Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Exp Neurol ; 263: 200-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25263581

ABSTRACT

Postnatal glucocorticoids (GCs) are widely used in the prevention of chronic lung disease in premature infants. Their pharmacologic use is associated with neurodevelopmental delay and cerebral palsy. However, the effect of GC dose and preparation (dexamethasone versus betamethasone) on short and long-term neurological outcomes remains undetermined, and the mechanisms of GC-induced brain injury are unclear. We hypothesized that postnatal GC would induce hypomyelination and motor impairment in a preparation- and dose-specific manner, and that GC receptor (GR) inhibition might restore myelination and neurological function in GC-treated animals. Additionally, GC-induced hypomyelination and neurological deficit might be transient. To test our hypotheses, we treated prematurely delivered rabbit pups with high (0.5mg/kg/day) or low (0.2mg/kg/day) doses of dexamethasone or betamethasone. Myelin basic protein (MBP), oligodendrocyte proliferation and maturation, astrocytes, transcriptomic profile, and neurobehavioral functions were evaluated. We found that high-dose GC treatment, but not low-dose, reduced MBP expression and impaired motor function at postnatal day 14. High-dose dexamethasone induced astrogliosis, betamethasone did not. Mifepristone, a GR antagonist, reversed dexamethasone-induced myelination, but not astrogliosis. Both GCs inhibited oligodendrocyte proliferation and maturation. Moreover, high-dose dexamethasone altered genes associated with myelination, cell-cycle, GR, and mitogen-activated protein kinase. Importantly, GC-induced hypomyelination, gliosis, and motor-deficit, observed at day 14, completely recovered by day 21. Hence, high-dose, but not low-dose, postnatal GC causes reversible reductions in myelination and motor functions. GC treatment induces hypomyelination by GR-dependent genomic mechanisms, but astrogliosis by non-genomic mechanisms. GC-induced motor impairment and neurodevelopmental delay might be transient and recover spontaneously in premature infants.


Subject(s)
Brain/drug effects , Glucocorticoids/adverse effects , Myelin Sheath/drug effects , Animals , Animals, Newborn , Betamethasone/administration & dosage , Betamethasone/adverse effects , Blotting, Western , Brain/pathology , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gliosis/chemically induced , Gliosis/pathology , Glucocorticoids/administration & dosage , Immunohistochemistry , In Situ Nick-End Labeling , Myelin Sheath/pathology , Rabbits , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/metabolism
2.
Exp Neurol ; 247: 630-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23474192

ABSTRACT

Intraventricular hemorrhage (IVH) results in white matter injury and hydrocephalus in premature infants. Chondroitin sulfate proteoglycans (CSPGs)-neuorcan, brevican, versican, aggrecan and phosphacan-are unregulated in the extracellular matrix after brain injury, and their degradation enhances plasticity of the brain. Therefore, we hypothesized that CSPG levels were elevated in the forebrain of premature infants with IVH and that in vivo degradation of CSPGs would enhance maturation of oligodendrocyte, augment myelination, promote neurological recovery, and minimize hydrocephalus. We found that levels of neurocan, brevican, aggrecan, phosphacan, and versican were elevated, whereas NG2 expression was reduced in premature rabbit pups and human infants with IVH compared to controls. Intracerebroventricular chondroitinase ABC (ChABC) reduced the expression of neuorcan, brevican, versican and aggrecan, but not NG2. However, ChABC treatment did not enhance maturation of oligodendrocytes, myelination, or neurological recovery in the pups with IVH. Moreover, ChABC did not reduce gliosis or ventriculomegaly. Our results demonstrate that IVH induces distinct changes in the components of CSPGs, and that reversing these changes by in vivo ChABC treatment neither promotes clinical recovery, myelination, nor reduces ventriculomegaly in preterm rabbit pups.


Subject(s)
Cerebral Hemorrhage , Chondroitin Sulfate Proteoglycans/metabolism , Gene Expression Regulation, Developmental/physiology , Recovery of Function/physiology , Age Factors , Animals , Animals, Newborn , Antigens/genetics , Antigens/metabolism , Cell Proliferation/drug effects , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Chondroitin ABC Lyase/administration & dosage , Chondroitin Sulfate Proteoglycans/genetics , Disease Models, Animal , Female , Fetus , Gestational Age , Humans , Infant, Newborn , Male , Motor Activity/drug effects , Motor Activity/physiology , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Pregnancy , Proteoglycans/genetics , Proteoglycans/metabolism , Rabbits , Recovery of Function/drug effects , Time Factors
3.
J Neurosci ; 33(2): 411-23, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23303921

ABSTRACT

Premature infants exhibit neurodevelopmental delay and reduced growth of the cerebral cortex. However, the underlying mechanisms have remained elusive. Therefore, we hypothesized that neurogenesis in the ventricular and subventricular zones of the cerebral cortex would continue in the third trimester of pregnancy and that preterm birth would suppress neurogenesis. To test our hypotheses, we evaluated autopsy materials from human fetuses and preterm infants of 16-35 gestational weeks (gw). We noted that both cycling and noncycling Sox2(+) radial glial cells and Tbr2(+) intermediate progenitors were abundant in human preterm infants until 28 gw. However, their densities consistently decreased from 16 through 28 gw. To determine the effect of premature birth on neurogenesis, we used a rabbit model and compared preterm [embryonic day 29 (E29), 3 d old] and term (E32, <2 h old) pups at an equivalent postconceptional age. Glutamatergic neurogenesis was suppressed in preterm rabbits, as indicated by the reduced number of Tbr2(+) intermediate progenitors and the increased number of Sox2(+) radial glia. Additionally, hypoxia-inducible factor-1α, vascular endothelial growth factor, and erythropoietin were higher in term than preterm pups, reflecting the hypoxic intrauterine environment of just-born term pups. Proneural genes, including Pax6 and Neurogenin-1 and -2, were higher in preterm rabbit pups compared with term pups. Importantly, neurogenesis and associated factors were restored in preterm pups by treatment with dimethyloxallyl glycine, a hypoxia mimetic agent. Hence, glutamatergic neurogenesis continues in the premature infants, preterm birth suppresses neurogenesis, and hypoxia-mimetic agents might restore neurogenesis, enhance cortical growth, and improve neurodevelopmental outcome of premature infants.


Subject(s)
Neurogenesis/physiology , Pregnancy Trimester, Third/physiology , Premature Birth/physiopathology , Adult , Animals , Cell Count , Cerebral Ventricles/growth & development , Erythropoietin/physiology , Female , Gestational Age , Glycine/pharmacology , Humans , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1/biosynthesis , Hypoxia-Inducible Factor 1/physiology , Immunohistochemistry , Infant, Newborn , Infant, Premature , Male , Nerve Tissue Proteins/biosynthesis , Neural Stem Cells/physiology , Pregnancy , Rabbits , Signal Transduction/physiology , Telencephalon/growth & development , Vascular Endothelial Growth Factor A/physiology , Wnt Proteins/physiology , beta Catenin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL