Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 297(4): 101196, 2021 10.
Article in English | MEDLINE | ID: mdl-34529976

ABSTRACT

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.


Subject(s)
Calcium Signaling , Dynamins/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Dynamics , Cell Line , Dynamins/genetics , Fatty Acids/genetics , Fatty Acids/metabolism , Humans , Mitochondria, Muscle/genetics , Oxidation-Reduction
2.
J Neurophysiol ; 123(6): 2122-2135, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32347148

ABSTRACT

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.


Subject(s)
Astrocytes/physiology , Chemoreceptor Cells/physiology , Excitatory Postsynaptic Potentials/physiology , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamic Acid/metabolism , Hypoxia , Signal Transduction/physiology , Sleep Apnea, Obstructive , Solitary Nucleus , Animals , Disease Models, Animal , Glutamate Plasma Membrane Transport Proteins/antagonists & inhibitors , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Rats , Rats, Sprague-Dawley , Sleep Apnea, Obstructive/metabolism , Sleep Apnea, Obstructive/physiopathology , Solitary Nucleus/metabolism , Solitary Nucleus/physiopathology
3.
Am J Physiol Regul Integr Comp Physiol ; 318(6): R1068-R1077, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32320636

ABSTRACT

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.


Subject(s)
Astrocytes/drug effects , Blood Glucose , Neurons/drug effects , Rhombencephalon/drug effects , Thrombin/pharmacology , Animals , Male , Phrenic Nerve/drug effects , Rats , Rats, Sprague-Dawley , Respiratory Rate/drug effects , Solitary Nucleus/drug effects
4.
Transl Psychiatry ; 10(1): 90, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152264

ABSTRACT

Previous studies identify a role for hypothalamic glia in energy balance regulation; however, a narrow hypothalamic focus provides an incomplete understanding of how glia throughout the brain respond to and regulate energy homeostasis. We examined the responses of glia in the dorsal vagal complex (DVC) to the adipokine leptin and high fat diet-induced obesity. DVC astrocytes functionally express the leptin receptor; in vivo pharmacological studies suggest that DVC astrocytes partly mediate the anorectic effects of leptin in lean but not diet-induced obese rats. Ex vivo calcium imaging indicated that these changes were related to a lower proportion of leptin-responsive cells in the DVC of obese versus lean animals. Finally, we investigated DVC microglia and astroglia responses to leptin and energy balance dysregulation in vivo: obesity decreased DVC astrogliosis, whereas the absence of leptin signaling in Zucker rats was associated with extensive astrogliosis in the DVC and decreased hypothalamic micro- and astrogliosis. These data uncover a novel functional heterogeneity of astrocytes in different brain nuclei of relevance to leptin signaling and energy balance regulation.


Subject(s)
Hypothalamus , Leptin , Animals , Energy Metabolism , Hypothalamus/metabolism , Leptin/metabolism , Neuroglia/metabolism , Rats , Rats, Zucker
5.
Am J Physiol Regul Integr Comp Physiol ; 318(3): R545-R564, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31967862

ABSTRACT

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.


Subject(s)
Amino Acid Transport System X-AG/drug effects , Aspartic Acid/analogs & derivatives , Astrocytes/metabolism , Neurons/metabolism , Receptors, Metabotropic Glutamate/metabolism , Amino Acid Transport System X-AG/metabolism , Animals , Aspartic Acid/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Neurons/drug effects , Receptors, Metabotropic Glutamate/drug effects , Solitary Nucleus/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
6.
Am J Physiol Regul Integr Comp Physiol ; 318(1): R38-R48, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31596114

ABSTRACT

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.


Subject(s)
Astrocytes/physiology , Calcium/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Glucose/metabolism , Rhombencephalon/cytology , Type C Phospholipases/metabolism , Anilides/pharmacology , Animals , Antioxidants/pharmacology , Boron Compounds/pharmacology , Calcium/pharmacology , Dantrolene/pharmacology , Estrenes/pharmacology , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Phlorhizin/pharmacology , Prodrugs , Pyrrolidinones/pharmacology , Quercetin/pharmacology , Rats , Rats, Long-Evans , Type C Phospholipases/antagonists & inhibitors
7.
Physiol Behav ; 204: 140-150, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30797812

ABSTRACT

Hindbrain astrocytes are emerging as critical components in the regulation of homeostatic functions by either modulating synaptic activity or serving as primary detectors of physiological parameters. Recent studies have suggested that the glucose counter-regulation response (CRR), a critical defense against hypoglycemic emergencies, is dependent on glucoprivation-sensitive astrocytes in the hindbrain. This subpopulation of astrocytes produces a robust calcium signal in response to glucopenic stimuli. Both ex vivo and in vivo evidence suggest that low-glucose sensitive astrocytes utilize purinergic gliotransmission to activate catecholamine neurons in the hindbrain that are critical to the generation of the integrated CRR. Lastly, reports in the clinical literature suggest that an uncontrolled activation of CRR may as part of the pathology of severe traumatic injury. Work in our laboratory also suggests that this pathological hyperglycemia resulting from traumatic injury may be caused by the action of thrombin (generated by tissue trauma or bleeding) on hindbrain astrocytes. Similar to their glucopenia-sensitive neighbors, these hindbrain astrocytes may trigger hyperglycemic responses by their interactions with catecholaminergic neurons.


Subject(s)
Astrocytes/physiology , Glucose/metabolism , Rhombencephalon/cytology , Rhombencephalon/physiology , Animals , Homeostasis , Humans , Metabolism/physiology
8.
Am J Physiol Regul Integr Comp Physiol ; 315(1): R153-R164, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29590557

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Calcium Signaling , Catecholamines/metabolism , Glucose/deficiency , Neurons/metabolism , Rhombencephalon/metabolism , Animals , Female , Genes, Reporter , Glutamic Acid/metabolism , Immunohistochemistry , In Vitro Techniques , Male , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Receptors, Purinergic P2/metabolism , Rhombencephalon/cytology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
9.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1102-8, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27101298

ABSTRACT

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.


Subject(s)
Adenosine/metabolism , Blood Glucose/metabolism , Deoxyglucose/administration & dosage , Hypoglycemia/metabolism , Rhombencephalon/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blood Glucose/drug effects , Female , Homeostasis/drug effects , Infusions, Intraventricular , Male , Rats , Rats, Long-Evans , Rhombencephalon/pathology , Up-Regulation/drug effects
10.
J Neurosci ; 36(12): 3531-40, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27013681

ABSTRACT

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.


Subject(s)
Appetite Regulation/physiology , Astrocytes/physiology , Feeding Behavior/physiology , Glucagon-Like Peptide-1 Receptor/metabolism , Homeostasis/physiology , Medulla Oblongata/metabolism , Animals , Energy Metabolism/physiology , Feedback, Physiological/physiology , Male , Rats , Rats, Long-Evans , Rats, Sprague-Dawley
11.
Am J Physiol Endocrinol Metab ; 309(8): E715-26, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26306596

ABSTRACT

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.


Subject(s)
Chemokines/metabolism , Diabetes Mellitus/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Interleukin-1beta/metabolism , NF-kappa B/metabolism , RNA, Messenger/metabolism , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Chemokines/genetics , Electron Spin Resonance Spectroscopy , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoblotting , Insulin/genetics , Insulin Secretion , Insulinoma , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxygen Consumption , Pancreatic Neoplasms , Patch-Clamp Techniques , Rats , Rats, Wistar , Rats, Zucker , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S9 , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Tumor Cells, Cultured
12.
J Neurosci ; 35(2): 776-85, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25589770

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Neurons, Afferent/physiology , Receptor, PAR-1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Solitary Nucleus/physiology , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Aspartic Acid/pharmacology , Astrocytes/drug effects , Calcium/metabolism , Citrates/pharmacology , Excitatory Amino Acid Antagonists , Excitatory Postsynaptic Potentials , Female , Glutamic Acid/metabolism , Male , Miniature Postsynaptic Potentials , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Peptide Fragments/pharmacology , Rats , Rats, Long-Evans , Receptor, PAR-1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Solitary Nucleus/cytology , Solitary Nucleus/metabolism , Synapses/metabolism , Synapses/physiology , Vagus Nerve/cytology , Vagus Nerve/metabolism , Vagus Nerve/physiology
13.
Brain Res ; 1595: 84-91, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25446446

ABSTRACT

Previous behavioral studies have demonstrated that presynaptic N-methyl-d-aspartate (NMDA) receptors expressed on vagal afferent terminals are involved in food intake and satiety. Therefore, using in vitro live cell calcium imaging of prelabeled rat hindbrain slices, we characterized which NMDA receptor GluN2 subunits may regulate vagal afferent activity. The nonselective NMDA receptor antagonist d,l-2-amino-5-phosphonopentanoic acid (d,l-AP5) significantly inhibited vagal terminal calcium influx, while the excitatory amino acid reuptake inhibitor d,l-threo-ß-benzyloxyaspartic acid (TBOA), significantly increased terminal calcium levels following pharmacological stimulation with ATP. Subunit-specific NMDA receptor antagonists and potentiators were used to identify which GluN2 subunits mediate the NMDA receptor response on the vagal afferent terminals. The GluN2B-selective antagonist, ifenprodil, selectively reduced vagal calcium influx with stimulation compared to the time control. The GluN2A-selective antagonist, 3-chloro-4-fluoro-N-[4-[[2-(phenylcarbonyl)hydrazino]carbonyl] benzyl]benzenesulfonamide (TCN 201) produced smaller but not statistically significant effects. Furthermore, the GluN2A/B-selective potentiator (pregnenolone sulfate) and the GluN2C/D-selective potentiator [(3-chlorophenyl)(6,7-dimethoxy-1-((4-methoxyphenoxy)methyl)-3,4-dihydroisoquinolin-2(1H)-yl)methanone; (CIQ)] enhanced vagal afferent calcium influx during stimulation. These data suggest that presynaptic NMDA receptors with GluN2B, GluN2C, and GluN2D subunits may predominantly control vagal afferent excitability in the nucleus of the solitary tract.


Subject(s)
Afferent Pathways/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Solitary Nucleus/physiology , Vagus Nerve/physiology , 2-Amino-5-phosphonovalerate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Aspartic Acid/pharmacology , Benzimidazoles/pharmacology , Calcium/metabolism , Excitatory Amino Acid Antagonists/pharmacology , In Vitro Techniques , Piperidines/pharmacology , Pregnenolone/pharmacology , Rats , Rats, Long-Evans , Solitary Nucleus/drug effects , Sulfonamides/pharmacology
14.
Neurosci Lett ; 582: 115-9, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25220699

ABSTRACT

Artemisia extracts have been used as remedies for a variety of maladies related to metabolic and gastrointestinal control. Because the vagal afferent-nucleus of the solitary tract (NST) synapse regulates the same homeostatic functions affected by Artemisia, it is possible that these extracts may have activity at the synaptic level in the NST. Therefore, we evaluated how extracts of three common medicinal Artemisia species, Artemisia santolinifolia (SANT), Artemisia scoparia (SCO), and Artemisia dracunculus L (PMI-5011), modulate the excitability of the glutamatergic vagal afferent-NST synapse. Our in vitro live cell calcium imaging data from prelabeled vagal afferent terminals show that SANT extract is a positive modulator of vagal afferent calcium levels, as the extract significantly increased the calcium signal relative to the time control. Neither SCO nor PMI-5011 extract altered the vagal calcium signals compared to the time control. Furthermore, whole cell voltage-clamp recordings from NST neurons corroborated the vagal terminal calcium data in that SANT extract also significantly increased miniature excitatory postsynaptic current (mEPSC) frequency in NST neurons. These data suggest that SANT extract could be a pharmacologically significant mediator of glutamatergic neurotransmission within the CNS.


Subject(s)
Artemisia/chemistry , Glutamic Acid/metabolism , Plant Extracts/pharmacology , Solitary Nucleus/drug effects , Synaptic Transmission/drug effects , Afferent Pathways , Animals , Calcium Signaling , Excitatory Postsynaptic Potentials , In Vitro Techniques , Miniature Postsynaptic Potentials , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Rats, Long-Evans , Solitary Nucleus/physiology , Vagus Nerve/physiology
15.
J Neurosci ; 34(32): 10488-96, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25100584

ABSTRACT

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.


Subject(s)
Citrates/pharmacology , Gastrointestinal Motility/drug effects , Neurons/drug effects , Reflex/drug effects , Rhombencephalon/cytology , Vagus Nerve/drug effects , Action Potentials/drug effects , Analysis of Variance , Animals , Deoxyglucose/pharmacology , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Insulin/pharmacology , Neurons/physiology , Rats , Reflex/physiology , Vagus Nerve/physiology
16.
Nutrition ; 30(7-8 Suppl): S37-42, 2014.
Article in English | MEDLINE | ID: mdl-24985104

ABSTRACT

OBJECTIVE: St. John's Wort (SJW) extract, which is commonly used to treat depression, inhibits the reuptake of several neurotransmitters, including glutamate, serotonin, norepinephrine, and dopamine. Glutamatergic visceral vagal afferents synapse upon neurons of the solitary tract (NST); thus, the aim of this study was to evaluate whether SJW extract modulates glutamatergic neurotransmission within the NST. METHODS: We used live cell calcium imaging to evaluate whether SJW and its isolated components hypericin and hyperforin increase the excitability of prelabeled vagal afferent terminals synapsing upon the NST. We used voltage-clamp recordings of spontaneous miniature excitatory postsynaptic currents (mEPSCs) to evaluate whether SJW alters glutamate release from vagal afferents onto NST neurons. RESULTS: Our imaging data show that SJW (50 µg/mL) increased the intracellular calcium levels of stimulated vagal afferent terminals compared with the bath control. This increase in presynaptic vagal afferent calcium by the extract coincides with an increase in neurotransmitter release within the nucleus of the solitary tract, as the frequency of mEPSCs is significantly higher in the presence of the extract compared with the control. Finally, our imaging data show that hyperforin, a known component of SJW extract, also significantly increases terminal calcium levels. CONCLUSION: These data suggest that SJW extract can significantly increase the probability of glutamate release from vagal afferents onto the NST by increasing presynaptic calcium. The in vitro vagal afferent synapse with NST neurons is an ideal model system to examine the mechanism of action of botanical agents on glutamatergic neurotransmission.


Subject(s)
Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Hypericum/chemistry , Plant Extracts/pharmacology , Solitary Nucleus/drug effects , Synaptic Transmission/drug effects , Animals , Anthracenes , Calcium/metabolism , Depression , Female , Male , Neurons, Afferent/metabolism , Perylene/analogs & derivatives , Perylene/pharmacology , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Rats, Long-Evans , Synapses , Terpenes/pharmacology , Vagus Nerve/metabolism
17.
Auton Neurosci ; 175(1-2): 61-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23313342

ABSTRACT

Glucoprivation is a strong signal for the initiation of gastrointestinal contractions. While this relationship between utilizable nutrient levels and gastric motility has been recognized for more than 100 years, the explanation of this phenomenon has remained incomplete. Using widely differing approaches, recent work has suggested that the hindbrain is responsible for this chemoreflex effect. Surprisingly, astrocytes may be the main glucodetector elements under hypoglycemic conditions. Our own work using in vitro live cell calcium imaging shows that astrocytes in the NST increase cytoplasmic calcium in a concentration dependent manner in reaction to reductions in glucose. This effect is reversed on restoration of normal glucose concentrations. In vivo single unit neurophysiological recordings show that brainstem neurons driving gastric motility are activated by glucoprivic stimuli. Studies in intact animals verify that both dorsal medullary and systemic glucoprivation significantly increases gastric motility. Astrocyte inactivation with fluorocitrate blocks the pro-motility effects of glucoprivation. Thus, it appears that intact astrocyte signaling may be essential to glucoregulatory control over gastric motility.


Subject(s)
Astrocytes/physiology , Gastrointestinal Motility/physiology , Glucose/metabolism , Rhombencephalon/physiology , Animals , Blood Glucose/physiology , Chemoreceptor Cells/physiology , Humans
18.
Brain Res ; 1492: 46-52, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23178697

ABSTRACT

Previous studies from our laboratory illustrated the potential for stromal cell-derived factor one [CXCL12; also referred to as SDF-1] to act on its receptor [CXCR4] within the dorsal vagal complex [DVC] of the hindbrain to suppress gastric motility (Hermann et al., 2008). While CXCR4 receptors are essential for normal brain development, they also play a critical role in the proliferation of the HIV virus and initiation of metastatic cell growth in the brain. Anorexia, nausea, and failed autonomic regulation of gastrointestinal function are significant causes of morbidity and are contributory factors in the mortality associated with these disease states. The implication of our previous study was that CXCL12 caused gastric stasis by acting on gastric reflex circuit elements in the DVC. This hindbrain complex includes vagal afferent terminations in the solitary nucleus, neurons in the solitary nucleus (NST) and visceral efferent motorneurons in the dorsal motor nucleus (DMN) that are responsible for the regulation of digestive functions from the oral cavity to the transverse colon. In the current study, in vivo single-unit neurophysiological recordings from physiologically-identified NST and DMN components of the gastric accommodation reflex show that while injection of femtomole doses of CXCL12 onto NST or DMN neurons has no effect on their basal activity, CXCL12 amplifies the effect of gastric vagal mechanosensory input to activate the NST and, in turn, inhibit DMN motor activity.


Subject(s)
Chemokine CXCL12/metabolism , Gastrointestinal Motility/physiology , Medulla Oblongata/metabolism , Neurons, Efferent/metabolism , Neurons/metabolism , Animals , Chemokine CXCL12/pharmacology , Female , Male , Medulla Oblongata/drug effects , Neurons/drug effects , Neurons, Efferent/drug effects , Patch-Clamp Techniques , Rats , Rats, Long-Evans , Reflex/drug effects , Reflex/physiology , Vagus Nerve/drug effects , Vagus Nerve/metabolism
19.
Front Neurosci ; 7: 249, 2013.
Article in English | MEDLINE | ID: mdl-24391532

ABSTRACT

Glucose homeostasis is maintained through interplay between central and peripheral control mechanisms which are aimed at storing excess glucose following meals and mobilizing these same stores during periods of fasting. The nucleus of the solitary tract (NST) in the dorsal medulla has long been associated with the central detection of glucose availability and the control of glucose homeostasis. Recent evidence has emerged which supports the involvement of astrocytes in glucose homeostasis. The aim of the present study was to investigate whether NST-astrocytes respond to physiologically relevant decreases in glucose availability, in vitro, as well as to the presence of the glucoprivic compound 2-deoxy-D-Glucose. This report demonstrates that some NST-astrocytes are capable of responding to low glucose or glucoprivation by increasing cytoplasmic calcium; a change that reverses with restoration of normal glucose availability. While some NST-neurons also demonstrate an increase in calcium signaling during low glucose availability, this effect is smaller and somewhat delayed compared to those observed in adjacent astrocytes. TTX did not abolish these hypoglycemia mediated responses of astrocytes, suggesting that NST-astrocytes may be directly sensing low glucose levels as opposed to responding to neuronal detection of hypoglycemia. Thus, chemodetection of low glucose by NST-astrocytes may play an important role in the autonomic regulation of glucose homeostasis.

20.
J Neurosci ; 32(15): 5237-41, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496569

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Cannabinoids/pharmacology , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Vagus Nerve/drug effects , Vagus Nerve/physiology , Animals , Brain Stem/physiology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Diffusion Chambers, Culture , Electric Stimulation , Female , Image Processing, Computer-Assisted , Isoquinolines/pharmacology , Male , Microscopy, Confocal , Presynaptic Terminals/drug effects , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Long-Evans , Receptor, Cannabinoid, CB1/agonists , Solitary Nucleus/physiology , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...