Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Neuroinflammation ; 21(1): 49, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355633

RESUMEN

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) constitute a recently discovered bone-marrow-derived cell type useful for dealing with neuroinflammatory disorders. However, these cells are only formed during inflammatory conditions from immature myeloid cells (IMCs) that acquire immunosuppressive activity, thus being commonly gathered from diseased animals. Then, to obtain a more clinically feasible source, we characterized IMCs directly derived from healthy bone marrow and proved their potential immunosuppressive activity under pathological conditions in vitro. We then explored their neuroprotective potential in a model of human cerebellar ataxia, the Purkinje Cell Degeneration (PCD) mouse, as it displays a well-defined neurodegenerative and neuroinflammatory process that can be also aggravated by invasive surgeries. METHODS: IMCs were obtained from healthy bone marrow and co-cultured with activated T cells. The proliferation and apoptotic rate of the later were analyzed with Tag-it Violet. For in vivo studies, IMCs were transplanted by stereotactic surgery into the cerebellum of PCD mice. We also used sham-operated animals as controls of the surgical effects, as well as their untreated counterparts. Motor behavior of mice was assessed by rotarod test. The Purkinje cell density was measured by immunohistochemistry and cell death assessed with the TUNEL technique. We also analyzed the microglial phenotype by immunofluorescence and the expression pattern of inflammation-related genes by qPCR. Parametric tests were applied depending on the specific experiment: one or two way ANOVA and Student's T test. RESULTS: IMCs were proven to effectively acquire immunosuppressive activity under pathological conditions in vitro, thus acting as MDSCs. Concerning in vivo studios, sham-operated PCD mice suffered detrimental effects in motor coordination, Purkinje cell survival and microglial activation. After intracranial administration of IMCs into the cerebellum of PCD mice, no special benefits were detected in the transplanted animals when compared to untreated mice. Nonetheless, this transplant almost completely prevented the impairments caused by the surgery in PCD mice, probably by the modulation of the inflammatory patterns. CONCLUSIONS: Our work comprise two main translational findings: (1) IMCs can be directly used as they behave as MDSCs under pathological conditions, thus avoiding their gathering from diseased subjects; (2) IMCs are promising adjuvants when performing neurosurgery.


Asunto(s)
Cerebelo , Células Mieloides , Ratones , Humanos , Animales , Células Mieloides/metabolismo , Células de Purkinje/patología , Monocitos , Inmunosupresores
2.
Cell Mol Neurobiol ; 44(1): 36, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637346

RESUMEN

Surgical brain injury (SBI), induced by neurosurgical procedures or instruments, has not attracted adequate attention. The pathophysiological process of SBI remains sparse compared to that of other central nervous system diseases thus far. Therefore, novel and effective therapies for SBI are urgently needed. In this study, we found that neutrophil extracellular traps (NETs) were present in the circulation and brain tissues of rats after SBI, which promoted neuroinflammation, cerebral edema, neuronal cell death, and aggravated neurological dysfunction. Inhibition of NETs formation by peptidylarginine deiminase (PAD) inhibitor or disruption of NETs with deoxyribonuclease I (DNase I) attenuated SBI-induced damages and improved the recovery of neurological function. We show that SBI triggered the activation of cyclic guanosine monophosphate-adenosine monophosphate synthase stimulator of interferon genes (cGAS-STING), and that inhibition of the cGAS-STING pathway could be beneficial. It is worth noting that DNase I markedly suppressed the activation of cGAS-STING, which was reversed by the cGAS product cyclic guanosine monophosphate-adenosine monophosphate (cGMP-AMP, cGAMP). Furthermore, the neuroprotective effect of DNase I in SBI was also abolished by cGAMP. NETs may participate in the pathophysiological regulation of SBI by acting through the cGAS-STING pathway. We also found that high-dose vitamin C administration could effectively inhibit the formation of NETs post-SBI. Thus, targeting NETs may provide a novel therapeutic strategy for SBI treatment, and high-dose vitamin C intervention may be a promising translational therapy with an excellent safety profile and low cost.


Asunto(s)
Lesiones Encefálicas , Trampas Extracelulares , Animales , Ratas , Encéfalo , Lesiones Encefálicas/tratamiento farmacológico , Ácido Ascórbico , Desoxirribonucleasa I/farmacología
3.
Clin Exp Pharmacol Physiol ; 50(12): 927-935, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37675456

RESUMEN

Ferroptosis is a recently discovered non-apoptotic form of cellular death. Acyl-CoA synthetase long-chain family number 4 (ACSL4) is necessary for iron-dependent cellular death, and reactive oxygen species (ROS) produced by ACSL4 are the executioners of ferroptosis. Rosiglitazone improves ferroptosis by inhibiting ACSL4. There is no research indicating whether ACSL4 plays a role in cell death after surgical brain injury (SBI). This study aimed to investigate the role of ACSL4 in SBI via the ferroptosis pathway. Ninety male Sprague-Dawley rats were examined using a model of SBI. Subsequently, the inhibitory effect of rosiglitazone on ACSL4 was assessed via western blot, real-time polymerase chain reaction (PCR), immunofluorescence, fluoro-jade C staining, Perl's staining, ROS assay, and neurological scoring. The results showed that compared with the Sham group, the protein levels of ACSL4 and transferrin were significantly increased after SBI. Administration of rosiglitazone significantly reduced neuronal necrosis, iron deposition, brain water content and ROS in brain tissue and ameliorated neurological deficits at 48 h after SBI, which was concomitant with decreased transferrin expression. These findings demonstrate that SBI-induced upregulation of ACSL4 may be partly mediated by the ferroptosis pathway, which can be reversed by rosiglitazone administration.


Asunto(s)
Lesiones Encefálicas , Neoplasias Encefálicas , Ratas , Masculino , Animales , Rosiglitazona/farmacología , Especies Reactivas de Oxígeno/metabolismo , Ratas Sprague-Dawley , Encéfalo/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Hierro , Transferrinas/metabolismo , Ligasas/metabolismo
4.
J Neurosci Res ; 98(1): 155-167, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31257634

RESUMEN

Neurosurgical procedures result in surgically induced brain injury (SBI) that causes postoperative complications including brain edema and neuronal apoptosis in the surrounding brain tissue. SBI leads to the release of cytokines that indirectly cause the stimulation of kynurenine 3-monooxygenase (KMO) and the release of neurotoxic quinolinic acid (QUIN). This study tested a KMO inhibitor, RO 61-8048, to prevent postoperative brain edema and consequent neuronal apoptosis in an in vivo model of SBI. A rodent model of SBI was utilized which involves partial resection of the right frontal lobe. A total of 127 Sprague-Dawley male rats (weight 275-325 g) were randomly divided into the following groups: Sham surgical group, SBI, SBI + DMSO, SBI + RO 61-8048 (10 mg/kg), SBI + RO 61-8048 (40 mg/kg), and SBI + RO 61-8048 (40 mg/kg) + KAT II inhibitor PF-04859989 (5 mg/kg). RO 61-8048 was administered by intraperitoneal injection after SBI. Postoperative assessment at different time points included brain water content (brain edema), neurological scoring, and western blot. SBI increased brain water content (ipsilateral frontal lobe), decreased neurological function, and increased apoptotic markers compared with sham animals. Treatment with RO 61-8048 (40 mg/kg) reduced brain water content and improved long-term neurological function after SBI. RO 61-8048 increased the expression of kynurenic acid while reducing QUIN and apoptotic markers in the surrounding brain tissue after SBI. These neuroprotective effects were reversed by PF-04859989. This study suggests KMO inhibition via RO 61-8048 as a potential postoperative therapy following neurosurgical procedures.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Quinurenina 3-Monooxigenasa/metabolismo , Quinurenina/metabolismo , Fármacos Neuroprotectores/farmacología , Sulfonamidas/farmacología , Tiazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Edema Encefálico/metabolismo , Lesiones Encefálicas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Sulfonamidas/uso terapéutico , Tiazoles/uso terapéutico
5.
Neurochem Res ; 44(11): 2470-2481, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31482256

RESUMEN

Neuroinflammation and brain edema are major complications in the pathophysiology of surgical brain injury (SBI). Programmed death-ligand 1 (PD-L1), an immune inhibitory receptor ligand, has been increasingly investigated for inhibition of T cell-mediated immunity and braking inflammatory response. However, the negative immunomodulatory capacity of PD-L1 and their possible mechanism in SBI is not yet clear. This study aimed to evaluate the expression and the role of PD-L1 in a mouse model of SBI induced inflammation and to further study the potential therapeutic effects of PD-L1 on SBI. Here we showed that PD-L1 expression was markedly elevated in the surrounding peri-resection brain tissue post-SBI in vivo. PD-L1 was up-regulated through ERK signal pathway in LPS-treated BV-2 cells in vitro. Furthermore, blockade of the PD-L1 checkpoint using PD-L1 antibody significantly enhanced brain edema, exacerbated apoptosis and increased neurodeficits post-SBI. Moreover, activated PD-1/PD-L1 with PD-L1 protein significantly attenuated the inflammation responses and brain edema post-SBI. These results suggest that enhanced expression of PD-L1 post-SBI exerts self-protection from inflammation and promotes neurological repair. PD-L1 signal may have therapeutic potential for neurodegenerative disorders.


Asunto(s)
Antígeno B7-H1/metabolismo , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Inflamación/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/patología , Encéfalo/cirugía , Edema Encefálico/metabolismo , Línea Celular , Femenino , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos C57BL , Microglía/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
Neurobiol Dis ; 107: 66-72, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28286182

RESUMEN

INTRODUCTION: Postoperative cerebral edema is a devastating complication in neurosurgical patients. Loss of blood-brain barrier integrity has been shown to lead to the development of brain edema following neurosurgical procedures. The aim of this study was to evaluate preconditioning with Crotalus helleri venom (Cv-PC) as a potential preventive therapy for reducing postoperative brain edema in the rodent SBI model. C. helleri venom is known to contain phospholipase A2 (PLA2), an enzyme upstream to cyclooxygenase-2 (COX-2) in the inflammatory cascade, acts to increase the production of inflammatory mediators, such as prostaglandins. We hypothesize that Cv-PC will downregulate the response of the COX-2 pathway to injury, thereby reducing the inflammatory response and the development of brain edema after SBI. MATERIALS AND METHODS: 75 male Sprague Dawley rats (280-330g) were divided to the following groups-naïve+vehicle, naïve+Cv-PC, sham, vehicle, Cv-PC, Cv-PC+NS398 (COX-2 inhibitor). Vehicle preconditioned and Cv-PC animals received either three daily subcutaneous doses of saline or C. helleri venom at 72h, 48h, and 24h prior to surgery. In Cv-PC+NS398 animals, NS398 was administered intraperitoneally 1h prior to each Cv-PC injection. Sham-operated animals received craniotomy only, whereas SBI animals received a partial right frontal lobectomy. Neurological testing and brain water content were assessed at 24h and 72h after SBI; COX-2 and PGE2 expression was assessed at 24h postoperatively by Western blot and immunohistochemistry, respectively. RESULTS: At 24h after SBI, the vehicle-treated animals were observed to have increased brain water content (83.1±0.2%) compared to that of sham animals (80.2±0.1%). The brain water content of vehicle-treated animals at 72h post-SBI was elevated at 83.3±0.2%. Cv-PC-treated animals with doses of 10% LD50 had significantly reduced brain water content of 81.92±0.7% and 81.82±0.3% at 24h and 72h, respectively, after SBI compared to that of vehicle-treated animals, while Cv-PC with 5% LD50 doses showed brain water content that trended lower but did not reach statistical significance. At 24h and 72h post-SBI, Cv-PC-treated animals had significantly higher neurological score than vehicle-treated animals. The COX-2 over-expression characterized in SBI was attenuated in Cv-PC-treated animals; NS398 reversed the protective effect of Cv-PC on COX-2 expression. Cv-PC tempered the over-expression of the inflammatory marker PGE2. CONCLUSION: Our findings indicate that Cv-PC may provide a promising therapy for reducing postoperative edema and improving neurological function after neurosurgical procedures.


Asunto(s)
Edema Encefálico/prevención & control , Encéfalo/cirugía , Lóbulo Frontal/lesiones , Complicaciones Intraoperatorias/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Complicaciones Posoperatorias/tratamiento farmacológico , Venenos de Serpiente/administración & dosificación , Animales , Agua Corporal/efectos de los fármacos , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Crotalus , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Epidermis/efectos de los fármacos , Epidermis/inmunología , Epidermis/patología , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/patología , Lóbulo Frontal/cirugía , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Complicaciones Intraoperatorias/metabolismo , Complicaciones Intraoperatorias/patología , Masculino , Procedimientos Neuroquirúrgicos , Nitrobencenos/farmacología , Complicaciones Posoperatorias/metabolismo , Complicaciones Posoperatorias/patología , Ratas Sprague-Dawley , Sulfonamidas/farmacología
7.
Neurobiol Dis ; 85: 164-173, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26550694

RESUMEN

BACKGROUND AND PURPOSE: Peripheral immune cell infiltration to the brain tissue at the perisurgical site can promote neuroinflammation after surgical brain injury (SBI). Slit2, an extracellular matrix protein, has been reported to reduce leukocyte migration. This study evaluated the effect of recombinant Slit2 and the role of its receptor roundabout1 (Robo1) and its downstream mediator Slit-Robo GTPase activating protein 1 (srGAP1)-Cdc42 on peripheral immune cell infiltration after SBI in a rat model. METHODS: One hundred and fifty-three adult male Sprague-Dawley rats (280-350 g) were used. Partial resection of right frontal lobe was performed to induce SBI. Slit2 siRNA was administered by intracerebroventricular injection 24h before SBI. Recombinant Slit2 was injected intraperitoneally 1h before SBI. Recombinant Robo1 used as a decoy receptor was co-administered with recombinant Slit2. srGAP1 siRNA was administered by intracerebroventricular injection 24h before SBI. Post-assessments included brain water content measurement, neurological tests, ELISA, Western blot, immunohistochemistry, and Cdc42 activity assay. RESULTS: Endogenous Slit2 was increased after SBI. Robo1 was expressed by peripheral immune cells. Endogenous Slit2 knockdown worsened brain edema after SBI. Recombinant Slit2 administration reduced brain edema, neurological deficits, and pro-inflammatory cytokines after SBI. Recombinant Slit2 reduced peripheral immune cell markers cluster of differentiation 45 (CD45) and myeloperoxidase (MPO), as well as Cdc42 activity in the perisurgical brain tissue which was reversed by recombinant Robo1 co-administration and srGAP1 siRNA. CONCLUSIONS: Recombinant Slit2 improved outcomes by reducing neuroinflammation after SBI, possibly by decreasing peripheral immune cell infiltration to the perisurgical site through Robo1-srGAP1 mediated inhibition of Cdc42 activity. These results suggest that Slit2 may be beneficial to reduce SBI-induced neuroinflammation.


Asunto(s)
Lesiones Encefálicas/inmunología , Lóbulo Frontal/inmunología , Lóbulo Frontal/lesiones , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Complicaciones Intraoperatorias/inmunología , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Animales , Edema Encefálico/etiología , Edema Encefálico/inmunología , Edema Encefálico/terapia , Lesiones Encefálicas/etiología , Lesiones Encefálicas/terapia , Modelos Animales de Enfermedad , Lóbulo Frontal/cirugía , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Técnicas de Silenciamiento del Gen , Terapia Genética/métodos , Infusiones Intraventriculares , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Complicaciones Intraoperatorias/terapia , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas Sprague-Dawley , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes/genética , Proteínas Roundabout
8.
Acta Neurochir Suppl ; 121: 299-304, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463965

RESUMEN

The surgical brain injury model replicates neurosurgical brain parenchymal damage. Postsurgical brain edema correlates with postoperative neurological dysfunction. Intranasal administration is a proven method of delivering therapies to brain tissue. Thrombin preconditioning decreased brain edema and improved neurological outcomes in models of ischemic brain injury. We hypothesized thrombin preconditioning in surgical brain injury may improve postoperative brain edema and neurological outcomes. Adult male Sprague-Dawley rats (n = 78) weighing 285-355 g were randomly assigned to sham or pre-injury treatment: one-time pretreatment 1 day prior, one-time pretreatment 5 days prior, and daily preconditioning for 5 days prior. Treatment arms were divided into vehicle or thrombin therapies, and subdivided into intranasal (thrombin 5 units/50 µL 0.9 % saline) or intracerebral ventricular (thrombin 0.1 unit/10 µL 0.9 % saline) administration. Blinded observers performed neurological testing 24 h after brain injury followed immediately by measurement of brain water content. There was a significant difference in ipsilateral brain water content and neurological outcomes between all treatment groups and the sham group. However, there was no change in brain water content or neurological outcomes between thrombin- and vehicle-treated animals. Thrombin preconditioning did not significantly improve brain edema or neurological function in surgical brain injury in rats.


Asunto(s)
Conducta Animal/efectos de los fármacos , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Encéfalo/efectos de los fármacos , Hemostáticos/farmacología , Precondicionamiento Isquémico , Procedimientos Neuroquirúrgicos , Trombina/farmacología , Administración Intranasal , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Edema Encefálico/etiología , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Modelos Animales de Enfermedad , Lóbulo Frontal/cirugía , Inyecciones Intraventriculares , Complicaciones Intraoperatorias , Masculino , Ratas , Ratas Sprague-Dawley
9.
Acta Neurochir Suppl ; 121: 317-21, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463968

RESUMEN

No matter how carefully a neurosurgical procedure is performed, it is intrinsically linked to postoperative deficits resulting in delayed healing caused by direct trauma, hemorrhage, and brain edema, termed surgical brain injury (SBI). Cerebral edema occurs several hours after SBI and is a major contributor to patient morbidity, resulting in increased postoperative care. Currently, the correlation between functional recovery and brain edema after SBI remains unknown. Here we examine the correlation between neurological function and brain water content in rats 42 h after SBI. SBI was induced in male Sprague-Dawley rats via frontal lobectomy. Twenty-four hours post-ictus animals were subjected to four neurobehavior tests: composite Garcia neuroscore, beam walking test, corner turn test, and beam balance test. Animals were then sacrificed for right-frontal brain water content measurement via the wet-dry method. Right-frontal lobe brain water content was found to significantly correlate with neurobehavioral deficits in the corner turn and beam balance tests: the number of left turns (percentage of total turns) for the corner turn test and distance traveled for the beam balance test were both inversely proportional with brain water content. No correlation was observed for the composite Garcia neuroscore or the beam walking test.


Asunto(s)
Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Procedimientos Neuroquirúrgicos , Animales , Conducta Animal , Edema Encefálico/etiología , Lesiones Encefálicas/complicaciones , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Lóbulo Frontal/cirugía , Complicaciones Intraoperatorias , Masculino , Ratas , Ratas Sprague-Dawley , Trastornos de la Sensación/etiología , Trastornos de la Sensación/fisiopatología
10.
Brain Inj ; 28(12): 1594-601, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25093611

RESUMEN

BACKGROUND: Dexamethasone (DEXA) is commonly used to reduce brain swelling during neurosurgical procedures. DEXA, however, has many side-effects that can increase the risks of post-operative complications. In contrast, progesterone (PRO) has fewer side-effects and has been found to be neuroprotective on traumatic brain injury (TBI). Whether PRO may be used as an alternative to DEXA during routine procedures has not been fully explored. OBJECT: To compare the effects of DEXA and PRO on surgical brain injury (SBI). METHODS: Seventy-five adult male Sprague Dawley rats were randomized into five groups: (1) SBI + drug vehicle (peanut oil, 1 ml kg(-1)); (2) SBI + DEXA (1 mg kg(-1)); (3) SBI + low-dose PRO (10 mg kg(-1)); (4) SBI + high-dose PRO (20 mg kg(-1)); and (5) sham SBI + drug vehicle. Magnetic resonance imaging study and assessments of brain water content (BWC), blood-brain barrier (BBB) permeability, cellular inflammatory responses and matrix metalloproteinase 9 (MMP-9) expression were conducted. RESULTS: This model consistently resulted in increased BWC and BBB disruption. PRO reduced astrocyte and microglia responses and attenuated brain oedema with preservation of BBB. A significant down-regulation of MMP-9 expression occurred in the PRO 20 group. CONCLUSIONS: PRO is as effective as DEXA in reducing brain oedema and inflammation following SBI; 10 mg kg(-1) of PRO was demonstrated to be more effective in relieving acute cellular inflammatory responses.


Asunto(s)
Edema Encefálico/metabolismo , Lesiones Encefálicas/metabolismo , Dexametasona/farmacología , Glucocorticoides/farmacología , Inflamación/metabolismo , Procedimientos Neuroquirúrgicos/efectos adversos , Progesterona/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Western Blotting , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/inmunología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/cirugía , Modelos Animales de Enfermedad , Regulación hacia Abajo , Inflamación/tratamiento farmacológico , Masculino , Inhibidores de la Metaloproteinasa de la Matriz , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
11.
Brain Res Bull ; 215: 111033, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39032586

RESUMEN

Brain Muscle ARNT-Like Protein 1 (BMAL1) suppresses oxidative stress in brain injury during surgery. Epigallocatechin-3-gallate (EGCG), a monomer in green tea, has been identified as an antioxidant and a potential agonist for BMAL1. In this work, the mechanism by which BMAL1 is regulated was investigated, as well as the therapeutic effect of EGCG on surgically injured rats. The pathological environment after brain injury during surgery was simulated by excising the right frontal lobe of rats. Rats received an intraperitoneal injection of EGCG immediately after surgery. Neurological scores and cerebral edema were recorded after surgery. Fluoro-Jade C staining, TUNEL staining, western blot, and lipid peroxidation analyses were conducted 3 days later. Here we show that the endogenous BMAL1 level decreased after brain injury. Postoperative administration of EGCG up-regulated the content of BMAL1 around the cerebral cortex, reduced the oxidative stress level, reduced neuronal apoptosis and the number of degenerated neurons, alleviated cerebral edema, and improved neurological scores in rats. This suggests that BMAL1 is an effective target for treating surgical brain injury, as well as that EGCG may be a promising agent for alleviating postoperative brain injury.


Asunto(s)
Factores de Transcripción ARNTL , Catequina , Ratas Sprague-Dawley , Regulación hacia Arriba , Animales , Catequina/análogos & derivados , Catequina/farmacología , Factores de Transcripción ARNTL/metabolismo , Masculino , Regulación hacia Arriba/efectos de los fármacos , Ratas , Estrés Oxidativo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Modelos Animales de Enfermedad , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Edema Encefálico/metabolismo , Edema Encefálico/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Antioxidantes/farmacología
12.
Heliyon ; 9(7): e18160, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37539293

RESUMEN

Histone deacetylase 3 (HDAC3) restores chromatin nucleosomes to a transcriptional repression state, thereby inhibiting gene expression. Studies have found that HDAC3 expression is upregulated in a variety of pathological states of the central nervous system and related to its neurotoxicity. However, the role of HDAC3 in surgical brain injury (SBI) has not been thoroughly explored. OBJECTIVE: To observe the role of HDAC3 in SBI and the outcome of SBI after its suppression. METHODS: Rat SBI model was used, and intraperitoneal injection of RGFP966 (HDAC3 specific inhibitor) was used to detect the changes of HDAC3 expression and neuronal apoptosis indexes in the surrounding cortex of SBI rats, and the cerebral edema and neurological outcome of rats were observed. RESULTS: The expression of HDAC3 in the peripheral cortex of SBI rats was increased, and RGFP966 inhibited the upregulation of HDAC3 and saved the nerve cells around the damaged area. In addition, RGFP966 increased the expression of anti-oxidative stress proteins such as heme oxygenase-1 (HO-1) and superoxide dismutase 2 (SOD2). At the same time, the expression of apoptotic marker protein cleaved-caspase-3 (cle-caspase-3) was decreased, while the expression level of apoptotic protective marker protein B-cell lymphoma 2 (Bcl-2) was increased. In addition, this research demonstrated that in the RGFP966 rat SBI model, the expression level of antioxidant modifier nuclear factor-erythroid 2-related factor 2 (Nrf2) was increased. CONCLUSION: RGFP966 might activate HDAC3/Nrf2 signaling pathway by inhibiting HDAC3, regulated oxidative stress and nerve cell apoptosis induced by SBI in rat SBI model, reduced brain edema, and had a protective effect on nerve injury. It might be a potential target of SBI pathology.

14.
Brain Res Bull ; 183: 116-126, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35247489

RESUMEN

Surgical brain injury (SBI) is unavoidable in neurosurgery, and could aggravate secondary brain injury. Post-brain injury, multiple inflammatory factors are released, resulting in neuroinflammation and cell apoptosis, with subsequent brain edema and nerve function injury. TREM2, an immune protein mainly expressed in microglia, is an important link for nerve cells to participate in the inflammatory response. TREM2 and nuclear factor кB (NF-кB) are indeed closely associated with the release of inflammatory cytokines following brain injury. This work aimed to determine the inflammatory function of TREM2 in SBI, and to investigate whether TREM2 regulates interleukin-1 beta (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α) release through the NF-кB p65 signaling pathway. We established a rat model of SBI, and performed Western blotting (WB), immunofluorescence (IF) and enzyme-linked immunosorbent assay (ELISA) for further analysis. Next, brain edema and neurological score analyses were performed. Finally, whether TREM2 regulating NF-кB p65 signaling affects blood-brain barrier (BBB) permeability and nerve cell apoptosis was examined. We found that post-SBI, TREM2 was upregulated, and inflammation and brain injury were aggravated. After TREM2 downregulation, NF-кB p65 production, inflammation and brain injury were enhanced, suggesting that TREM2 may play a protective role by inhibiting NF-кB p65 production after SBI. Overall, these findings suggest that TREM2 in SBI may have protective effects on postoperative nerve and BBB damage, possibly in part via the NF-κB p65 pathway.


Asunto(s)
Barrera Hematoencefálica , FN-kappa B , Animales , Apoptosis , Barrera Hematoencefálica/metabolismo , Regulación hacia Abajo , FN-kappa B/metabolismo , Ratas , Transducción de Señal
15.
World Neurosurg ; 168: e595-e606, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36280046

RESUMEN

BACKGROUND: Surgical resection is a key method for glioma treatment. This inherently invasive procedure alters the tumor microenvironment of glioma cells that cannot be removed by surgery. However, few studies have focused on the impact of this microenvironment change on the growth of glioma cells. METHODS: The authors preconstructed a surgical brain injury model, and then C6 glioma cells were transplanted. HE staining was used to observe the general morphology of tumor cells, and immunohistochemistry of MMP-2, MMP-9, GFAP, and CD31 was used to evaluate the invasiveness of glioma cells and activation of astrocytes and calculate microvessel density. In vitro, primary rat astrocytes were exposed to different temperature gradients. The supernatant was made into conditioned medium for culturing C6 glioma cells. The scratch test and transwell test were used to evaluate the migration and invasion of tumor cells. RESULTS: GFAP expression was stronger in surgical brain injury rats, C6 cells implanted in these rats showed stronger expression of MMP-2 and MMP-9, and CD31 was expressed in more microvessels. Astrocytes exposed to high temperatures of 40°C and 43°C expressed stronger GFAP, and C6 cells cultured in their supernatants had stronger scratch healing ability and the ability to cross transwell chambers. CONCLUSIONS: The microenvironment changes caused by surgical brain injury will enhance the migration and invasion of glioma cells and increase the microvessel density in the tumor. This effect may be related to the activation of astrocytes caused by the thermal injury of bipolar coagulation during surgery.


Asunto(s)
Lesiones Encefálicas , Neoplasias Encefálicas , Glioma , Ratas , Animales , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias Encefálicas/patología , Astrocitos/metabolismo , Glioma/patología , Lesiones Encefálicas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Microambiente Tumoral
16.
Galen Med J ; 10: 1-10, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36340958

RESUMEN

Background: Glioblastoma (GBM) is the most common primary central nervous system malignancy with a low survival without extra logistics. Currently, there is no definitive chemotherapy among the studied options. This study aims to evaluate the neuroprotective effects of dimethyl fumarate (DMF) on surgical brain injuries in patients treated for GBM. Materials and Methods: This randomized, phase II, placebo, triple-blinded, controlled trial was performed on 36 patients with a diagnosis of GBM. All the patients received DMF (240 mg, three-times per day) or placebo (with the same shape and administration route) one week before surgery. Also, patients in both groups after the operation received standard treatments (radiotherapy plus chemotherapy). In addition, Kanofsky's performance status (KPS) score was evaluated at baseline and one month later. Also, serum S100ß was measured 48 hours before and after surgery. Results: There was no significant difference among DMF and control groups with regard to age, gender, and the extent of resections (P˃0.05). The most adverse event in both groups was a headache. Although the serum S100ß level was not markedly changed after surgery, the mean KPS in the DMF group was higher than in the control group after surgery. Conclusion: The DMF could be a possible good regime for the treatment of GBM; however, questions are raised regarding its efficacy and application for the addition to standard treatment.

17.
Front Surg ; 8: 749310, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35071308

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is considered as a potential target for the treatment of Parkinson's disease. This protein is expressed in the brain and has been associated with various diseases and lysosomal maintenance. Rab10 is a member of the Rab protein GTPase family that has been recently shown to be a kinase substrate of LRRK2. In addition, LRRK2 and its kinase substrate Rab10 constitute a key stress response pathway during lysosomal overload stress. This study aimed to investigate the potential role and mechanism underlying LRRK2 and its kinase substrate Rab10 involving surgical brain injury (SBI). One hundred and forty-four male Sprague-Dawley rats were examined using an SBI model, and some had received the LRRK2-specific inhibitor PF-06447475. Thereafter, western blotting, immunofluorescence, brain water content analysis, neuronal apoptosis assay, and neurological score analysis were conducted. The results showed that after SBI, LRRK2 and phosphorylated Rab10 (p-Rab10) expression in neuronal cells were upregulated, and administration of PF-06447475 significantly reduced neuronal apoptosis, neuroinflammation, and brain water content 12 h after SBI and improved neurological deficit 72 h after SBI, which is related to the decreased expression of LRRK2 and p-Rab10, and the lessening of lysosomal overload stress. Our research suggests that the inhibition of LRRK2 can effectively interfere with the role of p-Rab10 in promoting the secretion of lysosomal hydrolase in lysosomal overload stress after SBI, thereby reducing neuronal apoptosis and inflammation after SBI and playing a major role in brain protection.

18.
Cell Signal ; 85: 110048, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34015470

RESUMEN

Integrated stress response (ISR) contributes to various neuropathological processes and acting as a therapy target in CNS injuries. However, the fundamental role of ISR in regulating microglial polarization remains largely unknown. Currently no proper pharmacological approaches to reverse microglia-driven neuroinflammation in surgical brain injury (SBI) have been reported. Here we found that inhibition of the crucial ISR effector, activating transcription factor 4 (ATF4), using the RNA interference suppressed the lipopolysaccharide (LPS)-stimulated microglial M1 polarization in vitro. Interestingly, counteracting ISR with a small-molecule ISR inhibitor (ISRIB) resulted in a significant microglial M1 towards M2 phenotype switching after LPS treatment. The potential underlying mechanisms may related to downregulate the intracellular NADPH oxidase 4 (NOX4) expression under the neuroinflammatory microenvironment. Notably, ISRIB ameliorated the infiltration of microglia and improved the neurobehavioral outcomes in the SBI rat model. Overall, our findings suggest that targeting ISR exerts a novel anti-inflammatory effect on microglia via regulating M1/M2 phenotype and may represent a potential therapeutic target to overcome neuroinflammation following SBI.


Asunto(s)
Lesiones Encefálicas , Microglía , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Polaridad Celular , Ratones , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias , Ratas , Transducción de Señal
19.
Surg Neurol Int ; 12: 210, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34084637

RESUMEN

BACKGROUND: Surgical brain injury (SBI) impacts roughly 800,000 people who undergo neurosurgical procedures each year. SBI is the result of unavoidable parenchymal damage, vessel disruption, and thermal injury that is an inherent part of all neurosurgical procedures. Clinically, SBI has been associated with postoperative seizures and long-term neurobehavioral deficits. Current therapies are aimed at providing symptom relief by reducing swelling and preventing seizures. However, there are no therapies aimed at reducing the extent of SBI preoperatively. The microbiome-gut-brain axis may serve as a potential target for the development of new preventative therapies due to its extensive involvement in central nervous system function. METHODS: An extensive literature review was conducted to determine whether there is a potential role for dysbiosis treatment in reducing the extent of SBI. RESULTS: Treatment of gut dysbiosis deserves further exploration as a potential means of reducing the extent of unavoidable SBI. Dysbiosis has been correlated with increased neuroinflammation through impaired immune regulation, increased blood-brain barrier permeability, and increased production of reactive metabolites. Recently, dysbiosis has also been linked to acute neurological dysfunction in the postoperative state. Importantly, treatment of dysbiosis has been correlated with better patient outcomes and decreased length of stay in surgical patients. CONCLUSION: Current literature supports the role of dysbiosis treatment in the preoperative setting as a means of optimizing neurological recovery following unavoidable SBI that results from all neurosurgical procedures.

20.
Front Mol Neurosci ; 14: 641993, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33867933

RESUMEN

Surgical brain injury (SBI) triggers microglia to release numerous inflammatory factors, leading to brain edema and neurological dysfunction. Reducing neuroinflammation and protecting the blood-brain barrier (BBB) are key factors to improve the neurological function and prognosis after SBI. Na+-K+-Cl- cotransporter 1 (NKCC1) and nuclear factor κB (NF-κB) have been implicated in the secretion of inflammatory cytokines by microglia in brain injury. This study aimed to establish the role of NKCC1 in inducing inflammation in SBI, as well as to determine whether NKCC1 controls the release of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) via phosphorylation of NF-κB in microglia, thus affecting BBB permeability and neuronal cell apoptosis. Male Sprague-Dawley (SD) rats were used to establish an SBI model. This study revealed that compared with the sham group, the expression levels of p-NKCC1, p-p65-NF-κB, and related inflammatory factor proteins in SBI model group significantly increased. After p-NKCC1 was inhibited, p-p65-NF-κB, IL-6, IL-1ß, and TNF-α were downregulated, and nerve cell apoptosis and BBB permeability were significantly reduced. These findings suggest that the SBI-induced increase in p-NKCC1 exacerbates neuroinflammation, brain edema, and nerve function injury, which may be mediated by regulating the activity of p65-NF-κB that in turn influences the release of inflammatory factors.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA