Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nature ; 583(7814): E14, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32533095

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nature ; 582(7812): 395-398, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32494010

RESUMEN

Neuroprotectant strategies that have worked in rodent models of stroke have failed to provide protection in clinical trials. Here we show that the opposite circadian cycles in nocturnal rodents versus diurnal humans1,2 may contribute to this failure in translation. We tested three independent neuroprotective approaches-normobaric hyperoxia, the free radical scavenger α-phenyl-butyl-tert-nitrone (αPBN), and the N-methyl-D-aspartic acid (NMDA) antagonist MK801-in mouse and rat models of focal cerebral ischaemia. All three treatments reduced infarction in day-time (inactive phase) rodent models of stroke, but not in night-time (active phase) rodent models of stroke, which match the phase (active, day-time) during which most strokes occur in clinical trials. Laser-speckle imaging showed that the penumbra of cerebral ischaemia was narrower in the active-phase mouse model than in the inactive-phase model. The smaller penumbra was associated with a lower density of terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL)-positive dying cells and reduced infarct growth from 12 to 72 h. When we induced circadian-like cycles in primary mouse neurons, deprivation of oxygen and glucose triggered a smaller release of glutamate and reactive oxygen species, as well as lower activation of apoptotic and necroptotic mediators, in 'active-phase' than in 'inactive-phase' rodent neurons. αPBN and MK801 reduced neuronal death only in 'inactive-phase' neurons. These findings suggest that the influence of circadian rhythm on neuroprotection must be considered for translational studies in stroke and central nervous system diseases.


Asunto(s)
Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Modelos Animales de Enfermedad , Neuronas/patología , Neuroprotección , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/prevención & control , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/prevención & control , Glucosa/deficiencia , Humanos , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Infarto de la Arteria Cerebral Media/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Accidente Cerebrovascular/fisiopatología , Investigación Biomédica Traslacional , Insuficiencia del Tratamiento
3.
Stroke ; 53(12): e507-e511, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36321457

RESUMEN

BACKGROUND: The immune response to acute cerebral ischemia is a major factor in stroke pathobiology. Circadian biology modulates some aspects of immune response. The goal of this study is to compare key parameters of immune response during the active/awake phase versus inactive/sleep phase in a mouse model of transient focal cerebral ischemia. METHODS: Mice were housed in normal or reversed light cycle rooms for 3 weeks, and then they were blindly subjected to transient focal cerebral ischemia. Flow cytometry was used to examine immune responses in blood, spleen, and brain at 3 days after ischemic onset. RESULTS: In blood, there were higher levels of circulating T cells in mice subjected to focal ischemia during zeitgeber time (ZT)1-3 (inactive or sleep phase) versus ZT13-15 mice (active or awake phase). In the spleen, organ weight and immune cell numbers were lower in ZT1-3 versus ZT13-15 mice. Consistent with these results, there was an increased infiltration of activated T cells into brain at ZT1-3 compared with ZT13-15. CONCLUSIONS: This proof-of-concept study indicates that there are significant diurnal effects on the immune response after focal cerebral ischemia in mice. Hence, therapeutic strategies focused on immune targets should be reassessed to account for the effects of diurnal rhythms and circadian biology in nocturnal rodent models of stroke.


Asunto(s)
Isquemia Encefálica , Ataque Isquémico Transitorio , Accidente Cerebrovascular , Animales , Ratones , Bazo , Ratones Endogámicos C57BL , Encéfalo , Infarto Cerebral , Isquemia , Inmunidad
4.
Stroke ; 53(12): 3741-3750, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36252110

RESUMEN

BACKGROUND: It has been reported that the S1P (sphingosine 1-phosphate) receptor modulator fingolimod reduces infarction in rodent models of stroke. Recent studies have suggested that circadian rhythms affect stroke and neuroprotection. Therefore, this study revisited the use of fingolimod in mouse focal cerebral ischemia to test the hypothesis that efficacy might depend on whether experiments were performed during the inactive sleep or active wake phases of the circadian cycle. METHODS: Two different stroke models were implemented in male C57Bl/6 mice-transient middle cerebral artery occlusion and permanent distal middle cerebral artery occlusion. Occlusion occurred either during inactive or active circadian phases. Mice were treated with 1 mg/kg fingolimod at 30- or 60-minute postocclusion and 1 day later for permanent and transient middle cerebral artery occlusion, respectively. Infarct volume, brain swelling, hemorrhagic transformation, and behavioral outcome were assessed at 2 or 3 days poststroke. Three independent experiments were performed in 2 different laboratories. RESULTS: Fingolimod decreased peripheral lymphocyte number in naive mice, as expected. However, it did not significantly affect infarct volume, brain swelling, hemorrhagic transformation, or behavioral outcome at 2 or 3 days after transient or permanent focal cerebral ischemia during inactive or active circadian phases of stroke onset. CONCLUSIONS: Outcomes were not improved by fingolimod in either transient or permanent focal cerebral ischemia during both active and inactive circadian phases. These negative findings suggest that further testing of fingolimod in clinical trials may not be warranted unless translational studies can identify factors associated with fingolimod's efficacy or lack thereof.


Asunto(s)
Edema Encefálico , Isquemia Encefálica , Accidente Cerebrovascular , Animales , Ratones , Masculino , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Edema Encefálico/tratamiento farmacológico , Esfingosina , Accidente Cerebrovascular/tratamiento farmacológico , Ratones Endogámicos C57BL , Hemorragia/tratamiento farmacológico , Modelos Animales de Enfermedad
5.
Nature ; 535(7613): 551-5, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27466127

RESUMEN

Neurons can release damaged mitochondria and transfer them to astrocytes for disposal and recycling. This ability to exchange mitochondria may represent a potential mode of cell-to-cell signalling in the central nervous system. Here we show that astrocytes in mice can also release functional mitochondria that enter neurons. Astrocytic release of extracellular mitochondrial particles was mediated by a calcium-dependent mechanism involving CD38 and cyclic ADP ribose signalling. Transient focal cerebral ischaemia in mice induced entry of astrocytic mitochondria into adjacent neurons, and this entry amplified cell survival signals. Suppression of CD38 signalling by short interfering RNA reduced extracellular mitochondria transfer and worsened neurological outcomes. These findings suggest a new mitochondrial mechanism of neuroglial crosstalk that may contribute to endogenous neuroprotective and neurorecovery mechanisms after stroke.


Asunto(s)
Astrocitos/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Neuronas/patología , Accidente Cerebrovascular/patología , ADP-Ribosil Ciclasa 1/deficiencia , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Astrocitos/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Calcio/metabolismo , Supervivencia Celular , ADP-Ribosa Cíclica/metabolismo , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal , Neuronas/metabolismo , Factores Protectores , ARN Interferente Pequeño/genética , Transducción de Señal , Estrés Fisiológico , Accidente Cerebrovascular/metabolismo
6.
Stroke ; 52(6): 2180-2190, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33940951

RESUMEN

Circadian biology modulates almost all aspects of mammalian physiology, disease, and response to therapies. Emerging data suggest that circadian biology may significantly affect the mechanisms of susceptibility, injury, recovery, and the response to therapy in stroke. In this review/perspective, we survey the accumulating literature and attempt to connect molecular, cellular, and physiological pathways in circadian biology to clinical consequences in stroke. Accounting for the complex and multifactorial effects of circadian rhythm may improve translational opportunities for stroke diagnostics and therapeutics.


Asunto(s)
Ritmo Circadiano/fisiología , Mediadores de Inflamación/fisiología , Acoplamiento Neurovascular/fisiología , Accidente Cerebrovascular/fisiopatología , Accidente Cerebrovascular/terapia , Animales , Ensayos Clínicos como Asunto/métodos , Humanos , Accidente Cerebrovascular/diagnóstico
7.
Stroke ; 51(9): 2844-2853, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32772683

RESUMEN

BACKGROUND AND PURPOSE: Although VEGF165 (vascular endothelial growth factor-165) is able to enhance both angiogenesis and neurogenesis, it also increases vascular permeability through the blood-brain barrier. Heparan sulfate (HS) sugars play important roles in regulating VEGF bioactivity in the pericellular compartment. Here we asked whether an affinity-purified VEGF165-binding HS (HS7) could augment endogenous VEGF activity during stroke recovery without affecting blood-brain barrier function. METHODS: Both rat brain endothelial cell line 4 and primary rat neural progenitor cells were used to evaluate the potential angiogenic and neurogenic effects of HS7 in vitro. For in vivo experiments, male Sprague-Dawley rats were subjected to 100 minutes of transient focal cerebral ischemia, then treated after 4 days with either PBS or HS7. One week later, infarct volume, behavioral sequelae, immunohistochemical markers of angiogenesis and neural stem cell proliferation were assessed. RESULTS: HS7 significantly enhanced VEGF165-mediated angiogenesis in rat brain endothelial cell line 4 brain endothelial cells, and increased the proliferation and differentiation of primary neural progenitor cells, both via the VEGFR2 (vascular endothelial growth factor receptor 2) pathway. Intracerebroventricular injection of HS7 improved neurological outcome in ischemic rats without changing infarct volumes. Immunostaining of the compromised cerebrum demonstrated increases in collagen IV/Ki67 and nestin/Ki67 after HS7 exposure, consistent with its ability to promote angiogenesis and neurogenesis, without compromising blood-brain barrier integrity. CONCLUSIONS: A VEGF-activating glycosaminoglycan sugar, by itself, is able to enhance endogenous VEGF165 activity during the post-ischemic recovery phase of stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Heparitina Sulfato/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Heparitina Sulfato/administración & dosificación , Infarto de la Arteria Cerebral Media/prevención & control , Inyecciones Intraventriculares , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/fisiopatología , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Stroke ; 49(6): 1534-1537, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29712880

RESUMEN

BACKGROUND AND PURPOSE: The efficacy of neuroprotective approaches in stroke may be influenced by existing comorbidities. Here, we compared the effects of normobaric hyperoxia (NBO) in normotensive versus hypertensive rats subjected to transient focal cerebral ischemia. METHODS: Male Sprague-Dawley and spontaneously hypertensive rats were subjected to transient focal ischemia via intraluminal filament occlusions of the middle cerebral artery. NBO was started 15 minutes after ischemic onset and stopped at the time of reperfusion. Acute neurological deficits and tetrazolium-stained infarct volumes were quantified at 24 hours. RESULTS: NBO reduced mean infarct volumes by ≈50% (P=0.0064) in normotensive Sprague-Dawley rats subjected to 100 minutes transient ischemia. No effects of NBO were observed in hypertensive spontaneously hypertensive rats subjected to either 100 minutes or 75 minutes of transient ischemia. No significant changes in neurological outcomes were detectable in any group. CONCLUSIONS: NBO reduced infarction in Sprague-Dawley but not in spontaneously hypertensive rats. These findings suggest that comorbidities may influence responses to potential treatments after stroke.


Asunto(s)
Isquemia Encefálica/terapia , Ataque Isquémico Transitorio/terapia , Oxígeno/farmacología , Accidente Cerebrovascular/terapia , Animales , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/terapia , Masculino , Arteria Cerebral Media/fisiopatología , Oxígeno/metabolismo , Ratas Sprague-Dawley
9.
J Neurochem ; 146(2): 160-172, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29570780

RESUMEN

Ischemic postconditioning is increasingly being investigated as a therapeutic approach for cerebral ischemia. However, the majority of studies are focused on the acute protection of neurons per se. Whether and how postconditioning affects multiple cells in the recovering neurovascular unit remains to be fully elucidated. Here, we asked whether postconditioning may modulate help-me signaling between injured neurons and reactive microglia. Rats were subjected to 100 min of focal cerebral ischemia, then randomized into a control versus postconditioning group. After 3 days of reperfusion, infarct volumes were significantly reduced in animals treated with postconditioning, along with better neurologic outcomes. Immunostaining revealed that ischemic postconditioning increased expression of vascular endothelial growth factor (VEGF) in neurons within peri-infarct regions. Correspondingly, we confirmed that VEGFR2 was expressed on Iba1-positive microglia/macrophages, and confocal microscopy showed that in postconditioned rats, these cells were polarized to a ramified morphology with higher expression of M2-like markers. Treating rats with a VEGF receptor 2 kinase inhibitor negated these effects of postconditioning on microglia/macrophage polarization. In vitro, postconditoning after oxygen-glucose deprivation up-regulated VEGF release in primary neuron cultures, and adding VEGF to microglial cultures partly shifted their M2-like markers. Altogether, our findings support the idea that after postconditioning, injured neurons may release VEGF as a 'help-me' signal that promotes microglia/macrophage polarization into potentially beneficial phenotypes.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Polaridad Celular/fisiología , Poscondicionamiento Isquémico/métodos , Microglía/patología , Neuronas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Infarto Encefálico/etiología , Proteínas de Unión al Calcio/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosa/deficiencia , Infusiones Intraventriculares , Masculino , Proteínas de Microfilamentos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
Stroke ; 48(8): 2231-2237, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28663512

RESUMEN

BACKGROUND AND PURPOSE: Recent studies suggest that extracellular mitochondria may be involved in the pathophysiology of stroke. In this study, we assessed the functional relevance of endogenous extracellular mitochondria in cerebrospinal fluid (CSF) in rats and humans after subarachnoid hemorrhage (SAH). METHODS: A standard rat model of SAH was used, where an intraluminal suture was used to perforate a cerebral artery, thus leading to blood extravasation into subarachnoid space. At 24 and 72 hours after SAH, neurological outcomes were measured, and the standard JC1 (5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-benzimidazolylcarbocyanineiodide) assay was used to quantify mitochondrial membrane potentials in the CSF. To further support the rat model experiments, CSF samples were obtained from 41 patients with SAH and 27 control subjects. Mitochondrial membrane potentials were measured with the JC1 assay, and correlations with clinical outcomes were assessed at 3 months. RESULTS: In the standard rat model of SAH, extracellular mitochondria was detected in CSF at 24 and 72 hours after injury. JC1 assays demonstrated that mitochondrial membrane potentials in CSF were decreased after SAH compared with sham-operated controls. In human CSF samples, extracellular mitochondria were also detected, and JC1 levels were also reduced after SAH. Furthermore, higher mitochondrial membrane potentials in the CSF were correlated with good clinical recovery at 3 months after SAH onset. CONCLUSIONS: This proof-of-concept study suggests that extracellular mitochondria may provide a biomarker-like glimpse into brain integrity and recovery after injury.


Asunto(s)
Líquido Extracelular/metabolismo , Mitocondrias/metabolismo , Recuperación de la Función/fisiología , Hemorragia Subaracnoidea/líquido cefalorraquídeo , Animales , Biomarcadores/líquido cefalorraquídeo , Humanos , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/ultraestructura , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/patología
12.
Stroke ; 46(9): 2691-4, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26243221

RESUMEN

BACKGROUND AND PURPOSE: Postconditioning may be a clinically feasible way to protect the brain after a stroke. However, its effects during the recovery phase post stroke remain to be fully elucidated. Here, we examine the hypothesis that ischemic postconditioning amplifies neurogenesis and angiogenesis during stroke recovery. METHODS: Male Sprague-Dawley rats were subjected to 100-minute transient middle cerebral artery occlusion (MCAO) or postconditioning (100-minute middle cerebral artery occlusion plus 10-minute reperfusion plus 10-minute reocclusion). After 2 weeks, infarct volumes, behavioral outcomes, and immunohistochemical markers of neurogenesis and angiogenesis were quantified. RESULTS: Postconditioning significantly reduced infarction and improved neurological outcomes. Concomitantly, brains subjected to postconditioning showed an increase in doublecortin/BrdU and collagen-IV/Ki67-positive cells. CONCLUSIONS: These results suggest that therapeutic effects of postconditioning may involve the promotion of neurogenesis and angiogenic remodeling during the recovery phase after focal cerebral ischemia.


Asunto(s)
Infarto de la Arteria Cerebral Media/terapia , Poscondicionamiento Isquémico/métodos , Neovascularización Fisiológica/fisiología , Neurogénesis/fisiología , Recuperación de la Función/fisiología , Reperfusión/métodos , Animales , Proteína Doblecortina , Masculino , Ratas , Ratas Sprague-Dawley
13.
Commun Biol ; 7(1): 229, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38402351

RESUMEN

Crosstalk between central nervous system (CNS) and systemic responses is important in many pathological conditions, including stroke, neurodegeneration, schizophrenia, epilepsy, etc. Accumulating evidence suggest that signals for central-systemic crosstalk may utilize glymphatic and lymphatic pathways. The glymphatic system is functionally connected to the meningeal lymphatic system, and together these pathways may be involved in the distribution of soluble proteins and clearance of metabolites and waste products from the CNS. Lymphatic vessels in the dura and meninges transport cerebrospinal fluid, in part collected from the glymphatic system, to the cervical lymph nodes, where solutes coming from the brain (i.e., VEGFC, oligomeric α-syn, ß-amyloid) might activate a systemic inflammatory response. There is also an element of time since the immune system is strongly regulated by circadian rhythms, and both glymphatic and lymphatic dynamics have been shown to change during the day and night. Understanding the mechanisms regulating the brain-cervical lymph node (CLN) signaling and how it might be affected by diurnal or circadian rhythms is fundamental to find specific targets and timing for therapeutic interventions.


Asunto(s)
Sistema Nervioso Central , Vasos Linfáticos , Vasos Linfáticos/fisiología , Encéfalo/metabolismo , Sistema Linfático , Meninges
14.
Neurobiol Dis ; 54: 105-14, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23454199

RESUMEN

It has recently been hypothesized that a sub-lethal ischemic insult induced in one organ is able to protect from a harmful ischemia occurring in a different organ. The objective of this study is to identify new putative mechanisms of neuroprotection elicited by remote ischemic femoral postconditioning. A 50% reduction in the infarct volume was observed when 100min of middle cerebral artery occlusion was followed, 10min later, by the remote postconditioning stimulus represented by 20min of femoral artery occlusion. The use of in vivo silencing strategy allowed to demonstrate that NO production through nNOS mediates part of the neuroprotection. Indeed, whereas CNS nNOS expression was up-regulated by remote postconditioning, the pharmacological inhibition of nNOS or its silencing-mediated knocking-down partially prevented this neuroprotective effect. This nNOS overexpression seemed to be p-ERK dependent. In fact, p-ERK expression increased in brain cortex after remote postconditioning, and its pharmacological inhibition prevented both nNOS overexpression and remote postconditioning-mediated neuroprotection. Interestingly, neuroprotection induced by remote postconditioning was partially prevented when ganglion transmission was pharmacologically interrupted by hexamethonium, thus showing that neural factors are involved in this phenomenon. Collectively, the present study demonstrates that p-ERK and nNOS take part to the complex cascade of events triggered by ischemic remote postconditioning.


Asunto(s)
Encéfalo/irrigación sanguínea , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Poscondicionamiento Isquémico , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Western Blotting , Encéfalo/fisiopatología , Arteria Femoral , Técnicas de Silenciamiento del Gen , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley
15.
BMC Neurosci ; 14: 129, 2013 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-24138708

RESUMEN

BACKGROUND: Intracerebral hemorrhage is a subtype of stroke that has a poor prognosis without an adequate therapy. Recently, the use of anesthetics such as isoflurane has been shown to be protective after cerebral ischemia. However, the potential therapeutic effect of isoflurane after intracerebral hemorrhage (ICH) has not been fully explored. RESULTS: In this study, male Sprague-Dawley rats (SD) were subjected to ICH and randomized into controls and 1.2% or 1.5% isoflurane posttreatment groups. Brain water content, neurological outcomes and matrix metalloproteinase-2 and -9 (MMP2-MMP9) plasma levels were quantified at 24 hours. Isoflurane treatment did not reduce brain edema compared with controls in any of the applied isoflurane concentrations. Moreover, consistent with this lack of effect on brain edema, isoflurane posttreatment did not affect neurological outcomes in any of the tests used. Plasma MMP levels did not change. CONCLUSION: Our data suggested that there is no neuroprotection after isoflurane posttreatment in a rat model of ICH.


Asunto(s)
Encéfalo/efectos de los fármacos , Hemorragia Cerebral/patología , Isoflurano/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Anestésicos por Inhalación/administración & dosificación , Animales , Encéfalo/metabolismo , Encéfalo/patología , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Masculino , Metaloproteinasa 2 de la Matriz/análisis , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos
16.
Adv Exp Med Biol ; 961: 223-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23224883

RESUMEN

Ischemic preconditioning is a neuroprotective mechanism in which a brief non-injurious episode of ischemia protects the brain from a subsequent lethal insult. Recently, it has been reported that modified reperfusion subsequent to a prolonged ischemic episode may also confer neuroprotection, a phenomenon termed postconditioning. Mitogen-activated protein kinases (MAPK) play a key role in these two neuroprotective mechanisms. The aim of this study was to evaluate whether Na(+)/Ca(2+) exchangers (NCXs), a family of ionic transporters that contribute to the maintenance of intracellular ionic homeostasis, contribute to the neuroprotection elicited by ischemic preconditioning and postconditioning.Results of this study indicated that (1) NCX1 and NCX3 are upregulated in those brain regions protected by preconditioning, while (2) postconditioning treatment induces an upregulation only in NCX3 expression. (3) NCX1 upregulation and NCX3 upregulation are mediated by p-AKT since its inhibition reverted the neuroprotective effect of preconditioning and postconditioning and prevented NCXs overexpression. (4) The involvement of NCX in preconditioning and postconditioning neuroprotection is further supported by the results of experiments showing that a partial reversion of the protective effect induced by preconditioning was obtained by silencing NCX1 or NCX3, while the silencing of NCX3 was able to mitigate the protection induced by ischemic postconditioning.Altogether, the data presented here suggest that NCX1 and NCX3 -represent two promising druggable targets for setting on new strategies in stroke therapy.


Asunto(s)
Encéfalo/metabolismo , Precondicionamiento Isquémico , Proteínas del Tejido Nervioso/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Encéfalo/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Intercambiador de Sodio-Calcio/genética , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología
17.
Adv Exp Med Biol ; 961: 137-45, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23224876

RESUMEN

The ubiquitous sodium-calcium exchanger isoform 1 (NCX1) is a -bidirectional transporter that plays a relevant role under physiological and pathophysiological conditions including brain ischemia by regulating intraneuronal Ca(2+) and Na(+) homeostasis. Although changes in ncx1 protein and transcript expression have been detected during stroke, its transcriptional regulation is still largely unexplored. Here, we reviewed our recent findings on several transcription factors including cAMP response element-binding protein (CREB), nuclear factor kappa B (NF-κB), and hypoxia-inducible factor-1 (HIF-1) in the control of the ncx1 gene expression in neuronal cells.


Asunto(s)
Isquemia Encefálica/metabolismo , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Intercambiador de Sodio-Calcio/biosíntesis , Accidente Cerebrovascular/metabolismo , Transcripción Genética , Animales , Encéfalo , Isquemia Encefálica/patología , Calcio/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Neuronas/patología , Sodio/metabolismo , Accidente Cerebrovascular/patología
18.
Front Cell Neurosci ; 17: 1260389, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37744881

RESUMEN

Background and purpose: Experimental studies suggest that ischemic postconditioning interferes with cell death mechanisms and reduces infarction during the acute phase after focal cerebral ischemia. Postconditioning may be a practically feasible way to promote stroke recovery, but many drawbacks prevent its clinical translation. First, all existing studies are mostly on acute 24 h outcomes. Second, the mechanisms of protection and augmented long-term benefits remain unclear. Our study aims to define some of the mechanisms that explain long-term benefits of improved recovery. Methods: Male Sprague-Dawley rats were subjected to 100-min transient middle cerebral artery occlusion (MCAO) or postconditioning (100-min middle cerebral artery occlusion plus 10-min reperfusion plus 10-min reocclusion). After 3 days or 2 weeks, infarct volumes, western blot, and immunohistochemical markers of neurogenesis and angiogenesis were quantified. Fluorocitrate (FC) or saline were administrated ICV (intraventricular injection) every other day starting on day 5 after focal cerebral ischemia, animals were recovered for 2 weeks. Results: After postconditioning BDNF protein expression levels increased compared to animals subjected to MCAO. Immunostaining showed that BDNF increased specifically in astrocytes. Moreover, when astrocytes were metabolically inhibited by fluorocitrate the postconditioning neuroprotective effect together with the postconditioning-dependent new angiogenesis and neurogenesis, were no longer observed. Conclusion: These results suggest for the first time that therapeutic effects of postconditioning may involve the promotion of neurogenesis and angiogenic remodeling, via BDNF released by astrocytes, during the recovery phase after focal cerebral ischemia.

19.
Commun Med (Lond) ; 3(1): 169, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38007588

RESUMEN

BACKGROUND: Transplantation of mitochondria is increasingly explored as a novel therapy in central nervous system (CNS) injury and disease. However, there are limitations in safety and efficacy because mitochondria are vulnerable in extracellular environments and damaged mitochondria can induce unfavorable danger signals. METHODS: Mitochondrial O-GlcNAc-modification was amplified by recombinant O-GlcNAc transferase (OGT) and UDP-GlcNAc. O-GlcNAcylated mitochondrial proteins were identified by mass spectrometry and the antiglycation ability of O-GlcNAcylated DJ1 was determined by loss-of-function via mutagenesis. Therapeutic efficacy of O-GlcNAcylated mitochondria was assessed in a mouse model of transient focal cerebral ischemia-reperfusion. To explore translational potential, we evaluated O-GlcNAcylated DJ1 in CSF collected from patients with subarachnoid hemorrhagic stroke (SAH). RESULTS: We show that isolated mitochondria are susceptible to advanced glycation end product (AGE) modification, and these glycated mitochondria induce the receptor for advanced glycation end product (RAGE)-mediated autophagy and oxidative stress when transferred into neurons. However, modifying mitochondria with O-GlcNAcylation counteracts glycation, diminishes RAGE-mediated effects, and improves viability of mitochondria recipient neurons. In a mouse model of stroke, treatment with extracellular mitochondria modified by O-GlcNAcylation reduces neuronal injury and improves neurologic deficits. In cerebrospinal fluid (CSF) samples from SAH patients, levels of O-GlcNAcylation in extracellular mitochondria correlate with better clinical outcomes. CONCLUSIONS: These findings suggest that AGE-modification in extracellular mitochondria may induce danger signals, but O-GlcNAcylation can prevent glycation and improve the therapeutic efficacy of transplanted mitochondria in the CNS.


Mitochondria are the part of a cell that generate most of its energy to perform its functions. In injury or disease, mitochondrial function can become disrupted. Transplantation of healthy mitochondria is being explored as a potential therapy to replace damaged mitochondria and restore normal cellular function. However, this approach is difficult to perform because mitochondria are not able to maintain their healthy state outside of cells. Here, we show that one of the reasons for this is due to a molecular process called advanced glycation end product modification. We show that simple modification of mitochondria with a sugar prevents this process and helps to improve the success of therapeutic mitochondrial transplantation in cells and in a mouse model of stroke. Our findings may help to guide future efforts to develop therapies based on mitochondrial transplantation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA