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










Base de dados
Intervalo de ano de publicação
1.
Hum Cell ; 34(3): 808-818, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33616869

RESUMO

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.


Assuntos
Anestésicos Inalatórios/efeitos adversos , Quinases Lim/metabolismo , MicroRNAs/metabolismo , Fármacos Neuroprotetores , Proteínas Nucleares/farmacologia , Proteínas Nucleares/fisiologia , Complicações Cognitivas Pós-Operatórias/tratamento farmacológico , Complicações Cognitivas Pós-Operatórias/genética , Sevoflurano/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Hipocampo/citologia , L-Lactato Desidrogenase/metabolismo , Quinases Lim/genética , Camundongos , MicroRNAs/genética , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Nucleares/administração & dosagem , Complicações Cognitivas Pós-Operatórias/induzido quimicamente
2.
Hum Exp Toxicol ; 40(7): 1095-1111, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33375867

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Lesões Encefálicas Traumáticas/prevenção & controle , Fator 2 Relacionado a NF-E2/administração & dosagem , Fármacos Neuroprotetores , Proteínas Nucleares/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Células Cultivadas/efeitos dos fármacos , Humanos , Masculino , Camundongos , Modelos Animais
3.
Nat Cell Biol ; 22(10): 1276-1285, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33005030

RESUMO

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.


Assuntos
Neoplasias Encefálicas/radioterapia , Neoplasias da Mama/radioterapia , Dano ao DNA , Reparo do DNA , Proteínas Nucleares/metabolismo , Radiossensibilizantes/metabolismo , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Feminino , Raios gama , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Nucleares/administração & dosagem , Fosforilação , Radiossensibilizantes/administração & dosagem , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nanomedicine (Lond) ; 13(4): 353-372, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29338617

RESUMO

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.


Assuntos
Antineoplásicos/química , Neoplasias da Mama/tratamento farmacológico , Carbono/química , Proteínas de Ciclo Celular/administração & dosagem , Proteínas de Ligação a DNA/administração & dosagem , Portadores de Fármacos/química , Nanosferas/química , Proteínas Nucleares/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclina D1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Liberação Controlada de Fármacos , Feminino , Humanos , Camundongos Endogâmicos BALB C , Proteínas Nucleares/metabolismo , Domínios Proteicos , Proteínas Recombinantes/administração & dosagem , Distribuição Tecidual
5.
J Cereb Blood Flow Metab ; 37(4): 1447-1460, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27381825

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Retroalimentação Fisiológica/fisiologia , Hipóxia-Isquemia Encefálica/metabolismo , Proteínas Nucleares/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/patologia , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/uso terapêutico , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/genética , Proteínas Nucleares/uso terapêutico , Fosforilação , RNA Interferente Pequeno/genética , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais , Serina-Treonina Quinases TOR/genética
6.
Sci Rep ; 6: 30626, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27464702

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Regulação para Baixo , Proteínas de Ligação a RNA/metabolismo , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Proteínas de Choque Térmico HSP72/metabolismo , Humanos , Camundongos , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/metabolismo , Ligação Proteica , Resultado do Tratamento , Ubiquitina-Proteína Ligases/metabolismo
7.
Int J Nanomedicine ; 11: 2039-51, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27274234

RESUMO

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.


Assuntos
Carbono/química , Proteínas de Ciclo Celular/administração & dosagem , Proteínas de Ciclo Celular/uso terapêutico , Proteínas de Ligação a DNA/administração & dosagem , Proteínas de Ligação a DNA/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Nanosferas/química , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/uso terapêutico , Animais , Diferenciação Celular/efeitos dos fármacos , Encefalomielite Autoimune Experimental/patologia , Endocitose/efeitos dos fármacos , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nanosferas/toxicidade , Nanosferas/ultraestrutura , Fator de Transcrição STAT3/metabolismo , Células Th17/citologia , Células Th17/efeitos dos fármacos
9.
Oncotarget ; 6(22): 19148-62, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25895131

RESUMO

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.


Assuntos
Neoplasias da Vesícula Biliar/terapia , Terapia Genética/métodos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/administração & dosagem , Fosfoproteínas Fosfatases/genética , Transporte Ativo do Núcleo Celular , Adulto , Idoso , Animais , Apoptose/genética , Proliferação de Células/genética , Feminino , Neoplasias da Vesícula Biliar/enzimologia , Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/patologia , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Fosforilação , Prognóstico , Distribuição Aleatória , Survivina , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Eur J Pharm Biopharm ; 91: 91-102, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25660910

RESUMO

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.


Assuntos
Materiais Biocompatíveis/química , Proteínas de Ciclo Celular/química , Sistemas de Liberação de Medicamentos , Reativadores Enzimáticos/química , Nanopartículas/química , Proteínas Nucleares/química , Fragmentos de Peptídeos/química , Animais , Materiais Biocompatíveis/efeitos adversos , Transporte Biológico , Proteínas de Ciclo Celular/administração & dosagem , Proteínas de Ciclo Celular/efeitos adversos , Proteínas de Ciclo Celular/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/efeitos adversos , Peptídeos Penetradores de Células/química , Células Cultivadas , Fenômenos Químicos , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/efeitos adversos , Preparações de Ação Retardada/química , Preparações de Ação Retardada/uso terapêutico , Composição de Medicamentos , Sistemas de Liberação de Medicamentos/efeitos adversos , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Disceratose Congênita/tratamento farmacológico , Reativadores Enzimáticos/administração & dosagem , Reativadores Enzimáticos/efeitos adversos , Reativadores Enzimáticos/uso terapêutico , Humanos , Ácido Láctico/efeitos adversos , Ácido Láctico/química , Camundongos , Nanopartículas/efeitos adversos , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/efeitos adversos , Proteínas Nucleares/genética , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/genética , Poliaminas/efeitos adversos , Poliaminas/química , Polieletrólitos , Polietilenoglicóis/efeitos adversos , Polietilenoglicóis/química , Poliglactina 910/efeitos adversos , Poliglactina 910/química , Ácido Poliglicólico/efeitos adversos , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Estabilidade Proteica , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico
11.
J Control Release ; 202: 14-20, 2015 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-25626085

RESUMO

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.


Assuntos
Azidas/química , Compostos Bicíclicos com Pontes/química , Corantes/química , Lipossomos/química , Proteínas Nucleares/química , Rodaminas/química , Repetição de Anquirina , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Colesterol/química , Química Click , Cobre , Molécula de Adesão da Célula Epitelial , Células HT29 , Humanos , Proteínas Nucleares/administração & dosagem , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Polietilenoglicóis/química
12.
IUBMB Life ; 64(4): 331-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22362485

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Proteína Smad2/metabolismo , Transativadores/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Primers do DNA/genética , Sinergismo Farmacológico , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutagênese Sítio-Dirigida , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteína Smad2/genética , Transativadores/administração & dosagem , Transativadores/genética , Fator de Crescimento Transformador beta3/administração & dosagem , Regulação para Cima/efeitos dos fármacos
13.
J Immunol ; 186(7): 4164-74, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21389254

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/transplante , Fertilização in vitro , Sobrevivência de Enxerto/imunologia , Transplante de Coração/imunologia , Proteínas Nucleares/administração & dosagem , Transferência Adotiva , Animais , Linhagem Celular , Feminino , Rejeição de Enxerto/prevenção & controle , Transplante de Coração/métodos , Transplante de Coração/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Distribuição Aleatória , Transplante Homólogo/imunologia , Transplante Homólogo/patologia
14.
PLoS One ; 6(1): e16465, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21304583

RESUMO

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.


Assuntos
Bactérias/metabolismo , Reprogramação Celular , Sistemas de Liberação de Medicamentos/métodos , Proteínas Nucleares/administração & dosagem , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/administração & dosagem , ADP Ribose Transferases/administração & dosagem , ADP Ribose Transferases/farmacocinética , Animais , Sistemas de Secreção Bacterianos , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/farmacocinética , Diferenciação Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Células Cultivadas , Humanos , Integrases , Camundongos , Sinais de Localização Nuclear , Proteínas Nucleares/farmacocinética , Pseudomonas aeruginosa/química , Recombinação Genética , Fatores de Transcrição/farmacocinética
15.
Jpn J Clin Oncol ; 40(12): 1184-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20656693

RESUMO

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.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Desoxicitidina/análogos & derivados , Dermatite/etiologia , Neoplasias Hepáticas/tratamento farmacológico , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/efeitos adversos , Pele/efeitos dos fármacos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/secundário , Idoso , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/efeitos adversos , Neoplasias Ósseas/radioterapia , Neoplasias Ósseas/secundário , Vacinas Anticâncer/imunologia , Proteínas de Ciclo Celular , Quimioterapia Adjuvante , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/secundário , Desoxicitidina/administração & dosagem , Desoxicitidina/efeitos adversos , Dermatite/imunologia , Esquema de Medicação , Feminino , Neoplasias da Vesícula Biliar/patologia , Humanos , Injeções Intradérmicas/efeitos adversos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Linfócitos/imunologia , Proteínas Nucleares/imunologia , Cuidados Paliativos/métodos , Fatores de Processamento de RNA , Radioterapia Adjuvante , Pele/imunologia , Gencitabina
16.
Proc Natl Acad Sci U S A ; 105(43): 16719-24, 2008 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-18940925

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/farmacologia , Mitose , Neoplasias/etiologia , Proteínas Nucleares/genética , Proteínas Nucleares/farmacologia , Aneuploidia , Animais , Proteínas de Ciclo Celular/administração & dosagem , Proteínas de Ciclo Celular/genética , Instabilidade Cromossômica , Doxiciclina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Cinetocoros , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos , Neoplasias/genética , Proteínas Nucleares/administração & dosagem , Distribuição Tecidual
17.
Blood ; 112(6): 2474-83, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18574026

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Produtos do Gene tat/uso terapêutico , Leucemia/tratamento farmacológico , Proteínas Nucleares/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Animais , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Inflamação , Leucemia/patologia , Camundongos , NF-kappa B/antagonistas & inibidores , Células-Tronco Neoplásicas/patologia , Nucleofosmina , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/uso terapêutico
18.
Oncogene ; 27(30): 4242-8, 2008 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-18345029

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/efeitos da radiação , Técnicas de Transferência de Genes , Microinjeções/métodos , Neoplasias Induzidas por Radiação/genética , Animais , Animais Geneticamente Modificados , Fase de Clivagem do Zigoto , Proteínas de Ligação a DNA/administração & dosagem , Proteínas de Ligação a DNA/genética , Embrião não Mamífero , Genes bcl-2 , Genes myc , Genes p53 , Proteínas de Fluorescência Verde/administração & dosagem , Proteínas de Fluorescência Verde/genética , Proteínas Luminescentes/administração & dosagem , Proteínas Luminescentes/genética , Proteínas Mutantes/genética , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Transgenes , Peixe-Zebra/embriologia , Proteínas ras/genética , Proteína Vermelha Fluorescente
19.
Acta Oncol ; 46(6): 773-81, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17653900

RESUMO

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.


Assuntos
Adenovírus Humanos , Neoplasias da Mama/genética , Ácido Graxo Sintases/uso terapêutico , Terapia Genética , Proteínas Recombinantes de Fusão/uso terapêutico , Proteínas Supressoras de Tumor/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/terapia , Proteínas de Ligação a DNA/administração & dosagem , Proteínas de Ligação a DNA/uso terapêutico , Ácido Graxo Sintases/efeitos dos fármacos , Feminino , Ganciclovir/uso terapêutico , Expressão Gênica , Genes Transgênicos Suicidas , Vetores Genéticos , Humanos , Técnicas In Vitro , Proteínas Nucleares/administração & dosagem , Proteínas Nucleares/uso terapêutico , Fenótipo , Prognóstico , Proteínas Recombinantes de Fusão/administração & dosagem , Falha de Tratamento , Resultado do Tratamento , Proteínas Supressoras de Tumor/administração & dosagem
20.
Immunobiology ; 211(1-2): 29-36, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16446168

RESUMO

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.


Assuntos
Autoantígenos/administração & dosagem , Células Dendríticas/imunologia , Células Dendríticas/transplante , Imunoterapia Adotiva , Proteínas Nucleares/administração & dosagem , Transativadores/administração & dosagem , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/sangue , Autoantígenos/genética , Autoantígenos/imunologia , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Células Dendríticas/metabolismo , Citometria de Fluxo , Células HeLa , Humanos , Imunoterapia Adotiva/métodos , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Transativadores/genética , Transativadores/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...