Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(4): e0298065, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38626211

RESUMO

Anoxia in the mammalian brain leads to hyper-excitability and cell death; however, this cascade of events does not occur in the anoxia-tolerant brain of the western painted turtle, Chrysemys picta belli. The painted turtle has become an important anoxia-tolerant model to study brain, heart, and liver function in the absence of oxygen, but being anoxia-tolerant likely means that decapitation alone is not a suitable method of euthanasia. Many anesthetics have long-term effects on ion channels and are not appropriate for same day experimentation. Using whole-cell electrophysiological techniques, we examine the effects of the anesthetic, Alfaxalone, on pyramidal cell action potential amplitude, threshold, rise and decay time, width, frequency, whole cell conductance, and evoked GABAA receptors currents to determine if any of these characteristics are altered with the use of Alfaxalone for animal sedation. We find that Alfaxalone has no long-term impact on action potential parameters or whole-cell conductance. When acutely applied to naïve tissue, Alfaxalone did lengthen GABAA receptor current decay rates by 1.5-fold. Following whole-animal sedation with Alfaxalone, evoked whole cell GABAA receptor current decay rates displayed an increasing trend with 1 and 2 hours after brain sheet preparation, but showed no significant change after a 3-hour washout period. Therefore, we conclude that Alfaxalone is a suitable anesthetic for same day use in electrophysiological studies in western painted turtle brain tissue.


Assuntos
Anestésicos , Hipóxia Encefálica , Pregnanodionas , Tartarugas , Animais , Tartarugas/fisiologia , Receptores de GABA-A/metabolismo , Células Piramidais/metabolismo , Hipóxia/metabolismo , Anestésicos/farmacologia , Mamíferos
2.
FEBS Open Bio ; 14(4): 555-573, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38342633

RESUMO

Anesthetics have varying physiological effects, but most notably alter ion channel kinetics. Alfaxalone is a rapid induction and washout neuroactive anesthetic, which potentiates γ-aminobutyric acid (GABA)-activated GABAA receptor (GABAA-R) currents. This study aims to identify any long-term effects of alfaxalone sedation on pyramidal neuron action potential and GABAA-R properties, to determine if its impact on neuronal function can be reversed in a sufficiently short timeframe to allow for same-day electrophysiological studies in goldfish brain. The goldfish (Carassius auratus) is an anoxia-tolerant vertebrate and is a useful model to study anoxia tolerance mechanisms. The results show that alfaxalone sedation did not significantly impact action potential properties. Additionally, the acute application of alfaxalone onto naive brain slices caused the potentiation of whole-cell GABAA-R current decay time and area under the curve. Following whole-animal sedation with alfaxalone, a 3-h wash of brain slices in alfaxalone-free saline, with saline exchanged every 30 min, was required to remove any potentiating impact of alfaxalone on GABAA-R whole-cell currents. These results demonstrate that alfaxalone is an effective anesthetic for same-day electrophysiological experiments with goldfish brain slices.


Assuntos
Anestésicos , Pregnanodionas , Receptores de GABA-A , Animais , Receptores de GABA-A/fisiologia , Potenciais de Ação , Carpa Dourada/fisiologia , Ácido gama-Aminobutírico , Células Piramidais/fisiologia , Anestésicos/farmacologia , Hipóxia
3.
J Biol Chem ; 300(3): 105727, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38325739

RESUMO

Hypoxia is a significant source of metabolic stress that activates many cellular pathways involved in cellular differentiation, proliferation, and cell death. Hypoxia is also a major component in many human diseases and a known driver of many cancers. Despite the challenges posed by hypoxia, there are animals that display impressive capacity to withstand lethal levels of hypoxia for prolonged periods of time and thus offer a gateway to a more comprehensive understanding of the hypoxic response in vertebrates. The weakly electric fish genus Brachyhypopomus inhabits some of the most challenging aquatic ecosystems in the world, with some species experiencing seasonal anoxia, thus providing a unique system to study the cellular and molecular mechanisms of hypoxia tolerance. In this study, we use closely related species of Brachyhypopomus that display a range of hypoxia tolerances to probe for the underlying molecular mechanisms via hypoxia inducible factors (HIFs)-transcription factors known to coordinate the cellular response to hypoxia in vertebrates. We find that HIF1⍺ from hypoxia tolerant Brachyhypopomus species displays higher transactivation in response to hypoxia than that of intolerant species, when overexpressed in live cells. Moreover, we identified two SUMO-interacting motifs near the oxygen-dependent degradation and transactivation domains of the HIF1⍺ protein that appear to boost transactivation of HIF1, regardless of the genetic background. Together with computational analyses of selection, this shows that evolution of HIF1⍺ are likely to underlie adaptations to hypoxia tolerance in Brachyhypopomus electric fishes, with changes in two SUMO-interacting motifs facilitating the mechanism of this tolerance.


Assuntos
Peixe Elétrico , Subunidade alfa do Fator 1 Induzível por Hipóxia , Oxigênio , Animais , Ecossistema , Peixe Elétrico/genética , Peixe Elétrico/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Anaerobiose , Oxigênio/metabolismo
4.
Front Physiol ; 13: 1031264, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36523555

RESUMO

Skeletal muscle regulation is responsible for voluntary muscular movement in vertebrates. The genes of two essential proteins, teneurins and latrophilins (LPHN), evolving in ancestors of multicellular animals form a ligand-receptor pair, and are now shown to be required for skeletal muscle function. Teneurins possess a bioactive peptide, termed the teneurin C-terminal associated peptide (TCAP) that interacts with the LPHNs to regulate skeletal muscle contractility strength and fatigue by an insulin-independent glucose importation mechanism in rats. CRISPR-based knockouts and siRNA-associated knockdowns of LPHN-1 and-3 in the C2C12 mouse skeletal cell line shows that TCAP stimulates an LPHN-dependent cytosolic Ca2+ signal transduction cascade to increase energy metabolism and enhance skeletal muscle function via increases in type-1 oxidative fiber formation and reduce the fatigue response. Thus, the teneurin/TCAP-LPHN system is presented as a novel mechanism that regulates the energy requirements and performance of skeletal muscle.

5.
Artigo em Inglês | MEDLINE | ID: mdl-35288242

RESUMO

Symbiosis is found throughout nature, but perhaps nowhere is it more fundamental than mitochondria in all eukaryotes. Since mitochondria were discovered and mechanisms of oxygen reduction characterized, an understanding gradually emerged that these organelles were involved not just in the combustion of oxygen, but also in the sensing of oxygen. While multiple hypotheses exist to explain the mitochondrial involvement in oxygen sensing, key elements are developing that include potassium channels and reactive oxygen species. To understand how mitochondria contribute to oxygen sensing, it is informative to study a model system which is naturally adapted to survive extended periods without oxygen. Amongst air-breathing vertebrates, the most highly adapted are western painted turtles (Chrysemys picta bellii), which overwinter in ice-covered and anoxic water bodies. Through research of this animal, it was postulated that metabolic rate depression is key to anoxic survival and that mitochondrial regulation is a key aspect. When faced with anoxia, excitatory neurotransmitter receptors in turtle brain are inhibited through mitochondrial calcium release, termed "channel arrest". Simultaneously, inhibitory GABAergic signalling contributes to the "synaptic arrest" of excitatory action potential firing through a pathway dependent on mitochondrial depression of ROS generation. While many pathways are implicated in mitochondrial oxygen sensing in turtles, such as those of adenosine, ATP turnover, and gaseous transmitters, an apparent point of intersection is the mitochondria. In this review we will explore how an organelle that was critical for organismal complexity in an oxygenated world has also become a potentially important oxygen sensor.


Assuntos
Hipóxia , Tartarugas , Animais , Hipóxia/metabolismo , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Tartarugas/metabolismo
6.
J Exp Biol ; 224(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34498078

RESUMO

In most vertebrates, anoxia drastically reduces the production of the essential adenosine triphosphate (ATP) to power its many necessary functions, and, consequently, cell death occurs within minutes. However, some vertebrates, such as the painted turtle (Chrysemys picta bellii), have evolved the ability to survive months without oxygen by simultaneously decreasing ATP supply and demand, surviving the anoxic period without any apparent cellular damage. The impact of anoxia on the metabolic function of painted turtles has received a lot of attention. However, the impact of low temperature has received less attention and the interactive effect of anoxia and temperature even less. In the present study, we investigated the interactive impacts of reduced temperature and severe hypoxia on the electrophysiological properties of pyramidal neurons in painted turtle cerebral cortex. Our results show that an acute reduction in temperature from 20 to 5°C decreases membrane potential, action potential width and amplitude, and whole-cell conductance. Importantly, acute exposure to 5°C considerably slows membrane repolarization by voltage-gated K+ channels. Exposing pyramidal cells to severe hypoxia in addition to an acute temperature change slightly depolarized membrane potential but did not alter action potential amplitude or width and whole-cell conductance. These results suggest that acclimation to low temperatures, preceding severe environmental hypoxia, induces cellular responses in pyramidal neurons that facilitate survival under low oxygen concentrations. In particular, our results show that temperature acclimation invokes a change in voltage-gated K+ channel kinetics that overcomes the acute inhibition of the channel.


Assuntos
Tartarugas , Animais , Encéfalo , Temperatura Baixa , Hipóxia , Temperatura
7.
Metabolites ; 11(8)2021 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-34436502

RESUMO

Polymerization of actin filaments and microtubules constitutes a ubiquitous demand for cellular adenosine-5'-triphosphate (ATP) and guanosine-5'-triphosphate (GTP). In anoxia-tolerant animals, ATP consumption is minimized during overwintering conditions, but little is known about the role of cell structure in anoxia tolerance. Studies of overwintering mammals have revealed that microtubule stability in neurites is reduced at low temperature, resulting in withdrawal of neurites and reduced abundance of excitatory synapses. Literature for turtles is consistent with a similar downregulation of peripheral cytoskeletal activity in brain and liver during anoxic overwintering. Downregulation of actin dynamics, as well as modification to microtubule organization, may play vital roles in facilitating anoxia tolerance. Mitochondrial calcium release occurs during anoxia in turtle neurons, and subsequent activation of calcium-binding proteins likely regulates cytoskeletal stability. Production of reactive oxygen species (ROS) formation can lead to catastrophic cytoskeletal damage during overwintering and ROS production can be regulated by the dynamics of mitochondrial interconnectivity. Therefore, suppression of ROS formation is likely an important aspect of cytoskeletal arrest. Furthermore, gasotransmitters can regulate ROS levels, as well as cytoskeletal contractility and rearrangement. In this review we will explore the energetic costs of cytoskeletal activity, the cellular mechanisms regulating it, and the potential for cytoskeletal arrest being an important mechanism permitting long-term anoxia survival in anoxia-tolerant species, such as the western painted turtle and goldfish.

8.
J Exp Biol ; 224(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-34047778

RESUMO

Goldfish are one of a few species able to avoid cellular damage during month-long periods in severely hypoxic environments. By suppressing action potentials in excitatory glutamatergic neurons, the goldfish brain decreases its overall energy expenditure. Coincident with reductions in O2 availability is a natural decrease in cellular reactive oxygen species (ROS) generation, which has been proposed to function as part of a low-oxygen signal transduction pathway. Using live-tissue fluorescence microscopy, we found that ROS production decreased by 10% with the onset of anoxia in goldfish telencephalic brain slices. Employing whole-cell patch-clamp recording, we found that, similar to severe hypoxia, the ROS scavengers N-acetyl cysteine (NAC) and MitoTEMPO, added during normoxic periods, depolarized membrane potential (severe hypoxia -73.6 to -61.4 mV, NAC -76.6 to -66.2 mV and MitoTEMPO -71.5 mV to -62.5 mV) and increased whole-cell conductance (severe hypoxia 5.7 nS to 8.0 nS, NAC 6.0 nS to 7.5 nS and MitoTEMPO 6.0 nS to 7.6 nS). Also, in a subset of active pyramidal neurons, these treatments reduced action potential firing frequency (severe hypoxia 0.18 Hz to 0.03 Hz, NAC 0.27 Hz to 0.06 Hz and MitoTEMPO 0.35 Hz to 0.08 Hz). Neither severe hypoxia nor ROS scavenging impacted action potential threshold. The addition of exogenous hydrogen peroxide could reverse the effects of the antioxidants. Taken together, this supports a role for a reduction in [ROS] as a low-oxygen signal in goldfish brain.


Assuntos
Carpa Dourada , Hipóxia , Animais , Oxigênio , Técnicas de Patch-Clamp , Células Piramidais , Espécies Reativas de Oxigênio
9.
J Neurophysiol ; 125(2): 321-330, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33296606

RESUMO

Mammalian neurons undergo rapid excitotoxic cell death when deprived of oxygen; however, the common goldfish (Carassius auratus) has the unique ability of surviving in oxygen-free waters, under anoxia. This organism utilizes γ-amino butyric acid (GABA) signaling to suppress excitatory glutamatergic activity during anoxic periods. Although GABAA receptor antagonists are not deleterious to the cellular survival, coinhibition of GABAA and GABAB receptors is detrimental by abolishing anoxia-induced neuroprotective mechanisms. Here we show that blocking the anoxic GABAergic neurotransmission induces seizure-like activity (SLA) analogous to a paroxysmal depolarization shift (PDS), with hyperpolarization of action potential (AP) threshold and elevation of threshold currents. The observed PDS was attributed to an increase in excitatory postsynaptic currents (EPSCs) that are normally attenuated with decreasing oxygen levels. Furthermore, for the first time, we show that in addition to PDS, some neurons undergo depolarization block and do not generate AP despite a suprathreshold membrane potential. In conclusion, our results indicate that with severe hypoxia and absence of GABA receptor activity, telencephalic neurons of C. auratus manifest a paroxysmal depolarization shift, a key feature of epileptic discharge.NEW & NOTEWORTHY This work shows that the combination of anoxia and inhibition of GABA receptors induces seizure-like activities in goldfish telencephalic pyramidal and stellate neurons. Importantly, to prevent seizure-like activity, an intact GABA-mediated inhibitory pathway is required.


Assuntos
Potenciais de Ação , Antagonistas GABAérgicos/farmacologia , Hipóxia/fisiopatologia , Neurônios/fisiologia , Animais , Proteínas de Peixes/metabolismo , Carpa Dourada , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oxigênio/metabolismo , Receptores de GABA/metabolismo
10.
J Neurophysiol ; 122(2): 466-479, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31141433

RESUMO

The western painted turtle (Chrysemys picta bellii) has the unique ability of surviving several months in the absence of oxygen, which is termed anoxia. One major protective strategy that the turtle employs during anoxia is a reduction in neuronal electrical activity, which may result from a natural reduction in reactive oxygen species (ROS). We previously linked a reduction in ROS levels to an increase in γ-amino butyric acid (GABA) receptor currents. The purpose of this study is to understand how fast-spiking, GABA-releasing neurons respond to reductions in ROS and how this affects GABA release. Using a fluorescence-coupled enzymatic microplate assay for GABA, we found that anoxia, the ROS scavenger N-(2-mercaptopriopionyl)glycine (MPG), or the mitochondria-specific ROS scavenger MitoTEMPO resulted in a 2.5-, 2.0-, and 2.5-fold increase in extracellular GABA concentration, respectively. This phenomenon could be blocked with TTX, indicating that it is activity dependent. Using whole cell patch-clamping techniques, we found that fast-spiking, burst-firing GABAergic turtle neurons increase the duration and number of action potentials per burst by 26% and 42%, respectively, in response to ROS scavenging via MPG. These results suggest that the reduction in mitochondrially produced ROS that occurs during anoxia leads to increased GABA release, which promotes postsynaptic inhibitory activity through activation of GABA receptors.NEW & NOTEWORTHY This is a novel study examining the response of cerebral cortical stellate interneurons to anoxia and mitochondrial reactive oxygen species (ROS) scavenging with MitoTEMPO. Under both conditions burst firing increases in these cells, and we show that extracellular GABA release increases in the presence of the ROS scavenger. We conclude that in the anoxia-tolerant painted turtle brain, a decrease in ROS levels is an important low oxygen signal.


Assuntos
Córtex Cerebral/fisiologia , Neurônios GABAérgicos/fisiologia , Hipóxia/metabolismo , Interneurônios/fisiologia , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Tartarugas/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Córtex Cerebral/metabolismo , Neurônios GABAérgicos/metabolismo , Interneurônios/metabolismo , Técnicas de Patch-Clamp , Tartarugas/metabolismo
11.
FEBS Open Bio ; 9(4): 571-581, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30984533

RESUMO

The western painted turtle (Chrysemys picta bellii) can survive extended periods of anoxia via a series of mechanisms that serve to reduce its energetic needs. Central to these mechanisms is the response of mitochondria, which depolarize in response to anoxia in turtle pyramidal neurons due to an influx of K+. It is currently unknown how mitochondrial matrix pH is affected by this response and we hypothesized that matrix pH acidifies during anoxia due to increased K+/H+ exchanger activity. Inhibition of K+/H+ exchange via quinine led to a collapse of mitochondrial membrane potential (Ψm) during oxygenated conditions in turtle cortical neurons, as indicated by rhodamine-123 fluorescence, and this occurred twice as quickly during anoxia which indicates an elevation in K+ conductance. Mitochondrial matrix pH acidified during anoxia, as indicated by SNARF-1 fluorescence imaged via confocal microscopy, and further acidification occurred during anoxia when the F1Fo-ATPase was inhibited with oligomycin-A, indicating that ΔpH collapse is prevented during anoxic conditions. Collectively, these results indicate that the mitochondrial proton electrochemical gradient is actively preserved during anoxia to prevent a collapse of Ψm and ΔpH.


Assuntos
Mitocôndrias/química , ATPases Mitocondriais Próton-Translocadoras/genética , Canais de Potássio/metabolismo , Células Piramidais/fisiologia , Proteínas de Répteis/genética , Tartarugas/fisiologia , Anaerobiose , Animais , Concentração de Íons de Hidrogênio , Potencial da Membrana Mitocondrial/fisiologia , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Antiportadores de Potássio-Hidrogênio/metabolismo , Proteínas de Répteis/metabolismo
12.
J Exp Biol ; 221(Pt 21)2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30237241

RESUMO

Unlike anoxia-intolerant mammals, painted turtles can survive extended periods without oxygen. This is partly accomplished by an anoxia-mediated increase in gamma-aminobutyric acid (GABA) release, which activates GABA receptors and mediates spike arrest in turtle neurons via shunting inhibition. Extracellular taurine levels also increase during anoxia; why this occurs is unknown but it is speculated that glycine and/or GABAA/B receptors are involved. Given the general importance of inhibitory neurotransmission in the anoxia-tolerant painted turtle brain, we investigated the function of taurine as an inhibitory neuromodulator in turtle pyramidal neurons. Using whole-cell patch-clamp electrophysiological methods to record from neurons within a cortical brain sheet, we found that taurine depolarized membrane potential by ∼8 mV, increased whole-cell conductance ∼2-fold, and induced an inward current that possessed characteristics similar to GABA- and glycine-evoked currents. These effects were mitigated following glycine receptor antagonism with strychnine and GABAA receptor antagonism with gabazine, bicuculine or picrotoxin, but were unchanged following GABAB or glutamatergic receptor inhibition. These data indicate that a high concentration of taurine in vitro mediates its effects through both glycine and GABAA receptors, and suggests that taurine, in addition to GABA, inhibits neuronal activity during anoxia in the turtle cortex.


Assuntos
Células Piramidais/fisiologia , Receptores de GABA-A/fisiologia , Receptores de Glicina/fisiologia , Taurina/farmacologia , Tartarugas/fisiologia , Potenciais de Ação/fisiologia , Anaerobiose , Animais , Técnicas de Patch-Clamp/veterinária , Células Piramidais/efeitos dos fármacos , Proteínas de Répteis/fisiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-29223874

RESUMO

The ion channel arrest hypothesis has been the foundation of three decades of research into the underlying mechanisms of hypoxia/anoxia tolerance in several key species, including: painted turtles, goldfish, crucian carp, naked mole rats, and arctic and ground squirrels. The hypothesis originally stated that hypoxia/anoxia tolerant species ought to have fewer ion channels per area membrane and/or mechanisms to regulate the conductance of ion channels. Today we can add to this and include mechanisms to remove channels from membranes and the expression of low conductance isoforms. Furthermore, possible oxygen sensing mechanisms in brain include a link to mitochondrial function, changes in the concentration of intracellular Ca2+ and reactive oxygen species, and activation of protein kinase C and a phosphatase. Importantly ion channel arrest leads to a decrease in metabolic rate that is fundamental to survival without oxygen and in brain is reflected in decreased action potential frequency or spike arrest. This results not only from a decrease in excitatory glutamatergic receptor currents but also by an increase in inhibitory GABAergic receptor currents. The surprising finding that ionic conductance through some ion channels increases is novel and contrary to the ion channel arrest hypothesis. The major insight that this offers is that key regulatory events are occurring at the level of the synapse and we therefore propose the "synaptic arrest hypothesis".


Assuntos
Potenciais de Ação , Hipóxia Encefálica/metabolismo , Sinapses/metabolismo , Transmissão Sináptica , Animais , Canais Iônicos/metabolismo , Mitocôndrias/metabolismo , Proteína Quinase C/metabolismo , Receptores de Glutamato/metabolismo
14.
J Exp Biol ; 220(Pt 4): 695-704, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27923876

RESUMO

With oxygen deprivation, the mammalian brain undergoes hyper-activity and neuronal death while this does not occur in the anoxia-tolerant goldfish (Carassius auratus). Anoxic survival of the goldfish may rely on neuromodulatory mechanisms to suppress neuronal hyper-excitability. As γ-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the brain, we decided to investigate its potential role in suppressing the electrical activity of goldfish telencephalic neurons. Utilizing whole-cell patch-clamp recording, we recorded the electrical activities of both excitatory (pyramidal) and inhibitory (stellate) neurons. With anoxia, membrane potential (Vm) depolarized in both cell types from -72.2 mV to -57.7 mV and from -64.5 mV to -46.8 mV in pyramidal and stellate neurons, respectively. While pyramidal cells remained mostly quiescent, action potential frequency (APf) of the stellate neurons increased 68-fold. Furthermore, the GABAA receptor reversal potential (E-GABA) was determined using the gramicidin perforated-patch-clamp method and found to be depolarizing in pyramidal (-53.8 mV) and stellate neurons (-42.1 mV). Although GABA was depolarizing, pyramidal neurons remained quiescent as EGABA was below the action potential threshold (-36 mV pyramidal and -38 mV stellate neurons). Inhibition of GABAA receptors with gabazine reversed the anoxia-mediated response. While GABAB receptor inhibition alone did not affect the anoxic response, co-antagonism of GABAA and GABAB receptors (gabazine and CGP-55848) led to the generation of seizure-like activities in both neuron types. We conclude that with anoxia, Vm depolarizes towards EGABA which increases APf in stellate neurons and decreases APf in pyramidal neurons, and that GABA plays an important role in the anoxia tolerance of goldfish brain.


Assuntos
Potenciais de Ação , Proteínas de Peixes/metabolismo , Carpa Dourada/fisiologia , Oxigênio/metabolismo , Células Piramidais/metabolismo , Receptores de GABA-A/metabolismo , Receptores de GABA-B/metabolismo , Potenciais de Ação/efeitos dos fármacos , Anaerobiose , Animais , Antagonistas de Receptores de GABA-A/farmacologia , Antagonistas de Receptores de GABA-B/farmacologia , Humanos , Hipóxia/metabolismo , Técnicas de Patch-Clamp , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Telencéfalo/citologia , Telencéfalo/fisiologia
15.
Artigo em Inglês | MEDLINE | ID: mdl-27664385

RESUMO

The pond snail Lymnaea stagnalis is reported to be anoxia-tolerant and if the tolerance mechanism is similar to that of the anoxia-tolerant painted turtle, GABA should play an important role. A potentially confounding factor investigating the role of GABA in anoxia tolerance are reports that GABA has both inhibitory and excitatory effects within L. stagnalis central ganglion. We therefore set out to determine if seasonality or photoperiod has an impact on: 1) the anoxia-tolerance of the intact pond snail, and 2) the response of isolated neuroganglia cluster F neurons to exogenous GABA application. L. stagnalis maintained on a natural summer light cycle were unable to survive any period of anoxic exposure, while those maintained on a natural winter light cycle survived a maximum of 4h. Using intracellular sharp electrode recordings from pedal ganglia cluster F neurons we show that there is a photoperiod dependent shift in the response to GABA. Snails exposed to a 16h:8h light:dark cycle in an environmental chamber (induced summer phenotype) exhibited hyperpolarizing inhibitory responses and those exposed to a 8h:16h light:dark cycle (induced winter phenotype) exhibited depolarizing excitatory responses to GABA application. Using gramicidin-perforated patch recordings we also found a photoperiod dependent shift in the reversal potential for GABA. We conclude that the opposing responses of L. stagnalis central neurons to GABA results from a shift in intracellular chloride concentration that is photoperiod dependent and is likely mediated through the relative efficacy of cation chloride co-transporters. Although the physiological ramifications of the photoperiod dependent shift are unknown this work potentially has important implications for the impact of artificial light pollution on animal health.


Assuntos
Neurônios GABAérgicos/fisiologia , Gânglios dos Invertebrados/fisiologia , Lymnaea/fisiologia , Receptores de GABA-A/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Hipóxia Celular , Polaridade Celular/efeitos dos fármacos , Cloro/metabolismo , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Agonistas de Receptores de GABA-A/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/efeitos dos fármacos , Gânglios dos Invertebrados/citologia , Gânglios dos Invertebrados/efeitos dos fármacos , Gramicidina/farmacologia , Técnicas In Vitro/veterinária , Ionóforos/farmacologia , Lymnaea/citologia , Microdissecção/veterinária , Técnicas de Patch-Clamp/veterinária , Fotoperíodo , Receptores de GABA-A/química , Estações do Ano , Transdução de Sinais/efeitos dos fármacos , Ácido gama-Aminobutírico/química
16.
Artigo em Inglês | MEDLINE | ID: mdl-27280321

RESUMO

Neurons from the western painted turtle (Chrysemys picta bellii) are remarkably resilient to anoxia. This is partly due to a reduction in the permeability of excitatory glutamatergic ion channels, initiated by mitochondrial ATP-sensitive K(+) (mK(+)ATP) channel activation. The aim of this study was to determine if: 1) PKCε, a kinase associated with hypoxic stress tolerance, is more highly expressed in turtle brain than the anoxia-intolerant rat brain; 2) PKCε translocates to the mitochondrial membrane during anoxia; 3) PKCε modulates mK(+)ATP channels at the Thr-224 phosphorylation site on the Kir6.2 subunit; and 4) Thr-224 phosphorylation sensitises mK(+)ATP channels to anoxia. Soluble and mitochondrial-rich particulate fractions of turtle and rat cerebral cortex were isolated and PKCε expression was determined by Western blot, which revealed that turtle cortical PKCε expression was half that of the rat. Following the transition to anoxia, no changes in PKCε expression in either the soluble or particulate fraction of the turtle cortex were observed. Furthermore, incubation of tissue with tat-conjugated activator or inhibitor peptides had no effect on the amount of PKCε in either fraction. However, we observed a 2-fold increase in Thr-224 phosphorylation following 1h of anoxia. The increased Thr-224 phosphorylation was blocked by the general kinase inhibitor staurosporine but this did not affect the latency or magnitude of mK(+)ATP channel-mediated mitochondrial depolarization following anoxia, as indicated by rhodamine-123. We conclude that PKCε does not play a role in the onset of mitochondrial depolarization and therefore glutamatergic channel arrest in turtle cerebral cortex.


Assuntos
Encéfalo/citologia , Mitocôndrias/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteína Quinase C-épsilon/metabolismo , Tartarugas , Animais , Encéfalo/metabolismo , Regulação Enzimológica da Expressão Gênica , Hipóxia/metabolismo , Mitocôndrias/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/química , Proteína Quinase C-épsilon/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Ratos , Solubilidade , Treonina/metabolismo
17.
Ann N Y Acad Sci ; 1365(1): 43-58, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25959851

RESUMO

Surviving hypoxia is one of the most critical challenges faced by vertebrates. Most species have adapted to changing levels of oxygen in their environment with specialized organs that sense hypoxia, while only few have been uniquely adapted to survive prolonged periods of anoxia. The goal of this review is to present the most recent research on oxygen sensing, adaptation to hypoxia, and mechanisms of anoxia tolerance in nonmammalian vertebrates. We discuss the respiratory structures in fish, including the skin, gills, and air-breathing organs, and recent evidence for chemosensory neuroepithelial cells (NECs) in these tissues that initiate reflex responses to hypoxia. The use of the zebrafish as a genetic and developmental model has allowed observation of the ontogenesis of respiratory and chemosensory systems, demonstration of a putative intracellular O2 sensor in chemoreceptors that may initiate transduction of the hypoxia signal, and investigation into the effects of extreme hypoxia on cardiorespiratory development. Other organisms, such as goldfish and freshwater turtles, display a high degree of anoxia tolerance, and these models are revealing important adaptations at the cellular level, such as the regulation of glutamatergic and GABAergic neurotransmission in defense of homeostasis in central neurons.


Assuntos
Adaptação Fisiológica/fisiologia , Células Quimiorreceptoras/metabolismo , Hipóxia/metabolismo , Animais , Brânquias/metabolismo , Homeostase/fisiologia , Humanos , Hipóxia/fisiopatologia , Pele/metabolismo , Pele/fisiopatologia , Peixe-Zebra
18.
PLoS One ; 10(7): e0131669, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26147940

RESUMO

Painted turtles are the most anoxia-tolerant tetrapods known, capable of surviving without oxygen for more than four months at 3°C and 30 hours at 20°C. To investigate the transcriptomic basis of this ability, we used RNA-seq to quantify mRNA expression in the painted turtle ventricle and telencephalon after 24 hours of anoxia at 19°C. Reads were obtained from 22,174 different genes, 13,236 of which were compared statistically between treatments for each tissue. Total tissue RNA contents decreased by 16% in telencephalon and 53% in ventricle. The telencephalon and ventricle showed ≥ 2x expression (increased expression) in 19 and 23 genes, respectively, while only four genes in ventricle showed ≤ 0.5x changes (decreased expression). When treatment effects were compared between anoxic and normoxic conditions in the two tissue types, 31 genes were increased (≥ 2x change) and 2 were decreased (≤ 0.5x change). Most of the effected genes were immediate early genes and transcription factors that regulate cellular growth and development; changes that would seem to promote transcriptional, translational, and metabolic arrest. No genes related to ion channels, synaptic transmission, cardiac contractility or excitation-contraction coupling changed. The generalized expression pattern in telencephalon and across tissues, but not in ventricle, correlated with the predicted metabolic cost of transcription, with the shortest genes and those with the fewest exons showing the largest increases in expression.


Assuntos
Ventrículos do Coração/fisiopatologia , Hipóxia/genética , Telencéfalo/fisiologia , Transcriptoma/genética , Tartarugas/genética , Animais , Expressão Gênica/genética , Ventrículos do Coração/metabolismo , Hipóxia/metabolismo , RNA Mensageiro/genética , Telencéfalo/metabolismo , Fatores de Transcrição/genética
19.
J Physiol ; 593(10): 2311-26, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25781154

RESUMO

Anoxia induces hyper-excitability and cell death in mammalian brain but in the anoxia-tolerant western painted turtle (Chrysemys picta bellii) neuronal electrical activity is suppressed (i.e. spike arrest), adenosine triphosphate (ATP) consumption is reduced, and cell death does not occur. Electrical suppression is primarily the result of enhanced γ-aminobutyric acid (GABA) transmission; however, the underlying mechanism responsible for initiating oxygen-sensitive GABAergic spike arrest is unknown. In turtle cortical pyramidal neurons there are three types of GABA(A) receptor-mediated currents: spontaneous inhibitory postsynaptic currents (IPSCs), giant IPSCs and tonic currents. The aim of this study was to assess the effects of reactive oxygen species (ROS) scavenging on these three currents since ROS levels naturally decrease with anoxia and may serve as a redox signal to initiate spike arrest. We found that anoxia, pharmacological ROS scavenging, or inhibition of mitochondrial ROS generation enhanced all three types of GABA currents, with tonic currents comprising ∼50% of the total current. Application of hydrogen peroxide inhibited all three GABA currents, demonstrating a reversible redox-sensitive signalling mechanism. We conclude that anoxia-mediated decreases in mitochondrial ROS production are sufficient to initiate a redox-sensitive inhibitory GABA signalling cascade that suppresses electrical activity when oxygen is limited. This unique strategy for reducing neuronal ATP consumption during anoxia represents a natural mechanism in which to explore therapies to protect mammalian brain from low-oxygen insults.


Assuntos
Potenciais de Ação/fisiologia , Hipóxia/fisiopatologia , Mitocôndrias/metabolismo , Células Piramidais/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de GABA-A/fisiologia , Tartarugas/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Feminino , Peróxido de Hidrogênio/farmacologia , Masculino , Oxirredução , Técnicas de Patch-Clamp , Espécies Reativas de Oxigênio/farmacologia , Receptores de GABA-A/efeitos dos fármacos , Transdução de Sinais/fisiologia , Potenciais Sinápticos/fisiologia
20.
Proteomics ; 15(9): 1587-97, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25583675

RESUMO

During anoxia, overall protein synthesis is almost undetectable in the brain of the western painted turtle. The aim of this investigation was to address the question of whether there are alterations to specific proteins by comparing the normoxic and anoxic brain proteomes. Reductions in creatine kinase, hexokinase, glyceraldehyde-3-phosphate dehydrogenase, and pyruvate kinase reflected the reduced production of adenosine triphosphate (ATP) during anoxia while the reduction in transitional endoplasmic reticulum ATPase reflected the conservation of ATP or possibly a decrease in intracellular Ca(2+). In terms of neural protection programed cell death 6 interacting protein (PDCD6IP; a protein associated with apoptosis), dihydropyrimidinase-like protein, t-complex protein, and guanine nucleotide protein G(o) subunit alpha (Go alpha; proteins associated with neural degradation and impaired cognitive function) also declined. A decline in actin, gelsolin, and PDCD6IP, together with an increase in tubulin, also provided evidence for the induction of a neurological repair response. Although these proteomic alterations show some similarities with the crucian carp (another anoxia-tolerant species), there are species-specific responses, which supports the theory of no single strategy for anoxia tolerance. These findings also suggest the anoxic turtle brain could be an etiological model for investigating mammalian hypoxic damage and clinical neurological disorders.


Assuntos
Encéfalo/fisiologia , Hipóxia/metabolismo , Proteoma/metabolismo , Tartarugas/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Encéfalo/citologia , Cognição , Glicólise , Hipóxia/fisiopatologia , Proteoma/análise , Proteômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...