Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 134(6): 695-710, 2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38484025

RESUMEN

Circadian and diurnal variation in cerebral blood flow directly contributes to the diurnal variation in the risk of stroke, either through factors that trigger stroke or due to impaired compensatory mechanisms. Cerebral blood flow results from the integration of systemic hemodynamics, including heart rate, cardiac output, and blood pressure, with cerebrovascular regulatory mechanisms, including cerebrovascular reactivity, autoregulation, and neurovascular coupling. We review the evidence for the circadian and diurnal variation in each of these mechanisms and their integration, from the detailed evidence for mechanisms underlying the nocturnal nadir and morning surge in blood pressure to identifying limited available evidence for circadian and diurnal variation in cerebrovascular compensatory mechanisms. We, thus, identify key systemic hemodynamic factors related to the diurnal variation in the risk of stroke but particularly identify the need for further research focused on cerebrovascular regulatory mechanisms.


Asunto(s)
Accidente Cerebrovascular , Humanos , Presión Sanguínea/fisiología , Hemodinámica , Ritmo Circadiano , Circulación Cerebrovascular/fisiología
2.
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.

3.
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
4.
Stroke ; 55(7): 1904-1913, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38913800

RESUMEN

BACKGROUND: The mitochondrial unfolded protein response (UPRmt) is an evolutionarily conserved mitochondrial response that is critical for maintaining mitochondrial and energetic homeostasis under cellular stress after tissue injury and disease. Here, we ask whether UPRmt may be a potential therapeutic target for ischemic stroke. METHODS: We performed the middle cerebral artery occlusion and oxygen-glucose deprivation models to mimic ischemic stroke in vivo and in vitro, respectively. Oligomycin and meclizine were used to trigger the UPRmt. We used 2,3,5-triphenyltetrazolium chloride staining, behavioral tests, and Nissl staining to evaluate cerebral injury in vivo. The Cell Counting Kit-8 assay and the Calcein AM Assay Kit were conducted to test cerebral injury in vitro. RESULTS: Inducing UPRmt with oligomycin protected neuronal cultures against oxygen-glucose deprivation. UPRmt could also be triggered with meclizine, and this Food and Drug Administration-approved drug also protected neurons against oxygen-glucose deprivation. Blocking UPRmt with siRNA against activating transcription factor 5 eliminated the neuroprotective effects of meclizine. In a mouse model of focal cerebral ischemia, pretreatment with meclizine was able to induce UPRmt in vivo, which reduced infarction and improved neurological outcomes. CONCLUSIONS: These findings suggest that the UPRmt is important in maintaining the survival of neurons facing ischemic/hypoxic stress. The UPRmt mechanism may provide a new therapeutic avenue for ischemic stroke.


Asunto(s)
Isquemia Encefálica , Glucosa , Mitocondrias , Neuronas , Respuesta de Proteína Desplegada , Animales , Masculino , Ratones , Isquemia Encefálica/metabolismo , Células Cultivadas , Glucosa/deficiencia , Infarto de la Arteria Cerebral Media/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oxígeno/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos
5.
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
6.
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
7.
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
8.
Int J Mol Sci ; 21(23)2020 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-33260683

RESUMEN

A-kinase anchor protein 12 (AKAP12) is a scaffolding protein that associates with intracellular molecules to regulate multiple signal transductions. Although the roles of AKAP12 in the central nervous system are still relatively understudied, it was previously shown that AKAP12 regulates blood-retinal barrier formation. In this study, we asked whether AKAP12 also supports the function and integrity of the blood-brain barrier (BBB). In a mouse model of focal ischemia, the expression level of AKAP12 in cerebral endothelial cells was upregulated during the acute phase of stroke. Also, in cultured cerebral endothelial cells, oxygen-glucose deprivation induced the upregulation of AKAP12. When AKAP12 expression was suppressed by an siRNA approach in cultured endothelial cells, endothelial permeability was increased along with the dysregulation of ZO-1/Claudin 5 expression. In addition, the loss of AKAP12 expression caused an upregulation/activation of the Rho kinase pathway, and treatment of Rho kinase inhibitor Y-27632 mitigated the increase of endothelial permeability in AKAP12-deficient endothelial cell cultures. These in vitro findings were confirmed by our in vivo experiments using Akap12 knockout mice. Compared to wild-type mice, Akap12 knockout mice showed a larger extent of BBB damage after stroke. However, the inhibition of rho kinase by Y-27632 tightened the BBB in Akap12 knockout mice. These data may suggest that endogenous AKAP12 works to alleviate the damage and dysfunction of the BBB caused by ischemic stress. Therefore, the AKAP12-rho-kinase signaling pathway represents a novel therapeutic target for stroke.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Proteínas de Ciclo Celular/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Animales , Permeabilidad de la Membrana Celular , Endotelio Vascular/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Quinasas Asociadas a rho/metabolismo
9.
Stem Cells ; 36(9): 1404-1410, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29781122

