Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Cell Rep ; 42(12): 113586, 2023 12 26.
Article En | MEDLINE | ID: mdl-38113139

Melanoma is the deadliest form of skin cancer due to its propensity to metastasize. It arises from melanocytes, which are attached to keratinocytes within the basal epidermis. Here, we hypothesize that, in addition to melanocyte-intrinsic modifications, dysregulation of keratinocyte functions could initiate early-stage melanoma cell invasion. We identified the lysolipid sphingosine 1-phosphate (S1P) as a tumor paracrine signal from melanoma cells that modifies the keratinocyte transcriptome and reduces their adhesive properties, leading to tumor invasion. Mechanistically, tumor cell-derived S1P reduced E-cadherin expression in keratinocytes via S1P receptor dependent Snail and Slug activation. All of these effects were blocked by S1P2/3 antagonists. Importantly, we showed that epidermal E-cadherin expression was inversely correlated with the expression of the S1P-producing enzyme in neighboring tumors and the Breslow thickness in patients with early-stage melanoma. These findings support the notion that E-cadherin loss in the epidermis initiates the metastatic cascade in melanoma.


Melanoma , Humans , Melanoma/pathology , Sphingolipids/metabolism , Paracrine Communication , Keratinocytes/metabolism , Cadherins/metabolism , Sphingosine/metabolism , Lysophospholipids/metabolism
2.
Clin Epigenetics ; 14(1): 156, 2022 11 28.
Article En | MEDLINE | ID: mdl-36443814

The protocadherin proteins are cell adhesion molecules at the crossroad of signaling pathways playing a major role in neuronal development. It is now understood that their role as signaling hubs is not only important for the normal physiology of cells but also for the regulation of hallmarks of cancerogenesis. Importantly, protocadherins form a cluster of genes that are regulated by DNA methylation. We have identified for the first time that PCDHB15 gene is DNA-hypermethylated on its unique exon in the metastatic melanoma-derived cell lines and patients' metastases compared to primary tumors. This DNA hypermethylation silences the gene, and treatment with the DNA demethylating agent 5-aza-2'-deoxycytidine reinduces its expression. We explored the role of PCDHB15 in melanoma aggressiveness and showed that overexpression impairs invasiveness and aggregation of metastatic melanoma cells in vitro and formation of lung metastasis in vivo. These findings highlight important modifications of the methylation of the PCDHß genes in melanoma and support a functional role of PCDHB15 silencing in melanoma aggressiveness.


Lung Neoplasms , Melanoma , Humans , DNA Methylation , Melanoma/genetics , Signal Transduction , Exons , Lung Neoplasms/genetics
3.
Elife ; 112022 09 20.
Article En | MEDLINE | ID: mdl-36125262

Aberrant DNA methylation is a well-known feature of tumours and has been associated with metastatic melanoma. However, since melanoma cells are highly heterogeneous, it has been challenging to use affected genes to predict tumour aggressiveness, metastatic evolution, and patients' outcomes. We hypothesized that common aggressive hypermethylation signatures should emerge early in tumorigenesis and should be shared in aggressive cells, independent of the physiological context under which this trait arises. We compared paired melanoma cell lines with the following properties: (i) each pair comprises one aggressive counterpart and its parental cell line and (ii) the aggressive cell lines were each obtained from different host and their environment (human, rat, and mouse), though starting from the same parent cell line. Next, we developed a multi-step genomic pipeline that combines the DNA methylome profile with a chromosome cluster-oriented analysis. A total of 229 differentially hypermethylated genes was commonly found in the aggressive cell lines. Genome localization analysis revealed hypermethylation peaks and clusters, identifying eight hypermethylated gene promoters for validation in tissues from melanoma patients. Five Cytosine-phosphate-Guanine (CpGs) identified in primary melanoma tissues were transformed into a DNA methylation score that can predict survival (log-rank test, p=0.0008). This strategy is potentially universally applicable to other diseases involving DNA methylation alterations.


Melanoma , Skin Neoplasms , Animals , Chromosomes , CpG Islands , Cytosine , DNA Methylation , Epigenesis, Genetic , Epigenome , Gene Expression Regulation, Neoplastic , Guanine , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Phosphates , Rats , Skin Neoplasms/genetics , Melanoma, Cutaneous Malignant
4.
Cancer Immunol Res ; 9(5): 568-582, 2021 05.
Article En | MEDLINE | ID: mdl-33727246

Dysregulation of lipid metabolism affects the behavior of cancer cells, but how this happens is not completely understood. Neutral sphingomyelinase 2 (nSMase2), encoded by SMPD3, catalyzes the breakdown of sphingomyelin to produce the anti-oncometabolite ceramide. We found that this enzyme was often downregulated in human metastatic melanoma, likely contributing to immune escape. Overexpression of nSMase2 in mouse melanoma reduced tumor growth in syngeneic wild-type but not CD8-deficient mice. In wild-type mice, nSMase2-overexpressing tumors showed accumulation of both ceramide and CD8+ tumor-infiltrating lymphocytes, and this was associated with increased level of transcripts encoding IFNγ and CXCL9. Overexpressing the catalytically inactive nSMase2 failed to alter tumor growth, indicating that the deleterious effect nSMase2 has on melanoma growth depends on its enzymatic activity. In vitro, small extracellular vesicles from melanoma cells overexpressing wild-type nSMase2 augmented the expression of IL12, CXCL9, and CCL19 by bone marrow-derived dendritic cells, suggesting that melanoma nSMase2 triggers T helper 1 (Th1) polarization in the earliest stages of the immune response. Most importantly, overexpression of wild-type nSMase2 increased anti-PD-1 efficacy in murine models of melanoma and breast cancer, and this was associated with an enhanced Th1 response. Therefore, increasing SMPD3 expression in melanoma may serve as an original therapeutic strategy to potentiate Th1 polarization and CD8+ T-cell-dependent immune responses and overcome resistance to anti-PD-1.


CD8-Positive T-Lymphocytes/immunology , Melanoma/immunology , Melanoma/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Immunity , Immunotherapy , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/genetics , Th1 Cells/immunology
5.
Cancers (Basel) ; 12(11)2020 Oct 27.
Article En | MEDLINE | ID: mdl-33121001

Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid ß-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is (i) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, (ii) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and (iii) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.

6.
Front Pharmacol ; 10: 443, 2019.
Article En | MEDLINE | ID: mdl-31114500

Sphingolipid (SL) metabolism alterations have been frequently reported in cancer including in melanoma, a bad-prognosis skin cancer. In normal cells, de novo synthesized ceramide is mainly converted to sphingomyelin (SM), the most abundant SL, by sphingomyelin synthase 1 (SMS1) and, albeit to a lesser extent, SMS2, encoded by the SGMS1 and SGMS2 genes, respectively. Alternatively, ceramide can be converted to glucosylceramide (GlcCer) by the GlcCer synthase (GCS), encoded by the UGCG gene. Herein, we provide evidence for the first time that SMS1 is frequently downregulated in various solid cancers, more particularly in melanoma. Accordingly, various human melanoma cells displayed a SL metabolism signature associated with (i) a robust and a low expression of UGCG and SGMS1/2, respectively, (ii) higher in situ enzyme activity of GCS than SMS, and (iii) higher intracellular levels of GlcCer than SM. SMS1 was expressed at low levels in most of the human melanoma biopsies. In addition, several mutations and increased CpG island methylation in the SGMS1 gene were identified that likely affect SMS1 expression. Finally, low SMS1 expression was associated with a worse prognosis in metastatic melanoma patients. Collectively, our study indicates that SMS1 downregulation in melanoma enhances GlcCer synthesis, triggering an imbalance in the SM/GlcCer homeostasis, which likely contributes to melanoma progression. Evaluating SMS1 expression level in tumor samples might serve as a biomarker to predict clinical outcome in advanced melanoma patients.

7.
Clin Epigenetics ; 11(1): 9, 2019 01 16.
Article En | MEDLINE | ID: mdl-30651148

BACKGROUND: Efficient treatments against metastatic melanoma dissemination are still lacking. Here, we report that low-cytotoxic concentrations of 5-aza-2'-deoxycytidine, a DNA demethylating agent, prevent in vitro 3D invasiveness of metastatic melanoma cells and reduce lung metastasis formation in vivo. RESULTS: We unravelled that this beneficial effect is in part due to MIR-199A2 re-expression by promoter demethylation. Alone, this miR showed an anti-invasive and anti-metastatic effect. Throughout integration of micro-RNA target prediction databases with transcriptomic analysis after 5-aza-2'-deoxycytidine treatments, we found that miR-199a-3p downregulates set of genes significantly involved in invasion/migration processes. In addition, analysis of data from melanoma patients showed a stage- and tissue type-dependent modulation of MIR-199A2 expression by DNA methylation. CONCLUSIONS: Thus, our data suggest that epigenetic- and/or miR-based therapeutic strategies can be relevant to limit metastatic dissemination of melanoma.


DNA Methylation/drug effects , Decitabine/pharmacology , Lung Neoplasms/secondary , Melanoma/genetics , MicroRNAs/genetics , Spheroids, Cellular/cytology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Melanoma/drug therapy , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Promoter Regions, Genetic , Sequence Analysis, RNA , Spheroids, Cellular/drug effects , Up-Regulation
8.
Oncotarget ; 6(17): 15265-82, 2015 Jun 20.
Article En | MEDLINE | ID: mdl-25948775

5-azacytidine and 5-aza-2'-deoxycytidine are clinically used to treat patients with blood neoplasia. Their antileukemic property is mediated by the trapping and the subsequent degradation of a family of proteins, the DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B) leading to DNA demethylation, tumor suppressor gene re-expression and DNA damage. Here we studied the respective role of each DNMT in the human leukemia KG1 cell line using a RNA interference approach. In addition we addressed the role of DNA damage formation in DNA demethylation by 5-aza-2'-deoxycytidine. Our data show that DNMT1 is the main DNMT involved in DNA methylation maintenance in KG1 cells and in mediating DNA damage formation upon exposure to 5-aza-2'-deoxycytidine. Moreover, KG1 cells express the DNMT1 protein at a level above the one required to ensure DNA methylation maintenance, and we identified a threshold for DNMT1 depletion that needs to be exceeded to achieve DNA demethylation. Most interestingly, by combining DNMT1 siRNA and treatment with low dose of 5-aza-2'-deoxycytidine, it is possible to uncouple DNA damage formation from DNA demethylation. This work strongly suggests that a direct pharmacological inhibition of DNMT1, unlike the use of 5-aza-2'-deoxycytidine, should lead to tumor suppressor gene hypomethylation and re-expression without inducing major DNA damage in leukemia.


Azacitidine/analogs & derivatives , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Leukemia/drug therapy , Azacitidine/pharmacology , Cell Line, Tumor , Cell Proliferation/genetics , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Damage/genetics , DNA Methylation/drug effects , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Decitabine , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Histones/metabolism , Humans , Nuclear Proteins/genetics , Phosphorylation , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , DNA Methyltransferase 3B
9.
Epigenetics ; 10(1): 82-91, 2015.
Article En | MEDLINE | ID: mdl-25531272

DNA methylation is a chemical modification of DNA involved in the regulation of gene expression by controlling the access to the DNA sequence. It is the most stable epigenetic mark and is widely studied for its role in major biological processes. Aberrant DNA methylation is observed in various pathologies, such as cancer. Therefore, there is a great interest in analyzing subtle changes in DNA methylation induced by biological processes or upon drug treatments. Here, we developed an improved methodology based on flow cytometry to measure variations of DNA methylation level in melanoma and leukemia cells. The accuracy of DNA methylation quantification was validated with LC-ESI mass spectrometry analysis. The new protocol was used to detect small variations of cytosine methylation occurring in individual cells during their cell cycle and those induced by the demethylating agent 5-aza-2'-deoxycytidine (5AzadC). Kinetic experiments confirmed that inheritance of DNA methylation occurs efficiently in S phase and revealed a short delay between DNA replication and completion of cytosine methylation. In addition, this study suggests that the uncoupling of 5AzadC effects on DNA demethylation and cell proliferation might be related to the duration of the DNA replication phase.


Cell Cycle , DNA Methylation , DNA, Neoplasm/genetics , Flow Cytometry/methods , Cell Line, Tumor , Humans
10.
PLoS One ; 7(5): e36762, 2012.
Article En | MEDLINE | ID: mdl-22675422

Metastatic melanoma is the most aggressive skin cancer. Recently, phenotypically distinct subpopulations of tumor cells were identified. Among them, ABCB5-expressing cells were proposed to display an enhanced tumorigenicity with stem cell-like properties. In addition, ABCB5(+) cells are thought to participate to chemoresistance through a potential efflux function of ABCB5. Nevertheless, the fate of these cells upon drugs that are used in melanoma chemotherapy remains to be clarified. Here we explored the effect of anti-melanoma treatments on the ABCB5-expressing cells. Using a melanoma xenograft model (WM266-4), we observed in vivo that ABCB5-expressing cells are enriched after a temozolomide treatment that induces a significant tumor regression. These results were further confirmed in a preliminary study conducted on clinical samples from patients that received dacarbazine. In vitro, we showed that ABCB5-expressing cells selectively survive when exposed to dacarbazine, the reference treatment of metastatic melanoma, but also to vemurafenib, a new inhibitor of the mutated kinase V600E BRAF and other various chemotherapeutic drugs. Our results show that anti-melanoma chemotherapy might participate to the chemoresistance acquisition by selecting tumor cell subpopulations expressing ABCB5. This is of particular importance in understanding the relapses observed after anti-melanoma treatments and reinforces the interest of ABCB5 and ABCB5-expressing cells as potential therapeutic targets in melanoma.


ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Melanoma/drug therapy , Melanoma/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival/drug effects , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm, Residual , Xenograft Model Antitumor Assays
11.
Cancer Immun ; 9: 10, 2009 Nov 02.
Article En | MEDLINE | ID: mdl-19877577

Cell surface expression of MHC class I molecules by tumor cells is determinant in the interplay between tumor cells and the immune system. Nevertheless, the mechanisms which regulate MHCI expression on tumor cells are not clear. We previously showed that immune innate cells from the spleen can regulate MHCI expression on MHCI(low) tumor cells. Here, using the murine model of B16 melanoma, we demonstrate that the MHCI status of tumor cells in vivo is regulated by the microenvironment. In subcutaneous grafts, induction of MHCI molecules on tumor cells is concomitant to the recruitment of lymphocytes and relies on an IFNgamma-mediated mechanism. gammadelta T and NK cells are essential to this regulation. A small proportion of tumor-infiltrating NK cells and gammadelta T cells were found to produce IFNgamma, suggesting a possible direct participation to the MHCI increase on the tumor cells upon tumor cell recognition. Depletion of gammadelta T cells increases the tumor growth rate, confirming their anti-tumoral role in our model. Taken together, our results demonstrate that in vivo, NK and gammadelta T cells play a dual role during the early growth of MHCI(low) tumor cells. In addition to controlling the growth of tumor cells, they contribute to modifying the immunogenic profile of residual tumor cells.


Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Melanoma/immunology , Melanoma/pathology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Progression , Histocompatibility Antigens Class I/metabolism , Interferon-gamma/immunology , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Receptors, Antigen, T-Cell, gamma-delta/immunology , Up-Regulation
12.
Cancer Immun ; 8: 14, 2008 Sep 11.
Article En | MEDLINE | ID: mdl-18783178

Tumor cells and the immune system play a lethal "pas de deux" during tumor development. However, it is not clear which role the innate immune system plays in these interactions. We studied the interaction of normal spleen cells (NSCs) with tumor cells expressing low levels of MHCI on the cell surface. This interaction induces increased MHCI expression on the MHCI(low) tumor cells by a cell-cell contact-dependent, IFN-gamma-mediated mechanism. The effector cells responsible for the increased IFN-gamma production were identified as CD4+ CD1d-independent NKT cells, NK1.1+ NK cells and CD4+ CD11c+ DCs. The possible three cell collaboration is not activated by MHCI(high) tumor cells or normal fibroblasts. Kinetic experiments showed that the increase in IFN-gamma production induced by MHCI(low) tumor cells happens in two consecutive waves, an early peak around 12 hours, followed by a second more important peak around day 2-3. Thus, we propose that CD4+ CD1d-independent NKT cells are activated by the MHCI(low) tumor cells, they release IFN-gamma stimulating DCs to produce IL-12, which in turn activates NK cells to produce large amounts of IFN-gamma. The recognition mechanism used by the CD4+ CD1d-independent non-classical NKT cells is unknown. Monoclonal antibody (mAb) blocking experiments using antibodies against either activating or inhibitory receptors or co-receptors on NKT/NK cells gave no conclusive results. Moreover, NSCs from either normal or MHCII(-/-) mice augmented MHCI expression on MHCI(low) tumors, excluding a significant role of CD4-MHCII interactions in the system. Hence the initial recognition mechanism in this system still awaits further experimentation.


Histocompatibility Antigens Class I/immunology , Immunity, Innate/immunology , Neoplasms/immunology , Animals , Antigens, CD1/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Communication , Cell Line, Tumor , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/immunology
13.
Eur J Immunol ; 35(8): 2284-94, 2005 Aug.
Article En | MEDLINE | ID: mdl-16021601

We have investigated the density of peptides required to elicit different biological responses in cytotoxic T lymphocytes (CTL), including trogocytosis (i.e., the phenomenon whereby the lymphocytes actively capture fragments of plasma membrane from those cells with which they establish an immune synapse). We have used two separate mouse models of CTL recognising defined peptides presented by MHC class I molecules. In both systems, triggering of cytotoxicity and capture of membrane components reached saturation with low densities of ligand. On the other hand, down-modulation of cell-surface levels of TCR, induction of IFN-gamma production and detection of peptide captured required much higher ligand densities. Interestingly, fratricide (i.e., killing between CTL sharing the same specificity), a mechanism proposed to account for CTL exhaustion, was detected only at antigen concentrations still well above that second threshold leading to full blown activation. Taken together, our results show that the different thresholds that govern the elicitation of different CTL functions correlate with different proportions of antigen among the target cell components being captured via trogocytosis.


Antigens/biosynthesis , Lymphocyte Activation/immunology , Plasma Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Biotin/metabolism , Cell Line, Tumor , Cells, Cultured , Cytokines/metabolism , Histocompatibility Antigens/immunology , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Peptide Fragments/immunology , Plasma Cells/metabolism , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/metabolism
...