Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Vis ; 18: 1457-66, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22736937

RESUMEN

PURPOSE: Hypertension and diabetes are known risk factors for retinal microvascular damage. However, the combined effects of diabetes with early and established stages of hypertension on retinal microvascular degeneration remain incompletely understood. METHODS: Male spontaneously hypertensive rats (SHR) were compared to SHR with streptozotocin-induced diabetes (SHR+D) for 6 or 10 weeks and Wistar rats as controls. RESULTS: Hypertension alone (the SHR group) or in combination with diabetes (the SHR+D group) for 6 weeks induced additive increases in total retinal cell death, compared to the Wistar controls. This increase was associated with significant increases in phosphorylated-Jun N-terminal kinase (pJNK) activation, phosphorylated-Akt inhibition, plasma and retinal lipid peroxides, and soluble intracellular adhesion molecule-1 (sICAM-1) levels. After 10 weeks, a similar trend was still observed in retinal nitrotyrosine, nuclear factor kappaB p65, and tumor necrosis factor-α expression, associated with exacerbated pJNK activation and formation of acellular capillaries. CONCLUSIONS: In conclusion, combining diabetes and hypertension-potentiated retinal oxidative/inflammatory stress promoted imbalance between the JNK stress and survival Akt pathways resulting in accelerated retinal cell death and acellular capillary formation.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Hipertensión/metabolismo , Inflamación/metabolismo , Retina/metabolismo , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Retinopatía Diabética/complicaciones , Retinopatía Diabética/genética , Expresión Génica , Hipertensión/complicaciones , Hipertensión/genética , Inflamación/complicaciones , Inflamación/genética , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Peroxidación de Lípido , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Masculino , FN-kappa B/genética , FN-kappa B/metabolismo , Estrés Oxidativo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Retina/patología , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
2.
Mol Vis ; 18: 2993-3003, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23288991

RESUMEN

PURPOSE: Neurotrophins, including nerve growth factor (NGF), are secreted by glia as a pro-form (proNGF) that is normally cleaved into the mature ligand. Increases of proNGF has been well documented in retinal neurodegenerative diseases. Since systemic overexpression of proNGF exhibits embryonic lethality, we aimed to establish a model that specifically and stably overexpresses a cleavage-resistant mutant of proNGF (proNGF123) plasmid in the retina using electroporation. METHODS: Male Sprague-Dawley rats were injected intravitreally with pGFP or pGFP-proNGF123 plasmids, then electroporated with various settings for optimization. Retinal cell death and ganglion cell count were assessed by TUNEL and immunostaining with anti-Brn3. Expression of proNGF, NGF, and their receptors was examined by western blot. Retinal vascular permeability was assessed by extravasation of bovine serum albumin-fluorescein. Development of acellular capillaries was assessed by periodic acid-Schiff and hematoxylin staining. RESULTS: Successful pGFP-proNGF123 gene delivery and expression of proNGF was demonstrated by western blot and extensive proNGF immunostaining in retina sections. Overexpression of proNGF reduced NGF expression while inducing the expression of neurotrophin receptors, including p75(NTR) and tyrosine receptor kinase A, but not sortilin. Overexpression of proNGF resulted in ~50% reduction in ganglion cell count and fivefold increase in TUNEL-positive cells in rat retina. In addition, overexpression of proNGF induced breakdown of the blood-retina barrier evident by twofold increase in extravasation of bovine serum albumin-fluorescein after 1 week and induced the development of acellular capillaries after 4 weeks. CONCLUSIONS: Electroporation can successfully incorporate and express biologically active cleavage-resistant proNGF locally in rat retinas. Overexpression of cleavage-resistant proNGF can be a useful tool to investigate specific molecular mechanisms by which proNGF causes neurodegeneration and vascular injury in the retina.


Asunto(s)
Barrera Hematorretinal/patología , Factores de Crecimiento Nervioso/metabolismo , Precursores de Proteínas/metabolismo , Neuronas Retinianas/patología , Vasos Retinianos/patología , Animales , Barrera Hematorretinal/metabolismo , Permeabilidad Capilar , Supervivencia Celular , Electroporación , Expresión Génica , Técnicas de Transferencia de Gen , Inyecciones Intravítreas , Masculino , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso , Precursores de Proteínas/genética , Proteolisis , Ratas , Ratas Sprague-Dawley , Receptor trkA/genética , Receptor trkA/metabolismo , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Neuronas Retinianas/metabolismo , Vasos Retinianos/metabolismo , Factor de Transcripción Brn-3/genética , Factor de Transcripción Brn-3/metabolismo , Transgenes
3.
Am J Pathol ; 177(3): 1187-97, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20651233

RESUMEN

We have previously shown a causal role of peroxynitrite in mediating retinal ganglion cell (RGC) death in diabetic and neurotoxicity models. In the present study, the role of peroxynitrite in altering the antioxidant and antiapoptotic thioredoxin (Trx) system will be investigated as well as the subsequent effects on glial activation and capillary degeneration. Excitotoxicity of the retina was induced by intravitreal injection of N-methyl-d-aspartate (NMDA) in rats, which also received the peroxynitrite decomposition catalyst FeTPPs. RGC loss was assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and GC count. Glial activation and nitrotyrosine were assessed by immunohistochemistry. Acellular capillaries and pericytes were counted in retinal trypsin digest. NMDA-induced peroxynitrite formation caused RGC loss, which was associated with enhanced expression of Trx and its endogenous inhibitor thioredoxin interacting protein. The results also showed enhanced thioredoxin interacting protein/Trx binding and disruption of the Trx/apoptosis signal-regulating kinase 1 "inhibitory complex," leading to release of apoptosis signal-regulating kinase 1 and activation of the apoptotic pathway, as evidenced by p38 MAPK and poly-ADP-ribose polymerase activation. Furthermore, NMDA caused glial activation and compromised retinal vasculature, as indicated by acellular-capillary formation and pericyte loss. Treatment with FeTPPs blocked these effects. In conclusion, NMDA-induced retinal neuro/vascular injury is mediated by peroxynitrite-altered Trx antioxidant defense, which in turn activates the apoptosis signal-regulating kinase-1 apoptotic pathway. In addition to acute RGC death, an NMDA model can be a useful tool to study glial activation and capillary degeneration in retinal neurodegenerative disorders, including diabetic retinopathy.


Asunto(s)
Apoptosis/efectos de los fármacos , Retinopatía Diabética/metabolismo , Metaloporfirinas/farmacología , N-Metilaspartato/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Vasos Retinianos/efectos de los fármacos , Análisis de Varianza , Animales , Western Blotting , Recuento de Células , Retinopatía Diabética/etiología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Inyecciones Intravítreas , MAP Quinasa Quinasa Quinasa 5/metabolismo , Masculino , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Ácido Peroxinitroso/metabolismo , Ratas , Ratas Sprague-Dawley , Células Ganglionares de la Retina/metabolismo , Vasos Retinianos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
4.
J Pharmacol Exp Ther ; 332(1): 125-34, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19815813

RESUMEN

Diabetic retinopathy and retinopathy of prematurity are blinding disorders that follow a pathological pattern of ischemic retinopathy and affect premature infants and working-age adults. Yet, the treatment options are limited to laser photocoagulation. The goal of this study is to elucidate the molecular mechanism and examine the therapeutic effects of inhibiting tyrosine nitration on protecting early retinal vascular cell death and late neovascularization in the ischemic retinopathy model. Ischemic retinopathy was developed by exposing neonatal mice to 75% oxygen [postnatal day (p) 7-p12] followed by normoxia (21% oxygen) (p12-p17). Peroxynitrite decomposition catalyst 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron III chloride (FeTPPS) (1 mg/kg), the nitration inhibitor epicatechin (10 mg/kg) or the thiol donor N-acetylcysteine (NAC, 150 mg/kg) were administered (p7-p12) or (p7-p17). Vascular endothelial cells were incubated at hyperoxia (40% oxygen) or normoxia (21% oxygen) for 48 h. Vascular density was determined in retinal flat mounts labeled with isolectin B4. Expression of vascular endothelial growth factor, caspase-3, and poly(ADP ribose) polymerase (PARP), activation of Akt and p38 mitogen-activated protein kinase (MAPK), and tyrosine nitration of the phosphatidylinositol (PI) 3-kinase p85 subunit were analyzed by Western blot. Hyperoxia-induced peroxynitrite caused endothelial cell apoptosis as indicated by expression of cleaved caspase-3 and PARP leading to vaso-obliteration. These effects were associated with significant tyrosine nitration of the p85 subunit of PI 3-kinase, decreased Akt activation, and enhanced p38 MAPK activation. Blocking tyrosine nitration of PI 3-kinase with epicatechin or NAC restored Akt phosphorylation, and inhibited vaso-obliteration at p12 and neovascularization at p17 comparable with FeTPPS. Early inhibition of tyrosine nitration with use of epicatechin or NAC can represent safe and effective vascular-protective agents in ischemic retinopathy.


Asunto(s)
Isquemia/tratamiento farmacológico , Ácido Peroxinitroso/metabolismo , Sustancias Protectoras/uso terapéutico , Neovascularización Retiniana/prevención & control , Vasos Retinianos/patología , Tirosina/análogos & derivados , Acetilcisteína/administración & dosificación , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Western Blotting , Catequina/administración & dosificación , Catequina/farmacología , Catequina/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Glutatión/metabolismo , Hiperoxia/enzimología , Hiperoxia/metabolismo , Hiperoxia/patología , Hipoxia/enzimología , Hipoxia/metabolismo , Hipoxia/patología , Isquemia/enzimología , Isquemia/metabolismo , Isquemia/patología , Peroxidación de Lípido/efectos de los fármacos , Metaloporfirinas/administración & dosificación , Metaloporfirinas/farmacología , Metaloporfirinas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/farmacología , Neovascularización Retiniana/enzimología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos/enzimología , Vasos Retinianos/metabolismo , Tirosina/metabolismo
5.
Neuropsychopharmacology ; 33(8): 1942-51, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17805306

RESUMEN

Functional alterations in the neurotrophin, brain-derived neurotrophic factor (BDNF) have recently been implicated in the pathophysiology of schizophrenia. Furthermore, animal studies have indicated that several antipsychotic drugs have time-dependent (and differential) effects on BDNF levels in the brain. For example, our previous studies in rats indicated that chronic treatment with the conventional antipsychotic, haloperidol, was associated with decreases in BDNF (and other neurotrophins) in the brain as well as deficits in cognitive function (an especially important consideration for the therapeutics of schizophrenia). Additional studies indicate that haloperidol has other deleterious effects on the brain (eg increased apoptosis). Despite such limitations, haloperidol remains one of the more commonly prescribed antipsychotic agents worldwide due to its efficacy for the positive symptoms of schizophrenia and its low cost. Interestingly, the hematopoietic hormone, erythropoietin, in its recombinant human form rhEPO has been reported to increase the expression of BDNF in neuronal tissues and to have neuroprotective effects. Such observations provided the impetus for us to investigate in the present study whether co-treatment of rhEPO with haloperidol could sustain the normal levels of BDNF in vivo in rats and in vitro in cortical neuronal cultures and further, whether BDNF could prevent haloperidol-induced apoptosis through the regulation of key apoptotic/antiapoptotic markers. The results indicated that rhEPO prevented the haloperidol-induced reduction in BDNF in both in vivo and in vitro experimental conditions. The sustained levels of BDNF in rats with rhEPO prevented the haloperidol-induced increase in caspase-3 (p<0.05) and decrease in Bcl-xl (p<0.01) protein levels. Similarly, in vitro experiments showed that rhEPO prevented (p<0.001) the haloperidol-induced neuronal cell death as well as the decrease in Bcl-xl levels (p<0.01). These findings may have significant implications for the development of neuroprotective strategies to improve clinical outcomes when antipsychotic drugs are used chronically.


Asunto(s)
Antipsicóticos/antagonistas & inhibidores , Antipsicóticos/farmacología , Apoptosis/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Eritropoyetina/farmacología , Haloperidol/antagonistas & inhibidores , Haloperidol/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Western Blotting , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes , Proteína Letal Asociada a bcl/metabolismo
6.
Transl Stroke Res ; 2(2): 179-85, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21904593

RESUMEN

We have shown that reduction of blood pressure (BP) immediately after the onset of reperfusion reduced neurovascular damage and improved functional outcome after experimental cerebral ischemia and candesartan is particularly effective in improving long-term functional outcome. In this study, we sought to determine if early BP lowering with candesartan, in the presence of an occluded cerebral artery, will reduce injury and improve outcome after experimental stroke. Male Wistar rats underwent 24 h or 7 days of middle cerebral artery occlusion (MCAO). A single dose of 1 mg/kg candesartan was administered intravenously at 3 h after MCAO. Animals received neurobehavioral testing at 3 h, 24 h, and 7 days, and blood pressure was measured by telemetry. Animals had brain tissue collected for infarct size (24 h and 7 days), hemoglobin content, matrix metalloproteinase (MMP) activity, and vascular endothelial growth factor (VEGF) expression (24 h only). Candesartan significantly decreased blood pressure, infarct size (-20%; p=0.021), hemoglobin excess (-50%; p=0.0013), and edema (-35%; p=0.0005) at 24 h after MCAO. This resulted in a reduced cerebral perfusion deficit (p=0.034) in the ischemic hemisphere compared with saline and significantly improved Bederson scores and paw grasp. MMP-2, MMP-9, and VEGF were significantly increased by MCAO, but there were no differences between candesartan- and saline-treated animals. There were no significant differences in behavioral outcome at day 7. BP lowering with candesartan reduces early brain injury after experimental stroke even when the artery remains occluded. The early benefits were not sustained at 7 days, as seen in reperfused animals, however. The neuroprotection and neurorestorative properties of candesartan may occur by separate distinct mechanisms.

7.
Br J Pharmacol ; 164(1): 170-80, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21434880

RESUMEN

BACKGROUND AND PURPOSE: Up-regulation of thioredoxin interacting protein (TXNIP), an endogenous inhibitor of thioredoxin (Trx), compromises cellular antioxidant and anti-apoptotic defences and stimulates pro-inflammatory cytokines expression, implying a role for TXNIP in apoptosis. Here we have examined the causal role of TXNIP expression in mediating retinal neurotoxicity and assessed the neuroprotective actions of verapamil, a calcium channel blocker and an inhibitor of TXNIP expression. EXPERIMENTAL APPROACH: Retinal neurotoxicity was induced by intravitreal injection of NMDA in Sprague-Dawley rats, which received verapamil (10 mg·kg(-1), p.o.) or vehicle. Neurotoxicity was examined by terminal dUTP nick-end labelling assay and ganglion cell count. Expression of TXNIP, apoptosis signal-regulating kinase 1 (ASK-1), NF-κB, p38 MAPK, JNK, cleaved poly-ADP-ribose polymerase (PARP), caspase-3, nitrotyrosine and 4-hydroxy-nonenal were examined by Western and slot-blot analysis. Release of TNF-α and IL-1ß was examined by elisa. KEY RESULTS: NMDA injection enhanced TXNIP expression, decreased Trx activity, causing increased oxidative stress, glial activation and release of TNF-α and IL-1ß. Enhanced TXNIP expression disrupted Trx/ASK-1 inhibitory complex leading to release of ASK-1 and activation of the pro-apoptotic p38 MAPK/JNK pathway, as indicated by cleaved PARP and caspase-3 expression. Treatment with verapamil blocked these effects. CONCLUSION AND IMPLICATIONS: Elevated TXNIP expression contributed to retinal neurotoxicity by three different mechanisms, inducing release of inflammatory mediators such as TNF-α and IL-1ß, altering antioxidant status and disrupting the Trx-ASK-1 inhibitory complex leading to activation of the p38 MAPK/JNK apoptotic pathway. Targeting TXNIP expression is a potential therapeutic target for retinal neurodegenerative disease.


Asunto(s)
Proteínas Portadoras/biosíntesis , Oftalmopatías/metabolismo , Inflamación/metabolismo , Retina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 3/metabolismo , Inhibidores de Caspasas , Proteínas de Ciclo Celular , Oftalmopatías/genética , Oftalmopatías/patología , Inflamación/genética , Inflamación/patología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 5/metabolismo , Masculino , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Neuroglía/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Tiorredoxinas/antagonistas & inhibidores , Tiorredoxinas/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Regulación hacia Arriba/genética , Verapamilo/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Diabetes ; 57(4): 889-98, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18285558

RESUMEN

OBJECTIVE: Recently we have shown that diabetes-induced retinal neurodegeneration positively correlates with oxidative stress and peroxynitrite. Studies also show that peroxynitrite impairs nerve growth factor (NGF) survival signaling in sensory neurons. However, the causal role of peroxynitrite and the impact of tyrosine nitration on diabetes-induced retinal neurodegeneration and NGF survival signaling have not been elucidated. RESEARCH DESIGN AND METHODS: Expression of NGF and its receptors was examined in retinas from human and streptozotocin-induced diabetic rats and retinal ganglion cells (RGCs). Diabetic animals were treated with FeTPPS (15 mg x kg(-1) x day(-1) ip), which catalytically decomposes peroxynitrite to nitrate. After 4 weeks of diabetes, retinal cell death was determined by TUNEL assay. Lipid peroxidation and nitrotyrosine were determined using MDA assay, immunofluorescence, and Slot-Blot analysis. Expression of NGF and its receptors was determined by enzyme-linked immunosorbent assay (ELISA), real-time PCR, immunoprecipitation, and Western blot analyses. RESULTS: Analyses of retinal neuronal death and NGF showed ninefold and twofold increases, respectively, in diabetic retinas compared with controls. Diabetes also induced increases in lipid peroxidation, nitrotyrosine, and the pro-apoptotic p75(NTR) receptor in human and rat retinas. These effects were associated with tyrosine nitration of the pro-survival TrkA receptor, resulting in diminished phosphorylation of TrkA and its downstream target, Akt. Furthermore, peroxynitrite induced neuronal death, TrkA nitration, and activation of p38 mitogen-activated protein kinase (MAPK) in RGCs, even in the presence of exogenous NGF. FeTPPS prevented tyrosine nitration, restored NGF survival signal, and prevented neuronal death in vitro and in vivo. CONCLUSIONS: Together, these data suggest that diabetes-induced peroxynitrite impairs NGF neuronal survival by nitrating TrkA receptor and enhancing p75(NTR) expression.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus/fisiopatología , Neuropatías Diabéticas/tratamiento farmacológico , Degeneración Nerviosa/prevención & control , Factor de Crecimiento Nervioso/genética , Nervio Óptico/fisiopatología , Ácido Peroxinitroso/uso terapéutico , Animales , Cadáver , Muerte Celular , Diabetes Mellitus/patología , Humanos , Peroxidación de Lípido/efectos de los fármacos , Persona de Mediana Edad , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , ARN Mensajero/genética , Ratas , Valores de Referencia , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Estreptozocina , Tirosina/análogos & derivados , Tirosina/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA