Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Stroke Cerebrovasc Dis ; 31(3): 106281, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35026495

RESUMEN

OBJECTIVE: Hyperglycemia is often observed in the patients after acute stroke. This study aims to elucidate the potential effect and mechanism of hyperglycemia by screening microRNAs expression in intracerebral hemorrhage mice. METHODS: We employed the collagenase model of intracerebral hemorrhage. Twenty male C57BL/6 mice were used and randomly divided in normo- and hyperglycemic. The hyperglycemia was induced by intraperitoneally injection of 50% of Dextrose (8 mL/kg) 3 hours after intracerebral hemorrhage. The neurologic impairment was investigated by neurologic deficit scale. To study the specific mechanisms of hyperglycemia, microRNAs expression in perihematomal area was investigated by RNA sequencing. MicroRNAs expression in hyperglycemic intracerebral hemorrhage animals were compared normoglycemic mice. Functional annotation analysis was used to indicate potential pathological pathway, underlying observed effects. Finally, polymerase chain reaction validation was administered. RESULTS: Intraperitoneal injection of dextrose significantly increased blood glucose level. That was associated with aggravation of neurological deficits in hyperglycemic compared to normoglycemic animals. A total of 73 differentially expressed microRNAs were identified via transcriptomics analysis. Bioinformatics analyses showed that these microRNAs were significantly altered in several signaling pathways, of which the hedgehog signaling pathway was regarded as the most potential pathway associated with the effect of hyperglycemia on acute intracerebral hemorrhage. Furthermore, polymerase chain reaction results validated the correlation between microRNAs and hedgehog signaling pathway. CONCLUSIONS: MicroRNA elevated in hyperglycemia group may be involved in worsening the neurological function via inhibiting the hedgehog signaling, which provides a novel molecular physiological mechanism and lays the foundation for treatment of intracerebral hemorrhage.


Asunto(s)
Proteínas Hedgehog , MicroARNs , Transducción de Señal , Transcriptoma , Animales , Hemorragia Cerebral/genética , Modelos Animales de Enfermedad , Glucosa/toxicidad , Proteínas Hedgehog/metabolismo , Hiperglucemia/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Transcriptoma/genética
2.
Stroke ; 52(12): 4043-4053, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34807744

RESUMEN

BACKGROUND AND PURPOSE: Sirt5 (Sirtuin 5) desuccinylates multiple metabolic enzymes and plays an important role in maintaining energy homeostasis. The goal of this study was to determine whether Sirt5-mediated desuccinylation restores the energy metabolism and protects brain against subarachnoid hemorrhage (SAH). METHODS: Male C57BL/6 or Sirt5-/- mice were used. The endovascular perforation SAH model was applied. Protein lysine succinylation in the brain cortex was examined using liquid chromatography-tandem mass spectrometry analysis. The brain metabolism was evaluated by measurement of brain pH as well as ATP and reactive oxygen species level. Neuronal cell death and neurobehavioral deficits were assessed 24 hours after SAH. The expression and desuccinylation activity of Sirt5, lysine succinylation of citrate synthase and ATP synthase subunits were investigated by Western blot, immunohistochemistry, and ELISA in SAH mice and patients. Furthermore, the benefits of resveratrol-mediated Sirt5 activation were investigated. RESULTS: A total of 211 lysine succinylation sites were differentially expressed on 170 proteins in mice brain after SAH. Thirty-nine percent of these succinylated proteins were localized in mitochondria and they are related to energy metabolism. SAH caused a decrease of Sirt5 expression and succinylated citrate synthase as well as the subunits of ATP synthase, subsequently lowered brain pH, reduced ATP and increased reactive oxygen species production, leading to neuronal cell death, and neurological deficits. Knockdown of Sirt5 aggravated SAH-induced effects, mentioned above. Administration of resveratrol resulted in activation of Sirt5. The activation was accompanied both with restoration of the mitochondrial metabolism and alleviation of early brain injury as well as with desuccinylating citrate synthase and ATP synthase. CONCLUSIONS: Protein lysine succinylation is a biochemical hallmark of metabolic crisis after SAH, and disruption of lysine succinylation through activation of Sirt5 might be a promising therapeutic strategy for the treatment of SAH.


Asunto(s)
Metabolismo Energético/fisiología , Lisina/metabolismo , Mitocondrias/metabolismo , Sirtuinas/metabolismo , Hemorragia Subaracnoidea/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hemorragia Subaracnoidea/patología
3.
Stroke ; 50(11): 3246-3254, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31558140

RESUMEN

Background and Purpose- Perihemorrhagic edema (PHE) is associated with poor outcome after intracerebral hemorrhage (ICH). Infiltration of immune cells is considered a major contributor of PHE. Recent studies suggest that immunomodulation via S1PR (sphingosine-1-phosphate receptor) modulators improve outcome in ICH. Siponimod, a selective modulator of sphingosine 1-phosphate receptors type 1 and type 5, demonstrated an excellent safety profile in a large study of patients with multiple sclerosis. Here, we investigated the impact of siponimod treatment on perihemorrhagic edema, neurological deficits, and survival in a mouse model of ICH. Methods- ICH was induced by intracranial injection of 0.075 U of bacterial collagenase in 123 mice. Mice were randomly assigned to different treatment groups: vehicle, siponimod given as a single dosage 30 minutes after the operation or given 3× for 3 consecutive days starting 30 minutes after operation. The primary outcome of our study was evolution of PHE measured by magnetic resonance-imaging on T2-maps 72 hours after ICH, secondary outcomes included evolution of PHE 24 hours after ICH, survival and neurological deficits, as well as effects on circulating blood cells and body weight. Results- Siponimod significantly reduced PHE measured by magnetic resonance imaging (P=0.021) as well as wet-dry method (P=0.04) 72 hours after ICH. Evaluation of PHE 24 hours after ICH showed a tendency toward attenuated brain edema in the low-dosage group (P=0.08). Multiple treatments with siponimod significantly improved neurological deficits measured by Garcia Score (P=0.03). Survival at day 10 was improved in mice treated with multiple dosages of siponimod (P=0.037). Mice treated with siponimod showed a reduced weight loss after ICH (P=0.036). Conclusions- Siponimod (BAF-312) attenuated PHE after ICH, increased survival, and reduced ICH-induced sensorimotor deficits in our experimental ICH-model. Findings encourage further investigation of inflammatory modulators as well as the translation of BAF-312 to a human study of ICH patients.


Asunto(s)
Azetidinas/farmacología , Compuestos de Bencilo/farmacología , Edema Encefálico , Hemorragia Cerebral , Transducción de Señal/efectos de los fármacos , Animales , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/fisiopatología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/fisiopatología , Modelos Animales de Enfermedad , Masculino , Ratones , Receptores de Esfingosina-1-Fosfato/metabolismo
4.
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
5.
Stroke ; 48(6): 1655-1664, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28495827

RESUMEN

BACKGROUND AND PURPOSE: Energy depletion is a critical factor leading to cell death and brain dysfunction after ischemic stroke. In this study, we investigated whether energy depletion is involved in hyperglycemia-induced hemorrhagic transformation after ischemic stroke and determined the pathway underlying the beneficial effects of hyperbaric oxygen (HBO). METHODS: After 2-hour middle cerebral artery occlusion, hyperglycemia was induced by injecting 50% dextrose (6 mL/kg) intraperitoneally at the onset of reperfusion. Immediately after it, rats were exposed to HBO at 2 atmospheres absolutes for 1 hour. ATP synthase inhibitor oligomycin A, nicotinamide phosphoribosyl transferase inhibitor FK866, or silent mating type information regulation 2 homolog 1 siRNA was administrated for interventions. Infarct volume, hemorrhagic volume, and neurobehavioral deficits were recorded; the level of blood glucose, ATP, and nicotinamide adenine dinucleotide and the activity of nicotinamide phosphoribosyl transferase were monitored; the expression of silent mating type information regulation 2 homolog 1, acetylated p53, acetylated nuclear factor-κB, and cleaved caspase 3 were detected by Western blots; and the activity of matrix metalloproteinase-9 was assayed by zymography. RESULTS: Hyperglycemia deteriorated energy metabolism and reduced the level of ATP and nicotinamide adenine dinucleotide and exaggerated hemorrhagic transformation, blood-brain barrier disruption, and neurological deficits after middle cerebral artery occlusion. HBO treatment increased the levels of the ATP and nicotinamide adenine dinucleotide and consequently increased silent mating type information regulation 2 homolog 1, resulting in attenuation of hemorrhagic transformation, brain infarction, as well as improvement of neurological function in hyperglycemic middle cerebral artery occlusion rats. CONCLUSIONS: HBO induced activation of ATP/nicotinamide adenine dinucleotide/silent mating type information regulation 2 homolog 1 pathway and protected blood-brain barrier in hyperglycemic middle cerebral artery occlusion rats. HBO might be promising approach for treatment of acute ischemic stroke patients, especially patients with diabetes mellitus or treated with r-tPA (recombinant tissue-type plasminogen activator).


Asunto(s)
Adenosina Trifosfato/metabolismo , Isquemia Encefálica , Hemorragia Cerebral , Oxigenoterapia Hiperbárica/métodos , Hiperglucemia/metabolismo , Infarto de la Arteria Cerebral Media , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Sirtuina 1/metabolismo , Accidente Cerebrovascular , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/terapia , Modelos Animales de Enfermedad , Hiperglucemia/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/terapia , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/terapia
6.
J Neurochem ; 143(6): 750-760, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29030969

RESUMEN

Adropin is expressed in the CNS and plays a crucial role in the development of stroke. However, little is currently known about the effects of adropin on the blood-brain barrier (BBB) function after intracerebral hemorrhage (ICH). In this study, the role of adropin in collagenase-induced ICH was investigated in mice. At 1-h post-ICH, mice were administered with recombinant human adropin by intranasal. Brain water +content, BBB permeability, and neurological function were measured at different time intervals. Proteins were quantified using western blot analysis, and the localizations of adropin and Notch1 were visualized via immunofluorescence staining. It is shown that adropin reduced brain water content and improved neurological functions. Adropin preserved the functionality of BBB by increasing N-cadherin expression and reducing extravasation of albumin. Moreover, in vivo knockdown of Notch1 and Hes1 both abolished the protective effects of adropin. Taken together, our data demonstrate that adropin constitutes a potential treatment value for ICH by preserving BBB and improving functional outcomes through the Notch1 signaling pathway.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Barrera Hematoencefálica/fisiología , Hemorragia Cerebral/metabolismo , Péptidos/metabolismo , Receptor Notch1/metabolismo , Factor de Transcripción HES-1/metabolismo , Animales , Proteínas Sanguíneas/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Hemorragia Cerebral/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Péptidos/farmacología , Transducción de Señal/fisiología
7.
Stroke ; 47(5): 1319-27, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27006454

RESUMEN

BACKGROUND AND PURPOSE: Recombinant osteopontin (rOPN) has been reported to be neuroprotective in stroke animal models. The purpose of this study is to investigate a potential role and mechanism of nasal administration of rOPN on preserving the vascular smooth muscle phenotype in early brain injury after subarachnoid hemorrhage (SAH). METHODS: One hundred and ninety-two male adult Sprague-Dawley rats were used. The SAH model was induced by endovascular perforation. Integrin-linked kinase small interfering RNA was intracerebroventricularly injected 48 hours before SAH. The integrin receptor antagonist fibronectin-derived peptide Gly-Arg-Gly-Asp-Ser-Pro (GRGDSP), focal adhesion kinase inhibitor Fib-14, and Rac-1 inhibitor NSC23766 were administered 1 hour before SAH induction. rOPN was administered via the intracerebroventricular and nasal route after SAH. SAH grade, neurological scores, brain water content, brain swelling, hematoxylin and eosin staining, India ink angiography, Western blots, and immunofluorescence were used to study the mechanisms of rOPN on the vascular smooth muscle phenotypic transformation. RESULTS: The marker proteins of vascular smooth muscle phenotypic transformation α-smooth muscle actin decreased and embryonic smooth muscle myosin heavy chain (SMemb) increased significantly at 24 and 72 hours in the cerebral arteries after SAH. rOPN prevented the changes of α-smooth muscle actin and SMemb and significantly alleviated neurobehavioral dysfunction, increased the cross-sectional area and the lumen diameter of the cerebral arteries, reduced the brain water content and brain swelling, and improved the wall thickness of cerebral arteries. These effects of rOPN were abolished by GRGDSP, integrin-linked kinase small interfering RNA, and NSC23766. Intranasal application of rOPN at 3 hours after SAH also reduced neurological deficits. CONCLUSIONS: rOPN prevented the vascular smooth muscle phenotypic transformation and improved the neurological outcome, which was possibly mediated by the integrin receptor/integrin-linked kinase/Rac-1 pathway.


Asunto(s)
Arterias Cerebrales/efectos de los fármacos , Integrinas/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Osteopontina/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Hemorragia Subaracnoidea/tratamiento farmacológico , Proteína de Unión al GTP rac1/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Masculino , Fármacos Neuroprotectores/administración & dosificación , Osteopontina/administración & dosificación , Fenotipo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes
8.
Crit Care Med ; 44(6): e390-402, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26646459

RESUMEN

OBJECTIVE: Platelet-derived growth factor-BB activates platelet-derived growth factor receptor-ß and promotes vascular smooth muscle cell phenotypic transformation. Elevated levels of non-muscle myosin IIB (SMemb) are found in secretory smooth muscle cells along with inflammatory mediators, such as intercellular adhesion molecule-1, which can amplify neutrophil infiltration into the brain. In the present study, we investigated the role of platelet-derived growth factor-BB/platelet-derived growth factor receptor-ß following intracerebral hemorrhage-induced brain injury in mice, with emphasis on its ability to promote vascular smooth muscle cell phenotypic transformation followed by increased intercellular adhesion molecule-1 expression and elevated neutrophil infiltration in the vicinity of the hematoma. We also determined the extent to which plasmin from the hematoma influences the platelet-derived growth factor-BB/platelet-derived growth factor receptor-ß system subsequent to intracerebral hemorrhage. DESIGN: Controlled in vivo laboratory study. SETTING: Animal research laboratory. SUBJECTS: One hundred and fifty six eight-week-old male CD1 mice. INTERVENTIONS: Brain injury was induced by autologous arterial blood or plasmin injection into mouse brains. Small interfering RNA targeting platelet-derived growth factor receptor-ß was administered 24 hours before intracerebral hemorrhage. A platelet-derived growth factor receptor antagonist, Gleevec, was administered following intracerebral hemorrhage. A mitogen-activated protein kinase-activated protein kinase 2 inhibitor (KKKALNRQLGVAA) was delivered with platelet-derived growth factor-BB in naïve animals. Platelet-derived growth factor-BB was injected with a plasmin inhibitor (ε-aminocaproic acid) in intracerebral hemorrhage mice. Plasmin-injected mice were given platelet-derived growth factor receptor-ß small interfering RNA 24 hours before the operation. Neurological deficits, brain edema, western blots, and immunofluorescence were evaluated. MEASUREMENTS AND MAIN RESULTS: Platelet-derived growth factor receptor-ß small interfering RNA attenuated SMemb and intercellular adhesion molecule-1 expression and neutrophil infiltration at 24 hours post injury and reduced neurological deficits and brain edema at 24 and 72 hours following intracerebral hemorrhage. The platelet-derived growth factor receptor antagonist, Gleevec, reduced SMemb and intercellular adhesion molecule-1 expression. Platelet-derived growth factor receptor-ß activation led to increased expression of intercellular adhesion molecule-1 and was reversed by KKKALNRQLGVAA in naïve mice. Plasmin inhibition suppressed platelet-derived growth factor receptor-ß activation and neutrophil infiltration, whereas exogenous platelet-derived growth factor-BB increased platelet-derived growth factor receptor-ß activation, regardless of plasmin inhibition. Platelet-derived growth factor receptor-ß small interfering RNA decreased the expression of intercellular adhesion molecule-1 by plasmin injection. CONCLUSION: The platelet-derived growth factor-BB/platelet-derived growth factor receptor-ß system contributes to neuroinflammation through vascular smooth muscle cell phenotypic transformation near the hematoma via the p38 mitogen-activated protein kinase/mitogen-activated protein kinase-activated protein kinase 2 pathway following intracerebral hemorrhage. Plasmin is hypothesized to be upstream of the proposed neuroinflammatory system. The therapeutic intervention targeting the platelet-derived growth factor-BB/platelet-derived growth factor receptor-ß is a novel strategy to prevent plasmin-induced brain injury following intracerebral hemorrhage.


Asunto(s)
Hemorragia Cerebral/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Músculo Liso Vascular/metabolismo , Miosina Tipo IIB no Muscular/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Actinas/metabolismo , Animales , Becaplermina , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Hemorragia Cerebral/complicaciones , Fibrinolisina/antagonistas & inhibidores , Fibrinolisina/farmacología , Fibrinolíticos/farmacología , Mesilato de Imatinib/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Músculo Liso Vascular/citología , Neutrófilos/fisiología , Miosina Tipo IIB no Muscular/genética , Fenotipo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , ARN Interferente Pequeño/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1093-R1104, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27707720

RESUMEN

This study explored the hypothesis that intracerebral hemorrhage (ICH) promotes release of diffusible factors that can significantly influence the structure and function of cerebral arteries remote from the site of injury, through action on platelet-derived growth factor (PDGF) receptors. Four groups of adult male Sprague-Dawley rats were studied (n = 8 each): 1) sham; 2) sham + 60 mg/kg ip imatinib; 3) ICH (collagenase method); and 4) ICH + 60 mg/kg ip imatinib given 60 min after injury. At 24 h after injury, sham artery passive diameters (+3 mM EGTA) averaged 244 ± 7 µm (at 60 mmHg). ICH significantly increased passive diameters up to 6.4% and decreased compliance up to 42.5%. For both pressure- and potassium-induced contractions, ICH decreased calcium mobilization up to 26.2% and increased myofilament calcium sensitivity up to 48.4%. ICH reduced confocal colocalization of smooth muscle α-actin (αActin) with nonmuscle myosin heavy chain (MHC) and increased its colocalization with smooth muscle MHC, suggesting that ICH promoted contractile differentiation. ICH also enhanced colocalization of myosin light chain kinase (MLCK) with both αActin and regulatory 20-kDa myosin light chain. All effects of ICH on passive diameter, compliance, contractility, and contractile protein colocalization were significantly reduced or absent in arteries from animals treated with imatinib. These findings support the hypothesis that ICH promotes release into the cerebrospinal fluid of vasoactive factors that can diffuse to and promote activation of cerebrovascular PDGF receptors, thereby altering the structure, contractile protein organization, contractility, and smooth muscle phenotype of cerebral arteries remote from the site of hemorrhage.


Asunto(s)
Arterias Cerebrales/fisiopatología , Hemorragia Cerebral/fisiopatología , Trastornos Cerebrovasculares/prevención & control , Trastornos Cerebrovasculares/fisiopatología , Mesilato de Imatinib/administración & dosificación , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Arterias Cerebrales/efectos de los fármacos , Hemorragia Cerebral/tratamiento farmacológico , Circulación Cerebrovascular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Fenotipo , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Resultado del Tratamiento
10.
Stroke ; 46(9): 2637-46, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26251254

RESUMEN

BACKGROUND AND PURPOSE: Inflammatory injury plays a critical role in intracerebral hemorrhage (ICH)-induced secondary brain injury. Recently, dopamine D2 receptor (DRD2) is identified as an important component controlling innate immunity and inflammatory response in central nervous system, and αB-crystallin (CRYAB) is a potent negative regulator on inflammatory pathways. Here, we sought to investigate the role of DRD2 on neuroinflammation after experimental ICH and the potential mechanism mediated by CRYAB. METHODS: Two hundred and twenty-four (224) male CD-1 mice were subjected to intrastriatal infusion of bacterial collagenase or autologous blood. Two DRD2 agonists quinpirole and ropinirole were administrated by daily intraperitoneal injection starting at 1 hour after ICH. DRD2 and CRYAB in vivo knockdown was performed 48 hours before ICH insult. Behavioral deficits and brain water content, Western blots, immunofluorescence staining, coimmunoprecipitation (Co-IP) assay, and proteome cytokine array were evaluated. RESULTS: Endogenous DRD2 and CRYAB expressions were increased after ICH. DRD2 knockdown aggravated the neurobehavioral deficits and the pronounced cytokine expressions. DRD2 activation by quinpirole and ropinirole ameliorated neurological outcome, brain edema, interleukin-1ß, and monocyte chemoattractant protein-1 expression, as well as microglia/macrophages activation, in the perihematomal region. These effects were abolished by pretreatment with CRYAB siRNAs. Quinpirole enhanced cytoplasmic binding activity between CRYAB and NF-κB and decreased nuclear NF-κB expression. Similar therapeutic benefits were observed using autologous blood injection model and intranasal delivery of quinpirole. CONCLUSIONS: DRD2 may have anti-inflammatory effects after ICH. DRD2 agonists inhibited neuroinflammation and attenuated brain injury after ICH, which is probably mediated by CRYAB and enhanced cytoplasmic binding activity with NF-κB.


Asunto(s)
Núcleo Celular/metabolismo , Hemorragia Cerebral/metabolismo , Inflamación/metabolismo , FN-kappa B/metabolismo , Receptores de Dopamina D2/metabolismo , Cadena B de alfa-Cristalina/metabolismo , Transporte Activo de Núcleo Celular , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos
11.
Stroke ; 46(2): 485-91, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25523052

RESUMEN

BACKGROUND AND PURPOSE: 17ß-estradiol (E2) has been reported to reduce bleeding and brain injury in experimental intracerebral hemorrhage (ICH) model. However, it is not clear if E2 can prevent early hematoma expansion (HE) induced by hyperglycemia in acute ICH. The aim of this study is to evaluate the effects of E2 on HE and its potential mechanisms in hyperglycemic ICH mice. METHODS: Two hundred, 8-week-old male CD1 mice were used. ICH was performed by collagenase injection. 50% dextrose (8 mL/kg) was injected intraperitoneally 3 hours after ICH to induce acute HE (normal saline was used as control). The time course of HE was measured 6, 24, and 72 hours after ICH. Two dosages (100 and 300 µg/kg) of E2 were administrated 1 hour after ICH intraperitoneally. Neurobehavioral deficits, hemorrhage volume, blood glucose level, and blood-brain barrier disruption were measured. To study the mechanisms of E2, estrogen receptor α (ERα) inhibitor methyl-piperidino-pyrazole, silent information regulator 1 (Sirt1) siRNA was administered, respectively. Protein expression of ERα, Sirt1, and acetylated nuclear factor-kappa B, and activity of matrix metalloproteinases-9 were detected. RESULTS: Hyperglycemia enhanced HE and deteriorated neurological deficits after ICH from 6 hours after ICH. E2 treatment prevented blood-brain barrier disruption and improved neurological deficits 24 and 72 hours after ICH. E2 reduced HE by activating its receptor ERα, decreasing the expression of Sirt1, deacelylation of nuclear factor-kappa B, and inhibiting the activity of matrix metalloproteinases-9. ERα inhibitor methyl-piperidino-pyrazole and Sirt1 siRNA removed these effects of E2. CONCLUSIONS: E2 treatment prevented hyperglycemia-enhanced HE and improved neurological deficits in ICH mice mediated by ERα/Sirt1/nuclear factor-kappa B pathway. E2 may serve as an alternative treatment to decrease early HE after ICH.


Asunto(s)
Hemorragia Cerebral/metabolismo , Estradiol/uso terapéutico , Receptor alfa de Estrógeno/metabolismo , Hematoma/metabolismo , Hiperglucemia/metabolismo , FN-kappa B/metabolismo , Sirtuina 1/metabolismo , Animales , Hemorragia Cerebral/tratamiento farmacológico , Modelos Animales de Enfermedad , Estradiol/farmacología , Receptor alfa de Estrógeno/agonistas , Estrógenos/farmacología , Estrógenos/uso terapéutico , Hematoma/prevención & control , Hiperglucemia/tratamiento farmacológico , Masculino , Ratones
12.
Stroke ; 46(6): 1710-3, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25931468

RESUMEN

BACKGROUND AND PURPOSE: This study examines the role of thrombin's protease-activated receptor (PAR)-1, PAR-4 in mediating cyclooxygenase-2 and mammalian target of rapamycin after germinal matrix hemorrhage. METHODS: Germinal matrix hemorrhage was induced by intraparenchymal infusion of bacterial collagenase into the right ganglionic eminence of P7 rat pups. Animals were treated with PAR-1, PAR-4, cyclooxygenase-2, or mammalian target of rapamycin inhibitors by 1 hour, and ≤5 days. RESULTS: We found increased thrombin activity 6 to 24 hours after germinal matrix hemorrhage, and PAR-1, PAR-4, inhibition normalized cyclooxygenase-2, and mammalian target of rapamycin by 72 hours. Early treatment with NS398 or rapamycin substantially improved long-term outcomes in juvenile animals. CONCLUSIONS: Suppressing early PAR signal transduction, and postnatal NS398 or rapamycin treatment, may help reduce germinal matrix hemorrhage severity in susceptible preterm infants.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Inmunosupresores/farmacología , Nitrobencenos/farmacología , Receptor PAR-1/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Sulfonamidas/farmacología , Animales , Animales Recién Nacidos , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Ciclooxigenasa 2/metabolismo , Ratas
13.
Stroke ; 45(3): 828-34, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24425124

RESUMEN

BACKGROUND AND PURPOSE: Transforming growth factor-ß (TGF-ß) overproduction and activation of the TGF-ß pathway are associated with the development of brain injury following germinal matrix hemorrhage (GMH) in premature infants. We examined the effects of GMH on the level of TGF-ß1 in a novel rat collagenase-induced GMH model and determined the effect of inhibition of the TGF receptor I. METHODS: In total, 92 seven-day old (P7) rats were used. Time-dependent effects of GMH on the level of TGF-ß1 and TGF receptor I were evaluated by Western blot. A TGF receptor I inhibitor (SD208) was administered daily for 3 days, starting either 1 hour or 3 days after GMH induction. The effects of GMH and SD208 on the TGF-ß pathway were evaluated by Western blot at day 3. The effects of GMH and SD208 on cognitive and motor function were also assessed. The effects of TGF receptor I inhibition by SD208 on GMH-induced brain injury and underlying molecular pathways were investigated by Western blot, immunofluorescence, and morphology studies 24 days after GMH. RESULTS: GMH induced significant delay in development, caused impairment in both cognitive and motor functions, and resulted in brain atrophy in rat subjects. GMH also caused deposition of both vitronectin (an extracellular matrix protein) and glial fibrillary acidic protein in perilesion areas, associated with development of hydrocephalus. SD208 ameliorated GMH-induced developmental delay, improved cognitive and motor functions, and attenuated body weight loss. SD208 also decreased vitronectin and glial fibrillary acidic protein deposition and decreased GMH-induced brain injury. CONCLUSIONS: Increased level of TGF-ß1 and activation of the TGF-ß pathway associate with the development of brain injury after GMH. SD208 inhibits GMH-induced activation of the TGF-ß pathway and leads to an improved developmental profile, partial recovery of cognitive and motor functions, and attenuation of GMH-induced brain atrophy and hydrocephalus.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/fisiopatología , Hemorragias Intracraneales/tratamiento farmacológico , Hemorragias Intracraneales/fisiopatología , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/fisiopatología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Adulto , Animales , Atrofia , Western Blotting , Ventrículos Cerebrales/patología , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Hidrocefalia/etiología , Hidrocefalia/patología , Inmunohistoquímica , Enfermedades del Sistema Nervioso/etiología , Examen Neurológico , Embarazo , Pteridinas/farmacología , Pteridinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Sobrevida , Vitronectina/metabolismo , Pérdida de Peso/efectos de los fármacos
14.
Stroke ; 45(12): 3691-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25342030

RESUMEN

BACKGROUND AND PURPOSE: Milk fat globule-EGF factor-8 (MFGE8) has been reported to be neuroprotective in ischemic stroke. However, the effects of MFGE8 in early brain injury after subarachnoid hemorrhage (SAH) have not been investigated. We investigated the role of MFGE8 in early brain injury and the potential mechanisms in antioxidation after SAH. METHODS: Two dosages (1 µg and 3.3 µg) of recombinant human MFGE8 were injected intracerebroventricularly at 1.5 hours after SAH. SAH grades, neurological scores, and brain water content were measured at 24 and 72 hours. For mechanistic study, MFGE8 siRNA, integrin ß3 siRNA, and heme oxygenase (HO) inhibitor SnPP IX were used for intervention. The oxidative stress and expression of MFGE8, integrin ß3, HO-1, extracellular signal-regulated kinase, and nuclear factor erythroid 2-related factor 2 were measured by Western blots 24 hours after SAH. RESULTS: The expression of MFGE8 and HO-1 increased and peaked 24 hours after SAH. Administration of recombinant human MFGE8 decreased brain water content and improved neurological functions both at 24 hours and at 72 hours after SAH. Recombinant human MFGE8 reduced oxidative stress and enhanced the expression of extracellular signal-regulated kinase, nuclear factor erythroid 2-related factor 2, and HO-1; and the effects were abolished by integrin ß3 siRNA and HO inhibitor SnPP IX. CONCLUSIONS: Recombinant MFGE8 attenuated oxidative stress that may be mediated by integrin ß3/nuclear factor erythroid 2-related factor 2/HO pathway after SAH. Recombinant MFGE8 may serve as an alternative treatment to ameliorate early brain injury for SAH patients.


Asunto(s)
Antígenos de Superficie/farmacología , Proteínas de la Leche/farmacología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Hemorragia Subaracnoidea/metabolismo , Animales , Antígenos de Superficie/metabolismo , Western Blotting , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Hemo Oxigenasa (Desciclizante)/metabolismo , Humanos , Integrina beta3/metabolismo , Masculino , Proteínas de la Leche/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología
15.
J Hepatol ; 61(5): 1048-55, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24972044

RESUMEN

BACKGROUND & AIMS: Hepatic ischemia and reperfusion (I/R) injury is a major complication of liver transplantation, hepatic resection and trauma. Helium preconditioning (HPC) exerts protection against ischemic stress. We investigated potential beneficial effects of HPC on I/R-induced liver injury and investigated mechanisms underlying HPC-induced protection. METHODS: We employed a model of segmental warm hepatic I/R on BALB/c mice. Serum ALT was measured and livers were analysed by histology, RT-PCR and western blot. HPC was induced by inhalation of a 70% helium/30% oxygen mixture for three 5-min periods, interspersed with three 5-min washout periods by room air. We tested which component of HPC (the helium/air mixture inhalation, the air room gap, or the interaction between these two factors) is protective. RESULTS: We found that HPC caused a significant increase in Akt phosphorylation in hepatocytes. The HPC-induced Akt phosphorylation resulted in decreased hepatocellular injury and improved survival rate of the treated animals. PI3K inhibitors abolished HPC induced effects. HPC-induced Akt phosphorylation affected expression of its downstream molecules. The effects of HPC on the PI3K/Akt pathway were attenuated by adenosine A2A receptor blockade, but could be re-established by PTEN inhibition. We demonstrated that the interaction of helium/air breathing and air gaps is responsible for the observed effects of HPC. CONCLUSIONS: HPC may be a promising strategy leading to a decrease in I/R induced liver injury in clinical settings. Additionally, the PI3K/Akt pathway plays an essential role in the protective effects of HPC in hepatic I/R injury.


Asunto(s)
Helio/uso terapéutico , Precondicionamiento Isquémico/métodos , Trasplante de Hígado , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/prevención & control , Acondicionamiento Pretrasplante/métodos , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Hígado/efectos de los fármacos , Hígado/lesiones , Hígado/metabolismo , Trasplante de Hígado/efectos adversos , Trasplante de Hígado/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosforilación , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal , Triazinas/farmacología , Triazoles/farmacología , Isquemia Tibia
16.
Exp Biol Med (Maywood) ; 249: 10117, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38590360

RESUMEN

The risk factors and causes of intracerebral hemorrhage (ICH) and the degree of functional recovery after ICH are distinct between young and elderly patients. The increasing incidence of ICH in young adults has become a concern; however, research on the molecules and pathways involved ICH in subjects of different ages is lacking. In this study, tandem mass tag (TMT)-based proteomics was utilized to examine the protein expression profiles of perihematomal tissue from young and aged mice 24 h after collagenase-induced ICH. Among the 5,129 quantified proteins, ICH induced 108 and 143 differentially expressed proteins (DEPs) in young and aged mice, respectively; specifically, there were 54 common DEPs, 54 unique DEPs in young mice and 89 unique DEPs in aged mice. In contrast, aging altered the expression of 58 proteins in the brain, resulting in 39 upregulated DEPs and 19 downregulated DEPs. Bioinformatics analysis indicated that ICH activated different proteins in complement pathways, coagulation cascades, the acute phase response, and the iron homeostasis signaling pathway in mice of both age groups. Protein-protein interaction (PPI) analysis and ingenuity pathway analysis (IPA) demonstrated that the unique DEPs in the young and aged mice were related to lipid metabolism and carbohydrate metabolism, respectively. Deeper paired-comparison analysis demonstrated that apolipoprotein M exhibited the most significant change in expression as a result of both aging and ICH. These results help illustrate age-related protein expression changes in the acute phase of ICH.


Asunto(s)
Hemorragia Cerebral , Proteómica , Anciano , Humanos , Ratones , Animales , Proteómica/métodos , Hemorragia Cerebral/metabolismo , Encéfalo/metabolismo , Envejecimiento , Proteínas/metabolismo
17.
Stroke ; 44(5): 1410-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23539525

RESUMEN

BACKGROUND AND PURPOSE: Plasma thrombin concentration is increased after subarachnoid hemorrhage (SAH). However, the role of thrombin receptor (protease-activated receptor-1 [PAR-1]) in endothelial barrier disruption has not been studied. The aims of this study were to investigate the role of PAR-1 in orchestrating vascular permeability and to assess the potential therapeutics of a PAR-1 antagonist, SCH79797, through maintaining vascular integrity. METHODS: SCH79797 was injected intraperitoneally into male Sprauge-Dawley rats undergoing SAH by endovascular perforation. Assessment was conducted at 24 hours after SAH for brain water content, Evans blue content, and neurobehavioral testing. To explore the role of PAR-1 activation and the specific mechanism of SCH79797's effect after SAH, Western blot, immunoprecipitation, and immunofluorescence of hippocampus tissue were performed. A p21-activated kinase-1 (PAK1) inhibitor, IPA-3, was used to explore the underlying protective mechanism of SCH79797. RESULTS: At 24 hours after SAH, animals treated with SCH79797 demonstrated a reduction in brain water content, Evans blue content, and neurobehavioral deficits. SCH79797 also attenuated PAR-1 expression and maintained the level of vascular endothelial-cadherin, an important component of adherens junctions. Downstream to PAR-1, c-Src-dependent activation of p21-activated kinase-1 led to an increased serine/threonine phosphorylation of vascular endothelial-cadherin; immunoprecipitation results revealed an enhanced binding of phosphorylated vascular endothelial-cadherin with endocytosis orchestrator ß-arrestin-2. These pathological states were suppressed after SCH79797 treatment. CONCLUSIONS: PAR-1 activation after SAH increases microvascular permeability, at least, partly through a PAR-1-c-Src-p21-activated kinase-1-vascular endothelial-cadherin phosphorylation pathway. Through suppressing PAR-1 activity, SCH79797 plays a protective role in maintaining microvascular integrity after SAH.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Pirroles/uso terapéutico , Quinazolinas/uso terapéutico , Receptor PAR-1/antagonistas & inhibidores , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Disulfuros/farmacología , Inhibidores Enzimáticos/farmacología , Masculino , Naftoles/farmacología , Permeabilidad/efectos de los fármacos , Pirroles/farmacología , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/patología , Quinasas p21 Activadas/antagonistas & inhibidores
18.
Neurobiol Dis ; 50: 13-20, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23000356

RESUMEN

Neurosurgical procedures inevitably produce intraoperative hemorrhage. The subsequent entry of blood into the brain parenchyma results in the release of large amounts of thrombin, a known contributor to perihematomal edema formation and apoptosis after brain injury. The present study seeks to test 1) the effect of surgically induced brain injury (SBI) on thrombin activity, expression of thrombin's receptor PAR-1, and PAR-1 mediated apoptosis; 2) the effect of thrombin inhibition by argatroban and PAR-1 inhibition by SCH79797 on the development of secondary brain injury in the SBI model on rats. A total of 88 Sprague-Dawley male rats were randomly divided into sham, vehicle-, argatroban-, or SCH79797-treated groups. SBI involved partial resection of the right frontal lobe under inhalation isoflurane anesthesia. Sham-operated animals received only craniotomy. Thrombin activity, brain water content, and neurological deficits were measured at 24 h following SBI. Involvement of the Ask1/JNK pathway in PAR-1-induced post-SBI apoptosis was characterized by using Ask1 or JNK inhibitors. We observed that SBI increased thrombin activity, yet failed to demonstrate any effect on PAR-1 expression. Argatroban and SCH79797 reduced SBI-induced brain edema and neurological deficits in a dose-dependent manner. SBI-induced apoptosis seemed mediated by the PAR-1/Ask1/JNK pathways. Administration of SCH79797 ameliorated the apoptosis following SBI. Our findings indicate that PAR-1 antagonist protects against secondary brain injury after SBI by decreasing both brain edema and apoptosis by inactivating PAR-1/Ask1/JNK pathway. The anti-apoptotic effect of PAR-1 antagonists may provide a promising path for therapy following SBI.


Asunto(s)
Apoptosis/efectos de los fármacos , Lesiones Encefálicas/metabolismo , Complicaciones Intraoperatorias/metabolismo , Procedimientos Neuroquirúrgicos/efectos adversos , Pirroles/farmacología , Quinazolinas/farmacología , Receptor PAR-1/antagonistas & inhibidores , Animales , Apoptosis/fisiología , Western Blotting , Encéfalo/cirugía , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Inmunohistoquímica , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Masculino , Ratas , Ratas Sprague-Dawley , Trombina/metabolismo
19.
Crit Care Med ; 41(5): 1266-75, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23388512

RESUMEN

OBJECTIVE: Hydrogen inhalation was neuroprotective in several brain injury models. Its mechanisms are believed to be related to antioxidative stress. We investigated the potential neurovascular protective effect of hydrogen inhalation especially effect on mast cell activation in a mouse model of intracerebral hemorrhage. DESIGN: Controlled in vivo laboratory study. SETTING: Animal research laboratory. SUBJECTS: One hundred seventy-one 8-week-old male CD-1 mice were used. INTERVENTIONS: Collagenase-induced intracerebral hemorrhage model in 8-week-old male CD-1 mice was used. Hydrogen was administrated via spontaneous inhalation. The blood-brain barrier permeability and neurologic deficits were investigated at 24 and 72 hours after intracerebral hemorrhage. Mast cell activation was evaluated by Western blot and immuno-staining. The effects of hydrogen inhalation on mast cell activation were confirmed in an autologous blood injection model intracerebral hemorrhage. MEASUREMENT AND MAIN RESULTS: At 24 and 72 hours post intracerebral hemorrhage, animals showed blood-brain barrier disruption, brain edema, and neurologic deficits, accompanied with phosphorylation of Lyn kinase and release of tryptase, indicating mast cell activation. Hydrogen treatment diminished phosphorylation of Lyn kinase and release of tryptase, decreased accumulation and degranulation of mast cells, attenuated blood-brain barrier disruption, and improved neurobehavioral function. CONCLUSION: Activation of mast cells following intracerebral hemorrhage contributed to increase of blood-brain barrier permeability and brain edema. Hydrogen inhalation preserved blood-brain barrier disruption by prevention of mast cell activation after intracerebral hemorrhage.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Lesiones Encefálicas/prevención & control , Hemorragia Cerebral/tratamiento farmacológico , Hidrógeno/administración & dosificación , Mastocitos/efectos de los fármacos , Administración por Inhalación , Análisis de Varianza , Animales , Western Blotting , Edema Encefálico/etiología , Edema Encefálico/patología , Edema Encefálico/prevención & control , Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Movimiento Celular/efectos de los fármacos , Hemorragia Cerebral/complicaciones , Colagenasas/farmacología , Modelos Animales de Enfermedad , Hidrógeno/farmacocinética , Peróxido de Hidrógeno/metabolismo , Inmunohistoquímica , Masculino , Mastocitos/fisiología , Ratones , Ratones Endogámicos , Distribución Aleatoria , Valores de Referencia , Tasa de Supervivencia
20.
Exp Neurol ; 368: 114507, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37598880

RESUMEN

Despite decades of intensive research, there are still very limited options for the effective treatment of intracerebral hemorrhage (ICH). Recently, mounting evidence has indicated that the ultra-early stage (<3 h), serving as the primary phase of ICH, plays a pivotal role and may even surpass other stages in terms of its significance. Therefore, uncovering the metabolic alterations induced by ICH in the ultra-early stage is of crucial importance. To investigate this, the collagenase ICH mouse model was employed in this study. ICH or sham-operated mice were euthanized at the ultra-early stage of 3 h and the acute stage of 24 h and 72 h after the operation. Then, the metabolic changes in the perihematomal tissues were detected by liquid chromatography coupled with tandem mass spectrometry. In total, alterations in the levels of 465 metabolites were detected. A total of 136 metabolites were significantly changed at 3 h. At 24 h and 72 h, the amounts were 132 and 126, respectively. Additionally, the key corresponding metabolic pathways for these time points were analyzed through KEGG. To gather additional information, quantitative real-time transcription polymerase chain reaction, enzyme-linked immunosorbent assay and Western blots were performed to validate the metabolic changes. Overall, ICH significantly alters important physiological functions such as cysteine metabolism, purine metabolism, synaptic alterations, the synaptic vesicle cycle, and the ATP-binding cassette transporter system. These might be the key pathologic mechanisms of the ultra-early stage induced by ICH.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Metabolómica , Animales , Ratones , Hemorragia Cerebral , Cromatografía Liquida , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA