Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 44(21)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38569926

RESUMEN

Proteoglycans containing link domains modify the extracellular matrix (ECM) to regulate cellular homeostasis and can also sensitize tissues/organs to injury and stress. Hypoxic-ischemic (H-I) injury disrupts cellular homeostasis by activating inflammation and attenuating regeneration and repair pathways. In the brain, the main component of the ECM is the glycosaminoglycan hyaluronic acid (HA), but whether HA modifications of the ECM regulate cellular homeostasis and response to H-I injury is not known. In this report, employing both male and female mice, we demonstrate that link-domain-containing proteoglycan, TNFα-stimulated gene-6 (TSG-6), is active in the brain from birth onward and differentially modifies ECM HA during discrete neurodevelopmental windows. ECM HA modification by TSG-6 enables it to serve as a developmental switch to regulate the activity of the Hippo pathway effector protein, yes-associated protein 1 (YAP1), in the maturing brain and in response to H-I injury. Mice that lack TSG-6 expression display dysregulated expression of YAP1 targets, excitatory amino acid transporter 1 (EAAT1; glutamate-aspartate transporter) and 2 (EAAT2; glutamate transporter-1). Dysregulation of YAP1 activation in TSG-6-/- mice coincides with age- and sex-dependent sensitization of the brain to H-I injury such that 1-week-old neonates display an anti-inflammatory response in contrast to an enhanced proinflammatory injury reaction in 3-month-old adult males but not females. Our findings thus support that a key regulator of age- and sex-dependent H-I injury response in the mouse brain is modulation of the Hippo-YAP1 pathway by TSG-6-dependent ECM modifications.


Asunto(s)
Moléculas de Adhesión Celular , Matriz Extracelular , Hipoxia-Isquemia Encefálica , Proteínas Señalizadoras YAP , Animales , Femenino , Masculino , Moléculas de Adhesión Celular/metabolismo , Ratones , Matriz Extracelular/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Proteínas Señalizadoras YAP/metabolismo , Ratones Endogámicos C57BL , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Ácido Hialurónico/metabolismo , Ratones Noqueados , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética
2.
Ann Neurol ; 94(6): 1048-1066, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37605362

RESUMEN

OBJECTIVE: Because the role of white matter (WM) degenerating microglia (DM) in remyelination failure is unclear, we sought to define the core features of this novel population of aging human microglia. METHODS: We analyzed postmortem human brain tissue to define a population of DM in aging WM lesions. We used immunofluorescence staining and gene expression analysis to investigate molecular mechanisms related to the degeneration of DM. RESULTS: We found that DM, which accumulated myelin debris were selectively enriched in the iron-binding protein light chain ferritin, and accumulated PLIN2-labeled lipid droplets. DM displayed lipid peroxidation injury and enhanced expression for TOM20, a mitochondrial translocase, and a sensor of oxidative stress. DM also displayed enhanced expression of the DNA fragmentation marker phospho-histone H2A.X. We identified a unique set of ferroptosis-related genes involving iron-mediated lipid dysmetabolism and oxidative stress that were preferentially expressed in WM injury relative to gray matter neurodegeneration. INTERPRETATION: Ferroptosis appears to be a major mechanism of WM injury in Alzheimer's disease and vascular dementia. WM DM are a novel therapeutic target to potentially reduce the impact of WM injury and myelin loss on the progression of cognitive impairment. ANN NEUROL 2023;94:1048-1066.


Asunto(s)
Ferroptosis , Sustancia Blanca , Humanos , Microglía/metabolismo , Sustancia Blanca/patología , Envejecimiento/patología , Encéfalo/patología
3.
Dev Neurosci ; 44(4-5): 186-193, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35263745

RESUMEN

Perinatal hypoxic-ischemic (HI) brain damage has long been a major cause of acute mortality and chronic neurological morbidity in infants and children. Experimental animal models are essential to gain insights into the pathogenesis and management of perinatal HI brain damage. Prior to 1980, only large animal models were available. The first small animal model was developed in the postnatal 7 (P7) rat in 1981, now known as the Vannucci model. This model combines unilateral carotid artery ligation with subsequent hypoxia to produce transient hemispheric hypoxia-ischemia in the hemisphere ipsilateral to the ligation while the contralateral hemisphere is exposed to hypoxia only. This model has been characterized with studies of cerebral hemodynamics, cerebral metabolic changes, and acute and chronic neuropathology. Over the past 40 year, this animal model has been utilized in numerous laboratories around the world, has been adapted to the immature mouse, as well as to immature rodents at various stages of development. This brief review describes the validation and characterization studies of the original model and some of the adaptations. A discussion of all of the studies focused on specific cell types is beyond the scope of this review. Rather, we present the application of the model to the study of a specific cell type, the pre-oligodendrocyte, and the role this cell plays in the development of white matter injury in the preterm brain.


Asunto(s)
Hipoxia-Isquemia Encefálica , Roedores , Animales , Animales Recién Nacidos , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Hipoxia , Hipoxia-Isquemia Encefálica/patología , Isquemia , Ratones , Embarazo , Ratas
4.
J Neurosci ; 39(40): 7853-7871, 2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31455661

RESUMEN

Children who survive premature birth often exhibit reductions in hippocampal volumes and deficits in working memory. However, it is unclear whether synaptic plasticity and cellular mechanisms of learning and memory can be elicited or disrupted in the preterm fetal hippocampus. CA1 hippocampal neurons were exposed to two common insults to preterm brain: transient hypoxia-ischemia (HI) and hypoxia (Hx). We used a preterm fetal sheep model using both sexes in twin 0.65 gestation fetuses that reproduces the spectrum of injury and abnormal growth in preterm infants. Using Cavalieri measurements, hippocampal volumes were reduced in both Hx and HI fetuses compared with controls. This volume loss was not the result of neuronal cell death. Instead, morphometrics revealed alterations in both basal and apical dendritic arborization that were significantly associated with the level of systemic hypoxemia and metabolic stress regardless of etiology. Anatomical alterations of CA1 neurons were accompanied by reductions in probability of presynaptic glutamate release, long-term synaptic plasticity and intrinsic excitability. The reduction in intrinsic excitability was in part due to increased activity of the channels underlying the fast and slow component of the afterhyperpolarization in Hx and HI. Our studies suggest that even a single brief episode of hypoxemia can markedly disrupt hippocampal maturation. Hypoxemia may contribute to long-term working memory disturbances in preterm survivors by disrupting neuronal maturation with resultant functional disturbances in hippocampal action potential throughput. Strategies directed at limiting the duration or severity of hypoxemia during brain development may mitigate disturbances in hippocampal maturation.SIGNIFICANCE STATEMENT Premature infants commonly sustain hypoxia-ischemia, which results in reduced hippocampal growth and life-long disturbances in learning and memory. We demonstrate that the circuitry related to synaptic plasticity and cellular mechanisms of learning and memory (LTP) are already functional in the fetal hippocampus. Unlike adults, the fetal hippocampus is surprisingly resistant to cell death from hypoxia-ischemia. However, the hippocampus sustains robust structural and functional disturbances in the dendritic maturation of CA1 neurons that are significantly associated with the magnitude of a brief hypoxic stress. Since transient hypoxic episodes occur commonly in preterm survivors, our findings suggest that the learning problems that ensue may be related to the unique susceptibility of the hippocampus to brief episodes of hypoxemia.


Asunto(s)
Región CA1 Hipocampal/patología , Hipoxia/patología , Células Piramidales/patología , Ovinos/fisiología , Animales , Región CA1 Hipocampal/crecimiento & desarrollo , Dendritas/patología , Espinas Dendríticas/patología , Femenino , Desarrollo Fetal , Masculino , Memoria a Largo Plazo , Memoria a Corto Plazo , Plasticidad Neuronal , Embarazo , Nacimiento Prematuro , Estrés Fisiológico , Transmisión Sináptica
5.
Glia ; 68(2): 263-279, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31490574

RESUMEN

Myelination delay and remyelination failure following insults to the central nervous system (CNS) impede axonal conduction and lead to motor, sensory and cognitive impairments. Both myelination and remyelination are often inhibited or delayed due to the failure of oligodendrocyte progenitor cells (OPCs) to mature into myelinating oligodendrocytes (OLs). Digestion products of the glycosaminoglycan hyaluronan (HA) have been implicated in blocking OPC maturation, but how these digestion products are generated is unclear. We tested the possibility that hyaluronidase activity is directly linked to the inhibition of OPC maturation by developing a novel modified flavonoid that functions as a hyaluronidase inhibitor. This compound, called S3, blocks some but not all hyaluronidases and only inhibits matrix metalloproteinase activity at high concentrations. We find that S3 reverses HA-mediated inhibition of OPC maturation in vitro, an effect that can be overcome by excess recombinant hyaluronidase. Furthermore, we find that hyaluronidase inhibition by S3 accelerates OPC maturation in an in vitro model of perinatal white matter injury. Finally, blocking hyaluronidase activity with S3 promotes functional remyelination in mice with lysolecithin-induced demyelinating corpus callosum lesions. All together, these findings support the notion that hyaluronidase activity originating from OPCs in CNS lesions is sufficient to prevent OPC maturation, which delays myelination or blocks remyelination. These data also indicate that modified flavonoids can act as selective inhibitors of hyaluronidase activity and can promote OPC maturation, making them excellent candidates to accelerate myelination or promote remyelination following perinatal and adult CNS insults.


Asunto(s)
Enfermedades Desmielinizantes/patología , Células Precursoras de Oligodendrocitos/citología , Oligodendroglía/metabolismo , Remielinización/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Neurogénesis/fisiología , Células Madre/metabolismo
6.
Neurochem Res ; 45(3): 672-683, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31542857

RESUMEN

Although the extra cellular matrix (ECM) comprises a major proportion of the CNS parenchyma, new roles for the ECM in regeneration and repair responses to CNS injury have only recently been appreciated. The ECM undergoes extensive remodeling following injury to the developing or mature CNS in disorders that -include perinatal hypoxic-ischemic cerebral injury, multiple sclerosis and age-related vascular dementia. Here we focus on recently described mechanisms involving hyaluronan (HA), which negatively impact myelin repair after cerebral white matter injury. Injury induced depolymerization of hyaluronan (HA)-a component of the neural ECM-can inhibit myelin repair through the actions of specific sizes of HA fragments. These bioactive fragments selectively block the maturation of late oligodendrocyte progenitors via an immune tolerance-like pathway that suppresses pro-myelination signaling. We highlight emerging new pathophysiological roles of the neural ECM, particularly of those played by HA fragments (HAf) after injury and discuss strategies to promoter repair and regeneration of chronic myelination failure.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Matriz Extracelular/metabolismo , Homeostasis , Ácido Hialurónico/metabolismo , Sustancia Blanca/fisiopatología , Animales , Lesiones Encefálicas/metabolismo , Humanos , Transducción de Señal , Sustancia Blanca/lesiones , Sustancia Blanca/metabolismo
7.
Ann Neurol ; 83(1): 142-152, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29283444

RESUMEN

OBJECTIVE: Microvascular brain injury (mVBI) is a common pathological correlate of vascular contributions to cognitive impairment and dementia (VCID) that leads to white matter (WM) injury (WMI). VCID appears to arise from chronic recurrent white matter ischemia that triggers oxidative stress and an increase in total oligodendrocyte lineage cells. We hypothesized that mVBI involves vasodilator dysfunction of white matter penetrating arterioles and aberrant oligodendrocyte progenitor cell (OPC) responses to WMI. METHODS: We analyzed cases of mVBI with low Alzheimer's disease neuropathological change in prefrontal cortex WM from rapid autopsies in a population-based cohort where VCID frequently occurs. Arteriolar vasodilator function was quantified by videomicroscopy. OPC maturation was quantified using lineage specific markers. RESULTS: Acetylcholine-mediated arteriolar dilation in mVBI was significantly reduced in WM penetrators relative to pial arterioles. Astrogliosis-defined WMI was positively associated with increased OPCs and was negatively associated with decreased mature oligodendrocytes. INTERPRETATION: Selectively impaired vasodilator function of WM penetrating arterioles in mVBI occurs in association with aberrant differentiation of OPCs in WMI, which supports that myelination disturbances in VCID are related to disrupted maturation of myelinating oligodendrocytes. Ann Neurol 2018;83:142-152.


Asunto(s)
Envejecimiento/patología , Oligodendroglía/patología , Vasodilatación , Sustancia Blanca/patología , Acetilcolina/farmacología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Arteriolas/patología , Autopsia , Linaje de la Célula , Trastornos del Conocimiento/patología , Estudios de Cohortes , Femenino , Gliosis/patología , Humanos , Inmunohistoquímica , Masculino , Células-Madre Neurales/patología , Corteza Prefrontal/patología
8.
J Neurosci ; 37(49): 11912-11929, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-29089437

RESUMEN

Preterm infants are at risk for a broad spectrum of neurobehavioral disabilities associated with diffuse disturbances in cortical growth and development. During brain development, subplate neurons (SPNs) are a largely transient population that serves a critical role to establish functional cortical circuits. By dynamically integrating into developing cortical circuits, they assist in consolidation of intracortical and extracortical circuits. Although SPNs reside in close proximity to cerebral white matter, which is particularly vulnerable to oxidative stress, the susceptibility of SPNs remains controversial. We determined SPN responses to two common insults to the preterm brain: hypoxia-ischemia and hypoxia. We used a preterm fetal sheep model using both sexes that reproduces the spectrum of human cerebral injury and abnormal cortical growth. Unlike oligodendrocyte progenitors, SPNs displayed pronounced resistance to early or delayed cell death from hypoxia or hypoxia-ischemia. We thus explored an alternative hypothesis that these insults alter the maturational trajectory of SPNs. We used DiOlistic labeling to visualize the dendrites of SPNs selectively labeled for complexin-3. SPNs displayed reduced basal dendritic arbor complexity that was accompanied by chronic disturbances in SPN excitability and synaptic activity. SPN dysmaturation was significantly associated with the level of fetal hypoxemia and metabolic stress. Hence, despite the resistance of SPNs to insults that trigger white matter injury, transient hypoxemia disrupted SPN arborization and functional maturation during a critical window in cortical development. Strategies directed at limiting the duration or severity of hypoxemia during brain development may mitigate disturbances in cerebral growth and maturation related to SPN dysmaturation.SIGNIFICANCE STATEMENT The human preterm brain commonly sustains blood flow and oxygenation disturbances that impair cerebral cortex growth and cause life-long cognitive and learning disabilities. We investigated the fate of subplate neurons (SPNs), which are a master regulator of brain development that plays critical roles in establishing cortical connections to other brain regions. We used a preterm fetal sheep model that reproduces key features of brain injury in human preterm survivors. We analyzed the responses of fetal SPNs to transient disturbances in fetal oxygenation. We discovered that SPNs are surprisingly resistant to cell death from low oxygen states but acquire chronic structural and functional changes that suggest new strategies to prevent learning problems in children and adults that survive preterm birth.


Asunto(s)
Hipoxia/patología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Dendritas/fisiología , Femenino , Hipoxia/complicaciones , Masculino , Degeneración Nerviosa/etiología , Degeneración Nerviosa/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Ovinos , Factores de Tiempo
9.
Acta Neuropathol ; 134(3): 331-349, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28534077

RESUMEN

The human preterm brain is particularly susceptible to cerebral white matter injury (WMI) that disrupts the normal progression of developmental myelination. Advances in the care of preterm infants have resulted in a sustained reduction in the severity of WMI that has shifted from more severe focal necrotic lesions to milder diffuse WMI. Nevertheless, WMI remains a global health problem and the most common cause of chronic neurological morbidity from cerebral palsy and diverse neurobehavioral disabilities. Diffuse WMI involves maturation-dependent vulnerability of the oligodendrocyte (OL) lineage with selective degeneration of late oligodendrocyte progenitors (preOLs) triggered by oxidative stress and other insults. The magnitude and distribution of diffuse WMI are related to both the timing of appearance and regional distribution of susceptible preOLs. Diffuse WMI disrupts the normal progression of OL lineage maturation and myelination through aberrant mechanisms of regeneration and repair. PreOL degeneration is accompanied by early robust proliferation of OL progenitors that regenerate and augment the preOL pool available to generate myelinating OLs. However, newly generated preOLs fail to differentiate and initiate myelination along their normal developmental trajectory despite the presence of numerous intact-appearing axons. Disrupted preOL maturation is accompanied by diffuse gliosis and disturbances in the composition of the extracellular matrix and is mediated in part by inhibitory factors derived from reactive astrocytes. Signaling pathways implicated in disrupted myelination include those mediated by Notch, WNT-beta catenin, and hyaluronan. Hence, there exists a potentially broad but still poorly defined developmental window for interventions to promote white matter repair and myelination and potentially reverses the widespread disturbances in cerebral gray matter growth that accompanies WMI.


Asunto(s)
Encéfalo/patología , Leucomalacia Periventricular/patología , Fibras Nerviosas Mielínicas/patología , Sustancia Blanca/patología , Humanos , Recién Nacido , Recien Nacido Prematuro , Neuroglía/patología
10.
Neurobiol Dis ; 92(Pt A): 90-101, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26477300

RESUMEN

In this review, we highlight critical unresolved questions in the etiology and mechanisms causing preterm brain injury. Involvement of neurons, glia, endogenous factors and exogenous exposures is considered. The structural and functional correlates of interrupted development and injury in the premature brain are under active investigation, with the hope that the cellular and molecular mechanisms underlying developmental abnormalities in the human preterm brain can be understood, prevented or repaired.


Asunto(s)
Lesiones Encefálicas/embriología , Lesiones Encefálicas/fisiopatología , Encéfalo/embriología , Encéfalo/fisiopatología , Animales , Encéfalo/diagnóstico por imagen , Lesiones Encefálicas/diagnóstico por imagen , Humanos
11.
Mol Genet Metab ; 114(4): 527-36, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25684057

RESUMEN

Leukodystrophies are a heterogeneous, often progressive group of disorders manifesting a wide range of symptoms and complications. Most of these disorders have historically had no etiologic or disease specific therapeutic approaches. Recently, a greater understanding of the pathologic mechanisms associated with leukodystrophies has allowed clinicians and researchers to prioritize treatment strategies and advance research in therapies for specific disorders, some of which are on the verge of pilot or Phase I/II clinical trials. This shifts the care of leukodystrophy patients from the management of the complex array of symptoms and sequelae alone to targeted therapeutics. The unmet needs of leukodystrophy patients still remain an overwhelming burden. While the overwhelming consensus is that these disorders collectively are symptomatically treatable, leukodystrophy patients are in need of advanced therapies and if possible, a cure.


Asunto(s)
Enfermedades Desmielinizantes/terapia , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/terapia , Leucodistrofia Metacromática/terapia , Leucoencefalopatías/terapia , Encefalopatías/prevención & control , Encefalopatías/terapia , Enfermedades Desmielinizantes/prevención & control , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/prevención & control , Humanos , Leucodistrofia Metacromática/prevención & control , Leucoencefalopatías/prevención & control
12.
Ann Neurol ; 75(4): 469-86, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24615937

RESUMEN

With advances in neonatal care, preterm neonates are surviving with an evolving constellation of motor and cognitive disabilities that appear to be related to widespread cellular maturational disturbances that target cerebral gray and white matter. Whereas preterm infants were previously at high risk for destructive brain lesions that resulted in cystic white matter injury and secondary cortical and subcortical gray matter degeneration, contemporary cohorts of preterm survivors commonly display less severe injury that does not appear to involve pronounced glial or neuronal loss. Nevertheless, these milder forms of injury are also associated with reduced cerebral growth. Recent human and experimental studies support that impaired cerebral growth is related to disparate responses in gray and white matter. Myelination disturbances in cerebral white matter are related to aberrant regeneration and repair responses to acute death of premyelinating late oligodendrocyte progenitors (preOLs). In response to preOL death, early oligodendrocyte progenitors rapidly proliferate and differentiate, but the regenerated preOLs fail to normally mature to myelinating cells required for white matter growth. Although immature neurons appear to be more resistant to cell death from hypoxia-ischemia than glia, they display widespread disturbances in maturation of their dendritic arbors, which further contribute to impaired cerebral growth. These complex and disparate responses of neurons and preOLs thus result in large numbers of cells that fail to fully mature during a critical window in development of neural circuitry. These recently recognized forms of cerebral gray and white matter dysmaturation raise new diagnostic challenges and suggest new therapeutic directions centered on reversal of the processes that promote dysmaturation.


Asunto(s)
Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Discapacidades del Desarrollo/etiología , Recien Nacido Prematuro , Degeneración Nerviosa/etiología , Trastornos del Conocimiento/etiología , Humanos , Recién Nacido , Fibras Nerviosas Mielínicas/patología
13.
Ann Neurol ; 75(4): 533-41, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24633673

RESUMEN

OBJECTIVE: White matter (WM) injury due to myelination defects is believed to be responsible for the motor deficits seen in cerebral palsy. We tested the hypothesis that the predominant injury is to functional electrical connectivity in unmyelinated WM fibers by conducting a longitudinal study of central WM tracts in newborn rabbit kits with hypertonia in our model of cerebral palsy. METHODS: Pregnant rabbits at 70% gestation underwent 40-minute uterine ischemia. Motor deficits in newborn kits, including muscle hypertonia, were assessed by neurobehavioral testing. Major central WM tracts, including internal capsule, corpus callosum, anterior commissure, and fimbria hippocampi, were investigated for structural and functional injury using diffusion tensor magnetic resonance imaging (MRI), electrophysiological recordings of fiber conductivity in perfused brain slices, electron microscopy, and immunohistochemistry of oligodendrocyte lineage. RESULTS: Motor deficits were observed on postnatal day 1 (P1) when WM tracts were unmyelinated. Myelination occurred later and was obvious by P18. Hypertonia was associated with microstructural WM injury and unmyelinated axon loss at P1, diagnosed by diffusion tensor MRI and electron microscopy. Axonal conductivity from electrophysiological recordings in hypertonic P18 kits decreased only in unmyelinated fibers, despite a loss in both myelinated and unmyelinated axons. INTERPRETATION: Motor deficits in cerebral palsy were associated with loss of unmyelinated WM tracts. The contribution of injury to myelinated fibers that was observed at P18 is probably a secondary etiological factor in the motor and sensory deficits in the rabbit model of cerebral palsy.


Asunto(s)
Hipoxia Fetal/complicaciones , Trastornos del Movimiento/etiología , Hipertonía Muscular/etiología , Fibras Nerviosas Amielínicas/patología , Animales , Animales Recién Nacidos , Imagen de Difusión Tensora , Electroencefalografía , Potenciales Evocados/fisiología , Femenino , Leucoencefalopatías/etiología , Leucoencefalopatías/patología , Microscopía Electrónica de Transmisión , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Embarazo , Conejos
14.
Ann Neurol ; 75(4): 508-24, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24395459

RESUMEN

OBJECTIVE: Recently, we reported that the neocortex displays impaired growth after transient cerebral hypoxia-ischemia (HI) at preterm gestation that is unrelated to neuronal death but is associated with decreased dendritic arbor complexity of cortical projection neurons. We hypothesized that these morphological changes constituted part of a more widespread neuronal dysmaturation response to HI in the caudate nucleus (CN), which contributes to motor and cognitive disability in preterm survivors. METHODS: Ex vivo magnetic resonance imaging (MRI), immunohistochemistry, and Golgi staining defined CN growth, cell death, proliferation, and dendritic maturation in preterm fetal sheep 4 weeks after HI. Patch-clamp recording was used to analyze glutamatergic synaptic currents in CN neurons. RESULTS: MRI-defined growth of the CN was reduced after ischemia compared to controls. However, no significant acute or delayed neuronal death was seen in the CN or white matter. Nor was there significant loss of calbindin-positive medium spiny projection neurons (MSNs) or CN interneurons expressing somatostatin, calretinin, parvalbumin, or tyrosine hydroxylase. Morphologically, ischemic MSNs showed a markedly immature dendritic arbor, with fewer dendritic branches, nodes, endings, and spines. The magnitude and kinetics of synaptic currents, and the relative contribution of glutamate receptor subtypes in the CN were significantly altered. INTERPRETATION: The marked MSN dendritic and functional abnormalities after preterm cerebral HI, despite the marked resistance of immature CN neurons to cell death, are consistent with widespread susceptibility of projection neurons to HI-induced dysmaturation. These global disturbances in dendritic maturation and glutamatergic synaptic transmission suggest a new mechanism for long-term motor and behavioral disabilities in preterm survivors via widespread disruption of neuronal connectivity.


Asunto(s)
Isquemia Encefálica/patología , Núcleo Caudado/patología , Hipoxia Fetal/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/patología , Nacimiento Prematuro/fisiopatología , Potenciales de Acción/efectos de los fármacos , Animales , Isquemia Encefálica/sangre , Caspasa 3/metabolismo , Dendritas/patología , Dendritas/ultraestructura , Modelos Animales de Enfermedad , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , GABAérgicos/farmacología , Cabras , Antígeno Ki-67/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/ultraestructura , Embarazo , Factores de Tiempo
15.
Glia ; 62(11): 1790-815, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24687630

RESUMEN

Injury to the preterm brain has a particular predilection for cerebral white matter. White matter injury (WMI) is the most common cause of brain injury in preterm infants and a major cause of chronic neurological morbidity including cerebral palsy. Factors that predispose to WMI include cerebral oxygenation disturbances and maternal-fetal infection. During the acute phase of WMI, pronounced oxidative damage occurs that targets late oligodendrocyte progenitors (pre-OLs). The developmental predilection for WMI to occur during prematurity appears to be related to both the timing of appearance and regional distribution of susceptible pre-OLs that are vulnerable to a variety of chemical mediators including reactive oxygen species, glutamate, cytokines, and adenosine. During the chronic phase of WMI, the white matter displays abberant regeneration and repair responses. Early OL progenitors respond to WMI with a rapid robust proliferative response that results in a several fold regeneration of pre-OLs that fail to terminally differentiate along their normal developmental time course. Pre-OL maturation arrest appears to be related in part to inhibitory factors that derive from reactive astrocytes in chronic lesions. Recent high field magnetic resonance imaging (MRI) data support that three distinct forms of chronic WMI exist, each of which displays unique MRI and histopathological features. These findings suggest the possibility that therapies directed at myelin regeneration and repair could be initiated early after WMI and monitored over time. These new mechanisms of acute and chronic WMI provide access to a variety of new strategies to prevent or promote repair of WMI in premature infants.


Asunto(s)
Leucoencefalopatías/etiología , Leucoencefalopatías/patología , Neuroglía/fisiología , Nacimiento Prematuro/fisiopatología , Animales , Humanos , Vaina de Mielina/patología , Neuroglía/patología , Estrés Oxidativo/fisiología , Receptores de Glutamato/metabolismo
16.
Ann Neurol ; 73(2): 266-80, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23463525

RESUMEN

OBJECTIVE: Oligodendrocyte progenitor cells (OPCs) recruited to demyelinating lesions often fail to mature into oligodendrocytes (OLs) that remyelinate spared axons. The glycosaminoglycan hyaluronan (HA) accumulates in demyelinating lesions and has been implicated in the failure of OPC maturation and remyelination. We tested the hypothesis that OPCs in demyelinating lesions express a specific hyaluronidase, and that digestion products of this enzyme inhibit OPC maturation. METHODS: Mouse OPCs grown in vitro were analyzed for hyaluronidase expression and activity. Gain of function studies were used to define the hyaluronidases that blocked OPC maturation. Mouse and human demyelinating lesions were assessed for hyaluronidase expression. Digestion products from different hyaluronidases and a hyaluronidase inhibitor were tested for their effects on OPC maturation and functional remyelination in vivo. RESULTS: OPCs demonstrated hyaluronidase activity in vitro and expressed multiple hyaluronidases, including HYAL1, HYAL2, and PH20. HA digestion by PH20 but not other hyaluronidases inhibited OPC maturation into OLs. In contrast, inhibiting HA synthesis did not influence OPC maturation. PH20 expression was elevated in OPCs and reactive astrocytes in both rodent and human demyelinating lesions. HA digestion products generated by the PH20 hyaluronidase but not another hyaluronidase inhibited remyelination following lysolecithin-induced demyelination. Inhibition of hyaluronidase activity lead to increased OPC maturation and promoted increased conduction velocities through lesions. INTERPRETATION: We determined that PH20 is elevated in demyelinating lesions and that increased PH20 expression is sufficient to inhibit OPC maturation and remyelination. Pharmacological inhibition of PH20 may therefore be an effective way to promote remyelination in multiple sclerosis and related conditions.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Hialuronoglucosaminidasa/metabolismo , Regeneración Nerviosa/fisiología , Células-Madre Neurales/enzimología , Oligodendroglía/citología , Potenciales de Acción/fisiología , Animales , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inducido químicamente , Inhibidores Enzimáticos/farmacología , Femenino , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/antagonistas & inhibidores , Hialuronoglucosaminidasa/genética , Lisofosfatidilcolinas/toxicidad , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Oligodendroglía/fisiología
17.
Anesthesiology ; 120(3): 626-38, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24158051

RESUMEN

BACKGROUND: The authors have previously shown that exposure of the neonatal nonhuman primate (NHP) brain to isoflurane for 5 h causes widespread acute apoptotic degeneration of neurons and oligodendrocyte. The current study explored the potential apoptogenic action of isoflurane in the fetal NHP brain. METHODS: Fetal rhesus macaques at gestational age of 120 days (G120) were exposed in utero for 5 h to isoflurane anesthesia (n = 5) or to no anesthesia (control condition; n = 4), and all regions of the brain were systematically evaluated 3 h later for evidence of apoptotic degeneration of neurons or glia. RESULTS: Exposure of the G120 fetal NHP brain to isoflurane caused a significant increase in apoptosis of neurons and of oligodendrocytes at a stage when oligodendrocytes were just beginning to myelinate axons. The neuroapoptosis response was most prominent in the cerebellum, caudate, putamen, amygdala, and several cerebrocortical regions. Oligodendrocyte apoptosis was diffusely distributed over many white matter regions. The total number of apoptotic profiles (neurons + oligodendrocytes) in the isoflurane-exposed brains was increased 4.1-fold, compared with the brains from drug-naive controls. The total number of oligodendrocytes deleted by isoflurane was higher than the number of neurons deleted. CONCLUSIONS: Isoflurane anesthesia for 5 h causes death of neurons and oligodendrocytes in the G120 fetal NHP brain. In the fetal brain, as the authors previously found in the neonatal NHP brain, oligodendrocytes become vulnerable when they are just achieving myelination competence. The neurotoxic potential of isoflurane increases between the third trimester (G120) and the neonatal period in the NHP brain.


Asunto(s)
Anestésicos por Inhalación/toxicidad , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Isoflurano/toxicidad , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Animales , Animales Recién Nacidos , Encéfalo/embriología , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Macaca mulatta , Neuronas/patología , Oligodendroglía/patología
18.
Pediatr Res ; 75(1-2): 227-33, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24336432

RESUMEN

Children surviving premature birth have a high risk of cognitive and learning disabilities and attention deficit. In turn, adverse outcomes are associated with persistent reductions in cerebral growth on magnetic resonance imaging (MRI). It is striking that modern care has been associated with a dramatic reduction in the risk of cystic white matter damage, but modest improvements in terms of neurodevelopmental impairment. This review will explore the hypothesis that the disability is primarily associated with impaired neural connectivity rather than cell death alone. Very preterm infants exhibit reduced thalamocortical connectivity and cortical neuroplasticity compared with term-born controls. In preterm fetal sheep, moderate cerebral ischemia with no neuronal loss, but significant diffuse failure of maturation of cortical pyramidal neurons, was associated with impaired dendritic growth and synapse formation, consistent with altered connectivity. These changes were associated with delayed decline in cortical fractional anisotropy (FA) on MRI. Supporting these preclinical findings, preterm human survivors showed similar enduring impairment of microstructural development of the cerebral cortex defined by FA, consistent with delayed formation of neuronal processes. These findings offer the promise that better understanding of impairment of neural connectivity may allow us to promote normal development and growth of the cortex after preterm birth.


Asunto(s)
Encéfalo/fisiopatología , Desarrollo Infantil , Cognición , Discapacidades del Desarrollo/etiología , Recien Nacido Prematuro , Factores de Edad , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/fisiopatología , Discapacidades del Desarrollo/psicología , Edad Gestacional , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Vías Nerviosas/fisiopatología , Plasticidad Neuronal , Factores de Riesgo , Factores de Tiempo
19.
Ann Neurol ; 72(6): 936-51, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23280842

RESUMEN

OBJECTIVE: Developing central white matter is subject to ischemic-type injury during the period that precedes myelination. At this stage in maturation, central axons initiate a program of radial expansion and ion channel redistribution. Here we test the hypothesis that during radial expansion axons display heightened ischemic sensitivity, when clusters of Ca(2+) channels decorate future node of Ranvier sites. METHODS: Functionality and morphology of central axons and glia were examined during and after a period of modeled ischemia. Pathological changes in axons undergoing radial expansion were probed using electrophysiological, quantitative ultrastructural, and morphometric analysis in neonatal rodent optic nerve and periventricular white matter axons studied under modeled ischemia in vitro or after hypoxia-ischemia in vivo. RESULTS: Acute ischemic injury of central axons undergoing initial radial expansion was mediated by Ca(2+) influx through Ca(2+) channels expressed in axolemma clusters. This form of injury operated only in this axon population, which was more sensitive to injury than neighboring myelinated axons, smaller axons yet to initiate radial expansion, astrocytes, or oligodendroglia. A pharmacological strategy designed to protect both small and large diameter premyelinated axons proved 100% protective against acute ischemia studied under modeled ischemia in vitro or after hypoxia-ischemia in vivo. INTERPRETATION: Recent clinical data highlight the importance of axon pathology in developing white matter injury. The elevated susceptibility of early maturing axons to ischemic injury described here may significantly contribute to selective white matter pathology and places these axons alongside preoligodendrocytes as a potential primary target of both injury and therapeutics.


Asunto(s)
Axones/metabolismo , Hipoxia-Isquemia Encefálica/patología , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/patología , Nervio Óptico/patología , Factores de Edad , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Astrocitos/efectos de los fármacos , Astrocitos/patología , Axones/efectos de los fármacos , Axones/ultraestructura , Modelos Animales de Enfermedad , Maleato de Dizocilpina/uso terapéutico , Glucosa/deficiencia , Proteínas Fluorescentes Verdes/genética , Hipoxia/patología , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Ratones , Ratones Transgénicos , Degeneración Nerviosa/etiología , Degeneración Nerviosa/prevención & control , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/ultraestructura , Fármacos Neuroprotectores/uso terapéutico , Oligodendroglía/metabolismo , Oligodendroglía/patología , Oligodendroglía/ultraestructura , Nervio Óptico/crecimiento & desarrollo , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Antígenos Thy-1/genética , omega-Agatoxina IVA/uso terapéutico
20.
Ann Neurol ; 71(1): 93-109, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22275256

RESUMEN

OBJECTIVE: The major form of magnetic resonance imaging-defined white matter injury (WMI) comprises diffuse lesions where the burden of small necrotic foci (microscopic necrosis) is poorly defined. We hypothesized that myelination failure associated with diffuse WMI involves an aberrant injury response linked to arrested preoligodendrocyte (preOL) maturation in reactive astrocyte-rich lesions. METHODS: A retrospective autopsy series (1983-2000) was selected for cases with diffuse WMI and analyzed relative to prospectively collected contemporary cases (2003-2010). Controls were age- and region-matched to address regional variation in preOL maturation. Successive oligodendrocyte stages were analyzed with lineage-specific markers. Microscopic necrosis was quantified with microglial markers. Axon injury markers defined the burden of axonopathy. Extracellular matrix remodeling was defined by detection of hyaluronic acid (HA), an inhibitor of preOL maturation, and the HA receptor, CD44. RESULTS: In the contemporary case series, diffuse WMI was accompanied by a significant reduction in the burden of microscopic necrosis and axonopathy. Diffuse astrogliosis extended into the lesion surround with elevated HA and astrocyte-expressed CD44. The total population of OL lineage stages was significantly increased in lesions. This increase coincided with significant expansion of the preOL pool. INTERPRETATION: Although these data confirm that microscopic necrosis occurs in contemporary cases, the markedly decreased burden supports that it does not contribute substantially to myelination failure. The primary mechanism of myelination failure involves a disrupted cellular response whereby preOLs fail to differentiate in diffuse astrogliotic lesions. PreOL maturation arrest converts chronic WMI to a more immature state related to the burden of astrogliosis.


Asunto(s)
Proliferación Celular , Enfermedades del Prematuro/patología , Vaina de Mielina/patología , Oligodendroglía/patología , Células Madre/patología , Astrocitos/patología , Diferenciación Celular/fisiología , Estudios de Cohortes , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Leucoencefalopatías/patología , Masculino , Necrosis , Fibras Nerviosas Mielínicas/patología , Estudios Prospectivos , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA