Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Neuroinflammation ; 18(1): 26, 2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33468172

RESUMEN

BACKGROUND: Microglia-mediated neuroinflammation plays a crucial role in the pathogenesis of hypoxic-ischemic (HI)-induced brain injury. Activation of melanocortin-1 receptor (MC1R) has been shown to exert anti-inflammatory and neuroprotective effects in several neurological diseases. In the present study, we have explored the role of MC1R activation on neuroinflammation and the potential underlying mechanisms after neonatal hypoxic-ischemic brain injury in rats. METHODS: A total of 169 post-natal day 10 unsexed rat pups were used. HI was induced by right common carotid artery ligation followed by 2.5 h of hypoxia. BMS-470539, a specific selective MC1R agonist, was administered intranasally at 1 h after HI induction. To elucidate the potential underlying mechanism, MC1R CRISPR KO plasmid or Nurr1 CRISPR KO plasmid was administered via intracerebroventricular injection at 48 h before HI induction. Percent brain infarct area, short- and long-term neurobehavioral tests, Nissl staining, immunofluorescence staining, and Western blot were conducted. RESULTS: The expression levels of MC1R and Nurr1 increased over time post-HI. MC1R and Nurr1 were expressed on microglia at 48 h post-HI. Activation of MC1R with BMS-470539 significantly reduced the percent infarct area, brain atrophy, and inflammation, and improved short- and long-term neurological deficits at 48 h and 28 days post-HI. MC1R activation increased the expression of CD206 (a microglial M2 marker) and reduced the expression of MPO. Moreover, activation of MC1R with BMS-470539 significantly increased the expression levels of MC1R, cAMP, p-PKA, and Nurr1, while downregulating the expression of pro-inflammatory cytokines (TNFα, IL-6, and IL-1ß) at 48 h post-HI. However, knockout of MC1R or Nurr1 by specific CRISPR reversed the neuroprotective effects of MC1R activation post-HI. CONCLUSIONS: Our study demonstrated that activation of MC1R with BMS-470539 attenuated neuroinflammation, and improved neurological deficits after neonatal hypoxic-ischemic brain injury in rats. Such anti-inflammatory and neuroprotective effects were mediated, at least in part, via the cAMP/PKA/Nurr1 signaling pathway. Therefore, MC1R activation might be a promising therapeutic target for infants with hypoxic-ischemic encephalopathy (HIE).


Asunto(s)
Encéfalo/efectos de los fármacos , Hipoxia-Isquemia Encefálica/metabolismo , Imidazoles/farmacología , Receptor de Melanocortina Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Ratas , Ratas Sprague-Dawley
2.
J Cell Mol Med ; 24(21): 12318-12330, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33034416

RESUMEN

This study aimed to investigate the effects of PPAR-ß/δ receptor agonist GW0742 on neuroinflammation in a rat model of hypoxia-ischaemia (HI) and in PC12 cells in OGD model. HI was induced by ligating the common carotid artery and inducing hypoxia for 150 minutes. Immunofluorescence was used for quantification of microglia activation and for determining cellular localization of PPAR-ß/δ. Expression of proteins was measured by Western blot. Activation of miR-17-5p by GW0742 was assessed in PC12 cells by Dual-Luciferase Reporter Gene Assay. The endogenous expression of TXNIP, NLRP3, cleaved caspase-1 and IL-1ß was increased after HI. GW0742 treatment significantly reduced the number of activated pro-inflammatory microglia in ipsilateral hemisphere after HI. Mechanistically, GW0742 significantly decreased the expression of TXNIP, NLRP3, IL-6 and TNF-α. Either PPAR-ß/δ antagonist GSK3787, miR-17-5p inhibitor, or TXNIP CRISPR activation abolished the anti-inflammatory effects of GW0742. Activation of PPAR-ß/δ by GW0742 activated miR-17-5p expression in PC12 cells and increased cell viability after OGD, which was accompanied by decreased expression of TXNIP and reduced secretion of IL-1ß and TNF-α. In conclusion, GW0742 may be a promising neurotherapeutic for the management of HI patients.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Hipoxia/metabolismo , Inflamación/metabolismo , Isquemia/metabolismo , MicroARNs/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Tiazoles/farmacología , Animales , Glucemia/metabolismo , Supervivencia Celular , Femenino , Glucosa/metabolismo , Inflamasomas , Masculino , Microscopía Fluorescente , Oxígeno/metabolismo , Células PC12 , Ratas , Ratas Sprague-Dawley
3.
Stroke ; 51(11): 3320-3331, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32933418

RESUMEN

BACKGROUND AND PURPOSE: Neuroinflammation has been proven to play an important role in the pathogenesis of early brain injury after subarachnoid hemorrhage (SAH). EZH2 (enhancer of zeste homolog 2)-mediated H3K27Me3 (trimethylation of histone 3 lysine 27) has been recognized to play a critical role in multiple inflammatory diseases. However, there is still a lack of evidence to address the effect of EZH2 on the immune response of SAH. Therefore, the aim of this study was to determine the role of EZH2 in SAH-induced neuroinflammation and explore the effect of EZH2 inhibition with its specific inhibitor EPZ6438. METHODS: The endovascular perforation method was performed on rats to induce subarachnoid hemorrhage. EPZ6438, a specific EZH2 inhibitor, was administered intraperitoneally at 1 hour after SAH. SOCS3 (Suppressor of cytokine signaling 3) siRNA and H3K27me3 CRISPR were administered intracerebroventricularly at 48 hours before SAH to explore potential mechanisms. The SAH grade, short-term and long-term neurobehavioral tests, immunofluorescence staining, and western blots were performed after SAH. RESULTS: The expression of EZH2 and H3K27me3 peaked at 24 hours after SAH. In addition, inhibition of EZH2 with EPZ6438 significantly improved neurological deficits both in short-term and long-term outcome studies. Moreover, EPZ6438 treatment significantly decreased the levels of EZH2, H3K27Me3, pathway-related proteins TRAF6 (TNF [tumor necrosis factor] receptor family 6), NF-κB (nuclear factor-κB) p65, proinflammatory cytokines TNF-α, IL (interleukin)-6, IL-1ß, but increased the expression levels of SOCS3 and anti-inflammatory cytokine IL-10. Furthermore, administration of SOCS3 siRNA and H3k27me3-activating CRISPR partly abolished the neuroprotective effect of EPZ6438, which indicated that the neuroprotective effect of EPZ6438 acted, at least partly, through activation of SOCS3. CONCLUSIONS: In summary, the inhibition of EZH2 by EPZ6438 attenuated neuroinflammation via H3K27me3/SOCS3/TRAF6/NF-κB signaling pathway after SAH in rats. By targeting EZH2, this study may provide an innovative method to ameliorate early brain injury after SAH.


Asunto(s)
Encéfalo/inmunología , Proteína Potenciadora del Homólogo Zeste 2/inmunología , Histonas/metabolismo , Inflamación/inmunología , FN-kappa B/inmunología , Hemorragia Subaracnoidea/inmunología , Proteína 3 Supresora de la Señalización de Citocinas/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología , Animales , Benzamidas/farmacología , Compuestos de Bifenilo , Encéfalo/efectos de los fármacos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Código de Histonas , Histonas/efectos de los fármacos , Masculino , Microglía/efectos de los fármacos , Microglía/inmunología , Morfolinas , Prueba del Laberinto Acuático de Morris , FN-kappa B/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Piridonas/farmacología , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante , Transducción de Señal , Hemorragia Subaracnoidea/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/efectos de los fármacos , Factor 6 Asociado a Receptor de TNF/efectos de los fármacos
4.
J Neuroinflammation ; 17(1): 152, 2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375838

RESUMEN

BACKGROUND: Inhibition of inositol-requiring enzyme-1 alpha (IRE1α), one of the sensor signaling proteins associated with endoplasmic reticulum (ER) stress, has been shown to alleviate brain injury and improve neurological behavior in a neonatal hypoxic-ischemic encephalopathy (HIE) rat model. However, there is no information about the role of IRE1α inhibitor as well as its molecular mechanisms in preventing neuronal pyroptosis induced by NLRP1 (NOD-, LRR- and pyrin domain-containing 1) inflammasome. In the present study, we hypothesized that IRE1α can degrade microRNA-125-b-2-3p (miR-125-b-2-3p) and activate NLRP1/caspased-1 pathway, and subsequently promote neuronal pyroptosis in HIE rat model. METHODS: Ten-day old unsexed rat pups were subjected to hypoxia-ischemia (HI) injury, and the inhibitor of IRE1α, STF083010, was administered intranasally at 1 h after HI induction. AntimiR-125 or NLRP1 activation CRISPR was administered by intracerebroventricular (i.c.v) injection at 24 h before HI induction. Immunofluorescence staining, western blot analysis, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), brain infarct volume measurement, neurological function tests, and Fluoro-Jade C staining were performed. RESULTS: Endogenous phosphorylated IRE1α (p-IRE1α), NLRP1, cleaved caspase-1, interleukin-1ß (IL-1ß), and interleukin-18 (IL-18) were increased and miR-125-b-2-3p was decreased in HIE rat model. STF083010 administration significantly upregulated the expression of miR-125-b-2-3p, reduced the infarct volume, improved neurobehavioral outcomes and downregulated the protein expression of NLRP1, cleaved caspase-1, IL-1ß and IL-18. The protective effects of STF083010 were reversed by antimiR-125 or NLRP1 activation CRISPR. CONCLUSIONS: IRE1α inhibitor, STF083010, reduced neuronal pyroptosis at least in part via miR-125/NLRP1/caspase-1 signaling pathway after HI.


Asunto(s)
Endorribonucleasas/antagonistas & inhibidores , Hipoxia-Isquemia Encefálica/patología , MicroARNs/metabolismo , Complejos Multienzimáticos/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica/metabolismo , Inflamasomas/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Piroptosis/efectos de los fármacos , Piroptosis/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sulfonamidas/farmacología , Tiofenos/farmacología
5.
J Neuroinflammation ; 17(1): 144, 2020 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366312

RESUMEN

BACKGROUND: Cardiac arrest survivors suffer from neurological dysfunction including cognitive impairment. Cerebral mast cells, the key regulators of neuroinflammation contribute to neuroinflammation-associated cognitive dysfunction. Mast cell tryptase was demonstrated to have a proinflammatory effect on microglia via the activation of microglial protease-activated receptor-2 (PAR-2). This study investigated the potential anti-neuroinflammatory effect of mast cell tryptase inhibition and the underlying mechanism of PAR-2/p-p38/NFκB signaling following asphyxia-induced cardiac arrest in rats. METHODS: Adult male Sprague-Dawley rats resuscitated from 10 min of asphyxia-induced cardiac arrest were randomized to four separate experiments including time-course, short-term outcomes, long-term outcomes and mechanism studies. The effect of mast cell tryptase inhibition on asphyxial cardiac arrest outcomes was examined after intranasal administration of selective mast cell tryptase inhibitor (APC366; 50 µg/rat or 150 µg/rat). AC55541 (selective PAR-2 activator; 30 µg/rat) and SB203580 (selective p38 inhibitor; 300 µg/rat) were used for intervention. Short-term neurocognitive functions were evaluated using the neurological deficit score, number of seizures, adhesive tape removal test, and T-maze test, while long-term cognitive functions were evaluated using the Morris water maze test. Hippocampal neuronal degeneration was evaluated by Fluoro-Jade C staining. RESULTS: Mast cell tryptase and PAR-2 were dramatically increased in the brain following asphyxia-induced cardiac arrest. The inhibition of mast cell tryptase by APC366 improved both short- and long-term neurological outcomes in resuscitated rats. Such behavioral benefits were associated with reduced expressions of PAR-2, p-p38, NFκB, TNF-α, and IL-6 in the brain as well as less hippocampal neuronal degeneration. The anti-neuroinflammatory effect of APC366 was abolished by AC55541, which when used alone, indeed further exacerbated neuroinflammation, hippocampal neuronal degeneration, and neurologic deficits following cardiac arrest. The deleterious effects aggregated by AC55541 were minimized by p38 inhibitor. CONCLUSIONS: The inhibition of mast cell tryptase attenuated neuroinflammation, led to less hippocampal neuronal death and improved neurological deficits following cardiac arrest. This effect was at least partly mediated via inhibiting the PAR-2/p-p38/NFκB signaling pathway. Thus, mast cell tryptase might be a novel therapeutic target in the management of neurological impairment following cardiac arrest.


Asunto(s)
Encéfalo/patología , Paro Cardíaco/complicaciones , Hipoxia-Isquemia Encefálica/etiología , Inflamación/metabolismo , Transducción de Señal/fisiología , Triptasas/antagonistas & inhibidores , Animales , Asfixia/complicaciones , Encéfalo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Inflamación/etiología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor PAR-2/metabolismo
6.
J Neurosci Res ; 98(11): 2275-2289, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32772463

RESUMEN

Role of peroxisome proliferator-activated receptors (PPARs) in the pathophysiology of stroke and protective effects of PPAR ligands have been widely investigated in the last 20 years. Activation of all three PPAR isoforms, but especially PPAR-γ, was documented to limit postischemic injury in the numerous in vivo, as well as in in vitro studies. PPARs have been demonstrated to act on multiple mechanisms and were shown to activate multiple protective pathways related to inflammation, apoptosis, BBB protection, neurogenesis, and oxidative stress. The aim of this review was to summarize two decades of PPAR research in stroke with emphasis on in vivo animal studies. We focus on each PPAR receptor separately and detail their implication in stroke. This review also discusses recent clinical efforts in the field and the epidemiological data with regard to role of PPAR polymorphisms in susceptibility to stroke, and tries to draw conclusions and describe future perspectives.


Asunto(s)
Receptores Activados del Proliferador del Peroxisoma/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/prevención & control , Animales , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/fisiopatología , Accidente Cerebrovascular Isquémico/prevención & control , Receptores Activados del Proliferador del Peroxisoma/genética , Accidente Cerebrovascular/fisiopatología
7.
Neurochem Res ; 39(1): 68-75, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24185491

RESUMEN

Perinatal brain insult mostly resulting from hypoxia-ischemia (H-I) often brings lifelong permanent disability, which has a major impact on the life of individuals and their families. The lack of progress in clinically-applicable neuroprotective strategies for birth asphyxia has led to an increasing interest in alternative methods of therapy, including induction of brain tolerance by pre- and particularly postconditioning. Hypoxic postconditioning represents a promising strategy for preventing ischemic brain damage. The aim of this study was to investigate the potential neuroprotective effect of hypobaric hypoxia (HH) postconditioning applied to 7-day old rats after H-I insult. The mild hypobaric conditions (0.47 atm) used in this study imitate an altitude of 5,000 m. We show that application of mild hypobaric hypoxia at relatively short time intervals (1-6 h) after H-I, repeated for two following days leads to significant neuroprotection, manifested by a reduction in weight loss of the ipsilateral hemisphere observed 14 days after H-I. HH postconditioning results in decrease in reactive oxygen species level observed in all experimental groups. The increase in superoxide dismutase activity observed after H-I is additionally enhanced by HH postconditioning applied 1 h after H-I. The increase observed 3 and 6 h after H-I was not statistically significant. Postconditioning with HH suppresses the glutathione concentration decrease evoked by H-I and increased glutathione peroxidase activity and this effect is not dependent on the time of postconditioning initiation. HH postconditioning had no effect on catalase activity. We show for the first time that HH postconditioning reduces brain damage resulting from H-I in immature rats and that the mechanism potentially involved in this effect is related to antioxidant defense mechanisms of immature brain.


Asunto(s)
Asfixia Neonatal/terapia , Hipoxia-Isquemia Encefálica/fisiopatología , Poscondicionamiento Isquémico , Oxígeno/administración & dosificación , Animales , Catalasa/metabolismo , Glutatión Peroxidasa/metabolismo , Hipoxia-Isquemia Encefálica/terapia , Poscondicionamiento Isquémico/métodos , Modelos Animales , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo , Ratas , Superóxido Dismutasa/metabolismo
8.
Exp Neurol ; 371: 114607, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37935323

RESUMEN

Delayed recanalization at days or weeks beyond the therapeutic window was shown to improve functional outcomes in acute ischemic stroke (AIS) patients. However, the underlying mechanisms remain unclear. Previous preclinical study reported that trefoil factor 3 (TFF3) was secreted by liver after cerebral ischemia and acted a distant neuroprotective factor. Here, we investigated the liver-derived TFF3-mediated neuroprotective mechanism enhanced by delayed recanalization after AIS. A total of 327 male Sprague-Dawley rats and the model of middle cerebral artery occlusion (MCAO) with permanent occlusion (pMCAO) or with delayed recanalization at 3 d post-occlusion (rMCAO) were used. Partial hepatectomy was performed within 5 min after MCAO. Leucine-rich repeat and immunoglobulin-like domain-containing nogo receptor-interacting protein 2 (LINGO2) siRNA was administered intracerebroventricularly at 48 h after MCAO. Recombinant rat TFF3 (rr-TFF3, 30 µg/Kg) or recombinant rat epidermal growth factor (rr-EGF, 100 µg/Kg) was administered intranasally at 1 h after recanalization, and EGFR inhibitor Gefitinib (75 mg/Kg) was administered intranasally at 30 min before recanalization. The evaluation of outcomes included neurobehavior, ELISA, western blot and immunofluorescence staining. TFF3 in hepatocytes and serum were upregulated in a similar time-dependent manner after MCAO. Compared to pMCAO, delayed recanalization increased brain TFF3 levels and attenuated brain damage with the reduction in neuronal apoptosis, infarct volume and neurological deficits. Partial hepatectomy reduced TFF3 levels in serum and ipsilateral brain hemisphere, and abolished the benefits of delayed recanalization on neuronal apoptosis and neurobehavioral deficits in rMCAO rats. Intranasal rrTFF3 treatment reversed the changes associated with partial hepatectomy. Delayed recanalization after MCAO increased the co-immunoprecipitation of TFF3 and LINGO2, as well as expressions of p-EGFR, p-Src and Bcl-2 in the brain. LINGO2 siRNA knockdown or EGFR inhibitor reversed the effects of delayed recanalization on apoptosis and brain expressions of LINGO2, p-EGFR, p-Src and Bcl-2 in rMCAO rats. EGFR activator abolished the deleterious effects of LINGO2 siRNA. In conclusion, our investigation demonstrated for the first time that delayed recanalization may enhance the entry of liver-derived TFF3 into ischemic brain upon restoring blood flow after MCAO, which attenuated neuronal apoptosis and neurological deficits at least in part via activating LINGO2/EGFR/Src pathway.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Fármacos Neuroprotectores , Humanos , Ratas , Masculino , Animales , Ratas Sprague-Dawley , Neuroprotección , Infarto de la Arteria Cerebral Media/metabolismo , Factor Trefoil-3/farmacología , Factor Trefoil-3/uso terapéutico , Transducción de Señal , Apoptosis , Receptores ErbB/metabolismo , Receptores ErbB/farmacología , Receptores ErbB/uso terapéutico , Hígado , ARN Interferente Pequeño/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
9.
Transl Stroke Res ; 14(2): 250-262, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35867328

RESUMEN

Current approved therapies for acute ischemic stroke have a restricted therapeutic time window. Delayed recanalization, which has been utilized clinically in patients who have missed the time window for administration, may be a promising alternative for stroke patients. However, the underlying molecular mechanisms remain undiscovered. Herein, we hypothesized that delayed recanalization would increase M2 microglial polarization through the IL-4R (interleukin-4 receptor)/STAT6 (signal transducer and activators of transcription 6)/PPARγ (peroxisome proliferator-activated receptor γ) pathway, subsequently promoting stroke recovery in rats. The permanent middle cerebral artery occlusion (pMCAO) model was induced via intravascular filament insertion. Recanalization was induced by withdrawing the filament at 3 days after MCAO (rMCAO). Interleukin (IL)-4 was administered intranasally at 3 days after pMCAO. AS1517499, a specific STAT6 inhibitor, was administered intranasally at 3 days after MCAO induction. Immunofluorescence staining, enzyme-linked immunosorbent assay (ELISA), western blot analysis, volumetric measurements of brain infarct, and neurological behavior tests were conducted. Delayed recanalization at 3 days after MCAO increased the polarization of M2 microglia, decreased inflammation, and improved neurological behavior. IL-4 treatment administered on the 3rd day after pMCAO increased M2 microglial polarization, improved neurological behavior, and reduced infarction volume of pMCAO rats. The inhibition of STAT6 decreased the level of p-STAT6 and PPARγ in rats treated with delayed recanalization. Delayed recanalization improved neurological function by increasing microglial M2 polarization, possibly involved with the IL-4R/STAT6/PPARγ pathway after MCAO in rats.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratas , Animales , Infarto de la Arteria Cerebral Media/metabolismo , Microglía/metabolismo , PPAR gamma , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico
10.
Oxid Med Cell Longev ; 2022: 4054938, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35140838

RESUMEN

Neuronal apoptosis induced by oxidative stress plays an important role in the pathogenesis and progression of hypoxic-ischemic encephalopathy (HIE). Previous studies reported that activation of melanocortin-1 receptor (MC1R) exerts antioxidative stress, antiapoptotic, and neuroprotective effects in various neurological diseases. However, whether MC1R activation can attenuate oxidative stress and neuronal apoptosis after hypoxic-ischemic- (HI-) induced brain injury remains unknown. Herein, we have investigated the role of MC1R activation with BMS-470539 in attenuating oxidative stress and neuronal apoptosis induced by HI and the underlying mechanisms. 159 ten-day-old unsexed Sprague-Dawley rat pups were used. HI was induced by right common carotid artery ligation followed by 2.5 h of hypoxia. The novel-selective MC1R agonist BMS-470539 was administered intranasally at 1 h after HI induction. MC1R CRISPR KO plasmid and Nurr1 CRISPR KO plasmid were administered intracerebroventricularly at 48 h before HI induction. Percent brain infarct area, short-term neurobehavioral tests, Western blot, immunofluorescence staining, Fluoro-Jade C staining, and MitoSox Staining were performed. We found that the expression of MC1R and Nurr1 increased, peaking at 48 h post-HI. MC1R and Nurr1 were expressed on neurons at 48 h post-HI. BMS-470539 administration significantly attenuated short-term neurological deficits and infarct area, accompanied by a reduction in cleaved caspase-3-positive neurons at 48 h post-HI. Moreover, BMS-470539 administration significantly upregulated the expression of MC1R, cAMP, p-PKA, Nurr1, HO-1, and Bcl-2. However, it downregulated the expression of 4-HNE and Bax, as well as reduced FJC-positive cells, MitoSox-positive cells, and 8-OHdG-positive cells at 48 h post-HI. MC1R CRISPR and Nurr1 CRISPR abolished the antioxidative stress, antiapoptotic, and neuroprotective effects of BMS-470539. In conclusion, our findings demonstrated that BMS-470539 administration attenuated oxidative stress and neuronal apoptosis and improved neurological deficits in a neonatal HI rat model, partially via the MC1R/cAMP/PKA/Nurr1 signaling pathway. Early administration of BMS-470539 may be a novel therapeutic strategy for infants with HIE.


Asunto(s)
Antioxidantes/administración & dosificación , Apoptosis/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/metabolismo , Imidazoles/administración & dosificación , Neuronas/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Receptor de Melanocortina Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Administración Intranasal , Animales , Animales Recién Nacidos , Femenino , Técnicas de Inactivación de Genes/métodos , Masculino , Neuronas/efectos de los fármacos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/genética , Transducción de Señal/genética , Resultado del Tratamiento
11.
Transl Stroke Res ; 12(3): 382-393, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33215347

RESUMEN

Stroke has become the second most prevalent cause of mortality in the world. Currently, the treatment of ischemic stroke is based on thrombolytic and thrombectomy therapy shortly after the ischemic event (≤ 4.5 h for thrombolytic strategies; ≤ 6 h for thrombectomy strategies). However, the majority of patients are unable to receive prompt treatment, particularly in undeveloped countries. Alternative solutions are lacking for those patients that miss the optimal window of opportunity for treatment. Recently, new developments in imaging techniques and intravascular interventional devices enable the expansion of the window of opportunity for treating stroke patients. Clinical studies have reported that delayed recanalization at 24 h, or even more than 1 month, was beneficial for some patients. However, the mechanisms of neuroprotection that underly the delayed recanalization in these ischemic stroke patients remain unclear. In this review, we will summarize the clinical studies of delayed recanalization, and organize them according to the duration of occlusion. Additionally, we will discuss the changing guidelines and possible mechanisms based on animal research, and attempt to draw conclusions and future perspectives.


Asunto(s)
Isquemia Encefálica , Procedimientos Endovasculares , Accidente Cerebrovascular , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/terapia , Fibrinolíticos/uso terapéutico , Humanos , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/terapia , Trombectomía , Terapia Trombolítica , Resultado del Tratamiento
12.
Curr Neuropharmacol ; 18(12): 1237-1249, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32469699

RESUMEN

It is well-known that stroke is one of the leading causes of death and disability all over the world. After a stroke, the blood-brain barrier subsequently breaks down. The BBB consists of endothelial cells surrounded by astrocytes. Microglia, considered the long-living resident immune cells of the brain, play a vital role in BBB function. M1 microglia worsen BBB disruption, while M2 microglia assist in repairing BBB damage. Microglia can also directly interact with endothelial cells and affect BBB permeability. In this review, we are going to discuss the mechanisms responsible for the dual role of microglia in BBB dysfunction after stroke.


Asunto(s)
Barrera Hematoencefálica , Accidente Cerebrovascular , Astrocitos , Células Endoteliales , Humanos , Microglía
13.
Exp Neurol ; 326: 113203, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31954682

RESUMEN

INTRODUCTION: Disruption of the blood brain barrier (BBB) and subsequent cerebral edema formation is one of the major adverse effects of brain surgery, leading to postoperative neurological dysfunction. Recently, Mfsd2a has been shown to have a crucial role for the maintenance of BBB functions. In this study, we aimed to evaluate the role of Mfsd2a on BBB disruption following surgical brain injury (SBI) in rats. MATERIALS AND METHODS: Rats were subjected to SBI by partial resection of the right frontal lobe. To evaluate the effect of Mfsd2a on BBB permeability and neurobehavior outcome following SBI, Mfsd2a was either overexpressed or downregulated in the brain by administering Mfsd2a CRISPR activation or knockout plasmids, respectively. The potential mechanism of Mfsd2a-mediated BBB protection through the cav-1/Nrf-2/HO-1 signaling pathway was evaluated. RESULTS: Mfsd2a levels were significantly decreased while cav-1, Nrf-2 and HO-1 levels were increased in the right frontal perisurgical area following SBI. When overexpressed, Mfsd2a attenuated brain edema and abolished neurologic impairment caused by SBI while downregulation of Mfsd2a expression further deteriorated BBB functions and worsened neurologic performance following SBI. The beneficial effect of Mfsd2a overexpression on BBB functions was associated with diminished expression of cav-1, increased Keap-1/Nrf-2 dissociation and further augmented levels of Nrf-2 and HO-1 in the right frontal perisurgical area, leading to enhanced levels of tight junction proteins following SBI. The BBB protective effect of Mfsd2a was blocked by selective inhibitors of Nrf-2 and HO-1. CONCLUSIONS: Mfsd2a attenuates BBB disruption through cav-1/Nrf-2/HO-1 signaling pathway in rats subjected to experimental SBI.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Lesiones Encefálicas/fisiopatología , Transducción de Señal/genética , Animales , Conducta Animal , Agua Corporal/metabolismo , Lesiones Encefálicas/genética , Lesiones Encefálicas/terapia , Caveolina 1/genética , Lóbulo Frontal/lesiones , Terapia Genética , Hemo Oxigenasa (Desciclizante)/genética , Proteína 1 Asociada A ECH Tipo Kelch/genética , Masculino , Factor 2 Relacionado con NF-E2/genética , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
14.
Exp Neurol ; 330: 113359, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32428505

RESUMEN

The activation of tyrosine kinase receptor c-Met by hepatocyte growth factor (HGF) showed an anti-apoptotic effect in numerous disease models. This study aimed to investigate the neuroprotective mechanism of the HGF/c-Met axis-mediated anti-apoptosis underlying the delayed recanalization in a rat model of middle cerebral artery occlusion (MCAO). Permanent MCAO model (pMCAO) was induced by intravascular filament insertion. Recanalization was induced by withdrawing the filament at 3 days after MCAO (rMCAO). HGF levels in the blood serum and brain tissue expressions of HGF, c-Met, phosphorylated-STAT3 (p-STAT3), STAT3, Bcl-2, Bax, cleaved caspase-3(CC3) were assessed using ELISA and western blot, respectively. To study the mechanism, HGF small interfering ribonucleic acid (siRNA) and c-Met inhibitor, su11274, were administered intracerebroventricularly (i.c.v.) or intranasally, respectively. The concentration of HGF in the serum was increased significantly after MCAO. Brain expression of HGF was increased after MCAO and peaked at 3 days after recanalization. HGF and c-Met were both co-localized with neurons. Compared to rats received permanent MCAO, delayed recanalization after MCAO decreased the infarction volume, inhibited neuronal apoptosis, and improved neurobehavioral function, increased expressions of p-STAT3 and its downstream Bcl-2. Mechanistic studies indicated that HGF siRNA and su11274 reversed the neuroprotection including anti-apoptotic effects provided by delayed recanalization. In conclusion, the delayed recanalization after MCAO increased the expression of HGF in the brain, and reduced the infarction and neuronal apoptosis after MCAO, partly via the activation of the HGF/c-Met/STAT3/Bcl-2 signaling pathway. The delayed recanalization may serve as a therapeutic alternative for a subset of ischemic stroke patients.


Asunto(s)
Apoptosis/fisiología , Factor de Crecimiento de Hepatocito/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Infarto de la Arteria Cerebral Media/patología , Accidente Cerebrovascular Isquémico/patología , Masculino , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología
15.
Mol Neurobiol ; 57(6): 2600-2619, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32253733

RESUMEN

cGAS is a sensor of cytosolic DNA and responds equally to exogenous and endogenous DNA. After recognition of cytosolic dsDNA or ssDNA, cGAS synthesizes the second messenger 2'3'-cGAMP, which then binds to and activates stimulator of interferon genes (STING). STING plays an essential role in responding to pathogenic DNA and self-DNA in the context of autoimmunity. In pathologic conditions, such as stroke or hypoxia-ischemia (HI), DNA can gain access into the cytoplasm of the cell and leak from the dying cells into the extracellular environment, which potentially activates cGAS/STING. Recent in vivo studies of myocardial ischemia, traumatic brain injury, and liver damage models suggest that activation of cGAS/STING is not only a side-effect of the injury, but it can also actively contribute to cell death and apoptosis. We found, for the first time, that cGAS/STING pathway becomes activated between 24 and 48 h after HI in a 10-day-old rat model. Silencing STING with siRNA resulted in decreased infarction area, reduced cortical neurodegeneration, and improved neurobehavior at 48 h, suggesting that STING can contribute to injury progression after HI. STING colocalized with lysosomal marker LAMP-1 and blocking STING reduced the expression of cathepsin B and decreased the expression of Bax and caspase 3 cleavage. We observed similar protective effects after intranasal treatment with cGAS inhibitor RU.521, which were reversed by administration of STING agonist 2'3'-cGAMP. Additionally, we showed that long interspersed element 1 (LINE-1) retrotransposon, a potential upstream activator of cGAS/STING pathway was induced at 48 h after HI, which was evidenced by increased expression of ORF1p and ORF2p proteins and increased LINE-1 DNA content in the cytosol. Blocking LINE-1 with the nucleoside analog reverse-transcriptase inhibitor (NRTI) stavudine reduced infarction area, neuronal degeneration in the cerebral cortex, and reduced the expression of Bax and cleaved caspase 3. Thus, our results identify the cGAS/STING pathway as a potential therapeutic target to inhibit delayed neuronal death after HI.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Elementos de Nucleótido Esparcido Largo/fisiología , Proteínas de la Membrana/metabolismo , Degeneración Nerviosa/metabolismo , Nucleotidiltransferasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Catepsina B/metabolismo , Muerte Celular/fisiología , Modelos Animales de Enfermedad , Silenciador del Gen , Hipoxia-Isquemia Encefálica/patología , Proteínas de Membrana de los Lisosomas/metabolismo , Proteínas de la Membrana/genética , Degeneración Nerviosa/patología , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/genética , ARN Interferente Pequeño , Ratas , Transducción de Señal/fisiología , Proteína X Asociada a bcl-2/metabolismo
16.
Neurotherapeutics ; 17(4): 1954-1972, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32918234

RESUMEN

Brain edema is a vital contributor to early brain injury after subarachnoid hemorrhage (SAH), which is responsible for prolonged hospitalization and poor outcomes. Pharmacological therapeutic targets on edema formation have been the focus of research for decades. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to participate in neural development and brain injury. Here, we used PACAP knockout CRISPR to demonstrate that endogenous PACAP plays an endogenous neuroprotective role against brain edema formation after SAH in rats. The exogenous PACAP treatment provided both short- and long-term neurological benefits by preserving the function of the blood-brain barrier and glymphatic system after SAH. Pretreatment of inhibitors of PACAP receptors showed that the PACAP-involved anti-edema effect and neuroprotection after SAH was facilitated by the selective PACAP receptor (PAC1). Further administration of adenylyl cyclase (AC) inhibitor and sulfonylurea receptor 1 (SUR1) CRISPR activator suggested that the AC-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) axis participated in PACAP signaling after SAH, which inhibited the expression of edema-related proteins, SUR1 and aquaporin-4 (AQP4), through SUR1 phosphorylation. Thus, PACAP may serve as a potential clinical treatment to alleviate brain edema in patients with SAH.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/tratamiento farmacológico , Sistema Glinfático/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Sistema Glinfático/metabolismo , Sistema Glinfático/patología , Masculino , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/patología
17.
Exp Neurol ; 320: 112982, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31247196

RESUMEN

Neuroinflammation plays a crucial role in the pathological development after neonatal hypoxia-ischemia (HI). Resolvin D1 (RvD1), an agonist of formyl peptide receptor 2 (FPR2), has been shown to exert anti-inflammatory effects in many diseases. The objective of this study was to explore the protective role of RvD1 through reducing inflammation after HI and to study the contribution of Ras-related C3 botulinum toxin substrate 1 (Rac1)/nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) pathways in RvD1-mediated protection. Rat pups (10-day old) were subjected to HI or sham surgery. RvD1 was administrated by intraperitoneal injection 1 h after HI. FPR2 small interfering ribonucleic acid (siRNA) and Rac1 activation CRISPR were administered prior to RvD1 treatment to elucidate the possible mechanisms. Time course expression of FPR2 by Western blot and RvD1 by ELISA were conducted at 6 h, 12 h, 24 h, 48 h and 72 h post HI. Infarction area, short-term neurological deficits, immunofluorescent staining and Western blot were conducted at 24 h post HI. Long-term neurological behaviors were evaluated at 4 weeks post HI. Endogenous expression levels of RvD1 decreased in time dependent manner while the expression of FPR2 increased after HI, peaking at 24 h post HI. Activation of FPR2, with RvD1, reduced percent infarction area, and alleviated short- and long-term neurological deficits. Administration of RvD1 attenuated inflammation after HI, while, either inhibition of FPR2 with siRNA or activation of Rac1 with CRISPR reversed those effects. Our results showed that RvD1 attenuated neuroinflammation through FPR2, which then interacted with Rac1/NOX2 signaling pathway, thereby reducing infarction area and alleviating neurological deficits after HI in neonatal rat pups. RvD1 may be a potential therapeutic approach to reduce inflammation after HI.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Hipoxia-Isquemia Encefálica/metabolismo , Inflamación/metabolismo , Fármacos Neuroprotectores/farmacología , Receptores de Lipoxina/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Animales Recién Nacidos , Ácidos Docosahexaenoicos/metabolismo , Hipoxia-Isquemia Encefálica/patología , Inflamación/patología , NADPH Oxidasa 2/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Proteína de Unión al GTP rac1/metabolismo
18.
Free Radic Biol Med ; 141: 322-337, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31279091

RESUMEN

Neuronal apoptosis induced by oxidative stress is one of the major pathological processes involved in neurological impairment after hypoxic-ischemic encephalopathy (HIE). Ghrelin, the unique endogenous ligand for the growth hormone secretagogue receptor-1α (GHSR-1α), could take an anti-apoptotic role in the brain. However, whether ghrelin can attenuate neuronal apoptosis by attenuating oxidative stress after hypoxia-ischemia (HI) insult remains unknown. To investigate the beneficial effects of ghrelin on oxidative stress injury and neuronal apoptosis induced by HI, ten-day old unsexed rat pups were subjected to HI injury and exogenous recombinant human ghrelin(rh-Ghrelin) was administered intranasally at 1 h and 24 h after HI induction. [D-Lys3]-GHRP-6, a selective inhibitor of GHSR-1α and Ex527, a selective inhibitor of GHSR-1α were administered intranasally at 1 h before HI induction respectively. Small interfering ribonucleic acid (siRNA) for GHSR-1α were administered by intracerebroventricular (i.c.v) injection at 24 h before HI induction. Neurological tests, immunofluorescence, MitoSox staining, Fluoro-Jade C staining, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, and western blot experiments were performed. Our results indicated that ghrelin significantly improved neurobehavioral outcomes and reduced oxidative stress and neuronal apoptosis. Moreover, ghrelin treatment significantly promoted phosphorylation of AMPK, upregulated the expression of Sirt1, PGC-1α, UCP2 and the ratio of Bcl2/Bax, while it downregulated cleaved caspase-3 levels. The protective effects of ghrelin were reversed by [D-Lys3]-GHRP-6, GHSR-1α siRNA or Ex527. In conclusion, our data demonstrated that ghrelin reduced oxidative stress injury and neuronal apoptosis which was in part via the GHSR-1α/AMPK/Sirt1/PGC-1α/UCP2 signalling pathway after HI. Ghrelin may be a novel therapeutic target for treatment after neonatasl HI injury.


Asunto(s)
Ghrelina/farmacología , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Proteínas Quinasas/genética , Ratas , Receptores de Ghrelina/genética , Transducción de Señal/efectos de los fármacos , Sirtuina 1/genética , Proteína Desacopladora 2/genética
19.
Neuropharmacology ; 140: 150-161, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30086290

RESUMEN

Activation of peroxisome proliferator-activated receptor beta/delta (PPAR-ß/δ), a nuclear receptor acting as a transcription factor, was shown to be protective in various models of neurological diseases. However, there is no information about the role of PPAR-ß/δ as well as its molecular mechanisms in neonatal hypoxia-ischemia (HI). In the present study, we hypothesized that PPAR-ß/δ agonist GW0742 can activate miR-17-5p, consequently inhibiting TXNIP and ASK1/p38 pathway leading to attenuation of apoptosis. Ten-day-old rat pups were subjected to right common carotid artery ligation followed by 2.5 h hypoxia. GW0742 was administered intranasally 1 and 24 h post HI. PPAR-ß/δ receptor antagonist GSK3787 was administered intranasally 1 h before and 24 h after HI, antimir-17-5p and TXNIP CRISPR activation plasmid were administered intracerebroventricularly 24 and 48 h before HI, respectively. Brain infarct area measurement, neurological function tests, western blot, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), Fluoro-Jade C and immunofluorescence staining were conducted. GW0742 reduced brain infarct area, brain atrophy, apoptosis, and improved neurological function at 72 h and 4 weeks post HI. Furthermore, GW0742 treatment increased PPAR-ß/δ nuclear expression and miR-17-5p level and reduced TXNIP in ipsilateral hemisphere after HI, resulting in inhibition of ASK1/p38 pathway and attenuation of apoptosis. Inhibition of PPAR-ß/δ receptor and miR-17-5p and activation of TXNIP reversed the protective effects. For the first time, we provide evidence that intranasal administration of PPAR-ß/δ agonist GW0742 attenuated neuronal apoptosis at least in part via PPAR-ß/δ/miR-17/TXNIP pathway. GW0742 could represent a therapeutic target for treatment of neonatal hypoxic ischemic encephalopathy (HIE).


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/fisiología , Hipoxia-Isquemia Encefálica/fisiopatología , MicroARNs/fisiología , PPAR delta/fisiología , PPAR-beta/fisiología , Animales , Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/patología , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Neuronas/patología , PPAR delta/agonistas , PPAR delta/antagonistas & inhibidores , PPAR delta/biosíntesis , PPAR-beta/agonistas , PPAR-beta/antagonistas & inhibidores , PPAR-beta/biosíntesis , Ratas , Transducción de Señal/fisiología , Sulfonas/farmacología , Tiazoles/farmacología , Tiazoles/uso terapéutico
20.
Pharmacol Rep ; 68(5): 1076-83, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27552063

RESUMEN

BACKGROUND: Perinatal hypoxia-ischemia causes brain injury in neonates, but a fully successful treatment to prevent changes in the brain has yet to be developed. The aim of this study was to evaluate the effect of combining memantine treatment with HBO (2.5 ATA) or HH (0.47 ATA) on neonatal hypoxia-ischemia brain injury. METHODS: 7-day old rats were subjected to hypoxia-ischemia (H-I) and treated with combination of memantine and HBO or HH. The brain damage was evaluated by examination of infarct area and the number of apoptotic cells in CA1 region of hippocampus. Additionally, the level of reactive oxygen species (ROS) was measured. RESULTS: Memantine, HBO or HH postconditioning applied at short time (1-6h) after H-I, and repeated for two subsequent days, resulted in significant neuroprotection. The reduction in ipsilateral hemisphere weight deficit and in the size of infarct area was observed 14days after H-I. A reduction in apoptosis and ROS level was also observed. Combining memantine with HBO or HH resulted in a loss of neuroprotection. CONCLUSIONS: Our results show that, combining HBO or HH postconditioning with memantine produce no additive increase in the neuroprotective effect. On the contrary, combining the treatments resulted in lower neuroprotection in comparison to the effects of memantine, HBO or HH alone.


Asunto(s)
Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Memantina/farmacología , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oxígeno/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Modelos Animales de Enfermedad , Oxigenoterapia Hiperbárica/métodos , Hipoxia/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA