Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(14): 3806-3821, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34099492

ABSTRACT

Overcoming acquired drug resistance is a primary challenge in cancer treatment. Notably, more than 50% of patients with BRAFV600E cutaneous metastatic melanoma (CMM) eventually develop resistance to BRAF inhibitors. Resistant cells undergo metabolic reprogramming that profoundly influences therapeutic response and promotes tumor progression. Uncovering metabolic vulnerabilities could help suppress CMM tumor growth and overcome drug resistance. Here we identified a drug, HA344, that concomitantly targets two distinct metabolic hubs in cancer cells. HA344 inhibited the final and rate-limiting step of glycolysis through its covalent binding to the pyruvate kinase M2 (PKM2) enzyme, and it concurrently blocked the activity of inosine monophosphate dehydrogenase, the rate-limiting enzyme of de novo guanylate synthesis. As a consequence, HA344 efficiently targeted vemurafenib-sensitive and vemurafenib-resistant CMM cells and impaired CMM xenograft tumor growth in mice. In addition, HA344 acted synergistically with BRAF inhibitors on CMM cell lines in vitro. Thus, the mechanism of action of HA344 provides potential therapeutic avenues for patients with CMM and a broad range of different cancers. SIGNIFICANCE: Glycolytic and purine synthesis pathways are often deregulated in therapy-resistant tumors and can be targeted by the covalent inhibitor described in this study, suggesting its broad application for overcoming resistance in cancer.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Carrier Proteins/antagonists & inhibitors , IMP Dehydrogenase/antagonists & inhibitors , Melanoma/drug therapy , Membrane Proteins/antagonists & inhibitors , Ribonucleotides/pharmacology , Skin Neoplasms/drug therapy , Aged , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , Melanoma/enzymology , Melanoma/pathology , Mice , Mice, Nude , Random Allocation , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Thyroid Hormones , Xenograft Model Antitumor Assays , Thyroid Hormone-Binding Proteins , Melanoma, Cutaneous Malignant
2.
Antioxidants (Basel) ; 10(5)2021 May 04.
Article in English | MEDLINE | ID: mdl-34064498

ABSTRACT

Specific inhibition of NADPH oxidases (NOX) and NO-synthases (NOS), two enzymes associated with redox stress in tumor cells, has aroused great pharmacological interest. Here, we show how these enzymes distinguish between isomeric 2'- and 3'-phosphate derivatives, a difference used to improve the specificity of inhibition by isolated 2'- and 3'-phosphate isomers of our NADPH analogue NS1. Both isomers become fluorescent upon binding to their target proteins as observed by in vitro assay and in vivo imaging. The 2'-phosphate isomer of NS1 exerted more pronounced effects on NOS and NOX-dependent physiological responses than the 3'-phosphate isomer did. Docking and molecular dynamics simulations explain this specificity at the level of the NADPH site of NOX and NOS, where conserved arginine residues distinguished between the 2'-phosphate over the 3'-phosphate group, in favor of the 2'-phosphate.

3.
Future Med Chem ; 13(14): 1157-1173, 2021 07.
Article in English | MEDLINE | ID: mdl-34096325

ABSTRACT

Background: In line with our recent discovery of an efficient anticancer thiazolebenzenesulfonamide framework HA15 (1) based on a remarkable endoplasmic reticulum stress inducement mode of action, we report herein a series of innovative constrained HA15 analogs, featuring four types of bicylic derivatives. Results: The structure-activity relationship analysis, using a cell line assay, led us to identify a novel version of HA15: a new benzothiazole derivative (10b) exhibiting important anti-melanoma effect against sensitive and resistant melanoma cells. Meanwhile, compound 10b induced a significant tumor growth inhibition in vivo with no apparent signs of toxicity. Conclusion: These results consistently open new directions to improve and develop more powerful anticancer therapeutics harboring this type of fused framework.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzothiazoles/chemistry , Melanoma/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Humans , Melanoma/pathology , Mice , Mice, Nude , Structure-Activity Relationship , Transplantation, Heterologous
4.
Cell Death Dis ; 12(1): 64, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431809

ABSTRACT

In the search of biguanide-derived molecules against melanoma, we have discovered and developed a series of bioactive products and identified the promising new compound CRO15. This molecule exerted anti-melanoma effects on cells lines and cells isolated from patients including the ones derived from tumors resistant to BRAF inhibitors. Moreover, CRO15 was able to decrease viability of cells lines from a broad range of cancer types. This compound acts by two distinct mechanisms. First by activating the AMPK pathway induced by a mitochondrial disorder. Second by inhibition of MELK kinase activity, which induces cell cycle arrest and activation of DNA damage repair pathways by p53 and REDD1 activation. All of these mechanisms activate autophagic and apoptotic processes resulting in melanoma cell death. The strong efficacy of CRO15 to reduce the growth of melanoma xenograft sensitive or resistant to BRAF inhibitors opens interesting perspective.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Melanoma/genetics , Protein Serine-Threonine Kinases/metabolism , Cell Death , Cell Proliferation , Humans , Melanoma/pathology , Signal Transduction
5.
Nat Commun ; 10(1): 2178, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097717

ABSTRACT

T-cells play a crucial role in progression of autoimmunity, including vitiligo, yet the initial steps triggering their activation and tissue damage remain unknown. Here we demonstrate increased presence of type-1 innate lymphoid cells (NK and ILC1)-producing interferon gamma (IFNγ) in the blood and in non-lesional skin of vitiligo patients. Melanocytes of vitiligo patients have strong basal expression of chemokine-receptor-3 (CXCR3) isoform B which is directly regulated by IFNγ. CXCR3B activation by CXCL10 at the surface of cultured human melanocytes induces their apoptosis. The remaining melanocytes, activated by the IFNγ production, express co-stimulatory markers which trigger T-cell proliferation and subsequent anti-melanocytic immunity. Inhibiting the CXCR3B activation prevents this apoptosis and the further activation of T cells. Our results emphasize the key role of CXCR3B in apoptosis of melanocytes and identify CXCR3B as a potential target to prevent and to treat vitiligo by acting at the early stages of melanocyte destruction.


Subject(s)
Autoimmunity , Melanocytes/immunology , Receptors, CXCR3/metabolism , T-Lymphocytes/immunology , Vitiligo/immunology , Adult , Aged , Apoptosis/immunology , Biopsy , Cells, Cultured , Chemokine CXCL10/metabolism , Female , Humans , Immunity, Innate , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation , Male , Melanocytes/metabolism , Middle Aged , Primary Cell Culture , Protein Isoforms/immunology , Protein Isoforms/metabolism , Receptors, CXCR3/immunology , Skin/cytology , Skin/pathology , T-Lymphocytes/metabolism , Vitiligo/blood , Vitiligo/pathology
6.
Cell Death Dis ; 9(5): 527, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29743521

ABSTRACT

Melanoma is one of the most lethal cancers when it reaches a metastatic stage. Despite advancements in targeted therapies (BRAF inhibitors) or immunotherapies (anti-CTLA-4 or anti-PD1), most patients with melanoma will need additional treatment. Thus, there is an urgent need to develop new therapeutical approaches to bypass resistance and achieve more prolonged responses. In this context, we were interested in E2F1, a transcription factor that plays a major role in the control of cell cycle under physiological and pathological conditions. Here we confirmed that E2F1 is highly expressed in melanoma cells. Inhibition of E2F1 activity further increased melanoma cell death and senescence, both in vitro and in vivo. Moreover, blocking E2F1 also induced death of melanoma cells resistant to BRAF inhibitors. In conclusion, our studies suggest that targeting the E2F1 signaling pathway may be therapeutically relevant for melanoma.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cell Death/drug effects , E2F1 Transcription Factor , Melanoma, Experimental , Signal Transduction/drug effects , Animals , Cell Line, Tumor , E2F1 Transcription Factor/antagonists & inhibitors , E2F1 Transcription Factor/metabolism , Female , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Nude , Neoplasm Metastasis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism
7.
Bioorg Med Chem Lett ; 27(10): 2192-2196, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28372910

ABSTRACT

We recently described a new family of bioactive molecules with interesting anti-cancer activities: the N-(4-(3-aminophenyl)thiazol-2-yl)acetamides. The lead compound of the series (1) displays significant anti-proliferative and cytotoxic activities against a panel of cancer cell lines, either sensitive or resistant to standard treatments. This molecule also shows a good pharmacological profile and high in vivo potency towards mice xenografts, without signs of toxicity on the animals. In the present article, we disclose the structure-activity relationships of this lead compound, which have provided clear information about the replacement of the acetamide function and the substitution pattern of the benzenesulfonamide ring. An improved high-yielding synthetic procedure towards these compounds has also been developed. Our drug design resulted in potency enhancement of 1, our new optimized lead compound being 19. These findings are of great interest to further improve this scaffold for the development of future clinical candidates.


Subject(s)
Antineoplastic Agents/chemistry , Sulfonamides/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Drug Design , Drug Resistance, Neoplasm/drug effects , Humans , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/toxicity , Benzenesulfonamides
9.
J Med Chem ; 59(18): 8276-92, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27575313

ABSTRACT

Cancer is the second cause of deaths worldwide and is forecasted to affect more that 22 million people in 2020. Despite dramatic improvement in its care over the last two decades, the treatment of resistant forms of cancer is still an unmet challenge. Thus, innovative and efficient treatments are still needed. In this context, we report herein the synthesis and the evaluation of a new class of bioactive molecules belonging to the N-(4-(3-aminophenyl(thiazol-2-yl)acetamide family. Structure-activity relationships could be driven and resulted in the discovery of lead compound 6b. The latter display high in vitro potency against both sensitive and resistant cancer cell lines on three models: melanoma, pancreatic cancer, and chronic myeloid leukemia (CML). 6b leads to cell death by concomitant induction of apoptosis and autophagy, shows good pharmacokinetic properties, and demonstrates a significant reduction of tumor growth in vivo on A375 xenograft model in mice.


Subject(s)
Acetamides/chemistry , Acetamides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Acetamides/pharmacokinetics , Acetamides/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Melanoma/drug therapy , Mice , Pancreatic Neoplasms/drug therapy , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Thiazoles/therapeutic use
10.
Cancer Cell ; 29(6): 805-819, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27238082

ABSTRACT

We have discovered and developed a series of molecules (thiazole benzenesulfonamides). HA15, the lead compound of this series, displayed anti-cancerous activity on all melanoma cells tested, including cells isolated from patients and cells that developed resistance to BRAF inhibitors. Our molecule displayed activity against other liquid and solid tumors. HA15 also exhibited strong efficacy in xenograft mouse models with melanoma cells either sensitive or resistant to BRAF inhibitors. Transcriptomic, proteomic, and biochemical studies identified the chaperone BiP/GRP78/HSPA5 as the specific target of HA15 and demonstrated that the interaction increases ER stress, leading to melanoma cell death by concomitant induction of autophagic and apoptotic mechanisms.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum Stress/drug effects , Melanoma/drug therapy , Sulfonamides/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Melanoma/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
11.
Cell Discov ; 1: 15030, 2015.
Article in English | MEDLINE | ID: mdl-27462428

ABSTRACT

Specific BRAFV600E inhibitors (BRAFi) are highly effective in the treatment of melanoma. However, acquired drug resistances invariably develop after the initial response. Therefore, the identification of new mechanisms of acquired resistance gives important clues towards the development of therapies that could elicit long lasting responses. Here we report that CD271 confers resistance to BRAFi in melanoma cells. The expression of CD271 is increased by BRAFi through a stimulation of tumor necrosis factor-alpha (TNFα) secretion that leads to NF-κB signaling pathway activation. CD271 is upregulated in a subset of BRAFi-resistant melanoma cells. The inhibition of TNFα/NF-κB pathway and CD271 silencing restore the BRAFi sensitivity of resistant melanoma cells. Finally, increase of CD271 expression is validated in BRAFi-resistant xenografts tumors and also in tumors from the patients who relapsed under BRAFi. In summary, these results reveal a novel TNFα/NF-κB/CD271 axis whose activation contributes to the acquisition of resistance to BRAFi and therefore may represent a novel therapeutic target to improve the efficacy of therapy in melanoma.

12.
J Invest Dermatol ; 134(10): 2589-2597, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24756109

ABSTRACT

Several reports have demonstrated the inhibitory effect of metformin, a widely used drug in the treatment of type 2 diabetes, on the proliferation of many cancers including melanoma. Recently, it has been shown that metformin is able to modulate the cAMP level in the liver. As cAMP has a crucial role in melanin synthesis and skin pigmentation, we investigated the effect of metformin on melanogenesis both in vitro and in vivo. We showed that metformin led to reduced melanin content in melanoma cells and in normal human melanocytes by decreasing cAMP accumulation and cAMP-responsive element-binding protein phosphorylation. This inhibitory effect is correlated with decreased expression of master genes of melanogenesis, microphthalmia-associated transcription factor, tyrosinase, dopachrome tautomerase, and tyrosinase-related protein 1. Furthermore, we demonstrated that the antimelanogenic effect of metformin is independent of the AMPK pathway. Interestingly, topical application of metformin induced tail whitening in mice. Finally, we confirmed the antimelanogenic effect of metformin on reconstituted human epidermis and on human skin biopsies. These data emphasize the depigmenting effect of metformin and suggest a clinical strategy for using metformin in the topical treatment of hyperpigmentation disorders.


Subject(s)
Hypoglycemic Agents/pharmacology , Melanins/metabolism , Melanocytes/drug effects , Melanocytes/metabolism , Metformin/pharmacology , Skin/drug effects , Skin/metabolism , Animals , Biopsy , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Microphthalmia-Associated Transcription Factor/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Pigmentation/drug effects , Time Factors
13.
Mol Cancer Ther ; 12(8): 1605-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23741061

ABSTRACT

Metformin was reported to inhibit the proliferation of many cancer cells, including melanoma cells. In this report, we investigated the effect of metformin on melanoma invasion and metastasis development. Using different in vitro approaches, we found that metformin inhibits cell invasion without affecting cell migration and independently of antiproliferation action. This inhibition is correlated with modulation of expression of proteins involved in epithelial-mesenchymal transition such as Slug, Snail, SPARC, fibronectin, and N-cadherin and with inhibition of MMP-2 and MMP-9 activation. Furthermore, our data indicate that this process is dependent on activation of AMPK and tumor suppressor protein p53. Finally, we showed that metformin inhibits melanoma metastasis development in mice using extravasation and metastasis models. The presented data reinforce the fact that metformin might be a good candidate for clinical trial in melanoma treatment.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Melanoma/metabolism , Melanoma/pathology , Metformin/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Melanoma/genetics , Metalloendopeptidases/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Metastasis
14.
Cancer Res ; 69(7): 2748-56, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19293188

ABSTRACT

Loss of tumor-suppressive pathways that control cellular senescence is a crucial step in malignant transformation. Spleen tyrosine kinase (Syk) is a cytoplasmic tyrosine kinase that has been recently implicated in tumor suppression of melanoma, a deadly skin cancer derived from pigment-producing melanocytes. However, the mechanism by which Syk suppresses melanoma growth remains unclear. Here, we report that reexpression of Syk in melanoma cells induces a p53-dependent expression of the cyclin-dependent kinase (cdk) inhibitor p21 and a senescence program. We first observed that Syk expression is lost in a subset of melanoma cell lines, primarily by DNA methylation-mediated gene silencing and restored after treatment with the demethylating agent 5-aza-2-deoxycytidine. We analyzed the significance of epigenetic inactivation of Syk and found that reintroduction of Syk in melanoma cells dramatically reduces clonogenic survival and three-dimensional tumor spheroid growth and invasion. Remarkably, melanoma cells reexpressing Syk display hallmarks of senescent cells, including reduction of proliferative activity and DNA synthesis, large and flattened morphology, senescence-associated beta-galactosidase activity, and heterochromatic foci. This phenotype is accompanied by hypophosphorylated retinoblastoma protein (Rb) and accumulation of p21, which depends on functional p53. Our results highlight a new role for Syk tyrosine kinase in regulating cellular senescence and identify Syk-mediated senescence as a novel tumor suppressor pathway the inactivation of which may contribute to melanoma tumorigenicity.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Melanoma/enzymology , Melanoma/pathology , Protein-Tyrosine Kinases/physiology , Cell Growth Processes/physiology , Cellular Senescence/physiology , Chemotaxis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Methylation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Melanoma/genetics , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Spheroids, Cellular , Syk Kinase , Transfection , Tumor Suppressor Protein p53/metabolism , Up-Regulation , src Homology Domains
15.
J Biol Chem ; 283(18): 12635-42, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18281284

ABSTRACT

Melanosomes are lysosome-related organelles specialized in melanin synthesis and transport. In this study, we show that microphthalmia-associated transcription factor (MITF) silencing induces melanosome gathering around the nucleus and causes the relocalization of Rab27A, Slac2a-Mlph, and Myo5a that control the transport of melanosomes on the actin network. In an attempt to elucidate the mechanism by which MITF controls melanosome distribution, we identify RAB27A as a new MITF target gene. Indeed, MITF silencing leads to a dramatic decrease in Rab27A expression and blocks the stimulation of Rab27A expression evoked by cAMP. Further, forced expression of MITF increases Rab27A expression, indicating that MITF is required and sufficient for Rab27A expression in melanoma cells. MITF binds to two E-boxes in the proximal region of the Rab27A promoter and stimulates its transcriptional activity. Finally, re-expression of Rab27A, in MITF-depleted cells, restores the transport of melanosomes to the cell periphery. These results show that RAB27A is a new direct transcriptional target of MITF and link MITF to melanosome transport, another key parameter of melanocyte differentiation and skin pigmentation. Interestingly, Rab27A is involved in other fundamental physiological functions, such as the transport of lytic granules and insulin secretion. Thus our results, deciphering the mechanism of Rab27A transcriptional regulation, have an interest that goes beyond the skin pigmentation field.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanosomes/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , rab GTP-Binding Proteins/genetics , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Base Sequence , Biological Transport , Cell Line, Tumor , E-Box Elements/genetics , Gene Silencing , Humans , Mice , Microphthalmia-Associated Transcription Factor/deficiency , Molecular Sequence Data , Myosin Type V/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Transcription, Genetic , rab GTP-Binding Proteins/metabolism , rab27 GTP-Binding Proteins
16.
J Invest Dermatol ; 127(2): 400-10, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16960555

ABSTRACT

Melanomas are malignant tumors of melanocytes that, if not detected early, are highly aggressive and poorly treatable. Activation of extracellular signal-regulated (ERK)/mitogen-activated protein (MAP) kinase signaling is commonly found in melanomas mainly through oncogenic mutations of B-Raf. We previously reported that activation of ERK/MAP kinase stimulates synthesis of fibronectin by upregulating the transcription factor early growth response-1 (Egr-1). To further analyze the link between ERK/MAP kinase pathway and fibronectin in melanoma, we have studied the regulation and role of fibronectin produced by melanoma cells bearing oncogenic B-Raf mutation. We show that fibronectin is expressed in situ during tumor progression and that high fibronectin and Egr-1 levels are found in cells expressing this mutation. Expression of active mutants of B-Raf induces fibronectin, whereas endogenous fibronectin is inhibited by small interfering RNA (siRNA)-mediated depletion of B-Raf or Egr-1. In contrast, stimulation of ERK pathway is insufficient to promote fibronectin upregulation in normal melanocytes. Finally, we show that suppression of fibronectin by siRNA leads to decreased melanoma cell invasiveness in vitro. These results reveal a tumor-specific regulation of fibronectin by constitutive ERK/MAP kinase signaling and indicate that self-production of fibronectin may play a role in melanoma tumorigenesis, by promoting tumor cell invasion.


Subject(s)
Fibronectins/metabolism , Melanoma/metabolism , Melanoma/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , Cells, Cultured , Disease Progression , Early Growth Response Protein 1/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibronectins/biosynthesis , Glutamic Acid , Humans , Melanocytes/metabolism , Mutation , Neoplasm Invasiveness , Valine
17.
Cancer Res ; 66(15): 7516-23, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885349

ABSTRACT

During progression of melanoma, loss of the cell-cell adhesion molecule E-cadherin contributes to uncontrolled growth and invasive behavior of transformed melanocytes. Secreted protein acidic and rich in cysteine (SPARC) is a nonstructural matricellular protein that regulates cell-matrix interactions leading to alterations in cell adhesion and proliferation. Overexpression of SPARC has been associated with progression of various cancers, including melanoma; however, its role in primary tumor development is not well defined. We show that normal human melanocytes overexpressing SPARC adopt a fibroblast-like morphology, concomitant with loss of E-cadherin and P-cadherin expression, and increased expression of mesenchymal markers. Concurrent with these changes, SPARC expression stimulates melanocyte motility and melanoma cell invasion. Expression of SPARC results in transcriptional down-regulation of E-cadherin that correlates with induction of Snail, a repressor of E-cadherin. Conversely, SPARC depletion leads to up-regulation of E-cadherin and reduces Snail levels, and SPARC-null cells exhibit a marked change in their mesenchymal phenotype. Finally, analysis of SPARC, Snail, and E-cadherin levels in melanocytes and malignant melanoma cell lines further supports the functional relationship among these proteins during melanoma progression. Our findings provide evidence for the role of SPARC in early transformation of melanocytes and identify a novel mechanism, whereby tumor-derived SPARC promotes tumorigenesis by mediating Snail induction and E-cadherin suppression.


Subject(s)
Cadherins/metabolism , Carrier Proteins/biosynthesis , Melanoma/metabolism , Melanoma/pathology , Transcription Factors/metabolism , Cadherins/biosynthesis , Cadherins/genetics , Calcium/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Melanocytes/metabolism , Melanocytes/pathology , Mesoderm/metabolism , Mesoderm/pathology , Neoplasm Invasiveness , Promoter Regions, Genetic , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Up-Regulation
18.
Oncogene ; 24(8): 1423-33, 2005 Feb 17.
Article in English | MEDLINE | ID: mdl-15608673

ABSTRACT

The matrix fibronectin protein is a multifunctional adhesive molecule that promotes migration and invasiveness of many tumors including melanomas. Increased fibronectin synthesis has been associated with the metastatic potential of melanoma cells; however, the molecular mechanisms underlying fibronectin overexpression during melanoma development are poorly understood. We report that hepatocyte growth factor/scatter factor (HGF) induces fibronectin expression and its extracellular assembly on the surface of melanoma cells through activation of mitogen-activated protein (MAP) kinase pathway, and induction and transcriptional activation of Early growth response-1 (Egr-1). Inhibition of B-RAF/MAP kinase pathway by dominant-negative mutants and by U0126-abrogated HGF-induced Egr-1, and chromatin immunoprecipitation showed that Egr-1 is bound to the fibronectin promoter in response to HGF. Exogenously expressed Egr-1 increased fibronectin levels, while blockage of Egr-1 activation by expression of the Egr-1 corepressor NAB2 interfered with the upregulation of fibronectin synthesis induced by HGF, indicating that Egr-1 exerts a significant role in fibronectin expression in response to HGF. Finally, analysis of the expression pattern of fibronectin in melanoma cells demonstrated that fibronectin levels are correlated with constitutive MAP kinase signaling. Our data define a novel mechanism that might have important implications in regulation of melanoma progression by autocrine HGF signaling or by constitutive activation of MAP kinase pathway.


Subject(s)
DNA-Binding Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/physiology , Fibronectins/biosynthesis , Hepatocyte Growth Factor/physiology , Immediate-Early Proteins/genetics , MAP Kinase Signaling System , Melanoma/metabolism , Skin Neoplasms/metabolism , Transcription Factors/genetics , Autocrine Communication/physiology , Butadienes/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/physiology , Early Growth Response Protein 1 , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibronectins/genetics , Genes, Reporter/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/pharmacology , Humans , Immediate-Early Proteins/physiology , Luciferases/analysis , Luciferases/genetics , Melanoma/genetics , Nitriles/pharmacology , Phosphorylation , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Signal Transduction , Skin Neoplasms/genetics , Transcription Factors/physiology , Up-Regulation/genetics , ras Proteins/physiology
19.
Pigment Cell Res ; 16(4): 374-82, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12859621

ABSTRACT

Microphthalmia-associated transcription factor (MITF) plays a pivotal role in melanocyte survival and differentiation. Nevertheless, until now it has not been possible to show that MITF regulates the expression of the endogenous tyrosinase or Tyrp1. Further, a direct involvement of MITF in the regulation of melanin synthesis, a key parameter of melanocyte differentiation, remains to be demonstrated. In the present report, using recombinant adenovirus encoding the wild-type or a dominant negative form of MITF, as well as stable cell lines expressing tetracycline inducible wild-type MITF, we reassessed the role of MITF in melanocyte differentiation and in the regulation of melanin synthesis. Immunofluorescence studies, as well as Western blot analyses, show that infection of B16 mouse melanoma cells or human melanocytes with adenovirus encoding wild-type MITF does not increase the expression of the endogenous melanogenic enzymes. However, infection with the MITF dominant negative mutant inhibits the expression of endogenous tyrosinase and Tyrp1 proteins and blocks cAMP-induced melanin synthesis. Thus, MITF is required but does not seem to be sufficient to induce the expression of melanogenic enzymes and we show for the first time a direct involvement of MITF in the regulation of melanin pigment synthesis. As a whole, our data point to the existence of still unknown regulatory mechanisms that co-operate or synergize with MITF to control melanogenic gene expression and melanin synthesis. The identification of such mechanisms will greatly improve our understanding of the melanocyte differentiation processes.


Subject(s)
DNA-Binding Proteins/physiology , Melanins/biosynthesis , Melanocytes/metabolism , Membrane Glycoproteins , Oxidoreductases , Transcription Factors/physiology , Adenoviridae/genetics , Animals , Cell Differentiation , Cell Line, Tumor , Cyclic AMP/pharmacology , Gene Expression , Genetic Vectors , Humans , Melanocytes/enzymology , Melanoma/metabolism , Mice , Microphthalmia-Associated Transcription Factor , Monophenol Monooxygenase/metabolism , Proteins/metabolism , Tetracycline/pharmacology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...