Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Am J Cardiovasc Drugs ; 19(1): 11-20, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30144016

RESUMEN

Distributive shock is a subset of shock marked by decreased systemic vascular resistance, organ hypoperfusion and altered oxygen extraction. Despite the use of intravenous fluids and either higher dose of catecholamines or other additional exogenous vasopressors to maintain blood pressure in the target range, the rate of mortality remains higher in patients with septic shock. Therefore, there is clearly an unmet need for additional safe and effective treatments. The use of angiotensin II to raise the mean arterial pressure (MAP) could provide additional therapy and the opportunity to evaluate a catecholamine-sparing effect by decreasing the dose of concomitant catecholamines while maintaining a target MAP. ATHOS-3 (Angiotensin II for the Treatment of High-Output Shock phase 3; ClinicalTrials.gov number, NCT02338843) was an adequate and well-controlled trial. The primary endpoint was the rate of MAP response at hour 3 of treatment with study drug, defined as either a 10-mmHg increase from baseline in MAP or a MAP of at least 75 mmHg. The secondary endpoints were changes from baseline in Sequential Organ Failure Assessment (SOFA) scores (total and cardiovascular). Mortality was an exploratory endpoint. The trial provided substantial evidence of the effectiveness of angiotensin II in raising blood pressure over placebo in patients with distributive shock, while keeping catecholamine levels constant. There was no change in the secondary endpoint of total SOFA scores relative to placebo when catecholamine use was reduced in lieu of angiotensin II treatment. There was a slight decrease in the secondary endpoint of cardiovascular SOFA score relative to placebo during the catecholamine-sparing phase, reflecting the catecholamine-sparing effect. There was a consistent trend in decreased mortality relative to placebo over the 28-day study period. Based on the agreements emanating from the special protocol assessment to assess blood pressure effects, the data from this single study supported approval of angiotensin II by the Food and Drug Administration for marketing in the USA.


Asunto(s)
Angiotensina II/uso terapéutico , Hipotensión/tratamiento farmacológico , Choque Séptico/tratamiento farmacológico , Vasoconstrictores/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Presión Sanguínea/efectos de los fármacos , Sistema Cardiovascular/efectos de los fármacos , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Estados Unidos , United States Food and Drug Administration , Adulto Joven
3.
Brain Res ; 1395: 94-107, 2011 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565333

RESUMEN

Effects of silencing ectopically expressed hSNCA in rat substantia nigra (SN) were examined as a novel therapeutic approach to Parkinson's disease (PD). AAV-hSNCA with or without an AAV harboring a short-hairpin (sh)RNA targeting hSNCA or luciferase was injected into one SN. At 9weeks, hSNCA-expressing rats had reduced SN dopamine (DA) neurons and exhibited a forelimb deficit. AAV-shRNA-SNCA silenced hSNCA and protected against the forelimb deficit. However, AAV-shRNA-SNCA also led to DA neuron loss suggesting undesirable effects of chronic shRNA expression. Effects on nigrostriatal-projecting neurons were examined using a retrograde tract tracer. Loss of striatal-projecting DA neurons was evident in the vector injection site, whereas DA neurons outside this site were lost in hSNCA-expressing rats, but not in hSNCA-silenced rats. These observations suggest that high levels of shRNA-SNCA were toxic to DA neurons, while neighboring neurons exposed to lower levels were protected by hSNCA gene silencing. Also, data collected on DA levels suggest that neurons other than or in addition to nigrostriatal DA neurons contributed to protection of forelimb use. Our observations suggest that while hSNCA gene silencing in DA neurons holds promise as a novel PD therapy, further development of silencing technology is required.


Asunto(s)
Dependovirus/genética , Silenciador del Gen/fisiología , Terapia Genética/métodos , Vectores Genéticos/farmacología , Vectores Genéticos/toxicidad , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/terapia , alfa-Sinucleína/genética , Animales , Modelos Animales de Enfermedad , Dopamina/fisiología , Miembro Anterior/fisiopatología , Células HEK293 , Humanos , Masculino , Neuronas/metabolismo , Neuronas/patología , Trastornos Parkinsonianos/fisiopatología , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/toxicidad , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , alfa-Sinucleína/fisiología
4.
Brain Res ; 1386: 15-24, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21338582

RESUMEN

Alpha-synuclein (SNCA), an abundantly expressed presynaptic protein, is implicated in Parkinson's disease (PD). Since over-expression of human SNCA (hSNCA) leads to death of dopaminergic (DA) neurons in human, rodent and fly brain, hSNCA gene silencing may reduce levels of toxic forms of SNCA and ameliorate degeneration of DA neurons in PD. To begin to develop a gene therapy for PD based on hSNCA gene silencing, two AAV gene silencing vectors were designed, and tested for efficiency and specificity of silencing, as well as toxicity in vitro. The same hSNCA silencing sequence (shRNA) was used in both vectors, but in one vector, the shRNA was embedded in a microRNA backbone and driven by a pol II promoter, and in the other the shRNA was not embedded in a microRNA and was driven by a pol III promoter. Both vectors silenced hSNCA to the same extent in 293T cells transfected with hSNCA. In DA PC12 cells, neither vector decreased expression of rat SNCA, tyrosine hydroxylase (TH), dopamine transporter (DAT) or the vesicular monoamine transporter (VMAT). However, the mir30 embedded vector was significantly less toxic to both PC12 and SH-SY5Y cells. Our in vitro data suggest that this miRNA-embedded silencing vector may be ideal for chronic in vivo SNCA gene silencing in DA neurons.


Asunto(s)
Dependovirus/genética , Dopamina/metabolismo , Silenciador del Gen/fisiología , Vectores Genéticos/toxicidad , MicroARNs/toxicidad , Neuronas/metabolismo , alfa-Sinucleína/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Vectores Genéticos/fisiología , Células HEK293 , Humanos , Ratones , MicroARNs/genética , Neuronas/patología , Células PC12 , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Plásmidos/genética , Ratas , alfa-Sinucleína/genética
5.
Exp Neurol ; 198(2): 382-90, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16455076

RESUMEN

Human alpha-synuclein overexpression and its toxic accumulation in neurons or glia are known to play key roles in the pathogenesis of Parkinson's disease and other related neurodegenerative synucleinopathies. Several single point mutations in the alpha-synuclein gene, as well as gene duplication and triplication, have been linked to familial Parkinson's disease. Moreover, genetic variability of the alpha-synuclein gene promoter is associated with idiopathic Parkinson's disease. Silencing of the human alpha-synuclein gene by vector-based RNA interference (RNAi) is a promising therapeutic approach for synucleinopathies. Here, we report identification of a 21-nucleotide sequence in the coding region of human alpha-synuclein that constitutes an effective target for robust silencing by RNAi and demonstrate allele-specific silencing of the A53T mutant of human alpha-synuclein. Furthermore, we have developed a plasmid vector-based RNAi for silencing of human alpha-synuclein in vitro. Lastly, using a dual cassette lentivirus that co-expresses an alpha-synuclein-targeting small hairpin RNA (shRNA) and enhanced green fluorescent protein (EGFP) as a marker gene, we demonstrate effective silencing of endogenous human alpha-synuclein in vitro in the human dopaminergic cell line SH-SY5Y and also of experimentally expressed human alpha-synuclein in vivo in rat brain. Our results demonstrate potent silencing of human alpha-synuclein expression in vitro and in vivo by viral vector-based RNAi and provide the tools for developing effective gene silencing therapeutics for synucleinopathies, including Parkinson's disease.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica/fisiología , Lentivirus/fisiología , Interferencia de ARN/fisiología , alfa-Sinucleína/metabolismo , Alelos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/virología , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Masculino , Proteínas Mutantes/fisiología , Fosfopiruvato Hidratasa/metabolismo , Interferencia de ARN/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Ratas , Ratas Wistar , Transfección/métodos , alfa-Sinucleína/genética
6.
J Neurosci Methods ; 114(1): 99-106, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11850044

RESUMEN

Several methodological limitations have emerged in the use of viral gene transfer into skeletal muscle. First, because the nuclei of mature muscle fibers do not undergo division, the use of strategies involving replicative integration of exogenous DNA is greatly limited. Another important limitation concerns the maturation-dependent loss in muscle fiber infectivity with adenoviral vectors. In this study, we investigated the possibility that high-titer infections with recombinant adenovirus, expressing a foreign marker gene under the control of a strong viral promoter, can significantly improve the efficiency of gene transfer in vivo into neonatal and adult rat skeletal muscle. High-titer (2 x 10(10) plaque forming units) intramuscular injection of replication-defective adenovirus vector, expressing green fluorescent protein (GFP) under the control of cytomegalovirus promoter, resulted in GFP expression in 99 +/- 0.34% of fibers in the adult soleus muscle and in approximately 85 +/- 1.44% of fibers in the adult tibialis anterior muscle. Interestingly, reduction in injected adenoviral dose significantly reduced the number of GFP-positive fibers in the adult tibialis anterior muscle, but not in the soleus muscle. However, in neonates, adenoviral infection resulted in GFP expression in 96-99% of the fibers in the tibialis anterior and the gastrocnemius muscles regardless of administered adenoviral dose.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Músculo Esquelético/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Línea Celular , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes , Indicadores y Reactivos/metabolismo , Proteínas Luminiscentes/genética , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA