1
|
Yao X, Gao S, Yan N. Structural biology of voltage-gated calcium channels. Channels (Austin) 2024; 18:2290807. [PMID: 38062897 PMCID: PMC10761187 DOI: 10.1080/19336950.2023.2290807] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Voltage-gated calcium (Cav) channels mediate Ca2+ influx in response to membrane depolarization, playing critical roles in diverse physiological processes. Dysfunction or aberrant regulation of Cav channels can lead to life-threatening consequences. Cav-targeting drugs have been clinically used to treat cardiovascular and neuronal disorders for several decades. This review aims to provide an account of recent developments in the structural dissection of Cav channels. High-resolution structures have significantly advanced our understanding of the working and disease mechanisms of Cav channels, shed light on the molecular basis for their modulation, and elucidated the modes of actions (MOAs) of representative drugs and toxins. The progress in structural studies of Cav channels lays the foundation for future drug discovery efforts targeting Cav channelopathies.
Collapse
Affiliation(s)
- Xia Yao
- TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Shuai Gao
- TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| |
Collapse
|
2
|
Tian L, Andrews C, Yan Q, Yang JJ. Molecular regulation of calcium-sensing receptor (CaSR)-mediated signaling. Chronic Dis Transl Med 2024; 10:167-194. [PMID: 39027195 PMCID: PMC11252437 DOI: 10.1002/cdt3.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 07/20/2024] Open
Abstract
Calcium-sensing receptor (CaSR), a family C G-protein-coupled receptor, plays a crucial role in regulating calcium homeostasis by sensing small concentration changes of extracellular Ca2+, Mg2+, amino acids (e.g., L-Trp and L-Phe), small peptides, anions (e.g., HCO3 - and PO4 3-), and pH. CaSR-mediated intracellular Ca2+ signaling regulates a diverse set of cellular processes including gene transcription, cell proliferation, differentiation, apoptosis, muscle contraction, and neuronal transmission. Dysfunction of CaSR with mutations results in diseases such as autosomal dominant hypocalcemia, familial hypocalciuric hypercalcemia, and neonatal severe hyperparathyroidism. CaSR also influences calciotropic disorders, such as osteoporosis, and noncalciotropic disorders, such as cancer, Alzheimer's disease, and pulmonary arterial hypertension. This study first reviews recent advances in biochemical and structural determination of the framework of CaSR and its interaction sites with natural ligands, as well as exogenous positive allosteric modulators and negative allosteric modulators. The establishment of the first CaSR protein-protein interactome network revealed 94 novel players involved in protein processing in endoplasmic reticulum, trafficking, cell surface expression, endocytosis, degradation, and signaling pathways. The roles of these proteins in Ca2+-dependent cellular physiological processes and in CaSR-dependent cellular signaling provide new insights into the molecular basis of diseases caused by CaSR mutations and dysregulated CaSR activity caused by its protein interactors and facilitate the design of therapeutic agents that target CaSR and other family C G-protein-coupled receptors.
Collapse
Affiliation(s)
- Li Tian
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging FacilityGeorgia State UniversityAtlantaGeorgiaUSA
| | - Corey Andrews
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging FacilityGeorgia State UniversityAtlantaGeorgiaUSA
| | - Qiuyun Yan
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging FacilityGeorgia State UniversityAtlantaGeorgiaUSA
| | - Jenny J. Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging FacilityGeorgia State UniversityAtlantaGeorgiaUSA
| |
Collapse
|
3
|
Maraslioglu-Sperber A, Pizzi E, Fisch JO, Kattler K, Ritter T, Friauf E. Molecular and functional profiling of cell diversity and identity in the lateral superior olive, an auditory brainstem center with ascending and descending projections. Front Cell Neurosci 2024; 18:1354520. [PMID: 38846638 PMCID: PMC11153811 DOI: 10.3389/fncel.2024.1354520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/15/2024] [Indexed: 06/09/2024] Open
Abstract
The lateral superior olive (LSO), a prominent integration center in the auditory brainstem, contains a remarkably heterogeneous population of neurons. Ascending neurons, predominantly principal neurons (pLSOs), process interaural level differences for sound localization. Descending neurons (lateral olivocochlear neurons, LOCs) provide feedback into the cochlea and are thought to protect against acoustic overload. The molecular determinants of the neuronal diversity in the LSO are largely unknown. Here, we used patch-seq analysis in mice at postnatal days P10-12 to classify developing LSO neurons according to their functional and molecular profiles. Across the entire sample (n = 86 neurons), genes involved in ATP synthesis were particularly highly expressed, confirming the energy expenditure of auditory neurons. Two clusters were identified, pLSOs and LOCs. They were distinguished by 353 differentially expressed genes (DEGs), most of which were novel for the LSO. Electrophysiological analysis confirmed the transcriptomic clustering. We focused on genes affecting neuronal input-output properties and validated some of them by immunohistochemistry, electrophysiology, and pharmacology. These genes encode proteins such as osteopontin, Kv11.3, and Kvβ3 (pLSO-specific), calcitonin-gene-related peptide (LOC-specific), or Kv7.2 and Kv7.3 (no DEGs). We identified 12 "Super DEGs" and 12 genes showing "Cluster similarity." Collectively, we provide fundamental and comprehensive insights into the molecular composition of individual ascending and descending neurons in the juvenile auditory brainstem and how this may relate to their specific functions, including developmental aspects.
Collapse
Affiliation(s)
- Ayse Maraslioglu-Sperber
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Erika Pizzi
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Jonas O. Fisch
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Kathrin Kattler
- Genetics/Epigenetics Group, Department of Biological Sciences, Saarland University, Saarbrücken, Germany
| | - Tamara Ritter
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| |
Collapse
|
4
|
del Rivero Morfin PJ, Kochiss AL, Liedl KR, Flucher BE, Fernández-Quintero ML, Ben-Johny M. Asymmetric contribution of a selectivity filter gate in triggering inactivation of CaV1.3 channels. J Gen Physiol 2024; 156:e202313365. [PMID: 38175169 PMCID: PMC10771039 DOI: 10.1085/jgp.202313365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 10/08/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Voltage-dependent and Ca2+-dependent inactivation (VDI and CDI, respectively) of CaV channels are two biologically consequential feedback mechanisms that fine-tune Ca2+ entry into neurons and cardiomyocytes. Although known to be initiated by distinct molecular events, how these processes obstruct conduction through the channel pore remains poorly defined. Here, focusing on ultrahighly conserved tryptophan residues in the interdomain interfaces near the selectivity filter of CaV1.3, we demonstrate a critical role for asymmetric conformational changes in mediating VDI and CDI. Specifically, mutagenesis of the domain III-IV interface, but not others, enhanced VDI. Molecular dynamics simulations demonstrate that mutations in distinct selectivity filter interfaces differentially impact conformational flexibility. Furthermore, mutations in distinct domains preferentially disrupt CDI mediated by the N- versus C-lobes of CaM, thus uncovering a scheme of structural bifurcation of CaM signaling. These findings highlight the fundamental importance of the asymmetric arrangement of the pseudotetrameric CaV pore domain for feedback inhibition.
Collapse
Affiliation(s)
| | - Audrey L. Kochiss
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Klaus R. Liedl
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Bernhard E. Flucher
- Department of Physiology and Medical Physics, Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| |
Collapse
|
5
|
Zhang H, Hu S, Yang P, Long H, Ma Q, Yin D, Xu G. HDAC9-mediated calmodulin deacetylation induces memory impairment in Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14573. [PMID: 38421101 PMCID: PMC10850929 DOI: 10.1111/cns.14573] [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: 06/12/2023] [Revised: 11/20/2023] [Accepted: 12/04/2023] [Indexed: 03/02/2024] Open
Abstract
AIMS Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive dysfunction and memory impairment. AD pathology involves protein acetylation. Previous studies have mainly focused on histone acetylation in AD, however, the roles of nonhistone acetylation in AD are less explored. METHODS The protein acetylation and expression levels were detected by western blotting and co-immunoprecipitation. The stoichiometry of acetylation was measured by home-made and site-specific antibodies against acetylated-CaM (Ac-CaM) at K22, K95, and K116. Hippocampus-dependent learning and memory were evaluated by using the Morris water maze, novel object recognition, and contextual fear conditioning tests. RESULTS We showed that calmodulin (CaM) acetylation is reduced in plasma of AD patients and mice. CaM acetylation and its target Ca2+ /CaM-dependent kinase II α (CaMKIIα) activity were severely impaired in AD mouse brain. The stoichiometry showed that Ac-K22, K95-CaM acetylation were decreased in AD patients and mice. Moreover, we screened and identified that lysine deacetylase 9 (HDAC9) was the main deacetylase for CaM. In addition, HDAC9 inhibition increased CaM acetylation and CaMKIIα activity, and hippocampus-dependent memory in AD mice. CONCLUSIONS HDAC9-mediated CaM deacetylation induces memory impairment in AD, HDAC9, or CaM acetylation may become potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Hai‐Long Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSuzhou Medical College of Soochow University, Medical Center of Soochow UniversitySuzhouChina
| | - Shufen Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSuzhou Medical College of Soochow University, Medical Center of Soochow UniversitySuzhouChina
| | - Pin Yang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life ScienceEast China Normal UniversityShanghaiChina
| | - Han‐Chun Long
- Department of NeurologyThe Affiliated Xingyi City Hospital of Guizhou Medical UniversityXingyiChina
| | - Quan‐Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSuzhou Medical College of Soochow University, Medical Center of Soochow UniversitySuzhouChina
| | - Dong‐Min Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life ScienceEast China Normal UniversityShanghaiChina
| | - Guang‐Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSuzhou Medical College of Soochow University, Medical Center of Soochow UniversitySuzhouChina
| |
Collapse
|
6
|
Archer C. Isothermal Titration Calorimetry for Fragment-Based Analysis of Ion Channel Interactions. Methods Mol Biol 2024; 2796:271-289. [PMID: 38856907 DOI: 10.1007/978-1-0716-3818-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Ion channels are membrane proteins that may also have intracellular and extracellular domains that interact with other ligands. In many cases, these interaction sites are highly mobile and may undergo changes in the configuration upon binding with regulatory signaling molecules. Isothermal titration calorimetry (ITC) is a powerful technique to quantify protein-ligand interactions of purified samples in solution. This chapter describes a fragment-based analysis method using ITC to quantify the interactions between a domain of the voltage-gated Kv7 channel and the calcium-regulated protein calmodulin. This example can be used to quantify the interactions between specific domains of other ion channels and their regulatory signaling proteins.
Collapse
Affiliation(s)
- Crystal Archer
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
7
|
del Rivero Morfin PJ, Kochiss AL, Liedl KR, Flucher BE, Fernández-Quintero ML, Ben-Johny M. Asymmetric Contribution of a Selectivity Filter Gate in Triggering Inactivation of Ca V1.3 Channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558864. [PMID: 37790368 PMCID: PMC10542529 DOI: 10.1101/2023.09.21.558864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Voltage-dependent and Ca2+-dependent inactivation (VDI and CDI, respectively) of CaV channels are two biologically consequential feedback mechanisms that fine-tune Ca2+ entry into neurons and cardiomyocytes. Although known to be initiated by distinct molecular events, how these processes obstruct conduction through the channel pore remains poorly defined. Here, focusing on ultra-highly conserved tryptophan residues in the inter-domain interfaces near the selectivity filter of CaV1.3, we demonstrate a critical role for asymmetric conformational changes in mediating VDI and CDI. Specifically, mutagenesis of the domain III-IV interface, but not others, enhanced VDI. Molecular dynamics simulations demonstrate that mutations in distinct selectivity filter interfaces differentially impact conformational flexibility. Furthermore, mutations in distinct domains preferentially disrupt CDI mediated by the N- versus C-lobes of CaM, thus uncovering a scheme of structural bifurcation of CaM signaling. These findings highlight the fundamental importance of the asymmetric arrangement of the pseudo-tetrameric CaV pore domain for feedback inhibition.
Collapse
Affiliation(s)
| | - Audrey L. Kochiss
- Department of Physiology and Cellular Biophysics, Columbia University
| | - Klaus R. Liedl
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Bernhard E. Flucher
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | | | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University
| |
Collapse
|
8
|
Barret D, Schuster D, Rodrigues M, Leitner A, Picotti P, Schertler G, Kaupp U, Korkhov V, Marino J. Structural basis of calmodulin modulation of the rod cyclic nucleotide-gated channel. Proc Natl Acad Sci U S A 2023; 120:e2300309120. [PMID: 37011209 PMCID: PMC10104587 DOI: 10.1073/pnas.2300309120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/06/2023] [Indexed: 04/05/2023] Open
Abstract
Calmodulin (CaM) regulates many ion channels to control calcium entry into cells, and mutations that alter this interaction are linked to fatal diseases. The structural basis of CaM regulation remains largely unexplored. In retinal photoreceptors, CaM binds to the CNGB subunit of cyclic nucleotide-gated (CNG) channels and, thereby, adjusts the channel's Cyclic guanosine monophosphate (cGMP) sensitivity in response to changes in ambient light conditions. Here, we provide the structural characterization for CaM regulation of a CNG channel by using a combination of single-particle cryo-electron microscopy and structural proteomics. CaM connects the CNGA and CNGB subunits, resulting in structural changes both in the cytosolic and transmembrane regions of the channel. Cross-linking and limited proteolysis-coupled mass spectrometry mapped the conformational changes induced by CaM in vitro and in the native membrane. We propose that CaM is a constitutive subunit of the rod channel to ensure high sensitivity in dim light. Our mass spectrometry-based approach is generally relevant for studying the effect of CaM on ion channels in tissues of medical interest, where only minute quantities are available.
Collapse
Affiliation(s)
- Diane C. A. Barret
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
| | - Dina Schuster
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8049Zürich, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zürich, 8049Zurich, Switzerland
| | - Matthew J. Rodrigues
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
| | - Alexander Leitner
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8049Zürich, Switzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8049Zürich, Switzerland
| | | | - U. Benjamin Kaupp
- Life and Medical Sciences Institute, University of Bonn, 53115Bonn, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077Göttingen, Germany
| | - Volodymyr M. Korkhov
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zürich, 8049Zurich, Switzerland
| | - Jacopo Marino
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
| |
Collapse
|
9
|
Kameyama M, Minobe E, Shao D, Xu J, Gao Q, Hao L. Regulation of Cardiac Cav1.2 Channels by Calmodulin. Int J Mol Sci 2023; 24:ijms24076409. [PMID: 37047381 PMCID: PMC10094977 DOI: 10.3390/ijms24076409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
Cav1.2 Ca2+ channels, a type of voltage-gated L-type Ca2+ channel, are ubiquitously expressed, and the predominant Ca2+ channel type, in working cardiac myocytes. Cav1.2 channels are regulated by the direct interactions with calmodulin (CaM), a Ca2+-binding protein that causes Ca2+-dependent facilitation (CDF) and inactivation (CDI). Ca2+-free CaM (apoCaM) also contributes to the regulation of Cav1.2 channels. Furthermore, CaM indirectly affects channel activity by activating CaM-dependent enzymes, such as CaM-dependent protein kinase II and calcineurin (a CaM-dependent protein phosphatase). In this article, we review the recent progress in identifying the role of apoCaM in the channel ‘rundown’ phenomena and related repriming of channels, and CDF, as well as the role of Ca2+/CaM in CDI. In addition, the role of CaM in channel clustering is reviewed.
Collapse
Affiliation(s)
- Masaki Kameyama
- Department of Physiology, Graduate School of Medical & Dental Sciences, Kagoshima University, Sakura-ga-oka, Kagoshima 890-8544, Japan
- Correspondence:
| | - Etsuko Minobe
- Department of Physiology, Graduate School of Medical & Dental Sciences, Kagoshima University, Sakura-ga-oka, Kagoshima 890-8544, Japan
| | - Dongxue Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110012, China (L.H.)
| | - Jianjun Xu
- Department of Physiology, Graduate School of Medical & Dental Sciences, Kagoshima University, Sakura-ga-oka, Kagoshima 890-8544, Japan
| | - Qinghua Gao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110012, China (L.H.)
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110012, China (L.H.)
| |
Collapse
|
10
|
Zuidscherwoude M, van Goor MK, Roig SR, Thijssen N, van Erp M, Fransen J, van der Wijst J, Hoenderop JG. Functional basis for calmodulation of the TRPV5 calcium channel. J Physiol 2023; 601:859-878. [PMID: 36566502 DOI: 10.1113/jp282952] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/06/2022] [Indexed: 12/26/2022] Open
Abstract
Within the transient receptor potential (TRP) superfamily of ion channels, TRPV5 is a highly Ca2+ -selective channel important for active reabsorption of Ca2+ in the kidney. Its channel activity is controlled by a negative feedback mechanism involving calmodulin (CaM) binding. Combining advanced microscopy techniques and biochemical assays, this study characterized the dynamic lobe-specific CaM regulation. We demonstrate for the first time that functional (full-length) TRPV5 interacts with CaM in the absence of Ca2+ , and this interaction is intensified at increasing Ca2+ concentrations sensed by the CaM C-lobe that achieves channel pore blocking. Channel inactivation occurs without requiring CaM N-lobe calcification. Moreover, we show a Ca2+ -dependent binding stoichiometry at the single channel level. In conclusion, our study proposes a new model for CaM-dependent regulation - calmodulation - of this uniquely Ca2+ -selective TRP channel TRPV5 that involves apoCaM interaction and lobe-specific actions, which may be of significant physiological relevance given its role as gatekeeper of Ca2+ transport in the kidney. KEY POINTS: The renal Ca2+ channel TRPV5 is an important player in maintenance of the body's Ca2+ homeostasis. Activity of TRPV5 is controlled by a negative feedback loop that involves calmodulin (CaM), a protein with two Ca2+ -binding lobes. We investigated the dynamics of the interaction between TRPV5 and CaM with advanced fluorescence microscopy techniques. Our data support a new model for CaM-dependent regulation of TRPV5 channel activity with CaM lobe-specific actions and demonstrates Ca2+ -dependent binding stoichiometries. This study improves our understanding of the mechanism underlying fast channel inactivation, which is physiologically relevant given the gatekeeper function of TRPV5 in Ca2+ reabsorption in the kidney.
Collapse
Affiliation(s)
- Malou Zuidscherwoude
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark K van Goor
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sara R Roig
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Imaging Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Niky Thijssen
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Merijn van Erp
- Radboudumc Technology Centre Microscopy, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jack Fransen
- Radboudumc Technology Centre Microscopy, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Ramis R, Ballesteros ÓR, Muguruza-Montero A, M-Alicante S, Núñez E, Villarroel Á, Leonardo A, Bergara A. Molecular dynamics simulations of the calmodulin-induced α-helix in the SK2 calcium-gated potassium ion channel. J Biol Chem 2022; 299:102850. [PMID: 36587765 PMCID: PMC9874072 DOI: 10.1016/j.jbc.2022.102850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
The family of small-conductance Ca2+-activated potassium ion channels (SK channels) is composed of four members (SK1, SK2, SK3, and SK4) involved in neuron-firing regulation. The gating of these channels depends on the intracellular Ca2+ concentration, and their sensitivity to this ion is provided by calmodulin (CaM). This protein binds to a specific region in SK channels known as the calmodulin-binding domain (CaMBD), an event which is essential for their gating. While CaMBDs are typically disordered in the absence of CaM, the SK2 channel subtype displays a small prefolded α-helical region in its CaMBD even if CaM is not present. This small helix is known to turn into a full α-helix upon CaM binding, although the molecular-level details for this conversion are not fully understood yet. In this work, we offer new insights on this physiologically relevant process by means of enhanced sampling, atomistic Hamiltonian replica exchange molecular dynamics simulations, providing a more detailed understanding of CaM binding to this target. Our results show that CaM is necessary for inducing a full α-helix along the SK2 CaMBD through hydrophobic interactions with V426 and L427. However, it is also necessary that W431 does not compete for these interactions; the role of the small prefolded α-helix in the SK2 CaMBD would be to stabilize W431 so that this is the case. In conclusion, our findings provide further insight into a key interaction between CaM and SK channels that is important for channel sensitivity to Ca2+.
Collapse
Affiliation(s)
- Rafael Ramis
- Donostia International Physics Center, Donostia, Spain; Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain.
| | - Óscar R. Ballesteros
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Centro de Física de Materiales CFM, CSIC-UPV/EHU, Donostia, Spain
| | | | - Sara M-Alicante
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Instituto Biofisika, CSIC-UPV/EHU, Leioa, Spain
| | - Eider Núñez
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Instituto Biofisika, CSIC-UPV/EHU, Leioa, Spain
| | | | - Aritz Leonardo
- Donostia International Physics Center, Donostia, Spain,Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain
| | - Aitor Bergara
- Donostia International Physics Center, Donostia, Spain,Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Centro de Física de Materiales CFM, CSIC-UPV/EHU, Donostia, Spain
| |
Collapse
|
12
|
Prodan N, Ershad F, Reyes-Alcaraz A, Li L, Mistretta B, Gonzalez L, Rao Z, Yu C, Gunaratne PH, Li N, Schwartz RJ, McConnell BK. Direct reprogramming of cardiomyocytes into cardiac Purkinje-like cells. iScience 2022; 25:105402. [PMID: 36388958 PMCID: PMC9646947 DOI: 10.1016/j.isci.2022.105402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/30/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Currently, there are no treatments that ameliorate cardiac cell death, the underlying basis of cardiovascular disease. An unexplored cell type in cardiac regeneration is cardiac Purkinje cells; specialized cells from the cardiac conduction system (CCS) responsible for propagating electrical signals. Purkinje cells have tremendous potential as a regenerative treatment because they may intrinsically integrate with the CCS of a recipient myocardium, resulting in more efficient electrical conduction in diseased hearts. This study is the first to demonstrate an effective protocol for the direct reprogramming of human cardiomyocytes into cardiac Purkinje-like cells using small molecules. The cells generated were genetically and functionally similar to native cardiac Purkinje cells, where expression of key cardiac Purkinje genes such as CNTN2, ETV1, PCP4, IRX3, SCN5a, HCN2 and the conduction of electrical signals with increased velocity was observed. This study may help to advance the quest to finding an optimized cell therapy for heart regeneration.
Collapse
Affiliation(s)
- Nicole Prodan
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
| | - Faheem Ershad
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Arfaxad Reyes-Alcaraz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
| | - Luge Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brandon Mistretta
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, UH-Sequencing & Gene Editing Core, University of Houston, Houston, TX 77204, USA
| | - Lei Gonzalez
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Zhoulyu Rao
- Department of Mechanical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Cunjiang Yu
- Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
- Department of Mechanical Engineering, Cullen College of Engineering, University of Houston, Houston, TX 77204, USA
| | - Preethi H. Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, UH-Sequencing & Gene Editing Core, University of Houston, Houston, TX 77204, USA
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert J. Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Bradley K. McConnell
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd, Health-2 (H2) Building, Room 5024, Houston, TX 77204-5037, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
13
|
Munk M, Villalobo E, Villalobo A, Berchtold MW. Differential expression of the three independent CaM genes coding for an identical protein: Potential relevance of distinct mRNA stability by different codon usage. Cell Calcium 2022; 107:102656. [DOI: 10.1016/j.ceca.2022.102656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/01/2022] [Accepted: 09/25/2022] [Indexed: 11/24/2022]
|
14
|
Zhao J, Segura E, Marsolais M, Parent L. A CACNA1C variant associated with cardiac arrhythmias provides mechanistic insights in the calmodulation of L-type Ca 2+ channels. J Biol Chem 2022; 298:102632. [PMID: 36273583 PMCID: PMC9691931 DOI: 10.1016/j.jbc.2022.102632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/12/2022] [Accepted: 10/15/2022] [Indexed: 11/07/2022] Open
Abstract
We recently reported the identification of a de novo single nucleotide variant in exon 9 of CACNA1C associated with prolonged repolarization interval. Recombinant expression of the glycine to arginine variant at position 419 produced a gain in the function of the L-type CaV1.2 channel with increased peak current density and activation gating but without significant decrease in the inactivation kinetics. We herein reveal that these properties are replicated by overexpressing calmodulin (CaM) with CaV1.2 WT and are reversed by exposure to the CaM antagonist W-13. Phosphomimetic (T79D or S81D), but not phosphoresistant (T79A or S81A), CaM surrogates reproduced the impact of CaM WT on the function of CaV1.2 WT. The increased channel activity of CaV1.2 WT following overexpression of CaM was found to arise in part from enhanced cell surface expression. In contrast, the properties of the variant remained unaffected by any of these treatments. CaV1.2 substituted with the α-helix breaking proline residue were more reluctant to open than CaV1.2 WT but were upregulated by phosphomimetic CaM surrogates. Our results indicate that (1) CaM and its phosphomimetic analogs promote a gain in the function of CaV1.2 and (2) the structural properties of the first intracellular linker of CaV1.2 contribute to its CaM-induced modulation. We conclude that the CACNA1C clinical variant mimics the increased activity associated with the upregulation of CaV1.2 by Ca2+-CaM, thus maintaining a majority of channels in a constitutively active mode that could ultimately promote ventricular arrhythmias.
Collapse
Affiliation(s)
- Juan Zhao
- Centre de recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Emilie Segura
- Centre de recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec, Canada,Département de Pharmacologie et Physiologie, Faculté de Médecine, Montréal, Québec, Canada
| | - Mireille Marsolais
- Centre de recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec, Canada,Département de Pharmacologie et Physiologie, Faculté de Médecine, Montréal, Québec, Canada
| | - Lucie Parent
- Centre de recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec, Canada,Département de Pharmacologie et Physiologie, Faculté de Médecine, Montréal, Québec, Canada,For correspondence: Lucie Parent
| |
Collapse
|
15
|
Calmodulin in Paramecium: Focus on Genomic Data. Microorganisms 2022; 10:microorganisms10101915. [PMID: 36296191 PMCID: PMC9608856 DOI: 10.3390/microorganisms10101915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022] Open
Abstract
Calcium (Ca2+) is a universal second messenger that plays a key role in cellular signaling. However, Ca2+ signals are transduced with the help of Ca2+-binding proteins, which serve as sensors, transducers, and elicitors. Among the collection of these Ca2+-binding proteins, calmodulin (CaM) emerged as the prototypical model in eukaryotic cells. This is a small protein that binds four Ca2+ ions and whose functions are multiple, controlling many essential aspects of cell physiology. CaM is universally distributed in eukaryotes, from multicellular organisms, such as human and land plants, to unicellular microorganisms, such as yeasts and ciliates. Here, we review most of the information gathered on CaM in Paramecium, a group of ciliates. We condense the information here by mentioning that mature Paramecium CaM is a 148 amino acid-long protein codified by a single gene, as in other eukaryotic microorganisms. In these ciliates, the protein is notoriously localized and regulates cilia function and can stimulate the activity of some enzymes. When Paramecium CaM is mutated, cells show flawed locomotion and/or exocytosis. We further widen this and additional information in the text, focusing on genomic data.
Collapse
|
16
|
Martinac B, Kung C. The force-from-lipid principle and its origin, a ‘ what is true for E. coli is true for the elephant’ refrain. J Neurogenet 2022; 36:44-54. [DOI: 10.1080/01677063.2022.2097674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Ching Kung
- Laboratory of Molecular Biology and the Department of Genetics, University of Wisconsin–Madison, Madison, WI, USA
| |
Collapse
|
17
|
Plattner H. Ciliate Research. From Myth to Trendsetting Science. J Eukaryot Microbiol 2022; 69:e12926. [PMID: 35608570 DOI: 10.1111/jeu.12926] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 11/28/2022]
Abstract
This special issue of the Journal of Eukaryotic Microbiology (JEM) summarizes achievements obtained by generations of researchers with ciliates in widely different disciplines. In fact, ciliates range among the first cells seen under the microscope centuries ago. Their beauty made them an object of scientia amabilis and their manifold reactions made them attractive for college experiments and finally challenged causal analyses at the cellular level. Some of this work was honored by a Nobel Prize. Some observations yielded a baseline for additional novel discoveries, occasionally facilitated by specific properties of some ciliates. This also offers some advantage in the exploration of closely related parasites (malaria). Articles contributed here by colleagues from all over the world encompass a broad spectrum of ciliate life, from genetics to evolution, from molecular cell biology to ecology, from intercellular signaling to epigenetics etc. This introductory chapter, largely based on my personal perception, aims at integrating work presented in this special issue of JEM into a broader historical context up to current research.
Collapse
|
18
|
Boamah GA, Huang Z, Shen Y, Lu Y, Wang Z, Su Y, Xu C, Luo X, Ke C, You W. Transcriptome analysis reveals fluid shear stress (FSS) and atherosclerosis pathway as a candidate molecular mechanism of short-term low salinity stress tolerance in abalone. BMC Genomics 2022; 23:392. [PMID: 35606721 PMCID: PMC9128277 DOI: 10.1186/s12864-022-08611-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
Background Transcriptome sequencing is an effective tool to reveal the essential genes and pathways underlying countless biotic and abiotic stress adaptation mechanisms. Although severely challenged by diverse environmental conditions, the Pacific abalone Haliotis discus hannai remains a high-value aquaculture mollusk and a Chinese predominantly cultured abalone species. Salinity is one of such environmental factors whose fluctuation could significantly affect the abalone’s cellular and molecular immune responses and result in high mortality and reduced growth rate during prolonged exposure. Meanwhile, hybrids have shown superiority in tolerating diverse environmental stresses over their purebred counterparts and have gained admiration in the Chinese abalone aquaculture industry. The objective of this study was to investigate the molecular and cellular mechanisms of low salinity adaptation in abalone. Therefore, this study used transcriptome analysis of the gill tissues and flow cytometric analysis of hemolymph of H. discus hannai (DD) and interspecific hybrid H. discus hannai ♀ x H. fulgens ♂ (DF) during low salinity exposure. Also, the survival and growth rate of the species under various salinities were assessed. Results The transcriptome data revealed that the differentially expressed genes (DEGs) were significantly enriched on the fluid shear stress and atherosclerosis (FSS) pathway. Meanwhile, the expression profiles of some essential genes involved in this pathway suggest that abalone significantly up-regulated calmodulin-4 (CaM-4) and heat-shock protein90 (HSP90), and significantly down-regulated tumor necrosis factor (TNF), bone morphogenetic protein-4 (BMP-4), and nuclear factor kappa B (NF-kB). Also, the hybrid DF showed significantly higher and sustained expression of CaM and HSP90, significantly higher phagocytosis, significantly lower hemocyte mortality, and significantly higher survival at low salinity, suggesting a more active molecular and hemocyte-mediated immune response and a more efficient capacity to tolerate low salinity than DD. Conclusions Our study argues that the abalone CaM gene might be necessary to maintain ion equilibrium while HSP90 can offset the adverse changes caused by low salinity, thereby preventing damage to gill epithelial cells (ECs). The data reveal a potential molecular mechanism by which abalone responds to low salinity and confirms that hybridization could be a method for breeding more stress-resilient aquatic species. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08611-8.
Collapse
Affiliation(s)
- Grace Afumwaa Boamah
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China.,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China.,College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Zekun Huang
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China.,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China.,College of the Environment and Ecology, Xiamen University, 361102, Xiamen, PR China
| | - Yawei Shen
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China.,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China.,College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Yisha Lu
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China.,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China.,College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Zhixuan Wang
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China.,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China.,College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Ying Su
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China.,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China.,College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Changan Xu
- Third Institute of Oceanography, MNR, Xiamen, 361005, China
| | - Xuan Luo
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China.,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China.,College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Caihuan Ke
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China. .,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China. .,College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China.
| | - Weiwei You
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, People's Republic of China. .,Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, Xiamen University, Xiamen, 361102, People's Republic of China. .,College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China.
| |
Collapse
|
19
|
Spolaor S, Rovetta M, Nobile MS, Cazzaniga P, Tisi R, Besozzi D. Modeling Calcium Signaling in S. cerevisiae Highlights the Role and Regulation of the Calmodulin-Calcineurin Pathway in Response to Hypotonic Shock. Front Mol Biosci 2022; 9:856030. [PMID: 35664674 PMCID: PMC9158465 DOI: 10.3389/fmolb.2022.856030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/04/2022] [Indexed: 01/17/2023] Open
Abstract
Calcium homeostasis and signaling processes in Saccharomyces cerevisiae, as well as in any eukaryotic organism, depend on various transporters and channels located on both the plasma and intracellular membranes. The activity of these proteins is regulated by a number of feedback mechanisms that act through the calmodulin-calcineurin pathway. When exposed to hypotonic shock (HTS), yeast cells respond with an increased cytosolic calcium transient, which seems to be conditioned by the opening of stretch-activated channels. To better understand the role of each channel and transporter involved in the generation and recovery of the calcium transient—and of their feedback regulations—we defined and analyzed a mathematical model of the calcium signaling response to HTS in yeast cells. The model was validated by comparing the simulation outcomes with calcium concentration variations before and during the HTS response, which were observed experimentally in both wild-type and mutant strains. Our results show that calcium normally enters the cell through the High Affinity Calcium influx System and mechanosensitive channels. The increase of the plasma membrane tension, caused by HTS, boosts the opening probability of mechanosensitive channels. This event causes a sudden calcium pulse that is rapidly dissipated by the activity of the vacuolar transporter Pmc1. According to model simulations, the role of another vacuolar transporter, Vcx1, is instead marginal, unless calcineurin is inhibited or removed. Our results also suggest that the mechanosensitive channels are subject to a calcium-dependent feedback inhibition, possibly involving calmodulin. Noteworthy, the model predictions are in accordance with literature results concerning some aspects of calcium homeostasis and signaling that were not specifically addressed within the model itself, suggesting that it actually depicts all the main cellular components and interactions that constitute the HTS calcium pathway, and thus can correctly reproduce the shaping of the calcium signature by calmodulin- and calcineurin-dependent complex regulations. The model predictions also allowed to provide an interpretation of different regulatory schemes involved in calcium handling in both wild-type and mutants yeast strains. The model could be easily extended to represent different calcium signals in other eukaryotic cells.
Collapse
Affiliation(s)
- Simone Spolaor
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| | - Mattia Rovetta
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| | - Marco S. Nobile
- Department of Environmental Sciences, Informatics and Statistics, Ca’ Foscari University of Venice, Venice, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre—B4, Milan, Italy
- SYSBIO/ISBE.IT Centre of Systems Biology, Milan, Italy
| | - Paolo Cazzaniga
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre—B4, Milan, Italy
- SYSBIO/ISBE.IT Centre of Systems Biology, Milan, Italy
- Department of Human and Social Sciences, University of Bergamo, Bergamo, Italy
| | - Renata Tisi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
- *Correspondence: Renata Tisi, ; Daniela Besozzi,
| | - Daniela Besozzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre—B4, Milan, Italy
- SYSBIO/ISBE.IT Centre of Systems Biology, Milan, Italy
- *Correspondence: Renata Tisi, ; Daniela Besozzi,
| |
Collapse
|
20
|
Kato K, Isbell HM, Fressart V, Denjoy I, Debbiche A, Itoh H, Poinsot J, George AL, Coulombe A, Shea MA, Guicheney P. Novel CALM3 Variant Causing Calmodulinopathy With Variable Expressivity in a 4-Generation Family. Circ Arrhythm Electrophysiol 2022; 15:e010572. [PMID: 35225649 DOI: 10.1161/circep.121.010572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND CaM (calmodulin), encoded by 3 separate genes (CALM1, CALM2, and CALM3), is a multifunctional Ca2+-binding protein involved in many signal transduction events including ion channel regulation. CaM variants may present with early-onset long QT syndrome (LQTS), catecholaminergic polymorphic ventricular tachycardia, or sudden cardiac death. Most reported variants occurred de novo. We identified a novel CALM3 variant, p.Asn138Lys (N138K), in a 4-generation family segregating with LQTS. The aim of this study was to elucidate its pathogenicity and to compare it with that of p.D130G-CaM-a variant associated with a severe LQTS phenotype. METHODS We performed whole exome sequencing for a large, 4-generation family affected by LQTS. To assess the effect of the detected CALM3 variant, the intrinsic Ca2+-binding affinity was measured by stoichiometric Ca2+ titrations and equilibrium titrations. L-type Ca2+ and slow delayed rectifier potassium currents (ICaL and IKs) were recorded by whole-cell patch-clamp. Cav1.2 and Kv7.1 membrane expression were determined by optical fluorescence assays. RESULTS We identified 14 p.N138K-CaM carriers in a family where 2 sudden deaths occurred in children. Several members were only mildly affected compared with CaM-LQTS patients to date described in literature. The intrinsic Ca2+-binding affinity of the CaM C-terminal domain was 10-fold lower for p.N138K-CaM compared with wild-type-CaM. ICaL inactivation was slowed in cells expressing p.N138K-CaM but less than in p.D130G-CaM cells. Unexpectedly, a larger IKs current density was observed in cells expressing p.N138K-CaM, but not for p.D130G-CaM, compared with wild-type-CaM. CONCLUSIONS The p.N138K CALM3 variant impairs Ca2+-binding affinity of CaM and ICaL inactivation but potentiates IKs. The variably expressed phenotype of this variant compared with previously published de novo LQTS-CaM variants is likely explained by a milder impairment of ICaL inactivation combined with IKs augmentation.
Collapse
Affiliation(s)
- Koichi Kato
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France (K.K., V.F., I.D., A.D., A.C., P.G.).,Department of Cardiovascular Medicine, Shiga University of Medical Science, Otsu, Japan (K.K.)
| | - Holly M Isbell
- Department of Biochemistry, Carver College of Medicine, University of Iowa (H.M.I., M.A.S.)
| | - Véronique Fressart
- AP-HP, Pitié-Salpêtrière Hospital, Functional Unit of Cardiogenetics and Myogenetics, Paris, France (V.F.)
| | - Isabelle Denjoy
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France (K.K., V.F., I.D., A.D., A.C., P.G.).,Cardiology Department, Referring Center for Heritable or Rare Cardiac Diseases, AP-HP, Bichat Hospital, HUPNVS, Referring Center for Rare Cardiac Diseases, Sorbonne University, Paris, France (I.D.)
| | - Amal Debbiche
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France (K.K., V.F., I.D., A.D., A.C., P.G.)
| | - Hideki Itoh
- Division of Patient Safety, Hiroshima University Hospital, Japan (H.I.)
| | - Jacques Poinsot
- Unité de cardio-pediatrie, service de medecine pediatrique, Centre Hospitalier Universitaire de Tours, Tours, France (J.P.)
| | - Alfred L George
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (A.L.G.)
| | - Alain Coulombe
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France (K.K., V.F., I.D., A.D., A.C., P.G.)
| | - Madeline A Shea
- Department of Biochemistry, Carver College of Medicine, University of Iowa (H.M.I., M.A.S.)
| | - Pascale Guicheney
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France (K.K., V.F., I.D., A.D., A.C., P.G.)
| |
Collapse
|
21
|
Bousova K, Zouharova M, Herman P, Vymetal J, Vetyskova V, Jiraskova K, Vondrasek J. TRPM5 Channel Binds Calcium-Binding Proteins Calmodulin and S100A1. Biochemistry 2022; 61:413-423. [PMID: 35225608 DOI: 10.1021/acs.biochem.1c00647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Melastatin transient receptor potential (TRPM) channels belong to one of the most significant subgroups of the transient receptor potential (TRP) channel family. Here, we studied the TRPM5 member, the receptor exposed to calcium-mediated activation, resulting in taste transduction. It is known that most TRP channels are highly modulated through interactions with extracellular and intracellular agents. The binding sites for these ligands are usually located at the intracellular N- and C-termini of the TRP channels, and they can demonstrate the character of an intrinsically disordered protein (IDP), which allows such a region to bind various types of molecules. We explored the N-termini of TRPM5 and found the intracellular regions for calcium-binding proteins (CBPs) the calmodulin (CaM) and calcium-binding protein S1 (S100A1) by in vitro binding assays. Furthermore, molecular docking and molecular dynamics simulations (MDs) of the discovered complexes confirmed their known common binding interface patterns and the uniqueness of the basic residues present in the TRPM binding regions for CaM/S100A1.
Collapse
Affiliation(s)
- Kristyna Bousova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, 16000 Prague, Czech Republic
| | - Monika Zouharova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, 16000 Prague, Czech Republic.,Second Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic
| | - Petr Herman
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 5, 12116 Prague, Czech Republic
| | - Jiri Vymetal
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, 16000 Prague, Czech Republic
| | - Veronika Vetyskova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, 16000 Prague, Czech Republic.,Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, Czech Republic
| | - Katerina Jiraskova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, 16000 Prague, Czech Republic
| | - Jiri Vondrasek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, 16000 Prague, Czech Republic
| |
Collapse
|
22
|
Xu Z, Zhang N, Fu H, Wang F, Wen M, Chang H, Wu J, Abdelaala WB, Luo Q, Li Y, Li C, Wang Q, Wang ZY. Salt Stress Modulates the Landscape of Transcriptome and Alternative Splicing in Date Palm ( Phoenix dactylifera L.). FRONTIERS IN PLANT SCIENCE 2022; 12:807739. [PMID: 35126432 PMCID: PMC8810534 DOI: 10.3389/fpls.2021.807739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/20/2021] [Indexed: 05/14/2023]
Abstract
Date palm regards as a valuable genomic resource for exploring the tolerance genes due to its ability to survive under the sever condition. Although a large number of differentiated genes were identified in date palm responding to salt stress, the genome-wide study of alternative splicing (AS) landscape under salt stress conditions remains unknown. In the current study, we identified the stress-related genes through transcriptomic analysis to characterize their function under salt. A total of 17,169 genes were differentially expressed under salt stress conditions. Gene expression analysis confirmed that the salt overly sensitive (SOS) pathway genes, such as PdSOS2;1, PdSOS2;2, PdSOS4, PdSOS5, and PdCIPK11 were involved in the regulation of salt response in date palm, which is consistent with the physiological analysis that high salinity affected the Na+/K+ homeostasis and amino acid profile of date palm resulted in the inhibition of plant growth. Interestingly, the pathway of "spliceosome" was enriched in the category of upregulation, indicating their potential role of AS in date palm response to salt stress. Expectedly, many differentially alternative splicing (DAS) events were found under salt stress conditions, and some splicing factors, such as PdRS40, PdRSZ21, PdSR45a, and PdU2Af genes were abnormally spliced under salt, suggesting that AS-related proteins might participated in regulating the salt stress pathway. Moreover, the number of differentially DAS-specific genes was gradually decreased, while the number of differentially expressed gene (DEG)-specific genes was increased with prolonged salt stress treatment, suggesting that AS and gene expression could be distinctively regulated in response to salt stress. Therefore, our study highlighted the pivotal role of AS in the regulation of salt stress and provided novel insights for enhancing the resistance to salt in date palm.
Collapse
Affiliation(s)
- Zhongliang Xu
- Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan, China
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan, China
| | - Ning Zhang
- Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan, China
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan, China
| | - Haiquan Fu
- Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan, China
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan, China
| | - Fuyou Wang
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan, China
| | - Mingfu Wen
- Institute of Nanfan & Seed Industry, Guangdong Academy of Sciences, Zhanjiang, China
- Zhanjiang Sugarcane Research Center, Guangzhou Sugarcane Industry Research Institute, Guangzhou, China
| | - Hailong Chang
- Institute of Nanfan & Seed Industry, Guangdong Academy of Sciences, Zhanjiang, China
| | - Jiantao Wu
- Institute of Nanfan & Seed Industry, Guangdong Academy of Sciences, Zhanjiang, China
| | - Walid Badawy Abdelaala
- Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Hainan, China
- Central Laboratory for Date Palm Research and Development of Agriculture Research Center, Giza, Egypt
| | - Qingwen Luo
- Institute of Nanfan & Seed Industry, Guangdong Academy of Sciences, Zhanjiang, China
- Zhanjiang Sugarcane Research Center, Guangzhou Sugarcane Industry Research Institute, Guangzhou, China
| | - Yang Li
- Institute of Nanfan & Seed Industry, Guangdong Academy of Sciences, Zhanjiang, China
- Zhanjiang Sugarcane Research Center, Guangzhou Sugarcane Industry Research Institute, Guangzhou, China
| | - Cong Li
- Institute of Nanfan & Seed Industry, Guangdong Academy of Sciences, Zhanjiang, China
- Zhanjiang Sugarcane Research Center, Guangzhou Sugarcane Industry Research Institute, Guangzhou, China
| | - Qinnan Wang
- Institute of Nanfan & Seed Industry, Guangdong Academy of Sciences, Zhanjiang, China
| | - Zhen-Yu Wang
- Institute of Nanfan & Seed Industry, Guangdong Academy of Sciences, Zhanjiang, China
- Zhanjiang Sugarcane Research Center, Guangzhou Sugarcane Industry Research Institute, Guangzhou, China
| |
Collapse
|
23
|
Peracchia C, Leverone Peracchia LM. Calmodulin-Connexin Partnership in Gap Junction Channel Regulation-Calmodulin-Cork Gating Model. Int J Mol Sci 2021; 22:ijms222313055. [PMID: 34884859 PMCID: PMC8658047 DOI: 10.3390/ijms222313055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 01/19/2023] Open
Abstract
In the past four decades numerous findings have indicated that gap junction channel gating is mediated by intracellular calcium concentrations ([Ca2+i]) in the high nanomolar range via calmodulin (CaM). We have proposed a CaM-based gating model based on evidence for a direct CaM role in gating. This model is based on the following: CaM inhibitors and the inhibition of CaM expression to prevent chemical gating. A CaM mutant with higher Ca2+ sensitivity greatly increases gating sensitivity. CaM co-localizes with connexins. Connexins have high-affinity CaM-binding sites. Connexin mutants paired to wild type connexins have a higher gating sensitivity, which is eliminated by the inhibition of CaM expression. Repeated trans-junctional voltage (Vj) pulses progressively close channels by the chemical/slow gate (CaM’s N-lobe). At the single channel level, the gate closes and opens slowly with on-off fluctuations. Internally perfused crayfish axons lose gating competency but recover it by the addition of Ca-CaM to the internal perfusion solution. X-ray diffraction data demonstrate that isolated gap junctions are gated at the cytoplasmic end by a particle of the size of a CaM lobe. We have proposed two types of CaM-driven gating: “Ca-CaM-Cork” and “CaM-Cork”. In the first, the gating involves Ca2+-induced CaM activation. In the second, the gating occurs without a [Ca2+]i rise.
Collapse
|
24
|
Guo L, Li C, Coupland G, Liang P, Chu D. Up-regulation of calmodulin involved in the stress response to cyantraniliprole in the whitefly, Bemisia tabaci (Hemiptera: Aleyrodidae). INSECT SCIENCE 2021; 28:1745-1755. [PMID: 33200870 DOI: 10.1111/1744-7917.12887] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 06/11/2023]
Abstract
Cyantraniliprole is the first diamide insecticide to have cross-spectrum activity against a broad range of insect orders. The insecticide, like other diamides, selectively acts on ryanodine receptor, destroys Ca2+ homeostasis, and ultimately causes insect death. Although expression regulations of genes associated with calcium signaling pathways are known to be involved in the response to diamides, little is known regarding the function of calmodulin (CaM), a typical Ca2+ sensor central in regulating Ca2+ homeostasis, in the stress response of insects to the insecticide. In this study, we cloned and identified the full-length complementary DNA of CaM in the whitefly, Bemisia tabaci (Gennadius), named BtCaM. Quantitative real-time reverse transcription polymerase chain reaction-based analyses showed that the messenger RNA level of BtCaM was rapidly induced from 1.51- to 2.43-fold by cyantraniliprole during 24 h. Knockdown of BtCaM by RNA interference increased the toxicity of cyantraniliprole in whiteflies by 42.85%. In contrast, BtCaM expression in Sf9 cells significantly increased the cells' tolerance to cyantraniliprole as much as 2.91-fold. In addition, the expression of BtCaM in Sf9 cells suppressed the rapid increase of intracellular Ca2+ after exposure to cyantraniliprole, and the maximum amplitude in the Sf9-BtCaM cells was only 34.9% of that in control cells (Sf9-PIZ/V5). These results demonstrate that overexpression of BtCaM is involved in the stress response of B. tabaci to cyantraniliprole through regulation of Ca2+ concentration. As CaM is one of the most evolutionarily conserved Ca2+ sensors in insects, outcomes of this study may provide the first details of a universal insect response to diamide insecticides.
Collapse
Affiliation(s)
- Lei Guo
- Key Lab of Integrated Crop Pest Management of Shandong Province, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, Shangdong, 266109, China
| | - Changyou Li
- Key Lab of Integrated Crop Pest Management of Shandong Province, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, Shangdong, 266109, China
| | - Grey Coupland
- Harry Butler Institute, Murdoch University, Murdoch, WA, 6150, Australia
| | - Pei Liang
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Dong Chu
- Key Lab of Integrated Crop Pest Management of Shandong Province, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, Shangdong, 266109, China
| |
Collapse
|
25
|
Vuong-Brender TT, Flynn S, Vallis Y, Sönmez SE, de Bono M. Neuronal calmodulin levels are controlled by CAMTA transcription factors. eLife 2021; 10:e68238. [PMID: 34499028 PMCID: PMC8428840 DOI: 10.7554/elife.68238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/28/2021] [Indexed: 01/18/2023] Open
Abstract
The ubiquitous Ca2+ sensor calmodulin (CaM) binds and regulates many proteins, including ion channels, CaM kinases, and calcineurin, according to Ca2+-CaM levels. What regulates neuronal CaM levels, is, however, unclear. CaM-binding transcription activators (CAMTAs) are ancient proteins expressed broadly in nervous systems and whose loss confers pleiotropic behavioral defects in flies, mice, and humans. Using Caenorhabditis elegans and Drosophila, we show that CAMTAs control neuronal CaM levels. The behavioral and neuronal Ca2+ signaling defects in mutants lacking camt-1, the sole C. elegans CAMTA, can be rescued by supplementing neuronal CaM. CAMT-1 binds multiple sites in the CaM promoter and deleting these sites phenocopies camt-1. Our data suggest CAMTAs mediate a conserved and general mechanism that controls neuronal CaM levels, thereby regulating Ca2+ signaling, physiology, and behavior.
Collapse
Affiliation(s)
- Thanh Thi Vuong-Brender
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Sean Flynn
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Yvonne Vallis
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Saliha E Sönmez
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Mario de Bono
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
26
|
Ftuwi H, Parri R, Mohammed AR. Novel, Fully Characterised Bovine Taste Bud Cells of Fungiform Papillae. Cells 2021; 10:2285. [PMID: 34571933 PMCID: PMC8469975 DOI: 10.3390/cells10092285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/23/2021] [Accepted: 08/28/2021] [Indexed: 01/14/2023] Open
Abstract
Current understanding of functional characteristics and biochemical pathways in taste bud cells have been hindered due the lack of long-term cultured cells. To address this, we developed a holistic approach to fully characterise long term cultured bovine taste bud cells (BTBCs). Initially, cultured BTBCs were characterised using RT-PCR gene expression profiling, immunocytochemistry, flowcytometry and calcium imaging, that confirmed the cells were mature TBCs that express taste receptor genes, taste specific protein markers and capable of responding to taste stimuli, i.e., denatonium (2 mM) and quinine (462.30 μM). Gene expression analysis of forty-two genes implicated in taste transduction pathway (map04742) using custom-made RT-qPCR array revealed high and low expressed genes in BTBCs. Preliminary datamining and bioinformatics demonstrated that the bovine α-gustducin, gustatory G-protein, have higher sequence similarity to the human orthologue compared to rodents. Therefore, results from this work will replace animal experimentation and provide surrogate cell-based throughput system to study human taste transduction.
Collapse
Affiliation(s)
| | | | - Afzal R. Mohammed
- Aston Pharmacy School, Aston University, Birmingham B4 7ET, UK; (H.F.); (R.P.)
| |
Collapse
|
27
|
Ledford HA, Park S, Muir D, Woltz RL, Ren L, Nguyen PT, Sirish P, Wang W, Sihn CR, George AL, Knollmann BC, Yamoah EN, Yarov-Yarovoy V, Zhang XD, Chiamvimonvat N. Different arrhythmia-associated calmodulin mutations have distinct effects on cardiac SK channel regulation. J Gen Physiol 2021; 152:211546. [PMID: 33211795 PMCID: PMC7681919 DOI: 10.1085/jgp.202012667] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/25/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Calmodulin (CaM) plays a critical role in intracellular signaling and regulation of Ca2+-dependent proteins and ion channels. Mutations in CaM cause life-threatening cardiac arrhythmias. Among the known CaM targets, small-conductance Ca2+-activated K+ (SK) channels are unique, since they are gated solely by beat-to-beat changes in intracellular Ca2+. However, the molecular mechanisms of how CaM mutations may affect the function of SK channels remain incompletely understood. To address the structural and functional effects of these mutations, we introduced prototypical human CaM mutations in human induced pluripotent stem cell–derived cardiomyocyte-like cells (hiPSC-CMs). Using structural modeling and molecular dynamics simulation, we demonstrate that human calmodulinopathy-associated CaM mutations disrupt cardiac SK channel function via distinct mechanisms. CaMD96V and CaMD130G mutants reduce SK currents through a dominant-negative fashion. By contrast, specific mutations replacing phenylalanine with leucine result in conformational changes that affect helix packing in the C-lobe, which disengage the interactions between apo-CaM and the CaM-binding domain of SK channels. Distinct mutant CaMs may result in a significant reduction in the activation of the SK channels, leading to a decrease in the key Ca2+-dependent repolarization currents these channels mediate. The findings in this study may be generalizable to other interactions of mutant CaMs with Ca2+-dependent proteins within cardiac myocytes.
Collapse
Affiliation(s)
- Hannah A Ledford
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Seojin Park
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Duncan Muir
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Ryan L Woltz
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Lu Ren
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Phuong T Nguyen
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Wenying Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Choong-Ryoul Sihn
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Björn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, School of Medicine, Vanderbilt University, Nashville, TN
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA
| | - Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA
| |
Collapse
|
28
|
Gap Junction Channelopathies and Calmodulinopathies. Do Disease-Causing Calmodulin Mutants Affect Direct Cell-Cell Communication? Int J Mol Sci 2021; 22:ijms22179169. [PMID: 34502077 PMCID: PMC8431743 DOI: 10.3390/ijms22179169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 11/24/2022] Open
Abstract
The cloning of connexins cDNA opened the way to the field of gap junction channelopathies. Thus far, at least 35 genetic diseases, resulting from mutations of 11 different connexin genes, are known to cause numerous structural and functional defects in the central and peripheral nervous system as well as in the heart, skin, eyes, teeth, ears, bone, hair, nails and lymphatic system. While all of these diseases are due to connexin mutations, minimal attention has been paid to the potential diseases of cell–cell communication caused by mutations of Cx-associated molecules. An important Cx accessory protein is calmodulin (CaM), which is the major regulator of gap junction channel gating and a molecule relevant to gap junction formation. Recently, diseases caused by CaM mutations (calmodulinopathies) have been identified, but thus far calmodulinopathy studies have not considered the potential effect of CaM mutations on gap junction function. The major goal of this review is to raise awareness on the likely role of CaM mutations in defects of gap junction mediated cell communication. Our studies have demonstrated that certain CaM mutants affect gap junction channel gating or expression, so it would not be surprising to learn that CaM mutations known to cause diseases also affect cell communication mediated by gap junction channels.
Collapse
|
29
|
Zhang HL, Zhao B, Han W, Sun YB, Yang P, Chen Y, Ni D, Zhang J, Yin DM. Acetylation of calmodulin regulates synaptic plasticity and fear learning. J Biol Chem 2021; 297:101034. [PMID: 34339735 PMCID: PMC8383114 DOI: 10.1016/j.jbc.2021.101034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/20/2021] [Accepted: 07/29/2021] [Indexed: 11/26/2022] Open
Abstract
Synaptic plasticity is critical for brain function, including learning and memory. It is regulated by gene transcription and protein synthesis as well as posttranslational modifications at synapses. Although protein acetylation has been shown to be involved in the regulation of synaptic plasticity, this was mainly for histone protein acetylation. To investigate whether acetylation of nonhistone proteins is important for synaptic plasticity, we analyzed mouse brain acetylome and found that calmodulin (CaM), a ubiquitous Ca2+ sensor, was acetylated on three lysine residues, which were conserved across species. NMDA receptor-dependent long-term potentiation (LTP) is considered the most compelling form of synaptic plasticity. During LTP induction, activation of NMDA receptor triggers Ca2+ influx, and the Ca2+ binds with CaM and activates calcium/calmodulin-dependent protein kinase IIα (CaMKIIα), which is essential for LTP induction. By using home-generated and site-specific antibodies against acetylated CaM, we show that CaM acetylation is upregulated by neural activities in an NMDA receptor-dependent manner. Moreover, mutation of acetyllysines in CaM1 proteins disrupts synaptic plasticity and fear learning in a mouse model. We further demonstrate that acetylation of CaM reduces the binding free energy and increases the binding affinity toward CaMKIIα, a protein kinase pivotal to synaptic plasticity and learning. Taken together, our results demonstrate importance of CaM acetylation in regulating synaptic plasticity and learning.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Bing Zhao
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China
| | - Wei Han
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China
| | - Yi-Bei Sun
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China
| | - Pin Yang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China
| | - Yongjun Chen
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Duan Ni
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pharmacy, Clinical and Fundamental Research Center, Renji Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jian Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pharmacy, Clinical and Fundamental Research Center, Renji Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Dong-Min Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, China.
| |
Collapse
|
30
|
CRISPR/Cas9-mediated tryptophan hydroxylase 1 knockout decreases calcium transportation in goat mammary epithelial cells. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.108015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
31
|
Val‐Blasco A, Gil‐Fernández M, Rueda A, Pereira L, Delgado C, Smani T, Ruiz Hurtado G, Fernández‐Velasco M. Ca 2+ mishandling in heart failure: Potential targets. Acta Physiol (Oxf) 2021; 232:e13691. [PMID: 34022101 DOI: 10.1111/apha.13691] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Department of Biochemistry Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV‐IPN) México City Mexico
| | - Laetitia Pereira
- INSERM UMR‐S 1180 Laboratory of Ca Signaling and Cardiovascular Physiopathology University Paris‐Saclay Châtenay‐Malabry France
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols Madrid Spain
- Department of Metabolism and Cell Signalling Biomedical Research Institute "Alberto Sols" CSIC‐UAM Madrid Spain
| | - Tarik Smani
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
- Department of Medical Physiology and Biophysics University of Seville Seville Spain
- Group of Cardiovascular Pathophysiology Institute of Biomedicine of Seville University Hospital of Virgen del Rocío, University of Seville, CSIC Seville Spain
| | - Gema Ruiz Hurtado
- Cardiorenal Translational Laboratory Institute of Research i+12 University Hospital 12 de Octubre Madrid Spain
- CIBER‐CV University Hospita1 12 de Octubre Madrid Spain
| | - Maria Fernández‐Velasco
- La Paz University Hospital Health Research Institute IdiPAZ Madrid Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
| |
Collapse
|
32
|
Guo Y, Yu ZY, Wu J, Gong H, Kesteven S, Iismaa SE, Chan AY, Holman S, Pinto S, Pironet A, Cox CD, Graham RM, Vennekens R, Feneley MP, Martinac B. The Ca 2+-activated cation channel TRPM4 is a positive regulator of pressure overload-induced cardiac hypertrophy. eLife 2021; 10:66582. [PMID: 34190686 PMCID: PMC8245133 DOI: 10.7554/elife.66582] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/20/2021] [Indexed: 01/19/2023] Open
Abstract
Pathological left ventricular hypertrophy (LVH) occurs in response to pressure overload and remains the single most important clinical predictor of cardiac mortality. The molecular pathways in the induction of pressure overload LVH are potential targets for therapeutic intervention. Current treatments aim to remove the pressure overload stimulus for LVH, but do not completely reverse adverse cardiac remodelling. Although numerous molecular signalling steps in the induction of LVH have been identified, the initial step by which mechanical stretch associated with cardiac pressure overload is converted into a chemical signal that initiates hypertrophic signalling remains unresolved. In this study, we show that selective deletion of transient receptor potential melastatin 4 (TRPM4) channels in mouse cardiomyocytes results in an approximately 50% reduction in the LVH induced by transverse aortic constriction. Our results suggest that TRPM4 channel is an important component of the mechanosensory signalling pathway that induces LVH in response to pressure overload and represents a potential novel therapeutic target for the prevention of pathological LVH.
Collapse
Affiliation(s)
- Yang Guo
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Ze-Yan Yu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jianxin Wu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Hutao Gong
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Scott Kesteven
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Siiri E Iismaa
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Andrea Y Chan
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Sara Holman
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Silvia Pinto
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Andy Pironet
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Robert M Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Michael P Feneley
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Department of Cardiology, St Vincent's Hospital, Sydney, Australia
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
33
|
Han Y, Tang Y, Sun S, Kim T, Ju K, Ri S, Du X, Zhou W, Shi W, Li S, Liu G. Modulatory function of calmodulin on phagocytosis and potential regulation mechanisms in the blood clam Tegillarca granosa. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 116:103910. [PMID: 33129883 DOI: 10.1016/j.dci.2020.103910] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 06/11/2023]
Abstract
Unlike vertebrate species, invertebrates lack antigen-antibody mediated immune response and mainly rely on haemocyte phagocytosis to fight against pathogen infection. Recently, studies conducted in model vertebrates demonstrated that the multifunctional protein calmodulin (CaM) plays an important role in regulating immune responses. However, the intrinsic relation between CaM and phagocytosis process remains poorly understood in invertebrate species such as bivalve mollusks. Therefore, in the present study, the immunomodulatory function of CaM on haemocyte phagocytosis was verified in the blood clam, Tegillarca granosa, using the CaM-specific inhibitor N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide hydrochloride (W-7). Results obtained show that CaM inhibition significantly suppressed the phagocytic activity of haemocytes. In addition, CaM inhibition constrained intracellular Ca2+ elevation, hampered actin cytoskeleton assembly, suppressed calcineurin (CaN) activity, and disrupted NF-κB activation in haemocytes upon LPS induction. Furthermore, expression of seven selected genes from the actin cytoskeleton regulation- and immune-related pathways were significantly downregulated whereas those of CaM and CaN from the Ca2+-signaling pathway were significantly upregulated by in vitro incubation of haemocytes with W-7. For the first time, the present study demonstrated that CaM play an important role in phagocytosis modulation in bivalve species. In addition, the intracellular Ca2+ and downstream Ca2+-signaling-, actin cytoskeleton regulation-, and immune-related pathways offer candidate routes through which CaM modulates phagocytosis.
Collapse
Affiliation(s)
- Yu Han
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yu Tang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Shuge Sun
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Tongchol Kim
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China; College of Life Science, Kim Hyong Jik University of Education, Pyongyang, 99903, Republic of Korea
| | - Kwangjin Ju
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China; College of Aquaculture, Wonsan Fisheries University, Wonsan, 999093, Republic of Korea
| | - Sanghyok Ri
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China; College of Life Science, Kim Hyong Jik University of Education, Pyongyang, 99903, Republic of Korea
| | - Xueying Du
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Weishang Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Wei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Shiguo Li
- Key Laboratory of Environmental Biotechnology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China.
| | - Guangxu Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
34
|
Jain K, Lavista-Llanos S, Grabe V, Hansson BS, Wicher D. Calmodulin regulates the olfactory performance in Drosophila melanogaster. Sci Rep 2021; 11:3747. [PMID: 33580172 PMCID: PMC7881240 DOI: 10.1038/s41598-021-83296-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/01/2021] [Indexed: 12/01/2022] Open
Abstract
Insect odorant receptors (ORs) detect volatile chemical cues with high sensitivity. These ORs operate as ligand-gated ion channels and are formed by heptahelical OrX and Orco (co-receptor) proteins. A highly conserved calmodulin (CaM) binding site (CBS) 336SAIKYWVER344 within the second intracellular loop of Drosophila melanogaster Orco constitutes a target for regulating OR performance. Here we asked how a point mutation K339N in this CBS affects the olfactory performance of Drosophila melanogaster. We first asked how this mutation would affect the odor responses of olfactory sensory neurons (OSNs). Using Ca2+ imaging in an ex-vivo antenna preparation, we activated all OR (OrX/Orco) expressing neurons using the synthetic agonist VUAA1. In a next attempt, we restricted the OR spectrum to Or22a expressing neurons (Or22a/Orco) and stimulated these OSNs with the ligand ethyl hexanoate. In both approaches, we found that flies carrying the K339N point mutation in Orco display a reduced olfactory response. We also found that the mutation abolishes the capability of OSNs to sensitize by repeated weak odor stimuli. Next, we asked whether OrcoK339N might affect the odor localization performance. Using a wind tunnel bioassay, we found that odor localization in flies carrying the OrcoK339N mutation was severely diminished.
Collapse
Affiliation(s)
- Kalpana Jain
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Hans-Knöll-Strasse 8, 07745, Jena, Germany
| | - Sofia Lavista-Llanos
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Hans-Knöll-Strasse 8, 07745, Jena, Germany
| | - Veit Grabe
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Hans-Knöll-Strasse 8, 07745, Jena, Germany
| | - Bill S Hansson
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Hans-Knöll-Strasse 8, 07745, Jena, Germany
| | - Dieter Wicher
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Hans-Knöll-Strasse 8, 07745, Jena, Germany.
| |
Collapse
|
35
|
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
|
36
|
Intrinsically disordered protein domain of human ameloblastin in synthetic fusion with calmodulin increases calmodulin stability and modulates its function. Int J Biol Macromol 2020; 168:1-12. [PMID: 33290768 DOI: 10.1016/j.ijbiomac.2020.11.216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 11/21/2022]
Abstract
Constantly increasing attention to bioengineered proteins has led to the rapid development of new functional targets. Here we present the biophysical and functional characteristics of the newly designed CaM/AMBN-Ct fusion protein. The two-domain artificial target consists of calmodulin (CaM) and ameloblastin C-terminus (AMBN-Ct). CaM as a well-characterized calcium ions (Ca2+) binding protein offers plenty of options in terms of Ca2+ detection in biomedicine and biotechnologies. Highly negatively charged AMBN-Ct belongs to intrinsically disordered proteins (IDPs). CaM/AMBN-Ct was designed to open new ways of communication synergies between the domains with potential functional improvement. The character and function of CaM/AMBN-Ct were explored by biophysical and molecular modelling methods. Experimental studies have revealed increased stability and preserved CaM/AMBN-Ct function. The results of molecular dynamic simulations (MDs) outlined different interface patterns between the domains with potential allosteric communication within the fusion.
Collapse
|
37
|
Chakouri N, Diaz J, Yang PS, Ben-Johny M. Ca V channels reject signaling from a second CaM in eliciting Ca 2+-dependent feedback regulation. J Biol Chem 2020; 295:14948-14962. [PMID: 32820053 DOI: 10.1074/jbc.ra120.013777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/18/2020] [Indexed: 11/06/2022] Open
Abstract
Calmodulin (CaM) regulation of voltage-gated calcium (CaV1-2) channels is a powerful Ca2+-feedback mechanism to adjust channel activity in response to Ca2+ influx. Despite progress in resolving mechanisms of CaM-CaV feedback, the stoichiometry of CaM interaction with CaV channels remains ambiguous. Functional studies that tethered CaM to CaV1.2 suggested that a single CaM sufficed for Ca2+ feedback, yet biochemical, FRET, and structural studies showed that multiple CaM molecules interact with distinct interfaces within channel cytosolic segments, suggesting that functional Ca2+ regulation may be more nuanced. Resolving this ambiguity is critical as CaM is enriched in subcellular domains where CaV channels reside, such as the cardiac dyad. We here localized multiple CaMs to the CaV nanodomain by tethering either WT or mutant CaM that lack Ca2+-binding capacity to the pore-forming α-subunit of CaV1.2, CaV1.3, and CaV2.1 and/or the auxiliary β2A subunit. We observed that a single CaM tethered to either the α or β2A subunit tunes Ca2+ regulation of CaV channels. However, when multiple CaMs are localized concurrently, CaV channels preferentially respond to signaling from the α-subunit-tethered CaM. Mechanistically, the introduction of a second IQ domain to the CaV1.3 carboxyl tail switched the apparent functional stoichiometry, permitting two CaMs to mediate functional regulation. In all, Ca2+ feedback of CaV channels depends exquisitely on a single CaM preassociated with the α-subunit carboxyl tail. Additional CaMs that colocalize with the channel complex are unable to trigger Ca2+-dependent feedback of channel gating but may support alternate regulatory functions.
Collapse
Affiliation(s)
- Nourdine Chakouri
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York, USA
| | - Johanna Diaz
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York, USA
| | - Philemon S Yang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York, USA.
| |
Collapse
|
38
|
Su J, Gao Q, Yu L, Sun X, Feng R, Shao D, Yuan Y, Zhu Z, Sun X, Kameyama M, Hao L. The LQT-associated calmodulin mutant E141G induces disturbed Ca 2+-dependent binding and a flickering gating mode of the Ca V1.2 channel. Am J Physiol Cell Physiol 2020; 318:C991-C1004. [PMID: 32186935 DOI: 10.1152/ajpcell.00019.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Calmodulin (CaM) mutations are associated with congenital long QT (LQT) syndrome (LQTS), which may be related to the dysregulation of the cardiac-predominant Ca2+ channel isoform CaV1.2. Among various mutants, CaM-E141G was identified as a critical missense variant. However, the interaction of this CaM mutant with the CaV1.2 channel has not been determined. In this study, by utilizing a semiquantitative pull-down assay, we explored the interaction of CaM-E141G with CaM-binding peptide fragments of the CaV1.2 channel. Using the patch-clamp technique, we also investigated the electrophysiological effects of the mutant on CaV1.2 channel activity. We found that the maximum binding (Bmax) of CaM-E141G to the proximal COOH-terminal region, PreIQ-IQ, PreIQ, IQ, and NT (an NH2-terminal peptide) was decreased (by 17.71-59.26%) compared with that of wild-type CaM (CaM-WT). In particular, the Ca2+-dependent increase in Bmax became slower with the combination of CaM-E141G + PreIQ and IQ but faster in the case of NT. Functionally, CaM-WT and CaM-E141G at 500 nM Ca2+ decreased CaV1.2 channel activity to 24.88% and 55.99%, respectively, compared with 100 nM Ca2+, showing that the inhibitory effect was attenuated in CaM-E141G. The mean open time of the CaV1.2 channel was increased, and the number of blank traces with no channel opening was significantly decreased. Overall, CaM-E141G exhibits disrupted binding with the CaV1.2 channel and induces a flickering gating mode, which may result in the dysfunction of the CaV1.2 channel and, thus, the development of LQTS. The present study is the first to investigate the detailed binding properties and single-channel gating mode induced by the interaction of CaM-E141G with the CaV1.2 channel.
Collapse
Affiliation(s)
- Jingyang Su
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Qinghua Gao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China.,Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Lifeng Yu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xuanxuan Sun
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Rui Feng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Dongxue Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yuan Yuan
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Zhengnan Zhu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xuefei Sun
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Masaki Kameyama
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
39
|
Baik JY, Park EYJ, So I. Ca 2+/calmodulin-dependent regulation of polycystic kidney disease 2-like-1 by binding at C-terminal domain. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:277-286. [PMID: 32392919 PMCID: PMC7193909 DOI: 10.4196/kjpp.2020.24.3.277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 11/27/2022]
Abstract
Polycystic kidney disease 2-like-1 (PKD2L1), also known as polycystin-L or TRPP3, is a non-selective cation channel that regulates intracellular calcium concentration. Calmodulin (CaM) is a calcium binding protein, consisting of N-lobe and C-lobe with two calcium binding EF-hands in each lobe. In previous study, we confirmed that CaM is associated with desensitization of PKD2L1 and that CaM N-lobe and PKD2L1 EF-hand specifically are involved. However, the CaM-binding domain (CaMBD) and its inhibitory mechanism of PKD2L1 have not been identified. In order to identify CaM-binding anchor residue of PKD2L1, single mutants of putative CaMBD and EF-hand deletion mutants were generated. The current changes of the mutants were recorded with whole-cell patch clamp. The calmidazolium (CMZ), a calmodulin inhibitor, was used under different concentrations of intracellular. Among the mutants that showed similar or higher basal currents with that of the PKD2L1 wild type, L593A showed little change in current induced by CMZ. Co-expression of L593A with CaM attenuated the inhibitory effect of PKD2L1 by CaM. In the previous study it was inferred that CaM C-lobe inhibits channels by binding to PKD2L1 at 16 nM calcium concentration and CaM N-lobe at 100 nM. Based on the results at 16 nM calcium concentration condition, this study suggests that CaM C-lobe binds to Leu-593, which can be a CaM C-lobe anchor residue, to regulate channel activity. Taken together, our results provide a model for the regulation of PKD2L1 channel activity by CaM.
Collapse
Affiliation(s)
- Julia Young Baik
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Eunice Yon June Park
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
40
|
Peracchia C. Calmodulin-Mediated Regulation of Gap Junction Channels. Int J Mol Sci 2020; 21:E485. [PMID: 31940951 PMCID: PMC7014422 DOI: 10.3390/ijms21020485] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 12/25/2022] Open
Abstract
Evidence that neighboring cells uncouple from each other as one dies surfaced in the late 19th century, but it took almost a century for scientists to start understanding the uncoupling mechanism (chemical gating). The role of cytosolic free calcium (Ca2+i) in cell-cell channel gating was first reported in the mid-sixties. In these studies, only micromolar [Ca2+]i were believed to affect gating-concentrations reachable only in cell death, which would discard Ca2+i as a fine modulator of cell coupling. More recently, however, numerous researchers, including us, have reported the effectiveness of nanomolar [Ca2+]i. Since connexins do not have high-affinity calcium sites, the effectiveness of nanomolar [Ca2+]i suggests the role of Ca-modulated proteins, with calmodulin (CaM) being most obvious. Indeed, in 1981 we first reported that a CaM-inhibitor prevents chemical gating. Since then, the CaM role in gating has been confirmed by studies that tested it with a variety of approaches such as treatments with CaM-inhibitors, inhibition of CaM expression, expression of CaM mutants, immunofluorescent co-localization of CaM and gap junctions, and binding of CaM to peptides mimicking connexin domains identified as CaM targets. Our gating model envisions Ca2+-CaM to directly gate the channels by acting as a plug ("Cork" gating model), and probably also by affecting connexin conformation.
Collapse
Affiliation(s)
- Camillo Peracchia
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
41
|
New Insights in the IP 3 Receptor and Its Regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:243-270. [PMID: 31646513 DOI: 10.1007/978-3-030-12457-1_10] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) is a Ca2+-release channel mainly located in the endoplasmic reticulum (ER). Three IP3R isoforms are responsible for the generation of intracellular Ca2+ signals that may spread across the entire cell or occur locally in so-called microdomains. Because of their ubiquitous expression, these channels are involved in the regulation of a plethora of cellular processes, including cell survival and cell death. To exert their proper function a fine regulation of their activity is of paramount importance. In this review, we will highlight the recent advances in the structural analysis of the IP3R and try to link these data with the newest information concerning IP3R activation and regulation. A special focus of this review will be directed towards the regulation of the IP3R by protein-protein interaction. Especially the protein family formed by calmodulin and related Ca2+-binding proteins and the pro- and anti-apoptotic/autophagic Bcl-2-family members will be highlighted. Finally, recently identified and novel IP3R regulatory proteins will be discussed. A number of these interactions are involved in cancer development, illustrating the potential importance of modulating IP3R-mediated Ca2+ signaling in cancer treatment.
Collapse
|
42
|
Dual action of amitriptyline on NMDA receptors: enhancement of Ca-dependent desensitization and trapping channel block. Sci Rep 2019; 9:19454. [PMID: 31857688 PMCID: PMC6923474 DOI: 10.1038/s41598-019-56072-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 12/04/2019] [Indexed: 12/18/2022] Open
Abstract
Although the tricyclic antidepressant amitriptyline (ATL) is widely used in the clinic, the mechanism underlying its high therapeutic efficacy against neuropathic pain remains unclear. NMDA receptors (NMDARs) represent a target for ATL and are involved in sensitization of neuropathic pain. Here we describe two actions of ATL on NMDARs: 1) enhancement of Ca2+-dependent desensitization and 2) trapping channel block. Inhibition of NMDARs by ATL was found to be dependent upon external Ca2+ concentration ([Ca2+]) in a voltage-independent manner, with an IC50 of 0.72 μM in 4 mM [Ca2+]. The ATL IC50 value increased exponentially with decreasing [Ca2+], with an e-fold change observed per 0.69 mM decrease in [Ca2+]. Loading neurons with BAPTA abolished Ca2+-dependent inhibition, suggesting that Ca2+ affects NMDARs from the cytosol. Since there is one known Ca2+-dependent process in gating of NMDARs, we conclude that ATL most likely promotes Ca2+-dependent desensitization. We also found ATL to be a trapping open-channel blocker of NMDARs with an IC50 of 220 µM at 0 mV. An e-fold change in ATL IC50 was observed to occur with a voltage shift of 50 mV in 0.25 mM [Ca2+]. Thus, we disclose here a robust dependence of ATL potency on extracellular [Ca2+], and demonstrate that ATL bound in the NMDAR pore can be trapped by closure of the channel.
Collapse
|
43
|
Boczek T, Radzik T, Ferenc B, Zylinska L. The Puzzling Role of Neuron-Specific PMCA Isoforms in the Aging Process. Int J Mol Sci 2019; 20:ijms20246338. [PMID: 31888192 PMCID: PMC6941135 DOI: 10.3390/ijms20246338] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 01/02/2023] Open
Abstract
The aging process is a physiological phenomenon associated with progressive changes in metabolism, genes expression, and cellular resistance to stress. In neurons, one of the hallmarks of senescence is a disturbance of calcium homeostasis that may have far-reaching detrimental consequences on neuronal physiology and function. Among several proteins involved in calcium handling, plasma membrane Ca2+-ATPase (PMCA) is the most sensitive calcium detector controlling calcium homeostasis. PMCA exists in four main isoforms and PMCA2 and PMCA3 are highly expressed in the brain. The overall effects of impaired calcium extrusion due to age-dependent decline of PMCA function seem to accumulate with age, increasing the susceptibility to neurotoxic insults. To analyze the PMCA role in neuronal cells, we have developed stable transfected differentiated PC12 lines with down-regulated PMCA2 or PMCA3 isoforms to mimic age-related changes. The resting Ca2+ increased in both PMCA-deficient lines affecting the expression of several Ca2+-associated proteins, i.e., sarco/endoplasmic Ca2+-ATPase (SERCA), calmodulin, calcineurin, GAP43, CCR5, IP3Rs, and certain types of voltage-gated Ca2+ channels (VGCCs). Functional studies also demonstrated profound changes in intracellular pH regulation and mitochondrial metabolism. Moreover, modification of PMCAs membrane composition triggered some adaptive processes to counterbalance calcium overload, but the reduction of PMCA2 appeared to be more detrimental to the cells than PMCA3.
Collapse
Affiliation(s)
- Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University, 92-215 Lodz, Poland; (T.B.); (T.R.); (B.F.)
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Tomasz Radzik
- Department of Molecular Neurochemistry, Medical University, 92-215 Lodz, Poland; (T.B.); (T.R.); (B.F.)
| | - Bozena Ferenc
- Department of Molecular Neurochemistry, Medical University, 92-215 Lodz, Poland; (T.B.); (T.R.); (B.F.)
| | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University, 92-215 Lodz, Poland; (T.B.); (T.R.); (B.F.)
- Correspondence: ; Tel.: +48-42-272-5680
| |
Collapse
|
44
|
Yamaguchi N. Molecular Insights into Calcium Dependent Regulation of Ryanodine Receptor Calcium Release Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1131:321-336. [DOI: 10.1007/978-3-030-12457-1_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
45
|
Polat OK, Uno M, Maruyama T, Tran HN, Imamura K, Wong CF, Sakaguchi R, Ariyoshi M, Itsuki K, Ichikawa J, Morii T, Shirakawa M, Inoue R, Asanuma K, Reiser J, Tochio H, Mori Y, Mori MX. Contribution of Coiled-Coil Assembly to Ca 2+/Calmodulin-Dependent Inactivation of TRPC6 Channel and its Impacts on FSGS-Associated Phenotypes. J Am Soc Nephrol 2019; 30:1587-1603. [PMID: 31266820 DOI: 10.1681/asn.2018070756] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND TRPC6 is a nonselective cation channel, and mutations of this gene are associated with FSGS. These mutations are associated with TRPC6 current amplitude amplification and/or delay of the channel inactivation (gain-of-function phenotype). However, the mechanism of the gain-of-function in TRPC6 activity has not yet been clearly solved. METHODS We performed electrophysiologic, biochemical, and biophysical experiments to elucidate the molecular mechanism underlying calmodulin (CaM)-mediated Ca2+-dependent inactivation (CDI) of TRPC6. To address the pathophysiologic contribution of CDI, we assessed the actin filament organization in cultured mouse podocytes. RESULTS Both lobes of CaM helped induce CDI. Moreover, CaM binding to the TRPC6 CaM-binding domain (CBD) was Ca2+-dependent and exhibited a 1:2 (CaM/CBD) stoichiometry. The TRPC6 coiled-coil assembly, which brought two CBDs into adequate proximity, was essential for CDI. Deletion of the coiled-coil slowed CDI of TRPC6, indicating that the coiled-coil assembly configures both lobes of CaM binding on two CBDs to induce normal CDI. The FSGS-associated TRPC6 mutations within the coiled-coil severely delayed CDI and often increased TRPC6 current amplitudes. In cultured mouse podocytes, FSGS-associated channels and CaM mutations led to sustained Ca2+ elevations and a disorganized cytoskeleton. CONCLUSIONS The gain-of-function mechanism found in FSGS-causing mutations in TRPC6 can be explained by impairments of the CDI, caused by disruptions of TRPC's coiled-coil assembly which is essential for CaM binding. The resulting excess Ca2+ may contribute to structural damage in the podocytes.
Collapse
Affiliation(s)
- Onur K Polat
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering
| | - Masatoshi Uno
- Department of Biophysics, Graduate School of Science.,Department of Molecular Engineering, Graduate School of Engineering
| | - Terukazu Maruyama
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering
| | - Ha Nam Tran
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering.,Department of Technology and Ecology, Laboratory of Environmental Systems Biology, Graduate School of Global Environmental Studies
| | - Kayo Imamura
- Department of Biophysics, Graduate School of Science
| | - Chee Fah Wong
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering.,Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, Perak, Malaysia
| | - Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering.,Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Mariko Ariyoshi
- Department of Molecular Engineering, Graduate School of Engineering
| | - Kyohei Itsuki
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Jun Ichikawa
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Takashi Morii
- Institute of Advanced Energy, Kyoto University, Kyoto, Japan
| | | | - Ryuji Inoue
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Katsuhiko Asanuma
- Department of Nephrology, School of Medicine, Chiba University, Chiba, Japan
| | - Jochen Reiser
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | | | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering
| | - Masayuki X Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering,
| |
Collapse
|
46
|
Edington SC, Halling DB, Bennett SM, Middendorf TR, Aldrich RW, Baiz CR. Non-Additive Effects of Binding Site Mutations in Calmodulin. Biochemistry 2019; 58:2730-2739. [PMID: 31124357 DOI: 10.1021/acs.biochem.9b00096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Despite decades of research on ion-sensing proteins, gaps persist in the understanding of ion binding affinity and selectivity even in well-studied proteins such as calmodulin. Site-directed mutagenesis is a powerful and popular tool for addressing outstanding questions about biological ion binding and is employed to selectively deactivate binding sites and insert chromophores at advantageous positions within ion binding structures. However, even apparently nonperturbative mutations can distort the binding dynamics they are employed to measure. We use Fourier transform infrared (FTIR) and ultrafast two-dimensional infrared (2D IR) spectroscopy of the carboxylate asymmetric stretching mode in calmodulin as a mutation- and label-independent probe of the conformational perturbations induced in calmodulin's binding sites by two classes of mutation, tryptophan insertion and carboxylate side-chain deletion, commonly used to study ion binding in proteins. Our results show that these mutations not only affect ion binding but also induce changes in calmodulin's conformational landscape along coordinates not probed by vibrational spectroscopy, remaining invisible without additional perturbation of binding site structure. Comparison of FTIR line shapes with 2D IR diagonal slices provides a clear example of how nonlinear spectroscopy produces well-resolved line shapes, refining otherwise featureless spectral envelopes into more informative vibrational spectra of proteins.
Collapse
Affiliation(s)
- Sean C Edington
- Department of Chemistry , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - D Brent Halling
- Department of Neuroscience , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Suzanna M Bennett
- Department of Neuroscience , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Thomas R Middendorf
- Department of Neuroscience , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Richard W Aldrich
- Department of Neuroscience , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Carlos R Baiz
- Department of Chemistry , The University of Texas at Austin , Austin , Texas 78712 , United States
| |
Collapse
|
47
|
Villalobo A, González-Muñoz M, Berchtold MW. Proteins with calmodulin-like domains: structures and functional roles. Cell Mol Life Sci 2019; 76:2299-2328. [PMID: 30877334 PMCID: PMC11105222 DOI: 10.1007/s00018-019-03062-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 02/26/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022]
Abstract
The appearance of modular proteins is a widespread phenomenon during the evolution of proteins. The combinatorial arrangement of different functional and/or structural domains within a single polypeptide chain yields a wide variety of activities and regulatory properties to the modular proteins. In this review, we will discuss proteins, that in addition to their catalytic, transport, structure, localization or adaptor functions, also have segments resembling the helix-loop-helix EF-hand motifs found in Ca2+-binding proteins, such as calmodulin (CaM). These segments are denoted CaM-like domains (CaM-LDs) and play a regulatory role, making these CaM-like proteins sensitive to Ca2+ transients within the cell, and hence are able to transduce the Ca2+ signal leading to specific cellular responses. Importantly, this arrangement allows to this group of proteins direct regulation independent of other Ca2+-sensitive sensor/transducer proteins, such as CaM. In addition, this review also covers CaM-binding proteins, in which their CaM-binding site (CBS), in the absence of CaM, is proposed to interact with other segments of the same protein denoted CaM-like binding site (CLBS). CLBS are important regulatory motifs, acting either by keeping these CaM-binding proteins inactive in the absence of CaM, enhancing the stability of protein complexes and/or facilitating their dimerization via CBS/CLBS interaction. The existence of proteins containing CaM-LDs or CLBSs substantially adds to the enormous versatility and complexity of Ca2+/CaM signaling.
Collapse
Affiliation(s)
- Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Arturo Duperier 4, 28029, Madrid, Spain.
- Instituto de Investigaciones Sanitarias, Hospital Universitario La Paz, Edificio IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.
| | - María González-Muñoz
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Arturo Duperier 4, 28029, Madrid, Spain
| | - Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, 2100, Copenhagen, Denmark.
| |
Collapse
|
48
|
Burgoyne RD, Helassa N, McCue HV, Haynes LP. Calcium Sensors in Neuronal Function and Dysfunction. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035154. [PMID: 30833454 DOI: 10.1101/cshperspect.a035154] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium signaling in neurons as in other cell types can lead to varied changes in cellular function. Neuronal Ca2+ signaling processes have also become adapted to modulate the function of specific pathways over a wide variety of time domains and these can have effects on, for example, axon outgrowth, neuronal survival, and changes in synaptic strength. Ca2+ also plays a key role in synapses as the trigger for fast neurotransmitter release. Given its physiological importance, abnormalities in neuronal Ca2+ signaling potentially underlie many different neurological and neurodegenerative diseases. The mechanisms by which changes in intracellular Ca2+ concentration in neurons can bring about diverse responses is underpinned by the roles of ubiquitous or specialized neuronal Ca2+ sensors. It has been established that synaptotagmins have key functions in neurotransmitter release, and, in addition to calmodulin, other families of EF-hand-containing neuronal Ca2+ sensors, including the neuronal calcium sensor (NCS) and the calcium-binding protein (CaBP) families, play important physiological roles in neuronal Ca2+ signaling. It has become increasingly apparent that these various Ca2+ sensors may also be crucial for aspects of neuronal dysfunction and disease either indirectly or directly as a direct consequence of genetic variation or mutations. An understanding of the molecular basis for the regulation of the targets of the Ca2+ sensors and the physiological roles of each protein in identified neurons may contribute to future approaches to the development of treatments for a variety of human neuronal disorders.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Hannah V McCue
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
49
|
Park EYJ, Baik JY, Kwak M, So I. The role of calmodulin in regulating calcium-permeable PKD2L1 channel activity. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:219-227. [PMID: 31080352 PMCID: PMC6488707 DOI: 10.4196/kjpp.2019.23.3.219] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 11/15/2022]
Abstract
Polycystic kidney disease 2-like-1 (PKD2L1), polycystin-L or transient receptor potential polycystin 3 (TRPP3) is a TRP superfamily member. It is a calcium-permeable non-selective cation channel that regulates intracellular calcium concentration and thereby calcium signaling. Although the calmodulin (CaM) inhibitor, calmidazolium, is an activator of the PKD2L1 channel, the activating mechanism remains unclear. The purpose of this study is to clarify whether CaM takes part in the regulation of the PKD2L1 channel, and if so, how. With patch clamp techniques, we observed the current amplitudes of PKD2L1 significantly reduced when coexpressed with CaM and CaMΔN. This result suggests that the N-lobe of CaM carries a more crucial role in regulating PKD2L1 and guides us into our next question on the different functions of two lobes of CaM. We also identified the predicted CaM binding site, and generated deletion and truncation mutants. The mutants showed significant reduction in currents losing PKD2L1 current-voltage curve, suggesting that the C-terminal region from 590 to 600 is crucial for maintaining the functionality of the PKD2L1 channel. With PKD2L1608Stop mutant showing increased current amplitudes, we further examined the functional importance of EF-hand domain. Along with co-expression of CaM, ΔEF-hand mutant also showed significant changes in current amplitudes and potentiation time. Our findings suggest that there is a constitutive inhibition of EF-hand and binding of CaM C-lobe on the channel in low calcium concentration. At higher calcium concentration, calcium ions occupy the N-lobe as well as the EF-hand domain, allowing the two to compete to bind to the channel.
Collapse
Affiliation(s)
- Eunice Yon June Park
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Julia Young Baik
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Misun Kwak
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
50
|
Greenwald EC, Mehta S, Zhang J. Genetically Encoded Fluorescent Biosensors Illuminate the Spatiotemporal Regulation of Signaling Networks. Chem Rev 2018; 118:11707-11794. [PMID: 30550275 PMCID: PMC7462118 DOI: 10.1021/acs.chemrev.8b00333] [Citation(s) in RCA: 316] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cellular signaling networks are the foundation which determines the fate and function of cells as they respond to various cues and stimuli. The discovery of fluorescent proteins over 25 years ago enabled the development of a diverse array of genetically encodable fluorescent biosensors that are capable of measuring the spatiotemporal dynamics of signal transduction pathways in live cells. In an effort to encapsulate the breadth over which fluorescent biosensors have expanded, we endeavored to assemble a comprehensive list of published engineered biosensors, and we discuss many of the molecular designs utilized in their development. Then, we review how the high temporal and spatial resolution afforded by fluorescent biosensors has aided our understanding of the spatiotemporal regulation of signaling networks at the cellular and subcellular level. Finally, we highlight some emerging areas of research in both biosensor design and applications that are on the forefront of biosensor development.
Collapse
Affiliation(s)
- Eric C Greenwald
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Sohum Mehta
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Jin Zhang
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| |
Collapse
|