1
|
Yang S, Song C. Multiple-Basin Go̅-Martini for Investigating Conformational Transitions and Environmental Interactions of Proteins. J Chem Theory Comput 2025; 21:5304-5321. [PMID: 40359486 DOI: 10.1021/acs.jctc.5c00256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Proteins are inherently dynamic molecules, and their conformational transitions among various states are essential for numerous biological processes, which are often modulated by their interactions with surrounding environments. Although molecular dynamics (MD) simulations are widely used to investigate these transitions, all-atom (AA) methods are often limited by short time scales and high computational costs, and coarse-grained (CG) implicit-solvent Go̅-like models are usually incapable of studying the interactions between proteins and their environments. Here, we present an approach called Multiple-basin Go̅-Martini, which combines the recent Go̅-Martini model with an exponential mixing scheme to facilitate the simulation of spontaneous protein conformational transitions in explicit environments. We demonstrate the versatility of our method through five diverse case studies: GlnBP, Arc, Hinge, SemiSWEET, and TRAAK, representing ligand-binding proteins, fold-switching proteins, de novo designed proteins, transporters, and mechanosensitive ion channels, respectively. Multiple-basin Go̅-Martini offers a new computational tool for investigating protein conformational transitions, identifying key intermediate states, and elucidating essential interactions between proteins and their environments, particularly protein-membrane interactions. In addition, this approach can efficiently generate thermodynamically meaningful data sets of protein conformational space, which may enhance deep learning-based models for predicting protein conformation distributions.
Collapse
Affiliation(s)
- Song Yang
- Center for Quantitative Biology, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- School of Medicine, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Chen Song
- Center for Quantitative Biology, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
2
|
Roy-Chowdhury S, Jang S, Abderemane-Ali F, Naughton F, Grabe M, Minor DL. Structure of the human K 2P13.1 channel reveals a hydrophilic pore restriction and lipid cofactor site. Nat Struct Mol Biol 2025:10.1038/s41594-024-01476-3. [PMID: 40011746 DOI: 10.1038/s41594-024-01476-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/17/2024] [Indexed: 02/28/2025]
Abstract
Polyunsaturated fatty acid (PUFA) lipids modulate the neuronal and microglial leak potassium channel K2P13.1 (THIK1) and other voltage-gated ion channel (VGIC) superfamily members through poorly understood mechanisms. Here we present cryo-electron microscopy structures of human THIK1 and mutants, revealing a unique two-chamber aqueous inner cavity obstructed by a hydrophilic barrier important for gating, the flow restrictor, and a P1-M4 intersubunit interface lipid at a site, the PUFA site, corresponding to the K2P small-molecule modulator pocket. This overlap, together with functional studies, indicates that PUFA site lipids are THIK1 cofactors. Comparison with a PUFA-responsive VGIC, Kv7.1, reveals a shared modulatory role for the pore domain intersubunit interface, providing a framework for understanding PUFA action on the VGIC superfamily. Our findings reveal the distinct THIK1 architecture, highlight the importance of the P1-M4 interface for K2P control by natural and synthetic ligands and should aid in the development of THIK subfamily modulators for neuroinflammation and autism.
Collapse
Affiliation(s)
| | - Seil Jang
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Fiona Naughton
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
| | - Michael Grabe
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA.
- Departments of Biochemistry and Biophysics and Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
3
|
Cox SS, Connolly DJ, Peng X, Badran BW. A Comprehensive Review of Low-Intensity Focused Ultrasound Parameters and Applications in Neurologic and Psychiatric Disorders. Neuromodulation 2025; 28:1-15. [PMID: 39230530 PMCID: PMC11700779 DOI: 10.1016/j.neurom.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/25/2024] [Accepted: 07/19/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVES Low-intensity focused ultrasound (LIFU) is gaining increased interest as a potential therapeutic modality for a range of neuropsychiatric diseases. Current neuromodulation modalities often require a choice between high spatial fidelity or invasiveness. LIFU is unique in this regard because it provides high spatial acuity of both superficial and deep neural structures while remaining noninvasive. This new form of noninvasive brain stimulation may provide exciting potential treatment options for a variety of neuropsychiatric disorders involving aberrant neurocircuitry within deep brain structures, including pain and substance use disorders. Furthermore, LIFU is compatible with noninvasive neuroimaging techniques, such as functional magnetic resonance imaging and electroencephalography, making it a useful tool for more precise clinical neuroscience research to further understand the central nervous system. MATERIALS AND METHODS In this study, we provide a review of the most recent LIFU literature covering three key domains: 1) the history of focused ultrasound technology, comparing it with other forms of neuromodulation, 2) the parameters and most up-to-date proposed mechanisms of LIFU, and finally, 3) a consolidation of the current literature to date surrounding the clinical research that has used LIFU for the modification or amelioration of several neuropsychiatric conditions. RESULTS The impact of LIFU including poststroke motor changes, pain, mood disorders, disorders of consciousness, dementia, and substance abuse is discussed. CONCLUSIONS Although still in its infancy, LIFU is a promising tool that has the potential to change the way we approach and treat neuropsychiatric disorders. In this quickly evolving field, this review serves as a snapshot of the current understanding of LIFU in neuropsychiatric research.
Collapse
Affiliation(s)
- Stewart S Cox
- Department of Psychiatry and Behavioral Sciences, Neuro-X Lab, Medical University of South Carolina, Charleston, SC, USA.
| | - Dillon J Connolly
- Department of Psychiatry and Behavioral Sciences, Neuro-X Lab, Medical University of South Carolina, Charleston, SC, USA
| | - Xiaolong Peng
- Department of Psychiatry and Behavioral Sciences, Neuro-X Lab, Medical University of South Carolina, Charleston, SC, USA
| | - Bashar W Badran
- Department of Psychiatry and Behavioral Sciences, Neuro-X Lab, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
4
|
Huang Y, Zhang Z, Hattori M. Recent Advances in Expression Screening and Sample Evaluation for Structural Studies of Membrane Proteins. J Mol Biol 2024; 436:168809. [PMID: 39362625 DOI: 10.1016/j.jmb.2024.168809] [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: 08/27/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Membrane proteins are involved in numerous biological processes and represent more than half of all drug targets; thus, structural information on these proteins is invaluable. However, the low expression level of membrane proteins, as well as their poor stability in solution and tendency to precipitate and aggregate, are major bottlenecks in the preparation of purified membrane proteins for structural studies. Traditionally, the evaluation of membrane protein constructs for structural studies has been quite time consuming and expensive since it is necessary to express and purify the proteins on a large scale, particularly for X-ray crystallography. The emergence of fluorescence detection size exclusion chromatography (FSEC) has drastically changed this situation, as this method can be used to rapidly evaluate the expression and behavior of membrane proteins on a small scale without the need for purification. FSEC has become the most widely used method for the screening of expression conditions and sample evaluation for membrane proteins, leading to the successful determination of numerous structures. Even in the era of cryo-EM, FSEC and the new generation of FSEC derivative methods are being widely used in various manners to facilitate structural analysis. In addition, the application of FSEC is not limited to structural analysis; this method is also widely used for functional analysis of membrane proteins, including for analysis of oligomerization state, screening of antibodies and ligands, and affinity profiling. This review presents the latest advances and applications in membrane protein expression screening and sample evaluation, with a particular focus on FSEC methods.
Collapse
Affiliation(s)
- Yichen Huang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ziyi Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
5
|
Yang S, Yang S, Li P, Gou S, Cheng Y, Jia Q, Du Z. Advanced neuroprosthetic electrode design optimized by electromagnetic finite element simulation: innovations and applications. Front Bioeng Biotechnol 2024; 12:1476447. [PMID: 39574462 PMCID: PMC11579925 DOI: 10.3389/fbioe.2024.1476447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Based on electrophysiological activity, neuroprostheses can effectively monitor and control neural activity. Currently, electrophysiological neuroprostheses are widely utilized in treating neurological disorders, particularly in restoring motor, visual, auditory, and somatosensory functions after nervous system injuries. They also help alleviate inflammation, regulate blood pressure, provide analgesia, and treat conditions such as epilepsy and Alzheimer's disease, offering significant research, economic, and social value. Enhancing the targeting capabilities of neuroprostheses remains a key objective for researchers. Modeling and simulation techniques facilitate the theoretical analysis of interactions between neuroprostheses and the nervous system, allowing for quantitative assessments of targeting efficiency. Throughout the development of neuroprostheses, these modeling and simulation methods can save time, materials, and labor costs, thereby accelerating the rapid development of highly targeted neuroprostheses. This article introduces the fundamental principles of neuroprosthesis simulation technology and reviews how various simulation techniques assist in the design and performance enhancement of neuroprostheses. Finally, it discusses the limitations of modeling and simulation and outlines future directions for utilizing these approaches to guide neuroprosthesis design.
Collapse
Affiliation(s)
- Shu Yang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Siyi Yang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peixuan Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuchun Gou
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuhang Cheng
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qinggang Jia
- Institute of Applied Physics and Computational Mathematics, Beijing, China
| | - Zhanhong Du
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Yu H, Wang W. Modulation of heteromeric glycine receptor function through high concentration clustering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618879. [PMID: 39464082 PMCID: PMC11507885 DOI: 10.1101/2024.10.17.618879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Ion channels are targeted by many drugs for treating neurological, musculoskeletal, renal and other diseases. These drugs bind to and alter the function of individual channels to achieve desired therapeutic effects. However, many ion channels function in high concentration clusters in their native environment. It is unclear if and how clustering modulates ion channel function. Human heteromeric glycine receptors (GlyRs) are the major inhibitory neurotransmitter receptors in the spinal cord and are active targets for developing chronic pain medications. We show that the α2β heteromeric GlyR assembles with the master postsynaptic scaffolding gephyrin (GPHN) into micron-sized clustered at the plasma membrane after heterologous expression. The inhibitory trans- synaptic adhesion protein neuroligin-2 (NL2) further increases both the cluster sizes and GlyR concentration. The apparent glycine affinity increases monotonically as a function of GlyR concentration but not with cluster size. We also show that ligand re-binding to adjacent GlyRs alters kinetics but not chemical equilibrium. A positively charged N- terminus sequence of the GlyR β subunit was further identified essential for glycine affinity modulation through clustering. Taken together, we propose a mechanism where clustering enhances local electrostatic potential, which in turn concentrates ions and ligands, modulating the function of GlyR. This mechanism is likely universal across ion channel clusters found ubiquitously in biology and provides new perspectives in possible pharmaceutical development.
Collapse
|
7
|
Escobedo Jr. G, Wu Y, Ogawa Y, Ding X, Rasband MN. An evolutionarily conserved AnkyrinG-dependent motif clusters axonal K2P K+ channels. J Cell Biol 2024; 223:e202401140. [PMID: 39078369 PMCID: PMC11289519 DOI: 10.1083/jcb.202401140] [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: 01/25/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
The evolution of ion channel clustering at nodes of Ranvier enabled the development of complex vertebrate nervous systems. At mammalian nodes, the K+ leak channels TRAAK and TREK-1 underlie membrane repolarization. Despite the molecular similarities between nodes and the axon initial segment (AIS), TRAAK and TREK-1 are reportedly node-specific, suggesting a unique clustering mechanism. However, we show that TRAAK and TREK-1 are enriched at both nodes and AIS through a common mechanism. We identified a motif near the C-terminus of TRAAK that is necessary and sufficient for its clustering. The motif first evolved among cartilaginous fish. Using AnkyrinG (AnkG) conditional knockout mice, CRISPR/Cas9-mediated disruption of AnkG, co-immunoprecipitation, and surface recruitment assays, we show that TRAAK forms a complex with AnkG and that AnkG is necessary for TRAAK's AIS and nodal clustering. In contrast, TREK-1's clustering requires TRAAK. Our results expand the repertoire of AIS and nodal ion channel clustering mechanisms and emphasize AnkG's central role in assembling excitable domains.
Collapse
Affiliation(s)
| | - Yu Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yuki Ogawa
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
8
|
Tang J, Feng M, Wang D, Zhang L, Yang K. Recent advancement of sonogenetics: A promising noninvasive cellular manipulation by ultrasound. Genes Dis 2024; 11:101112. [PMID: 38947740 PMCID: PMC11214298 DOI: 10.1016/j.gendis.2023.101112] [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: 02/04/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 07/02/2024] Open
Abstract
Recent advancements in biomedical research have underscored the importance of noninvasive cellular manipulation techniques. Sonogenetics, a method that uses genetic engineering to produce ultrasound-sensitive proteins in target cells, is gaining prominence along with optogenetics, electrogenetics, and magnetogenetics. Upon stimulation with ultrasound, these proteins trigger a cascade of cellular activities and functions. Unlike traditional ultrasound modalities, sonogenetics offers enhanced spatial selectivity, improving precision and safety in disease treatment. This technology broadens the scope of non-surgical interventions across a wide range of clinical research and therapeutic applications, including neuromodulation, oncologic treatments, stem cell therapy, and beyond. Although current literature predominantly emphasizes ultrasonic neuromodulation, this review offers a comprehensive exploration of sonogenetics. We discuss ultrasound properties, the specific ultrasound-sensitive proteins employed in sonogenetics, and the technique's potential in managing conditions such as neurological disorders, cancer, and ophthalmic diseases, and in stem cell therapies. Our objective is to stimulate fresh perspectives for further research in this promising field.
Collapse
Affiliation(s)
- Jin Tang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mingxuan Feng
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ke Yang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| |
Collapse
|
9
|
Luque-Fernández V, Vanspauwen SK, Landra-Willm A, Arvedsen E, Besquent M, Sandoz G, Rasmussen HB. An ankyrin G-binding motif mediates TRAAK periodic localization at axon initial segments of hippocampal pyramidal neurons. Proc Natl Acad Sci U S A 2024; 121:e2310120121. [PMID: 39058579 PMCID: PMC11295008 DOI: 10.1073/pnas.2310120121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The axon initial segment (AIS) is a critical compartment in neurons. It converts postsynaptic input into action potentials that subsequently trigger information transfer to target neurons. This process relies on the presence of several voltage-gated sodium (NaV) and potassium (KV) channels that accumulate in high densities at the AIS. TRAAK is a mechanosensitive leak potassium channel that was recently localized to the nodes of Ranvier. Here, we uncover that TRAAK is also present in AISs of hippocampal and cortical neurons in the adult rat brain as well as in AISs of cultured rat hippocampal neurons. We show that the AIS localization is driven by a C-terminal ankyrin G-binding sequence that organizes TRAAK in a 190 nm spaced periodic pattern that codistributes with periodically organized ankyrin G. We furthermore uncover that while the identified ankyrin G-binding motif is analogous to known ankyrin G-binding motifs in NaV1 and KV7.2/KV7.3 channels, it was acquired by convergent evolution. Our findings identify TRAAK as an AIS ion channel that convergently acquired an ankyrin G-binding motif and expand the role of ankyrin G to include the nanoscale organization of ion channels at the AIS.
Collapse
Affiliation(s)
- Virginia Luque-Fernández
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200, Denmark
| | - Sam K. Vanspauwen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200, Denmark
| | - Arnaud Landra-Willm
- Université Côte d’Azur, CNRS, INSERM, Institut de Biologie Valrose, Nice06108, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice06100, France
- Fédération Hospitalo-Universitaire InovPain, Côte d’Azur University, University Hospital Centre Nice, Nice06000, France
| | - Emil Arvedsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200, Denmark
| | - Maïlys Besquent
- Université Côte d’Azur, CNRS, INSERM, Institut de Biologie Valrose, Nice06108, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice06100, France
- Fédération Hospitalo-Universitaire InovPain, Côte d’Azur University, University Hospital Centre Nice, Nice06000, France
| | - Guillaume Sandoz
- Université Côte d’Azur, CNRS, INSERM, Institut de Biologie Valrose, Nice06108, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice06100, France
- Fédération Hospitalo-Universitaire InovPain, Côte d’Azur University, University Hospital Centre Nice, Nice06000, France
| | - Hanne B. Rasmussen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200, Denmark
| |
Collapse
|
10
|
Deal PE, Lee H, Mondal A, Lolicato M, Mendonça PRFD, Black H, Jang S, El-Hilali X, Bryant C, Isacoff EY, Renslo AR, Minor DL. Development of covalent chemogenetic K 2P channel activators. Cell Chem Biol 2024; 31:1305-1323.e9. [PMID: 39029456 PMCID: PMC11433823 DOI: 10.1016/j.chembiol.2024.06.006] [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: 11/01/2023] [Revised: 04/19/2024] [Accepted: 06/19/2024] [Indexed: 07/21/2024]
Abstract
K2P potassium channels regulate excitability by affecting cellular resting membrane potential in the brain, cardiovascular system, immune cells, and sensory organs. Despite their important roles in anesthesia, arrhythmia, pain, hypertension, sleep, and migraine, the ability to control K2P function remains limited. Here, we describe a chemogenetic strategy termed CATKLAMP (covalent activation of TREK family K+ channels to clamp membrane potential) that leverages the discovery of a K2P modulator pocket site that reacts with electrophile-bearing derivatives of a TREK subfamily small-molecule activator, ML335, to activate the channel irreversibly. We show that CATKLAMP can be used to probe fundamental aspects of K2P function, as a switch to silence neuronal firing, and is applicable to all TREK subfamily members. Together, our findings exemplify a means to alter K2P channel activity that should facilitate molecular and systems level studies of K2P function and enable the search for new K2P modulators.
Collapse
Affiliation(s)
- Parker E Deal
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Haerim Lee
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Abhisek Mondal
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | | | - Holly Black
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Seil Jang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Xochina El-Hilali
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Clifford Bryant
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Weill Neurohub, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 93858-2330, USA.
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 93858-2330, USA; California Institute for Quantitative Biomedical Research, University of California, San Francisco, San Francisco, CA 93858-2330, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 93858-2330, USA.
| |
Collapse
|
11
|
Roy-Chowdhury S, Jang S, Abderemane-Ali F, Naughton F, Grabe M, Minor DL. Structure of the human K 2P13.1(THIK-1) channel reveals a novel hydrophilic pore restriction and lipid cofactor site. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600491. [PMID: 38979306 PMCID: PMC11230452 DOI: 10.1101/2024.06.26.600491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The halothane-inhibited K2P leak potassium channel K2P13.1 (THIK-1)1-3 is found in diverse cells1,4 including neurons1,5 and microglia6-8 where it affects surveillance6, synaptic pruning7, phagocytosis7, and inflammasome-mediated interleukin-1β release6,8,9. As with many K2Ps1,5,10-14 and other voltage-gated ion channel (VGIC) superfamily members3,15,16, polyunsaturated fatty acid (PUFA) lipids modulate K2P13.1 (THIK-1)1,5,14,17 via a poorly understood mechanism. Here, we present cryo-electronmicroscopy (cryo-EM) structures of human K2P13.1 (THIK-1) and mutants in lipid nanodiscs and detergent. These reveal that, unlike other K2Ps13,18-24, K2P13.1 (THIK-1) has a two-chamber aqueous inner cavity obstructed by a M4 transmembrane helix tyrosine (Tyr273, the flow restrictor). This hydrophilic barrier can be opened by an activatory mutation, S136P25, at natural break in the M2 transmembrane helix and by intrinsic channel dynamics. The structures also reveal a buried lipid in the P1/M4 intersubunit interface at a location, the PUFA site, that coincides with the TREK subfamily K2P modulator pocket for small molecule agonists18,26,27. This overlap, together with the effects of mutation on K2P13.1 (THIK-1) PUFA responses, indicates that the PUFA site lipids are K2P13.1 (THIK-1) cofactors. Comparison with the PUFA-responsive VGIC Kv7.1 (KCNQ1)28-31 reveals a shared role for the equivalent pore domain intersubunit interface in lipid modulation, providing a framework for dissecting the effects of PUFAs on the VGIC superfamily. Our findings reveal the unique architecture underlying K2P13.1 (THIK-1) function, highlight the importance of the P1/M4 interface in control of K2Ps by both natural and synthetic agents, and should aid development of THIK subfamily modulators for diseases such as neuroinflammation6,32 and autism6.
Collapse
Affiliation(s)
- Shatabdi Roy-Chowdhury
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Seil Jang
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Fiona Naughton
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Michael Grabe
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California 93858-2330 USA
| | - Daniel L. Minor
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
- Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, California 93858-2330 USA
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, California 93858-2330 USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California 93858-2330 USA
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| |
Collapse
|
12
|
Li B, Zhao A, Tian T, Yang X. Mechanobiological insight into brain diseases based on mechanosensitive channels: Common mechanisms and clinical potential. CNS Neurosci Ther 2024; 30:e14809. [PMID: 38923822 PMCID: PMC11197048 DOI: 10.1111/cns.14809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND As physical signals, mechanical cues regulate the neural cells in the brain. The mechanosensitive channels (MSCs) perceive the mechanical cues and transduce them by permeating specific ions or molecules across the plasma membrane, and finally trigger a series of intracellular bioelectrical and biochemical signals. Emerging evidence supports that wide-distributed, high-expressed MSCs like Piezo1 play important roles in several neurophysiological processes and neurological disorders. AIMS To systematically conclude the functions of MSCs in the brain and provide a novel mechanobiological perspective for brain diseases. METHOD We summarized the mechanical cues and MSCs detected in the brain and the research progress on the functional roles of MSCs in physiological conditions. We then concluded the pathological activation and downstream pathways triggered by MSCs in two categories of brain diseases, neurodegenerative diseases and place-occupying damages. Finally, we outlined the methods for manipulating MSCs and discussed their medical potential with some crucial outstanding issues. RESULTS The MSCs present underlying common mechanisms in different brain diseases by acting as the "transportation hubs" to transduce the distinct signal patterns: the upstream mechanical cues and the downstream intracellular pathways. Manipulating the MSCs is feasible to alter the complicated downstream processes, providing them promising targets for clinical treatment. CONCLUSIONS Recent research on MSCs provides a novel insight into brain diseases. The common mechanisms mediated by MSCs inspire a wide range of therapeutic potentials targeted on MSCs in different brain diseases.
Collapse
Affiliation(s)
- Bolong Li
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- College of Life SciencesUniversity of Chinese Academy of ScienceBeijingChina
| | - An‐ran Zhao
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- College of Life SciencesUniversity of Chinese Academy of ScienceBeijingChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| | - Tian Tian
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| | - Xin Yang
- Shenzhen Key Laboratory of Translational Research for Brain Diseases, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Faculty of Life and Health SciencesShenzhen University of Advanced TechnologyShenzhenGuangdongChina
| |
Collapse
|
13
|
Sorum B, Docter T, Panico V, Rietmeijer RA, Brohawn SG. Tension activation of mechanosensitive two-pore domain K+ channels TRAAK, TREK-1, and TREK-2. Nat Commun 2024; 15:3142. [PMID: 38605031 PMCID: PMC11009253 DOI: 10.1038/s41467-024-47208-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
TRAAK, TREK-1, and TREK-2 are mechanosensitive two-pore domain K+ (K2P) channels that contribute to action potential propagation, sensory transduction, and muscle contraction. While structural and functional studies have led to models that explain their mechanosensitivity, we lack a quantitative understanding of channel activation by membrane tension. Here, we define the tension response of mechanosensitive K2Ps using patch-clamp recording and imaging. All are low-threshold mechanosensitive channels (T10%/50% 0.6-2.7 / 4.4-6.4 mN/m) with distinct response profiles. TRAAK is most sensitive, TREK-1 intermediate, and TREK-2 least sensitive. TRAAK and TREK-1 are activated broadly over a range encompassing nearly all physiologically relevant tensions. TREK-2, in contrast, activates over a narrower range like mechanosensitive channels Piezo1, MscS, and MscL. We further show that low-frequency, low-intensity focused ultrasound increases membrane tension to activate TRAAK and MscS. This work provides insight into tension gating of mechanosensitive K2Ps relevant to understanding their physiological roles and potential applications for ultrasonic neuromodulation.
Collapse
Affiliation(s)
- Ben Sorum
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
| | - Trevor Docter
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA
| | - Vincent Panico
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA
| | - Robert A Rietmeijer
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA
| | - Stephen G Brohawn
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
14
|
Prieto ML, Maduke M. Towards an ion-channel-centric approach to ultrasound neuromodulation. Curr Opin Behav Sci 2024; 56:101355. [PMID: 38505510 PMCID: PMC10947167 DOI: 10.1016/j.cobeha.2024.101355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Ultrasound neuromodulation is a promising technology that could revolutionize study and treatment of brain conditions ranging from mood disorders to Alzheimer's disease and stroke. An understanding of how ultrasound directly modulates specific ion channels could provide a roadmap for targeting specific neurological circuits and achieving desired neurophysiological outcomes. Although experimental challenges make it difficult to unambiguously identify which ion channels are sensitive to ultrasound in vivo, recent progress indicates that there are likely several different ion channels involved, including members of the K2P, Piezo, and TRP channel families. A recent result linking TRPM2 channels in the hypothalamus to induction of torpor by ultrasound in rodents demonstrates the feasibility of targeting a specific ion channel in a specific population of neurons.
Collapse
Affiliation(s)
- Martin Loynaz Prieto
- Department of Molecular and Cellular Physiology, Stanford University, 279 Campus Drive West, B151 Beckman Center, Stanford, CA 94305
| | - Merritt Maduke
- Department of Molecular and Cellular Physiology, Stanford University, 279 Campus Drive West, B155 Beckman Center, Stanford, CA 94305
| |
Collapse
|
15
|
Melrose J. Hippo cell signaling and HS-proteoglycans regulate tissue form and function, age-dependent maturation, extracellular matrix remodeling, and repair. Am J Physiol Cell Physiol 2024; 326:C810-C828. [PMID: 38223931 DOI: 10.1152/ajpcell.00683.2023] [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: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
This review examined how Hippo cell signaling and heparan sulfate (HS)-proteoglycans (HSPGs) regulate tissue form and function. Despite being a nonweight-bearing tissue, the brain is regulated by Hippo mechanoresponsive cell signaling pathways during embryonic development. HS-proteoglycans interact with growth factors, morphogens, and extracellular matrix components to regulate development and pathology. Pikachurin and Eyes shut (Eys) interact with dystroglycan to stabilize the photoreceptor axoneme primary cilium and ribbon synapse facilitating phototransduction and neurotransduction with bipolar retinal neuronal networks in ocular vision, the primary human sense. Another HSPG, Neurexin interacts with structural and adaptor proteins to stabilize synapses and ensure specificity of neural interactions, and aids in synaptic potentiation and plasticity in neurotransduction. HSPGs also stabilize the blood-brain barrier and motor neuron basal structures in the neuromuscular junction. Agrin and perlecan localize acetylcholinesterase and its receptors in the neuromuscular junction essential for neuromuscular control. The primary cilium is a mechanosensory hub on neurons, utilized by YES associated protein (YAP)-transcriptional coactivator with PDZ-binding motif (TAZ) Hippo, Hh, Wnt, transforming growth factor (TGF)-β/bone matrix protein (BMP) receptor tyrosine kinase cell signaling. Members of the glypican HSPG proteoglycan family interact with Smoothened and Patched G-protein coupled receptors on the cilium to regulate Hh and Wnt signaling during neuronal development. Control of glycosyl sulfotransferases and endogenous protease expression by Hippo TAZ YAP represents a mechanism whereby the fine structure of HS-proteoglycans can be potentially modulated spatiotemporally to regulate tissue morphogenesis in a similar manner to how Hippo signaling controls sialyltransferase expression and mediation of cell-cell recognition, dysfunctional sialic acid expression is a feature of many tumors.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Sydney Medical School-Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
16
|
Looser ZJ, Faik Z, Ravotto L, Zanker HS, Jung RB, Werner HB, Ruhwedel T, Möbius W, Bergles DE, Barros LF, Nave KA, Weber B, Saab AS. Oligodendrocyte-axon metabolic coupling is mediated by extracellular K + and maintains axonal health. Nat Neurosci 2024; 27:433-448. [PMID: 38267524 PMCID: PMC10917689 DOI: 10.1038/s41593-023-01558-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
The integrity of myelinated axons relies on homeostatic support from oligodendrocytes (OLs). To determine how OLs detect axonal spiking and how rapid axon-OL metabolic coupling is regulated in the white matter, we studied activity-dependent calcium (Ca2+) and metabolite fluxes in the mouse optic nerve. We show that fast axonal spiking triggers Ca2+ signaling and glycolysis in OLs. OLs detect axonal activity through increases in extracellular potassium (K+) concentrations and activation of Kir4.1 channels, thereby regulating metabolite supply to axons. Both pharmacological inhibition and OL-specific inactivation of Kir4.1 reduce the activity-induced axonal lactate surge. Mice lacking oligodendroglial Kir4.1 exhibit lower resting lactate levels and altered glucose metabolism in axons. These early deficits in axonal energy metabolism are associated with late-onset axonopathy. Our findings reveal that OLs detect fast axonal spiking through K+ signaling, making acute metabolic coupling possible and adjusting the axon-OL metabolic unit to promote axonal health.
Collapse
Affiliation(s)
- Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Zainab Faik
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Henri S Zanker
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
17
|
Pillai EK, Franze K. Mechanics in the nervous system: From development to disease. Neuron 2024; 112:342-361. [PMID: 37967561 DOI: 10.1016/j.neuron.2023.10.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 11/17/2023]
Abstract
Physical forces are ubiquitous in biological processes across scales and diverse contexts. This review highlights the significance of mechanical forces in nervous system development, homeostasis, and disease. We provide an overview of mechanical signals present in the nervous system and delve into mechanotransduction mechanisms translating these mechanical cues into biochemical signals. During development, mechanical cues regulate a plethora of processes, including cell proliferation, differentiation, migration, network formation, and cortex folding. Forces then continue exerting their influence on physiological processes, such as neuronal activity, glial cell function, and the interplay between these different cell types. Notably, changes in tissue mechanics manifest in neurodegenerative diseases and brain tumors, potentially offering new diagnostic and therapeutic target opportunities. Understanding the role of cellular forces and tissue mechanics in nervous system physiology and pathology adds a new facet to neurobiology, shedding new light on many processes that remain incompletely understood.
Collapse
Affiliation(s)
- Eva K Pillai
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany; Developmental Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK; Institute of Medical Physics and Microtissue Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestraße 91, 91052 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Kussmaulallee 1, 91054 Erlangen, Germany.
| |
Collapse
|
18
|
Zhu Y, Yun SD, Zhang T, Chang JY, Stover L, Laganowsky A. Native mass spectrometry of proteoliposomes containing integral and peripheral membrane proteins. Chem Sci 2023; 14:14243-14255. [PMID: 38098719 PMCID: PMC10718073 DOI: 10.1039/d3sc04938h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/18/2023] [Indexed: 12/17/2023] Open
Abstract
Cellular membranes are critical to the function of membrane proteins, whether they are associated (peripheral) or embedded (integral) within the bilayer. While detergents have contributed to our understanding of membrane protein structure and function, there remains challenges in characterizing protein-lipid interactions within the context of an intact membrane. Here, we developed a method to prepare proteoliposomes for native mass spectrometry (MS) studies. We first use native MS to detect the encapsulation of soluble proteins within liposomes. We then find the peripheral Gβ1γ2 complex associated with the membrane can be ejected and analyzed using native MS. Four different integral membrane proteins (AmtB, AqpZ, TRAAK, and TREK2), all of which have previously been characterized in detergent, eject from the proteoliposomes as intact complexes bound to lipids that have been shown to tightly associate in detergent, drawing a correlation between the two approaches. We also show the utility of more complex lipid environments, such as a brain polar lipid extract, and show TRAAK ejects from liposomes of this extract bound to lipids. These findings underscore the capability to eject protein complexes from membranes bound to both lipids and metal ions, and this approach will be instrumental in the identification of key protein-lipid interactions.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Sangho D Yun
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Jing-Yuan Chang
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Lauren Stover
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| |
Collapse
|
19
|
Moschetta M, Vurro V, Sesti V, Bertarelli C, Paternò GM, Lanzani G. Modulation of Mechanosensitive Potassium Channels by a Membrane-targeted Nongenetic Photoswitch. J Phys Chem B 2023; 127:8869-8878. [PMID: 37815392 PMCID: PMC10591468 DOI: 10.1021/acs.jpcb.3c04551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/11/2023] [Indexed: 10/11/2023]
Abstract
Mechanosensitive ion channels are present in the plasma membranes of all cells. They play a fundamental role in converting mechanical stimuli into biochemical signals and are involved in several physiological processes such as touch sensation, hearing, and blood pressure regulation. This protein family includes TWIK-related arachidonic acid-stimulated K+ channel (TRAAK), which is specifically implicated in the maintenance of the resting membrane potential and in the regulation of a variety of important neurobiological functions. Dysregulation of these channels has been linked to various diseases, including blindness, epilepsy, cardiac arrhythmia, and chronic pain. For these reasons, mechanosensitive channels are targets for the treatment of several diseases. Here, we propose a new approach to investigate TRAAK ion channel modulation that is based on nongenetic photostimulation. We employed an amphiphilic azobenzene, named Ziapin2. In the dark, Ziapin2 preferentially dwells in the plasma membrane, causing a thinning of the membrane. Upon light irradiation, an isomerization occurs, breaking the dimers and inducing membrane relaxation. To study the effect of Ziapin2 on the mechanosensitive channels, we expressed human TRAAK (hTRAAK) channels in HEK293T cells. We observed that Ziapin2 insertion in the membrane is able per se to recruit hTRAAK, permitting the exit of K+ ions outside the cells with a consequent hyperpolarization of the cell membrane. During light stimulation, membrane relaxation induces hTRAAK closure, generating a consistent and compensatory depolarization. These results add information to the Ziapin2 mechanism and suggest that membrane deformation can be a tool for the nonselective modulation of mechanosensitive channels.
Collapse
Affiliation(s)
- Matteo Moschetta
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, Via Rubattino, 81, 20134 Milano, Italy
| | - Vito Vurro
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, Via Rubattino, 81, 20134 Milano, Italy
| | - Valentina Sesti
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, Via Rubattino, 81, 20134 Milano, Italy
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Chiara Bertarelli
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, Via Rubattino, 81, 20134 Milano, Italy
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Giuseppe Maria Paternò
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, Via Rubattino, 81, 20134 Milano, Italy
- Department
of Physics, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Guglielmo Lanzani
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, Via Rubattino, 81, 20134 Milano, Italy
- Department
of Physics, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| |
Collapse
|
20
|
Deal PE, Lee H, Mondal A, Lolicato M, de Mendonca PRF, Black H, El-Hilali X, Bryant C, Isacoff EY, Renslo AR, Minor DL. Development of covalent chemogenetic K 2P channel activators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.15.561774. [PMID: 37905049 PMCID: PMC10614804 DOI: 10.1101/2023.10.15.561774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
K2P potassium channels regulate excitability by affecting cellular resting membrane potential in the brain, cardiovascular system, immune cells, and sensory organs. Despite their important roles in anesthesia, arrhythmia, pain, hypertension, sleep, and migraine, the ability to control K2P function remains limited. Here, we describe a chemogenetic strategy termed CATKLAMP (Covalent Activation of TREK family K+ channels to cLAmp Membrane Potential) that leverages the discovery of a site in the K2P modulator pocket that reacts with electrophile-bearing derivatives of a TREK subfamily small molecule activator, ML335, to activate the channel irreversibly. We show that the CATKLAMP strategy can be used to probe fundamental aspects of K2P function, as a switch to silence neuronal firing, and is applicable to all TREK subfamily members. Together, our findings exemplify a new means to alter K2P channel activity that should facilitate studies both molecular and systems level studies of K2P function and enable the search for new K2P modulators.
Collapse
Affiliation(s)
- Parker E. Deal
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 93858-2330 USA
| | - Haerim Lee
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Abhisek Mondal
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | | | - Holly Black
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Xochina El-Hilali
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 93858-2330 USA
| | - Clifford Bryant
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 93858-2330 USA
| | - Ehud Y. Isacoff
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California 94720, United States
- Weill Neurohub, University of California, Berkeley, Berkeley, California 94720, United States
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 93858-2330 USA
| | - Daniel L. Minor
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
- Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, California 93858-2330 USA
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, California 93858-2330 USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California 93858-2330 USA
| |
Collapse
|
21
|
Bogdanović I, Opačić M, Baščarević V, Raičević S, Ilić R, Grujičić D, Spasojević I, Ristić AJ. A potential role of mechano-gated potassium channels in meningioma-related seizures. Heliyon 2023; 9:e20761. [PMID: 37860528 PMCID: PMC10582377 DOI: 10.1016/j.heliyon.2023.e20761] [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: 06/08/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023] Open
Abstract
Every third patient with intracranial meningioma develops seizures of poorly understood etiology. Tumor and peritumoral edema may exert mechanical pressure on the cortex that may affect mechano-gated potassium channels - KCNK2 and KCNK4. These channels regulate neuron excitability and have been related to seizures in some other conditions. The objective of the present study was to explore a potential relation between the levels of these proteins in tumor tissue and adjacent cortex and seizures development. The study included 19 meningioma patients that presented one or more preoperative seizures and 24 patients with no seizures. Tissue samples were collected in the course of surgical removal of the meningioma. Postoperative seizure freedom was achieved in 11 out of 19 patients. The relative level of KCNK2 in the cortical tissue was lower in patients with preoperative seizures. On the other hand, cortical tissue level of KCNK4 was higher in patients that became seizure-free after the surgery. In addition, relative levels of KCNK4 in the cortical and tumor tissue appear to be lowered by the treatment with anti-seizure medication levetiracetam. These results imply that KCNK2 and KCNK4 may be involved in the development of meningioma-related seizures and may represent promising therapeutic targets.
Collapse
Affiliation(s)
- Ivan Bogdanović
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Miloš Opačić
- University of Belgrade - Institute for Multidisciplinary Research, Life Sciences Department, Kneza Višeslava 1, 11030, Belgrade, Serbia
| | - Vladimir Baščarević
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Savo Raičević
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Rosanda Ilić
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Danica Grujičić
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Ivan Spasojević
- University of Belgrade - Institute for Multidisciplinary Research, Life Sciences Department, Kneza Višeslava 1, 11030, Belgrade, Serbia
| | - Aleksandar J. Ristić
- Center for Epilepsy and Sleep Disorders, Neurology Clinic, Clinical Center of Serbia, Dr Subotića Starijeg 6, 11000, Belgrade, Serbia
| |
Collapse
|
22
|
Zhu Y, Odenkirk MT, Qiao P, Zhang T, Schrecke S, Zhou M, Marty MT, Baker ES, Laganowsky A. Combining native mass spectrometry and lipidomics to uncover specific membrane protein-lipid interactions from natural lipid sources. Chem Sci 2023; 14:8570-8582. [PMID: 37593000 PMCID: PMC10430552 DOI: 10.1039/d3sc01482g] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023] Open
Abstract
While it is known that lipids play an essential role in regulating membrane protein structure and function, it remains challenging to identify specific protein-lipid interactions. Here, we present an innovative approach that combines native mass spectrometry (MS) and lipidomics to identify lipids retained by membrane proteins from natural lipid extracts. Our results reveal that the bacterial ammonia channel (AmtB) enriches specific cardiolipin (CDL) and phosphatidylethanolamine (PE) from natural headgroup extracts. When the two extracts are mixed, AmtB retains more species, wherein selectivity is tuned to bias headgroup selection. Using a series of natural headgroup extracts, we show TRAAK, a two-pore domain K+ channel (K2P), retains specific acyl chains that is independent of the headgroup. A brain polar lipid extract was then combined with the K2Ps, TRAAK and TREK2, to understand lipid specificity. More than a hundred lipids demonstrated affinity for each protein, and both channels were found to retain specific fatty acids and lysophospholipids known to stimulate channel activity, even after several column washes. Natural lipid extracts provide the unique opportunity to not only present natural lipid diversity to purified membrane proteins but also identify lipids that may be important for membrane protein structure and function.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Melanie T Odenkirk
- Department of Chemistry, North Carolina State University Raleigh NC 27695 USA
| | - Pei Qiao
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Samantha Schrecke
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| | - Ming Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine Houston TX 77030 USA
| | - Michael T Marty
- Department of Chemistry and Biochemistry, The University of Arizona Tucson AZ 85721 USA
| | - Erin S Baker
- Department of Chemistry, University of North Carolina Chapel Hill NC 27514 USA
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University College Station TX 77843 USA
| |
Collapse
|
23
|
Dolma S, Joshi A. The Node of Ranvier as an Interface for Axo-Glial Interactions: Perturbation of Axo-Glial Interactions in Various Neurological Disorders. J Neuroimmune Pharmacol 2023; 18:215-234. [PMID: 37285016 DOI: 10.1007/s11481-023-10072-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/19/2023] [Indexed: 06/08/2023]
Abstract
The action potential conduction along the axon is highly dependent on the healthy interactions between the axon and myelin-producing glial cells. Myelin, which facilitates action potential, is the protective insulation around the axon formed by Schwann cells and oligodendrocytes in the peripheral (PNS) and central nervous system (CNS), respectively. Myelin is a continuous structure with intermittent gaps called nodes of Ranvier, which are the sites enriched with ion channels, transmembrane, scaffolding, and cytoskeletal proteins. Decades-long extensive research has identified a comprehensive proteome with strictly regularized localization at the node of Ranvier. Concurrently, axon-glia interactions at the node of Ranvier have gathered significant attention as the pathophysiological targets for various neurodegenerative disorders. Numerous studies have shown the alterations in the axon-glia interactions culminating in neurological diseases. In this review, we have provided an update on the molecular composition of the node of Ranvier. Further, we have discussed in detail the consequences of disruption of axon-glia interactions during the pathogenesis of various CNS and PNS disorders.
Collapse
Affiliation(s)
- Sonam Dolma
- Department of Pharmacy, Birla Institute of Technology and Sciences- Pilani, Hyderabad campus, Telangana state, India
| | - Abhijeet Joshi
- Department of Pharmacy, Birla Institute of Technology and Sciences- Pilani, Hyderabad campus, Telangana state, India.
| |
Collapse
|
24
|
Pérez-Mitta G, MacKinnon R. Freestanding lipid bilayer tensiometer for the study of mechanosensitive ion channels. Proc Natl Acad Sci U S A 2023; 120:e2221541120. [PMID: 36913590 PMCID: PMC10041094 DOI: 10.1073/pnas.2221541120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/12/2023] [Indexed: 03/15/2023] Open
Abstract
Mechanical forces modify the cell membrane potential by opening mechanosensitive ion channels. We report the design and construction of a lipid bilayer tensiometer to study channels that respond to lateral membrane tension, [Formula: see text] , in the range 0.2 to 1.4 [Formula: see text] (0.8 to 5.7 [Formula: see text] ). The instrument consists of a black-lipid-membrane bilayer, a custom-built microscope, and a high-resolution manometer. Values of [Formula: see text] are obtained from the determination of the bilayer curvature as a function of applied pressure by means of the Young-Laplace equation. We demonstrate that [Formula: see text] can be determined by calculating the bilayer radius of curvature from fluorescence microscopy imaging or from measurements of the bilayer's electrical capacitance, both yielding similar results. Using electrical capacitance, we show that the mechanosensitive potassium channel TRAAK responds to [Formula: see text] , not curvature. TRAAK channel open probability increases as [Formula: see text] is increased from 0.2 to 1.4 [Formula: see text] but open probability never reaches 0.5. Thus, TRAAK opens over a wide range of [Formula: see text] , but with a tension sensitivity about one-fifth that of the bacterial mechanosensitive channel MscL.
Collapse
Affiliation(s)
- Gonzalo Pérez-Mitta
- HHMI, The Rockefeller University, New York, NY, 10065
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY, 10065
| | - Roderick MacKinnon
- HHMI, The Rockefeller University, New York, NY, 10065
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY, 10065
| |
Collapse
|
25
|
Hopper AJ, Beswick‐Jones H, Brown AM. Resilience of compound action potential peaks to high-frequency firing in the mouse optic nerve. Physiol Rep 2023; 11:e15606. [PMID: 36807847 PMCID: PMC9937793 DOI: 10.14814/phy2.15606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/16/2023] [Accepted: 01/23/2023] [Indexed: 02/19/2023] Open
Abstract
Action potential conduction in axons triggers trans-membrane ion movements, where Na+ enters and K+ leaves axons, leading to disruptions in resting trans-membrane ion gradients that must be restored for optimal axon conduction, an energy dependent process. The higher the stimulus frequency, the greater the ion movements and the resulting energy demand. In the mouse optic nerve (MON), the stimulus evoked compound action potential (CAP) displays a triple peaked profile, consistent with subpopulations of axons classified by size producing the distinct peaks. The three CAP peaks show differential sensitivity to high-frequency firing, with the large axons, which contribute to the 1st peak, more resilient than the small axons, which produce the 3rd peak. Modeling studies predict frequency dependent intra-axonal Na+ accumulation at the nodes of Ranvier, sufficient to attenuate the triple peaked CAP. Short bursts of high-frequency stimulus evoke transient elevations in interstitial K+ ([K+ ]o ), which peak at about 50 Hz. However, powerful astrocytic buffering limits the [K+ ]o increase to levels insufficient to cause CAP attenuation. A post-stimulus [K+ ]o undershoot below baseline coincides with a transient increase in the amplitudes of all three CAP peaks. The volume specific scaling relating energy expenditure to increasing axon size dictates that large axons are more resilient to high-frequency firing than small axons.
Collapse
Affiliation(s)
- Amy J. Hopper
- School of Life SciencesUniversity of NottinghamNottinghamUK
| | | | - Angus M. Brown
- School of Life SciencesUniversity of NottinghamNottinghamUK
- Department of Neurology, School of MedicineUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
26
|
Tonomura S, Gu JG. Saltatory conduction and intrinsic electrophysiological properties at the nodes of ranvier of Aα/β-afferent fibers and Aα-efferent fibers in rat sciatic nerves. Mol Pain 2023; 19:17448069231187366. [PMID: 37369680 PMCID: PMC10413906 DOI: 10.1177/17448069231187366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/05/2023] [Accepted: 06/23/2023] [Indexed: 06/29/2023] Open
Abstract
Large-diameter myelinated fibers in sciatic nerves are composed of both Aα/β-afferent fibers and Aα-efferent fibers to convey sensory and motor impulses, respectively, via saltatory conduction for rapid leg responses. Saltatory conduction and electrophysiological properties at the nodes of Ranvier (NRs) of these sciatic nerve fibers have not been directly studied. We used ex vivo sciatic nerve preparations from rats and applied patch-clamp recordings at the NRs of both Aα/β-afferent fibers and Aα-efferent fibers in the sciatic nerves to characterize their saltatory conduction and intrinsic electrophysiological properties. The velocity and frequency of saltatory conduction in both types of fibers were similar. Resting membrane potentials (RMPs), input resistance, action potential (AP) threshold, and AP rheobase were also not significantly different at the NRs of the two types of fibers in the sciatic nerves. In comparison with Aα/β-afferent fibers, Aα-efferent fibers in the sciatic nerves show higher amplitude and broader width of APs at their NRs. At the NRs of both types of fibers, depolarizing voltages evoked transient inward currents followed by non-inactivating outward currents, and the inward currents and non-inactivating outward currents at the NRs were not significantly different between the two types of fibers. Using AP-clamp, inward currents during AP upstroke were found to be insignificant difference, but amplitudes of non-inactivating outward currents during AP repolarization were significantly lower at the NRs of Aα-efferent fibers than at the NRs of Aα/β-afferent fibers in the sciatic nerves. Collectively, saltatory conduction, ionic currents, and intrinsic electrophysiological properties at the NRs of Aα/β-afferent fibers and Aα-efferent fibers in the sciatic nerves are generally similar, but some differences were also observed.
Collapse
Affiliation(s)
- Sotatsu Tonomura
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama, Birmingham, AL, USA
| | - Jianguo G Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama, Birmingham, AL, USA
| |
Collapse
|
27
|
Abstract
The ankyrin proteins (Ankyrin-R, Ankyrin-B, and Ankyrin-G) are a family of scaffolding, or membrane adaptor proteins necessary for the regulation and targeting of several types of ion channels and membrane transporters throughout the body. These include voltage-gated sodium, potassium, and calcium channels in the nervous system, heart, lungs, and muscle. At these sites, ankyrins recruit ion channels, and other membrane proteins, to specific subcellular domains, which are then stabilized through ankyrin's interaction with the submembranous spectrin-based cytoskeleton. Several recent studies have expanded our understanding of both ankyrin expression and their ion channel binding partners. This review provides an updated overview of ankyrin proteins and their known channel and transporter interactions. We further discuss several potential avenues of future research that would expand our understanding of these important organizational proteins.
Collapse
Affiliation(s)
- Sharon R. Stevens
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA,CONTACT Matthew N. Rasband Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX77030, USA
| |
Collapse
|
28
|
Tanner GR, Tzingounis AV. The mammalian nodal action potential: new data bring new perspectives. ADVANCES IN PHYSIOLOGY EDUCATION 2022; 46:693-702. [PMID: 36173340 DOI: 10.1152/advan.00171.2021] [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: 11/01/2021] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Since its discovery in the mid-20th century, the Hodgkin-Huxley biophysical model of the squid giant axon's (SGA's) neurophysiology has traditionally served as the basis for the teaching of action potential (AP) dynamics in the physiology classroom. This model teaches that leak conductances set membrane resting potential; that fast, inactivating, voltage-gated sodium channels effect the SGA AP upstroke; and that delayed, rectifying, noninactivating voltage-gated potassium channels carry AP repolarization and the early part of the afterhyperpolarization (AHP). This model serves well to introduce students to the fundamental ideas of resting potential establishment and maintenance, as well as basic principles of AP generation and propagation. Furthermore, the Hodgkin-Huxley SGA model represents an excellent and accessible starting point for discussion of the concept of AP threshold and the role of passive electrical properties of the neuron. Additionally, the introduction of the Hodgkin-Huxley model of the SGA AP permits the integration of physiological principles, as instructors ask students to apply previously studied principles of transporter and channel biophysics to the essential physiological phenomenon of electrical signal conduction. However, both some early observations as well as more recent evidence strongly suggest that this seminal invertebrate model of AP dynamics does not appropriately capture the full story for mammalian axons. We review recent evidence that mammalian axonal nodes of Ranvier repolarize largely (though not exclusively) through the activity of leak potassium-ion (K+) conductances carried through two-pore domain (K2P) channels. We call for changes to physiology textbooks and curricula to highlight this remarkable difference in invertebrate and mammalian AP repolarization mechanisms.NEW & NOTEWORTHY Historically, physiology courses have typically taught that action potential repolarization occurs exclusively due to the activation of delayed-rectifier voltage-gated potassium channels. Here, we review and highlight recent evidence that leak potassium channels of the two-pore domain (K2P) class may largely serve this repolarization role at mammalian nodes of Ranvier. We call for the inclusion of these ideas in physiology curricula at all levels, from high school to graduate school.
Collapse
Affiliation(s)
- Geoffrey R Tanner
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
- Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut
| | - Anastasios V Tzingounis
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
- Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
29
|
Zhang Y, Shen B, Wu T, Zhao J, Jing JC, Wang P, Sasaki-Capela K, Dunphy WG, Garrett D, Maslov K, Wang W, Wang LV. Ultrafast and hypersensitive phase imaging of propagating internodal current flows in myelinated axons and electromagnetic pulses in dielectrics. Nat Commun 2022; 13:5247. [PMID: 36068212 PMCID: PMC9448739 DOI: 10.1038/s41467-022-33002-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/25/2022] [Indexed: 12/30/2022] Open
Abstract
Many ultrafast phenomena in biology and physics are fundamental to our scientific understanding but have not yet been visualized owing to the extreme speed and sensitivity requirements in imaging modalities. Two examples are the propagation of passive current flows through myelinated axons and electromagnetic pulses through dielectrics, which are both key to information processing in living organisms and electronic devices. Here, we demonstrate differentially enhanced compressed ultrafast photography (Diff-CUP) to directly visualize propagations of passive current flows at approximately 100 m/s along internodes, i.e., continuous myelinated axons between nodes of Ranvier, from Xenopus laevis sciatic nerves and of electromagnetic pulses at approximately 5 × 107 m/s through lithium niobate. The spatiotemporal dynamics of both propagation processes are consistent with the results from computational models, demonstrating that Diff-CUP can span these two extreme timescales while maintaining high phase sensitivity. With its ultrahigh speed (picosecond resolution), high sensitivity, and noninvasiveness, Diff-CUP provides a powerful tool for investigating ultrafast biological and physical phenomena.
Collapse
Affiliation(s)
- Yide Zhang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Binglin Shen
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Tong Wu
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Key Laboratory of Space Photoelectric Detection and Perception, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Jerry Zhao
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Joseph C Jing
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Peng Wang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Kanomi Sasaki-Capela
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - William G Dunphy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - David Garrett
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Konstantin Maslov
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Weiwei Wang
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lihong V Wang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
30
|
Sarkar D, Kang J, Wassie AT, Schroeder ME, Peng Z, Tarr TB, Tang AH, Niederst ED, Young JZ, Su H, Park D, Yin P, Tsai LH, Blanpied TA, Boyden ES. Revealing nanostructures in brain tissue via protein decrowding by iterative expansion microscopy. Nat Biomed Eng 2022; 6:1057-1073. [PMID: 36038771 PMCID: PMC9551354 DOI: 10.1038/s41551-022-00912-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 06/22/2022] [Indexed: 12/25/2022]
Abstract
Many crowded biomolecular structures in cells and tissues are inaccessible to labelling antibodies. To understand how proteins within these structures are arranged with nanoscale precision therefore requires that these structures be decrowded before labelling. Here we show that an iterative variant of expansion microscopy (the permeation of cells and tissues by a swellable hydrogel followed by isotropic hydrogel expansion, to allow for enhanced imaging resolution with ordinary microscopes) enables the imaging of nanostructures in expanded yet otherwise intact tissues at a resolution of about 20 nm. The method, which we named 'expansion revealing' and validated with DNA-probe-based super-resolution microscopy, involves gel-anchoring reagents and the embedding, expansion and re-embedding of the sample in homogeneous swellable hydrogels. Expansion revealing enabled us to use confocal microscopy to image the alignment of pre-synaptic calcium channels with post-synaptic scaffolding proteins in intact brain circuits, and to uncover periodic amyloid nanoclusters containing ion-channel proteins in brain tissue from a mouse model of Alzheimer's disease. Expansion revealing will enable the further discovery of previously unseen nanostructures within cells and tissues.
Collapse
Affiliation(s)
- Deblina Sarkar
- Media Lab, MIT, Cambridge, MA, 02139, USA.,MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Jinyoung Kang
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Asmamaw T Wassie
- Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Margaret E. Schroeder
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA
| | - Zhuyu Peng
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Tyler B. Tarr
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ai-Hui Tang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, China
| | - Emily D. Niederst
- The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Jennie, Z. Young
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Hanquan Su
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Demian Park
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA
| | - Peng Yin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA. .,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Correspondence and requests for materials should be addressed to Li-Huei Tsai, Thomas A. Blanpied or Edward S. Boyden. , ,
| | - Edward S. Boyden
- MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, 02139, USA.,MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA.,Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,Koch Institute, MIT, Cambridge, MA, 02139, USA.,Howard Hughes Medical Institute, Cambridge, MA, 02139, USA.,Media Arts and Sciences, MIT, Cambridge, MA, 02139, USA.,K. Lisa Yang Center for Bionics, MIT, Cambridge, MA, 02139, USA.,Correspondence and requests for materials should be addressed to Li-Huei Tsai, Thomas A. Blanpied or Edward S. Boyden. , ,
| |
Collapse
|
31
|
Tonomura S, Ling J, Gu JG. Function of KCNQ2 channels at nodes of Ranvier of lumbar spinal ventral nerves of rats. Mol Brain 2022; 15:64. [PMID: 35858950 PMCID: PMC9297653 DOI: 10.1186/s13041-022-00949-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/07/2022] [Indexed: 11/28/2022] Open
Abstract
Previous immunohistochemical studies have shown the expression of KCNQ2 channels at nodes of Ranvier (NRs) of myelinated nerves. However, functions of these channels at NRs remain elusive. In the present study, we addressed this issue by directly applying whole-cell patch-clamp recordings at NRs of rat lumbar spinal ventral nerves in ex vivo preparations. We show that depolarizing voltages evoke large non-inactivating outward currents at NRs, which are partially inhibited by KCNQ channel blocker linopirdine and potentiated by KCNQ channel activator retigabine. Furthermore, linopirdine significantly alters intrinsic electrophysiological properties of NRs to depolarize resting membrane potential, increase input resistance, prolong AP width, reduce AP threshold, and decrease AP amplitude. On the other hand, retigabine significantly decreases input resistance and increases AP rheobase at NRs. Moreover, linopirdine increases excitability at NRs by converting single AP firing into multiple AP firing at many NRs. Saltatory conduction velocity is significantly reduced by retigabine, and AP success rate at high stimulation frequency is significantly increased by linopirdine. Collectively, KCNQ2 channels play a significant role in regulating intrinsic electrophysiological properties and saltatory conduction at NRs of motor nerve fibers of rats. These findings may provide insights into how the loss-of-function mutation in KCNQ2 channels can lead to neuromuscular disorders in human patients.
Collapse
Affiliation(s)
- Sotatsu Tonomura
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jennifer Ling
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jianguo G Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
32
|
Tonomura S, Gu JG. Role of Voltage-Gated K + Channels and K2P Channels in Intrinsic Electrophysiological Properties and Saltatory Conduction at Nodes of Ranvier of Rat Lumbar Spinal Ventral Nerves. J Neurosci 2022; 42:4980-4994. [PMID: 35606142 PMCID: PMC9233441 DOI: 10.1523/jneurosci.0514-22.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/02/2022] [Accepted: 05/17/2022] [Indexed: 12/24/2022] Open
Abstract
Ion channels at the nodes of Ranvier (NRs) are believed to play essential roles in intrinsic electrophysiological properties and saltatory conduction of action potentials (AP) at the NRs of myelinated nerves. While we have recently shown that two-pore domain potassium (K2P) channels play a key role at the NRs of Aβ-afferent nerves, K+ channels and their functions at the NRs of mammalian motor nerves remain elusive. Here we addressed this issue by using ex vivo preparations of lumbar spinal ventral nerves from both male and female rats and the pressure-patch-clamp recordings at their NRs. We found that depolarizing voltages evoked large noninactivating outward currents at NRs. The outward currents could be partially inhibited by voltage-gated K+ channel blockers, largely inhibited by K2P blockers and cooling temperatures. Inhibition of the outward currents by voltage-gated K+ channel blockers, K2P blockers, or cooling temperatures significantly altered electrophysiological properties measured at the NRs, including resting membrane potential, input resistance, AP width, AP amplitude, AP threshold, and AP rheobase. Furthermore, K2P blockers and cooling temperatures significantly reduced saltatory conduction velocity and success rates of APs in response to high-frequency stimulation. Voltage-gated K+ channel blockers reduced AP success rates at high-frequency stimulation without significantly affecting saltatory conduction velocity. Collectively, both K2P and voltage-gated K+ channels play significant roles in intrinsic electrophysiological properties and saltatory conduction at NRs of motor nerve fibers of rats. The effects of cooling temperatures on saltatory conduction are at least partially mediated by K2P channels at the NRs.SIGNIFICANCE STATEMENT Ion channels localized at the NRs are believed to be key determinants of saltatory conduction on myelinated nerves. However, ion channels and their functions at the NRs have not been fully studied in different types of mammalian myelinated nerves. Here we use the pressure-patch-clamp recordings to show that both K2P and voltage-gated K+ channels play significant roles in intrinsic electrophysiological properties and saltatory conduction at NRs of lumbar spinal ventral nerves of rats. Furthermore, cooling temperatures exert effects on saltatory conduction via inhibition of ion channels at the NRs. Our results provide new insights into saltatory conduction on myelinated nerves and may have physiological as well as pathologic implications.
Collapse
Affiliation(s)
- Sotatsu Tonomura
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jianguo G Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
33
|
Sato K, Sasaki R, Matsuda R, Nakagawa M, Ekimoto T, Yamane T, Ikeguchi M, Tabata KV, Noji H, Kinbara K. Supramolecular Mechanosensitive Potassium Channel Formed by Fluorinated Amphiphilic Cyclophane. J Am Chem Soc 2022; 144:11802-11809. [PMID: 35727684 DOI: 10.1021/jacs.2c04118] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Inspired by mechanosensitive potassium channels found in nature, we developed a fluorinated amphiphilic cyclophane composed of fluorinated rigid aromatic units connected via flexible hydrophilic octa(ethylene glycol) chains. Microscopic and emission spectroscopic studies revealed that the cyclophane could be incorporated into the hydrophobic layer of the lipid bilayer membranes and self-assembled to form a supramolecular transmembrane ion channel. Current recording measurements using cyclophane-containing planer lipid bilayer membranes successfully demonstrated an efficient transmembrane ion transport. We also demonstrated that the ion transport property was sensitive to the mechanical forces applied to the membranes. In addition, ion transport assays using pH-sensitive fluorescence dye revealed that the supramolecular channel possesses potassium ion selectivity. We also performed all-atom hybrid quantum-mechanical/molecular mechanical simulations to assess the channel structures at atomic resolution and the mechanism of selective potassium ion transport. This research demonstrated the first example of a synthetic mechanosensitive potassium channel, which would open a new door to sensing and manipulating biologically important processes and purification of key materials in industries.
Collapse
Affiliation(s)
- Kohei Sato
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Ryo Sasaki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Ryoto Matsuda
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Mayuko Nakagawa
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Toru Ekimoto
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Tsutomu Yamane
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Mitsunori Ikeguchi
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Kazuhito V Tabata
- Department of Applied Chemistry, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hiroyuki Noji
- Department of Applied Chemistry, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kazushi Kinbara
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.,World Research Hub Initiative, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| |
Collapse
|
34
|
Turney TS, Li V, Brohawn SG. Structural Basis for pH-gating of the K + channel TWIK1 at the selectivity filter. Nat Commun 2022; 13:3232. [PMID: 35680900 PMCID: PMC9184524 DOI: 10.1038/s41467-022-30853-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/20/2022] [Indexed: 11/11/2022] Open
Abstract
TWIK1 (K2P1.1, KCNK1) is a widely expressed pH-gated two-pore domain K+ channel (K2P) that contributes to cardiac rhythm generation and insulin release from pancreatic beta cells. TWIK1 displays unique properties among K2Ps including low basal activity and inhibition by extracellular protons through incompletely understood mechanisms. Here, we present cryo-EM structures of TWIK1 in lipid nanodiscs at high and low pH that reveal a previously undescribed gating mechanism at the K+ selectivity filter. At high pH, TWIK1 adopts an open conformation. At low pH, protonation of an extracellular histidine results in a cascade of conformational changes that close the channel by sealing the top of the selectivity filter, displacing the helical cap to block extracellular ion access pathways, and opening gaps for lipid block of the intracellular cavity. These data provide a mechanistic understanding for extracellular pH-gating of TWIK1 and illustrate how diverse mechanisms have evolved to gate the selectivity filter of K+ channels.
Collapse
Affiliation(s)
- Toby S Turney
- Biophysics Graduate Program, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California Berkeley, Berkeley, CA, 94720, USA
| | - Vivian Li
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences (QB3), University of California Berkeley, Berkeley, CA, 94720, USA
| | - Stephen G Brohawn
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
- California Institute for Quantitative Biosciences (QB3), University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
35
|
Panasawatwong A, Pipatpolkai T, Tucker SJ. Transition between conformational states of the TREK-1 K2P channel promoted by interaction with PIP 2. Biophys J 2022; 121:2380-2388. [PMID: 35596528 DOI: 10.1016/j.bpj.2022.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/02/2022] [Accepted: 05/16/2022] [Indexed: 11/02/2022] Open
Abstract
Members of the TREK family of two-pore domain (K2P) potassium channels are highly sensitive to regulation by membrane lipids, including phosphatidylinositol-4,5-bisphosphate (PIP2). Previous studies have demonstrated that PIP2 increases TREK1 channel activity, however, the mechanistic understanding of the conformational transitions induced by PIP2 remain unclear. Here, we used coarse-grained molecular dynamics (CG-MD) and atomistic MD simulations to model the PIP2 binding site on both the up and down state conformations of TREK-1. We also calculated the free energy of PIP2 binding relative to other anionic phospholipids in both conformational states using potential of mean force (PMF) and free energy perturbation (FEP) calculations. Our results identify state-dependent binding of PIP2 to sites involving the proximal C-terminus and we show that PIP2 promotes a conformational transition from a down state towards an intermediate that resembles the up state. These results are consistent with functional data for PIP2 regulation and together provide evidence for a structural mechanism of TREK-1 channel activation by phosphoinositides.
Collapse
Affiliation(s)
| | - Tanadet Pipatpolkai
- Department of Physiology Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, U.K.; Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, U.K.; OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PT, U.K..
| | - Stephen J Tucker
- Department of Physics, University of Oxford, Parks Road, Oxford, OX1 3PU, U.K.; OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PT, U.K.; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, U.K..
| |
Collapse
|
36
|
Zhu Y, Schrecke S, Tang S, Odenkirk MT, Walker T, Stover L, Lyu J, Zhang T, Russell D, Baker ES, Yan X, Laganowsky A. Cupric Ions Selectively Modulate TRAAK-Phosphatidylserine Interactions. J Am Chem Soc 2022; 144:7048-7053. [PMID: 35421309 DOI: 10.1021/jacs.2c00612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
TRAAK and TREK2 are two-pore domain K+ (K2P) channels and are modulated by diverse factors including temperature, membrane stretching, and lipids, such as phosphatidic acid. In addition, copper and zinc, both of which are essential for life, are known to regulate TREK2 and a number of other ion channels. However, the role of ions in the association of lipids with integral membrane proteins is poorly understood. Here, we discover cupric ions selectively modulate the binding of phosphatidylserine (PS) to TRAAK but not TREK2. Other divalent cations (Ca2+, Mg2+, and Zn2+) bind both channels but have no impact on binding PS and other lipids. Additionally, TRAAK binds more avidly to Cu2+ and Zn2+ than TREK2. In the presence of Cu2+, TRAAK binds similarly to PS with different acyl chains, indicating a crucial role of the serine headgroup in coordinating Cu2+. High-resolution native mass spectrometry (MS) enables the determination of equilibrium binding constants for distinct Cu2+-bound stoichiometries and uncovered the highest coupling factor corresponds to a 1:1 PS-to-Cu2+ ratio. Interestingly, the next three highest coupling factors had a ∼1.5:1 PS-to-Cu2+ ratio. Our findings bring forth the role of cupric ions as an essential cofactor in selective TRAAK-PS interactions.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Samantha Schrecke
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Shuli Tang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Melanie T Odenkirk
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Thomas Walker
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Lauren Stover
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Jixing Lyu
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - David Russell
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Erin S Baker
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Xin Yan
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
37
|
Pope L, Minor DL. The Polysite Pharmacology of TREK K 2P Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1349:51-65. [PMID: 35138610 DOI: 10.1007/978-981-16-4254-8_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
K2P (KCNK) potassium channels form "background" or "leak" currents that have critical roles in cell excitability control in the brain, cardiovascular system, and somatosensory neurons. Similar to many ion channel families, studies of K2Ps have been limited by poor pharmacology. Of six K2P subfamilies, the thermo- and mechanosensitive TREK subfamily comprising K2P2.1 (TREK-1), K2P4.1 (TRAAK), and K2P10.1 (TREK-2) are the first to have structures determined for each subfamily member. These structural studies have revealed key architectural features that underlie K2P function and have uncovered sites residing at every level of the channel structure with respect to the membrane where small molecules or lipids can control channel function. This polysite pharmacology within a relatively small (~70 kDa) ion channel comprises four structurally defined modulator binding sites that occur above (Keystone inhibitor site), at the level of (K2P modulator pocket), and below (Fenestration and Modulatory lipid sites) the C-type selectivity filter gate that is at the heart of K2P function. Uncovering this rich structural landscape provides the framework for understanding and developing subtype-selective modulators to probe K2P function that may provide leads for drugs for anesthesia, pain, arrhythmia, ischemia, and migraine.
Collapse
Affiliation(s)
- Lianne Pope
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, US
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, US. .,Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA. .,California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, USA. .,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, USA. .,Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
38
|
Riel EB, Jürs BC, Cordeiro S, Musinszki M, Schewe M, Baukrowitz T. The versatile regulation of K2P channels by polyanionic lipids of the phosphoinositide and fatty acid metabolism. J Gen Physiol 2022; 154:212926. [PMID: 34928298 PMCID: PMC8693234 DOI: 10.1085/jgp.202112989] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/01/2021] [Indexed: 12/29/2022] Open
Abstract
Work over the past three decades has greatly advanced our understanding of the regulation of Kir K+ channels by polyanionic lipids of the phosphoinositide (e.g., PIP2) and fatty acid metabolism (e.g., oleoyl-CoA). However, comparatively little is known regarding the regulation of the K2P channel family by phosphoinositides and by long-chain fatty acid–CoA esters, such as oleoyl-CoA. We screened 12 mammalian K2P channels and report effects of polyanionic lipids on all tested channels. We observed activation of members of the TREK, TALK, and THIK subfamilies, with the strongest activation by PIP2 for TRAAK and the strongest activation by oleoyl-CoA for TALK-2. By contrast, we observed inhibition for members of the TASK and TRESK subfamilies. Our results reveal that TASK-2 channels have both activatory and inhibitory PIP2 sites with different affinities. Finally, we provided evidence that PIP2 inhibition of TASK-1 and TASK-3 channels is mediated by closure of the recently identified lower X-gate as critical mutations within the gate (i.e., L244A, R245A) prevent PIP2-induced inhibition. Our findings establish that K+ channels of the K2P family are highly sensitive to polyanionic lipids, extending our knowledge of the mechanisms of lipid regulation and implicating the metabolism of these lipids as possible effector pathways to regulate K2P channel activity.
Collapse
Affiliation(s)
- Elena B Riel
- Institute of Physiology, Kiel University, Kiel, Germany
| | - Björn C Jürs
- Institute of Physiology, Kiel University, Kiel, Germany.,Medical School Hamburg, University of Applied Sciences and Medical University, Hamburg, Germany
| | | | | | - Marcus Schewe
- Institute of Physiology, Kiel University, Kiel, Germany
| | | |
Collapse
|
39
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
40
|
McCoull D, Veale EL, Walsh Y, Byrom L, Avkiran T, Large JM, Vaitone E, Gaffey F, Jerman J, Mathie A, Wright PD. Aprepitant is a novel, selective activator of the K2P channel TRAAK. Biochem Biophys Res Commun 2021; 588:41-46. [PMID: 34942533 DOI: 10.1016/j.bbrc.2021.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 11/02/2022]
Abstract
TRAAK (KCNK4, K2P4.1) is a mechanosensitive two-pore domain potassium (K2P) channel. Due to its expression within sensory neurons and genetic link to neuropathic pain it represents a promising potential target for novel analgesics. In common with many other channels in the wider K2P sub-family, there remains a paucity of small molecule pharmacological tools. Specifically, there is a lack of molecules selective for TRAAK over the other members of the TREK subfamily of K2P channels. We developed a thallium flux assay to allow high throughput screening of compounds and facilitate the identification of novel TRAAK activators. Using a library of ∼1200 drug like molecules we identified Aprepitant as a small molecule activator of TRAAK. Aprepitant is an NK-1 antagonist used to treat nausea and vomiting. Close structural analogues of Aprepitant and a range of NK-1 antagonists were also selected or designed for purchase or brief chemical synthesis and screened for their ability to activate TRAAK. Electrophysiology experiments confirmed that Aprepitant activates both the 'long' and 'short' transcript variants of TRAAK. We also demonstrated that Aprepitant is selective and does not activate other members of the K2P superfamily. This work describes the development of a high throughput assay to identify potential TRAAK activators and subsequent identification and confirmation of the novel TRAAK activator Aprepitant. This discovery identifies a useful tool compound which can be used to further probe the function of TRAAK K2P channels.
Collapse
Affiliation(s)
- D McCoull
- LifeArc, Accelerator Building, Open Innovation Campus, Stevenage, SG1 2FX, UK.
| | - E L Veale
- Medway School of Pharmacy, University of Greenwich and University of Kent, Anson Building, Central Avenue, Chatham Maritime, Kent, ME4 4TB, UK
| | - Y Walsh
- Medway School of Pharmacy, University of Greenwich and University of Kent, Anson Building, Central Avenue, Chatham Maritime, Kent, ME4 4TB, UK
| | - L Byrom
- LifeArc, Accelerator Building, Open Innovation Campus, Stevenage, SG1 2FX, UK
| | - T Avkiran
- LifeArc, Accelerator Building, Open Innovation Campus, Stevenage, SG1 2FX, UK
| | - J M Large
- LifeArc, Accelerator Building, Open Innovation Campus, Stevenage, SG1 2FX, UK
| | - E Vaitone
- LifeArc, Accelerator Building, Open Innovation Campus, Stevenage, SG1 2FX, UK
| | - F Gaffey
- LifeArc, Accelerator Building, Open Innovation Campus, Stevenage, SG1 2FX, UK
| | - J Jerman
- LifeArc, Accelerator Building, Open Innovation Campus, Stevenage, SG1 2FX, UK
| | - A Mathie
- Medway School of Pharmacy, University of Greenwich and University of Kent, Anson Building, Central Avenue, Chatham Maritime, Kent, ME4 4TB, UK; School of Engineering, Arts, Science and Technology, University of Suffolk, Ipswich, IP4 1QJ, UK
| | - P D Wright
- LifeArc, Accelerator Building, Open Innovation Campus, Stevenage, SG1 2FX, UK
| |
Collapse
|
41
|
Lezmy J, Arancibia-Carcamo L, Quintela-Lopez T, Sherman DL, Brophy PJ, Attwell D. Astrocyte Ca 2+-evoked ATP release regulates myelinated axon excitability and conduction speed. Science 2021; 374:eabh2858. [PMID: 34648330 PMCID: PMC7611967 DOI: 10.1126/science.abh2858] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the brain’s gray matter, astrocytes regulate synapse properties, but their role is unclear for the white matter, where myelinated axons rapidly transmit information between gray matter areas. We found that in rodents, neuronal activity raised the intracellular calcium concentration ([Ca2+]i) in astrocyte processes located near action potential–generating sites in the axon initial segment (AIS) and nodes of Ranvier of myelinated axons. This released adenosine triphosphate, which was converted extracellularly to adenosine and thus, through A2a receptors, activated HCN2-containing cation channels that regulate two aspects of myelinated axon function: excitability of the AIS and speed of action potential propagation. Variations in astrocyte-derived adenosine level between wake and sleep states or during energy deprivation could thus control white matter information flow and neural circuit function.
Collapse
Affiliation(s)
- Jonathan Lezmy
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Lorena Arancibia-Carcamo
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
- Dementia Research Institute, Francis Crick Institute 1 Midland Rd, London, NW1 1AT, UK
| | - Tania Quintela-Lopez
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Diane L. Sherman
- Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor’s Building, Edinburgh, EH16 4SB
| | - Peter J. Brophy
- Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor’s Building, Edinburgh, EH16 4SB
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| |
Collapse
|
42
|
Liu J, Ganeshbabu N, Shalaby N, Chen L, Guo T, Feng B. Targeting Two-Pore-Domain Potassium Channels by Mechanical Stretch Instantaneously Modulates Action Potential Transmission in Mouse Sciatic Nerves. ACS Chem Neurosci 2021; 12:3558-3566. [PMID: 34423641 PMCID: PMC9012044 DOI: 10.1021/acschemneuro.1c00052] [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: 11/28/2022] Open
Abstract
Recent reports indicate dominant roles of TRAAK and TREK-1 channels, i.e., mechanosensitive two-pore-domain potassium channels (K2P) at the nodes of Ranvier for action potential repolarization in mammalian peripheral nerves. Functional changes in mammalian peripheral nerve conduction by mechanical stretch studied by recording compound action potentials lack the necessary resolution to detect subtle neuromodulatory effects on conduction velocity. In this study, we developed a novel in vitro approach that enables single-fiber recordings from individual mouse sciatic nerve axons while delivering computer-controlled stepped stretch to the sciatic nerve trunk. Axial stretch instantaneously increased the conduction delay in both myelinated A-fibers and unmyelinated C-fibers. Increases in conduction delay linearly correlated with increases in axial stretch ratio for both A- and C-fibers. The slope of the increase in conduction delay versus stretch ratio was steeper in C-fibers than in A-fibers. Moderate axial stretch (14-19% of in vitro length) reversibly blocked 37.5% of unmyelinated C-fibers but none of the eight myelinated A-fibers tested. Application of arachidonic acid, an agonist to TRAAK and TREK-1 to sciatic nerve trunk, blocks axonal transmission in both A- and C-fibers with delayed onset and prolonged block. Also, the application of an antagonist ruthenium red showed a tendency of suppressing the stretch-evoked increase in conduction delay. These results could draw focused research on pharmacological and mechanical activation of K2P channels as a novel neuromodulatory strategy to achieve peripheral nerve block.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, United States
| | - Nishanth Ganeshbabu
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, United States
| | - Noha Shalaby
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, United States
| | - Longtu Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, United States
| | - Tiantian Guo
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, United States
| | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, United States
| |
Collapse
|
43
|
Rietmeijer RA, Sorum B, Li B, Brohawn SG. Physical basis for distinct basal and mechanically gated activity of the human K + channel TRAAK. Neuron 2021; 109:2902-2913.e4. [PMID: 34390650 PMCID: PMC8448962 DOI: 10.1016/j.neuron.2021.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/10/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
TRAAK is a mechanosensitive two-pore domain K+ (K2P) channel localized to nodes of Ranvier in myelinated neurons. TRAAK deletion in mice results in mechanical and thermal allodynia, and gain-of-function mutations cause the human neurodevelopmental disorder FHEIG. TRAAK displays basal and stimulus-gated activities typical of K2Ps, but the mechanistic and structural differences between these modes are unknown. Here, we demonstrate that basal and mechanically gated openings are distinguished by their conductance, kinetics, and structure. Basal openings are low conductance, short duration, and due to a conductive channel conformation with the interior cavity exposed to the surrounding membrane. Mechanically gated openings are high conductance, long duration, and due to a channel conformation in which the interior cavity is sealed to the surrounding membrane. Our results explain how dual modes of activity are produced by a single ion channel and provide a basis for the development of state-selective pharmacology with the potential to treat disease.
Collapse
Affiliation(s)
- Robert A Rietmeijer
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Biophysics Graduate Program, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Ben Sorum
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Baobin Li
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Stephen G Brohawn
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
44
|
Prieto ML, Firouzi K, Khuri-Yakub BT, Madison DV, Maduke M. Spike frequency-dependent inhibition and excitation of neural activity by high-frequency ultrasound. J Gen Physiol 2021; 152:182190. [PMID: 33074301 PMCID: PMC7534904 DOI: 10.1085/jgp.202012672] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/14/2020] [Indexed: 01/09/2023] Open
Abstract
Ultrasound can modulate action potential firing in vivo and in vitro, but the mechanistic basis of this phenomenon is not well understood. To address this problem, we used patch-clamp recording to quantify the effects of focused, high-frequency (43 MHz) ultrasound on evoked action potential firing in CA1 pyramidal neurons in acute rodent hippocampal brain slices. We find that ultrasound can either inhibit or potentiate firing in a spike frequency–dependent manner: at low (near-threshold) input currents and low firing frequencies, ultrasound inhibits firing, while at higher input currents and higher firing frequencies, ultrasound potentiates firing. The net result of these two competing effects is that ultrasound increases the threshold current for action potential firing, the slope of frequency-input curves, and the maximum firing frequency. In addition, ultrasound slightly hyperpolarizes the resting membrane potential, decreases action potential width, and increases the depth of the after-hyperpolarization. All of these results can be explained by the hypothesis that ultrasound activates a sustained potassium conductance. According to this hypothesis, increased outward potassium currents hyperpolarize the resting membrane potential and inhibit firing at near-threshold input currents but potentiate firing in response to higher-input currents by limiting inactivation of voltage-dependent sodium channels during the action potential. This latter effect is a consequence of faster action potential repolarization, which limits inactivation of voltage-dependent sodium channels, and deeper (more negative) after-hyperpolarization, which increases the rate of recovery from inactivation. Based on these results, we propose that ultrasound activates thermosensitive and mechanosensitive two-pore-domain potassium (K2P) channels through heating or mechanical effects of acoustic radiation force. Finite-element modeling of the effects of ultrasound on brain tissue suggests that the effects of ultrasound on firing frequency are caused by a small (<2°C) increase in temperature, with possible additional contributions from mechanical effects.
Collapse
Affiliation(s)
- Martin Loynaz Prieto
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| | - Kamyar Firouzi
- E.L. Ginzton Laboratory, Stanford University, Stanford, CA
| | | | - Daniel V Madison
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| | - Merritt Maduke
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| |
Collapse
|
45
|
Kanda H, Tonomura S, Gu JG. Effects of Cooling Temperatures via Thermal K2P Channels on Regeneration of High-Frequency Action Potentials at Nodes of Ranvier of Rat Aβ-Afferent Nerves. eNeuro 2021; 8:ENEURO.0308-21.2021. [PMID: 34462308 PMCID: PMC8445039 DOI: 10.1523/eneuro.0308-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 12/02/2022] Open
Abstract
Temperature-sensitive two-pore domain potassium channels (thermal K2P) are recently shown to cluster at nodes of Ranvier (NRs) and play a key role in action potential (AP) regeneration and conduction on Aβ-afferent nerves. Cooling temperatures affect AP regeneration and conduction on Aβ-afferent nerves but the underlying mechanisms are not completely understood. Here, we have performed patch-clamp recordings directly at the NR in an ex vivo trigeminal nerve preparation. We have characterized the effects of cooling temperatures on intrinsic electrophysiological properties and AP regeneration at the NR on rat Aβ-afferent nerves, and determined whether and how thermal K2P channels may be involved in the effects of cooling temperatures. We show that cooling temperatures from 35°C to 15°C decrease outward leak currents, increase input resistance, depolarize resting membrane potential (RMP), broaden AP width and increase latency of AP threshold at the NR. We further demonstrate that cooling temperatures impair regeneration of high-frequency AP trains at the NR. The effects of cooling temperatures on the intrinsic electrophysiological properties and regeneration of high-frequency AP trains at the NR can be partially reversed by BL-1249 (BL), arachidonic acid (AA), and intra-axonal protons, three thermal K2P activators, indicating the involvement of thermal K2P channels. Moreover, we show that at cooling temperatures there are interplays among thermal K2P channels, RMPs, and voltage-gated Na+ channels, which together limit regeneration of high-frequency AP trains at the NR. Our findings demonstrate a new role of thermal K2P channels in temperature-dependent conduction of high-frequency sensory signals.
Collapse
Affiliation(s)
- Hirosato Kanda
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Pharmacology, Hyogo University of Health Sciences, Kobe, Hyogo 650-8530, Japan
| | - Sotatsu Tonomura
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jianguo G Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
46
|
Handler A, Ginty DD. The mechanosensory neurons of touch and their mechanisms of activation. Nat Rev Neurosci 2021; 22:521-537. [PMID: 34312536 PMCID: PMC8485761 DOI: 10.1038/s41583-021-00489-x] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Our sense of touch emerges from an array of mechanosensory structures residing within the fabric of our skin. These tactile end organ structures convert innocuous forces acting on the skin into electrical signals that propagate to the CNS via the axons of low-threshold mechanoreceptors (LTMRs). Our rich capacity for tactile discrimination arises from the dissimilar intrinsic properties of the LTMR subtypes that innervate different regions of the skin and the structurally distinct end organ complexes with which they associate. These end organ structures comprise a range of non-neuronal cell types, which may themselves actively contribute to the transformation of tactile forces into neural impulses within the LTMR afferents. Although the mechanism and the site of transduction across end organs remain unclear, PIEZO2 has emerged as the principal mechanosensitive channel involved in light touch of the skin. Here we review the physiological properties of LTMR subtypes and discuss how features of their cutaneous end organ complexes shape subtype-specific tuning.
Collapse
Affiliation(s)
- Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Ronzano R, Roux T, Thetiot M, Aigrot MS, Richard L, Lejeune FX, Mazuir E, Vallat JM, Lubetzki C, Desmazières A. Microglia-neuron interaction at nodes of Ranvier depends on neuronal activity through potassium release and contributes to remyelination. Nat Commun 2021; 12:5219. [PMID: 34471138 PMCID: PMC8410814 DOI: 10.1038/s41467-021-25486-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 08/11/2021] [Indexed: 12/19/2022] Open
Abstract
Microglia, the resident immune cells of the central nervous system, are key players in healthy brain homeostasis and plasticity. In neurological diseases, such as Multiple Sclerosis, activated microglia either promote tissue damage or favor neuroprotection and myelin regeneration. The mechanisms for microglia-neuron communication remain largely unkown. Here, we identify nodes of Ranvier as a direct site of interaction between microglia and axons, in both mouse and human tissues. Using dynamic imaging, we highlight the preferential interaction of microglial processes with nodes of Ranvier along myelinated fibers. We show that microglia-node interaction is modulated by neuronal activity and associated potassium release, with THIK-1 ensuring their microglial read-out. Altered axonal K+ flux following demyelination impairs the switch towards a pro-regenerative microglia phenotype and decreases remyelination rate. Taken together, these findings identify the node of Ranvier as a major site for microglia-neuron interaction, that may participate in microglia-neuron communication mediating pro-remyelinating effect of microglia after myelin injury. Microglia are important for brain homeostasis and plasticity. The mechanisms underlying microglia-neuron interactions are still unclear. Here, the authors show that microglia preferentially interact with the nodes of Ranvier along axons. This interaction is modulated by neuronal activity and contributes to remyelination in mice.
Collapse
Affiliation(s)
- R Ronzano
- Sorbonne Université, Paris Brain Institute (ICM), INSERM U1127, CNRS UMR 7225, Hopital Pitié-Salpétrière, Paris, France
| | - T Roux
- Sorbonne Université, Paris Brain Institute (ICM), INSERM U1127, CNRS UMR 7225, Hopital Pitié-Salpétrière, Paris, France.,Assistance Publique des Hôpitaux de Paris (APHP), Hopital Pitié-Salpétrière, Département de Neurologie, Paris, France
| | - M Thetiot
- Sorbonne Université, Paris Brain Institute (ICM), INSERM U1127, CNRS UMR 7225, Hopital Pitié-Salpétrière, Paris, France
| | - M S Aigrot
- Sorbonne Université, Paris Brain Institute (ICM), INSERM U1127, CNRS UMR 7225, Hopital Pitié-Salpétrière, Paris, France
| | - L Richard
- Centre de Référence National des Neuropathies Périphériques Rares et Département de Neurologie, Hopital Universitaire, Limoges, France
| | - F X Lejeune
- Sorbonne Université, Paris Brain Institute (ICM), INSERM U1127, CNRS UMR 7225, Hopital Pitié-Salpétrière, Paris, France.,Paris Brain Institute's Data and Analysis Core, University Hospital Pitié-Salpêtrière, Paris, France
| | - E Mazuir
- Sorbonne Université, Paris Brain Institute (ICM), INSERM U1127, CNRS UMR 7225, Hopital Pitié-Salpétrière, Paris, France
| | - J M Vallat
- Centre de Référence National des Neuropathies Périphériques Rares et Département de Neurologie, Hopital Universitaire, Limoges, France
| | - C Lubetzki
- Sorbonne Université, Paris Brain Institute (ICM), INSERM U1127, CNRS UMR 7225, Hopital Pitié-Salpétrière, Paris, France.,Assistance Publique des Hôpitaux de Paris (APHP), Hopital Pitié-Salpétrière, Département de Neurologie, Paris, France
| | - A Desmazières
- Sorbonne Université, Paris Brain Institute (ICM), INSERM U1127, CNRS UMR 7225, Hopital Pitié-Salpétrière, Paris, France.
| |
Collapse
|
48
|
Schwarz JR. Function of K2P channels in the mammalian node of Ranvier. J Physiol 2021; 599:4427-4439. [PMID: 34425634 DOI: 10.1113/jp281723] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/09/2021] [Indexed: 11/08/2022] Open
Abstract
In myelinated nerve fibres, action potentials are generated at nodes of Ranvier. These structures are located at interruptions of the myelin sheath, forming narrow gaps with small rings of axolemma freely exposed to the extracellular space. The mammalian node contains a high density of Na+ channels and K+ -selective leakage channels. Voltage-dependent Kv1 channels are only present in the juxta-paranode. Recently, the leakage channels have been identified as K2P channels (TRAAK, TREK-1). K2P channels are K+ -selective 'background' channels, characterized by outward rectification and their ability to be activated, e.g. by temperature, mechanical stretch or arachidonic acid. We are only beginning to elucidate the peculiar functions of nodal K2P channels. I will discuss two functions of the nodal K2P-mediated conductance. First, at body temperature K2P channels have a high open probability, thereby inducing a resting potential of about -85 mV. This negative resting potential reduces steady-state Na+ channel inactivation and ensures a large Na+ inward current upon a depolarizing stimulus. Second, the K2P conductance is involved in nodal action potential repolarization. The identification of nodal K2P channels is exciting since it shows that the nodal K+ conductance is not a fixed value but can be changed: it can be increased or decreased by a broad range of K2P modulators, thereby modulating, for example, the resting potential. The functional importance of nodal K2P channels will be exemplified by describing in more detail the function of the K2P conductance increase by raising the temperature from room temperature to 37°C.
Collapse
Affiliation(s)
- Jürgen R Schwarz
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
49
|
Lengyel M, Enyedi P, Czirják G. Negative Influence by the Force: Mechanically Induced Hyperpolarization via K 2P Background Potassium Channels. Int J Mol Sci 2021; 22:ijms22169062. [PMID: 34445768 PMCID: PMC8396510 DOI: 10.3390/ijms22169062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 02/08/2023] Open
Abstract
The two-pore domain K2P subunits form background (leak) potassium channels, which are characterized by constitutive, although not necessarily constant activity, at all membrane potential values. Among the fifteen pore-forming K2P subunits encoded by the KCNK genes, the three members of the TREK subfamily, TREK-1, TREK-2, and TRAAK are mechanosensitive ion channels. Mechanically induced opening of these channels generally results in outward K+ current under physiological conditions, with consequent hyperpolarization and inhibition of membrane potential-dependent cellular functions. In the past decade, great advances have been made in the investigation of the molecular determinants of mechanosensation, and members of the TREK subfamily have emerged among the best-understood examples of mammalian ion channels directly influenced by the tension of the phospholipid bilayer. In parallel, the crucial contribution of mechano-gated TREK channels to the regulation of membrane potential in several cell types has been reported. In this review, we summarize the general principles underlying the mechanical activation of K2P channels, and focus on the physiological roles of mechanically induced hyperpolarization.
Collapse
|
50
|
Mathie A, Veale EL, Golluscio A, Holden RG, Walsh Y. Pharmacological Approaches to Studying Potassium Channels. Handb Exp Pharmacol 2021; 267:83-111. [PMID: 34195873 DOI: 10.1007/164_2021_502] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this review, we consider the pharmacology of potassium channels from the perspective of these channels as therapeutic targets. Firstly, we describe the three main families of potassium channels in humans and disease states where they are implicated. Secondly, we describe the existing therapeutic agents which act on potassium channels and outline why these channels represent an under-exploited therapeutic target with potential for future drug development. Thirdly, we consider the evidence desired in order to embark on a drug discovery programme targeting a particular potassium channel. We have chosen two "case studies": activators of the two-pore domain potassium (K2P) channel TREK-2 (K2P10.1), for the treatment of pain and inhibitors of the voltage-gated potassium channel KV1.3, for use in autoimmune diseases such as multiple sclerosis. We describe the evidence base to suggest why these are viable therapeutic targets. Finally, we detail the main technical approaches available to characterise the pharmacology of potassium channels and identify novel regulatory compounds. We draw particular attention to the Comprehensive in vitro Proarrhythmia Assay initiative (CiPA, https://cipaproject.org ) project for cardiac safety, as an example of what might be both desirable and possible in the future, for ion channel regulator discovery projects.
Collapse
Affiliation(s)
- Alistair Mathie
- Medway School of Pharmacy, University of Kent, Kent, UK. .,Medway School of Pharmacy, University of Greenwich, London, UK. .,School of Engineering, Arts, Science and Technology, University of Suffolk, Ipswich, UK.
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Alessia Golluscio
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Robyn G Holden
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| | - Yvonne Walsh
- Medway School of Pharmacy, University of Kent, Kent, UK.,Medway School of Pharmacy, University of Greenwich, London, UK
| |
Collapse
|