Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
2.
Cardiovasc Drugs Ther ; 28(6): 501-11, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25348824

ABSTRACT

INTRODUCTION: K(+) channels are central to vascular pathophysiology. Previous results demonstrated that phenotypic modulation associates with a change in Kv1.3 to Kv1.5 expression, and that Kv1.3 blockade inhibits proliferation of VSMCs cultures. PURPOSE: To explore whether the Kv1.3 to Kv1.5 switch could be a marker of the increased risk of intimal hyperplasia in essential hypertension and whether systemic treatment with Kv1.3 blockers can prevent intimal hyperplasia after endoluminal lesion . METHODS: Morphometric and immunohistochemical analysis were performed in arterial segments following arterial injury and constant infusion of the Kv1.3 blocker PAP-1 during 28 days. Differential expression of K(+) channel genes was studied in VSMC from hypertensive (BPH) and normotensive (BPN) mice, both in control and after endoluminal lesion. Finally, the migration and proliferation rate of BPN and BPH VSMCs was explored in vitro. RESULTS: Changes in mRNA expression led to an increased Kv1.3/Kv1.5 ratio in BPH VSMC. Consistent with this, arterial injury in BPH mice induced a higher degree of luminal stenosis, (84 ± 4% vs. 70 ± 5% in BPN, p < 0.01), although no differences in migration and proliferation rate were observed in cultured VSMCs. The in vivo proliferative lesions were significantly decreased upon PAP-1 systemic infusion (18 ± 6% vs. 58 ± 20% with vehicle, p < 0.05). CONCLUSIONS: Hypertension leads to a higher degree of luminal stenosis in our arterial injury model, that correlates with a decreased expression of Kv1.5 channels. Kv1.3 blockers decreased in vitro VSMCs proliferation, migration, and in vivo intimal hyperplasia formation, pointing to Kv1.3 channels as promising therapeutical targets against restenosis.


Subject(s)
Arteries/drug effects , Hyperplasia/metabolism , Hypertension/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/metabolism , Potassium Channel Blockers/pharmacology , Tunica Intima/drug effects , Animals , Arteries/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Essential Hypertension , Female , Hyperplasia/drug therapy , Hypertension/drug therapy , Male , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Pancreatitis-Associated Proteins , Tunica Intima/metabolism
3.
Adv Exp Med Biol ; 648: 73-82, 2009.
Article in English | MEDLINE | ID: mdl-19536467

ABSTRACT

Chemoreceptor cells from rabbit carotid body (CB) exhibit transient outward currents reversibly inhibited by low P(o2). Molecular and functional dissection of the components of these outward currents indicates that at least two different channels (Kv4.3 and Kv3.4) contribute to this current. Furthermore, several lines of evidence support the conclusion that Kv4 channel subfamily members (either Kv4.3 alone or Kv4.3/Kv4.1 heteromultimers) are the oxygen sensitive K channels (K(o2)) in rabbit CB chemoreceptor cells. However, the pharmacological characterization of these currents shows that they are almost completely blocked by high external TEA concentrations, while Kv4 channels have been shown to be TEA-insensitive. We hypothesized that the expression of regulatory subunits in chemoreceptor cells could modify TEA sensitivity of Kv4 channels. Here, we explore the presence and functional contribution of DPPX to K(o2) currents in rabbit CB chemoreceptor cells by using DPPX functional knockdown with siRNA. Our data suggest that DPPX proteins are integral components of K(o2) currents, and that their association with Kv4 subunits modulate the pharmacological profile of the heteromultimers.


Subject(s)
Carotid Body/drug effects , Carotid Body/metabolism , Shal Potassium Channels/metabolism , Tetraethylammonium/pharmacology , Xanthines/pharmacology , Animals , Base Sequence , Dose-Response Relationship, Drug , Electric Conductivity , Extracellular Space/drug effects , Extracellular Space/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rabbits , Time Factors
4.
J Physiol ; 566(Pt 2): 491-503, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15890701

ABSTRACT

The carotid body (CB) is an arterial chemoreceptor, bearing specialized type I cells that respond to hypoxia by closing specific K+ channels and releasing neurotransmitters to activate sensory axons. Despite having detailed information on the electrical and neurochemical changes triggered by hypoxia in CB, the knowledge of the molecular components involved in the signalling cascade of the hypoxic response is fragmentary. This study analyses the mouse CB transcriptional changes in response to low PO2 by hybridization to oligonucleotide microarrays. The transcripts were obtained from whole CBs after mice were exposed to either normoxia (21% O2), or physiological hypoxia (10% O2) for 24 h. The CB transcriptional profiles obtained under these environmental conditions were subtracted from the profile of control non-chemoreceptor adrenal medulla extracted from the same animals. Given the common developmental origin of these two organs, they share many properties but differ specifically in their response to O2. Our analysis revealed 751 probe sets regulated specifically in CB under hypoxia (388 up-regulated and 363 down-regulated). These results were corroborated by assessing the transcriptional changes of selected genes under physiological hypoxia with quantitative RT-PCR. Our microarray experiments revealed a number of CB-expressed genes (e.g. TH, ferritin and triosephosphate isomerase) that were known to change their expression under hypoxia. However, we also found novel genes that consistently changed their expression under physiological hypoxia. Among them, a group of ion channels show specific regulation in CB: the potassium channels Kir6.1 and Kcnn4 are up-regulated, while the modulatory subunit Kcnab1 is down-regulated by low PO2 levels.


Subject(s)
Adrenal Medulla/metabolism , Carotid Body/metabolism , Gene Expression/physiology , Hypoxia/metabolism , Adrenal Medulla/cytology , Animals , Carotid Body/cytology , Cells, Cultured , Computational Biology , DNA Primers , Female , In Situ Hybridization , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Potassium Channels/biosynthesis , Potassium Channels/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
5.
J Physiol ; 554(Pt 1): 126-44, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14678497

ABSTRACT

Hypoxia increases the release of neurotransmitters from chemoreceptor cells of the carotid body (CB) and the activity in the carotid sinus nerve (CSN) sensory fibers, elevating ventilatory drive. According to previous reports, perinatal hyperoxia causes CSN hypotrophy and varied diminishment of CB function and the hypoxic ventilatory response. The present study aimed to characterize the presumptive hyperoxic damage. Hyperoxic rats were born and reared for 28 days in 55%-60% O2; subsequent growth (to 3.5-4.5 months) was in a normal atmosphere. Hyperoxic and control rats (born and reared in a normal atmosphere) responded with a similar increase in ventilatory frequency to hypoxia and hypercapnia. In comparison with the controls, hyperoxic CBs showed (1) half the size, but comparable percentage area positive to tyrosine hydroxylase (chemoreceptor cells) in histological sections; (2) a twofold increase in dopamine (DA) concentration, but a 50% reduction in DA synthesis rate; (3) a 75% reduction in hypoxia-evoked DA release, but normal high [K+]0-evoked release; (4) a 75% reduction in the number of hypoxia-sensitive CSN fibers (although responding units displayed a nearly normal hypoxic response); and (5) a smaller percentage of chemoreceptor cells that increased [Ca2+]1 in hypoxia, although responses were within the normal range. We conclude that perinatal hyperoxia causes atrophy of the CB-CSN complex, resulting in a smaller number of chemoreceptor cells and fibers. Additionally, hyperoxia damages O2-sensing, but not exocytotic, machinery in most surviving chemoreceptor cells. Although hyperoxic CBs contain substantially smaller numbers of chemoreceptor cells/sensory fibers responsive to hypoxia they appear sufficient to evoke normal increases in ventilatory frequency.


Subject(s)
Carotid Body/cytology , Carotid Body/physiology , Hyperoxia/physiopathology , Respiratory Mechanics/physiology , Age Factors , Animals , Calcium/metabolism , Calcium/pharmacokinetics , Cells, Cultured , Chemoreceptor Cells/cytology , Chemoreceptor Cells/physiology , Female , Hypoxia/physiopathology , Membrane Potentials/physiology , Motor Activity , Oxygen/pharmacology , Potassium/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Tritium
8.
Circ Res ; 89(5): 430-6, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11532904

ABSTRACT

Hypoxic inhibition of large-conductance Ca(2+)-dependent K(+) channels (maxiK) of rat carotid body type I cells is a well-established fact. However, the molecular mechanisms of such inhibition and the role of these channels in the process of hypoxic transduction remain unclear. We have examined the mechanisms of interaction of O(2) with maxiK channels exploring the effect of hypoxia on maxiK currents recorded with the whole-cell and the inside-out configuration of the patch-clamp technique. Hypoxia inhibits channel activity both in whole-cell and in excised membrane patches. This effect is strongly voltage- and Ca(2+)-dependent, being maximal at low [Ca(2+)] and low membrane potential. The analysis of single-channel kinetics reveals a gating scheme comprising three open and five closed states. Hypoxia inhibits channel activity increasing the time the channel spends in the longest closed states, an effect that could be explained by a decrease in the Ca(2+) sensitivity of those closed states. Reducing maxiK channels with dithiothreitol (DTT) increases channel open probability, whereas oxidizing the channels with 2,2'-dithiopyridine (DTDP) has the opposite effect. These results suggest that hypoxic inhibition is not related with a reduction of channel thiol groups. However, CO, a competitive inhibitor of O(2) binding to hemoproteins, fully reverts hypoxic inhibition, both at the whole-cell and the single-channel level. We conclude that O(2) interaction with maxiK channels does not require cytoplasmic mediators. Such interaction could be mediated by a membrane hemoprotein that, as an O(2) sensor, would modulate channel activity.


Subject(s)
Carbon Monoxide/pharmacology , Chemoreceptor Cells/drug effects , Membrane Potentials/drug effects , Oxygen/pharmacology , Potassium Channels, Calcium-Activated , Potassium Channels/physiology , Animals , Calcium/metabolism , Cells, Cultured , Chemoreceptor Cells/cytology , Chemoreceptor Cells/physiology , Dose-Response Relationship, Drug , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Large-Conductance Calcium-Activated Potassium Channels , Oxidation-Reduction , Patch-Clamp Techniques , Rats , Rats, Wistar
9.
J Neurosci ; 20(15): 5689-95, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10908607

ABSTRACT

Hypoxia initiates the neurosecretory response of the carotid body (CB) by inhibiting one or more potassium channels in the chemoreceptor cells. Oxygen-sensitive K(+) channels were first described in rabbit CB chemoreceptor cells, in which a transient outward K(+) current was reported to be reversibly inhibited by hypoxia. Although progress has been made to characterize this current with electrophysiological and pharmacological tools, no attempts have been made to identify which Kv channel proteins are expressed in rabbit CB chemoreceptor cells and to determine their contribution to the native O(2)-sensitive K(+) current. To probe the molecular identity of this current, we have used dominant-negative constructs to block the expression of functional Kv channels of the Shaker (Kv1.xDN) or the Shal (Kv4.xDN) subfamilies, because members of these two subfamilies contribute to the transient outward K(+) currents in other preparations. Delivery of the constructs into chemoreceptor cells has been achieved with adenoviruses that enabled ecdysone-inducible expression of the dominant-negative constructs and reporter genes in polycistronic vectors. In voltage-clamp experiments, we found that, whereas adenoviral infections of chemoreceptor cells with Kv1.xDN did not modify the O(2)-sensitive K(+) current, infections with Kv4.xDN suppressed the transient outward current in a time-dependent manner, significantly depolarized the cells, and abolished the depolarization induced by hypoxia. Our work demonstrate that genes of the Shal K(+) channels underlie the transient outward, O(2)-sensitive, K(+) current of rabbit CB chemoreceptor cells and that this current contributes to the cell depolarization in response to low pO(2).


Subject(s)
Adenoviridae/genetics , Chemoreceptor Cells/physiology , Gene Transfer Techniques , Oxygen/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium Channels/metabolism , Animals , CHO Cells , Carotid Body/chemistry , Carotid Body/physiology , Chemoreceptor Cells/chemistry , Cricetinae , Electrophysiology , Gene Expression/physiology , Genes, Dominant , Humans , Hypoxia/metabolism , Hypoxia/physiopathology , Kidney/cytology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutagenesis/physiology , Potassium/metabolism , Rabbits , Shaker Superfamily of Potassium Channels , Shal Potassium Channels , Tetrodotoxin/pharmacology , Transfection
10.
Am J Physiol Cell Physiol ; 279(1): C51-61, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898716

ABSTRACT

The notion that intracellular Ca(2+) (Ca(i)(2+)) stores play a significant role in the chemoreception process in chemoreceptor cells of the carotid body (CB) appears in the literature in a recurrent manner. However, the structural identity of the Ca(2+) stores and their real significance in the function of chemoreceptor cells are unknown. To assess the functional significance of Ca(i)(2+) stores in chemoreceptor cells, we have monitored 1) the release of catecholamines (CA) from the cells using an in vitro preparation of intact rabbit CB and 2) the intracellular Ca(2+) concentration ([Ca(2+)](i)) using isolated chemoreceptor cells; both parameters were measured in the absence or the presence of agents interfering with the storage of Ca(2+). We found that threshold [Ca(2+)](i) for high extracellular K(+) (K(e)(+)) to elicit a release response is approximately 250 nM. Caffeine (10-40 mM), ryanodine (0.5 microM), thapsigargin (0.05-1 microM), and cyclopiazonic acid (10 microM) did not alter the basal or the stimulus (hypoxia, high K(e)(+))-induced release of CA. The same agents produced Ca(i)(2+) transients of amplitude below secretory threshold; ryanodine (0.5 microM), thapsigargin (1 microM), and cyclopiazonic acid (10 microM) did not alter the magnitude or time course of the Ca(i)(2+) responses elicited by high K(e)(+). Several potential activators of the phospholipase C system (bethanechol, ATP, and bradykinin), and thereby of inositol 1,4,5-trisphosphate receptors, produced minimal or no changes in [Ca(2+)](i) and did not affect the basal release of CA. It is concluded that, in the rabbit CB chemoreceptor cells, Ca(i)(2+) stores do not play a significant role in the instant-to-instant chemoreception process.


Subject(s)
Calcium/metabolism , Carotid Body/metabolism , Chemoreceptor Cells/metabolism , Intracellular Membranes/metabolism , Animals , Caffeine/pharmacology , Carotid Body/cytology , Carotid Body/drug effects , Catecholamines/metabolism , Cell Hypoxia/physiology , Cell Separation , Chemoreceptor Cells/physiology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Extracellular Space/metabolism , Potassium/administration & dosage , Potassium/metabolism , Potassium/pharmacology , Rabbits , Ryanodine/pharmacology , Thapsigargin/pharmacology , Type C Phospholipases/metabolism
12.
J Gen Physiol ; 113(6): 897-907, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10352037

ABSTRACT

Voltage-gated K+ (KV) channels are protein complexes composed of ion-conducting integral membrane alpha subunits and cytoplasmic modulatory beta subunits. The differential expression and association of alpha and beta subunits seems to contribute significantly to the complexity and heterogeneity of KV channels in excitable cells, and their functional expression in heterologous systems provides a tool to study their regulation at a molecular level. Here, we have studied the effects of Kvbeta1.2 coexpression on the properties of Shaker and Kv4.2 KV channel alpha subunits, which encode rapidly inactivating A-type K+ currents, in transfected HEK293 cells. We found that Kvbeta1.2 functionally associates with these two alpha subunits, as well as with the endogenous KV channels of HEK293 cells, to modulate different properties of the heteromultimers. Kvbeta1.2 accelerates the rate of inactivation of the Shaker currents, as previously described, increases significantly the amplitude of the endogenous currents, and confers sensitivity to redox modulation and hypoxia to Kv4.2 channels. Upon association with Kvbeta1.2, Kv4.2 can be modified by DTT (1,4 dithiothreitol) and DTDP (2,2'-dithiodipyridine), which also modulate the low pO2 response of the Kv4.2+beta channels. However, the physiological reducing agent GSH (reduced glutathione) did not mimic the effects of DTT. Finally, hypoxic inhibition of Kv4.2+beta currents can be reverted by 70% in the presence of carbon monoxide and remains in cell-free patches, suggesting the presence of a hemoproteic O2 sensor in HEK293 cells and a membrane-delimited mechanism at the origin of hypoxic responses. We conclude that beta subunits can modulate different properties upon association with different KV channel subfamilies; of potential relevance to understanding the molecular basis of low pO2 sensitivity in native tissues is the here described acquisition of the ability of Kv4. 2+beta channels to respond to hypoxia.


Subject(s)
Hypoxia/physiopathology , Ion Channel Gating/drug effects , Oxygen/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , 2,2'-Dipyridyl/analogs & derivatives , 2,2'-Dipyridyl/pharmacology , Antioxidants/pharmacology , Cell Membrane/chemistry , Cell Membrane/metabolism , Cells, Cultured , Cloning, Molecular , Disulfides/pharmacology , Dithiothreitol/pharmacology , Glutathione/pharmacology , Humans , Hypoxia/metabolism , Ion Channel Gating/physiology , Kidney/cytology , Kinetics , Kv1.2 Potassium Channel , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Potassium Channels/chemistry , Potassium Channels/genetics , Shaker Superfamily of Potassium Channels , Shal Potassium Channels , Sulfhydryl Reagents/pharmacology , Transfection
13.
Mol Pharmacol ; 53(2): 330-9, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9463492

ABSTRACT

Almitrine is a drug used in the treatment of hypoxemic chronic lung diseases such as bronchitis and emphysema because it is a potent stimulant of the carotid bodies in human and different animal species that produces a long-lasting enhancement of alveolar ventilation, ameliorating arterial blood gases. However, the mechanism of action of almitrine remains unknown. We investigated the effect of almitrine on ionic currents of chemoreceptor cells isolated from the carotid body of rat and rabbits by using the whole-cell and inside-out configurations of the patch-clamp technique. Almitrine at concentrations up to 10 microM did not affect whole-cell voltage-dependent K+, Ca2+, or Na+ currents in rat or rabbit cells. However, this concentration of almitrine significantly inhibited the Ca2+-dependent component of K+ currents in rat chemoreceptor cells. This effect of almitrine on the Ca2+-dependent component of K+ currents was investigated further at the single-channel level in excised patches in the inside-out configuration. In this preparation, almitrine inhibited the activity of a high-conductance (152 +/- 13 pS), Ca2+-dependent K+ channel by decreasing its open probability. The IC50 value of the effect was 0. 22 microM. The inhibitory effect of almitrine on Ca2+-dependent K+ channels also was observed in GH3 cells. We conclude that almitrine inhibits selectively the Ca2+-dependent K+ channel and that in rat chemoreceptor cells, this inhibition could represent an important mechanism of action underlying the therapeutic actions of the drug.


Subject(s)
Almitrine/pharmacology , Carotid Body/drug effects , Chemoreceptor Cells/drug effects , Animals , Calcium/physiology , Calcium Channels/drug effects , Cells, Cultured , Electric Conductivity , Ion Channel Gating/drug effects , Membrane Potentials , Oxygen/physiology , Patch-Clamp Techniques , Potassium Channels/drug effects , Rabbits , Rats
14.
Pflugers Arch ; 434(6): 698-704, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9306001

ABSTRACT

Hypoxia activates erythropoietin-producing cells, chemoreceptor cells of the carotid body and pulmonary artery smooth muscle cells (PSMC) with a comparable arterial PO2 threshold of some 70 mmHg. The inhibition by CO of the hypoxic responses in the two former cell types has led to the proposal that a haemoprotein is involved in the detection of the PO2 levels. Here, we report the effect of CO on the hypoxic pulmonary vasoconstriction (HPV). Pulmonary arterial pressure (PAP) was measured in an in situ, blood-perfused lung preparation. PAP in normoxia (20% O2, 5% CO2) was 15.2+/-1.8 mmHg, and hypoxia (2% O2, 5% CO2) produced a DeltaPAP of 6.3+/-0.4 mmHg. Addition of 8% or 15% CO to the hypoxic gas mixture reduced the DeltaPAP by 88.3+/-2.7% and 78.2+/-6.1% respectively. The same levels of CO did not affect normoxic PAP nor reduced the DeltaPAP produced by angiotensin II. The effect of CO was studied after inhibition of the NO-cyclic guanosine monophosphate (cGMP) cascade with N-methyl-l-arginine (5.10(-5) M) or methylene blue (1.4.10(-4) M). It was found that both inhibitors more than doubled the hypoxic DeltaPAP without altering the effectiveness of CO to inhibit the HPV. In in vitro experiments we verified the inhibition of guanylate cyclase by measuring the levels of cGMP in segments of the pulmonary artery. Cyclic GMP levels were 1.4+/-0.2 (normoxia), 2.5+/-0.3 (hypoxia) and 3.3+/-0.5 pmole/mg tissue (hypoxia plus 8% CO); sodium nitroprusside increased normoxic cGMP levels about fourfold. Methylene blue reduced cGMP levels to less than 10% in all cases, and abolished the differences among normoxic, hypoxic and hypoxic plus CO groups. It is concluded that CO inhibits HPV by a NO-cGMP independent mechanism and it is proposed that a haemoprotein could be involved in O2-sensing in PSMC.


Subject(s)
Carbon Monoxide/pharmacology , Cyclic GMP/physiology , Hypoxia/physiopathology , Pulmonary Circulation/drug effects , Vasoconstriction/drug effects , Vasoconstriction/physiology , Animals , Cyclic GMP/metabolism , Enzyme Inhibitors/pharmacology , Female , Guanylate Cyclase/antagonists & inhibitors , In Vitro Techniques , Methylene Blue/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , Pulmonary Artery/metabolism , Rats , Rats, Wistar , Solubility
15.
J Physiol ; 499 ( Pt 2): 429-41, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9080372

ABSTRACT

1. The electrical properties of chemoreceptor cells from neonatal rat and adult rabbit carotid bodies (CBs) are strikingly different. These differences have been suggested to be developmental and/or species related. To distinguish between the two possibilities, the whole-cell configuration of the patch-clamp technique was used to characterize the ionic currents present in isolated chemoreceptor cells from adult rat CBs. Since hypoxia-induced inhibition of O2-sensitive K+ currents is considered a crucial step in O2 chemoreception, the effect of hypoxia on the adult rat chemoreceptor cell currents was also studied. 2. Outward currents were carried mainly by K+, and two different components could be distinguished: a Ca(2+)-dependent K+ current (IK(Ca)) sensitive to Cd2+ and charybdotoxin (CTX), and a Ca(2+)-insensitive, voltage-dependent K+ current (IK(V)). IK(V) showed a slow voltage-dependent activation (time constant (tau) of 87.4 ms at -20 mV and 8.8 ms at +60 mV) and a very slow inactivation, described by the sum of two exponentials (tau 1 = 684 +/- 150 ms and tau 2 = 4.96 +/- 0.76 s at + 30 mV), that was almost voltage insensitive. The kinetic and pharmacological properties of IK(V) are typical of a delayed rectifier K+ channel. 3. Voltage-dependent Ca2+ currents (ICa) were present in nineteen of twenty-seven cells. TTX-sensitive Na+ currents were also observed in about 10% of the cells. 4. Low PO2 (< 10 mmHg) reduced the whole outward current amplitude by 22.17 +/- 1.96% (n = 27) at +20 mV. This effect was absent in the presence of Cd2+. Since low PO2 did not affect ICa, we conclude that hypoxia selectively blocks IK(Ca). 5. The properties of the currents recorded in adult rat chemoreceptor cells, including the specific inhibition of IK(Ca) by hypoxia, are similar to those reported in neonatal rat CB cells, implying that the differences between rat and rabbit chemoreceptor cells are species related.


Subject(s)
Carotid Body/metabolism , Chemoreceptor Cells/metabolism , Oxygen/metabolism , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Animals , Cadmium/pharmacology , Charybdotoxin/pharmacology , Large-Conductance Calcium-Activated Potassium Channels , Rabbits , Rats , Rats, Wistar , Tetraethylammonium , Tetraethylammonium Compounds/pharmacology
17.
Respir Physiol ; 102(2-3): 137-47, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8904006

ABSTRACT

The carotid bodies (CB) are arterial chemoreceptors that by sensing changes of arterial PO2, PCO2 and pH can initiate and modify ventilatory and cardiovascular reflexes in order to maintain PO2, PCO2 and pH within physiological levels. It is now generally accepted that the glomus or type I cells of the CB are the transducers of hypoxic stimuli, and relay chemosensory information to the brainstem via neurotransmitter release at synaptic contacts with afferent terminals of the carotid sinus nerve. This article reviews the mechanisms of the O2-sensing process at the cellular level. We consider first the transduction of the hypoxic stimulus, in which most of the experimental evidence currently favors a mechanism involving modulation of the electrical properties of type I cells. The last part of the article deals with the transmission of the stimulus between type I cells and afferent nerve terminals, and we present an overview on the issue of neurotransmission in the CB, summarizing the actions of the main neurotransmitters present in the organ.


Subject(s)
Carotid Body/metabolism , Chemoreceptor Cells/metabolism , Oxygen/metabolism , Acetylcholine/pharmacology , Calcium/metabolism , Carotid Body/anatomy & histology , Dopamine/metabolism , Electrophysiology , Hypoxia/metabolism , NADPH Oxidases/metabolism , Neurotransmitter Agents/chemistry , Neurotransmitter Agents/metabolism , Substance P/pharmacology
18.
Science ; 268(5213): 1042-5, 1995 May 19.
Article in English | MEDLINE | ID: mdl-7754383

ABSTRACT

Excitation-contraction coupling was studied in mammalian cardiac cells in which the opening probability of L-type calcium (Ca2+) channels was reduced. Confocal microscopy during voltage-clamp depolarization revealed distinct local transients in the concentration of intracellular calcium ions ([Ca2+]i). When voltage was varied, the latency to occurrence and the relative probability of occurrence of local [Ca2+]i transients varied as predicted if Ca2+ release from the sarcoplasmic reticulum (SR) was linked tightly to Ca2+ flux through L-type Ca2+ channels but not to that through the Na-Ca exchanger or to average [Ca2+]i. Voltage had no effect on the amplitude of local [Ca2+]i transients. Thus, the most efficacious "Ca2+ signal" for activating Ca2+ release from the SR may be a transient microdomain of high [Ca2+]i beneath an individual, open L-type Ca2+ channel.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Myocardium/metabolism , Animals , Calcium Channels/drug effects , Electric Stimulation , Guinea Pigs , In Vitro Techniques , Ion Channel Gating/physiology , Membrane Potentials/physiology , Microscopy, Confocal , Patch-Clamp Techniques , Probability , Sarcoplasmic Reticulum/metabolism , Verapamil/pharmacology
19.
Ciba Found Symp ; 188: 146-60; discussion 160-4, 1995.
Article in English | MEDLINE | ID: mdl-7587615

ABSTRACT

In heart cells, several distinct kinds of transient spatial patterns of cytoplasmic calcium ion concentration ([Ca2+]i) can be observed: (1) [Ca2+]i waves, in which regions of spontaneously increased [Ca2+]i propagate at high velocity (100 microns/s) through the cell; (2) Ca2+ 'sparks', which are spontaneous, non-propagating changes in [Ca2+]i that are localized in small (approximately 2 microns) subcellular regions; and (3) evoked [Ca2+]i transients that are elicited by electrical depolarization, in association with normal excitation-contraction (E-C) coupling. In confocal [Ca2+]i images, evoked [Ca2+]i transients appear to be nearly spatially uniform throughout the cell, except during their rising phase or during small depolarizations. In contrast to [Ca2+]i waves and spontaneous Ca2+ sparks, evoked [Ca2+]i transients are triggered by L-type Ca2+ channel current and they are 'controlled', in the sense that stopping the L-type Ca2+ current stops them. Despite their different characteristics, all three types of Ca2+ transient involve Ca(2+)-induced release of Ca2+ from the sarcoplasmic reticulum. Here, we address the question of how the autocatalytic process of Ca(2+)-induced Ca2+ release, which can easily be understood to underlie spontaneous regenerative ('uncontrolled'), propagating [Ca2+]i waves, might be 'harnessed', under other circumstances, to produce controlled changes in [Ca2+]i, as during normal excitation-contraction coupling, or changes in [Ca2+]i that do not propagate. We discuss our observations of Ca2+ waves, Ca2+ sparks and normal Ca2+ transients in heart cells and review our results on the 'gain' of Ca(2+)-induced Ca2+ release. We discuss a model involving Ca2+ microdomains beneath L-type Ca2+ channels, and clusters of Ca(2+)-activated Ca2+ release channels in the sarcoplasmic reticulum which may form the basis of the answer to this question.


Subject(s)
Calcium/metabolism , Myocardium/metabolism , Signal Transduction/physiology , Animals , Myocardium/cytology
20.
J Physiol ; 480 ( Pt 1): 21-9, 1994 Oct 01.
Article in English | MEDLINE | ID: mdl-7853223

ABSTRACT

1. Confocal microscopy and the fluorescent Ca2+ indicator fluo-3 (K+ salt) were used to measure cytosolic free calcium ion concentration ([Ca2+]) during excitation-contraction (E-C) coupling in single, voltage-clamped, rat cardiac ventricular cells. 2. Local [Ca2+]i transients were measured nearly simultaneously in different, separate, subcellular volumes of approximately 2.0 microns 3. During depolarization, local [Ca2+]i transients were distinctly different from each other and from whole-cell [Ca2+]i transients. These differences were particularly apparent during small depolarizations, and were substantially reduced by ryanodine. 3. Components of the local [Ca2+]i transients, particularly those evoked by small depolarizations, were closely similar, in time course and amplitude, to spontaneous local [Ca2+]i transients, or 'sparks' (which have been shown previously to be Ca2+ released from sarcoplasmic reticulum). 4. Analysis of local [Ca2+]i transients in the spatial frequency domain (power spectrum) revealed that high power at spatial frequencies of 0.05-0.2 microns-1 was always associated with spontaneous calcium 'sparks' and with local [Ca2+]i transients evoked by small depolarizing pulses (e.g. to -31 mV). Evoked local [Ca2+]o transients in the presence of ryanodine, and those evoked by depolarization to very positive clamp-pulse potentials (+45 mV), were associated with considerably lower power at this frequency. 5. The results suggest that whole-cell [Ca2+]i transients evoked by voltage-clamp depolarization, and thus by L-type Ca2+ current, are comprised of local [Ca2+]i transients that are similar to the spontaneous calcium 'sparks'. At very positive clamp-pulse potentials, however, the electrically evoked local [Ca2+]i transients may be smaller, perhaps as a result of smaller unitary L-type Ca2+ current.


Subject(s)
Calcium/metabolism , Myocardium/metabolism , Aniline Compounds , Animals , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium Channels/physiology , Fluorescent Dyes , Heart/drug effects , In Vitro Techniques , Membrane Potentials/physiology , Microscopy, Confocal , Myocardium/cytology , Patch-Clamp Techniques , Rats , Rats, Wistar , Ryanodine/pharmacology , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Xanthenes
SELECTION OF CITATIONS
SEARCH DETAIL
...