1
|
Northington FJ, Kratimenos P, Turnbill V, Flock DL, Asafu-Adjaye D, Chavez-Valdez R, Martin LJ. Basal forebrain magnocellular cholinergic systems are damaged in mice following neonatal hypoxia-ischemia. J Comp Neurol 2022; 530:1148-1163. [PMID: 34687459 PMCID: PMC9014889 DOI: 10.1002/cne.25263] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/02/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) causes lifelong neurologic disability. Despite the use of therapeutic hypothermia, memory deficits and executive functions remain severely affected. Cholinergic neurotransmission from the basal forebrain to neocortex and hippocampus is central to higher cortical functions. We examined the basal forebrain by light microscopy and reported loss of choline acetyltransferase-positive (ChAT)+ neurons, at postnatal day (P) 40, in the ipsilateral medial septal nucleus (MSN) after neonatal hypoxia-ischemia (HI) in mice. There was no loss of ChAT+ neurons in the ipsilateral nucleus basalis of Meynert (nbM) and striatum. Ipsilateral striatal and nbM ChAT+ neurons were abnormal with altered immunoreactivity for ChAT, shrunken and crenated somas, and dysmorphic appearing dendrites. Using confocal images with 3D reconstruction, nbM ChAT+ dendrites in HI mice were shorter than sham (p = .0001). Loss of ChAT+ neurons in the MSN directly correlated with loss of ipsilateral hippocampal area. In the nbM and striatum, percentage of abnormal ChAT+ neurons correlated with loss of ipsilateral cerebral cortical and striatal area, respectively. Acetylcholinesterase (AChE) activity increased in adjacent ipsilateral cerebral cortex and hippocampus and the increase was linearly related to loss of cortical and hippocampal area. Numbers and size of cathepsin D+ lysosomes increased in large neurons in the ipsilateral nbM. After neonatal HI, abnormalities were found throughout the major cholinergic systems in relationship to amount of forebrain area loss. There was also an upregulation of cathepsin D+ particles within the nbM. Cholinergic neuropathology may underlie the permanent dysfunction in learning, memory, and executive function after neonatal brain injury.
Collapse
Affiliation(s)
- Frances J. Northington
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA,Corresponding Author: CMSC 6-104, Johns Hopkins Hospital, 600 North Wolfe Street, Baltimore, MD 21287,
| | - Panagiotis Kratimenos
- Department of Pediatrics and Neuroscience, Children’s National Hospital & The George Washington University School of Medicine, Washington, D.C
| | - Victoria Turnbill
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Debra L. Flock
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Daniella Asafu-Adjaye
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Raul Chavez-Valdez
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lee J. Martin
- Department of Neuroscience, Pathology, and Anesthesiology & Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Deng C, Li J, Li L, Sun F, Xie J. Effects of hypoxia ischemia on caspase-3 expression and neuronal apoptosis in the brain of neonatal mice. Exp Ther Med 2019; 17:4517-4521. [PMID: 31086583 PMCID: PMC6488988 DOI: 10.3892/etm.2019.7487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 02/01/2019] [Indexed: 02/06/2023] Open
Abstract
Effects of hypoxia ischemia on caspase-3 expression and neuronal apoptosis in the brain of neonatal mice were investigated. Twenty-five neonatal CD1 mice aged 1 week were selected and randomly divided into sham-operation group (n=8) and newborn hypoxia ischemia encephalopathy (NHIE) model group (n=17). The messenger ribonucleic acid (mRNA) expression levels of caspase-3 and Fas ligand (FasL) in brain tissues of mice in both groups were detected via reverse transcription-polymerase chain reaction (RT-PCR). The protein expression levels of caspase-3 and FasL in mice in both groups were detected via western blotting. Moreover, apoptosis of brain tissues was detected using the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), and caspase-3 protein expression level in brain tissues was detected using immunohistochemical methods. Results of RT-PCR and western blotting revealed that compared with those in sham-operation group, caspase-3 and FasL expression levels in model group were significantly increased. Results of TUNEL showed that the number of apoptotic neurons in model group was significantly increased. Besides, results of immunohistochemical detection manifested that the caspase-3 protein expression level in model group was obviously increased. Hypoxia ischemia can lead to significant increase of caspase-3 expression and increase of neuronal apoptosis in the brain of neonatal mice.
Collapse
Affiliation(s)
- Changbo Deng
- Department of Pediatrics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Juan Li
- Department of Pediatrics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Luyi Li
- Department of Pediatrics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Fengjie Sun
- Department of Pediatrics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Jiqing Xie
- Department of Pediatrics, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| |
Collapse
|
3
|
Miguel PM, Deniz BF, Deckmann I, Confortim HD, Diaz R, Laureano DP, Silveira PP, Pereira LO. Prefrontal cortex dysfunction in hypoxic-ischaemic encephalopathy contributes to executive function impairments in rats: Potential contribution for attention-deficit/hyperactivity disorder. World J Biol Psychiatry 2018; 19:547-560. [PMID: 28105895 DOI: 10.1080/15622975.2016.1273551] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The attention-deficit/hyperactivity disorder (ADHD) compromises the quality of life of individuals including adaptation to the social environment. ADHD aetiology includes perinatal conditions such as hypoxic-ischaemic events; preclinical studies have demonstrated attentional deficits and impulsive-hyperactive outcomes after neonatal hypoxic and/or ischaemic intervention, but data are missing to understand this relationship. Thus, the aim of this study was to evaluate executive function (EF) and impulsivity, and tissue integrity and dopaminergic function in the prefrontal cortex (PFC) of rats submitted to hypoxia-ischaemia (HI). METHODS At postnatal day (PND) 7, male Wistar rats were divided into control (n = 10) and HI groups (n = 11) and the HI procedure was conducted. At PND60, the animals were tested in the attentional set-shifting (ASS) task to EF and in the tolerance to delay of reward for assessment of impulsivity. After, morphological analysis and the dopaminergic system were evaluated in the PFC. RESULTS Animals subjected to HI had impairments in EF evidenced by a behavioural inflexibility that was correlated to PFC atrophy. Moreover, HI animals presented reduced D2 receptors in the ipsilateral side of ischaemia in the PFC. CONCLUSIONS Animals submitted to HI presented impaired EF associated with tissue atrophy and dopaminergic disturbance in the PFC.
Collapse
Affiliation(s)
- Patrícia Maidana Miguel
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Bruna Ferrary Deniz
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Iohanna Deckmann
- b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Heloísa Deola Confortim
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Ramiro Diaz
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Daniela Pereira Laureano
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Patrícia Pelufo Silveira
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,c Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,d Ludmer Centre for Neuroinformatics and Mental Health , Douglas Mental Health University Institute, McGill University , Montreal , QC , Canada
| | - Lenir Orlandi Pereira
- a Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS) , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil.,b Departamento de Ciências Morfológicas, ICBS , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| |
Collapse
|
4
|
Hypoxia-Induced Neuroinflammation and Learning-Memory Impairments in Adult Zebrafish Are Suppressed by Glucosamine. Mol Neurobiol 2018; 55:8738-8753. [PMID: 29589284 DOI: 10.1007/s12035-018-1017-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 03/16/2018] [Indexed: 10/17/2022]
Abstract
This study investigated changes in neuroinflammation and cognitive function in adult zebrafish exposed to acute hypoxia and protective effects of glucosamine (GlcN) against hypoxia-induced brain damage. The survival rate of zebrafish following exposure to hypoxia was improved by GlcN pretreatment. Moreover, hypoxia-induced upregulation of neuroglobin, NOS2α, glial fibrillary acidic protein, and S100β in zebrafish was suppressed by GlcN. Hypoxia stimulated cell proliferation in the telencephalic ventral domain and in cerebellum subregions. GlcN decreased the number of bromodeoxyuridine (BrdU)-positive cells in the telencephalon region, but not in cerebellum regions. Transient motor neuron defects, assessed by measuring the locomotor and exploratory activity of zebrafish exposed to hypoxia recovered quickly. GlcN did not affect hypoxia-induced motor activity changes. In passive avoidance tests, hypoxia impaired learning and memory ability, deficits that were rescued by GlcN. A learning stimulus increased the nuclear translocation of phosphorylated cAMP response element binding protein (p-CREB), an effect that was greatly inhibited by hypoxia. GlcN restored nuclear p-CREB after a learning trial in hypoxia-exposed zebrafish. The neurotransmitters, γ-aminobutyric acid and glutamate, were increased after hypoxia in the zebrafish brain, and GlcN further increased their levels. In contrast, acetylcholine levels were reduced by hypoxia and restored by GlcN. Acetylcholinesterase inhibitor physostigmine partially reversed the impaired learning and memory of hypoxic zebrafish. This study represents the first examination of the molecular mechanisms underlying hypoxia-induced memory and learning defects in a zebrafish model. Our results further suggest that GlcN-associated hexosamine metabolic pathway could be an important therapeutic target for hypoxic brain damage.
Collapse
|
5
|
|
6
|
Grothe MJ, Scheef L, Bäuml J, Meng C, Daamen M, Baumann N, Zimmer C, Teipel S, Bartmann P, Boecker H, Wolke D, Wohlschläger A, Sorg C. Reduced Cholinergic Basal Forebrain Integrity Links Neonatal Complications and Adult Cognitive Deficits After Premature Birth. Biol Psychiatry 2017; 82:119-126. [PMID: 28129944 DOI: 10.1016/j.biopsych.2016.12.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 11/23/2016] [Accepted: 12/09/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Prematurely born individuals have an increased risk for long-term neurocognitive impairments. In animal models, development of the cholinergic basal forebrain (cBF) is selectively vulnerable to adverse effects of perinatal stressors, and impaired cBF integrity results in lasting cognitive deficits. We hypothesized that cBF integrity is impaired in prematurely born individuals and mediates adult cognitive impairments associated with prematurity. METHODS We used magnetic resonance imaging-based volumetric assessments of a cytoarchitectonically defined cBF region of interest to determine differences in cBF integrity between 99 adults who were born very preterm and/or with very low birth weight and 106 term-born control subjects from the same birth cohort. Magnetic resonance imaging-derived cBF volumes were studied in relation to neonatal clinical complications after delivery and intelligence measures (IQ) in adulthood. RESULTS In adults who were born very preterm and/or with very low birth weight, cBF volumes were significantly reduced compared with term-born adults (-4.5% [F1,202 = 11.82, p = .001]). Lower cBF volume in adults who were born very preterm and/or with very low birth weight was specifically associated with both neonatal complications (rpart,92 = -.35, p < .001) and adult IQ (rpart,88 = .33, p = .001) even after controlling for global gray matter and white matter volume. In a path analytic model, cBF volume significantly mediated the association between neonatal complications and adult cognitive deficits. CONCLUSIONS We provide first-time evidence in humans that cBF integrity is impaired after premature birth and links neonatal complications with long-term cognitive outcome. Data suggest that cholinergic system abnormalities may play a relevant role for long-term neurocognitive impairments associated with premature delivery.
Collapse
Affiliation(s)
| | - Lukas Scheef
- Functional Neuroimaging Group, University Hospital Bonn, Bonn
| | - Josef Bäuml
- Department of Neuroradiology, Technische Universität München, Munich, Germany; Technische Universität München-Neuroimaging Center of Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Chun Meng
- Department of Neuroradiology, Technische Universität München, Munich, Germany; Technische Universität München-Neuroimaging Center of Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Marcel Daamen
- Functional Neuroimaging Group, University Hospital Bonn, Bonn
| | - Nicole Baumann
- Department of Psychology, University of Warwick, Coventry, United Kingdom
| | - Claus Zimmer
- Department of Neuroradiology, Technische Universität München, Munich, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Peter Bartmann
- Department of Radiology, and Department of Neonatology, University Hospital Bonn, Bonn
| | - Henning Boecker
- Functional Neuroimaging Group, University Hospital Bonn, Bonn
| | - Dieter Wolke
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Afra Wohlschläger
- Technische Universität München-Neuroimaging Center of Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Sorg
- Department of Psychiatry, Technische Universität München, Munich, Germany
| |
Collapse
|
7
|
Ota A, Ikeda T, Xia XY, Xia YX, Ikenoue T. Hypoxic-Ischemic Tolerance Induced by Hyperthermic Pretreatment in Newborn Rats. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155760000700204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Arturo Ota
- Department of Obstetrics and Gynecology, Miyazaki Medical College, Miyazaki, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Miyazaki Medical College, Miyazaki Japan
| | | | | | - Tsuyomu Ikenoue
- Department of Obstetrics and Gynecology, Miyazaki Medical College, Miyazaki, Japan
| |
Collapse
|
8
|
Seo H, Lim KH, Choi JH, Jeong SM. Similar neuroprotective effects of ischemic and hypoxic preconditioning on hypoxia-ischemia in the neonatal rat: a proton MRS study. Int J Dev Neurosci 2013; 31:616-23. [PMID: 23958850 DOI: 10.1016/j.ijdevneu.2013.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/05/2013] [Accepted: 08/05/2013] [Indexed: 10/26/2022] Open
Abstract
The aim of this study was to evaluate the effect of ischemic and hypoxic preconditioning on hypoxia-ischemia (HI) in the neonatal rat. Seven-day-old Sprague-Dawley rats were divided into four groups: control, sham, ischemic preconditioning, and hypoxic preconditioning. Ischemic preconditioning with a 10-min occlusion of the right carotid artery and hypoxic preconditioning with 4-h of hypoxia (8% oxygen) were performed 24-h before HI. For HI, all rats underwent right carotid artery ligature, followed by 2.5-h of hypoxia. Proton magnetic resonance spectroscopy ((1)H MRS) and TUNEL staining were evaluated at 1 and 7 days after HI. At 2 weeks after HI, all rats were sacrificed for morphological analysis. The lipid (Lip), N-acetyl aspartate (NAA), creatine (Cr), and choline-ratios were calculated and compared with TUNEL staining and brain morphologies. Both the ischemic and hypoxic preconditioning groups showed lower Lip/NAA and Lip/Cr ratios and morphological scores, and fewer TUNEL-positive cells than the control and sham groups (P < 0.05). There were no significant differences between the two preconditioning groups. In addition, the ratios correlated with the TUNEL staining and the degrees of morphological changes in all of the groups (P < 0.05). These results suggest that ischemic and hypoxic preconditioning in neonatal rats with HI similarly attenuate brain injury. Moreover, Lip/NAA and Lip/Cr ratios may be used as markers for assessing the extent of brain damage.
Collapse
Affiliation(s)
- Hyungseok Seo
- Department of Anesthesiology and Pain Medicine, College of Medicine, University of Ulsan, Asan Medical Center, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
9
|
Furukawa S, Sameshima H, Yang L, Ikenoue T. Activation of acetylcholine receptors and microglia in hypoxic-ischemic brain damage in newborn rats. Brain Dev 2013; 35:607-13. [PMID: 23141185 DOI: 10.1016/j.braindev.2012.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/10/2012] [Accepted: 10/14/2012] [Indexed: 02/05/2023]
Abstract
OBJECTIVE We previously showed that acetylcholine receptor (AChR) agonist reduced hypoxic-ischemic brain damage in the newborn rats. To further investigated the interaction between hypoxia and chorinergic anti-inflammatory pathway, we examined the effect of AChR antagonist on brain damage and to see the relation between microglial activation and protective effect of AChR agonist. STUDY DESIGN Seven-day-old Wistar rats were divided into 2 groups, one receiving AChR antagonists to see if they have deleterious effects on hypoxic-ischemic brain damage, and the other receiving AChR agonist, carbachol, to investigate the emergence of microglia in the hippocampus. Rats were subjected to left carotid artery ligation followed by 8% hypoxia. Brains were analyzed histologically and immunohistochemically. RESULTS Antagonists of AChRs significantly enhanced brain damage in 1-h hypoxia-ischemia. In particular, the nicotinic AChR antagonist showed a marked enhancement of brain damage compared to the saline controls (p<0.01). The hippocampal CA1 was most vulnerable to any AChR antagonists, while the cortex was least vulnerable and only responsive to a higher dose of non-selective nAChR antagonist. Carbachol showed significantly less accumulation of microglia in the hippocampus than the saline controls (p<0.01) in hypoxia-ischemia. CONCLUSION An AchR-responsive pathway in the brain plays an important role in modifying perinatal brain damage, in which microglial accumulation may be involved.
Collapse
Affiliation(s)
- Seishi Furukawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| | | | | | | |
Collapse
|
10
|
Northington FJ, Chavez-Valdez R, Martin LJ. Neuronal cell death in neonatal hypoxia-ischemia. Ann Neurol 2011; 69:743-58. [PMID: 21520238 DOI: 10.1002/ana.22419] [Citation(s) in RCA: 269] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Perinatal hypoxic-ischemic encephalopathy (HIE) is a significant cause of mortality and morbidity in infants and young children. Therapeutic opportunities are very limited for neonatal and pediatric HIE. Specific neural systems and populations of cells are selectively vulnerable in HIE; however, the mechanisms of degeneration are unresolved. These mechanisms involve oxidative stress, excitotoxicity, inflammation, and the activation of several different cell death pathways. Decades ago the structural and mechanistic basis of the cellular degeneration in HIE was thought to be necrosis. Subsequently, largely due to advances in cell biology and to experimental animal studies, emphasis has been switched to apoptosis or autophagy mediated by programmed cell death (PCD) mechanisms as important forms of degeneration in HIE. We have conceptualized based on morphological and biochemical data that this degeneration is better classified according to an apoptosis-necrosis cell death continuum and that programmed cell necrosis has prominent contribution in the neurodegeneration of HIE in animal models. It is likely that neonatal HIE evolves through many cell death chreodes influenced by the dynamic injury landscape. The relevant injury mechanisms remain to be determined in human neonatal HIE, though preliminary work suggests a complexity in the cell death mechanisms greater than that anticipated from experimental animal models. The accurate identification of the various cell death chreodes and their mechanisms unfolding within the immature brain matrix could provide fresh insight for developing meaningful therapies for neonatal and pediatric HIE.
Collapse
Affiliation(s)
- Frances J Northington
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | |
Collapse
|
11
|
Furukawa S, Sameshima H, Yang L, Ikenoue T. Acetylcholine receptor agonist reduces brain damage induced by hypoxia-ischemia in newborn rats. Reprod Sci 2010; 18:172-9. [PMID: 20959640 DOI: 10.1177/1933719110385129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The newborn rat model has been developed to elucidate the mechanism and management of perinatal brain damage. Our study hypothesis is that an acetylcholine receptor agonist (carbachol) reduces hypoxia-ischemia (HI)-induced brain damage in a well-established newborn rat model. STUDY DESIGN 7-day-old Wistar rats were divided into 3 groups at random: carbachol preinjection and HI (Carb/HI), saline preinjection and HI (Saline/HI), and only HI (HI). Rats were subjected to left carotid artery ligation followed by 2 hours of hypoxia (8% oxygen). We injected carbachol or saline before hypoxic loading. After 7 days, we checked for brain damage. RESULTS In the cerebral cortex, 25% of the Carb/HI group showed mild neural damage, and the remaining 75% showed no damage. In contrast, more than 80% of the Saline/HI and HI groups had severe neural damage. Similarly, neural damage significantly decreased in Carb/HI compared with Saline/HI and HI for CA1, CA2, CA3, and the dentate gyrus of hippocampal regions. CONCLUSION Acetylcholine receptor agonist has a potent effect by reducing perinatal brain damage induced by HI in newborn rats.
Collapse
Affiliation(s)
- Seishi Furukawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| | | | | | | |
Collapse
|
12
|
Abstract
BACKGROUND The present study was performed to investigate the effect of neonatal hypoxic-ischemic encephalopathy (HIE) on the neurotransmitter neurokinin A (NKA) and determine its relation to the severity of neonatal hypoxia. METHODS Eighteen neonates suffering from HIE were compared to 10 clinically healthy full-term neonates acting as the control group. Maternal history of each neonate was collected, then deliveries were attended, resuscitation details including the Apgar score and thorough clinical examination of the neonates were performed. Routine laboratory work-up was done for the enrolled neonates, including complete blood count and C-reactive protein as well as estimation of NKA by enzyme-linked immunosorbent assay in the cord blood and after clinical stabilization. RESULTS NKA was significantly lower in HIE patients compared to the controls at delivery with improvement in the follow-up sample. Additionally, the maximum decrease was detected in the neonates who suffered severe hypoxia compared to those who suffered mild hypoxia. Significant positive correlations were demonstrated between NKA at birth and Apgar scores at the 10th and 15th min. Regression showed that stage of HIE was the strongest determinant factor for the level of NKA at birth. CONCLUSION NKA levels are decreased in HIE and this is more profound in the severe degrees of hypoxia compared to the mild ones. This emphasizes its role in pathogenesis of HIE and further proves that an imbalance in the central neuropeptide system results from HIE in the neonatal period.
Collapse
Affiliation(s)
- May F Nassar
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | | | | | | |
Collapse
|
13
|
Hefner KE, Butler R, Ravindra A, Ahmad Z, Molina D, Turner CP. Dipyridamole promotes changes in calbindin-D28k and tyrosine hydroxylase expression in neonatal rats. Neonatology 2007; 91:222-32. [PMID: 17568153 DOI: 10.1159/000098169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Accepted: 08/28/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND Perinatal hypoxia alters the concentration of many neurochemicals in the brain, including adenosine, and promotes central nervous system (CNS) disorders in human infants such as periventricular leukomalacia or encephalopathy. OBJECTIVE Using the postnatal rat as a model of perinatal human development, we examined the effects of sustained increases in brain adenosine on CNS regions thought to be involved with both planning and execution of motor activity. METHODS To simulate hypoxia-induced changes in adenosine, Sprague-Dawley rats were injected twice daily from postnatal day (P) 3 to P14, with the adenosine uptake inhibitor dipyridamole (DIP) or the A(1) adenosine receptor agonist N(6)-cyclopentyladenosine (CPA). Vehicle-injected animals served as controls. Immunohistochemical and morphological analyses were then performed to examine the expression of calbindin D-28k (CB) and the thickness of the external granule cell layer (eGL) in the cerebellum. Additionally tyrosine hydroxylase (TH) expression in the caudate putamen and ventricular size were also examined. RESULTS In the cerebellum, both DIP and CPA reduced the number of CB-positive Purkinje cells as well as decreased the thickness of the eGL compared to vehicle. In the caudate putamen we found that DIP but not CPA decreased TH expression when compared to vehicle. Neither agent significantly altered ventricular size when compared to vehicle. CONCLUSIONS These observations suggest that elevations in brain adenosine, which can occur following hypoxia, leads to both neurochemical and cellular changes in regions of the brain which control the planning and execution of motor activity. Thus, therapeutic strategies that target brain regions most sensitive to adenosine may prevent or control at least some of the CNS damage observed following perinatal hypoxia.
Collapse
Affiliation(s)
- K E Hefner
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1010, USA
| | | | | | | | | | | |
Collapse
|
14
|
Spandou E, Papadopoulou Z, Soubasi V, Karkavelas G, Simeonidou C, Pazaiti A, Guiba-Tziampiri O. Erythropoietin prevents long-term sensorimotor deficits and brain injury following neonatal hypoxia-ischemia in rats. Brain Res 2005; 1045:22-30. [PMID: 15910759 DOI: 10.1016/j.brainres.2005.03.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 02/27/2005] [Accepted: 03/01/2005] [Indexed: 11/29/2022]
Abstract
Perinatal asphyxia accounts for behavioral dysfunctions that often manifest as sensorimotor, learning or memory disabilities throughout development and into maturity. Erythropoietin (Epo) has been shown to exert neuroprotective effects in different models of brain injury including experimental models of perinatal asphyxia. However, the effect of Epo on functional abilities following cerebral hypoxia-ischemia (HI) in neonatal rats is not known. The aim of the present study is to investigate the effect of Epo on sensorimotor deficits and brain injury induced by hypoxia-ischemia. Seven-day-old rats underwent unilateral, permanent carotid artery ligation followed by 1 h of hypoxia. Epo was administered as a single dose immediately after the hypoxic insult (2000 U/kg). The neuroprotective effect of Epo was evaluated at postnatal day 42 by using a battery of behavioral tests and histological analysis. The results of the present study suggest that Epo treatment immediately after HI insult significantly facilitated recovery of sensorimotor function. Consistently, histopathological evaluation demonstrated that Epo significantly attenuated brain injury and preserved the integrity of cerebral cortex. These findings indicate that long-term neuroprotective effect of Epo on neonatal HI-induced brain injury might be associated with the preservation of sensorimotor functions.
Collapse
Affiliation(s)
- Evangelia Spandou
- Department of Physiology and Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | | | | | | | | | | | | |
Collapse
|
15
|
Ma D, Hossain M, Chow A, Arshad M, Battson RM, Sanders RD, Mehmet H, Edwards AD, Franks NP, Maze M. Xenon and hypothermia combine to provide neuroprotection from neonatal asphyxia. Ann Neurol 2005; 58:182-93. [PMID: 16049939 DOI: 10.1002/ana.20547] [Citation(s) in RCA: 216] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Perinatal asphyxia can result in neuronal injury with long-term neurological and behavioral consequences. Although hypothermia may provide some modest benefit, the intervention itself can produce adverse consequences. We have investigated whether xenon, an antagonist of the N-methyl-D-aspartate subtype of the glutamate receptor, can enhance the neuroprotection provided by mild hypothermia. Cultured neurons injured by oxygen-glucose deprivation were protected by combinations of interventions of xenon and hypothermia that, when administered alone, were not efficacious. A combination of xenon and hypothermia administered 4 hours after hypoxic-ischemic injury in neonatal rats provided synergistic neuroprotection assessed by morphological criteria, by hemispheric weight, and by functional neurological studies up to 30 days after the injury. The protective mechanism of the combination, in both in vitro and in vivo models, involved an antiapoptotic action. If applied to humans, these data suggest that low (subanesthetic) concentrations of xenon in combination with mild hypothermia may provide a safe and effective therapy for perinatal asphyxia.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Apoptosis/physiology
- Asphyxia/prevention & control
- Blotting, Western/methods
- Body Temperature/physiology
- Brain/drug effects
- Brain/pathology
- Caspase 3
- Caspases/metabolism
- Cells, Cultured
- Cerebral Cortex/cytology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Embryo, Mammalian
- Excitatory Amino Acid Antagonists/pharmacology
- Flow Cytometry/methods
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/radiation effects
- Glucose/deficiency
- Hypothermia
- Hypoxia
- Hypoxia-Ischemia, Brain/physiopathology
- Hypoxia-Ischemia, Brain/therapy
- Immunohistochemistry/methods
- Indoles/pharmacology
- L-Lactate Dehydrogenase/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Motor Activity/drug effects
- Motor Activity/physiology
- Necrosis/therapy
- Neurons/drug effects
- Neurons/physiology
- Neuroprotective Agents/therapeutic use
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Rats
- Rats, Sprague-Dawley
- Recovery of Function/drug effects
- Temperature
- Time Factors
- Xenon/therapeutic use
- bcl-2-Associated X Protein
- bcl-X Protein
Collapse
Affiliation(s)
- Daqing Ma
- Department of Anaesthetics and Intensive Care, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Eisenhofer G, Kopin IJ, Goldstein DS. Catecholamine metabolism: a contemporary view with implications for physiology and medicine. Pharmacol Rev 2005; 56:331-49. [PMID: 15317907 DOI: 10.1124/pr.56.3.1] [Citation(s) in RCA: 660] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This article provides an update about catecholamine metabolism, with emphasis on correcting common misconceptions relevant to catecholamine systems in health and disease. Importantly, most metabolism of catecholamines takes place within the same cells where the amines are synthesized. This mainly occurs secondary to leakage of catecholamines from vesicular stores into the cytoplasm. These stores exist in a highly dynamic equilibrium, with passive outward leakage counterbalanced by inward active transport controlled by vesicular monoamine transporters. In catecholaminergic neurons, the presence of monoamine oxidase leads to formation of reactive catecholaldehydes. Production of these toxic aldehydes depends on the dynamics of vesicular-axoplasmic monoamine exchange and enzyme-catalyzed conversion to nontoxic acids or alcohols. In sympathetic nerves, the aldehyde produced from norepinephrine is converted to 3,4-dihydroxyphenylglycol, not 3,4-dihydroxymandelic acid. Subsequent extraneuronal O-methylation consequently leads to production of 3-methoxy-4-hydroxyphenylglycol, not vanillylmandelic acid. Vanillylmandelic acid is instead formed in the liver by oxidation of 3-methoxy-4-hydroxyphenylglycol catalyzed by alcohol and aldehyde dehydrogenases. Compared to intraneuronal deamination, extraneuronal O-methylation of norepinephrine and epinephrine to metanephrines represent minor pathways of metabolism. The single largest source of metanephrines is the adrenal medulla. Similarly, pheochromocytoma tumor cells produce large amounts of metanephrines from catecholamines leaking from stores. Thus, these metabolites are particularly useful for detecting pheochromocytomas. The large contribution of intraneuronal deamination to catecholamine turnover, and dependence of this on the vesicular-axoplasmic monoamine exchange process, helps explain how synthesis, release, metabolism, turnover, and stores of catecholamines are regulated in a coordinated fashion during stress and in disease states.
Collapse
Affiliation(s)
- Graeme Eisenhofer
- Clinical Neurocardiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Dr., MSC-1620, Bethesda, MD 20892-1620, USA.
| | | | | |
Collapse
|
17
|
Zaidi AU, Bessert DA, Ong JE, Xu H, Barks JDE, Silverstein FS, Skoff RP. New oligodendrocytes are generated after neonatal hypoxic-ischemic brain injury in rodents. Glia 2004; 46:380-90. [PMID: 15095368 DOI: 10.1002/glia.20013] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neonatal hypoxic-ischemic (HI) white matter injury is a major contributor to chronic neurological dysfunction. Immature oligodendrocytes (OLGs) are highly vulnerable to HI injury. As little is known about in vivo OLG repair mechanisms in neonates, we studied whether new OLGs are generated after HI injury in P7 rats. Rats received daily BrdU injections at P12-14 or P21-22 and sacrificed at P14 to study the level of cell proliferation or at P35 to permit dividing OLG precursors to differentiate. In P14 HI-injured animals, the number of BrdU+ cells in the injured hemisphere is consistently greater than controls. At P35, sections were double-labeled for BrdU and markers for OLGs, astrocytes, and microglia. Double-labeled BrdU+/myelin basic protein+ and BrdU+/carbonic anhydrase+ OLGs are abundant in the injured striatum, corpus callosum, and the infarct core. Quantitative studies show four times as many OLGs are generated from P21-35 in HI corpora callosa than controls. Surprisingly, the infarct core contains many newly generated OLGs in addition to hypertrophied astrocytes and activated microglia. These glia and non-CNS cells may stimulate OLG progenitor proliferation or induce their migration. At P35, astrogliosis and microgliosis are dramatic ipsilaterally but only a few microglia and some astrocytes are BrdU+. This finding indicates microglial and astrocytic hyperplasia occurs shortly after HI but before the P21 BrdU injections. Although the neonatal brain undergoes massive cell death and atrophy the first week after injury, it retains the potential to generate new OLGs up to 4 weeks after injury within and surrounding the infarct.
Collapse
Affiliation(s)
- Aliya U Zaidi
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Yang L, Sameshima H, Ikeda T, Ikenoue T. Lipopolysaccharide administration enhances hypoxic-ischemic brain damage in newborn rats. J Obstet Gynaecol Res 2004; 30:142-7. [PMID: 15009619 DOI: 10.1111/j.1447-0756.2003.00174.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM To determine whether inflammation and hypoxic-ischemic insult (HI) act additively to cause brain damage in perinatal animals by examining the dose-response effect of lipopolysaccharide (LPS) administration on HI insult in neonatal rat pups. METHODS Seven-day-old Wistar rats (n = 119) were divided into three groups: (i) a group that received a pre-injection of LPS and HI (LPS/HI, 1 mg/kg, n = 31; 0.5 mg/kg, n = 20; 0.1 mg/kg, n = 17); (ii) a group that received a pre-injection of saline and HI (saline/HI, n = 35); and (iii) those that received LPS alone (1 mg/kg, n = 16). At 4 h after the injection, rat pups from groups (i) and (11) were exposed to unilateral carotid artery ligation, followed by 1 h of hypoxia (8% oxygen in 92% nitrogen) at 33 degrees C. Seven days after the insult, they were sacrificed and their brains removed for histological study. Neuronal damage was categorized as mild, < or =25%; moderate, 25-50%; and severe, > or =50% of surface area on a single section. RESULTS Mortality rate during the experiment was significantly increased in the 1 mg/kg of LPS/HI group (12 of 31, 39%) compared with the saline/HI group (0%). No neuronal damage was observed in the LPS only group. However, when LPS was added to HI, neuronal loss in the cerebral cortex and hippocampus was significantly increased in a dose-response manner. CONCLUSION LPS potentiates hypoxic-ischemic insult in a dose-dependent fashion to cause brain damage in neonatal rats.
Collapse
Affiliation(s)
- Li Yang
- Department of Obstetrics and Gynecology, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan
| | | | | | | |
Collapse
|
19
|
Xia YX, Sameshima H, Ikeda T, Higo T, Ikenoue T. Cerebral blood flow distribution and hypoxic-ischemic brain damage in newborn rats. J Obstet Gynaecol Res 2002; 28:320-6. [PMID: 12512930 DOI: 10.1046/j.1341-8076.2002.00063.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Our purpose was to assess the cerebral blood flow distribution and resulting grade of hypoxicischemic brain damage in newborn rats. METHODS Seven-day-old Wistar rats (n = 75) underwent left common carotid artery ligation followed by 2 hours hypoxia (8% oxygen in nitrogen) at 33 degrees C. The control animals were exposed to hypoxia without ligation (n = 8). Colored microspheres of 15 microm in diameter were administered into the left cardiac ventricle percutaneously at the end of hypoxia. They were killed 24 hours after induced injury. Brain sections 2 mm in thickness were removed for microtubule-associated protein 2 (MAP-2) staining, and remaining parts were separated into left and right hemispheres for counting the microspheres. The blood flow distribution to the ligated side was expressed as the difference from the non-ligated control side. Severity of MAP-2 disappearance was ranked as normal, mild or severe. RESULTS In the control rats, there was no loss of MAP-2 staining. The blood flow equally distributed into both cerebral hemispheres. The cerebral blood flow distribution on the side of carotid artery ligation decreased by 44.7 +/- 21.9% in the mildly damaged group and 65.8 +/- 16.8% in the severely damaged group. CONCLUSION The greater the percentage difference of blood flow distribution from the non-ligated side, the more severe the brain damage.
Collapse
Affiliation(s)
- Yi Xin Xia
- Department of Obstetrics and Gynecology, Miyazaki Medical College, Miyazaki, Japan
| | | | | | | | | |
Collapse
|
20
|
Han BH, Xu D, Choi J, Han Y, Xanthoudakis S, Roy S, Tam J, Vaillancourt J, Colucci J, Siman R, Giroux A, Robertson GS, Zamboni R, Nicholson DW, Holtzman DM. Selective, reversible caspase-3 inhibitor is neuroprotective and reveals distinct pathways of cell death after neonatal hypoxic-ischemic brain injury. J Biol Chem 2002; 277:30128-36. [PMID: 12058036 DOI: 10.1074/jbc.m202931200] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypoxia-ischemia (H-I) in the developing brain results in brain injury with prominent features of both apoptosis and necrosis. A peptide-based pan-caspase inhibitor is neuroprotective against neonatal H-I brain injury, suggesting a central role of caspases in brain injury. Because previously studied peptide-based caspase inhibitors are not potent and are only partially selective, the exact contribution of specific caspases and other proteases to injury after H-I is not clear. In this study, we explored the neuroprotective effects of a small, reversible caspase-3 inhibitor M826. M826 selectively and potently inhibited both caspase-3 enzymatic activity and apoptosis in cultured cells in vitro. In a rat model of neonatal H-I, M826 blocked caspase-3 activation and cleavage of its substrates, which begins 6 h and peaks 24 h after H-I. Although M826 significantly reduced DNA fragmentation and brain tissue loss, it did not prevent calpain activation in the cortex. This activation, which is associated with excitotoxic/necrotic cell injury, occurred within 30 min to 2 h after H-I even in the presence of M826. Similar to calpain activation, we found evidence of caspase-2 processing within 30 min to 2 h after H-I that was not affected by M826. Caspase-2 processing appeared to be secondary to calpain-mediated cleavage and was not associated with caspase-2 activation. These data suggest that caspase-3 specifically contributes to delayed cell death and brain injury after neonatal H-I and that calpain activation is associated with and likely a marker for the early component of excitotoxic/necrotic brain injury previously demonstrated in this model.
Collapse
Affiliation(s)
- Byung Hee Han
- Department of Neurology, Washington University, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Van de Berg WDJ, Schmitz C, Steinbusch HWM, Blanco CE. Perinatal asphyxia induced neuronal loss by apoptosis in the neonatal rat striatum: a combined TUNEL and stereological study. Exp Neurol 2002; 174:29-36. [PMID: 11869031 DOI: 10.1006/exnr.2001.7855] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Perinatal asphyxia can lead to cell damage in various regions of the brain, such as the neostriatum. In this study, we investigated the mechanism of cell death that leads to neuron loss in the neostriatum of rat pups. Asphyxia was induced by immersing fetus-containing uterus horns in a water bath at 37 degrees C for 20 min. This led to an increase in mortality rate (+/- 40%) compared to control pups (0%). TUNEL-positive cell profiles were visible in all groups at postnatal day (P) 2, P8, and P15, peaking at P8. A significant increase of 40% at P8 and 45% at P15 in the number of TUNEL-positive cell profiles was observed in asphyctic rats compared to control rats. Nuclear condensation and fragmentation was visible with the DNA stain Hoechst 33342. Furthermore, laser-scanning confocal microscopy showed multiple DNA fragments in TUNEL-positive cell profiles. We found a decrease of 16% in the total number of striatal neurons in the asphyctic pups compared to the control pups at 21 days postasphyxia using stereology. These data show that asphyxia causes exaggerated apoptotic cell death during the first week of life and as a consequence a small amount of neuron loss in the neostriatum.
Collapse
|
22
|
Abstract
In the premature infant, hypoxic-ischemic damage to the cerebral white matter [periventricular leukomalacia (PVL)] is a common and leading cause of brain injury that often results in chronic neurologic disability from cerebral palsy. The cellular basis for the propensity of white matter injury to occur in the developing brain and the greater resistance of the adult white matter to similar injury remains unknown. By using a neonatal rat model of hypoxic-ischemic injury, we found that the mechanism of perinatal white matter injury involved maturation-dependent vulnerability in the oligodendroctye (OL) lineage. The timing of appearance of late OL progenitors was the major developmental factor that accounted for the susceptibility of the neonatal white matter to injury. Late OL progenitors were the major OL lineage stage killed by apoptosis, whereas early OL progenitors and more mature OLs were highly resistant. The density of pyknotic late OL progenitors was significantly increased in the ischemic hemisphere (67 +/- 31 cells/mm2) versus the control hemisphere (2.2 +/- 0.4 cells/mm2; mean +/- SEM; p = 0.05), which resulted in the death of 72 +/- 6% of this OL stage. Surviving late OL progenitors displayed a reactive response in which an increase in cell density was accompanied by accelerated maturation to a P27/kip1-positive oligodendrocyte. Because we showed recently that late OL progenitors populate human cerebral white matter during the high risk period for PVL (Back et al., 2001), maturation-dependent vulnerability of OL progenitors to hypoxia-ischemia may underlie the selective vulnerability to PVL of the white matter in the premature infant.
Collapse
|
23
|
Gibson ME, Han BH, Choi J, Knudson CM, Korsmeyer SJ, Parsadanian M, Holtzman DM. BAX Contributes to Apoptotic-Like Death Following Neonatal Hypoxia-Ischemia: Evidence for Distinct Apoptosis Pathways. Mol Med 2001. [DOI: 10.1007/bf03401871] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
24
|
Chang HM, Lue JH, Wen CY, Shieh JY. Axotomy along with hypoxia enhances the neuronal NADPH-d/NOS expression in lower brain stem motor neurons of adult rats. Exp Neurol 2001; 171:116-26. [PMID: 11520126 DOI: 10.1006/exnr.2001.7731] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study was aimed to determine whether axotomy coupled with hypoxia would exert a more profound effect on injury-induced neuronal nitric oxide synthase (NOS) expression. In this connection, the vagus and the hypoglossal nerves of adult rats were transected unilaterally in the same animal, and half of the operated animals were subjected to hypoxia treatment. Both the neuronal NOS immunohistochemistry and the nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) histochemistry were used to assess the neuronal NOS expression. The present results have shown that the number of NADPH-d/NOS-positive [NADPH-d/NOS(1)] neurons in the hypoglossal nucleus (HN) peaked at 14 days after axotomy, while that in dorsal motor nucleus of vagus (DMN) and nucleus ambiguus (NA) was progressively increased up to 60 days. The up-regulation of NADPH-d/NOS in HN and DMN was more pronounced in hypoxic than in normoxic animals, a feature that was not evident in the NA. Quantitative analysis showed that the number of surviving motoneurons in normoxic animals was significantly higher than those subjected to hypoxia at 14 days postaxotomy in HN and at all postaxotomy time points in DMN. The difference may be attributed to their different functional components. Since O2 deprivation leads to poor cellular function, the stronger expression of NADPH-d/NOS and the more drastic neuronal loss following nerve transection in the hypoxic animals compared with the controls suggest that hypoxia plays an important role in peripheral neuropathies in which NO is implicated.
Collapse
Affiliation(s)
- H M Chang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, 100, Republic of China
| | | | | | | |
Collapse
|
25
|
Abstract
Birth asphyxia can cause moderate to severe brain injury. It is unclear to what degree apoptotic or necrotic mechanisms of cell death account for damage after neonatal hypoxia-ischemia (HI). In a 7-d-old rat HI model, we determined the contributions of apoptosis and necrosis to neuronal injury in adjacent Nissl-stained, hematoxylin and eosin-stained, and terminal deoxynucleotidyl transferase-mediated UTP nick end-labeled sections. We found an apoptotic-necrotic continuum in the morphology of injured neurons in all regions examined. Eosinophilic necrotic neurons, typical in adult models, were rarely observed in neonatal HI. Electron microscopic analysis showed "classic" apoptotic and necrotic neurons and "hybrid" cells with intermediate characteristics. The time course of apoptotic injury varied regionally. In CA3, dentate gyrus, medial habenula, and laterodorsal thalamus, the density of apoptotic cells was highest at 24-72 hr after HI and then declined. In contrast, densities remained elevated from 12 hr to 7 d after HI in most cortical areas and in the basal ganglia. Temporal and regional patterns of neuronal death were compared with expression of caspase-3, a cysteine protease involved in the execution phase of apoptosis. Immunocytochemical and Western blot analyses showed increased caspase-3 expression in damaged hemispheres 24 hr to 7 d after HI. A p17 peptide fragment, which results from the proteolytic activation of the caspase-3 precursor, was detected in hippocampus, thalamus, and striatum but not in cerebral cortex. The continued expression of activated caspase-3 and the persistence of cells with an apoptotic morphology for days after HI suggests a prolonged role for apoptosis in neonatal hypoxic ischemic brain injury.
Collapse
|
26
|
Almli CR, Levy TJ, Han BH, Shah AR, Gidday JM, Holtzman DM. BDNF protects against spatial memory deficits following neonatal hypoxia-ischemia. Exp Neurol 2000; 166:99-114. [PMID: 11031087 DOI: 10.1006/exnr.2000.7492] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypoxic-ischemic (H-I) brain injury in the human perinatal period often leads to significant long-term neurobehavioral dysfunction in the cognitive and sensory-motor domains. Using a neonatal H-I injury model (unilateral carotid ligation followed by hypoxia) in postnatal day seven rats, previous studies have shown that neurotrophins, such as brain-derived neurotrophic factor (BDNF), can be protective against neural tissue loss. The present study explored potential relationships between neural protective and behavioral protective strategies in this neonatal H-I model by determining if neonatal H-I was associated with behavioral spatial learning and memory deficits and whether the neurotrophin BDNF was protective against both brain injury and spatial learning/memory dysfunction. Postnatal day seven rats received vehicle or BDNF pretreatments (intracerebroventricular injections) followed by H-I or sham treatments and then tested for spatial learning and memory on the simple place task in the Morris water maze from postnatal days 20 to 30, and their brains were histologically analyzed at 4 weeks following treatments. H-I rats with vehicle pretreatment displayed significant tissue loss in the hippocampus (including CA1 neurons), cortex, and striatum, as well as severe spatial memory deficits (e.g., short probe times). BDNF pretreatment resulted in significant protection against both H-I-induced brain tissue losses and spatial memory impairments. These findings indicate that unilateral H-I brain injury in a neonatal rodent model is associated with cognitive deficits, and that BDNF pretreatment is protective against both brain injury and spatial memory impairment.
Collapse
Affiliation(s)
- C R Almli
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63108-2212, USA
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Neurotrophins activate several different intracellular signaling pathways that in some way exert neuroprotective effects. In vitro studies of sympathetic and cerebellar granule neurons have demonstrated that the survival effects of neurotrophins can be mediated via activation of the phosphatidylinositol 3-kinase (PI3-kinase) pathway. Neurotrophin-mediated protection of other neuronal types in vitro can be mediated via the extracellular signal-related protein kinase (ERK) pathway. Whether either of these pathways contributes to the neuroprotective effects of neurotrophins in the brain in vivo has not been determined. Brain-derived neurotrophic factor (BDNF) is markedly neuroprotective against neonatal hypoxic-ischemic (H-I) brain injury in vivo. We assessed the role of the ERK and PI3-kinase pathways in neonatal H-I brain injury in the presence and absence of BDNF. Intracerebroventricular administration of BDNF to postnatal day 7 rats resulted in phosphorylation of ERK1/2 and the PI3-kinase substrate AKT within minutes. Effects were greater on ERK activation and occurred in neurons. Pharmacological inhibition of ERK, but not the PI3-kinase pathway, inhibited the ability of BDNF to block H-I-induced caspase-3 activation and tissue loss. These findings suggest that neuronal ERK activation in the neonatal brain mediates neuroprotection against H-I brain injury, a model of cerebral palsy.
Collapse
|
28
|
Chalon S, Cantagrel S. [Monoaminergic neurotransmission and brain dysfunctions in the newborn]. Arch Pediatr 2000; 6 Suppl 2:197s-198s. [PMID: 10370479 DOI: 10.1016/s0929-693x(99)80411-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- S Chalon
- Inserm U316, Faculté de pharmacie, Tours, France
| | | |
Collapse
|
29
|
Adelson PD, Dixon CE, Kochanek PM. Long-term dysfunction following diffuse traumatic brain injury in the immature rat. J Neurotrauma 2000; 17:273-82. [PMID: 10776912 DOI: 10.1089/neu.2000.17.273] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Children often suffer sustained cognitive dysfunction after severe diffuse traumatic brain injury (TBI). To study the effects of diffuse injury in the immature brain, we developed a model of severe diffuse impact (DI) acceleration TBI in immature rats and previously described the early motor and cognitive dysfunction posttrauma. In the present study, we investigated the long-term functional ability after DI (150 gm/2 m) compared to sham in the immature (PND 17) rat. Beam balance and inclined plane latencies were measured daily for 10 days after injury to assess gross vestibulomotor function. The Morris water maze (MWM) paradigm was evaluated monthly up to 3 months after DI and sham injuries. Reduced latencies on the balance beam and inclined plane were observed in DI rats (p < 0.05 vs. sham [n = 10 per group]) at 24 h and persisted for 10 days postinjury. DI produced sustained MWM performance deficits (p < 0.05 vs. sham) as indicated by the greater latencies to find the hidden platform remarkably through 90 days after injury. Lastly, the brain and body weights of the injured animals were less than sham (p < 0.05) after 3 months. We conclude that a diffuse TBI in the immature rat: (a) created a consistent, marked, but reversible motor deficit up to 10 days following injury; (b) produced a long-term, sustained performance deficit in the MWM up to 3 months posttrauma; and (c) affected body and brain weight gain in the developing rat through 3 months after injury. This TBI model should be useful for the testing of novel therapies and their effect on long-term outcome and development in the immature rat.
Collapse
Affiliation(s)
- P D Adelson
- Department of Pediatric Neurosurgery, Children's Hospital of Pittsburgh, Pennsylvania 15213-2583, USA.
| | | | | |
Collapse
|
30
|
Ikeda T, Ikenoue T, Xia XY, Xia YX. Important role of 72-kd heat shock protein expression in the endothelial cell in acquisition of hypoxic-ischemic tolerance in the immature rat. Am J Obstet Gynecol 2000; 182:380-6. [PMID: 10694341 DOI: 10.1016/s0002-9378(00)70228-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Hypoxic-ischemic tolerance can be induced in neonatal rats through hyperthermic preconditioning. The purposes of this study were to determine the interval between hyperthermic preconditioning and a subsequent hypoxic-ischemic insult that would provide optimal neuroprotection against the insult and to examine the relationship between tolerance induction and heat shock protein expression. STUDY DESIGN On postnatal day 7 Wistar rat pups were separated into the following 2 groups: a heated group (those exposed to 15 minutes of hyperthermic pretreatment at a brain temperature of 41.5 degrees C-42.0 degrees C) and an unheated control group. At 6, 12, 24, 48, and 72 hours after the hyperthermic stress, rats from both groups were exposed to left carotid artery ligation followed by 2 hours of hypoxia (8% oxygen and 92% nitrogen) at 33 degrees C. Twenty animals from each group were used at each time point. All rats were killed at 1 week after hypoxia-ischemia, at which time the brains were processed and neuronal damage in the cortex and hippocampus was assessed histologically. Another set of 7-day-old rats (n = 30) was studied immunohistochemically at 6, 12, 24, 48, and 72 hours after the same hyperthermic treatment. Expression of 72-kd heat shock protein was measured in neuronal, glial, and vascular endothelial cells. RESULTS Hyperthermia-induced hypoxic-ischemic tolerance was observed at 6, 12, and 24 hours but not at 48 and 72 hours after hyperthermic preconditioning. Heat shock protein 72 expression in the vascular endothelial cells, rather than in the glial or neuronal cells, was most strongly associated with hypoxic-ischemic tolerance. CONCLUSION These findings suggest that heat shock protein 72 in endothelial cells plays an important role in the acquisition of hypoxic-ischemic tolerance at postnatal day 7, a time when maximal angiogenesis occurs and the blood-brain barrier is still immature.
Collapse
Affiliation(s)
- T Ikeda
- Department of Obstetrics and Gynecology, Miayzaki Medical College, Japan
| | | | | | | |
Collapse
|
31
|
Han BH, D'Costa A, Back SA, Parsadanian M, Patel S, Shah AR, Gidday JM, Srinivasan A, Deshmukh M, Holtzman DM. BDNF blocks caspase-3 activation in neonatal hypoxia-ischemia. Neurobiol Dis 2000; 7:38-53. [PMID: 10671321 DOI: 10.1006/nbdi.1999.0275] [Citation(s) in RCA: 227] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Hypoxic-ischemic (H-I) injury to the brain in the perinatal period often leads to significant long-term neurological deficits. In a model of neonatal H-I injury in postnatal day 7 rats, our previous data have shown that cell death with features of apoptosis is prominent between 6 and 24 h after H-I and that neurotrophins, particularly BDNF, can markedly protect against tissue loss. During brain development, caspase-3 is required for normal levels of programmed cell death. Utilizing an antibody specific for the activated form of caspase-3, CM1, we now show that caspase-3 is specifically activated in neuronal cell bodies and their processes beginning at 6 h and peaking 24 h following unilateral carotid ligation and exposure to hypoxia in postnatal day 7 rats. Caspase-3 activation began to occur in cortex at 6 h and in striatum and hippocampus at 12-18 h. Caspase-3 activation was also observed in developing oligodendrocytes. Intracerebroventricular injection of BDNF prior to H-I injury almost completely abolished evidence of H-I-induced caspase-3 activation in vivo. Utilizing a specific molecular marker of an apoptotic pathway, these findings demonstrate that H-I injury to the developing brain is a strong apoptotic stimulus leading to caspase-3 activation, that BDNF can block this process in vivo, and that the ability of BDNF to inhibit caspase activation and subsequent apoptosis likely accounts in large part for its protection against neuronal injury in this model.
Collapse
Affiliation(s)
- B H Han
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Oorschot DE, Black MJ, Rangi F, Scarr E. Is Fos protein expressed by dying striatal neurons after immature hypoxic-ischemic brain injury? Exp Neurol 2000; 161:227-33. [PMID: 10683289 DOI: 10.1006/exnr.1999.7248] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transient induction of mRNA for the immediate-early gene c-fos has been reported following hypoxic-ischemic brain injury in the immature brain. However, no studies have examined the temporal expression of Fos protein, which is the functionally relevant product of c-fos gene expression. Increased expression of Fos protein has been linked to cell death. We therefore examined whether Fos protein is expressed by dying neurons after immature hypoxic-ischemic brain injury. A well characterized immature rat model of hypoxic-ischemic injury at postnatal day (PN) 7 was used. Three hypoxic-ischemic and three normoxic control pups were studied per time point (i.e., 0, 2, 12, 24, 48, and 72 h posttreatment). Expression of Fos within striatal and other neurons was detected immunocytochemically. Fos protein was expressed within dying striatal neurons at 0-12 h after hypoxia-ischemia. However, detection was only seen in 2 of 17 hypoxic-ischemic pups. These 2 pups had >/=80% of their striatal neurons dying within their right, hypoxic-ischemic-exposed hemisphere. Fos protein expression after severe injury may, therefore, be a response to extraordinary or extreme stress. The absence of Fos protein expression in the majority of hypoxic-ischemic pups, which all exhibited striatal neuronal death, suggests that Fos expression is not necessary for cell death to occur. Therapies directed against Fos protein expression may therefore have limited usefulness in immature hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- D E Oorschot
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | | | | | | |
Collapse
|
33
|
Abstract
Experimental data indicate a particular vulnerability of striatal neurons in the developing brain, and together with the idea that the striatum is important for context recognition and behavior, these data have led the author to search for subtle striatal lesions, in the form of biochemical changes, in children who have suffered perinatal adverse events. Evidence is presented to demonstrate that the composition of metabolites in the striatum is altered, primarily in the form of an elevated level of lactate, in human neonates who have suffered various perinatal disorders, such as germinal matrix hemorrhage, intrauterine growth retardation, and asphyxia. An elevated level of lactate suggests tissue hypoxia, which may interfere with the formation of frontostriatal circuits and may play a role in the pathogenesis of the behavioral disturbances observed in a proportion of children with a history of perinatal adverse events.
Collapse
Affiliation(s)
- P B Toft
- Danish Research Center of Magnetic Resonance, Hvidovre
| |
Collapse
|
34
|
Galvin KA, Oorschot DE. Postinjury magnesium sulfate treatment is not markedly neuroprotective for striatal medium spiny neurons after perinatal hypoxia/ischemia in the rat. Pediatr Res 1998; 44:740-5. [PMID: 9803456 DOI: 10.1203/00006450-199811000-00017] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hypoxic/ischemic (H/I) brain injury is thought to be mediated via the N-methyl-D-aspartate receptor complex, which can be blocked by the magnesium ion. Striatal medium spiny neurons abundantly express N-methyl-D-aspartate receptors and are known to be injured after H/I. Thus, the aim of this study was to investigate the effect of postinjury magnesium treatment on the total number of medium spiny neurons in the striatum after perinatal H/I injury in the rat. Anesthetized postnatal day (PN) 7 rats underwent common carotid artery ligation followed 2 h later by exposure to hypoxia for 1.5 h. Contralateral hemispheres served as controls as did animals exposed to normoxia. Immediately after hypoxia or normoxia, the magnesium groups received s.c. injections of 300 mg/kg MgSO4. Control, hypoxic or normoxic animals received NaCl injections. This continued daily until PN13. Eleven matched-for-weight H/I pups were injected in total. A power calculation showed that 11 pups per treatment group would permit detection of a treatment difference of 32% or more. Animals were killed on PN18, and 40-micron serial sections were cut through each entire striatum. The total number of the predominant medium spiny neurons within each striatum was stereologically determined via the use of an unbiased optical dissector/Cavalieri combination. It was found that postinjury magnesium treatment did not improve neuronal survival by 32% or more in the striatum. The results suggest that magnesium treatment after perinatal H/I damage in the rat is not markedly neuroprotective for striatal medium spiny neurons.
Collapse
Affiliation(s)
- K A Galvin
- Department of Anatomy and Structural Biology, University of Otago Medical School, Dunedin, New Zealand
| | | |
Collapse
|
35
|
Ota A, Ikeda T, Abe K, Sameshima H, Xia XY, Xia YX, Ikenoue T. Hypoxic-ischemic tolerance phenomenon observed in neonatal rat brain. Am J Obstet Gynecol 1998; 179:1075-8. [PMID: 9790401 DOI: 10.1016/s0002-9378(98)70218-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Our purpose was to determine whether hypoxic-ischemic brain damage would be protected against by advance conditioning of the animal with 4 hours of hypoxic exposure. STUDY DESIGN Neonatal rats were exposed on postnatal day 7 to (1) 4 hours of hypoxia with 8% oxygen (preconditioning hypoxic group) or (2) 4 hours of normoxia (sham-preconditioning group). At 24 hours after the conditioning, rats from both groups were exposed to left carotid artery ligation followed by 2 hours of hypoxia (8% oxygen/92% nitrogen). All the rats were killed 1 week after hypoxia-ischemia, and their brains were extracted for histologic study. RESULTS Two types of brain damage were histologically observed at 1 week after hypoxia-ischemia in both groups: (1) generalized infarction in the ligated hemisphere and (2) predominant neuronal loss in the hippocampal region. The total incidence of brain damage was significantly decreased in the preconditioning hypoxic group (10/24 rats, 41.7%) compared with the sham-preconditioning hypoxic group (17/22 rats, 77.3%; P < .05). CONCLUSION Our results show that the hypoxic-ischemic tolerance phenomenon may be induced in the hypoxic-ischemic brain damage model by conditioning the animal before the insult with 4 hours of hypoxic exposure.
Collapse
Affiliation(s)
- A Ota
- Department of Obstetrics and Gynecology, Miyazaki Medical College, Kiyotake, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Tanaka H, Oki J, Takahashi S, Miyamoto A, Cho K, Okuno A. Effects of neonatal hypoxia on the medulla-spinal cord descending neurons. Pediatr Neurol 1998; 19:204-10. [PMID: 9806138 DOI: 10.1016/s0887-8994(98)00054-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Hypoxic changes in the medulla-spinal cord descending neurons were studied morphologically using a retrograde neurotracer, choleratoxin B subunit (CTb). On postnatal day 7, Sprague-Dawley rats were subjected to a hypoxic load of 8% oxygen for 5 hours. In the rats that survived, CTb was injected into the lumbar enlargement at postnatal day 26, and they were killed at postnatal day 28 for histologic analysis. Retrograde transported CTb was visualized by immunohistochemistry. The results were compared with those obtained from control rats. In the control rats, CTb-positive cells were observed in the nucleus reticularis gigantocellularis, nucleus reticularis magnocellularis, nucleus raphe magnus, nucleus raphe obscurus, and nucleus raphe pallidus. In the hypoxic rats, although CTb-positive cells were detected in the same areas as the control rats, there was a noteworthy decrease in the number of CTb-positive cells in all areas, and there were many cells with hypoxic degeneration. In all of the nuclei a marked decrease in the number of CTb-positive cells was observed. Because medulla-spinal cord descending neurons have important roles in the regulation of postural muscle tone, these results may account for the pathophysiology of abnormal muscle tonus accompanying hypoxic brain damage.
Collapse
Affiliation(s)
- H Tanaka
- Department of Pediatrics, Asahikawa Habilitation Center for Disabled Children, Hokkaido, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Cheng Y, Deshmukh M, D'Costa A, Demaro JA, Gidday JM, Shah A, Sun Y, Jacquin MF, Johnson EM, Holtzman DM. Caspase inhibitor affords neuroprotection with delayed administration in a rat model of neonatal hypoxic-ischemic brain injury. J Clin Invest 1998; 101:1992-9. [PMID: 9576764 PMCID: PMC508786 DOI: 10.1172/jci2169] [Citation(s) in RCA: 416] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Programmed cell death (apoptosis) is a normal process in the developing nervous system. Recent data suggest that certain features seen in the process of programmed cell death may be favored in the developing versus the adult brain in response to different brain injuries. In a well characterized model of neonatal hypoxia-ischemia, we demonstrate marked but delayed cell death in which there is prominent DNA laddering, TUNEL-labeling, and nuclei with condensed chromatin. Caspase activation, which is required in many cases of apoptotic cell death, also followed a delayed time course after hypoxia-ischemia. Administration of boc-aspartyl(OMe)-fluoromethylketone, a pan-caspase inhibitor, was significantly neuroprotective when given by intracerebroventricular injection 3 h after cerebral hypoxia-ischemia. In addition, systemic injections of boc-aspartyl(OMe)-fluoromethylketone also given in a delayed fashion, resulted in significant neuroprotection. These findings suggest that caspase inhibitors may be able to provide benefit over a prolonged therapeutic window after hypoxic-ischemic events in the developing brain, a major contributor to static encephalopathy and cerebral palsy.
Collapse
Affiliation(s)
- Y Cheng
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
McDonald JW, Shapiro SM, Silverstein FS, Johnston MV. Role of glutamate receptor-mediated excitotoxicity in bilirubin-induced brain injury in the Gunn rat model. Exp Neurol 1998; 150:21-9. [PMID: 9514835 DOI: 10.1006/exnr.1997.6762] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Severe hyperbilirubinemia in neonates with prematurity and/or systemic illnesses such as hemolytic disease, acidosis, and hypoxemia enhances their risk for developing cerebral palsy, paralysis of ocular upgaze, and deafness. This neurologic syndrome has been associated with selective neuronal vulnerability in the basal ganglia, certain brainstem nuclei, and Purkinje cells. However, the mechanism by which bilirubin damages neurons remains unclear. In these studies, we found that intracerebral injection of N-methyl-D-aspartate (NMDA), an excitotoxic analogue of glutamate, caused greater injury in jaundiced 7-day-old Gunn (jj) rat pups than in nonjaundiced heterozygous (Nj) littermate controls. NMDA injection caused even greater injury when protein-bound bilirubin was displaced with the sulfonamide drug sulfadimethoxine in jaundiced homozygous pups. In additional experiments, the acute signs of bilirubin-mediated neuronal injury, induced in homozygous (jj) Gunn rats by treatment with sulfonamide, were reduced by concurrent treatment with the NMDA-type glutamate channel antagonist (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohept-5,10-imine (MK-801, dizocilpine). The results suggest that bilirubin may cause encephalopathy and neuronal injury, at least in part, through an NMDA receptor-mediated excitotoxic mechanism. This conclusion is consistent with clinical observations that bilirubin encephalopathy is synergistically worsened by hypoxemia, which also shares an excitotoxic mechanism of neuronal injury.
Collapse
Affiliation(s)
- J W McDonald
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
39
|
Abstract
Bcl-xL is a death-inhibiting member of the Bcl-2/Ced9 family of proteins which either promote or inhibit apoptosis. Gene targeting has revealed that Bcl-xL is required for neuronal survival during brain development; however, Bcl-xL knock-out mice do not survive past embryonic day 13.5, precluding an analysis of Bcl-xL function at later stages of development. Bcl-xL expression is maintained at a high level postnatally in the CNS, suggesting that it may also regulate neuron survival in the postnatal period. To explore functions of Bcl-xL related to neuron survival in postnatal life, we generated transgenic mice overexpressing human Bcl-xL under the control of a pan-neuronal promoter. A line that showed strong overexpression in brainstem and a line that showed overexpression in hippocampus and cortex were chosen for analysis. We asked whether overexpression of Bcl-xL influences neuronal survival in the postnatal period by studying two injury paradigms that result in massive neuronal apoptosis. In the standard neonatal facial axotomy paradigm, Bcl-xL overexpression had substantial effects, with survival of 65% of the motor neurons 7 d after axotomy, as opposed to only 15% in nontransgenic littermates. To investigate whether Bcl-xL regulates survival of CNS neurons in the forebrain, we used a hypoxia-ischemia paradigm in neonatal mice. We show here that hypoxia-ischemia leads to substantial apoptosis in the hippocampus and cortex of wild-type neonatal mice. Furthermore, we show that overexpression of Bcl-xL is neuroprotective in this paradigm. We conclude that levels of Bcl-xL in postnatal neurons may be a critical determinant of their susceptibility to apoptosis.
Collapse
|
40
|
Flagmeyer I, Haas HL, Stevens DR. Adenosine A1 receptor-mediated depression of corticostriatal and thalamostriatal glutamatergic synaptic potentials in vitro. Brain Res 1997; 778:178-85. [PMID: 9462890 DOI: 10.1016/s0006-8993(97)01060-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Electrophysiological recordings in rat brain slices have been used to study the actions of adenosine on striatal neurons and striatal excitatory amino acid neurotransmission originating in the cortex or the thalamus. Adenosine had no effects on membrane properties of striatal neurons. Adenosine and the A1 agonist N6-Cyclopentyl adenosine reduced EPSPs of both cortical and thalamic origin by more than 50%. Depression of EPSPs was associated with an increase in paired-pulse facilitation, suggesting a presynaptic locus of action. EPSP depression was blocked by the A1 antagonist, 8-Cyclopentyl-1,3-dipropyl xanthine. The A2 agonist 5'-(N-cyclopropyl)-carboxamidoadenosine had no effect on excitatory amino acid neurotransmission. The A1 antagonist alone enhanced the synaptic component of the evoked field potential (23 +/- 12%). These results indicate that endogenous adenosine, acting via A1 receptors, limits striatal glutamatergic neurotransmission, serving a modulatory and neuroprotective role.
Collapse
Affiliation(s)
- I Flagmeyer
- Physiologisches Institut II, Heinrich-Heine Universität, Düsseldorf, Germany
| | | | | |
Collapse
|
41
|
Ota A, Ikeda T, Ikenoue T, Toshimori K. Sequence of neuronal responses assessed by immunohistochemistry in the newborn rat brain after hypoxia-ischemia. Am J Obstet Gynecol 1997; 177:519-26. [PMID: 9322617 DOI: 10.1016/s0002-9378(97)70139-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Our purpose was to study the neuronal responses of heat shock protein-72 (a stress-inducible protein) and microtubule-associated protein-2 (a constitutive protein of the neuronal cytoskeleton) after hypoxia-ischemia and their relationship with permanent damage in the newborn rat brain. STUDY DESIGN Seven-day-old rats were exposed to unilateral carotid artery ligation followed by 2 hours of hypoxia (8% oxygen/92% nitrogen) and then killed at time points ranging from 1 to 72 hours after injury. Brains were removed for immunohistochemical and routine staining. RESULTS Heat shock protein-72 appearance and microtubule-associated protein-2 disappearance occurred from 1 hour after injury, mainly in the dentate gyrus of the hippocampal formation and the cerebral cortex. Such alterations reached maximal levels at 24 hours for both proteins. Microtubule-associated protein-2 staining recovered in almost all parts of the brain. However, the hippocampal CA3 showed a delay in the responses for both proteins, and microtubule-associated protein-2 did not recover the response to immunostaining. Histologic evaluation at 72 hours after hypoxia by routine methods showed predominant damage in the hippocampal CA3. CONCLUSION Our results show that delayed responses of heat shock protein-72 and microtubule-associated protein-2 are related to a high incidence of neuronal cell loss in the hippocampal CA3 region.
Collapse
Affiliation(s)
- A Ota
- Department of Obstetrics and Gynecology, Miyazaki Medical College, Japan
| | | | | | | |
Collapse
|
42
|
Martin LJ, Brambrink AM, Lehmann C, Portera-Cailliau C, Koehler R, Rothstein J, Traystman RJ. Hypoxia-ischemia causes abnormalities in glutamate transporters and death of astroglia and neurons in newborn striatum. Ann Neurol 1997; 42:335-48. [PMID: 9307255 DOI: 10.1002/ana.410420310] [Citation(s) in RCA: 228] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The neonatal striatum degenerates after hypoxia-ischemia (H-I). We tested the hypothesis that damage to astrocytes and loss of glutamate transporters accompany striatal neurodegeneration after H-I. Newborn piglets were subjected to 30 minutes of hypoxia (arterial O2 saturation, 30%) and then 7 minutes of airway occlusion (O2 saturation, 5%), producing cardiac arrest, followed by cardiopulmonary resuscitation. Piglets recovered for 24, 48, or 96 hours. At 24 hours, 66% of putaminal neurons were injured, without differing significantly thereafter, but neuronal densities were reduced progressively (21-44%). By DNA nick-end labeling, the number of dying putaminal cells per square millimeter was increased maximally at 24 to 48 hours. Glial fibrillary acidic protein-positive cell body densities were reduced 48 to 55% at 24 to 48 hours but then recovered by 96 hours. Early postischemia, subsets of astrocytes had fragmented DNA; later postischemia, subsets of astrocytes proliferated. By immunocytochemistry, glutamate transporter 1 (GLT1) was lost after ischemia in the astroglial compartment but gained in cells appearing as neurons, whereas neuronal excitatory amino acid carrier 1 (EAAC1) dissipated. By immunoblotting, GLT1 and EAAC1 levels were 85% and 45% of control, respectively, at 24 hours of recovery. Thus, astroglial and neuronal injury occurs rapidly in H-I newborn striatum, with early gliodegeneration and glutamate transporter abnormalities possibly contributing to neurodegeneration.
Collapse
Affiliation(s)
- L J Martin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2196, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Cheng Y, Gidday JM, Yan Q, Shah AR, Holtzman DM. Marked age-dependent neuroprotection by brain-derived neurotrophic factor against neonatal hypoxic-ischemic brain injury. Ann Neurol 1997; 41:521-9. [PMID: 9124810 DOI: 10.1002/ana.410410416] [Citation(s) in RCA: 150] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Hypoxic-ischemic brain injury in survivors of perinatal asphyxia is a frequently encountered clinical problem for which there is currently no effective therapy. Neurotrophins, such as brain-derived neurotrophic factor (BDNF), can protect responsive neurons against cell death in some injury paradigms. While the role of BDNF in hypoxic-ischemic brain injury is not clear, evidence suggests that BDNF may have different effects in the developing, as opposed to the adult, brain. We found that a single intracerebroventricular (ICV) injection of BDNF resulted in rapid and robust phosphorylation of trk receptors in multiple brain regions in the postnatal day (PD) 7 rat brain. BDNF also markedly protected against hypoxic-ischemic brain injury at PD7. It protected against 90% of tissue loss due to hypoxic-ischemia when given just prior to the insult and against 50% of tissue loss when give after the insult. In contrast, ICV injection of BDNF in PD21 and adult rats resulted in little trk phosphorylation and less dramatic protection against unilateral hypoxic-ischemic injury at PD21. Because of its potent neuroprotective actions in the developing brain, BDNF may be a potential treatment for asphyxia and other forms of acute injury in the perinatal period.
Collapse
Affiliation(s)
- Y Cheng
- Department of Neurology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
44
|
Roohey T, Raju TN, Moustogiannis AN. Animal models for the study of perinatal hypoxic-ischemic encephalopathy: a critical analysis. Early Hum Dev 1997; 47:115-46. [PMID: 9039963 DOI: 10.1016/s0378-3782(96)01773-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We critically evaluated various design features from 292 animal studies related to perinatal hypoxic-ischemic encephalopathy (HIE). Rodents were the most frequently used animals in HIE research (26%), followed by piglets (23%) and sheep (22%). Asphyxia with or without ischemia was the most predominant method of producing experimental brain damage, but there were significant variations in specific details, particularly regarding the method and duration of brain insult. In 71% (207/292) of studies the CNS outcomes were tested within 24 h of experimental insult and in 29% (85/292) they were tested 24 h or more after the insult. Acute CNS metabolic end-points were assessed in 82-100% of all studies. In 90% of studies the chronological age of the animal was equivalent to that of human term newborn infant. However, in only 23% (67/292) were clinical neurological, developmental or behavioral outcomes evaluated, and in only 26% (76/292) was neuropathology assessed. While no single animal model was found to be ideal for all HIE research, some models were distinctly superior to others, depending upon the specific research question. The fetal sheep, newborn lamb and piglet models are well suited for the study of acute and subacute metabolic and physiologic endpoints, whereas the rodent and primate models could be used for long-term neurological and behavioral outcome experiments as well. We also feel that standardizing the study design features, including an HI insult method that produces consistent and predictable brain damage is urgently needed. Studies in neuro-ethology should explore how well brains of various animals compare with that of the newborn human infant. There is also a need for developing animal models that mimic clinical entities in which long-term neuro-developmental and behavioral outcomes can be assessed.
Collapse
Affiliation(s)
- T Roohey
- Department of Pediatrics, University of Illinois, Chicago 60612, USA
| | | | | |
Collapse
|
45
|
|
46
|
Abstract
Recent developments in understanding the pathophysiology of disordered motor control in cerebral palsy are reviewed. In spastic cerebral palsy, evidence for abnormal segmental as well as supraspinal control of motor neuron output exists. Impaired Ia inhibition of antagonist muscles has been suggested but recently contested. Evidence also supports the role of decreased presynaptic inhibition of Ia afferents and decreased nonreciprocal Ib inhibition. Furthermore, early cerebral injury results in reorganization of supraspinal (corticospinal) inputs to motor neuron pools. In extrapyramidal cerebral palsy, injury of basal ganglia or thalamus has been demonstrated. A scheme for understanding the neurochemical circuitry of the extrapyramidal system is discussed. Animal models and certain specific human diseases provide examples of how this circuitry may be disturbed, thereby resulting in an imbalance between the direct and indirect striatal output systems and in impaired motor control. Future studies employing postmortem neurochemical analysis, functional magnetic resonance imaging, and positron emission tomographic scanning may foster progress in this area.
Collapse
Affiliation(s)
- F M Filloux
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, USA
| |
Collapse
|
47
|
Abstract
Movement disorders are a well-recognized feature of some patients with cerebral palsy and often require treatment. However, treatments have been symptomatic and empiric, and there have been few pharmacologic studies. The major movement disorders in cerebral palsy are dystonia and the hyperkinesias choreoathetosis and myoclonus. They may occur in combination, often accompanied by spasticity and sometimes by epilepsy. Some drugs are useful treatments for all of these problems, but others may improve one while worsening another. Pitfalls in management include not diagnosing metabolic/degenerative disorders, which may mimic cerebral palsy, or not recognizing reversible complications of cerebral palsy, which may exacerbate symptoms. This review attempts to summarize empiric drug use and recommendations for therapy, drug studies in extrapyramidal cerebral palsy, and prospects for new drugs or models for the problem. Many new pharmacologic agents are available for study in cerebral palsy. Better methods of detecting basal ganglia injury after perinatal injury in asymptomatic infants may allow early intervention in the biologic process of recovery and adaptation.
Collapse
Affiliation(s)
- M R Pranzatelli
- Department of Pharmacology, George Washington University, Washington, DC, USA
| |
Collapse
|
48
|
Jansen EM, Low WC. Long-term effects of neonatal ischemic-hypoxic brain injury on sensorimotor and locomotor tasks in rats. Behav Brain Res 1996; 78:189-94. [PMID: 8864051 DOI: 10.1016/0166-4328(95)00248-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Perinatal ischemia and/or hypoxia in humans are major risk factors for neurologic injury that often manifest as sensorimotor and locomotor deficits throughout development and into maturity. In these studies, we utilized an established model of neonatal ischemic-hypoxia that creates unilateral striatal, cortical, and hippocampal damage (Rice III, J.E., Vanucci, R.C. and Brierley, J.B., Ann. Neurol., 9 (1981) 131-141) to investigate sensorimotor and locomotor deficits in these animals during development and as adults. Sensorimotor deficits were examined by measuring the amount of time that the animals were able to remain on a rotating treadmill. Locomotor abnormalities were assessed by measuring apomorphine-induced rotational asymmetry. Following the neonatal ischemic-hypoxic episode, at 3-9 weeks of age, animals were not able to remain on the treadmill as long as their normal littermate controls. In addition, these animals demonstrated an abnormal, ipsiversive rotational asymmetry in response to systemic administration of apomorphine. When these animals reached adulthood, the degree of atrophy in specific regions of the damaged hemisphere was quantified using measurements of cross-sectional area. The mean cross-sectional area of the striatum was decreased by 29%, the sensorimotor cortex area by 26%, and the dorsal hippocampus cross-sectional area was approximately 6% of its normal size. These data suggest that this rodent model of neonatal ischemic-hypoxic brain injury results in cerebral atrophy and long-lasting sensorimotor and locomotor deficits. These particular behavioral tasks may be used in future studies to assess locomotor and sensorimotor deficits following neonatal ischemic-hypoxic brain injury.
Collapse
Affiliation(s)
- E M Jansen
- Department of Neurosurgery, Graduate Program in Neuroscience, University of Minnesota Medical School, Minneapolis, USA
| | | |
Collapse
|
49
|
Nanda D, Tolputt J, Collard KJ. Changes in brain glutathione levels during postnatal development in the rat. ACTA ACUST UNITED AC 1996. [DOI: 10.1016/s0165-3806(96)80016-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
50
|
Filloux FM, Adair J, Narang N. The temporal evolution of striatal dopamine receptor binding and mRNA expression following hypoxia-ischemia in the neonatal rat. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1996; 94:81-91. [PMID: 8816280 DOI: 10.1016/0165-3806(96)00053-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neonatal hypoxic-ischemic (HI) brain injury in the rat alters dopamine receptors. To determine whether such changes are permanent, dopamine receptors and corresponding mRNA were examined at various time points after neonatal HI using receptor autoradiography and in situ hybridization. Rat pups underwent ligation of the left common carotid artery followed by hypoxic exposure (8.5% O2 for 3 h). Controls underwent sham surgery alone. Animals surviving for 2-80 days following HI were studied. Striatal D1 receptors (labeled by [3H]SCH23390) were reduced as early as 2 days following HI, remained depressed for 21 days, but recovered to control levels by young adulthood (3 months of age). D2 receptors (labeled by [125I] iodosulpride) did not decline until 10 days after HI, and remained uniformly depressed throughout the caudate-putamen thereafter. Changes in D1 receptor mRNA transcripts closely paralleled alterations in receptors: early reductions in D1 mRNA signal recovered by young adulthood. D2 mRNA exhibited a unique temporal profile with an early decrease (2 days following HI), and prompt, persistent recovery. Dopamine receptors and transcripts are differentially affected by HI injury early in development. Whereas D1 receptor expression recovers from neonatal HI injury, D2 receptors remain permanently affected despite the presence of normal levels of D2 receptor transcripts. A persistent, post-transcriptional effect of HI on D2 receptor expression is suggested.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Autoradiography
- Base Sequence
- Benzazepines/pharmacology
- Brain Ischemia/genetics
- Brain Ischemia/physiopathology
- Dopamine Antagonists/pharmacology
- Dopamine D2 Receptor Antagonists
- Female
- Hypoxia, Brain/genetics
- Hypoxia, Brain/physiopathology
- In Situ Hybridization
- Iodine Radioisotopes
- Molecular Sequence Data
- Neostriatum/chemistry
- Neostriatum/physiology
- Neuronal Plasticity/physiology
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Dopamine D1/antagonists & inhibitors
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Sulpiride/analogs & derivatives
- Sulpiride/pharmacology
- Time Factors
- Tritium
Collapse
Affiliation(s)
- F M Filloux
- Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, USA.
| | | | | |
Collapse
|