Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
CNS Neurol Disord Drug Targets ; 7(3): 227-34, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18673207

RESUMEN

Erythropoietin (EPO) was first identified as a hematopoietic cytokine that stimulates proliferation and differentiation of erythroid progenitor cells and was approved by the Food and Drug Administration as a treatment for chronic renal disease patients with anemia. In neural tissues, EPO is working via EPO receptors and induces non-hematopoietic effects. Recent studies have demonstrated that EPO exerts therapeutic potentials on neurological disorders such as ischemic stroke, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and Parkinson's disease. EPO treatment has been shown to reduce the ischemic infarct and hemorrhage volume, decrease neuronal death including apoptosis, and improve survival rates in animal models. The mechanism of EPO action in neurological disorders involves neuroprotection and promotion of neurogenesis and angiogenesis. Clinical trials of EPO treatments in neurological diseases have accumulated positive results. In stroke patients, EPO treatment may reduce infarct volume and improve functional outcomes. EPO administration has proven safe in animal studies and adult human patients, although safety and efficacy data in neonates and infants are incomplete and long-term multi-center patient evaluations are necessary. Available information suggests that EPO is a promising therapeutic drug for the treatment of neurological diseases.


Asunto(s)
Eritropoyetina/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Eritropoyetina/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Enfermedades del Sistema Nervioso/clasificación , Enfermedades del Sistema Nervioso/metabolismo , Fármacos Neuroprotectores/metabolismo , Receptores de Eritropoyetina/metabolismo
2.
J Pharmacol Exp Ther ; 322(2): 521-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17494864

RESUMEN

Cerebral ischemia disrupts the neurovascular unit, involving death of neuronal, glial, and endothelial cells (ECs) in the core and penumbra regions. Whereas the neuroprotective effect of recombinant human erythropoietin (rhEPO) has been widely investigated, its effects on ECs remain elusive. We now report the effects of rhEPO treatment on EC death and neurovasculature repair following a focal ischemic stroke in postnatal day 7 neonatal rats. rhEPO (5000 U/kg i.p.) was administered 60 min after ischemia and for the next 3 days. Western blot analysis revealed increased expression of neurovascular remodeling proteins, including Tie-1, angiopoietin-2, and basic fibroblast growth factor in rhEPO-treated pups. rhEPO treatment significantly reduced EC death in the ischemic penumbra region 12 to 72 h after ischemia examined by immunostaining of terminal deoxynucleotidyl transferase dUTP nick-end labeling and EC marker glucose transporter-1 (GLUT-1). Treatment with rhEPO increased proliferation of ECs and neuronal cells, revealed by costaining of 5-bromo-2'-deoxyuridine with GLUT-1 or with the neuronal marker protein (NeuN) 7 to 21 days after stroke. Specifically, rhEPO increased number of NeuN-positive cells in close proximity to proliferating microvessels. These results suggest for the first time that, in addition to its protection on neural cells, EPO protects ECs and promotes the neurovascular unit repair, which may contribute to its therapeutic benefits after neonatal ischemic stroke.


Asunto(s)
Eritropoyetina/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Angiopoyetina 2/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Western Blotting , Arterias Carótidas/cirugía , Proliferación Celular/efectos de los fármacos , Arterias Cerebrales/cirugía , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Eritropoyetina/uso terapéutico , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Inmunohistoquímica , Ligadura , Sistema Nervioso/irrigación sanguínea , Sistema Nervioso/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Wistar , Receptor TIE-1/metabolismo , Proteínas Recombinantes , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
J Cereb Blood Flow Metab ; 27(5): 1043-54, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17077815

RESUMEN

Restoration of local blood supply in the post-ischemic brain plays a critical role in tissue repair and functional recovery. The present investigation explored beneficial effects of recombinant human erythropoietin (rhEPO) on vascular endothelial cell survival, angiogenesis, and restoration of local cerebral blood flow (LCBF) after permanent focal cerebral ischemia in adult mice. Saline or rhEPO (5,000 U/kg, intraperitoneal) was administered 30 mins before ischemia and once daily after ischemic stroke. Immunohistochemistry showed an enhancing effect of rhEPO on expression of EPO receptor (EPOR) of endothelial cells in the penumbra region 3 to 21 days after the ischemic insult. The treatment with rhEPO decreased ischemia-induced cell death and infarct volume 3 days after stroke. Specifically, rhEPO reduced the number of terminal deoxynucleotidyl transferase biotin-dUPT nick end labeling- and caspase-3-positive endothelial cells in the penumbra region. Colocalization of the vessel marker glucose transporter-1 (Glut-1) and cell proliferation marker 5-bromo-2'-deoxyuridine indicated enhanced angiogenic activity in rhEPO-treated mice 7 to 21 days after stroke. Western blot showed upregulation of the expression of angiogenic factors Tie-2, Angiopoietin-2, and vascular endothelial growth factor in rhEPO-treated animals. Local cerebral blood flow was measured by laser scanning imaging 3 to 21 days after stroke. At 14 days, LCBF in the penumbra was recovered to preischemia levels in rhEPO-treated mice but not in control mice. Our data suggest that rhEPO treatment upregulates the EPOR level in vascular endothelial cells, confers neurovascular protection, and enhances angiogenesis. We further show a promoting effect of rhEPO on LCBF recovery in the ischemic brain. These rhEPO-induced effects may contribute to therapeutic benefits in the treatment of ischemic stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Circulación Cerebrovascular/efectos de los fármacos , Eritropoyetina/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Animales , Antimetabolitos/farmacología , Presión Sanguínea/efectos de los fármacos , Western Blotting , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Bromodesoxiuridina/farmacología , Infarto Cerebral/patología , Técnica del Anticuerpo Fluorescente , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Etiquetado Corte-Fin in Situ , Flujometría por Láser-Doppler , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes
4.
J Pharmacol Exp Ther ; 317(1): 109-16, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16357210

RESUMEN

Cell death induced by the combined insult of hypoxia-ischemia in neonatal rodents has been extensively investigated. Ischemia-only-induced cell death, however, has been much less characterized. Based on the notion that 1) ischemic stroke is a relatively common disorder in human neonates, and 2) developing cells are more susceptible to apoptosis, the present study examined whether typical apoptosis was induced by cerebral ischemia in a new neonatal rat model. Erythropoietin (EPO; Epoetin) was tested for its protective effect against ischemia-induced cell death. Postnatal day 7 rats were subjected to permanent occlusion of the middle cerebral artery branch supplying the right whisker-barrel cortex. Terminal deoxynucleotidyl transferase biotin-dUTP nick end-labeled-positive cells in the ischemic region were detectable 4 h after ischemia and reached a peak level 16 h later. The cell death was preceded by caspase activation and cytochrome c release. Cell body shrinkage was evident among damaged cells. Agarose gel electrophoresis showed DNA damage with a smear pattern as well as DNA laddering. Electron microscopy demonstrated apoptotic features such as cell shrinkage, chromatin condensation, and fragmentation; meanwhile, necrotic alterations coexisted in the cytoplasm. EPO treatment increased signal transducers and activators of transcription-5 and Bcl-2 levels, markedly attenuated apoptotic cell death, and reduced ischemic infarct in the cortex. It is suggested that focal ischemia in the developing brain causes cell death with prominent apoptotic features coexisting with some characteristics of necrosis. This is consistent with the concept of hybrid death described previously in cultures and adult or developing brain. EPO may be explored as a potential therapy for neonatal ischemic stroke.


Asunto(s)
Apoptosis/efectos de los fármacos , Isquemia Encefálica/prevención & control , Corteza Cerebral/ultraestructura , Eritropoyetina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Animales Recién Nacidos , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Eritropoyetina/farmacología , Femenino , Etiquetado Corte-Fin in Situ , Masculino , Microscopía Electrónica de Transmisión , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar , Proteínas Recombinantes
5.
Pathophysiology ; 12(1): 47-62, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15927824

RESUMEN

Ischemic stroke is a leading cause of human death and disability. Although stroke survivors may gain spontaneous partial functional recovery, they often suffer from sensory-motor dysfunctions, behavioral/neurological alterations, and various degrees of paralysis. Currently, limited clinical intervention is available to prevent ischemic damage and restore lost function in stroke victims. In addition to the extensive research on protective maneuvers against ischemia-induced cell death, increasing attention has been focused on potential strategies of promoting tissue repair and functional recovery in the damaged post-ischemic brain. Angiogenesis, or the growth of new blood vessels, may contribute to cell survival and functional recovery of the area of insult. The study of angiogenesis will increase the understanding of the mechanism underlying post-ischemia neurovascular plasticity and regeneration. Additionally, stem cell transplantation has emerged in the last few years as a potential therapy for ischemic stroke, because of their capability to differentiate into multiple cell types and the possibility that they may provide trophic support for cell survival, tissue repair, and functional recovery.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA