1
|
Morales A, Mohan R, Chen X, Coffman BL, Bendahmane M, Watch L, West JL, Bakshi S, Traynor JR, Giovannucci DR, Kammermeier PJ, Axelrod D, Currie KP, Smrcka AV, Anantharam A. PACAP and acetylcholine cause distinct Ca2+ signals and secretory responses in chromaffin cells. J Gen Physiol 2023; 155:e202213180. [PMID: 36538657 PMCID: PMC9770323 DOI: 10.1085/jgp.202213180] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/22/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022] Open
Abstract
The adrenomedullary chromaffin cell transduces chemical messages into outputs that regulate end organ function throughout the periphery. At least two important neurotransmitters are released by innervating preganglionic neurons to stimulate exocytosis in the chromaffin cell-acetylcholine (ACh) and pituitary adenylate cyclase activating polypeptide (PACAP). Although PACAP is widely acknowledged as an important secretagogue in this system, the pathway coupling PACAP stimulation to chromaffin cell secretion is poorly understood. The goal of this study is to address this knowledge gap. Here, it is shown that PACAP activates a Gαs-coupled pathway that must signal through phospholipase C ε (PLCε) to drive Ca2+ entry and exocytosis. PACAP stimulation causes a complex pattern of Ca2+ signals in chromaffin cells, leading to a sustained secretory response that is kinetically distinct from the form stimulated by ACh. Exocytosis caused by PACAP is associated with slower release of peptide cargo than exocytosis stimulated by ACh. Importantly, only the secretory response to PACAP, not ACh, is eliminated in cells lacking PLCε expression. The data show that ACh and PACAP, acting through distinct signaling pathways, enable nuanced and variable secretory outputs from chromaffin cells.
Collapse
Affiliation(s)
- Alina Morales
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Ramkumar Mohan
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Xiaohuan Chen
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | | | | | - Lester Watch
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Joshua L. West
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Shreeya Bakshi
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - John R. Traynor
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | | | - Paul J. Kammermeier
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, USA
| | - Daniel Axelrod
- Department of Physics and LSA Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Kevin P.M. Currie
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Arun Anantharam
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
2
|
Tamang HK, Yang R, Song Z, Hsu S, Peng C, Tung Y, Tzeng B, Chen C. Ca v 3.2 T-type calcium channel regulates mouse platelet activation and arterial thrombosis. J Thromb Haemost 2022; 20:1887-1899. [PMID: 35490411 PMCID: PMC9541131 DOI: 10.1111/jth.15745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cav 3.2 is a T-type calcium channel that causes low-threshold exocytosis. T-type calcium channel blockers reduce platelet granule exocytosis and aggregation. However, studies of the T-type calcium channel in platelets are lacking. OBJECTIVE To examine the expression and role of Cav 3.2 in platelet function. METHODS Global Cav 3.2-/- and platelet-specific Cav 3.2-/- mice and littermate controls were used for this study. Western blot analysis was used to detect the presence of Cav 3.2 and activation of the calcium-responsive protein extracellular signal-regulated kinase (ERK). Fura-2 dye was used to assess platelet calcium. Flow cytometry and light transmission aggregometry were used to evaluate platelet activation markers and aggregation, respectively. FeCl3 -induced thrombosis and a microfluidic flow device were used to assess in vivo and ex vivo thrombosis, respectively. RESULTS Cav 3.2 was expressed in mouse platelets. As compared with wild-type controls, Cav 3.2-/- mouse platelets showed reduced calcium influx. Similarly, treatment with the T-type calcium channel inhibitor Ni2+ decreased the calcium influx in wild-type platelets. As compared with controls, both Cav 3.2-/- and Ni2+ -treated wild-type platelets showed reduced activation of ERK. ATP release, P-selectin exposure, and αIIb β3 activation were reduced in Cav 3.2-/- and Ni2+ -treated wild-type platelets, as was platelet aggregation. On in vivo and ex vivo thrombosis assay, Cav3.2 deletion caused delayed thrombus formation. However, tail bleeding assay showed intact hemostasis. CONCLUSION These results suggest that Cav 3.2 is required for the optimal activation of platelets.
Collapse
Affiliation(s)
- Hem Kumar Tamang
- Taiwan International Graduate Program in Molecular MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipeiTaiwan
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Ruey‐Bing Yang
- Taiwan International Graduate Program in Molecular MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipeiTaiwan
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Zong‐Han Song
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Shao‐Chun Hsu
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | | | - Yi‐Chung Tung
- Research Center for Applied SciencesAcademia SinicaTaipeiTaiwan
| | - Bing‐Hsiean Tzeng
- Division of CardiologyFar Eastern Memorial Hospital and Tri‐Service General HospitalNational Defense Medical CenterTaipeiTaiwan
| | - Chien‐Chang Chen
- Taiwan International Graduate Program in Molecular MedicineNational Yang Ming Chiao Tung University and Academia SinicaTaipeiTaiwan
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| |
Collapse
|
3
|
Carbone E, Borges R, Eiden LE, García AG, Hernández-Cruz A. Chromaffin Cells of the Adrenal Medulla: Physiology, Pharmacology, and Disease. Compr Physiol 2019; 9:1443-1502. [PMID: 31688964 DOI: 10.1002/cphy.c190003] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Chromaffin cells (CCs) of the adrenal gland and the sympathetic nervous system produce the catecholamines (epinephrine and norepinephrine; EPI and NE) needed to coordinate the bodily "fight-or-flight" response to fear, stress, exercise, or conflict. EPI and NE release from CCs is regulated both neurogenically by splanchnic nerve fibers and nonneurogenically by hormones (histamine, corticosteroids, angiotensin, and others) and paracrine messengers [EPI, NE, adenosine triphosphate, opioids, γ-aminobutyric acid (GABA), etc.]. The "stimulus-secretion" coupling of CCs is a Ca2+ -dependent process regulated by Ca2+ entry through voltage-gated Ca2+ channels, Ca2+ pumps, and exchangers and intracellular organelles (RE and mitochondria) and diffusible buffers that provide both Ca2+ -homeostasis and Ca2+ -signaling that ultimately trigger exocytosis. CCs also express Na+ and K+ channels and ionotropic (nAChR and GABAA ) and metabotropic receptors (mACh, PACAP, β-AR, 5-HT, histamine, angiotensin, and others) that make CCs excitable and responsive to autocrine and paracrine stimuli. To maintain high rates of E/NE secretion during stressful conditions, CCs possess a large number of secretory chromaffin granules (CGs) and members of the soluble NSF-attachment receptor complex protein family that allow docking, fusion, and exocytosis of CGs at the cell membrane, and their recycling. This article attempts to provide an updated account of well-established features of the molecular processes regulating CC function, and a survey of the as-yet-unsolved but important questions relating to CC function and dysfunction that have been the subject of intense research over the past 15 years. Examples of CCs as a model system to understand the molecular mechanisms associated with neurodegenerative diseases are also provided. Published 2019. Compr Physiol 9:1443-1502, 2019.
Collapse
Affiliation(s)
- Emilio Carbone
- Laboratory of Cellular and Molecular Neuroscience, Department of Drug Science, N.I.S. Centre, University of Torino, Torino, Italy
| | - Ricardo Borges
- Unidad de Farmacología, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Antonio G García
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, Universidad Autónoma de Madrid, Madrid, Spain
| | - Arturo Hernández-Cruz
- Departamento de Neurociencia Cognitiva and Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular, Universidad Nacional Autonoma de México, Ciudad Universitaria, CDMX, México
| |
Collapse
|
4
|
Rodrigues AL, Brescia M, Koschinski A, Moreira TH, Cameron RT, Baillie G, Beirão PSL, Zaccolo M, Cruz JS. Increase in Ca 2+ current by sustained cAMP levels enhances proliferation rate in GH3 cells. Life Sci 2017; 192:144-150. [PMID: 29183797 DOI: 10.1016/j.lfs.2017.11.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 11/20/2017] [Accepted: 11/24/2017] [Indexed: 12/12/2022]
Abstract
AIMS Ca2+ and cAMP are important intracellular modulators. In order to generate intracellular signals with various amplitudes, as well as different temporal and spatial properties, a tightly and precise control of these modulators in intracellular compartments is necessary. The aim of this study was to evaluate the effects of elevated and sustained cAMP levels on voltage-dependent Ca2+ currents and proliferation in pituitary tumor GH3 cells. MAIN METHODS Effect of long-term exposure to forskolin and dibutyryl-cyclic AMP (dbcAMP) on Ca2+ current density and cell proliferation rate were determined by using the whole-cell patch-clamp technique and real time cell monitoring system. The cAMP levels were assayed, after exposing transfected GH3 cells with the EPAC-1 cAMP sensor to forskolin and dbcAMP, by FRET analysis. KEY FINDINGS Sustained forskolin treatment (24 and 48h) induced a significant increase in total Ca2+ current density in GH3 cells. Accordingly, dibutyryl-cAMP incubation (dbcAMP) also elicited increase in Ca2+ current density. However, the maximum effect of dbcAMP occurred only after 72h incubation, whereas forskolin showed maximal effect at 48h. FRET-experiments confirmed that the time-course to elevate intracellular cAMP was distinct between forskolin and dbcAMP. Mibefradil inhibited the fast inactivating current component selectively, indicating the recruitment of T-type Ca2+ channels. A significant increase on cell proliferation rate, which could be related to the elevated and sustained intracellular levels of cAMP was observed. SIGNIFICANCE We conclude that maintaining high levels of intracellular cAMP will cause an increase in Ca2+ current density and this phenomenon impacts proliferation rate in GH3 cells.
Collapse
Affiliation(s)
- Andréia Laura Rodrigues
- Laboratório CaCIA, Faculdade de Ciências Humanas Sociais e da Saúde, Universidade FUMEC, Brazil.
| | - Marcella Brescia
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Andreas Koschinski
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Thaís Helena Moreira
- Laboratório de Membranas Excitáveis e de Biologia Cardiovascular, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ryan T Cameron
- Institute of Cardiovascular and Medical Sciences, Glasgow University, Glasgow, UK
| | - George Baillie
- Institute of Cardiovascular and Medical Sciences, Glasgow University, Glasgow, UK
| | - Paulo S L Beirão
- Laboratório de Membranas Excitáveis e de Biologia Cardiovascular, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, UK
| | - Jader S Cruz
- Laboratório de Membranas Excitáveis e de Biologia Cardiovascular, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
5
|
L-type calcium channels in exocytosis and endocytosis of chromaffin cells. Pflugers Arch 2017; 470:53-60. [DOI: 10.1007/s00424-017-2064-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 11/25/2022]
|
6
|
Roles of Na +, Ca 2+, and K + channels in the generation of repetitive firing and rhythmic bursting in adrenal chromaffin cells. Pflugers Arch 2017; 470:39-52. [PMID: 28776261 DOI: 10.1007/s00424-017-2048-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/23/2017] [Indexed: 12/30/2022]
Abstract
Adrenal chromaffin cells (CCs) are the main source of circulating catecholamines (CAs) that regulate the body response to stress. Release of CAs is controlled neurogenically by the activity of preganglionic sympathetic neurons through trains of action potentials (APs). APs in CCs are generated by robust depolarization following the activation of nicotinic and muscarinic receptors that are highly expressed in CCs. Bovine, rat, mouse, and human CCs also express a composite array of Na+, K+, and Ca2+ channels that regulate the resting potential, shape the APs, and set the frequency of AP trains. AP trains of increasing frequency induce enhanced release of CAs. If the primary role of CCs is simply to relay preganglionic nerve commands to CA secretion, why should they express such a diverse set of ion channels? An answer to this comes from recent observations that, like in neurons, CCs undergo complex firing patterns of APs suggesting the existence of an intrinsic CC excitability (non-neurogenically controlled). Recent work has shown that CCs undergo occasional or persistent burst firing elicited by altered physiological conditions or deletion of pore-regulating auxiliary subunits. In this review, we aim to give a rationale to the role of the many ion channel types regulating CC excitability. We will first describe their functional properties and then analyze how they contribute to pacemaking, AP shape, and burst waveforms. We will also furnish clear indications on missing ion conductances that may be involved in pacemaking and highlight the contribution of the crucial channels involved in burst firing.
Collapse
|
7
|
Scott AL, Zhang M, Nurse CA. Enhanced BDNF signalling following chronic hypoxia potentiates catecholamine release from cultured rat adrenal chromaffin cells. J Physiol 2016; 593:3281-99. [PMID: 26095976 DOI: 10.1113/jp270725] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/06/2015] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS We investigated the role of the neurotrophin BDNF signalling via the TrkB receptor in rat adrenomedullary chromaffin cells (AMCs) exposed to normoxia (Nox; 21% O2) and chronic hypoxia (CHox; 2% O2) in vitro for ∼ 48 h. TrkB receptor expression was upregulated in primary AMCs and in immortalized chromaffin (MAH) cells exposed to CHox; this effect was absent in MAH cells deficient in the transcription factor, hypoxia inducible factor (HIF)-2α. Relative to normoxic controls, activation of the TrkB receptor in chronically hypoxic AMCs led to a marked increase in membrane excitability, intracellular [Ca(2+)], and catecholamine secretion. The BDNF-induced rise of intracellular [Ca(2+)] in CHox cells was sensitive to the selective T-type Ca(2+) channel blocker TTA-P2 and tetrodotoxin (TTX), suggesting key roles of low threshold T-type Ca(2+) and voltage-gated Na(+) channels in the signalling pathway. Environmental stressors, including chronic hypoxia, enhance the ability of adrenomedullary chromaffin cells (AMCs) to secrete catecholamines; however, the underlying molecular mechanisms remain unclear. Here, we investigated the role of brain-derived neurotrophic factor (BDNF) signalling in rat AMCs exposed to chronic hypoxia. In rat adrenal glands, BDNF and its tropomyosin-related kinase B (TrkB) receptor are highly expressed in the cortex and medulla, respectively. Exposure of AMCs to chronic hypoxia (2% O2; 48 h) in vitro caused a significant increase to TrkB mRNA expression. A similar increase was observed in an immortalized chromaffin cell line (MAH cells); however, it was absent in MAH cells deficient in the transcription factor HIF-2α. A specific TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF), stimulated quantal catecholamine secretion from chronically hypoxic (CHox; 2% O2) AMCs to a greater extent than normoxic (Nox; 21% O2) controls. Activation of TrkB by BDNF or 7,8-DHF increased intracellular Ca(2+) ([Ca(2+)]i), an effect that was significantly larger in CHox cells. The 7,8-DHF-induced [Ca(2+)]i rise was sensitive to the tyrosine kinase inhibitor K252a and nickel (2 mm), but not the Ca(2+) store-depleting agent cyclopiazonic acid. Blockade of T-type calcium channels with TTA-P2 (1 μm) or voltage-gated Na(+) channels with TTX inhibited BDNF-induced [Ca(2+)]i increases. BDNF also induced a dose-dependent enhancement of action potential firing in CHox cells. These data demonstrate that during chronic hypoxia, enhancement of BDNF-TrkB signalling increases voltage-dependent Ca(2+) influx and catecholamine secretion in chromaffin cells, and that T-type Ca(2+) channels play a key role in the signalling pathway.
Collapse
Affiliation(s)
- Angela L Scott
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Min Zhang
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Colin A Nurse
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| |
Collapse
|
8
|
Gavello D, Vandael D, Gosso S, Carbone E, Carabelli V. Dual action of leptin on rest-firing and stimulated catecholamine release via phosphoinositide 3-kinase-driven BK channel up-regulation in mouse chromaffin cells. J Physiol 2015; 593:4835-53. [PMID: 26282459 DOI: 10.1113/jp271078] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/12/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Leptin is an adipokine produced by the adipose tissue regulating body weight through its appetite-suppressing effect and, as such, exerts a relevant action on the adipo-adrenal axis. Leptin has a dual action on adrenal mouse chromaffin cells both at rest and during stimulation. At rest, the adipokine inhibits the spontaneous firing of most cells by enhancing the probability of BK channel opening through the phosphoinositide 3-kinase signalling cascade. This inhibitory effect is absent in db(-) /db(-) mice deprived of Ob receptors. During sustained stimulation, leptin preserves cell excitability by generating well-adapted action potential (AP) trains of lower frequency and broader width and increases catecholamine secretion by increasing the size of the ready-releasable pool and the rate of vesicle release. In conclusion, leptin dampens AP firing at rest but preserves AP firing and enhances catecholamine release during sustained stimulation, highlighting the importance of the adipo-adrenal axis in the leptin-mediated increase of sympathetic tone and catecholamine release. ABSTRACT Leptin is an adipokine produced by the adipose tissue regulating body weight through its appetite-suppressing effect. Besides being expressed in the hypothalamus and hippocampus, leptin receptors (ObRs) are also present in chromaffin cells of the adrenal medulla. In the present study, we report the effect of leptin on mouse chromaffin cell (MCC) functionality, focusing on cell excitability and catecholamine secretion. Acute application of leptin (1 nm) on spontaneously firing MCCs caused a slowly developing membrane hyperpolarization followed by complete blockade of action potential (AP) firing. This inhibitory effect at rest was abolished by the BK channel blocker paxilline (1 μm), suggesting the involvement of BK potassium channels. Single-channel recordings in 'perforated microvesicles' confirmed that leptin increased BK channel open probability without altering its unitary conductance. BK channel up-regulation was associated with the phosphoinositide 3-kinase (PI3K) signalling cascade because the PI3K specific inhibitor wortmannin (100 nm) fully prevented BK current increase. We also tested the effect of leptin on evoked AP firing and Ca(2+) -driven exocytosis. Although leptin preserves well-adapted AP trains of lower frequency, APs are broader and depolarization-evoked exocytosis is increased as a result of the larger size of the ready-releasable pool and higher frequency of vesicle release. The kinetics and quantal size of single secretory events remained unaltered. Leptin had no effect on firing and secretion in db(-) /db(-) mice lacking the ObR gene, confirming its specificity. In conclusion, leptin exhibits a dual action on MCC activity. It dampens AP firing at rest but preserves AP firing and increases catecholamine secretion during sustained stimulation, highlighting the importance of the adipo-adrenal axis in the leptin-mediated increase of sympathetic tone and catecholamine release.
Collapse
Affiliation(s)
- Daniela Gavello
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,NIS Center, CNISM, University of Torino, Torino, Italy
| | - David Vandael
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,NIS Center, CNISM, University of Torino, Torino, Italy.,Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg, Austria
| | - Sara Gosso
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,NIS Center, CNISM, University of Torino, Torino, Italy
| | - Emilio Carbone
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,NIS Center, CNISM, University of Torino, Torino, Italy
| | - Valentina Carabelli
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,NIS Center, CNISM, University of Torino, Torino, Italy
| |
Collapse
|
9
|
Vandael DHF, Marcantoni A, Carbone E. Cav1.3 Channels as Key Regulators of Neuron-Like Firings and Catecholamine Release in Chromaffin Cells. Curr Mol Pharmacol 2015; 8:149-61. [PMID: 25966692 PMCID: PMC5384372 DOI: 10.2174/1874467208666150507105443] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 01/31/2015] [Accepted: 04/20/2015] [Indexed: 12/19/2022]
Abstract
Neuronal and neuroendocrine L-type calcium channels (Cav1.2, Cav1.3) open readily at relatively low membrane potentials and allow Ca(2+) to enter the cells near resting potentials. In this way, Cav1.2 and Cav1.3 shape the action potential waveform, contribute to gene expression, synaptic plasticity, neuronal differentiation, hormone secretion and pacemaker activity. In the chromaffin cells (CCs) of the adrenal medulla, Cav1.3 is highly expressed and is shown to support most of the pacemaking current that sustains action potential (AP) firings and part of the catecholamine secretion. Cav1.3 forms Ca(2+)-nanodomains with the fast inactivating BK channels and drives the resting SK currents. These latter set the inter-spike interval duration between consecutive spikes during spontaneous firing and the rate of spike adaptation during sustained depolarizations. Cav1.3 plays also a primary role in the switch from "tonic" to "burst" firing that occurs in mouse CCs when either the availability of voltage-gated Na channels (Nav) is reduced or the β2 subunit featuring the fast inactivating BK channels is deleted. Here, we discuss the functional role of these "neuron-like" firing modes in CCs and how Cav1.3 contributes to them. The open issue is to understand how these novel firing patterns are adapted to regulate the quantity of circulating catecholamines during resting condition or in response to acute and chronic stress.
Collapse
Affiliation(s)
| | | | - Emilio Carbone
- Department of Drug Science, Corso Raffaello 30, I - 10125 Torino, Italy.
| |
Collapse
|
10
|
T-type channel-mediated neurotransmitter release. Pflugers Arch 2014; 466:677-87. [PMID: 24595475 DOI: 10.1007/s00424-014-1489-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 02/18/2014] [Indexed: 10/25/2022]
Abstract
Besides controlling a wide variety of cell functions, T-type channels have been shown to regulate neurotransmitter release in peripheral and central synapses and neuroendocrine cells. Growing evidence over the last 10 years suggests a key role of Cav3.2 and Cav3.1 channels in controlling basal neurosecretion near resting conditions and sustained release during mild stimulations. In some cases, the contribution of low-voltage-activated (LVA) channels is not directly evident but requires either the activation of coupled presynaptic receptors, block of ion channels, or chelation of metal ions. Concerning the coupling to the secretory machinery, T-type channels appear loosely coupled to neurotransmitter and hormone release. In neurons, Cav3.2 and Cav3.1 channels mainly control the asynchronous appearance of "minis" [miniature inhibitory postsynaptic currents (mIPSCs) and miniature excitatory postsynaptic currents (mEPSCs)]. The same loose coupling is evident from membrane capacity and amperometric recordings in chromaffin cells and melanotropes where the low-threshold-driven exocytosis possesses the same linear Ca(2+) dependence of the other voltage-gated Ca(2+) channels (Cav1 and Cav2) that is strongly attenuated by slow calcium buffers. The intriguing issue is that, despite not expressing a consensus "synprint" site, Cav3.2 channels do interact with syntaxin 1A and SNAP-25 and, thus, may form nanodomains with secretory vesicles that can be regulated at low voltages. In this review, we discuss all the past and recent issues related to T-type channel-secretion coupling in neurons and neuroendocrine cells.
Collapse
|
11
|
Inhibition of catecholamine secretion by iron-rich and iron-deprived multiwalled carbon nanotubes in chromaffin cells. Neurotoxicology 2013; 39:84-94. [PMID: 23999117 DOI: 10.1016/j.neuro.2013.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 07/23/2013] [Accepted: 08/21/2013] [Indexed: 01/26/2023]
Abstract
The assay of the toxic effects of carbon nanotubes (CNTs) on human health is a stringent need in view of their expected increasing exploitation in industrial and biomedical applications. Most studies so far have been focused on lung toxicity, as the respiratory tract is the main entry of airborne particulate, but there is also recent evidence on the existence of toxic effects of multiwalled carbon nanotubes (MWCNTs) on neuronal and neuroendocrine cells (Belyanskaya et al., 2009; Xu et al., 2009; Gavello et al., 2012). Commercial MWCNTs often contain large amounts of metals deriving from the catalyst used during their synthesis. Since metals, particularly iron, may contribute to the toxicity of MWCNTs, we compared here the effects of two short MWCNTs samples (<5μm length), differing only in their iron content (0.5 versus 0.05% w/w) on the secretory responses of neurotransmitters in mouse chromaffin cells. We found that both iron-rich (MWCNT+Fe) and iron-deprived (MWCNT-Fe) samples enter chromaffin cells after 24h exposure, even though incorporation was attenuated in the latter case (40% versus 78% of cells). As a consequence of MWCNT+Fe or MWCNT-Fe exposure (50-263μg/ml, 24h), catecholamine secretion of chromaffin cells is drastically impaired because of the decreased Ca(2+)-dependence of exocytosis, reduced size of ready-releasable pool and lowered rate of vesicle release. On the contrary, both MWCNTs were ineffective in changing the kinetics of neurotransmitter release of single chromaffin granules and their quantal content. Overall, our data indicate that both MWCNT samples dramatically impair secretion in chromaffin cells, thus uncovering a true depressive action of CNTs mainly associated to their structure and degree of aggregation. This cellular "loss-of-function" is only partially attenuated in iron-deprived samples, suggesting a minor role of iron impurities on MWCNTs toxicity in chromaffin cells exocytosis.
Collapse
|
12
|
Álvarez YD, Belingheri AV, Perez Bay AE, Javis SE, Tedford HW, Zamponi G, Marengo FD. The immediately releasable pool of mouse chromaffin cell vesicles is coupled to P/Q-type calcium channels via the synaptic protein interaction site. PLoS One 2013; 8:e54846. [PMID: 23382986 PMCID: PMC3559834 DOI: 10.1371/journal.pone.0054846] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 12/18/2012] [Indexed: 01/17/2023] Open
Abstract
It is generally accepted that the immediately releasable pool is a group of readily releasable vesicles that are closely associated with voltage dependent Ca2+ channels. We have previously shown that exocytosis of this pool is specifically coupled to P/Q Ca2+ current. Accordingly, in the present work we found that the Ca2+ current flowing through P/Q-type Ca2+ channels is 8 times more effective at inducing exocytosis in response to short stimuli than the current carried by L-type channels. To investigate the mechanism that underlies the coupling between the immediately releasable pool and P/Q-type channels we transiently expressed in mouse chromaffin cells peptides corresponding to the synaptic protein interaction site of Cav2.2 to competitively uncouple P/Q-type channels from the secretory vesicle release complex. This treatment reduced the efficiency of Ca2+ current to induce exocytosis to similar values as direct inhibition of P/Q-type channels via ω-agatoxin-IVA. In addition, the same treatment markedly reduced immediately releasable pool exocytosis, but did not affect the exocytosis provoked by sustained electric or high K+ stimulation. Together, our results indicate that the synaptic protein interaction site is a crucial factor for the establishment of the functional coupling between immediately releasable pool vesicles and P/Q-type Ca2+ channels.
Collapse
Affiliation(s)
- Yanina D. Álvarez
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (CONICET), Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Verónica Belingheri
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (CONICET), Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andrés E. Perez Bay
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (CONICET), Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Scott E. Javis
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - H. William Tedford
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Gerald Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Fernando D. Marengo
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (CONICET), Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
13
|
Vandael DHF, Mahapatra S, Calorio C, Marcantoni A, Carbone E. Cav1.3 and Cav1.2 channels of adrenal chromaffin cells: emerging views on cAMP/cGMP-mediated phosphorylation and role in pacemaking. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1608-18. [PMID: 23159773 DOI: 10.1016/j.bbamem.2012.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 11/05/2012] [Accepted: 11/08/2012] [Indexed: 12/21/2022]
Abstract
Voltage-gated Ca²⁺ channels (VGCCs) are voltage sensors that convert membrane depolarizations into Ca²⁺ signals. In the chromaffin cells of the adrenal medulla, the Ca²⁺ signals driven by VGCCs regulate catecholamine secretion, vesicle retrievals, action potential shape and firing frequency. Among the VGCC-types expressed in these cells (N-, L-, P/Q-, R- and T-types), the two L-type isoforms, Ca(v)1.2 and Ca(v)1.3, control key activities due to their particular activation-inactivation gating and high-density of expression in rodents and humans. The two isoforms are also effectively modulated by G protein-coupled receptor pathways delimited in membrane micro-domains and by the cAMP/PKA and NO/cGMP/PKG phosphorylation pathways which induce prominent Ca²⁺ current changes if opposingly regulated. The two L-type isoforms shape the action potential and directly participate to vesicle exocytosis and endocytosis. The low-threshold of activation and slow rate of inactivation of Ca(v)1.3 confer to this channel the unique property of carrying sufficient inward current at subthreshold potentials able to activate BK and SK channels which set the resting potential, the action potential shape, the cell firing mode and the degree of spike frequency adaptation during spontaneous firing or sustained depolarizations. These properties help chromaffin cells to optimally adapt when switching from normal to stress-mimicking conditions. Here, we will review past and recent findings on cAMP- and cGMP-mediated modulations of Ca(v)1.2 and Ca(v)1.3 and the role that these channels play in the control of chromaffin cell firing. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- D H F Vandael
- Department of Drug Science, Laboratory of Cellular & Molecular Neuroscience, NIS Center, CNISM, University of Torino, Italy
| | | | | | | | | |
Collapse
|
14
|
Zamponi GW, Currie KPM. Regulation of Ca(V)2 calcium channels by G protein coupled receptors. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1629-43. [PMID: 23063655 DOI: 10.1016/j.bbamem.2012.10.004] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 10/02/2012] [Accepted: 10/04/2012] [Indexed: 12/29/2022]
Abstract
Voltage gated calcium channels (Ca²⁺ channels) are key mediators of depolarization induced calcium influx into excitable cells, and thereby play pivotal roles in a wide array of physiological responses. This review focuses on the inhibition of Ca(V)2 (N- and P/Q-type) Ca²⁺-channels by G protein coupled receptors (GPCRs), which exerts important autocrine/paracrine control over synaptic transmission and neuroendocrine secretion. Voltage-dependent inhibition is the most widespread mechanism, and involves direct binding of the G protein βγ dimer (Gβγ) to the α1 subunit of Ca(V)2 channels. GPCRs can also recruit several other distinct mechanisms including phosphorylation, lipid signaling pathways, and channel trafficking that result in voltage-independent inhibition. Current knowledge of Gβγ-mediated inhibition is reviewed, including the molecular interactions involved, determinants of voltage-dependence, and crosstalk with other cell signaling pathways. A summary of recent developments in understanding the voltage-independent mechanisms prominent in sympathetic and sensory neurons is also included. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, Canada
| | | |
Collapse
|
15
|
Control of low-threshold exocytosis by T-type calcium channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1579-86. [PMID: 22885170 DOI: 10.1016/j.bbamem.2012.07.031] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 07/30/2012] [Accepted: 07/30/2012] [Indexed: 12/11/2022]
Abstract
Low-voltage-activated (LVA) T-type Ca²⁺ channels differ from their high-voltage-activated (HVA) homologues by unique biophysical properties. Hence, whereas HVA channels convert action potentials into intracellular Ca²⁺ elevations, T-type channels control Ca²⁺ entry during small depolarizations around the resting membrane potential. They play an important role in electrical activities by generating low-threshold burst discharges that occur during various physiological and pathological forms of neuronal rhythmogenesis. In addition, they mediate a previously unrecognized function in the control of synaptic transmission where they directly trigger the release of neurotransmitters at rest. In this review, we summarize our present knowledge of the role of T-type Ca²⁺ channels in vesicular exocytosis, and emphasize the critical importance of localizing the exocytosis machinery close to the Ca²⁺ source for reliable synaptic transmission. This article is part of a Special Issue entitled: Calcium channels.
Collapse
|
16
|
Rosa JM, Nanclares C, Orozco A, Colmena I, de Pascual R, García AG, Gandía L. Regulation by L-Type Calcium Channels of Endocytosis: An Overview. J Mol Neurosci 2012; 48:360-7. [DOI: 10.1007/s12031-012-9786-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 04/22/2012] [Indexed: 11/29/2022]
|
17
|
Calcium channel types contributing to chromaffin cell excitability, exocytosis and endocytosis. Cell Calcium 2012; 51:321-30. [DOI: 10.1016/j.ceca.2012.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 01/10/2012] [Accepted: 01/12/2012] [Indexed: 11/18/2022]
|
18
|
|
19
|
Functional chromaffin cell plasticity in response to stress: focus on nicotinic, gap junction, and voltage-gated Ca2+ channels. J Mol Neurosci 2012; 48:368-86. [PMID: 22252244 DOI: 10.1007/s12031-012-9707-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 01/04/2012] [Indexed: 10/14/2022]
Abstract
An increase in circulating catecholamines constitutes one of the mechanisms whereby human body responds to stress. In response to chronic stressful situations, the adrenal medullary tissue exhibits crucial morphological and functional changes that are consistent with an improvement of chromaffin cell stimulus-secretion coupling efficiency. Stimulus-secretion coupling encompasses multiple intracellular (chromaffin cell excitability, Ca(2+) signaling, exocytosis, endocytosis) and intercellular pathways (splanchnic nerve-mediated synaptic transmission, paracrine and endocrine communication, gap junctional coupling), each of them being potentially subjected to functional remodeling upon stress. This review focuses on three chromaffin cell incontrovertible actors, the cholinergic nicotinic receptors and the voltage-dependent T-type Ca(2+) channels that are directly involved in Ca(2+)-dependent events controlling catecholamine secretion and electrical activity, and the gap junctional communication involved in the modulation of catecholamine secretion. We show here that these three actors react differently to various stressors, sometimes independently, sometimes in concert or in opposition.
Collapse
|
20
|
Hill J, Chan SA, Kuri B, Smith C. Pituitary adenylate cyclase-activating peptide (PACAP) recruits low voltage-activated T-type calcium influx under acute sympathetic stimulation in mouse adrenal chromaffin cells. J Biol Chem 2011; 286:42459-42469. [PMID: 22009744 PMCID: PMC3234986 DOI: 10.1074/jbc.m111.289389] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 10/17/2011] [Indexed: 02/02/2023] Open
Abstract
Low voltage-activated T-type Ca(v)3.2 calcium channels are expressed in neurosecretory chromaffin cells of the adrenal medulla. Previous studies have shown that naïve adrenal chromaffin cells express a nominal Ca(v)3.2-dependent conductance. However, Ca(v)3.2 conductance is up-regulated following chronic hypoxia or long term exposure to cAMP analogs. Thus, although a link between chronic stressors and up-regulation of Ca(v)3.2 exists, there are no reports testing the specific role of Ca(v)3.2 channels in the acute sympathoadrenal stress response. In this study, we examined the effects of acute sympathetic stress on T-type Ca(v)3.2 calcium influx in mouse chromaffin cells in situ. Pituitary adenylate cyclase-activating peptide (PACAP) is an excitatory neuroactive peptide transmitter released by the splanchnic nerve under elevated sympathetic activity to stimulate the adrenal medulla. PACAP stimulation did not evoke action potential firing in chromaffin cells but did cause a persistent subthreshold membrane depolarization that resulted in an immediate and robust Ca(2+)-dependent catecholamine secretion. Moreover, PACAP-evoked secretion was sensitive to block by nickel chloride and was acutely inhibited by protein kinase C blockers. We utilized perforated patch electrophysiological recordings conducted in adrenal tissue slices to investigate the mechanism of PACAP-evoked calcium entry. We provide evidence that stimulation with exogenous PACAP and native neuronal stress stimulation both lead to a protein kinase C-mediated phosphodependent recruitment of a T-type Ca(v)3.2 Ca(2+) influx. This in turn evokes catecholamine release during the acute sympathetic stress response.
Collapse
Affiliation(s)
- Jacqueline Hill
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Shyue-An Chan
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Barbara Kuri
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Corey Smith
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
21
|
Rogers GJ, Tolhurst G, Ramzan A, Habib AM, Parker HE, Gribble FM, Reimann F. Electrical activity-triggered glucagon-like peptide-1 secretion from primary murine L-cells. J Physiol 2011; 589:1081-93. [PMID: 21224236 PMCID: PMC3060588 DOI: 10.1113/jphysiol.2010.198069] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 12/31/2010] [Indexed: 01/21/2023] Open
Abstract
Glucagon like peptide 1 (GLP-1) based therapies are now widely used for the treatment of type 2 diabetes. Developing our understanding of intestinal GLP-1 release may facilitate the development of new therapeutics aimed at targeting the GLP-1 producing L-cells. This study was undertaken to characterise the electrical activity of primary L-cells and the importance of voltage gated sodium and calcium channels for GLP-1 secretion. Primary murine L-cells were identified and purified using transgenic mice expressing a fluorescent protein driven by the proglucagon promoter. Fluorescent L-cells were identified within primary colonic cultures for patch clamp recordings. GLP-1 secretion was measured from primary colonic cultures. L-cells purified by flow cytometry were used to measure gene expression by microarray and quantitative RT-PCR. Electrical activity in L-cells was due to large voltage gated sodium currents, inhibition of which by tetrodotoxin reduced both basal and glutamine-stimulated GLP-1 secretion. Voltage gated calcium channels were predominantly of the L-type, Q-type and T-type, by expression analysis, consistent with the finding that GLP-1 release was blocked both by nifedipine and ω-conotoxin MVIIC. We observed large voltage-dependent potassium currents, but only a small chromanol sensitive current that might be attributable to KCNQ1. GLP-1 release from primary L-cells is linked to electrical activity and activation of L-type and Q-type calcium currents. The concept of an electrically excitable L-cell provides a basis for understanding how GLP-1 release may be modulated by nutrient, hormonal and pharmaceutical stimuli.
Collapse
Affiliation(s)
- G J Rogers
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Addenbrooke's Hospital, Box 139, Hills Road, Cambridge CB2 0XY, UK
| | | | | | | | | | | | | |
Collapse
|
22
|
Karmažínová M, Baumgart JP, Perez-Reyes E, Lacinová L. The voltage dependence of gating currents of the neuronal CA(v)3.3 channel is determined by the gating brake in the I-II loop. Pflugers Arch 2011; 461:461-8. [PMID: 21340458 DOI: 10.1007/s00424-011-0937-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 02/06/2011] [Accepted: 02/08/2011] [Indexed: 01/10/2023]
Abstract
Low-voltage-activated Ca(v)3 Ca(2+) channels have an activation threshold around -60 mV, which is lower than the activation threshold of other voltage-dependent calcium channels (VDCCs). The kinetics of their activation at membrane voltages just above the activation threshold is much slower than the activation kinetics of other VDCCs. It was demonstrated recently that the intracellular loop connecting repeats I and II of all three Ca(v)3 channels contains a so-called gating brake. Disruption of this brake yields channels that activate at even more hyperpolarized potentials with significantly accelerated kinetics. We have compared gating of a wild-type Ca(v)3.3 channel and a mutated ID12 channel, in which the putative gating brake at the proximal part of the I-II loop was removed. Voltage dependence of the gating current activation was shifted by 34.6 mV towards more hyperpolarized potentials in ID12 channel. ON-charge movement was significantly faster in the ID12 channel, while the kinetics of the off-charge was not altered by the mutation. We conclude that the putative gating brake in I-II loop hinders not only the opening of the conducting pore but also the activating movement of voltage-sensing S4 segments, stabilizing the channel in its closed state.
Collapse
Affiliation(s)
- Mária Karmažínová
- Institute of Molecular Physiology and Genetics, Centre of Excellence for Cardiovascular Research, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | | | | |
Collapse
|
23
|
Álvarez YD, Marengo FD. The immediately releasable vesicle pool: highly coupled secretion in chromaffin and other neuroendocrine cells. J Neurochem 2010; 116:155-63. [DOI: 10.1111/j.1471-4159.2010.07108.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
24
|
Zhang Y, Kolli T, Hivley R, Jaber L, Zhao FI, Yan J, Herness S. Characterization of the expression pattern of adrenergic receptors in rat taste buds. Neuroscience 2010; 169:1421-37. [PMID: 20478367 DOI: 10.1016/j.neuroscience.2010.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 05/03/2010] [Accepted: 05/11/2010] [Indexed: 12/01/2022]
Abstract
Taste buds signal the presence of chemical stimuli in the oral cavity to the central nervous system using both early transduction mechanisms, which allow single cells to be depolarized via receptor-mediated signaling pathways, and late transduction mechanisms, which involve extensive cell-to-cell communication among the cells in the bud. The latter mechanisms, which involve a large number of neurotransmitters and neuropeptides, are less well understood. Among neurotransmitters, multiple lines of evidence suggest that norepinephrine plays a yet unknown role in the taste bud. This study investigated the expression pattern of adrenergic receptors in the rat posterior taste bud. Expression of alpha1A, alpha1B, alpha1D, alpha2A, alpha2B, alpha2C, beta1, and the beta2 adrenoceptor subtypes was observed in taste buds using RT-PCR and immunocytochemical techniques. Taste buds also expressed the biosynthetic enzyme for norepinephrine, dopamine beta-hydroxylase (DbetaH), as well as the norepinephrine transporter. Further, expression of the epinephrine synthetic enzyme, phenylethanolamine N-methyltransferase (PNMT), was observed suggesting a possible role for this transmitter in the bud. Phenotyping adrenoceptor expression patterns with double labeling experiments to gustducin, synaptosomal-associated protein 25 (SNAP-25), and neural cell adhesion molecule (NCAM) suggests they are prominently expressed in subsets of cells known to express taste receptor molecules but segregated from cells known to have synapses with the afferent nerve fiber. Alpha and beta adrenoceptors co-express with one another in unique patterns as observed with immunocytochemistry and single cell reverse transcription polymerase chain reaction (RT-PCR). These data suggest that single cells express multiple adrenergic receptors and that adrenergic signaling may be particularly important in bitter, sweet, and umami taste qualities. In summary, adrenergic signaling in the taste bud occurs through complex pathways that include presynaptic and postsynaptic receptors and likely play modulatory roles in processing of gustatory information similar to other peripheral sensory systems such as the retina, cochlea, and olfactory bulb.
Collapse
Affiliation(s)
- Y Zhang
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, School of Medicine, Xi'an Jiaotong University, 76# West Yanta Road, Xi'an 710061, PR China
| | | | | | | | | | | | | |
Collapse
|
25
|
Carbone E, Carabelli V. O2 sensing in chromaffin cells: new duties for T-type channels. J Physiol 2009; 587:1859-60. [PMID: 19406883 DOI: 10.1113/jphysiol.2009.172197] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
26
|
Levitsky KL, López-Barneo J. Developmental change of T-type Ca2+ channel expression and its role in rat chromaffin cell responsiveness to acute hypoxia. J Physiol 2009; 587:1917-29. [PMID: 19273573 DOI: 10.1113/jphysiol.2009.168989] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neonatal chromaffin cells of the adrenal medulla (AM) are intrinsic chemoreceptors that secrete catecholamines in response to hypoxia, thus contributing to fetal adaptation to extrauterine life. In most mammals studied, oxygen sensitivity of AM cells disappears a few days after birth, possibly due to innervation of the adrenal gland by the cholinergic fibres of the splanchnic nerve (approximately postnatal day 7 in the rat). The mechanisms underlying these homeostatic changes in chromaffin cells are unknown. Low voltage-activated, T-type, Ca(2+) channels regulate cell excitability and their expression is up-regulated by hypoxia. Hence, we hypothesized that these channels contribute to the developmental changes in the chemoreceptive properties of AM chromaffin cells. Using electrophysiological, immunocytochemical and molecular biology methodologies we show here that neonatal AM chromaffin cells express T-type Ca(2+) channels (of alpha1H or Ca(v)3.2 sub-type) and that the function of these channels is necessary for catecholamine release in response to acute hypoxia. T-type Ca(2+) channel expression, as well as chromaffin cell responsiveness to hypoxia, decrease with postnatal maturation. Adult chromaffin cell sensitivity to hypoxia reappears after AM denervation in parallel with the recruitment of T-type Ca(2+) channels. These observations indicate that T-type Ca(2+) channels are essential for the acute response of chromaffin cells to hypoxia and help explain the disappearance of O(2) sensitivity in adult AM chromaffin cells. Our results may also be relevant for understanding the pathogenesis of disorders associated with chronic hypoxia or maternal nicotine consumption.
Collapse
Affiliation(s)
- Konstantin L Levitsky
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Sevilla, Spain
| | | |
Collapse
|
27
|
Borland G, Smith BO, Yarwood SJ. EPAC proteins transduce diverse cellular actions of cAMP. Br J Pharmacol 2009; 158:70-86. [PMID: 19210747 DOI: 10.1111/j.1476-5381.2008.00087.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
It has now been over 10 years since efforts to completely understand the signalling actions of cAMP (3'-5'-cyclic adenosine monophosphate) led to the discovery of exchange protein directly activated by cAMP (EPAC) proteins. In the current review we will highlight important advances in the understanding of EPAC structure and function and demonstrate that EPAC proteins mediate multiple actions of cAMP in cells, revealing future targets for pharmaceutical intervention. It has been known for some time that drugs that elevate intracellular cAMP levels have proven therapeutic benefit for diseases ranging from depression to inflammation. The challenge now is to determine which of these positive actions of cAMP involve activation of EPAC-regulated signal transduction pathways. EPACs are specific guanine nucleotide exchange factors for the Ras GTPase homologues, Rap1 and Rap2, which they activate independently of the classical routes for cAMP signalling, cyclic nucleotide-gated ion channels and protein kinase A. Rather, EPAC activation is triggered by internal conformational changes induced by direct interaction with cAMP. Leading from this has been the development of EPAC-specific agonists, which has helped to delineate numerous cellular actions of cAMP that rely on subsequent activation of EPAC. These include regulation of exocytosis and the control of cell adhesion, growth, division and differentiation. Recent work also implicates EPAC in the regulation of anti-inflammatory signalling in the vascular endothelium, namely negative regulation of pro-inflammatory cytokine signalling and positive support of barrier function. Further elucidation of these important signalling mechanisms will no doubt support the development of the next generation of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Gillian Borland
- Division of Molecular and Cellular Biology, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | |
Collapse
|
28
|
Cueni L, Canepari M, Adelman JP, Lüthi A. Ca2+ signaling by T-type Ca2+ channels in neurons. Pflugers Arch 2008; 457:1161-72. [DOI: 10.1007/s00424-008-0582-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 08/15/2008] [Indexed: 10/21/2022]
|
29
|
PDE type-4 inhibition increases L-type Ca2+ currents, action potential firing, and quantal size of exocytosis in mouse chromaffin cells. Pflugers Arch 2008; 457:1093-110. [DOI: 10.1007/s00424-008-0584-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 08/22/2008] [Indexed: 01/21/2023]
|
30
|
Cheng X, Ji Z, Tsalkova T, Mei F. Epac and PKA: a tale of two intracellular cAMP receptors. Acta Biochim Biophys Sin (Shanghai) 2008; 40:651-62. [PMID: 18604457 PMCID: PMC2630796 DOI: 10.1111/j.1745-7270.2008.00438.x] [Citation(s) in RCA: 290] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
cAMP-mediated signaling pathways regulate a multitude of important biological processes under both physiological and pathological conditions, including diabetes, heart failure and cancer. In eukaryotic cells, the effects of cAMP are mediated by two ubiquitously expressed intracellular cAMP receptors, the classic protein kinase A (PKA)/cAMP-dependent protein kinase and the recently discovered exchange protein directly activated by camp (Epac)/cAMP-regulated guanine nucleotide exchange factors. Like PKA, Epac contains an evolutionally conserved cAMP binding domain that acts as a molecular switch for sensing intracellular second messenger cAMP levels to control diverse biological functions. The existence of two families of cAMP effectors provides a mechanism for a more precise and integrated control of the cAMP signaling pathways in a spatial and temporal manner. Depending upon the specific cellular environments as well as their relative abundance, distribution and localization, Epac and PKA may act independently, converge synergistically or oppose each other in regulating a specific cellular function.
Collapse
Affiliation(s)
- Xiaodong Cheng
- Department of Pharmacology and Toxicology, Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-1031, USA.
| | | | | | | |
Collapse
|
31
|
Marcantoni A, Carabelli V, Comunanza V, Hoddah H, Carbone E. Calcium channels in chromaffin cells: focus on L and T types. Acta Physiol (Oxf) 2008; 192:233-46. [PMID: 18021322 DOI: 10.1111/j.1748-1716.2007.01815.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Voltage-gated Ca2+ channels (Cav) are highly expressed in the adrenal chromaffin cells of mammalian species. Besides shaping action potential waveforms, they are directly involved in the excitation-secretion coupling underlying catecholamine release and, possibly, control other Ca2+-dependent events that originate near the membrane. These functions are shared by a number of Cav channel types (L, N, P/Q, R and T) which have different structure-function characteristics and whose degree of expression changes remarkably among mammalian species. Understanding precisely the functioning of each voltage-gated Ca2+ channels is a crucial task that helps clarifying the Ca2+-dependent mechanisms controlling exocytosis during physiological and pathological conditions. In this paper, we focus on classical and new roles that L- and T-type channels play in the control of chromaffin cell excitability and neurotransmitter release. Interestingly, L-type channels are shown to be implicated in the spontaneous autorhythmicity of chromaffin cells, while T-type channels, which are absent in adult chromaffin cells, are coupled with secretion and can be recruited following long-term beta-adrenergic stimulation or chronic hypoxia. This suggests that like other cells, adrenal chromaffin cells undergo effective remodelling of membrane ion channels and cell functioning during prolonged stress conditions.
Collapse
Affiliation(s)
- A Marcantoni
- Department of Neuroscience, NIS Centre of Excellence, CNISM Research Unit, Torino, Italy
| | | | | | | | | |
Collapse
|
32
|
Gackière F, Bidaux G, Delcourt P, Van Coppenolle F, Katsogiannou M, Dewailly E, Bavencoffe A, Van Chuoï-Mariot MT, Mauroy B, Prevarskaya N, Mariot P. CaV3.2 T-type calcium channels are involved in calcium-dependent secretion of neuroendocrine prostate cancer cells. J Biol Chem 2008; 283:10162-73. [PMID: 18230611 DOI: 10.1074/jbc.m707159200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Because prostate cancer is, in its early stages, an androgen-dependent pathology, treatments aiming at decreasing testosterone plasma concentration have been developed for many years now. However, a significant proportion of patients suffer a relapse after a few years of hormone therapy. The androgen-independent stage of prostate cancer has been shown to be associated with the development of neuroendocrine differentiation. We previously demonstrated that neuroendocrine prostate cancer cells derived from LNCaP cells overexpress CaV3.2 T-type voltage-dependent calcium channels. We demonstrate here using prostatic acid phosphatase as a marker of prostate secretion and FM1-43 fluorescence imaging of membrane trafficking that neuroendocrine differentiation is associated with an increase in calcium-dependent secretion which critically relies on CaV3.2 T-type calcium channel activity. In addition, we show that these channels are expressed by neuroendocrine cells in prostate cancer tissues obtained from patients after surgery. We propose that CaV3.2 T-type calcium channel up-regulation may account for the alteration of secretion during prostate cancer development and that these channels, by promoting the secretion of potential mitogenic factors, could participate in the progression of the disease toward an androgen-independent stage.
Collapse
Affiliation(s)
- Florian Gackière
- INSERM U800, Laboratoire de Physiologie Cellulaire, Equipe Labellisée par la Ligue contre le Cancer and Université des Sciences et Technologies de Lille, Villeneuve d'Ascq, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Carabelli V, Marcantoni A, Comunanza V, de Luca A, Díaz J, Borges R, Carbone E. Chronic hypoxia up-regulates alpha1H T-type channels and low-threshold catecholamine secretion in rat chromaffin cells. J Physiol 2007; 584:149-65. [PMID: 17690152 PMCID: PMC2277059 DOI: 10.1113/jphysiol.2007.132274] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Accepted: 08/03/2007] [Indexed: 12/24/2022] Open
Abstract
alpha(1H) T-type channels recruited by beta(1)-adrenergic stimulation in rat chromaffin cells (RCCs) are coupled to fast exocytosis with the same Ca(2+) dependence of high-threshold Ca(2+) channels. Here we show that RCCs exposed to chronic hypoxia (CH) for 12-18 h in 3% O(2) express comparable densities of functional T-type channels that depolarize the resting cells and contribute to low-voltage exocytosis. Following chronic hypoxia, most RCCs exhibited T-type Ca(2+) channels already available at -50 mV with the same gating, pharmacological and molecular features as the alpha(1H) isoform. Chronic hypoxia had no effects on cell size and high-threshold Ca(2+) current density and was mimicked by overnight incubation with the iron-chelating agent desferrioxamine (DFX), suggesting the involvement of hypoxia-inducible factors (HIFs). T-type channel recruitment occurred independently of PKA activation and the presence of extracellular Ca(2+). Hypoxia-recruited T-type channels were partially open at rest (T-type 'window-current') and contributed to raising the resting potential to more positive values. Their block by 50 microm Ni(2+) caused a 5-8 mV hyperpolarization. The secretory response associated with T-type channels could be detected following mild cell depolarizations, either by capacitance increases induced by step depolarizations or by amperometric current spikes induced by increased [KCl]. In the latter case, exocytotic bursts could be evoked even with 2-4 mm KCl and spike frequency was drastically reduced by 50 microm Ni(2+). Chronic hypoxia did not alter the shape of spikes, suggesting that hypoxia-recruited T-type channels increase the number of secreted vesicles at low voltages, without altering the mechanism of catecholamine release and the quantal content of released molecules.
Collapse
Affiliation(s)
- V Carabelli
- Department of Neuroscience, NIS Center of Excellence, CNISM Research Unit, 10125 Torino, Italy
| | | | | | | | | | | | | |
Collapse
|
34
|
Carabelli V, Marcantoni A, Comunanza V, Carbone E. Fast exocytosis mediated by T- and L-type channels in chromaffin cells: distinct voltage-dependence but similar Ca2+ -dependence. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2007; 36:753-62. [PMID: 17340096 DOI: 10.1007/s00249-007-0138-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 01/11/2007] [Accepted: 01/27/2007] [Indexed: 12/01/2022]
Abstract
Expression, spatial distribution and specific roles of different Ca(2+) channels in stimulus-secretion coupling of chromaffin cells are intriguing issues still open to discussion. Most of the evidence supports a role of high-voltage activated (HVA) Ca(2+) channels (L-, N-, P/Q- and R-types) in the control of exocytosis: some suggesting a preferential coupling of specific Ca(2+) channel subunits with the secretory apparatus, others favoring the idea of a contribution to secretion proportional to the expression density and gating properties of Ca(2+) channels. In this work we review recent findings and bring new evidence in favor of the hypothesis that also the LVA (low-voltage-activated, T-type) Ca(2+) channels effectively control fast exocytosis near resting potential in adrenal chromaffin cells of adult rats. T-type channels recruited after long-term treatments with pCPT-cAMP (or chronic hypoxia) are shown to control exocytosis with the same efficacy of L-type channels, which are the dominant Ca(2+) channel types expressed in rodent chromaffin cells. A rigorous comparison of T- and L-type channel properties shows that, although operating at different potentials and with different voltage-sensitivity, the two channels possess otherwise similar Ca(2+)-dependence of exocytosis, size and kinetics of depletion of the immediately releasable pool and mobilize vesicles of the same quantal size. Thus, T- and L-type channels are coupled with the same Ca(2+)-efficiency to the secretory apparatus and deplete the same number of vesicles ready for release. The major difference of the secretory signals controlled by the two channels appear to be the voltage range of operation, suggesting the idea that stressful conditions (hypoxia and persistent beta-adrenergic stimulation) can lower the threshold of cell excitability by recruiting new Ca(2+) channels and activate an additional source of catecholamine secretion.
Collapse
Affiliation(s)
- V Carabelli
- Department of Neuroscience, Centre of Excellence NIS, CNISM UdR, Corso Raffaello 30, Turin, Italy.
| | | | | | | |
Collapse
|
35
|
Tang KS, Wang N, Tse A, Tse FW. Influence of quantal size and cAMP on the kinetics of quantal catecholamine release from rat chromaffin cells. Biophys J 2007; 92:2735-46. [PMID: 17237205 PMCID: PMC1831689 DOI: 10.1529/biophysj.106.088997] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Using carbon fiber amperometry, we exploited the natural variation in quantal size (Q) among individual granules in rat chromaffin cells to examine the influence of Q on quantal release kinetics. Although it is generally accepted that granules with larger Q have slower kinetics of release, we found that this trend was applicable only to granules with Q(1/3) < 0.6 pC(1/3). Granules with larger Q adapted specific mechanisms to maintain a rapid kinetic of release. The semistable fusion pores in the large-Q granules persisted for a longer duration and could reach a bigger size before the onset of very rapid dilation to allow a longer and larger foot signal. Most importantly, a large proportion of large-Q granules maintained a relatively short half-width in the main spike. This suggests that the most rapid phase of fusion pore dilation in many large-Q granules may be faster than that in small-Q granules. Moreover, cAMP selectively advanced the onset of the rapid dilation of the fusion pore in the large- but not the small-Q granules. Thus, our finding raises the possibility that fusion pore and/or granule matrix in small- and large-Q granules may have different molecular structures.
Collapse
Affiliation(s)
- Kim San Tang
- Department of Pharmacology and Centre for Neuroscience, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
36
|
García AG, García-De-Diego AM, Gandía L, Borges R, García-Sancho J. Calcium Signaling and Exocytosis in Adrenal Chromaffin Cells. Physiol Rev 2006; 86:1093-131. [PMID: 17015485 DOI: 10.1152/physrev.00039.2005] [Citation(s) in RCA: 259] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
At a given cytosolic domain of a chromaffin cell, the rate and amplitude of the Ca2+concentration ([Ca2+]c) depends on at least four efficient regulatory systems: 1) plasmalemmal calcium channels, 2) endoplasmic reticulum, 3) mitochondria, and 4) chromaffin vesicles. Different mammalian species express different levels of the L, N, P/Q, and R subtypes of high-voltage-activated calcium channels; in bovine and humans, P/Q channels predominate, whereas in felines and murine species, L-type channels predominate. The calcium channels in chromaffin cells are regulated by G proteins coupled to purinergic and opiate receptors, as well as by voltage and the local changes of [Ca2+]c. Chromaffin cells have been particularly useful in studying calcium channel current autoregulation by materials coreleased with catecholamines, such as ATP and opiates. Depending on the preparation (cultured cells, adrenal slices) and the stimulation pattern (action potentials, depolarizing pulses, high K+, acetylcholine), the role of each calcium channel in controlling catecholamine release can change drastically. Targeted aequorin and confocal microscopy shows that Ca2+entry through calcium channels can refill the endoplasmic reticulum (ER) to nearly millimolar concentrations, and causes the release of Ca2+(CICR). Depending on its degree of filling, the ER may act as a sink or source of Ca2+that modulates catecholamine release. Targeted aequorins with different Ca2+affinities show that mitochondria undergo surprisingly rapid millimolar Ca2+transients, upon stimulation of chromaffin cells with ACh, high K+, or caffeine. Physiological stimuli generate [Ca2+]cmicrodomains in which the local subplasmalemmal [Ca2+]crises abruptly from 0.1 to ∼50 μM, triggering CICR, mitochondrial Ca2+uptake, and exocytosis at nearby secretory active sites. The fact that protonophores abolish mitochondrial Ca2+uptake, and increase catecholamine release three- to fivefold, support the earlier observation. This increase is probably due to acceleration of vesicle transport from a reserve pool to a ready-release vesicle pool; this transport might be controlled by Ca2+redistribution to the cytoskeleton, through CICR, and/or mitochondrial Ca2+release. We propose that chromaffin cells have developed functional triads that are formed by calcium channels, the ER, and the mitochondria and locally control the [Ca2+]cthat regulate the early and late steps of exocytosis.
Collapse
Affiliation(s)
- Antonio G García
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, and Servicio de Farmacología Clínica e Instituto Universitario de Investigación Gerontológica y Metabólica, Hospital Universitario de la Princesa, Madrid, Spain.
| | | | | | | | | |
Collapse
|
37
|
Kim JA, Park JY, Kang HW, Huh SU, Jeong SW, Lee JH. Augmentation of Cav3.2 T-type calcium channel activity by cAMP-dependent protein kinase A. J Pharmacol Exp Ther 2006; 318:230-7. [PMID: 16569752 DOI: 10.1124/jpet.106.101402] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ca2+ influx through T-type Ca2+ channels is crucial for important physiological activities such as hormone secretion and neuronal excitability. However, it is not clear whether these channels are regulated by cAMP-dependent protein kinase A (PKA). In the present study, we examined whether PKA modulates Cav3.2 T-type channels reconstituted in Xenopus oocytes. Application of 10 microM forskolin, an adenylyl cyclase stimulant, increased Cav3.2 channel activity by 40+/-4% over 30 min and negatively shifted the steady-state inactivation curve (V50=-61.4+/-0.2 versus -65.5+/-0.1 mV). Forskolin did not affect other biophysical properties of Cav3.2 channels, including activation curve, current kinetics, and recovery from inactivation. Similar stimulation was achieved by applying 200 microM 8-bromo-cAMP, a membrane-permeable cAMP analog. The augmentation of Cav3.2 channel activity by forskolin was strongly inhibited by preincubation with 20 microM N-[2-(4-bromocinnamylamino)ethyl]-5-isoquinoline (H89), and reversed by subsequent application of 500 nM protein kinase A inhibitor peptide. The stimulation of Cav3.2 channel activity by PKA was mimicked by serotonin when 5HT7 receptor was coexpressed with Cav3.2 in Xenopus oocytes. Finally, using chimeric channels constructed by replacing individual cytoplasmic loops of Cav3.2 with those of the Nav1.4 channel, which is insensitive to PKA, we localized a region required for the PKA-mediated augmentation to the II-III loop of the Cav3.2.
Collapse
Affiliation(s)
- Jin-Ah Kim
- Department of Life Science, Sogang University, Mapo-Gu, Sinsu-Dong 1, Seoul 121-742, Korea
| | | | | | | | | | | |
Collapse
|
38
|
Carbone E, Marcantoni A, Giancippoli A, Guido D, Carabelli V. T-type channels-secretion coupling: evidence for a fast low-threshold exocytosis. Pflugers Arch 2006; 453:373-83. [PMID: 16758226 DOI: 10.1007/s00424-006-0100-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Accepted: 05/29/2006] [Indexed: 10/24/2022]
Abstract
T-type channels are transient low-voltage-activated (LVA) Ca(2+) channels that control Ca(2+) entry in excitable cells during small depolarizations around resting potential. Studies in the past 20 years focused on the biophysical, physiological, and molecular characterization of T-type channels in most tissues. This led to a well-defined picture of the functional role of LVA channels in controlling low-threshold spikes, oscillatory cell activity, muscle contraction, hormone release, cell growth and differentiation. So far, little attention has been devoted to the role of T-type channels in transmitter release, which mainly involves channel types belonging to the high-voltage-activated (HVA) Ca(2+) channel family. However, evidence is accumulating in favor of a unique participation of T-type channels in fast transmitter release. Clear data are now reported in reciprocal synapses of the retina and olfactory bulb, synaptic contacts between primary afferent and second order nociceptive neurons, rhythmic inhibitory interneurons of invertebrates and clonal cell lines transfected with recombinant alpha(1) channel subunits. T-type channels also regulate the large dense-core vesicle release of neuroendocrine cells where Ca(2+) dependence, rate of vesicle release, and size of readily releasable pool appear comparable to those associated to HVA channels. This suggests that when sufficiently expressed and properly located near the release zones, T-type channels can trigger fast low-threshold secretion. In this study, we will review the main findings that assign a specific task to T-type channels in fast exocytosis, discussing their possible involvement in the control of the Ca(2+)-dependent processes regulating exocytosis like vesicle depletion and vesicle recycling.
Collapse
Affiliation(s)
- E Carbone
- Department of Neuroscience, NIS Center of Excellence, CNISM Research Unit, Torino, 10125, Italy.
| | | | | | | | | |
Collapse
|