Next Article in Journal
UPF1—From mRNA Degradation to Human Disorders
Next Article in Special Issue
Kinetics of Plasma Cell-Free DNA under a Highly Standardized and Controlled Stress Induction
Previous Article in Journal
RIPK1 Regulates Microglial Activation in Lipopolysaccharide-Induced Neuroinflammation and MPTP-Induced Parkinson’s Disease Mouse Models
Previous Article in Special Issue
Renal Ischemia Tolerance Mediated by eIF5A Hypusination Inhibition Is Regulated by a Specific Modulation of the Endoplasmic Reticulum Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Survival Mechanisms of Metastatic Melanoma Cells: The Link between Glucocorticoids and the Nrf2-Dependent Antioxidant Defense System

by
Elena Obrador
1,2,*,
Rosario Salvador-Palmer
1,
Rafael López-Blanch
1,2,
María Oriol-Caballo
1,2,
Paz Moreno-Murciano
2 and
José M. Estrela
1,2,3,*
1
Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
2
Scientia BioTech S.L., 46002 Valencia, Spain
3
Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
*
Authors to whom correspondence should be addressed.
Cells 2023, 12(3), 418; https://doi.org/10.3390/cells12030418
Submission received: 25 November 2022 / Revised: 11 January 2023 / Accepted: 23 January 2023 / Published: 26 January 2023
(This article belongs to the Special Issue Molecular Mechanism of Stress, Stress Response, and Adaptation)

Abstract

:
Circulating glucocorticoids increase during stress. Chronic stress, characterized by a sustained increase in serum levels of cortisol, has been associated in different cases with an increased risk of cancer and a worse prognosis. Glucocorticoids can promote gluconeogenesis, mobilization of amino acids, fat breakdown, and impair the body’s immune response. Therefore, conditions that may favor cancer growth and the acquisition of radio- and chemo-resistance. We found that glucocorticoid receptor knockdown diminishes the antioxidant protection of murine B16-F10 (highly metastatic) melanoma cells, thus leading to a drastic decrease in their survival during interaction with the vascular endothelium. The BRAFV600E mutation is the most commonly observed in melanoma patients. Recent studies revealed that VMF/PLX40-32 (vemurafenib, a selective inhibitor of mutant BRAFV600E) increases mitochondrial respiration and reactive oxygen species (ROS) production in BRAFV600E human melanoma cell lines. Early-stage cancer cells lacking Nrf2 generate high ROS levels and exhibit a senescence-like growth arrest. Thus, it is likely that a glucocorticoid receptor antagonist (RU486) could increase the efficacy of BRAF-related therapy in BRAFV600E-mutated melanoma. In fact, during early progression of skin melanoma metastases, RU486 and VMF induced metastases regression. However, treatment at an advanced stage of growth found resistance to RU486 and VMF. This resistance was mechanistically linked to overexpression of proteins of the Bcl-2 family (Bcl-xL and Mcl-1 in different human models). Moreover, melanoma resistance was decreased if AKT and NF-κB signaling pathways were blocked. These findings highlight mechanisms by which metastatic melanoma cells adapt to survive and could help in the development of most effective therapeutic strategies.

1. Introduction

Stressful events may precede cancer and stress-related psychosocial factors appear associated with higher cancer incidence and poorer survival [1]. The question of whether there is a link between stress and cancer has confused and interested both researchers and patients. Study after study has asked whether people who develop cancer have experienced more stress in the years before diagnosis and, conversely, if people who have experienced extreme stress are more likely to develop cancer. In this regard, epidemiological evidence continues to accumulate on the effect of psychosocial, behavioral and physic stress in relation to cancer risk, progression, and mortality [2,3,4]. Consequently, stress management appears essential for cancer patients, and particularly in the case of melanoma, a pathology in which there is a narrow barrier between benign lesions, malignant transformation, and metastatic spread [5].
Stress-induced diseases are the consequence of an excessive adrenergic response and mainly due to a glucocorticoid-dependent deterioration of the immune system. Stress is linked to a lower efficiency of natural cell repair processes [6], and emerging evidence suggest that DNA damage is increased by exposure to psychological stress and stress hormones [7]. Nevertheless, the key question is whether stress mediators may have a direct impact on DNA mutations, repair mechanisms, epigenetic changes, cancer spread and metastasis. In this sense, results from recent research are mixed since some experts suggest that stress can cause cancer, while others believe it may only contribute to the condition, see, e.g., [2,8].
Recent studies indicating that stress could facilitate cancer growth and even metastatic spread are mainly animal studies [3]. As pointed out by Eckerling et al. [3], “the stress response can facilitate cancer growth and metastasis through a direct action on the molecular characteristics of malignant tissue, on its microenvironment, on antitumor immune activity, and on other indirect modulators of progression”. For instance, cortisol increases the expression of the HPV16 E6 and E7 oncogenes, which facilitate degradation of p53 and, thereby, tumor initiation [9]. Moreover, stress conditions and/or increased levels of cortisol are associated to poor/bad nutrition, poor sleep and vitamin D deficiency, all factors that may favor the development of cancer [1].
In colon and gynecological cancers (endometrial, ovarian and triple negative breast cancers), it has been demonstrated that high glucocorticoid expression or glucocorticoid receptor (GR) activation are linked with cancer progression, development of treatment resistance, and/or a poorer patient prognosis [10,11,12,13,14,15]. Moreover, overexpression of GR induces cisplatin resistance through p38 MAP kinase in cervical cancer patients [16]. Prolonged serum elevation of glucocorticoid levels can negatively influence mitochondrial function leading to mitochondrial damage with a negative impact on cellular metabolism [17]. In addition, stress can reduce the body’s resistance to some types of viruses, which are now known to be significantly involved in the initiation of around 15% of cancer cases. Human papillomavirus [18], Epstein-Barr virus [19], Kaposi sarcoma-associated herpesvirus [20], and hepatitis C and B viruses [21] can be reactivated by catecholamines and glucocorticoids (e.g., [22,23]). Glucocorticoids are among the most potent immunosuppressive agents and, thus, may favor the progression of cancer [24]. They inhibit the synthesis of almost all known cytokines and of several cell surface molecules required for immune function [25]. Around 15–20% of all cancer cases are preceded by infection or chronic inflammation at the same organ site and, in many cases, inflammation exists long before tumor formation [26]. All these facts suggest that stress and glucocorticoids, in particular, could be involved in the initiation of the carcinogenic process, and favor the transformation of a physiological microenvironment into a protumoral milieu.
Nevertheless, moving from the level of animal research to human clinical research, both epidemiological studies and clinical trials have generated somewhat uncertain results, indicating only a small, not necessarily significant, effect of stress on cancer progression [3,27,28] (www.clinicaltrials.gov accessed on 5 January 2023). Consequently, the current medical routine does not include specific measures designed to prevent stress responses as a means of improving cancer survival [3]. Nevertheless, it seems reasonable to suggest that stress management interventions should be tested in the critical periods that affect cancer progression, especially in the short postoperative period and adjuvant treatments. In addition, more experimental studies will be needed to assess the long-term effects of treatments.

2. Melanoma Incidence, Prognosis and Therapeutic Challenges

Skin melanoma accounted for 4% of all new cancer diagnoses in EU-27 countries in 2020 (all cancers, excluding non-melanoma skin cancers) and for 1.3% of all deaths due to cancer (https://ecis.jrc.ec.europa.eu accessed on 3 January 2023). Ultraviolet (UV) solar radiation is the main source of skin damage that can lead to skin cancer. Both UV radiation and stress cause the body to produce reactive oxygen species (ROS) [29]. Those can increase inflammation and damage our skin’s DNA, leading to mutations and, possibly, skin cancer [29,30]. In fact, coupled with genetic and environmental factors, inflammation and stress have been suggested to play a role in melanoma formation and progression [31,32].
As in other tumors, the best prognosis is achieved by early removal of tumors. The two most common types of melanomas are [33]:
(a)
Superficial spreading melanoma: this type accounts for 70% of melanomas and most often affects the legs of women and the torsos of men. Tumor cells usually have mutations in the BRAF gene.
(b)
Nodular melanoma: 15 to 30% of melanomas, appears anywhere on the body, and grows rapidly.
The shallower the melanoma is in the skin, the greater the chance that surgery will cure it. Almost 100% of shallow and newer melanomas are cured by surgery. However, melanomas deeper than about 1 mm in the skin have a higher risk of metastasis to lymph nodes and spread through the blood. Once the melanoma has reached the lymph nodes, the 5-year survival rate varies between 25% and 70%. Once melanoma has metastasized to distant parts of the body, the 5-year survival rate is about 10% [34].
If the melanoma has spread to distant areas, surgery is not usually an option. Chemotherapy, such as dacarbazine and temozolomide, can be given intravenously to treat melanomas that have spread, but they still do not prolong survival and are given to people who have no other options. Radiation therapy may be used when the cancer has metastatized the brain (www.cancer.org accessed on 15 December 2022)
BRAF (vemurafenib, dabrafenib and encorafenib) and MEK (trametinib, cobimetinib and binimetinib) inhibitors have proven to improve survival in BRAF-mutant unresectable or metastatic melanoma [35], although half of the patients develop resistance within a year [36]. As the concurrent inhibition of the BRAF and MEK proteins could decrease MAPK-acquired resistance, and lead to a longer duration of response and overall survival, BRAF/MEK inhibitors are currently used in combinations in the clinical practice [37].
PD1, PD-L1, CTLA-4 and LAG-3 are immune checkpoint proteins physiologically expressed by immunocompetent cells to maintain immunological homeostasis and prevent autoimmunity. However, they can be used by cancer cells to down-regulate antitumor responses and evade the immune response [38]. Melanoma was the first malignancy to be treated with immunological checkpoint inhibitors (ICIs) [39]. ICIs are monoclonal antibodies that selectively bind to these proteins and reestablish the anti-tumor immune responses. Four classes of ICIs are approved by the FDA for the treatment of melanoma: ipilimumab (an antagonist of the cytotoxic-T lymphocytes antigen 4, CTLA-4) [40]; nivolumab and pembrolizumab, antagonists of programmed cell death protein 1 (PD-1) [41]; atezolizumab, an antagonist of programmed cell death ligand 1 (PD-L1) [42]; and relatlimab-rmbw, (a combination of the LAG-3-blocking antibody relatlimab and the programmed death receptor 1-blocking antibody nivolumab) [42]. Despite the increase in survival associated with the introduction of ICIs therapies [43], approximately half of the patients with melanoma do not obtain a lasting benefit [44,45].
In addition, over 350 NCI-registered clinical trials are currently being conducted. Many trials have been carried out to treat advanced or metastatic melanoma by using monotherapy or combination therapy of chemotherapy, immunotherapy, radiotherapy, and targeted therapy, and also new dosage forms or delivery systems (see [46]).

3. Intracellular Redox State and Oxidative Stress in Melanoma Initiation and Progression

Oxidative stress associates to an excessive ROS production, byproducts of O2 metabolism which also have key roles in cell signaling [47]. ROS-induced molecular damages and signaling activation of specific pathways can affect carcinogenesis and tumor progression [48,49]. Oxidative stress and/or redox status alterations may lead to cell transitions from quiescent to proliferative status, growth arrest and/or cell death depending on the importance of the redox imbalance [50]. Therefore, although oxidative stress and redox status shifts can cause cancer cell death, it is also feasible that they help to generate cell subsets capable of adapting and survive.
In addition to ROS, other reactive species can have a significant impact on the intracellular redox status, i.e., reactive nitrogen species (RNS), reactive sulfur species, reactive carbonyl species, reactive selenium species, chlorine and bromine species, also prooxidants such as transition metals (e.g., Mg2+, Cu2+ or Fe2+) or vitamins (e.g., Vitamin C), and some chemotherapeutic drugs (e.g., adriamycin and other anthracyclines, bleomycin, and cisplatin which bind to DNA and generate ROS, or quinones, highly redox active molecules which can cycle with their semiquinone radicals, leading to formation of ROS) [29].
Cancer cells, including melanoma cells, overproduce ROS compared to their normal cell counterparts [51,52,53]. ROS can be generated by mitochondria, the melanosomes, NADPH oxidase family enzymes, different arachidonic acid oxygenase activities, and the nitric oxide synthase activities [54]. In addition, an increased metabolism, as compared to normal melanocytes, interaction with immune and endothelial cells, UV radiation, and changes in the antioxidant system are factors that must be also taken into account to evaluate ROS levels and their effects on the growing melanoma [29]. For instance, H2O2 production is higher in melanoma cells than in melanocytes, and H2O2 induces higher tyrosinase activity (the rate-limiting enzyme in melanin synthesis) [55]. Moreover, melanin synthesis associates with higher levels of ROS, which turns the melanin/ROS ratio into a vicious cycle that favors the progression of melanoma [56]. Melanin, which is usually in an antioxidative reduced state within the melanosome, evolves during the pathogenesis of melanoma into a pro-oxidant substance that generates superoxide anion [57,58]. The important role of the antioxidant response during melanomagenesis is suggested by the overexpression of heme-oxygenase-1 (HO-1), a Nrf2 target. HO-1 is upregulated in B16-F10 murine melanoma cells and in different melanoma tumor models growing in vivo. Cells overexpressing HO-1, compared to controls, had increased proliferation rate, improved resistance to H2O2-induced oxidative stress, higher angiogenic activity, augmented metastatic activity, and decreased survival [59].
Cancer cells face replication stress caused by ROS-induced DNA damage, by oncogenic stress associated to dysregulation of fork progression, or by chemotherapy and radiotherapy. NOK-SI cells (human oral keratinocytes) stimulated with norepinephrine or cortisol showed higher DNA damage compared to untreated cells, whereas the hormone-induced DNA damage was reversed by pre-treatment with the β-adrenergic blocker propranolol [60].
Hence, there is much accumulated evidence of oxidative stress in melanoma cells growing in vitro and in murine models growing in vivo (e.g., [54,57,61,62,63,64,65,66]). Consequently, upregulation of their antioxidant defenses appears necessary to guarantee their survival, or at least that of the most resistant clones.

4. Stress Hormones and Melanoma Growth

As suggested by different studies and as pointed out by Sanzo et al. [67], chronic stress, involving environmental and psychological factors, could be a relevant cofactor in melanoma progression and spreading. In this, different risk factors have been suggested, i.e., excessive body mass index, high stress-related activities, or immunosuppression [68]. Moreover, stress hormones can cause upregulation of cytokines, i.e., VEGF, TGF, IL6 or IL8, which are proangiogenic and/or favor tumor progression [32,69]. Therefore, it seems plausible to infer that melanoma progression may be inhibited by blocking the molecular signaling cascades involving specific cytokines.
IL-6 is dysregulated in many types of cancers, and increased serum levels of IL-6 have been correlated with a worse prognosis in patients bearing different cancers, including melanoma [29,70]. In this regard, it has been shown that solid tumor cells may secrete high levels of IL-6, which is involved in fundamental processes in cancer metastasis, i.e., angiogenesis, proliferation, attachment, and invasion (e.g., [71,72]). In the classic B16-F10 melanoma model, known for its high metastatic potential, we observed that IL-6 (mainly derived from the melanoma cells) promotes the release of glutathione (GSH) from hepatocytes to the circulating blood [73]. This facilitates GSH to reach distant growing metastases. The plasma membrane-bound γ-glutamyl transpeptidase (GGT) enzyme degrades extracellular GSH, thus releasing cysteinyl-glycine (further metabolized by dipeptidases) and γ-glutamyl amino acids [74,75,76]. Free cysteine, glycine and γ-glutamyl-amino acids are taken up by the melanoma cells and can be used as GSH precursors [77]. Indeed, in the B16-F10 model, GGT activity and the interorgan transport of GSH promote the synthesis of GSH in the melanoma cells and their metastatic growth [78]. In this mechanism, the liver plays an essential role because it is the major physiological reservoir of GSH [79].
The neuroendocrine and immune systems work in order of maintaining homeostasis under conditions that favor overproduction of cytokines [80]. The hypothalamus-pituitary-adrenal (HPA) axis can be stimulated by cytokines (e.g., IL-1, IL-6, or αTNF), which are overproduced in many different immune, inflammatory or neoplastic processes [81]. Consequently, the HPA axis increases secretion of ACTH, thereby activating the synthesis and release of glucocorticoids from the adrenal glands [82]. Interestingly, pathophysiological concentrations of cortisol have been shown to increase IL-6 production by, e.g., human squamous cell carcinoma cells [83]. Moreover, in patients with advanced ovarian cancer, increased levels of IL-6 in ascitic fluid correlated with increased salivary cortisol [84]. More importantly, tumor-derived IL-6 impairs the ketogenic response to reduced caloric intake, thus promoting a systemic metabolic stress response that blocks anti-cancer immunotherapy [85]. Thus, suggesting a role of IL-6 to increase glucocorticoid secretion, and the consequent immune suppression. Facts that raise the question of whether glucocorticoids should be targeted in conjunction with immunotherapy interventions.
Glucocorticoids are useful in the primary combination chemotherapy of both acute and chronic lymphocytic leukemias, Hodgkin’s and non-Hodgkin’s lymphomas, multiple myeloma and breast cancer [86]. Glucocorticoids work through their receptors to perform a variety of functions, including arresting growth or inducing apoptosis in lymphocytes [86]. The glucocorticoid-induced apoptosis appears to involve multiple signaling pathways, i.e., transactivation of apoptosis inducing genes, such as Bim, and the negative modulation of survival cytokines through inhibition of AP-1 and NF-κB mediated transcriptions [87,88,89]. However, they seem to blunt different chemotherapeutics, as it occurs, e.g., in ovarian cancers [90] or in many other tumors [91]. Moreover, glucocorticoids are also likely to blunt immunotherapies by interfering with immune responses [92,93]. Thus, it seems reasonable to think that inhibition of the GRs may help to prevent these problems.
Furthermore, pathophysiological levels of noradrenaline favor overexpression of VEGF, IL-8, and IL-6 in different human melanoma cell lines, and cytokine production are progressively increased in the metastatic phenotypes [31]. β-adrenoceptors are upregulated in human melanoma and their activation releases pro-tumorigenic cytokines [94], whereas α-adrenoceptor stimulation appears to attenuate melanoma growth in mice [95]. Furthermore, catecholamines have been found to increase proliferation of murine melanoma B16-F10 cells [96]. Based on the results of retrospective and prospective observational studies, Giorgi et al. recently suggested that β-blockers treatments should be considered as a treatment in melanoma, although clinical trials would obviously be needed to test their efficacy [97].
Therefore, based on this background, it is plausible that glucocorticoids and catecholamines may influence melanoma growth and IL-6 production in its metastatic cells. Further work in the B16-F10 melanoma model showed that plasma levels of ACTH, corticosterone and noradrenaline increase in mice bearing B16-F10 lung or liver metastases, as compared to non-tumor-bearing controls [98]. Corticosterone and noradrenaline, at pathophysiological levels, increased expression and secretion of IL-6 in the B16-F10 cells, which involves changes in the DNA binding activity of NF-κB, cAMP response element-binding protein, AP-1, and nuclear factor for IL-6 [98]. Moreover, in vivo inoculation of B16-F10 cells transfected with anti-IL-6-siRNA, treatment with the GR blocker RU-486 or with propranolol (a β-adrenoceptor blocker), increased hepatic GSH whereas decreased plasma IL-6 levels and metastatic growth [98]. In addition, IL-6 may also promote mechanisms to avoid the stress- and/or cytotoxic drug-induced metastatic cell death (e.g., increased expression of several survival proteins, such as Bcl-2, Bcl-xL, Mcl-1, survivin, and XIAP) [98,99].
Figure 1 schematically describes the pathophysiology of stress and its metabolic consequences in metastatic melanoma-bearing mammals.

5. Glucocorticoids and the Antioxidant Defense of Melanoma Cells

5.1. Glucocorticoids, Nrf2 and the Antioxidant Defense of Melanoma Cells

The transcription activator Nrf2 is the master regulator of the antioxidant response and upregulates the expression of antioxidant and detoxifying enzymes [100]. Nrf2 has a protective role in UV-induced oxidative stress, DNA damage, and apoptosis of melanocytes [101]. Given the important contribution of UV radiation for ROS formation, it is not surprising that Nrf2 activity is induced by UV in melanocytes [100]. Nrf1 and Nrf2 transcription factors, upon activation by oxidative stress, form heterodimers with different factors, i.e., Maf and Jun, to bind to the antioxidant/electrophile response element (ARE/EpRE) and regulate the transcription of oxidative stress/cytoprotection-related genes [59,102]. This is important because oncogene (i.e., KRAS, BRAF or MYC)-induced Nrf2 transcription activity associates to increases in melanoma growth and pharmacological resistance [103,104,105]. Elevated Nrf2 expression and a high GSH/GSSG ratio in melanoma are correlated with a deeper Breslow index, invasive/metastatic phenotype, and poor survival [106,107,108]. In that sense, it was shown that GSH protects melanoma from oxidative stress, contributing to its survival [107,109]. These results are in agreement with Beberok et al. who demonstrated that treatment with the antibiotic Lomefloxacin induce oxidative stress, GSH depletion and apoptosis in the COLO829 melanoma cell line [110]. Moreover, N-acetylcysteine (a classical mucolytic drug) can promote melanoma metastases spread, a fact suggesting that caution should be taken when administering GSH promoters to cancer patients [111]. Furthermore, the link between Nrf2 and immune tolerance has already been shown in lung adenocarcinoma, where Keap1 mutations are present in up to 20% and lead to permanent Nrf2 activation [112]. This is an important question in the case of metastatic melanomas where the anti-PD-1 immune therapy represent the only optional treatment [113,114].
Since glucocorticoids increase ROS generation in metastatic B16-F10 melanoma cells [98] and also in breast cancer cells [115], we investigated if the decrease in antioxidant enzyme activities in invasive B16-F10 cells (iB16) knockdown for the GR (iB16-shGR) was associated with changes in nuclear Nrf1 and/or Nrf2. Nuclear Nrf2, and not Nrf1, decreased in iB16-shGR cells isolated from lung or liver metastatic foci compared to control iB16 cells [116]. This is a fact that may be key since increased Nrf2 transcription activity has been correlated with aggressiveness in different human cancers [117,118,119]. However, other authors have found just opposite results postulating that GR signaling represses the antioxidant response, e.g., by inhibiting the histone acetylation mediated by Nrf2 [120], or by forming a glucocorticoid-GR complex that migrates to the nucleus where it binds to glucocorticoid response elements and ARE/EpRE sequences [121,122]. These controversial results should be analyzed in the light of the actual doses of glucocorticoids administered. Ligand-occupied GR induces or represses the transcription of thousands of genes by direct binding to DNA response elements and/or by physically associating with other transcription factors, thus involving a vast array of molecular interactions [123]. It is then essential to differentiate between pathophysiological and pharmacological levels, the latter being much higher (as reported in [120,122]) and potentially causing very different results. Indeed, biological stressors can positively or negatively affect antioxidant enzymes depending on the time and levels of exposure [124]. In fact, exposure to physiological stressors induces the production of ROS and oxidative stress in, e.g., the rat liver [125]. More importantly, a meta-analysis of glucocorticoids as modulators of oxidative stress concluded that glucocorticoids promote oxidative stress [126], which cannot be the result of improving the antioxidant defenses. Although, glucocorticoids are currently used against different cancers [127], they can also induce cancer resistance, a still unclear effect that may promote growth and metastases [127,128]. In fact, at pathophysiological levels, glucocorticoid signaling is antiapoptotic in cells of epithelial origin and in many malignant solid tumors subjected to cytotoxic therapy [89,129,130,131]. It was shown, in the immortalized human mammary epithelial cell line MCF10A, that GR-mediated protection from apoptosis is associated with induction of the serine/threonine survival kinase gene, sgk-1 [132]. In agreement with these ideas, GR antagonism has been shown to promote apoptosis in solid tumor cells [133].
Importantly, at high levels or long-term exposure of ROS, p53 expression (promoted by DNA damage) increases, activating prooxidant genes, interfering with the Nrf2-dependent transcription of ARE/EpRE-containing promoters (and, thereby, inhibiting the Nrf2-mediated survival response), and potentially resulting in cell death [134]. However, particularly in highly aggressive human cancers, the p53 protein is reduced, lost, or mutated [135]. In this scenario, we used AS101 (ammonium tri-chloro(dioxoethylene-O,O′-)tellurate), a synthetic compound which has immunomodulating properties [136] and increases expression of wild-type p53 [137]. We observed that AS101-induced up-regulation of p53 in iB16 melanoma cells caused a decrease in antioxidant enzyme expression without affecting the nuclear levels of Nrf2 [116]. An effect that was reversed by using anti-p53 antisense oligonucleotides [116]. Thus, proving that p53 can suppress the Nrf2-dependent transcription of antioxidant enzymes in metastatic melanoma cells. Interestingly GR activation may lead to inhibition of p53-induced apoptosis, an effect observed in, e.g., MCF-10Amyc cells [138]. In agreement with this concept, in estrogen receptor-positive breast cancer cells, low GR expression has been linked to higher p53 expression [139]. Indeed, p53 can form a complex with the glucocorticoid that causes a cytoplasmic sequestration of both molecular structures [140]. These facts suggest a close link among GRs, p53 and Nrf2 which could be involved in regulating growth and spread of BRAFV600E-mutated melanoma cells. Figure 2 summarizes, as a working hypothesis, potential molecular interactions that may involve GRs and the Nrf2-dependent antioxidant defenses in melanoma cells.
Upon interaction of circulating melanoma cells with the vascular endothelium, a cascade of molecular events associates to the classical docking and rolling, i.e., attachment to the endothelial cells, release of proinflammatory cytokines, ROS and RNS, and tumor cytotoxicity [141]. Independently of the tumor location (liver, lung, or subcutaneous), in metastatic melanoma cells, GR knockdown decreased the expression and activities of γ-GCS, superoxide dismutase 1 and 2, catalase, glutathione peroxidase, and glutathione reductase, inducing a reduction in GSH levels [111]. Facts showing that GR knockdown compromises the antioxidant defense of melanoma cells, and increases the endothelium-induced tumor cytotoxicity [116]. Hence limiting their invasive capacity. This opens up potential therapeutic application in case selective GR blockers may show pharmacological efficacy under in vivo conditions.

5.2. Combined Glucocorticoid Receptor Antagonism and BRAF Inhibition Promotes Regression of Early Melanoma Metastases

Mifepristone (RU486) is a steroidal antiprogestogen (IC50 = 0.025 nM), as well as an antiglucocorticoid (IC50 = 2.2 nM), and antiandrogen (IC50 = 10 nM) to a much lesser extent [148]. Its relative binding affinity at the GR is more than three times that of dexamethasone and more than ten times that of cortisol [149]. The proposed mechanism of action of RU486 it is a competitive binding to the GR that prevents the dissociation of the heat shock proteins from the receptor avoiding its subsequent translocation to the nucleus and transcriptional activity [150]. RU486 does not bind to the estrogen receptor or the mineralocorticoid receptor [151]. Research work has revealed that progesterone can inhibit human melanoma cell growth. The mechanism of inhibition is due to autophagy and this effect of progesterone is not mediated through progesterone receptor [152]. Down-modulation or pharmacological inhibition of androgen receptors suppresses melanomagenesis, with increased intratumoral infiltration of macrophages and, in an immune-competent mouse model, cytotoxic T cells [153]. However, intracellular signaling derived from activation of progesterone or androgen receptors is different from that derived from GRs [154]. Recent studies have demonstrated cytotoxic and anti-metastatic effects of RU486 in vitro and in clinical trials involving meningioma, colon, breast, and ovarian cancers (e.g., Ritch et al. [155]), whereas Alvarez et al. [156] demonstrated that RU486 impedes the proliferation of uveal melanoma cells (a highly metastatic and drug resistant cancer). Furthermore, metapristone (the most active metabolite of RU486) inhibited cell viability and induced early and late apoptosis in B16-F10 and A375 melanoma cells [157]. Metapristone treatment resulted in decreased of Akt and ERK phosphorylation and of Bcl-2 and facilitated overexpression of p53 and Bax in A375 cells. In addition, metapristone suppressed cell migration and invasion by down-regulating the expression of matrix metalloproteinases (2 and 9), N-cadherin and vimentin, while E-cadherin expression was up-regulated [157].
The BRAFV600E mutation is the most commonly observed in patients, represents more than 90% of BRAF mutations in melanoma, and can be detected early during melanoma development [158]. B-Raf signaling create a balance between a pro-oncogenic signal and a senescent proliferative arrest. Interestingly, in human fibroblasts BRAFV600E-induced senescence was bypassed by the addition of glucocorticoids (albeit at pharmacological doses), which allowed their cancer transformation [159].
Vemurafenib (VMF)/PLX40-32 (a selective inhibitor of mutant BRAFV600E) was the first molecularly targeted therapy to be licensed in the US and Europe for treatment of advanced melanoma. Its mechanism of action involves selective inhibition of the mutated BRAF V600E kinase that leads to reduced signaling through the aberrant mitogen-activated protein kinase pathway [160]. It has been reported that VMF increases mitochondrial respiration-linked ROS generation in BRAFV600E melanoma cell lines [161]. However, VMF also induces HO-1 upregulation in primary BRAFV600E melanoma cell lines, limiting the efficacy of the drug and reducing the cancer cell recognition and killing by natural killer cells [162]. Thus, possibly, a GR antagonist could increase the efficacy of BRAF-related therapy in BRAFV600E-mutated melanoma. To test this hypothesis, we studied the effect of RU486 [163] on the antioxidant defense of different human BRAFV600E melanoma cell lines. We found that in vivo administration of RU486 to mice bearing metastatic BRAFV600E-mutated melanoma cells decreases Nrf2- and redox state-related enzyme activities and, in parallel, increases ROS production [109]. Further experiments showed that combined treatment with RU486 and VMF strongly inhibits BRAFV600E-mutated metastatic melanoma growth in vivo [109]. Importantly, melanoma growth inhibition was only observed if RU486 and VMF were administered simultaneously. However, if administration of VMF was delayed, the inhibitory effect of the association practically disappeared [109]. Thus, suggesting that, despite RU486 administration, melanoma cells can spontaneously develop anti-VMF resistance. Indeed, it is well known that the anti-melanoma effects of VMF are sort-lived, and that patients present tumor relapse in a short period after treatment [164,165]. In fact, melanoma cells showing acquired resistance to VMF have high rates of mitochondrial respiration associated with elevated mitochondrial oxidative stress [161]. Thus, suggesting that targeting the antioxidant defense could be the right therapeutic choice.
It is also worth to mention that the most common adverse effects of VMF treatment, i.e., pyrexia, arthralgia or skin rash, are usually treated with dexamethasone [166,167]. However, based on the above discussion, this therapy should be reconsidered as it has been recently recommended by the Oncological Endocrinology research group of the Italian Society of Endocrinology [168].

5.3. Anti-Death Adaptations Related to the Bcl-2 Family of Proteins in Advanced BRAFV600E-Mutated Melanoma Metastases

Recent research indicate that a stress-like state promotes overexpression of fos, hsp70 and ubb, all required for adaptation to diverse cellular stresses. This state has a higher tumor seeding capabilities compared to non-stressed cells, and confers intrinsic resistance to MEK inhibitors, commonly used in melanoma treatment [169]. Furthermore, this stress-like program can be induced by, e.g., heat shock, and promotes resistance to both MEK and BRAF inhibitors in human melanomas [169]. Further mechanisms of acquired melanoma resistance involve activation of the MAPK pathway. The PI3K-PTEN-AKT pathway is a 2nd resistance pathway, which often overlaps with the MAPK pathway [170,171]. Acquired resistance to MAPK pathway targeted therapies (BRAF/MEK inhibitors) develops in most patients at approx. 12 months [172]. Interestingly, it was also shown that GR-induced MAPK phosphatase-1 (MPK-1) expression inhibits paclitaxel-associated MAPK activation and contributes to breast cancer cell survival [173]. Moreover, the MEK/ERK signaling pathway regulates expression of different Bcl-2-related proteins and survival in, e.g., human pancreatic cancer cells [174,175]. We observed that different melanoma cells, surviving after RU486 treatment, down regulated expression of different Bcl-2-related pro-death genes (i.e., bax, bak, bid), whereas upregulated anti-death bcl-xl and mcl-1 [109]. Thus, we investigated if inhibition of Bcl-xL or Mcl-1 could improve the anti-melanoma effects of RU486 and VMF. Previously, dexamethasone was found to inhibit TRAIL-induced apoptosis of thyroid cancer cells via Bcl-xL induction [176]. Indeed, the treatment with RU486 + VMF + UMI-77 (UMI77 is a selective small-molecule inhibitor of Mcl-1 [177]) or RU486 + VMF + WEHI77 (WEHI77 is Bcl-xL-selective BH3 mimetic [178]) almost induced a complete regression of advanced BRAFV600E-mutated melanoma metastases depending on which of these Bcl-2-related proteins was preferentially overexpressed in the different human BRAFV600E melanomas tested [109]. These findings are very relevant since melanoma regression was also associated to an increase in host survival [109], and because BRAFV600E mutation can also be present in other malignant neoplasms such as hairy-cell leukemia, colon carcinoma, ovarian low-grade serous carcinoma, Langerhans cell histiocytosis and Erdheim-Chester disease, glial neoplasms and thyroid carcinoma [179].

6. Conclusions

To date, and based on the epidemiological studies carried out, there is insufficient evidence to establish a conclusive relationship between stress and the incidence/progression of melanomas. However, the experimental and clinical evidence mentioned and discussed in this review do make us suspect the existence of such a relationship. In this sense, and specifically regarding glucocorticoids, we may summarize the following facts: (a) although glucocorticoids are widely used in cancer therapy due to their proapoptotic properties in different tumor cells (i.e., acute and chronic lymphocytic leukemias, Hodgkin’s and non-Hodgkin’s lymphomas, multiple myeloma and breast cancer), they may also induce a yet undefined resistant phenotype which may facilitate tumor growth and metastases. In fact, different studies have demonstrated that glucocorticoids can suppress tumor progression, whereas other investigations reported that glucocorticoids inhibit chemotherapy-induced cancer cell death. This controversial phenomenon may result from different cancer subtypes, differential GR expression, different interactions at the transcription level and the dosage of glucocorticoid given; (b) glucocorticoids, at pathophysiological levels (non-pharmacological), can induce antiapoptotic signals which are associated with cancer resistance; (c) during early progression of skin melanoma metastases, RU486 (mifepristone, a GR antagonist) and VMF (vemurafenib, a BRAF inhibitor approved by the FDA for the treatment of late stage melanoma) induced a drastic metastases regression; (d) treatment at an advanced stage of growth is associated to the development of resistance to RU486 and VMF; (e) this resistance was mechanistically linked to overexpression of specific proteins of the Bcl-2 family (i.e., Bcl-xL and Mcl-1); (f) melanoma resistance was decreased if AKT and NF-κB signaling pathways were blocked; (g) the use of GR antagonists could increase the efficacy of the anti-melanoma immunotherapy. These facts highlight underlying mechanisms by which metastatic melanoma cells resist and adapt to survive. Consequently, it seems evident that elucidating the mechanisms involved in the pathophysiology of melanoma and the response to treatment is essential in advancing towards the development of personalized therapies.

Author Contributions

E.O., R.S.-P. and J.M.E. conceptualization. E.O. and J.M.E. manuscript writing. R.S.-P. manuscript editing. E.O., R.S.-P., R.L.-B., M.O.-C., P.M.-M. and J.M.E. have participated in the collection and analysis of information. All authors have read and agreed to the published version of the manuscript.

Funding

The research of our group in this field was supported by grants from the Ministerio de Economía y Competitividad (SAF2017-83458-R) (Spain), and from the University of Valencia (OTR2016-16618INVES) (Spain).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. O’Brien, K.; Ried, K.; Binjemain, T.; Sali, A. Integrative Approaches to the Treatment of Cancer. Cancers 2022, 14, 5933. [Google Scholar] [CrossRef] [PubMed]
  2. Dai, S.; Mo, Y.; Wang, Y.; Xiang, B.; Liao, Q.; Zhou, M.; Li, X.; Li, Y.; Xiong, W.; Li, G.; et al. Chronic Stress Promotes Cancer Development. Front. Oncol. 2020, 10, 1492. [Google Scholar] [CrossRef] [PubMed]
  3. Eckerling, A.; Ricon-Becker, I.; Sorski, L.; Sandbank, E.; Ben-Eliyahu, S. Stress and Cancer: Mechanisms, Significance and Future Directions. Nat. Rev. Cancer 2021, 21, 767–785. [Google Scholar] [CrossRef] [PubMed]
  4. Mohan, A.; Huybrechts, I.; Michels, N. Psychosocial Stress and Cancer Risk: A Narrative Review. Eur. J. Cancer Prev. Off. J. Eur. Cancer Prev. Organ. ECP 2022, 31, 585–599. [Google Scholar] [CrossRef]
  5. Lopes, J.; Rodrigues, C.M.P.; Gaspar, M.M.; Reis, C.P. Melanoma Management: From Epidemiology to Treatment and Latest Advances. Cancers 2022, 14, 4652. [Google Scholar] [CrossRef]
  6. Moreno-Villanueva, M.; Bürkle, A. Stress Hormone-Mediated DNA Damage Response--Implications for Cellular Senescence and Tumour Progression. Curr. Drug Targets 2016, 17, 398–404. [Google Scholar] [CrossRef]
  7. Flint, M.S.; Baum, A.; Chambers, W.H.; Jenkins, F.J. Induction of DNA Damage, Alteration of DNA Repair and Transcriptional Activation by Stress Hormones. Psychoneuroendocrinology 2007, 32, 470–479. [Google Scholar] [CrossRef]
  8. Perego, M.; Tyurin, V.A.; Tyurina, Y.Y.; Yellets, J.; Nacarelli, T.; Lin, C.; Nefedova, Y.; Kossenkov, A.; Liu, Q.; Sreedhar, S.; et al. Reactivation of Dormant Tumor Cells by Modified Lipids Derived from Stress-Activated Neutrophils. Sci. Transl. Med. 2020, 12, eabb5817. [Google Scholar] [CrossRef]
  9. Feng, Z.; Liu, L.; Zhang, C.; Zheng, T.; Wang, J.; Lin, M.; Zhao, Y.; Wang, X.; Levine, A.J.; Hu, W. Chronic Restraint Stress Attenuates P53 Function and Promotes Tumorigenesis. Proc. Natl. Acad. Sci. USA 2012, 109, 7013–7018. [Google Scholar] [CrossRef] [Green Version]
  10. Tian, D.; Tian, M.; Han, G.; Li, J.-L. Increased Glucocorticoid Receptor Activity and Proliferation in Metastatic Colon Cancer. Sci. Rep. 2019, 9, 11257. [Google Scholar] [CrossRef]
  11. Bakour, N.; Moriarty, F.; Moore, G.; Robson, T.; Annett, S.L. Prognostic Significance of Glucocorticoid Receptor Expression in Cancer: A Systematic Review and Meta-Analysis. Cancers 2021, 13, 1649. [Google Scholar] [CrossRef]
  12. Pan, D.; Kocherginsky, M.; Conzen, S.D. Activation of the Glucocorticoid Receptor Is Associated with Poor Prognosis in Estrogen Receptor-Negative Breast Cancer. Cancer Res. 2011, 71, 6360–6370. [Google Scholar] [CrossRef] [Green Version]
  13. Stringer-Reasor, E.M.; Baker, G.M.; Skor, M.N.; Kocherginsky, M.; Lengyel, E.; Fleming, G.F.; Conzen, S.D. Glucocorticoid Receptor Activation Inhibits Chemotherapy-Induced Cell Death in High-Grade Serous Ovarian Carcinoma. Gynecol. Oncol. 2015, 138, 656–662. [Google Scholar] [CrossRef] [Green Version]
  14. Veneris, J.T.; Darcy, K.M.; Mhawech-Fauceglia, P.; Tian, C.; Lengyel, E.; Lastra, R.R.; Pejovic, T.; Conzen, S.D.; Fleming, G.F. High Glucocorticoid Receptor Expression Predicts Short Progression-Free Survival in Ovarian Cancer. Gynecol. Oncol. 2017, 146, 153–160. [Google Scholar] [CrossRef]
  15. Tangen, I.L.; Veneris, J.T.; Halle, M.K.; Werner, H.M.; Trovik, J.; Akslen, L.A.; Salvesen, H.B.; Conzen, S.D.; Fleming, G.F.; Krakstad, C. Expression of Glucocorticoid Receptor Is Associated with Aggressive Primary Endometrial Cancer and Increases from Primary to Metastatic Lesions. Gynecol. Oncol. 2017, 147, 672–677. [Google Scholar] [CrossRef]
  16. Han, G.H.; Yun, H.; Kim, J.; Chung, J.-Y.; Kim, J.-H.; Cho, H. Overexpression of Glucocorticoid Receptor Promotes the Poor Progression and Induces Cisplatin Resistance through P38 MAP Kinase in Cervical Cancer Patients. Am. J. Cancer Res. 2022, 12, 3437–3454. [Google Scholar]
  17. Irie, M.; Asami, S.; Nagata, S.; Miyata, M.; Kasai, H. Relationships between Perceived Workload, Stress and Oxidative DNA Damage. Int. Arch. Occup. Environ. Health 2001, 74, 153–157. [Google Scholar] [CrossRef]
  18. Sund, D.T.; Brouwer, A.F.; Walline, H.M.; Carey, T.E.; Meza, R.; Jackson, T.; Eisenberg, M.C. Understanding the Mechanisms of HPV-Related Carcinogenesis: Implications for Cell Cycle Dynamics. J. Theor. Biol. 2022, 551–552, 111235. [Google Scholar] [CrossRef]
  19. Yang, Y.; Yin, L.; Liu, Q.; Sun, J.; Adami, H.-O.; Ye, W.; Zhang, Z.; Fang, F. Hospital-Treated Infections and Increased Risk of Two EBV-Related Malignancies: A Nested Case-Control Study. Cancers 2022, 14, 3804. [Google Scholar] [CrossRef]
  20. Méndez-Solís, O.; Bendjennat, M.; Naipauer, J.; Theodoridis, P.R.; Ho, J.J.D.; Verdun, R.E.; Hare, J.M.; Cesarman, E.; Lee, S.; Mesri, E.A. Kaposi’s Sarcoma Herpesvirus Activates the Hypoxia Response to Usurp HIF2α-Dependent Translation Initiation for Replication and Oncogenesis. Cell Rep. 2021, 37, 110144. [Google Scholar] [CrossRef]
  21. Elpek, G.O. Molecular Pathways in Viral Hepatitis-Associated Liver Carcinogenesis: An Update. World J. Clin. Cases 2021, 9, 4890–4917. [Google Scholar] [CrossRef] [PubMed]
  22. Sheridan, J.F.; Dobbs, C.; Jung, J.; Chu, X.; Konstantinos, A.; Padgett, D.; Glaser, R. Stress-Induced Neuroendocrine Modulation of Viral Pathogenesis and Immunity. Ann. N. Y. Acad. Sci. 1998, 840, 803–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ives, A.M.; Bertke, A.S. Stress Hormones Epinephrine and Corticosterone Selectively Modulate Herpes Simplex Virus 1 (HSV-1) and HSV-2 Productive Infections in Adult Sympathetic, but Not Sensory, Neurons. J. Virol. 2017, 91, e00582-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Melnikova, V.O.; Bar-Eli, M. Inflammation and Melanoma Metastasis. Pigment Cell Melanoma Res. 2009, 22, 257–267. [Google Scholar] [CrossRef] [PubMed]
  25. Auphan, N.; DiDonato, J.A.; Rosette, C.; Helmberg, A.; Karin, M. Immunosuppression by Glucocorticoids: Inhibition of NF-Kappa B Activity through Induction of I Kappa B Synthesis. Science 1995, 270, 286–290. [Google Scholar] [CrossRef]
  26. Kundu, J.K.; Surh, Y.-J. Emerging Avenues Linking Inflammation and Cancer. Free Radic. Biol. Med. 2012, 52, 2013–2037. [Google Scholar] [CrossRef]
  27. Mravec, B.; Tibensky, M.; Horvathova, L. Stress and Cancer. Part II: Therapeutic Implications for Oncology. J. Neuroimmunol. 2020, 346, 577312. [Google Scholar] [CrossRef]
  28. Gosain, R.; Gage-Bouchard, E.; Ambrosone, C.; Repasky, E.; Gandhi, S. Stress Reduction Strategies in Breast Cancer: Review of Pharmacologic and Non-Pharmacologic Based Strategies. Semin. Immunopathol. 2020, 42, 719–734. [Google Scholar] [CrossRef]
  29. 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. [Google Scholar] [CrossRef] [Green Version]
  30. Narendhirakannan, R.T.; Hannah, M.A.C. Oxidative Stress and Skin Cancer: An Overview. Indian J. Clin. Biochem. IJCB 2013, 28, 110–115. [Google Scholar] [CrossRef] [Green Version]
  31. Yang, E.V.; Kim, S.; Donovan, E.L.; Chen, M.; Gross, A.C.; Webster Marketon, J.I.; Barsky, S.H.; Glaser, R. Norepinephrine Upregulates VEGF, IL-8, and IL-6 Expression in Human Melanoma Tumor Cell Lines: Implications for Stress-Related Enhancement of Tumor Progression. Brain. Behav. Immun. 2009, 23, 267–275. [Google Scholar] [CrossRef]
  32. Sinnya, S.; De’Ambrosis, B. Stress and Melanoma: Increasing the Evidence towards a Causal Basis. Arch. Dermatol. Res. 2013, 305, 851–856. [Google Scholar] [CrossRef]
  33. Longo, C.; Pellacani, G. Melanomas. Dermatol. Clin. 2016, 34, 411–419. [Google Scholar] [CrossRef]
  34. Khan, J.; Ullah, A.; Matolo, N.; Waheed, A.; Nama, N.; Sharma, N.; Ballur, K.; Gilstrap, L.; Singh, S.G.; Ghleilib, I.; et al. Prognostic Value of Lymph Node Ratio in Cutaneous Melanoma: A Systematic Review. Cureus 2021, 13, e19117. [Google Scholar] [CrossRef]
  35. Corrie, P.; Meyer, N.; Berardi, R.; Guidoboni, M.; Schlueter, M.; Kolovos, S.; Macabeo, B.; Trouiller, J.-B.; Laramée, P. Comparative Efficacy and Safety of Targeted Therapies for BRAF-Mutant Unresectable or Metastatic Melanoma: Results from a Systematic Literature Review and a Network Meta-Analysis. Cancer Treat. Rev. 2022, 110, 102463. [Google Scholar] [CrossRef]
  36. Kasakovski, D.; Skrygan, M.; Gambichler, T.; Susok, L. Advances in Targeting Cutaneous Melanoma. Cancers 2021, 13, 2090. [Google Scholar] [CrossRef]
  37. Smalley, K.S.M.; Flaherty, K.T. Integrating BRAF/MEK Inhibitors into Combination Therapy for Melanoma. Br. J. Cancer 2009, 100, 431–435. [Google Scholar] [CrossRef] [Green Version]
  38. Morganti, S.; Curigliano, G. Combinations Using Checkpoint Blockade to Overcome Resistance. eCancerMedicalScience 2020, 14, 1148. [Google Scholar] [CrossRef]
  39. Queirolo, P.; Boutros, A.; Tanda, E.; Spagnolo, F.; Quaglino, P. Immune-Checkpoint Inhibitors for the Treatment of Metastatic Melanoma: A Model of Cancer Immunotherapy. Semin. Cancer Biol. 2019, 59, 290–297. [Google Scholar] [CrossRef]
  40. Atkins, M.B.; Lee, S.J.; Chmielowski, B.; Tarhini, A.A.; Cohen, G.I.; Truong, T.-G.; Moon, H.H.; Davar, D.; O’Rourke, M.; Stephenson, J.J.; et al. Combination Dabrafenib and Trametinib Versus Combination Nivolumab and Ipilimumab for Patients with Advanced BRAF-Mutant Melanoma: The DREAMseq Trial—ECOG-ACRIN EA6134. J. Clin. Oncol. 2023, 41, 186–197. [Google Scholar] [CrossRef]
  41. Moser, J.C.; Wei, G.; Colonna, S.V.; Grossmann, K.F.; Patel, S.; Hyngstrom, J.R. Comparative-Effectiveness of Pembrolizumab vs. Nivolumab for Patients with Metastatic Melanoma. Acta Oncol. Stockh. Swed. 2020, 59, 434–437. [Google Scholar] [CrossRef] [PubMed]
  42. Kuryk, L.; Bertinato, L.; Staniszewska, M.; Pancer, K.; Wieczorek, M.; Salmaso, S.; Caliceti, P.; Garofalo, M. From Conventional Therapies to Immunotherapy: Melanoma Treatment in Review. Cancers 2020, 12, 3057. [Google Scholar] [CrossRef] [PubMed]
  43. Fujimura, T.; Muto, Y.; Asano, Y. Immunotherapy for Melanoma: The Significance of Immune Checkpoint Inhibitors for the Treatment of Advanced Melanoma. Int. J. Mol. Sci. 2022, 23, 15720. [Google Scholar] [CrossRef] [PubMed]
  44. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Long-Term Outcomes with Nivolumab Plus Ipilimumab or Nivolumab Alone Versus Ipilimumab in Patients with Advanced Melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 127–137. [Google Scholar] [CrossRef]
  45. Ziogas, D.C.; Theocharopoulos, C.; Koutouratsas, T.; Haanen, J.; Gogas, H. Mechanisms of Resistance to Immune Checkpoint Inhibitors in Melanoma: What We Have to Overcome? Cancer Treat. Rev. 2022, 113, 102499. [Google Scholar] [CrossRef]
  46. Wang, H.; Tran, T.T.; Duong, K.T.; Nguyen, T.; Le, U.M. Options of Therapeutics and Novel Delivery Systems of Drugs for the Treatment of Melanoma. Mol. Pharm. 2022, 19, 4487–4505. [Google Scholar] [CrossRef]
  47. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef]
  48. Milkovic, L.; Siems, W.; Siems, R.; Zarkovic, N. Oxidative Stress and Antioxidants in Carcinogenesis and Integrative Therapy of Cancer. Curr. Pharm. Des. 2014, 20, 6529–6542. [Google Scholar] [CrossRef]
  49. Gill, J.G.; Piskounova, E.; Morrison, S.J. Cancer, Oxidative Stress, and Metastasis. Cold Spring Harb. Symp. Quant. Biol. 2016, 81, 163–175. [Google Scholar] [CrossRef] [Green Version]
  50. Ekshyyan, O.; Aw, T.Y. Decreased Susceptibility of Differentiated PC12 Cells to Oxidative Challenge: Relationship to Cellular Redox and Expression of Apoptotic Protease Activator Factor-1. Cell Death Differ. 2005, 12, 1066–1077. [Google Scholar] [CrossRef]
  51. Szatrowski, T.P.; Nathan, C.F. Production of Large Amounts of Hydrogen Peroxide by Human Tumor Cells. Cancer Res. 1991, 51, 794–798. [Google Scholar]
  52. Meierjohann, S. Oxidative Stress in Melanocyte Senescence and Melanoma Transformation. Eur. J. Cell Biol. 2014, 93, 36–41. [Google Scholar] [CrossRef]
  53. Liu-Smith, F.; Dellinger, R.; Meyskens, F.L. Updates of Reactive Oxygen Species in Melanoma Etiology and Progression. Arch. Biochem. Biophys. 2014, 563, 51–55. [Google Scholar] [CrossRef] [Green Version]
  54. Denat, L.; Kadekaro, A.L.; Marrot, L.; Leachman, S.A.; Abdel-Malek, Z.A. Melanocytes as Instigators and Victims of Oxidative Stress. J. Investig. Dermatol. 2014, 134, 1512–1518. [Google Scholar] [CrossRef] [Green Version]
  55. Karg, E.; Odh, G.; Wittbjer, A.; Rosengren, E.; Rorsman, H. Hydrogen Peroxide as an Inducer of Elevated Tyrosinase Level in Melanoma Cells. J. Investig. Dermatol. 1993, 100, 209S–213S. [Google Scholar] [CrossRef]
  56. Jenkins, N.C.; Grossman, D. Role of Melanin in Melanocyte Dysregulation of Reactive Oxygen Species. BioMed Res. Int. 2013, 2013, 908797. [Google Scholar] [CrossRef]
  57. Meyskens, F.L.; 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. [Google Scholar] [CrossRef] [Green Version]
  58. Gidanian, S.; Mentelle, M.; Meyskens, F.L.; Farmer, P.J. Melanosomal Damage in Normal Human Melanocytes Induced by UVB and Metal Uptake--a Basis for the pro-Oxidant State of Melanoma. Photochem. Photobiol. 2008, 84, 556–564. [Google Scholar] [CrossRef] [Green Version]
  59. Carpenter, E.L.; Becker, A.L.; Indra, A.K. NRF2 and Key Transcriptional Targets in Melanoma Redox Manipulation. Cancers 2022, 14, 1531. [Google Scholar] [CrossRef]
  60. Valente, V.B.; de Melo Cardoso, D.; Kayahara, G.M.; Nunes, G.B.; Tjioe, K.C.; Biasoli, É.R.; Miyahara, G.I.; Oliveira, S.H.P.; Mingoti, G.Z.; Bernabé, D.G. Stress Hormones Promote DNA Damage in Human Oral Keratinocytes. Sci. Rep. 2021, 11, 19701. [Google Scholar] [CrossRef]
  61. Yamanishi, D.T.; Buckmeier, J.A.; Meyskens, F.L. Expression of C-Jun, Jun-B, and c-Fos Proto-Oncogenes in Human Primary Melanocytes and Metastatic Melanomas. J. Investig. Dermatol. 1991, 97, 349–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Meyskens, F.L.; Chau, H.V.; Tohidian, N.; Buckmeier, J. Luminol-Enhanced Chemiluminescent Response of Human Melanocytes and Melanoma Cells to Hydrogen Peroxide Stress. Pigment Cell Res. 1997, 10, 184–189. [Google Scholar] [CrossRef] [PubMed]
  63. Sander, C.S.; Chang, H.; Hamm, F.; Elsner, P.; Thiele, J.J. Role of Oxidative Stress and the Antioxidant Network in Cutaneous Carcinogenesis. Int. J. Dermatol. 2004, 43, 326–335. [Google Scholar] [CrossRef] [PubMed]
  64. Ortega, A.; Carretero, J.; Obrador, E.; Estrela, J.M. Tumoricidal Activity of Endothelium-Derived NO and the Survival of Metastatic Cells with High GSH and Bcl-2 Levels. Nitric Oxide Biol. Chem. 2008, 19, 107–114. [Google Scholar] [CrossRef]
  65. Liu, F.; Gomez Garcia, A.M.; Meyskens, F.L. NADPH Oxidase 1 Overexpression Enhances Invasion via Matrix Metalloproteinase-2 and Epithelial-Mesenchymal Transition in Melanoma Cells. J. Investig. Dermatol. 2012, 132, 2033–2041. [Google Scholar] [CrossRef] [Green Version]
  66. Obrador, E.; Salvador, R.; López-Blanch, R.; Jihad-Jebbar, A.; Alcácer, J.; Benlloch, M.; Pellicer, J.A.; Estrela, J.M. Melanoma in the Liver: Oxidative Stress and the Mechanisms of Metastatic Cell Survival. Semin. Cancer Biol. 2021, 71, 109–121. [Google Scholar] [CrossRef]
  67. Sanzo, M.; Colucci, R.; Arunachalam, M.; Berti, S.; Moretti, S. Stress as a Possible Mechanism in Melanoma Progression. Dermatol. Res. Pract. 2010, 2010, 483493. [Google Scholar] [CrossRef]
  68. Rigel, D.S. Epidemiology of Melanoma. Semin. Cutan. Med. Surg. 2010, 29, 204–209. [Google Scholar] [CrossRef]
  69. Simonetti, O.; Lucarini, G.; Brancorsini, D.; Nita, P.; Bernardini, M.L.; Biagini, G.; Offidani, A. Immunohistochemical Expression of Vascular Endothelial Growth Factor, Matrix Metalloproteinase 2, and Matrix Metalloproteinase 9 in Cutaneous Melanocytic Lesions. Cancer 2002, 95, 1963–1970. [Google Scholar] [CrossRef]
  70. Hoejberg, L.; Bastholt, L.; Schmidt, H. Interleukin-6 and Melanoma. Melanoma Res. 2012, 22, 327–333. [Google Scholar] [CrossRef]
  71. Moreno-Smith, M.; Lutgendorf, S.K.; Sood, A.K. Impact of Stress on Cancer Metastasis. Future Oncol. Lond. Engl. 2010, 6, 1863–1881. [Google Scholar] [CrossRef] [Green Version]
  72. Manore, S.G.; Doheny, D.L.; Wong, G.L.; Lo, H.-W. IL-6/JAK/STAT3 Signaling in Breast Cancer Metastasis: Biology and Treatment. Front. Oncol. 2022, 12, 866014. [Google Scholar] [CrossRef]
  73. Obrador, E.; Benlloch, M.; Pellicer, J.A.; Asensi, M.; Estrela, J.M. Intertissue Flow of Glutathione (GSH) as a Tumor Growth-Promoting Mechanism: Interleukin 6 Induces GSH Release from Hepatocytes in Metastatic B16 Melanoma-Bearing Mice. J. Biol. Chem. 2011, 286, 15716–15727. [Google Scholar] [CrossRef] [Green Version]
  74. Meister, A. Selective Modification of Glutathione Metabolism. Science 1983, 220, 472–477. [Google Scholar] [CrossRef]
  75. Hanigan, M.H. Expression of Gamma-Glutamyl Transpeptidase Provides Tumor Cells with a Selective Growth Advantage at Physiologic Concentrations of Cyst(e)Ine. Carcinogenesis 1995, 16, 181–185. [Google Scholar] [CrossRef]
  76. Zhang, H.; Forman, H.J.; Choi, J. Gamma-Glutamyl Transpeptidase in Glutathione Biosynthesis. Methods Enzymol. 2005, 401, 468–483. [Google Scholar] [CrossRef]
  77. Meister, A. Glutathione Deficiency Produced by Inhibition of Its Synthesis, and Its Reversal; Applications in Research and Therapy. Pharmacol. Ther. 1991, 51, 155–194. [Google Scholar] [CrossRef]
  78. Obrador, E.; Carretero, J.; Ortega, A.; Medina, I.; Rodilla, V.; Pellicer, J.A.; Estrela, J.M. Gamma-Glutamyl Transpeptidase Overexpression Increases Metastatic Growth of B16 Melanoma Cells in the Mouse Liver. Hepatol. Baltim. Md 2002, 35, 74–81. [Google Scholar] [CrossRef] [Green Version]
  79. Ookhtens, M.; Kaplowitz, N. Role of the Liver in Interorgan Homeostasis of Glutathione and Cyst(e)Ine. Semin. Liver Dis. 1998, 18, 313–329. [Google Scholar] [CrossRef]
  80. Sternberg, E.M. Neural-Immune Interactions in Health and Disease. J. Clin. Investig. 1997, 100, 2641–2647. [Google Scholar] [CrossRef]
  81. Besedovsky, H.O.; del Rey, A.; Klusman, I.; Furukawa, H.; Monge Arditi, G.; Kabiersch, A. Cytokines as Modulators of the Hypothalamus-Pituitary-Adrenal Axis. J. Steroid Biochem. Mol. Biol. 1991, 40, 613–618. [Google Scholar] [CrossRef] [PubMed]
  82. Fauci, A.S. Mechanisms of the Immunosuppressive and Anti-Inflammatory Effects of Glucocorticosteroids. J. Immunopharmacol. 1978, 1, 1–25. [Google Scholar] [CrossRef] [PubMed]
  83. Bernabé, D.G.; Tamae, A.C.; Biasoli, É.R.; Oliveira, S.H.P. Stress Hormones Increase Cell Proliferation and Regulates Interleukin-6 Secretion in Human Oral Squamous Cell Carcinoma Cells. Brain. Behav. Immun. 2011, 25, 574–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Schrepf, A.; Thaker, P.H.; Goodheart, M.J.; Bender, D.; Slavich, G.M.; Dahmoush, L.; Penedo, F.; DeGeest, K.; Mendez, L.; Lubaroff, D.M.; et al. Diurnal Cortisol and Survival in Epithelial Ovarian Cancer. Psychoneuroendocrinology 2015, 53, 256–267. [Google Scholar] [CrossRef] [Green Version]
  85. Flint, T.R.; Janowitz, T.; Connell, C.M.; Roberts, E.W.; Denton, A.E.; Coll, A.P.; Jodrell, D.I.; Fearon, D.T. Tumor-Induced IL-6 Reprograms Host Metabolism to Suppress Anti-Tumor Immunity. Cell Metab. 2016, 24, 672–684. [Google Scholar] [CrossRef]
  86. Pufall, M.A. Glucocorticoids and Cancer. Adv. Exp. Med. Biol. 2015, 872, 315–333. [Google Scholar] [CrossRef] [Green Version]
  87. Greenstein, S.; Ghias, K.; Krett, N.L.; Rosen, S.T. Mechanisms of Glucocorticoid-Mediated Apoptosis in Hematological Malignancies. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2002, 8, 1681–1694. [Google Scholar]
  88. Wang, Z.; Malone, M.H.; He, H.; McColl, K.S.; Distelhorst, C.W. Microarray Analysis Uncovers the Induction of the Proapoptotic BH3-Only Protein Bim in Multiple Models of Glucocorticoid-Induced Apoptosis. J. Biol. Chem. 2003, 278, 23861–23867. [Google Scholar] [CrossRef] [Green Version]
  89. Volden, P.A.; Conzen, S.D. The Influence of Glucocorticoid Signaling on Tumor Progression. Brain Behav. Immun. 2013, 30, S26–S31. [Google Scholar] [CrossRef] [Green Version]
  90. Karvonen, H.; Arjama, M.; Kaleva, L.; Niininen, W.; Barker, H.; Koivisto-Korander, R.; Tapper, J.; Pakarinen, P.; Lassus, H.; Loukovaara, M.; et al. Glucocorticoids Induce Differentiation and Chemoresistance in Ovarian Cancer by Promoting ROR1-Mediated Stemness. Cell Death Dis. 2020, 11, 790. [Google Scholar] [CrossRef]
  91. Zhang, C.; Beckermann, B.; Kallifatidis, G.; Liu, Z.; Rittgen, W.; Edler, L.; Büchler, P.; Debatin, K.-M.; Büchler, M.W.; Friess, H.; et al. Corticosteroids Induce Chemotherapy Resistance in the Majority of Tumour Cells from Bone, Brain, Breast, Cervix, Melanoma and Neuroblastoma. Int. J. Oncol. 2006, 29, 1295–1301. [Google Scholar] [CrossRef] [Green Version]
  92. Kalfeist, L.; Galland, L.; Ledys, F.; Ghiringhelli, F.; Limagne, E.; Ladoire, S. Impact of Glucocorticoid Use in Oncology in the Immunotherapy Era. Cells 2022, 11, 770. [Google Scholar] [CrossRef]
  93. Adorisio, S.; Cannarile, L.; Delfino, D.V.; Ayroldi, E. Glucocorticoid and PD-1 Cross-Talk: Does the Immune System Become Confused? Cells 2021, 10, 2333. [Google Scholar] [CrossRef]
  94. Moretti, S.; Massi, D.; Farini, V.; Baroni, G.; Parri, M.; Innocenti, S.; Cecchi, R.; Chiarugi, P. β-Adrenoceptors Are Upregulated in Human Melanoma and Their Activation Releases pro-Tumorigenic Cytokines and Metalloproteases in Melanoma Cell Lines. Lab. Investig. J. Tech. Methods Pathol. 2013, 93, 279–290. [Google Scholar] [CrossRef] [Green Version]
  95. Maccari, S.; Buoncervello, M.; Ascione, B.; Stati, T.; Macchia, D.; Fidanza, S.; Catalano, L.; Matarrese, P.; Gabriele, L.; Marano, G. α-Adrenoceptor Stimulation Attenuates Melanoma Growth in Mice. Br. J. Pharmacol. 2022, 179, 1371–1383. [Google Scholar] [CrossRef]
  96. Caruntu, C. Catecholamines Increase In Vitro Proliferation of Murine B16F10 Melanoma Cells. Acta Endocrinol. Buchar. 2014, 10, 545–558. [Google Scholar] [CrossRef] [Green Version]
  97. De Giorgi, V.; Geppetti, P.; Lupi, C.; Benemei, S. The Role of β-Blockers in Melanoma. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2020, 15, 17–26. [Google Scholar] [CrossRef]
  98. Valles, S.L.; Benlloch, M.; Rodriguez, M.L.; Mena, S.; Pellicer, J.A.; Asensi, M.; Obrador, E.; Estrela, J.M. Stress Hormones Promote Growth of B16-F10 Melanoma Metastases: An Interleukin 6- and Glutathione-Dependent Mechanism. J. Transl. Med. 2013, 11, 72. [Google Scholar] [CrossRef] [Green Version]
  99. Kumari, N.; Dwarakanath, B.S.; Das, A.; Bhatt, A.N. Role of Interleukin-6 in Cancer Progression and Therapeutic Resistance. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2016, 37, 11553–11572. [Google Scholar] [CrossRef]
  100. Friedmann Angeli, J.P.; Meierjohann, S. NRF2-Dependent Stress Defense in Tumor Antioxidant Control and Immune Evasion. Pigment Cell Melanoma Res. 2021, 34, 268–279. [Google Scholar] [CrossRef]
  101. Jeayeng, S.; Wongkajornsilp, A.; Slominski, A.T.; Jirawatnotai, S.; Sampattavanich, S.; Panich, U. Nrf2 in Keratinocytes Modulates UVB-Induced DNA Damage and Apoptosis in Melanocytes through MAPK Signaling. Free Radic. Biol. Med. 2017, 108, 918–928. [Google Scholar] [CrossRef] [PubMed]
  102. Nguyen, T.; Sherratt, P.J.; Pickett, C.B. Regulatory Mechanisms Controlling Gene Expression Mediated by the Antioxidant Response Element. Annu. Rev. Pharmacol. Toxicol. 2003, 43, 233–260. [Google Scholar] [CrossRef] [PubMed]
  103. DeNicola, G.M.; Karreth, F.A.; Humpton, T.J.; Gopinathan, A.; Wei, C.; Frese, K.; Mangal, D.; Yu, K.H.; Yeo, C.J.; Calhoun, E.S.; et al. Oncogene-Induced Nrf2 Transcription Promotes ROS Detoxification and Tumorigenesis. Nature 2011, 475, 106–109. [Google Scholar] [CrossRef] [PubMed]
  104. Rocha, C.R.R.; Kajitani, G.S.; Quinet, A.; Fortunato, R.S.; Menck, C.F.M. NRF2 and Glutathione Are Key Resistance Mediators to Temozolomide in Glioma and Melanoma Cells. Oncotarget 2016, 7, 48081–48092. [Google Scholar] [CrossRef] [Green Version]
  105. Schmidlin, C.J.; Tian, W.; Dodson, M.; Chapman, E.; Zhang, D.D. FAM129B-Dependent Activation of NRF2 Promotes an Invasive Phenotype in BRAF Mutant Melanoma Cells. Mol. Carcinog. 2021, 60, 331–341. [Google Scholar] [CrossRef]
  106. Hintsala, H.-R.; Jokinen, E.; Haapasaari, K.-M.; Moza, M.; Ristimäki, A.; Soini, Y.; Koivunen, J.; Karihtala, P. Nrf2/Keap1 Pathway and Expression of Oxidative Stress Lesions 8-Hydroxy-2′-Deoxyguanosine and Nitrotyrosine in Melanoma. Anticancer Res. 2016, 36, 1497–1506. [Google Scholar]
  107. Benlloch, M.; Obrador, E.; Valles, S.L.; Rodriguez, M.L.; Sirerol, J.A.; Alcácer, J.; Pellicer, J.A.; Salvador, R.; Cerdá, C.; Sáez, G.T.; et al. Pterostilbene Decreases the Antioxidant Defenses of Aggressive Cancer Cells In Vivo: A Physiological Glucocorticoids- and Nrf2-Dependent Mechanism. Antioxid Redox Signal 2016, 24, 974–990. [Google Scholar] [CrossRef] [Green Version]
  108. Arslanbaeva, L.R.; Santoro, M.M. Adaptive Redox Homeostasis in Cutaneous Melanoma. Redox Biol. 2020, 37, 101753. [Google Scholar] [CrossRef]
  109. Estrela, J.M.; Salvador, R.; Marchio, P.; Valles, S.L.; López-Blanch, R.; Rivera, P.; Benlloch, M.; Alcácer, J.; Pérez, C.L.; Pellicer, J.A.; et al. Glucocorticoid Receptor Antagonism Overcomes Resistance to BRAF Inhibition in BRAFV600E-Mutated Metastatic Melanoma. Am. J. Cancer Res. 2019, 9, 2580–2598. [Google Scholar]
  110. Beberok, A.; Wrześniok, D.; Szlachta, M.; Rok, J.; Rzepka, Z.; Respondek, M.; Buszman, E. Lomefloxacin Induces Oxidative Stress and Apoptosis in COLO829 Melanoma Cells. Int. J. Mol. Sci. 2017, 18, 2194. [Google Scholar] [CrossRef]
  111. Obrador, E.; Salvador-Palmer, R.; López-Blanch, R.; Oriol-Caballo, M.; Moreno-Murciano, P.; Estrela, J.M. N-Acetylcysteine Promotes Metastatic Spread of Melanoma in Mice. Cancers 2022, 14, 3614. [Google Scholar] [CrossRef]
  112. Rojo de la Vega, M.; Chapman, E.; Zhang, D.D. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018, 34, 21–43. [Google Scholar] [CrossRef]
  113. Gide, T.N.; Quek, C.; Menzies, A.M.; Tasker, A.T.; Shang, P.; Holst, J.; Madore, J.; Lim, S.Y.; Velickovic, R.; Wongchenko, M.; et al. Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy. Cancer Cell 2019, 35, 238–255.e6. [Google Scholar] [CrossRef] [Green Version]
  114. Carlino, M.S.; Larkin, J.; Long, G.V. Immune Checkpoint Inhibitors in Melanoma. Lancet 2021, 398, 1002–1014. [Google Scholar] [CrossRef]
  115. Flaherty, R.L.; Owen, M.; Fagan-Murphy, A.; Intabli, H.; Healy, D.; Patel, A.; Allen, M.C.; Patel, B.A.; Flint, M.S. Glucocorticoids Induce Production of Reactive Oxygen Species/Reactive Nitrogen Species and DNA Damage through an INOS Mediated Pathway in Breast Cancer. Breast Cancer Res. BCR 2017, 19, 35. [Google Scholar] [CrossRef]
  116. Obrador, E.; Valles, S.L.; Benlloch, M.; Sirerol, J.A.; Pellicer, J.A.; Alcácer, J.; Coronado, J.A.-F.; Estrela, J.M. Glucocorticoid Receptor Knockdown Decreases the Antioxidant Protection of B16 Melanoma Cells: An Endocrine System-Related Mechanism That Compromises Metastatic Cell Resistance to Vascular Endothelium-Induced Tumor Cytotoxicity. PLoS ONE 2014, 9, e96466. [Google Scholar] [CrossRef]
  117. Rotblat, B.; Melino, G.; Knight, R.A. NRF2 and P53: Januses in Cancer? Oncotarget 2012, 3, 1272–1283. [Google Scholar] [CrossRef] [Green Version]
  118. Kitamura, H.; Motohashi, H. NRF2 Addiction in Cancer Cells. Cancer Sci. 2018, 109, 900–911. [Google Scholar] [CrossRef] [Green Version]
  119. 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. [Google Scholar] [CrossRef]
  120. Alam, M.M.; Okazaki, K.; Nguyen, L.T.T.; Ota, N.; Kitamura, H.; Murakami, S.; Shima, H.; Igarashi, K.; Sekine, H.; Motohashi, H. Glucocorticoid Receptor Signaling Represses the Antioxidant Response by Inhibiting Histone Acetylation Mediated by the Transcriptional Activator NRF2. J. Biol. Chem. 2017, 292, 7519–7530. [Google Scholar] [CrossRef] [Green Version]
  121. Ki, S.H.; Cho, I.J.; Choi, D.W.; Kim, S.G. Glucocorticoid Receptor (GR)-Associated SMRT Binding to C/EBPbeta TAD and Nrf2 Neh4/5: Role of SMRT Recruited to GR in GSTA2 Gene Repression. Mol. Cell. Biol. 2005, 25, 4150–4165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Giudice, A.; Aliberti, S.M.; Barbieri, A.; Pentangelo, P.; Bisogno, I.; D’Arena, G.; Cianciola, E.; Caraglia, M.; Capunzo, M. Potential Mechanisms by Which Glucocorticoids Induce Breast Carcinogenesis through Nrf2 Inhibition. Front. Biosci. Landmark Ed. 2022, 27, 223. [Google Scholar] [CrossRef] [PubMed]
  123. Oakley, R.H.; Cidlowski, J.A. The Biology of the Glucocorticoid Receptor: New Signaling Mechanisms in Health and Disease. J. Allergy Clin. Immunol. 2013, 132, 1033–1044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Gonçalves, L.; Dafre, A.L.; Carobrez, S.G.; Gasparotto, O.C. A Temporal Analysis of the Relationships between Social Stress, Humoral Immune Response and Glutathione-Related Antioxidant Defenses. Behav. Brain Res. 2008, 192, 226–231. [Google Scholar] [CrossRef]
  125. Jafari, M.; Salehi, M.; Zardooz, H.; Rostamkhani, F. Response of Liver Antioxidant Defense System to Acute and Chronic Physical and Psychological Stresses in Male Rats. EXCLI J. 2014, 13, 161–171. [Google Scholar]
  126. Costantini, D.; Marasco, V.; Møller, A.P. A Meta-Analysis of Glucocorticoids as Modulators of Oxidative Stress in Vertebrates. J. Comp. Physiol. 2011, 181, 447–456. [Google Scholar] [CrossRef]
  127. Schlossmacher, G.; Stevens, A.; White, A. Glucocorticoid Receptor-Mediated Apoptosis: Mechanisms of Resistance in Cancer Cells. J. Endocrinol. 2011, 211, 17–25. [Google Scholar] [CrossRef] [Green Version]
  128. Obradović, M.M.S.; Hamelin, B.; Manevski, N.; Couto, J.P.; Sethi, A.; Coissieux, M.-M.; Münst, S.; Okamoto, R.; Kohler, H.; Schmidt, A.; et al. Glucocorticoids Promote Breast Cancer Metastasis. Nature 2019, 567, 540–544. [Google Scholar] [CrossRef]
  129. Zhang, C.; Wenger, T.; Mattern, J.; Ilea, S.; Frey, C.; Gutwein, P.; Altevogt, P.; Bodenmüller, W.; Gassler, N.; Schnabel, P.A.; et al. Clinical and Mechanistic Aspects of Glucocorticoid-Induced Chemotherapy Resistance in the Majority of Solid Tumors. Cancer Biol. Ther. 2007, 6, 278–287. [Google Scholar] [CrossRef] [Green Version]
  130. Herr, I.; Büchler, M.W.; Mattern, J. Glucocorticoid-Mediated Apoptosis Resistance of Solid Tumors. Results Probl. Cell Differ. 2009, 49, 191–218. [Google Scholar] [CrossRef]
  131. Moran, T.J.; Gray, S.; Mikosz, C.A.; Conzen, S.D. The Glucocorticoid Receptor Mediates a Survival Signal in Human Mammary Epithelial Cells. Cancer Res. 2000, 60, 867–872. [Google Scholar]
  132. Mikosz, C.A.; Brickley, D.R.; Sharkey, M.S.; Moran, T.W.; Conzen, S.D. Glucocorticoid Receptor-Mediated Protection from Apoptosis Is Associated with Induction of the Serine/Threonine Survival Kinase Gene, Sgk-1. J. Biol. Chem. 2001, 276, 16649–16654. [Google Scholar] [CrossRef] [Green Version]
  133. Greenstein, A.E.; Hunt, H.J. Glucocorticoid Receptor Antagonism Promotes Apoptosis in Solid Tumor Cells. Oncotarget 2021, 12, 1243–1255. [Google Scholar] [CrossRef]
  134. Chen, W.; Jiang, T.; Wang, H.; Tao, S.; Lau, A.; Fang, D.; Zhang, D.D. Does Nrf2 Contribute to P53-Mediated Control of Cell Survival and Death? Antioxid. Redox Signal. 2012, 17, 1670–1675. [Google Scholar] [CrossRef] [Green Version]
  135. Muller, P.A.J.; Vousden, K.H.; Norman, J.C. P53 and Its Mutants in Tumor Cell Migration and Invasion. J. Cell Biol. 2011, 192, 209–218. [Google Scholar] [CrossRef]
  136. Sredni, B.; Caspi, R.R.; Klein, A.; Kalechman, Y.; Danziger, Y.; Ben Ya’akov, M.; Tamari, T.; Shalit, F.; Albeck, M. A New Immunomodulating Compound (AS-101) with Potential Therapeutic Application. Nature 1987, 330, 173–176. [Google Scholar] [CrossRef]
  137. Derech-Haim, S.; Teiblum, G.; Kadosh, R.; Rahav, G.; Bonda, E.; Sredni, B.; Bakhanashvili, M. Ribonuclease Activity of P53 in Cytoplasm in Response to Various Stress Signals. Cell Cycle Georget. Tex 2012, 11, 1400–1413. [Google Scholar] [CrossRef]
  138. Aziz, M.H.; Shen, H.; Maki, C.G. Glucocorticoid Receptor Activation Inhibits P53-Induced Apoptosis of MCF10Amyc Cells via Induction of Protein Kinase Cε. J. Biol. Chem. 2012, 287, 29825–29836. [Google Scholar] [CrossRef] [Green Version]
  139. Abduljabbar, R.; Negm, O.H.; Lai, C.-F.; Jerjees, D.A.; Al-Kaabi, M.; Hamed, M.R.; Tighe, P.J.; Buluwela, L.; Mukherjee, A.; Green, A.R.; et al. Clinical and Biological Significance of Glucocorticoid Receptor (GR) Expression in Breast Cancer. Breast Cancer Res. Treat. 2015, 150, 335–346. [Google Scholar] [CrossRef]
  140. Yu, C.; Yap, N.; Chen, D.; Cheng, S. Modulation of Hormone-Dependent Transcriptional Activity of the Glucocorticoid Receptor by the Tumor Suppressor P53. Cancer Lett. 1997, 116, 191–196. [Google Scholar] [CrossRef]
  141. Anasagasti, M.J.; Martin, J.J.; Mendoza, L.; Obrador, E.; Estrela, J.M.; McCuskey, R.S.; Vidal-Vanaclocha, F. Glutathione Protects Metastatic Melanoma Cells against Oxidative Stress in the Murine Hepatic Microvasculature. Hepatology 1998, 27, 1249–1256. [Google Scholar] [CrossRef] [PubMed]
  142. Gerber, A.N.; Newton, R.; Sasse, S.K. Repression of Transcription by the Glucocorticoid Receptor: A Parsimonious Model for the Genomics Era. J. Biol. Chem. 2021, 296, 100687. [Google Scholar] [CrossRef] [PubMed]
  143. Kumar, R.; Thompson, E.B. Gene Regulation by the Glucocorticoid Receptor: Structure:Function Relationship. J. Steroid Biochem. Mol. Biol. 2005, 94, 383–394. [Google Scholar] [CrossRef] [PubMed]
  144. Heck, S.; Kullmann, M.; Gast, A.; Ponta, H.; Rahmsdorf, H.J.; Herrlich, P.; Cato, A.C. A Distinct Modulating Domain in Glucocorticoid Receptor Monomers in the Repression of Activity of the Transcription Factor AP-1. EMBO J. 1994, 13, 4087–4095. [Google Scholar] [CrossRef]
  145. Caldenhoven, E.; Liden, J.; Wissink, S.; Van de Stolpe, A.; Raaijmakers, J.; Koenderman, L.; Okret, S.; Gustafsson, J.A.; Van der Saag, P.T. Negative Cross-Talk between RelA and the Glucocorticoid Receptor: A Possible Mechanism for the Antiinflammatory Action of Glucocorticoids. Mol. Endocrinol. 1995, 9, 401–412. [Google Scholar] [CrossRef] [Green Version]
  146. Ito, K.; Jazrawi, E.; Cosio, B.; Barnes, P.J.; Adcock, I.M. P65-Activated Histone Acetyltransferase Activity Is Repressed by Glucocorticoids: Mifepristone Fails to Recruit HDAC2 to the P65-HAT Complex. J. Biol. Chem. 2001, 276, 30208–30215. [Google Scholar] [CrossRef] [Green Version]
  147. Ding, X.F.; Anderson, C.M.; Ma, H.; Hong, H.; Uht, R.M.; Kushner, P.J.; Stallcup, M.R. Nuclear Receptor-Binding Sites of Coactivators Glucocorticoid Receptor Interacting Protein 1 (GRIP1) and Steroid Receptor Coactivator 1 (SRC-1): Multiple Motifs with Different Binding Specificities. Mol. Endocrinol. 1998, 12, 302–313. [Google Scholar] [CrossRef]
  148. Zhang, J.; Tsai, F.T.F.; Geller, D.S. Differential Interaction of RU486 with the Progesterone and Glucocorticoid Receptors. J. Mol. Endocrinol. 2006, 37, 163–173. [Google Scholar] [CrossRef] [Green Version]
  149. Heikinheimo, O.; Kekkonen, R.; Lähteenmäki, P. The Pharmacokinetics of Mifepristone in Humans Reveal Insights into Differential Mechanisms of Antiprogestin Action. Contraception 2003, 68, 421–426. [Google Scholar] [CrossRef]
  150. Baulieu, E.E. The Steroid Hormone Antagonist RU486. Mechanism at the Cellular Level and Clinical Applications. Endocrinol. Metab. Clin. N. Am. 1991, 20, 873–891. [Google Scholar] [CrossRef]
  151. Brogden, R.N.; Goa, K.L.; Faulds, D. Mifepristone. A Review of Its Pharmacodynamic and Pharmacokinetic Properties, and Therapeutic Potential. Drugs 1993, 45, 384–409. [Google Scholar] [CrossRef]
  152. Ramaraj, P.; Cox, J.L. In Vitro Effect of Progesterone on Human Melanoma (BLM) Cell Growth. Int. J. Clin. Exp. Med. 2014, 7, 3941–3953. [Google Scholar]
  153. Ma, M.; Ghosh, S.; Tavernari, D.; Katarkar, A.; Clocchiatti, A.; Mazzeo, L.; Samarkina, A.; Epiney, J.; Yu, Y.-R.; Ho, P.-C.; et al. Sustained Androgen Receptor Signaling Is a Determinant of Melanoma Cell Growth Potential and Tumorigenesis. J. Exp. Med. 2021, 218, e20201137. [Google Scholar] [CrossRef]
  154. Treviño, L.S.; Gorelick, D.A. The Interface of Nuclear and Membrane Steroid Signaling. Endocrinology 2021, 162, bqab107. [Google Scholar] [CrossRef]
  155. Ritch, S.J.; Noman, A.S.M.; Goyeneche, A.A.; Telleria, C.M. The Metastatic Capacity of High-Grade Serous Ovarian Cancer Cells Changes along Disease Progression: Inhibition by Mifepristone. Cancer Cell Int. 2022, 22, 397. [Google Scholar] [CrossRef]
  156. Alvarez, P.B.; Laskaris, A.; Goyeneche, A.A.; Chen, Y.; Telleria, C.M.; Burnier, J.V. Anticancer Effects of Mifepristone on Human Uveal Melanoma Cells. Cancer Cell Int. 2021, 21, 607. [Google Scholar] [CrossRef]
  157. Zheng, N.; Chen, J.; Liu, W.; Wang, J.; Liu, J.; Jia, L. Metapristone (RU486 Derivative) Inhibits Cell Proliferation and Migration as Melanoma Metastatic Chemopreventive Agent. Biomed. Pharmacother. Biomed. Pharmacother. 2017, 90, 339–349. [Google Scholar] [CrossRef]
  158. Mourah, S.; Denis, M.G.; Narducci, F.E.; Solassol, J.; Merlin, J.-L.; Sabourin, J.-C.; Scoazec, J.-Y.; Ouafik, L.; Emile, J.-F.; Heller, R.; et al. Detection of BRAF V600 Mutations in Melanoma: Evaluation of Concordance between the Cobas® 4800 BRAF V600 Mutation Test and the Methods Used in French National Cancer Institute (INCa) Platforms in a Real-Life Setting. PLoS ONE 2015, 10, e0120232. [Google Scholar] [CrossRef]
  159. Carvalho, C.; L’Hôte, V.; Courbeyrette, R.; Kratassiouk, G.; Pinna, G.; Cintrat, J.-C.; Denby-Wilkes, C.; Derbois, C.; Olaso, R.; Deleuze, J.-F.; et al. Glucocorticoids Delay RAF-Induced Senescence Promoted by EGR1. J. Cell Sci. 2019, 132, jcs230748. [Google Scholar] [CrossRef] [Green Version]
  160. Sharma, A.; Shah, S.R.; Illum, H.; Dowell, J. Vemurafenib: Targeted Inhibition of Mutated BRAF for Treatment of Advanced Melanoma and Its Potential in Other Malignancies. Drugs 2012, 72, 2207–2222. [Google Scholar] [CrossRef]
  161. Corazao-Rozas, P.; Guerreschi, P.; Jendoubi, M.; André, F.; Jonneaux, A.; Scalbert, C.; Garçon, G.; Malet-Martino, M.; Balayssac, S.; Rocchi, S.; et al. Mitochondrial Oxidative Stress Is the Achille’s Heel of Melanoma Cells Resistant to Braf-Mutant Inhibitor. Oncotarget 2013, 4, 1986–1998. [Google Scholar] [CrossRef] [Green Version]
  162. López-Cobo, S.; Pieper, N.; Campos-Silva, C.; García-Cuesta, E.M.; Reyburn, H.T.; Paschen, A.; Valés-Gómez, M. Impaired NK Cell Recognition of Vemurafenib-Treated Melanoma Cells Is Overcome by Simultaneous Application of Histone Deacetylase Inhibitors. Oncoimmunology 2018, 7, e1392426. [Google Scholar] [CrossRef] [PubMed]
  163. Sun, Y.; Fang, M.; Davies, H.; Hu, Z. Mifepristone: A Potential Clinical Agent Based on Its Anti-Progesterone and Anti-Glucocorticoid Properties. Gynecol. Endocrinol. Off. J. Int. Soc. Gynecol. Endocrinol. 2014, 30, 169–173. [Google Scholar] [CrossRef] [PubMed]
  164. Manzano, J.L.; Layos, L.; Bugés, C.; de Los Llanos Gil, M.; Vila, L.; Martínez-Balibrea, E.; Martínez-Cardús, A. Resistant Mechanisms to BRAF Inhibitors in Melanoma. Ann. Transl. Med. 2016, 4, 237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Radić, M.; Vlašić, I.; Jazvinšćak Jembrek, M.; Horvat, A.; Tadijan, A.; Sabol, M.; Dužević, M.; Herak Bosnar, M.; Slade, N. Characterization of Vemurafenib-Resistant Melanoma Cell Lines Reveals Novel Hallmarks of Targeted Therapy Resistance. Int. J. Mol. Sci. 2022, 23, 9910. [Google Scholar] [CrossRef]
  166. Daud, A.; Tsai, K. Management of Treatment-Related Adverse Events with Agents Targeting the MAPK Pathway in Patients with Metastatic Melanoma. Oncologist 2017, 22, 823–833. [Google Scholar] [CrossRef] [Green Version]
  167. Crozier, M.; Tubman, J.; Fifield, B.-A.; Ferraiuolo, R.-M.; Ritchie, J.; Zuccato, K.; Mailloux, E.; Sinha, I.; Hamm, C.; Porter, L.A. Frequently Used Antiemetic Agent Dexamethasone Enhances the Metastatic Behaviour of Select Breast Cancer Cells. PLoS ONE 2022, 17, e0274675. [Google Scholar] [CrossRef]
  168. Faggiano, A.; Mazzilli, R.; Natalicchio, A.; Adinolfi, V.; Argentiero, A.; Danesi, R.; D’Oronzo, S.; Fogli, S.; Gallo, M.; Giuffrida, D.; et al. Corticosteroids in Oncology: Use, Overuse, Indications, Contraindications. An Italian Association of Medical Oncology (AIOM)/ Italian Association of Medical Diabetologists (AMD)/ Italian Society of Endocrinology (SIE)/ Italian Society of Pharmacology (SIF) Multidisciplinary Consensus Position Paper. Crit. Rev. Oncol. Hematol. 2022, 180, 103826. [Google Scholar] [CrossRef]
  169. Baron, M.; Tagore, M.; Hunter, M.V.; Kim, I.S.; Moncada, R.; Yan, Y.; Campbell, N.R.; White, R.M.; Yanai, I. The Stress-Like Cancer Cell State Is a Consistent Component of Tumorigenesis. Cell Syst. 2020, 11, 536–546.e7. [Google Scholar] [CrossRef]
  170. Spagnolo, F.; Ghiorzo, P.; Queirolo, P. Overcoming Resistance to BRAF Inhibition in BRAF-Mutated Metastatic Melanoma. Oncotarget 2014, 5, 10206–10221. [Google Scholar] [CrossRef] [Green Version]
  171. Savoia, P.; Zavattaro, E.; Cremona, O. Clinical Implications of Acquired BRAF Inhibitors Resistance in Melanoma. Int. J. Mol. Sci. 2020, 21, 9730. [Google Scholar] [CrossRef]
  172. Welsh, S.J.; Rizos, H.; Scolyer, R.A.; Long, G.V. Resistance to Combination BRAF and MEK Inhibition in Metastatic Melanoma: Where to Next? Eur. J. Cancer 2016, 62, 76–85. [Google Scholar] [CrossRef]
  173. Wu, W.; Pew, T.; Zou, M.; Pang, D.; Conzen, S.D. Glucocorticoid Receptor-Induced MAPK Phosphatase-1 (MPK-1) Expression Inhibits Paclitaxel-Associated MAPK Activation and Contributes to Breast Cancer Cell Survival. J. Biol. Chem. 2005, 280, 4117–4124. [Google Scholar] [CrossRef] [Green Version]
  174. Boucher, M.J.; Morisset, J.; Vachon, P.H.; Reed, J.C.; Lainé, J.; Rivard, N. MEK/ERK Signaling Pathway Regulates the Expression of Bcl-2, Bcl-X(L), and Mcl-1 and Promotes Survival of Human Pancreatic Cancer Cells. J. Cell. Biochem. 2000, 79, 355–369. [Google Scholar] [CrossRef]
  175. Chipuk, J.E. BCL-2 Proteins: Melanoma Lives on the Edge. Oncoscience 2015, 2, 729–730. [Google Scholar] [CrossRef]
  176. Petrella, A.; Ercolino, S.F.; Festa, M.; Gentilella, A.; Tosco, A.; Conzen, S.D.; Parente, L. Dexamethasone Inhibits TRAIL-Induced Apoptosis of Thyroid Cancer Cells via Bcl-XL Induction. Eur. J. Cancer 2006, 42, 3287–3293. [Google Scholar] [CrossRef]
  177. Abulwerdi, F.; Liao, C.; Liu, M.; Azmi, A.S.; Aboukameel, A.; Mady, A.S.A.; Gulappa, T.; Cierpicki, T.; Owens, S.; Zhang, T.; et al. A Novel Small-Molecule Inhibitor of Mcl-1 Blocks Pancreatic Cancer Growth in Vitro and in Vivo. Mol. Cancer Ther. 2014, 13, 565–575. [Google Scholar] [CrossRef] [Green Version]
  178. Lessene, G.; Czabotar, P.E.; Sleebs, B.E.; Zobel, K.; Lowes, K.N.; Adams, J.M.; Baell, J.B.; Colman, P.M.; Deshayes, K.; Fairbrother, W.J.; et al. Structure-Guided Design of a Selective BCL-X(L) Inhibitor. Nat. Chem. Biol. 2013, 9, 390–397. [Google Scholar] [CrossRef]
  179. Rossi, E.D.; Martini, M.; Bizzarro, T.; Schmitt, F.; Longatto-Filho, A.; Larocca, L.M. Somatic Mutations in Solid Tumors: A Spectrum at the Service of Diagnostic Armamentarium or an Indecipherable Puzzle? The Morphological Eyes Looking for BRAF and Somatic Molecular Detections on Cyto-Histological Samples. Oncotarget 2017, 8, 3746–3760. [Google Scholar] [CrossRef]
Figure 1. Pathophysiology of Stress in Metastatic Melanoma. Stress-induced dysregulation of central pacemakers and IL-6 (mainly from metastatic cells) favor the release of pituitary ACTH. IL-6 and catecholamines increase glutathione (GSH) release from the liver. Metastatic cell γ-glutamyl transpeptidase degrades plasma GSH, providing extra cysteine for GSH synthesis. GSH is a main physiological antioxidant involved in promoting metastases growth. Glucocorticoids (GCs) upregulate the Nrf2-dependent defense system in metastatic cells. Stress hormones decrease the immune response and facilitate the provision of nutrients to the growing metastases. Tissue-specific microenvironments or the influence of tumor innervation can also be decisive in the behavior of metastatic cells. ACTH, adrenocorticotropic hormone; CRH, corticotropin releasing hormone; AAs, amino acids, FFAs, free fatty acids.
Figure 1. Pathophysiology of Stress in Metastatic Melanoma. Stress-induced dysregulation of central pacemakers and IL-6 (mainly from metastatic cells) favor the release of pituitary ACTH. IL-6 and catecholamines increase glutathione (GSH) release from the liver. Metastatic cell γ-glutamyl transpeptidase degrades plasma GSH, providing extra cysteine for GSH synthesis. GSH is a main physiological antioxidant involved in promoting metastases growth. Glucocorticoids (GCs) upregulate the Nrf2-dependent defense system in metastatic cells. Stress hormones decrease the immune response and facilitate the provision of nutrients to the growing metastases. Tissue-specific microenvironments or the influence of tumor innervation can also be decisive in the behavior of metastatic cells. ACTH, adrenocorticotropic hormone; CRH, corticotropin releasing hormone; AAs, amino acids, FFAs, free fatty acids.
Cells 12 00418 g001
Figure 2. Glucocorticoids and the Antioxidant Defense of Melanoma Cells. The GR is encoded by the NRC31 gene which can produce a number of receptor isoforms, the GRα being the primary receptor involved in glucocorticoid (GC) signaling. The cytosolic GR complexes with different proteins, i.e., Hsp90, Hsp70 and the FK506-binding protein 4. GCs diffuse through the plasma membrane into the cytoplasm and binds to the GR resulting in the release of heat shock proteins. Based on the two-part model proposed by Gerber et al. [142], the cytoplasmic GR interacts with glucocorticoids, thus causing a conformational change and nuclear translocation. GR interacts with both the DNA and other transcriptional machinery to orchestrate its genomic effects through three main mechanisms: direct binding to glucocorticoid response elements, transcription factor tethering, and binding of composite elements within the DNA [143]. Hypothetically, at high pharmacological levels of GCs, primary repression could result from an excessive amount of GR monomers tethering to the ARE-Nrf2 complex (Nrf2 dimerizes with a basic region-leucine zipper bZIP protein and binds to the ARE to activate gene transcription), then leading to recruitment of corepressors. In this mechanism, GR associates with NF-κB or AP-1 [144,145], thus resulting in repression of their activity in a process attributed to recruitment of transcriptional corepressors such as the nuclear receptor co-repressor 1 and the histone deacetylase 2 [146]. At lower extracellular levels of GCs (pathophysiological levels), GR homodimers (predominantly formed) would recruit coactivators (such as the steroid receptor coactivator-1, SRC-1; or the GR-interacting protein-1, GRIP-1) [147] and induce the transcription of genes encoding antioxidant/defense enzyme activities. This double model may help to reconcile the controversy in the results obtained by different groups.
Figure 2. Glucocorticoids and the Antioxidant Defense of Melanoma Cells. The GR is encoded by the NRC31 gene which can produce a number of receptor isoforms, the GRα being the primary receptor involved in glucocorticoid (GC) signaling. The cytosolic GR complexes with different proteins, i.e., Hsp90, Hsp70 and the FK506-binding protein 4. GCs diffuse through the plasma membrane into the cytoplasm and binds to the GR resulting in the release of heat shock proteins. Based on the two-part model proposed by Gerber et al. [142], the cytoplasmic GR interacts with glucocorticoids, thus causing a conformational change and nuclear translocation. GR interacts with both the DNA and other transcriptional machinery to orchestrate its genomic effects through three main mechanisms: direct binding to glucocorticoid response elements, transcription factor tethering, and binding of composite elements within the DNA [143]. Hypothetically, at high pharmacological levels of GCs, primary repression could result from an excessive amount of GR monomers tethering to the ARE-Nrf2 complex (Nrf2 dimerizes with a basic region-leucine zipper bZIP protein and binds to the ARE to activate gene transcription), then leading to recruitment of corepressors. In this mechanism, GR associates with NF-κB or AP-1 [144,145], thus resulting in repression of their activity in a process attributed to recruitment of transcriptional corepressors such as the nuclear receptor co-repressor 1 and the histone deacetylase 2 [146]. At lower extracellular levels of GCs (pathophysiological levels), GR homodimers (predominantly formed) would recruit coactivators (such as the steroid receptor coactivator-1, SRC-1; or the GR-interacting protein-1, GRIP-1) [147] and induce the transcription of genes encoding antioxidant/defense enzyme activities. This double model may help to reconcile the controversy in the results obtained by different groups.
Cells 12 00418 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Obrador, E.; Salvador-Palmer, R.; López-Blanch, R.; Oriol-Caballo, M.; Moreno-Murciano, P.; Estrela, J.M. Survival Mechanisms of Metastatic Melanoma Cells: The Link between Glucocorticoids and the Nrf2-Dependent Antioxidant Defense System. Cells 2023, 12, 418. https://doi.org/10.3390/cells12030418

AMA Style

Obrador E, Salvador-Palmer R, López-Blanch R, Oriol-Caballo M, Moreno-Murciano P, Estrela JM. Survival Mechanisms of Metastatic Melanoma Cells: The Link between Glucocorticoids and the Nrf2-Dependent Antioxidant Defense System. Cells. 2023; 12(3):418. https://doi.org/10.3390/cells12030418

Chicago/Turabian Style

Obrador, Elena, Rosario Salvador-Palmer, Rafael López-Blanch, María Oriol-Caballo, Paz Moreno-Murciano, and José M. Estrela. 2023. "Survival Mechanisms of Metastatic Melanoma Cells: The Link between Glucocorticoids and the Nrf2-Dependent Antioxidant Defense System" Cells 12, no. 3: 418. https://doi.org/10.3390/cells12030418

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop