Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 310: 306-21, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26404874

RESUMO

The state of protein folding in the endoplasmic reticulum (ER), via the unfolded protein response (UPR), regulates a pro- or anti-apoptotic cell fate. Hypoxic preconditioning (HPC) is a potent anti-apoptotic stimulus, wherein ischemic neural injury is averted by a non-damaging exposure to hypoxia. We tested if UPR modulation contributes to the pro-survival/anti-apoptotic phenotype in neurons preconditioned with hypoxia, using organotypic cultures of rat hippocampus as a model system. Pharmacologic induction of the UPR with tunicamycin increased mRNA of 79 of 84 UPR genes and replicated the pro-survival phenotype of HPC, whereas only small numbers of the same mRNAs were upregulated at 0, 6 and 24h after HPC. During the first 24h after HPC, protein signals in all 3 UPR pathways increased at various times: increased ATF4, phosphorylation of eif2α and IRE1, cleavage of xbb1 mRNA and cleavage of ATF6. Pharmacologic inhibition of ATF6 and IRE1 blocked HPC. Ischemia-like conditions (oxygen/glucose deprivation, OGD) caused extensive neuron cell damage and involved some of the same UPR protein signals as HPC. In distinction to HPC and tunicamycin, OGD caused widespread suppression of UPR genes: 55 of 84 UPR gene mRNAs were numerically downregulated. We conclude that although HPC and ischemic cell death in hippocampal neurons involve protein-based signaling in all 3 UPR pathways, these processes co-opt only a subset of the genomic response elicited by agents known to cause protein misfolding, possibly because of persistent transcription/translation arrest induced by hypoxia and especially OGD.


Assuntos
Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Hipóxia/metabolismo , Precondicionamento Isquêmico , Neurônios/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas , Animais , Isquemia Encefálica/genética , Morte Celular , Hipóxia Celular , Expressão Gênica , Glucose/metabolismo , Hipóxia/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
2.
Anaesthesia ; 68(12): 1220-3, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23992483

RESUMO

Pulse oximetry is a standard of care during anaesthesia in high-income countries. However, 70% of operating environments in low- and middle-income countries have no pulse oximeter. The 'Lifebox' oximetry project set out to bridge this gap with an inexpensive oximeter meeting CE (European Conformity) and ISO (International Organization for Standardization) standards. To date, there are no performance-specific accuracy data on this instrument. The aim of this study was to establish whether the Lifebox pulse oximeter provides clinically reliable haemoglobin oxygen saturation (Sp O2 ) readings meeting USA Food and Drug Administration 510(k) standards. Using healthy volunteers, inspired oxygen fraction was adjusted to produce arterial haemoglobin oxygen saturation (Sa O2 ) readings between 71% and 100% measured with a multi-wavelength oximeter. Lifebox accuracy was expressed using bias (Sp O2 - Sa O2 ), precision (SD of the bias) and the root mean square error (Arms). Simultaneous readings of Sa O2 and Sp O2 in 57 subjects showed a mean (SD) bias of -0.41% (2.28%) and Arms 2.31%. The Lifebox pulse oximeter meets current USA Food and Drug Administration standards for accuracy, thus representing an inexpensive solution for patient monitoring without compromising standards.


Assuntos
Hipóxia/diagnóstico , Monitorização Fisiológica/instrumentação , Monitorização Fisiológica/normas , Oximetria/instrumentação , Oximetria/normas , Adulto , Feminino , Voluntários Saudáveis , Humanos , Hipóxia/sangue , Masculino , Monitorização Fisiológica/métodos , Oximetria/métodos , Reprodutibilidade dos Testes
3.
Neuroscience ; 207: 316-25, 2012 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-22265728

RESUMO

This study examines the causes of hypothermia and rewarming injury in CA1, CA3, and dentate neurons in rat hippocampal slice cultures. Neuronal death, assessed with propidium iodide or Sytox fluorescence, Fluoro-Jade labeling, and Cresyl Violet staining, depended on the severity and duration of hypothermia. More than 6 h at temperatures less than 12 °C followed by rewarming to 37 °C (profound hypothermia and rewarming, PH/RW) caused swelling and death in large number of neurons in CA1, CA3, and dentate. During PH, [ATP] decreased and [Ca(2+)](I) and extracellular [glutamate] increased, with neuron rupture and nuclear condensation following RW. The data support the hypothesis that neuronal death from PH/RW is excitotoxic, due to ATP loss, glutamate receptor activation and Ca(2+) influx. We found that antagonism of N-methyl-D-aspartate (NMDA) receptors, but not 2-amino-3-(5-methyl-3-oxo-1,2- oxazol-4-yl) propanoic acid or metabotropic glutamate receptors, decreased neuron death and prevented increases in [Ca(2+)](I) caused by PH/RW. Chelating extracellular Ca(2+) decreased PH/RW injury, but inhibiting L- and T-type voltage-gated Ca(2+) channels, K+ channels, Ca(2+) release from the endoplasmic reticulum, and reverse Na(+)/Ca(2+) exchange did not affect the Ca(2+) changes or cell death. We conclude that the mechanism of PH/RW neuronal injury in hippocampal slices primarily involves intracellular Ca(2+) accumulation mediated by NMDA receptors that activates necrotic, but not apoptotic processes.


Assuntos
Sinalização do Cálcio/fisiologia , Ácido Glutâmico/fisiologia , Hipocampo/fisiopatologia , Hipotermia Induzida/efeitos adversos , Líquido Intracelular/fisiologia , Degeneração Neural/fisiopatologia , Animais , Temperatura Corporal/fisiologia , Hipocampo/metabolismo , Hipocampo/patologia , Líquido Intracelular/metabolismo , Degeneração Neural/etiologia , Degeneração Neural/metabolismo , Neurônios/patologia , Neurônios/fisiologia , Neurotoxinas/farmacologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
4.
Neuroscience ; 160(1): 51-60, 2009 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-19217932

RESUMO

Exposure of neurons to a non-lethal hypoxic stress greatly reduces cell death during subsequent severe ischemia (hypoxic preconditioning, HPC). In organotypic cultures of rat hippocampus, we demonstrate that HPC requires inositol triphosphate (IP3) receptor-dependent Ca2+ release from the endoplasmic reticulum (ER) triggered by increased cytosolic NAD(P)H. Ca2+ chelation with intracellular BAPTA, ER Ca2+ store depletion with thapsigargin, IP3 receptor block with xestospongin, and RNA interference against subtype 1 of the IP3 receptor all blunted the moderate increases in [Ca2+](i) (50-100 nM) required for tolerance induction. Increases in [Ca2+](i) during HPC and neuroprotection following HPC were not prevented with NMDA receptor block or by removing Ca2+ from the bathing medium. Increased NAD(P)H fluorescence in CA1 neurons during hypoxia and demonstration that NADH manipulation increases [Ca2+](i) in an IP3R-dependent manner revealed a primary role of cellular redox state in liberation of Ca2+ from the ER. Blockade of IP3Rs and intracellular Ca2+ chelation prevented phosphorylation of known HPC signaling targets, including MAPK p42/44 (ERK), protein kinase B (Akt) and CREB. We conclude that the endoplasmic reticulum, acting via redox/NADH-dependent intracellular Ca2+ store release, is an important mediator of the neuroprotective response to hypoxic stress.


Assuntos
Sinalização do Cálcio/fisiologia , Hipocampo/fisiopatologia , Hipóxia Encefálica/fisiopatologia , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , NADP/metabolismo , Neurônios/fisiologia , Animais , Isquemia Encefálica/prevenção & controle , Cálcio/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/fisiologia , Hipocampo/efeitos dos fármacos , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , NAD/metabolismo , Neurônios/efeitos dos fármacos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo
5.
Neuroscience ; 127(3): 673-83, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15283966

RESUMO

Although large increases in neuronal intracellular calcium concentrations ([Ca(2+)](i)) are lethal, moderate increases in [Ca(2+)](i) of 50-200 nM may induce immediate or long-term tolerance of ischemia or other stresses. In neurons in rat hippocampal slice cultures, we determined the relationship between [Ca(2+)](i), cell death, and Ca(2+)-dependent neuroprotective signals before and after a 45 min period of oxygen and glucose deprivation (OGD). Thirty minutes before OGD, [Ca(2+)](i) was increased in CA1 neurons by 40-200 nM with 1 nM-1 microM of a Ca(2+)-selective ionophore (calcimycin or ionomycin-"Ca(2+) preconditioning"). Ca(2+) preconditioning greatly reduced cell death in CA1, CA3 and dentate during the following 7 days, even though [Ca(2+)](i) was similar (approximately 2 microM) in preconditioned and control neurons 1 h after the OGD. When pre-OGD [Ca(2+)](i) was lowered to 25 nM (10 nM ionophore in Ca(2+)-free medium) or increased to 8 microM (10 microM ionophore), more than 90% of neurons died. Increased levels of the anti-apoptotic protein protein kinase B (Akt) and the MAP kinase ERK (p42/44) were present in preconditioned slices after OGD. Reducing Ca(2+) influx, inhibiting calmodulin, and preventing Akt or MAP kinase p42/44 upregulation prevented Ca(2+) preconditioning, supporting a specific role for Ca(2+) in the neuroprotective process. Further, in continuously oxygenated cultured hippocampal/cortical neurons, preconditioning for 30 min with 10 nM ionomycin reduced cell death following a 4 microM increase in [Ca(2+)](i) elicited by 1 microM ionomycin. Thus, a zone of moderately increased [Ca(2+)](i) before a potentially lethal insult promotes cell survival, uncoupling subsequent large increases in [Ca(2+)](i) from initiating cell death processes.


Assuntos
Apoptose/fisiologia , Cálcio/metabolismo , Hipocampo/metabolismo , Precondicionamento Isquêmico , Neurônios/metabolismo , Animais , Glucose/farmacologia , Hipocampo/citologia , Ionomicina/farmacologia , Ionóforos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Técnicas de Cultura de Órgãos , Oxigênio/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley
6.
J Neurochem ; 88(4): 878-84, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14756808

RESUMO

The effects of 30 s to 10 min hypoxia (PO2-10 mmHg) on glutamate receptor activity were studied in murine cortical neurons. Receptor activity was assessed as a rise in intracellular calcium concentration ([Ca2+]i) following a 10 s application of 1 mm glutamate or 100 micro mN-methy-d-aspartate (NMDA) in the presence of 0.1 mm Mg2+ and 10 micro m glycine. Change in [Ca2+]i elicited by glutamate increased 26% (n = 192, p < 0.001) and that to NMDA by 74% (n = 9, p < 0.01) during a 100-s period of hypoxia. After 10 min hypoxia, responses to glutamate were 62% smaller than those in normoxia, with increased basal intracellular [Ca2+]i predicting reduced receptor activity. When neurons were exposed to NMDA after 10 min of hypoxia, [Ca2+]i increases were 12% smaller than after 100 s hypoxia, but still 53% larger than in oxygenated neurons (n = 9, p = 0.01). Neurons expressed relatively similar amounts of NR2A, -B, -C, and -D subunits. The phosphorylation of NMDA NR1 subunits increased during hypoxia. Pre-treatment of neurons with a protein kinase C (PKC) inhibitor (chelerythrine, 10 micro m) prevented increases in N-methy-d-aspartate receptor (NMDAR) activity during hypoxia and reduced the phosphorylation of NR1 subunits. These results suggest that enhancement of glutamate receptor activity during the first minutes of hypoxia is mediated by phosphorylation of NMDARs by PKC and that other mechanisms, possibly involving intracellular calcium, limit glutamate receptor-mediated calcium influx during longer periods of hypoxia.


Assuntos
Cálcio/metabolismo , Córtex Cerebral/citologia , Neurônios/metabolismo , Oxigênio/metabolismo , Proteína Quinase C/metabolismo , Receptores de Glutamato/metabolismo , Alcaloides , Animais , Benzofenantridinas , Western Blotting , Hipóxia Celular , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Agonistas de Aminoácidos Excitatórios , Ácido Glutâmico/farmacologia , Imuno-Histoquímica , Hibridização In Situ , Camundongos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Fenantridinas/farmacologia , Fosforilação , Fatores de Tempo
7.
Neuroscience ; 118(1): 25-35, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12676134

RESUMO

Neonatal rats survive and avoid brain injury during periods of anoxia 25 times longer than adults. We hypothesized that oxygen activates and hypoxia suppresses NMDA receptor (NMDAR) responses in neonatal rat neurons, explaining the innate hypoxia tolerance of these cells. In CA1 neurons isolated from neonatal rat hippocampus (mean postnatal age [P] 5.8 days), hypoxia (PO(2) 10 mm Hg) reduced NMDA receptor-channel open-time percentage and NMDA-induced increase in [Ca(2+)](i) (NMDA DeltaCa(2+)) by 38 and 68% (P<0.01), respectively. In P20 neurons the reductions were not significant. In P3-10 CA1 neurons within intact hippocampal slices, hypoxia reduced NMDA DeltaCa(2+) by 52% (P=0.002) and decreased NMDA-induced death by 45% (P=0.004). Phalloidin, a microtubule stabilizer, prevented hypoxia-induced inhibition of NMDA DeltaCa(2+) in P3-10 neurons. To test whether NMDARs prevalent in neonates (NR1 plus NR2B or NR2D subunits) are inhibited by hypoxia compared with those in mature neurons (NR2A and NR2C), we expressed these receptors in Xenopus oocytes. Compared with responses in 21% O(2), hypoxia (PO(2) 17 mm Hg) reduced currents from neonatal type NR1/NR2D receptors by 25%, increased currents from NR1/NR2C by 18%, and had no effect on NR1/NR2A or NR1/NR2B. Modulation of NMDARs by hypoxia may play an important role in the hypoxia tolerance of the mammalian neonate. In addition, oxygen sensing by NMDARs could play a significant role in postnatal brain development.


Assuntos
Animais Recém-Nascidos/metabolismo , Asfixia Neonatal/metabolismo , Encéfalo/metabolismo , Hipóxia Encefálica/metabolismo , Neurônios/metabolismo , Oxigênio/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Envelhecimento/metabolismo , Animais , Asfixia Neonatal/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Cálcio/metabolismo , Hipocampo/metabolismo , Humanos , Recém-Nascido , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Oócitos/metabolismo , Técnicas de Cultura de Órgãos , Oxigênio/farmacologia , Faloidina/farmacologia , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Xenopus laevis
8.
Brain Res ; 958(1): 43-51, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12468029

RESUMO

The mechanism of the neuroprotective action of the glycolytic pathway intermediate fructose-1,6-bisphosphate (FBP) may involve activation of a phospholipase-C (PLC) dependent MAP kinase signaling pathway. In this study, we determined whether FBP's capacity to decrease delayed cell death in hippocampal slice cultures is dependent on PLC signaling or activation of the intracellular Ca(2+)-MEK/ERK neuroprotective signaling cascade. FBP (3.5 mM) reduced delayed death from oxygen/glucose deprivation in CA1, CA3 and dentate neurons in slice cultures. The phospholipase-C inhibitor U73122 and the MEK1/2 inhibitor U0126 prevented this protection. In hippocampal and cortical neurons, FBP increased phospho-ERK1/2 (p42/44) immunostaining during hypoxic, but not normoxic conditions. Increased phospho-ERK immunostaining was dependent on PLC and also on MEK 1/2, an upstream regulator of ERK. Further, we found that FBP enhancement of phospho-ERK immunostaining depended on [Ca(2+)](i): PLC inhibition and the IP(3) receptor blocker xestospongin C prevented FBP from increasing [Ca(2+)](i) and increasing phospho-ERK levels. However, while FBP-induced increases in [Ca(2+)](i) were blocked by xestospongin and a PLC inhibitor, [Ca(2+)](i) increases induced by the neuroprotective growth factor BDNF were not prevented. We conclude that during hypoxia FBP initiates a series of neuroprotective signals which include PLC activation, small increases in [Ca(2+)](i), and increased activity of the MEK/ERK signaling pathway.


Assuntos
Encéfalo/enzimologia , Frutosedifosfatos/metabolismo , Hipóxia-Isquemia Encefálica/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/enzimologia , Fármacos Neuroprotetores/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Frutosedifosfatos/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/fisiopatologia , Imuno-Histoquímica , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/metabolismo , MAP Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo
9.
Brain Res ; 917(2): 158-66, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11640901

RESUMO

The neuroprotectant fructose-1,6-bisphosphate (FBP) preserves cellular [ATP] and prevents catastrophic increases in [Ca2+]i during hypoxia. Because FBP does not enter neurons or glia, the mechanism of protection is not clear. In this study, we show that FBP's capacity to protect neurons and stabilize [Ca2+]i during hypoxia derives from signaling by a phospholipase-C-intracellular Ca2+-protein kinases pathway, rather than Ca2+ chelation or glutamate receptor inhibition. FBP reduced [Ca2+]i changes in hypoxic hippocampal neurons, regardless of [Ca2+]e, and preserved cellular integrity as measured by trypan blue or propidium iodide exclusion and [ATP]. FBP also prevented hypoxia-induced increases in [Ca2+]i when glucose was absent and when [Ca2+]e was increased to negate Ca2+ chelation by FBP. These protective effects were observed equally in postnatal day 2 (P2) and P16 neurons. Inhibiting glycolysis with iodoacetate eliminated the protective effects of FBP in P16 neurons. FBP did not alter Ca2+ influx stimulated by brief applications of NMDA or glutamate during normoxia or hypoxia, but did reduce the increase in [Ca2+]i produced by 10 min of glutamate exposure during hypoxia. Because FBP increases basal [Ca2+]i and stimulates membrane lipid hydrolysis, we tested whether FBP's protective action was dependent on phospholipase C signaling. The phospholipase C inhibitor U73122 prevented FBP-induced increases in [Ca2+]i and eliminated FBP's ability to stabilize [Ca2+]i and increase survival during anoxia. Similarly, FBP's protection was eliminated in the presence of the mitogen/extracellular signal protein kinase (MEK) inhibitor U0126. We conclude that FBP may produce neuroprotection via activation of neuroprotective signaling pathways that modulate Ca2+ homeostasis.


Assuntos
Isquemia Encefálica/metabolismo , Cálcio/metabolismo , Frutosedifosfatos/farmacologia , Hipóxia/metabolismo , Membranas Intracelulares/metabolismo , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Técnicas In Vitro , Camundongos , Concentração Osmolar , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/fisiologia
11.
J Neurosurg Anesthesiol ; 13(1): 19-25, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11145473

RESUMO

Cocktails of neuroprotectants acting at different parts of the ischemic injury cascade may have advantages over single agents. This study investigated, singly and in combination, the neuroprotective efficacy of an energy substrate (3.5 mM fructose 1,6-bisphosphate, FBP), an antagonist of NMDA receptors (1 and 10 microM MK-801), a free-radical scavenger (100 microM ascorbate), an adenosine A1 receptor agonist (10 microM 2-chloroadenosine), and an inhibitor of neurotransmission (2% isoflurane). These agents were evaluated for their ability to prevent loss and morphologic damage of CA1 neurons in rat hippocampal slices when these agents were administered during 30 minutes in vitro ischemia (combined oxygen/glucose deprivation at 37 degrees C) followed by 5 hours of recovery. Ten microM MK-801, alone or in combination with the other compounds, prevented loss of CA1 neurons and preserved their histologic appearance. Isoflurane, which prevents glutamate receptor-dependent cell death in this model, was also protective. Protection against neuron loss was also found when a subtherapeutic concentration of MK-801 (1 microM) was combined with 2-chloroadenosine (which indirectly causes NMDA receptor suppression), but not FBP or ascorbate. The authors conclude that in this model, the strategy of antagonizing NMDA receptors appears more protective than fructose-1,6-bisphosphate, 2-chloroadenosine or ascorbate.


Assuntos
Isquemia Encefálica/patologia , Hipocampo/patologia , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Glucose/deficiência , Hipocampo/efeitos dos fármacos , Hipocampo/ultraestrutura , Técnicas In Vitro , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley
12.
J Neurosci ; 20(10): 3522-8, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10804192

RESUMO

Hypoxia-induced suppression of NMDA receptors (NMDARs) in western painted turtle (Chrysemys picta) cortical neurons may be critical for surviving months of anoxic dormancy. We report that NMDARs are silenced by at least three different mechanisms operating at different times during anoxia. In pyramidal neurons from cerebrocortex, 1-8 min anoxia suppressed NMDAR activity (Ca(2+) influx and open probability) by 50-60%. This rapid decrease in receptor activity was controlled by activation of phosphatase 1 or 2A but was not associated with an increase in [Ca(2+)](i). However, during 2 hr of anoxia, [Ca(2+)](i) in cerebrocortical neurons increased by 35%, and suppression of NMDARs was predicted by the increase of [Ca(2+)](i) and controlled by calmodulin. An additional mechanism of NMDAR silencing, reversible removal of receptors from the cell membrane, was found in cerebrocortex of turtles remaining anoxic at 3 degrees C for 3-21 d. When suppression of NMDARs was prevented with phosphatase inhibitors, tolerance of anoxia was lost. Silencing of NMDARs is thus critical to the remarkable ability of C. picta to tolerate life without oxygen.


Assuntos
Regulação para Baixo/fisiologia , Hipóxia Encefálica/metabolismo , Neurônios/enzimologia , Receptores de N-Metil-D-Aspartato/metabolismo , Tartarugas/fisiologia , Adaptação Fisiológica/fisiologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Membrana Celular/química , Membrana Celular/metabolismo , Sobrevivência Celular/fisiologia , Córtex Cerebral/química , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Feminino , Masculino , Neurônios/química , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação
13.
Anesthesiology ; 92(5): 1343-9, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10781280

RESUMO

BACKGROUND: General anesthetics reduce neuron loss following focal cerebral ischemia in rodents. The relative efficacy of this action among different anesthetics clinically used for neuroprotection is uncertain. In addition, it remains unclear how anesthetics compare to neuroprotection afforded by mild hypothermia. This study was performed to evaluate the comparative effects of isoflurane, sodium pentothal, and mild hypothermia in a hippocampal slice model of cerebral ischemia and to determine if the mechanism of neuroprotection of isoflurane involves inhibition of glutamate excitotoxicity. METHODS: Survival and morphology of CA1, CA3, and dentate gyrus neurons in rat hippocampal slices were examined after 10 or 20 min of combined oxygen-glucose deprivation (in vitro ischemia) followed by a 5-h recovery period. RESULTS: 10 or 20 min in vitro ischemia at 37 degrees C killed 35-40% of neurons in CA1 (P < 0.001), 6% in CA3 (not significant) and 18% in dentate (P < 0.05). Isoflurane (0.7 and 2.0%, approximately 0.45 and 1.5 minimum alveolar concentration), pentothal (50 microm, approximately 1 minimum alveolar concentration equivalent) and mild hypothermia (34 degrees C) all reduced CA1 cell loss and morphologic damage to similar degrees in 10- and 20-min periods of ischemia (P < 0.001). The noncompetitive N-methyl-D-aspartate antagonist MK-801 prevented cell damage, showing that N-methyl-D-aspartate receptor activation is an important mechanism of injury in this model. Glutamate (1 mm) produced cell loss similar to in vitro ischemia. Isoflurane (2%) prevented cell damage from glutamate exposure. CONCLUSIONS: In hippocampal slices, neuron death from simulated ischemia was predominately due to activation of glutamate receptors. Isoflurane, sodium pentothal, an N-methyl-D-aspartate receptor antagonist, and mild hypothermia prevented cell death to similar degrees. For isoflurane, the mechanism appears to involve attenuation of glutamate excitotoxicity.


Assuntos
Anestésicos/uso terapêutico , Isquemia Encefálica/terapia , Hipocampo/efeitos dos fármacos , Hipotermia/metabolismo , Isoflurano/uso terapêutico , Neurônios/efeitos dos fármacos , Tiopental/uso terapêutico , Anestésicos/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Hipóxia/metabolismo , Isoflurano/farmacologia , Neurônios/citologia , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/efeitos dos fármacos , Tiopental/farmacologia
14.
Anesth Analg ; 89(4): 1040-5, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10512287

RESUMO

UNLABELLED: Volatile anesthetics may decrease synaptic transmission at central neurons by presynaptic and/or postsynaptic actions. Nonimmobilizers are volatile compounds with lipophilicities that suggest that they should (but do not) prevent motor responses to surgical stimuli. However, nonimmobilizers interfere with learning and memory, and, thus, might be predicted to depress synaptic transmission in areas of the brain mediating memory (e.g., hippocampal CA1 neurons). To test this possibility, we stimulated the Schaffer collaterals of rat hippocampal slices and recorded from stratum pyramidale of CA1 neurons. At approximately 0.5 MAC (MAC is the minimum alveolar anesthetic concentration at one standard atmosphere that is required to eliminate movement in response to noxious stimulation in 50% of subjects), halothane decreased population spike amplitude 37% +/- 21% (mean +/- SD), increased latency 15% +/- 9%, and decreased excitatory postsynaptic potentials 16% +/- 10%. In contrast, at concentrations below (0.4 times) predicted MAC, the nonimmobilizer, 1,2 dichlorohexafluorocyclobutane (2N), slightly (not significantly) increased population spike amplitude, decreased population spike latency 9% +/- 4%, and increased excitatory postsynaptic potentials 22% +/- 16%. At concentrations above (2 times) predicted MAC, 2N did not significantly increase population spike, decreased latency 10% +/- 4%, and did not significantly change excitatory postsynaptic potentials. At 0.1 predicted MAC, a second nonimmobilizer, perfluoropentane, tended (P = 0.05) to increase (11% +/- 9%) population spike amplitude, decreased population spike latency 8% +/- 2%, and tended (P = 0.06) to increase excitatory postsynaptic potentials (9% +/- 8%). We conclude that clinically relevant concentrations of halothane depress synaptic transmission at Schaffer collateral-CA1 synapses and that the nonimmobilizers 2N and perfluoropentane have no effect or are excitatory. The Schaffer collateral-CA1 synapse may serve as a useful model for the production of immobility by volatile anesthetics, but is flawed as a model for the capacity of volatile anesthetics to interfere with memory and learning. IMPLICATIONS: Halothane, but not the nonimmobilizers 1,2-dichlorohexafluorocyclobutane and perfluoropentane, inhibits hippocampal synaptic transmission at Schaffer collateral-CA1 synapses.


Assuntos
Anestésicos Inalatórios/farmacologia , Anestésicos/farmacologia , Clorofluorcarbonetos/farmacologia , Ciclobutanos/farmacologia , Fluorocarbonos/farmacologia , Halotano/farmacologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Anestésicos/administração & dosagem , Anestésicos Inalatórios/administração & dosagem , Animais , Clorofluorcarbonetos/administração & dosagem , Ciclobutanos/administração & dosagem , Modelos Animais de Doenças , Potenciais Evocados/efeitos dos fármacos , Fluorocarbonos/administração & dosagem , Halotano/administração & dosagem , Aprendizagem/efeitos dos fármacos , Memória/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Sinapses/efeitos dos fármacos
15.
Anesth Analg ; 88(5): 1168-74, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10320189

RESUMO

UNLABELLED: Anesthetics cause a reduction in excitatory neurotransmission that may be important in the mechanisms of in vivo anesthetic action. Because glutamate is the major excitatory neurotransmitter in mammalian brain, evaluation of anesthetic effects on induced glutamate release is relevant for studying this potential mechanism of anesthetic action. In the present study, we compared the effects of anesthetics and nonanesthetics (halogenated alkanes that disobey the Meyer-Overton hypothesis) on depolarization-evoked glutamate release. Glutamate released from rat cortical brain slices after chemically induced depolarization (50 mM KCl) was measured continuously using an enzymatic fluorescence assay. The effects of the volatile anesthetics isoflurane and enflurane were compared with the effects of the transitional compound 1,1,2-trichlorotrifluoroethane, the nonanesthetic compound 1,2-dichlorohexafluorocyclobutane, and other polyhalogenated alkanes. Tested concentrations included effective anesthetic concentrations for the anesthetics and transitional compounds, and concentrations predicted to be anesthetic based on lipid solubility for the nonanesthetics. Isoflurane dose-dependently reduced depolarization-evoked glutamate release in cortical brain slices. Isoflurane and enflurane at concentrations equivalent to 1 minimum alveolar anesthetic concentration (MAC) reduced the KCl-evoked release to 20% and 17% of control, respectively. The transitional compound 1,1,2-trichlorotrifluoroethane at 210 microM (approximately 1.2 MAC) reduced glutamate release to 47%, and the nonanesthetic 1,2-dichlorohexafluorocyclobutane increased glutamate release at 70 microM (approximately 3 MAC). These findings support the hypothesis that the modulation of excitatory neurotransmission might be responsible, in part, for in vivo anesthetic action. IMPLICATIONS: The volatile anesthetics isoflurane and enflurane reduce depolarization-evoked glutamate release in rat brain slices. The transitional compound 1,1,2-trichlorotrifluoroethane reduces glutamate release to a much lesser extent, and the nonanesthetic 1,2-dichlorohexafluorocyclobutane does not reduce glutamate release. These findings support the hypothesis that the modulation of excitatory neurotransmission might be responsible, in part, for in vivo anesthetic action.


Assuntos
Alcanos/farmacologia , Anestésicos Inalatórios/farmacologia , Córtex Cerebral/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Animais , Córtex Cerebral/metabolismo , Relação Dose-Resposta a Droga , Potenciais da Membrana , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley
16.
Anesthesiology ; 90(4): 1137-45, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10201687

RESUMO

BACKGROUND: An increase in intracellular calcium concentration ([Ca2+]i) in neurons has been proposed as an important effect of volatile anesthetics, because they alter signaling pathways that influence neurotransmission. However, the existing data for anesthetic-induced increases in [Ca2+]i conflict. METHODS: Changes in [Ca2+]i were measured using fura-2 fluorescence spectroscopy in rat cortical brain slices at 90, 185, 370, and 705 microM isoflurane. To define the causes of an increase in [Ca2+]i, slices were studied in Ca2+-free medium, in the presence of Ca2+-channel blockers, and in the presence of the Ca2+-release inhibitor azumolene. The authors compared the effect of the volatile anesthetic with that of the nonanesthetic compound 1,2-dichlorohexafluorocyclobutane. Single-dose experiments in CA1 neurons in hippocampal slices with halothane (360 microM) and in acutely dissociated CA1 neurons with halothane (360 microM) and isoflurane (445 microM) also were performed. RESULTS: Isoflurane at 0.5, 1, and 2 minimum alveolar concentrations increased basal [Ca2+]i in cortical slices in a dose-dependent manner (P < 0.05). This increase was not altered by Ca2+-channel blockers or Ca2+-free medium but was reduced 85% by azumolene. The nonanesthetic 1,2-dichlorohexafluorocyclobutane did not increase [Ca2+]i. In dissociated CA1 neurons, isoflurane reversibly increased basal [Ca2+]i by 15 nM (P < 0.05). Halothane increased [Ca2+]i in dissociated CA1 neurons and CA1 neurons in hippocampal slices by approximately 30 nM (P < 0.05). CONCLUSIONS: (1) Isoflurane and halothane reversibly increase [Ca2+]i in isolated neurons and in neurons within brain slices. (2) The increase in [Ca2+]i is caused primarily by release from intracellular stores. (3) Increases in [Ca2+]i occur with anesthetics but not with the nonanesthetic 1,2-dichlorohexafluorocyclobutane.


Assuntos
Anestésicos Inalatórios/farmacologia , Cálcio/metabolismo , Córtex Cerebral/efeitos dos fármacos , Halotano/farmacologia , Hipocampo/efeitos dos fármacos , Isoflurano/farmacologia , Animais , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Ratos , Ratos Sprague-Dawley
17.
Am J Physiol ; 275(1): R86-91, 1998 07.
Artigo em Inglês | MEDLINE | ID: mdl-9688964

RESUMO

Survival of brain anoxia during months of winter dormancy by the Western painted turtle, Chrysemys picta, may rely on inactivation of neuronal ion channels. During 2 h of anoxia, Ca2+ influx via the N-methyl-D-aspartate (NMDA) subtype of glutamate receptor decreases 30-40%, but it is not known if prolonged anoxic dormancy is associated with even more profound downregulation of this important channel. Because ionized Ca2+ in cerebrospinal fluid (CSF) increases five- to sixfold during prolonged anoxia, the potential for uncontrolled Ca2+ influx and neurotoxicity is increased. To study the regulation of NMDA receptor activity, we measured NMDA-mediated changes in intracellular Ca2+ (NMDA-DeltaCa2+) in turtle cerebrocortical sheets with fura 2. Turtles were kept in N2-bubbled aquariums for 2 h to 6 wk at 2-3 degrees C. NMDA-DeltaCa2+ decreased 60 +/- 14% (P < 0.05) after 2 h of anoxia and did not decrease further for 6 wk. Intracellular Ca2+ increased from 135 to 183 nM (P < 0.05) after 3 wk of anoxia and thereafter returned toward preanoxic levels. When NMDA receptor activity was assessed in artificial CSF containing the ions found in anoxic brain CSF (pH 7. 25, 69 mM lactate, 8.4 mM Ca2+, and 5.1 mM Mg2+), NMDA-DeltaCa2+ was twice control initially but was 21% less than in normoxic artificial CSF after the end of 6 wk, suggesting altered sensitivity of the NMDA receptor to ionized Ca2+ during prolonged anoxia. Regulation of NMDA receptor activity in turtle cerebrocortex during 6 wk of anoxia thus results in depression of NMDA receptor Ca2+ flux, despite a sixfold increase in ionized extracellular Ca2+.


Assuntos
Cálcio/metabolismo , Córtex Cerebral/metabolismo , Hipóxia/fisiopatologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/biossíntese , Animais , Regulação para Baixo , Valores de Referência , Fatores de Tempo , Tartarugas
18.
J Exp Biol ; 201(Pt 8): 1141-52, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9510526

RESUMO

Down-regulation of ion channel activity ('channel arrest'), which aids in preserving critical ion gradients in concert with greatly diminished energy production, is one important strategy by which anoxia-tolerant neurons adapt to O2 shortage. Channel arrest results in the elimination of action potentials and neurotransmission and also decreases the need for ion transport, which normally requires a large energy expenditure. Important targets of this down-regulation may be channels in which activity would otherwise result in the toxic increases in intracellular [Ca2+] characteristic of anoxia-sensitive mammalian neurons. In turtles, Na+ channels and the Ca2+-permeable ion channel of the N-methyl-d-aspartate (NMDA)-type glutamate receptor undergo down-regulation during anoxia. Inactivation of NMDA receptors during hypoxia occurs by a variety of mechanisms, including alterations in the phosphorylation state of ion channel subunits, Ca2+-dependent second messenger activation, changes in Ca2+-dependent polymerization/depolymerization of actin to postsynaptic receptors and activation of other G-protein-coupled receptors. Release of inhibitory neurotransmitters (e.g. gamma-aminobutyrate) and neuromodulators (e.g. adenosine) into the brain extracellular fluids may play an important role in the down-regulation of these and other types of ion channels.


Assuntos
Adaptação Fisiológica , Cálcio/metabolismo , Cálcio/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Vertebrados/fisiologia , Animais , Hipóxia Celular/fisiologia
19.
Brain Res Dev Brain Res ; 106(1-2): 57-69, 1998 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-9554954

RESUMO

Neurons in the neonatal mammalian brain survive greater degrees of hypoxic stress than those in the mature brain. To investigate how developmental changes in glutamate receptor-mediated neurotoxicity contribute to this difference, we measured hypoxia-evoked glutamate release, glutamate receptor contribution to hypoxia-evoked intracellular calcium changes, and survival of hypoxia-/ischemia-sensitive CA1 neurons in rat hippocampus. Glutamate release was measured by a fluorescence assay, calcium changes in CA1 neurons with fura-2, and cell viability using Nissl and fluorescence staining with calcein-AM/ethidium homodimer, all in 300-micron thick hippocampal slices from 3-30 post-natal day (PND) rats. Glutamate released from PND 3-7 slices during hypoxia (PO2 = 5 mmHg) was only one third that of PND 18-22 slices. In PND 3-7 slices, survival of CA1 neurons after 5 min of hypoxia and 6 h of recovery was significantly greater than in PND 18-22 slices (viability indices 0.60 and 0.28, respectively, (p < 0.05). Five min of anoxia significantly altered Nissl staining pattern and morphology of CA1 neurons in PND 18-22 but not PND 3-7 slices. Hypoxia (PO2 = 5 mm Hg) caused three to five times greater increases in [Ca2+]i in PND 18-22 slices than in PND 3-7 slices (p < 0.001). During re-oxygenation, [Ca2+]i returned to baseline in PND 3-7 slices, but remained elevated in PND 18-22 slices. Glutamate receptor-mediated calcium changes in CA1 during hypoxia were 33% and 62% of the total calcium change in PND 3-7 and PND 18-22 CA1, respectively. We conclude that survival of CA1 neurons in PND 3-7 slices following hypoxic stress is associated with smaller increases and enhanced recovery of [Ca2+]i, less accumulation of glutamate, and less glutamate receptor-mediated calcium influx than in PND 18-22 slices.


Assuntos
Cálcio/metabolismo , Hipóxia Celular/fisiologia , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Animais Recém-Nascidos , Sobrevivência Celular/fisiologia , Citosol/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , N-Metilaspartato/farmacologia , Neurônios/citologia , Ratos , Ratos Sprague-Dawley
20.
J Exp Biol ; 201(Pt 2): 289-97, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9405320

RESUMO

During normoxia, glutamate and the glutamate family of ion channels play a key role in mediating rapid excitatory synaptic transmission in the central nervous system. However, during hypoxia, intracellular [Ca2+] increases to neurotoxic levels, mediated largely by the N-methyl-D-aspartate (NMDA) subfamily of glutamate receptors. Adenosine has been shown to decrease the magnitude of the hypoxia-induced increase in [Ca2+]i in mammalian brain slices, delaying tissue injury. Turtle brain is remarkably tolerant of anoxia, maintaining a pre-anoxic [Ca2+]i while cerebral adenosine levels increase 12-fold. Employing cell-attached single-channel patch-clamp techniques, we studied the effect of adenosine (200 micromol l-1) and anoxia on NMDA receptor open probability (Popen) and current amplitude. After 60 min of anoxic perfusion, channel Popen decreased by 65 % (from 6.8+/-1.6 to 2.4+/-0.8 %) an effect that could also be achieved with a normoxic perfusion of 200 micromol l-1 adenosine (Popen decreased from 5.8+/-1.1 to 2.3+/-1.2 %). The inclusion of 10 micromol l-1 8-phenyltheophylline, an A1 receptor blocker, prevented the adenosine- and anoxia-induced decrease in Popen. Mean single-channel current amplitude remained at approximately 2.7+/-0.23 pA under all experimental conditions. To determine whether a change in the membrane potential could be part of the mechanism by which Popen decreases, membrane and threshold potential were measured following each experiment. Membrane potential did not change significantly under any condition, ranging from -76.8 to -80.6 mV. Therefore, during anoxia, NMDA receptors cannot be regulated by Mg2+ in a manner dependent on membrane potential. Threshold potentials did decrease significantly following 60 min of anoxic or adenosine perfusion (control -33.3+/-1.9 mV, anoxia -28.4+/-1.5 mV, adenosine -23.4+/-2.8 mV). We conclude that anoxia modulates NMDA receptor activity and that adenosine plays a key role in mediating this change. This is the first direct measurement of ion channel activity in anoxic turtle brain and demonstrates that ion channel regulation is part of the naturally evolved anoxic defence mechanism of this species.


Assuntos
Adenosina/farmacologia , Córtex Cerebral/metabolismo , Hipóxia/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Cálcio/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Magnésio/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/fisiologia , Tartarugas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA