Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mamm Genome ; 22(9-10): 602-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21626174

ABSTRACT

Cutaneous melanoma arises from transformed melanocytes and is caused mainly by environmental effects such as ultraviolet radiation and to a lesser extent by predisposing genetic variants. Only a few susceptibility genes for cutaneous melanoma have been identified so far in human; therefore, animal models represent a valuable alternative for genetic studies of this disease. In a previous quantitative trait locus (QTL) study, several susceptibility regions were identified in a swine biomedical model, the MeLiM (Melanoblastoma-bearing Libechov minipig) pigs. This article details the fine-mapping of a QTL located on SSC13 (Sus scrofa chromosome 13) through an increase in marker density. New microsatellites were used to confirm the results of the first analysis, and MITF (microphthalmia-associated transcription factor) was selected as a candidate gene for melanoma development. A single-marker association analysis was performed with single-nucleotide polymorphisms (SNPs) spread over the locus, but it did not reveal a significant association with diverse melanoma-related traits. In parallel, MITF alternative transcripts were characterized and their expression was investigated in different porcine tissues. The obtained results showed a complex transcriptional regulation concordant with the one present in other mammals. Notably, the ratio between MITF+ and MITF- isoforms in melanoma samples followed the same pattern as in human tumors, which highlights the adequacy of the MeLiM pig as a model for human melanoma. In conclusion, although MITF does not seem to be the causal gene of the QTL initially observed, we do not exclude a prominent role of its transcription and function in the outbreak and evolution of the tumors observed in pigs.


Subject(s)
Genetic Predisposition to Disease , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , Skin Neoplasms/genetics , Animals , Female , Gene Expression Regulation , Genetic Linkage , Male , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Molecular Sequence Data , Polymorphism, Single Nucleotide , Quantitative Trait Loci , RNA, Messenger , Skin Neoplasms/metabolism , Sus scrofa
2.
Mol Cancer ; 7: 34, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-18442364

ABSTRACT

BACKGROUND: Metastatic melanoma is a severe disease. Few experimental animal models of metastatic melanoma exist. MeLiM minipigs exhibit spontaneous melanoma. Cutaneous and metastatic lesions are histologically similar to human's. However, most of them eventually spontaneously regress. Our purpose was to investigate whether the MeLiM model could reveal markers of malignancy in human melanocytic proliferations. RESULTS: We compared the serial analysis of gene expression (SAGE) between normal pig skin melanocytes and melanoma cells from an early pulmonary metastasis of MeLiM minipigs. Tag identification revealed 55 regulated genes, including GNB2L1 which was found upregulated in the melanoma library. In situ hybridisation confirmed GNB2L1 overexpression in MeLiM melanocytic lesions. GNB2L1 encodes the adaptor protein RACK1, recently shown to influence melanoma cell lines tumorigenicity. We studied the expression of RACK1 by immunofluorescence and confocal microscopy in tissues specimens of normal skin, in cutaneous and metastatic melanoma developped in MeLiM minipigs and in human patients. In pig and human samples, the results were similar. RACK1 protein was not detected in normal epidermal melanocytes. By contrast, RACK1 signal was highly increased in the cytoplasm of all melanocytic cells of superficial spreading melanoma, recurrent dermal lesions and metastatic melanoma. RACK1 partially colocalised with activated PKCalphabeta. In pig metastases, additional nuclear RACK1 did not associate to BDNF expression. In human nevi, the RACK1 signal was low. CONCLUSION: RACK1 overexpression detected in situ in human melanoma specimens characterized cutaneous and metastatic melanoma raising the possibility that RACK1 can be a potential marker of malignancy in human melanoma. The MeLiM strain provides a relevant model for exploring mechanisms of melanocytic malignant transformation in humans. This study may contribute to a better understanding of melanoma pathophysiology and to progress in diagnosis.


Subject(s)
Cell Proliferation , GTP-Binding Proteins/genetics , Melanoma/pathology , Neoplasm Proteins/genetics , Receptors, Cell Surface/genetics , Skin Neoplasms/pathology , Transcription, Genetic , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Disease Models, Animal , Enzyme Activation , Female , GTP-Binding Proteins/metabolism , Humans , In Situ Hybridization , Male , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/genetics , Melanoma/metabolism , Microscopy, Confocal , Neoplasm Proteins/metabolism , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors for Activated C Kinase , Receptors, Cell Surface/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Swine , Swine, Miniature
3.
Int J Cancer ; 122(5): 1019-29, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-17957794

ABSTRACT

Tumors are complex tissues composed of neoplastic cells, soluble and insoluble matrix components and stromal cells. Here we report that in melanoma, turn-over of type I collagen (Col(I)), the predominant matrix protein in dermal stroma affects melanoma progression. Fibroblasts juxtaposed to melanoma cell nests within the papillary dermis display high levels of Col(I) mRNA expression. These nests are enveloped by collagen fibers. In contrast, melanoma-associated fibroblasts within the reticular dermis express Col(I) mRNA at a level that is comparable to its expression in uninvolved dermis and reduced amount of collagen protein can be observed. To determine the significance of Col(I) expression in melanoma, we pharmacologically inhibited its transcription in a porcine cutaneous melanoma model by oral administration of halofuginone. When administered before melanoma development, it reduced melanoma incidence and diminished the transition from microinvasive toward deeply invasive growth by limiting the development of a tumor vasculature. Whereas invasive melanoma growth has been correlated with increased blood vessel density previously, our data for the first time demonstrate that the proangiogenic effect of Col(I) expression by fibroblasts and vascular cells precedes the development of invasive melanomas in a de novo tumor model.


Subject(s)
Collagen Type I/metabolism , Melanoma/metabolism , Neoplasm Invasiveness , Neovascularization, Pathologic/metabolism , Skin Neoplasms/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Collagen Type I/drug effects , Humans , Immunohistochemistry , In Situ Hybridization , Melanoma/blood supply , Melanoma/pathology , Neoplasm Invasiveness/physiopathology , Piperidines/pharmacology , Quinazolinones/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Swine , Swine, Miniature
4.
Int J Cancer ; 120(2): 303-20, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17066441

ABSTRACT

Human cutaneous melanoma is a complex trait inherited in about 10% of cases. Although 2 high-risk genes, CDKN2A and CDK4, and 1 low risk gene, MC1R, have been identified, susceptibility genes remain to be discovered. Here, we attempted to determine new genomic regions linked to melanoma using the pig MeLiM strain, which develops hereditary cutaneous melanomas. We applied quantitative trait loci (QTL) mapping method to a significant genome-wide scan performed on 331 backcross pigs derived from this strain. QTLs were detected at chromosome-wide level for a melanoma synthetic trait corresponding to the development of melanoma. The peak positions on Sus scrofa chromosomes (SSC) were at 49.4 and 88.0 cM (SSC1), 56.0 cM (SSC13), 86.5 cM (SSC15) and 39.8 cM (SSC17), and, on SSC2, at 16.9 cM, in families derived from F1 males only (p < 0.05, except for SSC13, p < 0.01). Analysis of 7 precise specific traits revealed highly significant QTLs on SSC10 (ulceration), on SSC12 (presence of melanoma at birth), on SSC13 (lesion type), and on SSC16 and SSC17 (number of aggressive melanomas) at the respective positions 42.0, 95.6, 81.0, 45.3 and 44.8 cM (p < 0.001 and p < 0.05 respectively at the chromosome- and genome-wide levels). We also showed that MeLiM MC1R*2 allele, which determines black coat colour in pigs, predisposes significantly to melanoma. Interactions were observed between MC1R and markers located on SSC1 (p < 0.05). Taken together, these results indicate that MeLiM swine is a model for human multigenic diseases. Comparative mapping revealed human regions of interest to search for new melanoma susceptibility candidates.


Subject(s)
Disease Models, Animal , Melanoma/genetics , Quantitative Trait Loci , Skin Neoplasms/genetics , Swine, Miniature/genetics , Swine/genetics , Alleles , Animals , Chromosome Mapping , Female , Genetic Predisposition to Disease , Humans , Male , Phylogeny , Receptor, Melanocortin, Type 1/genetics , Swine/classification , Swine, Miniature/classification
5.
Int J Cancer ; 110(2): 232-8, 2004 Jun 10.
Article in English | MEDLINE | ID: mdl-15069687

ABSTRACT

Genetic alterations implicated in malignant melanoma are still poorly understood. Malignant melanomas present highly variable histologic and cytologic patterns. The aim of the present study is to define genomic imbalances associated with the development of 2 histologic types of swine hereditary cutaneous melanoma. We have investigated 11 swine tumors by comparative genomic hybridization (CGH), 4 superficial spreading melanomas (SSMs) and 7 nodular melanomas (NMs). Following laser capture microdissection and degenerate oligonucleotide primed-polymerase chain reaction, we were able to isolate and then amplify DNA from the 2 histologic subtypes. Consensus regions of chromosome gains were identified on both histologic subtypes, on swine chromosomes 3p13-p17 (75% of the SSMs and 71% of the NMs), 12q (100% of the SSMs and 57% of the NMs) and 14q11-q21 (75% of the SSMs and 42% of NMs). Chromosomal loss was restricted to NM lesions and the swine 13q36-49 region was lost in 100% of the NMs. Interphase fluorescence in situ hybridization with a probe mapping to the 13q41-q42 region indicates loss of the corresponding region on NM lesions. Taking into account this CGH analysis and the comparative genomic data between swine and human genomes, we suggest that a role for the human chromosomes 3p11-qter and chromosome 21 losses should be investigated in human nodular melanoma progression.


Subject(s)
Chromosome Deletion , Melanoma/genetics , Skin Neoplasms/genetics , Animals , Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 3 , Female , Humans , Male , Melanoma/pathology , Nucleic Acid Hybridization , Skin Neoplasms/pathology , Swine
6.
Int J Cancer ; 110(1): 39-50, 2004 May 20.
Article in English | MEDLINE | ID: mdl-15054867

ABSTRACT

In human familial melanoma, 3 risk susceptibility genes are already known, CDKN2A, CDK4 and MC1R. However, various observations suggest that other melanoma susceptibility genes have not yet been identified. To search for new susceptibility loci, we used the MeLiM swine as an animal model of hereditary melanoma to perform a genome scan for linkage to melanoma. Founders of the affected MeLiM stock were crossed with each other and with healthy Duroc pigs, generating MeLiM, F1 and backcross families. As we had previously excluded the MeLiM CDKN2A gene, we paid special attention to CDK4 and MC1R, as well as to other candidates such as BRAF and the SLA complex, mapping them on the swine radiation hybrid map and/or isolating close microsatellite markers to introduce them into the genome scan. The results revealed, first, that swine melanoma was inherited as an autosomal dominant trait with incomplete penetrance, preferably in black animals. Second, 4 chromosomal regions potentially involved in melanoma susceptibility were identified on Sus Scrofa chromosomes (SSC) 1, 2, 7 and 8, respectively, in intervals 44-103, 1.9-18, 59-73 and 47-62 cM. A fifth region close to MC1R was revealed on SSC 6 by analyzing an individual marker located at position 7.5 cM. Lastly, CDK4 and BRAF were unlikely to be melanoma susceptibility genes in the MeLiM swine model. The 3 regions on SSC 1, 6 and 7, respectively, have counterparts on human chromosomes (HSA) 9p, 16q and 6p, harboring melanoma candidate loci. The 2 others, on SSC 2 and 8, have counterparts on HSA 11 and 4, which might therefore be of interest for human studies.


Subject(s)
Chromosome Mapping , Cyclin-Dependent Kinases/genetics , Disease Models, Animal , Genetic Predisposition to Disease , Melanoma/genetics , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins , Receptor, Melanocortin, Type 1/genetics , Animals , Cyclin-Dependent Kinase 4 , Female , Genome , Genotype , Male , Microsatellite Repeats , Proto-Oncogene Proteins B-raf , Swine
7.
Pigment Cell Res ; 17(1): 24-35, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14717842

ABSTRACT

Spontaneous animal tumors appear to be highly suitable models to study human oncology and cancer therapy. The aim of this study was to characterize the clinical and histological features of hereditary melanocytic lesions found in the French herd of melanoblastoma-bearing Libechov minipigs (MeLiM) and their Duroc crossbreeds. Clinically, we discriminated between three types of melanocytic skin lesions, which offer a lesion continuum from lentigo to metastatic melanomas. More than 70% of these lesions appear on piglets before they are 3 months old and preferentially on homogeneous black coat piglets. The incidence of melanoma reaches 50% in MeLiM. Most of the highly invasive melanomas regressed spontaneously in the first year of the piglet's life and the regression was followed by hair, skin and iris depigmentation. A histopathological study was conducted according to the human melanoma classification. Except for lentigo maligna, we observed the three main types of human melanoma in swine [superficial spreading melanoma (SSM), nodular or unclassified melanoma] with an excess of SSM (59-67%). The histological events leading to total spontaneous regression are chronologically described. The genetic predisposition, the high incidence of melanoma, the clinical and histopathological features similar to the human disease and the high rate of spontaneous regression offer an opportunity to use this model for studying genetic events controlling melanoma development and regression and the biological mechanisms involved in oncogenesis and anti-cancerous self-defense.


Subject(s)
Disease Models, Animal , Hutchinson's Melanotic Freckle/pathology , Melanoma/pathology , Skin Neoplasms/pathology , Animals , Female , Hutchinson's Melanotic Freckle/genetics , Male , Melanoma/genetics , Neoplasm Regression, Spontaneous/pathology , Skin Neoplasms/genetics , Swine , Swine, Miniature
8.
Eur J Nucl Med Mol Imaging ; 30(6): 826-34, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12682788

ABSTRACT

The aim of this study was to evaluate the Melanoblastoma-bearing Libechov Minipigs (MeLiM) as an animal model of melanoma for in vivo imaging. Serial whole-body 2-deoxy-2-[(18)F]fluoro- d-glucose positron emission tomography (FDG PET) scans were conducted on five MeLiM. In order to explore different clinical stages of the tumoural lesions, each animal was scanned two to four times, at intervals of 30-155 days. PET images were analysed by a semiquantitative method based on the tumour to muscle metabolic ratio. Histology was performed on biopsies taken between or after the scans and the histological grading of the tumours was compared with the FDG uptake. The overall sensitivity of FDG PET for the detection of cutaneous melanoma was 75%; 62.5% of involved lymph nodes were positive. Sensitivity was better for tumours with vertical growth than for flat lesions. FDG PET did not detect tumours with epidermal involvement only, nor did it detect small metastatic foci. The metabolic ratio was correlated with the evolution of the melanoma. FDG PET is effective in the staging of cutaneous melanoma and the follow-up of tumoural extension and regression in Melanoblastoma-bearing Libechov Minipigs. The results obtained in this animal model correlate well with those described in human melanoma. Accordingly, this model may be useful in testing new tracers specific for melanoma and in helping to detect molecules expressed early during tumoural regression.


Subject(s)
Fluorodeoxyglucose F18 , Melanoma/veterinary , Radiopharmaceuticals , Skin Neoplasms/veterinary , Swine Diseases/diagnostic imaging , Tomography, Emission-Computed/methods , Animals , Disease Models, Animal , Fluorodeoxyglucose F18/pharmacokinetics , Melanoma/diagnostic imaging , Melanoma/pathology , Neoplasm Staging , Radiopharmaceuticals/pharmacokinetics , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/pathology , Swine , Swine Diseases/pathology , Swine, Miniature
9.
Int J Cancer ; 103(5): 631-5, 2003 Feb 20.
Article in English | MEDLINE | ID: mdl-12494470

ABSTRACT

Some herds of miniature swine are genetically predisposed to cutaneous melanoma. To test if swine melanoma susceptibility could be linked to the CDKN2A gene, which is involved in a proportion of 9p21-linked human familial melanoma, we performed a genetic analysis of miniature pigs of the MeLiM strain. F(1) and backcross animals were generated by crossing 1 MeLiM boar with healthy Duroc sows. We isolated the swine CDKN2A gene and characterized a linked informative microsatellite marker, the S0644 marker. Using this marker and 2 flanking markers, we analyzed the segregation of the CDKN2A gene in a 3-generation pedigree. Allelic association, linkage analysis and haplotype analysis of these data led to exclusion of the CDKN2A gene as a candidate for melanoma susceptibility. Nonetheless, this analysis suggests an association with the swine 1q25 chromosomal region, which is homologous to the human 9p21 region.


Subject(s)
Genes, p16 , Genetic Predisposition to Disease , Melanoma/veterinary , Polymorphism, Genetic , Skin Neoplasms/veterinary , Swine Diseases/genetics , Alleles , Animals , Chromosome Segregation , DNA Mutational Analysis , DNA Primers/chemistry , DNA, Neoplasm/analysis , Female , Genetic Linkage , Genetic Markers , Haplotypes , Humans , Male , Melanoma/genetics , Melanoma/pathology , Microsatellite Repeats , Pedigree , Polymerase Chain Reaction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Swine, Miniature
SELECTION OF CITATIONS
SEARCH DETAIL
...