- Review
- Open access
- Published:
Unraveling the role of hypoxia-inducible factors in cutaneous melanoma: from mechanisms to therapeutic opportunities
Cell Communication and Signaling volume 23, Article number: 177 (2025)
Abstract
Hypoxia is a common feature of solid malignancies, including cutaneous melanoma (CM). Hypoxia-inducible factor (HIF)-1α and HIF-2α orchestrate cellular responses to hypoxia and coordinate a transcriptional program that promote several aggressive features in CM, such as angiogenesis, epithelial-mesenchymal transition, metastasis formation, metabolic rewiring, and immune escape. BRAFV600E, which is the most frequent mutation observed in CM patients, usually increases HIF-α signaling not only in hypoxia, but also in normoxic CM cells, enabling HIF-1α and HIF-2α to continuously activate downstream molecular pathways. In this review, we aim to provide a comprehensive overview of the intricate role and regulation of HIF-1α and HIF-2α in CM, with a brief focus on the complex interactions between HIF-α subunits and non-coding RNAs. We also discuss HIF-α-mediated cellular responses in normoxia along with the mechanisms that allow HIF-α subunits to maintain their stability under normal oxygen conditions. Finally, we resume available evidence on potential therapeutic approaches aimed at targeting HIF-1α and/or HIF-2α.
Introduction
Oxygen is crucial for cell survival and function. Therefore, a prolonged reduction of oxygen availability (hypoxia) usually represents a lethal condition for cells and tissues. On the contrary, about 50–60% of solid tumors are commonly affected by hypoxia [1]. In fact, the rapid and uncontrolled growth of tumor cells commonly outstrips the oxygen supply from the preexisting blood vessels [2].
The adaptive response to hypoxia is mainly mediated by the hypoxia-inducible factors 1 (HIF-1), 2 (HIF-2), and 3 (HIF-3). HIF proteins are heterodimers composed of an oxygen sensitive α subunit (HIF-1α, -2α or -3α) and the HIF-1β subunit, which is constitutively expressed, independently from oxygen levels. In contrast, in normoxia, cytoplasmic α subunits show a very short half-life, since they are continuously degraded via the ubiquitin-proteasome system, whereas under hypoxic conditions, α subunits are stabilized via post-translational modifications and translocated into the nucleus, where they dimerize with HIF-1β [3].
HIF-1 was initially discovered through a study on the erythropoietin gene [4]. Subsequently, the structural analysis of the HIF-1α protein revealed four distinct domains: a basic helix-loop-helix (bHLH) domain for DNA binding and dimerization, two Per-ARNT-Sim (PAS) domains for dimerization and target gene specificity, an oxygen-dependent degradation (ODD) domain, and two transactivation domains (N-TAD and C-TAD) [5]. HIF-2α is closely related to HIF-1α, whereas HIF-3α misses the C-TAD domain, which is required for HIF-1α and HIF-2α transcriptional activity. In fact, the C-TAD was found to interact with CREB-binding protein (CBP) and p300 co-activators to activate gene transcription [6]. As shown in Fig. 1, α subunits are distinct from HIF-1β since they all possess the ODD domain, which is crucial for preventing their degradation in hypoxic condition [7].
Schematic illustration of HIF-1α, HIF-2α, HIF-3α isoforms and HIF-1β protein. As shown, HIF-α proteins possess a bHLH and two PAS domains, responsible for the heterodimerization. Instead of having the C-TAD domain, HIF-3α has a LZIP domain that allows the protein interaction. The constitutively expressed HIF-1β does not contain the ODD, N-TAD and ID domains. Abbreviations are listed in the legend section. Created in BioRender (https://BioRender.com/s38c417)
The existence of three different α subunits evokes questions about target genes exclusively induced by each of them, as well as the specificity of their regulated phenotypes. Unlike HIF-1α and HIF-2α, that are widely studied and clearly linked to development and carcinogenesis, HIF-3α functions are still largely unknown, especially in the context of malignancies [8,9,10,11]. The presence of multiple transcript variants of HIF-3α, showing tissue-specific expression and potential divergent roles in the regulation of gene expression, makes harder to determine its molecular functions [12]. Differently, HIF-1α and HIF-2α are well characterized, with a list of both common and unique target genes, which reflect their similar as well as independent roles in different tumor types (for review see [13]). Importantly, N-TAD domain, that differs between the two isoforms, has been shown to contribute to HIF-1α and HIF-2α target genes specificity, whereas the C-TAD domain is homologous and promotes the expression of HIF-1/HIF-2 common target genes [14].
Knockout mice have showed that both HIF-1α and HIF-2α are not-redundant and essential in development, since the targeted independent disruption of both subunits resulted in embryonic lethality. Nevertheless, embryos died of different causes and at different age. For instance, HIF-1α -/- embryos did not survive beyond cardiac and vascular defects, whereas HIF-2α -/- had normal systemic vasculature but suffered of bradycardia, and did not exhibit completed lung maturation [15]. Accordingly, HIF-1α and HIF-2α pattern of expression vary among tissues. Specifically, HIF-1α is detected ubiquitously, whereas HIF-2α exhibits a more tissue-specific expression pattern [16]. In response to reduced oxygenation, HIF-1α primarily mediates acute responses, whereas the chronic hypoxic response involves HIF-2α activation [17]. Although both subunits have been delineated as key factors in promoting tumor progression, however, the specificity of their functions remains to be fully elucidated.
Epidemiological characteristics and genetic alterations in cutaneous melanoma
Cutaneous melanoma (CM) represents a very aggressive neoplasm originating from the malignant transformation of melanocytes, accounting for about 20% of all cutaneous cancers [18]. Although it is the most lethal form of skin cancer, the 5-year relative survival is above 80%, with worse prognosis for nodular CM compared to lentigo maligna or superficial spreading CM [19]. CM incidence has dramatically increased in recent decades among light-skinned populations. Approximately 85% of CM occurring annually affect populations from North America, Europe and Oceania, with Australia and New Zealand showing the highest CM age-standardized incidence rates (40–50 cases/100000 per year) [18]. In Europe, the average incidence of CM is 9 cases/100,000 per year, with higher prevalence in Nordic populations (about 20 cases/100000 per year) [18].
Genetic alterations are common in both familial and sporadic CM, with frequently alteration of the MAPK/ERK signaling. In particular, the constitutive activation of the MAPK/ERK signaling caused by mutations within BRAF gene appears as a major driver of CM tumorigenic potential and survival [20, 21]. BRAF is a serine/threonine protein kinase encoded by the BRAF gene located on chromosome 7. At present, it is well known that over 90% of BRAF mutations occur in exon 15 and include substitutions of valine at position 600 (V600) with another aminoacid. In particular, the substitution of glutamic acid for valine (V600E, codon GTG > GAG), accounts for 70–80% of the BRAFV600 mutations. The second most common mutation is BRAFV600K substituting lysine for valine (GTG > AAG), followed by BRAFV600R (GTG > AGG), and by an infrequent two-nucleotide variation of the predominant mutation that includes the BRAFV600 ′E2′ (GTG > GAA) and the BRAFV600D (GTG > GAT) [22]. BRAF mutations are able to produce an activation of the kinase domain of the protein, leading to an uncontrolled stimulation of cell proliferation [23]. NRAS is the second most frequent mutation in CM (15–20%), after BRAF [24]. In the majority of cases (> 80%), NRAS gene presents a missense mutation within codon 61, which disrupts the GTPase activity of NRAS, locking it in its active conformation, independent of upstream RTK activation [25]. Besides BRAF and NRAS, a small subset of CM presents NF1 (10–15%) and c-Kit (1–3%) mutations [25]. Finally, a high frequency of PTEN mutations have been reported in highly metastatic CM, where they frequently coexist with BRAF mutations, but not with NRAS [25].
Constitutive expression of HIF-1α and HIF-2α has been frequently detected in CM [26]. BRAF-mutant CM, in particular, usually exhibits dramatic activation of HIF-1α signaling, often uncoupled from hypoxia [27,28,29]. Along this line, microarray profiling on 30 cell lines derived from various stages of CM progression and 5 melanocytes showed that HIF-1α expression was significantly higher in BRAF-mutant CM cell lines respect to cells with wild-type BRAF or melanocytes. Accordingly, suppression of BRAFV600E decreased HIF-1α expression and impaired CM cell survival and proliferation, whereas its overexpression increased hypoxic tolerance through HIF-1α up-regulation, suggesting that the oncogenic activity of BRAFV600E might be partially mediated directly through the HIF-α signaling pathway [27].
In the last years, a better understanding of the CM biology has resulted in the development of different FDA approved therapeutic strategies relying on the use of small molecule inhibitors of BRAF (BRAFi) and MEK (MEKi) and blockers of the immune checkpoint molecules. The advent of targeted and immune-based therapies has not only expanded the treatment options for CM but also proven clinical benefits in patients with unresectable or metastatic CM. However, primary and acquired resistance often limit the clinical effectiveness of these therapies. Importantly, aberrant expression of HIF-1α and HIF-2α might influence response to therapy and impact on drug resistance in CM.
Hence, this review aims to provide an overview of the activity and the regulation of HIF-1α and HIF-2α in CM, not only under hypoxia, which occurs when oxygen (O2) levels fall below 1 kPa, but also in normoxic environments. With the term “normoxia”, we refer to the atmospheric oxygen (O2) levels of 18 kPa at which the majority of pre-clinical studies with cells are conducted. This O2 tension is clearly higher respect to that measured in physiological tissues (i.e. 3–8 kPa in the skin), and termed “physoxia” [30, 31]. Based on these considerations, a list of papers with the terms “CM”, “hypoxia”, “normoxia” and/or “HIF” were retrieved and selected, mainly focusing on those published in the last two decades. The potential therapeutic targeting of HIFs to improve treatment efficacy in CM will be also discussed.
HIF-1α-related targets in hypoxia and normoxia in CM
In CM, HIF-1α mediates the transcription of genes involved in several processes, including angiogenesis and vascular remodeling, epithelial to mesenchymal transition (EMT), metastasis, and cellular metabolism, in both hypoxic and normoxic conditions. A selection of the most frequently HIF-1α-target genes in CM is given in Table 1.
