Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Am J Physiol Regul Integr Comp Physiol ; 325(3): R229-R237, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37424401

RESUMO

To investigate the role of glial cells in the regulation of glucoprivic responses in rats, a chemogenetic approach was used to activate astrocytes neighboring catecholamine (CA) neurons in the ventromedial medulla (VLM) where A1 and C1 CA cell groups overlap (A1/C1). Previous results indicate that activation of CA neurons in this region is necessary and sufficient for feeding and corticosterone release in response to glucoprivation. However, it is not known whether astrocyte neighbors of CA neurons contribute to glucoregulatory responses. Hence, we made nanoinjections of AAV5-GFAP-hM3D(Gq)-mCherry to selectively transfect astrocytes in the A1/C1 region with the excitatory designer receptor exclusively activated by designer drugs (DREADDs), hM3D(Gq). After allowing time for DREADD expression, we evaluated the rats for increased food intake and corticosterone release in response to low systemic doses of the antiglycolytic agent, 2-deoxy-d-glucose (2DG), alone and in combination with the hM3D(Gq) activator clozapine-n-oxide (CNO). We found that DREADD-transfected rats ate significantly more food when 2DG and CNO were coadministered than when either 2DG or CNO was injected alone. We also found that CNO significantly enhanced 2DG-induced FOS expression in the A1/C1 CA neurons, and that corticosterone release also was enhanced when CNO and 2DG were administered together. Importantly, CNO-induced activation of astrocytes in the absence of 2DG did not trigger food intake or corticosterone release. Our results indicate that during glucoprivation, activation of VLM astrocytes cells markedly increases the sensitivity or responsiveness of neighboring A1/C1 CA neurons to glucose deficit, suggesting a potentially important role for VLM astrocytes in glucoregulation.


Assuntos
Astrócitos , Corticosterona , Ratos , Animais , Astrócitos/metabolismo , Desoxiglucose/farmacologia , Ratos Sprague-Dawley , Bulbo/metabolismo , Glucose/metabolismo , Catecolaminas/metabolismo
2.
J Biol Chem ; 297(4): 101196, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34529976

RESUMO

Mitochondria undergo continuous cycles of fission and fusion to promote inheritance, regulate quality control, and mitigate organelle stress. More recently, this process of mitochondrial dynamics has been demonstrated to be highly sensitive to nutrient supply, ultimately conferring bioenergetic plasticity to the organelle. However, whether regulators of mitochondrial dynamics play a causative role in nutrient regulation remains unclear. In this study, we generated a cellular loss-of-function model for dynamin-related protein 1 (DRP1), the primary regulator of outer membrane mitochondrial fission. Loss of DRP1 (shDRP1) resulted in extensive ultrastructural and functional remodeling of mitochondria, characterized by pleomorphic enlargement, increased electron density of the matrix, and defective NADH and succinate oxidation. Despite increased mitochondrial size and volume, shDRP1 cells exhibited reduced cellular glucose uptake and mitochondrial fatty acid oxidation. Untargeted transcriptomic profiling revealed severe downregulation of genes required for cellular and mitochondrial calcium homeostasis, which was coupled to loss of ATP-stimulated calcium flux and impaired substrate oxidation stimulated by exogenous calcium. The insights obtained herein suggest that DRP1 regulates substrate oxidation by altering whole-cell and mitochondrial calcium dynamics. These findings are relevant to the targetability of mitochondrial fission and have clinical relevance in the identification of treatments for fission-related pathologies such as hereditary neuropathies, inborn errors in metabolism, cancer, and chronic diseases.


Assuntos
Sinalização do Cálcio , Dinaminas/metabolismo , Mitocôndrias Musculares/metabolismo , Dinâmica Mitocondrial , Linhagem Celular , Dinaminas/genética , Ácidos Graxos/genética , Ácidos Graxos/metabolismo , Humanos , Mitocôndrias Musculares/genética , Oxirredução
3.
J Neurophysiol ; 123(6): 2122-2135, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32347148

RESUMO

Peripheral viscerosensory afferent signals are transmitted to the nucleus tractus solitarii (nTS) via release of glutamate. Following release, glutamate is removed from the extrasynaptic and synaptic cleft via excitatory amino acid transporters (EAATs), thus limiting glutamate receptor activation or over activation, and maintaining its working range. We have shown that EAAT block with the antagonist threo-ß-benzyloxyaspartic acid (TBOA) depolarized nTS neurons and increased spontaneous excitatory postsynaptic current (sEPSC) frequency yet reduced the amplitude of afferent (TS)-evoked EPSCs (TS-EPSCs). Interestingly, chronic intermittent hypoxia (CIH), a model of obstructive sleep apnea (OSA), produces similar synaptic responses as EAAT block. We hypothesized EAAT expression or function are downregulated after CIH, and this reduction in glutamate removal contributes to the observed neurophysiological responses. To test this hypothesis, we used brain slice electrophysiology and imaging of glutamate release and TS-afferent Ca2+ to compare nTS properties of rats exposed to 10 days of normoxia (Norm; 21%O2) or CIH. Results show that EAAT blockade with (3S)-3-[[3-[[4-(trifluoromethyl)benzoyl]-amino]phenyl]methoxy]-l-aspartic acid (TFB-TBOA) in Norm caused neuronal depolarization, generation of an inward current, and increased spontaneous synaptic activity. The latter augmentation was eliminated by inclusion of tetrodotoxin in the perfusate. TS stimulation during TFB-TBOA also elevated extracellular glutamate and decreased presynaptic Ca2+ and TS-EPSC amplitude. In CIH, the effects of EAAT block are eliminated or attenuated. CIH reduced EAAT expression in nTS, which may contribute to the attenuated function seen in this condition. Therefore, CIH reduces EAAT influence on synaptic and neuronal activity, which may lead to the physiological consequences seen in OSA and CIH.NEW & NOTEWORTHY Removal of excitatory amino acid transporter (EAAT) restraint increases spontaneous synaptic activity yet decreases afferent [tractus solitarius (TS)]-driven excitatory postsynaptic current (EPSC) amplitude. In the chronic intermittent hypoxia model of obstructive sleep apnea, this restraint is lost due to reduction in EAAT expression and function. Thus EAATs are important in controlling elevated glutamatergic signaling, and loss of such control results in maladaptive synaptic signaling.


Assuntos
Astrócitos/fisiologia , Células Quimiorreceptoras/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Ácido Glutâmico/metabolismo , Hipóxia , Transdução de Sinais/fisiologia , Apneia Obstrutiva do Sono , Núcleo Solitário , Animais , Modelos Animais de Doenças , Proteínas de Transporte de Glutamato da Membrana Plasmática/antagonistas & inibidores , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley , Apneia Obstrutiva do Sono/metabolismo , Apneia Obstrutiva do Sono/fisiopatologia , Núcleo Solitário/metabolismo , Núcleo Solitário/fisiopatologia
4.
Am J Physiol Regul Integr Comp Physiol ; 318(6): R1068-R1077, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32320636

RESUMO

Severe trauma can produce a postinjury "metabolic self-destruction" characterized by catabolic metabolism and hyperglycemia. The severity of the hyperglycemia is highly correlated with posttrauma morbidity and mortality. Although no mechanism has been posited to connect severe trauma with a loss of autonomic control over metabolism, traumatic injury causes other failures of autonomic function, notably, gastric stasis and ulceration ("Cushing's ulcer"), which has been connected with the generation of thrombin. Our previous studies established that proteinase-activated receptors (PAR1; "thrombin receptors") located on astrocytes in the autonomically critical nucleus of the solitary tract (NST) can modulate gastric control circuit neurons to cause gastric stasis. Hindbrain astrocytes have also been implicated as important detectors of low glucose or glucose utilization. When activated, these astrocytes communicate with hindbrain catecholamine neurons that, in turn, trigger counterregulatory responses (CRR). There may be a convergence between the effects of thrombin to derange hindbrain gastrointestinal control and the hindbrain circuitry that initiates CRR to increase glycemia in reaction to critical hypoglycemia. Our results suggest that thrombin acts within the NST to increase glycemia through an astrocyte-dependent mechanism. Blockade of purinergic gliotransmission pathways interrupted the effect of thrombin to increase glycemia. Our studies also revealed that thrombin, acting in the NST, produced a rapid, dramatic, and potentially lethal suppression of respiratory rhythm that was also a function of purinergic gliotransmission. These results suggest that the critical connection between traumatic injury and a general collapse of autonomic regulation involves thrombin action on astrocytes.


Assuntos
Astrócitos/efeitos dos fármacos , Glicemia , Neurônios/efeitos dos fármacos , Rombencéfalo/efeitos dos fármacos , Trombina/farmacologia , Animais , Masculino , Nervo Frênico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Taxa Respiratória/efeitos dos fármacos , Núcleo Solitário/efeitos dos fármacos
5.
Am J Physiol Regul Integr Comp Physiol ; 318(3): R545-R564, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31967862

RESUMO

Astrocytic excitatory amino acid transporters (EAATs) are critical to restraining synaptic and neuronal activity in the nucleus tractus solitarii (nTS). Relief of nTS EAAT restraint generates two opposing effects, an increase in neuronal excitability that reduces blood pressure and breathing and an attenuation in afferent [tractus solitarius (TS)]-driven excitatory postsynaptic current (EPSC) amplitude. Although the former is due, in part, to activation of ionotropic glutamate receptors, there remains a substantial contribution from another unidentified glutamate receptor. In addition, the mechanism(s) by which EAAT inhibition reduced TS-EPSC amplitude is unknown. Metabotropic glutamate receptors (mGluRs) differentially modulate nTS excitability. Activation of group I mGluRs on nTS neuron somas leads to depolarization, whereas group II/III mGluRs on sensory afferents decrease TS-EPSC amplitude. Thus we hypothesize that EAATs control postsynaptic excitability and TS-EPSC amplitude via restraint of mGluR activation. To test this hypothesis, we used in vivo recording, brain slice electrophysiology, and imaging of glutamate release and TS-afferent Ca2+. Results show that EAAT blockade in the nTS with (3S)-3-[[3-[[4-(trifluoromethyl)benzoyl]amino]phenyl]methoxy]-l-aspartic acid (TFB-TBOA) induced group I mGluR-mediated depressor, bradycardic, and apneic responses that were accompanied by neuronal depolarization, elevated discharge, and increased spontaneous synaptic activity. Conversely, upon TS stimulation TFB-TBOA elevated extracellular glutamate to decrease presynaptic Ca2+ and TS-EPSC amplitude via activation of group II/III mGluRs. Together, these data suggest an important role of EAATs in restraining mGluR activation and overall cardiorespiratory function.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/efeitos dos fármacos , Ácido Aspártico/análogos & derivados , Astrócitos/metabolismo , Neurônios/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Ácido Aspártico/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/metabolismo , Neurônios/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Núcleo Solitário/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
6.
Am J Physiol Regul Integr Comp Physiol ; 318(1): R38-R48, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31596114

RESUMO

Astrocytes generate robust cytoplasmic calcium signals in response to reductions in extracellular glucose. This calcium signal, in turn, drives purinergic gliotransmission, which controls the activity of catecholaminergic (CA) neurons in the hindbrain. These CA neurons are critical to triggering glucose counter-regulatory responses (CRRs) that, ultimately, restore glucose homeostasis via endocrine and behavioral means. Although the astrocyte low-glucose sensor involvement in CRR has been accepted, it is not clear how astrocytes produce an increase in intracellular calcium in response to a decrease in glucose. Our ex vivo calcium imaging studies of hindbrain astrocytes show that the glucose type 2 transporter (GLUT2) is an essential feature of the astrocyte glucosensor mechanism. Coimmunoprecipitation assays reveal that the recombinant GLUT2 binds directly with the recombinant Gq protein subunit that activates phospholipase C (PLC). Additional calcium imaging studies suggest that GLUT2 may be connected to a PLC-endoplasmic reticular-calcium release mechanism, which is amplified by calcium-induced calcium release (CICR). Collectively, these data help outline a potential mechanism used by astrocytes to convert information regarding low-glucose levels into intracellular changes that ultimately regulate the CRR.


Assuntos
Astrócitos/fisiologia , Cálcio/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glucose/metabolismo , Rombencéfalo/citologia , Fosfolipases Tipo C/metabolismo , Anilidas/farmacologia , Animais , Antioxidantes/farmacologia , Compostos de Boro/farmacologia , Cálcio/farmacologia , Dantroleno/farmacologia , Estrenos/farmacologia , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Florizina/farmacologia , Pró-Fármacos , Pirrolidinonas/farmacologia , Quercetina/farmacologia , Ratos , Ratos Long-Evans , Fosfolipases Tipo C/antagonistas & inibidores
7.
Brain ; 142(11): 3382-3397, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31637422

RESUMO

CTP:phosphoethanolamine cytidylyltransferase (ET), encoded by PCYT2, is the rate-limiting enzyme for phosphatidylethanolamine synthesis via the CDP-ethanolamine pathway. Phosphatidylethanolamine is one of the most abundant membrane lipids and is particularly enriched in the brain. We identified five individuals with biallelic PCYT2 variants clinically characterized by global developmental delay with regression, spastic para- or tetraparesis, epilepsy and progressive cerebral and cerebellar atrophy. Using patient fibroblasts we demonstrated that these variants are hypomorphic, result in altered but residual ET protein levels and concomitant reduced enzyme activity without affecting mRNA levels. The significantly better survival of hypomorphic CRISPR-Cas9 generated pcyt2 zebrafish knockout compared to a complete knockout, in conjunction with previously described data on the Pcyt2 mouse model, indicates that complete loss of ET function may be incompatible with life in vertebrates. Lipidomic analysis revealed profound lipid abnormalities in patient fibroblasts impacting both neutral etherlipid and etherphospholipid metabolism. Plasma lipidomics studies also identified changes in etherlipids that have the potential to be used as biomarkers for ET deficiency. In conclusion, our data establish PCYT2 as a disease gene for a new complex hereditary spastic paraplegia and confirm that etherlipid homeostasis is important for the development and function of the brain.


Assuntos
Fosfatidiletanolaminas/biossíntese , RNA Nucleotidiltransferases/genética , Paraplegia Espástica Hereditária/genética , Adolescente , Alelos , Animais , Atrofia , Encéfalo/patologia , Criança , Pré-Escolar , Deficiências do Desenvolvimento/genética , Epilepsia/genética , Feminino , Técnicas de Inativação de Genes , Variação Genética , Humanos , Lipidômica , Masculino , Camundongos , RNA Nucleotidiltransferases/deficiência , Adulto Jovem , Peixe-Zebra
8.
Am J Physiol Regul Integr Comp Physiol ; 315(1): R153-R164, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29590557

RESUMO

Hindbrain catecholaminergic (CA) neurons are required for critical autonomic, endocrine, and behavioral counterregulatory responses (CRRs) to hypoglycemia. Recent studies suggest that CRR initiation depends on hindbrain astrocyte glucose sensors (McDougal DH, Hermann GE, Rogers RC. Front Neurosci 7: 249, 2013; Rogers RC, Ritter S, Hermann GE. Am J Physiol Regul Integr Comp Physiol 310: R1102-R1108, 2016). To test the proposition that hindbrain CA responses to glucoprivation are astrocyte dependent, we utilized transgenic mice in which the calcium reporter construct (GCaMP5) was expressed selectively in tyrosine hydroxylase neurons (TH-GCaMP5). We conducted live cell calcium-imaging studies on tissue slices containing the nucleus of the solitary tract (NST) or the ventrolateral medulla, critical CRR initiation sites. Results show that TH-GCaMP5 neurons are robustly activated by a glucoprivic challenge and that this response is dependent on functional astrocytes. Pretreatment of hindbrain slices with fluorocitrate (an astrocytic metabolic suppressor) abolished TH-GCaMP5 neuronal responses to glucoprivation, but not to glutamate. Pharmacologic results suggest that the astrocytic connection with hindbrain CA neurons is purinergic via P2 receptors. Parallel imaging studies on hindbrain slices of NST from wild-type C57BL/6J mice, in which astrocytes and neurons were prelabeled with a calcium reporter dye and an astrocytic vital dye, show that both cell types are activated by glucoprivation but astrocytes responded significantly sooner than neurons. Pretreatment of these hindbrain slices with P2 antagonists abolished neuronal responses to glucoprivation without interruption of astrocyte responses; pretreatment with fluorocitrate eliminated both astrocytic and neuronal responses. These results support earlier work suggesting that the primary detection of glucoprivic signals by the hindbrain is mediated by astrocytes.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio , Catecolaminas/metabolismo , Glucose/deficiência , Neurônios/metabolismo , Rombencéfalo/metabolismo , Animais , Feminino , Genes Reporter , Ácido Glutâmico/metabolismo , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Receptores Purinérgicos P2/metabolismo , Rombencéfalo/citologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Semin Immunol ; 26(5): 402-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24486057

RESUMO

The sympathetic nervous system (SNS) is part of an integrative network that functions to restore homeostasis following injury and infection. The SNS can provide negative feedback control over inflammation through the secretion of catecholamines from postganglionic sympathetic neurons and adrenal chromaffin cells (ACCs). Central autonomic structures receive information regarding the inflammatory status of the body and reflexively modulate SNS activity. However, inflammation and infection can also directly regulate SNS function by peripheral actions on postganglionic cells. The present review discusses how inflammation activates autonomic reflex pathways and compares the effect of localized and systemic inflammation on ACCs and postganglionic sympathetic neurons. Systemic inflammation significantly enhanced catecholamine secretion through an increase in Ca(2+) release from the endoplasmic reticulum. In contrast, acute and chronic GI inflammation reduced voltage-gated Ca(2+) current. Thus it appears that the mechanisms underlying the effects of peripheral and systemic inflammation neuroendocrine function converge on the modulation of intracellular Ca(2+) signaling.


Assuntos
Cálcio/metabolismo , Catecolaminas/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Neurônios/metabolismo , Sepse/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Cálcio/imunologia , Sinalização do Cálcio , Catecolaminas/imunologia , Células Cromafins/imunologia , Células Cromafins/metabolismo , Células Cromafins/patologia , Citocinas/genética , Citocinas/imunologia , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Neurônios/imunologia , Neurônios/patologia , Sepse/genética , Sepse/imunologia , Sepse/patologia , Sistema Nervoso Simpático/imunologia , Sistema Nervoso Simpático/patologia
10.
J Neurosci ; 36(12): 3531-40, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27013681

RESUMO

Astrocytes are well established modulators of extracellular glutamate, but their direct influence on energy balance-relevant behaviors is largely understudied. As the anorectic effects of glucagon-like peptide-1 receptor (GLP-1R) agonists are partly mediated by central modulation of glutamatergic signaling, we tested the hypothesis that astrocytic GLP-1R signaling regulates energy balance in rats. Central or peripheral administration of a fluorophore-labeled GLP-1R agonist, exendin-4, localizes within astrocytes and neurons in the nucleus tractus solitarius (NTS), a hindbrain nucleus critical for energy balance control. This effect is mediated by GLP-1R, as the uptake of systemically administered fluorophore-tagged exendin-4 was blocked by central pretreatment with the competitive GLP-1R antagonist exendin-(9-39). Ex vivo analyses show prolonged exendin-4-induced activation (live cell calcium signaling) of NTS astrocytes and neurons; these effects are also attenuated by exendin-(9-39), indicating mediation by the GLP-1R. In vitro analyses show that the application of GLP-1R agonists increases cAMP levels in astrocytes. Immunohistochemical analyses reveal that endogenous GLP-1 axons form close synaptic apposition with NTS astrocytes. Finally, pharmacological inhibition of NTS astrocytes attenuates the anorectic and body weight-suppressive effects of intra-NTS GLP-1R activation. Collectively, data demonstrate a role for NTS astrocytic GLP-1R signaling in energy balance control. SIGNIFICANCE STATEMENT: Glucagon-like peptide-1 receptor (GLP-1R) agonists reduce food intake and are approved by the Food and Drug Administration for the treatment of obesity, but the cellular mechanisms underlying the anorectic effects of GLP-1 require further investigation. Astrocytes represent a major cellular population in the CNS that regulates neurotransmission, yet the role of astrocytes in mediating energy balance is largely unstudied. The current data provide novel evidence that astrocytes within the NTS are relevant for energy balance control by GLP-1 signaling. Here, we report that GLP-1R agonists activate and internalize within NTS astrocytes, while behavioral data suggest the pharmacological relevance of NTS astrocytic GLP-1R activation for food intake and body weight. These findings support a previously unknown role for CNS astrocytes in energy balance control by GLP-1 signaling.


Assuntos
Regulação do Apetite/fisiologia , Astrócitos/fisiologia , Comportamento Alimentar/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Homeostase/fisiologia , Bulbo/metabolismo , Animais , Metabolismo Energético/fisiologia , Retroalimentação Fisiológica/fisiologia , Masculino , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley
11.
J Neurosci ; 35(2): 776-85, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25589770

RESUMO

Severe autonomic dysfunction, including the loss of control of the cardiovascular, respiratory, and gastrointestinal systems, is a common comorbidity of stroke and other bleeding head injuries. Previous studies suggest that this collapse of autonomic control may be caused by thrombin acting on astrocytic protease-activated receptors (PAR1) in the hindbrain. Using calcium imaging and electrophysiological techniques, we evaluated the mechanisms by which astrocytic PAR1s modulate the activity of presynaptic vagal afferent terminals and postsynaptic neurons in the rat nucleus of the solitary tract (NST). Our calcium-imaging data show that astrocytic and neuronal calcium levels increase after brain slices are treated with the PAR1 agonist SFLLRN-NH2. This increase in activity is blocked by pretreating the slices with the glial metabolic blocker fluorocitrate. In addition, PAR1-activated astrocytes communicate directly with NST neurons by releasing glutamate. Calcium responses to SFLLRN-NH2 in the astrocytes and neurons significantly increase after bath application of the excitatory amino acid transporter blocker DL-threo-ß-benzyloxyaspartic acid (TBOA) and significantly decrease after bath application of the NMDA receptor antagonist DL-2-amino-5-phosphonopentanoic acid (DL-AP5). Furthermore, astrocytic glutamate activates neuronal GluN2B-containing NMDA receptors. Voltage-clamp recordings of miniature EPSCs (mEPSCs) from NST neurons show that astrocytes control presynaptic vagal afferent excitability directly under resting and activated conditions. Fluorocitrate significantly decreases mEPSC frequency and SFLLRN-NH2 significantly increases mEPSC frequency. These data show that astrocytes act within a tripartite synapse in the NST, controlling the excitability of both postsynaptic NST neurons and presynaptic vagal afferent terminals.


Assuntos
Astrócitos/metabolismo , Neurônios Aferentes/fisiologia , Receptor PAR-1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleo Solitário/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , Animais , Ácido Aspártico/farmacologia , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Citratos/farmacologia , Antagonistas de Aminoácidos Excitatórios , Potenciais Pós-Sinápticos Excitadores , Feminino , Ácido Glutâmico/metabolismo , Masculino , Potenciais Pós-Sinápticos em Miniatura , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Long-Evans , Receptor PAR-1/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Núcleo Solitário/citologia , Núcleo Solitário/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia , Nervo Vago/citologia , Nervo Vago/metabolismo , Nervo Vago/fisiologia
12.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1102-8, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27101298

RESUMO

The hindbrain contains critical neurocircuitry responsible for generating defensive physiological responses to hypoglycemia. This counter-regulatory response (CRR) is evoked by local hindbrain cytoglucopenia that causes an autonomically mediated increase in blood glucose, feeding behavior, and accelerated digestion; that is, actions that restore glucose homeostasis. Recent reports suggest that CRR may be initially triggered by astrocytes in the hindbrain. The present studies in thiobutabarbital-anesthetized rats show that exposure of the fourth ventricle (4V) to 2-deoxyglucose (2DG; 15 µmol) produced a 35% increase in circulating glucose relative to baseline levels. While the 4V application of the astrocytic signal blocker, fluorocitrate (FC; 5 nmol), alone, had no effect on blood glucose levels, 2DG-induced increases in glucose were blocked by 4V FC. The 4V effect of 2DG to increase glycemia was also blocked by the pretreatment with caffeine (nonselective adenosine antagonist) or a potent adenosine A1 antagonist (8-cyclopentyl-1,3-dipropylxanthine; DPCPX) but not the NMDA antagonist (MK-801). These results suggest that CNS detection of glucopenia is mediated by astrocytes and that astrocytic release of adenosine that occurs after hypoglycemia may cause the activation of downstream neural circuits that drive CRR.


Assuntos
Adenosina/metabolismo , Glicemia/metabolismo , Desoxiglucose/administração & dosagem , Hipoglicemia/metabolismo , Rombencéfalo/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Glicemia/efeitos dos fármacos , Feminino , Homeostase/efeitos dos fármacos , Infusões Intraventriculares , Masculino , Ratos , Ratos Long-Evans , Rombencéfalo/patologia , Regulação para Cima/efeitos dos fármacos
13.
J Neurosci ; 34(32): 10488-96, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-25100584

RESUMO

Fasting and hypoglycemia elicit powerful gastrointestinal contractions. Whereas the relationship between utilizable nutrient and gastric motility is well recognized, the explanation of this phenomenon has remained incomplete. A relatively recent controversial report suggested that astrocytes in the dorsal hindbrain may be the principal detectors of glucoprivic stimuli. Our own studies also show that a subset of astrocytes in the solitary nucleus (NST) is activated by low glucose. It is very likely that information about glucopenia may directly impact gastric control because the hindbrain is also the location of the vago-vagal reflex circuitry regulating gastric motility. Our in vivo single unit neurophysiological recordings in intact rats show fourth ventricular application of 2-deoxyglucose (2-DG) inhibits NST neurons and activates dorsal motor nucleus (DMN) neurons involved in the gastric accommodation reflex. Additionally, as shown in earlier studies, either systemic insulin or central 2-DG causes an increase in gastric motility. These effects on motility were blocked by fourth ventricle pretreatment with the astrocyte inactivator fluorocitrate. Fluorocitrate administered alone has no effect on gastric-NST or -DMN neuron responsiveness, or on gastric motility. These results suggest that glucoprivation-induced increases in gastric motility are dependent on intact hindbrain astrocytes.


Assuntos
Citratos/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Reflexo/efeitos dos fármacos , Rombencéfalo/citologia , Nervo Vago/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Desoxiglucose/farmacologia , Inibidores Enzimáticos/farmacologia , Glucose/metabolismo , Insulina/farmacologia , Neurônios/fisiologia , Ratos , Reflexo/fisiologia , Nervo Vago/fisiologia
14.
Am J Physiol Endocrinol Metab ; 309(8): E715-26, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26306596

RESUMO

Proinflammatory cytokines impact islet ß-cell mass and function by altering the transcriptional activity within pancreatic ß-cells, producing increases in intracellular nitric oxide abundance and the synthesis and secretion of immunomodulatory proteins such as chemokines. Herein, we report that IL-1ß, a major mediator of inflammatory responses associated with diabetes development, coordinately and reciprocally regulates chemokine and insulin secretion. We discovered that NF-κB controls the increase in chemokine transcription and secretion as well as the decrease in both insulin secretion and proliferation in response to IL-1ß. Nitric oxide production, which is markedly elevated in pancreatic ß-cells exposed to IL-1ß, is a negative regulator of both glucose-stimulated insulin secretion and glucose-induced increases in intracellular calcium levels. By contrast, the IL-1ß-mediated production of the chemokines CCL2 and CCL20 was not influenced by either nitric oxide levels or glucose concentration. Instead, the synthesis and secretion of CCL2 and CCL20 in response to IL-1ß were dependent on NF-κB transcriptional activity. We conclude that IL-1ß-induced transcriptional reprogramming via NF-κB reciprocally regulates chemokine and insulin secretion while also negatively regulating ß-cell proliferation. These findings are consistent with NF-κB as a major regulatory node controlling inflammation-associated alterations in islet ß-cell function and mass.


Assuntos
Quimiocinas/metabolismo , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Interleucina-1beta/metabolismo , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Quimiocinas/genética , Espectroscopia de Ressonância de Spin Eletrônica , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Immunoblotting , Insulina/genética , Secreção de Insulina , Insulinoma , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Consumo de Oxigênio , Neoplasias Pancreáticas , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Ratos Zucker , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína S9 Ribossômica , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Células Tumorais Cultivadas
15.
J Neurosci ; 32(15): 5237-41, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22496569

RESUMO

The early proinflammatory cytokine tumor necrosis factor (TNF) is released in significant quantities by the activated immune system in response to infection, leukemia, autoimmune disorders, and radiation sickness. Nausea, emesis, and anorexia are common features of these disorders. TNF action on vagal afferent terminals in the brainstem is a likely cause of the malaise associated with these disorders. Our previous work has shown that TNF action to excite vagal afferents occurs as a result of sensitization of ryanodine channels in afferent nerve terminals. For millennia, cannabinoids (CB) have been used to combat the visceral malaise associated with chronic disease, although the mechanism of action has not been clear. Previous work in culture systems suggests that CB1 agonists can suppress neurotransmission by downregulating ryanodine channels through a protein kinase A (PKA)-dependent mechanism. Laser confocal calcium imaging methods were used to directly examine effects of CB1 cannabinoid agonists and TNF on visceral afferent signaling in the rat hindbrain. CB1 agonists blocked the effects of TNF to amplify vagal afferent responsiveness; blockade of PKA with H89 also eliminated the TNF amplification effect. These results help to explain the effectiveness of cannabinoids in blocking the malaise generated by TNF-releasing disease processes by opposing effects on ryanodine channels.


Assuntos
Cálcio/metabolismo , Canabinoides/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia , Animais , Tronco Encefálico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Cultura em Câmaras de Difusão , Estimulação Elétrica , Feminino , Processamento de Imagem Assistida por Computador , Isoquinolinas/farmacologia , Masculino , Microscopia Confocal , Terminações Pré-Sinápticas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Long-Evans , Receptor CB1 de Canabinoide/agonistas , Núcleo Solitário/fisiologia , Sulfonamidas/farmacologia
16.
J Neurosci ; 31(39): 14037-45, 2011 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-21957265

RESUMO

The nucleus of the solitary tract (NST), located in the dorsomedial medulla, is the site of visceral sensory modulation of a variety of homeostatic reflexes. Given recent advancements in the understanding of active regulation of synaptic information flow by astrocytes, we sought to determine whether afferent sensory inputs to NST neurons also activates NST astrocytes. Using confocal, live-cell calcium imaging of brainstem slices, we investigated the possibility that stimulation of vagal sensory afferents, the major sensory input into the NST, activated NST astrocytes, as indicated by increases in astrocytic intracellular calcium concentrations ([Ca²âº](i)). Astrocytes and neurons were preloaded with the calcium reporter dye Calcium Green, and astrocytes were selectively stained by sulforhodamine 101. Electrical stimulation of vagal afferent axons produced rapid increases in [Ca²âº](i) in NST astrocytes as well as neurons. Surprisingly, this effect on astrocytes was blocked by the AMPA receptor antagonist NBQX and was unaffected by antagonism of NMDA and metabotropic glutamate receptors. Bath application of AMPA also activated astrocytes. This activation was dependent on extracellular Ca²âº influx through both typical AMPA receptors and calcium-permeable AMPA receptors. This AMPA-mediated Ca²âº influx was further amplified by actions of the ryanodine receptor by way of calcium-induced calcium release. Our immunohistochemical staining of NST cells further verified the presence of the AMPAR subunit GluR1 on astrocytes. These observations suggest that NST astrocytes may be active participants in the regulation of autonomic reflexes even in the normal, healthy state.


Assuntos
Astrócitos/fisiologia , Tronco Encefálico/fisiologia , Neuroglia/fisiologia , Receptores de AMPA/fisiologia , Núcleo Solitário/fisiologia , Nervo Vago/fisiologia , Vias Aferentes/citologia , Vias Aferentes/fisiologia , Animais , Tronco Encefálico/citologia , Comunicação Celular/fisiologia , Feminino , Masculino , Neurônios/fisiologia , Ratos , Ratos Long-Evans , Núcleo Solitário/citologia , Núcleo Solitário/metabolismo
17.
J Physiol ; 590(3): 631-46, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22155934

RESUMO

Cholecystokinin (CCK) is a potent regulator of visceral functions as a consequence of its actions on vago-vagal reflex circuit elements. This paper addresses three current controversies regarding the role of CCK to control gastric function via vago-vagal reflexes. Specifically: (a) whether CNS vs. peripheral (vagal afferent) receptors are dominant, (b) whether the long (58) vs. short (8) isoform is more potent and (c) whether nutritional status impacts the gain or even the direction of vago-vagal reflexes. Our in vivo recordings of physiologically identified gastric vagal motor neurones (gastric-DMN) involved in the gastric accommodation reflex (GAR) show unequivocally that: (a) receptors in the coeliac-portal circulation are more sensitive in amplifying gastric vagal reflexes; (b) in the periphery, CCK8 is more potent than CCK58; and (c) the nutritional status has a marginal effect on gastric reflex control. While the GAR reflex is more sensitive in the fasted rat, CCK amplifies this sensitivity. Thus, our results are in stark contrast to recent reports which have suggested that vago-vagal reflexes are inverted by the metabolic status of the animal and that this inversion could be mediated by CCK within the CNS.


Assuntos
Colecistocinina/fisiologia , Jejum/fisiologia , Neurônios Motores/fisiologia , Sincalida/fisiologia , Nervo Vago/fisiologia , Animais , Feminino , Masculino , Ratos , Ratos Long-Evans , Reflexo/fisiologia , Estômago/inervação , Estômago/fisiologia
18.
J Biol Chem ; 286(13): 11659-71, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21296886

RESUMO

An ATP-Mg(2+/)P(i) inner mitochondrial membrane solute transporter (SLC25A25), which is induced during adaptation to cold stress in the skeletal muscle of mice with defective UCP1/brown adipose tissue thermogenesis, has been evaluated for its role in metabolic efficiency. SLC25A25 is thought to control ATP homeostasis by functioning as a Ca(2+)-regulated shuttle of ATP-Mg(2+) and P(i) across the inner mitochondrial membrane. Mice with an inactivated Slc25a25 gene have reduced metabolic efficiency as evidenced by enhanced resistance to diet-induced obesity and impaired exercise performance on a treadmill. Mouse embryo fibroblasts from Slc25a25(-/-) mice have reduced Ca(2+) flux across the endoplasmic reticulum, basal mitochondrial respiration, and ATP content. Although Slc25a25(-/-) mice are metabolically inefficient, the source of the inefficiency is not from a primary function in thermogenesis, because Slc25a25(-/-) mice maintain body temperature upon acute exposure to the cold (4 °C). Rather, the role of SLC25A25 in metabolic efficiency is most likely linked to muscle function as evidenced from the physical endurance test of mutant mice on a treadmill. Consequently, in the absence of SLC25A25 the efficiency of ATP production required for skeletal muscle function is diminished with secondary effects on adiposity. However, in the absence of UCP1-based thermogenesis, induction of Slc25a25 in mice with an intact gene may contribute to an alternative thermogenic pathway for the maintenance of body temperature during cold stress.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Metabolismo Energético/fisiologia , Proteínas Mitocondriais/metabolismo , Resistência Física/fisiologia , Termogênese/fisiologia , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Adiposidade/fisiologia , Animais , Proteínas de Ligação ao Cálcio/genética , Resposta ao Choque Frio/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Obesidade/genética , Obesidade/metabolismo , Condicionamento Físico Animal , Proteína Desacopladora 1
19.
J Neurophysiol ; 106(4): 1822-32, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21734104

RESUMO

Within the brain stem, the nucleus tractus solitarii (NTS) serves as a principal central site for sensory afferent integration from the cardiovascular and respiratory reflexes. Neuronal activity and synaptic transmission in the NTS are highly pliable and subject to neuromodulation. In the central nervous system, hydrogen sulfide (H2S) is a gasotransmitter generated primarily by the enzyme cystathionine-ß-synthase (CBS). We sought to determine the role of H2S, and its generation by CBS, in NTS excitability. Real-time RT-PCR, immunoblot, and immunohistochemistry analysis identified the presence of CBS in the NTS. Patch-clamp electrophysiology in brain stem slices examined excitatory postsynaptic currents (EPSCs) and membrane properties in monosynaptically driven NTS neurons. Confocal imaging of labeled afferent synaptic terminals in NTS slices monitored intracellular calcium. Exogenous H2S significantly increased the amplitude of evoked solitary tract (TS)-EPSCs, frequency of miniature (m)EPSCs, and presynaptic terminal calcium fluorescence in the NTS. H2S did not alter action potential discharge or postsynaptic properties. On the other hand, the CBS inhibitor aminooxyacetate (AOA) significantly reduced the amplitude of TS-EPSCs and presynaptic terminal calcium fluorescence in the NTS without altering postsynaptic properties. Taken together, these data support a presynaptic role for endogenous H2S in modulation of excitatory neurotransmission in the NTS.


Assuntos
Sulfeto de Hidrogênio/farmacologia , Núcleo Solitário/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Ácido Amino-Oxiacético/farmacologia , Animais , Sinalização do Cálcio , Cistationina beta-Sintase/antagonistas & inibidores , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Masculino , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/fisiologia , Técnicas de Patch-Clamp , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Núcleo Solitário/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA