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










Database
Language
Publication year range
1.
J Cereb Blood Flow Metab ; 32(11): 2033-43, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22805876

ABSTRACT

Anoxia or ischemia causes hyperexcitability and cell death in mammalian neurons. Conversely, in painted turtle brain anoxia increases γ-amino butyric acid (GABA)ergic suppression of spontaneous electrical activity, and cell death is prevented. To examine ischemia tolerance in turtle neurons, we treated cortical sheets with an in vitro mimic of the penumbral region of stroke-afflicted mammalian brain (ischemic solution, IS). We found that during IS perfusion, neuronal membrane potential (V(m)) and the GABA(A) receptor reversal potential depolarized to a similar steady state (-92 ± 2 to -28 ± 3 mV, and -75 ± 1 to -35 ± 3 mV, respectively), and whole-cell conductance (G(w)) increased >3-fold (from 4 ± 0.2 to 15 ± 1 nS). These neurons were electrically quiet and changes reversed after reperfusion. GABA receptor antagonism prevented the IS-mediated increase in G(w) and neurons exhibited enhanced electrical excitability and rapid and irreversible rundown of V(m) during reperfusion. These results suggest that inhibitory GABAergic mechanisms also suppress electrical activity in ischemic cortex. Indeed, after 4 hours of IS treatment neurons did not exhibit any apparent damage; while at 24 hours, only early indicators of apoptosis were present. We conclude that anoxia-tolerant turtle neurons are tolerant of exposure to a mammalian ischemic penumbral mimic solution.


Subject(s)
Brain Ischemia/pathology , Stroke/pathology , Turtles/physiology , Action Potentials/physiology , Animals , Cell Survival/physiology , Cerebral Cortex/pathology , Cerebrovascular Circulation/physiology , Disease Models, Animal , Hypoxia, Brain/pathology , Hypoxia, Brain/physiopathology , Membrane Potentials/physiology , Neurons/pathology , Neurotransmitter Agents/metabolism , Patch-Clamp Techniques , Potassium/pharmacology , Presynaptic Terminals/physiology , Pyramidal Cells/pathology , Solutions , gamma-Aminobutyric Acid/physiology
2.
Article in English | MEDLINE | ID: mdl-18519169

ABSTRACT

Neuronal membrane potential (E(m)) regulates the activity of excitatory voltage-sensitive channels. Anoxic insults lead to a severe loss of E(m) and excitotoxic cell death (ECD) in mammalian neurons. Conversely, anoxia-tolerant freshwater turtle neurons depress energy usage during anoxia by altering ionic conductance to reduce neuronal excitability and ECD is avoided. This wholesale alteration of ion channel and pump activity likely has a significant effect on E(m). Using the whole-cell patch clamp technique we recorded changes in E(m) from turtle cortical neurons during a normoxic to anoxic transition in the presence of various ion channel/pump modulators. E(m) did not change with normoxic perfusion but underwent a reversible, mild depolarization of 8.1+/-0.2 mV following anoxic perfusion. This mild anoxic depolarization (MAD) was not prevented by the manipulation of any single ionic conductance, but was partially reduced by pre-treatment with antagonists of GABA(A) receptors (5.7+/-0.5 mV), cellular bicarbonate production (5.3+/-0.2 mV) or K(+) channels (6.0+/-0.2 mV), or by perfusion of reactive oxygen species scavengers (5.2+/-0.3 mV). Furthermore, all of these treatments induced depolarization in normoxic neurons. Together these data suggest that the MAD may be due to the summation of numerous altered ion conductance states during anoxia.


Subject(s)
Hypoxia , Membrane Potentials , Neurons/metabolism , Animals , Bicarbonates/chemistry , Cell Death , Ion Channels/chemistry , Ions , Models, Biological , Oxygen/metabolism , Patch-Clamp Techniques , Potassium/chemistry , Reactive Oxygen Species , Receptors, GABA-A/chemistry , Turtles
3.
FEBS Lett ; 582(12): 1738-42, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18466771

ABSTRACT

Increased nitric oxide (NO) production from hypoxic mammalian neurons increases cerebral blood flow (CBF) but also glutamatergic excitotoxicity and DNA fragmentation. Anoxia-tolerant freshwater turtles have evolved NO-independent mechanisms to increase CBF; however, the mechanism(s) of NO regulation are not understood. In turtle cortex, anoxia or NMDAR blockade depressed NO production by 27+/-3% and 41+/-5%, respectively. NMDAR antagonists also reduced the subsequent anoxic decrease in NO by 74+/-6%, suggesting the majority of the anoxic decrease is due to endogenous suppression of NMDAR activity. Prevention of NO-mediated damage during the transition to and from anoxia may be incidental to natural reductions of NMDAR activity in the anoxic turtle cortex.


Subject(s)
Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Nitric Oxide/antagonists & inhibitors , Oxygen/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Turtles/metabolism , Anaerobiosis , Animals , Cerebrovascular Circulation , Nitric Oxide/biosynthesis
4.
Brain Res ; 1213: 27-34, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18455705

ABSTRACT

Adenosine is a defensive metabolite that is critical to anoxic neuronal survival in the freshwater turtle. Channel arrest of the N-methyl-d-aspartate receptor (NMDAR) is a hallmark of the turtle's remarkable anoxia tolerance and adenosine A1 receptor (A1R)-mediated depression of normoxic NMDAR activity is well documented. However, experiments examining the role of A1Rs in regulating NMDAR activity during anoxia have yielded inconsistent results. The aim of this study was to examine the role of A1Rs in the normoxic and anoxic regulation of turtle brain NMDAR activity. Whole-cell NMDAR currents were recorded for up to 2 h from turtle cortical pyramidal neurons exposed to pharmacological A1R or Gi protein modulation during normoxia (95% O(2)/5% CO2) and anoxia (95% N2/5% CO2). NMDAR currents were unchanged during normoxia and decreased 51+/-4% following anoxic exposure. Normoxic agonism of A1Rs with adenosine or N6-cyclopentyladenosine (CPA) decreased NMDAR currents 57+/-11% and 59+/-6%, respectively. The A1R antagonist 8-cyclopentyl-1,3-dimethylxanthine (DPCPX) had no effect on normoxic NMDAR currents and prevented the adenosine and CPA-mediated decreases in NMDAR activity. DPCPX partially reduced the anoxic decrease at 20 but not 40 min of treatment. The Gi protein inhibitor pertussis toxin (PTX) prevented both the CPA and anoxia-mediated decreases in NMDAR currents and calcium chelation or blockade of mitochondrial ATP-sensitive K+ channels also prevented the CPA-mediated decreases. Our results suggest that the long-term anoxic decrease in NMDAR activity is activated by a PTX-sensitive mechanism that is independent of A1R activity.


Subject(s)
Anorexia/pathology , Cerebral Cortex/pathology , Neurons/drug effects , Pertussis Toxin/pharmacology , Receptor, Adenosine A1/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Analysis of Variance , Animals , Anti-Arrhythmia Agents/pharmacology , Chelating Agents/pharmacology , Decanoic Acids/pharmacology , Drug Interactions , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Hydroxy Acids/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/metabolism , Patch-Clamp Techniques/methods , Time Factors , Turtles , Xanthines/pharmacology
5.
J Physiol ; 586(4): 1043-58, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18079161

ABSTRACT

Hypoxic mammalian neurons undergo excitotoxic cell death, whereas painted turtle neurons survive prolonged anoxia without apparent injury. Anoxic survival is possibly mediated by a decrease in N-methyl-d-aspartate receptor (NMDAR) activity and maintenance of cellular calcium concentrations ([Ca(2+)](c)) within a narrow range during anoxia. In mammalian ischaemic models, activation of mitochondrial ATP-sensitive K(+) (mK(ATP)) channels partially uncouples mitochondria resulting in a moderate increase in [Ca(2+)](c) and neuroprotection. The aim of this study was to determine the role of mK(ATP) channels in anoxic turtle NMDAR regulation and if mitochondrial uncoupling and [Ca(2+)](c) changes underlie this regulation. In isolated mitochondria, the K(ATP) channel activators diazoxide and levcromakalim increased mitochondrial respiration and decreased ATP production rates, indicating mitochondria were 'mildly' uncoupled by 10-20%. These changes were blocked by the mK(ATP) antagonist 5-hydroxydecanoic acid (5HD). During anoxia, [Ca(2+)](c) increased 9.3 +/- 0.3% and NMDAR currents decreased 48.9 +/- 4.1%. These changes were abolished by K(ATP) channel blockade with 5HD or glibenclamide, Ca(2+)(c) chelation with 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) or by activation of the mitochondrial Ca(2+) uniporter with spermine. Similar to anoxia, diazoxide or levcromakalim increased [Ca(2+)](c) 8.9 +/- 0.7% and 3.8 +/- 0.3%, while decreasing normoxic whole-cell NMDAR currents by 41.1 +/- 6.7% and 55.4 +/- 10.2%, respectively. These changes were also blocked by 5HD or glibenclamide, BAPTA, or spermine. Blockade of mitochondrial Ca(2+)-uptake decreased normoxic NMDAR currents 47.0 +/- 3.1% and this change was blocked by BAPTA but not by 5HD. Taken together, these data suggest mK(ATP) channel activation in the anoxic turtle cortex uncouples mitochondria and reduces mitochondrial Ca(2+) uptake via the uniporter, subsequently increasing [Ca(2+)](c) and decreasing NMDAR activity.


Subject(s)
Cerebral Cortex/physiology , KATP Channels/physiology , Mitochondria, Heart/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Turtles/physiology , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cromakalim/pharmacology , Decanoic Acids/pharmacology , Diazoxide/pharmacology , Glyburide/pharmacology , Hydroxy Acids/pharmacology , Hypoxia/physiopathology , Patch-Clamp Techniques , Potassium Channel Blockers
6.
Am J Physiol Regul Integr Comp Physiol ; 294(2): R606-13, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18056983

ABSTRACT

Without oxygen, all mammals suffer neuronal injury and excitotoxic cell death mediated by overactivation of the glutamatergic N-methyl-D-aspartate receptor (NMDAR). The western painted turtle can survive anoxia for months, and downregulation of NMDAR activity is thought to be neuroprotective during anoxia. NMDAR activity is related to the activity of another glutamate receptor, the alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR). AMPAR blockade is neuroprotective against anoxic insult in mammals, but the role of AMPARs in the turtle's anoxia tolerance has not been investigated. To determine whether AMPAR activity changes during hypoxia or anoxia in the turtle cortex, whole cell AMPAR currents, AMPAR-mediated excitatory postsynaptic potentials (EPSPs), and excitatory postsynaptic currents (EPSCs) were measured. The effect of AMPAR blockade on normoxic and anoxic NMDAR currents was also examined. During 60 min of normoxia, evoked peak AMPAR currents and the frequencies and amplitudes of EPSPs and EPSCs did not change. During anoxic perfusion, evoked AMPAR peak currents decreased 59.2 +/- 5.5 and 60.2 +/- 3.5% at 20 and 40 min, respectively. EPSP frequency (EPSP(f)) and amplitude decreased 28.7 +/- 6.4% and 13.2 +/- 1.7%, respectively, and EPSC(f) and amplitude decreased 50.7 +/- 5.1% and 51.3 +/- 4.7%, respectively. In contrast, hypoxic (Po(2) = 5%) AMPAR peak currents were potentiated 56.6 +/- 20.5 and 54.6 +/- 15.8% at 20 and 40 min, respectively. All changes were reversed by reoxygenation. AMPAR currents and EPSPs were abolished by 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX). In neurons pretreated with CNQX, anoxic NMDAR currents were reversibly depressed by 49.8 +/- 7.9%. These data suggest that AMPARs may undergo channel arrest in the anoxic turtle cortex.


Subject(s)
Adaptation, Physiological/physiology , Cerebral Cortex/physiology , Hypoxia/physiopathology , Receptors, AMPA/physiology , Turtles/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , Oxygen/pharmacology , Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate/physiology
7.
J Comp Physiol B ; 177(4): 473-81, 2007 May.
Article in English | MEDLINE | ID: mdl-17347830

ABSTRACT

The Western painted turtle survives months without oxygen. A key adaptation is a coordinated reduction of cellular ATP production and utilization that may be signaled by changes in the concentrations of reactive oxygen species (ROS) and cyclic nucleotides (cAMP and cGMP). Little is known about the involvement of cyclic nucleotides in the turtle's metabolic arrest and ROS have not been previously measured in any facultative anaerobes. The present study was designed to measure changes in these second messengers in the anoxic turtle. ROS were measured in isolated turtle brain sheets during a 40-min normoxic to anoxic transition. Changes in cAMP and cGMP were determined in turtle brain, pectoralis muscle, heart and liver throughout 4 h of forced submergence at 20-22 degrees C. Turtle brain ROS production decreased 25% within 10 min of cyanide or N(2)-induced anoxia and returned to control levels upon reoxygenation. Inhibition of electron transfer from ubiquinol to complex III caused a smaller decrease in [ROS]. Conversely, inhibition of complex I increased [ROS] 15% above controls. In brain [cAMP] decreased 63%. In liver [cAMP] doubled after 2 h of anoxia before returning to control levels with prolonged anoxia. Conversely, skeletal muscle and heart [cAMP] remained unchanged; however, skeletal muscle [cGMP] became elevated sixfold after 4 h of submergence. In liver and heart [cGMP] rose 41 and 127%, respectively, after 2 h of anoxia. Brain [cGMP] did not change significantly during 4 h of submergence. We conclude that turtle brain ROS production occurs primarily between mitochondrial complexes I and III and decreases during anoxia. Also, cyclic nucleotide concentrations change in a manner suggestive of a role in metabolic suppression in the brain and a role in increasing liver glycogenolysis.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Hypoxia/metabolism , Reactive Oxygen Species/metabolism , Turtles/physiology , Adaptation, Physiological/physiology , Animals , Cerebral Cortex/metabolism , Glycogenolysis/physiology , Hypoxia/physiopathology , Liver/metabolism , Oxygen/blood , Signal Transduction/physiology
8.
Article in English | MEDLINE | ID: mdl-16139540

ABSTRACT

Excitotoxic cell death (ECD) is characteristic of mammalian brain following min of anoxia, but is not observed in the western painted turtle following days to months without oxygen. A key event in ECD is a massive increase in intracellular Ca(2+) by over-stimulation of N-methyl-d-aspartate receptors (NMDARs). The turtle's anoxia tolerance may involve the prevention of ECD by attenuating NMDAR-induced Ca(2+) influx. The goal of this study was to determine if protein phosphatases (PPs) and intracellular calcium mediate reductions in turtle cortical neuron whole-cell NMDAR currents during anoxia, thereby preventing ECD. Whole-cell NMDAR currents did not change during 80 min of normoxia, but decreased 56% during 40 min of anoxia. Okadaic acid and calyculin A, inhibitors of serine/threonine PP1 and PP2A, potentiated NMDAR currents during normoxia and prevented anoxia-mediated attenuation of NMDAR currents. Decreases in NMDAR activity during anoxia were also abolished by inclusion of the Ca(2+) chelator -- BAPTA and the calmodulin inhibitor -- calmidazolium. However, cypermethrin, an inhibitor of the Ca(2+)/calmodulin-dependent PP2B (calcineurin), abolished the anoxic decrease in NMDAR activity at 20, but not 40 min suggesting that this phosphatase might play an early role in attenuating NMDAR activity during anoxia. Our results show that PPs, Ca(2+) and calmodulin play an important role in decreasing NMDAR activity during anoxia in the turtle cortex. We offer a novel mechanism describing this attenuation in which PP1 and 2A dephosphorylate the NMDAR (NR1 subunit) followed by calmodulin binding, a subsequent dissociation of alpha-actinin-2 from the NR1 subunit, and a decrease in NMDAR activity.


Subject(s)
Calcium/pharmacology , Cerebral Cortex/physiopathology , Hypoxia/physiopathology , Phosphoprotein Phosphatases/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Turtles/physiology , Animals , Calmodulin/antagonists & inhibitors , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Female , Imidazoles/pharmacology , Marine Toxins , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Patch-Clamp Techniques , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Phosphatase 1 , Pyrethrins/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...