Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Anesthesiology ; 120(3): 626-38, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24158051

RESUMO

BACKGROUND: The authors have previously shown that exposure of the neonatal nonhuman primate (NHP) brain to isoflurane for 5 h causes widespread acute apoptotic degeneration of neurons and oligodendrocyte. The current study explored the potential apoptogenic action of isoflurane in the fetal NHP brain. METHODS: Fetal rhesus macaques at gestational age of 120 days (G120) were exposed in utero for 5 h to isoflurane anesthesia (n = 5) or to no anesthesia (control condition; n = 4), and all regions of the brain were systematically evaluated 3 h later for evidence of apoptotic degeneration of neurons or glia. RESULTS: Exposure of the G120 fetal NHP brain to isoflurane caused a significant increase in apoptosis of neurons and of oligodendrocytes at a stage when oligodendrocytes were just beginning to myelinate axons. The neuroapoptosis response was most prominent in the cerebellum, caudate, putamen, amygdala, and several cerebrocortical regions. Oligodendrocyte apoptosis was diffusely distributed over many white matter regions. The total number of apoptotic profiles (neurons + oligodendrocytes) in the isoflurane-exposed brains was increased 4.1-fold, compared with the brains from drug-naive controls. The total number of oligodendrocytes deleted by isoflurane was higher than the number of neurons deleted. CONCLUSIONS: Isoflurane anesthesia for 5 h causes death of neurons and oligodendrocytes in the G120 fetal NHP brain. In the fetal brain, as the authors previously found in the neonatal NHP brain, oligodendrocytes become vulnerable when they are just achieving myelination competence. The neurotoxic potential of isoflurane increases between the third trimester (G120) and the neonatal period in the NHP brain.


Assuntos
Anestésicos Inalatórios/toxicidade , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Isoflurano/toxicidade , Neurônios/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Encéfalo/embriologia , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Macaca mulatta , Neurônios/patologia , Oligodendroglia/patologia
2.
Ann Neurol ; 72(4): 525-35, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23109147

RESUMO

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


Assuntos
Anestésicos Inalatórios/toxicidade , Apoptose/efeitos dos fármacos , Encéfalo/patologia , Isoflurano/toxicidade , Oligodendroglia/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Axônios/efeitos dos fármacos , Axônios/fisiologia , Caspases/fisiologia , Morte Celular/fisiologia , Imuno-Histoquímica , Macaca mulatta , Bainha de Mielina/fisiologia , Regeneração Nervosa/fisiologia , Fixação de Tecidos
3.
Anesthesiology ; 116(2): 372-84, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22222480

RESUMO

BACKGROUND: Exposure of rhesus macaque fetuses for 24 h or neonates for 9 h to ketamine anesthesia causes neuroapoptosis in the developing brain. The current study clarifies the minimum exposure required for and the extent and spatial distribution of ketamine-induced neuroapoptosis in rhesus fetuses and neonates. METHOD: Ketamine was administered by IV infusion for 5 h to postnatal day 6 rhesus neonates or to pregnant rhesus females at 120 days' gestation (full term = 165 days). Three hours later, fetuses were delivered by cesarean section, and the fetal and neonatal brains were studied for evidence of apoptotic neurodegeneration, as determined by activated caspase-3 staining. RESULTS: Both the fetal (n = 3) and neonatal (n = 4) ketamine-exposed brains had a significant increase in apoptotic profiles compared with drug-naive controls (fetal n = 4; neonatal n = 5). Loss of neurons attributable to ketamine exposure was 2.2 times greater in fetuses than in neonates. The pattern of neurodegeneration in fetuses was different from that in neonates, and all subjects exposed at either age had a pattern characteristic for that age. CONCLUSION: The developing rhesus macaque brain is sensitive to the apoptogenic action of ketamine at both a fetal and neonatal age, and exposure duration of 5 h is sufficient to induce a significant neuroapoptosis response at either age. The pattern of neurodegeneration induced by ketamine in fetuses was different from that in neonates, and loss of neurons attributable to ketamine exposure was 2.2 times greater in the fetal than neonatal brains.


Assuntos
Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Feto/efeitos dos fármacos , Ketamina/toxicidade , Degeneração Neural/induzido quimicamente , Animais , Animais Recém-Nascidos , Apoptose/fisiologia , Encéfalo/patologia , Feminino , Feto/patologia , Infusões Intravenosas , Ketamina/administração & dosagem , Macaca mulatta , Degeneração Neural/patologia , Gravidez , Distribuição Aleatória
4.
Am J Pathol ; 176(6): 2695-706, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20472884

RESUMO

Several different deletions within the N-terminal tail of the prion protein (PrP) induce massive neuronal death when expressed in transgenic mice. This toxicity is dose-dependently suppressed by coexpression of full-length PrP, suggesting that it results from subversion of a normal physiological activity of cellular PrP. We performed a combined biochemical and morphological analysis of Tg(DeltaCR) mice, which express PrP carrying a 21-aa deletion (residues 105-125) within a highly conserved region of the protein. Death of cerebellar granule neurons in Tg(DeltaCR) mice is not accompanied by activation of either caspase-3 or caspase-8 or by increased levels of the autophagy marker, LC3-II. In electron micrographs, degenerating granule neurons displayed a unique morphology characterized by heterogeneous condensation of the nuclear matrix without formation of discrete chromatin masses typical of neuronal apoptosis. Our data demonstrate that perturbations in PrP functional activity induce a novel, nonapoptotic, nonautophagic form of neuronal death whose morphological features are reminiscent of those associated with excitotoxic stress.


Assuntos
Morte Celular/fisiologia , Cerebelo/citologia , Neurônios/fisiologia , Proteínas PrPC/toxicidade , Animais , Apoptose/fisiologia , Autofagia/fisiologia , Biomarcadores/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Forma Celular , Ativação Enzimática , Camundongos , Camundongos Endogâmicos CBA , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Neurônios/patologia , Neurônios/ultraestrutura , Proteínas PrPC/genética , Príons/genética , Príons/metabolismo
5.
Neurobiol Dis ; 40(1): 200-6, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20580929

RESUMO

The ability of brief exposure to alcohol to cause widespread neuroapoptosis in the developing rodent brain and subsequent long-term neurocognitive deficits has been proposed as a mechanism underlying the neurobehavioral deficits seen in fetal alcohol spectrum disorder (FASD). It is unknown whether brief exposure to alcohol causes apoptosis in the fetal primate brain. Pregnant fascicularis macaques at various stages of gestation (G105 to G155) were exposed to alcohol for 8h, then the fetuses were delivered by caesarean section and their brains perfused with fixative and evaluated for apoptosis. Compared to saline control brains, the ethanol-exposed brains displayed a pattern of neuroapoptosis that was widespread and similar to that caused by alcohol in infant rodent brain. The observed increase in apoptosis was on the order of 60-fold. We propose that the apoptogenic action of alcohol could explain many of the neuropathological changes and long-term neuropsychiatric disturbances associated with human FASD.


Assuntos
Transtornos do Sistema Nervoso Induzidos por Álcool/patologia , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/patologia , Animais , Apoptose/fisiologia , Encéfalo/fisiopatologia , Contagem de Células , Depressores do Sistema Nervoso Central/toxicidade , Modelos Animais de Doenças , Esquema de Medicação , Feminino , Macaca fascicularis , Degeneração Neural/induzido quimicamente , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Tempo
6.
Anesthesiology ; 112(4): 834-41, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20234312

RESUMO

BACKGROUND: Brief isoflurane anesthesia induces neuroapoptosis in the developing rodent brain, but susceptibility of non-human primates to the apoptogenic action of isoflurane has not been studied. Therefore, we exposed postnatal day 6 (P6) rhesus macaques to a surgical plane of isoflurane anesthesia for 5 h, and studied the brains 3 h later for histopathologic changes. METHOD: With the same intensity of physiologic monitoring typical for human neonatal anesthesia, five P6 rhesus macaques were exposed for 5 h to isoflurane maintained between 0.7 and 1.5 end-tidal Vol% (endotracheally intubated and mechanically ventilated) and five controls were exposed for 5 h to room air without further intervention. Three hours later, the brains were harvested and serially sectioned across the entire forebrain and midbrain, and stained immunohistochemically with antibodies to activated caspase-3 for detection and quantification of apoptotic neurons. RESULTS: Quantitative evaluation of brain sections revealed a median of 32.5 (range, 18.0-48.2) apoptotic cells/mm of brain tissue in the isoflurane group and only 2.5 (range, 1.1-5.2) in the control group (difference significant at P = 0.008). Apoptotic neuronal profiles were largely confined to the cerebral cortex. In the control brains, they were sparse and randomly distributed, whereas in the isoflurane brains they were abundant and preferentially concentrated in specific cortical layers and regions. CONCLUSION: The developing non-human primate brain is sensitive to the apoptogenic action of isoflurane and displays a 13-fold increase in neuroapoptosis after 5 h exposure to a surgical plane of isoflurane anesthesia.


Assuntos
Anestésicos Inalatórios/toxicidade , Animais Recém-Nascidos/fisiologia , Apoptose/efeitos dos fármacos , Encéfalo/citologia , Isoflurano/toxicidade , Neurônios/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Caspase 3/metabolismo , Hemodinâmica/fisiologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Intubação Intratraqueal , Macaca mulatta , Células Piramidais/efeitos dos fármacos , Células Piramidais/ultraestrutura
7.
Anesth Analg ; 110(2): 442-8, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19955510

RESUMO

Millions of human fetuses, infants, and children are exposed to anesthetic drugs every year in the United States and throughout the world. Anesthesia administered during critical stages of neurodevelopment has been considered safe and without adverse long-term consequences. However, recent reports provide mounting evidence that exposure of the immature animal brain to anesthetics during the period of rapid synaptogenesis, also known as the brain growth spurt period, triggers widespread apoptotic neurodegeneration, inhibits neurogenesis, and causes significant long-term neurocognitive impairment. Herein, we summarize currently available evidence for anesthesia-induced pathological changes in the brain and associated long-term neurocognitive deficits and discuss promising strategies for protecting the developing brain from the potentially injurious effects of anesthetic drugs while allowing the beneficial actions of these drugs to be realized.


Assuntos
Anestésicos/toxicidade , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Anestésicos/efeitos adversos , Animais , Anticonvulsivantes/farmacologia , Temperatura Corporal , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Etanol/farmacologia , Feto/efeitos dos fármacos , Humanos , Hipotermia Induzida , Camundongos , Doenças Neurodegenerativas/induzido quimicamente
8.
Neurobiol Dis ; 33(1): 111-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18992344

RESUMO

Although a wide range of developmental disabilities following fetal alcohol exposure are observed clinically, the molecular factors that determine the severity of these sequelae remain undefined. In mice exposed to ethanol, deletion of adenylyl cyclases (ACs) 1 and 8 exacerbates the neuroapoptosis that occurs in a prolonged post-treatment period; however, it remains unclear whether AC1 and AC8 are critical to the primary or secondary mechanisms underlying ethanol-induced neurodegeneration. Here we demonstrate that mice lacking AC1 and AC8 (DKO) display significantly increased apoptosis in the striatum, a region sensitive to neuroapoptosis in the acute post-treatment period, compared to WT controls. The enhanced neuroapoptotic response observed in the striatum of DKO mice is accompanied by significant reductions in phosphorylation of known pro-survival proteins, insulin receptor substrate-1 (IRS-1), Akt and extracellular signal-regulated kinases (ERKs). These data suggest that AC1/AC8 are crucial activators of cell survival signaling pathways acutely following ethanol exposure and represent molecular factors that may directly modulate the severity of symptoms associated with Fetal Alcohol Syndrome.


Assuntos
Adenilil Ciclases/metabolismo , Apoptose/fisiologia , Corpo Estriado/fisiologia , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Adenilil Ciclases/genética , Animais , Animais Recém-Nascidos , Caspase 3/metabolismo , Sobrevivência Celular , Corpo Estriado/citologia , Corpo Estriado/crescimento & desenvolvimento , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Transtornos do Espectro Alcoólico Fetal/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/fisiologia , Fosforilação , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Neurobiol Dis ; 34(1): 1-10, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19100327

RESUMO

Dimethyl sulfoxide (DMSO) is a solvent that is routinely used as a cryopreservative in allogous bone marrow and organ transplantation. We exposed C57Bl/6 mice of varying postnatal ages (P0-P30) to DMSO in order to study whether DMSO could produce apoptotic degeneration in the developing CNS. DMSO produced widespread apoptosis in the developing mouse brain at all ages tested. Damage was greatest at P7. Significant elevations above the background rate of apoptosis occurred at the lowest dose tested, 0.3 ml/kg. In an in vitro rat hippocampal culture preparation, DMSO produced neuronal loss at concentrations of 0.5% and 1.0%. The ability of DMSO to damage neurons in dissociated cultures indicates that the toxicity likely results from a direct cellular effect. Because children, who undergo bone marrow transplantation, are routinely exposed to DMSO at doses higher than 0.3 ml/kg, there is concern that DMSO might be producing similar damage in human children.


Assuntos
Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Dimetil Sulfóxido/toxicidade , Neurônios/efeitos dos fármacos , Envelhecimento , Animais , Animais Recém-Nascidos , Encéfalo/ultraestrutura , Caspase 3/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/induzido quimicamente , Neurônios/fisiologia , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley , Solventes/toxicidade
10.
Anesthesiology ; 110(4): 862-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19293695

RESUMO

BACKGROUND: Ethanol and anesthetic drugs trigger neuroapoptosis in the developing mouse brain. Recently, it was found that ethanol-induced neuroapoptosis is preceded by suppressed phosphorylation of extracellular signal-regulated protein kinase (ERK), and lithium counteracts both the phosphorylated ERK suppressant action and ethanol-induced neuroapoptosis. The current study was undertaken to address the following questions. (1) Do ketamine and propofol mimic ethanol in suppressing ERK phosphorylation? (2) If they do, does lithium prevent this suppressant action and also prevent these anesthetic drugs from triggering neuroapoptosis? METHOD: Postnatal day 5 mice were treated with propofol, ketamine, lithium, a combination of propofol or ketamine with lithium or saline, and their brains were prepared for Western blot analysis or histology. For Western blot, cytosolic lysates of caudate putamen were analyzed for expression of phosphorylated ERK and phosphorylated serine/threonine-specific protein kinase. For histology, brains were stained immunohistochemically with antibodies to activated caspase-3, and the density of activated caspase-3 positive cells was determined. RESULTS: Ketamine and propofol suppressed phosphorylated ERK, and lithium counteracted both the phosphorylated ERK suppressant action and neuroapoptotic action of these anesthetic drugs. CONCLUSION: If further testing finds lithium to be safe for use in pediatric/obstetric medicine, administration of a single dose of lithium before anesthesia induction may be a suitable means of mitigating the risk of anesthesia-induced developmental neuroapoptosis.


Assuntos
Antipsicóticos/uso terapêutico , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Carbonato de Lítio/uso terapêutico , Neurônios/efeitos dos fármacos , Anestésicos Intravenosos/toxicidade , Animais , Western Blotting , Encéfalo/embriologia , Encéfalo/patologia , Caspase 3/efeitos dos fármacos , Ketamina/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Propofol/toxicidade , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Distribuição Aleatória , Resultado do Tratamento
11.
J Neurosci ; 27(8): 2081-90, 2007 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-17314303

RESUMO

Neuronal ceroid lipofuscinosces/Batten disease (NCL) is a devastating group of neurodegenerative diseases caused by genetic disruptions in lysosomal function. Cathepsin D (CD) is a major lysosomal protease, and mutations in CD that render it enzymatically defective have been reported recently in subsets of NCL patients. The targeted deletion of CD in mice results in extensive neuropathology, including biochemical and morphological evidence of apoptosis and autophagic stress (aberrant autophagosome accumulation), effects that are similar to those observed in NCL. To determine the contribution of Bax-dependent apoptosis in this mouse model of NCL, combined Bax- and CD-deficient mice were generated. Morphological analysis of CD-deficient mouse brains indicated large numbers of pyknotic neurons and neurons with marked cytoplasmic swellings containing undigested lipofuscin. Cell death and apoptosis were evidenced by increases in terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL) reactivity and activation of caspase-3, respectively. DeOlmos silver-positive neurons were abundant in CD-deficient brain and correlated with neuron loss, as indicated by significant decreases in NeuN (neuronal nuclear antigen)-positive neurons. Lysosome dysfunction and autophagic stress were apparent in CD-deficient brain as indicated by the accumulation of autofluorescent storage material and by increased levels of LC3-II (light chain 3-II, a selective autophagosome marker), respectively. Bax deletion significantly inhibited caspase-3 activation and hippocampal TUNEL reactivity but did not prevent the majority of CD deficiency-induced neuropathology, including the persistence of pyknotic neurons, elevated cortical TUNEL reactivity, lysosome dysfunction and autophagic stress, neurodegeneration, and neuron loss. Together, these results suggest that CD deficiency-induced neuropathology does not require Bax-dependent apoptosis and highlights the importance of caspase-independent neuron death and neurodegeneration resulting from the genetic disruption of lysosome function.


Assuntos
Apoptose , Catepsina D/deficiência , Degeneração Neural/etiologia , Degeneração Neural/fisiopatologia , Proteína X Associada a bcl-2/deficiência , Animais , Autofagia , Encéfalo/metabolismo , Encéfalo/patologia , Caspase 3/metabolismo , Morte Celular , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Ativação Enzimática , Hipocampo/fisiopatologia , Marcação In Situ das Extremidades Cortadas , Lipofuscina/metabolismo , Lisossomos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Proteínas do Tecido Nervoso/metabolismo , Lipofuscinoses Ceroides Neuronais , Neurônios/metabolismo , Neurônios/patologia , Proteínas Nucleares/metabolismo
12.
Neurobiol Dis ; 31(3): 355-60, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18595723

RESUMO

Transient exposure of immature animals during the brain growth spurt period to ethanol triggers neuroapoptosis in the developing brain. Here we report that lithium, when administered in a single, well-tolerated dose to infant mice, suppresses spontaneous neuroapoptosis that occurs naturally in the developing brain, and prevents ethanol from triggering neuroapoptosis. To explore lithium's mechanism of action, we focused on kinase signaling systems (ERK, Akt, JNK) that are believed to play a regulatory role in cell survival, and found that very rapidly after ethanol administration there is a suppression of ERK phosphorylation, and that lithium stimulates ERK phosphorylation and prevents ethanol from suppressing this phosphorylation process. Ethanol also suppressed pAKT, but lithium did not counteract this effect. We also found that ethanol activates the JNK system, but this cannot explain the neurotoxic action of ethanol, because JNK activation did not occur in the same neuronal populations that are killed by ethanol.


Assuntos
Transtornos do Sistema Nervoso Induzidos por Álcool/enzimologia , Transtornos do Sistema Nervoso Induzidos por Álcool/prevenção & controle , Encéfalo/efeitos dos fármacos , Etanol/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Compostos de Lítio/farmacologia , Transtornos do Sistema Nervoso Induzidos por Álcool/fisiopatologia , Animais , Animais Recém-Nascidos , Antimaníacos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Encéfalo/enzimologia , Depressores do Sistema Nervoso Central/antagonistas & inibidores , Depressores do Sistema Nervoso Central/toxicidade , Modelos Animais de Doenças , Interações Medicamentosas/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Etanol/toxicidade , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/tratamento farmacológico , Degeneração Neural/enzimologia , Degeneração Neural/fisiopatologia , Fosforilação/efeitos dos fármacos
13.
Anesth Analg ; 106(6): 1712-4, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18499599

RESUMO

Drugs that block N-methyl-d-aspartate glutamate receptors or that promote gamma-aminobutyric acid type A inhibition trigger neuroapoptosis in the developing rodent brain. Propofol reportedly interacts with both gamma-aminobutyric acid type A and N-methyl-d-aspartate glutamate receptors, but has not been adequately evaluated for its ability to induce developmental neuroapoptosis. Here we determined that the intraperitoneal (i.p.) dose of propofol required to induce a surgical plane of anesthesia in the infant mouse is 200 mg/kg. We then administered graduated doses of propofol (25-300 mg/kg i.p.) and found that doses >or=50 mg/kg induce a significant neuroapoptosis response. We conclude that propofol induces neuroapoptosis at 1/4 the dose required for surgical anesthesia.


Assuntos
Anestésicos Gerais/toxicidade , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Propofol/toxicidade , Anestésicos Gerais/administração & dosagem , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Estado de Consciência/efeitos dos fármacos , Relação Dose-Resposta a Droga , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Propofol/administração & dosagem , Reflexo/efeitos dos fármacos
14.
J Neurosurg Anesthesiol ; 20(1): 21-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18157021

RESUMO

Drugs that suppress neuronal activity, including general anesthetics used in pediatric and obstetric medicine, trigger neuroapoptosis in the developing rodent brain. Exposure of infant rats for 6 hours to a combination of anesthetic drugs (midazolam, nitrous oxide, isoflurane) reportedly causes widespread apoptotic neurodegeneration, followed by lifelong cognitive deficits. Isoflurane, the dominant ingredient in this triple cocktail, has not been evaluated individually for apoptogenic potential. It was recently reported that (1) the minimum alveolar concentration (MAC) for anesthetizing infant mice with isoflurane is 2.26%, and; (2) that infant mice, without assisted respiration, maintain normal arterial oxygen values but become hypoglycemic when exposed to isoflurane 3% for 30 minutes, then 1.8% for 1 hour (1.46 MAC-hours). In the present experiments, infant mice were exposed to isoflurane at various sub-MAC concentrations and durations, and the brains were evaluated quantitatively 5 hours after initiation of anesthesia exposure to determine the number of neuronal profiles undergoing apoptosis. Blood glucose values were also determined under each of these conditions. All conditions tested (isoflurane at 0.75% for 4 h, 1.5% for 2 h, 2.0% for 1 h) triggered a statistically significant increase in neuroapoptosis compared with the rate of spontaneous apoptosis in littermate controls. Blood glucose determinations ruled out hypoglycemia as a potential cause of the brain damage. It is concluded that exposure to sub-MAC concentrations of isoflurane for one or more hours triggers neuroapoptosis in the infant mouse brain. These findings are consistent with other recent evidence demonstrating that brief exposure to ethanol, ketamine, or midazolam triggers neuroapoptosis in the developing mouse brain.


Assuntos
Anestésicos Inalatórios/toxicidade , Apoptose/efeitos dos fármacos , Encéfalo/patologia , Isoflurano/toxicidade , Neurônios/patologia , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Encéfalo/crescimento & desenvolvimento , Contagem de Células , Hipoglicemia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo
15.
Sci Rep ; 8(1): 5302, 2018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29593226

RESUMO

Caffeine is the most frequently used medication in premature infants. It is the respiratory stimulant of choice for apnea associated with prematurity and has been called the silver bullet in neonatology because of many proven benefits and few known risks. Research has revealed that sedative/anesthetic drugs trigger apoptotic death of neurons and oligodendrocytes in developing mammalian brains. Here we evaluated the influence of caffeine on the neurotoxicity of anesthesia in developing nonhuman primate brains. Fetal macaques (n = 7-8/group), at a neurodevelopmental age comparable to premature human infants, were exposed in utero for 5 hours to no drug (control), isoflurane, or isoflurane + caffeine and examined for evidence of apoptosis. Isoflurane exposure increased apoptosis 3.3 fold for neurons and 3.4 fold for oligodendrocytes compared to control brains. Isoflurane + caffeine caused neuronal apoptosis to increase 8.0 fold compared to control levels but did not augment oligoapoptosis. Neuronal death was particularly pronounced in the basal ganglia and cerebellum. Higher blood levels of caffeine within the range considered therapeutic and safe for human infants correlated with increased neuroapoptosis. Caffeine markedly augments neurotoxicity of isoflurane in the fetal macaque brain and challenges the assumption that caffeine is safe for premature infants.


Assuntos
Cafeína/efeitos adversos , Desenvolvimento Fetal/efeitos dos fármacos , Isoflurano/efeitos adversos , Anestesia/efeitos adversos , Anestésicos Inalatórios/efeitos adversos , Anestésicos Inalatórios/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Cafeína/farmacologia , Morte Celular/efeitos dos fármacos , Feminino , Isoflurano/farmacologia , Macaca mulatta/embriologia , Masculino , Neurônios/fisiologia , Oligodendroglia/efeitos dos fármacos , Gravidez
16.
J Neurosci ; 26(15): 3923-32, 2006 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-16611808

RESUMO

Memantine, a drug recently approved for treatment of Alzheimer's disease, has been characterized as a unique NMDA antagonist that confers protection against excitotoxic neurodegeneration without the serious side effects that other NMDA antagonists are known to cause. In the present study, we determined what dose of memantine is required to protect the adult rat brain against an NMDA receptor-mediated excitotoxic process and then tested that dose and a range of lower doses to determine whether the drug in this dose range is associated with significant side effects. Consistent with previous research, we found that memantine confers a neuroprotective effect beginning at an intraperitoneal dose of 20 mg/kg, a dose that we found, contrary to previous reports, produces locomotor disturbances severe enough to preclude testing for learning and memory effects. We then determined that, at intraperitoneal doses of 10 and 5 mg/kg, memantine disrupts both memory and locomotor behaviors. Rats treated with these doses performed at control-like levels in learning a hole-board task but were significantly impaired in demonstrating what they had learned when tested 24 h later. This impairment of memory retention was not state dependent in that it was demonstrable regardless of whether the rats were or were not exposed to memantine on the day of retention testing. We conclude that, in the adult rat, memantine behaves like other NMDA antagonists in that it is neuroprotective only at doses that produce intolerable side effects, including memory impairment.


Assuntos
Antagonistas de Aminoácidos Excitatórios/farmacologia , Memantina/farmacologia , Memória/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Animais , Antiparkinsonianos/farmacologia , Edema Encefálico/induzido quimicamente , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Memória/fisiologia , Transtornos da Memória/induzido quimicamente , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley
17.
J Neurosci ; 26(24): 6618-26, 2006 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-16775150

RESUMO

Neurons engage compensatory, homeostatic synaptic changes to maintain their overall firing rate. We examined the induction and expression of a persistent presynaptic adaptation. We explored the effect of mild extracellular potassium elevation to increase hippocampal pyramidal neuron spiking over a physiological range. With several days of mild depolarization, glutamate release adapted, as revealed by an increased mismatch between the number of active, FM1-43-positive, glutamatergic synapses and the total number of synapses defined by vesicular glutamate transporter-1 antibody staining. Surprisingly, the adaptation of glutamate terminals was all-or-none; recycling vesicle pool size at remaining active synapses was not significantly altered by the adaptation. Tetrodotoxin (TTX), but not postsynaptic receptor blockade, reversed depolarization-induced adaptation, and TTX added to normal incubation medium increased the number of active synapses, suggesting that normal spiking activity sustains a steady-state percentage of inactive terminals. Chronic mild depolarization depressed EPSCs and decreased the size of the readily releasable pool of vesicles (RRP). Several hours of 10 Hz electrical stimulation also depressed the RRP size, confirming that spiking alone induces adaptation and that strong stimulation induces more rapid presynaptic adaptation. Despite the importance of RRP alteration to the adaptation, ultrastructural experiments revealed no changes in docked or total synaptic vesicle numbers. Furthermore, alpha-latrotoxin induced vesicle release at adapted synapses, consistent with the idea that adaptation resulted from changes in vesicle priming. These results show that glutamatergic neurotransmission persistently adapts to changes in electrical activity over a wide physiological range.


Assuntos
Inibição Neural/fisiologia , Neurônios/citologia , Sinapses/fisiologia , Vesículas Sinápticas/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , Potenciais de Ação/fisiologia , Potenciais de Ação/efeitos da radiação , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/citologia , Imuno-Histoquímica/métodos , Microscopia Eletrônica de Transmissão/métodos , Inibição Neural/efeitos dos fármacos , Inibição Neural/efeitos da radiação , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neurônios/efeitos da radiação , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp/métodos , Cloreto de Potássio/farmacologia , Compostos de Piridínio/farmacocinética , Compostos de Amônio Quaternário/farmacocinética , Quinoxalinas/farmacologia , Ratos , Sinapses/efeitos dos fármacos , Sinapses/efeitos da radiação , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/ultraestrutura , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
18.
J Neurosci ; 25(9): 2376-85, 2005 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-15745964

RESUMO

Fetal alcohol exposure results in cognitive and neurobehavioral deficits, but the effects of modifying genetic loci on the severity of these sequelas have not been well characterized. Although the cAMP signaling pathway has been shown to be an important modulator of ethanol sensitivity in adult mice, its potential role in modulating ethanol-induced neurodegeneration has not been examined. Adenylyl cyclases (ACs) 1 and 8 produce cAMP in response to intracellular calcium elevation and modulate several aspects of neuronal function, including ethanol sensitivity. AC1 and AC8 are expressed widely throughout the brain of neonatal mice, and genetic deletion of both AC1 and AC8 in double-knock-out (DKO) mice enhances ethanol-induced neurodegeneration in the brains of neonatal mice. In addition, ethanol treatment induces significantly greater levels of caspase-3 activation in the brains of DKO mice compared with wild-type (WT) mice, reflecting higher numbers of apoptotic neurons. Administration of the NMDA receptor antagonist MK801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d] cyclohepten-5,10-imine hydrogen maleate] or the GABA(A) receptor potentiator phenobarbital, which mimics components of the effects of ethanol on neurons, results in significantly greater neurodegeneration in the brains of neonatal DKO mice than WT mice. Furthermore, loss of a single calcium-stimulated AC isoform potentiates neurodegeneration after administration of ethanol, MK801, or phenobarbital. In contrast, the levels of physiological cell death, death after hypoxia/ischemia, and excitotoxic cell death are not increased in the brains of DKO mice. Thus, AC1 and AC8 are critical modulators of neurodegeneration induced by activity blockade in the neonatal brain and represent genetic loci that may potentially modify the severity of fetal alcohol syndrome.


Assuntos
Adenilil Ciclases/metabolismo , Encéfalo/efeitos dos fármacos , Cálcio/farmacologia , Etanol , Doenças Neurodegenerativas/induzido quimicamente , Anilidas/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Animal , Western Blotting/métodos , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Caspase 3 , Caspases/metabolismo , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Etanol/sangue , Moduladores GABAérgicos/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Hipóxia/metabolismo , Hipóxia/patologia , Hibridização In Situ/métodos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Oligopeptídeos/metabolismo , Fenobarbital/farmacologia , Coloração pela Prata/métodos , Fatores de Tempo
19.
Brain Res ; 1107(1): 70-81, 2006 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-16822489

RESUMO

Closed head injury to the developing rat brain causes an acute excitotoxic lesion and axonal disruption at the impact site followed by a delayed pattern of apoptotic damage at various distant sites. Using an electromagnetic impact device to deliver a precisely controlled degree of mechanical deformation to the P7 infant rat skull, we studied the distribution of distant apoptotic lesions and the sequence and time course with which these lesions evolve following relatively mild closed head injury. The first major wave of apoptotic neurodegeneration occurred at 8 h postimpact in the retrosplenial cortex and pre- and parasubiculum. The next major wave occurred in the 16- to 24-h interval and was localized to the anterior thalamic nuclei. A third wave was detected at 36 to 48 h in the mammillary nuclei. We propose that the first and second waves were triggered by injury to a specific fiber tract, the corpus callosum/cingulum bundle that conveys reciprocal connections between the anterior thalamic nuclei and retrosplenial/pre- and parasubicular neurons. This fiber tract passes through a zone of maximum mechanical strain, as measured by tagged MRI. The third wave affecting mammillary neurons occurred because the principal synaptic targets of these neurons are the anterior thalamic neurons that were destroyed in the second wave of degeneration. Prevention of these apoptotic waves of brain damage is a realistic goal in view of the long delay between the impact event and onset of apoptotic degeneration.


Assuntos
Apoptose , Lesões Encefálicas/complicações , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Degeneração Neural/etiologia , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Caspase 3 , Caspases/metabolismo , Fenômenos Eletromagnéticos/métodos , Imuno-Histoquímica/métodos , Microscopia Eletrônica de Transmissão/métodos , Ratos , Ratos Sprague-Dawley , Coloração pela Prata/métodos , Fatores de Tempo
20.
Sci Rep ; 6: 22427, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26951756

RESUMO

Exposure of infant animals, including non-human primates (NHPs), to anaesthetic drugs causes apoptotic death of neurons and oligodendrocytes (oligos) and results in long-term neurodevelopmental impairment (NDI). Moreover, retrospective clinical studies document an association between anaesthesia exposure of human infants and significant increase in NDI. These findings pose a potentially serious dilemma because millions of human infants are exposed to anaesthetic drugs every year as part of routine medical care. Lithium (Li) at clinically established doses is neuroprotective in various cerebral injury models. We therefore investigated whether Li also protects against anaesthesia neurotoxicity in infant NHPs. On postnatal day 6 NHPs were anaesthetized with the widely used anaesthetic isoflurane (ISO) for 5 h employing the same standards as in a human pediatric surgery setting. Co-administration of Li completely prevented the acute ISO-induced neuroapoptosis and significantly reduced ISO-induced apoptosis of oligodendroglia. Our findings are highly encouraging as they suggest that a relatively simple pharmacological manipulation might protect the developing primate brain against the neurotoxic action of anaesthetic drugs while not interfering with the beneficial actions of these drugs. Further research is needed to determine Li's potential to prevent long-term NDI resulting from ISO anaesthesia, and to establish its safety in human infants.


Assuntos
Anestésicos Inalatórios/toxicidade , Apoptose/efeitos dos fármacos , Isoflurano/toxicidade , Lítio/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Lítio/farmacocinética , Macaca mulatta , Transtornos do Neurodesenvolvimento/induzido quimicamente , Neurônios/efeitos dos fármacos , Neurônios/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA