Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2152 2022-04-03 01:03:55 |
2 update layout and references -43 word(s) 2109 2022-04-06 04:17:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Indra, A.; Carpenter, E.; Becker, A. NRF2 and Key Transcriptional Targets. Encyclopedia. Available online: https://encyclopedia.pub/entry/21329 (accessed on 27 July 2024).
Indra A, Carpenter E, Becker A. NRF2 and Key Transcriptional Targets. Encyclopedia. Available at: https://encyclopedia.pub/entry/21329. Accessed July 27, 2024.
Indra, Arup, Evan Carpenter, Alyssa Becker. "NRF2 and Key Transcriptional Targets" Encyclopedia, https://encyclopedia.pub/entry/21329 (accessed July 27, 2024).
Indra, A., Carpenter, E., & Becker, A. (2022, April 03). NRF2 and Key Transcriptional Targets. In Encyclopedia. https://encyclopedia.pub/entry/21329
Indra, Arup, et al. "NRF2 and Key Transcriptional Targets." Encyclopedia. Web. 03 April, 2022.
NRF2 and Key Transcriptional Targets
Edit

Melanocytes are dendritic, pigment-producing cells located in the skin and are responsible for its protection against the deleterious effects of solar ultraviolet radiation (UVR), which include DNA damage and elevated reactive oxygen species (ROS). They do so by synthesizing photoprotective melanin pigments and distributing them to adjacent skin cells (e.g., keratinocytes). However, melanocytes encounter a large burden of oxidative stress during this process, due to both exogenous and endogenous sources.To protect themselves, they utilize numerous antioxidant systems to reduce the amount of reactive oxygen and nitrogen species present in the cell and this activity then contributes towards the prevention of cancer formation. However, after the formation of melanoma these same antioxidant systems are often coopted by the cancer in order to promote its uncontrolled growth and metastasis.

NRF2 glutathione thioredoxin peroxiredoxin

1. Introduction

Melanoma is the deadliest form of skin cancer and the leading cause of death due to skin disease [1]. It has exhibited an increased incidence in the United States, with an average annual percentage change (AAPC) of 2.2% in males and 2.3% in females between 2013 and 2017. Fortunately, the rate of death between 2013 and 2018 has started to trend downward with an AAPC of −5.7% and −4.4% in males and females, respectively [2]. These decreases are likely due to a combination of more successful prevention and early detection strategies, as well as the development of increasingly efficacious therapies such as targeted BRAF or MEK inhibitors, immunotherapy, and all combinations thereof.
Melanomas arise from the malignant transformation of melanocytes, which are dendritic, pigment-producing cells located in the basal layer of the epidermis, hair follicles, inner ear, and uvea of the eye [3]. Normally, melanocytes are tasked with the protection of adjacent skin cells from solar ultraviolet radiation (UVR) by synthesizing and distributing photoprotective melanin pigment [4]. However, melanocytes are still susceptible to UV-induced DNA damage, which can lead to the formation of key driver mutations in oncogenes, including proliferation/survival regulators such as BRAF and NRAS and cell-cycle regulators such as CDK4 [5][6][7]. Additionally, UVR also leads to the rapid accumulation of passenger mutations, which might not be drivers of melanomagenesis themselves, but collectively serve to cause heightened tumor heterogeneity that complicates treatment [8]. Upon metastasis, melanomas are highly aggressive, often migrating to the brain and lungs, and five-year survival rates are 19% for patients diagnosed with stage IV disease [9].
Current treatment modalities for melanoma include surgical resection, radiotherapy, chemotherapy, immunotherapy, biochemotherapy, and targeted therapy [10]. These agents may be used alone or in combination depending on the tumor’s stage, location, and genetic profile, as well as the patient’s goals for treatment. Earlier stage disease (stages I-IIIB) can usually be fully resected [11]. Surgical margins depend on the depth of invasion: 0.5 cm for in situ melanomas, 1 cm for tumors ≤2 mm in thickness, and 2 cm for tumors >2 mm in thickness [10][12]. Surgical excision (metastasectomy) is also part of the standard of care in the treatment of a solitary site of metastasis. Radiotherapy, although rarely used in treatment of the primary tumor, can also be used for metastatic lesions, especially of the skin, bone, and brain [13]. In high-risk melanomas (stages IIC-IV), adjuvant therapy is recommended following a complete resection of the primary tumor [14].
Untargeted chemotherapies such as dacarbazine and temozolomide were the first pharmaceutical options available for advanced melanomas; however, they have been largely unsuccessful and typically do not improve overall survival [15]. Resistance to apoptosis is thought to be the major mechanism of chemotherapeutic drug failure in melanomas [16]. Untargeted chemotherapies have largely been replaced by newer options but may still have some utility in palliative treatment [15].
Approximately 70% of patients with cutaneous melanoma have oncogenic mutations in signaling pathways involving cell proliferation; targeted therapeutic strategies use small molecule inhibitors or antibodies that affect these mutated proteins [17]. FDA-approved targeted therapies for melanoma include BRAFV600E and MEK inhibitors. Compared to chemotherapy, BRAF inhibitors improve clinical response rates, progression-free survival, and overall survival in metastatic melanoma patients with BRAF mutations [18][19]. However, their efficacy can be limited due to the rapid development of multiple mechanisms of resistance [20]. In addition to possible drug resistance, BRAF inhibitor monotherapy can also be associated with cutaneous toxicities secondary to paradoxical MAPK pathway activation [19][21]. Combined therapeutic strategies are utilized to attempt to overcome these issues. Targeting signals downstream of BRAF, such as MEK, decreases MAPK-driven acquired resistance and toxicity, resulting in a greater duration of any response to therapy and fewer side effects [22][23]. While combined anti-BRAF/MEK therapy is initially highly effective in patients with melanomas harboring mutant BRAF, eventual resistance is not uncommon [24][25][26][27][28].
Improved understanding of the pathophysiology of the role of the immune system in tumorigenesis has led to the development and approval of several immunotherapies that show promising efficacy [29][30][31]. Combination immunotherapies show even greater promise; the CheckMate 067 trial demonstrated that, among patients with advanced melanoma, sustained long-term survival was seen in a greater percentage of patients who received nivolumab (anti-PD-1) plus ipilimumab (anti-CTLA-4) compared to ipilimumab alone [32][33]. Remarkably, nivolumab/ipilimumab combination therapy is the only currently available treatment for metastatic melanoma with a median overall survival greater than five years [33]. While immunotherapies show great promise, it is possible for primary and acquired resistance to occur, especially in the case of monotherapy, which may be due to the lack of recognition by T-cells, interactions with the cancer immune cycle, and/or complex signaling pathways within the tumor microenvironment [34][35][36].
Biochemotherapy combines the apoptotic and DNA-damaging mechanisms of chemotherapy with the immunomodulatory effects of immunotherapy [10]. Compared to chemotherapy monotherapy, biochemotherapy improves median progression-free survival but does not improve overall survival and can be associated with severe toxicity [10]. Therefore, further understanding of the metastatic progression of melanomas could provide insight into novel therapies that contribute to prevention and treatment.
Oxidative stress is an important aspect involved in normal melanocyte physiology, melanoma initiation, and disease progression, as well as a potential source for novel therapies going forward. Even in the absence of disease, melanocytes are subject to a significant degree of oxidative stress that originates both exogenously, from sources such as UVR, and endogenously, from metabolic pathways including mitochondrial respiration and melanogenesis [37][38][39]. These sources of oxidative stress can then contribute towards the regulation of nuclear factor erythroid 2-like 2 (NFE2L2 encoding NRF2) related signaling pathways [40].
Notably, UV irradiation of melanocytes in vitro has been shown to induce the activation of NRF2 and the subsequent expression of antioxidant target genes including GCLM, GCLC, NQO1, and HMOX1 [41]. Furthermore, in vivo human skin UV irradiation has been shown to induce the expression of another NRF2 target, thioredoxin reductase 1, in melanocytic nevi [42]. Researchers have also shown, using a tissue-specific knockout mouse model, that melanocytic thioredoxin reductase 1 contributes to both melanocyte development and photobiology, as they observed pigmentation defects and elevated UV-mediated 8-oxo-2′-deoxyguanosine DNA damage in its absence [43]. Overall, NRF2 and its antioxidant target genes are important to normal melanocyte physiology and the response to UVR and act as tumor suppressors in this context.
As with many cancers, melanomas exhibit elevated oxidative stress both intracellularly as well as in the tumor microenvironment [44][45]. This elevation is due, in part, to altered metabolic pathways that cancer cells engage to sustain their dysregulated proliferation, which NRF2 contributes to [46][47][48]. Classically, increased oxidative stress has been held to simply contribute to melanoma initiation and progression by inducing a microenvironment conducive to tumor heterogeneity and metastasis. This is due to the increased mutational burden, as well as the activated signaling pathways associated with heightened tissue damage, such as inflammation [49]. It would then follow that exogenous antioxidant therapies should be highly efficacious in reducing proliferation and progression. However, it is currently understood that the role played by oxidative stress in melanomas is highly complex, as cancer cells establish a reprogrammed redox homeostasis that enables the tumor-promoting aspects of oxidative stress while avoiding its associated toxicity [50]. This is often seen by the activation of NRF2 in melanomas, particularly in those harboring KEAP1 mutations, during progression with the upregulation of key downstream redox regulatory factors including elements of the glutathione (GSH) system, thioredoxin (TRX) system, cysteine cycling, nicotinamide adenine dinucleotide phosphate (NADPH): quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HO-1), heat shock protein 70 (HSP70), and peroxiredoxins [51][52][53][54][55][56][57][58][59][60][61].

Furthermore, the complexity of this reprogrammed redox homeostasis also explains the lack in efficacy often exhibited by exogenous antioxidant treatments thus far [62]. Altogether, it is certain that further research into how oxidative stress contributes towards melanocyte physiology and melanoma pathophysiology is needed for potential anti- or pro-oxidant therapies to come to fruition. This review primarily focuses on the NRF2 pathway and potential transcriptional targets controlling redox signaling in melanoma.

2. Conclusion and Future Directions

In this entry researchers have highlighted how protective endogenous antioxidant systems are coopted during melanoma initiation and progression. The net effect of this cooption is an increased antioxidant capacity within melanoma cells which allows for their unrestrained proliferation, metastasis, and drug resistance [63]. Fortunately, this reliance on endogenous antioxidant systems for progression and metastasis reveals potential targets for novel therapies. This is seen in the glutathione antioxidant system where there is upregulation of GSH synthesis genes, increased recycling of GSH mediated by GSR, and increased expression of antioxidant genes associated with drug-resistance like GPX2, GPX4, and GSTs [54]. Furthermore, NRF2 contributes to the elevated expression of G6PD which is an important source for NADPH necessary for the activity of both glutathione and thioredoxin antioxidant systems [54]. With the thioredoxin antioxidant system, there is an upregulation of both TXN and TXNRD1 expression that contributes to PRDX function and the clearance of cellular free radicals [54]. Both of these systems are dependent on the amino acid cysteine which is imported through the cystine/glutamate antiporter xCT (system xc-) comprised in part by the NRF2 transcriptional target SLC7A11 that also has upregulated expression in melanoma [54].

Three potential anti-melanoma targets within these antioxidant systems could include (but are not limited to) glutathione synthesis, thioredoxin reductase enzymatic activity, and the cystine/cysteine cycle. A combination of these redox targets would then be combined with conventional melanoma therapies. For example, it has been previously shown that combined inhibition of GSH synthesis and cystine/cysteine cycling was the more effective in killing melanoma cells than either treatment alone [52]. This blockade of coopted antioxidant function could then be combined with MAPK inhibition or immunotherapy. Knockdown of thioredoxin reductase 1 alone in melanoma alone does not prevent metastasis, but does induce a dependency on glycolysis and a complete knockout of TXNRD1 in melanocytes increases nuclear localization of NRF2 and synthesis of GSH post-UVB irradiation [43][53]. Interestingly, acquired resistance to BRAF inhibition induces a shift to oxidative phosphorylation [64]. So, it would be interesting to determine the effect of combinatorial inhibition of MAPK signaling and thioredoxin reductase 1 would have on melanoma cells.

Another avenue to treatment through intracellular redox manipulation is through the targeting of mitochondria. Researchers recently performed a high-throughput screen to identify compounds effective against drug-resistant melanoma and found that compounds that target the electron transport chain were the most potent and others have studied different compounds that act similarly [65][66]. Inhibition of the electron transport chain leads to the accumulation of oxidative stress making it a pro-oxidant treatment [67]. Also promising are compounds capable of disrupting mitochondrial membrane potential as exhibited by natural product polyphenols acting as proton shuttles that interfere with the electrochemical gradient across the inner mitochondrial membrane [68][69]. Additionally, polyphenols can also inhibit the electron transport chain and thereby further increase the oxidative stress present in cancer cells.

It is also important to examine NRF2 and its transcriptional targets in the context of the immune response. Melanoma cell NRF2 has an indirect connection with an immune response through its negative regulation of the transcription factor RXRα where researchers have previously shown that knockout of RXRα and RXRβ in melanocytes alters the expression of cytokines/chemokines that recruit IFN-γ secreting immune cells [70]. However, targeting of NRF2 directly in the attempt to improve an antimelanoma immune response is not likely to be effective as it has been previously shown that NRF2-null mice have increased susceptibility to xenograft of B16-F10 melanoma cells possibly due in part to an impaired immune system [71]. These data indicate any intervention that might also compromise NRF2 function in nonmelanoma cancer-adjacent cells could be detrimental, and therefore, endogenous antioxidants downstream of NRF2/KEAP1/ARE signaling could be better targets to benefit immunotherapy. These targets could include thioredoxin secretion, heme oxygenase 1, and cell surface expression of heat shock protein 70 previously shown to play a role in the immune response to melanoma as discussed above.

It should be noted that there has been some efficacy seen in clinical studies investigating the role of exogenous antioxidants in the prevention of melanoma but its utility in treatment requires further detailed studies on intervention at different stages of melanoma progression and metastasis to better understand and subsequently avoid any unintended detrimental effects.

References

  1. Lim, H.W.; Collins, S.A.B.; Resneck, J.S., Jr.; Bolognia, J.L.; Hodge, J.A.; Rohrer, T.A.; Van Beek, M.J.; Margolis, D.J.; Sober, A.J.; Weinstock, M.A.; et al. The burden of skin disease in the United States. J. Am. Acad. Dermatol. 2017, 76, 958–972.
  2. Islami, F.; Ward, E.M.; Sung, H.; Cronin, K.A.; Tangka, F.K.L.; Sherman, R.L.; Zhao, J.; Anderson, R.N.; Henley, S.J.; Yabroff, K.R.; et al. Annual Report to the Nation on the Status of Cancer, Part 1: National Cancer Statistics. J. Natl. Cancer Inst. 2021, 113, 1648–1669.
  3. Lin, J.Y.; Fisher, D.E. Melanocyte biology and skin pigmentation. Nature 2007, 445, 843–850.
  4. Boissy, R.E. Melanosome transfer to and translocation in the keratinocyte. Exp. Dermatol. 2003, 12 (Suppl. 2), S5–S12.
  5. Mullenders, L.H.F. Solar UV damage to cellular DNA: From mechanisms to biological effects. Photochem. Photobiol. Sci. 2018, 17, 1842–1852.
  6. Davis, E.J.; Johnson, D.B.; Sosman, J.A.; Chandra, S. Melanoma: What do all the mutations mean? Cancer 2018, 124, 3490–3499.
  7. Schadendorf, D.; van Akkooi, A.C.J.; Berking, C.; Griewank, K.G.; Gutzmer, R.; Hauschild, A.; Stang, A.; Roesch, A.; Ugurel, S. Melanoma. Lancet 2018, 392, 971–984.
  8. Hodis, E.; Watson, I.R.; Kryukov, G.V.; Arold, S.T.; Imielinski, M.; Theurillat, J.P.; Nickerson, E.; Auclair, D.; Li, L.; Place, C.; et al. A landscape of driver mutations in melanoma. Cell 2012, 150, 251–263.
  9. Miller, K.D.; Nogueira, L.; Mariotto, A.B.; Rowland, J.H.; Yabroff, K.R.; Alfano, C.M.; Jemal, A.; Kramer, J.L.; Siegel, R.L. Cancer treatment and survivorship statistics, 2019. CA Cancer J. Clin. 2019, 69, 363–385.
  10. Domingues, B.; Lopes, J.M.; Soares, P.; Populo, H. Melanoma treatment in review. Immunotargets Ther. 2018, 7, 35–49.
  11. Batus, M.; Waheed, S.; Ruby, C.; Petersen, L.; Bines, S.D.; Kaufman, H.L. Optimal management of metastatic melanoma: Current strategies and future directions. Am. J. Clin. Dermatol. 2013, 14, 179–194.
  12. Van Zeijl, M.C.; van den Eertwegh, A.J.; Haanen, J.B.; Wouters, M.W. (Neo)adjuvant systemic therapy for melanoma. Eur. J. Surg. Oncol. 2017, 43, 534–543.
  13. Garbe, C.; Peris, K.; Hauschild, A.; Saiag, P.; Middleton, M.; Bastholt, L.; Grob, J.J.; Malvehy, J.; Newton-Bishop, J.; Stratigos, A.J.; et al. Diagnosis and treatment of melanoma. European consensus-based interdisciplinary guideline—Update 2016. Eur. J. Cancer 2016, 63, 201–217.
  14. Christofyllakis, K.; Pföhler, C.; Bewarder, M.; Müller, C.S.L.; Thurner, L.; Rixecker, T.; Vogt, T.; Stilgenbauer, S.; Yordanova, K.; Kaddu-Mulindwa, D. Adjuvant therapy of high-risk (stages IIC-IV) malignant melanoma in the post interferon-alpha era: A systematic review and meta-analysis. Front. Oncol. 2021, 10, 637161.
  15. Wilson, M.A.; Schuchter, L.M. Chemotherapy for Melanoma. Cancer Treat. Res. 2016, 167, 209–229.
  16. Soengas, M.S.; Lowe, S.W. Apoptosis and melanoma chemoresistance. Oncogene 2003, 22, 3138–3151.
  17. Flaherty, K.T. Targeting metastatic melanoma. Annu. Rev. Med. 2012, 63, 171–183.
  18. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 2011, 364, 2507–2516.
  19. Livingstone, E.; Zimmer, L.; Vaubel, J.; Schadendorf, D. BRAF, MEK and KIT inhibitors for melanoma: Adverse events and their management. Chin. Clin. Oncol. 2014, 3, 29.
  20. Rizos, H.; Menzies, A.M.; Pupo, G.M.; Carlino, M.S.; Fung, C.; Hyman, J.; Haydu, L.E.; Mijatov, B.; Becker, T.M.; Boyd, S.C.; et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: Spectrum and clinical impact. Clin. Cancer Res. 2014, 20, 1965–1977.
  21. Eroglu, Z.; Ribas, A. Combination Therapy with BRAF and MEK Inhibitors for Melanoma: Latest Evidence and Place in Therapy. Ther. Adv. Med. Oncol. 2016, 8, 48–56.
  22. 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.
  23. Wright, C.J.M.; McCormack, P.L. Trametinib: First Global Approval. Drugs 2013, 73, 1245–1254.
  24. Larkin, J.; Ascierto, P.A.; Dreno, B.; Atkinson, V.; Liszkay, G.; Maio, M.; Mandala, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N. Engl. J. Med. 2014, 371, 1867–1876.
  25. Karimkhani, C.; Gonzalez, R.; Dellavalle, R.P. A review of novel therapies for melanoma. Am. J. Clin. Dermatol. 2014, 15, 323–337.
  26. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.J.; et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015, 386, 444–451.
  27. Pelster, M.S.; Amaria, R.N. Combined Targeted Therapy and Immunotherapy in Melanoma: A Review of the Impact on the Tumor Microenvironment and Outcomes of Early Clinical Trials. Ther. Adv. Med. Oncol. 2019, 11, 1758835919830826.
  28. Van Breeschoten, J.; Wouters, M.W.J.M.; Hilarius, D.L.; Haanen, J.B.; Blank, C.U.; Aarts, M.J.B.; van den Berkmortel, F.W.P.J.; de Groot, J.-W.B.; Hospers, G.A.P.; Kapiteijn, E.; et al. First-Line BRAF/MEK Inhibitors versus Anti-PD-1 Monotherapy in BRAFV600-Mutant Advanced Melanoma Patients: A Propensity-Matched Survival Analysis. Br. J. Cancer 2021, 124, 1222–1230.
  29. Gata, V.A.; Lisencu, C.I.; Vlad, C.I.; Piciu, D.; Irimie, A.; Achimas-Cadariu, P. Tumor infiltrating lymphocytes as a prognostic factor in malignant melanoma. Review of the literature. J. Buon. 2017, 22, 592–598.
  30. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570.
  31. Gasser, S.; Lim, L.H.K.; Cheung, F.S.G. The role of the tumour microenvironment in immunotherapy. Endocr. Relat. Cancer 2017, 24, T283–T295.
  32. Khair, D.O.; Bax, H.J.; Mele, S.; Crescioli, S.; Pellizzari, G.; Khiabany, A.; Nakamura, M.; Harris, R.J.; French, E.; Hoffmann, R.M.; et al. Combining Immune Checkpoint Inhibitors: Established and Emerging Targets and Strategies to Improve Outcomes in Melanoma. Front. Immunol. 2019, 10, 453.
  33. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546.
  34. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330.
  35. Gide, T.N.; Wilmott, J.S.; Scolyer, R.A.; Long, G.V. Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Metastatic Melanoma. Clin. Cancer Res. 2018, 24, 1260–1270.
  36. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723.
  37. D’Orazio, J.; Jarrett, S.; Amaro-Ortiz, A.; Scott, T. UV radiation and the skin. Int. J. Mol. Sci. 2013, 14, 12222–12248.
  38. Handy, D.E.; Loscalzo, J. Redox regulation of mitochondrial function. Antioxid. Redox Signal. 2012, 16, 1323–1367.
  39. Denat, L.; Kadekaro, A.L.; Marrot, L.; Leachman, S.A.; Abdel-Malek, Z.A. Melanocytes as instigators and victims of oxidative stress. J. Invest. Dermatol. 2014, 134, 1512–1518.
  40. Ryoo, I.G.; Kwak, M.K. Regulatory crosstalk between the oxidative stress-related transcription factor Nfe2l2/Nrf2 and mitochondria. Toxicol. Appl. Pharmacol. 2018, 359, 24–33.
  41. Marrot, L.; Jones, C.; Perez, P.; Meunier, J.R. The significance of Nrf2 pathway in (photo)-oxidative stress response in melanocytes and keratinocytes of the human epidermis. Pigment. Cell Melanoma Res. 2008, 21, 79–88.
  42. Cassidy, P.B.; Liu, T.; Florell, S.R.; Honeggar, M.; Leachman, S.A.; Boucher, K.M.; Grossman, D. A Phase II Randomized Placebo-Controlled Trial of Oral N-acetylcysteine for Protection of Melanocytic Nevi against UV-Induced Oxidative Stress In Vivo. Cancer Prev. Res. 2017, 10, 36–44.
  43. Carpenter, E.L.; Wyant, M.B.; Indra, A.; Ito, S.; Wakamatsu, K.; Merrill, G.F.; Moos, P.J.; Cassidy, P.B.; Leachman, S.A.; Ganguli-Indra, G.; et al. Thioredoxin Reductase 1 Modulates Pigmentation and Photobiology of Murine Melanocytes in vivo. J. Invest. Dermatol. 2022.
  44. Meyskens, F.L., Jr.; McNulty, S.E.; Buckmeier, J.A.; Tohidian, N.B.; Spillane, T.J.; Kahlon, R.S.; Gonzalez, R.I. Aberrant redox regulation in human metastatic melanoma cells compared to normal melanocytes. Free Radic. Biol. Med. 2001, 31, 799–808.
  45. Liu-Smith, F.; Dellinger, R.; Meyskens, F.L., Jr. Updates of reactive oxygen species in melanoma etiology and progression. Arch. Biochem. Biophys. 2014, 563, 51–55.
  46. Warburg, O.; Wind, F.; Negelein, E. The Metabolism of Tumors in the Body. J. Gen. Physiol. 1927, 8, 519–530.
  47. Abildgaard, C.; Guldberg, P. Molecular drivers of cellular metabolic reprogramming in melanoma. Trends Mol. Med. 2015, 21, 164–171.
  48. Mitsuishi, Y.; Taguchi, K.; Kawatani, Y.; Shibata, T.; Nukiwa, T.; Aburatani, H.; Yamamoto, M.; Motohashi, H. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 2012, 22, 66–79.
  49. Liou, G.Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496.
  50. Obrador, E.; Liu-Smith, F.; Dellinger, R.W.; Salvador, R.; Meyskens, F.L.; Estrela, J.M. Oxidative stress and antioxidants in the pathophysiology of malignant melanoma. Biol. Chem. 2019, 400, 589–612.
  51. Carretero, J.; Obrador, E.; Anasagasti, M.J.; Martin, J.J.; Vidal-Vanaclocha, F.; Estrela, J.M. Growth-associated changes in glutathione content correlate with liver metastatic activity of B16 melanoma cells. Clin. Exp. Metastasis 1999, 17, 567–574.
  52. Vene, R.; Castellani, P.; Delfino, L.; Lucibello, M.; Ciriolo, M.R.; Rubartelli, A. The cystine/cysteine cycle and GSH are independent and crucial antioxidant systems in malignant melanoma cells and represent druggable targets. Antioxid. Redox Signal. 2011, 15, 2439–2453.
  53. Cassidy, P.B.; Honeggar, M.; Poerschke, R.L.; White, K.; Florell, S.R.; Andtbacka, R.H.; Tross, J.; Anderson, M.; Leachman, S.A.; Moos, P.J. The role of thioredoxin reductase 1 in melanoma metabolism and metastasis. Pigment. Cell Melanoma Res. 2015, 28, 685–695.
  54. Rotblat, B.; Grunewald, T.G.; Leprivier, G.; Melino, G.; Knight, R.A. Anti-oxidative stress response genes: Bioinformatic analysis of their expression and relevance in multiple cancers. Oncotarget 2013, 4, 2577–2590.
  55. Bisevac, J.P.; Djukic, M.; Stanojevic, I.; Stevanovic, I.; Mijuskovic, Z.; Djuric, A.; Gobeljic, B.; Banovic, T.; Vojvodic, D. Association Between Oxidative Stress and Melanoma Progression. J. Med. Biochem. 2018, 37, 12–20.
  56. Sander, C.S.; Hamm, F.; Elsner, P.; Thiele, J.J. Oxidative stress in malignant melanoma and non-melanoma skin cancer. Br. J. Dermatol. 2003, 148, 913–922.
  57. Garate, M.; Wani, A.A.; Li, G. The NAD(P)H:Quinone Oxidoreductase 1 induces cell cycle progression and proliferation of melanoma cells. Free Radic. Biol. Med. 2010, 48, 1601–1609.
  58. Liu, L.; Wu, Y.; Bian, C.; Nisar, M.F.; Wang, M.; Hu, X.; Diao, Q.; Nian, W.; Wang, E.; Xu, W.; et al. Heme oxygenase 1 facilitates cell proliferation via the B-Raf-ERK signaling pathway in melanoma. Cell Commun. Signal. 2019, 17, 3.
  59. Cannavo, S.P.; Tonacci, A.; Bertino, L.; Casciaro, M.; Borgia, F.; Gangemi, S. The role of oxidative stress in the biology of melanoma: A systematic review. Pathol. Res. Pract. 2019, 215, 21–28.
  60. Ramasamy, P.; Larkin, A.M.; Linge, A.; Tiernan, D.; McAree, F.; Horgan, N.; Moriarty, P.; Beatty, S.; Murphy, C.C.; Clynes, M.; et al. PRDX3 is associated with metastasis and poor survival in uveal melanoma. J. Clin. Pathol. 2020, 73, 408–412.
  61. Lokaj, K.; Meierjohann, S.; Schutz, C.; Teutschbein, J.; Schartl, M.; Sickmann, A. Quantitative differential proteome analysis in an animal model for human melanoma. J. Proteome Res. 2009, 8, 1818–1827.
  62. Le Gal, K.; Ibrahim, M.X.; Wiel, C.; Sayin, V.I.; Akula, M.K.; Karlsson, C.; Dalin, M.G.; Akyurek, L.M.; Lindahl, P.; Nilsson, J.; et al. Antioxidants can increase melanoma metastasis in mice. Sci. Transl. Med. 2015, 7, 308re8.
  63. Paudel, B.B.; Lewis, J.E.; Hardeman, K.N.; Hayford, C.E.; Robbins, C.J.; Stauffer, P.E.; Codreanu, S.G.; Sherrod, S.D.; McLean, J.A.; Kemp, M.L.; et al. An Integrative Gene Expression and Mathematical Flux Balance Analysis Identifies Targetable Redox Vulnerabilities in Melanoma Cells. Cancer Res. 2020, 80, 4565–4577.
  64. Corazo-Rozas, P.; Guerreschi, P.; André, F.; Gabert, P.; Lancel, S.; Dekiouk, S.; Fontaine, D.; Tardivel, M.; Savina, A.; Quesnel, B.; et al. Mitochondrial oxidative phosphorylation controls cancer cell’s life and death descisions upon exposure to MAPK inhibitors. Oncotarget 2016, 7, 39473–39485.
  65. Carpenter, E.L.; Chagani, S.; Nelson, D.; Cassidy, P.B.; Laws, M.; Ganguli-Indra, G.; Indra, A.K. Mitochondrial complex I inhibitor deguelin induces metabolic reprogramming and sensitizes vemurafenib-resistant BRAFV600E mutation bearing metastatic melanoma cells. Mol. Carcinog. 2019, 58, 1680–1690.
  66. Pizzimenti, S.; Ribero, S.; Cucci, M.A.; Grattarola, M.; Monge, C.; Dianzani, C.; Barrera, G.; Muzio, G. Oxidative Stress-Related Mechanisms in Melanoma and in the Acquired Resistance to Targeted Therapies. Antioxidants 2021, 10, 1942.
  67. Vrijsen, S.; Besora-Casals, L.; van Veen, S.; Zielich, J.; van den Haute, C.; Hamouda, N.N.; Fischer, C.; Ghesquière, B.; Tournev, I.; Agostinis, P. ATP13A2-mediated endo-lysosomal polyamine export counters mitochondrial oxidative stress. Proc. Natl. Acad. Sci. USA 2020, 117, 31198–31207.
  68. Diaconeasa, Z.; Ayvaz, H.; Ruginǎ, D.; Leopold, L.; Stǎnilǎ, A.; Socaciu, C.; Tăbăran, F.; Luput, L.; Mada, D.C.; Pintea, A.; et al. Melanoma inhibition by anthocyanins is associated with the reduction of oxidative stress biomarkers and changes in mitochondrial membrane potential. Plant. Foods Hum. Nutr. 2017, 72, 404–410.
  69. Stevens, J.F.; Revel, J.S.; Maier, C.S. Mitochondira-centric review of polyphenol bioactivity in cancer models. Antioxid. Redox Signal. 2018, 29, 1589–1611.
  70. Coleman, D.J.; Garcia, G.; Hyter, S.; Jang, H.S.; Chagani, S.; Liang, X.; Larue, L.; Ganguli-Indra, G.; Indra, A.K. Retinoid-X-receptors (alpha/beta) in melanocytes modulate innate immune responses and differentially regulate cell survival following UV irradiation. PLoS Genet. 2014, 10, e1004321.
  71. Zhu, H.; Jia, Z.; Trush, M.A.; Li, Y.R. Nrf2 deficiency promotes melanoma growth and lung metastasis. React. Oxyg. Species 2016, 2, 308–314.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 336
Revisions: 2 times (View History)
Update Date: 06 Apr 2022
1000/1000
Video Production Service