1
|
Getsy PM, Coffee GA, Bates JN, Baby SM, Seckler JM, Palmer LA, Lewis SJ. Functional evidence that S-nitroso-L-cysteine may be a candidate carotid body neurotransmitter. Neuropharmacology 2025; 265:110229. [PMID: 39577762 DOI: 10.1016/j.neuropharm.2024.110229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
The primary objective of the present study is to provide further evidence that the endogenous S-nitrosothiol, S-nitroso-L-cysteine (L-CSNO), plays an essential role in signaling the hypoxic ventilatory response (HVR) in rodents. Key findings were that (1) injection of L-CSNO (50 nmol/kg, IV) caused a pronounced increase in frequency of breathing (Freq), tidal volume (TV) and minute ventilation (MV) in naïve C57BL/6 mice, whereas injection of D-CSNO (50 nmol/kg, IV) elicited minimal responses; (2) L-CSNO elicited minor responses in (a) C57BL/6 mice with bilateral carotid sinus nerve transection (CSNX), (b) C57BL/6 mice treated neonatally with capsaicin (CAP) to eliminate small-diameter C-fibers, and (c) C57BL/6 mice receiving continuous infusion of L-CSNO receptor antagonists, S-methyl-L-cysteine and S-ethyl-L-cysteine (L-SMC + L-SEC, both at 5 μmol/kg/min, IV); and (3) injection of S-nitroso-L-glutathione (L-GSNO, 50 nmol/kg, IV) elicited pronounced ventilatory responses that were not inhibited by L-SMC + L-SEC. Subsequent exposure of naïve C57BL/6 mice to a hypoxic gas challenge (HXC; 10% O2, 90% N2) elicited pronounced increases in Freq, TV and MV that were subject to roll-off. These HXC responses were markedly reduced in CSNX, CAP, and L-SMC + L-SEC-infused C57BL/6 mice. Subsequent exposure of all C57BL/6 mice (naïve, CSNX, CAP, and L-SMC + L-SEC) to a hypercapnic gas challenge (5% CO2, 21% O2, 74% N2) elicited similar robust increases in Freq, TV and MV. Taken together, these findings provide evidence that an endogenous factor with pharmacodynamic properties similar to those of L-CSNO, rather than L-GSNO, mediates the HVR in male C57BL/6 mice.
Collapse
Affiliation(s)
- Paulina M Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA.
| | - Gregory A Coffee
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - James N Bates
- Department of Anesthesiology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Santhosh M Baby
- Section of Biology, Galleon Pharmaceuticals, Inc, Horsham, PA, USA
| | - James M Seckler
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Lisa A Palmer
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - Stephen J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA; Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA; Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
2
|
Magliocca G, Esposito E, Tufano M, Piccialli I, Rubino V, Tedeschi V, Sisalli MJ, Carriero F, Ruggiero G, Secondo A, Annunziato L, Scorziello A, Pannaccione A. Involvement of K V3.4 Channel in Parkinson's Disease: A Key Player in the Control of Midbrain and Striatum Differential Vulnerability during Disease Progression? Antioxidants (Basel) 2024; 13:999. [PMID: 39199246 PMCID: PMC11351402 DOI: 10.3390/antiox13080999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024] Open
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disease in the elderly, is characterized by selective loss of dopaminergic neurons and accumulation of α-synuclein (α-syn), mitochondrial dysfunction, Ca2+ dyshomeostasis, and neuroinflammation. Since current treatments for PD merely address symptoms, there is an urgent need to identify the PD pathophysiological mechanisms to develop better therapies. Increasing evidence has identified KV3.4, a ROS-sensitive KV channel carrying fast-inactivating currents, as a potential therapeutic target against neurodegeneration. In fact, it has been hypothesized that KV3.4 channels could play a role in PD etiopathogenesis, controlling astrocytic activation and detrimental pathways in A53T mice, a well-known model of familial PD. Here, we showed that the A53T midbrain, primarily involved in the initial phase of PD pathogenesis, displayed an early upregulation of the KV3.4 channel at 4 months, followed by its reduction at 12 months, compared with age-matched WT. On the other hand, in the A53T striatum, the expression of KV3.4 remained high at 12 months, decreasing thereafter, in 16-month-old mice. The proteomic profile highlighted a different detrimental phenotype in A53T brain areas. In fact, the A53T striatum and midbrain differently expressed neuroprotective/detrimental pathways, with the variation of astrocytic p27kip1, XIAP, and Smac/DIABLO expression. Of note, a switch from protective to detrimental phenotype was characterized by the upregulation of Smac/DIABLO and downregulation of p27kip1 and XIAP. This occurred earlier in the A53T midbrain, at 12 months, compared with the striatum proteomic profile. In accordance, an upregulation of Smac/DIABLO and a downregulation of p27kip1 occurred in the A53T striatum only at 16 months, showing the slowest involvement of this brain area. Of interest, HIF-1α overexpression was associated with the detrimental profile in midbrain and its major vulnerability. At the cellular level, patch-clamp recordings revealed that primary A53T striatum astrocytes showed hyperpolarized resting membrane potentials and lower firing frequency associated with KV3.4 ROS-dependent hyperactivity, whereas primary A53T midbrain astrocytes displayed a depolarized resting membrane potential accompanied by a slight increase of KV3.4 currents. Accordingly, intracellular Ca2+ homeostasis was significantly altered in A53T midbrain astrocytes, in which the ER Ca2+ level was lower than in A53T striatum astrocytes and the respective littermate controls. Collectively, these results suggest that the early KV3.4 overexpression and ROS-dependent hyperactivation in astrocytes could take part in the different vulnerabilities of midbrain and striatum, highlighting astrocytic KV3.4 as a possible new therapeutic target in PD.
Collapse
Affiliation(s)
- Giorgia Magliocca
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy; (G.M.); (E.E.); (M.T.); (I.P.); (V.T.); (A.S.)
| | - Emilia Esposito
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy; (G.M.); (E.E.); (M.T.); (I.P.); (V.T.); (A.S.)
| | - Michele Tufano
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy; (G.M.); (E.E.); (M.T.); (I.P.); (V.T.); (A.S.)
| | - Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy; (G.M.); (E.E.); (M.T.); (I.P.); (V.T.); (A.S.)
| | - Valentina Rubino
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (V.R.); (M.J.S.); (G.R.)
| | - Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy; (G.M.); (E.E.); (M.T.); (I.P.); (V.T.); (A.S.)
| | - Maria Jose Sisalli
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (V.R.); (M.J.S.); (G.R.)
| | - Flavia Carriero
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy;
| | - Giuseppina Ruggiero
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (V.R.); (M.J.S.); (G.R.)
| | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy; (G.M.); (E.E.); (M.T.); (I.P.); (V.T.); (A.S.)
| | | | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy; (G.M.); (E.E.); (M.T.); (I.P.); (V.T.); (A.S.)
| | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini, 5, 80131 Naples, Italy; (G.M.); (E.E.); (M.T.); (I.P.); (V.T.); (A.S.)
| |
Collapse
|
3
|
Holmes AP, Ray CJ, Coney AM, Kumar P. Is Carotid Body Physiological O 2 Sensitivity Determined by a Unique Mitochondrial Phenotype? Front Physiol 2018; 9:562. [PMID: 29867584 PMCID: PMC5964187 DOI: 10.3389/fphys.2018.00562] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
The mammalian carotid body (CB) is the primary arterial chemoreceptor that responds to acute hypoxia, initiating systemic protective reflex responses that act to maintain O2 delivery to the brain and vital organs. The CB is unique in that it is stimulated at O2 levels above those that begin to impact on the metabolism of most other cell types. Whilst a large proportion of the CB chemotransduction cascade is well defined, the identity of the O2 sensor remains highly controversial. This short review evaluates whether the mitochondria can adequately function as acute O2 sensors in the CB. We consider the similarities between mitochondrial poisons and hypoxic stimuli in their ability to activate the CB chemotransduction cascade and initiate rapid cardiorespiratory reflexes. We evaluate whether the mitochondria are required for the CB to respond to hypoxia. We also discuss if the CB mitochondria are different to those located in other non-O2 sensitive cells, and what might cause them to have an unusually low O2 binding affinity. In particular we look at the potential roles of competitive inhibitors of mitochondrial complex IV such as nitric oxide in establishing mitochondrial and CB O2-sensitivity. Finally, we discuss novel signaling mechanisms proposed to take place within and downstream of mitochondria that link mitochondrial metabolism with cellular depolarization.
Collapse
Affiliation(s)
| | | | | | - Prem Kumar
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
4
|
Gattuso A, Garofalo F, Cerra MC, Imbrogno S. Hypoxia Tolerance in Teleosts: Implications of Cardiac Nitrosative Signals. Front Physiol 2018; 9:366. [PMID: 29706897 PMCID: PMC5906588 DOI: 10.3389/fphys.2018.00366] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/26/2018] [Indexed: 12/18/2022] Open
Abstract
Changes in environmental oxygen (O2) are naturally occurring phenomena which ectotherms have to face on. Many species exhibit a striking capacity to survive and remain active for long periods under hypoxia, even tolerating anoxia. Some fundamental adaptations contribute to this capacity: metabolic suppression, tolerance of pH and ionic unbalance, avoidance and/or repair of free-radical-induced cell injury during reoxygenation. A remarkable feature of these species is their ability to preserve a normal cardiovascular performance during hypoxia/anoxia to match peripheral (tissue pO2) requirements. In this review, we will refer to paradigms of hypoxia- and anoxia-tolerant teleost fish to illustrate cardiac physiological strategies that, by involving nitric oxide and its metabolites, play a critical role in the adaptive responses to O2 limitation. The information here reported may contribute to clarify the molecular and cellular mechanisms underlying heart vulnerability vs. resistance in relation to O2 availability.
Collapse
Affiliation(s)
- Alfonsina Gattuso
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Filippo Garofalo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Maria C Cerra
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Sandra Imbrogno
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
5
|
Kang D, Wang J, Hogan JO, Kim D. TASK-1 (K2P3) and TASK-3 (K2P9) in Rabbit Carotid Body Glomus Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1071:35-41. [DOI: 10.1007/978-3-319-91137-3_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
6
|
Wang J, Kim D. Activation of voltage-dependent K + channels strongly limits hypoxia-induced elevation of [Ca 2+ ] i in rat carotid body glomus cells. J Physiol 2017; 596:3119-3136. [PMID: 29160573 DOI: 10.1113/jp275275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/10/2017] [Indexed: 01/01/2023] Open
Abstract
KEY POINTS We studied the role of the large-conductance Ca2+ -activated K+ channel (BK) and voltage-dependent K+ channels (Kv) on [Ca2+ ]i responses to a wide range of hypoxia at different resting cell membrane potential (Em ). BK/Kv were mostly closed at rest in normoxia. BK/Kv became basally active when cells were depolarized by elevated [KCl]o (>12 mm). Regardless of whether BK/Kv were closed or basally open, hypoxia-induced elevation of [Ca2+ ]i was enhanced 2- to 3-fold by inhibitors of BK/Kv. Hypoxia-induced elevation of [Ca2+ ]i was enhanced ∼2-fold by an inhibitor of Kv2, a major Kv in rat glomus cells. Hypoxia did not inhibit BK in inside-out patches. Our study supports a scheme in which activation of BK/Kv strongly limits the magnitude of hypoxia-induced [Ca2+ ]i rise, with Kv having a much greater effect than BK. ABSTRACT Large-conductance KCa (BK) and other voltage-dependent K+ channels (Kv) are highly expressed in carotid body (CB) glomus cells, but their role in hypoxia-induced excitation is still not well defined and remains controversial. We addressed this issue by studying the effects of inhibitors of BK (IBTX) and BK/Kv (TEA/4-AP) on [Ca2+ ]i responses to a wide range of hypoxia at different levels of resting cell membrane potential (Em ). IBTX and TEA/4-AP did not affect the basal [Ca2+ ]i in isolated glomus cells bathed in 5 mm KClo , but elicited transient increases in [Ca2+ ]i in cells that were moderately depolarized (11-20 mV) by elevation of [KCl]o (12-20 mm). Thus, BK and Kv were mostly closed at rest and activated by depolarization. Four different levels of hypoxia (mild, moderate, severe, anoxia) were used to produce a wide range of [Ca2+ ]i elevation (0-700 nm). IBTX did not affect the rise in [Ca2+ ]i , but TEA/4-AP strongly (∼3-fold) enhanced [Ca2+ ]i rise by moderate and severe levels of hypoxia. Guangxitoxin, a Kv2 blocker, inhibited the whole-cell current by ∼50%, and enhanced 2-fold the [Ca2+ ]i rise elicited by moderate and severe levels of hypoxia. Anoxia did not directly affect BK, but activated BK via depolarization. Our findings do not support the view that hypoxia inhibits BK/Kv to initiate or maintain the hypoxic response. Rather, our results show that BK/Kv are activated as glomus cells depolarize in response to hypoxia, which then limits the rise in [Ca2+ ]i . Inhibition of Kv may provide a mechanism to enhance the chemosensory activity of the CB and ventilation.
Collapse
Affiliation(s)
- Jiaju Wang
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Donghee Kim
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
7
|
Zachar PC, Pan W, Jonz MG. Characterization of ion channels and O 2 sensitivity in gill neuroepithelial cells of the anoxia-tolerant goldfish ( Carassius auratus). J Neurophysiol 2017; 118:3014-3023. [PMID: 28904098 DOI: 10.1152/jn.00237.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 09/07/2017] [Accepted: 09/07/2017] [Indexed: 01/10/2023] Open
Abstract
The neuroepithelial cell (NEC) of the fish gill is an important model for O2 sensing in vertebrates; however, a complete picture of the chemosensory mechanisms in NECs is lacking, and O2 chemoreception in vertebrates that are tolerant to anoxia has not yet been explored. Using whole cell patch-clamp recording, we characterized four types of ion channels in NECs isolated from the anoxia-tolerant goldfish. A Ca2+-dependent K+ current (IKCa) peaked at ~20 mV, was potentiated by increased intracellular Ca2+, and was reduced by 100 μM Cd2+ A voltage-dependent inward current in Ba2+ solution, with peak at 0 mV, confirmed the presence of Ca2+ channels. A voltage-dependent K+ current (IKV) was inhibited by 20 mM tetraethylammonium and 5 mM 4-aminopyridine, revealing a background K+ current (IKB) with open rectification. Mean resting membrane potential of -45.2 ± 11.6 mV did not change upon administration of hypoxia (Po2 = 11 mmHg), nor were any of the K+ currents sensitive to changes in Po2 during whole cell recording. By contrast, when the membrane and cytosol were left undisturbed during fura-2 or FM 1-43 imaging experiments, hypoxia increased intracellular Ca2+ concentration and initiated synaptic vesicle activity. 100 μM Cd2+ and 50 μM nifedipine eliminated uptake of FM 1-43. We conclude that Ca2+ influx via L-type Ca2+ channels is correlated with vesicular activity during hypoxic stimulation. In addition, we suggest that expression of IKCa in gill NECs is species specific and, in goldfish, may contribute to an attenuated response to acute hypoxia.NEW & NOTEWORTHY This study provides the first physiological characterization of oxygen chemoreceptors from an anoxia-tolerant vertebrate. Neuroepithelial cells (NECs) from the gills of goldfish displayed L-type Ca2+ channels and three types of K+ channels, one of which was dependent upon intracellular Ca2+ Although membrane currents were not inhibited by hypoxia during patch-clamp recording, this study is the first to show that NECs with an undisturbed cytosol responded to hypoxia with increased intracellular Ca2+ and synaptic vesicle activity.
Collapse
Affiliation(s)
- Peter C Zachar
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Wen Pan
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael G Jonz
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
8
|
Chang AJ. Acute oxygen sensing by the carotid body: from mitochondria to plasma membrane. J Appl Physiol (1985) 2017; 123:1335-1343. [PMID: 28819004 DOI: 10.1152/japplphysiol.00398.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/11/2017] [Accepted: 08/12/2017] [Indexed: 12/12/2022] Open
Abstract
Maintaining oxygen homeostasis is crucial to the survival of animals. Mammals respond acutely to changes in blood oxygen levels by modulating cardiopulmonary function. The major sensor of blood oxygen that regulates breathing is the carotid body (CB), a small chemosensory organ located at the carotid bifurcation. When arterial blood oxygen levels drop in hypoxia, neuroendocrine cells in the CB called glomus cells are activated to signal to afferent nerves that project to the brain stem. The mechanism by which hypoxia stimulates CB sensory activity has been the subject of many studies over the past 90 years. Two discrete models emerged that argue for the seat of oxygen sensing to lie either in the plasma membrane or mitochondria of CB cells. Recent studies are bridging the gap between these models by identifying hypoxic signals generated by changes in mitochondrial function in the CB that can be sensed by plasma membrane proteins on glomus cells. The CB is important for physiological adaptation to hypoxia, and its dysfunction contributes to sympathetic hyperactivity in common conditions such as sleep-disordered breathing, chronic heart failure, and insulin resistance. Understanding the basic mechanism of oxygen sensing in the CB could allow us to develop strategies to target this organ for therapy. In this short review, I will describe two historical models of CB oxygen sensing and new findings that are integrating these models.
Collapse
Affiliation(s)
- Andy J Chang
- Department of Physiology and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| |
Collapse
|
9
|
Álvarez-Miguel I, Cidad P, Pérez-García MT, López-López JR. Differences in TRPC3 and TRPC6 channels assembly in mesenteric vascular smooth muscle cells in essential hypertension. J Physiol 2016; 595:1497-1513. [PMID: 27861908 DOI: 10.1113/jp273327] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/07/2016] [Indexed: 01/29/2023] Open
Abstract
KEY POINTS Canonical transient receptor potential (TRPC)3 and TRPC6 channels of vascular smooth muscle cells (VSMCs) mediate stretch- or agonist-induced cationic fluxes, contributing to membrane potential and vascular tone. Native TRPC3/C6 channels can form homo- or heterotetrameric complexes, which can hinder individual TRPC channel properties. The possibility that the differences in their association pattern may change their contribution to vascular tone in hypertension is unexplored. Functional characterization of heterologously expressed channels showed that TRPC6-containing complexes exhibited Pyr3/Pyr10-sensitive currents, whereas TRPC3-mediated currents were blocked by anti-TRPC3 antibodies. VSMCs from hypertensive (blood pressure high; BPH) mice have larger cationic basal currents insensitive to Pyr10 and sensitive to anti-TRPC3 antibodies. Consistently, myography studies showed a larger Pyr3/10-induced vasodilatation in BPN (blood pressure normal) mesenteric arteries. We conclude that the increased TRPC3 channel expression in BPH VSMCs leads to changes in TRPC3/C6 heteromultimeric assembly, with a higher TRPC3 channel contribution favouring depolarization of hypertensive VSMCs. ABSTRACT Increased vascular tone in essential hypertension involves a sustained rise in total peripheral resistance. A model has been proposed in which the combination of membrane depolarization and higher L-type Ca2+ channel activity generates augmented Ca2+ influx into vascular smooth muscle cells (VSMCs), contraction and vasoconstriction. The search for culprit ion channels responsible for membrane depolarization has provided several candidates, including members of the canonical transient receptor potential (TRPC) family. TRPC3 and TRPC6 are diacylglycerol-activated, non-selective cationic channels contributing to stretch- or agonist-induced depolarization. Conflicting information exists regarding changes in TRPC3/TRPC6 functional expression in hypertension. However, although TRPC3-TRPC6 channels can heteromultimerize, the possibility that differences in their association pattern may change their functional contribution to vascular tone is largely unexplored. We probe this hypothesis using a model of essential hypertension (BPH mice; blood pressure high) and its normotensive control (BPN mice; blood pressure normal). First, non-selective cationic currents through homo- and heterotetramers recorded from transfected Chinese hamster ovary cells indicated that TRPC currents were sensitive to the selective antagonist Pyr10 only when TRPC6 was present, whereas intracellular anti-TRPC3 antibody selectively blocked TRPC3-mediated currents. In mesenteric VSMCs, basal and agonist-induced currents were more sensitive to Pyr3 and Pyr10 in BPN cells. Consistently, myography studies showed a larger Pyr3/10-induced vasodilatation in BPN mesenteric arteries. mRNA and protein expression data supported changes in TRPC3 and TRPC6 proportions and assembly, with a higher TRPC3 channel contribution in BPH VSMCs that could favour cell depolarization. These differences in functional and pharmacological properties of TRPC3 and TRPC6 channels, depending on their assembly, could represent novel therapeutical opportunities.
Collapse
Affiliation(s)
- Inés Álvarez-Miguel
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - M Teresa Pérez-García
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - José Ramón López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| |
Collapse
|
10
|
Zhou T, Chien MS, Kaleem S, Matsunami H. Single cell transcriptome analysis of mouse carotid body glomus cells. J Physiol 2016; 594:4225-51. [PMID: 26940531 DOI: 10.1113/jp271936] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/24/2016] [Indexed: 01/12/2023] Open
Abstract
KEY POINTS Carotid body (CB) glomus cells mediate acute oxygen sensing and the initiation of the hypoxic ventilatory response, yet the gene expression profile of these cells is not available. We demonstrate that the single cell RNA-Seq method is a powerful tool for identifying highly expressed genes in CB glomus cells. Our single cell RNA-Seq results characterized novel CB glomus cell genes, including members of the G protein-coupled receptor signalling pathway, ion channels and atypical mitochondrial electron transport chain subunits. A heterologous cell-based screening identified acetate (which is known to affect CB glomus cell activity) as an agonist for the most highly abundant G protein-coupled receptor (Olfr78) in CB glomus cells. These data established the first transcriptome profile of CB glomus cells, highlighting genes with potential implications in CB chemosensory function. ABSTRACT The carotid body (CB) is a major arterial chemoreceptor containing glomus cells whose activities are regulated by changes in arterial blood content, including oxygen. Despite significant advancements in the characterization of their physiological properties, our understanding of the underlying molecular machinery and signalling pathway in CB glomus cells is still limited. To overcome this, we employed the single cell RNA-Seq method by performing next-generation sequencing on single glomus cell-derived cDNAs to eliminate contamination of genes derived from other cell types present in the CB. Using this method, we identified a set of genes abundantly expressed in glomus cells, which contained novel glomus cell-specific genes. Transcriptome and subsequent in situ hybridization and immunohistochemistry analyses identified abundant G protein-coupled receptor signalling pathway components and various types of ion channels, as well as members of the hypoxia-inducible factors pathway. A short-chain fatty acid olfactory receptor Olfr78, recently implicated in CB function, was the most abundant G protein-coupled receptor. Two atypical mitochondrial electron transport chain subunits (Ndufa4l2 and Cox4i2) were among the most specifically expressed genes in CB glomus cells, highlighting their potential roles in mitochondria-mediated oxygen sensing. The wealth of information provided by the present study offers a valuable foundation for identifying molecules functioning in the CB.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Molecular Genetics and Microbiology, Duke University Medical Centre, Durham, NC, USA
| | - Ming-Shan Chien
- Department of Molecular Genetics and Microbiology, Duke University Medical Centre, Durham, NC, USA
| | - Safa Kaleem
- Department of Molecular Genetics and Microbiology, Duke University Medical Centre, Durham, NC, USA
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology, Duke University Medical Centre, Durham, NC, USA.,Department of Neurobiology and Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| |
Collapse
|
11
|
Role of BK Channels in Murine Carotid Body Neural Responses in vivo. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 860:325-33. [PMID: 26303497 DOI: 10.1007/978-3-319-18440-1_37] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The aim of this study was to explore the role of BK channels in the hypoxic sensitivity of the in vivo murine carotid body (CB). Four strains of mice (DBA/2J, A/J, BKα1 knockout and BKα1 wild type - FVB background) were used. The mice were anesthetized, paralyzed and mechanically ventilated (PaCO(2) ~ 35 mmHg, PO(2) > 300 mmHg). We measured carotid sinus nerve (CSN) activity during three gas challenges (F(I)O(2): 0.21, 0.15 and 0.10). CSN activity was analyzed with time-variant spectral analysis with frequency domain conversion (Fast Fourier Transforms). Afferent CSN activity increased with lowering F(I)O(2) in the DBA/2J, BKKO and BKWT mice with the most robust response in 600-800 frequencies. No substantial changes were observed in the A/J mice. Although maximal neural output was similar between the BKKO and BKWT mice, the BKWT had a higher early response compared to BKKO. Thus, BK channels may play a role in the initial response of the CB to hypoxia. The contribution of BKβ subunits or the importance of frequency specific responses was unable to be determined by the current study.
Collapse
|
12
|
Pandit JJ. Volatile anaesthetic depression of the carotid body chemoreflex-mediated ventilatory response to hypoxia: directions for future research. SCIENTIFICA 2014; 2014:394270. [PMID: 24808974 PMCID: PMC3997855 DOI: 10.1155/2014/394270] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 03/06/2014] [Indexed: 06/03/2023]
Abstract
In assessing whether volatile anaesthetics directly depress the carotid body response to hypoxia it is necessary to combine in meta-analysis studies of when it is "functionally isolated" (e.g., recordings are made from its afferent nerve). Key articles were retrieved (full papers in English) and subjected to quantitative analysis to yield an aggregate estimate of effect. Results from articles that did not use such methodology were assessed separately from this quantitative approach, to see what could be learned also from a nonquantitative overview. Just 7 articles met the inclusion criteria for hypoxia and just 6 articles for hypercapnia. Within these articles, the anaesthetic (mean dose 0.75, standard deviation (SD) 0.40 minimum alveolar concentration, MAC) statistically significantly depressed carotid body hypoxic response by 24% (P = 0.041), but a similar dose (mean 0.81 (0.42) MAC) did not affect the hypercapnic response. The articles not included in the quantitative analysis (31 articles), assessed qualitatively, also indicated that anaesthetics depress carotid body function. This conclusion helps direct future research on the anaesthetic effects on putative cellular/molecular processes that underlie the transduction of hypoxia in the carotid body.
Collapse
Affiliation(s)
- J. J. Pandit
- Nuffield Department of Anaesthetics, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
13
|
Abstract
SIGNIFICANCE Ventilatory responses to hypoxia are initiated by the carotid body, where inhibition of specific K(+) channels causes cell depolarization, voltage-gated Ca(2+) influx, and neurotransmitter release. The identity of the upstream oxygen (O2) sensor is still controversial. RECENT ADVANCES The activity of BKCa channels is regulated by O2, carbon monoxide (CO), and hydrogen sulfide (H2S), suggesting that integration of these signals may be crucial to the physiological response of this tissue. BKCa is colocalized with hemeoxygenase-2, an enzyme that generates CO in the presence of O2, and CO is a BKCa channel opener. Reduced CO during hypoxia results in channel closure, conferring a degree of O2 sensitivity to the BKCa channel. Conversely, H2S is a potent BKCa inhibitor. H2S is produced endogenously by cystathionine-β-synthase and cystathionine-γ-lyase in the rat carotid body, and its intracellular concentration is dependent upon the balance between its enzymatic generation and its mitochondrial breakdown. During hypoxia, mitochondrial oxidation of H2S in many tissues is reduced, leading to hypoxia-evoked rises in its concentration. This may be sufficient to inhibit K(+) channels and lead to carotid body excitation. CRITICAL ISSUES Carotid body function is heavily dependent upon regulated production and breakdown of CO and H2S and integration of signals from these newly emerging gasotransmitters, in combination with several other proposed mechanisms, may refine, or even define, responses of this tissue to hypoxia. FUTURE DIRECTIONS Since several other sensors have been postulated, the challenge of future research is to begin to integrate each in a unifying mechanism, as has been attempted for the first time herein.
Collapse
Affiliation(s)
- Paul J Kemp
- Division of Pathophysiology and Repair, School of Biosciences, Cardiff University , Cardiff, United Kingdom
| | | |
Collapse
|
14
|
Dallas ML, Deuchars SA, Deuchars J. Immunopharmacology: utilizing antibodies as ion channel modulators. Expert Rev Clin Pharmacol 2014; 3:281-9. [DOI: 10.1586/ecp.10.18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
15
|
Ortega-Sáenz P, Pardal R, Levitsky K, Villadiego J, Muñoz-Manchado AB, Durán R, Bonilla-Henao V, Arias-Mayenco I, Sobrino V, Ordóñez A, Oliver M, Toledo-Aral JJ, López-Barneo J. Cellular properties and chemosensory responses of the human carotid body. J Physiol 2013; 591:6157-73. [PMID: 24167224 DOI: 10.1113/jphysiol.2013.263657] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The carotid body (CB) is the major peripheral arterial chemoreceptor in mammals that mediates the acute hyperventilatory response to hypoxia. The CB grows in response to sustained hypoxia and also participates in acclimatisation to chronic hypoxaemia. Knowledge of CB physiology at the cellular level has increased considerably in recent times thanks to studies performed on lower mammals, and rodents in particular. However, the functional characteristics of human CB cells remain practically unknown. Herein, we use tissue slices or enzymatically dispersed cells to determine the characteristics of human CB cells. The adult human CB parenchyma contains clusters of chemosensitive glomus (type I) and sustentacular (type II) cells as well as nestin-positive progenitor cells. This organ also expresses high levels of the dopaminotrophic glial cell line-derived neurotrophic factor (GDNF). We found that GDNF production and the number of progenitor and glomus cells were preserved in the CBs of human subjects of advanced age. Moreover, glomus cells exhibited voltage-dependent Na(+), Ca(2+) and K(+) currents that were qualitatively similar to those reported in lower mammals. These cells responded to hypoxia with an external Ca(2+)-dependent increase of cytosolic Ca(2+) and quantal catecholamine secretion, as reported for other mammalian species. Interestingly, human glomus cells are also responsive to hypoglycaemia and together these two stimuli can potentiate each other's effects. The chemosensory responses of glomus cells are also preserved at an advanced age. These new data on the cellular and molecular physiology of the CB pave the way for future pathophysiological studies involving this organ in humans.
Collapse
Affiliation(s)
- Patricia Ortega-Sáenz
- J. López-Barneo: Instituto de Biomedicina de Sevilla (IBiS), Campus Hospital Universitario Virgen del Rocío, Avenida Manuel Siurot s/n, 41013 Seville, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
The discovery of the sensory nature of the carotid body dates back to the beginning of the 20th century. Following these seminal discoveries, research into carotid body mechanisms moved forward progressively through the 20th century, with many descriptions of the ultrastructure of the organ and stimulus-response measurements at the level of the whole organ. The later part of 20th century witnessed the first descriptions of the cellular responses and electrophysiology of isolated and cultured type I and type II cells, and there now exist a number of testable hypotheses of chemotransduction. The goal of this article is to provide a comprehensive review of current concepts on sensory transduction and transmission of the hypoxic stimulus at the carotid body with an emphasis on integrating cellular mechanisms with the whole organ responses and highlighting the gaps or discrepancies in our knowledge. It is increasingly evident that in addition to hypoxia, the carotid body responds to a wide variety of blood-borne stimuli, including reduced glucose and immune-related cytokines and we therefore also consider the evidence for a polymodal function of the carotid body and its implications. It is clear that the sensory function of the carotid body exhibits considerable plasticity in response to the chronic perturbations in environmental O2 that is associated with many physiological and pathological conditions. The mechanisms and consequences of carotid body plasticity in health and disease are discussed in the final sections of this article.
Collapse
Affiliation(s)
- Prem Kumar
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, The University of Birmingham, Birmingham, United Kingdom.
| | | |
Collapse
|
17
|
Kefaloyianni E, Coetzee WA. Transcriptional remodeling of ion channel subunits by flow adaptation in human coronary artery endothelial cells. J Vasc Res 2011; 48:357-67. [PMID: 21389733 DOI: 10.1159/000323475] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 11/23/2010] [Indexed: 12/20/2022] Open
Abstract
Endothelial cells (ECs) are constantly exposed to blood flow-induced shear forces in the vessels and this is a major determinant of endothelial function. Ion channels have a major role in endothelial function and in the control of vascular tone. We hypothesized that shear force is a general regulator of ion channel expression, which will have profound effects on endothelial function. We examined this hypothesis using large-scale quantitative real-time RT-PCR. Human coronary artery ECs were exposed to two levels of flow-induced shear stress for 24 h, while control cells were grown under static conditions. The expression of ion channel subunits was compared between control and flow-adapted cells. We used primers against 55 ion channel and exchanger subunits and were able to detect 54 subunits. Five dyn/cm(2) of shear induced downregulation of 1 (NCX1) and upregulation of 18 subunits, including K(Ca)2.2, K(Ca)2.3, CX37, K(v)1.5 and HCN2. Fifteen dyn/cm(2) of shear stress induced the expression of 30 ion channel subunits, including K(Ca)2.3, K(Ca)2.2, CX37, K(ir)2.3 and K(Ca)3.1. Our data demonstrate that substantial remodeling of endothelial ion channel subunit expression occurs with flow adaptation and suggest that altered ion channel expression may significantly contribute to vascular pathology associated with flow-induced alterations.
Collapse
Affiliation(s)
- Eirini Kefaloyianni
- Department of Pediatrics, New York University School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
18
|
Gonzalez C, Agapito MT, Rocher A, Gomez-Niño A, Rigual R, Castañeda J, Conde SV, Obeso A. A revisit to O2 sensing and transduction in the carotid body chemoreceptors in the context of reactive oxygen species biology. Respir Physiol Neurobiol 2010; 174:317-30. [PMID: 20833275 DOI: 10.1016/j.resp.2010.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2010] [Revised: 08/31/2010] [Accepted: 09/01/2010] [Indexed: 11/25/2022]
Abstract
Oxygen-sensing and transduction in purposeful responses in cells and organisms is of great physiological and medical interest. All animals, including humans, encounter in their lifespan many situations in which oxygen availability might be insufficient, whether acutely or chronically, physiologically or pathologically. Therefore to trace at the molecular level the sequence of events or steps connecting the oxygen deficit with the cell responses is of interest in itself as an achievement of science. In addition, it is also of great medical interest as such knowledge might facilitate the therapeutical approach to patients and to design strategies to minimize hypoxic damage. In our article we define the concepts of sensors and transducers, the steps of the hypoxic transduction cascade in the carotid body chemoreceptor cells and also discuss current models of oxygen- sensing (bioenergetic, biosynthetic and conformational) with their supportive and unsupportive data from updated literature. We envision oxygen-sensing in carotid body chemoreceptor cells as a process initiated at the level of plasma membrane and performed by a hemoprotein, which might be NOX4 or a hemoprotein not yet chemically identified. Upon oxygen-desaturation, the sensor would experience conformational changes allosterically transmitted to oxygen regulated K+ channels, the initial effectors in the transduction cascade. A decrease in their opening probability would produce cell depolarization, activation of voltage dependent calcium channels and release of neurotransmitters. Neurotransmitters would activate the nerve endings of the carotid body sensory nerve to convey the information of the hypoxic situation to the central nervous system that would command ventilation to fight hypoxia.
Collapse
Affiliation(s)
- C Gonzalez
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular y CIBER de Enfermedades Respiratorias, Universidad de Valladolid, Consejo Superior de Investigaciones Científicas e Instituto Carlos III, Facultad de Medicina, 47005 Valladolid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Peers C, Wyatt CN, Evans AM. Mechanisms for acute oxygen sensing in the carotid body. Respir Physiol Neurobiol 2010; 174:292-8. [PMID: 20736087 DOI: 10.1016/j.resp.2010.08.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 08/16/2010] [Accepted: 08/17/2010] [Indexed: 12/22/2022]
Abstract
Hypoxic chemotransduction in the carotid body requires release of excitatory transmitters from type I cells that activate afferent sensory neurones. Transmitter release is dependent on voltage-gated Ca2+ entry which is evoked by membrane depolarization. This excitatory response to hypoxia is initiated by inhibition of specific O2 sensitive K+ channels, of which several types have been reported. Here, we discuss mechanisms which have been put forward to account for hypoxic inhibition of type I cell K+ channels. Whilst evidence indicates that one O2 sensitive K+ channel, BKCa, may be regulated by gasotransmitters (CO and H2S) in an O2-dependent manner, other studies now indicate that activation of AMP-activated protein kinase (AMPK) accounts for inhibition of both BKCa and 'leak' O2 sensitive K+ channels, and perhaps also other O2 sensitive K+ channels reported in different species. We propose that type I cell AMPK activation occurs as a result of inhibition of mitochondrial oxidative phosphorylation, and does not require increased production of reactive oxygen species. Thus, AMPK activation provides the basis for unifying the 'membrane' and 'mitochondrial' hypotheses, previously regarded as disparate, to account for hypoxic chemotransduction.
Collapse
Affiliation(s)
- Chris Peers
- Division of Cardiovascular and Neuronal Remodelling, Leeds Institute for Genetics, Health and Therapeutics (LIGHT), Faculty of Medicine and Health, Worsley Building (Level 10), University of Leeds, Clarendon Way, Leeds LS2 9JT, UK.
| | | | | |
Collapse
|
20
|
Evans AM, Hardie DG, Peers C, Wyatt CN, Viollet B, Kumar P, Dallas ML, Ross F, Ikematsu N, Jordan HL, Barr BL, Rafferty JN, Ogunbayo O. Ion channel regulation by AMPK: the route of hypoxia-response coupling in thecarotid body and pulmonary artery. Ann N Y Acad Sci 2009; 1177:89-100. [PMID: 19845611 DOI: 10.1111/j.1749-6632.2009.05041.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vital homeostatic mechanisms monitor O2 supply and adjust respiratory and circulatory function to meet demand. The pulmonary arteries and carotid bodies are key systems in this respect. Hypoxic pulmonary vasoconstriction (HPV) aids ventilation-perfusion matching in the lung by diverting blood flow from areas with an O2 deficit to those rich in O2, while a fall in arterial pO2 increases sensory afferent discharge from the carotid body to elicit corrective changes in breathing patterns. We discuss here the new concept that hypoxia, by inhibiting oxidative phosphorylation, activates AMP-activated protein kinase (AMPK) leading to consequent phosphorylation of target proteins, such as ion channels, which initiate pulmonary artery constriction and carotid body activation. Consistent with this view, AMPK knockout mice exhibit an impaired ventilatory response to hypoxia. Thus, AMPK may be sufficient and necessary for hypoxia-response coupling and may regulate O2 and thereby energy (ATP) supply at the whole body as well as the cellular level.
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Pandit J, Buckler K. Differential effects of halothane and sevoflurane on hypoxia-induced intracellular calcium transients of neonatal rat carotid body type I cells † †This work was presented in part at the 82nd Annual Meeting of the International Anesthetic Research Society, San Francisco, CA, USA, March 29–April 1, 2008. Br J Anaesth 2009; 103:701-10. [DOI: 10.1093/bja/aep223] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
22
|
Kemp PJ, Telezhkin V, Wilkinson WJ, Mears R, Hanmer SB, Gadeberg HC, Müller CT, Riccardi D, Brazier SP. Enzyme-Linked Oxygen Sensing by Potassium Channels. Ann N Y Acad Sci 2009; 1177:112-8. [DOI: 10.1111/j.1749-6632.2009.05025.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
23
|
Rocher A, Caceres AI, Almaraz L, Gonzalez C. EPAC signalling pathways are involved in low PO2 chemoreception in carotid body chemoreceptor cells. J Physiol 2009; 587:4015-27. [PMID: 19581380 DOI: 10.1113/jphysiol.2009.172072] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Chemoreceptor cells of the carotid bodies (CB) are activated by hypoxia and acidosis, responding with an increase in their rate of neurotransmitter release, which in turn increases the electrical activity in the carotid sinus nerve and evokes a homeostatic hyperventilation. Studies in isolated chemoreceptor cells have shown that moderate hypoxias ( 46 mmHg) produces smaller depolarisations and comparable Ca(2+) transients but a much higher catecholamine (CA) release response in intact CBs than intense acidic/hypercapnic stimuli (20% CO(2), pH 6.6). Similarly, intense hypoxia ( 20 mmHg) produces smaller depolarizations and Ca(2+) transients in isolated chemoreceptor cells but a higher CA release response in intact CBs than a pure depolarizing stimulus (30-35 mm external K(+)). Studying the mechanisms responsible for these differences we have found the following. (1) Acidic hypercapnia inhibited I(Ca) (60%; whole cell) and CA release (45%; intact CB) elicited by ionomycin and high K(+). (2) Adenylate cyclase inhibition (SQ-22536; 80 microm) inhibited the hypoxic release response (>50%) and did not affect acidic/hypercapnic release, evidencing that the high gain of hypoxia to elicit neurotransmitter release is cAMP dependent. (3) The last effect was independent of PKA activation, as three kinase inhibitors (H-89, KT 5720 and Rp-cAMP; 10 x IC(50)) did not alter the hypoxic release response. (4) The Epac (exchange protein activated by cAMP) activator (8-pCPT-2-O-Me-cAMP, 100 microm) reversed the effects of the cyclase inhibitor. (5) The Epac inhibitor brefeldin A (100 microm) inhibited (54%) hypoxic induced release. Our findings show for the first time that an Epac-mediated pathway mediates O(2) sensing/transduction in chemoreceptor cells.
Collapse
Affiliation(s)
- Asuncion Rocher
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina-IBGM, Universidad de Valladolid - CSIC, C/Ramon y Cajal no. 7, 47005 Valladolid. Spain
| | | | | | | |
Collapse
|
24
|
DeSimone CV, Lu Y, Bondarenko VE, Morales MJ. S3b amino acid substitutions and ancillary subunits alter the affinity of Heteropoda venatoria toxin 2 for Kv4.3. Mol Pharmacol 2009; 76:125-33. [PMID: 19357248 PMCID: PMC2701457 DOI: 10.1124/mol.109.055657] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 04/08/2009] [Indexed: 01/12/2023] Open
Abstract
Heteropoda venatoria toxin 2 (HpTx2) is an inhibitor cystine knot (ICK)-gating modifier toxin that selectively inhibits Kv4 channels. To characterize the molecular determinants of interaction, we performed alanine scanning of the Kv4.3 S3b region. HpTx2-Kv4.3 interaction had an apparent K(d) value of 2.3 microM. Two alanine mutants in Kv4.3 increased K(d) values to 6.4 microM for V276A and 25 microM for L275A. Simultaneous mutation of both amino acids to alanine nearly eliminated toxin interaction. Unlike Hanatoxin and other well characterized ICK toxins, HpTx2 binding does not require a charged amino acid for interaction. To determine whether the identity of the S3b binding site amino acids altered HpTx2 specificity, we constructed Kv4.3 [LV275IF]. This mutation decreased the K(d) value to 0.54 microM, suggesting that the hydrophobic character of the putative binding site is the most important property for interaction with HpTx2. One mutant, N280A, caused stronger interaction of HpTx2 with Kv4.3; the K(d) value for Kv4.3 [N280A] was 0.26 microM. To understand Kv4.3-based transient outward currents in native tissues, we tested the affinity of HpTx2 for Kv4.3 coexpressed with KChIP2b. The toxin's K(d) value for Kv4.3 + KChIP2b was 0.95 microM. KChIP2b stabilizes the closed state of Kv4.3, suggesting that the increased toxin affinity is due to increased stabilization of the closed state. These data show that HpTx2 binding to Kv4.3 has aspects in common with other ICK gating modifier toxins but that the interventions that increase toxin affinity suggest flexibility toward channel binding that belies its unusual specificity for Kv4 channels.
Collapse
Affiliation(s)
- Christopher V DeSimone
- Department of Physiology and Biophysics, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
25
|
DPPX modifies TEA sensitivity of the Kv4 channels in rabbit carotid body chemoreceptor cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009. [PMID: 19536467 DOI: 10.1007/978-90-481-2259-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Chemoreceptor cells from rabbit carotid body (CB) exhibit transient outward currents reversibly inhibited by low P(o2). Molecular and functional dissection of the components of these outward currents indicates that at least two different channels (Kv4.3 and Kv3.4) contribute to this current. Furthermore, several lines of evidence support the conclusion that Kv4 channel subfamily members (either Kv4.3 alone or Kv4.3/Kv4.1 heteromultimers) are the oxygen sensitive K channels (K(o2)) in rabbit CB chemoreceptor cells. However, the pharmacological characterization of these currents shows that they are almost completely blocked by high external TEA concentrations, while Kv4 channels have been shown to be TEA-insensitive. We hypothesized that the expression of regulatory subunits in chemoreceptor cells could modify TEA sensitivity of Kv4 channels. Here, we explore the presence and functional contribution of DPPX to K(o2) currents in rabbit CB chemoreceptor cells by using DPPX functional knockdown with siRNA. Our data suggest that DPPX proteins are integral components of K(o2) currents, and that their association with Kv4 subunits modulate the pharmacological profile of the heteromultimers.
Collapse
|
26
|
Moreno-Domínguez A, Cidad P, Miguel-Velado E, López-López JR, Pérez-García MT. De novo expression of Kv6.3 contributes to changes in vascular smooth muscle cell excitability in a hypertensive mice strain. J Physiol 2008; 587:625-40. [PMID: 19074965 DOI: 10.1113/jphysiol.2008.165217] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Essential hypertension involves a gradual and sustained increase in total peripheral resistance, reflecting an increased vascular tone. This change associates with a depolarization of vascular myocytes, and relies on a change in the expression profile of voltage-dependent ion channels (mainly Ca(2+) and K(+) channels) that promotes arterial contraction. However, changes in expression and/or modulation of voltage-dependent K(+) channels (Kv channels) are poorly defined, due to their large molecular diversity and their vascular bed-specific expression. Here we endeavor to characterize the molecular and functional expression of Kv channels in vascular smooth muscle cells (VSMCs) and their regulation in essential hypertension, by using VSMCs from resistance (mesenteric) or conduit (aortic) arteries obtained from a hypertensive inbred mice strain, BPH, and the corresponding normotensive strain, BPN. Real-time PCR reveals a differential distribution of Kv channel subunits in the different vascular beds as well as arterial bed-specific changes under hypertension. In mesenteric arteries, the most conspicuous change was the de novo expression of Kv6.3 (Kcng3) mRNA in hypertensive animals. The functional relevance of this change was studied by using patch-clamp techniques. VSMCs from BPH arteries were more depolarized than BPN ones, and showed significantly larger capacitance values. Moreover, Kv current density in BPH VSMCs is decreased mainly due to the diminished contribution of the Kv2 component. The kinetic and pharmacological profile of Kv2 currents suggests that the expression of Kv6.3 could contribute to the natural development of hypertension.
Collapse
Affiliation(s)
- Alejandro Moreno-Domínguez
- Departamento de Fisiología, Universidad de Valladolid, Instituto de Biología y Genética Molecular, c/Sanz y Forés s/n, Valladolid, Spain
| | | | | | | | | |
Collapse
|
27
|
Buniel M, Glazebrook PA, Ramirez-Navarro A, Kunze DL. Distribution of voltage-gated potassium and hyperpolarization-activated channels in sensory afferent fibers in the rat carotid body. J Comp Neurol 2008; 510:367-77. [PMID: 18668683 DOI: 10.1002/cne.21796] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The chemosensory glomus cells of the carotid body (CB) detect changes in O2 tension. Carotid sinus nerve fibers, which originate from peripheral sensory neurons located within the petrosal ganglion, innervate the CB. Release of transmitter from glomus cells activates the sensory afferent fibers to transmit information to the nucleus of the solitary tract in the brainstem. The ion channels expressed within the sensory nerve terminals play an essential role in the ability of the terminal to initiate action potentials in response to transmitter-evoked depolarization. However, with a few exceptions, the identity of ion channels expressed in these peripheral nerve fibers is unknown. This study addresses the expression of voltage-gated channels in the sensory fibers with a focus on channels that set the resting membrane potential and regulate discharge patterns. By using immunohistochemistry and fluorescence confocal microscopy, potassium channel subunits and HCN (hyperpolarization-activated) family members were localized both in petrosal neurons that expressed tyrosine hydroxylase and in the CSN axons within the carotid body. Channels contributing to resting membrane potential, including HCN2 responsible in part for I(h) current and the KCNQ2 and KCNQ5 subunits thought to underlie the neuronal "M current," were identified in the sensory neurons and their axons innervating the carotid body. In addition, the results presented here demonstrate expression of several potassium channels that shape the action potential and the frequency of discharge, including Kv1.4, Kv1.5, Kv4.3, and K(Ca) (BK). The role of these channels should be considered in interpretation of the fiber discharge in response to perturbation of the carotid body environment.
Collapse
Affiliation(s)
- Maria Buniel
- Rammelkamp Center for Education and Research, MetroHealth Campus of Case Western Reserve University, Cleveland, Ohio 44109-1998, USA
| | | | | | | |
Collapse
|
28
|
Colinas O, Pérez-Carretero FD, López-López JR, Pérez-García MT. A role for DPPX modulating external TEA sensitivity of Kv4 channels. ACTA ACUST UNITED AC 2008; 131:455-71. [PMID: 18411327 PMCID: PMC2346566 DOI: 10.1085/jgp.200709912] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Shal-type (Kv4) channels are expressed in a large variety of tissues, where they contribute to transient voltage-dependent K+ currents. Kv4 are the molecular correlate of the A-type current of neurons (ISA), the fast component of ITO current in the heart, and also of the oxygen-sensitive K+ current (KO2) in rabbit carotid body (CB) chemoreceptor cells. The enormous degree of variability in the physiological properties of Kv4-mediated currents can be attributable to the complexity of their regulation together with the large number of ancillary subunits and scaffolding proteins that associate with Kv4 proteins to modify their trafficking and their kinetic properties. Among those, KChIPs and DPPX proteins have been demonstrated to be integral components of ISA and ITO currents, as their coexpression with Kv4 subunits recapitulates the kinetics of native currents. Here, we explore the presence and functional contribution of DPPX to KO2 currents in rabbit CB chemoreceptor cells by using DPPX functional knockdown with siRNA. Additionally, we investigate if the presence of DPPX endows Kv4 channels with new pharmacological properties, as we have observed anomalous tetraethylammonium (TEA) sensitivity in the native KO2 currents. DPPX association with Kv4 channels induced an increased TEA sensitivity both in heterologous expression systems and in CB chemoreceptor cells. Moreover, TEA application to Kv4-DPPX heteromultimers leads to marked kinetic effects that could be explained by an augmented closed-state inactivation. Our data suggest that DPPX proteins are integral components of KO2 currents, and that their association with Kv4 subunits modulate the pharmacological profile of the heteromultimers.
Collapse
Affiliation(s)
- Olaia Colinas
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain
| | | | | | | |
Collapse
|
29
|
Salom JB, Castelló-Ruiz M, Burguete MC, Guzmán C, Jover-Mengual T, Torregrosa G, Jover R, Lizasoain I, Alborch E. Role of K+ and Ca2+ fluxes in the cerebroarterial vasoactive effects of sildenafil. Eur J Pharmacol 2007; 581:138-47. [PMID: 18155692 DOI: 10.1016/j.ejphar.2007.11.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Revised: 11/05/2007] [Accepted: 11/15/2007] [Indexed: 11/28/2022]
Abstract
The aim of this study was to assess the role of K(+) and Ca(2+) fluxes in the cerebroarterial vasoactive effects of the phosphodiesterase-5 inhibitor sildenafil. We used isolated rabbit basilar arteries to assess the effects of extracellular K(+) raising on sildenafil-induced vasodilatation, and studied the pharmacological interaction of sildenafil with selective modulators of membrane K(+) and Ca(2+) channels. Expression of Kv1 subunits of K(+) channels was assessed at messenger and protein levels. Parallel experiments were carried out with zaprinast for comparison. Sildenafil (10 nM-0.1 mM) induced concentration-dependent relaxation of endothelin-1 (10 nM)-precontracted arteries, which was partially inhibited by depolarization with KCl (50 mM), 3 mM tetraethylammonium (non-selective K(+) channel blocker) or 1 mM aminopyridine (inhibitor of K(v) channels), but not by 1 microM glibenclamide (inhibitor of K(ATP) channels) or 50 nM iberiotoxin (inhibitor of K(Ca) channels). Arterial smooth muscle expressed messengers for Kv1.2, Kv1.3, Kv1.4, Kv1.5 and Kv1.6, and proteins of Kv1.1, Kv1.2 and Kv1.4. CaCl(2) (10 microM- 10 mM) induced concentration-dependent contraction in Ca(2+)-free, depolarizing (50 mM KCl) medium. Sildenafil (0.1-100 microM) produced reversible concentration-dependent inhibition of the response to CaCl(2), which was completely abolished by the highest sildenafil concentration. By contrast, only 100 microM zaprinast inhibited the response to CaCl(2). The L-type Ca(2+) channel activator Bay K 8644 (0.1 nM-1 microM) induced concentration-dependent potentiation of the response to CaCl(2) inhibited by 100 microM sildenafil. Moreover, Bay K 8644 (0.1 nM-1 microM) induced concentration-dependent contraction in slightly depolarizing (15 mM) medium, which was inhibited to the same extent and in a concentration-dependent way by sildenafil (0.1-100 microM) and zaprinast (1 or 100 microM). These results show that sildenafil relaxes the rabbit basilar artery by increasing K(+) efflux through K(v) channels, which in turn may affect Ca(2+) signalling. Expression of Kv1 subunits involved in this pharmacological effect occurs at the messenger and, in some cases, at the protein level. In addition to this phosphodiesterase-5-related effect, sildenafil and zaprinast inhibit cerebroarterial vasoconstriction at least in part by directly blocking L-type Ca(2+) channels, although a decrease in the sensitivity of the contractile apparatus to Ca(2+) can not be discarded.
Collapse
Affiliation(s)
- Juan B Salom
- Centro de Investigación, Hospital Universitario La Fe, Valencia, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Dallas ML, Morris NP, Lewis DI, Deuchars SA, Deuchars J. Voltage-gated potassium currents within the dorsal vagal nucleus: inhibition by BDS toxin. Brain Res 2007; 1189:51-7. [PMID: 18048010 DOI: 10.1016/j.brainres.2007.10.090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 10/19/2007] [Accepted: 10/31/2007] [Indexed: 10/22/2022]
Abstract
Voltage-gated potassium (Kv) channels are essential components of neuronal excitability. The Kv3.4 channel protein is widely distributed throughout the central nervous system (CNS), where it can form heteromeric or homomeric Kv3 channels. Electrophysiological studies reported here highlight a functional role for this channel protein within neurons of the dorsal vagal nucleus (DVN). Current clamp experiments revealed that blood depressing substance (BDS) and intracellular dialysis of an anti-Kv3.4 antibody prolonged the action potential duration. In addition, a BDS sensitive, voltage-dependent, slowly inactivating outward current was observed in voltage clamp recordings from DVN neurons. Electrical stimulation of the solitary tract evoked EPSPs and IPSPs in DVN neurons and BDS increased the average amplitude and decreased the paired pulse ratio, consistent with a presynaptic site of action. This presynaptic modulation was action potential dependent as revealed by ongoing synaptic activity. Given the role of the Kv3 proteins in shaping neuronal excitability, these data highlight a role for homomeric Kv3.4 channels in spike timing and neurotransmitter release in low frequency firing neurons of the DVN.
Collapse
Affiliation(s)
- Mark L Dallas
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | | | |
Collapse
|
31
|
Abstract
The ability to sense and react to changes in environmental oxygen levels is crucial to the survival of all aerobic life forms. In mammals, specialized tissues have evolved which can sense and rapidly respond to an acute reduction in oxygen and central to this ability in many is dynamic modulation of ion channels by hypoxia. The most widely studied oxygen-sensitive ion channels are potassium channels but oxygen sensing by members of both the calcium and sodium channel families has also been demonstrated. This chapter will focus on mechanisms of physiological oxygen sensing by ion channels, with particular emphasis on potassium channel function, and will highlight some of the consensuses and controversies within the field. Where data are available, this chapter will also make use of information gleaned from heterologous expression of recombinant proteins in an attempt to consolidate what we know currently about the molecular mechanisms of acute oxygen sensing by ion channels.
Collapse
Affiliation(s)
- Paul J Kemp
- Cardiff School of Bioscience, Cardiff University, Museum Avenue, Cardiff CF10 3US, UK.
| | | |
Collapse
|
32
|
Caceres AI, Obeso A, Gonzalez C, Rocher A. Molecular identification and functional role of voltage-gated sodium channels in rat carotid body chemoreceptor cells. Regulation of expression by chronic hypoxia in vivo. J Neurochem 2007; 102:231-45. [PMID: 17564680 DOI: 10.1111/j.1471-4159.2007.04465.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have assessed the expression, molecular identification and functional role of Na+ channels (Na(v)) in carotid bodies (CB) obtained from normoxic and chronically hypoxic adult rats. Veratridine evoked release of catecholamines (CA) from an in vitro preparation of intact CBs obtained from normoxic animals, the response being Ca2+ and Na+-dependent and sensitive to tetrodotoxin (TTX). TTX inhibited by 25-50% the CA release response evoked by graded hypoxia. Immunoblot assays demonstrated the presence of Na(v)alpha-subunit (c. 220 kDa) in crude homogenates from rat CBs, being evident an up-regulation (60%) of this protein in the CBs obtained from chronically hypoxic rats (10% O2; 7 days). This up-regulation was accompanied by an enhanced TTX-sensitive release response to veratridine, and by an enhanced ventilatory response to acute hypoxic stimuli. RT-PCR studies demonstrated the expression of mRNA for Na(v)1.1, Na(v)1.2, Na(v)1.3, Na(v)1.6 and Na(v)1.7 isoforms. At least three isoforms, Na(v)1.1, Na(v)1.3 and Na(v)1.6 co-localized with tyrosine hydroxylase in all chemoreceptor cells. RT-PCR and immunocytochemistry indicated that Na(v)1.1 isoform was up-regulated by chronic hypoxia in chemoreceptor cells. We conclude that Na(v) up-regulation represents an adaptive mechanism to increase chemoreceptor sensitivity during acclimatization to sustained hypoxia as evidenced by enhanced ventilatory responses to acute hypoxic tests.
Collapse
Affiliation(s)
- Ana I Caceres
- Departamento de Bioquímica, Biología Molecular y Fisiología, Facultad de Medicina/Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid/CSIC, Valladolid, Spain
| | | | | | | |
Collapse
|
33
|
Fu XW, Nurse C, Cutz E. Characterization of slowly inactivating KV{alpha} current in rabbit pulmonary neuroepithelial bodies: effects of hypoxia and nicotine. Am J Physiol Lung Cell Mol Physiol 2007; 293:L892-902. [PMID: 17644754 DOI: 10.1152/ajplung.00098.2007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary neuroepithelial bodies (NEB) form innervated cell clusters that express voltage-activated currents and function as airway O(2) sensors. We investigated A-type K(+) currents in NEB cells using neonatal rabbit lung slice preparation. The whole cell K(+) current was slowly inactivating with activation threshold of approximately -30 mV. This current was blocked approximately 27% by blood-depressing substance I (BDS-I; 3 microM), a selective blocker of Kv3.4 subunit, and reduced approximately 20% by tetraethylammonium (TEA; 100 microM). The BDS-I-sensitive component had an average peak value of 189 +/- 14 pA and showed fast inactivation kinetics that could be fitted by one-component exponential function with a time constant of (tau1) 77 +/- 10 ms. This Kv slowly inactivating current was also blocked by heteropodatoxin-2 (HpTx-2; 0.2 microM), a blocker of Kv4 subunit. The HpTx-2-sensitive current had an average peak value of 234 +/- 23 pA with a time constant (tau) 82 +/- 11 ms. Hypoxia (Po(2) = 15-20 mmHg) inhibited the slowly inactivating K(+) current by approximately 47%, during voltage steps from -30 to +30 mV, and no further inhibition occurred when TEA was combined with hypoxia. Nicotine at concentrations of 50 and 100 microM suppressed the slowly inactivating K(+) current by approximately 24 and approximately 40%, respectively. This suppression was not reversed by mecamylamine suggesting a direct effect of nicotine on these K(+) channels. In situ hybridization experiments detected expression of mRNAs for Kv3.4 and Kv4.3 subunits, while double-label immunofluorescence confirmed membrane localization of respective channel proteins in NEB cells. These studies suggest that the hypoxia-sensitive current in NEB cells is carried by slowly inactivating A-type K(+) channels, which underlie their oxygen-sensitive potassium currents, and that exposure to nicotine may directly affect their function, contributing to smoking-related lung disease.
Collapse
Affiliation(s)
- Xiao Wen Fu
- Division of Pathology, Department of Pediatric Laboratory Medicine, The Research Institute, The Hospital for Sick Children and University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | | | | |
Collapse
|
34
|
López-López JR, Pérez-García MT. Oxygen sensitive Kv channels in the carotid body. Respir Physiol Neurobiol 2007; 157:65-74. [PMID: 17442633 DOI: 10.1016/j.resp.2007.01.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2006] [Revised: 01/17/2007] [Accepted: 01/22/2007] [Indexed: 11/25/2022]
Abstract
Hypoxic inhibition of K(+) channels has been documented in many native chemoreceptor cells, and is crucial to initiate reflexes directed to improve tissue O(2) supply. In the carotid body (CB) chemoreceptors, there is a general consensus regarding the facts that a decrease in P(O2) leads to membrane depolarization, increase of Ca(2+) entry trough voltage-dependent Ca(2+) channels and Ca(2+)-dependent release of neurotransmitters. Central to this pathway is the modulation by hypoxia of K(+) channels that triggers depolarization. However, the details of this process are still controversial, and even the molecular nature of these oxygen-sensitive K(+) (K(O2)) channels in the CB is hotly debated. Clearly there are inter-species differences, and even in the same preparation more that one K(O2) may be present. Here we recapitulate our present knowledge of the role of voltage dependent K(+) channels as K(O2) in the CB from different species, and their functional contribution to cell excitability in response to acute and chronic exposure to hypoxia.
Collapse
Affiliation(s)
- José Ramón López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina e Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, Valladolid, Spain
| | | |
Collapse
|
35
|
Wyatt CN, Evans AM. AMP-activated protein kinase and chemotransduction in the carotid body. Respir Physiol Neurobiol 2007; 157:22-9. [PMID: 17409030 DOI: 10.1016/j.resp.2007.01.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2006] [Revised: 01/18/2007] [Accepted: 01/22/2007] [Indexed: 10/23/2022]
Abstract
AMP-activated protein kinase (AMPK) is a key component of a kinase cascade that regulates energy balance at the cellular level. Our recent research has raised the possibility that AMPK may also function to couple hypoxic inhibition of mitochondrial oxidative phosphorylation to O(2)-sensitive K(+) channel inhibition and hence underpin carotid body type I cell excitation. Thus, in addition to maintaining the cellular energy state AMPK may act as the primary metabolic sensor and effector of hypoxic chemotransduction in type I cells. These findings provide a unifying link between two previously separate theories pertaining to O(2)-sensing in the carotid body, namely the 'membrane hypothesis' and the 'mitochondrial hypothesis'. Furthermore, our data suggest that in addition to its effects at the cellular level the AMPK signalling cascade can mediate vital physiological mechanisms essential for meeting the metabolic needs of the whole organism.
Collapse
|
36
|
Schultz HD, Li YL. Carotid body function in heart failure. Respir Physiol Neurobiol 2007; 157:171-85. [PMID: 17374517 PMCID: PMC1965591 DOI: 10.1016/j.resp.2007.02.011] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2007] [Revised: 02/07/2007] [Accepted: 02/08/2007] [Indexed: 11/16/2022]
Abstract
In this review, we summarize the present state of knowledge of the functional characteristics of the carotid body (CB) chemoreflex with respect to control of sympathetic nerve activity (SNA) in chronic heart failure (CHF). Evidence from both CHF patients and animal models of CHF has clearly established that the CB chemoreflex is enhanced in CHF and contributes to the tonic elevation in SNA. This adaptive change derives from altered function at the level of both the afferent and central nervous system (CNS) pathways of the reflex arc. At the level of the CB, an elevation in basal afferent discharge occurs under normoxic conditions in CHF rabbits, and the discharge responsiveness to hypoxia is enhanced. Outward voltage-gated K(+) currents (I(K)) are suppressed in CB glomus cells from CHF rabbits, and their sensitivity to hypoxic inhibition is enhanced. These changes in I(K) derive partly from downregulation of nitric oxide synthase (NOS)/NO signaling and upregulation of angiotensin II (Ang II)/Ang II receptor (AT(1)R) signaling in glomus cells. At the level of the CNS, interactions of the enhanced input from CB chemoreceptors with altered input from baroreceptor and cardiac afferent pathways and from central Ang II further enhance sympathetic drive. In addition, impaired function of NO in the paraventricular nucleus of the hypothalamus participates in the increased SNA response to CB chemoreceptor activation. These results underscore the principle that multiple mechanisms involving Ang II and NO at the level of both the CB and CNS represent complementary and perhaps redundant adaptive mechanisms to enhance CB chemoreflex function in CHF.
Collapse
Affiliation(s)
- Harold D Schultz
- Department of Cellular and Integrative Physiology, University of Nebraska College of Medicine, 985850 Nebraska Medical Center, Omaha, NE 68198-5850, USA.
| | | |
Collapse
|
37
|
Gonzalez C, Agapito MT, Rocher A, Gonzalez-Martin MC, Vega-Agapito V, Gomez-Niño A, Rigual R, Castañeda J, Obeso A. Chemoreception in the context of the general biology of ROS. Respir Physiol Neurobiol 2007; 157:30-44. [PMID: 17331812 DOI: 10.1016/j.resp.2007.01.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 01/23/2007] [Accepted: 01/23/2007] [Indexed: 11/28/2022]
Abstract
Superoxide anion is the most important reactive oxygen species (ROS) primarily generated in cells. The main cellular constituents with capabilities to generate superoxide anion are NADPH oxidases and mitochondrial respiratory chain. The emphasis of our article is centered in critically examining hypotheses proposing that ROS generated by NADPH oxidase and mitochondria are key elements in O(2)-sensing and hypoxic responses generation in carotid body chemoreceptor cells. Available data indicate that chemoreceptor cells express a specific isoform of NADPH oxidase that is activated by hypoxia; generated ROS acting as negative modulators of the carotid body (CB) hypoxic responses. Literature is also consistent in supporting that poisoned respiratory chain can produce high amounts of ROS, making mitochondrial ROS potential triggers-modulators of the CB activation elicited by mitochondrial venoms. However, most data favour the notion that levels of hypoxia, capable of strongly activating chemoreceptor cells, would not increase the rate of ROS production in mitochondria, making mitochondrial ROS unlikely triggers of hypoxic responses in the CB. Finally, we review recent literature on heme oxygenases from two perspectives, as potential O(2)-sensors in chemoreceptor cells and as generators of bilirubin which is considered to be a ROS scavenger of major quantitative importance in mammalian cells.
Collapse
Affiliation(s)
- C Gonzalez
- Departamento de Bioquímica y Biología Molecular y Fisiología e, Instituto de Biología y Genética Molecular, Facultad de Medicina, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, Valladolid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fitzgerald RS, Shirahata M, Balbir A, Grossman CE. Oxygen sensing in the carotid body and its relation to heart failure. Antioxid Redox Signal 2007; 9:745-9. [PMID: 17511590 DOI: 10.1089/ars.2007.1546] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This brief review first touches on the origins of the earth's oxygen. It then identifies and locates the principal oxygen sensor in vertebrates, the carotid body (CB). The CB is unique in that in human subjects, it is the only sensor of lower than normal levels in the partial pressure of oxygen (hypoxia, HH). Another oxygen sensor, the aortic bodies, are mostly vestigial in higher vertebrates. At least they play a much smaller role than the CB. In such an important role, the many reflexes in response to CB stimulation by HH are presented. After briefly reviewing what CB stimulation does, the next topic is to describe how the CB chemotransduces HH into neural signals to the brain. Several mechanisms are known, but critical steps in the mechanisms of chemosensation and chemotransduction are still under investigation. Finally, a brief glance at the operation of the CB in chronic heart failure patients is presented. Specifically, the role of nitric oxide, NO, is discussed.
Collapse
Affiliation(s)
- Robert S Fitzgerald
- Department of Environmental Health Sciences, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA.
| | | | | | | |
Collapse
|
39
|
Ward DS, Voter WA, Karan S. The effects of hypo- and hyperglycaemia on the hypoxic ventilatory response in humans. J Physiol 2007; 582:859-69. [PMID: 17478538 PMCID: PMC2075331 DOI: 10.1113/jphysiol.2007.130112] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Animal and tissue studies have indicated that the carotid bodies are sensitive to glucose concentrations within the physiological range. This glucose sensitivity may modulate the ventilatory response to hypoxia, with hyperglycaemia suppressing the hypoxic response and hypoglycaemia stimulating it. This study was designed to determine whether hypo- and hyperglycaemia modulate the hypoxic ventilatory response in humans. In 11 normal research participants, glucose levels were clamped at 2.8 and 11.2 mmol l(-1) for 30 min. At the start and end of each clamp, blood was drawn for hormone measurement and the isocapnic hypoxic ventilatory response was measured. Because generation of reactive oxygen species may be a common pathway for the interaction between glucose and oxygen levels, the experiments were repeated with and without pretreatment for 1 week with vitamins C and E. Hypoglycaemia caused an increase in the counter-regulatory hormones, a 54% increase in isocapnic ventilation, and a 108% increase in the hypoxic ventilatory response. By contrast, hyperglycaemia resulted in small but significant increases in both ventilation and the hypoxic ventilatory response. Antioxidant vitamin pretreatment altered neither response. In conclusion, the stimulant effect of hypoglycaemia on the hypoxic ventilatory response is consistent with a direct effect on the carotid body, but an indirect effect through the activation of the counter-regulatory response cannot be excluded. The mechanisms behind the mild stimulating effect of hyperglycaemia remain to be elucidated.
Collapse
Affiliation(s)
- Denham S Ward
- Department of Anesthesiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
40
|
Balbir A, Lee H, Okumura M, Biswal S, Fitzgerald RS, Shirahata M. A search for genes that may confer divergent morphology and function in the carotid body between two strains of mice. Am J Physiol Lung Cell Mol Physiol 2007; 292:L704-15. [PMID: 17098806 DOI: 10.1152/ajplung.00383.2006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The carotid body (CB) is the primary hypoxic chemosensory organ. Its hypoxic response appears to be genetically controlled. We have hypothesized that: 1) genes related to CB function are expressed less in the A/J mice (low responder to hypoxia) compared with DBA/2J mice (high responder to hypoxia); and 2) gene expression levels of morphogenic and trophic factors of the CB are significantly lower in the A/J mice than DBA/2J mice. This study utilizes microarray analysis to test these hypotheses. Three sets of CBs were harvested from both strains. RNA was isolated and used for global gene expression profiling (Affymetrix Mouse 430 v2.0 array). Statistically significant gene expression was determined as a minimum six counts of nine pairwise comparisons, a minimum 1.5-fold change, and P ≤ 0.05. Our results demonstrated that 793 genes were expressed less and that 568 genes were expressed more in the A/J strain vs. the DBA/2J strain. Analysis of individual genes indicates that genes encoding ion channels are differentially expressed between the two strains. Genes related to neurotransmitter metabolism, synaptic vesicles, and the development of neural crest-derived cells are expressed less in the A/J CB vs. the DBA/2J CB. Through pathway analysis, we have constructed a model that shows gene interactions and offers a roadmap to investigate CB development and hypoxic chemosensing/chemotransduction processes. Particularly, Gdnf, Bmp2, Kcnmb2, Tph1, Hif1a, and Arnt2 may contribute to the functional differences in the CB between the two strains. Bmp2, Phox2b, Dlx2, and Msx2 may be important for the morphological differences.
Collapse
Affiliation(s)
- Alexander Balbir
- Division of Physiology, Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, E7610, 615 N. Wolfe St., Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
41
|
Shirahata M, Balbir A, Otsubo T, Fitzgerald RS. Role of acetylcholine in neurotransmission of the carotid body. Respir Physiol Neurobiol 2007; 157:93-105. [PMID: 17284361 DOI: 10.1016/j.resp.2006.12.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Revised: 12/11/2006] [Accepted: 12/27/2006] [Indexed: 01/19/2023]
Abstract
Acetylcholine (ACh) has been considered an important excitatory neurotransmitter in the carotid body (CB). Its physiological and pharmacological effects, metabolism, release, and receptors have been well documented in several species. Various nicotinic and muscarinic ACh receptors are present in both afferent nerve endings and glomus cells. Therefore, ACh can depolarize or hyperpolarize the cell membrane depending on the available receptor type in the vicinity. Binding of ACh to its receptor can create a wide variety of cellular responses including opening cation channels (nicotinic ACh receptor activation), releasing Ca(2+) from intracellular storage sites (via muscarinic ACh receptors), and modulating activities of K(+) and Ca(2+) channels. Interactions between ACh and other neurotransmitters (dopamine, adenosine, nitric oxide) have been known, and they may induce complicated responses. Cholinergic biology in the CB differs among species and even within the same species due to different genetic composition. Development and environment influence cholinergic biology. We discuss these issues in light of current knowledge of neuroscience.
Collapse
Affiliation(s)
- Machiko Shirahata
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
42
|
Peers C, Wyatt CN. The role of maxiK channels in carotid body chemotransduction. Respir Physiol Neurobiol 2006; 157:75-82. [PMID: 17157084 DOI: 10.1016/j.resp.2006.10.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 10/27/2006] [Accepted: 10/28/2006] [Indexed: 01/10/2023]
Abstract
MaxiK channels are a unique class of K(+) channels activated by both voltage and intracellular Ca(2+). Derived from a single gene, their diversity arises from extensive splicing, and their wide distribution has led to their implication in a large variety of cellular functions. In the carotid body, they have been proposed to contribute to the resting membrane potential of type I cells, and also to be O(2) sensitive. Thus, they have been suggested to have an important role in hypoxic chemotransduction. Their O(2) sensitivity is preserved when the channels are expressed in HEK 293 cells, permitting detailed studies of candidate mechanisms underlying hypoxic inhibition of maxiK channels. In this article, we review evidence for and against an important role for maxiK channels in chemotransduction. We also consider different mechanisms proposed to account for hypoxic channel inhibition and suggest that, although our understanding of this important physiological process has advanced significantly in recent years, there remain important, unanswered questions as to the importance of maxiK in carotid body chemoreception.
Collapse
Affiliation(s)
- Chris Peers
- School of Medicine, University of Leeds, Leeds, UK.
| | | |
Collapse
|
43
|
Colinas O, Gallego M, Setién R, López-López JR, Pérez-García MT, Casis O. Differential modulation of Kv4.2 and Kv4.3 channels by calmodulin-dependent protein kinase II in rat cardiac myocytes. Am J Physiol Heart Circ Physiol 2006; 291:H1978-87. [PMID: 16648177 DOI: 10.1152/ajpheart.01373.2005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this work we have combined biochemical and electrophysiological approaches to explore the modulation of rat ventricular transient outward K+ current ( Ito) by calmodulin kinase II (CaMKII). Intracellular application of CaMKII inhibitors KN93, calmidazolium, and autocamtide-2-related inhibitory peptide II (ARIP-II) accelerated the inactivation of Ito, even at low [Ca2+]. In the same conditions, CaMKII coimmunoprecipitated with Kv4.3 channels, suggesting that phosphorylation of Kv4.3 channels modulate inactivation of Ito. Because channels underlying Ito are heteromultimers of Kv4.2 and Kv4.3, we have explored the effect of CaMKII on human embryonic kidney (HEK) cells transfected with either of those Kvα-subunits. Whereas Kv4.3 inactivated faster upon inhibition of CaMKII, Kv4.2 inactivation was insensitive to CaMKII inhibitors. However, Kv4.2 inactivation became slower when high Ca2+ was used in the pipette or when intracellular [Ca2+] ([Ca2+]i) was transiently increased. This effect was inhibited by KN93, and Western blot analysis demonstrated Ca2+-dependent phosphorylation of Kv4.2 channels. On the contrary, CaMKII coimmunoprecipitated with Kv4.3 channels without a previous Ca2+ increase, and the association was inhibited by KN93. These results suggest that both channels underlying Ito are substrates of CaMKII, although with different sensitivities; Kv4.2 remain unphosphorylated unless [Ca2+]i increases, whereas Kv4.3 are phosphorylated at rest. In addition to the functional impact that phosphorylation of Kv4 channels could cause on the shape of action potential, association of CaMKII with Kv4.3 provides a new role of Kv4.3 subunits as molecular scaffolds for concentrating CaMKII in the membrane, allowing Ca2+-dependent modulation by this enzyme of the associated Kv4.2 channels.
Collapse
Affiliation(s)
- Olaia Colinas
- Departamento de Bioquímica y Biología Molecular y Fisiología, Edificio IBGM, Universidad de Valladolid, C/ Sanz y Forés s/n, 47003 Valladolid, Spain
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
The majority of physiological processes proceed most favourably when O(2) is in plentiful supply. However, there are a number of physiological and pathological circumstances in which this supply is reduced either acutely or chronically. A crucial homeostatic response to such arterial hypoxaemia is carotid body excitation and a resultant increase in ventilation. Central to this response in carotid body, and many other chemosensory tissues, is the rapid inhibition of ion channels by hypoxia. Since the first direct demonstration of hypoxia-evoked depression in K(+) channel activity, the numbers of mechanisms which have been proposed to serve as the primary O(2) sensor have been almost as numerous as the experimental strategies with which to probe their nature. Three of the current favourite candidate mechanisms are mitochondria, AMP-activated kinase and haemoxygenase-2; a fourth proposal has been NADPH oxidase, but recent evidence suggests that this enzyme plays a secondary role in the O(2)-sensing process. All of these proposals have attractive points, but none can fully reconcile all of the data which have accumulated over the last two decades or so, suggesting that there may, in fact, not be a unique sensing system even within a single cell type. This latter point is key, because it implies that the ability of a cell to respond appropriately to decreased O(2) availability is biologically so important that several mechanisms have evolved to ensure that cellular function is never compromised during moderate to severe hypoxic insult.
Collapse
Affiliation(s)
- Paul J Kemp
- School of Biosciences, Museum Avenue, Cardiff University, Cardiff CF11 9BX, UK.
| |
Collapse
|
45
|
Li YL, Schultz HD. Enhanced sensitivity of Kv channels to hypoxia in the rabbit carotid body in heart failure: role of angiotensin II. J Physiol 2006; 575:215-27. [PMID: 16777942 PMCID: PMC1819433 DOI: 10.1113/jphysiol.2006.110700] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Angiotensin II (Ang II) plays an important role in the enhanced chemoreflex function that occurs in congestive heart failure (CHF), but the mechanism of this effect within the carotid body (CB) is not known. We investigated the sensitivity of Ca2+-independent, voltage-gated K+ (Kv) channels to hypoxia in CB glomus cells from CHF rabbits, and whether endogenous angiotensin II (Ang II) modulates this action. Using the conventional whole-cell patch clamp technique, we found that Kv currents (IK) under normoxic conditions were blunted in the CB glomus cells from CHF rabbits compared with sham rabbits. In addition, the inhibition of IK and the decrease of resting membrane potential (RMP) induced by hypoxia were greater in CHF versus sham glomus cells. Ang II, at 100 pM, had no direct effect on IK at constant normoxic PO2, but increased the sensitivity of IK and RMP to hypoxia in sham glomus cells. In CHF glomus cells, an AT1 receptor (AT1R) antagonist, L-158 809 (1 microM), alone did not affect IK at normoxia, but it decreased the sensitivity of IK and RMP to hypoxia. At higher concentrations, Ang II dose dependently (0.1-100 nM) reduced IK under constant normoxic conditions in sham and CHF glomus cells, with threshold concentrations of about 900 and 600 pM, respectively. Immunocytochemical and Western blot assessments demonstrated the down-expression of Kv3.4 but not Kv4.3 channels in CHF glomus cells. These results indicate that: (1) Ang II/AT1R signalling increases the sensitivity of Kv channels to hypoxia in CB glomus cells from CHF rabbits; (2) high concentrations of Ang II (> 1 nM) directly inhibit IK in CB glomus cells from sham and CHF rabbits; (3) changes in Kv channel protein expression (Kv3.4 versus Kv4.3) in the CB glomus cell may contribute to the suppression of IK and enhanced sensitivity of IK to hypoxia in CHF.
Collapse
Affiliation(s)
- Yu-Long Li
- Department of Cellular and Integrative Physiology, University of, Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | | |
Collapse
|
46
|
Abstract
All cells respond to metabolic stress. However, a variety of specialized cells, commonly referred to as O2-sensing cells, are acutely sensitive to relatively small changes in PO2. Within a variety of organisms such O2-sensing cells have evolved as vital homeostatic mechanisms that monitor O2 supply and alter respiratory and circulatory function, as well as the capacity of the blood to transport O2. Thereby, arterial PO2 may be maintained within physiological limits. In mammals, for example, two key tissues that contribute to this process are the pulmonary arteries and the carotid bodies. Constriction of pulmonary arteries by hypoxia optimizes ventilation-perfusion matching in the lung, whilst carotid body excitation by hypoxia initiates corrective changes in breathing patterns via increased sensory afferent discharge to the brain stem. Despite extensive investigation, the precise mechanism(s) by which hypoxia mediates these responses has remained elusive. It is clear, however, that hypoxia inhibits mitochondrial function in O2-sensing cells over a range of PO2 that has no such effect on other cell types. This raised the possibility that AMP-activated protein kinase might function to couple mitochondrial oxidative phosphorylation to Ca2+ signalling mechanisms in O2-sensing cells and thereby underpin pulmonary artery constriction and carotid body excitation by hypoxia. Our recent investigations have provided significant evidence in support of this view.
Collapse
Affiliation(s)
- A Mark Evans
- Division of Biomedical Sciences, School of Biology, Bute Building, University of St Andrews, St Andrews, Fife KY16 9TS, UK.
| |
Collapse
|
47
|
Fu XW, Cutz E. Identification and characterization of hypoxia sensitive Kvalpha subunits in pulmonary neuroepithelial bodies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 580:313-8; discussion 351-9. [PMID: 16683737 DOI: 10.1007/0-387-31311-7_48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Affiliation(s)
- X W Fu
- Division of Pathology, Department of Pediatric Laboratory Medicine, The Research Institute, The Hospital for Sick Children and University of Toronto, Toronto, Canada
| | | |
Collapse
|
48
|
Han P, Lucero MT. Pituitary adenylate cyclase activating polypeptide reduces expression of Kv1.4 and Kv4.2 subunits underlying A-type K(+) current in adult mouse olfactory neuroepithelia. Neuroscience 2006; 138:411-9. [PMID: 16426762 DOI: 10.1016/j.neuroscience.2005.11.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2005] [Revised: 11/09/2005] [Accepted: 11/20/2005] [Indexed: 10/25/2022]
Abstract
A-type K(+) currents (I(A)) in olfactory receptor neurons have been characterized electrophysiologically but the molecular identities of the underlying channel subunits have not been determined. Using RT-PCR, immunoblot and immunohistochemistry, we found that the two candidate channel families underlying I(A), shaker and shal, are expressed in olfactory epithelia of Swiss Webster mice. Specifically, Kv1.4, the only I(A) candidate from the shaker family, and Kv4.2 and Kv4.3 from the shal family were expressed, but Kv4.1 mRNA was not amplified from the olfactory epithelia. Immunoblot and immunohistochemical studies confirmed the existence of Kv1.4 and Kv4.2/3 subunits. Furthermore, quantitative RT-PCR showed that pituitary adenylate cyclase activating polypeptide (PACAP) reduced the expression of Kv1.4 and Kv4.2 but did not reduce the already low expression of Kv4.3. The PACAP-induced reduction of Kv4.1 and Kv4.2 expression was completely blocked by inhibiting the phospholipase C (PLC) pathway but was still significantly downregulated by PACAP when the cyclic AMP pathway was inhibited. In addition, downstream of the PLC pathway, calcium mediated the reduction of both Kv1.4 and Kv4.2 expression and I(A) current density. Phosphokinase C (PKC) activation did not affect Kv1.4 and Kv4.2 mRNA expression, even though PKC reduced I(A) current density. Together with our previous studies, our data suggest that A-type K(+) currents in olfactory receptor neurons are composed of multiple K(+) channel subunits, among which Kv1.4 and Kv4.2 are subject to transcriptional modulation by PACAP. We also found that PACAP predominately uses a PLC-calcium pathway to modulate Kv4.1 and Kv4.2 expression. Modulation of A-type K(+) current expression may contribute to the previously observed neuroprotective effects of PACAP on olfactory receptor neurons.
Collapse
Affiliation(s)
- P Han
- Interdepartmental Neuroscience Program, University of Utah, 20 North 1900 East, Salt Lake City, UT 84132-3401, USA
| | | |
Collapse
|
49
|
Yamamoto Y, Taniguchi K. Expression of Tandem P Domain K+ Channel, TREK-1, in the Rat Carotid Body. J Histochem Cytochem 2006; 54:467-72. [PMID: 16344329 DOI: 10.1369/jhc.5a6755.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
TREK-1 is one of the important potassium channels for regulating membrane excitability. To examine the distribution of TREK-1 in the rat carotid body, we performed RT-PCR for mRNA expression and in situ hybridization and immunohistochemistry for tissue distribution of TREK-1. RT-PCR detected mRNA expression of TREK-1 in the carotid body. Furthermore, in situ hybridization revealed the localization of TREK-1 mRNA in the glomus cells. TREK-1 immunoreactivity was mainly distributed in the glomus cells and nerve fibers in the carotid body. TREK-1 may modulate potassium current of glomus cells and/or afferent nerve endings in the rat carotid body.
Collapse
Affiliation(s)
- Y Yamamoto
- Laboratory of Veterinary Anatomy, Faculty of Agriculture, Iwate University, Morioka, Iwate 080-8550, Japan.
| | | |
Collapse
|
50
|
Yamamoto Y, Taniguchi K. Immunolocalization of tandem pore domain K+ channels in the rat carotid body. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 580:9-14; discussion 351-9. [PMID: 16683691 DOI: 10.1007/0-387-31311-7_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Affiliation(s)
- Yoshio Yamamoto
- Laboratory of Veterinary Anatomy, Faculty of Agriculture, Iwate University, 18-8, Ueda 3-chome, Morioka, Iwate 020-8550, Japan
| | | |
Collapse
|