RESUMEN

Endothelial progenitor cells (EPCs) have been pursued as a potential cellular therapy for stroke and central nervous system injury. However, their underlying mechanisms remain to be fully defined. Recent experimental studies suggest that mitochondria may be released and transferred between cells. In this proof-of-concept study, we asked whether beneficial effects of EPCs may partly involve a mitochondrial phenomenon as well. First, EPC-derived conditioned medium was collected and divided into supernatant and particle fractions after centrifugation. Electron microscopy, Western blots, and flow cytometry showed that EPCs were able to release mitochondria. ATP and oxygen consumption assays suggested that these extracellular mitochondria may still be functionally viable. Confocal microscopy confirmed that EPC-derived extracellular mitochondria can be incorporated into normal brain endothelial cells. Adding EPC particles to brain endothelial cells promoted angiogenesis and decreased the permeability of brain endothelial cells. Next, we asked whether EPC-derived mitochondria may be protective. As expected, oxygen-glucose deprivation (OGD) increased brain endothelial permeability. Adding EPC-derived mitochondria particles to the damaged brain endothelium increased levels of mitochondrial protein TOM40, mitochondrial DNA copy number, and intracellular ATP. Along with these indirect markers of mitochondrial transfer, endothelial tightness was also restored after OGD. Taken together, these findings suggest that EPCs may support brain endothelial energetics, barrier integrity, and angiogenic function partly through extracellular mitochondrial transfer. Stem Cells 2018;36:1404-1410.


Asunto(s)
Encéfalo/metabolismo , Células Progenitoras Endoteliales/metabolismo , Endotelio/metabolismo , Mitocondrias/metabolismo , Humanos , Transducción de Señal
10.
Stem Cells ; 36(5): 751-760, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29314444

RESUMEN

Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes in cerebral white matter. However, the underlying mechanisms that regulate this process remain to be fully defined, especially in adult brains. Recently, it has been suggested that signaling via A-kinase anchor protein 12 (AKAP12), a scaffolding protein that associates with intracellular molecules such as protein kinase A, may be involved in Schwann cell homeostasis and peripheral myelination. Here, we asked whether AKAP12 also regulates the mechanisms of myelination in the CNS. AKAP12 knockout mice were compared against wild-type (WT) mice in a series of neurochemical and behavioral assays. Compared with WTs, 2-months old AKAP12 knockout mice exhibited loss of myelin in white matter of the corpus callosum, along with perturbations in working memory as measured by a standard Y-maze test. Unexpectedly, very few OPCs expressed AKAP12 in the corpus callosum region. Instead, pericytes appeared to be one of the major AKAP12-expressing cells. In a cell culture model system, conditioned culture media from normal pericytes promoted in-vitro OPC maturation. However, conditioned media from AKAP12-deficient pericytes did not support the OPC function. These findings suggest that AKAP12 signaling in pericytes may be required for OPC-to-oligodendrocyte renewal to maintain the white matter homeostasis in adult brain. Stem Cells 2018;36:751-760.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/fisiología , Células-Madre Neurales/citología , Oligodendroglía/metabolismo , Sustancia Blanca/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Envejecimiento , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Medios de Cultivo Condicionados , Ratones Noqueados , Vaina de Mielina/metabolismo , Neurogénesis/fisiología , Oligodendroglía/citología , Sustancia Blanca/citología
11.
Neuroimage ; 178: 713-720, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29738908

RESUMEN

Magnetic Particle Imaging (MPI) is a rapidly developing imaging modality that directly measures and maps the concentration of injected superparamagnetic iron oxide nanoparticles (SPIOs). Since the agent does not cross the blood-brain barrier, cerebral SPIO concentration provides a direct probe of Cerebral Blood Volume (CBV). Here we provide an initial demonstration of the ability of MPI to detect functional CBV changes (fCBV) by monitoring SPIO concentration during hypercapnic manipulation in a rat model. As a tracer detection method, MPI offers a more direct probe of agent concentration and therefore fCBV than MRI measurements in which the agent is indirectly detected through perturbation of water relaxation time constants such as T2∗. We found that MPI detection could measure CBV changes during hypercapnia with high CNR (CNR = 50) and potentially with high temporal resolution. Although the detection process more closely resembles a tracer method, we also identify evidence of physiological noise in the MPI time-series, with higher time-series variance at higher concentration levels. Our findings suggest that CBV-based MPI can provide a detection modality for hemodynamic changes. Further investigation with tomographic imaging is needed to assess tomographic ability of the method and further study the presence of time-series fluctuations which scale with signal level similar to physiological noise in resting fMRI time-courses.


Asunto(s)
Determinación del Volumen Sanguíneo/métodos , Encéfalo/irrigación sanguínea , Volumen Sanguíneo Cerebral , Óxido Ferrosoférrico/farmacocinética , Neuroimagen/métodos , Animales , Determinación del Volumen Sanguíneo/instrumentación , Hipercapnia/fisiopatología , Ratas , Ratas Sprague-Dawley
12.
AJR Am J Roentgenol ; 210(4): 720-727, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29470156

RESUMEN

OBJECTIVE: Diffusion kurtosis imaging (DKI) has emerged as a new acute stroke imaging approach, augmenting routine DWI. Although it has been shown that a diffusion lesion without kurtosis abnormality is more likely to recover after reperfusion, whereas a kurtosis lesion shows poor response, little is known about the underlying pathophysiologic profile of the kurtosis lesion versus the kurtosis lesion-diffusion lesion mismatch. MATERIALS AND METHODS: We performed multiparametric MRI, including arterial spin labeling, pH-sensitive amide proton transfer, and DKI, in a rodent model of acute stroke caused by embolic middle cerebral artery occlusion. Diffusion and kurtosis lesions were semiautomatically segmented, and multiparametric MRI indexes were compared among the kurtosis lesion, diffusion lesion, kurtosis lesion-diffusion lesion mismatch, and the contralateral normal tissue area. RESULTS: We confirmed a significant difference between diffusion lesion and kurtosis lesion volumes (mean [± SD] volume, 151 ± 65 vs 125 ± 47 mm3; p < 0.05). Although ischemic lesions have significantly reduced cerebral blood flow compared with contralateral normal tissue, we did not find a significant difference in cerebral blood flow between the kurtosis lesion and the kurtosis lesion-diffusion lesion mismatch (mean cerebral blood flow, 0.53 ± 0.10 vs 0.47 ± 0.14 mL/g of tissue per minute; p > 0.05). Of importance, the pH of the kurtosis lesion was significantly lower than that of the lesion mismatch (mean pH, 6.81 ± 0.08 vs 6.89 ± 0.09; p < 0.01). CONCLUSION: The present study confirms that DKI provides an expedient approach for refining the heterogeneous DWI lesion that is associated with graded metabolic derangement, which is promising for improving the infarction core definition and ultimately helping to guide stroke treatment.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/métodos , Accidente Cerebrovascular/diagnóstico por imagen , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Imagen Eco-Planar , Interpretación de Imagen Asistida por Computador , Masculino , Ratas , Ratas Wistar , Marcadores de Spin
13.
J Neurosci ; 35(8): 3663-75, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716864

RESUMEN

The blood oxygenation level-dependent (BOLD) contrast is widely used in functional magnetic resonance imaging (fMRI) studies aimed at investigating neuronal activity. However, the BOLD signal reflects changes in blood volume and oxygenation rather than neuronal activity per se. Therefore, understanding the transformation of microscopic vascular behavior into macroscopic BOLD signals is at the foundation of physiologically informed noninvasive neuroimaging. Here, we use oxygen-sensitive two-photon microscopy to measure the BOLD-relevant microvascular physiology occurring within a typical rodent fMRI voxel and predict the BOLD signal from first principles using those measurements. The predictive power of the approach is illustrated by quantifying variations in the BOLD signal induced by the morphological folding of the human cortex. This framework is then used to quantify the contribution of individual vascular compartments and other factors to the BOLD signal for different magnet strengths and pulse sequences.


Asunto(s)
Encéfalo/irrigación sanguínea , Interpretación de Imagen Asistida por Computador/métodos , Angiografía por Resonancia Magnética/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Modelos Cardiovasculares , Animales , Encéfalo/fisiología , Colorantes Fluorescentes , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno , Ratas , Ratas Sprague-Dawley
14.
Stroke ; 47(4): 1094-100, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26965847

RESUMEN

BACKGROUND AND PURPOSE: Pentraxin 3 (PTX3) is released on inflammatory responses in many organs. However, roles of PTX3 in brain are still mostly unknown. Here we asked whether and how PTX3 contributes to blood-brain barrier dysfunction during the acute phase of ischemic stroke. METHODS: In vivo, spontaneously hypertensive rats were subjected to focal cerebral ischemia by transient middle cerebral artery occlusion. At day 3, brains were analyzed to evaluate the cellular origin of PTX3 expression. Correlations with blood-brain barrier breakdown were assessed by IgG staining. In vitro, rat primary astrocytes and rat brain endothelial RBE.4 cells were cultured to study the role of astrocyte-derived PTX3 on vascular endothelial growth factor-mediated endothelial permeability. RESULTS: During the acute phase of stroke, reactive astrocytes in the peri-infarct area expressed PTX3. There was negative correlation between gradients of IgG leakage and PTX3-positive astrocytes. Cell culture experiments showed that astrocyte-conditioned media increased levels of tight junction proteins and reduced endothelial permeability under normal conditions. Removing PTX3 from astrocyte-conditioned media by immunoprecipitation increased endothelial permeability. PTX3 strongly bound vascular endothelial growth factor in vitro and was able to decrease vascular endothelial growth factor-induced endothelial permeability. CONCLUSIONS: Astrocytes in peri-infarct areas upregulate PTX3, which may support blood-brain barrier integrity by regulating vascular endothelial growth factor-related mechanisms. This response in astrocytes may comprise a compensatory mechanism for maintaining blood-brain barrier function after ischemic stroke.


Asunto(s)
Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Proteína C-Reactiva/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Componente Amiloide P Sérico/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular , Medios de Cultivo Condicionados , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratas , Ratas Endogámicas SHR , Accidente Cerebrovascular/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Neuroimage ; 141: 242-249, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27444569

RESUMEN

pH-sensitive amide proton transfer (APT) MRI provides a surrogate metabolic biomarker that complements the widely-used perfusion and diffusion imaging. However, the endogenous APT MRI is often calculated using the asymmetry analysis (MTRasym), which is susceptible to an inhomogeneous shift due to concomitant semisolid magnetization transfer (MT) and nuclear overhauser (NOE) effects. Although the intact brain tissue has little pH variation, white and gray matter appears distinct in the MTRasym image. Herein we showed that the heterogeneous MTRasym shift not related to pH highly correlates with MT ratio (MTR) and longitudinal relaxation rate (R1w), which can be reasonably corrected using the multiple regression analysis. Because there are relatively small MT and R1w changes during acute stroke, we postulate that magnetization transfer and relaxation-normalized APT (MRAPT) analysis increases MRI specificity to acidosis over the routine MTRasym image, hence facilitates ischemic lesion segmentation. We found significant differences in perfusion, pH and diffusion lesion volumes (P<0.001, ANOVA). Furthermore, MRAPT MRI depicted graded ischemic acidosis, with the most severe acidosis in the diffusion lesion (-1.05±0.29%/s), moderate acidification within the pH/diffusion mismatch (i.e., metabolic penumbra, -0.67±0.27%/s) and little pH change in the perfusion/pH mismatch (i.e., benign oligemia, -0.04±0.14%/s), providing refined stratification of ischemic tissue injury.


Asunto(s)
Amidas/química , Química Encefálica , Encéfalo/diagnóstico por imagen , Concentración de Iones de Hidrógeno , Imagen por Resonancia Magnética/métodos , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/metabolismo , Algoritmos , Amidas/metabolismo , Animales , Biomarcadores/química , Interpretación de Imagen Asistida por Computador/métodos , Campos Magnéticos , Masculino , Protones , Ratas , Ratas Wistar , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
16.
NMR Biomed ; 29(12): 1670-1677, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27696558

RESUMEN

Diffusion kurtosis imaging (DKI) has been shown to augment diffusion-weighted imaging (DWI) for the definition of irreversible ischemic injury. However, the complexity of cerebral structure/composition makes the kurtosis map heterogeneous, limiting the specificity of kurtosis hyperintensity to acute ischemia. We propose an Inherent COrrelation-based Normalization (ICON) analysis to suppress the intrinsic kurtosis heterogeneity for improved characterization of heterogeneous ischemic tissue injury. Fast DKI and relaxation measurements were performed on normal (n = 10) and stroke rats following middle cerebral artery occlusion (MCAO) (n = 20). We evaluated the correlations between mean kurtosis (MK), mean diffusivity (MD) and fractional anisotropy (FA) derived from the fast DKI sequence and relaxation rates R1 and R2 , and found a highly significant correlation between MK and R1 (p < 0.001). We showed that ICON analysis suppressed the intrinsic kurtosis heterogeneity in normal cerebral tissue, enabling automated tissue segmentation in an animal stroke model. We found significantly different kurtosis and diffusivity lesion volumes: 147 ± 59 and 180 ± 66 mm3 , respectively (p = 0.003, paired t-test). The ratio of kurtosis to diffusivity lesion volume was 84% ± 19% (p < 0.001, one-sample t-test). We found that relaxation-normalized MK (RNMK), but not MD, values were significantly different between kurtosis and diffusivity lesions (p < 0.001, analysis of variance). Our study showed that fast DKI with ICON analysis provides a promising means of demarcation of heterogeneous DWI stroke lesions.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/patología , Imagen de Difusión por Resonancia Magnética/métodos , Interpretación de Imagen Asistida por Computador/métodos , Infarto de la Arteria Cerebral Media/diagnóstico por imagen , Infarto de la Arteria Cerebral Media/patología , Reconocimiento de Normas Patrones Automatizadas/métodos , Enfermedad Aguda , Algoritmos , Animales , Aumento de la Imagen/métodos , Aprendizaje Automático , Masculino , Modelos Biológicos , Modelos Estadísticos , Ratas , Ratas Wistar , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Estadística como Asunto
17.
Bioorg Med Chem Lett ; 25(17): 3748-53, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26142946

RESUMEN

A novel neurogenic compound (1), discovered from a mouse neural progenitor cell (NPC) screen, showed profound neurogenic effect on human NPCs. Synthesis and SAR of this novel 2,3,11,11a-tetrahydro-1H-pyrazino[1,2-b]isoquinoline-1,4(6H)-dione series are described. Compound 20 is brain penetrable in rodents, and promotes neurogenesis in wild type mice, therefore it is a good tool molecule to study neurogenesis induction as a potential treatment for conditions associated with neurogenesis impairment diseases.


Asunto(s)
Isoquinolinas/química , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Pirazinas/farmacocinética , Relación Estructura-Actividad , Administración Oral , Animales , Encéfalo/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Química Sintética , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos/métodos , Semivida , Humanos , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacocinética , Isoquinolinas/farmacología , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Pirazinas/administración & dosificación , Pirazinas/química
18.
Proc Natl Acad Sci U S A ; 109(51): 20826-31, 2012 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-23213211

RESUMEN

We report a group of optical imaging probes, comprising upconverting lanthanide nanoparticles (UCNPs) and polyanionic dendrimers. Dendrimers with rigid cores and multiple carboxylate groups at the periphery are able to tightly bind to surfaces of UCNPs pretreated with NOBF(4), yielding stable, water-soluble, biocompatible nanomaterials. Unlike conventional linear polymers, dendrimers adhere to UCNPs by donating only a fraction of their peripheral groups to the UCNP-surface interactions. The remaining termini make up an interface between the nanoparticle and the aqueous phase, enhancing solubility and offering multiple possibilities for subsequent modification. Using optical probes as dendrimer cores makes it possible to couple the UCNPs signal to analyte-sensitive detection via UCNP-to-chromophore excitation energy transfer (EET). As an example, we demonstrate that UCNPs modified with porphyrin-dendrimers can operate as upconverting ratiometric pH nanosensors. Dendritic UCNPs possess excellent photostability, solubility, and biocompatibility, which make them directly suitable for in vivo imaging. Polyglutamic dendritic UCNPs injected in the blood of a mouse allowed mapping of the cortical vasculature down to 400 µm under the tissue surface, thus demonstrating feasibility of in vivo high-resolution two-photon microscopy with continuous wave (CW) excitation sources. Dendrimerization as a method of solubilization of UCNPs opens up numerous possibilities for use of these unique agents in biological imaging and sensing.


Asunto(s)
Dendritas/fisiología , Microscopía/métodos , Nanopartículas/química , Animales , Aniones , Materiales Biocompatibles/química , Técnicas Biosensibles , Medios de Contraste/farmacología , Concentración de Iones de Hidrógeno , Ligandos , Masculino , Ensayo de Materiales , Ratones , Ratones Endogámicos C57BL , Modelos Químicos , Nanotecnología/métodos , Péptidos/química , Polímeros/química , Porfirinas/química , Solubilidad
19.
Stroke ; 45(2): 619-22, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24368559

RESUMEN

BACKGROUND AND PURPOSE: Tissue-type plasminogen activator (tPA) in combination with recombinant annexin A2 (rA2) is known to reduce acute brain damage after focal ischemia. Here, we ask whether tPA-plus-rA2 combination therapy can lead to sustained long-term neurological improvements as well. METHODS: We compared the effects of intravenous high-dose tPA alone (10 mg/kg) versus a combination of low-dose tPA (5 mg/kg) plus 10 mg/kg rA2 in a model of focal embolic cerebral ischemia in rats. All rats were treated at 3 hours after embolization. Brain tissue and neurological outcomes were assessed at 1 month. Surrogate biomarkers for endogenous neurovascular remodeling in peri-infarct area were analyzed by immunohistochemistry. RESULTS: Compared with high-dose tPA alone, low-dose tPA-plus-rA2 significantly decreased infarction and improved neurological function at 1-month poststroke. In peri-infarct areas, tPA-plus-rA2 combination therapy also significantly augmented microvessel density, vascular endothelial growth factor, and synaptophysin expression. CONCLUSIONS: Compared with conventional high-dose tPA alone, combination low-dose tPA plus rA2 therapy may provide a safe and effective way to improve long-term neurological outcomes after stroke.


Asunto(s)
Anexina A2/uso terapéutico , Antifibrinolíticos/uso terapéutico , Embolia Intracraneal/complicaciones , Accidente Cerebrovascular/terapia , Activador de Tejido Plasminógeno/uso terapéutico , Animales , Isquemia Encefálica/patología , Capilares/patología , Infarto Cerebral/patología , Terapia Combinada , Quimioterapia Combinada , Embolización Terapéutica , Inmunohistoquímica , Embolia Intracraneal/mortalidad , Masculino , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/fisiopatología , Ratas , Ratas Wistar , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/mortalidad , Sinaptofisina/biosíntesis , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/biosíntesis
20.
Stroke ; 45(7): 2085-92, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24916903

RESUMEN

BACKGROUND AND PURPOSE: We explored the hypothesis that injured neurons release lipocalin-2 as a help me signal. METHODS: In vivo lipocalin-2 responses were assessed in rat focal cerebral ischemia and human stroke brain samples using a combination of ELISA and immunostaining. In vitro, microglia and astrocytes were exposed to lipocalin-2, and various markers and assays of glial activation were quantified. Functional relevance of neuron-to-glia lipocalin-2 signaling was examined by transferring conditioned media from lipocalin-2-activated microglia and astrocytes onto neurons to see whether activated glia could protect neurons against oxygen-glucose deprivation and promote neuroplasticity. RESULTS: In human stroke samples and rat cerebral ischemia, neuronal expression of lipocalin-2 was significantly increased. In primary cell cultures, exposing microglia and astrocytes to lipocalin-2 resulted in glial activation. In microglia, lipocalin-2 converted resting ramified shapes into a long-rod morphology with reduced branching, increased interleukin-10 release, and enhanced phagocytosis. In astrocytes, lipocalin-2 upregulated glial fibrillary acid protein, brain-derived neurotropic factor, and thrombospondin-1. Conditioned media from lipocalin-2-treated astrocytes upregulated synaptotagmin, and conditioned media from lipocalin-2-treated microglia upregulated synaptophysin and post-synaptic density 95 (PSD95) and protected neurons against oxygen-glucose deprivation. CONCLUSIONS: These findings provide proof of concept that lipocalin-2 is released by injured neurons as a help me distress signal that activates microglia and astrocytes into potentially prorecovery phenotypes.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Astrocitos/metabolismo , Isquemia Encefálica/metabolismo , Corteza Cerebral/metabolismo , Lipocalinas/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Accidente Cerebrovascular/metabolismo , Anciano , Animales , Biomarcadores/metabolismo , Corteza Cerebral/patología , Femenino , Humanos , Lipocalina 2 , Masculino , Plasticidad Neuronal/fisiología , Fenotipo , Ratas , Ratas Wistar , Método Simple Ciego
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA