Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Stroke ; 53(3): 904-912, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732071

RESUMEN

BACKGROUND: Inhalational anesthetics were associated with reduced incidence of angiographic vasospasm and delayed cerebral ischemia (DCI) in patients with aneurysmal subarachnoid hemorrhage (SAH). Whether intravenous anesthetics provide similar level of protection is not known. METHODS: Anesthetic data were collected retrospectively for patients with SAH who received general anesthesia for aneurysm repair between January 1, 2014 and May 31, 2018, at 2 academic centers in the United States (one employing primarily inhalational and the other primarily intravenous anesthesia with propofol). We compared the outcomes of angiographic vasospasm, DCI, and neurological outcome (measured by disposition at hospital discharge), between the 2 sites, adjusting for potential confounders. RESULTS: We compared 179 patients with SAH receiving inhalational anesthetics at one institution to 206 patients with SAH receiving intravenous anesthetics at the second institution. The rates of angiographic vasospasm between inhalational versus intravenous anesthetic groups were 32% versus 52% (odds ratio, 0.49 [CI, 0.32-0.75]; P=0.001) and DCI were 21% versus 40% (odds ratio, 0.47 [CI, 0.29-0.74]; P=0.001), adjusting for imbalances between sites/groups, Hunt-Hess and Fisher grades, type of aneurysm treatment, and American Society of Anesthesiology status. No impact of anesthetics on neurological outcome at time of discharge was noted with rates of good discharge outcome between inhalational versus intravenous anesthetic groups at (78% versus 72%, P=0.23). CONCLUSIONS: Our data suggest that those who received inhalational versus intravenous anesthetic for ruptured aneurysm repair had significant protection against SAH-induced angiographic vasospasm and DCI. Although we cannot fully disentangle site-specific versus anesthetic effects in this comparative study, these results, when coupled with preclinical data demonstrating a similar protective effect of inhalational anesthetics on vasospasm and DCI, suggest that inhalational anesthetics may be preferable for patients with SAH undergoing aneurysm repair. Additional investigations examining the effect of inhalational anesthetics on other SAH outcomes such as early brain injury and long-term neurological outcomes are warranted.


Asunto(s)
Anestésicos Intravenosos/uso terapéutico , Isquemia Encefálica/prevención & control , Propofol/uso terapéutico , Hemorragia Subaracnoidea/complicaciones , Adulto , Anciano , Anestésicos Intravenosos/administración & dosificación , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/etiología , Angiografía Cerebral , Femenino , Humanos , Masculino , Persona de Mediana Edad , Propofol/administración & dosificación , Estudios Retrospectivos , Hemorragia Subaracnoidea/diagnóstico por imagen
2.
Physiol Genomics ; 52(1): 20-34, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31762411

RESUMEN

Recent evidence from our laboratory documents functional resilience to retinal ischemic injury in untreated mice derived from parents exposed to repetitive hypoxic conditioning (RHC) before breeding. To begin to understand the epigenetic basis of this intergenerational protection, we used methylated DNA immunoprecipitation and sequencing to identify genes with differentially methylated promoters (DMGPs) in the prefrontal cortex of mice treated directly with the same RHC stimulus (F0-RHC) and in the prefrontal cortex of their untreated F1-generation offspring (F1-*RHC). Subsequent bioinformatic analyses provided key mechanistic insights into how changes in gene expression secondary to promoter hypo- and hypermethylation might afford such protection within and across generations. We found extensive changes in DNA methylation in both generations consistent with the expression of many survival-promoting genes, with twice the number of DMGPs in the cortex of F1*RHC mice relative to their F0 parents that were directly exposed to RHC. In contrast to our hypothesis that similar epigenetic modifications would be realized in the cortices of both F0-RHC and F1-*RHC mice, we instead found relatively few DMGPs common to both generations; in fact, each generation manifested expected injury resilience via distinctly unique gene expression profiles. Whereas in the cortex of F0-RHC mice, predicted protein-protein interactions reflected activation of an anti-ischemic phenotype, networks activated in F1-*RHC cortex comprised networks indicative of a much broader cytoprotective phenotype. Altogether, our results suggest that the intergenerational transfer of an acquired phenotype to offspring does not necessarily require the faithful recapitulation of the conditioning-modified DNA methylome of the parent.


Asunto(s)
Metilación de ADN/genética , Hipoxia/genética , Animales , Región CA1 Hipocampal/patología , Supervivencia Celular , Femenino , Redes Reguladoras de Genes , Masculino , Ratones , Linaje , Regiones Promotoras Genéticas , Células Piramidales/patología , Transducción de Señal/genética
3.
Exp Brain Res ; 237(6): 1493-1502, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30915491

RESUMEN

Remote limb ischemic conditioning (RLIC) is a technique in which tissues distant from the target organ are exposed to brief, sub-lethal bouts of ischemia. The effects of remotely applied ischemic conditioning are systemically transferred to the target organ, and typically manifested as protection from subsequent ischemic injury. Previous studies in our lab have found and confirmed that RLIC enhances learning and retention during motor training on a balance task. The current study tested the effect of RLIC dose (number of cycles) on learning enhancement in young, healthy adults. Forty healthy participants age 18-40 years were randomized to receive 5 cycles of sham conditioning (n = 9), 3 cycles of RLIC (n = 11), 4 cycles of RLIC (n = 10), or 5 cycles of RLIC (n = 10) using a blood pressure cuff around the upper arm once a day for 7 consecutive weekdays (Days 1-7). Participants concurrently trained on a balance task, bimanual cup stacking task, and a discrete sequence production task on Days 3-7. Change in performance on each of the three tasks was compared across groups. Participants in all four groups improved their performance on each of the three tasks over time. However, RLIC at any dose did not enhance learning on any of the three tasks. While RLIC is safe, inexpensive, and clinically feasible, reproducibility may be challenged by unidentified factors, raising critical challenges to the straightforward translation of RLIC for improving rehabilitation outcomes in individuals recovering from neurological injury.


Asunto(s)
Brazo/irrigación sanguínea , Aprendizaje/fisiología , Equilibrio Postural/fisiología , Desempeño Psicomotor/fisiología , Flujo Sanguíneo Regional/fisiología , Adolescente , Adulto , Femenino , Humanos , Precondicionamiento Isquémico , Masculino , Placebos , Adulto Joven
4.
Mol Vis ; 24: 875-889, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30713425

RESUMEN

Purpose: Diverse groups of proteins play integral roles in both the physiology and pathophysiology of the retina. However, thorough proteomic analyses of retinas of experimental species are currently unavailable. The purpose of the present paper is providing the field with a comprehensive proteomic characterization of the retina of a commonly used laboratory mouse using a discovery-based mass spectrometry (MS) approach. Methods: Retinas from eight male and eight female 30-week-old outbred ND4 Swiss Webster mice were harvested and immediately processed for MS analysis on a Thermo Fisher (TF) Fusion Orbitrap MS. The retinal proteome and phosphoproteome were identified and subsequently analyzed using Proteome Discoverer 2.2 and Panther-GeneGo. SEQUEST-HT scoring was used for analysis, and the reference protein FASTA database was from Mus musculus. Specifically, three technical repeats were performed for each biological sample. For characterization, only high-scoring peptides were considered, with a false discovery rate (FDR) of <1%. Downstream bioinformatic analysis used Ingenuity Pathway Analysis (IPA; Qiagen). Results: Using Proteome Discoverer 2.2, 4,767 different proteins were identified and segregated into 26 major protein classes, 9 functional molecular classes, and 12 categories of biological processes. The five largest protein classes included the following: nucleic acid binding (17%), hydrolases (13%), enzyme modulators (10%), transferases (9%), and oxidoreductases (6%). "Binding" and "catalytic" proteins contributed to 81% of the molecular function class at 37% and 42%, respectively. "Cellular processing" and "metabolic processes" contributed the most to biologic activity, at 31% and 26%, respectively. Phosphopeptide enrichment yielded the identification of 610 additional unique proteins that were not originally identified. The two datasets combined produced an adult mouse retinal proteome consisting of 5,377 unique proteins. Overall, 41% of the retinal proteome was phosphorylated. The overwhelming diversity of retinal protein functionality was reflected through further analyses revealing 2,086 unique pathway hits across 241 different pathways (TF). A core analysis summary report was performed in IPA (Qiagen) to analyze the top signaling networks, protein-protein interaction (PPI) enrichments, and canonical pathways. Conclusions: Using this high-throughput technique, we have further deciphered and updated the diverse proteome of the mouse retina, including the phosphoproteome, thereby providing the most comprehensive proteomic profile for this tissue known to date. These findings, and the bioinformatic analyses we also provided, establish a platform for future studies, facilitating the elucidation of the relevance of these proteins to the molecular and cellular pathologies that underlie retinal function and disease.


Asunto(s)
Redes Reguladoras de Genes , Fosfoproteínas/genética , Proteoma/genética , Retina/metabolismo , Animales , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Masculino , Espectrometría de Masas/métodos , Redes y Vías Metabólicas/genética , Ratones , Anotación de Secuencia Molecular , Fosfoproteínas/clasificación , Fosfoproteínas/aislamiento & purificación , Fosfoproteínas/metabolismo , Proteoma/clasificación , Proteoma/aislamiento & purificación , Proteoma/metabolismo , Retina/química
5.
Mol Vis ; 19: 2360-72, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24319330

RESUMEN

PURPOSE: We recently demonstrated in a mouse model of glaucoma that endogenous epigenetic mechanisms can be activated by a repetitive hypoxic preconditioning (RHP) stimulus to provide robust retinal ganglion cell (RGC) protection. Although we also provided evidence that RHP prevents or delays the apoptotic demise of the RGC soma, the mechanisms responsible for signaling this epigenetic response, as well as the effectors of the glaucoma-tolerant phenotype at the somatic and axonal levels, remain unidentified. In the present study, we used conditional mutant mice lacking hypoxia-inducible factor-1α (HIF-1α) in RGCs (HIF-1α RGC-knockout [KO] mice) to test the hypothesis that RHP-mediated activation of this transcription factor in these cells protects them from glaucomatous injury. METHODS: Adult HIF-1α RGC-KO mice, generated by mating floxed HIF-1α mice with math5-Cre mice, were used. Experimental glaucoma was induced unilaterally in the HIF-1α RGC-KO mice and matched wild-types by elevating the intraocular pressure to 16-20 mmHg for 3 consecutive weeks, secondary to episcleral vein ligation. Mice of each genotype were randomized to either an RHP protocol (six total exposures to systemic hypoxia [11% oxygen], interspersed over a 2-week period, completed 3 days before ligation surgery) or to an untreated group. RGC soma and axon injury was quantified with Neuronal Nuclei (NeuN) immunohistochemistry in retinal flat mounts and SMI32 immunohistochemistry in cross sections of the post-laminar optic nerve, respectively. RESULTS: HIF-1α RGC-KO mice exhibited normal retinal function and morphology, and crosses of math5-Cre mice with floxed ROSA26 reporter mice confirmed Cre recombinase activity was confined to the RGC axons and soma. Untreated wild-type mice exhibited a 30±2% loss of RGC soma and a 31±3% loss of RGC axons after 3 weeks of intraocular hypertension (both p<0.05 versus fellow eye). The 90% and 81% improvement in soma and axon survival, respectively, observed in the wild-type mice treated with RHP (both p<0.05 versus the glaucoma eye in the untreated mice) was still observed to a near identical extent in the RHP-treated HIF-1α RGC-KO mice. RHP had no effect on the magnitude of intraocular pressure elevation in either the KO or wild-type groups, indicating that protection was realized in both genotypes in the face of ongoing intraocular hypertension. CONCLUSIONS: These findings indicate that the robust, glaucomatous protection of the RGC soma and axons induced by RHP does not require HIF-1α-mediated transcription of survival genes and other adaptive responses within the RGCs themselves. Rather, we infer that RGC survival is augmented secondary to the activation of other hypoxia-sensitive transcription factors in RGCs and/or the action of diffusible HIF-1α target gene proteins released from neighboring retinal cells. Ideally, the involvement of such autocrine- and/or paracrine-based mechanisms would be confirmed in future studies, but distinct components of the integrated, pleiotropic, and multicellular basis of this endogenous epigenetic response may prove difficult to demonstrate experimentally, as we found in the present study.


Asunto(s)
Axones/efectos de los fármacos , Glaucoma/tratamiento farmacológico , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hipoxia/genética , Hipertensión Ocular/tratamiento farmacológico , Oxígeno/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Axones/metabolismo , Axones/patología , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Femenino , Glaucoma/genética , Glaucoma/metabolismo , Glaucoma/patología , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Inmunohistoquímica , Presión Intraocular , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Hipertensión Ocular/genética , Hipertensión Ocular/metabolismo , Hipertensión Ocular/patología , Nervio Óptico/efectos de los fármacos , Nervio Óptico/metabolismo , Nervio Óptico/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Vena Retiniana/cirugía , Transducción de Señal , Tonometría Ocular , Transcripción Genética
6.
Behav Neurosci ; 137(3): 178-183, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36862475

RESUMEN

We showed previously in a mouse model of vascular cognitive impairment and dementia involving chronic cerebral hypoperfusion (CCH) that repetitive hypoxic conditioning (RHC) of both parents results in the epigenetic, intergenerational transmission of resilience to recognition memory loss in adult progeny, as assessed by the novel object recognition test. The present study was undertaken in the same model to determine whether RHC treatment of one or both parents is required to confer dementia resilience intergenerationally. We found inherited resilience to 3 months of CCH in males is maternally mediated (p = .006). Statistically, we observed a strong trend for the paternal germline to contribute as well (p = .052). We also found that, in contrast to what is widely observed in males, females display intact recognition memory (p = .001) after 3 months of CCH, revealing a heretofore unidentified sexual dimorphism with respect to cognitive impact during disease progression. Overall, results of our study strongly implicate epigenetic changes in maternal germ cells, induced by our repetitive systemic hypoxic stimulus, contributing to a modified differentiation program capable of establishing a dementia-resilient phenotype in adult male first-generation progeny. (PsycInfo Database Record (c) 2023 APA, all rights reserved).


Asunto(s)
Isquemia Encefálica , Disfunción Cognitiva , Demencia , Ratones , Animales , Femenino , Masculino , Trastornos de la Memoria/genética , Disfunción Cognitiva/genética , Hipoxia , Epigénesis Genética , Modelos Animales de Enfermedad
7.
J Neurochem ; 123(6): 954-62, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23043544

RESUMEN

The induction of ischemic tolerance by preconditioning provides a platform to elucidate endogenous mechanisms of stroke protection. In these studies, we characterize the relationship between hypoxia-inducible factor (HIF), sphingosine kinase 2 (SphK2), and chemokine (C-C motif) ligand 2 (CCL2) in models of hypoxic or pharmacological preconditioning-induced ischemic tolerance. A genetics-based approach using SphK2- and CCL2-null mice showed both SphK2 and CCL2 to be necessary for the induction of ischemic tolerance following preconditioning with hypoxia, the hypoxia-mimetic cobalt chloride, or the sphingosine-1-phosphate (S1P) agonist FTY720. A pharmacological approach confirmed the necessity of HIF signaling for all three preconditioning stimuli, and showed that the SphK/S1P pathway transduces tolerance via the S1P(1) receptor. In addition, our data suggest significant cross-talk between HIF and SphK2-produced S1P signaling, which together act to up-regulate CCL2 expression. Overall, HIF, SphK, S1P, and CCL2 participate in a signaling cascade to induce the gene expression responsible for the stroke-tolerant phenotype established by hypoxic and FTY720 preconditioning. The identification of these common molecular mediators involved in signaling the genomic response to multiple preconditioning stimuli provides several targets for therapeutic manipulation.


Asunto(s)
Quimiocina CCL2/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Hipoxia-Isquemia Encefálica/metabolismo , Precondicionamiento Isquémico/métodos , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Transducción de Señal/fisiología , Accidente Cerebrovascular/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Inhibidores Enzimáticos/farmacología , Femenino , Hipoxia-Isquemia Encefálica/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/enzimología
8.
Mol Med ; 18: 697-706, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22396016

RESUMEN

Like all cells, neurons adapt to stress by transient alterations in phenotype, an epigenetic response that forms the basis for preconditioning against acute ischemic injury in the central nervous system. We recently showed that a modified repetitive hypoxic preconditioning (RHP) regimen significantly extends the window of ischemic tolerance to acute retinal ischemic injury from days to months. The present study was undertaken to determine if this uniquely protracted neuroprotective phenotype would also confer resistance to glaucomatous neurodegeneration. Retinal ganglion cell death at somatic and axonal levels was assessed after both 3 and 10 wks of sustained intraocular hypertension in an adult mouse model of inducible, open-angle glaucoma, with or without RHP before intraocular pressure elevation. Loss of brn3-positive ganglion cell soma after 3 wks of experimental glaucoma, along with increases in several apoptotic endpoints, were all significantly and robustly attenuated in mice subjected to RHP. Soma protection by RHP was also confirmed after 10 wks of intraocular hypertension by brn3 and SMI32 immunostaining. In addition, quantification of axon density in the postlaminar optic nerve documented robust preservation in RHP-treated mice, and neurofilament immunostaining also revealed preconditioning-induced improvements in axon integrity/survival in both retina and optic nerve after 10 wks of experimental glaucoma. This uniquely protracted period of phenotypic change, established in retinal ganglion cells by the activation of latent antiapoptotic, prosurvival mechanisms at both somatic and axonal levels, reflects a novel form of inducible neuronal plasticity that may provide innovative therapeutic targets for preventing and treating glaucoma and other neurodegenerative diseases.


Asunto(s)
Glaucoma/patología , Glaucoma/prevención & control , Células Ganglionares de la Retina/patología , Animales , Apoptosis , Axones/metabolismo , Hipoxia de la Célula , Modelos Animales de Enfermedad , Presión Intraocular , Masculino , Ratones , Ratones Endogámicos C57BL , Células Ganglionares de la Retina/metabolismo
9.
J Neuroinflammation ; 9: 33, 2012 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-22340958

RESUMEN

BACKGROUND: A brief exposure to systemic hypoxia (i.e., hypoxic preconditioning; HPC) prior to transient middle cerebral artery occlusion (tMCAo) reduces infarct volume, blood-brain barrier disruption, and leukocyte migration. CCL2 (MCP-1), typically regarded as a leukocyte-derived pro-inflammatory chemokine, can also be directly upregulated by hypoxia-induced transcription. We hypothesized that such a hypoxia-induced upregulation of CCL2 is required for HPC-induced ischemic tolerance. METHODS: Adult male SW/ND4, CCL2-null, and wild-type mice were used in these studies. Cortical CCL2/CCR2 message, protein, and cell-type specific immunoreactivity were determined following HPC (4 h, 8% O2) or room air control (21% O2) from 6 h through 2 weeks following HPC. Circulating leukocyte subsets were determined by multi-parameter flow cytometry in naïve mice and 12 h after HPC. CCL2-null and wild-type mice were exposed to HPC 2 days prior to tMCAo, with immunoneutralization of CCL2 during HPC achieved by a monoclonal CCL2 antibody. RESULTS: Cortical CCL2 mRNA and protein expression peaked at 12 h after HPC (both p < 0.01), predominantly in cortical neurons, and returned to baseline by 2 days. A delayed cerebral endothelial CCL2 message expression (p < 0.05) occurred 2 days after HPC. The levels of circulating monocytes (p < 0.0001), T lymphocytes (p < 0.0001), and granulocytes were decreased 12 h after HPC, and those of B lymphocytes were increased (p < 0.0001), but the magnitude of these respective changes did not differ between wild-type and CCL2-null mice. HPC did decrease the number of circulating CCR2+ monocytes (p < 0.0001) in a CCL2-dependent manner, but immunohistochemical analyses at this 12 h timepoint indicated that this leukocyte subpopulation did not move into the CNS. While HPC reduced infarct volumes by 27% (p < 0.01) in wild-type mice, CCL2-null mice subjected to tMCAo were not protected by HPC. Moreover, administration of a CCL2 immunoneutralizing antibody prior to HPC completely blocked (p < 0.0001 vs. HPC-treated mice) the development of ischemic tolerance. CONCLUSIONS: The early expression of CCL2 in neurons, the delayed expression of CCL2 in cerebral endothelial cells, and CCL2-mediated actions on circulating CCR2+ monocytes, appear to be required to establish ischemic tolerance to focal stroke in response to HPC, and thus represent a novel role for this chemokine in endogenous neurovascular protection.


Asunto(s)
Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Quimiocina CCL2/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Precondicionamiento Isquémico/métodos , Regulación hacia Arriba/fisiología , Animales , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunoglobulina G/uso terapéutico , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/fisiología , NADH Deshidrogenasa/genética , NADH Deshidrogenasa/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/genética , Receptores CCR2/genética , Receptores CCR2/metabolismo , Factores de Tiempo
10.
Ann Neurol ; 69(6): 975-85, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21437933

RESUMEN

OBJECTIVE: Brief systemic hypoxia protects the rodent brain from subsequent ischemic injury, although the protection wanes within days. We hypothesized that the duration of ischemic tolerance could be extended from days to months by repeated intermittent hypoxia of varying magnitude and duration. METHODS: Infarction volumes following a 60-minute transient middle cerebral artery occlusion were determined in adult male mice 2 days through 8 weeks after completion of a 2-week repetitive hypoxic preconditioning (RHP) protocol. Separate cohorts were studied for the protective effects of RHP on postischemic and cytokine-induced cerebrovascular inflammation, and for potential deleterious effects of the RHP stimulus itself. RESULTS: RHP protection against transient focal stroke persisted for 8 weeks. Leukocyte adherence to cortical venules was attenuated in response to stroke, as well as following tumor necrosis factor-α administration, indicating that reductions in postischemic inflammation were not secondary to smaller infarct volumes. RHP reduced poststroke leukocyte diapedesis concomitant with a long-lasting downregulation of endothelial adhesion molecule mRNAs, and also reduced postischemic blood--brain barrier permeability to endogenous immunoglobulin G. RHP was without effect on hippocampal CA1 pyramidal cell viability, only transiently elevated hematocrit, and did not affect the magnitude of cerebral blood flow during and after ischemia. INTERPRETATION: Taken together, our findings reveal a novel form of epigenetic neurovascular plasticity characterized by a prominent anti-inflammatory phenotype that provides protection against stroke many weeks longer than previously established windows of preconditioning-induced tolerance. Translating these endogenous protective mechanisms into therapeutics could afford sustained periods of cerebroprotection in subpopulations of individuals at identified risk for stroke.


Asunto(s)
Circulación Cerebrovascular/fisiología , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/prevención & control , Precondicionamiento Isquémico , Análisis de Varianza , Animales , Barrera Hematoencefálica/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Selectina E/genética , Selectina E/metabolismo , Lateralidad Funcional , Proteínas Fluorescentes Verdes/genética , Hematócrito/métodos , Hipocampo/metabolismo , Hipocampo/patología , Infarto de la Arteria Cerebral Media/patología , Selectina L/genética , Selectina L/metabolismo , Leucocitos/patología , Masculino , Ratones , Ratones Transgénicos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/efectos adversos , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
Stroke ; 42(3): 776-82, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21317271

RESUMEN

BACKGROUND AND PURPOSE: Vasospasm-induced delayed cerebral ischemia remains a major source of morbidity in patients with aneurysmal subarachnoid hemorrhage (SAH). We hypothesized that activating innate neurovascular protective mechanisms by preconditioning (PC) may represent a novel therapeutic approach against SAH-induced vasospasm and neurological deficits and, secondarily, that the neurovascular protection it provides is mediated by endothelial nitric oxide synthase (eNOS). METHODS: Wild-type mice were subjected to hypoxic PC or normoxia followed 24 hours later by SAH. Neurological function was analyzed daily; vasospasm was assessed on post-surgery Day 2. Nitric oxide availability, eNOS expression, and eNOS activity were also assessed. In a separate experiment, wild-type and eNOS-null mice were subjected to hypoxic PC or normoxia followed by SAH and assessed for vasospasm and neurological deficits. RESULTS: PC nearly completely prevented SAH-induced vasospasm and neurological deficits. It also prevented SAH-induced reduction in nitric oxide availability and increased eNOS activity in mice with and without SAH. PC-induced protection against vasospasm and neurological deficits was lost in wild-type mice treated with the nitric oxide synthase inhibitor N(G)-nitro-l-arginine methyl ester and in eNOS-null mice. CONCLUSIONS: Endogenous protective mechanisms against vasospasm exist, are powerful, and can be induced by PC. eNOS-derived nitric oxide is a critical mediator of PC-induced neurovascular protection. These data provide strong "proof-of-principle" evidence that PC represents a promising new strategy to reduce vasospasm and delayed cerebral ischemia after SAH.


Asunto(s)
Precondicionamiento Isquémico/métodos , Óxido Nítrico Sintasa de Tipo III/fisiología , Hemorragia Subaracnoidea/enzimología , Hemorragia Subaracnoidea/prevención & control , Vasoespasmo Intracraneal/enzimología , Vasoespasmo Intracraneal/prevención & control , Animales , Conducta Animal/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Hemorragia Subaracnoidea/fisiopatología , Vasoespasmo Intracraneal/fisiopatología
12.
J Cereb Blood Flow Metab ; 41(2): 397-412, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32241204

RESUMEN

Sex differences in mitochondrial numbers and function are present in large cerebral arteries, but it is unclear whether these differences extend to the microcirculation. We performed an assessment of mitochondria-related proteins in cerebral microvessels (MVs) isolated from young, male and female, Sprague-Dawley rats. MVs composed of arterioles, capillaries, and venules were isolated from the cerebrum and used to perform a 3 versus 3 quantitative, multiplexed proteomics experiment utilizing tandem mass tags (TMT), coupled with liquid chromatography/mass spectrometry (LC/MS). MS data and bioinformatic analyses were performed using Proteome Discoverer version 2.2 and Ingenuity Pathway Analysis. We identified a total of 1969 proteins, of which 1871 were quantified by TMT labels. Sixty-four proteins were expressed significantly (p < 0.05) higher in female samples compared with male samples. Females expressed more mitochondrial proteins involved in energy production, mitochondrial membrane structure, anti-oxidant enzyme proteins, and those involved in fatty acid oxidation. Conversely, males had higher expression levels of mitochondria-destructive proteins. Our findings reveal, for the first time, the full extent of sexual dimorphism in the mitochondrial metabolic protein profiles of MVs, which may contribute to sex-dependent cerebrovascular and neurological pathologies.


Asunto(s)
Biología Computacional/métodos , Microvasos/metabolismo , Mitocondrias/metabolismo , Proteómica/métodos , Animales , Femenino , Masculino , Ratas , Ratas Sprague-Dawley
13.
J Am Heart Assoc ; 10(20): e021113, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34622677

RESUMEN

Background Many therapies designed to prevent delayed cerebral ischemia (DCI) and improve neurological outcome in aneurysmal subarachnoid hemorrhage (SAH) have failed, likely because of targeting only one element of what has proven to be a multifactorial disease. We previously demonstrated that initiating hypoxic conditioning before SAH (hypoxic preconditioning) provides powerful protection against DCI. Here, we expanded upon these findings to determine whether hypoxic conditioning delivered at clinically relevant time points after SAH (hypoxic postconditioning) provides similarly robust DCI protection. Methods and Results In this study, we found that hypoxic postconditioning (8% O2 for 2 hours) initiated 3 hours after SAH provides strong protection against cerebral vasospasm, microvessel thrombi, and neurological deficits. By pharmacologic and genetic inhibition of SIRT1 (sirtuin 1) using EX527 and global Sirt1-/- mice, respectively, we demonstrated that this multifaceted DCI protection is SIRT1 mediated. Moreover, genetic overexpression of SIRT1 using Sirt1-Tg mice, mimicked the DCI protection afforded by hypoxic postconditioning. Finally, we found that post-SAH administration of resveratrol attenuated cerebral vasospasm, microvessel thrombi, and neurological deficits, and did so in a SIRT1-dependent fashion. Conclusions The present study indicates that hypoxic postconditioning provides powerful DCI protection when initiated at clinically relevant time points, and that pharmacologic augmentation of SIRT1 activity after SAH can mimic this beneficial effect. We conclude that conditioning-based therapies administered after SAH hold translational promise for patients with SAH and warrant further investigation.


Asunto(s)
Isquemia Encefálica , Hemorragia Subaracnoidea , Vasoespasmo Intracraneal , Animales , Infarto Cerebral , Humanos , Hipoxia/complicaciones , Ratones , Sirtuina 1/genética , Hemorragia Subaracnoidea/complicaciones
14.
Invest Ophthalmol Vis Sci ; 61(11): 15, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32910134

RESUMEN

Purpose: Stress can lead to short- or long-term changes in phenotype. Accumulating evidence also supports the transmission of maladaptive phenotypes, induced by adverse stressors, through the germline to manifest in subsequent generations, providing a novel mechanistic basis for the heritability of disease. In the present study in mice, we tested the hypothesis that repeated presentations of a nonharmful conditioning stress, demonstrated previously to protect against retinal ischemia, will also provide ischemic protection in the retinae of their untreated, first-generation (F1) adult offspring. Methods: Swiss-Webster ND4 outbred mice were mated following a 16-week period of brief, every-other-day conditioning exposures to mild systemic hypoxia (repetitive hypoxic conditioning, RHC). Retinae of their 5-month-old F1 progeny were subjected to unilateral ischemia. Scotopic electroretinography quantified postischemic outcomes. The injury-resilient retinal proteome was revealed by quantitative mass spectrometry, and bioinformatic analyses identified the biochemical pathways and networks in which these differentially expressed proteins operate. Results: Significant resilience to injury in both sexes was documented in F1 mice derived from RHC-treated parents, relative to matched F1 adult progeny derived from normoxic control parents. Ischemia-induced increases and decreases in the expression of many visual transduction proteins that are integral to photoreceptor function were abrogated by parental RHC, providing a molecular basis for the observed functional protection. Conclusions: Our proteomic analyses provided mechanistic insights into the molecular manifestation of the inherited, injury-resilient phenotype. To our knowledge, this is the first study in a mammalian model documenting the reprogramming of heritability to promote disease resilience in the next generation.


Asunto(s)
Isquemia/prevención & control , Precondicionamiento Isquémico/métodos , Neuroprotección , Proteoma/metabolismo , Proteómica/métodos , Enfermedades de la Retina/prevención & control , Vasos Retinianos/patología , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Femenino , Isquemia/diagnóstico , Isquemia/metabolismo , Masculino , Ratones , Ratones Endogámicos , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/metabolismo
15.
Exp Neurol ; 334: 113484, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33010255

RESUMEN

BACKGROUND AND PURPOSE: Vasospasm and delayed cerebral ischemia (DCI) contribute significantly to the morbidity/mortality associated with aneurysmal subarachnoid hemorrhage (SAH). While considerable research effort has focused on preventing or reversing vasospasm, SAH-induced brain injury occurs in response to a multitude of concomitantly acting pathophysiologic mechanisms. In this regard, the pleiotropic epigenetic responses to conditioning-based therapeutics may provide an ideal SAH therapeutic strategy. We previously documented the ability of hypoxic preconditioning (PC) to attenuate vasospasm and neurological deficits after SAH, in a manner that depends on the activity of endothelial nitric oxide synthase. The present study was undertaken to elucidate whether the NAD-dependent protein deacetylase sirtuin isoform SIRT1 is an upstream mediator of hypoxic PC-induced protection, and to assess the efficacy of the SIRT1-activating polyphenol Resveratrol as a pharmacologic preconditioning therapy. METHODS: Wild-type C57BL/6J mice were utilized in the study and subjected to normoxia or hypoxic PC. Surgical procedures included induction of SAH via endovascular perforation or sham surgery. Multiple endpoints were assessed including cerebral vasospasm, neurobehavioral deficits, SIRT1 expression via quantitative real-time PCR for mRNA, and western blot for protein quantification. Pharmacological agents utilized in the study include EX-527 (SIRT1 inhibitor), and Resveratrol (SIRT1 activator). RESULTS: Hypoxic PC leads to rapid and sustained increase in cerebral SIRT1 mRNA and protein expression. SIRT1 inhibition blocks the protective effects of hypoxic PC on vasospasm and neurological deficits. Resveratrol pretreatment dose-dependently abrogates vasospasm and attenuates neurological deficits following SAH - beneficial effects that were similarly blocked by pharmacologic inhibition of SIRT1. CONCLUSION: SIRT1 mediates hypoxic preconditioning-induced protection against neurovascular dysfunction after SAH. Resveratrol mimics this neurovascular protection, at least in part, via SIRT1. Activation of SIRT1 is a promising, novel, pleiotropic therapeutic strategy to combat DCI after SAH.


Asunto(s)
Hipoxia-Isquemia Encefálica/metabolismo , Precondicionamiento Isquémico/métodos , Sirtuina 1/metabolismo , Hemorragia Subaracnoidea/metabolismo , Vasoespasmo Intracraneal/metabolismo , Animales , Antioxidantes/farmacología , Carbazoles/farmacología , Hipoxia-Isquemia Encefálica/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Resveratrol/farmacología , Sirtuina 1/antagonistas & inhibidores , Hemorragia Subaracnoidea/patología , Vasoespasmo Intracraneal/patología , Vasoespasmo Intracraneal/prevención & control
16.
PLoS One ; 15(2): e0227263, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32017777

RESUMEN

Remote limb ischemic conditioning (RLIC) is a clinically feasible method in which brief, sub-lethal bouts of ischemia protects remote organs or tissues from subsequent ischemic injury. A single session of RLIC can improve exercise performance and increase muscle activation. The purpose of this study, therefore, was to assess the effects of a brief, two-week protocol of repeated RLIC combined with strength training on strength gain and neural adaptation in healthy young adults. Participants age 18-40 years were randomized to receive either RLIC plus strength training (n = 15) or sham conditioning plus strength training (n = 15). Participants received RLIC or sham conditioning over 8 visits using a blood pressure cuff on the dominant arm with 5 cycles of 5 minutes each alternating inflation and deflation. Visits 3-8 paired conditioning with wrist extensors strength training on the non-dominant (non-conditioned) arm using standard guidelines. Changes in one repetition maximum (1 RM) and electromyography (EMG) amplitude were compared between groups. Both groups were trained at a similar workload. While both groups gained strength over time (P = 0.001), the RLIC group had greater strength gains (9.38 ± 1.01 lbs) than the sham group (6.3 ± 1.08 lbs, P = 0.035). There was not a significant group x time interaction in EMG amplitude (P = 0.231). The RLIC group had larger percent changes in 1 RM (43.8% vs. 26.1%, P = 0.003) and EMG amplitudes (31.0% vs. 8.6%, P = 0.023) compared to sham conditioning. RLIC holds promise for enhancing muscle strength in healthy young and older adults, as well as clinical populations that could benefit from strength training.


Asunto(s)
Extremidades/irrigación sanguínea , Precondicionamiento Isquémico/métodos , Fuerza Muscular/fisiología , Músculo Esquelético/irrigación sanguínea , Entrenamiento de Fuerza/métodos , Adolescente , Adulto , Femenino , Voluntarios Sanos , Humanos , Masculino , Adulto Joven
17.
Stroke ; 40(10): 3342-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19644058

RESUMEN

BACKGROUND AND PURPOSE: The importance of bioactive lipid signaling under physiological and pathophysiological conditions is progressively becoming recognized. The disparate distribution of sphingosine kinase (SphK) isoform activity in normal and ischemic brain, particularly the large excess of SphK2 in cerebral microvascular endothelial cells, suggests potentially unique cell- and region-specific signaling by its product sphingosine-1-phosphate. The present study sought to test the isoform-specific role of SphK as a trigger of hypoxic preconditioning (HPC)-induced ischemic tolerance. METHODS: Temporal changes in microvascular SphK activity and expression were measured after HPC. The SphK inhibitor dimethylsphingosine or sphingosine analog FTY720 was administered to adult male Swiss-Webster ND4 mice before HPC. Two days later, mice underwent a 60-minute transient middle cerebral artery occlusion and at 24 hours of reperfusion, infarct volume, neurological deficit, and hemispheric edema were measured. RESULTS: HPC rapidly increased microvascular SphK2 protein expression (1.7+/-0.2-fold) and activity (2.5+/-0.6-fold), peaking at 2 hours, whereas SphK1 was unchanged. SphK inhibition during HPC abrogated reductions in infarct volume, neurological deficit, and ipsilateral edema in HPC-treated mice. FTY720 given 48 hours before stroke also promoted ischemic tolerance; when combined with HPC, even greater (and dimethylsphingosine-reversible) protection was noted. CONCLUSIONS: These findings indicate hypoxia-sensitive increases in SphK2 activity may serve as a proximal trigger that ultimately leads to sphingosine-1-phosphate-mediated alterations in gene expression that promote the ischemia-tolerant phenotype. Thus, components of this bioactive lipid signaling pathway may be suitable therapeutic targets for protecting the neurovascular unit in stroke.


Asunto(s)
Arterias Cerebrales/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/terapia , Precondicionamiento Isquémico/métodos , Microcirculación/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Arteriolas/metabolismo , Arteriolas/fisiopatología , Edema Encefálico/etiología , Edema Encefálico/fisiopatología , Edema Encefálico/terapia , Arterias Cerebrales/fisiopatología , Circulación Cerebrovascular/fisiología , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Hipoxia-Isquemia Encefálica/fisiopatología , Inmunosupresores/farmacología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Infarto de la Arteria Cerebral Media/terapia , Lisofosfolípidos/metabolismo , Masculino , Ratones , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Glicoles de Propileno/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/terapia , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología
18.
Neurobiol Dis ; 35(1): 82-90, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19393318

RESUMEN

Neutrophil elastase (NE) degrades basal lamina and extracellular matrix molecules, and recruits leukocytes during inflammation; however, a basic understanding of the role of NE in stroke pathology is lacking. We measured an increased number of extravascular NE-positive cells, as well as increased levels of tissue elastase protein and activity, following transient middle cerebral artery occlusion (tMCAo). Both pharmacologic inhibition of NE with ZN200355 (ZN), and genetic deletion of NE, significantly reduced infarct volume, blood-brain barrier disruption, vasogenic edema, and leukocyte-endothelial adherence 24 h after tMCAo. ZN also reduced infarct volume in MMP9-null mice following tMCAo. There were, however, no reductions in infarct volume or vasogenic edema in NE-null mice in two models of permanent middle cerebral artery occlusion. Our findings confirm the involvement of NE in neurovascular stroke pathology, when reperfusion allows neutrophils access to vulnerable brain, with pharmacologic or genetic inhibition of NE being both neuro- and vasculo-protective in this setting.


Asunto(s)
Infarto Encefálico/etiología , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/metabolismo , Daño por Reperfusión/patología , Análisis de Varianza , Animales , Análisis de los Gases de la Sangre , Presión Sanguínea/genética , Barrera Hematoencefálica/fisiopatología , Encéfalo/irrigación sanguínea , Edema Encefálico/patología , Infarto Encefálico/prevención & control , Adhesión Celular , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Flujometría por Láser-Doppler/métodos , Elastasa de Leucocito/antagonistas & inhibidores , Leucocitos/patología , Masculino , Metaloproteinasa 9 de la Matriz/deficiencia , Ratones , Ratones Noqueados , Daño por Reperfusión/metabolismo , Sales de Tetrazolio
19.
Transl Stroke Res ; 10(4): 362-371, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30088217

RESUMEN

Remote limb ischemic conditioning (RLIC) is a clinically feasible method of promoting tissue protection against subsequent ischemic insult. Recent findings from our lab demonstrated that RLIC robustly enhances motor learning in young, healthy humans. The next step is to determine which individuals would receive maximum benefit from RLIC before applying these findings to clinical rehabilitation populations such as stroke. Numerous factors, such as age, sex, body mass index (BMI), and cardiovascular comorbidities may influence the response. Sixty-nine participants aged 40-80 were randomized to receive either RLIC (n = 33) or sham (n = 36) conditioning. Participants underwent seven consecutive sessions consisting of RLIC or sham conditioning with a blood pressure cuff on the upper extremity and motor training on a stability platform balance task, with two follow-up sessions. Balance change (post-test-pre-test) was compared across participants, groups, and the factors of age, sex, BMI, and comorbidities. Participants in both groups improved their performance on the balance task from pre- to post-test. Overall balance change was independently associated with age and BMI. There was no difference in balance change between RLIC and Sham groups. However, RLIC significantly enhanced balance performance in participants with no comorbidities. Compared with our previous study in young adults, middle-aged and older adults demonstrated smaller improvements on the balance task. RLIC enhanced learning in middle-aged and older adults only in the absence of pre-defined comorbidities. RLIC may be a promising tool for enhancing motor recovery, but the accumulation of comorbidity with age may decrease its effectiveness.


Asunto(s)
Isquemia/prevención & control , Precondicionamiento Isquémico/métodos , Aprendizaje/fisiología , Destreza Motora/fisiología , Extremidad Superior/irrigación sanguínea , Extremidad Superior/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Humanos , Isquemia/fisiopatología , Precondicionamiento Isquémico/instrumentación , Masculino , Persona de Mediana Edad , Equilibrio Postural/fisiología
20.
Front Neurol ; 10: 447, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31118919

RESUMEN

Background: Asphyxia is the most common cause of brain damage in newborns. Substantial evidence indicates that leukocyte recruitment in the cerebral vasculature during asphyxia contributes to this damage. We tested the hypothesis that superoxide radical ( O 2 ⋅ _ ) promotes an acute post-asphyxial inflammatory response and blood-brain barrier (BBB) breakdown. We investigated the effects of removing O 2 ⋅ _ by superoxide dismutase (SOD) or C3, the cell-permeable SOD mimetic, in protecting against asphyxia-related leukocyte recruitment. We also tested the hypothesis that xanthine oxidase activity is one source of this radical. Methods: Anesthetized piglets were tracheostomized, ventilated, and equipped with closed cranial windows for the assessment of post-asphyxial rhodamine 6G-labeled leukocyte-endothelial adherence and microvascular permeability to sodium fluorescein in cortical venules. Asphyxia was induced by discontinuing ventilation. SOD and C3 were administered by cortical superfusion. The xanthine oxidase inhibitor oxypurinol was administered intravenously. Results: Leukocyte-venular adherence significantly increased during the initial 2 h of post-asphyxial reperfusion. BBB permeability was also elevated relative to non-asphyxial controls. Inhibition of O 2 ⋅ _ production by oxypurinol, or elimination of O 2 ⋅ _ by SOD or C3, significantly reduced rhodamine 6G-labeled leukocyte-endothelial adherence and improved BBB integrity, as measured by sodium fluorescein leak from cerebral microvessels. Conclusion: Using three different strategies to either prevent formation or enhance elimination of O 2 ⋅ _ during the post-asphyxial period, we saw both reduced leukocyte adherence and preserved BBB function with treatment. These findings suggest that agents which lower O 2 ⋅ _ in brain may be attractive new therapeutic interventions for the protection of the neonatal brain following asphyxia.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA