Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 7113, 2022 11 19.
Article in English | MEDLINE | ID: mdl-36402789

ABSTRACT

NRAS-mutated melanoma lacks a specific line of treatment. Metabolic reprogramming is considered a novel target to control cancer; however, NRAS-oncogene contribution to this cancer hallmark is mostly unknown. Here, we show that NRASQ61-mutated melanomas specific metabolic settings mediate cell sensitivity to sorafenib upon metabolic stress. Mechanistically, these cells are dependent on glucose metabolism, in which glucose deprivation promotes a switch from CRAF to BRAF signaling. This scenario contributes to cell survival and sustains glucose metabolism through BRAF-mediated phosphorylation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-2/3 (PFKFB2/PFKFB3). In turn, this favors the allosteric activation of phosphofructokinase-1 (PFK1), generating a feedback loop that couples glycolytic flux and the RAS signaling pathway. An in vivo treatment of NRASQ61 mutant melanomas, including patient-derived xenografts, with 2-deoxy-D-glucose (2-DG) and sorafenib effectively inhibits tumor growth. Thus, we provide evidence for NRAS-oncogene contributions to metabolic rewiring and a proof-of-principle for the treatment of NRASQ61-mutated melanoma combining metabolic stress (glycolysis inhibitors) and previously approved drugs, such as sorafenib.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Sorafenib/pharmacology , Cell Line, Tumor , Mutation , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Glycolysis/genetics , Glucose/metabolism , Stress, Physiological , Phosphofructokinase-2/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism
2.
Commun Biol ; 3(1): 366, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32647375

ABSTRACT

Elucidating the contribution of somatic mutations to cancer is essential for personalized medicine. STK11 (LKB1) appears to be inactivated in human cancer. However, somatic missense mutations also occur, and the role/s of these alterations to this disease remain unknown. Here, we investigated the contribution of four missense LKB1 somatic mutations in tumor biology. Three out of the four mutants lost their tumor suppressor capabilities and showed deficient kinase activity. The remaining mutant retained the enzymatic activity of wild type LKB1, but induced increased cell motility. Mechanistically, LKB1 mutants resulted in differential gene expression of genes encoding vesicle trafficking regulating molecules, adhesion molecules and cytokines. The differentially regulated genes correlated with protein networks identified through comparative secretome analysis. Notably, three mutant isoforms promoted tumor growth, and one induced inflammation-like features together with dysregulated levels of cytokines. These findings uncover oncogenic roles of LKB1 somatic mutations, and will aid in further understanding their contributions to cancer development and progression.


Subject(s)
Biomarkers, Tumor/genetics , Cell Movement , Inflammation/pathology , Lung Neoplasms/pathology , Melanoma/pathology , Mutation, Missense , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Cell Cycle , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Nude , Phosphorylation , Protein Isoforms , Protein Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
J Clin Invest ; 128(9): 3887-3905, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29944140

ABSTRACT

Dormant or slow-cycling tumor cells can form a residual chemoresistant reservoir responsible for relapse in patients, years after curative surgery and adjuvant therapy. We have adapted the pulse-chase expression of H2BeGFP for labeling and isolating slow-cycling cancer cells (SCCCs). SCCCs showed cancer initiation potential and enhanced chemoresistance. Cells at this slow-cycling status presented a distinctive nongenetic and cell-autonomous gene expression profile shared across different tumor types. We identified TET2 epigenetic enzyme as a key factor controlling SCCC numbers, survival, and tumor recurrence. 5-Hydroxymethylcytosine (5hmC), generated by TET2 enzymatic activity, labeled the SCCC genome in carcinomas and was a predictive biomarker of relapse and survival in cancer patients. We have shown the enhanced chemoresistance of SCCCs and revealed 5hmC as a biomarker for their clinical identification and TET2 as a potential drug target for SCCC elimination that could extend patients' survival.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Cycle , Cell Line, Tumor , Cell Survival/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Dioxygenases , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasms/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Recurrence , Xenograft Model Antitumor Assays
4.
Oncotarget ; 7(19): 28086-95, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27057633

ABSTRACT

Melanomas arising in association with a common or cellular blue nevus (MABN) comprise a relatively rare and heterogeneous group of lethal melanomas. Although GNAQ is known to be frequently mutated in common blue nevus, cellular blue nevus (CBN) and MABN and these malignant lesions present gross chromosome alterations harboring BAP1 mutations, little is known about other mutations that contribute to the development and progression of these neoplasms. Thus, the genetic profile of these tumors is important to increase the number of intervention and treatment modalities. Here, we characterized and genetically profiled two different sections of a rare MABN and two CBNs from three different patients. All of the samples harbored a GNAQ mutation, exhibited RAS pathway activation, and harbored additional mutations in genes associated with genomic instability and epigenetic regulation (KMT2C, FANCD2, ATR, ATRX, NBN, ERCC2, SETD2, and WHSC1). In addition, all neoplasms harbored mutations that directly or indirectly affected either the regulation or activation of the PI3K pathway (PIK3CA, NF1, INPP5B and GSK3B). Our results not only help understand the genetic complexity of these blue melanocytic lesions but provide a rationale to use the combination of PI3K/MTOR and MEK1/2 inhibitors against these types of tumors.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Nevus, Blue/genetics , Phosphatidylinositol 3-Kinases/genetics , Skin Neoplasms/genetics , DNA Mutational Analysis , Genomic Instability , Humans , Mutation , Nevus, Blue/pathology , Skin Neoplasms/pathology
5.
Pigment Cell Melanoma Res ; 29(2): 247-53, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26701415

ABSTRACT

Melanoma presents molecular alterations based on its anatomical location and exposure to environmental factors. Due to its intrinsic genetic heterogeneity, a simple snapshot of a tumor's genetic alterations does not reflect the tumor clonal complexity or specific gene-gene cooperation. Here, we studied the genetic alterations and clonal evolution of a unique patient with a Nevus of Ota that developed into a recurring uveal-like dermal melanoma. The Nevus of Ota and ulterior lesions contained GNAQ mutations were c-KIT positive, and tumors showed an increased RAS pathway activity during progression. Whole-exome sequencing of these lesions revealed the acquisition of BAP1 and TP53 mutations during tumor evolution, thereby unmasking clonal heterogeneity and allowing the identification of cooperating genes within the same tumor. Our results highlight the importance of studying tumor genetic evolution to identify cooperating mechanisms and delineate effective therapies.


Subject(s)
Head and Neck Neoplasms/genetics , Nevus of Ota/genetics , Skin Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Adult , Female , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Head and Neck Neoplasms/pathology , Humans , Nevus of Ota/pathology , Skin Neoplasms/pathology
6.
PLoS One ; 8(6): e66933, 2013.
Article in English | MEDLINE | ID: mdl-23825589

ABSTRACT

Aberrant activation of MAP kinase signaling pathway and loss of tumor suppressor LKB1 have been implicated in lung cancer development and progression. Although oncogenic KRAS mutations are frequent, BRAF mutations (BRAF(V600E)) are found in 3% of human non-small cell lung cancers. Contrary to KRAS mutant tumors, BRAF(V600E)-induced tumors are benign adenomas that fail to progess. Interestingly, loss of tumor supressor LKB1 coexists with KRAS oncogenic mutations and synergizes in tumor formation and progression, however, its cooperation with BRAF(V600E) oncogene is unknown. Our results describe a lung cell population in neonates mice where expression of BRAF(V600E) leads to lung adenoma development. Importantly, expression of BRAF(V600E) concomitant with the loss of only a single-copy of Lkb1, overcomes senencence-like features of BRAF(V600E)-mutant adenomas leading malignization to carcinomas. These results posit LKB1 haploinsufficiency as a risk factor for tumor progression of BRAF(V600E) mutated lung adenomas in human cancer patients.


Subject(s)
Adenoma/pathology , Disease Progression , Gene Deletion , Lung Neoplasms/pathology , Mutation , Protein Serine-Threonine Kinases/deficiency , Proto-Oncogene Proteins B-raf/genetics , AMP-Activated Protein Kinases , Adenoma/enzymology , Adenoma/genetics , Animals , Animals, Newborn , Cadherins/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Mice , Mice, Transgenic , Oncogenes/genetics , Protein Serine-Threonine Kinases/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tumor Suppressor Protein p53/metabolism
7.
Dig Dis ; 30(5): 524-31, 2012.
Article in English | MEDLINE | ID: mdl-23108309

ABSTRACT

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is a chemoresistant tumor strongly associated with chronic hepatitis. Identification of molecular links connecting inflammation with cell growth/survival, and characterization of pro-tumorigenic intracellular pathways is therefore of therapeutic interest. The epidermal growth factor receptor (EGFR) signaling system stands at a crossroad between inflammatory signals and intracellular pathways associated with hepatocarcinogenesis. We investigated the regulation and activity of different components of the EGFR system, including the EGFR ligand amphiregulin (AR) and its sheddase ADAM17, and the modulation of intracellular EGFR signaling by a novel mechanism involving protein methylation. METHODS: ADAM17 protein expression was examined in models of liver injury and carcinogenesis. Crosstalk between tumor necrosis factor (TNF)-α, AR and EGFR signaling was evaluated in human HCC cells and mouse hepatocytes. Modulation of EGFR signaling and biological responses by methylation reactions was evaluated in AML12 mouse hepatocytes. RESULTS: ADAM17 was upregulated in liver injury and hepatocarcinogenesis. TNF-α triggered AR shedding and EGFR transactivation in HCC cells. AR was necessary for TNF-α activation of ERK1/2 and Akt signaling in hepatocytes. Inhibition of methylation reactions increased the ERK1/2 signal amplitude triggered by AR/EGFR and reduced DNA synthesis in AML12 cells. CONCLUSIONS: Increased ADAM17 in pre-neoplastic liver injury further supports its implication in hepatocarcinogenesis. AR release and EGFR transactivation by TNF-α constitutes a novel link between inflammatory signals and pro-tumorigenic mechanisms in liver cells. Finally, the identification of a new mechanism controlling growth factor signaling, and biological responses, involving methylation reactions within the RAS/RAF/MEK/ERK pathway, exposes a new target for antineoplastic intervention.


Subject(s)
Carcinoma, Hepatocellular/metabolism , ErbB Receptors/metabolism , Inflammation/metabolism , Liver Neoplasms/metabolism , Signal Transduction/physiology , ADAM Proteins/metabolism , ADAM17 Protein , Amphiregulin , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , EGF Family of Proteins , Gene Expression Regulation/physiology , Glycoproteins/metabolism , Hepatocytes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Liver/injuries , Liver/metabolism , MAP Kinase Signaling System/physiology , Methylation , Mice , Proto-Oncogene Proteins c-akt , Tumor Necrosis Factor-alpha/metabolism
8.
Sci Signal ; 4(190): ra58, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21917714

ABSTRACT

The RAS to extracellular signal-regulated kinase (ERK) signal transduction cascade is crucial to cell proliferation, differentiation, and survival. Although numerous growth factors activate the RAS-ERK pathway, they can have different effects on the amplitude and duration of the ERK signal and, therefore, on the biological consequences. For instance, nerve growth factor, which elicits a larger and more sustained increase in ERK phosphorylation in PC12 cells than does epidermal growth factor (EGF), stimulates PC12 cell differentiation, whereas EGF stimulates PC12 cell proliferation. Here, we show that protein arginine methylation limits the ERK1/2 signal elicited by particular growth factors in different cell types from various species. We found that this restriction in ERK1/2 phosphorylation depended on methylation of RAF proteins by protein arginine methyltransferase 5 (PRMT5). PRMT5-dependent methylation enhanced the degradation of activated CRAF and BRAF, thereby reducing their catalytic activity. Inhibition of PRMT5 activity or expression of RAF mutants that could not be methylated not only affected the amplitude and duration of ERK phosphorylation in response to growth factors but also redirected the response of PC12 cells to EGF from proliferation to differentiation. This additional level of regulation within the RAS pathway may lead to the identification of new targets for therapeutic intervention.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Methyltransferases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Animals , COS Cells , Cell Differentiation/drug effects , Chlorocebus aethiops , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Methyltransferases/genetics , Protein-Arginine N-Methyltransferases , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/genetics , Rats
9.
J Transl Med ; 9: 76, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21609436

ABSTRACT

BACKGROUND: A molecular linkage between the MAPK and the LKB1-AMPK energy sensor pathways suggests that combined MAPK oncogene inhibition and metabolic modulation of AMPK would be more effective than either manipulation alone in melanoma cell lines. MATERIALS AND METHODS: The combination of the BRAF inhibitor vemurafenib (formerly PLX4032) and metformin were tested against a panel of human melanoma cell lines with defined BRAF and NRAS mutations for effects on viability, cell cycle and apoptosis. Signaling molecules in the MAPK, PI3K-AKT and LKB1-AMPK pathways were studied by Western blot. RESULTS: Single agent metformin inhibited proliferation in 12 out of 19 cell lines irrespective of the BRAF mutation status, but in one NRASQ61K mutant cell line it powerfully stimulated cell growth. Synergistic anti-proliferative effects of the combination of metformin with vemurafenib were observed in 6 out of 11 BRAFV600E mutants, including highly synergistic effects in two BRAFV600E mutant melanoma cell lines. Antagonistic effects were noted in some cell lines, in particular in BRAFV600E mutant cell lines resistant to single agent vemurafenib. Seven out of 8 BRAF wild type cell lines showed marginally synergistic anti-proliferative effects with the combination, and one cell line had highly antagonistic effects with the combination. The differential effects were not dependent on the sensitivity to each drug alone, effects on cell cycle or signaling pathways. CONCLUSIONS: The combination of vemurafenib and metformin tended to have stronger anti-proliferative effects on BRAFV600E mutant cell lines. However, determinants of vemurafenib and metformin synergism or antagonism need to be understood with greater detail before any potential clinical utility of this combination.


Subject(s)
Indoles/therapeutic use , Melanoma/drug therapy , Melanoma/genetics , Metformin/therapeutic use , Mutation/genetics , Sulfonamides/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media/pharmacology , DNA Mutational Analysis , Drug Synergism , Flow Cytometry , Glucose/pharmacology , Humans , Indoles/pharmacology , Metformin/pharmacology , Phosphorylation/drug effects , Phosphothreonine/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides/pharmacology , Time Factors , Vemurafenib
10.
BMC Cancer ; 10: 265, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20529342

ABSTRACT

BACKGROUND: Melanoma is the most deadly form of skin cancer without effective treatment. Methylthioadenosine (MTA) is a naturally occurring nucleoside with differential effects on normal and transformed cells. MTA has been widely demonstrated to promote anti-proliferative and pro-apoptotic responses in different cell types. In this study we have assessed the therapeutic potential of MTA in melanoma treatment. METHODS: To investigate the therapeutic potential of MTA we performed in vitro proliferation and viability assays using six different mouse and human melanoma cell lines wild type for RAS and BRAF or harboring different mutations in RAS pathway. We also have tested its therapeutic capabilities in vivo in a xenograft mouse melanoma model and using variety of molecular techniques and tissue culture we investigated its anti-proliferative and pro-apoptotic properties. RESULTS: In vitro experiments showed that MTA treatment inhibited melanoma cell proliferation and viability in a dose dependent manner, where BRAF mutant melanoma cell lines appear to be more sensitive. Importantly, MTA was effective inhibiting in vivo tumor growth. The molecular analysis of tumor samples and in vitro experiments indicated that MTA induces cytostatic rather than pro-apoptotic effects inhibiting the phosphorylation of Akt and S6 ribosomal protein and inducing the down-regulation of cyclin D1. CONCLUSIONS: MTA inhibits melanoma cell proliferation and in vivo tumor growth particularly in BRAF mutant melanoma cells. These data reveal a naturally occurring drug potentially useful for melanoma treatment.


Subject(s)
Adenosine/pharmacology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Thionucleosides/pharmacology , Adenosine/analogs & derivatives , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D1/metabolism , Dose-Response Relationship, Drug , Genes, ras , Humans , Male , Melanoma/genetics , Melanoma/pathology , Mice , Mutation , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Time Factors , Tumor Burden , Xenograft Model Antitumor Assays
11.
Int J Cancer ; 126(7): 1549-61, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-19810100

ABSTRACT

Melanoma is the most lethal human skin cancer. If metastatic, it becomes very aggressive and resistant to standard modalities of anticancer treatment. During the last 10 years, several therapeutic strategies have been tested including the use of single and combined small drugs. Experimental results indicate that RAS and PI3K pathways are important for the development and maintenance of melanoma. In this study, we assessed the in vitro and in vivo inhibition potential of PI-103, a PI3K (p110alpha)/mTOR inhibitor and sorafenib, a BRAF inhibitor, as single agents and in combination in primary melanoma cell lines. Although PI-103 and sorafenib inhibited melanoma in vitro cell proliferation and viability, the inhibition of RAS pathway appeared to be more effective. The combination of the two agents in in vitro showed a synergistic effect inhibiting RAS and PI3K pathways in a cell line dependent manner. However, no cooperative effect was observed in blocking in vivo tumor growth in immunocompetent mice. In contrary to the expected, the data indicate that PI-103 induced immunosuppression promoting in vivo tumor growth and inhibiting apoptosis. Furthermore, in vitro studies examining the effects of the PI3K/mTOR inhibitor in tumor derived cell lines indicated that PI-103 induced the anti-apoptotic BH3 family proteins Mcl1, Bcl2 and Bcl(xL) favoring, the in vitro survival of sorafenib treated melanoma cells. These data certainly makes an argument for investigating unexpected effects of rational drug combinations on immunocompetent animal models prior to conducting clinical studies.


Subject(s)
Benzenesulfonates/pharmacology , Furans/pharmacology , Immunosuppression Therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Melanoma/pathology , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Blotting, Western , Cell Proliferation , Colony-Forming Units Assay , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Intercellular Signaling Peptides and Proteins/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Melanoma/immunology , Melanoma/mortality , Mice , Mice, Inbred BALB C , Mice, Nude , Niacinamide/analogs & derivatives , Phenylurea Compounds , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sorafenib , Survival Rate , TOR Serine-Threonine Kinases , Tumor Cells, Cultured , ras Proteins/genetics , ras Proteins/metabolism
12.
PLoS One ; 5(12): e15690, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21209952

ABSTRACT

BACKGROUND: Inflammation and fibrogenesis are directly related to chronic liver disease progression, including hepatocellular carcinoma (HCC) development. Currently there are few therapeutic options available to inhibit liver fibrosis. We have evaluated the hepatoprotective and anti-fibrotic potential of orally-administered 5'-methylthioadenosine (MTA) in Mdr2(-/-) mice, a clinically relevant model of sclerosing cholangitis and spontaneous biliary fibrosis, followed at later stages by HCC development. METHODOLOGY: MTA was administered daily by gavage to wild type and Mdr2(-/-) mice for three weeks. MTA anti-inflammatory and anti-fibrotic effects and potential mechanisms of action were examined in the liver of Mdr2(-/-) mice with ongoing fibrogenesis and in cultured liver fibrogenic cells (myofibroblasts). PRINCIPAL FINDINGS: MTA treatment reduced hepatomegaly and liver injury. α-Smooth muscle actin immunoreactivity and collagen deposition were also significantly decreased. Inflammatory infiltrate, the expression of the cytokines IL6 and Mcp-1, pro-fibrogenic factors like TGFß2 and tenascin-C, as well as pro-fibrogenic intracellular signalling pathways were reduced by MTA in vivo. MTA inhibited the activation and proliferation of isolated myofibroblasts and down-regulated cyclin D1 gene expression at the transcriptional level. The expression of JunD, a key transcription factor in liver fibrogenesis, was also reduced by MTA in activated myofibroblasts. CONCLUSIONS/SIGNIFICANCE: Oral MTA administration was well tolerated and proved its efficacy in reducing liver inflammation and fibrosis. MTA may have multiple molecular and cellular targets. These include the inhibition of inflammatory and pro-fibrogenic cytokines, as well as the attenuation of myofibroblast activation and proliferation. Downregulation of JunD and cyclin D1 expression in myofibroblasts may be important regarding the mechanism of action of MTA. This compound could be a good candidate to be tested for the treatment of (biliary) liver fibrosis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Adenosine/analogs & derivatives , Fibrosis/drug therapy , Liver Diseases/genetics , Liver Diseases/pathology , Thionucleosides/administration & dosage , Adenosine/administration & dosage , Animals , Cyclin D1/metabolism , Cytokines/metabolism , Disease Progression , Fibroblasts/metabolism , Inflammation , Liver/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-jun/metabolism , ATP-Binding Cassette Sub-Family B Member 4
13.
J Biol Chem ; 283(19): 13450-8, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18316372

ABSTRACT

In response to oncogenic signals, cells have developed safe mechanisms to avoid transformation through activation of a senescence program. Upon v-H-Ras overexpression, normal cells undergo senescence through several cellular processes, including activation of the ERK1/2 pathway. Interestingly, the E1a gene from adenovirus 5 has been shown to rescue cells from senescence by a yet unknown mechanism. We investigated whether E1a was able to interfere with the ERK1/2 signaling pathway to rescue cells from v-H-Ras-mediated senescence. Our results show that, E1a overexpression blocks v-H-Ras-mediated ERK1/2 activation by two different and concomitant mechanisms. E1a through its ability to interfere with PKB/Akt activation induces the down-regulation of the PEA15 protein, an ERK1/2 nuclear export factor, leading to nuclear accumulation of ERK1/2. In addition to this, we show that E1a increases the expression of the inducible ERK1/2 nuclear phosphatases (MAPK phosphatases) MKP1/DUSP1 and DUSP5, which leads to ERK1/2 dephosphorylation. We confirmed our observations in the human normal diploid fibroblasts IMR90, in which we could also show that an E1a mutant, unable to bind retinoblastoma protein (pRb), cannot rescue cells from v-H-Ras-induced senescence. In conclusion, E1a is able to rescue from Ras-induced senescence by affecting ERK1/2 localization and phosphorylation.


Subject(s)
Adenovirus E1A Proteins/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Oncogene Protein p21(ras)/metabolism , Up-Regulation , Active Transport, Cell Nucleus , Adenovirus E1A Proteins/genetics , Animals , Cell Line , Cell Nucleus/enzymology , Cellular Senescence/physiology , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Enzyme Activation , Gene Expression Regulation , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Oncogene Protein p21(ras)/genetics , Protein Binding , Retinoblastoma Protein/metabolism , Serum
14.
Cancer Res ; 63(7): 1576-82, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12670907

ABSTRACT

The hepatocyte growth factor/scatter factor (HGF/SF) receptor c-Met and variants of the CD44 family of surface adhesion molecules, including CD44v6, have been implicated in cancer progression and metastasis. CD44 isoforms bearing heparin sulfate chains can bind to HGF/SF and facilitate its presentation to c-Met. Here, we demonstrate that HGF/SF-Met binding up-regulates the expression of CD44v6 in murine melanoma cells, serving to compensate for loss by internalization. c-Met-mediated CD44v6 up-regulation was achieved through transcriptional activation of the immediate early gene egr-1. Enhanced egr-1 expression was apparent at the level of RNA 40 min after exposure to HGF/SF, and Egr-1 protein was detectable between 1 and 2 h post-treatment. CD44v6 RNA levels were correspondingly elevated 2 h after HGF/SF exposure. HGF/SF induced egr-1 activation via the Ras>Erk1/2 pathway but not through either phosphatidylinositol 3'-kinase or protein kinase C. Binding of NK2, a naturally occurring splice variant of HGF/SF, to c-Met failed to induce either Egr-1 or CD44v6, accounting at least in part for its antagonistic behavior. We also identified an Egr-1-binding site in the mouse CD44 gene promoter that accounts for its responsiveness to HGF/SF in melanoma cells. The compensatory up-regulation of both c-Met and CD44v6 in response to HGF/SF has important implications with respect to strategies used by cancer cells to sustain stimulation of growth- and metastasis-promoting pathways associated with tumor progression.


Subject(s)
DNA-Binding Proteins/physiology , Glycoproteins/biosynthesis , Hepatocyte Growth Factor/physiology , Hyaluronan Receptors/biosynthesis , Immediate-Early Proteins , Melanoma, Experimental/immunology , Transcription Factors/physiology , Animals , Antigen Presentation , Base Sequence , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Feedback/physiology , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Glycoproteins/immunology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , MAP Kinase Signaling System/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , Protein Structure, Tertiary , Proto-Oncogene Proteins c-met/immunology , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcriptional Activation , Up-Regulation
15.
Cancer Res ; 62(22): 6724-30, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12438273

ABSTRACT

Cutaneous malignant melanoma (CMM), already known for its highly aggressive behavior and resistance to conventional therapy, has evolved into a health crisis by virtue of a dramatic elevation in incidence. The underlying genetic basis for CMM, as well as the fundamental role for UV radiation in its etiology, is now widely accepted. However, the only bona fide genetic locus to emerge from extensive analysis of CMM suppressor candidates is INK4a/ARF at 9p21, which is lost frequently in familial and occasionally in somatic CMM. The functional relationship between INK4a/ARF and UV radiation in the pathogenesis of CMM is largely unknown. Recently, we reported that hepatocyte growth factor/scatter factor (HGF/SF)-transgenic mice develop melanomas after a single erythemal dose of neonatal UV radiation, supporting epidemiological data implicating childhood sunburn in CMM. Here we show that neonatal UV irradiation induces a full spectrum of melanocyte pathology from early premalignant lesions through distant metastases. Cutaneous melanomas arise with histopathological and molecular pathogenetic features remarkably similar to CMM, including loss of ink4a/arf. A role for ink4a/arf in UV-induced melanomagenesis was directly assessed by placing the HGF/SF transgene on a genetic background devoid of ink4a/arf. Median time to melanoma development induced by UV radiation was only 50 days in HGF/SF ink4a/arf(-/-) mice, compared with 152 and 238 days in HGF/SF ink4a/arf(+/-) and HGF/SF ink4a/arf(+/+) mice, respectively. These studies provide experimental evidence that ink4a/arf plays a critical role in UV-induced melanomagenesis and strongly suggest that sunburn is a highly significant risk factor, particularly in families harboring germ-line mutations in INK4a/ARF.


Subject(s)
Cocarcinogenesis , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Melanoma, Experimental/etiology , Ultraviolet Rays/adverse effects , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Models, Animal , Hepatocyte Growth Factor/genetics , Humans , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
Nat Med ; 8(11): 1276-80, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12368906

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma in children, yet molecular events associated with the genesis and progression of this potentially fatal disease are largely unknown. For the molecules and pathways that have been implicated, genetic validation has been impeded by lack of a mouse model of RMS. Here we show that simultaneous loss of Ink4a/Arf function and disruption of c-Met signaling in Ink4a/Arf(-/-) mice transgenic for hepatocyte growth factor/scatter factor (HGF/SF) induces RMS with extremely high penetrance and short latency. In cultured myoblasts, c-Met activation and Ink4a/Arf loss suppress myogenesis in an additive fashion. Our data indicate that human c-MET and INK4a/ARF, situated at the nexus of pathways regulating myogenic growth and differentiation, represent critical targets in RMS pathogenesis. The marked synergism in mice between aberrant c-Met signaling and Ink4a/Arf inactivation, lesions individually implicated in human RMS, suggests a therapeutic combination to combat this devastating childhood cancer.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Hepatocyte Growth Factor/metabolism , Rhabdomyosarcoma/metabolism , Signal Transduction , Soft Tissue Neoplasms/metabolism , Tumor Suppressor Protein p14ARF/genetics , Animals , Cells, Cultured , Humans , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-met/metabolism , Rhabdomyosarcoma/genetics , Soft Tissue Neoplasms/genetics
17.
Cancer Res ; 62(9): 2690-4, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11980669

ABSTRACT

Promotion of cellular resistance to stressful stimuli, including ionizing radiation and chemotherapeutic drugs, contributes to the transforming activity of the PCPH oncogene. The mechanism of this action, however, has remained unknown. Consistent with its intrinsic ATP diphosphohydrolase activity, expression of the PCPH oncoprotein in cultured cells has now been shown to result in partial depletion of intracellular ATP and consequent inhibition of the c-JUN NH2-terminal kinase-mediated stress signaling pathway. Supplementation of cells expressing the PCPH oncoprotein with exogenous ATP restored both stress-response signaling and sensitivity to cisplatin-induced apoptosis. In contrast, overexpression of the wild-type PCPH protein had a minimal effect on stress-induced signaling and on the cellular ATP content and did not protect cells from apoptosis. These results suggest that the PCPH oncoprotein confers resistance to stressors by reducing the cellular ATP concentration to levels below those required for optimal stress-induced signaling and apoptosis. Treatment with adenosine or nucleoside analogues may thus enhance the response to radiation or chemotherapy of tumors that express the PCPH oncogene.


Subject(s)
Adenosine Triphosphate/deficiency , Fungal Proteins , MAP Kinase Kinase Kinase 1 , Oncogene Proteins/physiology , 3T3 Cells , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Survival/physiology , DNA-Binding Proteins/antagonists & inhibitors , Enzyme Activation , GATA Transcription Factors , MAP Kinase Signaling System , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Stress, Physiological/enzymology , Stress, Physiological/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcriptional Activation/physiology , Transfection
18.
Oncogene ; 21(7): 1000-8, 2002 Feb 07.
Article in English | MEDLINE | ID: mdl-11850817

ABSTRACT

Members of the mitogen-activated protein kinase (MAPK) superfamily, including p38 kinase and SAPK/JNK, play a central role in mediating cellular response to environmental stress, growth factors and cytokines. Hepatocyte growth factor/scatter factor (HGF/SF) is a multifunctional cytokine capable of eliciting mitogenic, motogenic and morphogenetic activities in responsive cells, and has been implicated in tumor development and metastasis. Binding of HGF/SF to its tyrosine kinase receptor c-Met stimulates multiple signal transduction pathways, leading to the activation of numerous transcription factors. We here report that HGF/SF can induce cyclin D1 expression in mouse melanoma cells, and that this up-regulation is mediated in part by the activating transcription factor-2 (ATF-2). HGF/SF-mediated phosphorylation of ATF-2 was reduced in the presence of either the p38 kinase-specific inhibitor SB203580, a dominant negative p38 mutant, the SAPK/JNK inhibitor JNK-interacting protein-1 (JIP-1), or the phosphatidylinositol 3-kinase (PI3K)-specific inhibitor LY294002. Activation of p38 kinase by HGF/SF was partially blocked by the PI3K-specific inhibitor as well. The upstream kinases for p38, MKK3/6, did not become activated following HGF/SF exposure, and ATF-2 activation was undiminished by transient transfection of a dominant negative MKK6 mutant. However, transcriptional up-regulation of cyclin D1 by HGF/SF was partially inhibited by the p38 kinase-specific inhibitor, and cyclin D1 protein induction was partially blocked by a dominant negative ATF-2 mutant. Notably, the p38 kinase-specific inhibitor was able to block melanoma cell proliferation but not motility. We conclude that the ATF-2 transcription factor becomes activated by HGF/SF through p38 MAPK and SAPK/JNK. Moreover, the p38-ATF-2 pathway can help mediate proliferation signals in tumor cells through transcriptional activation of key cell cycle regulators.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Cyclin D1/physiology , Hepatocyte Growth Factor/pharmacology , Melanoma/metabolism , Mitogen-Activated Protein Kinases/physiology , Transcription Factors/physiology , Activating Transcription Factor 2 , Animals , Cell Division , Cell Movement , Cyclin D1/genetics , Kinetics , Melanoma/genetics , Melanoma/pathology , Mice , Phosphatidylinositol 3-Kinases/physiology , RNA, Messenger/biosynthesis , Signal Transduction , Transcriptional Activation , Tumor Cells, Cultured , Up-Regulation , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...