Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Dev Neurosci ; 46(2): 98-111, 2024.
Article in English | MEDLINE | ID: mdl-37231852

ABSTRACT

The developing brain is uniquely susceptible to oxidative stress, and endogenous antioxidant mechanisms are not sufficient to prevent injury from a hypoxic-ischemic challenge. Glutathione peroxidase (GPX1) activity reduces hypoxic-ischemic injury. Therapeutic hypothermia (HT) also reduces hypoxic-ischemic injury, in the rodent and the human brain, but the benefit is limited. Here, we combined GPX1 overexpression with HT in a P9 mouse model of hypoxia-ischemia (HI) to test the effectiveness of both treatments together. Histological analysis showed that wild-type (WT) mice with HT were less injured than WT with normothermia. In the GPX1-tg mice, however, despite a lower median score in the HT-treated mice, there was no significant difference between HT and normothermia. GPX1 protein expression was higher in the cortex of all transgenic groups at 30 min and 24 h, as well as in WT 30 min after HI, with and without HT. GPX1 was higher in the hippocampus of all transgenic groups and WT with HI and normothermia, at 24 h, but not at 30 min. Spectrin 150 was higher in all groups with HI, while spectrin 120 was higher in HI groups only at 24 h. There was reduced ERK1/2 activation in both WT and GPX1-tg HI at 30 min. Thus, with a relatively moderate insult, we see a benefit with cooling in the WT but not the GPX1-tg mouse brain. The fact that we see no benefit with increased GPx1 here in the P9 model (unlike in the P7 model) may indicate that oxidative stress in these older mice is elevated to an extent that increased GPx1 is insufficient for reducing injury. The lack of benefit of overexpressing GPX1 in conjunction with HT after HI indicates that pathways triggered by GPX1 overexpression may interfere with the neuroprotective mechanisms provided by HT.


Subject(s)
Hypothermia, Induced , Hypothermia , Hypoxia-Ischemia, Brain , Animals , Mice , Humans , Animals, Newborn , Spectrin , Hypoxia-Ischemia, Brain/pathology , Hypoxia , Glutathione Peroxidase/metabolism , Antioxidants , Ischemia
2.
Curr Neuropharmacol ; 20(7): 1400-1412, 2022.
Article in English | MEDLINE | ID: mdl-34766894

ABSTRACT

The central nervous system (CNS) is enriched with important classes of lipids, in which cholesterol is known to make up a major portion of myelin sheaths, besides being a structural and functional unit of CNS cell membranes. Unlike in the adult brain, where the cholesterol pool is relatively stable, cholesterol is synthesized and accumulated at the highest rate in the developing brain to meet the needs of rapid brain growth at this stage, which is also a critical period for neuroplasticity. In addition to its biophysical role in membrane organization, cholesterol is crucial for brain development due to its involvement in brain patterning, myelination, neuronal differentiation, and synaptogenesis. Thus any injuries to the immature brain that affect cholesterol homeostasis may have long-term adverse neurological consequences. In this review, we describe the unique features of brain cholesterol biosynthesis and metabolism, cholesterol trafficking between different cell types, and highlight cholesterol-dependent biological processes during brain maturation. We also discuss the association of impaired cholesterol homeostasis with several forms of perinatal brain disorders in term and preterm newborns, including hypoxic-ischemic encephalopathy. Strategies targeting the cholesterol pathways may open new avenues for the diagnosis and treatment of developmental brain injury.


Subject(s)
Brain Injuries , Hypoxia-Ischemia, Brain , Adult , Brain/metabolism , Brain Injuries/metabolism , Central Nervous System/metabolism , Cholesterol/metabolism , Female , Humans , Hypoxia-Ischemia, Brain/metabolism , Infant, Newborn , Pregnancy
3.
J Neurosci Res ; 99(11): 2964-2975, 2021 11.
Article in English | MEDLINE | ID: mdl-34487578

ABSTRACT

The cellular responses to hypoxia or hypoxia-ischemia (HI) are governed largely by the hypoxia-inducible factor (HIF) family of transcription factors. Our previous studies show that HIF-1α induction is an important factor that mediates protective effects in the brain after neonatal HI. In the present study, we investigated the contribution of another closely related HIF α isoform, HIF-2α, specifically the neuronal HIF-2α, to brain HI injury. Homozygous transgenic mice with a floxed exon 2 of HIF-2α were bred with CaMKIIα-Cre mice to generate a mouse line with selective deletion of HIF-2α in forebrain neurons. These mice, along with their wildtype littermates, were subjected to HI at postnatal day 9. Brain injury at different ages was evaluated by the levels of cleaved caspase-3 and spectrin breakdown products at 24 hr; and histologically at 6 days or 3 months after HI. Multiple behavioral tests were performed at 3 months, prior to sacrifice. Loss of neuronal HIF-2α exacerbated brain injury during the acute (24 hr) and subacute phases (6 days), with a trend toward more severe volume loss in the adult brain. The long-term brain function for coordinated movement and recognition memory, however, were not impacted in the neuronal HIF-2α deficient mice. Our data suggest that, similar to HIF-1α, neuronal HIF-2α promotes cell survival in the immature mouse brain. The two HIF alpha isoforms may act through partially overlapping or distinct transcriptional targets to mediate their intrinsic protective responses against neonatal HI brain injury.


Subject(s)
Brain Injuries , Neurons , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Injuries/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Transgenic , Neurons/metabolism
4.
J Cereb Blood Flow Metab ; 41(2): 312-323, 2021 02.
Article in English | MEDLINE | ID: mdl-32169014

ABSTRACT

The major pathway of brain cholesterol turnover relies on its hydroxylation into 24S-hydroxycholesterol (24S-HC) using brain-specific cytochrome P450 46A1 (CYP46A1). 24S-HC produced exclusively in the brain normally traverses the blood-brain barrier to enter the circulation to the liver for excretion; therefore, the serum 24S-HC level is an indication of cholesterol metabolism in the brain. We recently reported an upregulation of CYP46A1 following hypoxia-ischemia (HI) in the neonatal mouse brain and a correlation between serum 24S-HC levels and acute brain damage. Here, we performed a longitudinal study to investigate whether the serum 24S-HC concentrations predict long-term brain structural and functional outcomes. In postnatal day 9 mice subjected to HI, the serum 24S-HC levels increased at 6 h and 24 h after HI and correlated with the infarct volumes measured histologically or by T2-weighted MRI. The 24 h levels were associated with white matter volume loss quantified by MBP immunostaining and luxol fast blue staining. The animals with higher serum 24S-HC at 6 h and 24 h corresponded to those with more severe motor and cognitive deficits at 35-40 days after HI. These data suggest that 24S-HC could be a novel and early blood biomarker for severity of neonatal HI brain damage and associated functional impairments.


Subject(s)
Biomarkers/metabolism , Hydroxycholesterols/metabolism , Hypoxia-Ischemia, Brain/metabolism , Animals , Animals, Newborn , Male , Mice , Treatment Outcome
5.
Dev Neurosci ; : 1-10, 2019 Mar 05.
Article in English | MEDLINE | ID: mdl-30836371

ABSTRACT

Hypoxia-inducible factor 1α (HIF1α) is a key regulator of oxygen homeostasis, and its target genes mediate adaptive, protective, and pathological processes. The role of HIF1α in neuronal survival is controversial and the brain maturation stage is important in determining its function in brain ischemia or hypoxia-ischemia (HI). In this study, we used neuron-specific HIF1α knockout mice at postnatal day 9 (P9), and immature cortical neurons (days 7-8 in vitro) treated with the HIF1α inhibitor 2-methoxyestradiol (2ME2) or stabilizer dimethyloxalylglycine (DMOG), to examine the function of neuronal HIF1α in neonatal HI in vivo (Vannucci model) and in vitro (oxygen glucose deprivation, OGD). Inhibition of HIF1α with 2ME2 in primary neurons or deletion of neuronal HIF1α in P9 mice increased both necrotic and apoptotic cell death following HI, as evaluated by the protein levels of 145/150-kDa and 120-kDa spectrin breakdown products 24 h after HI. DMOG attenuated neuronal death right after OGD. Acute pharmacological manipulation of HIF1α synchronously regulated the expression of its targets, vascular endothelial growth factor (VEGF) and erythropoietin (Epo), in the same manner. The in vivo findings agree with our previous data using the same HIF1α-deficient mice at an earlier age. This study confirms the role of neuronal HIF1α signaling in the endogenous protective responses following HI in the developing brain.

6.
Physiol Behav ; 206: 76-83, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30904570

ABSTRACT

Fluoride ingestion has been linked to changes in behavior in mice and rats, related to dose, sex of the animal, and the timing of exposure. Previous studies have shown the behavior of female rats to be most affected by postnatal fluoride exposure, and in this study we determined the effects of postnatal fluoride exposure on anxiety related behavior and serotonin. Mice given 50 ppm fluoride in drinking water had increased entries in the open arms of the elevated plus maze, suggesting reduced anxiety. Both peripheral and central serotonin was increased in the fluoride treated mice. In a cohort of children drinking water containing 2.5 ppm fluoride, serum serotonin was also increased as compared to controls. The mechanisms by which fluoride results in an increase peripheral and central serotonin are not well understood, but warrant further study, as these effects may also be relevant to prenatal fluoride related changes in behavior in both mice and humans.


Subject(s)
Behavior, Animal/drug effects , Fluorides/administration & dosage , Maze Learning/drug effects , Motor Activity/drug effects , Serotonin/blood , Social Behavior , Administration, Oral , Animals , Brain Chemistry , Female , Fluorides/analysis , Mice
7.
Sci Rep ; 8(1): 17522, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504838

ABSTRACT

NMDA-type glutamate receptors (NMDAR) trigger superoxide production by neuronal NADPH oxidase-2 (NOX2), which if sustained leads to cell death. This process involves Ca2+ influx through NMDAR channels. By contrast, comparable Ca2+ influx by other routes does not induce NOX2 activation or cell death. This contrast has been attributed to site-specific effects of Ca2+ flux through NMDAR. Here we show instead that it stems from non-ionotropic signaling by NMDAR GluN2B subunits. To evaluate non-ionotropic effects, mouse cortical neurons were treated with NMDA together with 7-chlorokynurenate, L-689,560, or MK-801, which block Ca2+ influx through NMDAR channels but not NMDA binding. NMDA-induced superoxide formation was prevented by the channel blockers, restored by concurrent Ca2+ influx through ionomycin or voltage-gated calcium channels, and not induced by the Ca2+ influx in the absence of NMDAR ligand binding. Neurons expressing either GluN2B subunits or chimeric GluN2A/GluN2B C-terminus subunits exhibited NMDA-induced superoxide production, whereas neurons expressing chimeric GluN2B/GluN2A C-terminus subunits did not. Neuronal NOX2 activation requires phosphoinositide 3-kinase (PI3K), and NMDA binding to NMDAR increased PI3K association with NMDA GluN2B subunits independent of Ca2+ influx. These findings identify a non-ionotropic signaling pathway that links NMDAR to NOX2 activation through the C-terminus domain of GluN2B.


Subject(s)
Cell Death , Neurons/cytology , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Superoxides/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Ion Transport , Ionomycin/pharmacology , Mice , NADPH Oxidase 2/metabolism , Neurons/drug effects , Neurons/metabolism
8.
Pediatr Res ; 83(6): 1218-1227, 2018 06.
Article in English | MEDLINE | ID: mdl-29718007

ABSTRACT

BackgroundMaintenance of cholesterol homeostasis is crucial for brain development. Brain cholesterol relies on de novo synthesis and is cleared primarily by conversion to 24S-hydroxycholesterol (24S-HC) with brain-specific cholesterol 24-hydroxylase (CYP46A1). We aimed to investigate the impact of hypoxia-ischemia (HI) on brain cholesterol metabolism in the neonatal mice.MethodsPostnatal day 9 C57BL/6 pups were subjected to HI using the Vannucci model. CYP46A1 expression was assessed with western blotting and its cellular localization was determined using immunofluorescence staining. The amount of brain cholesterol, 24S-HC in the cortex and in the serum, was measured with enzyme-linked immunosorbent assay (ELISA).ResultsThere was a transient cholesterol loss at 6 h after HI. CYP46A1 was significantly upregulated at 6 and 24 h following HI with a concomitant increase of 24S-HC in the ipsilateral cortex and in the serum. The serum levels of 24S-HC correlated with those in the brain, as well as with necrotic and apoptotic cell death evaluated by the expression of spectrin breakdown products and cleaved caspase-3 at 6 and 24 h after HI.ConclusionEnhanced cholesterol turnover by activation of CYP46A1 represents disrupted brain cholesterol homeostasis early after neonatal HI. 24S-HC might be a novel blood biomarker for severity of hypoxic-ischemic encephalopathy with potential clinical application.


Subject(s)
Brain/metabolism , Cholesterol 24-Hydroxylase/metabolism , Cholesterol/metabolism , Gene Expression Regulation, Enzymologic , Hypoxia-Ischemia, Brain/pathology , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/growth & development , Cerebral Cortex/metabolism , Enzyme-Linked Immunosorbent Assay , Hydroxycholesterols/chemistry , Hypoxia , Hypoxia-Ischemia, Brain/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Oligodendroglia/metabolism , Up-Regulation
9.
Exp Neurol ; 299(Pt A): 65-74, 2018 01.
Article in English | MEDLINE | ID: mdl-28993251

ABSTRACT

The N-methyl-d-aspartate-type glutamate receptor (NMDAR)-associated multiprotein complexes are indispensable for synaptic plasticity and cognitive functions. While purification and proteomic analyses of these signaling complexes have been performed in adult rodent and human brain, much less is known about the protein composition of NMDAR complexes in the developing brain and their modifications by neonatal hypoxic-ischemic (HI) brain injury. In this study, the postsynaptic density proteins were prepared from postnatal day 9 naïve, sham-operated and HI-injured mouse cortex. The GluN2B-containing NMDAR complexes were purified by immunoprecipitation with a mouse GluN2B antibody and subjected to mass spectrometry analysis for determination of the GluN2B binding partners. A total of 71 proteins of different functional categories were identified from the naïve animals as native GluN2B-interacting partners in the developing mouse brain. Neonatal HI reshaped the postsynaptic GluN2B interactome by recruiting new proteins, including multiple kinases, into the complexes; and modifying the existing associations within 1h of reperfusion. The early responses of postsynaptic NMDAR complexes and their related signaling networks may contribute to molecular processes leading to cell survival or death, brain damage and/or neurological disorders in term infants with neonatal encephalopathy.


Subject(s)
Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Neurotransmitter/metabolism , Animals , Animals, Newborn , Brain/growth & development , Brain Chemistry/genetics , Female , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Neural Pathways/anatomy & histology , Neural Pathways/growth & development , Neurons/metabolism , Pregnancy , Primary Cell Culture , Protein Kinases/metabolism , Proteomics
10.
Dev Neurosci ; 39(1-4): 66-81, 2017.
Article in English | MEDLINE | ID: mdl-28315865

ABSTRACT

Proteomics of the synapses and postsynaptic densities (PSDs) have provided a deep understanding of protein composition and signal networks in the adult brain, which underlie neuronal plasticity and neurodegenerative or psychiatric disorders. However, there is a paucity of knowledge about the architecture and organization of PSDs in the immature brain, and how it is modified by brain injury in an early developing stage. Mass spectrometry (MS)-based proteomic analysis was performed on PSDs prepared from cortices of postnatal day 9 naïve mice or pups which had suffered hypoxic-ischemic (HI) brain injury. 512 proteins of different functional groups were identified from PSDs collected 1 h after HI injury, among which 60 have not been reported previously. Seven newly identified proteins involved in neural development were highlighted. HI injury increased the yield of PSDs at early time points upon reperfusion, and multiple proteins were recruited into PSDs following the insult. Quantitative analysis was performed using spectral counting, and proteins whose relative expression was more than 50% up- or downregulated compared to the sham animals 1 h after HI insult were reported. Validation with Western blotting demonstrated changes in expression and phosphorylation of the N-methyl-D-aspartate receptor, activation of a series of postsynaptic protein kinases and dysregulation of scaffold and adaptor proteins in response to neonatal HI insult. This work, along with other recent studies of synaptic protein profiling in the immature brain, builds a foundation for future investigation on the molecular mechanisms underlying developing plasticity. Furthermore, it provides insights into the biochemical changes of PSDs following early brain hypoxia-ischemia, which is helpful for understanding not only the injury mechanisms, but also the process of repair or replenishment of neuronal circuits during recovery from brain damage.


Subject(s)
Asphyxia Neonatorum/pathology , Cerebral Cortex/pathology , Hypoxia-Ischemia, Brain/pathology , Post-Synaptic Density/pathology , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Cerebral Cortex/metabolism , Female , Humans , Hypoxia-Ischemia, Brain/metabolism , Male , Mice , Mice, Inbred C57BL , Post-Synaptic Density/metabolism , Proteomics
11.
Stroke ; 45(10): 3040-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25158771

ABSTRACT

BACKGROUND AND PURPOSE: The NR2B subunit of the N-methyl-d-aspartate (NMDA) receptor is phosphorylated by the Src family kinase Fyn in brain, with tyrosine (Y) 1472 as the major phosphorylation site. Although Y1472 phosphorylation is important for synaptic plasticity, it is unknown whether it is involved in NMDA receptor-mediated excitotoxicity in neonatal brain hypoxia-ischemia (HI). This study was designed to elucidate the specific role of Y1472 phosphorylation of NR2B in neonatal HI in vivo and in NMDA-mediated neuronal death in vitro. METHODS: Neonatal mice with a knockin mutation of Y1472 to phenylalanine (YF-KI) and their wild-type littermates were subjected to HI using the Vannucci model. Brains were scored 5 days later for damage using cresyl violet and iron staining. Western blotting and immunoprecipitation were performed to determine NR2B tyrosine phosphorylation. Expression of NADPH oxidase subunits and superoxide production were measured in vivo. NMDA-induced calcium response, superoxide formation, and cell death were evaluated in primary cortical neurons. RESULTS: After neonatal HI, YF-KI mice have reduced expression of NADPH oxidase subunit gp91phox and p47phox and superoxide production, lower activity of proteases implicated in necrotic and apoptotic cell death, and less brain damage when compared with the wild-type mice. In vitro, YF-KI mutation diminishes superoxide generation in response to NMDA without effect on calcium accumulation and inhibits NMDA and glutamate-induced cell death. CONCLUSIONS: Upregulation of NR2B phosphorylation at Y1472 after neonatal HI is involved in superoxide-mediated oxidative stress and contributes to brain injury.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Animals, Newborn , Blotting, Western , Disease Models, Animal , Fluorescent Antibody Technique , Gene Knock-In Techniques , Immunoprecipitation , Mice , Mice, Inbred C57BL , Oxidative Stress , Phosphorylation , Tyrosine/metabolism
12.
Chin J Traumatol ; 10(6): 349-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18045516

ABSTRACT

OBJECTIVE: To analyze the application of vertebral pedicle screw fixation in the treatment of burst fracture of thoracolumbar vertebrae. METHODS: A total of 48 cases (31 males and 17 females, aged from 18-72 years, mean: 41.3 years) with thoracolumbar vertebrae burst fracture were treated by pedicle screw system since January 2004. According to the AO classification of thoracolumbar vertebrae fracture, there are 36 cases of Type A, 9 of Type B and 3 of Type C. RESULTS: All patients were followed up for 6-25 months (average 12 months), no secondary nerve root injury, spinal cord injury, loosening or breakage of pedicle screw were observed. The nerve function of 29 patients with cauda equina nerve injury was restored to different degrees. The vertebral body height returned to normal level and posterior process angle was rectified after operation. CONCLUSIONS: The vertebral pedicle screw internal fixation was technologically applicable, which can efficiently reposition and stabilize the bursting fractured vertebrae, indirectly decompress canalis spinalis, maintain spine stability, scatter stress of screw system, reduce the risk of loosening or breakage of screw and loss of vertebral height, and prevent the formation of posterior convex after operation.


Subject(s)
Bone Screws , Internal Fixators , Lumbar Vertebrae/injuries , Spinal Fractures/surgery , Thoracic Vertebrae/injuries , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...