1
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
2
|
Lawson K. Identification of the Involvement of Potassium Channels in Fibromyalgia. Front Biosci (Schol Ed) 2022; 14:29. [PMID: 36575839 DOI: 10.31083/j.fbs1404029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/05/2022] [Accepted: 09/28/2022] [Indexed: 01/05/2023]
Abstract
Fibromyalgia is a central sensitivity syndrome that presents with chronic pain, fatigue, cognitive dysfunction, and disordered sleep. The pathophysiology which due to multisensory hypersensitivity of the central nervous system involves neuronal excitability leading to central sensitization. Treatments of the challenges associated with the complexities of fibromyalgia involve combinations of pharmacological and non-pharmacological therapeutic approaches which often offer limited benefit. Potassium (K+) channels play a fundamental role in establishing and maintaining stability of neuronal activity. The large molecular diversity and distribution of K+ channels support involvement in a broad range of physiological functions. In nociceptive pathways, neuronal hyperexcitability leading to pain sensation has been associated with reduced function of K+ channels and loss of cellular control. This article reviews the evidence of involvement of K+ channels in fibromyalgia. A potential role both in the pathophysiological processes responsible for the symptoms of fibromyalgia and as therapeutic targets for the management of the condition is considered.
Collapse
Affiliation(s)
- Kim Lawson
- Department of Biosciences and Chemistry, Biomolecular Sciences Research Centre, Sheffield Hallam University, Faculty of Health and Wellbeing, S1 1WB Sheffield, UK
| |
Collapse
|
3
|
Cheng L, Fu H, Wang X, Ye L, Lakhani I, Tse G, Zhang Z, Liu T, Li G. Effects of ticagrelor pretreatment on electrophysiological properties of stellate ganglion neurons following myocardial infarction. Clin Exp Pharmacol Physiol 2020; 47:1932-1942. [PMID: 33459403 DOI: 10.1111/1440-1681.13385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 06/21/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022]
Abstract
Higher sympathetic activity predisposes to malignant ventricular arrhythmias in the context of myocardial infarction (MI). This is, in part, mediated by the electrical activity of the stellate ganglion (SG). The aim of this study is to examine the effects of ticagrelor pretreatment on the electrophysiological properties of SG neurons following MI in rabbits. MI was induced by isoproterenol (ISO) of 150 mg kg-1 d-1 (twice at an interval of 24 hours). Ticagrelor pretreatment was administered at low- (10 mg kg-1 d-1) or high-dose (20 mg kg-1 d-1). Protein and RNA expression were determined by immunohistochemical analysis and real-time PCR, respectively. The activity of sodium channel current (INa), delayed rectifier potassium current (IKDR), M-type potassium current (IKM) as well as action potentials (APs) from SG neurons were measured by whole-cell patch-clamp. Intracellular calcium concentrations were measured by confocal microscopy. Compared with the control group, the MI group exhibited a greater amplitude of INa, IKDR and IKM, significantly altered activation and inactivation characteristics of INa, no significant alterations in protein or mRNA expression of sodium and M-type potassium channels, along with higher AP amplitude and frequency and intracellular calcium concentrations. Most of these abnormalities were prevented by pretreatment with low- or high-dose ticagrelor. Our data suggest that ticagrelor exerts cardioprotective effects, potentially through modulating the activity of different ion channels in SG neurons.
Collapse
Affiliation(s)
- Lijun Cheng
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Huaying Fu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xinghua Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Lan Ye
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ishan Lakhani
- Laboratory of Cardiovascular Physiology, Li Ka Shing Institute of Health Sciences, Hong Kong, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiwei Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Ion Channels Involved in Substance P-Mediated Nociception and Antinociception. Int J Mol Sci 2019; 20:ijms20071596. [PMID: 30935032 PMCID: PMC6479580 DOI: 10.3390/ijms20071596] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023] Open
Abstract
Substance P (SP), an 11-amino-acid neuropeptide, has long been considered an effector of pain. However, accumulating studies have proposed a paradoxical role of SP in anti-nociception. Here, we review studies of SP-mediated nociception and anti-nociception in terms of peptide features, SP-modulated ion channels, and differential effector systems underlying neurokinin 1 receptors (NK1Rs) in differential cell types to elucidate the effect of SP and further our understanding of SP in anti-nociception. Most importantly, understanding the anti-nociceptive SP-NK1R pathway would provide new insights for analgesic drug development.
Collapse
|
5
|
Sanson C, Schombert B, Filoche-Rommé B, Partiseti M, Bohme GA. Electrophysiological and Pharmacological Characterization of Human Inwardly Rectifying K ir2.1 Channels on an Automated Patch-Clamp Platform. Assay Drug Dev Technol 2019; 17:89-99. [PMID: 30835490 PMCID: PMC6479253 DOI: 10.1089/adt.2018.882] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Inwardly rectifying IK1 potassium currents of the heart control the resting membrane potential of ventricular cardiomyocytes during diastole and contribute to their repolarization after each action potential. Mutations in the gene encoding Kir2.1 channels, which primarily conduct ventricular IK1, are associated with inheritable forms of arrhythmias and sudden cardiac death. Therefore, potential iatrogenic inhibition of Kir2.1-mediated IK1 currents is a cardiosafety concern during new drug discovery and development. Kir2.1 channels are part of the panel of cardiac ion channels currently considered for refined early compound risk assessment within the Comprehensive in vitro Proarrhythmia Assay initiative. In this study, we have validated a cell-based assay allowing functional quantification of Kir2.1 inhibitors using whole-cell recordings of Chinese hamster ovary cells stably expressing human Kir2.1 channels. We reproduced key electrophysiological and pharmacological features known for native IK1, including current enhancement by external potassium and voltage- and concentration-dependent blockade by external barium. Furthermore, the Kir inhibitors ML133, PA-6, and chloroquine, as well as the multichannel inhibitors chloroethylclonidine, chlorpromazine, SKF-96365, and the class III antiarrhythmic agent terikalant demonstrated slowly developing inhibitory activity in the low micromolar range. The robustness of this assay authorizes medium throughput screening for cardiosafety purposes and could help to enrich the currently limited Kir2.1 pharmacology.
Collapse
Affiliation(s)
- Camille Sanson
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Brigitte Schombert
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Bruno Filoche-Rommé
- 2 Integrated Drug Discovery, Medicinal Chemistry, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Michel Partiseti
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - G Andrees Bohme
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| |
Collapse
|
6
|
Rubi L, Koenig X, Kubista H, Todt H, Hilber K. Decreased inward rectifier potassium current I K1 in dystrophin-deficient ventricular cardiomyocytes. Channels (Austin) 2016; 11:101-108. [PMID: 27560040 PMCID: PMC5398571 DOI: 10.1080/19336950.2016.1228498] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Kir2.x channels in ventricular cardiomyocytes (most prominently Kir2.1) account for the inward rectifier potassium current IK1, which controls the resting membrane potential and the final phase of action potential repolarization. Recently it was hypothesized that the dystrophin-associated protein complex (DAPC) is important in the regulation of Kir2.x channels. To test this hypothesis, we investigated potential IK1 abnormalities in dystrophin-deficient ventricular cardiomyocytes derived from the hearts of Duchenne muscular dystrophy mouse models. We found that IK1 was substantially diminished in dystrophin-deficient cardiomyocytes when compared to wild type myocytes. This finding represents the first functional evidence for a significant role of the DAPC in the regulation of Kir2.x channels.
Collapse
Affiliation(s)
- Lena Rubi
- a Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology , Medical University of Vienna , Vienna , Austria
| | - Xaver Koenig
- a Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology , Medical University of Vienna , Vienna , Austria
| | - Helmut Kubista
- a Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology , Medical University of Vienna , Vienna , Austria
| | - Hannes Todt
- a Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology , Medical University of Vienna , Vienna , Austria
| | - Karlheinz Hilber
- a Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
7
|
Abstract
INTRODUCTION Cardiac K(+) channels play a critical role in maintaining the normal electrical activity of the heart by setting the cell resting membrane potential and by determining the shape and duration of the action potential. Drugs that block the rapid (IKr) and slow (IKs) components of the delayed rectifier K(+) current have been widely used as class III antiarrhythmic agents. In addition, drugs that selectively target the ultra-rapid delayed rectifier current (IKur) and the acetylcholine-gated inward rectifier current (IKAch) have shown efficacy in the treatment of patients with atrial fibrillation. In order to meet the future demand for new antiarrhythmic agents, novel approaches for cardiac K(+) channel drug discovery will need to be developed. Further, K(+) channel screening assays utilizing primary and stem cell-derived cardiomyocytes will be essential for evaluating the cardiotoxicity of potential drug candidates. AREAS COVERED In this review, the author provides a brief background on the structure, function and pharmacology of cardiac voltage-gated and inward rectifier K(+) channels. He then focuses on describing and evaluating current technologies, such as ion flux and membrane potential-sensitive dye assays, used for cardiac K(+) channel drug discovery. EXPERT OPINION Cardiac K(+) channels will continue to represent significant clinical targets for drug discovery. Although fluorescent high-throughput screening (HTS) assays and automated patch clamp systems will remain the workhorse technologies for identifying lead compounds, innovations in the areas of microfluidics, micropatterning and biosensor fabrication will allow further growth of technologies using primary and stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Kenneth B Walsh
- University of South Carolina, School of Medicine, Department of Pharmacology, Physiology and Neuroscience , Columbia, SC 29209 , USA +1 803 216 3519 ; +1 803 216 3538 ;
| |
Collapse
|
8
|
Adachi T, Huxtable AG, Fang X, Funk GD. Substance P Modulation of Hypoglossal Motoneuron Excitability During Development: Changing Balance Between Conductances. J Neurophysiol 2010; 104:854-72. [DOI: 10.1152/jn.00016.2010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although Substance P (SP) acts primarily through neurokinin 1 (NK1) receptors to increase the excitability of virtually all motoneurons (MNs) tested, the ontogeny of this transmitter system is not known for any MN pool. Hypoglossal (XII) MNs innervate tongue protruder muscles and participate in several behaviors that must be functional from birth including swallowing, suckling and breathing. We used immunohistochemistry, Western immunoblotting, and whole cell recording of XII MNs in brain stem slices from rats ranging in age from postnatal day zero (P0) to P23 to explore developmental changes in: NK1 receptor expression; currents evoked by SPNK1 (an NK1-selective SP receptor agonist) and; the efficacy of transduction pathways transforming ligand binding into channel modulation. Despite developmental reductions in XII MN NK1 receptor expression, SPNK1 current density remained constant at 6.1 ± 1.0 (SE) pA/pF. SPNK1 activated at least two conductances. Activation of a pH-insensitive Na+ conductance dominated in neonates (P0–P5), but its contribution fell from ∼80 to ∼55% in juveniles (P14–P23). SPNK1 also inhibited a pH-sensitive, two-pore domain K+ (TASK)-like K+ current. Its contribution increased developmentally. First, the density of this pH-sensitive K+ current doubled between P0 and P23. Second, SPNK1 did not affect this current in neonates, but reduced it by 20% at P7–P10 and 80% in juveniles. In addition, potentiation of repetitive firing was greatest in juveniles. These data establish that despite apparent reductions in NK1 receptor density, SP remains an important modulator of XII MN excitability throughout postnatal development due, in part, to increased expression of a pH-sensitive, TASK-like conductance.
Collapse
Affiliation(s)
- Tadafumi Adachi
- Department of Physiology, School of Molecular and Systems Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
- Department of Physiology, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand
| | - Adrianne G. Huxtable
- Department of Physiology, School of Molecular and Systems Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
| | - X. Fang
- Department of Physiology, School of Molecular and Systems Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
| | - Gregory D. Funk
- Department of Physiology, School of Molecular and Systems Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
- Department of Physiology, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand
| |
Collapse
|
9
|
Abstract
Cough is the most common symptom for which individuals seek medical attention and spend health-care dollars. Despite the burden induced by cough, the current treatments for cough are only partially effective. Delineating the sites and mechanisms in the cough central network for changes in the cough reflex could lead to new therapeutic strategies and drug target sites for more effective treatments. The first synaptic target in the CNS for the cough-related sensory input is the second-order neurons in the nucleus tractus solitarius (NTS); these neurons reorganize the primary sensory information into a coherent output. The NTS neurons have been shown to undergo neuroplasticity under a variety of conditions, such as respiratory disorders, stress, and exposures to environmental pollutants. The NTS contains a rich innervation of substance P immunoreactive nerve terminals, suggesting that substance P might be important in altered cough reflex response. This chapter summarizes our current findings on the role of substance P in enhanced cough reflex as well as the potential NTS targets for the action of substance P.
Collapse
Affiliation(s)
- C-Y Chen
- Department of Pharmacology, University of California, Davis School of Medicine, 4150 V Street, 1104 PSSB, Sacramento, CA 95817, USA
| | | | | | | |
Collapse
|
10
|
Tompkins JD, Parsons RL. Exocytotic release of ATP and activation of P2X receptors in dissociated guinea pig stellate neurons. Am J Physiol Cell Physiol 2006; 291:C1062-71. [PMID: 16760262 DOI: 10.1152/ajpcell.00472.2005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of P2X receptors by a Ca(2+)- and soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein-dependent release of ATP was measured using patch-clamp recordings from dissociated guinea pig stellate neurons. Asynchronous transient inward currents (ASTICs) were activated by depolarization or treatment with the Ca(2+) ionophore ionomycin (1.5 and 3 microM). During superfusion with a HEPES-buffered salt solution containing 2.5 mM Ca(2+), depolarizing voltage steps (-60 to 0 mV, 500 ms) evoked ASTICs on the decaying phase of a larger, transient inward current. Equimolar substitution of Ba(2+) for Ca(2+) augmented the postdepolarization frequency of ASTICs, while eliminating the larger transient current. Perfusion with an ionomycin-containing solution elicited a sustained activation of ASTICs, allowing quantitative analysis over a range of holding potentials. Under these conditions, increasing extracellular [Ca(2+)] to 5 mM increased ASTIC frequency, whereas no events were observed following replacement of Ca(2+) with Mg(2+), demonstrating a Ca(2+) requirement. ASTICs were Na(+) dependent, inwardly rectifying, and reversed near 0 mV. Treatment with the nonselective purinergic receptor antagonist pyridoxal phosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) (10 microM) blocked all events under both conditions, whereas the ganglionic nicotinic antagonist hexamethonium (100 microM and 1 mM) had no effect. PPADS also blocked the macroscopic inward current evoked by exogenously applied ATP (300 microM). The presence of botulinum neurotoxin E (BoNT/E) in the whole-cell recording electrode significantly attenuated the ionomycin-induced ASTIC activity, whereas phorbol ester treatment potentiated this activity. These results suggest that ASTICs are mediated by vesicular release of ATP and activation of P2X receptors.
Collapse
Affiliation(s)
- John D Tompkins
- University of Vermont, College of Medicine, Dept. of Anatomy and Neurobiology, Burlington, VT 05405, USA.
| | | |
Collapse
|
11
|
Fernández-Velasco M, Ruiz-Hurtado G, Delgado C. I K1 and I f in ventricular myocytes isolated from control and hypertrophied rat hearts. Pflugers Arch 2006; 452:146-54. [PMID: 16395601 DOI: 10.1007/s00424-005-0024-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2005] [Revised: 11/02/2005] [Accepted: 11/11/2005] [Indexed: 11/26/2022]
Abstract
Electrophysiological properties of inward rectifier potassium current (I (K1)) and hyperpolarization-activated inward current (I (f)) and the protein expression of the Kir2.1 subfamily and the hyperpolarization-activated cation channel 2 (HCN2) and HCN4 were studied in control and hypertrophied myocytes. Electrophysiological experiments were conducted by whole-cell patch-clamp technique, and protein levels of Kir2.1 subfamily and HCN2 and HCN4 isoforms were analysed by Western blot technique. The density of I (f) as well as the protein expression levels of the HCN2 isoform was found to be significantly higher in hypertrophied myocytes, whereas the protein expression level of HCN4 was not detected in any group. I (K1) density and Kir 2.1 protein expression were similar in control and hypertrophied myocytes, but the time-course of the currents was slower in hypertrophied myocytes. Analysis of I (f) and I (K1) in the same control and hypertrophied myocyte at -80 mV showed that cells in which I (f) was present had values of I (K1) density similar to those cells in which I (f) was not observed. In conclusion, although left ventricular hypertrophy involves an up-regulation of I (f) and its molecular correlate HCN2 in the rat ventricle, its contribution to diastolic depolarization would be limited by the low values of I (f) density at potentials close to the resting potential of the ventricular cells.
Collapse
Affiliation(s)
- María Fernández-Velasco
- Institute of Pharmacology and Toxicology (CSIC-UCM), School of Medicine, Universidad Complutense, 28040 Madrid, Spain
| | | | | |
Collapse
|
12
|
Priori SG, Pandit SV, Rivolta I, Berenfeld O, Ronchetti E, Dhamoon A, Napolitano C, Anumonwo J, di Barletta MR, Gudapakkam S, Bosi G, Stramba-Badiale M, Jalife J. A novel form of short QT syndrome (SQT3) is caused by a mutation in the KCNJ2 gene. Circ Res 2005; 96:800-7. [PMID: 15761194 DOI: 10.1161/01.res.0000162101.76263.8c] [Citation(s) in RCA: 407] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Short QT syndrome (SQTS) leads to an abbreviated QTc interval and predisposes patients to life-threatening arrhythmias. To date, two forms of the disease have been identified: SQT1, caused by a gain of function substitution in the HERG (I(Kr)) channel, and SQT2, caused by a gain of function substitution in the KvLQT1 (I(Ks)) channel. Here we identify a new variant, "SQT3", which has a unique ECG phenotype characterized by asymmetrical T waves, and a defect in the gene coding for the inwardly rectifying Kir2.1 (I(K1)) channel. The affected members of a single family had a G514A substitution in the KCNJ2 gene that resulted in a change from aspartic acid to asparagine at position 172 (D172N). Whole-cell patch-clamp studies of the heterologously expressed human D172N channel demonstrated a larger outward I(K1) than the wild-type (P<0.05) at potentials between -75 mV and -45 mV, with the peak current being shifted in the former with respect to the latter (WT, -75 mV; D172N, -65 mV). Coexpression of WT and mutant channels to mimic the heterozygous condition of the proband yielded an outward current that was intermediate between WT and D172N. In computer simulations using a human ventricular myocyte model the increased outward I(K1) greatly accelerated the final phase of repolarization, and shortened the action potential duration. Hence, unlike the known mutations in the two other SQTS forms (N588K in HERG and V307L in KvLQT1), simulations using the D172N and WT/D172N mutations fully accounted for the ECG phenotype of tall and asymmetrically shaped T waves. Although we were unable to test for inducibility of arrhythmia susceptibility due to lack of patients' consent, our computer simulations predict a steeper steady-state restitution curve for the D172N and WT/D172N mutation, compared with WT or to HERG or KvLQT1 mutations, which may predispose SQT3 patients to a greater risk of reentrant arrhythmias.
Collapse
Affiliation(s)
- Silvia G Priori
- Molecular Cardiology, IRCCS Fondazione Maugeri, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Endoh T. Modulation of voltage-dependent calcium channels by neurotransmitters and neuropeptides in parasympathetic submandibular ganglion neurons. Arch Oral Biol 2004; 49:539-57. [PMID: 15126136 DOI: 10.1016/j.archoralbio.2004.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2004] [Indexed: 12/20/2022]
Abstract
The control of saliva secretion is mainly under parasympathetic control, although there also could be a sympathetic component. Sympathetic nerves are held to have a limited action in secretion in submandibular glands because, on electrical stimulation, only a very small increase to the normal background, basal secretion occurs. Parasympathetic stimulation, on the other hand, caused a good flow of saliva with moderate secretion of acinar mucin, plus an extensive secretion of granules from the granular tubules. The submandibular ganglion (SMG) is a parasympathetic ganglion which receives inputs from preganglionic cholinergic neurons, and innervates the submandibular salivary gland to control saliva secretion. Neurotransmitters and neuropeptides acting via G-protein coupled receptors (GPCRs) change the electrical excitability of neurons. In these neurons, many neurotransmitters and neuropeptides modulate voltage-dependent calcium channels (VDCCs). The modulation is mediated by a family of GPCRs acting either directly through the membrane delimited G-proteins or through second messengers. However, the mechanism of modulation and the signal transduction pathway linked to an individual GPCRs depend on the animal species. This review reports how neurotransmitters and neuropeptides modulate VDCCs and how these modulatory actions are integrated in SMG systems. The action of neurotransmitters and neuropeptides on VDCCs may provide a mechanism for regulating SMG excitability and also provide a cellular mechanism of a variety of neuronal Ca(2+)-dependent processes.
Collapse
Affiliation(s)
- Takayuki Endoh
- Department of Physiology, Tokyo Dental College, 1-2-2 Masago, Mihama-ku, Chiba 261-8502, Japan.
| |
Collapse
|
14
|
Fuller BC, Sumner AD, Kutzler MA, Ruiz-Velasco V. A novel approach employing ultrasound guidance for percutaneous cardiac muscle injection to retrograde label rat stellate ganglion neurons. Neurosci Lett 2004; 363:252-6. [PMID: 15182954 DOI: 10.1016/j.neulet.2004.03.087] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2004] [Revised: 03/26/2004] [Accepted: 03/30/2004] [Indexed: 11/18/2022]
Abstract
Stellate ganglion (SG) neurons provide the main sympathetic innervation to the heart and help to regulate cardiac function. The purpose of this study was to determine if ultrasound imaging could be employed to retrograde label rat SG neurons innervating the heart without employing thoracotomy. In addition, electrophysiological experiments were performed to characterize the modulation of Ca(2+) channels by neurotransmitters in unlabeled and dye-labeled SG neurons. Fluorescence imaging of actutely isolated cells revealed that dye uptake was successful within five days following injection of dye in the cardiac muscle. Whole-cell voltage-clamp recordings revealed that the majority of the Ca(2+) current was carried by N-type Ca(2+) channels. Finally, fluorescence dye uptake did not appear to affect the modulation of Ca(2+) currents following exposure of SG neurons to norepinephrine, adenosine and neurokinin A. These results demonstrate that ultrasound imaging-guided percutaneous injection can be effectively employed to retrograde label neurons innervating the heart.
Collapse
Affiliation(s)
- Brad C Fuller
- Vascular Laboratories, Guthrie Healthcare System, Sayre, PA 18840, USA
| | | | | | | |
Collapse
|
15
|
Sun W, Ding DL, Wang P, Sun J, Jin X, Salvi RJ. Substance P inhibits potassium and calcium currents in inner ear spiral ganglion neurons. Brain Res 2004; 1012:82-92. [PMID: 15158164 DOI: 10.1016/j.brainres.2004.03.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2004] [Indexed: 10/26/2022]
Abstract
Substance P (SP), a member of the tachykinin family of neurotransmitters and neuromodulators, has been identified on spiral ganglion neurons (SGNs) in the inner ear; however, its high affinity receptor, neurokinin-1 (NK1), has not been identified and the physiological effects of SP on SGNs are not well understood. To address these issues, immunolabeling, RT-PCR, Western blots and whole-cell patch-clamp recordings were made from SGNs in P0-P5 mouse cochlear organotypic cultures. The NK1 receptor was detected on SGNs by immunocytochemistry, the protein was detected in cochlear tissues by Western blots, and the mRNA for the NK1 receptor was also found in cochlear tissues of postnatal mice (P2) by RT-PCR. Application of SP (1 to 25 microM) significantly increased the latency of SGN action potentials (APs) (mean increase 7.8 +/- 4 ms; 25 microM of SP), prolonged the duration of the action potential and made the resting potential (RP) more positive (mean 9.0 +/- 7 mV) relative to normal values (-54 +/- 6 mV). SP (1 to 25 microM) also suppressed voltage-activated potassium currents (IK+) and calcium currents (ICa2+). Puffing 25 microM of SP onto SGNs suppressed IK+ by 43 +/- 9% (n = 7) and ICa2+ by 40.6 +/- 5.6% (n = 7); both currents recovered when SP was washed out. A SP antagonist blocked the SP-induced suppression of IK+ and ICa2+. These results indicate that SP acting through NK1 receptors can have direct neuromodulatory effects on SGNs.
Collapse
Affiliation(s)
- Wei Sun
- Center for Hearing and Deafness, Hear. Res. Lab, SUNY University at Buffalo, 215 Parker Hall South Campus of UB, UB3435 Main Street, Buffalo, NY 14214, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Locknar SA, Barstow KL, Tompkins JD, Merriam LA, Parsons RL. Calcium-induced calcium release regulates action potential generation in guinea-pig sympathetic neurones. J Physiol 2004; 555:627-35. [PMID: 14724192 PMCID: PMC1664869 DOI: 10.1113/jphysiol.2003.059485] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2003] [Accepted: 01/14/2004] [Indexed: 11/08/2022] Open
Abstract
Experiments were done using guinea-pig sympathetic neurones dissociated from the stellate ganglia to establish whether calcium-induced calcium release (CICR) modulated action potential (AP) generation in mammalian neurones. Using measurements of intracellular calcium ([Ca(2+)](i)) with the Ca(2+)-sensitive dye fluo-3, we demonstrated that 10 mM caffeine activated ryanodine receptors and caused a rise in [Ca(2+)](i) in both Ca(2+)-containing and Ca(2+)-deficient solutions. We also demonstrated that combined treatment with caffeine and 1 microm thapsigargin or caffeine and 20 microm ryanodine blocked subsequent caffeine-induced elevations of [Ca(2+)](i). Treatment with thapsigargin, ryanodine or 200 microM Cd(2+) to disrupt CICR decreased the latency to AP generation during 400 ms depolarizing current ramps using the perforated patch whole cell patch clamp in current clamp mode. Treatment with 500 microM tetraethylammonium also decreased the latency to AP generation during depolarizing current ramps in control cells, but not in cells pretreated with thapsigargin to deplete internal Ca(2+) stores. In summary, we propose that an outward current, carried at least in part through BK channels, is activated by CICR at membrane voltages approaching the threshold for AP initiation and that this current opposed depolarizing current ramps applied to guinea-pig sympathetic stellate neurones.
Collapse
Affiliation(s)
- Sarah A Locknar
- Department of Anatomy and Neurobiology, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | | | |
Collapse
|
17
|
Collins A, Larson M. Regulation of inward rectifier K+ channels by shift of intracellular pH dependence. J Cell Physiol 2004; 202:76-86. [PMID: 15389543 DOI: 10.1002/jcp.20093] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The mechanistic link between mitochondrial metabolism and inward rectifier K+ channel activity was investigated by studying the effects of a mitochondrial inhibitor, carbonyl cyanide p-trifluoromethoxyphenylhydrazone (FCCP) on inward rectifiers of the Kir2 subfamily expressed in Xenopus oocytes, using two-electrode voltage-clamp, patch-clamp, and intracellular pH recording. FCCP inhibited Kir2.2 and Kir2.3 currents and decreased intracellular pH, but the pH change was too small to account for the inhibitory effect by itself. However, pre-incubation of oocytes with imidazole prevented both the pH decrease and the inhibition of Kir2.2 and Kir2.3 currents by FCCP. The pH dependence of Kir2.2 was shifted to higher pH in membrane patches from FCCP-treated oocytes compared to control oocytes. Therefore, the inhibition of Kir2.2 by FCCP may involve a combination of intracellular acidification and a shift in the intracellular pH dependence of these channels. To investigate the sensitivity of heteromeric channels to FCCP, we studied its effect on currents expressed by heteromeric tandem dimer constructs. While Kir2.1 homomeric channels were insensitive to FCCP, both Kir2.1-Kir2.2 and Kir2.1-Kir2.3 heterotetrameric channels were inhibited. These data support the notion that mitochondrial dysfunction causes inhibition of heteromeric inward rectifier K+ channels. The reduction of inward rectifier K+ channel activity observed in heart failure and ischemia may result from the mitochondrial dysfunction that occurs in these conditions.
Collapse
Affiliation(s)
- Anthony Collins
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon 97331-3507, USA.
| | | |
Collapse
|
18
|
Sculptoreanu A, de Groat WC. Protein kinase C is involved in neurokinin receptor modulation of N- and L-type Ca2+ channels in DRG neurons of the adult rat. J Neurophysiol 2003; 90:21-31. [PMID: 12660348 DOI: 10.1152/jn.00108.2003] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Whole cell patch-clamp techniques were used to examine neurokinin receptor modulation of Ca2+ channels in small to medium size dorsal root ganglia neurons (<40 pF) that express mainly N- and L-type Ca2+ currents. Low concentrations of substance P enhanced Ca2+ currents (5-40%, <0.2 microM), while higher concentrations applied cumulatively reversed these enhancements (5-28% reductions, >0.5 microM). This apparent inhibition by high concentrations of substance P was blocked by the administration of the NK3 antagonist SB 235,375 (0.2 microM). The NK1 agonist, [Sar9,Met11]-substance P (0.05 to 1.0 microM) did not alter Ca2+ currents; whereas the NK2 agonist, [betaAla8]-neurokinin A (4-10), enhanced Ca2+ currents (5-36% increase, 0.05-0.5 microM). The enhancement was reversed by the NK2 antagonist MEN 10,376 (0.2 microM) but unaffected by the NK3 antagonist SB 235,375 (0.2 microM). The NK3 agonist [MePhe7]-neurokinin B (0.5-1.0 microM) inhibited Ca2+ currents (6-24% decrease). This inhibition was not prevented by the NK2 antagonist MEN 10,376 (0.2 microM) but was blocked by the NK3 antagonist SB 235,375 (0.2 microM). Both the enhancement and inhibition of Ca2+ currents by neurokinin agonists were reversed by the protein kinase C inhibitor bisindolylmaleimide I HCl (0.2-0.5 microM). Following inhibition of Ca2+ channels by [MePhe7]-neurokinin the facilitatory effect of BayK 8644 (5 microM) was increased and the inhibitory effect of the N-type Ca2+ channel blocker w -conotoxin GVIA (1 microM) was diminished, suggesting that the NK3 agonist inhibits N-type Ca2+ channels. Similarly, block of all but N-type Ca2+ channels, revealed that [betaAla8]-neurokinin A (4-10) enhanced the currents while [MePhe7]-neurokinin B inhibited the currents. Inhibition of all but L-type Ca2+ channels, revealed that [betaAla8]-neurokinin A (4-10) enhanced the currents while [MePhe7]-neurokinin B had no effect. Activation of protein kinase C with low concentrations of phorbol-12,13-dibutyrate enhanced Ca2+ currents, but high concentrations inhibited N- and L-type Ca2+ currents. In summary, these data suggest that in adult rat dorsal root ganglia neurons, NK2 receptors enhance both L- and N-type Ca2+ channels and NK3 receptors inhibit N-type Ca2+ channels and that these effects are mediated by protein kinase C phosphorylation of Ca2+ channels.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- Calcium/physiology
- Calcium Channel Agonists/pharmacology
- Calcium Channel Blockers/pharmacology
- Calcium Channels, L-Type/metabolism
- Calcium Channels, L-Type/physiology
- Calcium Channels, N-Type/metabolism
- Calcium Channels, N-Type/physiology
- Cell Culture Techniques
- Enzyme Inhibitors/pharmacology
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/physiology
- Indoles/pharmacology
- Male
- Maleimides/pharmacology
- Neurotransmitter Agents/metabolism
- Neurotransmitter Agents/pharmacology
- Patch-Clamp Techniques
- Phosphorylation/drug effects
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C/metabolism
- Rats
- Receptors, Neurokinin-1/agonists
- Receptors, Neurokinin-1/metabolism
- Receptors, Neurokinin-2/agonists
- Receptors, Neurokinin-2/antagonists & inhibitors
- Receptors, Neurokinin-2/metabolism
- Receptors, Neurokinin-3/antagonists & inhibitors
- Receptors, Neurokinin-3/metabolism
- Receptors, Tachykinin/drug effects
- Receptors, Tachykinin/metabolism
- Receptors, Tachykinin/physiology
- Substance P/metabolism
- Substance P/pharmacology
Collapse
Affiliation(s)
- Adrian Sculptoreanu
- Department of Pharmacology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | |
Collapse
|
19
|
Miake J, Marbán E, Nuss HB. Functional role of inward rectifier current in heart probed by Kir2.1 overexpression and dominant-negative suppression. J Clin Invest 2003. [DOI: 10.1172/jci200317959] [Citation(s) in RCA: 167] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
20
|
Stanfield PR, Nakajima S, Nakajima Y. Constitutively active and G-protein coupled inward rectifier K+ channels: Kir2.0 and Kir3.0. Rev Physiol Biochem Pharmacol 2002; 145:47-179. [PMID: 12224528 DOI: 10.1007/bfb0116431] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Peter R Stanfield
- Molecular Physiology Group, Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | | | | |
Collapse
|
21
|
Dewson G, Conley EC, Bradding P. Multigene family isoform profiling from blood cell lineages. BMC Genomics 2002; 3:22. [PMID: 12167175 PMCID: PMC122081 DOI: 10.1186/1471-2164-3-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2002] [Accepted: 08/07/2002] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Analysis of cell-selective gene expression for families of proteins of therapeutic interest is crucial when deducing the influence of genes upon complex traits and disease susceptibility. Presently, there is no convenient tool for examining isoform-selective expression for large gene families. A multigene isoform profiling strategy was developed and used to investigate the inwardly rectifying K+ (Kir) channel family in human leukocytes. Comprised of seven subfamilies, Kir channels have important roles in setting the resting membrane potential in excitable and non-excitable cells. RESULTS Gene sequence alignment allowed determination of "islands" of amino acid homology, and sub-family "centred" priming permitted simultaneous co-amplification of each family member. Validation and cross-priming analysis was performed against a panel of cognate Kir channel clones. Radiolabelling and diagnostic restriction digestion of pooled PCR products enabled determination of distinct Kir gene expression profiles in pure populations of human neutrophils, eosinophils and lung mast cells, with conservation of Kir2.0 isoforms amongst the leukocyte subsets. We also identified a Kir2.0 channel product, which may potentially represent a novel family member. CONCLUSIONS We have developed a novel, rapid and flexible strategy for the determination of gene family isoform composition in any cell type with the additional capacity to detect hitherto unidentified family members and verified its application in a study of Kir channel isoform expression in human leukocytes.
Collapse
Affiliation(s)
- Grant Dewson
- Biochem. Toxicol., CMHT, Leicester University, Lancaster Road, Leicester, LE1 9HN, UK
| | - Edward C Conley
- APBiotech Inc. R&D Unit, Forest Farm Laboratories, Whitchurch, Cardiff, CF14 7YT, UK
| | - Peter Bradding
- Division of Respiratory Medicine, Institute for Lung Health, University of Leicester Medical School, Glenfield Hospital, Leicester, LE3 9QP, UK
| |
Collapse
|
22
|
Schram G, Pourrier M, Melnyk P, Nattel S. Differential distribution of cardiac ion channel expression as a basis for regional specialization in electrical function. Circ Res 2002; 90:939-50. [PMID: 12016259 DOI: 10.1161/01.res.0000018627.89528.6f] [Citation(s) in RCA: 311] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cardiac electrical system is designed to ensure the appropriate rate and timing of contraction in all regions of the heart, which are essential for effective cardiac function. Well-controlled cardiac electrical activity depends on specialized properties of various components of the system, including the sinoatrial node, atria, atrioventricular node, His-Purkinje system, and ventricles. Cardiac electrical specialization was first recognized in the mid 1800s, but over the past 15 years, an enormous amount has been learned about how specialization is achieved by differential expression of cardiac ion channels. More recently, many aspects of the molecular basis have been revealed. Although the field is potentially vast, an appreciation of key elements is essential for any clinician or researcher wishing to understand modern cardiac electrophysiology. This article reviews the major regionally determined features of cardiac electrical function, discusses underlying ionic bases, and summarizes present knowledge of ion channel subunit distribution in relation to functional specialization.
Collapse
Affiliation(s)
- Gernot Schram
- Department of Medicine, University of Montreal, Research Center, Montreal Heart Institute, Quebec, Canada
| | | | | | | |
Collapse
|
23
|
Oonuma H, Iwasawa K, Iida H, Nagata T, Imuta H, Morita Y, Yamamoto K, Nagai R, Omata M, Nakajima T. Inward rectifier K(+) current in human bronchial smooth muscle cells: inhibition with antisense oligonucleotides targeted to Kir2.1 mRNA. Am J Respir Cell Mol Biol 2002; 26:371-9. [PMID: 11867346 DOI: 10.1165/ajrcmb.26.3.4542] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Inward rectifier K(+) (Kir) channels play an important role in forming membrane potential and then modulating muscle tone in certain types of smooth muscles. In cultured human bronchial smooth muscle cells (hBSMCs), Kir current was identified using whole-cell voltage clamp techniques and explored by using RT-PCR analysis of mRNA, Western blotting, and antisense oligonucleotide methods to block the synthesis of Kir channel protein. The K(+) current with strong inward rectification and high K(+) ion selectivity was observed. The current was unaffected by 4-aminopyridine, glibenclamide, and charybdotoxin, and hardly inhibited by tetraethylammonium, but was potently inhibited by extracellular Ba(2+). The IC(50) value of external Ba(2+) was approximately 1.3 microm. RT-PCR analysis of mRNA showed transcripts for Kir2.1, but not Kir1.1, Kir2.2, or Kir2.3. Treatment of cells with antisense oligonucleotides targeted to Kir2.1 resulted in a decrease in the current density of the Kir current and Kir protein expression, as compared with the mismatch-treated cells, whereas the current density of 4-AP-sensitive K(+) currents (K(V)) remained unaffected. The application of Ba(2+) markedly depolarized the membrane. These results demonstrate that Kir channel is present in human bronchial smooth muscle cells, and the Kir2.1 gene encodes the Kir channel protein in these cells.
Collapse
Affiliation(s)
- Hitoshi Oonuma
- Department of Respiratory Medicine, University of Tokyo, Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhang L, Tompkins JD, Hancock JC, Hoover DB. Substance P modulates nicotinic responses of intracardiac neurons to acetylcholine in the guinea pig. Am J Physiol Regul Integr Comp Physiol 2001; 281:R1792-800. [PMID: 11705763 DOI: 10.1152/ajpregu.2001.281.6.r1792] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Application of substance P (SP) to intracardiac neurons of the guinea pig causes slow depolarization and increases neuronal excitability. The present study was done to determine the influence of SP on fast excitatory responses of intracardiac neurons to ACh. Intracellular recording methods were used to measure responses of intracardiac neurons in whole mount preparations of atrial ganglionated nerve plexus from guinea pig hearts. Local pressure ejection of 100 microM SP (1 s) from a glass micropipette caused slow depolarization of all neurons (n = 38) and triggered action potential generation in 47% of the cells tested. Bath application of SP (0.5-100 microM) caused a dose-dependent depolarization of intracardiac neurons but rarely evoked action potentials, even at the highest concentration. However, such treatment with SP enhanced nicotinic responses evoked by local pressure ejections of ACh (10 mM, 10- to 100-ms duration) in 77% of intracardiac neurons studied (n = 52). A significant increase in amplitude of ACh-evoked fast depolarization occurred during treatment with 0.5 microM SP (13.0 +/- 1.8 mV for control vs. 17.7 +/- 1.9 mV with SP present, n = 7, P = 0.019). At higher concentrations of SP, enhancement of the response to ACh resulted mainly in action potential generation. However, responses to ACh were attenuated by SP in 15% of the intracardiac neurons studied. This attenuation occurred primarily during exposure to 10 and 100 microM SP and was manifest as a reduction in amplitude of nicotinic fast depolarization or inhibition of ACh-evoked action potentials. These findings support the conclusion that SP could function as a neuromodulator and neurotransmitter in intracardiac ganglia of the guinea pig.
Collapse
Affiliation(s)
- L Zhang
- Department of Pharmacology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | | | | | | |
Collapse
|
25
|
Nerbonne JM, Nichols CG, Schwarz TL, Escande D. Genetic manipulation of cardiac K(+) channel function in mice: what have we learned, and where do we go from here? Circ Res 2001; 89:944-56. [PMID: 11717150 DOI: 10.1161/hh2301.100349] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the mammalian myocardium, potassium (K(+)) channels control resting potentials, action potential waveforms, automaticity, and refractory periods and, in most cardiac cells, multiple types of K(+) channels that subserve these functions are expressed. Molecular cloning has revealed the presence of a large number of K(+) channel pore forming (alpha) and accessory (beta) subunits in the heart, and considerable progress has been made recently in defining the relationships between expressed K(+) channel subunits and functional cardiac K(+) channels. To date, more than 20 mouse models with altered K(+) channel expression/functioning have been generated using dominant-negative transgenic and targeted gene deletion approaches. In several instances, the genetic manipulation of K(+) channel subunit expression has revealed the role of specific K(+) channel subunit subfamilies or individual K(+) channel subunit genes in the generation of myocardial K(+) channels. In other cases, however, the phenotypic consequences have been unexpected. This review summarizes what has been learned from the in situ genetic manipulation of cardiac K(+) channel functioning in the mouse, discusses the limitations of the models developed to date, and explores the likely directions of future research.
Collapse
Affiliation(s)
- J M Nerbonne
- Department of Molecular Biology, Washington University Medical School, St Louis, MO, USA
| | | | | | | |
Collapse
|
26
|
Jobling P, Messenger JP, Gibbins IL. Differential Expression of Functionally Identified and Immunohistochemically Identified NK1 Receptors on Sympathetic Neurons. J Neurophysiol 2001; 85:1888-98. [PMID: 11353005 DOI: 10.1152/jn.2001.85.5.1888] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have used multiple-labeling immunohistochemistry, intracellular dye-filling, and intracellular microelectrode recordings to characterize the distribution of tachykinin receptors and substance P boutons on subpopulations of neurons within the guinea pig celiac ganglion. Superfusion of substance P (SP, 1 μM for 1 min) depolarized 42% of tonic neurons and inhibited afterhyperpolarizations in 66% of long afterhyperpolarizing (LAH) neurons without significant desensitization. Twenty-one percent of tonic neurons and 24% of LAH neurons responded to the NK3 agonist senktide but did not respond to SP, indicating SP did not activate NK3 receptors at this concentration. All effects of SP were abolished by the selective NK1 receptor antagonist, SR140333, but not by the selective NK3 receptor antagonist, SR142801, suggesting that exogenous SP activated a receptor with NK1 pharmacology. No dye-filled LAH neuron and only 50% of tonic neurons responding to SP expressed NK1 receptor immunoreactivity (NK1-IR). All neurons responding to SP had SP immunoreactive fibers within one cell diameter, indicating good spatial matching between SP release sites and target neurons. These results indicate that SP may act via a receptor with NK1-like pharmacology that has a C terminus not recognized by antibodies to the intracellular domain of the conventional NK1 receptor. Inward currents evoked by SP acting on this NK1-like receptor or senktide acting through NK3 receptors had identical current-voltage relationships. In LAH neurons, both agonists suppressed I sAHP without reducing I AHP. Responses evoked by SP and senktide were resistant to PKC inhibitors, suggesting that the transduction mechanisms for the NK1-like receptor and the NK3 receptor may be similar.
Collapse
Affiliation(s)
- P Jobling
- Department of Anatomy and Histology and Centre for Neuroscience, Flinders University of South Australia, Adelaide, SA 5001, Australia.
| | | | | |
Collapse
|
27
|
Abdulla FA, Stebbing MJ, Smith PA. Effects of substance P on excitability and ionic currents of normal and axotomized rat dorsal root ganglion neurons. Eur J Neurosci 2001; 13:545-52. [PMID: 11168562 DOI: 10.1046/j.0953-816x.2000.01429.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Substance P (SP) may act within dorsal root ganglia (DRG) to modulate the transmission of nociceptive information. Because peripheral nerve injury (axotomy) alters the peptide content of sensory neurons, we used whole-cell recording to examine the effects of sciatic nerve section on the sensitivity of rat lumbar DRG neurons to SP (0.3--1 microM). At 1 microM, SP increased the excitability of 'small', putative nociceptive neurons but had little effect on the excitability of 'large' neurons. Two-four weeks after sciatic nerve section, however, the effect of SP on 'large' axotomized neurons was increased and its effect on 'small' neurons was decreased. SP did not affect Ca(2+) channel currents in control or axotomized neurons. The effects of SP on the current-voltage (I--V) relationship of 77% of neurons involved increased inward current at potentials below -30 mV and suppressed outward current at potentials above -20 mV. The effects of SP on the I--V relationship were similar in control and in axotomized neurons and the altered sensitivity of 'small' and 'large' cells could not be attributed to axotomy-induced changes in input resistance or membrane potential. The possible relevance of alterations in sensitivity, of 'large' DRG neurons to SP, to the generation of neuropathic pain is discussed.
Collapse
Affiliation(s)
- F A Abdulla
- Department of Physical Therapy, School of Allied Health Professions, Tennessee State University, Nashville, Tennessee 37209, USA
| | | | | |
Collapse
|
28
|
Cuevas J, Adams DJ. Substance P preferentially inhibits large conductance nicotinic ACh receptor channels in rat intracardiac ganglion neurons. J Neurophysiol 2000; 84:1961-70. [PMID: 11024089 DOI: 10.1152/jn.2000.84.4.1961] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effects of substance P (SP) on nicotinic acetylcholine (ACh)-evoked currents were investigated in parasympathetic neurons dissociated from neonatal rat intracardiac ganglia using standard whole cell, perforated patch, and outside-out recording configurations of the patch-clamp technique. Focal application of SP onto the soma reversibly decreased the peak amplitude of the ACh-evoked current with half-maximal inhibition occurring at 45 microM and complete block at 300 microM SP. Whole cell current-voltage (I-V) relationships obtained in the absence and presence of SP indicate that the block of ACh-evoked currents by SP is voltage independent. The rate of decay of ACh-evoked currents was increased sixfold in the presence of SP (100 microM), suggesting that SP may increase the rate of receptor desensitization. SP-induced inhibition of ACh-evoked currents was observed following cell dialysis and in the presence of either 1 mM 8-Br-cAMP, a membrane-permeant cAMP analogue, 5 microM H-7, a protein kinase C inhibitor, or 2 mM intracellular AMP-PNP, a nonhydrolyzable ATP analogue. These data suggest that a diffusible cytosolic second messenger is unlikely to mediate SP inhibition of neuronal nicotinic ACh receptor (nAChR) channels. Activation of nAChR channels in outside-out membrane patches by either ACh (3 microM) or cytisine (3 microM) indicates the presence of at least three distinct conductances (20, 35, and 47 pS) in rat intracardiac neurons. In the presence of 3 microM SP, the large conductance nAChR channels are preferentially inhibited. The open probabilities of the large conductance classes activated by either ACh or cytisine were reversibly decreased by 10- to 30-fold in the presence of SP. The single-channel conductances were unchanged, and mean apparent channel open times for the large conductance nAChR channels only were slightly decreased by SP. Given that individual parasympathetic neurons of rat intracardiac ganglia express a heterogeneous population of nAChR subunits represented by the different conductance levels, SP appears to preferentially inhibit those combinations of nAChR subunits that form the large conductance nAChR channels. Since ACh is the principal neurotransmitter of extrinsic (vagal) innervation of the mammalian heart, SP may play an important role in modulating autonomic control of the heart.
Collapse
Affiliation(s)
- J Cuevas
- Department of Physiology and Pharmacology, University of Queensland, Brisbane, QLD 4072, Australia
| | | |
Collapse
|
29
|
Hogan QH, McCallum JB, Sarantopoulos C, Aason M, Mynlieff M, Kwok WM, Bosnjak ZJ. Painful neuropathy decreases membrane calcium current in mammalian primary afferent neurons. Pain 2000; 86:43-53. [PMID: 10779659 DOI: 10.1016/s0304-3959(99)00313-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Hyperexcitability of the primary afferent neuron leads to neuropathic pain following injury to peripheral axons. Changes in calcium channel function of sensory neurons following injury have not been directly examined at the channel level, even though calcium is a primary second messenger-regulating neuronal function. We compared calcium currents (I(Ca)) in 101 acutely isolated dorsal root ganglion neurons from 31 rats with neuropathic pain following chronic constriction injury (CCI) of the sciatic nerve, to cells from 25 rats with normal sensory function following sham surgery. Cells projecting to the sciatic nerve were identified with a fluorescent label applied at the CCI site. Membrane function was determined using patch-clamp techniques in current clamp mode, and in voltage-clamp mode using solutions and conditions designed to isolate I(Ca). Somata of peripheral sensory neurons from hyperalgesic rats demonstrated decreased I(Ca). Peak calcium channel current density was diminished by injury from 3.06+/-0.30 pS/pF to 2. 22+/-0.26 pS/pF in medium neurons, and from 3.93+/-0.38 pS/pF to 2. 99+/-0.40 pS/pF in large neurons. Under these voltage and pharmacologic conditions, medium-sized neuropathic cells lacked obvious T-type calcium currents which were present in 25% of medium-sized cells from control animals. Altered Ca(2+) signalling in injured sensory neurons may contribute to hyperexcitability leading to neuropathic pain.
Collapse
Affiliation(s)
- Q H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee 53226, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Levite M, Cahalon L, Peretz A, Hershkoviz R, Sobko A, Ariel A, Desai R, Attali B, Lider O. Extracellular K(+) and opening of voltage-gated potassium channels activate T cell integrin function: physical and functional association between Kv1.3 channels and beta1 integrins. J Exp Med 2000; 191:1167-76. [PMID: 10748234 PMCID: PMC2193178 DOI: 10.1084/jem.191.7.1167] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Elevated extracellular K(+) ([K(+)](o)), in the absence of "classical" immunological stimulatory signals, was found to itself be a sufficient stimulus to activate T cell beta1 integrin moieties, and to induce integrin-mediated adhesion and migration. Gating of T cell voltage-gated K(+) channels (Kv1.3) appears to be the crucial "decision-making" step, through which various physiological factors, including elevated [K(+)](o) levels, affect the T cell beta1 integrin function: opening of the channel leads to function, whereas its blockage prevents it. In support of this notion, we found that the proadhesive effects of the chemokine macrophage-inflammatory protein 1beta, the neuropeptide calcitonin gene-related peptide (CGRP), as well as elevated [K(+)](o) levels, are blocked by specific Kv1.3 channel blockers, and that the unique physiological ability of substance P to inhibit T cell adhesion correlates with Kv1.3 inhibition. Interestingly, the Kv1.3 channels and the beta1 integrins coimmunoprecipitate, suggesting that their physical association underlies their functional cooperation on the T cell surface. This study shows that T cells can be activated and driven to integrin function by a pathway that does not involve any of its specific receptors (i.e., by elevated [K(+)](o)). In addition, our results suggest that undesired T cell integrin function in a series of pathological conditions can be arrested by molecules that block the Kv1.3 channels.
Collapse
Affiliation(s)
- M Levite
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Off-axis illumination elicits lateral inhibition at the primary visual synapse in crustacea and insects. The evidence suggests that the inhibitory action is presynaptic (i.e., on the photoreceptor terminal) and that the amacrine neurons of the lamina ganglionaris (the first synaptic layer) may be part of the inhibitory pathway. The neurotransmitters and the synaptic mechanisms are unknown. We show by immunocytochemistry that GABA and a tachykinin-related peptide (TRP) are localized in the amacrine neurons of the crayfish lamina ganglionaris. Indirect evidence suggests that GABA and TRP may be colocalized in these neurons. The extensive processes of the amacrine neurons occupy lamina layers containing the terminals of photoreceptors. Application of exogenous GABA and TRP to photoreceptor terminals produces a short-latency, dose-dependent hyperpolarization with a decay time constant on the order of a few seconds. TRP also exhibits actions that evolve over several minutes. These include a reduction of the receptor potential (and the light-elicited current) by approximately 40% and potentiation of the action of GABA by approximately 100%. The mechanisms of TRP action in crayfish are not known, but a plausible pathway is a TRP-dependent elevation of intracellular Ca(2+) that reduces photoreceptor sensitivity in arthropods. Although the mechanisms are not established, the results indicate that in crayfish photoreceptors TRP displays actions on two time scales and can exert profound modulatory control over cell function.
Collapse
|
32
|
Ptak K, Konrad M, Di Pasquale E, Tell F, Hilaire G, Monteau R. Cellular and synaptic effect of substance P on neonatal phrenic motoneurons. Eur J Neurosci 2000; 12:126-38. [PMID: 10651867 DOI: 10.1046/j.1460-9568.2000.00886.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Experiments were carried out on the in vitro brainstem-spinal cord preparation of the newborn rat to analyse the effects of substance P (SP) on phrenic motoneuron (PMN) activity. In current-clamp mode, SP significantly depolarized PMNs, increased their input resistance, decreased the rheobase current and shifted the firing frequency-intensity relationships leftwards, but did not affect spike frequency adaptation or single spike configuration. The neurokinin receptor agonist NK1 had SP-mimetic effects, whereas the NK3 and NK2 receptor agonists were less effective and ineffective, respectively. In a tetrodotoxin-containing aCSF, only SP or the NK1 receptor agonist were still active. No depolarization was observed when the NK1 receptor agonist was applied in the presence of muscarine. In voltage-clamp mode, SP or the NK1 receptor agonist produced an inward current (ISP) which was not significantly reduced by extracellular application of tetraethylammonium, Co2+, 4-aminopyridine or Cs+. In aCSF containing tetrodotoxin, Co2+ and Cs+, ISP was blocked by muscarine. No PMN displayed any M-type potassium current but only a current showing no voltage sensitivity over the range -100 to 0 mV, reversing near the expected EK +, hence consistent with a leak current. SP application to the spinal cord only (using a partitioned chamber) significantly increased the phrenic activity. Pretreatment with the NMDA receptor antagonist DL-2-amino-5-phosphonovaleric acid (AP5) decreased the C4 discharge duration and blocked the effect of SP, thus exhibiting an NMDA potentiation by SP. In conclusion, SP modulates postsynaptically the response of phrenic motoneurons to the inspiratory drive through the reduction of a leak conductance and the potentiation of the NMDA component of the synaptic input.
Collapse
Affiliation(s)
- K Ptak
- ESA CNRS 6034, Faculté des Sciences de St Jérôme, 13397 Marseille cedex 20, France
| | | | | | | | | | | |
Collapse
|
33
|
Nakamura TY, Lee K, Artman M, Rudy B, Coetzee WA. The role of Kir2.1 in the genesis of native cardiac inward-rectifier K+ currents during pre- and postnatal development. Ann N Y Acad Sci 1999; 868:434-7. [PMID: 10414316 DOI: 10.1111/j.1749-6632.1999.tb11308.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Our results demonstrate that (a) the Kir2.1 gene encodes a native K+ channel protein with a 21-pS conductance; (b) this channel has an important role in the genesis of adult ventricular 1K1; and (c) the contribution of Kir2.1 channel proteins to 1K1 changes during development. The lack of contribution of Kir2.1 to fetal 1K1 channels is interesting from the point of view of possible future generation of knockout mice lacking Kir2.1, since cardiac abnormalities would not be expected to result in fetal lethality. These observations provide further support for a generalized hypothesis that different genes may code for 1K1 channel proteins at various developmental stages. However, the effects of these AS-oligos must first be examined on native 1K1 channels in cardiac myocytes before definite conclusions can be reached.
Collapse
Affiliation(s)
- T Y Nakamura
- Department of Pediatrics, New York University Medical Center, New York 10016, USA
| | | | | | | | | |
Collapse
|
34
|
Coetzee WA, Amarillo Y, Chiu J, Chow A, Lau D, McCormack T, Moreno H, Nadal MS, Ozaita A, Pountney D, Saganich M, Vega-Saenz de Miera E, Rudy B. Molecular diversity of K+ channels. Ann N Y Acad Sci 1999; 868:233-85. [PMID: 10414301 DOI: 10.1111/j.1749-6632.1999.tb11293.x] [Citation(s) in RCA: 877] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
K+ channel principal subunits are by far the largest and most diverse of the ion channels. This diversity originates partly from the large number of genes coding for K+ channel principal subunits, but also from other processes such as alternative splicing, generating multiple mRNA transcripts from a single gene, heteromeric assembly of different principal subunits, as well as possible RNA editing and posttranslational modifications. In this chapter, we attempt to give an overview (mostly in tabular format) of the different genes coding for K+ channel principal and accessory subunits and their genealogical relationships. We discuss the possible correlation of different principal subunits with native K+ channels, the biophysical and pharmacological properties of channels formed when principal subunits are expressed in heterologous expression systems, and their patterns of tissue expression. In addition, we devote a section to describing how diversity of K+ channels can be conferred by heteromultimer formation, accessory subunits, alternative splicing, RNA editing and posttranslational modifications. We trust that this collection of facts will be of use to those attempting to compare the properties of new subunits to the properties of others already known or to those interested in a comparison between native channels and cloned candidates.
Collapse
Affiliation(s)
- W A Coetzee
- Department of Physiology, New York University School of Medicine, New York 10016, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yamada T, Endoh T, Suzuki T. Inhibition of calcium channels by neurokinin receptor and signal transduction in hamster submandibular ganglion cells. JOURNAL OF THE AUTONOMIC NERVOUS SYSTEM 1999; 76:1-8. [PMID: 10323301 DOI: 10.1016/s0165-1838(98)00183-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Both substance P (SP) and neurokinin A (NKA) are known as neurotransmitters of the submandibular ganglion (SMG) neurons. SP released from collaterals of the sensory nerves also regulates the excitability of SMG neurons. It has recently been shown that neurokinins (NK) inhibit calcium channels in various neurons. In this study, the effects of NK on voltage-dependent calcium channel current (I(Ca)) in SMG cells were investigated using the whole-cell patch-clamp recording method. NK-1 receptor agonist and SP caused inhibition of I(Ca) in SMG cells in a dose-dependent manner. NK-1 receptor agonist inhibited L-, N- and P/Q-type I(Ca) components. GDP-beta-S included in the pipette solution reduced the NK-1 receptor agonist-induced inhibition of I(Ca). In addition, NK-1 receptor agonist-induced inhibition of I(Ca) was reduced by stimulation of protein kinase C (PKC) but not cyclic AMP-dependent protein kinase (PKA). The results provided evidence for a signal transduction pathway in which calcium channel inhibition by NK receptors required activation of G-protein and PKC-affected step phosphorylation in SMG neurons.
Collapse
Affiliation(s)
- T Yamada
- Department of Physiology, Tokyo Dental College, Chiba, Japan
| | | | | |
Collapse
|
36
|
Abstract
Nicotine is the main constituent of tobacco smoke responsible for the elevated risk of the cardiovascular disease and sudden coronary death associated with smoking, presumably by provoking cardiac arrhythmias. The cellular mechanisms may be related to the ability of nicotine to prolong action potentials and to depolarize membrane potential. However, the underlying ionic mechanisms remained unknown. We showed here that nicotine blocked multiple types of K+ currents, including the native currents in canine ventricular myocytes and the cloned channels expressed in Xenopus oocytes: A-type K+ currents (I(to)/Kv4.3), delayed rectifier K+ currents (I(Kr)/HERG) and inward rectifier K+ currents (I(K1)/Kir2.1). Most noticeably, nicotine at a concentration as low as of 10 nM significantly suppressed I(to) and Kv4.3 by approximately 20%. The effects of nicotine were independent of nicotinic receptor simulation or catecholamine release. Our results indicate that nicotine is a non-specific blocker of K+ channels and the inhibitory effects are the consequence of direct interactions between nicotine molecules and the channel proteins. Our study provided for the first time the evidence for the direct inhibition of cardiac K+ channels by nicotine and established a novel aspect of nicotine pharmacology.
Collapse
Affiliation(s)
- H Wang
- Research Center, Montreal Heart Institute, Quebec, Canada
| | | | | |
Collapse
|