Angiogenesis and vasculogenic mimicry. It has become increasingly clear that the interaction of endothelial cells (EC) and cancer cells affects tumor growth and vascularization. Hypoxia and HIF-α signature are master regulators of microcirculation acquisition, thus supporting malignant growth and hematogenous dissemination. The delineation of molecular mechanisms of neo-angiogenesis in cancer has revealed a critical role for HIF-1α in positively regulating pro-angiogenic factors, including vascular endothelial growth factor (VEGF) and its receptors (i.e. FLK-1 and FTL-1), angiopoietins and platelet-derived growth factor beta [32]. To date, although the hypoxia-induced new vessel formation is a common feature of CM, the number of studies directly focused on HIF-1α and angiogenesis in CM progression and treatment are still limited. However, it has become clear that HIF-1α fosters pro-angiogenic factors expression in CM [33]. Indeed, Trisciuoglio et al. reported that Bcl-2, via BH4 domain, increased HIF-1α half-life by counteracting its ubiquitination, in order to enhance HIF-1α-mediated VEGF expression and secretion in CM cells exposed to hypoxia [34]. Of interest, vasculogenic mimicry, a process that cooperates with neo-angiogenesis for the recruitment of new vessels, was firstly identified in CM and uveal melanoma through in vitro and in vivo studies [35, 36]. In particular, vasculogenic mimicry describes the ability of CM cells to express endothelium-associated genes and ECM-remodeling proteins to mimic the presence and function of EC and to support a capillary-like structure formation [35, 36]. Mouse CM cells injected in mouse ischemic limbs increased the expression of HIF-1α and VEGF to support the formation of vasculogenic mimicry channels [37]. In a study by Comito et al., once stabilized upon hypoxia-induced accumulation of mitochondrial reactive oxygen species (ROS), HIF-1α activated MET expression in CM cells to promote capillary-like structures formation and vasculogenic mimicry [38]. In a subsequent study by Spinella et al., hypoxia induced production of endothelin-1 (ET-1) that, in turn, sustained HIF-1α/HIF-2α-mediated VEGF-A and VEGF-C expression in CM cells and EC, suggesting HIFs involvement in promoting endothelia remodeling and vasculogenic mimicry. Consistent with this hypothesis, silencing of HIF-1α/HIF-2α completely abrogated cell migration and hypoxia-induced capillary-like structure acquisition, in both CM and EC exposed to CM cells secretome. Furthermore, ET-1 capacity to increase VEGF-A and VEGF-C mRNA and protein expression in CM and EC was reduced [39].
EMT and metastasis. The expression of HIF transcription factors has been suggested to correlate with the increased malignant potential of melanocytes. In fact, several genes that act as key players in melanogenesis and melanocyte behavior contain putative functional hypoxia response elements (HREs) and, therefore, might be directly targeted by HIF transcription factors [26]. In another study, transcriptomic analysis of murine immortalized melanocyte cells exposed to hypoxia identified a signature that included thirteen EMT-associated genes, of which only two, Plod1 and Plod2, exhibited HRE motifs within their promoters [40], supporting that HIF-1α regulation might occur through both direct and indirect mechanisms.
In hypoxic conditions, HIF-1α has been demonstrated to contribute to CM heterogeneity, and to trigger metastatic progression by reprogramming gene expression patterns in proliferative CM cells, making them more invasive in in vitro assays. The incubation of a set of human CM cells in hypoxia chamber allowed to identify eighteen HIF1α-dependent genes that were differentially regulated in the switch from a proliferative to an invasive phenotype in CM cell cultures. Interestingly, the microphthalmia-associated transcription factor (MITF) and several of its target (i.e. TRP1, TNFRSF14, CAPN3, GPM6B, and DAPK1) were among repressed genes [41], indicating that HIF1α-regulated MITF might act as a key regulator of the process of hypoxia-induced phenotype switching in CM. In accordance, a study by Cheli et al. revealed that, in hypoxic condition, HIF-1α positively regulated the expression of the transcription factor basic helix-loop-helix protein 2 (BHLHB2) to inhibit MITF, thus increasing the tumorigenic and metastatic properties of CM cells [42]. The interplay between HIF-1α and the receptor tyrosine kinase-like orphan receptor (ROR)-1 and ROR-2 has been intimately involved in the switch from a proliferative to an invasive phenotype as well. More specifically, following hypoxia exposure, Wnt5A stabilized HIF-1α, resulting in ROR-2 up-regulation and decreased expression of MITF and ROR-1. ROR2 expression associated with a more aggressive phenotype and was critical for Wnt5A-mediated invasion and metastasis in CM [43].
HIF-1α was also shown to orchestrate EMT and metastasis in CM by increasing the expression of proteins and enzymes directly implicated in extracellular-matrix (ECM) remodeling, including matrix metalloproteinase-2 (MMP-2) and urokinase-type plasminogen activator receptor (uPAR). Once activated by hypoxia, HIF-1α enhanced the invasion and metastatic potential of CM cells by fostering MMP-2 expression through the involvement of CD147 [44], a key inducer of MMPs [45]. Chromosome immunoprecipitation assay supported that HIF-1α directly bound to CD147 promoter. Indeed, point mutations within CD147 promoter attenuated MMP-2 response to hypoxia both in vitro and in vivo [44]. The interaction between urokinase-type plasminogen activator receptor (uPAR) and its ligand, which is mainly involved in pericellular proteolysis, is a central step in mesenchymal motility. By binding to the HRE within uPAR promoter, HIF-1α mediated uPAR transcription in various cancers under hypoxia [46, 47]. In CM, in particular, uPAR overexpression promoted spontaneous lymph nodes metastasis in in vivo models [48]. In addition to MMP-2 and uPAR, hypoxia might induce CM metastasis through a putative cooperation between HIF-1α and thymosin beta-4 (Tβ4) [49], an actin-sequestering molecule involved in cytoskeletal reorganization [50]. At present, the molecular mechanism by which the HIF-1α/Tβ4 axis contributes to CM metastasis is still largely unknown. Nevertheless, in a study by Makowiecka et al., Tβ4 was not only a component of focal adhesions, but also directly regulated their formation, thus altering the adhesion abilities of CM cells [51].
As described above, the ROS/HIF-1α/MET signaling axis has been strongly involved in hypoxia mediated vasculogenic mimicry in CM. In the same study, MET expression and activation were also correlated with enhanced CM cells migration in vitro and lung metastasis formation in vivo [38], thereby suggesting that HIF-1α and MET might cooperate in eliciting an escape strategy to avoid the hypoxic CM environment.
More recently, Xu et al. unveiled baculoviral IAP repeat-containing protein 7 (BIRC7) as a downstream HIF-1α target gene. Although BIRC7 is known to act as an apoptosis inhibitor, its knockdown inhibited the invasion of A875 and M14 CM cells in both hypoxia and normoxia [52], indicating that even in normoxic condition HIF-1α could sustain invasiveness. Under lactate stimulation, HIF-1α was found to drive lung metastasis in B16F10 transplanted mice, specifically by regulating Rab27 expression and extracellular vesicles secretion [53].
Metabolic reprogramming. HIF-1α represents the main transcription factor for the induction of genes encoding glucose transporters (GLUT 1/4) and glycolytic enzymes, such as aldolase, enolase, lactate dehydrogenase (LDHA) and phosphoglycerate kinase-1, allowing hypoxic tumors cells to take up glucose more efficiently. Additionally, HIF-1α can suppress oxidative phosphorylation (OXPHOS) by reducing metabolites entry into the tricarboxylic acid cycle (TCA), via the direct induction of the key glycolytic enzyme pyruvate dehydrogenase kinase (PDK) 1 that, in turn, phosphorylates the pyruvate dehydrogenase (PDH) complex, thus preventing the conversion of pyruvate into acetyl-CoA [54]. Respect to melanocytes, malignant CM cells, particularly those harboring the BRAFV600E mutation, exhibit the Warburg effect with high glucose consumption and more lactate production even in the presence of oxygen and fully functioning mitochondria [55]. The BRAFV600E mutation might transactivate a panel of transcriptional regulators of glycolysis, including HIF-1α [28] and its target genes GLUT1, hexokinase 2 (HK2), LDHA [56], and PDK1 [29]. Under hypoxia, HIF-1α-dependent upregulation was accompanied by a decreased expression of several components of the OXPHOS, including ATP5ME, NDUFA6, NDUFB5, NDUFB6 and NDUFS8 [57]. HIF-1α activation could also up-regulate pyruvate kinase M2 (PKM2), a rate-limiting enzyme of glycolysis, further minimizing OXPHOS [58].
HIF-1α-dependent glycolytic program offers metabolic plasticity but also growth advantage for CM cells. For example, Liu et al. demonstrated that HIF-1α enhanced PDK1 transcription by recruiting the Ku80 protein to PDK1 promoter. Consistently, treatment with melatonin, which affects the stability of HIF-1α at the protein level, suppressed the binding of Ku80 to the PDK1 promoter and inhibited CM growth [59]. By reprogramming the glucose metabolic pathway and ensuring scavenging of mitochondrial free radicals, HIF-1α enhances the antioxidant capacity of tumor cells, fostering radioresistance [60]. In this context, it has been demonstrated that HIF-1α could indirectly decrease the expression of the ganglioside GM3 synthase, thus reducing synthesis of glycosphingolipids and increasing resistance of CM cells to oxidative stress and radiation therapy [61].
Inflammation. In silico analysis suggested that inflammatory response, together with increased recruitment of T-cells, B-cells, NK-cells, monocytes and macrophages, were strongly associated with CM [62]. Intriguingly, HIF-1α has emerged as one the main pathways implicated in the regulation of the inflammatory response. Atypical chemokine receptor 2 (ACKR2), which regulates a number of pro-inflammatory chemokines reported to drive cytotoxic cells in tumor microenvironment (TME), has been recently identified as a direct transcriptional target of HIF-1α. In a study by Benoit et al., the decreased ACKR2 expression observed following HIF-1α depletion was associated with an augment of Chemokine (C-C motif) ligand 5 levels in hypoxic B16F10 CM cells, providing novel evidence on how hypoxia might impair pro-inflammatory TME and hamper immune infiltration [63].
HIF-2α-related phenotypes in hypoxia and normoxia in CM
While HIF-2α has been largely characterized in renal cell carcinoma, where it has been shown to represent a key driver of this disease, studies focused on its function in other human malignancies, including CM, are still in their infancy. At present, however, it is known that, similarly to HIF-1α, HIF-2α can regulate several pathways, thus contributing to the malignant behaviors of CM cells (Table 1).
Angiogenesis and vasculogenic mimicry. Unlike HIF-1α, the role of HIF-2α in regulating angiogenesis is still ambiguous and not complete clarified. As stated above, ET-1 production sustained HIF-1α/HIF-2α-mediated VEGF-A and VEGF-C expression in CM and EC, thus promoting neo-vascularization [39]. Surprisingly, and in contrast with the study by Spinella et al., a putative tumor-suppressor role for HIF-2α has been also described. In fact, stabilization of HIF-2α with the proline hydroxylases (PHD) 3 inhibitor AKB-6899 decreased angiogenesis in B16F10 CM-bearing mice and in A375 CM cell-line derived xenograft, by promoting secretion in the tumor environment of a soluble form of the VEGF receptor (VEGFR), which usually inhibits VEGF activity. On the contrary, HIF-1α accumulation and VEGF production were not affected by AKB-6899 treatment [64].
EMT, metastasis and stemness. In the last years, a number of evidence has confirmed that aberrant HIF-2α activation contributes to CM metastasis. Under hypoxia, HIF-2α promotes EMT and cell migration by enhancing the expression of specific target genes, which can be, or not, commonly regulated by HIF-1α. For instance, HIF-2α, but not HIF-1α, directly enhanced Snail expression in CM cells, fostering features associated with stemness in vitro and increasing metastatic capacity in vivo [65]. A mass spectrometry analysis of HIF-2α interactome identified specific HIF-2α interactors involved in CM development, including MITF, SRY-box transcription factor (SOX) 10, and adaptor protein-2α (AP2α). Although MITF and SOX10 were confirmed as HIF-1α partners as well, the transcription factor AP2α was not. Furthermore, HIF-2α and AP2α interaction had functional consequences for the invasive properties of CM cells since HIF-2α overexpression in AP2α-expressing cells could reduce their invasion potential [66]. On the other hand, an independent silencing of HIF-1α and HIF-2α performed in metastatic CM cells K457 by using short hairpin RNA (shRNA) identified connective tissue growth factor (CTGF), an invasion positive MMPs regulator, as a specific target of both isoforms. Inhibition of CTGF decreased invasion and migration of CM cells along with reduced MMP-9 expression [67]. shRNA constructs against HIF1α and HIF2α did not affect tumor initiation and progression in PTEN-deficient, BRAF-mutant genetically engineered mouse model of CM, but clearly abrogated metastasis at lymph node. Mechanistically, HIF-1α and HIF-2α drove CM invasion through Platelet Derived Growth Factor Receptor Alpha (PDGFRα) and Focal Adhesion Kinase (FAK)-dependent SRC activation, and by directly regulating the expression of MMPs involved in invadopodia-associated ECM degradation. Notably, HIF-1α and HIF-2α acted independently since HIF-1α positively regulated PDGFRα-dependent SRC activation and Metallothionein 1-MMP expression, whereas HIF-2α specifically fostered FAK signaling and MMP-2 expression in CM cells cultured in hypoxic conditions [68].
Immunomodulation. Regulatory T (Treg) cells usually prevent effective anti-tumor immune response and promote immune evasion, thus playing a crucial role in CM initiation and progression [69]). However, some Treg cells can be converted into effector T cells, which lose suppressive capacity and release pro-inflammatory cytokines to promote inflammation [70]. In this context, Treg-selective HIF-2α deletion or treatment with the HIF-2α PT2385 inhibitor reduced Treg capacity to impair the activities of effector T cells, and suppressed cells dissemination in immunogenic B16F10 CM mouse models, apparently via increasing HIF-1α expression [70]. In contrast, in a study by Doedens et al., HIF-2α, along with HIF-1α maintains the functions of CD8+ cytotoxic T lymphocytes (CTL) [71], which are the front-line defense in the elimination of infected or tumor cells [72]. In fact, in CTL, HIF-1α and HIF-2α sustained the expression of effector molecules, co-stimulatory receptors and key transcriptional regulators, thus hampering tumor growth in CM mouse models [71].
HIF-α regulation in CM
Canonical-oxygen dependent negative regulation of HIF-α signaling is commonly driven by the PHD 1–3 proteins. PHDs hydroxylate two conserved proline residues in the ODD motif of HIF-α subunits, in order to promote the binding of HIF-α with the von Hippel-Lindau protein (VHL) that leads to its subsequent proteasome-dependent degradation [2]. Surprisingly, Liu et al. found that psychological stress enhanced EMT and vasculogenic mimicry in CM by reducing the ubiquitin-mediated degradation of HIF-1α through the activity of the D2 dopamine receptor (DRD2), which expression was upregulated in tumor tissues under stress condition. More precisely, DRD2 could interact with VHL and competitively inhibit the HIF-1α/VHL interaction, resulting in increased HIF-1α stability [73]. In addition, the transcriptional activity of both HIF-1α and HIF-2α is also negatively regulated through the hydroxylation of an asparagine residue in the C-TAD domain, mediated by the factor-inhibiting HIF, that blocks the HIF-α binding to CBP/p300 co-transcription factors [74]. These processes are hampered under hypoxic conditions, since HIF-α subunits accumulate and heterodimerize with HIF-1β to form a transcriptional complex that translocates into the nucleus [75].
In addition to the canonical regulation, supplementary factors can influence HIF-1α transcription, protein stability and activity in response to hypoxia in CM (Fig. 2). Among them, poly (ADP-ribose) polymerase 1 (PARP-1) has recently emerged as a novel HIF-1α regulator in CM. Mechanistically, following incubation in a hypoxia chamber, Martí JM et al. observed an early ROS induction that led to PARP-1 activation in C8161 metastatic CM cells. Once activated, PARP-1 stabilized HIF-1α by PARylation at specific lysine and arginine residues within the C-terminus domain. Consistently, PARP-1 and HIF-1α expression were strongly associated in tissues from CM patients, thus suggesting that PARP inhibitors might be considered as a potential therapeutic approach in CM displaying HIF-1α overactivation [76]. Intriguingly, Lakhter et al. unveiled that HIF-1α could localize at Golgi compartment in CM cells cultured in hypoxia chamber. Treatment with brefeldin A, an ER-Golgi transport inhibitor, not only induced HIF-1α protein accumulation in the nucleus, but also reduced its transcriptional activity, indicating that ER-Golgi transport is essential to fine-tuning the transcriptional activity of HIF-1α [77].
Overview of the multiple pathways impacting on HIF-1α in CM. In the upper panel, a graphic representation of the main pathways regulated by HIF-1α under hypoxic conditions (1 kPa O2 levels) in CM is shown. Conversely, the lower panel resumes pathways that are controlled by HIF-1α in normoxia (18 kPa O2 levels). Panels are divided into pro-tumorigenic (blue) and anti-tumorigenic (orange). Created in BioRender (https://BioRender.com/p73u915). Abbreviations: HIF: Hypoxia-inducible factor; ROS: Reactive oxygen species; PARP1: Poly (ADP-ribose) polymerase; PAR: Poly ADP-ribose; HDAC8: Histone deacetylase 8; IL-18R: interleukin-18 receptor; RAC1: Rac Family Small GTPase 1; NF-kB: The Nuclear Factor Kappa B; VEGF: Vascular endothelial growth factor; ER: endoplasmic reticulum; NLGN4X: Neuroligin; VBP1: VHL Binding Protein 1; VHL: Von Hippel-Lindau; DRD2: D2 dopamine receptor; GPR81: G protein-coupled receptor 81; PKA: protein kinase A; TEV: tumour-derived extracellular vesicles; SOX2: SRY-box transcription factor; TCA: Tricarboxylic acid; GPX3: Glutathione Peroxidase 3; NOX5: NADPH oxidase 5; EMT: Epithelial to mesenchymal transition; BNIP3: BCL-2 interacting protein 3; NCOA4: Nuclear Receptor Coactivator 4; MITF: Microphthalmia-associated transcription factor; ANXA3: Annexin A3; PDK1: Pyruvate dehydrogenase kinase; UPR: unfolded protein response
In cancer, hypoxia is only one factor among several HIF-α-activating mechanisms. Interestingly, normoxic activation of HIF-1α has been detected in several cancer cell types, including CM, where contributes to the acquisition and maintenance of a malignant phenotype [78, 79] (Fig. 2). Accordingly, Martí-Díaz et al. observed that, in normoxic conditions, the inhibition of HIF-1α reduced glucose viability, impaired glucose metabolism, and suppressed CM cell survival [80]. In line with these findings, in a study by Andreucci et al., the SOX2-driven HIF-1α inhibition decreased the glycolytic activity and induced a shift towards oxidative phosphorylation in CM cells in normoxia [81].
Different non-hypoxic stimuli could enhance HIF-1α signaling, including lipopolysaccharides [81], thrombin, angiotensin I and cytokines [82]. As reported above, a study by Luo et al. showed an interplay between lactate stimulation and HIF-1α-driven CM metastasis formation in vivo. In detail, lactate could bind to G protein-coupled receptor 81 (GPR81) to decrease cAMP levels and the subsequent protein kinase A (PKA) activation, finally suppressing the ubiquitin-mediated proteolysis of HIF-1α [53].
At present, several proteins have been listed as HIF-1α non-hypoxic activators in CM. In a recent paper by Schörghofer et al., decreased amount of the adhesion molecule Neuroligin (NLGN4X) has been correlated with increased HIF-1α accumulation and migratory phenotypes in metastatic CM. The same group further demonstrated that HIF-1α regulation by NLGN4X was primarily mediated by Von Hippel-Lindau Binding Protein 1, a member of the canonical prefoldin complex [83]. In hypoxia, MITF expression was found to be suppressed following HIF-1α activation in CM [41,42,43]. However, in response to cAMP, MITF bound to the HIF-1α promoter to stimulate its transcriptional activity in normoxic melanocytes and CM cells [84]. Mitochondrial ROS have been involved in HIF-1α regulation in hypoxia in CM [38]. Besides mitochondria, ROS can be produced in the cytosol as well, and their accumulation can also occur in non-hypoxic conditions. For instance, cytosolic NADPH oxidase 5 (NOX5) contributed to sustain CM cell proliferation, at least in part, by generating high local concentration of ROS that in turn, regulated normoxic HIF-1α expression [85]. ROS are known to activate NF-κB [86], and NF-κB activation might enhance HIF-1α transcription under hypoxic and normoxic environments [87, 88], suggesting a potential crosstalk among ROS, NF-κB and HIF-1α. Consistent with this hypothesis, NF-κB activation triggered by ROS increased the normoxic HIF-1α activity, whereas NF-κB inhibition attenuated accumulation in CM cell lines [79]. In another study by Kim J et al., interleukin (IL)-18 signaling induced HIF-1α expression through the Rac1/NF-κB pathway in the mouse CM B16F10 cell line. Of interest, following treatment with the antioxidants N-acetyl-l-cysteine and pyrrolidine dithiocarbamate (PDTC), IL-18 was not able to up-regulate HIF-1α expression. Furthermore, PDTC alone, or in combination with an inactive form of Rac1, markedly reduced NF-κB activation, suggesting that once activated by IL-8 signaling, Rac1 promoted ROS generation to mediate NF-κB activation and HIF-1α expression. Even though the molecular mechanism has been investigated under normoxic conditions, hypoxic stimulation dramatically increased IL-18 and HIF-1α mRNA expression, indicating that IL-18 might perform regulatory functions in HIF-1α expression under both normoxic and hypoxic conditions [89]. Although the molecular mechanism is yet to be defined, the calcium-dependent phospholipid-binding protein annexin A3 (ANXA3) has been implicated in HIF-1α aberrant activation in CM as well. In fact, in a study by Xu et al., ANXA3 overexpression increased HIF-1α and VEGF protein expression levels [90]. In CM cells, under conditions of oxygen availability, BCL-2 interacting protein 3 (BNIP3) could act as the upstream regulator of the HIF-1α-mediated glycolytic program. More specifically, by controlling the intracellular availability of iron and the mediated autophagic degradation of ferritin, BNIP3 was found to regulate HIF-1α stability [91]. Since BNIP3 represents one of the main HIF-1α target genes, results from this study indicated that this bidirectional loop between BNIP3 and HIF-1α fostered normoxic HIF-1α-linked glycolysis and pro-tumorigenic program in CM. Finally, the histone deacetylase 8 (HDAC8) directly bound to HIF-1α to sustain the stabilization of the protein in both hypoxia and normoxia in CM [92]. On the other hand, the glutathione peroxidase 3, an enzyme involved in protecting cells from oxidative stress, and often downregulated in CM, was reported to reduce HIF-1α and HIF-2α stability and activity by inactivating ROS production [93].
Non-coding RNAs associated to HIF-α signaling
Functional RNAs that are not translated into proteins are called non-coding RNA (ncRNA) (for review see [94]). In the last years, a growing number of studies have revealed their importance as key molecular mediators in several many critical aspects of tumor biology, including hypoxia and HIF-α signaling [95, 96].
MicroRNAs involved in HIF-α regulation in CM
MicroRNAs (miRNAs) are small single-stranded ncRNAs whose length ranges between 18 and 25 nucleotides. MiRNAs are transcribed as pri-miRNAs by RNA polymerase II. Pri-miRNAs are then processed by the complex formed by DROSHA/DGCR8 to produce miRNA precursors (pre-miRNAs). Thereafter, pre-miRNAs are transported to the cytoplasm where they are processed by the RNase III enzyme DICER/TRBP2 to form the mature miRNA duplex. This miRNA duplex includes the two-strand pair miR-3p/miR-5p, each of which may be selected by the ARGONAUTE proteins associated with the RNA-induced silencing complex. The final single-stranded miRNA can regulate gene expression at the post-transcriptional level through fine-tuning of target protein expression. It has become increasingly clear that miRNAs deregulation is a hallmark of cancer, where they might function as oncogenes to promote tumor formation (oncomiRs), or act as tumor suppressors. Not surprisingly, miRNAs are involved in cancer development and progression, including the metastatic process and the TME remodeling (for review see [97]).
Regulation of HIF-1α on miRNAs is common during cancer initiation and progression (for review see [98]). However, recent evidence has documented complex interactions between HIF-1α and miRNAs since, besides to be directly or indirectly regulated by HIF-1α, miRNAs can modulate the expression or activity of HIF-1α as well (Fig. 3) [96, 99, 100].
Schematic representation of non-coding RNAs involved in HIF-1α pathway regulation in CM. In the upper semicircle, a summary of the current advances about the bidirectional interactions between HIF-1α and miRNAs in CM (blue: suppressor miRNAs; orange: oncogenic miRNAs). The lower semicircle illustrates a graphic representation of the mechanism by which the lncRNA linc00518 modulates HIF-1α expression in CM. Created in BioRender (https://BioRender.com/833o824). Abbreviations: HIF: Hypoxia-inducible factor; miR: mirna; CM: cutaneous melanoma; ROS: Reactive oxygen species; MNT: MAX Network Transcriptional Repressor; CTL: cytotoxic T lymphocyte; TCA: Tricarboxylic acid; PDK4: Pyruvate Dehydrogenase Kinase 4; LDHA: Lactate dehydrogenase A; PTPN1:Protein Tyrosine Phosphatase Non-Receptor Type 1; HOXA1: Homeobox A1; TP53I11. Tumor Protein P53 Inducible Protein 11.LDHA: Lactate dehydrogenase A; PTPN1:Protein Tyrosine Phosphatase Non-Receptor Type 1; HOXA1: Homeobox A1; TP53I11. Tumor Protein P53 Inducible Protein 11
MiRNAs that negatively modulate HIF-1α. HIF-1α was identified as a target of miR-199a-5p in several tumors, including CM. In fact, miR-199a-5p mimics impaired CM cell proliferation by decreasing HIF-1α mRNA and protein expression both in vitro and in vivo. Consistently, low expression levels of miR-199a-5p were observed in CM, particularly in tissue samples from patients with advanced tumor stage [101]. Zhou et al. reported that miR-33a expression in metastatic CM cells was also relatively low. Interestingly, when overexpressed, miR-33a inhibited HIF-1α and, consequently, reduced BRAF-mutant CM cell proliferation and invasion in vitro, and tumor growth in vivo [102]. Subsequently, Cao et al. confirmed that miR-33a-5p expression was decreased in CM cells, and that miR-33a-5p negatively regulated HIF-1α, thereby repressing glycolysis and increasing radiosensitivity of CM cells [103]. In addition to miR-199a-5p and miR-33a, Mazar et al. found that miR-211-5p indirectly reduced HIF-1α protein levels and decreased CM cell growth in hypoxia. In detail, when overexpressed in A375 BRAF-mutant CM cells, miR-211-5p improved mitochondrial respiration by targeting PDK4. On the contrary, miR-211 down-regulation led to increased PDK4 protein expression, which finally resulted in less oxidative phosphorylation, reduced mitochondrial activity and HIF-1α stabilization. Accordingly, miR-211-5p was found to be downregulated during CM progression [104]. Intriguingly, although miR-211-5p overexpression was more frequent in primary tumors respect to metastases, in a study by Moritz et al., high miR-211-5p expression in CM metastases, but not primary tumors, was associated with worse overall survival [105]. In line with these findings, miR-211-5p was upregulated in CM cells resistant to BRAFi, and involved in the early phases of resistance acquisition to targeted therapy through HIF-1α-independent mechanisms [106, 107].
HIF-associated oncogenic miRNAs. Among miRNAs that positively support HIF-α signaling, miR-675-3p was significantly up-regulated in CM cell lines, tissues and peripheral blood from CM patients. In vitro analyses indicated that miR-675-3p might function as a mediator of many cellular pathways in CM, since the use of a miR-675 mimic could activate TGFβ and HIF-1α signaling pathways [108]. In a subsequent study by Hwang et al., high levels of HIF-1α transcriptional activity were detected in a panel of TGFβ1 positive CM cell lines under normoxic conditions, and HIF-1α silencing downregulated several miRNAs, including miR-210, miR-218, miR-224 and miR-452 [109]. MiR-210 is a direct target of both HIF-1α and HIF-2α, and its expression has been correlated with metastasis, poor prognosis and early risk of recurrence in CM [110, 111]. HIF-1α-dependent miR-210 upregulation impaired the ability of immune system to target CM cells. Indeed, miR-210 overexpression promoted immune escape from CTL-mediated lysis by degrading protein tyrosine phosphatase non-receptor type 1, homeobox A1, and tumor protein p53 inducible protein 11 [112]. Besides to impact on immune response, miR-210 might also influence mitochondrial activity in CM. In fact, miR-210 silencing accelerated mitochondrial respiratory, thus improving ROS elimination and labelling CM cells for self-destruction, in both normoxic and hypoxic conditions [113, 114]. Therefore, miR-210 might act as a key factor in HIF-1α-mediated tumorigenesis, thus representing a good prognostic and therapeutic target in both normoxic and hypoxic CM.
At present, differently from HIF-1α, only a study by Hao et al. has focused on HIF-2α-associated miRNA in CM. More specifically, HIF-2α protein was found to mediate stemness of CM cells by regulating the miRNA-363-3p/p21 axis [115]. Nevertheless, the deeper correlation between HIF-2α and miRNA-363-3p remains to be further explored.
Long non-coding RNAs involved in HIF-α regulation in CM
Long non-coding RNAs (lncRNAs) are defined as ncRNAs of more than 200 nucleotides with little or no coding information. LncRNAs can be located either in the nucleus or the cytoplasm, or in both compartments simultaneously, and can be classified into five categories: sense, antisense, bidirectional, intronic, and intergenic. LncRNAs have been involved in a wide range of cellular mechanisms in several tumors, including CM (for review see [94]).
To our knowledge, LINC00518 is the only lncRNA that has been characterized for its role in regulating HIF-1α expression in CM [116]. More precisely, LINC00518 sponged miR-33a-3p to increase the expression of HIF-1α, thus accelerating glycolytic metabolism and triggering radioresistance in BRAF-mutant CM cells. Additionally, HIF-1α recruited HDAC2 to the miR-33a-3p promoter region to decrease its expression. Hence, the LINC00518/miR-33a-3p/HIF-1α axis might represent an efficient target to increase the therapeutic efficiency of radiotherapy in CM [116].
Therapeutic opportunity in CM
As mentioned above, HIF-α subunits not only play a crucial role in the response of CM cells to hypoxia but also regulate different biological processes under normoxic conditions, thus representing attractive therapeutic molecular targets. In the last years, several anticancer drugs that modulate HIF-1α expression and/or activity without directly targeting it have been described.
HIF-1α significantly impacts on CM cell metabolism as it influences the expression of different genes involved in glycolysis. Among the key effectors of HIF-1α-mediated metabolic remodeling, the 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) enzyme plays a crucial role in glycolysis since, once activated, it stimulates allosterically the activity of PFK-1 in BRAF-mutant CM, fostering the Warburg’s effect. Intriguingly, (2E)-3-(3-Pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO), an inhibitor of PFKFB3, was shown to interfere with the HIF-1α/PFKFB3/PFK-1 axis, and to induce cell cycle arrest in G1/0, apoptosis and glucose uptake reduction in BRAF-mutant A375 CM cells [117].
In a study by Furfaro et al., MeOV-1 CM cells cultured under 18 kPa O2 (normoxia), 5 kPa O2 (physioxia), and 1 kPa O2 (hypoxia) were treated with BRAFi. Of interest, HIF-1α expression was significantly decreased by the treatment under all O2 tensions tested [118]. Consistently, BRAFi vemurafenib (PLX4032) suppressed HIF-1α expression at the mRNA and protein levels in a panel of BRAFV600 CM cell lines cultured under standard conditions. More interestingly, HIF-1α expression was also clearly decreased early on treatment with BRAFi and restored after progression in a subset of CM biopsies. Similarly, the expression of several glycolysis-associated genes (i.e. HK2, SLC2A1 and SLC2A3) was significantly decreased following BRAFi treatment and restored in CM patients that developed drug resistance [28], suggesting a possible involvement of HIF-1α in resistance to BRAF-targeted therapies. Under hypoxia, HIF-1α levels were decreased in CM cells exposed to PLX4032 in a cell type-dependent manner [82, 119], further suggesting that, besides to favor tumor heterogeneity, HIF-1α might influence the response to the drug treatment. Accordingly, Qui et al. demonstrated that hypoxia, by inducing MET and HGF via HIF-1α, prompted the acquisition of vemurafenib resistance in CM BRAF-mutant models both in vitro and in vivo [120]. Blocking MET signalling potentiated the antitumor effect of vemurafenib, indicating that MET inhibition might represent a potential therapeutic strategy for overcoming HIF-1α-driven PLX4032 resistance in CM patients.
In NRAS-driven CM, the growth factor receptor-binding protein 2-associated protein 2 (GAB2) has been reported to enhance tumor formation and angiogenesis by stabilizing HIF-1α at the post-transcriptional level. Histological examination of GAB2wt/NRASG12D xenografts showed longer vessels with markedly dilated lumina, together with increased expression of CD31, VEGFR2, VEGF, respect to NRASG12D tumors. Pharmacological inhibition of MEK significantly suppressed angiogenesis, providing evidence that HIF-1α mediated angiogenic response was dependent on RAS-RAF-MEK-ERK signaling in GAB2/NRAS-driven tumorigenesis [121].
At present, no study has evaluated the potential contribution of NF1 mutation in HIFs signalling activation in CM. Conversely, in c-Kit mutated melanocytes, HIF-1α overexpression was found to promote oncogenic transformation. Treatment with imatinib, a c-Kit inhibitor, reversed proliferation and anchorage-independent growth of melanocytes, confirming that HIF-1α induced melanocytes transformation in a c-Kit-dependent manner [122].
PTEN has been clearly identified as a potent negative regulator of angiogenesis via inhibition of HIF-1α signaling. In normoxic conditions, PTEN enhances HIF-1α ubiquitination and degradation, whereas PTEN inactivation increases the transcriptional activity of HIF-1α in hypoxia. Hence, to restore PTEN expression or activity might impair HIF-1-α-induced angiogenesis, with potential therapeutic benefit in PTEN-deficient CM. In this context, it has been proposed that the administration of plant-derived natural bioactive compounds with demethylating effects, such as phytochemicals, might favor PTEN upregulation [123].
So far, a broad spectrum of molecules of natural origin has proven to affect HIF-1α expression. Among them, isoliquiritigenin was effective in suppressing cell proliferation and in inducing apoptosis via reduced HIF-1α protein stability and mRNA expression of a number of glycolysis-related genes in B16F10 transplanted mice [124]. Vanillin was also reported to possess anti-cancer and anti-metastatic activities in CM. In particular, vanillin treatment significantly decreases HIF-1α mRNA expression by suppressing STAT3 transcriptional activity in CM cells [125]. A subsequent study by Li et al. indicated that luteolin, a natural flavonoid, exhibited a strong anti-cancer activity in CM by impairing angiogenesis and EMT both in vitro and in vivo. This effect was mainly achieved through the disruption of the HIF-1α/VEGF signaling pathway [126]. Finally, celastrol, a chemical compound extracted from the root extract of the Tripterygium Wilfordii, reduced the expression of HIF-1α mRNA by inhibiting the PI3K/Akt/mTOR signaling, leading to ROS accumulation, a decrease in that mitochondrial membrane potential that promoted cell cycle arrest and apoptosis [127].
Recent evidence has unveiled that HIF-1α has an important role in modulating recruitment and functions of immune cells to create an anti-tumorigenic microenvironment. Chromatin immunoprecipitation and luciferase reporter assay revealed that HIF-1α directly bound to a transcriptionally active HRE within the PD-L1 proximal promoter. Hence, hypoxia could modulate HIF-1α-transcriptional activity in order to increase PD-L1 expression on macrophages, dendritic cells and CM mouse models [128]. Along this line, in a study by Barsoum et al., HIF-1α induced PD-L1 expression in human breast and prostate cancer cells, as well as in mouse CM and mammary carcinoma cells, leading to resistance to CTL-mediated lysis [129]. More recently, high-throughput analyses performed in metastatic CM patients correlated hypoxia with non-response to anti-PD-1 and with immunosuppression and changes in tumor-stromal communication in TME. Intriguingly, this response was mainly orchestrated by the expression of HIF-2α in EC and fibroblasts within CM microenvironment. Indeed, HIF-2α inhibition with the small molecule PT2399, in combination with anti-PD1, delayed tumor growth in mice implanted with B16F10 murine CM cells. However, enhanced HIF-1α expression was observed in intra-tumoral EC following the anti-PD-1/PT2399 treatment. These findings suggest that the lack of HIF-2α could provoke a compensatory increase of the HIF-1α signaling to improve tumor growth control [130], thus impairing the effect of the combined anti-PD-1/HIF-2α inhibition.
In an in vivo model proposed by Wei et al., tumors form the aggressive B16F10 CM line grew significantly slower in mice lacking HIF-1α in their CD8+ T cells with respect to those implanted in wild-type mice. Similar effects were observed in CD8+ T cells treated with the HIF-1α inhibitor acriflavine alone, whereas the combinatorial treatment using acriflavine and the Treg depletion agent cytoxan further suppressed tumor development by improving CD8+ T cell responses [131]. Consistently, in a study by Lequez et al., the impairment of the transcriptional activity of HIF-1α significantly reduced B16F10 CM tumor growth by enhancing NK, CD4+, and CD8+ T cell infiltration. Acriflavine ameliorated the therapeutic benefit of anti-PD-1 immune checkpoint- and TRP-2 vaccine-based cancer immunotherapy [132]. Another HIF-1α inhibitor, namely IDF-11,774, also improved CD8+ T cell infiltration and impaired invasiveness of B16F10 CM cells in vivo by reducing the expression of the EMT markers SNAIL and N-cadherin [133]. On the other hand, dimethyloxalylglycine, a hypoxia mimetic agent, induced HIF-1α, HIF-2α and HIF-1β binding to the genes encoding the costimulatory receptors CD81, GITR, OX40 and 4-1BB, rising their expression in CD8+ T cells, and increasing T-cell mediated killing of CM cells in vivo. Similar effects were observed in CD8+ T-cells treated with the PHDs inhibitor molidustat [134]. Altogether, these results suggest that, although to target intra-tumoral hypoxia might improve the response to immunotherapy [135], HIF-α signaling activation in T-cells might further stimulate anti-tumor immunity in CM.
Conclusion
HIF-α subunits play an essential role in cancer since they modulate the transcription of several genes involved in tumour progression and response to therapies. As we have discussed in this review, although HIF-1α and HIF-2α are closely related and share similar domain structures, it is worth noting that they are mainly non-redundant and have distinct target genes in CM. Therefore, HIF-α subunits are driver of multiple aggressive features in CM, including angiogenesis, EMT and metastasis, metabolic alterations, but also immune escape.
The pleiotropic effects of HIF-α subunits in CM suggest that inhibiting both HIF-1α and HIF-2α might offer greater therapeutic benefits respect to target either protein alone. In this setting, combination therapies that incorporate HIF-α inhibitors with existing treatments, such as BRAF-targeted therapy and/or immune checkpoint inhibitors, might provide novel therapeutic options for CM patients with recurrent or metastatic disease. At present, however, the majority of studies with molecules that inhibit HIF-α expression or promote its degradation are mostly restricted to pre-clinical models. In the future, CM humanized mouse models may allow to accurately assess the efficacy of combining HIF-α inhibitors and immunotherapy.
HIF-1α and HIF-2α expression is regulated by different pathways involving both oxygen-dependent and oxygen-independent mechanisms. While the canonical oxygen-dependent HIF-α regulation has been well described, the mechanisms by which HIF-α subunits maintain their stability under normoxic conditions in CM are still less understood, highlighting a critical area for further investigation. These observations implicate that future drug development should focus on defining additional molecular pathways that allow HIF-1α subunits to escape degradation in normoxia and elucidating how this stability might contribute to CM development and progression.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- ACKR2:
-
Atypical chemokine receptor 2
- ADAM12:
-
ADAM Metallopeptidase Domain 12
- ADRP:
-
Adipose differentiation-related protein
- ANXA3:
-
Annexin A3
- AP2α:
-
Adaptor protein-2α
- AXL:
-
AXL Receptor Tyrosine Kinase
- ATP5ME:
-
ATP Synthase Membrane Subunit E
- bHLH:
-
Basic helix-loop-helix
- BIRC7:
-
Baculoviral IAP repeat-containing protein 7
- BNIP3:
-
BCL-2 interacting protein 3
- BRAFi:
-
BRAF inhibitor
- CAPN3:
-
Calpain-3
- CBP:
-
CREB-binding protein
- CM:
-
Cutaneous melanoma
- COL13A1:
-
Collagen type XIII alpha 1 chain
- CRIM1:
-
Cysteine-rich motor neuron 1
- CTGF:
-
Connective tissue growth factor
- CTL:
-
Cytotoxic T lymphocyte
- DAPK1:
-
Death Associated Protein Kinase 1
- DRD2:
-
D2 dopamine receptor
- EC:
-
Endothelial cells
- ECM:
-
Extracellular-matrix
- EMT:
-
Epithelial to mesenchymal transition
- ET-1:
-
Endothelin-1
- FABP3:
-
Fatty acid binding protein 3
- FABP7:
-
Fatty acid binding protein 7
- FAK:
-
Focal adhesion kinase
- FGF2:
-
Fibroblast Growth Factor 2
- FLNB:
-
Filamin B
- GALNT3:
-
Polypeptide N-Acetylgalactosaminyltransferase 3
- GLUT:
-
Glucose transporters
- GM3S:
-
Ganglioside GM3 synthase
- GPM6B:
-
Glycoprotein M6B
- GPR81:
-
G protein-coupled receptor 81
- HDAC:
-
Histone deacetylase
- HIF:
-
Hypoxia-inducible factors
- HK2:
-
Hexokinase 2
- HRE:
-
Hypoxia response elements
- HS3ST3A1:
-
Heparan Sulfate-Glucosamine 3-Sulfotransferase 3A1
- IL:
-
Interleukin
- ITGA3:
-
Integrin Subunit Alpha 3
- KCNMA1:
-
Potassium calcium-activated channel subfamily M alpha 1
- LDHA:
-
Lactate dehydrogenase
- lncRNA:
-
Long non-coding RNA
- miRNA:
-
MicroRNAs
- MITF:
-
Microphthalmia-associated transcription factor
- MMP:
-
Matrix metalloproteinase
- MT1A:
-
Metallothionein 1 A
- MYO1D:
-
Myosin ID
- ncRNA:
-
Non-coding RNA
- NDUFA6:
-
NADH: Ubiquinone Oxidoreductase Subunit
- NLGN4X:
-
Neuroligin
- NOX5:
-
NADPH oxidase 5
- ODD:
-
Oxygen-dependent degradation
- OXPHOS:
-
Oxidative phosphorylation
- PARP:
-
Poly (ADP-ribose) polymerase
- PAS:
-
Per-ARNT-Sim
- PDGFRα:
-
Platelet Derived Growth Factor Receptor Alpha
- PDH:
-
Pyruvate dehydrogenase
- PDK:
-
Pyruvate dehydrogenase kinase
- PDTC:
-
Pyrrolidine dithiocarbamate
- PFKFB3:
-
6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3
- PHD:
-
Prolyl hidroxylases
- PKA:
-
Protein kinase A
- PKM2:
-
Pyruvate kinase M2
- pre-miRNA:
-
miRNA precursor
- ROR:
-
Receptor tyrosine kinase-like orphan receptor
- ROS:
-
Reactive oxygen species
- SFK:
-
Src family kinases
- shRNA:
-
Short hairpin RNA
- SOX:
-
SRY-box transcription factor
- TAD:
-
Transactivation domain
- Tβ4:
-
Thymosin beta-4
- TCA:
-
Tricarboxylic acid
- TME:
-
Tumor microenvironment
- TNFRSF14:
-
TNF Receptor Superfamily Member 14
- Treg:
-
Regulatory T lymphocyte
- TYRP1:
-
Tyrosinase related protein 1
- uPAR:
-
Urokinase-type plasminogen activator receptor
- VEGF:
-
Vascular endothelial growth factor
- VEGFR:
-
VEGF receptor
- VHL:
-
Von Hippel-Lindau
- WT:
-
Wild type
References
Bigos KJ, Quiles CG, Lunj S, Smith DJ, Krause M, Troost EG, et al. Tumour response to hypoxia: Understanding the hypoxic tumour microenvironment to improve treatment outcome in solid tumours. Front Oncol. 2024;14:1331355. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2024.1331355.
Chen Z, Han F, Du Y, Shi H, Zhou W. Hypoxic microenvironment in cancer: molecular mechanisms and therapeutic interventions. Signal Transduct Target Therapy. 2023;8(1):70. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-023-01332-8.
Emami Nejad A, Najafgholian S, Rostami A, Sistani A, Shojaeifar S, Esparvarinha M, et al. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell Int. 2021;21(1):62. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-020-01719-5.
Semenza GL, Jiang BH, Leung SW, Passantino R, Concordet JP, Maire P, et al. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J Biol Chem. 1996;271(51):32529–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.271.51.32529.
Masoud GN, Li W. HIF-1α pathway: role, regulation and intervention for cancer therapy. Acta Pharm Sinica B. 2015;5(5):378–89. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.apsb.2015.05.007.
Guo Y, Xiao Z, Yang L, Gao Y, Zhu Q, Hu L, et al. Hypoxia–inducible factors in hepatocellular carcinoma (Review). Oncol Rep. 2020;43(1):3–15. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/or.2019.7397.
Albadari N, Deng S, Li W. The transcriptional factors HIF-1 and HIF-2 and their novel inhibitors in cancer therapy. Expert Opin Drug Discov. 2019;14(7):667–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/17460441.2019.1613370.
Ravenna L, Salvatori L, Russo MA. HIF3α: the little we know. Febs J. 2016;283(6):993–1003. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/febs.13572.
Zhou X, Guo X, Chen M, Xie C, Jiang J. HIF-3α promotes metastatic phenotypes in pancreatic cancer by transcriptional regulation of the RhoC–ROCK1 signaling pathway. Mol Cancer Res. 2018;16(1):124–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1541-7786.Mcr-17-0256.
Gao Q, Ren Z, Jiao S, Guo J, Miao X, Wang J, et al. HIF-3α-Induced miR-630 expression promotes cancer hallmarks in cervical cancer cells by forming a positive feedback loop. J Immunol Res. 2022;2022(1):5262963. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2022/5262963.
Kyurkchiyan SG, Stancheva G, Petkova V, Hadzhiev Y, Dobriyanova V, Popova D, et al.Exploration of the association between HIF3α mRNA and lncRNA MALAT1 in laryngeal squamous cell carcinoma by correlation analysis. Oncol Lett. 2024;28(1):292. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/ol.2024.14425.
Duan C. Hypoxia-inducible factor 3 biology: complexities and emerging themes. Am J Physiol Cell Physiol. 2016;310(4):C260–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/ajpcell.00315.2015.
Keith B, Johnson RS, Simon MC. HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat Rev Cancer. 2011;12(1):9–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrc3183.
Hu CJ, Sataur A, Wang L, Chen H, Simon MC. The N-terminal transactivation domain confers target gene specificity of hypoxia-inducible factors HIF-1alpha and HIF-2alpha. Mol Biol Cell. 2007;18(11):4528–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1091/mbc.e06-05-0419.
Huang LE, Bunn HF. Hypoxia-inducible factor and its biomedical relevance. J Biol Chem. 2003;278(22):19575–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.R200030200.
Contenti J, Guo Y, Larcher M, Mirabal-Ortega L, Rouleau M, Irondelle M, et al. HIF-1 inactivation empowers HIF-2 to drive hypoxia adaptation in aggressive forms of Medulloblastoma. Cell Death Discovery. 2024;10(1):338. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41420-024-02100-5.
Downes NL, Laham-Karam N, Kaikkonen MU, Ylä-Herttuala S. Differential but complementary HIF1α and HIF2α transcriptional regulation. Mol Ther. 2018;26(7):1735–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ymthe.2018.05.004.
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74(3):229–63. https://doiorg.publicaciones.saludcastillayleon.es/10.3322/caac.21834.
Crocetti E, Mallone S, Robsahm TE, Gavin A, Agius D, Ardanaz E, et al. Survival of patients with skin melanoma in Europe increases further: results of the EUROCARE-5 study. Eur J Cancer. 2015;51(15):2179–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejca.2015.07.039.
Sosman JA, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, Weber JS, et al. Survival in BRAF V600-mutant advanced melanoma treated with Vemurafenib. N Engl J Med. 2012;366(8):707–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa1112302.
Castellani G, Buccarelli M, Arasi MB, Rossi S, Pisanu ME, Bellenghi M, et al. BRAF Mutations in Melanoma: Biological Aspects, Therapeutic Implications, and Circulating Biomarkers. Cancers (Basel). 2023;15(16). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers15164026.
Ascierto PA, Kirkwood JM, Grob J-J, Simeone E, Grimaldi AM, Maio M, et al. The role of BRAF V600 mutation in melanoma. J Transl Med. 2012;10:85. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1479-5876-10-85.
Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature00766. https://www.nature.com/articles/nature00766#supplementary-information.
Akbani R, Akdemir Kadir C, Aksoy BA, Albert M, Ally A, Amin Samirkumar B, et al. Genomic classification of cutaneous melanoma. Cell. 2015;161(7):1681–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2015.05.044.
Reddy BY, Miller DM, Tsao H. Somatic driver mutations in melanoma. Cancer. 2017;123(S11):2104–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cncr.30593.
Zbytek B, Peacock DL, Seagroves TN, Slominski A. Putative role of HIF transcriptional activity in melanocytes and melanoma biology. Dermatoendocrinol. 2013;5(2):239–51. https://doiorg.publicaciones.saludcastillayleon.es/10.4161/derm.22678.
Kumar SM, Yu H, Edwards R, Chen L, Kazianis S, Brafford P, et al. Mutant V600E BRAF increases hypoxia inducible Factor-1α expression in melanoma. Cancer Res. 2007;67(7):3177–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.Can-06-3312.
Parmenter TJ, Kleinschmidt M, Kinross KM, Bond ST, Li J, Kaadige MR, et al. Response of BRAF-mutant melanoma to BRAF Inhibition is mediated by a network of transcriptional regulators of Glycolysis. Cancer Discov. 2014;4(4):423–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2159-8290.Cd-13-0440.
Montico B, Giurato G, Guerrieri R, Colizzi F, Salvati A, Nassa G, et al. Suppression of Spry1 reduces HIF1α-dependent Glycolysis and impairs angiogenesis in BRAF-mutant cutaneous melanoma. J Exp Clin Cancer Res. 2025;44(1):53. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13046-025-03289-8.
Wenger RH, Kurtcuoglu V, Scholz CC, Marti HH, Hoogewijs D. Frequently asked questions in hypoxia research. Hypoxia (Auckland NZ). 2015;3:35–43. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/hp.S92198.
Steinberger KJ, Eubank TD. The underexplored landscape of Hypoxia-Inducible factor 2 alpha and potential roles in tumor macrophages: A review. Oxygen. 2023;3(1):45–76.
Yang Y, Sun M, Wang L, Jiao B. HIFs, angiogenesis, and cancer. J Cell Biochem. 2013;114(5):967–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcb.24438.
Malekan M, Ebrahimzadeh MA, Sheida F. The role of Hypoxia-Inducible Factor-1alpha and its signaling in melanoma. Biomed Pharmacother. 2021;141:111873. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2021.111873.
Trisciuoglio D, Gabellini C, Desideri M, Ragazzoni Y, De Luca T, Ziparo E, et al. Involvement of BH4 domain of bcl-2 in the regulation of HIF-1-mediated VEGF expression in hypoxic tumor cells. Cell Death Differ. 2011;18(6):1024–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cdd.2010.175.
Folberg R, Hendrix MJC, Maniotis AJ. Vasculogenic mimicry and tumor angiogenesis. Am J Pathol. 2000;156(2):361–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0002-9440(10)64739-6.
Hendrix MJ, Seftor EA, Hess AR, Seftor RE. Vasculogenic mimicry and tumour-cell plasticity: lessons from melanoma. Nat Rev Cancer. 2003;3(6):411–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrc1092.
Sun B, Zhang D, Zhang S, Zhang W, Guo H, Zhao X. Hypoxia influences vasculogenic mimicry channel formation and tumor invasion-related protein expression in melanoma. Cancer Lett. 2007;249(2):188–97. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.canlet.2006.08.016.
Comito G, Calvani M, Giannoni E, Bianchini F, Calorini L, Torre E, et al. HIF-1α stabilization by mitochondrial ROS promotes Met-dependent invasive growth and vasculogenic mimicry in melanoma cells. Free Radic Biol Med. 2011;51(4):893–904. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.freeradbiomed.2011.05.042.
Spinella F, Caprara V, Cianfrocca R, Rosanò L, Di Castro V, Garrafa E, et al. The interplay between hypoxia, endothelial and melanoma cells regulates vascularization and cell motility through endothelin-1 and vascular endothelial growth factor. Carcinogenesis. 2014;35(4):840–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/carcin/bgu018.
Loftus SK, Baxter LL, Cronin JC, Fufa TD, Program NCS, Pavan WJ. Hypoxia-induced HIF1α targets in melanocytes reveal a molecular profile associated with poor melanoma prognosis. Pigment Cell Melanoma Res. 2017;30(3):339–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/pcmr.12579.
Widmer DS, Hoek KS, Cheng PF, Eichhoff OM, Biedermann T, Raaijmakers MIG, et al. Hypoxia contributes to melanoma heterogeneity by triggering HIF1α-dependent phenotype switching. J Invest Dermatol. 2013;133(10):2436–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/jid.2013.115.
Cheli Y, Giuliano S, Fenouille N, Allegra M, Hofman V, Hofman P, et al. Hypoxia and MITF control metastatic behaviour in mouse and human melanoma cells. Oncogene. 2012;31(19):2461–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/onc.2011.425.
O’Connell MP, Marchbank K, Webster MR, Valiga AA, Kaur A, Vultur A, et al. Hypoxia induces phenotypic plasticity and therapy resistance in melanoma via the tyrosine kinase receptors ROR1 and ROR2. Cancer Discov. 2013;3(12):1378–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/2159-8290.Cd-13-0005.
Zeng W, Su J, Wu L, Yang D, Long T, Li D, et al. CD147 promotes melanoma progression through hypoxia-induced MMP2 activation. Curr Mol Med. 2014;14(1):163–73. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/15665240113136660077.
Hu X, Su J, Zhou Y, Xie X, Peng C, Yuan Z et al. Repressing CD147 is a novel therapeutic strategy for malignant melanoma. Oncotarget. 2017;8(15).
Büchler P, Reber HA, Tomlinson JS, Hankinson O, Kallifatidis G, Friess H, et al. Transcriptional regulation of urokinase-type plasminogen activator receptor by hypoxia-inducible factor 1 is crucial for invasion of pancreatic and liver cancer. Neoplasia (New York NY). 2009;11(2):196–206. https://doiorg.publicaciones.saludcastillayleon.es/10.1593/neo.08734.
Nishi H, Sasaki T, Nagamitsu Y, Terauchi F, Nagai T, Nagao T, et al. Hypoxia inducible factor-1 mediates upregulation of urokinase-type plasminogen activator receptor gene transcription during hypoxia in cervical cancer cells. Oncol Rep. 2016;35(2):992–8. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/or.2015.4449.
Rofstad EK, Rasmussen H, Galappathi K, Mathiesen B, Nilsen K, Graff BA. Hypoxia promotes lymph node metastasis in human melanoma xenografts by Up-Regulating the Urokinase-Type plasminogen activator Receptor1. Cancer Res. 2002;62(6):1847–53.
Ryu Y-K, Im Y-S, Moon E-Y. Cooperation of actin-sequestering protein, thymosin β-4 and hypoxia inducible factor-1α in tumor cell migration. Oncol Rep. 2010;24(5):1389–94. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/or_00000997.
Bjørklund G, Dadar M, Aaseth J, Chirumbolo S. Thymosin Β4: A Multi-Faceted tissue repair stimulating protein in heart injury. Curr Med Chem. 2020;27(37):6294–305. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/0929867326666190716125456.
Makowiecka A, Malek N, Mazurkiewicz E, Mrówczyńska E, Nowak D, Mazur AJ. Thymosin Β4 regulates focal adhesion formation in human melanoma cells and affects their migration and invasion. Front Cell Dev Biology. 2019;7:304. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcell.2019.00304.
Xu H, Liu H, Li Z, Xu Q, Lin N, Li X. BIRC7 is beneficial for melanoma progression and hypoxic response. Clin Cosmet Investig Dermatol. 2022;15:1109–17. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/ccid.S370969.
Luo M, Zhu J, Ren J, Tong Y, Wang L, Ma S, et al. Lactate increases tumor malignancy by promoting tumor small extracellular vesicles production via the GPR81-cAMP-PKA-HIF-1α axis. Front Oncol. 2022;12. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fonc.2022.1036543.
Kierans SJ, Taylor CT. Regulation of Glycolysis by the hypoxia-inducible factor (HIF): implications for cellular physiology. J Physiol. 2021;599(1):23–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1113/JP280572.
Scott DA, Richardson AD, Filipp FV, Knutzen CA, Chiang GG, Ronai ZA, et al. Comparative metabolic flux profiling of melanoma cell lines: Beyond the warburg effect*. J Biol Chem. 2011;286(49):42626–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M111.282046.
Koch A, Ebert EV, Seitz T, Dietrich P, Berneburg M, Bosserhoff A, et al. Characterization of glycolysis-related gene expression in malignant melanoma. Pathol - Res Pract. 2020;216(1):152752. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.prp.2019.152752.
Datta KK, Periasamy P, Mohan SV, Ziegman R, Gowda H. Temporal quantitative proteomics reveals proteomic and phosphoproteomic alterations associated with adaptive response to hypoxia in melanoma cells. Cancers (Basel). 2021;13(9):2175.
Lakhter AJ, Hamilton J, Dagher PC, Mukkamala S, Hato T, Dong XC, et al. Ferroxitosis: a cell death from modulation of oxidative phosphorylation and PKM2-dependent Glycolysis in melanoma. Oncotarget. 2014;5(24):12694–703. https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.3031.
Liu T, Jin L, Chen M, Zheng Z, Lu W, Fan W, et al. Ku80 promotes melanoma growth and regulates antitumor effect of melatonin by targeting HIF1-α dependent PDK-1 signaling pathway. Redox Biol. 2019;25:101197. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2019.101197.
Nagao A, Kobayashi M, Koyasu S, Chow CCT, Harada H. HIF-1-Dependent reprogramming of glucose metabolic pathway of cancer cells and its therapeutic significance. Int J Mol Sci. 2019;20(2):238.
Shimizu T, Nagane M, Suzuki M, Yamauchi A, Kato K, Kawashima N, et al. Tumor hypoxia regulates ganglioside GM3 synthase, which contributes to oxidative stress resistance in malignant melanoma. Biochimica et biophysica acta (BBA) -. Gen Subj. 2020;1864(12):129723. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbagen.2020.129723.
Jin L, Yang G, Liu Y, Rang Z, Cui F. Bioinformatics data combined with single-cell analysis reveals patterns of Immunoinflammatory infiltration and cell death in melanoma. Int Immunopharmacol. 2024;143:113347. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.intimp.2024.113347.
Benoit A, Lequeux A, Harter P, Berchem G, Janji B. Atypical chemokine receptor 2 expression is directly regulated by hypoxia inducible factor-1 alpha in cancer cells under hypoxia. Sci Rep. 2024;14(1):26589. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-77628-8.
Roda JM, Wang Y, Sumner LA, Phillips GS, Marsh CB, Eubank TD. Stabilization of HIF-2α induces sVEGFR-1 production from tumor-associated macrophages and decreases tumor growth in a murine melanoma model. J Immunol. 2012;189(6):3168–77. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.1103817.
Liu S, Kumar SM, Martin JS, Yang R, Xu X. Snail1 mediates hypoxia-induced melanoma progression. Am J Pathol. 2011;179(6):3020–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ajpath.2011.08.038.
Steunou AL, Ducoux-Petit M, Lazar I, Monsarrat B, Erard M, Muller C, et al. Identification of the hypoxia-inducible factor 2α nuclear interactome in melanoma cells reveals master proteins involved in melanoma development. Mol Cell Proteom. 2013;12(3):736–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/mcp.M112.020727.
Finger EC, Cheng CF, Williams TR, Rankin EB, Bedogni B, Tachiki L, et al. CTGF is a therapeutic target for metastatic melanoma. Oncogene. 2014;33(9):1093–100. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/onc.2013.47.
Hanna SC, Krishnan B, Bailey ST, Moschos SJ, Kuan PF, Shimamura T, et al. HIF1α and HIF2α independently activate SRC to promote melanoma metastases. J Clin Invest. 2013;123(5):2078–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/jci66715.
Ibrahim YS, Amin AH, Jawhar ZH, Alghamdi MA, Al-Awsi GRL, Shbeer AM, et al. To be or not to be: regulatory T cells in melanoma. Int Immunopharmacol. 2023;118:110093. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.intimp.2023.110093.
Hsu T-S, Lin Y-L, Wang Y-A, Mo S-T, Chi P-Y, Lai AC-Y, et al. HIF-2α is indispensable for regulatory T cell function. Nat Commun. 2020;11(1):5005. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-020-18731-y.
Doedens AL, Phan AT, Stradner MH, Fujimoto JK, Nguyen JV, Yang E, et al. Hypoxia-inducible factors enhance the effector responses of CD8 + T cells to persistent antigen. Nat Immunol. 2013;14(11):1173–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ni.2714.
Halle S, Halle O, Förster R. Mechanisms and dynamics of T Cell-Mediated cytotoxicity in vivo. Trends Immunol. 2017;38(6):432–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.it.2017.04.002.
Liu H, Yang J, Zhang Y, Han J, Yang Y, Zhao Z, et al. Psychologic stress drives progression of malignant tumors via DRD2/HIF1α signaling. Cancer Res. 2021;81(20):5353–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.Can-21-1043.
Lando D, Peet DJ, Whelan DA, Gorman JJ, Whitelaw ML. Asparagine hydroxylation of the HIF transactivation domain: A hypoxic switch. Science. 2002;295(5556):858–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.1068592.
Yuan X, Ruan W, Bobrow B, Carmeliet P, Eltzschig HK. Targeting hypoxia-inducible factors: therapeutic opportunities and challenges. Nat Rev Drug Discovery. 2024;23(3):175–200. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41573-023-00848-6.
Martí JM, Garcia-Diaz A, Delgado-Bellido D, O’Valle F, González-Flores A, Carlevaris O, et al. Selective modulation by PARP-1 of HIF-1α-recruitment to chromatin during hypoxia is required for tumor adaptation to hypoxic conditions. Redox Biol. 2021;41:101885. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2021.101885.
Lakhter AJ, Lahm T, Broxmeyer HE, Naidu SR. Golgi associated HIF1a serves as a reserve in melanoma cells. J Cell Biochem. 2016;117(4):853–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcb.25381.
Mills CN, Joshi SS, Niles RM. Expression and function of hypoxia inducible factor-1 alpha in human melanoma under non-hypoxic conditions. Mol Cancer. 2009;8(1):104. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1476-4598-8-104.
Kuphal S, Winklmeier A, Warnecke C, Bosserhoff A-K. Constitutive HIF-1 activity in malignant melanoma. Eur J Cancer. 2010;46(6):1159–69. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejca.2010.01.031.
Martí-Díaz R, Montenegro MF, Cabezas-Herrera J, Goding CR, Rodríguez-López JN, Sánchez-del-Campo L. Acriflavine, a potent inhibitor of HIF-1α, disturbs glucose metabolism and suppresses ATF4-Protective pathways in melanoma under Non-Hypoxic conditions. Cancers (Basel). 2021;13(1):102.
Andreucci E, Pietrobono S, Peppicelli S, Ruzzolini J, Bianchini F, Biagioni A, et al. SOX2 as a novel contributor of oxidative metabolism in melanoma cells. Cell Communication Signal. 2018;16(1):87. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-018-0297-z.
Kuschel A, Simon P, Tug S. Functional regulation of HIF-1α under normoxia–is there more than post-translational regulation? J Cell Physiol. 2012;227(2):514–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcp.22798.
Schörghofer D, Vock L, Mirea MA, Eckel O, Gschwendtner A, Neesen J, et al. Late stage melanoma is hallmarked by low NLGN4X expression leading to HIF1A accumulation. Br J Cancer. 2024;131(3):468–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41416-024-02758-9.
Buscà R, Berra E, Gaggioli Cd, Khaled M, Bille K, Marchetti B, et al. Hypoxia-inducible factor 1α is a new target of microphthalmia-associated transcription factor (MITF) in melanoma cells. J Cell Biol. 2005;170(1):49–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1083/jcb.200501067.
Antony S, Jiang G, Wu Y, Meitzler JL, Makhlouf HR, Haines DC, et al. NADPH oxidase 5 (NOX5)—induced reactive oxygen signaling modulates normoxic HIF-1α and p27Kip1 expression in malignant melanoma and other human tumors. Mol Carcinog. 2017;56(12):2643–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/mc.22708.
Lingappan K. NF-κB in oxidative stress. Curr Opin Toxicol. 2018;7:81–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cotox.2017.11.002.
Li ZL, Ji JL, Wen Y, Cao JY, Kharbuja N, Ni WJ, et al. HIF-1α is transcriptionally regulated by NF-κB in acute kidney injury. Am J Physiol Ren Physiol. 2021;321(2):F225–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/ajprenal.00119.2021.
Liu H, Li Y, Xiong J. The role of Hypoxia-Inducible Factor-1 alpha in renal disease. Molecules. 2022;27(21). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/molecules27217318.
Kim J, Shao Y, Kim SY, Kim S, Song HK, Jeon JH, et al. Hypoxia-induced IL-18 increases hypoxia-inducible factor-1alpha expression through a Rac1-dependent NF-kappaB pathway. Mol Biol Cell. 2008;19(2):433-44. https://doiorg.publicaciones.saludcastillayleon.es/10.1091/mbc.e07-02-0182.
Xu B, Zhang X, Gao Y, Song J, Shi B. Microglial Annexin A3 promoted the development of melanoma via activation of hypoxia-inducible factor-1α/vascular endothelial growth factor signaling pathway. J Clin Lab Anal. 2021;35(2):e23622. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcla.23622.
Vara-Pérez M, Rossi M, Van den Haute C, Maes H, Sassano ML, Venkataramani V, et al. BNIP3 promotes HIF-1α-driven melanoma growth by curbing intracellular iron homeostasis. EMBO J. 2021;40(10):e106214. https://doiorg.publicaciones.saludcastillayleon.es/10.15252/embj.2020106214.
Kim JY, Cho H, Yoo J, Kim GW, Jeon YH, Lee SW, et al. HDAC8 deacetylates HIF-1α and enhances its protein stability to promote tumor growth and migration in melanoma. Cancers (Basel). 2023;15(4):1123.
Yi Z, Jiang L, Zhao L, Zhou M, Ni Y, Yang Y, et al. Glutathione peroxidase 3 (GPX3) suppresses the growth of melanoma cells through reactive oxygen species (ROS)-dependent stabilization of hypoxia-inducible factor 1-α and 2-α. J Cell Biochem. 2019;120(11):19124–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcb.29240.
Montico B, Giurato G, Pecoraro G, Salvati A, Covre A, Colizzi F, et al. The pleiotropic roles of circular and long noncoding RNAs in cutaneous melanoma. Mol Oncol. 2022;16(3):565–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/1878-0261.13034.
Ghafouri-Fard S, Hussen BM, Shoorei H, Abak A, Poornajaf Y, Taheri M, et al. Interactions between non-coding RNAs and HIF-1α in the context of cancer. Eur J Pharmacol. 2023;943:175535. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejphar.2023.175535.
Jawad SF, Altalbawy FMA, Hussein RM, Fadhil AA, Jawad MA, Zabibah RS, et al. The strict regulation of HIF-1α by non-coding RNAs: new insight towards proliferation, metastasis, and therapeutic resistance strategies. Cancer Metastasis Rev. 2024;43(1):5–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10555-023-10129-8.
Shang R, Lee S, Senavirathne G, Lai EC. MicroRNAs in action: biogenesis, function and regulation. Nat Rev Genet. 2023;24(12):816–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41576-023-00611-y.
Peng X, Gao H, Xu R, Wang H, Mei J, Liu C. Correction to: the interplay between HIF-1α and noncoding RNAs in cancer. J Exp Clin Cancer Res. 2020;39(1):45. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13046-020-01544-8.
Silina MV, Dzhalilova DS, Makarova OV. Role of MicroRNAs in regulation of cellular response to hypoxia. Biochem (Mosc). 2023;88(6):741–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1134/s0006297923060032.
Valencia-Cervantes J, Sierra-Vargas MP. Regulation of Cancer-Associated MiRNAs expression under hypoxic conditions. Anal Cell Pathol (Amst). 2024;2024:5523283. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2024/5523283.
Yang X, Lei S, Long J, Liu X, Wu Q. MicroRNA-199a-5p inhibits tumor proliferation in melanoma by mediating HIF-1α. Mol Med Rep. 2016;13(6):5241–7. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/mmr.2016.5202.
Zhou J, Xu D, Xie H, Tang J, Liu R, Li J, et al. miR-33a functions as a tumor suppressor in melanoma by targeting HIF-1α. Cancer Biol Ther. 2015;16(6):846–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/15384047.2015.1030545.
Cao K, Li J, Chen J, Qian L, Wang A, Chen X et al. microRNA-33a-5p increases radiosensitivity by inhibiting Glycolysis in melanoma. Oncotarget. 2017;8(48).
Mazar J, Qi F, Lee B, Marchica J, Govindarajan S, Shelley J, et al. MicroRNA 211 functions as a metabolic switch in human melanoma cells. Mol Cell Biol. 2016;36(7):1090–108. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/MCB.00762-15.
Moritz RKC, Ebelt N, Rattay T, Ehrenreich J, Sunderkötter C, Gerloff D. The expression of miR-211-5p in Sentinel lymph node metastases of malignant melanoma is a potential marker for poor prognosis. Int J Mol Sci. 2024;25(19):10859.
Díaz-Martínez M, Benito-Jardón L, Alonso L, Koetz-Ploch L, Hernando E, Teixidó J. miR-204-5p and miR-211-5p contribute to BRAF inhibitor resistance in melanoma. Cancer Res. 2018;78(4):1017–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.Can-17-1318.
Sahoo A, Sahoo SK, Joshi P, Lee B, Perera RJ. MicroRNA-211 loss promotes metabolic vulnerability and BRAF inhibitor sensitivity in melanoma. J Invest Dermatol. 2019;139(1):167–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jid.2018.06.189.
Zhao CC, Guo H, Wang Y, Li JH. Comprehensive upstream and downstream regulatory analyses identify miR-675-3p as a potential prognostic biomarker in melanoma. Hum Cell. 2021;34(2):654–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13577-020-00473-0.
Hwang HW, Baxter LL, Loftus SK, Cronin JC, Trivedi NS, Borate B, et al. Distinct MicroRNA expression signatures are associated with melanoma subtypes and are regulated by HIF1A. Pigment Cell Melanoma Res. 2014;27(5):777–87. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/pcmr.12255.
Zhang Z, Sun H, Dai H, Walsh R, Imakura M, Schelter J, et al. MicroRNA miR-210 modulates cellular response to hypoxia through the MYC antagonist MNT. Cell Cycle. 2009;8(17):2756–68. https://doiorg.publicaciones.saludcastillayleon.es/10.4161/cc.8.17.9387.
Ono S, Oyama T, Lam S, Chong K, Foshag LJ, Hoon DSB. A direct plasma assay of Circulating microRNA-210 of hypoxia can identify early systemic metastasis recurrence in melanoma patients. Oncotarget. 2015;6(9).
Noman MZ, Buart S, Romero P, Ketari S, Janji B, Mari B, et al. Hypoxia-Inducible miR-210 regulates the susceptibility of tumor cells to Lysis by cytotoxic T cells. Cancer Res. 2012;72(18):4629–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.Can-12-1383.
Špaková I, Graier WF, Rabajdová M, Dubayová K, Nagyová V, Mareková M. Hypoxia factors suppression effect on the energy metabolism of a malignant melanoma cell SK-MEL-30. Eur Rev Med Pharmacol Sci. 2020;24(9):4909–20. https://doiorg.publicaciones.saludcastillayleon.es/10.26355/eurrev_202005_21180.
Špaková I, Rabajdová M, Mičková H, Graier WF, Mareková M. Effect of hypoxia factors gene Silencing on ROS production and metabolic status of A375 malignant melanoma cells. Sci Rep. 2021;11(1):10325. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-021-89792-2.
Hao T, Li CX, Ding XY, Xing XJ. MicroRNA-363-3p/p21(Cip1/Waf1) axis is regulated by HIF-2α in mediating stemness of melanoma cells. Neoplasma. 2019;66(3):427–36. https://doiorg.publicaciones.saludcastillayleon.es/10.4149/neo_2018_180828N655.
Liu Y, He D, Xiao M, Zhu Y, Zhou J, Cao K. Long noncoding RNA LINC00518 induces radioresistance by regulating Glycolysis through an miR-33a-3p/HIF-1α negative feedback loop in melanoma. Cell Death Dis. 2021;12(3):245. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-021-03523-z.
Kotowski K, Supplitt S, Wiczew D, Przystupski D, Bartosik W, Saczko J, et al. 3PO as a Selective Inhibitor of 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 3 in A375 Human Melanoma Cells. Anticancer Res. 2020;40(5):2613–25. https://doiorg.publicaciones.saludcastillayleon.es/10.21873/anticanres.14232.
Furfaro AL, Loi G, Ivaldo C, Passalacqua M, Pietra G, Mann GE, et al. HO-1 limits the efficacy of Vemurafenib/PLX4032 in BRAF(V600E) mutated melanoma cells adapted to physiological normoxia or hypoxia. Antioxid (Basel Switzerland). 2022;11(6). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antiox11061171.
Pucciarelli D, Lengger N, Takáčová M, Csaderova L, Bartosova M, Breiteneder H, et al. Hypoxia increases the heterogeneity of melanoma cell populations and affects the response to Vemurafenib. Mol Med Rep. 2016;13(4):3281–8. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/mmr.2016.4888.
Qin Y, Roszik J, Chattopadhyay C, Hashimoto Y, Liu C, Cooper ZA, et al. Hypoxia-Driven mechanism of Vemurafenib resistance in melanoma. Mol Cancer Ther. 2016;15(10):2442–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1535-7163.Mct-15-0963.
Yang Y, Wu J, Demir A, Castillo-Martin M, Melamed RD, Zhang G, et al. GAB2 induces tumor angiogenesis in NRAS-driven melanoma. Oncogene. 2013;32(31):3627–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/onc.2012.367.
Monsel G, Ortonne N, Bagot M, Bensussan A, Dumaz N. c-Kit mutants require hypoxia-inducible factor 1α to transform melanocytes. Oncogene. 2010;29(2):227–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/onc.2009.320.
Maphutha J, Twilley D, Lall N. The role of the PTEN tumor suppressor gene and its Anti-Angiogenic activity in melanoma and other cancers. Molecules. 2024;29(3):721.
Wang Y, Ma J, Yan X, Chen X, Si L, Liu Y, et al. Isoliquiritigenin inhibits proliferation and induces apoptosis via alleviating hypoxia and reducing Glycolysis in mouse melanoma B16F10 cells. Recent Pat Anticancer Drug Discov. 2016;11(2):215–27. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1573406412666160307151904.
Park E-J, Lee Y-M, Oh T-I, Kim BM, Lim B-O, Lim J-H. Vanillin suppresses cell motility by inhibiting STAT3-Mediated HIF-1α mRNA expression in malignant melanoma cells. Int J Mol Sci. 2017;18(3):532.
Li C, Wang Q, Shen S, Wei X, Li G. HIF-1α/VEGF signaling-mediated epithelial–mesenchymal transition and angiogenesis is critically involved in anti-metastasis effect of Luteolin in melanoma cells. Phytother Res. 2019;33(3):798–807. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ptr.6273.
Zhao P, He X-B, Chen X-Y, Li Z-L, Xing W-J, Liu W, et al. Celastrol inhibits mouse B16-F10 melanoma cell survival by regulating the PI3K/AKT/mTOR signaling pathway and repressing HIF-1α expression. Discover Oncol. 2024;15(1):178. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12672-024-01045-6.
Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct target of HIF-1α, and its Blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211(5):781–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1084/jem.20131916.
Barsoum IB, Smallwood CA, Siemens DR, Graham CH. A mechanism of Hypoxia-Mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014;74(3):665–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.Can-13-0992.
Huang AY, Burke KP, Porter R, Meiger L, Fatouros P, Yang J, et al. Stratified analysis identifies HIF-2α as a therapeutic target for highly immune-infiltrated melanomas. BioRxiv: Preprint Serv Biology. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/2024.10.29.620300.
Wei P, Kou W, Riaz F, Zhang K, Fu J, Pan F. Combination therapy of HIFα inhibitors and Treg depletion strengthen the anti-tumor immunity in mice. Eur J Immunol. 2023;53(12):2250182. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/eji.202250182.
Lequeux A, Noman MZ, Xiao M, Van Moer K, Hasmim M, Benoit A, et al. Targeting HIF-1 alpha transcriptional activity drives cytotoxic immune effector cells into melanoma and improves combination immunotherapy. Oncogene. 2021;40(28):4725–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41388-021-01846-x.
Kim NH, Jeong JH, Park YJ, Shin HY, Choi WK, Lee K, et al. Anti-Tumor effect of IDF-11774, an inhibitor of Hypoxia-Inducible Factor-1, on melanoma. Biomol Ther (Seoul). 2022;30(5):465–72. https://doiorg.publicaciones.saludcastillayleon.es/10.4062/biomolther.2022.061.
Yang Y, Salman S, Dordai D, Lyu Y, Datan E, Drehmer D, et al. Pharmacologic HIF stabilization activates costimulatory receptor expression to increase antitumor efficacy of adoptive T cell therapy. Sci Adv. 2024;10(35):eadq2366. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.adq2366.
Semenza GL. Targeting intratumoral hypoxia to enhance anti-tumor immunity. Semin Cancer Biol. 2023;96:5–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcancer.2023.09.002.
Acknowledgements
Not applicable.
Funding
This work was supported by the Italian Ministry of Health (Ricerca Corrente, grant GR-2018-12366312 to EF) and 5 × 1000 Institutional Grant from CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) (seed grant to BM and EF; no grant number provided).
Author information
Authors and Affiliations
Contributions
AB, BM and EF conceptualised the article, performed literature search and wrote the initial manuscript. AB prepared the table. BM designed figures. RG, FC and JP contributed to writing the manuscript. EF revised the manuscript and provided substantial feedback throughout the writing process. All authors approved the final version of the manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
All the authors agree to publish this paper.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Bellazzo, A., Montico, B., Guerrieri, R. et al. Unraveling the role of hypoxia-inducible factors in cutaneous melanoma: from mechanisms to therapeutic opportunities. Cell Commun Signal 23, 177 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-025-02173-4
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-025-02173-4