Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nature ; 632(8023): 139-146, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38961289

RESUMO

Brain computation performed by billions of nerve cells relies on a sufficient and uninterrupted nutrient and oxygen supply1,2. Astrocytes, the ubiquitous glial neighbours of neurons, govern brain glucose uptake and metabolism3,4, but the exact mechanisms of metabolic coupling between neurons and astrocytes that ensure on-demand support of neuronal energy needs are not fully understood5,6. Here we show, using experimental in vitro and in vivo animal models, that neuronal activity-dependent metabolic activation of astrocytes is mediated by neuromodulator adenosine acting on astrocytic A2B receptors. Stimulation of A2B receptors recruits the canonical cyclic adenosine 3',5'-monophosphate-protein kinase A signalling pathway, leading to rapid activation of astrocyte glucose metabolism and the release of lactate, which supplements the extracellular pool of readily available energy substrates. Experimental mouse models involving conditional deletion of the gene encoding A2B receptors in astrocytes showed that adenosine-mediated metabolic signalling is essential for maintaining synaptic function, especially under conditions of high energy demand or reduced energy supply. Knockdown of A2B receptor expression in astrocytes led to a major reprogramming of brain energy metabolism, prevented synaptic plasticity in the hippocampus, severely impaired recognition memory and disrupted sleep. These data identify the adenosine A2B receptor as an astrocytic sensor of neuronal activity and show that cAMP signalling in astrocytes tunes brain energy metabolism to support its fundamental functions such as sleep and memory.


Assuntos
Adenosina , Astrócitos , Encéfalo , Metabolismo Energético , Glucose , Plasticidade Neuronal , Neurônios , Receptor A2B de Adenosina , Transdução de Sinais , Animais , Astrócitos/metabolismo , Adenosina/metabolismo , Camundongos , Encéfalo/metabolismo , Encéfalo/citologia , Masculino , Receptor A2B de Adenosina/metabolismo , Receptor A2B de Adenosina/deficiência , Glucose/metabolismo , Neurônios/metabolismo , Sono/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ácido Láctico/metabolismo , Hipocampo/metabolismo , Hipocampo/citologia , Feminino , AMP Cíclico/metabolismo , Camundongos Endogâmicos C57BL
2.
J Physiol ; 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38843467

RESUMO

The brain requires an uninterrupted supply of oxygen and nutrients to support the high metabolic needs of billions of nerve cells processing information. In low oxygen conditions, increases in cerebral blood flow maintain brain oxygen delivery, but the cellular and molecular mechanisms responsible for dilation of cerebral blood vessels in response to hypoxia are not fully understood. This article presents a systematic review and analysis of data reported in studies of these mechanisms. Our primary outcome measure was the percent reduction of the cerebrovascular response to hypoxia in conditions of pharmacological or genetic blockade of specific signaling mechanisms studied in experimental animals or in humans. Selection criteria were met by 28 articles describing the results of animal studies and six articles describing the results of studies conducted in humans. Selected studies investigated the potential involvement of various neurotransmitters, neuromodulators, vasoactive molecules and ion channels. Of all the experimental conditions, blockade of adenosine-mediated signaling and inhibition of ATP-sensitive potassium (KATP) channels had the most significant effect in reducing the cerebrovascular response to hypoxia (by 49% and 37%, respectively). Various degree reductions of the hypoxic response were also reported in studies which investigated the roles of nitric oxide, arachidonic acid derivates, catecholamines and hydrogen sulphide, amongst others. However, definitive conclusions about the importance of these signaling pathways cannot be drawn from the results of this analysis. In conclusion, there is significant evidence that one of the key mechanisms of hypoxic cerebral vasodilation (accounting for ∼50% of the response) involves the actions of adenosine and modulation of vascular KATP channels. However, recruitment of other vasodilatory signaling mechanisms is required for the full expression of the cerebrovascular response to hypoxia.

3.
Mol Cell Neurosci ; 124: 103806, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36592801

RESUMO

Previously, we have shown that purinergic signalling is involved in the control of hyperosmotic-induced sympathoexcitation at the level of the PVN, via activation of P2X receptors. However, the source(s) of ATP that drives osmotically-induced increases in sympathetic outflow remained undetermined. Here, we tested the two competing hypotheses that either (1) higher extracellular ATP in PVN during salt loading (SL) is a result of a failure of ectonucleotidases to metabolize ATP; and/or (2) SL can stimulate PVN astrocytes to release ATP. Rats were salt loaded with a 2 % NaCl solution replacing drinking water up to 4 days, an experimental model known to cause a gradual increase in blood pressure and plasma osmolarity. Immunohistochemical assessment of glial-fibrillary acidic protein (GFAP) revealed increased glial cell reactivity in the PVN of rats after 4 days of high salt exposure. ATP and adenosine release measurements via biosensors in hypothalamic slices showed that baseline ATP release was increased 17-fold in the PVN while adenosine remained unchanged. Disruption of Ca2+-dependent vesicular release mechanisms in PVN astrocytes by virally-driven expression of a dominant-negative SNARE protein decreased the release of ATP. The activity of ectonucleotidases quantified in vitro by production of adenosine from ATP was increased in SL group. Our results showed that SL stimulates the release of ATP in the PVN, at least in part, from glial cells by a vesicle-mediated route and likely contributes to the neural control of circulation during osmotic challenges.


Assuntos
Núcleo Hipotalâmico Paraventricular , Cloreto de Sódio , Ratos , Animais , Núcleo Hipotalâmico Paraventricular/metabolismo , Cloreto de Sódio/metabolismo , Cloreto de Sódio na Dieta/metabolismo , Astrócitos/metabolismo , Trifosfato de Adenosina/metabolismo , Adenosina
4.
J Neurosci ; 40(49): 9364-9371, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33122390

RESUMO

Mechanosensitivity is a well-known feature of astrocytes, however, its underlying mechanisms and functional significance remain unclear. There is evidence that astrocytes are acutely sensitive to decreases in cerebral perfusion pressure and may function as intracranial baroreceptors, tuned to monitor brain blood flow. This study investigated the mechanosensory signaling in brainstem astrocytes, as these cells reside alongside the cardiovascular control circuits and mediate increases in blood pressure and heart rate induced by falls in brain perfusion. It was found that mechanical stimulation-evoked Ca2+ responses in astrocytes of the rat brainstem were blocked by (1) antagonists of connexin channels, connexin 43 (Cx43) blocking peptide Gap26, or Cx43 gene knock-down; (2) antagonists of TRPV4 channels; (3) antagonist of P2Y1 receptors for ATP; and (4) inhibitors of phospholipase C or IP3 receptors. Proximity ligation assay demonstrated interaction between TRPV4 and Cx43 channels in astrocytes. Dye loading experiments showed that mechanical stimulation increased open probability of carboxyfluorescein-permeable membrane channels. These data suggest that mechanosensory Ca2+ responses in astrocytes are mediated by interaction between TRPV4 and Cx43 channels, leading to Cx43-mediated release of ATP which propagates/amplifies Ca2+ signals via P2Y1 receptors and Ca2+ recruitment from the intracellular stores. In astrocyte-specific Cx43 knock-out mice the magnitude of heart rate responses to acute increases in intracranial pressure was not affected by Cx43 deficiency. However, these animals displayed lower heart rates at different levels of cerebral perfusion, supporting the hypothesis of connexin hemichannel-mediated release of signaling molecules by astrocytes having an excitatory action on the CNS sympathetic control circuits.SIGNIFICANCE STATEMENT There is evidence suggesting that astrocytes may function as intracranial baroreceptors that play an important role in the control of systemic and cerebral circulation. To function as intracranial baroreceptors, astrocytes must possess a specialized membrane mechanism that makes them exquisitely sensitive to mechanical stimuli. This study shows that opening of connexin 43 (Cx43) hemichannels leading to the release of ATP is the key central event underlying mechanosensory Ca2+ responses in astrocytes. This astroglial mechanism plays an important role in the autonomic control of heart rate. These data add to the growing body of evidence suggesting that astrocytes function as versatile surveyors of the CNS metabolic milieu, tuned to detect conditions of potential metabolic threat, such as hypoxia, hypercapnia, and reduced perfusion.


Assuntos
Astrócitos/fisiologia , Mecanotransdução Celular/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Tronco Encefálico/citologia , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Circulação Cerebrovascular/fisiologia , Conexina 43/antagonistas & inibidores , Conexina 43/genética , Feminino , Frequência Cardíaca/fisiologia , Masculino , Mecanotransdução Celular/efeitos dos fármacos , Camundongos , Camundongos Knockout , Peptídeos/antagonistas & inibidores , Peptídeos/genética , Estimulação Física , Ratos , Receptores Purinérgicos P2Y1/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética
5.
J Neurosci ; 40(15): 3052-3062, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32132265

RESUMO

Maintenance of cardiorespiratory homeostasis depends on autonomic reflexes controlled by neuronal circuits of the brainstem. The neurophysiology and neuroanatomy of these reflex pathways are well understood, however, the mechanisms and functional significance of autonomic circuit modulation by glial cells remain largely unknown. In the experiments conducted in male laboratory rats we show that astrocytes of the nucleus of the solitary tract (NTS), the brain area that receives and integrates sensory information from the heart and blood vessels, respond to incoming afferent inputs with [Ca2+]i elevations. Astroglial [Ca2+]i responses are triggered by transmitters released by vagal afferents, glutamate acting at AMPA receptors and 5-HT acting at 5-HT2A receptors. In conscious freely behaving animals blockade of Ca2+-dependent vesicular release mechanisms in NTS astrocytes by virally driven expression of a dominant-negative SNARE protein (dnSNARE) increased baroreflex sensitivity by 70% (p < 0.001). This effect of compromised astroglial function was specific to the NTS as expression of dnSNARE in astrocytes of the ventrolateral brainstem had no effect. ATP is considered the principle gliotransmitter and is released by vesicular mechanisms blocked by dnSNARE expression. Consistent with this hypothesis, in anesthetized rats, pharmacological activation of P2Y1 purinoceptors in the NTS decreased baroreflex gain by 40% (p = 0.031), whereas blockade of P2Y1 receptors increased baroreflex gain by 57% (p = 0.018). These results suggest that glutamate and 5-HT, released by NTS afferent terminals, trigger Ca2+-dependent astroglial release of ATP to modulate baroreflex sensitivity via P2Y1 receptors. These data add to the growing body of evidence supporting an active role of astrocytes in brain information processing.SIGNIFICANCE STATEMENT Cardiorespiratory reflexes maintain autonomic balance and ensure cardiovascular health. Impaired baroreflex may contribute to the development of cardiovascular disease and serves as a robust predictor of cardiovascular and all-cause mortality. The data obtained in this study suggest that astrocytes are integral components of the brainstem mechanisms that process afferent information and modulate baroreflex sensitivity via the release of ATP. Any condition associated with higher levels of "ambient" ATP in the NTS would be expected to decrease baroreflex gain by the mechanism described here. As ATP is the primary signaling molecule of glial cells (astrocytes, microglia), responding to metabolic stress and inflammatory stimuli, our study suggests a plausible mechanism of how the central component of the baroreflex is affected in pathological conditions.


Assuntos
Astrócitos/fisiologia , Barorreflexo/fisiologia , Núcleo Solitário/fisiologia , Trifosfato de Adenosina/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Masculino , Neurônios Aferentes/metabolismo , Neurotransmissores/metabolismo , Neurotransmissores/fisiologia , Agonistas do Receptor Purinérgico P2Y/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor 5-HT2A de Serotonina/efeitos dos fármacos , Receptores de AMPA/efeitos dos fármacos , Receptores Purinérgicos P2Y1/efeitos dos fármacos , Proteínas SNARE/fisiologia , Serotonina/farmacologia , Estimulação do Nervo Vago
6.
Basic Res Cardiol ; 116(1): 32, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33942194

RESUMO

Stroke remains one of the most common causes of death and disability worldwide. Several preclinical studies demonstrated that the brain can be effectively protected against ischaemic stroke by two seemingly distinct treatments: remote ischaemic conditioning (RIC), involving cycles of ischaemia/reperfusion applied to a peripheral organ or tissue, or by systemic administration of glucagon-like-peptide-1 (GLP-1) receptor (GLP-1R) agonists. The mechanisms underlying RIC- and GLP-1-induced neuroprotection are not completely understood. In this study, we tested the hypothesis that GLP-1 mediates neuroprotection induced by RIC and investigated the effect of GLP-1R activation on cerebral blood vessels, as a potential mechanism of GLP-1-induced protection against ischaemic stroke. A rat model of ischaemic stroke (90 min of middle cerebral artery occlusion followed by 24-h reperfusion) was used. RIC was induced by 4 cycles of 5 min left hind limb ischaemia interleaved with 5-min reperfusion periods. RIC markedly (by ~ 80%) reduced the cerebral infarct size and improved the neurological score. The neuroprotection established by RIC was abolished by systemic blockade of GLP-1R with a specific antagonist Exendin(9-39). In the cerebral cortex of GLP-1R reporter mice, ~ 70% of cortical arterioles displayed GLP-1R expression. In acute brain slices of the rat cerebral cortex, activation of GLP-1R with an agonist Exendin-4 had a strong dilatory effect on cortical arterioles and effectively reversed arteriolar constrictions induced by metabolite lactate or oxygen and glucose deprivation, as an ex vivo model of ischaemic stroke. In anaesthetised rats, Exendin-4 induced lasting increases in brain tissue PO2, indicative of increased cerebral blood flow. These results demonstrate that neuroprotection against ischaemic stroke established by remote ischaemic conditioning is mediated by a mechanism involving GLP-1R signalling. Potent dilatory effect of GLP-1R activation on cortical arterioles suggests that the neuroprotection in this model is mediated via modulation of cerebral blood flow and improved brain perfusion.


Assuntos
Arteríolas/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Membro Posterior/irrigação sanguínea , Incretinas/farmacologia , Infarto da Artéria Cerebral Média/prevenção & controle , Precondicionamento Isquêmico , AVC Isquêmico/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Arteríolas/metabolismo , Arteríolas/fisiopatologia , Modelos Animais de Doenças , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , AVC Isquêmico/metabolismo , AVC Isquêmico/fisiopatologia , Masculino , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional
7.
Glia ; 67(12): 2264-2278, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31318482

RESUMO

The electrogenic sodium bicarbonate cotransporter 1, NBCe1 (SLC4A4), is the major bicarbonate transporter expressed in astrocytes. It is highly sensitive for bicarbonate and the main regulator of intracellular, extracellular, and synaptic pH, thereby modulating neuronal excitability. However, despite these essential functions, the molecular mechanisms underlying NBCe1-mediated astrocytic response to extracellular pH changes are mostly unknown. Using primary mouse cortical astrocyte cultures, we investigated the effect of long-term extracellular metabolic alkalosis on regulation of NBCe1 and elucidated the underlying molecular mechanisms by immunoblotting, biotinylation of surface proteins, intracellular H+ recording using the H+ -sensitive dye 2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein, and phosphoproteomic analysis. The results showed significant downregulation of NBCe1 activity following metabolic alkalosis without influencing protein abundance or surface expression of NBCe1. During alkalosis, the rate of intracellular H+ changes upon challenging NBCe1 was decreased in wild-type astrocytes, but not in cortical astrocytes from NBCe1-deficient mice. Alkalosis-induced decrease of NBCe1 activity was rescued after activation of mTOR signaling. Moreover, mass spectrometry revealed constitutively phosphorylated S255-257 and mutational analysis uncovered these residues being crucial for NBCe1 transport activity. Our results demonstrate a novel mTOR-regulated mechanism by which NBCe1 functional expression is regulated. Such mechanism likely applies not only for NBCe1 in astrocytes, but in epithelial cells as well.


Assuntos
Astrócitos/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Simportadores de Sódio-Bicarbonato/biossíntese , Serina-Treonina Quinases TOR/fisiologia , Alcalose/metabolismo , Alcalose/patologia , Animais , Células Cultivadas , Feminino , Expressão Gênica , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/fisiologia , Simportadores de Sódio-Bicarbonato/genética
9.
J Cell Physiol ; 234(1): 89-99, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-30132845

RESUMO

Sodium plays a major role in different astrocytic functions, including maintenance of ion homeostasis and uptake of neurotransmitters and metabolites, which are mediated by different Na+ -coupled transporters. In the current study, the role of an electrogenic sodium-bicarbonate cotransporter (NBCe1), a sodium-potassium-chloride transporter 1 (NKCC1) and sodium-potassium ATPase (Na+ -K+ -ATPase) for the maintenance of [Na+ ]i was investigated in cultured astrocytes of wild-type (WT) and of NBCe1-deficient (NBCe1-KO) mice using the Na+ -sensitive dye, asante sodium green-2. Our results suggest that cytosolic Na+ was higher in the presence of CO2 /HCO3- (15 mM) than CO2 /HCO3- -free, HEPES-buffered solution in WT, but not in NBCe1-KO astrocytes (12 mM). Surprisingly, there was a strong dependence of cytosolic [Na+ ] on the extracellular [HCO3- ] attributable to NBCe1 activity. Pharmacological blockage of NKCC1 with bumetanide led to a robust drop in cytosolic Na+ in both WT and NBCe1-KO astrocytes by up to 6 mM. There was a strong dependence of the cytosolic [Na+ ] on the extracellular [K+ ]. Inhibition of the Na+ -K+ -ATPase led to larger increase in cytosolic Na+ , both in the absence of K+ as compared with the presence of ouabain and in NBCe1-KO astrocytes as compared with WT astrocytes. Our results show that cytosolic Na+ in mouse cortical astrocytes can vary considerably and depends greatly on the concentrations of HCO3- and K+ , attributable to the activity of the Na+ -K+ -ATPase, of NBCe1 and NKCC1.


Assuntos
Simportadores de Sódio-Bicarbonato/genética , ATPase Trocadora de Sódio-Potássio/genética , Sódio/metabolismo , Membro 2 da Família 12 de Carreador de Soluto/genética , Animais , Astrócitos/metabolismo , Bicarbonatos/metabolismo , Transporte Biológico/genética , Córtex Cerebral/metabolismo , Citosol/metabolismo , Camundongos , Camundongos Knockout , Ouabaína/farmacologia , Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores
10.
Glia ; 66(6): 1185-1199, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29274121

RESUMO

Astrocytes support neuronal function by providing essential structural and nutritional support, neurotransmitter trafficking and recycling and may also contribute to brain information processing. In this article we review published results and report new data suggesting that astrocytes function as versatile metabolic sensors of central nervous system (CNS) milieu and play an important role in the maintenance of brain metabolic homeostasis. We discuss anatomical and functional features of astrocytes that allow them to detect and respond to changes in the brain parenchymal levels of metabolic substrates (oxygen and glucose), and metabolic waste products (carbon dioxide). We report data suggesting that astrocytes are also sensitive to circulating endocrine signals-hormones like ghrelin, glucagon-like peptide-1 and leptin, that have a major impact on the CNS mechanisms controlling food intake and energy balance. We discuss signaling mechanisms that mediate communication between astrocytes and neurons and consider how these mechanisms are recruited by astrocytes activated in response to various metabolic challenges. We review experimental data suggesting that astrocytes modulate the activities of the respiratory and autonomic neuronal networks that ensure adaptive changes in breathing and sympathetic drive in order to support the physiological and behavioral demands of the organism in ever-changing environmental conditions. Finally, we discuss evidence suggesting that altered astroglial function may contribute to the pathogenesis of disparate neurological, respiratory and cardiovascular disorders such as Rett syndrome and systemic arterial hypertension.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Animais , Humanos
11.
J Neurosci ; 36(42): 10750-10758, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27798130

RESUMO

Ventral regions of the medulla oblongata of the brainstem are populated by astrocytes sensitive to physiological changes in PCO2/[H+]. These astrocytes respond to decreases in pH with elevations in intracellular Ca2+ and facilitated exocytosis of ATP-containing vesicles. Released ATP propagates Ca2+ excitation among neighboring astrocytes and activates neurons of the brainstem respiratory network triggering adaptive increases in breathing. The mechanisms linking increases in extracellular and/or intracellular PCO2/[H+] with Ca2+ responses in chemosensitive astrocytes remain unknown. Fluorescent imaging of changes in [Na+]i and/or [Ca2+]i in individual astrocytes was performed in organotypic brainstem slice cultures and acute brainstem slices of adult rats. It was found that astroglial [Ca2+]i responses triggered by decreases in pH are preceded by Na+ entry, markedly reduced by inhibition of Na+/HCO3- cotransport (NBC) or Na+/Ca2+ exchange (NCX), and abolished in Na+-free medium or by combined NBC/NCX blockade. Acidification-induced [Ca2+]i responses were also dramatically reduced in brainstem astrocytes of mice deficient in the electrogenic Na+/HCO3- cotransporter NBCe1. Sensitivity of astrocytes to changes in pH was not affected by inhibition of Na+/H+ exchange or blockade of phospholipase C. These results suggest that in pH-sensitive astrocytes, acidification activates NBCe1, which brings Na+ inside the cell. Raising [Na+]i activates NCX to operate in a reverse mode, leading to Ca2+ entry followed by activation of downstream signaling pathways. Coupled NBC and NCX activities are, therefore, suggested to be responsible for functional CO2/H+ sensitivity of astrocytes that contribute to homeostatic regulation of brain parenchymal pH and control of breathing. SIGNIFICANCE STATEMENT: Brainstem astrocytes detect physiological changes in pH, activate neurons of the neighboring respiratory network, and contribute to the development of adaptive respiratory responses to the increases in the level of blood and brain PCO2/[H+]. The mechanisms underlying astroglial pH sensitivity remained unknown and here we show that in brainstem astrocytes acidification activates Na+/HCO3- cotransport, which brings Na+ inside the cell. Raising [Na+]i activates the Na+/Ca2+ exchanger to operate in a reverse mode leading to Ca2+ entry. This identifies a plausible mechanism of functional CO2/H+ sensitivity of brainstem astrocytes, which play an important role in homeostatic regulation of brain pH and control of breathing.


Assuntos
Astrócitos/efeitos dos fármacos , Dióxido de Carbono/farmacologia , Hidrogênio/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Astrócitos/metabolismo , Bicarbonatos/metabolismo , Sinalização do Cálcio , Exocitose , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Ratos , Respiração , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sódio/metabolismo , Simportadores de Sódio-Bicarbonato/antagonistas & inibidores , Simportadores de Sódio-Bicarbonato/genética , Simportadores de Sódio-Bicarbonato/metabolismo , Trocador de Sódio e Cálcio/antagonistas & inibidores , Trocador de Sódio e Cálcio/metabolismo
12.
J Physiol ; 595(8): 2569-2585, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-27981578

RESUMO

KEY POINTS: The present study suggests that the electrogenic sodium-bicarbonate cotransporter, NBCe1, supported by carbonic anhydrase II, CAII, provides an efficient mechanism of bicarbonate sensing in cortical astrocytes. This mechanism is proposed to play a major role in setting the pHi responses to extracellular acid/base challenges in astrocytes. A decrease in extracellular [HCO3- ] during isocapnic acidosis and isohydric hypocapnia, or an increase in intracellular [HCO3- ] during hypercapnic acidosis, was effectively sensed by NBCe1, which carried bicarbonate out of the cells under these conditions, and caused an acidification and sodium fall in WT astrocytes, but not in NBCe1-knockout astrocytes. Isocapnic acidosis, hypercapnic acidosis and isohydric hypocapnia evoked inward currents in NBCe1- and CAII-expressing Xenopus laevis oocytes, but not in native oocytes, suggesting that NBCe1 operates in the outwardly directed mode under these conditions consistent with our findings in astrocytes. We propose that bicarbonate sensing of astrocytes may have functional significance during extracellular acid/base disturbances in the brain, as it not only alters intracellular pH/[HCO3- ]-dependent functions of astrocytes, but also modulates the extracellular pH/[HCO3- ] in brain tissue. ABSTRACT: Extracellular acid/base status of the mammalian brain undergoes dynamic changes during many physiological and pathological events. Although intracellular pH (pHi ) of astrocytes responds to extracellular acid/base changes, the mechanisms mediating these changes have remained unresolved. We have previously shown that the electrogenic sodium-bicarbonate cotransporter, NBCe1, is a high-affinity bicarbonate carrier in cortical astrocytes. In the present study, we investigated whether NBCe1 plays a role in bicarbonate sensing in astrocytes, and in determining the pHi responses to extracellular acid/base challenges. We measured changes in intracellular H+ and Na+ in astrocytes from wild-type (WT) and from NBCe1-knockout (KO) mice, using ion-selective dyes, during isocapnic acidosis, hypercapnic acidosis and hypocapnia. We also analysed NBCe1-mediated membrane currents in Xenopus laevis oocytes under similar conditions. Comparing WT and NBCe1-KO astrocytes, we could dissect the contribution of NBCe1, of diffusion of CO2 across the cell membrane and, after blocking carbonic anhydrase (CA) activity with ethoxyzolamide, of the role of CA, for the amplitude and rate of acid/base fluxes. Our results suggest that NBCe1 transport activity in astrocytes, supported by CA activity, renders astrocytes bicarbonate sensors in the mouse cortex. NBCe1 carried bicarbonate into and out of the cell by sensing the variations of transmembrane [HCO3- ], irrespective of the changes in intra- and extracellular pH, and played a major role in setting pHi responses to the extracellular acid/base challenges. We propose that bicarbonate sensing of astrocytes may have potential functional significance during extracellular acid/base alterations in the brain.


Assuntos
Astrócitos/metabolismo , Bicarbonatos/metabolismo , Córtex Cerebral/metabolismo , Antiportadores de Cloreto-Bicarbonato/metabolismo , Líquido Extracelular/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Bicarbonatos/farmacologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Líquido Extracelular/efeitos dos fármacos , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Xenopus laevis
13.
J Biol Chem ; 291(36): 19108-17, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27422823

RESUMO

Glycolysis is the primary step for major energy production in the cell. There is strong evidence suggesting that glucose consumption and rate of glycolysis are highly modulated by cytosolic pH/[H(+)], but those can also be stimulated by an increase in the intracellular [HCO3 (-)]. Because proton and bicarbonate shift concomitantly, it remained unclear whether enhanced glucose consumption and glycolytic rate were mediated by the changes in intracellular [H(+)] or [HCO3 (-)]. We have asked whether glucose metabolism is enhanced by either a fall in intracellular [H(+)] or a rise in intracellular [HCO3 (-)], or by both, in mammalian astrocytes. We have recorded intracellular glucose in mouse astrocytes using a FRET-based nanosensor, while imposing different intracellular [H(+)] and [CO2]/[HCO3 (-)]. Glucose consumption and glycolytic rate were augmented by a fall in intracellular [H(+)], irrespective of a concomitant rise or fall in intracellular [HCO3 (-)]. Transport of HCO3 (-) into and out of astrocytes by the electrogenic sodium bicarbonate cotransporter (NBCe1) played a crucial role in causing changes in intracellular pH and [HCO3 (-)], but was not obligatory for the pH-dependent changes in glucose metabolism. Our results clearly show that it is the cytosolic pH that modulates glucose metabolism in cortical astrocytes, and possibly also in other cell types.


Assuntos
Astrócitos/metabolismo , Bicarbonatos/metabolismo , Glucose/metabolismo , Glicólise/fisiologia , Prótons , Simportadores de Sódio-Bicarbonato/metabolismo , Animais , Transferência Ressonante de Energia de Fluorescência , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Knockout , Simportadores de Sódio-Bicarbonato/genética
14.
J Neurosci ; 34(4): 1148-57, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24453308

RESUMO

The electrogenic sodium bicarbonate cotransporter NBCe1 (SLC4A4) is a robust regulator of intracellular H(+) and a significant base carrier in many cell types. Using wild-type (WT) and NBCe1-deficient (NBC-KO) mice, we have studied the role of NBCe1 in cortical astrocytes in culture and in situ by monitoring intracellular H(+) using the H(+)-sensitive dye BCECF [2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein] in wide-field and confocal microscopy. Adding 0.1-3 mm HCO3(-) to an O2-gassed, HEPES-buffered saline solution lowered the intracellular H(+) concentration with a Km of 0.65 mm HCO3(-) in WT astrocytes, but slowly raised [H(+)]i in NBCe1-KO astrocytes. Human NBCe1 heterologously expressed in Xenopus oocytes could be activated by adding 1-3 mm HCO3(-), and even by residual HCO3(-) in a nominally CO2/HCO3(-)-free saline solution. Our results demonstrate a surprisingly high apparent bicarbonate sensitivity mediated by NBCe1 in cortical astrocytes, suggesting that NBCe1 may operate over a wide bicarbonate concentration in these cells.


Assuntos
Astrócitos/metabolismo , Córtex Cerebral/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Animais , Humanos , Concentração de Íons de Hidrogênio , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Técnicas de Patch-Clamp
15.
J Physiol ; 593(16): 3533-47, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25990710

RESUMO

KEY POINTS: The regulation of H(+) i from cytosolic alkalosis has generally been attributed to the activity of Cl(-) -coupled acid loaders/base extruders in most cell types, including brain cells. The present study demonstrates that outwardly-directed sodium bicarbonate cotransport via electrogenic sodium bicarbonate cotransporter 1 (NBCe1) mediates the major fraction of H(+) i regulation from cytosolic alkalosis in mouse cortical astrocytes. Cl(-) -coupled acid-loading transporters play only a minor role in the regulation of H(+) i from alkalosis in mouse cortical astrocytes. NBCe1-mediated H(+) i regulation from alkalosis was dominant, with the support of intracellular carbonic anhydrase II, even when the intra- and extracellular [HCO3 (-) ] was very low (<1mM), as in nominally CO2 /HCO3 (-) free condition. A reversed NBCe1 in astrocytes may also be significant for stabilizing extracellular pH in brain tissue. ABSTRACT: Recovery of intracellular pH from cytosolic alkalosis has been attributed primarily to Cl(-) coupled acid loaders/base extruders such as Cl(-) /HCO3 (-) or Cl(-) /OH(-) exchangers. We have studied this process in cortical astrocytes from wild-type and transgenic mouse models with gene deletion for the electrogenic sodium bicarbonate cotransporter 1 (NBCe1) and for carbonic anhydrase (CA) isoform II. An acute cytosolic alkalosis was induced by the removal of either CO2 /HCO3 (-) or butyric acid, and the subsequent acid loading was analysed by monitoring changes in cytosolic H(+) or Na(+) using ion-sensitive fluorescent dyes. We have identified that NBCe1 reverses during alkalosis and contributes more than 70% to the rate of recovery from alkalosis by extruding Na(+) and HCO3 (-) . After CA inhibition or in CAII-knockout (KO) cells, the rate of recovery was reduced by 40%, and even by 70% in the nominal absence of CO2 /HCO3 (-) . Increasing the extracellular K(+) concentration modulated the rate of acid loading in wild-type cells, but not in NBCe1-KO cells. Removing chloride had only a minor effect on the recovery from alkalosis. Reversal of NBCe1 by reducing pH/[HCO3 (-) ] was demonstrated in astrocytes and in Xenopus oocytes, in which human NBCe1 was heterologously expressed. The results obtained suggest that reversed NBCe1, supported by CAII activity, plays a major role in acid-loading cortical astrocytes to support recovery from cytosolic alkalosis.


Assuntos
Astrócitos/fisiologia , Simportadores de Sódio-Bicarbonato/fisiologia , Alcalose , Animais , Bicarbonatos/metabolismo , Ácido Butírico/metabolismo , Dióxido de Carbono/metabolismo , Células Cultivadas , Citosol/metabolismo , Feminino , Concentração de Íons de Hidrogênio , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/fisiologia , Sódio/metabolismo , Xenopus laevis
16.
Glia ; 63(9): 1581-94, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25820238

RESUMO

Cytosolic H(+) buffering plays a major role for shaping intracellular H(+) shifts and hence for the availability of H(+) for biochemical reactions and acid/base-coupled transport processes. H(+) buffering is one of the prime means to protect the cell from large acid/base shifts. We have used the H(+) indicator dye BCECF and confocal microscopy to monitor the cytosolic H(+) concentration, [H(+)]i, in cultured cortical astrocytes of wild-type mice and of mice deficient in sodium/bicarbonate cotransporter NBCe1 (NBCe1-KO) or in carbonic anhydrase isoform II (CAII-KO). The steady-state buffer strength was calculated from the amplitude of [H(+)]i transients as evoked by CO2/HCO3(-) and by butyric acid in the presence and absence of CO2/HCO3(-). We tested the hypotheses if, in addition to instantaneous physicochemical H(+) buffering, rapid acid/base transport across the cell membrane contributes to the total, "effective" cytosolic H(+) buffering. In the presence of 5% CO2/26 mM HCO3(-), H(+) buffer strength in astrocytes was increased 4-6 fold, as compared with that in non-bicarbonate, HEPES-buffered solution, which was largely attributable to fast HCO3 (-) transport into the cells via NBCe1, supported by CAII activity. Our results show that within the time frame of determining physiological H(+) buffering in cells, fast transport and equilibration of CO2/H(+)/HCO3(-) can make a major contribution to the total "effective" H(+) buffer strength. Thus, "effective" cellular H(+) buffering is, to a large extent, attributable to membrane transport of base equivalents rather than a purely passive physicochemical process, and can be much larger than reported so far. Not only physicochemical H(+) buffering, but also rapid import of HCO3(-) via the electrogenic sodium-bicarbonate cotransporter NBCe1, supported by carbonic anhydrase II (CA II), was identified to enhance cytosolic H(+) buffer strength substantially.


Assuntos
Astrócitos/metabolismo , Transporte Biológico/fisiologia , Córtex Cerebral/metabolismo , Citosol/metabolismo , Prótons , Simportadores de Sódio-Bicarbonato/metabolismo , Animais , Bicarbonatos/metabolismo , Células Cultivadas , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos , Simportadores de Sódio-Bicarbonato/genética , Xenopus laevis
17.
Glia ; 63(7): 1226-39, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25755028

RESUMO

The electrogenic sodium bicarbonate cotransporter NBCe1 (SLC4A4) is expressed in many cell types and is a major regulator of intracellular, and extracellular pH. In astrocytes, membrane depolarization leads to intracellular alkalinization through the activation of NBCe1. However, the molecular mechanisms regulating functional expression of NBCe1 in astrocytes are largely unknown. Astrocytes also express voltage-dependent K(+) channels that are activated after depolarization and are sensitive to the K(+) blocker 4-aminopyridine (4AP). Using acute hippocampal slices and primary hippocampal and cortical astrocyte cultures, we have investigated the role of 4AP for the regulation of NBCe1 and elucidated the underlying signaling pathways by quantitative RT-PCR, immunoblotting, biotinylation of surface proteins, immunofluorescence, and intracellular H(+) recording using the H(+) -sensitive dye 2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein. The results show significant upregulation of NBCe1 transcript, protein, and surface expression after the application of 4AP in both hippocampal slices and astrocyte cultures, effects that were suppressed after the inhibition of c-jun N-terminal kinase (JNK), proto-oncogene tyrosine-protein kinase Src, and Src/extracellular-signal-regulated kinases signaling. In the presence of 4AP, the rate and amplitude of intracellular H(+) changes upon challenging NBCe1 increased in wild-type astrocytes but not in cortical astrocytes from NBCe1-deficient mice. 4AP-dependent effects were suppressed after the inhibition of JNK and Src signaling. Our results demonstrate that transcriptional regulation and targeting of NBCe1, as well as functional operation of NBCe1, may occur through multiple signaling pathways.


Assuntos
Astrócitos/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , 4-Aminopiridina/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Knockout , Bloqueadores dos Canais de Potássio/farmacologia , Prótons , RNA Mensageiro/metabolismo , Ratos Wistar , Simportadores de Sódio-Bicarbonato/genética , Técnicas de Cultura de Tecidos , Quinases da Família src/metabolismo
18.
Cell Calcium ; 120: 102882, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38631162

RESUMO

Strict homeostatic control of pH in both intra- and extracellular compartments of the brain is fundamentally important, primarily due to the profound impact of free protons ([H+]) on neuronal activity and overall brain function. Astrocytes, crucial players in the homeostasis of various ions in the brain, actively regulate their intracellular [H+] (pHi) through multiple membrane transporters and carbonic anhydrases. The activation of astroglial pHi regulating mechanisms also leads to corresponding alterations in the acid-base status of the extracellular fluid. Notably, astrocyte pH regulators are modulated by various neuronal signals, suggesting their pivotal role in regulating brain acid-base balance in both health and disease. This review presents the mechanisms involved in pH regulation in astrocytes and discusses their potential impact on extracellular pH under physiological conditions and in brain disorders. Targeting astrocytic pH regulatory mechanisms represents a promising therapeutic approach for modulating brain acid-base balance in diseases, offering a potential critical contribution to neuroprotection.


Assuntos
Astrócitos , Encéfalo , Astrócitos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Animais , Encéfalo/metabolismo , Encefalopatias/metabolismo , Encefalopatias/patologia , Homeostase
19.
Cell Rep ; 42(12): 113514, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38041814

RESUMO

During hypoxia, increases in cerebral blood flow maintain brain oxygen delivery. Here, we describe a mechanism of brain oxygen sensing that mediates the dilation of intraparenchymal cerebral blood vessels in response to reductions in oxygen supply. In vitro and in vivo experiments conducted in rodent models show that during hypoxia, cortical astrocytes produce the potent vasodilator nitric oxide (NO) via nitrite reduction in mitochondria. Inhibition of mitochondrial respiration mimics, but also occludes, the effect of hypoxia on NO production in astrocytes. Astrocytes display high expression of the molybdenum-cofactor-containing mitochondrial enzyme sulfite oxidase, which can catalyze nitrite reduction in hypoxia. Replacement of molybdenum with tungsten or knockdown of sulfite oxidase expression in astrocytes blocks hypoxia-induced NO production by these glial cells and reduces the cerebrovascular response to hypoxia. These data identify astrocyte mitochondria as brain oxygen sensors that regulate cerebral blood flow during hypoxia via release of nitric oxide.


Assuntos
Hipóxia Encefálica , Nitritos , Humanos , Nitritos/metabolismo , Astrócitos/metabolismo , Óxido Nítrico/metabolismo , Molibdênio/metabolismo , Hipóxia/metabolismo , Oxigênio/metabolismo , Mitocôndrias/metabolismo , Hipóxia Encefálica/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Circulação Cerebrovascular
20.
Nat Commun ; 13(1): 2125, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35440557

RESUMO

Neurovascular coupling is a fundamental brain mechanism that regulates local cerebral blood flow (CBF) in response to changes in neuronal activity. Functional imaging techniques are commonly used to record these changes in CBF as a proxy of neuronal activity to study the human brain. However, the mechanisms of neurovascular coupling remain incompletely understood. Here we show in experimental animal models (laboratory rats and mice) that the neuronal activity-dependent increases in local CBF in the somatosensory cortex are prevented by saturation of the CO2-sensitive vasodilatory brain mechanism with surplus of exogenous CO2 or disruption of brain CO2/HCO3- transport by genetic knockdown of electrogenic sodium-bicarbonate cotransporter 1 (NBCe1) expression in astrocytes. A systematic review of the literature data shows that CO2 and increased neuronal activity recruit the same vasodilatory signaling pathways. These results and analysis suggest that CO2 mediates signaling between neurons and the cerebral vasculature to regulate brain blood flow in accord with changes in the neuronal activity.


Assuntos
Acoplamento Neurovascular , Animais , Dióxido de Carbono/metabolismo , Córtex Cerebral/metabolismo , Circulação Cerebrovascular , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Simportadores de Sódio-Bicarbonato/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA