Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 30(6): 1680-1688, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38740994

RESUMO

Emotional distress (ED), commonly characterized by symptoms of depression and/or anxiety, is prevalent in patients with cancer. Preclinical studies suggest that ED can impair antitumor immune responses, but few clinical studies have explored its relationship with response to immune checkpoint inhibitors (ICIs). Here we report results from cohort 1 of the prospective observational STRESS-LUNG study, which investigated the association between ED and clinical efficacy of first-line treatment of ICIs in patients with advanced non-small-cell lung cancer. ED was assessed by Patient Health Questionnaire-9 and Generalized Anxiety Disorder 7-item scale. The study included 227 patients with 111 (48.9%) exhibiting ED who presented depression (Patient Health Questionnaire-9 score ≥5) and/or anxiety (Generalized Anxiety Disorder 7-item score ≥5) symptoms at baseline. On the primary endpoint analysis, patients with baseline ED exhibited a significantly shorter median progression-free survival compared with those without ED (7.9 months versus 15.5 months, hazard ratio 1.73, 95% confidence interval 1.23 to 2.43, P = 0.002). On the secondary endpoint analysis, ED was associated with lower objective response rate (46.8% versus 62.1%, odds ratio 0.54, P = 0.022), reduced 2-year overall survival rate of 46.5% versus 64.9% (hazard ratio for death 1.82, 95% confidence interval 1.12 to 2.97, P = 0.016) and detriments in quality of life. The exploratory analysis indicated that the ED group showed elevated blood cortisol levels, which was associated with adverse survival outcomes. This study suggests that there is an association between ED and worse clinical outcomes in patients with advanced non-small-cell lung cancer treated with ICIs, highlighting the potential significance of addressing ED in cancer management. ClinicalTrials.gov registration: NCT05477979 .


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Inibidores de Checkpoint Imunológico , Neoplasias Pulmonares , Angústia Psicológica , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/efeitos adversos , Feminino , Masculino , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Pessoa de Meia-Idade , Idoso , Estudos Prospectivos , Depressão/tratamento farmacológico , Ansiedade/tratamento farmacológico , Resultado do Tratamento , Intervalo Livre de Progressão , Adulto , Idoso de 80 Anos ou mais
2.
Mol Imaging Biol ; 15(6): 748-57, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23689985

RESUMO

PURPOSE: A novel caspase-3 substrate-based probe [(18)F]-CP18 was evaluated as an in vivo positron emission tomography (PET) imaging agent for monitoring apoptosis in tumors. METHODS: Uptake of [(18)F]-CP18 in cell assays and tumors was measured. Caspase-3/7 activities in cell lysates and tumor homogenates were determined. Autoradiography,Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and cleaved caspase-3 immunostaining were performed on adjacent tumor sections to identify areas of apoptosis. RESULTS: The in vitro cell assays showed caspase-3-dependent uptake of [(18)F]-CP18 in tumor cells when treated with an apoptosis inducer. The in vivo microPET imaging signal of [(18)F]-CP18 in xenograft tumors correlated with the ex vivo caspase-3/7 activities in these tumors. Furthermore, tumor autoradiographies of [(18)F]-CP18 in tumor sections matched adjacent sections stained by TUNEL and caspase-3 immunohistochemistry (IHC). CONCLUSIONS: [(18)F]-CP18 demonstrated high affinity and selectivity for activated caspase-3 both in vitro and in vivo, and the results support [(18)F]-CP18 as a promising new PET imaging agent for apoptosis.


Assuntos
Apoptose , Glicopeptídeos/farmacocinética , Imagem Molecular/métodos , Neoplasias/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Animais , Caspase 3/análise , Caspase 3/metabolismo , Caspase 7/análise , Caspase 7/metabolismo , Linhagem Celular Tumoral , Glicopeptídeos/química , Humanos , Modelos Lineares , Camundongos , Compostos Radiofarmacêuticos/química , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Imaging Biol ; 15(6): 739-47, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23681757

RESUMO

PURPOSE: We identified and validated [(18)F]-CP18, a DEVD (the caspase 3 substrate recognition motif) containing substrate-based compound as an imaging tracer for caspase-3 activity in apoptotic cells. PROCEDURES: CP18 was radiolabeled with fluorine-18 using click chemistry. The affinity and selectivity of CP18 for caspase-3 were evaluated in vitro. The biodistribution and metabolism pattern of [(18)F]-CP18 were assessed in vivo. [(18)F]-CP18 positron emission tomography (PET) scans were performed in a dexamethasone-induced thymic apoptosis mouse model. After imaging, the mice were sacrificed, and individual organs were collected, measured in a gamma counter, and tested for caspase-3 activity. RESULTS: In vitro enzymatic caspase-3 assay demonstrated specific cleavage of CP18. In vivo, [(18)F]-CP18 is predominantly cleared through the kidneys and urine, and is rapidly eliminated from the bloodstream. There was a sixfold increase in caspase activity and a fourfold increase of [(18)F]-CP18 retention in the dexamethasone-induced thymus of treated versus control mice. CONCLUSIONS: We report the use [(18)F]-CP18 as a PET tracer for imaging apoptosis. Our data support further development of this tracer for clinical PET applications.


Assuntos
Apoptose , Glicopeptídeos/farmacocinética , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Análise de Variância , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Dexametasona/efeitos adversos , Glicopeptídeos/química , Humanos , Modelos Lineares , Camundongos , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Timo/química , Timo/efeitos dos fármacos , Distribuição Tecidual
4.
Mol Pharm ; 6(3): 747-51, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19093871

RESUMO

Delivery of short interfering RNA (siRNA) to cells in culture, and in vivo, is possible with combined use of a receptor-specific monoclonal antibody (mAb) and avidin-biotin technology. In the present studies, the luciferase gene is transiently expressed in human 293 epithelial cells. The siRNA delivery system is composed of the siRNA, monobiotinylated on the 3'-terminus of the sense strand, and a conjugate of streptavidin (SA) and a mAb to the human insulin receptor (HIR). Exposure of cells to 3'-biotinyl-siRNA bound to the HIRMAb/SA conjugate, but not to unconjugated SA, avidin, or the HIRMAb, causes a >90% reduction in luciferase gene expression. The receptor-targeted siRNA effect is maximal at 48 h after delivery of the siRNA to the cells, and the effect is lost by 7 days after a single application of the targeted siRNA in culture. The KI of the receptor-targeted siRNA inhibition of gene expression is 30.5 +/- 11.7 nM, and significant inhibition is observed with siRNA concentrations as low as 3 nM. In conclusion, the combination of a receptor-specific targeting ligand, such as the HIRMAb, and avidin-biotin technology allows for high affinity capture of the monobiotinylated siRNA by the targeting mAb. The siRNA is effectively delivered to the cytosol of cells, and knockdown of gene expression with the HIRMAb/SA delivery system is comparable to RNA interference effects obtained with cationic polyplexes. Whereas the use of cationic polyplexes in vivo is problematic, the bond between the targeting mAb and the siRNA is stable with avidin-biotin technology, and RNAi effects at distant sites such as brain are observed in vivo following an intravenous administration of the targeted siRNA.


Assuntos
Anticorpos Monoclonais/química , Antígenos CD/imunologia , Avidina/química , Biotina/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/fisiologia , Receptor de Insulina/imunologia , Animais , Anticorpos Monoclonais/imunologia , Linhagem Celular , Humanos , Luciferases/genética , Camundongos , Interferência de RNA/fisiologia , Estreptavidina/química , Fatores de Tempo
5.
Bioconjug Chem ; 19(3): 731-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18278853

RESUMO

The genetic engineering, expression, and validation of a fusion protein of avidin (AV) and a chimeric monoclonal antibody (mAb) to the human insulin receptor (HIR) is described. The 15 kDa avidin monomer was fused to the carboxyl terminus of the heavy chain of the HIRMAb. The fusion protein heavy chain reacted with antibodies specific for human IgG and avidin, and had the same affinity for binding to the HIR extracellular domain as the original chimeric HIRMAb. The fusion protein qualitatively bound biotinylated ligands, but was secreted fully saturated with biotin by COS cells, owing to the high level of biotin in tissue culture medium. Chinese hamster ovary (CHO) cells were permanently transfected with a tandem vector expressing the fusion protein genes, and high expressing cell lines were isolated by methotrexate amplification and dilutional cloning. The product expressed by CHO cells had high binding to the HIR, and migrated as a homogeneous species in size exclusion HPLC and native polyacrylamide gel electrophoresis. The CHO cells were adapted to a 4 week culture in biotin depleted medium, and the HIRMAb-AV fusion protein expressed under these conditions had 1 unoccupied biotin binding site per molecule, based on a [3H]-biotin ultrafiltration assay. The HIRMAb-AV increased biotin uptake by human cells >15-fold, and mediated the endocytosis of fluorescein-biotin, as demonstrated by confocal microscopy. In summary, the HIRMAb-AV fusion protein is a new drug targeting system for humans that can be adapted to monobiotinylated drugs or nucleic acids.


Assuntos
Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Biotina/química , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/farmacologia , Receptores de Droga/efeitos dos fármacos , Animais , Anticorpos Monoclonais/biossíntese , Avidina/química , Biotina/farmacocinética , Western Blotting , Células CHO , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão , Cricetinae , Cricetulus , DNA Complementar/biossíntese , DNA Complementar/genética , Diálise , Sistemas de Liberação de Medicamentos , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Engenharia Genética , Humanos , Microscopia Confocal , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Soroalbumina Bovina/química , Ultrafiltração
6.
Pharm Res ; 24(12): 2309-16, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17926121

RESUMO

PURPOSE: The effective delivery of short interfering RNA (siRNA) to brain following intravenous administration requires the development of a delivery system for transport of the siRNA across the brain capillary endothelial wall, which forms the blood-brain barrier in vivo. METHODS: siRNA was delivered to brain in vivo with the combined use of a receptor-specific monoclonal antibody delivery system, and avidin-biotin technology. The siRNA was mono-biotinylated on either terminus of the sense strand, in parallel with the production of a conjugate of the targeting MAb and streptavidin. RESULTS: Rat glial cells (C6 or RG-2) were permanently transfected with the luciferase gene, and implanted in the brain of adult rats. Following the formation of intra-cranial tumors, the rats were treated with a single intravenous injection of 270 microg/kg of biotinylated siRNA attached to a transferrin receptor antibody via a biotin-streptavidin linker. The intravenous administration of the siRNA caused a 69-81% decrease in luciferase gene expression in the intracranial brain cancer in vivo. CONCLUSIONS: Brain delivery of siRNA following intravenous administration is possible with siRNAs that are targeted to brain with the combined use of receptor specific antibody delivery systems and avidin-biotin technology.


Assuntos
Anticorpos Monoclonais/metabolismo , Biotina/metabolismo , Neoplasias Encefálicas/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores da Transferrina/imunologia , Estreptavidina/metabolismo , Animais , Biotinilação , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Genes Reporter , Humanos , Luciferases/genética , Masculino , RNA Interferente Pequeno/administração & dosagem , Ratos , Ratos Endogâmicos F344 , Receptor de Insulina/imunologia , Fatores de Tempo , Transfecção
7.
Hypertension ; 47(4): 752-61, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16534002

RESUMO

Kallikrein cleaves low molecular weight kininogen to generate vasoactive kinins, which bind to the kinin B2 receptor, triggering a host of biological effects. Kallikrein gene delivery has been shown previously to reduce ischemia/reperfusion-induced cerebral infarction. In this study, we tested the hypothesis that the kinin B2 receptor plays a protective role in ischemic brain injury using kinin B2 receptor gene knockout (B2R-KO) mice subjected to middle cerebral artery occlusion (MCAO). The mortality rate and neurological deficit scores of B2R-KO mice (n=48) after MCAO were significantly increased compared with wild-type (WT) mice (n=40) when examined over a 14-day period. In addition, the infarct volume in B2R-KO mice was significantly larger than in WT mice at days 1 and 3 after MCAO. Similarly, apoptotic cells, detected by TUNEL labeling counterstained with propidium iodide, and caspase-3 activity in the ischemic brain increased significantly in B2R-KO mice at days 1 and 3 after MCAO. Furthermore, the accumulation of neutrophils in the ischemic brain of B2R-KO mice after MCAO increased when compared with WT mice and was associated with elevated tumor necrosis factor alpha expression. These alterations in B2R-KO mice correlated with decreased NO levels, Akt, and glycogen synthase kinase-3beta phosphorylation and increased nicotinamide-adenine dinucleotide oxidase activity. These results indicate that the kinin B2 receptor promotes survival and protects against brain injury by suppression of apoptosis and inflammation induced by ischemic stroke.


Assuntos
Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Receptor B2 da Bradicinina/deficiência , Animais , Apoptose , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Isquemia Encefálica/complicações , Isquemia Encefálica/mortalidade , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multienzimáticos/metabolismo , NADH NADPH Oxirredutases/metabolismo , Doenças do Sistema Nervoso/etiologia , Fármacos Neuroprotetores/metabolismo , Infiltração de Neutrófilos , Nitratos/metabolismo , Nitritos/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor B2 da Bradicinina/metabolismo , Acidente Vascular Cerebral/mortalidade , Fator de Necrose Tumoral alfa/metabolismo
8.
Hum Gene Ther ; 17(2): 206-19, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16454654

RESUMO

Stroke-induced neurological deficits and mortality are often associated with timing of treatment after the onset of stroke. We showed that local delivery of the human tissue kallikrein gene into rat brain immediately after middle cerebral artery occlusion (MCAO) exerts neuroprotection. In this study, we investigated the effect of systemic delivery of the kallikrein gene 8 hr after MCAO. Expression of recombinant human tissue kallikrein after gene transfer was identified in the ischemic brain region and blood vessels. Intravenous injection of adenovirus encoding the kallikrein gene significantly reduced neurological deficit scores 2 and 7 days after gene transfer. Kallikrein gene transfer also reduced ischemia-reperfusion (I/R)-induced cerebral infarction and promoted the survival and migration of glial cells from penumbra to the ischemic core from 3 to 14 days after gene delivery. Kallikrein reduced I/R-induced apoptosis of neuronal cells and inhibited inflammatory cell accumulation in the ischemic brain. These effects were blocked by the kinin B2 receptor antagonist icatibant. In addition, kallikrein enhanced angiogenesis and promoted neurogenesis after I/R and the stimulatory effect of kinin on neuronal cell proliferation was confirmed in primary cultured neuronal cells. The protective effects of kallikrein, through the kinin B2 receptor, were accompanied by increased cerebral nitric oxide and Bcl-2 levels, Akt phosphorylation, and reduced NAD(P)H oxidase activity, superoxide production, Bax levels, and caspase-3 activity. These results indicate that delayed systemic administration of the kallikrein gene after onset of stroke protects against ischemic brain injury by inhibiting apoptosis and inflammation and by promoting angiogenesis and neurogenesis.


Assuntos
Apoptose/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Inflamação/tratamento farmacológico , Calicreínas/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/prevenção & controle , Animais , Apoptose/genética , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Infarto Encefálico/tratamento farmacológico , Infarto Encefálico/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Células Cultivadas , Técnicas de Transferência de Genes , Humanos , Calicreínas/genética , Masculino , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico/metabolismo , Proteína Oncogênica v-akt/efeitos dos fármacos , Proteína Oncogênica v-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Traumatismo por Reperfusão/terapia
9.
Nephrol Dial Transplant ; 21(3): 624-33, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16401625

RESUMO

BACKGROUND: Our previous study showed that kallikrein gene transfer protects against gentamicin-induced nephrotoxicity and enhances renal function. In this study, we investigated the effects and potential mechanisms of kallikrein/kinin on inflammation and apoptosis induced by gentamicin. METHODS: Rats were injected subcutaneously with gentamicin daily for 10 days and received an intravenous injection of adenovirus carrying the human tissue kallikrein gene or control virus on the first day of gentamicin administration. RESULTS: After 10 days of gentamicin treatment, kallikrein gene transfer significantly attenuated gentamicin-induced tubular dilatation and lumenal protein casts. Moreover, kallikrein gene transfer reduced monocyte/macrophage infiltration, monocyte chemoattractant peptide-1 expression and renal cell apoptosis. Kallikrein's protective effects were accompanied by increased nitric oxide formation, and reduced NADH oxidase activity and superoxide production. Suppression of oxidative stress was associated with diminished c-jun N-terminal kinase activation and intercellular adhesion molecule-1 and transforming growth factor-beta protein levels. These biochemical effects were blocked by icatibant, indicating a kinin B2 receptor-mediated signalling event. CONCLUSIONS: This study indicates that kallikrein/kinin protects against gentamicin-induced nephrotoxicity by inhibiting inflammatory cell recruitment and apoptosis through suppression of oxidative stress-mediated signalling pathways. These findings raise the potential of applying kallikrein therapy approaches in treating aminoglycoside-induced renal damage.


Assuntos
Injúria Renal Aguda/prevenção & controle , Apoptose/efeitos dos fármacos , Rim/patologia , Cininas/uso terapêutico , Calicreínas Teciduais/uso terapêutico , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Adenoviridae/genética , Animais , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Técnicas de Transferência de Genes , Gentamicinas/toxicidade , Rim/efeitos dos fármacos , Rim/metabolismo , Cininas/farmacocinética , Masculino , Ratos , Ratos Sprague-Dawley , Calicreínas Teciduais/farmacocinética
10.
Exp Neurol ; 197(2): 521-30, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16343485

RESUMO

Adrenomedullin (AM) is a peptide hormone widely distributed in the central nervous system. Our previous study showed that AM gene delivery immediately after middle cerebral artery occlusion (MCAO) protected against cerebral ischemia/reperfusion (I/R) injury by promoting glial cell survival and migration. In the present study, we investigated the effect of delayed AM peptide infusion on ischemic brain injury at 24 h after MCAO. AM infusion significantly reduced neurological deficit scores at days 2, 4, and 8 after cerebral I/R. AM reduced cerebral infarct size at 8 and 15 days after surgery as determined by quantitative analysis. Double staining showed that AM infusion reduced TUNEL-positive apoptotic cells in both neurons and glial cells, as well as reduced caspase-3 activity in the ischemic area of the brain. In addition, AM treatment increased capillary density in the ischemic region at 15 days after I/R injury. Parallel studies revealed that AM treatment enhanced the proliferation of cultured endothelial cells as measured by both (3)H-thymidine incorporation and in situ BrdU labeling. Both in vitro and in vivo AM effects were blocked by calcitonin gene-related peptide (8-37), an AM receptor antagonist. Moreover, AM's effects were associated with increased cerebral nitric oxide (NO) levels, as well as decreased NAD(P)H oxidase activities and superoxide anion production. These results indicate that a continuous supply of exogenous AM peptide protects against I/R injury by improving the survival of neuronal and glial cells, and promoting angiogenesis through elevated NO formation and suppression of oxidative stress.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Apoptose/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Peptídeos/administração & dosagem , Acidente Vascular Cerebral/prevenção & controle , Adrenomedulina , Análise de Variância , Animais , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Infarto Encefálico/prevenção & controle , Isquemia Encefálica/fisiopatologia , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Caspase 3 , Caspases/metabolismo , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Células Endoteliais/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica/métodos , Marcação In Situ das Extremidades Cortadas/métodos , Masculino , NADPH Oxidases/metabolismo , Exame Neurológico , Nitratos/metabolismo , Nitritos/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
11.
Am J Physiol Renal Physiol ; 289(3): F622-31, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15886273

RESUMO

In DOCA-salt hypertension, renal kallikrein levels are increased and may play a protective role in renal injury. We investigated the effect of enhanced kallikrein levels on kidney remodeling of DOCA-salt hypertensive rats by systemic delivery of adenovirus containing human tissue kallikrein gene. Recombinant human kallikrein was detected in the urine and serum of rats after gene delivery. Kallikrein gene transfer significantly decreased DOCA- and salt-induced proteinuria, glomerular sclerosis, tubular dilatation, and luminal protein casts. Sirius red staining showed that kallikrein gene transfer reduced renal fibrosis, which was confirmed by decreased collagen I and fibronectin levels. Furthermore, kallikrein gene delivery diminished myofibroblast accumulation in the interstitium of the cortex and medulla, as well as transforming growth factor (TGF)-beta1 immunostaining in glomeruli. Western blot analysis and ELISA verified the decrease in immunoreactive TGF-beta1 levels. Kallikrein gene transfer also significantly reduced kidney weight, glomerular size, proliferating tubular epithelial cells, and macrophages/monocytes. Reduction of proliferation and hypertrophy was associated with reduced levels of the cyclin-dependent kinase inhibitor p27(Kip1), and the phosphorylation of c-Jun NH2-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK). The protective effects of kallikrein were accompanied by increased urinary nitrate/nitrite and cGMP levels, and suppression of superoxide formation. These results indicate that kallikrein protects against mineralocorticoid-induced renal fibrosis glomerular hypertrophy, and renal cell proliferation via inhibition of oxidative stress, JNK/ERK activation, and p27(Kip1) and TGF-beta1 expression.


Assuntos
Terapia Genética/métodos , Hipertensão Renal/terapia , Calicreínas/genética , Proteinúria/terapia , Animais , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , GMP Cíclico/urina , Inibidor de Quinase Dependente de Ciclina p27 , Desoxicorticosterona/farmacologia , Modelos Animais de Doenças , Matriz Extracelular/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrose , Técnicas de Transferência de Genes , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Hipertrofia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Nitratos/urina , Estresse Oxidativo , Proteinúria/metabolismo , Proteinúria/patologia , Ratos , Ratos Sprague-Dawley , Cloreto de Sódio na Dieta/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , Proteínas Supressoras de Tumor/metabolismo
12.
Life Sci ; 76(21): 2457-71, 2005 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15763077

RESUMO

Nitric oxide (NO) has been shown to play a key role in the regulation of cardiac hypertrophy and fibrosis in response to myocardial ischemia in part by antagonizing the action of angiotensin II (Ang II). In this study, we investigated the potential protective role of human endothelial nitric oxide synthase (eNOS) in left ventricular (LV) remodeling after myocardial infarction (MI) by a somatic gene transfer approach. Male Wistar rats underwent coronary artery ligation to induce MI. One week after surgery, adenovirus encoding the human eNOS or luciferase gene under the control of the CMV promoter/enhancer was injected into rats via the tail vein, and animals were sacrificed at 1 and 5 weeks after gene transfer. Successful gene transfer was evaluated based on increased levels of NO and cGMP in the heart, measured at one week after eNOS gene delivery. Six weeks after MI, the LV end-diastolic pressure, heart weight, LV axis length and cardiomyocyte size were markedly increased compared to the Sham group, while eNOS gene delivery significantly reduced these parameters. Rats receiving control virus developed considerably more fibrotic lesions identified by Sirius Red staining and collagen I immunostaining compared to Sham rats, and eNOS gene delivery significantly reduced collagen accumulation. eNOS gene transfer also reduced TUNEL-positive apoptotic cells. The cardioprotective effect of NO was accompanied by reduced NADH and NADPH oxidase activities and superoxide formation, TGF-beta1 and p27 levels, JNK activation, NF-kappa B nuclear translocation, and caspase-3 activity. This study shows that NO may play an important role in attenuating cardiac remodeling and apoptosis after myocardial infarction via suppression of oxidative stress-mediated signaling pathways.


Assuntos
Terapia Genética/métodos , Infarto do Miocárdio/terapia , Óxido Nítrico Sintase/genética , Estresse Oxidativo/genética , Remodelação Ventricular/genética , Adenoviridae , Animais , Apoptose/fisiologia , Compostos Azo , Caspase 3 , Caspases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Colágeno , Vasos Coronários/cirurgia , GMP Cíclico/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Vetores Genéticos , Ventrículos do Coração/patologia , Humanos , Immunoblotting , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Ligadura , Luciferases/genética , MAP Quinase Quinase 4 , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NAD/metabolismo , NADP/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III , Tamanho do Órgão , Ratos , Ratos Wistar , Superóxidos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , Proteínas Supressoras de Tumor/metabolismo , Função Ventricular
13.
Hypertension ; 43(2): 452-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14698996

RESUMO

Kallikrein/kinin has been shown to protect against ischemia/reperfusion-induced myocardial infarction and apoptosis. In the present study, we examined the potential neuroprotective action of kallikrein gene transfer in cerebral ischemia. Adult, male Sprague-Dawley rats were subjected to a 1-hour occlusion of the middle cerebral artery followed by intracerebroventricular injection of adenovirus harboring either the human tissue kallikrein gene or the luciferase gene. Kallikrein gene transfer significantly reduced ischemia-induced locomotor deficit scores and cerebral infarction after cerebral ischemia injury. Expression of recombinant human tissue kallikrein was identified and localized in monocytes/macrophages of rat ischemic brain by double immunostaining. Morphological analyses showed that kallikrein gene transfer enhanced the survival and migration of glial cells into the ischemic penumbra and core, as identified by immunostaining with glial fibrillary acidic protein. Cerebral ischemia markedly increased apoptotic cells, and kallikrein gene delivery reduced apoptosis to near-normal levels as seen in sham control rats. In primary cultured glial cells, kinin stimulated cell migration but inhibited hypoxia/reoxygenation-induced apoptosis in a dose-dependent manner. The effects of kinin on both migration and apoptosis were abolished by icatibant, a bradykinin B2 receptor antagonist. Enhanced cell survival after kallikrein gene transfer occurred in conjunction with markedly increased cerebral nitric oxide levels and phospho-Akt and Bcl-2 levels but reduced caspase-3 activation, NAD(P)H oxidase activity, and superoxide production. These results indicate that kallikrein gene transfer provides neuroprotection against cerebral ischemia injury by enhancing glial cell survival and migration and inhibiting apoptosis through suppression of oxidative stress and activation of the Akt-Bcl-2 signaling pathway.


Assuntos
Apoptose , Isquemia Encefálica/prevenção & controle , Calicreínas/genética , Neuroglia/fisiologia , Fármacos Neuroprotetores , Acidente Vascular Cerebral/prevenção & controle , Adenoviridae/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/metabolismo , Caspases/metabolismo , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Córtex Cerebral/patologia , Infarto Cerebral/patologia , Infarto Cerebral/prevenção & controle , Vetores Genéticos , Masculino , NADPH Oxidases/metabolismo , Neuroglia/citologia , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/patologia , Superóxidos/metabolismo , Calicreínas Teciduais/análise , Calicreínas Teciduais/metabolismo
14.
Hum Gene Ther ; 15(12): 1243-54, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15684700

RESUMO

Adrenomedullin (AM) has been shown to protect against ischemia/reperfusion-induced myocardial infarction and apoptosis. In the present study, we examined the potential neuroprotective action of delayed AM gene transfer in cerebral ischemia. Three days after a 1-hr occlusion of the middle cerebral artery (MCAO), rats were injected intravenously with adenovirus harboring human AM cDNA. The experiment was terminated 7 days after MCAO. AM gene transfer significantly reduced cerebral infarct size compared with that of rats before virus injection and compared with that of rats injected with control virus. The expression of recombinant human AM was identified in ischemic brain by immunostaining. Morphological analyses showed that AM gene transfer enhanced the survival and migration of astrocytes into the ischemic core. Cerebral ischemia markedly increased astrocyte apoptosis, and AM gene delivery significantly reduced apoptosis to near normal levels as seen in sham control rats. Similarly, in primary cultured astrocytes, AM stimulated cell migration and inhibited hypoxia/reoxygenation-induced apoptosis. The effects of AM on both migration and apoptosis were abolished by calcitonin gene-related peptide [CGRP(8-37)], an AM receptor antagonist. Enhanced cell survival after AM gene transfer was accompanied by markedly increased cerebral nitric oxide and Bcl-2 levels, as well as Akt and GSK-3beta phosphorylation, but reduced NADPH oxidase activity and superoxide production. Inactivation of GSK-3beta by phosphorylation led to reduced GSK-3beta activity and caspase- 3 activation. These results indicate that exogenous AM provides neuroprotection against cerebral ischemia injury by enhancing astrocyte survival and migration and inhibiting apoptosis through suppression of oxidative stress-mediated signaling events.


Assuntos
Astrócitos/citologia , Isquemia Encefálica/prevenção & controle , Movimento Celular/genética , Técnicas de Transferência de Genes , Fármacos Neuroprotetores , Peptídeos/genética , Adrenomedulina , Animais , Apoptose/genética , Astrócitos/fisiologia , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Caspases/metabolismo , Sobrevivência Celular , Células Cultivadas , Infarto Cerebral/patologia , Infarto Cerebral/prevenção & controle , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Imuno-Histoquímica , Masculino , NADPH Oxidases/metabolismo , Óxido Nítrico/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-bcl-2 , Ratos , Ratos Sprague-Dawley , Superóxidos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA