Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Neuroscience ; 194: 195-207, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21840378

ABSTRACT

Our previous studies show that insulin-like growth factor-1 (IGF-1) can either protect against or increase lipopolysaccharide (LPS)-induced damage in the developing brain, depending on the dose, when it is co-administered with LPS through intracerebral injection. To further explore effects of IGF-1 on central inflammation associated brain injury, IGF-1 was administered through intranasal infusion in the current study. Postnatal day 5 (P5) rats were exposed to LPS at a dose of 1 µg/g body weight or sterile saline through intracerebral injection. Recombinant human insulin-like growth factor-1 (rhIGF-1) at a dose of 50 µg/pup or vehicle was administered intranasally 1 or 2 h after the LPS injection. Neonatal LPS exposure resulted in oligodendrocyte (OL) and white matter injury in the P6 or P21 rat brain. The damages include dilatation of lateral ventricles, pyknotic cell death, loss of OL progenitor cells and mature OLs in the cingulum area, and impairment of myelination at the corpus callosum area. Neurological dysfunctions were observed in juvenile rats with neonatal LPS exposure. Intranasal IGF-1 treatment at either 1 or 2 h after LPS exposure significantly attenuated LPS-induced brain injury and improved some behavioral deficits. Intranasal IGF-1 treatment also reduced infiltration of polymorphonuclear (PMN) leukocytes and activation of microglia in the rat brain 24 h after LPS exposure, but it did not prevent the elevation in concentrations of interleukin-1ß (IL-1ß) and tumor necrosis factor alpha (TNFα) in the LPS-exposed rat brain during the first 24 h. This is an indication that direct anti-inflammation might not be the primary mechanism for the protection of IGF-1, and other mechanisms, such as anti-apoptotic effects, are likely involved in its protective effects.


Subject(s)
Brain Chemistry/physiology , Brain/metabolism , Inflammation Mediators/administration & dosage , Insulin-Like Growth Factor I/administration & dosage , Lipopolysaccharides/administration & dosage , Wallerian Degeneration/physiopathology , Wallerian Degeneration/therapy , Administration, Intranasal , Animals , Animals, Newborn , Brain/growth & development , Brain/pathology , Disease Models, Animal , Female , Humans , Inflammation Mediators/physiology , Insulin-Like Growth Factor I/physiology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Pregnancy , Rats , Rats, Sprague-Dawley , Wallerian Degeneration/pathology
2.
Neuroscience ; 179: 223-32, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21277350

ABSTRACT

Dexamethasone (Dex) provides neuroprotection against subsequent hypoxia ischemia (HI) in newborn rats, but the mechanism of this neuroprotection is not well understood. It is known that vascular endothelial growth factor A (VEGF) has neuroprotective effects. The objective of this study was to evaluate the role of the VEGF signaling pathway in the Dex-induced neuroprotection in newborn rats. Seven-day-old rat pups had the right carotid artery permanently ligated followed by 140 or 160 min of hypoxia (8% oxygen). Rat pups received two i.p. injections of either saline or Dex (0.25 mg/kg) at 24 and 4 h before HI exposure. To quantify the effects of a glucocorticoid receptor (GR) blocker, on postnatal day (PD) 6 and 15 min prior to Dex treatment rat pups received s.c. vehicle or RU486 (GR blocker, 60 mg/kg). After 24 h at PD 7, all rat pups had HI as described earlier. To quantify the effects of a VEGFR 2 blocker, at 24 h after Dex/Veh treatment (PD7), SU5416, a VEGFR 2 inhibitor or vehicle was injected intracerebroventricularly in the right hemisphere at 30 min before and 2 h after HI. Dex pre-treatment reduced brain injury and enhanced the HI-induced brain VEGF protein while a GR blocker inhibited these effects. Treatment with VEGFR 2 blocker decreased Dex-induced neuroprotection also. Dex pre-treatment enhanced the HI-induced increase in mRNA expression of VEGF splice variants and decreased the HI-induced reduction of Akt phosphorylation. Additionally, it also decreased HI-induced increase of caspase-3 activity and DNA fragments in neonatal rat brain. We conclude that Dex provides robust neuroprotection against subsequent HI in newborn rats via GR likely with the partial involvement of VEGF signaling pathway.


Subject(s)
Brain/drug effects , Dexamethasone/pharmacology , Hypoxia-Ischemia, Brain/prevention & control , Neuroprotective Agents/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Animals, Newborn , Blotting, Western , Brain/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Hypoxia-Ischemia, Brain/metabolism , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
3.
Neuroscience ; 168(1): 240-52, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20346393

ABSTRACT

Our previous study showed that perinatal exposure to interleukin-1beta (IL-1beta), an inflammatory cytokine, induces acute injury to developing white matter in the neonatal rat brain, and alpha-phenyl-n-tert-butyl-nitrone (PBN), a free radical scavenger and antioxidant, protects against IL-1beta-induced acute brain injury. The objective of the present study was to further examine whether perinatal exposure to IL-1beta resulted in persistent brain damage and neurological disabilities, and whether PBN offers lasting protection. Intracerebral injection of IL-1beta (1 microg/kg) was performed in postnatal day 5 (P5) Sprague-Dawley rat pups and PBN (100 mg/kg) or saline was administered intraperitoneally 5 min after IL-1beta injection. Perinatal IL-1beta exposure significantly affected neurobehavioral functions in juvenile rats. Although some neurobehavioral deficits such as performance in negative geotaxis, cliff avoidance, beam walking, and locomotion were spontaneously reversible, sustained deficits such as poor performance in the vibrissa-elicited forelimb-placing test, the pole test, the passive avoidance task, and the elevated plus-maze task were still observable at P21. Perinatal IL-1beta exposure resulted in persistent brain damage including enlargement of ventricles, loss of mature oligodendrocytes, impaired myelination as indicated by the decrease in myelin basic protein immunostaining, axonal and dendritic injury, and loss of hippocampal CA1 neurons and tyrosine hydroxylase positive neurons in the substantia nigra and ventral tegmental areas of the rat brain. Treatments with PBN provided lasting protection against the IL-1beta-induced brain injury and improved the associated neurological dysfunctions in juvenile rats, suggesting that prompt treatments for brain injury induced by perinatal infection/inflammation might have important long-term consequences.


Subject(s)
Behavior, Animal/drug effects , Cerebrovascular Disorders/prevention & control , Cyclic N-Oxides/therapeutic use , Interleukin-1beta/physiology , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Animals , Animals, Newborn , Avoidance Learning/drug effects , Axons/drug effects , Axons/pathology , Brain/drug effects , Brain/pathology , Cerebrovascular Disorders/chemically induced , Cerebrovascular Disorders/psychology , Female , Interleukin-1beta/toxicity , Lipopolysaccharides/pharmacology , Male , Maze Learning/drug effects , Microglia/drug effects , Microglia/physiology , Neurons/pathology , Rats , Tyrosine 3-Monooxygenase/metabolism
4.
Neuroscience ; 158(2): 823-32, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19007863

ABSTRACT

The use of dexamethasone (Dex) in premature infants to prevent and/or treat bronchopulmonary dysplasia can adversely affect early neurodevelopment and probably result in loss of cerebral volume. Vascular endothelial growth factor A (VEGF), specifically VEGF(164) isoform has neurotrophic, neuroprotective and neurogenesis enhancing effects. Previous studies have demonstrated that Dex usually down-regulates VEGF. In the present study we investigated the effect of Dex on brain growth and VEGF in the neonatal rat brain. The pups in each litter were divided into the vehicle (n=84) or Dex-treated (n=98) groups. Rat pups in the Dex group received one of three different regimens of i.p. Dex which included tapering doses on postnatal days 3-6 (0.5, 0.25, 0.125 and 0.06 mg/kg, respectively), or repeated doses of 0.5 or 1 mg/kg/day on postnatal days 4-6 or single dose of 0.031, 0.06, 0.125, 0.25 or 0.5 mg/kg on postnatal day 6. The total VEGF protein and mRNA expression of the three main VEGF splice variants (VEGF(120), VEGF(164), and VEGF(188)) were measured in the rat pup brain using enzyme-linked immunosorbent assay and real-time reverse transcription polymerase chain reaction, respectively. Treatment with Dex significantly decreased the gain of body and brain weight. The tapering and repeated doses of Dex significantly increased caspase-3 activity, VEGF protein and the expression of mRNA of VEGF(164) and VEGF(188) splice variants but the single dose did not. We conclude that Dex is neurodegenerative in the developing brain but also increases VEGF which may play a neurotrophic and neuroprotective role.


Subject(s)
Brain/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Neurogenesis/drug effects , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Age Factors , Animals , Animals, Newborn , Body Weight/drug effects , Caspase 3/metabolism , Dose-Response Relationship, Drug , Female , Male , Organ Size/drug effects , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/genetics
5.
Neuroscience ; 151(3): 737-44, 2008 Feb 06.
Article in English | MEDLINE | ID: mdl-18191905

ABSTRACT

Although white matter damage is a fundamental neuropathological feature of periventricular leukomalacia (PVL), the motor and cognitive deficits observed later in infants with PVL indicate the possible involvement of cerebral neuronal dysfunction. Using a previously developed rat model of white matter injury induced by cerebral lipopolysaccharide (LPS) injection, we investigated whether LPS exposure also results in neuronal injury in the neonatal brain and whether alpha-phenyl-n-tert-butyl-nitrone (PBN), an antioxidant, offers protection against LPS-induced neuronal injury. A stereotactic intracerebral injection of LPS (1 mg/kg) was performed in Sprague-Dawley rats (postnatal day 5) and control rats were injected with sterile saline. LPS exposure resulted in axonal and neuronal injury in the cerebral cortex as indicated by elevated expression of beta-amyloid precursor protein, altered axonal length and width, and increased size of cortical neuronal nuclei. LPS exposure also caused loss of tyrosine hydroxylase positive neurons in the substantia nigra and the ventral tegmental areas of the rat brain. Treatments with PBN (100 mg/kg) significantly reduced LPS-induced neuronal and axonal damage. The protection of PBN was associated with an attenuation of oxidative stress induced by LPS as indicated by the reduced number of 4-hydroxynonenal, malondialdehyde or nitrotyrosine positive cells in the cortical area following LPS exposure, and with the reduction in microglial activation stimulated by LPS. The finding that an inflammatory environment may cause both white matter and neuronal injury in the neonatal brain supports the possible anatomical correlate for the intellectual deficits and the other cortical and deep gray neuronal dysfunctions associated with PVL. The protection of PBN may indicate the potential usefulness of antioxidants for treatment of these neuronal dysfunctions.


Subject(s)
Brain Injuries/pathology , Cyclic N-Oxides/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Axons/drug effects , Brain Injuries/chemically induced , CD11b Antigen/metabolism , Cerebral Ventricles/drug effects , Dendrites/drug effects , Female , Interleukin-1beta/metabolism , Lipopolysaccharides , Male , Nerve Tissue Proteins/metabolism , Neurons/pathology , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
6.
Neuroscience ; 141(2): 745-755, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16713113

ABSTRACT

There are increasing data in support of the hypothesis that inflammatory cytokines are involved in neonatal white matter damage. Despite extensive study of the proinflammatory cytokines tumor necrosis factor-alpha and interleukin-1beta, the role of interleukin-6 in the development of white matter damage is largely unknown. In the present study, the role(s) of interleukin-6 in mediating lipopolysaccharide-induced brain injury and behavioral changes was investigated by the intracerebral injection of lipopolysaccharide with interleukin-6 neutralizing antibody in the 5-day-old rat brain. Brain injury was examined in brain sections at postnatal day 8 and postnatal day 21. Behavioral tests including righting reflex, wire hanging maneuver, cliff avoidance, locomotor activity, gait analysis, responses in the elevated plus-maze and passive avoidance were performed from postnatal day 3 to postnatal day 21. Changes in astroglia, microglia and oligodendrocytes were studied using immunohistochemistry in the postnatal day 21 rat brain. Our results show that interleukin-6 antibody attenuated lipopolysaccharide-induced brain lateral ventricle dilation and improved neurobehavioral performance. Interleukin-6 antibody also suppressed lipopolysaccharide-induced astrogliosis and microglial activation, and increased the number of oligodendrocytes in white matter. However, no changes of tumor necrosis factor-alpha and interleukin-1beta were detected. In contrast, no histopathological changes and glial activation were observed in rats injected with only interleukin-6. The present study indicates that the contribution to brain injury by interleukin-6 depends on its interaction with other lipopolysaccharide-induced agents and not on interleukin-6 alone.


Subject(s)
Behavior, Animal/drug effects , Brain Injuries/chemically induced , Brain Injuries/physiopathology , Interleukin-6/physiology , Lipopolysaccharides/toxicity , Age Factors , Animals , Animals, Newborn , Antibodies/administration & dosage , Avoidance Learning/drug effects , Body Weight/drug effects , Brain Injuries/pathology , Cell Count/methods , Enzyme-Linked Immunosorbent Assay/methods , Female , Immunohistochemistry/methods , Interleukin-1/metabolism , Interleukin-6/immunology , Male , Maze Learning/drug effects , Motor Activity/drug effects , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
7.
Neuroscience ; 137(2): 425-35, 2006.
Article in English | MEDLINE | ID: mdl-16289838

ABSTRACT

The role of minocycline in preventing white matter injury, in particular the injury to developing oligodendrocytes was examined in a neonatal rat model of hypoxia-ischemia. Hypoxia-ischemia was achieved through bilateral carotid artery occlusion followed by exposure to hypoxia (8% oxygen) for 15 min in postnatal day 4 Sprague-Dawley rats. A sham operation was performed in control rats. Minocycline (45 mg/kg) or normal phosphate-buffered saline was administered intraperitoneally 12 h before and immediately after bilateral carotid artery occlusion+hypoxia and then every 24 h for 3 days. Nissl staining revealed pyknotic cells in the white matter area of the rat brain 1 and 5 days after hypoxia-ischemia. Hypoxia-ischemia insult also resulted in apoptotic oligodendrocyte cell death, loss of O4+ and O1+ oligodendrocyte immunoreactivity, and hypomyelination as indicated by decreased myelin basic protein immunostaining and by loss of mature oligodendrocytes in the rat brain. Minocycline significantly attenuated hypoxia-ischemia-induced brain injury. The protective effect of minocycline was associated with suppression of hypoxia-ischemia-induced microglial activation as indicated by the decreased number of activated microglia, which were also interleukin-1beta and inducible nitric oxide synthase expressing cells. The protective effect of minocycline was also linked with reduction in hypoxia-ischemia-induced oxidative and nitrosative stress as indicated by 4-hydroxynonenal and nitrotyrosine positive oligodendrocytes, respectively. The reduction in hypoxia-ischemia-induced oxidative stress was also evidenced by the decreases in the content of 8-isoprostane in the minocycline-treated hypoxia-ischemia rat brain as compared with that in the vehicle-treated hypoxia-ischemia rat brain. The overall results suggest that reduction in microglial activation may protect developing oligodendrocytes in the neonatal brain from hypoxia-ischemia injury.


Subject(s)
Brain/drug effects , Hypoxia-Ischemia, Brain/drug therapy , Minocycline/pharmacology , Oligodendroglia/drug effects , Animals , Animals, Newborn , Antigens, Surface/metabolism , Biomarkers/metabolism , Brain/metabolism , Brain/physiopathology , Carotid Artery, Common , Cell Differentiation/drug effects , Cell Differentiation/physiology , Disease Models, Animal , Free Radicals/metabolism , Gliosis/drug therapy , Gliosis/physiopathology , Gliosis/prevention & control , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Ligation , Microglia/drug effects , Microglia/metabolism , Minocycline/therapeutic use , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/metabolism , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oligodendroglia/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Stem Cells/metabolism , Treatment Outcome
8.
Neuroscience ; 133(1): 159-68, 2005.
Article in English | MEDLINE | ID: mdl-15893639

ABSTRACT

Our previous studies have shown that intracerebral administration of endotoxin, lipopolysaccharide (LPS), induces selective white matter injury and hypomyelination in the neonatal rat brain and that the LPS-induced brain injury is associated with activation of microglia. To test the hypothesis that inhibition of microglial activation may protect against LPS-induced white matter injury, we examined roles of minocycline, a putative suppressor of microglial activation, on LPS-induced brain injury in the neonatal rat. A stereotactic intracerebral injection of LPS (1 mg/kg) was performed in postnatal day 5 Sprague-Dawley rats and control rats were injected with sterile saline. Minocycline (45 mg/kg) was administered intraperitoneally 12 h before and immediately after LPS injection and then every 24 h for 3 days. Inflammatory responses, activation of microglia and brain injury were examined 1 and 3 days after LPS injection. LPS injection resulted in brain injury in selective brain areas, including bilateral ventricular enlargement, cell death at the sub- and periventricular areas, loss of O4+ and O1+ oligodendrocyte (OL) immunoreactivity and hypomyelination, as indicated by decreased myelin basic protein immunostaining, in the neonatal rat brain. Minocycline administration significantly attenuated LPS-induced brain injury in these rat brains. The protective effect of minocycline was associated with suppressed microglial activation as indicated by the decreased number of activated microglial cells following LPS stimulation and with consequently decreased elevation of interleukin 1beta and tumor necrosis factor-alpha concentrations induced by LPS and a reduced number of inducible nitric oxide synthase expressing cells. Protection of minocycline was also linked with the reduction in LPS-induced oxidative stress, as indicated by 4-hydroxynonenal positive OLs. The overall results suggest that reduction in microglial activation may protect the neonatal brain from LPS-induced white matter injury and inhibition of microglial activation might be an effective approach for the therapeutic treatment of infection-induced white matter injury.


Subject(s)
Anti-Bacterial Agents/pharmacology , Brain Diseases/chemically induced , Brain Diseases/prevention & control , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Minocycline/pharmacology , Neuroprotective Agents , Animals , Animals, Newborn , Brain/growth & development , Brain Diseases/pathology , Cerebral Ventricles/pathology , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Injections , Interleukin-1/analysis , Interleukin-1/biosynthesis , Macrophage Activation/drug effects , Male , Microglia/drug effects , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/biosynthesis
9.
Brain Res ; 914(1-2): 15-22, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11578593

ABSTRACT

We used cDNA arrays to investigate differentially expressed genes in astrocytes challenged with lipopolysaccharide (LPS). Astrocyte cultures were prepared from 1-day-old rat brains. Purified astrocytes were treated with LPS (1 microg/ml) for 2, 8 and 48 h. Differentially expressed genes in these astrocytes were examined with Atlas rat cDNA arrays. At all the three time points studied, three genes were found consistently up-regulated: I-kappaB alpha chain, NF-kappaB, and interferon induced protein. In addition to these three, six other genes were also up-regulated at 2 and 8 h. They were genes encoding vascular cell adhesion protein 1 (VCAM-1), interferon regulatory factor 1 (IRF-1), mitochondrial hydroxymethylglutaryl-CoA synthase (HMG-CoA synthase), aldehyde dehydrogenase 2, macrophage inflammatory protein 1 (MIP-1) and neurotensin receptor 2. At these two time points, three genes were down-regulated: copper-zinc-containing superoxide dismutase 1 (SOD-1), insulin-like growth factor binding protein 1 (IGFBP-1), and insulin-like growth factor binding protein 3 (IGFBP-3). Expression of several differentially expressed genes in cDNA array (I-kappaB, VCAM-1 and MIP-3) were further confirmed by reverse transcription polymerase chain reaction study. The prominently modulated genes could be classified into three categories: nuclear transcription factors, pro-inflammatory cytokines/chemokines and metabolic enzymes. Application of pyrrolidine dithiocarbamate, an inhibitor of nuclear factor-kB (NF-kappaB), prior to LPS stimulation not only prevented up-regulation of NF-kappaB gene expression, but also completely blocked up-regulation of pro-inflammatory cytokine genes (TNF-alpha and interleukin-1beta) and two chemokine genes: CXC chemokine LIX and CC chemokine MIP-3 alpha. These results indicate that both up-regulation of inflammatory cytokine expression and down-regulation of growth factor expression are probably involved in the response of astrocytes upon exposure to LPS.


Subject(s)
Astrocytes/drug effects , Cytokines/genetics , Encephalitis/genetics , Gene Expression Regulation/immunology , Growth Substances/genetics , Lipopolysaccharides/pharmacology , Transcription Factors/genetics , Animals , Animals, Newborn , Astrocytes/immunology , Astrocytes/metabolism , Bacterial Infections/complications , Bacterial Infections/immunology , Bacterial Infections/metabolism , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cytokines/immunology , Cytokines/metabolism , Down-Regulation/genetics , Encephalitis/immunology , Encephalitis/metabolism , Growth Substances/immunology , Growth Substances/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Transcription Factors/immunology , Transcription Factors/metabolism , Up-Regulation/genetics
10.
Neurochem Res ; 26(5): 487-95, 2001 May.
Article in English | MEDLINE | ID: mdl-11513474

ABSTRACT

Effects of intrauterine hypoxia-ischemia (HI) on expression of the NMDA receptor subunits as well as on [3H]MK-801 binding of the NMDA receptor were studied in 1-day to 30-day old rat brain. Intrauterine HI conditions were achieved on gestation day 17 by clamping the uterine vasculature for 30 min followed by removal of the clamps to permit reperfusion. As determined by reverse-transcriptase polymerase chain reaction, prenatal HI significantly reduced mRNA expression of the NRI subunit of the NMDA receptor in the hippocampus of 4, 8, and 30-day old rat brains. NR2A and NR2B subunit mRNAs were expressed in the hippocampus and the cortex of both the control and the prenatal HI rat brains. Intrauterine HI did not significantly affect expression of either the NR2A or NR2B subunit mRNA. Consistent with the RT-PCR data, protein expression of the NRI subunit in the hippocampus, but not the cortex, of 21-day old prenatal HI rat brains was significantly decreased as compared to the control rat brain. Intrauterine HI also significantly reduced binding affinity, but not the number of binding sites, of the NMDA receptor to [3H]MK-801, a noncompetitive antagonist of the NMDA receptor, in the hippocampus of 21-day old rat brain. The overall results suggest that prenatal HI-induced reduction of NRI expression and the altered binding ability of the NMDA receptor in the young rat brain may contribute to other long-lasting effects of intrauterine HI that we reported previously.


Subject(s)
Animals, Newborn/metabolism , Brain Ischemia/metabolism , Brain/metabolism , Fetal Diseases/metabolism , Hypoxia/metabolism , Prenatal Exposure Delayed Effects , Receptors, N-Methyl-D-Aspartate/metabolism , Aging/metabolism , Animals , Animals, Newborn/growth & development , Brain/growth & development , Dizocilpine Maleate/metabolism , Excitatory Amino Acid Antagonists/metabolism , Female , Pregnancy , RNA, Messenger/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/genetics
11.
Brain Res ; 898(1): 126-35, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11292456

ABSTRACT

Chronic ischemic brain injuries were studied in 7- and 14-day-old rat pups, which were subjected to bilateral carotid artery occlusion (BCAO) on postnatal day 1. BCAO preferentially injured white matter in the corpus callosum, subcortex and internal capsule areas while largely spared cortical neurons. White matter rarefaction in the corpus callosum was observed in 12 out of the 17 BCAO rat brains and significantly enlarged lateral ventricles were found in five out of seven P14 BCAO rat brains. These white matter changes were similar to injuries found in newborn infants with periventricular leukomalacia (PVL). White matter injuries in the 7-day-old BCAO rat brain were accompanied with increased activation of microglia/macrophages, as indicated by ED1 and OX42 positive immunostaining. Immature oligodendrocytes in the 7-day-old BCAO rat brain, as indicated by O4+/O1+ staining, were much fewer than in the sham-operated rat brain. Immunostaining for myelin basic protein (MBP) at the fimbria hippocampus and the internal capsule areas in the 7-day-old BACO rat brain was also much less than in the control rat brain. Consistent with the immunostaining data, MBP mRNA expression in the 7-day-old, but not in the 14-day-old, BCAO rat brain was significantly less than in the control rat brain. The overall results suggest that pre-oligodendrocytes and immature oligodendrocytes might be major targets for chronic ischemic insults and activated microglia/macrophages are possibly involved in the process of white matter injury.


Subject(s)
Brain Ischemia/pathology , Brain/pathology , Animals , Animals, Newborn/physiology , Biomarkers , Brain/metabolism , Brain Ischemia/metabolism , Chronic Disease , Immunohistochemistry/methods , Macrophages/pathology , Microglia/pathology , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Oligodendroglia/pathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Staining and Labeling
12.
J Neurosci Res ; 62(4): 510-20, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11070494

ABSTRACT

Oligodendrocytes are the primary cells injured in periventricular leukomalacia (PVL), a predominant form of brain white matter lesion in preterm infants. To explore the possible linkage between white matter injury and maternal infection, purified rat O-2A progenitor (Oligodendrocyte-type 2 astrocyte progenitor) cell cultures were used as a model in studying the effects of lipopolysaccharide (LPS), an endotoxin, on survival and differentiation of oligodendrocytes and the involvement of other glial cells in the effects of LPS. O-2A progenitor cells were cultured from optic nerves of 7-day-old rat pups in a chemically defined medium (CDM). Astrocyte and microglia cell cultures were prepared from the cortex of 1-day-old rat brains in the CDM. Direct treatment of LPS (1 microg/ml) to O-2A cells had no effect on viability or differentiation of these cells. When O-2A progenitor cells were cultured in the conditioned medium obtained from either astrocyte or microglial cell cultures for 48 hr, survival rate and differentiation of O-2A cells into mature oligodendrocytes were greatly enhanced as measured by the MTT assay and immunocytochemistry. The conditioned medium obtained from astrocytes or microglia treated with LPS for 48 hr, however, failed to show such a promotional effect on viability and differentiation of O-2A cells. When 5 microg/ml LPS was used to stimulate astrocytes or microglia, the conditioned medium from these glial cell cultures caused O-2A cell injury. The overall results indicate that astrocytes and microglia may promote viability and differentiation of O-2A progenitor cells under physiological conditions, but they may also mediate cytotoxic effects of LPS on oligodendrocytes under an infectious disease biochemical environment.


Subject(s)
Astrocytes/drug effects , Leukomalacia, Periventricular/physiopathology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Oligodendroglia/drug effects , Stem Cells/drug effects , Animals , Animals, Newborn , Astrocytes/metabolism , Bacterial Infections/complications , Cell Communication/drug effects , Cell Communication/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Culture Media, Conditioned/pharmacology , Cytokines/adverse effects , Endotoxins/adverse effects , Female , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical , Leukomalacia, Periventricular/etiology , Leukomalacia, Periventricular/pathology , Lipopolysaccharides/metabolism , Microglia/metabolism , Oligodendroglia/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/metabolism
13.
Int J Dev Neurosci ; 18(6): 585-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10884603

ABSTRACT

Phospholipase D (PLD) is a ubiquitous enzyme involved in many important cellular functions. Rat brain PLD isoforms have recently been cloned and characterized, but the expression of this gene has not been well studied. We thus examined the developmental expression of PLD2 in postnatal brains by Northern blotting employing a non-radioactive RNA probe. Two PLD2 mRNA transcripts of 3.9 and 10.8 kb were detected in different brain regions and various tissues of the rat. In postnatal brains, the level of PLD2 mRNA transcripts are low 1 day after birth, increase progressively during development, and reach the maximum level in the adult brain. We thus conclude that rat brain PLD2 mRNA is developmentally regulated.


Subject(s)
Brain/enzymology , Gene Expression Regulation, Developmental/genetics , Phospholipase D/genetics , RNA, Messenger/biosynthesis , Aging/metabolism , Animals , Blotting, Northern , Brain/growth & development , Brain Chemistry , Organ Specificity , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
14.
Neurosci Lett ; 285(1): 5-8, 2000 May 05.
Article in English | MEDLINE | ID: mdl-10788694

ABSTRACT

To explore the role of nitric oxide (NO) in the hypoxic-ischemic (HI) tolerance phenomenon, NO production and brain injury following neonatal hypoxia-ischemia (induced by unilateral common carotid artery ligation followed by hypoxic exposure) were assessed in rat pups with or without HI preconditioning. A previously demonstrated prenatal HI rat model of preconditioning was used in this study. On G17, rat fetuses were subjected to either HI in utero (PreHI) for 30 min or a sham operation (SH). The PreHI treatment provided significant protection against neonatal HI-induced brain injury, as indicated by decreased ipsilateral brain weight reduction, less severe tissue damage, and decreased activation of caspase-3. Concomitant with the protective effect of prenatal HI preconditioning, elevation of nitrite/nitrate content in the ipsilateral cortex of the brain, as an indirect measure of NO production, was significantly lower in the PreHI group than in the SH group following neonatal HI. The protective effect of prenatal HI preconditioning could be reversed by sodium nitroprusside (SNP), a spontaneous NO donor, while SNP had no effect on neonatal HI-induced brain injury in the SH group. Intraperitoneal administration of SNP to pups from the PreHI group (2 mg/kg, 24 and 1.5 h before neonatal HI) increased neonatal HI-induced brain injury similar to that observed in the SH group. On the other hand, L-N(G)-nitro-arginine (2 mg/kg, i.p., 1.5 h before the hypoxic exposure), an NO synthase inhibitor, significantly attenuated neonatal HI-induced brain injury in the SH group. The overall results indicate that reduced NO production in the preconditioned rat brain contributes to prenatal HI-induced tolerance to neonatal HI brain injury.


Subject(s)
Brain Ischemia/metabolism , Fetal Hypoxia/metabolism , Hypoxia-Ischemia, Brain/metabolism , Nitric Oxide/metabolism , Animals , Animals, Newborn , Brain Ischemia/enzymology , Brain Ischemia/physiopathology , Brain Ischemia/prevention & control , Female , Fetal Hypoxia/enzymology , Fetal Hypoxia/physiopathology , Fetal Hypoxia/prevention & control , Hypoxia-Ischemia, Brain/enzymology , Hypoxia-Ischemia, Brain/physiopathology , Hypoxia-Ischemia, Brain/prevention & control , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type I , Oxidation-Reduction , Pregnancy , Rats , Rats, Sprague-Dawley
15.
Pediatr Res ; 47(1): 64-72, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10625084

ABSTRACT

Induction of proinflammatory cytokines has been proposed to be a link between prenatal maternal intrauterine infection and neonatal brain damage. It is known that the endotoxin, lipopolysaccharide (LPS), released during bacterial infection crosses the placenta. Cytokine induction in the fetal rat brain after maternal administration of LPS was determined by reverse transcriptase-polymerase chain reaction method. LPS suspension in pyrogen-free saline was administered (i.p.) to pregnant rats at 18 d of gestation. The control group was treated with pyrogen-free saline. Expression of the proinflammatory cytokines, tumor necrosis factor-alpha and IL-1beta mRNA, in the fetal rat brain was increased in a dose-dependent manner at 1 h after LPS administration. The great increase in expression of IL-1beta mRNA was only observed at 1 h after injection of LPS (4 mg/kg), whereas the increased expression of tumor necrosis factor-alpha was still detectable from 4 to 24 h after LPS administration. Brain injuries were examined by immunohistochemistry in 8-d-old rat pups born to the dams that were consecutively treated with LPS (500 microg/kg) or pyrogen-free saline on gestation d 18 and 19. No apparent necrotic tissue damage was found in either the LPS group or the control group. Myelin basic protein staining, as a marker of myelin, was clearly observed in the internal capsule and the fimbria hippocampus in the rat brain from the control group. Myelin basic protein staining was much less and weaker in the brains of the LPS-treated group. Glial fibrillary acidic protein-positive astrocytes were observed in both the control and the LPS-treated groups. The LPS-treated group appeared to have more glial fibrillary acidic protein-positive astrocytes in the hippocampal and the cortex areas of the brain than the control group. Immunoblotting data showed that glial fibrillary acidic protein content in the cortex or the hippocampus of the LPS-treated rat brain was higher than in the control group. OX-42-positive staining (a marker of the type 3 complement receptors) of microglial cells was greatly reduced in the 8-d-old rat brain after maternal LPS administration. However, histochemistry with tomato lectin showed that staining of both amoeboid and ramified microglial cells in the LPS-treated rat brain was similar to that in the control group. The overall results indicate that maternal LPS administration induces an increased expression of IL-1beta and tumor necrosis factor-alpha mRNA in the fetal brain. Maternal LPS administration also increases glial fibrillary acidic protein-positive astrocytes, decreases myelin basic protein and alters immunoreactivity of microglia in the brain of offspring. Although results from the current study do not provide direct evidence linking LPS-induced cytokines with the abnormalities in the neonatal rat brain, our animal model may be appropriate for exploring the mechanisms involved in the effects of maternal infection on glial cells in the brains of offspring.


Subject(s)
Brain/drug effects , Interleukin-1/biosynthesis , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Base Sequence , Blotting, Western , Brain/embryology , Brain/metabolism , DNA Primers , Female , Fetus/drug effects , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Interleukin-1/genetics , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Necrosis Factor-alpha/genetics
16.
Acta Paediatr ; 88(8): 874-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10503688

ABSTRACT

This prospective study was designed to identify the role of postnatal penicillin prophylaxis in the prevention of neonatal group B streptococcus (GBS) infection. We studied 10 998 infants. Of these, 5389 were in the penicillin prophylaxis group (PP) and 5609 infants did not receive penicillin prophylaxis (NPP). Infants were allocated to treatment by month of birth, alternating 3-mo blocks or 2-mo blocks to the two groups after the first block was randomly assigned. The use of PP reduced the incidence of clinical sepsis (1.7% PP versus 2.5% NPP, p < 0.01), GBS infection (0.4% PP versus 0.9% NPP, p < 0.001) and deaths from sepsis (0.1% PP versus 0.3% NPP, p < 0.05). We conclude that the routine use of postnatal penicillin prophylaxis appears to be effective in reducing the incidence of clinical sepsis and death from sepsis in neonates.


Subject(s)
Antibiotic Prophylaxis , Penicillins/therapeutic use , Streptococcal Infections/prevention & control , Streptococcus agalactiae , Female , Humans , Infant, Newborn , Male , Penicillins/administration & dosage , Pregnancy , Pregnancy, High-Risk , Prospective Studies , Sepsis/mortality , Sepsis/prevention & control , Streptococcal Infections/microbiology , Streptococcus agalactiae/isolation & purification
17.
Neurochem Res ; 24(8): 975-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10478935

ABSTRACT

Phospholipase D (PLD) is emerging as a major player in many novel signaling pathways. Based on recent studies correlating membrane composition with enzyme function, we speculated that feeding of dietary lipids to the newborns has a major impact on brain PLD activity. To test this hypothesis, the rat dams were fed fat-free powder containing either safflower oil or fish oil, and a control powdered chow. The pups were weaned onto the diet and sacrificed at 30 days of age. PLD activity was measured by transphosphatidylation assays using rat brain membranes. This study shows that microsome GTPgammaS-dependent PLD activity in rats fed safflower oil or fish oil was significantly reduced by 38% and 30% respectively compared to controls. Oleate-dependent PLD activity in the safflower oil group, however, was significantly increased by 38%. In contrast, synaptosome membrane (P2) GTPgammaS-dependent PLD activity in rats consuming safflower oil was significantly increased by 29%, but there was no difference in oleate-dependent PLD activity. Likewise, no difference was observed in microsome oleate-dependent PLD and P2 GTPgammaS-dependent PLD activity between the fish oil and the control groups. These results indicate that dietary lipid intake appears to modulate phospholipid metabolism and differential expression of PLD isozymes in the brain.


Subject(s)
Brain/drug effects , Dietary Fats/pharmacology , Phospholipase D/metabolism , Animals , Brain/enzymology , Brain/metabolism , Female , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Microsomes/drug effects , Microsomes/enzymology , Microsomes/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley
18.
Brain Res Bull ; 49(5): 359-65, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10452357

ABSTRACT

Inhibition of nitric oxide synthase (NOS) is known to possibly impair learning and memory. Our previous studies have demonstrated that prenatal hypoxia-ischemia (HI) decreases NOS expression and NOS activity in the neonatal rat brain. To investigate whether effects of prenatal HI on NOS expression continue and whether prenatal HI affects learning and memory in young rats, NOS expression and NOS activity were determined in the hippocampus of rat brains at 28 days of age following a prenatal HI insult on G17. Performances in the passive avoidance test and the Morris water maze test were also studied in these young rats prior to sampling. Rat fetuses were subjected to either a 30-min prenatal HI insult or a sham operation (SH) on gestation day 17 and rat pups were delivered naturally. Increased locomotor activity was observed in the prenatal HI rats as compared to the SH rats on postnatal days 13 and 15, but not on postnatal days 20 and 30. Prenatal HI affected learning ability in these young rats at 28 days of age, as indicated by a delayed acquisition of passive avoidance and by longer escape latency in the Morris water maze test as compared to the SH group. Prenatal HI did not affect retention of passive avoidance and spatial memory. Concomitant with these learning deficits, expression of neuronal NOS and endothelial NOS mRNAs as well as Ca2(+)-dependent NOS activity in the hippocampus of the prenatal HI rat brain were significantly decreased as compared to the SH group. These results suggest that a 30-min prenatal HI insult on gestation day 17 in rats has long-lasting effects on NOS expression and NOS activity in the offspring brain and on learning ability of these young rats. The learning deficit in offspring is possibly associated with the reduction in expression of NOS mRNA and NOS activity in the hippocampus of these animals.


Subject(s)
Brain Ischemia/enzymology , Brain/enzymology , Fetal Diseases/enzymology , Learning Disabilities/etiology , Nitric Oxide Synthase/metabolism , Prenatal Exposure Delayed Effects , Animals , Avoidance Learning/physiology , Brain Ischemia/psychology , Female , Fetal Diseases/psychology , Maze Learning/physiology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type III , Pregnancy , RNA, Messenger/metabolism , Rats , Swimming
19.
Am J Obstet Gynecol ; 180(6 Pt 1): 1349-53, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10368470

ABSTRACT

OBJECTIVE: Our purpose was to evaluate the neonatal adrenal gland by provocative testing in neonates of mothers who had received multiple courses of betamethasone to enhance fetal lung maturity. STUDY DESIGN: Infants of mothers who had received >/=3 courses of betamethasone for fetal lung maturation were enrolled in the study. Twenty-four hours after delivery a baseline serum cortisol concentration was obtained. A synthetic adrenocorticotropic hormone (Cortrosyn) was administered (0.25 mg/1.73 m2). Two hours later a second serum cortisol concentration was obtained. An increase in serum cortisol in response to Cortrosyn was considered a positive test result. Nominal data were compared by means of the Student t test. RESULTS: There were 9 infants enrolled in the study. The mean number of betamethasone treatment cycles was 4.8 +/- 1.09. The mean baseline cortisol level was 2.23 +/- 0.52 microgram/dL, and the mean post-adrenocorticotropic hormone cortisol level was 9.86 +/- 1.70 microgram/dL. All neonates had a positive adrenocorticotropic hormone test result. Stepwise linear regression showed no association between the number of courses of betamethasone treatment cycles and the post-adrenocorticotropic hormone cortisol concentration. CONCLUSION: Multiple weekly treatment cycles of betamethasone for fetal lung maturity administered between 24 and 34 weeks' gestation do not appear to cause adrenal suppression.


Subject(s)
Adrenal Glands/drug effects , Betamethasone/administration & dosage , Betamethasone/adverse effects , Fetal Organ Maturity/drug effects , Lung/embryology , Prenatal Exposure Delayed Effects , Betamethasone/therapeutic use , Cosyntropin , Female , Humans , Hydrocortisone/blood , Infant, Newborn , Pregnancy
20.
Neuroreport ; 10(18): 3927-31, 1999 Dec 16.
Article in English | MEDLINE | ID: mdl-10716235

ABSTRACT

Neuroprotective effects of a Group II metabotropic glutamate receptor agonist, LY379268, were examined in a neonatal rat model of hypoxia-ischemia (unilateral common carotid artery ligation followed by hypoxic exposure for 1.5h in 7-day-old rat pups). LY379268 administered 5 min after hypoxic exposure (2, 5, or 10 mg/kg, i.p.) significantly reduced brain injury as measured by reductions in the ipsilateral brain weight and in CA1 hippocampal neuron density. The significant neuroprotective effects were also observed when this compound (5 mg/kg) was administered 30 min, but not 60 min, after hypoxic exposure. The neonatal hypoxia-ischemia (HI) procedure significantly increased caspase-3 activity and induced DNA fragmentation in the ipsilateral cortex compared with that in the contralateral cortex 24 and 72h after the insult, respectively. LY379268 did not prevent this increase in caspase-3 activity and DNA fragmentation in the ipsilateral cortex. These results suggest that activation of Group II metabotropic glutamate receptors may provide neuroprotection against HI brain injury. However, blockade of caspase-3 activation and the apoptotic pathway appears not to be involved in the neuroprotective effects of LY379268 observed in the neonatal rat model of HI.


Subject(s)
Amino Acids/pharmacology , Animals, Newborn/physiology , Brain Ischemia/pathology , Brain/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Hypoxia/pathology , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Amino Acids/administration & dosage , Animals , Brain/drug effects , Brain/enzymology , Brain Ischemia/enzymology , Brain Ischemia/genetics , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Caspase 3 , Caspases/metabolism , Cell Count/drug effects , DNA Fragmentation , Drug Administration Schedule , Excitatory Amino Acid Agonists/administration & dosage , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Hypoxia/enzymology , Hypoxia/genetics , Neurons/pathology , Neuroprotective Agents/administration & dosage , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...