Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem J ; 477(17): 3131-3145, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32766732

RESUMEN

The Hedgehog-regulated transcription factors GLI1 and GLI2 play overlapping roles in development and disease; however, the mechanisms underlying their interplay remain elusive. We report for the first time that GLI1 and GLI2 physically and functionally interact in cancer cells. GLI1 and GLI2 were shown to co-immunoprecipitate in PANC1 pancreatic cancer cells and RMS13 rhabdomyosarcoma cells. Mapping analysis demonstrated that the zinc finger domains of both proteins are required for their heteromerization. RNAi knockdown of either GLI1 or GLI2 inhibited expression of many well-characterized GLI target genes (BCL2, MYCN, PTCH2, IL7 and CCND1) in PANC1 cells, whereas PTCH1 expression was only inhibited by GLI1 depletion. qPCR screening of a large set of putative canonical and non-canonical Hedgehog/GLI targets identified further genes (e.g. E2F1, BMP1, CDK2) strongly down-regulated by GLI1 and/or GLI2 depletion in PANC1 cells, and demonstrated that ANO1, AQP1 and SOCS1 are up-regulated by knockdown of either GLI1 or GLI2. Chromatin immunoprecipitation showed that GLI1 and GLI2 occupied the same regions at the BCL2, MYCN and CCND1 promoters. Furthermore, depletion of GLI1 inhibited GLI2 occupancy at these promoters, suggesting that GLI1/GLI2 interaction is required for the recruitment of GLI2 to these sites. Together, these findings indicate that GLI1 and GLI2 co-ordinately regulate the transcription of some genes, and provide mechanistic insight into the roles of GLI proteins in carcinogenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/metabolismo , Rabdomiosarcoma/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína Gli2 con Dedos de Zinc/metabolismo , Línea Celular Tumoral , Proteínas Hedgehog/genética , Humanos , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Multimerización de Proteína , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Proteína con Dedos de Zinc GLI1/genética , Proteína Gli2 con Dedos de Zinc/genética
3.
Dev Cell ; 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38834071

RESUMEN

Normal cells coordinate proliferation and differentiation by precise tuning of gene expression based on the dynamic shifts of the epigenome throughout the developmental timeline. Although non-mutational epigenetic reprogramming is an emerging hallmark of cancer, the epigenomic shifts that occur during the transition from normal to malignant cells remain elusive. Here, we capture the epigenomic changes that occur during tumorigenesis in a prototypic embryonal brain tumor, medulloblastoma. By comparing the epigenomes of the different stages of transforming cells in mice, we identify nuclear factor I family of transcription factors, known to be cell fate determinants in development, as oncogenic regulators in the epigenomes of precancerous and cancerous cells. Furthermore, genetic and pharmacological inhibition of NFIB validated a crucial role of this transcription factor by disrupting the cancer epigenome in medulloblastoma. Thus, this study exemplifies how epigenomic changes contribute to tumorigenesis via non-mutational mechanisms involving developmental transcription factors.

4.
J Biol Chem ; 287(22): 17996-8004, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22496449

RESUMEN

The melanocyte-specific transcription factor M-MITF is involved in numerous aspects of melanoblast lineage biology including pigmentation, survival, and migration. It plays complex roles at all stages of melanoma progression and metastasis. We established previously that GLI2, a Kruppel-like transcription factor that acts downstream of Hedgehog signaling, is a direct transcriptional target of the TGF-ß/SMAD pathway and contributes to melanoma progression, exerting antagonistic activities against M-MITF to control melanoma cell invasiveness. Herein, we dissected the molecular mechanisms underlying both TGF-ß and GLI2-driven M-MITF gene repression. Using transient cell transfection experiments with M-MITF promoter constructs, chromatin immunoprecipitation, site-directed mutagenesis, and electrophoretic mobility shift assays, we identified a GLI2 binding site within the -334/-296 region of the M-MITF promoter, critical for GLI2-driven transcriptional repression. This region is, however, not needed for inhibition of M-MITF promoter activity by TGF-ß. We determined that TGF-ß rapidly repressed protein kinase A activity, thus reducing both phospho-cAMP-response element-binding protein (CREB) levels and CREB-dependent transcription of the M-MITF promoter. Increased GLI2 binding to its cognate cis-element, associated with reduced CREB-dependent transcription, allowed maximal inhibition of the M-MITF promoter via two distinct mechanisms.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/fisiología , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Proteínas Nucleares/fisiología , Factor de Crecimiento Transformador beta/fisiología , Secuencia de Bases , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cartilla de ADN , ADN de Neoplasias/genética , Progresión de la Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica , Humanos , Factor de Transcripción Asociado a Microftalmía/fisiología , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Transcripción Genética , Proteína Gli2 con Dedos de Zinc
5.
Nat Mater ; 9(2): 172-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20010827

RESUMEN

In the domain of health, one important challenge is the efficient delivery of drugs in the body using non-toxic nanocarriers. Most of the existing carrier materials show poor drug loading (usually less than 5 wt% of the transported drug versus the carrier material) and/or rapid release of the proportion of the drug that is simply adsorbed (or anchored) at the external surface of the nanocarrier. In this context, porous hybrid solids, with the ability to tune their structures and porosities for better drug interactions and high loadings, are well suited to serve as nanocarriers for delivery and imaging applications. Here we show that specific non-toxic porous iron(III)-based metal-organic frameworks with engineered cores and surfaces, as well as imaging properties, function as superior nanocarriers for efficient controlled delivery of challenging antitumoural and retroviral drugs (that is, busulfan, azidothymidine triphosphate, doxorubicin or cidofovir) against cancer and AIDS. In addition to their high loadings, they also potentially associate therapeutics and diagnostics, thus opening the way for theranostics, or personalized patient treatments.


Asunto(s)
Diagnóstico por Imagen , Portadores de Fármacos/química , Nanoestructuras/química , Compuestos Organometálicos/química , Animales , Línea Celular Tumoral , Medios de Contraste/química , Medios de Contraste/metabolismo , Medios de Contraste/toxicidad , Portadores de Fármacos/metabolismo , Portadores de Fármacos/toxicidad , Femenino , Humanos , Macrófagos/efectos de los fármacos , Imagen por Resonancia Magnética , Ratones , Compuestos Organometálicos/metabolismo , Compuestos Organometálicos/toxicidad , Tamaño de la Partícula , Porosidad , Ratas
6.
Mol Cancer ; 9: 103, 2010 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-20459741

RESUMEN

BACKGROUND: Aberrant expression of cyclin D1 is a common feature in multiple myeloma (MM) and always associated with mantle cell lymphoma (MCL). CCND1 gene is alternatively spliced to produce two cyclin D1 mRNA isoforms which are translated in two proteins: cyclin D1a and cyclin D1b. Both isoforms are present in MM cell lines and primary cells but their relative role in the tumorigenic process is still elusive. RESULTS: To test the tumorigenic potential of cyclin D1b in vivo, we generated cell clones derived from the non-CCND1 expressing MM LP-1 cell line, synthesizing either cyclin D1b or cyclin K, a structural homolog and viral oncogenic form of cyclin D1a. Immunocompromised mice injected s.c. with LP-1K or LP-1D1b cells develop tumors at the site of injection. Genome-wide analysis of LP-1-derived cells indicated that several cellular processes were altered by cyclin D1b and/or cyclin K expression such as cell metabolism, signal transduction, regulation of transcription and translation. Importantly, cyclin K and cyclin D1b have no major action on cell cycle or apoptosis regulatory genes. Moreover, they impact differently cell functions. Cyclin K-expressing cells have lost their migration properties and display enhanced clonogenic capacities. Cyclin D1b promotes tumorigenesis through the stimulation of angiogenesis. CONCLUSIONS: Our study indicates that cyclin D1b participates into MM pathogenesis via previously unrevealed actions.


Asunto(s)
Ciclina D1/metabolismo , Ciclinas/metabolismo , Mieloma Múltiple/metabolismo , Animales , Ciclo Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Separación Celular , Embrión de Pollo , Ciclina D1/genética , Ciclinas/genética , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Desnudos , Mieloma Múltiple/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Breast Cancer Res Treat ; 122(1): 145-58, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19771505

RESUMEN

The suppression of oestrogen receptor alpha (ERalpha) functions by silencing RNAs in association with or not with anti-oestrogens (AEs) both in vitro and in breast cancer cell xenografts was assessed. In vitro, a prolonged decrease in ERalpha protein expression and an enhanced AE-induced inhibition of ERalpha-mediated transcription, together with antiproliferative activity, were observed. Incorporation of ERalpha-siRNAs in pegylated nanocapsules (NC) was achieved; and their intravenous injections in MCF-7 xenografts, in contrast to scramble siRNA containing NCs, lead to decrease in ERalpha protein content and Ki67 labelling in tumour cells. The pure AE RU58668 (RU) both free and entrapped in stealth nanospheres (NS) at very low concentration (8 microg/kg/week) had no effect on tumour growth evolution. However, coinjection of the two nanocarriers potentiated the decrease in ERalpha protein, concomitantly with decreasing tumour vasculature and glucose transporter-1. These data support that the targeted delivery of ERalpha-siRNA in breast tumours potentiates the inhibition of E(2)-induced proliferative activity by encapsulated AE through enhanced anti-vascular activity. In the hormone-independent MDA-MB-231 xenograft model, RU-NS at 4 mg/kg/week induce also a strong tumour vascular normalisation. Together, these findings suggest that the anti-oestrogen activity of RU as well as that of targeted ERalpha-siRNA leads to anti-angiogenic activity. Their delivery in stealth nanocarriers may constitute a new anti-cancer therapeutic strategy in solid tumours.


Asunto(s)
Adenocarcinoma/patología , Neoplasias de la Mama/patología , Estradiol/análogos & derivados , Moduladores de los Receptores de Estrógeno/uso terapéutico , Receptor alfa de Estrógeno/antagonistas & inhibidores , Estrógenos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Nanocápsulas/administración & dosificación , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Hormono-Dependientes/patología , ARN Interferente Pequeño/uso terapéutico , Animales , División Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/trasplante , Sinergismo Farmacológico , Estradiol/farmacología , Estradiol/uso terapéutico , Moduladores de los Receptores de Estrógeno/farmacología , Receptor alfa de Estrógeno/biosíntesis , Receptor alfa de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inyecciones Intravenosas , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Nanocápsulas/química , Nanosferas/administración & dosificación , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Poliésteres , Polietilenglicoles , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , Organismos Libres de Patógenos Específicos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Pharm Res ; 27(2): 327-39, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20033476

RESUMEN

PURPOSE: To determine the better liposomal formulation incorporating the active metabolite of tamoxifen, 4-hydroxy-tamoxifen (4HT) and the biological impact of 4HT-pH-gradient liposomes on response to in vivo treatment. METHODS: Several pegylated liposomes were formulated by varying the composition of lipids, increasing external pH from 7.4 to 9.0 and doubling the lipid concentration. Dipalmitoylphosphatidylcholine / cholesterol / distearoylphosphoethanolamine poly(ethylene)glycol liposomes (DL-9 liposomes) were chosen for their physico-chemical properties. Toxicity and release kinetics were assessed in breast cancer MCF-7 as well as in multiple myeloma (MM) cells. In vivo antitumor activity and bio-distribution were measured in the RPMI8226 MM model. RESULTS: Compared to conventional non-pH-gradient liposomes, 4HT-DL-9 liposomes resulted in concentration of up to 1 mM 4HT, greater stability, relative toxicity and slow 4HT release. Intravenous injections of 4HT-DL-9 liposomes at 4 mg/kg/week blocked MM tumor growth. Ki67 and CD34 labeling decreased in treated tumors, concomitantly with increase of activated caspase-3 supporting a cell proliferation arrest, a decrease of tumor vasculature and the induction of tumor cell death. CONCLUSION: This antitumor effect was assumed to be the result of a modified biodistribution of 4HT once trapped in DL-9 liposomes. Such 4HT-containing pH-gradient Stealth nanocarriers could be helpful for MM treatment.


Asunto(s)
Modelos Animales de Enfermedad , Mieloma Múltiple/tratamiento farmacológico , Fuerza Protón-Motriz/efectos de los fármacos , Tamoxifeno/análogos & derivados , Animales , Línea Celular Tumoral , Femenino , Humanos , Liposomas , Ratones , Ratones Desnudos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Fuerza Protón-Motriz/fisiología , Tamoxifeno/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Sci Rep ; 10(1): 14491, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32879407

RESUMEN

GLI1 expression is broadly accepted as a marker of Hedgehog pathway activation in tumors. Efficacy of Hedgehog inhibitors is essentially limited to tumors bearing activating mutations of the pathway. GLI2, a critical Hedgehog effector, is necessary for GLI1 expression and is a direct transcriptional target of TGF-ß/SMAD signaling. We examined the expression correlations of GLI1/2 with TGFB and HH genes in 152 distinct transcriptome datasets totaling over 23,500 patients and representing 37 types of neoplasms. Their prognostic value was measured in over 15,000 clinically annotated tumor samples from 26 tumor types. In most tumor types, GLI1 and GLI2 follow a similar pattern of expression and are equally correlated with HH and TGFB genes. However, GLI1/2 broadly share prognostic value with TGFB genes and a mesenchymal/EMT signature, not with HH genes. Our results provide a likely explanation for the frequent failure of anti-Hedgehog therapies in tumors, as they suggest a key role for TGF-ß, not Hedgehog, ligands, in tumors with elevated GLI1/2-expression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/genética , Neoplasias/diagnóstico , Proteínas Nucleares/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína Gli2 con Dedos de Zinc/genética , Biología Computacional , Perfilación de la Expresión Génica , Humanos , Ligandos , Análisis Multivariante , Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Modelos de Riesgos Proporcionales , Factores de Riesgo , Transducción de Señal/genética , Transcriptoma
10.
J Dermatol Sci ; 94(3): 321-329, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31208857

RESUMEN

BACKGROUND: Tyrosinase-Related Protein 2 (TRP2) is an enzyme involved in melanogenesis, that also exerts proliferative, anti-apoptotic and immunogenic functions in melanoma cells. TRP2 transcription is regulated by the melanocytic master transcription factor MITF. GLI2, a transcription factor that acts downstream of Hedgehog signaling, is also a direct transcriptional target of the TGF-ß/SMAD pathway that contributes to melanoma progression and exerts transcriptional antagonistic activities against MITF. OBJECTIVES: To characterize the molecular events responsible for TGF-ß and GLI2 repression of TRP2 expression. METHODS: In silico promoter analysis, transient cell transfection experiments with 5'-end TRP2 promoter deletion constructs, chromatin immuno-precipitation, and site-directed promoter mutagenesis were used to dissect the molecular mechanisms of TRP2 gene regulation by TGF-ß and GLI2. RESULTS: We demonstrate that TGF-ß and GLI2-specific TRP2 repression involves direct mechanisms that occur in addition to MITF downregulation by TGF-ß and GLI2. We identify two functional GLI2 binding sites within the TRP2 promoter that are critical for TGF-ß and GLI2 responsiveness, one of them overlapping a CREB binding site. GLI2 and CREB competing for the same cis-element is associated with opposite transcriptional outcome. CONCLUSION: Our results further refine the understanding of how TGF-ß and GLI2 control the phenotypic plasticity of melanoma cells. In particular, we identify critical GLI2-binding cis-elements within the TRP2 promoter region that allow for its transcriptional repression independently from MITF concomitant downregulation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Oxidorreductasas Intramoleculares/genética , Melanoma/genética , Proteínas Nucleares/metabolismo , Neoplasias Cutáneas/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteína Gli2 con Dedos de Zinc/metabolismo , Línea Celular Tumoral , Biología Computacional , Humanos , Melanoma/patología , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas/genética , Neoplasias Cutáneas/patología , Transcripción Genética
11.
Int J Cancer ; 122(9): 2130-41, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18183592

RESUMEN

Multiple myeloma (MM) is a malignancy characterized by the accumulation of tumoral plasma cells in bone marrow. This disease remains incurable and the development of new therapeutic strategies is urgently required. We have studied the effects of 2 selective estrogen receptor disrupters (SERDs), RU 58668 (RU) and ICI 182,780 (ICI) or pure antiestrogens (AEs) on MM cell lines. Both compounds have antimyeloma activity through either cell cycle arrest or induction of apoptosis. To analyze the molecular mechanisms of SERD action, we choose 2 differently responding cell lines as models. In LP-1 cells, RU blocked cell cycle at the G1 phase. RU treatment induced a rapid decrease of c-Myc, an upregulation of p27(Kip1), and the subsequent decreased activity of cyclin-dependent kinase, CDK6 and associated cyclin D3, impairing the inactivation of the retinoblastoma protein (pRb). In RPMI 8226 cells, RU induced apoptosis by recruiting endoplasmic reticulum- as well as mitochondria-associated caspases. Moreover, RU interfered with the NF-kappaB survival pathway, often deregulated in MM malignancy. Antimyeloma activities were observed in dexamethasone (Dex)- and RU-resistant cells when RU was combined with bortezomib; Dex and bortezomib being frequently used in MM therapy. RU induced the death of CD138+ cells purified from MM patients but not CD19+ normal cells obtained from tonsils. Therefore, RU mediates the inhibition of survival, the activation of apoptosis and finally potentiates anticancer drug. Those combinatory effects provide a basis for the potential use of pure AEs in MM treatment.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Caspasas/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Ciclinas/metabolismo , Retículo Endoplásmico/metabolismo , Estradiol/análogos & derivados , Moduladores de los Receptores de Estrógeno/farmacología , Fase G1/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/farmacología , Western Blotting , Bortezomib , Proliferación Celular/efectos de los fármacos , Colorimetría , Ciclina D3 , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Estradiol/farmacología , Citometría de Flujo , Humanos , Inmunoprecipitación , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Transducción de Señal , Células Tumorales Cultivadas
12.
Biomacromolecules ; 9(10): 2881-90, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18788777

RESUMEN

Specific siRNAs that target estrogen receptor alpha (ERalpha) were encapsulated in nanocapsules (NCs). We produced small (approximately 100-200 nm) ERalpha-siRNA NCs with a water core by incorporating two mixed duplexes of specific ERalpha-siRNAs (ERalpha-mix-siRNA) into NCs. The encapsulation yield that was obtained with poly(iso-butylcyanoacrylate) (PIBCA) NCs was low, whereas no release of trapped siRNA was observed for poly(ethylene)glycol-poly(D,L-lactide-co-glycolide) (PEG-PLGA) NCs. High levels of ERalpha-siRNA incorporation into PEG-epsilon-caprolactone-malic acid (PEG-PCL/MA) NCs (3.3 microM in a polymer solution at 16 mg/mL) were observed (72% yield). No difference in size or zeta potential was observed between siRNA NCs that were based on PEG-PCL/MA and empty NCs. Fluorescence quenching assays confirmed the incorporation of siRNA into the NC core. A persistent loss of ERalpha (90% over 5 days) was observed in MCF-7 human breast cancer cells that were exposed to PEG-PCL/MA NCs that were loaded with ERalpha-siRNA. The intravenous injection of these NCs into estradiol-stimulated MCF-7 cell xenografts led to a significant decrease in tumor growth and a decrease in ERalpha expression in tumor cells. These data indicate that a novel strategy, based on ERalpha-siRNA delivery, could be developed for the treatment of hormone-dependent breast cancers.


Asunto(s)
Materiales Biocompatibles/química , Química Física/métodos , Receptor alfa de Estrógeno/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Emulsiones , Femenino , Humanos , Ensayo de Materiales , Ratones , Ratones Desnudos , Nanocápsulas/química , Trasplante de Neoplasias
13.
Bioorg Med Chem Lett ; 18(7): 2495-8, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18304811

RESUMEN

A new series of coumarin inhibitors of hsp90 lacking the noviose moiety as well as substituents on C-7 and C-8 positions of the aromatic ring was synthesised and their hsp90 inhibitory activity has been delineated: for example, their capacity to induce the degradation of client proteins and to inhibit estradiol-induced transcription in human breast cancer cells. In cell proliferation assay, the most active compound 5g was approximately 8 times more potent than the parent novobiocin natural compound.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Cumarinas/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Novobiocina/farmacología , Antibióticos Antineoplásicos/síntesis química , Sitios de Unión , Neoplasias de la Mama/patología , Línea Celular Tumoral/efectos de los fármacos , Cumarinas/química , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Estradiol/farmacología , Humanos , Novobiocina/síntesis química , Relación Estructura-Actividad , Transcripción Genética/efectos de los fármacos
14.
Int J Pharm ; 347(1-2): 128-35, 2008 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-17643877

RESUMEN

For the first time, two organometallic triphenylethylene compounds (Fc-diOH and DFO), with strong antiproliferative activity in breast cancer cells, but insoluble in biological fluids, were incorporated in two types of stealth nanoparticles (NP): PEG/PLA nanospheres (NS) and nanocapsules (NC). Their physicochemical parameters were measured (size, zeta potential, encapsulation and loading efficiency), and their biological activity was assessed. In vitro drug release after high dilution of loaded NPs was measured by estradiol binding competition in MELN cells. The influence of the encapsulated drugs on the cell cycle and apoptosis was studied by flow cytometry analyses. Notwithstanding potential drug adsorption at the NP surface, Fc-diOH and DFO were incorporated efficiently in NC and NS, which slowly released both compounds. They arrested the cell cycle in the S-phase and induced apoptosis, whose activity is increased by loaded NS. A decrease in their antiproliferative activity by the antioxidant alpha-tocopherol indicated that reactive oxygen species (ROS) may be involved. Therefore, nanosystems, containing for the first time a high load of anticancer organometallic triphenylethylenes, have been developed. Their small size and delayed drug release, combined with their enhanced apoptotic potential, are compatible with an increased persistence in the blood and a promising antitumour activity.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos Ferrosos/química , Nanopartículas/química , Tamoxifeno/farmacología , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Estradiol/química , Estradiol/farmacología , Antagonistas de Estrógenos/química , Antagonistas de Estrógenos/farmacología , Femenino , Humanos , Metalocenos , Estructura Molecular , Nanocápsulas/química , Nanosferas/química , Tamaño de la Partícula , Receptores de Estrógenos/antagonistas & inhibidores , Electricidad Estática , Propiedades de Superficie , Tamoxifeno/química , alfa-Tocoferol/química , alfa-Tocoferol/farmacología
15.
J Med Chem ; 50(24): 6189-200, 2007 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-17979263

RESUMEN

Selective hsp90 inhibitors simultaneously destabilize and deplete key signaling proteins involved in cell proliferation and survival, angiogenesis, and metastasis. Investigation of novobiocin analogues lacking the noviose moiety as novel inhibitors of hsp90 was carried out. A novel series of 3-aminocoumarin analogues has been produced and screened in cell proliferation, and the molecular signature of hsp90 inhibition was assessed by depletion of estrogen receptor, HER2, Raf-1, and cdk4 in human breast cancer cells. This structure-activity relationship study highlights the crucial role of the C-4 and/or C-7 positions of coumarin which appeared to be essential for degradation of hsp90 client proteins. Removal of the noviose moiety in novobiocin together with introduction of a tosyl substituent at C-4 or C-7 coumarins provides 6e and 6f as lead structures which compared favorably with novobiocin as demonstrated by enhanced rates of cell death. The processing and activation of caspases 7 and 8 and the subsequent cleavage of PARP by 6e suggest stimulation of the extrinsic apoptosis pathway.


Asunto(s)
Antineoplásicos/síntesis química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Novobiocina/análogos & derivados , Novobiocina/síntesis química , Antineoplásicos/farmacología , Apoptosis , Neoplasias de la Mama , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Citometría de Flujo , Humanos , Novobiocina/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Transcripción Genética/efectos de los fármacos
16.
J Drug Target ; 15(2): 146-53, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17365286

RESUMEN

The folate receptor (FR) is a highly selective tumor marker over expressed in many human cancers and it constitutes a useful target for tumor-specific drug delivery. Thus, the conjugation of folic acid to different drugs or drug carriers may enhance the delivery of the therapeutic agent to FR-positive tumor cells. The aim of this study was to investigate the interactions of folate-conjugated polyalkylcyanoacrylate nanoparticles with tumor cells overexpressing the FR. For this purpose, nanoparticles were prepared by nanoprecipitation of the poly[aminopoly(ethylene glycol) cyanoacrylate-co-hexadecyl cyanoacrylate] [poly(H2NPEGCA-co-HDCA)] copolymer and labeled with the hydrophobic fluorescent dye nile red. Nile red-loaded nanoparticles were then conjugated to folic acid via the PEG terminal amino groups. Four human cancer cell lines were then tested by western blot in order to evaluate the FR expression levels. KB3-1 cell line showed the higher expression level, while MCF-7 cells were taken as a control. After measuring the cytotoxicity of the nanoparticles on these two cell lines, fluorescent folate-nanoparticles were incubated with them and the cellular uptake was evaluated by confocal microscopy and flow cytometry. KB3-1 cells showed a greater nanoparticle internalization, when compared to MCF-7 cells.


Asunto(s)
Cianoacrilatos/química , Ácido Fólico/química , Modelos Biológicos , Nanopartículas , Western Blotting , Línea Celular Tumoral , Citometría de Flujo , Humanos , Microscopía Confocal , Microscopía Fluorescente
17.
Int J Pharm ; 344(1-2): 71-7, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17651931

RESUMEN

The aim of this study was to develop both a physical and a chemical protection of the anticancer drug gemcitabine, which suffers from a rapid plasmatic metabolization. For this purpose, we used a series of lipophilic derivatives of gemcitabine in which an acyl chain is covalently coupled to the 4-amino group of gemcitabine; moreover, a physical protection of the drug was attempted by incorporating these lipophilic derivatives into poly(H(2)NPEGCA-co-HDCA) nanospheres and nanocapsules. Nanoparticles were prepared by nanoprecipitation of the poly(H(2)NPEGCA-co-HDCA) copolymer and their size, zeta potential and encapsulation efficiency were further characterized. These results have been relied on lipophilicity and flexibility studies. Data showed that only the more lipophilic derivative, 4-(N)-stearoylgemcitabine, was incorporated with a high yield. Thus, 4-(N)-stearoylgemcitabine-containing nanospheres and nanocapsules were further analyzed by differential scanning calorimetry. Their cytotoxicity was tested on two human cancer cell lines and compared to that of gemcitabine and free 4-(N)-stearoylgemcitabine.


Asunto(s)
Antimetabolitos Antineoplásicos/química , Cianoacrilatos , Desoxicitidina/análogos & derivados , Nanocápsulas , Nanosferas , Antimetabolitos Antineoplásicos/farmacología , Rastreo Diferencial de Calorimetría , Línea Celular Tumoral , Desoxicitidina/química , Desoxicitidina/farmacología , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Humanos , Polímeros , Profármacos/química , Relación Estructura-Actividad , Gemcitabina
18.
J Steroid Biochem Mol Biol ; 102(1-5): 114-27, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17056251

RESUMEN

Anti-oestrogens (AEs) are currently used for treating hormone-dependent breast cancers. They specifically bind to oestrogen receptors (ERs) and inhibit their transactivation capacity. However, ERs are present in various other tissues in which AEs may have either a beneficial or detrimental action. AE administration via systems targeting breast tumours may be an important therapeutic improvement. Thus, several biodegradable drug delivery systems containing either "mixed" (4-hydroxytamoxifen - 4-HT) or "pure" (RU 58668 - RU) AEs were prepared. Liposomes and nanospheres (NS, composed of non-toxic and biodegradable lipids and poly(d,l-lactic acid) incorporated up to 1 and 0.5 mM AE, respectively. Nanocapsules (NCs) in which an oily core solubilises the AE incorporated no more than 0.02 mM of the drug. PEG-functionalised nanoparticles survived longer in plasma and had better controlled release of the drug. The small size of the vectors (100-250 nm) was compatible with their extravasation through the discontinuous endothelium of tumour vasculature, allowing their accumulation in MCF-7 cell xenografts and leading to a prolonged exposure of the tumour to AEs. In these tumours and in MCF-7/ras xenografts, RU-NS and RU-NC (6.5mg/kg/week and 0.27 mg/kg/week, respectively, doses at which free RU had a very weak effect), both inhibited tumour growth. Entrapped RU significantly induced involution of tumours and strongly induced apoptosis in tumour cells, concomitantly with inhibiting tumour angiogenesis. 4-HT-nanoparticles also arrest oestradiol-induced tumour growth, inducing apoptosis and inhibiting angiogenesis. However, unlike RU-nanoparticles, they did not promote ERalpha subtype loss in tumour cells. Subcutaneous administration of both RU- and 4-HT-NS in MCF-7 xenografts strongly arrested tumour growth for prolonged periods and RUNS decreased the number of tumour epithelial cells. Analysis of the proteins involved in cell cycle proliferation and apoptosis confirmed that RU-nanoparticles were more efficient than 4-HT-nanoparticles. Their lack of toxicity and high anti-tumour potency that affects only tumour cells in the xenograft models mean these AE-loaded colloidal systems are a breakthrough in hormone-dependent breast cancer treatment.


Asunto(s)
Coloides , Sistemas de Liberación de Medicamentos , Moduladores de los Receptores de Estrógeno/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Nanocápsulas , Receptores de Estrógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Nanopartículas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Steroid Biochem Mol Biol ; 100(1-3): 67-78, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16753295

RESUMEN

In most of multiple myeloma (MM) cells, the "pure" antiestrogen (AE) RU 58668 (RU) induced either a G1-arrest (LP-1, OPM-2, NCI-H929, U266 cells) or apoptosis (RPMI 8226 cells). In RPMI 8226 cells, RU activates a caspase-dependent cell death pathway leading to the release of cytochrome c, the decrease of the essential MM survival factor Mcl-1, the cleavage of Bid and the activation of caspases-3 and -8. Incorporation of RU in pegylated cholesterol-containing liposomes allowed a controlled RU release, improving its anti-proliferative and apoptotic effects in cells. In RPMI 8226 xenografts, i.v. injected RU-liposomes but not free RU, exhibited antitumor activity. In vivo, RU-liposomes triggered the mitochondrial death pathway, concomitantly with a down-regulation of Mcl-1 and Bid cleavage. The decrease of CD34 immunoreactivity indicated a reduction of angiogenesis. The decrease of VEGF secretion in vitro supported a direct effect of RU on angiogenesis. These pro-apoptotic and antiangiogenic effects were explained by a prolonged exposure to the drug and to the endocytosis capacity of liposomes which might increase RU uptake and bypass a membrane export of free RU. Thus, these combined enhanced activities of RU-liposomes support that such a delivery of an AE may constitute a strategy of benefit for MM treatment.


Asunto(s)
Antineoplásicos/farmacología , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Liposomas , Mieloma Múltiple/tratamiento farmacológico , Animales , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Preparaciones de Acción Retardada , Sistemas de Liberación de Medicamentos/métodos , Estradiol/aislamiento & purificación , Estradiol/farmacología , Antagonistas de Estrógenos/aislamiento & purificación , Moduladores de los Receptores de Estrógeno/administración & dosificación , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Modelos Biológicos , Mieloma Múltiple/patología , Tonsila Palatina/citología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Clin Cancer Res ; 11(6): 2345-54, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15788686

RESUMEN

PURPOSE: Multiple myeloma is an incurable B-cell malignancy requiring new therapeutic strategies. Our approach was to analyze the in vitro effects of a selective estrogen receptor modulator, 4-hydroxytamoxifen (4-OHT), on six multiple myeloma cell lines. EXPERIMENTAL DESIGN: Cultured multiple myeloma cells were treated with various 4-OHT concentrations and the cellular response was studied: cell proliferation, cell viability, induction of apoptosis, caspase activities, and expression of signaling proteins. RESULTS: We found that pharmacologic concentrations of 4-OHT inhibit cell proliferation (4 of 6 cell lines). This inhibition is achieved by two independent events: a block at the G(1) phase of the cell cycle and the induction of apoptotic death. The cellular response to 4-OHT depends on the presence of functional estrogen receptors. 4-OHT treatment activates an intrinsic mitochondrial caspase-9-dependent pathway but not the Fas/FasL death pathway. Signaling pathways known to be involved in the survival and/or proliferation of multiple myeloma cells are not affected by 4-OHT treatment. 4-OHT-induced G(1) arrest is accompanied by the up-regulation of the cell cycle inhibitor p27(Kip1) and the down-regulation of c-Myc. Among the Bcl-2 family members tested, the proapoptotic BimS protein is induced whereas the antiapoptotic protein Mcl-1 is decreased. CONCLUSIONS: Although the effects of 4-OHT are observed at micromolar concentrations, cellular mechanisms responsible for G(1) arrest, as well as apoptosis induction, are similar to those observed in breast cancer cells. Our data support the concept that 4-OHT may represent an alternative approach to inhibit proliferation and induce apoptosis of multiple myeloma cells.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Mieloma Múltiple/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Proteínas Supresoras de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 9 , Caspasas/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Regulación hacia Abajo , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mieloma Múltiple/patología , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA