Next Article in Journal
Cellular Reparative Mechanisms of Mesenchymal Stem Cells for Retinal Diseases
Next Article in Special Issue
Pigmented Epithelioid Melanocytoma (PEM)/Animal Type Melanoma (ATM): Quest for an Origin. Report of One Unusual Case Indicating Follicular Origin and Another Arising in an Intradermal Nevus
Previous Article in Journal
A Simple Procedure for the Evaluation of Bone Vitality by Staining with a Tetrazolium Salt
Previous Article in Special Issue
Dermal Delivery of Constructs Encoding Cre Recombinase to Induce Skin Tumors in PtenLoxP/LoxP;BrafCA/+ Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The HGF/SF Mouse Model of UV-Induced Melanoma as an In Vivo Sensor for Metastasis-Regulating Gene

by
M. Kathryn Leonard
1,2,
Nidhi Pamidimukkala
1,2,
Gemma S. Puts
1,2,
Devin E. Snyder
1,2,
Andrzej T. Slominski
3,4,5 and
David M. Kaetzel
1,2,*
1
Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland-Baltimore, Baltimore, MD 21201, USA
2
University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland-Baltimore, Baltimore, MD 21201, USA
3
Department of Dermatology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35249, USA
4
Cancer Chemoprevention Program, and Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35249, USA
5
Pathology and Laboratory Medicine Service, VA Medical Center, Birmingham, AL 35249, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(8), 1647; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081647
Submission received: 12 June 2017 / Revised: 19 July 2017 / Accepted: 24 July 2017 / Published: 28 July 2017
(This article belongs to the Special Issue Animal Models of Melanoma)

Abstract

:
Cutaneous malignant melanoma is an aggressive and potentially lethal form of skin cancer, particularly in its advanced and therapy-resistant stages, and the need for novel therapeutics and prognostic tools is acute. Incidence of melanoma has steadily increased over the past few decades, with exposure to the genome-damaging effects of ultraviolet radiation (UVR) well-recognized as a primary cause. A number of genetically-engineered mouse models (GEMMs) have been created that exhibit high incidence of spontaneous and induced forms of melanoma, and a select subset recapitulates its progression to aggressive and metastatic forms. These GEMMs hold considerable promise for providing insights into advanced stages of melanoma, such as potential therapeutic targets and prognostic markers, and as in vivo systems for testing of novel therapies. In this review, we summarize how the HGF/SF transgenic mouse has been used to reveal metastasis-regulating activity of four different genes (CDK4R24C, survivin and NME1/NME2) in the context of UV-induced melanoma. We also discuss how these models can potentially yield new strategies for clinical management of melanoma in its most aggressive forms.

1. Introduction

Melanoma is the most lethal variant of skin cancer, with melanoma incidence and mortality rising dramatically over the past 20 years in both men and women [1]. Exposure to ultraviolet radiation (UVR) is now widely accepted as a major cause of malignant cutaneous melanoma and skin cancer-related deaths [2]. While surgical resection of skin-localized melanomas in their early stages (radial growth phase) is frequently curative, invasive melanomas at vertical growth phase often progress to metastatic disease and death. Despite the advent of inhibitors of constitutively-active BRAF (e.g., vemurafenib) and immune checkpoints (e.g., antibodies against programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1) or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), current therapies rarely provide durable responses in advanced melanoma. Moreover, clinical management of melanoma patients is impaired by a lack of molecular markers that reliably identify lesions with elevated risk for metastasis and/or disease progression.
Genes that regulate the metastatic process in melanoma provide unique tools for discovery of new therapies and prognostic markers. To date, candidate metastasis-regulating genes (MRGs) have been identified in studies using established melanoma cell lines, with a spectrum of cell culture and mouse xenograft models used to assess their impacts on growth and metastatic phenotypes. While melanoma cell lines have proven invaluable models for discovery, they are imperfect representations of the melanoma tumors from which they were isolated. Most were selected for proliferation on artificial two-dimensional plastic surfaces, maintained under a continual mitotic stimulus of fetal bovine serum, and subjected to extensive passaging that frequently leads to alterations in genotype and phenotype. Genetically-engineered mouse models (GEMMs) of cancer represent a powerful in vivo approach for measuring metastasis-regulating activity of candidate MRGs in the context of an intact tumor microenvironment and immune system. A myriad of GEMMs have been developed for study of melanoma [3], some of which can indeed progress to metastatic disease and are discussed in articles within this compendium. This review is focused specifically on the hepatocyte growth factor/scatter factor (HGF/SF) mouse model of UV-induced melanoma as a means for measuring metastasis-regulating activity of genes in vivo. Such applications of the HGF/SF model hold promise for elucidating molecular mechanisms underlying the metastatic process, and for the development of new therapeutic and prognostic tools to manage the disease more effectively.

2. Application of the Hepatocyte Growth Factor/Scatter Factor (HGF/SF) Transgenic Mouse Model as a Sensor of Metastasis-Regulating Genes (MRG) Activity: Working Principles

The HGF/SF strain of transgenic mice overexpresses HGF/SF in tissues under control of the metallothionein (MT) gene promoter [4]. HGF/SF mice also exhibit spontaneous neoplasms of both mesenchymal and epithelial cell origins, a profile likely determined by MT promoter activity across diverse cell types. Importantly, HGF/SF mice display a high incidence of subcutaneous melanoma when subjected shortly after birth (postnatal day 4, P4; Figure 1) to a single erythemal dose of ultraviolet radiation (UVR; 4–8 kJ/M2; 35% UV-A/65% UV-B/0.3% UV-C) [5]. The sensitivity of HGF/SF mice to UVR-induced melanoma has been attributed in large part to an HGF-induced shift of mouse skin melanocytes from their normal localization within the hair follicle to more external sites (i.e., UV-exposed) within the epidermis and at the dermal–epidermal junction. The externalization of melanocytes in HGF/SF mouse skin resembles that seen in human skin and is rarely observed in other GEMMs of melanoma [4]. Interestingly, the UV-induced melanoma phenotype of the HGF/SF mouse has been demonstrated in both the non-pigmented FVB and highly-pigmented C57BL/6 genetic backgrounds [6].
The K14-SCF mouse model, which expresses stem cell factor (SCF) in epidermal keratinocytes under control of the keratin 14 promoter [7], also exhibits retention of melanocytes in the interfollicular layer of skin yet is melanoma-resistant [8]. This strongly suggests externalization of melanocytes is not sufficient for UV-induced melanomagenesis and that expression of the HGF/SF transgene itself contributes to the melanoma phenotype.
Cutaneous melanoma lesions usually first appear at 6–12 months post-UVR treatment in 60–70% of irradiated HGF/SF mice (Figure 1), and they resemble human melanoma in a number of respects. For example, melanoma cells form junctions with epithelial cells in the epidermal layer of skin [4,9,10], a similarity with human melanomas rarely seen in other GEMM systems (Figure 1). During malignant progression, UV-induced HGF/SF melanomas also frequently exhibit loss or inactivating mutations within regions of the p16INK4ACDKN2A gene that encode the p16INK4A tumor suppressor protein [11]. Loss of p16INK4A expression and/or function is also exhibited in the progression of many human melanomas [12]. In addition, constitutive expression of HGF/SF results in activation of the receptor tyrosine kinase and proto-oncogene MET, a growth-promoting autocrine loop characteristic of many human melanomas [10]. The oncogenic signaling loop of HGF/SF and MET is also not represented in other GEMM systems as they often utilize the BRAFV600E mutation, which is only present in 35–50% of human melanomas, to aid in the induction of melanomas. As such, the HGF/SF GEMM may better model the other 50% of human melanomas not expressing mutant BRAF.
UV-induced melanomas in the HGF/SF model are composed of heavily pigmented epithelioid and dendritic melanocytes with high proliferative potential. Although they readily invade the dermis and adjacent subcutaneous structures (Figure 1), they rarely show evidence of metastasis within four months of their initiation. In this sense, they can be viewed as poised for progression to metastatic activity upon acquisition of gene expression or mutational profiles that drive further progression or the metastatic phenotype itself. This “non-metastatic but poised state” renders them highly suitable for in vivo validation of candidate MRGs (Figure 2). To date, this principle has been exploited to validate MRG activity of four genes: CDK4R24C [2,13], survivin [14], and NME1/NME2 [15]. Each of these applications of the HGF/SF model as a sensor of MRG activity are reviewed individually in the following section.

3. Validation of MRGs Using the HGF/SF Model

3.1. Validation of Two Metastasis Driver Genes (MDGs): CDK4R24C and Survivin

3.1.1. CDK4R24C

Cyclin-dependent kinases (CDKs) are serine/threonine kinases best known for their cell cycle-regulating functions. CDK4 acts in conjunction with the D-type cyclin proteins to promote the G1-to-S transition, and is opposed in its actions by the CDK inhibitor p16INK4A. The CDK4 gene is often amplified and overexpressed in a variety of cancers including melanoma [16,17,18]. Activating mutations in CDK4 have also been observed in cancer and are usually directed to the p16INK4A-binding domain encoded within exon 2 [17]. An example is the arginine24-to-cysteine substitution (CDK4R24C), which stabilizes the CDK4 molecule by preventing its physical interaction with p16INK4A [19]. In turn, constitutive activity of CDK4R24C overrides the ability of p16INK4A to function as a mediator of oncogene-induced senescence [20,21] A transgenic mouse strain engineered on a C57BL/6 genetic background for constitutive expression of CDK4R24C was shown to be prone to two step chemical (7,12-dimethylbenz[a]anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA))- and UV-induced melanomas, although the tumors exhibited little metastatic potential [21,22]. Moreover, lifespan of the mice was limited by the incidence of unrelated tumors such as sarcomas and lymphomas. Further evidence of the importance of these pathways is the observation that loss of the CDKN2A locus encoding p16INK4A and p14ARF, which, respectively, inhibit CDK4/6 and activate the p53 tumor suppressor, are among the most commonly inactivated genes in cancer (including melanoma) [23,24].
To generate a more aggressive mouse model of CDK4R24C-dependent melanoma, the CDK4R24C strain was crossed with HGF/SF mice. This hybrid indeed exhibited a more rapid onset of DMBA/TPA- and UVR-induced melanomas than either of the parental strains [25]. Moreover, DMBA/TPA-induced melanomas were more invasive in the hybrid, with hybrid tumors displaying enhanced metastasis to regional lymph nodes and lungs. Organ metastasis was limited to lung tissue, with no lesions detected in liver, spleen, kidney or brain. In a later study, however, liver metastasis was indeed observed in UVB-induced melanomas with the CDK4R24C × HGF/SF model [26]. In effect, these studies represented an early application of the HGF/SF mouse as a sensor for metastasis-regulating activity of a test gene, CDK4R24C. A caveat of the study is that both parental strains are prone to induced forms of melanoma, conferring uncertainty as to the relative roles of each transgene in melanoma initiation and growth. Another consideration is that primary melanomas of the hybrid strain grew more rapidly and with more lesions per mouse, suggesting increased cumulative tumor mass could have contributed to the metastatic phenotype in addition to, or in lieu of, a direct impact of CDK4R24C on metastatic potential of the melanoma cell itself. In addition, the germline transmission and constitutive expression of the CDK4R24C transgene across all tissues suggests the possibility that impacts of CDK4R24C on cells in the tumor microenvironment (alone or in combination with HGF/SF) could have contributed to the elevated metastatic activity of melanomas in the hybrid strain. Indeed, paracrine actions of HGF/SF in the HGF/SF model have been shown to impact lung colonization activity of melanoma cell allografts [27]. Overall, however, multiple studies have provided valuable proof-of-principle for the HGF/SF model of UV-induced melanoma as a sensor of MSG activity. To date, the HGF/SF × CDK4R24C hybrid has proven a valuable model of metastatic melanoma for studies of immunological tolerance [22], and impacts of inflammation on resistance to T-cell therapy [28] and metastatic potential [2,25].

3.1.2. Survivin

Survivin is a member of the “Inhibitors of Apoptosis (IAP)” family of proteins, which were first identified in viruses and later in metazoans (reviewed in [29]). Evidence suggests IAP activity of survivin is mediated at least in part via inhibition of caspase activity (primarily caspases 3 and 7) and structural-functional interactions with the protein, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Survivin expression is associated with fetal tissues, but is not normally seen in adult melanocytes [30] or other differentiated cells [31]. Survivin expression is a hallmark of melanoma [32] and a host of other human cancers, including those of the lung, colon, breast, prostate, and neuroblastoma [33,34,35]. In melanoma, strong positive correlations are seen among survivin expression, metastatic disease, and poor survival [36,37]. Consistent with a cancer-driving function, survivin plays key roles in mitotic control, specifically cytokinesis [38], and chromosomal alignment during mitosis [39]. Using a variety of melanoma cell lines, multiple groups have demonstrated that disruption of survivin expression and/or function confers sensitivity to apoptotic signals, chemosensitization, and inhibits growth as tumor xenografts in mice [40,41]. Survivin has also been shown to confer metastatic activity to the mammary adenocarcinoma cell line R3230AC [42], suggesting a potential metastasis-driving function. Another study has implicated α5β1 integrin as a mediator of survivin-driven metastasis in melanoma cell lines [43].
To investigate potential roles of survivin in the initiation and progression of melanoma in vivo, a transgenic mouse strain was created that provided melanocyte-specific expression of survivin under the transcriptional control of the dopachrome tautomerase (Dct) promoter [14]. Melanocytes isolated from these mice (Dct-survivin) were resistant to UV-induced apoptosis, but their proliferative rate was unaltered. Likewise, formation of DMBA-induced melanocytic nevi was unaffected in the Dct-survivin strain. To assess the impact of survivin in the context of a full melanoma phenotype, Dct-survivin mice were bred with the HGF/SF strain. Melanocytes in the HGF/SF × Dct-survivin hybrid strain exhibited higher resistance via anti-apoptotic mechanisms to UVR treatment in vivo than the parental HGF/SF strain, consistent with the survivin-dependent resistance seen with cultured Dct-survivin melanocytes ex vivo. Survivin overexpression in the hybrid strain also accelerated the onset and density of UV-induced melanomas. Notably, larger melanomas (>5 mm) in the hybrid strain were associated with a significantly higher incidence of lymph node involvement and lung metastasis than seen with similarly-sized melanomas of the HGF/SF strain. The increased metastatic activity of melanomas in hybrid mice was accompanied by more prominent association of melanoma cells with blood and lymphatic vessels (angiotropism), as well as adjacent muscle tissue.
In addition to providing in vivo validation of MDG activity for survivin, the model also yielded in vivo confirmation of earlier observations that survivin confers resistance to apoptosis in melanoma cells. Melanomas that metastasized in the hybrid strain indeed contained significantly fewer apoptotic cells than matched, non-metastatic tumors from HGF/SF mice. Suppression of apoptosis has been suggested as a metastasis-promoting function of cancer cells [44], conferring survival advantage to tumor cells upon acquisition of potential metastasis-driving mutations as well as during dissemination and colonization steps of metastasis. A complicating factor with the model was a significantly higher tumor load in the Dct-survivin × HGF/SF hybrid (two-fold), which could have contributed to the metastasis phenotype. On the other hand, the increase was relatively small, and taken together with minimal effects of survivin on growth rates of melanoma lesions would seem unlikely to account completely for the strong metastasis observed. In addition to revealing MDG activity for survivin in vivo, the Dct-survivin × HGF/SF hybrid strain may be suitable as an in vivo model for testing of survivin-inhibitory therapies in advanced melanoma. Overall, this study provided a further demonstration of the utility of the HGF/SF mouse strain as a sensor for validating the MRG activity of a test gene (survivin) in vivo. In light of the utility of the HGF/SF mouse in revealing metastasis-driving activity of survivin, it could also be of potential application to analysis of other apoptosis-inhibiting proteins such as other IAP factors and members of the Bcl-2 family in a wild-type BRAF background [45]. Nearly all prior studies with anti-apoptotic proteins were conducted in a mutant BRAF background, despite the understanding that HGF-cMET signaling (like that in the HGF/SF model) activates AKT which can lead to suppression of many apoptosis inhibiting genes [45,46].

3.2. MSGs: NME1 and NME2

The NME1 (initially designated nm23, for Negative in Metastasis clone 23”) gene was identified by virtue of reduced expression of its cognate transcript in a metastatic clone of the mouse melanoma cell line K-1735 [47]. Enforced expression of NME1 was shown to suppress metastatic potential in a variety of cell lines, and with the notable absence of impacts on proliferative or tumor-forming capacity (reviewed in [48]). An inhibitory activity of NME1 in cancer metastasis was further suggested by an inverse correlation between NME1 expression and poor clinical outcome observed across a spectrum of human cancers, including melanoma [49,50]. Together, these findings led to the designation of NME1 as a metastasis suppressor gene (MSG), a term subsequently extended to all genes sharing the unique ability to suppress metastatic potential of cancer cells without impacting primary tumor growth [51,52,53]. At least 23 MSGs have been described that affect a broad spectrum of cellular processes, and are operative in many different settings of cancer [51]. Due to their selective impact on metastasis, additional study of MSGs holds great potential for elucidating molecular events and processes that specifically regulate the metastatic process. The metastasis-directed actions of MSGs stand in contrast with those of oncogenes and tumor suppressors, whose impacts on tumor growth can obscure their effects on the metastatic process per se.
In addition to NME1, nine other homologs (NME2–NME10) have been identified [54]. NME homologs 1–4 each harbor a nucleoside diphosphate kinase (NDPK) activity in vitro that catalyzes transfer of γ-phosphate residues between NTPs and NDPs [55] via a phospho-histidine intermediate. Our laboratory also demonstrated NME1 harbors a 3′–5′ exonuclease activity directed to single-stranded DNA substrates [56]. Both enzymatic activities have been suggested to contribute to the metastasis suppressor function of NME1 in melanoma cells [57], although the underlying molecular mechanisms have yet to be identified.
We have shown that NME1 expression promotes repair of UV-induced DNA damage via the nucleotide excision repair pathway in yeast [58] and mammalian cells [59]. Moreover, mice rendered hemizygous-null at the tandemly-arranged NME1 and NME2 loci (herein designated NME1/2+/−) are highly vulnerable to UV-induced melanoma in situ and epidermal inclusion cyst formation on tail skin [59], consistent with a deficit in DNA repair. The DNA repair-promoting activity of NME1 and/or NME2 suggests loss of their expression could promote acquisition of mutations that accelerate melanoma progression. Interestingly, expressions of genes involved in multiple pathways of DNA repair are upregulated in metastatic melanoma [60,61,62,63]. This suggests that viability of metastatic cells may require a return to genomic stability in order for proliferation to be both fast and free of errors, and may indeed contribute to the resistance of metastatic melanoma to therapy [64].
We and others have also shown that NME1 regulates a broad spectrum of RNAs [65,66,67], some of which may mediate its metastasis suppressor function. For example, NME1 regulates expression of numerous RNA species in breast carcinoma cells, with the lysophosphatidic acid (LPA) receptor EDG2 identified as a key target [67]. Both NME1 and NME2 exhibit binding and DNA-cleaving activities upon single-stranded motifs in the CMYC [68] and platelet derived growth factor A (PDGFA) [56,69] promoter regions, and appear to regulate transcription via those interactions [70]. Chromatin immunoprecipitation analyses have confirmed physical association of NME1 with non-B-form elements in the promoter regions of multiple genes, including PDGFA and others [71,72]. Taken together, these studies suggest direct participation of NME1 and NME2 proteins in both DNA repair and gene transcription processes. Such functional duality is not unusual, with a recent report identifying more than 2500 unique pairwise protein associations that encompass the processes of DNA repair and gene transcription [73]. Clearly, both processes could be central to the metastasis suppressor function of NME proteins.
While studies conducted in cancer cell lines strongly suggested NME1 possesses metastasis suppressor activity, the activity had yet to be validated in a model of spontaneous melanoma in vivo. To this end, we crossed the HGF/SF and NME1/2+/− mouse strains in a C57BL/6 genetic background to measure the impact of NME1/NME2 deficiency on the metastatic potential of UV-induced melanoma [15]. It should be noted that the homozygous-null NME1/2−/− genotype could not be used in this study, due to death in late gestation or early postnatal ages from severe anemia and other manifestations of impaired erythroid development [74,75]. While UV-induced melanomas of the HGF/SF strain failed to exhibit metastasis to lymph nodes or distal organs, robust metastatic activity was seen in HGF/SF × NME1/2+/− hybrids bearing large (500 mm3) melanomas on back skin (Figure 3). Heavily pigmented metastases to draining lymph nodes were observed in 9/10 of these mice, with a majority (7/10) also exhibiting a high number of melanotic metastases in lung. A significant number (3/10) of mice also had substantial metastatic involvement in liver and the thoracic cavity, and to a lesser extent in bone and brain (Figure 3). Metastasis was not observed in melanomas of either strain that arose in other UV-exposed sites, such as the tail, leg/paw or ear. Such melanomas were delayed in their appearance and had much slower growth rates than those arising on back skin. Consistent with the classical definition of an MSG, the NME1/2+/− genotype did not affect the average growth rate of primary tumors, time of onset of UV-induced melanomas (~180 days), or penetrance of the melanoma phenotype (~70% of irradiated mice). Somewhat unexpectedly, no difference was observed in the invasive index of melanomas between the two strains, suggesting the high metastatic potential conferred by NME deficiency was due to impacts on subsequent steps of the metastatic cascade. Cumulatively, these findings confirmed the HGF/SF model as a robust sensor for MRG activity of the test genes NME1 and NME2. Interestingly, a metastatic phenotype was conferred even in the face of one intact copy of NME1 and NME2 and retention of a 50% expression level for both proteins, highlighting the sensitivity of the HGF/SF model. Moreover, the HGF/SF model system revealed prototypical MSG activity for NME1 and/or NME2 for the first time in an in vivo setting of melanoma, with suppression of metastasis in the absence of an effect on primary tumor growth. Another group has also verified the metastasis suppressor function of NME1 in the context of a mouse model of carcinogen-induced hepatocellular carcinoma [54].
As with the studies of HGF/SF × CDK4R24C hybrid, our NME1/NME2 deletion was germline in nature and is thus found in all tissues of the NME1/2+/− genotype. This again suggests the possibility that impacts of a test gene (e.g., NME1/2 locus) on metastasis of UV-induced HGF/SF melanomas might be exerted on the tumor microenvironment in addition to, or in the absence of, effects on the melanoma cell itself. While no overt evidence of changes within the microenvironment was apparent in the histological analysis of tumors in our study, the question has yet to be addressed in detail.
Our laboratory is currently exploiting the HGF/SF and NME1/2+/− model system on a number of fronts. The system is now being used to measure the separate impacts of NME1 and NME2 on metastatic potential, by crossing individual mouse strains that harbor homozygous-null deletions of either the NME1 [76] or NME2 genes [77] with HGF/SF mice. We have also generated a number of stable cell lines from primary and metastatic lesions in HGF/SF and NME1/2+/− mice for genetic manipulation and functional studies. Their C57BL/6 origin will permit allografting in syngeneic C57/BL/6 mice harboring an intact immune system. In light of the DNA repair and transcription functions of NME1, we are also using NextGen sequencing to compare mutational and transcriptional profiles in UV-induced melanomas (both primary and metastatic tumors) between the HGF/SF and HGF/SF × NME1/2+/− hybrid strains. These studies will be facilitated by the low frequency of melanoma lesions per mouse (usually one), facilitating comparisons of matched primary and metastatic tumors. It is anticipated that genomic and transcriptomic alterations will be identified that are correlated with the metastatic phenotype and, thus, potentially metastasis-regulating in nature. A clear advantage of this model system for these studies is the genetic homogeneity of the mouse strains under study, with all strains having been incorporated (>6 generations) into C57BL/6 genetic background of our own colony. This provides a considerable advantage over genomic analyses in human melanomas, which are subject to considerable heterogeneity from polymorphisms and mutations in the human population. Candidate metastasis-regulating alterations will be validated to the extent possible by analyzing their impacts on growth and metastatic phenotypes in cell culture systems, as well searching for their potential occurrence in human melanomas. In principle, such alterations could prove valuable as prognostic markers for melanoma progression in human patients.

4. Optimization of the HGF/SF Model as a Sensor for MRG Activity

While the HGF/SF model has been used effectively as a sensor for MRG activity, the application can be further optimized in a number of ways. A current impediment to progress is the length of time of a typical experiment, which can take up to a year after the initiating UVR treatment of neonatal mice. We are currently incorporating a melanocyte-specific, homozygous-null of the p16INK4Aainactivation of the p16INK4A coding region of the CDKN2A gene into the HGF/SF model to accelerate our ongoing studies of MRG activity of NME genes. Mice bearing a melanocyte-directed inactivation of the p16INK4AP16INK4A locus should exhibit a longer lifespan than those bearing a germline deletion, which die prematurely. Introduction of the p16INK4AP16INK4A-null genotype into HGF/SF mice was shown previously to cut melanoma onset time by more than half, from 6–12 months to 2–3 months [78]. That study also showed that the p16INK4A-null condition maintained the average multiplicity of UV-induced melanoma lesions to one per mouse, an important consideration for pairwise comparisons between primary and metastatic tumors. Another potential complication was posed in the CDKR24C and NME gene studies, in which the genetic alterations were implemented in all cell types/tissues of the mouse. This raised the issue of whether effects on tumor and/or metastatic potential were exerted not only on the melanoma cell itself but also other cells, such as those of the TME. The survivin study [14] revealed how a melanocyte-specific promoter such as the Dct promoter can be used effectively to limit expression of MRG candidates to tumor tissue. The utility of a Dct-driven, doxycycline-inducible transgene encoding a fusion of histone 2B and green fluorescence protein [79,80] illustrates the feasibility of such an approach. In addition, standard Cre-lox approaches to conditional (i.e., melanocyte-specific and time-controlled) deletion should be considered for future analysis of candidate MSGs. Another limitation of the HGF/SF sensor model is that mice must be sacrificed to determine if metastatic lesions are present, unless expensive alternatives such as magnetic resonance imaging (MRI) are available [81,82]. Incorporation of a melanocyte-specific transgene such as a Dct-luciferase module would enable monitoring of primary and metastatic melanoma lesions in living animals with standard imaging equipment.

5. Conclusions

With studies in melanoma cell lines providing an increasing list of genes, genetic lesions, and gene expression profiles that exert metastasis-regulating activity, the HGF/SF model of UV-induced melanoma represents a valuable approach for measuring their respective activities in vivo. A primary attribute of the HGF/SF model is the UV-initiated character of its melanomas [83], which recapitulates the UV-dependence of most human melanomas [84]. HGF/SF melanomas also exhibit a number of other characteristics that resemble the human form of the disease, such as the junctional activity observed between melanoma and epithelial cells in skin [4,10], and loss of the tumor suppressor CDKN2A during malignant progression [11]. Importantly, melanomas of hybrid crosses of the HGF/SF mouse with three different strains of mice harboring genetic alterations give rise to lymph node and organ metastasis [14,15,25]. In particular, the HGF/SF × NME1/2+/− hybrid exhibits a tropism of metastases (lymph node > lung > liver > bone, brain, etc.) that is nearly identical to that observed in human melanoma [85].
To our knowledge, impacts of the known melanoma drivers BRAFV600E and PTEN inactivation/deletion have not been examined in the context of the HGF/SF mouse model. While the conditional BRAFV600E/PTEN knockout mouse model is well-known for its robust metastatic melanoma phenotype [86], and PI3K/AKT signaling indeed underlies the phenotype [87], these lesions would seem unlikely to provide strong pro-metastatic effects in the HGF/SF mouse where signaling via the RAS/RAF/MAPK and PI3K pathways is already constitutively active [88]. Nevertheless, boosting of signaling pathways initiated by BRAFV600E expression and PTEN inactivation could have unanticipated benefits in accelerating UV-induced melanoma initiation and progression and, thus, may warrant experimental testing.
In addition to providing a system for quantifying MRG activity, HGF/SF hybrids that exhibit robust metastatic melanoma are promising in vivo models for testing of novel therapeutics specifically directed to various aspects of the metastatic process. Of particular interest is the elimination of metastatic cells in both growing and dormant forms, disruption of individual steps in the metastatic cascade, and functional assessment of other relevant molecular targets. As discussed above, the HGF/SF model may not be appropriate for testing of inhibitors of mutant BRAF, the constitutive MET activity and consequent downstream activation of BRAF in HGF/SF melanomas likely precludes the genesis of BRAF-activating mutations. Nevertheless, metastatic variants of the HGF/SF model appear to be a viable option for testing of novel metastasis-directed therapeutics alone and in combination with MAPK inhibitors, immune checkpoint inhibitors, and even conventional therapeutic regimens. As discussed above, detailed comparisons of molecular signatures between non-metastatic melanomas of the HGF/SF strain with metastatic melanomas of HGF/SF hybrid crosses may provide an ideal discovery platform for the diagnostic and prognostic markers that are so acutely needed for management of melanoma patients.

Acknowledgments

The authors would like to express their gratitude to Glenn Merlino and Chi-Peng Day for their critical review of this manuscript. Work from the Kaetzel laboratory cited in this review was funded by grants from the National Cancer Institute (R01 CA83237, R01 CA159871 and R01 CA159871-S1).

Author Contributions

M. Kathryn Leonard and David M. Kaetzel edited the manuscript. Andrzej T. Slominski contributed clarifications and assistance on the manuscript. M. Kathryn Leonard, Nidhi Pamidimukkala, Gemma S. Puts, Devin E. Snyder and David M. Kaetzel collected the literature and helped writing the paper.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CDKCyclin-Dependent Kinase
DctDopachrome Tautomerase
GEMMGenetically-Engineered Mouse model
IAPInhibitor of Apoptosis
HGF/SFHepatocyte Growth Factor/Scatter Growth Factor
MRGsMetastasis-Regulating Genes
MDGMetastasis-Driving Gene
MSGMetastasis Suppressor Gene
MTMetallothionine
UVRUltraviolet Light Radiation
DMBA7,12-Dimethylbenz[a]anthracene
TPA12-O-Tetradecanoylphorbol-13-acetate
NF-κBNuclear Factor Kappa-light-chain-enhancer of activated B cells
LPALysophosphatidic Acid
PDGFAPlatelet Derived Growth Factor A

References

  1. Shaikh, W.R.; Dusza, S.W.; Weinstock, M.A.; Oliveria, S.A.; Geller, A.C.; Halpern, A.C. Melanoma thickness and survival trends in the United States, 1989–2009. J. Nat. Cancer Inst. 2016, 108, djv294. [Google Scholar] [CrossRef] [PubMed]
  2. Bald, T.; Quast, T.; Landsberg, J.; Rogava, M.; Glodde, N.; Lopez-Ramos, D.; Kohlmeyer, J.; Riesenberg, S.; van den Boorn-Konijnenberg, D.; Homig-Holzel, C.; et al. Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature 2014, 507, 109–113. [Google Scholar] [CrossRef] [PubMed]
  3. Kuzu, O.F.; Nguyen, F.D.; Noory, M.A.; Sharma, A. Current state of animal (mouse) modeling in melanoma research. Cancer Growth Metastasis 2015, 8, 81–94. [Google Scholar] [CrossRef] [PubMed]
  4. Takayama, H.; LaRochelle, W.J.; Sharp, R.; Otsuka, T.; Kriebel, P.; Anver, M.; Aaronson, S.A.; Merlino, G. Diverse tumorigenesis associated with aberrant development in mice overexpressing hepatocyte growth factor/scatter factor. Proc. Natl. Acad. Sci. USA 1997, 94, 701–706. [Google Scholar] [CrossRef] [PubMed]
  5. Noonan, F.P.; Recio, J.A.; Takayama, H.; Duray, P.; Anver, M.R.; Rush, W.L.; de Fabo, E.C.; Merlino, G. Neonatal sunburn and melanoma in mice. Nature 2001, 413, 271–272. [Google Scholar] [CrossRef] [PubMed]
  6. Noonan, F.P.; Zaidi, M.R.; Wolnicka-Glubisz, A.; Anver, M.R.; Bahn, J.; Wielgus, A.; Cadet, J.; Douki, T.; Mouret, S.; Tucker, M.A.; et al. Melanoma induction by ultraviolet A but not ultraviolet B radiation requires melanin pigment. Nat. Commun. 2012, 3, 884. [Google Scholar] [CrossRef] [PubMed]
  7. Kunisada, T.; Yoshida, H.; Yamazaki, H.; Miyamoto, A.; Hemmi, H.; Nishimura, E.; Shultz, L.D.; Nishikawa, S.; Hayashi, S. Transgene expression of steel factor in the basal layer of epidermis promotes survival, proliferation, differentiation and migration of melanocyte precursors. Development 1998, 125, 2915–2923. [Google Scholar] [PubMed]
  8. D’Orazio, J.A.; Nobuhisa, T.; Cui, R.; Arya, M.; Spry, M.; Wakamatsu, K.; Igras, V.; Kunisada, T.; Granter, S.R.; Nishimura, E.K.; et al. Topical drug rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning. Nature 2006, 443, 340–344. [Google Scholar] [CrossRef] [PubMed]
  9. Vanover, J.C.; Spry, M.L.; Hamilton, L.; Wakamatsu, K.; Ito, S.; D’Orazio, J.A. Stem cell factor rescues tyrosinase expression and pigmentation in discreet anatomic locations in albino mice. Pigment Cell Melanoma Res. 2009, 22, 827–838. [Google Scholar] [CrossRef] [PubMed]
  10. Otsuka, T.; Takayama, H.; Sharp, R.; Celli, G.; LaRochelle, W.J.; Bottaro, D.P.; Ellmore, N.; Vieira, W.; Owens, J.W.; Anver, M.; et al. c-Met autocrine activation induces development of malignant melanoma and acquisition of the metastatic phenotype. Cancer Res. 1998, 58, 5157–5167. [Google Scholar] [PubMed]
  11. Recio, J.A.; Noonan, F.P.; Takayama, H.; Anver, M.R.; Duray, P.; Rush, W.L.; Lindner, G.; de Fabo, E.C.; DePinho, R.A.; Merlino, G. INK4A/ARF deficiency promotes ultraviolet radiation-induced melanomagenesis. Cancer Res. 2002, 62, 6724–6730. [Google Scholar] [PubMed]
  12. Sharpless, E.; Chin, L. The INK4a/ARF locus and melanoma. Oncogene 2003, 22, 3092–3098. [Google Scholar] [CrossRef] [PubMed]
  13. Barnhill, R.L.; Lugassy, C. Angiotropic malignant melanoma and extravascular migratory metastasis: Description of 36 cases with emphasis on a new mechanism of tumour spread. Pathology 2004, 36, 485–490. [Google Scholar] [CrossRef] [PubMed]
  14. Thomas, J.; Liu, T.; Cotter, M.A.; Florell, S.R.; Robinette, K.; Hanks, A.N.; Grossman, D. Melanocyte expression of survivin promotes development and metastasis of UV-induced melanoma in HGF-transgenic mice. Cancer Res. 2007, 67, 5172–5178. [Google Scholar] [CrossRef] [PubMed]
  15. Jarrett, S.G.; Novak, M.; Harris, N.; Merlino, G.; Slominski, A.; Kaetzel, D.M. NM23 deficiency promotes metastasis in a UV radiation-induced mouse model of human melanoma. Clin. Exp. Metastasis 2013, 30, 25–36. [Google Scholar] [CrossRef] [PubMed]
  16. Muthusamy, V.; Hobbs, C.; Nogueira, C.; Cordon-Cardo, C.; McKee, P.H.; Chin, L.; Bosenberg, M.W. Amplification of CDK4 and MDM2 in malignant melanoma. Genes Chromosomes Cancer 2006, 45, 447–454. [Google Scholar] [CrossRef] [PubMed]
  17. Wolfel, T.; Hauer, M.; Schneider, J.; Serrano, M.; Wolfel, C.; Klehmann-Hieb, E.; de Plaen, E.; Hankeln, T.; Meyer zum Buschenfelde, K.H.; Beach, D. A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science 1995, 269, 1281–1284. [Google Scholar] [CrossRef] [PubMed]
  18. Zuo, L.; Weger, J.; Yang, Q.; Goldstein, A.M.; Tucker, M.A.; Walker, G.J.; Hayward, N.; Dracopoli, N.C. Germline mutations in the p16INK4a binding domain of CDK4 in familial melanoma. Nat. Genet. 1996, 12, 97–99. [Google Scholar] [CrossRef] [PubMed]
  19. Bartkova, J.; Lukas, J.; Guldberg, P.; Alsner, J.; Kirkin, A.F.; Zeuthen, J.; Bartek, J. The p16-cyclin D/Cdk4-pRb pathway as a functional unit frequently altered in melanoma pathogenesis. Cancer Res. 1996, 56, 5475–5483. [Google Scholar] [PubMed]
  20. Michaloglou, C.; Vredeveld, L.C.; Soengas, M.S.; Denoyelle, C.; Kuilman, T.; van der Horst, C.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] [PubMed]
  21. Sotillo, R.; Garcia, J.F.; Ortega, S.; Martin, J.; Dubus, P.; Barbacid, M.; Malumbres, M. Invasive melanoma in Cdk4-targeted mice. Proc. Natl. Acad. Sci. USA 2001, 98, 13312–13317. [Google Scholar] [CrossRef] [PubMed]
  22. Steitz, J.; Buchs, S.; Tormo, D.; Ferrer, A.; Wenzel, J.; Huber, C.; Wolfel, T.; Barbacid, M.; Malumbres, M.; Tuting, T. Evaluation of genetic melanoma vaccines in cdk4-mutant mice provides evidence for immunological tolerance against authochthonous melanomas in the skin. Int. J. Cancer 2006, 118, 373–380. [Google Scholar] [CrossRef] [PubMed]
  23. Hayward, N. New developments in melanoma genetics. Curr. Oncol. Rep. 2000, 2, 300–306. [Google Scholar] [CrossRef] [PubMed]
  24. Begg, C.B.; Orlow, I.; Hummer, A.J.; Armstrong, B.K.; Kricker, A.; Marrett, L.D.; Millikan, R.C.; Gruber, S.B.; Anton-Culver, H.; Zanetti, R.; et al. Lifetime risk of melanoma in CDKN2A mutation carriers in a population-based sample. J. Natl. Cancer Inst. 2005, 97, 1507–1515. [Google Scholar] [CrossRef] [PubMed]
  25. Tormo, D.; Ferrer, A.; Gaffal, E.; Wenzel, J.; Basner-Tschakarjan, E.; Steitz, J.; Heukamp, L.C.; Gutgemann, I.; Buettner, R.; Malumbres, M.; et al. Rapid growth of invasive metastatic melanoma in carcinogen-treated hepatocyte growth factor/scatter factor-transgenic mice carrying an oncogenic CDK4 mutation. Am. J. Pathol. 2006, 169, 665–672. [Google Scholar] [CrossRef] [PubMed]
  26. Gaffal, E.; Landsberg, J.; Bald, T.; Sporleder, A.; Kohlmeyer, J.; Tuting, T. Neonatal UVB exposure accelerates melanoma growth and enhances distant metastases in Hgf-Cdk4R24C C57BL/6 mice. Int. J. Cancer 2011, 129, 285–294. [Google Scholar] [CrossRef] [PubMed]
  27. Yu, Y.; Merlino, G. Constitutive c-Met signaling through a nonautocrine mechanism promotes metastasis in a transgenic transplantation model. Cancer Res. 2002, 62, 2951–14956. [Google Scholar] [PubMed]
  28. Landsberg, J.; Kohlmeyer, J.; Renn, M.; Bald, T.; Rogava, M.; Cron, M.; Fatho, M.; Lennerz, V.; Wolfel, T.; Holzel, M.; et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature 2012, 490, 412–416. [Google Scholar] [CrossRef] [PubMed]
  29. LaCasse, E.C.; Baird, S.; Korneluk, R.G.; MacKenzie, A.E. The inhibitors of apoptosis (IAPs) and their emerging role in cancer. Oncogene 1998, 17, 3247–3259. [Google Scholar] [CrossRef] [PubMed]
  30. Bowen, A.R.; Hanks, A.N.; Murphy, K.J.; Florell, S.R.; Grossman, D. Proliferation, apoptosis, and survivin expression in keratinocytic neoplasms and hyperplasias. Am. J. Dermatopathol. 2004, 26, 177–181. [Google Scholar] [CrossRef] [PubMed]
  31. Ambrosini, G.; Adida, C.; Altieri, D.C. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat. Med. 1997, 3, 917–921. [Google Scholar] [CrossRef] [PubMed]
  32. Grossman, D.; McNiff, J.M.; Li, F.; Altieri, D.C. Expression and targeting of the apoptosis inhibitor, survivin, in human melanoma. J. Investig. Dermatol. 1999, 113, 1076–1081. [Google Scholar] [CrossRef] [PubMed]
  33. Adida, C.; Berrebi, D.; Peuchmaur, M.; Reyes-Mugica, M.; Altieri, D.C. Anti-apoptosis gene, survivin, and prognosis of neuroblastoma. Lancet 1998, 351, 882–883. [Google Scholar] [CrossRef]
  34. Lu, C.D.; Altieri, D.C.; Tanigawa, N. Expression of a novel antiapoptosis gene, survivin, correlated with tumor cell apoptosis and p53 accumulation in gastric carcinomas. Cancer Res. 1998, 58, 1808–1812. [Google Scholar] [PubMed]
  35. Kawasaki, H.; Altieri, D.C.; Lu, C.D.; Toyoda, M.; Tenjo, T.; Tanigawa, N. Inhibition of apoptosis by survivin predicts shorter survival rates in colorectal cancer. Cancer Res. 1998, 58, 5071–5074. [Google Scholar] [PubMed]
  36. Gradilone, A.; Gazzaniga, P.; Ribuffo, D.; Scarpa, S.; Cigna, E.; Vasaturo, F.; Bottoni, U.; Innocenzi, D.; Calvieri, S.; Scuderi, N.; et al. Survivin, bcl-2, bax, and bcl-X gene expression in sentinel lymph nodes from melanoma patients. J. Clin. Oncol. 2003, 21, 306–312. [Google Scholar] [CrossRef] [PubMed]
  37. Takeuchi, H.; Morton, D.L.; Elashoff, D.; Hoon, D.S.B. Survivin expression by metastatic melanoma predicts poor disease outcome in patients receiving adjuvant polyvalent vaccine. Int. J. Cancer 2005, 117, 1032–1038. [Google Scholar] [CrossRef] [PubMed]
  38. Li, F.; Ambrosini, G.; Chu, E.Y.; Plescia, J.; Tognin, S.; Marchisio, P.C.; Altieri, D.C. Control of apoptosis and mitotic spindle checkpoint by survivin. Nature 1998, 396, 580–584. [Google Scholar] [CrossRef] [PubMed]
  39. Vong, Q.P.; Cao, K.; Li, H.Y.; Iglesias, P.A.; Zheng, Y. Chromosome alignment and segregation regulated by ubiquitination of survivin. Science 2005, 310, 1499–1504. [Google Scholar] [CrossRef] [PubMed]
  40. Grossman, D.; Kim, P.J.; Schechner, J.S.; Altieri, D.C. Inhibition of melanoma tumor growth in vivo by survivin targeting. Proc. Natl. Acad. Sci. USA 2001, 98, 635–640. [Google Scholar] [CrossRef] [PubMed]
  41. Pennati, M.; Binda, M.; de Cesare, M.; Pratesi, G.; Folini, M.; Citti, L.; Daidone, M.G.; Zunino, F.; Zaffaroni, N. Ribozyme-mediated down-regulation of survivin expression sensitizes human melanoma cells to topotecan in vitro and in vivo. Carcinogenesis 2004, 25, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
  42. Hoon, D.B.; Wang, H.C.; Ramshaw, I.A. Increased metastatic ability and bone formation of a mammary adenocarcinoma in vivo after in vitro passaging. Eur. J. Cancer Clin. Oncol. 1984, 20, 1517–1526. [Google Scholar] [CrossRef]
  43. McKenzie, J.A.; Liu, T.; Jung, J.Y.; Jones, B.B.; Ekiz, H.A.; Welm, A.L.; Grossman, D. Survivin promotion of melanoma metastasis requires upregulation of α 5 integrin. Carcinogenesis 2013, 34, 2137–2144. [Google Scholar] [CrossRef] [PubMed]
  44. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  45. Mukherjee, N.; Schwan, J.V.; Fujita, M.; Norris, D.A.; Shellman, Y.G. Alternative treatments for melanoma: Targeting BCL-2 family members to de-bulk and kill cancer stem cells. J. Investig. Dermatol. 2015, 135, 2155–2161. [Google Scholar] [CrossRef] [PubMed]
  46. Ye, M.; Hu, D.; Tu, L.; Zhou, X.; Lu, F.; Wen, B.; Wu, W.; Lin, Y.; Zhou, Z.; Qu, J. Involvement of PI3K/Akt signaling pathway in hepatocyte growth factor-induced migration of uveal melanoma cells. Investig. Ophthalmol. Vis. Sci. 2008, 49, 497–504. [Google Scholar] [CrossRef] [PubMed]
  47. Steeg, P.S.; Bevilacqua, G.; Kopper, L.; Thorgerisson, U.R.; Talmadge, J.E.; Liotta, L.A.; Sobel, M. Evidence for a novel gene associated with low tumor metastatic potential. J. Natl. Cancer Inst. 1988, 80, 200–205. [Google Scholar] [CrossRef] [PubMed]
  48. Hartsough, M.T.; Steeg, P.S. Nm23/nucleoside diphosphate kinase in human cancers. J. Bioenerg. Biomembr. 2000, 32, 301–308. [Google Scholar] [CrossRef] [PubMed]
  49. Leone, A.; Flatow, U.; King, C.R.; Sandeen, M.A.; Margulies, I.M.; Liotta, L.A.; Steeg, P.S. Reduced tumor incidence, metastatic potential, and cytokine responsiveness of nm23-transfected melanoma cells. Cell 1991, 65, 25–35. [Google Scholar] [CrossRef]
  50. Florenes, V.A.; Aamdal, S.; Myklebost, O.; Maelandsmo, G.M.; Bruland, O.S.; Fodstad, O. Levels of nm23 messenger RNA in metastatic malignant melanomas: Inverse correlation to disease progression. Cancer Res. 1992, 52, 6088–6091. [Google Scholar] [PubMed]
  51. Smith, S.C.; Theodorescu, D. Learning therapeutic lessons from metastasis suppressor proteins. Nat. Rev. Cancer 2009, 9, 253–264. [Google Scholar] [CrossRef] [PubMed]
  52. Hurst, D.R.; Welch, D.R. Metastasis suppressor genes at the interface between the environment and tumor cell growth. Int. Rev. Cell Mol. Biol. 2011, 286, 107–180. [Google Scholar] [CrossRef] [PubMed]
  53. Marino, N.; Nakayama, J.; Collins, J.W.; Steeg, P.S. Insights into the biology and prevention of tumor metastasis provided by the Nm23 metastasis suppressor gene. Cancer Metastasis Rev. 2012, 31, 593–603. [Google Scholar] [CrossRef] [PubMed]
  54. Boissan, M.; Dabernat, S.; Peuchant, E.; Schlattner, U.; Lascu, I.; Lacombe, M.L. The mammalian Nm23/NDPK family: From metastasis control to cilia movement. Mol. Cell Biochem. 2009, 329, 51–62. [Google Scholar] [CrossRef] [PubMed]
  55. Agarwal, R.P.; Robinson, B.; Parks, R.E. Nucleoside diphosphokinase from erythrocytes. Methods Enzymol. 1978, 51, 376–386. [Google Scholar] [PubMed]
  56. Ma, D.; McCorkle, J.R.; Kaetzel, D.M. The metastasis suppressor NM23-H1 possesses 3′–5′ exonuclease activity. J. Biol. Chem. 2004, 279, 18073–18084. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, Q.; McCorkle, J.R.; Novak, M.; Yang, M.; Kaetzel, D.M. Metastasis suppressor function of NM23-H1 requires its 3′–5′ exonuclease activity. Int. J. Cancer 2011, 128, 40–50. [Google Scholar] [CrossRef] [PubMed]
  58. Yang, M.; Jarrett, S.G.; Craven, R.; Kaetzel, D.M. YNK1, the yeast homolog of human metastasis suppressor NM23, is required for repair of UV radiation- and etoposide-induced DNA damage. Mutat. Res. 2009, 660, 74–78. [Google Scholar] [CrossRef] [PubMed]
  59. Jarrett, S.G.; Novak, M.; Dabernat, S.; Daniel, J.Y.; Mellon, I.; Zhang, Q.; Harris, N.; Ciesielski, M.J.; Fenstermaker, R.A.; Kovacic, D.; et al. Metastasis suppressor NM23-H1 promotes repair of UV-induced DNA damage and suppresses UV-induced melanomagenesis. Cancer Res. 2012, 72, 133–143. [Google Scholar] [CrossRef] [PubMed]
  60. Wei, Q.; Cheng, L.; Xie, K.; Bucana, C.D.; Dong, Z. Direct correlation between DNA repair capacity and metastatic potential of K-1735 murine melanoma cells. J. Investig. Dermatol. 1997, 108, 3–6. [Google Scholar] [CrossRef] [PubMed]
  61. Kauffmann, A.; Rosselli, F.; Lazar, V.; Winnepenninckx, V.; Mansuet-Lupo, A.; Dessen, P.; van den Oord, J.J.; Spatz, A.; Sarasin, A. High expression of DNA repair pathways is associated with metastasis in melanoma patients. Oncogene 2008, 27, 565–573. [Google Scholar] [CrossRef] [PubMed]
  62. Jewell, R.; Conway, C.; Mitra, A.; Randerson-Moor, J.; Lobo, S.; Nsengimana, J.; Harland, M.; Marples, M.; Edward, S.; Cook, M.; et al. Patterns of expression of DNA repair genes and relapse from melanoma. Clin. Cancer Res. 2010, 16, 5211–5221. [Google Scholar] [CrossRef] [PubMed]
  63. Li, C.; Yin, M.; Wang, L.-E.; Amos, C.I.; Zhu, D.; Lee, J.E.; Gershenwald, J.E.; Grimm, E.A.; Wei, Q. Polymorphisms of nucleotide excision repair genes predict melanoma survival. J. Investig. Dermatol. 2014, 133, 1813–1821. [Google Scholar] [CrossRef] [PubMed]
  64. 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] [PubMed]
  65. McCorkle, J.R.; Leonard, M.K.; Kraner, S.D.; Blalock, E.M.; Ma, D.; Zimmer, S.G.; Kaetzel, D.M. The metastasis suppressor NME1 regulates expression of genes linked to metastasis and patient outcome in melanoma and breast carcinoma. Cancer Genom. Proteom. 2014, 11, 175–194. [Google Scholar]
  66. Horak, C.E.; Lee, J.H.; Elkahloun, A.G.; Boissan, M.; Dumont, S.; Maga, T.K.; Arnaud-Dabernat, S.; Palmieri, D.; Stetler-Stevenson, W.G.; Lacombe, M.L.; et al. Nm23-H1 suppresses tumor cell motility by down-regulating the lysophosphatidic acid receptor EDG2. Cancer Res. 2007, 67, 7238–7246. [Google Scholar] [CrossRef] [PubMed]
  67. Horak, C.E.; Mendoza, A.; Vega-Valle, E.; Albaugh, M.; Graff-Cherry, C.; McDermott, W.G.; Hua, E.; Merino, M.J.; Steinberg, S.M.; Khanna, C.; et al. Nm23-H1 suppresses metastasis by inhibiting expression of the lysophosphatidic acid receptor EDG2. Cancer Res. 2007, 67, 11751–11759. [Google Scholar] [CrossRef] [PubMed]
  68. Postel, E.H.; Berberich, S.J.; Flint, S.J.; Ferrone, C.A. Human c-myc transcription factor PuF identified as nm23-H2 nucleoside diphosphate kinase, a candidate suppressor of tumor metastasis. Science 1993, 261, 478–480. [Google Scholar] [CrossRef] [PubMed]
  69. Ma, D.; Xing, Z.; Liu, B.; Pedigo, N.; Zimmer, S.; Bai, Z.; Postel, E.; Kaetzel, D.M. NM23-H1 cleaves and represses transcriptional activity of nuclease-hypersensitive elements in the PDGF-A promoter. J. Biol. Chem. 2002, 277, 1560–1567. [Google Scholar] [CrossRef] [PubMed]
  70. Postel, E.; Berberich, S.J.; Rooney, J.W.; Kaetzel, D.M. Human NM23/nucleoside diphosphate kinase regulates gene expression through DNA binding to nuclease-hypersensitive elements. J. Bioenerg. Biomembr. 2000, 32, 277–284. [Google Scholar] [CrossRef] [PubMed]
  71. Cervoni, L.; Pietrangeli, P.; Chichiarelli, S.; Altieri, F.; Egistelli, L.; Turano, C.; Lascu, I.; Giartosio, A. In vivo cross-linking of nm23/nucleoside diphosphate kinase to the PDGF-A gene promoter. Mol. Biol. Rep. 2003, 30, 33–40. [Google Scholar] [CrossRef] [PubMed]
  72. Cervoni, L.; Egistelli, L.; Eufemi, M.; d’Abusco, A.S.; Altieri, F.; Lascu, I.; Turano, C.; Giartosio, A. DNA sequences acting as binding sites for NM23/NDPK proteins in melanoma M14 cells. J. Cell Biochem. 2006, 98, 421–428. [Google Scholar] [CrossRef] [PubMed]
  73. Moses, R.E.; O’Malley, B.W. DNA transcription and repair: A confluence. J. Biol. Chem. 2012, 287, 23266–23270. [Google Scholar] [CrossRef] [PubMed]
  74. Postel, E.H.; Wohlman, I.; Zou, X.; Juan, T.; Sun, N.; D’Agostin, D.; Cuellar, M.; Choi, T.; Notterman, D.A.; la Perle, K.M. Targeted deletion of Nm23/nucleoside diphosphate kinase A and B reveals their requirement for definitive erythropoiesis in the mouse embryo. Dev. Dyn. 2009, 238, 775–787. [Google Scholar] [CrossRef] [PubMed]
  75. Postel, E.H.; Zou, X.; Notterman, D.A.; la Perle, K.M. Double knockout Nme1/Nme2 mouse model suggests a critical role for NDP kinases in erythroid development. Mol. Cell Biochem. 2009, 329, 45–50. [Google Scholar] [CrossRef] [PubMed]
  76. Arnaud-Dabernat, S.; Bourbon, P.M.; Dierich, A.; le Meur, M.; Daniel, J.-Y. Knockout mice as model systems for studying nm23/NDP kinase gene functions. Application to the nm23-M1 gene. J. Bioenerg. Biomembr. 2003, 35, 19–30. [Google Scholar] [CrossRef] [PubMed]
  77. Di, L.; Srivastava, S.; Zhdanova, O.; Sun, Y.; Li, Z.; Skolnik, E.Y. Nucleoside diphosphate kinase B knock-out mice have impaired activation of the K+ channel KCa3.1, resulting in defective T cell activation. J. Biol. Chem. 2010, 285, 38765–38771. [Google Scholar] [PubMed]
  78. Ha, L.; Ichikawa, T.; Anver, M.; Dickins, R.; Lowe, S.; Sharpless, N.E.; Krimpenfort, P.; DePinho, R.A.; Bennett, D.C.; Sviderskaya, E.V.; et al. ARF functions as a melanoma tumor suppressor by inducing p53-independent senescence. Proc. Natl. Acad. Sci. USA 2007, 104, 10968–10973. [Google Scholar] [CrossRef] [PubMed]
  79. Zaidi, M.R.; Hornyak, T.J.; Merlino, G. A genetically engineered mouse model with inducible GFP expression in melanocytes. Pigment Cell Melanoma Res. 2011, 24, 393–394. [Google Scholar] [CrossRef] [PubMed]
  80. Zaidi, M.R.; Davis, S.; Noonan, F.P.; Graff-Cherry, C.; Hawley, T.S.; Walker, R.L.; Feigenbaum, L.; Fuchs, E.; Lyakh, L.; Young, H.A.; et al. Interferon-γ links ultraviolet radiation to melanomagenesis in mice. Nature 2011, 469, 548–553. [Google Scholar] [CrossRef] [PubMed]
  81. Foster, P.J.; Dunn, E.A.; Karl, K.E.; Snir, J.A.; Nycz, C.M.; Harvey, A.J.; Pettis, R.J. Cellular magnetic resonance imaging: In vivo imaging of melanoma cells in lymph nodes of mice. Neoplasia 2008, 10, 207–216. [Google Scholar]
  82. Puaux, A.L.; Ong, L.C.; Jin, Y.; Teh, I.; Hong, M.; Chow, P.K.; Golay, X.; Abastado, J.P. A comparison of imaging techniques to monitor tumor growth and cancer progression in living animals. Int. J. Mol. Imaging 2011, 2011, 321538. [Google Scholar] [CrossRef] [PubMed]
  83. Noonan, F.P.; Otsuka, T.; Bang, S.; Anver, M.R.; Merlino, G. Accelerated ultraviolet radiation-induced carcinogenesis in hepatocyte growth factor/scatter factor transgenic mice. Cancer Res. 2000, 60, 3738–3743. [Google Scholar] [PubMed]
  84. Shain, A.H.; Bastian, B.C. From melanocytes to melanomas. Nat. Rev. Cancer 2016, 16, 345–358. [Google Scholar] [CrossRef] [PubMed]
  85. Tas, F. Metastatic behavior in melanoma: Timing, pattern, survival, and influencing factors. J. Oncol. 2012, 2012, 647684. [Google Scholar] [CrossRef] [PubMed]
  86. Dankort, D.; Curley, D.P.; Cartlidge, R.A.; Nelson, B.; Karnezis, A.N.; Damsky, W.E., Jr.; You, M.J.; DePinho, R.A.; McMahon, M.; Bosenberg, M. BrafV600E cooperates with Pten loss to induce metastatic melanoma. Nat. Genet. 2009, 41, 544–552. [Google Scholar] [CrossRef] [PubMed]
  87. Kircher, D.A.; Arave, R.A.; Cho, J.H.; Holmen, S.L. Melanoma metastases caught in the AKT. Mol. Cell Oncol. 2016, 3, e1128516. [Google Scholar] [CrossRef] [PubMed]
  88. Ha, L.; Noonan, F.P.; de Fabo, E.C.; Merlino, G. Animal models of melanoma. J. Investig. Dermatol. Symp. Proc. 2005, 10, 86–88. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Ultraviolet radiation-induced melanoma in the hepatocyte HGF/SF transgenic mouse (Jarrett et al., 2013) [15]. EPI-MEL JUNCs, epithelial-melanoma junctions; TDI, transdermal invasion with invasion of subcutis.
Figure 1. Ultraviolet radiation-induced melanoma in the hepatocyte HGF/SF transgenic mouse (Jarrett et al., 2013) [15]. EPI-MEL JUNCs, epithelial-melanoma junctions; TDI, transdermal invasion with invasion of subcutis.
Ijms 18 01647 g001
Figure 2. The HGF/SF transgenic mouse as a genetically-engineered mouse model (GEMM) sensor for metastasis-regulating activity (MRG) of genes. Schematic depicts the HGF/SF transgenic mouse as a “Sensor” to be bred (X) with “Test” GEMMs harboring altered expression of candidate MRGs.
Figure 2. The HGF/SF transgenic mouse as a genetically-engineered mouse model (GEMM) sensor for metastasis-regulating activity (MRG) of genes. Schematic depicts the HGF/SF transgenic mouse as a “Sensor” to be bred (X) with “Test” GEMMs harboring altered expression of candidate MRGs.
Ijms 18 01647 g002
Figure 3. Ablation of the NME1/NME1 locus confers lymph node and organ metastasi to UV-induces melanomas in the HGF/SF transgenic mouse. Schematic depicts the HGF/SF transgenic mouse as a “Sensor GEMM” and the NME1+/−;NME2+/− transgenic strain as the “Test GEMM.” Percentages represent percent incidence of mice bearing melanin-pigment metastatic lesions in organs shown. Images have appeared previously in ref. 10 and are used herein with the permission of the journal.
Figure 3. Ablation of the NME1/NME1 locus confers lymph node and organ metastasi to UV-induces melanomas in the HGF/SF transgenic mouse. Schematic depicts the HGF/SF transgenic mouse as a “Sensor GEMM” and the NME1+/−;NME2+/− transgenic strain as the “Test GEMM.” Percentages represent percent incidence of mice bearing melanin-pigment metastatic lesions in organs shown. Images have appeared previously in ref. 10 and are used herein with the permission of the journal.
Ijms 18 01647 g003

Share and Cite

MDPI and ACS Style

Leonard, M.K.; Pamidimukkala, N.; Puts, G.S.; Snyder, D.E.; Slominski, A.T.; Kaetzel, D.M. The HGF/SF Mouse Model of UV-Induced Melanoma as an In Vivo Sensor for Metastasis-Regulating Gene. Int. J. Mol. Sci. 2017, 18, 1647. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081647

AMA Style

Leonard MK, Pamidimukkala N, Puts GS, Snyder DE, Slominski AT, Kaetzel DM. The HGF/SF Mouse Model of UV-Induced Melanoma as an In Vivo Sensor for Metastasis-Regulating Gene. International Journal of Molecular Sciences. 2017; 18(8):1647. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081647

Chicago/Turabian Style

Leonard, M. Kathryn, Nidhi Pamidimukkala, Gemma S. Puts, Devin E. Snyder, Andrzej T. Slominski, and David M. Kaetzel. 2017. "The HGF/SF Mouse Model of UV-Induced Melanoma as an In Vivo Sensor for Metastasis-Regulating Gene" International Journal of Molecular Sciences 18, no. 8: 1647. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18081647

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