Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
1.
J Stroke Cerebrovasc Dis ; 33(7): 107738, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38701940

RESUMEN

OBJECTIVES: Edaravone dexborneol is neuroprotective against ischemic stroke, with free radical-scavenging and anti-inflammatory effects, but its effects in hemorrhagic stroke remain unclear. We evaluated whether edaravone dexborneol has a neuroprotective effect in intracerebral hemorrhage, and its underlying mechanisms. MATERIALS AND METHODS: Bioinformatics were used to predict the pathway of action of edaravone dexborneol. An intracerebral hemorrhage model was established using type IV collagenase in edaravone dexborneol, intracerebral hemorrhage, Sham, adeno-associated virus + edaravone dexborneol, and adeno-associated virus + intracerebral hemorrhage groups. The modified Neurological Severity Score was used to evaluate neurological function in rats. Brain water content was measured using the dry-wet weight method. Tumor necrosis factor-α, interleukin-1ß, inducible nitric oxide synthase, and γ-aminobutyric acid levels were determined by enzyme-linked immunosorbent assay. The expression levels of neurofilament light chain and γ-aminobutyric acid transaminase were determined by western blot. Nissl staining was used to examine neuronal morphology. Cognitive behavior was evaluated using a small-animal treadmill. RESULTS: Edaravone dexborneol alleviated neurological defects, improved cognitive function, and reduced cerebral edema, neuronal degeneration, and necrosis in rats with cerebral hemorrhage. The expression levels of neurofilament light chain, tumor necrosis factor-α, interleukin-1ß, inducible nitric oxide synthase, and γ-aminobutyric acid were decreased, while γ-aminobutyric acid transaminase expression was up-regulated. CONCLUSIONS: Edaravone dexborneol regulates γ-aminobutyric acid content by acting on the γ-aminobutyric acid transaminase signaling pathway, thus alleviating oxidative stress, neuroinflammation, neuronal degeneration, and death caused by excitatory toxic injury of neurons after intracerebral hemorrhage.


Asunto(s)
Edema Encefálico , Modelos Animales de Enfermedad , Edaravona , Interleucina-1beta , Fármacos Neuroprotectores , Ratas Sprague-Dawley , Animales , Edaravona/farmacología , Masculino , Fármacos Neuroprotectores/farmacología , Interleucina-1beta/metabolismo , Edema Encefálico/patología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/metabolismo , Edema Encefálico/enzimología , Edema Encefálico/prevención & control , 4-Aminobutirato Transaminasa/metabolismo , 4-Aminobutirato Transaminasa/antagonistas & inhibidores , Conducta Animal/efectos de los fármacos , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Hemorragia Cerebral/enzimología , Antiinflamatorios/farmacología , Cognición/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/metabolismo , Encéfalo/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Mediadores de Inflamación/metabolismo
2.
J Cell Physiol ; 234(4): 5304-5318, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30216439

RESUMEN

Inflammation and apoptosis are two key factors contributing to secondary brain injury after intracerebral hemorrhage (ICH). In the present study, we explored the neuroprotective role of methylene blue (MB) in ICH rats and studied the potential mechanisms involved. Rats were subjected to local injection of collagenase IV in the striatum or sham surgery. We observed that MB treatment could exert a neuroprotective effect on ICH by promoting neurological scores, decreasing the brain water content, alleviating brain-blood barrier disruption, and improving the histological damages in the perihematomal areas. Furthermore, we demonstrated that the various mechanisms underlying MB's neuroprotective effects linked to inhibited apoptosis and inhibited neuroinflammation. In addition, wortmannin, a selective inhibitor of phosphoinositide 3-kinase (PI3K), could reverse the antiapoptotic and anti-inflammatory effects of MB, which suggested that the PI3K-Akt pathway played an important role. In conclusion, these data suggested that MB could inhibit apoptosis and ameliorate neuroinflammation after ICH, and its neuroprotective effects might be exerted via the activation of the PI3K/Akt/GSK3ß pathway.


Asunto(s)
Antiinflamatorios/farmacología , Encéfalo/efectos de los fármacos , Hemorragia Cerebral/tratamiento farmacológico , Encefalitis/prevención & control , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Azul de Metileno/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/enzimología , Barrera Hematoencefálica/patología , Encéfalo/enzimología , Encéfalo/patología , Edema Encefálico/enzimología , Edema Encefálico/patología , Edema Encefálico/prevención & control , Permeabilidad Capilar/efectos de los fármacos , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/enzimología , Encefalitis/etiología , Encefalitis/patología , Masculino , Microglía/efectos de los fármacos , Microglía/enzimología , Microglía/patología , Neuronas/enzimología , Neuronas/patología , Infiltración Neutrófila/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal
3.
J Stroke Cerebrovasc Dis ; 28(6): 1718-1725, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30922669

RESUMEN

OBJECTIVE: Intracerebral hemorrhage affects approximately 2 million individuals per year. While the incidence is roughly equal in men and women, few studies have examined the influence of sex on secondary injury and associated long-term functional outcomes. Matrix metalloproteinases (MMPs) promote vessel rupture and worsen outcomes by potentiating blood-brain barrier breakdown after injury. We hypothesized that different MMP isoform levels would be predictive of injury severity, secondary injury, and long-term functional outcomes in males and females, respectively. METHODS: We examined the levels of MMP isoforms in serum samples from a prospective patient biobank (n = 55). Baseline clinical, radiographic, and laboratory data were also analyzed. RESULTS: We found that MMP-1 (P = .036), MMP-2 (P = .014), MMP-3 (P < .001), and MMP-9 (P = .02) levels gradually increased over time in male patients until 10 DPI. In female patients, we found a different pattern of activation: MMP-8 (P = .02) was the only isoform that significantly changed with time, which reached a peak at 3-5 days postinjury. Several MMP isoforms correlated with markers of secondary injury in female patients (all P < .05). Additionally, serum levels of MMP-3 (P = .011) in males and MMP-10 (P = .044) in females were significantly associated with long-term functional outcomes in a sex-specific manner. CONCLUSIONS: This is the first sex-specific study to examine serum MMP levels and their correlation with clinicoradiologic measures after intracerebral hemorrhage, and identifies potential biomarkers of secondary injury and long-term outcomes in both sexes.


Asunto(s)
Hemorragia Cerebral/enzimología , Metaloproteinasas de la Matriz/sangre , Adulto , Anciano , Biomarcadores/sangre , Hemorragia Cerebral/sangre , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/diagnóstico por imagen , Bases de Datos Factuales , Evaluación de la Discapacidad , Edema/sangre , Edema/enzimología , Edema/etiología , Femenino , Escala de Coma de Glasgow , Humanos , Puntaje de Gravedad del Traumatismo , Isoenzimas , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo , Factores Sexuales , Factores de Tiempo , Tomografía Computarizada por Rayos X
4.
J Stroke Cerebrovasc Dis ; 27(12): 3563-3569, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30201457

RESUMEN

BACKGROUND: Fabry disease is an X-linked lysosomal storage disorder caused by mutations in GLA, which encodes the enzyme α-galactosidase A (α-Gal A). Although the prevalence of Fabry disease in patients with stroke has been reported to range from 0% to 4%, few cohort studies have examined Japanese stroke patients. We aimed to clarify the prevalence of Fabry disease and the frequency of GLA mutations among patients with young-onset stroke in Japan. METHODS: From April 2015 to December 2016, we enrolled patients with young-onset (≤60 years old) ischemic stroke or intracerebral hemorrhage. We measured α-Gal A activity and the concentration of globotriaosylsphingosine in plasma. Genetic evaluations were performed in patients with low α-Gal A activity or high concentrations of globotriaosylsphingosine. RESULTS: Overall, 516 patients (median age of onset, 52 years old; 120 women) were consecutively enrolled in this study. Five patients (4 men and 1 woman) had low α-Gal A activity, and no patients were detected with the screen for plasma globotriaosylsphingosine levels. The genetic analysis did not identify a causative mutation responsible for classic Fabry disease in any of the patients, but 2 patients (.4%) carried the p.E66Q in GLA. CONCLUSIONS: No patient with Fabry disease was detected in our young-onset stroke cohort.


Asunto(s)
Isquemia Encefálica/sangre , Hemorragia Cerebral/sangre , Enfermedad de Fabry/sangre , Glucolípidos/sangre , Esfingolípidos/sangre , Accidente Cerebrovascular/sangre , alfa-Galactosidasa/sangre , Adulto , Edad de Inicio , Isquemia Encefálica/enzimología , Isquemia Encefálica/epidemiología , Isquemia Encefálica/genética , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/epidemiología , Hemorragia Cerebral/genética , Enfermedad de Fabry/enzimología , Enfermedad de Fabry/genética , Femenino , Humanos , Japón , Masculino , Persona de Mediana Edad , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/genética , Adulto Joven , alfa-Galactosidasa/genética
5.
J Stroke Cerebrovasc Dis ; 27(7): 1905-1913, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29598904

RESUMEN

BACKGROUND: Cerebral microhemorrhage (CMH) is a neuropathological term that could be easily found in cerebral amyloid angiopathy, intracerebral hemorrhages, etc. CMHs could be detected clearly in vivo by magnetic resonance imaging (MRI)-susceptibility-weighted imaging or MRI T2* scan. This terminology is now accepted in the area of neuroimaging. CMHs are quite common in elderly patients and are associated with several other neuropsychiatric disorders. The causes of CMHs are complicated, and neuroinflammation is considered as one of the well-accepted mechanical factors. This study investigated whether lipopolysaccharide (LPS)-induced CMHs occur through the regulation of nitric oxide synthase (NOS) isoforms and reveals the exact underlying mechanism of LPS-induced CMHs. METHODS: Our work successfully developed a subacute model of CMHs in rats. LPS was intraperitoneally injected into rats at 0, 6, and 24 hours, which induced typical CMH features 7 days after the injection. These could be detected on the brain surface or parenchyma by hematoxylin and eosin staining and MRI. RESULTS: LPS-treated rats showed significant activation of astrocytes and microglia, as well as loss of pericytes and disruption of blood-brain barrier. Meanwhile, both astrocytes and microglia were positively correlated with CMH numbers. Furthermore, the expressions of NOS isoforms were also examined, and the levels of neuronal NOS and endothelial NOS were found to be elevated. CONCLUSIONS: These results implied that the NOS isoforms might be involved in the subacute model of CMHs in rats induced by LPS.


Asunto(s)
Encéfalo/enzimología , Hemorragia Cerebral/enzimología , Modelos Animales de Enfermedad , Óxido Nítrico Sintasa/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Permeabilidad Capilar/fisiología , Hemorragia Cerebral/diagnóstico por imagen , Hemorragia Cerebral/patología , Inflamación/diagnóstico por imagen , Inflamación/enzimología , Inflamación/patología , Isoenzimas/metabolismo , Lipopolisacáridos , Masculino , Microglía/enzimología , Microglía/patología , Neuroinmunomodulación/fisiología , Neuronas/enzimología , Neuronas/patología , Ratas Sprague-Dawley , Tirosina/análogos & derivados , Tirosina/metabolismo
6.
Stroke ; 48(2): 445-451, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28057806

RESUMEN

BACKGROUND AND PURPOSE: For stroke prevention, patients with atrial fibrillation typically receive oral anticoagulation. The commonly used anticoagulant warfarin increases the risk of hemorrhagic transformation (HT) when a stroke occurs; tissue-type plasminogen activator treatment is therefore restricted in these patients. This study was designed to test the hypothesis that 12/15-lipoxygenase (12/15-LOX) inhibition would reduce HT in warfarin-treated mice subjected to experimental stroke. METHODS: Warfarin was dosed orally in drinking water, and international normalized ratio values were determined using a Coaguchek device. C57BL6J mice or 12/15-LOX knockout mice were subjected to transient middle cerebral artery occlusion with 3 hours severe ischemia (model A) or 2 hours ischemia and tissue-type plasminogen activator infusion (model B), with or without the 12/15-LOX inhibitor ML351. Hemoglobin was determined in brain homogenates, and hemorrhage areas on the brain surface and in brain sections were measured. 12/15-LOX expression was detected by immunohistochemistry. RESULTS: Warfarin treatment resulted in reproducible increased international normalized ratio values and significant HT in both models. 12/15-LOX knockout mice suffered less HT after severe ischemia, and ML351 reduced HT in wild-type mice. When normalized to infarct size, ML351 still independently reduced hemorrhage. HT after tissue-type plasminogen activator was similarly reduced by ML351. CONCLUSIONS: In addition to its benefits in infarct size reduction, 12/15-LOX inhibition also may independently reduce HT in warfarin-treated mice. ML351 should be further evaluated as stroke treatment in anticoagulated patients suffering a stroke, either alone or in conjunction with tissue-type plasminogen activator.


Asunto(s)
Anticoagulantes/toxicidad , Araquidonato 12-Lipooxigenasa/deficiencia , Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/deficiencia , Araquidonato 15-Lipooxigenasa/metabolismo , Hemorragia Cerebral/enzimología , Accidente Cerebrovascular/enzimología , Warfarina/toxicidad , Animales , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Accidente Cerebrovascular/tratamiento farmacológico
7.
Stroke ; 48(11): 3108-3116, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29018127

RESUMEN

BACKGROUND AND PURPOSE: Intracerebral hemorrhage (ICH) is a subtype of stroke with highest mortality and morbidity. Pronounced inflammation plays a significant role in the development of the secondary brain injury after ICH. Recently, SIK-2 (salt-inducible kinase-2) was identified as an important component controlling inflammatory response. Here we sought to investigate the role of SIK-2 in post-ICH inflammation and potential protective effects of SIK-2 inhibition after ICH. METHODS: Two hundred and ninety-three male CD-1 mice were used. ICH was induced via injection of 30 µL of autologous blood. Recombinant SIK-2 was administrated 1 hour after ICH intracerebroventricularly. SIK-2 small interfering RNA was injected intracerebroventricularly 24 hours before ICH. Bosutinib, a clinically approved tyrosine kinase inhibitor with affinity to SIK-2, was given intranasally 1 hour or 6 hours after ICH. Effects of treatments were evaluated by neurological tests and brain water content calculation. Molecular pathways were investigated by Western blots and immunofluorescence studies. RESULTS: Endogenous SIK-2 was expressed in microglia and neurons. SIK-2 expression was reduced after ICH. Exogenous SIK-2 aggravated post-ICH inflammation, leading to brain edema and the neurobehavioral deficits. SIK-2 inhibition attenuated post-ICH inflammation, reducing brain edema and ameliorating neurological dysfunctions. Bosutinib inhibited SIK-2-attenuating ICH-induced brain damage. Protective effects of Bosutinib were mediated, at least partly, by CRTC3 (cyclic amp-response element binding protein-regulated transcription coactivator 3)/cyclic amp-response element binding protein/NF-κB (nuclear factor-κB) pathway. CONCLUSIONS: SIK-2 participates in inflammation induction after ICH. SIK-2 inhibition via Bosutinib or small interfering RNA decreased inflammation, attenuating brain injury. SIK-2 effects are, at least partly, mediated by CRTC3-cyclic amp-response element binding protein-NF-κB signaling pathway.


Asunto(s)
Compuestos de Anilina/farmacología , Hemorragia Cerebral/tratamiento farmacológico , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Nitrilos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinolinas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/patología , Masculino , Ratones , Microglía/enzimología , Microglía/patología , Neuronas/enzimología , Neuronas/patología , Proteínas Serina-Treonina Quinasas/biosíntesis , Factores de Transcripción/metabolismo
8.
Neurobiol Dis ; 102: 140-146, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28323022

RESUMEN

Pharmacotherapies that increase CNS expression of heme oxygenase-1 (HO-1) and other antioxidant proteins have improved outcome in experimental models of spontaneous intracerebral hemorrhage (ICH). In order to more specifically investigate the relationship between HO-1 and ICH outcome, mice expressing human HO-1 driven by the glial fibrillary acidic protein (GFAP) promoter (GFAP·HMOX1 mice) were tested in a model of in situ parenchymal hemorrhage. Injection of collagenase into the striata of wild-type (WT) mice resulted in a 26.3% mortality rate, with deaths equally distributed between males and females. Mortality was reduced to 4.48% in GFAP·HMOX1 mice. Cell viability in the injected striata of surviving WT mice was reduced by about half at one week and was significantly increased in transgenics; this benefit persisted over a 22day observation period. Cell counts guided by design-based stereology indicated loss of ~40% of neurons in WT hemorrhagic striata at one week, which was decreased by half in transgenics; no significant differences in microglia or astrocyte numbers were observed. Blood-brain barrier disruption and short-term neurological deficits were also mitigated in GFAP·HMOX1 mice, but long-term outcome did not differ from that of WT survivors. These results suggest that astrocyte HO-1 overexpression provides robust neuroprotection after acute intracerebral hemorrhage. Further investigation of drug or genetic therapies that selectively increase astrocyte HO-1 is warranted.


Asunto(s)
Astrocitos/enzimología , Hemorragia Cerebral/enzimología , Hemo-Oxigenasa 1/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Permeabilidad Capilar/fisiología , Supervivencia Celular/fisiología , Hemorragia Cerebral/mortalidad , Hemorragia Cerebral/patología , Hemorragia Cerebral/psicología , Colagenasas , Cuerpo Estriado/enzimología , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hemo-Oxigenasa 1/genética , Humanos , Masculino , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Neuroprotección/fisiología
9.
J Neuroinflammation ; 14(1): 230, 2017 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-29178914

RESUMEN

BACKGROUND: Inflammatory responses significantly contribute to neuronal damage and poor functional outcomes following intracerebral hemorrhage (ICH). Soluble epoxide hydrolase (sEH) is known to induce neuroinflammatory responses via degradation of anti-inflammatory epoxyeicosatrienoic acids (EET), and sEH is upregulated in response to brain injury. The present study investigated the involvement of sEH in ICH-induced neuroinflammation, brain damage, and functional deficits using a mouse ICH model and microglial cultures. METHODS: ICH was induced by injecting collagenase in both wild-type (WT) C57BL/6 mice and sEH knockout (KO) mice. WT mice were injected intracerebroventricularly with 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA), a selective sEH inhibitor, 30 min before ICH. Expression of sEH in the hemorrhagic hemisphere was examined by immunofluorescence and Western blot analysis. The effects of genetic deletion or pharmacological inhibition of sEH by AUDA on neuroinflammatory responses, EET degradation, blood-brain barrier (BBB) permeability, histological damage, and functional deficits were evaluated. The anti-inflammatory mechanism of sEH inactivation was investigated in thrombin- or hemin-stimulated cultured microglia. RESULTS: ICH induced an increase in sEH protein levels in the hemorrhagic hemisphere from 3 h to 4 days. sEH was expressed in microglia/macrophages, astrocytes, neurons, and endothelial cells in the perihematomal region. Genetic deletion of sEH significantly attenuated microglia/macrophage activation and expression of inflammatory mediators and reduced EET degradation at 1 and 4 days post-ICH. Deletion of sEH also reduced BBB permeability, matrix metalloproteinase (MMP)-9 activity, neutrophil infiltration, and neuronal damage at 1 and 4 days. Likewise, administration of AUDA attenuated proinflammatory microglia/macrophage activation and EET degradation at 1 day post-ICH. These findings were associated with a reduction in functional deficits and brain damage for up to 28 days. AUDA also ameliorated neuronal death, BBB disruption, MMP-9 activity, and neutrophil infiltration at 1 day. However, neither gene deletion nor pharmacological inhibition of sEH altered the hemorrhage volume following ICH. In primary microglial cultures, genetic deletion or pharmacological inhibition of sEH by AUDA reduced thrombin- and hemin-induced microglial activation. Furthermore, AUDA reduced thrombin- and hemin-induced P38 MAPK and NF-κB activation in BV2 microglia cultures. Ultimately, AUDA attenuated N2A neuronal death that was induced by BV2 microglial conditioned media. CONCLUSIONS: Our results suggest that inhibition of sEH may provide a potential therapy for ICH by suppressing microglia/macrophage-mediated neuroinflammation.


Asunto(s)
Lesiones Encefálicas/enzimología , Hemorragia Cerebral/patología , Epóxido Hidrolasas/metabolismo , Inflamación/enzimología , Animales , Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/enzimología , Inflamación/etiología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
10.
J Neuroinflammation ; 14(1): 114, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28583195

RESUMEN

BACKGROUND: Cerebral microbleeds (CMB) are MRI-demonstrable cerebral microhemorrhages (CMH) which commonly coexist with ischemic stroke. This creates a challenging therapeutic milieu, and a strategy that simultaneously protects the vessel wall and provides anti-thrombotic activity is an attractive potential approach. Phosphodiesterase 3A (PDE3A) inhibition is known to provide cerebral vessel wall protection combined with anti-thrombotic effects. As an initial step in the development of a therapy that simultaneously treats CMB and ischemic stroke, we hypothesized that inhibition of the PDE3A pathway is protective against CMH development. METHODS: The effect of PDE3A pathway inhibition was studied in the inflammation-induced and cerebral amyloid angiopathy (CAA)-associated mouse models of CMH. The PDE3A pathway was modulated using two approaches: genetic deletion of PDE3A and pharmacological inhibition of PDE3A by cilostazol. The effects of PDE3A pathway modulation on H&E- and Prussian blue (PB)-positive CMH development, BBB function (IgG, claudin-5, and fibrinogen), and neuroinflammation (ICAM-1, Iba-1, and GFAP) were investigated. RESULTS: Robust development of CMH in the inflammation-induced and CAA-associated spontaneous mouse models was observed. Inflammation-induced CMH were associated with markers of BBB dysfunction and inflammation, and CAA-associated spontaneous CMH were associated primarily with markers of neuroinflammation. Genetic deletion of the PDE3A gene did not alter BBB function, microglial activation, or CMH development, but significantly reduced endothelial and astrocyte activation in the inflammation-induced CMH mouse model. In the CAA-associated CMH mouse model, PDE3A modulation via pharmacological inhibition by cilostazol did not alter BBB function, neuroinflammation, or CMH development. CONCLUSIONS: Modulation of the PDE3A pathway, either by genetic deletion or pharmacological inhibition, does not alter CMH development in an inflammation-induced or in a CAA-associated mouse model of CMH. The role of microglial activation and BBB injury in CMH development warrants further investigation.


Asunto(s)
Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/deficiencia , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Microvasos/efectos de los fármacos , Inhibidores de Fosfodiesterasa 3/uso terapéutico , Animales , Hemorragia Cerebral/enzimología , Cilostazol , Eliminación de Gen , Ratones , Ratones Noqueados , Ratones Transgénicos , Microvasos/enzimología , Microvasos/patología , Inhibidores de Fosfodiesterasa 3/farmacología , Tetrazoles , Resultado del Tratamiento
11.
Cell Tissue Res ; 368(1): 13-27, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27726026

RESUMEN

Recently, NIX, a pro-apoptotic BH3-only protein, was found to be a novel p75 neurotrophin receptor (p75NTR) binding protein by screening a human fetal brain two-hybrid library in our laboratory. We further study the interaction of these two proteins and the possible roles of p75NTR and NIX in intracerebral hemorrhage (ICH)-induced neuronal death. Using the split-ubiquitin yeast two-hybrid system, we found that the "Copper" domain in p75NTR and the TM region in NIX were sufficient for the interaction of these two proteins. Co-immunoprecipitation and in vitro binding assays demonstrated the direct interaction between p75NTR and NIX. NIX protein was stabilized by p75NTR at post-translational levels. Moreover, p75NTR was able to work together with NIX to promote apoptosis and affected the NIX-induced JNK-p53-Bax pathway in neuronal PC12 cells. Previous work has indicated that p75NTR and NIX are induced in neurons in human ICH and the rat ICH model, respectively. We confirm that both p75NTR and NIX levels were up-regulated in glutamate-treated primary cortical neurons (a cellular in vitro model for ICH) and in the rat ICH model. Glutamate exposure increased the association between p75NTR and NIX and elevated the activation of the JNK-p53-Bax pathway and neuronal apoptosis; all of these observations were similar in the rat ICH model. Importantly, p75NTR and NIX appeared to be involved in cortical neuronal apoptosis, because knockdown of p75NTR or NIX not only inhibited the JNK pathway but also impaired neuronal apoptosis. Thus, p75NTR and NIX may play critical roles in ICH-induced neuronal apoptosis in vitro and in vivo.


Asunto(s)
Apoptosis , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Neuronas/patología , Proteínas Proto-Oncogénicas/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/patología , Hemorragia Cerebral/enzimología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Ácido Glutámico/farmacología , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Proteínas de la Membrana/química , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/química , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso/química , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/química , Técnicas del Sistema de Dos Híbridos , Ubiquitina/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
12.
Cell Mol Neurobiol ; 37(2): 251-261, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27053350

RESUMEN

The hematopoietic cell kinase (Hck) is a member of the Src family protein kinases which regulates many signal transduction pathways including cell growth, proliferation, differentiation, migration, and apoptosis. However, the expression and function of Hck after intracerebral hemorrhage (ICH) are unknown. Western blot, immunohistochemistry, and immunofluorescence showed that Hck was obviously up-regulation in neurons adjacent to the hematoma after ICH. In addition, the temporary raise of Hck expression was paralleled with the expression of p53, Bax, and active caspase-3, suggesting that Hck was involved in neuronal apoptosis. Hck siRNA dramatically decrease hemin-induced expression of p53, Bax, and active caspase-3 as well as the amount of apoptotic SH-SY5Y cells in vitro. Furthermore, Hck interacted with p53. Hence, Hck might promote neuronal apoptosis via p53 signaling pathway after ICH.


Asunto(s)
Apoptosis/fisiología , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/patología , Neuronas/enzimología , Neuronas/patología , Proteínas Proto-Oncogénicas c-hck/biosíntesis , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
13.
Cell Mol Neurobiol ; 37(5): 831-841, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27568302

RESUMEN

Isocitrate dehydrogenase 1 (IDH1), one member of the IDH family can convert isocitrate to α-ketoglutarate (α-KG) via oxidative decarboxylation. IDH1 and IDH2 mutations have been identified in multiple tumor types and the mutations confer neomorphic activity in the mutant protein, resulting in the conversion of α-KG to the oncometabolite, D-2-hydroxyglutarate (2-HG). The subsequent accumulation of 2-HG results in epigenetic dysregulation via inhibition of α-KG-dependent histone and DNA demethylase. And the glutamate levels are reduced in IDH mutant cells compared to wild-type. We have known that diffuse gliomas contain a high frequency of mutations in the IDH1 gene. However, the expression of IDH1 and its roles in Intracranial hemorrhage (ICH) remain largely unknown. We observed increased expression of IDH1 in neurons after intracerebral hemorrhage. Up-regulation of IDH1 was found to be accompanied by the increased expression of active caspase-3 and pro-apoptotic Bcl-2-associated X protein and decreased expression of anti-apoptotic protein B cell lymphoma-2 in vivo and vitro studies. So we hypothesized that IDH1 was involved in the regulation of neuronal apoptosis. The present research for the first time detected the expression and variation of IDH1 surrounding the hematoma, and all data proved the involvement of IDH1 in neuronal apoptosis following ICH.


Asunto(s)
Envejecimiento/patología , Apoptosis , Encéfalo/patología , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/patología , Isocitrato Deshidrogenasa/metabolismo , Neuronas/enzimología , Neuronas/patología , Animales , Conducta Animal , Biomarcadores/metabolismo , Western Blotting , Modelos Animales de Enfermedad , Masculino , Células PC12 , Ratas , Ratas Sprague-Dawley
14.
Cell Mol Neurobiol ; 37(3): 427-435, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27114249

RESUMEN

Ubiquitinating enzymes catalyze protein ubiquitination, a reversible process countered by deubiquitinating enzyme (DUB) action. Ubiquitin-specific protease 4 (USP4) is a member of the ubiquitin-specific protease (USP) family of DUBs that has a role in spliceosome regulation. In the present study, we demonstrated that USP4 may be involved in neuronal apoptosis in the processes of intracerebral hemorrhage (ICH). We obtained a significant up-regulation of USP4 in neurons adjacent to the hematoma following ICH by the results of Western blot, immunohistochemistry, and immunofluorescence. Increasing USP4 level was found to be accompanied by the up-regulation of active caspase-3, γH2AX, Bax, and decreased expression of Bcl-2. In addition, USP4 co-localized well with γH2AX in the nucleus in the ICH model and hemin-induced apoptosis model. Moreover, in vitro study, knocking down USP4 by USP4-specific siRNA in PC12 cells reduced active caspase-3 expression. All these results above suggested that USP4 may be involved in neuronal apoptosis after ICH.


Asunto(s)
Envejecimiento/metabolismo , Apoptosis , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/patología , Neuronas/enzimología , Neuronas/patología , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Biomarcadores/metabolismo , Técnica del Anticuerpo Fluorescente , Masculino , Fenotipo , Ratas Sprague-Dawley , Ubiquitina-Proteína Ligasas
15.
Cell Mol Neurobiol ; 37(3): 527-536, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27316643

RESUMEN

Contrary to cell cycle-associated cyclin-dependent kinases, CDK5 is best known for its regulation of signaling processes in regulating mammalian CNS development. Studies of CDK5 have focused on its phosphorylation, although the diversity of CDK5 functions in the brain suggests additional forms of regulation. Here we expanded on the functional roles of CDK5 glycosylation in neurons. We showed that CDK5 was dynamically modified with O-GlcNAc in response to neuronal activity and that glycosylation represses CDK5-dependent apoptosis by impairing its association with p53 pathway. Blocking glycosylation of CDK5 alters cellular function and increases neuronal apoptosis in the cell model of the ICH. Our findings demonstrated a new role for O-glycosylation in neuronal apoptosis and provided a mechanistic understanding of how glycosylation contributes to critical neuronal functions. Moreover, we identified a previously unknown mechanism for the regulation of activity-dependent gene expression, neural development, and apoptosis.


Asunto(s)
Acetilglucosamina/metabolismo , Apoptosis , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/patología , Quinasa 5 Dependiente de la Ciclina/metabolismo , Neuronas/enzimología , Neuronas/patología , Animales , Apoptosis/efectos de los fármacos , Glicosilación/efectos de los fármacos , Células HEK293 , Hemina/farmacología , Humanos , Modelos Biológicos , Neuronas/efectos de los fármacos , Células PC12 , Ratas
16.
Toxicol Mech Methods ; 27(1): 18-23, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27841083

RESUMEN

CONTEXT: Studies have shown that thrombin activation played a central role in cell injuries associated with intracerebral hemorrhage (ICH). OBJECTIVE: Here, our study investigated the cytotoxicity of thrombin on neurons, and determined the involvement of JNK pathways in thrombin-induced neuronal apoptosis. MATERIALS AND METHODS: Primary cultured neurons were treated with different doses of thrombin. Some neurons were given either SP600125 or vehicle. LDH release assay and flow cytometry were used to measure neuronal apoptosis caused by thrombin. The activation of JNK and capases-3 were measured by Western blot. RESULTS: Our results showed large doses of thrombin that increased the LDH release, the level of cleaved caspase-3 and apoptosis rate of neurons. JNK was activated by thrombin in a time-dependent manner. Administration of SP600125 protects neurons from thrombin-induced apoptosis. CONCLUSION: These data indicate that the activation of JNK is crucial for thrombin-induced neuronal apoptosis, and inhibition of JNK may be a potential therapeutic target for ICH.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Trombina/farmacología , Animales , Antracenos/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Hemorragia Cerebral/enzimología , Hemorragia Cerebral/patología , Relación Dosis-Respuesta a Droga , Activación Enzimática , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Neuronas/enzimología , Neuronas/patología , Cultivo Primario de Células , Ratas , Trombina/fisiología
17.
Stroke ; 47(3): 843-51, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26839355

RESUMEN

BACKGROUND AND PURPOSE: Acute hyperglycemia worsens the clinical outcomes and exacerbates cerebral hemorrhage after stroke. The mediators of hemorrhagic transformation (HT) in hyperglycemic stroke are not fully understood. Matrix metalloproteinase 3 (MMP3) plays a critical role in the tissue-type plasminogen activator-induced HT. However, the role of MMP3 in exacerbating the HT and worsening the functional outcomes in hyperglycemic stroke remains unknown. METHODS: Control/normoglycemic and hyperglycemic (blood glucose, 140-200 mg/dL) male Wistar rats were subjected to middle cerebral artery occlusion for 90 minutes and either 24 hours or 7 days reperfusion. MMP3 was inhibited pharmacologically (UK 356618, 15 mg/kg IV at reperfusion) or knocked down in the brain by shRNA lentiviral particles (injected intracerebroventricular). Neurovascular injury was assessed at 24 hours, and functional outcomes were assessed at 24 hours, day 3, and day 7. MMP3 activity was measured in brain homogenate and cerebral macrovessels. Localization of MMP3 within the neurovascular unit after hyperglycemic stroke was demonstrated by immunohistochemistry. RESULTS: Hyperglycemia significantly increased MMP3 activity in the brain after stroke, and this was associated with exacerbated HT and worsened functional outcomes. MMP3 inhibition significantly reduced HT and improved functional outcomes. CONCLUSIONS: MMP3 plays a critical role in mediating cerebrovascular injury in hyperglycemic stroke. Our findings point out MMP3 as a potential therapeutic target in hyperglycemic stroke.


Asunto(s)
Hemorragia Cerebral/enzimología , Hiperglucemia/enzimología , Metaloproteinasa 3 de la Matriz/biosíntesis , Recuperación de la Función/fisiología , Accidente Cerebrovascular/enzimología , Animales , Hemorragia Cerebral/patología , Técnicas de Silenciamiento del Gen/métodos , Hiperglucemia/patología , Masculino , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratas , Ratas Wistar , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/patología , Resultado del Tratamiento
18.
Neurochem Res ; 41(12): 3308-3321, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27662850

RESUMEN

The JNKs have been implicated in a variety of biological functions in mammalian cells, including apoptosis and the responses to stress. However, the physiological role of these pathways in the intracerebral hemorrhage (ICH) has not been fully elucidated. In this study, we identified a MAPK kinase kinase (MAPKKK), MEKK1, may be involved in neuronal apoptosis in the processes of ICH through the activation of JNKs. From the results of western blot, immunohistochemistry and immunofluorescence, we obtained a significant up-regulation of MEKK1 in neurons adjacent to the hematoma following ICH. Increasing MEKK1 level was found to be accompanied with the up-regulation of p-JNK 3, p53, and c-jun. Besides, MEKK1 co-localized well with p-JNK in neurons, indicating its potential role in neuronal apoptosis. What's more, our in vitro study, using MEKK1 siRNA interference in PC12 cells, further confirmed that MEKK1 might exert its pro-apoptotic function on neuronal apoptosis through extrinsic pathway. Thus, MEKK1 may play a role in promoting the brain damage following ICH.


Asunto(s)
Apoptosis , Ganglios Basales/enzimología , Hemorragia Cerebral/enzimología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Neuronas/enzimología , Animales , Hemorragia Cerebral/patología , Hemorragia Cerebral/fisiopatología , Masculino , Neuronas/patología , Ratas Sprague-Dawley
19.
Mol Cell Biochem ; 422(1-2): 171-180, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27629786

RESUMEN

OTUB1 is a member of deubiquitinating enzymes, which was shown as a proteasome-associated DUB to be involved in the proteins Ub-dependent degradation. Previous studies have indicated that OTUB1 was expressed in brain. But its distribution and function in the brain remain unclear. In this study, we explored the roles of OTUB1 protein in the pathophysiology of intracerebral hemorrhage (ICH). From the results of Western blot, immunohistochemistry, and immunofluorescence, we found an obvious up-regulation of OTUB1 in neurons adjacent to the hematoma after ICH. Furthermore, we also found that the increase of OTUB1 expression was accompanied by the enhanced expression of Bax and active caspase-3, and decreased expression of Bcl-2 in the pathological process of rat ICH. What's more, our in vitro study, using OTUB1 RNA interference in PC12 cells, suggested that OTUB1 might exert its anti-apoptotic function in neuronal apoptosis. Therefore, OTUB1 may play a role in protecting the brain from secondary damage following ICH.


Asunto(s)
Apoptosis , Hemorragia Cerebral/enzimología , Endopeptidasas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Neuronas/enzimología , Animales , Hemorragia Cerebral/patología , Masculino , Neuronas/patología , Células PC12 , Ratas , Ratas Sprague-Dawley
20.
Stroke ; 46(4): 1093-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25690543

RESUMEN

BACKGROUND AND PURPOSE: Heme oxygenase-1 (HO-1) catalyzes the rate-limiting reaction of heme breakdown and may have both antioxidant and pro-oxidant effects. In previous studies, HO-1 overexpression protected astrocytes from heme-mediated injury in vitro. In the present study, we tested the hypothesis that selective astrocyte overexpression of HO-1 improves outcome after intracerebral hemorrhage. METHODS: Male and female transgenic mice overexpressing human HO-1 driven by the GFAP promoter (GFAP.HMOX1) and wild-type controls received striatal injections of autologous blood (25 µL). Blood-brain barrier disruption was assessed by Evans blue assay and striatal cell viability by methylthiazolyldiphenyl-tetrazolium bromide assay. Neurological deficits were quantified by digital analysis of spontaneous cage activity, adhesive removal, and elevated body swing tests. RESULTS: Mortality rate for wild-type mice was 34.8% and was similar for males and females; all GFAP.HMOX1 mice survived. Striatal Evans blue leakage at 24 hours was 23.4±3.2 ng in surviving wild-type mice, compared with 10.9±1.8 ng in transgenics. Perihematomal cell viability was reduced to 61±4% of contralateral at 3 days in wild-type mice, versus 80±4% in transgenics. Focal neurological deficits were significantly reduced and spontaneous cage activity was increased in GFAP.HMOX1 mice. CONCLUSIONS: Selective HO-1 overexpression in astrocytes reduces mortality, blood-brain barrier disruption, perihematomal cell injury, and neurological deficits in an autologous blood injection intracerebral hemorrhage model. Genetic or pharmacological therapies that acutely increase astrocyte HO-1 may be beneficial after intracerebral hemorrhage.


Asunto(s)
Astrocitos/enzimología , Hemorragia Cerebral/enzimología , Hemo-Oxigenasa 1/metabolismo , Animales , Femenino , Proteína Ácida Fibrilar de la Glía , Hemo-Oxigenasa 1/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA