Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
2.
Int J Mol Sci ; 25(3)2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38338726

RESUMEN

Phenobarbital (PB) remains the first-line medication for neonatal seizures. Yet, seizures in many newborns, particularly those associated with perinatal ischemia, are resistant to PB. Previous animal studies have shown that in postnatal day P7 mice pups with ischemic stroke induced by unilateral carotid ligation, the tyrosine receptor kinase B (TrkB) antagonist ANA12 (N-[2-[[(hexahydro-2-oxo-1H-azepin-3-yl)amino]carbonyl]phenyl]-benzo[b]thiophene-2-carboxamide, 5 mg/kg) improved the efficacy of PB in reducing seizure occurrence. To meet optimal standards of effectiveness, a wider range of ANA12 doses must be tested. Here, using the unilateral carotid ligation model, we tested the effectiveness of higher doses of ANA12 (10 and 20 mg/kg) on the ability of PB to reduce seizure burden, ameliorate cell death (assessed by Fluoro-Jade staining), and affect neurodevelopment (righting reflex, negative geotaxis test, open field test). We found that a single dose of ANA12 (10 or 20 mg/kg) given 1 h after unilateral carotid ligation in P7 pups reduced seizure burden and neocortical and striatal neuron death without impairing developmental reflexes. In conclusion, ANA12 at a range of doses (10-20 mg/kg) enhanced PB effectiveness for the treatment of perinatal ischemia-related seizures, suggesting that this agent might be a clinically safe and effective adjunctive agent for the treatment of pharmacoresistant neonatal seizures.


Asunto(s)
Epilepsia , Hipoxia-Isquemia Encefálica , Animales , Ratones , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Animales Recién Nacidos , Modelos Animales de Enfermedad , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Convulsiones/metabolismo , Fenobarbital/farmacología , Fenobarbital/uso terapéutico , Epilepsia/tratamiento farmacológico , Isquemia/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico
3.
J Neurochem ; 168(5): 910-954, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38183680

RESUMEN

Although we have learned much about how the brain fuels its functions over the last decades, there remains much still to discover in an organ that is so complex. This article lays out major gaps in our knowledge of interrelationships between brain metabolism and brain function, including biochemical, cellular, and subcellular aspects of functional metabolism and its imaging in adult brain, as well as during development, aging, and disease. The focus is on unknowns in metabolism of major brain substrates and associated transporters, the roles of insulin and of lipid droplets, the emerging role of metabolism in microglia, mysteries about the major brain cofactor and signaling molecule NAD+, as well as unsolved problems underlying brain metabolism in pathologies such as traumatic brain injury, epilepsy, and metabolic downregulation during hibernation. It describes our current level of understanding of these facets of brain energy metabolism as well as a roadmap for future research.


Asunto(s)
Encéfalo , Metabolismo Energético , Animales , Humanos , Encéfalo/metabolismo
4.
Physiol Rep ; 11(7): e15656, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37038896

RESUMEN

Hypoxia in the neonatal period is associated with early manifestations of adverse cardiovascular health in adulthood including higher risk of hypertension and atherosclerosis. We hypothesize that this occurs due to activation of lysyl oxidases (LOXs) and the remodeling of the large conduit vessels, leading to early arterial stiffening. Newborn C57Bl/6 mice were exposed to hypoxia (FiO2  = 11.5%) from postnatal day 1 (P1) to postnatal day 11 (P11), followed by resumption of normoxia. Controls were maintained in normoxia. Using in vivo (pulse wave velocity; PWV) and ex vivo (tensile testing) arterial stiffness indexes, we determined that mice exposed to neonatal hypoxia had significantly higher arterial stiffness compared with normoxia controls by young adulthood (P60), and it increased further by P120. Echocardiography performed at P60 showed that mice exposed to hypoxia displayed a compensated dilated cardiomyopathy. Western blotting revelated that neonatal hypoxia accelerated age-related increase in LOXL2 protein expression in the aorta and elevated LOXL2 expression in the PA at P11 with a delayed decay toward normoxic controls. In the heart and lung, gene and protein expression of LOX/LOXL2 were upregulated at P11, with a delayed decay when compared to normoxic controls. Neonatal hypoxia results in a significant increase in arterial stiffness in early adulthood due to aberrant LOX/LOXL2 expression. This suggests an acceleration in the mechanical decline of the cardiovascular system, that contributes to increased risk of hypertension in young adults exposed to neonatal hypoxia that may increase susceptibility to further insults.


Asunto(s)
Hipertensión , Rigidez Vascular , Ratones , Animales , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , Análisis de la Onda del Pulso , Hipoxia , Aorta/metabolismo , Rigidez Vascular/fisiología
6.
J Neurosci ; 42(24): 4812-4827, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35589394

RESUMEN

Neonatal brain injury renders the developing brain vulnerable to oxidative stress, leading to cognitive deficit. However, oxidative stress-induced damage to hippocampal circuits and the mechanisms underlying long-term changes in memory and learning are poorly understood. We used high oxygen tension or hyperoxia (HO) in neonatal mice of both sexes to investigate the role of oxidative stress in hippocampal damage. Perinatal HO induces reactive oxygen species and cell death, together with reduced interneuron maturation, inhibitory postsynaptic currents, and dentate progenitor proliferation. Postinjury interneuron stimulation surprisingly improved inhibitory activity and memory tasks, indicating reversibility. With decreased hippocampal levels of Wnt signaling components and somatostatin, HO aberrantly activated glycogen synthase kinase 3 ß activity. Pharmacological inhibition or ablation of interneuron glycogen synthase kinase 3 ß during HO challenge restored progenitor cell proliferation, interneuron development, inhibitory/excitatory balance, as well as hippocampal-dependent behavior. Biochemical targeting of interneuron function may benefit learning deficits caused by oxidative damage.SIGNIFICANCE STATEMENT Premature infants are especially vulnerable to oxidative stress, as their antioxidant defenses are underdeveloped. Indeed, high oxygen tension is associated with poor neurologic outcomes. Because of its sustained postnatal development and role in learning and memory, the hippocampus is especially vulnerable to oxidative damage in premature infants. However, the role of oxidative stress in the developing hippocampus has yet to be explored. With ever-rising rates of neonatal brain injury and no universally viable approach to maximize functional recovery, a better understanding of the mechanisms underlying neonatal brain injury is needed. Addressing this need, this study uses perinatal hyperoxia to study cognitive deficits, pathophysiology, and molecular mechanisms of oxidative damage in the developing hippocampus.


Asunto(s)
Lesiones Encefálicas , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/metabolismo , Hiperoxia , Estrés Oxidativo , Animales , Femenino , Hipocampo/crecimiento & desarrollo , Humanos , Hiperoxia/metabolismo , Masculino , Ratones , Oxígeno/metabolismo , Embarazo
7.
Dev Neurosci ; 44(4-5): 266-276, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35358965

RESUMEN

Cerebral palsy (CP) is the most common cause of physical disability for children worldwide. Many infants and toddlers are not diagnosed with CP until they fail to achieve obvious motor milestones. Currently, there are no effective pharmacologic interventions available for infants and toddlers to substantially improve their trajectory of neurodevelopment. Because children with CP from preterm birth also exhibit a sustained immune system hyper-reactivity, we hypothesized that neuro-immunomodulation with a regimen of repurposed endogenous neurorestorative medications, erythropoietin (EPO) and melatonin (MLT), could improve this trajectory. Thus, we administered EPO + MLT to rats with CP during human infant-toddler equivalency to determine whether we could influence gait patterns in mature animals. After a prenatal injury on embryonic day 18 (E18) that mimics chorioamnionitis at ∼25 weeks human gestation, rat pups were born and raised with their dam. Beginning on postnatal day 15 (P15), equivalent to human infant ∼1 year, rats were randomized to receive either a regimen of EPO + MLT or vehicle (sterile saline) through P20. Gait was assessed in young adult rats at P30 using computerized digital gait analyses including videography on a treadmill. Results indicate that gait metrics of young adult rats treated with an infantile cocktail of EPO + MLT were restored compared to vehicle-treated rats (p < 0.05) and similar to sham controls. These results provide reassuring evidence that pharmacological interventions may be beneficial to infants and toddlers who are diagnosed with CP well after the traditional neonatal window of intervention.


Asunto(s)
Lesiones Encefálicas , Eritropoyetina , Melatonina , Nacimiento Prematuro , Animales , Lesiones Encefálicas/tratamiento farmacológico , Eritropoyetina/farmacología , Femenino , Marcha , Humanos , Lactante , Melatonina/farmacología , Embarazo , Ratas
8.
NMR Biomed ; 34(2): e4451, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33258202

RESUMEN

The study of cerebral metabolites relies heavily on detection methods and sample preparation. Animal experiments in vivo require anesthetic agents that can alter brain metabolism, whereas ex vivo experiments demand appropriate fixation methods to preserve the tissue from rapid postmortem degradation. In this study, the metabolic profiles of mouse hippocampi using proton magnetic resonance spectroscopy (1 H-MRS) were compared in vivo and in situ with or without focused beam microwave irradiation (FBMI) fixation. Ten major brain metabolites, including lactate (Lac), N-acetylaspartate (NAA), total choline (tCho), myo-inositol (mIns), glutamine (Gln), glutamate (Glu), aminobutyric acid (GABA), glutathione (GSH), total creatine (tCr) and taurine (Tau), were analyzed using LCModel. After FBMI fixation, the concentrations of Lac, tCho and mIns were comparable with those obtained in vivo under isoflurane, whereas other metabolites were significantly lower. Except for a decrease in NAA and an increase in Tau, all the other metabolites remained stable over 41 hours in FBMI-fixed brains. Without FBMI, the concentrations of mIns (before 2 hours), tCho and GABA were close to those measured in vivo. However, higher Lac (P < .01) and lower NAA, Gln, Glu, GSH, tCr and Tau were observed (P < .01). NAA, Gln, Glu, GSH, tCr and Tau exhibited good temporal stability for at least 20 hours in the unfixed brain, whereas a linear increase of tCho, mIns and GABA was observed. Possible mechanisms of postmortem degradation are discussed. Our results indicate that a proper fixation method is required for in situ detection depending on the targeted metabolites of specific interests in the brain.


Asunto(s)
Hipocampo/diagnóstico por imagen , Espectroscopía de Resonancia Magnética/métodos , Neuroimagen/métodos , Espectroscopía de Protones por Resonancia Magnética/métodos , Animales , Agua Corporal , Femenino , Hipocampo/metabolismo , Lípidos/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Microondas , Cambios Post Mortem , Distribución Aleatoria , Fijación del Tejido/métodos
9.
Nat Commun ; 11(1): 964, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32075970

RESUMEN

Hypoxic damage to the developing brain due to preterm birth causes many anatomical changes, including damage to the periventricular white matter. This results in the loss of glial cells, significant disruptions in myelination, and thereby cognitive and behavioral disabilities seen throughout life. Encouragingly, these neurological morbidities can be improved by environmental factors; however, the underlying cellular mechanisms remain unknown. We found that early and continuous environmental enrichment selectively enhances endogenous repair of the developing white matter by promoting oligodendroglial maturation, myelination, and functional recovery after perinatal brain injury. These effects require increased exposure to socialization, physical activity, and cognitive enhancement of surroundings-a complete enriched environment. Using RNA-sequencing, we identified oligodendroglial-specific responses to hypoxic brain injury, and uncovered molecular mechanisms involved in enrichment-induced recovery. Together, these results indicate that myelin plasticity induced by modulation of the neonatal environment can be targeted as a therapeutic strategy for preterm birth.


Asunto(s)
Lesiones Encefálicas/rehabilitación , Ambiente , Neuroprotección , Sustancia Blanca/fisiología , Animales , Animales Recién Nacidos , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Hipoxia/patología , Hipoxia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Vaina de Mielina/fisiología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Oligodendroglía/fisiología , RNA-Seq , Recuperación de la Función , Sustancia Blanca/citología , Sustancia Blanca/lesiones , Sustancia Blanca/metabolismo
10.
Exp Neurol ; 316: 74-84, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30951705

RESUMEN

Improved patient survival following pediatric traumatic brain injury (TBI) has uncovered a currently limited understanding of both the adaptive and maladaptive metabolic perturbations that occur during the acute and long-term phases of recovery. While much is known about the redundancy of metabolic pathways that provide adequate energy and substrates for normal brain growth and development, the field is only beginning to characterize perturbations in these metabolic pathways after pediatric TBI. To date, the majority of studies have focused on dysregulated oxidative glucose metabolism after injury; however, the immature brain is well-equipped to use alternative substrates to fuel energy production, growth, and development. A comprehensive understanding of metabolic changes associated with pediatric TBI cannot be limited to investigations of glucose metabolism alone. All energy substrates used by the brain should be considered in developing nutritional and pharmacological interventions for pediatric head trauma. This review summarizes post-injury changes in brain metabolism of glucose, lipids, ketone bodies, and amino acids with discussion of the therapeutic potential of altering substrate utilization to improve pediatric TBI outcomes.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Glucosa/metabolismo , Adolescente , Aminoácidos/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/patología , Niño , Preescolar , Metabolismo Energético , Humanos , Lactante , Recién Nacido , Cuerpos Cetónicos/metabolismo , Metabolismo de los Lípidos
11.
Nat Rev Neurosci ; 20(5): 298-313, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30923348

RESUMEN

The human cerebellum has a protracted developmental timeline compared with the neocortex, expanding the window of vulnerability to neurological disorders. As the cerebellum is critical for motor behaviour, it is not surprising that most neurodevelopmental disorders share motor deficits as a common sequela. However, evidence gathered since the late 1980s suggests that the cerebellum is involved in motor and non-motor function, including cognition and emotion. More recently, evidence indicates that major neurodevelopmental disorders such as intellectual disability, autism spectrum disorder, attention-deficit hyperactivity disorder and Down syndrome have potential links to abnormal cerebellar development. Out of recent findings from clinical and preclinical studies, the concept of the 'cerebellar connectome' has emerged that can be used as a framework to link the role of cerebellar development to human behaviour, disease states and the design of better therapeutic strategies.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Cerebelo/fisiopatología , Conectoma , Red Nerviosa/fisiopatología , Trastornos del Neurodesarrollo/fisiopatología , Animales , Humanos
12.
Cancer Res ; 78(8): 2081-2095, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29559476

RESUMEN

Newly developed targeted anticancer drugs inhibit signaling pathways commonly altered in adult and pediatric cancers. However, as these pathways are also essential for normal brain development, concerns have emerged of neurologic sequelae resulting specifically from their application in pediatric cancers. The neural substrates and age dependency of these drug-induced effects in vivo are unknown, and their long-term behavioral consequences have not been characterized. This study defines the age-dependent cellular and behavioral effects of these drugs on normally developing brains and determines their reversibility with post-drug intervention. Mice at different postnatal ages received short courses of molecularly targeted drugs in regimens analagous to clinical treatment. Analysis of rapidly developing brain structures important for sensorimotor and cognitive function showed that, while adult administration was without effect, earlier neonatal administration of targeted therapies attenuated white matter oligodendroglia and hippocampal neuronal development more profoundly than later administration, leading to long-lasting behavioral deficits. This functional impairment was reversed by rehabilitation with physical and cognitive enrichment. Our findings demonstrate age-dependent, reversible effects of these drugs on brain development, which are important considerations as treatment options expand for pediatric cancers.Significance: Targeted therapeutics elicit age-dependent long-term consequences on the developing brain that can be ameliorated with environmental enrichment. Cancer Res; 78(8); 2081-95. ©2018 AACR.


Asunto(s)
Factores de Edad , Antineoplásicos/farmacología , Conducta Animal/efectos de los fármacos , Gefitinib/farmacología , Hipocampo/efectos de los fármacos , Terapia Molecular Dirigida , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Sirolimus/farmacología , Sunitinib/farmacología , Animales , Femenino , Hipocampo/citología , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Oligodendroglía/citología
13.
Epilepsia ; 59(5): e63-e67, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29600511

RESUMEN

Arterial spin labeling (ASL) magnetic resonance imaging (MRI) can assess cerebral blood flow (CBF) without using radiolabeled tracers. It is unknown whether regional increases in CBF on ASL MRI correlate with seizure location in newborns. We report 3 newborns with focal seizures localized on continuous video electroencephalogram (cEEG), anatomical brain MRI, and ASL MRI. Each patient underwent pseudocontinuous ASL with segmented 3-dimensional fast spin echo readout as part of standard care. Case 1 is a term male infant presenting with left temporal status epilepticus and recurrent cEEG seizures from an idiopathic large left intraventricular hemorrhage. ASL images demonstrated left mesial temporal lobe increased CBF. Case 2 is a late preterm male infant presenting with recurrent cEEG seizures due to focal right megalencephaly. Ictal EEG and ASL images coincided with the focal dysplasia. Case 3 is a dysmorphic term female infant with nonconvulsive partial status epilepticus identified by focal increased CBF of the left temporal lobe on ASL images. The area of increased CBF was within an area of extensive left hemisphere dysplasia. To our knowledge, this is the first report of regional increases in CBF on ASL MRI correlating with ictal cEEG in newborns.


Asunto(s)
Encéfalo/diagnóstico por imagen , Circulación Cerebrovascular/fisiología , Imagen por Resonancia Magnética/métodos , Convulsiones/diagnóstico por imagen , Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Electroencefalografía , Femenino , Humanos , Recién Nacido , Masculino , Convulsiones/fisiopatología , Marcadores de Spin
14.
Pediatr Res ; 83(1-2): 345-355, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28922350

RESUMEN

Cell-based therapies hold significant promise for infants at risk for cerebral palsy (CP) from perinatal brain injury (PBI). PBI leading to CP results from multifaceted damage to neural cells. Complex developing neural networks are injured by neural cell damage plus unique perturbations in cell signaling. Given that cell-based therapies can simultaneously repair multiple injured neural components during critical neurodevelopmental windows, these interventions potentially offer efficacy for patients with CP. Currently, the use of cell-based interventions in infants at risk for CP is limited by critical gaps in knowledge. In this review, we will highlight key questions facing the field, including: Who are optimal candidates for treatment? What are the goals of therapeutic interventions? What are the best strategies for agent delivery, including timing, dosage, location, and type? And, how are short- and long-term efficacy reliably tracked? Challenges unique to treating PBI with cell-based therapies, and lessons learned from cell-based therapies in closely related neurological disorders in the mature central nervous system, will be reviewed. Our goal is to update pediatric specialists who may be counseling families about the current state of the field. Finally, we will evaluate how rigor can be increased in the field to ensure the safety and best interests of this vulnerable patient population.


Asunto(s)
Lesiones Encefálicas/terapia , Parálisis Cerebral/terapia , Sangre Fetal/citología , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Ensayos Clínicos como Asunto , Trasplante de Células Madre de Sangre del Cordón Umbilical , Humanos , Lactante , Recién Nacido , Fenotipo
15.
Ann Neurol ; 82(1): 121-127, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28556287

RESUMEN

To characterize the mechanism of Zika virus (ZIKV)-associated microcephaly, we performed immunolabeling on brain tissue from a 20-week fetus with intrauterine ZIKV infection. Although ZIKV demonstrated a wide range of neuronal and non-neuronal tropism, the infection rate was highest in intermediate progenitor cells and immature neurons. Apoptosis was observed in both infected and uninfected bystander cortical neurons, suggesting a role for paracrine factors in induction of neuronal apoptosis. Our results highlight differential neuronal susceptibility and neuronal apoptosis as potential mechanisms in the development of ZIKV-associated microcephaly, and may provide insights into the design and best timing of future therapy. Ann Neurol 2017;82:121-127.


Asunto(s)
Feto/patología , Feto/virología , Neuronas/patología , Neuronas/virología , Infección por el Virus Zika/patología , Apoptosis , Encéfalo/patología , Encéfalo/virología , Susceptibilidad a Enfermedades , Humanos , Infección por el Virus Zika/virología
16.
Oncotarget ; 6(14): 12141-55, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25987129

RESUMEN

Diffuse intrinsic pontine gliomas (DIPGs) have a dismal prognosis and are poorly understood brain cancers. Receptor tyrosine kinases stabilized by neuron-glial antigen 2 (NG2) protein are known to induce gliomagenesis. Here, we investigated NG2 expression in a cohort of DIPG specimens (n= 50). We demonstrate NG2 expression in the majority of DIPG specimens tested and determine that tumors harboring histone 3.3 mutation express the highest NG2 levels. We further demonstrate that microRNA 129-2 (miR129-2) is downregulated and hypermethylated in human DIPGs, resulting in the increased expression of NG2. Treatment with 5-Azacytidine, a methyltransferase inhibitor, results in NG2 downregulation in DIPG primary tumor cells in vitro. NG2 expression is altered (symmetric segregation) in mitotic human DIPG and mouse tumor cells. These mitotic cells co-express oligodendrocyte (Olig2) and astrocyte (glial fibrillary acidic protein, GFAP) markers, indicating lack of terminal differentiation. NG2 knockdown retards cellular migration in vitro, while NG2 expressing neurospheres are highly tumorigenic in vivo, resulting in rapid growth of pontine tumors. NG2 expression is targetable in vivo using miR129-2 indicating a potential avenue for therapeutic interventions. This data implicates NG2 as a molecule of interest in DIPGs especially those with H3.3 mutation.


Asunto(s)
Antígenos/genética , Antígenos/metabolismo , Neoplasias del Tronco Encefálico/genética , Glioma/genética , Proteoglicanos/genética , Proteoglicanos/metabolismo , Adolescente , Animales , Neoplasias del Tronco Encefálico/patología , Niño , Perfilación de la Expresión Génica , Glioma/patología , Humanos , Ratones , Mutación
17.
Nat Neurosci ; 18(5): 674-82, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25821912

RESUMEN

Diffuse white matter injury (DWMI), a leading cause of neurodevelopmental disabilities in preterm infants, is characterized by reduced oligodendrocyte formation. NG2-expressing oligodendrocyte precursor cells (NG2 cells) are exposed to various extrinsic regulatory signals, including the neurotransmitter GABA. We investigated GABAergic signaling to cerebellar white matter NG2 cells in a mouse model of DWMI (chronic neonatal hypoxia). We found that hypoxia caused a loss of GABAA receptor-mediated synaptic input to NG2 cells, extensive proliferation of these cells and delayed oligodendrocyte maturation, leading to dysmyelination. Treatment of control mice with a GABAA receptor antagonist or deletion of the chloride-accumulating transporter NKCC1 mimicked the effects of hypoxia. Conversely, blockade of GABA catabolism or GABA uptake reduced NG2 cell numbers and increased the formation of mature oligodendrocytes both in control and hypoxic mice. Our results indicate that GABAergic signaling regulates NG2 cell differentiation and proliferation in vivo, and suggest that its perturbation is a key factor in DWMI.


Asunto(s)
Cerebelo/patología , Enfermedades Desmielinizantes/etiología , Hipoxia Encefálica/fisiopatología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Oligodendroglía/citología , Receptores de GABA-A/fisiología , Sustancia Blanca/lesiones , Ácido gamma-Aminobutírico/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Asfixia Neonatal/patología , Carbacol/farmacología , Recuento de Células , Células Cultivadas , Cerebelo/crecimiento & desarrollo , Enfermedades Desmielinizantes/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Antagonistas de Receptores de GABA-A/toxicidad , Hipoxia Encefálica/patología , Interneuronas/patología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neurogénesis/efectos de los fármacos , Ácidos Nipecóticos/farmacología , Ácidos Nipecóticos/uso terapéutico , Células de Purkinje/patología , Miembro 2 de la Familia de Transportadores de Soluto 12/deficiencia , Miembro 2 de la Familia de Transportadores de Soluto 12/fisiología , Tiagabina , Vigabatrin/farmacología , Vigabatrin/uso terapéutico
18.
J Hum Genet ; 60(7): 363-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25809939

RESUMEN

Pontocerebellar hypoplasia (PCH) is characterized by hypoplasia and atrophy of the cerebellum, variable pontine atrophy, microcephaly, severe mental and motor impairments and seizures. Mutations in 11 genes have been reported in 8 out of 10 forms of PCH. Recessive mutations in the mitochondrial arginyl-transfer RNA synthetase gene (RARS2) have been recently associated with PCH type 6, which is characterized by early-onset encephalopathy with signs of oxidative phosphorylation defect. Here we describe the clinical presentation, neuroimaging findings and molecular characterizations of two siblings with a clinical diagnosis of PCH who displayed a novel variant (c.-2A>G) in the 5'-UTR of the RARS2 gene in the homozygous state. This variant was identified through next-generation sequencing testing of a panel of nine genes known to be involved in PCH. Gene expression and functional studies demonstrated that the c.-2A>G sequence change directly leads to a reduced RARS2 messenger RNA expression in the patients by decreasing RARS2 promoter activity, thus providing evidence that mutations in the RARS2 promoter are likely to represent a new causal mechanism of PCH6.


Asunto(s)
Arginino-ARNt Ligasa/genética , Regiones Promotoras Genéticas , Secuencia de Bases , Enfermedades Cerebelosas/diagnóstico , Enfermedades Cerebelosas/genética , Preescolar , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Mutación Missense , Mutación Puntual
19.
Nat Neurosci ; 17(3): 341-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24569830

RESUMEN

Every year in the United States, an estimated 500,000 babies are born preterm (before 37 completed weeks of gestation), and this number is rising, along with the recognition of brain injuries due to preterm delivery. A common underlying pathogenesis appears to be perinatal hypoxia induced by immature lung development, which causes injury to vulnerable neurons and glia. Abnormal growth and maturation of susceptible cell types, particularly neurons and oligodendrocytes, in preterm babies with very low birth weight is associated with decreased cerebral and cerebellar volumes and increases in cerebral ventricular size. Here we reconcile these observations with recent studies using models of perinatal hypoxia that show perturbations in the maturation and function of interneurons, oligodendrocytes and astroglia. Together, these findings suggest that the global mechanism by which perinatal hypoxia alters development is through a delay in maturation of affected cell types, including astroglia, oligodendroglia and neurons.


Asunto(s)
Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Hipoxia Fetal/complicaciones , Enfermedades del Prematuro/fisiopatología , Neuroglía/citología , Neuronas/citología , Animales , Astrocitos/citología , Lesiones Encefálicas/etiología , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/etiología , Enfermedades del Prematuro/patología , Oligodendroglía/citología
20.
Pediatr Res ; 75(5): 618-25, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24488087

RESUMEN

BACKGROUND: White matter (WM) injury is common after cardiopulmonary bypass or deep hypothermic circulatory arrest in neonates who have cerebral immaturity secondary to in utero hypoxia. The mechanism remains unknown. We investigated effects of preoperative hypoxia on deep hypothermic circulatory arrest-induced WM injury using a combined experimental paradigm in rodents. METHODS: Mice were exposed to hypoxia (prehypoxia). Oxygen-glucose deprivation was performed under three temperatures to simulate brain conditions of deep hypothermic circulatory arrest including ischemia-reperfusion/reoxygenation under hypothermia. RESULTS: WM injury in prenormoxia was identified after 35 °C-oxygen-glucose deprivation. In prehypoxia, injury was displayed in all groups. Among oligodendrocyte stages, the preoligodendrocyte was the most susceptible, while the oligodendrocyte progenitor was resistant to insult. When effects of prehypoxia were assessed, injury of mature oligodendrocytes and oligodendrocyte progenitors in prehypoxia significantly increased as compared with prenormoxia, indicating that mature oligodendrocytes and progenitors that had developed under hypoxia had greater vulnerability. Conversely, damage of oligodendrocyte progenitors in prehypoxia were not identified after 15 °C-oxygen-glucose deprivation, suggesting that susceptible oligodendrocytes exposed to hypoxia are protected by deep hypothermia. CONCLUSION: Developmental alterations due to hypoxia result in an increased WM susceptibility to injury. Promoting WM regeneration by oligodendrocyte progenitors after earlier surgery using deep hypothermia is the most promising approach for successful WM development in congenital heart disease patients.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Puente Cardiopulmonar/efectos adversos , Hipoxia , Sustancia Blanca/patología , Animales , Encéfalo/patología , Linaje de la Célula , Modelos Animales de Enfermedad , Glucosa/química , Proteínas Fluorescentes Verdes/química , Hipotermia , Ratones , Oligodendroglía/citología , Oxígeno/química , Perfusión , Daño por Reperfusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA