Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Mol Sci ; 24(23)2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38069249

ABSTRACT

Seizures are common in preterm newborns and are associated with poor neurodevelopmental outcomes. Current anticonvulsants have poor efficacy, and many have been associated with upregulation of apoptosis in the developing brain. Apigenin, a natural bioactive flavonoid, is a potent inhibitor of hyaluronidase and reduces seizures in adult animal models. However, its impact on perinatal seizures is unclear. In the present study, we examined the effect of apigenin and S3, a synthetic, selective hyaluronidase inhibitor, on seizures after cerebral ischemia in preterm fetal sheep at 0.7 gestation (98-99 days, term ~147 days). Fetuses received sham ischemia (n = 9) or ischemia induced by bilateral carotid occlusion for 25 min. Immediately after ischemia, fetuses received either a continuous infusion of vehicle (0.036% dimethyl sulfoxide, n = 8) or apigenin (50 µM, n = 6). In a pilot study, we also tested infusion of S3 (2 µM, n = 3). Fetuses were monitored continuously for 72 h after ischemia. Infusion of apigenin or S3 were both associated with reduced numbers of animals with seizures, total seizure time, and mean seizure burden. S3 was also associated with a reduction in the total number of seizures over the 72 h recovery period. In animals that developed seizures, apigenin was associated with earlier cessation of seizures. However, apigenin or S3 treatment did not alter recovery of electroencephalographic power or spectral edge frequency. These data support that targeting brain hyaluronidase activity with apigenin or S3 may be an effective strategy to reduce perinatal seizures following ischemia. Further studies are required to determine their effects on neurohistological outcomes.


Subject(s)
Apigenin , Hypoxia-Ischemia, Brain , Pregnancy , Female , Sheep , Animals , Apigenin/pharmacology , Apigenin/therapeutic use , Hyaluronoglucosaminidase , Pilot Projects , Seizures/drug therapy , Fetus/pathology , Ischemia , Electroencephalography , Hypoxia-Ischemia, Brain/pathology
2.
Children (Basel) ; 9(6)2022 Jun 12.
Article in English | MEDLINE | ID: mdl-35740812

ABSTRACT

Thoracic outlet syndrome (TOS) is a condition that results from the compression of neurovascular structures as they exit the thorax. Arterial ischemic stroke can occur in TOS due to retrograde embolism from the subclavian artery. We describe a 15-year-old girl who presented with left hemiplegia after 2 weeks of right arm numbness and tingling. Imaging showed an acute ischemic stroke due to a right middle cerebral artery occlusion. She was treated with intravenous tissue plasminogen activator at 1.3 h and mechanical thrombectomy at 2.4 h with successful recanalization. Review of her neck computed tomography angiogram suggested a right subclavian artery aneurysm, and upper-extremity imaging also demonstrated distal thrombosis and fusion of right first and second ribs, which was consistent with thoracic outlet syndrome. Three days later, she underwent a right subclavian artery aneurysm repair, right brachial and ulnar artery thrombectomy, and first rib resection. Three months later, she demonstrated good neurologic recovery. TOS is an uncommon cause of stroke in children, which may be heralded by upper-extremity symptoms. Interventionalists should be aware of the possibility of vascular anomalies in children; however, this finding does not exclude the possibility of acute stroke intervention.

3.
PLoS One ; 9(11): e112800, 2014.
Article in English | MEDLINE | ID: mdl-25390897

ABSTRACT

OBJECTIVE: Although the spectrum of white matter injury (WMI) in preterm infants is shifting from cystic necrotic lesions to milder forms, the factors that contribute to this changing spectrum are unclear. We hypothesized that recurrent hypoxia-ischemia (rHI) will exacerbate the spectrum of WMI defined by markers of inflammation and molecules related to the extracellular matrix (hyaluronan (HA) and the PH20 hyaluronidase) that regulate maturation of the oligodendrocyte (OL) lineage after WMI. METHODS: We employed a preterm fetal sheep model of in utero moderate hypoxemia and global severe but not complete cerebral ischemia that reproduces the spectrum of human WMI. The response to rHI was compared against corresponding early or later single episodes of HI. An ordinal rating scale of WMI was compared against an unbiased quantitative image analysis protocol that provided continuous histo-pathological outcome measures for astrogliosis and microglial activation. Late oligodendrocyte progenitors (preOLs) were quantified by stereology. Analysis of hyaluronan and the hyaluronidase PH20 defined the progressive response of the extracellular matrix to WMI. RESULTS: rHI resulted in a more severe spectrum of WMI with a greater burden of necrosis, but an expanded population of preOLs that displayed reduced susceptibility to cell death. WMI from single episodes of HI or rHI was accompanied by elevated HA levels and increased labeling for PH20. Expression of PH20 in fetal ovine WMI was confirmed by RT-PCR and RNA-sequencing. CONCLUSIONS: rHI is associated with an increased risk for more severe WMI with necrosis, but reduced risk for preOL degeneration compared to single episodes of HI. Expansion of the preOL pool may be linked to elevated hyaluronan and PH20.


Subject(s)
Hypoxia-Ischemia, Brain/pathology , White Matter/injuries , White Matter/pathology , Animals , Animals, Newborn , Cell Adhesion Molecules/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fetus/metabolism , Fetus/pathology , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/metabolism , Hypoxia-Ischemia, Brain/metabolism , Necrosis/metabolism , Necrosis/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Sheep
4.
Ann Neurol ; 75(4): 508-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24395459

ABSTRACT

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


Subject(s)
Brain Ischemia/pathology , Caudate Nucleus/pathology , Fetal Hypoxia/pathology , Gene Expression Regulation, Developmental/physiology , Neurons/pathology , Premature Birth/physiopathology , Action Potentials/drug effects , Animals , Brain Ischemia/blood , Caspase 3/metabolism , Dendrites/pathology , Dendrites/ultrastructure , Disease Models, Animal , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Female , GABA Agents/pharmacology , Goats , Ki-67 Antigen/metabolism , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Pregnancy , Time Factors
5.
Pediatr Res ; 75(1-2): 227-33, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24336432

ABSTRACT

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


Subject(s)
Brain/physiopathology , Child Development , Cognition , Developmental Disabilities/etiology , Infant, Premature , Age Factors , Animals , Brain/embryology , Brain/growth & development , Brain/pathology , Developmental Disabilities/diagnosis , Developmental Disabilities/physiopathology , Developmental Disabilities/psychology , Gestational Age , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Neural Pathways/physiopathology , Neuronal Plasticity , Risk Factors , Time Factors
6.
Sci Transl Med ; 5(168): 168ra7, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23325800

ABSTRACT

Children who survive preterm birth exhibit persistent unexplained disturbances in cerebral cortical growth with associated cognitive and learning disabilities. The mechanisms underlying these deficits remain elusive. We used ex vivo diffusion magnetic resonance imaging to demonstrate in a preterm large-animal model that cerebral ischemia impairs cortical growth and the normal maturational decline in cortical fractional anisotropy (FA). Analysis of pyramidal neurons revealed that cortical deficits were associated with impaired expansion of the dendritic arbor and reduced synaptic density. Together, these findings suggest a link between abnormal cortical FA and disturbances of neuronal morphological development. To experimentally investigate this possibility, we measured the orientation distribution of dendritic branches and observed that it corresponds with the theoretically predicted pattern of increased anisotropy within cases that exhibited elevated cortical FA after ischemia. We conclude that cortical growth impairments are associated with diffuse disturbances in the dendritic arbor and synapse formation of cortical neurons, which may underlie the cognitive and learning disabilities in survivors of preterm birth. Further, measurement of cortical FA may be useful for noninvasively detecting neurological disorders affecting cortical development.


Subject(s)
Brain Ischemia/pathology , Cerebral Cortex/pathology , Dendrites/pathology , Diffusion Magnetic Resonance Imaging , Premature Birth/pathology , Animals , Anisotropy , Cerebral Cortex/growth & development , Dendritic Spines/pathology , Disease Models, Animal , Pyramidal Cells/pathology , Sheep
7.
PLoS One ; 8(12): e82940, 2013.
Article in English | MEDLINE | ID: mdl-24416093

ABSTRACT

BACKGROUND AND PURPOSE: Although the spectrum of perinatal white matter injury (WMI) in preterm infants is shifting from cystic encephalomalacia to milder forms of WMI, the factors that contribute to this changing spectrum are unclear. We hypothesized that the variability in WMI quantified by immunohistochemical markers of inflammation could be correlated with the severity of impaired blood oxygen, glucose and lactate. METHODS: We employed a preterm fetal sheep model of in utero moderate hypoxemia and global severe but not complete cerebral ischemia that reproduces the spectrum of human WMI. Since there is small but measurable residual brain blood flow during occlusion, we sought to determine if the metabolic state of the residual arterial blood was associated with severity of WMI. Near the conclusion of hypoxia-ischemia, we recorded cephalic arterial blood pressure, blood oxygen, glucose and lactate levels. To define the spectrum of WMI, an ordinal WMI rating scale was compared against an unbiased quantitative image analysis protocol that provided continuous histo-pathological outcome measures for astrogliosis and microgliosis derived from the entire white matter. RESULTS: A spectrum of WMI was observed that ranged from diffuse non-necrotic lesions to more severe injury that comprised discrete foci of microscopic or macroscopic necrosis. Residual arterial pressure, oxygen content and blood glucose displayed a significant inverse association with WMI and lactate concentrations were directly related. Elevated glucose levels were the most significantly associated with less severe WMI. CONCLUSIONS: Our results suggest that under conditions of hypoxemia and severe cephalic hypotension, WMI severity measured using unbiased immunohistochemical measurements correlated with several physiologic parameters, including glucose, which may be a useful marker of fetal response to hypoxia or provide protection against energy failure and more severe WMI.


Subject(s)
Brain Ischemia/physiopathology , Fetal Hypoxia/physiopathology , Myelin Sheath/pathology , Analysis of Variance , Animals , Blood Chemical Analysis , Blood Glucose , Blood Pressure , Brain Ischemia/metabolism , Fetal Hypoxia/metabolism , Hemodynamics , Immunohistochemistry , Lactic Acid/blood , Myelin Sheath/metabolism , Necrosis , Oxygen/blood , Premature Birth , Sheep
8.
Sci Transl Med ; 4(155): 155ra136, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23052293

ABSTRACT

Shiverer-immunodeficient (Shi-id) mice demonstrate defective myelination in the central nervous system (CNS) and significant ataxia by 2 to 3 weeks of life. Expanded, banked human neural stem cells (HuCNS-SCs) were transplanted into three sites in the brains of neonatal or juvenile Shi-id mice, which were asymptomatic or showed advanced hypomyelination, respectively. In both groups of mice, HuCNS-SCs engrafted and underwent preferential differentiation into oligodendrocytes. These oligodendrocytes generated compact myelin with normalized nodal organization, ultrastructure, and axon conduction velocities. Myelination was equivalent in neonatal and juvenile mice by quantitative histopathology and high-field ex vivo magnetic resonance imaging, which, through fractional anisotropy, revealed CNS myelination 5 to 7 weeks after HuCNS-SC transplantation. Transplanted HuCNS-SCs generated functional myelin in the CNS, even in animals with severe symptomatic hypomyelination, suggesting that this strategy may be useful for treating dysmyelinating diseases.


Subject(s)
Demyelinating Diseases/therapy , Myelin Sheath/metabolism , Neural Stem Cells/cytology , Animals , Brain/cytology , Brain/metabolism , Brain/pathology , Central Nervous System/cytology , Central Nervous System/metabolism , Central Nervous System/pathology , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Neural Stem Cells/physiology , Stem Cell Transplantation
9.
Ann Neurol ; 72(4): 525-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23109147

ABSTRACT

OBJECTIVE: Previously we reported that exposure of 6-day-old (P6) rhesus macaques to isoflurane for 5 hours triggers a robust neuroapoptosis response in developing brain. We have also observed (unpublished data) that isoflurane causes apoptosis of cellular profiles in the white matter that resemble glia. We analyzed the cellular identity of the apoptotic white matter profiles and determined the magnitude of this cell death response to isoflurane. METHODS: Neonatal (P6) rhesus macaques were exposed for 5 hours to isoflurane anesthesia according to current clinical standards in pediatric anesthesia. Brains were collected 3 hours later and examined immunohistochemically to analyze apoptotic neuronal and glial death. RESULTS: Brains exposed to isoflurane displayed significant apoptosis in both the white and gray matter throughout the central nervous system. Approximately 52% of the dying cells were glia, and 48% were neurons. Oligodendrocytes (OLs) engaged in myelinogenesis were selectively vulnerable, in contrast to OL progenitors, astrocytes, microglia, and interstitial neurons. When adjusted for control rates of OL apoptosis, the percentage of OLs that degenerated in the forebrain white matter of the isoflurane-treated group was 6.3% of the total population of myelinating OLs. INTERPRETATION: Exposure of the infant rhesus macaque brain to isoflurane for 5 hours is sufficient to cause widespread apoptosis of neurons and OLs throughout the developing brain. Deletion of OLs at a stage when they are just beginning to myelinate axons could potentially have adverse long-term neurobehavioral consequences that might be additive to the potential consequences of isoflurane-induced neuroapoptosis.


Subject(s)
Anesthetics, Inhalation/toxicity , Apoptosis/drug effects , Brain/pathology , Isoflurane/toxicity , Oligodendroglia/drug effects , Animals , Animals, Newborn , Axons/drug effects , Axons/physiology , Caspases/physiology , Cell Death/physiology , Immunohistochemistry , Macaca mulatta , Myelin Sheath/physiology , Nerve Regeneration/physiology , Tissue Fixation
10.
Neurotherapeutics ; 9(2): 359-70, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22399133

ABSTRACT

Despite advances in neonatal intensive care, survivors of premature birth remain highly susceptible to unique patterns of developmental brain injury that manifest as cerebral palsy and cognitive-learning disabilities. The developing brain is particularly susceptible to cerebral white matter injury related to hypoxia-ischemia. Cerebral white matter development in fetal sheep shares many anatomical and physiological similarities with humans. Thus, the fetal sheep has provided unique experimental access to the complex pathophysiological processes that contribute to injury to the human brain during successive periods in development. Recent refinements have resulted in models that replicate major features of acute and chronic human cerebral injury and have provided access to complex clinically relevant studies of cerebral blood flow and neuroimaging that are not feasible in smaller laboratory animals. Here, we focus on emerging insights and methodologies from studies in fetal sheep that have begun to define cellular and vascular factors that contribute to white matter injury. Recent advances include spatially defined measurements of cerebral blood flow in utero, the definition of cellular maturational factors that define the topography of injury and the application of high-field magnetic resonance imaging to define novel neuroimaging signatures for specific types of chronic white matter injury. Despite the higher costs and technical challenges of instrumented preterm fetal sheep models, they provide powerful access to clinically relevant studies that provide a more integrated analysis of the spectrum of insults that appear to contribute to cerebral injury in human preterm infants.


Subject(s)
Brain Injuries/pathology , Cerebrum/pathology , Disease Models, Animal , Nerve Fibers, Myelinated/pathology , Sheep , Animals , Brain Injuries/physiopathology , Brain Injuries/therapy , Cerebrovascular Circulation/physiology , Cerebrum/blood supply , Female , Fetus , Humans , Infant, Newborn , Infant, Premature , Pregnancy
11.
Ann Neurol ; 71(1): 93-109, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22275256

ABSTRACT

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


Subject(s)
Cell Proliferation , Infant, Premature, Diseases/pathology , Myelin Sheath/pathology , Oligodendroglia/pathology , Stem Cells/pathology , Astrocytes/pathology , Cell Differentiation/physiology , Cohort Studies , Female , Humans , Infant, Newborn , Infant, Premature , Leukoencephalopathies/pathology , Male , Necrosis , Nerve Fibers, Myelinated/pathology , Prospective Studies , Retrospective Studies
12.
Ann Neurol ; 72(6): 936-51, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23280842

ABSTRACT

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


Subject(s)
Axons/metabolism , Hypoxia-Ischemia, Brain/pathology , Myelin Sheath/metabolism , Nerve Fibers, Myelinated/pathology , Optic Nerve/pathology , Age Factors , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Astrocytes/drug effects , Astrocytes/pathology , Axons/drug effects , Axons/ultrastructure , Disease Models, Animal , Dizocilpine Maleate/therapeutic use , Glucose/deficiency , Green Fluorescent Proteins/genetics , Hypoxia/pathology , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/drug therapy , Mice , Mice, Transgenic , Nerve Degeneration/etiology , Nerve Degeneration/prevention & control , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/ultrastructure , Neuroprotective Agents/therapeutic use , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglia/ultrastructure , Optic Nerve/growth & development , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Thy-1 Antigens/genetics , omega-Agatoxin IVA/therapeutic use
13.
Stroke ; 43(1): 178-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22076007

ABSTRACT

BACKGROUND AND PURPOSE: White matter injury (WMI) is the leading cause of brain injury in preterm survivors and results in myelination failure. Although axonal degeneration occurs in necrotic lesions, the role of axonopathy in myelination failure remains controversial for diffuse non-necrotic WMI, which is currently the major form of WMI. We determined the burden of axonopathy in diffuse lesions. METHODS: We analyzed WMI in a preterm fetal sheep model of global cerebral ischemia that replicates the relative burden of necrotic and non-necrotic human WMI. WMI was analyzed at 1 or 2 weeks after ischemia and identified by ex vivo high-field (11.7 Tesla) magnetic resonance imaging of fixed brain tissue. Axonal integrity was analyzed by immunohistochemical detection of axon injury markers and by transmission electron microscopy to quantify axon loss and degeneration in magnetic resonance imaging-defined lesions. RESULTS: Axonal degeneration, defined by staining for neurofilament protein and ß-amyloid precursor protein, was restricted to discrete necrotic foci with robust microglial activation. Unexpectedly, axonal degeneration was not visualized in the major form of WMI, which comprised large non-necrotic lesions with diffuse reactive astrogliosis. In these major lesions, quantitative electron microscopy studies confirmed no significant differences in the density of intact and degenerating axons or in the distribution of axon diameters relative to controls. CONCLUSIONS: The mechanism of myelination failure differs significantly in perinatal WMI dependent on the burden of necrosis. Axonopathy is associated with focal necrotic injury but not with primary diffuse non-necrotic lesions, which supports that intact axons in the primary lesions are potential targets for myelination.


Subject(s)
Axons/pathology , Brain/pathology , Hypoxia-Ischemia, Brain/pathology , Myelin Sheath/pathology , Nerve Fibers, Myelinated/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Axons/metabolism , Brain/metabolism , Female , Hypoxia-Ischemia, Brain/metabolism , Myelin Sheath/metabolism , Necrosis/metabolism , Necrosis/pathology , Nerve Fibers, Myelinated/metabolism , Neurofilament Proteins/metabolism , Pregnancy , Sheep
14.
Mol Neurodegener ; 6: 46, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21729326

ABSTRACT

BACKGROUND: CNS myelination disturbances commonly occur in chronic white matter lesions in neurodevelopmental and adult neurological disorders. Recent studies support that myelination failure can involve a disrupted cellular repair mechanism where oligodendrocyte (OL) progenitor cells (OPCs) proliferate in lesions with diffuse astrogliosis, but fail to fully differentiate to mature myelinating OLs. There are no in vitro models that reproduce these features of myelination failure. RESULTS: Forebrain coronal slices from postnatal day (P) 0.5/1 rat pups were cultured for 1, 5, or 9 days in vitro (DIV). Slices rapidly exhibited diffuse astrogliosis and accumulation of the extracellular matrix glycosaminoglycan hyaluronan (HA), an inhibitor of OPC differentiation and re-myelination. At 1 DIV ~1.5% of Olig2+ OLs displayed caspase-3 activation, which increased to ~11.5% by 9 DIV. At 1 DIV the density of PDGFRα+ and PDGFRα+/Ki67+ OPCs were significantly elevated compared to 0 DIV (P < 0.01). Despite this proliferative response, at 9 DIV ~60% of white matter OLs were late progenitors (preOLs), compared to ~7% in the postnatal day 10 rat (P < 0.0001), consistent with preOL maturation arrest. Addition of HA to slices significantly decreased the density of MBP+ OLs at 9 DIV compared to controls (217 ± 16 vs. 328 ± 17 cells/mm2, respectively; P = 0.0003), supporting an inhibitory role of HA in OL lineage progression in chronic lesions. CONCLUSIONS: Diffuse white matter astrogliosis and early OPC proliferation with impaired OL maturation were reproduced in this model of myelination failure. This system may be used to define mechanisms of OPC maturation arrest and myelination failure related to astrogliosis and HA accumulation.

15.
Ann Neurol ; 70(3): 493-507, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21796666

ABSTRACT

OBJECTIVE: Although magnetic resonance imaging (MRI) is the optimal imaging modality to define cerebral white-matter injury (WMI) in preterm survivors, the histopathological features of MRI-defined chronic lesions are poorly defined. We hypothesized that chronic WMI is related to a combination of delayed oligodendrocyte (OL) lineage cell death and arrested maturation of preoligodendrocytes (preOLs). We determined whether ex vivo MRI can distinguish distinct microglial and astroglial responses related to WMI progression and arrested preOL differentiation. METHODS: We employed a preterm fetal sheep model of global cerebral ischemia in which acute WMI results in selective preOL degeneration. We developed novel algorithms to register histopathologically-defined lesions with contrast-weighted and diffusion-weighted high-field ex vivo MRI data. RESULTS: Despite mild delayed preOL degeneration, preOL density recovered to control levels by 7 days after ischemia and was ~2 fold greater at 14 days. However, premyelinating OLs were significantly diminished at 7 and 14 days. WMI evolved to mostly gliotic lesions where arrested preOL differentiation was directly proportional to the magnitude of astrogliosis. A reduction in cerebral WM volume was accompanied by four classes of MRI-defined lesions. Each lesion type displayed unique astroglial and microglial responses that corresponded to distinct forms of necrotic or non-necrotic injury. High-field MRI defined 2 novel hypointense signal abnormalities on T(2) -weighted images that coincided with microscopic necrosis or identified astrogliosis with high sensitivity and specificity. INTERPRETATION: These studies support the potential of high-field MRI for early identification of microscopic necrosis and gliosis with preOL maturation arrest, a common form of WMI in preterm survivors.


Subject(s)
Animals, Newborn/physiology , Brain Damage, Chronic/pathology , Brain/pathology , Magnetic Resonance Imaging/methods , Algorithms , Animals , Brain/growth & development , Brain Ischemia/pathology , Calcium-Binding Proteins , Caspase 3/metabolism , Cell Proliferation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Disease Progression , Electromagnetic Fields , Enzyme Activation/physiology , Female , Fetus/pathology , Glial Fibrillary Acidic Protein/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Microfilament Proteins , Neural Stem Cells/pathology , Oligodendroglia/pathology , Pregnancy , Sheep , Tissue Fixation
16.
J Cereb Blood Flow Metab ; 30(5): 1053-65, 2010 May.
Article in English | MEDLINE | ID: mdl-20068573

ABSTRACT

Emerging evidence supports that premature infants are susceptible to both cerebral white and gray matter injury. In a fetal rabbit model of placental insufficiency, preterm rabbits at embryonic day 22 (E22) exhibited histologic evidence of gray matter injury but minimal white matter injury after global hypoxia-ischemia (H-I). We hypothesized that the dissociation between susceptibility to gray and white matter injury at E22 was related to the timing of appearance of late oligodendrocyte progenitors (preOLs) that are particularly vulnerable in preterm human white matter lesions. During normal rabbit oligodendrocyte (OL) lineage progression, early OL progenitors predominated at E22. PreOL density increased between E24 and E25 in major forebrain white matter tracts. After H-I at E22 and E25, we observed a similar magnitude of cerebral H-I, assessed by cortical microvascular blood flow, and gray matter injury, assessed by caspase activation. However, the increased preOL density at E25 was accompanied by a significant increase in acute white matter injury after H-I that coincided with enhanced preOL degeneration. At E29, significant white matter atrophy developed after H-I at E25 but not E22. Thus, the timing of appearance of preOLs coincided with onset of a developmental window of enhanced white but not gray matter susceptibility to H-I.


Subject(s)
Cerebral Cortex , Hypoxia-Ischemia, Brain , Nerve Fibers, Myelinated , Oligodendroglia/physiology , Stem Cells/physiology , Animals , Basal Ganglia/cytology , Basal Ganglia/embryology , Caspase 3/metabolism , Cell Lineage , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Female , Fetus/pathology , Fetus/physiopathology , Gestational Age , Humans , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/physiology , Oligodendroglia/cytology , Pregnancy , Rabbits , Stem Cells/cytology
17.
Ann Neurol ; 63(4): 520-30, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18393269

ABSTRACT

OBJECTIVE: Abnormal myelination is a major pathological sequela of chronic periventricular white matter injury in survivors of premature birth. We tested the hypothesis that myelination failure in chronic hypoxia-ischemia-induced periventricular white matter injury is related to persistent depletion of the oligodendrocyte (OL) precursor pool required to generate mature myelinating OLs. METHODS: A neonatal rat model of hypoxia-ischemia was used where acute degeneration of late OL progenitors (preOLs) occurs via a mostly caspase-independent mechanism. The fate of OL lineage cells in chronic cerebral lesions was defined with OL lineage-specific markers. RESULTS: Acute caspase-3-independent preOL degeneration from hypoxia-ischemia was significantly augmented by delayed preOL death that was caspase-3-dependent. Degeneration of preOLs was offset by a robust regenerative response that resulted in a several-fold expansion in the pool of surviving preOLs in chronic lesions. However, these preOLs displayed persistent maturation arrest with failure to differentiate and generate myelin. When preOL-rich chronic lesions sustained recurrent hypoxia-ischemia at a time in development when white matter is normally resistant to injury, an approximately 10-fold increase in caspase-dependent preOL degeneration occurred relative to lesions caused by a single episode of hypoxia-ischemia. INTERPRETATION: The mechanism of myelination failure in chronic white matter lesions is related to a combination of delayed preOL degeneration and preOL maturation arrest. The persistence of a susceptible population of preOLs renders chronic white matter lesions markedly more vulnerable to recurrent hypoxia-ischemia. These data suggest that preOL maturation arrest may predispose to more severe white matter injury in preterm survivors that sustain recurrent hypoxia-ischemia.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Nerve Fibers, Myelinated/pathology , Oligodendroglia/cytology , Oligodendroglia/pathology , Animals , Animals, Newborn , Cell Differentiation/physiology , Cell Lineage/physiology , Chronic Disease , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Nerve Fibers, Myelinated/physiology , Oligodendroglia/physiology , Rats , Rats, Sprague-Dawley , Stem Cells/pathology
18.
J Cereb Blood Flow Metab ; 28(5): 995-1008, 2008 May.
Article in English | MEDLINE | ID: mdl-18091757

ABSTRACT

Periventricular white matter (PVWM) injury is the leading cause of neurologic disability in survivors of prematurity. To address the role of ischemia in PVWM and cerebral cortical injury, we hypothesized that immaturity of spatially distal vascular 'end zones' or 'border zones' predisposes PVWM to greater decreases in cerebral blood flow (CBF) than more proximal structures. We quantified regional CBF with fluorescently labeled microspheres in 0.65 gestation fetal sheep in histopathologically defined three-dimensional regions by post hoc digital dissection and coregistration algorithms. Basal flow in PVWM was significantly lower than in gyral white matter and cortex, but was equivalent in superficial, middle, and deep PVWM. Absolute and relative CBF (expressed as percentage of basal) did not differ significantly during ischemia or reperfusion between PVWM, gyral white matter, or cortex. Moreover, CBF during ischemia-reperfusion was equivalent in three adjacent PVWM levels and was not consistent with the magnitude of severity of PVWM injury, defined by TUNEL (terminal deoxynucleotidyltransferase-mediated dUPT nick end labeling) staining. However, the magnitude of ischemia was predicted by the severity of discrete cortical lesions. Hence, unlike cerebral cortex, unique CBF disturbances did not account for the distribution of PVWM injury. Previously defined cellular maturational factors, thus, appear to have a greater influence on PVWM vulnerability to ischemic injury than the presence of immature vascular boundary zones.


Subject(s)
Cerebral Cortex/blood supply , Cerebrovascular Circulation/physiology , Corpus Callosum/blood supply , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Nerve Fibers, Myelinated/pathology , Animals , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Corpus Callosum/embryology , Corpus Callosum/pathology , Electroencephalography , Epilepsy/pathology , Epilepsy/physiopathology , Female , Gestational Age , In Situ Nick-End Labeling , Microspheres , Nerve Fibers, Myelinated/physiology , Oligodendroglia/pathology , Oligodendroglia/physiology , Predictive Value of Tests , Pregnancy , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Severity of Illness Index , Sheep
19.
J Neurochem ; 100(3): 628-38, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17263792

ABSTRACT

Multipotent neural stem cells (NSCs) are competent for commitment to the oligodendrocyte (OL) lineage both in vitro and in vivo. We exploited this property to develop a rat neurospheres (NS)/oligospheres (OS)-based culture system to generate large numbers of highly enriched late OL progenitors (preOLs) and mature OLs (MatOLs). CNS neuroblastoma cell line B104-derived conditioned medium promoted the generation of nearly pure populations of preOLs from dissociated OS. The subsequent culture of preOLs with ciliary neurotrophic factor (CNTF) and 3,3',5'-triiodo-L-thyronine (T(3)) generated nearly pure populations of MatOLs. OL lineage specificity was confirmed by immunocytochemistry, quantitative RT-PCR and gene expression profiling, which demonstrated large differences between preOLs and MatOLs. The insulin-like growth factors (IGFs) are potent neuro-protective agents required for OL survival. We used this system to systematically define maturation-dependent changes in IGF signaling during the course of OL differentiation. The IGF-I and insulin receptors, insulin receptor substrate-1 (IRS-1) and IRS-2, protein kinase B (PKB)/Akt and Janus kinase (JNK) were expressed at higher levels in NS and preOLs compared with OS and MatOLs. Erk expression increased markedly from NS to OS, decreased only partially upon commitment to preOLs, and, in MatOLs, returned to a low level similar to NS. IGF activation of the generally proliferative Erk pathway was gradually acquired during NSC differentiation, whereas IGF activation of the generally pro-survival, anti-apoptotic PI3K/PKB pathway was consistently robust at each developmental stage.


Subject(s)
Brain Tissue Transplantation/methods , Cell Differentiation/physiology , Multipotent Stem Cells/metabolism , Oligodendroglia/metabolism , Somatomedins/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , Cell Lineage/physiology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/pharmacology , Culture Media, Conditioned/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/metabolism , Janus Kinase 1/drug effects , Janus Kinase 1/metabolism , Multipotent Stem Cells/drug effects , Oligodendroglia/drug effects , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Somatomedins/pharmacology , Stem Cells/drug effects , Triiodothyronine/metabolism , Triiodothyronine/pharmacology
20.
Stroke ; 38(2 Suppl): 724-30, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17261726

ABSTRACT

Survivors of premature birth have a predilection for perinatal brain injury, especially to periventricular cerebral white matter. Periventricular white matter injury (PWMI) is now the most common cause of brain injury in preterm infants and the leading cause of chronic neurological morbidity. The spectrum of chronic PWMI includes focal cystic necrotic lesions (periventricular leukomalacia) and diffuse myelination disturbances. Recent neuroimaging studies support that the incidence of periventricular leukomalacia is declining, whereas focal or diffuse noncystic injury is emerging as the predominant lesion. In a significant number of infants, PWMI appears to be initiated by perturbations in cerebral blood flow that reflect anatomic and physiological immaturity of the vasculature. Ischemic cerebral white matter is susceptible to pronounced free radical-mediated injury that particularly targets immature stages of the oligodendrocyte lineage. Emerging experimental data supports that pronounced ischemia in the periventricular white matter is necessary but not sufficient to generate the initial injury that leads to PWMI. The developmental predilection for PWMI to occur during prematurity appears to be related to both the timing of appearance and regional distribution of susceptible oligodendrocyte progenitors. Injury to oligodendrocyte progenitors may contribute to the pathogenesis of PWMI by disrupting the maturation of myelin-forming oligodendrocytes. There has been substantial recent progress in the understanding of the cellular and molecular pathogenesis of PWMI. The oligodendrocyte progenitor is a key target for preventive strategies to reduce ischemic cerebral white matter injury in premature infants.


Subject(s)
Brain Injuries/pathology , Myelin Sheath/pathology , Premature Birth/pathology , Brain Injuries/diagnosis , Humans , Infant, Newborn , Infant, Premature , Premature Birth/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...