Next Article in Journal
Atypical Endometrial Hyperplasia and Concurrent Cancer: A Comprehensive Overview on a Challenging Clinical Condition
Previous Article in Journal
Pathogenesis of Warthin’s Tumor: Neoplastic or Non-Neoplastic?
Previous Article in Special Issue
The Effect of Non-Overlapping Somatic Mutations in BRAF, NRAS, NF1, or CKIT on the Incidence and Outcome of Brain Metastases during Immune Checkpoint Inhibitor Therapy of Metastatic Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Keratinocyte in the Picture Cutaneous Melanoma Microenvironment

by
Ramona Marrapodi
and
Barbara Bellei
*
Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144 Rome, Italy
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(5), 913; https://doi.org/10.3390/cancers16050913
Submission received: 22 January 2024 / Revised: 8 February 2024 / Accepted: 20 February 2024 / Published: 23 February 2024

Abstract

:

Simple Summary

The tumor environment is the place where the tumor resides implying that it might evolve in the role of disease progression, particularly in the phase of tumor dissemination. For cutaneous melanoma, in the early phase of disease advancement, the microenvironment is confined to the epidermal compartment since the dermo-epidermal basement membrane prevents dermal invasion. Here, poorly aggressive melanoma cells gain the aggressiveness necessary for disease progression. After dermo-epidermal membrane breakdown, the melanoma cells progressively lose keratinocyte control and engage in unusual partnerships mainly with dermal fibroblasts.

Abstract

Melanoma progression is a multistep evolution from a common melanocytic nevus through a radial superficial growth phase, the invasive vertical growth phase finally leading to metastatic dissemination into distant organs. Melanoma aggressiveness largely depends on the propensity to metastasize, which means the capacity to escape from the physiological microenvironment since tissue damage due to primary melanoma lesions is generally modest. Physiologically, epidermal melanocytes are attached to the basement membrane, and their adhesion/migration is under the control of surrounding keratinocytes. Thus, the epidermal compartment represents the first microenvironment responsible for melanoma spread. This complex process involves cell–cell contact and a broad range of secreted bioactive molecules. Invasion, or at the beginning of the microinvasion, implies the breakdown of the dermo-epidermal basement membrane followed by the migration of neoplastic melanocytic cells in the superficial papillary dermis. Correspondingly, several experimental evidences documented the structural and functional rearrangement of the entire tissue surrounding neoplasm that in some way reflects the atypia of tumor cells. Lastly, the microenvironment must support the proliferation and survival of melanocytes outside the normal epidermal–melanin units. This task presumably is mostly delegated to fibroblasts and ultimately to the self-autonomous capacity of melanoma cells. This review will discuss remodeling that occurs in the epidermis during melanoma formation as well as skin changes that occur independently of melanocytic hyperproliferation having possible pro-tumoral features.

1. Introduction

Melanoma is a malignant tumor, caused by the transformation of melanocytes responsible for melanin production [1]. Melanoma represents only 1% of skin cancers but it accounts for most deaths due to cutaneous neoplasms [2]. Its incidence has increased in the last decade especially in the Caucasian population since it affects fair-skinned people with blond, red hair and light-colored eyes more frequently [3,4]. Furthermore, according to the Global Cancer Observatory, there were approximately 325,000 new cases of melanoma worldwide in 2020, of which about 150,000 were diagnosed in Europe [5]. The overall incidence is slightly higher in men than in women with Age Standardized Incidence Rates of 3.8/100,000 and 3.0/100,000 respectively [5]. The average age of melanoma diagnosis is 57 years. Between the ages of 25 and 40, the incidence is higher among women, but after 75 years of age, the incidence for men is 3 times higher than for women [6]. Although melanoma is considered a multifactorial disease, prolonged and unprotected natural sunlight exposure or indoor UV lamps are recognized as the main causes of disease onset [7,8]. On the Earth’s surface, UV rays are composed of UVA and UVB since UVC is absorbed by the ozone layer. UVB is the main cause of sunburn. UVB, having a shortened wavelength, directly causes DNA damage, while UVA, having a longer wavelength, acts indirectly by inducing the generation of reactive oxygen species (ROS) responsible for lipid peroxidation, protein carbonylation, and DNA lesions [9,10,11]. The consequence of intracellular signaling alteration contributes to dermis remodeling, inflammation, hyperpigmentation, and acceleration of the aging process [10]. UV-caused damage is cumulative and results in a collection of random unrepaired mutations that when involved in genes implicated in cell cycle regulation might cause hyperproliferation of skin cells. Indeed, in line with the general notion that old organisms are more sensitive to stress, the capacity to repair DNA defects is subject to age-dependent decline [12]. Accordingly, the number of somatic mutations found in normal human tissue as well as in tumors increases with age [13]. On the other hand, the accumulation of mutations may contribute to aging and apoptosis perturbing the physiological balance existing between cell death and cell renewal and exacerbating tissue functional decline [14]. Several studies evidenced that a family history of skin cancers is strongly associated with an increased risk of melanoma in both sexes [15]. A history of non-melanoma skin cancer increases the risk of melanoma and also in the skin of color subjects having a generally lower incidence of skin cancer [16]. A number of phenotypic traits connected to pigmentation portend increased risk for melanoma. People with fairer skin phenotypes (pale white skin, light-colored eyes, low tanning ability, burning propensity, high freckling tendency) are more likely to develop cutaneous melanoma [9,17] as well as people habitually exposed to the sun for prolonged periods [18]. Family history is one of the most significant risk factors for melanoma [9]. Approximately 8–12% of melanoma cases occur in multiple-case families and can be inherited in an autosomal dominant fashion [19]. Interestingly, risk based on family history depends not only on the number of individuals in the family who experience melanoma but also on the number of melanomas in each family member [20]. Germline mutations of CDKN2A, CDK4, TERT, and POT1 genes have been identified as high-risk factors for melanoma susceptibility [21,22,23], whereas MC1R (melanocortin 1 receptor) polymorphic variants are reported in 60.5–82.1% of melanoma [24,25]. Currently, technological advances have allowed the identification of additional genes involved in melanoma susceptibility: BRCA2, BAP1, and MITF [26]. Also, pathogenic variants in albinism genes may account for a minor part of familiar melanoma [27]. More recently, CDH23, ARHGEF40, BRD9 [28], and CASP8 [29] have been proposed as candidate susceptibility genes in hereditary melanoma [30,31]. However, in general, it is important to take into consideration that inhered cancer-associated gene variants might influence the biology of all cell types including those of the tumor microenvironment augmenting or reducing the personal oncologic risk. Common variants of the MC1R gene play a relevant role also in sporadic melanoma onset [32,33]. Engagement of the MC1R expressed on the cell membrane by agonists activates the cAMP pathway leading to pro-melanogenic gene transcription and enhancement of DNA repair and antioxidant capacity [34]. This is explained by the evidence that several MC1R polymorphisms are associated with light-skin phenotype and attenuated hormonal stimulation of the melanogenic biosynthetic pathway [35]. Consistently, evolutionary constraints have determined the geographical distribution of undermelanized variants. MC1R nonsynonymous variants are largely present in Asian and European populations but not in African ones [36,37]. However, increased melanoma risk for individuals carrying polymorphic forms of MC1R is not restricted to cAMP signaling and resultant weakened pigment-dependent function (prevalent synthesis of reddish pheomelanin having photosensitizer properties and insufficient UV-ray absorption), but is additionally attributed to reduced protection against oxidative damage, attenuated MAPKs (mitogen-activated protein kinases), AKT (also known as protein kinase B, PKB) and PI3K (phosphoinositide 3-kinase) signaling activation [38,39,40,41,42], immune responses, glucose metabolism, and energy homeostasis modulation [43]. The increased risk associated with MC1R variants spans all histological subtypes and it occurs on both chronically and intermittently sun-exposed skin [44]. MC1R genetic variants are determinant also in the acquisition of skin aging-related features [45,46] including specific dermal variation during photoaging [47,48]. Since properties referred to functional aging of the skin have been associated with the potential pro-tumorigenic phenotype of fibroblast [49,50], impaired functionality of the (α-melanocyte stimulating hormone) α-MSH/MC1R axis might contribute to creating a favorable milieu for hyperproliferative diseases in photo-exposed bodies’ area. Notably, in the skin, the features of senescent fibroblast largely overlap with those of fibroblast associated with neoplastic cells [51].
MC1R signaling favors tolerance to environmental stressors by increasing the capacity to repair DNA damage for genomic maintenance and cell survival [40,52,53]. Consequently, a higher somatic mutation burden has been demonstrated in melanoma from subjects with MC1R variants compared to individuals with the wild-type sequence [54]. Accordingly, independent of skin type and hair color, some genetic variants of MC1R confer susceptibility also to non-melanoma (basal cell carcinoma, BCC, and squamous cell carcinoma, SCC) skin cancer [32,55]. MC1R activation lessens T-cell infiltration in the tumor microenvironment and causes a melanoma-specific mechanism of checkpoint blockade therapy resistance and immune evasion [56]. Moreover, MC1R variants correlate with poor response to BRAF inhibitors [57]. As part of the “qualitative” consideration concerning non-synonymous genomic variants, very recently, high MC1R levels of expression in primary and metastatic melanoma have been associated with poor prognosis [58]. Inverting the viewpoint, paradoxically a possible explanation for this observation might reside in the overexpression of DNA repair and anti-apoptosis pathways that support the persistence of highly injured cells [59].
Concerning the identification of melanoma risk factors, awareness is also addressed to melanocytic nevi that represent a benign lesion resulting from an abnormal proliferation of melanocytes. Since nearly 25% of melanomas appear in pre-existing nevi [60,61], an elevated number of nevi and the presence of atypical nevi constitute an important risk factor for the onset of melanoma [62]. Melanocytic nevi are precancerous lesions resulting from a focal and limited proliferation of melanocytes driven by the mutational activation of an oncogene such as BRAF [63]. After forming a nevus, nevomelanocytes undergo an enduring exit from the cell cycle that prevents them from progressing to melanoma. Melanoma can also originate from lentigo maligna, a slowly growing lesion in situ, which generally arises in the face of elderly people with photodamaged skin [62]. It has been estimated that 2–5% of lentigo maligna progress to invasive melanoma [64] and that lentigo maligna cover about 10% of all melanomas [65]. Desmoplastic melanoma partially overlaps with lentigo maligna due to the common link to sun-damaged skin but presents a distinct genetic profile [66].
In the context of cancer, the tissue surrounding pathologic cells acquired more and more interest.
During its evolution, melanoma progressively shapes host tissue into a favorable milieu promoting cancer dissemination, resistance to therapy, and immune evasion [37,67,68]. However, some inherited or acquired characteristics including chronic inflammation, fibrosis, and immunosuppression might predispose to hyperproliferative diseases [69,70]. Current studies have highlighted the role of aging in shaping the skin into a pro-tumorigenic condition [51,71,72,73]. Correspondingly, the incidence of tumors positively correlates with age [74] and progeroid syndrome, a heritable autosomal recessive human disorder characterized by the premature onset of numerous age-related diseases, predisposing to cancers [75]. Properly, skin cancers are the most common malignancies affecting older adults [76]. While in normal aging, the acquisition of cell-senescent phenotype and functional decline over time acts synchronously in various tissues, premature senescence may be limited to one or a few organs such as the skin since it is continuously exposed to external aggression. Extrinsic aging is superimposed on intrinsic aging resulting in a more intense rate of senescent cell accumulation than in normal aging. In neoplasms, crosstalk between melanoma and dermal fibroblasts also referred to as cancer-associated fibroblasts (CAFs) or more precisely melanoma-associated fibroblasts (MAFs) plays a relevant role in tumor growth, invasive abilities, and resistance to therapy [37,77,78]. Other cutaneous cells are involved in melanocyte transformation into melanoma and disease progression. Among these, keratinocytes forming the multilayer outer skin compartment, being in direct contact with neoplastic cells in the melanoma niche are deeply implicated in melanomagenesis, especially during early phases. However, compared to other components of the melanoma stroma, including mesenchymal and immune cells (natural killer cells, T lymphocytes, B-lymphocytes, dendritic cells, macrophages, and adipocytes), keratinocytes have been less investigated yet. This review will present the current knowledge concerning the direct and paracrine communication between keratinocytes and melanocytes in melanomagenesis.

2. Material and Methods

This manuscript considered data from reviews and experimental in vitro as well as in vivo studies with the thematic of melanoma and keratinocyte (epidermis) with a specific emphasis on articles comprising both topics. This analysis covers a multitude of recent and current peer-reviewed manuscripts without any restriction related to the publication period. Since this review has been designed as a narrative review, it synthesizes without systematic methods.

3. Skin Physiology

3.1. Skin Structure

Human skin covering the entire body acts as a barrier preventing the loss of water and protecting it from physical, chemical, and microbial damage. It is structured in three layers: epidermis, dermis, and hypodermis. The epidermis is mainly composed of keratinocytes, sparse immune cells, and melanocytes allocated at the base of the epidermis (Figure 1A).
The dermis is the middle layer of the skin. It confers flexibility and strength to the cutis due to the abundance of well-organized extracellular matrix proteins (collagen and elastin, glycoproteins, and glycosaminoglycans) [79,80]. Even if cellularity is modest in the dermal compartment, it hosts the highest functional and morphological cell diversity [81]. In addition to sparse fibroblasts, the most abundant cell type, the dermis, contains mesenchymal stem cells, lymphocytes, natural killer cells, Langerhans cells, Merkel cells, macrophages, mast cells, and dendritic cells [82]. The dermis also encloses blood and lymph vessels, sweat and sebaceous glands, hair follicles, nerves, and a variety of sensory nerve receptors. The subcutaneous fat layer (or hypodermis) is predominantly composed of adipocytes, pre-adipocytes, and mesenchymal stem cells but also of fibroblasts, pericytes, macrophages, T cells, and erythrocytes in a stromal network of collagen, vascular elements, nerve endings, and muscles [83]. The primary function of the hypodermis is to regulate the body’s temperature, and storage energy, and protect from injury underlying structures such as muscle and bones. Recent work has demonstrated a remarkable plasticity of adipocyte-lineage cells (stem cells and mature cells) during tissue repair indicating a central role of these cells in normal and pathological tissue remodeling. Further, due to the dynamic nature of adipocytes as they deliver bioactive products, lipids, and adipokines, hypodermic fat is emerging as an essential regulator of several physiological skin processes such as wound healing, pigmentation, hair growth, and innate immune response [84,85]. More, some evidence argues for the role of subcutaneous adipocytes in skin tumorigenesis [86,87]. Adipocyte-derived lipids represent a potent energy source capable of increasing melanoma proliferation and invasion in vivo and in vitro models [86,88]. Transferring high amounts of fatty acids, adipocytes alter the mitochondria metabolism of adjacent cells rendering melanoma cells less dependent on de novo intrinsic lipogenesis and accelerating melanoma development in the zebrafish model [88]. Since to be in direct contact with adipocytes, melanoma cells require a breakdown of the dermal-epidermal junction of the skin and the gain of the vertical growth phase, adipocytes are likely important for the metastatic process in melanoma. Lastly, adipocytes are a local source of CAF [89] since this type of cell might phenotypically change to generate fibroblast-like cells, termed adipose-derived fibroblasts, expressing some specific CAF markers and a marked pro-inflammatory phenotype [90,91].

3.2. Epidermal Organization

Epidermal cells are organized in a multilayered stratified structure prevalently occupied by keratinocytes at different levels of differentiation [92]. Epidermal thickness varies depending on the anatomical location and the presence of the appendix. The basal membrane, also known as basal laminae, provides the primary function of anchoring down the basal keratinocytes of the epidermis to the connective tissue of the papillary dermis. Keratinocytes are anchored to the basal membrane through hemidesmosomes and to adjacent cells with desmosomes (specialized adhesion structures) [92,93]. In healthy skin, melanocytes are confined in the basal layer. As discussed below, passing the basal membrane and invading the dermis is a crucial step for the acquisition of metastatic competence of melanoma cells [37].
Keratinocytes modify their appearance from one layer to the next with the function of completing a convoluted differentiation process. Starting from the bottom of the epidermis upwards, we find the stratum basale, stratum spinosum, stratum granulosum, and stratum corneum. Stratum basale is a germinative layer and it is the deepest layer of the epidermis. It comprises a monolayer of keratinocytes with high mitotic activity, responsible for the intrinsic continuous renewal of the skin. Here, the self-renewing cell population (stem cells) ensures continuous epidermal renewal in the course of the human lifetime. During the migration, cells gradually fill the cytoplasm with intermediate filament proteins (keratins 1 and 10 and filaggrin) and lipids which conifer toughness to the epidermis [92]. At the same time, keratinocytes change architecture from cuboidal to polygonal shape that progresses through largely flattered morphology culminating in the disk-like shape of corneocytes (also called squamous cells). As part of the differentiation program, keratinocytes lose their nucleus and the major part of cytoplasmic organelles through a degenerative evolution partially overlapping those of the apoptotic process [94,95]. Finally, to complete their differentiation (cornification), a cornified envelope replaces the plasma membrane and keratinocytes undergo a specific pH- and calcium-dependent cell death (corneoptosis) [96]. Indeed, the stratum corneum is composed of 20–30 layers of corneocytes filled with large granules of keratohyalin of which the major constituent is pro-filaggrin [97]. Within the cells, there are also lamellar bodies, consisting of lipids (mainly glycolipids, cholesterol, and ceramides); the lipids released into the extracellular space through exocytosis form a hydrophilic barrier on the corneocytes (corneocyte lipid envelope) and confer waterproofness to the cutis [98]. The protein fraction of the corneocyte envelope includes proteins like involucrin, loricrin, envoplakin, and periplakin cross-linked to lipids [96]. Stratum corneum represents the first line of defense against several insults, protecting the skin from UV-irradiation and chemical injury, preventing toxins and microorganisms from entering the body, and playing a crucial role in trans-epidermal water loss [99]. Corneocytes are held together through corner-desmosomes until cells leave the stratum corneum by desquamation, the process of cell shedding from the skin’s surface [92].
The epidermis hosts a relevant number of resident immune cells and derived molecular mediators. Leucocytes, dendritic T cells, macrophages, mast cells, natural killer, and Langerhans cells are strategically disseminated along all epidermal layers [100,101]. Stratum corneum includes several microbial sensors such as Toll-like receptors, antimicrobial peptides, and cytokines produced physiologically and in response to microbial invasion [102].

3.3. Epidermal–Melanin Units

In normal epidermis, the structural and functional architecture enclosing keratinocytes and melanocytes in an intimate relationship with each other is referred to as an “epidermal–melanin unit” (Figure 1B). The epidermal–melanin unit denotes the symbiotic relationship in which one melanocyte has the task of carrying melanosomes to neighboring keratinocytes. The transfer of melanin protects keratinocytes from UV-induced nuclear damage, which in turn controls the proliferation and differentiation of melanocytes in the basal layer of the epidermis [103]. The number of melanocytes, per unit area of the epidermis is the same in all the human skin phenotypes, hence the skin color is a function of the quantity and the type of melanin produced, the number, and size of melanosomes transferred into the surrounding keratinocytes [104,105]. Caucasians have small melanosomes, lightly pigmented usually absent in the most superficial layers of the epidermis, while in black subjects melanosomes are larger, darker, and almost reach the stratum corneum [106,107]. According to Iozumi K. et al. (1993), differences between white-skin melanocytes and dark-skin melanocytes are related to the intensity of tyrosinase activity [108]. Furthermore, the quantity and quality of melanin are influenced by a variety of genes including MC1R, OCA2, DRD2, EGFR, DCT, KITLG, SLC24A5, SLC24A5, TYRP1, DTNBP1, MYO5A, MFSD1, DDB1, and HERC2, having different impacts in the function of the ethnicity [36,105].
Hyperpigmentation can occur pathologically, as in cutaneous melasma due to an increase in melanin production and transfer with a change in the density of melanocytes resulting in dark patches on the skin, in nevi and melanoma due to benign and malignant hyperproliferation of melanin-producing cells respectively. On the other hand, the whole absence of pigmentation results in albinism, a genetic condition, and the partial absence in skin patch results in vitiligo (acquired condition) and piebaldism (genetic disorder). Post-inflammatory dermal and epidermal hyperpigmentation as well as permanent hypopigmentation are also frequently observed in human skin [109]. From the functional point of view, keratinocytes of the epidermal–melanin unit elaborate several intrinsic and extrinsic stimuli and converge them in a mechanism appropriated to support melanocyte homeostasis and melanogenesis. The transportation of melanosomes, specialized lysosome-derived organelles, where melanin synthesis and packaging occur, is allowed by dendritic protrusions of epidermal melanocytes, which can reach the upper layers of the epidermis. The mechanism of the transfer process of melanosomes from melanocytes to keratinocytes has not been completely defined. Four mechanisms of melanosome transfer have been suggested: (i) direct inoculation of melanosomes into the cytoplasm of keratinocytes via keratinocyte–melanocyte membrane fusion; (ii) release of single melanosomes from melanocytes and their subsequent endocytosis by keratinocytes; (iii) the shedding of melanosome-laden globules by melanocytes, and (iv) direct cytophagocytosis of melanocyte dendrite tips containing melanosomes by adjacent keratinocytes [110,111]. Once incorporated into recipient keratinocytes, melanosomes are predominantly arranged in a supranuclear cap and degraded as the keratinocytes undergo terminal differentiation and desquamation. In dark-skinned individuals, large melanosomes are maintained as individual organelles throughout the cytosol of the keratinocyte. In contrast, in light-skinned individuals, melanosomes are smaller and are aggregated in clusters of 4–8 units [112].
Skin melanocytes are considered intermittent mitotic cells since this type of cell proliferates on demand in case of UV exposure or regenerative processes [113]. Similar to most of the human cell types, an inverse relationship between age and the proliferative activity of melanocytes has been observed [114,115]. Melanosome transfer mainly involves the epidermal basal layer to ensure effective photoprotection for progenitor keratinocytes and stem cells that reside in the microenvironment of the basal epidermis. In the skin, the major types of melanin are brown-black eumelanin and yellow-red pheomelanin. Eumelanin is characterized by high protective properties against DNA damage induced by UV radiation, whereas pheomelanin pigment exhibits a phototoxic pro-oxidant behavior [116]. Tyrosine, a nonessential amino acid, serves as the precursor molecule for the production of all types of melanins. The key enzymes involved in this process include tyrosinase, which catalyzes the initial step of converting tyrosine into dopaquinone, tyrosinase-related protein 1 (TRP1), and dopachrome tautomerase (DCT), which further serves for eumelanin biosynthesis. Independently of the presence of TRP1 and DCT, in the presence of cysteine, dopaquinone is converted into cysteinyldopa and then into pheomelanin by oxidation. Thus, the function of the relative expression of these three enzymes depends on the ratio of eumelanin and pheomelanin and ultimately the skin color. Independently of the presence of TRP1 and DCT after conjugation with cysteine, dopaquinone is converted into pheomelanin [117]. The gene expression of Tyrosinase, TRP1, and DCT, is under the control of MITF (microphthalmia-associated transcription factor), the key transcription factor in melanocyte differentiation. MITF also enhances melanogenesis, activating the transcription of PMEL (a melanosome protein also reported as PMEL-17, gp100, ME20, HMB-45, or silver protein), GPR143 (G protein-coupled receptor 143), and MLANA (Melan-a), coding for structural melanosome-associated proteins [118,119]. MITF is mainly regulated by cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA), cAMP-response element binding protein (CREB) transcription factor, and Wnt/β-catenin signal pathways may function as an oncogene, reinforcing the existing link between melanogenesis and melanomagenesis [120,121]. MITF function is additionally controlled at post-transcriptional and post-translational levels and the availability of transcriptional partners [122]. Excluding mutations and other genetic alterations occurring in melanoma cells, the elevated number of intrinsic and microenvironmental factors contributing to MITF function modulation explains the intra- and inter-tumoral remarkable heterogeneity reported [123,124].

3.4. Physiological Keratinocyte-Melanocyte Communication

The interaction between keratinocytes and melanocytes is relevant to provide the homeostasis of the epidermis. First, to ensure functional optimization of the epidermal–melanin unit, keratinocytes and melanocytes are precisely organized in the space; one post-mitotic melanocyte is in contact with their dendrites with about 36–40 keratinocytes [125]. Keratinocyte–melanocyte interaction occurs through direct cell–cell physical contact as well as through intense bidirectional paracrine communication.

3.4.1. Cell–Cell Contact in Melanocyte-Keratinocyte Interaction

Cell adhesion molecules such as E (epithelial)-cadherin, P (placental)-cadherin, and Desmocollin-1 are important regulators for keratinocyte differentiation by modulating homotypic and heterotypic cell–cell interactions [126,127]. Furthermore, E-cadherin is not only critical to balance the differentiation/proliferation states of keratinocytes and melanocytes but also to ensure epidermal cell survival [128]. Cadherins are transmembrane proteins included in the adherens junction with multiple Ca2+-dependent extracellular domains and a relatively short cytoplasmic domain that connects them to the cytoskeleton [128]. Keratinocytes and melanocytes express on the surface both E-cadherin and P-cadherin but E-cadherin is primarily responsible for the adhesion of human melanocytes to keratinocytes [129]. The membrane expression of E-cadherin and other filopodia-associated proteins is important for the transfer of melanosomes from producing cells to keratinocytes following UV exposure [130,131]. In partial contradiction to the pro-melanogenic role of cadherin-mediated adhesion, UV irradiation enhancing ET-1 (endothelin-1) release by keratinocytes downregulates E-cadherin in melanocytes and melanoma cells [132]. Recently, Pmel17 delivery in the extracellular environment by melanocytes has been described as an additional UV-induced mechanism for temporally weak melanocyte–keratinocyte contact [133]. In this study, Hu and co-authors demonstrated that a soluble form of Pmel17 when internalized in the keratinocyte interacts with the scaffold protein FHL2 (four-and-a-half LIM-domain protein 2) triggering actin cytoskeleton remodeling and E-cadherin reduction, that compressively favor melanocyte migration. However, normally this mechanism is part of the formation of a new epidermal–melanin unit that imposes the rapid recoupling with keratinocytes [134]. Dendrites retraction and melanocyte decoupling from neighboring keratinocytes is a prerequisite for melanin-producing cell division. Impairment of this process might facilitate melanoma development. The intracellular domain of E-cadherin binds β-catenin limiting the nuclear localization and function of this co-transcription factor. On the other side, nuclear β-catenin when complexed with LEF-1 represses E-cadherin transcription reinforcing the loss of adhesion [135]. GPNMB (Glycoprotein nonmetastatic melanoma protein B), another MITF-regulated melanosome-associated glycoprotein, also exists in a soluble form [136]. GPNMB also functions as an adhesion protein between basal keratinocytes and melanocytes since its extracellular domain binds to integrin in the process of maintaining cell–cell adhesion in a calcium-dependent way [137]. Both keratinocytes and melanocytes express and secrete GPNMB and the amount of this protein is augmented after UVA irradiation, by α-MSH (α-melanocyte stimulating hormone) and pro-inflammatory cytokines like IFN-γ and TNF-α. Interestingly, lack of GPNMB expression is a specific characteristic of depigmented vitiligo skin [138]. Evidence that GPNMB protects melanocytes from cytotoxicity induced by oxidative stress further argues for a role in the vitiligo pathogenic mechanism [139]. Recently, a pro-metastatic function has been attributed to the melanoma-derived soluble form of GPNMB due to its capacity to exclude T-lymphocytes from the pre-metastatic niches [136].
Desmosomes are formed by glycoproteins such as desmogleins (or desmosomal cadherins) and desmocollins. Desmoglein 1 (Dsg1), in addition to its adhesive function, can modulate keratinocytes–melanocytes paracrine communication. In particular, it has been demonstrated that melanocytes within a Dsg1-deficient 3D human skin exhibited increased pigmentation, increased production of paracrine factors such as cytokines/chemokines, and altered dendritic protrusions. Therefore, this scenario highlights that Dsg1 deficiency may contribute to pagetoid behavior, such as occurring in early melanoma development [140]. Different types of cell–cell adhesion molecules are connexin, integral membrane proteins clustered in the gap junctions, and occludins and claudins included in the tight junctions. Tight junctions allow passage of ions and small molecules between adjacent cells. Gap junctions are channels in which secondary messengers and metabolic products are easily exchanged between coupled cells [141]. Alteration of gap junction communication mostly reflects an atypical cadherin expression profile [142]. It has been demonstrated that blocking gap junctions in a co-culture system of human keratinocytes and melanocytes reduces the expression of key regulatory genes of melanogenesis such as tyrosinase and MITF in a dose-dependent manner [143]. UVB and UVA transiently alter the integrity of epidermal gap junctions and cancer cells frequently present downregulated levels of gap junctions suggesting a role of these complexes in the regulation of UV-induced inflammation and skin carcinogenesis [131,144].
In normal epidermis, integrins are important for the connection of basal cells to extracellular matrix components such as collagen, laminin, and fibronectin and for any type of tissue remodeling including normal migration of cells and wound healing. Alpha2/beta 2 integrins are implicated in resident lymphocyte recruitment due to the binding to ligands of the Ig immunoglobulin family, such as ICAM-1, ICAM-2, and ICAM-3 [145].

3.4.2. Cross-Talk in the Epidermal–Melanin Unit

The most evident exchange between cells in the epidermis is the melanin-containing melanosome transfer that aims to protect keratinocytes from the deleterious effect of UV irradiation [146]. Keratinocytes, on the other hand, control the proliferation, adhesion, migration, and differentiation of melanocytes (including the melanin biosynthetic pathway) through the secretion of growth factors and cytokines in a paracrine manner [147,148]. Several molecules are identified as involved in melanocyte growth, such as basic fibroblast growth factor (bFGF), stem cell factor (SCF), endothelin 1 (ET1), ET3, and hepatocyte growth factor (HGF) [149,150]. Among these mitogenic factors, ETs, SCF, and HGF are mainly provided by keratinocytes. In physiological conditions, the production of POMC, the α-MSH precursor, bFGF, and ET-1 is mostly due to p53-dependent transcriptional activation in response to UV [40,151]. These mitogens display a synergistic effect on melanocytes, activating the MAP Kinase pathway, especially the kinase ERK2 [152]. P53 plays a central role in hyperpigmentation [153]. At variance, mutations in the p53 gene are rare in primary melanoma [154]. In contrast, repeated daily exposure to UVB induces a p53-dependent melanocyte senescence [155]. Keratinocytes release a significant amount of bFGF after UV irradiation increasing expression of focal adhesion kinase p125FAK on melanocytes and facilitating their migration [156]. Transwell-based assay of normal human epidermal melanocytes demonstrated that PI3K/Akt-Rac1-FAK-JNK-ERK signaling pathways lead to cytoskeleton reorganization contributing to melanocyte migration after bFGF stimulation [157]. Similarly, HGF-binding transmembrane tyrosine kinase receptor-c-Met stimulates NF-kB and MAPK increasing the proliferation and motility of melanocytes [158]. Upon ligand binding, c-Met activates PI3K MAP Kinases through the Gab1/Grb2-SOS Ras pathway [159,160]. After UVB irradiation, epidermal cells strongly induce HGF expression. Further, keratinocytes produce a large amount of IL-1α that stimulates HGF production by dermal fibroblasts. At the same time, UVA directly upregulates HGF mRNA and protein [161].
SCF/KIT pathway is important for normal melanocytic function; experiments carried out with adult human skin xenografts injected with recombinant human SCF or a KIT-inhibitory antibody demonstrated that SCF/KIT pathway impairment elicited a melanocyte loss and a reduced expression of melanocytic differentiation antigens TRP-1 and pmel17. Hence, this investigation suggests that the SCF/KIT pathway plays a critical role in the control of human melanocyte homeostasis [162]. Keratinocytes-derived SCF has pleiotropic effects. It can modulate integrin expression at the protein level through SCF facilitating melanocyte attachment and migration on ECM ligands, a process needed during embryogenesis and the repigmentation process [163].
In the presence of IL-1α and after UVB exposure, keratinocytes produce a high amount of ET-1 triggering a stimulatory effect on DNA synthesis and melanization in cultured human melanocytes [164]. The signaling mechanism involved is represented by the crosstalk between PKC, cyclin AMP system, and MAP kinase [165]. A recent study demonstrated that after UV irradiation, ET-1 protects human melanocytes, inhibiting apoptosis and reducing DNA photoproducts in a dose-dependent manner. ET-1 enhances the phosphorylation of JNK and p38 that target the transcription factor ATF-2. ET-1 synergistically with α-MSH regulates DNA repair to in response to UV. After binding to the receptor, ET-1 causes phosphorylation of repair-DNA sensors ATM/ATR, therefore enhancing the level of DNA damage recognition proteins like XPC, VCP, XPA, and γH2AX [52].
Cell migration is essential in inflammatory reactions, remodeling, and healing. Keratinocytes express several MMPs (matrix metalloproteinases) that can cleave components of the extracellular matrix, preparing an optimal context for cell mobilization. However, normally the expression of MMPs and ADAMs (a disintegrin and metalloproteinase) are very low and their function is regulated by proteolytic activation to ensure fast remodeling of the dermis, as it became necessary. Several MMPs are UV-responsive genes. On keratinocytes, UVB activates the transcription of a cluster of MMP genes indicating that coordinated regulation of these proteins is important for damage response [166]. Specifically, MMP9 and MMP2 secreted in response to UV or proinflammatory cytokines such as IFN-γ, TNF-α, IL-1β, and IL-6 are involved in inflammatory reactions, remodeling, and healing [167,168]. In this process, keratinocytes have a direct as well as an indirect role since UVB-irradiated keratinocytes releasing IL-1 activate the expression of MMP1 in dermal fibroblasts [169]. On the other hand, MMP1 in addition to collagen damage exacerbates the skin inflammatory response [170]. Dysregulation of MMPs affects skin homeostasis by premature aging and facilitating disease development. ADAM12 is upregulated in the non-healing edge of chronic skin ulcers [171], whereas ADAM10, ADAM12, and ADAM17 [172], MMP2, MMP9, MM12, and MMP7 [173,174] are highly expressed in psoriasis, a common hyperproliferative and inflammatory skin condition. MMP9 produced by keratinocytes in response to IFN-γ has been also implicated in melanocyte detachment from the basal epidermal layer in vitiligo skin [175]. Notably, since the ability of melanocyte migration during re-pigmentation also largely depends on MMP9 activity [176], impaired MMP activity has been proposed to explain the reduced migration of melanocyte precursor from the outer root sheath in vitiligo skin [177].

4. Remodeling of Epidermal Microenvironment during Melanoma Onset and Progression

To replace old cells, and repair or expand the skin, melanocytes need to retract their dendrites, separate from the basement membrane and neighboring keratinocytes, proliferate, and migrate along the basement membrane before they reach a new epidermal–melanin unit. Since these processes are largely regulated locally by surrounding cells, it is reasonable that melanocytic hyperproliferations are due to the mistaken escape from keratinocyte control. Accordingly, in vitro, keratinocytes control proliferation to maintain the original keratinocyte/melanocyte ratio in a cell–cell direct contact-dependent manner [178]. Mechanistically, escape from keratinocyte control might be due to loss of contact with keratinocytes, modification of keratinocytes’ secretory behavior, or alternatively, the acquisition of unresponsiveness to inhibitory growth factors by melanocytes. During the initial radial growth phase, melanoma cells are still confined in the epidermal compartment. Thus, it is conceivable that poorly aggressive melanoma acquires aggressiveness being still surrounded by normal keratinocytes (Figure 2). This process is likely dependent on the modification of cell adhesion properties and the paracrine activity of keratinocytes.

4.1. Alteration of Melanocyte–Keratinocyte Physical Interaction during Melanoma Inception and Progression

Melanocytes and corresponding early hyperproliferative melanocytic cells are fully surrounded by keratinocyte layers that physiologically protect long-living pigment-producing cells. Thus, the simple reduction of these activities by stressed or damaged keratinocytes might represent in theory a pro-melanogenic condition. Sun exposure is the major environmental risk for epidermal homeostasis. Frequent excessive exposure to UV light is associated with alterations in the composition of the basement membrane and dermal extracellular matrix that might facilitate disease occurrence. Chronic UV irradiation decreases the expression of epidermal type VII collagen that serves for dermal–epidermal junction integrity and fibrillins in the upper dermis [179,180]. Moreover, the expression of β1 integrins that serve for the connection of basal keratinocytes to the basement membrane is severely compromised in sun-damaged skin [181]. At the same time, higher expression of involucrin in the stratum corneum and lower β1 integrins in the basal epidermis argue for impaired self-renewal and adhesive properties of UV-damaged keratinocytes [181]. In chronically sun-exposed skin, qualitative and quantitative modifications of extracellular matrix (ECM) proteins cause loss of tensile strength, increase fragility, and lessen reparative ability [73,182]. Even so, the tissue surrounding benign nevi and melanomas displays greater stiffness than normal skin evidencing that mechanical properties of the matrix have a role in melanoma initiation and control progression of the invasive process in advanced melanoma [183,184]. UV-irradiated skin produces also several MMPs, which degrade dermal collagen and elastic fibers causing an overall tissue remodeling that is part of the photoaging process [185]. Photoaging is characterized by poorly organized ECM, wrinkling, irregular pigmentation, telangiectasia, thinned or fragile skin, and dermal and epidermal atrophy. Epidermis-releasing MMPs display an accelerated melanocytic cell invasion of the dermis due to basal membrane destruction [186]. Instead, in the context of melanoma, excessive production of MMP1, MMP2, MMP9, and MMP19 has been frequently associated with invasion behavior of tumor cells [187,188]. Overall, modification of the balance MMPs/TIMPs (tissue inhibitors of MMPs) is critical for melanoma invasion [189]. MMP9 overexpression is frequently associated with aberrant MAPK pathway activation, a very frequent event in melanomagenesis [190]. Other mechanisms for MMP9 induction are the dysregulation of TGFβ [191], mutation in the MMP9 gene [190], and epigenetic events [192]. TGFβ elevation in the tumor microenvironment is due to autocrine production by tumor cells as well as to the release by keratinocytes [193] and fibroblasts [194] subsequent to UV radiation. Furthermore, the expression of several components of ADAM family proteins is remarkably increased in melanoma cells, with significant differences between metastases and primary melanocytic lesions [195]. ECM degradation favors melanoma spreading due to the activation of proangiogenic factors in the surrounding tissue. Overexpression of ADAMs and MMPs involves melanoma peritumoral stromal fibroblasts [196]. A possible similar activation of keratinocytes surrounding melanoma has been suggested by in vitro evidence that medium conditioned by melanoma cells stimulates the production of MMP3, MMP9, and MMP14 by Hacat cells while lowering TIMP expression [197].
Cell adhesion molecules including cadherin, integrin, immunoglobulin, and selectin have been related to the melanoma metastatic process [128]. E-cadherin is the most critical molecule for keratinocyte–melanocyte interaction because melanocytes not expressing E-cadherin lose most of the physiological regulation by keratinocytes [198].
From the early stages of melanoma, loss of E-cadherin expression and acquisition of N (neuronal)-cadherin expression allows the melanoma cells to preferentially bind to fibroblasts that abundantly express this cadherin, and thus promote invasion into the dermis [198]. E-cadherin/N-cadherin shift represents the hallmark of epithelial-to-mesenchymal transition (Figure 3).
Distraction of E-cadherin from the membrane surface has also been implicated in the regulation of Wnt/β-catenin since its intracellular domain binds β-catenin, counteracting the nuclear localization of this key co-transcription factor for melanocyte biology. Β-catenin membrane-to-nuclear translocation and gain of nuclear localization enable the binding to members of lymphoid enhancer-binding factor (LEF)/T-cell specific factor (TCF) family and some other co-regulators to promote the transcription of ubiquitous genes (Jun, c-Myc and CyclinD-1) [199] and melanocyte-lineage restricted genes including MITF [200], DCT, and Brn-2 [201,202,203]. Some recent data suggested that stromal cells might cause atypical melanocyte detachment and exclusion from the normal epidermal–melanin unit. Using a trans-well co-culture system to keep in touch with melanoma cell lines with poor aggressive behavior and healthy epidermal keratinocytes, Untiveros and collaborators demonstrated that tumor cells progressively assume a mesenchymal-like phenotype resulting in a decrease in E-cadherin expression and activating stabilization of β-catenin [204]. Similar results were recorded with dermal fibroblasts with the difference that keratinocytes additionally activated ERK signaling [204]. Discrepancies emerged comparing in vivo and in vitro studies regarding the effect of Wnt/β-catenin signaling on proliferation [205,206], invasion [202,207], and migration [208] of melanoma cells. Further, in vivo, observations demonstrated that β-catenin nuclear localization correlates with improved patient survival [208,209]. Following the loss of E-cadherin, the dropping of Dgs1 expression in keratinocytes discontinues control of melanocytes contributing to melanomagenesis [140]. Melanocytes in Dsg1-silenced skin equivalents delocalize suprabasally, contributing to the pagetoid behavior occurring in early melanoma development. In a co-culture model, silencing Dsg1 expression in keratinocytes increases the production of the melanocortins precursor POMC and increases melanin production and distribution even in the absence of UV stimulation [140]. Reinforcing the relevance of Dsg1 in melanocyte biology, Dsg1 is reduced in response to acute UV exposure [210]. Elevated levels of Tspan8 (Tetraspanin8) expression on the melanoma membrane confer the capacity to invade the dermis [211]. The mechanism of Tspan8-dependent dissolution of dermo-epidermal junction depends on the presence of keratinocytes and specifically on their capacity to produce MMPs and TIMPs [211]. Due to the tight correlation between Tspan8 expression and invasive capacity, Tspan8 has been proposed to predict metastasis risk in individual patients [212].
Melanoma cells lose the capacity to form gap junction communication with keratinocytes but still use these structures to communicate with themselves and with dermal fibroblasts [141]. Interestingly, several studies described the role of gap junctions in melanoma-immune cell interaction [213,214] indicating implications in anti-melanoma immune response and disease control. In skin cancer, Cx43 (connexin-43) is overexpressed in malignant melanomas when compared with the common and benign nevi, and higher levels of expression were found in patients with lymph node metastases. The upregulation of Cx43 was associated with tumor spreading since Cx43 mediates intracellular communication between the tumor microenvironment and the metastatic melanoma cells [215]. In contrast, abundant cytoplasmic Cx43 observed at later stages has been linked to invasion and metastasis [216]. Thus, the definition of oncogene has been proposed for Cx43 in melanoma; however, in human melanoma cell lines, Cx43 overexpression mitigates melanoma growth and metastasis and facilitates TNFα-induced cell apoptosis [217]. Cx31·1 and Cx26 are significantly downregulated in metastases compared with vertical growth phase melanomas. Cx26 expression in melanoma tissue has been shown to promote a metastatic cell phenotype and enhance the establishment of new tumor niches through cell-to-cell communication with the surrounding tissue. Very recently, Cx31 downregulation has been associated with melanoma metastasis, MAPK inhibitor resistance, and worse overall survival of patients [218].
The selectin family plays a major role in the progression of melanoma, as well as initiating complications in cancer patients such as coagulation defects, leading to a poorer prognosis [219]. Selectins are glycoproteins with a key role in cancer cell rolling, adhesion, extravasation, and the establishment of metastatic lesions [128]. E-selectin, (faintly expressed in normal melanocytes) has been shown to regulate the transepithelial migratory pathway through p38 and ERK kinases [219] and to facilitate immune evasion [128]. Melanocyte–keratinocyte physical interaction is not only critical to confine melanocytes in their physiological niche but also to maintain the regular protein surface profile of pigment cells. Accordingly, after isolation from skin normal human melanocytes in vitro express some melanoma-associated antigens [220].

4.2. Keratinocyte–Melanoma Cells Cross-Talk

Melanoma cells at the early stage are still fully embedded in the epidermis which is quite a homogenous environment. Thus, spontaneous as well as melanoma-driven alterations in keratinocyte layers undoubtedly serve for melanoma niche formation and progression. However, concerning the paracrine function of epidermal cells in melanoma, it is necessary to also take into consideration the role of intercellular communication with distant cells. Particularly, the complex melanocyte–keratinocyte–fibroblast network is critical for melanocyte homeostasis. Keratinocytes directly offer several mitogenic factors and indirectly stimulate the secretion of growth factors by fibroblasts. The acquisition of growth factor independence is a significant step in melanoma progression. Until the melanoma mutational profile does not involve key cell cycle regulators, the up modulation of growth factors by stromal cells appears to be the turning point for pro-tumorigenic switching of normal skin. While the hypersecretion of mitogenic molecules is a well-defined feature of CAFs, until now the corresponding secretory profile of melanoma-associated keratinocytes has not been fully elucidated. In wild-type BRAF melanocytes, the activation of the MAPK signaling can be because of abnormal production of mitogens by surrounding cells and by autocrine production of the growth factors bFGF and HGF [221,222]. Keratinocytes upregulate mitogenic factors in response to UV and during wound healing. Keratinocyte-derived NGF (nerve growth factor) serves to counteract UV-induced depletion of melanocytes by increasing the level of the anti-apoptotic BCL-2 protein [223], a mechanism that might help melanoma cells bypass critical phases. HGF, PDGF, and ET-1 decrease the expression of E-cadherin, leading to decoupling from keratinocytes [132,224]. Overall, these data evidence that E-cadherin is a very dynamic molecule that can relatively easily be down-modulated. Decoupling from keratinocytes does not necessarily reflect only E-cadherin levels in tumors since melanoma cells might orchestrate the remodeling of surface protein on keratinocytes. Emerging literature indicates that in addition to cell proliferation and survival, sphingolipid metabolism is implicated in the modulation of cell adhesion properties. Lipid mediators such as sphingosine 1-phosphate (S1P) released by melanoma cells reduce E-cadherin on the keratinocyte membrane and stimulate MMP9 expression in the same cells [225]. Correspondingly, melanoma-derived S1P activates stromal fibroblasts and promotes transdifferentiation into myofibroblasts [226].
The interaction of growth factors with proper receptors activates MAPK intracellular signaling and a cascade of events converging in cell division stimulation. However, chronic MAPK activation occurring in the case of BRAF mutations does not necessarily lead to loss of growth controls. Nevi harboring BRAF mutation with a frequency near 80% still require exogenous growth factors and have a finite life span similar to healthy melanocytes [63,221,222,227]. It is well-known that these precancerous focal proliferations of melanocytes stop growth due to the acquisition of oncogene-induced senescent features [228]. The exit of nevi from persistent cell senescence is paradigmatic in cancer biology. Using a microfluids system to leave intact indirect melanocyte/keratinocyte communication, a recent study showed that keratinocyte as well as dermal fibroblasts might alter the secretion of both pro- and anti-tumorigenic factors by transformed melanocytes leading to BRAF-induced senescence escape [229]. From the same study, emerged a phototype-dependent disparity in the secretome of melanocytes regardless of the neovascularization stimulation.
Expanding the forms of melanocyte–keratinocyte communication, Tagore et al. recently described a pro-melanomagenesis interaction based on GABAenergic (gamma-aminobutyrric acid) signaling [230]. Considering the embryological origin of melanocytes, it is not unexpected that GABA-synthesizing enzymes are expressed by pigment cells. However, the augmented level of GAD1 (glutamic acid decarboxylase) upon BRAFV600E activation is associated with a high level of GABA receptors in non-excitable keratinocytes directly adjacent to melanoma cells, suggesting an electrical activity-based communication is more surprising [230].
The contribution of epidermal keratinocytes to melanoma progression seems to be not restricted to cells residing in the proximity of melanocytic hyperprolipheration since in a very interesting investigation Golan and collaborators demonstrated that differentiated keratinocyte and not undifferentiated ones induce melanoma invasion via a Notch-dependent mechanism [231]. Confirming that keratinocytes are not unique static elements of the epidermis, when in contact with undifferentiated keratinocytes, melanocytes exhibit features of healthy melanocytes but when in contact with differentiated keratinocytes, melanocytes acquire the expression of phenotypic characteristics of nevus and melanoma cells [178].
Tumor lesions are frequently surrounded by persistent inflammation [232]. NALP3 (NRLR family pyrin domain containing 3) and additional components of the inflammasome multiprotein complex are highly activated in melanoma [233]. Inflammasome features correlate to a favorable prognosis in melanoma patients to the efficacy of immune checkpoint blockade therapy [233]. Local inflammatory mechanisms are associated with higher levels of circulating proinflammatory cytokines such as IL-8 (interleukin-8), IL-6, and INFα (interferon-α) [234]. Ultraviolet irradiation stimulates keratinocytes to secrete numerous cytokines that are responsible for the proliferation of melanocytes and transient melanogenesis activation. Chronic UVB results in IL-1, IL-8, and TNF (tumor necrosis factor) secretion by keratinocytes [235]. However, UV radiation is prevalently immune suppressive, a fact that might enable malignant cells to escape local (skin) immune control. Complicating the scenario, IL-12 in addition to the immune system regulatory function is involved in DNA damage repair of keratinocytes indicating that suppression of local immune response is necessary to restore compromised cells. Exposing the skin to solar-simulated UV radiation enhances the production of immunomodulatory lipids. Changes in lipid metabolism directly affect immune-cell phenotype and function, including increasing the production of cytokines that suppress the immune system. Interestingly, microvesicles generated by UVB-treated epidermal layers containing immunosuppressive molecules allow systemic effects [236]. Thus, body areas not exposed directly to UV radiation might receive immunosuppressive mediators derived from irradiated keratinocytes and released in blood circulation [237].

5. Conclusions

Rapidly dividing cells, such as keratinocytes, are quickly replaced by new differentiating cells due to the physiological turnover, whereas melanocytes are long-living cells constantly undergoing damage accumulation. Despite this significant difference, these two types of cells exist in a symbiotic relationship rarely perturbed during normal life. Skin regeneration, wound healing, and tanning response are tightly regulated processes that ensure the unmodified melanocyte/keratinocyte ratio. Also, cell adhesion features and direct contact between melanocytes and keratinocytes are only transiently modulated during skin remodeling. However, melanocytic hyperproliferation imposes a redesign of the epidermal picture. In the case of focal benign hyperproliferation such as in the case of nevi skin remodel reaches a stable point of equilibrium that rarely involves the dermis. By contrast, in the case of melanoma, tumor cells digress from the epidermis invading the dermis. This step is often associated with the switch from an epithelial-like to a mesenchymal-like phenotype of tumor cells. Thus, understanding the phenomena underlying the loss of epidermal homeostasis during the initial stages of melanoma is essential to optimize the diagnosis and treatment of early lesions.

Author Contributions

Conceptualization, Writing, Review, and Editing: B.B.; Data Curation Writing and Editing: R.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Italian Ministry of Health (Ricerca Corrente).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Long, G.V.; Swetter, S.M.; Menzies, A.M.; Gershenwald, J.E.; Scolyer, R.A. Cutaneous Melanoma. Lancet 2023, 402, 485. [Google Scholar] [CrossRef]
  2. Saginala, K.; Barsouk, A.; Aluru, J.S.; Rawla, P.; Barsouk, A. Epidemiology of Melanoma. Med. Sci. 2021, 9, 63. [Google Scholar] [CrossRef]
  3. Karimkhani, C.; Green, A.C.; Nijsten, T.; Weinstock, M.A.; Dellavalle, R.P.; Naghavi, M.; Fitzmaurice, C. The Global Burden of Melanoma: Results from the Global Burden of Disease Study 2015. Br. J. Dermatol. 2017, 177, 134. [Google Scholar] [CrossRef]
  4. Teixido, C.; Castillo, P.; Martinez-Vila, C.; Arance, A.; Alos, L. Molecular Markers and Targets in Melanoma. Cells 2021, 10, 2320. [Google Scholar] [CrossRef]
  5. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  6. Carr, S.; Smith, C.; Wernberg, J. Epidemiology and Risk Factors of Melanoma. Surg. Clin. N. Am. 2020, 100, 1–12. [Google Scholar] [CrossRef]
  7. Nilsen, L.T.N.; Hannevik, M.; Veierød, M.B. Ultraviolet Exposure from Indoor Tanning Devices: A Systematic Review. Br. J. Dermatol. 2016, 174, 730–740. [Google Scholar] [CrossRef] [PubMed]
  8. Boniol, M.; Autier, P.; Boyle, P.; Gandini, S. Cutaneous Melanoma Attributable to Sunbed use: Systematic Review and Meta-Analysis. BMJ 2012, 345, e4757. [Google Scholar] [CrossRef] [PubMed]
  9. Dzwierzynski, W.W. Melanoma Risk Factors and Prevention. Clin. Plast. Surg. 2021, 48, 543–550. [Google Scholar] [CrossRef]
  10. Bernerd, F.; Passeron, T.; Castiel, I.; Marionnet, C. The Damaging Effects of Long UVA (UVA1) Rays: A Major Challenge to Preserve Skin Health and Integrity. Int. J. Mol. Sci. 2022, 23, 8243. [Google Scholar] [CrossRef]
  11. Jin, S.; Padron, F.; Pfeifer, G.P. UVA Radiation, DNA Damage, and Melanoma. ACS Omega 2022, 7, 32936–32948. [Google Scholar] [CrossRef]
  12. Gorbunova, V.; Seluanov, A.; Mao, Z.; Hine, C. Changes in DNA Repair during Aging. Nucleic Acids Res. 2007, 35, 7466–7474. [Google Scholar] [CrossRef]
  13. Ren, P.; Dong, X.; Vijg, J. Age-Related Somatic Mutation Burden in Human Tissues. Front. Aging 2022, 3, 1018119. [Google Scholar] [CrossRef] [PubMed]
  14. Maslov, A.Y.; Vijg, J. Somatic Mutation Burden in Relation to Aging and Functional Life Span: Implications for Cellular Reprogramming and Rejuvenation. Curr. Opin. Genet. Dev. 2023, 83, 102132. [Google Scholar] [CrossRef] [PubMed]
  15. Wei, E.X.; Li, X.; Nan, H. Having a First-Degree Relative with Melanoma Increases Lifetime Risk of Melanoma, Squamous Cell Carcinoma, and Basal Cell Carcinoma. J. Am. Acad. Dermatol. 2019, 81, 489–499. [Google Scholar] [CrossRef] [PubMed]
  16. Mohr, C.; Li, Y.; Navsaria, L.J.; Hinkston, C.L.; Margolis, D.J.; Wehner, M.R. Melanoma Risk in Skin of Color Patients with a History of a Keratinocyte Carcinoma. Br. J. Dermatol. 2023, 190, 449–451. [Google Scholar] [CrossRef] [PubMed]
  17. Mercieca, L.; Aquilina, S.; Calleja, N.; Boffa, M.J. Cutaneous Melanoma More Likely to be Invasive in Fairer Skin Phototypes: A Retrospective Observational Study. Skinmed 2021, 19, 280–283. [Google Scholar] [PubMed]
  18. Strashilov, S.; Yordanov, A. Aetiology and Pathogenesis of Cutaneous Melanoma: Current Concepts and Advances. Int. J. Mol. Sci. 2021, 22, 6395. [Google Scholar] [CrossRef] [PubMed]
  19. Bishop, J.N.; Harland, M.; Bishop, D.T. The Genetics of Melanoma. Br. J. Hosp. Med. 2006, 67, 299–304. [Google Scholar] [CrossRef] [PubMed]
  20. Chen, T.; Hemminki, K.; Kharazmi, E.; Ji, J.; Sundquist, K.; Fallah, M. Multiple Primary (Even in Situ) Melanomas in a Patient Pose Significant Risk to Family Members. Eur. J. Cancer 2014, 50, 2659–2667. [Google Scholar] [CrossRef]
  21. Hussussian, C.J.; Struewing, J.P.; Goldstein, A.M.; Higgins, P.A.T.; Ally, D.S.; Sheahan, M.D.; Clark, W.H.; Tucker, M.A.; Dracopoli, N.C. Germline p16 Mutations in Familial Melanoma. Nat. Genet. 1994, 8, 15–21. [Google Scholar] [CrossRef] [PubMed]
  22. Horn, S.; Figl, A.; Rachakonda, P.S.; Fischer, C.; Sucker, A.; Gast, A.; Kadel, S.; Moll, I.; Nagore, E.; Hemminki, K.; et al. TERT Promoter Mutations in Familial and Sporadic Melanoma. Science 2013, 339, 959–961. [Google Scholar] [CrossRef] [PubMed]
  23. Robles-Espinoza, C.D.; Harland, M.; Ramsay, A.J.; Aoude, L.G.; Quesada, V.; Ding, Z.; Pooley, K.A.; Pritchard, A.L.; Tiffen, J.C.; Petljak, M.; et al. POT1 Loss-of-Function Variants Predispose to Familial Melanoma. Nat. Genet. 2014, 46, 478–481. [Google Scholar] [CrossRef] [PubMed]
  24. Puntervoll, H.E.; Yang, X.R.; Vetti, H.H.; Bachmann, I.M.; Avril, M.F.; Benfodda, M.; Catricalà, C.; Dalle, S.; Duval-Modeste, A.B.; Ghiorzo, P.; et al. Melanoma Prone Families withCDK4germline Mutation: Phenotypic Profile and Associations with MC1Rvariants. J. Med. Genet. 2013, 50, 264. [Google Scholar] [CrossRef] [PubMed]
  25. Pastorino, L.; Bonelli, L.; Ghiorzo, P.; Queirolo, P.; Battistuzzi, L.; Balleari, E.; Nasti, S.; Gargiulo, S.; Gliori, S.; Savoia, P.; et al. CDKN2A Mutations and MC1R Variants in Italian Patients with Single or Multiple Primary Melanoma. Pigment Cell Melanoma Res. 2008, 21, 700–709. [Google Scholar] [CrossRef]
  26. O’Neill, C.H.; Scoggins, C.R. Melanoma. J. Surg. Oncol. 2019, 120, 873–881. [Google Scholar] [CrossRef] [PubMed]
  27. Goldstein, A.M.; Xiao, Y.; Sampson, J.; Zhu, B.; Rotunno, M.; Bennett, H.; Wen, Y.; Jones, K.; Vogt, A.; Burdette, L.; et al. Rare Germline Variants in Known Melanoma Susceptibility Genes in Familial Melanoma. Hum. Mol. Genet. 2017, 26, 4886–4895. [Google Scholar] [CrossRef] [PubMed]
  28. Campos, C.; Fragoso, S.; Luís, R.; Pinto, F.; Brito, C.; Esteves, S.; Pataco, M.; Santos, S.; Machado, P.; Vicente, J.B.; et al. High-Throughput Sequencing Identifies 3 Novel Susceptibility Genes for Hereditary Melanoma. Genes 2020, 11, 403. [Google Scholar] [CrossRef]
  29. Castaneda-Garcia, C.; Iyer, V.; Nsengimana, J.; Trower, A.; Droop, A.; Brown, K.M.; Choi, J.; Zhang, T.; Harland, M.; Newton-Bishop, J.A.; et al. Defining Novel Causal SNPs and Linked Phenotypes at Melanoma-Associated Loci. Hum. Mol. Genet. 2022, 31, 2845–2856. [Google Scholar] [CrossRef]
  30. Bruno, W.; Dalmasso, B.; Barile, M.; Andreotti, V.; Elefanti, L.; Colombino, M.; Vanni, I.; Allavena, E.; Barbero, F.; Passoni, E.; et al. Predictors of Germline Status for Hereditary Melanoma: 5 Years of Multi-Gene Panel Testing within the Italian Melanoma Intergroup. ESMO Open 2022, 7, 100525. [Google Scholar] [CrossRef]
  31. Potjer, T.P.; Bollen, S.; Grimbergen, A.J.E.M.; van Doorn, R.; Gruis, N.A.; van Asperen, C.J.; Hes, F.J.; van der Stoep, N. Multigene Panel Sequencing of Established and Candidate Melanoma Susceptibility Genes in a Large Cohort of Dutch Non-CDKN2A/CDK4 Melanoma Families. Int. J. Cancer 2019, 144, 2453–2464. [Google Scholar] [CrossRef]
  32. 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 2021, 12, 1093. [Google Scholar] [CrossRef] [PubMed]
  33. Tagliabue, E.; Gandini, S.; Bellocco, R.; Maisonneuve, P.; Newton-Bishop, J.; Polsky, D.; Lazovich, D.; Kanetsky, P.; Ghiorzo, P.; Gruis, N.; et al. MC1R Variants as Melanoma Risk Factors Independent of at-Risk Phenotypic Characteristics: A Pooled Analysis from the M-SKIP Project. CMAR 2018, 10, 1143. [Google Scholar] [CrossRef] [PubMed]
  34. Swope, V.B.; Abdel-Malek, Z.A. MC1R: Front and Center in the Bright Side of Dark Eumelanin and DNA Repair. Int. J. Mol. Sci. 2018, 19, 2667. [Google Scholar] [CrossRef] [PubMed]
  35. Rees, J.L. Genetics of Hair and Skin Color. Annu. Rev. Genet. 2003, 37, 67–90. [Google Scholar] [CrossRef] [PubMed]
  36. Feng, Y.; McQuillan, M.A.; Tishkoff, S.A. Evolutionary Genetics of Skin Pigmentation in African Populations. Hum. Mol. Genet. 2021, 30, R88–R97. [Google Scholar] [CrossRef]
  37. Bellei, B.; Migliano, E.; Picardo, M. A Framework of Major Tumor-Promoting Signal Transduction Pathways Implicated in Melanoma-Fibroblast Dialogue. Cancers 2020, 12, 3400. [Google Scholar] [CrossRef]
  38. Cerdido, S.; Sánchez-Beltrán, J.; Lambertos, A.; Abrisqueta, M.; Padilla, L.; Herraiz, C.; Olivares, C.; Jiménez-Cervantes, C.; García-Borrón, J.C. A Side-by-Side Comparison of Wildtype and Variant Melanocortin 1 Receptor Signaling with Emphasis on Protection Against Oxidative Damage to DNA. Int. J. Mol. Sci. 2023, 24, 14381. [Google Scholar] [CrossRef] [PubMed]
  39. Abdel-Malek, Z.A.; Ruwe, A.; Kavanagh-Starner, R.; Kadekaro, A.L.; Swope, V.; Haskell-Luevano, C.; Koikov, L.; Knittel, J.J. A-MSH Tripeptide Analogs Activate the Melanocortin 1 Receptor and Reduce UV-Induced DNA Damage in Human Melanocytes. Pigment Cell Melanoma Res. 2009, 22, 635–644. [Google Scholar] [CrossRef]
  40. Kadekaro, A.L.; Chen, J.; Yang, J.; Chen, S.; Jameson, J.; Swope, V.B.; Cheng, T.; Kadakia, M.; Abdel-Malek, Z. Alpha-Melanocyte–Stimulating Hormone Suppresses Oxidative Stress through a p53-Mediated Signaling Pathway in Human Melanocytes. Mol. Cancer Res. 2012, 10, 778–786. [Google Scholar] [CrossRef]
  41. Herraiz, C.; Journé, F.; Abdel-Malek, Z.; Ghanem, G.; Jiménez-Cervantes, C.; García-Borrón, J.C. Signaling from the Human Melanocortin 1 Receptor to ERK1 and ERK2 Mitogen-Activated Protein Kinases Involves Transactivation of cKIT. Mol. Endocrinol. 2011, 25, 138–156. [Google Scholar] [CrossRef]
  42. Herraiz, C.; Martínez-Vicente, I.; Maresca, V. The A-Melanocyte-Stimulating Hormone/Melanocortin-1 Receptor Interaction: A Driver of Pleiotropic Effects Beyond Pigmentation. Pigment Cell Melanoma Res. 2021, 34, 748–761. [Google Scholar] [CrossRef]
  43. Zhou, Y.; Cai, M. Novel Approaches to the Design of Bioavailable Melanotropins. Expert Opin. Drug Discov. 2017, 12, 1023–1030. [Google Scholar] [CrossRef]
  44. Caini, S.; Gandini, S.; Botta, F.; Tagliabue, E.; Raimondi, S.; Nagore, E.; Zanna, I.; Maisonneuve, P.; Newton-Bishop, J.; Polsky, D.; et al. MC1R Variants and Cutaneous Melanoma Risk According to Histological Type, Body Site, and Breslow Thickness: A Pooled Analysis from the M-SKIP Project. Melanoma Res. 2020, 30, 500–510. [Google Scholar] [CrossRef]
  45. Latreille, J.; Ezzedine, K.; Elfakir, A.; Ambroisine, L.; Jdid, R.; Galan, P.; Hercberg, S.; Gruber, F.; Malvy, D.; Tschachler, E.; et al. MC1R Polymorphisms and Facial Photoaging. Ann. Dermatol. Venereol. 2011, 138, 385–389. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, Y.; André, M.; Adhikari, K.; Blin, M.; Bonfante, B.; Mendoza-Revilla, J.; Fuentes-Guajardo, M.; Palmal, S.; Chacón-Duque, J.C.; Hurtado, M.; et al. A Genome-Wide Association Study Identifies Novel Gene Associations with Facial Skin Wrinkling and Mole Count in Latin Americans. Br. J. Dermatol. 2021, 185, 988–998. [Google Scholar] [CrossRef] [PubMed]
  47. Guida, S.; Ciardo, S.; De Pace, B.; De Carvalho, N.; Peccerillo, F.; Manfredini, M.; Farnetani, F.; Chester, J.; Kaleci, S.; Manganelli, M.; et al. The Influence of MC1R On dermal Morphological Features of Photo-Exposed Skin in Women Revealed by Reflectance Confocal Microscopy and Optical Coherence Tomography. Exp. Dermatol. 2019, 28, 1321–1327. [Google Scholar] [CrossRef] [PubMed]
  48. Law, M.H.; Medland, S.E.; Zhu, G.; Yazar, S.; Viñuela, A.; Wallace, L.; Shekar, S.N.; Duffy, D.L.; Bataille, V.; Glass, D.; et al. Genome-Wide Association shows that Pigmentation Genes Play a Role in Skin Aging. J. Investig. Dermatol. 2017, 137, 1887–1894. [Google Scholar] [CrossRef] [PubMed]
  49. Carroll, T.D.; Wong, T.; Morris, M.K.; Di Germanio, C.; Ma, Z.; Stone, M.; Ball, E.; Fritts, L.; Rustagi, A.; Simmons, G.; et al. Administration of Vaccine-Boosted COVID-19 Convalescent Plasma to SARS-CoV-2 Infected Hamsters Decreases Virus Replication in Lungs and Hastens Resolution of the Infection Despite Transiently Enhancing Disease and Lung Pathology. bioRxiv 2023. [Google Scholar] [CrossRef]
  50. Cirri, P.; Chiarugi, P. Cancer Associated Fibroblasts: The Dark Side of the Coin. Am. J. Cancer Res. 2011, 1, 482–497. [Google Scholar] [PubMed]
  51. Bellei, B.; Picardo, M. Premature Cell Senescence in Human Skin: Dual Face in Chronic Acquired Pigmentary Disorders. Ageing Res. Rev. 2020, 57, 100981. [Google Scholar] [CrossRef] [PubMed]
  52. Swope, V.B.; Starner, R.J.; Rauck, C.; Abdel-Malek, Z.A. Endothelin-1 and A-Melanocortin have Redundant Effects on Global Genome Repair in UV-Irradiated Human Melanocytes Despite Distinct Signaling Pathways. Pigment Cell Melanoma Res. 2020, 33, 293–304. [Google Scholar] [CrossRef] [PubMed]
  53. Yin, K.; Sturm, R.A.; Smith, A.G. MC1R and NR4A Receptors in Cellular Stress and DNA Repair: Implications for UVR Protection. Exp. Dermatol. 2014, 23, 449–452. [Google Scholar] [CrossRef] [PubMed]
  54. Castejón-Griñán, M.; Herraiz, C.; Olivares, C.; Jiménez-Cervantes, C.; García-Borrón, J.C. cAMP-Independent Non-Pigmentary Actions of Variant Melanocortin 1 Receptor: AKT-Mediated Activation of Protective Responses to Oxidative DNA Damage. Oncogene 2018, 37, 3631–3646. [Google Scholar] [CrossRef] [PubMed]
  55. Bastiaens, M.T.; Huurne, J.A.C.t.; Kielich, C.; Gruis, N.A.; Westendorp, R.G.J.; Vermeer, B.J.; Bavinck, J.N.B. Melanocortin-1 Receptor Gene Variants Determine the Risk of Nonmelanoma Skin Cancer Independently of Fair Skin and Red Hair. Am. J. Hum. Genet. 2001, 68, 884–894. [Google Scholar] [CrossRef] [PubMed]
  56. Cui, Y.; Miao, Y.; Cao, L.; Guo, L.; Cui, Y.; Yan, C.; Zeng, Z.; Xu, M.; Han, T. Activation of Melanocortin-1 Receptor Signaling in Melanoma Cells Impairs T Cell Infiltration to Dampen Antitumor Immunity. Nat. Commun. 2023, 14, 5740. [Google Scholar] [CrossRef] [PubMed]
  57. Guida, M.; Strippoli, S.; Ferretta, A.; Bartolomeo, N.; Porcelli, L.; Maida, I.; Azzariti, A.; Tommasi, S.; Grieco, C.; Guida, S.; et al. Detrimental Effects of Melanocortin-1 Receptor (MC1R) Variants on the Clinical Outcomes of BRAF V600 Metastatic Melanoma Patients Treated with BRAF Inhibitors. Pigment Cell Melanoma Res. 2016, 29, 679–687. [Google Scholar] [CrossRef]
  58. Su, D.; Djureinovic, D.; Schoenfeld, D.; Marquez-Nostra, B.; Olino, K.; Jilaveanu, L.; Kluger, H. Melanocortin 1 Receptor (MC1R) Expression as a Marker of Progression in Melanoma. Res. Sq. 2023. [Google Scholar] [CrossRef]
  59. Sarasin, A.; Kauffmann, A. Overexpression of DNA Repair Genes is Associated with Metastasis: A New Hypothesis. Mutat. Res. 2008, 659, 49–55. [Google Scholar] [CrossRef]
  60. Zhang, G.; Herlyn, M. Human Nevi: No Longer Precursors of Melanomas? J. Investig. Dermatol. 2012, 132, 2133–2134. [Google Scholar] [CrossRef]
  61. Tsao, H.; Bevona, C.; Goggins, W.; Quinn, T. The Transformation Rate of Moles (Melanocytic Nevi) into Cutaneous Melanoma: A Population-Based Estimate. Arch. Dermatol. (1960) 2003, 139, 282–288. [Google Scholar] [CrossRef] [PubMed]
  62. Shreberk-Hassidim, R.; Ostrowski, S.M.; Fisher, D.E. The Complex Interplay between Nevi and Melanoma: Risk Factors and Precursors. Int. J. Mol. Sci. 2023, 24, 3541. [Google Scholar] [CrossRef] [PubMed]
  63. Pollock, P.M.; Harper, U.L.; Hansen, K.S.; Yudt, L.M.; Stark, M.; Robbins, C.M.; Moses, T.Y.; Hostetter, G.; Wagner, U.; Kakareka, J.; et al. High Frequency of BRAF Mutations in Nevi. Nat. Genet. 2003, 33, 19–20. [Google Scholar] [CrossRef] [PubMed]
  64. Requena, C.; Manrique, E.; Nagore, E. Update on Lentigo Maligna: Diagnostic Signs and Treatment. Actas Dermosifiliogr. 2023, 114, 413–424. [Google Scholar] [CrossRef] [PubMed]
  65. Brînzea, A.; Nedelcu, R.I.; Ion, D.A.; Turcu, G.; Antohe, M.; Hodorogea, A.; Călinescu, A.; Pirici, D.; Popescu, R.; Popescu, C.M.; et al. Matrix Metalloproteinases Expression in Lentigo Maligna∕lentigo Maligna Melanoma—A Review of the Literature and Personal Experience. Rom. J. Morphol. Embryol. 2019, 60, 1091–1095. [Google Scholar] [PubMed]
  66. DeWane, M.E.; Kelsey, A.; Oliviero, M.; Rabinovitz, H.; Grant-Kels, J.M. Melanoma on Chronically Sun-Damaged Skin: Lentigo Maligna and Desmoplastic Melanoma. J. Am. Acad. Dermatol. 2019, 81, 823–833. [Google Scholar] [CrossRef] [PubMed]
  67. Kharouf, N.; Flanagan, T.W.; Hassan, S.; Shalaby, H.; Khabaz, M.; Hassan, S.; Megahed, M.; Haikel, Y.; Santourlidis, S.; Hassan, M. Tumor Microenvironment as a Therapeutic Target in Melanoma Treatment. Cancers 2023, 15, 3147. [Google Scholar] [CrossRef]
  68. Zhang, G.; Ji, P.; Xia, P.; Song, H.; Guo, Z.; Hu, X.; Guo, Y.; Yuan, X.; Song, Y.; Shen, R.; et al. Identification and Targeting of Cancer-Associated Fibroblast Signature Genes for Prognosis and Therapy in Cutaneous Melanoma. Comput. Biol. Med. 2023, 167, 107597. [Google Scholar] [CrossRef]
  69. Anderson-Crannage, M.; Ascensión, A.M.; Ibanez-Solé, O.; Zhu, H.; Schaefer, E.; Ottomanelli, D.; Hochberg, B.; Pan, J.; Luo, W.; Tian, M.; et al. Inflammation-Mediated Fibroblast Activation and Immune Dysregulation in Collagen VII-Deficient Skin. Front. Immunol. 2023, 14, 1211505. [Google Scholar] [CrossRef]
  70. Wu, B.; Sodji, Q.H.; Oyelere, A.K. Inflammation, Fibrosis and Cancer: Mechanisms, Therapeutic Options and Challenges. Cancers 2022, 14, 552. [Google Scholar] [CrossRef]
  71. Bottazzi, B.; Riboli, E.; Mantovani, A. Aging, Inflammation and Cancer. Semin. Immunol. 2018, 40, 74–82. [Google Scholar] [CrossRef] [PubMed]
  72. Chin, T.; Lee, X.E.; Ng, P.Y.; Lee, Y.; Dreesen, O. The Role of Cellular Senescence in Skin Aging and Age-Related Skin Pathologies. Front. Physiol. 2023, 14, 1297637. [Google Scholar] [CrossRef] [PubMed]
  73. D’Arino, A.; Caputo, S.; Eibenschutz, L.; Piemonte, P.; Buccini, P.; Frascione, P.; Bellei, B. Skin Cancer Microenvironment: What we can Learn from Skin Aging? Int. J. Mol. Sci. 2023, 24, 14043. [Google Scholar] [CrossRef]
  74. Chatsirisupachai, K.; Lagger, C.; de Magalhães, J.P. Age-Associated Differences in the Cancer Molecular Landscape. Trends Cancer 2022, 8, 962–971. [Google Scholar] [CrossRef]
  75. Nicolas, E.; Golemis, E.A.; Arora, S. POLD1: Central Mediator of DNA Replication and Repair, and Implication in Cancer and Other Pathologies. Gene 2016, 590, 128–141. [Google Scholar] [CrossRef]
  76. Council, M.L.; Sheinbein, D.M. Common Skin Cancers in Older Adults Approach to Diagnosis and Management. Clin. Geriatr. Med. 2024, 40, 25–36. [Google Scholar] [CrossRef]
  77. Papaccio, F.; Kovacs, D.; Bellei, B.; Caputo, S.; Migliano, E.; Cota, C.; Picardo, M. Profiling Cancer-Associated Fibroblasts in Melanoma. Int. J. Mol. Sci. 2021, 22, 7255. [Google Scholar] [CrossRef]
  78. Liu, J.; Zheng, R.; Zhang, Y.; Jia, S.; He, Y.; Liu, J. The Cross Talk between Cellular Senescence and Melanoma: From Molecular Pathogenesis to Target Therapies. Cancers 2023, 15, 2640. [Google Scholar] [CrossRef]
  79. Huang, J.; Heng, S.; Zhang, W.; Liu, Y.; Xia, T.; Ji, C.; Zhang, L. Dermal Extracellular Matrix Molecules in Skin Development, Homeostasis, Wound Regeneration and Diseases. Semin. Cell Dev. Biol. 2022, 128, 137–144. [Google Scholar] [CrossRef]
  80. Tracy, L.E.; Minasian, R.A.; Caterson, E.J. Extracellular Matrix and Dermal Fibroblast Function in the Healing Wound. Adv. Wound Care 2016, 5, 119–136. [Google Scholar] [CrossRef]
  81. Driskell, R.R.; Lichtenberger, B.M.; Hoste, E.; Kretzschmar, K.; Simons, B.D.; Charalambous, M.; Ferron, S.R.; Herault, Y.; Pavlovic, G.; Ferguson-Smith, A.C.; et al. Distinct Fibroblast Lineages Determine Dermal Architecture in Skin Development and Repair. Nature 2013, 504, 277–281. [Google Scholar] [CrossRef] [PubMed]
  82. Tsepkolenko, A.; Tsepkolenko, V.; Dash, S.; Mishra, A.; Bader, A.; Melerzanov, A.; Giri, S. The Regenerative Potential of Skin and the Immune System. Clin. Cosmet. Investig. Dermatol. 2019, 12, 519–532. [Google Scholar] [CrossRef] [PubMed]
  83. Nyström, A.; Bruckner-Tuderman, L. Matrix Molecules and Skin Biology. Semin. Cell Dev. Biol. 2019, 89, 136–146. [Google Scholar] [CrossRef] [PubMed]
  84. Guerrero-Juarez, C.F.; Plikus, M.V. Emerging Nonmetabolic Functions of Skin Fat. Nat. Rev. Endocrinol. 2018, 14, 163–173. [Google Scholar] [CrossRef] [PubMed]
  85. Nicu, C.; O’Sullivan, J.D.B.; Ramos, R.; Timperi, L.; Lai, T.; Farjo, N.; Farjo, B.; Pople, J.; Bhogal, R.; Hardman, J.A.; et al. Dermal Adipose Tissue Secretes HGF to Promote Human Hair Growth and Pigmentation. J. Investig. Dermatol. 2021, 141, 1633–1645.e13. [Google Scholar] [CrossRef] [PubMed]
  86. Chi, M.; Chen, J.; Ye, Y.; Tseng, H.; Lai, F.; Tay, K.H.; Jin, L.; Guo, S.T.; Jiang, C.C.; Zhang, X.D. Adipocytes Contribute to Resistance of Human Melanoma Cells to Chemotherapy and Targeted Therapy. Curr. Med. Chem. 2014, 21, 1255–1267. [Google Scholar] [CrossRef]
  87. Kwan, H.Y.; Fu, X.; Liu, B.; Chao, X.; Chan, C.L.; Cao, H.; Su, T.; Tse, A.K.W.; Fong, W.F.; Yu, Z. Subcutaneous Adipocytes Promote Melanoma Cell Growth by Activating the Akt Signaling Pathway: Role of Palmitic Acid. J. Biol. Chem. 2014, 289, 30525–30537. [Google Scholar] [CrossRef]
  88. Zhang, M.; Di Martino, J.S.; Bowman, R.L.; Campbell, N.R.; Baksh, S.C.; Simon-Vermot, T.; Kim, I.S.; Haldeman, P.; Mondal, C.; Yong-Gonzales, V.; et al. Adipocyte-Derived Lipids Mediate Melanoma Progression Via FATP Proteins. Cancer Discov. 2018, 8, 1006–1025. [Google Scholar] [CrossRef]
  89. Okumura, T.; Ohuchida, K.; Kibe, S.; Iwamoto, C.; Ando, Y.; Takesue, S.; Nakayama, H.; Abe, T.; Endo, S.; Koikawa, K.; et al. Adipose Tissue-Derived Stromal Cells are Sources of Cancer-Associated Fibroblasts and Enhance Tumor Progression by Dense Collagen Matrix. Int. J. Cancer 2019, 144, 1401–1413. [Google Scholar] [CrossRef]
  90. Jung, Y.Y.; Lee, Y.K.; Koo, J.S. Expression of Cancer-Associated Fibroblast-Related Proteins in Adipose Stroma of Breast Cancer. Tumour Biol. 2015, 36, 8685–8695. [Google Scholar] [CrossRef]
  91. Bochet, L.; Lehuédé, C.; Dauvillier, S.; Wang, Y.Y.; Dirat, B.; Laurent, V.; Dray, C.; Guiet, R.; Maridonneau-Parini, I.; Le Gonidec, S.; et al. Adipocyte-Derived Fibroblasts Promote Tumor Progression and Contribute to the Desmoplastic Reaction in Breast Cancer. Cancer Res. 2013, 73, 5657–5668. [Google Scholar] [CrossRef]
  92. Menon, G.K. Skin Basics; Structure and Function. In Lipids and Skin Health; Springer International Publishing: Cham, Switzerland, 2014; pp. 9–23. [Google Scholar]
  93. Yousef, H.; Alhajj, M.; Sharma, S. Anatomy, Skin (Integument), Epidermis. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2023. [Google Scholar]
  94. Gdula, M.R.; Poterlowicz, K.; Mardaryev, A.N.; Sharov, A.A.; Peng, Y.; Fessing, M.Y.; Botchkarev, V.A. Remodeling of Three-Dimensional Organization of the Nucleus during Terminal Keratinocyte Differentiation in the Epidermis. J. Investig. Dermatol. 2013, 133, 2191–2201. [Google Scholar] [CrossRef]
  95. Monteleon, C.L.; Agnihotri, T.; Dahal, A.; Liu, M.; Rebecca, V.W.; Beatty, G.L.; Amaravadi, R.K.; Ridky, T.W. Lysosomes Support the Degradation, Signaling, and Mitochondrial Metabolism Necessary for Human Epidermal Differentiation. J. Investig. Dermatol. 2018, 138, 1945–1954. [Google Scholar] [CrossRef]
  96. Murata, T.; Honda, T.; Mostafa, A.; Kabashima, K. Stratum Corneum as Polymer Sheet: Concept and Cornification Processes. Trends Mol. Med. 2022, 28, 350–359. [Google Scholar] [CrossRef] [PubMed]
  97. Kolarsick, P.A.J.; Kolarsick, M.A.; Goodwin, C. Anatomy and Physiology of the Skin. J. Dermatol. Nurses’ Assoc. 2011, 3, 203–213. [Google Scholar] [CrossRef]
  98. Venus, M.; Waterman, J.; McNab, I. Basic Physiology of the Skin. Surgery 2010, 28, 469–472. [Google Scholar]
  99. Proksch, E.; Brandner, J.M.; Jensen, J. The Skin: An Indispensable Barrier. Exp. Dermatol. 2008, 17, 1063–1072. [Google Scholar] [CrossRef] [PubMed]
  100. Williams, I.R.; Kupper, T.S. Immunity at the Surface: Homeostatic Mechanisms of the Skin Immune System. Life Sci. 1996, 58, 1485–1507. [Google Scholar] [CrossRef] [PubMed]
  101. Chambers, E.S.; Vukmanovic-Stejic, M. Skin Barrier Immunity and Ageing. Immunology 2020, 160, 116–125. [Google Scholar] [CrossRef]
  102. Morizane, S.; Mukai, T.; Sunagawa, K.; Tachibana, K.; Kawakami, Y.; Ouchida, M. “Input/Output Cytokines” in Epidermal Keratinocytes and the Involvement in Inflammatory Skin Diseases. Front. Immunol. 2023, 14, 1239598. [Google Scholar] [CrossRef]
  103. Park, H.Y.; Kosmadaki, M.; Yaar, M.; Gilchrest, B.A. Cellular Mechanisms Regulating Human Melanogenesis. Cell. Mol. Life Sci. 2009, 66, 1493–1506. [Google Scholar] [CrossRef] [PubMed]
  104. Casalou, C.; Moreiras, H.; Mayatra, J.M.; Fabre, A.; Tobin, D.J. Loss of ‘Epidermal Melanin Unit’ Integrity in Human Skin during Melanoma-Genesis. Front. Oncol. 2022, 12, 878336. [Google Scholar] [CrossRef]
  105. Naik, P.P.; Farrukh, S.N. Influence of Ethnicities and Skin Color Variations in Different Populations: A Review. Skin. Pharmacol. Physiol. 2022, 35, 65–76. [Google Scholar] [CrossRef] [PubMed]
  106. Olson, R.L.; Gaylor, J.; Everett, M.A. Skin Color, Melanin, and Erythema. Arch. Dermatol. (1960) 1973, 108, 541–544. [Google Scholar] [CrossRef]
  107. Montagna, W.; Carlisle, K. The Architecture of Black and White Facial Skin. J. Am. Acad. Dermatol. 1991, 24, 929–937. [Google Scholar] [CrossRef] [PubMed]
  108. Iozumi, K.; Hoganson, G.E.; Pennella, R.; Everett, M.A.; Fuller, B.B. Role of Tyrosinase as the Determinant of Pigmentation in Cultured Human Melanocytes. J. Investig. Dermatol. 1993, 100, 806–811. [Google Scholar] [CrossRef]
  109. Markiewicz, E.; Karaman-Jurukovska, N.; Mammone, T.; Idowu, O.C. Post-Inflammatory Hyperpigmentation in Dark Skin: Molecular Mechanism and Skincare Implications. Clin. Cosmet. Investig. Dermatol. 2022, 15, 2555–2565. [Google Scholar] [CrossRef]
  110. Van Den Bossche, K.; Naeyaert, J.; Lambert, J. The Quest for the Mechanism of Melanin Transfer. Traffic 2006, 7, 769–778. [Google Scholar] [CrossRef]
  111. Bento-Lopes, L.; Cabaço, L.C.; Charneca, J.; Neto, M.V.; Seabra, M.C.; Barral, D.C. Melanin’s Journey from Melanocytes to Keratinocytes: Uncovering the Molecular Mechanisms of Melanin Transfer and Processing. Int. J. Mol. Sci. 2023, 24, 11289. [Google Scholar] [CrossRef]
  112. Costin, G.; Hearing, V.J. Human Skin Pigmentation: Melanocytes Modulate Skin Color in Response to Stress. FASEB J. 2007, 21, 976–994. [Google Scholar] [CrossRef]
  113. Cichorek, M.; Wachulska, M.; Stasiewicz, A.; Tymińska, A. Skin Melanocytes: Biology and Development. Postep. Dermatol. Alergol. 2013, 30, 30–41. [Google Scholar] [CrossRef]
  114. Nordlund, J.J. The Lives of Pigment Cells. Dermatol. Clin. 1986, 4, 407–418. [Google Scholar] [CrossRef]
  115. Gilchrest, B.A.; Blog, F.B.; Szabo, G. Effects of Aging and Chronic Sun Exposure on Melanocytes in Human Skin. J. Investig. Dermatol. 1979, 73, 141–143. [Google Scholar] [CrossRef] [PubMed]
  116. Simon, J.D.; Peles, D.; Wakamatsu, K.; Ito, S. Current Challenges in Understanding Melanogenesis: Bridging Chemistry, Biological Control, Morphology, and Function. Pigment Cell Melanoma Res. 2009, 22, 563–579. [Google Scholar] [CrossRef]
  117. Land, E.J.; Riley, P.A. Spontaneous Redox Reactions of Dopaquinone and the Balance between the Eumelanic and Phaeomelanic Pathways. Pigment Cell Res. 2000, 13, 273–277. [Google Scholar] [CrossRef] [PubMed]
  118. Hertzman Johansson, C.; Azimi, A.; Frostvik Stolt, M.; Shojaee, S.; Wiberg, H.; Grafström, E.; Hansson, J.; Egyházi Brage, S. Association of MITF and Other Melanosome-Related Proteins with Chemoresistance in Melanoma Tumors and Cell Lines. Melanoma Res. 2013, 23, 360–365. [Google Scholar] [CrossRef] [PubMed]
  119. Juntti-Berggren, L.; Lindh, U.; Berggren, P.O. Starvation is Associated with Changes in the Elemental Composition of the Pancreatic Beta-Cell. Biosci. Rep. 1991, 11, 73–84. [Google Scholar] [CrossRef] [PubMed]
  120. Kawakami, A.; Fisher, D.E. The Master Role of Microphthalmia-Associated Transcription Factor in Melanocyte and Melanoma Biology. Lab. Investig. 2017, 97, 649–656. [Google Scholar] [CrossRef] [PubMed]
  121. Slominski, R.M.; Sarna, T.; Płonka, P.M.; Raman, C.; Brożyna, A.A.; Slominski, A.T. Melanoma, Melanin, and Melanogenesis: The Yin and Yang Relationship. Front. Oncol. 2022, 12, 842496. [Google Scholar] [CrossRef] [PubMed]
  122. Hartman, M.L.; Czyz, M. MITF in Melanoma: Mechanisms Behind its Expression and Activity. Cell. Mol. Life Sci. 2015, 72, 1249–1260. [Google Scholar] [CrossRef] [PubMed]
  123. Kovacs, D.; Migliano, E.; Muscardin, L.; Silipo, V.; Catricalà, C.; Picardo, M.; Bellei, B. The Role of Wnt/Β-Catenin Signaling Pathway in Melanoma Epithelial-to-Mesenchymal-Like Switching: Evidences from Patients-Derived Cell Lines. Oncotarget 2016, 7, 43295–43314. [Google Scholar] [CrossRef]
  124. Hossain, S.M.; Eccles, M.R. Phenotype Switching and the Melanoma Microenvironment; Impact on Immunotherapy and Drug Resistance. Int. J. Mol. Sci. 2023, 24, 1601. [Google Scholar] [CrossRef]
  125. Jimbow, K.; Quevedo, W.C.; Fitzpatrick, T.B.; Szabo, G. Some Aspects of Melanin Biology: 1950–1975. J. Investig. Dermatol. 1976, 67, 72–89. [Google Scholar] [CrossRef]
  126. Hardman, M.J.; Liu, K.; Avilion, A.A.; Merritt, A.; Brennan, K.; Garrod, D.R.; Byrne, C. Desmosomal Cadherin Misexpression Alters Beta-Catenin Stability and Epidermal Differentiation. Mol. Cell. Biol. 2005, 25, 969–978. [Google Scholar] [CrossRef] [PubMed]
  127. Charest, J.L.; Jennings, J.M.; King, W.P.; Kowalczyk, A.P.; García, A.J. Cadherin-Mediated Cell-Cell Contact Regulates Keratinocyte Differentiation. J. Investig. Dermatol. 2009, 129, 564–572. [Google Scholar] [CrossRef] [PubMed]
  128. D’Arcy, C.; Kiel, C. Cell Adhesion Molecules in Normal Skin and Melanoma. Biomolecules 2021, 11, 1213. [Google Scholar] [CrossRef]
  129. Tang, A.; Eller, M.S.; Hara, M.; Yaar, M.; Hirohashi, S.; Gilchrest, B.A. E-Cadherin is the Major Mediator of Human Melanocyte Adhesion to Keratinocytes In Vitro. J. Cell Sci. 1994, 107 Pt 4, 983–992. [Google Scholar] [CrossRef]
  130. Hung, C.; Chiang, H.; Lo, H.; Jian, J.; Wu, W. E-Cadherin and its Downstream Catenins are Proteolytically Cleaved in Human HaCaT Keratinocytes Exposed to UVB. Exp. Dermatol. 2006, 15, 315–321. [Google Scholar] [CrossRef]
  131. Gambichler, T.; Rotterdam, S.; Tigges, C.; Altmeyer, P.; Bechara, F.G. Impact of Ultraviolet Radiation on the Expression of Marker Proteins of Gap and Adhesion Junctions in Human Epidermis. Photodermatol. Photoimmunol. Photomed. 2008, 24, 318–321. [Google Scholar] [CrossRef]
  132. Jamal, S.; Schneider, R.J. UV-Induction of Keratinocyte Endothelin-1 Downregulates E-Cadherin in Melanocytes and Melanoma Cells. J. Clin. Investig. 2002, 110, 443–452. [Google Scholar] [CrossRef] [PubMed]
  133. Hu, S.; Jiang, S.; Miao, F.; Lei, T. sPmel17 Secreted by Ultraviolet B-Exposed Melanocytes Alters the Intercellular Adhesion of Keratinocytes. Oxid. Med. Cell. Longev. 2022, 2022, 1856830. [Google Scholar] [CrossRef]
  134. Shain, A.H.; Bastian, B.C. From Melanocytes to Melanomas. Nat. Rev. Cancer 2016, 16, 345–358. [Google Scholar] [CrossRef]
  135. Huber, O.; Bierkamp, C.; Kemler, R. Cadherins and Catenins in Development. Curr. Opin. Cell Biol. 1996, 8, 685–691. [Google Scholar] [CrossRef]
  136. Ramani, V.; Teshima, T.; Tamura, K.; Chung, J.; Kobayashi, M.; Cruz, P.D.; Ariizumi, K. Melanoma-Derived Soluble DC-HIL/GPNMB Promotes Metastasis by Excluding T-Lymphocytes from the Pre-Metastatic Niches. J. Investig. Dermatol. 2018, 138, 2443–2451. [Google Scholar] [CrossRef]
  137. Tomihari, M.; Hwang, S.; Chung, J.; Cruz, P.D.; Ariizumi, K. Gpnmb is a Melanosome-Associated Glycoprotein that Contributes to Melanocyte/Keratinocyte Adhesion in a RGD-Dependent Fashion. Exp. Dermatol. 2009, 18, 586–595. [Google Scholar] [CrossRef] [PubMed]
  138. Biswas, K.B.; Takahashi, A.; Mizutani, Y.; Takayama, S.; Ishitsuka, A.; Yang, L.; Yang, F.; Iddamalgoda, A.; Katayama, I.; Inoue, S. GPNMB is Expressed in Human Epidermal Keratinocytes but Disappears in the Vitiligo Lesional Skin. Sci. Rep. 2020, 10, 4930. [Google Scholar] [CrossRef] [PubMed]
  139. Wang, Q.; Kuroda, Y.; Yang, L.; Lai, S.; Mizutani, Y.; Iddamalgoda, A.; Guo, J.; Yamamoto, A.; Murase, D.; Takahashi, Y.; et al. GPNMB Extracellular Fragment Protects Melanocytes from Oxidative Stress by Inhibiting AKT Phosphorylation Independent of CD44. Int. J. Mol. Sci. 2021, 22, 10843. [Google Scholar] [CrossRef]
  140. Arnette, C.R.; Roth-Carter, Q.R.; Koetsier, J.L.; Broussard, J.A.; Burks, H.E.; Cheng, K.; Amadi, C.; Gerami, P.; Johnson, J.L.; Green, K.J. Keratinocyte Cadherin Desmoglein 1 Controls Melanocyte Behavior through Paracrine Signaling. Pigment Cell Melanoma Res. 2020, 33, 305–317. [Google Scholar] [CrossRef]
  141. Hsu, M.; Andl, T.; Li, G.; Meinkoth, J.L.; Herlyn, M. Cadherin Repertoire Determines Partner-Specific Gap Junctional Communication during Melanoma Progression. J. Cell Sci. 2000, 113 Pt 9, 1535–1542. [Google Scholar] [CrossRef]
  142. Haass, N.K.; Wladykowski, E.; Kief, S.; Moll, I.; Brandner, J.M. Differential Induction of Connexins 26 and 30 in Skin Tumors and their Adjacent Epidermis. J. Histochem. Cytochem. 2006, 54, 171–182. [Google Scholar] [CrossRef] [PubMed]
  143. Zhou, J.Z.; Jiang, J.X. Gap Junction and Hemichannel-Independent Actions of Connexins on Cell and Tissue Functions—An Update. FEBS Lett. 2014, 588, 1186–1192. [Google Scholar] [CrossRef]
  144. Bellei, B.; Mastrofrancesco, A.; Briganti, S.; Aspite, N.; Ale-Agha, N.; Sies, H.; Picardo, M. Ultraviolet A Induced Modulation of Gap Junctional Intercellular Communication by P38 MAPK Activation in Human Keratinocytes. Exp. Dermatol. 2008, 17, 115–124. [Google Scholar] [CrossRef] [PubMed]
  145. Dedrick, R.L.; Walicke, P.; Garovoy, M. Anti-Adhesion Antibodies Efalizumab, a Humanized Anti-CD11a Monoclonal Antibody. Transpl. Immunol. 2002, 9, 181–186. [Google Scholar] [CrossRef] [PubMed]
  146. Upadhyay, P.R.; Ho, T.; Abdel-Malek, Z.A. Participation of Keratinocyte- and Fibroblast-Derived Factors in Melanocyte Homeostasis, the Response to UV, and Pigmentary Disorders. Pigment Cell Melanoma Res. 2021, 34, 762–776. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, J.X.; Fukunaga-Kalabis, M.; Herlyn, M. Crosstalk in Skin: Melanocytes, Keratinocytes, Stem Cells, and Melanoma. J. Cell Commun. Signal. 2016, 10, 191–196. [Google Scholar] [CrossRef]
  148. Hirobe, T. Keratinocytes Regulate the Function of Melanocytes. Dermatol. Sin. 2014, 32, 200–204. [Google Scholar] [CrossRef]
  149. Haass, N.K.; Smalley, K.S.M.; Li, L.; Herlyn, M. Adhesion, Migration and Communication in Melanocytes and Melanoma. Pigment Cell Res. 2005, 18, 150–159. [Google Scholar] [CrossRef]
  150. Imokawa, G. Autocrine and Paracrine Regulation of Melanocytes in Human Skin and in Pigmentary Disorders. Pigment Cell Res. 2004, 17, 96–110. [Google Scholar] [CrossRef]
  151. Schiller, M.; Brzoska, T.; Böhm, M.; Metze, D.; Scholzen, T.E.; Rougier, A.; Luger, T.A. Solar-Simulated Ultraviolet Radiation-Induced Upregulation of the Melanocortin-1 Receptor, Proopiomelanocortin, and Alpha-Melanocyte-Stimulating Hormone in Human Epidermis In Vivo. J. Investig. Dermatol. 2004, 122, 468–476. [Google Scholar]
  152. Swope, V.B.; Medrano, E.E.; Smalara, D.; Abdel-Malek, Z.A. Long-Term Proliferation of Human Melanocytes is Supported by the Physiologic Mitogens Alpha-Melanotropin, Endothelin-1, and Basic Fibroblast Growth Factor. Exp. Cell Res. 1995, 217, 453–459. [Google Scholar] [CrossRef]
  153. Murase, D.; Hachiya, A.; Amano, Y.; Ohuchi, A.; Kitahara, T.; Takema, Y. The Essential Role of p53 in Hyperpigmentation of the Skin Via Regulation of Paracrine Melanogenic Cytokine Receptor Signaling. J. Biol. Chem. 2009, 284, 4343–4353. [Google Scholar] [CrossRef]
  154. Lübbe, J.; Reichel, M.; Burg, G.; Kleihues, P. Absence of p53 Gene Mutations in Cutaneous Melanoma. J. Investig. Dermatol. 1994, 102, 819–821. [Google Scholar] [CrossRef]
  155. Choi, S.; Bin, B.; Kim, W.; Lee, E.; Lee, T.R.; Cho, E. Exposure of Human Melanocytes to UVB Twice and Subsequent Incubation Leads to Cellular Senescence and Senescence-Associated Pigmentation through the Prolonged p53 Expression. J. Dermatol. Sci. 2018, 90, 303–312. [Google Scholar] [CrossRef]
  156. Wu, C.; Lan, C.E.; Chiou, M.; Yu, H. Basic Fibroblast Growth Factor Promotes Melanocyte Migration Via Increased Expression of p125(FAK) on Melanocytes. Acta Derm. Venereol. 2006, 86, 498–502. [Google Scholar] [CrossRef]
  157. Shi, H.; Lin, B.; Huang, Y.; Wu, J.; Zhang, H.; Lin, C.; Wang, Z.; Zhu, J.; Zhao, Y.; Fu, X.; et al. Basic Fibroblast Growth Factor Promotes Melanocyte Migration Via Activating PI3K/Akt-Rac1-FAK-JNK and ERK Signaling Pathways. IUBMB Life 2016, 68, 735–747. [Google Scholar] [CrossRef]
  158. Halaban, R.; Rubin, J.S.; Funasaka, Y.; Cobb, M.; Boulton, T.; Faletto, D.; Rosen, E.; Chan, A.; Yoko, K.; White, W. Met and Hepatocyte Growth Factor/Scatter Factor Signal Transduction in Normal Melanocytes and Melanoma Cells. Oncogene 1992, 7, 2195–2206. [Google Scholar]
  159. Weidner, K.M.; Di Cesare, S.; Sachs, M.; Brinkmann, V.; Behrens, J.; Birchmeier, W. Interaction between Gab1 and the C-Met Receptor Tyrosine Kinase is Responsible for Epithelial Morphogenesis. Nature 1996, 384, 173–176. [Google Scholar] [CrossRef] [PubMed]
  160. Ponzetto, C.; Bardelli, A.; Zhen, Z.; Maina, F.; dalla Zonca, P.; Giordano, S.; Graziani, A.; Panayotou, G.; Comoglio, P.M. A Multifunctional Docking Site Mediates Signaling and Transformation by the Hepatocyte Growth Factor/Scatter Factor Receptor Family. Cell 1994, 77, 261–271. [Google Scholar] [CrossRef] [PubMed]
  161. Mildner, M.; Mlitz, V.; Gruber, F.; Wojta, J.; Tschachler, E. Hepatocyte Growth Factor Establishes Autocrine and Paracrine Feedback Loops for the Protection of Skin Cells After UV Irradiation. J. Investig. Dermatol. 2007, 127, 2637–2644. [Google Scholar] [CrossRef] [PubMed]
  162. Grichnik, J.M.; Burch, J.A.; Burchette, J.; Shea, C.R. The SCF/KIT Pathway Plays a Critical Role in the Control of Normal Human Melanocyte Homeostasis. J. Investig. Dermatol. 1998, 111, 233–238. [Google Scholar] [CrossRef] [PubMed]
  163. Scott, G.; Ewing, J.; Ryan, D.; Abboud, C. Stem Cell Factor Regulates Human Melanocyte-Matrix Interactions. Pigment Cell Res. 1994, 7, 44–51. [Google Scholar] [CrossRef]
  164. Imokawa, G.; Yada, Y.; Miyagishi, M. Endothelins Secreted from Human Keratinocytes are Intrinsic Mitogens for Human Melanocytes. J. Biol. Chem. 1992, 267, 24675–24680. [Google Scholar] [CrossRef]
  165. Imokawa, G.; Yada, Y.; Kimura, M. Signalling Mechanisms of Endothelin-Induced Mitogenesis and Melanogenesis in Human Melanocytes. Biochem. J. 1996, 314 Pt 1, 305–312. [Google Scholar] [CrossRef] [PubMed]
  166. Ujfaludi, Z.; Tuzesi, A.; Majoros, H.; Rothler, B.; Pankotai, T.; Boros, I.M. Coordinated Activation of a Cluster of MMP Genes in Response to UVB Radiation. Sci. Rep. 2018, 8, 2660. [Google Scholar] [CrossRef]
  167. Yoshihisa, Y.; Norisugi, O.; Matsunaga, K.; Nishihira, J.; Shimizu, T. Involvement of MIF in Basement Membrane Damage in Chronically UVB-Exposed Skin in Mice. PLoS ONE 2014, 9, e89569. [Google Scholar] [CrossRef] [PubMed]
  168. Purwar, R.; Kraus, M.; Werfel, T.; Wittmann, M. Modulation of Keratinocyte-Derived MMP-9 by IL-13: A Possible Role for the Pathogenesis of Epidermal Inflammation. J. Investig. Dermatol. 2008, 128, 59–66. [Google Scholar] [CrossRef]
  169. Wang, X.; Bi, Z.; Chu, W.; Wan, Y. IL-1 Receptor Antagonist Attenuates MAP Kinase/AP-1 Activation and MMP1 Expression in UVA-Irradiated Human Fibroblasts Induced by Culture Medium from UVB-Irradiated Human Skin Keratinocytes. Int. J. Mol. Med. 2005, 16, 1117–1124. [Google Scholar] [CrossRef]
  170. Lee, M.J.; Oh, J.; Park, C.; Kim, K.H.; Lee, D.H.; Chung, J.H. Galanin Contributes to Ultraviolet Irradiation-Induced Inflammation in Human Skin. Exp. Dermatol. 2017, 26, 744–747. [Google Scholar] [CrossRef]
  171. Harsha, A.; Stojadinovic, O.; Brem, H.; Sehara-Fujisawa, A.; Wewer, U.; Loomis, C.A.; Blobel, C.P.; Tomic-Canic, M. ADAM12: A Potential Target for the Treatment of Chronic Wounds. J. Mol. Med. 2008, 86, 961–969. [Google Scholar] [CrossRef]
  172. Oh, S.T.; Schramme, A.; Stark, A.; Tilgen, W.; Gutwein, P.; Reichrath, J. Overexpression of ADAM 10 and ADAM 12 in Lesional Psoriatic Skin. Br. J. Dermatol. 2008, 158, 1371–1373. [Google Scholar] [CrossRef]
  173. Abbes, A.; Zayani, Y.; Zidi, W.; Hammami, M.B.; Mebazaa, A.; El Euch, D.; Ben Ammar, A.; Sanhaji, H.; El May, M.V.; Mokni, M.; et al. Matrix Metalloproteinase-7 could be a Predictor for Acute Inflammation in Psoriatic Patients. Cytokine 2020, 134, 155195. [Google Scholar] [CrossRef]
  174. Suomela, S.; Kariniemi, A.L.; Snellman, E.; Saarialho-Kere, U. Metalloelastase (MMP-12) and 92-kDa Gelatinase (MMP-9) as Well as their Inhibitors, TIMP-1 and -3, are Expressed in Psoriatic Lesions. Exp. Dermatol. 2001, 10, 175–183. [Google Scholar] [CrossRef]
  175. Boukhedouni, N.; Martins, C.; Darrigade, A.; Drullion, C.; Rambert, J.; Barrault, C.; Garnier, J.; Jacquemin, C.; Thiolat, D.; Lucchese, F.; et al. Type-1 Cytokines Regulate MMP-9 Production and E-Cadherin Disruption to Promote Melanocyte Loss in Vitiligo. JCI Insight 2020, 5, e133772. [Google Scholar]
  176. Su, M.; Miao, F.; Jiang, S.; Shi, Y.; Luo, L.; He, X.; Wan, J.; Xu, S.; Lei, T. Role of the p53-TRPM1/miR-211-MMP9 Axis in UVB-induced Human Melanocyte Migration and its Potential in Repigmentation. Int. J. Mol. Med. 2020, 45, 1017–1026. [Google Scholar] [CrossRef] [PubMed]
  177. Kumar, R.; Parsad, D.; Kanwar, A.J.; Kaul, D. Altered Levels of Ets-1 Transcription Factor and Matrix Metalloproteinases in Melanocytes from Patients with Vitiligo. Br. J. Dermatol. 2011, 165, 285–291. [Google Scholar] [CrossRef] [PubMed]
  178. Valyi-Nagy, I.T.; Hirka, G.; Jensen, P.J.; Shih, I.M.; Juhasz, I.; Herlyn, M. Undifferentiated Keratinocytes Control Growth, Morphology, and Antigen Expression of Normal Melanocytes through Cell-Cell Contact. Lab. Investig. 1993, 69, 152–159. [Google Scholar]
  179. Le Varlet, B.; Chaudagne, C.; Saunois, A.; Barré, P.; Sauvage, C.; Berthouloux, B.; Meybeck, A.; Dumas, M.; Bonté, F. Age-Related Functional and Structural Changes in Human Dermo-Epidermal Junction Components. J. Investig. Dermatol. Symp. Proc. 1998, 3, 172–179. [Google Scholar] [CrossRef]
  180. Craven, N.M.; Watson, R.E.; Jones, C.J.; Shuttleworth, C.A.; Kielty, C.M.; Griffiths, C.E. Clinical Features of Photodamaged Human Skin are Associated with a Reduction in Collagen VII. Br. J. Dermatol. 1997, 137, 344–350. [Google Scholar] [CrossRef]
  181. Bosset, S.; Bonnet-Duquennoy, M.; Barré, P.; Chalon, A.; Lazou, K.; Kurfurst, R.; Bonté, F.; Schnébert, S.; Disant, F.; Le Varlet, B.; et al. Decreased Expression of Keratinocyte Beta1 Integrins in Chronically Sun-Exposed Skin In Vivo. Br. J. Dermatol. 2003, 148, 770–778. [Google Scholar] [CrossRef] [PubMed]
  182. Zhang, S.; Duan, E. Fighting Against Skin Aging: The Way from Bench to Bedside. Cell Transplant. 2018, 27, 729–738. [Google Scholar] [CrossRef] [PubMed]
  183. Miskolczi, Z.; Smith, M.P.; Rowling, E.J.; Ferguson, J.; Barriuso, J.; Wellbrock, C. Collagen Abundance Controls Melanoma Phenotypes through Lineage-Specific Microenvironment Sensing. Oncogene 2018, 37, 3166–3182. [Google Scholar] [CrossRef]
  184. Kirkpatrick, S.J.; Wang, R.K.; Duncan, D.D.; Kulesz-Martin, M.; Lee, K. Imaging the Mechanical Stiffness of Skin Lesions by in Vivo Acousto-Optical Elastography. Opt. Express 2006, 14, 9770–9779. [Google Scholar] [CrossRef]
  185. Naylor, E.C.; Watson, R.E.B.; Sherratt, M.J. Molecular Aspects of Skin Ageing. Maturitas 2011, 69, 249–256. [Google Scholar] [CrossRef] [PubMed]
  186. Taloni, A.; Alemi, A.A.; Ciusani, E.; Sethna, J.P.; Zapperi, S.; La Porta, C.A.M. Mechanical Properties of Growing Melanocytic Nevi and the Progression to Melanoma. PLoS ONE 2014, 9, e94229. [Google Scholar] [CrossRef] [PubMed]
  187. Napoli, S.; Scuderi, C.; Gattuso, G.; Bella, V.D.; Candido, S.; Basile, M.S.; Libra, M.; Falzone, L. Functional Roles of Matrix Metalloproteinases and their Inhibitors in Melanoma. Cells 2020, 9, 1151. [Google Scholar] [CrossRef] [PubMed]
  188. Müller, M.; Beck, I.M.; Gadesmann, J.; Karschuk, N.; Paschen, A.; Proksch, E.; Djonov, V.; Reiss, K.; Sedlacek, R. MMP19 is Upregulated during Melanoma Progression and Increases Invasion of Melanoma Cells. Mod. Pathol. 2010, 23, 511–521. [Google Scholar] [CrossRef]
  189. Hofmann, U.B.; Westphal, J.R.; Zendman, A.J.; Becker, J.C.; Ruiter, D.J.; van Muijen, G.N. Expression and Activation of Matrix Metalloproteinase-2 (MMP-2) and its Co-Localization with Membrane-Type 1 Matrix Metalloproteinase (MT1-MMP) Correlate with Melanoma Progression. J. Pathol. 2000, 191, 245–256. [Google Scholar] [CrossRef]
  190. Salemi, R.; Falzone, L.; Madonna, G.; Polesel, J.; Cinà, D.; Mallardo, D.; Ascierto, P.A.; Libra, M.; Candido, S. MMP-9 as a Candidate Marker of Response to BRAF Inhibitors in Melanoma Patients with BRAFV600E Mutation Detected in Circulating-Free DNA. Front. Pharmacol. 2018, 9, 856. [Google Scholar] [CrossRef]
  191. Guarneri, C.; Bevelacqua, V.; Polesel, J.; Falzone, L.; Cannavò, P.S.; Spandidos, D.A.; Malaponte, G.; Libra, M. NF-κB Inhibition is Associated with OPN/MMP-9 Downregulation in Cutaneous Melanoma. Oncol. Rep. 2017, 37, 737–746. [Google Scholar] [CrossRef]
  192. Frank, A.; David, V.; Aurelie, T.; Florent, G.; William, H.; Philippe, B. Regulation of MMPs during Melanoma Progression: From Genetic to Epigenetic. Anticancer Agents Med. Chem. 2012, 12, 773–782. [Google Scholar] [CrossRef]
  193. Moustakas, A. TGF-Beta Targets PAX3 to Control Melanocyte Differentiation. Dev. Cell 2008, 15, 797–799. [Google Scholar] [CrossRef]
  194. Brenner, M.; Degitz, K.; Besch, R.; Berking, C. Differential Expression of Melanoma-Associated Growth Factors in Keratinocytes and Fibroblasts by Ultraviolet A and Ultraviolet B Radiation. Br. J. Dermatol. 2005, 153, 733–739. [Google Scholar] [CrossRef]
  195. Lee, S.B.; Schramme, A.; Doberstein, K.; Dummer, R.; Abdel-Bakky, M.S.; Keller, S.; Altevogt, P.; Oh, S.T.; Reichrath, J.; Oxmann, D.; et al. ADAM10 is Upregulated in Melanoma Metastasis Compared with Primary Melanoma. J. Investig. Dermatol. 2010, 130, 763–773. [Google Scholar] [CrossRef]
  196. Kawaguchi, M.; Hearing, V.J. The Roles of ADAMs Family Proteinases in Skin Diseases. Enzym. Res. 2011, 2011, 482498. [Google Scholar] [CrossRef] [PubMed]
  197. Mazurkiewicz, J.; Simiczyjew, A.; Dratkiewicz, E.; Kot, M.; Pietraszek-Gremplewicz, K.; Wilk, D.; Ziętek, M.; Matkowski, R.; Nowak, D. Melanoma Stimulates the Proteolytic Activity of HaCaT Keratinocytes. Cell Commun. Signal. 2022, 20, 146. [Google Scholar] [CrossRef]
  198. Loh, C.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef]
  199. Karim, R.; Tse, G.; Putti, T.; Scolyer, R.; Lee, S. The Significance of the Wnt Pathway in the Pathology of Human Cancers. Pathology 2004, 36, 120–128. [Google Scholar] [CrossRef] [PubMed]
  200. Dorsky, R.I.; Raible, D.W.; Moon, R.T. Direct Regulation of Nacre, a Zebrafish MITF Homolog Required for Pigment Cell Formation, by the Wnt Pathway. Genes Dev. 2000, 14, 158–162. [Google Scholar] [CrossRef]
  201. Bellei, B.; Pitisci, A.; Catricalà, C.; Larue, L.; Picardo, M. Wnt/Β-Catenin Signaling is Stimulated by A-Melanocyte-Stimulating Hormone in Melanoma and Melanocyte Cells: Implication in Cell Differentiation. Pigment Cell Melanoma Res. 2011, 24, 309–325. [Google Scholar] [CrossRef]
  202. Gallagher, S.J.; Rambow, F.; Kumasaka, M.; Champeval, D.; Bellacosa, A.; Delmas, V.; Larue, L. Beta-Catenin Inhibits Melanocyte Migration but Induces Melanoma Metastasis. Oncogene 2013, 32, 2230–2238. [Google Scholar] [CrossRef] [PubMed]
  203. Saito, H.; Yasumoto, K.; Takeda, K.; Takahashi, K.; Yamamoto, H.; Shibahara, S. Microphthalmia-Associated Transcription Factor in the Wnt Signaling Pathway. Pigment Cell Res. 2003, 16, 261–265. [Google Scholar] [CrossRef]
  204. Untiveros, G.; Dezi, L.; Gillette, M.; Sidor, J.; Strizzi, L. Normal Skin Cells Increase Aggressiveness of Cutaneous Melanoma by Promoting Epithelial-to-Mesenchymal Transition Via Nodal and Wnt Activity. Int. J. Mol. Sci. 2021, 22, 11719. [Google Scholar] [CrossRef]
  205. Chien, A.J.; Moore, E.C.; Lonsdorf, A.S.; Kulikauskas, R.M.; Rothberg, B.G.; Berger, A.J.; Major, M.B.; Hwang, S.T.; Rimm, D.L.; Moon, R.T. Activated Wnt/Beta-Catenin Signaling in Melanoma is Associated with Decreased Proliferation in Patient Tumors and a Murine Melanoma Model. Proc. Natl. Acad. Sci. USA 2009, 106, 1193–1198. [Google Scholar] [CrossRef]
  206. Widlund, H.R.; Horstmann, M.A.; Price, E.R.; Cui, J.; Lessnick, S.L.; Wu, M.; He, X.; Fisher, D.E. Beta-Catenin-Induced Melanoma Growth Requires the Downstream Target Microphthalmia-Associated Transcription Factor. J. Cell Biol. 2002, 158, 1079–1087. [Google Scholar] [CrossRef]
  207. Arozarena, I.; Bischof, H.; Gilby, D.; Belloni, B.; Dummer, R.; Wellbrock, C. In Melanoma, Beta-Catenin is a Suppressor of Invasion. Oncogene 2011, 30, 4531–4543. [Google Scholar] [CrossRef]
  208. Bachmann, I.M.; Straume, O.; Puntervoll, H.E.; Kalvenes, M.B.; Akslen, L.A. Importance of P-Cadherin, Beta-Catenin, and Wnt5a/Frizzled for Progression of Melanocytic Tumors and Prognosis in Cutaneous Melanoma. Clin. Cancer Res. 2005, 11, 8606–8614. [Google Scholar] [CrossRef] [PubMed]
  209. Zhou, Y.; Wang, X.; Pang, D.; Wang, Y.; Bai, J.; Tian, F.; Han, D.; Shi, S.; Hu, L. Nomogram Incorporating the WNT/Β-Catenin Signaling Pathway for Predicting the Survival of Cutaneous Melanoma. Int. J. Gen. Med. 2021, 14, 2751–2761. [Google Scholar] [CrossRef] [PubMed]
  210. Johnson, J.L.; Koetsier, J.L.; Sirico, A.; Agidi, A.T.; Antonini, D.; Missero, C.; Green, K.J. The Desmosomal Protein Desmoglein 1 Aids Recovery of Epidermal Differentiation After Acute UV Light Exposure. J. Investig. Dermatol. 2014, 134, 2154–2162. [Google Scholar] [CrossRef] [PubMed]
  211. El Kharbili, M.; Cario, M.; Béchetoille, N.; Pain, C.; Boucheix, C.; Degoul, F.; Masse, I.; Berthier-Vergnes, O. Tspan8 Drives Melanoma Dermal Invasion by Promoting ProMMP-9 Activation and Basement Membrane Proteolysis in a Keratinocyte-Dependent Manner. Cancers 2020, 12, 1297. [Google Scholar] [CrossRef]
  212. Berthier-Vergnes, O.; Barbollat-Boutrand, L.; Pommier, R.M.; de la Fouchardière, A.; Combemale, P.; Grimont, M.; Lopez-Ramirez, N.; Caramel, J.; Dalle, S.; Perrot, J.; et al. Tetraspanin8 Expression Predicts an Increased Metastatic Risk and is Associated with Cancer-Related Death in Human Cutaneous Melanoma. Mol. Cancer 2021, 20, 127. [Google Scholar] [CrossRef]
  213. Navarrete, M.; Salazar-Onfray, F.; Tittarelli, A. Flow Cytometry Evaluation of Gap Junction-Mediated Intercellular Communication between Cytotoxic T Cells and Target Tumor Cells. Methods Mol. Biol. 2021, 2346, 225–236. [Google Scholar]
  214. Tittarelli, A.; Mendoza-Naranjo, A.; Farías, M.; Guerrero, I.; Ihara, F.; Wennerberg, E.; Riquelme, S.; Gleisner, A.; Kalergis, A.; Lundqvist, A.; et al. Gap Junction Intercellular Communications Regulate NK Cell Activation and Modulate NK Cytotoxic Capacity. J. Immunol. 2014, 192, 1313–1319. [Google Scholar] [CrossRef] [PubMed]
  215. Mendoza-Naranjo, A.; Cormie, P.; Serrano, A.E.; Wang, C.M.; Thrasivoulou, C.; Sutcliffe, J.E.S.; Gilmartin, D.J.; Tsui, J.; Serena, T.E.; Phillips, A.R.J.; et al. Overexpression of the Gap Junction Protein Cx43 as found in Diabetic Foot Ulcers can Retard Fibroblast Migration. Cell Biol. Int. 2012, 36, 661–667. [Google Scholar] [CrossRef]
  216. Trosko, J.E.; Ruch, R.J. Cell-Cell Communication in Carcinogenesis. Front. Biosci. 1998, 3, 208. [Google Scholar] [CrossRef]
  217. Tittarelli, A.; Guerrero, I.; Tempio, F.; Gleisner, M.A.; Avalos, I.; Sabanegh, S.; Ortíz, C.; Michea, L.; López, M.N.; Mendoza-Naranjo, A.; et al. Overexpression of Connexin 43 Reduces Melanoma Proliferative and Metastatic Capacity. Br. J. Cancer 2015, 113, 259–267. [Google Scholar] [CrossRef]
  218. Scatolini, M.; Patel, A.; Grosso, E.; Mello-Grand, M.; Ostano, P.; Coppo, R.; Vitiello, M.; Venesio, T.; Zaccagna, A.; Pisacane, A.; et al. GJB5 Association with BRAF Mutation and Survival in Cutaneous Malignant Melanoma. Br. J. Dermatol. 2022, 186, 117–128. [Google Scholar] [CrossRef] [PubMed]
  219. Läubli, H.; Borsig, L. Selectins Promote Tumor Metastasis. Semin. Cancer Biol. 2010, 20, 169–177. [Google Scholar] [CrossRef]
  220. Shih, I.M.; Elder, D.E.; Hsu, M.Y.; Herlyn, M. Regulation of Mel-CAM/MUC18 Expression on Melanocytes of Different Stages of Tumor Progression by Normal Keratinocytes. Am. J. Pathol. 1994, 145, 837–845. [Google Scholar]
  221. Brose, M.S.; Volpe, P.; Feldman, M.; Kumar, M.; Rishi, I.; Gerrero, R.; Einhorn, E.; Herlyn, M.; Minna, J.; Nicholson, A.; et al. BRAF and RAS Mutations in Human Lung Cancer and Melanoma. Cancer Res. 2002, 62, 6997–7000. [Google Scholar]
  222. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF Gene in Human Cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
  223. Stefanato, C.M.; Yaar, M.; Bhawan, J.; Phillips, T.J.; Kosmadaki, M.G.; Botchkarev, V.; Gilchrest, B.A. Modulations of Nerve Growth Factor and Bcl-2 in Ultraviolet-Irradiated Human Epidermis. J. Cutan. Pathol. 2003, 30, 351–357. [Google Scholar] [CrossRef]
  224. Li, G.; Schaider, H.; Satyamoorthy, K.; Hanakawa, Y.; Hashimoto, K.; Herlyn, M. Downregulation of E-Cadherin and Desmoglein 1 by Autocrine Hepatocyte Growth Factor during Melanoma Development. Oncogene 2001, 20, 8125–8135. [Google Scholar] [CrossRef] [PubMed]
  225. Noujarède, J.; Carrié, L.; Garcia, V.; Grimont, M.; Eberhardt, A.; Mucher, E.; Genais, M.; Schreuder, A.; Carpentier, S.; Ségui, B.; et al. Sphingolipid Paracrine Signaling Impairs Keratinocyte Adhesion to Promote Melanoma Invasion. Cell Rep. 2023, 42, 113586. [Google Scholar] [CrossRef]
  226. Albinet, V.; Bats, M.-L.; Huwiler, A.; Rochaix, P.; Chevreau, C.; Ségui, B.; Levade, T.; Andrieu-Abadie, N. Dual Role of Sphingosine Kinase-1 in Promoting the Differentiation of Dermal Fibroblasts and the Dissemination of Melanoma Cells. Oncogene 2014, 33, 3364–3373. [Google Scholar] [CrossRef] [PubMed]
  227. Mancianti, M.L.; Herlyn, M.; Weil, D.; Jambrosic, J.; Rodeck, U.; Becker, D.; Diamond, L.; Clark, W.H.; Koprowski, H. Growth and Phenotypic Characteristics of Human Nevus Cells in Culture. J. Investig. Dermatol. 1988, 90, 134–141. [Google Scholar] [CrossRef]
  228. Michaloglou, C.; Vredeveld, L.C.W.; Soengas, M.S.; Denoyelle, C.; Kuilman, T.; van der Horst, C.M.A.M.; Majoor, D.M.; Shay, J.W.; Mooi, W.J.; Peeper, D.S. BRAFE600-Associated Senescence-Like Cell Cycle Arrest of Human Naevi. Nature 2005, 436, 720–724. [Google Scholar] [CrossRef]
  229. Sadangi, S.; Milosavljevic, K.; Castro-Perez, E.; Lares, M.; Singh, M.; Altameemi, S.; Beebe, D.J.; Ayuso, J.M.; Setaluri, V. Role of the Skin Microenvironment in Melanomagenesis: Epidermal Keratinocytes and Dermal Fibroblasts Promote BRAF Oncogene-Induced Senescence Escape in Melanocytes. Cancers 2022, 14, 1233. [Google Scholar] [CrossRef] [PubMed]
  230. Tagore, M.; Hergenreder, E.; Perlee, S.C.; Cruz, N.M.; Menocal, L.; Suresh, S.; Chan, E.; Baron, M.; Melendez, S.; Dave, A.; et al. GABA Regulates Electrical Activity and Tumor Initiation in Melanoma. Cancer Discov. 2023, 13, 2270–2291. [Google Scholar] [CrossRef] [PubMed]
  231. Golan, T.; Messer, A.R.; Amitai-Lange, A.; Melamed, Z.; Ohana, R.; Bell, R.E.; Kapitansky, O.; Lerman, G.; Greenberger, S.; Khaled, M.; et al. Interactions of Melanoma Cells with Distal Keratinocytes Trigger Metastasis Via Notch Signaling Inhibition of MITF. Mol. Cell 2015, 59, 664–676. [Google Scholar] [CrossRef]
  232. Hou, J.; Karin, M.; Sun, B. Targeting Cancer-Promoting Inflammation—Have Anti-Inflammatory Therapies Come of Age? Nat. Rev. Clin. Oncol. 2021, 18, 261–279. [Google Scholar] [CrossRef]
  233. Sheng, Y.; Liu, J.; Zhang, M.; Zheng, S. Unveiling the Link between Inflammasomes and Skin Cutaneous Melanoma: Insights into Expression Patterns and Immunotherapy Response Prediction. Math. Biosci. Eng. 2023, 20, 19912–19928. [Google Scholar] [CrossRef] [PubMed]
  234. Kučera, J.; Strnadová, K.; Dvořánková, B.; Lacina, L.; Krajsová, I.; Štork, J.; Kovářová, H.; Skalníková, H.K.; Vodička, P.; Motlík, J.; et al. Serum Proteomic Analysis of Melanoma Patients with Immunohistochemical Profiling of Primary Melanomas and Cultured Cells: Pilot Study. Oncol. Rep. 2019, 42, 1793–1804. [Google Scholar] [CrossRef] [PubMed]
  235. Smatlik, N.; Drexler, S.K.; Burian, M.; Röcken, M.; Yazdi, A.S. ASC Speck Formation After Inflammasome Activation in Primary Human Keratinocytes. Oxid. Med. Cell. Longev. 2021, 2021, 7914829. [Google Scholar] [CrossRef] [PubMed]
  236. Liu, L.; Awoyemi, A.A.; Fahy, K.E.; Thapa, P.; Borchers, C.; Wu, B.Y.; McGlone, C.L.; Schmeusser, B.; Sattouf, Z.; Rohan, C.A.; et al. Keratinocyte-Derived Microvesicle Particles Mediate Ultraviolet B Radiation-Induced Systemic Immunosuppression. J. Clin. Investig. 2021, 131, e144963. [Google Scholar] [CrossRef]
  237. Katiyar, S.K. UV-Induced Immune Suppression and Photocarcinogenesis: Chemoprevention by Dietary Botanical Agents. Cancer Lett. 2007, 255, 1–11. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of skin and epidermal–melanin unit. (A) The skin structure is shaped in three layers: epidermis, the top layer; dermis, the middle layer; hypodermis, the bottom or fatty layer. The principal cells of the epidermis are keratinocytes at different levels of differentiation. The epidermis, composed of several connected cell sheets, is subdivided into four layers: the stratum basale (the deepest portion of the epidermis), stratum spinosum, stratum granulosum, stratum lucidum, and stratum corneum. Immediately below the epidermis is the basal membrane, a specialized structure that lies between the epidermis and dermis. The dermis is made of an abundant extracellular matrix composed of an interconnected mesh of elastin collagens produced by mesenchymal cells. The dermis also contains nerves and blood vessels. The hypodermis layer contains fat cells for energy reserve, additional connective tissue, and larger blood vessels. It gives elasticity and strength to the skin. (B) Details of the epidermal–melanin unit. The epidermal–melanin unit denotes the symbiotic relationship between a melanocyte and a group of associated keratinocytes. Melanocytes are highly dendritic, pigment-producing cells located in the deeper basal layer. The pigment is synthesized and packaged within specialized lysosome-derived organelles named melanosome and transferred to neighboring keratinocytes after movement across melanocyte dendrites. Keratinocytes accumulate melanin perinuclear as a cap around the nucleus. Within the epidermis, there are also sparse immune cells like Langerhans cells, Natural killer cells, and lymphocytes.
Figure 1. Schematic representation of skin and epidermal–melanin unit. (A) The skin structure is shaped in three layers: epidermis, the top layer; dermis, the middle layer; hypodermis, the bottom or fatty layer. The principal cells of the epidermis are keratinocytes at different levels of differentiation. The epidermis, composed of several connected cell sheets, is subdivided into four layers: the stratum basale (the deepest portion of the epidermis), stratum spinosum, stratum granulosum, stratum lucidum, and stratum corneum. Immediately below the epidermis is the basal membrane, a specialized structure that lies between the epidermis and dermis. The dermis is made of an abundant extracellular matrix composed of an interconnected mesh of elastin collagens produced by mesenchymal cells. The dermis also contains nerves and blood vessels. The hypodermis layer contains fat cells for energy reserve, additional connective tissue, and larger blood vessels. It gives elasticity and strength to the skin. (B) Details of the epidermal–melanin unit. The epidermal–melanin unit denotes the symbiotic relationship between a melanocyte and a group of associated keratinocytes. Melanocytes are highly dendritic, pigment-producing cells located in the deeper basal layer. The pigment is synthesized and packaged within specialized lysosome-derived organelles named melanosome and transferred to neighboring keratinocytes after movement across melanocyte dendrites. Keratinocytes accumulate melanin perinuclear as a cap around the nucleus. Within the epidermis, there are also sparse immune cells like Langerhans cells, Natural killer cells, and lymphocytes.
Cancers 16 00913 g001
Figure 2. Cell interaction in normal versus pathological skin. (A) In healthy skin, epidermal cells are tightly interconnected. Through their dendrites, melanocytes interact with about 36–40 keratinocytes. The integral basal membrane preserves the separation of dermal and epidermal compartments. The dermis is made of abundant extracellular matrix produced by normal tissue-associated fibroblasts (NAF). (B) In pathological skin, for example during melanoma development, a loss of connections takes place between melanocytes with keratinocytes and basal membrane. During melanocytic abnormal proliferation, the entire cutis undergoes progressive structural and functional rearrangement. If this process is limited to a group of cells the ultimate lesion consists of nevomelanocytes presenting features of senescent cells. Normal melanocyte mutates into melanoma cells in a multi-step process. The uncontrolled horizontal or radial growth phase is the first step towards the invasive phenotype, in which melanocytes display a hyperproliferative phenotype and undergo alterations that offer a survival advantage. This is followed by a vertical growth phase, in which tumor cells deeply invade the dermis. This step implies damage to the basal membrane. Hyperproliferative melanocytic cells escape keratinocyte control and instead interact with fibroblasts and other hyperproliferative melanocytic cells. Melanoma progressively shapes the surrounding microenvironment in a more favorable milieu, a process that is mostly due to the transformation of NAF (light green) in cancer-associated fibroblasts CAF (dark green).
Figure 2. Cell interaction in normal versus pathological skin. (A) In healthy skin, epidermal cells are tightly interconnected. Through their dendrites, melanocytes interact with about 36–40 keratinocytes. The integral basal membrane preserves the separation of dermal and epidermal compartments. The dermis is made of abundant extracellular matrix produced by normal tissue-associated fibroblasts (NAF). (B) In pathological skin, for example during melanoma development, a loss of connections takes place between melanocytes with keratinocytes and basal membrane. During melanocytic abnormal proliferation, the entire cutis undergoes progressive structural and functional rearrangement. If this process is limited to a group of cells the ultimate lesion consists of nevomelanocytes presenting features of senescent cells. Normal melanocyte mutates into melanoma cells in a multi-step process. The uncontrolled horizontal or radial growth phase is the first step towards the invasive phenotype, in which melanocytes display a hyperproliferative phenotype and undergo alterations that offer a survival advantage. This is followed by a vertical growth phase, in which tumor cells deeply invade the dermis. This step implies damage to the basal membrane. Hyperproliferative melanocytic cells escape keratinocyte control and instead interact with fibroblasts and other hyperproliferative melanocytic cells. Melanoma progressively shapes the surrounding microenvironment in a more favorable milieu, a process that is mostly due to the transformation of NAF (light green) in cancer-associated fibroblasts CAF (dark green).
Cancers 16 00913 g002
Figure 3. Concise representation of melanocyte–keratinocyte interaction during melanomagenesis. The release of several soluble factors normally released by keratinocytes for paracrine functions (A) are up-regulated during melanomagenesis (B). Among these factors, mitogens and inflammatory mediators play the most relevant role. MMPs, ADAMs and TIMPs secreted by melanoma and stromal cells are responsible of tissue remodeling including basal membrane (BM) invasion and the entrance in the dermis where fibroblasts assume CAF phenotype.
Figure 3. Concise representation of melanocyte–keratinocyte interaction during melanomagenesis. The release of several soluble factors normally released by keratinocytes for paracrine functions (A) are up-regulated during melanomagenesis (B). Among these factors, mitogens and inflammatory mediators play the most relevant role. MMPs, ADAMs and TIMPs secreted by melanoma and stromal cells are responsible of tissue remodeling including basal membrane (BM) invasion and the entrance in the dermis where fibroblasts assume CAF phenotype.
Cancers 16 00913 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Marrapodi, R.; Bellei, B. The Keratinocyte in the Picture Cutaneous Melanoma Microenvironment. Cancers 2024, 16, 913. https://doi.org/10.3390/cancers16050913

AMA Style

Marrapodi R, Bellei B. The Keratinocyte in the Picture Cutaneous Melanoma Microenvironment. Cancers. 2024; 16(5):913. https://doi.org/10.3390/cancers16050913

Chicago/Turabian Style

Marrapodi, Ramona, and Barbara Bellei. 2024. "The Keratinocyte in the Picture Cutaneous Melanoma Microenvironment" Cancers 16, no. 5: 913. https://doi.org/10.3390/cancers16050913

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

Article Metrics

Back to TopTop