1
|
Abstract
G protein-coupled receptors (GPCRs) are critical cellular sensors that mediate numerous physiological processes. In the heart, multiple GPCRs are expressed on various cell types, where they coordinate to regulate cardiac function by modulating critical processes such as contractility and blood flow. Under pathological settings, these receptors undergo aberrant changes in expression levels, localization and capacity to couple to downstream signalling pathways. Conventional therapies for heart failure work by targeting GPCRs, such as β-adrenergic receptor and angiotensin II receptor antagonists. Although these treatments have improved patient survival, heart failure remains one of the leading causes of mortality worldwide. GPCR kinases (GRKs) are responsible for GPCR phosphorylation and, therefore, desensitization and downregulation of GPCRs. In this Review, we discuss the GPCR signalling pathways and the GRKs involved in the pathophysiology of heart disease. Given that increased expression and activity of GRK2 and GRK5 contribute to the loss of contractile reserve in the stressed and failing heart, inhibition of overactive GRKs has been proposed as a novel therapeutic approach to treat heart failure.
Collapse
|
2
|
Age-related homeostatic midchannel proteolysis of neuronal L-type voltage-gated Ca²⁺ channels. Neuron 2014; 82:1045-57. [PMID: 24908485 DOI: 10.1016/j.neuron.2014.04.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2014] [Indexed: 12/11/2022]
Abstract
Neural circuitry and brain activity depend critically on proper function of voltage-gated calcium channels (VGCCs), whose activity must be tightly controlled. We show that the main body of the pore-forming α1 subunit of neuronal L-type VGCCs, Cav1.2, is proteolytically cleaved, resulting in Cav1.2 fragment channels that separate but remain on the plasma membrane. This "midchannel" proteolysis is regulated by channel activity, involves the Ca(2+)-dependent protease calpain and the ubiquitin-proteasome system, and causes attenuation and biophysical alterations of VGCC currents. Recombinant Cav1.2 fragment channels mimicking the products of midchannel proteolysis do not form active channels on their own but, when properly paired, produce currents with distinct biophysical properties. Midchannel proteolysis increases dramatically with age and can be attenuated with an L-type VGCC blocker in vivo. Midchannel proteolysis represents a novel form of homeostatic negative-feedback processing of VGCCs that could profoundly affect neuronal excitability, neurotransmission, neuroprotection, and calcium signaling in physiological and disease states.
Collapse
|
3
|
Hofmann F, Flockerzi V, Kahl S, Wegener JW. L-type CaV1.2 calcium channels: from in vitro findings to in vivo function. Physiol Rev 2014; 94:303-26. [PMID: 24382889 DOI: 10.1152/physrev.00016.2013] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The L-type Cav1.2 calcium channel is present throughout the animal kingdom and is essential for some aspects of CNS function, cardiac and smooth muscle contractility, neuroendocrine regulation, and multiple other processes. The L-type CaV1.2 channel is built by up to four subunits; all subunits exist in various splice variants that potentially affect the biophysical and biological functions of the channel. Many of the CaV1.2 channel properties have been analyzed in heterologous expression systems including regulation of the L-type CaV1.2 channel by Ca(2+) itself and protein kinases. However, targeted mutations of the calcium channel genes confirmed only some of these in vitro findings. Substitution of the respective serines by alanine showed that β-adrenergic upregulation of the cardiac CaV1.2 channel did not depend on the phosphorylation of the in vitro specified amino acids. Moreover, well-established in vitro phosphorylation sites of the CaVβ2 subunit of the cardiac L-type CaV1.2 channel were found to be irrelevant for the in vivo regulation of the channel. However, the molecular basis of some kinetic properties, such as Ca(2+)-dependent inactivation and facilitation, has been approved by in vivo mutagenesis of the CaV1.2α1 gene. This article summarizes recent findings on the in vivo relevance of well-established in vitro results.
Collapse
|
4
|
Beta-adrenergic-regulated phosphorylation of the skeletal muscle Ca(V)1.1 channel in the fight-or-flight response. Proc Natl Acad Sci U S A 2010; 107:18712-7. [PMID: 20937870 DOI: 10.1073/pnas.1012384107] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ca(V)1 channels initiate excitation-contraction coupling in skeletal and cardiac muscle. During the fight-or-flight response, epinephrine released by the adrenal medulla and norepinephrine released from sympathetic nerves increase muscle contractility by activation of the β-adrenergic receptor/cAMP-dependent protein kinase pathway and up-regulation of Ca(V)1 channels in skeletal and cardiac muscle. Although the physiological mechanism of this pathway is well defined, the molecular mechanism and the sites of protein phosphorylation required for Ca(V)1 channel regulation are unknown. To identify the regulatory sites of phosphorylation under physiologically relevant conditions, Ca(V)1.1 channels were purified from skeletal muscle and sites of phosphorylation on the α1 subunit were identified by mass spectrometry. Two phosphorylation sites were identified in the proximal C-terminal domain, serine 1575 (S1575) and threonine 1579 (T1579), which are conserved in cardiac Ca(V)1.2 channels (S1700 and T1704, respectively). In vitro phosphorylation revealed that Ca(V)1.1-S1575 is a substrate for both cAMP-dependent protein kinase and calcium/calmodulin-dependent protein kinase II, whereas Ca(V)1.1-T1579 is a substrate for casein kinase 2. Treatment of rabbits with isoproterenol to activate β-adrenergic receptors increased phosphorylation of S1575 in skeletal muscle Ca(V)1.1 channels in vivo, and treatment with propranolol to inhibit β-adrenergic receptors reduced phosphorylation. As S1575 and T1579 in Ca(V)1.1 channels and their homologs in Ca(V)1.2 channels are located at a key regulatory interface between the distal and proximal C-terminal domains, it is likely that phosphorylation of these sites in skeletal and cardiac muscle is directly involved in calcium channel regulation in response to the sympathetic nervous system in the fight-or-flight response.
Collapse
|
5
|
Buvinic S, Almarza G, Bustamante M, Casas M, López J, Riquelme M, Sáez JC, Huidobro-Toro JP, Jaimovich E. ATP released by electrical stimuli elicits calcium transients and gene expression in skeletal muscle. J Biol Chem 2009; 284:34490-505. [PMID: 19822518 DOI: 10.1074/jbc.m109.057315] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
ATP released from cells is known to activate plasma membrane P2X (ionotropic) or P2Y (metabotropic) receptors. In skeletal muscle cells, depolarizing stimuli induce both a fast calcium signal associated with contraction and a slow signal that regulates gene expression. Here we show that nucleotides released to the extracellular medium by electrical stimulation are partly involved in the fast component and are largely responsible for the slow signals. In rat skeletal myotubes, a tetanic stimulus (45 Hz, 400 1-ms pulses) rapidly increased extracellular levels of ATP, ADP, and AMP after 15 s to 3 min. Exogenous ATP induced an increase in intracellular free Ca(2+) concentration, with an EC(50) value of 7.8 +/- 3.1 microm. Exogenous ADP, UTP, and UDP also promoted calcium transients. Both fast and slow calcium signals evoked by tetanic stimulation were inhibited by either 100 mum suramin or 2 units/ml apyrase. Apyrase also reduced fast and slow calcium signals evoked by tetanus (45 Hz, 400 0.3-ms pulses) in isolated mouse adult skeletal fibers. A likely candidate for the ATP release pathway is the pannexin-1 hemichannel; its blockers inhibited both calcium transients and ATP release. The dihydropyridine receptor co-precipitated with both the P2Y(2) receptor and pannexin-1. As reported previously for electrical stimulation, 500 mum ATP significantly increased mRNA expression for both c-fos and interleukin 6. Our results suggest that nucleotides released during skeletal muscle activity through pannexin-1 hemichannels act through P2X and P2Y receptors to modulate both Ca(2+) homeostasis and muscle physiology.
Collapse
Affiliation(s)
- Sonja Buvinic
- Centro de Estudios Moleculares de la Célula, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Casilla 70005, Santiago 7, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Jeftinija DM, Wang QB, Hebert SL, Norris CM, Yan Z, Rich MM, Kraner SD. The Ca(V) 1.2 Ca(2+) channel is expressed in sarcolemma of type I and IIa myofibers of adult skeletal muscle. Muscle Nerve 2007; 36:482-90. [PMID: 17636479 PMCID: PMC2756106 DOI: 10.1002/mus.20842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Although Ca(2+)-dependent signaling pathways are important for skeletal muscle plasticity, the sources of Ca(2+) that activate these signaling pathways are not completely understood. Influx of Ca(2+) through surface membrane Ca(2+) channels may activate these pathways. We examined expression of two L-type Ca(2+) channels in adult skeletal muscle, the Ca(V) 1.1 and Ca(V) 1.2, with isoform-specific antibodies in Western blots and immunocytochemistry assays. Consistent with a large body of work, expression of the Ca(V) 1.1 was restricted to skeletal muscle where it was expressed in T-tubules. Ca(V) 1.2 was also expressed in skeletal muscle, in the sarcolemma of type I and IIa myofibers. Exercise-induced alterations in muscle fiber types cause a concomitant increase in the number of both Ca(V) 1.2 and type IIa-positive fibers. Taken together, these data suggest that the Ca(V) 1.2 Ca(2+) channel is expressed in adult skeletal muscle in a fiber type-specific manner, which may help to maintain oxidative muscle phenotype.
Collapse
Affiliation(s)
- Dusan M Jeftinija
- Department of Molecular and Biomedical Pharmacology, University of Kentucky Medical Center, Lexington, KY 40536, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Yang SN, Berggren PO. The role of voltage-gated calcium channels in pancreatic beta-cell physiology and pathophysiology. Endocr Rev 2006; 27:621-76. [PMID: 16868246 DOI: 10.1210/er.2005-0888] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Voltage-gated calcium (CaV) channels are ubiquitously expressed in various cell types throughout the body. In principle, the molecular identity, biophysical profile, and pharmacological property of CaV channels are independent of the cell type where they reside, whereas these channels execute unique functions in different cell types, such as muscle contraction, neurotransmitter release, and hormone secretion. At least six CaValpha1 subunits, including CaV1.2, CaV1.3, CaV2.1, CaV2.2, CaV2.3, and CaV3.1, have been identified in pancreatic beta-cells. These pore-forming subunits complex with certain auxiliary subunits to conduct L-, P/Q-, N-, R-, and T-type CaV currents, respectively. beta-Cell CaV channels take center stage in insulin secretion and play an important role in beta-cell physiology and pathophysiology. CaV3 channels become expressed in diabetes-prone mouse beta-cells. Point mutation in the human CaV1.2 gene results in excessive insulin secretion. Trinucleotide expansion in the human CaV1.3 and CaV2.1 gene is revealed in a subgroup of patients with type 2 diabetes. beta-Cell CaV channels are regulated by a wide range of mechanisms, either shared by other cell types or specific to beta-cells, to always guarantee a satisfactory concentration of Ca2+. Inappropriate regulation of beta-cell CaV channels causes beta-cell dysfunction and even death manifested in both type 1 and type 2 diabetes. This review summarizes current knowledge of CaV channels in beta-cell physiology and pathophysiology.
Collapse
Affiliation(s)
- Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology L1:03, Karolinska University Hospital Solna, SE-171 76 Stockholm, Sweden.
| | | |
Collapse
|
8
|
Hulme JT, Yarov-Yarovoy V, Lin TWC, Scheuer T, Catterall WA. Autoinhibitory control of the CaV1.2 channel by its proteolytically processed distal C-terminal domain. J Physiol 2006; 576:87-102. [PMID: 16809371 PMCID: PMC1995633 DOI: 10.1113/jphysiol.2006.111799] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Voltage-gated Ca(2+) channels of the Ca(V)1 family initiate excitation-contraction coupling in cardiac, smooth, and skeletal muscle and are primary targets for regulation by the sympathetic nervous system in the 'fight-or-flight' response. In the heart, activation of beta-adrenergic receptors greatly increases the L-type Ca(2+) current through Ca(V)1.2 channels, which requires phosphorylation by cyclic AMP-dependent protein kinase (PKA) anchored via an A-kinase anchoring protein (AKAP15). Surprisingly, the site of interaction of PKA and AKAP15 lies in the distal C-terminus, which is cleaved from the remainder of the channel by in vivo proteolytic processing. Here we report that the proteolytically cleaved distal C-terminal domain forms a specific molecular complex with the truncated alpha(1) subunit and serves as a potent autoinhibitory domain. Formation of the autoinhibitory complex greatly reduces the coupling efficiency of voltage sensing to channel opening and shifts the voltage dependence of activation to more positive membrane potentials. Ab initio structural modelling and site-directed mutagenesis revealed a binding interaction between a pair of arginine residues in a predicted alpha-helix in the proximal C-terminal domain and a set of three negatively charged amino acid residues in a predicted helix-loop-helix bundle in the distal C-terminal domain. Disruption of this interaction by mutation abolished the inhibitory effects of the distal C-terminus on Ca(V)1.2 channel function. These results provide the first functional characterization of this autoinhibitory complex, which may be a major form of the Ca(V)1 family Ca(2+) channels in cardiac and skeletal muscle cells, and reveal a unique ion channel regulatory mechanism in which proteolytic processing produces a more effective autoinhibitor of Ca(V)1.2 channel function.
Collapse
Affiliation(s)
- Joanne T Hulme
- Department of Pharmacology, Mailstop 357280, University of Washington, Seattle, 98195-7280, USA
| | | | | | | | | |
Collapse
|
9
|
Hulme JT, Konoki K, Lin TWC, Gritsenko MA, Camp DG, Bigelow DJ, Catterall WA. Sites of proteolytic processing and noncovalent association of the distal C-terminal domain of CaV1.1 channels in skeletal muscle. Proc Natl Acad Sci U S A 2005; 102:5274-9. [PMID: 15793008 PMCID: PMC555994 DOI: 10.1073/pnas.0409885102] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In skeletal muscle cells, voltage-dependent potentiation of Ca2+ channel activity requires phosphorylation by cAMP-dependent protein kinase (PKA) anchored via an A-kinase anchoring protein (AKAP15), and the most rapid sites of phosphorylation are located in the C-terminal domain. Surprisingly, the site of interaction of the complex of PKA and AKAP15 with the alpha1-subunit of Ca(V)1.1 channels lies in the distal C terminus, which is cleaved from the remainder of the channel by in vivo proteolytic processing. Here we report that the distal C terminus is noncovalently associated with the remainder of the channel via an interaction with a site in the proximal C-terminal domain when expressed as a separate protein in mammalian nonmuscle cells. Deletion mapping of the C terminus of the alpha1-subunit using the yeast two-hybrid assay revealed that a distal C-terminal peptide containing amino acids 1802-1841 specifically interacts with a region in the proximal C terminus containing amino acid residues 1556-1612. Analysis of the purified alpha1-subunit of Ca(V)1.1 channels from skeletal muscle by saturation sequencing of the intracellular peptides by tandem mass spectrometry identified the site of proteolytic processing as alanine 1664. Our results support the conclusion that a noncovalently associated complex of the alpha1-subunit truncated at A1664 with the proteolytically cleaved distal C-terminal domain, AKAP15, and PKA is the primary physiological form of Ca(V)1.1 channels in skeletal muscle cells.
Collapse
Affiliation(s)
- Joanne T Hulme
- Department of Pharmacology, University of Washington, Mailstop 357280, Seattle, WA 98195-7280, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Hulme JT, Ahn M, Hauschka SD, Scheuer T, Catterall WA. A novel leucine zipper targets AKAP15 and cyclic AMP-dependent protein kinase to the C terminus of the skeletal muscle Ca2+ channel and modulates its function. J Biol Chem 2002; 277:4079-87. [PMID: 11733497 DOI: 10.1074/jbc.m109814200] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In skeletal muscle, voltage-dependent potentiation of L-type Ca(2+) channel (Ca(V)1.1) activity requires phosphorylation by cyclic AMP-dependent protein kinase (PKA) anchored via an A kinase-anchoring protein (AKAP15). However, the mechanism by which AKAP15 targets PKA to L-type Ca(2+) channels has not been elucidated. Here we report that AKAP15 directly interacts with the C-terminal domain of the alpha(1) subunit of Ca(V)1.1 via a leucine zipper (LZ) motif. Disruption of the LZ interaction effectively inhibits voltage-dependent potentiation of L-type Ca(2+) channels in skeletal muscle cells. Our results reveal a novel mechanism whereby anchoring of PKA to Ca(2+) channels via LZ interactions ensures rapid and efficient phosphorylation of Ca(2+) channels in response to local signals such as cAMP and depolarization.
Collapse
Affiliation(s)
- Joanne T Hulme
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280, USA
| | | | | | | | | |
Collapse
|
11
|
Keef KD, Hume JR, Zhong J. Regulation of cardiac and smooth muscle Ca(2+) channels (Ca(V)1.2a,b) by protein kinases. Am J Physiol Cell Physiol 2001; 281:C1743-56. [PMID: 11698232 DOI: 10.1152/ajpcell.2001.281.6.c1743] [Citation(s) in RCA: 178] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High voltage-activated Ca(2+) channels of the Ca(V)1.2 class (L-type) are crucial for excitation-contraction coupling in both cardiac and smooth muscle. These channels are regulated by a variety of second messenger pathways that ultimately serve to modulate the level of contractile force in the tissue. The specific focus of this review is on the most recent advances in our understanding of how cardiac Ca(V)1.2a and smooth muscle Ca(V)1.2b channels are regulated by different kinases, including cGMP-dependent protein kinase, cAMP-dependent protein kinase, and protein kinase C. This review also discusses recent evidence regarding the regulation of these channels by protein tyrosine kinase, calmodulin-dependent kinase, purified G protein subunits, and identification of possible amino acid residues of the channel responsible for kinase regulation.
Collapse
Affiliation(s)
- K D Keef
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA.
| | | | | |
Collapse
|
12
|
Abstract
Voltage-gated Ca(2+) channels mediate Ca(2+) entry into cells in response to membrane depolarization. Electrophysiological studies reveal different Ca(2+) currents designated L-, N-, P-, Q-, R-, and T-type. The high-voltage-activated Ca(2+) channels that have been characterized biochemically are complexes of a pore-forming alpha1 subunit of approximately 190-250 kDa; a transmembrane, disulfide-linked complex of alpha2 and delta subunits; an intracellular beta subunit; and in some cases a transmembrane gamma subunit. Ten alpha1 subunits, four alpha2delta complexes, four beta subunits, and two gamma subunits are known. The Cav1 family of alpha1 subunits conduct L-type Ca(2+) currents, which initiate muscle contraction, endocrine secretion, and gene transcription, and are regulated primarily by second messenger-activated protein phosphorylation pathways. The Cav2 family of alpha1 subunits conduct N-type, P/Q-type, and R-type Ca(2+) currents, which initiate rapid synaptic transmission and are regulated primarily by direct interaction with G proteins and SNARE proteins and secondarily by protein phosphorylation. The Cav3 family of alpha1 subunits conduct T-type Ca(2+) currents, which are activated and inactivated more rapidly and at more negative membrane potentials than other Ca(2+) current types. The distinct structures and patterns of regulation of these three families of Ca(2+) channels provide a flexible array of Ca(2+) entry pathways in response to changes in membrane potential and a range of possibilities for regulation of Ca(2+) entry by second messenger pathways and interacting proteins.
Collapse
Affiliation(s)
- W A Catterall
- Department of Pharmacology, Box 357280, University of Washington, Seattle, Washington 98195-7280, USA.
| |
Collapse
|
13
|
Flucher BE, Kasielke N, Gerster U, Neuhuber B, Grabner M. Insertion of the full-length calcium channel alpha(1S) subunit into triads of skeletal muscle in vitro. FEBS Lett 2000; 474:93-8. [PMID: 10828458 DOI: 10.1016/s0014-5793(00)01583-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
A full-length and a C-terminally truncated form of the calcium channel alpha(1S) subunit can be isolated from skeletal muscle. Here we studied whether full-length alpha(1S) is functionally incorporated into the skeletal muscle excitation-contraction coupling apparatus. A fusion protein of alpha(1S) with the green fluorescent protein attached to its C-terminus (alpha(1S)-GFP) or alpha(1S) and GFP separately (alpha(1S)+GFP) were expressed in dysgenic myotubes, which lack endogenous alpha(1S). Full-length alpha(1S)-GFP was targeted into triad junctions and restored calcium currents and excitation-contraction coupling. GFP remained colocalized with alpha(1S), indicating that intact alpha(1S)-GFP was inserted into triads and that the C-terminus remained associated with the excitation-contraction coupling apparatus.
Collapse
Affiliation(s)
- B E Flucher
- Department of Physiology and Department of Biochemical Pharmacology, University of Innsbruck, Fritz-Pregl-Str. 3, A-6020, Innsbruck, Austria.
| | | | | | | | | |
Collapse
|
14
|
Zhang MI, O'Neil RG. The diversity of calcium channels and their regulation in epithelial cells. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1999; 46:43-83. [PMID: 10332501 DOI: 10.1016/s1054-3589(08)60469-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- M I Zhang
- Department of Integrative Biology, Pharmacology, and Physiology, University of Texas-Houston Health Science Center 77030, USA
| | | |
Collapse
|
15
|
Rossie S. Regulation of voltage-sensitive sodium and calcium channels by phosphorylation. ADVANCES IN SECOND MESSENGER AND PHOSPHOPROTEIN RESEARCH 1999; 33:23-48. [PMID: 10218113 DOI: 10.1016/s1040-7952(99)80004-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Affiliation(s)
- S Rossie
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, USA
| |
Collapse
|
16
|
Bae I, Yoon S, Yoon Y, Kim MK, Kim H. Identification of three types of voltage dependent Ca2+‐channels in mouse follicular oocytes. ACTA ACUST UNITED AC 1999. [DOI: 10.1080/12265071.1999.9647465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
17
|
Zhou J, Cribbs L, Yi J, Shirokov R, Perez-Reyes E, Ríos E. Molecular cloning and functional expression of a skeletal muscle dihydropyridine receptor from Rana catesbeiana. J Biol Chem 1998; 273:25503-9. [PMID: 9738021 DOI: 10.1074/jbc.273.39.25503] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In skeletal muscle the dihydropyridine receptor is the voltage sensor for excitation-contraction coupling and an L-type Ca2+ channel. We cloned a dihydropyridine receptor (named Fgalpha1S) from frog skeletal muscle, where excitation-contraction coupling has been studied most extensively. Fgalpha1S contains 5600 base pairs coding for 1688 amino acids. It is highly homologous with, and of the same length as, the C-truncated form predominant in rabbit muscle. The primary sequence has every feature needed to be an L-type Ca2+ channel and a skeletal-type voltage sensor. Currents expressed in tsA201 cells had rapid activation (5-10 ms half-time) and Ca2+-dependent inactivation. Although functional expression of the full Fgalpha1S was difficult, the chimera consisting of Fgalpha1S domain I in the rabbit cardiac Ca channel had high expression and a rapidly activating current. The slow native activation is therefore not determined solely by the alpha1 subunit sequence. Its Ca2+-dependent inactivation strengthens the notion that in rabbit skeletal muscle this capability is inhibited by a C-terminal stretch (Adams, B., and Tanabe, T. (1997) J. Gen. Physiol. 110, 379-389). This molecule constitutes a new tool for studies of excitation-contraction coupling, gating, modulation, and gene expression.
Collapse
Affiliation(s)
- J Zhou
- Department of Molecular Biophysics and Physiology, Rush University, Chicago, Illinois 60612, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Kuriyama H, Kitamura K, Itoh T, Inoue R. Physiological features of visceral smooth muscle cells, with special reference to receptors and ion channels. Physiol Rev 1998; 78:811-920. [PMID: 9674696 DOI: 10.1152/physrev.1998.78.3.811] [Citation(s) in RCA: 176] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Visceral smooth muscle cells (VSMC) play an essential role, through changes in their contraction-relaxation cycle, in the maintenance of homeostasis in biological systems. The features of these cells differ markedly by tissue and by species; moreover, there are often regional differences within a given tissue. The biophysical features used to investigate ion channels in VSMC have progressed from the original extracellular recording methods (large electrode, single or double sucrose gap methods), to the intracellular (microelectrode) recording method, and then to methods for recording from membrane fractions (patch-clamp, including cell-attached patch-clamp, methods). Remarkable advances are now being made thanks to the application of these more modern biophysical procedures and to the development of techniques in molecular biology. Even so, we still have much to learn about the physiological features of these channels and about their contribution to the activity of both cell and tissue. In this review, we take a detailed look at ion channels in VSMC and at receptor-operated ion channels in particular; we look at their interaction with the contraction-relaxation cycle in individual VSMC and especially at the way in which their activity is related to Ca2+ movements and Ca2+ homeostasis in the cell. In sections II and III, we discuss research findings mainly derived from the use of the microelectrode, although we also introduce work done using the patch-clamp procedure. These sections cover work on the electrical activity of VSMC membranes (sect. II) and on neuromuscular transmission (sect. III). In sections IV and V, we discuss work done, using the patch-clamp procedure, on individual ion channels (Na+, Ca2+, K+, and Cl-; sect. IV) and on various types of receptor-operated ion channels (with or without coupled GTP-binding proteins and voltage dependent and independent; sect. V). In sect. VI, we look at work done on the role of Ca2+ in VSMC using the patch-clamp procedure, biochemical procedures, measurements of Ca2+ transients, and Ca2+ sensitivity of contractile proteins of VSMC. We discuss the way in which Ca2+ mobilization occurs after membrane activation (Ca2+ influx and efflux through the surface membrane, Ca2+ release from and uptake into the sarcoplasmic reticulum, and dynamic changes in Ca2+ within the cytosol). In this article, we make only limited reference to vascular smooth muscle research, since we reviewed the features of ion channels in vascular tissues only recently.
Collapse
Affiliation(s)
- H Kuriyama
- Seinan Jogakuin University, Kokura-Kita, Fukuoka, Japan
| | | | | | | |
Collapse
|
19
|
Grabner M, Dirksen RT, Beam KG. Tagging with green fluorescent protein reveals a distinct subcellular distribution of L-type and non-L-type Ca2+ channels expressed in dysgenic myotubes. Proc Natl Acad Sci U S A 1998; 95:1903-8. [PMID: 9465115 PMCID: PMC19211 DOI: 10.1073/pnas.95.4.1903] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Expression of cardiac L-type Ca2+ channels in dysgenic myotubes results in large Ca2+ currents and electrically evoked contractions resulting from Ca2+-entry dependent release of Ca2+ from the sarcoplasmic reticulum. By contrast, expression of either P/Q-type or N-type Ca2+ channels in dysgenic myotubes does not result in electrically evoked contractions despite producing comparably large Ca2+ currents. In this work we examined the possibility that this discrepancy is caused by the preferential distribution of expressed L-type Ca2+ channels in close apposition to sarcoplasmic reticulum Ca2+ release channels. We tagged the N termini of different alpha1 subunits (classes A, B, C, and S) with a modified green fluorescent protein (GFP) and expressed each of the fusion channels in dysgenic myotubes. Each GFP-tagged alpha1 subunit exhibited Ca2+ channel activity that was indistinguishable from its wild-type counterpart. In addition, expression of GFP-alpha1S and GFP-alpha1C in dysgenic myotubes restored skeletal- and cardiac-type excitation-contraction (EC) coupling, respectively, whereas expression of GFP-alpha1A and GFP-alpha1B failed to restore EC coupling of any type. Laser-scanning confocal microscopy revealed a distinct expression pattern for L-type compared with non-L-type channels. After injection of cDNA into a single nucleus, GFP-alpha1S and GFP-alpha1C were present in the plasmalemma as small punctate foci along much of the longitudinal extent of the myotube. In contrast, GFP-alpha1A and GFP-alpha1B were not concentrated into punctate foci and primarily were found adjacent to the injected nucleus. Thus, L-type channels possess a targeting signal that directs their longitudinal transport and insertion into punctate regions of myotubes that presumably represent functional sites of EC coupling.
Collapse
Affiliation(s)
- M Grabner
- Department of Anatomy and Neurobiology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | | |
Collapse
|
20
|
Catterall WA. Modulation of sodium and calcium channels by protein phosphorylation and G proteins. ADVANCES IN SECOND MESSENGER AND PHOSPHOPROTEIN RESEARCH 1997; 31:159-81. [PMID: 9344250 DOI: 10.1016/s1040-7952(97)80017-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- W A Catterall
- Department of Pharmacology, University of Washington School of Medicine, Seattle 98195, USA
| |
Collapse
|
21
|
Gray PC, Tibbs VC, Catterall WA, Murphy BJ. Identification of a 15-kDa cAMP-dependent protein kinase-anchoring protein associated with skeletal muscle L-type calcium channels. J Biol Chem 1997; 272:6297-302. [PMID: 9045648 DOI: 10.1074/jbc.272.10.6297] [Citation(s) in RCA: 106] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Voltage-dependent potentiation of skeletal muscle L-type calcium channels requires phosphorylation by cAMP-dependent protein kinase (PKA) that is localized by binding to a cAMP-dependent protein kinase-anchoring protein (AKAP). L-type calcium channels purified from rabbit skeletal muscle contain an endogenous co-purifying protein kinase activity that phosphorylates the alpha1 and beta subunits of the channel. The co-purifying kinase also phosphorylates a known PKA peptide substrate, is stimulated by cAMP, and is inhibited by PKA inhibitor peptide-(5-24), indicating that it is PKA. PKA activity co-immunoprecipitates with the calcium channel, suggesting that the channel and the kinase are physically associated. Using biotinylated type II regulatory subunit of PKA (RII) as a probe, we have identified a 15-kDa RII-binding protein in purified calcium channel preparations, which we have designated AKAP-15. Anti-peptide antibodies directed against the alpha1 subunit of the calcium channel co-immunoprecipitate AKAP-15. Together, these findings demonstrate a physical link between PKA and the calcium channel and suggest that AKAP-15 may mediate their interaction.
Collapse
Affiliation(s)
- P C Gray
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280, USA
| | | | | | | |
Collapse
|
22
|
|
23
|
Zhang MI, O'Neil RG. Regulated calcium channel in apical membranes renal proximal tubule cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1996; 271:C1757-64. [PMID: 8944661 DOI: 10.1152/ajpcell.1996.271.5.c1757] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Using the single-channel patch-clamp technique, we identified a Ca2+ channel in the apical membranes rabbit cultured proximal tubule cells. The channel is permeable to both Ca2+ and Ba2+ but not to monovalent cations. In on-cell patches, the channel opened infrequently and had a conductance of 4.6 +/- 0.9 pS (n = 5) with 105 mM CaCl2 in the pipette. Although addition of forskolin (12.5 microM) Cl2 is without effect, addition of phorbol 12-myristate 13-acetate (PMA, 1 microM) activated the channel. At 0 mV pipette voltage (resting state) in the on-cell patches, PMA increased open probability (P0) from 0 to 6.9 +/- 2.3% (n = 5) within 1-3 min of PMA application. Likewise, stretching the membrane patch (-10 to -30 mmHg) activated this channel (P0 increased to 5.3 +/- 2.1%, n = 3, at 0 mV applied pipette potential), with results consistent with a mechanosensitive channel. The channel displayed only modest voltage sensitivity, with mild activation on membrane hyperpolarization but with inactivation on strong depolarization. The addition of the L-type Ca2+ channel blocker (antagonist), nifedipine (10 microM), completely blocked this channel in both on-cell and inside-out patches, whereas the agonist, BAY K 8644 (5 microM) was without effect. It is concluded that this channel is a nifedipine-sensitive, protein kinase C-regulated Ca2+ channel and that it may play a role in Ca2+ signaling in the proximal tubule cells, particularly during periods of mechanical stress.
Collapse
Affiliation(s)
- M I Zhang
- Department of Integrative Biology, University of Texas-Houston Health Science Center 77030, USA
| | | |
Collapse
|
24
|
Komalavilas P, Lincoln TM. Phosphorylation of the inositol 1,4,5-trisphosphate receptor. Cyclic GMP-dependent protein kinase mediates cAMP and cGMP dependent phosphorylation in the intact rat aorta. J Biol Chem 1996; 271:21933-8. [PMID: 8702997 DOI: 10.1074/jbc.271.36.21933] [Citation(s) in RCA: 169] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The effects of cyclic GMP (cGMP) and activation of cGMP-dependent protein kinase (PKG) on the phosphorylation of the inositol 1,4, 5-trisphosphate (IP3) receptor were examined in intact rat aorta using the technique of back phosphorylation. Aorta treated with the nitric oxide donors, S-nitroso-N-acetylpenicillamine and sodium nitroprusside, or the selective PKG activator, 8-(4-para-chlorophenylthio)-cGMP (8-CPT-cGMP), demonstrated increased IP3 receptor phosphorylation in situ, which was both time- and concentration-dependent with a stoichiometry of 0.5 mol of phosphate/mol of receptor above control. Treatment of aorta with the adenyl cyclase activator, forskolin, also demonstrated increased phosphorylation of the IP3 receptor on the PKG site, although the selective cAMP-dependent protein kinase activator, 8-(4-para-chlorophenylthio)-cAMP (8-CPT-cAMP), did not increase the phosphorylation of the IP3 receptor. Moreover, the PKG selective inhibitor, KT 5823, inhibited both sodium nitroprusside and forskolin-induced IP3 receptor phosphorylation more potently than the selective cAMP-dependent protein kinase inhibitor, KT 5720, suggesting that PKG mediates the increase in IP3 receptor phosphorylation by both cyclic nucleotides in intact aorta. These results provide further support for the notion that PKG is activated by both cAMP and cGMP in intact vascular smooth muscle and that PKG performs a critical role in cyclic nucleotide-dependent relaxation of blood vessels.
Collapse
Affiliation(s)
- P Komalavilas
- Department of Pathology, University of Alabama at Birmingham, 35294-0019, USA
| | | |
Collapse
|
25
|
Abstract
Voltage-gated sodium and calcium channels are responsible for inward movement of sodium and calcium during electrical signals in cell membranes. Their principal subunits are members of a gene family and can function as voltage-gated ion channels by themselves. They are expressed in association with one or more auxiliary subunits which increase functional expression and modify the functional properties of the principal subunits. Structural elements which are required for voltage-dependent activation, selective ion conductance, and inactivation have been identified, and their mechanisms of action are being explored through mutagenesis, expression in heterologous cells, and functional analysis. These experiments reveal that these two channels are built on a common structural theme with variations appropriate for functional specialization of each channel type.
Collapse
Affiliation(s)
- W A Catterall
- Department of Pharmacology, University of Washington, Seattle 98195-7280, USA
| |
Collapse
|
26
|
Fratacci MD, Shimahara T, Bournaud R, Atlan G. cAMP-dependent modulation of L-type calcium currents in mouse diaphragmatic cells. RESPIRATION PHYSIOLOGY 1996; 104:1-9. [PMID: 8865376 DOI: 10.1016/0034-5687(96)00031-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The regulation of calcium channels by cAMP-dependent phosphorylation was investigated in the diaphragm muscle. Experiments were performed on dissociated costal diaphragmatic cells from 16- to 17-day-old fetal mice. The ionic current through calcium channels was measured using the whole cell clamp technique with barium as the charge carrier. A depolarizing pulse delivered from a holding potential of -80 mV elicited a low-threshold dihydropyridine (DHP)-insensitive T-type current and a high-threshold DHP-sensitive L-type current. Agents that either increase intracellular cAMP levels (forskolin, 10(-4) M, and dibutyryladenosine 3'-5' cyclic monophosphate, 10(-4) M) or inhibit cAMP degradation (theophylline, 10(-4) M) produced relative increases in L-type current amplitude of 24.4 +/- 13.8%, 13.4 +/- 4.6%, and 15.9 +/- 2.8% (p < 0.05), respectively. Current intensity increased after application of the beta-adrenergic agonist isoproterenol (10(-5) M, 16.5 +/- 3.6%, P < 0.005). None of these agents affected the T-type current. These results suggest that L-type calcium channel activities of the diaphragm muscle are regulated by cAMP-dependent phosphorylation.
Collapse
Affiliation(s)
- M D Fratacci
- INSERM, U296, Faculté de Médecine, Créteil, France
| | | | | | | |
Collapse
|
27
|
Meissner G, Lu X. Dihydropyridine receptor-ryanodine receptor interactions in skeletal muscle excitation-contraction coupling. Biosci Rep 1995; 15:399-408. [PMID: 8825041 DOI: 10.1007/bf01788371] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Much recent progress has been made in our understanding of the mechanism of sarcoplasmic reticulum Ca2+ release in skeletal muscle. Vertebrate skeletal muscle excitation-contraction (E-C) coupling is thought to occur by a "mechanical coupling" mechanism involving protein-protein interactions that lead to activation of the sarcoplasmic reticulum (SR) ryanodine receptor (RyR)/Ca2+ release channel by the voltage-sensing transverse (T-) tubule dihydropyridine receptor (DHPR)/Ca2+ channel. In a subsequent step, the released Ca2+ amplify SR Ca2+ release by activating release channels that are not linked to the DHPR. Experiments with mutant muscle cells have indicated that skeletal muscle specific DHPR and RyR isoforms are required for skeletal muscle E-C coupling. A direct functional and structural interaction between a DHPR-derived peptide and the RyR has been described. The interaction between the DHPR and RyR may be stabilized by other proteins such as triadin (a SR junctional protein) and modulated by phosphorylation of the DHPR.
Collapse
Affiliation(s)
- G Meissner
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill 27599-7260, USA
| | | |
Collapse
|
28
|
Lu X, Xu L, Meissner G. Phosphorylation of dihydropyridine receptor II-III loop peptide regulates skeletal muscle calcium release channel function. Evidence for an essential role of the beta-OH group of Ser687. J Biol Chem 1995; 270:18459-64. [PMID: 7629172 DOI: 10.1074/jbc.270.31.18459] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In vertebrate skeletal muscle, excitation-contraction coupling may occur by a mechanical coupling mechanism involving protein-protein interactions between the dihydropyridine receptor (DHPR) of the transverse tubule membrane and the ryanodine receptor (RYR)/Ca2+ release channel of the sarcoplasmic reticulum membrane. We have previously shown that the cytoplasmic II-III loop peptides of the skeletal and cardiac muscle DHPR alpha 1 subunits (SDCL and CDCL, respectively) activate the skeletal muscle RYR. We now report that cyclic AMP-dependent protein kinase-mediated phosphorylation of Ser687 of SDCL yields a peptide that fails to activate the RYR, as determined in [3H]ryanodine binding and single channel measurements. The phosphorylated SDCL bound to the skeletal muscle but not cardiac muscle RYR, and the binding could be displaced by the unphosphorylated SDCL. A mutant SDCL with a Ser687-->Ala substitution failed to activate the RYR, but was still able to bind. Similarly, a Ser813-->Ala substitution in CDCL yielded a peptide that failed to activate the skeletal RYR. Use of three smaller overlapping peptides within the SDCL region identified an amino acid region from 666 to 726 including Ser687, which bound to and activated the skeletal muscle RYR. These results suggest that cyclic AMP-dependent protein kinase-mediated phosphorylation of the DHPR alpha 1 subunit may play a role in the functional interaction of the DHPR and RYR in skeletal muscle.
Collapse
Affiliation(s)
- X Lu
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill 27599-7260, USA
| | | | | |
Collapse
|
29
|
Rotman EI, Murphy BJ, Catterall WA. Sites of selective cAMP-dependent phosphorylation of the L-type calcium channel alpha 1 subunit from intact rabbit skeletal muscle myotubes. J Biol Chem 1995; 270:16371-7. [PMID: 7608207 DOI: 10.1074/jbc.270.27.16371] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The principal (alpha 1) subunit of purified skeletal muscle dihydropyridine-sensitive (L-type) calcium channels is present in full-length (212 kDa) and COOH-terminal truncated (190 kDa) forms, which are both phosphorylated by cAMP-dependent protein kinase (cA-PK) in vitro. Immunoprecipitation of the calcium channel from rabbit muscle myotubes in primary cell culture followed by phosphorylation with cA-PK, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and two-dimensional phosphopeptide mapping revealed comparable phosphorylation of three COOH-terminal phosphopeptides found in the purified full-length alpha 1 subunit. Stimulation of muscle myotubes with a permeant cAMP analogue, 8-(4-chlorophenylthio) adenosine 3',5'-cyclic monophosphate, prior to immunoprecipitation of alpha 1 results in a 60-80% reduction of cA-PK catalyzed "back" phosphorylation of each of these sites in vitro in calcium channels purified from the cells, indicating that these sites are phosphorylated in vivo in response to increased intracellular cAMP. Serine 687, the most rapidly phosphorylated site in the truncated 190-kDa alpha 1 subunit, was observed as a minor phosphopeptide whose level of phosphorylation was not significantly affected by stimulation of endogenous cA-PK in the myotubes. The COOH-terminal sites, designated tryptic phosphopeptides 4, 5, and 6, were identified as serine 1757 (phosphopeptides 4 and 6) and 1854 (phosphopeptide 5) by a combination of protease cleavage, phosphorylation of synthetic peptides and fusion proteins, specific immunoprecipitation, and phosphopeptide mapping. Phosphorylation of serines 1757 and 1854 in the COOH-terminal region of the 212-kDa alpha 1 subunit in intact skeletal muscle cells may play a pivotal role in the regulation of calcium channel function by cA-PK.
Collapse
Affiliation(s)
- E I Rotman
- Department of Pharmacology, University of Washington, Seattle 98195, USA
| | | | | |
Collapse
|
30
|
Melzer W, Herrmann-Frank A, Lüttgau HC. The role of Ca2+ ions in excitation-contraction coupling of skeletal muscle fibres. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1241:59-116. [PMID: 7742348 DOI: 10.1016/0304-4157(94)00014-5] [Citation(s) in RCA: 406] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- W Melzer
- Department of Cell Physiology, Ruhr-University, Bochum, Germany
| | | | | |
Collapse
|
31
|
Diversé-Pierluissi M, Dunlap K. Interaction of convergent pathways that inhibit N-type calcium currents in sensory neurons. Neuroscience 1995; 65:477-83. [PMID: 7777162 DOI: 10.1016/0306-4522(94)00476-l] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Norepinephrine and GABA inhibit omega-conotoxin GVIA-sensitive (N-type) calcium current in embryonic sensory neurons by separate pathways. We have investigated the mechanisms that limit the modulation of N current by varying the level of activation for a single pathway or simultaneously activating multiple pathways. Calcium currents were measured with tight-seal, whole-cell recording methods. Simultaneous application of the two transmitters at saturating concentrations produced a larger inhibition of the current than either transmitter by itself, but the maximal inhibition was not linearly additive. Maximal, direct activation of GTP-binding proteins by intracellular application of guanosine 5'-(3-O-thio)-triphosphate (GTP gamma S) resulted in a similar limit to the inhibition; furthermore, GTP gamma S did not enhance the maximal inhibition produced by co-application of transmitters. Interventions downstream in the modulatory pathway (e.g. direct activation of protein kinase C or inhibition of protein phosphatases) were also unable to alter the maximal limit for inhibition. These results suggest that transmitter-mediated inhibition is not limited by receptor number, levels of G-protein or protein kinase C activation, or degree of phosphorylation; rather, the extent of inhibition may be limited by the structural properties of the N channels themselves.
Collapse
Affiliation(s)
- M Diversé-Pierluissi
- Department of Physiology and Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | | |
Collapse
|
32
|
Ruiz-Velasco V, Mayer MB, Inscho EW, Hymel LJ. Modulation of dihydropyridine receptors in vascular smooth muscle cells by membrane potential and cell proliferation. Eur J Pharmacol 1994; 268:311-8. [PMID: 7805757 DOI: 10.1016/0922-4106(94)90055-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We have studied binding of isradipine to A7r5 vascular smooth muscle cells as a function of membrane potential and cell proliferation. Consistent with a voltage-modulated receptor model, two classes of binding sites were detected in confluent cultures: high-affinity sites under depolarizing (50 mM K+) conditions (Kd = 45 +/- 3 pM), and lower affinity sites under resting (5 mM K+) conditions (Kd = 181 +/- 20 pM). However, proliferating cells also displayed the high-affinity state at rest (Kd = 29 +/- 9 pM) in addition to a low-affinity site (Kd = 869 +/- 383 pM). Analysis of dissociation rates also revealed two receptor classes during proliferation. Proliferating cells showed a single class of high-affinity sites (Kd = 39 +/- 6 pM) when depolarized, similar to confluent cells. Receptor density in confluent monolayers increased from 15 +/- 3 fmol/10(6) cells at 5 days to 72 +/- 6 fmol/10(6) cells after 10 days. These results suggest (i) that some L-type Ca2+ channels are spontaneously active in proliferating vascular smooth muscle cells, but require depolarization to activate in a confluent monolayer, and (ii) that the density of dihydropyridine receptors increases after a monolayer becomes confluent.
Collapse
Affiliation(s)
- V Ruiz-Velasco
- Tulane University School of Medicine, Department of Physiology, New Orleans, LA 70112-2699
| | | | | | | |
Collapse
|
33
|
Vandaele SF, Rieger F. Co-localization of 1,4-dihydropyridine receptor alpha 2/delta subunit and N-CAM during early myogenesis in vitro. J Cell Sci 1994; 107 ( Pt 5):1217-27. [PMID: 7929631 DOI: 10.1242/jcs.107.5.1217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The surface distribution of the alpha 2/delta subunit of the 1,4-dihydropyridine receptor and its topographical relationship with the neural cell adhesion molecule (N-CAM) were investigated during early myogenesis in vitro, by double immunocytochemical labeling with the monoclonal antibody 3007 and an anti-N-CAM polyclonal antiserum. The monoclonal antibody 3007 has been previously shown to immunoprecipitate dihydropyridine receptor from skeletal muscle T-tubules. In further immunoprecipitation experiments on such preparations and muscle cell cultures, it was demonstrated here that the monoclonal antibody 3007 exclusively recognizes the alpha 2/delta subunit of the 1,4-dihydropyridine receptor. In rabbit muscle cell cultures, the labeling for both alpha 2/delta and N-CAM was first detected on myoblasts, in the form of spots on the membrane and perinuclear patches. Spots of various sizes organized in aggregates were then found on the membrane of myotubes. At fusion (T0), aggregates of N-CAM spots alone were found at the junction between fusing cells. At T6 and later stages, all alpha 2/delta aggregates present on myotubes co-localized with N-CAM, while less than 3% of N-CAM aggregates did not co-localize with alpha 2/delta. A uniform N-CAM staining also made its appearance. At T12, when myotubes showed prominent contractility, alpha 2/delta-N-CAM aggregates diminished in size. Dispersed alpha 2/delta spots of a small regular size spread over the whole surface of the myotubes and alignments of these spots became visible. Corresponding N-CAM spots were now occasionally seen, and uniform N-CAM staining was prominent. These results show that alpha 2/delta and N-CAM are co-localized and that their distributions undergo concomitant changes during early myogenesis until the T-tubule network starts to be organized. This suggest that these two proteins might jointly participate in morphogenetic events preceding the formation of T-tubules.
Collapse
Affiliation(s)
- S F Vandaele
- Département de Pathologie, Université de Montréal, QC, Canada
| | | |
Collapse
|
34
|
Hell J, Appleyard S, Yokoyama C, Warner C, Catterall W. Differential phosphorylation of two size forms of the N-type calcium channel alpha 1 subunit which have different COOH termini. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)37297-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
35
|
Maurer JA, McKay DB. Staurosporine-induced reduction of secretory function in cultured bovine adrenal chromaffin cells. Eur J Pharmacol 1994; 253:115-24. [PMID: 8013537 DOI: 10.1016/0014-2999(94)90765-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Staurosporine, a potent inhibitor of protein kinases, has been used to investigate the involvement of protein kinases in cellular processes such as secretory function and differentiation. We have been examining the effects of staurosporine on secretory function under the same conditions it induces dramatic changes in cell morphology in cultured bovine adrenal chromaffin cells. Our results show that treatment with 100 nM staurosporine reduces catecholamine release stimulated by 56 mM KCl, 10 microM nicotine, and 2 mM BaCl2 in a time-dependent manner (t1/2s, 42, 32, and 31 min, respectively). However, we demonstrate that the time-dependent effects on secretory function are not the direct result of staurosporine-induced changes in cell morphology. The effects of staurosporine on secretion stimulated by KCl, nicotine, and BaCl2 are concentration-dependent (IC50s, 6.3, 29.3, and 34.9 nM, respectively). Staurosporine pretreatment does not inhibit activated 45Ca2+ influx, but does reduce catecholamine release stimulated directly by Ca2+ from permeabilized cells. Furthermore, staurosporine also inhibits basal release with time- and concentration-dependencies (IC50, 9.3 nM and t1/2, 21 min) similar to those found for stimulated release. These results suggest that prolonged staurosporine pretreatment may result in the depletion/alteration of a component essential for the more terminal steps of the secretory process.
Collapse
Affiliation(s)
- J A Maurer
- Division of Pharmacology, Ohio State University, College of Pharmacy, Columbus 43210
| | | |
Collapse
|
36
|
Vaello M, Ruiz-Gómez A, Lerma J, Mayor F. Modulation of inhibitory glycine receptors by phosphorylation by protein kinase C and cAMP-dependent protein kinase. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)42127-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
37
|
Tanabe T, Mikami A, Niidome T, Numa S, Adams BA, Beam KG. Structure and function of voltage-dependent calcium channels from muscle. Ann N Y Acad Sci 1993; 707:81-6. [PMID: 9137544 DOI: 10.1111/j.1749-6632.1993.tb38044.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- T Tanabe
- Howard Hugber Medical Institute, Department of Cellular & Molecular Physiology, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, Connecticut 06536-0812, USA
| | | | | | | | | | | |
Collapse
|
38
|
Haase H, Karczewski P, Beckert R, Krause EG. Phosphorylation of the L-type calcium channel beta subunit is involved in beta-adrenergic signal transduction in canine myocardium. FEBS Lett 1993; 335:217-22. [PMID: 8253200 DOI: 10.1016/0014-5793(93)80733-b] [Citation(s) in RCA: 92] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cyclic AMP-mediated phosphorylation of calcium channel subunits was studied in vitro and in vivo in preparations from dog heart. Calcium channels in native cardiac membranes were phosphorylated by cAMP-dependent protein kinase (PKA) solubilized with digitonin and subsequently immunoprecipitated using a polyclonal antibody generated against the deduced carboxy-terminal sequence of the cardiac beta subunit. A 62 kDa protein was identified as the major PKA-substrate in the immunoprecipitates. In the intact myocardium, this putative beta subunit was found to be phosphorylated in response to cAMP elevating agents. In contrast, no phosphorylation of a protein with an electrophoretic mobility similar to the alpha 1 subunit was detected, although 1,4-dihydropyridine receptor sites were recovered in the immunoprecipitates. Thus, we suggest that PKA-mediated phosphorylation of the beta subunit is the major mechanism for beta-adrenergic regulation of cardiac L-type calcium channel activity.
Collapse
Affiliation(s)
- H Haase
- Max Delbrück Centre for Molecular Medicine, Molecular Cardiology, Berlin-Buch, Germany
| | | | | | | |
Collapse
|
39
|
|
40
|
Wang Z, Estacion M, Mordan LJ. Ca2+ influx via T-type channels modulates PDGF-induced replication of mouse fibroblasts. THE AMERICAN JOURNAL OF PHYSIOLOGY 1993; 265:C1239-46. [PMID: 8238477 DOI: 10.1152/ajpcell.1993.265.5.c1239] [Citation(s) in RCA: 138] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The role of low-threshold voltage-gated calcium channels (VGCC) in modulating extracellular calcium influx and proliferation was investigated in platelet-derived growth factor (PDGF)-stimulated C3H/10T1/2 mouse fibroblasts. Previous studies demonstrated that cell cycle progression after PDGF stimulation was dependent on extracellular calcium influx producing a sustained increase in the intracellular calcium concentration. In this study, PDGF-induced calcium influx, the sustained intracellular calcium increase, and progression to S phase were inhibited by nordihydroguariaretic acid (NDGA), an inhibitor of calcium influx through VGCC. With the use of the whole cell patch-clamp technique to measure calcium currents, NDGA inhibited inward calcium current through low-threshold VGCC, the only VGCC expressed in C3H/10T1/2 fibroblasts. The inhibitory effects of NDGA on calcium influx and cell proliferation each had a mean inhibitory dose of 2-3 microM. Although NDGA also effectively inhibits cyclooxygenase and lipoxygenase, the addition of prostaglandins or leukotrienes could not reverse this inhibition nor could it be replicated by other antioxidants. These data support the hypothesis that low-threshold VGCC can mediate extracellular calcium influx on the stimulation of cell proliferation by PDGF.
Collapse
Affiliation(s)
- Z Wang
- Cancer Research Center of Hawaii, University of Hawaii at Manoa, Honolulu 96813
| | | | | |
Collapse
|
41
|
Lai Y, Peterson BZ, Catterall WA. Selective dephosphorylation of the subunits of skeletal muscle calcium channels by purified phosphoprotein phosphatases. J Neurochem 1993; 61:1333-9. [PMID: 8397293 DOI: 10.1111/j.1471-4159.1993.tb13626.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Multiple sites on the alpha 1 and beta subunits of purified skeletal muscle calcium channels are phosphorylated by cyclic AMP-dependent protein kinase, resulting in three different tryptic phosphopeptides derived from each subunit. Phosphoprotein phosphatases dephosphorylated these sites selectively. Phosphoprotein phosphatase 1 (PP1) and phosphoprotein phosphatase 2A (PP2A) dephosphorylated both alpha 1 and beta subunits at similar rates, whereas calcineurin dephosphorylated beta subunits preferentially. PP1 dephosphorylated phosphopeptides 1 and 2 of the alpha 1 subunit more rapidly than phosphopeptide 3. In contrast, PP2A dephosphorylated phosphopeptide 3 of the alpha 1 subunit preferentially. All three phosphoprotein phosphatases preferentially dephosphorylated phosphopeptide 1 of the beta subunit and dephosphorylated phosphopeptides 2 and 3 more slowly. Mn2+ increased the rate and extent of dephosphorylation of all sites by calcineurin so that > 80% dephosphorylation of both alpha 1 and beta subunits was obtained. The results demonstrate selective dephosphorylation of different phosphorylation sites on the alpha 1 and beta subunits of skeletal muscle calcium channels by the three principal serine/threonine phosphoprotein phosphatases.
Collapse
Affiliation(s)
- Y Lai
- Department of Pharmacology, University of Washington, Seattle 98195
| | | | | |
Collapse
|
42
|
Differential phosphorylation of two size forms of the neuronal class C L-type calcium channel alpha 1 subunit. J Biol Chem 1993. [DOI: 10.1016/s0021-9258(19)36536-6] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
43
|
Catterall WA, de Jongh K, Rotman E, Hell J, Westenbroek R, Dubel SJ, Snutch TP. Molecular properties of calcium channels in skeletal muscle and neurons. Ann N Y Acad Sci 1993; 681:342-55. [PMID: 8395149 DOI: 10.1111/j.1749-6632.1993.tb22913.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- W A Catterall
- Department of Pharmacology, University of Washington, Seattle 98195
| | | | | | | | | | | | | |
Collapse
|
44
|
Lin LF, Kim KT, Westhead EW. Protein phosphorylation at a postreceptor site can block desensitization and induce potentiation of secretion in chromaffin cells. J Neurochem 1993; 60:1491-7. [PMID: 8384251 DOI: 10.1111/j.1471-4159.1993.tb03312.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Desensitization or habituation to repeated or prolonged stimulation is a common property of secretory cells. Phosphorylation of receptors mediates some desensitization processes, but the relationship of phosphorylation to desensitization at postreceptor sites is not well understood. We have tested the effect of protein phosphorylation on desensitization in bovine chromaffin cells. To increase protein phosphorylation, we have used the protein phosphatase inhibitor okadaic acid at 12.5 nM, 100 microM 8-bromo-cyclic AMP to activate protein kinase A, and 10 nM phorbol 12,13-dibutyrate to activate protein kinase C. During repeated 6-s stimulation at 5-min intervals, catecholamine secretion from control cells decreases. Cells exposed to 8-bromo-cyclic AMP or okadaic acid alone show slightly decreased rates of desensitization. In cells pretreated with phorbol 12,13-dibutyrate, desensitization is blocked. Okadaic acid-treated cells stimulated in the presence of 8-bromo-cyclic AMP show potentiation of secretion with repeated stimulation. The protein kinase inhibitor 1-(5-isoquinolinylsulfonyl)-2-methylpiperazine (H7) increases the desensitization rate. Because these phenomena are observed during secretion evoked with elevated K+ as well as by a nicotinic agonist, the effect of phosphorylation is at a postreceptor site. In contrast to desensitization to the repeated stimulations, desensitization to prolonged stimulation with high K+ is not altered by the above protocols in chromaffin cells.
Collapse
Affiliation(s)
- L F Lin
- Program in Molecular and Cellular Biology, University of Massachusetts, Amherst 01003
| | | | | |
Collapse
|
45
|
Westenbroek RE, Hell JW, Warner C, Dubel SJ, Snutch TP, Catterall WA. Biochemical properties and subcellular distribution of an N-type calcium channel alpha 1 subunit. Neuron 1992; 9:1099-115. [PMID: 1334419 DOI: 10.1016/0896-6273(92)90069-p] [Citation(s) in RCA: 462] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A site-directed anti-peptide antibody, CNB-1, that recognizes the alpha 1 subunit of rat brain class B calcium channels (rbB) immunoprecipitated 43% of the N-type calcium channels labeled by [125I]omega-conotoxin. CNB-1 recognized proteins of 240 and 210 kd, suggesting the presence of two size forms of this alpha 1 subunit. Calcium channels recognized by CNB-1 were localized predominantly in dendrites; both dendritic shafts and punctate synaptic structures upon the dendrites were labeled. The large terminals of the mossy fibers of the dentate gyrus granule neurons were heavily labeled, suggesting that the punctate labeling pattern represents calcium channels in nerve terminals. The pattern of immunostaining was cell specific. The cell bodies of some pyramidal cells in layers II, III, and V of the dorsal cortex, Purkinje cells, and scattered cell bodies elsewhere in the brain were also labeled at a low level. The results define complementary distributions of N- and L-type calcium channels in dendrites, nerve terminals, and cell bodies of most central neurons and support distinct functional roles in calcium-dependent electrical activity, intracellular calcium regulation, and neurotransmitter release for these two channel types.
Collapse
Affiliation(s)
- R E Westenbroek
- Department of Pharmacology, University of Washington, Seattle 98195
| | | | | | | | | | | |
Collapse
|
46
|
Beam KG, Adams BA, Niidome T, Numa S, Tanabe T. Function of a truncated dihydropyridine receptor as both voltage sensor and calcium channel. Nature 1992; 360:169-71. [PMID: 1331811 DOI: 10.1038/360169a0] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The skeletal muscle dihydropyridine (DHP) receptor serves dual functions, as a voltage sensor for excitation-contraction coupling and as an L-type calcium channel. Biochemical analysis indicates the presence of two forms of the DHP receptor polypeptide in skeletal muscle, a full-length translation product present as a minor species and a much more abundant form that has a truncated carboxy-terminus. On the basis of these and other observations, it has been proposed that, in skeletal muscle, only the full-length DHP receptor can function as a calcium channel and that the truncated form can only function as a voltage sensor for excitation-contraction coupling. To resolve this issue, we have now constructed a complementary DNA (pC6 delta 1) encoding a protein corresponding to the truncated DHP receptor in skeletal muscle. Expression of pC6 delta 1 in dysgenic myotubes fully restores both excitation-contraction coupling and calcium current, consistent with the idea that a single class of DHP receptors performs both functions.
Collapse
Affiliation(s)
- K G Beam
- Department of Physiology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins 80523
| | | | | | | | | |
Collapse
|
47
|
Yoshida A, Takahashi M, Nishimura S, Takeshima H, Kokubun S. Cyclic AMP-dependent phosphorylation and regulation of the cardiac dihydropyridine-sensitive Ca channel. FEBS Lett 1992; 309:343-9. [PMID: 1325377 DOI: 10.1016/0014-5793(92)80804-p] [Citation(s) in RCA: 94] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A polyclonal antibody, CR2, prepared using the C-terminal peptide of the alpha 1 subunit of the rabbit cardiac DHP-sensitive Ca channel, specifically immunoprecipitated the [3H]PN200-110-labeled Ca channel solubilized from cardiac microsomes. The antibody recognized 250 and 200-kDa cardiac microsomal proteins as determined by immunoblotting, and cAMP-dependent protein kinase phosphorylated the 250-kDa, but not the 200-kDa protein in vitro. CHO cells, transfected with the cardiac alpha 1 subunit cDNA carried by an expression vector, synthesized a 250-kDa protein which was recognized by CR2. Adding db-cAMP or forskolin to the transformed CHO cells induced phosphorylation of the 250-kDa protein and stimulated the DHP-sensitive Ba current under patch-clamp conditions. These results suggested that the cardiac DHP-sensitive Ca channel was regulated by cAMP-dependent phosphorylation of the alpha 1 subunit.
Collapse
Affiliation(s)
- A Yoshida
- Mitsubishi Kasei Institute of Life Sciences, Tokyo, Japan
| | | | | | | | | |
Collapse
|
48
|
Specific phosphorylation of a COOH-terminal site on the full-length form of the alpha 1 subunit of the skeletal muscle calcium channel by cAMP-dependent protein kinase. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(18)41972-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
49
|
|
50
|
Dihydropyridine-sensitive L-type Ca2+ channels in human foreskin fibroblast cells. Characterization of activation with the growth factor Lys-bradykinin. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(19)50048-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|