Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Pharmaceutics ; 13(11)2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34834374

RESUMEN

Bladder cancer is the 10th most diagnosed cancer, with almost 10 M cancer deaths last year worldwide. Currently, chemotherapy is widely used as adjuvant therapy after surgical transurethral resection. Paclitaxel (PTX) is one of the most promising drugs, but cancer cells acquire resistance, causing failure of this treatment and increasing the recurrence of the disease. This poor chemotherapeutic response has been associated with the overexpression of the protein survivin. In this work, we present a novel dual nano-treatment for bladder cancer based on the hypothesis that the inhibition of survivin in cancer cells, using a siRNA gene therapy strategy, could decrease their resistance to PTX. For this purpose, two different polymeric nanoparticles were developed to encapsulate PTX and survivin siRNA independently. PTX nanoparticles showed sizes around 150 nm, with a paclitaxel loading of around 1.5%, that produced sustained tumor cell death. In parallel, siRNA nanoparticles, with similar sizes and loading efficiency of around 100%, achieved the oligonucleotide transfection and knocking down of survivin expression that also resulted in tumor cell death. However, dual treatment did not show the synergistic effect expected. The root cause of this issue was found to be the cell cycle arrest produced by nuclear survivin silencing, which is incompatible with PTX action. Therefore, we concluded that although the vastly reported role of survivin in bladder cancer, its silencing does not sensitize cells to currently applied chemotherapies.

2.
J Neuromuscul Dis ; 8(5): 815-825, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34366365

RESUMEN

BACKGROUND: Duchenne Muscular Dystrophy (DMD) is one of the most common muscular dystrophies, caused by mutated forms of the dystrophin gene. Currently, the only treatment available is symptoms management. Novel approximations are trying to treat these patients with gene therapy, namely, using viral vectors. However, these vectors can be recognized by the immune system decreasing their therapeutic activity and making impossible a multidose treatment due to the induction of the humoral immunity following the first dose. OBJECTIVE: Our objective is to demonstrate the feasibility of using a hybrid vector to avoid immune clearance, based on the electrostatic coating of adeno-associated virus (AAVs) vectors with our proprietary polymers. METHODS: We coated model adeno-associated virus vectors by electrostatic interaction of our cationic poly (beta aminoester) polymers with the viral anionic capsid and characterized biophysical properties. Once the nanoformulations were designed, we studied their in vivo biodistribution by bioluminescence analysis and we finally studied the capacity of the polymers as potential coatings to avoid antibody neutralization. RESULTS: We tested two polymer combinations and we demonstrated the need for poly(ethylene glycol) addition to avoid vector aggregation after coating. In vivo biodistribution studies demonstrated that viral particles are located in the liver (short times) and also in muscles (long times), the target organ. However, we did not achieve complete antibody neutralization shielding using this electrostatic coating. CONCLUSIONS: The null hypothesis stands: although it is feasible to coat viral particles by electrostatic interaction with a proprietary polymer, this strategy is not appropriate for AAVs due to their small size, so other alternatives are required as a novel treatment for DMD patients.


Asunto(s)
Terapia Genética/métodos , Distrofia Muscular de Duchenne/inmunología , Electricidad Estática , Virión/inmunología , Animales , Dependovirus/inmunología , Distrofina/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Ratones , Distribución Tisular
3.
Theranostics ; 10(6): 2744-2758, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194832

