Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Carcinogenesis ; 36(11): 1372-80, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26271098

RESUMEN

Solar ultraviolet irradiation is an environmental carcinogen that causes skin cancer. Caspase-7 is reportedly expressed at reduced levels in many cancers. The present study was designed to examine the role of caspase-7 in solar-simulated light (SSL)-induced skin cancer and to elucidate its underlying molecular mechanisms. Our study revealed that mice with genetic deficiency of caspase-7 are highly susceptible to SSL-induced skin carcinogenesis. Epidermal hyperplasia, tumor volume and the average number of tumors were significantly increased in caspase-7 knockout (KO) mice compared with SKH1 wild-type mice irradiated with SSL. The expression of cell proliferation markers, such as survivin and Ki-67, was elevated in SSL-irradiated skin of caspase-7 KO mice compared with those observed in SSL-exposed wild-type SKH1 mouse skin. Moreover, SSL-induced apoptosis was abolished in skin from caspase-7 KO mice. Two-dimensional gel electrophoresis, followed by matrix-assisted laser desorption/ionization-time-of-flight analysis of skin tissue lysates from SSL-irradiated SKH1 wild-type and caspase-7 KO mice revealed an aberrant induction of keratin-17 in caspase-7 KO mice. Immunohistochemical analysis of skin tumors also showed an increase of keratin-17 expression in caspase-7 KO mice compared with SKH1 wild-type mice. The expression of keratin-17 was also elevated in SSL-irradiated caspase-7 KO keratinocytes as well as in human basal cell carcinomas. The in vitro caspase activity assay showed keratin-17 as a substrate of caspase-7, but not caspase-3. Overall, our study demonstrates that genetic loss of caspase-7 promotes SSL-induced skin carcinogenesis by blocking caspase-7-mediated cleavage of keratin-17.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Caspasa 7/genética , Queratinas/fisiología , Traumatismos Experimentales por Radiación/enzimología , Neoplasias Cutáneas/enzimología , Luz Solar/efectos adversos , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Caspasa 7/metabolismo , Células Cultivadas , Epidermis/enzimología , Epidermis/patología , Epidermis/efectos de la radiación , Femenino , Técnicas de Inactivación de Genes , Queratinocitos/enzimología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteolisis , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Carga Tumoral
2.
Mol Carcinog ; 54(11): 1513-20, 2015 11.
Artículo en Inglés | MEDLINE | ID: mdl-25307283

RESUMEN

Sulforaphane is a natural product found in broccoli, which is known to exert many different molecular effects in the cell, including inhibition of histone deacetylase (HDAC) enzymes. Here, we examine for the first time the potential for sulforaphane to inhibit HDACs in HaCaT keratinocytes and compare our results with those found using HCT116 colon cancer cells. Significant inhibition of HDAC activity in HCT116 nuclear extracts required prolonged exposure to sulforaphane in the presence of serum. Under the same conditions HaCaT nuclear extracts did not exhibit reduced HDAC activity with sulforaphane treatment. Both cell types displayed down-regulation of HDAC protein levels by sulforaphane treatment. Despite these reductions in HDAC family member protein levels, acetylation of marker proteins (acetylated Histone H3, H4, and tubulin) was decreased by sulforaphane treatment. Time-course analysis revealed that HDAC6, HDAC3, and acetylated histone H3 protein levels are significantly inhibited as early as 6 h into sulforaphane treatment. Transcript levels of HDAC6 are also suppressed after 48 h of treatment. These results suggest that HDAC activity noted in nuclear extracts is not always translated as expected to target protein acetylation patterns, despite dramatic inhibition of some HDAC protein levels. In addition, our data suggest that keratinocytes are at least partially resistant to the nuclear HDAC inhibitory effects of sulforaphane, which is exhibited in HCT116 and other cells.


Asunto(s)
Anticarcinógenos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Isotiocianatos/farmacología , Acetilación/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Células HCT116 , Histona Desacetilasa 6 , Histonas/metabolismo , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Sulfóxidos
3.
Arch Biochem Biophys ; 558: 143-52, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25004464

RESUMEN

One of the primary components of the East Indian sandalwood oil (EISO) is α-santalol, a molecule that has been investigated for its potential use as a chemopreventive agent in skin cancer. Although there is some evidence that α-santalol could be an effective chemopreventive agent, to date, purified EISO has not been extensively investigated even though it is widely used in cultures around the world for its health benefits as well as for its fragrance and as a cosmetic. In the current study, we show for the first time that EISO-treatment of HaCaT keratinocytes results in a blockade of cell cycle progression as well as a concentration-dependent inhibition of UV-induced AP-1 activity, two major cellular effects known to drive skin carcinogenesis. Unlike many chemopreventive agents, these effects were not mediated through an inhibition of signaling upstream of AP-1, as EISO treatment did not inhibit UV-induced Akt or MAPK activity. Low concentrations of EISO were found to induce HaCaT cell death, although not through apoptosis as annexin V and PARP cleavage were not found to increase with EISO treatment. However, plasma membrane integrity was severely compromised in EISO-treated cells, which may have led to cleavage of LC3 and the induction of autophagy. These effects were more pronounced in cells stimulated to proliferate with bovine pituitary extract and EGF prior to receiving EISO. Together, these effects suggest that EISO may exert beneficial effects upon skin, reducing the likelihood of promotion of pre-cancerous cells to actinic keratosis (AK) and skin cancer.


Asunto(s)
Anticarcinógenos/farmacología , Autofagia/efectos de los fármacos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Medicina Tradicional , Aceites de Plantas/farmacología , Sesquiterpenos/farmacología , Animales , Bovinos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Quimioprevención , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteolisis/efectos de los fármacos , Factor de Transcripción AP-1/antagonistas & inhibidores
4.
Mol Carcinog ; 47(5): 349-60, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17935202

RESUMEN

A cell line that produces mouse squamous cell carcinoma (6M90) was modified to develop a cell line with an introduced Tet-responsive catalase transgene (MTOC2). We have previously reported that the overexpressed catalase in the MTOC2 cells reverses the malignant phenotype in part by decreasing epidermal growth factor receptor (EGFR) signaling. With this work we expanded the investigation into the differences between these two cell lines. We found that the decreased EGFR pathway activity of the MTOC2 cells is not because of reduced autocrine secretion of an epidermal growth factor (EGF) ligand but rather because of lower basal receptor activity. Phosphorylated levels of the mitogen-activated protein kinase (MAPK) members JNK and p38 were both higher in the 6M90 cells with low catalase when compared with the MTOC2 cell line. Although treatment with an EGFR inhibitor, AG1478, blocked the increased activity of JNK in the 6M90 cells, a similar effect was not observed for p38. Basal levels of downstream c-jun transcription were also found to be higher in the 6M90 cells versus MTOC2 cells. Activated p38 was found to down-regulate the JNK MAPK pathway in the 6M90 cells. However, the 6M90 cells contain constitutively high levels of phosphorylated JNK, generating higher levels of phosphorylated c-jun and total c-jun than those in the MTOC2 cells. Inhibition of JNK activity in the 6M90 cells reduced AP-1 transcription and cell proliferation. The data confirm the inhibitory effects of catalase on tumor cell growth, specifically through a ligand-independent decrease in the stress activated JNK pathway.


Asunto(s)
Carcinoma de Células Escamosas/patología , Catalasa/metabolismo , Proliferación Celular , Receptores ErbB/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Queratinocitos/citología , Animales , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Catalasa/genética , Ciclo Celular , Células Cultivadas , Queratinocitos/metabolismo , Ratones , Fosforilación , Especies Reactivas de Oxígeno , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional , Transgenes/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Photochem Photobiol ; 84(2): 463-76, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18248498

RESUMEN

Acute UVB irradiation of mouse skin results in activation of phospatidyinositol-3 (PI-3) kinase and mitogen-activated protein kinase (MAPK) pathways leading to altered protein phosphorylation and downstream transcription of genes. We determined whether activation of these pathways also occurs in human skin exposed to 4x minimal erythemic dose of UVB in 23 volunteers. Biopsies were taken prior to, at 30 min, 1 and 24 h post-UVB. In agreement with mouse studies, the earliest UV-induced changes in epidermis were seen in phospho-CREB (two- and five-fold at 30 min and 1 h) and in phospho-MAPKAPK-2 (three-fold at both 30 min and 1 h). At 1 h, phospho-c-JUN and phospho-p38 were increased five- and two-fold, respectively. Moreover, phospho-c-JUN and phospho-p38 were further increased at 24 h (12- and six-fold, respectively). Phospho-GSK-3beta was similarly increased at all time points. Increases in phospho-p53 (12-fold), COX-2 (four-fold), c-FOS (14-fold) and apoptosis were not seen until 24 h. Our data suggest that UVB acts through MAPK p38 and PI-3 kinase with phosphorylation of MAPKAPK-2, CREB, c-JUN, p38, GSK-3beta and p53 leading to marked increases in c-FOS, COX-2 and apoptosis. Validation of murine models in human skin will aid in development of effective skin cancer chemoprevention and prevention strategies.


Asunto(s)
Transducción de Señal/efectos de la radiación , Piel/efectos de la radiación , Rayos Ultravioleta , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Piel/metabolismo
6.
Mol Cancer Res ; 3(2): 90-9, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15755875

RESUMEN

Activation of activator protein-1 (AP-1) and increased expression of cyclooxygenase-2 (COX-2) have been clearly shown to play a functional role in UVB-induced skin tumor promotion. In this study, we examined UVB-induced signal transduction pathways in SKH-1 mouse epidermis leading to increases in COX-2 expression and AP-1 activity. We observed rapid increases in p38 mitogen-activated protein kinase (MAPK) signaling through activation of p38 MAPK and its downstream target, MAPK activated protein kinase-2. UVB also increased phosphatidylinositol 3-kinase (PI3K) signaling as observed through increases in AKT and GSK-3beta phosphorylation. Activation of the p38 MAPK and PI3K pathways results in the phosphorylation of cyclic AMP-responsive element binding protein, which was also observed in UVB-irradiated SKH-1 mice. Topical treatment with SB202190 (a specific inhibitor of p38 MAPK) or LY294002 (a specific inhibitor of PI3K) significantly decreased UVB-induced AP-1 activation by 84% and 68%, respectively, as well as COX-2 expression. Our data show that in mouse epidermis, UVB activation of the p38 MAPK and PI3K pathways leads to AP-1 activation and COX-2 expression.


Asunto(s)
Epidermis/efectos de la radiación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Prostaglandina-Endoperóxido Sintasas/metabolismo , Factor de Transcripción AP-1/metabolismo , Rayos Ultravioleta , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Cromonas/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclooxigenasa 2 , Epidermis/enzimología , Imidazoles/farmacología , Ratones , Ratones Pelados , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
7.
Photochem Photobiol ; 82(2): 423-31, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16613495

RESUMEN

Inhibition of c-Jun N-terminal kinase (JNK) with the pharmacologic inhibitor SP600125 in UVA-irradiated HaCaT cells and human primary keratinocytes resulted in dramatic phenotypic changes indicative of cell death. These phenotypic changes correlated with caspase 8, 9 and 3 activations as well as cleavage of the caspase substrate polyADP-ribose polymerase (PARP). Morphologic analysis and analysis of sub-G0 DNA content confirmed apoptotic cell death in these keratinocytes after combination treatment. Addition of the general caspase inhibitor zVAD-fmk to combination-treated HaCaT cells was able to completely block caspase activation, PARP cleavage, the increase in sub-G0 DNA content and the classic morphologic features of apoptosis, indicating that this combination treatment resulted in caspase-dependent apoptotic cell death. zVAD-fmk treatment of primary keratinocytes was able to completely inhibit caspase activation and PARP cleavage, reduce morphologic apoptosis at lower concentrations of SP600125 and decrease the sub-G(0) DNA content detected after UVA + SP600125 treatment. However, cell death and a significant amount of debris was still detected after caspase inhibitor treatment, particularly with 125 nM SP600125. At subconfluent conditions and low passage, primary keratinocytes were more sensitive to UVA irradiation alone than HaCaT cells. In conclusion, we have observed that inhibition of UVA-induced JNK activity with the pharmacologic inhibitor SP600125 resulted in caspase-dependent apoptotic cell death in both the immortalized keratinocyte cell line HaCaT and primary keratinocytes. However, the increased sensitivity of primary keratinocytes to experimental stress may have also resulted in direct cellular injury and caspase-independent cell death.


Asunto(s)
Antracenos/farmacología , Apoptosis/efectos de los fármacos , Caspasas/fisiología , Fragmentación del ADN/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Queratinocitos/efectos de los fármacos , Rayos Ultravioleta , Clorometilcetonas de Aminoácidos/farmacología , Línea Celular , Fragmentación del ADN/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de la radiación , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Fenotipo , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factores de Tiempo
8.
Cancer Res ; 63(9): 2292-9, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12727852

RESUMEN

Degradation of the extracellular matrix by proteolytic enzymes is a central aspect of physiological and pathologic tissue-remodeling processes such as trophoblastic implantation, wound healing, and tumor invasion. We have hypothesized that prostate adenocarcinoma cell invasion through the normal basal lamina is attributable in part to metalloproteinase-induced cleavage of laminin-5 (Ln-5) and enhanced motility of the cancer cells. We studied the role of membrane type-1-matrix metalloproteinase (MT1-MMP) expressed on the surface of prostate tumor cells in cleaving Ln-5 and enhancing the migration of prostate tumor cells. We also determined the nature of the MT1-MMP cleavage of human Ln-5 and how this altered Ln-5 changes the migration of prostate carcinoma cells. We found that human MT1-MMP cleaves purified human Ln-5 to an 80-kDa fragment. Mass spectrometry analyses of the 80-kDa cleaved product by trypsin and chymotrypsin gave 14 and 9 different peptide sequences, respectively, that were identical to the expected amino acid sequence of the Ln-5-beta3 chain. The recovered peptides represent 14.4% (trypsin) and 10.3% (chymotrypsin) of Ln-5-beta3 chain by amino acid count. Both trypsin and chymotrypsin digestion of MT1-MMP-cleaved product of Ln-5 did not show any other peptides that were identical to the other chains of Ln-5. Using a linear migration assay we found that the Ln-5 cleaved by MT1-MMP enhanced the migration of DU-145 prostate carcinoma cells by 2-fold compared with uncleaved Ln-5. The use of blocked antisense MT1-MMP oligonucleotides inhibited the migration of DU-145 cells on Ln-5. We also found that the prostate carcinoma cells expressing high levels of MT1-MMP, such as PC3N and PPC, demonstrated enhanced migration on human Ln-5-coated substrate, and this migration was inhibited using blocked antisense MT1-MMP oligonucleotides. In conclusion, this is a novel and important finding where we have shown that beta3-chain is cleaved by MT1-MMP, and this cleavage enhances migration of prostate cancer cells.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/fisiología , Metaloendopeptidasas/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Moléculas de Adhesión Celular/inmunología , Humanos , Masculino , Metaloproteinasas de la Matriz Asociadas a la Membrana , Metaloendopeptidasas/biosíntesis , Metaloendopeptidasas/farmacología , Datos de Secuencia Molecular , Invasividad Neoplásica , Neoplasias de la Próstata/metabolismo , Proteínas Recombinantes/farmacología , Células Tumorales Cultivadas , Kalinina
9.
Neoplasia ; 7(9): 873-82, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16229810

RESUMEN

One of the puzzles in cancer predisposition is that women carrying BRCA-1 mutations preferentially develop tumors in epithelial tissues of the breast and ovary. Moreover, sporadic breast tumors contain lower levels of BRCA-1 in the absence of mutations in the BRCA-1 gene. The problem of tissue specificity requires analysis of factors that are unique to tissues of the breast. For example, the expression of estrogen receptor-alpha (ER alpha) is inversely correlated with breast cancer risk, and 90% of BRCA-1 tumors are negative for ER alpha. Here, we show that estrogen stimulates BRCA-1 promoter activity in transfected cells and the recruitment of ER alpha and its cofactor p300 to an AP-1 site that binds Jun/Fos transcription factors. The recruitment of ER alpha/p300 coincides with accumulation in the S-phase of the cell cycle and is antagonized by the antiestrogen tamoxifen. Conversely, we document that overexpression of wild-type p53 prevents the recruitment of ER alpha to the AP-1 site and represses BRCA-1 promoter activity. Taken together, our findings support a model in which an ER alpha/AP-1 complex modulates BRCA-1 transcription under conditions of estrogen stimulation. Conversely, the formation of this transcription complex is abrogated in cells overexpressing p53.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Receptor alfa de Estrógeno/metabolismo , Factor de Transcripción AP-1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína BRCA1/genética , Línea Celular Tumoral , ADN de Neoplasias/metabolismo , Estrógenos/farmacología , Células HCT116 , Células HeLa , Humanos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/genética
10.
Clin Cancer Res ; 8(1): 149-55, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11801552

RESUMEN

More than one million new skin cancers are diagnosed yearly in the United States creating the need for effective primary and chemopreventive strategies to reduce the incidence, morbidity, and mortality associated with skin cancer. Skin chemoprevention trials often focus on subjects at high risk of nonmelanoma skin cancers and include biological endpoints like number of actinic keratoses (AK) and measures of cell proliferation, apoptosis, and p53 expression and/or mutation. Difluoromethylornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase, suppresses increased polyamine synthesis and inhibits tumors in models of skin carcinogenesis. Thus, DFMO is a good candidate chemopreventive agent in humans at increased risk of NMSC. We reported previously results of a randomized, placebo-controlled trial of topical DFMO in 48 participants with AK. In this study there was a significant reduction in the number of AK (23.5%; P = 0.001) and the polyamine, spermidine (26%, P = 0.04; Alberts, D. S. et al. Cancer Epidemiol. Biomark. Prev., 9: 1281-2186, 2000). In skin biopsies from the same study, we demonstrate that topical DFMO significantly reduces the percentage of p53-positive cells (22%; P = 0.04); however, there were no significant changes in proliferating cell nuclear antigen or apoptotic indices, or in the frequency of p53 mutations (25% at baseline, 21% after placebo, and 26% after DFMO). We conclude that inhibition of the premalignant AK lesions as well as a reduction in the expression of p53 and in spermidine concentrations may serve as surrogate endpoint biomarkers of DFMO and possibly other topically administered skin cancer chemopreventive agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Eflornitina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Queratosis/prevención & control , Trastornos por Fotosensibilidad/prevención & control , Neoplasias Cutáneas/prevención & control , Anciano , Apoptosis/efectos de los fármacos , Biopsia , División Celular/efectos de los fármacos , Femenino , Humanos , Técnicas para Inmunoenzimas , Queratosis/metabolismo , Masculino , Mutación , Inhibidores de la Ornitina Descarboxilasa , Trastornos por Fotosensibilidad/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Riesgo , Neoplasias Cutáneas/metabolismo , Espermidina/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Neoplasia ; 5(4): 319-29, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14511403

RESUMEN

To further delineate ultraviolet A (UVA) signaling pathways in the human keratinocyte cell line HaCaT, we examined the potential role of mitogen-activated protein kinases (MAPKs) in UVA-induced activator protein-1 (AP-1) transactivation and c-Fos expression. UVA-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK) proteins was detected immediately after irradiation and disappeared after approximately 2 hours. Conversely, phosphorylation of extracellular signal-regulated kinase was significantly inhibited for up to 1 hour post-UVA irradiation. To examine the role of p38 and JNK MAPKs in UVA-induced AP-1 and c-fos transactivations, the selective pharmacologic MAPK inhibitors, SB202190 (p38 inhibitor) and SP600125 (JNK inhibitor), were used to independently treat stably transfected HaCaT cells in luciferase reporter assays. Both SB202190 and SP600125 dose-dependently inhibited UVA-induced AP-1 and c-fos transactivations. SB202190 (0.25-0.5 microM) and SP600125 (62-125 nM) treatments also primarily inhibited UVA-induced c-Fos expression. These results demonstrated that activation of both JNK and p38 play critical role in UVA-mediated AP-1 transactivation and c-Fos expression in these human keratinocyte cells. Targeted inhibition of these MAPKs with their selective pharmacologic inhibitors may be effective chemopreventive strategies for UVA-induced nonmelanoma skin cancer.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal , Factor de Transcripción AP-1/metabolismo , Antracenos/farmacología , Western Blotting , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Imidazoles/farmacología , Luciferasas/metabolismo , MAP Quinasa Quinasa 4 , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Piridinas/farmacología , Neoplasias Cutáneas/patología , Factores de Tiempo , Activación Transcripcional , Rayos Ultravioleta , Proteínas Quinasas p38 Activadas por Mitógenos
12.
Crit Rev Oncol Hematol ; 41(3): 269-85, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11880204

RESUMEN

The incidence of skin cancer has been rising in recent years with significant effects on public health. Primary prevention has proven inadequate in impacting the incidence of skin cancer, thus stimulating the development of chemopreventive strategies. The majority of skin cancer chemoprevention studies focus on occurrence of new nonmelanoma skin cancers (NMSC) in individuals with a previous NMSC, or on reduction in the number of premalignant skin lesions such as actinic keratoses (AK). Dysplastic nevi, a likely precursor of melanoma, are also potential targets for chemoprevention strategies. Premalignant lesions are especially attractive as endpoints since they are more common than frank cancer, resulting in reduced sample size, length, and cost of clinical trials. Development of new agents that affect the pathogenesis of skin cancer will be discussed, from elucidation of molecular targets to implementation of trials designed to determine the effects of chemopreventive interventions on human skin cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Cutáneas/prevención & control , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Suplementos Dietéticos , Humanos , Protectores contra Radiación/uso terapéutico , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/etiología , Rayos Ultravioleta/efectos adversos
13.
Recent Results Cancer Res ; 163: 151-64; discussion 264-6, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12903851

RESUMEN

There are over 1 million cases of skin cancer diagnosed yearly in the United States. The majority of these are nonmelanoma (NMSCs) and are associated with chronic exposure to ultraviolet light (UV). Actinic keratosis (AK) has been identified as a precursor for SCC, but not for BCC. AKs are far more common than SCC, making them excellent targets for chemoprevention. Cancer chemoprevention can prevent or delay the occurrence of cancer in high-risk populations using dietary or chemical interventions. We have developed strategies that have rational mechanisms of action and demonstrate activity in preclinical models of skin cancer. Promising agents proceed to phase I-III trials in subjects at high risk of skin cancer. UV light induces molecular signaling pathways and results in specific genetic alterations (i.e., mutation of p53) that are likely critical to skin cancer development. UVB-induced changes serve as a basis for the development of novel agents. Targets include inhibition of polyamine or prostaglandin synthesis, specific retinoid receptors, and components of the Ras and MAP kinase signaling pathways. Agents under study include: epigallocatechin gallate (EGCG), a green tea catechin with antioxidant and sunscreen activity, as well as UVB signal transduction blocking activity; perillyl alcohol, a monoterpene derived from citrus peel that inhibits Ras farnesylation; difluoromethylornithine (DFMO), an inhibitor of ornithine decarboxylase and polyamines; retinoids that target retinoid X receptors and AP-1 activity; and nonsteroidal anti-inflammatory agents that inhibit cylooxygenase and prostaglandin synthesis. We performed a series of Phase I-II trials in subjects with multiple AK. For example, a phase II randomized trial of topical DFMO reduced AK number, suppressed polyamines, and reduced p53 protein. Our goal is to develop agents for use in combination and/or incorporation into sunscreens to improve chemoprevention efficacy and reduce skin cancer incidence.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Cutáneas/prevención & control , Animales , Carcinoma Basocelular/prevención & control , Carcinoma de Células Escamosas/prevención & control , Quimioprevención , Ensayos Clínicos como Asunto , Humanos , Melanoma/prevención & control , Ratones , Neoplasias Inducidas por Radiación/prevención & control , Transducción de Señal/efectos de la radiación , Piel/efectos de la radiación , Rayos Ultravioleta/efectos adversos
14.
Photochem Photobiol ; 75(3): 302-10, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11950097

RESUMEN

To determine whether the transcription factor activator protein-1 (AP-1) could be modulated by ultraviolet A (UVA) exposure, we examined AP-1 DNA-binding activity and transactivation after exposure to UVA in the human immortalized keratinocyte cell line HaCaT. Maximal AP-1 transactivation was observed with 250 kJ/m2 UVA between 3 and 4 h after irradiation. DNA binding of AP-1 to the target 12-O-tetradecanoylphorbol-13-acetate response element sequence was maximally induced 1-3 h after irradiation. Both de novo transcription and translation contributed to the UVA-induced AP-1 DNA binding. c-Fos was implicated as a primary component of the AP-1 DNA-binding complex. Other components of the complex included Fra-2, c-Jun, JunB and JunD. UVA irradiation induced protein expression of c-Fos, c-Jun, Fra-1 and Fra-2. Phosphorylated forms of these induced proteins were determined at specific time points. A strong correlation existed between UVA-induced AP-1 activity and accumulation of c-Fos, c-Jun and Fra-1 proteins. UVA irradiation also induced c-fos and c-jun mRNA expression and transcriptional activation of the c-fos gene promoter. These results demonstrate that UVA irradiation activates AP-1 and that c-fos induction may play a critical role in the response of these human keratinocytes to UVA irradiation.


Asunto(s)
Queratinocitos/efectos de la radiación , Factor de Transcripción AP-1/metabolismo , Rayos Ultravioleta , Línea Celular Transformada , ADN/metabolismo , Humanos , Queratinocitos/metabolismo , Factor de Transcripción AP-1/fisiología , Activación Transcripcional/fisiología
15.
Cancer Prev Res (Phila) ; 5(3): 403-13, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22389437

RESUMEN

Reverse phase protein microarray analysis was used to identify cell signaling derangements in squamous cell carcinoma (SCC) compared with actinic keratosis (AK) and upper inner arm (UIA). We analyzed two independent tissue sets with isolation and enrichment of epithelial cells by laser capture microdissection. Set 1 served as a pilot and a means to identify protein pathway activation alterations that could be further validated in a second independent set. Set 1 was comprised of 4 AK, 13 SCC, and 20 UIA. Set 2 included 15 AK, 9 SCCs, and 20 UIAs. Activation of 51 signaling proteins, known to be involved in tumorigenesis, were assessed for set 1 and showed that the MEK-ERK [mitogen-activated protein (MAP)/extracellular signal-regulated (ERK; MEK)] pathway was activated in SCC compared with AK and UIA, and that epidermal growth factor receptor (EGFR) and mTOR pathways were aberrantly activated in SCC. Unsupervised two-way hierarchical clustering revealed that AK and UIA shared a common signaling network activation architecture while SCC was dramatically different. Statistical analysis found that prosurvival signaling through phosphorylation of ASK and 4EBP1 as well as increased Bax and Bak expression was higher in AK compared with UIA. We expanded pathway network activation mapping in set 2 to 101 key signaling proteins, which corroborated activation of MEK-ERK, EGFR, and mTOR pathways through discovery of a number of upstream and downstream signaling molecules within these pathways to conclude that SCC is indeed a pathway activation-driven disease. Pathway activation mapping of SCC compared with AK revealed several interconnected networks that could be targeted with drug therapy for potential chemoprevention and therapeutic applications.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Piel/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Femenino , Humanos , Captura por Microdisección con Láser , Masculino , Persona de Mediana Edad , Fosforilación , Pronóstico , Estudios Prospectivos , Neoplasias Cutáneas/patología , Adulto Joven
16.
Cancer Prev Res (Phila) ; 3(2): 160-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20103724

RESUMEN

The chemopreventive and antitumor properties of perillyl alcohol (POH) that were studied preclinically indicate that topical POH inhibits both UVB-induced murine skin carcinogenesis (squamous cell tumor models) and 7,12-dimethylbenz(a)anthracene-induced murine melanoma (transgenic models involving tyrosinase-driven Ras). A previous phase 1 clinical trial in participants with normal-appearing skin showed that topical POH cream was well tolerated at a dose of 0.76% (w/w). Here, we performed a 3-month, double-blind, randomized, placebo-controlled phase 2a trial of two different doses of topical POH in individuals with sun-damaged skin. Participants applied POH cream twice daily to each dorsal forearm. Baseline and end-of-study biopsies were taken from each participant to evaluate whether the topical application of POH was effective in reversing actinic damage as evidenced by normalization of quantitative skin histopathologic scores and change in nuclear chromatin pattern as measured by karyometric analysis. There was a borderline reduction in the histopathologic score of the lower-dose POH group compared with the placebo (P = 0.1), but this was not observed in the high-dose group. However, in the high-dose group, a statistically significant reduction in the proportion of nuclei deviating from normal was observed by the use of karyometric analysis (P < 0.01). There was no statistical significance shown in the lower-dose group. No changes were observed in p53 expression, cellular proliferation (by proliferating cell nuclear antigen expression), or apoptosis in either treatment group compared with the placebo group. These results suggest that whereas our karyometric analyses can detect a modest effect of POH in sun-damaged skin, improved delivery into the epidermis may be necessary.


Asunto(s)
Antineoplásicos/administración & dosificación , Monoterpenos/administración & dosificación , Neoplasias Cutáneas/prevención & control , Administración Tópica , Anciano , Apoptosis/efectos de los fármacos , Quimioprevención/métodos , Cromatina/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Inmunohistoquímica , Cariotipificación , Masculino , Persona de Mediana Edad
17.
Cancer Prev Res (Phila) ; 3(10): 1277-83, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20876729

RESUMEN

Cutaneous squamous cell carcinoma (SCC) occurs commonly and can metastasize. Identification of specific molecular aberrations and mechanisms underlying the development and progression of cutaneous SCC may lead to better prognostic and therapeutic approaches and more effective chemoprevention strategies. To identify genetic changes associated with early stages of cutaneous SCC development, we analyzed a series of 40 archived skin tissues ranging from normal skin to invasive SCC. Using high-resolution array-based comparative genomic hybridization, we identified deletions of a region on chromosome 10q harboring the INPP5A gene in 24% of examined SCC tumors. Subsequent validation by immunohistochemistry on an independent sample set of 71 SCC tissues showed reduced INPP5A protein levels in 72% of primary SCC tumors. Decrease in INPP5A protein levels seems to be an early event in SCC development, as it also is observed in 9 of 26 (35%) examined actinic keratoses, the earliest stage in SCC development. Importantly, further reduction of INPP5A levels is seen in a subset of SCC patients as the tumor progresses from primary to metastatic stage. The observed frequency and pattern of loss indicate that INPP5A, a negative regulator of inositol signaling, may play a role in development and progression of cutaneous SCC tumors.


Asunto(s)
Carcinoma de Células Escamosas/genética , Monoéster Fosfórico Hidrolasas/genética , Neoplasias Cutáneas/genética , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Hibridación Genómica Comparativa , Dosificación de Gen , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Inositol Polifosfato 5-Fosfatasas , Análisis de Secuencia por Matrices de Oligonucleótidos , Monoéster Fosfórico Hidrolasas/metabolismo , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología
18.
Cancer Res ; 69(17): 7103-10, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19671797

RESUMEN

Sulforaphane is an isothiocyanate derived from cruciferous vegetables that has been linked to decreased risk of certain cancers. Although the role of sulforaphane in the induction of the transcription factor Nrf2 has been studied extensively, there is also evidence that inhibition of the transcription factor activator protein-1 (AP-1) may contribute to the chemopreventive properties of this compound. In this study, we show for the first time that sulforaphane is effective at reducing the multiplicity and tumor burden of UVB-induced squamous cell carcinoma in a mouse model using cotreatment with the compound and the carcinogen. We also show that sulforaphane pretreatment is able to reduce the activity of AP-1 luciferase in the skin of transgenic mice after UVB. Chromatin immunoprecipitation analysis verified that a main constituent of the AP-1 dimer, cFos, is inhibited from binding to the AP-1 DNA binding site by sulforaphane. Electrophoretic mobility shift assay analysis of nuclear proteins also shows that sulforaphane and diamide, both known to react with cysteine amino acids, are effective at inhibiting AP-1 from binding to its response element. Using truncated recombinant cFos and cJun, we show that mutation of critical cysteines in the DNA-binding domain of these proteins (Cys(154) in cFos and Cys(272) in cJun) results in loss of sensitivity to both sulforaphane and diamide in electrophoretic mobility shift assay analysis. Together, these data indicate that inhibition of AP-1 activity may be an important molecular mechanism in chemoprevention of squamous cell carcinoma by sulforaphane.


Asunto(s)
Anticarcinógenos/farmacología , Carcinoma de Células Escamosas/prevención & control , Neoplasias Cutáneas/prevención & control , Tiocianatos/farmacología , Factor de Transcripción AP-1/antagonistas & inhibidores , Animales , Sitios de Unión , Carcinoma de Células Escamosas/metabolismo , Cisteína , Modelos Animales de Enfermedad , Femenino , Isotiocianatos , Ratones , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Neoplasias Cutáneas/metabolismo , Sulfóxidos , Factor de Transcripción AP-1/química , Factor de Transcripción AP-1/metabolismo , Rayos Ultravioleta
19.
J Biol Chem ; 279(41): 42658-68, 2004 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-15292226

RESUMEN

We examined the effect of inhibiting p38 MAPK on UVA-irradiated HaCaT cells, a spontaneously immortalized human keratinocyte cell line. Recent work from our laboratory has shown that UVA (250 kJ/m2) induces a rapid phosphorylation of p38 MAPK in the HaCaT cell line. Inhibition of p38 MAPK activity through the use of a specific inhibitor, SB202190, in combination with UVA treatment induced a rapid cleavage of caspase-9, caspase-8, and caspase-3, whereas UVA irradiation alone had no effect. Similarly, cleavage of the caspase substrate poly(ADP-ribose) polymerase was observed in UVA-irradiated HaCaT cells treated with SB202190 or in cells expressing a dominant-negative p38 MAPK. No effect of p38 MAPK inhibition upon caspase cleavage was observed in mock-irradiated HaCaT cells. In addition, increases in apoptosis were observed in UVA-irradiated cells treated with SB202190 by morphological analysis with no significant apoptosis occurring from UVA irradiation alone. Similar results were obtained by using normal human epidermal keratinocytes. UVA induced expression of the anti-apoptotic Bcl-2 family member, Bcl-XL, with abrogation of expression by using the p38 MAPK inhibitor SB202190. Overexpression of Bcl-XL prevented poly(ADP-ribose) polymerase cleavage induced by the combination of UVA and p38 MAPK inhibition. UVA enhanced the stability of Bcl-XL mRNA through increases in p38 MAPK activity. We determined that increases in UVA-induced expression of Bcl-XL occur through a posttranscriptional mechanism mediated by the 3'-untranslated region (UTR). We used Bcl-XL 3'-UTR luciferase constructs to determine the mechanism by which UVA increased Bcl-XL mRNA stability. Additionally, RNA binding studies indicate that UVA increases the binding of RNA-binding proteins to Bcl-XL 3'-UTR mRNA, which can be decreased by using SB202190. In conclusion, p38 MAPK and Bcl-XL expression play critical roles in the survival of UVA-irradiated HaCaT cells.


Asunto(s)
Queratinocitos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Procesamiento Postranscripcional del ARN , Rayos Ultravioleta , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Regiones no Traducidas 3' , Apoptosis , Western Blotting , Caspasa 3 , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Línea Celular , Línea Celular Tumoral , Citocromos c/metabolismo , Citoplasma/metabolismo , ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Genes Dominantes , Genes Reporteros , Humanos , Imidazoles/farmacología , Luciferasas/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , Fosforilación , Poli(ADP-Ribosa) Polimerasas/metabolismo , Piridinas/farmacología , ARN/metabolismo , ARN Mensajero/metabolismo , Retroviridae/genética , Factores de Tiempo , Transfección , Proteína bcl-X
20.
Prostate ; 53(1): 1-8, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12210475

RESUMEN

BACKGROUND: We have shown previously that interleukin (IL) -1 beta- and IL-6-induced promatrilysin expression is mediated by an indirect pathway that requires NF kappa B-dependent synthesis of IL-6 and STAT3 signaling. We now demonstrate that IL-1 beta-induced but not IL-6-induced promatrilysin expression can be blocked by androgens in the prostate carcinoma cell line LNCaP (lymph node-derived carcinoma cells of the prostate). METHODS: By using enzyme-linked immunosorbent assay analyses, promatrilysin was measured in LNCaP cells stimulated with IL-1 beta or IL-6 LNCaP-treated cells pretreated with testosterone. In addition, promatrilysin message was measured by using Northern analyses after IL-6-treated cells pretreated with testosterone. RESULTS: In LNCaP treated with testosterone before IL-1 beta stimulation induced promatrilysin expression was completely abrogated. Furthermore, testosterone completely abrogated NF kappa B transactivation activity and induction of IL-6 protein expression and mRNA. Testosterone and 5 alpha-dihydrotestosterone did not have an inhibitory effect on IL-6-induced promatrilysin expression. Testosterone also had no effect on basal promatrilysin expression or basal NF kappa B transactivation activity. CONCLUSION: From these data, we conclude that testosterone blocks IL-1 beta-induced promatrilysin expression by inhibition of NF kappa B transactivation activity, which in turn, blocks IL-6 expression. These data suggest a mechanism in vivo by which invasive and metastatic prostatic carcinoma cell clones refractory to hormone ablation therapy may develop after chemical or surgical castration. Furthermore, these data suggest that, perhaps, upstream targets such as the cytokines IL-1 beta and IL-6 may provide alternative drug targets for inhibiting prostate cancer progression.


Asunto(s)
Carcinoma/patología , Precursores Enzimáticos/biosíntesis , Interleucina-1/farmacología , Interleucina-6/farmacología , Metaloendopeptidasas/biosíntesis , Neoplasias de la Próstata/patología , Testosterona/farmacología , Northern Blotting , Resistencia a Antineoplásicos , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA