Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Dermatol ; 18(7): 611-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19320734

ABSTRACT

Please cite this paper as: Aldo-keto reductase 1C subfamily genes in skin are UV-inducible: possible role in keratinocytes survival. Experimental Dermatology 2009; 18: 611-618.Abstract: Human skin is endowed with the capacity to synthesize and metabolize steroid hormones, a function of importance in skin physiology and pathology. It is the hormone-regulatory enzymes, including the aldo-keto reductase 1C subfamily (AKR1Cs) that are largely responsible for the local levels of active steroid hormones. AKR1C1 and AKR1C2 inactivate progesterone and 5alpha-dihydrotestosterone, respectively, whereas AKR1C3 activates oestradiol and testosterone. Here, we show that AKR1C1-3 are expressed in keratinocytes and fibroblasts, with marginal expression in melanocytes. In human primary keratinocytes, AKR1C1 and -2 were UVB-inducible in a dose-dependent manner, as shown by quantitative PCR and Western blot analyses. The induction of AKR1C1 by UVB was concomitant with the presence of an apoptotic marker, the cleavage product of poly-ADP ribose polymerase. Similarly, the activation of AKR1C1 and -2 upon UVB exposure was demonstrated in swine skin in vivo and in human skin explants. As expected, hydrogen peroxide-derived reactive oxygen species also induced AKR1C1 and -2 mRNA and protein levels in keratinocytes in a dose-dependent manner. Furthermore, down-regulation of AKR1Cs by small interfering ribonucleic acid led to significantly reduced cell viability. Based on the combined evidence of the presence of an apoptotic marker in the UVB-exposed keratinocytes with increased AKR1Cs expression and reduced cell viability in down-regulated AKR1Cs, we suggest that AKR1C subfamily genes are stress-inducible and might function as survival factors in keratinocytes.


Subject(s)
20-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Hydroxyprostaglandin Dehydrogenases/metabolism , Hydroxysteroid Dehydrogenases/metabolism , Keratinocytes/cytology , Skin/metabolism , Skin/radiation effects , Ultraviolet Rays , 20-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , Aldo-Keto Reductase Family 1 Member C3 , Animals , Apoptosis/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Gene Expression Regulation/radiation effects , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/genetics , Keratinocytes/metabolism , Keratinocytes/radiation effects , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Skin/cytology , Swine
2.
Melanoma Res ; 17(5): 274-83, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17885582

ABSTRACT

Melanoma, the most deadly form of skin cancer, is very aggressive and resistant to present therapies. The transcription factor nuclear factor-kappa B (NF-kappaB) has been reported to be constitutively active in many types of cancer. Constitutively active NF-kappaB seen in melanoma likely plays a central role in cell survival and growth. We have established and characterized novel cell lines from our murine melanoma model. Here we report the constitutive activity of NF-kappaB in these melanoma-derived cells, as shown by electrophoretic mobility shift assay and reporter assays. We hypothesized that agents that inhibit NF-kappaB may also inhibit cell proliferation and may induce apoptosis in such melanoma cells. Curcumin has been shown to inhibit NF-kappaB activity in several cell types. In our system, curcumin selectively inhibited growth of melanoma cells, but not normal melanocytes. Curcumin induced melanoma cells to undergo apoptosis, as shown by caspase-3 activation, inversion of membrane phosphatidyl serine, and increases in cells in the sub-G1 phase. A curcumin dose-dependent inhibition of NF-kappaB-driven reporter activity correlated with decreased levels of phospho-IkappaBalpha, and decreased expression of NF-kappaB-target genes COX-2 and cyclin D1. This study demonstrates that the use of cells from our model system can facilitate studies of signaling pathways in melanoma. We furthermore conclude that curcumin, a natural and safe compound, inhibits NF-kappaB activity and the expression of its downstream target genes, and also selectively induces apoptosis of melanoma cells but not normal melanocytes. These encouraging in-vitro results support further investigation of curcumin for treatment of melanoma in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Curcumin/pharmacology , Melanoma, Experimental/pathology , NF-kappa B/metabolism , Animals , Annexin A5/metabolism , Caspase 3/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D , Cyclins/genetics , Cyclins/metabolism , Cyclooxygenase 2 , Down-Regulation , Electrophoretic Mobility Shift Assay , G1 Phase/drug effects , Luciferases/metabolism , Melanoma, Experimental/metabolism , Mice , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured
3.
Cancer Res ; 67(5): 2298-305, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332361

ABSTRACT

Recently, several laboratories have started to investigate the involvement of glutamate signaling in cancer. In previous studies, we reported on a transgenic mouse model that develops melanoma spontaneously. Subsequent studies in these mice identified that the aberrant expression of metabotropic glutamate receptor 1 (GRM1) in melanocytes played a critical role in the onset of melanoma. Confirmation of the etiologic role of GRM1 in melanoma development was shown in a second transgenic line with GRM1 expression under the regulation of a melanocyte-specific dopachrome tautomerase promoter. Ectopic expression of GRM1 was also detected in a subset of human melanoma cell lines and biopsies, suggesting that aberrant expression of GRM1 in melanocytes may contribute to the development of human melanoma. GRM1, a seven-transmembrane domain G protein-coupled receptor, is normally expressed and functional in neuronal cells, and its ligand, glutamate, is the major excitatory neurotransmitter. Human melanoma cells are shown here to release elevated levels of glutamate, implying a possible autocrine loop. Treatment of GRM1-expressing human melanoma cells with a GRM1 antagonist (LY367385 or BAY36-7620) or a glutamate release inhibitor (riluzole) leads to a suppression of cell proliferation as well as a decrease in levels of extracellular glutamate. Treatment of human melanoma cell xenografts with riluzole for 18 days via p.o. gavage or i.v. injection leads to inhibition of tumor growth by 50% in comparison with controls. These data suggest the importance of glutamate signaling in human melanoma and imply that the suppression of glutamate signaling may be a new target for melanoma therapy.


Subject(s)
Glutamic Acid/metabolism , Melanoma/etiology , Receptors, Metabotropic Glutamate/physiology , Skin Neoplasms/etiology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Humans , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , Mutant Proteins/physiology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/genetics , Riluzole/therapeutic use , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cell Signal ; 18(8): 1279-86, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16305822

ABSTRACT

Metabotropic glutamate receptor 1 (Grm1, formerly mGluR1) is a G protein coupled receptor (GPCR) normally expressed and functional in the central nervous system. Studies of our transgenic mouse melanoma model (TG-3) revealed that ectopic expression of Grm1 in melanocytes is sufficient to induce melanoma development in vivo [P.M. Pollock, K. Cohen-Solal, R. Sood, J. Namkoong, J.J. Martino, A. Koganti, H. Zhu, C. Robbins, I. Makalowska, S.S. Shin, Y. Marin, K.G. Roberts, L.M. Yudt, A. Chen, J. Cheng, A. Incao, H.W. Pinkett, C.L. Graham, K. Dunn, S.M. Crespo-Carbone, K.R. Mackason, K.B. Ryan, D. Sinsimer, J. Goydos, K.R. Reuhl, M. Eckhaus, P.S. Meltzer, W.J. Pavan, J.M. Trent, S. Chen, Nat. Genet. 34 (2003) 108-112.]. We have established and characterized several cell lines in vitro from independent mouse melanoma tumors [Y.E. Marín, J. Namkoong, S.S. Shin, J. Raines, K. Degenhardt, E. White, S. Chen, Neuropharmacol. 49 (2005) 70-79.]. These cell lines are useful tools in the studies of signaling events that may be mediated by Grm1 in transformed melanocytes. Here we show that stimulation of Grm1 by l-quisqualate, a group I metabotropic glutamate receptor agonist, results in inositol triphosphate (IP3) accumulation, and the activation of ERK1/2 in these cell lines. IP3 accumulation and ERK1/2 activation were inhibited by pretreatment of the tumor cells with a Grm1-specific antagonist (LY367385) or by dominant negative mutants of Grm1, demonstrating the specificity of these events. We also show that ERK1/2 activation by Grm1 was PKC-dependent, but cAMP and PKA-independent. PKCepsilon was shown to play a pivotal role in Grm1-mediated ERK1/2 phosphorylation. Insights into the signaling cascades mediated by Grm1 in melanoma cells may aid in the identification of key molecular targets for the future design of combined therapies for melanoma.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Melanoma/metabolism , Oncogenes/genetics , Protein Kinase C-epsilon/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Genes, Dominant/genetics , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Melanoma/pathology , Mice , Mutation/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/genetics , Quisqualic Acid/pharmacology
5.
Int J Oncol ; 27(4): 941-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142309

ABSTRACT

The phorbol ester, 12-O-tetradecanoylphorbol-13-acetate (TPA) is a potent stimulator of differentiation and apoptosis in myeloid leukemia cells. In the present study, we investigated the role of the transcription factor NF-kappaB in TPA-induced growth inhibition and apoptosis in the myeloid leukemia HL-60 cell line and its TPA-resistant cell variant HL-525. Unlike the parental cell line, HL-525 cells are protein kinase C (PKC)-beta deficient and resistant to TPA-induced differentiation and apoptosis. We found that treatment of HL-60 cells with TPA resulted in a concentration-dependent growth inhibition and an increase in apoptotic cells. TPA only had a small effect on growth and apoptosis in HL-525 cells. Treatment of HL-60 cells with TPA (0.64-3.2 nM) caused a rapid activation of NF-kappaB as determined by electrophoresis mobility shift assay (EMSA) and immunocytochemistry. Although the basal level of NF-kappaB activity was low in HL-60 cells, TPA-resistant HL-525 cells had a high basal level of NF-kappaB activity. Treatment of HL-525 cells with higher concentrations of TPA (16-80 nM) resulted in a further increase in NF-kappaB activity. (E)3-[(4-methylphenyl)-sulfonyl]-2-propenenitrile (BAY 11-7082; BAY), which inhibits IkappaB alpha phosphorylation and thus decreases NF-kappaB activation, was found to decrease TPA-induced nuclear translocation of NF-kappaB. Furthermore, BAY enhanced TPA-induced growth inhibition and apoptosis in both HL-60 and HL-525 cells. Results from the present study indicate that inhibition of NF-kappaB by BAY was associated with enhanced TPA-induced growth inhibition and apoptosis in human myeloid leukemia cells. TPA in combination with pharmacological inhibitors of NF-kappaB may improve the therapeutic efficacy of TPA and overcome the resistance to TPA in some myeloid leukemia patients.


Subject(s)
Apoptosis , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid/drug therapy , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Sulfones/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Active Transport, Cell Nucleus , Cell Adhesion , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , DNA/chemistry , DNA Damage , Dose-Response Relationship, Drug , HL-60 Cells , Humans , I-kappa B Proteins/metabolism , Immunophenotyping , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , Propidium/pharmacology , Protein Kinase C/metabolism , Protein Kinase C beta , Time Factors
6.
Neuropharmacology ; 49 Suppl 1: 70-9, 2005.
Article in English | MEDLINE | ID: mdl-16040064

ABSTRACT

Melanoma is the aberrant proliferation of melanocytes, the cells in the skin responsible for pigment (melanin) production. In its early stages, melanoma can be surgically removed with great success, however, advanced stages of melanoma have a high mortality rate due to the lack of responsiveness to currently available therapies. We have previously characterized a mouse melanoma model, TG-3, which has implicated the ectopic expression of metabotropic glutamate receptor 1 (Grm1, formerly mGluR1), in melanomagenesis and metastasis [Pollock et al., 2003. Melanoma mouse model implicates metabotropic glutamate signaling in melanocytic neoplasia. Nat Genet. 34, 108-112.]. Here we report the characterization of several in vitro cell lines derived from independent mouse melanoma tumors. These cell lines show characteristic phenotypes of transformed melanocytes, and express Grm1, and Grm5 (another metabotropic glutamate receptor), as well as melanocyte-specific protein markers. To investigate the possible role of Grm5 in vivo during melanoma development in our mice, we have crossed Grm5 null mice with TG-3, generating a new line of transgenic mice, TGM. TGMs, which are homozygote knockouts for Grm5 and carry the TG transgene, develop tumors with onset, progression, and metastasis very similar to that described for TG-3. Taken together, these results indicate that Grm1 can act as an oncogene in melanocytes independently of Grm5 expression.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Melanocytes/metabolism , Melanoma, Experimental/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/physiology , Animals , Blotting, Western/methods , Ear Neoplasms/metabolism , Ear Neoplasms/pathology , Fluorescent Antibody Technique/methods , Humans , Melanoma/metabolism , Melanoma/pathology , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Oxidoreductases/metabolism , RNA, Messenger/metabolism , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/deficiency , Reverse Transcriptase Polymerase Chain Reaction/methods , Tetrazolium Salts , Thiazoles , Time Factors , Transfection/methods , Tumor Cells, Cultured
7.
J Mol Med (Berl) ; 82(11): 735-49, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15322701

ABSTRACT

Melanoma is the aberrant proliferation of melanocytes, the cells in the skin responsible for pigment production. In the United States the current lifetime risk of melanoma development is 1 in 57 in males and 1 in 81 in females. In its early stages melanoma can be surgically removed with great success; however, advanced stages of melanoma have a high mortality rate due to the lack of responsiveness to currently available therapies. The development of animal models to be used in the studies of melanoma will provide the means for developing improved and targeted treatments for this disease. This review focuses on the recent report of a mouse melanoma model, TG-3, which has implicated the ectopic expression of the metabotropic glutamate receptor 1 (Grm1), a G protein coupled receptor (GPCR), in melanomagenesis and metastasis. The involvement of other GPCRs in cellular transformation, particularly GPCRs in melanoma biology, and signaling of Grm1 are also discussed.


Subject(s)
Melanoma/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Disease Models, Animal , Female , Humans , Male , Melanocytes/metabolism , Melanoma/etiology , Melanoma/genetics , Mice , Mice, Transgenic , Mutation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...