Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 26(16): 4359-69, 2006 Apr 19.
Article in English | MEDLINE | ID: mdl-16624956

ABSTRACT

Perinatal hypoxia/ischemia (H/I) is the leading cause of neurologic injury resulting from birth complications. Recent advances in critical care have dramatically improved the survival rate of infants suffering this insult, but approximately 50% of survivors will develop neurologic sequelae such as cerebral palsy, epilepsy or cognitive deficits. Here we demonstrate that tripotential neural stem/progenitor cells (NSPs) participate in the regenerative response to perinatal H/I as their numbers increase 100% by 3 d and that they alter their intrinsic properties to divide using expansive symmetrical cell divisions. We further show that production of new striatal neurons follows the expansion of NSPs. Increased proliferation within the NSP niche occurs at 2 d after perinatal H/I, and the proliferating cells express nestin. Of those stem-cell related genes that change, the membrane receptors Notch1, gp-130, and the epidermal growth factor receptor, as well as the downstream transcription factor Hes5, which stimulate NSP proliferation and regulate stem cellness are induced before NSP expansion. The mechanisms for the reactive expansion of the NSPs reported here reveal potential therapeutic targets that could be exploited to amplify this response, thus enabling endogenous precursors to restore a normal pattern of brain development after perinatal H/I.


Subject(s)
Cell Proliferation , Hypoxia-Ischemia, Brain/metabolism , Nerve Regeneration/physiology , Neurons/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Female , Hypoxia-Ischemia, Brain/pathology , Neurons/cytology , Pregnancy , Rats , Rats, Wistar , Stem Cells/cytology
2.
Stroke ; 36(10): 2226-31, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16179572

ABSTRACT

BACKGROUND AND PURPOSE: Interleukin-1 (IL-1) is a proinflammatory cytokine implicated in multiple neurodegenerative diseases, including stroke. However, to date, there is no consensus regarding which receptor(s) mediates the detrimental effects of IL-1. We hypothesized that abrogating IL-1 type 1 receptor (IL-1R1) signaling would reduce edema, chemokine expression, and leukocyte infiltration; lower levels of iNOS; and, consequently, decrease free radical damage after mild hypoxia/ischemia (H/I), thus preserving brain cells. METHODS: IL-1R1 null mice and wild-type mice were subjected to a mild H/I insult. MRI was used to measure the area affected at 30 minutes and 48 hours after H/I. An RNAse protection assay was used to evaluate changes in chemokine mRNA expression. RT-PCR was used to assess inducible nitric oxide synthase (iNOS) and endothelial nitric oxide synthase mRNA levels. Immunohistochemistry was used to assess leukocyte infiltration. Western blots were used to assess iNOS and glutamate aspartate transporter protein levels. RESULTS: IL-1R1 null mice had reduced cytotoxic and vasogenic edema. The volume of hyperintense signal on T2-weighted images was reduced on average by 90% at 48 hours after H/I. The induction of multiple chemokine mRNAs was significantly reduced in IL-1R1 null mice compared with wild-type mice at 18 and 72 hours after H/I, which correlated with fewer infiltrating CD3+ leukocytes. Levels of iNOS protein and mRNA (but not glutamate aspartate transporter) were significantly reduced in the IL-1R1 mouse brain. CONCLUSIONS: These findings indicate that abrogating IL-1R1 signaling could protect brain cells subsequent to a mild stroke by reducing edema and immune cell recruitment, as well as by limiting iNOS-mediated free radical damage.


Subject(s)
Edema/pathology , Inflammation/pathology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/physiology , Amino Acid Transport System X-AG/metabolism , Animals , Blotting, Western , Brain/pathology , CD3 Complex/biosynthesis , Chemokines/metabolism , Crosses, Genetic , Cytokines/metabolism , DNA Primers/chemistry , Diffusion Magnetic Resonance Imaging , Disease Models, Animal , Free Radicals , Hypoxia/pathology , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , Interleukin-1/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism , Reperfusion Injury/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/metabolism , Signal Transduction , Stroke/pathology , Time Factors
3.
Dev Neurosci ; 27(2-4): 149-59, 2005.
Article in English | MEDLINE | ID: mdl-16046849

ABSTRACT

With significant improvements in neonatal care, fewer infants sustain severe injury as a consequence of hypoxia/ischemia (H/I). However, the majority of experimental studies have inflicted moderate to severe injuries, or they have assessed damage to the caudal forebrain; therefore, to better understand how a mild H/I episode affects the structures and cells of the rostral forebrain, we assessed the relative vulnerabilities of cells in the neocortex, striatum, corpus callosum, choroid plexus and subventricular zone (SVZ). To inflict mild H/I injury, the right common carotid artery was ligated followed by 1 h of hypoxia (8% O(2)) at 37 degrees C. Regional vulnerabilities were assessed using TUNEL, active caspase-3 and hematoxylin and eosin staining at 24 and 48 h of recovery. Scattered columns of cell death were observed in the neocortex with deep-layer neurons more vulnerable than more superficial neurons. The majority of these dying neurons appeared to be dying apoptotic rather than necrotic deaths. In addition, approximately 1/3 of the apoptotic cells in the neocortex were O4+ oligodendrocyte progenitors. We also observed a decrease in NG2 staining within the affected regions of the forebrain. By contrast, active caspase-3+/S-100beta+ astrocytes were not observed. Neurons and O4+ oligodendrocyte progenitors also died apoptotic deaths within the striatum. The lining cells of the choroid plexus also sustained damage. Elevated numbers of apoptotic cells were observed in the most lateral region of the SVZ and some of these dying cells were O4+. The most novel finding of this study, that oligodendrocyte progenitors in the gray matter are damaged and eliminated as a consequence of perinatal H/I, provides new insights into the histopathology and neurological deficits observed in infants who sustain mild H/I brain injuries.


Subject(s)
Brain/pathology , Hypoxia-Ischemia, Brain/pathology , Neurons/pathology , Oligodendroglia/pathology , Stem Cells/pathology , Animals , Animals, Newborn , Apoptosis/physiology , Brain/blood supply , Carotid Arteries/surgery , Caspase 3 , Caspases/metabolism , Disease Models, Animal , Female , In Situ Nick-End Labeling , Ligation , Male , Rats
4.
J Cereb Blood Flow Metab ; 25(1): 17-29, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15678109

ABSTRACT

Excessive inflammation has been implicated in the progressive neurodegeneration that occurs in multiple neurological diseases, including cerebral ischemia, and elevated levels of the proinflammatory cytokine interleukin-1 (IL-1) have been shown to exacerbate brain damage, whereas diminishing IL-1 levels limits the extent of injury. However, to date there is no consensus regarding which receptor(s) mediates the detrimental effects of IL-1. Because we have previously demonstrated that signaling through the IL-1 type 1 receptor (IL-1R1) is necessary for microglial activation and because results from other studies have implicated microglia as effectors of neurodegeneration, we hypothesized that inactivating the IL-1R1 would decrease the extent of damage caused by a hypoxic-ischemic (H/I) insult. It is shown that a mild insult initiates progressive neurodegeneration that leads to cystic infarcts, which can be prevented by inactivating the IL-1R1. The IL-1R1 null mice also show preserved sensorimotor function at 1 month's recovery. The mild insult induces multiple proinflammatory cytokines and activates microglia, and these responses are dramatically curtailed in mice lacking the IL-1R1. Importantly, the neuroinflammation precedes the progressive enlargement of the infarct, suggesting that the inflammation is causal rather than a consequence of the brain damage. These findings show that abrogating the inflammation consequent to a mild H/I insult will prevent brain damage and preserve neurological function. Additionally, these data incriminate the IL-1R1 as a master proinflammatory cytokine receptor.


Subject(s)
Brain Injuries/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Hypoxia-Ischemia, Brain/metabolism , Receptors, Interleukin-1/metabolism , Recovery of Function/physiology , Animals , Brain Injuries/etiology , Brain Injuries/pathology , Heredodegenerative Disorders, Nervous System/etiology , Heredodegenerative Disorders, Nervous System/pathology , Hypoxia-Ischemia, Brain/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Receptors, Interleukin-1/genetics , Recovery of Function/genetics
5.
J Cereb Blood Flow Metab ; 24(7): 814-25, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15241190

ABSTRACT

Perinatal hypoxic-ischemic (H/I) brain injury remains a major cause of neurologic disability. Because we have previously demonstrated that this insult depletes cells from the subventricular zone (SVZ), the goal of the present investigation was to compare the relative vulnerability to H/I of neural stem cells versus progenitors. The dorsolateral SVZs of P6 rats were examined at 2 to 48 hours of recovery from H/I using hematoxylin and eosin, in situ end labeling (ISEL), terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling (TUNEL), electron microscopy, and immunofluorescence. Pyknotic nuclei and ISEL cells were observed by 4 hours of recovery, peaked at 12 hours, and persisted for at least 48 hours. Many active-caspase-3 cells were observed at 12 hours and they comprised one third of the total TUNEL population. Electron microscopy revealed that hybrid cell deaths predominated at 12 hours of recovery. Importantly, few dying cells were observed in the medial SVZ, where putative stem cells reside, and no nestin medial SVZ cells showed caspase-3 activation. By contrast, active-caspase-3/PSA-NCAM progenitors were prominent in the lateral SVZ. These data demonstrate that early progenitors are vulnerable to H/I, whereas neural stem cells are resilient. The demise of these early progenitors may lead to the depletion of neuronal and late oligodendrocyte progenitors, contributing to cerebral dysgenesis after perinatal insults.


Subject(s)
Brain/pathology , Hypoxia-Ischemia, Brain/pathology , Stem Cells/physiology , Animals , Brain/embryology , Caspase 3 , Caspases/metabolism , Cell Count , Cell Death , Cell Nucleus/ultrastructure , Enzyme Activation , Female , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Microscopy, Electron , Neurons/physiology , Oligodendroglia/physiology , Pregnancy , Rats , Rats, Wistar , Time Factors
6.
Dev Neurosci ; 26(2-4): 266-74, 2004.
Article in English | MEDLINE | ID: mdl-15711066

ABSTRACT

Hypoxia-ischemia (H/I) as a result of asphyxia at term remains a major cause of neurologic disability. Our previous studies in the P7 rat model of perinatal H/I have shown that progenitors within the subventricular zone (SVZ) are vulnerable to this insult. Since many investigators are using transgenic and knockout mice to determine the importance of specific molecules in the evolution of damage after a stroke, there is a need to perform comparative studies on the relative vulnerability of the mouse SVZ. Here we assess damage to the SVZ of 5-, 7- and 10-day-old C57BL/6 mice after unilateral common carotid artery cauterization followed by 70 min of H/I (10% O2). Whereas 5- and 7-day-old mice sustained little SVZ damage as assessed by hematoxylin and eosin staining, there was a 16% reduction of cellularity in 10-day-old animals by 18 h of recovery. Additionally, swollen cells were observed in the medial region of the SVZ of 10-day-old mice. However, few caspase-3+ and TUNEL+ cells were observed in this region, which contains the putative neural stem cells. Rather, the majority of the dying cells were situated in the mediolateral and lateral tail of the SVZ. At 18 h of recovery, there was a 2-fold increase in the frequency of TUNEL+ cells in the ipsilateral SVZ as well as a 3-fold increase in the frequency of active-caspase-3+ cells. We conclude that progenitors within the neonatal mouse SVZ are vulnerable to hypoxic/ischemic insult. The demise of these early progenitors likely leads to depletion of neuronal and late oligodendrocyte progenitors, contributing to cerebral dysgenesis.


Subject(s)
Cerebral Cortex/pathology , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Lateral Ventricles/pathology , Neurons/pathology , Stem Cells/pathology , Age Factors , Animals , Animals, Newborn , Apoptosis/physiology , Caspase 3 , Caspases/metabolism , Causality , Cerebral Cortex/growth & development , Cerebral Cortex/physiopathology , Disease Models, Animal , Female , In Situ Nick-End Labeling , Lateral Ventricles/growth & development , Lateral Ventricles/physiopathology , Male , Mice , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/metabolism , Stem Cells/metabolism
7.
Prog Neurobiol ; 69(1): 49-69, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12637172

ABSTRACT

There has been enormous progress in uncovering the contributions of the subventricular zone (SVZ) to the developing brain. Here, we review the roles of four anatomically defined embryologic divisions of the SVZ of the mammalian brain: the lateral ganglionic eminence (LGE), the medial ganglionic eminence (MGE), the caudal ganglionic eminence (CGE), and the fetal neocortical SVZ (SVZn), as well as the roles of the two major anatomically defined regions of the postnatal SVZ, the anterior SVZ (SVZa) and the dorsolateral SVZ (SVZdl). We describe the types of cells within each subdivision of the SVZ, the types of brain cells that they generate during embryonic, fetal, and perinatal development, and when known the mechanisms that regulate their differentiation. This review provides a critical analysis of the literature, from which current and future studies on the SVZ can be formulated and evaluated.


Subject(s)
Brain/embryology , Brain/growth & development , Cerebral Ventricles/embryology , Cerebral Ventricles/growth & development , Neuroglia , Neurons , Animals , Cell Differentiation , Cell Movement , Humans , Neocortex/embryology , Neocortex/growth & development , Prosencephalon/embryology , Prosencephalon/growth & development , Stem Cells
8.
Dev Neurosci ; 24(5): 426-36, 2002.
Article in English | MEDLINE | ID: mdl-12640182

ABSTRACT

Cerebral hypoxia/ischemia (H/I) of the premature infant is a major cause of cerebral palsy and mental retardation. An important determinant of the ultimate outcome from this insult is the extent to which the stem cells and progenitors in the brain are affected. Irreversible injury to these cells will impair normal development of the infant's brain and, hence, its function. In the present study, we examine early intervals after H/I to identify which cells in the periventricular region are most vulnerable. At 0 h of recovery from a perinatal H/I insult, the choroid plexus shows extensive necrotic damage. The adjacent ependymal and subependymal cells are also affected. Swelling of the ependymal and medial subependymal cells is observed; however, these cells rarely sustain permanent damage. By contrast, cells in the most lateral aspect of the subventricular zone (SVZ) show more delayed, but extensive apoptotic and hybrid cell deaths. Interestingly, activated macrophages/microglia are observed adjacent to the swollen ependymal cells as well as within the affected subependyma. We conclude that the choroid plexus is an especially vulnerable structure in the immature brain, whereas the ependymal and adjacent subependymal cells are relatively resistant to damage. As the medial aspect of the SVZ contains neural stem cells, we predict that neural stem cells will be especially resistant to perinatal H/I brain damage.


Subject(s)
Choroid Plexus/pathology , Hypoxia-Ischemia, Brain/pathology , Animals , Animals, Newborn , Apoptosis , Choroid Plexus/metabolism , Choroid Plexus/ultrastructure , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corpus Striatum/ultrastructure , Ependyma/metabolism , Ependyma/pathology , Ependyma/ultrastructure , Hypoxia-Ischemia, Brain/metabolism , Immunohistochemistry , Lectins/metabolism , Microscopy, Electron , Necrosis , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...