Skip to main content

Alpha-melanocyte stimulating hormone (α-MSH): biology, clinical relevance and implication in melanoma

Abstract

Alpha-melanocyte stimulating hormone (α-MSH) and its receptor, melanocortin 1 receptor (MC1R), have been proposed as potential target for anti-cancer strategies in melanoma research, due to their tissue specific expression and involvement in melanocyte homeostasis. However, their role in prevention and treatment of melanoma is still debated and controversial. Although a large body of evidence supports α-MSH in preventing melanoma development, some preclinical findings suggest that the α-MSH downstream signalling may promote immune escape and cancer resistance to therapy. Additionally, in metastatic melanoma both MC1R and α-MSH have been reported to be overexpressed at levels much higher than normal cells. Furthermore, targeted therapy (e.g. BRAF inhibition in BRAFV600E mutant tumours) has been shown to enhance this phenomenon. Collectively, these data suggest that targeting MC1R could serve as an approach in the treatment of metastatic melanoma. In this review, we explore the molecular biology of α-MSH with particular emphasis into its tumor-related properties, whilst elaborating the experimental evidence currently available regarding the interplay between α-MSH/MC1R axis, melanoma and antitumor strategies.

Introduction

Melanocortins are peptidic pituitary hormones produced by the cleavage and posttranslational modifications of pro-opiomelanocortin hormone (POMC). The family of melanocortins includes Adrenocorticotropic Hormone (ACTH), Melanocyte Stimulating Hormone (MSH) and endorphins, that activate five forms of membrane receptors called Melanocortin Receptors (MCRs) with different affinities. MSH consists of the three forms α-, β- and γ-MSH. Among them α-MSH is well-characterized and first described for its melanin-inducing activity in frogs. α-MSH is a 13 amino acid neuropeptide secreted by melanocytes and keratinocytes after ultraviolet light exposure and it is responsible of the melanin synthesis, being the main actor of skin pigmentation [1,2,3]. Moreover, it has been shown that α-MSH and analogues have anti-inflammatory and anti-microbial properties, activating melanocortin receptors (MCR) signaling [4, 5]. α-MSH binds to four out of five MCR subtypes (MC1R, MC3R, MC4R, MC5R), regulating several downstream cascades in different cell types. Notably, in melanocytes MC1R is highly expressed and the binding of α-MSH promotes the expression of melanogenesis enzyme genes via Adenylyl Cyclase (AC)/cyclic AMP (cAMP)/Protein Kinase A (PKA) pathway. Beyond melanin synthesis, the α-MSH/MC1R axis controls a plethora of important processes such as DNA damage repair, reduction of free radical production and cell proliferation among others. For the broad spectrum of properties, the use of α-MSH or its synthetic analogs has been proposed for several pathologic conditions. The primary target cell for α-MSH is the melanocyte, in which, despite the proven efficacy in the prevention of melanoma development, its role in malignant melanoma, and in particular in metastatic stage disease still remains underinvestigated [6].

2- Molecular biology of α-MSH

α-MSH production and melanocortin receptors

Human POMC gene is located on chromosome 2p23.3 and it is expressed in a variety of tissues but broadly in testis, pancreas and fat tissue. The early encoded protein undergoes extensive posttranslational processing via prohormone convertases cleavage, in order to produce at least ten active peptides mainly synthesized in corticotroph cells of the anterior pituitary. Among them, ACTH is essential for physiologic steroidogenesis whereas in other tissues such as placenta and epithelium, proteolytic cleavage gives rise to peptides with roles in energy homeostasis, melanocyte stimulation, and immune modulation. These include several distinct melanotropins (or melanocortins): α-, β- and γ-MSH. All forms of MSHs bind to four well characterised G-Protein Coupled Receptor (GPCR) subtypes: Melanocortin Receptors (MC1R, MC3R, MC4R, and MC5R), whereas MC2R is specific for the binding with ACTH [7,8,9,10].

MC1R is an intron less gene encoding seven pass transmembrane GPCR, preferentially expressed on cell membrane of melanocytes and mainly recognized as the key regulator of the synthesis of epidermal melanin pigments [11, 12]. MC1R gene is polymorphic and frequent variants are associated not only with hair/skin phenotypes but also with increased melanoma risk [13,14,15,16]. MC1R is also the target of the α-MSH antagonists Agouti protein and Agouti related protein (Agrp), both responsible for the inhibition of eumelanin production in favour of pheomelanin [17].

MC3R and MC4R genes encode the GPCRs for MSH and ACTH and are expressed in tissues other than the adrenal cortex and melanocytes. Studies suggest a function role of MC3R and MC4R in the regulation of energy homeostasis and food intake. Mutations of this receptors have been correlated to susceptibility to obesity and anorexia in humans [18,19,20,21,22]. Evidence suggests that MC5R plays a key role in the regulation of sexual behaviour, thermoregulation and exocrine secretion (sebogenesis) but also in immune reaction and inflammatory response via cAMP signal transduction [23, 24].

α-MSH regulation of melanocyte function: MC1R/cAMP signaling cascade

MC1R plays a key role in cutaneous homeostasis and photoprotection as it is coupled to the stimulatory G protein Gα which in turn activates AC switching on the cAMP/PKA pathway [25].

PKA phosphorylates the transcription factor cAMP Response Element Binding Protein (CREB) that stimulates the Microphthalmia inducing Transcription Factor (MiTF) which in turn promotes the expression of melanogenesis enzyme genes Tyrosinase (TYR), Tyrosinase Related Protein 1 and 2 (TRP1,TRP2) and Dopachrome Tautomerase (DCT) [26, 27]. MiTF coordinates a broad range of biological processes including cell survival, differentiation, proliferation, migration, invasion, senescence, metabolism, and DNA damage repair (Fig. 1).

Fig. 1
figure 1

A Physiological condition. B Pathological condition (advanced stage melanoma). In physiological condition melanocytes express a membrane receptor (MC1R) that controls the melanin synthesis process. A Upon UV exposure, alpha-melanocyte stimulating hormone (α-MSH) is released by keratinocytes: the binding of α-MSH to MC1R activates Adenyl Ciclase (AC) that stimulates cilic AMP (cAMP) production and the activation of Protein Kinase A (PKA). PKA phosphorylates the transcription factor CREB that stimulates the transcription factor MiTF which in turn promotes the expression of melanogenesis enzyme genes TYR, TRP1 and DCT. In our working hypothesis B in advanced stage of melanoma, tumour cells overexpress MC1R and BRAF inhibitor treatment significantly increase this MC1R expression via MiTF-dependent pathways, leading to enhanced ligand binding on the cell surface. As a consequence, the cAMP/PKA pathway is aberrantly altered and might promote tumour migration , growth and proliferation. PM: Plasmatic Membrane; Gα, Gβ, Gγ: G proteins; CREB: cAMP Response Element Binding protein; RTK Tyrosine Kinase Receptor. This figure was created with www.BioRender.com

α-MSH stimulated MC1R triggers the production of both free radicals (ROS) and brown/black eumelanin, acting as a filter against UV. MC1R polymorphisms are associated with pigmentary phenotypes such as Red-Hair-Colour (RHC) and light skin [28, 29]. Patients carrying these variants show a reduced ability to produce eumelanin and therefore pheomelanin synthesis prevails. Pheomelanin acts as a photosensitizer and these patients are more susceptible to skin cancer development, both by UV- dependent and independent mechanisms [30].

Other pathways connected with α-MSH/MC1R signal

The Mitogen-Activated Protein Kinase (MAPK) signal transduction cascades are highly conserved regulators of cell proliferation, differentiation and survival which are activated by signals as cytokines, growth factors and other stress inducers. The most widely studied MAPK pathway is the RAS/RAF/MEK/ERK cascade that controls melanogenesis and it is aberrantly activated in 90% of human cutaneous melanomas as well as in several type of cancers. Gain of Function (GoF) mutations in N-RAS and B-RAF are common drivers for melanoma development (~ 25% for N-RAS and ~ 60% for B-RAF) as they are responsible for dysregulated cell cycle and proliferation [31,32,33,34,35]. Multiple stimuli such as growth factors, cytokines, viruses, GPCR ligands and oncogenes can sequentially activate the ERK pathway and result in ERK1/2 phosphorylation that regulates different transcription factors, including c-FOS, cJUN, ELK-1, c-MYC, and ATF-2 controlling cell growth, migration, and differentiation [36]. Noteworthy, ERK1/2 can phosphorylate MiTF decreasing its protein levels and leading to a negative regulation of melanogenic enzymes, inhibiting melanogenesis process. In human melanocytic cells ERK activation upon α-MSH binding to MC1R is a cAMP-independent process, it occurs through a transactivation mechanism of the Tyrosine Kinase Receptor (RTK) c-KIT and plays an important role in melanogenesis [37,38,39].

Another pathway linked to α-MSH/MC1R axis is PI3K/AKT, an intracellular signal transduction cascade that, through the phosphorylation of several downstream substrates, is involved in cellular functions such as cell growth, proliferation, and differentiation. The key molecules involved in this signalling pathway are RTKs, phosphatidylinositol 3-kinase (PI3K), phosphatidylinositol-4,5-bisphosphate (PIP2), phosphatidylinositol-3,4,5-triphosphate (PIP3) and AKT/protein kinase B. The binding of RTK with growth factors and various stimuli activates PI3K which in turn phosphorylates PIP2 leading to the production of the second messenger PIP3 that regulates metabolic processes by recruiting signaling proteins, including AKT/Protein kinase B (PKB) [40]. PTEN (Phosphatase and TENsin homolog) is a phosphatase responsible for the conversion of PIP3 to PIP2, acting as an antagonist of the PI3K/AKT response. Investigating the interaction between MC1R and PI3K/PTEN signaling, it has been shown that upon α-MSH binding, MC1R interacts with PTEN and, by preventing its degradation, inactivates AKT. It has also been shown that RHC MC1R allelic variants have an impaired ability to interact with PTEN, thus increasing AKT signaling and predisposing melanocytes to melanomagenesis [41]. Studies with a synthetic analog of α-MSH revealed that the stimulation of RHC MC1R variants activates DNA repair pathways through a cAMP-independent mechanism mediated by AKT activation [42]. On the other hand, it has been shown that the binding of α-MSH to MC1R activates DNA repair and antioxidant signals in a cAMP-dependent manner with decreased AKT phosphorylation [43]. Moreover an interplay between α-MSH/MC1R and Peroxisome Proliferator-Activated Receptor-γ (PPAR-γ) has been reported [44]. Briefly, α-MSH induces the release of calcium (Ca2+) from endoplasmic reticulum (ER) by a phospholipase C (PLC) dependent mechanism and Ca2+ efflux is connected with the translocation of PPARγ into the nucleus, where it promotes the transcription of target genes involved in lipid metabolism, adipogenesis, maintenance of metabolic homeostasis, inflammation and anticancer effects in a variety of human tumours [45].

α-MSH/MC1R: range of action

Maintenance of cell integrity and DNA damage repair. MC1R polymorphism

In physiologic conditions, the main role of α-MSH is to protect skin from UV exposure by coordinating the production of eumelanin. However, both in melanocytes and keratinocytes, several studies have established that the α-MSH/MC1R-cAMP axis is also involved in additional responses, like antioxidant defences and DNA damage repair [42, 46]. UV radiation and melanin synthesis process are sources of ROS among which hydrogen peroxide (H2O2), that is able to injure all cell compartments [47]. After UV exposure, human melanocytes stimulate the generation of H2O2 with a concomitant decrease in the activity of catalase, the enzyme most involved in H2O2 neutralization [48]. Therefore, it has been shown that treatment with α-MSH protects melanocytes from oxidative stress since α-MSH through MC1R induces both the activation and overexpression of catalase, reducing H2O2 production [49, 50].

Exposure to UV radiation is considered the most common environmental risk factor for skin melanoma. The high prevalence of polymorphisms of MC1R, with more than 300 variants, makes it the best-established susceptibility gene for cutaneous melanoma [25, 51]. The association between some MC1R polymorphisms and red hair, freckles, and inability to tan (the RHC phenotype) was first reported in 1995 by Valverde et al. [52]. An extensive body of research shows that inactivating variants of MC1R are the main contributors to the increased risk of melanoma development, because the functions of UV protection and DNA damage repair are lost. According to their penetrance RHC MC1R alleles have been classified as high (R) or low (r) variants. “R” variants include D84E, R142H, R151C, R160W, and D294H and people carrying these variants MC1R have the highest risk of developing melanoma and non-melanoma skin cancers whereas “r” variants: V60L, V92M, and R163Q showed a weaker association with the RHC phenotype [52,53,54,55,56].

In keratinocytes, the canonical α-MSH/MC1R-cAMP-PKA pathway enhances Nucleotide Excision Repair (NER) activity: PKA directly phosphorylates the DNA damage sensors Ataxia Telangiectasia Mutated (ATM) and Rad3 related (ATR) which actively recruits the key NER protein Xeroderma Pigmentosum complementation group A (XPA) to sites of nuclear UV damage, thus accelerating the clearance of UV-induced lesions and reducing the mutagenesis rate [57].

It has been reported that α-MSH-MC1R axis can induce cutaneous carcinogenesis other than melanoma. Regarding Non-Melanoma Skin Cancers (NMSCs), it must be highlighted that carriers of two MC1R variant alleles have a higher risk of developing NMSC than the WT. However, it is not clear whether MC1R variants confer a relevant contribution in the genesis of skin carcinomas [58].

Anti inflammatory and immunomodulatory properties

In addition to its effects on melanocytes, α-MSH has potent anti-inflammatory effects when administered systemically or locally [59]. Its immunomodulating properties rely mainly on the binding with MC1R that is also expressed on monocytes, macrophages, and dendritic cells (DCs). α-MSH downregulates the production of pro-inflammatory cytokines IL-1, IL-6, TNF-α, IL-2, IFN-γ, IL-4, IL-13 and in contrast, anti-inflammatory IL-10 production is upregulated. At the molecular level, α-MSH affects several pathways implicated in the regulation of transcription factors such as NFκB thus modulating inflammatory cell proliferation, activity and migration. NFκB regulates the transcription of genes involved in cell survival, and inhibition of NFκB activation has been considered as a strategy for the treatment of melanoma [60,61,62,63]. α-MSH was discovered to be an ancient natural antimicrobial agent against two representative pathogens Staphylococcus A. and Candida A., enhancing the local inflammatory reaction. It has been described that the candidacidal activity is mostly based on increasing intracellular cAMP levels that interferes with microbial regulatory pathway thus reducing fungal viability and germ tube formation [64].

From an oncological perspective, in human melanoma cells, an anti-inflammatory and anti-invasive effects of α-MSH have been reported [65].

Broad spectrum of α-MSH applications

The pivotal role of α-MSH in stimulating skin pigmentation and protecting from UV damage led to propose its topical application as strategy to improve a “sunless tanning” both for cosmetic purpose and mostly as skin cancer prevention. Therefore, by boosting the α-MSH/MC1R-cAMP/PKA pathway activation and MiTF transcription, melanogenesis and DNA damage repair apparatus are enhanced [66].

Moreover, studies revealed that α-MSH and synthetic analog peptides could be resolutive for other conditions as Hypoactive Sexual Desire Disorders (HSDD) or be neuroprotective against cerebral ischemia/reperfusion injury as well as neovascularization inhibition [67,68,69]. Additionally α-MSH was found to be involved in appetite regulation (suppressor), in the pathogenesis of restless legs syndrome and in insulin resistance/sensitivity [70,71,72,73].

α-MSH/MC1R and cancer

Melanoma

Cutaneous malignant melanoma arises from melanocytes, the pigment producing cells, and remains a challenging disease due both to difficult early diagnoses and to the tendency to metastasize quickly to lymph nodes and distant organs such as liver, lung and brain. Although melanoma accounts for only about 10% of skin cancers it is responsible for the vast majority of deaths [74, 75].

Mortality is correlated with the stage at diagnosis and, to date, the management of metastatic disease remains a relevant clinical issue. Genetic mutations in oncogenes and tumour suppressor genes affecting the RAS-RAF-MEK-ERK signalling pathway (MAPK) are the main drivers in most cutaneous melanomas. A common mutation found in melanoma patients is BRAFV600E whereas tumours bearing NRAS mutations are less frequent but more aggressive and associated with shorter survival [76]. The MAPK cascade leads to activation of ERK1 and ERK2 which translocate into the nucleus to regulate MiTF, cMYC and other transcription factors to sustain cell cycle progression, tumor invasion, metastasis and immune evasion [77].

The BRAFV600E mutation is found only in about 50% of melanoma and this fact limits the use of BRAF inhibitors (BRAFi). Moreover, most of patients in BRAFi therapy for metastatic melanoma relapses early after an initial partial response. The development of drug resistance within some metastatic clones causes the relapse of disease.

MC1R overexpression in melanoma

α-MSH/MC1R/cAMP axis converges to the regulation of MiTF expression with a pivotal role for homeostasis but when impaired in melanoma environment it takes a role in tumor progression and survival. It has been reported that MiTF is a factor that supports melanoma stem cells properties [27, 78, 79].

Many studies showed increased levels of MC1R expression on the surface of most melanomas (either primary or metastatic tissues) but not in carcinoma cell lines making it a valuable marker of melanoma cells [80, 81]. Moreover, the tumor itself overproduces α-MSH, leading to an autocrine hyperproliferative process, described in melanoma metastases [82].

EPAC in melanoma

cAMP regulates a wide range of physiologic processes in melanocyte homeostasis mainly by acting through the canonical PKA-CREB pathway. During melanoma initiation the system might switch and impaired cAMP signaling might sustain the tumor environment in a way that need to be explored deeply. However Rodriguez et al. showed that topical application of forskolin that directly activates AC, increases the level of cAMP, speeding melanoma tumor development in BRAFV600E/PTEN mouse model of melanoma and stimulating the proliferation of mouse and human primary melanoma cells in vitro. Although the process was cAMP-driven, an alternative downstream effector called Exchange Protein directly Activated by cAMP (EPAC) is involved. EPAC has been identified in 1998 and it acts as a guanine nucleotide exchange factor for the GTPase Ras family: RAP1 and RAP2 [83]. Modulating different signaling pathways, EPAC is involved in several cellular processes such as cell proliferation, migration, apoptosis and adhesion in various tissues [84]. In addition it has been shown that MC1R-cAMP-EPAC cascade promotes DNA repair by increasing the nuclear translocation of XPA protein in keratinocytes [57]. On the other hand, EPAC has shown to have a pro-metastatic role as it acts by activating ERK pathway and αvβ3 integrin through RAP1 thus promoting tumorigenesis and migration in human lung cancer cells but also by influencing other signalling cascades in cells derived from human metastatic melanomas, in human melanoma samples and melanoma cell lines [85,86,87]. The current hypothesis is that EPAC could have a different function during different stages of melanoma progression, with EPAC-RAP1 axis showing both a pro-survival role in primary melanoma and an anti-survival role in metastatic melanoma. Hence, it could be speculated that proliferation is inhibited during metastasis promoting an invasive phenotype [88, 89].

α-MSH-based strategies in melanoma treatment

The MC1R receptor is recognized to play a key role in melanocyte, melanosome, and melanoma cell (patho)physiology. Regarding metastasis, overexpression levels of MC1R, and MSH production by the neoplastic tissue itself are well-established data in the scientific literature. In this way, metastasis creates and self-maintains an autocrine loop that stimulates the growth, proliferation and invasiveness of the neoplasm, with the possibility of recurrence at metastatic sites, progression and dissemination, creating new metastatic sites and thus making the patient life-threatening.

This mechanism may also play a role in resistance to targeted therapy against mutated B-RAF (V600E B-RAF) where this phenomenon is described to be enhanced, suggesting that MC1R activation may contribute to the development of cancer resistance to dabrafenib. For these reasons, our group among others posits MC1R inhibition as a possible strategy to counteract this autocrine loop that intervenes in metastatic disease.

MC1R potentially constitutes an ideal target for design of novel anticancer drugs both for its involvement in melanocytic pathophysiology anf for its high levels of tissue-specific expression in melanoma cells.

At present, many works have shown promising results using the tissue specificity of MC1R for melanocytic tissues as an antitumor strategy.

Liu and collaborators reported that the immunotoxin α-MSH-PE38KDEL, constructed by connecting the α-MSH gene to PE38KDEL (a mutated and truncated form of a bacterial toxin), showed in vitro high cytotoxicity on MC1R positive melanoma cell lines, promoting apoptosis via Erk1/2/MITF/TYR signaling modulation in a MC1R-dependent manner [90]. They demonstrated that MC1R is essential for the immunotoxin-mediated cytotoxicity, promoting melanoma cell apoptosis inhibiting MITF and TYR expression. In fact, the overexpression of MITF or TYR abolishes α-MSH-PE38KDEL induction of apoptosis in mouse melanoma B16-F10 cell line. The authors demonstrated that the same pathway modulation significantly inhibited the in vivo tumor-forming ability of B16-F10 cells, when injected into athymic BALB-C nude mice.

Other works, using radionuclide-αMSH analogs conjugates, depicted interesting results in a theranostic settingThese studies, conducted in melanoma-bearing mouse models, demonstrated the high specificity of those molecules for MC1R, with a good bioavailability and renal clearance. In in vivo preclinical experimental animal model bearing mouse B16F1 or B16F10 melanoma radiolabelled peptides targeting MC1R, the radionuclide-αMSH analogs conjugates are able to selectively and specifically kill melanoma cells, sparing healthy cells and normal tissue. These studies are reviewed and summarised in two recent works [91, 92] Shi and collaborators considered studies about molecular probes for melanoma theranostics targeting either MC1R or melanine. These MC1R targeted radiotracers, displaying a good tumor uptake and retention, could potentially be used for inmaging of MC1R expressing melanoma in clinic. These imaging probes could be transformed into therapeutic radiopharmaceuticals through radiolabeling with beta- or alpha emitters. [91].

These novel sensitive and specific MC1R targeted radiotracers can overcome the actual limitation of (18)F FDG PET (I.e poor selectivity for distinguishing tumor from inflammatory tissue and low sensitivity in the detection of both nodal and lung and brain metastases) [92]. Furthermore, in a potential clinical application, cytotoxic radiation generated by therapeutic radionuclides could help treat remnant metastatic deposits, in an adjuvant setting, after surgical excision of the tumor.

Notheworthely, the group of Cachin reported the results of a multicenter phase III clinical trial [93]. This trial evaluated the accuracy of a new benzamide-derivative melanin targeted radiotracer, the (123)I-BZA2 radiopharmaceutical. This trial was prematurely closed after the enrollment of 87 patients, because of the low sensitivity of the radioconjugate in comparison to (18)F FDG when considering both a patient-based and a lesion-based analyses. However, (123)I-BZA2 demonstrated higher specificity than (18)F FDG for diagnosis of melanoma metastasis in a lesion-based analysis.

Further clinical studies are needed to validate the results of promising pre-clinical works.

Conclusions and perspectives

In this study we reviewed and summarised the molecular biology of α-MSH/MC1R, their range of action beyond pigmentation, the role of α-MSH/MC1R axis in melanoma and the MC1R targeting therapeutic strategies that have been proposed for melanoma.

α-MSH is the key hormone for melanocytic metabolism. It is not only the main actor of skin pigmentation but it displays also anti-inflammatory and anti-microbal properties. Among melanocortin receptors, melanocytes mainly express MC1R, whose binding with α-MSH promotes both the production of eumelanin, through the activation of AC/cAMP/PKA pathway, and melanocytic proliferation, survival and migration.

In summary, this review shows the ambivalence in the relationship between α-MSH and its membrane receptor. In physiological condition, the intracellular pathways elicited by this bond ensure skin pigmentation, DNA repair and anti-microbal and inflammatory defense. On the other hand, in pathological conditions, the overstimulation of the α-MSH/MC1R axis can lead to survival, uncontrolled proliferation, and invasion of cancer cells in metastatic melanoma. Moreover, some reports showed that synthetic alpha-MSH analogues (MC1R agonists) could lead to proliferation of melanocytic cells in predisposed patients, representing an increased risk for atypical naevi and melanoma development [94, 95]. Notheworthely, Kansal et al., reported that the inhibition of MC1R diminishes melanoma growth and increases survival of mice bearing melanoma [96]. Being overexpressed in metastatic melanoma, and particularly in targeted therapy-resistant clones, MC1R could represent a molecular target for metastatic melanoma and its inhibition a molecular strategy to delay resistance.

Availability of data and materials

Not applicable.

Abbreviations

AC:

Adenylyl Cyclase

ACTH:

Adrenocorticotropic Hormone

Agrp:

Agouti related protein

α-MSH:

Alpha-melanocyte stimulating hormone

ATM:

Ataxia Telangiectasia Mutated

BRAFi:

BRAF inhibitors

Ca2+ :

Calcium

cAMP:

Cyclic AMP

Dcs:

Dendritic cells

DCT:

Dopachrome Tautomerase

EPAC:

Exchange Protein directly Activated by cAMP

ER:

Endoplasmic reticulum

GoF:

Gain of Function

GPCR:

G-Protein Coupled Receptor

HSDD:

Hypoactive Sexual Desire Disorders

IFN-γ:

Interferon-gamma

MAPK:

Mitogen-Activated Protein Kinase

MC1R:

Melanocortin 1 receptor

MCRs:

Melanocortin Receptors

MiTF:

Microphthalmia inducing Transcription Factor

NER:

Nucleotide Excision Repair

NfκB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

PI3K:

Phosphatidylinositol 3-kinase

PIP2:

Phosphatidylinositol-4,5-bisphosphate

PIP3:

Phosphatidylinositol-3,4,5-triphosphate

PKA:

Protein Kinase A

PLC:

Phospholipase C

POMC:

Pro-opiomelanocortin hormone

PPAR-γ:

Peroxisome Proliferator-Activated Receptor-γ

PTEN:

Phosphatase and TENsin homolog

RHC:

Red-Hair-Colour

RTK:

Tyrosine Kinase Receptor

TYR:

Tyrosinase

TNF-α:

Tumor Necrosis Factor alpha

TRP:

Tyrosinase Related Protein

XPA:

Xeroderma Pigmentosum complementation group A

References

  1. Navarro S, Soletto L, Puchol S, Rotllant J, Soengas JL, Cerdá-Reverter JM. 60 years of POMC: POMC: an evolutionary perspective. J Mol Endocrinol. 2016;56(4):T113-118.

    Article  PubMed  CAS  Google Scholar 

  2. Schauer E, Trautinger F, Köck A, Schwarz A, Bhardwaj R, Simon M, et al. Proopiomelanocortin-derived peptides are synthesized and released by human keratinocytes. J Clin Invest. 1994;93(5):2258–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Chakraborty AK, Funasaka Y, Slominski A, Ermak G, Hwang J, Pawelek JM, et al. Production and release of proopiomelanocortin (POMC) derived peptides by human melanocytes and keratinocytes in culture: regulation by ultraviolet B. Biochim Biophys Acta. 1996;1313(2):130–8.

    Article  PubMed  Google Scholar 

  4. Tiwari K, Singh M, Kumar P, Mukhopadhyay K. Binding of cationic analogues of α-MSH to lipopolysaccharide and disruption of the cytoplasmic membranes caused bactericidal action against Escherichia coli. Sci Rep. 2022;12(1):1987.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Cutuli M, Cristiani S, Lipton JM, Catania A. Antimicrobial effects of alpha-MSH peptides. J Leukoc Biol. 2000;67(2):233–9.

    Article  PubMed  CAS  Google Scholar 

  6. Slominski RM, Sarna T, Płonka PM, Raman C, Brożyna AA, Slominski AT. Melanoma, melanin, and melanogenesis: the yin and yang relationship. Front Oncol. 2022;12: 842496.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Schiöth HB, Mutulis F, Muceniece R, Prusis P, Wikberg JE. Discovery of novel melanocortin4 receptor selective MSH analogues. Br J Pharmacol. 1998;124(1):75–82.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Cai M, Hruby VJ. The melanocortin receptor system: a target for multiple degenerative diseases. Curr Protein Pept Sci. 2016;17(5):488–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Mountjoy KG, Robbins LS, Mortrud MT, Cone RD. The cloning of a family of genes that encode the melanocortin receptors. Science. 1992;257(5074):1248–51.

    Article  PubMed  CAS  Google Scholar 

  10. Fridmanis D, Roga A, Klovins J. ACTH receptor (MC2R) specificity: what do we know about underlying molecular mechanisms? Front Endocrinol (Lausanne). 2017;8:13.

    Article  PubMed  Google Scholar 

  11. Guida S, Guida G, Goding CR. MC1R functions, expression, and implications for targeted therapy. J Invest Dermatol. 2022;142(2):293-302.e1.

    Article  PubMed  CAS  Google Scholar 

  12. D’Orazio JA, Nobuhisa T, Cui R, Arya M, Spry M, Wakamatsu K, et al. Topical drug rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning. Nature. 2006;443(7109):340–4.

    Article  PubMed  Google Scholar 

  13. Nasti TH, Timares L. MC1R, eumelanin and pheomelanin: their role in determining the susceptibility to skin cancer. Photochem Photobiol. 2015;91(1):188–200.

    Article  PubMed  CAS  Google Scholar 

  14. Herraiz C, Garcia-Borron JC, Jiménez-Cervantes C, Olivares C. MC1R signaling Intracellular partners and pathophysiological implications. Biochim Biophys Acta Mol Basis Dis. 2017;186(10):2448–61.

    Article  Google Scholar 

  15. Peng L, Chang J, Liu X, Lu S, Ren H, Zhou X, et al. MC1R is a prognostic marker and its expression is correlated with msi in colorectal cancer. Curr Issues Mol Biol. 2021;43(3):1529–47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott T. UV radiation and the skin. Int J Mol Sci. 2013;14(6):12222–48.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Lu D, Willard D, Patel IR, Kadwell S, Overton L, Kost T, et al. Agouti protein is an antagonist of the melanocyte-stimulating-hormone receptor. Nature. 1994;371(6500):799–802.

    Article  PubMed  CAS  Google Scholar 

  18. Li WD, Joo EJ, Furlong EB, Galvin M, Abel K, Bell CJ, et al. Melanocortin 3 receptor (MC3R) gene variants in extremely obese women. Int J Obes Relat Metab Disord. 2000;24(2):206–10.

    Article  PubMed  CAS  Google Scholar 

  19. Tao YX. Mutations in the melanocortin-3 receptor (MC3R) gene: impact on human obesity or adiposity. Curr Opin Investig Drugs. 2010;11(10):1092–6.

    PubMed  CAS  Google Scholar 

  20. Sweeney P, Bedenbaugh MN, Maldonado J, Pan P, Fowler K, Williams SY, et al. The melanocortin-3 receptor is a pharmacological target for the regulation of anorexia. Sci Transl Med. 2021;13(590):eabd6434.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Ericson MD, Doering SR, Larson CM, Freeman KT, LaVoi TM, Donow HM, et al. Functional mixture-based positional scan identifies a library of antagonist tetrapeptide sequences (LAtTeS) with nanomolar potency for the melanocortin-4 receptor and equipotent with the endogenous AGRP(86–132) antagonist. J Med Chem. 2021;64(19):14860–75.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Yu K, Li L, Zhang L, Guo L, Wang C. Association between MC4R rs17782313 genotype and obesity: a meta-analysis. Gene. 2020;5(733): 144372.

    Article  Google Scholar 

  23. Springer MS, Gatesy J. Evolution of the MC5R gene in placental mammals with evidence for its inactivation in multiple lineages that lack sebaceous glands. Mol Phylogenet Evol. 2018;120:364–74.

    Article  PubMed  CAS  Google Scholar 

  24. Xu Y, Guan X, Zhou R, Gong R. Melanocortin 5 receptor signaling pathway in health and disease. Cell Mol Life Sci. 2020;77(19):3831–40.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. García-Borrón JC, Abdel-Malek Z, Jiménez-Cervantes C. MC1R, the cAMP pathway, and the response to solar UV: extending the horizon beyond pigmentation. Pigment Cell Melanoma Res. 2014;27(5):699–720.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Rodríguez CI, Setaluri V. Cyclic AMP (cAMP) signaling in melanocytes and melanoma. Arch Biochem Biophys. 2014;1(563):22–7.

    Article  Google Scholar 

  27. Goding CR, Arnheiter H. MITF-the first 25 years. Genes Dev. 2019;33(15–16):983–1007.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Makova K, Norton H. Worldwide polymorphism at the MC1R locus and normal pigmentation variation in humans. Peptides. 2005;26(10):1901–8.

    Article  PubMed  CAS  Google Scholar 

  29. Branicki W, Brudnik U, Kupiec T, Wolañska-Nowak P, Wojas-Pelc A. Determination of phenotype associated SNPs in the MC1R gene. J Forensic Sci. 2007;52(2):349–54.

    Article  PubMed  CAS  Google Scholar 

  30. Mitra D, Luo X, Morgan A, Wang J, Hoang MP, Lo J, et al. An ultraviolet-radiation-independent pathway to melanoma carcinogenesis in the red hair/fair skin background. Nature. 2012;491(7424):449–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Katz M, Amit I, Yarden Y. Regulation of MAPKs by growth factors and receptor tyrosine kinases. Biochim Biophys Acta. 2007;1773(8):1161–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Colombino M, Capone M, Lissia A, Cossu A, Rubino C, De Giorgi V, et al. BRAF/NRAS mutation frequencies among primary tumors and metastases in patients with melanoma. J Clin Oncol. 2012;30(20):2522–9.

    Article  PubMed  Google Scholar 

  33. Dhomen N, Marais R. BRAF signaling and targeted therapies in melanoma. Hematol Oncol Clin North Am. 2009;23(3):529–45, ix.

    Article  PubMed  Google Scholar 

  34. Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP, et al. A landscape of driver mutations in melanoma. Cell. 2012;150(2):251–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Krauthammer M, Kong Y, Ha BH, Evans P, Bacchiocchi A, McCusker JP, et al. Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Nat Genet. 2012;44(9):1006–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Murphy LO, Blenis J. MAPK signal specificity: the right place at the right time. Trends Biochem Sci. 2006;31(5):268–75.

    Article  PubMed  CAS  Google Scholar 

  37. Hemesath TJ, Price ER, Takemoto C, Badalian T, Fisher DE. MAP kinase links the transcription factor Microphthalmia to c-Kit signalling in melanocytes. Nature. 1998;391(6664):298–301.

    Article  PubMed  CAS  Google Scholar 

  38. Herraiz C, Journé F, Abdel-Malek Z, Ghanem G, Jiménez-Cervantes C, García-Borrón JC. Signaling from the human melanocortin 1 receptor to ERK1 and ERK2 mitogen-activated protein kinases involves transactivation of cKIT. Mol Endocrinol. 2011;25(1):138–56.

    Article  PubMed  CAS  Google Scholar 

  39. Herraiz C, Sánchez-Laorden BL, Jiménez-Cervantes C, García-Borrón JC. N-glycosylation of the human melanocortin 1 receptor: occupancy of glycosylation sequons and functional role. Pigment Cell Melanoma Res. 2011;24(3):479–89.

    Article  PubMed  CAS  Google Scholar 

  40. Vanhaesebroeck B, Whitehead MA, Piñeiro R. Molecules in medicine mini-review: isoforms of PI3K in biology and disease. J Mol Med (Berl). 2016;94(1):5–11.

    Article  PubMed  CAS  Google Scholar 

  41. Cao J, Wan L, Hacker E, Dai X, Lenna S, Jimenez-Cervantes C, et al. MC1R is a potent regulator of PTEN after UV exposure in melanocytes. Mol Cell. 2013;51(4):409–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Castejón-Griñán M, Herraiz C, Olivares C, Jiménez-Cervantes C, García-Borrón JC. cAMP-independent non-pigmentary actions of variant melanocortin 1 receptor: AKT-mediated activation of protective responses to oxidative DNA damage. Oncogene. 2018;37(27):3631–46.

    Article  PubMed  Google Scholar 

  43. Herraiz C, Martínez-Vicente I, Maresca V. The α-melanocyte-stimulating hormone/melanocortin-1 receptor interaction: a driver of pleiotropic effects beyond pigmentation. Pigment Cell Melanoma Res. 2021;34(4):748–61.

    Article  PubMed  CAS  Google Scholar 

  44. Maresca V, Flori E, Camera E, Bellei B, Aspite N, Ludovici M, et al. Linking αMSH with PPARγ in B16–F10 melanoma. Pigment Cell Melanoma Res. 2013;26(1):113–27.

    Article  PubMed  CAS  Google Scholar 

  45. Fanale D, Amodeo V, Caruso S. The interplay between metabolism, PPAR signaling pathway, and cancer. PPAR Res. 2017;2017:1830626.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Deraredj Nadim W, Hassanaly S, Bénédetti H, Kieda C, Grillon C, Morisset-Lopez S. The GTPase-activating protein-related domain of neurofibromin interacts with MC1R and regulates pigmentation-mediated signaling in human melanocytes. Biochem Biophys Res Commun. 2021;1(534):758–64.

    Article  Google Scholar 

  47. Calabrese G, Peker E, Amponsah PS, Hoehne MN, Riemer T, Mai M, et al. Hyperoxidation of mitochondrial peroxiredoxin limits H2 O2 -induced cell death in yeast. EMBO J. 2019;38(18): e101552.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Song X, Mosby N, Yang J, Xu A, Abdel-Malek Z, Kadekaro AL. alpha-MSH activates immediate defense responses to UV-induced oxidative stress in human melanocytes. Pigment Cell Melanoma Res. 2009;22(6):809–18.

    Article  PubMed  CAS  Google Scholar 

  49. Kadekaro AL, Leachman S, Kavanagh RJ, Swope V, Cassidy P, Supp D, et al. Melanocortin 1 receptor genotype: an important determinant of the damage response of melanocytes to ultraviolet radiation. FASEB J. 2010;24(10):3850–60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Maresca V, Flori E, Bellei B, Aspite N, Kovacs D, Picardo M. MC1R stimulation by alpha-MSH induces catalase and promotes its re-distribution to the cell periphery and dendrites. Pigment Cell Melanoma Res. 2010;23(2):263–75.

    Article  PubMed  CAS  Google Scholar 

  51. Tagliabue E, Fargnoli MC, Gandini S, Maisonneuve P, Liu F, Kayser M, et al. MC1R gene variants and non-melanoma skin cancer: a pooled-analysis from the M-SKIP project. Br J Cancer. 2015;113(2):354–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Valverde P, Healy E, Jackson I, Rees JL, Thody AJ. Variants of the melanocyte-stimulating hormone receptor gene are associated with red hair and fair skin in humans. Nat Genet. 1995;11(3):328–30.

    Article  PubMed  CAS  Google Scholar 

  53. Kennedy C, ter Huurne J, Berkhout M, Gruis N, Bastiaens M, Bergman W, et al. Melanocortin 1 receptor (MC1R) gene variants are associated with an increased risk for cutaneous melanoma which is largely independent of skin type and hair color. J Invest Dermatol. 2001;117(2):294–300.

    Article  PubMed  CAS  Google Scholar 

  54. Palmer JS, Duffy DL, Box NF, Aitken JF, O’Gorman LE, Green AC, et al. Melanocortin-1 receptor polymorphisms and risk of melanoma: is the association explained solely by pigmentation phenotype? Am J Hum Genet. 2000;66(1):176–86.

    Article  PubMed  CAS  Google Scholar 

  55. Sánchez Más J, Olivares Sánchez C, Ghanem G, Haycock J, Lozano Teruel JA, García-Borrón JC, et al. Loss-of-function variants of the human melanocortin-1 receptor gene in melanoma cells define structural determinants of receptor function. Eur J Biochem. 2002;269(24):6133–41.

    Article  PubMed  Google Scholar 

  56. Sturm RA, Duffy DL, Box NF, Newton RA, Shepherd AG, Chen W, et al. Genetic association and cellular function of MC1R variant alleles in human pigmentation. Ann NY Acad Sci. 2003;994:348–58.

    Article  PubMed  CAS  Google Scholar 

  57. Dong L, Wen J, Pier E, Zhang X, Zhang B, Dong F, et al. Melanocyte-stimulating hormone directly enhances UV-Induced DNA repair in keratinocytes by a xeroderma pigmentosum group A-dependent mechanism. Cancer Res. 2010;70(9):3547–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Manganelli M, Guida S, Ferretta A, Pellacani G, Porcelli L, Azzariti A, Guida G. Behind the scene: exploiting MC1R in skin cancer risk and prevention. Genes (Basel). 2021;12(7):1093.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Lipton JM, Catania A. Anti-inflammatory actions of the neuroimmunomodulator alpha-MSH. Immunol Today. 1997;18(3):140–5.

    Article  PubMed  CAS  Google Scholar 

  60. Poźniak J, Nsengimana J, Laye JP, O’Shea SJ, Diaz JMS, Droop AP, et al. Genetic and environmental determinants of immune response to cutaneous melanoma. Cancer Res. 2019;79(10):2684–96.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Luger TA, Scholzen TE, Brzoska T, Böhm M. New insights into the functions of alpha-MSH and related peptides in the immune system. Ann NY Acad Sci. 2003;994:133–40.

    Article  PubMed  CAS  Google Scholar 

  62. Luger TA, Brzoska T. alpha-MSH related peptides: a new class of anti-inflammatory and immunomodulating drugs. Ann Rheum Dis. 2007;66(3):iii52-55.

    PubMed  PubMed Central  CAS  Google Scholar 

  63. Singh M, Mukhopadhyay K. Alpha-melanocyte stimulating hormone: an emerging anti-inflammatory antimicrobial peptide. Biomed Res Int. 2014;2014: 874610.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Catania A, Colombo G, Rossi C, Carlin A, Sordi A, Lonati C, et al. Antimicrobial properties of alpha-MSH and related synthetic melanocortins. ScientificWorldJournal. 2006;2(6):1241–6.

    Article  Google Scholar 

  65. Eves P, Haycock J, Layton C, Wagner M, Kemp H, Szabo M, et al. Anti-inflammatory and anti-invasive effects of alpha-melanocyte-stimulating hormone in human melanoma cells. Br J Cancer. 2003;89(10):2004–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Rachmin I, Ostrowski SM, Weng QY, Fisher DE. Topical treatment strategies to manipulate human skin pigmentation. Adv Drug Deliv Rev. 2020;1(153):65–71.

    Article  Google Scholar 

  67. Simon JA, Kingsberg SA, Portman D, Jordan R, Lucas J, Sadiq A, et al. Prespecified and integrated subgroup analyses from the RECONNECT phase 3 studies of bremelanotide. J Womens Health (Larchmt). 2022;31(3):391–400.

    Article  PubMed  Google Scholar 

  68. Guo X, Yuan J, Li M, Wang M, Lv P. Neuroprotection of intermedin against cerebral ischemia/reperfusion injury through cerebral microcirculation improvement and apoptosis inhibition. J Mol Neurosci. 2021;71(4):767–77.

    Article  PubMed  CAS  Google Scholar 

  69. Weng WT, Wu CS, Wang FS, Wu CY, Ma YL, Chan HH, et al. α-melanocyte-stimulating hormone attenuates neovascularization by inducing nitric oxide deficiency via MC-Rs/PKA/NF-κB signaling. Int J Mol Sci. 2018;19(12):E3823.

    Article  Google Scholar 

  70. Kleinau G, Heyder NA, Tao YX, Scheerer P. Structural complexity and plasticity of signaling regulation at the melanocortin-4 receptor. Int J Mol Sci. 2020;21(16):5728.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Koo BB, Feng P, Dostal J, Strohl KP. Alpha-melanocyte stimulating hormone and adrenocorticotropic hormone: an alternative approach when thinking about restless legs syndrome? Mov Disord. 2008;23(9):1234–42.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Costa JL, Hochgeschwender U, Brennan M. The role of melanocyte-stimulating hormone in insulin resistance and type 2 diabetes mellitus. Treat Endocrinol. 2006;5(1):7–13.

    Article  PubMed  CAS  Google Scholar 

  73. Goit RK, Taylor AW, Lo ACY. The central melanocortin system as a treatment target for obesity and diabetes: a brief overview. Eur J Pharmacol. 2022;5(924): 174956.

    Article  Google Scholar 

  74. Owens B. Melanoma. Nature. 2014;515(7527):S109.

    Article  PubMed  Google Scholar 

  75. Rastrelli M, Tropea S, Rossi CR, Alaibac M. Melanoma: epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo. 2014;28(6):1005–11.

    PubMed  Google Scholar 

  76. Echevarría-Vargas IM, Villanueva J. COMBATING NRAS MUTANT MELANOMA: FROM BENCH TO BEDSIDE. Melanoma Manag. 2017;4(4):183–6.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Amann VC, Ramelyte E, Thurneysen S, Pitocco R, Bentele-Jaberg N, Goldinger SM, et al. Developments in targeted therapy in melanoma. Eur J Surg Oncol. 2017;43(3):581–93.

    Article  PubMed  CAS  Google Scholar 

  78. Bell RE, Levy C. The three M’s: melanoma, microphthalmia-associated transcription factor and microRNA. Pigment Cell Melanoma Res. 2011;24(6):1088–106.

    Article  PubMed  CAS  Google Scholar 

  79. Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S, et al. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 2005;65(20):9328–37.

    Article  PubMed  CAS  Google Scholar 

  80. Salazar-Onfray F, López M, Lundqvist A, Aguirre A, Escobar A, Serrano A, et al. Tissue distribution and differential expression of melanocortin 1 receptor, a malignant melanoma marker. Br J Cancer. 2002;87(4):414–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Rosenkranz AA, Slastnikova TA, Durymanov MO, Sobolev AS. Malignant melanoma and melanocortin 1 receptor. Biochemistry (Mosc). 2013;78(11):1228–37.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Loir B, Bouchard B, Morandini R, Del Marmol V, Deraemaecker R, Garcia-Borron JC, et al. Immunoreactive alpha-melanotropin as an autocrine effector in human melanoma cells. Eur J Biochem. 1997;244(3):923–30.

    Article  PubMed  CAS  Google Scholar 

  83. de Rooij J, Zwartkruis FJ, Verheijen MH, Cool RH, Nijman SM, Wittinghofer A, et al. Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP. Nature. 1998;396(6710):474–7.

    Article  PubMed  Google Scholar 

  84. de Rooij J, Rehmann H, van Triest M, Cool RH, Wittinghofer A, Bos JL. Mechanism of regulation of the Epac family of cAMP-dependent RapGEFs. J Biol Chem. 2000;275(27):20829–36.

    Article  PubMed  Google Scholar 

  85. Lim JA, Juhnn YS. Isoproterenol increases histone deacetylase 6 expression and cell migration by inhibiting ERK signaling via PKA and Epac pathways in human lung cancer cells. Exp Mol Med. 2016;48(1): e204.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Gao L, Feng Y, Bowers R, Becker-Hapak M, Gardner J, Council L, et al. Ras-associated protein-1 regulates extracellular signal-regulated kinase activation and migration in melanoma cells: two processes important to melanoma tumorigenesis and metastasis. Cancer Res. 2006;66(16):7880–8.

    Article  PubMed  CAS  Google Scholar 

  87. Baljinnyam E, Umemura M, De Lorenzo MS, Iwatsubo M, Chen S, Goydos JS, et al. Epac1 promotes melanoma metastasis via modification of heparan sulfate. Pigment Cell Melanoma Res. 2011;24(4):680–7.

    Article  PubMed  CAS  Google Scholar 

  88. Rodriguez CI, Setaluri V. EPAC mediates the dual role of cAMP signaling in melanoma. Oncoscience. 2019;6(1–2):283–4.

    PubMed  Google Scholar 

  89. Rodríguez CI, Castro-Pérez E, Prabhakar K, Block L, Longley BJ, Wisinski JA, et al. EPAC-RAP1 axis-mediated switch in the response of primary and metastatic melanoma to cyclic AMP. Mol Cancer Res. 2017;15(12):1792–802.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Liu X, Li H, Cong X, Huo D, Cong L, Wu G. α-MSH-PE38KDEL kills melanoma cells via modulating Erk1/2/MITF/TYR signaling in an MC1R-dependent manner. Onco Targets Ther. 2020;13:12457–69.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Shi H, Cheng Z. MC1R and melanin-based molecular probes for theranostic of melanoma and beyond. Acta Pharmacol Sin. 2022;43(12):3034–44.

    Article  PubMed  CAS  Google Scholar 

  92. Wei W, Ehlerding EB, Lan X, Luo Q, Cai W. PET and SPECT imaging of melanoma: the state of the art. Eur J Nucl Med Mol Imaging. 2018;45(1):132–50.

    Article  PubMed  CAS  Google Scholar 

  93. Cachin F, Miot-Noirault E, Gillet B, Isnardi V, Labeille B, Payoux P, Meyer N, Cammilleri S, Gaudy C, Razzouk-Cadet M, Lacour JP, Granel-Brocard F, Tychyj C, Benbouzid F, Grange JD, Baulieu F, Kelly A, Merlin C, Mestas D, Gachon F, Chezal JM, Degoul F, D’Incan M. (123)I-BZA2 as a melanin-targeted radiotracer for the identification of melanoma metastases: results and perspectives of a multicenter phase III clinical trial. J Nucl Med. 2014;55(1):15–22. https://doi.org/10.2967/jnumed.113.123554.

    Article  PubMed  CAS  Google Scholar 

  94. Habbema L, Halk AB, Neumann M, Bergman W. Risks of unregulated use of alpha-melanocyte-stimulating hormone analogues: a review. Int J Dermatol. 2017;56(10):975–80.

    Article  PubMed  Google Scholar 

  95. Ong S, Bowling J. Melanotan-associated melanoma in situ. Australas J Dermatol. 2012;53(4):301–2.

    Article  PubMed  Google Scholar 

  96. Kansal RG, McCravy MS, Basham JH, Earl JA, McMurray SL, Starner CJ, et al. Inhibition of melanocortin 1 receptor slows melanoma growth, reduces tumor heterogeneity and increases survival. Oncotarget. 2016;7(18):26331–45.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank Associazione Piccoli Punti and Mr Nicolò Socal for their support during the preparation and the revision of the manuscript.

Funding

Open access funding provided by Università degli Studi di Padova within the CRUI-CARE Agreement. The fellowship of NP was supported by the “5 × 1000” IOV grant.

Author information

Authors and Affiliations

Authors

Contributions

LDO and NP were responsible for conceiving the ideas. All authors wrote different parts of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Luigi Dall’Olmo.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dall’Olmo, L., Papa, N., Surdo, N.C. et al. Alpha-melanocyte stimulating hormone (α-MSH): biology, clinical relevance and implication in melanoma. J Transl Med 21, 562 (2023). https://doi.org/10.1186/s12967-023-04405-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-023-04405-y

Keywords