RESUMEN

Oncolytic adenoviruses are used as agents for the treatment of cancer. However, their potential is limited due to the high seroprevalence of anti-adenovirus neutralizing antibodies (nAbs) within the population and the rapid liver sequestration when systemically administered. To overcome these challenges, we explored using nanoparticle formulation to boost the efficacy of systemic oncolytic adenovirus administration. Methods: Adenovirus were conjugated with PEGylated oligopeptide-modified poly(ß-amino ester)s (OM-pBAEs). The resulting coated viral formulation was characterized in terms of surface charge, size, aggregation state and morphology and tested for anti-adenovirus nAbs evasion and activity in cancer cells. In vivo pharmacokinetics, biodistribution, tumor targeting, and immunogenicity studies were performed. The antitumor efficacy of the oncolytic adenovirus AdNuPARmE1A coated with OM-pBAEs (SAG101) in the presence of nAbs was evaluated in pancreatic ductal adenocarcinoma (PDAC) mouse models. Toxicity of the coated formulation was analyzed in vivo in immunocompetent mice. Results: OM-pBAEs conjugated to adenovirus and generated discrete nanoparticles with a neutral charge and an optimal size. The polymeric coating with the reporter AdGFPLuc (CPEG) showed enhanced transduction and evasion of antibody neutralization in vitro. Moreover, systemic intravenous administration of the formulation showed improved blood circulation and reduced liver sequestration, substantially avoiding activation of nAb production. OM-pBAEs coating of the oncolytic adenovirus AdNuPARmE1A (SAG101) improved its oncolytic activity in vitro and enhanced antitumor efficacy in PDAC mouse models. The coated formulation protected virions from neutralization by nAbs, as antitumor efficacy was preserved in their presence but was completely lost in mice that received the non-formulated AdNuPARmE1A. Finally, coated-AdNuPARmE1A showed reduced toxicity when high doses of the formulation were administered. Conclusions: The developed technology represents a promising improvement for future clinical cancer therapy using oncolytic adenoviruses.


Asunto(s)
Antineoplásicos , Neoplasias/terapia , Oligopéptidos , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Polímeros , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/química , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Polímeros/química , Polímeros/farmacocinética , Polímeros/farmacología , Células RAW 264.7
4.
Adv Healthc Mater ; 8(19): e1900849, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31478348

RESUMEN

One of the main bottlenecks in the translation of nanomedicines from research to clinics is the difficulty in designing nanoparticles actively vectorized to the target tissue, a key parameter to ensure efficacy and safety. In this group, a library of poly(beta aminoester) polymers is developed, and it is demonstrated that adding specific combinations of terminal oligopeptides (OM-PBAE), in vitro transfection is cell selective. The current study aims to actively direct the nanoparticles to the liver by the addition of a targeting molecule. To achieve this objective, retinol, successfully attached to OM-PBAE, is selected as hepatic targeting moiety. It is demonstrated that organ biodistribution is tailored, achieving the desired liver accumulation. Regarding cell type transfection, antigen presenting cells in the liver are those showing the highest transfection. Thanks to proteomics studies, organ but not cellular biodistribution can be explained by the formation of differential protein coronas. Therefore, organ biodistribution is governed by differential protein corona formed when retinol is present, while cellular biodistribution is controlled by the end oligopeptides type. In summary, this work is a proof of concept that demonstrates the versatility of these OM-PBAE nanoparticles, in terms of the modification of the biodistribution of OM-PBAE nanoparticles adding active targeting moieties.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Polímeros/química , Corona de Proteínas/química , Animales , Supervivencia Celular , Citometría de Flujo , Proteínas Fluorescentes Verdes/química , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Nanomedicina , Oligonucleótidos Antisentido , Oligopéptidos/química , Tamaño de la Partícula , Proteómica , Células RAW 264.7 , Vitamina A/química
5.
Nanoscale ; 11(38): 17869-17877, 2019 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-31552987

RESUMEN

The future of gene therapy relies on the development of efficient and safe delivery vectors. Poly(ß-amino ester)s are promising cationic polymers capable of condensing oligonucleotides into nanoparticles - polyplexes - and deliver them into the cell nucleus, where the gene material would be expressed. The complexation state during the crossing of biological barriers is crucial: polymers should tightly complex DNA before internalization and then release to allow free DNA to reach the nucleus. However, measuring the complexation state in cells is challenging due to the nanometric size of polyplexes and the difficulties to study the two components (polymer and DNA) independently. Here we propose a method to visualize and quantify the two components of a polyplex inside cells, with nanometre scale resolution, using two-colour direct stochastic reconstruction super-resolution microscopy (dSTORM). With our approach, we tracked the complexation state of pBAE polyplexes from cell binding to DNA release and nuclear entry revealing time evolution and the final fate of DNA and pBAE polymers in mammalian cells.


Asunto(s)
Núcleo Celular/metabolismo , ADN/química , Nanopartículas/química , Oligonucleótidos/química , Polímeros/química , Transfección , Animales , Células COS , Chlorocebus aethiops , ADN/farmacología , Terapia Genética , Humanos , Oligonucleótidos/farmacología , Polímeros/farmacología
6.
Int J Pharm ; 553(1-2): 169-185, 2018 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-30321641

RESUMEN

A dual-targeting drug delivery system for paclitaxel (PTX) was developed by functionalizing novel polyester-based nanoparticles (NPs) with peptides possessing special affinity for low-density lipoprotein receptor (LDLR), overcoming the limitations of the current chemotherapeutics, to transport drug from blood to brain, and then target glioma cells. Employing novel biodegradable block co-polymers (P and 2P), PTX loaded and peptide-functionalized nanoparticles were prepared by a modified nano-co-precipitation method, carried out in one step only without emulsifier, allowing to obtain spherical nanometric (<200 nm), monodisperse (PDI ∼ 0.1), Poly (Ethylene Glycol) (PEG)-coated and high PTX loaded NPs with a slow and controlled release rate for a prolonged period of time. Peptide functionalization, confirmed by fluorimetric assay and HPLC amino acids analysis, enhanced the cellular uptake of functionalized-PTX-NPs by human primary glioblastoma cell line (U-87 MG) and Bovine Brain Endothelial Cells (BBMVECs), compared with non-functionalized-PTX-NPs. To confirm dual-targeting effect, transendothelial transport experiments in an in vitro BBB model and in vitro anti-tumoral activity against U-87 MG revealed that peptide-functionalized-PTX-NPs significantly increased the transport ratio of PTX across the BBB along with an improved anti-proliferative efficiency. Pharmacokinetics and biodistribution studies in rats, carried out by in vivo experiments with 125I radiolabelled dual-targeting PTX-NPs, confirmed the stealthy behavior of NPs and indicated slightly lower levels of penetration into brain tissue in comparison with peptides known to be able to cross the BBB. These promising results suggested that the dual-targeting drug delivery system might have great potential for glioma therapy in clinical applications.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Glioblastoma/tratamiento farmacológico , Paclitaxel/administración & dosificación , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/farmacología , Barrera Hematoencefálica , Neoplasias Encefálicas/patología , Bovinos , Línea Celular Tumoral , Células Cultivadas , Preparaciones de Acción Retardada , Células Endoteliales/metabolismo , Glioblastoma/patología , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Nanopartículas , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Péptidos/química , Polímeros/química , Ratas , Ratas Sprague-Dawley , Receptores de LDL/metabolismo , Distribución Tisular
7.
Adv Healthc Mater ; 7(17): e1800335, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29923337

RESUMEN

The encapsulation of mRNA in nanosystems as gene vaccines for immunotherapy purposes has experienced an exponential increase in recent years. Despite the many advantages envisaged within these approaches, their application in clinical treatments is still limited due to safety issues. These issues can be attributed, in part, to liver accumulation of most of the designed nanosystems and to the inability to transfect immune cells after an intravenous administration. In this context, this study takes advantage of the known versatile properties of the oligopeptide end-modified poly (ß-amino esters) (OM-PBAEs) to complex mRNA and form discrete nanoparticles. Importantly, it is demonstrated that the selection of the appropriate end-oligopeptide modifications enables the specific targeting and major transfection of antigen-presenting cells (APC) in vivo, after intravenous administration, thus enabling their use for immunotherapy strategies. Therefore, with this study, it can be confirmed that OM-PBAE are appropriate systems for the design of mRNA-based immunotherapy approaches aimed to in vivo transfect APCs and trigger immune responses to fight either tumors or infectious diseases.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , ARN Mensajero/administración & dosificación , ARN Mensajero/metabolismo , Animales , Línea Celular , Supervivencia Celular , Portadores de Fármacos/química , Células HeLa , Humanos , Inmunoterapia , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Polímeros/química , Células RAW 264.7
8.
Drug Deliv ; 25(1): 1137-1146, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29779394

RESUMEN

A dual doxorubicin/camptothecin (DOX/CPT) pH-triggered drug delivery mesoporous silica nanoparticle (MSN)-based nano-vehicle has been prepared. In this drug-delivery system (DDS), CPT is loaded inside the pores of the MSNs, while DOX is covalently attached to the surface of an aldehyde-functionalized MSN through a dihydrazide-polyethylene glycol chain. Thus, DOX and the linker act as pH-sensitive gatekeeper. The system is versatile and easy to assemble, not requiring the chemical modification of the drugs. While at physiological conditions the release of the drugs is negligible, at acidic pH a burst release of DOX and a gradual release of CPT take place. In vitro cytotoxicity tests have demonstrated that this DDS can deliver efficiently DOX and CPT for combination therapy.


Asunto(s)
Camptotecina/química , Doxorrubicina/química , Portadores de Fármacos/química , Nanopartículas/química , Dióxido de Silicio/química , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Excipientes/química , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Porosidad
9.
Drug Deliv ; 25(1): 472-483, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29412012

RESUMEN

Glioblastoma multiforme (GBM) is the most devastating primary brain tumor due to its infiltrating and diffuse growth characteristics, a situation compounded by the lack of effective treatments. Currently, many efforts are being devoted to find novel formulations to treat this disease, specifically in the nanomedicine field. However, due to the lack of comprehensive characterization that leads to insufficient data on reproducibility, only a reduced number of nanomedicines have reached clinical phases. In this context, the aim of the present study was to use a cascade of assays that evaluate from physical-chemical and structural properties to biological characteristics, both in vitro and in vivo, and also to check the performance of nanoparticles for glioma therapy. An amphiphilic block copolymer, composed of polyester and poly(ethylene glycol; PEG) blocks, has been synthesized. Using a mixture of this copolymer and a polymer containing an active targeting moiety to the Blood Brain Barrier (BBB; Seq12 peptide), biocompatible and biodegradable polymeric nanoparticles have been prepared and extensively characterized. In vitro studies demonstrated that nanoparticles are safe for normal cells but cytotoxic for cancer cells. In vivo studies in mice demonstrated the ability of the Seq12 peptide to cross the BBB. Finally, in vivo efficacy studies using a human tumor model in SCID mice resulted in a significant 50% life-span increase, as compared with non-treated animals. Altogether, this assay cascade provided extensive pre-clinical characterization of our polymeric nanoparticles, now ready for clinical evaluation.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Nanopartículas/administración & dosificación , Polímeros/administración & dosificación , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Bovinos , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos/métodos , Glioma/metabolismo , Glioma/patología , Humanos , Masculino , Ratones , Ratones SCID , Nanopartículas/metabolismo , Polímeros/metabolismo , Ratas , Ratas Wistar , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
11.
Acta Biomater ; 20: 82-93, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25839122

RESUMEN

Here we present an extended family of pBAEs that incorporate terminal oligopeptide moieties synthesized from both positive and negative amino acids. Polymer formulations of mixtures of negative and positive oligopeptide-modified pBAEs are capable of condensing siRNA into discrete nanoparticles. We have demonstrated that efficient delivery of nucleic acids in a cell-type dependent manner can be achieved by careful control of the pBAE formulation. In addition, our approach of adding differently charged oligopeptides to the termini of poly(ß-amino ester)s is of great interest for the design of tailored complexes having specific features, such as tuneable zeta potential. We anticipate that this surface charge tunability may be a powerful strategy to control unwanted electrostatic interactions, while preserving high silencing efficiency and reduced toxicity.


Asunto(s)
Silenciador del Gen , Oligopéptidos/química , Polímeros/química , Electricidad Estática , Supervivencia Celular , Dispersión Dinámica de Luz , Ensayo de Cambio de Movilidad Electroforética , Endocitosis , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Hidrodinámica , Nanopartículas/química , Oligopéptidos/síntesis química , Tamaño de la Partícula , Polímeros/síntesis química , ARN Interferente Pequeño/metabolismo , Propiedades de Superficie , Transfección
12.
J Mol Biol ; 426(20): 3467-77, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-24747049

RESUMEN

Neural stem cell (NSC) state and fate depend on spatially and temporally synchronized transcriptional and epigenetic regulation of the expression of extrinsic signaling factors and intrinsic cell-specific genes, but the functional roles for chromatin-modifying enzymes in neural differentiation remain poorly understood. Here we show that the histone demethylases KDM4A (JMJD2A) and KDM4C (JMJD2C) are essential for proper differentiation of NSCs in vitro and in vivo. KDM4A/C were required for neuronal differentiation, survival and expression of the neurotrophic signaling factor BDNF in association with promoter H3K9 demethylation and RNA polymerase II recruitment. Unexpectedly, KDM4A/C were essential for selective H3K36 demethylation and loss of RNA polymerase II recruitment in transcribed regions of the astrocyte-characteristic gene GFAP, thereby in parallel repressing astrocytic differentiation by control of elongation. We propose that gene- and lysine-specific KDM4A/C-mediated control of histone methylation and thereby regulation of intrinsic factors and signaling factors such as BDNF provide a novel control mechanism of lineage decision.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diferenciación Celular , Proteína Ácida Fibrilar de la Glía/metabolismo , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células-Madre Neurales/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Células Cultivadas , Regulación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Immunoblotting , Histona Demetilasas con Dominio de Jumonji/genética , Lisina/metabolismo , Metilación , Ratones , Microscopía Fluorescente , Células-Madre Neurales/citología , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Polimerasa II/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Acta Biomater ; 10(5): 2147-58, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24406199

RESUMEN

The main limitation of gene therapy towards clinics is the lack of robust, safe and efficient gene delivery vectors. This paper describes new polycations for gene delivery based on poly(ß-amino ester)s (pBAE) containing terminal oligopeptides. The authors developed oligopeptide-modified pBAE-pDNA nanoparticles that achieve better cellular viability and higher transfection efficacy than other end-modified pBAE and commercial transfection agents. Gene expression in highly permissive cell lines was remarkably high, but transfection efficiency in less-permissive cell lines was highly dependent on oligopeptide composition and nanoparticle formulation. Moreover, the use of selected oligopeptides in the pBAE formulation led to preferential intracellular localization of the particles. Particle analysis of highly efficient pBAE formulations revealed different particle sizes and charge features, which indicates chemical pseudotyping of the particle surface, related to the oligopeptide chemical nature. In conclusion, chemical modification at the termini of pBAE with amine-rich oligopeptides is a powerful strategy for developing delivery systems for future gene therapy applications.


Asunto(s)
Técnicas de Transferencia de Gen , Espacio Intracelular/metabolismo , Oligopéptidos/química , Polímeros/química , Secuencia de Aminoácidos , Animales , Línea Celular , Supervivencia Celular , Ensayo de Cambio de Movilidad Electroforética , Endocitosis , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Datos de Secuencia Molecular , Tamaño de la Partícula , Plásmidos/metabolismo , Polímeros/síntesis química , Protones , Análisis Espectral , Electricidad Estática , Transfección
14.
Mol Cancer Ther ; 10(3): 505-17, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21388975

RESUMEN

Gemcitabine is a nucleoside analogue with anticancer activity. Inside the cell, it is sequentially phosphorylated to generate the active drug. Phosphorylated nucleoside analogues have been shown to traffic through gap junctions. We investigated the participation of gap junctional intercellular communication (GJIC) as a possible mechanism spreading gemcitabine cytotoxicity in pancreatic tumors. Immunohistochemical analysis of pancreatic cancer biopsies revealed increased connexin 26 (Cx26) content but loss of connexins 32 (Cx32) and 43 (Cx43) expression. Cx26 abundance in neoplastic areas was confirmed by Cx26 mRNA in situ hybridization. Heterogeneity on the expression levels and the localization of Cx26, Cx32, and Cx43 were identified in pancreatic cancer cells and found to be associated with the extent of GJIC, and correlated with gemcitabine bystander cytotoxic effect. The abundance of Cx26 at the contact points in tumoral regions prompted us to study the involvement of Cx26 in the GJIC of gemcitabine toxic metabolites and their influence on the antitumoral effects of gemcitabine. Knockdown of Cx26 led to decreased GJIC and reduced gemcitabine bystander killing whereas overexpression of Cx26 triggered increased GJIC and enhanced the gemcitabine cytotoxic bystander effect. Gemcitabine treatment of mice bearing tumors, with a high GJIC capacity, resulted in a significant delay in tumor progression. Interestingly, gemcitabine administration in mice bearing tumors that overexpress Cx26 triggered a dramatic tumor regression of 50% from the initial volume. This study shows that Cx26 participates in the gap junction-mediated bystander cytoxic effect of gemcitabine and provides evidence that upregulation of Cx26 improves gemcitabine anticancer efficacy.


Asunto(s)
Efecto Espectador/fisiología , Conexinas/metabolismo , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/metabolismo , Animales , Comunicación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular , Conexina 26 , Conexina 43/genética , Conexinas/antagonistas & inhibidores , Conexinas/genética , Desoxicitidina/farmacología , Uniones Comunicantes/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Fosforilación , ARN Mensajero/genética , Gemcitabina , Proteína beta1 de Unión Comunicante
15.
J Biol Chem ; 285(5): 3462-9, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-19917601

RESUMEN

Proinsulin C-peptide is internalized into cells, but a function of its intracellular localization has not been established. We now demonstrate that, upon cellular entry, C-peptide is localized to the nucleoli, where it promotes transcription of genes encoding for ribosomal RNA. We find that C-peptide binds to histones and enhances acetylation of lysine residue 16 of histone H4 at the promoter region of genes for ribosomal RNA. In agreement with synchrony of ribosomal RNA synthesis and cell proliferation, we show that C-peptide stimulates proliferation in chondrocytes and HEK-293 cells. This regulation of ribosomal RNA provides a mechanism by which C-peptide can exert transcriptional effects and implies that the peptide has growth factor activity.


Asunto(s)
Péptido C/química , Regulación de la Expresión Génica , ARN Ribosómico/metabolismo , Células 3T3 , Animales , Ciclo Celular , Línea Celular , Nucléolo Celular/metabolismo , Proliferación Celular , Condrocitos/metabolismo , Epigénesis Genética , Histonas/química , Humanos , Ratones , Transcripción Genética
16.
Mol Cell Biol ; 29(7): 1814-25, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19158275

RESUMEN

The transcription factor p73, a member of the p53 family of proteins, is involved in the regulation of cell cycle progression and apoptosis. However, the regulatory mechanisms controlling the distinct roles for p73 in these two processes have remained unclear. Here, we report that p73 is able to induce cell cycle arrest independently of its amino-terminal transactivation domain, whereas this domain is crucial for p73 proapoptotic functions. We also characterized a second transactivation domain in the carboxy terminus of p73 within amino acid residues 381 to 399. This carboxy terminus transactivation domain was found to preferentially regulate genes involved in cell cycle progression. Moreover, its activity is regulated throughout the cell cycle and modified by protein kinase C-dependent phosphorylation at serine residue 388. Our results suggest that this novel posttranslational modification within the p73 carboxy terminus transactivation domain is involved in the context-specific guidance of p73 toward the selective induction of cell cycle arrest.


Asunto(s)
Ciclo Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteína Quinasa C/metabolismo , Activación Transcripcional/genética , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proteína Vmw65 de Virus del Herpes Simple/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Especificidad de Órganos , Fosforilación , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ratas , Serina/metabolismo , Relación Estructura-Actividad
17.
J Biol Chem ; 284(6): 3672-81, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19001364

RESUMEN

Bone morphogenetic proteins such as BMP4 are essential for proper development of telencephalic forebrain structures and induce differentiation of telencephalic neural stem cells into a variety of cellular fates, including astrocytic, neuronal, and mesenchymal cells. Little is yet understood regarding the mechanisms that underlie the spatiotemporal differences in progenitor response to BMP4. In a screen designed to identify novel targets of BMP4 signaling in telencephalic neural stem cells, we found the mRNA levels of the previously uncharacterized factor CXXC5 reproducibly up-regulated upon BMP4 stimulation. In vivo, CXXC5 expression overlapped with BMP4 adjacent to Wnt3a expression in the dorsal regions of the telencephalon, including the developing choroid plexus. CXXC5 showed partial homology with Idax, a related protein previously shown to interact with the Wnt-signaling intermediate Dishevelled (Dvl). Indeed CXXC5 and Dvl co-localized in the cytoplasm and interacted in co-immunoprecipitation experiments. Moreover, fluorescence resonance energy transfer (FRET) experiments verified that CXXC5 and Dvl2 were located in close spatial proximity in neural stem cells. Studies of the functional role of CXXC5 revealed that overexpression of CXXC5 or exposure to BMP4 repressed the levels of the canonical Wnt signaling target Axin2, and CXXC5 attenuated Wnt3a-mediated increase in TOPflash reporter activity. Accordingly, RNA interference of CXXC5 attenuated the BMP4-mediated decrease in Axin2 levels and facilitated the response to Wnt3a in neural stem cells. We propose that CXXC5 is acting as a BMP4-induced inhibitor of Wnt signaling in neural stem cells.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Telencéfalo/embriología , Factores de Transcripción/biosíntesis , Proteínas Wnt/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Células Cultivadas , Plexo Coroideo/citología , Plexo Coroideo/embriología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas Dishevelled , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Telencéfalo/citología , Regulación hacia Arriba/fisiología , Proteína Wnt3
18.
Hum Gene Ther ; 16(12): 1377-88, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16390269

RESUMEN

Suicide gene therapy using the herpes simplex virus thymidine kinase (TK) gene in combination with ganciclovir (GCV) has been shown to produce therapeutic, but limited, efficacy because of poor gene transfer efficiency and reduced bystander effect. Here we report that fusion of TK to an eight-amino acid peptide from the basic domain of the human immunodeficiency virus (HIV) Tat protein significantly increases the cytotoxic efficacy of the TK/GCV system in pancreatic cancer cells. We demonstrate that Tat8-TK protein is released from the intracellular compartment of Tat8-TK-expressing cells to the extracellular medium after GCV treatment. Interestingly, we show that this conditioned medium is then able to mediate cytotoxicity of wildtype cultures, suggesting the internalization of the Tat8-TK protein. Moreover, a strong antitumoral effect of Tat8-TK/GCV treatment could be achieved by two different in vivo approaches. Tumors injected with NIH 3T3/Tat8-TK cells attached to microcarriers (MC+Tat8-TK) and treated with GCV led to a 35.6% reduction in the initial tumor volume and to 50% tumor eradication. Furthermore, electrogene transfer of TK or Tat8-TK followed by administration of high doses of GCV led to an overall statistically significant reduction in tumor growth. However, the reduction in initial tumor volume was statistically significant only for the Tat8-TK group (59.5% reduction). Moreover, in this group 50% complete tumor eradication was achieved. When moderate doses of GCV were administered, the overall reduction in tumor growth was statistically significant only in the Tat8-TK group. Therefore, our results suggest that fusion of TK to the Tat8 peptide enhances TK/GCV suicide gene therapy.


Asunto(s)
Ganciclovir/uso terapéutico , Productos del Gen tat/genética , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias Pancreáticas/terapia , Timidina Quinasa/genética , Células 3T3 , Animales , Técnicas de Transferencia de Gen , Masculino , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Neoplasias Pancreáticas/genética , Timidina Quinasa/metabolismo , Transfección , Células Tumorales Cultivadas
19.
Clin Cancer Res ; 10(4): 1454-62, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14977849

RESUMEN

PURPOSE: Gene transfer of a truncated variant of the retinoblastoma (RB) gene encoding a M(r) 94000 protein that lacks the NH(2)-terminal 112 amino acid residues, termed RB94, has been shown to inhibit proliferation of several human tumor cell types. We have assessed its therapeutic effectiveness on pancreatic cancer, one of the most aggressive and therapy-resistant types of cancer. For this purpose, preclinical studies aimed to evaluate the therapeutic potential of RB94 gene transfer in pancreatic cancer were carried out. EXPERIMENTAL DESIGN: We have compared the antiproliferative effects of adenovirus-mediated gene transfer of RBwt and RB94 at the in vitro and in vivo levels in three RB-positive human pancreatic tumor cell lines: (a). NP-9; (b). NP-18; and (c). NP-31. We have also examined their effects on cell cycle and their capacity to induce apoptosis. RESULTS: In vitro results indicate that RB94 gene transfer has stronger antiproliferative effects compared with RBwt. RB94 transduction correlated with accumulation at the S-G(2) phase of the cell cycle in the three cell lines tested and induction of apoptosis in two of them. In vivo studies show significant decreases in the growth rate of tumors treated with Ad-RB94 when compared with those treated with Ad-RBwt. Moreover, terminal deoxynucleotidyl transferase-mediated nick end labeling analyses of Ad-RB94-treated tumor sections revealed that only RB94 is able to significantly induce apoptosis. CONCLUSIONS: RB94 gene expression has antiproliferative effects also in human pancreatic tumor cells, being more effective than wild-type RB in preventing tumor growth.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Neoplasias Pancreáticas/terapia , Proteína de Retinoblastoma/genética , Animales , Anexina A5/farmacología , Apoptosis , Western Blotting , Ciclo Celular , División Celular , Línea Celular Tumoral , Colorantes/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/genética , Estructura Terciaria de Proteína , Factores de Tiempo
20.
J Gene Med ; 4(2): 141-9, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11933215

RESUMEN

BACKGROUND: Pancreatic cancer is one of the most aggressive human tumors and the development of new therapeutic approaches is particularly urgent since current therapies are not effective. The use of pro-drug-activating genes is a possible approach for cancer gene therapy. METHODS: The present study evaluated the efficiency of the cytochrome P4502B1 (CYP2B1) suicide gene that encodes the enzyme responsible for activating the pro-drug cyclophosphamide (CPA), in pancreatic tumor cells invitro and in vivo. The effects on tumor growth of the combination of two suicide systems, CYP2B1/CPA and herpes simplex virus thymidine kinase gene/ganciclovir (HSVtk/GCV), were also studied. RESULTS: Retroviral CYP2B1 transfer followed by CPA treatment highly sensitized pancreatic tumor cells NP-9, NP-18, and NP-31, and led to stabilization of tumor growth in a pancreatic tumor model. Differences in tumor volume at the end of the treatment were statistically significant when compared with animals injected with CPA alone. The combination of both suicide systems CYP2B1/CPA and HSVtk/GCV in vitro resulted in a potentiation of the killing effect. However, no potentiation was achieved in vivo, although retardation in tumor growth was evident. CONCLUSIONS: The results show that in situ transduction of pancreatic tumor cells with the CYP2B1 gene by retroviral vectors clearly increases the sensitivity to CPA. Moreover, they suggest that in order to achieve a potentiation on cell killing when the two suicide systems HSVtk/GCV and CYP2B1/CPA are combined, co-expression of both genes in the same tumor cell would be necessary.


Asunto(s)
Ciclofosfamida/farmacología , Citocromo P-450 CYP2B1/metabolismo , Técnicas de Transferencia de Gen , Retroviridae/genética , Animales , Antineoplásicos Alquilantes/farmacología , Antivirales/farmacología , Diferenciación Celular , Medios de Cultivo Condicionados/farmacología , Relación Dosis-Respuesta a Droga , Ganciclovir/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Profármacos , Timidina Quinasa/genética , Factores de Tiempo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA