Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cancer Res ; 75(24): 5228-34, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26627015

ABSTRACT

Merkel cell carcinoma (MCC) is an uncommon, but highly malignant, cutaneous tumor. Merkel cell polyoma virus (MCV) has been implicated in a majority of MCC tumors; however, viral-negative tumors have been reported to be more prevalent in some geographic regions subject to high sun exposure. While the impact of MCV and viral T-antigens on MCC development has been extensively investigated, little is known about the etiology of viral-negative tumors. We performed targeted capture and massively parallel DNA sequencing of 619 cancer genes to compare the gene mutations and copy number alterations in MCV-positive (n = 13) and -negative (n = 21) MCC tumors and cell lines. We found that MCV-positive tumors displayed very low mutation rates, but MCV-negative tumors exhibited a high mutation burden associated with a UV-induced DNA damage signature. All viral-negative tumors harbored mutations in RB1, TP53, and a high frequency of mutations in NOTCH1 and FAT1. Additional mutated or amplified cancer genes of potential clinical importance included PI3K (PIK3CA, AKT1, PIK3CG) and MAPK (HRAS, NF1) pathway members and the receptor tyrosine kinase FGFR2. Furthermore, looking ahead to potential therapeutic strategies encompassing immune checkpoint inhibitors such as anti-PD-L1, we also assessed the status of T-cell-infiltrating lymphocytes (TIL) and PD-L1 in MCC tumors. A subset of viral-negative tumors exhibited high TILs and PD-L1 expression, corresponding with the higher mutation load within these cancers. Taken together, this study provides new insights into the underlying biology of viral-negative MCC and paves the road for further investigation into new treatment opportunities.


Subject(s)
Carcinoma, Merkel Cell/genetics , DNA Damage/radiation effects , Skin Neoplasms/genetics , Ultraviolet Rays/adverse effects , Blotting, Western , Cells, Cultured , DNA Mutational Analysis , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Merkel cell polyomavirus , Mutation , Polymerase Chain Reaction , Transcriptome
2.
Exp Dermatol ; 23(12): 916-21, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25346513

ABSTRACT

Dopachrome tautomerase (DCT) is involved in the formation of the photoprotective skin pigment eumelanin and has also been shown to have a role in response to apoptotic stimuli and oxidative stress. The effect of DCT on UVR DNA damage responses and survival pathways in human melanocytic cells was examined by knockdown experiments using melanoma cells, neonatal foreskin melanoblasts (MB) in monoculture and in co-culture with human keratinocytes. MB cell strains genotyped as either MC1R WT or MC1R RHC homozygotes, which are known to be deficient in DCT, were transduced with lentivirus vectors for either DCT knockdown or overexpression. We found melanoma cell survival was reduced by DCT depletion and by UVR over time. UVR-induced p53 and pp53-Ser15 levels were reduced with DCT depletion. Knockdown of DCT in MC1R WT and MC1R RHC MB cells reduced their survival after UVR exposure, whereas increased DCT protein levels enhanced survival. DCT depletion reduced p53 and pp53-Ser15 levels in WM266-4 melanoma and MC1R WT MB cells, while MC1R RHC MB cells displayed variable levels. Both MC1R WT and RHC genotypes of MB cells were responsive to UVR at 3 h with increases in both p53 and pp53-Ser15 proteins. MC1R WT MB cell strains in coculture with keratinocytes have an increased cell survival after UVR exposure when compared to those in monoculture, a protective effect which appears to be conferred by the keratinocytes.


Subject(s)
Cell Survival/radiation effects , Intramolecular Oxidoreductases/metabolism , Melanocytes/metabolism , Melanocytes/radiation effects , Ultraviolet Rays/adverse effects , Coculture Techniques , DNA Damage , Gene Knockdown Techniques , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/genetics , Keratinocytes/metabolism , Melanocytes/cytology , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Up-Regulation
3.
J Invest Dermatol ; 132(5): 1452-61, 2012 May.
Article in English | MEDLINE | ID: mdl-22336944

ABSTRACT

Variant alleles of the human melanocortin 1 receptor (MC1R) reduce the ability of melanocytes to produce the dark pigment eumelanin, with R alleles being most deficient. Cultured melanocytes of MC1R R/R variant genotype give reduced responses to [Nle(4), D-Phe(7)]α-melanocyte-stimulating hormone (NDP-MSH) ligand stimulation and lower levels of DNA repair than MC1R wild-type strains. p38 controls xeroderma pigmentosum (XP)-C recruitment to DNA damage sites through regulating ubiquitylation of the DNA damage-binding protein 2 (DDB2) protein, and p53 is implicated in the nuclear excision repair process through its regulation of XP-C and DDB2 protein expression. We report the effects of MC1R ligand treatment and UVR exposure on phosphorylation of p38 and p53, and DDB2 protein expression in MC1R variant strains. Wild-type MC1R melanocyte strains grown together with keratinocytes in coculture, when treated with NDP-MSH and exposed to UVR, gave synergistic activation of p38 and p53 phosphorylation, and were not replicated by R/R variant melanocytes, which have lower basal levels of phosphorylated forms of p38. Minor increases in p38 phosphorylation status in R/R variant melanocyte cocultures could be attributed to the keratinocytes alone. We also found that MC1R wild-type strains regulate DDB2 protein levels through p38, but MC1R R/R variant melanocytes do not. This work confirms the important functional role that the MC1R receptor plays in UVR stress-induced DNA repair.


Subject(s)
Alleles , DNA-Binding Proteins/metabolism , Receptor, Melanocortin, Type 1/genetics , Tumor Suppressor Protein p53/metabolism , alpha-MSH/analogs & derivatives , p38 Mitogen-Activated Protein Kinases/metabolism , Cells, Cultured , Coculture Techniques , DNA Repair/genetics , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/radiation effects , Humans , Keratinocytes , Melanocytes/drug effects , Melanocytes/radiation effects , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/radiation effects , Signal Transduction , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/radiation effects , Ultraviolet Rays , alpha-MSH/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/radiation effects
4.
J Clin Virol ; 52(3): 272-5, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21907614

ABSTRACT

BACKGROUND: Merkel cell polyomavirus (MCPyV) is present in approximately 80% of human Merkel cell carcinomas (MCCs). A previous in silico prediction suggested MCPyV encodes a microRNA (miRNA) that may regulate cellular and viral genes. OBJECTIVES: To determine the presence and prevalence of a putative MCPyV-encoded miRNA in human MCC tumors. STUDY DESIGN: Over 30 million small RNAs from 7 cryopreserved MCC tumors and 1 perilesional sample were sequenced. 45 additional MCC tumors were examined for expression of an MCPyV-encoded mature miRNA by reverse transcription real-time PCR. RESULTS: An MCPyV-encoded mature miRNA, "MCV-miR-M1-5p", was detected by direct sequencing in 2 of 3 MCPyV-positive MCC tumors. Although a precursor miRNA, MCV-miR-M1, had been predicted in silico and studied in vitro by Seo et al., no MCPyV-encoded miRNAs have been directly detected in human tissues. Importantly, the mature sequence of MCV-miR-M1 found in vivo was identical in all 79 reads obtained but differed from the in silico predicted mature miRNA by a 2-nucleotide shift, resulting in a distinct seed region and a different set of predicted target genes. This mature miRNA was detected by real-time PCR in 50% of MCPyV-positive MCCs (n = 38) and in 0% of MCPyV-negative MCCs (n = 13). CONCLUSIONS: MCV-miR-M1-5p is expressed at low levels in 50% of MCPyV-positive MCCs. This virus-encoded miRNA is predicted to target genes that may play a role in promoting immune evasion and regulating viral DNA replication.


Subject(s)
Carcinoma, Merkel Cell/virology , Merkel cell polyomavirus/genetics , MicroRNAs/genetics , Polyomavirus Infections/virology , Base Sequence , Carcinoma, Merkel Cell/diagnosis , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/pathology , DNA, Viral/biosynthesis , Humans , MicroRNAs/biosynthesis , Polyomavirus Infections/genetics , Polyomavirus Infections/pathology , RNA, Viral/biosynthesis , Real-Time Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, RNA
5.
Exp Dermatol ; 20(8): 681-4, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21615505

ABSTRACT

A co-culture system of melanocytic cells and keratinocytes was used to examine dendricity and dopachrome tautomerase (DCT) responses in low penetrant 'r' homozygote and 'R/+' heterozygote MC1R variant allele expressing cells compared to that of wild-type (WT) cells. The V60L-/- homozygote r variant cells showed similar responses to ligand as WT MC1R strains, while V92M-/- homozygote r variant cells were generally shown to have greater dendricity and express higher DCT than the WT cells, even at basal levels. The R151C+/- heterozygote cells showed similar responses to WT cells, while the R160W+/- and D294H+/- variant cells were reduced in their responses to NDP-MSH, but still had an active cAMP response with forskolin treatment. These responses are consistent with the dominant negative effect of these alleles on the MC1R WT allele that has previously been demonstrated genetically and biochemically.


Subject(s)
Alleles , Intramolecular Oxidoreductases/metabolism , Keratinocytes/metabolism , Melanocyte-Stimulating Hormones/pharmacology , Melanocytes/metabolism , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Coculture Techniques , Colforsin/pharmacology , Cyclic AMP/metabolism , Heterozygote , Homozygote , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Male , Melanocytes/cytology , Melanocytes/drug effects
6.
J Clin Oncol ; 29(12): 1539-46, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21422430

ABSTRACT

PURPOSE: Merkel cell carcinoma (MCC) is a polyomavirus-associated skin cancer that is frequently lethal and lacks established prognostic biomarkers. This study sought to identify biomarkers that improve prognostic accuracy and provide insight into MCC biology. PATIENTS AND METHODS: Gene expression profiles of 35 MCC tumors were clustered based on prognosis. The cluster of genes overexpressed in good-prognosis tumors was tested for biologic process enrichment. Relevant mRNA expression differences were confirmed by quantitative polymerase chain reaction and immunohistochemistry. An independent set of 146 nonoverlapping MCC tumors (median follow-up, 25 months among 116 living patients) was employed for biomarker validation. Univariate and multivariate Cox regression analyses were performed. RESULTS: Immune response gene signatures were prominent in patients with good prognoses. In particular, genes associated with cytotoxic CD8+ lymphocytes were overexpressed in tumors from patients with favorable prognoses. In the independent validation set, cases with robust intratumoral CD8+ lymphocyte infiltration had improved outcomes (100% MCC-specific survival, n = 26) compared with instances characterized by sparse infiltration (60% survival, n = 120). Only stage and intratumoral CD8 infiltration (but not age, sex, or CD8+ lymphocytes localized to the tumor-stroma interface) were significant in both univariate and multivariate Cox regression analyses. Notably, traditional histologic identification of tumor-infiltrating lymphocytes was not a significant independent predictor of survival. CONCLUSION: Intratumoral CD8+ lymphocyte infiltration can be readily assessed on paraffin-embedded tissue, is independently associated with improved MCC-specific survival, and therefore, may provide prognostic information that enhances established MCC staging protocols.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Merkel Cell/diagnosis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Lymphocytes, Tumor-Infiltrating/immunology , Skin Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Austria , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/immunology , Carcinoma, Merkel Cell/mortality , Carcinoma, Merkel Cell/pathology , Cluster Analysis , Female , Gene Expression Profiling/methods , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Staging , Paraffin Embedding , Prognosis , Proportional Hazards Models , Queensland , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Risk Assessment , Risk Factors , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Rate , Time Factors , Washington
7.
Eur J Pharmacol ; 660(1): 103-10, 2011 Jun 11.
Article in English | MEDLINE | ID: mdl-21199646

ABSTRACT

The melanocortin MC(1) receptor is a G-protein coupled receptor expressed in the melanocytes of the skin and hair and is known for its key role in the regulation of human pigmentation. Melanocortin MC(1) receptor activation after ultraviolet radiation exposure results in a switch from the red/yellow pheomelanin to the brown/black eumelanin pigment synthesis within cutaneous melanocytes; this pigment is then transferred to the surrounding keratinocytes of the skin. The increase in melanin maturation and uptake results in tanning of the skin, providing a physical protection of skin cells from ultraviolet radiation induced DNA damage. Melanocortin MC(1) receptor polymorphism is widespread within the Caucasian population and some variant alleles are associated with red hair colour, fair skin, poor tanning and increased risk of skin cancer. Here we will discuss the use of mouse coat colour models, human genetic association studies, and in vitro cell culture studies to determine the complex functions of the melanocortin MC(1) receptor and the molecular mechanisms underlying the association between melanocortin MC(1) receptor variant alleles and the red hair colour phenotype. Recent research indicates that melanocortin MC(1) receptor has many non-pigmentary functions, and that the increased risk of skin cancer conferred by melanocortin MC(1) receptor variant alleles is to some extent independent of pigmentation phenotypes. The use of new transgenic mouse models, the study of novel melanocortin MC(1) receptor response genes and the use of more advanced human skin models such as 3D skin reconstruction may provide key elements in understanding the pharmacogenetics of human melanocortin MC(1) receptor polymorphism.


Subject(s)
Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Animals , Cell Line , Genotype , Humans , Melanocytes/cytology , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Phenotype , Pigmentation/genetics
8.
J Invest Dermatol ; 129(6): 1547-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19020549

ABSTRACT

Merkel cell carcinoma (MCC) is an aggressive neuroendocrine skin cancer with poorly characterized genetics. We performed high resolution comparative genomic hybridization on 25 MCC specimens using a high-density oligonucleotide microarray. Tumors frequently carried extra copies of chromosomes 1, 3q, 5p, and 6 and lost chromosomes 3p, 4, 5q, 7, 10, and 13. MCC tumors with less genomic aberration were associated with improved survival (P=0.04). Tumors from 13 of 22 MCC patients had detectable Merkel cell polyomavirus DNA, and these tumors had fewer genomic deletions. Three regions of genomic alteration were of particular interest: a deletion of 5q12-21 occurred in 26% of tumors, a deletion of 13q14-21 was recurrent in 26% of tumors and contains the well-characterized tumor suppressor RB1, and a previously unreported focal amplification at 1p34 was present in 39% of tumors and centers on L-Myc (MYCL1). L-Myc is related to the c-Myc proto-oncogene, has transforming activity, and is amplified in the closely related small cell lung cancer. Normal skin showed no L-Myc expression, whereas 4/4 MCC specimens tested expressed L-Myc RNA in relative proportion to the DNA copy number gain. These findings suggest several genes that may contribute to MCC pathogenesis, most notably L-Myc.


Subject(s)
Carcinoma, Merkel Cell/genetics , Comparative Genomic Hybridization , Gene Expression Regulation, Neoplastic , Genes, myc , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Skin Neoplasms/metabolism , Small Cell Lung Carcinoma/genetics , Aged , Aged, 80 and over , Cell Line, Tumor , Chromosome Mapping , Female , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Proto-Oncogene Mas , Recurrence
10.
J Invest Dermatol ; 129(2): 392-405, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18650849

ABSTRACT

Single nucleotide polymorphisms (SNPs) within the SLC45A2/MATP, SLC24A5/NCKX5, and OCA2/P genes have been associated with natural variation of pigmentation traits in human populations. Here, we describe the characterization of human primary melanocytic cells genotyped for polymorphisms within the MATP, NCKX5, or OCA2 loci. On the basis of genotype, these cultured cells reflect the phenotypes observed by others in terms of both melanin content and tyrosinase (TYR) activity when comparing skin designated as either "White" or "Black". We found a statistically significant association of MATP-374L (darker skin) with higher TYR protein abundance that was not observed for any NCKX5-111 or OCA2 rs12913832 allele. MATP-374L/L homozygous strains displayed significantly lower MATP transcript levels compared to MATP-374F/F homozygous cells, but this did not reach statistical significance based on NCKX5 or OCA2 genotype. Similarly, we observed significantly increased levels of OCA2 mRNA in rs12913832-T (brown eye) homozygotes compared to rs12913832-C (blue eye) homozygous strains, which was not observed for MATP or NCKX5 gene transcripts. In genotype-phenotype associations performed on a collection of 226 southern European individuals using these same SNPs, we were able to show strong correlations in MATP-L374F, OCA2, and melanocortin-1 receptor with skin, eye, and hair color variation, respectively.


Subject(s)
Antigens, Neoplasm/genetics , Antiporters/genetics , Melanocytes/physiology , Membrane Transport Proteins/genetics , Skin Pigmentation/genetics , Cells, Cultured , Eye Color/genetics , Gene Expression Regulation/physiology , Genotype , Hair Color/genetics , Humans , Melanins/metabolism , Melanocytes/cytology , Microphthalmia-Associated Transcription Factor/metabolism , Phenotype , Polymorphism, Single Nucleotide/physiology , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Receptor, Melanocortin, Type 1/genetics , Skin/cytology , White People/genetics
11.
J Cell Physiol ; 215(2): 344-55, 2008 May.
Article in English | MEDLINE | ID: mdl-17960564

ABSTRACT

The occurrence of red hair and pale skin in individuals, which is associated with UV-radiation sensitivity and increased skin cancer risk, is mainly due to polymorphisms in the melanocortin-1 receptor (MC1R) expressed in melanocytes. We have established a serum free human melanocyte-keratinocyte coculture system to study the behavior and functional abilities of melanocytes expressing MC1R red hair color (RHC) variants in order to identify differences from their wild type (WT) counterparts. This model revealed the importance of elevated calcium levels in promoting strong melanocyte interaction with the surrounding keratinocytes and resulted in a dendritic melanocyte morphology similar to that in skin. However, the dendricity response following agonist activation of the MC1R receptor by NDP-MSH peptide, was markedly enhanced in WT melanocytes in comparison to RHC strains. Analysis of mRNA expression and protein levels of the major pigmentation markers following NDP-MSH treatment distinguished the enzyme dopachrome tautomerase as preferentially upregulated in cocultures of WT strains, with negligible or a much reduced response in melanocytes with RHC variant alleles. These results highlight the use of the coculture system in determining fundamental differences in the physiology of melanocytes expressing RHC MC1R receptors and those of WT genotype, which are likely to contribute to the increased skin cancer risk for individuals that carry these variants.


Subject(s)
Hair Color/genetics , Keratinocytes/physiology , Melanocyte-Stimulating Hormones/metabolism , Melanocytes/physiology , Polymorphism, Genetic , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Skin Pigmentation/physiology , Biomarkers , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Ligands , Melanocytes/cytology , Melanocytes/drug effects , Melanocytes/metabolism , Osmolar Concentration , RNA, Messenger/metabolism , Up-Regulation , Vimentin/metabolism , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
12.
Peptides ; 28(12): 2387-96, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18006116

ABSTRACT

Variant alleles of the human MC1R gene are strongly associated with red hair color, fair skin and poor tanning ability (RHC-trait). Recently, we demonstrated that melanocytes harboring RHC-associated alleles have markedly reduced surface expression and/or impaired G-protein coupling of the corresponding receptor protein. The consequences of such a deficit on MC1R-mediated signaling pathways have now been quantitatively evaluated utilizing strains of human primary melanocytes homozygous for RHC-associated variant alleles and comparing responses to wild-type strains. The ability of melanocortin peptides to increase transcription of cAMP-dependent pigmentation genes, including MITF and SLC45A2, was abrogated in melanocytes with RHC-associated variant alleles, an effect that may contribute to the RHC phenotype. Activation of the c-Fos transcription factor gene was also severely compromised, a finding of potential relevance for non-pigmentary roles of MC1R. We also confirmed p38 signaling as an MC1R-regulated pathway and identified a large synergistic interaction between UV irradiation and MC1R stimulation for the activation of p38. This synergism was impaired in melanocytes expressing RHC variants of MC1R which may be relevant for the poor tanning ability associated with individuals possessing these alleles.


Subject(s)
Melanocytes/metabolism , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Alleles , Cells, Cultured , Cyclic AMP/metabolism , Genetic Variation , Hair Color/genetics , Humans , Melanocytes/drug effects , Melanocytes/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/radiation effects , Skin Pigmentation/genetics , Ultraviolet Rays , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Invest Dermatol ; 127(9): 2216-27, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17460731

ABSTRACT

Upregulation of microphthalmia-associated transcription factor (MITF) expression has been proposed to mediate melanogenesis stimulated by cAMP, whereas downregulation of MITF has been suggested to underlie the depigmentary effects of resveratrol, a promising chemotherapeutic found in red wine. We have assessed the contribution of MITF to pigmentation regulation by treating primary cultures of normal human melanocytes with the adenylate cyclase activator forskolin and/or resveratrol, then quantifying mRNA and protein levels for MITF, tyrosinase, tyrosinase-related protein-1, and dopachrome tautomerase (DCT). The inhibition of tyrosinase activity by resveratrol was not due to alterations in MITF, but instead was explained by both direct tyrosinase inhibition and a post-transcriptional effect that reduced the amount of fully processed tyrosinase. Glycosidase digestion revealed that the basis for the tyrosinase decrease was the retention of an immature form in the ER and subsequent loss of the mature, Golgi-processed enzyme. Elevation of intracellular cAMP by forskolin markedly increased protein levels for MITF, tyrosinase and DCT, however there was no concomitant increase in tyrosinase or DCT mRNA. This indicated that elevated levels of MITF were not sufficient to promote transcription of these melanogenic genes and that the increase in their protein abundance appeared to be predominantly mediated through post-transcriptional processing events.


Subject(s)
Cyclic AMP/metabolism , Gene Expression Regulation , Melanins/biosynthesis , RNA Processing, Post-Transcriptional , Stilbenes/pharmacology , Cells, Cultured/metabolism , Colforsin/metabolism , Enzyme Inhibitors/pharmacology , Golgi Apparatus/metabolism , Humans , Intramolecular Oxidoreductases/metabolism , Levodopa/metabolism , Melanocytes/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Resveratrol , Transcription, Genetic
14.
J Invest Dermatol ; 126(5): 1135-42, 2006 May.
Article in English | MEDLINE | ID: mdl-16498399

ABSTRACT

Merkel cell carcinoma (MCC) is a highly metastatic skin tumor. To assess the relevance of the Ras/Raf/MEK/MAP kinase pathway, we analyzed for activating B-Raf mutations and we elucidated the presence of the Raf Kinase Inhibitor Protein (RKIP) and extracellular signal-regulated kinase (ERK) as well as the phosphorylation status of ERK. All MCC samples were negative for the B-Raf(V600E) mutation. Remarkably, RKIP, which was shown to interfere with the activation of MEK by Raf, was highly expressed in primary as well as in metastatic MCC. Immunohistochemical analysis of the phosphorylation status of ERK revealed in 42 out of 44 samples a complete lack of activated ERK in the tumor cells although ERK is expressed; in the two positive cases phosphorylated ERK was restricted to a minor fraction of the tumor cells. Western blot analysis of three MCC-derived cell lines revealed in one case the pattern present in situ (i.e. high RKIP expression and complete absence of phosphorylated ERK). In summary, our data demonstrate the inactivity of the classical MAP kinase signal transduction pathway in MCC, which seems to be because of lack of activation as well as active deactivation. These findings should be accounted for in future therapeutic approaches for this tumor.


Subject(s)
Carcinoma, Merkel Cell/metabolism , MAP Kinase Signaling System/physiology , Skin Neoplasms/metabolism , Androgen-Binding Protein/analysis , Carcinoma, Merkel Cell/pathology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunohistochemistry , Mutation , Phosphatidylethanolamine Binding Protein , Phosphorylation , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/pathology
15.
Hum Mol Genet ; 14(15): 2145-54, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15972726

ABSTRACT

The human melanocortin-1 receptor gene (MC1R) encodes a G-protein coupled receptor that is primarily expressed on melanocytes, where it plays a key role in pigmentation regulation. Variant alleles are associated with red hair colour and fair skin, known as the RHC phenotype, as well as skin cancer risk. The R151C, R160W and D294H alleles, designated 'R', are strongly associated with the RHC phenotype and have been proposed to result in loss of function receptors due to impaired G-protein coupling. We recently provided evidence that the R151C and R160W variants can efficiently couple to G-proteins in response to alpha-melanocyte stimulating hormone. The possibility that altered cellular localization of the R151C and R160W variant receptors could underlie their association with RHC was therefore considered. Using immunofluorescence and ligand binding studies, we found that melanocytic cells exogenously or endogenously expressing MC1R show strong surface localization of the wild-type and D294H alleles but markedly reduced cell surface expression of the R151C and R160W receptors. In additional exogenous expression studies, the R variant D84E and the rare I155T variant, also demonstrated a significant reduction in plasma membrane receptor numbers. The V60L, V92M and R163Q weakly associated RHC alleles, designated 'r', were expressed with normal or intermediate cell surface receptor levels. These results indicate that reduced receptor coupling activity may not be the only contributing factor to the genetic association between the MC1R variants and the RHC phenotype, with MC1R polymorphisms now linked to a change in receptor localization.


Subject(s)
Alleles , Hair Color/genetics , Receptor, Melanocortin, Type 1/metabolism , Skin Neoplasms/genetics , Cell Line , Cell Membrane/metabolism , Fluorescent Antibody Technique , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Melanocytes/metabolism , Odds Ratio , Phenotype , Radioligand Assay , Receptor, Melanocortin, Type 1/genetics , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Transfection , Tumor Cells, Cultured
16.
Exp Cell Res ; 308(1): 222-35, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15896776

ABSTRACT

Investigations into pigment cell biology have relied on the ability to culture both murine and human melanocytes, numerous melanoma cell lines and more recently, murine and human melanoblasts. Melanoblast culture requires medium supplemented with a range of growth factors including Stem Cell Factor, Endothelin-3 and Fibroblast Growth Factor-2, withdrawal of which causes the cells to differentiate into melanocytes. Using the human melanoblast culture system, we have now examined the expression and/or DNA binding activity of several transcription factors implicated in melanocytic development and differentiation. Of these, the POU domain factor BRN2 and the SOX family member SOX10 are both highly expressed in unpigmented melanocyte precursors but are down-regulated upon differentiation. In contrast, the expression levels of the previously described MITF and PAX3 transcription factors remain relatively constant during the melanoblast-melanocyte transition. Moreover, BRN2 ablated melanoma cells lack expression of SOX10 and MITF but retain PAX3. A novel finding implicates a second SOX protein, SOX9, as a potential melanogenic transcriptional regulator, as its expression level is increased following the down-regulation of BRN2 and SOX10 in differentiated melanoblasts. Our results suggest that a complex network of transcription factor interactions requiring proper temporal coordination is necessary for acquisition and maintenance of the melanocytic phenotype.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation , High Mobility Group Proteins/genetics , Melanocytes/cytology , Melanocytes/metabolism , Transcription Factors/genetics , Cell Differentiation/physiology , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/drug effects , Down-Regulation , High Mobility Group Proteins/biosynthesis , High Mobility Group Proteins/drug effects , Homeodomain Proteins/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , POU Domain Factors , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Receptor, Endothelin B/drug effects , Receptor, Endothelin B/genetics , SOX9 Transcription Factor , SOXE Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/drug effects , Transcription Factors/metabolism
17.
Oncogene ; 23(15): 2732-42, 2004 Apr 08.
Article in English | MEDLINE | ID: mdl-14755241

ABSTRACT

Merkel cell carcinoma (MCC) is a rare aggressive skin tumor which shares histopathological and genetic features with small-cell lung carcinoma (SCLC), both are of neuroendocrine origin. Comparable to SCLC, MCC cell lines are classified into two different biochemical subgroups designated as 'Classic' and 'Variant'. With the aim to identify typical gene-expression signatures associated with these phenotypically different MCC cell lines subgroups and to search for differentially expressed genes between MCC and SCLC, we used cDNA arrays to profile 10 MCC cell lines and four SCLC cell lines. Using significance analysis of microarrays, we defined a set of 76 differentially expressed genes that allowed unequivocal identification of Classic and Variant MCC subgroups. We assume that the differential expression levels of some of these genes reflect, analogous to SCLC, the different biological and clinical properties of Classic and Variant MCC phenotypes. Therefore, they may serve as useful prognostic markers and potential targets for the development of new therapeutic interventions specific for each subgroup. Moreover, our analysis identified 17 powerful classifier genes capable of discriminating MCC from SCLC. Real-time quantitative RT-PCR analysis of these genes on 26 additional MCC and SCLC samples confirmed their diagnostic classification potential, opening opportunities for new investigations into these aggressive cancers.


Subject(s)
Carcinoma, Merkel Cell/genetics , Carcinoma, Small Cell/genetics , Gene Expression Profiling/methods , Lung Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Skin Neoplasms/genetics , Biomarkers, Tumor , Cell Line, Tumor , Cluster Analysis , DNA, Complementary/metabolism , Diagnosis, Differential , Gene Expression Regulation, Neoplastic , Humans , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
18.
Pigment Cell Res ; 16(3): 198-207, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12753386

ABSTRACT

Recent population studies have demonstrated an association with the red-hair and fair-skin phenotype with variant alleles of the melanocortin-1 receptor (MC1R) which result in amino acid substitutions within the coding region leading to an altered receptor activity. In particular, Arg151Cys, Arg160Trp and Asp294His were the most commonly associated variants seen in the south-east Queensland population with at least one of these alleles found in 93% of those with red hair. In order to study the individual effects of these variants on melanocyte biology and melanocytic pigmentation, we established a series of human melanocyte strains genotyped for the MC1R receptor which included wild-type consensus, variant heterozygotes, compound heterozygotes and homozygotes for Arg151Cys, Arg160Trp, Val60Leu and Val92Met alleles. These strains ranged from darkly pigmented to amelanotic, with all strains of consensus sequence having dark pigmentation. UV sensitivity was found not to be associated with either MC1R genotype or the level of pigmentation with a range of sensitivities seen across all genotypes. Ultrastructural analysis demonstrated that while consensus strains contained stage IV melanosomes in their terminal dendrites, Arg151Cys and Arg160Trp homozygote strains contained only stage II melanosomes. This was despite being able to show expression of tyrosinase and tyrosinase-related protein-1 markers, although at reduced levels and an ability to convert exogenous 3,4-dihydroxyphenyl-alanine (DOPA) to melanin in these strains.


Subject(s)
Melanocytes/metabolism , Melanocytes/ultrastructure , alpha-MSH/biosynthesis , alpha-MSH/genetics , Alleles , Antigens/chemistry , Arginine/chemistry , Cell Survival , Cells, Cultured , Cystine/chemistry , DNA/metabolism , Dihydroxyphenylalanine/chemistry , Dihydroxyphenylalanine/metabolism , Genetic Markers , Genotype , Heterozygote , Homozygote , Humans , Male , Melanins/chemistry , Melanins/metabolism , Melanoma/metabolism , Melanosomes/metabolism , Microscopy, Electron , Microscopy, Fluorescence , Monophenol Monooxygenase/metabolism , Phenotype , Pigmentation , Tryptophan/chemistry , Ultraviolet Rays
19.
Pigment Cell Res ; 16(3): 266-72, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12753400

ABSTRACT

We have examined melanocortin-1 receptor (MC1R) variant allele frequencies in the general population and in a collection of adolescent dizygotic and monozygotic twins to determine statistical associations of pigmentation phenotypes with increased skin cancer risk. This included hair and skin color, freckling, mole count and sun exposed skin reflectance. Nine variants were studied and designated as either strong R (OR = 63; 95% CI 32-140) or weak r (OR = 5; 95% CI 3-11) red hair alleles. Penetrance of each MC1R variant allele was consistent with an allelic model where effects were multiplicative for red hair but additive for skin reflectance. To assess the interaction of the brown eye color gene BEY2/OCA2 on the phenotypic effects of variant MC1R alleles we imputed OCA2 genotype in the twin collection. A modifying effect of OCA2 on MC1R variant alleles was seen on constitutive skin color, freckling and mole count. In order to study the individual effects of these variants on pigmentation phenotype we have established a series of human primary melanocyte strains genotyped for the MC1R receptor. These include strains which are MC1R wild-type consensus, variant heterozygotes, and homozygotes for strong R alleles Arg151Cys and Arg160Trp. Ultrastructural analysis demonstrated that only consensus strains contained stage III and IV melanosomes in their terminal dendrites whereas Arg151Cys and Arg160Trp homozygous strains contained only immature stage I and II melanosomes. Such genetic association studies combined with the functional analysis of MC1R variant alleles in melanocytic cells should provide a link in understanding the association between pigmentary phototypes and skin cancer risk.


Subject(s)
Polymorphism, Genetic , Receptor, Melanocortin, Type 1/genetics , Skin Neoplasms/genetics , Alleles , Cells, Cultured , Diseases in Twins , Eye Color/genetics , Genetic Variation , Genotype , Hair Color/genetics , Heterozygote , Homozygote , Humans , Melanocytes/metabolism , Phenotype , Risk , Twins, Dizygotic , Twins, Monozygotic
20.
J Invest Dermatol ; 121(5): 1150-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14708619

ABSTRACT

The BRN2 transcription factor (POU3F2, N-Oct-3) has been implicated in development of the melanocytic lineage and in melanoma. Using a low calcium medium supplemented with stem cell factor, fibroblast growth factor-2, endothelin-3 and cholera toxin, we have established and partially characterised human melanocyte precursor cells, which are unpigmented, contain immature melanosomes and lack L-dihydroxyphenylalanine reactivity. Melanoblast cultures expressed high levels of BRN2 compared to melanocytes, which decreased to a level similar to that of melanocytes when cultured in medium that contained phorbol ester but lacked endothelin-3, stem cell factor and fibroblast growth factor-2. This decrease in BRN2 accompanied a positive L-dihydroxyphenylalanine reaction and induction of melanosome maturation consistent with melanoblast differentiation seen during development. Culture of primary melanocytes in low calcium medium supplemented with stem cell factor, fibroblast growth factor-2 and endothelin-3 caused an increase in BRN2 protein levels with a concomitant change to a melanoblast-like morphology. Synergism between any two of these growth factors was required for BRN2 protein induction, whereas all three factors were required to alter melanocyte morphology and for maximal BRN2 protein expression. These finding implicate BRN2 as an early marker of melanoblasts that may contribute to the hierarchy of melanocytic gene control.


Subject(s)
Endothelin-3/pharmacology , Fibroblast Growth Factor 2/pharmacology , Melanocytes/cytology , Neural Crest/cytology , Stem Cell Factor/pharmacology , Stem Cells/cytology , Transcription Factors/analysis , Antigens, Neoplasm , Cell Differentiation , Cells, Cultured , Homeodomain Proteins , Humans , Immunohistochemistry , MART-1 Antigen , Melanocytes/chemistry , Monophenol Monooxygenase/analysis , Neoplasm Proteins/analysis , POU Domain Factors , Stem Cells/chemistry , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...