Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 68(14): 5743-52, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632627

RESUMEN

Although many melanomas harbor either activating mutations in BRAF or NRAS, there remains a substantial, yet little known, group of tumors without either mutation. Here, we used a genomic strategy to define a novel group of melanoma cell lines with co-overexpression of cyclin-dependent kinase 4 (CDK4) and KIT. Although this subgroup lacked any known KIT mutations, they had high phospho-KIT receptor expression, indicating receptor activity. Quantitative PCR confirmed the existence of a similar KIT/CDK4 subgroup in human melanoma samples. Pharmacologic studies showed the KIT/CDK4-overexpressing subgroup to be resistant to BRAF inhibitors but sensitive to imatinib in both in vitro and in vivo melanoma models. Mechanistically, imatinib treatment led to increased apoptosis and G(1) phase cell cycle arrest associated with the inhibition of phospho-ERK and increased expression of p27(KIP). Other melanoma cell lines, which retained some KIT expression but lacked phospho-KIT, were not sensitive to imatinib, suggesting that KIT expression alone is not predictive of response. We suggest that co-overexpression of KIT/CDK4 is a potential mechanism of oncogenic transformation in some BRAF/NRAS wild-type melanomas. This group of melanomas may be a subpopulation for which imatinib or other KIT inhibitors may constitute optimal therapy.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Animales , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Esferoides Celulares/metabolismo , Células Tumorales Cultivadas/metabolismo , Proteínas ras/metabolismo
2.
Cancer Res ; 68(9): 3413-20, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18451169

RESUMEN

In this study, we investigated the mechanism of apoptosis induction of obatoclax (GX15-070), a novel Bcl-2 homology domain-3 (BH3) mimetic, in acute myeloid leukemia (AML) cell lines and primary AML samples. Obatoclax inhibited cell growth of HL-60, U937, OCI-AML3, and KG-1 cell lines. Apoptosis induction contributed to the observed antiproliferative effects at concentrations of this agent that mirror its affinity for antiapoptotic Bcl-2 proteins. We show that obatoclax can promote the release of cytochrome c from isolated leukemia cell mitochondria and that apoptosis induced by this agent is preceded by the release of Bak from Mcl-1, liberation of Bim from both Bcl-2 and Mcl-1, and the formation of an active Bak/Bax complex. Notably, apoptosis was diminished, but not fully prevented, in the absence of Bak/Bax or Bim, suggesting that obatoclax has additional targets that contribute to its cytotoxicity. At growth inhibitory doses that did not induce apoptosis or decrease viability, obatoclax induced an S-G(2) cell-cycle block. Obatoclax induced apoptosis in AML CD34+ progenitor cells with an average IC(50) of 3.59 +/- 1.23 micromol/L although clonogenicity was inhibited at concentrations of 75 to 100 nmol/L. Obatoclax synergized with the novel BH3 mimetic ABT-737 to induce apoptosis in OCI-AML3 cells and synergistically induced apoptosis in combination with AraC in leukemic cell lines and in primary AML samples. In conclusion, we show that obatoclax potently induces apoptosis and decreases leukemia cell proliferation and may be used in a novel therapeutic strategy for AML alone and in combination with other targeted agents and chemotherapeutics.


Asunto(s)
Leucemia/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Pirroles/farmacología , Pirroles/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Factor de Transcripción de AraC/farmacología , Biomimética , Compuestos de Bifenilo/farmacología , Células Cultivadas , Sinergismo Farmacológico , Células HL-60 , Humanos , Indoles , Ratones , Ratones Noqueados , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/metabolismo , Nitrofenoles/farmacología , Piperazinas/farmacología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/farmacología , Ensayo de Tumor de Célula Madre , Células U937
3.
Clin Cancer Res ; 14(1): 230-9, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18172275

RESUMEN

PURPOSE: Disseminated melanoma is highly therapy resistant. The finding that 66% of melanomas harbor the activating BRAF(V600E) mutation has raised expectations for targeting the Ras/RAF/mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK pathway in melanoma. This study addresses the anti-melanoma activity of the MEK inhibitor AZD6244 (ARRY-142886). EXPERIMENTAL DESIGN: We recently have shown that growing melanoma cells as three-dimensional collagen-implanted spheroids enhances resistance to the MEK inhibitor U0126. Here, we investigated the anti-melanoma activity of AZD6244 in two-dimensional cell culture, the three-dimensional spheroid model, and an in vivo model. RESULTS: In two-dimensional cell culture, AZD6244 was cytostatic and reduced the growth of melanoma cells in a concentration-dependent fashion through the induction of G(1)-phase cell cycle arrest. In our three-dimensional spheroid model, the effects of AZD6244 were largely cytostatic and reversible, with drug washout leading to spheroid regrowth. Finally, 1205Lu cells were grown as tumor xenografts in severe combined immunodeficient mice. After tumor establishment, mice were dosed twice daily with 0, 10, or 30 mg/kg AZD6244 p.o. AZD6244 treatment decreased phospho-ERK in the tumors and significantly suppressed tumor growth. The original tumors remained viable, suggesting that AZD6244 monotherapy was largely cytostatic, and not proapoptotic in this model. Further studies showed that co-administration of AZD6244 (30 mg/kg) with docetaxel (15 mg/kg) led to tumor regression, indicating the potential for MEK inhibitor/chemotherapy drug combinations. CONCLUSIONS: Inhibition of MEK is cytostatic as a monotherapy in melanoma, but cytotoxic when combined with docetaxel.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bencimidazoles/administración & dosificación , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Melanoma/tratamiento farmacológico , Animales , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Citostáticos/administración & dosificación , Citotoxinas/administración & dosificación , Docetaxel , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Etiquetado Corte-Fin in Situ , Melanoma/enzimología , Ratones , Ratones SCID , Hibridación de Ácido Nucleico , Taxoides/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Biol Ther ; 6(12): 1967-77, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18075297

RESUMEN

Acute promyelocytic leukemia (APL) is associated with oncogenic PML-RARalpha that acts as a dominant negative transcriptional repressor of retinoic acid (RA) receptor target genes by recruiting histone deacetylase (HDAC). The peroxisome proliferator-activated receptor-gamma (PPARgamma) is a member of the nuclear receptor family that forms heterodimers with retinoid X receptor (RXR). In addition to RAR targets, PML-RARalpha silence a wide range of nuclear receptor target genes including PPARgamma targets. All-trans-retinoic acid (ATRA), a ligand for the RA receptor (RAR), restores normal retinoid signaling and induces terminal differentiation of APL cells; however, APL cells can develop resistance to ATRA. Using ATRA sensitive NB4 and ATRA-resistant derivative MR2 cell lines, we demonstrate that PPARgamma ligand 2-cyano-3, 12-dioxooleana-1, 9-dien-28-oic acid (CDDO) enhances pro-apoptotic and differentiating effects of ATRA in ATRA-sensitive NB4 cells and partially reverses ATRA resistance in MR2 cells. The CDDO/ATRA combination synergistically induces RARbeta2 expression both in ATRA-sensitive and -resistant APL cells. RARbeta2 MrNA induction by CDDO/ATRA was mediated in part by enhanced H3-Lys9 acetylation in the RARbeta2 promoter which in turn increased the affinity of RARbeta for betaRARE. PPARgamma specific inhibitor T007 and silencing of PPARgamma by siRNA diminished CDDO-induced maturation and RARbeta2 mRNA along with PPARgamma induction indicating that PPARgamma activation is at least partially responsible for the RARbeta2 transcription and maturation induction. In an in vivo mouse model of APL, CDDO derivative CDDO-methyl ester markedly enhanced ATRA-induced maturation and extended the survival of mice. In summary, these results provide rationale for the combined targeting of RAR and PPARgamma nuclear receptors in the therapy of APL.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Ácido Oleanólico/análogos & derivados , PPAR gamma/efectos de los fármacos , Tretinoina/farmacología , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Ácidos Nicotínicos/farmacología , Ácido Oleanólico/farmacología , Ácido Oleanólico/uso terapéutico , Regiones Promotoras Genéticas/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Receptores de Ácido Retinoico/genética , Elementos de Respuesta/efectos de los fármacos , Tetrahidronaftalenos/farmacología , Tretinoina/agonistas
5.
Cancer Res ; 67(1): 209-17, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17210701

RESUMEN

Unlike other tumors, melanomas harbor wild-type (WT) p53 but exhibit impaired p53-dependent apoptosis. The mechanisms for the impaired p53 activation are poorly understood but may be linked to the high expression of the p53 suppressor Mdm2, which is found in >50% of melanoma lesions. Here, we describe an organometallic glycogen synthase kinase 3beta (GSK3beta) inhibitor (DW1/2) as a potent activator of p53 and inducer of cell death in otherwise highly chemoresistant melanoma cells. Using RNA interference and pharmacologic approaches, we show that p53 is required for the cytotoxic effects of this organometallic inhibitor. The DW1/2 compound was barely able to induce cell death in melanoma cells with p53 mutations, further confirming the requirement for p53-WT in the cytotoxic effects of the GSK3beta inhibition. Mechanistic analysis of the p53-dependent cell death indicated an apoptotic mechanism involving depolarization of mitochondrial membrane potential, caspase cleavage, and elevated NOXA expression. The effect of p53 was not simply due to passive up-regulation of protein expression as adenoviral-mediated overexpression of p53 was not able to induce cell death. Treatment of melanoma cells with DW1/2 was instead found to decrease levels of Mdm2 and Mdm4. The importance of Mdm2 down-regulation in DW1/2-induced apoptosis was confirmed by treating the p53-WT cells with the p53/Mdm2 antagonist Nutlin-3. Taken together, our data provide a new strategy for the pharmacologic activation of p53 in melanoma, which may be a viable approach for overcoming apoptotic resistance in melanoma and offer new hope for rational melanoma therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Melanoma/tratamiento farmacológico , Compuestos Organometálicos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Melanoma/metabolismo , Melanoma/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Rutenio/química , Rutenio/farmacología , Proteína p53 Supresora de Tumor/biosíntesis , Regulación hacia Arriba
6.
Cancer Cell ; 10(5): 375-88, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17097560

RESUMEN

BCL-2 proteins are critical for cell survival and are overexpressed in many tumors. ABT-737 is a small-molecule BH3 mimetic that exhibits single-agent activity against lymphoma and small-cell lung cancer in preclinical studies. We here report that ABT-737 effectively kills acute myeloid leukemia blast, progenitor, and stem cells without affecting normal hematopoietic cells. ABT-737 induced the disruption of the BCL-2/BAX complex and BAK-dependent but BIM-independent activation of the intrinsic apoptotic pathway. In cells with phosphorylated BCL-2 or increased MCL-1, ABT-737 was inactive. Inhibition of BCL-2 phosphorylation and reduction of MCL-1 expression restored sensitivity to ABT-737. These data suggest that ABT-737 could be a highly effective antileukemia agent when the mechanisms of resistance identified here are considered.


Asunto(s)
Apoptosis/fisiología , Compuestos de Bifenilo , Resistencia a Antineoplásicos/fisiología , Leucemia Mieloide Aguda , Nitrofenoles , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas , Animales , Compuestos de Bifenilo/metabolismo , Compuestos de Bifenilo/uso terapéutico , Línea Celular , Dimerización , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/metabolismo , Nitrofenoles/metabolismo , Nitrofenoles/uso terapéutico , Piperazinas/metabolismo , Piperazinas/uso terapéutico , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sulfonamidas/metabolismo , Sulfonamidas/uso terapéutico , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/química , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
7.
Ann Hematol ; 85(10): 689-704, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16832676

RESUMEN

The acute promyelocytic leukemia-specific PML-RARalpha fusion protein is a dominant-negative transcriptional repressor of retinoic acid receptor (RAR) target genes, which recruits HDAC and corepressor proteins and inhibits coactivators. Another oncogenic transcription factor, AML1-ETO, was proposed to cause an HDAC-dependent repression of RAR target genes. The RAR target RARbeta2 gene has been reported to be frequently silenced by hypermethylation in many types of cancer cells. We examined the methylation status of the RARbeta2 and asked if demethylation could reverse ATRA resistance in ATRA-resistant PML-RARalpha and AML1-ETO-positive cells. PML-RARalpha positive NB4 and its ATRA-resistant subvariant MR2 and AML1-ETO expressing Kasumi-1 cells had heterozygous methylation of RARbeta2. Although DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine partially reversed RARbeta2 CpG methylation in these cells, it did not significantly enhance ATRA-induced RARbeta2 mRNA expression and induction of maturation. However, the histone acetylase inhibitor SAHA combined with ATRA significantly reactivated RARbeta2 mRNA both in NB4 and MR2 cells with degradation of PML-RARalpha, which was associated with maturation. In contrast, SAHA did not affect AML1-ETO levels and failed to induce RARbeta2 expression and maturation in Kasumi-1 cells. In primary AML samples, RARbeta2 expression was uniformly low; however, no specific correlation was observed between the methylation of the RARbeta2 gene and expression of the fusion proteins, PML-RARalpha, and AML1-ETO. These results demonstrate that oncogenic PML-RARalpha and AML1-ETO translocations are rarely associated with RARbeta2 promoter methylation in primary AML samples.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/biosíntesis , Metilación de ADN , Leucemia Promielocítica Aguda/metabolismo , Proteínas de Fusión Oncogénica/biosíntesis , Regiones Promotoras Genéticas , Receptores de Ácido Retinoico/biosíntesis , Translocación Genética , Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Decitabina , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células HL-60 , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Humanos , Células K562 , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Proteína 1 Compañera de Translocación de RUNX1 , Receptores de Ácido Retinoico/genética , Translocación Genética/efectos de los fármacos , Translocación Genética/genética , Tretinoina/farmacología , Células U937
8.
Mol Pharmacol ; 69(4): 1182-93, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16410408

RESUMEN

Methyl-2-cyano-3,12 dioxoolean-1,9 diene-28-oate (CDDO-Me) is a synthetic oleanolic acid derivative that displays antitumorigenic and anti-inflammatory activities, and we have previously reported that this agent potently activates the intrinsic apoptotic pathway in leukemia cells. In this study, we demonstrate that mitochondrial dysfunction induced by CDDO-Me is mediated by direct permeabilization of the inner mitochondrial membrane, which results in the rapid depletion of mitochondrial glutathione (GSXm), loss of cardiolipin, and inhibition of mitochondrial respiration. More importantly, we demonstrate that in addition to activating the intrinsic apoptotic pathway, the mitochondrial effects of CDDO-Me may mediate its anti-inflammatory activity by modulating the generation of superoxide anion (O2*). It is noteworthy that CDDO-Me did not increase the generation of O2* and pretreatment of leukemia cells with CDDO-Me prevented the increase of this reactive oxygen species elicited by inhibition of complex I or III in the absence of de novo protein synthesis. CDDO-Me, but not other inhibitors of respiration, induced a time- and dose-dependent, cyclosporin A-independent permeability transition (PT) of isolated mitochondria that was sensitive to sulfhydryl antioxidants but not to EDTA. PT induced by CDDO-Me and Ca2+ was accompanied by loss of GSXm, suggesting that the increased permeability of the inner mitochondrial membrane facilitates the loss of this antioxidant. Finally, transmission electron microscopy revealed that CDDO-Me rapidly induced caspase-independent mitochondrial swelling and loss of inner membrane structure before the release of cytochrome c. Taken together, our results indicate that CDDO-Me is a novel mitochondriotoxic agent that induces apoptosis and inhibits mitochondrial electron transport via perturbations in inner mitochondrial membrane integrity.


Asunto(s)
Apoptosis/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Cardiolipinas/metabolismo , Línea Celular , Transporte de Electrón , Humanos , Membranas Intracelulares/efectos de los fármacos
9.
Blood ; 107(4): 1546-54, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16223781

RESUMEN

The multidrug resistance 1 (MDR1) gene product P-glycoprotein (P-gp) is frequently implicated in cross-resistance of tumors to chemotherapeutic drugs. In contrast, acute promyelocytic leukemia (APL) cells do not express MDR1 and are highly sensitive to anthracyclines. The combination of ATRA and the novel histone deacetylase inhibitor (HDACI) depsipeptide (FK228) induced P-gp expression and prevented growth inhibition and apoptosis in NB4 APL cells subsequently exposed to doxorubicin (DOX). ATRA/FK228 treatment after exposure to DOX, however, enhanced apoptosis. Both agents, ATRA or FK228, induced MDR1 mRNA. This effect was significantly enhanced by ATRA/FK228 administered in combination, due in part to increased H4 and H3-Lys9 acetylation of the MDR1 promoter and recruitment of the nuclear transcription factor Y alpha (NFYA) transcription activator to the CCAAT box. Cotreatment with specific P-gp inhibitor PSC833 reversed cytoprotective effects of ATRA/FK228. G1 cell-cycle arrest and p21 mRNA induction were also observed in response to ATRA/FK228, which may restrict DOX-induced apoptosis of cells in G2 phase. These results indicate that epigenetic mechanisms involving NF-YA transcription factor recruitment and histone acetylation are activated by ATRA and HDACI, induce MDR1 in APL cells, and point to the critical importance of mechanism-based sequential therapy in future clinical trials that combine HDAC inhibitors, ATRA, and anthracyclines.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Antibióticos Antineoplásicos/farmacología , Depsipéptidos/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/fisiología , Leucemia Promielocítica Aguda/genética , Ciclo Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Genes MDR , Inhibidores de Histona Desacetilasas , Humanos
10.
Cancer Res ; 65(21): 9962-70, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16267021

RESUMEN

Conditionally active forms of the Raf proteins (Raf-1, B-Raf, and A-Raf) were created by ligating NH2-terminal truncated activated forms (Delta) to the estrogen receptor (ER) hormone-binding domain resulting in estradiol-regulated constructs (DeltaRaf:ER). These different Raf:ER oncoproteins were introduced into the murine FDC-P1 hematopoietic cell line, and cells that grew in response to the three DeltaRaf:ER oncoproteins were isolated. The ability of FDC-P1, DeltaRaf-1:ER, DeltaA-Raf:ER, and DeltaB-Raf:ER cells to form tumors in severe combined immunodeficient mice was compared. Mice injected with DeltaRaf:ER cells were implanted with beta-estradiol pellets to induce the DeltaRaf:ER oncoprotein. Cytokine-dependent parental cell lines did not form tumors. Implantation of beta-estradiol pellets into mice injected with DeltaRaf:ER cells significantly accelerated tumor onset and tumor size. The recovered DeltaRaf:ER cells displayed induction of extracellular signal-regulated kinase (ERK) in response to beta-estradiol stimulation, indicating that they had retained conditional activation of ERK even when passed through a severe combined immunodeficient mouse. The DeltaRaf:ER cells were very sensitive to induction of apoptosis by the mitogen-activated protein/ERK kinase (MEK) 1 inhibitor CI1040 whereas parental cells were much less affected, demonstrating that the MEK1 may be useful in eliminating Ras/Raf/MEK-transformed cells. Furthermore, the effects of in vivo administration of the MEK1 inhibitor were evaluated and this inhibitor was observed to suppress the tumorigenicity of the injected cells. This DeltaRaf:ER system can serve as a preclinical model to evaluate the effects of signal transduction inhibitors which target the Raf and MEK proteins.


Asunto(s)
Transformación Celular Neoplásica/patología , Células Madre Hematopoyéticas/patología , Leucemia/patología , Quinasas raf/biosíntesis , Animales , Benzamidas/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Estradiol/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Leucemia/inducido químicamente , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Ratones , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores de Estrógenos/biosíntesis , Trasplante Heterólogo
11.
Cancer Res ; 65(7): 2890-8, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15805291

RESUMEN

We investigated the antileukemic activity and molecular mechanisms of action of a newly synthesized ring-substituted diindolylmethane derivative, 1,1-bis[3'-(5-methoxyindolyl)]-1-(p-t-butylphenyl) methane (DIM #34), in acute myelogenous leukemia (AML) cells. DIM #34 inhibited AML cell growth via the induction of apoptosis and abrogated clonogenic growth of primary AML samples. Exposure to DIM #34 induced loss of mitochondrial inner transmembrane potential, release of cytochrome c into the cytosol, and caspase activation. Bcl-2-overexpressing, Bax knockout, and caspase-9-deficient cells were partially resistant to cell death, suggesting the involvement of the intrinsic apoptotic pathway. Furthermore, DIM #34 transiently inhibited the phosphorylation and activity of the extracellular signal-regulated kinase and abrogated Bcl-2 phosphorylation. Because other methylene-substituted diindolylmethane analogues have been shown to transactivate the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma), we studied the role of PPARgamma in apoptosis induction. Cotreatment of cells with a selective PPARgamma antagonist or with retinoid X receptor and retinoic acid receptor ligands partially modulated apoptosis when combined with DIM #34, suggesting PPARgamma receptor-dependent and receptor-independent cell death. Together, these findings suggest that diindolylmethanes are a new class of compounds that selectively induce apoptosis in AML cells through the modulation of the extracellular signal-regulated kinase and PPARgamma signaling pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Indoles/farmacología , Leucemia Mieloide/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Enfermedad Aguda , Apoptosis/fisiología , Caspasas/metabolismo , Activación Enzimática/efectos de los fármacos , Células HL-60 , Humanos , Células Jurkat , Leucemia Monocítica Aguda/tratamiento farmacológico , Leucemia Monocítica Aguda/enzimología , Leucemia Monocítica Aguda/patología , Leucemia Mieloide/enzimología , Leucemia Mieloide/patología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Leucemia Mielomonocítica Aguda/tratamiento farmacológico , Leucemia Mielomonocítica Aguda/enzimología , Leucemia Mielomonocítica Aguda/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Células U937
12.
J Toxicol Environ Health A ; 67(1): 1-22, 2004 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-14668108

RESUMEN

In order to explore the potential of DNA methylation to serve as a biomarker of toxicity, thus establishing a link between exposure to environmental contaminants and physiologically significant changes in gene expression, tissue- and gender-specific methylation patterns in the promoter regions of estrogen receptor (ER) and aromatase genes of Japanese medaka (Oryzias latipes) were determined. Adult male and female medaka were exposed to either 0 or 500 ng/L 17 alpha-ethinylestradiol (EE) for 14d via a waterborne exposure. Livers, gonads, and brains were removed and genomic DNA was extracted. Samples of genomic DNA were then analyzed by bisulfite-mediated methylation-specific polymerase chain reaction (PCR) of an approximately 300-bp region containing suspected methylation sites from the two genes, amplified, cloned, and sequenced. ER protein content in exposed medaka was significantly induced in all male and female tissues compared to controls. Aromatase activity in exposed medaka was significantly increased in the male brain, testes, and female brain as compared to controls. The methylation changes described by these studies indicate the potential for anthropogenic alteration of the mechanisms controlling gene expression, as well as gender- and tissue-specific sensitivity. While methylation differences were not paralleled by changes in protein expression in this study, changes in methylation have the potential to impact the regulation of normal gene expression and these changes could be transmitted to offspring.


Asunto(s)
Aromatasa/efectos de los fármacos , Aromatasa/genética , Metilación de ADN/efectos de los fármacos , Monitoreo del Ambiente/métodos , Etinilestradiol/toxicidad , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Secuencia de Aminoácidos , Animales , Aromatasa/análisis , Biomarcadores/análisis , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Exposición a Riesgos Ambientales/efectos adversos , Exposición a Riesgos Ambientales/análisis , Monitoreo del Ambiente/normas , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Etinilestradiol/análisis , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Gónadas/química , Gónadas/efectos de los fármacos , Hígado/química , Hígado/efectos de los fármacos , Masculino , Datos de Secuencia Molecular , Oryzias , Reacción en Cadena de la Polimerasa , Receptores de Estrógenos/análisis , Caracteres Sexuales , Contaminantes Químicos del Agua/análisis , Contaminantes Químicos del Agua/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA