Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
J Neurophysiol ; 84(3): 1636-44, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980033

ABSTRACT

Previous studies have suggested that voltage-gated Ca(2+) influx in glomus cells plays a critical role in sensory transduction at the carotid body chemoreceptors. The purpose of the present study was to determine the effects of hypoxia on the Ca(2+) current in glomus cells and to elucidate the underlying mechanism(s). Experiments were performed on freshly dissociated glomus cells from rabbit carotid bodies. Ca(2+) current was monitored using the whole cell configuration of the patch-clamp technique, with Ba(2+) as the charge carrier. Hypoxia (pO(2) = 40 mmHg) augmented the Ca(2+) current by 24 +/- 3% (n = 42, at 0 mV) in a voltage-independent manner. This effect was seen in a CO(2)/HCO(3)(-)-, but not in a HEPES-buffered extracellular solution at pH 7.4 (n = 6). When the pH of a HEPES-buffered extracellular solution was lowered from 7.4 to 7. 0, hypoxia augmented the Ca(2+) current by 20 +/- 5% (n = 4, at 0 mV). Nisoldipine, an L-type Ca(2+) channel blocker (2 microM, n = 6), prevented, whereas, omega-conotoxin MVIIC (2 microM, n = 6), an inhibitor of N and P/Q type Ca(2+) channels, did not prevent augmentation of the Ca(2+) current by hypoxia, implying that low oxygen affects L-type Ca(2+) channels in glomus cells. Protein kinase C (PKC) inhibitors, staurosporine (100 nM, n = 6) and bisindolylmaleimide (2 microM, n = 8, at 0 mV), prevented, whereas, a protein kinase A inhibitor (4 nM PKAi, n = 10) did not prevent the hypoxia-induced increase of the Ca(2+) current. Phorbol 12-myristate 13-acetate (PMA, 100 nM), a PKC activator, augmented the Ca(2+) current by 20 +/- 3% (n = 8, at 0 mV). In glomus cells treated with PMA overnight (100 nM), hypoxia did not augment the Ca(2+) current (-3 + 4%, n = 5, at 0 mV). Immunocytochemical analysis revealed PKCdelta-like immunoreactivity in the cytosol of the glomus cells. Following hypoxia (6% O(2) for 5 min), PKCdelta-like immunoreactivity translocated to the plasma membrane in 87 +/- 3% of the cells, indicating PKC activation. These results demonstrate that hypoxia augments Ca(2+) current through L-type Ca(2+) channels via a PKC-sensitive mechanism.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Carotid Body/metabolism , Cell Hypoxia/physiology , Protein Kinase C/metabolism , Animals , Bicarbonates/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Carotid Body/cytology , Carotid Body/drug effects , Cell Membrane/enzymology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cytosol/enzymology , Extracellular Space/metabolism , HEPES/pharmacology , Ion Transport/drug effects , Ion Transport/physiology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Nisoldipine/pharmacology , Patch-Clamp Techniques , Protein Kinase C/antagonists & inhibitors , Rabbits , Signal Transduction/physiology , Tetradecanoylphorbol Acetate/pharmacology
3.
Respir Physiol ; 122(2-3): 209-21, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10967345

ABSTRACT

The purpose of this article is to highlight some recent concepts on oxygen sensing mechanisms at the carotid body chemoreceptors. Most available evidence suggests that glomus (type I) cells are the initial site of transduction and they release transmitters in response to hypoxia, which in turn depolarize the nearby afferent nerve ending, leading to an increase in sensory discharge. Two main hypotheses have been advanced to explain the initiation of the transduction process that triggers transmitter release. One hypothesis assumes that a biochemical event associated with a heme protein triggers the transduction cascade. Supporting this idea it has been shown that hypoxia affects mitochondrial cytochromes. In addition, there is a body of evidence implicating non-mitochondrial enzymes such as NADPH oxidases, NO synthases and heme oxygenases located in glomus cells. These proteins could contribute to transduction via generation of reactive oxygen species, nitric oxide and/or carbon monoxide. The other hypothesis suggests that a K(+) channel protein is the oxygen sensor and inhibition of this channel and the ensuing depolarization is the initial event in transduction. Several oxygen sensitive K(+) channels have been identified. However, their roles in initiation of the transduction cascade and/or cell excitability are unclear. In addition, recent studies indicate that molecular oxygen and a variety of neurotransmitters may also modulate Ca(2+) channels. Most importantly, it is possible that the carotid body response to oxygen requires multiple sensors, and they work together to shape the overall sensory response of the carotid body over a wide range of arterial oxygen tensions.


Subject(s)
Carotid Body/physiology , Chemoreceptor Cells/physiology , Hemeproteins/physiology , Ion Channels/physiology , Oxygen/physiology , Animals , Carotid Body/enzymology , Chemoreceptor Cells/enzymology , Humans , Oxygen/blood , Oxygen Consumption/physiology
4.
Adv Exp Med Biol ; 475: 101-9, 2000.
Article in English | MEDLINE | ID: mdl-10849652

ABSTRACT

Organisms respond to hypoxia through detection of blood oxygen levels by sensors at peripheral chemoreceptors and by receptors in certain key cells of the body. The pathways over which peripheral chemoreceptor signals are transmitted to respiratory muscles are well established. However, the intracellular pathways that transmit hypoxic stimulus to gene activation are just being identified. Using anti-sense c-fos strategy, we have shown that c-fos is essential for the activation of activator protein-1 transcription factor complex (AP-1) and subsequent stimulation of downstream genes such as tyrosine hydroxylase (TH; Mishra et al. 1998). The purpose of the present study was to identify intracellular pathways that link hypoxia to activation of c-fos. The results of the present study show that hypoxia causes Ca2+ influx through L-type voltage gated Ca2+ channels and that hypoxia-induced c-fos gene expression is Ca2+/calmodulin dependent. We also demonstrate that hypoxia activates the extracellular-regulated kinase (ERK) and p38, but not JNK. Further, phosphorylation of ERK is essential for c-fos activation via SRE cis-element. Further characterization of nuclear signalling pathways provides evidence for the involvement of Src, a non receptor protein tyrosine kinase, and Ras, a small G protein, in the hypoxia-induced c-fos gene expression. These results suggest a possible role for non-receptor protein tyrosine kinases in propagating signals from G-protein coupled receptors to the activation of immediate early genes such as c-fos during hypoxia.


Subject(s)
Genes, fos , Hypoxia/genetics , Hypoxia/metabolism , Transcription Factor AP-1/metabolism , Animals , Calcium Channels/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Mitogen-Activated Protein Kinases/metabolism , PC12 Cells , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Rats , Signal Transduction , Transcriptional Activation , p38 Mitogen-Activated Protein Kinases , src-Family Kinases/metabolism
5.
Adv Exp Med Biol ; 475: 241-8, 2000.
Article in English | MEDLINE | ID: mdl-10849664

ABSTRACT

Currently, it is not clear what type of K+ channel(s) is active at the resting membrane potential (RMP) in glomus cells of the carotid body (CB). HERG channels produce currents that are known to contribute to the RMP in other neuronal cells. The goal of the present study was to determine whether CB glomus cells express HERG-like (HL) K+ current, and if so, to determine whether HL currents regulate the RMP. With high [K+]o, depolarizing voltage steps from -85 mV revealed a slowly deactivating inward tail current indicative of HL K+ current in whole-cell, voltage clamped glomus cells. The HL currents were blocked by dofetilide (DOF) in a concentration-dependent manner (IC50 = 13 nM) and high concentrations (1 and 10 mM) of Ba2+. The steady-state activation properties of the HL current (Vh = -45 mV) suggest that it is active at the RMP in glomus cells. Whole-cell, current clamped glomus cells exhibited a RMP of -48 mV. 150 nM DOF caused a significant (14 mV) depolarizing shift in the RMP. In isolated glomus cells, [Ca2+]i increased in response to DOF (1 microM). In an in-vitro CB preparation, DOF increased basal sensory discharge in a concentration-dependent manner and significantly attenuated the sensory response to hypoxia. These results suggest that the HERG-like current is responsible for controlling the RMP in glomus cells of the rabbit CB, and that it is involved in the chemosensory response to hypoxia of the CB.


Subject(s)
Carotid Body/metabolism , Cation Transport Proteins , Chemoreceptor Cells/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Animals , Calcium/metabolism , Carotid Body/cytology , Carotid Body/drug effects , Cell Hypoxia/physiology , Chemoreceptor Cells/drug effects , Ether-A-Go-Go Potassium Channels , In Vitro Techniques , Membrane Potentials/drug effects , Phenethylamines/pharmacology , Potassium Channel Blockers , Rabbits , Sulfonamides/pharmacology
6.
Adv Exp Med Biol ; 475: 589-99, 2000.
Article in English | MEDLINE | ID: mdl-10849699

ABSTRACT

Several lines of evidence indicate that transduction of the hypoxic stimulus at the carotid body involves an increase in cytosolic Ca2+ ([Ca2+]i) via activation of voltage-gated Ca2+ channels in the glomus cells. However, reported responses to hypoxia include either no effect on or inhibition of Ca2+ current in glomus cells. The apparent discrepancy between the effects of hypoxia on [Ca2+]i and Ca2+ channel activity prompted us to re-examine the effects of low oxygen on Ca2+ currents in glomus cells. Experiments were performed on freshly dissociated glomus cells from rabbit carotid bodies. Ca2+ channel activity was monitored using the whole-cell configuration of the patch clamp technique with Ba2+ as the charge carrier. Hypoxia (pO2 = 40 mmHg) augmented the Ca2+ current by 24% (at 0 mV). This augmentation was seen in a CO2/HCO3- but not in a HEPES buffered extracellular solution. However, when the extracellular pH (pHo) of a HEPES buffered solution is lowered from 7.4 to 7.0, then the Ca2+ current in glomus cells is augmented by hypoxia by 20%. Nisoldipine, an L-type Ca2+ channel blocker (2 microM), prevented augmentation of the Ca2+ current by hypoxia. On the other hand, an N- and P-type Ca2+ channel blocker (2 microM omega-conotoxin MVIIC) did not prevent the augmentation of the Ca2+ current by hypoxia. Protein kinase C (PKC) inhibitors, staurosporine (100 nM) and bisindolylmaleimide (2 microM), prevented augmentation by hypoxia. Okadaic acid (100 nM), an inhibitor of serine/threonine phosphatases also prevented augmentation of Ca2+ current by hypoxia; whereas, norokadaone, an inactive analog of okadaic acid, had no effect. These results suggest that hypoxia augments Ca2+ current through L-type Ca2+ channels via a PKC and/or phosphatase-sensitive pathways.


Subject(s)
Calcium/metabolism , Carotid Body/metabolism , Cell Hypoxia/physiology , Animals , Buffers , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Carotid Body/cytology , Carotid Body/drug effects , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , In Vitro Techniques , Indoles/pharmacology , Maleimides/pharmacology , Membrane Potentials , Nisoldipine/pharmacology , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rabbits , Staurosporine/pharmacology
7.
Adv Exp Med Biol ; 475: 705-13, 2000.
Article in English | MEDLINE | ID: mdl-10849712

ABSTRACT

Carotid body expresses neutral endopeptidase (NEP)-like enzyme activity and phosphoramidon, an inhibitor of NEP augments sensory response of the carotid body to hypoxia (Kumar et al., 1990). NEP hydrolyzes substance P (SP) and methionine enkephalin (Met-ENK) in the nervous system. In the present study, we determined whether NEP hydrolyzes Met-ENK and SP in the carotid body and whether these peptides contribute to the phosphoramidon-induced potentiation of the sensory response to hypoxia. Experiments were performed on carotid bodies excised from anaesthetized adult cats. HPLC analysis showed that both SP and Met-ENK were hydrolyzed by the carotid body. Phosphoramidon (400 microM) markedly inhibited SP (approximately 90%) but had only marginal effect on Met-ENK hydrolysis (approximately 15%). Sensory responses of the carotid body in vitro to hypoxia (pO2, 68 +/- 6 mmHg) and SP (10 nmoles) were potentiated by phosphoramidon by approximately 80% and approximately 275% respectively (p < 0.01). SP-receptor antagonist abolished phosphoramidon-induced potentiation of the sensory response to hypoxia as well as to SP. These results demonstrate that SP is a preferred substrate for NEP in the carotid body and SP plays a major role in the potentiation of the hypoxic response of the carotid body by phosphoramidon.


Subject(s)
Carotid Body/metabolism , Hypoxia/metabolism , Neprilysin/metabolism , Substance P/metabolism , Animals , Carotid Body/drug effects , Cats , Enkephalin, Methionine/metabolism , Female , Glycopeptides/pharmacology , Hydrolysis , In Vitro Techniques , Male , Neurokinin-1 Receptor Antagonists , Substance P/analogs & derivatives , Substance P/pharmacology
8.
J Appl Physiol (1985) ; 88(5): 1898-906, 2000 May.
Article in English | MEDLINE | ID: mdl-10797155

ABSTRACT

In the present study we examined the intracellular pathways that link hypoxia to activation of c-fos gene expression. Experiments were performed on rat pheocromocytoma-12 (PC-12) cells. c-fos mRNA and promoter activities were analyzed by RT-PCR and reporter gene assays, respectively. BAPTA, a Ca(2+) chelator, inhibited c-fos mRNA and promoter activation by hypoxia. Nitrendipine, an L-type Ca(2+)-channel blocker, abolished, whereas BAY K 8644, an L-type channel agonist, enhanced c-fos activation by hypoxia. Ca(2+) currents were augmented reversibly by hypoxia, suggesting that Ca(2+) influx mediated by L-type Ca(2+) channels is essential for c-fos activation by hypoxia. We next determined downstream pathways activated by intracellular Ca(2+) concentration. Immunoblot analysis revealed Ca(2+)/calmodulin-dependent kinase II (CaMKII) protein in PC-12 cells and revealed that hypoxia increased the enzyme activity. KN-93, a CaMK inhibitor, blocked CaMKII activation and c-fos promoter stimulation by hypoxia. Ectopic expression of an active mutant of CaMKII (pCaMKII290) stimulated c-fos promoter activity under normoxia. Hypoxia increased phosphorylation of CREB at the serine residue 133 (Ser-133), and KN-93 attenuated this effect. Point mutations at the Ca(2+)/cAMP-responsive cis-element (Ca/CRE) attenuated, whereas point mutations in the serum-responsive cis-element (SRE) abolished transcriptional activation of c-fos by hypoxia. These results demonstrate that c-fos activation by hypoxia involves CaMK activation and CREB phosphorylation at Ser-133 and requires Ca/CRE and SRE. These observations demonstrate that Ca(2+)-dependent signaling pathways play a crucial role in induction of c-fos gene expression, which may underlie long-term adaptive responses to hypoxia.


Subject(s)
Calcium Channels, L-Type/physiology , Gene Expression Regulation/physiology , Hypoxia/physiopathology , Proto-Oncogene Proteins c-fos/genetics , Transcription, Genetic/physiology , Animals , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Chelating Agents/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Hypoxia/genetics , PC12 Cells , Phosphorylation , Point Mutation/physiology , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Rats
9.
J Neurophysiol ; 83(3): 1150-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712445

ABSTRACT

Direct evidence for a specific K(+) channel underlying the resting membrane potential in glomus cells of the carotid body has been absent. The product of the human ether-a-go-go-related gene (HERG) produces inward rectifier currents that are known to contribute to the resting membrane potential in other neuronal cells. The goal of the present study was to determine whether carotid body glomus cells express HERG-like K(+) current, and if so, to determine whether a HERG-like current regulates the resting membrane potential. Freshly dissociated rabbit glomus cells under whole cell voltage clamp exhibited slowly decaying outward currents that activated 20-30 mV positive to the resting membrane potential. Raising extracellular K(+) revealed a slowly deactivating inward tail current indicative of HERG-like K(+) current. HERG-like currents were not found in cells resembling type II cells. The HERG-like current was blocked by dofetilide (DOF) in a concentration-dependent manner (IC(50) = 13 +/- 4 nM, mean +/- SE) and high concentrations of Ba(2+) (1 and 10 mM). The biophysical and pharmacological characteristics of this inward tail current suggest that it is conducted by a HERG-like channel. The steady-state activation properties of the HERG-like current (V(h) = -44 +/- 2 mV) suggest that it is active at the resting membrane potential in glomus cells. In whole cell, current-clamped glomus cells (average resting membrane potential, - 48 +/- 4 mV), DOF, but not tetraethylammonium, caused a significant (13 mV) depolarizing shift in the resting membrane potential. Using fluorescence imaging, DOF increased [Ca(2+)](i) in isolated glomus cells. In an in-vitro carotid body preparation, DOF increased basal sensory discharge in the carotid sinus nerve in a concentration-dependent manner. These results demonstrate that glomus cells express a HERG-like current that is active at, and responsible for controlling the resting membrane potential.


Subject(s)
Carotid Body/physiology , Cation Transport Proteins , Neurons/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Animals , Barium/metabolism , Calcium/metabolism , Carotid Body/cytology , Electrophysiology , Ether-A-Go-Go Potassium Channels , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Patch-Clamp Techniques , Phenethylamines/pharmacology , Potassium Channel Blockers , Rabbits , Sulfonamides/pharmacology
10.
J Appl Physiol (1985) ; 88(1): 195-202, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10642381

ABSTRACT

Previously, we showed that carotid bodies express neutral endopeptidase (NEP)-like enzyme activity and that phosphoramidon, a potent inhibitor of NEP, potentiates the chemosensory response of the carotid body to hypoxia in vivo. NEP has been shown to hydrolyze methionine enkephalin (Met-Enk) and substance P (SP) in neuronal tissues. The purpose of the present study is to determine whether NEP hydrolyzes Met-Enk and SP in the carotid body and if so whether these peptides contribute to phosphoramidon-induced potentiation of the sensory response to hypoxia. Experiments were performed on carotid bodies excised from anesthetized adult cats (n = 72 carotid bodies). The hydrolysis of Met-Enk and SP was analyzed by HPLC. The results showed that both SP and Met-Enk were hydrolyzed by the carotid body, but the rate of Met-Enk hydrolysis was approximately fourfold higher than that of SP. Phosphoramidon (400 microM) markedly inhibited SP hydrolysis ( approximately 90%) but had only a marginal effect on Met-Enk hydrolysis ( approximately 15% inhibition). Hypoxia (PO(2), 68 +/- 6 Torr) as well as exogenous administration of SP (10 and 20 nmol) increased the sensory discharge of the carotid body in vitro. Sensory responses to hypoxia and SP (10 nmol) were potentiated by approximately 80 and approximately 275%, respectively (P < 0.01), in the presence of phosphoramidon. SP-receptor antagonists Spantide (peptidyl) and CP-96345 (nonpeptidyl) either abolished or markedly attenuated the phosphoramidon-induced potentiation of the sensory response of the carotid body to hypoxia as well as to SP. These results demonstrate that SP is a preferred substrate for NEP in the carotid body and that SP is involved in the potentiation of the hypoxic response of the carotid body by phosphoramidon.


Subject(s)
Carotid Body/enzymology , Carotid Body/physiology , Neprilysin/metabolism , Oxygen/physiology , Substance P/metabolism , Animals , Biphenyl Compounds/pharmacology , Carotid Body/drug effects , Carotid Body/metabolism , Cats , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Enkephalin, Methionine/metabolism , Female , Glycopeptides/antagonists & inhibitors , Glycopeptides/pharmacology , Hydrolysis/drug effects , Hypoxia/physiopathology , Kinetics , Male , Neprilysin/antagonists & inhibitors , Neurokinin-1 Receptor Antagonists , Receptors, Neurokinin-1/metabolism , Substance P/analogs & derivatives , Substance P/antagonists & inhibitors , Substance P/pharmacology
11.
J Neurophysiol ; 81(4): 1449-57, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10200181

ABSTRACT

Previous studies have shown that nitric oxide (NO) inhibits carotid body sensory activity. To begin to understand the cellular mechanisms associated with the actions of NO in the carotid body, we monitored the effects of NO donors on the macroscopic Ca2+ current in glomus cells isolated from rabbit carotid bodies. Experiments were performed on freshly dissociated glomus cells from adult rabbit carotid bodies using the whole cell configuration of the patch-clamp technique. The NO donors sodium nitroprusside (SNP; 600 microM, n = 7) and spermine nitric oxide (SNO; 100 microM, n = 7) inhibited the Ca2+ current in glomus cells in a voltage-independent manner. These effects of NO donors were rapid in onset and peaked within 1 or 2 min. In contrast, the outward K+ current was unaffected by SNP (600 microM, n = 6), indicating that the inhibition by SNP was not a nonspecific membrane effect. 2-(4-carboxyphenyl)-4,4,5, 5-tetramethyl-imidazoline-1-oxyl-3-oxide (carboxy-PTIO; 500 microM), an NO scavenger, prevented inhibition of the Ca2+ current by SNP (n = 7), whereas neither superoxide dismutase (SOD; 2,000 U/ml, n = 4), a superoxide scavenger, nor sodium hydrosulfite (SHS; 1 mM, n = 7), a reducing agent, prevented inhibition of the Ca2+ current by SNP. However, SNP inhibition of the Ca2+ current was reversible in the presence of either SOD or SHS. These results suggest that NO itself inhibits Ca2+ current in a reversible manner and that subsequent formation of peroxynitrites results in irreversible inhibition. SNP inhibition of the Ca2+ current was not affected by 30 microM LY 83, 583 (n = 7) nor was it mimicked by 600 microM 8-bromoguanosine 3':5'-cyclic monophosphate (8-Br-cGMP; n = 6), suggesting that the effects of NO on the Ca2+ current are mediated, in part, via a cGMP-independent mechanism. N-ethylmaleimide (NEM; 2.5 mM, n = 6) prevented the inhibition of the Ca2+ current by SNP, indicating that SNP is acting via a modification of sulfhydryl groups on Ca2+ channel proteins. Norepinephrine (NE; 10 microM) further inhibited the Ca2+ current in the presence of NEM (n = 7), implying that NEM did not nonspecifically eliminate Ca2+ current modulation. Nisoldipine, an L-type Ca2+ channel blocker (2 microM, n = 6), prevented the inhibition of Ca2+ current by SNP, whereas omega-conotoxin GVIA, an N-type Ca2+ channel blocker (1 microM, n = 9), did not prevent the inhibition of Ca2+ current by SNP. These results demonstrate that NO inhibits L-type Ca2+ channels in adult rabbit glomus cells, in part, due to a modification of calcium channel proteins. The inhibition might provide one plausible mechanism for efferent inhibition of carotid body activity by NO.


Subject(s)
Calcium Channels/physiology , Carotid Body/chemistry , Cyclic GMP/metabolism , Ion Channel Gating/drug effects , Nitric Oxide/pharmacology , Aminoquinolines/pharmacology , Animals , Calcium/metabolism , Calcium Channels, L-Type , Carotid Body/cytology , Carotid Body/drug effects , Cells, Cultured , Cyanides/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dithionite/pharmacology , Electrophysiology , Enzyme Inhibitors/pharmacology , Ethylmaleimide/pharmacology , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nitrites/metabolism , Nitroprusside/pharmacology , Potassium/metabolism , Potassium Channels/physiology , Rabbits , Superoxide Dismutase/pharmacology , Vasodilator Agents/pharmacology
12.
J Neurophysiol ; 81(1): 225-33, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9914283

ABSTRACT

Previous studies have demonstrated that endogenous norepinephrine (NE) inhibits carotid body (CB) sensory discharge, and the cellular actions of NE have been associated with inhibition of Ca2+ current in glomus cells. The purpose of the present study was to elucidate the characteristics and mechanism of NE inhibition of whole cell Ca2+ current isolated from rabbit CB glomus cells and to determine the type(s) of Ca2+ channel involved. NE (10 microM) inhibited 24 +/- 2% (SE) of the macroscopic Ca2+ current measured at the end of a 25 ms pulse to 0 mV and slowed activation of the current. The alpha2 adrenergic receptor antagonist, SK&F 86466, attenuated these effects. Inhibition by NE was fast and voltage-dependent i.e., maximal at -10 mV and then diminished with stronger depolarizations. This is characteristic of G protein betagamma subunit interaction with the alpha1 subunit of certain Ca2+ channels, which can be relieved by depolarizing steps. A depolarizing step (30 ms to +80 mV) significantly increased (14 +/- 1%) current in the presence of NE, whereas it had no effect before application of NE (1 +/- 1%). To further test for the involvement of G proteins, NE was applied to cells where intracellular GTP was replaced by GDP-betaS. NE had little or no effect on Ca2+ current in cells dialyzed with GDP-betaS. To determine whether NE was inhibiting N- and/or P/Q-type channels, we applied NE in the presence of omega-conotoxin MVIIC (MVIIC). In the presence of 2.5 microM MVIIC, NE was equally potent at inhibiting the Ca2+ current (23 +/- 4% vs. 23 +/- 4% in control), suggesting that NE was not exclusively inhibiting N- or P/Q-type channels. NE was also equally potent (30 +/- 2% vs. 26 +/- 4% in control) at inhibiting the Ca2+ current in the presence of 2 microM nisoldipine, suggesting that NE was not inhibiting L-type channels. Further, NE inhibited a significantly larger proportion (47 +/- 6%) of the resistant Ca2+ current remaining in the presence of NISO and MVIIC. These results suggest that NE inhibition of Ca2+ current in rabbit CB glomus cells is mediated in most part by effects on the resistant, non L-, N-, or P/Q-type channel and involves a direct G protein betagamma interaction with this channel.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Carotid Body/physiology , GTP-Binding Proteins/physiology , Norepinephrine/pharmacology , Adrenergic alpha-2 Receptor Agonists , Animals , Calcium Channels/metabolism , Carotid Body/cytology , Carotid Body/drug effects , Electric Stimulation , Electrophysiology , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Rabbits
13.
Am J Physiol ; 274(6): C1592-600, 1998 06.
Article in English | MEDLINE | ID: mdl-9611124

ABSTRACT

We examined the effects of hypoxia on the release of dopamine (DA) and norepinephrine (NE) from rat pheochromocytoma 12 (PC-12) cells and assessed the involvement of Ca2+ and protein kinases in stimulus-secretion coupling. Catecholamine release was monitored by microvoltammetry using a carbon fiber electrode as well as by HPLC coupled with electrochemical detection (ECD). Microvoltammetric analysis showed that hypoxia-induced catecholamine secretion (PO2 of medium approximately 40 mmHg) occurred within 1 min after the onset of the stimulus and reached a plateau between 10 and 15 min. HPLC-ECD analysis revealed that, at any level of PO2, the release of NE was greater than the release of DA. In contrast, in response to K+ (80 mM), DA release was approximately 11-fold greater than NE release. The magnitude of hypoxia-induced NE and DA releases depended on the passage, source, and culture conditions of the PC-12 cells. Omission of extracellular Ca2+ or addition of voltage-gated Ca2+ channel blockers attenuated hypoxia-induced release of both DA and NE to a similar extent. Protein kinase inhibitors, staurosporine (200 nM) and bisindolylmaleimide I (2 microM), on the other hand, attenuated hypoxia-induced NE release more than DA release. However, protein kinase inhibitors had no significant effect on K+-induced NE and DA releases. These results demonstrate that hypoxia releases catecholamines from PC-12 cells and that, for a given change in PO2, NE release is greater than DA release. It is suggested that protein kinases are involved in the enhanced release of NE during hypoxia.


Subject(s)
Cell Hypoxia , Dopamine/metabolism , Norepinephrine/metabolism , PC12 Cells/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Cell Survival , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Maleimides/pharmacology , Microelectrodes , Oxygen/administration & dosage , Potassium/pharmacology , Protein Kinase Inhibitors , Protein Kinases/metabolism , Rats , Staurosporine/pharmacology
14.
J Neurophysiol ; 78(5): 2467-74, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9356397

ABSTRACT

Ca2+ current in rabbit carotid body glomus cells is conducted by multiple types of high-voltage-activated Ca2+ channels. J. Neurophysiol. 78: 2467-2474, 1997. Carotid bodies are sensory organs that detect changes in arterial oxygen. Glomus cells are presumed to be the initial sites for sensory transduction, and Ca2+-dependent neurotransmitter release from glomus cells is believed to be an obligatory step in this response. Some information exists on the Ca2+ channels in rat glomus cells. However, relatively little is known about the types of Ca2+ channels present in rabbit glomus cells, the species in which most of the neurotransmitter release studies have been performed. Therefore we tested the effect of specific Ca2+ channel blockers on current recorded from freshly dissociated, adult rabbit carotid body glomus cells using the whole cell configuration of the patch-clamp technique. Macroscopic Ba2+ current elicited from a holding potential of -80 mV activated at a Vm of approximately -30 mV, peaked between 0 and +10 mV and did not inactivate during 25-ms steps to positive test potentials. Prolonged ( approximately 2 min) depolarized holding potentials inactivated the current with a V1/2 of -47 mV. There was no evidence for T-type channels. On steps to 0 mV, 6 mM Co2+ decreased peak inward current by 97 +/- 1% (mean +/- SE). Nisoldipine (2 mu M), 1 mu M omega-conotoxin GVIA, and 100 nM omega-agatoxin IVa each blocked a portion of the macroscopic Ca2+ current (30 +/- 5, 33 +/- 5, and 19 +/- 3% after rundown correction, respectively). Simultaneous application of these blockers revealed a resistant current that was not affected by 1 mu M omega-conotoxin MVIIC. This resistant current constituted 27 +/- 5% of the total macroscopic Ca2+ current. Each blocker had an effect in every cell so tested. However, the relative proportion of current blocked varied from cell to cell. These results suggest that L, N, P, and resistant channel types each conduct a significant proportion of the macroscopic Ca2+ current in rabbit glomus cells. Hypoxia-induced neurotransmitter release from glomus cells may involve one or more of these channels.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium/metabolism , Carotid Body/physiology , omega-Conotoxins , Animals , Calcium Channels/drug effects , Carotid Body/cytology , Carotid Body/drug effects , Cells, Cultured , Egtazic Acid/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nisoldipine/pharmacology , Patch-Clamp Techniques , Peptides/pharmacology , Rabbits , Rats , Spider Venoms/pharmacology , omega-Agatoxin IVA , omega-Conotoxin GVIA
15.
Brain Res ; 706(2): 297-302, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8822371

ABSTRACT

Previous investigators have reported that intracellular pH responds to hypoxia with a heterogenous pattern in individual glomus cells of the carotid body. The aim of the present study was to examine whether hypoxia had similar effects on cytosolic calcium ([Ca2+]i) in glomus cells, and if so, whether a heterogenous response pattern is also seen in other cell types. Experiments were performed on glomus cells from adult rat carotid bodies, rat pheochromocytoma (PC12) and vascular smooth muscle (A7r5) cells. Changes in [Ca2+]i in individual cells were determined by fluorescence imaging using Fura-2. Glomus cells were identified by catecholamine fluorescence. [Ca2+]i in glomus cells increased in response to hypoxia (pO2 = 35 +/- 8 mmHg; 5 min), whereas hypoxia induced decreases in [Ca2+]i were not seen. Increases in [Ca2+]i were observed in 20% of the isolated cells and strings of cells, but clustered glomus cells never responded. The magnitude of the calcium change in responding cells was proportional to the hypoxic stimulus. Under a given hypoxic challenge, there were marked variations in the response pattern between glomus cells. The response pattern characteristic of any given cell was reproducible. At comparable levels of hypoxia, PC12 cells also responded with an increase in [Ca2+]i with a heterogenous response pattern similar to that seen in glomus cells. In contrast, increases in [Ca2+]i in A7r5 cells could be seen only with sustained hypoxia (approximately 20 min), and little heterogeneity in the response patterns was evident. These results demonstrate that: (a) hypoxia increases cytosolic calcium in glomus cells; (b) response patterns were heterogeneous in individual cells; and (c) the pattern of the hypoxia-induced changes in [Ca2+]i is cell specific. These results suggest that hypoxia-induced increases in [Ca2+]i are faster in secretory than in non-secretory cells.


Subject(s)
Calcium/metabolism , Carotid Body/metabolism , Cell Hypoxia/physiology , Cytosol/metabolism , Animals , Carotid Body/cytology , Cell Line , Female , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , PC12 Cells , Rats , Rats, Sprague-Dawley , Reproducibility of Results
17.
Am J Physiol ; 268(3 Pt 1): C636-46, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7534982

ABSTRACT

Whole cell epithelial cystic fibrosis transmembrane conductance regulator (CFTR) Cl- currents exhibited a linear current-voltage (I-V) relationship with high symmetrical transmembrane Cl- concentrations. However, when intracellular Cl- (Cli-) was reduced by replacement with glutamate, I-V relationships were outwardly rectifying. Rectification was not affected by reducing extracellular Cl- to eliminate or reverse the gradient, indicating that rectification is not a function of the Cl- gradient. Rectification was affected by Cli- in a concentration-dependent manner, and it was weaker when Cli- was reduced by replacement with sucrose. These characteristics are identical to those of the cardiac isoform of CFTR, and the experimental data could be simulated by an Eyring rate theory model assuming that permeating anions interact at a single binding site within the channel pore. No evidence was found for multiple binding sites. These results indicate that rectification is a function of the concentration and permeability of the anions inside the cell. It is concluded that rectification of CFTR Cl- current is a property of ion channel permeation that would occur under physiological conditions and that permeation of the epithelial and cardiac isoforms of CFTR is identical.


Subject(s)
Chloride Channels/physiology , Membrane Proteins/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Adenosine Triphosphate/metabolism , Animals , Anions , Cell Line , Chlorides/pharmacology , Colforsin/pharmacology , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator , Electric Conductivity , Epithelium/physiology , Guinea Pigs , Heart/physiology , Membrane Proteins/genetics , Phosphorylation , Transfection
18.
Medinfo ; 8 Pt 2: 1545, 1995.
Article in English | MEDLINE | ID: mdl-8591496

ABSTRACT

Computer networks serve as convenient, efficient, and enduring vehicles for delivering nursing services to patients at home. The ComputerLink, a nurse-supervised computer network, provides information, decision support, and communication services to caregivers of people with Alzheimer's Disease [1]. The data obtained in the original experiment included daily logs of each caregiver's use of ComputerLink (n=606 days), records of each caregiver's ComputerLink access behavior (47 subjects; 3875 accesses), and transcripts of the public communications (n=749) posted on the ComputerLink's open electronic bulletin board. In this paper, we present a case study of an early user's pattern of connections to ComputerLink as an atypical example of how elders make use of computer networks.


Subject(s)
Caregivers/psychology , Community Networks/statistics & numerical data , Computer Communication Networks/statistics & numerical data , Aged , Female , Humans , Information Services/statistics & numerical data , Ohio , Social Support
19.
Medinfo ; 8 Pt 2: 1546, 1995.
Article in English | MEDLINE | ID: mdl-8591497

ABSTRACT

1. INTRODUCTION. Decision-making demands faced by homebound caregivers of persons with Alzheimer's Disease (AD) are numerous and complex. Although caregivers frequently know what to do in caring for the person with AD, they lack confidence in their decisions [1]. The challenge of caregiving decisions may be compounded by the lack of resources to aid in decision-making or affirm decisions made. Because many caregivers are homebound, they are unable to avail themselves of traditional health and social services. ComputerLink, computer network, was designed to address the needs of these homebound caregivers. 2. DESCRIPTION. The ComputerLink consisted of a set of specialized programs and utilities residing within a public computer network. The three main features of the ComputerLink, which may provide aid to caregivers in decision making, include: the electronic encyclopedia (EE); a communications utility composed of a question and answer bulletin board (Q&A) and public (Forum) and private mail services; and a decision support module (DSM). The EE contained illness-specific information to support decision-making. The Forum and private mail provided caregivers with a means to interact with peers and professionals about their caregiving decisions. Q&A provided the opportunity to ask questions about caregiving and receive feedback from professionals and peers. The DSM could be accessed to gain information about decision-making or to actually work through a decision problem. The DSM was built on a multi-attribute utility model which employed English-language questions to guide users through a decision analysis. The analysis strategy was designed to help the user focus on the values and trade-offs that occur during difficult choices. The impact of ComputerLink on decision confidence and skill was evaluated as part of a larger randomized experiment. 102 AD caregivers were recruited: 51 caregivers in the control group and 47 in the experimental group completed the 12-month study. The mean age of the caregivers was 60 years. Thirty-three percent were male and 28 percent were Black. The groups did not differ in demographic characteristics. The ComputerLink interface was menu-driven and the system was available 24 hours. ComputerLink use was captured through a passive monitoring system. AD caregivers accessed ComputerLink a total of 3,875 times, with a mean access per individual of 83 (range: 3 - 590). The mean length of an encounter was 13 minutes; subjects typically accessed two or more functions during this time. The decision support model was accessed 91 times. The communication functions were most frequently used (3,724 Forum accesses and 1,888 private mail); the information utility was accessed 518 times. 3. RESULTS. Decision confidence was objectively measured using a modification of the Saunders and Courtney confidence-in-decision making instrument. Caregivers who had access to ComputerLink experienced greater confidence in decision-making than caregivers in the control group (t = 2.73, p < .01). Access to ComputerLink did not alter decision skill as measured by the number of alternative solutions generated for two self-identified decisions problems (t = .18, p < .05). Decision-making confidence correlated positively with ComputerLink accesses (r = .32, p = < .02) and with length of time spent on ComputerLink (r = .30, p = < .02). 56% of the AD caregivers reported that use of ComputerLink increased both their confidence and skill in decision making, though only 38 percent reported using the DSM to make a decision. 4. DISCUSSION. Access to ComputerLink increased decision-making confidence, but not decision-making skill in AD caregivers. The impact of ComputerLink on decision confidence, and not skill, may be related to the differential use of the utilities. (abstract truncated)


Subject(s)
Alzheimer Disease/nursing , Caregivers/psychology , Computer Communication Networks/statistics & numerical data , Decision Making, Computer-Assisted , Community Networks , Female , Home Nursing/psychology , Humans , Male , Middle Aged , Ohio
20.
J Gen Physiol ; 102(5): 871-95, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8301261

ABSTRACT

The whole cell configuration of the patch clamp technique was used to investigate the mechanism underlying rectification of the isoproterenol-activated chloride (Cl-) current in isolated guinea pig ventricular myocytes. When extracellular Cl- was replaced with either bromide (Br-), glutamate (Glut), iodide (I-), isethionate (Iseth), or nitrate (NO3-), the magnitude of the shift in reversal potential of the macroscopic current suggested the following selectivity sequence: NO3- > Br- > or = Cl- > or = I- > Iseth > or = Glut. This information was used to investigate the role of permeant ions in rectification of this current. Consistent with previous observations, when the concentration of intracellular Cl- (Cli-) was less than the concentration of extracellular Cl- (Clo-) (40 mM Cli-/150 mM Clo-) the current exhibited outward rectification, but when Cli- was increased to equal that outside (150 Cli-/150 Clo-), the current no longer rectified. Rectification in the presence of asymmetrical concentrations of permeant ions on either side of the membrane is predicted by constant field theory, as described by the Goldman-Hodgkin-Katz current equation. However, when the Cl- gradient was reversed (150 Cli-/40 Clo-) the current did not rectify in the opposite direction, and in the presence of lower symmetrical concentrations of Cl- inside and out (40 Cli-/40 Clo-), outward rectification did not disappear. Reducing Cli- by equimolar replacement with glutamate caused a concentration dependent increase in the degree of rectification. However, when Cli- was replaced with more permeant anions (NO3- and Br-), rectification was not observed. These results can be explained by a single binding site model based on Eyring rate theory, indicating that rectification is a function of the concentration and the permeability of the anions in the intracellular solution.


Subject(s)
Chloride Channels/physiology , Heart/physiology , Isoproterenol/pharmacology , Myocardium/cytology , Animals , Biological Transport/physiology , Bromides/pharmacokinetics , Cell Membrane Permeability/physiology , Chloride Channels/analysis , Chlorides/pharmacokinetics , Female , Glutamates/pharmacokinetics , Guinea Pigs , Iodides/pharmacokinetics , Isethionic Acid/pharmacokinetics , Male , Myocardium/chemistry , Nitrates/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...