Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hum Pathol ; 45(8): 1630-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24837095

RESUMEN

Patients with cholangiocarcinoma often present with locally advanced or metastatic disease. There is a need for effective therapeutic strategies for advanced stage cholangiocarcinoma. Recently, FGFR2 translocations have been identified as a potential target for tyrosine kinase inhibitor therapies. This study evaluated 152 cholangiocarcinomas and 4 intraductal papillary biliary neoplasms of the bile duct for presence of FGFR2 translocations by fluorescence in situ hybridization and characterized the clinicopathologic features of cases with FGFR2 translocations. Thirteen (10 women, 3 men; 8%) of 156 biliary tumors harbored FGFR2 translocations, including 12 intrahepatic cholangiocarcinomas (12/96; 13%) and 1 intraductal papillary neoplasm of the bile duct. Histologically, cholangiocarcinomas with FGFR2 translocations displayed prominent intraductal growth (62%) or anastomosing tubular glands with desmoplasia (38%). Immunohistochemically, the tumors with FGFR2 translocations frequently showed weak and patchy expression of CK19 (77%). Markers of the stem cell phenotype in cholangiocarcinoma, HepPar1 and CK20, were negative in all cases. The median cancer-specific survival for patients whose tumors harbored FGFR2 translocations was 123 months compared to 37 months for cases without FGFR2 translocations (P = .039). This study also assessed 100 cholangiocarcinomas for ERBB2 amplification and ROS1 translocations. Of the cases tested, 3% and 1% were positive for ERBB2 amplification and ROS1 translocation, respectively. These results confirm that FGFR2, ERRB2, and ROS1 alterations are potential therapeutic targets for intrahepatic cholangiocarcinoma.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/metabolismo , Colangiocarcinoma/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Translocación Genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de los Conductos Biliares/mortalidad , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Colangiocarcinoma/mortalidad , Colangiocarcinoma/patología , Femenino , Humanos , Queratina-20/genética , Queratina-20/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Tasa de Supervivencia
2.
J Biol Chem ; 285(3): 1879-87, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19917613

RESUMEN

Protein kinase Cdelta (PKCdelta) is an essential component of the intrinsic apoptotic program. Following DNA damage, such as exposure to UV radiation, PKCdelta is cleaved in a caspase-dependent manner, generating a constitutively active catalytic fragment (PKCdelta-cat), which is necessary and sufficient for keratinocyte apoptosis. We found that in addition to inducing apoptosis, expression of PKCdelta-cat caused a pronounced G(2)/M cell cycle arrest in both primary human keratinocytes and immortalized HaCaT cells. Consistent with a G(2)/M arrest, PKCdelta-cat induced phosphorylation of Cdk1 (Tyr(15)), a critical event in the G(2)/M checkpoint. Treatment with the ATM/ATR inhibitor caffeine was unable to prevent PKCdelta-cat-induced G(2)/M arrest, suggesting that PKCdelta-cat is functioning downstream of ATM/ATR in the G(2)/M checkpoint. To better understand the role of PKCdelta and PKCdelta-cat in the cell cycle response to DNA damage, we exposed wild-type and PKCdelta null mouse embryonic fibroblasts (MEFs) to UV radiation. Wild-type MEFs underwent a pronounced G(2)/M arrest, Cdk1 phosphorylation, and induction of apoptosis following UV exposure, whereas PKCdelta null MEFs were resistant to these effects. Expression of PKCdelta-green fluorescent protein, but not caspase-resistant or kinase-inactive PKCdelta, was able to restore G(2)/M checkpoint integrity in PKCdelta null MEFs. The function of PKCdelta in the DNA damage-induced G(2)/M cell cycle checkpoint may be a critical component of its tumor suppressor function.


Asunto(s)
Dominio Catalítico , División Celular/fisiología , Daño del ADN , Fase G2/fisiología , Proteína Quinasa C-delta/química , Proteína Quinasa C-delta/metabolismo , Animales , Biocatálisis , División Celular/efectos de la radiación , Línea Celular , Fase G2/efectos de la radiación , Humanos , Recién Nacido , Ratones , Proteína Quinasa C-delta/deficiencia , Proteína Quinasa C-delta/genética , Rayos Ultravioleta/efectos adversos
3.
Pigment Cell Melanoma Res ; 23(2): 216-24, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20015121

RESUMEN

Protein kinase C (PKC) is a heterogeneous family of serine/threonine protein kinases that have different biological effects in normal and neoplastic melanocytes (MCs). To explore the mechanism behind their differential response to PKC activation, we analyzed the expression profile of all nine PKC isoforms in normal human MCs, HPV16 E6/E7 immortalized MCs, and a panel of melanoma cell lines. We found reduced PKCbeta and increased PKCzeta and PKCiota expression at both the protein and mRNA levels in immortalized MCs and melanoma lines. We focused on PKCbeta as it has been functionally linked to melanin production and oxidative stress response. Re-expression of PKCbeta in melanoma cells inhibited colony formation in soft agar, indicating that PKCbeta loss in melanoma is important for melanoma growth. PKCbetaII, but not PKCbetaI, was localized to the mitochondria, and inhibition of PKCbeta significantly reduced UV-induced reactive oxygen species (ROS) in MCs with high PKCbeta expression. Thus alterations in PKCbeta expression in melanoma contribute to their neoplastic phenotype, possibly by reducing oxidative stress, and may constitute a selective therapeutic target.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/enzimología , Proteína Quinasa C/genética , Línea Celular , Proliferación Celular , Perfilación de la Expresión Génica , Humanos , Melanocitos/enzimología , Melanocitos/metabolismo , Melanoma/metabolismo , Melanoma/patología , Estrés Oxidativo , Fenotipo , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas , Rayos Ultravioleta
4.
J Invest Dermatol ; 129(6): 1351-60, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19037233

RESUMEN

Rapid downregulation of the antiapoptotic Bcl-2 family protein myeloid cell leukemia 1 (Mcl-1) is required for UV-induced apoptosis, underlining an important role for Mcl-1 in epidermal pathology. To determine if Mcl-1 has a specific role in normal keratinocyte (KC) biology, Mcl-1 was downregulated in human KCs by RNAi and these KCs were induced to differentiate in organotypic raft cultures. Mcl-1 shRNA organotypic cultures showed increased levels of spontaneous premature apoptosis, implicating Mcl-1 as an essential KC survival protein. Mcl-1-downregulated cultures also had reduced granular and cornified layers, and produced lower levels of cross-linked protein and cornified envelopes. Cornification could only partially be rescued with the general caspase inhibitor z-VAD, suggesting that reduced cornification was not entirely because of premature apoptosis. Differentiation markers (K1, K10, filaggrin, loricrin, cleaved caspase-14) were normally expressed in control organotypic cultures, but were expressed at reduced levels in organotypic cultures with downregulated Mcl-1. The defect in differentiation marker expression was independent of apoptosis as it could not be rescued by z-VAD. Thus, Mcl-1 serves two important, independent functions in epidermal KCs: acting as a major survival protein by inhibiting premature apoptosis in the spinous and granular layers to promote conification, and promoting the robust induction of KC differentiation markers.


Asunto(s)
Diferenciación Celular , Epidermis/metabolismo , Queratinocitos/citología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Apoptosis , Caspasa 3/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Proteínas Filagrina , Regulación de la Expresión Génica , Humanos , Queratinocitos/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Piel/metabolismo
5.
Cancer Res ; 66(19): 9636-45, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17018621

RESUMEN

By deciphering the dysregulation of apoptosis in melanoma cells, new treatment approaches exploiting aberrant control mechanisms regulating cell death can be envisioned. Among the Bcl-2 family, a BH3-only member, NOXA, functions in a specific mitochondrial-based cell death pathway when melanoma cells are exposed to a proteasome inhibitor (e.g., bortezomib). Some therapeutic agents, such as bortezomib, not only induce proapoptotic Bcl-2 family members and active conformational changes in Bak and Bax but also are associated with undesirable effects, including accumulation of antiapoptotic proteins, such as Mcl-1. To enhance the bortezomib-mediated killing of melanoma cells, the apoptotic pathway involving NOXA was further investigated, leading to identification of an important target (i.e., the labile Bcl-2 homologue Mcl-1 but not other survival proteins). To reduce Mcl-1 levels, melanoma cells were pretreated with several different agents, including Mcl-1 small interfering RNA (siRNA), UV light, or the purine nucleoside analogue fludarabine. By simultaneously triggering production of NOXA (using bortezomib) as well as reducing Mcl-1 levels (using siRNA, UV light, or fludarabine), significantly enhanced killing of melanoma cells was achieved. These results show binding interactions between distinct Bcl-2 family members, such as NOXA and Mcl-1, in melanoma cells, paving the way for novel and rational therapeutic combination strategies, which target guardians of the proapoptotic Bak- and Bax-mediated pathways, against this highly aggressive and often fatal malignancy.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Melanoma/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Pirazinas/farmacología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/administración & dosificación , Bortezomib , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Línea Celular Tumoral/efectos de la radiación , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Melanoma/secundario , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Pirazinas/administración & dosificación , ARN Interferente Pequeño/farmacología , Rayos Ultravioleta , Vidarabina/administración & dosificación , Vidarabina/análogos & derivados , Vidarabina/farmacología , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
6.
J Biol Chem ; 281(40): 29703-10, 2006 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-16901898

RESUMEN

Proteolytic cleavage and subsequent activation of protein kinase C (PKC) delta is required for apoptosis induced by a variety of genotoxic agent, including UV radiation. In addition, overexpression of the constitutively active PKCdelta catalytic fragment (PKCdelta-cat) is sufficient to trigger Bax activation, cytochrome c release, and apoptosis. While PKCdelta is a key apoptotic effector, the downstream target(s) responsible for the mitochondrial apoptotic cascade are not known. We found that expression of the active PKCdelta-cat in HaCaT cells triggers a reduction in the anti-apoptotic protein Mcl-1, similar to UV radiation. The down-regulation of Mcl-1 induced by PKCdelta-cat was not at the mRNA level but was due to decreased protein half-life. Overexpression of Mcl-1 protected HaCaT cells from both UV and PKCdelta-cat-induced apoptosis and blocked the release of cytochrome c from the mitochondria, indicating that Mcl-1 down-regulation was required for apoptosis signaling. Indeed, down-regulation of Mcl-1 with siRNA slightly increased the basal apoptotic rate of HaCaT cells and dramatically sensitized them to UV or PKCdelta-cat-induced apoptosis. HaCaT cells with down-regulated Mcl-1 had higher activated Bax protein, as measured by Bax cross-linking, indicating that Mcl-1 down-regulation is sufficient for Bax activation. Finally, recombinant PKCdelta could phosphorylate Mcl-1 in vitro, identifying Mcl-1 as a direct target for PKCdelta. Overall our results identify Mcl-1 as an important target for PKCdelta-cat that can mediate its pro-apoptotic effects on mitochondria to amplify the apoptotic signaling induced by a wide range of apoptotic stimuli.


Asunto(s)
Apoptosis/fisiología , Dominio Catalítico/fisiología , Proteínas de Neoplasias/metabolismo , Fragmentos de Péptidos/fisiología , Proteína Quinasa C-delta/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Línea Celular Transformada , Citocromos c/antagonistas & inhibidores , Citocromos c/metabolismo , Retroalimentación Fisiológica/fisiología , Células HeLa , Humanos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteína Quinasa C-delta/biosíntesis , Proteína Quinasa C-delta/genética , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Transducción de Señal/fisiología , Rayos Ultravioleta
7.
Exp Dermatol ; 15(1): 14-22, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16364027

RESUMEN

Whether terminal differentiation/stratum corneum formation of keratinocytes (KCs) represents a form of programmed cell death, utilizing mediators of classical apoptosis, is unclear. Apoptosis, an evolutionarily conserved death process, is comprised of extrinsic and intrinsic pathways, which converge using caspase 3. To define upstream and downstream caspases involved in terminal differentiation, we utilized human epidermal equivalents (EEs). Using submerged cultures comprised of human KCs, EEs were sequentially analyzed before and after being raised to an air/liquid (A/L) interface at 3-24 h intervals. At each time point, EEs were analyzed morphologically and for specific enzyme activity to distinguish different initiator (caspases 1, 2, 8, 9) and effector caspases (3, 6, 7). Terminal differentiation began at 6-8 h, as defined by stratum corneum with loricirin expression and completed at 18-24 h producing an epidermis resembling normal skin. Enzyme activity for caspases 1, 2, 3, 6, 7, 8, and 9 (but not 4, 5) was enhanced (>two-fold nmol/mg/h) at 3-6 h compared with submerged cultures. Processing of caspase 14 occurred at 18 h, and cleaved caspase 14 was increased at 24 h. Activated caspase 3-positive and terminal deoxynucleotidyl transferase-mediated nick end labeling-positive KCs were identified in EEs at 3-6 h corresponding to initiation sites of terminal differentiation. Addition of caspase inhibitors reduced levels of involucrin and loricrin in EEs raised to an A/L interface. We conclude caspases function as important death effectors strategically positioned at intersection of intrinsic and extrinsic pathways in KCs undergoing stratum corneum formation.


Asunto(s)
Apoptosis/fisiología , Caspasas/fisiología , Epidermis/fisiología , Western Blotting , Caspasas/metabolismo , Diferenciación Celular/fisiología , Activación Enzimática , Humanos , Técnicas para Inmunoenzimas , Queratinocitos/fisiología , Modelos Biológicos
8.
Oncogene ; 24(34): 5299-312, 2005 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-15940268

RESUMEN

Ultraviolet (UV) light exposure is a common cause of epithelial-derived skin cancers, and the epidermal response to UV-light has been extensively studied using both mouse models and cultured human keratinocytes (KCs). Elimination of cells with UV-induced DNA damage via apoptosis provides a powerful mechanism to minimize retention or expansion of genetically abnormal cells. This cell editing function has largely been ascribed to the biological role of the p53 tumor suppressor gene, as mutations or deletions involving p53 have been linked to skin cancer development. Rather than introducing mutations, or using cells with complete loss of wild-type p53, we used an siRNA-based approach to knockdown, but not eliminate, p53 levels in primary cultures of human KCs followed by UV-irradiation. Surprisingly, when p53 levels were reduced by 50-80% the apoptosis induced by exposure to UV-light was accelerated and markedly enhanced (two- to three- fold) compared to control siRNA treated KCs. The p53 siRNA treated KCs were characterized by elevated E2F-1 levels accompanied by accelerated elimination of the Mcl-1 and Bcl-x(L) antiapoptotic proteins, as well as enhanced Bax oligomerization. Forced overexpression of either Mcl-1 or Bcl-x(L) reduced the UV-light enhanced apoptotic response in p53 siRNA treated KCs. We conclude that p53 not only can provide proapoptotic signals but also regulates a survival pathway influencing Mcl-1 and Bcl-x(L) levels. This overlooked survival function of p53 may explain previous paradoxical responses noted by investigators using p53 heterozygous and knockout mouse models, and opens up the possibility that not all liaisons within the cell involving p53 necessarily represent fatal attractions.


Asunto(s)
Apoptosis , Queratinocitos/fisiología , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Rayos Ultravioleta/efectos adversos , Animales , Apoptosis/efectos de la radiación , Supervivencia Celular , Daño del ADN , Regulación hacia Abajo , Humanos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Interferencia de ARN , Regulación hacia Arriba , Proteína bcl-X
9.
J Invest Dermatol ; 123(3): 434-43, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15304079

RESUMEN

The constitutively active catalytic domain of protein kinase C (PKC)delta is an apoptotic effector generated by caspase-3 cleavage of full-length PKCdelta in response to a wide variety of apoptotic stimuli, including UV radiation. The PKCdelta catalytic domain induces apoptosis when ectopically expressed, however, the mechanism of apoptosis induction is unclear. We constructed a chimeric protein encoding the PKCdelta catalytic domain fused to a mutated estrogen receptor ligand-binding domain in order to selectively activate the PKCdelta catalytic domain. The enzymatic activity of the PKCdelta catalytic domain fusion protein was induced in human keratinocytes treated with 4-hydroxytamoxifen, and its activation triggered loss of mitochondrial membrane potential and apoptosis. The apoptosis was associated with release of cytochrome c from the mitochondria and caspase activation, and was blocked by caspase inhibitors and the anti-apoptotic proteins Bcl-2, and Bcl-x(L), suggesting a role for mitochondrial pore formation. Consistent with this, the activated PKCdelta catalytic domain triggered the redistribution and activation of Bax, a Bcl-2 family protein that can directly induce cytochrome c release. In summary, despite being an apoptotic effector activated late in the apoptotic cascade, PKCdelta also activates upstream components of the death effector pathway to insure the demise of cells committed to apoptosis.


Asunto(s)
Apoptosis/fisiología , Queratinocitos/citología , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Caspasa 3 , Caspasas/metabolismo , Dominio Catalítico , Línea Celular , Citocromos c/metabolismo , Retroalimentación Fisiológica , Humanos , Queratinocitos/enzimología , Proteína Quinasa C/química , Proteína Quinasa C/genética , Proteína Quinasa C-delta , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2
10.
J Cell Physiol ; 200(1): 155-66, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15137068

RESUMEN

The impact of low-dose ultraviolet light (UV-light) on apoptotic susceptibility of keratinocytes (KCs) induced by TRAIL is unclear. Skin expresses a functional form of TRAIL, and while sun exposure influences TRAIL death receptors, a role for decoy receptors has not been evaluated. Unraveling mechanisms involving apoptotic sensitivity of KCs is important because skin is the first target of UV-light, and a site for commonly occurring cancers. Since apoptosis is a homeostatic process eliminating UV-light induced DNA damaged cells, elucidating molecular events regulating apoptosis enhances understanding of cutaneous photocarcinogenesis. Here we demonstrate low-dose UV-light enhances susceptibility of KCs to TRAIL-induced apoptosis. Low-dose UV-light selectively reduces decoy receptors, without influencing death receptor levels. UV-induced enhanced apoptotic susceptibility was reduced by over-expression of decoy receptor TRAIL-R4, but not TRAIL-R3; or treatment with thiol compound pyrrolidine dithiocarbamate (PDTC), which also enhanced TRAIL-R4 levels. Besides influencing decoy receptors, low-dose UV-light plus TRAIL also synergistically promoted cytochrome c and Smac release from mitochondria. Inhibitors directed against caspases 2, 3, 8, and 9 reduced the synergistic apoptotic response following low-dose UV-light plus TRAIL exposure; as did forced over-expression of Bcl-x and dominant negative (DN) constructs of FADD and caspase 9. Thus, relative levels of decoy receptors significantly influence susceptibility of KCs to TRAIL-induced apoptosis with concomitant low-dose UV-light exposure; in addition to the apoptotic pathway mediated by mitochondrial permeabilization.


Asunto(s)
Apoptosis/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Glicoproteínas de Membrana/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Reguladoras de la Apoptosis , Western Blotting , Proteínas Portadoras/metabolismo , Inhibidores de Caspasas , Caspasas/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Péptidos y Proteínas de Señalización Intracelular , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Luciferasas/genética , Masculino , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Proteínas Mitocondriales/metabolismo , Pene/anatomía & histología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/metabolismo , Retroviridae/genética , Piel/citología , Ligando Inductor de Apoptosis Relacionado con TNF , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta , Proteína bcl-X
11.
J Biol Chem ; 277(22): 19346-52, 2002 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-11919192

RESUMEN

UV radiation from the sun activates both the membrane death receptor and the intrinsic or mitochondrial apoptotic signaling pathways in epidermal keratinocytes, triggering apoptosis and affording protection against skin cancer formation. We have investigated the involvement of caspase-9 in the UV death effector pathway in human keratinocytes, since this is the initiating caspase in the mitochondrial pathway required for UV-induced apoptosis in some, but not all, cell types. UV radiation triggered activation of caspase-3, caspase-9, and caspase-8 with similar kinetics, although the rank order of activation was caspase-3 > caspase-9 > caspase-8. Inhibition of caspase-9 with either the peptide inhibitor benzyloxycarbonyl-Leu-Glu(OCH(3))-His-Asp(OCH(3))-fluoromethyl ketone, or expression of a catalytically inactive caspase-9 by retroviral transduction, protected normal keratinocytes from UV-induced apoptosis. HaCaT keratinocytes harboring mutant p53 alleles were also protected from UV-induced apoptosis by the dominant negative caspase-9. The dominant negative caspase-9 blocked UV-induced activation of caspase-3, caspase-9, and caspase-8, and also protected cells from the loss of mitochondrial membrane potential. In contrast, the dominant negative caspase-9 did not protect from anti-Fas-induced apoptosis or caspase activation. These results identify caspase-9 as the critical upstream caspase initiating apoptosis by UV radiation in human keratinocytes, the relevant cell type for this important environmental carcinogen.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Alelos , Western Blotting , Caspasa 8 , Caspasa 9 , Células Cultivadas , ADN Complementario/metabolismo , Activación Enzimática , Precursores Enzimáticos/metabolismo , Citometría de Flujo , Genes p53 , Humanos , Cinética , Microscopía Fluorescente , Mitocondrias/metabolismo , Modelos Biológicos , Mutación , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Factores de Tiempo , Rayos Ultravioleta , Receptor fas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA