Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Stroke ; 53(11): 3250-3259, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36065810

RESUMEN

BACKGROUND: Nelonemdaz is a multitarget neuroprotectant that selectively blocks N-methyl-D-aspartate receptors and scavenges free radicals, as proven in preclinical ischemia-reperfusion studies. We aimed to evaluate the safety and efficacy of nelonemdaz in patients with acute ischemic stroke receiving endovascular reperfusion therapy. METHODS: This phase II randomized trial involved participants with large-artery occlusion in the anterior circulation at baseline who received endovascular reperfusion therapy <8 hours from symptom onset at 7 referral stroke centers in South Korea between October 29, 2016, and June 1, 2020. Two hundred thirteen patients were screened and 209 patients were randomly assigned at a 1:1:1 ratio using a computer-generated randomization system. Patients were divided into 3 groups based on the medication received-placebo, low-dose (2750 mg) nelonemdaz, and high-dose (5250 mg) nelonemdaz. The primary outcome was the proportion of patients with modified Rankin Scale scores of 0-2 at 12 weeks. RESULTS: Two hundred eight patients were assigned to the placebo (n=70), low-dose (n=71), and high-dose (n=67) groups. The groups had similar baseline characteristics. The primary outcome was achieved in 183 patients, and it did not differ among the groups (33/61 [54.1%], 40/65 [61.5%], and 36/57 [63.2%] patients; P=0.5578). The common odds ratio (90% CI) indicating a favorable shift in the modified Rankin Scale scores at 12 weeks was 1.55 (0.92-2.60) between the placebo and low-dose groups and 1.61 (0.94-2.76) between the placebo and high-dose groups. No serious adverse events were reported. CONCLUSIONS: The study arms showed no significant difference in the proportion of patients achieving modified Rankin Scale scores of 0-2 at 12 weeks. Nevertheless, nelonemdaz-treated patients showed a favorable tendency toward achieving these scores at 12 weeks, without serious adverse effects. Thus, a large-scale phase III trial is warranted. REGISTRATION: URL: https://clinicaltrials.gov; Unique identifier: NCT02831088.


Asunto(s)
Isquemia Encefálica , Procedimientos Endovasculares , Accidente Cerebrovascular Isquémico , Fármacos Neuroprotectores , Accidente Cerebrovascular , Humanos , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/cirugía , Isquemia Encefálica/diagnóstico , Trombectomía/efectos adversos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/cirugía , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/cirugía , Fármacos Neuroprotectores/uso terapéutico , Receptores de N-Metil-D-Aspartato , Procedimientos Endovasculares/efectos adversos , Resultado del Tratamiento , Reperfusión
2.
Proc Natl Acad Sci U S A ; 110(29): 12066-71, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818595

RESUMEN

Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disorder characterized by loss of motor neurons. Dominant mutations in the gene for superoxide dismutase 1 (SOD1) give rise to familial ALS by an unknown mechanism. Here we show that genetic deficiency of mammalian sterile 20-like kinase 1 (MST1) delays disease onset and extends survival in mice expressing the ALS-associated G93A mutant of human SOD1. SOD1(G93A) induces dissociation of MST1 from a redox protein thioredoxin-1 and promotes MST1 activation in spinal cord neurons in a reactive oxygen species-dependent manner. Moreover, MST1 was found to mediate SOD1(G93A)-induced activation of p38 mitogen-activated protein kinase and caspases as well as impairment of autophagy in spinal cord motoneurons of SOD1(G93A) mice. Our findings implicate MST1 as a key determinant of neurodegeneration in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Activación Enzimática/fisiología , Neuronas Motoras/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Superóxido Dismutasa/genética , Adulto , Esclerosis Amiotrófica Lateral/metabolismo , Análisis de Varianza , Animales , Autofagia/genética , Autofagia/fisiología , Activación Enzimática/genética , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Mutación Missense/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Médula Espinal/citología , Superóxido Dismutasa-1 , Tiorredoxinas/metabolismo
3.
Neurobiol Dis ; 80: 63-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26002422

RESUMEN

Oxidative stress contributes to degeneration of motor neurons in patients with amyotrophic lateral sclerosis (ALS) as well as transgenic mice overexpressing ALS-associated human superoxide dismutase 1 (SOD1) mutants. However, the molecular mechanism by which the ALS-linked SOD1 mutants including SOD1(G93A) induce oxidative stress remains unclear. Here, we show that iron was accumulated in ventral motor neurons from SOD1(G93A)-transgenic mice even at 4 weeks of age, subsequently inducing oxidative stress. Iron chelation with deferoxamine mesylate delayed disease onset and extended lifespan of SOD1(G93A) mice. Furthermore, SOD1(G93A)-induced iron accumulation mediated the increase in the enzymatic activity of TNF-α converting enzyme (TACE), leading to secretion of TNF-α at least in part through iron-dependent oxidative stress. Our findings suggest iron as a key determinant of early motor neuron degeneration as well as proinflammatory responses at symptomatic stage in SOD1(G93A) mice.


Asunto(s)
Proteínas ADAM/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Hierro/toxicidad , Neuronas Motoras/metabolismo , Superóxido Dismutasa/genética , Factor de Necrosis Tumoral alfa/sangre , Proteína ADAM17 , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Modelos Animales de Enfermedad , Humanos , Hierro/metabolismo , Ratones , Ratones Transgénicos , Actividad Motora , Neuronas Motoras/patología , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología
4.
J Stroke ; 25(1): 160-168, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36746385

RESUMEN

BACKGROUND AND PURPOSE: Nelonemdaz (Neu2000) has both selective antagonism against 2B subunit of N-methyl-D-aspartate receptor and antioxidant activity. This drug provides sufficient evidence of neuroprotection in acute cerebral ischemia/reperfusion models. This phase III trial aims to determine this effect in patients. DESIGN: The Rescue on Reperfusion Damage in Cerebral Infarction by Nelonemdaz is a multicenter, double-blinded clinical trial. A total of 496 patients will be randomly assigned into the nelonemdaz (a total of 5,250 mg divided by 10 times for 5 days) and placebo groups. Patients will be included if they have an acute ischemic stroke (National Institutes of Health Stroke Scale score ≥8) caused by intracranial large vessel occlusion in the anterior circulation (Alberta Stroke Program Early CT Score ≥4), and if they are expected to undergo endovascular thrombectomy within 12 hours after stroke onset. ENDPOINTS: The primary endpoint is a favorable shift in the modified Rankin Scale (mRS) score at 90 days after the first dose of drug. The data will be analyzed by the Cochran-Mantel-Haenszel shift test. The secondary endpoints include functional independence (mRS 0-2) at 35 and 90 days, the favorable shift of mRS at 35 days, the proportion of mRS 0 at 35 and 90 days, and the occurrence rates of symptomatic intracranial hemorrhage within 7 days. CONCLUSION: This trial will clarify the efficacy and safety of nelonemdaz in patients with acute ischemic stroke and endovascular thrombectomy. This study has been registered at ClinicalTrials. gov (NCT05041010).

5.
J Neurochem ; 122(5): 952-61, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22537108

RESUMEN

While free radicals and inflammation constitute major routes of neuronal injury occurring in amyotrophic lateral sclerosis (ALS), neither antioxidants nor non-steroidal anti-inflammatory drugs have shown significant efficacy in human clinical trials. We examined the possibility that concurrent blockade of free radicals and prostaglandin E(2) (PGE(2))-mediated inflammation might constitute a safe and effective therapeutic approach to ALS. We have developed 2-hydroxy-5-[2-(4-trifluoromethylphenyl)-ethylaminobenzoic acid] (AAD-2004) as a derivative of aspirin. AAD-2004 completely removed free radicals at 50 nM as a potent spin-trapping molecule and inhibited microsomal PGE(2) synthase-1 (mPGES-1) activity in response to both lipopolysaccharide-treated BV2 cell with IC(50) of 230 nM and recombinant human mPGES-1 protein with IC(50) of 249 nM in vitro. In superoxide dismutase 1(G93A) transgenic mouse model of ALS, AAD-2004 blocked free radical production, PGE(2) formation, and microglial activation in the spinal cords. As a consequence, AAD-2004 reduced autophagosome formation, axonopathy, and motor neuron degeneration, improving motor function and increasing life span. In these assays, AAD-2004 was superior to riluzole or ibuprofen. Gastric bleeding was not induced by AAD-2004 even at a dose 400-fold higher than that required to obtain maximal therapeutic efficacy in superoxide dismutase 1(G93A). Targeting both mPGES-1-mediated PGE(2) and free radicals may be a promising approach to reduce neurodegeneration in ALS and possibly other neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Dinoprostona/metabolismo , Radicales Libres/metabolismo , Sulfasalazina/uso terapéutico , 8-Hidroxi-2'-Desoxicoguanosina , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Análisis de Varianza , Animales , Antiinflamatorios no Esteroideos/farmacología , Aspirina/análogos & derivados , Aspirina/farmacología , Aspirina/uso terapéutico , Proteínas de Unión al Calcio/metabolismo , Corteza Cerebral/patología , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Encefalitis/tratamiento farmacológico , Depuradores de Radicales Libres/metabolismo , Radicales Libres/antagonistas & inhibidores , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Ibuprofeno/farmacología , Ibuprofeno/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , Estrés Oxidativo/efectos de los fármacos , Riluzol/farmacología , Riluzol/uso terapéutico , Médula Espinal/patología , Sulfasalazina/farmacología , Superóxido Dismutasa/genética , Tirosina/análogos & derivados , Tirosina/metabolismo
6.
Trials ; 23(1): 587, 2022 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-35871083

RESUMEN

BACKGROUND: Ischemic brain injury is a major hurdle that limits the survival of resuscitated out-of-hospital cardiac arrest (OHCA). METHODS: The aim of this study is to assess the feasibility and potential for reduction of ischemic brain injury in adult OHCA patients treated with high- or low-dose Neu2000K, a selective blocker of N-methyl-D-aspartate (NMDA) type 2B receptor and also a free radical scavenger, or given placebo. This study is a phase II, multicenter, randomized, double-blinded, prospective, intention-to-treat, placebo-controlled, three-armed, safety and efficacy clinical trial. This trial is a sponsor-initiated trial supported by GNT Pharma. Successfully resuscitated OHCA patients aged 19 to 80 years would be included. The primary outcome is blood neuron-specific enolase (NSE) level on the 3rd day. The secondary outcomes are safety, efficacy defined by study drug administration within 4 h in > 90% of participants, daily NSE up to 5th day, blood S100beta, brain MRI apparent diffusion coefficient imaging, cerebral performance category (CPC), and Modified Rankin Scale (mRS) at 5th, 14th, and 90th days. Assuming NSE of 42 ± 80 and 80 ± 80 µg/L in the treatment (high- and low-dose Neu2000K) and control arms with 80% power, a type 1 error rate of 5%, and a 28% of withdrawal prior to the endpoint, the required sample size is 150 patients. DISCUSSION: The AWAKE trial explores a new multi-target neuroprotectant for the treatment of resuscitated OHCA patients. TRIAL REGISTRATION: ClinicalTrials.gov NCT03651557 . Registered on August 29, 2018.


Asunto(s)
Lesiones Encefálicas , Hipoxia Encefálica , Paro Cardíaco Extrahospitalario , Adulto , Antioxidantes/efectos adversos , Humanos , Paro Cardíaco Extrahospitalario/diagnóstico , Paro Cardíaco Extrahospitalario/tratamiento farmacológico , Estudios Prospectivos , Receptores de N-Metil-D-Aspartato/uso terapéutico , Resultado del Tratamiento , Vigilia
7.
J Pain Res ; 15: 3869-3879, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36531829

RESUMEN

Purpose: Nonsteroidal anti-inflammatory drugs (NSAIDs) and cyclooxygenase (COX)-2 selective inhibitors are the most widely used drugs to treat pain. Conventional NSAIDs and COX-2 selective inhibitors, however, cause several side effects such as gastric damage, kidney damage, and cardiovascular problems. Our previous study showed that 2-acetoxy-5-(2-4-(trifluoromethyl)-phenethylamino)-benzoic acid ie, flusalazine (also known as ND-07), which exerts dual actions by serving both as an anti-inflammatory agent and a free radical scavenger, is an effective and safe treatment for severe inflammatory diseases in mice. The goal of the present study was to examine the potential analgesic action and safety of flusalazine in mice models of pain. Methods and Results: Flusalazine showed a significant analgesic effect in an acetic acid-induced abdominal constriction model. Likewise, total paw licking was reduced significantly in neurogenic (early stage) and inflammatory (late stage) pain induced by formalin in flusalazine-treated mice. In the tail immersion test, flusalazine significantly increased tail withdrawal time at 2 h after its administration. Also, the formation of paw edema in the flusalazine-treated group was significantly inhibited in a carrageenan-induced inflammatory pain model. Gastric damage was not induced by flusalazine even up to 1000 mg/kg, while aspirin and indomethacin caused critical gastric bleeding. Conclusion: These findings suggest that flusalazine's safety profile and analgesic effects have high translational potential for the clinical treatment of patients experiencing pain.

8.
BMC Neurosci ; 12: 106, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-22029666

RESUMEN

BACKGROUND: Evidence suggests that rheumatoid arthritis (RA) may enhance or reduce the progression of Alzheimer's disease (AD). The present study was performed to directly explore the effects of collagen-induced rheumatoid arthritis (CIA) on amyloid plaque formation, microglial activation, and microvascular pathology in the cortex and hippocampus of the double transgenic APP/PS1 mouse model for AD. Wild-type or APP/PS1 mice that received type II collagen (CII) in complete Freund's adjuvant (CFA) at 2 months of age revealed characteristics of RA, such as joint swelling, synovitis, and cartilage and bone degradation 4 months later. Joint pathology was accompanied by sustained induction of IL-1ß and TNF-α in plasma over 4 weeks after administration of CII in CFA. RESULTS: CIA reduced levels of soluble and insoluble amyloid beta (Aß) peptides and amyloid plaque formation in the cortex and hippocampus of APP/PS1 mice, which correlated with increased blood brain barrier disruption, Iba-1-positive microglia, and CD45-positive microglia/macrophages. In contrast, CIA reduced vessel density and length with features of microvascular pathology, including vascular segments, thinner vessels, and atrophic string vessels. CONCLUSIONS: The present findings suggest that RA may exert beneficial effects against Aß burden and harmful effects on microvascular pathology in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/patología , Artritis Experimental/patología , Artritis Reumatoide/patología , Modelos Animales de Enfermedad , Microcirculación , Presenilina-1/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Amiloidosis/genética , Amiloidosis/fisiopatología , Animales , Artritis Experimental/genética , Artritis Experimental/fisiopatología , Artritis Reumatoide/genética , Artritis Reumatoide/fisiopatología , Colágeno Tipo II/toxicidad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Microcirculación/genética
9.
Acta Neuropathol ; 121(4): 459-73, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21120509

RESUMEN

Abnormal brain iron homeostasis has been proposed as a pathological event leading to oxidative stress and neuronal injury under pathological conditions. We examined the possibility that neuronal iron overload would mediate free radical production and delayed neuronal death (DND) in hippocampal CA1 area after transient forebrain ischemia (TFI). Mitochondrial free radicals (MFR) were biphasically generated in CA1 neurons 0.5-8 and 48-60 h after TFI. Treatment with Neu2000, a potent spin trapping molecule, as well as trolox, a vitamin E analogue, blocked the biphasic MFR production and attenuated DND in the CA1, regardless of whether it was administered immediately or even 24 h after reperfusion. The late increase in MFR was accompanied by iron accumulation and blocked by the administration of deferoxamine-an iron chelator. Iron accumulation was attributable to prolonged upregulation of the transferrin receptor and to increased uptake of peripheral iron through a leaky blood-brain barrier. Infiltration of iron-containing cells and iron accumulation were attenuated by depletion of circulating blood cells through X-ray irradiation of the whole body except the head. The present findings suggest that excessive iron transported from blood mediates slowly evolving oxidative stress and neuronal death in CA1 after TFI, and that targeting iron-mediated oxidative stress holds extended therapeutic time window against an ischemic event.


Asunto(s)
Hipocampo/metabolismo , Hipocampo/patología , Hierro/sangre , Ataque Isquémico Transitorio/patología , Neuronas/fisiología , Prosencéfalo/patología , 8-Hidroxi-2'-Desoxicoguanosina , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Autoantígenos/metabolismo , Muerte Celular , Células Cultivadas , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Azul de Evans , Glicoforinas/metabolismo , Hipocampo/efectos de los fármacos , Hierro/metabolismo , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/fisiopatología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Neuronas/efectos de los fármacos , Neuronas/enzimología , Peroxidasa/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Prosencéfalo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/metabolismo , Factores de Tiempo , Transferrina/metabolismo , Zinc/metabolismo
10.
J Neurochem ; 114(1): 160-70, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20374429

RESUMEN

Alternative splicing of tau exon 10 influences microtubule assembly and stability during development and in pathological processes of the central nervous system. However, the cellular events that underlie this pre-mRNA splicing remain to be delineated. In this study, we examined the possibility that ischemic injury, known to change the cellular distribution and expression of several RNA splicing factors, alters the splicing of tau exon 10. Transient occlusion of the middle cerebral artery reduced tau exon 10 inclusion in the ischemic cortical area within 12 h, resulting in the induction of three-repeat (3R) tau in cortical neurons. Ubiquitinated protein aggregates and reduced proteasome activity were also observed. Administration of proteasome inhibitors such as MG132, proteasome inhibitor I and lactacystin reduced tau exon 10 splicing in cortical cell cultures. Decreased levels of Tra2beta, an RNA splicing factor responsible for tau exon 10 inclusion, were detected both in cortical cell cultures exposed to MG132 and in cerebral cortex after ischemic injury. Taken together, these findings suggest that transient focal cerebral ischemia reduces tau exon 10 splicing through a mechanism involving proteasome-ubiquitin dysfunction and down-regulation of Tra2beta.


Asunto(s)
Hipoxia-Isquemia Encefálica/metabolismo , Ataque Isquémico Transitorio/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas tau/metabolismo , Empalme Alternativo , Animales , Células Cultivadas , Corteza Cerebral/metabolismo , Exones , Masculino , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Fosforilación , Inhibidores de Proteasoma , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Empalme Serina-Arginina , Ubiquitinación , Proteínas tau/genética
11.
Drug News Perspect ; 23(9): 549-56, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21152450

RESUMEN

Excess activation of ionotropic glutamate receptors, primarily N-methyl-D-aspartate (NMDA) receptors and free radicals, evoke nerve cell death following hypoxic-ischemic brain injury in various animal models. However, clinical trials in stroke patients using NMDA receptor antagonists have failed to show efficacy primarily due to the limited therapeutic time window for neuroprotection and a narrow therapeutic index. In comparison, antioxidants prolonged the time window for neuroprotection in animal models of ischemic stroke and showed greater therapeutic potential in clinical trials for ischemic stroke. Excess activation of NMDA receptors and free radicals mediate the two separate pathways of nerve cell death in stroke and a safe and multifunctional drug that can block both routes in the brain will likely provide a better therapeutic outcome in patients with stroke. Derivatives of the lead structures of sulfasalazine and aspirin have led to the discovery of a new molecule, Neu2000, that has demonstrated excellent neuroprotection against NMDA- and free radical-induced cell death. Neu2000 is an NR2B-selective, moderate NMDA receptor antagonist with potent cell-permeable, spin trapping antioxidant action even at nanomolar concentrations. Nonclinical and human phase I studies demonstrated that Neu2000 can be translated to treat patients with stroke with better efficacy and therapeutic time window.


Asunto(s)
Antioxidantes/farmacología , Benzoatos/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Antioxidantes/efectos adversos , Benzoatos/efectos adversos , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/fisiopatología , Sistemas de Liberación de Medicamentos , Fluorobencenos , Humanos , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Salicilatos , Detección de Spin , Accidente Cerebrovascular/fisiopatología , Factores de Tiempo , metaminobenzoatos
12.
Cerebrovasc Dis ; 29(5): 431-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20203485

RESUMEN

BACKGROUND: Most epidemiological studies have reported a significant association between elevated serum levels of uric acid (UA) and increased cardiovascular disease. On the other hand, UA is the most abundant antioxidant in the human body. We hypothesized that UA levels would change noticeably in association with the degree of oxidative stress in acute ischemic stroke. METHODS: We analyzed consecutive patients with acute ischemic stroke in the anterior circulation system within 24 h of symptom onset, confirmed by diffusion-weighted imaging (DWI), and with a modified NIH Stroke Scale (mNIHSS) score of 5 or greater. Baseline demographics, serial clinical scores, serial UA and allantoin (a nonenzymatic metabolite of UA) levels, UA change (baseline UA - UA at 48 h), and DWI lesion volumes were compared between 45 patients with recanalized vessels (RV) and 43 patients with nonrecanalized vessels (NV) in follow-up imaging. RESULTS: The RV (vs. NV) patients were more likely to receive thrombolytic treatment (p = 0.005), achieve a reduction in day-14-mNIHSS scores (p = 0.001), and greater changes in UA (p = 0.024) and allantoin levels (p = 0.003). The UA levels dropped at 48 h and gradually increased in a U-shaped pattern. UA change (r = 0.360; p = 0.001) rather than baseline UA (r = 0.044; p = 0.681) was significantly correlated with infarct volume. In the RV (vs. NV) patients, there was a stronger association between infarct volume and UA change (r = 0.483; p = 0.001) or allantoin levels (r = 0.466; p = 0.017). CONCLUSION: Our results suggest that UA might be a consumptive and reproducible antioxidant in acute ischemic stroke, and this pattern appears to be influenced by recanalization success and infarct volume size.


Asunto(s)
Circulación Cerebrovascular/fisiología , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/fisiopatología , Ácido Úrico/sangre , Grado de Desobstrucción Vascular/fisiología , Anciano , Alantoína/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estrés Oxidativo/fisiología , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Factores de Tiempo
13.
Nat Neurosci ; 23(12): 1555-1566, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33199896

RESUMEN

Although the pathological contributions of reactive astrocytes have been implicated in Alzheimer's disease (AD), their in vivo functions remain elusive due to the lack of appropriate experimental models and precise molecular mechanisms. Here, we show the importance of astrocytic reactivity on the pathogenesis of AD using GiD, a newly developed animal model of reactive astrocytes, where the reactivity of astrocytes can be manipulated as mild (GiDm) or severe (GiDs). Mechanistically, excessive hydrogen peroxide (H2O2) originated from monoamine oxidase B in severe reactive astrocytes causes glial activation, tauopathy, neuronal death, brain atrophy, cognitive impairment and eventual death, which are significantly prevented by AAD-2004, a potent H2O2 scavenger. These H2O2--induced pathological features of AD in GiDs are consistently recapitulated in a three-dimensional culture AD model, virus-infected APP/PS1 mice and the brains of patients with AD. Our study identifies H2O2 from severe but not mild reactive astrocytes as a key determinant of neurodegeneration in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Astrocitos/patología , Peróxido de Hidrógeno/metabolismo , Enfermedad de Alzheimer/psicología , Animales , Atrofia , Encéfalo/patología , Muerte Celular , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Humanos , Activación de Macrófagos , Ratones , Ratones Mutantes Neurológicos , Ratones Transgénicos , Monoaminooxidasa/metabolismo , Degeneración Nerviosa/patología , Neuroglía , Neuronas/patología , Memoria Espacial , Tauopatías/patología
14.
J Neurosci ; 28(7): 1721-7, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18272692

RESUMEN

Several studies report microglial accumulation and activation in the CA1 area in response to transient forebrain ischemia (TFI). Here we examine the possibility that free radicals and chemokines mediate the transient activation of microglia. Free radicals are produced primarily in CA1 pyramidal neurons within 2 h of TFI. Administration of trolox, a vitamin E analog, led to the inhibition of free radical production and recruitment of microglia in the CA1 area. In addition, intrahippocampal injection of Fe2+ triggered free radical production in CA1 neurons, followed by the recruitment and activation of microglial cells into this area. TFI-induced expression of macrophage inflammatory protein-1alpha (MIP-1alpha) was increased in CA1 neurons before microglial recruitment, and blocked by trolox. Moreover, the MIP-1alpha level was upregulated in cultured hippocampal neurons exposed to Fe2+, suggesting an essential role of free radicals in TFI-induced expression of MIP-1alpha. Intracerebroventricular injection of vMIP-2 (viral macrophage inflammatory protein-2), a broad-spectrum peptide antagonist of chemokine receptors, attenuated microglial recruitment and delayed CA1 neuronal degeneration after TFI. Our data suggest that free radicals produced in CA1 neurons contribute to the recruitment and activation of microglia and neurodegeneration through MIP-1alpha expression.


Asunto(s)
Muerte Celular , Quimiocina CCL3/metabolismo , Radicales Libres/metabolismo , Ataque Isquémico Transitorio/fisiopatología , Microglía/patología , Prosencéfalo/irrigación sanguínea , Células Piramidales/metabolismo , Animales , Antioxidantes/farmacología , Recuento de Células , Cloruros , Cromanos/farmacología , Compuestos Férricos/farmacología , Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/terapia , Masculino , Microglía/metabolismo , Células Piramidales/patología , Ratas , Ratas Sprague-Dawley , Reclutamiento Neurofisiológico , Reperfusión , Regulación hacia Arriba
15.
Glia ; 56(10): 1039-47, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381656

RESUMEN

Brain inflammation is a suggested risk factor for neurodegenerative disease. Interestingly, severe inflammation in the substantia nigra pars compacta (SNpc) accelerates the onset and progression of Parkinson's disease. In this study, we examined the underlying mechanisms of severe inflammation in the SNpc by comparing the inflammatory process with that in the cortex. In intact brain, the densities of CD11b(+) microglia were similar in the SNpc and cortex. However, lipopolysaccharide injection enhanced the CD11b(+) cell number in the SNpc, but not in the cortex. Previously, we reported that CD11b and myeloperoxidase (MPO) double-positive neutrophils infiltrate the SNpc following LPS injection (GLIA 55:1577-88). Notably, the MPO(+) neutrophil number increased dramatically in the SNpc, but only slightly in the cortex. The extent of neutrophil infiltration appeared to correlate with neuronal damage. We confirmed that loss of neurons in the SNpc was significantly reduced in neutropenic rats versus normal rats following LPS injection. In addition, the densities of astrocytes were much lower in the intact SNpc, compared with the cortex. Furthermore, after LPS injection, damage of endothelial cells and astrocytes, and blood-brain barrier (BBB) permeability was more pronounced in the SNpc. These results collectively suggest that excessive neutrophil infiltration and environmental factors, such as lower astrocyte density and higher BBB permeability, contribute to severe inflammation and neuronal death in the SNpc.


Asunto(s)
Corteza Cerebral/patología , Encefalitis/patología , Infiltración Neutrófila/fisiología , Sustancia Negra/patología , Animales , Recuento de Células/métodos , Masculino , Ratas , Ratas Sprague-Dawley
16.
Neurobiol Dis ; 30(2): 174-85, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18316197

RESUMEN

Cortical neurons deprived of serum undergo apoptosis that is sensitive to inhibitors of macromolecule synthesis. Proteomic analysis revealed differential expression of 49 proteins in cortical neurons 8 h after serum deprivation. Tissue inhibitor of metalloproteinases-3 (TIMP-3), a pro-apoptotic protein in various cancer cells, was increased during serum deprivation-induced apoptosis (SDIA), but not during necrosis induced by excitotoxicity or oxidative stress. Levels of TIMP-3 were markedly increased in degenerating motor neurons in a transgenic model of familial amyotrophic lateral sclerosis. The TIMP-3 expression was accompanied by increase in Fas-FADD interaction, activated caspase-8, and caspase-3 during SDIA and in vulnerable spinal cord of the ALS mouse. SDIA and activation of the Fas pathway were prevented by addition of an active MMP-3. Timp-3 deletion by RNA interference attenuated SDIA in N2a cells. These findings provide evidence that TIMP-3 is an upstream mediator of neuronal apoptosis and likely contributes to neuronal loss in neurodegenerative diseases such as amyotrophic lateral sclerosis.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Apoptosis/fisiología , Medio de Cultivo Libre de Suero/farmacología , Neuronas/enzimología , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Inhibidor Tisular de Metaloproteinasa-3/genética , Regulación hacia Arriba/genética , Receptor fas/fisiología , Alanina/genética , Sustitución de Aminoácidos/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Glicina/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Inhibidor Tisular de Metaloproteinasa-3/fisiología
17.
Cell Transplant ; 16(10): 1007-12, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18351016

RESUMEN

To understand the fates of human mesenchymal stem cells (hMSCs) following transplantation into a rodent model of middle cerebral artery occlusion (MCAo), magnetic resonance imaging (MRI) techniques were employed, hMSCs were labeled with ferumoxides (Feridex)--protamine sulfate complexes, which were visualized and examined by MRI up to 10 weeks following transplantation. Migration of the transplanted cells to the infarcted area was further confirmed by histological methods. We found that the hMSCs transplanted in MCAo models possess the capacity to migrate to the infarcted area extensively in both ipsilateral and contralateral injections, exhibiting a pathotropism. We also analyzed the detailed migration patterns of transplanted hMSCs. We speculate that the extensive migratory ability of hMSCs may represent a therapeutic potential for developing efficient cell transplantation strategies in stroke.


Asunto(s)
Infarto de la Arteria Cerebral Media/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Movimiento Celular , Medios de Contraste , Dextranos , Óxido Ferrosoférrico , Humanos , Infarto de la Arteria Cerebral Media/diagnóstico , Infarto de la Arteria Cerebral Media/patología , Hierro , Imagen por Resonancia Magnética , Nanopartículas de Magnetita , Masculino , Óxidos , Protaminas , Ratas , Ratas Sprague-Dawley
18.
J Neurol Sci ; 268(1-2): 40-7, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18054961

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a progressive disease which is caused by degeneration of motor neurons in the central nervous system. The incidence of ALS is higher in men than women, but the female advantage disappears with increased age. Here, we report evidence that the female advantage is due to the protective role of estrogen. In an ALS mouse model carrying the human Cu/Zn superoxide dismutase (hSOD1) G93A transgene, ovariectomy did not alter the onset age of the disease while reducing the female lifespan by 7 days and making it comparable to that of the male transgenic mice. Treatment of ovariectomized females with 17beta-estradiol (E2) did not delay the onset of disease, but prevented progression of ALS motor dysfunctions as shown by extension reflex test for a limited time window. Importantly, E2 treatment rescued the lifespans in overiectomized females. These findings will provide important new insights to interpretation of disease progression in post-menopausal female ALS patients.


Asunto(s)
Esclerosis Amiotrófica Lateral , Estradiol/uso terapéutico , Estrógenos/uso terapéutico , Actividad Motora/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Superóxido Dismutasa/genética , Edad de Inicio , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/mortalidad , Esclerosis Amiotrófica Lateral/fisiopatología , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Tamaño de los Órganos/efectos de los fármacos , Ovariectomía/métodos , Probabilidad , Tiempo de Reacción/efectos de los fármacos , Factores Sexuales
19.
J Cereb Blood Flow Metab ; 27(6): 1142-51, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17106444

RESUMEN

Excitotoxicity and oxidative stress mediate neuronal death after hypoxic-ischemic brain injury. We examined the possibility that targeting both N-methyl-D-aspartate (NMDA) receptor-mediated excitotoxicity and oxidative stress would result in enhanced neuroprotection against hypoxic-ischemia. 2-Hydroxy-5-(2,3,5,6-tetrafluoro-4-trifluoromethyl-benzylamino)-benzoic acid (Neu2000) was derived from aspirin and sulfasalazine to prevent both NMDA neurotoxicity and oxidative stress. In cortical cell cultures, Neu2000 was shown to be an uncompetitive NMDA receptor antagonist and completely blocked free radical toxicity at doses as low as 0.3 micromol/L. Neu2000 showed marked neuroprotection in a masked fashion using histology and behavioral testing in two rodent models of focal cerebral ischemia without causing neurotoxic side effects. Neu2000 protected against the effects of middle cerebral artery occlusion, even when delivered 8 h after reperfusion. Single bolus administration of the drug prevented gray and white matter degeneration and spared neurologic function for over 28 days after MACO. Neu2000 may be a novel therapy for combating both NMDA receptor-mediated excitotoxicity and oxidative stress, the two major routes of neuronal death in ischemia, offering profound neuroprotection and an extended therapeutic window.


Asunto(s)
Antioxidantes/farmacología , Benzoatos/farmacología , Isquemia Encefálica/prevención & control , N-Metilaspartato/antagonistas & inhibidores , Animales , Aspirina/química , Benzoatos/uso terapéutico , Isquemia Encefálica/tratamiento farmacológico , Células Cultivadas , Antagonistas de Aminoácidos Excitadores/química , Antagonistas de Aminoácidos Excitadores/farmacología , Fluorobencenos , Infarto de la Arteria Cerebral Media , Ratones , Estrés Oxidativo/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Salicilatos , Sulfasalazina/química , metaminobenzoatos
20.
Neuropharmacology ; 50(1): 1-15, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16169564

RESUMEN

Sulfasalazine (SULFA), of anti-inflammatory drugs, shows a protective action against NMDA-induced neuronal toxicity. Here, we used an electrophysiological study of the pharmacological effects of SULFA on NMDA receptors to examine the molecular mechanisms underlying the neuroprotective role of SULFA. The drug acted as a typical noncompetitive inhibitor with neither agonist- nor use-dependency, and antagonized NMDA-evoked responses in a voltage-independent manner, suggesting that SULFA is not an open channel blocker. Noise and single channel analyses showed that SULFA-blocked NMDA responses by reducing the number of NMDA channels available for activation, and also reduced the channel open probability without changing single channel conductance. Moreover, SULFA accelerated NMDA desensitization without affecting the affinity of the receptor for NMDA or glutamate. Taken together, these data indicate that SULFA blocks the NMDA response by reducing the number of NMDA channels available for activation. This appears to occur via a SULFA-induced decrease in the channel open probability, and a concomitant acceleration of the desensitization response, which is likely associated with a reduced affinity for glycine. SULFA indeed decreased the glycine-potentiated NMDA response without binding directly to the glycine site. Our results suggest that SULFA acts as a noncompetitive NMDA receptor antagonist with an allosteric glycine modulation.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Corteza Cerebral/citología , Neuronas/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sulfasalazina/farmacología , Algoritmos , Animales , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Electrofisiología , Glicina/metabolismo , Técnicas In Vitro , Cinética , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Técnicas de Placa-Clamp
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA