1
|
Yoshida A, Ito A, Yasui N, Yamashita A. Direct binding of calmodulin to the cytosolic C-terminal regions of sweet/umami taste receptors. J Biochem 2023; 174:451-459. [PMID: 37527916 PMCID: PMC11033526 DOI: 10.1093/jb/mvad060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023] Open
Abstract
Sweet and umami taste receptors recognize chemicals such as sugars and amino acids on their extracellular side and transmit signals into the cytosol of the taste cell. In contrast to ligands that act on the extracellular side of these receptors, little is known regarding the molecules that regulate receptor functions within the cytosol. In this study, we analysed the interaction between sweet and umami taste receptors and calmodulin, a representative Ca2+-dependent cytosolic regulatory protein. High prediction scores for calmodulin binding were observed on the C-terminal cytosolic side of mouse taste receptor type 1 subunit 3 (T1r3), a subunit that is common to both sweet and umami taste receptors. Pull-down assay and surface plasmon resonance analyses showed different affinities of calmodulin to the C-terminal tails of distinct T1r subtypes. Furthermore, we found that T1r3 and T1r2 showed the highest and considerable binding to calmodulin, whereas T1r1 showed weaker binding affinity. Finally, the binding of calmodulin to T1rs was consistently higher in the presence of Ca2+ than in its absence. The results suggested a possibility of the Ca2+-dependent feedback regulation process of sweet and umami taste receptor signaling by calmodulin.
Collapse
Affiliation(s)
- Atsuki Yoshida
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ayumi Ito
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Norihisa Yasui
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Atsuko Yamashita
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
2
|
Mahling R, Hovey L, Isbell HM, Marx DC, Miller MS, Kilpatrick AM, Weaver LD, Yoder JB, Kim EH, Andresen CNJ, Li S, Shea MA. Na V1.2 EFL domain allosterically enhances Ca 2+ binding to sites I and II of WT and pathogenic calmodulin mutants bound to the channel CTD. Structure 2021; 29:1339-1356.e7. [PMID: 33770503 PMCID: PMC8458505 DOI: 10.1016/j.str.2021.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 12/23/2020] [Accepted: 03/03/2021] [Indexed: 11/23/2022]
Abstract
Neuronal voltage-gated sodium channel NaV1.2 C-terminal domain (CTD) binds calmodulin (CaM) constitutively at its IQ motif. A solution structure (6BUT) and other NMR evidence showed that the CaM N domain (CaMN) is structurally independent of the C-domain (CaMC) whether CaM is bound to the NaV1.2IQp (1,901-1,927) or NaV1.2CTD (1,777-1,937) with or without calcium. However, in the CaM + NaV1.2CTD complex, the Ca2+ affinity of CaMN was more favorable than in free CaM, while Ca2+ affinity for CaMC was weaker than in the CaM + NaV1.2IQp complex. The CTD EF-like (EFL) domain allosterically widened the energetic gap between CaM domains. Cardiomyopathy-associated CaM mutants (N53I(N54I), D95V(D96V), A102V(A103V), E104A(E105A), D129G(D130G), and F141L(F142L)) all bound the NaV1.2 IQ motif favorably under resting (apo) conditions and bound calcium normally at CaMN sites. However, only N53I and A102V bound calcium at CaMC sites at [Ca2+] < 100 μM. Thus, they are expected to respond like wild-type CaM to Ca2+ spikes in excitable cells.
Collapse
Affiliation(s)
- Ryan Mahling
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Liam Hovey
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Holly M Isbell
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Dagan C Marx
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Mark S Miller
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Adina M Kilpatrick
- Department of Physics and Astronomy, Drake University, Des Moines, IA 50311-4516, USA
| | - Lisa D Weaver
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Jesse B Yoder
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Elaine H Kim
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Corinne N J Andresen
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Shuxiang Li
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Madeline A Shea
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA.
| |
Collapse
|
3
|
Wu X, Hong L. Calmodulin Interactions with Voltage-Gated Sodium Channels. Int J Mol Sci 2021; 22:ijms22189798. [PMID: 34575961 PMCID: PMC8472079 DOI: 10.3390/ijms22189798] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 02/06/2023] Open
Abstract
Calmodulin (CaM) is a small protein that acts as a ubiquitous signal transducer and regulates neuronal plasticity, muscle contraction, and immune response. It interacts with ion channels and plays regulatory roles in cellular electrophysiology. CaM modulates the voltage-gated sodium channel gating process, alters sodium current density, and regulates sodium channel protein trafficking and expression. Many mutations in the CaM-binding IQ domain give rise to diseases including epilepsy, autism, and arrhythmias by interfering with CaM interaction with the channel. In the present review, we discuss CaM interactions with the voltage-gated sodium channel and modulators involved in CaM regulation, as well as summarize CaM-binding IQ domain mutations associated with human diseases in the voltage-gated sodium channel family.
Collapse
|
4
|
Zybura A, Hudmon A, Cummins TR. Distinctive Properties and Powerful Neuromodulation of Na v1.6 Sodium Channels Regulates Neuronal Excitability. Cells 2021; 10:cells10071595. [PMID: 34202119 PMCID: PMC8307729 DOI: 10.3390/cells10071595] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Navs) are critical determinants of cellular excitability. These ion channels exist as large heteromultimeric structures and their activity is tightly controlled. In neurons, the isoform Nav1.6 is highly enriched at the axon initial segment and nodes, making it critical for the initiation and propagation of neuronal impulses. Changes in Nav1.6 expression and function profoundly impact the input-output properties of neurons in normal and pathological conditions. While mutations in Nav1.6 may cause channel dysfunction, aberrant changes may also be the result of complex modes of regulation, including various protein-protein interactions and post-translational modifications, which can alter membrane excitability and neuronal firing properties. Despite decades of research, the complexities of Nav1.6 modulation in health and disease are still being determined. While some modulatory mechanisms have similar effects on other Nav isoforms, others are isoform-specific. Additionally, considerable progress has been made toward understanding how individual protein interactions and/or modifications affect Nav1.6 function. However, there is still more to be learned about how these different modes of modulation interact. Here, we examine the role of Nav1.6 in neuronal function and provide a thorough review of this channel’s complex regulatory mechanisms and how they may contribute to neuromodulation.
Collapse
Affiliation(s)
- Agnes Zybura
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Biology Department, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Andy Hudmon
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA;
| | - Theodore R. Cummins
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Biology Department, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Correspondence:
| |
Collapse
|
5
|
Nathan S, Gabelli SB, Yoder JB, Srinivasan L, Aldrich RW, Tomaselli GF, Ben-Johny M, Amzel LM. Structural basis of cytoplasmic NaV1.5 and NaV1.4 regulation. J Gen Physiol 2020; 153:211587. [PMID: 33306788 PMCID: PMC7953540 DOI: 10.1085/jgp.202012722] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are membrane proteins responsible for the rapid upstroke of the action potential in excitable cells. There are nine human voltage-sensitive NaV1 isoforms that, in addition to their sequence differences, differ in tissue distribution and specific function. This review focuses on isoforms NaV1.4 and NaV1.5, which are primarily expressed in skeletal and cardiac muscle cells, respectively. The determination of the structures of several eukaryotic NaVs by single-particle cryo-electron microscopy (cryo-EM) has brought new perspective to the study of the channels. Alignment of the cryo-EM structure of the transmembrane channel pore with x-ray crystallographic structures of the cytoplasmic domains illustrates the complementary nature of the techniques and highlights the intricate cellular mechanisms that modulate these channels. Here, we review structural insights into the cytoplasmic C-terminal regulation of NaV1.4 and NaV1.5 with special attention to Ca2+ sensing by calmodulin, implications for disease, and putative channel dimerization.
Collapse
Affiliation(s)
- Sara Nathan
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sandra B Gabelli
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD.,Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jesse B Yoder
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lakshmi Srinivasan
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard W Aldrich
- Department of Neuroscience, University of Texas at Austin, Austin, TX
| | - Gordon F Tomaselli
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY
| | - L Mario Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
6
|
Tsai WC, Guo S, Olaopa MA, Field LJ, Yang J, Shen C, Chang CP, Chen PS, Rubart M. Complex Arrhythmia Syndrome in a Knock-In Mouse Model Carrier of the N98S Calm1 Mutation. Circulation 2020; 142:1937-1955. [PMID: 32929985 PMCID: PMC7867118 DOI: 10.1161/circulationaha.120.046450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/28/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Calmodulin mutations are associated with arrhythmia syndromes in humans. Exome sequencing previously identified a de novo mutation in CALM1 resulting in a p.N98S substitution in a patient with sinus bradycardia and stress-induced bidirectional ventricular ectopy. The objectives of the present study were to determine if mice carrying the N98S mutation knocked into Calm1 replicate the human arrhythmia phenotype and to examine arrhythmia mechanisms. METHODS Mouse lines heterozygous for the Calm1N98S allele (Calm1N98S/+) were generated using CRISPR/Cas9 technology. Adult mutant mice and their wildtype littermates (Calm1+/+) underwent electrocardiographic monitoring. Ventricular de- and repolarization was assessed in isolated hearts using optical voltage mapping. Action potentials and whole-cell currents and [Ca2+]i, as well, were measured in single ventricular myocytes using the patch-clamp technique and fluorescence microscopy, respectively. The microelectrode technique was used for in situ membrane voltage monitoring of ventricular conduction fibers. RESULTS Two biologically independent knock-in mouse lines heterozygous for the Calm1N98S allele were generated. Calm1N98S/+ mice of either sex and line exhibited sinus bradycardia, QTc interval prolongation, and catecholaminergic bidirectional ventricular tachycardia. Male mutant mice also showed QRS widening. Pharmacological blockade and activation of β-adrenergic receptors rescued and exacerbated, respectively, the long-QT phenotype of Calm1N98S/+ mice. Optical and electric assessment of membrane potential in isolated hearts and single left ventricular myocytes, respectively, revealed β-adrenergically induced delay of repolarization. β-Adrenergic stimulation increased peak density, slowed inactivation, and left-shifted the activation curve of ICa.L significantly more in Calm1N98S/+ versus Calm1+/+ ventricular myocytes, increasing late ICa.L in the former. Rapidly paced Calm1N98S/+ ventricular myocytes showed increased propensity to delayed afterdepolarization-induced triggered activity, whereas in situ His-Purkinje fibers exhibited increased susceptibility for pause-dependent early afterdepolarizations. Epicardial mapping of Calm1N98S/+ hearts showed that both reentry and focal mechanisms contribute to arrhythmogenesis. CONCLUSIONS Heterozygosity for the Calm1N98S mutation is causative of an arrhythmia syndrome characterized by sinus bradycardia, QRS widening, adrenergically mediated QTc interval prolongation, and bidirectional ventricular tachycardia. β-Adrenergically induced ICa.L dysregulation contributes to the long-QT phenotype. Pause-dependent early afterdepolarizations and tachycardia-induced delayed afterdepolarizations originating in the His-Purkinje network and ventricular myocytes, respectively, constitute potential sources of arrhythmia in Calm1N98S/+ hearts.
Collapse
Affiliation(s)
- Wen-Chin Tsai
- The Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cardiology, Cardiovascular Research Center, Buddhist Tzu Chi General Hospital and Tzu Chi University, Hualien, Taiwan
| | - Shuai Guo
- The Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Michael A. Olaopa
- The Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Loren J. Field
- The Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jin Yang
- The Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Changyu Shen
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ching-Pin Chang
- The Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peng-Sheng Chen
- The Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael Rubart
- The Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
7
|
Hanemaaijer NA, Popovic MA, Wilders X, Grasman S, Pavón Arocas O, Kole MH. Ca 2+ entry through Na V channels generates submillisecond axonal Ca 2+ signaling. eLife 2020; 9:54566. [PMID: 32553116 PMCID: PMC7380941 DOI: 10.7554/elife.54566] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/12/2020] [Indexed: 12/15/2022] Open
Abstract
Calcium ions (Ca2+) are essential for many cellular signaling mechanisms and enter the cytosol mostly through voltage-gated calcium channels. Here, using high-speed Ca2+ imaging up to 20 kHz in the rat layer five pyramidal neuron axon we found that activity-dependent intracellular calcium concentration ([Ca2+]i) in the axonal initial segment was only partially dependent on voltage-gated calcium channels. Instead, [Ca2+]i changes were sensitive to the specific voltage-gated sodium (NaV) channel blocker tetrodotoxin. Consistent with the conjecture that Ca2+ enters through the NaV channel pore, the optically resolved ICa in the axon initial segment overlapped with the activation kinetics of NaV channels and heterologous expression of NaV1.2 in HEK-293 cells revealed a tetrodotoxin-sensitive [Ca2+]i rise. Finally, computational simulations predicted that axonal [Ca2+]i transients reflect a 0.4% Ca2+ conductivity of NaV channels. The findings indicate that Ca2+ permeation through NaV channels provides a submillisecond rapid entry route in NaV-enriched domains of mammalian axons. Nerve cells communicate using tiny electrical impulses called action potentials. Special proteins termed ion channels produce these electric signals by allowing specific charged particles, or ions, to pass in or out of cells across its membrane. When a nerve cell ‘fires’ an action potential, specific ion channels briefly open to let in a surge of positively charged ions which electrify the cell. Action potentials begin in the same place in each nerve cell, at an area called the axon initial segment. The large number of sodium channels at this site kick-start the influx of positively charged sodium ions ensuring that every action potential starts from the same place. Previous research has shown that, when action potentials begin, the concentration of calcium ions at the axon initial segment also increases, but it was not clear which ion channels were responsible for this entry of calcium. Channels that are selective for calcium ions are the prime candidates for this process. However, research in squid nerve cells gave rise to an unexpected idea by suggesting that sodium channels may not exclusively let in sodium but also allow some calcium ions to pass through. Hanemaaijer, Popovic et al. therefore wanted to test the routes that calcium ions take and see whether the sodium channels in mammalian nerve cells are also permeable to calcium. Experiments using fluorescent dyes to track the concentration of calcium in rat and human nerve cells showed that calcium ions accumulated at the axon initial segment when action potentials fired. Most of this increase in calcium could be stopped by treating the neurons with a toxin that prevents sodium channels from opening. Electrical manipulations of the cells revealed that, in this context, the calcium ions were effectively behaving like sodium ions. Human kidney cells were then engineered to produce the sodium channel protein. This confirmed that calcium and sodium ions were indeed both passing through the same channel. These results shed new light on the relationship between calcium ions and sodium channels within the mammalian nervous system and that this interplay occurs at the axon initial segment of the cell. Genetic mutations that ‘nudge’ sodium channels towards favoring calcium entry are also found in patients with autism spectrum disorders, and so this new finding may contribute to our understanding of these conditions.
Collapse
Affiliation(s)
- Naomi Ak Hanemaaijer
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands.,Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Marko A Popovic
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Xante Wilders
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Sara Grasman
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Oriol Pavón Arocas
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Maarten Hp Kole
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands.,Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
8
|
Dürvanger Z, Harmat V. Structural Diversity in Calmodulin - Peptide Interactions. Curr Protein Pept Sci 2020; 20:1102-1111. [PMID: 31553290 DOI: 10.2174/1389203720666190925101937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/13/2019] [Accepted: 04/12/2019] [Indexed: 01/17/2023]
Abstract
Calmodulin (CaM) is a highly conserved eukaryotic Ca2+ sensor protein that is able to bind a large variety of target sequences without a defined consensus sequence. The recognition of this diverse target set allows CaM to take part in the regulation of several vital cell functions. To fully understand the structural basis of the regulation functions of CaM, the investigation of complexes of CaM and its targets is essential. In this minireview we give an outline of the different types of CaM - peptide complexes with 3D structure determined, also providing an overview of recently determined structures. We discuss factors defining the orientations of peptides within the complexes, as well as roles of anchoring residues. The emphasis is on complexes where multiple binding modes were found.
Collapse
Affiliation(s)
- Zsolt Dürvanger
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Veronika Harmat
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary.,MTA-ELTE Protein Modelling Research Group, Budapest, Hungary
| |
Collapse
|
9
|
Li L, Lai M, Cole S, Le Novère N, Edelstein SJ. Neurogranin stimulates Ca2+/calmodulin-dependent kinase II by suppressing calcineurin activity at specific calcium spike frequencies. PLoS Comput Biol 2020; 16:e1006991. [PMID: 32049957 PMCID: PMC7041932 DOI: 10.1371/journal.pcbi.1006991] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 02/25/2020] [Accepted: 11/18/2019] [Indexed: 11/18/2022] Open
Abstract
Calmodulin sits at the center of molecular mechanisms underlying learning and memory. Its complex and sometimes opposite influences, mediated via the binding to various proteins, are yet to be fully understood. Calcium/calmodulin-dependent protein kinase II (CaMKII) and calcineurin (CaN) both bind open calmodulin, favoring Long-Term Potentiation (LTP) or Depression (LTD) respectively. Neurogranin binds to the closed conformation of calmodulin and its impact on synaptic plasticity is less clear. We set up a mechanistic computational model based on allosteric principles to simulate calmodulin state transitions and its interactions with calcium ions and the three binding partners mentioned above. We simulated calcium spikes at various frequencies and show that neurogranin regulates synaptic plasticity along three modalities. At low spike frequencies, neurogranin inhibits the onset of LTD by limiting CaN activation. At intermediate frequencies, neurogranin facilitates LTD, but limits LTP by precluding binding of CaMKII with calmodulin. Finally, at high spike frequencies, neurogranin promotes LTP by enhancing CaMKII autophosphorylation. While neurogranin might act as a calmodulin buffer, it does not significantly preclude the calmodulin opening by calcium. On the contrary, neurogranin synchronizes the opening of calmodulin's two lobes and promotes their activation at specific frequencies. Neurogranin suppresses basal CaN activity, thus increasing the chance of CaMKII trans-autophosphorylation at high-frequency calcium spikes. Taken together, our study reveals dynamic regulatory roles played by neurogranin on synaptic plasticity, which provide mechanistic explanations for opposing experimental findings.
Collapse
Affiliation(s)
- Lu Li
- Babraham Institute, Cambridge, United Kingdom
| | - Massimo Lai
- Quantitative Systems Pharmacology, CERTARA, Canterbury, United Kingdom
| | - Stephen Cole
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | | | | |
Collapse
|
10
|
Pan Y, Cummins TR. Distinct functional alterations in SCN8A epilepsy mutant channels. J Physiol 2020; 598:381-401. [PMID: 31715021 PMCID: PMC7216308 DOI: 10.1113/jp278952] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/12/2019] [Indexed: 01/12/2023] Open
Abstract
KEY POINTS Mutations in the SCN8A gene cause early infantile epileptic encephalopathy. We characterize a new epilepsy-related SCN8A mutation, R850Q, in the human SCN8A channel and present gain-of-function properties of the mutant channel. Systematic comparison of R850Q with three other SCN8A epilepsy mutations, T761I, R1617Q and R1872Q, identifies one common dysfunction in resurgent current, although these mutations alter distinct properties of the channel. Computational simulations in two different neuron models predict an increased excitability of neurons carrying these mutations, which explains the over-excitation that underlies seizure activities in patients. These data provide further insight into the mechanism of SCN8A-related epilepsy and reveal subtle but potentially important distinction of functional characterization performed in the human vs. rodent channels. ABSTRACT SCN8A is a novel causal gene for early infantile epileptic encephalopathy. It is well accepted that gain-of-function mutations in SCN8A underlie the disorder, although the remarkable heterogeneity of its clinical presentation and poor treatment response demand a better understanding of the disease mechanisms. Here, we characterize a new epilepsy-related SCN8A mutation, R850Q, in human Nav1.6. We show that it is a gain-of-function mutation, with a hyperpolarizing shift in voltage dependence of activation, a two-fold increase of persistent current and a slowed decay of resurgent current. We systematically compare its biophysics with three other SCN8A epilepsy mutations, T767I, R1617Q and R1872Q, in the human Nav1.6 channel. Although all of these mutations are gain-of-function, the mutations affect different aspects of channel properties. One commonality that we discovered is an alteration of resurgent current kinetics, although the mechanisms by which resurgent currents are augmented remain unclear for all of the mutations. Computational simulations predict an increased excitability of neurons carrying these mutations with differential enhancement by open channel blockade.
Collapse
Affiliation(s)
- Yanling Pan
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, IN, USA
| | - Theodore R Cummins
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, IN, USA
- Department of Biology, School of Science, IUPUI, IN, USA
| |
Collapse
|
11
|
Zhang M, Li Z, Jang H, Hedman AC, Sacks DB, Nussinov R. Ca 2+-Dependent Switch of Calmodulin Interaction Mode with Tandem IQ Motifs in the Scaffolding Protein IQGAP1. Biochemistry 2019; 58:4903-4911. [PMID: 31724397 PMCID: PMC8195445 DOI: 10.1021/acs.biochem.9b00854] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
IQ domain GTPase-activating scaffolding protein 1 (IQGAP1) mediates cytoskeleton, cell migration, proliferation, and apoptosis events. Calmodulin (CaM) modulates IQGAP1 functions by binding to its four tandem IQ motifs. Exactly how CaM binds the IQ motifs and which functions of IQGAP1 CaM regulates and how are fundamental mechanistic questions. We combine experimental pull-down assays, mutational data, and molecular dynamics simulations to understand the IQ-CaM complexes with and without Ca2+ at the atomic level. Apo-CaM favors the IQ3 and IQ4 motifs but not the IQ1 and IQ2 motifs that lack two hydrophobic residues for interactions with apo-CaM's hydrophobic pocket. Ca2+-CaM binds all four IQ motifs, with both N- and C-lobes tightly wrapped around each motif. Ca2+ promotes IQ-CaM interactions and increases the amount of IQGAP1-loaded CaM for IQGAP1-mediated signaling. Collectively, we describe IQ-CaM binding in atomistic detail and feature the emergence of Ca2+ as a key modulator of the CaM-IQGAP1 interactions.
Collapse
Affiliation(s)
- Mingzhen Zhang
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 20892, United States
| | - Zhigang Li
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hyunbum Jang
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Andrew C. Hedman
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - David B. Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ruth Nussinov
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 20892, United States
- Sackler Institute of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
12
|
Kawasaki H, Soma N, Kretsinger RH. Molecular Dynamics Study of the Changes in Conformation of Calmodulin with Calcium Binding and/or Target Recognition. Sci Rep 2019; 9:10688. [PMID: 31337841 PMCID: PMC6650393 DOI: 10.1038/s41598-019-47063-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 07/10/2019] [Indexed: 11/12/2022] Open
Abstract
Calmodulin is a calcium binding protein with two lobes, N-lobe and C-lobe, which evolved from duplication and fusion of a single precursor lobe of a pair of EF-hand. These two lobes of calmodulin show subtle differences in calcium binding and target recognition; these are important for the functions of calmodulin. Since the structures, especially main chain conformations, of two EF-lobes in holo-form are quite similar; this is a good example to evaluate the effect of side chains for structural dynamics. We analyzed the structure of calmodulin using molecular dynamics and found differences in conformational ensembles between N- and C-lobes. We also showed the mutant structures created by homology modeling could reproduce the difference of dynamic motion between N- and C-lobes.
Collapse
Affiliation(s)
- Hiroshi Kawasaki
- Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan.
| | - Natsumi Soma
- Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Robert H Kretsinger
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA
| |
Collapse
|
13
|
Johnson CN. Calcium modulation of cardiac sodium channels. J Physiol 2019; 598:2835-2846. [PMID: 30707447 DOI: 10.1113/jp277553] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 01/08/2019] [Indexed: 12/28/2022] Open
Abstract
Modification of voltage-gated Na+ channel (NaV ) function by intracellular Ca2+ has been a topic of much controversy. Early studies relied on measuring NaV function in the absence or presence of intracellular Ca2+ , and generated seemingly disparate results. Subsequent investigations revealed the mechanism(s) of Ca2+ -driven NaV modulation are complex and involve multiple accessory proteins. The Ca2+ -sensing protein calmodulin (CaM) has a central role in tuning NaV function to [Ca2+ ]i , but the mechanism has been obscured by other proteins (such as fibroblast growth factors (FGF) or CaM-dependent kinase II (CaMKII)) that can also modify channel function or exert an influence in a Ca2+ -dependent manner. Significant progress has been made in understanding the architecture of full-length ion channels and the structural and biophysical details of NaV -accessory protein interactions. Interdisciplinary structure-function studies are beginning to resolve the effect each interaction has on NaV gating. Carefully designed structure-guided or strategically selected disease-associated mutations are able to impair NaV -accessory protein interactions without altering other properties of channel function. Recently CaM was found to engage part of NaV 1.5 that is required for channel inactivation with high affinity. Careful impairment of this interaction disrupted NaV 1.5's ability to recover from inactivation. Such results support a paradigm of CaM-facilitated recovery from inactivation (CFRI). How NaV -CaM, CaMKII and FGF/fibroblast growth factor homologous factor interactions affect the timing or function of CFRI in cardiomyocytes remain open questions that are discussed herein. Moreover whether CFRI dysfunction or premature activation underlie certain NaV channelopathies are important questions that will require further investigation.
Collapse
Affiliation(s)
- Christopher N Johnson
- The Ohio State Wexner Medical Centre, Dorothy M. Davis Heart & Lung Research Institute, Columbus, OH, USA.,Vanderbilt Centre for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Nashville, TN, USA
| |
Collapse
|
14
|
Isbell HM, Kilpatrick AM, Lin Z, Mahling R, Shea MA. Backbone resonance assignments of complexes of apo human calmodulin bound to IQ motif peptides of voltage-dependent sodium channels Na V1.1, Na V1.4 and Na V1.7. BIOMOLECULAR NMR ASSIGNMENTS 2018; 12:283-289. [PMID: 29728980 PMCID: PMC6274588 DOI: 10.1007/s12104-018-9824-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/29/2018] [Indexed: 06/08/2023]
Abstract
Human voltage-gated sodium (NaV) channels are critical for initiating and propagating action potentials in excitable cells. Nine isoforms have different roles but similar topologies, with a pore-forming α-subunit and auxiliary transmembrane β-subunits. NaV pathologies lead to debilitating conditions including epilepsy, chronic pain, cardiac arrhythmias, and skeletal muscle paralysis. The ubiquitous calcium sensor calmodulin (CaM) binds to an IQ motif in the C-terminal tail of the α-subunit of all NaV isoforms, and contributes to calcium-dependent pore-gating in some channels. Previous structural studies of calcium-free (apo) CaM bound to the IQ motifs of NaV1.2, NaV1.5, and NaV1.6 showed that CaM binding was mediated by the C-domain of CaM (CaMC), while the N-domain (CaMN) made no detectable contacts. To determine whether this domain-specific recognition mechanism is conserved in other NaV isoforms, we used solution NMR spectroscopy to assign the backbone resonances of complexes of apo CaM bound to peptides of IQ motifs of NaV1.1, NaV1.4, and NaV1.7. Analysis of chemical shift differences showed that peptide binding only perturbed resonances in CaMC; resonances of CaMN were identical to free CaM. Thus, CaMC residues contribute to the interface with the IQ motif, while CaMN is available to interact elsewhere on the channel.
Collapse
Affiliation(s)
- Holly M Isbell
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242-1109, USA
| | - Adina M Kilpatrick
- Department of Physics and Astronomy, Drake University, Des Moines, IA, 50311-4516, USA
| | - Zesen Lin
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242-1109, USA
| | - Ryan Mahling
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242-1109, USA
| | - Madeline A Shea
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242-1109, USA.
| |
Collapse
|
15
|
Protein Engineering Allows for Mild Affinity-based Elution of Therapeutic Antibodies. J Mol Biol 2018; 430:3427-3438. [DOI: 10.1016/j.jmb.2018.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/25/2018] [Accepted: 06/04/2018] [Indexed: 01/08/2023]
|
16
|
Scala F, Nenov MN, Crofton EJ, Singh AK, Folorunso O, Zhang Y, Chesson BC, Wildburger NC, James TF, Alshammari MA, Alshammari TK, Elfrink H, Grassi C, Kasper JM, Smith AE, Hommel JD, Lichti CF, Rudra JS, D'Ascenzo M, Green TA, Laezza F. Environmental Enrichment and Social Isolation Mediate Neuroplasticity of Medium Spiny Neurons through the GSK3 Pathway. Cell Rep 2018; 23:555-567. [PMID: 29642012 PMCID: PMC6150488 DOI: 10.1016/j.celrep.2018.03.062] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/05/2018] [Accepted: 03/14/2018] [Indexed: 11/29/2022] Open
Abstract
Resilience and vulnerability to neuropsychiatric disorders are linked to molecular changes underlying excitability that are still poorly understood. Here, we identify glycogen-synthase kinase 3β (GSK3β) and voltage-gated Na+ channel Nav1.6 as regulators of neuroplasticity induced by environmentally enriched (EC) or isolated (IC) conditions-models for resilience and vulnerability. Transcriptomic studies in the nucleus accumbens from EC and IC rats predicted low levels of GSK3β and SCN8A mRNA as a protective phenotype associated with reduced excitability in medium spiny neurons (MSNs). In vivo genetic manipulations demonstrate that GSK3β and Nav1.6 are molecular determinants of MSN excitability and that silencing of GSK3β prevents maladaptive plasticity of IC MSNs. In vitro studies reveal direct interaction of GSK3β with Nav1.6 and phosphorylation at Nav1.6T1936 by GSK3β. A GSK3β-Nav1.6T1936 competing peptide reduces MSNs excitability in IC, but not EC rats. These results identify GSK3β regulation of Nav1.6 as a biosignature of MSNs maladaptive plasticity.
Collapse
Affiliation(s)
- Federico Scala
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Biophysics Graduate Program, Institute of Human Physiology, Università Cattolica, Rome, Italy
| | - Miroslav N Nenov
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Elizabeth J Crofton
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Neuroscience Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Aditya K Singh
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Oluwarotimi Folorunso
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Yafang Zhang
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Brent C Chesson
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Norelle C Wildburger
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Thomas F James
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Neuroscience Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA; Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - Tahani K Alshammari
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA; Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - Hannah Elfrink
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Bench Tutorials Program: Scientific Research and Design, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - James M Kasper
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Ashley E Smith
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA; Cell Biology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Jonathan D Hommel
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Cheryl F Lichti
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Jai S Rudra
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | | | - Thomas A Green
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA.
| |
Collapse
|
17
|
Westerlund AM, Delemotte L. Effect of Ca2+ on the promiscuous target-protein binding of calmodulin. PLoS Comput Biol 2018; 14:e1006072. [PMID: 29614072 PMCID: PMC5898786 DOI: 10.1371/journal.pcbi.1006072] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/13/2018] [Accepted: 03/07/2018] [Indexed: 01/05/2023] Open
Abstract
Calmodulin (CaM) is a calcium sensing protein that regulates the function of a large number of proteins, thus playing a crucial part in many cell signaling pathways. CaM has the ability to bind more than 300 different target peptides in a Ca2+-dependent manner, mainly through the exposure of hydrophobic residues. How CaM can bind a large number of targets while retaining some selectivity is a fascinating open question. Here, we explore the mechanism of CaM selective promiscuity for selected target proteins. Analyzing enhanced sampling molecular dynamics simulations of Ca2+-bound and Ca2+-free CaM via spectral clustering has allowed us to identify distinct conformational states, characterized by interhelical angles, secondary structure determinants and the solvent exposure of specific residues. We searched for indicators of conformational selection by mapping solvent exposure of residues in these conformational states to contacts in structures of CaM/target peptide complexes. We thereby identified CaM states involved in various binding classes arranged along a depth binding gradient. Binding Ca2+ modifies the accessible hydrophobic surface of the two lobes and allows for deeper binding. Apo CaM indeed shows shallow binding involving predominantly polar and charged residues. Furthermore, binding to the C-terminal lobe of CaM appears selective and involves specific conformational states that can facilitate deep binding to target proteins, while binding to the N-terminal lobe appears to happen through a more flexible mechanism. Thus the long-ranged electrostatic interactions of the charged residues of the N-terminal lobe of CaM may initiate binding, while the short-ranged interactions of hydrophobic residues in the C-terminal lobe of CaM may account for selectivity. This work furthers our understanding of the mechanism of CaM binding and selectivity to different target proteins and paves the way towards a comprehensive model of CaM selectivity.
Collapse
Affiliation(s)
- Annie M. Westerlund
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Lucie Delemotte
- Science for Life Laboratory, Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
18
|
Direct visualization of interaction between calmodulin and connexin45. Biochem J 2017; 474:4035-4051. [PMID: 28963343 DOI: 10.1042/bcj20170426] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/12/2017] [Accepted: 09/25/2017] [Indexed: 01/21/2023]
Abstract
Calmodulin (CaM) is an intracellular Ca2+ transducer involved in numerous activities in a broad Ca2+ signaling network. Previous studies have suggested that the Ca2+/CaM complex may participate in gap junction regulation via interaction with putative CaM-binding motifs in connexins; however, evidence of direct interactions between CaM and connexins has remained elusive to date due to challenges related to the study of membrane proteins. Here, we report the first direct interaction of CaM with Cx45 (connexin45) of γ-family in living cells under physiological conditions by monitoring bioluminescence resonance energy transfer. The interaction between CaM and Cx45 in cells is strongly dependent on intracellular Ca2+ concentration and can be blocked by the CaM inhibitor, N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide hydrochloride (W7). We further reveal a CaM-binding site at the cytosolic loop (residues 164-186) of Cx45 using a peptide model. The strong binding (Kd ∼ 5 nM) observed between CaM and Cx45 peptide, monitored by fluorescence-labeled CaM, is found to be Ca2+-dependent. Furthermore, high-resolution nuclear magnetic resonance spectroscopy reveals that CaM and Cx45 peptide binding leads to global chemical shift changes of 15N-labeled CaM, but does not alter the size of the structure. Observations involving both N- and C-domains of CaM to interact with the Cx45 peptide differ from the embraced interaction with Cx50 from another connexin family. Such interaction further increases Ca2+ sensitivity of CaM, especially at the N-terminal domain. Results of the present study suggest that both helicity and the interaction mode of the cytosolic loop are likely to contribute to CaM's modulation of connexins.
Collapse
|
19
|
Mahling R, Kilpatrick AM, Shea MA. Backbone resonance assignments of complexes of human voltage-dependent sodium channel Na V1.2 IQ motif peptide bound to apo calmodulin and to the C-domain fragment of apo calmodulin. BIOMOLECULAR NMR ASSIGNMENTS 2017; 11:297-303. [PMID: 28823028 PMCID: PMC5791537 DOI: 10.1007/s12104-017-9767-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/12/2017] [Indexed: 06/07/2023]
Abstract
Human voltage-gated sodium channel NaV1.2 has a single pore-forming α-subunit and two transmembrane β-subunits. Expressed primarily in the brain, NaV1.2 is critical for initiation and propagation of action potentials. Milliseconds after the pore opens, sodium influx is terminated by inactivation processes mediated by regulatory proteins including calmodulin (CaM). Both calcium-free (apo) CaM and calcium-saturated CaM bind tightly to an IQ motif in the C-terminal tail of the α-subunit. Our thermodynamic studies and solution structure (2KXW) of a C-domain fragment of apo 13C,15N- CaM (CaMC) bound to an unlabeled peptide with the sequence of rat NaV1.2 IQ motif showed that apo CaMC (a) was necessary and sufficient for binding, and (b) bound more favorably than calcium-saturated CaMC. However, we could not monitor the NaV1.2 residues directly, and no structure of full-length CaM (including the N-domain of CaM (CaMN)) was determined. To distinguish contributions of CaMN and CaMC, we used solution NMR spectroscopy to assign the backbone resonances of a complex containing a 13C,15N-labeled peptide with the sequence of human NaV1.2 IQ motif (NaV1.2IQp) bound to apo 13C,15N-CaM or apo 13C,15N-CaMC. Comparing the assignments of apo CaM in complex with NaV1.2IQp to those of free apo CaM showed that residues within CaMC were significantly perturbed, while residues within CaMN were essentially unchanged. The chemical shifts of residues in NaV1.2IQp and in the C-domain of CaM were nearly identical regardless of whether CaMN was covalently linked to CaMC. This suggests that CaMN does not influence apo CaM binding to NaV1.2IQp.
Collapse
Affiliation(s)
- Ryan Mahling
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242-1109, USA
| | - Adina M Kilpatrick
- Department of Physics and Astronomy, Drake University, Des Moines, IA, 50311-4516, USA
| | - Madeline A Shea
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242-1109, USA.
| |
Collapse
|
20
|
Fischer C, DeFalco TA, Karia P, Snedden WA, Moeder W, Yoshioka K, Dietrich P. Calmodulin as a Ca2+-Sensing Subunit of Arabidopsis Cyclic Nucleotide-Gated Channel Complexes. PLANT & CELL PHYSIOLOGY 2017; 58:1208-1221. [PMID: 28419310 DOI: 10.1093/pcp/pcx052] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 04/06/2017] [Indexed: 05/23/2023]
Abstract
Ca2+ serves as a universal second messenger in eukaryotic signaling pathways, and the spatial and temporal patterns of Ca2+ concentration changes are determined by feedback and feed-forward regulation of the involved transport proteins. Cyclic nucleotide-gated channels (CNGCs) are Ca2+-permeable channels that interact with the ubiquitous Ca2+ sensor calmodulin (CaM). CNGCs interact with CaMs via diverse CaM-binding sites, including an IQ-motif, which has been identified in the C-termini of CNGC20 and CNGC12. Here we present a family-wide analysis of the IQ-motif from all 20 Arabidopsis CNGC isoforms. While most of their IQ-peptides interacted with conserved CaMs in yeast, some were unable to do so, despite high sequence conservation across the family. We showed that the CaM binding ability of the IQ-motif is highly dependent on its proximal and distal vicinity. We determined that two alanine residues positioned N-terminal to the core IQ-sequence play a significant role in CaM binding, and identified a polymorphism at this site that promoted or inhibited CaM binding in yeast. Through detailed biophysical analysis of the CNGC2 IQ-motif, we found that this polymorphism specifically affected the Ca2+-independent interactions with the C-lobe of CaM. This same polymorphism partially suppressed the induction of programmed cell death by CNGC11/12 in planta. Our work expands the model of CNGC regulation, and posits that the C-lobe of apo-CaM is permanently associated with the channel at the N-terminal part of the IQ-domain. This mode allows CaM to function as a Ca2+-sensing regulatory subunit of the channel complex, providing a mechanism by which Ca2+ signals may be fine-tuned.
Collapse
Affiliation(s)
- Cornelia Fischer
- Molecular Plant Physiology, Department of Biology, University of Erlangen-Nürnberg, Staudtstrasse 5, D-91058 Erlangen, Germany
| | - Thomas A DeFalco
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada, M5S 3B2
| | - Purva Karia
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada, M5S 3B2
| | - Wayne A Snedden
- Department of Biology, Biosciences Complex, Queen's University, Kingston, ON, Canada, K7L 3N6
| | - Wolfgang Moeder
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada, M5S 3B2
| | - Keiko Yoshioka
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada, M5S 3B2
- Center for the Analysis of Genome Evolution and Function (CAGEF), University of Toronto, Toronto, ON, Canada, M5S 3B2
| | - Petra Dietrich
- Molecular Plant Physiology, Department of Biology, University of Erlangen-Nürnberg, Staudtstrasse 5, D-91058 Erlangen, Germany
| |
Collapse
|
21
|
Hovey L, Fowler CA, Mahling R, Lin Z, Miller MS, Marx DC, Yoder JB, Kim EH, Tefft KM, Waite BC, Feldkamp MD, Yu L, Shea MA. Calcium triggers reversal of calmodulin on nested anti-parallel sites in the IQ motif of the neuronal voltage-dependent sodium channel Na V1.2. Biophys Chem 2017; 224:1-19. [PMID: 28343066 PMCID: PMC5503752 DOI: 10.1016/j.bpc.2017.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/23/2017] [Accepted: 02/23/2017] [Indexed: 01/26/2023]
Abstract
Several members of the voltage-gated sodium channel family are regulated by calmodulin (CaM) and ionic calcium. The neuronal voltage-gated sodium channel NaV1.2 contains binding sites for both apo (calcium-depleted) and calcium-saturated CaM. We have determined equilibrium dissociation constants for rat NaV1.2 IQ motif [IQRAYRRYLLK] binding to apo CaM (~3nM) and (Ca2+)4-CaM (~85nM), showing that apo CaM binding is favored by 30-fold. For both apo and (Ca2+)4-CaM, NMR demonstrated that NaV1.2 IQ motif peptide (NaV1.2IQp) exclusively made contacts with C-domain residues of CaM (CaMC). To understand how calcium triggers conformational change at the CaM-IQ interface, we determined a solution structure (2M5E.pdb) of (Ca2+)2-CaMC bound to NaV1.2IQp. The polarity of (Ca2+)2-CaMC relative to the IQ motif was opposite to that seen in apo CaMC-Nav1.2IQp (2KXW), revealing that CaMC recognizes nested, anti-parallel sites in Nav1.2IQp. Reversal of CaM may require transient release from the IQ motif during calcium binding, and facilitate a re-orientation of CaMN allowing interactions with non-IQ NaV1.2 residues or auxiliary regulatory proteins interacting in the vicinity of the IQ motif.
Collapse
Affiliation(s)
- Liam Hovey
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - C Andrew Fowler
- NMR Facility, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242-1109 Iowa City, United States
| | - Ryan Mahling
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Zesen Lin
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Mark Stephen Miller
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Dagan C Marx
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Jesse B Yoder
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Elaine H Kim
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Kristin M Tefft
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Brett C Waite
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Michael D Feldkamp
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States
| | - Liping Yu
- NMR Facility, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242-1109 Iowa City, United States
| | - Madeline A Shea
- Department of Biochemistry, University of Iowa, 52242-1109 Iowa City, United States.
| |
Collapse
|
22
|
Yan H, Wang C, Marx SO, Pitt GS. Calmodulin limits pathogenic Na+ channel persistent current. J Gen Physiol 2017; 149:277-293. [PMID: 28087622 PMCID: PMC5299624 DOI: 10.1085/jgp.201611721] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/13/2016] [Accepted: 12/19/2016] [Indexed: 01/29/2023] Open
Abstract
The molecular mechanisms controlling “persistent” current through voltage-gated Na+ channels are poorly understood. Yan et al. show that apocalmodulin binding to the intracellular C-terminal domain limits persistent Na+ flux and accelerates inactivation across the voltage-gated Na+ channel family. Increased “persistent” current, caused by delayed inactivation, through voltage-gated Na+ (NaV) channels leads to cardiac arrhythmias or epilepsy. The underlying molecular contributors to these inactivation defects are poorly understood. Here, we show that calmodulin (CaM) binding to multiple sites within NaV channel intracellular C-terminal domains (CTDs) limits persistent Na+ current and accelerates inactivation across the NaV family. Arrhythmia or epilepsy mutations located in NaV1.5 or NaV1.2 channel CTDs, respectively, reduce CaM binding either directly or by interfering with CTD–CTD interchannel interactions. Boosting the availability of CaM, thus shifting its binding equilibrium, restores wild-type (WT)–like inactivation in mutant NaV1.5 and NaV1.2 channels and likewise diminishes the comparatively large persistent Na+ current through WT NaV1.6, whose CTD displays relatively low CaM affinity. In cerebellar Purkinje neurons, in which NaV1.6 promotes a large physiological persistent Na+ current, increased CaM diminishes the persistent Na+ current, suggesting that the endogenous, comparatively weak affinity of NaV1.6 for apoCaM is important for physiological persistent current.
Collapse
Affiliation(s)
- Haidun Yan
- Ion Channel Research Unit, Duke University Medical Center, Durham, NC 27710
| | - Chaojian Wang
- Ion Channel Research Unit, Duke University Medical Center, Durham, NC 27710
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Geoffrey S Pitt
- Ion Channel Research Unit, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
23
|
Novel SCN9A mutations underlying extreme pain phenotypes: unexpected electrophysiological and clinical phenotype correlations. J Neurosci 2015; 35:7674-81. [PMID: 25995458 DOI: 10.1523/jneurosci.3935-14.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The importance of NaV1.7 (encoded by SCN9A) in the regulation of pain sensing is exemplified by the heterogeneity of clinical phenotypes associated with its mutation. Gain-of-function mutations are typically pain-causing and have been associated with inherited erythromelalgia (IEM) and paroxysmal extreme pain disorder (PEPD). IEM is usually caused by enhanced NaV1.7 channel activation, whereas mutations that alter steady-state fast inactivation often lead to PEPD. In contrast, nonfunctional mutations in SCN9A are known to underlie congenital insensitivity to pain (CIP). Although well documented, the correlation between SCN9A genotypes and clinical phenotypes is still unclear. Here we report three families with novel SCN9A mutations. In a multiaffected dominant family with IEM, we found the heterozygous change L245 V. Electrophysiological characterization showed that this mutation did not affect channel activation but instead resulted in incomplete fast inactivation and a small hyperpolarizing shift in steady-state slow inactivation, characteristics more commonly associated with PEPD. In two compound heterozygous CIP patients, we found mutations that still retained functionality of the channels, with two C-terminal mutations (W1775R and L1831X) exhibiting a depolarizing shift in channel activation. Two mutations (A1236E and L1831X) resulted in a hyperpolarizing shift in steady-state fast inactivation. To our knowledge, these are the first descriptions of mutations with some retained channel function causing CIP. This study emphasizes the complex genotype-phenotype correlations that exist for SCN9A and highlights the C-terminal cytoplasmic region of NaV1.7 as a critical region for channel function, potentially facilitating analgesic drug development studies.
Collapse
|
24
|
Abstract
The structures of the cytosolic portion of voltage activated sodium channels (CTNav) in complexes with calmodulin and other effectors in the presence and the absence of calcium provide information about the mechanisms by which these effectors regulate channel activity. The most studied of these complexes, those of Nav1.2 and Nav1.5, show details of the conformations and the specific contacts that are involved in channel regulation. Another voltage activated sodium channel, Nav1.4, shows significant calcium dependent inactivation, while its homolog Nav1.5 does not. The available structures shed light on the possible localization of the elements responsible for this effect. Mutations in the genes of these 3 Nav channels are associated with several disease conditions: Nav1.2, neurological conditions; Nav1.4, syndromes involving skeletal muscle; and Nav1.5, cardiac arrhythmias. Many of these disease-specific mutations are located at the interfaces involving CTNav and its effectors.
Collapse
Affiliation(s)
- Sandra B Gabelli
- a Structural Enzymology and Thermodynamics Group; Department of Biophysics and Biophysical Chemistry , Johns Hopkins University School of Medicine , Baltimore , MD USA.,b Division of Cardiology; Department of Medicine, Johns Hopkins University School of Medicine ; Baltimore , MD USA.,c Department of Oncology ; Johns Hopkins University School of Medicine ; Baltimore , MD USA
| | - Jesse B Yoder
- a Structural Enzymology and Thermodynamics Group; Department of Biophysics and Biophysical Chemistry , Johns Hopkins University School of Medicine , Baltimore , MD USA
| | - Gordon F Tomaselli
- b Division of Cardiology; Department of Medicine, Johns Hopkins University School of Medicine ; Baltimore , MD USA
| | - L Mario Amzel
- a Structural Enzymology and Thermodynamics Group; Department of Biophysics and Biophysical Chemistry , Johns Hopkins University School of Medicine , Baltimore , MD USA
| |
Collapse
|
25
|
Heterologous expression of NaV1.9 chimeras in various cell systems. Pflugers Arch 2015; 467:2423-35. [PMID: 25916202 DOI: 10.1007/s00424-015-1709-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 03/31/2015] [Accepted: 04/16/2015] [Indexed: 01/27/2023]
Abstract
SCN11A encodes the voltage-gated sodium channel NaV1.9, which deviates most strongly from the other eight NaV channels expressed in mammals. It is characterized by resistance to the prototypic NaV channel blocker tetrodotoxin and exhibits slow activation and inactivation gating. Its expression in dorsal root ganglia neurons suggests a role in motor or pain signaling functions as also recently demonstrated by the occurrence of various mutations in human SCN11A leading to altered pain sensation syndromes. The systematic investigation of human NaV1.9, however, is severely hampered because of very poor heterologous expression in host cells. Using patch-clamp and two-electrode voltage-clamp methods, we show that this limitation is caused by the C-terminal structure of NaV1.9. A chimera of NaV1.9 harboring the C terminus of NaV1.4 yields functional expression not only in neuronal cells but also in non-excitable cells, such as HEK 293T or Xenopus oocytes. The major functional difference of the chimeric channel with respect to NaV1.9 is an accelerated activation and inactivation. Since the entire transmembrane domain is preserved, it is suited for studying pharmacological properties of the channel and the functional impact of disease-causing mutations. Moreover, we demonstrate how mutation S360Y makes NaV1.9 channels sensitive to tetrodotoxin and saxitoxin and that the unusual slow open-state inactivation of NaV1.9 is also mediated by the IFM (isoleucine-phenylalanine-methionine) inactivation motif located in the linker connecting domains III and IV.
Collapse
|
26
|
Lai M, Brun D, Edelstein SJ, Le Novère N. Modulation of calmodulin lobes by different targets: an allosteric model with hemiconcerted conformational transitions. PLoS Comput Biol 2015; 11:e1004063. [PMID: 25611683 PMCID: PMC4303274 DOI: 10.1371/journal.pcbi.1004063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 11/26/2014] [Indexed: 01/30/2023] Open
Abstract
Calmodulin is a calcium-binding protein ubiquitous in eukaryotic cells, involved in numerous calcium-regulated biological phenomena, such as synaptic plasticity, muscle contraction, cell cycle, and circadian rhythms. It exibits a characteristic dumbell shape, with two globular domains (N- and C-terminal lobe) joined by a linker region. Each lobe can take alternative conformations, affected by the binding of calcium and target proteins. Calmodulin displays considerable functional flexibility due to its capability to bind different targets, often in a tissue-specific fashion. In various specific physiological environments (e.g. skeletal muscle, neuron dendritic spines) several targets compete for the same calmodulin pool, regulating its availability and affinity for calcium. In this work, we sought to understand the general principles underlying calmodulin modulation by different target proteins, and to account for simultaneous effects of multiple competing targets, thus enabling a more realistic simulation of calmodulin-dependent pathways. We built a mechanistic allosteric model of calmodulin, based on an hemiconcerted framework: each calmodulin lobe can exist in two conformations in thermodynamic equilibrium, with different affinities for calcium and different affinities for each target. Each lobe was allowed to switch conformation on its own. The model was parameterised and validated against experimental data from the literature. In spite of its simplicity, a two-state allosteric model was able to satisfactorily represent several sets of experiments, in particular the binding of calcium on intact and truncated calmodulin and the effect of different skMLCK peptides on calmodulin's saturation curve. The model can also be readily extended to include multiple targets. We show that some targets stabilise the low calcium affinity T state while others stabilise the high affinity R state. Most of the effects produced by calmodulin targets can be explained as modulation of a pre-existing dynamic equilibrium between different conformations of calmodulin's lobes, in agreement with linkage theory and MWC-type models.
Collapse
Affiliation(s)
- Massimo Lai
- Babraham Institute, Cambridge, United Kingdom
- * E-mail:
| | - Denis Brun
- EMBL-EBI, Hinxton, United Kingdom
- Amadeus IT Group, Sophia Antipolis, France
| | | | - Nicolas Le Novère
- Babraham Institute, Cambridge, United Kingdom
- EMBL-EBI, Hinxton, United Kingdom
| |
Collapse
|
27
|
James TF, Nenov MN, Wildburger NC, Lichti CF, Luisi J, Vergara F, Panova-Electronova NI, Nilsson CL, Rudra JS, Green TA, Labate D, Laezza F. The Nav1.2 channel is regulated by GSK3. Biochim Biophys Acta Gen Subj 2015; 1850:832-44. [PMID: 25615535 DOI: 10.1016/j.bbagen.2015.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/17/2014] [Accepted: 01/14/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Phosphorylation plays an essential role in regulating voltage-gated sodium (Na(v)) channels and excitability. Yet, a surprisingly limited number of kinases have been identified as regulators of Na(v) channels. We posited that glycogen synthase kinase 3 (GSK3), a critical kinase found associated with numerous brain disorders, might directly regulate neuronal Na(v) channels. METHODS We used patch-clamp electrophysiology to record sodium currents from Na(v)1.2 channels stably expressed in HEK-293 cells. mRNA and protein levels were quantified with RT-PCR, Western blot, or confocal microscopy, and in vitro phosphorylation and mass spectrometry to identify phosphorylated residues. RESULTS We found that exposure of cells to GSK3 inhibitor XIII significantly potentiates the peak current density of Na(v)1.2, a phenotype reproduced by silencing GSK3 with siRNA. Contrarily, overexpression of GSK3β suppressed Na(v)1.2-encoded currents. Neither mRNA nor total protein expression was changed upon GSK3 inhibition. Cell surface labeling of CD4-chimeric constructs expressing intracellular domains of the Na(v)1.2 channel indicates that cell surface expression of CD4-Na(v)1.2 C-tail was up-regulated upon pharmacological inhibition of GSK3, resulting in an increase of surface puncta at the plasma membrane. Finally, using in vitro phosphorylation in combination with high resolution mass spectrometry, we further demonstrate that GSK3β phosphorylates T(1966) at the C-terminal tail of Na(v)1.2. CONCLUSION These findings provide evidence for a new mechanism by which GSK3 modulates Na(v) channel function via its C-terminal tail. GENERAL SIGNIFICANCE These findings provide fundamental knowledge in understanding signaling dysfunction common in several neuropsychiatric disorders.
Collapse
Affiliation(s)
- Thomas F James
- Department of Pharmacology & Toxicology, USA; Neuroscience Graduate Program, USA
| | | | - Norelle C Wildburger
- Department of Pharmacology & Toxicology, USA; Neuroscience Graduate Program, USA
| | | | | | | | | | | | - Jai S Rudra
- Department of Pharmacology & Toxicology, USA
| | - Thomas A Green
- Department of Pharmacology & Toxicology, USA; Center for Addiction Research, USA
| | | | - Fernanda Laezza
- Department of Pharmacology & Toxicology, USA; Center for Addiction Research, USA; Center for Biomedical Engineering, USA; Mitchell Center for Neurodegenerative Diseases, USA.
| |
Collapse
|
28
|
Gabelli SB, Boto A, HalperinKuhns V, Bianchet MA, Farinelli F, Aripirala S, Yoder J, Jakoncic J, Tomaselli GF, Amzel LM. Regulation of the NaV1.5 cytoplasmic domain by calmodulin. Nat Commun 2014; 5:5126. [PMID: 25370050 PMCID: PMC4223872 DOI: 10.1038/ncomms6126] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 09/02/2014] [Indexed: 12/23/2022] Open
Abstract
Voltage-gated sodium channels (Na(v)) underlie the rapid upstroke of action potentials in excitable tissues. Binding of channel-interactive proteins is essential for controlling fast and long-term inactivation. In the structure of the complex of the carboxy-terminal portion of Na(v)1.5 (CTNa(v)1.5) with calmodulin (CaM)-Mg(2+) reported here, both CaM lobes interact with the CTNa(v)1.5. On the basis of the differences between this structure and that of an inactivated complex, we propose that the structure reported here represents a non-inactivated state of the CTNa(v), that is, the state that is poised for activation. Electrophysiological characterization of mutants further supports the importance of the interactions identified in the structure. Isothermal titration calorimetry experiments show that CaM binds to CTNa(v)1.5 with high affinity. The results of this study provide unique insights into the physiological activation and the pathophysiology of Na(v) channels.
Collapse
Affiliation(s)
- Sandra B. Gabelli
- Structural Enzymology and Thermodynamics Group. Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe St, WBSB 608, Baltimore, Maryland 21205, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Bldg. 844, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Agedi Boto
- Structural Enzymology and Thermodynamics Group. Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe St, WBSB 608, Baltimore, Maryland 21205, USA
| | - Victoria HalperinKuhns
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Bldg. 844, Baltimore, MD 21205, USA
| | - Mario A. Bianchet
- Structural Enzymology and Thermodynamics Group. Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe St, WBSB 608, Baltimore, Maryland 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Baltimore, MD 21287, USA
| | - Federica Farinelli
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Bldg. 844, Baltimore, MD 21205, USA
| | - Srinivas Aripirala
- Structural Enzymology and Thermodynamics Group. Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe St, WBSB 608, Baltimore, Maryland 21205, USA
| | - Jesse Yoder
- Structural Enzymology and Thermodynamics Group. Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe St, WBSB 608, Baltimore, Maryland 21205, USA
| | - Jean Jakoncic
- Brookhaven National Laboratory, National Synchrotron Light Source, Upton, NY 11973
| | - Gordon F. Tomaselli
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Bldg. 844, Baltimore, MD 21205, USA
| | - L. Mario Amzel
- Structural Enzymology and Thermodynamics Group. Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe St, WBSB 608, Baltimore, Maryland 21205, USA
| |
Collapse
|
29
|
Kursula P. The many structural faces of calmodulin: a multitasking molecular jackknife. Amino Acids 2014; 46:2295-304. [PMID: 25005783 DOI: 10.1007/s00726-014-1795-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 06/22/2014] [Indexed: 12/16/2022]
Abstract
Calmodulin (CaM) is a highly conserved protein and a crucial calcium sensor in eukaryotes. CaM is a regulator of hundreds of diverse target proteins. A wealth of studies has been carried out on the structure of CaM, both in the unliganded form and in complexes with target proteins and peptides. The outcome of these studies points toward a high propensity to attain various conformational states, depending on the binding partner. The purpose of this review is to provide examples of different conformations of CaM trapped in the crystal state. In addition, comparisons are made to corresponding studies in solution. The different CaM conformations in crystal structures are also compared based on the positions of the metal ions bound to their EF hands, in terms of distances, angles, and pseudo-torsion angles. Possible caveats and artifacts in CaM crystal structures are discussed, as well as the possibilities of trapping biologically relevant CaM conformations in the crystal state.
Collapse
Affiliation(s)
- Petri Kursula
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland,
| |
Collapse
|
30
|
Stoilova-McPhie S, Ali S, Laezza F. Protein-Protein Interactions as New Targets for Ion Channel Drug Discovery. AUSTIN JOURNAL OF PHARMACOLOGY AND THERAPEUTICS 2013; 1:5. [PMID: 25485305 PMCID: PMC4255474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Protein-protein interactions (PPI) are key molecular elements that provide the basis of signaling in virtually all cellular processes. The precision and specificity of these molecular interactions have ignited a strong interest in pursuing PPI surfaces as new targets for drug discovery, especially against ion channels in the central nervous system (CNS) where selectivity and specificity are vital for developing drugs with limited side effects. Ion channels are large transmembrane domain proteins assembled with multiple regulatory proteins binding to the intracellular portion of channels. These macromolecular complexes are difficult to isolate, purify and reconstitute, posing a significant barrier in targeting these PPI for drug discovery purposes. Here, we will provide a short overview of salient features of PPI and discuss successful studies focusing on protein-channel interactions that could inspire new drug discovery campaigns targeting ion channel complexes.
Collapse
Affiliation(s)
- Svetla Stoilova-McPhie
- Department of Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Structural Biology and Molecular Biophysics, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Syed Ali
- Department Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Fernanda Laezza
- Department Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|