Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Pharmacol Res ; 70(1): 27-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23287425

ABSTRACT

Hydrogen sulphide (H2S) has been recently hypothesized to be an endogenous adipocyte-derived relaxing factor, evoking vasorelaxation of conductance and resistance vessels. Although the activation of ATP-sensitive potassium channels is known to play a central role in H2S-induced vasorelaxation, activation of vascular Kv7 voltage-gated potassium channels has also been suggested. To investigate this possibility, the ability of selective activators and blockers of distinct classes of potassium channels to affect vasodilation induced by the H2S-donor NaHS, as well as NaHS-induced Rb(+) efflux in endothelium-denuded rat aortic rings, was investigated. NaHS-induced changes of membrane potential were fluorimetrically assessed on human vascular smooth muscle (VSM) cells. Modulation of Kv7.4 channels by NaHS was assessed by electrophysiological studies, upon their heterologous expression in CHO cells. In isolated aortic rings, NaHS evoked vasorelaxing responses associated with an increase of Rb(+)-efflux. NaHS promoted membrane hyperpolarization of human VSM cells. These effects were antagonized by selective blockers of Kv7 channels. The H2S-donor caused a left-shift of current activation threshold of Kv7.4 channels expressed in CHO cells. Altogether, these results suggest that the activation of Kv7.4 channels is a key mechanism in the vascular effects of H2S. Given the relevant roles played by Kv7.4 channels in VSM contractility and by H2S in circulatory homeostasis regulation, these findings provide interesting insights to improve our understanding of H2S pathophysiology and to focus on Kv7.4 channels as novel targets for therapeutic approaches via the "H2S-system".


Subject(s)
Aorta/drug effects , Hydrogen Sulfide/pharmacology , KCNQ Potassium Channels/metabolism , Muscle, Smooth, Vascular/drug effects , Sulfides/pharmacology , Vasodilation/drug effects , Animals , Aorta/metabolism , Cell Line , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Endothelium, Vascular/physiology , Humans , In Vitro Techniques , KCNQ Potassium Channels/biosynthesis , Male , Membrane Potentials/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Wistar , Vasoconstrictor Agents/pharmacology
2.
Biochim Biophys Acta ; 1830(9): 4365-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23220699

ABSTRACT

BACKGROUND: There is no doubt that future discoveries in the field of biochemistry will depend on the implementation of novel biosensing techniques, able to record biophysiological events with minimal biological interference. In this respect, organic electronics may represent an important new tool for the analysis of structures ranging from single molecules up to cellular events. Specifically, organic field-effect transistors (OFET) are potentially powerful devices for the real-time detection/transduction of bio-signals. Despite this interest, up to date, the experimental data useful to support the development of OFET-based biosensors are still few and, in particular, n-type (electron-transporting) devices, being fundamental to develop highly-performing circuits, have been scarcely investigated. METHODS: Here, films of N,N'-1H,1H-perfluorobutyldicyanoperylene-carboxydi-imide (PDIF-CN2) molecules, a recently-introduced and very promising n-type semiconductor, have been evaporated on glass and silicon dioxide substrates to test the biocompatibility of this compound and its capability to stay electrically-active even in liquid environments. RESULTS: We found that PDIF-CN2 transistors can work steadily in water for several hours. Biocompatibility tests, based on in-vitro cell cultivation, remark the need to functionalize the PDIF-CN2 hydrophobic surface by extra-coating layers (i.e. poly-l-lysine) to favor the growth of confluent cellular populations. CONCLUSIONS: Our experimental data demonstrate that PDIF-CN2 compound is an interesting organic semiconductor to develop electronic devices to be used in the biological field. GENERAL SIGNIFICANCE: This work contributes to define a possible strategy for the fabrication of low-cost and flexible biosensors, based on complex organic complementary metal-oxide-semiconductor (CMOS) circuitry including both p- (hole-transporting) and n-type transistors. This article is part of a Special Issue entitled Organic Bioelectronics-Novel Applications in Biomedicine.


Subject(s)
Biosensing Techniques/instrumentation , Biosensing Techniques/statistics & numerical data , Electronics, Medical/instrumentation , Electronics, Medical/methods , Imides/chemistry , Perylene/analogs & derivatives , Semiconductors , Transistors, Electronic , Animals , CHO Cells , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Materials Testing/methods , Metals/chemistry , Nitriles/chemistry , Oxides/chemistry , Perylene/chemistry , Water/chemistry
3.
Eur J Pain ; 16(8): 1148-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22392917

ABSTRACT

BACKGROUND: Gamma-aminobutyric acid (GABA) and glutamate (GLU) are involved in nociceptive signals processing in the trigeminal system. In this study, we investigated the influence of excitatory transmission on GABA release in nerve terminals isolated from the rat trigeminal caudal nucleus (TCN). METHODS: We utilize biochemical (superfused synaptosomes loaded with [(3) H]GABA) and morphological (immunofluorescence experiments with specific antibody) techniques. RESULTS: Our results show that GLU potentiates the release of [(3) H]GABA evoked by 9, 15 and 30 mM [K(+)](e); 15 mM [K(+)](e)-evoked [(3) H]GABA release was also reinforced by domoate and kainate (KA), two naturally occurring GLU-receptor agonists. The enhancement of 15 mM [K(+)](e)-evoked [(3) H]GABA release produced by 100 µM KA was abolished by NBQX, a mixed AMPA/KA receptor antagonist, but was not affected by GYKI52466, a selective AMPA receptor antagonist. ATPA, a selective agonist for KA receptors containing the GLUK1 subunit, had no effect on depolarization-induced [(3) H]GABA release, and UBP310, which selectively antagonizes these same receptors, failed to reverse the KA-induced potentiation of 15 mM [K(+)](e)-evoked [(3) H]GABA release. The KA-induced potentiation was also unaffected by concanavalin A (10 µM), a positive allosteric modulator of GLUK1- and GLUK2-containing KA receptors. Immunofluorescence experiments revealed that GABAergic nerve terminals in the TCN differentially expressed GLUK subunits, with GLUK2/3-positive terminals being twice more abundant than GLUK1-containing synaptosomes. CONCLUSIONS: These findings indicate that pre-synaptic KA receptors facilitating GABA release from TCN nerve terminals mainly express GLUK2/GLUK3 subunits, supporting the notion that different types of KA receptors are involved in the various stages of pain transmission.


Subject(s)
Receptors, Kainic Acid/metabolism , Trigeminal Caudal Nucleus/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Male , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Synaptosomes/drug effects , Synaptosomes/metabolism , Trigeminal Caudal Nucleus/drug effects , GluK2 Kainate Receptor , GluK3 Kainate Receptor
4.
Eur Biophys J ; 41(2): 249-56, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22237602

ABSTRACT

Impedance spectroscopy (IS) is a powerful technique for analysis of the complex electrical impedance of a large variety of biological systems, because it is sensitive both to surface phenomena and to changes of bulk properties. A simple and convenient method of analysis of cell properties by IS is described. An interdigitated electrodes configuration was used for the measurements; human epithelial cells were grown on the device to investigate the complex dielectric response as a function of frequency, in order to test the suitability of the device for use as a label-free biosensor. To test the ability of the device to detect channels in the cell membrane, the effect of drugs known to affect membrane integrity was also investigated. The frequency response of the admittance (i.e. the reciprocal of the impedance) can be well fitted by a model based on very simple assumptions about the cells coating the device surface and the current flow; from the calculations, membrane-specific capacitance and information about cell adhesion can be inferred. These preliminary efforts have shown that our configuration could lead to a label-free non-invasive technique for biosensing and cellular behavior monitoring which might prove useful in investigation of the basic properties of cells and the effect of drugs by estimation of some fundamental properties and modification of the electrical characteristics of the device.


Subject(s)
Biosensing Techniques/methods , Dielectric Spectroscopy/methods , Systems Integration , Biosensing Techniques/instrumentation , Cell Membrane/drug effects , Cell Membrane/metabolism , Electrodes , HeLa Cells , Humans , Nystatin/pharmacology , Octoxynol/pharmacology
5.
Pharmacol Res ; 64(4): 397-409, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21740972

ABSTRACT

Voltage-dependent type 7 K+ (KV7) channels play important physiological roles in neurons and muscle cells. The aims of the present study were to investigate the motor effects of KV7 channel modulators in the rat gastric fundus and the expression of KV7 channels in this tissue. Muscle tone and electrical field stimulation (EFS)-evoked relaxations of precontracted longitudinal muscle strips of the rat gastric fundus were investigated under nonadrenergic noncholinergic conditions by organ bath studies. Gene expression was studied by real-time PCR and tissue localization of channels was investigated by immunohistochemistry. The KV7 channel blocker XE-991 induced concentration-dependent contractions, with mean pD2 and Emax of 5.4 and 48% of the maximal U46619-induced contraction, respectively. The KV7 channel activators retigabine and flupirtine concentration-dependently relaxed U46619-precontracted strips, with pD2s of 4.7 and 4.4 and Emax of 93% and 91% of the maximal relaxation induced by papaverine, respectively. XE-991 concentration-dependently inhibited retigabine-induced relaxation with a pIC50 of 6.2. XE-991 and DMP-543, another KV7 channel blocker, increased by 13-25% or reduced by 11-21% the relaxations evoked by low- or high-frequency EFS, respectively. XE-991 also reduced the relaxation induced by vasoactive intestinal polypeptide (VIP) by 33% of controls. Transcripts encoded by all KV7 genes were detected in the fundus, with 7.4 and 7.5 showing the highest expression levels. KV7.4 and 7.5 channels were visualized by confocal immunofluorescence in both circular and longitudinal muscle layers. In conclusion, in the rat proximal stomach, KV7 channels appear to contribute to the resting muscle tone and to VIP- and high-frequency EFS-induced relaxation. KV7 channel activators could be useful relaxant agents of the gastric smooth muscle.


Subject(s)
Gastric Fundus/drug effects , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , Muscle Relaxation/drug effects , Muscle Tonus/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Anticonvulsants/pharmacology , Carbamates/pharmacology , Female , Gastric Fundus/metabolism , KCNQ Potassium Channels/metabolism , Male , Phenylenediamines/pharmacology , Rats , Rats, Wistar
6.
Neurochem Int ; 57(6): 623-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20673841

ABSTRACT

Glutamate (GLU) plays a key role in the transmission and modulation of sensory input to the trigeminal caudal nuclei (TCN). In the present study, we investigated the regulation of previously taken-up [3H]D-aspartate ([3H]D-ASP) release from nerve terminals isolated from rat caudal brainstem, in particular from the zone containing the TCN. TCN neurons can be considered integrative relay neurons linking peripheral and central pain mechanisms. Understanding the mechanisms that control the release of GLU in this area could lead to more effective treatment of migraines and other types of pain associated with the trigeminal nerve. In isolated rat caudal brainstem synaptosomes, exposure to AMPA dose-dependently potentiated [K+](e)-stimulated release of [3H]D-ASP (maximum increase: 218±13.08%; EC(50): 1.60±0.08 µM). This effect was inhibited by selective AMPA-receptor antagonists (competitive [NBQX] and non-competitive [GYKI52466]) but not by the kainate receptor subunit antagonists NS102 and ACET. AMPA-evoked responses were significantly enhanced by preventing AMPA receptor desensitization with cyclothiazide (10 µM). Basal release of [3H]D-ASP was stimulated by millimolar concentrations of ATP (maximum increase: 197.80±11.85%; EC(50): 545±3.15 µM) and by the selective P2X7-receptor agonist benzoylbenzoyl-ATP. ATP also potentiated the release of [3H]D-ASP induced by depolarization. Its effect on basal [3H]D-ASP release was inhibited by the selective P2X7-receptor antagonist A-438079 and by the non-selective antagonist PPADS, but it was only partially suppressed by the ionotropic purinergic receptor antagonist TNP-ATP. Our findings demonstrate that glutamatergic nerve terminals in rat caudal brainstem express AMPA receptors that can facilitate [3H]D-ASP during terminal depolarization and P2X7 receptors that can also enhance this release under basal conditions.


Subject(s)
Aspartic Acid/metabolism , Brain Stem/metabolism , Nerve Endings/metabolism , Receptors, AMPA/physiology , Receptors, Purinergic P2X7/physiology , Animals , Male , Rats , Rats, Wistar , Tritium
8.
Mol Pharmacol ; 72(3): 665-73, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17495071

ABSTRACT

The aim of the present study was to investigate whether K(V)3.4 channel subunits are involved in neuronal death induced by neurotoxic beta-amyloid peptides (Abeta). In particular, to test this hypothesis, three main questions were addressed: 1) whether the Abeta peptide can up-regulate both the transcription/translation and activity of K(V)3.4 channel subunit and its accessory subunit, MinK-related peptide 2 (MIRP2); 2) whether the increase in K(V)3.4 expression and activity can be mediated by the nuclear factor-kappaB (NF-kappaB) family of transcriptional factors; and 3) whether the specific inhibition of K(V)3.4 channel subunit reverts the Abeta peptide-induced neurodegeneration in hippocampal neurons and nerve growth factor (NGF)-differentiated PC-12 cells. We found that Abeta(1-42) treatment induced an increase in K(V)3.4 and MIRP2 transcripts and proteins, detected by reverse transcription-polymerase chain reaction and Western blot analysis, respectively, in NGF-differentiated PC-12 cells and hippocampal neurons. Patch-clamp experiments performed in whole-cell configuration revealed that the Abeta peptide caused an increase in I(A) current amplitude carried by K(V)3.4 channel subunits, as revealed by their specific blockade with blood depressing substance-I (BDS-I) in both hippocampal neurons and NGF-differentiated PC-12 cells. The inhibition of NF-kappaB nuclear translocation with the cell membrane-permeable peptide SN-50 prevented the increase in K(V)3.4 protein and transcript expression. In addition, the SN-50 peptide was able to block Abeta(1-42)-induced increase in K(V)3.4 K(+) currents and to prevent cell death caused by Abeta(1-42) exposure. Finally, BDS-I produced a similar neuroprotective effect by inhibiting the increase in K(V)3.4 expression. As a whole, our data indicate that K(V)3.4 channels could be a novel target for Alzheimer's disease pharmacological therapy.


Subject(s)
Amyloid beta-Peptides/toxicity , Apoptosis/drug effects , Neurons/drug effects , Peptide Fragments/toxicity , Peptides/metabolism , Shaw Potassium Channels/metabolism , Up-Regulation/drug effects , Amyloid beta-Peptides/chemistry , Animals , Cell Death/drug effects , Cells, Cultured , Cnidarian Venoms/pharmacology , Electrophysiology , Hippocampus/cytology , Hippocampus/embryology , NF-kappa B/antagonists & inhibitors , Neurons/cytology , Neurons/physiology , PC12 Cells , Patch-Clamp Techniques , Peptide Fragments/chemistry , Peptides/genetics , Peptides/pharmacology , RNA, Messenger/analysis , Rats , Rats, Wistar , Sea Anemones/chemistry , Shaw Potassium Channels/genetics
9.
Neurology ; 63(1): 57-65, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15249611

ABSTRACT

BACKGROUND: Benign familial neonatal convulsion (BFNC) is a rare autosomal dominant disorder caused by mutations in two genes, KCNQ2 and KCNQ3, encoding for potassium channel subunits underlying the M-current. This current limits neuronal hyperexcitability by causing spike-frequency adaptation. METHODS: The authors describe a BFNC family with four affected members: two of them exhibit BFNC only while the other two, in addition to BFNC, present either with a severe epileptic encephalopathy or with focal seizures and mental retardation. RESULTS: All affected members of this family carry a novel missense mutation in the KCNQ2 gene (K526N), disrupting the tri-dimensional conformation of a C-terminal region of the channel subunit involved in accessory protein binding. When heterologously expressed in CHO cells, potassium channels containing mutant subunits in homomeric or heteromeric configuration with wild-type KCNQ2 and KCNQ3 subunits exhibit an altered voltage-dependence of activation, without changes in intracellular trafficking and plasma membrane expression. CONCLUSION: The KCNQ2 K526N mutation may affect M-channel function by disrupting the complex biochemical signaling involving KCNQ2 C-terminus. Genetic rather than acquired factors may be involved in the pathophysiology of the phenotypic variability of the neurologic symptoms associated with BFNC in the described family.


Subject(s)
Amino Acid Substitution , Epilepsy, Benign Neonatal/genetics , Intellectual Disability/genetics , Mutation, Missense , Point Mutation , Potassium Channels, Voltage-Gated/genetics , Adult , Amino Acid Sequence , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , CHO Cells , Cricetinae , Cricetulus , Drug Resistance/genetics , Epilepsies, Partial/drug therapy , Epilepsies, Partial/genetics , Epilepsy, Benign Neonatal/drug therapy , Female , Humans , Infant, Newborn , Ion Channel Gating , Ion Transport , KCNQ2 Potassium Channel , Magnetic Resonance Imaging , Male , Molecular Sequence Data , Pedigree , Phenotype , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/physiology , Protein Conformation , Protein Subunits , Quadriplegia/genetics , Structure-Activity Relationship
11.
Neurology ; 61(1): 131-4, 2003 Jul 08.
Article in English | MEDLINE | ID: mdl-12847176

ABSTRACT

Patients with benign familial neonatal convulsions (BFNC) may develop various epilepsies or epilepsy-associated EEG traits. A heterozygous 1-base pair deletion (2043DeltaT) in the KCNQ2 gene encoding for K+ channel subunits was found in a patient with BFNC who showed centrotemporal spikes at age 3 years. Electrophysiologic studies showed that mutant K+ channel subunits failed to give rise to functional homomeric channels or exert dominant-negative effects when expressed with KCNQ2/KCNQ3 subunits.


Subject(s)
Epilepsy, Benign Neonatal/diagnosis , Epilepsy, Benign Neonatal/genetics , Mutation , Potassium Channels/genetics , Temporal Lobe/physiopathology , Action Potentials , Animals , CHO Cells , Cells, Cultured , Child , Child, Preschool , Cricetinae , DNA Mutational Analysis , Electroencephalography , Electrophysiology , Epilepsy, Benign Neonatal/physiopathology , Female , Gene Transfer Techniques , Humans , KCNQ2 Potassium Channel , Male , Oocytes/metabolism , Pedigree , Potassium Channels/metabolism , Potassium Channels, Voltage-Gated , Protein Subunits/genetics , Protein Subunits/metabolism , Xenopus
12.
Clin Exp Allergy ; 32(4): 489-98, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11972592

ABSTRACT

This review addresses novel concepts of histamine H1-receptor function and attempts to relate them to the anti-inflammatory effects of H1-antihistamines. Furthermore, the molecular mechanisms underlying the cardiotoxic effects of H1-antihistamines are discussed. H1-receptors are G-protein-coupled-receptors (GPCRs), the inactive and active conformations of which coexist in equilibrium. The degree receptor activation in the absence of histamine is its 'constitutive activity'. In this two-state model, histamine acts as an agonist by combining with and stabilizing the activated conformation of the H1-receptor to shift the equilibrium towards the activated state. Drugs classified previously as antagonists act as either inverse agonists or neutral antagonists. Inverse agonists combine with and stabilize the inactive conformation of the receptor to shift the equilibrium towards the inactive state. Thus, they may down-regulate constitutive receptor activity, even in the absence of histamine. Neutral antagonists combine equally with both conformations of the receptor, do not affect basal receptor activity but do interfere with agonist binding. All H1-antihistamines examined to date are inverse agonists. As the term 'H1-receptor antagonists' is obviously erroneous, we suggest that it be replaced by 'H1-antihistamines'. The observations that H1-receptors modulate NF-kappaB activation and that there are complex interactions between GPCRs, has allowed us to postulate receptor dependent-mechanisms for some anti-inflammatory effects of H1-antihistamines, e.g. inhibition of ICAM-1 expression and the effects of bradykinin. Finally, the finding that blockade of HERG1 K+ channels is the mechanism by which some H1-antihistamines may cause cardiac arrhythmias has allowed the development of preclinical tests to predict such activity.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cation Transport Proteins , DNA-Binding Proteins , Histamine H1 Antagonists/adverse effects , Histamine H1 Antagonists/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels , Trans-Activators , Animals , Anti-Inflammatory Agents/pharmacology , Bradykinin/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Histamine Agonists/pharmacology , Histamine Release/drug effects , Humans , Models, Biological , Potassium Channel Blockers , Transcriptional Regulator ERG
13.
Biochem Pharmacol ; 62(9): 1229-38, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11705456

ABSTRACT

In the present study, the effect of the blockade of membrane calcium channels activated by intracellular Ca(2+) store depletion on basal and depolarization-induced [3H]norepinephrine ([3H]NE) release from SH-SY5Y human neuroblastoma cells was examined. The second-generation H(1) receptor blockers astemizole, terfenadine, and loratadine, as well as the first-generation compound hydroxyzine, inhibited [3H]NE release induced by high extracellular K(+) concentration ([K(+)](e)) depolarization in a concentration-dependent manner (the IC(50)s were 2.3, 1.7, 4.8, and 9.4 microM, respectively). In contrast, the more hydrophilic second-generation H(1) receptor blocker cetirizine was completely ineffective (0.1-30 microM). The inhibition of high [K(+)](e)-induced [3H]NE release by H(1) receptor blockers seems to be related to their ability to inhibit Ca(2+) channels activated by Ca(i)(2+) store depletion (SOCs). In fact, astemizole, terfenadine, loratadine, and hydroxyzine, but not cetirizine, displayed a dose-dependent inhibitory action on the increase in intracellular Ca(2+) concentrations ([Ca(2+)](i)) obtained with extracellular Ca(2+) reintroduction after Ca(i)(2+) store depletion with thapsigargin (1 microM), an inhibitor of the sarcoplasmic-endoplasmic reticulum calcium ATPase (SERCA) pump. The rank order of potency for SOC inhibition by these compounds closely correlated with their inhibitory properties on depolarization-induced [3H]NE release from SH-SY5Y human neuroblastoma cells. Nimodipine (1 microM) plus omega-conotoxin (100 nM) did not interfere with the present model for SOC activation. In addition, the inhibition of depolarization-induced [3H]NE release does not seem to be attributable to the blockade of the K(+) currents carried by the K(+) channels encoded by the human Ether-a-Gogo Related Gene (I(HERG)) by these antihistamines. In fact, whole-cell voltage-clamp experiments revealed that the IC(50) for astemizole-induced hERG blockade is about 300-fold lower than that for the inhibition of high K(+)-induced [3H]NE release. Furthermore, current-clamp experiments in SH-SY5Y cells showed that concentrations of astemizole (3 microM) which were effective in preventing depolarization-induced [3H]NE release were unable to interfere with the cell membrane potential under depolarizing conditions (100 mM [K(+)](e)), suggesting that hERG K(+) channels do not contribute to membrane potential control during exposure to elevated [K(+)](e). Collectively, the results of the present study suggest that, in SH-SY5Y human neuroblastoma cells, the inhibition of SOCs by some second-generation antihistamines can prevent depolarization-induced neurotransmitter release.


Subject(s)
Calcium Channels/metabolism , Cation Transport Proteins , DNA-Binding Proteins , Histamine H1 Antagonists/pharmacology , Norepinephrine/metabolism , Potassium Channels, Voltage-Gated , Receptors, Histamine H1/metabolism , Trans-Activators , Astemizole/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Cetirizine/pharmacology , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Hydroxyzine/pharmacology , Loratadine/pharmacology , Neuroblastoma , Potassium Channels/metabolism , Terfenadine/pharmacology , Transcriptional Regulator ERG , Tritium , Tumor Cells, Cultured
15.
Drugs ; 61(2): 207-36, 2001.
Article in English | MEDLINE | ID: mdl-11270939

ABSTRACT

Second generation antihistamines are recognised as being highly effective treatments for allergy-based disease and are among the most frequently prescribed and safest drugs in the world. However, consideration of the therapeutic index or the benefit/risk ratio of the H1 receptor antagonists is of paramount importance when prescribing this class of compounds as they are used to treat non-life threatening conditions. There are many second generation antihistamines available and at first examination these appear to be comparable in terms of safety and efficacy. However, the newer antihistamines in fact represent a heterogeneous group of compounds, having markedly differing chemical structures, adverse effects, half-life, tissue distribution and metabolism, spectrum of antihistaminic properties, and varying degrees of anti-inflammatory effects. With regard to the latter, there is growing awareness that some of these compounds might represent useful adjunct medications in asthma therapy. In terms of safety issues, the current second generation grouping includes compounds with proven cardiotoxic effects and others with the potential for adverse drug interactions. Moreover, some of the second generation H1 antagonists have given cause for concern regarding their potential to cause a degree of somnolence in some individuals. It can be argued, therefore, that the present second generation grouping is too large and indistinct since this was based primarily on the concept of separating the first generation sedating compounds from nonsedating H1 antagonists. Although it is too early to talk about a third generation grouping of antihistamines, future membership of such a classification could be based on a low volume of distribution coupled with a lack of sedating effects, drug interactions and cardiotoxicity.


Subject(s)
Asthma/drug therapy , Histamine H1 Antagonists/adverse effects , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/pharmacokinetics , Anti-Asthmatic Agents/therapeutic use , Central Nervous System/drug effects , Contraindications , Heart/drug effects , Histamine H1 Antagonists/pharmacokinetics , Histamine H1 Antagonists/therapeutic use , Humans , Inflammation/drug therapy , Inflammation Mediators/metabolism , Receptors, Histamine H1/metabolism
16.
J Immunol ; 166(6): 4083-91, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238657

ABSTRACT

Increasing evidence suggests that a continuous release of histamine from mast cells occurs in the airways of asthmatic patients and that histamine may modulate functions of other inflammatory cells such as macrophages. In the present study histamine (10(-9)-10(-6) M) increased in a concentration-dependent fashion the basal release of beta-glucuronidase (EC(50) = 8.2 +/- 3.5 x 10(-9) M) and IL-6 (EC(50) = 9.3 +/- 2.9 x 10(-8) M) from human lung macrophages. Enhancement of beta-glucuronidase release induced by histamine was evident after 30 min and peaked at 90 min, whereas that of IL-6 required 2-6 h of incubation. These effects were reproduced by the H(1) agonist (6-[2-(4-imidazolyl)ethylamino]-N-(4-trifluoromethylphenyl)heptane carboxamide but not by the H(2) agonist dimaprit. Furthermore, histamine induced a concentration-dependent increase of intracellular Ca(2+) concentrations ([Ca(2+)](i)) that followed three types of response, one characterized by a rapid increase, a second in which [Ca(2+)](i) displays a slow but progressive increase, and a third characterized by an oscillatory pattern. Histamine-induced beta-glucuronidase and IL-6 release and [Ca(2+)](i) elevation were inhibited by the selective H(1) antagonist fexofenadine (10(-7)-10(-4) M), but not by the H(2) antagonist ranitidine. Inhibition of histamine-induced beta-glucuronidase and IL-6 release by fexofenadine was concentration dependent and displayed the characteristics of a competitive antagonism (K(d) = 89 nM). These data demonstrate that histamine induces exocytosis and IL-6 production from human macrophages by activating H(1) receptor and by increasing [Ca(2+)](i) and they suggest that histamine may play a relevant role in the long-term sustainment of allergic inflammation in the airways.


Subject(s)
Exocytosis/immunology , Histamine/analogs & derivatives , Histamine/physiology , Interleukin-6/biosynthesis , Lung/immunology , Lung/metabolism , Macrophages, Alveolar/metabolism , Receptors, Histamine H1/metabolism , Calcium/metabolism , Calcium/physiology , Cytosol/metabolism , Dimaprit/pharmacology , Dose-Response Relationship, Immunologic , Glucuronidase/metabolism , Histamine/pharmacology , Histamine Agonists/pharmacology , Histamine H1 Antagonists/pharmacology , Histamine H2 Antagonists/pharmacology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/cytology , Lung/enzymology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/immunology , RNA, Messenger/biosynthesis , Toluidines/pharmacology , Up-Regulation/immunology
17.
J Mol Cell Cardiol ; 32(12): 2327-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11113008

ABSTRACT

Mutations in the cardiac potassium channel HERG (KCNH2) cause chromosome 7-linked long QT syndrome (LQT2) characterized by a prolonged QT interval, recurrent syncope and sudden cardiac death. Most mutations in HERG exhibit "loss of function" phenotypes with defective channels either inserted into the plasma membrane or retained in the endoplasmic reticulum. "Loss of function" mutations reduce I(Kr), the cardiac delayed rectifier current encoded by HERG, due to haploinsufficiency or suppression of wild-type function by a dominant-negative mechanism. One explanation for dominant-negative current suppression is that mutant subunits render tetrameric channel complexes non-conducting on co-assembly. In the present paper we describe an alternative mechanism for this phenomenon. We show (1) that the dominant-negative HERG mutation A561V is retained in the endoplasmic reticulum and (2) that wild-type channels are tagged for retention in the ER by co-assembly with trafficking deficient A561V subunits. Thus, in HERG A561V dominant-negative suppression of wild-type function is the result of an acquired trafficking defect.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Endoplasmic Reticulum/metabolism , Long QT Syndrome/genetics , Mutation , Potassium Channels, Voltage-Gated , Trans-Activators , Animals , Blotting, Western , COS Cells , Cell Membrane/metabolism , Cloning, Molecular , Codon , DNA, Complementary/metabolism , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Genes, Dominant , Humans , Immunoblotting , Microscopy, Fluorescence , Patch-Clamp Techniques , Phenotype , Potassium Channels/genetics , Suppression, Genetic , Transcriptional Regulator ERG
18.
Br J Pharmacol ; 131(6): 1081-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11082114

ABSTRACT

1. Ventricular arrhythmias are rare but life-threatening side effects of therapy with the second-generation H(1) receptor antagonists terfenadine and astemizole. Blockade of the K(+) channels encoded by the Human Ether-à-go-go-Related Gene 1 (HERG1) K(+) channels, which is the molecular basis of the cardiac repolarizing current I(Kr), by prolonging cardiac repolarization, has been recognized as the mechanism underlying the cardiac toxicity of these compounds. 2. In the present study, the potential blocking ability of the novel second-generation H(1) receptor antagonist mizolastine of the HERG1 K(+) channels heterologously expressed in Xenopus oocytes and in HEK 293 cells or constitutively present in SH-SY5Y human neuroblastoma cells has been examined and compared to that of astemizole. 3. Mizolastine blocked HERG1 K(+) channels expressed in Xenopus oocytes with an estimated IC(50) of 3.4 microM. Mizolastine blockade was characterized by a fast dissociation rate when compared to that of astemizole; when fitted to a monoexponential function, the time constants for drug dissociation from the K(+) channel were 72.4+/-11.9 s for 3 microM mizolastine, and 1361+/-306 s for 1 microM astemizole. 4. In human embryonic kidney 293 cells (HEK 293 cells) stably transfected with HERG1 cDNA, extracellular application of mizolastine exerted a dose-related inhibitory action on I(HERG1), with an IC(50) of 350+/-76 nM. Furthermore, mizolastine dose-dependently inhibited HERG1 K(+) channels constitutively expressed in SH-SY5Y human neuroblastoma clonal cells. 5. The results of the present study suggest that the novel second-generation H(1) receptor antagonist mizolastine, in concentrations higher than those achieved in vivo during standard therapy, is able to block in some degree both constitutively and heterologously expressed HERG1 K(+) channels, and confirm the heterogeneity of molecules belonging to this therapeutical class with respect to their HERG1-inhibitory action.


Subject(s)
Astemizole/pharmacology , Benzimidazoles/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Histamine H1 Antagonists/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Trans-Activators , Animals , Cell Line , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Female , Humans , Potassium Channels/genetics , Transcriptional Regulator ERG , Xenopus
19.
Mol Pharmacol ; 58(5): 1115-28, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11040061

ABSTRACT

In the present study, the effects on intracellular calcium concentration ([Ca(2+)](i)) oscillations of the blockade of ether-a-go-go-related gene (ERG) K(+) channels and of Ca(2+) influx through store-operated channels (SOC) activated by [Ca(2+)](i) store depletion have been studied in GH(3) cells by means of a combination of single-cell fura-2 microfluorimetry and whole-cell mode of the patch-clamp technique. Nanomolar concentrations (1-30 nM) of the piperidinic second-generation antihistamines terfenadine and astemizole and of the class III antiarrhythmic methanesulfonanilide dofetilide, by blocking ERG K(+) channels, increased the frequency and the amplitude of [Ca(2+)](i) oscillations in resting oscillating GH(3) cells. These compounds also induced the appearance of an oscillatory pattern of [Ca(2+)](i) in a subpopulation of nonoscillating GH(3) cells. The effects of ERG K(+) channel blockade on [Ca(2+)](i) oscillations appeared to be due to the activation of L-type Ca(2+) channels, because they were prevented by 300 nM nimodipine. By contrast, the piperazinic second-generation antihistamine cetirizine (0.01-30 microM), which served as a negative control, failed to affect ERG K(+) channels and did not interfere with [Ca(2+)](i) oscillations in GH(3) cells. Interestingly, micromolar concentrations of terfenadine and astemizole (0.3-30 microM), but not of dofetilide (10-100 microM), produced an inhibition of the spontaneous oscillatory pattern of [Ca(2+)](i) changes. This effect was possibly related to an inhibition of SOC, because these compounds inhibited the increase of [Ca(2+)](i) achieved by extracellular calcium reintroduction after intracellular calcium store depletion with the sarcoplasmic or endoplasmic reticulum calcium ATPase pump inhibitor thapsigargin (10 microM) in an extracellular calcium-free medium. The same inhibitory effect on [Ca(2+)](i) oscillations and SOC was observed with the first-generation antihistamine hydroxyzine (1-30 microM), the more hydrophobic metabolic precursor of cetirizine. Collectively, the results of the present study obtained with compounds that interfere in a different concentration range with ERG K(+) channels or SOC suggest that 1) ERG K(+) channels play a relevant role in controlling the oscillatory pattern of [Ca(2+)](i) in resting GH(3) cells and 2) the inhibition of SOC might induce an opposite effect, i.e., an inhibition of [Ca(2+)](i) oscillations.


Subject(s)
Calcium/metabolism , Cation Transport Proteins , Histamine H1 Antagonists/pharmacology , Pituitary Gland/drug effects , Potassium Channel Blockers , Potassium Channels, Calcium-Activated , Potassium Channels, Voltage-Gated , Animals , Anti-Arrhythmia Agents/pharmacology , Astemizole/pharmacology , Biological Transport/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium Channels, L-Type/metabolism , Cells, Cultured , Cetirizine/pharmacology , Electrophysiology , Ether-A-Go-Go Potassium Channels , Hydroxyzine/pharmacology , Large-Conductance Calcium-Activated Potassium Channels , Nimodipine/pharmacology , Phenethylamines/pharmacology , Pituitary Gland/cytology , Pituitary Gland/metabolism , Potassium Channels/metabolism , Rats , Small-Conductance Calcium-Activated Potassium Channels , Sulfonamides/pharmacology , Terfenadine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...