1
|
Teimouri-Korani H, Hemmatinafar M, Willems MET, Rezaei R, Imanian B. Individual responses to encapsulated caffeine and caffeine chewing gum on strength and power in strength-trained males. J Int Soc Sports Nutr 2025; 22:2495228. [PMID: 40249126 PMCID: PMC12010647 DOI: 10.1080/15502783.2025.2495228] [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: 01/16/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Liquid-dissolved and encapsulated powder are two popular ways to consume caffeine for performance-enhancing effects. Caffeine in other delivery methods, such as chewing gums, orally dissolvable strips, gels, mouthwashes, energy drinks, and nasal sprays, is believed to be absorbed more quickly into the bloodstream. Inter-individual responses to caffeine's enhancing effects are recognized. The present study examined the inter-individual responses to the acute effects of encapsulated caffeine and caffeinated chewing gum on the lower-body isokinetic and isometric strength and power in strength-trained males. METHOD A randomized, cross-over, placebo-controlled study was conducted with 15 strength-trained males (age: 25 ± 4 years, height: 176 ± 7 cm, weight: 75 ± 11 kg, habitual caffeine intake: 66 ± 15 mg·day-1). Participants were randomly assigned to three conditions: i) caffeinated chewing gum (CG), ii) caffeine capsule (CC), and iii) starch capsule as a placebo (PLA). Participants consumed approximately 3 to 4.5 mg·kg-1 of caffeine 60 minutes before testing. The washout period between conditions was one week. Participants performed the Sargent jump test, followed by a 5-minute active recovery (walking). Subsequently, isokinetic strength and power (60°/s and 180°/s) and isometric strength (45° and 60°) parameters were measured for knee extensor and flexor muscles. Data were analyzed using one-way repeated measures ANOVA and Bonferroni post hoc tests, with significance set at p ≤ 0.05. Responders to the caffeine conditions were identified using the smallest worthwhile change (SWC) analysis. RESULTS In knee extensors, 1) average peak torque and power at 60°/s were higher in CC (p = 0.045; + 11.2% and p = 0.038; + 14.1%) and CG (p = 0.044; + 7.3% and p = 0.015; + 11.4%) compared to PLA with a co-response rate of 60% and 66%, 2) maximum voluntary isometric contraction at 45° (MVIC-45°) was higher in CC compared to PLA (p = 0.031; + 10.1%), and 3) MVIC-60° was higher in CG compared to PLA (p = 0.037; + 10.1%) with a co-response rate of 60%. In knee flexors, 1) time to peak torque at 60°/s was higher in CG compared to PLA (p = 0.011; + 18.2%) with a co-response rate of 46%, 2) average rate of force development at 60°/s was higher in CC (p = 0.007; + 24.1%) and CG (p = 0.050; + 20.6%) compared to PLA with a co-response rate of 53%, and 3) average power at 180°/s was higher in CC compared to PLA (p = 0.033; + 18%) with a co-response rate of 46%. However, there were no differences between other strength indicators in the knee extensors and flexors between the different conditions. Vertical jump height (VJH) was higher in CC (p = 0.001; + 5.5%) and CG (p = 0.001; + 6.) compared to PLA, with a co-response rate of 53%. CONCLUSION Caffeine supplementation in CC and CG forms significantly enhanced lower-body strength, power, and vertical jump height in strength-trained males, with over ~50% of participants exceeding the SWC thresholds across key performance metrics. CC showed slightly higher responder rates for strength parameters, while CG excelled in time-dependent measures, supporting their use as effective and flexible ergogenic aids.
Collapse
Affiliation(s)
- Hamed Teimouri-Korani
- Shahid Beheshti University, Department of Biological Sciences in Sports, Faculty of Sport Sciences and Health, Tehran, Iran
| | - Mohammad Hemmatinafar
- Shiraz University, Department of Sport Sciences, Faculty of Education and Psychology, Shiraz, Iran
| | - Mark ET Willems
- University of Chichester, Institute of Applied Sciences, Chichester, UK
| | - Rasoul Rezaei
- Shiraz University, Department of Sport Sciences, Faculty of Education and Psychology, Shiraz, Iran
| | - Babak Imanian
- Shiraz University, Department of Sport Sciences, Faculty of Education and Psychology, Shiraz, Iran
| |
Collapse
|
2
|
Chen M. Building molecular model series from heterogeneous CryoEM structures using Gaussian mixture models and deep neural networks. Commun Biol 2025; 8:798. [PMID: 40415012 DOI: 10.1038/s42003-025-08202-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
Cryogenic electron microscopy (CryoEM) produces structures of macromolecules at near-atomic resolution. However, building molecular models with good stereochemical geometry from those structures can be challenging and time-consuming, especially when many structures are obtained from datasets with conformational heterogeneity. Here we present a model refinement protocol that automatically generates series of molecular models from CryoEM datasets, which describe the dynamics of the macromolecular system and have near-perfect geometry scores. This method makes it easier to interpret the movement of the protein complex from heterogeneity analysis and to compare the structural dynamics observed from CryoEM data with results from other experimental and simulation techniques.
Collapse
Affiliation(s)
- Muyuan Chen
- Division of CryoEM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, USA.
| |
Collapse
|
3
|
Yu Y, Zhou Y, Zhang Y, Cong Z, Tong Z, Wang J, Feng L, Hou T, Li Z, Guo X, Qu Y. Beyond Hotspots: Functional Characterization of the Novel p.Asp2730Tyr Mutation in RYR1 Associated With Malignant Hyperthermia. Anesth Analg 2025:00000539-990000000-01311. [PMID: 40408285 DOI: 10.1213/ane.0000000000007584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
BACKGROUND Malignant hyperthermia (MH) is a life-threatening pharmacogenetic disorder triggered by certain anesthetics, characterized by muscle rigidity, elevated body temperature, and hypermetabolic crisis. This condition is primarily associated with genetic mutations in ryanodine receptor 1 (RYR1), which encodes the pivotal calcium release channel in the sarcoplasmic reticulum of skeletal muscle. While numerous hotspot mutations in RYR1 have been identified, the functional impact of nonhotspot mutations on channel activity related to MH remains insufficiently investigated. In this study, we identified a known pathogenic mutation (p.Arg2508His) and a novel variant (p.Asp2730Tyr), both located outside the conventional MH hotspots, in 2 patients with clinical suspicion of MH. Our objective was to investigate the functional implications of the p.Asp2730Tyr mutation in RYR1 on calcium release dynamics related to MH. METHODS We engineered a recombinant wild-type (WT) plasmid (pcDNA3.1-3Myc-His-RYR1-WT) to express the full-length mouse skeletal muscle RYR1 using seamless multi-fragment cloning techniques. Two RYR1 mutations, p.Arg2508His (used as positive control) and p.Asp2730Tyr, were separately introduced into the WT plasmid, generating 2 mutant constructs (pcDNA3.1-3Myc-His-RYR1-p.Arg2508His and pcDNA3.1-3Myc-His-RYR1-p.Asp2730Tyr). We utilized 293T cells expression system to express either the WT or mutant forms of mouse RYR1. Fluo-4 calcium imaging was conducted to evaluate the alterations in calcium release in response to RYR1 agonists, caffeine or 4-chloro-m-cresol (4CmC), for each mutation compared to WT. RESULTS Cells transfected with the p.Arg2508His or p.Asp2730Tyr mutation demonstrated a leftward shift in the caffeine and 4CmC concentration-response curves compared to WT, suggesting an increased channel sensitivity to caffeine and 4CmC (P < .001). The mean ± standard error of the mean (SEM) of the EC50 values for caffeine-induced calcium release was 2.56 ± 0.04 mM in WT, which significantly decreased to 1.32 ± 0.13 mM for p.Arg2508His (P < .001) and 1.12 ± 0.09 mM for p.Asp2730Tyr (P < .001). For 4CmC, the EC50 values were 43.2 ± 1.90 μM in WT, 17.2 ± 0.76 μM for p.Arg2508His (P < .001), and 21.8 ± 1.04 μM for p.Asp2730Tyr (P < .001), indicating enhanced calcium release in both mutations. CONCLUSIONS The p.Asp2730Tyr mutation, situated beyond the established RYR1 hotspot regions, significantly alters calcium release dynamics related to MH. A comprehensive investigation into the structural conformations, functional assays, and in vivo mechanisms associated with this mutation could yield a more profound understanding of the molecular underpinnings of MH pathogenesis.
Collapse
Affiliation(s)
- Yao Yu
- From the Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yang Zhou
- From the Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yiyin Zhang
- Department of Physiology, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Zhukai Cong
- From the Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Zexin Tong
- Department of Physiology, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Jiechu Wang
- From the Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Luyang Feng
- From the Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Tingting Hou
- Department of Physiology, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Zhengqian Li
- From the Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiangyang Guo
- From the Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yinyin Qu
- From the Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
4
|
Li X, Hu S, Yu Z, He F, Zhao X, Liu R. New Evidence for the Mechanisms of Nanoplastics Amplifying Cadmium Cytotoxicity: Trojan Horse Effect, Inflammatory Response, and Calcium Imbalance. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:9471-9485. [PMID: 40350783 DOI: 10.1021/acs.est.5c01254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Nanoplastics (NPs) are emerging pollutants worldwide. Particularly worrisome is that although studies have reported that NPs can amplify the biotoxicity of environmental pollutants, the specific mechanism remains unclear. Here, we found that NPs, even without significant toxicity (cell survival: 99.11%), amplified the hepatocyte toxicity of Cd2+. Mechanistically, higher Cd2+ uptake (Δ = 23.80%) combined with crucial intracellular desorption behavior of Cd2+ loaded in NPs (desorption rate: 82.70%) were identified as prerequisites for NPs amplifying Cd2+ cytotoxicity. As for toxigenic pathways, the inflammatory response and calcium (Ca) signaling pathway were identified as the primary molecular events leading to the amplification of Cd2+ cytotoxicity. Further phenotypic monitoring revealed that NPs synergized with Cd2+ to induce more severe pyroptosis and apoptosis by activating the inflammatory caspase-1-dependent and Ca2+-mitochondrial-caspase-3 pathways to a greater extent, respectively. This study reveals and proves for the first time the "Trojan horse" effects of NPs, thus elucidating the actual mechanisms by which NPs act as toxicity amplifiers of pollutants, providing significant insights into accurate risk assessment of NPs in composite pollution.
Collapse
Affiliation(s)
- Xiangxiang Li
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, 72# Jimo Binhai Road, Qingdao, Shandong 266237, China
| | - Shaoyang Hu
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, 72# Jimo Binhai Road, Qingdao, Shandong 266237, China
| | - Zelian Yu
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, 72# Jimo Binhai Road, Qingdao, Shandong 266237, China
| | - Falin He
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, 72# Jimo Binhai Road, Qingdao, Shandong 266237, China
| | - Xingchen Zhao
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, 72# Jimo Binhai Road, Qingdao, Shandong 266237, China
| | - Rutao Liu
- School of Environmental Science and Engineering, China-America CRC for Environment & Health, Shandong University, 72# Jimo Binhai Road, Qingdao, Shandong 266237, China
| |
Collapse
|
5
|
Zahradníková A, Pavelková J, Sabo M, Baday S, Zahradník I. Structure-based mechanism of RyR channel operation by calcium and magnesium ions. PLoS Comput Biol 2025; 21:e1012950. [PMID: 40300027 PMCID: PMC12119028 DOI: 10.1371/journal.pcbi.1012950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 05/28/2025] [Accepted: 03/11/2025] [Indexed: 05/01/2025] Open
Abstract
Ryanodine receptors (RyRs) serve for excitation-contraction coupling in skeletal and cardiac muscle cells in a noticeably different way, not fully understood at the molecular level. We addressed the structure of skeletal (RyR1) and cardiac (RyR2) isoforms relevant to gating by Ca2+ and Mg2+ ions (M2+). Bioinformatics analysis of RyR structures ascertained the EF-hand loops as the M2+ binding inhibition site and revealed its allosteric coupling to the channel gate. The intra-monomeric inactivation pathway interacts with the Ca2+-activation pathway in both RyR isoforms, and the inter-monomeric pathway, stronger in RyR1, couples to the gate through the S23*-loop of the neighbor monomer. These structural findings were implemented in the model of RyR operation based on statistical mechanics and the Monod-Wyman-Changeux theorem. The model, which defines closed, open, and inactivated macrostates allosterically coupled to M2+-binding activation and inhibition sites, approximated the open probability data for both RyR1 and RyR2 channels at a broad range of M2+ concentrations. The proposed mechanism of RyR operation provides a new interpretation of the structural and functional data of mammalian RyR channels on common grounds. This may provide a new platform for designing pharmacological interventions in the relevant diseases of skeletal and cardiac muscles. The synthetic approach developed in this work may find general use in deciphering mechanisms of ion channel functions.
Collapse
Affiliation(s)
- Alexandra Zahradníková
- Department of Cellular Cardiology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jana Pavelková
- Department of Cellular Cardiology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Miroslav Sabo
- Bioinformatics Laboratory, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Sefer Baday
- Applied Informatics Department, Informatics Institute, Istanbul Technical University, Istanbul, Türkiye
| | - Ivan Zahradník
- Department of Cellular Cardiology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
6
|
Dong M, Maturana AD. Effects of aging on calcium channels in skeletal muscle. Front Mol Biosci 2025; 12:1558456. [PMID: 40177518 PMCID: PMC11961898 DOI: 10.3389/fmolb.2025.1558456] [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: 01/10/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
In skeletal muscle, calcium is not only essential to stimulate and sustain their contractions but also for muscle embryogenesis, regeneration, energy production in mitochondria, and fusion. Different ion channels contribute to achieving the various functions of calcium in skeletal muscles. Muscle contraction is initiated by releasing calcium from the sarcoplasmic reticulum through the ryanodine receptor channels gated mechanically by four dihydropyridine receptors of T-tubules. The calcium influx through store-operated calcium channels sustains the contraction and stimulates muscle regeneration. Mitochondrial calcium uniporter allows the calcium entry into mitochondria to stimulate oxidative phosphorylation. Aging alters the expression and activity of these different calcium channels, resulting in a reduction of skeletal muscle force generation and regeneration capacity. Regular physical training and bioactive molecules from nutrients can prevent the effects of aging on calcium channels. This review focuses on the current knowledge of the effects of aging on skeletal muscles' calcium channels.
Collapse
Affiliation(s)
| | - Andrés Daniel Maturana
- Department of Applied Biosciences, Graduate School of Bioagricultural Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
7
|
Arige V, Wagner LE, Malik S, Baker MR, Fan G, Serysheva II, Yule DI. Functional investigation of a putative calcium-binding site involved in the inhibition of inositol 1,4,5-trisphosphate receptor activity. J Biol Chem 2025; 301:108302. [PMID: 39947469 PMCID: PMC11938044 DOI: 10.1016/j.jbc.2025.108302] [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: 11/19/2024] [Revised: 02/01/2025] [Accepted: 02/07/2025] [Indexed: 02/19/2025] Open
Abstract
The regulation of inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) activity is thought to define the spatiotemporal patterns of Ca2+ signals necessary for the appropriate activation of downstream effectors. The binding of both IP3 and Ca2+ is obligatory for IP3R channel opening. Ca2+ however regulates IP3R activity in a biphasic manner. Ca2+ binding to a high-affinity pocket formed by the third armadillo repeat domain and linker domain promotes IP3R channel opening without altering the Ca2+ dependency for channel inactivation. These data suggest that a distinct low-affinity Ca2+-binding site is responsible for the reduction in IP3R activity at higher [Ca2+]. We mutated a cluster of acidic residues in the second armadillo repeat domain and central linker domain of IP3R type 1, reported to coordinate Ca2+ in the cryo-EM structures of the IP3R type 3. This "CD Ca2+-binding site" is well conserved in all IP3R subtypes. CD site Ca2+-binding mutants where the negatively charged glutamic acid residues were mutated to alanine exhibited enhanced sensitivity to IP3-generating agonists. Ca2+-binding mutants displayed spontaneous elemental Ca2+ puffs, and the number of IP3-induced Ca2+ puffs was augmented in cells stably expressing Ca2+-binding site mutants. The inhibitory effect of high [Ca2+] on single-channel open probability (Po) was reduced in mutant channels, and this effect was dependent on [ATP]. This indicates that Ca2+ binding to the putative CD Ca2+ inhibitory site facilitates the reduction in IP3R channel activation at subsaturating, likely physiological cytosolic [ATP], and suggest that at higher [ATP], additional Ca2+-binding motifs may contribute to the biphasic regulation of IP3-induced Ca2+ release.
Collapse
Affiliation(s)
- Vikas Arige
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Larry E Wagner
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Mariah R Baker
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Guizhen Fan
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Irina I Serysheva
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA.
| |
Collapse
|
8
|
Wei R, Chen Q, Zhang L, Liu C, Liu C, Yin CC, Hu H. Structural insights into transmembrane helix S0 facilitated RyR1 channel gating by Ca 2+/ATP. Nat Commun 2025; 16:1936. [PMID: 39994184 PMCID: PMC11850639 DOI: 10.1038/s41467-025-57074-4] [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: 09/26/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
The type-1 ryanodine receptor (RyR1) is an intracellular calcium release channel for skeletal muscle excitation-contraction coupling. Previous structural studies showed that the RyR1 activity is modulated by the exogenous regulators including caffeine, ryanodine, PCB-95 and diamide. An additional transmembrane helix, located adjacent to S1 and S4, has been observed in some structures, although its function remains unclear. Here, we report that using a mild purification procedure, this helix is co-purified with RyR1 and is designated as S0. When RyR1 is coupled with S0, it can be activated by Ca2+ to an open state; however when decoupled from S0, it remains in primed state. S0 regulates the channel conformation by directly affecting the TM domain via the pVSD-S0-S4/S5 linker coupling, which facilitates the dilation of S6. Our results demonstrate that S0 is an essential component of RyR1 and plays a key role in the physiological regulation of RyR1 channel gating.
Collapse
Affiliation(s)
- Risheng Wei
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen; Shenzhen, Guangdong, 518172, China
| | - Lei Zhang
- Electron Microscopy Analysis Laboratory, Medical and Health Analysis Center, Peking University, Beijing, 100191, China
| | - Congcong Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; Shenzhen, Guangdong, 518112, China
| | - Chuang Liu
- Center for Biological Cryo-EM, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
| | - Chang-Cheng Yin
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen; Shenzhen, Guangdong, 518172, China.
| |
Collapse
|
9
|
Townson J, Progida C. The emerging roles of the endoplasmic reticulum in mechanosensing and mechanotransduction. J Cell Sci 2025; 138:JCS263503. [PMID: 39976266 DOI: 10.1242/jcs.263503] [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] [Indexed: 02/21/2025] Open
Abstract
Cells are continuously subjected to physical and chemical cues from the extracellular environment, and sense and respond to mechanical cues via mechanosensation and mechanotransduction. Although the role of the cytoskeleton in these processes is well known, the contribution of intracellular membranes has been long neglected. Recently, it has become evident that various organelles play active roles in both mechanosensing and mechanotransduction. In this Review, we focus on mechanosensitive roles of the endoplasmic reticulum (ER), the functions of which are crucial for maintaining cell homeostasis. We discuss the effects of mechanical stimuli on interactions between the ER, the cytoskeleton and other organelles; the role of the ER in intracellular Ca2+ signalling via mechanosensitive channels; and how the unfolded protein response and lipid homeostasis contribute to mechanosensing. The expansive structure of the ER positions it as a key intracellular communication hub, and we additionally explore how this may be leveraged to transduce mechanical signals around the cell. By synthesising current knowledge, we aim to shed light on the emerging roles of the ER in cellular mechanosensing and mechanotransduction.
Collapse
Affiliation(s)
- Jonathan Townson
- Department of Biosciences, University of Oslo, Blindernveien 31, 0316 Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, University of Oslo, Blindernveien 31, 0316 Oslo, Norway
| |
Collapse
|
10
|
Iyer KA, Kobayashi T, Murayama T, Samsó M. Dantrolene inhibition of ryanodine receptor 1 carrying the severe malignant hyperthermia mutation Y522S visualized by cryo-EM. Structure 2025; 33:338-348.e4. [PMID: 39708816 PMCID: PMC11805659 DOI: 10.1016/j.str.2024.11.018] [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: 07/25/2024] [Revised: 10/30/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024]
Abstract
Mutations in the skeletal isoform of the ryanodine receptor 1 (RyR1) pose grave risks during anesthesia or treatment with succinylcholine muscle relaxants. These can trigger a potentially lethal malignant hyperthermia (MH) episode via intracellular calcium increase mainly from RyR1 channel leakage. Dantrolene is the only known treatment option to prevent death. The main target of dantrolene is RyR1; however, little is known about the mechanism of inhibition. Cryoelectron microscopy (cryo-EM) structures of dantrolene bound to the severe MH Y522S RyR1 mutant in the closed and open states at 2.5-3.3 Å resolution revealed that the drug binds to the channel's cytoplasmic assembly, far from the ion gate, interacting with residues W882, W996, and R1000 in the P1 domain. The finding was validated by Ca2+ imaging and [3H]ryanodine binding in wild-type (WT) and alanine mutants. Dantrolene reduced channel opening probability by restricting the central activation module, "cooling down" the primed conformation caused by the mutation.
Collapse
Affiliation(s)
- Kavita A Iyer
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Takuya Kobayashi
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takashi Murayama
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Montserrat Samsó
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
11
|
Sadakierska-Chudy A, Szymanowski P, Szepieniec WK, Boniewska-Bernacka E, Pollak A. Whole Exome Sequencing Reveals Candidate Variants in Ion Channel Genes for Pelvic Muscle Dysfunction in Young Females with a Family History. Int Urogynecol J 2025; 36:457-468. [PMID: 39833541 DOI: 10.1007/s00192-025-06048-7] [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: 08/16/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
INTRODUCTION AND HYPOTHESIS Pelvic floor dysfunction usually results in pelvic organ prolapse (POP) and/or urinary incontinence. In women, several factors, including pregnancy and vaginal delivery, can affect pelvic muscle conditions. The aim of the study was to perform a genetic analysis in young women with a family history of pelvic floor dysfunction to find potentially harmful variants or variants that increase the risk of developing pelvic floor disorders. METHODS We employed whole exome sequencing to test ten young women with pelvic floor muscle dysfunction (along with their parents) and a family history. The average age of symptoms was 29.1 (± 3.98) years old, soon after their first delivery. RESULTS In five out of ten patients, trio-based WES analysis revealed potentially pathogenic, causative nonsense variants in ion channel genes, including ATP1A4, CLCN1, GRIN2C, and ORAI1, as well as missense variants in PIEZO1 and RYR1. Additionally, some of these patients had variants in genes related to muscle function (MUSK) and connective tissue disorder (FKBP14, p.Glu122ArgfsTer7). The variants found in this study, such as CLCN1 (p.Arg894Ter) and MUSK (p.Val790Met), have already been associated with neuromuscular channelopathy and severe muscle weakness. CONCLUSIONS The identified candidate genes encode mainly proteins involved in electrical action potential and mechanical muscle contraction. The results suggest that the identified genetic variants may result in skeletal muscle ion channelopathies that affect muscle function, gradually leading to muscle hypotonia and weakness.
Collapse
Affiliation(s)
- Anna Sadakierska-Chudy
- Department of Genetics, Faculty of Medicine, Collegium Medicum, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705, Krakow, Poland.
| | - Paweł Szymanowski
- Department of Gynecology and Urogynecology, Faculty of Medicine, Collegium Medicum, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705, Krakow, Poland
| | - Wioletta Katarzyna Szepieniec
- Department of Gynecology and Urogynecology, Faculty of Medicine, Collegium Medicum, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705, Krakow, Poland
| | - Ewa Boniewska-Bernacka
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052, Opole, Poland
| | - Agnieszka Pollak
- Department of Medical Genetics, Warsaw Medical University, Pawinskiego 3C, 02-106, Warsaw, Poland
| |
Collapse
|
12
|
van den Bersselaar LR, Schiemann AH, Yang CY, Voermans NC, Malagon I, Scheffer GJ, Bjorksten AR, Gillies R, Hellblom A, Kamsteeg EJ, Snoeck MMJ, Stowell KM. Pathogenicity assessment of seven RYR1 variants in patients with confirmed susceptibility to malignant hyperthermia in the Netherlands. Br J Anaesth 2025:S0007-0912(24)00773-6. [PMID: 39890490 DOI: 10.1016/j.bja.2024.11.043] [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: 06/15/2024] [Revised: 10/07/2024] [Accepted: 11/02/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Malignant hyperthermia (MH) susceptibility is associated with variants in RYR1, the gene encoding the skeletal muscle ryanodine receptor-1 (RyR1), in 70-75% of patients. Functional characterisation demonstrating an increased sensitivity to RyR1 agonists is necessary among other criteria for inclusion in the European Malignant Hyperthermia Group list of MH susceptibility diagnostic variants. METHODS Seven variants in the RYR1 gene, p.Glu342Lys, p.Leu2288Ser, p.Phe2340Leu, p.Arg2676Trp, p.Val3324Ala, p.Phe4076Leu, and p.Trp5020Cys, identified in MH-susceptible individuals were introduced into the cDNA for the human RYR1 gene. These variants were tested in cultured human embryonic kidney HEK293 cells for their effect on calcium release in response to the RyR1 agonist 4-chloro-m-cresol. Calcium release of each variant was compared with wild-type and benign and pathogenic controls. Each variant was subjected to curation using the European Malignant Hyperthermia Group scoring matrix and ClinGen RYR1 Variant Curation Expert Panel guidelines. RESULTS Six of seven RYR1 variants (p.Glu342Lys, p.Leu2288Ser, p.Phe2340Leu, p.Arg2676Trp, p.Val3324Ala, p.Phe4076Leu) showed hypersensitivity to 4-chloro-m-cresol compared with wild-type. The p.Trp5020Cys variant did not release calcium in response to 4-chloro-m-cresol. All variants had minor allele frequencies <0.1%. Rare exome variant ensemble learner scores of p.Glu342Lys, p.Leu2288Ser, p.Phe4076Leu, and p.Trp5020Cys were >0.85, supporting pathogenicity. CONCLUSIONS The variants p.Glu342Lys, p.Leu2288Ser p.Phe2340Leu, and p.Arg2676Trp are pathogenic or likely pathogenic for MH and can be used for presymptomatic testing for MH susceptibility. As current knowledge on the p.Val3324Ala, p.Phe4076Leu, and p.Trp5020Cys variants remains insufficient, they are still classified as variants of uncertain significance.
Collapse
Affiliation(s)
- Luuk R van den Bersselaar
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Malignant Hyperthermia Investigation Unit, Department of Anesthesiology, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands.
| | - Anja H Schiemann
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Chu-Ya Yang
- Malignant Hyperthermia Investigation Unit, Department of Anesthesiology, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Nicol C Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ignacio Malagon
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gert-Jan Scheffer
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andrew R Bjorksten
- Malignant Hyperthermia Diagnostic Unit, Department of Anaesthesia and Pain Management, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Robyn Gillies
- Malignant Hyperthermia Diagnostic Unit, Department of Anaesthesia and Pain Management, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Anna Hellblom
- Department of Intensive and Perioperative Care, Skane University Hospital, Lund, Sweden; Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marc M J Snoeck
- Malignant Hyperthermia Investigation Unit, Department of Anesthesiology, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Kathryn M Stowell
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
| |
Collapse
|
13
|
Zhang Y, Seidel M, Rabesahala de Meritens C, Beckmann A, Ahmed S, Hurtz M, Lai FA, Zorio E, Parthimos D, Zissimopoulos S. Disparate molecular mechanisms in cardiac ryanodine receptor channelopathies. Front Mol Biosci 2024; 11:1505698. [PMID: 39777228 PMCID: PMC11703740 DOI: 10.3389/fmolb.2024.1505698] [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: 10/03/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Aims Mutations in the cardiac ryanodine receptor (RyR2) are associated with catecholaminergic polymorphic ventricular tachycardia (CPVT). This study investigates the underlying molecular mechanisms for CPVT mutations within the RyR2 N-terminus domain (NTD). Methods and Results We consulted the high-resolution RyR2 structure in both open and closed configuration to identify mutations G357S/R407I and A77T, which lie within the NTD intra- and inter-subunit interface with the Core Solenoid (CSol), respectively. Their structural and functional roles were compared to R169L, a mutation that lies within the NTD-NTD inter-subunit interface. Using chemical cross-linking and co-immunoprecipitation assays, we show that R169L disrupts NTD tetramerization, while it does not alter the NTD-CSol interaction. Single cell Ca2+ imaging revealed that R169L increases the number of spontaneous Ca2+ transients and the proportion of oscillating cells, while reducing the Ca2+ store content. G357S and R407I do not affect NTD tetramerization, but they also do not alter the NTD-CSol interaction. Functionally, RyR2G357S-expressing cells have Ca2+ handling properties similar to RyR2WT. A77T enhances the NTD-CSol interaction, while it does not affect NTD tetramerization. Like R169L, A77T also increases the number of spontaneous Ca2+ transients and the proportion of oscillating cells, and it reduces the Ca2+ store content. However, unlike R169L that displays Ca2+ transients of normal amplitude and shorter duration, Ca2+ transients for A77T are of smaller amplitude and normal duration. Conclusion The NTD-CSol inter-subunit interface variant, A77T, produces a hyperactive channel by altering a different structure-function parameter to other CPVT mutations within the RyR2 NTD. Reduced NTD-NTD inter-subunit interaction and reinforced NTD inter-subunit interaction with CSol are distinct molecular mechanisms for gain-of-function RyR2 arrhythmogenic mutations.
Collapse
Affiliation(s)
- Yadan Zhang
- Swansea University Medical School, Institute of Life Science, Swansea, United Kingdom
| | - Monika Seidel
- Swansea University Medical School, Institute of Life Science, Swansea, United Kingdom
| | | | - Astrid Beckmann
- Swansea University Medical School, Institute of Life Science, Swansea, United Kingdom
| | - Syeda Ahmed
- Swansea University Medical School, Institute of Life Science, Swansea, United Kingdom
| | - Melanie Hurtz
- Swansea University Medical School, Institute of Life Science, Swansea, United Kingdom
| | - F. Anthony Lai
- College of Medicine and Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Esther Zorio
- Inherited Cardiac Disease Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Medicine Department, Universitat de València, Valencia, Spain
- Research group CB16/11/00261, Center for Biomedical Network Research on Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Dimitris Parthimos
- School of Medicine, Division of Cancer and Genetics, Cardiff University, Cardiff, United Kingdom
| | - Spyros Zissimopoulos
- Swansea University Medical School, Institute of Life Science, Swansea, United Kingdom
| |
Collapse
|
14
|
Montalvo-Alonso JJ, Munilla C, Garriga-Alonso L, Ferragut C, Valadés D, Gonzalo-Encabo P, Pérez-López A. Acute Co-Ingestion of Caffeine and Sodium Bicarbonate on Muscular Endurance Performance. Nutrients 2024; 16:4382. [PMID: 39771003 PMCID: PMC11677328 DOI: 10.3390/nu16244382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Caffeine and sodium bicarbonate individually enhance muscular endurance by delaying fatigue, but their combined effects have scarcely been studied. Objectives: This study aimed to evaluate the acute effects of co-ingesting caffeine and sodium bicarbonate on muscular endurance at different loads in bench press and back squat exercises. Methods: Twenty-seven recreationally trained participants (female/male: 14/14; age: 23 ± 3.6 years) were randomized to four conditions in a double-blind, crossover design: (a) sodium bicarbonate and caffeine (NaHCO3 + CAF); (b) sodium bicarbonate (NaHCO3); (c) caffeine (CAF); (d) placebo (PLA); ingesting 0.3 g/kg NaHCO3, 3 mg/kg caffeine or placebo (maltodextrin). Participants performed two muscle endurance tests on bench press and back squat exercises at 65% and 85% 1RM, performing as many repetitions as possible in one set until task failure. Results: CAF increased the number of repetitions (p < 0.001; ηp2 = 0.111), mean velocity (Vmean, p = 0.043, ηp2 = 0.16), and mean power output (Wmean, p = 0.034, ηp2 = 0.15) compared to placebo. These effects were observed in back squat exercise at 65%1RM in Vmean (3.7%, p = 0.050, g = 1.144) and Wmean (5.2%, p = 0.047, g = 0.986) and at 85%1RM in Vmean (5.4%, p = 0.043, g = 0.22) and Wmean (5.5%, p = 0.050, g = 0.25). No ergogenic effects were found in NaHCO3 + CAF) or NaHCO3 conditions. Conclusions: CAF increased muscular endurance performance in male and female participants by increasing the number of repetitions, mean velocity, and power output; however, when NaHCO3 was ingested, these effects were not detected.
Collapse
|
15
|
Steinz MM, Beard N, Shorter E, Lanner JT. Stable oxidative posttranslational modifications alter the gating properties of RyR1. J Gen Physiol 2024; 156:e202313515. [PMID: 39499505 PMCID: PMC11540854 DOI: 10.1085/jgp.202313515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/03/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024] Open
Abstract
The ryanodine receptor type 1 (RyR1) is a Ca2+ release channel that regulates skeletal muscle contraction by controlling Ca2+ release from the sarcoplasmic reticulum (SR). Posttranslational modifications (PTMs) of RyR1, such as phosphorylation, S-nitrosylation, and carbonylation are known to increase RyR1 open probability (Po), contributing to SR Ca2+ leak and skeletal muscle dysfunction. PTMs on RyR1 have been linked to muscle dysfunction in diseases like breast cancer, rheumatoid arthritis, Duchenne muscle dystrophy, and aging. While reactive oxygen species (ROS) and oxidative stress induce PTMs, the impact of stable oxidative modifications like 3-nitrotyrosine (3-NT) and malondialdehyde adducts (MDA) on RyR1 gating remains unclear. Mass spectrometry and single-channel recordings were used to study how 3-NT and MDA modify RyR1 and affect Po. Both modifications increased Po in a dose-dependent manner, with mass spectrometry identifying 30 modified residues out of 5035 amino acids per RyR1 monomer. Key modifications were found in domains critical for protein interaction and channel activation, including Y808/3NT in SPRY1, Y1081/3NT and H1254/MDA in SPRY2&3, and Q2107/MDA and Y2128/3NT in JSol, near the binding site of FKBP12. Though these modifications did not directly overlap with FKBP12 binding residues, they promoted FKBP12 dissociation from RyR1. These findings provide detailed insights into how stable oxidative PTMs on RyR1 residues alter channel gating, advancing our understanding of RyR1-mediated Ca2+ release in conditions associated with oxidative stress and muscle weakness.
Collapse
Affiliation(s)
- Maarten M. Steinz
- Department of Physiology and Pharmacology, Molecular Muscle Physiology and Pathophysiology lab, Karolinska Institutet, Stockholm, Sweden
| | - Nicole Beard
- Faculty or Science and Technology, University of Canberra, Canberra, Australia
| | - Emily Shorter
- Department of Physiology and Pharmacology, Molecular Muscle Physiology and Pathophysiology lab, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T. Lanner
- Department of Physiology and Pharmacology, Molecular Muscle Physiology and Pathophysiology lab, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
16
|
Pfeil-Gardiner O, Rosa HVD, Riedel D, Chen YS, Lörks D, Kükelhan P, Linck M, Müller H, Van Petegem F, Murphy BJ. Elemental mapping in single-particle reconstructions by reconstructed electron energy-loss analysis. Nat Methods 2024; 21:2299-2306. [PMID: 39448878 PMCID: PMC11621030 DOI: 10.1038/s41592-024-02482-5] [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: 01/16/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
For macromolecular structures determined by cryogenic electron microscopy, no technique currently exists for mapping elements to defined locations, leading to errors in the assignment of metals and other ions, cofactors, substrates, inhibitors and lipids that play essential roles in activity and regulation. Elemental mapping in the electron microscope is well established for dose-tolerant samples but is challenging for biological samples, especially in a cryo-preserved state. Here we combine electron energy-loss spectroscopy with single-particle image processing to allow elemental mapping in cryo-preserved macromolecular complexes. Proof-of-principle data show that our method, reconstructed electron energy-loss (REEL) analysis, allows a three-dimensional reconstruction of electron energy-loss spectroscopy data, such that a high total electron dose is accumulated across many copies of a complex. Working with two test samples, we demonstrate that we can reliably localize abundant elements. We discuss the current limitations of the method and potential future developments.
Collapse
Affiliation(s)
- Olivia Pfeil-Gardiner
- Redox and Metalloprotein Research Group, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Higor Vinícius Dias Rosa
- Redox and Metalloprotein Research Group, Max Planck Institute of Biophysics, Frankfurt, Germany
- Mattei Lab, Structural and Computational Biology Unit, EMBL Heidelberg, Heidelberg, Germany
| | - Dietmar Riedel
- Facility for Electron Microscopy, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Yu Seby Chen
- The University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | - Filip Van Petegem
- The University of British Columbia, Vancouver, British Columbia, Canada
| | - Bonnie J Murphy
- Redox and Metalloprotein Research Group, Max Planck Institute of Biophysics, Frankfurt, Germany.
| |
Collapse
|
17
|
Fernández-Morales JC, Toth N, Bayram P, Rienzo T, Morad M. Loss-of-function W4645R mutation in the RyR2-caffeine binding site: implications for synchrony and arrhythmogenesis. Cell Calcium 2024; 123:102925. [PMID: 38908063 PMCID: PMC11392648 DOI: 10.1016/j.ceca.2024.102925] [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: 02/17/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/24/2024]
Abstract
AIMS Previous studies have identified RyR2 W4645R mutation, located in the caffeine-binding site, to associate with CPVT1 pathology. Caffeine binding to its site is thought to displace the carboxyl-terminal domain to Ca2+-binding, allowing the tryptophan residue (W4645) to regulate Ca2+ sensitivity of RyR2. To gain insights into regulation of RyR2 Ca2+-binding and its interaction with caffeine-binding site, we introduced W4645R-RyR2 point mutation via CRISPR/Cas9 gene-editing in human induced pluripotent stem cell-derived cardiomyocytes (hiPSCCMs) and characterized their Ca2+-signaling phenotype compared to WT hiPSCCMs. METHODS AND RESULTS W4645R-RyR2 cardiomyocytes had: (1) no significant change in ICa magnitude or voltage-dependence; (2) slightly reduced CICR; (3) altered relaxation kinetics of Ca2+-transients with no change in isoproterenol sensitivity; (4) complete loss of caffeine-triggered Ca2+ release; (5) larger SR Ca2+ leak resulting in 40 % lower SR Ca2+ content, as determined by myocytes' response to 4-CmC; (6) lower incidence of calcium sparks and asynchronous spontaneous SR Ca2+ releases. CONCLUSIONS W4645R-RyR2 mutation induces loss of caffeine-triggered SR Ca2+ release and enhances SR Ca2+ leak that underlie asynchronous spontaneous Ca2+ releases, triggering arrhythmia and impairing cardiac function.
Collapse
Affiliation(s)
| | - Noemi Toth
- Cardiac Signaling Center of MUSC, USC and Clemson University, Charleston, SC, USA
| | - Pinar Bayram
- Cardiac Signaling Center of MUSC, USC and Clemson University, Charleston, SC, USA
| | - Taylor Rienzo
- Cardiac Signaling Center of MUSC, USC and Clemson University, Charleston, SC, USA
| | - Martin Morad
- Cardiac Signaling Center of MUSC, USC and Clemson University, Charleston, SC, USA; Department of Regenerative Medicine and Cell Biology, MUSC,Charleston, SC, USA.
| |
Collapse
|
18
|
Kim K, Li H, Yuan Q, Melville Z, Zalk R, des Georges A, Frank J, Hendrickson WA, Marks AR, Clarke OB. Structural identification of the RY12 domain of RyR1 as an ADP sensor and the target of the malignant hyperthermia therapeutic dantrolene. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619409. [PMID: 39484412 PMCID: PMC11526878 DOI: 10.1101/2024.10.21.619409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Malignant hyperthermia (MH) is a life-threatening pharmacogenetic condition triggered by volatile anesthetics, which activate pathogenic RyR1 mutants. The small molecule therapeutic dantrolene has long been used to treat MH. However, the binding site and mechanism of dantrolene remain unclear. Here, we present cryo-EM structures of RyR1 bound to dantrolene and the MH trigger agent 4-chloro-m-cresol (4CmC), revealing the dantrolene and 4CmC binding sites in atomic detail. Dantrolene binds stacked with ATP or ADP in the RY12 domain at the corner of the receptor, inducing a conformational change in this domain which is allosterically coupled to pore closure. Functional analyses revealed that ATP or ADP was required for dantrolene inhibition, and a single point mutation that disrupts the peripheral ATP binding site abolished ATP/ADP-dependent dantrolene inhibition. Strikingly, in the absence of dantrolene, this site selectively binds two ADP molecules, suggesting a possible role in ATP/ADP ratio sensing.
Collapse
Affiliation(s)
- Kookjoo Kim
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Irving Institute for Clinical and Translational Research, Columbia University, New York, NY 10032, USA
| | - Huan Li
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Irving Institute for Clinical and Translational Research, Columbia University, New York, NY 10032, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zephan Melville
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ran Zalk
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Amédée des Georges
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY 10010, USA
- Pain Research Center, New York University, New York, NY 10010, USA
| | - Joachim Frank
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Wayne A. Hendrickson
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Oliver B. Clarke
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Irving Institute for Clinical and Translational Research, Columbia University, New York, NY 10032, USA
| |
Collapse
|
19
|
Lin L, Wang C, Wang W, Jiang H, Murayama T, Kobayashi T, Hadiatullah H, Chen YS, Wu S, Wang Y, Korza H, Gu Y, Zhang Y, Du J, Van Petegem F, Yuchi Z. Cryo-EM structures of ryanodine receptors and diamide insecticides reveal the mechanisms of selectivity and resistance. Nat Commun 2024; 15:9056. [PMID: 39428398 PMCID: PMC11491487 DOI: 10.1038/s41467-024-53490-0] [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: 02/07/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
The resistance of pests to common insecticides is a global issue that threatens food production worldwide. Diamide insecticides target insect ryanodine receptors (RyRs), causing uncontrolled calcium release from the sarcoplasmic and endoplasmic reticulum. Despite their high potency and species selectivity, several resistance mutations have emerged. Using a chimeric RyR (chiRyR) approach and cryo-electron microscopy (cryo-EM), we investigate how insect RyRs engage two different diamide insecticides from separate families: flubendiamide, a phthalic acid derivative, and tetraniliprole, an anthranilic compound. Both compounds target the same site in the transmembrane region of the RyR, albeit with different poses, and promote channel opening through coupling with the pore-forming domain. To explore the resistance mechanisms, we also solve two cryo-EM structures of chiRyR carrying the two most common resistance mutations, I4790M and G4946E, both alone and in complex with the diamide insecticide chlorantraniliprole. The resistance mutations perturb the local structure, directly reducing the binding affinity and altering the binding pose. Our findings elucidate the mode of action of different diamide insecticides, reveal the molecular mechanism of resistance mutations, and provide important clues for the development of novel pesticides that can bypass the resistance mutations.
Collapse
Affiliation(s)
- Lianyun Lin
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Frontiers Science Center for Synthetic Biology, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, China
| | - Changshi Wang
- Institute of Plant and Food Science, Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Wenlan Wang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Frontiers Science Center for Synthetic Biology, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, China
| | - Heng Jiang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Frontiers Science Center for Synthetic Biology, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, China
| | - Takashi Murayama
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takuya Kobayashi
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hadiatullah Hadiatullah
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Frontiers Science Center for Synthetic Biology, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, China
| | - Yu Seby Chen
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shunfan Wu
- College of Plant Protection, State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yiwen Wang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Frontiers Science Center for Synthetic Biology, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Henryk Korza
- Syngenta Jealott's Hill International Research Centre, Bracknell, Berkshire, UK
| | - Yucheng Gu
- Syngenta Jealott's Hill International Research Centre, Bracknell, Berkshire, UK
| | - Yan Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Frontiers Science Center for Synthetic Biology, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, China
| | - Jiamu Du
- Institute of Plant and Food Science, Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Filip Van Petegem
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Zhiguang Yuchi
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Frontiers Science Center for Synthetic Biology, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, China.
- Guangdong Laboratory for Lingnan Modern Agriculture (Shenzhen Branch), Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong, China.
| |
Collapse
|
20
|
Stephenson DG. Modeling the mechanism of Ca2+ release in skeletal muscle by DHPRs easing inhibition at RyR I1-sites. J Gen Physiol 2024; 156:e202213113. [PMID: 39230559 PMCID: PMC11390858 DOI: 10.1085/jgp.202213113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/30/2023] [Accepted: 07/01/2024] [Indexed: 09/05/2024] Open
Abstract
Ca2+ release from the sarcoplasmic reticulum (SR) plays a central role in excitation-contraction coupling (ECC) in skeletal muscles. However, the mechanism by which activation of the voltage-sensors/dihydropyridine receptors (DHPRs) in the membrane of the transverse tubular system leads to activation of the Ca2+-release channels/ryanodine receptors (RyRs) in the SR is not fully understood. Recent observations showing that a very small Ca2+ leak through RyR1s in mammalian skeletal muscle can markedly raise the background [Ca2+] in the junctional space (JS) above the Ca2+ level in the bulk of the cytosol indicate that there is a diffusional barrier between the JS and the cytosol at large. Here, I use a mathematical model to explore the hypothesis that a sudden rise in Ca2+ leak through DHPR-coupled RyR1s, caused by reduced inhibition at the RyR1 Ca2+/Mg2+ inhibitory I1-sites when the associated DHPRs are activated, is sufficient to enable synchronized responses that trigger a regenerative rise of Ca2+ release that remains under voltage control. In this way, the characteristic response to Ca2+ of RyR channels is key not only for the Ca2+ release mechanism in cardiac muscle and other tissues, but also for the DHPR-dependent Ca2+ release in skeletal muscle.
Collapse
Affiliation(s)
- D. George Stephenson
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Australia
| |
Collapse
|
21
|
Chen YS, Garcia-Castañeda M, Charalambous M, Rossi D, Sorrentino V, Van Petegem F. Cryo-EM investigation of ryanodine receptor type 3. Nat Commun 2024; 15:8630. [PMID: 39366997 PMCID: PMC11452665 DOI: 10.1038/s41467-024-52998-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/27/2024] [Indexed: 10/06/2024] Open
Abstract
Ryanodine Receptor isoform 3 (RyR3) is a large ion channel found in the endoplasmic reticulum membrane of many different cell types. Within the hippocampal region of the brain, it is found in dendritic spines and regulates synaptic plasticity. It controls myogenic tone in arteries and is upregulated in skeletal muscle in early development. RyR3 has a unique functional profile with a very high sensitivity to activating ligands, enabling high gain in Ca2+-induced Ca2+ release. Here we solve high-resolution cryo-EM structures of RyR3 in non-activating and activating conditions, revealing structural transitions that occur during channel opening. Addition of activating ligands yields only open channels, indicating an intrinsically high open probability under these conditions. RyR3 has reduced binding affinity to the auxiliary protein FKBP12.6 due to several sequence variations in the binding interface. We map disease-associated sequence variants and binding sites for known pharmacological agents. The N-terminal region contains ligand binding sites for a putative chloride anion and ATP, both of which are targeted by sequence variants linked to epileptic encephalopathy.
Collapse
Affiliation(s)
- Yu Seby Chen
- Department of Biochemistry and Molecular Biology, the Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Maricela Garcia-Castañeda
- Department of Biochemistry and Molecular Biology, the Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Maria Charalambous
- Department of Biochemistry and Molecular Biology, the Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, the Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
22
|
Sanbonmatsu K. Supercomputing in the biological sciences: Toward Zettascale and Yottascale simulations. Curr Opin Struct Biol 2024; 88:102889. [PMID: 39163795 DOI: 10.1016/j.sbi.2024.102889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 08/22/2024]
Abstract
Molecular simulations of biological systems tend to be significantly more compute-intensive than those in materials science and astrophysics, due to important contributions of long-range electrostatic forces and large numbers of time steps (>1E9) required. Simulations of biomolecular complexes of microseconds to milliseconds are considered state-of-the-art today. However, these time scales are miniscule in comparison to physiological time scales relevant to molecular machine activity, drug action, and elongation cycles for protein synthesis, RNA synthesis, and DNA synthesis (seconds to days). While an exascale supercomputer has simulated an entire virus for nanoseconds, this supercomputer would need to be 10 billion times faster to simulate that virus for 3 hours of physiological time, demonstrating the insatiable need for computing power. With growing interest in computational drug design from the pharmaceutical sector, the biological sciences are positioned to be an industry driver in computing.
Collapse
Affiliation(s)
- Karissa Sanbonmatsu
- Los Alamos National Laboratory, United States; New Mexico Consortium, New Mexico.
| |
Collapse
|
23
|
Li C, Willegems K, Uchański T, Pardon E, Steyaert J, Efremov RG. Rapid small-scale nanobody-assisted purification of ryanodine receptors for cryo-EM. J Biol Chem 2024; 300:107734. [PMID: 39233227 PMCID: PMC11474372 DOI: 10.1016/j.jbc.2024.107734] [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: 04/18/2024] [Revised: 07/05/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024] Open
Abstract
Ryanodine receptors (RyRs) are large Ca2+ release channels residing in the endoplasmic or sarcoplasmic reticulum membrane. Three isoforms of RyRs have been identified in mammals, the disfunction of which has been associated with a series of life-threatening diseases. The need for large amounts of native tissue or eukaryotic cell cultures limits advances in structural studies of RyRs. Here, we report a method that utilizes nanobodies to purify RyRs from only 5 mg of total protein. The purification process, from isolated membranes to cryo-EM grade protein, is achieved within 4 h on the bench, yielding protein usable for cryo-EM analysis. This is demonstrated by solving the structures of rabbit RyR1, solubilized in detergent, reconstituted into lipid nanodiscs or liposomes, and bovine RyR2 reconstituted in nanodisc, and mouse RyR2 in detergent. The reported method facilitates structural studies of RyRs directed toward drug development and is useful in cases where the amount of starting material is limited.
Collapse
Affiliation(s)
- Chenyao Li
- Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium; Structural Biology Brussels, Department of Bioengineering Sciences, VUB, Brussels, Belgium
| | - Katrien Willegems
- Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium; Structural Biology Brussels, Department of Bioengineering Sciences, VUB, Brussels, Belgium
| | - Tomasz Uchański
- Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium; Structural Biology Brussels, Department of Bioengineering Sciences, VUB, Brussels, Belgium
| | - Els Pardon
- Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium; Structural Biology Brussels, Department of Bioengineering Sciences, VUB, Brussels, Belgium
| | - Jan Steyaert
- Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium; Structural Biology Brussels, Department of Bioengineering Sciences, VUB, Brussels, Belgium
| | - Rouslan G Efremov
- Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium; Structural Biology Brussels, Department of Bioengineering Sciences, VUB, Brussels, Belgium.
| |
Collapse
|
24
|
Chen M. Building molecular model series from heterogeneous CryoEM structures using Gaussian mixture models and deep neural networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615511. [PMID: 39386715 PMCID: PMC11463374 DOI: 10.1101/2024.09.27.615511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cryogenic electron microscopy (CryoEM) produces structures of macromolecules at near-atomic resolution. However, building molecular models with good stereochemical geometry from those structures can be challenging and time-consuming, especially when many structures are obtained from datasets with conformational heterogeneity. Here we present a model refinement protocol that automatically generates series of molecular models from CryoEM datasets, which describe the dynamics of the macromolecular system and have near-perfect geometry scores.
Collapse
Affiliation(s)
- Muyuan Chen
- Division of CryoEM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA 94025, USA
| |
Collapse
|
25
|
Murayama T, Otori Y, Kurebayashi N, Yamazawa T, Oyamada H, Sakurai T, Ogawa H. Dual role of the S5 segment in type 1 ryanodine receptor channel gating. Commun Biol 2024; 7:1108. [PMID: 39294299 PMCID: PMC11411075 DOI: 10.1038/s42003-024-06787-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 08/27/2024] [Indexed: 09/20/2024] Open
Abstract
The type 1 ryanodine receptor (RyR1) is a Ca2+ release channel in the sarcoplasmic reticulum that is essential for skeletal muscle contraction. RyR1 forms a channel with six transmembrane segments, in which S5 is the fifth segment and is thought to contribute to pore formation. However, its role in channel gating remains unclear. Here, we performed a functional analysis of several disease-associated mutations in S5 and interpreted the results with respect to the published RyR1 structures to identify potential interactions associated with the mutant phenotypes. We demonstrate that S5 plays a dual role in channel gating: the cytoplasmic side interacts with S6 to reduce the channel activity, whereas the luminal side forms a rigid structural base necessary for S6 displacement in channel opening. These results deepen our understanding of the molecular mechanisms of RyR1 channel gating and provide insight into the divergent disease phenotypes caused by mutations in S5.
Collapse
Affiliation(s)
- Takashi Murayama
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan.
| | - Yuya Otori
- Department of Structural Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Nagomi Kurebayashi
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Toshiko Yamazawa
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| | - Hideto Oyamada
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Takashi Sakurai
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Haruo Ogawa
- Department of Structural Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
26
|
Lopergolo D, Gallus GN, Pieraccini G, Boscaro F, Berti G, Serni G, Volpi N, Formichi P, Bianchi S, Cassandrini D, Sorrentino V, Rossi D, Santorelli FM, De Stefano N, Malandrini A. CCDC78: Unveiling the Function of a Novel Gene Associated with Hereditary Myopathy. Cells 2024; 13:1504. [PMID: 39273074 PMCID: PMC11394131 DOI: 10.3390/cells13171504] [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: 07/21/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
CCDC78 was identified as a novel candidate gene for autosomal dominant centronuclear myopathy-4 (CNM4) approximately ten years ago. However, to date, only one family has been described, and the function of CCDC78 remains unclear. Here, we analyze for the first time a family harboring a CCDC78 nonsense mutation to better understand the role of CCDC78 in muscle. METHODS We conducted a comprehensive histopathological analysis on muscle biopsies, including immunofluorescent assays to detect multiple sarcoplasmic proteins. We examined CCDC78 transcripts and protein using WB in CCDC78-mutated muscle tissue; these analyses were also performed on muscle, lymphocytes, and fibroblasts from healthy subjects. Subsequently, we conducted RT-qPCR and transcriptome profiling through RNA-seq to evaluate changes in gene expression associated with CCDC78 dysfunction in muscle. Lastly, coimmunoprecipitation (Co-Ip) assays and mass spectrometry (LC-MS/MS) studies were carried out on extracted muscle proteins from both healthy and mutated subjects. RESULTS The histopathological features in muscle showed novel histological hallmarks, which included areas of dilated and swollen sarcoplasmic reticulum (SR). We provided evidence of nonsense-mediated mRNA decay (NMD), identified the presence of novel CCDC78 transcripts in muscle and lymphocytes, and identified 1035 muscular differentially expressed genes, including several involved in the SR. Through the Co-Ip assays and LC-MS/MS studies, we demonstrated that CCDC78 interacts with two key SR proteins: SERCA1 and CASQ1. We also observed interactions with MYH1, ACTN2, and ACTA1. CONCLUSIONS Our findings provide insight, for the first time, into the interactors and possible role of CCDC78 in skeletal muscle, locating the protein in the SR. Furthermore, our data expand on the phenotype previously associated with CCDC78 mutations, indicating potential histopathological hallmarks of the disease in human muscle. Based on our data, we can consider CCDC78 as the causative gene for CNM4.
Collapse
Affiliation(s)
- Diego Lopergolo
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Gian Nicola Gallus
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Giuseppe Pieraccini
- CISM—Mass Spectrometry Centre, University of Florence, 50139 Florence, Italy
| | - Francesca Boscaro
- CISM—Mass Spectrometry Centre, University of Florence, 50139 Florence, Italy
| | - Gianna Berti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Giovanni Serni
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Nila Volpi
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Patrizia Formichi
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Silvia Bianchi
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Denise Cassandrini
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
- Interdepartmental Program of Molecular Diagnosis and Pathogenetic Mechanisms of Rare Genetic Diseases, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
- Interdepartmental Program of Molecular Diagnosis and Pathogenetic Mechanisms of Rare Genetic Diseases, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | - Alessandro Malandrini
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- UOC Neurologia, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| |
Collapse
|
27
|
Sun QA, Grimmett ZW, Hess DT, Perez LG, Qian Z, Chaube R, Venetos NM, Plummer BN, Laurita KR, Premont RT, Stamler JS. Physiological role for S-nitrosylation of RyR1 in skeletal muscle function and development. Biochem Biophys Res Commun 2024; 723:150163. [PMID: 38820626 DOI: 10.1016/j.bbrc.2024.150163] [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: 04/17/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024]
Abstract
Excitation-contraction coupling in skeletal muscle myofibers depends upon Ca2+ release from the sarcoplasmic reticulum through the ryanodine receptor/Ca2+-release channel RyR1. The RyR1 contains ∼100 Cys thiols of which ∼30 comprise an allosteric network subject to posttranslational modification by S-nitrosylation, S-palmitoylation and S-oxidation. However, the role and function of these modifications is not understood. Although aberrant S-nitrosylation of multiple unidentified sites has been associated with dystrophic diseases, malignant hyperthermia and other myopathic syndromes, S-nitrosylation in physiological situations is reportedly specific to a single (1 of ∼100) Cys in RyR1, Cys3636 in a manner gated by pO2. Using mice expressing a form of RyR1 with a Cys3636→Ala point mutation to prevent S-nitrosylation at this site, we showed that Cys3636 was the principal target of endogenous S-nitrosylation during normal muscle function. The absence of Cys3636 S-nitrosylation suppressed stimulus-evoked Ca2+ release at physiological pO2 (at least in part by altering the regulation of RyR1 by Ca2+/calmodulin), eliminated pO2 coupling, and diminished skeletal myocyte contractility in vitro and measures of muscle strength in vivo. Furthermore, we found that abrogation of Cys3636 S-nitrosylation resulted in a developmental defect reflected in diminished myofiber diameter, altered fiber subtypes, and altered expression of genes implicated in muscle development and atrophy. Thus, our findings establish a physiological role for pO2-coupled S-nitrosylation of RyR1 in skeletal muscle contractility and development and provide foundation for future studies of RyR1 modifications in physiology and disease.
Collapse
Affiliation(s)
- Qi-An Sun
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Zachary W Grimmett
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Douglas T Hess
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Lautaro G Perez
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Zhaoxia Qian
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Ruchi Chaube
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Nicholas M Venetos
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Bradley N Plummer
- Heart and Vascular Research Center, MetroHealth Campus of Case Western Reserve University, Cleveland, OH, 44109, USA
| | - Kenneth R Laurita
- Heart and Vascular Research Center, MetroHealth Campus of Case Western Reserve University, Cleveland, OH, 44109, USA
| | - Richard T Premont
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Jonathan S Stamler
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
28
|
Arige V, Wagner LE, Malik S, Baker MR, Fan G, Serysheva II, Yule DI. Functional investigation of a putative calcium-binding site involved in the inhibition of inositol 1,4,5-trisphosphate receptor activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608318. [PMID: 39211071 PMCID: PMC11360954 DOI: 10.1101/2024.08.16.608318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
A wide variety of factors influence inositol 1,4,5-trisphosphate (IP 3 ) receptor (IP 3 R) activity resulting in modulation of intracellular Ca 2+ release. This regulation is thought to define the spatio-temporal patterns of Ca 2+ signals necessary for the appropriate activation of downstream effectors. The binding of both IP 3 and Ca 2+ are obligatory for IP 3 R channel opening, however, Ca 2+ regulates IP 3 R activity in a biphasic manner. Mutational studies have revealed that Ca 2+ binding to a high-affinity pocket formed by the ARM3 domain and linker domain promotes IP 3 R channel opening without altering the Ca 2+ dependency for channel inactivation. These data suggest a distinct low-affinity Ca 2+ binding site is responsible for the reduction in IP 3 R activity at higher [Ca 2+ ]. We determined the consequences of mutating a cluster of acidic residues in the ARM2 and central linker domain reported to coordinate Ca 2+ in cryo-EM structures of the IP 3 R type 3. This site is termed the "CD Ca 2+ binding site" and is well-conserved in all IP 3 R sub-types. We show that the CD site Ca 2+ binding mutants where the negatively charged glutamic acid residues are mutated to alanine exhibited enhanced sensitivity to IP 3 -generating agonists. Ca 2+ binding mutants displayed spontaneous elemental Ca 2+ events (Ca 2+ puffs) and the number of IP 3 -induced Ca 2+ puffs was significantly augmented in cells stably expressing Ca 2+ binding site mutants. When measured with "on-nucleus" patch clamp, the inhibitory effect of high [Ca 2+ ] on single channel-open probability (P o ) was reduced in mutant channels and this effect was dependent on [ATP]. These results indicate that Ca 2+ binding to the putative CD Ca 2+ inhibitory site facilitates the reduction in IP 3 R channel activation when cytosolic [ATP] is reduced and suggest that at higher [ATP], additional Ca 2+ binding motifs may contribute to the biphasic regulation of IP 3 -induced Ca 2+ release.
Collapse
|
29
|
Weninger G, Miotto MC, Tchagou C, Reiken S, Dridi H, Brandenburg S, Riedemann GC, Yuan Q, Liu Y, Chang A, Wronska A, Lehnart SE, Marks AR. Structural insights into the regulation of RyR1 by S100A1. Proc Natl Acad Sci U S A 2024; 121:e2400497121. [PMID: 38917010 PMCID: PMC11228480 DOI: 10.1073/pnas.2400497121] [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: 01/09/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
S100A1, a small homodimeric EF-hand Ca2+-binding protein (~21 kDa), plays an important regulatory role in Ca2+ signaling pathways involved in various biological functions including Ca2+ cycling and contractile performance in skeletal and cardiac myocytes. One key target of the S100A1 interactome is the ryanodine receptor (RyR), a huge homotetrameric Ca2+ release channel (~2.3 MDa) of the sarcoplasmic reticulum. Here, we report cryoelectron microscopy structures of S100A1 bound to RyR1, the skeletal muscle isoform, in absence and presence of Ca2+. Ca2+-free apo-S100A1 binds beneath the bridging solenoid (BSol) and forms contacts with the junctional solenoid and the shell-core linker of RyR1. Upon Ca2+-binding, S100A1 undergoes a conformational change resulting in the exposure of the hydrophobic pocket known to serve as a major interaction site of S100A1. Through interactions of the hydrophobic pocket with RyR1, Ca2+-bound S100A1 intrudes deeper into the RyR1 structure beneath BSol than the apo-form and induces sideways motions of the C-terminal BSol region toward the adjacent RyR1 protomer resulting in tighter interprotomer contacts. Interestingly, the second hydrophobic pocket of the S100A1-dimer is largely exposed at the hydrophilic surface making it prone to interactions with the local environment, suggesting that S100A1 could be involved in forming larger heterocomplexes of RyRs with other protein partners. Since S100A1 interactions stabilizing BSol are implicated in the regulation of RyR-mediated Ca2+ release, the characterization of the S100A1 binding site conserved between RyR isoforms may provide the structural basis for the development of therapeutic strategies regarding treatments of RyR-related disorders.
Collapse
Affiliation(s)
- Gunnar Weninger
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Marco C. Miotto
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Carl Tchagou
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Haikel Dridi
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Sören Brandenburg
- Department of Cardiology and Pneumology, Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, 37075Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC 2067), University of Göttingen, 37075Göttingen, Germany
| | - Gabriel C. Riedemann
- Department of Cardiology and Pneumology, Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, 37075Göttingen, Germany
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Yang Liu
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Alexander Chang
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| | - Stephan E. Lehnart
- Department of Cardiology and Pneumology, Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, 37075Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC 2067), University of Göttingen, 37075Göttingen, Germany
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY10032
| |
Collapse
|
30
|
Hu M, Feng X, Liu Q, Liu S, Huang F, Xu H. The ion channels of endomembranes. Physiol Rev 2024; 104:1335-1385. [PMID: 38451235 PMCID: PMC11381013 DOI: 10.1152/physrev.00025.2023] [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: 06/30/2023] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 03/08/2024] Open
Abstract
The endomembrane system consists of organellar membranes in the biosynthetic pathway [endoplasmic reticulum (ER), Golgi apparatus, and secretory vesicles] as well as those in the degradative pathway (early endosomes, macropinosomes, phagosomes, autophagosomes, late endosomes, and lysosomes). These endomembrane organelles/vesicles work together to synthesize, modify, package, transport, and degrade proteins, carbohydrates, and lipids, regulating the balance between cellular anabolism and catabolism. Large ion concentration gradients exist across endomembranes: Ca2+ gradients for most endomembrane organelles and H+ gradients for the acidic compartments. Ion (Na+, K+, H+, Ca2+, and Cl-) channels on the organellar membranes control ion flux in response to cellular cues, allowing rapid informational exchange between the cytosol and organelle lumen. Recent advances in organelle proteomics, organellar electrophysiology, and luminal and juxtaorganellar ion imaging have led to molecular identification and functional characterization of about two dozen endomembrane ion channels. For example, whereas IP3R1-3 channels mediate Ca2+ release from the ER in response to neurotransmitter and hormone stimulation, TRPML1-3 and TMEM175 channels mediate lysosomal Ca2+ and H+ release, respectively, in response to nutritional and trafficking cues. This review aims to summarize the current understanding of these endomembrane channels, with a focus on their subcellular localizations, ion permeation properties, gating mechanisms, cell biological functions, and disease relevance.
Collapse
Affiliation(s)
- Meiqin Hu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Xinghua Feng
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiang Liu
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Siyu Liu
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Fangqian Huang
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Haoxing Xu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
31
|
Rebbeck RT, Svensson B, Zhang J, Samsó M, Thomas DD, Bers DM, Cornea RL. Kinetics and mapping of Ca-driven calmodulin conformations on skeletal and cardiac muscle ryanodine receptors. Nat Commun 2024; 15:5120. [PMID: 38879623 PMCID: PMC11180167 DOI: 10.1038/s41467-024-48951-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 05/16/2024] [Indexed: 06/19/2024] Open
Abstract
Calmodulin transduces [Ca2+] information regulating the rhythmic Ca2+ cycling between the sarcoplasmic reticulum and cytoplasm during contraction and relaxation in cardiac and skeletal muscle. However, the structural dynamics by which calmodulin modulates the sarcoplasmic reticulum Ca2+ release channel, the ryanodine receptor, at physiologically relevant [Ca2+] is unknown. Using fluorescence lifetime FRET, we resolve different structural states of calmodulin and Ca2+-driven shifts in the conformation of calmodulin bound to ryanodine receptor. Skeletal and cardiac ryanodine receptor isoforms show different calmodulin-ryanodine receptor conformations, as well as binding and structural kinetics with 0.2-ms resolution, which reflect different functional roles of calmodulin. These FRET methods provide insight into the physiological calmodulin-ryanodine receptor structural states, revealing additional distinct structural states that complement cryo-EM models that are based on less physiological conditions. This technology will drive future studies on pathological calmodulin-ryanodine receptor interactions and dynamics with other important ryanodine receptor bound modulators.
Collapse
Affiliation(s)
- Robyn T Rebbeck
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, USA.
| | - Bengt Svensson
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jingyan Zhang
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Montserrat Samsó
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Donald M Bers
- Department of Pharmacology, University of California at Davis, Davis, CA, USA
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
32
|
Taweechat P, Boonamnaj P, Samsó M, Sompornpisut P. Significance of Zn 2+ in RyR1 for Structural Integrity and Ligand Binding: Insight from Molecular Dynamics. J Phys Chem B 2024; 128:4670-4684. [PMID: 38717304 PMCID: PMC11103704 DOI: 10.1021/acs.jpcb.4c01189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024]
Abstract
Ryanodine receptor type 1 (RyR1) is a Ca2+-release channel central to skeletal muscle excitation-contraction (EC) coupling. RyR1's cryo-EM structures reveal a zinc-finger motif positioned within the cytoplasmic C-terminal domain (CTD). Yet, owing to limitations in cryo-EM resolution, RyR1 structures lack precision in detailing the metal coordination structure, prompting the need for an accurate model. In this study, we employed molecular dynamics (MD) simulations and the density functional theory (DFT) method to refine the binding characteristics of Zn2+ in the zinc-finger site of the RyR1 channel. Our findings also highlight substantial conformational changes in simulations conducted in the absence of Zn2+. Notably, we observed a loss of contact at the interface between protein domains proximal to the zinc-finger site, indicating a crucial role of Zn2+ in maintaining structural integrity and interdomain interactions within RyR1. Furthermore, this study provides valuable insights into the modulation of ATP, Ca2+, and caffeine binding, shedding light on the intricate relationship between Zn2+ coordination and the dynamic behavior of RyR1. Our integrative approach combining MD simulations and DFT calculations enhances our understanding of the molecular mechanisms governing ligand binding in RyR1.
Collapse
Affiliation(s)
- Panyakorn Taweechat
- Center
of Excellence in Computational Chemistry, Department of Chemistry,
Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panisak Boonamnaj
- Center
of Excellence in Computational Chemistry, Department of Chemistry,
Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Montserrat Samsó
- Department
of Physiology and Biophysics, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Pornthep Sompornpisut
- Center
of Excellence in Computational Chemistry, Department of Chemistry,
Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
33
|
Nayak AR, Rangubpit W, Will AH, Hu Y, Castro-Hartmann P, Lobo JJ, Dryden K, Lamb GD, Sompornpisut P, Samsó M. Interplay between Mg 2+ and Ca 2+ at multiple sites of the ryanodine receptor. Nat Commun 2024; 15:4115. [PMID: 38750013 PMCID: PMC11096358 DOI: 10.1038/s41467-024-48292-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 04/26/2024] [Indexed: 05/18/2024] Open
Abstract
RyR1 is an intracellular Ca2+ channel important in excitable cells such as neurons and muscle fibers. Ca2+ activates it at low concentrations and inhibits it at high concentrations. Mg2+ is the main physiological RyR1 inhibitor, an effect that is overridden upon activation. Despite the significance of Mg2+-mediated inhibition, the molecular-level mechanisms remain unclear. In this work we determined two cryo-EM structures of RyR1 with Mg2+ up to 2.8 Å resolution, identifying multiple Mg2+ binding sites. Mg2+ inhibits at the known Ca2+ activating site and we propose that the EF hand domain is an inhibitory divalent cation sensor. Both divalent cations bind to ATP within a crevice, contributing to the precise transmission of allosteric changes within the enormous channel protein. Notably, Mg2+ inhibits RyR1 by interacting with the gating helices as validated by molecular dynamics. This structural insight enhances our understanding of how Mg2+ inhibition is overcome during excitation.
Collapse
Affiliation(s)
- Ashok R Nayak
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Warin Rangubpit
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Alex H Will
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Yifan Hu
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Pablo Castro-Hartmann
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
- ThermoFisher Scientific, Cambridge, UK
| | - Joshua J Lobo
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Kelly Dryden
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Department of Chemistry and Biochemistry, UC Santa Barbara, Santa Barbara, CA, USA
| | - Graham D Lamb
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
| | - Pornthep Sompornpisut
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand.
| | - Montserrat Samsó
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
34
|
Benucci S, Ruiz A, Franchini M, Ruggiero L, Zoppi D, Sitsapesan R, Lindsay C, Pelczar P, Pietrangelo L, Protasi F, Treves S, Zorzato F. A novel, patient-derived RyR1 mutation impairs muscle function and calcium homeostasis in mice. J Gen Physiol 2024; 156:e202313486. [PMID: 38445312 PMCID: PMC10911087 DOI: 10.1085/jgp.202313486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/20/2023] [Accepted: 02/09/2024] [Indexed: 03/07/2024] Open
Abstract
RYR1 is the most commonly mutated gene associated with congenital myopathies, a group of early-onset neuromuscular conditions of variable severity. The functional effects of a number of dominant RYR1 mutations have been established; however, for recessive mutations, these effects may depend on multiple factors, such as the formation of a hypomorphic allele, or on whether they are homozygous or compound heterozygous. Here, we functionally characterize a new transgenic mouse model knocked-in for mutations identified in a severely affected child born preterm and presenting limited limb movement. The child carried the homozygous c.14928C>G RYR1 mutation, resulting in the p.F4976L substitution. In vivo and ex vivo assays revealed that homozygous mice fatigued sooner and their muscles generated significantly less force compared with their WT or heterozygous littermates. Electron microscopy, biochemical, and physiological analyses showed that muscles from RyR1 p.F4976L homozygous mice have the following properties: (1) contain fewer calcium release units and show areas of myofibrillar degeneration, (2) contain less RyR1 protein, (3) fibers show smaller electrically evoked calcium transients, and (4) their SR has smaller calcium stores. In addition, single-channel recordings indicate that RyR1 p.F4976L exhibits higher Po in the presence of 100 μM [Ca2+]. Our mouse model partly recapitulates the clinical picture of the homozygous human patient and provides significant insight into the functional impact of this mutation. These results will help understand the pathology of patients with similar RYR1 mutations.
Collapse
Affiliation(s)
- Sofia Benucci
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Alexis Ruiz
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Martina Franchini
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Lucia Ruggiero
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Dario Zoppi
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | | | - Chris Lindsay
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | - Laura Pietrangelo
- DMSI, Department of Medicine and Aging Sciences and CAST, Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Feliciano Protasi
- DMSI, Department of Medicine and Aging Sciences and CAST, Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Susan Treves
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesco Zorzato
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
35
|
Xu J, Liao C, Yin CC, Li G, Zhu Y, Sun F. In situ structural insights into the excitation-contraction coupling mechanism of skeletal muscle. SCIENCE ADVANCES 2024; 10:eadl1126. [PMID: 38507485 PMCID: PMC10954225 DOI: 10.1126/sciadv.adl1126] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/12/2024] [Indexed: 03/22/2024]
Abstract
Excitation-contraction coupling (ECC) is a fundamental mechanism in control of skeletal muscle contraction and occurs at triad junctions, where dihydropyridine receptors (DHPRs) on transverse tubules sense excitation signals and then cause calcium release from the sarcoplasmic reticulum via coupling to type 1 ryanodine receptors (RyR1s), inducing the subsequent contraction of muscle filaments. However, the molecular mechanism remains unclear due to the lack of structural details. Here, we explored the architecture of triad junction by cryo-electron tomography, solved the in situ structure of RyR1 in complex with FKBP12 and calmodulin with the resolution of 16.7 Angstrom, and found the intact RyR1-DHPR supercomplex. RyR1s arrange into two rows on the terminal cisternae membrane by forming right-hand corner-to-corner contacts, and tetrads of DHPRs bind to RyR1s in an alternating manner, forming another two rows on the transverse tubule membrane. This unique arrangement is important for synergistic calcium release and provides direct evidence of physical coupling in ECC.
Collapse
Affiliation(s)
- Jiashu Xu
- Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenyi Liao
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Chang-Cheng Yin
- Department of Biophysics, The Health Science Center, Peking University, Beijing 100191, China
- Electron Microscopy Analysis Laboratory, The Health Science Center, Peking University, Beijing 100191, China
- Center for Protein Science, Peking University, Beijing 100871, China
| | - Guohui Li
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yun Zhu
- Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Sun
- Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, 510005, China
| |
Collapse
|
36
|
Xia Y, Zhang XH, Yamaguchi N, Morad M. Point mutations in RyR2 Ca2+-binding residues of human cardiomyocytes cause cellular remodelling of cardiac excitation contraction-coupling. Cardiovasc Res 2024; 120:44-55. [PMID: 37890099 PMCID: PMC10898933 DOI: 10.1093/cvr/cvad163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/17/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
AIMS CRISPR/Cas9 gene edits of cardiac ryanodine receptor (RyR2) in human-induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) provide a novel platform for introducing mutations in RyR2 Ca2+-binding residues and examining the resulting excitation contraction (EC)-coupling remodelling consequences. METHODS AND RESULTS Ca2+-signalling phenotypes of mutations in RyR2 Ca2+-binding site residues associated with cardiac arrhythmia (RyR2-Q3925E) or not proven to cause cardiac pathology (RyR2-E3848A) were determined using ICa- and caffeine-triggered Ca2+ releases in voltage-clamped and total internal reflection fluorescence-imaged wild type and mutant cardiomyocytes infected with sarcoplasmic reticulum (SR)-targeted ER-GCaMP6 probe. (i) ICa- and caffeine-triggered Fura-2 or ER-GCaMP6 signals were suppressed, even when ICa was significantly enhanced in Q3925E and E3848A mutant cardiomyocytes; (ii) spontaneous beating (Fura-2 Ca2+ transients) persisted in mutant cells without the SR-release signals; (iii) while 5-20 mM caffeine failed to trigger Ca2+-release in voltage-clamped mutant cells, only ∼20% to ∼70% of intact myocytes responded respectively to caffeine; (iv) and 20 mM caffeine transients, however, activated slowly, were delayed, and variably suppressed by 2-APB, FCCP, or ruthenium red. CONCLUSION Mutating RyR2 Ca2+-binding residues, irrespective of their reported pathogenesis, suppressed both ICa- and caffeine-triggered Ca2+ releases, suggesting interaction between Ca2+- and caffeine-binding sites. Enhanced transmembrane calcium influx and remodelling of EC-coupling pathways may underlie the persistence of spontaneous beating in Ca2+-induced Ca2+ release-suppressed mutant myocytes.
Collapse
Affiliation(s)
- Yanli Xia
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, 68 President Street, Bioengineering building Rm 306, Charleston, SC 29425, USA
| | - Xiao-hua Zhang
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, 68 President Street, Bioengineering building Rm 306, Charleston, SC 29425, USA
| | - Naohiro Yamaguchi
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, 68 President Street, Bioengineering building Rm 306, Charleston, SC 29425, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 68 President Street, Bioengineering building Rm 306, Charleston, SC 29425, USA
| | - Martin Morad
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, 68 President Street, Bioengineering building Rm 306, Charleston, SC 29425, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 68 President Street, Bioengineering building Rm 306, Charleston, SC 29425, USA
| |
Collapse
|
37
|
Farrell RJ, Bredvik KG, Hoppa MB, Hennigan ST, Brown TA, Ryan TA. A ratiometric ER calcium sensor for quantitative comparisons across cell types and subcellular regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580492. [PMID: 38405980 PMCID: PMC10888930 DOI: 10.1101/2024.02.15.580492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The endoplasmic reticulum (ER) is an important regulator of Ca 2 + in cells and dysregulation of ER calcium homeostasis can lead to numerous pathologies. Understanding how various pharmacological and genetic perturbations of ER Ca 2 + homeostasis impacts cellular physiology would likely be facilitated by more quantitative measurements of ER Ca 2 + levels that allow easier comparisons across conditions. Here, we developed a ratiometric version of our original ER-GCaMP probe that allows for more quantitative comparisons of the concentration of Ca 2 + in the ER across cell types and sub-cellular compartments. Using this approach we show that the resting concentration of ER Ca2+ in primary dissociated neurons is substantially lower than that in measured in embryonic fibroblasts.
Collapse
Affiliation(s)
- Ryan J Farrell
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- David Rockefeller Graduate Program, The Rockefeller University, New York, NY, USA, 10065
- Present Address: Neuroscience Institute, NYU Medical Center, New York, NY, USA, 10016
| | - Kirsten G Bredvik
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- Tri-Institutional M.D./Ph.D. Program, Weill Cornell Medicine, New York, NY, USA, 10065
| | - Michael B Hoppa
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- Present Address: Department of Biology, Dartmouth College, Hanover, NH 03755
| | - S Thomas Hennigan
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, 20147
| | - Timothy A Brown
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, 20147
| | - Timothy A Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA, 10065
- Lead Contact: correspondence
| |
Collapse
|
38
|
Wegener JW, Mitronova GY, ElShareif L, Quentin C, Belov V, Pochechueva T, Hasenfuss G, Ackermann L, Lehnart SE. A dual-targeted drug inhibits cardiac ryanodine receptor Ca 2+ leak but activates SERCA2a Ca 2+ uptake. Life Sci Alliance 2024; 7:e202302278. [PMID: 38012000 PMCID: PMC10681910 DOI: 10.26508/lsa.202302278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
In the heart, genetic or acquired mishandling of diastolic [Ca2+] by ryanodine receptor type 2 (RyR2) overactivity correlates with risks of arrhythmia and sudden cardiac death. Strategies to avoid these risks include decrease of Ca2+ release by drugs modulating RyR2 activity or increase in Ca2+ uptake by drugs modulating SR Ca2+ ATPase (SERCA2a) activity. Here, we combine these strategies by developing experimental compounds that act simultaneously on both processes. Our screening efforts identified the new 1,4-benzothiazepine derivative GM1869 as a promising compound. Consequently, we comparatively studied the effects of the known RyR2 modulators Dantrolene and S36 together with GM1869 on RyR2 and SERCA2a activity in cardiomyocytes from wild type and arrhythmia-susceptible RyR2R2474S/+ mice by confocal live-cell imaging. All drugs reduced RyR2-mediated Ca2+ spark frequency but only GM1869 accelerated SERCA2a-mediated decay of Ca2+ transients in murine and human cardiomyocytes. Our data indicate that S36 and GM1869 are more suitable than dantrolene to directly modulate RyR2 activity, especially in RyR2R2474S/+ mice. Remarkably, GM1869 may represent a new dual-acting lead compound for maintenance of diastolic [Ca2+].
Collapse
Affiliation(s)
- Jörg W Wegener
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Gyuzel Y Mitronova
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Lina ElShareif
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
| | - Christine Quentin
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Vladimir Belov
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Tatiana Pochechueva
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Lutz Ackermann
- Georg-August University of Göttingen, Institute of Organic and Biomolecular Chemistry, Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Stephan E Lehnart
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center of Göttingen (UMG), Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
39
|
Chirasani VR, Elferdink M, Kral M, Carter JS, Heitmann S, Meissner G, Yamaguchi N. Structural and functional interactions between the EF hand domain and S2-S3 loop in the type-1 ryanodine receptor ion channel. J Biol Chem 2024; 300:105606. [PMID: 38159862 PMCID: PMC10832476 DOI: 10.1016/j.jbc.2023.105606] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024] Open
Abstract
Previous cryo-electron micrographs suggested that the skeletal muscle Ca2+ release channel, ryanodine receptor (RyR)1, is regulated by intricate interactions between the EF hand Ca2+ binding domain and the cytosolic loop (S2-S3 loop). However, the precise molecular details of these interactions and functional consequences of the interactions remain elusive. Here, we used molecular dynamics simulations to explore the specific amino acid pairs involved in hydrogen bond interactions within the EF hand-S2-S3 loop interface. Our simulations unveiled two key interactions: (1) K4101 (EF hand) with D4730 (S2-S3 loop) and (2) E4075, Q4078, and D4079 (EF hand) with R4736 (S2-S3 loop). To probe the functional significance of these interactions, we constructed mutant RyR1 complementary DNAs and expressed them in HEK293 cells for [3H]ryanodine binding assays. Our results demonstrated that mutations in the EF hand, specifically K4101E and K4101M, resulted in reduced affinities for Ca2+/Mg2+-dependent inhibitions. Interestingly, the K4101E mutation increased the affinity for Ca2+-dependent activation. Conversely, mutations in the S2-S3 loop, D4730K and D4730N, did not significantly change the affinities for Ca2+/Mg2+-dependent inhibitions. Our previous finding that skeletal disease-associated RyR1 mutations, R4736Q and R4736W, impaired Ca2+-dependent inhibition, is consistent with the current results. In silico mutagenesis analysis aligned with our functional data, indicating altered hydrogen bonding patterns upon mutations. Taken together, our findings emphasize the critical role of the EF hand-S2-S3 loop interaction in Ca2+/Mg2+-dependent inhibition of RyR1 and provide insights into potential therapeutic strategies targeting this domain interaction for the treatment of skeletal myopathies.
Collapse
Affiliation(s)
- Venkat R Chirasani
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; R.L. Juliano Structural Bioinformatics Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Millar Elferdink
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA; Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, Charleston, South Carolina, USA; College of Charleston Honors College, Charleston, South Carolina, USA
| | - MacKenzie Kral
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA; Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, Charleston, South Carolina, USA; College of Charleston Honors College, Charleston, South Carolina, USA
| | - Jordan S Carter
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA; Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, Charleston, South Carolina, USA
| | - Savannah Heitmann
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA; Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, Charleston, South Carolina, USA
| | - Gerhard Meissner
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Naohiro Yamaguchi
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA; Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, Charleston, South Carolina, USA.
| |
Collapse
|
40
|
Joseph TT, Bu W, Haji-Ghassemi O, Chen YS, Woll K, Allen PD, Brannigan G, van Petegem F, Eckenhoff RG. Propofol directly binds and inhibits skeletal muscle ryanodine receptor 1 (RyR1). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575040. [PMID: 38260485 PMCID: PMC10802444 DOI: 10.1101/2024.01.10.575040] [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
As the primary Ca 2+ release channel in skeletal muscle sarcoplasmic reticulum (SR), mutations in the type 1 ryanodine receptor (RyR1) or its binding partners underlie a constellation of muscle disorders, including malignant hyperthermia (MH). In patients with MH mutations, exposure to triggering drugs such as the halogenated volatile anesthetics biases RyR1 to an open state, resulting in uncontrolled Ca 2+ release, sarcomere tension and heat production. Restoration of Ca 2+ into the SR also consumes ATP, generating a further untenable metabolic load. When anesthetizing patients with known MH mutations, the non-triggering intravenous general anesthetic propofol is commonly substituted for triggering anesthetics. Evidence of direct binding of anesthetic agents to RyR1 or its binding partners is scant, and the atomic-level interactions of propofol with RyR1 are entirely unknown. Here, we show that propofol decreases RyR1 opening in heavy SR vesicles and planar lipid bilayers, and that it inhibits activator-induced Ca 2+ release from SR in human skeletal muscle. In addition to confirming direct binding, photoaffinity labeling using m- azipropofol (AziP m ) revealed several putative propofol binding sites on RyR1. Prediction of binding affinity by molecular dynamics simulation suggests that propofol binds at least one of these sites at clinical concentrations. These findings invite the hypothesis that in addition to propofol not triggering MH, it may also be protective against MH by inhibiting induced Ca 2+ flux through RyR1.
Collapse
|
41
|
Abstract
Changes in the intracellular concentration of free calcium (Ca2+) underpin egg activation and initiation of development in animals and plants. In mammals, the Ca2+ release is periodical, known as Ca2+ oscillations, and mediated by the type 1 inositol 1,4,5-trisphosphate receptor (IP3R1). Another divalent cation, zinc (Zn2+), increases exponentially during oocyte maturation and is vital for meiotic transitions, arrests, and polyspermy prevention. It is unknown if these pivotal cations interplay during fertilization. Here, using mouse eggs, we showed that basal concentrations of labile Zn2+ are indispensable for sperm-initiated Ca2+ oscillations because Zn2+-deficient conditions induced by cell-permeable chelators abrogated Ca2+ responses evoked by fertilization and other physiological and pharmacological agonists. We also found that chemically or genetically generated eggs with lower levels of labile Zn2+ displayed reduced IP3R1 sensitivity and diminished ER Ca2+ leak despite the stable content of the stores and IP3R1 mass. Resupplying Zn2+ restarted Ca2+ oscillations, but excessive Zn2+ prevented and terminated them, hindering IP3R1 responsiveness. The findings suggest that a window of Zn2+ concentrations is required for Ca2+ responses and IP3R1 function in eggs, ensuring optimal response to fertilization and egg activation.
Collapse
Affiliation(s)
- Hiroki Akizawa
- Department of Veterinary and Animal Sciences, University of Massachusetts AmherstAmherstUnited States
| | - Emily M Lopes
- Department of Veterinary and Animal Sciences, University of Massachusetts AmherstAmherstUnited States
- Molecular and Cellular Biology Graduate Program, University of MassachusettsAmherstUnited States
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts AmherstAmherstUnited States
| |
Collapse
|
42
|
Mitronova GY, Quentin C, Belov VN, Wegener JW, Kiszka KA, Lehnart SE. 1,4-Benzothiazepines with Cyclopropanol Groups and Their Structural Analogues Exhibit Both RyR2-Stabilizing and SERCA2a-Stimulating Activities. J Med Chem 2023; 66:15761-15775. [PMID: 37991191 PMCID: PMC10726367 DOI: 10.1021/acs.jmedchem.3c01235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023]
Abstract
To discover new multifunctional agents for the treatment of cardiovascular diseases, we designed and synthesized a series of compounds with a cyclopropyl alcohol moiety and evaluated them in biochemical assays. Biological screening identified derivatives with dual activity: preventing Ca2+ leak through ryanodine receptor 2 (RyR2) and enhancing cardiac sarco-endoplasmic reticulum (SR) Ca2+ load by activation of Ca2+-dependent ATPase 2a (SERCA2a). The compounds that stabilize RyR2 at micro- and nanomolar concentrations are either structurally related to RyR-stabilizing drugs or Rycals or have structures similar to them. The novel compounds also demonstrate a good ability to increase ATP hydrolysis mediated by SERCA2a activity in cardiac microsomes, e.g., the half-maximal effective concentration (EC50) was as low as 383 nM for compound 12a, which is 1,4-benzothiazepine with two cyclopropanol groups. Our findings indicate that these derivatives can be considered as new lead compounds to improve cardiac function in heart failure.
Collapse
Affiliation(s)
- Gyuzel Y. Mitronova
- Department
of NanoBiophotonics, Max Planck Institute
for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
- German
Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen 37075, Germany
| | - Christine Quentin
- Department
of NanoBiophotonics, Max Planck Institute
for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - Vladimir N. Belov
- Department
of NanoBiophotonics, Max Planck Institute
for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - Jörg W. Wegener
- Department
of Cardiology & Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Strasse 42a, Göttingen 37075, Germany
- German
Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen 37075, Germany
| | - Kamila A. Kiszka
- Department
of NanoBiophotonics, Max Planck Institute
for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | - Stephan E. Lehnart
- Department
of Cardiology & Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Robert-Koch-Strasse 42a, Göttingen 37075, Germany
- German
Centre for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen 37075, Germany
| |
Collapse
|
43
|
Song X, Kirtipal N, Lee S, Malý P, Bharadwaj S. Current therapeutic targets and multifaceted physiological impacts of caffeine. Phytother Res 2023; 37:5558-5598. [PMID: 37679309 DOI: 10.1002/ptr.8000] [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: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
Caffeine, which shares consubstantial structural similarity with purine adenosine, has been demonstrated as a nonselective adenosine receptor antagonist for eliciting most of the biological functions at physiologically relevant dosages. Accumulating evidence supports caffeine's beneficial effects against different disorders, such as total cardiovascular diseases and type 2 diabetes. Conversely, paradoxical effects are also linked to caffeine ingestion in humans including hypertension-hypotension and tachycardia-bradycardia. These observations suggest the association of caffeine action with its ingested concentration and/or concurrent interaction with preferential molecular targets to direct explicit events in the human body. Thus, a coherent analysis of the functional targets of caffeine, relevant to normal physiology, and disease pathophysiology, is required to understand the pharmacology of caffeine. This review provides a broad overview of the experimentally validated targets of caffeine, particularly those of therapeutic interest, and the impacts of caffeine on organ-specific physiology and pathophysiology. Overall, the available empirical and epidemiological evidence supports the dose-dependent functional activities of caffeine and advocates for further studies to get insights into the caffeine-induced changes under specific conditions, such as asthma, DNA repair, and cancer, in view of its therapeutic applications.
Collapse
Affiliation(s)
- Xinjie Song
- Zhejiang Provincial Key Lab for Chemical and Biological Processing Technology of Farm Product, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| | - Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| |
Collapse
|
44
|
Noda Y, Miyoshi H, Benucci S, Gonzalez A, Bandschapp O, Girard T, Treves S, Zorzato F. Functional characterization of RYR1 variants identified in malignant hyperthermia susceptible individuals. Neuromuscul Disord 2023; 33:951-963. [PMID: 37996280 DOI: 10.1016/j.nmd.2023.10.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023]
Abstract
Malignant hyperthermia is a pharmacogenetic disorder triggered by halogenated anesthetic agents in genetically predisposed individuals. Approximately 70 % of these individuals carry mutations in RYR1, the gene encoding the ryanodine receptor calcium channel of skeletal muscle. In this study, we performed functional analysis of 5 RYR1 variants identified in members from 8 families who had been diagnosed by the IVCT. Of the 68 individuals enrolled in the study, 43 were diagnosed as MHS, 23 as MHN, and 2 individuals were not tested. Here we demonstrate that the 5 RyR1 variants cause hypersensitivity to RyR1 agonist-mediated calcium release. According to the EMHG scoring matrix these five genetic variants can be classified as follows: c.8638G>A (p.E2880K) and c.11314C>T (p.R3772W) likely pathogenic, c.11416G>A (p.G3806R), c.14627A>G (p.K4876R) and c.14813T>C (p.I4938T), pathogenic (RefSeq NM_000540.3). We propose that the newly functionally characterized RYR1 variants, be included in the panel of variants to be used for the molecular diagnosis of MHS.
Collapse
Affiliation(s)
- Yuko Noda
- Departments of Biomedicine and Neurology, Basel University Hospital, Hebelstrasse 20, Basel 4031, Switzerland; Department of Anesthesiology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku Hiroshima, 734-8551, Japan
| | - Hirotsugu Miyoshi
- Department of Anesthesiology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku Hiroshima, 734-8551, Japan
| | - Sofia Benucci
- Departments of Biomedicine and Neurology, Basel University Hospital, Hebelstrasse 20, Basel 4031, Switzerland
| | | | | | - Thierry Girard
- Anesthesiology, Spitalstrasse 21, Basel 4031, Switzerland
| | - Susan Treves
- Departments of Biomedicine and Neurology, Basel University Hospital, Hebelstrasse 20, Basel 4031, Switzerland; Department of Life Science and Biotechnology, University of Ferrara, Via Borsari 46, Ferrara 44100, Italy.
| | - Francesco Zorzato
- Departments of Biomedicine and Neurology, Basel University Hospital, Hebelstrasse 20, Basel 4031, Switzerland; Department of Life Science and Biotechnology, University of Ferrara, Via Borsari 46, Ferrara 44100, Italy.
| |
Collapse
|
45
|
Akizawa H, Lopes E, Fissore RA. Zn 2+ is Essential for Ca 2+ Oscillations in Mouse Eggs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.13.536745. [PMID: 37131581 PMCID: PMC10153198 DOI: 10.1101/2023.04.13.536745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Changes in the intracellular concentration of free calcium (Ca2+) underpin egg activation and initiation of development in animals and plants. In mammals, the Ca2+ release is periodical, known as Ca2+ oscillations, and mediated by the type 1 inositol 1,4,5-trisphosphate receptor (IP3R1). Another divalent cation, zinc (Zn2+), increases exponentially during oocyte maturation and is vital for meiotic transitions, arrests, and polyspermy prevention. It is unknown if these pivotal cations interplay during fertilization. Here, using mouse eggs, we showed that basal concentrations of labile Zn2+ are indispensable for sperm-initiated Ca2+ oscillations because Zn2+-deficient conditions induced by cell-permeable chelators abrogated Ca2+ responses evoked by fertilization and other physiological and pharmacological agonists. We also found that chemically- or genetically generated eggs with lower levels of labile Zn2+ displayed reduced IP3R1 sensitivity and diminished ER Ca2+ leak despite the stable content of the stores and IP3R1 mass. Resupplying Zn2+ restarted Ca2+ oscillations, but excessive Zn2+ prevented and terminated them, hindering IP3R1 responsiveness. The findings suggest that a window of Zn2+ concentrations is required for Ca2+ responses and IP3R1 function in eggs, ensuring optimal response to fertilization and egg activation.
Collapse
Affiliation(s)
- Hiroki Akizawa
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, Massachusetts, 01003, United States
| | - Emily Lopes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, Massachusetts, 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, 01003, United States
| | - Rafael A. Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant Street, Amherst, Massachusetts, 01003, United States
| |
Collapse
|
46
|
Gong Y, Ge L, Li Q, Gong J, Chen M, Gao H, Kang J, Yu T, Li J, Xu H. Ethanol Causes Cell Death and Neuronal Differentiation Defect During Initial Neurogenesis of the Neural Retina by Disrupting Calcium Signaling in Human Retinal Organoids. Stem Cell Rev Rep 2023; 19:2790-2806. [PMID: 37603136 DOI: 10.1007/s12015-023-10604-3] [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] [Accepted: 07/31/2023] [Indexed: 08/22/2023]
Abstract
Fetal Alcohol Syndrome (FAS) affects a significant proportion, exceeding 90%, of afflicted children, leading to severe ocular aberrations such as microphthalmia and optic nerve hypoplasia. During the early stages of pregnancy, the commencement of neural retina neurogenesis represents a critical period for human eye development, concurrently exposing the developing retinal structures to the highest risk of prenatal ethanol exposure due to a lack of awareness. Despite the paramount importance of this period, the precise influence and underlying mechanisms of short-term ethanol exposure on the developmental process of the human neural retina have remained largely elusive. In this study, we utilize the human embryonic stem cells derived retinal organoids (hROs) to recapitulate the initial retinal neurogenesis and find that 1% (v/v) ethanol slows the growth of hROs by inducing robust cell death and retinal ganglion cell differentiation defect. Bulk RNA-seq analysis and two-photon microscope live calcium imaging reveal altered calcium signaling dynamics derived from ethanol-induced down-regulation of RYR1 and CACNA1S. Moreover, the calcium-binding protein RET, one of the downstream effector genes of the calcium signaling pathway, synergistically integrates ethanol and calcium signals to abort neuron differentiation and cause cell death. To sum up, our study illustrates the effect and molecular mechanism of ethanol on the initial neurogenesis of the human embryonic neural retina, providing a novel interpretation of the ocular phenotype of FAS and potentially informing preventative measures for susceptible populations.
Collapse
Affiliation(s)
- Yu Gong
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Lingling Ge
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Qiyou Li
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Jing Gong
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Min Chen
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Hui Gao
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Jiahui Kang
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Ting Yu
- Department of Clinical Laboratory, The 89th Hospital of The People's Liberation Army, Weifang, People's Republic of China
| | - Jiawen Li
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, Chongqing, China.
| | - Haiwei Xu
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China.
| |
Collapse
|
47
|
Paknejad N, Sapuru V, Hite RK. Structural titration reveals Ca 2+-dependent conformational landscape of the IP 3 receptor. Nat Commun 2023; 14:6897. [PMID: 37898605 PMCID: PMC10613215 DOI: 10.1038/s41467-023-42707-3] [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: 02/16/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are endoplasmic reticulum Ca2+ channels whose biphasic dependence on cytosolic Ca2+ gives rise to Ca2+ oscillations that regulate fertilization, cell division and cell death. Despite the critical roles of IP3R-mediated Ca2+ responses, the structural underpinnings of the biphasic Ca2+ dependence that underlies Ca2+ oscillations are incompletely understood. Here, we collect cryo-EM images of an IP3R with Ca2+ concentrations spanning five orders of magnitude. Unbiased image analysis reveals that Ca2+ binding does not explicitly induce conformational changes but rather biases a complex conformational landscape consisting of resting, preactivated, activated, and inhibited states. Using particle counts as a proxy for relative conformational free energy, we demonstrate that Ca2+ binding at a high-affinity site allows IP3Rs to activate by escaping a low-energy resting state through an ensemble of preactivated states. At high Ca2+ concentrations, IP3Rs preferentially enter an inhibited state stabilized by a second, low-affinity Ca2+ binding site. Together, these studies provide a mechanistic basis for the biphasic Ca2+-dependence of IP3R channel activity.
Collapse
Affiliation(s)
- Navid Paknejad
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Physiology, Biophysics, and Systems Biology (PBSB) Program, Weill Cornell Graduate School of Biomedical Sciences, 1300 York Avenue, New York, NY, 10065, USA
| | - Vinay Sapuru
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Physiology, Biophysics, and Systems Biology (PBSB) Program, Weill Cornell Graduate School of Biomedical Sciences, 1300 York Avenue, New York, NY, 10065, USA
| | - Richard K Hite
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
48
|
Šeflová J, Schwarz JA, Smith AN, Svensson B, Blackwell DJ, Phillips TA, Nikolaienko R, Bovo E, Rebbeck RT, Zima AV, Thomas DD, Van Petegem F, Knollmann BC, Johnston JN, Robia SL, Cornea RL. RyR2 Binding of an Antiarrhythmic Cyclic Depsipeptide Mapped Using Confocal Fluorescence Lifetime Detection of FRET. ACS Chem Biol 2023; 18:2290-2299. [PMID: 37769131 PMCID: PMC11648969 DOI: 10.1021/acschembio.3c00376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Hyperactivity of cardiac sarcoplasmic reticulum (SR) ryanodine receptor (RyR2) Ca2+-release channels contributes to heart failure and arrhythmias. Reducing the RyR2 activity, particularly during cardiac relaxation (diastole), is a desirable therapeutic goal. We previously reported that the unnatural enantiomer (ent) of an insect-RyR activator, verticilide, inhibits porcine and mouse RyR2 at diastolic (nanomolar) Ca2+ and has in vivo efficacy against atrial and ventricular arrhythmia. To determine the ent-verticilide structural mode of action on RyR2 and guide its further development via medicinal chemistry structure-activity relationship studies, here, we used fluorescence lifetime (FLT)-measurements of Förster resonance energy transfer (FRET) in HEK293 cells expressing human RyR2. For these studies, we used an RyR-specific FRET molecular-toolkit and computational methods for trilateration (i.e., using distances to locate a point of interest). Multiexponential analysis of FLT-FRET measurements between four donor-labeled FKBP12.6 variants and acceptor-labeled ent-verticilide yielded distance relationships placing the acceptor probe at two candidate loci within the RyR2 cryo-EM map. One locus is within the Ry12 domain (at the corner periphery of the RyR2 tetrameric complex). The other locus is sandwiched at the interface between helical domain 1 and the SPRY3 domain. These findings document RyR2-target engagement by ent-verticilide, reveal new insight into the mechanism of action of this new class of RyR2-targeting drug candidate, and can serve as input in future computational determinations of the ent-verticilide binding site on RyR2 that will inform structure-activity studies for lead optimization.
Collapse
Affiliation(s)
- Jaroslava Šeflová
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois 60153, United States
| | - Jacob A Schwarz
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Abigail N Smith
- Department of Chemistry & Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Bengt Svensson
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Daniel J Blackwell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Taylor A Phillips
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois 60153, United States
| | - Roman Nikolaienko
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois 60153, United States
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois 60153, United States
| | - Robyn T Rebbeck
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois 60153, United States
| | - David D Thomas
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Björn C Knollmann
- Department of Chemistry & Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jeffrey N Johnston
- Department of Chemistry & Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois 60153, United States
| | - Răzvan L Cornea
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
49
|
Raisch T, Raunser S. The modes of action of ion-channel-targeting neurotoxic insecticides: lessons from structural biology. Nat Struct Mol Biol 2023; 30:1411-1427. [PMID: 37845413 DOI: 10.1038/s41594-023-01113-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 08/31/2023] [Indexed: 10/18/2023]
Abstract
Insecticides are indispensable tools for plant protection in modern agriculture. Despite having highly heterogeneous structures, many neurotoxic insecticides use similar principles to inhibit or deregulate neuronal ion channels. Insecticides targeting pentameric ligand-gated channels are structural mimetics of neurotransmitters or manipulate and deregulate the proteins. Those binding to (pseudo-)tetrameric voltage-gated(-like) channels, on the other hand, are natural or synthetic compounds that directly block the ion-conducting pore or prevent conformational changes in the transmembrane domain necessary for opening and closing the pore. The use of a limited number of inhibition mechanisms can be problematic when resistances arise and become more widespread. Therefore, there is a rising interest in the development of insecticides with novel mechanisms that evade resistance and are pest-insect-specific. During the last decade, most known insecticide targets, many with bound compounds, have been structurally characterized, bringing the rational design of novel classes of agrochemicals within closer reach than ever before.
Collapse
Affiliation(s)
- Tobias Raisch
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| |
Collapse
|
50
|
Dridi H, Santulli G, Bahlouli L, Miotto MC, Weninger G, Marks AR. Mitochondrial Calcium Overload Plays a Causal Role in Oxidative Stress in the Failing Heart. Biomolecules 2023; 13:1409. [PMID: 37759809 PMCID: PMC10527470 DOI: 10.3390/biom13091409] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Heart failure is a serious global health challenge, affecting more than 6.2 million people in the United States and is projected to reach over 8 million by 2030. Independent of etiology, failing hearts share common features, including defective calcium (Ca2+) handling, mitochondrial Ca2+ overload, and oxidative stress. In cardiomyocytes, Ca2+ not only regulates excitation-contraction coupling, but also mitochondrial metabolism and oxidative stress signaling, thereby controlling the function and actual destiny of the cell. Understanding the mechanisms of mitochondrial Ca2+ uptake and the molecular pathways involved in the regulation of increased mitochondrial Ca2+ influx is an ongoing challenge in order to identify novel therapeutic targets to alleviate the burden of heart failure. In this review, we discuss the mechanisms underlying altered mitochondrial Ca2+ handling in heart failure and the potential therapeutic strategies.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Laith Bahlouli
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Marco C. Miotto
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Gunnar Weninger
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| |
Collapse
|