Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 589
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35101976

RESUMEN

Blood-brain barrier (BBB) breakdown and inflammation occurring at the BBB have a key, mainly a deleterious role in the pathophysiology of ischemic stroke. Neddylation is a ubiquitylation-like pathway that is critical in various cellular functions by conjugating neuronal precursor cell-expressed developmentally down-regulated protein 8 (NEDD8) to target proteins. However, the roles of neddylation pathway in ischemic stroke remain elusive. Here, we report that NEDD8 conjugation increased during acute phase after ischemic stroke and was present in intravascular and intraparenchymal neutrophils. Inhibition of neddylation by MLN4924, also known as pevonedistat, inactivated cullin-RING E3 ligase (CRL), and reduced brain infarction and improved functional outcomes. MLN4924 treatment induced the accumulation of the CRL substrate neurofibromatosis 1 (NF1). By using virus-mediated NF1 silencing, we show that NF1 knockdown abolished MLN4924-dependent inhibition of neutrophil trafficking. These effects were mediated through activation of endothelial P-selectin and intercellular adhesion molecule-1 (ICAM-1), and blocking antibodies against P-selectin or anti-ICAM-1 antibodies reversed NF1 silencing-induced increase in neutrophil infiltration in MLN4924-treated mice. Furthermore, we found that NF1 silencing blocked MLN4924-afforded BBB protection and neuroprotection through activation of protein kinase C δ (PKCδ), myristoylated alanine-rich C-kinase substrate (MARCKS), and myosin light chain (MLC) in cerebral microvessels after ischemic stroke, and treatment of mice with the PKCδ inhibitor rottlerin reduced this increased BBB permeability. Our study demonstrated that increased neddylation promoted neutrophil trafficking and thus exacerbated injury of the BBB and stroke outcomes. We suggest that the neddylation inhibition may be beneficial in ischemic stroke.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Ciclopentanos/farmacología , Proteína NEDD8/metabolismo , Proteínas del Tejido Nervioso , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirimidinas/farmacología , Ubiquitina-Proteína Ligasas , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/enzimología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/enzimología , Masculino , Ratones , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo
2.
Int J Mol Sci ; 22(23)2021 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-34884729

RESUMEN

The human brain requires a high rate of oxygen consumption to perform intense metabolic activities, accounting for 20% of total body oxygen consumption. This high oxygen uptake results in the generation of free radicals, including reactive oxygen species (ROS), which, at physiological levels, are beneficial to the proper functioning of fundamental cellular processes. At supraphysiological levels, however, ROS and associated lesions cause detrimental effects in brain cells, commonly observed in several neurodegenerative disorders. In this review, we focus on the impact of oxidative DNA base lesions and the role of DNA glycosylase enzymes repairing these lesions on brain function and disease. Furthermore, we discuss the role of DNA base oxidation as an epigenetic mechanism involved in brain diseases, as well as potential roles of DNA glycosylases in different epigenetic contexts. We provide a detailed overview of the impact of DNA glycosylases on brain metabolism, cognition, inflammation, tissue loss and regeneration, and age-related neurodegenerative diseases based on evidence collected from animal and human models lacking these enzymes, as well as post-mortem studies on patients with neurological disorders.


Asunto(s)
ADN Glicosilasas/metabolismo , Reparación del ADN , Enfermedades Neurodegenerativas/enzimología , Estrés Oxidativo , Animales , Encéfalo/fisiología , Lesiones Encefálicas/enzimología , Epigénesis Genética , Humanos , Regeneración
3.
Dis Markers ; 2021: 3682034, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790277

RESUMEN

OBJECTIVE: This study is aimed at exploring the possible neuroprotective mechanism of aspirin and the effect of bacterial endotoxin lipopolysaccharide (LPS) during cerebral ischaemia-reperfusion (CIRP) injury. METHODS: We established three animal models: the CIRP, LPS, and CIRP+LPS models. Mortality, the injured brain area, and the beam walking test were used to estimate the degree of cerebral injury among the rats. Immunohistochemistry and immunofluorescence were used to detect activated microglia, matrix metalloproteinase-3 (MMP-3), and osteopontin (OPN). RESULTS: The injured brain area and mortality were dramatically reduced (p < 0.01), and the beam walking test scores were elevated (p < 0.01) in the acetylsalicylic acid (ASA) group compared to the control group. The number of microglia-, MMP-3-, and OPN-positive cells also increased. Furthermore, the number of GSI-B4, OPN, and MMP-3 cells decreased in the ASA group compared to the control group. After LPS stimulation, the number of microglia reached a peak at 24 h; at 7 d, these cells disappeared. In the ASA group, the number of microglia was significantly smaller (p < 0.05), especially at 24 h (p < 0.01), compared to the LPS group. Moreover, the injured brain area and the mortality were dramatically increased and the beam walking test scores were reduced (p < 0.01) after LPS simulation following CIRP. The degree of injury in the ASA group resembled that in the control group. However, the number of MMP-3-immunoreactive neurons or microglia was significantly larger than that of the control group (p < 0.05). In the ASA group, the MMP-3 expression was also considerably decreased (p < 0.05). CONCLUSIONS: After CIRP, microglia were rapidly activated and the expression of MMP-3 and OPN significantly increased. For rats injected with LPS at reperfusion, the injured brain area and mortality also dramatically increased and the neurologic impairment worsened. However, ASA exhibited a neuroprotective effect during CIRP injury. Furthermore, ASA can reverse LPS-induced cerebral injury and inhibit the inflammatory reaction after CIRP injury.


Asunto(s)
Aspirina/farmacología , Lesiones Encefálicas/prevención & control , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/toxicidad , Metaloproteinasa 3 de la Matriz/química , Fármacos Neuroprotectores/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley
4.
Biochem Pharmacol ; 194: 114796, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34678224

RESUMEN

Extracellular Vesicles (EVs) are implicated in the spread of pathogenic proteinsin a growing number of neurological diseases. Given this, there is rising interest in developing inhibitors of Neutral Sphingomyelinase 2 (nSMase2), an enzyme critical in EV biogenesis. Our group recently discovered phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)carbamate (PDDC), the first potent, selective, orally-available, and brain-penetrable nSMase2 inhibitor, capable of dose-dependently reducing EVs release in vitro and in vivo. Herein, using multiplexed Surface Plasmon Resonance imaging (SPRi), we evaluated which brain cell-derived EVs were affected by PDDC following acute brain injury. Mice were fed PDDC-containing chow at doses which gave steady PDDC brain exposures exceeding its nSMase2 IC50. Mice were then administered an intra-striatal IL-1ß injection and two hours later plasma and brain were collected. IL-1ß injection significantly increased striatal nSMase2 activity which was completely normalized by PDDC. Using SPRi, we found that IL-1ß-induced injury selectively increased plasma levels of CD171 + and PLP1 + EVs; this EV increase was normalized by PDDC. In contrast, GLAST1 + EVs were unchanged by IL-1ß or PDDC. IL-1ß injection selectively increased EVs released from activated versus non-activated microglia, indicated by the CD11b+/IB4 + ratio. The increase in EVs from CD11b + microglia was dramatically attenuated with PDDC. Taken together, our data demonstrate that following acute injury, brain nSMase2 activity is elevated. EVs released from neurons, oligodendrocytes, and activated microglial are increased in plasma and inhibition of nSMase2 with PDDC reduced these IL-1ß-induced changes implicating nSMase2 inhibition as a therapeutic target for acute brain injury.


Asunto(s)
Lesiones Encefálicas/enzimología , Vesículas Extracelulares/enzimología , Microglía/enzimología , Neuronas/enzimología , Oligodendroglía/enzimología , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Lesiones Encefálicas/tratamiento farmacológico , Carnitina/administración & dosificación , Carnitina/análogos & derivados , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/enzimología , Vesículas Extracelulares/efectos de los fármacos , Inyecciones Intraventriculares , Interleucina-1beta/administración & dosificación , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Pirenos/administración & dosificación , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores
5.
J Biochem Mol Toxicol ; 35(9): e22855, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34309959

RESUMEN

Valproic acid (VPA) is an efficient antiepileptic drug widely used for the treatment of epilepsy and other seizures in both children and adults. It is also reported to have side and toxic effects on many organs and tissues. Vitamin B6 (Vit B6 ) is a well-described water-soluble vitamin, which has an antioxidant effect. In this study, we aimed to investigate the protective effect of Vit B6 on VPA-induced brain injury. Male Sprague-Dawley rats were divided into four groups. Group I, control animals; Group II, Vit B6 (50 mg/kg/day) given rats; Group III, VPA (500 mg/kg/day) given rats; Group IV, VPA and Vit B6 given rats at same dose and time. VPA and Vit B6 were administered intraperitoneally and orally, respectively, for 7 days. At the end of the experiments, the rats were sacrificed and brain tissues were taken. Protein carbonyl and sialic acid levels, xanthine oxidase, adenosine deaminase, acetylcholine esterase, lactate dehydrogenase, myeloperoxidase activities, total oxidant status, and reactive oxygen species levels were found to be increased, while glutathione and total antioxidant capacity levels, catalase, superoxide dismutase, glutathione-S-transferase, paraoxonase, and glutathione reductase activities were found to be decreased in the VPA group. Administration of Vit B6 reversed these defects in the VPA group. These findings indicate that Vit B6 has a protective effect on VPA-induced brain damage.


Asunto(s)
Lesiones Encefálicas , Síndromes de Neurotoxicidad , Ácido Valproico/efectos adversos , Vitamina B 6/farmacología , Animales , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/enzimología , Masculino , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/enzimología , Oxidorreductasas/metabolismo , Ratas , Ratas Sprague-Dawley , Ácido Valproico/farmacología
6.
Neurochem Res ; 45(10): 2245-2257, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32671628

RESUMEN

Inflammation secondary to tissue injuries serves as a double-edged sword that determines the prognosis of tissue repair. As one of the most important enzymes controlling the inflammation process by producing leukotrienes, 5-lipoxygenase (5-LOX, also called 5-LO) has been one of the therapeutic targets in regulating inflammation for a long time. Although a large number of 5-LOX inhibitors have been explored, only a few of them can be applied clinically. Surprisingly, phosphorylation of 5-LOX reveals great significance in regulating the subcellular localization of 5-LOX, which has proven to be an important mechanism underlying the enzymatic activities of 5-LOX. There are at least three phosphorylation sites in 5-LOX jointly to determine the final inflammatory outcomes, and adjustment of phosphorylation of 5-LOX at different phosphorylation sites brings hope to provide an unrecognized means to regulate inflammation. The present review intends to shed more lights into the set-point-like mechanisms of phosphorylation of 5-LOX and its possible clinical application by summarizing the biological properties of 5-LOX, the relationship of 5-LOX with neurodegenerative diseases and brain injuries, the phosphorylation of 5-LOX at different sites, the regulatory effects and mechanisms of phosphorylated 5-LOX upon inflammation, as well as the potential anti-inflammatory application through balancing the phosphorylation-depended set-point.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Lesiones Encefálicas/metabolismo , Inflamación/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Animales , Araquidonato 5-Lipooxigenasa/química , Encéfalo/enzimología , Encéfalo/metabolismo , Lesiones Encefálicas/enzimología , Humanos , Inflamación/enzimología , Inhibidores de la Lipooxigenasa/farmacología , Enfermedades Neurodegenerativas/enzimología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Serina/química
7.
Stroke ; 51(3): 975-985, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32078472

RESUMEN

Background and Purpose- Microglia/macrophages (Mi/MΦ) can profoundly influence stroke outcomes by acquiring functionally dominant phenotypes (proinflammatory or anti-inflammatory; deleterious or salutary). Identification of the molecular mechanisms that dictate the functional status of Mi/MΦ after brain ischemia/reperfusion may reveal novel therapeutic targets for stroke. We hypothesized that activation of TAK1 (transforming growth factor beta-activated kinase 1), a key MAP3K upstream of multiple inflammation-regulating pathways, drives Mi/MΦ toward a proinflammatory phenotype and potentiates ischemia/reperfusion brain injury. Methods- Young adult mice were subjected to 1 hour of middle cerebral artery occlusion (MCAO) followed by reperfusion. TAK1 was targeted by tamoxifen-induced Mi/MΦ-specific knockout or administration of a selective inhibitor 5Z-7-Oxozeaenol after MCAO. Neurobehavioral deficits and long-term gray matter and white matter injury were assessed up to 35 days after MCAO. Mi/MΦ functional status and brain inflammatory profiles were assessed 3 days after MCAO by RNA-seq, flow cytometry, and immunohistochemistry. Results- TAK1 Mi/MΦ-specific knockout markedly ameliorated neurological deficits in the rotarod and cylinder tests for at least 35 days after MCAO. Mechanistically, RNA-seq of purified brain Mi/MΦ demonstrated that proinflammatory genes and their predicted biological functions were downregulated or inhibited in microglia and macrophages from TAK1 Mi/MΦ-specific knockout mice versus WT mice 3 days after MCAO. Consistent with the anti-inflammatory phenotype of Mi/MΦ-specific knockout, oxozeaenol treatment mitigated neuroinflammation 3 days after MCAO, manifested by less Iba1+/CD16+ proinflammatory Mi/MΦ and suppressed brain invasion of various peripheral immune cells. Oxozeaenol treatment beginning 2 hours after MCAO improved long-term sensorimotor and cognitive functions in the foot fault, rotarod, and water maze tests. Furthermore, Oxozeaenol promoted both gray matter and white matter integrity 35 days after MCAO. Conclusions- TAK1 promotes ischemia/reperfusion-induced inflammation, brain injury, and maladaptive behavior by enhancing proinflammatory and deleterious Mi/MΦ responses. Therefore, TAK1 inhibition is a promising therapy to improve long-term stroke outcomes.


Asunto(s)
Conducta Animal , Lesiones Encefálicas/enzimología , Isquemia Encefálica/enzimología , Quinasas Quinasa Quinasa PAM/metabolismo , Daño por Reperfusión/enzimología , Accidente Cerebrovascular/enzimología , Animales , Lesiones Encefálicas/genética , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Quinasas Quinasa Quinasa PAM/genética , Macrófagos , Ratones , Ratones Noqueados , Microglía , Daño por Reperfusión/genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Factores de Tiempo , Zearalenona/análogos & derivados , Zearalenona/farmacología
8.
Front Neuroendocrinol ; 56: 100816, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31786088

RESUMEN

Aromatase is the requisite and limiting enzyme in the production of estrogens from androgens. Estrogens synthesized centrally have more recently emerged as potent neuroprotectants in the vertebrate brain. Studies in rodents and songbirds have identified key mechanisms that underlie both; the injury-dependent induction of central aromatization, and the protective effects of centrally synthesized estrogens. Injury-induced aromatase expression in astrocytes occurs following a broad range of traumatic brain damage including excitotoxic, penetrating, and concussive injury. Responses to neural insult such as edema and inflammation involve signaling pathways the components of which are excellent candidates as inducers of this astrocytic response. Finally, estradiol from astrocytes exerts a paracrine neuroprotective influence via the potent inhibition of inflammatory pathways. Taken together, these data suggest a novel role for neural aromatization as a protective mechanism against the threat of inflammation and suggests that central estrogen provision is a wide-ranging neuroprotectant in the vertebrate brain.


Asunto(s)
Aromatasa/metabolismo , Lesiones Encefálicas/enzimología , Encéfalo/enzimología , Animales , Aromatasa/genética , Astrocitos/enzimología , Edema Encefálico/enzimología , Estradiol/fisiología , Femenino , Humanos , Inflamación/enzimología , Masculino , Degeneración Nerviosa/enzimología , Neuroprotección/fisiología , Pájaros Cantores/fisiología , Regulación hacia Arriba
9.
J Lipid Res ; 60(3): 609-623, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30662008

RESUMEN

Traumatic brain injury (TBI) is one of the leading causes of disability worldwide and a prominent risk factor for neurodegenerative diseases. The expansion of nervous tissue damage after the initial trauma involves a multifactorial cascade of events, including excitotoxicity, oxidative stress, inflammation, and deregulation of sphingolipid metabolism that further mitochondrial dysfunction and secondary brain damage. Here, we show that a posttranscriptional activation of an acid sphingomyelinase (ASM), a key enzyme of the sphingolipid recycling pathway, resulted in a selective increase of sphingosine in mitochondria during the first week post-TBI that was accompanied by reduced activity of mitochondrial cytochrome oxidase and activation of the Nod-like receptor protein 3 inflammasome. TBI-induced mitochondrial abnormalities were rescued in the brains of ASM KO mice, which demonstrated improved behavioral deficit recovery compared with WT mice. Furthermore, an elevated autophagy in an ASM-deficient brain at the baseline and during the development of secondary brain injury seems to foster the preservation of mitochondria and brain function after TBI. Of note, ASM deficiency attenuated the early stages of reactive astrogliosis progression in an injured brain. These findings highlight the crucial role of ASM in governing mitochondrial dysfunction and brain-function impairment, emphasizing the importance of sphingolipids in the neuroinflammatory response to TBI.


Asunto(s)
Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Técnicas de Inactivación de Genes , Mitocondrias/patología , Recuperación de la Función , Esfingomielina Fosfodiesterasa/deficiencia , Animales , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/genética , Cognición , Activación Enzimática , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Esfingomielina Fosfodiesterasa/genética
10.
Stroke ; 50(2): 520-523, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30602353

RESUMEN

Background and Purpose- Subarachnoid hemorrhage (SAH) is a devastating form of stroke. Oxidative stress contributes to brain injury, but the mechanisms have been poorly studied. Here, we evaluated the role of 12/15-lipoxygenase (12/15-LOX), an enzyme known to cause cell death in ischemic stroke, on brain injury in a mouse model of SAH. Methods- C57Bl6 wild-type mice and Alox15 knockout mice were subjected to SAH using a direct blood injection technique. In SAH wild-type mice, half received the 12/15-LOX inhibitor ML351 and half received vehicle. Immunohistochemistry, brain edema, blood-brain barrier leakage and functional outcomes were assessed 1 and 3 days after SAH induction. Results- SAH led to increased 12/15-LOX in macrophages of the brain parenchyma, adjacent to the subarachnoid blood. Neuronal cell death after SAH was reduced by ML351 and in Alox15 knockout mice. Similarly, SAH induced brain edema, which was 12/15-LOX dependent. Finally, Alox15 gene knockout and inhibitor treatment in wild-type mice with SAH led to an improved behavioral outcome. Conclusions- 12/15-LOX is overexpressed in macrophages after SAH in mice, and inhibition of the 12/15-LOX pathway decreases brain injury and improves neurological outcome. This study suggests 12/15-LOX as a novel therapeutic target to limit brain injury after SAH.


Asunto(s)
Araquidonato 12-Lipooxigenasa , Araquidonato 15-Lipooxigenasa , Lesiones Encefálicas , Isoxazoles/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Macrófagos , Naftalenos/farmacología , Estrés Oxidativo , Hemorragia Subaracnoidea , Animales , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Macrófagos/enzimología , Macrófagos/patología , Ratones , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/enzimología , Hemorragia Subaracnoidea/genética , Hemorragia Subaracnoidea/patología
11.
Brain Res ; 1701: 112-125, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30205109

RESUMEN

Oxidative stress plays an important role in secondary brain injury (SBI) after intracerebral hemorrhage (ICH), but the underling mechanism has not been fully elucidated. Recently, the antioxidant enzyme glutathione peroxidase 4 (GPX4), has attracted increasing attention due to its ability to degrade reactive oxygen species (ROS) which are the major indicator of oxidative stress; However, the role of GPX4 in ICH has not been reported. This study was designed to investigate the changes in protein levels, as well as potential role and mechanism of GPX4 in SBI following ICH using a Sprague-Dawley (SD) rat model of ICH induced by autologous blood injection into the right basal ganglia. Firstly, GPX4 protein levels in the brain were reduced gradually and bottomed out at 24 h after ICH, compared with the Sham group. Secondly, genetic-overexpression of GPX4 effectively increased level of GPX4 in the brain, and clearly relieved neuronal dysfunction, brain edema, blood brain barrier (BBB) injury, oxidative stress and inflammation after ICH. In contrast, inhibiting GPX4 with a specific pharmacological inhibitor or genetic knockdown exacerbated SBI after ICH. Finally, Ferrostatin-1, a chemical inhibitor of ferroptosis, was used to explore the role of ferroptosis in brain injury after ICH. The results suggest that inhibiting ferroptosis can significantly alleviate SBI after ICH. In summary, our work indicated that GPX4 contributes to SBI following ICH by mediating ferroptosis. Therefore, inhibiting ferroptosis with specific inhibitors or upregulation of GPX4 may be a potential strategy to ameliorate brain injury induced by ICH.


Asunto(s)
Lesiones Encefálicas/enzimología , Hemorragia Cerebral/enzimología , Glutatión Peroxidasa/metabolismo , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Muerte Celular/fisiología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Ciclohexilaminas/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Fenilendiaminas/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
12.
J Thromb Haemost ; 16(7): 1413-1422, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29729651

RESUMEN

Essentials von Willebrand Factor (VWF) and ADAMTS13 may affect early injury after subarachnoid hemorrhage (SAH). Early brain injury was assessed in VWF-/- , ADAMTS13-/- and recombinant (r) ADAMTS13 treated mice. VWF-/- and rADAMTS13 treated mice had less brain injury than ADAMTS13-/- and wild-type mice. Early administration of rADAMTS13 may improve outcome after SAH by reducing early brain injury. SUMMARY: Background Early brain injury is an important determinant of poor functional outcome and case fatality after aneurysmal subarachnoid hemorrhage (SAH), and is associated with early platelet aggregation. No treatment exists for early brain injury after SAH. We investigated whether von Willebrand factor (VWF) is involved in the pathogenesis of early brain injury, and whether ultra-early treatment with recombinant ADAMTS-13 (rADAMTS-13) reduces early brain injury after experimental SAH. Methods Experimental SAH in mice was induced by prechiasmatic injection of non-anticoagulated blood from a littermate. The following experimental SAH groups were investigated: C57BL/6J control (n = 21), VWF-/- (n = 25), ADAMTS-13-/- (n = 23), and C57BL/6J treated with rADAMTS-13 (n = 26). Mice were killed at 2 h after SAH. Primary outcome measures were microglial activation (IBA-1 surface area) and neuronal injury (number of cleaved caspase-3-positive neurons). Results As compared with controls, microglial activation was decreased in VWF-/- mice (mean difference of - 20.0%, 95% confidence interval [CI] - 4.0% to - 38.6%), increased in ADAMTS-13-/- mice (mean difference of + 34.0%, 95% CI 16.2-51.7%), and decreased in rADAMTS-13-treated mice (mean difference of - 22.1%, 95% CI - 3.4% to - 39.1%). As compared with controls (185 neurons, interquartile range [IQR] 133-353), neuronal injury in the cerebral cortex was decreased in VWF-/- mice (63 neurons, IQR 25-78), not changed in ADAMTS-13-/- mice (53 neurons, IQR 26-221), and reduced in rADAMTS-13-treated mice (45 neurons, IQR 9-115). Conclusions Our findings suggest that VWF is involved in the pathogenesis of early brain injury, and support the further study of rADAMTS-13 as a treatment option for early brain injury after SAH.


Asunto(s)
Proteína ADAMTS13/metabolismo , Lesiones Encefálicas/etiología , Encéfalo/enzimología , Hemorragia Subaracnoidea/complicaciones , Factor de von Willebrand/metabolismo , Proteína ADAMTS13/administración & dosificación , Proteína ADAMTS13/deficiencia , Proteína ADAMTS13/genética , Animales , Apoptosis , Encéfalo/efectos de los fármacos , Encéfalo/patología , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/genética , Lesiones Encefálicas/prevención & control , Proteínas de Unión al Calcio/metabolismo , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Esquema de Medicación , Femenino , Predisposición Genética a la Enfermedad , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/enzimología , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fenotipo , Proteínas Recombinantes/administración & dosificación , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/enzimología , Hemorragia Subaracnoidea/genética , Factores de Tiempo , Factor de von Willebrand/genética
13.
Mol Med Rep ; 17(5): 6647-6654, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29512790

RESUMEN

N-acetyl cysteine (NAC) has been extensively reported to exert neuroprotective effects on the central nervous system. Oxidative stress may contribute to the underlying mechanisms causing Alzheimer's disease (AD). The effect of NAC against oxidative stress injury was investigated in a cellular model of AD in the present study and the underlying mechanisms were revealed. The neuroprotective action of NAC (1, 10, 100 and 1,000 µmol/l) on a cellular model of AD [hydrogen peroxide (H2O2)­induced (3, 30 and 300 µmol/l) toxicity in primary rat hippocampus neurons] demonstrated the underlying mechanisms. Cytotoxicity was measured using the MTT assay, and light microscopy and the dichloro-dihydro-fluorescein diacetate method were used to detect the reactive oxygen species (ROS) levels. Furthermore, the levels of mitogen-activated protein kinases (MAPKs) signal transduction and tau protein phosphorylation were measured via western blotting. NAC (100 µmol/l) protected hippocampus neurons against H2O2­mediated toxicity, as evidenced by enhanced cell viability. Using MTT assay and light microscopy for the observation of cell death, NAC ameliorated cell viability, which was induced by H2O2 injury (P<0.05). NAC was found to mitigate the excessive production of ROS (P<0.05). Another mechanism involved in the neuroprotective action of NAC may be its ability to inhibit MAPK signal transduction following H2O2 exposure. In addition, NAC may protect cells against H2O2­induced toxicity by attenuating increased tau phosphorylation. Thus, the protective ability of NAC is hypothesized to result from inhibition of oxidative stress and downregulation of MAPK signal transduction and tau phosphorylation.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/lesiones , Peróxido de Hidrógeno/efectos adversos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuronas/enzimología , Fármacos Neuroprotectores/farmacología , Animales , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/patología , Hipocampo/enzimología , Hipocampo/patología , Peróxido de Hidrógeno/farmacología , Neuronas/patología , Ratas , Ratas Sprague-Dawley
14.
Shock ; 49(6): 704-711, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28846566

RESUMEN

No pharmacological interventions are currently available to provide neuroprotection for patients suffering from cardiac arrest. Dichloroacetate (DCA) is a pyruvate dehydrogenase kinase inhibitor, which activates pyruvate dehydrogenase (PDH), and increases cell adenosine triphosphate (ATP) production by promoting influx of pyruvate into the Krebs cycle. In this study, we investigated the effects of DCA on post-resuscitation neurological injury in an asphyxial cardiac arrest rat model. Asphyxial cardiac arrest was established by endotracheal tube clamping. A total of 111 rats were randomized into three groups: Sham group, Control group, and DCA intervention group. Animals in DCA intervention group were intraperitoneally administered DCA with a loading dose of 80 mg/kg at 15 min after return of spontaneous circulation (ROSC), whereas rats in the Control group received equivalent volume of saline. DCA treatment increased 3-day survival time, and reduced neurologic deficit scores at 24, 48, and 72 h after ROSC. It also attenuated cellular apoptosis and neuronal damage in the hippocampal cornuammonis one region by hematoxylin-eosin staining and TdT-mediated dUTP nick-end labeling assay. In addition, DCA reduced the messenger RNA expression of tumor necrosis factor α and interleukin 1ß in brain hippocampus and cortex after ROSC. Furthermore, DCA treatment significantly increased ATP production, PDH activity, and decreased blood glucose, lactate, and brain pyruvate levels after ROSC. Our results suggested that DCA has neuroprotective effects on brain injury after cardiac arrest, and its salutary effects were associated with an increase of mitochondrial energy metabolism in the brain through activation of PDH activity.


Asunto(s)
Lesiones Encefálicas , Ácido Dicloroacético/farmacología , Paro Cardíaco , Fármacos Neuroprotectores/farmacología , Complejo Piruvato Deshidrogenasa/metabolismo , Animales , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/patología , Lesiones Encefálicas/prevención & control , Metabolismo Energético/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Paro Cardíaco/tratamiento farmacológico , Paro Cardíaco/enzimología , Paro Cardíaco/patología , Masculino , Oxidación-Reducción/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Resucitación
15.
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
16.
Biosci Trends ; 11(4): 439-449, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-28824025

RESUMEN

The purposes of the current study were to investigate the effects of a monoclonal antibody (mAb) on cytosolic phospholipase A2 alpha (cPLA2α) in mice with cerebral ischemia-reperfusion (IR) injury and to ascertain the potential mechanisms of those effects. This study evaluated whether the use of anti-cPLA2α mAb could reduce stroke injury in a mouse model of cerebral IR injury. The expression/activity of cPLA2α and cPLA2α- derived proinflammatory lipid mediators such as prostaglandin E2 (PGE2), leukotriene B4 (LTB4), lysophosphatidylcholine (LPC), and free fatty acids (FFA) was assessed. This study also evaluated neurological deficits, motor function, pathological changes, apoptosis, and the area of infarction in the injured mice. Mice treated with anti-cPLA2α mAb recovered neurological function and their condition improved, apoptosis in the brain decreased and infarct volume decreased, and expression of cPLA2α, 5-LOX, COX-2, FFA, LPC, PGE2, and LTB4 was attenuated. Our findings indicate that cPLA2α plays a key role in cerebral IR injury and that treatment with anti-cPLA2α mAb after cerebral IR injury helps to reduce levels of proinflammatory cytokines, alleviate tissue damage, and reduce levels of deleterious lipid mediators. Thus, anti-cPLA2α mAb treatment has the potential to treat ischemic brain damage.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/enzimología , Fosfolipasas A2 Grupo IV/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Encéfalo/patología , Encéfalo/cirugía , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Fosfolipasas A2 Grupo IV/metabolismo , Masculino , Ratones , Daño por Reperfusión/complicaciones , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/fisiopatología
17.
Biomed Res Int ; 2017: 2924848, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28698869

RESUMEN

BACKGROUND: Hypoxia ischemia (HI) to the developing brain occurs in 1-6 in 1000 live births. Large numbers of survivors have neurological long-term sequelae. However, mechanisms of recovery after HI are not understood and preventive measures or clinical treatments are not effective. Poly(ADP-ribose) polymerase-1 is overactivated in response to ischemia. In neonatal mice HI activates PARP-1 but its role in perinatal brain injury remains uncertain. OBJECTIVE: Aim of this study was to explore the effect of TES448 (PARP-1-inhibitor) and hypothermia after an ischemic insult. DESIGN AND METHODS: 10-day-old Wistar rats underwent HI. TES448 was given 10 min, 3 hrs, and 6 hrs after hypoxia. Hypothermia was started 30 min after HI and brains were dissected at P12. Western blotting and histological staining were used to evaluate for degree of injury. RESULTS: Protein expression of PARP-1 levels was diminished after TES448 treatment. Cresyl violet and TUNEL staining revealed decreased injury in male rat pups following TES448 and combined treatment. Female rats showed increased numbers of TUNEL-positive cells after combined therapy. TES448 inhibited microglia activation after hypoxic-ischemic injury. A cellular response including NeuN, Olig2, and MBP was not affected by PARP-1-inhibition. CONCLUSIONS: Inhibition of PARP-1 and hypothermia lead to an alteration of injury but this effect is sexually dimorphic.


Asunto(s)
Lesiones Encefálicas/enzimología , Isquemia Encefálica/enzimología , Encéfalo/enzimología , Regulación Enzimológica de la Expresión Génica , Poli(ADP-Ribosa) Polimerasa-1/biosíntesis , Animales , Encéfalo/patología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Masculino , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Ratas , Ratas Wistar
18.
Endocrinology ; 158(8): 2585-2592, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575175

RESUMEN

Injury to the vertebrate brain causes neuroinflammation, characterized in part by increases in prostaglandins. In rodents and songbirds, brain injury also induces the transcription and translation of aromatase in reactive astrocytes around the site of damage. Interestingly, this induction is more rapid in female zebra finches relative to males. Induced aromatization is neuroprotective, as inhibition of aromatase and estrogen replacement, increases and decreases the extent of damage, respectively. Although the consequences of induced astrocytic aromatization are intensely studied, little is known about what factors induce aromatase. Inflammation is sufficient to induce astrocytic aromatase suggesting that the link between inflammation and aromatase expression may be causal. To test this hypothesis, adult male and female zebra finches received bilateral mechanical injuries through which either the cyclooxygenase (COX)-1/2 inhibitor indomethacin or vehicle was administered into contralateral hemispheres. Subjects were killed either 6 or 24 hours after injury. In both sexes, an enzyme immunoassay for prostaglandin E2 (PGE2) revealed that indomethacin decreased PGE2 relative to the contralateral hemisphere at both time points, suggesting that the dose and mode of administration used were successful in affecting neuroinflammation locally. Indomethacin reduced aromatase expression and 17ß-estradiol (E2) content at 6 hours but not 24 hours following injury in females. However, in males, the inhibitory effect of indomethacin on aromatase and E2 was apparent at 24 but not 6 hours after treatment. These data suggest that COX activity, perhaps via consequent prostaglandin secretion, may induce aromatase expression and central E2, an effect that is detectable in temporally distinct patterns between sexes.


Asunto(s)
Aromatasa/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Encéfalo/metabolismo , Estradiol/metabolismo , Pinzones , Indometacina/farmacología , Animales , Antiinflamatorios no Esteroideos , Encéfalo/patología , Lesiones Encefálicas/enzimología , Dinoprostona/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Técnicas para Inmunoenzimas , Indometacina/administración & dosificación , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
19.
Harefuah ; 156(5): 280-284, 2017 May.
Artículo en Hebreo | MEDLINE | ID: mdl-28551908

RESUMEN

INTRODUCTION: Traumatic brain injuries (TBI) are a major cause of mortality and disability among young adults. TBI are characterized by primary injury, the result of a mechanical impact to the cranium and a secondary injury, a series of molecular mechanism processes developing thereafter. Cerebral cells modify their gene and protein expression as a result of the injury. Epigenetic modifications have a key role as regulators of gene transcription and may simultaneously be involved in the regulation of the molecular pathways following TBI. However, the mechanisms are unknown. OBJECTIVES: To clarify whether modification in the expression of Histone Acetyl Transferase1 (HAT1) and Histone deacetylase2 (HDAC 2) occurs during secondary brain damage. METHODS: Rat diffused head injury model was used; 72 hours post injury animals were sacrificed and the brains were removed for immunohistochemistry staining with Caspase 3, HAT1 and HDAC2 antibodies. We compared these stains in the perilesional versus the contralateral cortex. RESULTS: An increase of Caspase 3 stained cells were observed in the perilesional cortex. HAT1 expression was elevated and HDAC2 expression reduced in the injured cortex. CONCLUSIONS: TBI induced modifications in the expression of epigenetic factors were concomitant with increases in apoptotic cell death. The mitochondria involved in the apoptotic processes is a target for epigenetic regulation and also influences it at the same time. DISCUSSION: This study contributes to the understanding of epigenetic modification following TBI. Further study on the relationship between mitochondrial activity and epigenetic regulation has to be performed in order to develop novel drugs and therapies for TBI.


Asunto(s)
Muerte Encefálica , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/genética , Muerte Celular/genética , Epigénesis Genética , Regulación de la Expresión Génica/fisiología , Animales , Corteza Cerebral , Modelos Animales de Enfermedad , Humanos , Ratas
20.
Mol Med Rep ; 15(5): 2727-2731, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28447745

RESUMEN

The present study aimed to investigate the association between myocardial cell apoptosis and calpain-1/caspase-3 expression in a rat model of hypoxic-ischemic brain damage (HIBD). A total of 64 newborn rats were divided into control (n=8; sacrificed on day 7) and HIBD groups (n=56). HIBD group rats were sacrificed 2, 12 or 24 h, or 2, 3, 5 or 7 days following HIBD (n=8/group). A terminal deoxynucleotidyl transferase dUTP nick-end labeling assay was performed to detect myocardial apoptotic cells and calculate the apoptosis index (AI), reverse transcription-polymerase chain reaction was performed to detect myocardial calpain-1/caspase-3 mRNA expression levels and a western blot analysis was conducted to detect calpain­1 protein expression levels. The correlations between calpain­1 and caspase­3 expression levels and AI were analyzed. The results demonstrated that apoptotic myocardial cells in the HIBD groups were markedly increased compared with the control group, with AI peaking in the day 3 group. Caspase­3 and calpain­1 mRNA expression levels were increased from 2 and 12 h following HIBD, respectively, with the most elevated levels in the day 2 group. Compared with the control group, calpain­1 protein expression levels were increased from 2 h, with the greatest expression levels in the day 3 group (P<0.05). Calpain­1 mRNA and protein (76/80 kDa) expression levels demonstrated positive linear correlations with AI (r=0.786, P=0.001; and r=0.853, P=0.001, respectively) Caspase-3 mRNA expression levels were positively correlated with AI (r=0.894; P=0.001). In conclusion, the present study demonstrated that in rats with HIBD, there is a positive correlation between increased apoptosis of myocardial cells and expression levels of calpain-1 and caspase-3.


Asunto(s)
Apoptosis , Lesiones Encefálicas/enzimología , Calpaína/biosíntesis , Caspasa 3/biosíntesis , Regulación Enzimológica de la Expresión Génica , Hipoxia Encefálica/enzimología , Miocardio/enzimología , Animales , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Femenino , Hipoxia Encefálica/patología , Masculino , Miocardio/patología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...