Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cancer Cell ; 26(1): 61-76, 2014 Jul 14.
Article in English | MEDLINE | ID: mdl-24981740

ABSTRACT

Although common cancer hallmarks are well established, lineage-restricted oncogenes remain less understood. Here, we report an inherent dependency of melanoma cells on the small GTPase RAB7, identified within a lysosomal gene cluster that distinguishes this malignancy from over 35 tumor types. Analyses in human cells, clinical specimens, and mouse models demonstrated that RAB7 is an early-induced melanoma driver whose levels can be tuned to favor tumor invasion, ultimately defining metastatic risk. Importantly, RAB7 levels and function were independent of MITF, the best-characterized melanocyte lineage-specific transcription factor. Instead, we describe the neuroectodermal master modulator SOX10 and the oncogene MYC as RAB7 regulators. These results reveal a unique wiring of the lysosomal pathway that melanomas exploit to foster tumor progression.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Lineage , Lysosomes/enzymology , Melanoma/enzymology , Skin Neoplasms/enzymology , rab GTP-Binding Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Melanoma/genetics , Melanoma/mortality , Melanoma/secondary , Melanoma/therapy , Mice , Neoplasm Invasiveness , Neoplasm Staging , Protein Transport , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Time Factors , Transfection , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
2.
Cell Rep ; 5(2): 493-507, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24139804

ABSTRACT

Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.


Subject(s)
GMP Reductase/metabolism , Melanoma/enzymology , Purine Nucleosides/biosynthesis , Animals , Cell Line, Tumor , Cell Movement , Extracellular Matrix/metabolism , GMP Reductase/antagonists & inhibitors , GMP Reductase/genetics , Guanosine Triphosphate/metabolism , HCT116 Cells , Humans , IMP Dehydrogenase/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Transplantation, Heterologous , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
3.
Pigment Cell Melanoma Res ; 26(4): 555-70, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23470013

ABSTRACT

The role of caveolin-1 (CAV1) in cancer is highly controversial. CAV1 suppresses genes that favor tumor development, yet also promotes focal adhesion turnover and migration of metastatic cells. How these contrasting observations relate to CAV1 function in vivo is unclear. Our previous studies implicate E-cadherin in CAV1-dependent tumor suppression. Here, we use murine melanoma B16F10 cells, with low levels of endogenous CAV1 and E-cadherin, to unravel how CAV1 affects tumor growth and metastasis and to assess how co-expression of E-cadherin modulates CAV1 function in vivo in C57BL/6 mice. We find that overexpression of CAV1 in B16F10 (cav-1) cells reduces subcutaneous tumor formation, but enhances metastasis relative to control cells. Furthermore, E-cadherin expression in B16F10 (E-cad) cells reduces subcutaneous tumor formation and lung metastasis when intravenously injected. Importantly, co-expression of CAV1 and E-cadherin in B16F10 (cav-1/E-cad) cells abolishes tumor formation, lung metastasis, increased Rac-1 activity, and cell migration observed with B16F10 (cav-1) cells. Finally, consistent with the notion that CAV1 participates in switching human melanomas to a more malignant phenotype, elevated levels of CAV1 expression correlated with enhanced migration and Rac-1 activation in these cells.


Subject(s)
Cadherins/metabolism , Caveolin 1/metabolism , Melanoma/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Gene Expression Profiling , Humans , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neoplasm Metastasis , Neuropeptides/metabolism , Phenotype , Skin Neoplasms/metabolism , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism
4.
J Invest Dermatol ; 133(10): 2436-2443, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23474946

ABSTRACT

We have previously reported a model for melanoma progression in which oscillation between melanoma cell phenotypes characterized by invasion or proliferation is fundamental to tumor heterogeneity and disease progression. In this study we examine the possible role of hypoxia as one of the microenvironmental influences driving metastatic progression by promoting a switch from a proliferative to an invasive phenotype. Immunohistochemistry on primary human cutaneous melanoma biopsies showed intratumoral heterogeneity for cells expressing melanocytic markers, and a loss of these markers correlated with hypoxic regions. Furthermore, we show that the downregulation of melanocytic markers is dependent on hypoxia inducible factor 1α (HIF1α), a known regulator of the hypoxic response. In vitro invasion assays showed that a hypoxic environment increases the invasiveness of proliferative melanoma cell cultures in a HIF1α-dependent manner. In contrast, invasive phenotype melanoma cells showed no increase in invasive potential upon exposure to hypoxia. Thus, exposure of proliferative melanoma cells to hypoxic microenvironments is sufficient, in a HIF1α-dependent manner, to downregulate melanocytic marker expression and increase their invasive potential.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/pathology , Melanoma/pathology , Skin Neoplasms/pathology , Tumor Microenvironment/physiology , Biomarkers, Tumor/metabolism , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MART-1 Antigen/genetics , MART-1 Antigen/metabolism , Melanoma/genetics , Melanoma/metabolism , Neoplasm Invasiveness , Phenotype , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tumor Cells, Cultured
5.
Pigment Cell Melanoma Res ; 25(3): 343-53, 2012 May.
Article in English | MEDLINE | ID: mdl-22336146

ABSTRACT

There is growing evidence that the metastatic spread of melanoma is driven not by a linear increase in tumorigenic aggressiveness, but rather by switching back and forth between two different phenotypes of metastatic potential. In vitro these phenotypes are respectively defined by the characteristics of strong proliferation/weak invasiveness and weak proliferation/strong invasiveness. Melanoma cell phenotype is tightly linked to gene expression. Taking advantage of this, we have developed a gene expression-based tool for predicting phenotype called Heuristic Online Phenotype Prediction. We demonstrate the predictive utility of this tool by comparing phenotype-specific signatures with measurements of characteristics of melanoma phenotype-specific biology in different melanoma cell lines and short-term cultures. We further show that 86% of 536 tested melanoma lines and short-term cultures are significantly associated with the phenotypes we describe. These findings reinforce the concept that a two-state system, as described by the phenotype switching model, underlies melanoma progression.


Subject(s)
Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Profiling , Melanoma/classification , Skin Neoplasms/classification , Cell Proliferation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Genetic Association Studies , Humans , Melanoma/genetics , Melanoma/pathology , Microarray Analysis , Neoplasm Invasiveness , Phenotype , Reproducibility of Results , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tissue Distribution , Tumor Cells, Cultured/classification
6.
Pigment Cell Melanoma Res ; 24(5): 932-43, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21801332

ABSTRACT

We recently identified GLI2, the most active of GLI transcription factors, as a direct TGF-ß/SMAD target, whose expression in melanoma cells is associated with increased invasiveness and metastatic capacity. In this work, we provide evidence that high GLI2 expression is inversely correlated with that of the melanocyte-specific transcription factor M-microphthalmia transcription factor (M-MITF) and associated transcriptional program. GLI2-expressing cell lines were characterized by the loss of M-MITF-dependent melanocytic differentiation markers and reduced pigmentation. The balance between M-MITF and GLI2 expression did not correlate with the presence or absence of BRAF-activating mutations, but rather was controlled by two distinct pathways: the TGF-ß pathway, which favors GLI2 expression, and the protein kinase A (PKA)/cAMP pathway, which pushes the balance toward high M-MITF expression. Furthermore, overexpression and knockdown experiments demonstrated that GLI2 and M-MITF reciprocally repress each other's expression and control melanoma cell invasion in an opposite manner. These findings thus identify GLI2 as a critical transcription factor antagonizing M-MITF function to promote melanoma cell phenotypic plasticity and invasive behavior.


Subject(s)
Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Melanoma/genetics , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/metabolism , Nuclear Proteins/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kruppel-Like Transcription Factors/genetics , Melanoma/physiopathology , Mice , Mice, Nude , Microphthalmia-Associated Transcription Factor/genetics , Neoplasm Invasiveness , Nuclear Proteins/genetics , Pigmentation/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Skin Neoplasms/physiopathology , Smad Proteins/metabolism , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/pathology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Xenograft Model Antitumor Assays , Zinc Finger Protein Gli2
7.
Pigment Cell Melanoma Res ; 24(4): 631-42, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21599871

ABSTRACT

Recent observations suggest that melanoma cells drive disease progression by switching back and forth between phenotypic states of proliferation and invasion. Phenotype switching has been linked to changes in Wnt signalling, and we therefore looked for cell phenotype-specific differences in the levels and activity of ß-catenin and its LEF/TCF co-factors. We found that while cytosolic ß-catenin distribution is phenotype-specific (membrane-associated in proliferative cells and cytosolic in invasive cells), its nuclear distribution and activity is not. Instead, the expression patterns of two ß-catenin co-factors, LEF1 and TCF4, are both phenotype-specific and inversely correlated. LEF1 is preferentially expressed by differentiated/proliferative phenotype cells and TCF4 by dedifferentiated/invasive phenotype cells. Knock-down experiments confirmed that these co-factors are important for the phenotype-specific expression of M-MITF, WNT5A and other genes and that LEF1 suppresses TCF4 expression independently of ß-catenin. Our data show that melanoma cell phenotype switching behaviour is regulated by differential LEF1/TCF4 activity.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Models, Biological , Neoplasm Invasiveness , Phenotype , Transcription Factor 4 , Transcription Factors/metabolism , Wnt Proteins/pharmacology , beta Catenin/metabolism
9.
Pigment Cell Melanoma Res ; 24(2): 326-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21176117

ABSTRACT

Oncogenic mutations within the MAPK pathway are frequent in melanoma, and targeting of MAPK signaling has yielded spectacular responses in a significant number of patients that last for several months before relapsing. We investigated the effects of two different inhibitors of MAPK signaling in proliferative and invasive melanoma cell cultures with various mutations in the MAPK pathway. Proliferative melanoma cells were more susceptible to pathway inhibition than invasive phenotype cells, irrespective of BRAF mutation status, while invasive phenotype cell response was dependent on BRAF mutation status. Critically, MAPK pathway inhibition of proliferative phenotype cells resulted in acquisition of invasive phenotype characteristics. These results show that melanoma cell phenotype is an important factor in MAPK pathway inhibition response. This suggests that while current therapeutic strategies target proliferative melanoma cells, future approaches should also account for the invasive phenotype population.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phenotype , Proto-Oncogene Proteins B-raf/genetics , raf Kinases/antagonists & inhibitors , Adult , Aged , Cell Proliferation , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Profiling , Humans , MAP Kinase Signaling System/physiology , Male , Melanoma/drug therapy , Melanoma/metabolism , Microarray Analysis , Middle Aged , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Neoplasm Invasiveness , Proto-Oncogene Proteins B-raf/metabolism
11.
Pigment Cell Melanoma Res ; 23(6): 746-59, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20726948

ABSTRACT

Tumours comprise multiple phenotypically distinct subpopulations of cells, some of which are proposed to possess stem cell-like properties, being able to self-renew, seed and maintain tumours, and provide a reservoir of therapeutically resistant cells. Here, we use melanoma as a model to explore the validity of the cancer stem cell hypothesis in the light of accumulating evidence that melanoma progression may instead be driven by phenotype-switching triggered by genetic lesions that impose an increased sensitivity to changes in the tumour microenvironment. Although at any given moment cells within a tumour may exhibit differentiated, proliferative or invasive phenotypes, an ability to switch phenotypes implies that most cells will have the potential to adopt a stem cell-like identity. Insights into the molecular events underpinning phenotype-switching in melanoma highlight the close relationship between signalling pathways that generate, maintain and activate melanocyte stem cells as well as the inverse correlation between proliferation and invasive potentials. An understanding of phenotype-switching in melanoma, and in particular the signalling events that regulate the expression of the microphthalmia-associated transcription factor Mitf, points to new therapeutic opportunities aimed at eradicating therapeutically resistant stem cell-like melanoma cells.


Subject(s)
Melanoma/pathology , Neoplastic Stem Cells/pathology , Animals , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/genetics , Melanoma/therapy , Microphthalmia-Associated Transcription Factor/metabolism , Neoplastic Stem Cells/metabolism , Phenotype , Precancerous Conditions/pathology
12.
J Natl Cancer Inst ; 102(15): 1148-59, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20660365

ABSTRACT

BACKGROUND: The transforming growth factor-beta (TGF-beta) pathway, which has both tumor suppressor and pro-oncogenic activities, is often constitutively active in melanoma and is a marker of poor prognosis. Recently, we identified GLI2, a mediator of the hedgehog pathway, as a transcriptional target of TGF-beta signaling. METHODS: We used real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blotting to determine GLI2 expression in human melanoma cell lines and subsequently classified them as GLI2high or as GLI2low according to their relative GLI2 mRNA and protein expression levels. GLI2 expression was reduced in a GLI2high cell line with lentiviral expression of short hairpin RNA targeting GLI2. We assessed the role of GLI2 in melanoma cell invasiveness in Matrigel assays. We measured secretion of matrix metalloproteinase (MMP)-2 and MMP-9 by gelatin zymography and expression of E-cadherin by western blotting and RT-PCR. The role of GLI2 in development of bone metastases was determined following intracardiac injection of melanoma cells in immunocompromised mice (n = 5-13). Human melanoma samples (n = 79) at various stages of disease progression were analyzed for GLI2 and E-cadherin expression by immunohistochemistry, in situ hybridization, or RT-PCR. All statistical tests were two-sided. RESULTS: Among melanoma cell lines, increased GLI2 expression was associated with loss of E-cadherin expression and with increased capacity to invade Matrigel and to form bone metastases in mice (mean osteolytic tumor area: GLI2high vs GLI2low, 2.81 vs 0.93 mm(2), difference = 1.88 mm(2), 95% confidence interval [CI] = 1.16 to 2.60, P < .001). Reduction of GLI2 expression in melanoma cells that had expressed high levels of GLI2 substantially inhibited both basal and TGF-beta-induced cell migration, invasion (mean number of Matrigel invading cells: shGLI2 vs shCtrl (control), 52.6 vs 100, difference = 47.4, 95% CI = 37.0 to 57.8, P = .024; for shGLI2 + TGF-beta vs shCtrl + TGF-beta, 31.0 vs 161.9, difference = -130.9, 95% CI = -96.2 to -165.5, P = .002), and MMP secretion in vitro and the development of experimental bone metastases in mice. Within human melanoma lesions, GLI2 expression was heterogeneous, associated with tumor regions in which E-cadherin was lost and increased in the most aggressive tumors. CONCLUSION: GLI2 was directly involved in driving melanoma invasion and metastasis in this preclinical study.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Kruppel-Like Transcription Factors/metabolism , Melanoma/metabolism , Melanoma/pathology , Nuclear Proteins/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Animals , Blotting, Western , Cadherins/metabolism , Cell Line, Tumor , Collagen , Drug Combinations , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Immunocompromised Host , Immunohistochemistry , In Situ Hybridization , Kruppel-Like Transcription Factors/genetics , Laminin , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Melanoma/secondary , Mice , Neoplasm Invasiveness , Nuclear Proteins/genetics , Polymerase Chain Reaction , Proteoglycans , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation , Zinc Finger Protein Gli2
13.
Melanoma Res ; 20(4): 349-55, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20526217

ABSTRACT

To date there is no effective therapy for metastatic melanoma and at the molecular level the disease progression is poorly understood. A recent study by our group led to the development of a novel phenotype switching model for melanoma progression, wherein cells transition back-and-forth between states of proliferation and invasion to drive disease progression. To explore the model's clinical relevance we interrogated phenotype-specific expression patterns in human melanoma patient material. A matched primary/metastasis pair from a human melanoma patient was obtained and immunohistochemically stained for proliferative and invasive phenotype markers. These were also stained for hypoxia and blood vessel markers. Proliferative phenotype markers Melan-A and Mitf showed consistent anti-correlation with invasive phenotype marker Wnt5A and hypoxia marker Glut-1. These also correlated with observed intra-tumoural vascularization patterns. Similar pattern distributions were present in both primary and metastasis samples. Strikingly, we observed that late phase metastatic melanoma cells adopt morphologies and behaviours identical to very early phase cells. The expression patterns observed closely matched expectations derived from previous in vitro and xenografting experiments. These results highlight the likelihood that disease progression involves melanoma cells retaining the capacity to regulate the expression of metastatic potential critical factors according to changing microenvironmental conditions.


Subject(s)
Melanoma/pathology , Skin Neoplasms/pathology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Disease Progression , Female , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , MART-1 Antigen , Melanoma/genetics , Melanoma/metabolism , Melanoma/secondary , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
14.
Pigment Cell Melanoma Res ; 23(3): 306-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20236251

ABSTRACT

Coverage on: Berger, M.F. et al. (2010).Integrative analysis of the melanoma transcriptome.Genome Res. (doi: 10.1101/gr.103697.109).


Subject(s)
Melanoma/genetics , Sequence Analysis, DNA/methods , Fusion Proteins, bcr-abl/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Rearrangement/genetics , Humans , Oligonucleotide Array Sequence Analysis
15.
J Biol Chem ; 284(42): 28704-12, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19696445

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are important modulators for optimizing signal transduction of many pathways, including the Wnt pathways. We demonstrate that HSPG glycosaminoglycan levels increased with increasing metastatic potential of melanoma cells. Previous studies have demonstrated that Wnt5A increases the invasiveness of melanoma cells. We further demonstrate that HSPGs potentiate Wnt5A signaling, since enzymatic removal of the HSPG backbone resulted in a decrease in cellular Wnt5A levels, an increase in secreted Wnt5A in cell media, a decrease in downstream signaling, and ultimately, a decrease in invasiveness. Specifically, syndecan 1 and syndecan 4 expression correlated to Wnt5A expression and melanoma malignancy. Knockdown of syndecan 1 or 4 caused decreases in cell invasion, which could be restored by treating the cells with recombinant Wnt5A. These data indicate that syndecan 1 and 4 correlate to increased metastatic potential in melanoma patients and are an important component of the Wnt5A autocrine signaling loop, the activation of which leads to increased metastasis of melanoma.


Subject(s)
Gene Expression Regulation, Neoplastic , Heparan Sulfate Proteoglycans/metabolism , Melanoma/pathology , Proto-Oncogene Proteins/chemistry , Skin Neoplasms/pathology , Wnt Proteins/chemistry , Cell Line, Tumor , Cell Membrane/metabolism , Heparan Sulfate Proteoglycans/chemistry , Humans , Melanoma/metabolism , Neoplasm Metastasis , Recombinant Proteins/chemistry , Signal Transduction , Skin Neoplasms/metabolism , Syndecan-1/metabolism , Syndecan-4/metabolism , Wnt Proteins/metabolism , Wnt-5a Protein , Wound Healing
16.
Proc Natl Acad Sci U S A ; 106(32): 13493-8, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19651608

ABSTRACT

Cancer/testis (CT) genes are predominantly expressed in human germ line cells, but not somatic tissues, and frequently become activated in different cancer types. Several CT antigens have already proved to be useful biomarkers and are promising targets for therapeutic cancer vaccines. The aim of the present study was to investigate the expression of CT antigens in breast cancer. Using previously generated massively parallel signature sequencing (MPSS) data, together with 9 publicly available gene expression datasets, the expression pattern of CT antigens located on the X chromosome (CT-X) was interrogated. Whereas a minority of unselected breast cancers was found to contain CT-X transcripts, a significantly higher expression frequency was detected in estrogen and progesterone receptor (ER) negative breast cancer cell lines and primary breast carcinomas. A coordinated pattern of CT-X antigen expression was observed, with MAGEA and NY-ESO-1/CTAG1B being the most prevalent antigens. Immunohistochemical staining confirmed the correlation of CT-X antigen expression and ER negativity in breast tumors and demonstrated a trend for their coexpression with basal cell markers. Because of the limited therapeutic options for ER-negative breast cancers, vaccines based on CT-X antigens might prove to be useful.


Subject(s)
Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Antigens, Neoplasm/metabolism , Breast Neoplasms/classification , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Expressed Sequence Tags , Female , Humans , Immunohistochemistry , Membrane Proteins/genetics , Neoplasm Metastasis/pathology , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tissue Array Analysis
17.
Pigment Cell Melanoma Res ; 22(4): 445-53, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19368689

ABSTRACT

Proliferative resistance to transforming growth factor beta (TGF-beta) is regarded as a critical turning point in the malignant progression of many cancer types. In melanoma this resistance is associated with more aggressive metastatic behaviour. A recent study by our group identified proliferative and invasive subtypes of melanoma cultures and found that these are, respectively, susceptible and resistant to TGF-beta suppression of proliferation. Here, using previously characterised proliferative and invasive phenotype melanoma cultures, we explored molecular responses involved in modulating susceptibility to TGF-beta-mediated inhibition of proliferation. The Id2 gene was identified as being expressed more strongly in invasive phenotype cells less susceptible to TGF-beta repression than in proliferative phenotype cells. We correlated TGF-beta repression of Id2 gene expression in proliferative phenotype cells with p15(Ink4b) induction and cell cycle arrest. Furthermore, ectopic Id2 expression in proliferative phenotype cells counteracted p15(Ink4b) induction and consequently protected them from TGF-beta-mediated inhibition of proliferation. We conclude that transition to increased aggressiveness in melanoma cells requires Id2 upregulation to suppress TGF-beta induction of p15(Ink4b) and thus help to circumvent TGF-beta-mediated inhibition of proliferation.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Inhibitor of Differentiation Protein 2/physiology , Melanoma/metabolism , Transforming Growth Factor beta/physiology , Cell Line, Tumor , G1 Phase/physiology , Gene Expression Regulation, Neoplastic , Humans , Melanoma/pathology , Neoplasm Invasiveness
18.
Clin Cancer Res ; 15(6): 1954-63, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276289

ABSTRACT

PURPOSE: The sperm-derived SPANX family proteins can be found expressed in human tumors. Here, we aimed to perform a comprehensive study to evaluate immunotherapeutic relevance of one of its members, SPANX-B. We wanted to test its expression pattern in human tumors and to evaluate CD4(+) and CD8(+) T-cell responses in healthy humans after in vitro immunizations. EXPERIMENTAL DESIGN: Expression of SPANX-B in human malignancies, including a multitumor tissue array of 145 primary tumors, was assessed using reverse transcription-PCR, Western blotting, and immunohistochemical analysis. T-cell immunogenicity and immunodominant epitopes of SPANX-B were studied using in vitro immunizations of healthy human donor-derived leukocytes. RESULTS: SPANX-B was abundantly expressed in melanoma and carcinomas of lung, ovary, colon, and breast. In melanoma, tissue array data indicated that it was expressed in advanced and metastatic disease. Unlike most tumor-associated antigens, SPANX-B was an immunogenic antigen that was recognized by circulating T-cell precursors in healthy humans. Importantly, these T cells were readily expanded to generate SPANX-B-specific helper CD4(+) and cytolytic CD8(+) T cells that recognized unique immunodominant epitopes: at least one HLA-DR-restricted Pep-9 epitope (SPANX-B(12-23)) and two HLA-A2-restricted Pep-2 and Pep-4 epitopes (SPANX-B(23-31) and SPANX-B(57-65), respectively). CD8(+) T cells were fully functional to recognize and lyse HLA-A2-expressing tumors, including primary human melanomas. CONCLUSIONS: SPANX-B is an immunogenic sperm-derived antigen that is expressed in several human tumors. SPANX-B is also efficiently recognized by the human T-cell immune arm, indicating its significant value for the development of protective and therapeutic cancer vaccines.


Subject(s)
Antigens, Neoplasm/analysis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Nuclear Proteins/analysis , Amino Acid Sequence , Cancer Vaccines/immunology , Humans , Immunodominant Epitopes , Molecular Sequence Data , Neoplasms/therapy , Nuclear Proteins/immunology
19.
Endocrinology ; 150(6): 2551-60, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19213840

ABSTRACT

The actin-binding protein filamin A (FLNa) affects the intracellular trafficking of various classes of receptors and has a potential role in oncogenesis. However, it is unclear whether FLNa regulates the signaling capacity and/or down-regulation of the activated epidermal growth factor receptor (EGFR). Here it is shown that partial knockdown of FLNa gene expression blocked ligand-induced EGFR responses in metastatic human melanomas. To gain greater insights into the role of FLNa in EGFR activation and intracellular sorting, we used M2 melanoma cells that lack endogenous FLNa and a subclone in which human FLNa cDNA has been stably reintroduced (M2A7 cells). Both tyrosine phosphorylation and ubiquitination of EGFR were significantly lower in epidermal growth factor (EGF)-stimulated M2 cells when compared with M2A7 cells. Moreover, the lack of FLNa interfered with EGFR interaction with the ubiquitin ligase c-Cbl. M2 cells exhibited marked resistance to EGF-induced receptor degradation, which was very active in M2A7 cells. Despite comparable rates of EGF-mediated receptor endocytosis, internalized EGFR colocalized with the lysosomal marker lysosome-associated membrane protein-1 in M2A7 cells but not M2 cells, in which EGFR was found to be sequestered in large vesicles and subsequently accumulated in punctated perinuclear structures after EGF stimulation. These results suggest the requirement of FLNa for efficient EGFR kinase activation and the sorting of endocytosed receptors into the degradation pathway.


Subject(s)
Contractile Proteins/metabolism , ErbB Receptors/metabolism , Melanoma/metabolism , Microfilament Proteins/metabolism , Phosphotransferases/metabolism , Signal Transduction/physiology , Biopsy , Cell Line, Tumor , Contractile Proteins/genetics , DNA, Complementary/genetics , Down-Regulation/physiology , Endocytosis/physiology , Filamins , Humans , Lysosomes/metabolism , Melanoma/pathology , Microfilament Proteins/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Transfection
20.
BMC Cancer ; 9: 52, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19216735

ABSTRACT

BACKGROUND: The C-terminal binding protein 1 (CtBP1) is a known co-repressor of gene transcription. We recently revealed that CtBP1 expression is lost in melanoma cells and melanoma inhibitory activity (MIA) expression is subsequently increased. The present study was performed to evaluate a more general role of CtBP1 in human melanoma and identify further CtBP1-regulated target genes. METHODS: Sequence analysis and expression profile of CtBP1 in melanoma cell lines were done by PCR. Boyden Chamber assays and co-immunoprecipitation were performed to investigate the functional role of CtBP1. Gene expression analysis and micro array data were used to define target genes. RESULTS: Interestingly, we detected an alternative splice product of CtBP1 with unknown function whose expression is induced at reduction of full length CtBP1. Overexpression of full length CtBP1 in melanoma cells had no effect on cell proliferation but did influence cell migration and invasiveness. To understand the effect of CtBP1 we identified putative LEF/TCF target genes found to be strongly expressed in melanoma using DNA microarray analysis. We focused on fourteen genes not previously associated with melanoma. Detailed analysis revealed that most of these were known to be involved in tumor metastasis. Eleven genes had expression profiles associated with melanoma cell invasiveness. CONCLUSION: In summary, this study revealed that reduction of CtBP1 expression is correlated with migratory, invasive potential of melanoma cells.


Subject(s)
Alcohol Oxidoreductases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/pathology , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Base Sequence , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Humans , Melanoma/metabolism , Molecular Sequence Data , Neoplasm Invasiveness
SELECTION OF CITATIONS
SEARCH DETAIL
...