Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nat Commun ; 5: 5200, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25335460

ABSTRACT

Targeting the vasculature remains a promising approach for treating solid tumours; however, the mechanisms of tumour neovascularization are diverse and complex. Here we uncover a new subpopulation of melanoma cells that express the vascular cell adhesion molecule PECAM1, but not VEGFR-2, and participate in a PECAM1-dependent form of vasculogenic mimicry (VM). Clonally derived PECAM1(+) tumour cells coalesce to form PECAM1-dependent networks in vitro and they generate well-perfused, vascular endothelial growth factor (VEGF)-independent channels in mice. The neural crest specifier AP-2α is diminished in PECAM1(+) melanoma cells and is a transcriptional repressor of PECAM1. Re-introduction of AP-2α into PECAM1(+) tumour cells represses PECAM1 and abolishes tube-forming ability, whereas AP-2α knockdown in PECAM1(-) tumour cells upregulates PECAM1 expression and promotes tube formation. Thus, VM-competent subpopulations, rather than all cells within a tumour, may instigate VM, supplant host-derived endothelium, and form PECAM1-dependent conduits that are not diminished by neutralizing VEGF.


Subject(s)
Melanoma/metabolism , Neovascularization, Pathologic/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Angiogenesis Inhibitors/chemistry , Animals , Cell Line, Tumor , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , RNA, Small Interfering/metabolism , Skin Neoplasms/metabolism , Transcription Factor AP-2/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Environ Mol Mutagen ; 55(6): 457-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24616037

ABSTRACT

A systems biology approach was applied to investigate the mechanisms of chromosomal instability in melanoma cell lines. Chromosomal instability was quantified using array comparative genomic hybridization to identify somatic copy number alterations (deletions and duplications). Primary human melanocytes displayed an average of 8.5 alterations per cell primarily representing known polymorphisms. Melanoma cell lines displayed 25 to 131 alterations per cell, with an average of 68, indicative of chromosomal instability. Copy number alterations included approximately equal numbers of deletions and duplications with greater numbers of hemizygous (-1,+1) alterations than homozygous (-2,+2). Melanoma oncogenes, such as BRAF and MITF, and tumor suppressor genes, such as CDKN2A/B and PTEN, were included in these alterations. Duplications and deletions were functional as there were significant correlations between DNA copy number and mRNA expression for these genes. Spectral karyotype analysis of three lines confirmed extensive chromosomal instability with polyploidy, aneuploidy, deletions, duplications, and chromosome rearrangements. Bioinformatic analysis identified a signature of gene expression that was correlated with chromosomal instability but this signature provided no clues to the mechanisms of instability. The signature failed to generate a significant (P = 0.105) prediction of melanoma progression in a separate dataset. Chromosomal instability was not correlated with elements of DNA damage response (DDR) such as radiosensitivity, nucleotide excision repair, expression of the DDR biomarkers γH2AX and P-CHEK2, nor G1 or G2 checkpoint function. Chromosomal instability in melanoma cell lines appears to influence gene function but it is not simply explained by alterations in the system of DDR.


Subject(s)
Chromosomal Instability/genetics , Melanoma/genetics , Systems Biology/methods , Cell Line, Tumor , Comparative Genomic Hybridization , Computational Biology , DNA Copy Number Variations/genetics , DNA Damage/genetics , DNA Damage/physiology , Humans , Karyotyping , Oncogenes/genetics
3.
Pigment Cell Melanoma Res ; 27(4): 653-63, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24628946

ABSTRACT

Somatic sequencing of cancers has produced new insight into tumorigenesis, tumor heterogeneity, and disease progression, but the vast majority of genetic events identified are of indeterminate clinical significance. Here, we describe a NextGen sequencing approach to fully analyzing 248 genes, including all those of known clinical significance in melanoma. This strategy features solution capture of DNA followed by multiplexed, high-throughput sequencing and was evaluated in 31 melanoma cell lines and 18 tumor tissues from patients with metastatic melanoma. Mutations in melanoma cell lines correlated with their sensitivity to corresponding small molecule inhibitors, confirming, for example, lapatinib sensitivity in ERBB4 mutant lines and identifying a novel activating mutation of BRAF. The latter event would not have been identified by clinical sequencing and was associated with responsiveness to a BRAF kinase inhibitor. This approach identified focal copy number changes of PTEN not found by standard methods, such as comparative genomic hybridization (CGH). Actionable mutations were found in 89% of the tumor tissues analyzed, 56% of which would not be identified by standard-of-care approaches. This work shows that targeted sequencing is an attractive approach for clinical use in melanoma.


Subject(s)
High-Throughput Nucleotide Sequencing , Melanoma/genetics , Mutation , Neoplasm Proteins/genetics , Cell Line, Tumor , Female , Humans , Male
4.
J Histochem Cytochem ; 62(3): 185-96, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24309508

ABSTRACT

The DNA damage response (DDR) coordinates DNA repair with cell cycle checkpoints to ameliorate or mitigate the pathological effects of DNA damage. Automated quantitative analysis (AQUA) and Tissue Studio are commercial technologies that use digitized immunofluorescence microscopy images to quantify antigen expression in defined tissue compartments. Because DDR is commonly activated in cancer and may reflect genetic instability within the lesion, a method to quantify DDR in cancer offers potential diagnostic and/or prognostic value. In this study, both AQUA and Tissue Studio algorithms were used to quantify the DDR in radiation-damaged skin fibroblasts, melanoma cell lines, moles, and primary and metastatic melanomas. Digital image analysis results for three markers of DDR (γH2AX, P-ATM, P-Chk2) correlated with immunoblot data for irradiated fibroblasts, whereas only γH2AX and P-Chk2 correlated with immunoblot data in melanoma cell lines. Melanoma cell lines displayed substantial variation in γH2AX and P-Chk2 expression, and P-Chk2 expression was significantly correlated with radioresistance. Moles, primary melanomas, and melanoma metastases in brain, lung and liver displayed substantial variation in γH2AX expression, similar to that observed in melanoma cell lines. Automated digital analysis of immunofluorescent images stained for DDR biomarkers may be useful for predicting tumor response to radiation and chemotherapy.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Biomarkers, Tumor/metabolism , Checkpoint Kinase 2/metabolism , DNA Damage , Histones/metabolism , Cell Line, Tumor , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Image Processing, Computer-Assisted , Melanoma/diagnosis , Melanoma/metabolism , Neoplasm Metastasis , Nevus/diagnosis , Nevus/metabolism , Skin/cytology , Skin Neoplasms/diagnosis , Skin Neoplasms/metabolism
5.
J Clin Invest ; 123(5): 2257-67, 2013 May.
Article in English | MEDLINE | ID: mdl-23585477

ABSTRACT

Metastatic melanoma is one of the most aggressive forms of cutaneous cancers. Although recent therapeutic advances have prolonged patient survival, the prognosis remains dismal. C-MER proto-oncogene tyrosine kinase (MERTK) is a receptor tyrosine kinase with oncogenic properties that is often overexpressed or activated in various malignancies. Using both protein immunohistochemistry and microarray analyses, we demonstrate that MERTK expression correlates with disease progression. MERTK expression was highest in metastatic melanomas, followed by primary melanomas, while the lowest expression was observed in nevi. Additionally, over half of melanoma cell lines overexpressed MERTK compared with normal human melanocytes; however, overexpression did not correlate with mutations in BRAF or RAS. Stimulation of melanoma cells with the MERTK ligand GAS6 resulted in the activation of several downstream signaling pathways including MAPK/ERK, PI3K/AKT, and JAK/STAT. MERTK inhibition via shRNA reduced MERTK-mediated downstream signaling, reduced colony formation by up to 59%, and diminished tumor volume by 60% in a human melanoma murine xenograft model. Treatment of melanoma cells with UNC1062, a novel MERTK-selective small-molecule tyrosine kinase inhibitor, reduced activation of MERTK-mediated downstream signaling, induced apoptosis in culture, reduced colony formation in soft agar, and inhibited invasion of melanoma cells. This work establishes MERTK as a therapeutic target in melanoma and provides a rationale for the continued development of MERTK-targeted therapies.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Skin Neoplasms/metabolism , Animals , Apoptosis , Cell Line, Tumor , Disease Progression , Enzyme Inhibitors/pharmacology , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Melanocytes/metabolism , Mice , Mice, SCID , Microscopy, Fluorescence , Models, Biological , Mutation , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Mas , c-Mer Tyrosine Kinase
6.
Oncologist ; 17(12): 1496-503, 2012.
Article in English | MEDLINE | ID: mdl-23006498

ABSTRACT

PURPOSE: This phase I study assessed the toxicity and safety of combining daily lapatinib with radiation therapy. Sequential tumor biopsies were obtained to evaluate changes in biomarkers, such as epidermal growth factor receptor (EGFR) and human EGFR-2 (HER2) signaling pathways. METHODS: Eligibility for this dose-escalation study included unresectable and locally recurrent or chemotherapy-refractory and locally advanced breast cancer, and adequate organ function. Patients underwent three serial biopsies: at baseline, after 1 week of lapatinib alone, and after 1 week of lapatinib and radiation. Endpoints included determination of toxicity, maximum tolerated dose, and analysis of the effect of lapatinib with or without radiation on EGFR and HER2 signaling pathways by immunohistochemistry. RESULTS: Doses of lapatinib up to 1,500 mg/day were well tolerated. Toxicity of grade 3 or more was limited to radiation dermatitis and pain. Out of 19 patients treated, in field responses per response evaluation criteria in solid tumors criteria were complete in four patients and partial in six patients. Serial biopsies were obtained in 16 patients with no complications. Total Her2 was relatively unchanged while phospho-Her2, phospho-Akt, and phospho-ERK showed variable responses to both lapatinib alone and dual therapy with lapatinib and radiation. CONCLUSIONS: The combination of lapatinib and radiation was well tolerated in this patient cohort. Overall local response rates were comparable to those reported in other studies in this patient population. Biopsies were safely performed at all time points. Inhibition of HER2 and downstream signaling pathways was identified, although no strong correlation with response was seen.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Quinazolines/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Biopsy/methods , Cohort Studies , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Immunohistochemistry , Lapatinib , Maximum Tolerated Dose , Middle Aged , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction
7.
Pigment Cell Melanoma Res ; 25(4): 514-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22540896

ABSTRACT

Melanoma cell lines and normal human melanocytes (NHM) were assayed for p53-dependent G1 checkpoint response to ionizing radiation (IR)-induced DNA damage. Sixty-six percent of melanoma cell lines displayed a defective G1 checkpoint. Checkpoint function was correlated with sensitivity to IR with checkpoint-defective lines being radio-resistant. Microarray analysis identified 316 probes whose expression was correlated with G1 checkpoint function in melanoma lines (P≤0.007) including p53 transactivation targets CDKN1A, DDB2, and RRM2B. The 316 probe list predicted G1 checkpoint function of the melanoma lines with 86% accuracy using a binary analysis and 91% accuracy using a continuous analysis. When applied to microarray data from primary melanomas, the 316 probe list was prognostic of 4-yr distant metastasis-free survival. Thus, p53 function, radio-sensitivity, and metastatic spread may be estimated in melanomas from a signature of gene expression.


Subject(s)
G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Profiling , Melanoma/genetics , Skin Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Probes/metabolism , Gene Expression Regulation, Neoplastic , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/diagnosis , Melanoma/pathology , Prognosis , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
8.
Radiother Oncol ; 98(3): 394-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21295875

ABSTRACT

PURPOSE: To assess the relative radiosensitivities of a large collection of melanoma cell lines and to determine whether pharmacologic inhibition of mutant B-RAF with PLX-4032 can radiosensitize B-Raf+ melanoma cells. MATERIALS AND METHODS: A large collection of melanoma cell lines (n=37) were treated with 0-8Gy IR and clonogenic survival assays used to generate survival curves to rank relative radiosensitivities among the cell lines. The ability of a B-RAF inhibitor, PLX-4032, to radiosensitize highly radioresistant B-Raf+ cells was also assessed by clonogenic cell survival and spheroid invasion assays and the effects of treatment on the cell cycle assessed by FACS. RESULTS: Melanoma cell lines displayed a very large, heterogeneous range of SF2 values (1.002-0.053) with a mean of 0.51. Cell lines with surviving fractions of 0.29 or less at SF2 and SF4 were observed at a high frequency of 18.9% and 70.2%, respectively. Treatment of B-Raf+ cells with the B-RAF inhibitor PLX-4032 in combination with radiation provided enhanced inhibition of both colony formation and invasion, and radiosensitized cells through an increase in G(1) arrest. CONCLUSIONS: Our data suggest that melanomas are not uniformly radioresistant with a significant subset displaying inherent radiosensitivity. Pharmacologic inhibition of B-RAF with PLX-4032 effectively radiosensitized B-Raf+ melanoma cells suggesting that this combination approach could provide improved radiotherapeutic response in B-Raf+ melanoma patients.


Subject(s)
Indoles/pharmacology , Melanoma/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Sulfonamides/pharmacology , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib
9.
Cancer Res ; 70(12): 4922-30, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501836

ABSTRACT

Sunlight UV exposure produces DNA photoproducts in skin that are repaired solely by nucleotide excision repair in humans. A significant fraction of melanomas are thought to result from UV-induced DNA damage that escapes repair; however, little evidence is available about the functional capacity of normal human melanocytes, malignant melanoma cells, and metastatic melanoma cells to repair UV-induced photoproducts in DNA. In this study, we measured nucleotide excision repair in both normal melanocytes and a panel of melanoma cell lines. Our results show that in 11 of 12 melanoma cell lines tested, UV photoproduct repair occurred as efficiently as in primary melanocytes. Importantly, repair capacity was not affected by mutation in the N-RAS or B-RAF oncogenes, nor was a difference observed between a highly metastatic melanoma cell line (A375SM) or its parental line (A375P). Lastly, we found that although p53 status contributed to photoproduct removal efficiency, its role did not seem to be mediated by enhanced expression or activity of DNA binding protein DDB2. We concluded that melanoma cells retain capacity for nucleotide excision repair, the loss of which probably does not commonly contribute to melanoma progression.


Subject(s)
DNA Damage/radiation effects , DNA Repair , Genes, ras/physiology , Melanocytes/physiology , Melanoma/genetics , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/genetics , Cells, Cultured , Collagen/metabolism , DNA, Neoplasm/radiation effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Combinations , Humans , Laminin/metabolism , Melanoma/metabolism , Mutation/genetics , Proteoglycans/metabolism , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
10.
Int J Radiat Oncol Biol Phys ; 77(2): 575-81, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20457354

ABSTRACT

PURPOSE: To determine whether lapatinib, a dual epidermal growth factor receptor (EGFR)/HER2 kinase inhibitor, can radiosensitize EGFR+ or HER2+ breast cancer xenografts. METHODS AND MATERIALS: Mice bearing xenografts of basal-like/EGFR+ SUM149 and HER2+ SUM225 breast cancer cells were treated with lapatinib and fractionated radiotherapy and tumor growth inhibition correlated with alterations in ERK1 and AKT activation by immunohistochemistry. RESULTS: Basal-like/EGFR+ SUM149 breast cancer tumors were completely resistant to treatment with lapatinib alone but highly growth impaired with lapatinib plus radiotherapy, exhibiting an enhancement ratio average of 2.75 and a fractional tumor product ratio average of 2.20 during the study period. In contrast, HER2+ SUM225 breast cancer tumors were highly responsive to treatment with lapatinib alone and yielded a relatively lower enhancement ratio average of 1.25 during the study period with lapatinib plus radiotherapy. Durable tumor control in the HER2+ SUM225 model was more effective with the combination treatment than either lapatinib or radiotherapy alone. Immunohistochemical analyses demonstrated that radiosensitization by lapatinib correlated with ERK1/2 inhibition in the EGFR+ SUM149 model and with AKT inhibition in the HER2+ SUM225 model. CONCLUSION: Our data suggest that lapatinib combined with fractionated radiotherapy may be useful against EGFR+ and HER2+ breast cancers and that inhibition of downstream signaling to ERK1/2 and AKT correlates with sensitization in EGFR+ and HER2+ cells, respectively.


Subject(s)
Breast Neoplasms/radiotherapy , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Radiation Tolerance , Radiation-Sensitizing Agents/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Female , Humans , Lapatinib , Mice , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines , Receptor, ErbB-2/metabolism , Transplantation, Heterologous
11.
Clin Cancer Res ; 16(3): 912-23, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20103665

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) family members (e.g., EGFR, HER2, HER3, and HER4) are commonly overexpressed in pancreatic cancer. We investigated the effects of inhibition of EGFR/HER2 signaling on pancreatic cancer to elucidate the role(s) of EGFR/HER2 in radiosensitization and to provide evidence in support of further clinical investigations. EXPERIMENTAL DESIGN: Expression of EGFR family members in pancreatic cancer lines was assessed by quantitative reverse transcription-PCR. Cell growth inhibition was determined by MTS assay. The effects of inhibition of EGFR family receptors and downstream signaling pathways on in vitro radiosensitivity were evaluated using clonogenic assays. Growth delay was used to evaluate the effects of nelfinavir on in vivo tumor radiosensitivity. RESULTS: Lapatinib inhibited cell growth in four pancreatic cancer cell lines, but radiosensitized only wild-type K-ras-expressing T3M4 cells. Akt activation was blocked in a wild-type K-ras cell line, whereas constitutive phosphorylation of Akt and extracellular signal-regulated kinase (ERK) was seen in lines expressing mutant K-ras. Overexpression of constitutively active K-ras (G12V) abrogated lapatinib-mediated inhibition of both Akt phosphorylation and radiosensitization. Inhibition of MAP/ERK kinase/ERK signaling with U0126 had no effect on radiosensitization, whereas inhibition of activated Akt with LY294002 (enhancement ratio, 1.2-1.8) or nelfinavir (enhancement ratio, 1.2-1.4) radiosensitized cells regardless of K-ras mutation status. Oral nelfinavir administration to mice bearing mutant K-ras-containing Capan-2 xenografts resulted in a greater than additive increase in radiation-mediated tumor growth delay (synergy assessment ratio of 1.5). CONCLUSIONS: Inhibition of EGFR/HER2 enhances radiosensitivity in wild-type K-ras pancreatic cancer. Nelfinavir, and other phosphoinositide 3-kinase/Akt inhibitors, are effective pancreatic radiosensitizers regardless of K-ras mutation status.


Subject(s)
ErbB Receptors/metabolism , Genes, ras , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Lapatinib , Mice , Mice, Inbred BALB C , Mutation , Nelfinavir/pharmacology , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
Radiother Oncol ; 93(3): 639-44, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19853943

ABSTRACT

BACKGROUND AND PURPOSE: We recently showed that lapatinib, an EGFR/HER2 inhibitor, radiosensitized breast cancer cells of the basal and HER2+ subtypes. The purpose of this study was to identify the downstream signaling pathways responsible for lapatinib-mediated radiosensitization in breast cancer. MATERIALS AND METHODS: Response of EGFR downstream signaling pathways was assessed by Western blot and clonogenic cell survival assays in breast tumor cells after irradiation (5Gy), lapatinib, CI-1040, or combined treatment. RESULTS: In SUM102 cells, an EGFR+ basal breast cancer cell line, exposure to ionizing radiation elicited strong activation of ERK1/2 and JNK, which was blocked by lapatinib, and weak/no activation of p38, AKT or STAT3. Direct inhibition of MEK1 with CI-1040 resulted in 95% inhibition of surviving colonies when combined with radiation while inhibition of JNK with SP600125 had no effect. Lapatinib-mediated radiosensitization of SUM102 cells was completely abrogated with expression of constitutively active Raf. Treatment of lapatinib-resistant SUM185 cells with CI-1040 restored radiosensitization with 45% fewer surviving colonies when combined with radiation. CONCLUSIONS: These data suggest that radiosensitization by lapatinib is mediated largely through inhibition of MEK/ERK and that direct inhibition of this pathway may provide an additional avenue of radiosensitization in EGFR+ or HER2+ breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , ErbB Receptors/antagonists & inhibitors , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Quinazolines/pharmacology , Radiation Tolerance/drug effects , Anthracenes/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Lapatinib , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Tumor Cells, Cultured
13.
J Invest Dermatol ; 128(1): 175-87, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17597816

ABSTRACT

Defects in DNA damage responses may underlie genetic instability and malignant progression in melanoma. Cultures of normal human melanocytes (NHMs) and melanoma lines were analyzed to determine whether global patterns of gene expression could predict the efficacy of DNA damage cell cycle checkpoints that arrest growth and suppress genetic instability. NHMs displayed effective G1 and G2 checkpoint responses to ionizing radiation-induced DNA damage. A majority of melanoma cell lines (11/16) displayed significant quantitative defects in one or both checkpoints. Melanomas with B-RAF mutations as a class displayed a significant defect in DNA damage G2 checkpoint function. In contrast the epithelial-like subtype of melanomas with wild-type N-RAS and B-RAF alleles displayed an effective G2 checkpoint but a significant defect in G1 checkpoint function. RNA expression profiling revealed that melanoma lines with defects in the DNA damage G1 checkpoint displayed reduced expression of p53 transcriptional targets, such as CDKN1A and DDB2, and enhanced expression of proliferation-associated genes, such as CDC7 and GEMININ. A Bayesian analysis tool was more accurate than significance analysis of microarrays for predicting checkpoint function using a leave-one-out method. The results suggest that defects in DNA damage checkpoints may be recognized in melanomas through analysis of gene expression.


Subject(s)
DNA Damage , G1 Phase/physiology , G2 Phase/physiology , Gene Expression Profiling , Melanoma/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cells, Cultured , Chromosomal Instability , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Geminin , Humans , Melanocytes/cytology , Melanoma/genetics , Melanoma/metabolism , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Tumor Suppressor Protein p53/physiology
14.
J Clin Invest ; 117(12): 3922-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18008004

ABSTRACT

Immune-mediated antitumor responses occur in patients with metastatic melanoma (MM), and therapies designed to augment such responses are clinically beneficial. Despite the immunogenicity of melanoma, immunomodulatory therapies fail in the majority of patients with MM. An inability of DCs to sufficiently activate effector cells may, in part, underlie this failure of the antitumor response seen in most patients. In this work, we show that mutation of N-RAS or B-RAF, signature genetic lesions present in most MMs, potently induced the expression of cell-surface CD200, a repressor of DC function. Employing 2 independent, genome-wide microarray analyses, we identified CD200 as a highly dynamic, downstream target of RAS/RAF/MEK/ERK activation in melanoma. CD200 protein was similarly overexpressed in human melanoma cell lines and primary tumors. CD200 mRNA expression correlated with progression and was higher in melanoma than in other solid tumors or acute leukemia. Melanoma cell lines expressing endogenous CD200 repressed primary T cell activation by DCs, while knockdown of CD200 by shRNA abrogated this immunosuppressive effect. These data indicate that in addition to its effects on growth, survival, and motility, ERK activation in MM attenuates a host antitumor immune response, implicating CD200 and its interaction with the CD200 receptor as a potential therapeutic target for MM.


Subject(s)
Antigens, CD/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Gene Expression Regulation, Neoplastic/immunology , Melanoma/immunology , Acute Disease , Antigens, CD/genetics , Antigens, Surface/genetics , Antigens, Surface/immunology , Cell Movement/genetics , Cell Movement/immunology , Cell Survival/genetics , Cell Survival/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Leukemia/genetics , Leukemia/immunology , Leukemia/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mutation , Neoplasm Metastasis , Orexin Receptors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
15.
Mol Cancer Ther ; 6(8): 2220-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17699719

ABSTRACT

Mutational activation of Ras and a key downstream effector of Ras, the B-Raf serine/threonine kinase, has been observed in melanomas and colorectal carcinomas. These observations suggest that inhibition of B-Raf activation of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase kinase (MEK) and the extracellular signal-regulated kinase MAPK cascade may be an effective approach for the treatment of RAS and B-RAF mutation-positive melanomas and colon carcinomas. Although recent studies with interfering RNA (RNAi) and pharmacologic inhibitors support a critical role for B-Raf signaling in melanoma growth, whether mutant B-Raf has an equivalent role in promoting colorectal carcinoma growth has not been determined. In the present study, we used both RNAi and pharmacologic approaches to further assess the role of B-Raf activation in the growth of human melanomas and additionally determined if a similar role for mutant B-Raf is seen for colorectal carcinoma cell lines. We observed that RNAi suppression of mutant B-Raf(V600E) expression strongly suppressed the anchorage-dependent growth of B-RAF mutation-positive melanoma, but not colorectal carcinoma, cells. However, the anchorage-independent and tumorigenic growth of B-RAF mutation-positive colorectal carcinomas was dependent on mutant B-Raf function. Finally, pharmacologic inhibition of MEK and Raf was highly effective at inhibiting the growth of B-RAF mutation-positive melanomas and colorectal carcinoma cells, whereas inhibitors of other protein kinases activated by Ras (AKT, c-Jun NH(2)-terminal kinase, and p38 MAPK) were less effective. Our observations suggest that Raf and MEK inhibitors may be effective for the treatment of B-RAF mutation-positive colorectal carcinomas as well as melanomas.


Subject(s)
Colorectal Neoplasms/pathology , Melanoma/pathology , Mutant Proteins/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , Skin Neoplasms/pathology , Animals , Cell Communication/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/enzymology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/enzymology , Mice , Mice, Nude , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Skin Neoplasms/enzymology
16.
Cancer Res ; 67(4): 1502-12, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17308088

ABSTRACT

The majority of human melanomas harbor activating mutations of either N-RAS or its downstream effector B-RAF, which cause activation of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase and the ERK MAPK cascade. The melanoma-relevant effectors of ERK activation, however, are largely unknown. In this work, we show that increased ERK activation correlates strongly with mutational status of N-RAS or B-RAF in 21 melanoma cell lines. Melanoma lines that were wild-type for RAS/RAF showed low levels of ERK activation comparable with primary human melanocytes. Through supervised analysis of RNA expression profiles, we identified 82 genes, including TWIST1, HIF1alpha, and IL-8, which correlated with ERK activation across the panel of cell lines and which decreased with pharmacologic inhibition of ERK activity, suggesting that they are ERK transcriptional targets in melanoma. Additionally, lines lacking mutations of N-RAS and B-RAF were molecularly distinct and characterized by p53 inactivation, reduced ERK activity, and increased expression of epithelial markers. Analysis of primary human melanomas by tissue microarray confirmed a high correlation among expression of these epithelial markers in a heterogeneous sample of 570 primary human tumors, suggesting that a significant frequency of primary melanomas is of this "epithelial-like" subtype. These results show a molecularly distinct melanoma subtype that does not require ERK activation or epithelial-mesenchymal transformation for progression.


Subject(s)
MAP Kinase Signaling System , Melanoma/enzymology , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Epithelial Cells/pathology , Gene Expression Profiling , Humans , Melanocytes/enzymology , Melanoma/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , raf Kinases/genetics , ras Proteins/genetics
17.
Methods Enzymol ; 407: 556-74, 2006.
Article in English | MEDLINE | ID: mdl-16757353

ABSTRACT

Cancer is a multistep genetic process that includes mutational activation of oncogenes and inactivation of tumor suppressor genes. The Ras oncogenes are the most frequently mutated oncogenes in human cancers (30%), with a high frequency associated with cancers of the lung, colon, and pancreas. Mutational activation of Ras is commonly an early event in the development of these cancers. Thus, whether mutated Ras is required for tumor maintenance and what aspects of the complex malignant phenotype might be promoted by mutated Ras are issues that remain unresolved for these and other human cancers. The recent development of interfering RNA to selectively impair expression of mutated Ras provides a powerful approach to begin to resolve these issues. In this chapter, we describe the use of retrovirus-based RNA interference approaches to study the functions of Ras and Ras effectors (Raf, RalA, RalB, and Tiam1) in the growth of pancreatic carcinoma and other human tumor cell lines. Finally, we also compare the use of constitutive and inducible shRNA expression vectors for analyses of mutant Ras function.


Subject(s)
Genes, ras/physiology , Neoplasms/physiopathology , Retroviridae/physiology , ras Proteins/physiology , Base Sequence , Cell Line, Tumor , Genes, ras/drug effects , Humans , Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA Interference , RNA, Small Interfering/physiology , Transfection/methods , raf Kinases/metabolism
18.
J Biol Chem ; 280(24): 23363-70, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15831492

ABSTRACT

The apoptosis-promoting protein Par-4 has been shown to be down-regulated in Ras-transformed NIH 3T3 fibroblasts through the Raf/MEK/ERK MAPK pathway. Because mutations of the ras gene are most often found in tumors of epithelial origin, we explored the signaling pathways utilized by oncogenic Ras to down-regulate Par-4 in RIE-1 and rat ovarian surface epithelial (ROSE) cells. We determined that constitutive activation of the Raf, phosphatidylinositol 3-kinase, or Ral guanine nucleotide exchange factor effector pathway alone was not sufficient to down-regulate Par-4 in RIE-1 or ROSE cells. However, treatment of Ras-transformed RIE-1 or ROSE cells with the MEK inhibitors U0126 and PD98059 increased Par-4 protein expression. Thus, although oncogenic Ras utilizes the Raf/MEK/ERK pathway to down-regulate Par-4 in both fibroblasts and epithelial cells, Ras activation of an additional signaling pathway(s) is required to achieve the same outcome in epithelial cells. Methylation-specific PCR showed that the par-4 promoter is methylated in Ras-transformed cells through a MEK-dependent pathway and that treatment with the DNA methyltransferase inhibitor azadeoxycytidine restored Par-4 mRNA transcript and protein levels, suggesting that the mechanism for Ras-mediated down-regulation of Par-4 is by promoter methylation. Support for this possibility is provided by our observation that Ras transformation was associated with up-regulation of Dnmt1 and Dnmt3 DNA methyltransferase expression. Finally, ectopic Par-4 expression significantly reduced Ras-mediated growth in soft agar, but not morphological transformation, highlighting the importance of Par-4 down-regulation in specific aspects of Ras-mediated transformation of epithelial cells.


Subject(s)
Apoptosis , Azacitidine/analogs & derivatives , DNA Methylation , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction , raf Kinases/physiology , Agar/chemistry , Alleles , Animals , Apoptosis Regulatory Proteins , Azacitidine/pharmacology , Blotting, Northern , Blotting, Western , Butadienes/pharmacology , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA, Complementary/metabolism , Decitabine , Down-Regulation , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/metabolism , Flavonoids/pharmacology , Genetic Vectors , Humans , Mice , Mutation , NIH 3T3 Cells , Nitriles/pharmacology , Ovary/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Polymerase Chain Reaction , RNA/metabolism , RNA, Messenger/metabolism , Rats , Up-Regulation
19.
Oncogene ; 24(21): 3535-40, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15782137

ABSTRACT

The Raf-MEK-ERK protein kinase cascade is a highly conserved signaling pathway that is pivotal in relaying environmental cues from the cell surface to the nucleus. Three Raf isoforms, which share great sequence and structure similarities, have been identified in mammalian cells. We have previously identified Raf kinase inhibitor protein (RKIP) as a negative regulator of the Raf-MEK-ERK signaling pathway by specifically binding to the Raf-1 isoform. We show here that RKIP also antagonizes kinase activity of the B-Raf isoform. Yeast two-hybrid and coimmunoprecipitation experiments indicated that RKIP specifically interacted with B-Raf. Ectopic expression of RKIP antagonized the kinase activity of B-Raf. We showed that the effects of RKIP on B-Raf functions were independent of its known inhibitory action on Raf-1. The expression levels of RKIP in melanoma cancer cell lines are low relative to primary melanocytes. Forced expression of RKIP partially reverted the oncogenic B-Raf kinase-transformed melanoma cancer cell line SK-Mel-28. The low expression of RKIP and its antagonistic action on B-Raf suggests that RKIP may play an important role in melanoma turmorgenesis.


Subject(s)
Androgen-Binding Protein/pharmacology , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/pathology , Proto-Oncogene Proteins B-raf/pharmacology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Down-Regulation , Humans , Melanocytes/physiology , Phosphatidylethanolamine Binding Protein , Proto-Oncogene Proteins B-raf/biosynthesis , Saccharomyces cerevisiae/genetics , Signal Transduction , Tumor Cells, Cultured
20.
J Biol Chem ; 279(32): 33168-76, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15184373

ABSTRACT

Melanoma progresses as a multistep process where the thickness of the lesion and depth of tumor invasion are the best prognostic indicators of clinical outcome. Degradation of the interstitial collagens in the extracellular matrix is an integral component of tumor invasion and metastasis, and much of this degradation is mediated by collagenase-1 (MMP-1), a member of the matrix metalloproteinase (MMP) family. MMP-1 levels increase during melanoma progression where they are associated with shorter disease-free survival. The Ras/Raf/MEK/ERK mitogen-activated protein kinase (MAPK) pathway is a major regulator of melanoma cell proliferation. Recently, BRAF has been identified as a common site of activating mutations, and, although many reports focus on its growth-promoting effects, this pathway has also been implicated in progression toward metastatic disease. In this study, we describe four melanoma cell lines that produce high levels of MMP-1 constitutively. In each cell line the Ras/Raf/MEK/ERK pathway is constitutively active and is the dominant pathway driving the production of MMP-1. Activation of this pathway arises due to either an activating mutation in BRAF (three cell lines) or autocrine fibroblast growth factor signaling (one cell line). Furthermore, blocking MEK/ERK activity inhibits melanoma cell proliferation and abrogates collagen degradation, thus decreasing their metastatic potential. Importantly, this inhibition of invasive behavior can occur in the absence of any detectable changes in cell proliferation and survival. Thus, constitutive activation of this MAPK pathway not only promotes the increased proliferation of melanoma cells but is also important for the acquisition of an invasive phenotype.


Subject(s)
Gene Expression , Matrix Metalloproteinase 1/genetics , Melanoma/enzymology , Mitogen-Activated Protein Kinases/metabolism , Arsenites/pharmacology , Disease-Free Survival , Enzyme Activation/drug effects , Fibroblast Growth Factors/metabolism , Humans , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Matrix Metalloproteinase 1/analysis , Matrix Metalloproteinase 1/metabolism , Melanoma/genetics , Melanoma/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Neoplasm Invasiveness , Phosphorylation , Prognosis , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-raf/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sodium Compounds/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...