Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Mol Sci ; 17(3): 289, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26927081

RESUMO

The hematopoietic growth factor erythropoietin (EPO) has been shown to be neuroprotective against hypoxia-ischemia (HI) in Postnatal Day 7 (P7)-P10 or adult animal models. The current study was aimed to determine whether EPO also provides long-lasting neuroprotection against HI in P5 rats, which is relevant to immature human infants. Sprague-Dawley rats at P5 were subjected to right common carotid artery ligation followed by an exposure to 6% oxygen with balanced nitrogen for 1.5 h. Human recombinant EPO (rEPO, at a dose of 5 units/g) was administered intraperitoneally one hour before or immediately after insult, followed by additional injections at 24 and 48 h post-insult. The control rats were injected with normal saline following HI. Neurobehavioral tests were performed on P8 and P20, and brain injury was examined on P21. HI insult significantly impaired neurobehavioral performance including sensorimotor, locomotor activity and cognitive ability on the P8 and P20 rats. HI insult also resulted in brain inflammation (as indicated by microglia activation) and neuronal death (as indicated by Jade B positive staining) in the white matter, striatum, cortex, and hippocampal areas of the P21 rat. Both pre- and post-treatment with rEPO significantly improved neurobehavioral performance and protected against the HI-induced neuronal death, microglia activation (OX42+) as well as loss of mature oligodendrocytes (APC-CC1+) and hippocampal neurons (Nissl+). The long-lasting protective effects of rEPO in the neonatal rat HI model suggest that to exert neurotrophic activity in the brain might be an effective approach for therapeutic treatment of neonatal brain injury induced by hypoxia-ischemia.


Assuntos
Eritropoetina/uso terapêutico , Hipocampo/fisiopatologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Transtornos Motores/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Eritropoetina/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Humanos , Hipóxia-Isquemia Encefálica/complicações , Locomoção , Transtornos Motores/etiologia , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley
2.
Pediatr Res ; 71(6): 645-52, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22337231

RESUMO

INTRODUCTION: Intrauterine growth restriction (IUGR) alters fetal development and is associated with neurodevelopmental abnormalities. We hypothesized that growth restriction from reduced intrauterine perfusion would predispose neonatal rats to subsequent inflammatory brain injury. METHODS: In this study, IUGR was achieved by induced placental insufficiency in pregnant rats at 14 days of gestation. IUGR offspring and sham-operated control pups were subsequently injected with intracerebral lipopolysaccharide (LPS) as a model of periventricular leukomalacia (PVL). RESULTS: LPS similarly elevates proinflammatory cytokines in the brains of both IUGR and control rat pups. However, the chemokines cytokine-induced neutrophil chemoattractant-1 (CINC-1) and macrophage chemoattractant protein-1 (MCP-1), as well as microglia activation, were significantly higher in LPS-treated IUGR rat pups as compared with LPS-treated controls. In addition to the unique brain inflammatory response, IUGR rat pups demonstrated increased brain damage with an increased number of apoptotic cells, larger lateral ventricular size, and more severe impairment of myelination. DISCUSSION: This study provides evidence that placental insufficiency may sensitize the innate immune system in the immature brain and reveals a possible link between brain inflammation and injury.


Assuntos
Animais Recém-Nascidos/metabolismo , Encefalomalacia/patologia , Retardo do Crescimento Fetal/patologia , Lipopolissacarídeos/efeitos adversos , Animais , Apoptose , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Modelos Animais de Doenças , Encefalomalacia/induzido quimicamente , Encefalomalacia/metabolismo , Feminino , Retardo do Crescimento Fetal/metabolismo , Humanos , Recém-Nascido , Injeções Intraventriculares , Leucomalácia Periventricular/induzido quimicamente , Leucomalácia Periventricular/metabolismo , Leucomalácia Periventricular/patologia , Lipopolissacarídeos/administração & dosagem , Insuficiência Placentária/metabolismo , Insuficiência Placentária/patologia , Gravidez , Ratos , Ratos Sprague-Dawley
3.
Pediatr Res ; 71(5): 552-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22314662

RESUMO

INTRODUCTION: The aim of this study was to test whether dexamethasone (Dex) and betamethasone (Beta), two of the most commonly used corticosteroids, protect against lipopolysaccharide (LPS)-induced white matter damage and neurobehavioral dysfunction. METHODS: LPS or sterile saline was injected into the brain white matter of rat pups at postnatal day 5 (P5), and Dex or Beta was given intraperitoneally to the rat pups 1 h before the LPS microinjection. Brain inflammatory response, brain damage, and myelination were examined at P6, P8, and P14. Neurobehavioral tests were performed from P3 through P22. RESULTS: Our results demonstrate that Dex and Beta markedly diminish the LPS-induced brain inflammatory response, restore myelin basic protein (MBP) expression, and alleviate lateral ventricle dilation. Both corticosteroids demonstrate significant protection against most LPS-induced behavioral deficits, including those in rearing, vibrissa-elicited forelimb-placing, beam walking, learning, and elevated plus-maze test. Of note, only Beta improved the locomotion and stereotype dysfunction. In contrast to their beneficial effects, neither drug prevented LPS-induced delay in body weight gain from P6 through P21. DISCUSSION: Our study suggests that if their adverse effects are minimized, corticosteroids may be the potential candidate drugs to prevent brain damage in premature infants.


Assuntos
Betametasona/farmacologia , Lesões Encefálicas/prevenção & controle , Dexametasona/farmacologia , Lipopolissacarídeos/toxicidade , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/patologia , Imuno-Histoquímica , Inflamação/patologia , Ratos , Ratos Sprague-Dawley , Redução de Peso/efeitos dos fármacos
4.
Neurobiol Dis ; 44(3): 304-16, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21798348

RESUMO

Brain inflammation in early life has been proposed to play important roles in the development of neurodegenerative disorders in adult life. To test this hypothesis, we used a neonatal rat model of lipopolysaccharide (LPS) exposure (1000 EU/g body weight, intracerebral injection on P5) to produce brain inflammation. By P70, when LPS-induced behavioral deficits were spontaneously recovered, animals were challenged with rotenone, a commonly used pesticide, through subcutaneous mini-pump infusion at a dose of 1.25 mg/kg per day for 14 days. This rotenone treatment regimen ordinarily does not produce toxic effects on behaviors in normal adult rats. Our results show that neonatal LPS exposure enhanced the vulnerability of nigrostriatal dopaminergic neurons to rotenone neurotoxicity in later life. Rotenone treatment resulted in motor neurobehavioral impairments in rats with the neonatal LPS exposure, but not in those without the neonatal LPS exposure. Rotenone induced losses of tyrosine hydroxylase immunoreactive neurons in the substantia nigra and decreased mitochondrial complex I activity in the striatum of rats with neonatal LPS exposure, but not in those without this exposure. Neonatal LPS exposure with later exposure to rotenone decreased retrogradely labeled nigrostriatal dopaminergic projecting neurons. The current study suggests that perinatal brain inflammation may enhance adult susceptibility to the development of neurodegenerative disorders triggered later on by environmental toxins at an ordinarily non-toxic or sub-toxic dose. Our model may be useful for studying mechanisms involved in the pathogenesis of nonfamilial Parkinson's disease and the development of potential therapeutic treatments.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Inseticidas/toxicidade , Lipopolissacarídeos/farmacologia , Rotenona/toxicidade , Substância Negra/citologia , Fatores Etários , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Neurônios Dopaminérgicos/ultraestrutura , Sinergismo Farmacológico , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Infusões Subcutâneas/métodos , Masculino , Proteínas dos Microfilamentos/metabolismo , Microscopia Eletrônica de Transmissão , Atividade Motora/efeitos dos fármacos , Movimento/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Gravidez , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Comportamento Estereotipado/efeitos dos fármacos , Vibrissas/inervação
5.
Brain Behav Immun ; 25(2): 286-97, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20875849

RESUMO

Our previous studies have shown that neonatal exposure to lipopolysaccharide (LPS) resulted in motor dysfunction and dopaminergic neuronal injury in the juvenile rat brain. To further examine whether neonatal LPS exposure has persisting effects in adult rats, motor behaviors were examined from postnatal day 7 (P7) to P70 and brain injury was determined in P70 rats following an intracerebral injection of LPS (1 mg/kg) in P5 Sprague-Dawley male rats. Although neonatal LPS exposure resulted in hyperactivity in locomotion and stereotyped tasks, and other disturbances of motor behaviors, the impaired motor functions were spontaneously recovered by P70. On the other hand, neonatal LPS-induced injury to the dopaminergic system such as the loss of dendrites and reduced tyrosine hydroxylase immunoreactivity in the substantia nigra persisted in P70 rats. Neonatal LPS exposure also resulted in sustained inflammatory responses in the P70 rat brain, as indicated by an increased number of activated microglia and elevation of interleukin-1ß and interleukin-6 content in the rat brain. In addition, when challenged with methamphetamine (METH, 0.5 mg/kg) subcutaneously, rats with neonatal LPS exposure had significantly increased responses in METH-induced locomotion and stereotypy behaviors as compared to those without LPS exposure. These results indicate that although neonatal LPS-induced neurobehavioral impairment is spontaneously recoverable, the LPS exposure-induced persistent injury to the dopaminergic system and the chronic inflammation may represent the existence of silent neurotoxicity. Our data further suggest that the compromised dendritic mitochondrial function might contribute, at least partially, to the silent neurotoxicity.


Assuntos
Animais Recém-Nascidos/fisiologia , Encéfalo/patologia , Dopamina/fisiologia , Lipopolissacarídeos/farmacologia , Neurônios/patologia , Síndromes Neurotóxicas/patologia , Animais , Comportamento Animal/fisiologia , Estimulantes do Sistema Nervoso Central , Citocinas/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Membro Anterior/fisiologia , Imuno-Histoquímica , Metanfetamina , Atividade Motora/efeitos dos fármacos , Destreza Motora/fisiologia , Estimulação Física , Gravidez , Ratos , Ratos Sprague-Dawley , Comportamento Estereotipado/efeitos dos fármacos , Vibrissas/inervação , Vibrissas/fisiologia
6.
Pediatr Res ; 67(6): 579-84, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20220546

RESUMO

Periventricular leukomalacia (PVL) is a major form of brain damage in premature infants. This study was to test whether IGF-1 can prevent PVL-like brain damage induced by lipopolysaccharide (LPS) in the neonatal rat. Intraventricular delivery of LPS resulted in an acute brain inflammatory response, i.e., rapid recruitment of polymorphonuclear leukocytes (PMNs), activation of microglia and astrocytes, and induction of IL-1beta (IL1beta) expression. Brain inflammation was associated with the loss of O4+ preoligodendrocytes (preOLs), a decrease of myelin basic protein (MBP) in the white matter and an increase of pyknotic cells in the cortex. IGF-1 at a low dose significantly prevented LPS-induced deleterious effects without alteration of IL-1beta expression and microglia/astrocytes activation. On the other hand, the low dose of IGF-1 enhanced LPS-induced PMNs recruitment and blood-brain barrier (BBB) permeability, and caused intracerebral hemorrhage. At higher doses, co-application of IGF-1 with LPS resulted in a high mortality rate. Brains from the surviving rats showed massive PMN infiltration and intracerebral hemorrhage. However, these adverse effects were not found in rats treated with IGF-1 alone. This study provides the alarming evidence that in an acute inflammatory condition, IGF-1 may have severe, harmful effects on the developing brain.


Assuntos
Encéfalo/efeitos dos fármacos , Encefalite/prevenção & controle , Fator de Crescimento Insulin-Like I/administração & dosagem , Leucomalácia Periventricular/prevenção & controle , Fármacos Neuroprotetores/administração & dosagem , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/patologia , Permeabilidade Capilar/efeitos dos fármacos , Morte Celular , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/patologia , Hemorragia Cerebral/fisiopatologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalite/induzido quimicamente , Encefalite/metabolismo , Encefalite/patologia , Encefalite/fisiopatologia , Feminino , Humanos , Recém-Nascido , Mediadores da Inflamação/metabolismo , Injeções Intraventriculares , Fator de Crescimento Insulin-Like I/toxicidade , Interleucina-1beta/metabolismo , Leucomalácia Periventricular/induzido quimicamente , Leucomalácia Periventricular/metabolismo , Leucomalácia Periventricular/patologia , Leucomalácia Periventricular/fisiopatologia , Lipopolissacarídeos , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Fármacos Neuroprotetores/toxicidade , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem
7.
Eur J Neurosci ; 27(6): 1475-84, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18364024

RESUMO

Neonatal exposure to infectious agents may result in long-term neurological disability, and is particularly associated with the subsequent development of motor and cognitive disturbances. Our previous studies have shown that treatment with alpha-phenyl-n-tert-butyl-nitrone (PBN) following exposure to lipopolysaccharide (LPS) reduces LPS-induced brain injury in the neonatal rat. To examine whether PBN has long-lasting protective effects and ameliorates LPS-induced motor and cognitive dysfunction, PBN (100 mg/kg) was administered intraperitoneally 5 min after an LPS (1 mg/kg) intracerebral injection in postnatal day 5 (P5) Sprague-Dawley rat pups. Neurobehavioral tests were carried out from P3 to P21, and brain injury was examined at 24 h and 16 days after LPS injection. Neonatal LPS exposure resulted in hyperactivity from P13 to P17 in the open field task as compared with the control rat. Neurobehavioral deficits that were still observable at P21 included dysfunction in the beam-walking and pole tests, learning and memory deficits in the passive avoidance task, and less anxiety-like response in the elevated plus-maze task. These behavioral findings were matched by LPS-induced axonal injury in the CA1 region of the middle dorsal hippocampus (HP), reduction in the size of the HP and the number of neurons in the CA1 region of the middle dorsal HP, and loss of tyrosine hydroxylase immunoreactivity in neurons in the substantia nigra and ventral tegmental areas. Treatment with PBN provided long-lasting protection against the LPS-induced axonal injury and neuronal loss, and improved the associated neurological dysfunctions in juvenile rats.


Assuntos
Lesões Encefálicas/patologia , Lesões Encefálicas/prevenção & controle , Óxidos N-Cíclicos/uso terapêutico , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Aprendizagem/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Fármacos Neuroprotetores/uso terapêutico , Animais , Animais Recém-Nascidos , Anti-Inflamatórios não Esteroides/uso terapêutico , Lesões Encefálicas/induzido quimicamente , Feminino , Aprendizagem/fisiologia , Masculino , Memória/efeitos dos fármacos , Memória/fisiologia , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley
8.
J Neurosci Res ; 86(16): 3536-47, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18683243

RESUMO

Our previous study showed that treatment with alpha-phenyl-n-tert-butyl-nitrone (PBN) after exposure to lipopolysaccharide (LPS) reduced LPS-induced white matter injury in the neonatal rat brain. The object of the current study was to further examine whether PBN has long-lasting protective effects and ameliorates LPS-induced neurological dysfunction. Intracerebral (i.c.) injection of LPS (1 mg/kg) was performed in postnatal day (P) 5 Sprague Dawley rat pups and PBN (100 mg/kg) or saline was administered intraperitoneally 5 min after LPS injection. The control rats were injected (i.c.) with sterile saline. Neurobehavioral tests were carried out from P3 to P21, and brain injury was examined after these tests. LPS exposure resulted in severe brain damage, including enlargement of ventricles bilaterally, loss of mature oligodendrocytes, impaired myelination as indicated by the decrease in myelin basic protein immunostaining, and alterations in dendritic processes in the cortical gray matter of the parietal cortex. Electron microscopic examination showed that LPS exposure caused impaired myelination as indicated by the disintegrated myelin sheaths in the juvenile rat brain. LPS administration also significantly affected neurobehavioral functions such as performance in righting reflex, wire hanging maneuver, cliff avoidance, negative geotaxis, vibrissa-elicited forelimb-placing test, beam walking, and gait test. Treatment with PBN, a free radical scavenger and antioxidant, provided protection against LPS-induced brain injury and associated neurological dysfunction in juvenile rats, suggesting that antioxidation might be an effective approach for therapeutic treatment of neonatal brain injury induced by infection/inflammation.


Assuntos
Dano Encefálico Crônico/tratamento farmacológico , Infecções Bacterianas do Sistema Nervoso Central/transmissão , Óxidos N-Cíclicos/uso terapêutico , Transmissão Vertical de Doenças Infecciosas , Leucomalácia Periventricular/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Dano Encefálico Crônico/induzido quimicamente , Dano Encefálico Crônico/microbiologia , Infecções Bacterianas do Sistema Nervoso Central/microbiologia , Infecções Bacterianas do Sistema Nervoso Central/fisiopatologia , Modelos Animais de Doenças , Feminino , Transtornos Neurológicos da Marcha/induzido quimicamente , Transtornos Neurológicos da Marcha/tratamento farmacológico , Transtornos Neurológicos da Marcha/microbiologia , Humanos , Recém-Nascido , Leucomalácia Periventricular/microbiologia , Lipopolissacarídeos/toxicidade , Masculino , Transtornos dos Movimentos/tratamento farmacológico , Transtornos dos Movimentos/microbiologia , Transtornos dos Movimentos/fisiopatologia , Proteína Básica da Mielina/efeitos dos fármacos , Proteína Básica da Mielina/metabolismo , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Gravidez , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Reflexo/efeitos dos fármacos , Reflexo/fisiologia
9.
Pediatr Res ; 64(4): 370-4, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18535483

RESUMO

Vascular Endothelial Growth Factor (VEGF) protects the brain against ischemic injury in adult animals. We evaluated whether VEGF has neuroprotective effects against hypoxic-ischemic (HI) brain injury in newborn rats. Seven-day-old rat pups had the right carotid artery permanently ligated followed by 140 min of hypoxia (8% oxygen). VEGF (5, 10, 20, or 40 ng) or vehicle was administered intracerebroventricularly 5 min after reoxygenation following HI. Brain damage was evaluated by weight loss of the right hemisphere at 22 d after HI and by gross and microscopic morphology. Body weight, rectal temperature, and mortality were not significantly different in the VEGF and vehicle treated groups. VEGF treatment increased brain VEGF levels at 15 min after injection. VEGF (10 and 20 ng) significantly reduced brain weight loss (p < 0.05) and gross brain injury (p < 0.05); however, treatment with 5 or 40 ng did not. VEGF (10 ng) also decreased brain damage assessed by histologic scoring. VEGF increased phosphorylation of protein kinase B (Akt) and extracellular-signal regulated kinase 1/2 (ERK1/2) in the cortex (p < 0.05). These results suggest that VEGF has neuroprotective effects in the neonatal rat HI model that may be related to activation of the Akt/ERK signaling pathway.


Assuntos
Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Animais Recém-Nascidos , Western Blotting , Temperatura Corporal , Peso Corporal , Artérias Carótidas/cirurgia , Hipóxia-Isquemia Encefálica/patologia , Ligadura , Ratos , Estatísticas não Paramétricas , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Neurosci Lett ; 405(1-2): 52-6, 2006 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-16876321

RESUMO

Our previous study has demonstrated that alpha-phenyl-tert-butyl-nitrone (PBN) provided neuroprotection to the neonatal white matter following cerebral hypoxia-ischemia (HI). Free radical scavenging was involved in the neuroprotection of PBN. To investigate if other mechanisms contribute to the neuroprotection of PBN, postnatal day 4 SD rats were subjected to bilateral common carotid artery ligation, followed by 8% oxygen exposure for 20min. A single dose of PBN (100mg/kg, i.p.) was given prior to the hypoxic exposure. Expression of inflammatory cytokines: interleukin-1beta (IL-1beta), inducible nitric oxide synthase (iNOS) and tumor necrosis factor-alpha (TNF-alpha) was determined by RT-PCR, ELISA and immunohistochemistry. Activation of transcriptional factor nuclear factor-kappa B (NF-kappaB) was measured by ELISA. PBN significantly inhibited HI-induced up-regulation of IL-1beta, TNF-alpha and iNOS mRNA expression at 4h following HI. PBN treatment also reduced the brain concentration of IL-1beta significantly and decreased the number of IL-1beta- or iNOS-expressing cells in the white matter area at 12h following HI. Moreover, PBN suppressed the HI-induced NF-kappaB activation at 1h after HI. The overall results indicate that besides free radical scavenging, anti-inflammation might partly contribute to the neuroprotection afforded by PBN on neonatal white matter following cerebral HI.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Encéfalo/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Hipóxia-Isquemia Encefálica/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Óxidos de Nitrogênio/farmacologia , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Óxidos N-Cíclicos , Ensaio de Imunoadsorção Enzimática , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Imuno-Histoquímica , Interleucina-1/biossíntese , Interleucina-1/genética , NF-kappa B/biossíntese , NF-kappa B/genética , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , RNA Mensageiro/biossíntese , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
11.
Behav Brain Res ; 165(1): 80-90, 2005 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-16140403

RESUMO

Bilateral carotid artery occlusion (BCAO) followed by exposure to a hypoxic condition (8% oxygen for 10 or 15 min) was performed in postnatal day 4 SD rats. Brain injury and myelination changes were examined on postnatal day 21 (P21) and tests for neurobehavioral toxicity were performed from P3 to P21. BCAO followed by 10 or 15 min hypoxic insult resulted in mild and severe, respectively, brain injury, reduction in mature oligodendrocytes and tyrosine hydroxylase positive neurons and impaired myelination as indicated by decreased myelin basic protein immunostaining in the P21 rat brain. Hypoxia-ischemia also affected physical development (body weight gain and eye opening) and neurobehavioral performance, such as righting reflex, wire hanging maneuver, cliff avoidance, locomotor activity, gait analysis, responses in the elevated plus-maze and passive avoidance. BCAO followed by 15 min of hypoxia caused more severely impaired neurobehavioral performance as compared with BCAO followed by 10 min of hypoxia in the rat. The overall results demonstrate that hypoxia-ischemia-induced brain injury not only persists, but also is linked with neurobehavioral deficits in juvenile rats. The present data also indicate that the degree of brain injury and the deficits of neurobehavioral performance in the rat are dependent on the hypoxic-ischemic condition, i.e., the exposure time to hypoxia.


Assuntos
Comportamento Animal/fisiologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Leucomalácia Periventricular/fisiopatologia , Bainha de Mielina/patologia , Neurônios/patologia , Oligodendroglia/patologia , Animais , Animais Recém-Nascidos , Peso Corporal , Lesão Encefálica Crônica/complicações , Lesão Encefálica Crônica/patologia , Lesão Encefálica Crônica/fisiopatologia , Modelos Animais de Doenças , Humanos , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/patologia , Recém-Nascido , Leucomalácia Periventricular/etiologia , Leucomalácia Periventricular/patologia , Atividade Motora , Destreza Motora/fisiologia , Neurônios/enzimologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
12.
Brain Res ; 1063(1): 15-26, 2005 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-16259966

RESUMO

To investigate if insulin-like growth factor-1 (IGF-1) provides neuroprotection to oligodendrocyte progenitor cells (OPCs) following cerebral hypoxia-ischemia, a previously developed neonatal rat model of white matter damage was used in this study. Postnatal day 4 (P4) SD rat pups were subjected to bilateral common carotid artery ligation, followed by exposure to 8% oxygen for 10 min. IGF-1 (0.5 microg) or vehicle was injected into the left ventricle after artery ligation and before the hypoxic exposure. Cerebral hypoxia-ischemia caused death of O4+ late OPCs in the P5 rat brain and impaired myelination in the P9 and P21 rat brain. Caspase-3 activation was involved in the death of OPCs. Moreover, cerebral hypoxia-ischemia impaired neurobehavioral performance in juvenile rats. IGF-1 treatment attenuated damages to OPCs and improved neurological functions after cerebral hypoxia-ischemia. It reduced death of O4+ OPCs by 39% on P5 and enhanced myelination on P9 and P21. Bromodeoxyuridine uptake assay showed that cerebral hypoxia-ischemia inhibited proliferation of stem/progenitor cells in the subventricular zone and NG2+ early OPCs in the white matter area. IGF-1 treatment increased cell proliferation in the subventricular zone by 31% 1 day following hypoxic-ischemic insult. Proliferation of early and late OPCs in the IGF-1-treated group was 1.5- and 2.4-fold of that in the vehicle-treated group, respectively. In conclusion, IGF-1 provided potent neuroprotection to OPCs and improved neurological functions following cerebral hypoxia-ischemia in the neonatal rat. The neuroprotection of IGF-1 was associated with its antiapoptotic and mitogenic effects.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fármacos Neuroprotetores/metabolismo , Oligodendroglia/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Injeções Intraventriculares , Fator de Crescimento Insulin-Like I/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Oligodendroglia/patologia , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Células-Tronco/patologia
13.
Brain Res Dev Brain Res ; 157(2): 141-9, 2005 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-15878785

RESUMO

White matter damage (WMD) is an important cause of disability including cerebral palsy in preterm, low birth-weight infants. Maternal infection is now recognized as one of the risk factors for WMD. Previously we reported that intrauterine inoculation of Escherichia coli to pregnant rats resulted in WMD in offspring and interleukin-10 (IL-10) was protective against this damage. The objective of this study was to elucidate the mechanism involved in the protective effect of IL-10 against neonatal WMD. We found that E. coli treatment in dams resulted in significant apoptosis in periventricular white matter of rat pups on postnatal day 0 (P0). On P8, a remarkable increase in ED-1 immunostaining (indicating either microglial activation or macrophage infiltration) was detected in brains of pups in the E. coli-treated group. Astrogliosis was also noticed in brain white matter of pups in the E. coli-treated group. In addition to the strong activation of microglia and astrocytes, oligodendrocytes (OLs) were significantly reduced in periventricular areas in the brains of pups from the E. coli-treated group. Later, on P15, hypomyelination was also noticed in rat brains from the E. coli-treated group, using myelin basic protein (MBP) immunostaining. Treatment with IL-10 after E. coli inoculation significantly reduced TUNEL staining and caspase-3 activation, and partially restored the impaired immunostaining markers for immature and mature OLs, such as CNPase, O4, adenomatous polyposis coli (APC) and MBP. These results indicate that the protective effect of IL-10 against brain WMD is linked with suppression of microglial activation/macrophage infiltration, as shown by significantly reduced ED-1+ cells in the white matter.


Assuntos
Infecções Bacterianas/complicações , Dano Encefálico Crônico/fisiopatologia , Gliose/fisiopatologia , Interleucina-10/imunologia , Exposição Materna , Efeitos Tardios da Exposição Pré-Natal , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Infecções Bacterianas/imunologia , Biomarcadores , Dano Encefálico Crônico/imunologia , Dano Encefálico Crônico/prevenção & controle , Paralisia Cerebral/imunologia , Paralisia Cerebral/fisiopatologia , Paralisia Cerebral/prevenção & controle , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/imunologia , Modelos Animais de Doenças , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/imunologia , Feminino , Gliose/etiologia , Gliose/prevenção & controle , Interleucina-10/farmacologia , Microglia/efeitos dos fármacos , Microglia/imunologia , Degeneração Neural/imunologia , Degeneração Neural/fisiopatologia , Degeneração Neural/prevenção & controle , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/imunologia , Fibras Nervosas Mielinizadas/patologia , Fármacos Neuroprotetores/imunologia , Fármacos Neuroprotetores/farmacologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/imunologia , Gravidez , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
14.
Brain Res Dev Brain Res ; 156(1): 87-92, 2005 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-15862631

RESUMO

Focal cerebral hypoxia-ischemia due to isolated vascular insufficiency is well known to cause ipsilateral, but not contralateral, cerebral apoptosis. Hypoxic-ischemic damage to the cerebellum and brainstem in such a model has not been established. This experimental rodent study demonstrates, through deoxyribonucleic acid fragmentation and terminal deoxynucleotidyl transferase-mediated deoxyuridine 5'-triphosphate-digoxigenin nick end labeling analysis, that neuronal cells in these infratentorial regions also suffer mild apoptosis and necrosis after focal cerebral hypoxic-ischemic injury in the newborn rat. These data provide additional insight into the mechanisms of neurological injury in the cerebellum and brainstem areas resulting from a focal cerebral hypoxic-ischemic insult and demonstrate that future therapeutic interventions for hypoxic-ischemic encephalopathy system should deal with the entire central nervous system.


Assuntos
Apoptose/fisiologia , Tronco Encefálico/patologia , Cerebelo/patologia , Hipóxia-Isquemia Encefálica/patologia , Animais , Animais Recém-Nascidos , Southern Blotting/métodos , Tronco Encefálico/crescimento & desenvolvimento , Tronco Encefálico/fisiopatologia , Cerebelo/crescimento & desenvolvimento , Cerebelo/fisiopatologia , Fragmentação do DNA/fisiologia , Feminino , Hipóxia-Isquemia Encefálica/fisiopatologia , Marcação In Situ das Extremidades Cortadas/métodos , Masculino , Necrose , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
15.
Brain Res ; 1007(1-2): 132-41, 2004 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-15064144

RESUMO

White matter of the neonatal brain is highly sensitive to hypoxic-ischemic insult. The susceptibility of premature oligodendrocytes (OLs) to free radicals (FRs) produced during hypoxia-ischemia (HI) has been proposed as one of the mechanisms involved. To test this hypothesis, and to further investigate if the FR scavenger alpha-phenyl-N-tert-butyl-nitrone (PBN) attenuates hypoxic-ischemic white matter damage (WMD), postnatal day 4 (P4) SD rats were subjected to bilateral common carotid artery ligation (BCAL), followed by 8% oxygen exposure for 20 min. Pathological changes were evaluated on P6 and P9, 2 and 5 days after the HI insult. HI caused severe WMD including rarefaction, necrosis and cavity formation in the corpus callosum, external and internal capsule areas. OL injury was evidenced by degeneration of O4 positive OLs on P6. Disrupted myelination was verified by decreased immunostaining of myelin basic protein (MBP) on P9. Axonal injury was demonstrated by increased amyloid precursor protein (APP) immunostaining on both P6 and P9. Two lipid peroxidation end products, malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), showed a one-fold elevation within 1-24 h following HI. 4-HNE immunostaining was found to specifically localize in the white matter area. Furthermore, pyknotic O4+ OLs were double-labeled with 4-HNE. These findings suggest that FRs are involved in the pathogenesis of neonatal WMD. PBN (100 mg/kg, i.p.) treatment alleviated the pathological changes of WMD following HI. It improved the survival of O4 positive OLs, attenuated hypomyelination and reduced axonal damage. PBN treatment also decreased the brain concentration of MDA/4-HNE and positive 4-HNE staining in the white matter area. These findings indicate that in the current WMD model, PBN protects both OLs and axons, the two main components in the white matter, from neonatal HI insult. FR scavenging appears to be the primary mechanism underlying its neuroprotective effect.


Assuntos
Animais Recém-Nascidos , Sequestradores de Radicais Livres/uso terapêutico , Hipóxia-Isquemia Encefálica/prevenção & controle , Óxidos de Nitrogênio/uso terapêutico , Fatores Etários , Aldeídos/metabolismo , Peptídeos beta-Amiloides/metabolismo , Análise de Variância , Animais , Antígenos de Diferenciação/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Contagem de Células/métodos , Sobrevivência Celular/efeitos dos fármacos , Óxidos N-Cíclicos , Hipóxia-Isquemia Encefálica/etiologia , Hipóxia-Isquemia Encefálica/patologia , Imuno-Histoquímica/métodos , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Malondialdeído/metabolismo , Proteína Básica da Mielina/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Ratos , Coloração e Rotulagem/métodos , Fatores de Tempo
16.
Brain Res ; 975(1-2): 37-47, 2003 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-12763591

RESUMO

Increasing data provide support for the hypothesis that inflammatory cytokines mediate inflammation-induced injury to developing white matter. In the present study, roles of tumor necrosis factor-alpha (TNFalpha) and interleukin-1 beta (IL-1beta) in mediating lipopolysaccharide (LPS)-induced brain injury were investigated by co-administration of LPS with IL-1 receptor antagonist (IL-1ra) or TNFalpha antibody in the 5-day-old rat brain. Intracerebral injection of LPS and other agents was performed in a stereotaxic apparatus at the location of 1.0 mm posterior and 1.0 mm lateral to the bregma, and 2.0 mm deep to the skull surface at the left hemisphere. Brain injury was examined in brain sections 3 and 11 days after LPS injection. LPS-induced inflammatory responses were evidenced by great increases in TNFalpha and IL-1beta concentrations in the neonatal rat brain 6 h after LPS injection. White matter rarefaction was observed in 71% (five out of seven) of the rat brains 3 days after LPS injection and bilateral ventricle dilation was found in 71% (five out of seven) of the P8 rat brains and in 100% of the P16 rat brains (four out of four). These alterations were not found in the control rat brains. No apparent histological changes in gray matter were observed in the LPS-injected rat brains. LPS injection also resulted in injuries to oligodendrocytes (OLs) and hypomyelination, as indicated by reduced immunostaining for O4 and myelin basic protein (MBP). Increased astrogliosis, as indicated by increased glial fibrillary acidic protein (GFAP) immunostaining, was also observed in the LPS-injected, but not the control rat brain. Co-administration of LPS with IL-1ra, but not with TNFalpha antibody, significantly attenuated LPS-induced white matter injury, as indicated by decreases in ventricle dilation, white matter rarefaction, GFAP positive staining and by improved O4 and MBP immunostaining. Co-administration of LPS with IL-1ra significantly reduced LPS-induced elevation of caspase-3 activity in the rat brain. While TNFalpha antibody had no effect on LPS-induced elevation of caspase-3 activity, co-administration of LPS with TNFalpha antibody partially, but significantly, decreased LPS-stimulated increase in IL-1beta in the neonatal rat brain. These data suggest that IL-1beta may play an important role in mediating LPS-induced brain injury and TNFalpha may have complicated, probably dual, effects in LPS-induced brain injury.


Assuntos
Animais Recém-Nascidos/fisiologia , Encefalite/induzido quimicamente , Encefalite/patologia , Interleucina-1/fisiologia , Lipopolissacarídeos/toxicidade , Fator de Necrose Tumoral alfa/fisiologia , Animais , Astrócitos/patologia , Caspase 3 , Caspases/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Injeções Intraventriculares , Interleucina-1/antagonistas & inibidores , Interleucina-1/imunologia , Lipopolissacarídeos/administração & dosagem , Masculino , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/patologia , Oligodendroglia/patologia , Ratos , Ratos Sprague-Dawley , Técnicas Estereotáxicas , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/imunologia
17.
Eur J Pharmacol ; 437(3): 139-45, 2002 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-11890901

RESUMO

Neuroprotective effects of N-acetylaspartylglutamate (NAAG), the precursor of glutamate and a selective agonist at the Group II metabotropic glutamate (mGlu) receptor, against hypoxic-ischemic brain injury were examined in a neonatal rat model of cerebral hypoxia-ischemia. The neonatal hypoxia-ischemia procedure (unilateral carotid artery ligation followed by exposure to an 8% oxygen hypoxic condition for 1.5 h) was performed in 7-day-old rat pups. Following unilateral carotid artery ligation, NAAG (0.5 to 20 mg/kg, i.p.) was administered before or after the hypoxic exposure. Brain injury was examined 1-week later by weight reduction in the ipsilateral brain and by neuron density in the hippocampal CA1 area. In the saline-treated rat, neonatal hypoxia-ischemia resulted in severe brain injury as indicated by a 24% reduction in the ipsilateral brain weight. Low doses of NAAG (2-10 mg/kg, but not 0.5 mg/kg), administered before or even if 1 h after the hypoxic exposure, greatly reduced hypoxia-ischemia-induced brain injury (3.8-14.2% reduction in the ipsilateral brain weight). A high dose of NAAG (20 mg/kg) was ineffective. While L(+)-2-Amino-4-phosphonobutyric acid (L-AP4) and trans-[1S,3R]-1-Amino-cyclopentane-1, 3-dicarboxylic acid (t-ACPD) were unable to provide protection against hypoxic-ischemic brain injury, 2-(phosphonomethyl) pentanedioic acid (2-PMPA), an inhibitor of N-acetylated alpha-linked acidic dipeptidase (NAALADase), which hydrolyzes endogenous NAAG into N-acetyl-aspartate and glutamate, significantly reduced neonatal hypoxia-ischemia-induced brain injury. (alphaS)-alpha-Amino-alpha-[(1S, 2S)-2-carboxycyclopropyl]-9H-xanthine-9-propanoic acid (LY341495), a selective antagonist at the mGlu2/3 receptor, prevented the neuroprotective effect of NAAG. Neuron density data measured in the hippocampal CA1 area confirmed that ipsilateral brain weight reduction was a valid measure for hypoxic-ischemic brain injury. Neonatal hypoxia-ischemia stimulated an elevation of cyclic AMP (cAMP) concentration in the saline-treated rat brain. NAAG, L-AP4 and t-ACPD all significantly decreased hypoxia-ischemia-induced elevation of cAMP. LY341495 blocked the effect of NAAG, but not of L-AP4 or t-ACPD, on hypoxia-ischemia-stimulated cAMP elevation. The overall results suggest that the neuroprotective effect of NAAG is largely associated with activation of mGlu2/3 receptor.


Assuntos
Dipeptídeos/farmacologia , Hipóxia-Isquemia Encefálica/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Aminoácidos/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Carboxipeptidases/antagonistas & inibidores , AMP Cíclico/metabolismo , Cicloleucina/análogos & derivados , Cicloleucina/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glutamato Carboxipeptidase II , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Compostos Organofosforados/farmacologia , Propionatos/farmacologia , Ratos , Ratos Sprague-Dawley , Xantenos/farmacologia
18.
Brain Res Dev Brain Res ; 140(2): 205-14, 2003 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-12586426

RESUMO

Increasing data provide support for the hypothesis that brain inflammation plays an important role in injury to developing white matter. In the present study, inflammatory responses in the neonatal rat brain were investigated following lipopolysaccharide (LPS) administration at postnatal day 5. LPS-induced brain injury was examined in brain sections 24 h, 3 and 9 days after LPS injection. White matter rarefaction was observed in 50% of the rat brains (three out of six) 24 h after LPS injection. Lateral ventricle enlargement was found in 100% (four out of four) and 89% (eight out of nine) of rat brains 3 and 9 days after LPS administration, respectively. White matter necrosis was found in three out of nine brains injected with LPS on P14. None of these injuries was observed in any control rat brains. No histological changes in gray matter were noted in the LPS-injected rat brain. Proinflammatory cytokines, tumor necrosis factor-alpha (TNFalpha), interleukin-1beta (IL-1beta) and interleukin-6 (IL-6), and inducible nitric oxide synthase (iNOS) in the rat brain were greatly induced after LPS administration. Activated astrocytes and microglia/macrophages were found in the affected rat brains. Double-labeling showed that IL-1beta and iNOS expressing cells were microglia/macrophages. Injury to or delayed development of immature oligodendrocytes (OLs) was evident by decreased immunostaining for both O4 and O1 antibodies, markers for developing immature OLs, in the LPS-injected as compared to the control rat brain. LPS also resulted in hypomyelination, as indicated by reduced myelin basic protein (MBP) immunostaining in the P8 rat brain. Co-administration of IL-1 receptor antagonist (IL-1Ra) with LPS reduced brain injury by improving myelination and subsequent reduction of lateral ventricle enlargement. These results indicate that developing OLs may be the target cells for LPS-induced brain injury and inflammatory cytokines are possible mediators of LPS-induced brain injury.


Assuntos
Animais Recém-Nascidos/fisiologia , Encefalite/induzido quimicamente , Lipopolissacarídeos/administração & dosagem , Bainha de Mielina/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Animais , Encéfalo/patologia , Senescência Celular , Ventrículos Cerebrais/patologia , Citoproteção , Encefalite/patologia , Feminino , Gliose/patologia , Injeções , Proteína Antagonista do Receptor de Interleucina 1 , Masculino , Bainha de Mielina/fisiologia , Necrose , Oligodendroglia/fisiologia , Ratos , Ratos Sprague-Dawley , Sialoglicoproteínas/farmacologia , Coloração e Rotulagem
19.
Cardiovasc Diagn Ther ; 4(6): 487-94, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25610806

RESUMO

BACKGROUND: The Framingham risk score (FRS) has long been used as a global tool to estimate coronary heart disease (CHD) risk, but data has shown that subclinical CHD may exist in those classified as low risk by FRS, and as a result, there is potential for misclassification. Lipoprotein-associated phospholipase A2 (Lp-PLA2) and carotid intima-media thickness (CIMT) are two emerging risk markers that are predictive of future CHD events. PURPOSE: To examine Lp-PLA2 and CIMT values in low risk individuals, and to explore the relationship between Lp-PLA2 and CIMT. METHODS: A total of 229 men and women (age =53±7 years) underwent body composition analysis, objective physical activity measurement, fasting blood draw to determine standard lipid values and Lp-PLA2 mass, and CIMT measurement through ultrasound. RESULTS: For all subjects, mean CIMT was 0.61±0.1 mm, mean Lp-PLA2 mass was 197±45 ng/dL. A total of 19.5% and 34.6% of women and 4.6% and 73.8% of men were considered at elevated risk for CHD by CIMT (>75(th) percentile for age) and Lp-PLA2 mass (>200 ng/dL) standards, respectively. Both CIMT and Lp-PLA2 mass were significant independent predictors of each other, whereas traditional risk markers (lipids, glucose) were not. CONCLUSIONS: Results suggest that in those classified as low risk by FRS, evidence of increased CHD risk may exist through the use of newer risk markers like CIMT and Lp-PLA2. These emerging markers may aid in the earlier detection and intervention of subclinical CHD.

20.
Brain Behav ; 2(1): 53-67, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22574274

RESUMO

An in vitro myelination model derived from rat central nervous system (CNS) remains to be established. Here, we describe a simple and reproducible myelination culture method using dissociated neuron-oligodendrocyte (OL) co-cultures from either the embryonic day 16 (E16) rat spinal cord or cerebral cortex. The dissociated cells are plated directly on poly-L-lysine-coated cover slips and maintained in a modified myelination medium that supports both OL and neuron differentiation. The spinal cord derived OL progenitor cells develop quickly into myelin basic protein (MBP)+ mature OLs and start to myelinate axons around 17 days in vitro (DIV17). Myelination reaches its peak around six weeks (DIV40) and the typical nodes of Ranvier are revealed by paranodal proteins Caspr and juxaparanodal protein Kv1.2 immunoreactivity. Electron microscopy (EM) shows typical myelination cytoarchitecture and synaptic organization. In contrast, the cortical-derived co-culture requires triiodothyronine (T3) in the culture medium for myelination. Finally, either hypomyelination and/or demyelination can be induced by exposing proinflammatory cytokines or demyelinating agents to the co-culture, suggesting the feasibility of this modified in vitro myelination model for myelin-deficit investigation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA