1
|
Ong ST, Nam YW, Nasburg JA, Ramanishka A, Ng XR, Zhuang Z, Goay SSM, Nguyen HM, Singh L, Singh V, Rivera A, Eyster ME, Xu Y, Alper SL, Wulff H, Zhang M, Chandy KG. Design and structural basis of selective 1,4-dihydropyridine inhibitors of the calcium-activated potassium channel K Ca3.1. Proc Natl Acad Sci U S A 2025; 122:e2425494122. [PMID: 40294255 PMCID: PMC12067266 DOI: 10.1073/pnas.2425494122] [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: 12/15/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
The 1,4-dihydropyridines, drugs with well-established bioavailability and toxicity profiles, have proven efficacy in treating human hypertension, peripheral vascular disorders, and coronary artery disease. Every 1,4-dihydropyridine in clinical use blocks L-type voltage-gated calcium channels. We now report our development, using selective optimization of a side activity (SOSA), of a class of 1,4-dihydropyridines that selectively and potently inhibit the intermediate-conductance calcium-activated K+ channel KCa3.1, a validated therapeutic target for diseases affecting many organ systems. One of these 1,4-dihydropyridines, DHP-103, blocked KCa3.1 with an IC50 of 6 nM and exhibited exquisite selectivity over calcium channels and a panel of >100 additional molecular targets. Using high-resolution structure determination by cryogenic electron microscopy together with mutagenesis and electrophysiology, we delineated the drug binding pocket for DHP-103 within the water-filled central cavity of the KCa3.1 channel pore, where bound drug directly impedes ion permeation. DHP-103 inhibited gain-of-function mutant KCa3.1 channels that cause hereditary xerocytosis, suggesting its potential use as a therapeutic for this hemolytic anemia. In a rat model of acute ischemic stroke, the second leading cause of death worldwide, DHP-103 administered 12 h postischemic insult in proof-of-concept studies reduced infarct volume, improved balance beam performance (measure of proprioception) and decreased numbers of activated microglia in infarcted areas. KCa3.1-selective 1,4-dihydropyridines hold promise for the many diseases for which KCa3.1 has been experimentally confirmed as a therapeutic target.
Collapse
Affiliation(s)
- Seow Theng Ong
- Lee Kong Chian School of Medicine-Innovative CRO Explorer Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore636921, Singapore
| | - Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA92618
| | - Joshua A. Nasburg
- Department of Pharmacology, School of Medicine, University of California, Davis, CA95616
| | - Alena Ramanishka
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA92618
| | - Xuan Rui Ng
- Lee Kong Chian School of Medicine-Innovative CRO Explorer Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore636921, Singapore
| | - Zhong Zhuang
- Lee Kong Chian School of Medicine-Innovative CRO Explorer Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore636921, Singapore
| | - Stephanie Shee Min Goay
- Lee Kong Chian School of Medicine-Innovative CRO Explorer Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore636921, Singapore
| | - Hai M. Nguyen
- Department of Pharmacology, School of Medicine, University of California, Davis, CA95616
| | - Latika Singh
- Department of Pharmacology, School of Medicine, University of California, Davis, CA95616
| | - Vikrant Singh
- Department of Pharmacology, School of Medicine, University of California, Davis, CA95616
| | - Alicia Rivera
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
| | - M. Elaine Eyster
- Division of Blood and Vascular Disorders, Department of Medicine, Penn State College of Medicine, Milton S. Hershey Medical Center, Hershey, PA17033
| | - Yang Xu
- Division of Cryogenic Electron Microscopy and Bioimaging, Stanford Synchrotron Radiation Lightsource, Stanford Linear Accelerator Center National Accelerator Laboratory, Stanford University, Menlo Park, CA94025
| | - Seth L. Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA02215
- Department of Medicine, Harvard Medical School, Boston, MA02115
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA95616
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA92618
| | - K. George Chandy
- Lee Kong Chian School of Medicine-Innovative CRO Explorer Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore636921, Singapore
| |
Collapse
|
2
|
Nam YW, Im D, Garcia ASC, Tringides ML, Nguyen HM, Liu Y, Orfali R, Ramanishka A, Pintilie G, Su CC, Cui M, Logothetis DE, Yu EW, Wulff H, Chandy KG, Zhang M. Cryo-EM structures of the small-conductance Ca 2+-activated K Ca2.2 channel. Nat Commun 2025; 16:3690. [PMID: 40246884 PMCID: PMC12006403 DOI: 10.1038/s41467-025-59061-1] [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: 07/22/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Small-conductance Ca2+-activated K+ (KCa2.1-KCa2.3) channels modulate neuronal and cardiac excitability. We report cryo-electron microscopy structures of the KCa2.2 channel in complex with calmodulin and Ca2+, alone or bound to two small molecule inhibitors, at 3.18, 3.50, 2.99 and 2.97 angstrom resolution, respectively. Extracellular S3-S4 loops in β-hairpin configuration form an outer canopy over the pore with an aromatic box at the canopy's center. Each S3-S4 β-hairpin is tethered to the selectivity filter in the neighboring subunit by inter-subunit hydrogen bonds. This hydrogen bond network flips the aromatic residue (Tyr362) in the filter's GYG signature by 180°, causing the outer selectivity filter to widen and water to enter the filter. Disruption of the tether by a mutation narrows the outer selectivity filter, realigns Tyr362 to the position seen in other K+ channels, and significantly increases unitary conductance. UCL1684, a mimetic of the bee venom peptide apamin, sits atop the canopy and occludes the opening in the aromatic box. AP14145, an analogue of a therapeutic for atrial fibrillation, binds in the central cavity below the selectivity filter and induces closure of the inner gate. These structures provide a basis for understanding the small unitary conductance and pharmacology of KCa2.x channels.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Dohyun Im
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ana Santa Cruz Garcia
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy and Pharmaceutical Sciences, Boston, MA, USA
| | - Marios L Tringides
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Hai Minh Nguyen
- Department of Pharmacology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Yan Liu
- Division of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Alena Ramanishka
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Grigore Pintilie
- Department of Bioengineering and Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Chih-Chia Su
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy and Pharmaceutical Sciences, Boston, MA, USA
| | - Diomedes E Logothetis
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy and Pharmaceutical Sciences, Boston, MA, USA
| | - Edward W Yu
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California Davis, Davis, CA, USA
| | - K George Chandy
- LKCMedicine-ICE Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA.
| |
Collapse
|
3
|
Cichon J, Joseph TT, Lu X, Wasilczuk AZ, Kelz MB, Mennerick SJ, Zorumski CF, Nagele P. Nitrous oxide activates layer 5 prefrontal neurons via SK2 channel inhibition for antidepressant effect. Nat Commun 2025; 16:2999. [PMID: 40180931 PMCID: PMC11968965 DOI: 10.1038/s41467-025-57951-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Nitrous oxide (N2O) induces rapid and durable antidepressant effects. The cellular and circuit mechanisms mediating this process are not known. Here we find that a single dose of inhaled N2O induces rapid and specific activation of layer V (L5) pyramidal neurons in the cingulate cortex of rodents exposed to chronic stress conditions. N2O-induced L5 activation rescues a stress-associated hypoactivity state, persists following exposure, and is necessary for its antidepressant-like activity. Although NMDA-receptor antagonism is believed to be a primary mechanism of action for N2O, L5 neurons activate even when NMDA-receptor function is attenuated through both pharmacological and genetic approaches. By examining different molecular and circuit targets, we identify N2O-induced inhibition of calcium-sensitive potassium (SK2) channels as a key molecular interaction responsible for driving specific L5 activity along with ensuing antidepressant-like effects. These results suggest that N2O-induced L5 activation is crucial for its fast antidepressant action and this effect involves novel and specific molecular actions in distinct cortical cell types.
Collapse
Affiliation(s)
- Joseph Cichon
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Thomas T Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinguo Lu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrzej Z Wasilczuk
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Max B Kelz
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven J Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Nagele
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| |
Collapse
|
4
|
Orenbuch R, Shearer CA, Kollasch AW, Spinner HD, Hopf TA, van Niekerk L, Franceschi D, Dias M, Frazer J, Marks DS. Proteome-wide model for human disease genetics. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2023.11.27.23299062. [PMID: 38076790 PMCID: PMC10705666 DOI: 10.1101/2023.11.27.23299062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Identifying variants driving disease accelerates both genetic diagnosis and therapeutic development, but missense variants still present a bottleneck as their effects are less straightforward than truncations or nonsense mutations. While computational prediction methods are sufficiently accurate to be of clinical value for variants in known disease genes, they do not generalize well to other genes as the scores are not calibrated across the proteome 1-6 . To address this, we developed a deep generative model, popEVE, that combines evolutionary information with population sequence data 7 and achieves state-of-the-art performance on a suite of proteome-wide prediction tasks, without overestimating the prevalence of deleterious variants in the population. popEVE identifies 442 genes in a developmental disorder cohort 8 , including evidence of 123 novel candidates, many without the need for cohort-wide enrichment. Candidate genes are functionally similar to known developmental disorder genes and case variants tend to fall in functionally important regions of these genes. Finally, we show that these findings can be reproduced from analysis of the patient exomes alone, demonstrating that popEVE provides a new avenue for genetic analysis in situations where traditional methods fail, including genetic diagnosis of rare-as-one diseases, even in the absence of parent sequencing.
Collapse
|
5
|
Nasburg JA, Rouen KC, Dietrich CJ, Shim H, Zhang M, Vorobyov I, Wulff H. 6,7-Dichloro-1H-indole-2,3-dione-3-oxime functions as a superagonist for the intermediate-conductance Ca 2+-activated K + channel K Ca3.1. Mol Pharmacol 2025; 107:100018. [PMID: 40068526 DOI: 10.1016/j.molpha.2025.100018] [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] [Received: 09/09/2024] [Accepted: 01/26/2025] [Indexed: 04/01/2025] Open
Abstract
NS309 (6,7-dichloro-1H-indole-2,3-dione-3-oxime) is widely used as a pharmacological tool to increase the activity of small- and intermediate-conductance calcium-activated potassium channels. NS309 is assumed to function as a positive allosteric gating modulator. However, its binding site and the molecular details of its action remain unknown. Here, we show that NS309 has a profound effect on the calcium-dependent gating of the intermediate-conductance Ca2+-activated K+ channel KCa3.1. In inside-out experiments, 10 μM NS309 shifted the calcium EC50 from 430 to 31 nM. In whole-cell experiments, changing free intracellular calcium from 250 nM to 3 μM decreased the EC50 of NS309 from 74 to 8.6 nM. We further observed that NS309 could elicit greater responses than saturating calcium, making it a "superagonist." Molecular modeling suggested 2 possible binding sites for NS309 in KCa3.1, which we probed by mutagenesis and determined that NS309 is binding in the interface between the S45A segment of the intracellular S4-S5 linker and the N-lobe of the channel-associated calmodulin. Molecular dynamic simulations revealed that NS309 pushes several water molecules out of the interface pocket, establishes stable contacts with S181 and L185 in the S45A segment of KCa3.1 and E54 in calmodulin, and promotes longer sustained widening of the inner gate of KCa3.1 at V282 in the S6 segment. Polar substitutions of the hydrophobic-gating residues V282 and A279 resulted in constitutively open channels that could not be further potentiated by NS309, suggesting that NS309 produces its agonistic effects by increasing the open probability of the inner gate of KCa3.1. SIGNIFICANCE STATEMENT: The publication of the full-length cryo-electron microscopy structure of the intermediate-conductance Ca2+-activated K+ channel KCa3.1 suggested that the previously reported binding site of NS309 (6,7-dichloro-1H-indole-2,3-dione-3-oxime) was a crystallization artifact because this structure only included the C-terminus and the channel-associated calmodulin. This study demonstrates that the true binding site of NS309 is located between the S4 and S5 linker of KCa3.1 and the N-lobe of calmodulin. NS309 acts as a stabilizing force within the gating interface and increases the open probability of the inner hydrophobic gate.
Collapse
Affiliation(s)
- Joshua A Nasburg
- Department of Pharmacology, School of Medicine, University of California, Davis, California
| | - Kyle C Rouen
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California
| | - Connor J Dietrich
- Department of Pharmacology, School of Medicine, University of California, Davis, California
| | - Heesung Shim
- Physical and Life Sciences, Lawrence Livermore National Laboratory, Livermore, California
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| | - Igor Vorobyov
- Department of Pharmacology, School of Medicine, University of California, Davis, California; Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, California.
| |
Collapse
|
6
|
Jedele S, Allegrini B, Guizouarn H, Etchebest C. Investigating the Dynamics of the KCNN4 Channel: From the Determination of the Complete K + Permeation Pathway Across the Channel to Its Opening by PIP2. J Chem Inf Model 2025; 65:2116-2128. [PMID: 39928097 DOI: 10.1021/acs.jcim.4c01711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
KCNN4 is a calcium (Ca)-activated potassium channel for which Ca2+ sensitivity is conferred by calmodulin (CaM) that constitutively binds to the channel. Until the main part of the structure bound to CaM has been resolved, in silico studies had used homology models derived from the well characterized transmembrane domain of other K+ channels, limiting the functional investigation to this particular region. Thus, how the regulatory domains of KCNN4 communicate with each other and where the possible gates are located across the complete structure are still not well understood. Here we present for the first time results obtained from the investigation of full-length models of the channel in different conformational states and molecular contexts using classical all-atom molecular dynamic simulations. The simulations covered two activated states (open and closed) and a preactivated state of the channel embedded in a simple membrane model and a model of red blood cell membrane, where the channel is functionally expressed in vivo. Surprisingly, the intracellular domain was refractory to the entrance of K+, whatever the state of the channel was, allowing the K+ ions to enter and exit the channel only through two newly identified restrained diffusion spots. Inside the channel, the K+ flux was controlled by the V282 residue closing the pore region when the CaM N-lobes were not bound. This flux was compatible with the passage of fully or partially hydrated K+, depending on the opening level. Finally, the presence of phosphatidylinositol-4,5-bisphosphate (PIP2), a well-known K+-channel modulator, in a putative binding site of KCNN4 clearly facilitated the opening of the V282 restriction. Thus, in addition to the elucidation of the possible complete K+ permeation pathway throughout KCNN4, our results confirmed the direct activatory role of PIP2, associated with the channel opening, and provide a first insight into the architecture and the behavior of the complete intracellular region of KCNN4.
Collapse
Affiliation(s)
- Stephane Jedele
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
| | | | | | - Catherine Etchebest
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
| |
Collapse
|
7
|
Kuriyama M, Hirose H, Kawaguchi Y, Michibata J, Maekawa M, Futaki S. KCNN4 as a genomic determinant of cytosolic delivery by the attenuated cationic lytic peptide L17E. Mol Ther 2025; 33:595-614. [PMID: 39748507 PMCID: PMC11852704 DOI: 10.1016/j.ymthe.2024.12.050] [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] [Received: 07/27/2024] [Revised: 11/19/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
The development of a cytosolic delivery strategy for biopharmaceuticals is one of the central issues in drug development. Knowledge of the mechanisms underlying these processes may also pave the way for the discovery of novel delivery systems. L17E is an attenuated cationic amphiphilic lytic (ACAL) peptide developed by our research group that shows promise for cytosolic antibody delivery. In this study, given the high efficacy of L17E in cytosolic delivery, we investigated the mechanism of action of L17E in detail. L17E was found to achieve cytosolic delivery predominantly by transient disruption of the plasma membrane without the need for endocytosis. Importantly, the cell-line selectivity studies of L17E revealed a strong correlation between the efficiency of L17E-mediated delivery and the expression level of KCNN4, the gene encoding the calcium-activated potassium channel KCa3.1. Genetic and pharmacological regulation of KCNN4 expression and KCa3.1 activity, respectively, correlate closely with the efficiency of L17E-mediated cytosolic delivery, suggesting the importance of membrane-potential regulation by extracellular Ca2+ influx. Therefore, the activity of the L17E is relevant to the calcium-activated potassium channel.
Collapse
Affiliation(s)
- Masashi Kuriyama
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Hisaaki Hirose
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| | - Yoshimasa Kawaguchi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Junya Michibata
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Masashi Maekawa
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
8
|
Allegrini B, Mignotet M, Rapetti-Mauss R, Borgese F, Soriani O, Guizouarn H. A new regulation mechanism for KCNN4, the Ca 2+-dependent K + channel, by molecular interactions with the Ca 2+pump PMCA4b. J Biol Chem 2025; 301:108114. [PMID: 39716493 PMCID: PMC11787511 DOI: 10.1016/j.jbc.2024.108114] [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] [Received: 09/02/2024] [Revised: 11/20/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
KCNN4, a Ca2+-activated K+ channel, is involved in various physiological and pathological processes. It is essential for epithelial transport, immune system, and other physiological mechanisms, but its activation is also involved in cancer pathophysiology as well as red blood cell (RBC) disorders. The activation of KCNN4 in RBC leads to loss of KCl and water, a mechanism known as the "Gardos effect" described 70 years ago. This Ca2+-induced dehydration is irreversible in human RBC and must be tightly controlled to prevent not only hemolysis but also alterations in RBC rheological properties. In this study, we have investigated the regulation of KCNN4 activity after changes in RBC Ca2+ concentration. Using electrophysiology, immunoprecipitation, and proximity ligation assay in human embryonic kidney 293-transfected cells, K562 cells, or RBCs, we have found that KCNN4 and the Ca2+ pump PMCA4b (plasma membrane calcium-transporting ATPase 4b) interact tightly with each other, such that the C-terminal domain of PMCA4b regulates KCNN4 activity, independently of the Ca2+ extrusion activity of the pump. This regulation was not restricted to KCNN4: the small-conductance Ca2+-activated K+ channel KCNN2 was similarly regulated by the calcium pump. We propose a new mechanism that could control KCNN4 activity by a molecular inhibitory interaction with PMCA4b. It is suggested that this mechanism could attenuate erythrocyte dehydration in response to an increase in intracellular Ca2+.
Collapse
Affiliation(s)
- Benoit Allegrini
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | - Morgane Mignotet
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | | | - Franck Borgese
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | - Olivier Soriani
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France
| | - Hélène Guizouarn
- Université Côte d'Azur, CNRS, Inserm, Institut Biologie Valrose, Nice, France; Laboratory of Excellence for RBC, LABEX GR-Ex, Paris, France.
| |
Collapse
|
9
|
Thale I, Naß E, Vinnenberg L, Todesca LM, Budde T, Maisuls I, Strassert CA, Schwab A, Wünsch B. Fluorescent Probes to Image the K Ca3.1 Channel in Tumor Cells. Pharmaceutics 2025; 17:154. [PMID: 40006521 PMCID: PMC11859423 DOI: 10.3390/pharmaceutics17020154] [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: 12/19/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The Ca2+-activated K+ channel KCa3.1 is not only involved in physiological processes such as immune reactions and control of vascular tone, but is highly expressed in various tumor entities. Thus, imaging of KCa3.1 channels comes into focus for the localization of high channel density, i.e., for tumor diagnosis. In particular, the physicochemical properties of the fluorescent probes should be improved compared to existing probes. Methods: The small molecule inhibitor of the KCa3.1 channel, senicapoc, was used as a warhead and was coupled with different fluorescent dyes. After synthesis of the novel probes, their physicochemical properties (lipophilicity, photophysical properties) and their ability to image KCa3.1 channels in A549-3R lung tumor cells were determined. Results: In order to increase the polarity and quantum yield of reported fluorescent probes, three strategies were followed: (1) An F-atom at the B-atom of bodipy-labeled senicapoc derivatives 9a, 9b, and 15a was replaced by a OCH3 moiety, which decreased the logP value by one log-unit. (2) The p-phenylene moiety of the linker was replaced by an aliphatic tetramethylene linker decreasing the lipophilicity by 0.3-0.5 log-units. (3) Instead of bodipy dyes, fluorescein was coupled with the senicapoc warhead resulting in very polar probes 21a and 21b with low logP values of 1.5 and 1.3, respectively. Introduction of an ethyl moiety at the bodipy core increased the quantum yield, which resulted in the best punctate staining pattern of fixed and living A549-3R lung tumor cells with the ethylbodipy-labeled senicapoc derivative 10b. The specificity was shown by various control experiments. Co-staining with 10b and an antibody did not result in overlapping signals. Conclusions: The well-balanced lipophilicity and fluorescent quantum yield render the ethylbodipy-labeled senicapoc derivative 10b a very good probe to image selectively KCa3.1 ion channels in fixed and living tumor cells. It was hypothesized that the antibody binds selectively at the closed channel (58.5%), whereas the senicapoc-bodipy conjugate 10b binds selectively at the open channel (41.5%). The ratio 58.5:41.5 reflects the ratio of the ion channel in closed and open conformations.
Collapse
Affiliation(s)
- Insa Thale
- Chemical Biology of Ion Channels (Chembion), University of Münster, Corrensstraße 48, D-48149 Münster, Germany; (I.T.); (L.V.); (L.M.T.); (T.B.); (A.S.)
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Elke Naß
- Institute of Physiology I, University of Münster, University Hospital Münster, Robert-Koch-Straße 27a, D-48149 Münster, Germany;
| | - Laura Vinnenberg
- Chemical Biology of Ion Channels (Chembion), University of Münster, Corrensstraße 48, D-48149 Münster, Germany; (I.T.); (L.V.); (L.M.T.); (T.B.); (A.S.)
- Institute of Physiology I, University of Münster, University Hospital Münster, Robert-Koch-Straße 27a, D-48149 Münster, Germany;
| | - Luca Matteo Todesca
- Chemical Biology of Ion Channels (Chembion), University of Münster, Corrensstraße 48, D-48149 Münster, Germany; (I.T.); (L.V.); (L.M.T.); (T.B.); (A.S.)
- Institute of Physiology II, University of Münster, University Hospital Münster, Robert-Koch-Straße 27b, D-48149 Münster, Germany
- Department of Biology, University of Padua, Via U.Bassi 58/B, 35131 Padova, Italy
| | - Thomas Budde
- Chemical Biology of Ion Channels (Chembion), University of Münster, Corrensstraße 48, D-48149 Münster, Germany; (I.T.); (L.V.); (L.M.T.); (T.B.); (A.S.)
- Institute of Physiology I, University of Münster, University Hospital Münster, Robert-Koch-Straße 27a, D-48149 Münster, Germany;
| | - Ivan Maisuls
- Institute of Inorganic and Analytical Chemistry, University of Münster, CiMIC, SoN, Corrensstraße 28, D-48149 Münster, Germany; (I.M.); (C.A.S.)
- CeNTech, University of Münster, Heisenbergstraße 11, D-48149 Münster, Germany
| | - Cristian A. Strassert
- Institute of Inorganic and Analytical Chemistry, University of Münster, CiMIC, SoN, Corrensstraße 28, D-48149 Münster, Germany; (I.M.); (C.A.S.)
- CeNTech, University of Münster, Heisenbergstraße 11, D-48149 Münster, Germany
| | - Albrecht Schwab
- Chemical Biology of Ion Channels (Chembion), University of Münster, Corrensstraße 48, D-48149 Münster, Germany; (I.T.); (L.V.); (L.M.T.); (T.B.); (A.S.)
- Institute of Physiology II, University of Münster, University Hospital Münster, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Chemical Biology of Ion Channels (Chembion), University of Münster, Corrensstraße 48, D-48149 Münster, Germany; (I.T.); (L.V.); (L.M.T.); (T.B.); (A.S.)
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
10
|
Szollosi A. Functional Characterization of Ion Channels in Planar Lipid Bilayers. Methods Mol Biol 2025; 2908:141-161. [PMID: 40304908 DOI: 10.1007/978-1-0716-4434-8_10] [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: 05/02/2025]
Abstract
Ion channels form transmembrane protein pores allowing ion fluxes across biological membranes. Their function is indispensable for normal homeostasis; therefore, channelopathies lead to a large variety of diseases affecting almost all tissues and organs. In the last decades, the structure of hundreds of ion channels has been solved. In the majority of the solved structures the pore is closed, and for most of the protein preparations used in these studies, currently little data is available to support that the purified protein forms a functional ion channel capable of gating. Planar lipid bilayer technique is a powerful tool to address this issue. Purified ion channel protein is incorporated into a membrane bilayer separating two compartments in an experimental chamber. Channel currents are recorded between the two sides. Such setup allows characterization of gating and permeation properties of ion channels unbiased by auxiliary components present in native membranes. The planar lipid bilayer technique is thus discussed herein together with the robust protein expressing BacMam system.
Collapse
Affiliation(s)
- Andras Szollosi
- Semmelweis University, Department of Biochemistry, Budapest, Hungary.
- HUN-REN-SE Ion Channel Research Group, Budapest, Hungary.
- HCEMM-SE Molecular Channelopathies Research Group, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
11
|
Saljic A, Heijman J, Dobrev D. From Atrial Small-conductance Calcium-activated Potassium Channels to New Antiarrhythmics. Eur Cardiol 2024; 19:e26. [PMID: 39872420 PMCID: PMC11770539 DOI: 10.15420/ecr.2024.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/27/2024] [Indexed: 01/30/2025] Open
Abstract
Despite significant advances in its management, AF remains a major healthcare burden affecting millions of individuals. Rhythm control with antiarrhythmic drugs or catheter ablation has been shown to improve symptoms and outcomes in AF patients, but current treatment options have limited efficacy and/or significant side-effects. Novel mechanism-based approaches could potentially be more effective, enabling improved therapeutic strategies for managing AF. Small-conductance calcium-activated potassium (SK or KCa2.x) channels encoded by KCNN1-3 have recently gathered interest as novel antiarrhythmic targets with potential atrial-predominant effects. Here, the molecular composition of smallconductance calcium-activated potassium channels and their complex regulation in AF as the basis for understanding the distinct mechanism of action of pore-blockers (apamin, UCL1684, ICAGEN) and modulators of calcium-dependent activation (NS8593, AP14145, AP30663) are summarised. Furthermore, the preclinical and early clinical evidence for the role of small-conductance calcium-activated potassium channel inhibitors in the treatment of AF are reviewed.
Collapse
Affiliation(s)
- Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagen, Denmark
- Institute of Pharmacology, West German Heart and Vascular Center, University of Duisburg-EssenEssen, Germany
| | - Jordi Heijman
- Gottfried Schatz Research Centre, Division of Medical Physics & Biophysics, Medical University of GrazGraz, Austria
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht UniversityMaastricht, the Netherlands
| | - Dobromir Dobrev
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagen, Denmark
- Departments of Medicine and Research Centre, Montreal Heart Institute and Université de MontréalMontreal, Canada
- Department of Integrative Physiology, Baylor College of MedicineHouston, TX, US
| |
Collapse
|
12
|
Mkrtchyan L, Sahakyan H, Eldstrom J, Karapetyan T, Abrahamyan A, Nazaryan K, Schwarz JR, Kneussel M, Fedida D, Vardanyan V. Ion permeation through a narrow cavity constriction in KCNQ1 channels: Mechanism and implications for pathogenic variants. Proc Natl Acad Sci U S A 2024; 121:e2411182121. [PMID: 39671184 DOI: 10.1073/pnas.2411182121] [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: 06/04/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024] Open
Abstract
KCNQ1 potassium channels play a pivotal role in the physiology and pathophysiology of several human excitable and epithelial tissues. The latest cryo-electron microscopy (cryo-EM) structures provide unique insights into channel function and pharmacology, opening avenues for different therapeutic strategies against human diseases associated with KCNQ1 mutations. However, these structures also raise fundamental questions about the mechanisms of ion permeation. Cryo-EM structures thought to represent the open state of the channel feature a cavity region not wide enough for accommodation of hydrated K+. To understand how K+ passes through the cavity constriction, we utilized microsecond-scale molecular dynamics (MD) simulations using the KCNQ1/KCNE3 cryo-EM structure, characterized mutants at the G345 residue situated at the narrowest point of the cavity, and recorded single channels. The findings indicate that ions become partially dehydrated at the constriction, which enables permeation. MD simulations demonstrate that the constriction can impede the flow of ions through the channel's pore, a finding that is corroborated by mutational screening and single-channel recordings. Reduced channel conductance is the key mechanism underlying reported pathological KCNQ1 mutations at or near the constriction site.
Collapse
Affiliation(s)
- Liana Mkrtchyan
- Molecular Neuroscience Group, Institute of Molecular Biology, Yerevan 0014, Armenia
| | - Harutyun Sahakyan
- Laboratory of Computational Modeling of Biological Processes, Institute of Molecular Biology, Yerevan 0014, Armenia
| | - Jodene Eldstrom
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Tatev Karapetyan
- Molecular Neuroscience Group, Institute of Molecular Biology, Yerevan 0014, Armenia
| | - Astghik Abrahamyan
- Molecular Neuroscience Group, Institute of Molecular Biology, Yerevan 0014, Armenia
| | - Karen Nazaryan
- Laboratory of Computational Modeling of Biological Processes, Institute of Molecular Biology, Yerevan 0014, Armenia
| | - Jürgen R Schwarz
- Institute for Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Matthias Kneussel
- Institute for Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - David Fedida
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Vitya Vardanyan
- Molecular Neuroscience Group, Institute of Molecular Biology, Yerevan 0014, Armenia
| |
Collapse
|
13
|
Sanders JH, Taiwo KM, Adekanye GA, Bali A, Zhang Y, Paulsen CE. Calmodulin binding is required for calcium mediated TRPA1 desensitization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627969. [PMID: 39713425 PMCID: PMC11661184 DOI: 10.1101/2024.12.11.627969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Calcium (Ca2+) ions affect nearly all aspects of biology. Excessive Ca2+ entry is cytotoxic and Ca2+-mobilizing receptors have evolved diverse mechanisms for tight regulation that often include Calmodulin (CaM). TRPA1, an essential Ca2+-permeable ion channel involved in pain signaling and inflammation, exhibits complex Ca2+ regulation with initial channel potentiation followed by rapid desensitization. The molecular mechanisms of TRPA1 Ca2+ regulation and whether CaM plays a role remain elusive. We find that TRPA1 binds CaM best at basal Ca2+ concentration, that they co-localize in resting cells, and that CaM suppresses TRPA1 activity. Combining biochemical, biophysical, modeling, NMR spectroscopy, and functional approaches, we identify an evolutionarily conserved, high-affinity CaM binding element in the distal TRPA1 C-terminus (DCTCaMBE). Genetic or biochemical perturbation of Ca2+/CaM binding to the TRPA1 DCTCaMBE yields hyperactive channels that exhibit drastic slowing of desensitization with no effect on potentiation. Ca2+/CaM TRPA1 regulation does not require the N-lobe, raising the possibility that CaM is not the Ca2+ sensor, per se. Higher extracellular Ca2+ can partially rescue slowed desensitization suggesting Ca2+/CaM binding to the TRPA1 DCTCaMBE primes an intrinsic TRPA1 Ca2+ binding site that, upon binding Ca2+, triggers rapid desensitization. Collectively, our results identify a critical regulatory element in an unstructured TRPA1 region highlighting the importance of these domains, they reveal Ca2+/CaM is an essential TRPA1 auxiliary subunit required for rapid desensitization that establishes proper channel function with implications for all future TRPA1 work, and they uncover a mechanism for receptor regulation by Ca2+/CaM that expands the scope of CaM biology.
Collapse
Affiliation(s)
- Justin H. Sanders
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Kehinde M. Taiwo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Glory A. Adekanye
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Avnika Bali
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Yuekang Zhang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Candice E. Paulsen
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Wong BHS, Shim H, Goay SSM, Ong ST, Muhammad Taib NAB, Chai KXY, Lim K, Huang D, Ong CK, Vaiyapuri TS, Cheah YC, Wang Y, Wulff H, Webster RD, Shelat VG, Verma NK. The novel quinoline derivative SKA-346 as a K Ca3.1 channel selective activator. RSC Adv 2024; 14:38364-38377. [PMID: 39635364 PMCID: PMC11615718 DOI: 10.1039/d4ra07330d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/17/2024] [Indexed: 12/07/2024] Open
Abstract
The calcium-activated KCa3.1 channel plays a crucial role in T-cell immune response. Genetic manipulation of T-cells to upregulate the expression of K+ channels has been shown to boost T-cell cytotoxicity in cancer. Here, we aimed to identify and characterize an activator that would augment KCa3.1 currents without affecting other channels. We synthesized five quinoline derivatives and used electrophysiology to screen them on KCa3.1 and a panel of 14 other ion channels. One quinoline derivative, SKA-346, activated KCa3.1 with an EC50 of 1.9 μM and showed selectivity against the other channels. In silico analysis using RosettaLigand and GLIDE demonstrated a well-converged pose of SKA-346 in a binding pocket at the interface between the calmodulin N-lobe and the S45A helix in the S4-S5 linker of the KCa3.1 channel. SKA-346 (30 mg kg-1), tolerated by mice after intra-peritoneal administration, exhibited a peak plasma concentration of 6.29 μg mL-1 (29.2 μM) at 15 min and a circulating half-life (t 1/2) of 2.8 h. SKA-346 could serve as a template for the development of more potent KCa3.1 activators to enhance T-cell cytotoxicity in cancer.
Collapse
Affiliation(s)
- Brandon Han Siang Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- Interdisciplinary Graduate Programme, NTU Institute for Health Technologies (HealthTech NTU), Nanyang Technological University Singapore Singapore
| | - Heesung Shim
- Physical and Life Sciences, Lawrence Livermore National Laboratory Livermore CA USA
| | - Stephanie Shee Min Goay
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- LKCMedicine-ICE Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Seow Theng Ong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- LKCMedicine-ICE Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Nur Ayuni Binte Muhammad Taib
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Kelila Xin Ye Chai
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Kerry Lim
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Dachuan Huang
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
- Duke-NUS Medical School Singapore
| | - Choon Kiat Ong
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
- Duke-NUS Medical School Singapore
| | | | - Yeong Cheng Cheah
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Heike Wulff
- Department of Pharmacology, University of California Davis CA USA
| | - Richard D Webster
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University Singapore Singapore
| | - Vishalkumar G Shelat
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- Department of General Surgery, Tan Tock Seng Hospital Singapore
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| |
Collapse
|
15
|
Karimi R, Coupland CE, Rubinstein JL. Vesicle Picker: A tool for efficient identification of membrane protein complexes in vesicles. J Struct Biol 2024; 216:108148. [PMID: 39481498 DOI: 10.1016/j.jsb.2024.108148] [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] [Received: 07/15/2024] [Revised: 09/15/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Electron cryomicroscopy (cryo-EM) has recently allowed determination of near-atomic resolution structures of membrane proteins and protein complexes embedded in lipid vesicles. However, particle selection from electron micrographs of these vesicles can be challenging due to the strong signal contributed from the lipid bilayer. This challenge often requires iterative and laborious particle selection workflows to generate a dataset of high-quality particle images for subsequent analysis. Here we present Vesicle Picker, an open-source program built on the Segment Anything model. Vesicle Picker enables automatic identification of vesicles in cryo-EM micrographs with high recall and precision. It then exhaustively selects all potential particle locations, either at the perimeter or uniformly over the surface of the projection of the vesicle. The program is designed to interface with cryoSPARC, which performs both upstream micrograph processing and downstream single particle image analysis. We demonstrate Vesicle Picker's utility by determining a high-resolution map of the vacuolar-type ATPase from micrographs of native synaptic vesicles (SVs) and identifying an additional protein or protein complex in the SV membrane.
Collapse
Affiliation(s)
- Ryan Karimi
- Molecular Medicine Program, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Department of Medical Biophysics, The University of Toronto, Toronto M5G 1L7, Canada
| | - Claire E Coupland
- Molecular Medicine Program, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - John L Rubinstein
- Molecular Medicine Program, The Hospital for Sick Children, Toronto M5G 0A4, Canada; Department of Medical Biophysics, The University of Toronto, Toronto M5G 1L7, Canada; Department of Biochemistry, The University of Toronto, Toronto M5S 1A8, Canada.
| |
Collapse
|
16
|
Sharma P, Guo A, Poudel S, Boada-Romero E, Verbist KC, Palacios G, Immadisetty K, Chen MJ, Haydar D, Mishra A, Peng J, Babu MM, Krenciute G, Glazer ES, Green DR. An early, novel arginine methylation of KCa3.1 attenuates subsequent T cell exhaustion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593421. [PMID: 38798680 PMCID: PMC11118966 DOI: 10.1101/2024.05.09.593421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
T cell receptor (TCR) engagement initiates the activation process, and this signaling event is regulated in multifaceted ways. Nutrient availability in the immediate niche is one such mode of regulation 1-3 . Here, we investigated how the availability of an essential amino acid methionine (Met) and TCR signaling might interplay in the earliest events of T cell activation to affect subsequent T cell fate and function. We found that limiting Met during only the initial 30 minutes of CD8 + T cell activation increased Ca 2+ influx, Ca 2+ -mediated NFAT1 ( Nfatc2 ) activation, NFAT1 promoter occupancy, and T cell exhaustion. We identified changes in the protein arginine methylome during the initial 30 min of TCR engagement and discovered a novel arginine methylation of a Ca 2+ -activated potassium transporter, KCa3.1, which regulates Ca 2+ -mediated NFAT1 signaling to ensure optimal activation. Ablation of arginine methylation in KCa3.1 led to increased NFAT1 activation, rendering T cells dysfunctional in murine tumour and infection models. Furthermore, acute Met supplementation at early stages reduced nuclear NFAT1 in tumour-infiltrating T cells and augmented their anti-tumour activity. Our findings identify a metabolic event occurring early after T cell activation that influences the subsequent fate of the cell.
Collapse
|
17
|
Smorodina E, Tao F, Qing R, Yang S, Zhang S. Computational engineering of water-soluble human potassium ion channels through QTY transformation. Sci Rep 2024; 14:28159. [PMID: 39548172 PMCID: PMC11568286 DOI: 10.1038/s41598-024-76603-7] [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] [Received: 07/10/2023] [Accepted: 10/14/2024] [Indexed: 11/17/2024] Open
Abstract
Transmembrane potassium ion channels are crucial for ion transport, metabolism, and signaling, and serve as promising targets for anti-cancer therapies. However, their hydrophobic transmembrane nature requires detergents, posing a major bottleneck for experimental handling. In this paper, we present a structural bioinformatics study of six experimentally determined and twelve modeled potassium channel structures, in which hydrophobic amino acids (L, I/V, and F) were systematically replaced with neutral hydrophilic ones (Q, T, and Y), making the proteins more water-soluble. QTY (computationally predicted) and native (experimental and repredicted) variants show remarkable structural similarity (RMSD: ~0.50 Å - ~2.14 Å) despite significant sequence differences. QTY variants, both rigid and refined with MD simulations, maintain comparable to native variants stability, solvent-accessible surface area (SASA), and ionic, aromatic, and van der Waals interactions but differ in the grand average of hydropathy (GRAVY), solubility, and hydrophobic contacts. Overall, our study presents a computational approach for designing hydrophilic potassium ion channels while maintaining the native global structure that could potentially simplify their practical use by eliminating the need for detergents.
Collapse
Affiliation(s)
- Eva Smorodina
- Laboratory for Computational and Systems Immunology, Department of Immunology, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - Fei Tao
- Laboratory of Food Microbial Technology, State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiaotong University, Shanghai, 200240, China
| | - Rui Qing
- Laboratory of Food Microbial Technology, State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiaotong University, Shanghai, 200240, China
| | - Steve Yang
- PT Metiska Farma, Daerah Khusus Ibukota, Jakarta, 12220, Indonesia
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
18
|
Woltz RL, Zheng Y, Choi W, Ngo K, Trinh P, Ren L, Thai PN, Harris BJ, Han Y, Rouen KC, Mateos DL, Jian Z, Chen-Izu Y, Dickson EJ, Yamoah EN, Yarov-Yarovoy V, Vorobyov I, Zhang XD, Chiamvimonvat N. Atomistic mechanisms of the regulation of small-conductance Ca 2+-activated K + channel (SK2) by PIP2. Proc Natl Acad Sci U S A 2024; 121:e2318900121. [PMID: 39288178 PMCID: PMC11441529 DOI: 10.1073/pnas.2318900121] [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: 10/31/2023] [Accepted: 06/24/2024] [Indexed: 09/19/2024] Open
Abstract
Small-conductance Ca2+-activated K+ channels (SK, KCa2) are gated solely by intracellular microdomain Ca2+. The channel has emerged as a therapeutic target for cardiac arrhythmias. Calmodulin (CaM) interacts with the CaM binding domain (CaMBD) of the SK channels, serving as the obligatory Ca2+ sensor to gate the channels. In heterologous expression systems, phosphatidylinositol 4,5-bisphosphate (PIP2) coordinates with CaM in regulating SK channels. However, the roles and mechanisms of PIP2 in regulating SK channels in cardiomyocytes remain unknown. Here, optogenetics, magnetic nanoparticles, combined with Rosetta structural modeling, and molecular dynamics (MD) simulations revealed the atomistic mechanisms of how PIP2 works in concert with Ca2+-CaM in the SK channel activation. Our computational study affords evidence for the critical role of the amino acid residue R395 in the S6 transmembrane segment, which is localized in propinquity to the intracellular hydrophobic gate. This residue forms a salt bridge with residue E398 in the S6 transmembrane segment from the adjacent subunit. Both R395 and E398 are conserved in all known isoforms of SK channels. Our findings suggest that the binding of PIP2 to R395 residue disrupts the R395:E398 salt bridge, increasing the flexibility of the transmembrane segment S6 and the activation of the channel. Importantly, our findings serve as a platform for testing of structural-based drug designs for therapeutic inhibitors and activators of the SK channel family. The study is timely since inhibitors of SK channels are currently in clinical trials to treat atrial arrhythmias.
Collapse
Grants
- OT2 OD026580 NIH HHS
- T32 HL086350 NHLBI NIH HHS
- NIH R01 DC016099 HHS | NIH | National Institute on Deafness and Other Communication Disorders (NIDCD)
- I01 CX001490 CSRD VA
- T32 GM136597 NIGMS NIH HHS
- R01 DC016099 NIDCD NIH HHS
- NIH F32 HL151130 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Anton 2 allocation MCB210014P Pittsburgh Supercomputing Center
- NIH T32 HL86350 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL158961 NHLBI NIH HHS
- R01 HL137228 NHLBI NIH HHS
- T32 GM007377 NIGMS NIH HHS
- R01 HL174001 NHLBI NIH HHS
- F32 HL151130 NHLBI NIH HHS
- R01 HL128537 NHLBI NIH HHS
- NIH R01 HL085727 NIH R01 HL085844 NIH R01 HL137228 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL152681 NHLBI NIH HHS
- R01 HL085727 NHLBI NIH HHS
- R01 GM116961 NIGMS NIH HHS
- NIH R01 HL152681 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 AG060504 NIA NIH HHS
- R35 GM149211 NIGMS NIH HHS
- I01 BX000576 BLRD VA
- NIH R01 AG060504 and NIH 2P01 AG051443 HHS | NIH | National Institute on Aging (NIA)
- R01 HL085844 NHLBI NIH HHS
- NIH R01 HL158961 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH R35 GM149211 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- P01 AG051443 NIA NIH HHS
- NIH R01 HL128537 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R56 HL138392 NHLBI NIH HHS
Collapse
Affiliation(s)
- Ryan L. Woltz
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616
| | - Yang Zheng
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616
| | - Woori Choi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616
| | - Khoa Ngo
- Department of Physiology and Membrane Biology, University of California, Davis, CA95616
| | - Pauline Trinh
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616
| | - Lu Ren
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA94305
| | - Phung N. Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616
| | - Brandon J. Harris
- Department of Physiology and Membrane Biology, University of California, Davis, CA95616
| | - Yanxiao Han
- Department of Physiology and Membrane Biology, University of California, Davis, CA95616
| | - Kyle C. Rouen
- Department of Physiology and Membrane Biology, University of California, Davis, CA95616
| | - Diego Lopez Mateos
- Department of Physiology and Membrane Biology, University of California, Davis, CA95616
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, CA95616
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA95616
| | - Eamonn J. Dickson
- Department of Physiology and Membrane Biology, University of California, Davis, CA95616
| | - Ebenezer N. Yamoah
- Department of Translational Neuroscience, University of Arizona College of Medicine, Phoenix, AZ85004
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, CA95616
- Department of Anesthesiology and Pain Medicine, University of California, Davis, CA95616
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California, Davis, CA95616
- Department of Pharmacology, University of California, Davis, CA95616
| | - Xiao-Dong Zhang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA 95616
- Department of Pharmacology, University of California, Davis, CA95616
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA95655
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ85004
| |
Collapse
|
19
|
Gary CR, Acharige NPN, Oyewumi TO, Pflum MKH. Kinase-catalyzed biotinylation for discovery and validation of substrates to multispecificity kinases NME1 and NME2. J Biol Chem 2024; 300:107588. [PMID: 39032654 PMCID: PMC11375270 DOI: 10.1016/j.jbc.2024.107588] [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: 09/18/2023] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024] Open
Abstract
Protein phosphorylation by kinases regulates mammalian cell functions, such as growth, division, and signal transduction. Among human kinases, NME1 and NME2 are associated with metastatic tumor suppression but remain understudied due to the lack of tools to monitor their cellular substrates. In particular, NME1 and NME2 are multispecificity kinases phosphorylating serine, threonine, histidine, and aspartic acid residues of substrate proteins, and the heat and acid sensitivity of phosphohistidine and phosphoaspartate complicates substrate discovery and validation. To provide new substrate monitoring tools, we established the γ-phosphate-modified ATP analog, ATP-biotin, as a cosubstrate for phosphorylbiotinylation of NME1 and NME2 cellular substrates. Building upon this ATP-biotin compatibility, the Kinase-catalyzed Biotinylation with Inactivated Lysates for Discovery of Substrates method enabled validation of a known substrate and the discovery of seven NME1 and three NME2 substrates. Given the paucity of methods to study kinase substrates, ATP-biotin and the Kinase-catalyzed Biotinylation with Inactivated Lysates for Discovery of Substrates method are valuable tools to characterize the roles of NME1 and NME2 in human cell biology.
Collapse
Affiliation(s)
- Chelsea R Gary
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | | | | | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA.
| |
Collapse
|
20
|
Zhao Y, Zhang W, Hong J, Yang L, Wang Y, Qu F, Xu W. Mobility capillary electrophoresis-native mass spectrometry reveals the dynamic conformational equilibrium of calmodulin and its complexes. Analyst 2024; 149:3793-3802. [PMID: 38847183 DOI: 10.1039/d4an00378k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Benefitting from the rapid evolution of artificial intelligence and structural biology, an expanding collection of high-resolution protein structures has greatly improved our understanding of protein functions. Yet, proteins are inherently flexible, and these static structures can only offer limited snapshots of their true dynamic nature. The conformational and functional changes of calmodulin (CaM) induced by Ca2+ binding have always been a focus of research. In this study, the conformational dynamics of CaM and its complexes were investigated using a mobility capillary electrophoresis (MCE) and native mass spectrometry (native MS) based method. By analyzing the ellipsoidal geometries of CaM in the solution phase at different Ca2+ concentrations, it is interesting to discover that CaM molecules, whether bound to Ca2+ or not, possess both closed and open conformations. Moreover, each individual CaM molecule actively "jumps" (equilibrium exchange) between these two distinct conformations on a timescale ranging from milli- to micro-seconds. The binding of Ca2+ ions did not affect the structural dynamics of CaM, while the binding of a peptide ligand would stabilize CaM, leading to the observation of a single, compact conformation of the resulting protein complex. A target recognition mechanism was also proposed based on these new findings, suggesting that CaM's interaction with targets may favor a conformational selection model. This enriches our understanding of the binding principles between CaM and its numerous targets.
Collapse
Affiliation(s)
- Yi Zhao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Wenjing Zhang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Jie Hong
- Kunshan Nier Precision Instrumentation Inc. Kunshan, Suzhou, 215316, China
| | - Lei Yang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Yuanyuan Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Feng Qu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Wei Xu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
21
|
Trofimov YA, Krylov NA, Minakov AS, Nadezhdin KD, Neuberger A, Sobolevsky AI, Efremov RG. Dynamic molecular portraits of ion-conducting pores characterize functional states of TRPV channels. Commun Chem 2024; 7:119. [PMID: 38824263 PMCID: PMC11144267 DOI: 10.1038/s42004-024-01198-z] [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: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024] Open
Abstract
Structural biology is solving an ever-increasing number of snapshots of ion channel conformational ensembles. Deciphering ion channel mechanisms, however, requires understanding the ensemble dynamics beyond the static structures. Here, we present a molecular modeling-based approach characterizing the ion channel structural intermediates, or their "dynamic molecular portraits", by assessing water and ion conductivity along with the detailed evaluation of pore hydrophobicity and residue packing. We illustrate the power of this approach by analyzing structures of few vanilloid-subfamily transient receptor potential (TRPV) channels. Based on the pore architecture, there are three major states that are common for TRPVs, which we call α-closed, π-closed, and π-open. We show that the pore hydrophobicity and residue packing for the open state is most favorable for the pore conductance. On the contrary, the α-closed state is the most hydrophobic and always non-conducting. Our approach can also be used for structural and functional classification of ion channels.
Collapse
Affiliation(s)
- Yury A Trofimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay A Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
22
|
Baltasar-Marchueta M, Llona L, M-Alicante S, Barbolla I, Ibarluzea MG, Ramis R, Salomon AM, Fundora B, Araujo A, Muguruza-Montero A, Nuñez E, Pérez-Olea S, Villanueva C, Leonardo A, Arrasate S, Sotomayor N, Villarroel A, Bergara A, Lete E, González-Díaz H. Identification of Riluzole derivatives as novel calmodulin inhibitors with neuroprotective activity by a joint synthesis, biosensor, and computational guided strategy. Biomed Pharmacother 2024; 174:116602. [PMID: 38636396 DOI: 10.1016/j.biopha.2024.116602] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024] Open
Abstract
The development of new molecules for the treatment of calmodulin related cardiovascular or neurodegenerative diseases is an interesting goal. In this work, we introduce a novel strategy with four main steps: (1) chemical synthesis of target molecules, (2) Förster Resonance Energy Transfer (FRET) biosensor development and in vitro biological assay of new derivatives, (3) Cheminformatics models development and in vivo activity prediction, and (4) Docking studies. This strategy is illustrated with a case study. Firstly, a series of 4-substituted Riluzole derivatives 1-3 were synthetized through a strategy that involves the construction of the 4-bromoriluzole framework and its further functionalization via palladium catalysis or organolithium chemistry. Next, a FRET biosensor for monitoring Ca2+-dependent CaM-ligands interactions has been developed and used for the in vitro assay of Riluzole derivatives. In particular, the best inhibition (80%) was observed for 4-methoxyphenylriluzole 2b. Besides, we trained and validated a new Networks Invariant, Information Fusion, Perturbation Theory, and Machine Learning (NIFPTML) model for predicting probability profiles of in vivo biological activity parameters in different regions of the brain. Next, we used this model to predict the in vivo activity of the compounds experimentally studied in vitro. Last, docking study conducted on Riluzole and its derivatives has provided valuable insights into their binding conformations with the target protein, involving calmodulin and the SK4 channel. This new combined strategy may be useful to reduce assay costs (animals, materials, time, and human resources) in the drug discovery process of calmodulin inhibitors.
Collapse
Affiliation(s)
- Maider Baltasar-Marchueta
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | - Leire Llona
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | | | - Iratxe Barbolla
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | - Markel Garcia Ibarluzea
- Donostia International Physics Center, Donostia, Spain; Departament of Physics, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Rafael Ramis
- Donostia International Physics Center, Donostia, Spain; Departament of Physics, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Ane Miren Salomon
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | - Brenda Fundora
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | - Ariane Araujo
- Biofisika Institute, CSIC-UPV/EHU, Leioa 48940, Spain
| | | | - Eider Nuñez
- Biofisika Institute, CSIC-UPV/EHU, Leioa 48940, Spain
| | - Scarlett Pérez-Olea
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | - Christian Villanueva
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | - Aritz Leonardo
- Donostia International Physics Center, Donostia, Spain; Departament of Physics, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Sonia Arrasate
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | - Nuria Sotomayor
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain
| | | | - Aitor Bergara
- Donostia International Physics Center, Donostia, Spain; Departament of Physics, University of the Basque Country, UPV/EHU, Leioa, Spain.
| | - Esther Lete
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain.
| | - Humberto González-Díaz
- Department of Organic and Inorganic Chemistry, University of the Basque Country UPV/EHU, Leioa 48940, Spain; Biofisika Institute, CSIC-UPV/EHU, Leioa 48940, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48011, Spain.
| |
Collapse
|
23
|
Segura É, Zhao J, Broszczak M, Audet F, Sauvé R, Parent L. Investigating the Impact of Electrostatic Interactions on Calmodulin Binding and Ca 2+-Dependent Activation of the Calcium-Gated Potassium SK4 Channel. Int J Mol Sci 2024; 25:4255. [PMID: 38673845 PMCID: PMC11050286 DOI: 10.3390/ijms25084255] [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: 03/08/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Ca2+ binding to the ubiquitous Ca2+ sensing protein calmodulin (CaM) activates the intermediate conductance Ca2+-activated SK4 channel. Potential hydrophilic pockets for CaM binding have been identified at the intracellular HA and HB helices in the C-terminal of SK4 from the three published cryo-EM structures of SK4. Single charge reversal substitutions at either site, significantly weakened the pull-down of SK4 by CaM wild-type (CaM), and decreased the TRAM-34 sensitive outward K+ current densities in native HEK293T cells when compared with SK4 WT measured under the same conditions. Only the doubly substituted SK4 R352D/R355D (HB helix) obliterated the CaM-mediated pull-down and thwarted outward K+ currents. However, overexpression of CaM E84K/E87K, which had been predicted to face the arginine doublet, restored the CaM-mediated pull-down of SK4 R352D/R355D and normalized its whole-cell current density. Virtual analysis of the putative salt bridges supports a unique role for the positively charged arginine doublet at the HB helix into anchoring the interaction with the negatively charged CaM glutamate 84 and 87 CaM. Our findings underscore the unique contribution of electrostatic interactions in carrying CaM binding onto SK4 and support the role of the C-terminal HB helix to the Ca2+-dependent gating process.
Collapse
Affiliation(s)
- Émilie Segura
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (É.S.); (F.A.)
| | - Juan Zhao
- Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (J.Z.); (M.B.)
| | - Marlena Broszczak
- Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (J.Z.); (M.B.)
| | - Frédéric Audet
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (É.S.); (F.A.)
| | - Rémy Sauvé
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, 2900 Bd Édouard-Montpetit, Montréal, QC H3T 1J4, Canada;
| | - Lucie Parent
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (É.S.); (F.A.)
| |
Collapse
|
24
|
Cozzolino M, Panyi G. Intracellular acidity impedes KCa3.1 activation by Riluzole and SKA-31. Front Pharmacol 2024; 15:1380655. [PMID: 38638868 PMCID: PMC11024243 DOI: 10.3389/fphar.2024.1380655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 04/20/2024] Open
Abstract
Background The unique microenvironment in tumors inhibits the normal functioning of tumor-infiltrating lymphocytes, leading to immune evasion and cancer progression. Over-activation of KCa3.1 using positive modulators has been proposed to rescue the anti-tumor response. One of the key characteristics of the tumor microenvironment is extracellular acidity. Herein, we analyzed how intra- and extracellular pH affects K+ currents through KCa3.1 and if the potency of two of its positive modulators, Riluzole and SKA-31, is pH sensitive. Methods Whole-cell patch-clamp was used to measure KCa3.1 currents either in activated human peripheral lymphocytes or in CHO cells transiently transfected with either the H192A mutant or wild-type hKCa3.1 in combination with T79D-Calmodulin, or with KCa2.2. Results We found that changes in the intra- and extracellular pH minimally influenced the KCa3.1-mediated K+ current. Extracellular pH, in the range of 6.0-8.0, does not interfere with the capacity of Riluzole and SKA-31 to robustly activate the K+ currents through KCa3.1. Contrariwise, an acidic intracellular solution causes a slow, but irreversible loss of potency of both the activators. Using different protocols of perfusion and depolarization we demonstrated that the loss of potency is strictly time and pH-dependent and that this peculiar effect can be observed with a structurally similar channel KCa2.2. While two different point mutations of both KCa3.1 (H192A) and its associated protein Calmodulin (T79D) do not limit the effect of acidity, increasing the cytosolic Ca2+ concentration to saturating levels eliminated the loss-of-potency phenotype. Conclusion Based on our data we conclude that KCa3.1 currents are not sensitive the either the intracellular or the extracellular pH in the physiological and pathophysiological range. However, intracellular acidosis in T cells residing in the tumor microenvironment could hinder the potentiating effect of KCa3.1 positive modulators administered to boost their activity. Further research is warranted both to clarify the molecular interactions between the modulators and KCa3.1 at different intracellular pH conditions and to define whether this loss of potency can be observed in cancer models as well.
Collapse
Affiliation(s)
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
25
|
Van NTH, Kim WK, Nam JH. Challenges in the Therapeutic Targeting of KCa Channels: From Basic Physiology to Clinical Applications. Int J Mol Sci 2024; 25:2965. [PMID: 38474212 PMCID: PMC10932353 DOI: 10.3390/ijms25052965] [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: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 03/14/2024] Open
Abstract
Calcium-activated potassium (KCa) channels are ubiquitously expressed throughout the body and are able to regulate membrane potential and intracellular calcium concentrations, thereby playing key roles in cellular physiology and signal transmission. Consequently, it is unsurprising that KCa channels have been implicated in various diseases, making them potential targets for pharmaceutical interventions. Over the past two decades, numerous studies have been conducted to develop KCa channel-targeting drugs, including those for disorders of the central and peripheral nervous, cardiovascular, and urinary systems and for cancer. In this review, we synthesize recent findings regarding the structure and activating mechanisms of KCa channels. We also discuss the role of KCa channel modulators in therapeutic medicine. Finally, we identify the major reasons behind the delay in bringing these modulators to the pharmaceutical market and propose new strategies to promote their application.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| |
Collapse
|
26
|
Babini H, Jiménez-Sábado V, Stogova E, Arslanova A, Butt M, Dababneh S, Asghari P, Moore EDW, Claydon TW, Chiamvimonvat N, Hove-Madsen L, Tibbits GF. hiPSC-derived cardiomyocytes as a model to study the role of small-conductance Ca 2+-activated K + (SK) ion channel variants associated with atrial fibrillation. Front Cell Dev Biol 2024; 12:1298007. [PMID: 38304423 PMCID: PMC10830749 DOI: 10.3389/fcell.2024.1298007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
Atrial fibrillation (AF), the most common arrhythmia, has been associated with different electrophysiological, molecular, and structural alterations in atrial cardiomyocytes. Therefore, more studies are required to elucidate the genetic and molecular basis of AF. Various genome-wide association studies (GWAS) have strongly associated different single nucleotide polymorphisms (SNPs) with AF. One of these GWAS identified the rs13376333 risk SNP as the most significant one from the 1q21 chromosomal region. The rs13376333 risk SNP is intronic to the KCNN3 gene that encodes for small conductance calcium-activated potassium channels type 3 (SK3). However, the functional electrophysiological effects of this variant are not known. SK channels represent a unique family of K+ channels, primarily regulated by cytosolic Ca2+ concentration, and different studies support their critical role in the regulation of atrial excitability and consequently in the development of arrhythmias like AF. Since different studies have shown that both upregulation and downregulation of SK3 channels can lead to arrhythmias by different mechanisms, an important goal is to elucidate whether the rs13376333 risk SNP is a gain-of-function (GoF) or a loss-of-function (LoF) variant. A better understanding of the functional consequences associated with these SNPs could influence clinical practice guidelines by improving genotype-based risk stratification and personalized treatment. Although research using native human atrial cardiomyocytes and animal models has provided useful insights, each model has its limitations. Therefore, there is a critical need to develop a human-derived model that represents human physiology more accurately than existing animal models. In this context, research with human induced pluripotent stem cells (hiPSC) and subsequent generation of cardiomyocytes derived from hiPSC (hiPSC-CMs) has revealed the underlying causes of various cardiovascular diseases and identified treatment opportunities that were not possible using in vitro or in vivo studies with animal models. Thus, the ability to generate atrial cardiomyocytes and atrial tissue derived from hiPSCs from human/patients with specific genetic diseases, incorporating novel genetic editing tools to generate isogenic controls and organelle-specific reporters, and 3D bioprinting of atrial tissue could be essential to study AF pathophysiological mechanisms. In this review, we will first give an overview of SK-channel function, its role in atrial fibrillation and outline pathophysiological mechanisms of KCNN3 risk SNPs. We will then highlight the advantages of using the hiPSC-CM model to investigate SNPs associated with AF, while addressing limitations and best practices for rigorous hiPSC studies.
Collapse
Affiliation(s)
- Hosna Babini
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Verónica Jiménez-Sábado
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- IIB SANT PAU, and CIBERCV, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ekaterina Stogova
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Alia Arslanova
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Mariam Butt
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Parisa Asghari
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Edwin D. W. Moore
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Thomas W. Claydon
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | | | - Leif Hove-Madsen
- IIB SANT PAU, and CIBERCV, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - Glen F. Tibbits
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
27
|
Orenbuch R, Kollasch AW, Spinner HD, Shearer CA, Hopf TA, Franceschi D, Dias M, Frazer J, Marks DS. Deep generative modeling of the human proteome reveals over a hundred novel genes involved in rare genetic disorders. RESEARCH SQUARE 2024:rs.3.rs-3740259. [PMID: 38260496 PMCID: PMC10802723 DOI: 10.21203/rs.3.rs-3740259/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Identifying causal mutations accelerates genetic disease diagnosis, and therapeutic development. Missense variants present a bottleneck in genetic diagnoses as their effects are less straightforward than truncations or nonsense mutations. While computational prediction methods are increasingly successful at prediction for variants in known disease genes, they do not generalize well to other genes as the scores are not calibrated across the proteome1-6. To address this, we developed a deep generative model, popEVE, that combines evolutionary information with population sequence data7 and achieves state-of-the-art performance at ranking variants by severity to distinguish patients with severe developmental disorders8 from potentially healthy individuals9. popEVE identifies 442 genes in patients this developmental disorder cohort, including evidence of 123 novel genetic disorders, many without the need for gene-level enrichment and without overestimating the prevalence of pathogenic variants in the population. A majority of these variants are close to interacting partners in 3D complexes. Preliminary analyses on child exomes indicate that popEVE can identify candidate variants without the need for inheritance labels. By placing variants on a unified scale, our model offers a comprehensive perspective on the distribution of fitness effects across the entire proteome and the broader human population. popEVE provides compelling evidence for genetic diagnoses even in exceptionally rare single-patient disorders where conventional techniques relying on repeated observations may not be applicable.
Collapse
Affiliation(s)
- Rose Orenbuch
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Aaron W. Kollasch
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Hansen D. Spinner
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Courtney A. Shearer
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Dinko Franceschi
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Mafalda Dias
- Dias & Frazer Group, Centre for Genomic Regulation (CRG),The Barcelona Institute of Science and Technology, Barcelona, Spain
- University Pompeu Fabra, Barcelona, Spain
| | - Jonathan Frazer
- Dias & Frazer Group, Centre for Genomic Regulation (CRG),The Barcelona Institute of Science and Technology, Barcelona, Spain
- University Pompeu Fabra, Barcelona, Spain
| | - Debora S. Marks
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
28
|
Mӓnnikkӧ R, Kullmann DM. Structure-function and pharmacologic aspects of ion channels relevant to neurologic channelopathies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 203:1-23. [PMID: 39174242 DOI: 10.1016/b978-0-323-90820-7.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Ion channels are membrane proteins that allow the passage of ions across the membrane. They characteristically contain a pore where the selectivity of certain ion species is determined and gates that open and close the pore are found. The pore is often connected to additional domains or subunits that regulate its function. Channels are grouped into families based on their selectivity for specific ions and the stimuli that control channel opening and closing, such as voltage or ligands. Ion channels are fundamental to the electrical properties of excitable tissues. Dysfunction of channels can lead to abnormal electrical signaling of neurons and muscle cells, accompanied by clinical manifestations, known as channelopathies. Many naturally occurring toxins target ion channels and affect excitable cells where the channels are expressed. Furthermore, ion channels, as membrane proteins and key regulators of a number of physiologic functions, are an important target for drugs in clinical use. In this chapter, we give a general overview of the classification, genetics and structure-function features of the main ion channel families, and address some pharmacologic aspects relevant to neurologic channelopathies.
Collapse
Affiliation(s)
- Roope Mӓnnikkӧ
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
29
|
Charlick JN, Bozadzhieva D, Butler AS, Wilkinson KA, Marrion NV. A single coiled-coil domain mutation in hIKCa channel subunits disrupts preferential formation of heteromeric hSK1:hIKCa channels. Eur J Neurosci 2024; 59:3-16. [PMID: 38018635 PMCID: PMC10952195 DOI: 10.1111/ejn.16189] [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: 02/09/2023] [Revised: 09/22/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
The expression of IKCa (SK4) channel subunits overlaps with that of SK channel subunits, and it has been proposed that the two related subunits prefer to co-assemble to form heteromeric hSK1:hIKCa channels. This implicates hSK1:hIKCa heteromers in physiological roles that might have been attributed to activation of SK channels. We have used a mutation approach to confirm formation of heterometric hSK1:hIKCa channels. Introduction of residues within hSK1 that were predicted to impart sensitivity to the hIKCa current blocker TRAM-34 changed the pharmacology of functional heteromers. Heteromeric channels formed between wildtype hIKCa and mutant hSK1 subunits displayed a significantly higher sensitivity and maximum block to addition of TRAM-34 than heteromers formed between wildtype subunits. Heteromer formation was disrupted by a single point mutation within one COOH-terminal coiled-coil domain of the hIKCa channel subunit. This mutation only disrupted the formation of hSK1:hIKCa heteromeric channels, without affecting the formation of homomeric hIKCa channels. Finally, the Ca2+ gating sensitivity of heteromeric hSK1:hIKCa channels was found to be significantly lower than the Ca2+ gating sensitivity of homomeric hIKCa channels. These data confirmed the preferred formation of heteromeric channels that results from COOH-terminal interactions between subunits. The distinct sensitivity of the heteromer to activation by Ca2+ suggests that heteromeric channels fulfil a distinct function within those neurons that express both subunits.
Collapse
Affiliation(s)
- James N. Charlick
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Daniella Bozadzhieva
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Andrew S. Butler
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Kevin A. Wilkinson
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Neil V. Marrion
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| |
Collapse
|
30
|
Zuccolini P, Barbieri R, Sbrana F, Picco C, Gavazzo P, Pusch M. IK Channel-Independent Effects of Clotrimazole and Senicapoc on Cancer Cells Viability and Migration. Int J Mol Sci 2023; 24:16285. [PMID: 38003471 PMCID: PMC10671816 DOI: 10.3390/ijms242216285] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Many studies highlighted the importance of the IK channel for the proliferation and the migration of different types of cancer cells, showing how IK blockers could slow down cancer growth. Based on these data, we wanted to characterize the effects of IK blockers on melanoma metastatic cells and to understand if such effects were exclusively IK-dependent. For this purpose, we employed two different blockers, namely clotrimazole and senicapoc, and two cell lines: metastatic melanoma WM266-4 and pancreatic cancer Panc-1, which is reported to have little or no IK expression. Clotrimazole and senicapoc induced a decrease in viability and the migration of both WM266-4 and Panc-1 cells irrespective of IK expression levels. Patch-clamp experiments on WM266-4 cells revealed Ca2+-dependent, IK-like, clotrimazole- and senicapoc-sensitive currents, which could not be detected in Panc-1 cells. Neither clotrimazole nor senicapoc altered the intracellular Ca2+ concentration. These results suggest that the effects of IK blockers on cancer cells are not strictly dependent on a robust presence of the channel in the plasma membrane, but they might be due to off-target effects on other cellular targets or to the blockade of IK channels localized in intracellular organelles.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael Pusch
- Biophysics Institute, National Research Council, 16149 Genova, Italy; (P.Z.); (R.B.); (F.S.); (C.P.); (P.G.)
| |
Collapse
|
31
|
Stewart GW, Gibson JS, Rees DC. The cation-leaky hereditary stomatocytosis syndromes: A tale of six proteins. Br J Haematol 2023; 203:509-522. [PMID: 37679660 DOI: 10.1111/bjh.19093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023]
Abstract
This review concerns a series of dominantly inherited haemolytic anaemias in which the membrane of the erythrocyte 'leaks' the univalent cations, compromising the osmotic stability of the cell. The majority of the conditions are explained by mutations in one of six genes, coding for multispanning membrane proteins of different structure and function. These are: RhAG, coding for an ammonium carrier; SLC4A1, coding for the band 3 anion exchanger; PIEZO1, coding for a mechanosensitive cation channel; GLUT1, coding for a glucose transporter; KCNN4, coding for an internal-calcium-activated potassium channel; and ABCB6, coding for a porphyrin transporter. This review describes the five clinical syndromes associated with genetic defects in these genes and their variable genotype/phenotype relationships.
Collapse
Affiliation(s)
- Gordon W Stewart
- Division of Medicine, Faculty of Medical Sciences, University College London, London, UK
| | - John S Gibson
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - David C Rees
- Haematological Medicine, Kings College London, London, UK
| |
Collapse
|
32
|
Szollosi A, Almássy J. Functional characterization of the transient receptor potential melastatin 2 (TRPM2) cation channel from Nematostella vectensis reconstituted into lipid bilayer. Sci Rep 2023; 13:11471. [PMID: 37454209 PMCID: PMC10349829 DOI: 10.1038/s41598-023-38640-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Transient receptor potential melastatin 2 (TRPM2) cation channel activity is required for insulin secretion, immune cell activation and body heat control. Channel activation upon oxidative stress is involved in the pathology of stroke and neurodegenerative disorders. Cytosolic Ca2+, ADP-ribose (ADPR) and phosphatidylinositol-4,5-bisphosphate (PIP2) are the obligate activators of the channel. Several TRPM2 cryo-EM structures have been resolved to date, yet functionality of the purified protein has not been tested. Here we reconstituted overexpressed and purified TRPM2 from Nematostella vectensis (nvTRPM2) into lipid bilayers and found that the protein is fully functional. Consistent with the observations in native membranes, nvTRPM2 in lipid bilayers is co-activated by cytosolic Ca2+ and either ADPR or ADPR-2'-phosphate (ADPRP). The physiological metabolite ADPRP has a higher apparent affinity than ADPR. In lipid bilayers nvTRPM2 displays a large linear unitary conductance, its open probability (Po) shows little voltage dependence and is stable over several minutes. Po is high without addition of exogenous PIP2, but is largely blunted by treatment with poly-L-Lysine, a polycation that masks PIP2 headgroups. These results indicate that PIP2 or some other activating phosphoinositol lipid co-purifies with nvTRPM2, suggesting a high PIP2 binding affinity of nvTRPM2 under physiological conditions.
Collapse
Affiliation(s)
- Andras Szollosi
- Department of Biochemistry, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary.
- ELKH-SE Ion Channel Research Group, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary.
- HCEMM-SE Molecular Channelopathies Research Group, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary.
| | - János Almássy
- Department of Physiology, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary
| |
Collapse
|
33
|
d’Apolito M, Ceccarini C, Savino R, Adipietro I, di Bari I, Santacroce R, Curcetti M, D’Andrea G, Croce AI, Cesarano C, Polito AN, Margaglione M. A Novel KCNN2 Variant in a Family with Essential Tremor Plus: Clinical Characteristics and In Silico Analysis. Genes (Basel) 2023; 14:1380. [PMID: 37510285 PMCID: PMC10379157 DOI: 10.3390/genes14071380] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Essential tremor (ET) is one of the more common movement disorders. Current diagnosis is solely based on clinical findings. ET appears to be inherited in an autosomal dominant pattern. Several loci on specific chromosomes have been studied by linkage analysis, but the causes of essential tremor are still unknown in many patients. Genetic studies described the association of several genes with familial ET. However, they were found only in distinct families, suggesting that some can be private pathogenic variants. AIM OF THE STUDY to characterize the phenotype of an Italian family with ET and identify the genetic variant associated. METHODS Clinical and genetic examinations were performed. Genetic testing was done with whole-exome sequencing (WES) using the Illumina platform. Bidirectional capillary Sanger sequencing was used to investigate the presence of variant in all affected members of the family. In silico prediction of pathogenicity was used to study the effect of gene variants on protein structure. RESULTS The proband was a 15-year-old boy. The patient was the first of two children of a non-consanguineous couple. Family history was remarkable for tremor in the mother line. His mother suffered from bilateral upper extremity kinetic tremors (since she was 20 years old), anxiety, and depression. Other relatives referred bilateral upper extremity tremors. In the index case, WES analysis performed supposing a dominant mode of inheritance, identified a novel heterozygous missense variant in potassium calcium-activated channel subfamily N member 2 (KCNN2) (NM_021614.3: c.1145G>A, p.Gly382Asp). In the pedigree investigation, all carriers of the gene variant had ET and showed variable expressivity, the elder symptomatic relative showing cognitive impairment and hallucinations in the last decade, in addition to tremor since a young age. The amino acid residue #382 is located in a transmembrane region and in silico analysis suggested a causative role for the variant. Modelling of the mutant protein structure showed that the variant causes a clash in the protein structure. Therefore, the variant could cause a conformational change that alters the ability of the protein in the modulation of ion channels Conclusions: The KCNN2 gene variant identified could be associated with ET. The variant could modify a voltage-independent potassium channel activated by intracellular calcium.
Collapse
Affiliation(s)
- Maria d’Apolito
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Caterina Ceccarini
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Rosa Savino
- Neuropsychiatry for Child and Adolescent Unit, Department of Woman and Child, Policlinico Riuniti, 70122 Foggia, Italy; (R.S.); (A.N.P.)
| | - Iolanda Adipietro
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Ighli di Bari
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Rosa Santacroce
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Maria Curcetti
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Giovanna D’Andrea
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Anna-Irma Croce
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Carla Cesarano
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| | - Anna Nunzia Polito
- Neuropsychiatry for Child and Adolescent Unit, Department of Woman and Child, Policlinico Riuniti, 70122 Foggia, Italy; (R.S.); (A.N.P.)
| | - Maurizio Margaglione
- Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 70122 Foggia, Italy; (M.d.); (C.C.); (I.A.); (I.d.B.); (R.S.); (M.C.); (G.D.); (A.-I.C.); (C.C.)
| |
Collapse
|
34
|
Orfali R, AlFaiz A, Rahman MA, Lau L, Nam YW, Zhang M. K Ca2 and K Ca3.1 Channels in the Airways: A New Therapeutic Target. Biomedicines 2023; 11:1780. [PMID: 37509419 PMCID: PMC10376499 DOI: 10.3390/biomedicines11071780] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023] Open
Abstract
K+ channels are involved in many critical functions in lung physiology. Recently, the family of Ca2+-activated K+ channels (KCa) has received more attention, and a massive amount of effort has been devoted to developing selective medications targeting these channels. Within the family of KCa channels, three small-conductance Ca2+-activated K+ (KCa2) channel subtypes, together with the intermediate-conductance KCa3.1 channel, are voltage-independent K+ channels, and they mediate Ca2+-induced membrane hyperpolarization. Many KCa2 channel members are involved in crucial roles in physiological and pathological systems throughout the body. In this article, different subtypes of KCa2 and KCa3.1 channels and their functions in respiratory diseases are discussed. Additionally, the pharmacology of the KCa2 and KCa3.1 channels and the link between these channels and respiratory ciliary regulations will be explained in more detail. In the future, specific modulators for small or intermediate Ca2+-activated K+ channels may offer a unique therapeutic opportunity to treat muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
- Biomedical Research Administration, Research Centre, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia
| | - Ali AlFaiz
- Biomedical Research Administration, Research Centre, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Liz Lau
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| |
Collapse
|
35
|
Liu T, Li T, Xu D, Wang Y, Zhou Y, Wan J, Huang CLH, Tan X. Small-conductance calcium-activated potassium channels in the heart: expression, regulation and pathological implications. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220171. [PMID: 37122223 PMCID: PMC10150224 DOI: 10.1098/rstb.2022.0171] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/25/2022] [Indexed: 05/02/2023] Open
Abstract
Ca2+-activated K+ channels are critical to cellular Ca2+ homeostasis and excitability; they couple intracellular Ca2+ and membrane voltage change. Of these, the small, 4-14 pS, conductance SK channels include three, KCNN1-3 encoded, SK1/KCa2.1, SK2/KCa2.2 and SK3/KCa2.3, channel subtypes with characteristic, EC50 ∼ 10 nM, 40 pM, 1 nM, apamin sensitivities. All SK channels, particularly SK2 channels, are expressed in atrial, ventricular and conducting system cardiomyocytes. Pharmacological and genetic modification results have suggested that SK channel block or knockout prolonged action potential durations (APDs) and effective refractory periods (ERPs) particularly in atrial, but also in ventricular, and sinoatrial, atrioventricular node and Purkinje myocytes, correspondingly affect arrhythmic tendency. Additionally, mitochondrial SK channels may decrease mitochondrial Ca2+ overload and reactive oxygen species generation. SK channels show low voltage but marked Ca2+ dependences (EC50 ∼ 300-500 nM) reflecting their α-subunit calmodulin (CaM) binding domains, through which they may be activated by voltage-gated or ryanodine-receptor Ca2+ channel activity. SK function also depends upon complex trafficking and expression processes and associations with other ion channels or subunits from different SK subtypes. Atrial and ventricular clinical arrhythmogenesis may follow both increased or decreased SK expression through decreased or increased APD correspondingly accelerating and stabilizing re-entrant rotors or increasing incidences of triggered activity. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Ting Liu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Dandi Xu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Yan Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Yafei Zhou
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Juyi Wan
- Department of Cardiovascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Christopher L.-H. Huang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Physiological Laboratory and Department of Biochemistry, University of Cambridge, Cambridge CB2 3EG, UK
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
36
|
Zhang XD, Chiamvimonvat N. Targeting Small-Conductance Calcium-Activated Potassium Channels in Atrial Fibrillation: Therapeutic Opportunities. Circ Res 2023; 132:1104-1106. [PMID: 37104564 PMCID: PMC10155264 DOI: 10.1161/circresaha.123.322777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Affiliation(s)
- Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine (X.-D.Z., N.C.), School of Medicine, University of California, Davis
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine (X.-D.Z., N.C.), School of Medicine, University of California, Davis
- Department of Pharmacology (N.C.), School of Medicine, University of California, Davis
- Department of Veterans Affairs, Northern California Health Care System, Mather (N.C.)
| |
Collapse
|
37
|
Gu RX, de Groot BL. Central cavity dehydration as a gating mechanism of potassium channels. Nat Commun 2023; 14:2178. [PMID: 37069187 PMCID: PMC10110622 DOI: 10.1038/s41467-023-37531-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 03/21/2023] [Indexed: 04/19/2023] Open
Abstract
The hydrophobic gating model, in which ion permeation is inhibited by the hydrophobicity, rather than a physical occlusion of the nanopore, functions in various ion channels including potassium channels. Available research focused on the energy barriers for ion/water conduction due to the hydrophobicity, whereas how hydrophobic gating affects the function and structure of channels remains unclear. Here, we use potassium channels as examples and conduct molecular dynamics simulations to investigate this problem. Our simulations find channel activities (ion currents) highly correlated with cavity hydration level, implying insufficient hydration as a barrier for ion permeation. Enforced cavity dehydration successfully induces conformational transitions between known channel states, further implying cavity dewetting as a key step in the gating procedure of potassium channels utilizing different activation mechanisms. Our work reveals how the cavity dewetting is coupled to structural changes of potassium channels and how it affects channel activity. The conclusion may also apply to other ion channels.
Collapse
Affiliation(s)
- Ruo-Xu Gu
- School of Life Sciences and Biotechnology, Shanghai Jia Tong University, 800 Dongchuan Road, 200240, Shanghai, China
- Department of Theoretical and Computational Biophysics, Max-Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Göttingen, Germany
| | - Bert L de Groot
- Department of Theoretical and Computational Biophysics, Max-Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
38
|
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: 9] [Impact Index Per Article: 4.5] [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
|
39
|
van Herck IGM, Seutin V, Bentzen BH, Marrion NV, Edwards AG. Gating kinetics and pharmacological properties of small-conductance Ca 2+-activated potassium channels. Biophys J 2023; 122:1143-1157. [PMID: 36760125 PMCID: PMC10111258 DOI: 10.1016/j.bpj.2023.02.004] [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: 07/20/2022] [Revised: 11/20/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Small-conductance (SK) calcium-activated potassium channels are a promising treatment target in atrial fibrillation. However, the functional properties that differentiate SK inhibitors remain poorly understood. The objective of this study was to determine how two unrelated SK channel inhibitors, apamin and AP14145, impact SK channel function in excised inside-out single-channel recordings. Surprisingly, both apamin and AP14145 exert much of their inhibition by inducing a class of very-long-lived channel closures (apamin: τc,vl = 11.8 ± 7.1 s, and AP14145: τc,vl = 10.3 ± 7.2 s), which were never observed under control conditions. Both inhibitors also induced changes to the three closed and two open durations typical of normal SK channel gating. AP14145 shifted the open duration distribution to favor longer open durations, whereas apamin did not alter open-state kinetics. AP14145 also prolonged the two shortest channel closed durations (AP14145: τc,s = 3.50 ± 0.81 ms, and τc,i = 32.0 ± 6.76 ms versus control: τc,s = 1.59 ± 0.19 ms, and τc,i = 13.5 ± 1.17 ms), thus slowing overall gating kinetics within bursts of channel activity. In contrast, apamin accelerated intraburst gating kinetics by shortening the two shortest closed durations (τc,s = 0.75 ± 0.10 ms and τc,i = 5.08 ± 0.49 ms) and inducing periods of flickery activity. Finally, AP14145 introduced a unique form of inhibition by decreasing unitary current amplitude. SK channels exhibited two clearly distinguishable amplitudes (control: Ahigh = 0.76 ± 0.03 pA, and Alow = 0.54 ± 0.03 pA). AP14145 both reduced the fraction of patches exhibiting the higher amplitude (AP14145: 4/9 patches versus control: 16/16 patches) and reduced the mean low amplitude (0.38 ± 0.03 pA). Here, we have demonstrated that both inhibitors introduce very long channel closures but that each also exhibits unique effects on other components of SK gating kinetics and unitary current. The combination of these effects is likely to be critical for understanding the functional differences of each inhibitor in the context of cyclical Ca2+-dependent channel activation in vivo.
Collapse
Affiliation(s)
- Ilsbeth G M van Herck
- Computational Physiology Department, Simula Research Laboratory, Oslo, Norway; Institute of Informatics, University of Oslo, Oslo, Norway
| | - Vincent Seutin
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Bo H Bentzen
- Acesion Pharma, Copenhagen, Denmark; Biomedical Institute, University of Copenhagen, Copenhagen, Denmark
| | - Neil V Marrion
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Andrew G Edwards
- Computational Physiology Department, Simula Research Laboratory, Oslo, Norway; Department of Pharmacology, University of California, Davis, California.
| |
Collapse
|
40
|
Vera OD, Wulff H, Braun AP. Endothelial KCa channels: Novel targets to reduce atherosclerosis-driven vascular dysfunction. Front Pharmacol 2023; 14:1151244. [PMID: 37063294 PMCID: PMC10102451 DOI: 10.3389/fphar.2023.1151244] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
Elevated levels of cholesterol in the blood can induce endothelial dysfunction, a condition characterized by impaired nitric oxide production and decreased vasodilatory capacity. Endothelial dysfunction can promote vascular disease, such as atherosclerosis, where macrophages accumulate in the vascular intima and fatty plaques form that impair normal blood flow in conduit arteries. Current pharmacological strategies to treat atherosclerosis mostly focus on lipid lowering to prevent high levels of plasma cholesterol that induce endothelial dysfunction and atherosclerosis. While this approach is effective for most patients with atherosclerosis, for some, lipid lowering is not enough to reduce their cardiovascular risk factors associated with atherosclerosis (e.g., hypertension, cardiac dysfunction, stroke, etc.). For such patients, additional strategies targeted at reducing endothelial dysfunction may be beneficial. One novel strategy to restore endothelial function and mitigate atherosclerosis risk is to enhance the activity of Ca2+-activated K+ (KCa) channels in the endothelium with positive gating modulator drugs. Here, we review the mechanism of action of these small molecules and discuss their ability to improve endothelial function. We then explore how this strategy could mitigate endothelial dysfunction in the context of atherosclerosis by examining how KCa modulators can improve cardiovascular function in other settings, such as aging and type 2 diabetes. Finally, we consider questions that will need to be addressed to determine whether KCa channel activation could be used as a long-term add-on to lipid lowering to augment atherosclerosis treatment, particularly in patients where lipid-lowering is not adequate to improve their cardiovascular health.
Collapse
Affiliation(s)
- O. Daniel Vera
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, United States
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- *Correspondence: Andrew P. Braun,
| |
Collapse
|
41
|
Nam YW, Rahman MA, Yang G, Orfali R, Cui M, Zhang M. Loss-of-function K Ca2.2 mutations abolish channel activity. Am J Physiol Cell Physiol 2023; 324:C658-C664. [PMID: 36717104 PMCID: PMC10069973 DOI: 10.1152/ajpcell.00584.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
Small-conductance Ca2+-activated potassium channels subtype 2 (KCa2.2, also called SK2) are operated exclusively by a Ca2+-calmodulin gating mechanism. Heterozygous genetic mutations of KCa2.2 channels have been associated with autosomal dominant neurodevelopmental disorders including cerebellar ataxia and tremor in humans and rodents. Taking advantage of these pathogenic mutations, we performed structure-function studies of the rat KCa2.2 channel. No measurable current was detected from HEK293 cells heterologously expressing these pathogenic KCa2.2 mutants. When coexpressed with the KCa2.2_WT channel, mutations of the pore-lining amino acid residues (I360M, Y362C, G363S, and I389V) and two proline substitutions (L174P and L433P) dominant negatively suppressed and completely abolished the activity of the coexpressed KCa2.2_WT channel. Coexpression of the KCa2.2_I289N and the KCa2.2_WT channels reduced the apparent Ca2+ sensitivity compared with the KCa2.2_WT channel, which was rescued by a KCa2.2 positive modulator.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California, United States
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California, United States
| | - Grace Yang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California, United States
| | - Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California, United States
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy, Boston, Massachusetts, United States
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California, United States
| |
Collapse
|
42
|
Nam YW, Downey M, Rahman MA, Cui M, Zhang M. Channelopathy of small- and intermediate-conductance Ca 2+-activated K + channels. Acta Pharmacol Sin 2023; 44:259-267. [PMID: 35715699 PMCID: PMC9889811 DOI: 10.1038/s41401-022-00935-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023]
Abstract
Small- and intermediate-conductance Ca2+-activated K+ (KCa2.x/KCa3.1 also called SK/IK) channels are gated exclusively by intracellular Ca2+. The Ca2+ binding protein calmodulin confers sub-micromolar Ca2+ sensitivity to the channel-calmodulin complex. The calmodulin C-lobe is constitutively associated with the proximal C-terminus of the channel. Interactions between calmodulin N-lobe and the channel S4-S5 linker are Ca2+-dependent, which subsequently trigger conformational changes in the channel pore and open the gate. KCNN genes encode four subtypes, including KCNN1 for KCa2.1 (SK1), KCNN2 for KCa2.2 (SK2), KCNN3 for KCa2.3 (SK3), and KCNN4 for KCa3.1 (IK). The three KCa2.x channel subtypes are expressed in the central nervous system and the heart. The KCa3.1 subtype is expressed in the erythrocytes and the lymphocytes, among other peripheral tissues. The impact of dysfunctional KCa2.x/KCa3.1 channels on human health has not been well documented. Human loss-of-function KCa2.2 mutations have been linked with neurodevelopmental disorders. Human gain-of-function mutations that increase the apparent Ca2+ sensitivity of KCa2.3 and KCa3.1 channels have been associated with Zimmermann-Laband syndrome and hereditary xerocytosis, respectively. This review article discusses the physiological significance of KCa2.x/KCa3.1 channels, the pathophysiology of the diseases linked with KCa2.x/KCa3.1 mutations, the structure-function relationship of the mutant KCa2.x/KCa3.1 channels, and potential pharmacological therapeutics for the KCa2.x/KCa3.1 channelopathy.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Myles Downey
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy, Boston, MA, 02115, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA.
| |
Collapse
|
43
|
Numata T, Sato-Numata K, Yoshino M. Intermediate conductance Ca 2+-activated potassium channels are activated by functional coupling with stretch-activated nonselective cation channels in cricket myocytes. FRONTIERS IN INSECT SCIENCE 2023; 2:1100671. [PMID: 38468799 PMCID: PMC10926553 DOI: 10.3389/finsc.2022.1100671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/20/2022] [Indexed: 03/13/2024]
Abstract
Cooperative gating of localized ion channels ranges from fine-tuning excitation-contraction coupling in muscle cells to controlling pace-making activity in the heart. Membrane deformation resulting from muscle contraction activates stretch-activated (SA) cation channels. The subsequent Ca2+ influx activates spatially localized Ca2+-sensitive K+ channels to fine-tune spontaneous muscle contraction. To characterize endogenously expressed intermediate conductance Ca2+-activated potassium (IK) channels and assess the functional relevance of the extracellular Ca2+ source leading to IK channel activity, we performed patch-clamp techniques on cricket oviduct myocytes and recorded single-channel data. In this study, we first investigated the identification of IK channels that could be distinguished from endogenously expressed large-conductance Ca2+-activated potassium (BK) channels by adding extracellular Ba2+. The single-channel conductance of the IK channel was 62 pS, and its activity increased with increasing intracellular Ca2+ concentration but was not voltage-dependent. These results indicated that IK channels are endogenously expressed in cricket oviduct myocytes. Second, the Ca2+ influx pathway that activates the IK channel was investigated. The absence of extracellular Ca2+ or the presence of Gd3+ abolished the activity of IK channels. Finally, we investigated the proximity between SA and IK channels. The removal of extracellular Ca2+, administration of Ca2+ to the microscopic region in a pipette, and application of membrane stretching stimulation increased SA channel activity, followed by IK channel activity. Membrane stretch-induced SA and IK channel activity were positively correlated. However, the emergence of IK channel activity and its increase in response to membrane mechanical stretch was not observed without Ca2+ in the pipette. These results strongly suggest that IK channels are endogenously expressed in cricket oviduct myocytes and that IK channel activity is regulated by neighboring SA channel activity. In conclusion, functional coupling between SA and IK channels may underlie the molecular basis of spontaneous rhythmic contractions.
Collapse
Affiliation(s)
- Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| | - Masami Yoshino
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| |
Collapse
|
44
|
Thale I, Maskri S, Grey L, Todesca LM, Budde T, Maisuls I, Strassert CA, Koch O, Schwab A, Wünsch B. Imaging of K Ca 3.1 Channels in Tumor Cells with PET and Small-Molecule Fluorescent Probes. ChemMedChem 2023; 18:e202200551. [PMID: 36315933 PMCID: PMC10098740 DOI: 10.1002/cmdc.202200551] [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: 10/10/2022] [Revised: 10/31/2022] [Indexed: 01/20/2023]
Abstract
The Ca2+ activated K+ channel KCa 3.1 is overexpressed in several human tumor cell lines, e. g. clear cell renal carcinoma, prostate cancer, non-small cell lung cancer. Highly aggressive cancer cells use this ion channel for key processes of the metastatic cascade such as migration, extravasation and invasion. Therefore, small molecules, which are able to image this KCa 3.1 channel in vitro and in vivo represent valuable diagnostic and prognostic tool compounds. The [18 F]fluoroethyltriazolyl substituted senicapoc was used as positron emission tomography (PET) tracer and showed promising properties for imaging of KCa 3.1 channels in lung adenocarcinoma cells in mice. The novel senicapoc BODIPY conjugates with two F-atoms (9 a) and with a F-atom and a methoxy moiety (9 b) at the B-atom led to the characteristic punctate staining pattern resulting from labeling of single KCa 3.1 channels in A549-3R cells. This punctate pattern was completely removed by preincubation with an excess of senicapoc confirming the high specificity of KCa 3.1 labeling. Due to the methoxy moiety at the B-atom and the additional oxyethylene unit in the spacer, 9 b exhibits higher polarity, which improves solubility and handling without reduction of fluorescence quantum yield. Docking studies using a cryo-electron microscopy (EM) structure of the KCa 3.1 channel confirmed the interaction of 9 a and 9 b with a binding pocket in the channel pore.
Collapse
Affiliation(s)
- Insa Thale
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Sarah Maskri
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Lucie Grey
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Luca Matteo Todesca
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology II, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Thomas Budde
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology I, Robert-Koch-Straße 27a, 48149, Münster, Germany
| | - Ivan Maisuls
- Westfälische Wilhelms-Universität Münster, Institut für Anorganische und Analytische Chemie CiMIC, SoN, Corrensstraße 28, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, CeNTech, Heisenbergstraße 11, 48149, Münster, Germany
| | - Cristian A Strassert
- Westfälische Wilhelms-Universität Münster, Institut für Anorganische und Analytische Chemie CiMIC, SoN, Corrensstraße 28, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, CeNTech, Heisenbergstraße 11, 48149, Münster, Germany
| | - Oliver Koch
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| | - Albrecht Schwab
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Universitätsklinikum Münster, Institute of Physiology II, Robert-Koch-Straße 27b, 48149, Münster, Germany
| | - Bernhard Wünsch
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, 48149, Münster, Germany.,Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, 48149, Münster, Germany
| |
Collapse
|
45
|
Zahra A, Liu R, Han W, Meng H, Wang Q, Wang Y, Campbell SL, Wu J. K Ca-Related Neurological Disorders: Phenotypic Spectrum and Therapeutic Indications. Curr Neuropharmacol 2023; 21:1504-1518. [PMID: 36503451 PMCID: PMC10472807 DOI: 10.2174/1570159x21666221208091805] [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] [Received: 04/21/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 12/14/2022] Open
Abstract
Although potassium channelopathies have been linked to a wide range of neurological conditions, the underlying pathogenic mechanism is not always clear, and a systematic summary of clinical manifestation is absent. Several neurological disorders have been associated with alterations of calcium-activated potassium channels (KCa channels), such as loss- or gain-of-function mutations, post-transcriptional modification, etc. Here, we outlined the current understanding of the molecular and cellular properties of three subtypes of KCa channels, including big conductance KCa channels (BK), small conductance KCa channels (SK), and the intermediate conductance KCa channels (IK). Next, we comprehensively reviewed the loss- or gain-of-function mutations of each KCa channel and described the corresponding mutation sites in specific diseases to broaden the phenotypic-genotypic spectrum of KCa-related neurological disorders. Moreover, we reviewed the current pharmaceutical strategies targeting KCa channels in KCa-related neurological disorders to provide new directions for drug discovery in anti-seizure medication.
Collapse
Affiliation(s)
- Aqeela Zahra
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Department of Zoology, University of Sialkot, Sialkot 51310, Pakistan
| | - Ru Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Wenzhe Han
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Hui Meng
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Qun Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - YunFu Wang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Susan L. Campbell
- Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| |
Collapse
|
46
|
Halling DB, Philpo AE, Aldrich RW. Calcium dependence of both lobes of calmodulin is involved in binding to a cytoplasmic domain of SK channels. eLife 2022; 11:e81303. [PMID: 36583726 PMCID: PMC9803350 DOI: 10.7554/elife.81303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022] Open
Abstract
KCa2.1-3 Ca2+-activated K+-channels (SK) require calmodulin to gate in response to cellular Ca2+. A model for SK gating proposes that the N-terminal domain (N-lobe) of calmodulin is required for activation, but an immobile C-terminal domain (C-lobe) has constitutive, Ca2+-independent binding. Although structures support a domain-driven hypothesis of SK gate activation by calmodulin, only a partial understanding is possible without measuring both channel activity and protein binding. We measured SK2 (KCa2.2) activity using inside-out patch recordings. Currents from calmodulin-disrupted SK2 channels can be restored with exogenously applied calmodulin. We find that SK2 activity only approaches full activation with full-length calmodulin with both an N- and a C-lobe. We measured calmodulin binding to a C-terminal SK peptide (SKp) using both composition-gradient multi-angle light-scattering and tryptophan emission spectra. Isolated lobes bind to SKp with high affinity, but isolated lobes do not rescue SK2 activity. Consistent with earlier models, N-lobe binding to SKp is stronger in Ca2+, and C-lobe-binding affinity is strong independent of Ca2+. However, a native tryptophan in SKp is sensitive to Ca2+ binding to both the N- and C-lobes of calmodulin at Ca2+ concentrations that activate SK2, demonstrating that the C-lobe interaction with SKp changes with Ca2+. Our peptide-binding data and electrophysiology show that SK gating models need deeper scrutiny. We suggest that the Ca2+-dependent associations of both lobes of calmodulin to SKp are crucial events during gating. Additional investigations are necessary to complete a mechanistic gating model consistent with binding, physiology, and structure.
Collapse
Affiliation(s)
- David B Halling
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| | - Ashley E Philpo
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| | - Richard W Aldrich
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| |
Collapse
|
47
|
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
|
48
|
Tiffner A, Hopl V, Derler I. CRAC and SK Channels: Their Molecular Mechanisms Associated with Cancer Cell Development. Cancers (Basel) 2022; 15:101. [PMID: 36612099 PMCID: PMC9817886 DOI: 10.3390/cancers15010101] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Cancer represents a major health burden worldwide. Several molecular targets have been discovered alongside treatments with positive clinical outcomes. However, the reoccurrence of cancer due to therapy resistance remains the primary cause of mortality. Endeavors in pinpointing new markers as molecular targets in cancer therapy are highly desired. The significance of the co-regulation of Ca2+-permeating and Ca2+-regulated ion channels in cancer cell development, proliferation, and migration make them promising molecular targets in cancer therapy. In particular, the co-regulation of the Orai1 and SK3 channels has been well-studied in breast and colon cancer cells, where it finally leads to an invasion-metastasis cascade. Nevertheless, many questions remain unanswered, such as which key molecular components determine and regulate their interplay. To provide a solid foundation for a better understanding of this ion channel co-regulation in cancer, we first shed light on the physiological role of Ca2+ and how this ion is linked to carcinogenesis. Then, we highlight the structure/function relationship of Orai1 and SK3, both individually and in concert, their role in the development of different types of cancer, and aspects that are not yet known in this context.
Collapse
Affiliation(s)
- Adéla Tiffner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| |
Collapse
|
49
|
Xu H, Wang D, Ramponi C, Wang X, Zhang H. The P21-Activated Kinase 1 and 2 As Potential Therapeutic Targets for the Management of Cardiovascular Disease. INTERNATIONAL JOURNAL OF DRUG DISCOVERY AND PHARMACOLOGY 2022:5. [PMID: 39899001 PMCID: PMC7617276 DOI: 10.53941/ijddp.v1i1.179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Group I p21-activated kinases (Paks) are members of the serine/threonine protein kinase family. Paks are encoded by three genes (Pak 1 - 3) and are involved in the regulation of various biological processes. Pak1 and Pak2 are key members, sharing 91% sequence identity in their kinase domains. Recent studies have shown that Pak1/2 protect the heart from various types of stresses. Activated Pak1/2 participate in the maintenance of cellular homeostasis and metabolism, thus enhancing the adaptation and resilience of cardiomyocytes to stress. The structure, activation and function of Pak1/2 as well as their protective roles against the occurrence of cardiovascular disease are described in this review. The values of Pak1/2 as therapeutic targets are also discussed.
Collapse
Affiliation(s)
- Honglin Xu
- Michael Smith building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Dingwei Wang
- Michael Smith building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Chiara Ramponi
- Michael Smith building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Xin Wang
- Michael Smith building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Hongyuan Zhang
- Michael Smith building, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
50
|
Vaisey G, Banerjee P, North AJ, Haselwandter CA, MacKinnon R. Piezo1 as a force-through-membrane sensor in red blood cells. eLife 2022; 11:e82621. [PMID: 36515266 PMCID: PMC9750178 DOI: 10.7554/elife.82621] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Piezo1 is the stretch activated Ca2+ channel in red blood cells that mediates homeostatic volume control. Here, we study the organization of Piezo1 in red blood cells using a combination of super-resolution microscopy techniques and electron microscopy. Piezo1 adopts a non-uniform distribution on the red blood cell surface, with a bias toward the biconcave 'dimple'. Trajectories of diffusing Piezo1 molecules, which exhibit confined Brownian diffusion on short timescales and hopping on long timescales, also reflect a bias toward the dimple. This bias can be explained by 'curvature coupling' between the intrinsic curvature of the Piezo dome and the curvature of the red blood cell membrane. Piezo1 does not form clusters with itself, nor does it colocalize with F-actin, Spectrin, or the Gardos channel. Thus, Piezo1 exhibits the properties of a force-through-membrane sensor of curvature and lateral tension in the red blood cell.
Collapse
Affiliation(s)
- George Vaisey
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
| | - Priyam Banerjee
- Bio-Imaging Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Alison J North
- Bio-Imaging Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Christoph A Haselwandter
- Department of Physics and Astronomy and Department of Quantitative and Computational Biology, University of Southern CaliforniaLos AngelesUnited States
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
| |
Collapse
|