, 123, 2078–2093. [Google Scholar] [CrossRef] [Green Version]
- Bott, A.; Peng, I.-C.; Fan, Y.; Faubert, B.; Zhao, L.; Li, J.; Neidler, S.; Sun, Y.; Jaber, N.; Krokowski, D.; et al. Oncogenic Myc Induces Expression of Glutamine Synthetase through Promoter Demethylation. Cell Metab. 2015, 22, 1068–1077. [Google Scholar] [CrossRef] [Green Version]
- Hartman, M.L.; Czyz, M. MITF in melanoma: Mechanisms behind its expression and activity. Cell. Mol. Life Sci. 2015, 72, 1249–1260. [Google Scholar] [CrossRef] [Green Version]
- Baenke, F.; Chaneton, B.; Smith, M.; Van Den Broek, N.; Hogan, K.; Tang, H.; Viros, A.; Martin, M.; Galbraith, L.; Girotti, M.R.; et al. Resistance to BRAF inhibitors induces glutamine dependency in melanoma cells. Mol. Oncol. 2016, 10, 73–84. [Google Scholar] [CrossRef]
- LeBleu, V.S.; O’Connell, J.T.; Gonzalez Herrera, K.N.; Wikman, H.; Pantel, K.; Haigis, M.C.; De Carvalho, F.M.; Damascena, A.; Domingos Chinen, L.T.; Rocha, R.M.; et al. PGC-1α mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat. Cell Biol. 2014, 16, 992–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Delgado-Goni, T.; Miniotis, M.F.; Wantuch, S.; Parkes, H.G.; Marais, R.; Workman, P.; Leach, M.O.; Beloueche-Babari, M. The BRAF Inhibitor Vemurafenib Activates Mitochondrial Metabolism and Inhibits Hyperpolarized Pyruvate-Lactate Exchange in BRAF-Mutant Human Melanoma Cells. Mol. Cancer Ther. 2016, 15, 2987–2999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vazquez, F.; Lim, J.-H.; Chim, H.; Bhalla, K.; Girnun, G.; Pierce, K.; Clish, C.; Granter, S.R.; Widlund, H.; Spiegelman, B.M.; et al. PGC1α Expression Defines a Subset of Human Melanoma Tumors with Increased Mitochondrial Capacity and Resistance to Oxidative Stress. Cancer Cell 2013, 23, 287–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ratnikov, B.I.; Scott, D.; Osterman, A.L.; Smith, J.W.; Ronai, Z.A. Metabolic rewiring in melanoma. Oncogene 2017, 36, 147–157. [Google Scholar] [CrossRef] [Green Version]
- BuscàR; Berra, E.; Gaggioli, C.; Khaled, M.; Bille, K.; Marchetti, B.; Thyss, R.; Fitsialos, G.; Larribère, L.; Bertolotto, C.; et al. Hypoxia-inducible factor 1α is a new target of microphthalmia-associated transcription factor (MITF) in melanoma cells. J. Cell Biol. 2005, 170, 49–59. [Google Scholar] [CrossRef] [Green Version]
- Luebker, S.A.; Koepsell, S.A. Diverse Mechanisms of BRAF Inhibitor Resistance in Melanoma Identified in Clinical and Preclinical Studies. Front. Oncol. 2019, 9, 268. [Google Scholar] [CrossRef] [Green Version]
- Thompson, N.; Adams, D.J.; Ranzani, M. Synthetic lethality: Emerging targets and opportunities in melanoma. Pigment. Cell Melanoma Res. 2017, 30, 183–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Trunzer, K.; Pavlick, A.C.; Schuchter, L.; Gonzalez, R.; McArthur, G.A.; Hutson, T.E.; Moschos, S.J.; Flaherty, K.T.; Kim, K.B.; Weber, J.S.; et al. Pharmacodynamic Effects and Mechanisms of Resistance to Vemurafenib in Patients With Metastatic Melanoma. J. Clin. Oncol. 2013, 31, 1767–1774. [Google Scholar] [CrossRef] [PubMed]
- Hélias-Rodzewicz, Z.; Funck-Brentano, E.; Terrones, N.; Beauchet, A.; Zimmermann, U.; Marin, C.; Saiag, P.; Emile, J.-F. Variation of mutant allele frequency in NRAS Q61 mutated melanomas. BMC Dermatol. 2017, 17, 9. [Google Scholar] [CrossRef] [Green Version]
- Kunz, M.; Vera, J. Modelling of Protein Kinase Signaling Pathways in Melanoma and Other Cancers. Cancers 2019, 11, 465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carlino, M.S.; Fung, C.; Shahheydari, H.; Todd, J.R.; Boyd, S.C.; Irvine, M.; Nagrial, A.M.; Scolyer, R.A.; Kefford, R.F.; Long, G.V.; et al. Preexisting MEK1P124 Mutations Diminish Response to BRAF Inhibitors in Metastatic Melanoma Patients. Clin. Cancer Res. 2015, 21, 98–105. [Google Scholar] [CrossRef] [Green Version]
- Vergani, E.; Vallacchi, V.; Frigerio, S.; Deho, P.; Mondellini, P.; Perego, P.; Cassinelli, G.; Lanzi, C.; Testi, M.A.; Rivoltini, L.; et al. Identification of MET and SRC Activation in Melanoma Cell Lines Showing Primary Resistance to PLX4032. Neoplasia 2011, 13, 1132–1142. [Google Scholar] [CrossRef] [Green Version]
- Sabbatino, F.; Wang, Y.; Wang, X.; Flaherty, K.T.; Yu, L.; Pepin, D.; Scognamiglio, G.; Pepe, S.; Kirkwood, J.M.; Cooper, Z.A.; et al. PDGFRalpha up-regulation mediated by sonic hedgehog pathway activation leads to BRAF inhibitor resistance in melanoma cells with BRAF mutation. Oncotarget 2014, 5, 1926–1941. [Google Scholar] [CrossRef] [Green Version]
- Shi, H.; Hong, A.; Kong, X.; Koya, R.C.; Song, C.; Moriceau, G.; Hugo, W.; Yu, C.C.; Ng, C.; Chodon, T.; et al. A Novel AKT1 Mutant Amplifies an Adaptive Melanoma Response to BRAF Inhibition. Cancer Discov. 2014, 4, 69–79. [Google Scholar] [CrossRef] [Green Version]
- Govindarajan, B.; Sligh, J.E.; Vincent, B.J.; Li, M.; Canter, J.A.; Nickoloff, B.J.; Rodenburg, R.J.; Smeitink, J.A.; Oberley, L.; Zhang, Y.; et al. Overexpression of Akt converts radial growth melanoma to vertical growth melanoma. J. Clin. Investig. 2007, 117, 719–729. [Google Scholar] [CrossRef] [Green Version]
- Fisher, M.L.; Grun, D.; Adhikary, G.; Xu, W.; Eckert, R.L. Inhibition of YAP function overcomes BRAF inhibitor resistance in melanoma cancer stem cells. Oncotarget 2017, 8, 110257–110272. [Google Scholar] [CrossRef] [Green Version]
- Villanueva, J.; Vultur, A.; Herlyn, M. Resistance to BRAF Inhibitors: Unraveling Mechanisms and Future Treatment Options: Figure 1. Cancer Res. 2011, 71, 7137–7140. [Google Scholar] [CrossRef] [Green Version]
- Flaherty, K.T.; Robert, C.; Hersey, P.; Nathan, P.; Garbe, C.; Milhem, M.; Demidov, L.V.; Hassel, J.C.; Rutkowski, P.; Mohr, P.; et al. Improved Survival with MEK Inhibition in BRAF-Mutated Melanoma. N. Engl. J. Med. 2012, 367, 107–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benito-Jardón, L.; Díaz-Martínez, M.; Arellano-Sánchez, N.; Vaquero-Morales, P.; Esparís-Ogando, A.; Teixidó, J. Resistance to MAPK Inhibitors in Melanoma Involves Activation of the IGF1R–MEK5–Erk5 Pathway. Cancer Res. 2019, 79, 2244–2256. [Google Scholar] [CrossRef] [Green Version]
- Bridges, H.R.; Jones, A.J.Y.; Pollak, M.N.; Hirst, J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem. J. 2014, 462, 475–487. [Google Scholar] [CrossRef] [Green Version]
- Jaune, E.; Rocchi, S. Metformin: Focus on Melanoma. Front. Endocrinol. 2018, 9, 472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryabaya, O.; Prokofieva, A.; Akasov, R.; Khochenkov, D.; Emelyanova, M.; Burov, S.; Markvicheva, E.; Inshakov, A.; Stepanova, E. Metformin increases antitumor activity of MEK inhibitor binimetinib in 2D and 3D models of human metastatic melanoma cells. Biomed. Pharmacother. 2019, 109, 2548–2560. [Google Scholar] [CrossRef] [PubMed]
- McQuade, J.L.; Vashisht Gopal, Y. Counteracting oxidative phosphorylation-mediated resistance of melanomas to MAPK pathway inhibition. Mol. Cell. Oncol. 2015, 2, e991610. [Google Scholar] [CrossRef] [Green Version]
- Filipp, F.V.; Ratnikov, B.; De Ingeniis, J.; Smith, J.W.; Osterman, A.L.; Scott, D. Glutamine-fueled mitochondrial metabolism is decoupled from glycolysis in melanoma. Pigment. Cell Melanoma Res. 2012, 25, 732–739. [Google Scholar] [CrossRef] [Green Version]
- Nielson, T.C.; Le, H.V. Inhibition of Glycolysis and Glutaminolysis: An Emerging Drug Discovery Approach to Combat Cancer. Curr. Top. Med. Chem. 2018, 18, 494–504. [Google Scholar] [CrossRef]
- Strohecker, A.M.; White, E. Targeting Mitochondrial Metabolism by Inhibiting Autophagy in BRAF-Driven Cancers. Cancer Discov. 2014, 4, 766–772. [Google Scholar] [CrossRef] [Green Version]
- Xia, S.; Lin, R.; Jin, L.; Zhao, L.; Kang, H.-B.; Pan, Y.; Liu, S.; Qian, G.; Qian, Z.; Konstantakou, E.; et al. Prevention of Dietary-Fat-Fueled Ketogenesis Attenuates BRAF V600E Tumor Growth. Cell Metab. 2017, 25, 358–373. [Google Scholar] [CrossRef] [Green Version]
- Kang, H.-B.; Fan, J.; Lin, R.; Elf, S.; Ji, Q.; Zhao, L.; Jin, L.; Seo, J.H.; Shan, C.; Arbiser, J.L.; et al. Metabolic Rewiring by Oncogenic BRAF V600E Links Ketogenesis Pathway to BRAF-MEK1 Signaling. Mol. Cell 2015, 59, 345–358. [Google Scholar] [CrossRef] [Green Version]
- Aloia, A.; Müllhaupt, D.; Chabbert, C.D.; Eberhart, T.; Flückiger-Mangual, S.; Vukolic, A.; Eichhoff, O.M.; Irmisch, A.; Alexander, L.T.; Scibona, E.; et al. A Fatty Acid Oxidation-dependent Metabolic Shift Regulates the Adaptation of BRAF-mutated Melanoma to MAPK Inhibitors. Clin. Cancer Res. 2019, 25, 6852–6867. [Google Scholar] [CrossRef]
- Tian, Y.; Guo, W. A Review of the Molecular Pathways Involved in Resistance to BRAF Inhibitors in Patients with Advanced-Stage Melanoma. Med Sci. Monit. 2020, 26, e920957. [Google Scholar] [CrossRef]
- Dumaz, N.; Hayward, R.; Martin, J.; Ogilvie, L.; Hedley, D.; Curtin, J.A.; Bastian, B.C.; Springer, C.; Marais, R. In Melanoma, RASMutations Are Accompanied by Switching Signaling from BRAF to CRAF and Disrupted Cyclic AMP Signaling. Cancer Res. 2006, 66, 9483–9491. [Google Scholar] [CrossRef] [Green Version]
- Dhomen, N.; Marais, R. New insight into BRAF mutations in cancer. Curr. Opin. Genet. Dev. 2007, 17, 31–39. [Google Scholar] [CrossRef]
- Montagut, C.; Sharma, S.V.; Shioda, T.; McDermott, U.; Ulman, M.; Ulkus, L.E.; Dias-Santagata, D.; Stubbs, H.; Lee, D.Y.; Singh, A.; et al. Elevated CRAF as a Potential Mechanism of Acquired Resistance to BRAF Inhibition in Melanoma. Cancer Res. 2008, 68, 4853–4861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kholodenko, B.N. Drug Resistance Resulting from Kinase Dimerization Is Rationalized by Thermodynamic Factors Describing Allosteric Inhibitor Effects. Cell Rep. 2015, 12, 1939–1949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galmiche, A.; Fueller, J.; Santel, A.; Krohne, G.; Wittig, I.; Doye, A.; Rolando, M.; Flatau, G.; Lemichez, E.; Rapp, U.R. Isoform-specific Interaction of C-RAF with Mitochondria. J. Biol. Chem. 2008, 283, 14857–14866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smalley, K.S.M.; Xiao, M.; Villanueva, J.; Nguyen, T.K.; Flaherty, K.T.; Letrero, R.; Van Belle, P.; E Elder, D.; Wang, Y.; Nathanson, K.; et al. CRAF inhibition induces apoptosis in melanoma cells with non-V600E BRAF mutations. Oncogene 2008, 28, 85–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sundaresan, N.R.; Gupta, M.; Kim, G.; Rajamohan, S.B.; Isbatan, A.; Gupta, M.P. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J. Clin. Investig. 2009, 119, 2758–2771. [Google Scholar] [CrossRef] [Green Version]
- Zhang, S.; Xiong, X.; Sun, Y. Functional characterization of SOX2 as an anticancer target. Signal Transduct. Target. Ther. 2020, 5, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Pietrobono, S.; Morandi, A.; Gagliardi, S.; Gerlini, G.; Borgognoni, L.; Chiarugi, P.; Arbiser, J.L.; Stecca, B. Down-Regulation of SOX2 Underlies the Inhibitory Effects of the Triphenylmethane Gentian Violet on Melanoma Cell Self-Renewal and Survival. J. Investig. Dermatol. 2016, 136, 2059–2069. [Google Scholar] [CrossRef] [Green Version]
- Weina, K.; Wu, H.; Knappe, N.; Orouji, E.; Novak, D.; Bernhardt, M.; Hüser, L.; Larribere, L.; Umansky, V.; Gebhardt, C.; et al. TGF-βinduces SOX2 expression in a time-dependent manner in human melanoma cells. Pigment. Cell Melanoma Res. 2016, 29, 453–458. [Google Scholar] [CrossRef] [PubMed]
- Santini, R.; Pietrobono, S.; Pandolfi, S.; Montagnani, V.; Damico, M.; Penachioni, J.Y.; Vinci, M.C.; Borgognoni, L.; Stecca, B. SOX2 regulates self-renewal and tumorigenicity of human melanoma-initiating cells. Oncogene 2014, 33, 4697–4708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Girouard, S.D.; Laga, A.C.; Mihm, M.C.; Scolyer, R.A.; Thompson, J.F.; Zhan, Q.; Widlund, H.R.; Lee, C.-W.; Murphy, G.F. SOX2 contributes to melanoma cell invasion. Lab. Investig. 2012, 92, 362–370. [Google Scholar] [CrossRef] [Green Version]
- Schaefer, S.M.; Segalada, C.; Cheng, P.; Bonalli, M.; Parfejevs, V.; Levesque, M.P.; Dummer, R.; Nicolis, S.K.; Sommer, L. Sox2 is dispensable for primary melanoma and metastasis formation. Oncogene 2017, 36, 4516–4524. [Google Scholar] [CrossRef] [Green Version]
- Laga, A.C.; Lai, C.-Y.; Zhan, Q.; Huang, S.J.; Velazquez, E.F.; Yang, Q.; Hsu, M.-Y.; Murphy, G.F. Expression of The Embryonic Stem Cell Transcription Factor SOX2 in Human Skin: Relevance to Melanocyte and Merkel Cell Biology. Am. J. Pathol. 2010, 176, 903–913. [Google Scholar] [CrossRef] [Green Version]
- Novak, D.; Hüser, L.; Elton, J.J.; Umansky, V.; Altevogt, P.; Utikal, J. SOX2 in development and cancer biology. Semin. Cancer Biol. 2020, 67, 74–82. [Google Scholar] [CrossRef]
- Campbell, K.J.; Dhayade, S.; Ferrari, N.; Sims, A.; Johnson, E.; Mason, S.; Dickson, A.; Ryan, K.M.; Kalna, G.; Edwards, J.; et al. MCL-1 is a prognostic indicator and drug target in breast cancer. Cell Death Dis. 2018, 9, 1–14. [Google Scholar] [CrossRef]
- Abdulghani, J.; Gokare, P.; Gallant, J.-N.; Dicker, D.; Whitcomb, T.; Cooper, T.; Liao, J.; Derr, J.; Liu, J.; Goldenberg, D.; et al. Sorafenib and Quinacrine Target Anti-Apoptotic Protein MCL1: A Poor Prognostic Marker in Anaplastic Thyroid Cancer (ATC). Clin. Cancer Res. 2016, 22, 6192–6203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, K.; O’Neill, K.L.; Li, J.; Zhou, W.; Han, N.; Pang, X.; Wu, W.; Struble, L.; Borgstahl, G.; Liu, Z.; et al. BH3-only proteins target BCL-xL/MCL-1, not BAX/BAK, to initiate apoptosis. Cell Res. 2019, 29, 942–952. [Google Scholar] [CrossRef]
- Cook, S.J.; Stuart, K.; Gilley, R.; Sale, M.J. Control of cell death and mitochondrial fission by ERK1/2 MAP kinase signalling. FEBS J. 2017, 284, 4177–4195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sale, M.J.; Minihane, E.; Monks, N.R.; Gilley, R.; Richards, F.M.; Schifferli, K.P.; Andersen, C.L.; Davies, E.J.; Vicente, M.A.; Ozono, E.; et al. Targeting melanoma’s MCL1 bias unleashes the apoptotic potential of BRAF and ERK1/2 pathway inhibitors. Nat. Commun. 2019, 10, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bolomsky, A.; Vogler, M.; Köse, M.C.; Heckman, C.A.; Ehx, G.; Ludwig, H.; Caers, J. MCL-1 inhibitors, fast-lane development of a new class of anti-cancer agents. J. Hematol. Oncol. 2020, 13, 1–19. [Google Scholar] [CrossRef]
- Domina, A.M.; Vrana, J.; Gregory, M.; Hann, S.R.; Craig, R.W. MCL1 is phosphorylated in the PEST region and stabilized upon ERK activation in viable cells, and at additional sites with cytotoxic okadaic acid or taxol. Oncogene 2004, 23, 5301–5315. [Google Scholar] [CrossRef] [Green Version]
- McKee, C.S.; Hill, D.S.; Redfern, C.P.F.; Armstrong, J.L.; Lovat, P.E. Oncogenic BRAF signalling increases Mcl-1 expression in cutaneous metastatic melanoma. Exp. Dermatol. 2013, 22, 767–769. [Google Scholar] [CrossRef] [Green Version]
- Dai, W.; Liu, S.; Wang, S.; Zhao, L.; Yang, X.; Zhou, J.; Wang, Y.; Zhang, J.; Ding, K.; Li, Y.; et al. Activation of transmembrane receptor tyrosine kinase DDR1-STAT3 cascade by extracellular matrix remodeling promotes liver metastatic colonization in uveal melanoma. Signal Transduct. Target. Ther. 2021, 6, 1–16. [Google Scholar] [CrossRef]
- Lee, E.F.; Harris, T.J.; Tran, S.; Evangelista, M.; Arulananda, S.; John, T.; Ramnac, C.; Hobbs, C.; Zhu, H.; Gunasingh, G.; et al. BCL-XL and MCL-1 are the key BCL-2 family proteins in melanoma cell survival. Cell Death Dis. 2019, 10, 1–14. [Google Scholar] [CrossRef]
- Zang, H.; Qian, G.; Arbiser, J.; Owonikoko, T.K.; Ramalingam, S.S.; Fan, S.; Sun, S. Overcoming acquired resistance of EGFR-mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib, with the natural product honokiol. Mol. Oncol. 2020, 14, 882–895. [Google Scholar] [CrossRef] [Green Version]
- Elgendy, M.; Cirò, M.; Hosseini, A.; Weiszmann, J.; Mazzarella, L.; Ferrari, E.; Cazzoli, R.; Curigliano, G.; DeCensi, A.; Bonanni, B.; et al. Combination of Hypoglycemia and Metformin Impairs Tumor Metabolic Plasticity and Growth by Modulating the PP2A-GSK3β-MCL-1 Axis. Cancer Cell 2019, 35, 798–815.e5. [Google Scholar] [CrossRef]
- Guzzo, G.; Sciacovelli, M.; Bernardi, P.; Rasola, A. Inhibition of succinate dehydrogenase by the mitochondrial chaperone TRAP1 has anti-oxidant and anti-apoptotic effects on tumor cells. Oncotarget 2014, 5, 11897–11908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hua, G.; Zhang, Q.; Fan, Z. Heat Shock Protein 75 (TRAP1) Antagonizes Reactive Oxygen Species Generation and Protects Cells from Granzyme M-mediated Apoptosis. J. Biol. Chem. 2007, 282, 20553–20560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, S.; Wang, X.; Gan, S.; Tang, X.; Kang, X.; Zhu, S. The Mitochondrial Chaperone TRAP1 as a Candidate Target of Oncotherapy. Front. Oncol. 2021, 10. [Google Scholar] [CrossRef]
- Yoshida, S.; Tsutsumi, S.; Muhlebach, G.; Sourbier, C.; Lee, M.-J.; Lee, S.; Vartholomaiou, E.; Tatokoro, M.; Beebe, K.; Miyajima, N.; et al. Molecular chaperone TRAP1 regulates a metabolic switch between mitochondrial respiration and aerobic glycolysis. Proc. Natl. Acad. Sci. USA 2013, 110, E1604–E1612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Purushottam Dharaskar, S.; Paithankar, K.; Kanugovi Vijayavittal, A.; Shabbir Kara, H.; Amere Subbarao, S. Mitochondrial chaperone, TRAP1 modulates mitochondrial dynamics and promotes tumor metastasis. Mitochondrion 2020, 54, 92–101. [Google Scholar] [CrossRef]
- Selak, M.A.; Armour, S.M.; MacKenzie, E.D.; Boulahbel, H.; Watson, D.G.; Mansfield, K.D.; Pan, Y.; Simon, M.C.; Thompson, C.B.; Gottlieb, E. Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-α prolyl hydroxylase. Cancer Cell 2005, 7, 77–85. [Google Scholar] [CrossRef] [Green Version]
- Masgras, I.; Sanchez-Martin, C.; Colombo, G.; Rasola, A. The Chaperone TRAP1 As a Modulator of the Mitochondrial Adaptations in Cancer Cells. Front. Oncol. 2017, 7, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maddalena, F.; Condelli, V.; Matassa, D.S.; Pacelli, C.; Scrima, R.; Lettini, G.; Li Bergolis, V.; Pietrafesa, M.; Crispo, F.; Piscazzi, A.; et al. TRAP1 enhances Warburg metabolism through modulation of PFK1 expression/activity and favors resistance to EGFR inhibitors in human colorectal carcinomas. Mol. Oncol. 2020, 14, 3030–3047. [Google Scholar] [CrossRef]
- Maddalena, F.; Sisinni, L.; Lettini, G.; Condelli, V.; Matassa, D.S.; Piscazzi, A.; Amoroso, M.R.; La Torre, G.; Esposito, F.; Landriscina, M. Resistance to paclitxel in breast carcinoma cells requires a quality control of mitochondrial antiapoptotic proteins by TRAP1. Mol. Oncol. 2013, 7, 895–906. [Google Scholar] [CrossRef] [PubMed]
- Matassa, D.S.; Agliarulo, I.; Avolio, R.; Landriscina, M.; Esposito, F. TRAP1 Regulation of Cancer Metabolism: Dual Role as Oncogene or Tumor Suppressor. Genes 2018, 9, 195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, G.; Frederick, D.T.; Wu, L.; Wei, Z.; Krepler, C.; Srinivasan, S.; Chae, Y.C.; Xu, X.; Choi, H.; Dimwamwa, E.; et al. Targeting mitochondrial biogenesis to overcome drug resistance to MAPK inhibitors. J. Clin. Investig. 2016, 126, 1834–1856. [Google Scholar] [CrossRef] [PubMed]
- Basit, F.; Van Oppen, L.M.P.E.; Schöckel, L.; Bossenbroek, H.M.; Van Emst-de Vries, S.E.; Hermeling, J.C.W.; Grefte, S.; Kopitz, C.; Heroult, M.; Willems, P.H.; et al. Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase leading to necroptosis and ferroptosis in melanoma cells. Cell Death Dis. 2017, 8, e2716. [Google Scholar] [CrossRef] [PubMed]
- Li, X.-T.; Li, Y.-S.; Shi, Z.-Y.; Guo, X.-L. New insights into molecular chaperone TRAP1 as a feasible target for future cancer treatments. Life Sci. 2020, 254, 117737. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Martin, C.; Menon, D.; Moroni, E.; Ferraro, M.; Masgras, I.; Elsey, J.; Arbiser, J.L.; Colombo, G.; Rasola, A. Honokiol Bis-Dichloroacetate Is a Selective Allosteric Inhibitor of the Mitochondrial Chaperone TRAP1. Antioxidants Redox Signal. 2021, 34, 505–516. [Google Scholar] [CrossRef]
- Lionarons, D.A.; Hancock, D.C.; Rana, S.; East, P.; Moore, C.; Murillo, M.M.; Carvalho, J.; Spencer-Dene, B.; Herbert, E.; Stamp, G.; et al. RAC1P29S Induces a Mesenchymal Phenotypic Switch via Serum Response Factor to Promote Melanoma Development and Therapy Resistance. Cancer Cell 2019, 36, 68–83.e9. [Google Scholar] [CrossRef] [Green Version]
- Medjkane, S.; Perez-Sanchez, C.; Gaggioli, C.; Sahai, E.; Treisman, R. Myocardin-related transcription factors and SRF are required for cytoskeletal dynamics and experimental metastasis. Nat. Cell Biol. 2009, 11, 257–268. [Google Scholar] [CrossRef]
- Moore, A.S.; Wong, Y.C.; Simpson, C.L.; Holzbaur, E.L.F. Dynamic actin cycling through mitochondrial subpopulations locally regulates the fission–fusion balance within mitochondrial networks. Nat. Commun. 2016, 7, 12886. [Google Scholar] [CrossRef]
- Olson, E.N.; Nordheim, A. Linking actin dynamics and gene transcription to drive cellular motile functions. Nat. Rev. Mol. Cell Biol. 2010, 11, 353–365. [Google Scholar] [CrossRef] [Green Version]
- Youle, R.J.; van der Bliek, A.M. Mitochondrial Fission, Fusion, and Stress. Science 2012, 337, 1062–1065. [Google Scholar] [CrossRef] [Green Version]
- Dal Yontem, F.; Kim, S.-H.; Ding, Z.; Grimm, E.; Ekmekcioglu, S.; Akcakaya, H. Mitochondrial dynamic alterations regulate melanoma cell progression. J. Cell. Biochem. 2019, 120, 2098–2108. [Google Scholar] [CrossRef]
- Singleton, K.R.; Crawford, L.; Tsui, E.; Manchester, H.E.; Maertens, O.; Liu, X.; Liberti, M.; Magpusao, A.N.; Stein, E.M.; Tingley, J.P.; et al. Melanoma Therapeutic Strategies that Select against Resistance by Exploiting MYC-Driven Evolutionary Convergence. Cell Rep. 2017, 21, 2796–2812. [Google Scholar] [CrossRef] [Green Version]
- Dong, Y.; Tu, R.; Liu, H.; Qing, G. Regulation of cancer cell metabolism: Oncogenic MYC in the driver’s seat. Signal Transduct. Target. Ther. 2020, 5, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Alhazzazi, T.Y.; Kamarajan, P.; Verdin, E.; Kapila, Y.L. SIRT3 and cancer: Tumor promoter or suppressor? Biochim. Biophys. Acta (BBA) Bioenerg. 2011, 1816, 80–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kong, X.; Wang, R.; Xue, Y.; Liu, X.; Zhang, H.; Chen, Y.; Fang, F.; Chang, Y. Sirtuin 3, a New Target of PGC-1α, Plays an Important Role in the Suppression of ROS and Mitochondrial Biogenesis. PLoS ONE 2010, 5, e11707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mastropasqua, F.; Girolimetti, G.; Shoshan, M. PGC1α: Friend or Foe in Cancer? Genes 2018, 9, 48. [Google Scholar] [CrossRef] [Green Version]
- Torrens-Mas, M.; Oliver, J.O.; Roca, P.; Sastre-Serra, J. SIRT3: Oncogene and Tumor Suppressor in Cancer. Cancers 2017, 9, 90. [Google Scholar] [CrossRef] [Green Version]
- Torrens-Mas, M.; Pons, D.G.; Sastre-Serra, J.; Oliver, J.; Roca, P. SIRT3 Silencing Sensitizes Breast Cancer Cells to Cytotoxic Treatments Through an Increment in ROS Production. J. Cell. Biochem. 2017, 118, 397–406. [Google Scholar] [CrossRef]
- Alhazzazi, T.Y.; Kamarajan, P.; Verdin, E.; Kapila, Y.L. Sirtuin-3 (SIRT3) and the Hallmarks of Cancer. Genes Cancer 2013, 4, 164–171. [Google Scholar] [CrossRef] [Green Version]
- Cheng, Y.; Fu, L.L.; Wen, X.; Wang, X.Y.; Liu, J.; Huang, J. Sirtuin-3 (SIRT3), a therapeutic target with oncogenic and tumor-suppressive function in cancer. Cell Death Dis. 2014, 5, e1047. [Google Scholar] [CrossRef] [Green Version]
- Jung, S.-N.; Yang, W.K.; Kim, J.; Kim, H.S.; Kim, E.J.; Yun, H.; Park, H.; Kim, S.S.; Choe, W.; Kang, I.; et al. Reactive oxygen species stabilize hypoxia-inducible factor-1 alpha protein and stimulate transcriptional activity via AMP-activated protein kinase in DU145 human prostate cancer cells. Carcinogenesis 2008, 29, 713–721. [Google Scholar] [CrossRef] [Green Version]
- Singh, C.K.; George, J.; Chhabra, G.; Nihal, M.; Chang, H.; Ahmad, N. Genetic Manipulation of Sirtuin 3 Causes Alterations of Key Metabolic Regulators in Melanoma. Front. Oncol. 2021, 11. [Google Scholar] [CrossRef] [PubMed]
- Bonner, M.Y.; Karlsson, I.; Rodolfo, M.; Arnold, R.S.; Vergani, E.; Arbiser, J.L. Honokiol bis-dichloroacetate (Honokiol DCA) demonstrates activity in vemurafenib-resistant melanoma in vivo. Oncotarget 2016, 7, 12857–12868. [Google Scholar] [CrossRef]
- Qiao, A.; Wang, K.; Yuan, Y.; Guan, Y.; Ren, X.; Li, L.; Chen, X.; Li, F.; Chen, A.F.; Zhou, J.; et al. Sirt3-mediated mitophagy protects tumor cells against apoptosis under hypoxia. Oncotarget 2016, 7, 43390–43400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gonzalez Herrera, K.N.; Zaganjor, E.; Ishikawa, Y.; Spinelli, J.B.; Yoon, H.; Lin, J.-R.; Satterstrom, F.K.; Ringel, A.; Mulei, S.; Souza, A.; et al. Small-Molecule Screen Identifies De Novo Nucleotide Synthesis as a Vulnerability of Cells Lacking SIRT3. Cell Rep. 2018, 22, 1945–1955. [Google Scholar] [CrossRef] [Green Version]
- Wang, Q.; Beaumont, K.A.; Otte, N.J.; Font, J.; Bailey, C.G.; Van Geldermalsen, M.; Sharp, D.M.; Tiffen, J.C.; Ryan, R.M.; Jormakka, M.; et al. Targeting glutamine transport to suppress melanoma cell growth. Int. J. Cancer 2014, 135, 1060–1071. [Google Scholar] [CrossRef] [PubMed]
- Robertson, G.P. Functional and therapeutic significance of Akt deregulation in malignant melanoma. Cancer Metastasis Rev. 2005, 24, 273–285. [Google Scholar] [CrossRef] [PubMed]
- Theodosakis, N.; Micevic, G.; Kelly, D.P.; Bosenberg, M. Mitochondrial function in melanoma. Arch. Biochem. Biophys. 2014, 563, 56–59. [Google Scholar] [CrossRef]
- Bonner, M.Y.; Arbiser, J.L. Targeting NADPH oxidases for the treatment of cancer and inflammation. Cell. Mol. Life Sci. 2012, 69, 2435–2442. [Google Scholar] [CrossRef] [Green Version]
- Ciołczyk-Wierzbicka, D.; Gil, D.; Zarzycka, M.; Laidler, P. mTOR inhibitor everolimus reduces invasiveness of melanoma cells. Hum. Cell 2020, 33, 88–97. [Google Scholar] [CrossRef] [Green Version]
- Sathe, A.; Chalaud, G.; Oppolzer, I.; Wong, K.Y.; Von Busch, M.; Schmid, S.C.; Tong, Z.; Retz, M.; Gschwend, J.E.; Schulz, W.; et al. Parallel PI3K, AKT and mTOR inhibition is required to control feedback loops that limit tumor therapy. PLoS ONE 2018, 13, e0190854. [Google Scholar] [CrossRef] [Green Version]
- Kim, L.C.; Cook, R.S.; Chen, J. mTORC1 and mTORC2 in cancer and the tumor microenvironment. Oncogene 2017, 36, 2191–2201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, S. Role of mTOR Signaling in Tumor Cell Motility, Invasion and Metastasis. Curr. Protein Pept. Sci. 2011, 12, 30–42. [Google Scholar] [CrossRef] [PubMed]
- Mercurio, L.; Albanesi, C.; Madonna, S. Recent Updates on the Involvement of PI3K/AKT/mTOR Molecular Cascade in the Pathogenesis of Hyperproliferative Skin Disorders. Front. Med. 2021, 8, 665647. [Google Scholar] [CrossRef]
- Land, S.; Tee, A. Hypoxia-inducible Factor 1α Is Regulated by the Mammalian Target of Rapamycin (mTOR) via an mTOR Signaling Motif. J. Biol. Chem. 2007, 282, 20534–20543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arbiser, J.L.; Kau, T.; Konar, M.; Narra, K.; Ramchandran, R.; Summers, S.A.; Vlahos, C.J.; Ye, K.; Perry, B.N.; Matter, W.; et al. Solenopsin, the alkaloidal component of the fire ant (Solenopsis invicta), is a naturally occurring inhibitor of phosphatidylinositol-3-kinase signaling and angiogenesis. Blood 2007, 109, 560–565. [Google Scholar] [CrossRef] [Green Version]
- Gottlob, K.; Majewski, N.; Kennedy, S.; Kandel, E.; Robey, R.B.; Hay, N. Inhibition of early apoptotic events by Akt/PKB is dependent on the first committed step of glycolysis and mitochondrial hexokinase. Genes Dev. 2001, 15, 1406–1418. [Google Scholar] [CrossRef] [Green Version]
- Wang, B.; Zhang, W.; Zhang, G.; Kwong, L.; Lu, H.; Tan, J.; Sadek, N.; Xiao, M.; Zhang, J.; Labrie, M.; et al. Targeting mTOR signaling overcomes acquired resistance to combined BRAF and MEK inhibition in BRAF-mutant melanoma. Oncogene 2021, 40, 5590–5599. [Google Scholar] [CrossRef]
- Subbiah, V.; Sen, S.; Hess, K.R.; Janku, F.; Hong, D.S.; Khatua, S.; Karp, D.D.; Munoz, J.; Falchook, G.S.; Groisberg, R.; et al. Phase I Study of the BRAF Inhibitor Vemurafenib in Combination with the Mammalian Target of Rapamycin Inhibitor Everolimus in Patients With BRAF-Mutated Malignancies. JCO Precis. Oncol. 2018, 2, 1–12. [Google Scholar] [CrossRef]
- Wan, X.; Harkavy, B.; Shen, N.; Grohar, P.J.; Helman, L.J. Rapamycin induces feedback activation of Akt signaling through an IGF-1R-dependent mechanism. Oncogene 2006, 26, 1932–1940. [Google Scholar] [CrossRef] [Green Version]
- Chamcheu, J.C.; Roy, T.; Uddin, M.B.; Banang-Mbeumi, S.; Chamcheu, R.-C.N.; Walker, A.L.; Liu, Y.-Y.; Huang, S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019, 8, 803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abildgaard, C.; Rizza, S.; Christiansen, H.; Schmidt, S.; Dahl, C.; Abdul-Al, A.; Christensen, A.; Filomeni, G.; Guldberg, P. Screening of metabolic modulators identifies new strategies to target metabolic reprogramming in melanoma. Sci. Rep. 2021, 11, 1–12. [Google Scholar] [CrossRef]
- Liu, D.; Hu, J.; Agorreta, J.; Cesario, A.; Zhang, Y.; Harris, A.L.; Gatter, K.; Pezzella, F. Tumor necrosis factor receptor-associated protein 1(TRAP1) regulates genes involved in cell cycle and metastases. Cancer Lett. 2010, 296, 194–205. [Google Scholar] [CrossRef] [PubMed]
- Cabrita, R.; Mitra, S.; Sanna, A.; Ekedahl, H.; Lövgren, K.; Olsson, H.; Ingvar, C.; Isaksson, K.; Lauss, M.; Carneiro, A.; et al. The Role of PTEN Loss in Immune Escape, Melanoma Prognosis and Therapy Response. Cancers 2020, 12, 742. [Google Scholar] [CrossRef] [Green Version]
- Spranger, S.; Gajewski, T.F. Mechanisms of Tumor Cell–Intrinsic Immune Evasion. Annu. Rev. Cancer Biol. 2018, 2, 213–228. [Google Scholar] [CrossRef]
- Wu, R.; Wang, C.; Li, Z.; Xiao, J.; Li, C.; Wang, X.; Kong, P.; Cao, J.; Huang, F.; Huang, Y.; et al. SOX2 promotes resistance of melanoma with PD-L1 high expression to T-cell-mediated cytotoxicity that can be reversed by SAHA. J. Immunother. Cancer 2020, 8, e001037. [Google Scholar] [CrossRef]
- Staszkiewicz, J.; Power, R.A.; Harkins, L.L.; Barnes, C.W.; Strickler, K.L.; Rim, J.S.; Bondioli, K.R.; Eilersten, K.J. Silencing Histone Deacetylase–Specific Isoforms Enhances Expression of Pluripotency Genes in Bovine Fibroblasts. Cell. Reprogramming 2013, 15, 397–404. [Google Scholar] [CrossRef] [Green Version]
- Hepburn, A.C.; Steele, R.E.; Veeratterapillay, R.; Wilson, L.; Kounatidou, E.E.; Barnard, A.; Berry, P.; Cassidy, J.R.; Moad, M.; El-Sherif, A.; et al. The induction of core pluripotency master regulators in cancers defines poor clinical outcomes and treatment resistance. Oncogene 2019, 38, 4412–4424. [Google Scholar] [CrossRef]
- Caunt, C.J.; Sale, M.J.; Smith, P.D.; Cook, S.J. MEK1 and MEK2 inhibitors and cancer therapy: The long and winding road. Nat. Rev. Cancer 2015, 15, 577–592. [Google Scholar] [CrossRef]
Factor | Function | Main Roles in Melanoma Plasticity | Discussed Therapeutic Approaches |
---|---|---|---|
BRAF and RAF1 (CRAF) | Protein kinases in MAPK signaling pathway | - Activation of MAPK upregulates HIF1A activity, facilitating a shift to aerobic glycolysis (also known as the Warburg Effect) [5] - Suppresses MITF and PGC1A, factors that drive oxidative phosphorylation [34] - BRAF mutations, particularly BRAFV600E in melanoma, amplify these effects [5] - CRAF activation and other mutations emerge in resistance to treatment with BRAF inhibitors to reactivate MAPK [35] | Vemurafenib Development: FDA-approved for treating BRAFV600E melanoma Mechanism of action: selectively inhibits mutated BRAFV600E kinase [36] Dabrafenib Development: FDA-approved for treating BRAFV600E melanoma Mechanism of action: ATP-competitive binding to BRAF kinase [37] Binimetinib Development: FDA-approved for treating BRAFV600E melanoma in combination with encorafenib, a BRAF inhibitor Mechanism of action: selective MAP2K1/2 inhibitor [38] |
SOX2 | Transcription factor with multiple roles including maintaining pluripotency | - Enables plasticity in response to acidic extracellular environment, shifting metabolism to one favoring oxidative phosphorylation - Inhibits HIF1A by binding to its promotor [20] | Gefitnib Development: approved by FDA for treating non-small cell lung cancer with EGFR mutations Mechanism of action: selective small-molecule inhibitor of EGFR (a SOX2 upstream activator) [39] |
MCL1 | Anti-apoptotic protein | - Prevents apoptosis and keeps mitochondria intact - Matrix isoform promotes mitochondrial fusion and ATP production, enhancing oxidative phosphorylation efficiency [31] | AZD5991 Development: phase 1 clinical trial paused (ClinicalTrials.gov identifier: NCT03218683) Mechanism of action: selective MCL1 protein inhibitor, leading to increased apoptosis [40] |
TRAP1 | Mitochondrial heat shock protein, protein chaperone | - Inhibits complexes II and IV of the electron transport chain and SRC, downregulating oxidative phosphorylation - Stabilizes HIF1A via accumulation of succinate [41] - Activates BRAF signaling pathway [42] | Gamitrinib Development: in phase 1 clinical trial (ClinicalTrials.gov identifier: NCT04827810) Mechanism of action: small-molecule, mitochondrial-targeted HSP90 inhibitor [43] Honokiol bis-dichloroacetate Development: synthesized from the natural compound honokiol Mechanism of action: allosteric inhibitor of TRAP1 [41] |
RHOA and SRF | Signaling regulates actin dynamics and other pathways | - Induces polymerization of actin, resulting in subsequent mitochondrial fission [44] - Assists in using glutamine as a source of energy [44] | C3 Transferase Development: bacterial exoenzyme from clostridium botulinum [45] Mechanism of action: irreversible inactivation of RHOA GTP-ase protein [45] |
SIRT3 | Histone deacetylase protein | - Activates complexes I and II of the electron transport chain - Decreases oxidative stress and reactive oxygen species [32] - Indirectly destabilizes HIF1A [46] | Hexafluoro Development: Analog of natural compound honokiol Mechanism of action: induction of SIRT3 [47] |
PTEN | Tumor suppressor that dephosphorylates PIP3 | - Acts to maintain normal metabolism via the downregulation of PI3K/AKT1/mTORC1 signaling pathway - Deactivating mutations in PTEN often found in melanoma [48] | Therapies are aimed towards decreasing PI3K/AKT1/mTORC1 signaling, which is the role of PTEN |
AKT1 | Oncogene and key regulator of cellular growth | - Increased PI3K/AKT1/mTORC1 pathway signaling upregulates HIF1A, contributing to a glycolytic metabolism [49] - Increases coupling of efficiency of oxidative phosphorylation and glycolysis, enhancing bioenergetics in general [50] | Everolimus Development: FDA-approved for treating a variety of malignancies Mechanism of action: binds cyclophilin FKBP-12 which then binds mTOR, inhibiting mTORC1 complex formation [51] Rapamycin Development: natural compound from Streptomyces hygroscopicus, FDA approved for immunosupression Mechanism of action: forms complex with FKBP-12, which allostericaly inhibits mTORC1 [52] NVP-BEZ235 Development: in phase I/II clinical trial (ClinicalTrials.gov identifier: NCT00620594) Mechanism of action: ATP-competitive PI3K and mTOR inhibitor [53] Nexrutine Development: plant extract from Phellodendron trees [54] Mechanism of action: induction of oxidative stress [55] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Huang, C.; Radi, R.H.; Arbiser, J.L. Mitochondrial Metabolism in Melanoma. Cells 2021, 10, 3197. https://doi.org/10.3390/cells10113197
Huang C, Radi RH, Arbiser JL. Mitochondrial Metabolism in Melanoma. Cells. 2021; 10(11):3197. https://doi.org/10.3390/cells10113197
Chicago/Turabian StyleHuang, Christina, Rakan H. Radi, and Jack L. Arbiser. 2021. "Mitochondrial Metabolism in Melanoma" Cells 10, no. 11: 3197. https://doi.org/10.3390/cells10113197
APA StyleHuang, C., Radi, R. H., & Arbiser, J. L. (2021). Mitochondrial Metabolism in Melanoma. Cells, 10(11), 3197. https://doi.org/10.3390/cells10113197