Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hum Cell ; 34(3): 808-818, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33616869

RESUMEN

Sevoflurane could stimulate neurotoxicity and result in postoperative cognitive dysfunction (POCD). Long non-coding RNAs (lncRNAs) have been implicated in the regulation of nervous system disease. This study was performed to investigate role and mechanism of lncRNA Rian (RNA imprinted and accumulated in nucleus) in sevoflurane anesthesia-induced cognitive dysfunction. Mice post-sevoflurane anesthesia showed cognitive impairments and neuronal damage and apoptosis. However, intracerebroventricularly injection with Adenovirus (Ad) for the over-expression of Rian ameliorated sevoflurane-induced neuronal damage and apoptosis. Cognitive impairments induced by sevoflurane were attenuated by injection with Ad-Rian. Moreover, transfection with Ad-Rian also protected isolated primary hippocampal neurons against sevoflurane-induced decrease of cell viability and increase of lactic acid dehydrogenase (LDH) and apoptosis. Mechanistically, Rian bind to miR-143-3p, and decreased expression of LIMK1 (Lim kinase 1) through negative regulation of miR-143-3p. Knockdown of LIMK1 aggravated sevoflurane-induced decrease of cell viability and increase of LDH and apoptosis in neurons, while over-expression attenuated LIMK1 silence-induced neuronal damage post-sevoflurane anesthesia. In conclusion, Rian demonstrated neuroprotective effects against sevoflurane anesthesia-induced cognitive dysfunction through regulation of miR-143-3p/LIMK1 axis, providing promising target for sevoflurane anesthesia-induced cognitive dysfunction.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Quinasas Lim/metabolismo , MicroARNs/metabolismo , Fármacos Neuroprotectores , Proteínas Nucleares/farmacología , Proteínas Nucleares/fisiología , Complicaciones Cognitivas Postoperatorias/tratamiento farmacológico , Complicaciones Cognitivas Postoperatorias/genética , Sevoflurano/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Hipocampo/citología , L-Lactato Deshidrogenasa/metabolismo , Quinasas Lim/genética , Ratones , MicroARNs/genética , Neuronas/metabolismo , Neuronas/fisiología , Proteínas Nucleares/administración & dosificación , Complicaciones Cognitivas Postoperatorias/inducido químicamente
2.
Hum Exp Toxicol ; 40(7): 1095-1111, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33375867

RESUMEN

Sestrin2 (SESN2) is stress-inducible protein that confers cytoprotective effects against various noxious stimuli. Accumulating evidence has documented that SESN2 has potent anti-apoptosis and anti-oxidative stress functions. However, whether it provides neuroprotection in traumatic brain injury (TBI) models remains unexplored. The purpose of this study was to explore the regulatory effect of SESN2 on TBI using in vivo and in vitro models. We found that TBI resulted in a marked induction of SESN2 in the cerebral cortex tissues of mice. SESN2 overexpression in the brain by in vivo gene transfer significantly decreased neurological deficit, brain edema, and neuronal apoptosis of mice with TBI. Moreover, the overexpression of SESN2 significantly decreased the oxidative stress induced by TBI in mice. In vitro studies of TBI demonstrated that SESN2 overexpression decreased apoptosis and oxidative stress in scratch-injured cortical neurons. Notably, SESN2 overexpression increased the nuclear levels of nuclear factor-erythroid 2-related factor 2 (Nrf2) and enhanced the activation of Nrf2 antioxidant signaling in in vivo and in vitro models of TBI. In addition, the inhibition of Nrf2 significantly abolished SESN2-mediated neuroprotective effects in vivo and in vitro. In conclusion, these results of our work demonstrate that SESN2 protects against TBI by enhancing the activation of Nrf2 antioxidant signaling.


Asunto(s)
Apoptosis/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/prevención & control , Factor 2 Relacionado con NF-E2/administración & dosificación , Fármacos Neuroprotectores , Proteínas Nucleares/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Células Cultivadas/efectos de los fármacos , Humanos , Masculino , Ratones , Modelos Animales
3.
Nat Cell Biol ; 22(10): 1276-1285, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33005030

RESUMEN

Breast cancer brain metastasis (BCBM) is a devastating disease. Radiation therapy remains the mainstay for treatment of this disease. Unfortunately, its efficacy is limited by the dose that can be safely applied. One promising approach to overcoming this limitation is to sensitize BCBMs to radiation by inhibiting their ability to repair DNA damage. Here, we report a DNA repair suppressor, leucine-rich repeat-containing protein 31 (LRRC31), that was identified through a genome-wide CRISPR screen. We found that overexpression of LRRC31 suppresses DNA repair and sensitizes BCBMs to radiation. Mechanistically, LRRC31 interacts with Ku70/Ku80 and the ataxia telangiectasia mutated and RAD3-related (ATR) at the protein level, resulting in inhibition of DNA-dependent protein kinase, catalytic subunit (DNA-PKcs) recruitment and activation, and disruption of the MutS homologue 2 (MSH2)-ATR module. We demonstrate that targeted delivery of the LRRC31 gene via nanoparticles improves the survival of tumour-bearing mice after irradiation. Collectively, our study suggests LRRC31 as a major DNA repair suppressor that can be targeted for cancer radiosensitizing therapy.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Neoplasias de la Mama/radioterapia , Daño del ADN , Reparación del ADN , Proteínas Nucleares/metabolismo , Fármacos Sensibilizantes a Radiaciones/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Rayos gamma , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Nucleares/administración & dosificación , Fosforilación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Nanomedicine (Lond) ; 13(4): 353-372, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29338617

RESUMEN

AIM: To investigate anticancer activity of the DNA binding domain of SMAR1 (His 5) in vitro and in vivo. MATERIALS & METHODS: His 5 was conjugated to hydrothermally synthesized carbon nanospheres (CNs). Anticancer activity of CNs-His 5 was evaluated in vitro and in vivo. RESULTS: CNs- His 5 significantly reduced cyclin D1 levels in MDA-MB-231 cells. Tumor bearing Balb/c mice injected with CNs-His 5 showed approximately 62% tumor regression and significantly reduced 18FDG uptake. Caspases assay and IHC staining confirmed tumor growth inhibition, which could be attributed to apoptotic, antiproliferative and antiangiogenic activities of His 5. CONCLUSION: DNA binding domain of the SMAR1 protein (His 5) has potent anticancer activity and its CNs mediated delivery could control breast tumor in mice model.


Asunto(s)
Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Carbono/química , Proteínas de Ciclo Celular/administración & dosificación , Proteínas de Unión al ADN/administración & dosificación , Portadores de Fármacos/química , Nanosferas/química , Proteínas Nucleares/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclina D1/metabolismo , Proteínas de Unión al ADN/metabolismo , Liberación de Fármacos , Femenino , Humanos , Ratones Endogámicos BALB C , Proteínas Nucleares/metabolismo , Dominios Proteicos , Proteínas Recombinantes/administración & dosificación , Distribución Tisular
5.
J Cereb Blood Flow Metab ; 37(4): 1447-1460, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27381825

RESUMEN

Hypoxic-ischemic encephalopathy is a condition caused by reduced oxygen and cerebral blood flow to the brain resulting in neurological impairments. Effective therapeutic treatments to ameliorate these disabilities are still lacking. We sought to investigate the role of sestrin2, a highly conserved stress-inducible protein, in a neonatal rat hypoxic-ischemic encephalopathy model. Ten-day-old rat pups underwent right common carotid artery ligation followed by 2.5 h hypoxia. At 1 h post hypoxic-ischemic encephalopathy, rats were intranasally administered with recombinant human sestrin2 and sacrificed for brain infarct area measurement, Fluoro-Jade C, immunofluorescence staining, Western blot, and neurological function testing. rh-sestrin2 reduced brain infarct area, brain atrophy, apoptosis, ventricular area enlargement, and improved neurological function. Western blot showed that sestrin2 expression levels were increased after treatment with rh-sestrin2, and sestrin2 exerts neuroprotective effects via activation of the adenosine monophosphate-activated protein kinase pathway which in turn inhibits mammalian target of rapamycin signaling resulting in the attenuation of apoptosis. In conclusions: Sestrin2 plays an important neuroprotective role after hypoxic-ischemic encephalopathy via adenosine monophosphate-activated protein kinase signaling pathway and serves as a negative feedback regulator of mammalian target of rapamycin. Administration of rh-sestrin2 not only reduced infarct area and brain atrophy, but also significantly improved neurological function.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Retroalimentación Fisiológica/fisiología , Hipoxia-Isquemia Encefálica/metabolismo , Proteínas Nucleares/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/genética , Proteínas Nucleares/uso terapéutico , Fosforilación , ARN Interferente Pequeño/genética , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
6.
Sci Rep ; 6: 30626, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27464702

RESUMEN

Potential tumor suppressor p42, ErbB3-binding protein 1 (EBP1) inhibits phosphoinositide 3-kinase (PI3K) activity reducing the p85 regulatory subunit. In this study, we demonstrated that overexpression of p42 promoted not only a reduction of wild type of p85 subunit but also oncogenic mutant forms of p85 which were identified in human cancers. Moreover, we identified the small fragment of C-terminal domain of p42 is sufficient to exhibit tumor suppressing activity of p42-WT, revealing that this small fragment (280-394) of p42 is required for the binding of both HSP70 and CHIP for a degradation of p85. Furthermore, we showed the small fragment of p42 markedly inhibited the tumor growth in mouse xenograft models of brain and breast cancer, resembling tumor suppressing activity of p42. Through identification of the smallest fragment of p42 that is responsible for its tumor suppressor activity, our findings represent a novel approach for targeted therapy of cancers that overexpress PI3K.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Regulación hacia Abajo , Proteínas de Unión al ARN/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Proteínas del Choque Térmico HSP72/metabolismo , Humanos , Ratones , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/metabolismo , Unión Proteica , Resultado del Tratamiento , Ubiquitina-Proteína Ligasas/metabolismo
7.
Int J Nanomedicine ; 11: 2039-51, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27274234

RESUMEN

Owing to the suppression of immune responses and associated side effects, steroid based treatments for inflammatory encephalitis disease can be detrimental. Here, we demonstrate a novel carbon nanosphere (CNP) based treatment regime for encephalomyelitis in mice by exploiting the functional property of the nuclear matrix binding protein SMAR1. A truncated part of SMAR1 ie, the DNA binding domain was conjugated with hydrothermally synthesized CNPs. When administered intravenously, the conjugate suppressed experimental animal encephalomyelitis in T cell specific conditional SMAR1 knockout mice (SMAR(-/-)). Further, CNP-SMAR1 conjugate delayed the onset of the disease and reduced the demyelination significantly. There was a significant decrease in the production of IL-17 after re-stimulation with MOG. Altogether, our findings suggest a potential carbon nanomaterial based therapeutic intervention to combat Th17 mediated autoimmune diseases including experimental autoimmune encephalomyelitis.


Asunto(s)
Carbono/química , Proteínas de Ciclo Celular/administración & dosificación , Proteínas de Ciclo Celular/uso terapéutico , Proteínas de Unión al ADN/administración & dosificación , Proteínas de Unión al ADN/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Nanosferas/química , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/uso terapéutico , Animales , Diferenciación Celular/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/patología , Endocitosis/efectos de los fármacos , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Nanosferas/toxicidad , Nanosferas/ultraestructura , Factor de Transcripción STAT3/metabolismo , Células Th17/citología , Células Th17/efectos de los fármacos
9.
Oncotarget ; 6(22): 19148-62, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25895131

RESUMEN

Many factors regulate cancer cell apoptosis, among which Survivin has a strong anti-apoptotic effect and PHLPP is a tumor suppressor gene that can induce significant apoptosis. However, the relationship between PHLPP and Survivin in gallbladder carcinoma (GBC) has not been reported. This study found that PHLPP expression is decreased and Survivin expression is increased in GBC tissues and cell lines. Their expression levels showed an inverse relationship and were associated with poor prognosis of GBC patients. Loss of PHLPP can increase the level of phosphorylated Survivin and induce the nuclear export of Survivin, which thus inhibit cell apoptosis and promote cell proliferation in GBC cells. The process that PHLPP regulates Survivin phosphorylation and intracellular localization is involved in AKT activity. Re-overexpression of PHLPP in GBC cells can decrease AKT phosphorylation level. Reduced expression of PHLPP in GBC is associated with high expression of miR-495. Increasing PHLPP expression or inhibiting miR-495 expression can induce apoptosis and suppress tumor growth in GBC xenograft model in nude mice. The results revealed the role and mechanism of PHLPP and Survivin in GBC cells and proposed strategies for gene therapies targeting the miR-495 / PHLPP / AKT / Survivin regulatory pathway.


Asunto(s)
Neoplasias de la Vesícula Biliar/terapia , Terapia Genética/métodos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/administración & dosificación , Fosfoproteínas Fosfatasas/genética , Transporte Activo de Núcleo Celular , Adulto , Anciano , Animales , Apoptosis/genética , Proliferación Celular/genética , Femenino , Neoplasias de la Vesícula Biliar/enzimología , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/patología , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Fosforilación , Pronóstico , Distribución Aleatoria , Survivin , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Eur J Pharm Biopharm ; 91: 91-102, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25660910

RESUMEN

The aim of the present study was to develop a novel strategy to deliver intracellularly the peptide GSE24.2 for the treatment of Dyskeratosis congenita (DC) and other defective telomerase disorders. For this purpose, biodegradable polymeric nanoparticles using poly(lactic-co-glycolic acid) (PLGA NPs) or poly(lactic-co-glycolic acid)-poly ethylene glycol (PLGA-PEG NPs) attached to either polycations or cell-penetrating peptides (CPPs) were prepared in order to increase their cellular uptake. The particles exhibited an adequate size and zeta potential, with good peptide loading and a biphasic pattern obtained in the in vitro release assay, showing an initial burst release and a later sustained release. GSE24.2 structural integrity after encapsulation was assessed using SDS-PAGE, revealing an unaltered peptide after the NPs elaboration. According to the cytotoxicity results, cell viability was not affected by uncoated polymeric NPs, but the incorporation of surface modifiers slightly decreased the viability of cells. The intracellular uptake exhibited a remarkable improvement of the internalization, when the NPs were conjugated to the CPPs. Finally, the bioactivity, addressed by measuring DNA damage rescue and telomerase reactivation, showed that some formulations had the lowest cytotoxicity and highest biological activity. These results proved that GSE24.2-loaded NPs could be delivered to cells, and therefore, become an effective approach for the treatment of DC and other defective telomerase syndromes.


Asunto(s)
Materiales Biocompatibles/química , Proteínas de Ciclo Celular/química , Sistemas de Liberación de Medicamentos , Reactivadores Enzimáticos/química , Nanopartículas/química , Proteínas Nucleares/química , Fragmentos de Péptidos/química , Animales , Materiales Biocompatibles/efectos adversos , Transporte Biológico , Proteínas de Ciclo Celular/administración & dosificación , Proteínas de Ciclo Celular/efectos adversos , Proteínas de Ciclo Celular/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Péptidos de Penetración Celular/efectos adversos , Péptidos de Penetración Celular/química , Células Cultivadas , Fenómenos Químicos , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/efectos adversos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/uso terapéutico , Composición de Medicamentos , Sistemas de Liberación de Medicamentos/efectos adversos , Liberación de Fármacos , Estabilidad de Medicamentos , Disqueratosis Congénita/tratamiento farmacológico , Reactivadores Enzimáticos/administración & dosificación , Reactivadores Enzimáticos/efectos adversos , Reactivadores Enzimáticos/uso terapéutico , Humanos , Ácido Láctico/efectos adversos , Ácido Láctico/química , Ratones , Nanopartículas/efectos adversos , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/efectos adversos , Proteínas Nucleares/genética , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/genética , Poliaminas/efectos adversos , Poliaminas/química , Polielectrolitos , Polietilenglicoles/efectos adversos , Polietilenglicoles/química , Poliglactina 910/efectos adversos , Poliglactina 910/química , Ácido Poliglicólico/efectos adversos , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Estabilidad Proteica , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapéutico
11.
J Control Release ; 202: 14-20, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25626085

RESUMEN

The modification of liposomal surfaces is of interest for many different applications and a variety of chemistries are available that makes this possible. A major disadvantage of commonly used coupling chemistries (e.g. maleimide-thiol coupling) is the limited control over the site of conjugation in cases where multiple reactive functionalities are present, leading to heterogeneous products and in some cases dysfunctional conjugates. Bioorthogonal coupling approaches such as the well-established copper-catalyzed azide-alkyne cycloaddition (CuAAC) "click" reaction are attractive alternatives as the reaction kinetics are favorable and azide-containing reagents are widely available. In the work described here, we prepared lipids containing a reactive cyclooctyne group and, after incorporation into liposomes, demonstrated successful conjugation of both a small molecule dye (5'-TAMRA-azide) as well as a larger azide-containing model protein based upon a designed ankyrin repeat protein (azido-DARPin). By applying the strain-promoted azido-alkyne cycloaddition (SPAAC) the use of Cu(I) as a catalyst is avoided, an important advantage considering the known deleterious effects associated with copper in cell and protein studies. We demonstrate complete control over the number of ligands coupled per liposome when using a small molecule azide with conjugation occurring at a reasonable reaction rate. By comparison, the conjugation of a larger azide-modified protein occurs more slowly, however the number of protein ligands coupled was found to be sufficient for liposome targeting to cells. Importantly, these results provide a strong proof of concept for the site-specific conjugation of protein ligands to liposomal surfaces via SPAAC. Unlike conventional approaches, this strategy provides for the homogeneous coupling of proteins bearing a single site-specific azide modification and eliminates the chance of forming dysfunctional ligands on the liposome. Furthermore, the absence of copper in the reaction process should also make this approach much more compatible with cell-based and in vivo applications.


Asunto(s)
Azidas/química , Compuestos Bicíclicos con Puentes/química , Colorantes/química , Liposomas/química , Proteínas Nucleares/química , Rodaminas/química , Repetición de Anquirina , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Colesterol/química , Química Clic , Cobre , Molécula de Adhesión Celular Epitelial , Células HT29 , Humanos , Proteínas Nucleares/administración & dosificación , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Polietilenglicoles/química
12.
IUBMB Life ; 64(4): 331-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22362485

RESUMEN

Several reports demonstrated that mesenchymal stem cells (MSCs) might differentiate into smooth muscle cells (SMCs) in vitro and in vivo. It has been shown that myocardin protein is a strong inducer of smooth muscle genes and MSCs can differentiate into SMCs in response to transforming growth factor-ß (TGF-ß). However, the relationship or link between myocardin and TGF-ß3-induced MSC differentiation has not been fully elucidated. Here, we demonstrated that both myocardin and TGF-ß3 were able to induce differentiation of rat bone marrow-derived MSCs toward smooth-muscle-like cell types, as evidenced by increasing expression of SMC-specific genes. Of note, myocardin cooperated with Smad2 to synergistically activate SM22α promoter and significantly enhance the expression of SM22α. Report assays with site-direct mutation analysis of SM22α promoter demonstrated that myocardin and Smad2 coactivated SM22α promoter mainly depending on CArG box and less on smad binding elements (SBE) sites as well. These findings reveal the cooperation of myocardin and Smad2 in process of MSC differentiation into SMCs.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Proteína Smad2/metabolismo , Transactivadores/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cartilla de ADN/genética , Sinergismo Farmacológico , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutagénesis Sitio-Dirigida , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteína Smad2/genética , Transactivadores/administración & dosificación , Transactivadores/genética , Factor de Crecimiento Transformador beta3/administración & dosificación , Regulación hacia Arriba/efectos de los fármacos
13.
J Immunol ; 186(7): 4164-74, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21389254

RESUMEN

Tolerance induction toward allogeneic organ grafts represents one of the major aims of transplantation medicine. Stem cells are promising candidates for promoting donor-specific tolerance. In this study, we investigated the immunomodulatory properties of murine embryonic stem cells (ESCs), obtained either by in vitro fertilization (IVF-ESCs) or by nuclear transfer (NT-ESCs), in heart transplant mouse models. IVF-ESCs did not prolong the survival of fully allogeneic cardiac transplants but significantly prolonged the survival of semiallogeneic hearts from the same ESC donor strain for >100 d in 44% of the animals. However, 28% of transplanted animals infused with IVF-ESCs experienced development of a teratoma. NT-ESCs similarly prolonged semiallogeneic heart graft survival (>100 d in 40% of the animals) but were less teratogenic. By in vitro studies, IVF-ESC and NT-ESC immunoregulation was mediated both by cell contact-dependent mechanisms and by the release of soluble factors. By adding specific inhibitors, we identified PGE(2) as a soluble mediator of ESC immunoregulation. Expansion of regulatory T cells was found in lymphoid organs and in the grafts of IVF-ESC- and NT-ESC-tolerized mice. Our study demonstrates that both IVF-ESCs and NT-ESCs modulate recipient immune response toward tolerance to solid organ transplantation, and that NT-ESCs exhibit a lower tendency for teratoma formation. Because NT-ESCs are obtained by NT of a somatic cell from living individuals into an enucleated oocyte, they could represent a source of donor-derived stem cells to induce tolerance to solid organ allograft.


Asunto(s)
Células Madre Embrionarias/inmunología , Células Madre Embrionarias/trasplante , Fertilización In Vitro , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Proteínas Nucleares/administración & dosificación , Traslado Adoptivo , Animales , Línea Celular , Femenino , Rechazo de Injerto/prevención & control , Trasplante de Corazón/métodos , Trasplante de Corazón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Distribución Aleatoria , Trasplante Homólogo/inmunología , Trasplante Homólogo/patología
14.
PLoS One ; 6(1): e16465, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21304583

RESUMEN

Numerous Gram negative pathogens possess a type III secretion system (T3SS) which allows them to inject virulent proteins directly into the eukaryotic cell cytoplasm. Injection of these proteins is dependent on a variable secretion signal sequence. In this study, we utilized the N-terminal secretion signal sequence of Pseudomonas aeruginosa exotoxin ExoS to translocate Cre recombinase containing a nuclear localization sequence (Cre-NLS). Transient exposure of human sarcoma cell line, containing Cre-dependent lacZ reporter, resulted in efficient recombination in the host chromosome, indicating that the bacterially delivered protein was not only efficiently localized to the nucleus but also retained its biological function. Using this system, we also illustrate the ability of P. aeruginosa to infect mouse embryonic stem cells (mESC) and the susceptibility of these cells to bacterially delivered Cre-NLS. A single two-hour infection caused as high as 30% of the mESC reporter cells to undergo loxP mediated chromosomal DNA recombination. A simple antibiotic treatment completely eliminated the bacterial cells following the delivery, while the use of an engineered mutant strain greatly reduced cytotoxicity. Utility of the system was demonstrated by delivery of the Cre-NLS to induced pluripotent stem cells to excise the floxed oncogenic nuclear reprogramming cassette. These results validate the use of T3SS for the delivery of transcription factors for the purpose of cellular reprogramming.


Asunto(s)
Bacterias/metabolismo , Reprogramación Celular , Sistemas de Liberación de Medicamentos/métodos , Proteínas Nucleares/administración & dosificación , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/administración & dosificación , ADP Ribosa Transferasas/administración & dosificación , ADP Ribosa Transferasas/farmacocinética , Animales , Sistemas de Secreción Bacterianos , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/farmacocinética , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Células Cultivadas , Humanos , Integrasas , Ratones , Señales de Localización Nuclear , Proteínas Nucleares/farmacocinética , Pseudomonas aeruginosa/química , Recombinación Genética , Factores de Transcripción/farmacocinética
15.
Jpn J Clin Oncol ; 40(12): 1184-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20656693

RESUMEN

The skin toxicity of vaccine therapy at injection sites is generally limited to Grades 1-2 due to the nature of their function. We experienced two cases of severe and prolonged local adverse effects in 25 patients following a Phase I study of gemcitabine and Wilms tumor-1 peptide vaccine mixed with incomplete Freund's adjuvant for inoperable pancreatic or biliary tract cancer. These patients requested to continue the treatment after the study period; however, in the course of compassionate use, they developed unacceptable local skin reactions and terminated their vaccine treatment. One patient (human leukocyte antigen, A0201, 3 mg) developed Grade 3 ulceration at the 10th vaccination and another (human leukocyte antigen, A2402, 1 mg) developed Grade 2 indulation and fibrosis at the 16th vaccination. Skin toxicity occurred at 6.4-8.4 months and continued for several months after the final vaccination during gemcitabine treatment. In these cases, activation or induction of Wilms tumor-1-specific T lymphocytes was not apparent in the peripheral blood despite their severe local reactions. Therefore, we need to monitor patients for late-onset, severe and long-lasting skin reactions at injection sites in Wilms tumor-1 cancer vaccine therapy, particularly for combination treatment with gemcitabine.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Desoxicitidina/análogos & derivados , Dermatitis/etiología , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/efectos adversos , Piel/efectos de los fármacos , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/secundario , Anciano , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/efectos adversos , Neoplasias Óseas/radioterapia , Neoplasias Óseas/secundario , Vacunas contra el Cáncer/inmunología , Proteínas de Ciclo Celular , Quimioterapia Adyuvante , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/secundario , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Dermatitis/inmunología , Esquema de Medicación , Femenino , Neoplasias de la Vesícula Biliar/patología , Humanos , Inyecciones Intradérmicas/efectos adversos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Linfocitos/inmunología , Proteínas Nucleares/inmunología , Cuidados Paliativos/métodos , Factores de Empalme de ARN , Radioterapia Adyuvante , Piel/inmunología , Gemcitabina
16.
Proc Natl Acad Sci U S A ; 105(43): 16719-24, 2008 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-18940925

RESUMEN

Hec1 (Highly Expressed in Cancer 1) is one of four proteins of the outer kinetochore Ndc80 complex involved in the dynamic interface between centromeres and spindle microtubules. Its overexpression is seen in a variety of human tumors and correlates with tumor grade and prognosis. We show here that the overexpression of Hec1 in an inducible mouse model results in mitotic checkpoint hyperactivation. As previously observed with overexpression of the Mad2 gene, hyperactivation of the mitotic checkpoint leads to aneuploidy in vitro and is sufficient to generate tumors in vivo that harbor significant levels of aneuploidy. These results underscore the role of chromosomal instability as a result of mitotic checkpoint hyperactivation in the initiation of tumorigenesis.


Asunto(s)
Proteínas de Ciclo Celular/farmacología , Mitosis , Neoplasias/etiología , Proteínas Nucleares/genética , Proteínas Nucleares/farmacología , Aneuploidia , Animales , Proteínas de Ciclo Celular/administración & dosificación , Proteínas de Ciclo Celular/genética , Inestabilidad Cromosómica , Doxiciclina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Cinetocoros , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos , Neoplasias/genética , Proteínas Nucleares/administración & dosificación , Distribución Tisular
17.
Blood ; 112(6): 2474-83, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18574026

RESUMEN

Nucleophosmin (NPM) is frequently overexpressed in leukemias and other tumors. NPM has been reported to suppress oncogene-induced senescence and apoptosis and may represent a therapeutic target for cancer. We fused a NPM-derived peptide to the HIV-TAT (TAT-NPMDeltaC) and found that the fusion peptide inhibited proliferation and induced apoptotic death of primary fibroblasts and preleukemic stem cells. TAT-NPMDeltaC down-regulated several NF-kappaB-controlled survival and inflammatory proteins and suppressed NF-kappaB-driven reporter gene activities. Using an inflammation-associated leukemia model, we demonstrate that TAT-NPMDeltaC induced proliferative suppression and apoptosis of preleukemic stem cells and significantly delayed leukemic development in mice. Mechanistically, TAT-NPMDeltaC associated with wild-type NPM proteins and formed complexes with endogenous NPM and p65 at promoters of several antiapoptotic and inflammatory genes and abrogated their transactivation by NF-kappaB in leu-kemic cells. Thus, TAT-delivered NPM peptide may provide a novel therapy for inflammation-associated tumors that require NF-kappaB signaling for survival.


Asunto(s)
Apoptosis/efectos de los fármacos , Productos del Gen tat/uso terapéutico , Leucemia/tratamiento farmacológico , Proteínas Nucleares/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Inflamación , Leucemia/patología , Ratones , FN-kappa B/antagonistas & inhibidores , Células Madre Neoplásicas/patología , Nucleofosmina , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/uso terapéutico
18.
Oncogene ; 27(30): 4242-8, 2008 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-18345029

RESUMEN

The zebrafish has emerged as a powerful genetic model of cancer, but has been limited by the use of stable transgenic approaches to induce disease. Here, a co-injection strategy is described that capitalizes on both the numbers of embryos that can be microinjected and the ability of transgenes to segregate together and exert synergistic effects in forming tumors. Using this mosaic transgenic approach, gene pathways involved in tumor initiation and radiation sensitivity have been identified.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/efectos de la radiación , Técnicas de Transferencia de Gen , Microinyecciones/métodos , Neoplasias Inducidas por Radiación/genética , Animales , Animales Modificados Genéticamente , Fase de Segmentación del Huevo , Proteínas de Unión al ADN/administración & dosificación , Proteínas de Unión al ADN/genética , Embrión no Mamífero , Genes bcl-2 , Genes myc , Genes p53 , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Proteínas Luminiscentes/administración & dosificación , Proteínas Luminiscentes/genética , Proteínas Mutantes/genética , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Transgenes , Pez Cebra/embriología , Proteínas ras/genética , Proteína Fluorescente Roja
19.
Acta Oncol ; 46(6): 773-81, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17653900

RESUMEN

Gene therapy with adenoviral vectors is a promising new approach for the treatment of refractory advanced breast cancer. Strategies to restrict adenoviral-mediated therapeutic gene expression are important to avoid harming normal cells. Fatty acid synthase (FAS) is overexpressed in several human cancers. FAS is highly expressed in infiltrating breast cancer tissue, and always associated with malignant phenotypes and poor prognosis. In this study, expression of the FAS was evaluated in three breast cancer cell lines. A 680 bp-FAS promoter was cloned and its transcriptional activity was analyzed in breast cancer cell lines. We made a recombinant adenovirus construct carrying herpes simplex virus thymidine kinase (HSV-TK) driven by human FAS promoter (Ad-FAS-TK) and analyzed its target cytotoxicity in vitro and in vivo against human breast cancer cells combined with prodrug ganciclovir (GCV). The results show that the expression of FAS varies in the three breast cancer cell lines examined (respectively, SK-Br3>MCF-7>MDA-MB-231), but FAS promoter can initiate relative high transcriptional activities in all three kinds of cancer cells while little in normal fibroblast cells. Furthermore, FAS promoter can drive the therapeutic gene in a wider range of human breast cancers than cerbB2 promoter and exhibit a stronger activity than midkine (MK) promoter. Combination of Ad-FAS-TK and GCV treatment exhibited strong-targeted cytotoxic effect on breast cancer cells but showed little activity in normal fibroblast cells. The tumorigenic capability of breast cancer cells treated with Ad-FAS-TK/GCV was completely inhibited in vitro and in vivo assays. In conclusion, adenoviral-mediated suicide gene therapy controlled by tumor associated-FAS promoter can induce specific cytotoxic effect on human breast cancer cells in vitro and in vivo. So it is a promising target for the development of gene therapy against breast cancers.


Asunto(s)
Adenovirus Humanos , Neoplasias de la Mama/genética , Ácido Graso Sintasas/uso terapéutico , Terapia Genética , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Supresoras de Tumor/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/terapia , Proteínas de Unión al ADN/administración & dosificación , Proteínas de Unión al ADN/uso terapéutico , Ácido Graso Sintasas/efectos de los fármacos , Femenino , Ganciclovir/uso terapéutico , Expresión Génica , Genes Transgénicos Suicidas , Vectores Genéticos , Humanos , Técnicas In Vitro , Proteínas Nucleares/administración & dosificación , Proteínas Nucleares/uso terapéutico , Fenotipo , Pronóstico , Proteínas Recombinantes de Fusión/administración & dosificación , Insuficiencia del Tratamiento , Resultado del Tratamiento , Proteínas Supresoras de Tumor/administración & dosificación
20.
Immunobiology ; 211(1-2): 29-36, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16446168

RESUMEN

Dendritic cells (DCs) are the only professional antigen-presenting cells endowed with the ability to stimulate naïve T cells and initiate a primary immune response. For this reason, DC-based immunization has been shown to be highly effective in eliciting CTL responses to viruses and tumor-associated antigens. Here we report on the use of DC immunization to enhance the B cell-mediated humoral immune response to highly conserved proteins and the application of this approach to the generation of monoclonal antibodies (mAbs) against these proteins. To illustrate the technique we describe the production of mAbs to class II transactivator (CIITA), the major histocompatibility complex (MHC) CIITA, a difficult immunogen owing to its high degree of identity among species. We show that mice immunized with a combination of an intravenous injection of DCs pulsed with recombinant fragments of CIITA followed by intraperitoneal injection of the antigen in incomplete Freund's adjuvant induced a detectable antibody response against CIITA, while sera from mice immunized using the traditional method (i.e. intraperitoneal immunization with 50mug of protein in complete Freund's adjuvant) gave an almost undetectable response. Furthermore, a total of four fusion experiments demonstrate that immunization with Ag-pulsed DCs is necessary for the efficient generation of hybridomas and a good yield of mAbs specific for the recombinant and the native endogenous CIITA protein. Conversely, four independent fusions carried out with splenocytes from mice immunized using the traditional method failed to produce anti-CIITA hybridomas. We propose that immunization with antigen-loaded DCs should be the method of preference when attempting to raise mAbs against weak self-immunogens.


Asunto(s)
Autoantígenos/administración & dosificación , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Inmunoterapia Adoptiva , Proteínas Nucleares/administración & dosificación , Transactivadores/administración & dosificación , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/sangre , Autoantígenos/genética , Autoantígenos/inmunología , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Células Dendríticas/metabolismo , Citometría de Flujo , Células HeLa , Humanos , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Transactivadores/genética , Transactivadores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...