1
|
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
|
2
|
Wang S, Qiao ST, Li PZ, Xie Y, Guo FR, Liu JW, Hu WK, Gao MY, Zheng LJ, Yang FX, Yuchi ZG, Wu SF, Bass C, Gao CF. Y4667D Mutation in the Ryanodine Receptor Confers High Level Resistance to Diamide Insecticides in the Rice Stem Borer, Chilo suppressalis Walker (Lepidoptera: Crambidae). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9920-9931. [PMID: 40198889 DOI: 10.1021/acs.jafc.5c00470] [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: 04/10/2025]
Abstract
Chilo suppressalis is a major rice pest with escalating resistance to diamide insecticides, threatening sustainable management. However, the precise molecular mechanisms underlying this resistance remain unclear. In this study, we assessed the sensitivity of 71 C. suppressalis field populations in China to chlorantraniliprole during 2023-2024 and investigated target-site mutations associated with resistance. The proportion of highly resistant populations increased to 80% in 2023 (RR = 111.6-2706.4) and 90.3% in 2024 (RR = 160-1794.7). Multiple RyR mutations, including Y4667D, were identified in highly resistant populations. Introgressing the Y4667D mutation into a laboratory strain generated the 4667D strain, which exhibited high resistance to chlorantraniliprole and other diamides. Resistance showed autosomal inheritance with incomplete dominance. Modeling and molecular docking revealed that Y4667D reduced CsRyR binding affinity for chlorantraniliprole. Furthermore, Y4667D conferred significant fitness costs such as longer larval duration and reduced reproductive output. These findings provide insights into the molecular mechanisms of diamide resistance, inform pesticide management strategies, and aid the development of novel resistance-breaking pesticides.
Collapse
Affiliation(s)
- Shuai Wang
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Song-Tao Qiao
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Pei-Zhuo Li
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency; Collaborative Innovation Center of Chemical Science and Engineering; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yuan Xie
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Fang-Rui Guo
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Jin-Wei Liu
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Wen-Kai Hu
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Meng-Yue Gao
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Ling-Jun Zheng
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Feng-Xia Yang
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Zhi-Guang Yuchi
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency; Collaborative Innovation Center of Chemical Science and Engineering; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Shun-Fan Wu
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| | - Chris Bass
- Centre for Ecology and Conservation, University of Exeter, Penryn TR10 9FE, U.K
| | - Cong-Fen Gao
- College of Plant Protection, State and Local Joint Engineering Research Center of Green Pesticide Invention and Application, Nanjing Agricultural University, Weigang Road 1, Nanjing 210095, Jiangsu, China
| |
Collapse
|
3
|
Yuan S, Kuai Z, Zhao F, Xu D, Wu W. Improving effect of physical exercise on heart failure: Reducing oxidative stress-induced inflammation by restoring Ca 2+ homeostasis. Mol Cell Biochem 2025; 480:2471-2486. [PMID: 39365389 DOI: 10.1007/s11010-024-05124-8] [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: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024]
Abstract
Heart failure (HF) is associated with the occurrence of mitochondrial dysfunction. ATP produced by mitochondria through the tricarboxylic acid cycle is the main source of energy for the heart. Excessive release of Ca2+ from myocardial sarcoplasmic reticulum (SR) in HF leads to excessive Ca2+ entering mitochondria, which leads to mitochondrial dysfunction and REDOX imbalance. Excessive accumulation of ROS leads to mitochondrial structure damage, which cannot produce and provide energy. In addition, the accumulation of a large number of ROS can activate NF-κB, leading to myocardial inflammation. Energy deficit in the myocardium has long been considered to be the main mechanism connecting mitochondrial dysfunction and systolic failure. However, exercise can improve the Ca2+ imbalance in HF and restore the Ca2+ disorder in mitochondria. Similarly, exercise activates mitochondrial dynamics to improve mitochondrial function and reshape intact mitochondrial structure, rebalance mitochondrial REDOX, reduce excessive release of ROS, and rescue cardiomyocyte energy failure in HF. In this review, we summarize recent evidence that exercise can improve Ca2+ homeostasis in the SR and activate mitochondrial dynamics, improve mitochondrial function, and reduce oxidative stress levels in HF patients, thereby reducing chronic inflammation in HF patients. The improvement of mitochondrial dynamics is beneficial for ameliorating metabolic flow bottlenecks, REDOX imbalance, ROS balance, impaired mitochondrial Ca2+ homeostasis, and inflammation. Interpretation of these findings will lead to new approaches to disease mechanisms and treatment.
Collapse
Affiliation(s)
- Shunling Yuan
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
| | - Zhongkai Kuai
- Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, China
| | - Fei Zhao
- Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, China.
| | - Diqun Xu
- School of Physical Education, Minnan Normal University, Zhangzhou, China.
| | - Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, China.
| |
Collapse
|
4
|
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
|
5
|
Zhang J, Treinen LM, Mast SJ, McCarthy MR, Svensson B, Thomas DD, Cornea RL. Kinetics insight into the roles of the N- and C-lobes of calmodulin in RyR1 channel regulation. J Biol Chem 2025; 301:108258. [PMID: 39904484 PMCID: PMC11923823 DOI: 10.1016/j.jbc.2025.108258] [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: 01/31/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 02/06/2025] Open
Abstract
Calmodulin (CaM) activates the skeletal muscle Ca2+ release channel (ryanodine receptor, RyR1) at nanomolar Ca2+ and inhibits it at micromolar Ca2+. CaM conversion from RyR1 activator to inhibitor is due to structural changes induced by Ca2+ binding at CaM's two lobes. However, it remains unclear which lobe provides the switch for this conversion. Here, we attached the environment-sensitive fluorophore acrylodan (Acr) at either lobe of intact CaM or lobe-specific Ca2+-sensitive CaM mutants, and monitored the effects of Ca2+ binding via the fluorescence change of free or RyR1-bound AcrCaM. Using steady state measurements, we found that Ca2+ binding to free CaM causes a dramatic structural change in the N-lobe, but only a slight effect on the C-lobe of the Ca2+-sensitive lobe-specific mutants, in addition to the previously known higher Ca2+ affinity at the C-lobe versus the N-lobe. Using stopped-flow measurements, we found ∼30x faster Ca2+ dissociation from the N- versus C-lobe, and ∼20x slower Ca2+ association to the N-lobe versus C-lobe. These Ca2+ binding properties hold for the CaM/RyR1 complex, and Ca2+ affinity is enhanced at the CaM C-lobe but decreased at the N-lobe by RyR1 binding. We propose that fast Ca2+-binding at the C-lobe of CaM initiates its inhibition to RyR1 at high [Ca2+], while slow Ca2+ binding to the N-lobe is necessary for timely enhancement of the inhibitory effect. The dysregulation of RyR1 by M124Q-CaM may be explained by the lower Ca2+ affinity versus WT-CaM, as suggested by both steady-state and transient kinetics results.
Collapse
Affiliation(s)
- Jingyan Zhang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, USA
| | - Levy M Treinen
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, USA
| | - Skylar J Mast
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, USA
| | - Megan R McCarthy
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, USA
| | - Bengt Svensson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, USA
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, USA.
| |
Collapse
|
6
|
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
|
7
|
Richardson SJ, Thekkedam CG, Casarotto MG, Beard NA, Dulhunty AF. Complex Actions of FKBP12 on RyR1 Ion Channel Activity Consistent with Negative Co-Operativity in FKBP12 Binding to the RyR1 Tetramer. Cells 2025; 14:157. [PMID: 39936949 PMCID: PMC11817637 DOI: 10.3390/cells14030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
The association of the 12 KDa FK506 binding protein (FKBP12) with ryanodine receptor type 1 (RyR1) in skeletal muscle is thought to suppress RyR1 channel opening and contribute to healthy muscle function. The strongest evidence for this role is increased RyR1 channel activity following FKBP12 dissociation. However, the corollary that channel activity will decrease when FKBP12 is added back to FKBP12-depleted RyR1 is not well established, and when reported, the time- and concentration-dependence of inhibition vary over orders of magnitude. Here, we address this problem with an investigation of the molecular mechanisms of the FKBP12 regulation of RyR1. Muscle processing to obtain sarcoplasmic reticulum (SR) vesicle preparations enriched in RyR1 resulted in substantial FKBP12 dissociation from RyR1, indicating low-affinity binding. Conversely, high-affinity binding was indicated by some FKBP12 remaining bound to RyR1 after solubilization. We report, for the first time, an increase in the activity of FKBP12-depleted channels after the addition of exogenous FKBP12 (5 nM to 5 µM), followed by a reduction in activity consistent with inhibition after 20-30 min exposure to higher [FKBP12]s. Both the increase and later decline in activity were time- and concentration-dependent. The results suggest a high-affinity activation when FKBP12 binding sites on the RyR1 tetramer are partially occupied by FKBP12 and lower affinity inhibition as more RyR1 monomers become occupied. These novel results imply negative cooperativity in FKBP12 binding to RyR1 and a dynamic role for FKBP12/RyR1 interactions in intact muscle fibers.
Collapse
Affiliation(s)
| | - Chris G. Thekkedam
- Developmental and Regeneration Biology Laboratory, Victor Chang Cardiac Research Institute, 405 Liverpool St, Darlinghurst, NSW 2010, Australia;
| | - Marco G. Casarotto
- Biomolecular Interactions Group, Research School of Biology, Australian National University, Canberra, ACT 2601, Australia;
| | - Nicole A. Beard
- Muscle Proteomics Group, Centre for Research in Therapeutic Solutions, University of Canberra, Bruce, ACT 2617, Australia;
| | - Angela F. Dulhunty
- Muscle Research Group, Eccles Institute of Neuroscience, John Curtin School of Mecical Research, Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
8
|
García-Puga M, Gerenu G, Bargiela A, Espinosa-Espinosa J, Mosqueira-Martín L, Sagartzazu-Aizpurua M, Aizpurua JM, Vallejo-Illarramendi A, Artero R, López de Munain A, Matheu A. A Novel Class of FKBP12 Ligands Rescues Premature Aging Phenotypes Associated with Myotonic Dystrophy Type 1. Cells 2024; 13:1939. [PMID: 39682688 PMCID: PMC11639790 DOI: 10.3390/cells13231939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Myotonic dystrophy type 1 (DM1) is an autosomal dominant disorder clinically characterized by progressive muscular weakness and multisystem degeneration, which correlates with the size of CTG expansion and MBLN decrease. These changes induce a calcium and redox homeostasis imbalance in several models that recapitulate the features of premature tissue aging. In this study, we characterized the impact of a new family of FKBP12 ligands (generically named MPs or MP compounds) designed to stabilize FKBP12 binding to the ryanodine receptors and normalize calcium dysregulation under oxidative stress. Methods: Human primary fibroblasts from DM1 patients and control donors, treated with MP compounds or not, were used for functional studies of cell viability, proliferation, and metabolism. The gene expression profile in treated cells was determined using RNA sequencing. The impact of MP compounds in vivo was evaluated in a Drosophila model of the disease using locomotor activity and longevity studies. Results: The treatment with different MP compounds reversed oxidative stress and impaired cell viability and proliferation, mitochondrial activity, and metabolic defects in DM1-derived primary fibroblasts. RNA sequencing analysis confirmed the restoration of molecular pathways related to calcium and redox homeostasis and additional pathways, including the cell cycle and metabolism. This analysis also revealed the rescue of alternative splicing events in DM1 fibroblasts treated with MP compounds. Importantly, treatment with MP compounds significantly extended the lifespan and improved the locomotor activity of a Drosophila model of the DM1 disease, and restored molecular defects characteristic of the disease in vivo. Conclusions: Our results revealed that MP compounds rescue multiple premature aging phenotypes described in DM1 models and decipher the benefits of this new family of compounds in the pre-clinical setting of DM1.
Collapse
Affiliation(s)
- Mikel García-Puga
- Cellular Oncology Group, Biogipuzkoa Health Research Institute, Paseo Dr. Beguiristain s/n, 20014 San Sebastian, Spain;
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- CIBERNED, CIBER, Carlos III Institute, 28029 Madrid, Spain
- Department of Health Sciences, Public University of Navarre (UPNA), Health Sciences Campus, Avda. de Barañain s/n, 31008 Pamplona, Spain
| | - Gorka Gerenu
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- CIBERNED, CIBER, Carlos III Institute, 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Ariadna Bargiela
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Spain; (A.B.); (J.E.-E.); (R.A.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- CIBERER, CIBER, Carlos III Institute, 28029 Madrid, Spain
| | - Jorge Espinosa-Espinosa
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Spain; (A.B.); (J.E.-E.); (R.A.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- CIBERER, CIBER, Carlos III Institute, 28029 Madrid, Spain
| | - Laura Mosqueira-Martín
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country, 20014 San Sebastian, Spain
| | - Maialen Sagartzazu-Aizpurua
- Joxe Mari Korta R&D Center, Department of Organic Chemistry I, University of the Basque Country, 20014 San Sebastian, Spain; (M.S.-A.); (J.M.A.)
| | - Jesús M. Aizpurua
- Joxe Mari Korta R&D Center, Department of Organic Chemistry I, University of the Basque Country, 20014 San Sebastian, Spain; (M.S.-A.); (J.M.A.)
| | - Ainara Vallejo-Illarramendi
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- CIBERNED, CIBER, Carlos III Institute, 28029 Madrid, Spain
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country, 20014 San Sebastian, Spain
| | - Rubén Artero
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Spain; (A.B.); (J.E.-E.); (R.A.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- CIBERER, CIBER, Carlos III Institute, 28029 Madrid, Spain
| | - Adolfo López de Munain
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- CIBERNED, CIBER, Carlos III Institute, 28029 Madrid, Spain
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country, 20014 San Sebastian, Spain
- Neurology Department, Donostia University Hospital, Osakidetza, 20014 San Sebastian, Spain
- Department of Internal Medicine, Faculty of Health Sciences, University of Deusto, 48007 Bilbao, Spain
| | - Ander Matheu
- Cellular Oncology Group, Biogipuzkoa Health Research Institute, Paseo Dr. Beguiristain s/n, 20014 San Sebastian, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- CIBERFES, CIBER, Carlos III Institute, 28029 Madrid, Spain
| |
Collapse
|
9
|
Huang J, Pan X, Yan N. Structural biology and molecular pharmacology of voltage-gated ion channels. Nat Rev Mol Cell Biol 2024; 25:904-925. [PMID: 39103479 DOI: 10.1038/s41580-024-00763-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 08/07/2024]
Abstract
Voltage-gated ion channels (VGICs), including those for Na+, Ca2+ and K+, selectively permeate ions across the cell membrane in response to changes in membrane potential, thus participating in physiological processes involving electrical signalling, such as neurotransmission, muscle contraction and hormone secretion. Aberrant function or dysregulation of VGICs is associated with a diversity of neurological, psychiatric, cardiovascular and muscular disorders, and approximately 10% of FDA-approved drugs directly target VGICs. Understanding the structure-function relationship of VGICs is crucial for our comprehension of their working mechanisms and role in diseases. In this Review, we discuss how advances in single-particle cryo-electron microscopy have afforded unprecedented structural insights into VGICs, especially on their interactions with clinical and investigational drugs. We present a comprehensive overview of the recent advances in the structural biology of VGICs, with a focus on how prototypical drugs and toxins modulate VGIC activities. We explore how these structures elucidate the molecular basis for drug actions, reveal novel pharmacological sites, and provide critical clues to future drug discovery.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Xiaojing Pan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
| | - Nieng Yan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
Wang D, Jia H, Cao H, Hou X, Wang Q, Lin J, Liu J, Yang L, Liu J. A Dual-Channel Ca 2+ Nanomodulator Induces Intracellular Ca 2+ Disorders via Endogenous Ca 2+ Redistribution for Tumor Radiosensitization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401222. [PMID: 38690593 DOI: 10.1002/adma.202401222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/21/2024] [Indexed: 05/02/2024]
Abstract
Tumor cells harness Ca2+ to maintain cellular homeostasis and withstand external stresses from various treatments. Here, a dual-channel Ca2+ nanomodulator (CAP-P-NO) is constructed that can induce irreversible intracellular Ca2+ disorders via the redistribution of tumor-inherent Ca2+ for disrupting cellular homeostasis and thus improving tumor radiosensitivity. Stimulated by tumor-overexpressed acid and glutathione, capsaicin and nitric oxide are successively escaped from CAP-P-NO to activate the transient receptor potential cation channel subfamily V member 1 and the ryanodine receptor for the influx of extracellular Ca2+ and the release of Ca2+ in the endoplasmic reticulum, respectively. The overwhelming level of Ca2+ in tumor cells not only impairs the function of organelles but also induces widespread changes in the gene transcriptome, including the downregulation of a set of radioresistance-associated genes. Combining CAP-P-NO treatment with radiotherapy achieves a significant suppression against both pancreatic and patient-derived hepatic tumors with negligible side effects. Together, the study provides a feasible approach for inducing tumor-specific intracellular Ca2+ overload via endogenous Ca2+ redistribution and demonstrates the great potential of Ca2+ disorder therapy in enhancing the sensitivity for tumor radiotherapy.
Collapse
Affiliation(s)
- Dianyu Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Haixue Jia
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Hongmei Cao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Xiaoxue Hou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Qian Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jia Lin
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jinjian Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Lijun Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
Feng Y, Wang J, Cao J, Cao F, Chen X. Manipulating calcium homeostasis with nanoplatforms for enhanced cancer therapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230019. [PMID: 38854493 PMCID: PMC10867402 DOI: 10.1002/exp.20230019] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/28/2023] [Indexed: 06/11/2024]
Abstract
Calcium ions (Ca2+) are indispensable and versatile metal ions that play a pivotal role in regulating cell metabolism, encompassing cell survival, proliferation, migration, and gene expression. Aberrant Ca2+ levels are frequently linked to cell dysfunction and a variety of pathological conditions. Therefore, it is essential to maintain Ca2+ homeostasis to coordinate body function. Disrupting the balance of Ca2+ levels has emerged as a potential therapeutic strategy for various diseases, and there has been extensive research on integrating this approach into nanoplatforms. In this review, the current nanoplatforms that regulate Ca2+ homeostasis for cancer therapy are first discussed, including both direct and indirect approaches to manage Ca2+ overload or inhibit Ca2+ signalling. Then, the applications of these nanoplatforms in targeting different cells to regulate their Ca2+ homeostasis for achieving therapeutic effects in cancer treatment are systematically introduced, including tumour cells and immune cells. Finally, perspectives on the further development of nanoplatforms for regulating Ca2+ homeostasis, identifying scientific limitations and future directions for exploitation are offered.
Collapse
Affiliation(s)
- Yanlin Feng
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Jianlin Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Jimin Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingaporeSingapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Agency for Science, Technology, and Research (A*STAR)Institute of Molecular and Cell BiologySingaporeSingapore
| |
Collapse
|
22
|
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
|
23
|
Beauvieux A, Fromentin JM, Romero D, Couffin N, Brown A, Metral L, Bourjea J, Bertile F, Schull Q. Molecular fingerprint of gilthead seabream physiology in response to pollutant mixtures in the wild. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 340:122789. [PMID: 37913978 DOI: 10.1016/j.envpol.2023.122789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/29/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
The increase in trace element concentrations in the aquatic environment due to anthropogenic activities, urges the need for their monitoring and potential toxicity, persistence, bioaccumulation, and biomagnification at different trophic levels. Gilthead seabream is a species of commercial importance in the Mediterranean Sea, both for the aquaculture and fisheries sectors, however very little is known about their trace element contamination accumulation and the resulting effect on their health status. In the present study, 135 juveniles were collected from seven coastal lagoons known to be essential nursery areas for this species. We measured seventeen different inorganic contaminants at the individual level in fish muscle (namely Al, As, Be, Bi, Cd, Cr, Cu, Hg, Li, Ni, Pb, Rb, Sb, Sr, Ti, Tl and Zn). Our results revealed the accumulation of multiple trace elements in individuals and distinct contamination signatures between lagoons which might lead to contrasted quality as nurseries for juveniles of numerous ecologically and economically relevant fish species in addition to seabreams. We further evaluated the potential adverse effect of these complex contamination mixtures on the liver (the main organ implicated in the metabolism of xenobiotics) and red muscle (a highly metabolic organ) using a proteomic approach. Alterations in cellular organization pathways and protein transport were detected in both tissues (albeit they were not similarly regulated). Chromosome organization and telomere maintenance in the liver appeared to be affected by contaminant mixture which could increase mortality, age-related disease risk and shorter lifetime expectancy for these juveniles. Red muscle proteome also demonstrated an upregulation of pathways involved in metabolism in response to contamination which raises the issue of potential energy allocation trade-offs between the organisms' main functions such as reproduction and growth. This study provides new insights into the cellular and molecular responses of seabreams to environmental pollution and proposed biomarkers of health effects of trace elements that could serve as a starting point for larger-scale biomonitoring programs.
Collapse
Affiliation(s)
| | | | - Diego Romero
- Toxicology Department, Faculty of Veterinary Medicine, University of Murcia, 30100, Murcia, Spain
| | - Nathan Couffin
- Université de Strasbourg, CNRS, IPHC UMR 7178, 23 rue du Loess, 67037, Strasbourg Cedex 2, France; Infrastructure Nationale de Protéomique ProFI, FR2048 CNRS, CEA, Strasbourg, 67087, France
| | - Adrien Brown
- Université de Strasbourg, CNRS, IPHC UMR 7178, 23 rue du Loess, 67037, Strasbourg Cedex 2, France; Infrastructure Nationale de Protéomique ProFI, FR2048 CNRS, CEA, Strasbourg, 67087, France
| | - Luisa Metral
- MARBEC, Univ Montpellier, Ifremer, IRD, CNRS, Sète, France
| | - Jérôme Bourjea
- MARBEC, Univ Montpellier, Ifremer, IRD, CNRS, Sète, France
| | - Fabrice Bertile
- Université de Strasbourg, CNRS, IPHC UMR 7178, 23 rue du Loess, 67037, Strasbourg Cedex 2, France; Infrastructure Nationale de Protéomique ProFI, FR2048 CNRS, CEA, Strasbourg, 67087, France
| | - Quentin Schull
- MARBEC, Univ Montpellier, Ifremer, IRD, CNRS, Sète, France
| |
Collapse
|
24
|
Deniaud A, Kabasakal BV, Bufton JC, Schaffitzel C. Sample Preparation for Electron Cryo-Microscopy of Macromolecular Machines. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 3234:173-190. [PMID: 38507207 DOI: 10.1007/978-3-031-52193-5_12] [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: 03/22/2024]
Abstract
High-resolution structure determination by electron cryo-microscopy underwent a step change in recent years. This now allows study of challenging samples which previously were inaccessible for structure determination, including membrane proteins. These developments shift the focus in the field to the next bottlenecks which are high-quality sample preparations. While the amounts of sample required for cryo-EM are relatively small, sample quality is the key challenge. Sample quality is influenced by the stability of complexes which depends on buffer composition, inherent flexibility of the sample, and the method of solubilization from the membrane for membrane proteins. It further depends on the choice of sample support, grid pre-treatment and cryo-grid freezing protocol. Here, we discuss various widely applicable approaches to improve sample quality for structural analysis by cryo-EM.
Collapse
Affiliation(s)
- Aurélien Deniaud
- Univ. Grenoble Alpes, CNRS, CEA, IRIG - Laboratoire de Chimie et Biologie des Métaux, Grenoble, France
| | - Burak V Kabasakal
- School of Biochemistry, University of Bristol, Bristol, UK
- Turkish Accelerator and Radiation Laboratory, Gölbaşı, Ankara, Türkiye
| | | | | |
Collapse
|
25
|
Wang W, Zhang T, Du L, Li K, Zhang L, Li H, Gao X, Xu L, Li J, Gao H. Transcriptomic analysis reveals diverse expression patterns underlying the fiber diameter of oxidative and glycolytic skeletal muscles in steers. Meat Sci 2024; 207:109350. [PMID: 37844514 DOI: 10.1016/j.meatsci.2023.109350] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 08/18/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
Skeletal muscles consist of heterogeneous fibers with various contractile and metabolic properties that affect meat quality. The size of muscle fibers contributes to muscle mass and myopathy. Thus, improved understanding of the expression patterns underlying fiber size might open possibilities to change them using genetic methods. The aim of this study was to reveal transcriptomic landscapes of one oxidative (Psoas major) and three glycolytic (Longissimus lumborum, Triceps brachii, and Semimembranosus) muscles. Principal component analysis (PCA) showed significant differences in gene expression among the four muscles. Specifically, 2777 differentially expressed genes (DEGs) were detected between six pairwise comparisons of the four muscles. Weighted gene co-expression network analysis (WGCNA) identified six modules, which were significantly associated with muscle fiber diameter. We also identified 23 candidate genes, and enrichment analysis showed that biosynthesis of amino acids (bta01230), sarcomere (GO:0030017), and regulation of actin cytoskeleton (bta04810) overlapped in DEGs and WGCNA. Nineteen of these genes (e.g., EEF1A2, FARSB, and PINK1) have been reported to promote or inhibit muscle growth and development. Our findings contribute to the understanding of fiber size differences among oxidative and glycolytic muscles, which may provide a basis for breeding to improve meat yield.
Collapse
Affiliation(s)
- Wenxiang Wang
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Tianliu Zhang
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Lili Du
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Keanning Li
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Lupei Zhang
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Haipeng Li
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Xue Gao
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Lingyang Xu
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Junya Li
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Huijiang Gao
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
26
|
Torres R, Hidalgo C. Subcellular localization and transcriptional regulation of brain ryanodine receptors. Functional implications. Cell Calcium 2023; 116:102821. [PMID: 37949035 DOI: 10.1016/j.ceca.2023.102821] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/16/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
Ryanodine receptors (RyR) are intracellular Ca2+ channels localized in the endoplasmic reticulum, where they act as critical mediators of Ca2+-induced Ca2+ calcium release (CICR). In the brain, mammals express in both neurons, and non-neuronal cells, a combination of the three RyR-isoforms (RyR1-3). Pharmacological approaches, which do not distinguish between isoforms, have indicated that RyR-isoforms contribute to brain function. However, isoform-specific manipulations have revealed that RyR-isoforms display different subcellular localizations and are differentially associated with neuronal function. These findings raise the need to understand RyR-isoform specific transcriptional regulation, as this knowledge will help to elucidate the causes of neuronal dysfunction for a growing list of brain disorders that show altered RyR channel expression and function.
Collapse
Affiliation(s)
- Rodrigo Torres
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Lago Panguipulli 1390, 5501842, Puerto Montt, Chile.
| | - Cecilia Hidalgo
- Department of Neurosciences. Biomedical Neuroscience Institute, Physiology and Biophysics Program, Institute of Biomedical Sciences, Center for Exercise, Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago, 8380000, Chile
| |
Collapse
|
27
|
Dulhunty AF. Biophysical reviews top five: voltage-dependent charge movement in nerve and muscle. Biophys Rev 2023; 15:1903-1907. [PMID: 38192339 PMCID: PMC10771356 DOI: 10.1007/s12551-023-01165-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 01/10/2024] Open
Abstract
The discovery of gating currents and asymmetric charge movement in the early 1970s represented a remarkable leap forward in our understanding of the biophysical basis of voltage-dependent events that underlie electrical signalling that is vital for nerve and muscle function. Gating currents and charge movement reflect a fundamental process in which charged amino acid residues in an ion channel protein move in response to a change in the membrane electrical field and therefore activate the specific voltage-dependent response of that protein. The detection of gating currents and asymmetric charge movement over the past 50 years has been pivotal in unraveling the multiple molecular and intra-molecular processes which lead to action potentials in excitable tissues and excitation-contraction (EC) coupling in skeletal muscle. The recording of gating currents and asymmetric charge movement remains an essential component of investigations into the basic molecular mechanisms of neuronal conduction and muscle contraction.
Collapse
Affiliation(s)
- Angela F. Dulhunty
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, ACT, Canberra, 2601 Australia
| |
Collapse
|
28
|
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
|
29
|
Liu Y, Reiken S, Dridi H, Yuan Q, Mohammad KS, Trivedi T, Miotto MC, Wedderburn-Pugh K, Sittenfeld L, Kerley Y, Meyer JA, Peters JS, Persohn SC, Bedwell AA, Figueiredo LL, Suresh S, She Y, Soni RK, Territo PR, Marks AR, Guise TA. Targeting ryanodine receptor type 2 to mitigate chemotherapy-induced neurocognitive impairments in mice. Sci Transl Med 2023; 15:eadf8977. [PMID: 37756377 DOI: 10.1126/scitranslmed.adf8977] [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: 11/18/2022] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Chemotherapy-induced cognitive dysfunction (chemobrain) is an important adverse sequela of chemotherapy. Chemobrain has been identified by the National Cancer Institute as a poorly understood problem for which current management or treatment strategies are limited or ineffective. Here, we show that chemotherapy treatment with doxorubicin (DOX) in a breast cancer mouse model induced protein kinase A (PKA) phosphorylation of the neuronal ryanodine receptor/calcium (Ca2+) channel type 2 (RyR2), RyR2 oxidation, RyR2 nitrosylation, RyR2 calstabin2 depletion, and subsequent RyR2 Ca2+ leakiness. Chemotherapy was furthermore associated with abnormalities in brain glucose metabolism and neurocognitive dysfunction in breast cancer mice. RyR2 leakiness and cognitive dysfunction could be ameliorated by treatment with a small molecule Rycal drug (S107). Chemobrain was also found in noncancer mice treated with DOX or methotrexate and 5-fluorouracil and could be prevented by treatment with S107. Genetic ablation of the RyR2 PKA phosphorylation site (RyR2-S2808A) also prevented the development of chemobrain. Chemotherapy increased brain concentrations of the tumor necrosis factor-α and transforming growth factor-β signaling, suggesting that increased inflammatory signaling might contribute to oxidation-driven biochemical remodeling of RyR2. Proteomics and Gene Ontology analysis indicated that the signaling downstream of chemotherapy-induced leaky RyR2 was linked to the dysregulation of synaptic structure-associated proteins that are involved in neurotransmission. Together, our study points to neuronal Ca2+ dyshomeostasis via leaky RyR2 channels as a potential mechanism contributing to chemobrain, warranting further translational studies.
Collapse
Affiliation(s)
- Yang Liu
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Khalid S Mohammad
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Present address: College of Medicine, Alfaisal University, Box 50927, Riyadh 1153, Kingdom of Saudi Arabia
| | - Trupti Trivedi
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marco C Miotto
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Kaylee Wedderburn-Pugh
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Leah Sittenfeld
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Ynez Kerley
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jill A Meyer
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jonathan S Peters
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Scott C Persohn
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Amanda A Bedwell
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lucas L Figueiredo
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sukanya Suresh
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yun She
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Paul R Territo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Theresa A Guise
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
30
|
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
|
31
|
Dridi H, Liu Y, Reiken S, Liu X, Argyrousi EK, Yuan Q, Miotto MC, Sittenfeld L, Meddar A, Soni RK, Arancio O, Lacampagne A, Marks AR. Heart failure-induced cognitive dysfunction is mediated by intracellular Ca 2+ leak through ryanodine receptor type 2. Nat Neurosci 2023; 26:1365-1378. [PMID: 37429912 PMCID: PMC10400432 DOI: 10.1038/s41593-023-01377-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/12/2023] [Indexed: 07/12/2023]
Abstract
Cognitive dysfunction (CD) in heart failure (HF) adversely affects treatment compliance and quality of life. Although ryanodine receptor type 2 (RyR2) has been linked to cardiac muscle dysfunction, its role in CD in HF remains unclear. Here, we show in hippocampal neurons from individuals and mice with HF that the RyR2/intracellular Ca2+ release channels were subjected to post-translational modification (PTM) and were leaky. RyR2 PTM included protein kinase A phosphorylation, oxidation, nitrosylation and depletion of the stabilizing subunit calstabin2. RyR2 PTM was caused by hyper-adrenergic signaling and activation of the transforming growth factor-beta pathway. HF mice treated with a RyR2 stabilizer drug (S107), beta blocker (propranolol) or transforming growth factor-beta inhibitor (SD-208), or genetically engineered mice resistant to RyR2 Ca2+ leak (RyR2-p.Ser2808Ala), were protected against HF-induced CD. Taken together, we propose that HF is a systemic illness driven by intracellular Ca2+ leak that includes cardiogenic dementia.
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, USA.
| | - Yang Liu
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Xiaoping Liu
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Elentina K Argyrousi
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - 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, USA
| | | | | | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Alain Lacampagne
- PHYMEDEXP, University of Montpellier, CNRS, INSERM, CHU Montpellier, Montpellier, France
- LIA1185 CNRS, Montpellier, France
| | - 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, USA.
| |
Collapse
|
32
|
Wu M, Wu C, Song T, Pan K, Wang Y, Liu Z. Structure and transport mechanism of the human calcium pump SPCA1. Cell Res 2023; 33:533-545. [PMID: 37258749 PMCID: PMC10313705 DOI: 10.1038/s41422-023-00827-x] [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: 02/14/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023] Open
Abstract
Secretory-pathway Ca2+-ATPases (SPCAs) play critical roles in maintaining Ca2+ homeostasis, but the exact mechanism of SPCAs-mediated Ca2+ transport remains unclear. Here, we determined six cryo-electron microscopy (cryo-EM) structures of human SPCA1 (hSPCA1) in a series of intermediate states, revealing a near-complete conformational cycle. With the aid of molecular dynamics simulations, these structures offer a clear structural basis for Ca2+ entry and release in hSPCA1. We found that hSPCA1 undergoes unique conformational changes during ATP binding and phosphorylation compared to other well-studied P-type II ATPases. In addition, we observed a conformational distortion of the Ca2+-binding site induced by the separation of transmembrane helices 4L and 6, unveiling a distinct Ca2+ release mechanism. Particularly, we determined a structure of the long-sought CaE2P state of P-type IIA ATPases, providing valuable insights into the Ca2+ transport cycle. Together, these findings enhance our understanding of Ca2+ transport by hSPCA1 and broaden our knowledge of P-type ATPases.
Collapse
Affiliation(s)
- Mengqi Wu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Cang Wu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Tiefeng Song
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kewu Pan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- The Provincial International Science and Technology Cooperation Base on Engineering Biology, International Campus of Zhejiang University, Haining, Zhejiang, China.
| | - Zhongmin Liu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
33
|
Richardson SJ, Thekkedam CG, Casarotto MG, Beard NA, Dulhunty AF. FKBP12 binds to the cardiac ryanodine receptor with negative cooperativity: implications for heart muscle physiology in health and disease. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220169. [PMID: 37122219 PMCID: PMC10150220 DOI: 10.1098/rstb.2022.0169] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Cardiac ryanodine receptors (RyR2) release the Ca2+ from intracellular stores that is essential for cardiac myocyte contraction. The ion channel opening is tightly regulated by intracellular factors, including the FK506 binding proteins, FKBP12 and FKBP12.6. The impact of these proteins on RyR2 activity and cardiac contraction is debated, with often apparently contradictory experimental results, particularly for FKBP12. The isoform that regulates RyR2 has generally been considered to be FKBP12.6, despite the fact that FKBP12 is the major isoform associated with RyR2 in some species and is bound in similar proportions to FKBP12.6 in others, including sheep and humans. Here, we show time- and concentration-dependent effects of adding FKBP12 to RyR2 channels that were partly depleted of FKBP12/12.6 during isolation. The added FKBP12 displaced most remaining endogenous FKBP12/12.6. The results suggest that FKBP12 activates RyR2 with high affinity and inhibits RyR2 with lower affinity, consistent with a model of negative cooperativity in FKBP12 binding to each of the four subunits in the RyR tetramer. The easy dissociation of some FKBP12/12.6 could dynamically alter RyR2 activity in response to changes in in vivo regulatory factors, indicating a significant role for FKBP12/12.6 in Ca2+ signalling and cardiac function in healthy and diseased hearts. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- S J Richardson
- John Curtin School of Medical Research, Australian National University, Canberra, Australia, Australian Capital Territory 2601, Australia
| | - C G Thekkedam
- John Curtin School of Medical Research, Australian National University, Canberra, Australia, Australian Capital Territory 2601, Australia
| | - M G Casarotto
- John Curtin School of Medical Research, Australian National University, Canberra, Australia, Australian Capital Territory 2601, Australia
| | - N A Beard
- John Curtin School of Medical Research, Australian National University, Canberra, Australia, Australian Capital Territory 2601, Australia
| | - A F Dulhunty
- John Curtin School of Medical Research, Australian National University, Canberra, Australia, Australian Capital Territory 2601, Australia
| |
Collapse
|
34
|
Du J, Fu Y. Diamide insecticides targeting insect ryanodine receptors: Mechanism and application prospect. Biochem Biophys Res Commun 2023; 670:19-26. [PMID: 37271036 DOI: 10.1016/j.bbrc.2023.05.107] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
As a Lepidoptera pest, Spodoptera frugiperda has become one of the major migratory pests causing significant damage to crops. It should prevent and control Spodoptera frugiperda with strong reproductive ability, adaptability, and migration ability, and reduce economic losses as much as possible. Chemical insecticides are mainly used in the emergency control of Spodoptera frugiperda. Diamide insecticide is a kind of pesticide that specifically targets the ryanodine receptor of Lepidopteran pests, which makes it safe, effective, targeted, and low toxicity to mammals. So, it is one of the most concerned and fastest-growing pesticide products after neonicotinoid pesticides. Intracellular Ca2+ concentration can be regulated by ryanodine receptors, and the continuous release of Ca2+ eventually leads to the death of pests and achieve the insecticidal effect. This review introduces in detail diamide insecticides that mainly play roles in stomach toxicity, as well as its specific target-ryanodine receptor, and analyzes how the diamide insecticide acts on the ryanodine receptor and how its mechanism of action can provide a theoretical basis for the rational use of highly effective insecticides and solve the resistance problem. Moreover, we also propose several recommendations for reducing resistance to diamide insecticides, and provide a reference for chemical control and resistance studies of Spodoptera frugiperda, which has broad development prospects in today's increasingly concerned about the ecological environment and advocating green environmental protection.
Collapse
Affiliation(s)
- Juan Du
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, China
| | - Yuejun Fu
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
35
|
Daba MY, Fan Z, Li Q, Yuan X, Liu B. The Role of Calcium Channels in Prostate Cancer Progression and Potential as a Druggable Target for Prostate Cancer Treatment. Crit Rev Oncol Hematol 2023; 186:104014. [PMID: 37119879 DOI: 10.1016/j.critrevonc.2023.104014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/01/2023] Open
Abstract
Prostate cancer (PCa) is the most diagnosed cancer among men. Discovering novel prognostic biomarkers and potential therapeutic targets are critical. Calcium signaling has been implicated in PCa progression and development of treatment resistance. Altered modification of Ca2+ flows leads to serious pathophysiological processes, such as malignant transformation, tumor proliferation, epithelial to mesenchymal transition, evasion of apoptosis, and treatment resistance. Calcium channels control and contribute to these processes. PCa has shown defective Ca2+ channels, which subsequently promotes tumor metastasis and growth. Store-operated Ca2+ entry channels such as Orai and STIM channels and transient receptor potential channels play a significant role in PCa pathogenesis. Pharmacological modulation of these calcium channels or pumps has been suggested as a practical approach. In this review, we discuss the role of calcium channels in PCa development and progression, and we identify current novel discoveries of drugs that target specific calcium channels for the treatment of PCa.
Collapse
Affiliation(s)
- Motuma Yigezu Daba
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhijie Fan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Qinyu Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
36
|
Chiantia G, Hidisoglu E, Marcantoni A. The Role of Ryanodine Receptors in Regulating Neuronal Activity and Its Connection to the Development of Alzheimer's Disease. Cells 2023; 12:cells12091236. [PMID: 37174636 PMCID: PMC10177020 DOI: 10.3390/cells12091236] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Research into the early impacts of Alzheimer's disease (AD) on synapse function is one of the most promising approaches to finding a treatment. In this context, we have recently demonstrated that the Abeta42 peptide, which builds up in the brain during the processing of the amyloid precursor protein (APP), targets the ryanodine receptors (RyRs) of mouse hippocampal neurons and potentiates calcium (Ca2+) release from the endoplasmic reticulum (ER). The uncontrolled increase in intracellular calcium concentration ([Ca2+]i), leading to the development of Ca2+ dysregulation events and related excitable and synaptic dysfunctions, is a consolidated hallmark of AD onset and possibly other neurodegenerative diseases. Since RyRs contribute to increasing [Ca2+]i and are thought to be a promising target for AD treatment, the goal of this review is to summarize the current level of knowledge regarding the involvement of RyRs in governing neuronal function both in physiological conditions and during the onset of AD.
Collapse
Affiliation(s)
| | - Enis Hidisoglu
- Department of Drug and Science Technology, University of Torino, Corso Raffaello 30, 10125 Torino, Italy
| | - Andrea Marcantoni
- Department of Drug and Science Technology, University of Torino, Corso Raffaello 30, 10125 Torino, Italy
- N.I.S. Center, University of Torino, 10125 Turin, Italy
| |
Collapse
|
37
|
Zhang N, Feng S, Tian Y, Zhuang L, Cha G, Duan S, Li H, Nong X, Zhang Z, Tu X, Wang G. Identification, characterization and spatiotemporal expression analysis of the FKBP family genes in Locusta migratoria. Sci Rep 2023; 13:4048. [PMID: 36899085 PMCID: PMC10006077 DOI: 10.1038/s41598-023-30889-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
FK506 binding proteins (FKBPs) are a highly-conserved group of proteins known to bind to FK506, an immunosuppressive drug. They play different physiological roles, including transcription regulation, protein folding, signal transduction and immunosuppression. A number of FKBP genes have been identified in eukaryotes; however, very little information about these genes has been reported in Locusta migratoria. Here, we identified and characterized 10 FKBP genes from L. migratoria. Phylogenetic analysis and comparison of domain architectures indicated that the LmFKBP family can be divided into two subfamilies and five subclasses. Developmental and tissue expression pattern analysis revealed that all LmFKBPs transcripts, including LmFKBP46, LmFKBP12, LmFKBP47, LmFKBP79, LmFKBP16, LmFKBP24, LmFKBP44b, LmFKBP53, were periodically expressed during different developmental stages and mainly expressed in the fat body, hemolymph, testis, and ovary. In brief, our work depicts a outline but panoramic picture of LmFKBP family in L. migratoria, and provides a solid foundation to further investigate the molecular functions of LmFKBPs.
Collapse
Affiliation(s)
- Neng Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,Scientific Observation and Experimental Station of Pests in Xilingol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, 026000, China
| | - Shiqian Feng
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Ye Tian
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Ling Zhuang
- Bayannur Forestry and Grassland Development Center, Bayannur, 015000, China
| | - Gan Cha
- Bayannur Forestry and Grassland Development Center, Bayannur, 015000, China
| | - Saiya Duan
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hongmei Li
- MARA-CABI Joint Laboratory for Bio-Safety, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, 100193, China
| | - Xiangqun Nong
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zehua Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Xiongbing Tu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,Scientific Observation and Experimental Station of Pests in Xilingol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, 026000, China
| | - Guangjun Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China. .,Scientific Observation and Experimental Station of Pests in Xilingol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, 026000, China.
| |
Collapse
|
38
|
Zhang Y, Han S, Liu C, Zheng Y, Li H, Gao F, Bian Y, Liu X, Liu H, Hu S, Li Y, Chen ZJ, Zhao S, Zhao H. THADA inhibition in mice protects against type 2 diabetes mellitus by improving pancreatic β-cell function and preserving β-cell mass. Nat Commun 2023; 14:1020. [PMID: 36823211 PMCID: PMC9950491 DOI: 10.1038/s41467-023-36680-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Impaired insulin secretion is a hallmark in type 2 diabetes mellitus (T2DM). THADA has been identified as a candidate gene for T2DM, but its role in glucose homeostasis remains elusive. Here we report that THADA is strongly activated in human and mouse islets of T2DM. Both global and β-cell-specific Thada-knockout mice exhibit improved glycemic control owing to enhanced β-cell function and decreased β-cell apoptosis. THADA reduces endoplasmic reticulum (ER) Ca2+ stores in β-cells by inhibiting Ca2+ re-uptake via SERCA2 and inducing Ca2+ leakage through RyR2. Upon persistent ER stress, THADA interacts with and activates the pro-apoptotic complex comprising DR5, FADD and caspase-8, thus aggravating ER stress-induced apoptosis. Importantly, THADA deficiency protects mice from high-fat high-sucrose diet- and streptozotocin-induced hyperglycemia by restoring insulin secretion and preserving β-cell mass. Moreover, treatment with alnustone inhibits THADA's function, resulting in ameliorated hyperglycemia in obese mice. Collectively, our results support pursuit of THADA as a potential target for developing T2DM therapies.
Collapse
Affiliation(s)
- Yuqing Zhang
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Shan Han
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Congcong Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Yuanwen Zheng
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong, China
| | - Hao Li
- Shandong Provincial Qianfoshan Hospital, Shandong University, 250014, Jinan, Shandong, China
| | - Fei Gao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Science, 100101, Beijing, China
| | - Yuehong Bian
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Xin Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Shourui Hu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Yuxuan Li
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, 200135, Shanghai, China. .,Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, 250012, Jinan, China.
| | - Shigang Zhao
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China.
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China.
| |
Collapse
|
39
|
Zhang J, Qin W, Hu C, Gu S, Chai X, Yang M, Zhou F, Wang X, Chen K, Yan G, Wang G, Jiang C, Warren A, Xiong J, Miao W. Giant proteins in a giant cell: Molecular basis of ultrafast Ca 2+-dependent cell contraction. SCIENCE ADVANCES 2023; 9:eadd6550. [PMID: 36812318 PMCID: PMC9946354 DOI: 10.1126/sciadv.add6550] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
The giant single-celled eukaryote, Spirostomum, exhibits one of the fastest movements in the biological world. This ultrafast contraction is dependent on Ca2+ rather than ATP and therefore differs to the actin-myosin system in muscle. We obtained the high-quality genome of Spirostomum minus from which we identified the key molecular components of its contractile apparatus, including two major Ca2+ binding proteins (Spasmin 1 and 2) and two giant proteins (GSBP1 and GSBP2), which act as the backbone and allow for the binding of hundreds of spasmins. The evidence suggests that the GSBP-spasmin protein complex is the functional unit of the mesh-like contractile fibrillar system, which, coupled with various other subcellular structures, provides the mechanism for repetitive ultrafast cell contraction and extension. These findings improve our understanding of the Ca2+-dependent ultrafast movement and provide a blueprint for future biomimicry, design, and construction of this kind of micromachine.
Collapse
Affiliation(s)
- Jing Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Weiwei Qin
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Che Hu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- Harbin Normal University, Harbin 150025, China
| | - Siyu Gu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaocui Chai
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Mingkun Yang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Fang Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xueyan Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Guanxiong Yan
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Guangying Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Chuanqi Jiang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Alan Warren
- Department of Life Sciences, Natural History Museum, London SW7 5BD, UK
| | - Jie Xiong
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Wei Miao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Freshwater Ecology and Biotechnology of China, Wuhan 430072, China
- CAS Center for Excellence in Animal Evolution and Genetics, Kunming 650223, China
| |
Collapse
|
40
|
Abstract
This Review provides an update on ryanodine receptors (RyRs) and their role in human diseases of heart, muscle, and brain. Calcium (Ca2+) is a requisite second messenger in all living organisms. From C. elegans to mammals, Ca2+ is necessary for locomotion, bodily functions, and neural activity. However, too much of a good thing can be bad. Intracellular Ca2+ overload can result in loss of function and death. Intracellular Ca2+ release channels evolved to safely provide large, rapid Ca2+ signals without exposure to toxic extracellular Ca2+. RyRs are intracellular Ca2+ release channels present throughout the zoosphere. Over the past 35 years, our knowledge of RyRs has advanced to the level of atomic-resolution structures revealing their role in the mechanisms underlying the pathogenesis of human disorders of heart, muscle, and brain. Stress-induced RyR-mediated intracellular Ca2+ leak in the heart can promote heart failure and cardiac arrhythmias. In skeletal muscle, RyR1 leak contributes to muscle weakness in inherited myopathies, to age-related loss of muscle function and cancer-associated muscle weakness, and to impaired muscle function in muscular dystrophies, including Duchenne. In the brain, leaky RyR channels contribute to cognitive dysfunction in Alzheimer's disease, posttraumatic stress disorder, and Huntington's disease. Novel therapeutics targeting dysfunctional RyRs are showing promise.
Collapse
|
41
|
Greene D, Luchko T, Shiferaw Y. The role of subunit cooperativity on ryanodine receptor 2 calcium signaling. Biophys J 2023; 122:215-229. [PMID: 36348625 PMCID: PMC9822801 DOI: 10.1016/j.bpj.2022.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/09/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
The ryanodine receptor type 2 (RyR2) is composed of four subunits that control calcium (Ca) release in cardiac cells. RyR2 serves primarily as a Ca sensor and can respond to rapid sub-millisecond pulses of Ca while remaining shut at resting concentrations. However, it is not known how the four subunits interact for the RyR2 to function as an effective Ca sensor. To address this question, and to understand the role of subunit cooperativity in Ca-mediated signal transduction, we have developed a computational model of the RyR2 composed of four interacting subunits. We first analyze the statistical properties of a single RyR2 tetramer, where each subunit can exist in a closed or open conformation. Our findings indicate that the number of subunits in the open state is a crucial parameter that dictates RyR2 kinetics. We find that three or four open subunits are required for the RyR2 to harness cooperative interactions to respond to sub-millisecond changes in Ca, while at the same time remaining shut at the resting Ca levels in the cardiac cell. If the required number of open subunits is lowered to one or two, the RyR2 cannot serve as a robust Ca sensor, as the large cooperativity required to stabilize the closed state prevents channel activation. Using this four-subunit model, we analyze the kinetics of Ca release from a RyR2 cluster. We show that the closure of a cluster of RyR2 channels is highly sensitive to the balance of cooperative interactions between closed and open subunits. Based on this result, we analyze how specific interactions between RyR2 subunits can induce persistent Ca leak from the sarcoplasmic reticulum (SR), which is believed to be arrhythmogenic. Thus, these results provide a framework to analyze how a pharmacologic or genetic modification of RyR2 subunit cooperativity can induce abnormal Ca cycling that can potentially lead to life-threatening arrhythmias.
Collapse
Affiliation(s)
- D'Artagnan Greene
- Department of Physics & Astronomy, California State University, Northridge
| | - Tyler Luchko
- Department of Physics & Astronomy, California State University, Northridge
| | - Yohannes Shiferaw
- Department of Physics & Astronomy, California State University, Northridge.
| |
Collapse
|
42
|
Campiglio M, Dyrda A, Tuinte WE, Török E. Ca V1.1 Calcium Channel Signaling Complexes in Excitation-Contraction Coupling: Insights from Channelopathies. Handb Exp Pharmacol 2023; 279:3-39. [PMID: 36592225 DOI: 10.1007/164_2022_627] [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: 01/03/2023]
Abstract
In skeletal muscle, excitation-contraction (EC) coupling relies on the mechanical coupling between two ion channels: the L-type voltage-gated calcium channel (CaV1.1), located in the sarcolemma and functioning as the voltage sensor of EC coupling, and the ryanodine receptor 1 (RyR1), located on the sarcoplasmic reticulum serving as the calcium release channel. To this day, the molecular mechanism by which these two ion channels are linked remains elusive. However, recently, skeletal muscle EC coupling could be reconstituted in heterologous cells, revealing that only four proteins are essential for this process: CaV1.1, RyR1, and the cytosolic proteins CaVβ1a and STAC3. Due to the crucial role of these proteins in skeletal muscle EC coupling, any mutation that affects any one of these proteins can have devastating consequences, resulting in congenital myopathies and other pathologies.Here, we summarize the current knowledge concerning these four essential proteins and discuss the pathophysiology of the CaV1.1, RyR1, and STAC3-related skeletal muscle diseases with an emphasis on the molecular mechanisms. Being part of the same signalosome, mutations in different proteins often result in congenital myopathies with similar symptoms or even in the same disease.
Collapse
Affiliation(s)
- Marta Campiglio
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria.
| | - Agnieszka Dyrda
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Wietske E Tuinte
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Enikő Török
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
43
|
Abstract
Calcium ions (Ca2+) play a critical role in triggering neurotransmitter release. The rate of release is directly related to the concentration of Ca2+ at the presynaptic site, with a supralinear relationship. There are two main sources of Ca2+ that trigger synaptic vesicle fusion: influx through voltage-gated Ca2+ channels in the plasma membrane and release from the endoplasmic reticulum via ryanodine receptors. This chapter will cover the sources of Ca2+ at the presynaptic nerve terminal, the relationship between neurotransmitter release rate and Ca2+ concentration, and the mechanisms that achieve the necessary Ca2+ concentrations for triggering synaptic exocytosis at the presynaptic site.
Collapse
Affiliation(s)
- Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Sadaf Riaz
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Longgang Niu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
44
|
Greene D, Barton M, Luchko T, Shiferaw Y. Molecular Dynamics Simulations of the Cardiac Ryanodine Receptor Type 2 (RyR2) Gating Mechanism. J Phys Chem B 2022; 126:9790-9809. [PMID: 36384028 PMCID: PMC9720719 DOI: 10.1021/acs.jpcb.2c03031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mutations in the cardiac ryanodine receptor type 2 (RyR2) have been linked to fatal cardiac arrhythmias such as catecholaminergic polymorphic ventricular tachycardia (CPVT). While many CPVT mutations are associated with an increase in Ca2+ leak from the sarcoplasmic reticulum, the mechanistic details of RyR2 channel gating are not well understood, and this poses a barrier in the development of new pharmacological treatments. To address this, we explore the gating mechanism of the RyR2 using molecular dynamics (MD) simulations. We test the effect of changing the conformation of certain structural elements by constructing chimera RyR2 structures that are derived from the currently available closed and open cryo-electron microscopy (cryo-EM) structures, and we then use MD simulations to relax the system. Our key finding is that the position of the S4-S5 linker (S4S5L) on a single subunit can determine whether the channel as a whole is open or closed. Our analysis reveals that the position of the S4S5L is regulated by interactions with the U-motif on the same subunit and with the S6 helix on an adjacent subunit. We find that, in general, channel gating is crucially dependent on high percent occupancy interactions between adjacent subunits. We compare our interaction analysis to 49 CPVT1 mutations in the literature and find that 73% appear near a high percent occupancy interaction between adjacent subunits. This suggests that disruption of cooperative, high percent occupancy interactions between adjacent subunits is a primary cause of channel leak and CPVT in mutant RyR2 channels.
Collapse
|
45
|
Tsuboi Y, Oyama K, Kobirumaki-Shimozawa F, Murayama T, Kurebayashi N, Tachibana T, Manome Y, Kikuchi E, Noguchi S, Inoue T, Inoue YU, Nishino I, Mori S, Ishida R, Kagechika H, Suzuki M, Fukuda N, Yamazawa T. Mice with R2509C-RYR1 mutation exhibit dysfunctional Ca2+ dynamics in primary skeletal myocytes. J Gen Physiol 2022; 154:213526. [PMID: 36200983 PMCID: PMC9546722 DOI: 10.1085/jgp.202213136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/22/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022] Open
Abstract
Type 1 ryanodine receptor (RYR1) is a Ca2+ release channel in the sarcoplasmic reticulum (SR) of the skeletal muscle and plays a critical role in excitation-contraction coupling. Mutations in RYR1 cause severe muscle diseases, such as malignant hyperthermia, a disorder of Ca2+-induced Ca2+ release (CICR) through RYR1 from the SR. We recently reported that volatile anesthetics induce malignant hyperthermia (MH)-like episodes through enhanced CICR in heterozygous R2509C-RYR1 mice. However, the characterization of Ca2+ dynamics has yet to be investigated in skeletal muscle cells from homozygous mice because these animals die in utero. In the present study, we generated primary cultured skeletal myocytes from R2509C-RYR1 mice. No differences in cellular morphology were detected between wild type (WT) and mutant myocytes. Spontaneous Ca2+ transients and cellular contractions occurred in WT and heterozygous myocytes, but not in homozygous myocytes. Electron microscopic observation revealed that the sarcomere length was shortened to ∼1.7 µm in homozygous myocytes, as compared to ∼2.2 and ∼2.3 µm in WT and heterozygous myocytes, respectively. Consistently, the resting intracellular Ca2+ concentration was higher in homozygous myocytes than in WT or heterozygous myocytes, which may be coupled with a reduced Ca2+ concentration in the SR. Finally, using infrared laser-based microheating, we found that heterozygous myocytes showed larger heat-induced Ca2+ transients than WT myocytes. Our findings suggest that the R2509C mutation in RYR1 causes dysfunctional Ca2+ dynamics in a mutant-gene dose-dependent manner in the skeletal muscles, in turn provoking MH-like episodes and embryonic lethality in heterozygous and homozygous mice, respectively.
Collapse
Affiliation(s)
- Yoshitaka Tsuboi
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan.,Department of Molecular Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kotaro Oyama
- Quantum Beam Science Research Directorate, National Institutes for Quantum Science and Technology, Gunma, Japan.,Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Takashi Murayama
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nagomi Kurebayashi
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiaki Tachibana
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshinobu Manome
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Emi Kikuchi
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Satoru Noguchi
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takayoshi Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yukiko U Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shuichi Mori
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryosuke Ishida
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Madoka Suzuki
- Institute for Protein Research, Osaka University, Osaka, Japan
| | - Norio Fukuda
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshiko Yamazawa
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan.,Department of Molecular Physiology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
46
|
Abstract
The global burden caused by cardiovascular disease is substantial, with heart disease representing the most common cause of death around the world. There remains a need to develop better mechanistic models of cardiac function in order to combat this health concern. Heart rhythm disorders, or arrhythmias, are one particular type of disease which has been amenable to quantitative investigation. Here we review the application of quantitative methodologies to explore dynamical questions pertaining to arrhythmias. We begin by describing single-cell models of cardiac myocytes, from which two and three dimensional models can be constructed. Special focus is placed on results relating to pattern formation across these spatially-distributed systems, especially the formation of spiral waves of activation. Next, we discuss mechanisms which can lead to the initiation of arrhythmias, focusing on the dynamical state of spatially discordant alternans, and outline proposed mechanisms perpetuating arrhythmias such as fibrillation. We then review experimental and clinical results related to the spatio-temporal mapping of heart rhythm disorders. Finally, we describe treatment options for heart rhythm disorders and demonstrate how statistical physics tools can provide insights into the dynamics of heart rhythm disorders.
Collapse
Affiliation(s)
- Wouter-Jan Rappel
- Department of Physics, University of California San Diego, La Jolla, CA 92037
| |
Collapse
|
47
|
Rossi D, Catallo MR, Pierantozzi E, Sorrentino V. Mutations in proteins involved in E-C coupling and SOCE and congenital myopathies. J Gen Physiol 2022; 154:e202213115. [PMID: 35980353 PMCID: PMC9391951 DOI: 10.1085/jgp.202213115] [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: 01/31/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
In skeletal muscle, Ca2+ necessary for muscle contraction is stored and released from the sarcoplasmic reticulum (SR), a specialized form of endoplasmic reticulum through the mechanism known as excitation-contraction (E-C) coupling. Following activation of skeletal muscle contraction by the E-C coupling mechanism, replenishment of intracellular stores requires reuptake of cytosolic Ca2+ into the SR by the activity of SR Ca2+-ATPases, but also Ca2+ entry from the extracellular space, through a mechanism called store-operated calcium entry (SOCE). The fine orchestration of these processes requires several proteins, including Ca2+ channels, Ca2+ sensors, and Ca2+ buffers, as well as the active involvement of mitochondria. Mutations in genes coding for proteins participating in E-C coupling and SOCE are causative of several myopathies characterized by a wide spectrum of clinical phenotypes, a variety of histological features, and alterations in intracellular Ca2+ balance. This review summarizes current knowledge on these myopathies and discusses available knowledge on the pathogenic mechanisms of disease.
Collapse
Affiliation(s)
- Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Interdepartmental Program of Molecular Diagnosis and Pathogenetic Mechanisms of Rare Genetic Diseases, Azienda Ospedaliero Universitaria Senese, Siena, Italy
| | - Maria Rosaria Catallo
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Interdepartmental Program of Molecular Diagnosis and Pathogenetic Mechanisms of Rare Genetic Diseases, Azienda Ospedaliero Universitaria Senese, Siena, Italy
| |
Collapse
|
48
|
Bolaños P, Calderón JC. Excitation-contraction coupling in mammalian skeletal muscle: Blending old and last-decade research. Front Physiol 2022; 13:989796. [PMID: 36117698 PMCID: PMC9478590 DOI: 10.3389/fphys.2022.989796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The excitation–contraction coupling (ECC) in skeletal muscle refers to the Ca2+-mediated link between the membrane excitation and the mechanical contraction. The initiation and propagation of an action potential through the membranous system of the sarcolemma and the tubular network lead to the activation of the Ca2+-release units (CRU): tightly coupled dihydropyridine and ryanodine (RyR) receptors. The RyR gating allows a rapid, massive, and highly regulated release of Ca2+ from the sarcoplasmic reticulum (SR). The release from triadic places generates a sarcomeric gradient of Ca2+ concentrations ([Ca2+]) depending on the distance of a subcellular region from the CRU. Upon release, the diffusing Ca2+ has multiple fates: binds to troponin C thus activating the contractile machinery, binds to classical sarcoplasmic Ca2+ buffers such as parvalbumin, adenosine triphosphate and, experimentally, fluorescent dyes, enters the mitochondria and the SR, or is recycled through the Na+/Ca2+ exchanger and store-operated Ca2+ entry (SOCE) mechanisms. To commemorate the 7th decade after being coined, we comprehensively and critically reviewed “old”, historical landmarks and well-established concepts, and blended them with recent advances to have a complete, quantitative-focused landscape of the ECC. We discuss the: 1) elucidation of the CRU structures at near-atomic resolution and its implications for functional coupling; 2) reliable quantification of peak sarcoplasmic [Ca2+] using fast, low affinity Ca2+ dyes and the relative contributions of the Ca2+-binding mechanisms to the whole concert of Ca2+ fluxes inside the fibre; 3) articulation of this novel quantitative information with the unveiled structural details of the molecular machinery involved in mitochondrial Ca2+ handing to understand how and how much Ca2+ enters the mitochondria; 4) presence of the SOCE machinery and its different modes of activation, which awaits understanding of its magnitude and relevance in situ; 5) pharmacology of the ECC, and 6) emerging topics such as the use and potential applications of super-resolution and induced pluripotent stem cells (iPSC) in ECC. Blending the old with the new works better!
Collapse
Affiliation(s)
- Pura Bolaños
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| | - Juan C. Calderón
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellín, Colombia
- *Correspondence: Juan C. Calderón,
| |
Collapse
|
49
|
Ishida R, Mori S, Murayama T, Nakamichi A, Chai X, Kurebayashi N, Iinuma H, Kagechika H. Development of a water-soluble ryanodine receptor 1 inhibitor. Bioorg Med Chem 2022; 74:117027. [DOI: 10.1016/j.bmc.2022.117027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/25/2022]
|
50
|
Wang Y, Hou Y, Song S, Zuo Y, Yu Y, Chi Y, Zhang T. Harm of circadian misalignment to the hearts of the adolescent wistar rats. J Transl Med 2022; 20:352. [PMID: 35933342 PMCID: PMC9356460 DOI: 10.1186/s12967-022-03546-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/17/2022] [Indexed: 11/10/2022] Open
Abstract
PURPOSE The purpose of this study was to observe the harm of circadian misalignment (CM), caused by an inverted photoperiod (IP), on the hearts of the adolescent Wistar rats, and to explore the mechanisms leading to harm. METHODS An IP was used to create a CM model. A total of 174 Wistar rats were randomly divided into circadian alignment (CA) and CM groups (87 rats per group). The different activity rhythms of the two groups of rats were adjusted through different light/dark cycles for 90 days. We recorded the rhythmic activity trajectory and sleep time of the rats. After 90 days of modeling, we performed various analyses (i.e., blood pressure, weight, cardiac ultrasound tests, serological tests, cardiac tissue immunofluorescence, immunohistochemistry, transmission electron microscopy on myocardial mitochondria, western blotting, and quantitative polymerase chain reactions). RESULTS (1) The IP protocol caused CM in rats. (2) CM rats showed significantly higher blood pressure during the day (resting phase). They also showed significantly higher serum levels of angiotensin II and epinephrine during the day compared to the CA rats. (3) CM caused up-regulation of gene expression of adrenergic receptors α1 (α1-AR) and β1 (β1-AR) and down-regulation of the glucocorticoid receptor (Gr) gene expression in rat hearts. It also caused downregulation of Bmal1 expression. In addition, the changes in Bmal1 and Per2 correlated with the changes in β1-AR and α1-AR. (4) CM had adverse effects on multiple molecular proteins of the heart. (5) CM increased the collagen fibers in the rat heart and increased the destruction of mitochondria. (6) Eventually, CM caused a decrease in the pumping function of the heart and decreased the coronary blood flow rate. CONCLUSIONS (1) CM significantly affected the cardiac structure and function in the adolescent rats through a variety of mechanisms. (2) CM can regulate the expression of myocardial clock genes, and it is likely to have an impact on the heart through this pathway.
Collapse
Affiliation(s)
- YunLei Wang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China.,Beijing Bo'ai Hospital, China Rehabilitation Research Center, No.10 JiaoMen North Road, Fengtai District, Beijing, 100068, China.,Lab of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, 100068, China
| | - YuanYuan Hou
- School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China.,Beijing Bo'ai Hospital, China Rehabilitation Research Center, No.10 JiaoMen North Road, Fengtai District, Beijing, 100068, China.,Lab of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, 100068, China
| | - ShaoFei Song
- School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China.,Beijing Bo'ai Hospital, China Rehabilitation Research Center, No.10 JiaoMen North Road, Fengtai District, Beijing, 100068, China.,Lab of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, 100068, China
| | - Yao Zuo
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,University of Health and Rehabilitation Sciences, Qingdao, 266071, Shandong, China
| | - Yan Yu
- China Rehabilitation Science Institute, Beijing, 100068, China
| | - YaFei Chi
- Capital Medical University, Beijing, 100069, China
| | - Tong Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China. .,Beijing Bo'ai Hospital, China Rehabilitation Research Center, No.10 JiaoMen North Road, Fengtai District, Beijing, 100068, China. .,Lab of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, 100068, China.
| |
Collapse
|